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, this will not stop me, Only God and death can..........
DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai), INDIA, worlddrugtracker, 29Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.8 Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution
n, सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये।औकात बस इतनी देना,कि औरों का भला हो जाये।...........P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Friday 14 August 2015

SACUBITRIL


Sacubitril skeletal.svg

Sacubitril, AHU 377


5-(Biphenyl-4-yl)-4(S)-(3-carboxypropionamido)-2(R)-methylbutyric acid ethyl ester
N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-2R-methyl butanoic acid ethyl ester
[1,​1'-​Biphenyl]​-​4-​pentanoic acid, γ-​[(3-​carboxy-​1-​oxopropyl)​amino]​-​α-​methyl-​, α-​ethyl ester, (αR,​γS)​-
  • [1,1'-Biphenyl]-4-pentanoic acid, γ-[(3-carboxy-1-oxopropyl)amino]-α-methyl-, ethyl ester, [S-(R*,S*)]-
  • (2R,4S)-4-[(3-Carboxy-1-oxopropyl)amino]-4-[(p-phenylphenyl)methyl]-2-methylbutanoic acid ethyl ester
  • (2R,4S)-5-(Biphenyl-4-yl)-4-[(3-carboxypropionyl)amino]-2-methylpentanoic acid ethyl ester
  • FormulaC24H29NO5
    MW411.49 g/mol
AHU377; AHU-377; Sacubitril; 149709-62-6; UNII-17ERJ0MKGI; Alpha-ethyl (alphaR,gammaS)-gamma-<(3-carboxy-1-oxopropyl)amino>-alpha-methyl<1,1'-biphenyl>-4-pentanoate

Sacubitril is an antihypertensive drug used in combination with valsartan. The combination drug, valsartan/sacubitril, known during trials as LCZ696 and marketed under the brand name, Entresto, is a treatment for heart failure.[1] It was approved under the FDA'spriority review process for use in heart failure on July 7, 2015.
AHU377.png

Mechanism of action

Sacubitril is a prodrug that is activated to LBQ657 by de-ethylation via esterases.[2] LBQ657 inhibits the enzyme neprilysin,[3] which is responsible for the degradation of atrial and brain natriuretic peptide, two blood pressure lowering peptides that work mainly by reducing blood volume.[4]
Sacubitril activation to LBQ657


SYNTHESIS


WO-2008031567

http://www.google.com/patents/WO2008031567A1?cl=en
the following steps:
Figure imgf000040_0001
Figure imgf000040_0002
and optionally the following additional steps:
Figure imgf000041_0001
Figure imgf000041_0002

 

Example 1 :
(E)-(R)-5-biphenyl-4-yl-4-fert-butoxycarbonylamino-2-methylpent-2-enoic acid
Figure imgf000047_0001
(E)-(R)-5-Biphenyl-4-yl-4-tert-butoxycarbonylamino-2-methylpent-2-enoic acid ethyl ester (CAS# 149709-59-1) is hydrolysed using lithium hydroxide in ethanol to yield (£)-(f?)-5-biphenyl-4-yl-4-te/t-butoxycarbonylamino-2-methylpent-2-enoic acid as a white solid. δH (400 MHz; DMSO) 1.31 (9H1 s, (CH3J3), 1.59 (3H, s, 1- CH3), 2.68 (1H, dd, J 6.8, 13.2, 5-HA), 2.86 (1H, m, 5-HB), 4.44 (1H, m, 4-H), 6.51 (1H1 d, J 9.2, 3-H), 7.16 (1H, d, J 8.0, NH), 7.26 (2H, d, J 8.0, Ar-ortho- H(Ph)), 7.31 (1H, t, J 7.6, Ar-(Ph)-para-H), 7.40 (2H, t, J 8.0, Ar-(Ph)-metø-H), 7.54 (2H, d, J 8.0, Ar-mefa-H(Ph), 7.60 (2H, d, J 7.6, Ar-(Ph)-ort/vo-H), 12.26 (1H, s, CO2H); m/z (+ESI) 404 ([MNa]+, 17%), 382 ([MHf, 2), 326 (10), 264 (100), 167 (13).
Example 2:
(2/?,4S)-5-biphenyl-4-yl-4-fert-butoxycarbonylamino-2-methylpentanoic acid in crystalline form [2(i-a)]
Figure imgf000047_0002
2(i-a) To a suspension of (E)-(f?)-5-biphenyl-4-yl-4-te/t-butoxycarbonylamino-2- methylpent-2-enoic acid [2(ii-a)] (200 g, 524.3 mmol) in degassed ethanol (900 ml) at 40 °C a solution of diiodo(p-cymene)ruthenium(ll) dimer (0.052 g, 0.0524 mmol) and (αf?,αf?)-2,2>-bis(α-Λ/,Λ/-dimethylaminophenylmethyl)-(S,S)- 1 ,1'-bis[di(3,5-dimethyl-4-methoxyphenyl)phosphine]ferrocene (= Mandyphos SL-M004-1) (0.116 g, 0.110 mmol) is added in degassed ethanol (100 ml). The solution is degassed using vacuum and a pressure of 20 bar hydrogen applied. The mixture is stirred at 40 0C for 6 h. Vessel is then purged with nitrogen. Ethanol (700 ml) is removed by distillation, lsopropyl acetate (600 ml) is added. Solvent (600 ml) is removed by distillation, lsopropyl acetate (600 ml) is added. Solvent (600 ml) is removed by distillation, lsopropyl acetate (300 ml) is added and the solution is heated to reflux. Heptane fraction (1200 ml) is added and the mixture is cooled to room temperature. The solid is collected by filtration and washed with heptane fraction-isopropyl acetate 2 : 1 mixture (360 ml). The solid is dried overnight at 50 0C under 1-50 mbar vacuum to afford the title compound as a white/off-white solid [Ratio 2(i-a) : 2(i-b) 99 : 1, as determined by HPLC analysis]. Mpt 146-147 0C; δH (500 MHz; DMSO) 1.07 (3H1 d, J 7.0, 1-CH3), 1.34 (9H, s, (CH3)3), 1-38 (1H, m, 3-HA), 1.77 (1H, m, 3-HB), 2.43 (1H, m, 2-H), 2.70 (2H, d, J 7.0, 5-H)1 3.69 (1 H, m, 4-H), 6.74 (1 H, d, J 9.0, NH)1 7.27 (2H, d, J 8.0, Ar-ortA;o-H(Ph)), 7.36 (1H, t, J 7.0, Ar-(Ph)-para-H), 7.46 (2H, t, J 7.5, Ar-(Ph)- meta-H), 7.57 (2H, d, J 8.0, Ar-mefa-H(Ph), 7.64 (2H, d, J 7.5, Ar-(Ph)-orfΛo-H), 12.01 (1H, s, CO2H); δc (500 MHz, DMSO) 18.1 (1-CH3), 28.3 [(CH3)3], 35.9 (2- C), 37.9 (3-C), 40.7 (5-C), 50.0 (4-C), 77.4 [(C(CH3)3], 126.3, 126.5, 127.2, 128.9, 129.8 (Ar-CH), 137.7 (Ar-/pso-C(Ph)), 138.3 (Ar-para-C(Ph)), 140.1 (Ar- (Ph)-/pso-C), 155.2 (NCO), 177.2 (CO2H); mlz (+ESI) 406 ([MNa]+, 6%), 384 ([MH]+, 31 ), 328 (100), 284 (19); Found: [MH]+, 384.21691. C23H30NO4 requires MH 384.21693

 ...........

PATENT
http://www.google.com/patents/EP0555175A1
      Example 1....THE SODIUM SALT
    • To a solution of N-(3-carbo(t)butoxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-2R-methylbutanoic acid ethyl ester (0.80 g) in 15 ml of CH2CI2 at room temperature are added 3 ml of trifluoroacetic acid. The mixture is stirred overnight and concentrated. The residue is dissolved in tetrahydrofuran (THF), and 6.5 ml of 1 N NaOH is added. The mixture is concentrated and triturated with ether. The solid can be recrystallized from methylene chloride-hexane to give sodium N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-2R-methyl butanoic acid ethyl ester melting at 159-160°C; [a]D20 = - 11.4° (methanol).
      Figure imgb0018SODIUM SALT
The starting material is prepared as follows:
    • A solution of a-t-BOC-(R)-tyrosine methyl ester (5.9 g, 20 mmol) and pyridine (8 mL, 100 mmol) in methylene chloride (30 mL) is cooled to 0-5°C. Trifluoromethanesulfonic anhydride (4 mL, 23 mmol) is added at 0-5°C, and the resulting mixture is held for another 30 minutes. The reaction mixture is diluted with water (60 mL) and methylene chloride (100 mL), and washed sequentially with 0.5 N sodium hydroxide solution (1 x 50 mL), water (1 x 60 mL), 10% citric acid solution (2 x 75 mL) and water (1 x 60 mL). The organic phase is dried over MgS04 and concentrated to an oil. The oil is purified by column chromatography (silica gel, hexane/ethyl acetate, 2:1 to give methyl(R)-2-(t-butoxycarbonylamino)-3-[4-(trifluoromethylsulfonyloxy)phenyl]-propionate which crystallizes on standing; m.p. 46-48°C; [a]20 D-36.010 (c=1, CHCI3).
    • Nitrogen is passed through a suspension of (R)-2-(t-butoxycarbonylamino)-3-[4-(trifluoromethylsulfonyloxy)-phenyl]-propionate (1.75mmol), phenylboronic acid (3.5 mmol), anhydrous potassium carbonate (2.63 mmol) and toluene (17 mL) for 15 minutes. Tetrakis(triphenyiphosphine)paiiadium(0) is added, and the mixture is heated at 85-90° for 3 hours. The reaction mixture is cooled to 25°C, diluted with ethyl acetate (17 mL) and washed sequentially with saturated sodium bicarbonate (1 x 20 mL), water (1 x 20 mL), 10% citric acid (1 x 20 mL), water (1 x 20 mL) and saturated sodium chloride solution (1 x 20 mL). The organic phase is concentrated, and the residue is purified by column chromatography (silica gel, hexane/ethyl acetate 2:1) to yield methyl (R)-2-(t-butoxycarbonylamino)-3-(p-phenylphenyl)-propionate which can also be called N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine methyl ester.
    • To a solution of N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine methyl ester (6.8 g) in 60 ml of THF and 20 ml of methanol are added 20 ml of aqueous 1 N sodium hydroxide solution. The mixture is stirred for 1 h at room temperature and then acidified with 21 ml of 1 N hydrochloric acid. The aqueous solution is extracted 3x with ethyl acetate. The combined organic extracts are dried (MgS04), filtered and concentrated to give N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine, m.p. 98-99°C; [a]2°D -18.59° (c=1, methanol).
    • To a solution of N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine (4.8 g) in 70 ml of methylene chloride (CH2CI2) at 0°C with 1.65 g of N,O-dimethylhydroxylamine HCI, 1.7 g of triethylamine and 2.85 g of hydroxybenzotriazole are added 5.37 g of 1-[3-(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride. The mixture is stirred 17 h at room temperature. The mixture is concentrated taken up in ethyl acetate (EtOAc) and washed with saturated sodium bicarbonate, 1N HCI and brine, then dried (MgS04), filtered and concentrated to give N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine N,O-dimethyl hydroxylamine amide.
    • To a 0°C solution of N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine N,O-dimethyl hydroxylamine amide (5.2 g) in 250 ml of diethyl ether are added 0.64 g of lithium aluminum hydride. The reaction is stirred for 30 min. and quenched with aqueous potassium hydrogen sulfate. The mixture is stirred for additional 5 min., poured onto 1N HCI, extracted (3x) with EtOAc, dried (MgS04), filtered, and concentrated to give N-(R)-4-t-butoxycarbonyl-(p-phenylphenyl)-alanine carboxaldehyde as a colorless oil.
    • To a 0°C solution of N-(R)-t-butoxycarbonyl-(p-phenylphenyl)-alanine carboxaldehyde (4.4 g) in 200 ml of CH2CI2are added 10 g of carboethoxyethylidene phenyl phosphorane. The mixture is warmed to room temperature, stirred for 1 h, washed with brine, dried (MgS04), filtered and concentrated. The residue is chromatographed on silica gel eluting with (1:2) ether:hexane to give N-t-butoxycarbonyl-(4R)-(p-phenylphenylme- thyl)-4-amino-2-methyl-2-butenoic acid ethyl ester.
    • A solution of N-t-butoxycarbonyl-(4R)-(p-phenylphenylmethyl)-4-amino-2-methyl-2-butenoic acid ethyl ester (4.2 g) in 400 ml of ethanol is suspended with 2.0 g of 5% palladium on charcoal and then is hydrogenated at 50 psi for 6h. The catalyst is removed by filtration and the filtrate is concentrated to give N-t-butoxycarbonyl(4S)-(p-phenylphenylmethyl)-4-amino-2-methylbutanoic acid ethyl ester as a 80:20 mixture of diastereomers.
    • To the N-t-butoxycarbonyl(4S)-(p-phenylphenylmethyl)-4-amino-2-methylbutanoic acid ethyl ester (4.2 g) in 40 ml of CH2CI2 at 0°C is bubbled dry hydrogen chloride gas for 15 min. The mixture is stirred 2 h and concentrated to give (4S)-(p-phenylphenylmethyl)-4-amino-2-methylbutanoic acid ethyl ester hydrochloride as a 80:20 mixture of diastereomers.
    • To a room temperature solution of the above amine salt (3.12 g) in 15 ml of CH2CI2 and 15 ml of pyridine are added 13.5 g of succinic anhydride. The mixture is stirred for 17 h, concentrated, dissolved in ethyl acetate, washed with 1N HCI and brine, and dried (MgS04) to give N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenyl- methyl)-4-amino-2-methylbutanoic acid ethyl ester as a 80:20 mixture of diastereomers.
    • The above N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-2-methylbutanoic acid ethyl ester diastereomeric mixture (3.9 g) and N,N-dimethylformamide-di-t-butyl acetal (8.8 ml) are heated at 80°C in 40 ml of toluene for 2 h. The mixture is poured onto ice- 1N HCI, extracted with ether, chromatographed on silica gel eluting with (2:1) toluene:ethyl acetate to give N-(3-carbo(t)butoxy-1-oxopropyl)-(4S)-(p-phenylphe- nylmethyl)-4-amino-2R-methylbutanoic acid ethyl ester as the more polar material and the corresponding (S,S) diastereomer as the less polar material.
Example 2.........THE ACID
  • To a solution of N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-(2R)-methylbutanoic acid ethyl ester (0.33 g) in 20 ml of (1:1) ethanol:tetrahydrofuran (THF) at room temperature are added 5 ml of 1 N sodium hydroxide solution (NaOH) and stirred for 17 h. The mixture is concentrated, dissolved in water and washed with ether. The aqueous layer is acidified with 1 N hydrochloric acid (HCI), extracted 3x with ethyl acetate (EtOAc), dried over magnesium sulfate (MgS04), filtered and concentrated. The residue is triturated with ether to yield N-(3-carboxy-1-oxopropyl)-(4S)-(p-phenylphenylmethyl)-4-amino-(2R)-methylbutanoic acid melting at 158-164°C, [α]D 20= -23.5° (methanol).
.........................
CN 104557600
http://www.google.com/patents/CN104557600A?cl=zh
Figure CN104557600AC00021
United States Patent US5217996 and international patent W02008031567, W02010136474 and W02012025501 reported a synthetic route follows to the chiral amino alcohols as raw materials, oxidized to the aldehyde, Victoria ladder tin reaction, chiral hydrogenation and amidation condensation reaction to obtain the objective product.
Figure CN104557600AD00032
In addition, the international patent TO2008083967, TO2011088797, TO2012025502 and TO2014198195 reported that another type of preparation. The route through the 2-oxo-proline as raw material, carboxyl activating biphenyl substituted carbonyl reduction, chiral methylation, ring-opening reaction and amide condensation reaction to obtain the objective product.

Figure CN104557600AD00041


Figure CN104557600AD00051
Example Eight:
in the reaction flask was added (2R, 4S) -2- methyl-4-amino -5- (l, P- biphenyl-4-yl) - pentanoic acid ethyl ester (VII) (1.55g, 5mmol ), Jie of pyridine (1.2g, 15mmol) and dichloromethane burning 25mL, stirring to dissolve, butyric anhydride (1.0g, 10mmol), was heated to 4〇-45 ° C, the reaction was stirred for 6 hours. Fill Gaudin anhydride (0. 5g, 5mmol), the reaction was continued for 4 hours and the end of the reaction by TLC. Concentrated under reduced pressure, the residue was recrystallized from ethyl acetate and n-hexane to give an off-white solid sand sacubitril Kubica song (I) L 6g, a yield of 77.9%;
1H NMR (CDCl3) S 7.51 (d, 2H), 7.46 ( d, 2H), 7.36 (m, 2H), 7. 27 (m, 1H), 7. 17 (d, 2H), 5. 72 (d, 1H), 4. 19 (brs, 1H), 4. 06 (q, 2H), 2. 87-2. 72 (m, 2H), 2. 62-2. 54 (m, 2H), 2. 49 (brs, 1H), 2. 43-2. ​​33 ( m, 2H), I 88 (m 1H), I 54-1 43 (m, 1H), I. 18 (t, 3H), l 10 (d, 3H);.....
FAB-MSm / z : 412 [M + H] +.
......................
http://www.google.com/patents/WO2014198195A1?cl=en
Example 7
(2 Standby

Acetyl chloride (1 mL) 0 ° C was added ethanol (10 mL), and at room temperature for 0.5 hours, the compound (3R, 5S) -5- biphenyl-4-methyl-1- (2,2- methyl-propionyl) -3-methyl pyrrolidone (520 mg, 1.49 mmol), the reaction was refluxed for 3 days. After cooling to room temperature, and concentrated. The reaction mixture was dissolved in 8 mL of dichloromethane and pyridine 1: 1 mixed solution, and then butyryl anhydride (223 mg, 2.23 mmol). 30 ° C overnight. LC-MS detection, a small amount of starting material remaining, fill Gading anhydride (75 mg, 0.74 mmol), continue to reflect four hours. Concentrated and reverse phase column chromatography to give a white foam solid (2R, 4S) -5- biphenyl-4-yl-4- (3-carboxy - propionylamino) -2-methyl - acetic acid ester a (355 mg, 58%) and white solid (2R, 4S) -5- biphenyl-4-yl-4- (3-carboxy - propionylamino) -2-methyl - pentanoic acid b ( 13 mg, 2.3%).
a: 1H MR (400 MHz, CDCl 3 ) [delta] 7.51 (d, = 7.8 Hz, 2H), 7.46 (d, = 7.8 Hz, 2H), 7.36 (t, J = 7.6 Hz, 2H), 7.27 (t, J = 7.2 Hz, IH), 7.17 (d, J = 7.9 Hz, 2H), 5.72 (d, J = 8.1 Hz, IH), 4.19 (brs, IH), 4.06 (q, J = 7.0 Hz, 2H) , 2.87-2.72 (m, 2H), 2.62-2.54 (m, 2H), 2.49 (brs, IH), 2.43-2.33 (m, 2H), 1.88 (ddd, = 13.2, 9.5, 3.9 Hz, IH), 1.54-1.43 (m, IH), 1.18 (t, = 7.0 Hz, 3H), 1.10 (d, = 7.2 Hz, 3H).
LC-MS: t R = 3.43 min; [M + H] +: 412.0.

........................
Paper
JOURNAL OF MEDICINAL CHEMISTRY, vol. 38, no. 10, 1995, pages 1689-1700,
http://pubs.acs.org/doi/pdf/10.1021/jm00010a014
NOTE-----------ACID
(aR,yS)-y-[ (3-Carbo-1-oxopropyl)aminol-a-methyl- [l,l'-biphenyllpentanoic Acid (21a).
To the sodium salt of 19a (0.73 g, 1.68 mM) in 20 mL of THF:EtOH was added 1 N NaOH (5.0 mL, 5.0 "01). The reaction mixture was stirred overnight and then washed with ether. The aqueous layer was acidified with 1 N HCI, re-extracted with EtOAc (3 x 10 mL), dried (MgSO& and evaporated to dryness. The solid was recrystallized from ethanol to yield 435 mg of 21a ACID OF SACUBITRIL
melting at 165-167 "C:
[a] D25~ -28.73 (c = 10.1 in MeOH);
'H NMR, DMSOD6
PPM 12.0, (s, 2H), 7.75 (d, 1H), 7.62 (d, 2H), 7.55 (d, 2H), 7.45 (t, 2H), 7.32 (t, lH), 7.25 (d, 2H), 4.92 (m, lH), 2.70 (d, 2H), 2.35 (t, 3H), 2.25 (m, 2H), 1.75 (m, lH), 1.32 (m, lH), 1.03 (d, 3H).
Anal. (C22H25N05) C,H,N
NMR PREDICT
SACUBITRIL CHEMDOOCL

 1H NMR PREDICT



1HNMR GRAPH 1HNMR VAL
13C NMR PREDICT
13C NMR GRAPH 13C NMR VAL

 

COSY PREDICT

COSY

................

Formula Image

 

 

 

 

 

NMR.....http://www.chemietek.com/Files/Line3/CHEMIETEK,%20AHU-377%20,%20Lot%2001,%20NMR-MeOD,%201.1.pdf


Mol. Formula:C24H29NO5 ∙ C4H11NO3
MW:532.6
HPLC.........http://www.chemietek.com/Files/Line2/CHEMIETEK,%20AHU-377%20,%20Lot%2001,%20HPLC.pdf

References

  1. John J.V. McMurray, Milton Packer, Akshay S. Desai, et al. for the PARADIGM-HF Investigators and Committees (August 30, 2014)."Angiotensin–Neprilysin Inhibition versus Enalapril in Heart Failure". N Eng J Med 371. doi:10.1056/NEJMoa1409077.
  2. Solomon, SD. "HFpEF in the Future: New Diagnostic Techniques and Treatments in the Pipeline". Boston. p. 48. Retrieved 2012-01-26.
  3. Gu, J.; Noe, A.; Chandra, P.; Al-Fayoumi, S.; Ligueros-Saylan, M.; Sarangapani, R.; Maahs, S.; Ksander, G.; Rigel, D. F.; Jeng, A. Y.; Lin, T. H.; Zheng, W.; Dole, W. P. (2009). "Pharmacokinetics and Pharmacodynamics of LCZ696, a Novel Dual-Acting Angiotensin Receptor-Neprilysin Inhibitor (ARNi)". The Journal of Clinical Pharmacology 50 (4): 401–414. doi:10.1177/0091270009343932.PMID 19934029. edit
  4. Schubert-Zsilavecz, M; Wurglics, M. "Neue Arzneimittel 2010/2011." (in German)






WO2004085378A1 *Mar 15, 2004Oct 7, 2004Joseph D Armstrong IiiProcess for the preparation of chiral beta amino acid derivatives by asymmetric hydrogenation
WO2006057904A1 *Nov 18, 2005Jun 1, 2006Merck & Co IncStereoselective preparation of 4-aryl piperidine amides by asymmetric hydrogenation of a prochiral enamide and intermediates of this process
WO2006069617A1 *Dec 5, 2005Jul 6, 2006Dsm Fine Chem Austria GmbhProcess for transition metal-catalyzed asymmetric hydrogenation of acrylic acid derivatives, and a novel catalyst system for asymmetric transition metal catalysis
US5217996 *Jan 22, 1992Jun 8, 1993Ciba-Geigy CorporationBiaryl substituted 4-amino-butyric acid amides

NON-PATENT CITATIONS
Reference
1*KSANDER, GARY M. ET AL: "Dicarboxylic Acid Dipeptide Neutral Endopeptidase Inhibitors" JOURNAL OF MEDICINAL CHEMISTRY, vol. 38, no. 10, 1995, pages 1689-1700, XP002340280 cited in the application
PatentSubmittedGranted
ORGANIC COMPOUNDS [US2009156585]2009-06-18
METHODS OF TREATMENT AND PHARMACEUTICAL COMPOSITION [US8101659]2008-10-232012-01-24
Substituted Aminobutyric Derivatives as Neprilysin Inhibitors [US2010305145]2010-12-02
PROCESS FOR PREPARING BIARYL SUBSTITUTED 4-AMINO-BUTYRIC ACID OR DERIVATIVES THEREOF AND THEIR USE IN THE PRODUCTION OF NEP INHIBITORS [US2009326066]2009-12-31
Process for preparing 5-biphenyl-4-amino-2-methyl pentanoic acid [US8115016]2010-05-062012-02-14
Methods of treatment and pharmaceutical composition [US7468390]2003-07-312008-12-23
Process for Preparing 5-biphenyl-4-amino-2-methyl Pentanoic Acid [US2014249320]2014-03-252014-09-04
Substituted Aminobutyric Derivatives as Neprilysin Inhibitors [US2012252830]2012-06-072012-10-04
Process for preparing 5-biphenyl-4-amino-2-methyl pentanoic acid [US8716495]2011-12-212014-05-06
f
Sacubitril
Sacubitril skeletal.svg
Systematic (IUPAC) name
4-{[(2S,4R)-1-(4-Biphenylyl)-5-ethoxy-4-methyl-5-oxo-2-pentanyl]amino}-4-oxobutanoic acid
Clinical data
Legal status
  • Investigational
Identifiers
CAS Registry Number149709-62-6
ATC codeNone
PubChemCID: 9811834
ChemSpider7987587
SynonymsAHU-377; AHU377
Chemical data
FormulaC24H29NO5
Molecular mass411.49 g/mol

Relevant Clinical Literature
UK Guidance
Regulatory Literature
Other Literature









 सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये। औकात बस इतनी देना, कि औरों का भला हो जाये।
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE


Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK
Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus
Join me on Researchgate

Anthony Melvin Crasto Dr.

  amcrasto@gmail.com

09b37-misc2b027LIONEL MY SON
He was only in first standard in school when I was hit by a deadly one in a million spine stroke called acute transverse mylitis, it made me 90% paralysed and bound to a wheel chair, Now I keep him as my source of inspiration and helping millions, thanks to millions of my readers who keep me going and help me to keep my son happy


सुकून उतना ही देना प्रभू, जितने से
जिंदगी चल जाये।
औकात बस इतनी देना,
कि औरों का भला हो जाये।
Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL  


////// antihypertensive drugHeart Failure, Sacubitril, AHU 377

Monday 3 August 2015

LENVATINIB


Lenvatinib skeletal.svg

Lenvatinib

For the treatment of patients with progressive radioiodine-refractory, differentiated thyroid cancer (RR-DTC).
CAS  417716-92-8,
 CAS 857890-39-2 (lenvatinib mesylate)
E 7080, ER-203492-00, E7080, E 7080,
4-[3-Chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6-quinolinecarboxamide
Molecular Formula: C21H19ClN4O4
Molecular Weight: 426.85296

Eisai Co., Ltd INNOVATOR
Eisai R&D Management Co., Ltd.
 エーザイ・アール・アンド・ディー・マネジメント株式会社




Lenvatinib was granted Orphan Drug Designation for thyroid cancer by the health authorities in Japan in 2012, and in Europe and the U.S in 2013. The first application for marketing authorization of lenvatinib in the world was submitted in Japan on June 2014. Eisai is planning to submit applications for marketing authorization in Europe and the U.S. in the second quarter of fiscal 2014. Lenvatinib is an oral multiple receptor tyrosine kinase (RTK) inhibitor with a novel binding mode that selectively inhibits the kinase activities of vascular endothelial growth factor receptors (VEGFR), in addition to other proangiogenic and oncogenic pathway-related RTKs including fibroblast growth factor receptors (FGFR), the platelet-derived growth factor (PDGF) receptor PDGFRalpha, KIT and RET that are involved in tumor proliferation. This potentially makes lenvatinib a first-in-class treatment, especially given that it simultaneously inhibits the kinase activities of FGFR as well as VEGFR.

  Eisai's lenvatinib to get speedy review in Europe
  LENVATINIB BASE
  COSY PREDICT COSY  LENVA BASE
Systematic (IUPAC) name
4-[3-chloro-4-(cyclopropylcarbamoylamino)phenoxy]-7-methoxy-quinoline-6-carboxamide
Clinical data
Legal status Prescription only
Identifiers
CAS number
ATC code None
PubChem CID 9823820
ChemSpider 7999567 Yes
UNII EE083865G2 Yes
Chemical data
Formula C21H19ClN4O4 
Mol. mass 426.853 g/mol

Lenvatinib (E7080) is a multi-kinase inhibitor that is being investigated for the treatment of various types of cancer by Eisai Co. It inhibits both VEGFR2 and VEGFR3 kinases.[1] The substence was granted orphan drug status for the treatment of various types of thyroid cancer that do not respond toradioiodine; in the US and Japan in 2012 and in Europe in 2013[2] and is now approved for this use.

 

Clinical trials

Lenvatinib has had promising results from a phase I clinical trial in 2006[3] and is being tested in several phase II trials as of October 2011, for example against hepatocellular carcinoma.[4] After a phase II trial testing the treatment of thyroid cancer has been completed with modestly encouraging results,[5] the manufacturer launched a phase III trial in March 2011.[6] Chemical structure for Lenvatinib Lenvatinib Mesilate Molecular formula: C21H19ClN4O4,CH4O3S =523.0. CAS: 857890-39-2. UNII code: 3J78384F61.

About the Lenvatinib (E7080) Phase II Study The open-label, global, single-arm Phase II study of multi-targeted kinase inhibitor lenvatinib (E7080) in advanced radioiodine (RAI)-refractory differentiated thyroid cancer involved 58 patients with advanced RAI refractory DTC (papillary, follicular or Hurthle Cell) whose disease had progressed during the prior 12 months. (Disease progression was measured using Response Evaluation Criteria in Solid Tumors (RECIST).) The starting dose of lenvatinib was 24 mg once daily in repeated 28 day cycles until disease progression or development of unmanageable toxicities.

  2.   About Thyroid Cancer Thyroid cancer refers to cancer that forms in the tissues of the thyroid gland, located at the base of the throat or near the trachea. It affects more women than men and usually occurs between the ages of 25 and 65. The most common types of thyroid cancer, papillary and follicular (including Hurthle Cell), are classified as differentiated thyroid cancer and account for 95 percent of all cases. While most of these are curable with surgery and radioactive iodine treatment, a small percentage of patients do not respond to therapy. 3.   About Lenvatinib (E7080) Lenvatinib is multi-targeted kinase inhibitor with a unique receptor tyrosine kinase inhibitory profile that was discovered and developed by the Discovery Research team of Eisai's Oncology Unit using medicinal chemistry technology. As an anti-angiogenic agent, it inhibits tyrosine kinase of the VEGF (Vascular Endothelial Growth Factor) receptor, VEGFR2, and a number of other types of kinase involved in angiogenesis and tumor proliferation in balanced manner. It is a small molecular targeting drug that is currently being studied in a wide array of cancer types. 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (additional name: 4-[3-chloro-4-(N′-cyclopropylureido)phenoxy]-7-methoxyquinoline-6-carboxamide) is known to exhibit an excellent angiogenesis inhibition as a free-form product, as described in Example 368 of Patent Document 1. 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide is also known to exhibit a strong inhibitory action for c-Kit kinase (Non-Patent Document 1, Patent Document 2). However, there has been a long-felt need for the provision of a c-Kit kinase inhibitor or angiogenesis inhibitor that has high usability as a medicament and superior characteristics in terms of physical properties and pharmacokinetics in comparison with the free-form product of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide.
 [Patent Document 1] WO 02/32872
 [Patent Document 2] WO 2004/080462
 [Non-Patent Document 1] 95th Annual Meeting Proceedings, AACR (American Association for Cancer Research), Volume 45, Page 1070-1071, 2004

 ............................. PATENT
  http://www.google.co.in/patents/US8058474
 EXAMPLES Examples will now be described to facilitate understanding of the invention, but the invention is not limited to these examples. Example 1Phenyl N-(2-chloro-4-hydroxyphenyl)carbamate
After suspending 4-amino-3-chlorophenol (23.7 g) in N,N-dimethylformamide (100 mL) and adding pyridine (23.4 mL) while cooling on ice, phenyl chloroformate (23.2 ml) was added dropwise below 20° C. Stirring was performed at room temperature for 30 minutes, and then water (400 mL), ethyl acetate (300 mL) and 6N HCl (48 mL) were added, the mixture was stirred and the organic layer was separated. The organic layer was washed twice with 10% brine (200 mL), and dried over magnesium sulfate. The solvent was removed to give 46 g of the title compound as a solid. 1H-NMR (CDCl3): 5.12 (1h, br s), 6.75 (1H, dd, J=9.2, 2.8 Hz), 6.92 (1H, d, J=2.8 Hz), 7.18-7.28 (4H, m), 7.37-7.43 (2H, m), 7.94 (1H, br s) Example 21-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea
After dissolving phenyl N-(2-chloro-4-hydroxyphenyl)carbamate in N,N-dimethylformamide (100 mL), cyclopropylamine (22.7 mL) was added while cooling on ice and the mixture was stirred overnight at room temperature. Water (400 mL), ethyl acetate (300 mL) and 6N HCl (55 mL) were then added, the mixture was stirred and the organic layer was separated. The organic layer was washed twice with 10% brine (200 mL), and dried over magnesium sulfate. Prism crystals obtained by concentrating the solvent were filtered and washed with heptane to give 22.8 g of the title compound (77% yield from 4-amino-3-chlorophenol). 1H-NMR (CDCl3): 0.72-0.77 (2H, m), 0.87-0.95 (2H, m), 2.60-2.65 (1H, m), 4.89 (1H, br s), 5.60 (1H, br s), 6.71 (1H, dd, J=8.8, 2.8 Hz), 6.88 (1H, d, J=2.8 Hz), 7.24-7.30 (1H, br s), 7.90 (1H, d, J=8.8H) Example 34-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide
To dimethylsulfoxide (20 mL) were added 7-methoxy-4-chloro-quinoline-6-carboxamide (0.983 g), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (1.13 g) and cesium carbonate (2.71 g), followed by heating and stirring at 70° C. for 23 hours. After the reaction mixture was allowed to cool down to room temperature, water (50 mL) was added, and the produced crystals were collected by filtration to give 1.56 g of the title compound (88% yield). 1H-NMR (d6-DMSO): 0.41 (2H, m), 0.66 (2H, m), 2.56 (1H, m), 4.01 (3H, s), 6.51 (1H, d, J=5.6 Hz), 7.18 (1H, d, J=2.8 Hz), 7.23 (1H, dd, J=2.8, 8.8 Hz), 7.48 (1H, d, J=2.8 Hz), 7.50 (1H, s), 7.72 (1H, s), 7.84 (1H, s), 7.97 (1H, s), 8.25 (1H, d, J=8.8 Hz), 8.64 (1H, s), 8.65 (1H, d, J=5.6 Hz) 
Example 44-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide In a reaction vessel were placed 7-methoxy-4-chloro-quinoline-6-carboxamide (5.00 kg, 21.13 mol), dimethylsulfoxide (55.05 kg), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (5.75 kg, 25.35 mol) and potassium t-butoxide (2.85 kg, 25.35 mol) in that order, under a nitrogen atmosphere. After stirring at 20° C. for 30 minutes, the temperature was raised to 65° C. over a period of 2.5 hours. After stirring at the same temperature for 19 hours, 33% (v/v) acetone water (5.0 L) and water (10.0 L) were added dropwise over a period of 3.5 hours. Upon completion of the dropwise addition, the mixture was stirred at 60° C. for 2 hours, and 33% (v/v) acetone water (20.0 L) and water (40.0 L) were added dropwise at 55° C. or higher over a period of 1 hour. After then stirring at 40° C. for 16 hours, the precipitated crystals were collected by filtration using a nitrogen pressure filter, and the crystals were washed with 33% (v/v) acetone water (33.3 L), water (66.7 L) and acetone (50.0 L) in that order. The obtained crystals were dried at 60° C. for 22 hours using a conical vacuum drier to give 7.78 kg of the title compound (96.3% yield).
 ..............................

  SYNTHESIS SYN YAOPHA


1H NMR PREDICT NMR 1H GRAPH NMR 1H VAL

13 C NMR PREDICT

NMR 13C GRAPH NMR 13C VAL

.......................

PATENT
 http://www.google.co.in/patents/US7253286
 EX 368 Figure US07253286-20070807-C00838     Example 368 4-(3-Chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide The title compound (22.4 mg, 0.052 mmol, 34.8%) was obtained as white crystals from phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate (70 mg, 0.15 mmol) and cyclopropylamine, by the same procedure as in Example 11. 1H-NMR Spectrum (DMSO-d6) δ (ppm): 0.41 (2H, m), 0.66 (2H, m), 2.56 (1H, m), 4.01 (3H, s), 6.51 (1H, d, J=5.6 Hz), 7.18 (1H, d, J=2.8 Hz), 7.23 (1H, dd, J=2.8, 8.8 Hz), 7.48 (1H, d, J=2.8 Hz), 7.50 (1H, s), 7.72 (1H, s), 7.84 (1H, s), 7.97 (1H, s), 8.25 (1H, d, J=8.8 Hz), 8.64 (1H, s), 8.65 (1H, d, J=5.6 Hz). The starting material was synthesized in the following manner. Production Example 368-1Phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate The title compound (708 mg, 1.526 mmol, 87.4%) was obtained as light brown crystals from 4-(4-amino-3-chlorophenoxy)-7-methoxy-6-quinolinecarboxamide (600 mg, 1.745 mmol), by the same procedure as in Production Example 17. 1H-NMR Spectrum (CDCl3) δ (ppm): 4.14 (3H, s), 5.89 (1H, br), 6.50 (1H, d, J=5.6 Hz), 7.16 (2H, dd, J=2.4, 8.8 Hz), 7.22–7.30 (4H, m), 7.44 (2H, m), 7.55 (1H, s), 7.81 (1H, br), 8.31 (1H, d, J=8.8 Hz), 8.68 (1H, d, J=5.6 Hz), 9.27 (1H, s).

 ........................

  CRYSTALLINE FORM

  http://www.google.co.in/patents/US7612208 Preparation Example 1 Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (1) Phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate (17.5 g, 37.7 mmol) disclosed in WO 02/32872 was dissolved in N,N-dimethylformamide (350 mL), and then cyclopropylamine (6.53 mL, 94.25 mmol) was added to the reaction mixture under a nitrogen atmosphere, followed by stirring overnight at room temperature. To the mixture was added water (1.75 L), and the mixture was stirred. Precipitated crude crystals were filtered off, washed with water, and dried at 70° C. for 50 min. To the obtained crude crystals was added ethanol (300 mL), and then the mixture was heated under reflux for 30 min to dissolve, followed by stirring overnight to cool slowly down to room temperature. Precipitated crystals was filtered off and dried under vacuum, and then further dried at 70° C. for 8 hours to give the titled crystals (12.91 g; 80.2%). Preparation Example 2Preparation of 4-(3-cloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (2) (1) Preparation of phenyl N-(2-chloro-4-hydroxyphenyl)carbamate
To a suspension of 4-amino-3-chlorophenol (23.7 g) in N,N-dimethylformamide (100 mL) was added pyridine (23.4 mL) while cooling in an ice bath, and phenyl chloroformate (23.2 mL) was added dropwise below 20° C. After stirring at room temperature for 30 min, water (400 mL), ethyl acetate (300 mL), and 6N-HCl (48 mL) were added and stirred. The organic layer was separated off, washed twice with a 10% aqueous sodium chloride solution (200 mL), and dried over magnesium sulfate. The solvent was evaporated to give 46 g of the titled compound as a solid.
  • 1H-NMR Spectrum (CDCl3) δ(ppm): 5.12 (1H, br s), 6.75 (1H, dd, J=9.2, 2.8 Hz), 6.92 (1H, d, J=2.8 Hz), 7.18-7.28 (4H, m), 7.37-7.43 (2H, m), 7.94 (1H, br s). (2) Preparation of 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea
To a solution of phenyl N-(2-chloro-4-hydroxyphenyl)carbamate in N,N-dimethylformamide (100 mL) was added cyclopropylamine (22.7 mL) while cooling in an ice bath, and the stirring was continued at room temperature overnight. Water (400 mL), ethyl acetate (300 mL), and 6N-HCl (55 mL) were added thereto, and the mixture was stirred. The organic layer was then separated off, washed twice with a 10% aqueous sodium chloride solution (200 mL), and dried over magnesium sulfate. The solvent was evaporated to give prism crystals, which were filtered off and washed with heptane to give 22.8 g of the titled compound (yield from 4-amino-3-chlorophenol: 77%).
  • 1H-NMR Spectrum (CDCl3) δ(ppm): 0.72-0.77 (2H, m), 0.87-0.95 (2H, m), 2.60-2.65 (1H, m), 4.89 (1H, br s), 5.60 (1H, br s), 6.71 (1H, dd, J=8.8, 2.8 Hz), 6.88 (1H, d, J=2.8 Hz), 7.24-7.30 (1H, br s), 7.90 (1H, d, J=8.8 Hz) (3) Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide
To dimethyl sulfoxide (20 mL) were added 7-methoxy-4-chloroquinoline-6-carboxamide (0.983 g), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (1.13 g) and cesium carbonate (2.71 g), and the mixture was heated and stirred at 70° C. for 23 hours. The reaction mixture was cooled to room temperature, and water (50 mL) was added, and the resultant crystals were then filtered off to give 1.56 g of the titled compound (yield: 88%). Preparation Example 3Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (3) 7-Methoxy-4-chloroquinoline-6-carboxamide (5.00 kg, 21.13 mol), dimethyl sulfoxide (55.05 kg), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea 5.75 kg, 25.35 mol) and potassium t-butoxide (2.85 kg, 25.35 mol) were introduced in this order into a reaction vessel under a nitrogen atmosphere. The mixture was stirred for 30 min at 20° C., and the temperature was raised to 65° C. over 2.5 hours. The mixture was stirred at the same temperature for 19 hours. 33% (v/v) acetone-water (5.0 L) and water (10.0 L) were added dropwise over 3.5 hours. After the addition was completed, the mixture was stirred at 60° C. for 2 hours. 33% (v/v) acetone-water (20.0 L) and water (40.0 L) were added dropwise at 55° C. or more over 1 hour. After stirring at 40° C. for 16 hours, precipitated crystals were filtered off using a nitrogen pressure filter, and was washed with 33% (v/v) acetone-water (33.3 L), water (66.7 L), and acetone (50.0 L) in that order. The obtained crystals were dried at 60° C. for 22 hours using a conical vacuum dryer to give 7.78 kg of the titled compound (yield: 96.3%). 1H-NMR chemical, shift values for 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamides obtained in Preparation Examples 1 to 3 corresponded to those for 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide disclosed in WO 02/32872. Example 5 A Crystalline Form of the Methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form A) (Preparation Method 1) In a mixed solution of methanol (14 mL) and methanesulfonic acid (143 μL, 1.97 mmol) was dissolved 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (700 mg, 1.64 mmol) at 70° C. After confirming the dissolution of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide, the reaction mixture was cooled to room temperature over 5.5 hours, further stirred at room temperature for 18.5 hours, and crystals were filtered off. The resultant crystals were dried at 60° C. to give the titled crystals (647 mg). (Preparation Method 2) In a mixed solution of acetic acid (6 mL) and methanesulfonic acid (200 μL, 3.08 mmol) was dissolved 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (600 mg, 1.41 mmol) at 50° C. After confirming the dissolution of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide, ethanol (7.2 mL) and seed crystals of a crystalline form of the methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form A) (12 mg) were added in this order to the reaction mixture, and ethanol (4.8 mL) was further added dropwise over 2 hours. After the addition was completed, the reaction mixture was stirred at 40° C. for 1 hour then at room temperature for 9 hours, and crystals were filtered off. The resultant crystals were dried at 60° C. to give the titled crystals (545 mg). Example 6A Crystalline Form of the Methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form B) A crystalline form of the acetic acid solvate of the methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form I) (250 mg) obtained in Example 10 was dried under aeration at 30° C. for 3 hours and at 40° C. for 16 hours to give the titled crystals (240 mg).........MORE IN PATENT

...................................

 PATENT
https://www.google.com/patents/WO2014098176A1?cl=en
  According to the present invention 4- (3-chloro-4- (cyclopropylamino-carbonyl) aminophenoxy) -7-methoxy-6-quinolinecarboxamide amorphous is excellent in solubility in water. Example 1 4- (3-chloro-4- (cyclopropylamino-carbonyl) aminophenoxy) -7-methoxy-6-quinolinecarboxamide manufacture of amorphous amide 4- (3-chloro-4- (cyclopropylamino-carbonyl) amino phenoxy) -7-methoxy-6-quinolinecarboxamide B-type crystals (Patent Document 2) were weighed to 300mg, is placed in a beaker of 200mL volume, it was added tert- butyl alcohol (tBA) 40mL. This was heated to boiling on a hot plate, an appropriate amount of tBA to Compound A is dissolved, water was added 10mL. Then, the weakened heated to the extent that the solution does not boil, to obtain a sample solution. It should be noted, finally the solvent amount I was 60mL. 200mL capacity eggplant type flask (egg-plant shaped flask), and rotated in a state of being immersed in ethanol which had been cooled with dry ice. It was added dropwise a sample solution into the interior of the flask and frozen. After freezing the sample solution total volume, to cover the opening of the flask in wiping cloth, and freeze-dried. We got an amorphous A of 290mg.

  Patent Document 2: US Patent Application Publication No. 2007/0117842 Patent specification
Amorphous A 13 C-solid state NMR spectrum in Figure 2, the chemical shifts and I are shown in Table 3.
 
[Table 3] *: peak of t- butyl alcohol

  .............................
  Paper
 ACS Medicinal Chemistry Letters (2015), 6(1), 89-94 http://pubs.acs.org/doi/full/10.1021/ml500394m

 .................
  Paper
Journal of Pharmaceutical and Biomedical Analysis (2015), 114, 82-87 http://www.sciencedirect.com/science/article/pii/S0731708515002940


  KEEP WATCHING WILL BE UPDATED.............most of my posts are updated regularly

References

  1.  Matsui, J.; Funahashi, Y.; Uenaka, T.; Watanabe, T.; Tsuruoka, A.; Asada, M. (2008). "Multi-Kinase Inhibitor E7080 Suppresses Lymph Node and Lung Metastases of Human Mammary Breast Tumor MDA-MB-231 via Inhibition of Vascular Endothelial Growth Factor-Receptor (VEGF-R) 2 and VEGF-R3 Kinase". Clinical Cancer Research 14 (17): 5459–65.doi:10.1158/1078-0432.CCR-07-5270. PMID 18765537.
  2.  "Phae III trial shows lenvatinib meets primary endpoint of progression free surival benefit in treatment of radioiodine-refactory differentiated thyroid cancer". Eisai. 3 February 2014.
  3.  Glen, H; D. Boss; T. R. Evans; M. Roelvink; J. M. Saro; P. Bezodis; W. Copalu; A. Das; G. Crosswell; J. H. Schellens (2007). "A phase I dose finding study of E7080 in patients (pts) with advanced malignancies". Journal of Clinical Oncology, ASCO Annual Meeting Proceedings Part I 25 (18S): 14073.
  4.  ClinicalTrials.gov NCT00946153 Study of E7080 in Patients With Advanced Hepatocellular Carcinoma (HCC)
  5.  Gild, M. L.; Bullock, M.; Robinson, B. G.; Clifton-Bligh, R. (2011). "Multikinase inhibitors: A new option for the treatment of thyroid cancer". Nature Reviews Endocrinology 7 (10): 617–624.doi:10.1038/nrendo.2011.141. PMID 21862995. edit
  6.  ClinicalTrials.gov NCT01321554 A Trial of E7080 in 131I-Refractory Differentiated Thyroid Cancer
UPDATES
EXTRAS
...............

 Martin Schlumberger et al. A phase 3, multicenter, double-blind, placebo-controlled trial of lenvatinib(E7080) in patients with 131I-refractory differentiated thyroid cancer (SELECT). 2014 ASCO Annual Meeting. Abstract Number:LBA6008. Presented June 2, 2014. Citation: J Clin Oncol 32:5s, 2014 (suppl; abstr LBA6008). Clinical trial information: NCT01321554. Bando, Masashi. Quinoline derivative-​containing pharmaceutical composition. PCT Int. Appl. (2011), WO 2011021597 A1 Tomohiro Matsushima, four Nakamura, Kazuhiro Murakami, Atsushi Hoteido, Yusuke Ayat, Naoko Suzuki, Itaru Arimoto, Pinche Hirose, Masaharu Gotoda.Has excellent characteristics in terms of physical properties (particularly, dissolution rate) and pharmacokinetics (particularly, bioavailability), and is extremely useful as an angiogenesis inhibitor or c-Kit kinase inhibitor. US patent number US7612208  Also published as: CA2426461A1, CA2426461C, CN1308310C, CN1478078A, CN101024627A, DE60126997D1, DE60126997T2, DE60134679D1, DE60137273D1, EP1415987A1, EP1415987A4, EP1415987B1, EP1506962A2, EP1506962A3, EP1506962B1, EP1777218A1, EP1777218B1 , US7612092, US7973160, US8372981, US20040053908, US20060160832, US20060247259, US20100197911, US20110118470, WO2002032872A1, WO2002032872A8.Publication date: Aug 7, 2007 Original Assignee: Eisai Co., Ltd Funahashi, Yasuhiro et al.Preparation of urea derivatives containing nitrogenous aromatic ring compounds as inhibitors of angiogenesis. US patent number US7253286, Also published as:CA2426461A1, CA2426461C, CN1308310C, CN1478078A, CN101024627A, DE60126997D1, DE60126997T2, DE60134679D1, DE60137273D1, EP1415987A1, EP1415987A4, EP1415987B1, EP1506962A2, EP1506962A3, EP1506962B1, EP1777218A1, EP1777218B1, US7612092, US7973160, US8372981, US20040053908, US20060160832, US20060247259, US20100197911, US20110118470, WO2002032872A1, WO2002032872A8.Publication date:Aug 7, 2007. Original Assignee:Eisai Co., Ltd Sakaguchi, Takahisa; Tsuruoka, Akihiko. Preparation of amorphous salts of 4-​[3-​chloro-​4-​[(cyclopropylaminocarbonyl)​amino]​phenoxy]​-​7-​methoxy-​6-​quinolinecarboxamide as antitumor agents.  PCT Int. Appl. (2006), WO2006137474 A1 20061228. Naito, Toshihiko and Yoshizawa, Kazuhiro. Preparation of urea moiety-containing quinolinecarboxamide derivatives. PCT Int. Appl., WO2005044788, 19 May 2005 Itaru Arimoto et al. Crystal of salt of 4-​[3-​chloro-​4-​(cyclopropylaminocarbonyl)​amino-​phenoxy]​-​7-​methoxy-​6-​quinolinecarboxamide or solvate thereof and processes for producing these. PCT Int. Appl. (2005), WO2005063713 A1 20050714.
10-23-2009
ANTITUMOR AGENT FOR UNDIFFERENTIATED GASTRIC CANCER
10-2-2009
ANTI-TUMOR AGENT FOR MULTIPLE MYELOMA
8-21-2009
ANTITUMOR AGENT FOR THYROID CANCER
8-14-2009
THERAPEUTIC AGENT FOR LIVER FIBROSIS
2-27-2009
USE OF COMBINATION OF ANTI-ANGIOGENIC SUBSTANCE AND c-kit KINASE INHIBITOR
9-5-2008
Medicinal Composition
8-8-2007
Nitrogen-containing aromatic derivatives
5-25-2007
Polymorph of 4-[3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6- quinolinecarboxamide and a process for the preparation of the same
7-21-2006
Nitrogen-containing aromatic derivatives
6-23-2006
Use of sulfonamide-including compounds in combination with angiogenesis inhibitors
11-16-2011
UREA DERIVATIVE AND PROCESS FOR PREPARING THE SAME
8-10-2011
c-Kit kinase inhibitor
7-6-2011
Nitrogen-Containing Aromatic Derivatives
12-24-2010
COMBINED USE OF ANGIOGENESIS INHIBITOR AND TAXANE
9-24-2010
COMBINATION OF ANTI-ANGIOGENIC SUBSTANCE AND ANTI-TUMOR PLATINUM COMPLEX
4-30-2010
METHOD FOR PREDICTION OF THE EFFICACY OF VASCULARIZATION INHIBITOR
4-16-2010
METHOD FOR ASSAY ON THE EFFECT OF VASCULARIZATION INHIBITOR
3-24-2010
Urea derivative and process for preparing the same
2-26-2010
COMPOSITION FOR TREATMENT OF PANCREATIC CANCER
2-26-2010
COMPOSITION FOR TREATMENT OF UNDIFFERENTIATED GASTRIC CANCER
US7253286 * 18 Apr 2003 7 Aug 2007 Eisai Co., Ltd Nitrogen-containing aromatic derivatives
US20040053908 18 Apr 2003 18 Mar 2004 Yasuhiro Funahashi Nitrogen-containing aromatic derivatives
US20040242506 9 Aug 2002 2 Dec 2004 Barges Causeret Nathalie Claude Marianne Formed from paroxetine hydrochloride and ammonium glycyrrhyzinate by precipitation, spray, vacuum or freeze drying, or evaporation to glass; solid or oil; masks the bitter taste of paroxetine and has a distinctive licorice flavor; antidepressants; Parkinson's disease
US20040253205 10 Mar 2004 16 Dec 2004 Yuji Yamamoto c-Kit kinase inhibitor
US20070004773 * 22 Jun 2006 4 Jan 2007 Eisai R&D Management Co., Ltd. Amorphous salt of 4-(3-chiloro-4-(cycloproplylaminocarbonyl)aminophenoxy)-7-method-6-quinolinecarboxamide and process for preparing the same
US20070078159 22 Dec 2004 5 Apr 2007 Tomohiro Matsushima Has excellent characteristics in terms of physical properties (particularly, dissolution rate) and pharmacokinetics (particularly, bioavailability), and is extremely useful as an angiogenesis inhibitor or c-Kit kinase inhibitor
US20070117842 * 22 Apr 2004 24 May 2007 Itaru Arimoto Polymorph of 4-[3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6- quinolinecarboxamide and a process for the preparation of the same
EP0297580A1 30 Jun 1988 4 Jan 1989 E.R. SQUIBB &amp; SONS, INC. Amorphous form of aztreonam
JP2001131071A Title not available
JP2005501074A Title not available
JPS6422874U Title not available
WO2002032872A1 19 Oct 2001 25 Apr 2002 Itaru Arimoto Nitrogenous aromatic ring compounds
WO2003013529A1 9 Aug 2002 20 Feb 2003 Barges Causeret Nathalie Claud Paroxetine glycyrrhizinate
WO2004039782A1 29 Oct 2003 13 May 2004 Hirai Naoko QUINOLINE DERIVATIVES AND QUINAZOLINE DERIVATIVES INHIBITING AUTOPHOSPHORYLATION OF Flt3 AND MEDICINAL COMPOSITIONS CONTAINING THE SAME
WO2004080462A1 10 Mar 2004 23 Sep 2004 Eisai Co Ltd c-Kit KINASE INHIBITOR
WO2004101526A1 22 Apr 2004 25 Nov 2004 Itaru Arimoto Polymorphous crystal of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-qunolinecarboxamide and method for preparation thereof
WO2005044788A1 8 Nov 2004 19 May 2005 Eisai Co Ltd Urea derivative and process for producing the same
WO2005063713A1 22 Dec 2004 14 Jul 2005 Itaru Arimoto Crystal of salt of 4-(3-chloro-4-(cyclopropylaminocarbonyl)amino-phenoxy)-7-methoxy-6-quinolinecarboxamide or of solvate thereof and processes for producing these
WO2006030826A1 14 Sep 2005 23 Mar 2006 Eisai Co Ltd Medicinal composition
     

1H NMR PREDICT OF LENVATINIB BASE



 सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये। औकात बस इतनी देना, कि औरों का भला हो जाये।
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE


Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK
Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus
Join me on Researchgate

Anthony Melvin Crasto Dr.

  amcrasto@gmail.com

09b37-misc2b027LIONEL MY SON
He was only in first standard in school when I was hit by a deadly one in a million spine stroke called acute transverse mylitis, it made me 90% paralysed and bound to a wheel chair, Now I keep him as my source of inspiration and helping millions, thanks to millions of my readers who keep me going and help me to keep my son happy


सुकून उतना ही देना प्रभू, जितने से
जिंदगी चल जाये।
औकात बस इतनी देना,
कि औरों का भला हो जाये।
Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL