Home Cart 0 Sign in  
X

[ CAS No. 638-07-3 ] {[proInfo.proName]}

,{[proInfo.pro_purity]}
Cat. No.: {[proInfo.prAm]}
HazMat Fee +

There will be a HazMat fee per item when shipping a dangerous goods. The HazMat fee will be charged to your UPS/DHL/FedEx collect account or added to the invoice unless the package is shipped via Ground service. Ship by air in Excepted Quantity (each bottle), which is up to 1g/1mL for class 6.1 packing group I or II, and up to 25g/25ml for all other HazMat items.

Type HazMat fee for 500 gram (Estimated)
Excepted Quantity USD 0.00
Limited Quantity USD 15-60
Inaccessible (Haz class 6.1), Domestic USD 80+
Inaccessible (Haz class 6.1), International USD 150+
Accessible (Haz class 3, 4, 5 or 8), Domestic USD 100+
Accessible (Haz class 3, 4, 5 or 8), International USD 200+
3d Animation Molecule Structure of 638-07-3
Chemical Structure| 638-07-3
Chemical Structure| 638-07-3
Structure of 638-07-3 * Storage: {[proInfo.prStorage]}
Cart0 Add to My Favorites Add to My Favorites Bulk Inquiry Inquiry Add To Cart

Quality Control of [ 638-07-3 ]

Related Doc. of [ 638-07-3 ]

Alternatived Products of [ 638-07-3 ]

Product Details of [ 638-07-3 ]

CAS No. :638-07-3 MDL No. :MFCD00000939
Formula : C6H9ClO3 Boiling Point : -
Linear Structure Formula :- InChI Key :OHLRLMWUFVDREV-UHFFFAOYSA-N
M.W : 164.59 Pubchem ID :69484
Synonyms :

Calculated chemistry of [ 638-07-3 ]

Physicochemical Properties

Num. heavy atoms : 10
Num. arom. heavy atoms : 0
Fraction Csp3 : 0.67
Num. rotatable bonds : 5
Num. H-bond acceptors : 3.0
Num. H-bond donors : 0.0
Molar Refractivity : 37.24
TPSA : 43.37 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) : -6.69 cm/s

Lipophilicity

Log Po/w (iLOGP) : 1.51
Log Po/w (XLOGP3) : 0.87
Log Po/w (WLOGP) : 0.75
Log Po/w (MLOGP) : 0.64
Log Po/w (SILICOS-IT) : 1.35
Consensus Log Po/w : 1.02

Druglikeness

Lipinski : 0.0
Ghose : None
Veber : 0.0
Egan : 0.0
Muegge : 1.0
Bioavailability Score : 0.55

Water Solubility

Log S (ESOL) : -1.08
Solubility : 13.7 mg/ml ; 0.0835 mol/l
Class : Very soluble
Log S (Ali) : -1.36
Solubility : 7.11 mg/ml ; 0.0432 mol/l
Class : Very soluble
Log S (SILICOS-IT) : -1.73
Solubility : 3.09 mg/ml ; 0.0187 mol/l
Class : Soluble

Medicinal Chemistry

PAINS : 0.0 alert
Brenk : 2.0 alert
Leadlikeness : 1.0
Synthetic accessibility : 1.88

Safety of [ 638-07-3 ]

Signal Word:Danger Class:8,6.1
Precautionary Statements:P280-P301+P310-P305+P351+P338-P310 UN#:2922
Hazard Statements:H301-H314 Packing Group:
GHS Pictogram:

Application In Synthesis of [ 638-07-3 ]

* 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.

  • Upstream synthesis route of [ 638-07-3 ]
  • Downstream synthetic route of [ 638-07-3 ]

[ 638-07-3 ] Synthesis Path-Upstream   1~25

  • 1
  • [ 638-07-3 ]
  • [ 95-54-5 ]
  • [ 4857-04-9 ]
YieldReaction ConditionsOperation in experiment
82% With tin(ll) chloride In ethanol for 6 h; Reflux The synthesis of 2-(chloromethyl)-1H-benzo[d]imidazoles (3a–f) is the same as described for compound 3a. SnCl2 (0.1 mmol) was added to a stirred solution of ortho-phenylenediamine (1,1 mmol) and ethyl 4-chloro-3-oxobutanoate (2, 1 mmol) in EtOH (2mL) at rt.The mixture was stirred under reflux for 6 h. After completion of the reaction, monitored by thin-layer chromatography(TLC), the solvent was removed under reduced pressure. The crude product was subjected to column chromatography (hexane=AcOEt 84:16) to give the pure benzimidazoles.
Reference: [1] Green Chemistry, 2017, vol. 19, # 24, p. 5818 - 5830
[2] Synthetic Communications, 2015, vol. 45, # 14, p. 1642 - 1651
  • 2
  • [ 638-07-3 ]
  • [ 57-13-6 ]
  • [ 18592-13-7 ]
Reference: [1] Synthetic Communications, 2002, vol. 32, # 6, p. 851 - 855
[2] Heterocyclic Communications, 2013, vol. 19, # 6, p. 401 - 404
  • 3
  • [ 638-07-3 ]
  • [ 17356-08-0 ]
  • [ 53266-94-7 ]
YieldReaction ConditionsOperation in experiment
78% for 4 h; Reflux 4-chloroEthyl acetoacetate,100mmol thiourea, 50mL ethanol, reflux 4h, spin solvent was evaporated to yield an oilLiquid, was added 20mL of ethyl acetate was dissolved, was added dropwise under ice-cooling with stirring, concentrated hydrochloric acid, the precipitated white solid was suction filtered, ethyl acetateA white solid was washed ester 2-aminothiazol-4-acetate hydrochloride, the filter cake was dissolved in water, adjusted to pH 10 ammonia water, ethyl acetateExtracted, dried over anhydrous sodium sulfate, and rotation of the solvent afforded a white solid 2-aminothiazol-4-acetate, yield 78percent, m.p.94 ~ 95 .
Reference: [1] Patent: CN105541752, 2016, A, . Location in patent: Paragraph 0035; 0036; 0037
[2] Journal of Heterocyclic Chemistry, 1980, vol. 17, p. 1255 - 1257
[3] Synlett, 1999, # 8, p. 1239 - 1240
  • 4
  • [ 420-04-2 ]
  • [ 638-07-3 ]
  • [ 53266-94-7 ]
Reference: [1] Journal of Heterocyclic Chemistry, 2017, vol. 54, # 5, p. 2703 - 2707
  • 5
  • [ 638-07-3 ]
  • [ 108-46-3 ]
  • [ 69716-04-7 ]
YieldReaction ConditionsOperation in experiment
67%
Stage #1: at 20℃; for 2 h;
Stage #2: With sodium hydroxide In water for 2 h; Reflux
Weigh 4-ethyl chloroacetoacetate (8. 2g, 50mmol) was dissolved in 20mL of concentrated sulfuric acid in an ice-water bath was cooled to 0 ° C, the reaction system was added in divided portions resorcinol (5. 5g, 50mmol) and naturally to room temperature, stirring was continued for 2 hours, The starting material until the reaction was complete. The reaction solution was poured into 200mL ice water mixture, large amount of solid precipitated, was filtered, and the resulting solid was repeatedly washed with water, dried, and the resulting crude product was used without further purification in the next reaction. The resulting crude product from the previous step was dissolved 400ml 1M in aqueous NaOH and heated at reflux for 2 hours. The reaction was cooled to room temperature, the aqueous solution was washed twice with ethyl acetate, the aqueous phase retained, concentrated sulfuric acid, 200 mL ethyl acetate three times, the combined organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give The product YZ-5 (6. 3g), 67percent yield over two steps.
Reference: [1] Patent: CN103145663, 2016, B, . Location in patent: Paragraph 0116-0118
  • 6
  • [ 64-18-6 ]
  • [ 638-07-3 ]
  • [ 20485-39-6 ]
Reference: [1] J. Gen. Chem. USSR (Engl. Transl.), 1962, vol. 32, p. 2315 - 2320[2] Zhurnal Obshchei Khimii, 1962, vol. 32, p. 2348 - 2353
  • 7
  • [ 638-07-3 ]
  • [ 104-94-9 ]
  • [ 27143-07-3 ]
Reference: [1] Patent: US2006/69258, 2006, A1, . Location in patent: Page/Page column 11-12
  • 8
  • [ 638-07-3 ]
  • [ 108-46-3 ]
  • [ 25392-41-0 ]
YieldReaction ConditionsOperation in experiment
91% at 90℃; for 5 h; General procedure: A mixture of phenol (1.0 mmol), β-ketoester (1.5 mmol) and MNESA (0.075 g) was stirred at 90 °C in a round-bottomed flask for the appreciated time. After completion of the reaction as confirmed by TLC, the reaction mixture was cooled down to room temperature and the catalyst was separated from the reaction mixture using an external magnetic Some field. water was then added to the reaction mixture and the product was extracted using EtOAc (2 9 10 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuumto yield the crude product. For more purification, the crude product was purified by recrystallization in ethanol to obtain the desired purity.
91% With Ag supported on the hydroxyapatite-core–shell magnetic γ-Fe2O3 nanoparticles In neat (no solvent) at 80℃; for 0.333333 h; Green chemistry General procedure: In a round bottom flask, g-Fe2O3HAp-Ag NPs (10 mg) were added to the mixture of the phenolic compound (1 mmol) and ethyl acetoacetate (1 mmol) at 80 C and the reaction mixture stirred for the appropriate time (Table 3). The progress of the reaction was monitored by TLC (eluent, n-hexane:ethyl acetate, 4:1) analysis. Upon completion of the reaction, EtOH was added to the reaction mixture and the g-Fe2O3HAp-Ag NPs were separated with an external magnet. The solvent was then removed under reduced pressure and the resulting product was purified by recrystallization using ethanol. The coumarin derivatives were obtained in good to excellent isolated yields (83percent–96percent).
84% at 20℃; for 2 h; Reference Example 28; 4- (chloromethyl) -7-hydroxy-2H-chromen-2- one; <n="88"/>Under ice-cooling, ethyl 4-chloroacetoacetate (14.0 g, 85.0 mmol) was dissolved in concentrated sulfuric acid (30 mL) , resorcinol (8.81 g, 80.0 mmol) was added by small portions, and the mixture was stirred at room temperature for 2 hr. The reaction mixture was poured into ice water, and the precipitated solid was collected by filtration, washed with water, and air-dried to give the title compound (14.1 g, yield 84percent) as a beige powder. MS m/z 211 (M + H)+.
84% at 20℃; Cooling with ice Example 30: 7-(3-(4-(3-(6-fluoro-benzisoxazole)-1-piperidyl)-n-propoxy))-4-hydroxymethyl-2H-benzopyran-2-one (30) [0067] The target compound was synthesized according to Scheme 3. 1) 50 ml of concentrated sulfuric acid was stirred in an ice bath, to which was added 5.5 g of resorcinol and added 8 g of 4-chloro ethyl acetoacetate dropwsie. The solution turned yellowish and turbid slowly. The reaction was performed at room temperature overnight. The reaction liquid was poured into ice/water mixture, and a lot of white solid was precipitated, which was filtrated. The cake was washed with water. The cake was recrystallized with 40percent ethanol to give 7.5 g of white crystal. Melting point: 183-185°C, yield: 84percent.
84% at 20℃; 1) 50 ml of concentrated sulfuric acid was stirred in an ice bath, to which was added 5.5 g of resorcinol and added 8 g of 4-chloro ethyl acetoacetate dropwise. The solution turned yellowish and turbid slowly. The reaction was performed at room temperature overnight. The reaction liquid was poured into ice/water mixture, and a lot of white solid was precipitated, which was filtrated. The cake was washed with water. The cake was recrystallized with 40percent ethanol to give 7.5 g of white crystal. Melting point: 183-185° C., yield: 84percent.
84% With nanosilica molybdic acid 2 In neat (no solvent) at 80℃; for 0.333333 h; Green chemistry General procedure: In a general experimental procedure, β-ketoester 3(1 mmol) was added to a mixture of substituted phenol4 (1 mmol) and SMA NPs 2 (5 molpercent) in a solvent-freetube. The reaction mixture was stirred in a preheated oilbath (80 °C). After the completion of the reaction, the precipitateobtained was extracted with ethyl acetate, washedwith water (3 × 10 ml) and dried to obtain the product.The remaining insoluble solid catalyst in aqueous phasewas separated by filtration, washed with ethyl acetate
75.6% at 0 - 20℃; for 2 h; Intermediate 88: 4-(Chloromethyl)-7-hydroxy-2tf-chromen-2-oneTo a 250 mL RB flask fitted with magnetic stirrer was charged with Ethyl 4- chloroacetoacetate (22.12 g, 134.9 mmol) was dissolved in concentrated sulfuric acid (48 mL) at 0 , and resorcinol (14.0 g, 127.3 mmol) was ad ded portionwise. The mixture was stirred at room temperature for 2 h. The reaction mixture was poured into ice-water, and the resulting solid was collected by filtration, washed with water, and dried to give 4- (chloromethyl)-7-hydroxy-2H-chromen-2-one (21.0 g, 75.6 percent) as a beige solid.
70% at 65℃; for 5 h; A resorcinol (5.5 g, 0.05 mol)Ethyl 4-chloroacetoacetate (9.9 g, 0.06 mol)Anhydrous bismuth chloride (0.8 g, 2.5 mmol)Mixed in a 100 mL round bottom flask,It was heated in the absence of a solvent The reaction was stirred 65 , 5h,After the reaction,Add 16 mL of 50percent aqueous ethanol solution,Continue to stir at 65 ° C for 10 min,Cooled to room temperature, suction filtration, solid parts washed to neutral,Dried to give 7.45 g of a white solid,Is 4-chloromethyl-7-hydroxycoumarin, the yield of 70percent.
67% With sulfuric acid In acetic acid at 60℃; for 4 h; Cooling with ice M-bisphenol (27.5 g, 250 mmol) was placed in acetic acid (60mL) heated to 50 ° C for use, ethyl 4-chloroacetoacetate(20.5 g, 125 mmol) was dissolved in acetic acid (20 mL), cooled in an ice-water bath, concentrated sulfuric acid (10 mL) was slowly added, then, the acetic acid solution of m-bisphenol was added to the ice-water bath, stirred at room temperature for 1 hour, and then reacted at 60 deg. C for 3 hours. After completion of the reaction, water (300 mL) was added and the mixture was stirred at room temperature for 1 hour. The resulting white solid was suction-washed three times with water (100 mL) and dried to give the product (17.6g, 67percent yield).
60% With methanesulfonic acid In toluene at 80 - 110℃; Resorcinol (2 g, 18.18 mmol) was dissolved in 50 mL of hottoluene (80 oC). Chloroacetoacetate (3.5 g, 18.18 mmol, 3 mL)was added and allowed to dissolve. The solution was heated to110 oC and methanesulfonic acid (1 mL) was added. After 1 h,the hot toluene containing the product was decanted. The product crystallized uponcooling, it was filtered and dried under vacuum. The resulting gum from thedecanting step was re-suspended in toluene, heated and decanted. The procedurewas repeated until most of product 5 was recovered as a light brown solid (2.3 g,60percent yield).
45.7% at 0℃; for 2 h; 4-Chloromethyl-7-hydroxy-2H-chromen-2-oneResorcinol (7.0 g, 63.6 mmol), ethyl 4-chloroacetoacetate (9.5 ml, 69.9 mmol) and 104 ml of 96percent sulphuric acid were stirred for 2 hours at 0 0C. The reaction mixture was poured into ice water (200 ml) and extracted with ethyl acetate. The organic layers were collected, washed with NaHCO3 10percent aqueous solution, then with water, dried over sodium sulphate and evaporated under vacuum. The resulting oil was purified by column chromatography on silica gel (eluant CHCl3/AcOEt 7.5/2.5 v/v) yielding 5.22 g (45.7percent) of a white solid used without any further purification for the next step synthesis.1H-NMR (Acetone-d6) δ: 9.50 (s, IH, exchanges with D2O), 7.73 (d, IH5 J=8.8), 6.91(dd, IH, J=8.8, J=2.5), 6.80 (d, IH, J=2.5), 6.40 (s, IH), 4.92 (s, 2H).
23.4 g at 40 - 50℃; for 2 h; Step 1: 4-Chloromethyl-7-hydroxy-chromen-2-one [0239] 33.5 g (304 mmol) resorcine is dissolved in 105 ml acetic acid at 40° C. 25.0 g (152 mmol) 4-chloroacetic acid ethylester is added and the funnel is rinsed with 10 ml acetic acid. Then, 26.8 g (274 mmol) conc. H2SO4 (97percent) is added and the funnel is rinsed with 10 ml acetic acid. The mixture is heated to 50° C. for approx. 2 h. After full conversion, 330 ml water is added. The reaction mixture is cooled to r.t. and the suspension is stirred overnight. The product is filtered off, washed with water (2×50 ml) and dried. Yield: 23.4 g; Rf=0.42 (silica gel, PE/EtOAc=6/4); Mass spectrum (ESI): m/z=211 [M+H]+.
23.4 g With sulfuric acid In acetic acid at 40 - 50℃; for 0.2 h; Step 1:
4-chloromethyl-7-hydroxy-chromen-2-one
33.5 g (304 mmol) resorcine is dissolved in 105 ml acetic acid at 40 °C. 25.0 g (152 mmol) 4-chloroacetic acid ethylester is added and the funnel is rinsed with 10 ml acetic acid. Then, 26.8 g (274 mmol) cone. H2S04(97percent) is added and the funnel is rinsed with 10 ml acetic acid. The mixture is heated to 50 °C for approx. 2 h. After full conversion, 330 ml water is added. The reaction mixture is cooled to r.t. and the suspension is stirred overnight. The product is filtered off, washed with water (2 x 50 ml) and dried. Yield: 23.4 g; Rf= 0.42 (silica gel, PE/EtOAc = 6/4); Mass spectrum (ESI): m/z = 21 1 [M + H]+
5.6 g at 0℃; for 2 h; 4-chloroacetoacetate (4.25 ml, 31.43 mmol) was dissolved in 20 ml of concentrated sulfuric acid at 0 ° C, The resulting pale yellow viscous solution was placed in an ice bath and cooled to about _5 ° C, Resorcinol (3.15 g, 28.57 mmol) was added in portions, Control the internal temperature below 0 ° C, plus complete, room temperature stirring 2 h, The reaction solution was poured into 50 ml of ice water and precipitated as a white solid. The filtrate was filtered, washed with water (5 ml x2) and dried to obtain 5.6 g of a white solid and 82percent of the crude product. [0085] Take the crude product (2 g, 9.50 mmol) in 200 ml single-necked flask and add IN NaOH solution (100 ml). The solution immediately became concentrated yellow. The solution was heated in an oil bath for 2 h. Finished, cooled to room temperature, with concentrated sulfuric acid regulation PH to 2-3, the resulting solution was extracted with ethyl acetate (30 ml X4). The organic phases were combined, washed with saturated brine (20 ml X2), dried over anhydrous sodium sulfate, filtered and the filtrate was evaporated under reduced pressure. Brown columnar crystals 1.3 g, crude yield 71.2percent.(1 g, 5.20 mmol) was suspended in 10 ml of methanol, 0.5 ml of concentrated sulfuric acid was added dropwise, and the reaction was refluxed for about 4 hours. After completion of the reaction, the methanol was distilled off under reduced pressure and the residual liquid was poured into 30 ml The mixture was extracted with ethyl acetate (20 ml x3) and the organic phases were combined, washed with saturated sodium bicarbonate solution (15 ml X), washed with saturated brine (15 ml X), dried over anhydrous sodium sulfate, filtered, The filtrate was evaporated under reduced pressure to give a yellowish brown oil which was purified by column chromatography (petroleum ether / ethyl acetate, 80: 20, v / v) to give 0.75 g of a pale yellow solid in 70percent yield.
0.188 g at 0℃; Cooling with ice Under the condition of the ice, will be 1.10 g resorcinol is dissolved in concentrated sulfuric acid in the amount of drying, stirring to completely dissolve, slowly dropping 1.62 ml of 4 - chloro acetyl ethyl acetate, its temperature is 0 °C, after dropping, is moved to the reaction at room temperature, stirring overnight, the reaction [...] poured into ice water, stirring, to be solid after fully depositing, filtering and drying, to obtain 0.188 g intermediate 1, name for 4 - chloro methyl -7 - hydroxy coumarin

Reference: [1] Tetrahedron Letters, 2005, vol. 46, # 36, p. 6119 - 6121
[2] Journal of Organic Chemistry, 2008, vol. 73, # 1, p. 287 - 290
[3] Synthetic Communications, 2008, vol. 38, # 13, p. 2082 - 2088
[4] Journal of Chemical Research, 2008, # 4, p. 232 - 234
[5] Synthetic Communications, 2010, vol. 40, # 22, p. 3358 - 3364
[6] Tetrahedron Letters, 2002, vol. 43, # 50, p. 9195 - 9197
[7] Synthetic Communications, 2004, vol. 34, # 21, p. 3997 - 4003
[8] Tetrahedron Letters, 2006, vol. 47, # 19, p. 3279 - 3281
[9] Catalysis Communications, 2011, vol. 14, # 1, p. 62 - 67
[10] Research on Chemical Intermediates, 2016, vol. 42, # 6, p. 6089 - 6103
[11] Chinese Chemical Letters, 2017, vol. 28, # 1, p. 75 - 82
[12] Heterocycles, 2007, vol. 71, # 3, p. 677 - 682
[13] RSC Advances, 2016, vol. 6, # 24, p. 20269 - 20275
[14] Journal of the Chinese Chemical Society, 2014, vol. 61, # 2, p. 213 - 216
[15] Monatshefte fur Chemie, 2007, vol. 138, # 10, p. 997 - 999
[16] Journal of Materials Chemistry B, 2018, vol. 6, # 17, p. 2547 - 2556
[17] European Journal of Organic Chemistry, 2007, # 26, p. 4301 - 4304
[18] Patent: WO2008/1931, 2008, A2, . Location in patent: Page/Page column 86-87
[19] Journal of Medicinal Chemistry, 2012, vol. 55, # 4, p. 1538 - 1552
[20] Patent: EP2698369, 2014, A1, . Location in patent: Paragraph 0025; 0067
[21] Patent: US2014/113911, 2014, A1, . Location in patent: Paragraph 0066; 0204
[22] Journal of the Iranian Chemical Society, 2017, vol. 14, # 3, p. 655 - 663
[23] MedChemComm, 2014, vol. 5, # 5, p. 650 - 654
[24] Indian Journal of Chemistry - Section B Organic and Medicinal Chemistry, 2009, vol. 48, # 1, p. 120 - 123
[25] Bioorganic and Medicinal Chemistry Letters, 2018, vol. 28, # 18, p. 3050 - 3056
[26] Journal of Medicinal Chemistry, 2009, vol. 52, # 21, p. 6685 - 6706
[27] Beilstein Journal of Organic Chemistry, 2013, vol. 9, p. 254 - 259
[28] Chemical Communications, 2016, vol. 52, # 34, p. 5852 - 5855
[29] Patent: WO2012/11125, 2012, A1, . Location in patent: Page/Page column 136-137
[30] Antiviral Research, 2015, vol. 118, p. 103 - 109
[31] Molecules, 2013, vol. 18, # 5, p. 6057 - 6091
[32] Chemical Communications, 2016, vol. 52, # 85, p. 12574 - 12577
[33] Patent: CN107089963, 2017, A, . Location in patent: Paragraph 0019; 0031; 0032; 0033; 0034
[34] Patent: CN104250239, 2016, B, . Location in patent: Paragraph 0289; 0290; 0291
[35] Bioorganic and Medicinal Chemistry, 2010, vol. 18, # 1, p. 358 - 365
[36] Macromolecules, 2013, vol. 46, # 13, p. 5133 - 5140
[37] Farmaco, 1995, vol. 50, # 2, p. 125 - 130
[38] Journal of Medicinal Chemistry, 2013, vol. 56, # 11, p. 4671 - 4690
[39] Synthesis (Germany), 2018, vol. 50, # 4, p. 846 - 852
[40] Synthetic Communications, 2006, vol. 36, # 20, p. 2949 - 2956
[41] Heterocycles, 1995, vol. 41, # 4, p. 647 - 650
[42] Journal of the American Chemical Society, 2012, vol. 134, # 35, p. 14310 - 14313
[43] Patent: WO2006/102958, 2006, A1, . Location in patent: Page/Page column 29
[44] Journal of Medicinal Chemistry, 2004, vol. 47, # 27, p. 6792 - 6803
[45] Photochemistry and photobiology, 1993, vol. 58, # 4, p. 486 - 491
[46] Chemical Research in Toxicology, 1999, vol. 12, # 7, p. 555 - 559
[47] Organic Letters, 2005, vol. 7, # 25, p. 5565 - 5568
[48] Tetrahedron, 2007, vol. 63, # 6, p. 1353 - 1359
[49] Journal of Medicinal Chemistry, 2007, vol. 50, # 23, p. 5848 - 5852
[50] Patent: US5362628, 1994, A,
[51] Patent: US5576424, 1996, A,
[52] Bioorganic and Medicinal Chemistry Letters, 2010, vol. 20, # 1, p. 272 - 275
[53] Journal of Heterocyclic Chemistry, 2011, vol. 48, # 6, p. 1251 - 1257
[54] Patent: WO2012/111849, 2012, A1, . Location in patent: Page/Page column 141-142
[55] European Journal of Medicinal Chemistry, 2013, vol. 62, p. 11 - 19
[56] Patent: US2015/87829, 2015, A1, . Location in patent: Paragraph 0239
[57] Patent: WO2015/44073, 2015, A1, . Location in patent: Page/Page column 63
[58] Bioorganic and Medicinal Chemistry, 2015, vol. 23, # 23, p. 7448 - 7463
[59] Journal of Enzyme Inhibition and Medicinal Chemistry, 2016, vol. 31, # 5, p. 760 - 772
[60] Patent: CN105566267, 2016, A, . Location in patent: Paragraph 0092; 0094; 0095
[61] Bioorganic and Medicinal Chemistry Letters, 2016, vol. 26, # 23, p. 5732 - 5735
[62] Journal of Fluorescence, 2016, vol. 26, # 6, p. 2177 - 2185
[63] Patent: CN105566263, 2016, A, . Location in patent: Paragraph 0081-0085
[64] Patent: CN107298686, 2017, A, . Location in patent: Paragraph 0087; 0088
[65] New Journal of Chemistry, 2017, vol. 41, # 15, p. 7531 - 7543
[66] Patent: CN105017242, 2018, B, . Location in patent: Paragraph 0077-0080
  • 9
  • [ 638-07-3 ]
  • [ 109-73-9 ]
  • [ 5349-24-6 ]
YieldReaction ConditionsOperation in experiment
99% With 4-toluenesulfonyl azide In tetrahydrofuran at 20 - 25℃; General procedure: To a solution of the corresponding β-keto ester 1 (1.0 mmol) and TsN3 (197 mg, 1.0 mmol) in THF (2.0 mL) under stirring at 25 °C was added the amine (1.1 mmol). Then the reaction mixture was stirred at room temperature until consumption of the starting material (monitored by TLC: 30 min to 29 h, see Table 1 and Scheme 4). Next, the mixture was diluted in 5 mL of CH2Cl2 and concentrated under reduced pressure. After complete removal of the solvent, the residue was triturated in ethyl ether and the resulting mixture was again concentrated under reduced pressure. The final solid residue was repeatedly triturated with hexane (for amides 3 and 4) or a 9:1 hexane/CH2Cl2 mixture (for amides 5 and 6) to separate out the insoluble TsNH2 by decantation. The resulting supernatants were filtered and concentrated under reduced pressure to give the known amides25 3-6 as oils with high degree of purity. Alternatively, further purification through column chromatography on silica gel using gradient mixtures of hexane/EtOAc as eluent was employed to furnish pure products in 80-99percent yield.
Reference: [1] Tetrahedron, 2017, vol. 73, # 31, p. 4549 - 4559
  • 10
  • [ 141-97-9 ]
  • [ 638-07-3 ]
YieldReaction ConditionsOperation in experiment
100%
Stage #1: With n-butyllithium; diisopropylamine In tetrahydrofuran; hexane at 0℃; for 1 h; Inert atmosphere
Stage #2: With methyl chlorosulfate In tetrahydrofuran; hexane at -78℃; for 0.5 h; Inert atmosphere
General procedure: To a solution of diisopropylamine (340 µL, 2.4 eq.) in dry THF (2 mL) in a flame dried round bottom flask under argon at 0 °C was added n-butyllithium (1.4 mL, 1.6 M in hexanes, 2.2 eq.), and the reaction mixture was stirred at this temperature for 15 minutes. At the same temperature a solution of dicarbonyl compound (3n) (1 mmol) in THF (2 mL) was slowly added. After 1 hour of stirring at 0 °C the solution was cooled to -78 °C and methyl chlorosulfate (110 µL, 1.2 eq.) was then added. After stirring at –78 °C for 30 minutes, the reaction was quenched with saturated ammonium chloride aqueous solution (5 mL). The mixture was then extracted with dichloromethane (3 x 5 mL), the combined organic phases were dried with anhydrous magnesium sulfate and the solvent evaporated affording the desired 4-chloro dicarbonyl compound 5n.
Reference: [1] Tetrahedron Letters, 2018, vol. 59, # 13, p. 1233 - 1238
  • 11
  • [ 41295-64-1 ]
  • [ 64-17-5 ]
  • [ 638-07-3 ]
Reference: [1] Patent: EP2518043, 2012, A1, . Location in patent: Page/Page column 10; 11
[2] Patent: CN103787883, 2016, B, . Location in patent: Paragraph 0026
  • 12
  • [ 6082-74-2 ]
  • [ 638-07-3 ]
Reference: [1] Russian Journal of Organic Chemistry, 2004, vol. 40, # 7, p. 924 - 927
  • 13
  • [ 591-78-6 ]
  • [ 6082-74-2 ]
  • [ 638-07-3 ]
Reference: [1] Russian Journal of Organic Chemistry, 2004, vol. 40, # 7, p. 924 - 927
  • 14
  • [ 591-78-6 ]
  • [ 6082-74-2 ]
  • [ 638-07-3 ]
  • [ 141-97-9 ]
Reference: [1] Russian Journal of Organic Chemistry, 2004, vol. 40, # 7, p. 924 - 927
  • 15
  • [ 6082-74-2 ]
  • [ 100-52-7 ]
  • [ 638-07-3 ]
  • [ 1503-99-7 ]
Reference: [1] Russian Journal of Organic Chemistry, 2004, vol. 40, # 7, p. 924 - 927
  • 16
  • [ 67-56-1 ]
  • [ 638-07-3 ]
  • [ 66762-68-3 ]
YieldReaction ConditionsOperation in experiment
15.1% With sodium hydride In N,N-dimethyl-formamide at 0 - 20℃; for 16 h; NaH (7.71 g, 192.7 mmol) was suspended in DMF (110 mL). The solution of MeOH (2.64 g, 82.5 mmol) in DMF (55 mL) was added drop wise at 0° C. After that, the mixture was stirred at 0° C. for 30 min. The solution of compound 1 (9.54 g, 57.9 mmol) in DMF (55 mL) was added dropwise at 0° C., then the reaction mixture was allowed to warm slowly to RT and stirred for 16 h. The reaction mixture was diluted with 1N HCl (400 mL), extracted with MTBE (250 mL×3). The combined organic layer was washed with H2O (200 mL×5), dried over Na2SO4, filtered, concentrated, purified by silica gel column (PE:EA=10:1) afforded ethyl 4-methoxy-3-oxobutanoate (1.4 g, 15.1percent) as light yellow oil.
74 g With sodium hydride In tetrahydrofuran; mineral oil at 15 - 25℃; for 9 h; Inert atmosphere In 2L pre-reaction bottle by adding 250 ml tetrahydrofuran, under the protection of argon, are started to stir, the oven 25 °C, the inner temperature 15-25 °C added in batches under the condition of 55g (1.35mol) sodium hydride (containing 40percent mineral oil), continue adding tetrahydrofuran after adding 450 ml; the inner temperature 20 °C slowly dropping under the condition of 30g methanol and 100g (0.61mol) 4-chloro-acetyl-acetic acid ethyl ester of mixed solution, adjusting the stirring rate in 310 R/min the left and the right, about 4h finish; to temperature rise within 20-25 °C stirring 5h, TLC detection reaction finishes; cooling system, can be seen and the color is yellowish solution a large number of solid suspended, the inner temperature 11 °C add under the condition of 200 ml tetrahydrofuran, to be oven to -5 ° C slowly adding 100 ml molar concentration is 2mol/L hydrochloric acid solution, maintained in the oven 0 °C the following, about 20 min after adding, at this time the pH of the reaction system 11, in the process of dropping the solid significant increase in the reaction bottle, immediately after adding filtering, the filter cake is a white solid, cake obtained drying filters 92g solid; the resulting solid is added to the 770g in ethyl acetate, the temperature of the reaction system to 0 °C, slow instillment mole concentration is 6mol/L hydrochloric acid solution, maintaining the temperature of the reaction system is not variable, the dropping white solid gradually disappear in the process, the reaction system is adjusted to pH 3, reaction liquid contains a small amount of inorganic salt white solid, filtering the reaction solution, the filtrate layer, the organic phase is separated, the organic phase by adding 5g activated carbon, in the 30 °C under the condition of stirring 1h, filtered, filtrate in the 35 °C ethyl acetate under the condition of the vacuum distillation to the colorless liquid 74g, HPLC checking the purity of 99.6percent.
Reference: [1] Patent: JP2015/214548, 2015, A, . Location in patent: Paragraph 0992; 0993
[2] Patent: CN105481694, 2016, A, . Location in patent: Paragraph 0015; 0016; 0017; 0018; 0019; 0020; 0021 - 0023
  • 17
  • [ 638-07-3 ]
  • [ 66762-68-3 ]
Reference: [1] Patent: US2006/189664, 2006, A1, . Location in patent: Page/Page column 8
[2] Patent: US5767131, 1998, A,
  • 18
  • [ 638-07-3 ]
  • [ 100-51-6 ]
  • [ 67354-34-1 ]
YieldReaction ConditionsOperation in experiment
90%
Stage #1: With sodium tert-pentoxide In 1,3-dimethyl-2-imidazolidinone at 20 - 40℃; for 2 h; Inert atmosphere
Stage #2: at 0 - 20℃; for 5 h; Cooling with ice
Example 2 First StepA solution of benzyl alcohol (0.66 g, 6.1 mmol) in DMI (3 ml) was added to a suspension of sodium tert-pentoxide (1.67 g, 15.2 mmol) in DMI (4 ml) at room temperature under a nitrogen atmosphere, and the mixture was stirred at 40° C. for 2 hours. This reaction solution was cooled in an ice bath, and a solution of Compound 2A (1.10 g, 6.68 mmol) in DMI (3 ml) was added dropwise at 0 to 10° C. The reaction solution was stirred at 0 to 5° C. for 2 hours, and at room temperature for 3 hours, and 2N hydrochloric acid (15 ml) was added, followed by extraction with ethyl acetate two times. The combined extracts were washed sequentially with water, saturated sodium bicarbonate water, water and saturated sodium chloride water, and then dried with anhydrous sodium sulfate. The solvent was distilled off, and the resulting oil product was purified by silica gel column chromatography (n-hexane-ethyl acetate 4:1, v/v) to obtain 1.29 g (yield 90percent) of Compound 2B as an oil product.1H-NMR (CDCl3) δ: 1.25 (3H, t, J=7.2 Hz), 3.54 (2H, s), 4.14 (2H, s), 4.17 (2H, q, J=7.2 Hz), 4.59 (2H, s), 7.28-7.40 (5H, m).
90%
Stage #1: at 40℃; for 2 h; Inert atmosphere
Stage #2: at 0 - 20℃; for 5 h;
A DMI (3 ml) solution of benzyl alcohol (0.66 g, 6.1 mmol) was added to a DMI (4 ml) suspension of sodium tert-pentoxide (1.67 g, 15.2 mmol) at room temperature in a nitrogen atmosphere, and the mixture was stirred at 40°C for 2 hours. This reaction solution was cooled in an ice bath, and a DMI (3 ml) solution of compound 2A (1.10 g, 6.68 mmol) was added dropwise thereto at 0-10°C. The reaction solution was stirred at 0-5°C for 2 hours and at room temperature for 3 hours, and 2 N hydrochloric acid (15 ml) was then added thereto, followed by extraction two times with ethyl acetate. The combined extracts were washed with water, a saturated aqueous solution of sodium bicarbonate, water, and saturated saline in this order and then dried over anhydrous sodium sulfate. The solvent was distilled off, and the obtained oil was purified by silica gel column chromatography (n-hexane-ethyl acetate: 4:1, v/v) to obtain 1.29 g (yield: 90percent) of compound 2B as an oil. 1H-NMR (CDCl3) δ: 1.25 (3H, t, J = 7.2 Hz), 3.54 (2H, s), 4.14 (2H, s), 4.17 (2H, q, J = 7.2Hz), 4.59 (2H, s), 7.28-7.40 (5H, m).
85%
Stage #1: With sodium hydride In tetrahydrofuran; mineral oil at 10℃; for 1 h; Cooling with ice
Stage #2: at 20℃; for 0.666667 h;
Benzyl alcohol (6.90 g, 63.8 mmol) was added dropwise to a stirred suspension of sodium hydride (60percent, 5.37 g, 134 mmol) in dry THE (50 mL) under ice bath to keep the temperature below 10°C. The mixture was stirred for an hour. Ethyl 4-chloro-3-oxobutanoate (10.0 g,60.8 mmcl) was added dropwise within 40 mm at rt, and the resulting mixture was stirred overnight. The reaction mixture was carefully added to HCI (5percent, 100 mL) at 5 °C and then extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na2504, filtered and concentrated. The crude was purified by column chromatography (PE/EtOAc 10/1) to give the title compound (12.1 g, yield 85percent) as ayellow oil.LCMS: [mobile phase: 5-95percent CH3CN in 2.5 minI, Rt 1.55 mm, MS Calcd: 236; MS Found:237 [M+H].1H NMR (300 MHz, CDCI3): 6 7.38-7.30 (m, 5H), 4.59 (s, 2H), 4.22-4.10 (m, 4H), 3.53 (s,2H), 1.25 Ct, J = 7.2 Hz, 3H).
78%
Stage #1: With sodium tert-pentoxide In tetrahydrofuran at 40 - 45℃; for 2 h;
Stage #2: at 0 - 20℃; for 3 h;
Benzyl alcohol (95 mL, 1 eq) was added to a mixture of sodium tert- pentoxide (250 g, 2.5 eq) in tetrahydrofuran (750 mL, 5V) at 20-25°C. The reaction mass was heated to 40-45°C and stirred for 2 hrs. The reaction mass was then cooled to 0-5°C and ethyl-4-chloro acetoacetate (XII-C1,150 g, leq) in tetrahydrofuran (750 mL, 5V) was added to it. The reaction mass was then stirred for 3 hrs at room temperature. After reaction completion, the reaction mass was cooled to 0-5 °C and pH was adjusted to ~2 using 20percent hydrochloric acid. The reaction mass was extracted twice with ethyl acetate (2X5V). Organic layers were combined, washed with saturated bicarbonate solution (1X10V) followed by water (5V) and saturated brine solution (5V). Organic layer was dried over anhydrous sodium sulfate and distilled off the solvent completely to get the crude compound as brown oily thick liquid. The crude compound was purified by silica gel column chromatography (Eluent: EtOAc-hexane). The pure fractions were concentrated under vacuum to afford the title compound as pale yellow liquid (168 g, 78percent yield). MS (ES): m/z 237 (M+H)+.
69% With sodium hydride In tetrahydrofuran at 40℃; for 2 h; Ethyl 4-(benzyloxy)-3-oxobutanoate
Sodium hydride (145 g, 6.06 mol) was suspended in tetrahydrofuran (3.0 L).
Benzyl alcohol (328 g, 3.03 mol) and ethyl 4-chloro-3-oxobutanoate (500 g, 3.03 mol) were added to the suspension and the reaction mixture was stirred at 40° C. for 2 h.
The reaction mixture was cooled to room temperature, quenched with ice water and extracted with ethyl acetate.
The combined organic layers were dried over anhydrous sodium sulphate and concentrated under vacuum to afford 500 g (69percent) of Ethyl 4-(benzyloxy)-3-oxobutanoate.
1H NMR (400 MHz, CDCl3): δ1.29 (t, 3H), 3.53 (s, 2H), 4.22 (q, 2H), 4.22 (s, 2H), 4.60 (s, 2H), 7.38 (m, 5H).
20%
Stage #1: With sodium hydride In toluene at 20℃; for 1.66667 h;
Stage #2: at 20℃; for 18 h;
A suspension of sodium hydride (60percent in oil) in tolene (200 mL) was mechanically stirred at room temperature under argon and treated dropwise with benzyl alcohol in toluene (40 mL) over 40 minutes. The mixture was stirred for 1 hour, treated with ethyl 4-chloroacetate, and then stirred over 18 hours at room temperature. [0744] At end of the reaction, citric acid; was added and layers were separated. The organic layers was washed water, dried over MgSO4 and concentrated to give the the title compound (7.2 g, 20percent). MS(ES) m/z 237 (M+H)+

Reference: [1] Journal of Medicinal Chemistry, 2016, vol. 59, # 10, p. 5051 - 5062
[2] Journal of Organic Chemistry, 2011, vol. 76, # 22, p. 9444 - 9451
[3] Angewandte Chemie - International Edition, 2006, vol. 45, # 38, p. 6376 - 6380
[4] Patent: US2012/22251, 2012, A1, . Location in patent: Page/Page column 25
[5] Patent: EP2602260, 2013, A1, . Location in patent: Paragraph 0127
[6] Patent: WO2017/12576, 2017, A1, . Location in patent: Page/Page column 120
[7] Synlett, 2000, # 6, p. 844 - 846
[8] Organic and Biomolecular Chemistry, 2009, vol. 7, # 5, p. 962 - 975
[9] Patent: WO2015/110897, 2015, A2, . Location in patent: Paragraph 00138-00140
[10] Patent: WO2015/110897, 2015, A3, . Location in patent: Paragraph 00138-00140
[11] Patent: US2008/312255, 2008, A1, . Location in patent: Page/Page column 91
[12] New Journal of Chemistry, 2016, vol. 40, # 10, p. 8786 - 8808
[13] Tetrahedron Asymmetry, 2001, vol. 12, # 12, p. 1713 - 1718
[14] European Journal of Medicinal Chemistry, 2015, vol. 97, p. 42 - 54
[15] Patent: US2004/19190, 2004, A1, . Location in patent: Page 38
[16] Monatshefte fuer Chemie, 1989, vol. 120, p. 891 - 898
[17] Synthesis, 1995, # 8, p. 1014 - 1018
[18] Organic Letters, 2010, vol. 12, # 3, p. 416 - 419
[19] Patent: WO2011/76878, 2011, A1, . Location in patent: Page/Page column 52-53
[20] Patent: WO2015/164308, 2015, A1, . Location in patent: Page/Page column 90
[21] ChemMedChem, 2018, vol. 13, # 16, p. 1658 - 1663
  • 19
  • [ 638-07-3 ]
  • [ 88150-42-9 ]
Reference: [1] Journal of Medicinal Chemistry, 1986, vol. 29, # 9, p. 1696 - 1702
[2] Journal of Medicinal Chemistry, 1986, vol. 29, # 9, p. 1696 - 1702
  • 20
  • [ 638-07-3 ]
  • [ 10488-69-4 ]
YieldReaction ConditionsOperation in experiment
98% With hydrogen In ethanol at 95 - 105℃; for 0.5 - 8 h; Example 1 (Comparative . A 600ml stainless steel Parr reactor was charged with ethanol (340ml) and ethyl-4-chloroacetoacetate (53g). The reactor agitator was started and the speed set to 600rpm. The reactor was pressurised using nitrogen to 7 bar and stirring continued for 5 minutes. After 5 minutes the reactor was slowly vented to ambient pressure, the pressurisation/depressurisation cycle was repeated for a total of five times to ensure complete removal of dissolved oxygen. At the end of the last cycle the reactor set-point temperature was adjusted to 95°C. (R)-[RuCI2 (BINAP)] ncatalyst was accurately weighed (23mg) into a catalyst transfer vessel and the vessel then purged using nitrogen for 5 minutes. The catalyst was flushed from the transfer vessel using deoxygenated solvent into a 100ml stainless steel injection bomb which was attached to the Parr reactor. When the Parr reactor temperature was between 95°C and 100°C the injection bomb was pressurised to 100bar using hydrogen. Appropriate valves were then opened to transfer the catalyst mixture and hydrogen into the reactor. The contents of the reactor were stirred at 600rpm for 30 minutes before being cooled to less than 30°C. The reactor was then slowly vented to ambient pressure. The reactor contents were transferred into a 1L rotary film evaporator flask and the mixture evaporated to constant weight by application of vacuum and by using a heated water bath. The residue was subjected to pot to pot distillation under vacuum to afford a clear colourless oily liquid product of ethyl (S)- (-)-4-chloro-3-hydroxybutyrate in >98percent yield, >98percent purity and 94percent enantiomeric excess.; Example 2 A feed tank was charged with 3.6L ethanol solvent. The solvent was deoxygenated by pumping it through a spray nozzle whilst pressurising to 7bar with nitrogen and then depressurising through a needle valve at a controlled rate. The pressurisation/depressurisation cycle was repeated three times and the entire process automated using a PLC-based control system. In a similar manner a second feed tank was charged with ethyl-4-chloroacetoacetate (3.6L) and deoxygenated using the same protocol to that described above. The catalyst, (R)- [RuCl2 (BINAP)] (149mg) was charged into a transfer vessel and the vessel purged using nitrogen before transferring the catalyst into the solvent feed tank. The catalyst solution had a concentration of 52.2mg/Kg. The two feed systems were connected to the continuous hydrogenation reactor system via two high-pressure pumps. The continuous hydrogenation reactor system was constructed of Hastalloy 276 and comprised a number of in-line static mixers to give a residence time of between 30 and 35 seconds. The static mixers also ensured good mixing of the process streams and rapid absorption of hydrogen. The reactor system was equipped with a recycle pump and an in-line valve which enabled operation as either a plug flow reactor (PFR, valve closed) or a continuous loop reactor (CLR, valve open). The system was equipped with a gas/liquid separator and the liquid level inside the separator controlled using a differential pressure sensor, which in turn operated an exit flow control valve. The reactor system was controlled using a PLC based control system. The hydrogenation reactor was pressurised using hydrogen and the pressure maintained between 90 and 100 bar by continually feeding hydrogen through a mass flow controller at a rate of 2.7g/h. The reaction liquors passed through a heat exchanger using a pump such that the process temperature was maintained between 102°C and 105°C. The system above was operated as a plug flow reactor. The flow rate of the ethyl-4-chloroacetoacetate was set to 2. 6ml/minute and the flow rate of the catalyst solution set to 8. 9ml/min. These flows gave a process concentration of 30percent w/w and a substrate to catalyst ratio of 20,000 : 1. Over a series of continuous runs, each varying between 4 and 8 hours, the reactor consistently converted >99percent ethyl-4-chloroacetoacetate to (S) -ethyl-4-chloro-3-hydroxybutyrate which was isolated after removing the solvents by evaporation to give a chemical yield of >98percent and an enantiomeric excess of 98- 99percent.; Example 3 The reactor was set up as Example 2, except it was operated as a continuous loop reactor. The flow rate of the ethyl-4-chloroacetoacetate was set to 2. 55ml/minute and the flow rate of the ethanol catalyst solution set to 6. 60ml/min at a catalyst concentration of 45. 8mg/kg. These flows gave a process concentration of 37percent w/w and a substrate to catalyst ratio of 65,000 : 1. Over a series of continuous runs, each varying between 4 and 8 hours, the reactor consistently converted >99percent ethyl-4-chloroacetoacetate to (S) -ethyl-4-chloro-3-hydroxybutyrate which was isolated after removing the solvents by evaporation to give a chemical yield of >98percent and an enantiomeric excess of 98- 99percent.
96% With D-glucose In ethanol at 25℃; for 0.25 h; Microbiological reaction; aq. phosphate buffer General procedure: Cells harvested by centrifugation were washed and resuspended (19 g cell dry weight/l) in a 10 mM potassium phosphate buffer, pH 6.8. Cell suspension (25 ml) was taken in a 150 ml Erlenmeyer flask capped with a cotton plug and pre-incubated in a water-bath shaker at 200 rpm and 25 °C for 15 min after adding glucose (50 g/l). A solution of 0.12 g of COBE in 0.5 ml of ethanol was then added to the reaction flask. Incubation was continued for 15 min [for (R)-CHBE, the reaction time was 1 h] after the addition of COBE after which the reaction mixture was extracted with ethyl acetate (2 .x. 100 ml). The organic layer was dried over anhydrous sodium sulfate and the pure product was obtained by silica gel chromatography using hexane/ethyl acetate (9:1) as a mobile phase. In order to determine the rate of formation of CHBE, a 0.5 ml sample was taken every 5 min and centrifuged (10,000 rpm, 10 min) at 4 °C to remove the cells. The supernatant was extracted with ethyl acetate (1 ml) of which 1 μl was injected into the GC to determine the concentrations of COBE and CHBE.
87.4% With ketoreductase; NADP In acetic acid butyl ester; water for 7 - 18 h; Example 9: Preparation of Ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE from Ethyl 4-chloro- acetoacetate. To a 100 mL vessel connected to an automatic titrater by a pH electrode and a feeding tube for addition of base was charged a solution of glucose (7.5 g) in 100 mM TRIETHANOLAMINE pH 7 buffer (25 ML). To this solution was charged ketoreductase SEQ ID NO: 42 (100 mg); 50 mg GDH SEQ ID NO: 66 and NADP (6.25 mg). Butyl acetate (10 ml) was then charged. Then, ethyl 4-chloroacetoacetate (6 g) in butyl acetate (10 mL) was charged. The pH was maintained at 7 by the automatic titrater by the addition of 4M NaOH (7.5 mL) over 7 hrs. A sample of the reaction mixture was extracted with an equal volume of butyl acetate and the organic layer was analyzed by GC. The analysis showed 99percent conversion of the ethyl 4-CLLLOROACETOACETATE to ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE.Example 11 : Preparation of Ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE from Ethyl 4-chloro- acetoacetate. To a 100 mL vessel connected to an automatic titrater by a pH electrode and a feeding tube for addition of base was charged a solution of glucose (12. g) in water (30 mL). To this solution was charged ketoreductase SEQ ID NO: 42 (100 mg); 50 mg GDH SEQ ID NO: 66 and NADP (6.25 mg). Butyl acetate (10 ML) WAS then charged. Ethyl 4-chloroacetoacetate (10 g) was then charged via syringe pump as follows : 1 mL was charged rapidly and the remainder was then charged at a rate of 1 mL/hr). The pH was maintained at 7 by the automatic titrater by the addition OF 4M NAOH over 18 hours hrs. The stirring was stopped and the phases allowed to separate. The organic layer included some emulsion. The organic layer, including some emulsion, was separated and washed with 10 mL of water. The combined aqueous layers were extracted twice WITH 20 mL of butyl acetate. The organic extracts were combined and rotary evaporated under vacuum to remove water. Additional butyl acetate was added during the evaporation to help remove the water. When the water was removed the butyl acetate solution was decanted from solids in the flask. Evaporation of the solvent under vacuum then gave 8. 85 g of ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE (87.4percent yield) of very good purity.
87.4% With ketoreductase; NADP In acetic acid butyl ester; water for 7 - 18 h; Example 9: Preparation of Ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE from Ethyl 4-chloro- acetoacetate. To a 100 mL vessel connected to an automatic titrater by a pH electrode and a feeding tube for addition of base was charged a solution of glucose (7.5 g) in 100 mM TRIETHANOLAMINE pH 7 buffer (25 ML). To this solution was charged ketoreductase SEQ ID NO: 42 (100 mg); 50 mg GDH SEQ ID NO: 66 and NADP (6.25 mg). Butyl acetate (10 ml) was then charged. Then, ethyl 4-chloroacetoacetate (6 g) in butyl acetate (10 mL) was charged. The pH was maintained at 7 by the automatic titrater by the addition of 4M NaOH (7.5 mL) over 7 hrs. A sample of the reaction mixture was extracted with an equal volume of butyl acetate and the organic layer was analyzed by GC. The analysis showed 99percent conversion of the ethyl 4-CLLLOROACETOACETATE to ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE.Example 11 : Preparation of Ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE from Ethyl 4-chloro- acetoacetate. To a 100 mL vessel connected to an automatic titrater by a pH electrode and a feeding tube for addition of base was charged a solution of glucose (12. g) in water (30 mL). To this solution was charged ketoreductase SEQ ID NO: 42 (100 mg); 50 mg GDH SEQ ID NO: 66 and NADP (6.25 mg). Butyl acetate (10 ML) WAS then charged. Ethyl 4-chloroacetoacetate (10 g) was then charged via syringe pump as follows : 1 mL was charged rapidly and the remainder was then charged at a rate of 1 mL/hr). The pH was maintained at 7 by the automatic titrater by the addition OF 4M NAOH over 18 hours hrs. The stirring was stopped and the phases allowed to separate. The organic layer included some emulsion. The organic layer, including some emulsion, was separated and washed with 10 mL of water. The combined aqueous layers were extracted twice WITH 20 mL of butyl acetate. The organic extracts were combined and rotary evaporated under vacuum to remove water. Additional butyl acetate was added during the evaporation to help remove the water. When the water was removed the butyl acetate solution was decanted from solids in the flask. Evaporation of the solvent under vacuum then gave 8. 85 g of ethyl (S)-4-CHLORO-3-HYDROXYBUTYRATE (87.4percent yield) of very good purity.
50% at 37 - 40℃; for 60 h; Enzymatic reaction; Aqueous phosphate buffer The General Methodology for the Reduction of Substituted β-Ketoesters; 100 mg of each of the compounds in Table 3, entry Nos. 1-10 and their related substituted β-ketoesters were added to a 100 ml flask containing 2 gm of crude extract of Daucus carota, and 50 ml of 0.1 M sodium phosphate buffer pH 6.0 to 7.5 were added. The reaction was incubated in a shaker for 50 to 70 hours for maximum product formation. The product formed was isolated, purified by column chromatography and the structure of the compound was confirmed by spectral data.
97.6 % ee With hydrogen In ethanol at 80℃; for 2 h; In a 150 mL autoclave under argon atmosphere RuCl3 (10.2 mg, 0.049 mmol), (-)-5,5'-bis(diphenylphosphanyl)-2,2,2',2'-tetrafluoro-4,4'-bi[benzo-1,3-dioxolyl] (i.e. (-)-Fluoxphos) (34.2 mg, 0.050 mmol) and ethyl 4-chloro-3-oxobutyrate (0.81 g, 4.9 mmol, approx. 98.5percent) is dissolved in degassed ethanol (30 mL). After flushing the autoclave with argon hydrogenation is carried out during 2 hours at 80 °C and at 4 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and enantiomeric excess (ee) (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 97.6percent.
89.5 % ee With hydrogen In ethanol at 90℃; for 2.3 h; In a 250 mL autoclave in an argon atmosphere [Ru2Cl4(cym)2] (6.7 mg, 0.011 mmol), (-)-5,5'-bis(diphenylphosphanyl)-4,4'-bi[benzo-1,3-dioxolyl] (14.3 mg, 0.023 mmol) and ethyl 4-chloro-3-oxobutyrate (9.10 g, 54.5 mmol, approx. 98.5percent) is dissolved in degassed ethanol (30 mL). After flushing the autoclave with argon hydrogenation is carried out during 2.3 hours at 90 °C and at 30 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 89.5percent.
97.4 % ee With hydrogen In ethanol; dichloromethane at 100℃; for 2.83333 h; In a 1 L autoclave under argon atmosphere [RuI((-)-Fluoxphos)(cym)]I (35 mg, 0.030 mmol, prepared from (-)-Fluoxphos and [Ru2I4(cym)2] as disclosed in WO 00/29370, and ethyl 4-chloro-3-oxobutyrate (33.4 g, 200 mmol, approx. 98.5percent) is dissolved in degassed ethanol (340 mL) and dichloromethane (80 mL). After flushing the autoclave with argon hydrogenation is carried out during 170 minutes at 100 °C and at 22 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 97.4percent.
98.0 % ee With hydrogen In ethanol; dichloromethane at 100℃; for 3 h; In a 1 L autoclave under argon atmosphere [RuCl((-)-Fluoxphos)(cym)]BF4 (23 mg, 0.022 mmol, prepared from (-)-Fluoxphos and [Ru2Cl4(cym)2] and AgBF4 as disclosed in Mashima, K., J. Org. Chem. 1994, 59, 3064-3076, and ethyl 4-chloro-3-oxobutyrate (33.4 g, 200 mmol, approx. 98.5percent) is dissolved in degassed ethanol (340 mL) and dichloromethane (80 mL). After flushing the autoclave with argon hydrogenation is carried out during 180 minutes at 100 °C and at 22 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 98.0percent.
98.1 % ee With hydrogen In ethanol; dichloromethane at 110℃; for 1.16667 h; In a 1 L autoclave under argon atmosphere [Ru2Cl4(cym)2] (5.0 mg, 0.008 mmol, (-)-Fluoxphos (12.1 mg, 0.018 mmol) and ethyl 4-chloro-3-oxobutyrate (27.0 g, 162 mmol, approx. 98.5percent) is dissolved in degassed ethanol (340 mL) and dichloromethane (80 mL). After flushing the autoclave with argon hydrogenation is carried out during 70 minutes at 110 °C and at 22 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 98.1percent.
95.8 % ee With hydrogen In ethanol at 90℃; for 2.3 h; In a 250 mL autoclave in an argon atmosphere bis[(1-isopropyl-4-methylbenzene)dichloro ruthenium] (i.e. [Ru2Cl4(cym)2]) (6.7 mg, 0.011 mmol), (-)-Fluoxphos (16.0 mg, 0.023 mmol) and ethyl 4-chloro-3-oxobutyrate (9.11 g, 54.5 mmol, approx. 98.5percent) is dissolved in degassed ethanol (30 mL). After flushing the autoclave with argon hydrogenation is carried out during 2.3 hours at 90 °C and at 30 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 95.8percent.
80 % ee With hydrogen In ethanol at 80℃; for 3 h; Example 11: (5)-Ethyl4-chloro-3-hydrpxybutyrate; In a 150 mL autoclave under argon atmosphere bis(l-isopropyl-4-methylbenzene)dichloro-ruthenium (7.5 mg, 0.012 mmol), (+)-ligand Ic (14.4 mg, 0.025 mmol) and ethyl 4-chloro-3-oxobutyrate (0.83 g, 5.0 mmol) is dissolved in degassed ethanol (30 mL). After flushingthe autoclave with argon hydrogenation is carried out during 3 h at 80 °C and at 4 barhydrogen pressure. After cooling to room temperature the reaction solution is directlyanalyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column:Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 80percent.
80 % ee With (+)-6-dicyclohexylphosphanyl-2'-diphenylphosphanyl-2-methoxy-1,1'-biphenyl; hydrogen In ethanol at 80℃; for 3 h; In a 150 mL autoclave under argon atmosphere bis(1-isopropyl-4-methylbenzene)dichloro-ruthenium (7.5 mg, 0.012 mmol), (+)-ligand Ic (14.4 mg, 0.025 mmol) and ethyl 4-chloro-3-oxobutyrate (0.83 g, 5.0 mmol) is dissolved in degassed ethanol (30 mL). After flushing the autoclave with argon hydrogenation is carried out during 3 h at 80 °C and at 4 bar hydrogen pressure. After cooling to room temperature the reaction solution is directly analyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column: Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 80percent.
80 % ee With hydrogen In ethanol at 80℃; for 3 h; Example 16: (5)-Ethyl 4-chloro-3-hydroxybutyrate; In a 150 mL autoclave under argon atmosphere bis(l-isopropyl-4-methylbenzene)dichloro-ruthenium (7.5 mg, 0.012 mmol), (+)-ligand Ic (14.4 mg, 0.025 mmol) and ethyl 4-chloro-3-oxobutyrate (0.83 g, 5.0 mmol) is dissolved in degassed ethanol (30 mL). After flushingthe autoclave with argon hydrogenation is carried out during 3 h at 80 °C and at 4 barhydrogen pressure. After cooling to room temperature the reaction solution is directlyanalyzed by GC for conversion (column: HP-101 25 m / 0.2 mm) and ee (column:Lipodex-E 25 m / 0.25 mm). Conversion is 100percent at an ee of 80percent.
98 % ee With hydrogen In tetrahydrofuran; ethanol at 100℃; for 2 h; Examples 3 to 17The reaction of example 1 was carried out with different catalysts and under varying conditions. The specific catalysts, conditions and results are summarized in table 1 in figure 2. Unless specified otherwise in figure 2, the reaction was carried out as described above for example 1. The solvent used in all reactions was EtOH THF. The ratio substrate/catalyst (S/C) was relatively high in most experiments. Thus only low amounts of catalyst are necessary in the inventive reaction. Further, the concentration of the substrate S was selected relatively high to 3 or 4 M. This is equivalent to a concentration of approximately 50percent (w/w). The temperature was 100°C. The pressures were adjusted to relatively low levels of between 15 and 40 bar.The results show, that for almost all catalysts a high absolute yield (conversion) and a high enantiomeric yield (ee) were obtained, although the total amounts of catalyst were very low. Further, the pressure was relatively low, which is advantageous for large scale industrial applications. High yields could be obtained even after relatively short reaction times of between 1.3 and 3 hours (examples 8 to 13). Overall, the examples show that the inventive reaction is efficient and can be carried out with low amounts of catalyst and solvent. The reaction is also energy-efficient, because the reaction time is low, the volume to be heated is small (due to low solvent levels) and the pressure is low.
> 99.9 % ee at 30℃; for 8 h; Enzymatic reaction General procedure: Asymmetric reductions of various carbonyl compounds by the purified enzymes were carried out at 30°C for 8h with mild shaking in a reaction mixture containing 0.1M potassium phosphate buffer (pH 6.5), 1gL−1 substrate, 10mM NADPH, and the purified enzyme of appropriate amount in a total volume of 2mL. In order to determine the absolute configuration of chiral alcohols, the reaction products were extracted with ethyl acetate or hexane and the organic layer was used for analysis. The optical purity of the reaction products were determined by chiral HPLC (HP 1100, Agilent, USA) equipped with Chiralcel OB-H column (4.6mm×250mm; Daicel Chemical Ind. Ltd., Japan) or chiral GC (7890A, Agilent, USA) equipped with FID detector and Chrompack Chirasil-Dex CB chiral capillary column (25m×0.25mm; Varian, USA) [21].
> 99 % ee With D-glucose; nicotinamide adenine dinucleotide phosphate; recombinant Bacillus subtilis-derived glucose dehydrogenase; recombinant Gluconobacter oxydans carbonyl reductase 0525 In aq. phosphate buffer; ethanol at 30℃; for 12 h; Enzymatic reaction General procedure: The enzyme activity toward the reduction ofketones was determined by spectrophotometricallymeasuring the oxidation of NAD(P)H at 340 nm(ε = 6.22mM−1 cm−1) in the presence of an excess amount of ketones. The change in absorbance of NAD(P)H was monitored at 340 nm using an ultraviolet-visiblespectrophotometer with a temperature-controlledcuvette holder (Shimadzu Co., Kyoto, Japan). One unit(U) activity was defined as the amount of enzymesrequired to catalyze the oxidation of 1 μmol NAD(P)Hper minute at 30 °C. The data were expressed as U/mgof protein. The standard reaction mixture contained100mM phosphate buffer (pH 7.0), 0.1mM NAD(P)H,5 mM substrate, and enzyme solvent in a total volumeof 1 mL. The reaction was initiated by adding 20 μLsolvent containing 10 μg–40 μg of enzymes. Blankswithout the enzyme were carried out for each substrate,and data were collected in triplicate. Protein concentrationswere determined with the bicinchoninic acid assayusing bovine serum albumin as a standard.NAD(P)H or NADH was assayed under standardreaction conditions for the study of coenzyme dependence.A range of substrates from 1 to 20 mM concentrationwas assayed under the standard reactionconditions for the study of kinetics. Apparent values ofMichaelis constant (Km) and kcat were calculated by fittingthe data into Michaelis–Menten equation using theSigmaPlot (Systat Software Inc., San Jose, CA, USA).All reactions followed Michaelis–Menten-type kinetics.Enantioselective reduction of ketones. The enantioselectivityof the enzymes was determined byexamining the reduction of aryl ketones, ethyl 4-chloroacetoacetate(COBE), and ethyl 2-oxo-4-phenylbutyrate(OPBE) using an NAD(P)H regeneration system consistingof BsGDH and glucose. The general procedurewas as follows: D-glucose (0.5percent), recombinant BsGDH(10 U), NAD(P)+ (0.1 mM), the recombinant cell(30 g L−1, wet weight), and ketone solvated in ethanol[1 g L−1, 10percent(v/v)] were mixed in a potassium phosphatebuffer (10 mL, 100 mM, pH 7.0). The mixturewas shaken at 30 °C for 12 h.22) Upon termination ofthe reaction, each sample was extracted twice withequivalent ethyl acetate. The organic layer wasremoved, dried, diluted in the mobile phase, and thensubjected to chiral high-performance liquid chromatography(HPLC) to determine the conversion and enantiomericexcess (e.e.). Chiral HPLC analysis wasperformed on an Agilent 1100 series HPLC system witha UV detector.23) Chiral CHBE was analyzed on a chiracelOB-H column (Daicel, Japan) at λ210 nm using hexane/2-propanol (90/10, v/v) as eluent at a flow rate of0.8 mL min−1 and a temperature of 25 °C. Chiral HPBEand 4-phenyl-2-butanol were analyzed on a chiracelOD-H column at λ210 nm and λ254 nm using hexane/2-propanol (98/2, v/v) as eluent at a flow rate of 1.0 mLmin−1 and a temperature of 30 °C. Authentic (relevant)standards were used for peak identification, and quantificationwas based on the peak area that was suitablycalibrated with standards of known concentration.
99 % ee With D-glucose; sodium carbonate In aq. phosphate buffer; acetic acid butyl ester at 30℃; for 5 h; Microbiological reaction; Enzymatic reaction Cells were harvested by centrifugation (10,000 g, 20 min, 4 °C) and washed with 100 mM potassium phosphate buffer (pH 6.2). The bioconversion of COBE to (S)-CHBE was performed in an aqueous–butyl acetate (4:1, v/v) biphase system containing 100 mM potassium phosphate buffer (pH 6.2), 1500 mM COBE, 1550 mM glucose, 1550 mM Na2CO3, Triton X-100 (1‰, v/v), 0.1 g of dry cell weight (DCW) of E. coli Rosseta (pET-22b-SOU1), and E. coli Rosseta (pET-22b-GDH), respectively, at a total volume of 25 mL. The reaction was performed at 30 °C and 220 rpm for 5 h. The organic layer was isolated to determine the product concentration and optical purity.
> 99 % ee With aldo-keto reductase CaAKR; NADH In aq. phosphate buffer at 30℃; for 10 h; Enzymatic reaction General procedure: The bioreduction was performed in 1.5 mL Eppendorf tubes containing 200 mmol/L potassium phosphate buffer (pH 7.0), 1 mmol/L of each substrate, 0.5 mmol/L NADH, and 1 mg/mL purified CaAKR in a total volume of 1.0 mL, shaking for 10 h at 30 °C. The reaction mixture was extracted twice with an equivalent volume of 300 μL ethyl acetate. The extracts were combined, dried with anhydrous sodium sulfate. The concentrations for each product were determined by GC or HPLC analysis.
0.91% With alpha-D-glucopyranose In toluene at 50℃; for 11 h; Enzymatic reaction Then 1.0 g COBE (6.1 mmol) and 1.2 g glucose was added. The second batch was completed within 70 min. After-wards, another 1.0 g COBE (6.1 mmol) and 1.2 g glucose was fedwhen COBE was converted thoroughly. At the 7th batch, at least3 h was required for a complete reaction. All 7.0 g COBE was fullyreduced into (S)-CHBE by merely 0.1 g RpCR-GDH dry cells within11 h without addition of external NADP+. After extraction, about6.29 g (S)-CHBE (>99percent ee) was recovered, with a molar yield ofabout 91percent. The substrate to catalyst ratio and the space-time yieldwere about 70 and 1480 g L−1d−1, which was attributed to the highcatalytic efciency of RpCR in the biphasic system with substrate-feeding.
99.95 % ee With hydrogenchloride; dichloro-R-2,2′-bis(diphenylphosphino)-1,1′-binaphthyl ruthenium; hydrogen In methanol; water at 95 - 98℃; In a 500 ml autoclave in a hydrogen atmosphere, was added Compound A: ethyl chloroacetoacetate (50 g, 0.30 mol), hydrochloric acid 5ml (AR, content of 36percent to 38percent), methanol 300 ml, the reaction solution was stirred, a chiral ruthenium metal catalyst (R) -RuCl2 (BINAP) (50 mg) was added, the reaction solution was replaced with hydrogen twice, the pressure rose to 8 to 10 atmospheres, insulation 95 ~ 98 deg. C reaction 1 ~ 2h. The reaction solution was cooled to room temperature, and the reaction solution was directly analyzed by GC to measure the conversion (column: HP-10125 m / 0.2 mm) and the enantiomeric excess (column: Lipodex-E25 m / 0.25 mm). The enantiomeric excess was 99.95percent and the conversion was 100percent. The filtrate was dried and filtered, and the solvent was distilled off under reduced pressure to obtain the compound B, which was directly used in a one-step reaction.
458 g With sodium formate In ethanol at 50℃; 5 liters of reaction flask 500g (3.03 mol) of ethyl chloroacetoacetate and 2 liters of ethanol, after stirring, add a chiral catalyst (0.15 mol, 0.05 eq.) and 250g (3.68 mol, 1.2 eq.) of sodium formate, heating to 50 °C to react; the gas phase detection reaction was complete, the solvent was removed by distillation,And distilled under reduced pressure to give 485g of chiral ethyl 4-chloro-3-hydroxybutyrate;

Reference: [1] Bioscience, Biotechnology and Biochemistry, 2005, vol. 69, # 3, p. 544 - 552
[2] Tetrahedron Asymmetry, 2007, vol. 18, # 16, p. 1883 - 1887
[3] Patent: WO2003/97569, 2003, A1, . Location in patent: Page/Page column 10-13
[4] Organic Process Research and Development, 2002, vol. 6, # 4, p. 558 - 561
[5] Tetrahedron Asymmetry, 2005, vol. 16, # 4, p. 899 - 901
[6] Tetrahedron Letters, 1994, vol. 35, # 44, p. 8119 - 8122
[7] Green Chemistry, 2010, vol. 12, # 1, p. 81 - 86
[8] Tetrahedron Asymmetry, 2011, vol. 22, # 14-15, p. 1548 - 1552
[9] Catalysis Science and Technology, 2015, vol. 5, # 1, p. 105 - 108
[10] Tetrahedron Asymmetry, 2001, vol. 12, # 12, p. 1713 - 1718
[11] Journal of Organic Chemistry, 2003, vol. 68, # 6, p. 2376 - 2384
[12] Organic letters, 2001, vol. 3, # 25, p. 4011 - 4013
[13] Tetrahedron Asymmetry, 2001, vol. 12, # 20, p. 2835 - 2843
[14] Chemistry - A European Journal, 2013, vol. 19, # 14, p. 4532 - 4537
[15] Bioscience, Biotechnology and Biochemistry, 2012, vol. 76, # 6, p. 1210 - 1212
[16] Chinese Journal of Chemistry, 2013, vol. 31, # 3, p. 349 - 354
[17] RSC Advances, 2015, vol. 5, # 29, p. 22703 - 22711
[18] Tetrahedron Asymmetry, 2008, vol. 19, # 11, p. 1347 - 1351
[19] Asian Journal of Chemistry, 2012, vol. 24, # 11, p. 5151 - 5154
[20] Patent: EP1176135, 2002, A1, . Location in patent: Example 6
[21] Process Biochemistry, 2010, vol. 45, # 9, p. 1445 - 1449
[22] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 52-53
[23] Asian Journal of Chemistry, 2014, vol. 26, # 9, p. 2695 - 2698
[24] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 52-53
[25] Organic Process Research and Development, 2014, vol. 18, # 6, p. 739 - 743
[26] Journal of Organic Chemistry, 1991, vol. 56, # 15, p. 4778 - 4783
[27] Journal of Organic Chemistry, 1991, vol. 56, # 15, p. 4778 - 4783
[28] Bioscience, Biotechnology and Biochemistry, 2001, vol. 65, # 7, p. 1676 - 1679
[29] Tetrahedron Letters, 1988, vol. 29, # 20, p. 2453 - 2454
[30] Tetrahedron Asymmetry, 2005, vol. 16, # 15, p. 2539 - 2549
[31] Patent: US7056540, 2006, B2, . Location in patent: Page/Page column 6; 9
[32] Bioscience, Biotechnology and Biochemistry, 1998, vol. 62, # 2, p. 280 - 285
[33] Bulletin of the Chemical Society of Japan, 1994, vol. 67, # 12, p. 3314 - 3319
[34] Bulletin of the Chemical Society of Japan, 1994, vol. 67, # 12, p. 3314 - 3319
[35] Chemosphere, 1999, vol. 38, # 10, p. 2243 - 2246
[36] Chemosphere, 1999, vol. 38, # 10, p. 2243 - 2246
[37] Tetrahedron Asymmetry, 1998, vol. 9, # 24, p. 4395 - 4417
[38] Russian Chemical Bulletin, 2000, vol. 49, # 4, p. 728 - 731[39] Izvestiya Akademi Nauk, Seriya Khimicheskaya, 2000, vol. 49, # 4, p. 725 - 728
[40] Bioscience, Biotechnology and Biochemistry, 2000, vol. 64, # 7, p. 1430 - 1436
[41] Synlett, 2001, # SPEC. ISS, p. 1050 - 1054
[42] Bioscience, Biotechnology and Biochemistry, 2003, vol. 67, # 6, p. 1417 - 1420
[43] Journal of the American Chemical Society, 2004, vol. 126, # 40, p. 12827 - 12832
[44] Tetrahedron Letters, 2006, vol. 47, # 12, p. 1901 - 1903
[45] Journal of Organic Chemistry, 2006, vol. 71, # 11, p. 4202 - 4205
[46] Synthesis, 2005, # 20, p. 3666 - 3671
[47] Organic Letters, 2002, vol. 4, # 26, p. 4599 - 4602
[48] Bioscience, Biotechnology and Biochemistry, 2005, vol. 69, # 3, p. 544 - 552
[49] Bioscience, Biotechnology and Biochemistry, 2006, vol. 70, # 2, p. 418 - 426
[50] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 4
[51] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 6
[52] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 5
[53] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 5
[54] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 5
[55] Patent: EP1528053, 2005, A1, . Location in patent: Page/Page column 4
[56] Patent: WO2006/2731, 2006, A1, . Location in patent: Page/Page column 19
[57] Patent: US6075154, 2000, A,
[58] Patent: WO2006/131933, 2006, A1, . Location in patent: Page/Page column 7-8
[59] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 49-50
[60] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 59-60
[61] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 53
[62] Advanced Synthesis and Catalysis, 2008, vol. 350, # 14-15, p. 2322 - 2328
[63] Patent: EP1609795, 2005, A1, . Location in patent: Page/Page column 15
[64] Patent: WO2006/2730, 2006, A1, . Location in patent: Page/Page column 21-22
[65] Australian Journal of Chemistry, 2009, vol. 62, # 9, p. 1034 - 1039
[66] Tetrahedron Asymmetry, 2009, vol. 20, # 19, p. 2263 - 2266
[67] Organic and Biomolecular Chemistry, 2011, vol. 9, # 11, p. 4070 - 4078
[68] Advanced Synthesis and Catalysis, 2011, vol. 353, # 8, p. 1213 - 1217
[69] Biochimica et Biophysica Acta - Proteins and Proteomics, 2010, vol. 1804, # 9, p. 1841 - 1849
[70] Patent: WO2011/141160, 2011, A1, . Location in patent: Page/Page column 21
[71] Bioscience, Biotechnology and Biochemistry, 2011, vol. 75, # 6, p. 1055 - 1060
[72] Organic Letters, 2012, vol. 14, # 8, p. 1982 - 1985
[73] Mendeleev Communications, 2012, vol. 22, # 4, p. 184 - 186
[74] Journal of Molecular Catalysis B: Enzymatic, 2012, vol. 84, p. 15 - 21
[75] Advanced Synthesis and Catalysis, 2013, vol. 355, # 16, p. 3179 - 3190
[76] Journal of Molecular Catalysis B: Enzymatic, 2014, vol. 105, p. 66 - 73
[77] Bioscience, Biotechnology and Biochemistry, 2014, vol. 78, # 8, p. 1350 - 1356
[78] Chemical Communications, 2015, vol. 51, # 61, p. 12328 - 12331
[79] Bioscience, Biotechnology and Biochemistry, 2015, vol. 79, # 7, p. 1090 - 1093
[80] Chemical Communications, 2015, vol. 51, # 86, p. 15728 - 15731
[81] Journal of Molecular Catalysis B: Enzymatic, 2015, vol. 122, p. 44 - 50
[82] Journal of Molecular Catalysis B: Enzymatic, 2016, vol. 123, p. 67 - 72
[83] Patent: CN105566242, 2016, A, . Location in patent: Paragraph 0046; 0047; 0048
[84] Patent: CN107098872, 2017, A, . Location in patent: Paragraph 0028; 0032; 0033; 0034; 0041; 0046
[85] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 49-50
[86] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 53
[87] Patent: WO2005/18579, 2005, A2, . Location in patent: Page/Page column 59-60
  • 21
  • [ 638-07-3 ]
  • [ 10488-69-4 ]
  • [ 10488-69-4 ]
YieldReaction ConditionsOperation in experiment
82.30 % ee at 30℃; for 8 h; Enzymatic reaction General procedure: Asymmetric reductions of various carbonyl compounds by the purified enzymes were carried out at 30°C for 8h with mild shaking in a reaction mixture containing 0.1M potassium phosphate buffer (pH 6.5), 1gL−1 substrate, 10mM NADPH, and the purified enzyme of appropriate amount in a total volume of 2mL. In order to determine the absolute configuration of chiral alcohols, the reaction products were extracted with ethyl acetate or hexane and the organic layer was used for analysis. The optical purity of the reaction products were determined by chiral HPLC (HP 1100, Agilent, USA) equipped with Chiralcel OB-H column (4.6mm×250mm; Daicel Chemical Ind. Ltd., Japan) or chiral GC (7890A, Agilent, USA) equipped with FID detector and Chrompack Chirasil-Dex CB chiral capillary column (25m×0.25mm; Varian, USA) [21].
11.84 % ee at 30℃; for 8 h; Enzymatic reaction General procedure: Asymmetric reductions of various carbonyl compounds by the purified enzymes were carried out at 30°C for 8h with mild shaking in a reaction mixture containing 0.1M potassium phosphate buffer (pH 6.5), 1gL−1 substrate, 10mM NADPH, and the purified enzyme of appropriate amount in a total volume of 2mL. In order to determine the absolute configuration of chiral alcohols, the reaction products were extracted with ethyl acetate or hexane and the organic layer was used for analysis. The optical purity of the reaction products were determined by chiral HPLC (HP 1100, Agilent, USA) equipped with Chiralcel OB-H column (4.6mm×250mm; Daicel Chemical Ind. Ltd., Japan) or chiral GC (7890A, Agilent, USA) equipped with FID detector and Chrompack Chirasil-Dex CB chiral capillary column (25m×0.25mm; Varian, USA) [21].
70 % ee With D-glucose; D-glucose dehyrodenase; Lodderomyces elongisporus aldo–keto reductase 48; NADPH In aq. phosphate buffer at 30℃; Enzymatic reaction General procedure: The enantioselectivity of LEAKRs was determined by employingNADPH regeneration system composed of D-glucose dehydrogenase(GDH) and D-glucose. The reaction mixture included 20 mMethyl 4-chloroacetoacetate, 0.2 mM NADPH, 0.03 mg LEAKR(1.2 mg for LEAKR 49), 100 mM D-glucose, 0.1 mg GDH, and100 mM sodium phosphate buffer (pH 7.0), with the final volumeof 0.5 mL. After shaking constantly at 30 C, the reaction mixturewas extracted with 1 mL ethyl acetate. The organic layer collectedby centrifugation and dried with anhydrous sodium sulfate wasthen analyzed to determine the conversion of the substrate andenantiomeric excess (e. e.) of the products by employing GC andHPLC as efficient and sensitive tools. A GC station (Agilent6890N) with a capillary column (DB-5) was used to analyze theconversion of ethyl 4-chloroacetoacetate while an HPLC station(Agilent 1100) with a Chiralcel OB-H column was employed forthe optical assay of the products.
58 % ee With D-glucose; D-glucose dehyrodenase; Lodderomyces elongisporus aldo–keto reductase 49; NADPH In aq. phosphate buffer at 30℃; Enzymatic reaction The enantioselectivity of LEAKRs was determined by employingNADPH regeneration system composed of D-glucose dehydrogenase(GDH) and D-glucose. The reaction mixture included 20 mMethyl 4-chloroacetoacetate, 0.2 mM NADPH, 0.03 mg LEAKR(1.2 mg for LEAKR 49), 100 mM D-glucose, 0.1 mg GDH, and100 mM sodium phosphate buffer (pH 7.0), with the final volumeof 0.5 mL. After shaking constantly at 30 C, the reaction mixturewas extracted with 1 mL ethyl acetate. The organic layer collectedby centrifugation and dried with anhydrous sodium sulfate wasthen analyzed to determine the conversion of the substrate andenantiomeric excess (e. e.) of the products by employing GC andHPLC as efficient and sensitive tools. A GC station (Agilent6890N) with a capillary column (DB-5) was used to analyze theconversion of ethyl 4-chloroacetoacetate while an HPLC station(Agilent 1100) with a Chiralcel OB-H column was employed forthe optical assay of the products.
21.6 % ee With D-glucose; nicotinamide adenine dinucleotide phosphate; recombinant Bacillus subtilis-derived glucose dehydrogenase; recombinant Gluconobacter oxydans carbonyl reductase 0644 In aq. phosphate buffer; ethanol at 30℃; for 12 h; Enzymatic reaction General procedure: The enzyme activity toward the reduction ofketones was determined by spectrophotometricallymeasuring the oxidation of NAD(P)H at 340 nm(ε = 6.22mM−1 cm−1) in the presence of an excess amount of ketones. The change in absorbance of NAD(P)H was monitored at 340 nm using an ultraviolet-visiblespectrophotometer with a temperature-controlledcuvette holder (Shimadzu Co., Kyoto, Japan). One unit(U) activity was defined as the amount of enzymesrequired to catalyze the oxidation of 1 μmol NAD(P)Hper minute at 30 °C. The data were expressed as U/mgof protein. The standard reaction mixture contained100mM phosphate buffer (pH 7.0), 0.1mM NAD(P)H,5 mM substrate, and enzyme solvent in a total volumeof 1 mL. The reaction was initiated by adding 20 μLsolvent containing 10 μg–40 μg of enzymes. Blankswithout the enzyme were carried out for each substrate,and data were collected in triplicate. Protein concentrationswere determined with the bicinchoninic acid assayusing bovine serum albumin as a standard.NAD(P)H or NADH was assayed under standardreaction conditions for the study of coenzyme dependence.A range of substrates from 1 to 20 mM concentrationwas assayed under the standard reactionconditions for the study of kinetics. Apparent values ofMichaelis constant (Km) and kcat were calculated by fittingthe data into Michaelis–Menten equation using theSigmaPlot (Systat Software Inc., San Jose, CA, USA).All reactions followed Michaelis–Menten-type kinetics.Enantioselective reduction of ketones. The enantioselectivityof the enzymes was determined byexamining the reduction of aryl ketones, ethyl 4-chloroacetoacetate(COBE), and ethyl 2-oxo-4-phenylbutyrate(OPBE) using an NAD(P)H regeneration system consistingof BsGDH and glucose. The general procedurewas as follows: D-glucose (0.5percent), recombinant BsGDH(10 U), NAD(P)+ (0.1 mM), the recombinant cell(30 g L−1, wet weight), and ketone solvated in ethanol[1 g L−1, 10percent(v/v)] were mixed in a potassium phosphatebuffer (10 mL, 100 mM, pH 7.0). The mixturewas shaken at 30 °C for 12 h.22) Upon termination ofthe reaction, each sample was extracted twice withequivalent ethyl acetate. The organic layer wasremoved, dried, diluted in the mobile phase, and thensubjected to chiral high-performance liquid chromatography(HPLC) to determine the conversion and enantiomericexcess (e.e.). Chiral HPLC analysis wasperformed on an Agilent 1100 series HPLC system witha UV detector.23) Chiral CHBE was analyzed on a chiracelOB-H column (Daicel, Japan) at λ210 nm using hexane/2-propanol (90/10, v/v) as eluent at a flow rate of0.8 mL min−1 and a temperature of 25 °C. Chiral HPBEand 4-phenyl-2-butanol were analyzed on a chiracelOD-H column at λ210 nm and λ254 nm using hexane/2-propanol (98/2, v/v) as eluent at a flow rate of 1.0 mLmin−1 and a temperature of 30 °C. Authentic (relevant)standards were used for peak identification, and quantificationwas based on the peak area that was suitablycalibrated with standards of known concentration.
88.8 % ee With Kluyveromyces polysporus alcohol dehydrogenase S237R mutant; isopropyl alcohol; NADPH In aq. phosphate buffer at 30℃; Enzymatic reaction General procedure: Bioconversion was conducted with 20 mM 1a–10a,20 U·mL−1KpADH variants, 40 mM isopropanol in PBS buffer (pH 7.0,100 mM) in total volume of 2 mL at 30 °C and 180 rpm overnight. Then,1 mL of the reaction mixture was withdrawn and extracted with ethylacetate. The organic phase was isolated by centrifugation and driedover anhydrous MgSO4. The conversion rate and enantioselectivity ofthe products were analyzed as described in supporting information.

Reference: [1] Asian Journal of Chemistry, 2012, vol. 24, # 11, p. 5151 - 5154
[2] Asian Journal of Chemistry, 2012, vol. 24, # 11, p. 5151 - 5154
[3] Tetrahedron Letters, 1989, vol. 30, # 17, p. 2245 - 2246
[4] Chemistry Letters, 1987, p. 679 - 682
[5] Journal of the American Chemical Society, 1983, vol. 105, # 18, p. 5925 - 5926
[6] Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1992, # 6, p. 659 - 660
[7] Bioscience, Biotechnology and Biochemistry, 1994, vol. 58, # 9, p. 1666 - 1670
[8] Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1995, # 10, p. 1295 - 1298
[9] Bioscience, Biotechnology and Biochemistry, 1998, vol. 62, # 2, p. 280 - 285
[10] Tetrahedron Letters, 1988, vol. 29, # 13, p. 1555 - 1556
[11] Chemistry Letters, 1987, p. 679 - 682
[12] Journal of the American Chemical Society, 1985, vol. 107, # 10, p. 2993 - 2994
[13] Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1992, # 6, p. 659 - 660
[14] Bulletin of the Chemical Society of Japan, 1989, vol. 62, # 3, p. 875 - 879
[15] Tetrahedron Asymmetry, 1997, vol. 8, # 17, p. 2881 - 2884
[16] Tetrahedron Letters, 1990, vol. 31, # 2, p. 267 - 270
[17] Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1990, # 6, p. 1826 - 1828
[18] Tetrahedron Letters, 1988, vol. 29, # 13, p. 1555 - 1556
[19] Tetrahedron Letters, 1989, vol. 30, # 17, p. 2245 - 2246
[20] Tetrahedron Letters, 1991, vol. 32, # 33, p. 4163 - 4166
[21] Journal of the American Chemical Society, 1985, vol. 107, # 10, p. 2993 - 2994
[22] Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1992, # 6, p. 659 - 660
[23] Tetrahedron: Asymmetry, 1994, vol. 5, # 4, p. 675 - 690
[24] Bioscience, Biotechnology and Biochemistry, 1998, vol. 62, # 2, p. 280 - 285
[25] Chemosphere, 1999, vol. 38, # 10, p. 2243 - 2246
[26] Tetrahedron Asymmetry, 1998, vol. 9, # 24, p. 4395 - 4417
[27] Tetrahedron Asymmetry, 1999, vol. 10, # 3, p. 551 - 559
[28] Tetrahedron Asymmetry, 1999, vol. 10, # 3, p. 551 - 559
[29] Tetrahedron, 2001, vol. 57, # 13, p. 2563 - 2568
[30] Tetrahedron Asymmetry, 2001, vol. 12, # 15, p. 2103 - 2107
[31] Tetrahedron Letters, 2002, vol. 43, # 8, p. 1539 - 1543
[32] Tetrahedron Letters, 2002, vol. 43, # 15, p. 2789 - 2792
[33] Tetrahedron Asymmetry, 2004, vol. 15, # 14, p. 2253 - 2261
[34] Tetrahedron Asymmetry, 2004, vol. 15, # 22, p. 3591 - 3593
[35] Tetrahedron, 2006, vol. 62, # 5, p. 901 - 905
[36] Russian Chemical Bulletin, 2004, vol. 53, # 10, p. 2172 - 2174
[37] Tetrahedron Asymmetry, 2006, vol. 17, # 12, p. 1769 - 1774
[38] Journal of the American Chemical Society, 2006, vol. 128, # 17, p. 5955 - 5965
[39] Synlett, 2006, # 5, p. 766 - 770
[40] Tetrahedron Letters, 1985, vol. 26, # 35, p. 4213 - 4216
[41] Tetrahedron Letters, 1985, vol. 26, # 35, p. 4213 - 4216
[42] Journal of Organic Chemistry, 2002, vol. 67, # 11, p. 3900 - 3903
[43] Chemistry - A European Journal, 2002, vol. 8, # 4, p. 843 - 852
[44] Advanced Synthesis and Catalysis, 2007, vol. 349, # 3, p. 323 - 336
[45] Advanced Synthesis and Catalysis, 2008, vol. 350, # 14-15, p. 2322 - 2328
[46] Tetrahedron Asymmetry, 2009, vol. 20, # 19, p. 2263 - 2266
[47] Tetrahedron Asymmetry, 2010, vol. 21, # 9-10, p. 1211 - 1215
[48] Tetrahedron Asymmetry, 2010, vol. 21, # 9-10, p. 1211 - 1215
[49] Tetrahedron Asymmetry, 2011, vol. 22, # 14-15, p. 1548 - 1552
[50] Patent: WO2011/141160, 2011, A1, . Location in patent: Page/Page column 21
[51] Advanced Synthesis and Catalysis, 2013, vol. 355, # 16, p. 3179 - 3190
[52] Journal of Molecular Catalysis B: Enzymatic, 2014, vol. 105, p. 66 - 73
[53] Journal of Molecular Catalysis B: Enzymatic, 2014, vol. 105, p. 66 - 73
[54] Tetrahedron Asymmetry, 2014, vol. 25, # 23, p. 1501 - 1504
[55] Archives of Biochemistry and Biophysics, 2014, vol. 564, p. 219 - 228
[56] Archives of Biochemistry and Biophysics, 2014, vol. 564, p. 219 - 228
[57] Bioscience, Biotechnology and Biochemistry, 2014, vol. 78, # 8, p. 1350 - 1356
[58] Journal of Molecular Catalysis B: Enzymatic, 2016, vol. 123, p. 67 - 72
[59] European Journal of Inorganic Chemistry, 2017, vol. 2017, # 20, p. 2762 - 2773
[60] Organic Process Research and Development, 2017, vol. 21, # 9, p. 1349 - 1354
[61] Catalysis Communications, 2018, vol. 108, p. 1 - 6
  • 22
  • [ 638-07-3 ]
  • [ 10488-69-4 ]
Reference: [1] Journal of Molecular Catalysis B: Enzymatic, 2011, vol. 69, # 3-4, p. 89 - 94
  • 23
  • [ 3891-07-4 ]
  • [ 638-07-3 ]
  • [ 88150-75-8 ]
YieldReaction ConditionsOperation in experiment
93% With sodium hydride In tetrahydrofuran at -10 - 32℃; a, 2400 ml of tetrahydrofuran was charged into the reaction flask A, and the mixture was cooled to -10 °C or less, 750 g of N-hydroxyethyl phthalimide and 345 g of NaH were added successively (while stirring at a low speed, N-hydroxyethyl phthalimide, NaH added to the beginning of the dropwise addition of ethyl 4-chloroacetoacetate time interval within 30min); the reaction flask temperature down to -10 ° C below, 732 g of ethyl 4-chloroacetoacetate was added slowly dropwise, and the temperature was controlled below 0 °C and the addition was completed in 1.5 to 2 hours. The temperature was raised to 28 to 32 °C. After 5 to 6 hours of reaction, 1500 ml of toluene was added, The temperature dropped below 5 °C to give a toluene dilution of the reaction solution; b, add 1700 ml of toluene, 2800 ml of water and 1178 g of acetic acid to the reaction flask B at a temperature below 5 °C; add the toluene dilution of the above reaction solution with 1 to 1.5 h at a temperature below 10 °C; After stirring for 10 min, the mixture was allowed to stand for 15 min, and the aqueous layer was removed. The toluene layer was washed 5 times with a mass fraction of 4percent brine (2500 ml each). After the washed toluene solution was added anhydrous magnesium sulfate, And the toluene solution was concentrated under reduced pressure to remove toluene (the amount of toluene removed was 2700 ml or more and the toluene content was not more than a predetermined value) to give ethyl-4- (2-phthalimidoethoxy) acetoacetate in a yield of 93percent
51%
Stage #1: With sodium hydride In tetrahydrofuran at -10℃; for 0.583333 h;
Stage #2: at -10 - 20℃; for 18 h;
Preparation of Ethyl 4-[2-(PHTHALIMIDO) ETHOXY] ACEFOACETATE (X1 A 250 mL two-necked flask, equipped with a magnetic stirring bar, thermometer and a pressure equalized addition funnel was charged with 75 mL of tetrahydrofuran under nitrogen atmosphere. 7.53 g Sodium hydride (60percent dispersed in oil) was added and the resulting suspension was cooled to-10 °C and 20 g of N- (2-HYDROXYETHYL) PHTHALIMIDE was added slowly over 5 minutes. The resulting slurry was stirred at-10 °C for 30 minutes. To this mixture a solution of 16.35 g ethyl 4-chloroacetoacetate in 30 ML of TETRAHYDRAFURAN was added at-10 °C in 40 minutes. The reaction mixture was warmed to room temperature and then stirred at room temperature for 18 hours. The reaction mixture was placed in an ice bath and quenched by dropwise addition of 10 mL ethanol. The mixture was then poured into 150 mL of IN hydrochloric acid solution in crushed ice and 200 ML of ethyl acetate was added. The resulting mixture was transferred into a separatory funnel and the aqueous phase was separated. The organic phase was first washed with 70 mL of ethyl acetate was added. The resulting mixture was transferred into a separatory funnel and the aqueous phase was separated. The organic phase was first washed with 70 mL of 5 wt. percent OF NAHC03 solution, then with 150 ML of water, dried over 5 G of MgS04. MgS04 was filtered off and the filtrate was concentrated in vacuo to give a light brown oily product. The oil was washed with 15 mL of hexane to remove the mineral oil to give 17.2 g of ethyl 4- [2- (phthalimido) ethoxy] acetoacetate light brown product. (yield 51percent, rel. compound purity > 80percent). IR (KBR, CM~L) : 2995 and 1716. 1H-NMR (CDC13) 6 7.76 (dd, 2H), 7.65 (dd, 2H), 4.11 (S, 2H), 4.02 (q, 2H), 3.80 (t, 2H), 3.70 (t, 2H), 3.37 (s, 2H), 1. 16 (t, 3H). 13C-NMR (CDC13) 8 201.7, 168.4, 167.2, 134.3, 132.2, 123.5, 75.6, 68.7, 61.6, 46.0, 37.4, 14.3.
50% With sodium hydride In DMF (N,N-dimethyl-formamide) at 0 - 20℃; Example 7 : 2-({2-[2-(4-{(2S)-2-Hydroxy-3-[(methylethyl)amino]propoxy}phenoxy) acetylamino] ETHOXY} METHYL)-4-(2-CHLOROPHENYL)-5-CYANO-6-METLLYL-1, 4- dihydropyridine-3-carboxylate is synthesized according to Scheme VI. SCHEME VI 0 Ho- p OEt HO out C1 Ni C1 Me NC Me N N CHO CHO ci NC Me H 0 1. acetone, II. -- . HO /O 2. II wEthyl 4- [2-FL, 3-DIOXOISOMDOLM-2-YDETHOXY1-3-OXOBUTANOATE. A solution of 2- (2- hydroxyethyl) isoindoline-1, 3-dione (10 g; 52.31 mmol) in DMF (150 ml) is cooled to 0°C and treated with 1. 1 equivalents of a 60percent dispersion of sodium hydride in mineral oil. After gas evolution has ceased, ethyl 4-chloro-3-oxobutanoate (7.75 g; 47.08 mmol) in 20 ml of DMF is added drop-wise, and the resulting solution is stirred overnight at room temperature. The pH of the mixture is then adjusted to 6-7 by the addition of 1 N HC1, and the product is partitioned between H2O and EtOAc. The aqueous phase is extracted again with EtOAc, and the combined organic phases are washed with water and brine, dried over magnesium sulfate, and concentrated to a crude residue, which is purified on a silica gel column, eluting with 25percent EtOAc in hexane to obtain 7.52 g (23.54 mmol ; 50percent) of the product as an oil.
40.5%
Stage #1: With sodium hydride In tetrahydrofuran; N,N-dimethyl-formamide; mineral oil at 20℃; for 4 h;
Stage #2: at 0 - 20℃;
To a mechanically stirred suspension of NaH (15g, 0. 375 mol, 60percent dispersion in mineral oil) in THF (250 mL) and DMF (25 mL) was added N- (2-hydroxyethyl)-phtalimide (43g, 0. 225 mol). The mixture was stirred at room temperature for about 4 h and then cooled to about 0°C in an ice bath. Ethyl-4- chloroacetate (21. 4 mL, 0. 15 mol) in THF (4 mL) was then added via an addition funnel over a period of about 0. 5 h and the resulting mixture allowed to stir overnight. The mixture was then poured into a 2 L sepratory funnel containing 500 mL ice water and 700 mL EtOAc and separated. The organic layer was washed 2x with 500 mL of water and dried over MGS04. The solution was then filtered through a 4-inch plug of silica and then CONCENTRATED IN VACUO. The resulting yellow oil was then dissolved in 250 mL CH3CN and washed 2x with 30 mL hexane. The CH3CN layer was the concentrated in vacuo to yield 19. 4g (40. 5percent) OF 4- [2- (1, 3-DIOXO-1, 3-DIHYDRO-ISOINDOL-2-YL)-ETHOXY]-3-OXO-BUTYRIC acid ethyl ester as a yellow oil. This crude compound was used for the next step
40.5%
Stage #1: With sodium hydride In tetrahydrofuran; N,N-dimethyl-formamide; mineral oil at 20℃; for 4 h;
Stage #2: at 0 - 20℃;
To a mechanically stirred suspension of NaH (15g, 0. 375 mol, 60percent dispersion in mineral oil) in THF (250 mL) and DMF (25 mL) was added N- (2-hydroxyethyl)-phtalimide (43g, 0. 225 mol). The mixture was stirred at room temperature for about 4 h and then cooled to about 0°C in an ice bath. Ethyl-4- chloroacetate (21. 4 mL, 0. 15 mol) in THF (4 mL) was then added via an addition funnel over a period of about 0. 5 h and the resulting mixture allowed to stir overnight. The mixture was then poured into a 2 L sepratory funnel containing 500 mL ice water and 700 mL EtOAc and separated. The organic layer was washed 2x with 500 mL of water and dried over MGS04. The solution was then filtered through a 4-inch plug of silica and then CONCENTRATED IN VACUO. The resulting yellow oil was then dissolved in 250 mL CH3CN and washed 2x with 30 mL hexane. The CH3CN layer was the concentrated in vacuo to yield 19. 4g (40. 5percent) OF 4- [2- (1, 3-DIOXO-1, 3-DIHYDRO-ISOINDOL-2-YL)-ETHOXY]-3-OXO-BUTYRIC acid ethyl ester as a yellow oil. This crude compound was used for the next step

Reference: [1] Patent: CN106749187, 2017, A, . Location in patent: Paragraph 0031; 0032; 0033; 0034
[2] Patent: WO2004/58711, 2004, A1, . Location in patent: Page 18, 19
[3] Patent: WO2005/16885, 2005, A2, . Location in patent: Page/Page column 47-49
[4] Patent: WO2005/25507, 2005, A2, . Location in patent: Page/Page column 166-167
[5] Patent: WO2005/25507, 2005, A2, . Location in patent: Page/Page column 166-167
[6] Journal of Medicinal Chemistry, 1986, vol. 29, # 9, p. 1696 - 1702
[7] Patent: US2004/44218, 2004, A1, . Location in patent: Page 3
[8] Patent: US4572908, 1986, A,
[9] Patent: US4572909, 1986, A,
[10] Patent: US2007/260065, 2007, A1, . Location in patent: Page/Page column 5-6
[11] Patent: CN108358833, 2018, A, . Location in patent: Paragraph 0013; 0015; 0016
[12] Patent: CN108456160, 2018, A, . Location in patent: Paragraph 0006; 0014; 0015; 0016; 0020
  • 24
  • [ 504-29-0 ]
  • [ 638-07-3 ]
  • [ 101820-69-3 ]
YieldReaction ConditionsOperation in experiment
92.8% With copper(I) triflate; triethylamine In ethanol at 55 - 60℃; Dissolve 23.5 g (250 mmol) of 2-aminopyridine with 100 mL of absolute ethanol, add 35.4 g of triethylamine, 2.4 g of copper trifluoromethanesulfonate to the solution, and add dropwise 100 mL of absolute ethanol to the solution 47.3 while stirring. g ethyl 4-chloroacetoacetate, heated to 55-60° C., reacted for 4-5 hours, TLC (developer: chloroform:methanol=2:1) was used to monitor the reaction solution to 2-aminopyridine.The spots disappeared, and 1percent citric acid solution was added to the solution until the pH of the solution was 7.6. The insolubles were removed by filtration and the filtrate was extracted with ethyl acetate. The organic layer was collected, dried, filtered and concentrated to give a pale yellow viscous liquid. 47.4 g, yield 92.8percent (calculated as 2-aminopyridine), purity 97.57percent, impurity 2-(imidazo[1,2-a]pyridin-2-yl)acetic acid ethyl ester 0.43percent (HPLC normalization method) .
Reference: [1] Patent: CN104945436, 2017, B, . Location in patent: Paragraph 0035; 0036; 0037; 0038; 0039; 0040; 0041; 0042
  • 25
  • [ 25984-63-8 ]
  • [ 638-07-3 ]
  • [ 161797-99-5 ]
Reference: [1] New Journal of Chemistry, 2018, vol. 42, # 1, p. 76 - 84
Same Skeleton Products
Historical Records

Related Functional Groups of
[ 638-07-3 ]

Aliphatic Chain Hydrocarbons

Chemical Structure| 32807-28-6

[ 32807-28-6 ]

Methyl 4-chloro-3-oxobutanoate

Similarity: 0.94

Chemical Structure| 276249-18-4

[ 276249-18-4 ]

tert-Butyl 6-chloro-3,5-dioxohexanoate

Similarity: 0.87

Chemical Structure| 54527-68-3

[ 54527-68-3 ]

2-Chloroethyl 3-oxobutanoate

Similarity: 0.83

Chemical Structure| 609-15-4

[ 609-15-4 ]

Ethyl 2-chloroacetoacetate

Similarity: 0.80

Chemical Structure| 105484-55-7

[ 105484-55-7 ]

Ethyl 8-chlorooctanoate

Similarity: 0.79

Chlorides

Chemical Structure| 32807-28-6

[ 32807-28-6 ]

Methyl 4-chloro-3-oxobutanoate

Similarity: 0.94

Chemical Structure| 276249-18-4

[ 276249-18-4 ]

tert-Butyl 6-chloro-3,5-dioxohexanoate

Similarity: 0.87

Chemical Structure| 54527-68-3

[ 54527-68-3 ]

2-Chloroethyl 3-oxobutanoate

Similarity: 0.83

Chemical Structure| 609-15-4

[ 609-15-4 ]

Ethyl 2-chloroacetoacetate

Similarity: 0.80

Chemical Structure| 105484-55-7

[ 105484-55-7 ]

Ethyl 8-chlorooctanoate

Similarity: 0.79

Esters

Chemical Structure| 32807-28-6

[ 32807-28-6 ]

Methyl 4-chloro-3-oxobutanoate

Similarity: 0.94

Chemical Structure| 276249-18-4

[ 276249-18-4 ]

tert-Butyl 6-chloro-3,5-dioxohexanoate

Similarity: 0.87

Chemical Structure| 54527-68-3

[ 54527-68-3 ]

2-Chloroethyl 3-oxobutanoate

Similarity: 0.83

Chemical Structure| 609-15-4

[ 609-15-4 ]

Ethyl 2-chloroacetoacetate

Similarity: 0.80

Chemical Structure| 105484-55-7

[ 105484-55-7 ]

Ethyl 8-chlorooctanoate

Similarity: 0.79

Ketones

Chemical Structure| 32807-28-6

[ 32807-28-6 ]

Methyl 4-chloro-3-oxobutanoate

Similarity: 0.94

Chemical Structure| 276249-18-4

[ 276249-18-4 ]

tert-Butyl 6-chloro-3,5-dioxohexanoate

Similarity: 0.87

Chemical Structure| 54527-68-3

[ 54527-68-3 ]

2-Chloroethyl 3-oxobutanoate

Similarity: 0.83

Chemical Structure| 609-15-4

[ 609-15-4 ]

Ethyl 2-chloroacetoacetate

Similarity: 0.80

Chemical Structure| 141-97-9

[ 141-97-9 ]

Ethyl acetoacetate

Similarity: 0.79