DR ANTHONY MELVIN CRASTO,WorldDrugTracker, helping millions, A 90 % paralysed man in action for you, I am suffering from transverse mylitis and bound to a wheel chair, With death on the horizon, nothing will not stop me except God................DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai) , INDIA 25Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK GENERICS at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution

Monday 26 December 2016

One-step asymmetric synthesis of (R)- and (S)-rasagiline by reductive amination applying imine reductases

Graphical abstract: One-step asymmetric synthesis of (R)- and (S)-rasagiline by reductive amination applying imine reductases
One-step asymmetric synthesis of (R)- and (S)-rasagiline by reductive amination applying imine reductases
Green Chem., 2017, Advance Article
DOI: 10.1039/C6GC03023H, Communication
P. Matzel, M. Gand, M. Hohne
Imine reductases (IREDs) show great potential as catalysts for reductive amination of ketones to produce chiral secondary amines.
One-step asymmetric synthesis of (R)- and (S)-rasagiline by reductive amination applying imine reductases
Imine reductases (IREDs) show great potential as catalysts for reductive amination of ketones to produce chiral secondary amines. In this work, we explored this potential and synthesized the pharmaceutically relevant (R)-rasagiline in high yields (up to 81%) and good enantiomeric excess (up to 90% ee) from the ketone precursor. This one-step approach in aqueous medium represents the shortest synthesis route from achiral starting materials. Furthermore, we demonstrate for the first time that tertiary amines also can be accessed by this route, which provides new opportunities for eco-friendly enzymatic asymmetric syntheses of these important molecules.

One-step asymmetric synthesis of (R)- and (S)-rasagiline by reductive amination applying imine reductases

P. Matzel,a   M. Gandb and   M. Höhne*a  
*Corresponding authors
aInstitute of Biochemistry, Greifswald University, Felix-Hausdorff-Str. 4, 17487 Greifswald, Germany
E-mail: Matthias.Hoehne@uni-greifswald.de
bBiocenter Klein Flottbek, University of Hamburg, Ohnhorststr. 18, 22609 Hamburg, Germany
Green Chem., 2017, Advance Article
DOI: 10.1039/C6GC03023H
str0 str1 str2 str3 str4
 
////////////One-step, asymmetric synthesis,  (R)- ,  (S)-rasagiline,  reductive amination,  imine reductases

Saturday 24 December 2016

(3aS, 6aR)-3-Oxyhexahydrofuro [2, 3-b] furan


(3aS, 6aR)-3-Oxyhexahydrofuro [2, 3-b] furan

809286-93-9 cas
[ Molecular Formula ]:C6H8O3
[ Molecular Weight ]:128.12600

(6): Under a nitrogen atmosphere, a, solution containing 440 mg (3.38 mmol) (3S, 3aS, 6aR)-3-hydroxy-hexahydrofuro [2, 3-b] furan (5), 599 mg (5.11 mmol) 4-methylmorpholine-N-oxide, and 2 g 4 A molecular sieves in CH2Cl2 (30 mL) was stirred for 20 minutes. Then, tetrapropylammonium perruthenate (36 mg (0.10 mmol)) was added at room temperature. After 5-10 minutes, the reaction mixture was filtered through a pad of silica gel (5 g) with CH2Cl2 (100 mL). After evaporation, column chromatography (silica gel 70 g, ethyl acetate in hexanes 40%) gave compound (6) (409 mg. 94%) as a colorless solid, Rf=0.3, [α]25D -126.6°, c 0.8, CHCl3

IR (Neat) 1758, 1658, 1023 cm-1;

 1H-NMR (400 MHz, CDCl3) δ: 2.23 (mc, 1H), 2.96 (dd, 1H, J=6.8 Hz, J=6.8 Hz), 3.79 (mc, 1H), 3.99 (m, 1H), 4.11 (s, 2H), 6.02 (d, 1H, J=5.0 Hz).

 13C-NMR (100.6 MHz, CDCl3, Dept) δ: 30.38 (-), 49.58 (+), 67.65 (-), 71.69 (-), 107.89 (+), 215.52 (quat). 

C6H8O3;
Exact Mass: 128.05; Mol. Wt.: 128.13; C, 56.24, H, 6:29, 0, 37.46.



https://www.google.com/patents/US20040127727



/////////////

Friday 23 December 2016

(3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-ol , Furofuranol

str1
CAS :156928-09-5  
Molecular Formula:C6H10O3  
Molecular Weight:130.144
  • Furo[2,3-b]furan-3-ol, hexahydro-, [3R-(3α,3aβ,6aβ)]-
  • (3R,3aS,6aR)-Hexahydrofuro[2,3-b]furan-3-ol
  • 3R,3AS,6aR-hexahydrofuro[2,3-b]furan-3-ol
  • R,S,R-Bisfuran alcohol
WO2012075122  SP ROT= -13.2/1G/100ML, METHANOL
PATENT
The overall synthesis of the present invention is shown in the scheme 1:
Figure imgf000005_0003
Yet another aspect of present invention is to provide a process for the preparation of compound formula I as per below scheme 2.
Figure imgf000006_0001
str1

str2
(3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-ol
(3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-ol (7) as clear oil (7.8 g, 96.8 A% purity by GC-MS, 55.7 mmol, 74% yield). C6H10O3, GC-MS (EI): m/z 100 (M- H2CO).
1H NMR (CDCl3): 1.88 (m, 1H), 2.08 (bd, 1H, −OH), 2.31 (m, 1H), 2.87 (m, 1H), 3.64 (dd, J = 9.2, 7.0 Hz, 1H), 3.87–4.02 (abx system, 3H), 4.45 (m, 1H), 5.70 (d, J = 5.2 Hz, 1H).
13C NMR (CDCl3): 109.54, 73.15, 71.00, 69.90, 46.58, 24.86.
Diastereomeric ratio of 7 to 12 = 98.2:1.8.
GC retention time of 7= 3.20 min; 12 = 3.09 min.
Abstract Image
A practical synthesis of (3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-ol—a key intermediate in the synthesis of darunavir—from monopotassium isocitrate is described. The isocitric acid salt, obtained from a high-yielding fermentation fed by sunflower oil, was converted in several steps to a tertiary amide. This amide, along with the compound’s ester functionalities, was reduced with lithium aluminum hydride to give, on acidic workup, a transient aminal-triol. This was converted in situ to the title compound, the bicyclic acetal furofuranol side chain of darunavir, a protease inhibitor used in treatment of HIV/AIDS. Key to the success of this process was identifying an optimal amide that allowed for complete reaction and successful product isolation. N-Methyl aniline amide was identified as the most suitable substrate for the reduction and the subsequent cyclization to the desired product. Thus, the side chain is produced in 55% overall yield from monopotassium isocitrate.
Practical Synthesis of the Bicyclic Darunavir Side Chain: (3R,3aS,6aR)-Hexahydrofuro[2,3-b]furan-3-ol from Monopotassium Isocitrate
Clinton Health Access Initiative, 800 North Five Points Road, West Chester, Pennsylvania 19380, United States
Org. Process Res. Dev., Article ASAP
 
*E-mail address: tyue@clintonhealthaccess.org.
1H NMR PREDICT
str1 str2
 
13C NMR PREDICT
str1 str2
 PATENT
In particular, the following synthetic scheme (1) illustrates the present commercial method of synthesizing compound (I) . This synthesis is disclosed in detail in A.K. Ghosh et al . , Tetra- hedron Letters, 36 (4) , pp. 505-508 (1995), incorporated herein by reference. Also see, A.K. Ghosh et al., J. Med. Chem . , 39, pp. 3278-3290 (1996) for the synthesis of compound (I) and a related compound of structural formula (II) (i.e., (3S, 3aR, 7aS) -3- hydroxyhexahydrofuro [2, 3-b] pyran) .
Figure imgf000004_0001
Scheme 1 (prior art)
Figure imgf000004_0002
(91%)
Cobaloxime (catalytic) , NaBH4, EtOH
Figure imgf000004_0003
Figure imgf000005_0001
Alternatively,
-OAc
0 Immobilized Lipase 30
0- pH 7 buffer 23°C, 24 h
(+)
Figure imgf000005_0002
R=Ac
MeLi, THF
^ R=H (Compound (I))
The present method of synthesizing bis-THF is summarized as follows:
Figure imgf000007_0002
Figure imgf000008_0001
<
(78-100%;
Figure imgf000008_0002
(70-90%)
Figure imgf000008_0003
(65-80%;
Figure imgf000008_0004
2. NaBH4, EtOH (65-75%) -15°C, 1-3 h Compound (I) (bis-THF) Another aspect of the present invention is to provide a method of preparing a compound having a structure
Figure imgf000009_0001
then utilizing the benzyl-protected 5-hydroxymethyl- 5H-furan-2-one in the synthesis of compound (I) .
Another aspect of the present invention is to provide a method of preparing compounds related to bis-THF by using a starting material having a following structure:
Figure imgf000009_0002
Figure imgf000009_0003
X
I
R R
The synthesis of bis-THF (compound (I) ) is summarized below:
Figure imgf000012_0001
(1) (2)
Figure imgf000012_0002
(3)
Figure imgf000012_0003
15) (6)
Figure imgf000013_0001
(I)
(3R, 3aS , 6aR) -3-Hydroxyhexahydrofuro [2 , 3-b] uran (I)
Figure imgf000030_0001
(3R, 3aS, 6aR) -3-Hydroxyhexahydrofuxo [2, 3- b] furan (I) : To a solution containing 250 mg (1.95 mmol) (3aS, 6aR) -3-oxyhexahydrofuro [2, 3-b] furan (6) in EtOH (25 mL) was added '89 mg (2.35 mmol) NaBH4 at -18 °C. The reaction mixture was stirred at -18 °C for 2.5 hours, then the reaction was quenched with saturated NH4C1 solution (5 mL) and warmed to room temperature. The resulting mixture was concentrated under reduced pressure, and then 10 mL water was added. The aqueous layer was extracted with ethyl acetate (3 x 50 mL) and a solution of 70% CHC13, 20% MeOH, and 10% water (3 x 50 mL) . The combined organic extracts were dried over Na2S04. Column chromatography (silica gel 80 g, MeOH in CHC13 7%) gave compound (I) (178 mg. 70%) as a colorless solid, Rf=0.3, [α]25 D -12.4°, c 1.3, MeOH. IR (neat) 2951, 1641, 1211 cm"1XH-NMR (400 MHz CDC13) δ: 1.85 (mc, IH) , 1.94 (bs, IH) , 2.27 (mc, IH) , 2.84 (mc, IH) , 3.63 (dd, IH, J=7.1 Hz, J=9.2 Hz), 3.89 (mc, IH) , 3.97 (mc, IH) , 4.43 (dd, IH, J=6.8 Hz, J=14.5 " Hz), 5.68 (d, IH, J=5.2 Hz). 13C-NMR (125.8 MHz, CDC13, Dept) δ': 25.27 (-) , 46.97 (+) , 70.31 (-) , . 71.26 (-), 73.50 (+) , 109.93. (+) . C6H10O3; Exact Mass: 130.06; Mol. Wt . : 130.14; C, 55.37, H, 7.74, 0, 36.88.
Experimentals :
l-(Benzyloxy)-but-3-en-2-ol (±)-(8): To a solution of vinylmagnesium bromide (1 M in THF, 40 mL, 40 mmol) in THF (10 mL) at 0°C was added benz- yloxyacetaldehyde (7) (5 g, 33.3 mmol) dropwise. The mixture was stirred for 10 min at 0°C, and the reaction then was quenched with 20 L of saturated NaHC03 solution. The layers were separated, the aqueous layer was extracted with ethyl acetate (3 x 20 mL) , and the combined organic extracts were dried over sodium sulfate. Evaporation of solvent under reduced pressure, followed by column chromatography on silica gel (20% EtOAc in hexanes as the eluent) yielded alcohol (±)-8 (5.22 g, 88%) as a yellow oil, Rf=0.40 (30% EtOAc in hexanes); 1H-NMR (400 MHz, CDC13) δ: a 2.79 (bs, IH) , 3.39 (dd, IH, J=1.7, 7.85 Hz), 3.55 (dd, IH, J=3.35, 6.3 Hz), 4.35 (m, IH) , 4.58 (s, IH) , 5.21 (dt, IH, J=7.75, 1.4 Hz), 5.38 (dt, IH, J=14.18, 1.4 Hz), 5.84 (m, IH) , 7.30-7.38 (m, 5H) ; 13C-NMR (100.6 MHz, CDC13) δ: 71.52, 73.37, 74.02, 116.49, 127.85, 128.49, 136.58, 137.81. (S)-l-(Benzyloxy) -but-3-en-2-ol (9) and (R) -1- (benzyloxy) -but-3-en-2-oyl acetate (10):
A: To a solution of alcohol (±)-(8) (5.21 g, 29.3 mmol) in acetic anhydride (14 mL, 147 mmol) and tert-butyl methyl ether (70 mL, 586 mmol) was added immobilized lipase PS-30 (5.3 g ) on Celite 521 (Aldrich) . The mixture was stirred at room temperature for 20 h, and then filtered through Celite. Removal of solvent under reduced pressure followed, by column chromatography on silica gel (10 and 15% EtOAc in hexanes as the eluents) yielded acetate (10) (3.81 g, 54%) Rf=0.57 (30% EtOAc in hexanes) as a clear oil, [of]25 D -2° (c 1, CHC13) ; NMR (500 MHz, CDC13) δ: 2.10 (s, 3H) , 3.55-3.59 (m, 2H) , 4.56 (q, 2H, J=12.2, 14.0 Hz), 5.24 (d, IH, J-10.6 Hz), 5.32 (d, IH, J=17.3 Hz), 5.50 (m, IH) , 5.84 (m, IH) , 7.25-7.36 (m, 5H) ; 13C-NMR (125.8 MHz, CDC13) δ: 21.62, 71.67, 73.57, 73.59, 118.39, 128.14, 128.84, 133.77, 138.32, 170.63; alcohol 9 (2.34 g, 45%) as a yellow oil, Rf=0.40 (30% EtOAc in hexanes), [α]25 D- 8.3° (c 1.06, MeOH) .
B: To a solution of alcohol (±)-(8) (3.92 g, 22.0 mmol) in vinyl acetate (46 mL, 499 mmol) and ethylene glycol dimethyl ether (46 mL, 440 mmol) was added immobilized lipase PS-30 (4 g ) on Celite-545 (Aldrich) . The mixture was stirred at room temperature for 28 h, and then filtered through celite. Removal of solvent under reduced pressure, followed by column chromatography on silica gel (10 and 15% EtOAc in hexanes as the eluents) yielded acetate
(10) (2.20 g, 45%) Rf=0.57 (30% EtOAc in hexanes) as a clear oil, [ ]25 D -2.7° (c 1.35, MeOH); alcohol (9) (2.00 g, 51%) as a yellow oil, Rf=0.40 (30% EtOAc in hexanes), [α]25 D -11.4° (c 1.6, MeOH).
C: To a solution of alcohol (+)-(8) (30 mg, 0.168 mmol) in isopropenyl acetate (375 μL, 3.36 mmol) and ethylene glycol dimethyl ether (375 μL, 3.61mmol) was added immobilized lipase PS-30 (35 mg) on Celite-545 (Aldrich) . The mixture was stirred at room temperature for 23 h, and then filtered through celite. Removal of solvent under reduced pressure, followed by column chromatography on silica gel (10) and 15% EtOAc in hexanes as the eluents) yielded acetate 10 (20.3 mg, 54%) as an oil, Rf=0.57 (30% EtOAc in hexanes), [α]25 D -1.4° (c 1.02, MeOH); alcohol (9) (13 mg, 43%) as a yellow oil, Rf=0.40
(30% EtOAc in hexanes), [ ]25 D -13.5° (c 1.3, MeOH). (R) -1- (Benzyloxy) -but-3-en-2-ol (11): To a solution of acetate (10) (3.7 g, 16.9 mmol) in methanol (20 mL) was added K2C03 (7 g, 50.6 mmol). The mixture was stirred at room temperature for 35 min. Methanol then was removed under reduced pressure. The resulting solid residue was dissolved in ethyl acetate, washed with saturated NH4C1 solution and brine, and dried over sodium sulfate. Removal of ethyl acetate under reduced pressure yielded the crude alcohol (11) (3 g, 100%) as a yellow oil, Rf=0.40 (30% EtOAc in hexanes), [α]25 D 8.3° (c 1.06, MeOH) .
(S)-l- (Benzyloxy) -but-3-en-2-ol (9) from (11): To a solution of crude alcohol (5) (2 g, 11.2 mmol), triphenylphosphine (5.88 g , 22.4 mmol), and 4-nitrobenzoic acid (2.81 g, 16.8 mmol) in benzene (35 mL) was added at room temperature diisopropyl azodicarboxylate (4.35 mL, 22.4 mmol) dropwise. The mixture was stirred for 40 min, followed by the re- moval of solvent under reduced pressure. All of the crude ester then was dissolved in a mixture of MeOH:Et3N:H20 (20ml) in the ratio of 4:3:1 and reacted with LiOH (1.64 g, 39.3 mmol) at room temperature. The mixture was stirred for 2 h, followed by the removal of solvent. Column chromatography on silica gel (15% EtOAc in hexanes as the eluent) yielded alcohol (3) (1.64 g, 82%) as a yellow oil, Rf=0.40 (30% EtOAc in hexanes), [o;]25 D -7.3° (c 0.82, MeOH) . (S) -1- (Benzyloxy) -but-3-en-2-yl acrylate
(12): To a solution of alcohol (3) (1 g, 5.61 mmol) in CH2C12 (20 L) was added acryloyl chloride (685 μL, 8.41 mmol) dropwise, followed by the addition of Et3N (1.56 mL, 11.2 mmol). The resulting mixture was stirred for 10 min, and the solvent then was removed under reduced pressure. Filtration of the concentrated crude acrylate through a pad of silica gel using 15% EtOAc in hexanes, followed by the removal of solvent, yielded acrylate (12) (1.19 g, 92%) as a colorless oil, Rf=0.57 (30% EtOAc in hexanes), [α]25 D -5.7° (c 1.09, CHC13) ; 1H-NMR (500 MHz, CDC13) δ: 3.59-3.65 (m, 2H) , 4.56 (q, 2H, J=12.2, 14.65 Hz), 5.25 (d, IH, J=10.6 Hz), 5.33 (d, IH, J=16.8 Hz), 5.57 (m, IH) , 5.84-5.91 (m, 2H) , 6.17 (dd, IH, J=6.9, 10.4 Hz), 6.44 (dd, IH, J=1.3, 16.2 Hz), 7.27-7.36 (m, 5H) ; 13C-NMR (125.8 MHz, CDC13) δ: 71 . 62 , 73 . 58 , 73 . 77 , 118 . 49 , 128 . 05 , 128 . 85 , 131 . 52 , 133 . 62 , 138 . 31 , 165 . 79 .
(5S) -5- (Benzyloxymethyl) -5H-furan-2-one (13): To a solution of acrylate (12) (1.87 g, 8.05 mmol) in CH2C12 (700 mL) was added second generation Grubbs' catalyst (4 mol %, 170 mg, 0.322 mmol). The reaction mixture was refluxed for 5 hours, and the solvent then was removed under reduced pressure. Column chromatography on silica gel (30% EtOAc in hexanes as the eluent) yielded the furanone (13)
(1.62 g, 98%) as a brown oil, Rf=0.15 (30% EtOAc in hexanes), [α]25 D -81.3° (c 1.09, MeOH); αH-NMR (500 MHz, CDC13) δ: 3.66 (dd, IH, J=5.0, 5.5 Hz), 3.71 (dd, IH, J=5.0, 5.2 Hz), 4.57 (s, 2H) , 5.17 (m, IH) , 6.16 (dd, IH, J=1.9, 3.8 Hz), 7.29-7.37 (m, 5H) , 7.48 (dd, IH, J=1.4, 4.3 Hz); 13C-NMR (125.8 MHz, CDC13) δ: a 69.86, 74.18, 82.61, 123.03, 128.42, 128.95, 137.69, 154.32, 173.19.
(4S ,5S) -5- (Benzyloxymethyl) -4- [1 , 3] di- oxolan-2-yldihydrofuran-2-one (14) : A solution of furanone (13) (1.2 g, 5.88 mmols) and benzophenone
(108 mg, 0.588 mmols) in [1, 3] -dioxolane (108 mg) was degassed for 40 min in a stream of argon. The mixture then was irradiated using one 450 watt ACE glass medium pressure mercury lamp, from a distance of 15 cm, for 9 hours. Progress of this reaction was observed via 1H-NMR. As the reaction mixture was degassed, and throughout all of the irradiation time, the reaction flask was held in a water cooled cooling mantel. The temperature of the cooling water was constantly maintained near 0°C. Upon completion of the reaction, solvent was removed under reduced pressure, followed by column chromatography on silica gel (35% EtOAc in hexanes as the eluent), yielding the title compound (1.34 g, 82%) as a clear oil, Rf=0.14 (30% EtOAc in hexanes), [α]25 D 16.5° (c 1.2, CHC13) ; 1H-NMR (500 MHz, CDC13) δ: 2.50 (dd, IH, J=3.9, 12.9 Hz), 2.70-2.79 (m, 2H) , 3.58 (dd, IH, J=3.5, 7.2 Hz), 3.75 (dd, IH, J=2.8, 7.9 Hz), 3.87-3.92 (m, 2H) , 3.97-4.00 ( , 2H) , 4.51 (d, IH, J=11.9 Hz), 4.57-4.61 (m, 2H) , 4.88 (d, IH, J=3.6 Hz), 7.26-7.36 (m, 5H) ; 13C-NMR (125.8 MHz, CDC13) δ: 30.39, 40.53, 65.77, 71.74, 73.99, 79.52, 104.14, 128.00, 128.89, 138.07, 176.79.
(4S,5S) -4-[l,3]Dioxolan-2-yl-5-hydroxy- methyldihydrofuran-2-one (15) : To a solution of dihydrofuranone (14) (0.5 g, 1.79 mmol) in MeOH (30 mL) was added Pd/C (25 mg) . The mixture was stirred at room temperature under an H2 balloon for 24 hours, and then filtered over Celite. Removal of solvent under reduced pressure, followed by column chromatography on silica gel (35% EtOAc in hexanes as the eluent) yielded the compound (15) (301 mg, 89%) as a white solid, Rf=0.28 (50% EtOAc in hexanes), [ ]25 D 22° (c 1.32, CHC13) ; XH-NMR (500 MHz, CDC13) δ: 2.54 (dd, IH, J=6.0, 11.4 Hz), 2.68-2.81 (m, 2H) , 3.66
(dd, IH, J=3.9-8.5 Hz), 3.88-3.95 (m, 3H) , 3.97-4.02 (m, 2H) , 4.53 (m, IH) , 4.91 (d, IH, J=3.9 Hz); 13C- NMR (125.8 MHz, CDC13) δ: 30.68, 40.12, 64.36, 65.77, 81.07, 103.94, 176.83. (3S , 3aS , 6aR) -3-Hydroxyhexahydrofuro [2 , 3- b] furan (5) : To a solution of lithium aluminum hydride (76 mg, 1.98 mmols) in THF (10 ml) at 0°C was added dihydrofuranone 15 (275 mg , 1.46 mmol) in THF (30 mL ) dropwise. Upon completion of the reduction after 4 hours, the reaction was quenched with a saturated aqueous sodium sulfate solution at 0°C. The solvent then was decanted and the remaining residue was washed with THF (3x) , EtOAc (3x) , and CHC13 (3x) . The organic extracts were combined and the solvent was removed under reduced pressure, yielding a crude (2S, 3S) -3- [1, 3] dioxolan-2- ylpentane-1, 2, 5-triol, which was immediately used in the next reaction.
The crude triol was dissolved in a mixture of THF:H20 (8ml) in the ratio of a 5:1. This solu- tion then was acidified at room temperature to pH 2- 3 with 1 N hydrochloric acid, and was stirred for 40 hours. Removal of solvent with the aid of benzene under reduced pressure, followed by column chromatography purification on silica gel (5% MeOH in CHCI3 as the eluent) yielded the compound (5) (145 mg,
77%) as a white solid, Rf=0.40 (15% MeOH in CHC13) , [α]25 D -25.1° (c 1.05, CHC13) ; XH-NMR (500 MHz, CDCI3) δ: 1.67 ( , IH) , 2.13 (m, IH) , 2.31 (bs, IH) , 2.79 (m, IH) , 3.80-3.88 (m, 3H) , 3.95 (dd, IH, J=3.2, 7.1 Hz), 4.20 (d, IH, J=3.1 Hz), 5.86 (d, IH, J=4.9 Hz). Preparation of bis-THF derivative (I) (by Mitsunobu inversion of compound (5) ) : To a stirred solution of alcohol (5) (400 mg, 3.07 mmol), tri- phenylphosphine (1.6 g, 61.4 mmol), and p-nitroben- zoic acid (770 mg, 4.61 mmol) in dry benzene (30 mL) at 23 °C was added diisoproylazodicarboxylate (DIAD, 1.2 L, 6.14 mmol) dropwise. After 1.5 hours, the mixture was concentrated in vacuo, and the crude ester was dissolved in a (4:3:1) mixture of MeOH:Et3N:H20 (24 mL) , then treated with LiOH (450 mg, 10.7 mmol) . The solution was stirred at room temperature for 2 h. The mixture then was concentrated under reduced pressure and the residue was chromatographed over silica gel to provide the bis-
THF (I) (326 mg, 82%); [ ]25 D -12.4 (c 1.16 , MeOH)
 
In particular, the following synthetic scheme (1) illustrates the present commercial method of synthesizing compound (I). This synthesis is disclosed in detail in A.K. Ghosh et al., Tetra. hedron Letters, 36 (4) , pp. 505-508 (1995), incorporated herein by reference. Also see, A.K. Ghosh et al., J. Med. Chem . , 39, pp. 3278-3290 (1996) for the synthesis of compound (I) and a related compound of structural formula (II) (i.e., (3S, 3aR, 7aS) -3-hydroxyhexahydrofuro [2, 3-b] pyran).

The present method of synthesizing bis-THF is summarized as follows:

The synthesis of bis-THF (compound (I) ) is summarized below:
 
previously.

REF
//////////(3R,3aS,6aR)-hexahydrofuro[2,3-b]furan-3-ol, furofuranol, DARUNAVIR
O[C@H]1CO[C@H]2OCC[C@@H]12

Thursday 22 December 2016

(S)-2-(4-(Piperidin-3-yl)phenyl)-2H-indazole-7-carboxamide Tosylate Monohydrate

str1 str2
str1 as tosylate H2o
1613220-15-7 cas
free 1038915-60-4

(S)-2-(4-(Piperidin-3-yl)phenyl)-2H-indazole-7-carboxamide Tosylate Monohydrate 1
................... The solid was collected and dried in vacuo at 40 °C to afford 1 as the tosylate monohydrate salt (797 g, 86%, >99 wt %, >99%ee) as a tan-coloured solid.
Mp = 144 °C. 1H NMR (600 MHz, CD3OD) δ 8.95 (1H, s), 8.15 (1H, dd, J = 7.1, 1.2 Hz), 8.02 (2H, m), 8.00 (1H, dd, J = 8.3, 1.2 Hz), 7.72 (2H, m), 7.49 (2H, m), 7.25 (1H, dd, J = 8.3, 7.1 Hz), 7.22 (2H, d, J = 8.0 Hz), 3.49–3.43 (2H, m), 3.16–3.04 (3H, m), 2.34 (3H, s), 2.09–2.05 (2H, m), 1.96–1.82 (2H, m).
13C NMR (150.9 MHz, CD3OD) δ 169.7, 148.1, 143.7, 143.0, 141.9, 140.5, 131.8, 130.0, 129.8, 127.3, 127.1, 125.4, 124.2, 123.3, 122.4, 50.2, 45.2, 41.1, 30.9, 24.0, 21.4.
 
 
 

Discovery of 2-{4-[(3S)-Piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): A Novel Oral Poly(ADP-ribose)polymerase (PARP) Inhibitor Efficacious in BRCA-1 and -2 Mutant Tumors

IRBM/Merck Research Labs Rome, Via Pontina km 30,600, 00040 Pomezia, Italy
J. Med. Chem.200952 (22), pp 7170–7185
DOI: 10.1021/jm901188v, http://pubs.acs.org/doi/abs/10.1021/jm901188v?source=chemport
 
*To whom correspondence should be addressed. Current address: Department of Medicinal Chemistry, Merck Research Labs Boston, Avenue Louis Pasteur 33, Boston, MA 02115-5727. Phone: +1-617-992-2292. Fax: +1-617-992-2405. E-mail: philip_jones@merck.com.

Abstract

Abstract Image
We disclose the development of a novel series of 2-phenyl-2H-indazole-7-carboxamides as poly(ADP-ribose)polymerase (PARP) 1 and 2 inhibitors. This series was optimized to improve enzyme and cellular activity, and the resulting PARP inhibitors display antiproliferation activities against BRCA-1 and BRCA-2 deficient cancer cells, with high selectivity over BRCA proficient cells. Extrahepatic oxidation by CYP450 1A1 and 1A2 was identified as a metabolic concern, and strategies to improve pharmacokinetic properties are reported. These efforts culminated in the identification of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide 56 (MK-4827), which displays good pharmacokinetic properties and is currently in phase I clinical trials. This compound displays excellent PARP 1 and 2 inhibition with IC50 = 3.8 and 2.1 nM, respectively, and in a whole cell assay, it inhibited PARP activity with EC50 = 4 nM and inhibited proliferation of cancer cells with mutant BRCA-1 and BRCA-2 with CC50 in the 10−100 nM range. Compound 56 was well tolerated in vivo and demonstrated efficacy as a single agent in a xenograft model of BRCA-1 deficient cancer.


//////////1613220-15-7, 1038915-60-4, 2-[4-(3S)-3-Piperidinylphenyl]-2H-indazole-7-carboxamide, Niraparib, mk 4827

Efficient Transposition of the Sandmeyer Reaction from Batch to Continuous Process

Abstract Image
The transposition of Sandmeyer chlorination from a batch to a safe continuous-flow process was investigated. Our initial approach was to develop a cascade method using flow chemistry which involved the generation of a diazonium salt and its quenching with copper chloride. To achieve this safe continuous process diazotation, a chemometric approach (Simplex method) was used and extrapolated to establish a fully continuous-flow method. The reaction scope was also examined via the synthesis of several (het)aryl chlorides. Validation and scale-up of the process were also performed. A higher productivity was obtained with increased safety.

Efficient Transposition of the Sandmeyer Reaction from Batch to Continuous Process

 Institut de Chimie Organique et Analytique, Univ Orleans, UMR CNRS 7311, Rue de Chartres, BP 6759, 45067 CEDEX 2 Orléans, France
 ISOCHEM, 4 Rue Marc Sangnier, BP 16729, 45300 Pithiviers, France
§ Institut de Combustion, Aérothermique, Réactivité, et Environnement (ICARE), 1c, Avenue de la Recherche Scientifique, 45071 CEDEX 2 Orléans, France
Org. Process Res. Dev., Article ASAP
 
str1
1H NMR (250 MHz, Chloroform-d) δ 7.65 (dd, J = 2.1, 0.6 Hz, 1H, Har), 7.42 (dd, J = 8.7, 0.6 Hz, 1H, Har), 7.32 (dd, J = 8.7, 2.0 Hz, 1H, Har).
2,5-Dichloro-1,3-benzoxazole (33)
The reaction was carried out as described in general procedure B using 2-Amino-5-chlorobenzoxazole (221 mg, 1.31 mmol). After purification with silica flash chromatography (EP 100%), the product was isolated as a yellow oil (62 mg, 25%).
CAS number 3621-81-6.
1H NMR (250 MHz, Chloroform-d) δ 7.65 (dd, J = 2.1, 0.6 Hz, 1H, Har), 7.42 (dd, J = 8.7, 0.6 Hz, 1H, Har), 7.32 (dd, J = 8.7, 2.0 Hz, 1H, Har).
str1
 
13C NMR (101 MHz, Chloroform-d) δ 152.27 (C), 150.12 (C), 142.06 (C), 130.79 (C), 125.85 (CH), 119.78 (CH), 111.16 (CH).
 
HRMS [M + H]+ (EI) calcd for C7H4Cl2NO: 187.9664, found: 187.9663.
1H NMR PREDICT
str1 str2
13 C NMR PREDICT
str1 str2
/////////////