Home » Posts tagged 'NEW DRUGS'
Tag Archives: NEW DRUGS
Myself Anthony Crasto up for grabs as Advisor API & INT, Chem.
Admin note for myself . I am up for Grabs
I myself Dr Anthony Melvin Crasto Looking for a post retirement assignment as Advisor API & INT, Chem.
With 36 yrs rich experience, about dozen patents, 10000plus steps covered, 200 API targets, 30 plus products commercialization in plant in full career. Hands on knowledge of Synthesis, Process, scaleup, cost reduction, DOE , softwares etc
Kindly contact me
Dr Anthony Melvin Crasto
+919321316780
amcrasto@gmail.com
About myself
Dr Anthony Crasto
click on my website to know about me
Read http://amcrasto.weebly.com/
Also http://amcrasto.weebly.com/awards.html
Also
http://amcrasto.weebly.com/felicitations.html
1000 lakh google hits, 100lakh blog views, 10 lakh viewers in USA alone, all in 7 continents, 226 countries, 30 Indian and International awards, helping millions across the world

DIFLUPREDNATE

(1R,3aS,3bS,5S,9aS,9bR,10S,11aS)-1-[2-(acetyloxy)acetyl]-5,9b-difluoro-10-hydroxy-9a,11a-dimethyl-7-oxo-1H,2H,3H,3aH,3bH,4H,5H,7H,9aH,9bH,10H,11H,11aH-cyclopenta[a]phenanthren-1-yl butanoate
(6a,11b)-21-(Acetyloxy)-6,9-difluoro-11-hydroxy-17-(1-oxobutoxy)pregna-1,4-diene-3,20-dione
(6α,11β)-21-(acetyloxy)-6,9-difluoro-11-hydroxy-3,20-dioxopregna-1,4-dien-17-yl butanoate
(6α,11β)-21-Acetoxy-6,9-difluor-11-hydroxy-3,20-dioxopregna-1,4-dien-17-ylbutyrat[German][ACD/IUPAC Name]
(6α,11β)-21-Acetoxy-6,9-difluoro-11-hydroxy-3,20-dioxopregna-1,4-dien-17-yl butyrate[ACD/IUPAC Name]
23674-86-4[RN]
245-815-4[EINECS]
2652
6a,9a-Difluoroprednisolone-21-acetate-17-butyrate
DIFLUPREDNATE
CAS# 23674-86-4
- Molecular FormulaC27H34F2O7
- Average mass508.552 Da
- W 6309
- W-6309
- DFBA
- Difluoroprednisolone butyrate acetate
S8A06QG2QE
TU3831500
дифлупреднат[Russian][INN]
ديفلوبريدنات[Arabic][INN]
二氟泼尼酯[Chinese][INN]
(1R,3aS,3bS,5S,9aS,9bR,10S,11aS)-1-[2-(acetyloxy)acetyl]-5,9b-difluoro-10-hydroxy-9a,11a-dimethyl-7-oxo-1H,2H,3H,3aH,3bH,4H,5H,7H,9aH,9bH,10H,11H,11aH-cyclopenta[a]phenanthren-1-yl butanoate
(6a,11b)-21-(Acetyloxy)-6,9-difluoro-11-hydroxy-17-(1-oxobutoxy)pregna-1,4-diene-3,20-dione
(6α,11β)-21-(acetyloxy)-6,9-difluoro-11-hydroxy-3,20-dioxopregna-1,4-dien-17-yl butanoate
(6α,11β)-21-Acetoxy-6,9-difluoro-11-hydroxy-3,20-dioxopregna-1,4-dien-17-yl butyrate
23674-86-4[RN], 245-815-4[EINECS], 2652, 6a,9a-Difluoroprednisolone-21-acetate-17-butyrate
Difluprednate is a topical corticosteroid used for the symptomatic treatment of inflammation and pain associated with ocular surgery.
Difluprednate is a corticosteroid, It is chemically a butyrate ester of 6(alpha),9(alpha)-difluoro prednisolone acetate. Accordingly, difluprednate is sometimes abbreviated DFBA, for difluoroprednisolone butyrate acetate.
Difluprednate is a topical corticosteroid indicated for the treatment of infammation and pain associated with ocular surgery. It is a butyrate ester of 6(α), 9(α)-difluoro prednisolone acetate. Difluprednate is abbreviated DFBA, or difluoroprednisolone butyrate acetate. It is indicated for treatment of endogenous anterior uveiti.
Approval
On June 24, 2008, the US Food and Drug Administration (FDA) approved difluprednate for the treatment of post-operative ocular inflammation and pain.[1] It is marketed by Alcon under the tradename Durezol.
Depositor-Supplied Patent Identifiers
Publication Number | Title | Priority Date | Grant Date |
---|---|---|---|
US-2020325543-A1 | Diagnostic method | 2017-11-20 | |
WO-2012088044-A2 | Compositions and methods for improving ocular surface health, corneal clarity, optical function and maintaining visual acuity | 2010-12-20 | |
US-7790905-B2 | Pharmaceutical propylene glycol solvate compositions | 2002-02-15 | 2010-09-07 |
US-7927613-B2 | Pharmaceutical co-crystal compositions | 2002-02-15 | 2011-04-19 |
PATENT
WO/2022/118271DIFLUPREDNATE FOR REDUCING THE ADVERSE EFFECTS OF OCULAR INFLAMMATION
SYN 1
Synthetic Reference
Process for preparation of Difluprednate from sterol fermentation product; Ding, Kai; Xu, Feifei; Assignee Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Peop. Rep. China; East China University of Science and Technology; 2014; Patent Information; Aug 06, 2014; CN; 103965277; A

SYN 2
Synthetic Reference
Preparation method of Difluprednate; Tian, Yuan; Zhou, Shengan; Guo, Bin; Xu, Zhiguo; Assignee Guangzhou Renheng Pharmaceutical Technology Co., Ltd., Peop. Rep. China 2017; Patent Information; May 10, 2017; CN; 106632561; A

SYN3
Synthetic Reference
Shailesh, Singh; Bharat, Suthar; Jain, Ashish; Gaikwad, Vinod; Kulkarni, Kuldip. Process for preparing difluprednate. Assignee Ajanta Pharma Ltd., India. IN 2013MU02535. (2015).

SYN4
Synthetic Reference
Sun, Hongbin; Chen, Bo. Method for preparation of Difluprednate. Assignee China Pharmaceutical University, Peop. Rep. China. CN 103509075. (2014).


PATENT
https://patents.google.com/patent/CN103509075A/en

Embodiment 1:4, pregnant steroid-17 α of 9 (11)-diene, 21-dihydroxyl-3,20-diketone-21-acetic ester (formula III compound)
10g hydrocortisone-21 acetic ester (formula II compound) is joined in 250mL eggplant type bottle, add 50mL N, dinethylformamide and 8.8mL pyridine, slowly heat up and make material dissolution complete, slowly cooling afterwards, slowly be added dropwise to 4.4mL methylsulfonyl chloride, add rear solution to be yellow completely.Be warming up to 85 ℃ of stirrings, the reaction solution thick one-tenth that can slowly become sticky is faint yellow, adds slightly some DMFs and makes reaction solution dilution, can normally stir, and keeps this thermotonus one hour, and reaction solution slowly becomes grey black during this period.TLC follows the tracks of (sherwood oil: ethyl acetate=1: 1) show that reaction finishes.Stop heating, treat that the backward reaction solution of slow cooling adds 200mL methyl alcohol, stir 1min, reaction flask is placed in to crystallization under ice-water bath.Suction filtration after 1h, makes water and methanol wash filter cake, crude product productive rate 100%.With methyl alcohol-methylene dichloride mixed solvent system recrystallization, obtain sterling, M.P.231-235 ℃, productive rate 90%. 1H-NMR(300MHz,CDCl 3):δ(ppm)5.75(1H,s,4-H),5.55(1H,s,11-H),5.07(1H,d,J=5Hz,21-H),4.84(1H,d,J=5Hz,21-H),2.15(3H,s,H-21-OAc),1.31(3H,s,19-CH 3),0.65(3H,s,18-CH 3),0.66-2.90(m,17H,backbone).
Embodiment 2:4,9 (11)-diene-17 α, 21-dihydroxyl-3,20-diketone-21-acetic ester 17 iophenoxic acid esters (formula IV compound)
By 9.4g4, pregnant steroid-17 α of 9 (11)-diene, 21-dihydroxyl-3,20-diketone-21-acetic ester (formula III compound) and 10g4-Dimethylamino pyridine add in 1000mL eggplant-shape bottle, add again 50mL diethylene glycol dimethyl ether and 260mL methylene dichloride, heated and stirred makes dissolution of solid, slowly adds 32mL butyryl oxide slightly after cooling, is warming up to 80 ℃ of return stirrings.After 23h, TLC follows the tracks of, and raw material primitive reaction is complete, stops heating and stirs.Vacuum concentration is removed methylene dichloride.After being down to room temperature, add frozen water in reaction flask, white solid standing to be separated out.Suction filtration, saturated sodium bicarbonate aqueous solution washing leaching cake, dries under infrared lamp, obtain 4,9 (11)-diene-17 α, 21-dihydroxyl-3,20-ketone-21-acetic ester 17 iophenoxic acid esters (formula IV compound) sterling 10.65g, M.P220-224 ℃, productive rate 95.9%. 1H-NMR(500MHz,CDCl 3):δ(ppm)5.75(1H,s,4-H),5.54(1H,m,11-H),4.87(1H,d,J=4.8Hz,O=C-CH 2-O,21-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.75(2H,m,2-H),0.70(3H,s,18-CH 3),0.95(3H,t,J=4.4Hz),1.34(3H,s,18-CH 3),1.66(2H,m,-CH 2CH 3),2.17(3H,s,O=C-CH 3),2.32(2H,t,J=4.3Hz,O=C-CH 2),? 13C-NMR(75MHz,CDCl 3):δ(ppm)199.1,198.9,173.4,170.4,169.1,144.1,124.1,118.5,94.5,66.9,48.2,46.3,40.9,37.5,36.4,34.2,33.8,32.7,32.2,32.1,30.6,26.2,24.5,20.5,18.3,13.7,13.6;ESI-MS?m/z:457.2[M+H +],479.2[M+Na +];HRMS?for?C 27H 36O 6+Na +?calcd?479.2410,found479.2402.
Embodiment 3:3,5,9 (11) pregnant steroid-3 of triolefin, 17 α, 21 trihydroxy–3,20-diketone-3,21-diacetate esters 17 iophenoxic acid esters (formula V)
10g4, pregnant steroid-17 α of 9 (11)-diene, 21-dihydroxyl-3,20-diketone-21-acetic ester 17 iophenoxic acid esters add in 250mL eggplant type bottle, then add 80mL methylvinyl acetate, slowly drip while stirring the 1mL vitriol oil.Be warming up to 80 ℃ of stirring reactions, solution is thin out yellow clarification slowly.(sherwood oil: ethyl acetate=3: 1), raw material reaction is complete produces new point to TLC after 30min.Stop heating, wait to be cooled to 50 ℃, add 1mL triethylamine, be stirred to and be down to room temperature.Add water in reaction solution, ethyl acetate aqueous layer extracted three times, saturated common salt water washing organic phase twice, anhydrous sodium sulfate drying.After 30min, steam organic solvent and obtain brown color oily matter.Column chromatography is purified and is obtained 3,5,9 (11) pregnant steroid-3 of triolefin, 17 α, 21 trihydroxy–3,20-diketone-3,21-diacetate esters 17 iophenoxic acid esters, productive rate 90%. 1H-NMR(300MHz,CDCl 3):δ(ppm)5.74(1H,s,4-H),5.53(1H,s,11-H),5.45(1H,s,6-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.17(3H,s,-COCH 3),1.17(3H,s,19-CH 3),0.96(3H,t,J=7.5Hz),0.70(3H,s,18-CH 3).
Embodiment 4:4, fluoro-17 α of 9 (11)-diene-6-, 21-dihydroxyl-3,20-diketone-21-acetic ester 17 iophenoxic acid esters
10g3,5,9 (11) pregnant steroid-3 of triolefin, 17 α, 21 trihydroxy–3,20-diketone-3,21-diacetate esters 17 iophenoxic acid esters are dissolved in 60mL acetonitrile, and under nitrogen protection ,-4 ℃ are stirred half an hour.Slowly drip the acetonitrile suspension 40mL of Selecfluor in reaction flask, under nitrogen protection, react 2 hours, TLC (sherwood oil: ethyl acetate=3: 1) monitoring reaction, raw material reaction is complete.Stopped reaction, adds water in reaction flask, ethyl acetate extraction three times, saturated common salt water washing twice, anhydrous sodium sulfate drying.Vacuum concentration is removed organic solvent, obtain faint yellow solid 4,9 (11)-diene-6 α-fluoro-17 α, 21-dihydroxyl-3,20-diketone-21-acetic ester 17 iophenoxic acid esters (formula VII) and 9 (11)-diene-6 β-fluoro-17 α, 21-dihydroxyl-3, the mixture of 20-diketone-21-acetic ester 17 iophenoxic acid esters (formula VI), productive rate 85%. 1H-NMR(500MHz,CDC1 3):δ(ppm)5.90(1H,d,J=4.5Hz,4-H),5.59(1H,s,11-H),5.07(1H,m,6-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.17(3H,s,-COCH 3),1.46(3H,s,18-CH 3),0.96(3H,t,J=7.5Hz),0.73(3H,s,19-CH 3).
Embodiment 5:4,9 (11)-diene-6 α-fluoro-17 α, 21-dihydroxyl-3,20-diketone-21-acetic ester 17 iophenoxic acid esters (formula VII)
14g4, 9 (11)-diene-6 α-fluoro-17 α, 21-dihydroxyl-3, 20-diketone-21-acetic ester 17 iophenoxic acid esters (formula VII) and 9 (11)-diene-6 β-fluoro-17 α, 21-dihydroxyl-3, the mixture of 20-diketone-21-acetic ester 17 iophenoxic acid esters (formula VI) adds in dry three-necked bottle, add while stirring 400mL acetum, under room temperature, slowly pass into anhydrous hydrogen chloride gas (98% vitriol oil is added dropwise in 37% concentrated hydrochloric acid solution and makes) until saturated, be stirred to raw material and be dissolved into yellow solution completely, continue to stir 2h, TLC monitoring reacts completely, stop stirring, in reaction solution, add the aqueous solution, after separating out solid, suction filtration, saturated sodium bicarbonate aqueous solution washing, dry, be weighed as 13g, productive rate is 93%. 1H?NMR(300MHz,CDCl 3):δ(ppm)6.10(s,1H),5.61(s,1H),5.41-5.16(m,1H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.82(dd,J=28.3,15.7Hz,3H),2.50(s,2H),2.32(t,J=7.4Hz,2H),2.17(s,3H),1.96(s,5H),1.66(d,J=7.4Hz,2H),1.46(s,2H),1.33(s,3H),0.96(s,3H),0.71(s,3H).
Embodiment 6:6 α-fluoro-9 α-bromo-11 beta-hydroxies-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester (formula VIII)
13g 6 α-fluoro-4; 9; (11)-diene-pregnant steroid-3,20-22 ketone-17-butyric ester-20-acetic ester is dissolved in and fills 300mL1, in the eggplant type bottle of 4 dioxane; add while stirring 40mL 0.46mol/L high chloro acid solution; under room temperature, stir after several minutes, add 14g N-succinimide in reaction system, under nitrogen protection, stir; raw material dissolves gradually, and it is faint yellow that reaction solution is.(the sherwood oil: ethyl acetate=12: 5) monitoring, raw material primitive reaction is complete, adds 10%Na of TLC after 2h 2sO 3unnecessary N-succinimide is fallen in aqueous solution cancellation, and checks (it is blue that test paper no longer becomes) with starch-kalium iodide test paper.Add water in reaction flask, ethyl acetate extraction three times, twice of saturated common salt water washing organic phase, anhydrous sodium sulfate drying organic phase, after 30min, be spin-dried for organic phase, obtain faint yellow oily matter, column chromatography purification (sherwood oil: ethyl acetate=12: 1) obtain white solid 6 α-fluoro-9 α-bromo-11 beta-hydroxies-4-alkene-pregnant steroid-3, the about 14g of 20-diketone-17-butyric ester-20-acetic ester, productive rate is 89%. 1H-NMR(300MHz,CDCl 3):δ(ppm)5.93(1H,d,J=4.5,4-H),5.06(1H,m,6-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.17(3H,s,-COCH 3),1.84(3H,s,18-CH 3),0.96(3H,t,J=7.5Hz),1.02(3H,s,19-CH 3),4.72(1H,s,11-H);ESI-MS?m/z:593.3,595.3[M+Na +].
Embodiment 7:6 α-fluoro-9 β, 11 beta epoxides-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester (formula IX)
14g 6 α-fluoro-9 α-bromo-11 beta-hydroxies-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester drops in 500mL eggplant type bottle, adds 200mL acetone, stirs raw material is fully dissolved, and adds afterwards 3g Potassium ethanoate, is warming up to 60 ℃ of return stirring 13h.TLC (sherwood oil: ethyl acetate=2: 1) monitoring finds that new product occurs.Stop heating, in reaction solution, add water, ethyl acetate extraction, anhydrous sodium sulfate drying organic phase, after standing 30min, steams except organic solvent, obtains yellow oil, productive rate 96%.Column chromatography is purified, and obtains white solid powder, and nuclear-magnetism confirmation structure is 6 α-fluoro-9 β, 11 beta epoxides-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester. 1H-NMR(300MHz,CDC1 3):δ(ppm)6.11(1H,d,J=4.5Hz,4-H),5.31(1H,m,6-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.17(3H,s,-COCH 3),0.94(3H,s,18-CH 3),0.97(3H,t,J=7.5Hz),1.55(3H,s,19-CH 3),3.52(1H,s,11-H);ESI-MS?m/z:491.2[M+H +],513.2[M+Na +].
Embodiment 8:6 α, 9 α-fluoro-11 beta-hydroxies-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester (formula X)
100mg 6 α-fluoro-9 β, 11 beta epoxides-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester drops in the Plastic Bottle of tetrafluoroethylene, adds 2mL methylene dichloride to dissolve, and stirs at-20 ℃.1mL Olah reagent with under 1mL methylene dichloride low temperature, mix after, be slowly added dropwise in reaction system, maintain low temperature and stir 2 hours, TLC monitoring reaction finishes.Reaction flask shifts out low-temp reaction groove, is slowly added dropwise to the 1mol/L NaOH aqueous solution by excessive HF cancellation, is adjusted to pH7~8.Add chloroform in reaction system, extraction, organic layer is used respectively aqueous hydrochloric acid and the saturated common salt water washing of 3mol/L, anhydrous sodium sulfate drying, after standing 30min, steams except organic solvent, column chromatography is further purified and is obtained white solid powder 6 α, 9 α-fluoro-11 beta-hydroxies-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester, productive rate 90%. 1H-NMR(300MHz,CDCl 3):δ(ppm)?6.11(1H,d,J=4.5Hz,4-H),5.27(1H,m,6-H),4.64-4.91(2H,ABq,J=16.6Hz,21-H),2.17(3H,s,-COCH 3),4.40(1H,d,J=4.5Hz,11-H),1.02(3H,s,18-CH 3),0.96(3H,t,J=7.5Hz),1.52(3H,s,19-CH 3);ESI-MS?m/z:533.3[M+Na +]
Embodiment 9:6 α, 9 α-fluoro-11 beta-hydroxies-Isosorbide-5-Nitrae-diene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester (difluprednate) (formula I)
40mg 6 α, 9 α-fluoro-11 beta-hydroxies-4-alkene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester is dissolved in 3mL dioxane, adds 28mgDDQ, and 100 ℃ of return stirrings heat up.TLC monitoring reaction (sherwood oil: ethyl acetate=12: 8) after 13h, generate the larger product of polarity, steam except organic solvent dioxane, obtain brown color oily matter, add a small amount of methylene dichloride lysate, suction filtration, elimination solid residue, filtrate is washed with sodium bicarbonate aqueous solution after adding a small amount of methylene dichloride again, steams except organic phase rear pillar Chromatographic purification, obtain white solid powder 6 α, 9 α-fluoro-11 beta-hydroxies-Isosorbide-5-Nitrae-diene-pregnant steroid-3,20-diketone-17-butyric ester-20-acetic ester, be title molecule difluprednate, productive rate 70%. 1h-NMR (300MHz, CDCl 3): δ (ppm) 7.20 (1H, d, J=4.5Hz, 1-H), 6.43 (1H, s, 4-H), 6.38 (1H, d, J=6Hz, 2-H), 5.36 (1H, m, 6-H), 4.64-4.91 (2H, ABq, J=16.6Hz, 21-H), 4.43 (1H, d, J=4.5Hz, 11-H), 2.27 (2H, m ,-CH 2-CH 3), 2.17 (3H, s, O=C-CH 3), 1.55 (3H, s, 19-CH 3), 1.02 (3H, s, 18-CH 3), 0.93 (3H, t, J=4.5Hz, 0=C-CH 2cH 2cH 3); ESI-MS m/z:509.3[M+H +]; HRMS for C 27h 35o 7f 2+ H +calcd 509.2351, found 509.2356.M.P.188-190 ℃ (literature value M.P.190-194 ℃); [α] d22=+30.1 ° of (literature values [α] d22=+31.7 °).
Claims (6)
Hide Dependent
1. a method of preparing difluprednate, as following reaction formula:
Specifically comprise the following steps:
(1) by hydrocortisone-21-acetic ester (formula II compound):
Carry out dehydration reaction, generate formula III compound:
(2) formula III compound is carried out to butyric acid esterification, obtains formula IV compound:
(3) formula IV compound is carried out to the reaction of enolization esterifying reagent, obtains formula V compound:
(4) formula V compound is reacted with fluoro reagent and obtains formula VI and formula VII compound:
(5) by formula VI compound, through configuration reversal, reaction obtains formula VII compound;
(6) formula VII compound is reacted with N-bromo-succinimide and water, obtains formula VIII compound:
(7) formula VIII compound epoxidation under alkaline condition is obtained to formula IX compound:
(8) formula IX compound is reacted with fluorination reagent and obtains formula X compound:
(9) dehydrogenation of formula X compound oxidation is obtained to formula I compound (difluprednate).
2. method as claimed in claim 1, is characterized in that, in step (2), formula III compound is obtained to formula IV compound through fourth esterification, and the fourth esterifying reagent adopting is butyryl oxide or butyryl chloride; The alkaline catalysts adopting is pyridine, triethylamine or DMAP; The solvent adopting is methylene dichloride, diethylene glycol dimethyl ether, 1, the mixture of the optional solvents in 2-ethylene dichloride, dioxane, trichloromethane, DMF, methyl-sulphoxide, N,N-dimethylacetamide or above-mentioned solvent.
3. method as claimed in claim 1, is characterized in that, in step (3), formula IV compound is obtained to formula V compound through enolization esterification, and the enolization esterifying reagent adopting is diacetyl oxide, Acetyl Chloride 98Min., methylvinyl acetate or vinyl-acetic ester; The catalyzer adopting is the vitriol oil or tosic acid; The solvent adopting is the mixture of the optional solvents in methylene dichloride, chloroform, toluene, methylvinyl acetate, vinyl-acetic ester or above-mentioned solvent.
4. method as claimed in claim 1, is characterized in that, in step (4), formula V compound is obtained to formula VI compound and formula VII compound through fluoridizing, and the fluoro reagent adopting is Selectfluor or Accufluor; The solvent adopting is the mixture of the optional solvents in methylene dichloride, chloroform, toluene, acetonitrile or above-mentioned solvent.
5. method as claimed in claim 1, it is characterized in that, in step (8), formula IX compound is obtained to formula X compound through fluoridizing open loop, the fluorination reagent adopting is aqueous hydrogen fluoride solution, hydrogen fluoride pyridine solution (Olah reagent) or hydrogen fluoride triethylamine solution; The solvent adopting is methylene dichloride, chloroform, 1, the mixture of the optional solvents in 2-ethylene dichloride, tetrahydrofuran (THF), toluene or above-mentioned solvent; Range of reaction temperature is-50~50 ℃.
6. a key intermediate compound for synthetic difluprednate, shown in IV compound:

Patent
Publication numberPriority datePublication dateAssigneeTitle
US3780177A *1967-06-161973-12-18Warner Lambert Co17-butyrate,21-ester derivatives of 6alpha,9alpha-difluoroprednisolone,compositions and use
US4525303A *1982-06-211985-06-25Dainippon Ink And Chemicals Inc.Process for preparation of steroids
CN101397321A *2007-09-292009-04-01天津药业研究院有限公司Preparation of hydrocortisone and derivatives thereof
CN102076344A *2008-05-282011-05-25瓦利杜斯生物医药有限公司Non-hormonal steroid modulators of nf-kb for treatment of disease
CN102134266A *2010-12-302011-07-27北京市科益丰生物技术发展有限公司Preparation method of melengestrol acetate
Publication numberPriority datePublication dateAssigneeTitle
CN102964412A *2012-11-272013-03-13山东省医药工业研究所Novel crystal form and preparation method of difluprednate
CN103965277A *2014-05-192014-08-06中国科学院上海有机化学研究所Method for synthesizing difluprednate from sterol fermentation product
CN106632561A *2016-12-162017-05-10广州仁恒医药科技股份有限公司Method for preparing difluprednate
CN106749464A *2016-12-292017-05-31奥锐特药业有限公司Steroidal epoxide carries out open loop, the method for fluorination reaction and its device
CN107915766A *2016-10-112018-04-17江苏福锌雨医药科技有限公司A kind of preparation method of fludrocortison acetate
CN108503679A *2018-04-032018-09-07广州仁恒医药科技股份有限公司A kind of purification process of Difluprednate intermediate
//////////

AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

join me on Facebook
Anthony Melvin Crasto Dr. | Facebook
join me on twitter
Anthony Melvin Crasto Dr. | twitter
+919321316780 call whatsaapp
EMAIL. amcrasto@amcrasto
/////////////////////////////////////////////////////////////////////////////
Clinical trials
Difluprednate ophthalmic emulsion 0.05% is also being studied in other ocular inflammatory diseases, including a phase 3 study evaluating difluprednate for the treatment of anterior uveitis[2][3]

NEW DRUG APPROVALS
TO MAINTAIN THIS BLOG SUBSCRIPTIONS
$10.00
References
- ^ “Sirion Therapeutics Announces FDA Approval of Durezol for Treatment of Postoperative Ocular Inflammation and Pain” (Press release). Sirion Therapeutics, Inc. 2008-06-24. Retrieved 2008-06-30.
- ^ Clinical trial number NCT00501579 for “Study of Difluprednate in the Treatment of Uveitis” at ClinicalTrials.gov
- ^ Sheppard JD, Toyos MM, Kempen JH, Kaur P, Foster CS (May 2014). “Difluprednate 0.05% versus prednisolone acetate 1% for endogenous anterior uveitis: a phase III, multicenter, randomized study”. Investigative Ophthalmology & Visual Science. 55 (5): 2993–3002. doi:10.1167/iovs.13-12660. PMC 4581692. PMID 24677110.
Clinical data | |
---|---|
AHFS/Drugs.com | Monograph |
MedlinePlus | a609025 |
License data | US FDA: Difluprednate |
Routes of administration | eye drops |
ATC code | D07AC19 (WHO) |
Legal status | |
Legal status | US: ℞-only |
Identifiers | |
showIUPAC name | |
CAS Number | 23674-86-4 |
PubChem CID | 32037 |
DrugBank | DB06781 |
ChemSpider | 391990 |
UNII | S8A06QG2QE |
KEGG | D01266 |
ChEBI | CHEBI:31485 |
ChEMBL | ChEMBL1201749 |
CompTox Dashboard (EPA) | DTXSID0046773 |
ECHA InfoCard | 100.041.636 |
Chemical and physical data | |
Formula | C27H34F2O7 |
Molar mass | 508.559 g·mol−1 |
3D model (JSmol) | Interactive image |
showSMILES | |
showInChI | |
(what is this?) (verify) |
///////////////DIFLUPREDNATE, W 6309, W-6309, DFBA, Difluoroprednisolone butyrate acetate, S8A06QG2QE, TU3831500, дифлупреднат , ديفلوبريدنات , 二氟泼尼酯 , OCCULAR, PAIN
CCCC(=O)OC1(CCC2C1(CC(C3(C2CC(C4=CC(=O)C=CC43C)F)F)O)C)C(=O)COC(=O)C
Sept 2021, This blog New Drug Approvals approaching 34 lakh views

NEW DRUG APPROVALS FAST APPROACING 34 LAKH VIEWS in 225 countries 7 continents. 5 lakh viewers in USA alone
#worlddrugtracker#worldpeaceambassador#india#helpingmillions#amcrasto#medchem#blog#education#service#free#divyang#everydifficultyanopportunity#opensuperstar#pharmadon#qbdking#ict#ictian#pharma


NEW DRUG APPROVALS
ONE TIME TO MAINTAIN THIS BLOG
$10.00
NERATINIB, HKI 272, ..Puma presents positive results from phase II trial of its investigational drug PB272

PB-272
PF-0528767
WAY-179272
WAY-179272-B (maleate)
Neratinib – малая молекула класса 6,7-дизамещенных-4-anilinoquinoline-3-карбонитрила – ингибитор тирозинкиназы HER-2 с потенциальной противоопухолевой активностью. Neratinib связывается с рецептором HER-2 необратимо, снижая аутофосфорилирование в клетках, и направляя остаток цистеина в АТФ-связывающего кармана рецептора. Обработка раковых клеток с этим агентом приводит к торможению передачи сигнала клеточного цикла и в конечном счете уменьшает клеточную пролиферацию. Neratinib ингибирует рецептор EGFR киназы и распространение EGFR-зависимых клеток. |
|
Neratinib – small molecule 6,7-disubstituted class of 4-anilinoquinoline-3-carbonitrile – inhibitor of the HER-2 tyrosine kinase with potential antitumor activity. Neratinib binds to the receptor HER-2 irreversible, reducing autophosphorylation in cells and directing the cysteine residue in the ATP-binding pocket of the receptor. Treatment of cancer cells with this agent leads to inhibition of signal transduction and cell cycle ultimately reducescell proliferation. Neratinib inhibit EGFR kinase receptor and distribution of EGFR-dependent cells. |
EVER THE POST WAS WRITTEN IT GOT FDA APPROVAL
NERATINIB MALEATE
PUMA BIOTECH
Nerlynx | FDA | 7/17/2017 | To reduce the risk of breast cancer returning Press Release Drug Trials Snapshot |
Neratinib (HKI-272) is a tyrosine kinase inhibitor[1][2] under investigation for the treatment breast cancer[3] and other solid tumours.
It is in development for the treatment of early- and late-stage HER2-positive breast cancer.[4]
Like lapatinib and afatinib, it is a dual inhibitor of the human epidermal growth factor receptor 2 (Her2) and epidermal growth factor receptor (EGFR) kinases.[5]
Neratinib is a signal transduction pathway inhibitor and an irreversible inhibitor of HER-2 in early clinical trials for the treatment of advanced solid tumors in combination with paclitaxel. The company had also been developing the drug candidate for the treatment of non-small cell lung cancer (NSCLC); however, no recent development has been reported for the indication. In 2011, Pfizer discontinued development of the compound as monotherapy for the treatment of ErbB-2-positive breast cancer. A phase III clinical trial had been under way. Dana-Farber Cancer Institute is studying the compound for the treatment of patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer and brain metastases. Puma Biotechnology is conducting phase III trials for use as third-line treatment of HER2-positive metastatic breast cancer and phase II trials for the treatment of patients with HER2 activating mutations in Non-Small Cell Lung Cancer (as monotherapy or in combination with temsirolimus) as well as other solid tumors.
The drug candidate is a synthetic compound developed based on the chemical structure of EKB-569, an inhibitor of the epidermal growth factor receptor (EGFR) currently under clinical evaluation for the treatment of EGFR-positive tumors. In previous trials, neratinib inhibited kinase activity of HER-2 and EGFR by 50% while showing no effects on several serine-threonine kinases, and also inhibited the proliferation of two HER-2-positive breast cancer cell lines and a mouse fibroblast cell line transfected with the HER-2 oncogene.
In 2011, the compound was licensed to Puma by Pfizer for global development and commercialization.
HKI-272 (neratinib) has been described for the treatment of neoplasms [US Patent 6,288,082]. Neratinib is a potent irreversible pan erbB inhibitor. Neratinib is an orally available small molecule that inhibits erbB-1 , erbB-2 and erbB-4 at the intracellular tyrosine kinase domains, a mechanism of action that is different from trastuzumab. Neratinib reduces erbB-1 and erbB-2 autophosphorylation, downstream signaling, and the growth of erbB-1 and erbB-2 dependent cell lines.
Preclinical data suggest that neratinib will have antitumor activity in erbB-1 – and/or erbB 2-expressing carcinoma cell lines, with cellular IC50 <100 nM [Rabindran SK, et al. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Research. 2004;64(1 1 ):3958-65].
Neratanib is being developed by Puma Biotechnology. It will be included in the forthcoming I-SPY2breast cancer trial.[6]
neratinib refers to HKI-272, which has the following core structure:
in its free base form. Optionally, a pharmaceutically acceptable salt or hydrate thereof may be used. The core structure represented above is a particular HKI-272 compound, called HKI-272 or neratinib, which has the chemical name [(2E)-N-[4-[[3-chloro-4- [(pyridin-2-yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4- (dimethylamino)but-2-enamide]. Although currently less preferred, another HKI-272 compound may be used in the place of neratinib. “A HKI-272 compound” refers, in one embodiment, to a compound derived from the core structure of neratinib shown above
The preparation of HKI-272 compounds, of which neratinib is a species, are described in detail in US Patent Application Publication No. 2005/0059678, which is hereby incorporated by reference. See, also, US Patent Nos. 6,288,082, US Patent No. 6,002,008, US Patent No. 6,297,258 and US Patent Application Publication No. 2007/0104721 , which are hereby incorporated by reference. The methods described in these documents can also be used to prepare neratinib and/or the other HKI-272 and substituted 3-quinoline compounds used herein and are hereby incorporated by reference. In addition to the methods described in these documents, International Patent Publication Nos. WO-96/33978 and WO-96/33980, which are hereby incorporated by reference, describe methods that are useful for the preparation of these HKI-272 compounds. Although these methods describe the preparation of certain quinazolines, they are also applicable to the preparation of correspondingly substituted 3- cyanoquinolines and are hereby incorporated by reference.
The term “treating” or “treatment” refers to the administration of the neratinib to a subject to prevent or delay, to alleviate, or to arrest or inhibit development of the symptoms or conditions associated with neoplasms
(E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- (dimethylamino)-2-butenamide is an irreversible inhibitor to Her-2 (also known as ErbB-2 or neu) kinase, a member of the epidermal growth factor receptor (EGFR) family. EGFR family members have been implicated in tumorigenesis and associated with poor prognosis in tumor types in humans. The structure of the (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano- 7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide in the form of a free base is shown below:
The compound (E)-N-{4-[3-chloro-4 J-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide in the form of a free base is described in U.S. Patent No. 6,288,082. The compound is classified, based on the Biopharmaceutical Classification System, as a BCS Class IV compound (low water solubility and low permeability). The free base has low solubility in water, with a water solubility of about 1 μg/ml_ at about pH 7. The water solubility increases with decreasing pH as the compound becomes ionized. This compound is water soluble at gastrointestinal pH, and dissolution is not rate limiting.
Research on Chemical Intermediates, 2012, 09(22),6168
10.1007/s11164-012-0822-4
The Wittig–Horner reaction for the synthesis of neratinib
…………………
U.S. Patent No. 6,288,082
http://www.google.co.in/patents/US6288082
…………
WO2010048477A2
http://www.google.com/patents/WO2010048477A2?cl=en
U.S. Pat. No. 7,126,025 discloses certain novel 4-amino-2-butenoyl chlorides, processes for their preparation and their use as intermediates in the synthesis of pharmaceutically active protein kinase inhibitors, including but not limited to for example HKI-272 and EKB-569.
The sequence illustrated below and summarized in Scheme 1 describes one existing process for preparing HKI-272, (E)-Λ/-(4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-3- cyano-7-ethoxyquinolin-6-yl)-4-(dimethylamino)but-2-enamide in the form of the maleate salt, also known as Neratinib™.
1 95 eq (COCI)2, cat DMF
O
^
Step 5 OH 16 h HCI
Scheme 1
Scheme 2
Scheme 3. Formation of acid chloride with SOCI2 in DMAc and coupling with a substituted aniline.
SOCl2
/Nv^-^’C02H HCI DMAc HCI
Scheme 4. Formation of the MW 638 impurity.
Example 4: Process 3
4-Dimethylaminocrotonoyl chloride hydrochloride and its coupling with 6-amino- 4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-7-ethoxyquinoline-3-carbonitrile (procedure with thionyl chloride and DMAc).
A suspension of 4-dimethylaminocrotonic acid (17.0 g, 97.5 mmol) in DMAc (170 ml_) was cooled to -15 0C under nitrogen atmosphere. Neat thionyl chloride (12.8 g, 7.83 mmol) was added to the slurry at a rate to maintain the temperature in the reactor in the range of -18 to -14 0C (moderate exotherm). The reaction mixture was held at -17 to -15 0C for 4 hrs. A solution of the aminoquinoline (36.2 g, 81.3 mmol) in DMAc (440 ml_) was added to the reactor maintaining the temperature in the -14 to -19 0C range. The resulting mixture was held for 18 hr at approximately -15 0C. At this point HPLC analysis showed residual aniline level at 2.5%. The thick suspension of the hydrochloride salt of the coupled product was quenched with water (200 ml_) maintaining the batch temperature between -5 and -16 0C. The pH of the resulting clear solution was adjusted to 1 1 with a 13% aqueous solution of NaOH (approx. 210 ml_ of the solution was added). The suspension was further diluted with water (350 ml_) and the solids were filtered on a polypropylene cloth filter. The cake was washed with water until neutral pH of the washes and dried first in the nitrogen flow on the filter and then on a tray in vacuum at 45 to 50 0C to afford crude (.=)-/\/-(4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-3-cyano-7- ethoxyquinolin-6-yl)-4-(dimethylamino)but-2-enamide (42.0 g, 91 %) as a bright-yellow crystalline solid.
………………..
WO2004066919A2
http://www.google.com/patents/WO2004066919A2?cl=en
Reaction Scheme Example 1 :
SCHEME 1
(“)
6-(4-N,N-dimethylarninocrotonyt)amido- 4-(4-benzyioxy-3-chloro)arniπo-3-cyano- 7-ethoxyquiπoline, WAY-177820 C31H3[1CIN5θ3 MW 556.07
A suspension of 4-N,N-dimethylaminocrotonic acid hydrochloride in acetonitrile and a catalytic amount of DMF is cooled to 0-10° C. Oxalyl chloride (0.95 eq) is added dropwise and the mixture warmed to 25-30° C and stirred until the chlorinating agent is completely consumed. The light yellow solution is checked for complete consumption of oxalyl chloride by HPLC then cooled to 0-10° C. A cooled solution (0-10° C) of 4-[4-benzyloxy-3-chloro]amino-6-amino-3-cyano-7- ethoxyquinoline in NMP is added dropwise and the mixture is stirred until < 2% of the starting aniline remains. The mixture is added to saturated aqueous sodium bicarbonate, the yellow precipitates are filtered and washed with water. The wet solids are heated to reflux in acetonitrile and clarified hot to remove insolubles. The solution is cooled, the precipitated product filtered and washed with cold acetonitrile. The product is dried (40-50° C, 10 mm Hg, 24 hours) to obtain the final product. Reaction Scheme Example 2:
A solution of 4-N,N-dimethylaminocrotonic acid hydrochloride in tetrahydrofuran (THF) and a catalytic amount of dimethyiformamide (DMF) is cooled to 0-5s C. Oxalyl chloride (0.95 eq) is added dropwise and the mixture warmed to 25-302C and stirred until the chlorinating agent is completely consumed. The orange solution is checked for complete consumption of oxalyl chloride by high- pressure liquid chromatography (HPLC) then cooled to 0-52 C. A solution of 4-[4-(2- pyridylmethoxy)-3-chloro]amino-6-amino-3-cyano-7-ethoxyquinoline is added dropwise and the mixture is stirred until < 0.5% of the starting aniline remains. The reaction is quenched with water and the mixture warmed to 40s C. Aqueous sodium hydroxide is added to bring the pH to 10-11. The resulting precipitates are filtered hot and washed with water. The wet solids are heated to reflux (70-759 C) in acetonitrile:THF (1 :5:1) and the solution cooled slowly to room temperature. The product is filtered and washed with acetonitrile.THF. The product is dried (50e C, 10 mm Hg, 24 hours) to 80-85% yield.
Reaction Scheme Example 3:
4-Dirnethy!amino-but-2-enoic acid |4-(3-chloro-4-fluoro-phenylamino)-3-cvano-7- ethoxy-quinolin-6-vHamide
A. 4-(dimethylamino)-2-butenoyl chloride hydrochloride
A 1 L multi-neck flask equipped with agitator, thermometer, addition funnel, and nitrogen protection is charged with acetonitrile (0.67 kg, 0.85 L) followed by adding dimethylformamide (0.00086 kg, 0.91 mL, d=0.944 g/mL). At ambient temperature, is added 4-dimethylaminocrotonic acid hydrochloride (0.0709 kg) and the mixture stirred until homogeneous. Cool the reaction mixture to (0-10° C) and add oxalyl chloride (0.0473 kg, 0.0325 L, d = 1.45 g/mL) dropwise over (20 minutes) at (0-10° C) followed by a rinse with acetonitrile (0.02 kg, 0.03 L). The temperature (0-10°C) is maintained for about (20 minutes). The temperature of the reaction mixture is adjusted to (22-26° C) over (20 minutes) and maintained over (2 hours). The temperature of reaction mixture is adjusted to (40-45° C) and held for about (5 minutes). Cool the light suspension to about (20-25° C) and check for reaction completion by high-pressure liquid chromatography (HPLC). The reaction is complete when there is < 15 % of the starting material (4-dimethylaminocrotonic acid hydrochloride) present and/or < 2 % of oxalyl chloride (detected as the dimethyl oxalate).
B. 4-Dimethy!amino-but-2-enoic acid |4-(3-chloro-4-fluoro-phenylamino)-3-cyano-7- ethoxy-quinolin-6-yll-amide (crude)
A 3 L multi-neck flask equipped with agitator, thermometer, dip tube, and nitrogen protection is charged N-methyl pyrrolidinone (0.77 kg, 0.75 L, d=1.033 g/mL). At ambient temperature is added 4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7- ethoxy quinoline (0.0748 kg). The reaction mixture is heated to 40-45° C and maintained for about (15 minutes). The reaction mixture is cooled to (0-10° C) and the light suspension of 4-(dimethylamino)-2-butenoyl chloride hydrochloride in CH3CN added via dip tube and positive nitrogen pressure, over (30-45 minutes) while maintaining the temperature (0-10° C) for at least (2 hours). Reaction completion is monitored by HPLC. The reaction is complete when there is < 2 % of the starting material (4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7-ethoxy quinoline) present. To a 12 L multi-neck flask equipped with agitator, thermometer, dip tube, and nitrogen protection is charged with water (2.61 kg, 2.61 L) and sodium bicarbonate (0.209 kg) with stirring until a solution is obtained followed by cooling to (20-24° C) to which is transferred the reaction mixture above which contains < 2 % of the starting material (4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7-ethoxy quinoline), via dip tube and positive nitrogen pressure, to the 12 L flask over about (45-60 minutes) while maintaining the temperature at (20-24° C). The temperature is maintained at (20-24° C) for at least (1 hour). Filter the reaction mixture on a Buchner funnel, rinse with water (3 x 0.40 kg, 3 x 0.40 L), and maintain suction until dripping stops. Dry the product in a vacuum oven at about (50° C) and about (10 mm Hg) for about (28-30 hours). The yield is 78.5 g (86%) at 79.7% strength and 12.3% total impurities.
4-Dimethylamino-but-2-enoic acid r4-(3-chloro-4-fluoro-phenylamino -3-cyano-7- ethoxy-quinolin-6-vn-amide (purified small scale)
First crop: A 6 L multi-neck flask equipped with agitator, condenser, temperature probe, and nitrogen protection is charged with acetonitrile (3.14 kg, 4.00 L) followed by adding 4-dimethylamino-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-3-cyano-7- ethoxy-quinolin-6-yl]-amide (0.16 kg, 0.167 moles). Heat the mixture to (75-80° C) and hold it for (1 hour). Cool the mixture to (70-75° C) and filter on a pad of diatomaceous earth to remove inorganic salts. Wash the pad with acetonitrile (2 x 0.24 kg, 2x 0.30 L), preheated to (70-75° C). Concentrate the filtrate at (20-30 mm Hg) and a maximum temperature of (40-45° C) to a volume of ( 1.2 L). To the concentrate (slurry) add prefiltered tetrahydrofuran (0.53 kg, 0.60 L). Heat to (65-70° C) to obtain a complete solution. Cool the mixture to (40-45° C) over (0.3 hours). Add seeds and continue cooling to (20-25° C) over (1 hour). Hold at (20-25° C) for a minimum of (18 hours). Collect the solid on a Buchner funnel and wash the collected solid with a prefiltered and precooled at (0-5° C) mixture of acetonitrile/tetrahydrofuran (2/1 by volume) (2 x .06 kg, 2 x 0.08 L). Dry the product in a vacuum oven at (50° C) and (10 mm Hg) for (48 hours) to a loss on drying (LOD) of less than (0.5 %). All washes and concentrates (mother liquors) are saved for further purification.
Second crop:
A 3 L multi-neck flask equipped with agitator, temperature probe, nitrogen protection, and charge with the mother liquors and washes from above. Concentrate by distillation at (20-30 mm Hg) and a maximum temperature of (40-45° C) to a volume of (0.50 L). Collect the solid on a Buchner funnel and wash the solid with prefiltered acetonitrile (0.04 kg, 0.05 L). Dry the solid product in a vacuum oven at (50° C) and (10 mm Hg) for (18 hours). A 1 L multi-neck flask equipped with agitator, condenser, temperature probe, nitrogen protection and charge with prefiltered acetonitrile (0.47 kg, 0.60 L), and the collected solid is heated as a suspension to (70-75° C) over (0.5 hours). Add prefiltered tetrahydrofuran (0.03 kg, 0.03 L) to the suspension while maintaining the temperature at (70-75° C). Cool the solution to (40-45° C) and add seed crystals. Continue cooling to (20-25° C) over (1 hour) and hold for (2 hours). Collect the resulting solid on a Buchner funnel and wash the collected solid with a prefiltered and precooled to (5° C) mixture of acetonitrile/tetrahydrofuran (20/1 by volume) (2 x 0.02 kg, 2 x 0.03 L). Dry the collected solid in a vacuum oven at (50° C) and (10 mm Hg) for (24 hours) to an LOD of less than (0.5 %). The combined yield is 27.5 g + 30.5 g (73%) in 96.2-98.4% strength and 1.5-1.7% total impurities by high pressure liquid chromatography (HPLC).
4-Dimethylamino-but-2-enoic acid f4-(3-chloro-4-fluoro-phenylamino)-3-cvano-7- ethoxy-quinolin-6-vn-amide (purified larger scale)
Acetonitrile, practical (34.0 kg) and 4-dimethylamino-but-2-enoic acid [4-(3- chloro-4-fluoro-phenylamino)-3-cyano-7-ethoxy-quinolin-6-yl]-amide (2.69 kg crude, 1.53 kg at 100% strength) are charged to a purged (100 L) reactor. Acetonitrile, practical (2.0 kg) is used as rinse for funnel and vessel walls. The brown suspension is heated at 70 to 76° C using a jacket temperature not exceeding 85° C, then held at the latter temperature for a minimum of 45 minutes, not exceeding 60 minutes. The resulting suspension is then filtered on the warm-jacketed (70-76° C) 14″ Aurora filter, while maintaining the batch temperature at 70 to 76° C. The filtrates are collected by pump into a purged (100 L) receiver, while keeping their temperature below 50° C. The diatomaceous earth pad is then washed with warm (70 to 76° C) acetonitrile, practical (3 x 2.5 kg). The filtrates and washes in (100 L) receiver are cooled to 20 to 26° C, then transferred into a stainless steel drum. Acetonitrile, practical (2.0 kg) is used as rinse. After cleaning and purging both vessels, the contents of the stainless steel drum is transferred into the (100 L) receiver. Acetonitrile, practical (2.0 kg) is used as a rinse. The batch is heated at 70 to 76° C without exceeding jacket temperature of 85° C. The batch is filtered by pump through a .0 micron single cartridge filter, while maintaining the contents at 70 to 76° C. Warm (70-76° C) acetonitrile, practical (4.0 kg) is used as rinse for vessel, filters, pump and lines. The filtrate and rinse are collected and maintained below 50° C. The batch is adjusted to 10 to 16° C, then concentrated by vacuum distillation to 28 to 33 L volume: expected distillation temperature 20 to 30° C, distillate volume 32 to 37 L. The suspension is heated to 64 to 70° C without exceeding jacket temperature of 85° C. The resulting solution is cooled to 40 to 46° C, then seeded using 4-dimethylamino-but-2~enoic acid [4-(3-chloro-4-fluoro-phenylamino)-3-cyano- 7-ethoxy-quinolin-6-yl]-amide, purified (0.5 g). The mixture is cooled to 20 to 26° C over 1 hour, then held at the latter temperature for a minimum of 2 hours. The suspension is then cooled at -3 to 3° C over 1 hour, then held for a minimum of 1 hour. The solid product is collected on a 16″ Buchner, then washed with cold (0-5° C) acetonitrile-tetrahydrofuran (20-6 v/v) mixture (2 x 2.5 kg). The wet collected solid is recrystallized once more from acetonitrile-tetrahydrofuran (20-6 v/v) to desired purity. The material is dried in a vacuum oven first at 35 to 45° C (target 40° C) for 4 hours, liquid ring pump, then 45 to 55° C (target 50° C) for 4 hours. After high vacuum is applied at the latter temperature, until LOD <0.5% (90° C, 2 hours, full vacuum) and each of acetonitrile, tetrahydrofuran and 1-methyl-2-pyrrolidinone are below 0.2%. The purified drug substance is milled (Comil), then blended. The yield is 1.10 kg (70.1 %, corrected for starting material). The strength of the material is 98.3% and a total impurities of 1.27%.
………………….
N OXIDE
http://www.google.com/patents/US20130225594
EXAMPLE 19 Formula 57-Compound 19a
19a: (E)-4-((4-((3-Chloro-4-(pyridin-2-ylmethoxy)phenyl)amino)-3-cyano-7-ethoxyquinolin-6-yl)amino)-N,N-dimethyl-4-oxobut-2-en-1-amine oxide
To a solution of compound A (200 mg, 0.36 mmol, 1.0 eq) in CH2Cl2 (20 mL) was added m-CPBA (74 mg, 0.43 mmol, 1.2 eq) and the resulting mixture was stirred at room temperature for 4 h. A saturated aqueous solution of NaHCO3 (20 mL) was then added and the organic layer was separated, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by preparative TLC (CH2Cl2/MeOH, 10/1, v/v) to give (E)-4-((4-((3-chloro-4-(pyridin-2-ylmethoxy)phenyl)amino)-3-cyano-7-ethoxyquinolin-6-yl)amino)-N,N-dimethyl-4-oxobut-2-en-1-amine oxide (20 mg, 10%) as a yellow solid.
LC-MS (Agilent): Rt 3.03 min; m/z calculated for C30H29ClN6O4 [M+H]+ 573.19. found 573.2.
1H NMR: (400 MHz, CD3OD) δ (ppm): 8.98 (s, 1H), 8.57 (m, 1H), 8.39 (s, 1H), 7.92 (td, J=7.2, 1.6 Hz, 1H), 7.72 (d, J=8.0 Hz, 1H), 7.39 (m, 1H), 7.36 (d, J=2.4 Hz, 1H), 7.28 (s, 1H), 7.24-7.13 (m, 3H), 6.74 (d, J=15.6 Hz, 1H), 5.29 (s, 2H), 4.32 (q, J=6.8 Hz, 2H), 4.20 (d, J=7.2 Hz, 2H), 3.28 (s, 6H), 1.57 (t, J=6.8 Hz, 3H).
……………
http://www.google.fm/patents/EP1883631A1?cl=en
Scheme 2 and Scheme 3. Scheme 2
e-Acelamlno^chloro-S-cyano- 7-ethoxy quinoliπe C,4Hi2CIN2O2 +
MW 289.72
25 °C, 5 h 3-Chloro-4-(3-fluorobenzyl)oxy- anillne
C13Hi1CIFNO
MW 251.69
2 h
free base
Scheme 3
6-Acetamlno-4-chloro-3-cyanc~ 7-elhoxy qulnollne C,4H12CIN2O2 +
MW 28972
3-Chlorc-4-fluoronitrobenzene 2-Pyπdyl carblnol 3-Chloro-4-(3-pyndinylmethoxy) 3-Chloro-4-(2-pyrtdlnylmethewy)- C6H3CIFNO2 C6H7NO nitrobenzene anlllne
MW 17555 MW 109 13 C12H9CIN2O3 C12H11CIN2O d=1 1131 g/ml MW 26467 (EM 264) MW 23469
1 h
(HCI salt)
free base
maleate
Example 1
[0078] Synthesis of 3-chloro-4-(2-pyridylmethoxy)nitrobenzene
[0079] 2-pyridinyl carbinol (31.08 g, 1.05 eq) was dissolved in ACN (750 mL) and KOH flakes (85%) were added (20.6 g, 1.25 eq.). The resulting suspension was warmed to 35 °C. A solution of the 3-chloro-4-fluoronitrobenzene (50.0 g, 0.285 mol) in ACN (250 mL) was added at 35-40 °C. The mixture was held for 14 hours. The mixture was then cooled back to 20-25 °C, quenched with H2O (IL) and the resulting slurry filtered and washed with H2O (3 x 100 mL). The resulting product was isolated as a tan solid in 93% yield with a greater than 99.5% purity as determined by HPLC area. Example Ia
[0080] To accomplish the analogous synthesis of 3-chloro-4-(3-fluorobenzyloxy) nitrobenzene, 3-fluorobenzyl alcohol (0.30 kg, 2.39 mole, 1.05 eq) was dissolved in ACN (6.0 L) and to it was added potassium hydroxide flakes (85%) (0.16 kg, 1.25 eq). The resulting suspension was warmed to 35 0C. A solution of the 3-chloro-4-fluoronitrobenzene (0.40 kg, 2.28 mol) in ACN (2.0 L) was added at 35-40 °C. The mixture was held for 18 hours. The mixture was then cooled back to 20-25 °C, quenched with water (8 L) and the resulting slurry filtered and washed with water (2 x 0.40 L). The resulting product was dried at 45 °C, under 10 mm Hg pressure, for 25 hours to give 0.59 kg (92% yield). Example Ib
[0081] To prepare 4-(benzyloxy)3-chloronitrobenzene, benzyl alcohol (0.34 kg, 3.14 mole, 1.10 eq) was dissolved in acetonitrile (1.70 L) and to it was added potassium hydroxide flakes (85%) (0.24 kg, 1.50 eq). The resulting suspension was warmed to 25 0C. A solution of the 3- chloro-4-fluoronitrobenzene (0.50 kg, 2.85 mol, 1.0 eq) in acetonitrile (0.75 L) was added keeping the pot temperature < 45 0C. The mixture was held for 14 h. The mixture was then cooled back to 0-15 0C, quenched with water (2.5 L) and the resulting slurry was filtered and washed with water (2 x 0.50 L). The resulting product was dried at 50 0C, under 10 mm Hg pressure, for 24 hours to give 0.73 kg (97% yield). [0082] Experimental results for the reaction of Example 1 with different bases and solvents are shown in Table 1. The last three entries on Table 1 are large scale runs in which a 5% excess of pyridyl carbinol was used. Table 1 – Preparation of Nitroaryl Intermediate
NA = not applicable
RT = room temperature (20-25 °C)
Example 2
[0083] Preparation of 3-chloro-4-(2-pyridyhnethoxy)aniline from the nitrobenzene product of
Example 1 was accomplished with catalytic hydrogenation using platinum on carbon.
[0084] A typical hydrogenation was done using 6 volumes of THF, 2% by weight of 5%Pt/C (50% water wet), at 25 psi and at 25-30 0C for approximately 4-6 hours. The reaction is slightly exothermic and the temperature will rise to about 30-35 °C. Cooling is necessary to maintain the temperature below 30 0C.
[0085] As a specific example, a mixture of 3-chloro-4-(2-pyridylmethoxy)nitrobenzene (0.15 kg, 0.57 mole) and 2% (w/w) of 5% Pt/C (6.0 g) in tetrahydrofuran (0.90 L) was hydrogenated at 25 psi for at least 5 hours. The mixture was filtered through a celite pad and washed with tetrahydrofuran (0.60 L). The filtrate was distilled to a volume of about 0.75 L and ethanol (1.12 L) was added. Distillation was continued to a volume of about 0.75 L and ethanol (2.85 L) was added. The mixture may be used “as is” in the step of Example 3 below. Example 2 a
[0086] To accomplish an analogous synthesis of 3-chloro-4-(3-fluorobenzyloxy)aniline, zinc (0.464 kg) was added to a mixture of 3-chloro-4-(3-fluorobenzyloxy)nitrobenzene (0.40 kg, 1.42 mole) and ethanol (4.0 L). The mixture was heated to 40-50 °C. A solution of ammonium chloride (0.152 kg) in water (0.80 L) was added over 0.5 hour keeping the pot temperature at 40-50 °C. The mixture was stirred for 2 hours, filtered and washed with hot (40-50 °C) ethanol (2 x 0.40 L). The filtrate was distilled to a volume of about 0.80 L and 2- methyltetrahydrofuran (2.0 L) was added to dissolve the product. Water (0.80 L) and saturated brine (0.40 L) were added and the layers separated. The organic layer was washed with water (0.60 L), and distilled to a volume of about 0.40 L. Ethanol (2.0 L) was added and distillation continued to a volume of 1.2 L. Example 2b
[0087] To prepare 4-(benzyloxy)-3-chloroaniline, a mixture of 4-(benzyloxy)-3- chloronitrobenzene (0.325 kg, 1.23 mole, 1.0 eq) and 1% (w/w) of 5% Pt/C (3.25 g) in isopropanol (3.25 L) was hydrogenated at 25 psi for a minimum of 4.5 h. The mixture was filtered through a celite pad and washed with isopropanol (2.0 L). The filtrates were used as is in the next step.
[0088] Performing the hydrogenation in isopropyl alcohol (PA), methanol (MeOH), or ethanol
(EtOH) may result in the product being contaminated with late eluting impurity that partially precipitates out on standing in solution. It was found that performing the hydrogenation in a solvent where both the product and starting material are soluble, such as tetrahydrofuran
(THF), resulted in greater product purity and required much less solvent. Thus, THF is a preferred solvent for this step. Experimental results showing the effect of different reaction conditions are shown in Table 2. For the larger scale runs, the first aniline intermediate was not isolated (“NI”) before proceeding with the next step.
Table 2 – Hydrogenation to Form First Aniline Intermediate
* Solid impurities noted after reaction completion. ** percent by weight of starting material. Example 3
[0090] Following hydrogenation to form the first aniline intermediate, acid catalyzed coupling was performed to prepare 4~[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6-N- acetylaminoquinoline, as shown below:
[0091] To perform the coupling reaction, the two reactants were heated together in alcohol at 65-78°C over 4-6 hours, yielding the product. The reaction begins as an amber slurry and thickens to a lighter beige slurry as it approaches completion. Upon scaling up from 75 g to 350 g, it proved necessary to add a catalytic amount (0.025 eq.) of methanesulfonic acid to initiate the reaction. As a specific example, 4-chloro-3-cyano-7-ethoxy-6-N- acetylaminoquinoline (0.141 kg, 0.49 mole) was added to the mixture of Example 2, followed by ethanol (0.037 L) to give a suspension. A catalytic amount of methanesulfonic acid (1.17 g) was added at 20-25 C. The resulting slurry was heated to 70-75 C and held for a minimum of 4 hours. Thickening of the slurry was evident after 1.5 hours. Following reaction completion, the mixture was cooled to room temperature and may be used “as is” in the telescoped reaction of Example 4 below. Example 3 a
[0092] To prepare 6-acetamido-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline, ethanol (4.80 L) was added to the aniline solution followed by 4-chloro-3- cyano-7-ethoxy-6-N-acetylaminoquinoline (0.350 kg, 1.11 mole). A catalytic amount of methanesulfonic acid (2.0 ml) was added at 20-250C. The resulting suspension was heated to 70-750C and held for a minimum of 2 h. Thickening of the slurry was evident during this holding period. Following reaction completion, the mixture was used as is in the following telescoped reaction. Example 3 b
[0093] To prepare 6-acetainido-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-quinoline, isopropanol (6.75 L) was added to the aniline solution followed by 4-chloro-3-cyano-7-ethoxy- 6-N-acetylaminoquinoline (0.277 kg, 0.96 mole, 0.78 eq). A catalytic amount of methane sulfonic acid (3.50 ml) was added at 20-250C. The resulting suspension was heated to 80-850C and held for a minimum of 10 hr. Thickening of the slurry was evident during this holding period. Following reaction completion, the mixture was cooled to 25-35 0C, filtered and the cake washed with isopropanol (3 x 0.25 L). The cake was used as is in the following telescoped reaction.
[0094] As solvents EtOH, DMF or other suitable solvent may be used. Experimental results obtained using different solvents and reaction conditions are shown in Table 3. Difficulty filtering the product of this step (noted in several entries on Table 3) was circumvented by not isolating the solid at this point, but telescoping the reaction with the next step. It has been found that on the order of 20 volumes of EtOH were necessary to achieve reasonable stirring, but that the reaction can proceed in only 10 volumes of DMF, without significant loss in purity. [0095] In Table 3, where the entry is labelled NI , the intermediate product was not isolated, but carried into the next reaction step. Table 3 – Coupling Reaction
NR = no reaction, NI = not isolated; ND = not determined; NA = not available
1. Carried through to the deprotection and generation of free base to give 69.5% overall yield.
2. The overall yield after the deprotection and generation of the free base is 76.1%
3. This reaction was not filtered at all but taken as slurry to the next step.
Example 4 – Deprotection
[0096] The deprotection of the quinoline intermediate formed by the coupling reaction using
2N HCl in water is preferred as noted in Table 4 below. As in the previous Examples, the intermediate product of this step is advantageously not isolated, but carried over as a wet cake into the next step.
[0097] Preparation of 4-[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6- aminoquinoline hydrochloride.
[0098] The reaction mixture from the previous step (Example 3) was taken as is and to it was added 2.7N HCl (3.3L) in H2O (16.0 L). The slurry was heated to 700C and held for 19 hours. The resulting slurry was then filtered and rinsed with 1:1 EtOHTH2O (4 x 1.0 L). The product was isolated as a wet cake and carried through to the next step. A small sample was dried at this stage and analyzed. The HCl salt had a strength of 98.9%. Example 4a
[0099] To prepare 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline hydrochloride, the reaction mixture from the previous step was taken as is and to it was added ethanol (1.6 L) and concentrated hydrochloric acid (1.38 L) to bring the pH to 1-3. The suspension was held at 70-75 0C for a minimum of 2 h. After 1 h, the mixture thickens and ethanol (0.80 L) was added. After 2 h, water (6.80 L) was added, the mixture stirred for 1 h and then cooled to 35-45 0C and stirred overnight (12 h). The mixture was filtered and rinsed with 1 : 1 ethanol/water (2 x 0.84 L) at 35-45 0C. The product was isolated as a wet cake and carried through to the next step. Example 4b
[00100] To prepare 6-amino-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7- ethoxyquinoline hydrochloride, the wet cake from the previous step was taken as is and to it was added a 2 N solution of concentrated hydrochloric acid (1.16 L) in methanol (5.84 L). The suspension was heated to 63-68 0C and held for a minimum of 30 h. The mixture was cooled to 20-300C, filtered and rinsed with methanol (2 x 0.30 L). The product was isolated as a wet cake and carried through to the next step. Table 4 – Deprotection
ND = not determined (the product was used in the next step as a wet cake) NA = not available SM= starting material
Example 5 – Preparation of free base
[0100] The 4-[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6-aminoquinoline HCl salt was converted to the corresponding free base by treatment with 10% potassium carbonate (1.8 L) in MeOH (2.82 L). The mixture was stirred for a minimum of 2.5 hours and the pH was 9-10. The product was filtered, washed with 1:1 methanol/water (3 x 0.19 L) and dried (at 45-50 C at a pressure of 10 mm Hg, for 24 hours) to give 0.186 kg of product with an overall yield of 86% over 4 steps.
Example 5 a
[0101] To prepare 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline free base, the 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline hydrochloride salt was converted to its corresponding free base by treatment with 10% potassium carbonate (0.22 kg in 2.27 L water) in methanol (7.21 L) until pH was 10. The mixture was stirred for a minimum of 2 h. The beige suspension was filtered, washed with 1:1 methanol/water (2 x 0.84 L) and dried (45-50 0C, 10 mm Hg, 24 h) to give 0.51 kg of product with an overall yield of 99% over 4 steps. Example 5b
[0102] To prepare 6-amino-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxyquinolme free base, the 6-amino-4-[4-(benzyloxy)-3-chloroamlino]-3-cyano-7-ethoxyqumoline hydrochloride salt was converted to its corresponding free base by treatment with 10% aqueous potassium carbonate (0.213 kg in 2.13 L) in methanol (6.40 L). The mixture was stirred for a minimum of 1.5 h keeping the pH at 9-10. The product was filtered, washed with water (2 x 0.50 L) and dried (50-60 0C, 10 mm Hg, 20 h) to give 0.347 kg of product with an overall yield of 82% over 4 steps.
Example 6 – Side Chain Coupling
[0103] An acid chloride of formula RV(C=O)-Cl, a mixed anhydride or an activated carboxylase R’ 2-(C=O)-LG derived from the corresponding carboxylic acid, may be used to couple a side chain at the 6 position to form a 6-amido-4-amino-3 cyanoquinoline. R’2 may be alkyl of 1-6 carbon atoms, which may be mono- or di-substituted with amino groups or cycloamino groups, or R’2 may be alkenyl of 2-6 carbon atoms which may be mono- or di- substituted with amino groups or cycloamino groups. [0104] Using the 2-step sequence shown below, an activated carboxylate is prepared in situ and coupled with the aniline. Although the acid chloride can be prepared in acetonitile, a better yield was obtained when the acid chloride was prepared in THF. In both cases, the aniline should be dissolved in NMP before amidation. It is believed that formation of product is better due to better solubility of the aniline in a THF/NMP mixture rather than in an ACN/NMP combination.
[0105] The amount of 4-N,N-dimethylaminocrotonic acid needed was 2 equivalents with respect to aniline. A slight undercharge of 1.95 eq of oxalyl chloride was added along with a catalytic amount (3 mol %) of DMF. The acid chloride was formed via the Vilsmeier intermediate. The completion test for the acid chloride reaction consists of quenching an aliquot of the reaction into ethanol and detecting by HPLC the crotonic acid ethyl ester. This method serves as a check to ensure complete consumption of oxalyl chloride. Excess oxalyl chloride will form diethyl oxalate when quenched in ethanol. [0106] The acid chloride is stable after holding for up to 5 hours at 0-10 °C, when decomposition begins. After 20 hours, complete decomposition takes place. If the acid chloride is allowed to warm, decomposition occurs and its effectiveness is diminished. [0107] The quality of the starting crotonic acid also plays a role in this coupling reaction, as commercially available crotonic acid may contain acetic acid. Acetic acid is detrimental to this reaction. 6-N-acetyl quinoline can be formed which is difficult to remove from the final product. The acetic acid can be removed by re-slurrying the crotonic acid in 4 volumes of isopropanol at room tempature, filtering and drying preferably to a level of less than 0.01%. [0108] It was found that the addition of the aniline solution in NMP to the acid chloride gave a better yield as compared to adding the acid chloride to the aniline. The addition is done keeping the temperature at 0-5 °C. The coupling reaction is slow and requires holding overnight at this temperature. It is not desirable to raise the reaction temperature as the stability of the acid chloride diminishes.
[0109] The reaction is quenched using aqueous sodium hydroxide at 40 °C and then filtered at that temperature. Quenching the reaction at 40 0C gives bigger crystals that are easily filterable. It was observed that filtration at 40 °C was faster than at room temperature. The product is recrystallized from a 1.5:1 mixture of acetonitrile:THF (15 volumes) at 70-75 0C. This in-process purification beneficially removes unreacted aniline. The recovery yields are typically greater than 85%.
[0110] To demonstrate a specific synthesis of (E)-N- {4-[3-chloro-4-(2- pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide, a solution of 4-N,N-dimethylaminocrotonic acid hydrochloride (186 g, 1.12 mol) in THF (1.88 L) and a catalytic amount of DMF (2 mL) was cooled to 0-5 °C. Oxalyl chloride (97 mL, 1.09 mol, 0.95 eq) was added dropwise over 45 minutes. The mixture was then warmed to 25-30 °C and stirred for 2 hours. The yellow solution was checked for complete consumption of oxalyl chloride by HPLC, then cooled to 0-5 0C.
[0111] When the reaction is deemed complete, a solution of 4-[4-(2-pyridylniethoxy)-3- chloro]amino-6-amino-3-cyano-7-ethoxyquinoline (250 g, 0.56 mol) in N-methyl-2- pyrolidinone (1.88 L) was added dropwise over 2 hours keeping the temperature at 0-5 °C. The mixture was stirred for at least 3 hours until less than about 2% of the starting aniline remains by HPLC, which takes about 3 hours.
[0112] Upon completion, the reaction was quenched with water (3.0 L), held for 30 minutes and then warmed to 40 °C. Aqueous sodium hydroxide (170 g in 1.25 L water) was added over 1.25 hours to bring the pH to 10-11. The mixture was stirred for an hour, then cooled to room temperature and held for 3 hours. The resulting precipitates were filtered and washed with water (100 mL) and heptane (100 mL). The wet solids were heated to reflux (70-75 °C) in acetonitrile:THF and the solution cooled over 3 hours to room temperature. The product was filtered and washed with cold acetonitrile:THF. The product was dried (40-50 0C, 10 mm Hg, 24 hours) to give 83% uncorrected yield. Example 6a
[0113] In an analogous synthesis of (E)-N- {4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3- cyano-7-ethoxy-6-qumolmyl}-4-(dimethylamino)-2-butenamide, a solution of 4-N5N- dimethylaminocrotonic acid hydrochloride (108 g, 0.65 mole) in tetrahydrofuran (1.13 L) and a catalytic amount of dimethylformamide (1.2 mL) was cooled to 0-5 °c. Oxalyl chloride (55 mL, 0.62 mole, 0.95 eq) was added dropwise over 50 min. The mixture was then warmed to 25-30 °c and stirred for 2 h then cooled to 0-5 °c. N-methyl-2-pyrrolidinone (0.225 L) was added over 25 min followed by a solution of 6-amino-4-[3-chloro-4-(3- fluorobenzyloxy)]anilino-3-cyano-7-ethoxy-quinoline (150 g, 0.32 mol) in N-methyl-2- pyrrolidinone (1.20 L) added dropwise over 2 hours keeping the temperature 0-5 . The mixture was stirred for at least about 3 hours, warmed to 10-15 °c and stirred for a further 12 hours. The mixture is cooled to 0-10 c, quenched by adding water (1.8 L) over 2 hours, and stirred for 30 minutes. The mixture is warmed to 40 °c. Aqueous sodium hydroxide (101 g in 0.75 L water) was added over 1 hour to bring the pH to 10-11. The mixture was stirred for an hour, filtered warm (40 °c) and washed with water (2 x 0.30 L) until the pH of the last wash was about 7. The wet solids were recrystallized by heating to reflux (70-75 °c) in 60:40 acetonitrile:tetrahydrofuran (2.25 L) and the solution cooled over 3 hours to room temperature. The product was filtered and washed with cold 60:40 acetonitrile:tetrahydrofuran (2 x 0.30 L). The product was dried (40-50 °c, 10 mm Hg, 16 h) to give 0.154 kg (83% yield). Example 6b
[0114] To prepare (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide free base, a solution of 4-N,N-dimethylaminocrotonic acid hydrochloride (18.6 g, 112 mmole) in acetonitrile (295 ml) and a catalytic amount of dimethylformamide (0.25 mL) was cooled to 0-5 °c. Oxalyl chloride (9.3 mL, 106 mmole, 0.95
Op eq) was added dropwise over 5 min. The mixture was then warmed to 25-30 and stirred for 1-1.5 h then cooled to 0-10 °c. A solution of 6-amino-4-[4-(benzyloxy)-3-cliloroanilino]-3- cyano-7-ethoxy-quinoline (25 g, 56 mmole) in N-methyl-2-pyrrolidinone (175 ml) was added dropwise over 30 min keeping the temperature 0-10 °c. The mixture was stirred for a minimum of 1 h at 0-10 °c. After reaction completion, the mixture was quenched by dropwise addition to a solution of sodium bicarbonate (69.7 g in 870 ml water) over 30 mins. and stirred overnight while warming to room temperature. The mixture was filtered and washed with water (3 x 25 ml). The crude product was recrystallized in refluxing (80-82 °c) acetonitrile (570 ml). The product was dried (45-50 °c, 10 mm Hg, 28 h) to give 12.81 g (41% yield). 1H NMR : δ (DMSO-d6) 9.44 (s, IH, NH), 8.97 (s, IH, Ar), 8.44 (s, IH, Ar), 7.53-7.35 (m, 7H, Ar), 7.35- 7.10 (in, 2H, Ar), 6.78 (dt, IH, -CH2CH=CH-), 6.59 (d, IH, -CH2CH=CH-), 5.21 (s, 2H, OCH2Ph), 4.30 (q, 2H, OCH2CH3), 3.07 (s, 2H, NCH2), 2.18 (s, 6H, N(CHs)2), 1-47 (t, 3H, OCH2CH3).
[0115] Results obtained with different reaction procedures at different degrees of scale-up for synthesis of the 2-pyridylmethoxy analog are shown in Table 5. Table 5 – Side Chain Coupling
* TI = total impurities
[0116] Purificatiuon of the product is conducted by recrystallization in a suitable solvent followed by reslurrying with water followed by additional recrystallization, as necessary. As noted in Table 6, in the synthesis of the 2-pyridylmethoxy analog, several trials in different solvents did not result in the isolation of a single polymorphic form of the product. Table 6
Example 7 – Formation of Salt
[0117] The free base is hygroscopic and undergoes hydrolysis readily. Forming a salt of the compound, such as a fumarate or mesylate salt, stabilizes the molecule and renders the compound more soluble. The most preferred salt is a maleate salt, which has been found to be highly crystalline and to exist substantially as a single polymorph as shown by DSC thermogram in Fig. 1.
[0118] Recrystallizing the product in the presence of an acid has been found to yield a stable salt form of the product. Experimental results achieved utilizing different solvents for the recrystallization are set forth in Table 7. As seen in Table 7, an improvement is observed when n-propanol/water is used as the solvent system. A maleate salt is the most preferred, as it exists in a single polymorphic form. Table 7 – Recrystallization
Preparation of (E)-N- {4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6- quinolinyl} -4-(dimethylamino)-2-butenamide maleate, WAY- 179272-B
[0120] (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- dimethylamino)-2-butenamide crude free base (0.1 kg, 0.159 mole) and maleic acid (0.019 kg, 0.164 mole) were dissolved at 40-50 in a 10% water/n-propanol mixture (1.20 L). The hot solution was clarified and cooled over 2 h to room temperature and held for 12-15 hr. The product was filtered and washed with 10% water/n-propanol (2 x 0.15 L). The product was dried (50 °c, 10 mm Hg, 24 h) to give 94.4 g (88% yield). DSC: 204 °c (single crystal form). 1H NMR : δ (DMSO-d6) 9.73 (s, IH, NH), 9.62 (s, IH, NH), 8.93 (s, IH, Ar), 8.60 (dd, IH, Ar), 8.50 (s, IH, Ar), 7.88 (dd, IH, Ar), 7.58 (d, IH, Ar), 7.40 (m, 3H, Ar), 7.24 (m, 2H, Ar), 6.75 (d, 2H, -CH=CH-), 6.03 (s, 2H, HOOC-CH=CH-COOH), 5.29 (s, 2H, OCH2PVr), 4.33 (q, 2H, OCH2CH3), 3.89 (s, 2H, NCH2), 2.76 (s, 6H, N(CH3)2), 1.47 (t, 3H, OCH2CH3). 13C NMR : δ (DMSO-d6) 168.0, 163.2, 156.9, 154.2, 153.2, 151.9, 151.3, 149.8, 148.5, 137.8, 136.5, 134.7, 133.4, 132.2, 128.0, 126.6, 124.9, 123.8, 122.3, 122.2, 117.9, 116.4, 115.1, 113.9, 109.5, 88.1, 72.0, 65.3, 57.8, 43.1, 14.9.
Example 7a
To prepare (E)-N- {4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7-ethoxy-6- quinolinyl}-4-(dimethylamino)-2-butenamide dimaleate,
(E)-N- {4-[3-chloro-4-(3- fluorobenzyloxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-dimethylamino)-2-butenamide crude free base (0.516 kg, 0.90 mole) and maleic acid (0.214 kg, 1.84 mole) were dissolved at 40-50 °c in a 6.5% water/n-propanol mixture (12.60 L). The hot solution was clarified, rinsed with 5% water/n-propanol (0.52 L) and n-propanol (2.0 L). The mixture was held at 45 for 3 hr, cooled over 2 h to room temperature and held overnight. The mixture was further cooled to 5-10 °c. The product was filtered and washed with cold 5% water/n-propanol (0.52 L). The product was dried (45 °c, 10 mm Hg, 16-24 h) to give 0.586 kg (81% yield). DSC: 184 °c (single crystal form). 1HNMR : δ (DMSO-d6) 9.77 (s, IH, NH), 8.95 (s, IH, Ar), 8.53 (s, IH, Ar), 7.49-7.16 (m, 8H, Ar), 6.78 (m, 2H, -CH=CH-), 6.15 (s, 4H, 2 x HOOC-CH=CH-COOH), 5.26 (s, 2H, OCH2PyT), 4.33 (q, 2H, OCH2CH3), 3.97 (dd, 2H, NCH2), 2.82 (s, 6H, N(CEb)2), 1.47 (t, 3H, OCH2CH3). 13C NMR : δ (DMS0-d6) 167.0, 163.8, 162.3, 160.6, 153.6, 152.2, 151.3, 150.8, 139.5, 139.4, 133.7, 133.2, 132.2, 131.8, 130.5, 130.4, 127.4, 126.1, 124.3, 123.3, 121.7, 116.9, 115.7, 114.8, 114.5, 114.4, 114.1, 113.8, 113.1, 108.1, 87.2, 69.5, 64.6, 56.9, 42.1, 14.2. Example 7b
[0122] To prepare (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide maleate, (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano- 7-ethoxy-6-quinolinyl}-4-dimethylamino)-2-butenamide crude free base (2.0 g, 3.6 mmole) and maleic acid (0.43 g, 3.7 mmole) were mixed at 40-50 c in a 10% water/n-propanol mixture (24 ml) for 2 hr. The mixture was cooled to ambient temperature, filtered and washed with 10% water/n-propanol (2 x 3 ml). The product was dried (40 °c, 10 mm Hg, 24 h) to give 0.32 g (13% yield). 1HNMR : δ (DMSO-d6) 9.75 (s, IH, NH), 8.95 (s, IH, Ar), 8.49 (s, IH, Ar), 7.49-7.37 (m, 7H, Ar), 7.23 (dd, 2H, Ar), 6.78 (s, 2H, -CH2CH=CH-), 6.06 (s, 2H, HOOC- CH=CH-COOH), 5.22 (s, 2H, OCH2Ph), 4.31 (q, 2H, OCH2CH3), 3.93 (s, 2H, NCH2), 2.79 (s, 6H, N(CH3)2), 1.46 (t, 3H, OCH2CH3).13C NMR : δ (DMSO-d6) 167.9, 163.1, 154.2, 153.3, 152.1, 151.3, 148.5, 137.3, 136.3, 134.5, 133.2, 132.3, 129.3, 129.2, 128.7, 128.3, 128.2, 128.0, 126.7, 124.9, 122.4, 117.9, 116.4, 115.2, 113.9, 109.5, 88.0, 71.1, 65.3, 57.7, 43.0, 15.0. [0123] (E)-N-{4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- dimethylamino)-2-butenamide crude free base (2.0 g, 3.6 mmole) and maleic acid (0.43 g, 3.7 mmole) were mixed at 40-50 °c in a 10% water/n-propanol mixture (24 ml) for 2 hr. The mixture was cooled to ambient temperature, filtered and washed with 10% water/n-propanol (2 x 3 ml). The product was dried (40 °c, 10 mm Hg, 24 h) to give 0.32 g (13% yield). 1H NMR : δ (DMSO-d6) 9.75 (s, IH, NH), 8.95 (s, IH, Ar), 8.49 (s, IH, Ar), 7.49-7.37 (m, 7H, Ar), 7.23 (dd, 2H, Ar), 6.78 (s, 2H, -CH2CH=CH-), 6.06 (s, 2H, HOOC-CH=CH-COOH), 5.22 (s, 2H, OCH2Ph), 4.31 (q, 2H, OCH2CH3), 3.93 (s, 2H, NCH2), 2.79 (s, 6H, N(CH3)2), 1.46 (t, 3H, OCH2CH3). 13C NMR : δ (DMSO-d6) 167.9, 163.1, 154.2, 153.3, 152.1, 151.3, 148.5, 137.3, 136.3, 134.5, 133.2, 132.3, 129.3, 129.2, 128.7, 128.3, 128.2, 128.0, 126.7, 124.9, 122.4, 117.9,
116.4, 115.2, 113.9, 109.5, 88.0, 71.1, 65.3, 57.7, 43.0, 15.0.
……………….
http://www.google.com/patents/WO2009052264A2?cl=en
TABLE 1 1. STRUCTURES OF DEGRADATION PRODUCT AND PROCESS IMPURITIES
N-{4-[3-chloro-4-(2- (E)-4-({4-[3-chloro-4-(2- N -{4-[3-chloro-4-(2- pyrιdιnylmethoxy)anιlιno]-3-cyano-7- pyrιdιnylmethoxy)anιlιno]-3-cyano-7- pyrιdιnylmethoxy)anιlιno]-3-cyano-7-ethoxy- ethoxy-6-quιnolιnyl}acetamιde ethoxy-6-quιnolιnyl}amιno)-N,N,N- 6-quιnolιnyl}-N2,N2-dιmethylethanedιamιde trιmethyl-4-oxo-2-buten-1-amιnιum
Exact Mass 487 14 Exact Mass 544 16
Exact Mass 571 22
Process Impurity I Process Impurity J
SCHEME 1
The reaction of the free base and maleic acid occurs at an elevated temperature of from about 40 0C to about 60 0C, preferably between about 4O0C to about 5O0C. The ratio of watenn- propanol may vary, for example between about 1 :10 to about 1 :5, and the optimal ratio of watenn-propanol is about 1 :9. The water-alcohol solution may comprise from about 5% to about 20% by volume water and from about 80% to about 95% by volume alcohol. The alcohol may be n-propanol. In one embodiment, the water-alcohol solution comprises about 10% by volume water and about 90% by volume n-propanol. The volume of the solvent solution may be between about 8 to about 25 volumes, including about 10 to about 12 volumes. About 1.0-1.2 equivalents of maleic acid is used per equivalent of the free base, preferably about 1.03 equivalents of maleic acid per equivalent of the free base.
The resulting solution of the maleate salt may be clarified by filtration prior to cooling. The cooling step may be continued until the solution reaches a temperature of about 45°C or less, including a temperature of about 39°C or less, and more preferably to about 300C or less. In one embodiment, the solution is filtered after cooling to about room temperature, preferably from about 230C to about 25 0C. Typically, the maleate salt begins to crystallize out of solution once the temperature reaches 370C or below. The solution may be allowed to sit for at least 12 hours, preferably about 12 to about 15 hours at room temperature, and is then filtered and washed to recover the crystalline maleate salt product. The resulting filter cake may be washed with the same or a different water-alcohol solution to obtain the product. The product may be dried to obtain crystalline (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7- ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide maleate. At this point, the maleate salt product recovered and isolated is typically in the form of the monohydrate form of the maleate salt.
……………
PAPTENT
http://www.google.com/patents/CN102731395A?cl=en
The present invention relates to a process for preparing that imatinib (neratinib, HKI-272) is a new method for its preparation and its intermediates in the preparation to the application that imatinib
[0155] Example 14 (E)-N-(4 – (3 – chloro-4 – (2 – pyridyl) phenyl) amino] _3_ ethoxy-quinolin-6-cyano-_7_ – yl) -4 – dimethylamino-2 – butene amide
[0156]
Compound of Example 13 (20mg, 0. 037mmol) was dissolved in DMF was added potassium carbonate (10mg, 0. 07mmol), dimethylamine hydrochloride (5mg, 0. 06mmol), at room temperature for I hour, after , the reaction mixture was dropped into water, stirred for 10 minutes, filtered, washed with water and dried to give the title compound 1511 ^ 75% yield.1HNMR (300MHz, DMS0_d6): δ I. 5 (t, 3H, J = 6 · 8,13. 8), 2. 2 (br s, 6H), 3. I (d, 2H, J = 3. 8 ), 4. 3 (q, 2H, J = 7. 0,14. 2), 5. 2 (s, 2H),
6. 6 (d, 1H, J = 15. 0), 6. 8 (m, 1H), 7. 1-7. 3 (m, 2H), 7. 3-7. 4 (m, 3H), 7. 6 (d, 1H, J = 3. 9),
7. 9 (d, 1H, J = 3. 9), 8. 5 (s, 1H), 8. 6 (d, 1H, J = 3. 9), 9. 0 (s, 1H), 9. 5 (s, 1H), 9. 6 (s, 1H). ESI-MS: [M + H] + = 557. 3.
GOING BACKWARDS…………………
Example 13 (E) -4 – bromo-N-(4 – (3 – chloro-4 – (2 – pyridyl) phenyl) amino] _3_ cyano _7_ ethoxyquin -6 – yl) -2 – butene amide
Example 12 Compound (100mg, 0. 2mmol) was suspended in carbon tetrachloride was added NBS (40mg,
O. 22mmol), benzoyl peroxide (2mg, 0. Olmmol), nitrogen, refluxed for 10 hours, the reaction solution directly mixed baby gel, silica gel column chromatography to obtain the title compound isolated 60mg, yield 51%. 1HnmrgoomHz, cdci3): δ i.6 (t, 3H, J = 6. 8,13. 7), 2. 0 (d, 2H, J = 6. 9), 4. 3 (q, 2H, J = 7. 2,13. 8), 5. 3 (s, 2H), 6. I (d, 1H, J =
15. 0), 7. 0 (m, 1H), 7. 2 (m, 1H), 7. 3 (s, 1H), 7. 4 (s, 1H), 7. 6 (d, 1H, J = 8. 2), 7. 8 (d, 1H, J =
7. 6), 8. 0 (s, 1H), 8. 5 (s, 1H), 8. 6 (d, 1H, J = 4. 7), 9. 2 (s, 1H). ESI-MS: [M + H] + = 594. I.
……………
PAPER
Optimization of 6,7-disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity
J Med Chem 2005, 48(4): 1107
http://pubs.acs.org/doi/full/10.1021/jm040159c
(E)-N-{4-[3-Chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide (25o).
This compound was prepared as a yellow solid (0.86 g, 85%) by the method described for 25g using 0.65 g (1.81 mmol) of 23 and 0.42 g (3.62 mmol) of 3-chloro-4-(2-pyridinylmethoxy)aniline:
HRMS (ES+) m/z 557.205 89 (M + H)+1, Δ = −0.36 mmu;
1H NMR (DMSO-d6) δ 9.62 (s, 1H), 9.49 (s, 1H), 8.96 (s, 1H),
8.60 (d, 1H, J = 3.9 Hz), 8.47 (s, 1H),
7.88 (t, 1H, J = 3.9 Hz), 7.58 (d, 1H, J = 3.9 Hz),
7.39−7.35 (m, 3H), 7.26 (d, 1H, J = 7.8 Hz),
7.19 (d, 1H, J = 8.1 Hz), 6.81−6.73 (m, 1H),
6.59 (d, 1H, J = 7.8 Hz), 5.28 (s, 2H),
4.30 (q, 2H, J = 6.9 Hz),
3.07 (d, 2H, J = 3.9 Hz),
2.17 (s, 6H),
1.46 (t, 3H, J = 3.9 Hz).
Anal. (C30H29ClN6O3·1.1H2O) C, H, N.
INTERPRETATION
1H NMR : δ (DMSO-d6)
9.44 (s, IH, NH),
8.97 (s, IH, Ar),
8.44 (s, IH, Ar),
7.53-7.35 (m, 7H, Ar),
7.35- 7.10 (in, 2H, Ar),
6.78 (dt, IH, -CH2CH=CH-),
6.59 (d, IH, -CH2CH=CH-),
5.21 (s, 2H, OCH2Ph),
4.30 (q, 2H, OCH2CH3),
3.07 (s, 2H, NCH2),
2.18 (s, 6H, N(CHs)2),
1-47 (t, 3H, OCH2CH3).
References
- “Definition of neratinib – National Cancer Institute Drug Dictionary”. Retrieved 2008-12-01.
- Rabindran SK, Discafani CM, Rosfjord EC, et al. (June 2004). “Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase”. Cancer Res. 64 (11): 3958–65. doi:10.1158/0008-5472.CAN-03-2868. PMID 15173008.
- ClinicalTrials.gov NCT00398567 A Phase 1/2 Study Of HKI-272 In Combination With Herceptin In Subjects With Advanced Breast Cancer
- “Puma Acquires Global Rights to Pfizer’s Phase III Breast Cancer Drug Neratinib”.
- Minami Y, Shimamura T, Shah K, et al. (July 2007). “The major lung cancer-derived mutants of ERBB2 are oncogenic and are associated with sensitivity to the irreversible EGFR/ERBB2 inhibitor HKI-272”. Oncogene 26 (34): 5023–7. doi:10.1038/sj.onc.1210292.PMID 17311002.
- http://www.reuters.com/article/idUSN1612347120100317 “Breast cancer study aims to speed drugs, cooperation” March 2010
- Sequist L.V., Besse B., Lynch T.J. and all; Neratinib, an Irreversible Pan-ErbB Receptor Tyrosine Kinase Inhibitor: Results of a Phase II Trial in Patients With Advanced Non-Small-Cell Lung Cancer., J. Clin. Oncol., 2010, May 17.
PubMed PMID: 20479403. - Belani CP. The role of irreversible EGFR inhibitors in the treatment of non-small cell lung cancer: overcoming resistance to reversible EGFR inhibitors. Review. Cancer Invest. 2010, 28(4), 413-423. Review.
PubMed PMID: 20307200. - TSOU H-R ET AL: “Optimization of 6,7-Disubstituted-4-(arylamino)quinoline-3 -carbonitr iles as Orally Active, Irreverible Inhibitors of HEGFR-2 Kinase Activity” JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 48, 27 January 2005 (2005-01-27), pages 1107-1131, XP002414228 ISSN: 0022-2623 cited in the application
- Optimization of 6,7-disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity
J Med Chem 2005, 48(4): 1107
2-20-2009
|
Novel Combinational Use of Sulfonamide Compound
|
|
9-12-2008
|
Multi-Functional Small Molecules as Anti-Proliferative Agents
|
|
5-11-2007
|
Antineoplastic combinations with mTOR inhibitor,herceptin, and/or hki-272
|
|
11-31-2006
|
Methods of synthesizing substituted 3-cyanoquinolines and intermediates thereof
|
|
11-31-2006
|
Methods of synthesizing 6-alkylaminoquinoline derivatives
|
|
10-25-2006
|
Synthesis of 4-(amino)-2-butenoyl chlorides and their use in the preparation of 3-cyano quinolines
|
5-30-2012
|
Amide derivative for inhibiting the growth of cancer cells
|
|
9-21-2011
|
Maleate salts of (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide and crystalline forms thereof
|
|
8-26-2011
|
COMPOUNDS THAT MODULATE EGFR ACTIVITY AND METHODS FOR TREATING OR PREVENTING CONDITIONS THEREWITH
|
|
5-7-2010
|
Antineoplastic Combinations of 4-Anilino-3-Cyanoquinolines and Capecitabine
|
|
4-30-2010
|
METHOD FOR PREDICTION OF THE EFFICACY OF VASCULARIZATION INHIBITOR
|
|
4-16-2010
|
METHOD FOR ASSAY ON THE EFFECT OF VASCULARIZATION INHIBITOR
|
|
3-19-2010
|
PHARMACEUTICAL COMPOSITIONS OF AN SRC KINASE INHIBITOR AND AN AROMATASE INHIBITOR
|
|
2-26-2010
|
Heterocyclic N-Oxides as Hypoxic Selective Protein Kinase Inhibitors
|
|
12-18-2009
|
Antineoplastic Combinations Containing HKI-272 and Vinorelbine
|
|
12-4-2009
|
ANTINEOPLASTIC COMBINATIONS WITH mTOR INHIBITOR, TRASTUZUMAB, AND/OR HKI-272
|
NMR
Vorapaxar …FDA advisory panel votes to approve Merck & Co’s vorapaxar
VORAPAXAR
Thrombosis, Antiplatelet Therapy, PAR1 Antagonists , MERCK ..ORIGINATOR
Ethyl N-[(3R,3aS,4S,4aR,7R,8aR,9aR)-4-[(E)-2-[5-(3-fluorophenyl)-2-pyridyl]vinyl]-3-methyl-1-oxo-3a,4,4a,5,6,7,8,8a,9,9a-decahydro-3H-benzo[f]isobenzofuran-7-yl]carbamate
618385-01-6 CAS NO
Vorapaxar (formerly SCH 530348) is a thrombin receptor (protease-activated receptor, PAR-1) antagonist based on the natural product himbacine. Discovered by Schering-Plough and currently being developed by Merck & Co., it is an experimental pharmaceutical treatment for acute coronary syndrome chest pain caused by coronary artery disease.[1]
In January 2011, clinical trials being conducted by Merck were halted for patients with stroke and mild heart conditions.[2] In a randomized double-blinded trial comparing vorapaxar with placebo in addition to standard therapy in 12,944 patients who had acute coronary syndromes, there was no significant reduction in a composite end point of death from cardiovascular causes, myocardial infarction, stroke, recurrent ischemia with rehospitalization, or urgent coronary revascularization. However, there was increased risk of major bleeding.[3]
A trial published in February 2012, found no change in all cause mortality while decreasing the risk of cardiac death and increasing the risk of major bleeding.[4]
SCH-530348 is a protease-activated thrombin receptor (PAR-1) antagonist developed by Schering-Plough and waiting for approval in U.S. for the oral secondary prevention of cardiovascular events in patients with a history of heart attack and no history of stroke or transient ischemic attack. The drug candidate is being investigated to determine its potential to provide clinical benefit without the liability of increased bleeding; a tendency associated with drugs that block thromboxane or ADP pathways. In April 2006, SCH-530348 was granted fast track designation in the U.S. for the secondary prevention of cardiovascular morbidity and mortality outcomes in at-risk patients.
Vorapaxar was recommended for FDA approval on January 15, 2014.[5]
17 JAN 2014
FDA advisory panel votes to approve Merck & Co’s vorapaxar REF 6
VORAPAXAR SULPHATE
CAS Number: 705260-08-8
Molecular Formula: C29H33FN2O4.H2O4S
Molecular Weight: 590.7
Chemical Name: Ethyl [(1R,3aR,4aR,6R,8aR,9S,9aS)-9-[(1E)-2-[5-(3-fluorophenyl)pyridin-2- yl]ethenyl]-1-methyl-3-oxododecahydronaphtho[2,3-c]furan-6-yl]carbamate sulfate
Synonyms: Carbamic acid, [(1R,3aR,4aR,6R,8aR,9S,9aS)-9-[(1E)-2-[5-(3-fluorophenyl)-2- pyridinyl]ethenyl]dodecahydro-1-methyl-3-oxonaphtho[2,3-c]furan-6-yl]-,ethyl ester,sulfate; SCH-530348
Vorapaxar Sulfate (SCH 530348) a thrombin receptor (PAR-1) antagonist for the prevention and treatment of atherothrombosis.
……………………
GENERAL INTRO
SIMILAR NATURAL PRODUCT
+ HIMBACINE
Himbacine is an alkaloid muscarinic receptor antagonist displaying more potent activity associated with M2 and M2 subtypes over M1 or M3. Observations show himbacine bound tightly to various chimeric receptors in COS-7 cells as well as possessed the ability to bind to cardiac muscarinic receptors allosterically. Recent studies have produced series of thrombin receptor (PAR1) antagonists derived from himbacine Himbacine is an inhibitor of mAChR M2 and mAChR M4.
Physical State: | Solid |
Derived from: | Australian pine Galbulimima baccata |
Solubility: | Soluble in ethanol (50 mg/ml), methanol, and dichloromethane. Insoluble in water. |
Storage: | Store at -20° C |
Melting Point: | 132-134 °C |
Boiling Point: | 469.65 °C at 760 mmHg |
Density: | 1.08 g/cm3 |
Refractive Index: | n20D 1.57 |
Optical Activity: | α20/D +51.4º, c = 1.01 in chloroform |
Application: | An alkaloid muscarinic receptor antagonist |
CAS Number: | 6879-74-9 |
Molecular Weight: | 345.5 |
Molecular Formula: | C22H35NO2 |
general scheme:
……………………………
SYNTHESIS
THE EXACT BELOW COMPD IS 14
Example 2
Step 1 :
Phosphonate 7, described in US 6,063,847, (3.27 g, 8.1 mmol) was dissolved in THF (12 ml) and C(O)Oled to 0 °C, followed by addition of 2.5 M n- BuLi (3.2 ml, 8.1 mmol). The reaction mixture was stirred at 0 °C for 10 min and warmed up to rt. A solution of aldehyde 6, described in US 6,063,847, in THF (12 ml) was added to the reaction mixture. The reaction mixture was stirred for 30 min. Standard aqueous work-up, followed by column chromatography (30-50% EtOAc in hexane) afforded product 8. 1HNMR (CDCI3): δ 0.92-1.38 (m, 31 H), 1.41 (d, J= 6 Hz, 3H), 1.40-1.55 (m, 2H), 1.70-1.80 (m, 2H), 1.81-1.90 (m, 2H), 2.36 (m, 2H), 2.69 (m, 1 H), 3.89 (m, 4H), 4.75 (m, 1 H), 6.28-6.41 (m, 2H), 7.05-7.15 (m, 2H), 8.19 (br s, 1 H). Step 2:
Compound 8 (2.64 g, 4.8 mmol) was dissolved in THF (48 ml). The reaction mixture was C(O)Oled to 0 °C followed by addition of 1 M TBAF (4.8 ml). The reaction mixture was stirred for 5 min followed by standard aqueous work-up. Column chromatography (50% EtOAc/hexane) afforded product 9 (1.9 g, 100%). 1HNMR (CDCI3): δ 1.15-1.55 (m, 6H), 1.41 (d, J= 6 Hz, 3H), 1.70-1.82 (m, 3H), 1.85-1.90 (m, 1 H), 2.36 (m, 2H), 2.69 (m, 1 H), 3.91 (m, 4H), 4.75 (m, 1 H), 6.18- 6.45 (m, 2H), 7.19 (br s, 2H), 8.19 (br s, 1 H). Step 3:
To a solution of compound 9 (250 mg, 0.65 mmol) in pyridine (5 ml) C(O)Oled to 0 °C was added Tf2O (295 μL, 2.1 mmol). The reaction mixture was stirred overnight at rt. Standard aqueous work-up followed by column chromatography afforded product 10 (270 mg, 80%). 1HNMR (CDCI3): δ 1.15-1.55 (m, 6H), 1.41 (d, J= 6 Hz, 3H), 1.70-1.82 (m, 3H), 1.85-1.90 (m, 1 H), 2.36 (m, 2H), 2.69 (m, 1 H), 3.91 (m, 4H), 4.75 (m, 1 H), 6.42-6.68 (m, 2H), 7.25 (m, 1 H), 7.55 (m, 1 H), 8.49 (d, J= 2.8 Hz, 1 H).
Compound 10 (560 mg, 1.1 mmol), 3-fluorophenyl boronic acid (180 mg, 1.3 mmol) and K2CO3 (500 mg, 3.6 mmol) were mixed with toluene (4.4 ml), H2O (1.5 ml) and EtOH (0.7 ml) in a sealed tube. Under an atmosphere of N2, Pd(Ph3P)4 (110 mg, 0.13 mmol) was added. The reaction mixture was heated at 100 °C for 2 h under N2. The reaction mixture was C(O)Oled down to rt, poured to EtOAc (30 ml) and washed with water (2X20 ml). The EtOAc solution was dried with NaHCO3 and concentrated at reduced pressure to give a residue. Preparative TLC separation of the residue (50% EtOAc in hexane) afforded product 11 (445 mg, 89%). 1HNMR (CDCI3): δ 1.15-1.59 (m, 6H), 1.43 (d, J= 6 Hz, 3H), 1.70-1.79 (m, 2H), 1.82 (m, 1H), 1.91 (m, 2H), 2.41 (m, 2H), 2.69 (m, 1 H), 3.91 (m, 4H), 4.75 (m, 1 H), 6.52-6.68 (m, 2H), 7.15 (m, 1 H), 7.22 (m, 2H), 7.35 (m, 1 H), 7.44 (m, 1 H), 7.81 (m, 1 H), 8.77 (d, J= 1.2 Hz, 1 H). Step 5:
Compound 11 (445 mg, 0.96 mmol) was dissolved in a mixture of acetone (10 ml) and 1 N HCI (10 ml). The reaction mixture was heated at 50 °C for 1 h.
Standard aqueous work-up followed by preparative TLC separation (50% EtOAc in hexane) afforded product 12 (356 mg, 89%). 1HNMR (CDCI3): δ 1.21-1.45 (m, 2H), 1.47 (d, J= 5.6 Hz, 3H), 1.58-1.65 (m, 2H), 2.15 (m, 1 H), 2.18-2.28 (m, 2H), 2.35- 2.51 (m, 5H), 2.71 (m, 1 H), 4.79 (m, 1 H), 6.52-6.68 (m, 2H), 7.15 (m, 1 H), 7.22 (m, 2H), 7.35 (m, 1 H), 7.44 (m, 1 H), 7.81 (m, 1 H), 8.77 (d, J= 1.2 Hz, 1 H). Step 6:
Compound 12 (500 mg, 4.2 mmol) was dissolved in EtOH (40 ml) and CH2CI2 (15 ml) NH3 (g) was bubbled into the solution for 5 min. The reaction mixture was C(O)Oled to 0 °C followed by addition of Ti(O/‘Pr)4 (1.89 ml, 6.3 mmol). After stirring at 0 °C for 1 h, 1 M TiCI (6.3 ml, 6.3 mmol) was added. The reaction mixture was stirred at rt for 45 min and concentrated to dryness under reduced pressure. The residue was dissolved in CH3OH (10 ml) and NaBH3CN (510 mg, 8 mmol) was added. The reaction mixture was stirred overnight at rt. The reaction mixture was poured to 1 N NaOH (100 ml) and extracted with EtOAc (3x 100 ml). The organic layer was combined and dried with NaHC03. Removal of solvent and separation by PTLC (5% 2 M NH3 in CH3OH/ CH2CI2) afforded β-13 (spot 1 , 30 mg, 6%) and α-13 (spot 2, 98 mg, 20%). β-13: 1HNMR (CDCI3): δ 1.50-1.38 (m, 5H), 1.42 (d, J= 6 Hz, 3H), 1.51-1.75 (m, 5H), 1.84 (m, 2H), 2.38 (m, 1 H), 2.45 (m, 1 H), 3.38 (br s, 1 H), 4.78 (m, 1 H), 6.59 (m, 2H), 7.15 (m, 1 H), 7.26 (m, 2H), 7.36 (m, 1 H), 7.42 (m, 1 H), 7.82 (m, 1 H), 8.77 (d, J= 2 Hz, 1 H). α-13:1HNMR (CDCI3): δ 0.95 (m, 2H), 1.02-1.35 (m, 6H), 1.41 (d, J= 6 Hz, 3H), 1.82-1.95 (m, 4H), 2.37 (m; 2H), 2.69 (m, 2H), 4.71 (m, 1 H), 6.71 (m, 2H), 7.11 (m, 1 H), 7.25 (m, 2H), 7.38 (m, 1 H), 7.42 (m, 1 H), 7.80 (m, 1 H), 8.76 (d, J= 1.6 Hz, 1 H). Step 7:
Compound α-13 (300 mg, 0.71 mmol) was dissolved in CH2CI2 (10 ml) followed by addition of Et3N (0.9 ml). The reaction mixture was C(O)Oled to 0 °C and ethyl chloroformate (0.5 ml) was added. The reaction mixture was stirred at rt for 1 h. The reaction mixture was directly separated by preparative TLC (EtOAc/ hexane, 1 :1) to give the title compound (14) VORAPAXAR (300 mg, 86%). MS m/z 493 (M+1).
HRMS Calcd for C29H34N2O4F (M+1 ): 493.2503, found 493.2509.
…………………
SYNTHESIS 1
http://www.google.com/patents/WO2006076564A1
VORAPAXAR= COMPD A
Example 6 – Preparation of Compound A
To a three-neck flask equipped with an agitator, thermometer and nitrogen inertion was added 7A (13.0 g), THF (30 mL). The mixture was cooled to below -200C after which lithium diisopropylamide (2M, 20 mL) was slowly added. The reaction mixture was agitated for an additional hour (Solution A). To another flask was added 6 (10.0 g) and THF (75 mL) . The mixture was stirred for about 30 minutes and then slowly transferred into the solution A while maintaining the temperature below 200C. The mixture was stirred at below -200C for an additional hour before quenching the reaction by adding 20 mL of water. The reaction mixture was warmed to 00C and the pH was adjusted to about 7 by addition of 25% HaSO4 (11 mL). The mixture was further warmed to 200C and then diluted with 100 mL of ethyl acetate and 70 mL of water. The two phases that had formed were separated and the aqueous layer was extracted with 50 mL of ethyl acetate. The solvents THF and ethyl acetate were then replaced with ethanol, and the Compound A was precipitated out as a crystalline solid from ethanol with seeding at 35 to 4O0C. After cooling to O0C, the suspension was stirred for an additional hour and then the product was filtered and washed with cold ethanol. The product was dried at 50 – 600C under vacuum to provide an off-white solid. VORAPAXAR
Yield: 12.7 g, (90%). m.p. 104.90C (DSC onset point).
1H NMR (CDCl3) δ 8.88 (d, J = 2.4 Hz, IH), 8.10 (dd, J = 8.2, 2.4 Hz, IH), 7.64 (IH), 7.61 (d, J = 8.8 Hz, IH), 7.55 (m, J = 8.2, 6.2 Hz, IH), 7.51 (d, J = 8.0 Hz, IH), 7.25 (dt, J = 9.0, 2.3 Hz, IH), 7.08 (d, J = 8.0 Hz, IH), 6.68 (dd, J = 15.4, 9.4 Hz, IH), 6.58 (d, J = 9.6 Hz, IH), 4.85 (dd, J = 14.2, 7.2 Hz, IH), 3.95 (dd, J = 14.2, 7.1 Hz, 2H), 3.29 (m, IH), 2.66 (m, J = 12.0, 6.4 Hz, IH), 2.33 (m, 2H), 1.76 (m, 4H), 1.30 (d, J = 5.6 Hz, 3H), 1.19 (m, 4H), 1.14 (t, J = 7.2 Hz, 3H), 0.98 (m, IH), 0.84 (m, IH). MS (EI) m/z: calcd. 492, found 492.
BISULPHATE SALT
Example 7 – Preparation of an Acid Salt (bisulfate) of Compound A:
Compound IA (5 g) was dissolved in about 25 mL of acetonitrile.
The solution was agitated for about 10 minutes and then heated to about 50 0C. About 6 mL of 2M sulfuric acid in acetonitrile was added into the heated reaction mixture. The solid salt of Compound A precipitated out during the addition of sulfuric acid in acetonitrile. After addition of sulfuric acid solution, the reaction mixture was agitated for 1 hour before cooling to room temperature. The precipitated solid was filtered and washed with about 30 mL of acetonitrile. The wet solid was dried under vacuum at room temperature for 1 hour and at 80 0C for about 12 hours to provide about 5 g white solid (yield 85%). m.p. 217.0 0C. 1H NMR (DMSO) 9.04 (s, IH), 8.60 (d, J = 8.1 Hz, IH), 8.10 (d, J = 8.2 Hz, IH), 7.76 (d, J = 10.4, IH), 7.71 (d, J = 7.8 Hz, IH), 7.60 (dd, J = 8.4, 1.8 Hz, IH), 7.34 (dd, 8.4, 1.8 Hz, IH), 7.08 (d, J = 8.0 Hz, IH), 7.02 (m, IH), 6.69 (d, J = 15.8 Hz, IH), 4.82 (m, IH), 3.94 (dd, J = 14.0, 7.0 Hz, 2H), 3.35 (brs, IH), 2.68 (m, IH), 2.38 (m, 2H), 1.80-1.70 (m, 4H), 1.27 (d, J = 5.8 Hz, 3H), 1.21 (m, 2H), 1.13 (t, J = 7.0 Hz, 3H), 0.95 (m, IH, 0.85 (m, IH). MS (EI) m/z calcd. 590, found 492.
INTERMEDIATE 6
Example 5- Preparation of Compound 6
To a three-neck flask equipped with an agitator, thermometer and nitrogen inert were added the crude product solution of Compound 5 (containing about 31 g. of Compound 5 in 300 mL solution) and anhydrous DMF (0.05 mL). After the mixture was agitated for 5 minutes, oxalyl chloride (12.2 mL) was added slowly while maintaining the batch temperature between 15 and 25°C. The reaction mixture was agitated for about an hour after the addition and checked by NMR for completion of reaction. After the reaction was judged complete, the mixture was concentrated under vacuum to 135 mL while maintaining the temperature of the reaction mixture below 300C. The excess oxalyl chloride was removed completely by two cycles of vacuum concentration at below 500C with replenishment of toluene (315 mL) each time, resulting in a final volume of 68 mL. The reaction mixture was then cooled to 15 to 25°C, after which THF (160 mL) and 2,6-lutidine (22 mL) were added. The mixture was agitated for 16 hours at 20 to 25°C under 100 psi hydrogen in the presence of dry 5% Pd/C (9.0 g). After the reaction was judged complete, the reaction mixture was filtered through celite to remove catalyst. More THF was added to rinse the hydrogenator and catalyst, and the reaction mixture was again filtered through celite. Combined filtrates were concentrated under vacuum at below 25°C to 315 mL. MTBE (158 mL) and 10% aqueous solution of phosphoric acid (158 mL) were added for a thorough extraction at 100C to remove 2,6- lutidine. Then phosphoric acid was removed by extracting the organic layer with very dilute aqueous sodium bicarbonate solution (about 2%), which was followed by a washing with dilute brine. The organic solution was concentrated atmospherically to a volume of 90 mL for solvent replacement. IPA (315 mL) was added to the concentrated crude product solution. The remaining residual solvent was purged to <_ 0.5% of THF (by GC) by repeated concentration under vacuum to 68 mL, with replenishment of IPA (315 mL) before each concentration. The concentrated (68 mL) IPA solution was heated to 50°C, to initiate crystallization. To this mixture n-heptane (68 mL) was added very slowly while maintaining the batch temperature at 50°C. The crystallizing mixture was cooled very slowly over 2.5 hours to 25°C. Additional n- heptane (34 mL) was added very slowly into the suspension mixture at 250C. The mixture was further cooled to 200C, and aged at that temperature for about 20 hours. The solid was filtered and washed with a solvent mixture of 25% IPA in n-heptane, and then dried to provide
19.5 g of a beige colored solid of Compound 6. (Yield: 66%) m.p. 169.30C. IH NMR (CD3CN) δ 9.74 (d, J = 3.03 Hz, IH), 5.42 (br, IH), 4.69 (m, IH), 4.03 (q, J = 7.02 Hz, 2H), 3.43 (qt, J = 3.80, 7.84 Hz, IH), 2.67 (m, 2H), 2.50 (dt, J = 3.00, 8.52 Hz, IH), 1.93 (d, J = 12.0 Hz, 2H), 1.82 (dt, J = 3.28, 9.75 Hz, 2H), 1.54 (qd, J = 3.00, 10.5 Hz, IH), 1.27 (d, J = 5.97 Hz, 3H), 1.20 (m, 6H), 1.03 – 0.92 (m, 2H). MS (ESI) m/z (M++1): calcd. 324, found 324.
INTERMEDIATE 7A
Example 4 – Preparation of Compound 7A
+ 1-Pr2NLi + (EtO)2POCI – + LiCI
7A
To a 10 L three-necked round bottomed flask equipped with an agitator, thermometer and a nitrogen inlet tube, was added 20Og of
Compound 8 (1.07 mol, from Synergetica, Philadelphia, Pennsylvania). THF (1000 mL) was added to dissolve Compound 8. After the solution was cooled to -80 0C to -50 0C, 2.0 M LDA in hexane/THF(1175 mL, 2.2 eq) was added while maintaining the batch temperature below -50 0C. After about 15 minutes of agitation at -800C to -50 0C, diethyl chlorophosphate (185 mL, 1.2 eq) was added while maintaining the batch temperature below -50 0C. The mixture was agitated at a temperature from -800C to – 50 0C for about 15 minutes and diluted with n-heptane (1000 mL). This mixture was warmed up to about -35 0C and quenched with aqueous ammonium chloride (400 g in 1400 mL water) at a temperature below -10 0C. This mixture was agitated at -150C to -10 0C for about 15 minutes followed by agitation at 150C to 25 0C for about 15 minutes. The aqueous layer was split and extracted with toluene (400 mL). The combined organic layers were extracted with 2N hydrochloric acid (700 mL) twice. The product-containing hydrochloric acid layers were combined and added slowly to a mixture of toluene (1200 mL) and aqueous potassium carbonate (300 g in 800 mL water) at a temperature below 30 0C. The aqueous layer was extracted with toluene (1200 mL). The organic layers were combined and concentrated under vacuum to about 600 ml and filtered to remove inorganic salts. To the filtrate was added n-heptane (1000 ml) at about 55 0C. The mixture was cooled slowly to 40 0C, seeded, and cooled further slowly to -10 0C. The resulting slurry was aged at about -10 0C for 1 h, filtered, washed with n- heptane, and dried under vacuum to give a light brown solid (294 g, 85% yield), m.p. 52 0C (DSC onset point).1H NMR (CDCl3) δ 8.73 (d, J = 1.5 Hz, IH), 7.85 (dd, Ji = 8.0 Hz, J2 = 1.5 Hz, IH), 7.49 (dd, Ji = 8.0 Hz, J2 = 1.3 Hz, IH), 7.42 (m, IH), 7.32 (d, J = 7.8 Hz, IH), 7.24 (m, IH), 7.08 (dt, Ji = 8.3 Hz, J2 = 2.3 Hz, IH), 4.09 (m, 4H), 3.48 (d, J = 22.0 Hz, 2H), 1.27 (t, J = 7.0 Hz, 6H). MS (ESI) for M+H calcd. 324, found 324.
Example 3 – Preparation of Compound 5:
4 5
To a three-necked round bottomed flask equipped with an agitator, thermometer and a nitrogen inlet tube was added a solution of Compound 4 in aqueous ethanol (100 g active in 2870 ml). The solution was concentrated to about 700 ml under reduced pressure at 350C to 40°C to remove ethyl alcohol. The resultant homogeneous mixture was cooled to 200C to 300C and its pH was adjusted to range from 12 to 13 with 250 ml of 25% sodium hydroxide solution while maintaining the temperature at 20-300C. Then 82 ml of ethyl chloroformate was slowly added to the batch over a period of 1 hour while maintaining the batch temperature from 200C to 300C and aged for an additional 30 minutes. After the reaction was judged complete, the batch was acidified to pH 7 to 8 with 10 ml of concentrated hydrochloric acid (37%) and 750 ml of ethyl acetate. The pH of the reaction mixture was further adjusted to pH 2 to 3 with 35% aqueous hydrochloric acid solution. The organic layer was separated and the aqueous layer was extracted again with 750 ml of ethyl acetate. The combined organic layers were washed twice with water (200 ml) . Compound 5 was isolated from the organic layer by crystallization from ethyl acetate and heptane mixture (1: 1 mixture, 1500 ml) at about 700C to 80 0C. The solid was filtered at 500C to 60 °C, washed with heptane and then dried to provide an off-white solid (yield 50%). m.p. 197.7°C. 1HNMR (CD3CN) δ 5.31 (brs, IH), 4.67 (dt, J = 16.1, 5.9 Hz, IH), 4.03 (q, J = 7.1 Hz, 2H), 3.41 (m, IH), 2.55 – 2.70 (m, 2H), 1.87 – 1.92 (m, IH), 1.32 – 1.42 (m, IH), 1.30 (d, J = 5.92 Hz, 3H), 1.30 – 1.25 (m, 6H), 0.98 (qt, J = 15.7, 3.18 Hz, 2H). MS (ESI) M+l m/z calculated 340, found 340.
Example 2 – Preparation of Compound 4;
3 4
7.4 kg of ammonium formate was dissolved in 9L of water at 15- 250C, and then cooled to 0-100C. 8.9 kg of Compound 3 was charged at 0-150C followed by an addition of 89L of 2B ethyl alcohol. The batch was cooled to 0-50C 0.9 kg of 10% Palladium on carbon (50% wet) and 9 L of water were charged. The batch was then warmed to 18-280C and agitated for 5 hours, while maintaining the temperature between 18-28 0C. After the reaction was judged complete, 7 IL of water was charged. The batch was filtered and the wet catalyst cake was then washed with 8OL of water. The pH of the filtrate was adjusted to 1-2 with 4N aqueous hydrochloric acid solution. The solution was used in the next process step without further isolation. The yield is typically quantiative. m.p. 216.40C. IH NMR (D2O+1 drop HCl) δ 3.15 (m, IH), 2.76 (m, IH), 2.62 (m, IH), 2.48 (dd,J-5.75Hz, IH), 1.94 (m, 2H), 1.78 (m, 2H), 1.38 (m, 2H), 1.20 (m, 6H), 1.18 (m, IH), 0.98 (q,J=2.99Hz, IH).
Example 1 – Preparation of Compound 3
2B 3
To a reactor equipped with an agitator, thermometer and nitrogen, were added about 10.5 kg of 2B, 68 L of acetone and 68 L of IN aqueous hydrochloric acid solution. The mixture was heated to a temperature between 50 and 600C and agitated for about 1 hour before cooling to room temperature. After the reaction was judged complete, the solution was concentrated under reduced pressure to about 42 L and then cooled to a temperature between 0 and 50C. The cooled mixture was agitated for an additional hour. The product 3 was filtered, washed with cooled water and dried to provide an off-white solid (6.9 kg, yield 76%). m.p. 2510C. Η NMR (DMSO) δ 12.8 (s, IH), 4.72 (m, J = 5.90 Hz, IH), 2.58 (m, 2H), 2.40 (m, J = 6.03 Hz, 2H), 2.21 (dd, J = 19.0, 12.8 Hz, 3H), 2.05 (m, IH), 1.87 (q, J = 8.92 Hz, IH), 1.75 (m, IH), 1.55 (m, IH), 1.35 (q, J = 12.6 Hz, IH), 1.27 (d, J = 5.88 Hz, 3H). MS (ESI) M+l m/z calcd. 267, found 267.
NOTE
Compound 7A may be prepared from Compound 8 by treating Compound 8 with diethylchlorophosphate:
Compound 8 may be obtained by the process described by Kyoku, Kagehira et al in “Preparation of (haloaryl)pyridines,” (API Corporation, Japan). Jpn. Kokai Tokkyo Koho (2004). 13pp. CODEN: JKXXAF JP
2004182713 A2 20040702. Compound 8 is subsequently reacted with a phosphate ester, such as a dialkyl halophosphate, to yield Compound 7A. Diethylchlorophosphate is preferred. The reaction is preferably conducted in the presence of a base, such as a dialkylithium amide, for example diisopropyl lithium amide.
…………………………………..
J Med Chem 2008, 51(11): 3061
http://pubs.acs.org/doi/abs/10.1021/jm800180e
The discovery of an exceptionally potent series of thrombin receptor (PAR-1) antagonists based on the natural product himbacine is described. Optimization of this series has led to the discovery of 4 (SCH 530348), a potent, oral antiplatelet agent that is currently undergoing Phase-III clinical trials for acute coronary syndrome (unstable angina/non-ST segment elevation myocardial infarction) and secondary prevention of cardiovascular events in high-risk patients.
Ethyl [(3aR,4aR,8aR,9aS)-9(S)-[(E)-2-[5-(3-fluorophenyl)-2-
pyridinyl]ethenyl]dodecahydro-1(R)-methyl-3-oxonaphtho[2,3-c]furan-6(R)-yl]carbamate (4).
4 (300 mg, 86%). MS m/z 493 (M+1).
HRMS Calcd for C29H34N2O4F
(M+1): 493.2503, found 493.2509; mp125 °C;
[]D20 6.6 (c 0.5, MeOH).
1HNMR (CDCl3):
http://pubs.acs.org/doi/suppl/10.1021/jm800180e/suppl_file/jm800180e-file002.pdf
0.88-1.18 (m, 5 H), 1.22-1.30 (m, 3 H), 1.43 (d, J = 5.85 Hz, 3 H), 1.88-2.10 (m, 4 H), 2.33-2.42 (m, 2 H),
2.75-2.67 (m, 1 H), 3.52-3.60 (m, 1 H), 4.06-4.14 (m, 2 H), 4.54-4.80 (m, 1 H), 4.71-4.77 (m, 1 H),
6.55-6.63 (m, 2 H), 7.07-7.12 (m, 1 H), 7.26-7.29 (m, 2 H), 7.34 (d, J = 8.05 Hz, 1 H), 7.41-7.46 (m, 1 H), 7.80-7.82 (m, 1 H), 8.76-8.71 (m, 1 H).
……………………..
References
- Samuel Chackalamannil; Wang, Yuguang; Greenlee, William J.; Hu, Zhiyong; Xia, Yan; Ahn, Ho-Sam; Boykow, George; Hsieh, Yunsheng et al. (2008). “Discovery of a Novel, Orally Active Himbacine-Based Thrombin Receptor Antagonist (SCH 530348) with Potent Antiplatelet Activity”. Journal of Medicinal Chemistry 51 (11): 3061–4.doi:10.1021/jm800180e. PMID 18447380.
- Merck Blood Thinner Studies Halted in Select Patients, Bloomberg News, January 13, 2011
- Tricoci et al. (2012). “Thrombin-Receptor Antagonist Vorapaxar in Acute Coronary Syndromes”. New England Journal of Medicine 366 (1): 20–33.doi:10.1056/NEJMoa1109719. PMID 22077816.
- Morrow, DA; Braunwald, E; Bonaca, MP; Ameriso, SF; Dalby, AJ; Fish, MP; Fox, KA; Lipka, LJ; Liu, X; Nicolau, JC; Ophuis, AJ; Paolasso, E; Scirica, BM; Spinar, J; Theroux, P; Wiviott, SD; Strony, J; Murphy, SA; TRA 2P–TIMI 50 Steering Committee and, Investigators (Apr 12, 2012). “Vorapaxar in the secondary prevention of atherothrombotic events.”. The New England Journal of Medicine 366 (15): 1404–13. doi:10.1056/NEJMoa1200933.PMID 22443427.
- “Merck Statement on FDA Advisory Committee for Vorapaxar, Merck’s Investigational Antiplatelet Medicine”. Merck. Retrieved 16 January 2014.
- http://www.forbes.com/sites/larryhusten/2014/01/15/fda-advisory-panel-votes-in-favor-of-approval-for-mercks-vorapaxar/
- SCH-530348 (Vorapaxar) is an investigational candidate for the prevention of arterial thrombosis in patients with acute coronary syndrome and peripheral arterial disease. “Convergent Synthesis of Both Enantiomers of 4-Hydroxypent-2-ynoic Acid Diphenylamide for a Thrombin Receptor Antagonist Sch530348 and Himbacine Analogues.” Alex Zaks et al.: Adv. Synth. Catal. 2009, 351: 2351-2357 Full text;
- Discovery of a novel, orally active himbacine-based thrombin receptor antagonist (SCH 530348) with potent antiplatelet activity
J Med Chem 2008, 51(11): 3061
- Stu Borman (2005). “Hopes Ride on Drug Candidates: Researchers reveal potential new medicines for thrombosis, anxiety, diabetes, and cancer”. Chemical & Engineering News 83 (16): 40–44.
PATENTS
- WO 2003089428
- WO 2006076452
- US 6063847
- WO 2006076565
- WO 2008005344
- WO2010/141525
- WO2008/5353
- US2008/26050
- WO2006/76564 mp, nmr
3-21-2012
|
EXO-SELECTIVE SYNTHESIS OF HIMBACINE ANALOGS
|
|
10-14-2011
|
EXO- AND DIASTEREO- SELECTIVE SYNTHESIS OF HIMBACINE ANALOGS
|
|
8-3-2011
|
Exo- and diastereo-selective syntheses of himbacine analogs
|
|
3-18-2011
|
COMBINATION THERAPIES COMPRISING PAR1 ANTAGONISTS WITH NAR AGONISTS
|
|
8-11-2010
|
Exo-selective synthesis of himbacine analogs
|
|
6-4-2010
|
SYNTHESIS Of DIETHYLPHOSPHONATE
|
|
5-12-2010
|
THROMBIN RECEPTOR ANTAGONISTS
|
|
3-31-2010
|
Synthesis of diethyl{[5-(3-fluorophenyl)-pyridine-2yl]methyl}phosphonate
|
|
12-4-2009
|
Local Delivery of PAR-1 Antagonists to Treat Vascular Complications
|
|
12-2-2009
|
SYNTHESIS OF HIMBACINE ANALOGS
|
10-21-2009
|
Exo- and diastereo- selective syntheses of himbacine analogs
|
|
6-31-2009
|
Synthesis of 3-(5-nitrocyclohex-1-enyl) acrylic acid and esters thereof
|
|
6-3-2009
|
Synthesis of himbacine analogs
|
|
1-23-2009
|
METHODS AND COMPOSITIONS FOR TREATING CARDIAC DYSFUNCTIONS
|
|
9-26-2008
|
REDUCTION OF ADVERSE EVENTS AFTER PERCUTANEOUS INTERVENTION BY USE OF A THROMBIN RECEPTOR ANTAGONIST
|
|
2-8-2008
|
IMMEDIATE-RELEASE TABLET FORMULATIONS OF A THROMBIN RECEPTOR ANTAGONIST
|
|
1-32-2008
|
SOLID DOSE FORMULATIONS OF A THROMBIN RECEPTOR ANTAGONIST
|
|
12-5-2007
|
Thrombin receptor antagonists
|
|
11-23-2007
|
THROMBIN RECEPTOR ANTAGONISTS
|
|
8-31-2007
|
THROMBIN RECEPTOR ANTAGONISTS AS PROPHYLAXIS TO COMPLICATIONS FROM CARDIOPULMONARY SURGERY
|
8-31-2007
|
CRYSTALLINE POLYMORPH OF A BISULFATE SALT OF A THROMBIN RECEPTOR ANTAGONIST
|
|
6-27-2007
|
Crystalline polymorph of a bisulfate salt of a thrombin receptor antagonist
|
|
8-4-2006
|
Preparation of chiral propargylic alcohol and ester intermediates of himbacine analogs
|
|
9-31-2004
|
Methods of use of thrombin receptor antagonists
|
US6063847 * | Nov 23, 1998 | May 16, 2000 | Schering Corporation | Thrombin receptor antagonists |
US6326380 * | Apr 7, 2000 | Dec 4, 2001 | Schering Corporation | Thrombin receptor antagonists |
US20030216437 * | Apr 14, 2003 | Nov 20, 2003 | Schering Corporation | Thrombin receptor antagonists |
US20040176418 * | Jan 9, 2004 | Sep 9, 2004 | Schering Corporation | Crystalline polymorph of a bisulfate salt of a thrombin receptor antagonist |
WO2011128420A1 | Apr 14, 2011 | Oct 20, 2011 | Sanofi | Pyridyl-vinyl pyrazoloquinolines as par1 inhibitors |
DAPAGLIFLOZIN…FDA approves AZ diabetes drug Farxiga
DAPAGLIFLOZIN, BMS-512148
The US Food and Drug Administration has finally approved AstraZeneca’s diabetes drug Farxiga but is insisting on six post-marketing studies, including a cardiovascular outcomes trial.
The approval was expected given that the agency’s Endocrinologic and Metabolic Drugs Advisory Committee voted 13-1 last month that the benefits of Farxiga (dapagliflozin), already marketed in Europe as Forxiga, outweigh identified risks. The FDA rejected the drug in January 2012 due to concerns about possible liver damage and the potential link with breast and bladder cancer.

AVOSENTAN
AVOSENTAN
N-[6-Methoxy-5-(2-methoxyphenoxy)-2-(4-pyridyl)pyrimidin-4-yl]-5-methylpyridine-2-sulfonamide
5-methyl-pyridine-2-sulfonic acid [6-methoxy-5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)-pyrimidin-4-yl]-amide,
5-methyl-pyridine-2-sulfonic acid [6-methoxy-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide,
Endothelin ETA Receptor Antagonists
M.Wt: 479.51
Formula: C23H21N5O5S
Roche (Originator)
CAS No.: 290815-26-8
- RO 67-0565
- SPP 301
- UNII-L94KSX715K
PHASE 3
CLINICAL TRIALS
http://clinicaltrials.gov/search/intervention=spp301+OR+Avosentan
SPP-301 is an oral, once-daily, second-generation endothelin ETA receptor antagonist which had been in phase III clinical development at Speedel for the treatment of diabetic nephropathy. In December 2006, the company reported that the phase III trial had been stopped based on the recommendation from the trial’s Data Safety Monitoring Board (DSMB) to stop the trial following incidence of a significant imbalance in fluid retention in patients in the study arms. Speedel reported that the compound will be evaluated for potential new clinical development for the treatment of diabetic kidney disease and other indications.
Originally developed by Roche and specifically optimized for improved liver safety, SPP-301 was licensed to Speedel in October 2000. In 2003, Speedel exercised its option to license from Roche all rights to SPP-301, including exclusive worldwide rights for the full development and commercialization of the ETA antagonist. SPP-301 has fast track designation and has undergone a special protocol assessment (SPA) by the FDA. Speedel had been studying the drug for the treatment of hypertension.
AVOSENTAN
290815-26-8 CAS
PATENTS
2. WO 2004078104
3. WO 2005113543
4. WO 2007031501
5. WO 2008077916
Dutzler R, Ernstb B, Hediger MA, Keppler D, Mohr P, Neidhart W, Märki HP.Chimia (Aarau). 2010;64(9):662-6.
………………………
INTRODUCTION
-
Own investigations have shown that there exist two distinct crystalline forms, hereinafter referred to as form A and form B, as well as a number of further solvates, in particular the methanol, ethanol, isopropanol, dichloromethane, acetone, methyl ethyl ketone and tetrahydrofuran solvates.
-
It was further surprisingly found that the thermodynamically stable crystalline form – form B – can be prepared under controlled conditions and that said form B can be prepared with a reliable method in an industrial scale, which is easy to handle and to process in the manufacture and preparation of formulations.
………………..
4,6-Dichloro-5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)-pyrimidine (described in EP 0 799 209) can be transformed to the intermediate of formula (III)—according to scheme 1—on reaction with an appropriate sulfonamide of formula (II), wherein R1 is as defined in claim 1, in a suited solvent such as DMSO or DMF at room temperature or at elevated temperature and in the presence of a suited base such as potassium carbonate.
EXAMPLE 1
[0064] a) To a solution of 6.9 g sodium in MeOH (300 ml) were added 14.52 g of 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide at RT and the mixture was refluxed for 5 days until completion of the reaction according to TLC analysis. The reaction mixture was concentrated in vacuo to half its volume upon which the crude reaction product precipitated as a sodium salt. It was filtered off by suction and dried in a high vacuum. The solid was dissolved in water, which was then made acidic by addition of acetic acid. The precipitating free sulfonamide was extracted into Me2Cl2. The organic layer was dried over Mg2SO4, concentrated on a rotary evaporator, and the crystalline solid that had formed was filtered off. It was then dried in a high vacuum for 12 h at 120° C. to give the desired 5-methyl-pyridine-2-sulfonic acid [6-methoxy-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide as white crystals. Melting point 225-226° C. ISN mass spectrum, m/e 478.2 (M-1 calculated for C23H21N5O5S1: 478).
[0065] C23H21N5O5S1: Calc: C 57.61; H 4.41; N 14.61; S 6.69. Found: C 57.56; H 4.38; N 14.61; S 6.83
[0066] Preparation of the starting material:
[0067] b) 11.3 g of 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)-pyrimidine and 19.66 g of 5-methylpyridyl-2-sulfonamide potassium salt (preparations described in EP 0 799 209) were dissolved in DMF (255 ml) under argon. The solution was stirred for 2 h at 40° C. until completion of the reaction according to TLC analysis. The reaction mixture was cooled to RT and the solvent removed in a high vacuum. The residue was suspended in water (850 ml), acetic acid (85 ml) was added and the mixture was stirred for 30 minutes at RT. The solid that precipitated was collected by filtration and dried in a high vacuum at 60° C. for 16 h to give 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide ( CHLORO STARTING MATERIAL) as yellow crystals. Melting point 177-179° C. ISN mass spectrum, m/e 482.2 (M-1 calculated for C22H18ClN5O5S1: 482).
……………………………….
http://www.google.com/patents/US6417360
EXAMPLE 1
a) To a solution of 6.9 g sodium in MeOH (300 ml) were added 14.52 g of 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide at RT and the mixture was refluxed for 5 days until completion of the reaction according to TLC analysis. The reaction mixture was concentrated in vacuo to half its volume upon which the crude reaction product precipitated as a sodium salt. It was filtered off by suction and dried in a high vacuum. The solid was dissolved in water, which was then made acidic by addition of acetic acid. The precipitating free sulfonamide was extracted into Me2Cl2. The organic layer was dried over Mg2SO4, concentrated on a rotary evaporator, and the crystalline solid that had formed was filtered off. It was then dried in a high vacuum for 12 h at 120° C. to give the desired 5-methyl-pyridine-2-sulfonic acid [6-methoxy-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide as white crystals. Melting point 225-226° C. ISN mass spectrum, m/e 478.2 (M-1 calculated for C23H21N5O5S1: 478).
C23H21N5O5S1: Calc: C 57.61; H 4.41; N 14.61; S 6.69. Found: C 57.56; H 4.38; N 14.61; S 6.83
Preparation of the starting material:
b) 11.3 g of 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)-pyrimidine and 19.66 g of 5-methylpyridyl-2-sulfonamide potassium salt (preparations described in EP 0 799 209) were dissolved in DMF (255 ml) under argon. The solution was stirred for 2 h at 40° C. until completion of the reaction according to TLC analysis. The reaction mixture was cooled to RT and the solvent removed in a high vacuum. The residue was suspended in water (850 ml), acetic acid (85 ml) was added and the mixture was stirred for 30 minutes at RT. The solid that precipitated was collected by filtration and dried in a high vacuum at 60° C. for 16 h to give 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl]-amide as yellow crystals. Melting point 177-179° C. ISN mass spectrum, m/e 482.2 (M-1 calculated for C22H18ClN5O5S1: 482).
…………………….
http://www.google.com/patents/EP0799209B1
SYNTHESIS OF
4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)-pyrimidine
A BASIC STARTING MATERIAL FOR AVOSENTAN
- Preparation of the starting material
-
- b) 53.1 g of 4-cyano-pyridine (98%) are added all at once to a solution of 1.15 g of sodium in 200 ml of abs. MeOH. After 6 hours 29.5 g of NH4Cl are added while stirring vigorously. The mixture is stirred at room temperature overnight. 600 ml of ether are added thereto, whereupon the precipitate is filtered off under suction and thereafter dried at 50°C under reduced pressure. There is thus obtained 4-amidino-pyridine hydrochloride (decomposition point 245-247°C).
- c) 112.9 g of diethyl (2-methoxyphenoxy)malonate are added dropwise within 30 minutes to a solution of 27.60 g of sodium in 400 ml of MeOH. Thereafter, 74.86 g of the amidine hydrochloride obtained in b) are added all at once. The mixture is stirred at room temperature overnight and evaporated at 50°C under reduced pressure. The residue is treated with 500 ml of ether and filtered off under suction. The filter cake is dissolved in 1000 ml of H2O and treated little by little with 50 ml of CH3COOH. The precipitate is filtered off under suction, washed with 400 ml of H2O and dried at 80°C under reduced pressure. There is thus obtained 5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)-pyrimidine-4,6-diol (or tautomer), melting point above 250°C.
- d) A suspension of 154.6 g of 5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)-pyrimidine-4,6-diol (or tautomer) in 280 ml of POCl3 is heated at 120°C in an oil bath for 24 hours while stirring vigorously. The reaction mixture changes gradually into a dark brown liquid which is evaporated under reduced pressure and thereafter taken up three times with 500 ml of toluene and evaporated. The residue is dissolved in 1000 ml of CH2Cl2, treated with ice and H2O and thereafter adjusted with 3N NaOH until the aqueous phase has pH 8. The organic phase is separated and the aqueous phase is extracted twice with CH2Cl2. The combined CH2Cl2 extracts are dried with MgSO4, evaporated to half of the volume, treated with 1000 ml of acetone and the CH2Cl2remaining is distilled off at normal pressure. After standing in a refrigerator for 2 hours the crystals are filtered off under suction and dried at 50°C overnight. There is thus obtained 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)-pyrimidine, melting point 178-180°C.
…………………………
http://www.google.com/patents/WO2000052007A1
Preparation of the starting material:
5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2- methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl] -amide IE THE 6 CHLORO COMPD
b) 11.3 g of 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)-pyrimidine and 1 .66 g of 5-methylpyridyl-2-sulfonamide potassium salt (preparations described in EP 0 799 209) were dissolved in DMF (255 ml) under argon. The solution was stirred for 2 h at 40°C until completion of the reaction according to TLC analysis. The reaction mixture was cooled to RT and the solvent removed in a high vacuum. The residue was suspended in water (850 ml), acetic acid (85 ml) was added and the mixture was stirred for 30 minutes at RT. The solid that precipitated was collected by filtration and dried in a high vacuum at 60 °C for 16 h to give 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2- methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl] -amide as yellow crystals. Melting point 177-179 °C. ISN mass spectrum, m/e 482.2 (M-l calculated for C22Hi8ClN5O5Sι: 482).
………………………………………………………………………………………….
NEXT
Example 1AVOSENTAN
a) To a solution of 6.9 g sodium in MeOH (300 ml) were added 14.52 g of
5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl- pyrimidin-4-yl] -amide at RT and the mixture was refluxed for 5 days until completion of the reaction according to TLC analysis. The reaction mixture was concentrated in vacuo to half its volume upon which the crude reaction product precipitated as a sodium salt. It was filtered off by suction and dried in a high vacuum. The solid was dissolved in water, which was then made acidic by addition of acetic acid. The precipitating free sulfonamide was extracted into Me2Cl2. The organic layer was dried over Mg SO , concentrated on a rotary evaporator, and the crystalline solid that had formed was filtered off. It was then dried in a high vacuum for 12 h at 120 °C to give the desired 5-methyl-pyridine-2-sulfonic acid [6- methoxy-5-(2-methoxy-phenoxy)-2-pyridin-4-yl-pyrimidin-4-yl] -amide as white crystals. Melting point 225-226 °C. ISN mass spectrum, m/e 478.2 (M-l calculated for
C23H21N5O5S1: Calc: C 57.61; H 4.41; N 14.61; S 6.69. Found: C 57.56; H 4.38; N 14.61; S 6.83
…………………………………………….
IS DESCRIBED IN
http://www.google.com/patents/EP2331513A1?cl=en
ALSO
-
Diabetic nephropathy is the principle cause of end stage renal disease in the western world. It is a major cause of morbidity and mortality in Type-I Diabetes, but is an increasing problem in Type-II Diabetes and because the incidence of this is five times that of Type-I Diabetes, it contributes at least 50% of diabetics with end stage renal disease.
-
The initial stage of subtle morphologic changes in the renal glomeruli is followed by microalbuminuria. This is associated with a modestly rising blood pressure and an increased incidence of cardiovascular disease. There follows a continued increase in urinary protein excretion and declining glomerular filtration rate. Diabetic nephropathy has many possible underlying pathophysiological causes including metabolic, glycosylation of proteins, haemodynamics, altered flow/pressure in glomeruli, the development of hypertension and cytokine production; all of these are associated with the development of extracellular matrix and increased vascular permeability leading to glomerular damage and proteinuria.
WO2005113543A1 * | May 12, 2005 | Dec 1, 2005 | Alexander Bilz | Crystalline forms of a pyridinyl-sulfonamide and their use as endothelin receptor antagonists |
WO2007031501A2 * | Sep 11, 2006 | Mar 22, 2007 | Speedel Pharma Ag | Pyridylsulfonamidyl-pyrimidines for the prevention of blood vessel graft failure |
WO2008077916A1 * | Dec 21, 2007 | Jul 3, 2008 | Ovidiu Baltatu | Pharmaceutical composition using aliskiren and avosentan |
EP1454625A1 * | Mar 6, 2003 | Sep 8, 2004 | Speedel Development AG | Pyridylsulfonamidyl-pyrimidines for the treatment of diabetic nephropathies |
EP1595880A1 * | May 13, 2004 | Nov 16, 2005 | Speedel Pharma AG | Crystalline forms of a pyridinyl-sulfonamide and their use as endothelin receptor antagonists |
EP1938812A1 * | Dec 22, 2006 | Jul 2, 2008 | Speedel Pharma AG | Pharmaceutical composition using aliskiren and avosentan |
US6951856 | Jul 10, 2001 | Oct 4, 2005 | Actelion Pharmaceuticals Ltd. | Arylethene-sulfonamides |
US7402587 | May 12, 2005 | Jul 22, 2008 | Speedel Pharma Ag | Crystalline forms of a pyridinyl-sulfonamide and their use as endothelin receptor antagonists |
WO1996019459A1 * | Dec 8, 1995 | Jun 27, 1996 | Volker Breu | Novel sulfonamides |
EP0713875A1 * | Nov 13, 1995 | May 29, 1996 | F. Hoffmann-La Roche AG | Sulfonamides |
EP0897914A1 * | Aug 10, 1998 | Feb 24, 1999 | F. Hoffmann-La Roche Ag | Process for the preparation of 2,5-disubstitued pyridines |
READ MORE ON SNTAN SERIES……http://medcheminternational.blogspot.in/p/sentan-series.html
NETUPITANT
NETUPITANT
- Ro 67-3189/000
- UNII-7732P08TIR
- Ro-67-3189
- Netupitant, an NK-1 antagonist is under development for the treatment of overactive bladder. HELSINN GROUP
CAS: 290297-26-6
290296-54-7 (di HCl)
U.S. Pat. Nos. 6,303,790, 6,531,597, 6,297,375 and 6,479,483, 6,719,996 and 6,593,472 to Hoffmann La Roche(originator).
IUPAC/Chemical name:
2-(3,5-bis(trifluoromethyl)phenyl)-N,2-dimethyl-N-(6-(4-methylpiperazin-1-yl)-4-(o-tolyl)pyridin-3-yl)propanamide
Chemical Formula: C30H32F6N4O
Exact Mass: 578.24803
Molecular Weight: 578.59
Elemental Analysis: C, 62.28; H, 5.57; F, 19.70; N, 9.68; O, 2.77
Netupitant is another selective NKi receptor antagonist under development by Helsinn Healthcare, having the formula 2-[3,5-bis(trifluoromethyl)phenyl]-N,2-dimethyl-N-[4-(2- methylphenyl)-6-(4-methylpiperazin- l-yl)pyridin-3-yl]propanamide, or Benzeneacetamide, N,a,a-trimethyl-N-[4-(2-methylphenyl)-6-(4-methyl-l-piperazinyl)-3-pyridinyl]-3,5- bis(trifluoromethyl)-, and the below chemical structure:
Netupitant is a tachykinin NK-1 antagonist which had been in phase III clinical trials at Helsinn for the prophylaxis of chemotherapy-induced nausea and vomiting and in phase II clinical studies for the treatment of overactive bladder. However, no recent development has been reported for this research.
NK-1 receptor antagonists work by blocking the action of neurokinin-1 (Substance P), a naturally-occurring neurotransmitter in the brain that causes emesis. Netupitant was originally developed at Roche. In June 2005, Helsinn and Roche signed a licensing agreement granting Helsinn worldwide rights to the drug candidate.
Methods of synthesizing and formulating netupitant and its prodrugs are described in U.S. Patent Nos. 6,297,375, 6,719,996 and 6,593,472 to Hoffmann La Roche.
Netupitant is a highly selective NK1 receptor antagonist, which is thought to work by blocking the action of substance P, an endogenous neurotransmitter contained in high concentrations in the vomiting center of the brainstem that can stimulate the vomiting reflex. Netupitant is currently under phase III trials.
Chemotherapy is one of the treatment options utilized by oncologists in treating different types of cancers. Nausea and vomiting are the most common side-effects experienced by cancer patients when administered with chemotherapy. Netupitant-palonosetron, which is currently in Phase III trials helps in preventing CINV. The blockage of P/NK1 receptors by Netupitant in the central nervous system inhibits the binding of endogenous tachykinin neuropeptide substance and this result in preventing the chemotherapy-induced nausea and vomiting. Moreover, Palonosetron helps in the blockage of serotonin at 5-hydroxytryptamine type 3 (5-HT3) receptors and it also helps in the chemotherapy-induced nausea and vomiting.
Netupitant-Palonosetron FDC is estimated to answer significant unmet needs of the CINV market post its launch that is expected to be commercialized in 2014, as it would overcome the problems associated with current treatment with 5-HT3 receptor antagonists. Similar to Emend, Netupitant-Palonosetron FDC would gain considerable patient pool after its estimated launch in 2014, and subsequently match the patient share of Aloxi by 2018. Netupitant-Palonosetron FDC sales are expected to reach an estimated USD 515.0 million USD by 2018. FDC combination of 5-HT3 receptor antagonist and neurokinin-1 (NK1) receptor antagonist have shown better efficacy results in Phase II clinical trials for CINV patients and would thus lead to high uptake due to shifting physician and patient preference pattern towards better treatment for CINV.
Neurokinin 1 receptor antagonists are being developed for the treatment of a number of physiological disorders associated with an excess or imbalance of tachykinin, in particular substance P. Examples of conditions in which substance P has been implicated include disorders of the central nervous system such as anxiety, depression and psychosis (WO 95/16679, WO 95/18124 and WO 95/23798).
The neurokinin-1 receptor antagonists are further useful for the treatment of motion sickness and for treatment induced vomiting. The New England Journal of Medicine, Vol. 340, No. 3 190-195, 1999 has been described the reduction of cisplatin-induced emesis by a selective neurokinin-l-receptor antagonist. US5,972,938 describes a method for treating a psychoimmunologic or a psychosomatic disorder by administration of a tachykinin receptor, such as NK-1 receptor antagonist.
With the development of the 5-HT3 antagonist in the early 1990s, there emerged new strategies in the medical community to better control nausea and vomiting caused by various medical procedures, including chemotherapy (CINV), surgery (PONV), and radiation therapy (RINV). When added to steroids such as dexamethasone, several 5-HT3 antagonists have been demonstrated to significantly improve the standard of life for patients undergoing emetogenic medical procedures. Examples of 5-HT3 antagonists include ondansetron, marketed by
GlaxoSmithKline, and palonosetron, developed by Helsinn Healthcare.
Netupitant is another selective NKi receptor antagonist under development by Helsinn Healthcare, having the formula 2-[3,5-bis(trifluoromethyl)phenyl]-N,2-dimethyl-N-[4-(2- methylphenyl)-6-(4-methylpiperazin- l-yl)pyridin-3-yl]propanamide, or Benzeneacetamide, N,a,a-trimethyl-N-[4-(2-methylphenyl)-6-(4-methyl-l-piperazinyl)-3-pyridinyl]-3,5- bis(trifluoromethyl)-, and the below chemical structure:
Methods of synthesizing and formulating netupitant and its prodrugs are described in U.S. Patent Nos. 6,297,375, 6,719,996 and 6,593,472 to Hoffmann La Roche.
Other representative NKi antagonists include ZD4974 (developed by AstraZeneca), CGP49823 (developed by Ciba-Geigy), Lanepitant and LY686017 (developed by Eli Lilly), FK888 (developed by Fujisawa), Vofopitant, Vestipitant and Orvepitant (developed by
GlaxoSmithKline), Befetupitant (developed by Hoffmann-La Roche), Rl 16031 (developed by Janssen), L-733060 and L-736281 (developed by Merck), TKA731, NKP608 and DNK333 (developed by Novartis), CP-96345, CP-99994, CP- 122721, CJ-17493, CJ-11974 and CJ-11972 (developed by Pfizer), RP67580 and Dapitant (developed by Rhone-Poulenc Rorer),
Nolpitantium and SSR240600 (developed by Sanofi-Aventis), SCH388714 and Rolapitant (developed by Schering-Plough), TAK637 (developed by Takeda), HSP117 (developed by Hisamitsu), KRP103 (developed by Kyorin Pharm) and SLV317 (developed by Solvay).
Chemical structures of the above-mentioned NKi antagonists are shown below and discussion of those compounds as well as other NKi antagonists is present in Expert Opin. Ther. Patents (2010) 20(8), pp 1019- 1045 by Huang et al.
………………………………………………
WO 2013057554
WO 2011061622
WO 2010119347
WO 2003006016
WO 2006002860///
WO 2002085458
US 2002091265…….
…………………………………………………..
http://pubs.acs.org/doi/full/10.1021/jo0523666
…………………………………………..
https://www.google.co.in/patents/US6297375
(2-(3,5-bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide) which has the formula Ib
and to pharmaceutically acceptable acid addition salts thereof.
The compound of formula Ib and its salts is also characterized by valuable therapeutic properties as a highly selective antagonist of the Neurokinin 1 (NK-1, substance P) The present compound of formula lb and its pharmaceutically acceptable salts can be prepared by methods known in the art, for example, by processes described below, which process comprises
a) reacting the compound of formula
with the compound of formula
to the compound of formula
EXAMPLE 14
2-(3,5-Bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide hydrochloride (1:2)
a) 1-Methyl-4-(5-nitro-pyridin-2-yl)-piperazine
To a solution of 20 g (126 mmol) of 2-chloro-5-nitropyridine in 200 ml tetrahydrofuran were added dropwise 35 ml (315 mmol) 1-methylpiperazine within 10 min. The reaction mixture was refluxed for additional 1.5 h. After cooling to room temperature, the solvent was removed in vacuo and the residue was re-dissolved in 200 ml ethyl acetate. The organic phase was washed with 200 ml 1 N sodium bicarbonate solution, dried (magnesium sulfate) and evaporated to give 27.9 g (quantitative) of the title compound as a yellow solid.
MS m/e (%):223 (M+H+, 100).
b)2,2-Dimethyl-N-[6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-propionamide
To a solution of 27.9 g (125.5 mmol) of 1-methyl-4-(5-nitro-pyridin-2-yl)-piperazine in 400 ml methanol were added 2.6 g of 10% of palladium on activated charcoal. The reaction mixture was hydrogenated (room temperature to ca. 45° C., 1 bar) until the theoretical amount of hydrogen was taken up (about 2 h). The catalyst was filtered off and was washed twice with 100 ml portions of methanol. The filtrate was evaporated in vacuo to give 28 g of a purple oil which consisted to ca. 90% of the desired aniline derivative according to analysis by thin layer chromatography.
This crude product was dissolved in a mixture of 400 ml tetrahydrofuran and 100 ml diethyl ether. After cooling to 0° C., 30 ml (215 mmol) of triethylamine were added in one portion. Stirring was continued while 26 g (215 mmol) of pivaloyl chloride were added dropwise within a period of 10 min. The ice bath was removed and the reaction mixture was stirred for 1 h at room temperature. Then, the solvent was removed in vacuo and the residue was suspended in 200 ml 1 N sodium bicarbonate solution. The product was extracted three times with 200 ml portions of dichloromethane, dried (sodium sulfate) and purified by flash chromatography to give 30 g (86%) of the title compound as pink crystals.
MS m/e (%):277 (M+H+, 100).
c) N-[4-Iodo-6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-2,2-dimethyl-propionamide
A solution of 30 g (108 mmol) 2,2-dimethyl-N-[6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-propionamide and 58 ml (380 mmol) N,N,N′,N′-tetramethylethylenediamine under argon in 650 ml tetrahydrofuran was cooled in a dry ice bath to −78° C. Within lh, 239 ml (380 mmol) of a 1.6 N n-butyllithium solution in hexane were added dropwise. The reaction mixture was allowed to warm up to −30° C. overnight. After cooling again to −78° C., 43.6 g (170 mmol) iodine dissolved in 60 ml tetrahydrofuran were added dropwise during 15 min. The dry ice bath was replaced by an ice bath and a solution of 90 g (363 mmol) sodium thiosulfate pentahydrate in 250 ml water were added within 10 min when the temperature of the reaction mixture had reached 0° C. Then, 1000 ml diethyl ether were added and the organic layer was separated. The aqueous layer was extracted twice with 500 ml dichloromethane and the combined organic layers were dried (magnesium sulfate) and evaporated. Flash chromatography gave 18.5 g (42%) of the title compound as a light brown oil which crystallized upon standing at room temperature.
MS m/e (%): 403 (M+H+, 100).
d) 2,2-Dimethyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-propionamide
A mixture of 54 g (134 mmol) N-[4-iodo-6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-2,2-dimethyl-propionamide, 420 ml toluene, 150 ml 2 N sodium carbonate solution, 4.63 g (3.9 mmol) tetrakis(triphenylphosphine)palladium(0) and 20.16 g (147 mmol) o-tolylboronic acid was heated under argon at 80° C. for 12 h. After cooling to room temperature, the aqueous phase was separated and washed twice with toluene. The combined organic layers were washed with 50 ml brine, dried (sodium sulfate), evaporated and dried in vacuo to yield 49 g (quantitative) of the title compound as a brown oil.
MS m/e (%): 367 (M+H+, 100).
e) 6-(4-Methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-ylamine
A suspension of 56 g (152 mmol) 2,2-dimethyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-propionamide in 1300 ml 3 N hydrochloric acid solution was heated to 90-95° C. overnight. The reaction mixture was cooled to room temperature, washed with three 500 ml portions diethyl ether and filtered over celite. The filtrate was diluted with 500 ml water and was adjusted to pH 7-8 by addition of 28% sodium hydroxide solution under ice cooling. The product was extracted with four 1000 ml portions of dichloromethane. The combined organic layers were washed with 500 ml brine, dried (magnesium sulfate) and evaporated to give 35 g (82%) of the title compound as a light brown oil.
MS m/e (%):283 (M+H+, 100).
f) Methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine
A solution of 35 g (124 mmol) 6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-ylamine in 270 ml trimethyl orthoformate and 8 drops trifluoroacetic acid was heated for 3 h at 130° C. The reaction mixture was evaporated and dried in vacuo for 30 min. The residual oil was dissolved in 100 ml tetrahydrofuran and was added dropwise under ice cooling to 9.4 g (248 mmol) lithium aluminum hydride in 300 ml tetrahydrofuran. The reaction mixture was stirred for lh at room temperature, cooled to 0° C. again and acidified (pH 1-2) by addition of 28% hydrochloric acid solution. After stirring for 5 min, 28% sodium hydroxide solution was added to reach pH 10. The solution was filtered over celite, evaporated and purified by flash chromatography to give 23.6 g (64%) of the title compound as a light brown oil.
MS m/e (%):297 (M+H+, 100).
g) 2-(3,5-Bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide
A solution of 20 g (67.5 mmol) methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine and 17.5 ml (101 mmol) N-ethyldiisopropylamine in 200 ml dichloromethane was cooled in an ice bath and a solution of 24 g (75 mmol)2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionyl chloride in 50 ml dichloromethane was added dropwise. The reaction mixture was warmed to 35-40° C. for 3 h, cooled to room temperature again and was stirred with 250 ml saturated sodium bicarbonate solution. The organic layer was separated and the aqueous phase was extracted with dichloromethane. The combined organic layers were dried (magnesium sulfate) and evaporated. The residue was purified by flash chromatography to give 31.6 g (81%) of the title compound as white crystals. M.p. 155-157° C.
MS m/e (%): 579 (M+H+, 100).
h)2-(3,5-Bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide hydrochloride (1:2)
To a solution of 31.6 g (54.6 mmol)2-(3,5-bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide in 250 ml diethyl ether were added under ice cooling 60 ml 3 N hydrochloric acid solution in diethyl ether. After stirring for 15 min at 0° C., the suspension was evaporated to dryness, re-suspended in 100 ml diethyl ether, filtered and dried in vacuo to give 34.8 g (98%) of the title compound as white crystals. M.p. 235-238° C.
MS m/e (%): 579 (M+H+, 100).
……………………………….
2-(3,5-bis(trifluoromethyl)phenyl)-N,2-dimethyl-N-(6-(4-methylpiperazin-1-yl)-4-(o-tolyl)pyridin-3-yl)propanamide (Netupitant)
Other general procedures of preparing similar compounds to intermediate 1 of Scheme 1 are also disclosed in U.S. Pat. Nos. 6,303,790, 6,531,597, 6,297,375 and 6,479,483, the entirety of which are incorporated herein by reference.
Synthesis of methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine
Step 1:
13.0 g (82.5 mMol) 6-Chloro-nicotinic acid in 65 ml THF were cooled to 0° C. and 206.3 ml (206.3 mMol) o-tolylmagnesium chloride solution (1M in THF) were added over 45 minutes. The solution obtained was further stirred 3 hours at 0° C. and overnight at room temperature. It was cooled to −60° C. and 103.8 ml (1.8 Mol) acetic acid were added, followed by 35 ml THF and 44.24 g (165 mMol) manganese(III) acetate dihydrate. After 30 minutes at −60° C. and one hour at room temperature, the reaction mixture was filtered and THF removed under reduced pressure. The residue was partitioned between water and dichloromethane and extracted. The crude product was filtered on silica gel (eluent: ethyl acetate/toluene/formic acid 20:75:5) then partitioned between 200 ml aqueous half-saturated sodium carbonate solution and 100 ml dichloromethane. The organic phase was washed with 50 ml aqueous half-saturated sodium carbonate solution, The combined aqueous phases were acidified with 25 ml aqueous HCl 25% and extracted with dichloromethane. The organic extracts were dried (Na2SO4) and concentrated under reduced pressure to yield 10.4 g (51%) of 6-chloro-4-o-tolyl-nicotinic acid as a yellow foam. MS (ISN): 246 (M−H, 100), 202 (M-CO2H, 85), 166 (36).
Step 2:
To a solution of 8.0 g (32.3 mMol) 6-chloro-4-o-tolyl-nicotinic acid in 48.0 ml THF were added 3.1 ml (42.0 mMol) thionylchloride and 143 .mu.l (1.8 mMol) DMF. After 2 hours at 50° C., the reaction mixture was cooled to room temperature and added to a solution of 72.5 ml aqueous ammonium hydroxide 25% and 96 ml water cooled to 0″C. After 30 minutes at 0° C., THF was removed under reduced pressure and the aqueous layer was extracted with ethyl acetate. Removal of the solvent yielded 7.8 g (98%) 6-chloro-4-o-tolyl-nicotinamide as a beige crystalline foam. MS (ISP): 247 (M+H30 , 100).
Step 3:
1.0 g (4.05 mMol) 6-Chloro-4-o-tolyl-nicotinamidein 9.0 ml 1-methyl-piperazine was heated to 100° C. for 2 hours. The excess N-methyl-piperazine was removed under high vacuum and the residue was filtered on silica gel (eluent: dichloromethane) to yield 1.2 g (95%) 6-(4-methyl-piperazin-1yl)-4-o-tolyl-nicotinamide as a light yellow crystalline foam. MS (ISP): 311 (M+H+, 100), 254 (62).
Step 4:
A solution of 0.2 g (0.6 mMol) 6-(4-methyl-piperazin-1-yl)-4-o-tolyl-nicotinamide in 1.0 ml methanol was added to a solution of 103 mg (2.6 mMol) sodium hydroxide in 1.47 ml (3.2 mMol) NaOCl (13%) and heated for 2 hours at 70° C. After removal of methanol, the aqueous layer was extracted with ethyl acetate. The combined. organic extracts were dried (Na2SO4), concentrated under reduced pressure and the residue filtered on silica gel (eluent: dichloromethane/methanol 4:1) to yield 100 mg (70%) 6-(4-methyl-piperazine-1-yl)-4o-tolyl-pyridin-3-ylamine as a brown resin. MS (ISP): 283 (M+H+, 100), 226 (42).
Step 5:
2.15 ml (11.6 mMol) Sodium methoxide in methanol were added over 30 minutes to a suspension of 0.85 g (4.6 mMol) N-bromosuccinimide in 5.0 ml dichloromethane cooled to −5° C. The reaction mixture was stirred 16 hours at −5° C. Still at this temperature, a solution of 1.0 g (3.1 mMol) 6-(4-methyl-piperazin-1-yl)-4-o-tolyl-nicotinamide in 5.0 ml methanol was added over 20 minutes and stirred for 5 hours. 7.1 ml (7.1 mMol) Aqueous HCl 1N and 20 ml dichloromethane were added. The phases were separated and the organic phase was washed with deionized water. The aqueous phases were extracted with dichloromethane, brought to pH=8 with aqueous NaOH 1N and further extracted with dichloromethane. The latter organic, extracts were combined, dried (Na2SO4) and concentrated to yield 1.08 g (quant.) [6-(4-methyl-piperazin-1yl)-4-o-tolyl-pyridin-3-yl]-carbamic acid methyl ester as a grey foam. MS (ISP): 341 (M+H+, 100), 284 (35).
Step 6:
A solution of 0.5 g (1.4 mMol) [6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-carbamic acid methyl ester in 3.0 ml dichloromethane was added over 10 minutes to a solution of 1.98 ml (6.9 mMol) Red-Al.RTM. (70% in toluene) and 2.5 ml toluene (exothermic, cool with a water bath to avoid temperature to go >50° C.). The reaction mixture was stirred 2 hours at 50° C. in CH2Cl2, extracted with ethyl acetate and cooled to 0° C. 4 ml Aqueous NaOH 1N were carefully (exothermic) added over 15 minutes, followed by 20 ml ethyl acetate. The phases were separated and the aqueous phase was extracted with ethyl acetate. The combined organic extracts were washed with deionized water and brine, dried (Na2SO4) and concentrated under reduced pressure to yield 0.37 g (89%) methyl-[6-4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine as an orange resin. MS (ISP): 297 (M+H+, 100).
Synthesis of 2-(3,5-bis-Trifluoromethyl-phenyl)-2-methyl-propionyl Chloride
15.0 g (50 mmol) 2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionic acid were dissolved in 127.5 ml dichloromethane in the presence of 0.75 ml DMF. 8.76 ml (2 eq.) Oxalyl chloride were added and after 4.5 hours, the solution was rotary evaporated to dryness. 9 ml Toluene were added and the resulting solution was again rotary evaporated, then dried under high vacuum yielding 16.25 g (quant.) of 2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionyl chloride as a yellow oil of 86% purity according to HPLC analysis. NMR (250 MHz, CDCl3): 7.86 (br s, 1H); 7.77, (br s, 2H, 3 Harom); 1.77 (s, 6H, 2 CH3).
Synthesis of 2-(3,5-bis(trifluoromethyl)phenyl)-N,2-dimethyl-N-(6-(4-methylpiperazin-1-yl)-4-(o-tolyl)pyridin-3-yl)propanamide (Netupitant)
A solution of 20 g (67.5 mmol) methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine and 17.5 ml (101 mmol) N-ethyldiisopropylamine in 200 ml dichloromethane was cooled in an ice bath and a solution of 24 g (75 mmol)2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionyl chloride in 50 ml dichloromethane was added dropwise. The reaction mixture was warmed to 35-40° C. for 3 h, cooled to room temperature again and was stirred with 250 ml saturated sodium bicarbonate solution. The organic layer was separated and the aqueous phase was extracted with dichloromethane, The combined organic layers were dried (magnesium sulfate) and evaporated. The residue was purified by flash chromatography to give 31.6 g (81%) of 2-(3,5-bis(trifluoromethyl)phenyl)-N,2-dimethyl-N-(6-(4-methylpiperazin-1yl)-4-(o-tolyl)pyridin-3yl)propanamide as white crystals. M.P. 155-157° C.; MS m/e (%): 579 (M+H+, 100).
…………………………………..
http://www.google.com/patents/US20130231315
N OXIDE SYNTHESIS
Synthesis of 5-(2-(3,5-bis(trifluoromethyl)phenyl-N,2-dimethylpropanamido)2-(4-methylpiperazin-1yl)-4-(o-tolyl)pyridine 1-oxide
Step 1:
The solution of 6-chloropyridin-3-amine (115 g, 0.898 mol) and (Boc)2O (215.4 g, 0.988 mol) in 900 mL of dioxane was refluxed overnight. The resulting solution was poured into 1500 mL of water. The resulting solid was collected, washed with water and re-crystallized from EtOAc to afford 160 g tert-butyl (6-chloropyridin-3yl)carbamate as a white solid (Yield: 78.2%).
Step 2:
To the solution of tert-butyl (6-chloropyridin-3-yl)carbamate (160 g, 0.7 mol) in 1 L of anhydrous THF was added n-BuLi (600 mL, L5 ml) at −78° C. under N2 atmosphere. After the addition was finished, the solution was stirred at −78° C. for 30 min, and the solution of I2 (177.68 g, 0.7 mol) in 800 mL of anhydrous THF was added. Then the solution was stirred at −78° C. for 4 hrs, TLC indicated the reaction was over. Water was added for quench, and EtOAc was added to extract twice. The combined organic phases were washed with brine, dried over Na2SO4, filtered and purified by flash chromatography to afford 80 g of tert-butyl (6-chloro-4-iodopyridin-3-yl)carbamate as a yellow solid (32.3%).
Step 3:
To the solution of tert-butyl (6-chloro-4-iodopyridin-3-yl)carbamate (61 g, 0.172 mol) in 300 of anhydrous THF was added 60% NaH (7.6 g, 0.189 mol) at 0° C. under N2 atmosphere. After the addition was finished, the solution was stirred for 30 min, and then the solution of MeI (26.92 g, 0.189 mol) in 100 mL of dry THF was added. Then the solution was stirred at 0° C. for 3 hrs. TLC indicated the reaction was over. Water was added for quench, and EtOAc was added to extract twice. The combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated to afford 63 g of crude tert-butyl (6-chloro-4-iodopyridin-3-yl)methyl)carbamate used into the following de-protection without the further purification.
Step 4:
To the solution of tert-butyl (6-chloro-4-iodopyridin-3-yl)(methyl)carbamate (62.5 g, 0.172 mol) in 500 mL of anhydrous DCM was added 180 mL of TFA. Then the solution was stirred at room temperature for 4 hrs. Concentrated to remove the solvent, and purified by flash chromatography to afford 45.1 g 6-chloro-4-iodo-N-methylpyridin-3-amine as a yellow solid (Yield: 97.3%).
Step 5:
To the solution of 6-chloro-4-iodo-N-methylpyridin-3-amine (40.3 g, 0.15 mol) and 2-methylbenzene boric acid (24.5 g, 0.18 mol) in 600 mL of anhydrous toluene was added 400 mL of 2 N aq. Na2CO3 solution, Pd(OAc)2 (3.36 g, 15 mmol) and PPh3(7.87 g, 0.03 mmol), The solution was stirred at 100° C. for 2 hrs. Cooled to room temperature, and diluted with water. EtOAc was added to extract twice. The combined organic phases were washed with water and brine consecutively, dried over Na2SO4, concentrated and purified by flash chromatography to afford 19 g 6-chloro-N-methyl-4-(o-tolyl)pyridin-3-amine as a white solid (Yield: 54.6%).
Step 6:
To the solution of 6-chloro-N-methyl-4-(o-tolyl)pyridin-3-amine (18.87 g, 81.3 mmol) and DMAP (29.8 g, 243.9 mmol) in 200 mL of anhydrous toluene was added the solution of 2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionyl chloride (28.5 g, 89.4 mmol) in toluene under N2 atmosphere. The solution was heated at 120° C. for 23 hrs. Cooled to room temperature, poured into 1 L of 5% aq. NaHCO3 solution, and extracted with EtOAc twice. The combined organic phases were washed by water and brine consecutively, dried. over Na2SO4, filtered and purified by flash chromatography to afford 35 g 2-(3,5-bis(trifluoromethyl)phenyl)-N-(6-chloro-4-(4-tolyl)pyridin-3-yl)-N,2-dimethylpropanamide as a white solid (Yield: 83.9%).
Step 7:
To the solution of 2-(3,5-bis(trifluoromethyl)phenyl)-N-(6-chloro-4-(o-tolyl)pyridin-3-yl)-N,2-dimethylpropanamide (5.14 g, 10 mmol) in 60 mL of DCM was added m-CPBA (6.92 g, 40 mmol) at 0° C. under N2 atmosphere. Then the solution was stirred overnight at room temperature. 1 N aq. NaOH solution was added to wash twice for removing the excess m-CPBA. and a side product. The organic phase was washed by brine, dried over Na2SO4, filtered and concentrated to afford 5.11 g of crude 5-(2-(3,5-bis(trifluoromethyl)phenyl-N,2-dimethylpropanamido)-2-chloro-4(o-tolyl)pyridine 1-oxide as a white solid (Yield: 96.4%).
Step 8:
To the solution of crude 5-(2-(3,5-bis(trifluoromethyl)phenyl)-N,2-dimethylpropanamido)-2-chloro-4-(o-tolyl)pyridine 1-oxide (5.1 g, 9.62 mmol) in 80 mL of n-BuOH was added N-methylpiperazine (7.41 g, 74.1 mmol) under N2 atmosphere. Then the solution was stirred at 80° C. overnight. Concentrated and purified by flash chromatography to afford 4.98 g 5-(2-(3,5-bis(trifluoromethyl)phenyl-N,2-dimethylpropanamido)-2-(4-methylpiperazin-1-yl)-4-(o-tolyl)pyridine 1-oxide as a white solid (Yield: 87.2%), 1HNMR (CDCl3, 400 MHz) δ 8.15 (s, 1H), 7.93 (s, 1H), 7.78 (s, 2H), 7.38 (m, 2H), 7.28 (m, 1H), 7.17 (m, 1H), 7.07 (s, 1H), 5.50 (s, 3H), 2.72 (d, J=4.4 Hz, 4H), 2.57 (m, 3H), 2.40 (s, 3H), 2.23 (s, 3H), 1.45-1.20 (m, 6H).
………………………………….
https://www.google.co.in/patents/US6479483
EXAMPLE 14 2-(3,5-bis-Trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperan-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide Hydrochloride (1:2)
a) 1-Methyl-4-(5-nitro-pyridin-2-yl)-piperazine
To a solution of 20 g (126 mmol) of 2-chloro-5-nitropyridine in 200 ml tetrahydrofuran were added dropwise 35 ml (315 mmol) 1-methylpiperazine within 10 min. The reaction mixture was refluxed for additional 1.5 h. After cooling to room temperature, the solvent was removed in vacuo and the residue was re-dissolved in 200 ml ethyl acetate. The organic phase was washed with 200 ml 1 N sodium bicarbonate solution, dried (magnesium sulfate) and evaporated to give 27.9 g (quantitative) of the title compound as a yellow solid.
MS m/e (%): 223 (M+H+, 100).
b) 2,2-Dimethyl-N-[6-(4-methyl-piperazin-1-yl)-pyridin-3-yl-propionamide
To a solution of 27.9 g (125.5 mmol) of 1-methyl-4-(5-nitro-pyridin-2-yl)-piperazine in 400 ml methanol were added 2.6 g of 10% of palladium on activated charcoal. The reaction mixture was hydrogenated (room temperature to ca. 45° C., 1 bar) until the theoretical amount of hydrogen was taken up (about 2 h). The catalyst was filtered off and was washed twice with 100 ml portions of methanol. The filtrate was evaporated in vacuo to give 28 g of a purple oil which consisted to ca. 90% of the desired aniline derivative according to analysis by thin layer chromatography.
This crude product was dissolved in a mixture of 400 ml tetrahydrofuran and 100 ml diethyl ether. After cooling to 0° C., 30 ml (215 mmol) of triethylamine were added in one portion. Stirring was continued while 26 g (215 mmol) of pivaloyl chloride were added dropwise within a period of 10 min. The ice bath was removed and the reaction mixture was stirred for 1 h at room temperature. Then, the solvent was removed in vacuo and the residue was suspended in 200 ml 1 N sodium bicarbonate solution. The product was extracted three times with 200 ml portions of dichloromethane, dried (sodium sulfate) and purified by flash chromatography to give 30 g (86%) of the title compound as pink crystals.
MS m/e (%): 277 (M+H+, 100).
c) N-[4-Iodo-6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-2,2-dimethyl-propionamide
A solution of 30 g (108 mmol) 2,2-dimethyl-N-[6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-propionamide and 58 ml (380 mmol) N,N,N′,N′-tetramethylethylenediamine under argon in 650 ml tetrahydrofuran was cooled in a dry ice bath to −78° C. Within 1 h, 239 ml (380 mmol) of a 1.6 N n-butyllithium solution in hexane were added dropwise. The reaction mixture was allowed to warm up to −30° C. overnight. After cooling again to −78° C., 43.6 g (170 mmol) iodine dissolved in 60 ml tetrahydrofuran were added dropwise during 15 min. The dry ice bath was replaced by an ice bath and a solution of 90 g (363 mmol) sodium thiosulfate pentahydrate in 250 ml water were added within 10 min when the temperature of the reaction mixture had reached 0° C. Then, 1000 ml diethyl ether were added and the organic layer was separated. The aqueous layer was extracted twice with 500 ml dichloromethane and the combined organic layers were dried (magnesium sulfate) and evaporated. Flash chromatography gave 18.5 g (42%) of the tide compound as a light brown oil which crystallized upon standing at room temperature.
MS m/e (%): 403 (M+H+, 100).
d) 2,2-Dimethyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-propionamide
A mixture of 54 g (134 mmol) N-[4-iodo-6-(4-methyl-piperazin-1-yl)-pyridin-3-yl]-2,2-dimethyl-propionamide, 420 ml toluene, 150 ml 2 N sodium carbonate solution, 4.63 g (3.9 mmol) tetrakis(triphenylphosphine)palladium(0) and 20.16 g (147 mmol) o-tolylboronic acid was heated under argon at 80° C. for 12 h. After cooling to room temperature, the aqueous phase was separated and washed twice with toluene. The combined organic layers were washed with 50 ml brine, dried (sodium sulfate), evaporated and dried in vacuo to yield 49 g (quantitative) of the title compound as a brown oil.
MS m/e (%): 367 (M+H+, 100).
e) 6-(4-Methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-ylamine
A suspension of 56 g (152 mmol) 2,2-dimethyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-propionamide in 1300 ml 3 N hydrochloric acid solution was heated to 90-95° C. overnight. The reaction mixture was cooled to room temperature, washed with three 500 ml portions diethyl ether and filtered over celite. The filtrate was diluted with 500 ml water and was adjusted to pH 7-8 by addition of 28% sodium hydroxide solution under ice cooling. The product was extracted with four 1000 ml portions of dichloromethane. The combined organic layers were washed with 500 ml brine, dried (magnesium sulfate) and evaporated to give 35 g (82%) of the title compound as a light brown oil.
MS m/e (%):283 (M+H+, 100).
f) Methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-amine
A solution of 35 g (124 mmol) 6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-ylamine in 270 ml trimethyl orthoformate and 8 drops trifluoroacetic acid was heated for 3 h at 130° C. The reaction mixture was evaporated and dried in vacuo for 30 min. The residual oil was dissolved in 100 ml tetrahydrofuran and was added dropwise under ice cooling to 9.4 g (248 mmol) lithium aluminum hydride in 300 ml tetrahydrofuran. The reaction mixture was stirred for 1 h at room temperature, cooled to 0° C. again and acidified (pH 1-2) by addition of 28% hydrochloric acid solution. After stirring for 5 min, 28% sodium hydroxide solution was added to reach pH 10. The solution was filtered over celite, evaporated and purified by flash chromatography to give 23.6 g (64%) of the title compound as a light brown oil.
MS m/e (%): 297 (M+H+, 100).
g) 2-(3,5-bis-Trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide
A solution of 20 g (67.5 mmol) methyl-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]amine and 17.5 ml (101 mmol) N-ethyldiisopropylamine in 200 ml dichloromethane was cooled in an ice bath and a solution of 24 g (75 mmol) 2-(3,5-bis-trifluoromethyl-phenyl)-2-methyl-propionyl chloride in 50 ml dichloromethane was added dropwise. The reaction mixture was warmed to 35-40° C. for 3 h, cooled to room temperature again and was stirred with 250 ml saturated sodium bicarbonate solution. The organic layer was separated and the aqueous phase was extracted with dichloromethane. The combined organic layers were dried (magnesium sulfate) and evaporated. The residue was purified by flash chromatography to give 31.6 g (81%) of the title compound as white crystals. M.p. 155-157° C.
MS m/e (%): 579 (M+H+, 100).
h) 2-(3,5-bis-Trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide Hydrochloride (1:2)
To a solution of 31.6 g (54.6 mmol) 2-(3,5-bis-trifluoromethyl-phenyl)-N-methyl-N-[6-(4-methyl-piperazin-1-yl)-4-o-tolyl-pyridin-3-yl]-isobutyramide in 250 ml diethyl ether were added under ice cooling 60 ml 3 N hydrochloric acid solution in diethyl ether. After stirring for 15 min at 0° C., the suspension was evaporated to dryness, re-suspended in 100 ml diethyl ether, filtered and dried in vacuo to give 34.8 g (98%) of the title compound as white crystals. M.p. 235-238° C.
MS m/e (%): 579 (M+H+, 100).
…………………………….
Research and development of an efficient process for the construction of the 2,4,5-substituted pyridines of NK-1 receptor antagonists
Org Process Res Dev 2006, 10(6): 1157
Navari RM.
Drugs. 2013 Mar;73(3):249-62. doi: 10.1007/s40265-013-0019-1. Review.
-
Hoffmann-Emery F, Hilpert H, Scalone M, Waldmeier P.
J Org Chem. 2006 Mar 3;71(5):2000-8.
Hoffmann T, Bös M, Stadler H, Schnider P, Hunkeler W, Godel T, Galley G, Ballard TM, Higgins GA, Poli SM, Sleight AJ.
Bioorg Med Chem Lett. 2006 Mar 1;16(5):1362-5. Epub 2005 Dec 5.
http://www.sciencedirect.com/science/article/pii/S0960894X05014824
…………………………………….……………………………………………………….
US6897226 * | 9 Jul 2003 | 24 May 2005 | Hoffmann-La Roche Inc. | NK-1 receptor active amine oxide prodrugs |
US7211579 * | 15 Mar 2006 | 1 May 2007 | Hoffmann-La Roche Inc. | NK-1 receptor antagonists |
US8426450 | 23 May 2012 | 23 Apr 2013 | Helsinn Healthcare Sa | Substituted 4-phenyl pyridines having anti-emetic effect |
WO2011061622A1 | 18 Nov 2010 | 26 May 2011 | Helsinn Healthcare S.A. | Compositions for treating centrally mediated nausea and vomiting |
WO2013057554A2 | 10 Oct 2012 | 25 Apr 2013 | Helsinn Healthcare Sa | Therapeutic combinations of netupitant and palonosetron |
US8426450 | 23 May 2012 | 23 Apr 2013 | Helsinn Healthcare Sa | Substituted 4-phenyl pyridines having anti-emetic effect |
WO2011061622A1 | 18 Nov 2010 | 26 May 2011 | Helsinn Healthcare S.A. | Compositions for treating centrally mediated nausea and vomiting |
WO2013057554A2 | 10 Oct 2012 | 25 Apr 2013 | Helsinn Healthcare Sa | Therapeutic combinations of netupitant and palonosetron |
……………………………………………………………………………………….

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D
GLENMARK SCIENTIST , NAVIMUMBAI, INDIA
did you feel happy, a head to toe paralysed man’s soul in action for you round the clock
need help, email or call me
I was paralysed in dec2007
Sprout Pharmaceuticals Appeals FDA Decision on NDA for Flibanserin to Treat Hypoactive Sexual Desire Disorder in Premenopausal Women
Flibanserin, girosa
167933-07-5 cas no
147359-76-0 (monoHCl)
- Bimt 17
- BIMT 17 BS
- Bimt-17
- Flibanserin
- Girosa
- UNII-37JK4STR6Z
December 11, 2013 – Sprout Pharmaceuticals today announced that it has received and appealed the Food and Drug Administration’s (FDA) Complete Response Letter (CRL) for flibanserin through the Formal Dispute Resolution process.
Flibanserin is an investigational, once-daily treatment for Hypoactive Sexual Desire Disorder, or HSDD, in premenopausal women. HSDD is the most commonly reported form of female sexual dysfunction
read all here
A new drug being developed by Boehringer Ingelheim could give a boost to the sex drive of women with low libido. The drug, known as flibanserin, has been shown in clinical trials to increase their sexual desire when taken once a day at bedtime.
The results from four pivotal Phase III clinical trials on women with hypoactive sexual desire disorder (HSDD) were presented this week at the European Society for Sexual Medicine’s congress in Lyon, France. The trials showed that participants taking flibanserin had a significant improvement in their sexual desire compared to those given a placebo. They also experienced less of the distress associated with sexual dysfunction.
The drug was initially being investigated as a treatment for depression, and acts on the serotonin receptors in the brain – it is both a 5-HT1A receptor agonist and a 5-HT2A receptor antagonist. It is also a partial agonist at the dopamine D4 receptor.
Neurotransmitters such as serotonin are believed to be involved in sexual function, and antidepressants are commonly associated with a loss of libido, so this was an obvious side-effect to look out for during clinical trials in depression. But far from suppressing the libido in women, it appeared to have the opposite effect, so trials in women with HSDD were initiated.
Hormone replacement can improve the libido of women who have had their ovaries removed, but there is no available drug to treat those who have not. There have been accusations that pharma companies invent new diseases like HSDD in order to sell more medicines, but according to Kathleen Segraves, an assistant professor at Case Western Reserve University in the US who has worked in the field of sexual functioning for many years, this is not the case here. HSDD is a very real disorder, she says, and the potential for a treatment for these women is very exciting.
Flibanserin (code name BIMT-17; proposed trade name Girosa) is a drug that was investigated by Boehringer Ingelheim as a novel, non-hormonal treatment for pre-menopausal women with Hypoactive Sexual Desire Disorder (HSDD).[1][2] Development was terminated in October 2010 following a negative report by the U.S. Food and Drug Administration.[3]
HSDD is the most commonly reported female sexual complaint and characterized by a decrease in sexual desire that causes marked personal distress and/or personal difficulties. According to prevalence studies about 1 in 10 women reported low sexual desire with associated distress, which may be HSDD.[4] The neurobiological pathway of female sexual desire involves interactions among multiple neurotransmitters, sex hormones and various psychosocial factors. Sexual desire is modulated in distinct brain areas by a balance between inhibitory and excitatory neurotransmitters, serotonin acting as an inhibitor while dopamine and norepinephrine act as a stimulator of sexual desire.[5][6]Flibanserin is a 5-HT1A receptor agonist and 5-HT2A receptor antagonist that had initially been investigated as an antidepressant. Preclinical evidence suggested that flibanserin targets these receptors preferentially in selective brain areas and helps to restore a balance between these inhibitory and excitatory effects.[6] HSDD has been recognized as a distinct sexual function disorder for more than 30 years.
The proposed mechanism of action refers back to the Kinsey dual control model. Several sex steroids, neurotransmitters, and hormones have important excitatory or inhibitory effects on the sexual response. Among the neurotransmitters, the excitatory activity is driven by dopamine and norepinephrine, while the inhibitory activity is driven by serotonin. The balance between these systems is relevant for a healthy sexual response. By modulating these neurotransmitters in selective brain areas, flibanserin, a 5-HT1A receptoragonist and 5-HT2A receptor antagonist, is likely to restore the balance between these neurotransmitter systems.[6]
Several large pivotal Phase III studies with Flibanserin were conducted in the USA, Canada and Europe. They involved more than 5,000 pre-menopausal women with generalized acquired Hypoactive Sexual Desire Disorder (HSDD). The results of the Phase III North American Trials demonstrated that
Although the two North American trials that used the flibanserin 100 mg qhs dose showed a statistically significant difference between flibanserin and placebo for the endpoint of [satisfying sexual events], they both failed to demonstrate a statistically significant improvement on the co-primary endpoint of sexual desire. Therefore, neither study met the agreed-upon criteria for success in establishing the efficacy of flibanserin for the treatment of [Hypoactive Sexual Desire Disorder].
These data were first presented on November 16, 2009 at the congress of the European Society for Sexual Medicine in Lyon, France. The women receiving Flibanserin reported that the average number of times they had “satisfying sexual events” rose from 2.8 to 4.5 times a month. However, women receiving placebo reported also an increase of “satisfying sexual events” from 2.7 to 3.7 times a month.
Evaluation of the overall improvement of their condition and whether the benefit was meaningful to the women, showed a significantly higher rate of a meaningful benefit in the flibanserin-treated patient group versus the placebo group.The onset of the Flibanserin effect was seen from the first timepoint measured after 4 weeks of treatment and maintained throughout the treatment period.
The overall incidence of adverse events among women taking flibanserin was low, the majority of adverse events being mild to moderate and resolved during the treatment. The most commonly reported adverse events included dizziness, nausea, fatigue, somnolence and insomnia.
On June 18, 2010, a federal advisory panel to the U.S. Food and Drug Administration (FDA) unanimously voted against recommending approval of Flibanserin.
Earlier in the week, a FDA staff report also recommended non-approval of the drug. While the FDA still might approve Flibanserin, in the past, negative panel votes tended to cause the FDA not to approve.
On October 8, 2010, Boehringer Ingelheim announced that it would discontinue its development of flibanserin in light of the FDA advisory panel’s recommendation.
On June 27, 2013, Sprout Pharmaceuticals confirmed they had resubmitted flibanserin for FDA approval.
Flibanserin, chemically 1 -[2-(4-(3-trifluoromethylphenyl)piperazin-1 – yl)ethyl]-2,3-dihydro-1 H-benzimidazole-2-one was disclosed in form of its hydrochloride in European Patent No. 526,434 (‘434) and has the following chemical structure:
Process for preparation of flibanserin were disclosed in European Patent No. ‘434, U.S. Application Publication No. 2007/0032655 and Drugs of the future 1998, 23(1): 9-16.
According to European Patent No. ‘434 flibanserin is prepared by condensing 1-(2-chloroethyl)-2,3-dihydro-1 H-benzimidazol-one with m- trifluoromethyl phenyl piperazine. According to U.S. Application Publication No. 2007/0032655 flibanserin is prepared by condensing 1-[(3-trifluoromethyl)phenyl]-4-(2- chloroethyl)piperazine with 1 -(2-propenyl)-1 ,3-dihydro-benzimidazol-2H-one.
According to Drugs of the future 1998, 23(1): 9-16 flibanserin is prepared by reacting 1-(2-chloroethyl)-2,3-dihydro-1 H-benzimidazol-one with m- trifluoromethylphenylpiperazine.
…………………
1-[2-(4-(3-trifluoromethyl-phenyl)piperazin-1-yl)ethyl]-2,3-dihydro-1H-benzimidazol-2-one
Compound 3
- Hydrochloride salt (isopropanol) M.p. 230-231°C
Analysis
-
¹H NMR (DMSO-d₆/CDCL₃ 5:2) 11.09 (b, 1H), 11.04 (s, 1H), 7.5-6.9 (8H), 4.36 (t, 2H), 4.1-3.1 (10H)
…………………………………
The compound 1-[2-(4-(3-trifluoromethyl-phenyl)piperazin-1-yl)ethyl]-2,3-dihydro-1 H- benzimidazol-2-one (flibanserin) is disclosed in form of its hydrochlorid in European Patent Application EP-A-526434 and has the following chemical structure:
Flibanserin shows affinity for the 5-HTιA and 5-HT2-receptor. It is therefore a promising therapeutic agent for the treatment of a variety of diseases, for instance depression, schizophrenia, Parkinson, anxiety, sleep disturbances, sexual and mental disorders and age associated memory impairment.
EXAMPLE……… EP1518858A1
375 kg of 1-[(3-trifluoromethyl)phenyl]-4-(2-cloroethyl)piperazin are charged in a reactor with 2500 kg of water and 200 kg of aqueous Sodium Hydroxide 45%. Under stirring 169.2 kg of 1-(2-propenyl)-1,3-dihydro-benzimidazol-2H-one, 780 kg of isopropanol, 2000 kg of water and 220 kg of aqueous Sodium Hydroxide 45% are added. The reaction mixture is heated to 75-85° C. and 160 kg of concentrated hydrochloric acid and 200 kg of water are added.
The reaction mixture is stirred at constant temperature for about 45 minutes. After distillation of a mixture of water and Isopropanol (about 3000 kg) the remaining residue is cooled to about 65-75° C. and the pH is adjusted to 6.5-7.5 by addition of 125 kg of aqueous Sodium Hydroxide 45%. After cooling to a temperature of 45-50° C., the pH value is adjusted to 8-9 by addition of about 4 kg of aqueous Sodium Hydroxide 45%. Subsequently the mixture is cooled to 30-35° C. and centrifuged. The residue thus obtained is washed with 340 l of water and 126 l of isopropanol and then with water until chlorides elimination.
The wet product is dried under vacuum at a temperature of about 45-55° C. which leads to 358 kg of crude flibanserin polymorph A. The crude product thus obtained is loaded in a reactor with 1750 kg of Acetone and the resulting mixture is heated under stirring until reflux. The obtained solution is filtered and the filtrate is concentrated by distillation. The temperature is maintained for about 1 hour 0-5° C., then the precipitate solid is isolated by filtration and dried at 55° C. for at least 12 hours.
The final yield is 280 kg of pure flibanserin polymorph A.
………………………….
Flibanserin may be prepared by reacting 1-(phenylvinyl)-2,3-dihydro-1H-benzimidazol-2-one (I) with 1,2-dichloroethane (II) in the presence of NaH in warm dimethylformamide. The resulting 1-(2-chloroethyl)-2,3-dihydro-1H-benzimidazol-one (III) is in turn coupled with commercially available m-trifluoromethylphenylpiperazine hydrochloride (IV) in the presence of sodium carbonate and catalytic potassium iodide in refluxing ethanol. The crude flibanserin hydrochloride (V) is then dissolved in aqueous ethanol and the pure base is precipitated upon addition of sodium hydroxide.
PICK UP INTERMEDIATES FROM CHEM24H.COM
1-(1-phenylvinyl)-1,3-dihydro-2H-benzimidazol-2-one (I)
1,2-dichloroethane (II)
1-(2-chloroethyl)-1,3-dihydro-2H-benzimidazol-2-one (III)
1-[3-(trifluoromethyl)phenyl]piperazine; N-[3-(trifluoromethyl)phenyl]piperazine (IV)
1-(2-[4-[3-(trifluoromethyl)phenyl]piperazino]ethyl)-1,3-dihydro-2H-benzimidazol-2-one (V)
………………………..
According to another aspect of the present invention there is provided a novel compound or a salt thereof selected from the compounds of formula I, IV and VII:
Wherein R is hydrogen or an amino protecting group.
Preferable the amino protecting groups are selected from butyl, 1 ,1- diphenylmethyl, methoxymethyl, benzyloxymethyl, trichloroethoxymethyl, pyrrolidinomethyl, cyanomethyl, pivaloyloxymethyl, allyl, 2-propenyl, t- butyldimethylsilyl, methoxy, thiomethyl, phenylvinyl, 4-methoxyphenyl, benzyl, A- methoxybenzyl, 2,4-dimethoxybenzyl, 2-nitrobenzyl, t-butoxycarbonyl, benzyloxycarbonyl, phenoxycarbonyl, 4-chlorophenoxycarbonyl, A- nitrophenoxycarbonyl, methoxycarbonyl and ethoxycarbonyl. Still more preferable protecting groups are selected from t- butoxycarbonyl, ethoxycarbonyl, methoxycarbonyl, benzyloxycarbonyl, phenoxycarbonyl, phenylvinyl and 2-propenyl.
R1 is independently selected from chlorine, bromine, iodine, methanesulphonate, trifluoromethanesulphonate, paratoluenesulphonate or benzenesulphonate. Preferable R1 is independently selected from chlorine, bromine or iodine and more preferable R1 is chlorine.
Wherein R2 is hydrogen or an amino protecting group.
The amino protecting group may be any of the groups commonly used to protect the amino function such as alkyl, substituted alkyl, hetero substituted alkyl, substituted or unsubstituted unsaturated alkyl, alkyl substituted hetero atoms, substituted or unsubstituted phenyl, substituted or unsubstituted benzyl, alkyoxy carbonyl groups and aryloxy carbonyl groups.
Preferable the amino protecting groups are selected from butyl, 1 ,1 – diphenylmethyl, methoxymethyl, benzyloxymethyl, trichloroethoxymethyl, pyrrolidinomethyl, cyanomethyl, pivaloyloxymethyl, allyl, 2-propenyl, t- butyldimethylsilyl, methoxy, thiomethyl, phenylvinyl, 4-methoxyphenyl, benzyl, A- methoxybenzyl, 2,4-dimethoxybenzyl, 2-nitrobenzyl, t-butoxycarbonyl, benzyloxycarbonyl, phenoxycarbonyl, 4-chlorophenoxycarbonyl, A- nitrophenoxycarbonyl, methoxycarbonyl and ethoxycarbonyl. Still more preferable protecting groups are selected from t- butoxycarbonyl, ethoxycarbonyl, methoxycarbonyl, benzyloxycarbonyl, phenoxycarbonyl, phenylvinyl and 2-propenyl. The following examples are given for the purpose of illustrating the present invention and should not be considered as limitations on the scope and spirit of the invention.
EXAMPLES Example 1
A mixture of sodium hydroxide (47 gm) and i-(α-methylvinyl) benzimidazol-2-one (100 gm) in dimethylformamide (400 ml) was .stirred for 1 hour at room temperature. Dibromoethane (217 gm) was slowly added to the mixture and stirred at 1 hour 30 minutes. The resulting solution after addition water (500 ml) was extracted with ethyl acetate. The combined ethyl acetate extract washed with water. After drying the solvent was removed under vacuum to yield 132 gm of 1 ,3-dihydro-1-(2-bromoethyl)-3-isopropenyl-2H-benzimidazol- 2-one as a yellow oily liquid.
Example 2 A mixture of 1 ,3-dihydro-1-(2-bromoethyl)-3-isopropenyl-2H- benzimidazol-2-one (100 gm), diethanolamine (175 ml), sodium carbonate (40 gm) and potassium iodide (10 gm) was heated to 90 to 95 deg C and stirred for 2 hours. The reaction mass was cooled to room temperature and added water (500 ml). The resulting mixture extracted into ethyl acetate and the organic layer washed with water. After drying the solvent was removed under vacuum to yield 105 gm of 1 ,3-dihydro-1-[2-[N-bis-(2-hydroxyethyl)amino]ethyl]-3-isopropenyl- 2H-benzimidazol-2-one as a thick yellow oily liquid.
Example 3
To the mixture of 1 ,3-dihydro-1-[2-[N-bis-(2-hydroxyethyl)amino]ethyl]-3- isopropenyl-2H-benzimidazol-2-one (100 gm) obtained as in example 2 and chloroform (300 ml), thionyl chloride (95 ml) was slowly added. The mixture was heated to reflux and stirred for 2 hours. The excess thionyl chloride and chloroform was distilled off to yield 98 gm of 1 ,3-dihydro-1-[2-[N-[bis-(2- chloroethyl)amino]ethyl]-3-isopropenyl-2H-benzimidazol-2-one as a brown coloured sticky residue.
Example 4
1 ,3-dihydro-1-[2-[N-[bis-(2-chloroethyl)amino]ethyl]-3-isopropenyl-2H- benzimidazol-2-one (98 gm) obtained as in example 3 was added to water (500 ml) and concentrated hydrochloric acid (200 ml) mixture. The mixture was heated to 60 to 65 deg C and stirred for 1 hour. The contents of the flask cooled to room temperature and pH of the solution adjusted to 9 – 10 with 10% sodium hydroxide solution. The resulting solution extracted with ethyl acetate and washed the organic layer with water. Evaporate the solvent under reduced pressure to yield 82 gm of 1 ,3-dihydro-1-[2-[N-bis-(2-chloroethyl)amino]ethyl]- 2H-benzimidazol-2-one as a dark brown coloured oily liquid
Example 5
A mixture of 1 ,3-dihydro-1-[2-[N-bis-(2-chloroethyl)amino]ethyl]-1,2-H- benzimidazol-2-one (82 gm) obtained as in example 4, xylene (300 ml) and m- trifluoromethyl aniline (58 gm) was refluxed for 64 hours. The reaction mass was cooled to room temperature and filtered to obtain 1-[2-(4-(3- thfluoromethylphenyl)piperazin-1-yl)ethyl]-2,3-dihydro-1 H-benzimidazole-2-one hydrochloride (Flibanserin hydrochloride) as a light brown coloured solid.
The crude flibanserin hydrochloride was purified in isopropyl alcohol to give 85 gm of pure flibanserin hydrochloride as off white solid.
Example 6
Piperazine (12 gm), toluene(60 ml) and tetra butyl ammonium bromide (1 gm) mixture was heated to 60 deg C, added 1 ,3-dihydro-1-(2-bromoethyl)-3- isopropenyl-2H-benzimidazol-2-one (10 gm) and stirred for 4 hours at 90 to 95 deg C. The mixture was cooled to 60 deg C and added water (50 ml). The separated toluene layer distilled under vacuum to give 8.5 gm of 1 ,3-dihydro-1- (2-piperazinyl)ethyl-3-isopropenyl-2H-benzimidazol-2-one as a white solid.
Example 7
To the mixture of concentrated hydrochloric acid (20 ml) and water (100 ml) was added 1 ,3-dihydro-1-(2-piperazinylethyl)-3-isopropenyl-2H- benzimidazol-2-one (10 gm) obtained as in example 6 and heated to 60 to 65 deg C 1 hour. The mixture was cooled to room temperature and pH of the solution was adjusted to 9 – 10 with 10% sodium hydroxide solution, extracted with ethyl acetate and the organic layer was washed with water. After drying the solvent was removed under vacuum to yield 8.5 gm of 1 ,3-dihydro-1-(2- piperazinyl ethyl)-2H-benzimidazol-2-one as a white solid.
Example 8
3-trifluoromethylaniline (40 gm) and hydrobromic acid (85 ml; 48- 50%w/w) mixture was cooled to 0 to 5 deg C. To this mixture added sodium nitrite solution (18.5 gm in 25 ml of water) at 5 to 10 deg C and copper powder (1 gm). The temperature was slowly raised to 50 to 55 deg C and stirred for 30 minutes. Added water (200 ml) to reaction mass and applied steam distillation, collected m-trifluoromethylbromobenzene as oily liquid. The oily liquid washed with sulfuric acid for two times (2 X 10 ml) followed by washed with water (2 X 20 ml) and dried the liquid with sodium sulphate to give 22 gm of m- trifluoromethylbromobenzene.
Example 9
To a mixture of 1 ,3-dihydro-1-(2-piperazinyl ethyl)-2H-benzimidazol-2- one (10 gm) obtained as in example 7, m-trifluoromethylbromobenzene (9 gm) obtained as in example 8, sodium tert-butoxide (5.5 gm), palladium acetate (4.5 mg) and xylene (80 ml) was added tri-tert.-butylphosphine (0.2 ml). The mixture was heated to 120 deg C and stirred for 3 hours. The reaction mass was cooled, added water (100 ml) and extracted with ethyl acetate and the organic layer was washed with water. After drying the solvent was removed under vacuum to yield
10 gm of 1-[2-(4-(3-trifluoromethylphenyl)piperazin-1-yl)ethyl]-2,3-dihydro-1 H- benzimidazole-2-one (Flibanserin).
Example 10
To a mixture of 1 ,3-dihydro-1-[2-[N-bis-(2-hydroxyethyl)amino]ethyl]-3- isopropenyl-2H-benzimidazol-2-one (100 gm) obtained as in example 3, cyclohexane (400 ml) and sodium carbonate (35 gm) was added benzene sulfonyl chloride (116 gm) at room temperature. The mixture was heated to 80 to
85 deg C and stirred for 8 hours . The contents were cooled to room temperature and added water (500 ml). Distilled the organic layer to give 182 gm of 1 ,3-dihydro-1-[2-[N-[bis-(2-benzenesulfonyloxy)- ethyl]amino]ethyl]-3- isopropenyl- 2H-benzimidazol-2-one.
Example 11
1 ,3-dihydro-1 -[2-[N-[bis-(2-benzenesulfonyloxy)- ethyl]amino]ethyl]-3- isopropenyl- 2H-benzitηidazol-2-one (100 gm) obtained as in example 10, dimethylformamide (500 ml) and sodium corbonate (18 gm) was mixed and heated to 70 deg C. To the mixture was added m-trifluoromethyl aniline (27 gm) and heated to 80 to 85 deg C, stirred for 5 hours. The reaction mass was cooled and added water (2000 ml), filtered the solid to yield 1 ,3-dihydro-1-[2-[4-(3- trifluoromethylphenyl)piperazinyl]ethyl]-3-isopropenyl-2H benzimidazol-2-one. Example 12
1 ,3-dihydro-1-[2-[N-[bis-(2-benzenesulfonyloxy)- ethyl]amino]ethyl]-3- isopropenyl- 2H-benzimidazol-2-one (100 gm) obtained as in example 11 added to the mixture of water (500 ml) and concentrated hydrochloric acid (200 ml), heated to 65 deg C and stirred for 1 hour. The reaction mass was cooled to room temperature and pH adjusted to 10 to 10-5 with 10% sodium hydroxide solution. The resulting mixture was extracted with ethyl acetate and the organic
■ layer was washed with water. After drying the solvent was removed under vacuum to yield 87 gm of 1-[2-(4-(3-trifluoromethylphenyl)piperazin-1-yl)ethyl]- 2,3-dihydro-1 H-benzimidazole -2-one (Flibanserin).
…………………..
Paper
Journal of Pharmaceutical and Biomedical Analysis, v.57, 2012 Jan 5, p.104(5)
Isolation and structural elucidation of flibanserin as an adulterant in a health supplement used for female sexual performance enhancement
Low, Min-Yong et al
http://www.sciencedirect.com/science/article/pii/S0731708511004833
This proposed formula and structure was further confirmed by 1H and 13C NMR data which indicated the presence of 20 carbon atoms and 21 protons.
1H NMR
13C NMR
1D and 2DNMR data were used to assign the protons and carbon atoms.
In the1H NMR spectrum , a sharp singlet at 10.00 ppm integrating for one
proton is a typical proton attached to nitrogen. HMBC correlated this proton to C-2, C-4, and C-9 suggesting that it was H-3.
Complex signals were observedbetween 7.00 to 7.31 ppm, integrating for eight protons. A triplet at 7.31 ppm,integrating for a proton has a coupling constant of 8.0 Hz. HMBC correlated thisproton with C-16, C-19, and C-21 suggesting that it was H-20.
A double-doubletsplitting pattern at chemical shift 7.11 ppm, integrating for a proton, has couplingconstants of 6.3 Hz and 1.6 Hz.
HMBC correlated this proton to C-6, C-7, and C-9 showing that it was H-8. Overlapped signals were observed from 7.04 ppm to7.10 ppm, integrating for five protons. A double-doublet splitting pattern at 7.01ppm with coupling constant 8.0 Hz and 2.0 Hz, integrating for a proton was
observed.
HMBC correlated this proton to C-17 suggesting that it was either H-19or H-21. Four triplet signals were also observed from 2.73 ppm to 4.08 ppm,integrating for a total of twelve protons.
Two of these triplet signals at 2.74 ppmand 3.22 ppm integrated for four protons each, suggesting overlapping signals ofmethylene protons. This was further confirmed by 13C and DEPT NMR.
13C and DEPT NMR data showed the signals of four methylene, eight methineand six quaternary carbon atoms. The DEPT signals at 53.1 ppm and 48.6 ppmhave intensities which were double of those from the rest of the methylene carbonsignals, suggesting two methylene carbon atoms each contributing to the signal at 53.1 ppm and 48.6 ppm.
DEPT
HMQC results further indicated that these two methylene carbon signals at 53.1 ppm and 48.6 ppm were correlated to the protons signal at 2.73 ppm and 4.08 ppm respectively, which corresponded to four protons each. The finding confirmed overlapping methylene carbon signals (at 53.1 ppm and 48.6 ppm) and methylene proton signals (at 2.73 ppm and 4.08 ppm). Hence, the unknown compound has six methylene carbon atoms with a total of twelve methylene protons.
The chemical shifts of the twelve methylene protons suggested that they were attached to relatively electronegative atoms. It was speculated that the six methylene groups were attached to the nitrogen atoms and the electron withdrawing effect of these electronegative nitrogen atoms resulted in the deshielding of the protons. HMBC and COSY correlations were used to assign the rest of the protons
The 13C NMR data showed that there were two quaternary carbon at
155.6 ppm and 151.3 ppm. The carbon with chemical shift 155.6 ppm was C-2. Inthe structure of imidazolone, carbonyl carbon C-2 was attached to two nitrogenatoms which helped to withdraw electrons from oxygen to C-2. Hence, C-2 wasless deshielded as compared to a normal carbonyl carbon which has chemical shiftabove 170 ppm.
Eight methine carbons and two quaternary carbons with chemicalshifts above 108 ppm suggested the presence of two aromatic rings. Thequaternary carbon with chemical shift 125.4 ppm was C-22 which was attached tothree fluorine atoms. Due to the strong electron withdrawing effect of the fluorineatoms, C-22 was highly deshielded and had a high chemical shift.
The IR spectrum of the isolated compound showed absorption bands of amide (νC=O 1685 cm-1, νN-H (stretch) 3180 cm-1, νN-H (bending) 1610 cm-1), alkyl fluoride (νC-F1077 cm-1, 1112 cm-1, 1158 cm-1), aromatic ring (ν Ar-H 3028 cm-1, 3078 cm-1 andνC=C 1401 cm-1, 1446 cm-1, 1453 cm-1, 1468 cm-1, 1487 cm-1) and alkane (νC-H2891 cm-1, 2930 cm-1 2948 cm-).
COSY
……………………………….
US5576318, 1996
1 H NMR (DMSO-d6 /CDCL3 5:2) 11.09 (b, 1H), 11.04 (s, 1H), 7.5-6.9 (SH), 4.36 (t, 2H), 4.1-3.1 (10 H)
,,,,,,,,,,,,,,,,,,
- Borsini F, Evans K, Jason K, Rohde F, Alexander B, Pollentier S (summer 2002). “Pharmacology of flibanserin”. CNS Drug Rev. 8 (2): 117–142. doi:10.1111/j.1527-3458.2002.tb00219.x. PMID 12177684.
- Jolly E, Clayton A, Thorp J, Lewis-D’Agostino D, Wunderlich G, Lesko L (April 2008). “Design of Phase III pivotal trials of flibanserin in female Hypoactive Sexual Desire Disorder (HSDD)”. Sexologies 17 (Suppl 1): S133–4. doi:10.1016/S1158-1360(08)72886-X.
- Spiegel online: Pharmakonzern stoppt Lustpille für die Frau, 8 October 2010 (in German)
- Nygaard I (November 2008). “Sexual dysfunction prevalence rates: marketing or real?”. Obstet Gynecol 112 (5): 968–9.doi:10.1097/01.AOG.0000335775.68187.b2. PMID 18978094.
- Clayton AH (July 2010). “The pathophysiology of hypoactive sexual desire disorder in women”. Int J Gynaecol Obstet 110 (1): 7–11.doi:10.1016/j.ijgo.2010.02.014. PMID 20434725.
- Pfaus JG (June 2009). “Pathways of sexual desire”. J Sex Med 6 (6): 1506–33. doi:10.1111/j.1743-6109.2009.01309.x.PMID 19453889.
- Yves Aubert, Thesis, Leiden University. (Dec 11, 2012) Sex, aggression and pair-bond: a study on the serotonergic regulation of female sexual function in the marmoset monkey
- Viagra for women?
- Marazziti D, Palego L, Giromella A, et al. (June 2002). “Region-dependent effects of flibanserin and buspirone on adenylyl cyclase activity in the human brain”. Int. J. Neuropsychopharmacol. 5 (2): 131–40. doi:10.1017/S1461145702002869.PMID 12135537.
- Podhorna J, Brown RE (June 2000). “Flibanserin has anxiolytic effects without locomotor side effects in the infant rat ultrasonic vocalization model of anxiety”. Br J Pharmacol 130 (4): 739–746. doi:10.1038/sj.bjp.0703364. PMC 1572126.PMID 10864879.
- Brambilla A, Baschirotto A, Grippa N, Borsini F (December 1999). “Effect of flibanserin (BIMT 17), fluoxetine, 8-OH-DPAT and buspirone on serotonin synthesis in rat brain”. Eur Neuropsychopharmacol 10 (1): 63–7. doi:10.1016/S0924-977X(99)00056-5.PMID 10647099.
EP0200322A1 * | Mar 18, 1986 | Nov 5, 1986 | H. Lundbeck A/S | Heterocyclic compounds |
BE904945A1 * | Title not available | |||
GB2023594A * | Title not available | |||
US3472854 * | May 29, 1967 | Oct 14, 1969 | Sterling Drug Inc | 1-((benzimidazolyl)-lower-alkyl)-4-substituted-piperazines |
US4954503 * | Sep 11, 1989 | Sep 4, 1990 | Hoechst-Roussel Pharmaceuticals, Inc. | 3-(1-substituted-4-piperazinyl)-1H-indazoles |
FDA Advisory Committee Recommends Approval of Takeda’s Investigational Biologic Vedolizumab
Deerfield, Ill., December 9, 2013 and Osaka, Japan, December 10, 2013 — Takeda Pharmaceutical Company Limited (“Takeda”) and its wholly-owned subsidiary, Takeda Pharmaceuticals U.S.A., Inc., today announced that a joint panel of members from the Gastrointestinal Drugs and Drug Safety and Risk Management Advisory Committees of the United States (U.S.) Food and Drug Administration (FDA) voted to recommend approval of Takeda’s vedolizumab for the treatment of adults with moderately to severely active ulcerative colitis (UC) and Crohn’s disease (CD). All 21 committee members voted that based on currently available efficacy and safety data, the benefits outweigh the potential risks of vedolizumab to support approval for UC. Specifically, 13 committee members supported approval for UC patients who have failed steroids or immunosuppressants or TNF-α antagonists, while eight committee members supported approval for UC patients who have failed immunosuppressants or TNF-α antagonists (the indicated population would not include patients that failed steroids only). Twenty of the 21 committee members voted to support approval for CD. Specifically, 14 committee members supported approval for CD patients who have failed steroids or immunosuppressants or TNF-α antagonists while six supported approval for CD patients who have failed immunosuppressants or TNF-α antagonists (the indicated population would not include patients that failed steroids only).
read at
About Crohn’s disease and ulcerative colitis
Crohn’s disease (CD) and ulcerative colitis (UC) are the two most common forms of inflammatory bowel disease (IBD), which is marked by inflammation in the lining of the GI tract. CD can impact any part of the digestive tract, and common symptoms may include abdominal pain, diarrhea, rectal bleeding, weight loss, and/or fever. UC impacts the large intestine only, which includes the colon and the rectum. The most common symptoms of UC include abdominal discomfort and blood or pus in diarrhea. There is no known cause for CD or UC, although many researchers believe that the interaction of an outside agent, such as a virus or bacteria, with the body’s immune system may trigger them. No cure exists for CD or UC; the aim of IBD treatments is to induce and maintain remission, or achieve extended periods of time when patients do not experience symptoms.
About vedolizumab
Vedolizumab was developed for the treatment of CD and UC, as a gut-selective, humanized monoclonal antibody that specifically antagonizes the alpha4beta7 (α4β7) integrin, which is expressed on a subset of circulating white blood cells. These cells have been shown to play a role in mediating the inflammatory process in CD and UC. α4β7 binds with a specific adhesion molecule primarily expressed in the intestinal tract. Therefore, vedolizumab, by preventing this interaction, has a gut selective effect.
About Takeda Pharmaceutical Company Limited
Located in Osaka, Japan, Takeda is a research-based global company with its main focus on pharmaceuticals. As the largest pharmaceutical company in Japan and one of the global leaders of the industry, Takeda is committed to strive towards better health for patients worldwide through leading innovation in medicine. Additional information about Takeda is available through its corporate website, http://www.takeda.com.
Vedolizumab is a monoclonal antibody being developed by Millennium Pharmaceuticals, Inc. for the treatment of ulcerative colitis and Crohn’s disease.It binds to integrin α4β7(LPAM-1, lymphocyte Peyer’s patch adhesion molecule 1).[1][2]
The molecule was first identified by Dr. Andrew Lazarovits [1][2] as the murine MLN0002 homologue. His discovery of the mouse equivalent of this antibody—originally applied to anti-rejection strategies in kidney transplantation—was published in the journal Nature in 1996. The drug was then licensed to Millennium Pharmaceuticals of Boston for further development.
As of October 2009, vedolizumab is undergoing Phase III trials.[3] Clinical trials indicate that Vedolizumab was found safe and highly effective for inducing and maintaining clinical remission in patients with moderate to severe ulcerative colitis [3]. Dr. Brian Faegan, head researcher, reported an absence of any instances of progressive multifocal leukoencephalopathy (PML), which is a particularly important finding [4]. It looks like it will be an effective abiologic agent without some of the toxicity issues previously seen with anti-TNF drugs .
It is widely believed now that “vedolizumab can be used either as a first-line treatment or in case of anti-TNF failure”
- Statement On A Nonproprietary Name Adopted By The USAN Council – Vedolizumab, American Medical Association.
- Soler, D; Chapman, T; Yang, LL; Wyant, T; Egan, R; Fedyk, ER (2009). “The binding specificity and selective antagonism of vedolizumab, an anti-alpha4beta7 integrin therapeutic antibody in development for inflammatory bowel diseases”. The Journal of Pharmacology and Experimental Therapeutics 330 (3): 864–75. doi:10.1124/jpet.109.153973. PMID 19509315.
- ClinicalTrials.gov NCT00790933 Study of Vedolizumab (MLN0002) in Patients With Moderate to Severe Crohn’s Disease (GEMINI II)