New Drug Approvals

Home » FDA 2015 » Daiichi Sankyo receives FDA approval for anti-clotting drug Savaysa , EDOXABAN

Daiichi Sankyo receives FDA approval for anti-clotting drug Savaysa , EDOXABAN

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Archives

Categories

Recent Posts

Blog Stats

  • 4,479,880 hits

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,841 other subscribers

add to any

Share

Edoxaban, DU-176b

Edoxaban (DU-176b, trade names Savaysa, Lixiana) is an anticoagulant drug which acts as a direct factor Xa inhibitor. It was developed by Daiichi Sankyo and approved in July 2011 in Japan for prevention of venous thromboembolisms (VTE) following lower-limb orthopedic surgery.[1] It was also approved by the FDA in January 2015 for the prevention of stroke and non–central-nervous-system systemic embolism.[2]

Daiichi Sankyo receives FDA approval for anti-clotting drug Savaysa
Japanese drug-maker Daiichi Sankyo has obtained approval from US Food and Drug Administration (FDA) for its anti-clotting drug Savaysa (edoxaban tablets)..8 JAN 2015

Daiichi Sankyo, APPROVED IN JAPAN as tosylate monohydrate salt in 2011 for the prevention of venous embolism in patients undergoing total hip replacement surgery

for synthesis see….http://www.sciencedirect.com/science/article/pii/S0968089613002642  Bioorganic & Medicinal Chemistry 21 (2013) 2795–2825,  see s[pecific page 2808 for description  ie 14/31 of pdf

WO 2010071121, http://www.google.com/patents/WO2010071121A1

WO 2007032498

N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide

    N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide

Edoxaban (INN, codenamed DU-176b, trade name Lixiana) is an anticoagulant drug which acts as a direct factor Xa inhibitor. It is being developed by Daiichi Sankyo. It was approved in July 2011 in Japan for prevention of venous thromboembolisms (VTE) following lower-limb orthopedic surgery.[1]

 

In animal studies, edoxaban is potent, selective for factor Xa and has good oral bioavailability.[2]

Daichi Sankyo’s edoxaban tosilate is an orally administered
coagulation factor Xa inhibitor that was approved and launched
in Japan for the preventive treatment of venous thromboembolic
events (VTE) in patients undergoing total knee arthroplasty, total
hip arthroplasty, or hip fracture surgery. Edoxaban has been
shown to have a rapid onset of anticoagulant effect due to short
Tmax (1–2 h) after dosing and sustained for up to 24 h post-dose.
Marketed under the brand name Lixiana, it is currently in phase
III studies in the US for the prevention of stroke and systemic embolic
events in patients with atrial fibrillation (AF) and venous
thromboembolism (VTE).

Several Phase II clinical trials have been conducted, for example for thromboprophylaxis after total hip replacement[3] (phase III early results compare well to enoxaparin[4]), and for stroke prevention in patients with atrial fibrillation[5][6].Those papers follow similar recent major trials showing similar results for the other new factor Xa inhibitorsrivaroxaban and apixaban.

A large phase III trial showed that edoxaban was non inferior to warfarin in preventing recurrent venous thromboembolic events with fewer episodes of major bleeding.[7]

……………..

PATENT

http://www.google.com/patents/WO2014081047A1?cl=en

Chemically, edoxaban is

N1– (5-chloropyridin-2-yl) -N2– ( (IS, 2R/4S) -4- [ (dimethylamino) carbo nyl] -2- { [ ( 5-methyl-4 , 5,6, 7-tetrahydrothiazolo [5 , 4-c] pyridin-2-yl ) carbonyl] amino}eyelohexyl) ethanediamide , represented by the following formula (A) :

Figure imgf000003_0002

(A) The p-toluenesulfonic acid monohydrate salt of compound A is represented b the following formula (B) :

Figure imgf000004_0001

(B)

Edoxaban is known as a compound that exhibits an inhibitory effect on activated blood coagulation factor X (also referred to as activated factor X or FXa) , and is useful as a preventive and/or therapeutic drug for thrombotic diseases.

Several processes are known in the literature for preparing edoxaban for example, U.S. Patent No. 7365205; U.S. Publication No . 20090105491.

U.S. Patent No. 7365205 provides a process for the preparation of edoxaban, wherein the process involves the use of

(IS, 4S, 5S) -4-iodo-6-oxabicyclo [3.2.1] octan-7-one, represented by the following formula (C) :

Figure imgf000004_0002

(C)

as an intermediate.

The present inventors have identified that

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one, represented by the following formula (I) :

Figure imgf000005_0001

( I )

could also be used as an intermediate for the preparation of FXa inhibitory compounds like edoxaban. The present inventors have found that replacement of

(IS, 4S, 5S) -4-iodo-6-oxabicyclo [3.2.1] octan-7-one (C) with

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one (I) has a better atom economy and also an impact on cost.

A method for the synthesis of the

(IS, 4S, 5S) -4 -bromo- 6 -oxabicyclo [3.2.1] octan-7-one (I) was reported in Tetrahedron Letters, 51, (2010) Pages 3433-3435 which involves the reaction of ( IS) -cyclohex-3 -ene- 1-carboxylic acid represented by the following formula (II) :

Figure imgf000005_0002

( Π )

with N-bromosuccinimide in the presence of molecular sieves using dichloromethane as a solvent. However, this reaction is carried out in dark over a period of 7 hours and does not provide a pure product .

Tetrahedron, Vol. 28, Pages 3393 -3399 , 1972 provides a process for the preparation of 4 -bromo- 6 -oxabicyclo [3.2.1] octan-7-one which involves the addition of 20% excess of a 2M solution of bromine in chloroform to a stirred solution of cyclohex- 3 -ene- 1-carboxylic acid (0.04 mol) in chloroform (250 mL) in the absence of a base . Extraction with aqueous sodium bicarbonate followed by acidification gave, after extraction with ether and evaporation of the extract, a mixture of cis & trans 3 , 4-dibromocyclohexanecarboxylic acid (6.7 g) and evaporation of the chloroform layer afforded the bromolactone (0.59 g) . It further provides a process for the preparation of

4 -bromo-6 -oxabicyclo [3.2.1] octan-7-one which involves the treating of cyclohex-3-ene-l-carboxylic acid (0.08 mol) dissolved in chloroform (450 mL) with 20% excess bromine in the presence of an equimolar amount of triethylamine (8.1 g) . After extraction of the amine with 2N hydrochloric acid, and work-up, bromolactone (10.7 g) and a mixture of cis & trans 3 , 4 -dibromocyclohexanecarboxylic acid (6.6 g) were obtained.

Tetrahedron Vol. 48, No. 3, Pages 539-544, 1992 provides a process for the preparation of

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one (I) which involves the addition of 1M solution of bromine in chloroform (30 mL) at 0°C to a solution of ( IS) -cyclohex-3 -ene- 1-carboxylic acid (0.024 mol) of formula (II) in chloroform (600 mL) in the presence of an equimolar amount of triethylamine (3.33 mL) . After work-up, the crude bromolactone obtained was recrystallized from petroleum ether.

However, bromination using bromine does not provide a pure product in good yield.

Heterocycles, Vol. 23, No. 8, Pages 2035-2039, 1985 provides a process for the 4-bromo-6-oxabicyclo [3.2.1] octan-7-one which involves the addition of cyclohex-3-ene-l-carboxylic acid (1.0 mM) in 1 , 2 -dimethoxyethane (2 mL) to a stirred solution of 90% Lead (IV) acetate (1.1 or 2.2 mM) in 1 , 2 -dimethoxyethane (4 mL) followed by the addition of Zinc bromide (2.2 mM) in 1 , 2 -dimethoxyethane (4 mL) and continuing the stirring for 10-30 minutes at 0°C . The reaction mixture was poured into a solution of ice-cold water (30 mL) and 10% hydrochloric acid (10 mL) , and extracted with ether (50 mL X 3) . The combined ether extract was washed successively with saturated sodium hydrogen carbonate solution (20 mL) , 10% sodium thiosulphate solution (5 mL) , and brine (10 mL) , and dried over sodium sulphate. Evaporation of the solvent gave crude lactone which were separated and purified (42% yield) . However, this reaction does not provide a pure product in good yield.

Heterocycles, Vol. 31, No. 6, Pages 987-991, 1990 provides a method for bromolactonization using a

dimethylsulfoxide-trimethylsilyl bromide-amine system. The bromolactonization is carried out for 10 to 72 hours using different solvents and triethylamine or diisopropylethyl amine as base. However, this process does not provide a product in high yield. Further the process afforded the cis isomer exclusively. Journal of the Chemical Society, Perkin Transactions 1:

Organic and Bio-Organic Chemistry (1972-1999) (1994) , (7) , Pages 847-851 provides a method for bromolactonization using a

dimethylsulfoxide-trimethylsilyl bromide-amine system. The bromolactonization is carried out for 12 hours using

dimethylsulfoxide and chloroform solvent system and triethylamine or diisopropylethyl amine as base. However, this process resulted in a low yield of about 55%. Citation List

Patent Literature

PTLl: U.S. Patent No. 7365205

PTL2: U.S. Publication No. 20090105491.

Non Patent Reference

NPLl: Feng Chen et al . , Tetrahedron Letters, 51, (2010) Pages 3433-3435.

NPL2 : G. Belluci et al . , Tetrahedron, Vol. 28, No. 13, Pages 3393-3399, 1972.

NPL3 : Marco Chini et al ., Tetrahedron Vol .48, No. 3, Pages 539-544 , 1992.

NPL4 : Y. Fujimoto et al . , Heterocycles , Vol. 23, No. 8, Pages 2035-2039, 1985.

NPL5: C. Iwata et al . , Heterocycles, Vol. 31, No. 6, Pages 987-991, 1990. –

NPL6 : K. Miyashita et al . , Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999) (1994) , (7) , Pages 847-851.

Summary of Invention

Technical Problem

It is an object of the present invention to solve the problems associated with the prior art, and to provide an improved and efficient method for the preparation of

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one of formula (I).

Solution to Problem As a result of conducting diligent studies to attain the object, the present inventors have found that: surprisingly, the use of N-bromosuccinimide or bromohydantoin (representative is

1, 3-dibromo-5, 5-dimethylhydantoin) as brominating agent in the presence of a base selected from calcium oxide or calcium hydroxide, in specific mole ratios in a solvent selected from the group consisting of dichloromethane , toluene, tetrahydrofuran, ethyl acetate, hexanes, cyclopentyl methyl ether (CPME) or a mixture thereof can efficiently produce a pure

( IS , 4S , 5S) -4 -bromo- 6 -oxabicyclo [3.2.1] octan- 7 -one (I) in better yields. The process provides obvious benefits with respect to economics, convenience to operate at a commercial scale.

…………………………..

SEE

http://www.google.co.ug/patents/US20090105491

………………………….

PATENT

http://www.google.com/patents/EP2589590A1?cl=en

FREE BASE

      (Reference Example 6) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0052
  • Methanesulfonic acid (66 ml) was added to a suspension of tert-butyl [(1R,2,S,5S)-2-({[(5-chloropyridin-2-yl)amino](oxo)acetyl}amino)-5-(dimethylaminocarbonyl)cyclohexyl]carbamate (5) (95.1 g) in acetonitrile (1900 ml) at room temperature, and the mixture was stirred at this temperature for 2 hours. To the reaction solution, triethylamine (155 ml), 5-methyl-4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyrzdine-2-carboxylic acid hydrochloride (8) (52.5 g), 1-hydroxybenzotriazole (33.0 g), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (46.8 g) were added under ice cooling, and the mixture was stirred at room temperature for 16 hours. Triethylamine and water were added thereto, and the mixture was stirred for 1 hour under ice cooling. Then, crystals were collected by filtration to obtain the title compound (X) (103.2 g). 1H-NMR (CDCl3) δ : 1.60-1.98 (3H, m), 2.00-2.16 (3H, m), 2.52 (3H, s), 2.78-2.90 (3H, m), 2.92-2.98 (2H, m), 2.95 (3H, s), 3.06 (3H, s), 3.69 (1H, d, J = 15.4 Hz), 3.75 (1H, d, J = 15.4 Hz), 4.07-4.15 (1H, m), 4.66-4.72 (1H, m), 7.40 (1H, dd, J = 8.8, 0.6 Hz), 7. 68 (1H, dd, J = 8.8, 2.4 Hz), 8.03 (1H, d, J = 7.8 Hz), 8.16 (1H, dd, J = 8.8, 0.6 Hz), 8.30 (1H, dd, J = 2. 4, 0.6 Hz), 9.72 (1H, s). MS (ESI) m/z: 548 (M+H)+.

TOSYLATE

      (Reference Example 7) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide mono-p-toluenesulfonate monohydrate (X-a) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0053
  • N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (6.2 g) was dissolved in methylene chloride (120 ml). To the solution, a 1 mol/L solution of p-toluenesulfonic acid in ethanol (11.28 ml) was added, and the solvent was distilled off. To the residue, 15% hydrous ethanol (95 ml) was added, and the mixture was dissolved by stirring at 60°C. Then, the mixture was cooled to room temperature and stirred for 1 day. The precipitated crystals were collected by filtration, washed with ethanol, and then dried under reduced pressure at room temperature for 2 hours to obtain the title compound (X-a) (7.4 g).
    1H-NMR (DMSO-d6) δ : 1. 45-1. 54 (1H, m), 1.66-1.78 (3H, m), 2.03-2.10 (2H, m), 2.28 (3H, s), 2.79 (3H, s), 2.91-3.02 (1H, m), 2.93 (3H, s), 2.99 (3H, s), 3.13-3.24 (2H, m), 3.46-3.82 (2H, m), 3.98-4.04 (1H, m), 4.43-4.80 (3H, m), 7.11 (2H, d, J = 7.8 Hz), 7.46 (2H, d, J = 8.2 Hz), 8.01 (2H, d, J = 1.8 Hz), 8.46 (1H, t, J = 1.8 Hz), 8.75 (1H, d, J = 6.9 Hz), 9.10-9.28 (1H, br), 10.18 (1H, br), 10.29 (1H, s).
    MS (ESI) m/z: 548 (M+H)+.
    Anal.: C24H30ClN7O4S·C7H8O3S·H2O
    Theoretical: C; 50.43, H; 5.46, N; 13.28, Cl; 4.80, S; 8.69.
    Found: C; 50.25, H; 5.36, N; 13.32, Cl; 4.93, S; 8.79. mp (dec.): 245-248°C.

……………………………………………..

PATENT

http://www.google.com/patents/EP2589590A1?cl=en

    • A compound represented by the following formula (X) [hereinafter, also referred to as compound (X)] or a pharmacologically acceptable salt thereof, or a hydrate thereof is a compound that exhibits an FXa inhibitory effect, as disclosed in Patent Literatures 1 to 3, and is useful as a preventive and/or therapeutic drug for thrombotic and/or embolic diseases:
    • Figure imgb0001
    • The pamphlet of International Publication No. WO 2007/032498discloses a process for preparing an FXa inhibitor compound (X) or a pharmacologically acceptable salt thereof, or a hydrate thereof. The process for producing compound (X) disclosed therein involves, as shown in [Scheme A] below, azidifying compound (2) to produce azide compound (3), subsequently reducing compound (3) into amino compound (1a), subsequently treating compound (1a) with anhydrous oxalic acid to obtain compound (1), which is then treated with compound (4) (ethyl[5-chloropyridin-2-yl]amino](oxo)acetate hydrochloride) in the presence of a base to produce compound (5), followed by several steps from compound (5). This pamphlet also discloses crystals of the oxalate of compound (1) as a production intermediate.
    • Figure imgb0002
    • wherein Boc represents a tert-butoxycarbonyl group.

Citation ListPatent Literatures

  1.  Furugohri T, Isobe K, Honda Y, Kamisato-Matsumoto C, Sugiyama N, Nagahara T, Morishima Y, Shibano T (September 2008). “DU-176b, a potent and orally active factor Xa inhibitor: in vitro and in vivo pharmacological profiles”. J. Thromb. Haemost.6 (9): 1542–9. doi:10.1111/j.1538-7836.2008.03064.xPMID18624979.
  2.  Raskob, G.; Cohen, A. T.; Eriksson, B. I.; Puskas, D.; Shi, M.; Bocanegra, T.; Weitz, J. I. (2010). “Oral direct factor Xa inhibition with edoxaban for thromboprophylaxis after elective total hip replacement”. Thrombosis and Haemostasis104 (3): 642–649. doi:10.1160/TH10-02-0142.PMID20589317edit
  3. “Phase III Trial Finds Edoxaban Outclasses Enoxaparin in Preventing Venous Thromboembolic Events”. 8 Dec 2010.
  4.  Weitz JI, Connolly SJ, Patel I, Salazar D, Rohatagi S, Mendell J, Kastrissios H, Jin J, Kunitada S (September 2010). “Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation”. Thromb. Haemost.104 (3): 633–41. doi:10.1160/TH10-01-0066.
  5.  Edoxaban versus Warfarin in Patients with Atrial Fibrillation Robert P. Giugliano, M.D., Christian T. Ruff, M.D., M.P.H., Eugene Braunwald, M.D., Sabina A. Murphy, M.P.H., Stephen D. Wiviott, M.D., Jonathan L. Halperin, M.D., Albert L. Waldo, M.D., Michael D. Ezekowitz, M.D., D.Phil., Jeffrey I. Weitz, M.D., Jindřich Špinar, M.D., Witold Ruzyllo, M.D., Mikhail Ruda, M.D., Yukihiro Koretsune, M.D., Joshua Betcher, Ph.D., Minggao Shi, Ph.D., Laura T. Grip, A.B., Shirali P. Patel, B.S., Indravadan Patel, M.D., James J. Hanyok, Pharm.D., Michele Mercuri, M.D., and Elliott M. Antman, M.D. for the ENGAGE AF-TIMI 48 InvestigatorsDOI: 10.1056/NEJMoa1310907
  6.  “Edoxaban versus Warfarin for the Treatment of Symptomatic Venous Thromboembolism”. N. Engl. J. Med. August 2013. doi:10.1056/NEJMoa1306638PMID23991658.
  7. WO 03/000657 pamphlet WO 03/000680 pamphlet WO 03/016302 pamphlet WO 04/058715 pamphlet WO 05/047296 pamphlet WO 07/032498 pamphlet WO 08/129846 pamphlet WO 08/156159 pamphlet
  8. J Am Chem Soc 1978, 100(16): 5199
[1] 王利华, 赵丽嘉, 李文利, 等. 直接抑制凝血因子Xa 的口服抗凝药物Edoxaban Tosilate Hydrate [J]. 药物评价研究, 2011, 34(6): 478-481.
[2] Ohta T, Komoriya S, Yoshino T, et al. Preparation of N,N’-bis( heterocyclicacyl) cycloalkanediamine and heterocyclediamine derivatives as inhibitors of activated blood coagulation factor X (factor Xa): WO, 2003 000657 [P]. 2003-01-03. (CA 2003, 138: 73271)
[3] Ohta T, Komoriya S, Yoshino T, et al. Preparation of heterocyclic moiety-containing diamine derivatives as FXa inhibitors: WO, 2003 000680 [P]. 2003-01-03. (CA 2003, 138: 89801)
[4] Mochizuki A, Nagata T. Triamine derivative: WO, 2006106963 [P]. 2005-03-31. (CA 2006, 145: 419128)
[5] Kawanami K, Ishikawa H, Shoji M. Process for preparation of optically active (1S,3R,4R)-3-amino-4-hydroxy-N,Ndimethylcyclohexanecarboxamide derivative salt: WO, 2012002538 [P]. 2012-01-05. (CA 2012, 156: 122056)
[6] Sato K, Kubota K. Process for producing optically active carboxylic acid: WO, 2010067824 [P]. 2010-06-17. (CA 2010, 153: 36882)
[7] Yoshikawa K, Yokomizo A, Naito H, et al. Design, synthesis, and SAR of cis-1,2-diaminocyclohexane derivatives as potent factor Xa inhibitors. Part I: Exploration of 5-6fused rings
[8] as alternative S1 moieties [J]. Bioorg Med Chem, 2009, 17(24): 8206-8220.
[9] Sato K, Kawanami K, Yagi T. Process for the preparation of optically active cyclohexane-1,2-diamine derivative from 7-oxabicyclo[4.1.0]heptane compound: WO, 2007032498
[10] 2007-03-22. (CA 2007, 146: 358502)
[11] Kawanami K. Method for the preparation of optically active diamine derivative: WO, 2010104106 [P]. 2010-09-16. (CA 2010, 153: 406061)
[12] Koyama T, Kondo S. Process for the preparation of diamine derivative: WO, 2010104078 [P]. 2010-09-16. (CA 2010, 153: 382938)
[13] Suzuki T, Ono M. Crystal of diamine derivative and method of producing same: WO, 2011115066 [P]. 2011-09-22. (CA 2011, 155: 467954)

 

US8357808 9 Sep 2011 22 Jan 2013 Daiichi Sankyo Company, Limited Process for producing diamine derivative
US8394821 13 Jul 2011 12 Mar 2013 Daiichi Sankyo Company, Limited Activated blood coagulation factor inhibitor
US8404847 17 Jun 2011 26 Mar 2013 Daiichi Sankyo Company, Limited Method for producing diamine derivative
US8449896 16 Dec 2011 28 May 2013 Daiichi Sankyo Company, Limited Pharmaceutical composition having improved solubility
US8541443 19 Sep 2012 24 Sep 2013 Daiichi Sankyo Company, Limited Crystal of diamine derivative and method of producing same
US20130004550 * 22 Aug 2012 3 Jan 2013 Daiichi Sankyo Company, Limited Sustained-release solid preparation for oral use
WO2014081047A1 22 Nov 2013 30 May 2014 Daiichi Sankyo Company,Limited Process for the preparation of (1s,4s,5s)-4-bromo-6-oxabicyclo[3.2.1] octan-7-one

Molecular Formula C24H30ClN7O4S.C7H7HSO3
Molecular Weight 720.26
CAS Registry Number 480449-71-6 (912273-65-5)

Drug formulation , lixiana, edoxaban tosylate monohydrate, CAS 912273-65-5, C24 H30 Cl N7 O4 S . C7 H8 O3 S . H2 O, 738.274

    • N1-(5-chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide p-toluenesulfonic acid monohydrate represented by the following formula (A) (hereinafter, also referred to as compound A) :
    • Figure imgb0001
      Figure imgb0002
    • is known as a compound that exhibits an inhibitory effect on activated blood coagulation factor X (FXa), and is useful as a preventive and/or therapeutic drug for thrombotic diseases (Patent Literature 1 to 8).
    • For example, a method comprising mixing the free form of compound A represented by the following formula (B) (hereinafter, also referred to as compound B):
    • Figure imgb0003
    • with p-toluenesulfonic acid or p-toluenesulfonic acid monohydrate, followed by crystallization from aqueous ethanol, is known as a method for obtaining compound A (Patent Literature 1 to 8). These literature documents do not make any mention about adding p-toluenesulfonic acid or p-toluenesulfonic acid monohydrate in a stepwise manner in the step of obtaining compound A from compound B.

Citation ListPatent Literature

    • Patent Literature 1: International Publication No. WO 03/000657
    • Patent Literature 2: International Publication No. WO 03/000680
    • Patent Literature 3: International Publication No. WO 03/016302
    • Patent Literature 4: International Publication No. WO 04/058715
    • Patent Literature 5: International Publication No. WO 05/047296
    • Patent Literature 6: International Publication No. WO 07/032498
    • Patent Literature 7: International Publication No. WO 08/129846
    • Patent Literature 8: International Publication No. WO 08/156159

SIMILAR

OTHER SALTS

Edoxaban hydrochloride
CAS Number: 480448-29-1
Molecular Formula: C24H30ClN7O4S · HCl
Molecular Weight: 584.52 g.mol-1

Edoxaban is reported to be a member of the so-called “Xaban-group” and as such to be a low molecular inhibitor of the enzyme factor Xa, participating in the blood coagulation system. Therefore, edoxaban is classified as an antithrombotic drug and its possible medical indications are reported to be treatment of thrombosis and thrombosis prophylaxis after orthopaedic operations, such as total hip replacement, as well as for stroke prevention in patients with atrial fibrillation, the prophylaxis of the acute coronary syndrome and the prophylaxis after thrombosis and pulmonary embolism.

The IUPAC name for edoxaban is N’-(5-chloropyridin-2-yl)-N-[(15,2^,4S)-4- (dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[l ,3]thiazolo[5,4-c]pyridine-2- carbonyl)amino]cyclohexyl]oxamide. The chemical structure of edoxaban is shown in the formula (1) below:

Figure imgf000002_0001

formula ( 1 ) While Edoxaban is reported to be soluble in strongly acidic aqueous solutions, its solubility is considered to be very low in neutral or alkaline aqueous media. EP 2 140 867 A 1 claims an edoxaban-containing pharmaceutical composition comprising a water-swelling additive and/or a sugar alcohol. Further, it is alleged that compositions comprising lactose or cornstarch do not have good dissolution properties. The claimed pharmaceutical compositions in EP 2 140 867 Al are considered to show good dissolution properties in a neutral aqueous medium as well. Tablets comprising said composition were produced by wet granulation. However, it turned out that prior art pharmaceutical formulations comprising edoxaban being suitable for oral administration are still improvable with regards to dissolution rate and bioavailability. Further, stability and content uniformity of the known formulations could be improved. Further, due to the intolerance of many people to sugar alcohol(s), such as sorbitol, the use of sugar alcohol(s) should be avoided.

UPDATE

2-amino-5-methyl-4,5,6,7-tetrahydro thiazolone [5,4-c] pyridine 

WO2015125710

(Reference Example 1) 2-amino-5-methyl-4,5,6,7-tetrahydro thiazolone [5,4-c] pyridine (1-n) (The method described in WO 2005/047296 Pamphlet )
[0091]
[Of 35]  in 2-PrOH (1.44L) solution was heated to 50 ℃ 1- methyl-4-piperidone (180.0g), 2-PrOH (360mL) solution of cyanamide (67.0g), and sulfur powder (51.0 g) it was added. Pyrrolidine (13.3mL) was added to the reaction mixture, after stirring for 2 hours at 50 ℃, followed by stirring overnight and allowed to cool to room temperature.  The reaction mixture was cooled to 10 ℃ less in an ice water bath and stirred for 1 hour at the same temperature. Is filtered and the precipitated crystals were washed with 2-PrOH (540mL), the title compound was dried under reduced pressure at 40 ℃ (209.9g, 78%) was obtained.

[0092]
1 H-NMR (CDCl 3 ) ppm: 4.86 (Br, 2H), 3.47-3.46 (t, 2H, J = 1.9 Hz), 2.78-2.71 (M, 2H), 2.71-2.65 (M, 2H), 2.47 . (s, 3H)
MS (FAB) M / z: 170 (M + H) +
elemental analysis: C 7 H 11 N 3 as S,
theoretical value: C, 49.68; H, 6.55; N, 24.83; S, 18.95
measured value: C, 49.70; H, 6.39; N, 24.91; S, 19.00.

WO2015125710

 (Example 11) 1 – (5-Chloro-2-yl) -N 2 – [(1S, 2R, 4S)-4-(dimethylcarbamoyl) -2 – {[(5-methyl-4,5 , 6,7-tetrahydro [1,3] thiazolo [5,4-c] pyridin-2-yl) carbonyl] amino} cyclohexyl] Etanjiamido (X) [Production method via Compound (1-p2)]
[0137]
 In 10 mL test tube, compound (5-ms) (the compound of Reference Example 8) (100 mg, 0.216 mmol), Compound (1-p2) (81.4 mg, 0.216 mmol), K 3 PO 4 (91.7 mg, 0.432 mmol) and DMF (1 mL) was added, and the mixture was stirred at room temperature conditions for 3 hours. H To the reaction mixture 2 O (2 mL) was added and the resulting slurry was stirred at room temperature overnight, the solid was filtered. The resulting solid H 2 was washed with O (1 mL), was obtained by drying under reduced pressure the title compound (110.0 mg, 92.9%) as a solid.
[0138]
1 H-NMR (500 Hz, CDCl 3 ) delta: 9.72 (s, 1H), 8.30 (dd, 1H, J = 2.5, 0.5 Hz), 8.17 (dd, 1H, J = 9.0, 0.5 Hz), 8.03 (D , 1H, J = 8.5 Hz), 7.68 (dd, 1H, J = 9.0, 2.5 Hz), 7.39 (d, 1H, J = 8.5 Hz), 4.70-4.67 (m, 1H), 4.13-4.09 (m, 1H), 3.73 (d, 1H, J = 16.0 Hz), 3.70 (d, 1H, J = 16.0 Hz), 3.06 (s, 3H), 2.96-2.93 (m, 2H), 2.95 (s, 3H), 2.89-2.79 (m, 3H), 2.52 (s, 3H), 2.14-2.06 (m, 3H), 1.96-1.90 (m, 1H), 1.84-1.78 (m, 1H), 1.69-1.62 (m, 1H ).

UPDATE

Edoxaban, DU-176b

1H NMR PREDICTION

edoxaban NMR spectra analysis, Chemical CAS NO. 480449-70-5 NMR spectral analysis, edoxaban H-NMR spectrum

………….

13 C NMR

edoxaban NMR spectra analysis, Chemical CAS NO. 480449-70-5 NMR spectral analysis, edoxaban C-NMR spectrum

FREE BASE

      (Reference Example 6) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0052

  • Methanesulfonic acid (66 ml) was added to a suspension of tert-butyl [(1R,2,S,5S)-2-({[(5-chloropyridin-2-yl)amino](oxo)acetyl}amino)-5-(dimethylaminocarbonyl)cyclohexyl]carbamate (5) (95.1 g) in acetonitrile (1900 ml) at room temperature, and the mixture was stirred at this temperature for 2 hours. To the reaction solution, triethylamine (155 ml), 5-methyl-4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyrzdine-2-carboxylic acid hydrochloride (8) (52.5 g), 1-hydroxybenzotriazole (33.0 g), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (46.8 g) were added under ice cooling, and the mixture was stirred at room temperature for 16 hours. Triethylamine and water were added thereto, and the mixture was stirred for 1 hour under ice cooling. Then, crystals were collected by filtration to obtain the title compound (X) (103.2 g).

  • 1H-NMR (CDCl3) δ : 1.60-1.98 (3H, m), 2.00-2.16 (3H, m), 2.52 (3H, s), 2.78-2.90 (3H, m), 2.92-2.98 (2H, m), 2.95 (3H, s), 3.06 (3H, s), 3.69 (1H, d, J = 15.4 Hz), 3.75 (1H, d, J = 15.4 Hz), 4.07-4.15 (1H, m), 4.66-4.72 (1H, m), 7.40 (1H, dd, J = 8.8, 0.6 Hz), 7. 68 (1H, dd, J = 8.8, 2.4 Hz), 8.03 (1H, d, J = 7.8 Hz), 8.16 (1H, dd, J = 8.8, 0.6 Hz), 8.30 (1H, dd, J = 2. 4, 0.6 Hz), 9.72 (1H, s).

  • MS (ESI) m/z: 548 (M+H)+.


Molecular Formula C24H30ClN7O4S.C7H7HSO3
Molecular Weight 720.26
CAS Registry Number 480449-71-6 (912273-65-5)

TOSYLATE

      (Reference Example 7) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide mono-p-toluenesulfonate monohydrate (X-a) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0053

  • N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (6.2 g) was dissolved in methylene chloride (120 ml). To the solution, a 1 mol/L solution of p-toluenesulfonic acid in ethanol (11.28 ml) was added, and the solvent was distilled off. To the residue, 15% hydrous ethanol (95 ml) was added, and the mixture was dissolved by stirring at 60°C. Then, the mixture was cooled to room temperature and stirred for 1 day. The precipitated crystals were collected by filtration, washed with ethanol, and then dried under reduced pressure at room temperature for 2 hours to obtain the title compound (X-a) (7.4 g).

  • 1H-NMR (DMSO-d6) δ : 1. 45-1. 54 (1H, m), 1.66-1.78 (3H, m), 2.03-2.10 (2H, m), 2.28 (3H, s), 2.79 (3H, s), 2.91-3.02 (1H, m), 2.93 (3H, s), 2.99 (3H, s), 3.13-3.24 (2H, m), 3.46-3.82 (2H, m), 3.98-4.04 (1H, m), 4.43-4.80 (3H, m), 7.11 (2H, d, J = 7.8 Hz), 7.46 (2H, d, J = 8.2 Hz), 8.01 (2H, d, J = 1.8 Hz), 8.46 (1H, t, J = 1.8 Hz), 8.75 (1H, d, J = 6.9 Hz), 9.10-9.28 (1H, br), 10.18 (1H, br), 10.29 (1H, s).

    MS (ESI) m/z: 548 (M+H)+.

    Anal.: C24H30ClN7O4S·C7H8O3S·H2O

    Theoretical: C; 50.43, H; 5.46, N; 13.28, Cl; 4.80, S; 8.69.

    Found: C; 50.25, H; 5.36, N; 13.32, Cl; 4.93, S; 8.79. mp (dec.): 245-248°C.

1H NMR PREDICTION, TOSYLATE

CAS NO. 1229194-11-9, N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate H-NMR spectral analysis

N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate NMR spectra analysis, Chemical CAS NO. 1229194-11-9 NMR spectral analysis, N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate H-NMR spectrum

13 CNMR PREDICTION, TOSYLATE

CAS NO. 1229194-11-9, N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate C-NMR spectral analysis

N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate NMR spectra analysis, Chemical CAS NO. 1229194-11-9 NMR spectral analysis, N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate C-NMR spectrum

………………

WO2015125710

 (Example 11) 1 – (5-Chloro-2-yl) -N 2 – [(1S, 2R, 4S)-4-(dimethylcarbamoyl) -2 – {[(5-methyl-4,5 , 6,7-tetrahydro [1,3] thiazolo [5,4-c] pyridin-2-yl) carbonyl] amino} cyclohexyl] Etanjiamido (X) [Production method via Compound (1-p2)]

[0137]

 In 10 mL test tube, compound (5-ms) (the compound of Reference Example 8) (100 mg, 0.216 mmol), Compound (1-p2) (81.4 mg, 0.216 mmol), K 3 PO 4 (91.7 mg, 0.432 mmol) and DMF (1 mL) was added, and the mixture was stirred at room temperature conditions for 3 hours. H To the reaction mixture 2 O (2 mL) was added and the resulting slurry was stirred at room temperature overnight, the solid was filtered. The resulting solid H 2 was washed with O (1 mL), was obtained by drying under reduced pressure the title compound (110.0 mg, 92.9%) as a solid.

[0138]

1 H-NMR (500 Hz, CDCl 3 ) delta: 9.72 (s, 1H), 8.30 (dd, 1H, J = 2.5, 0.5 Hz), 8.17 (dd, 1H, J = 9.0, 0.5 Hz), 8.03 (D , 1H, J = 8.5 Hz), 7.68 (dd, 1H, J = 9.0, 2.5 Hz), 7.39 (d, 1H, J = 8.5 Hz), 4.70-4.67 (m, 1H), 4.13-4.09 (m, 1H), 3.73 (d, 1H, J = 16.0 Hz), 3.70 (d, 1H, J = 16.0 Hz), 3.06 (s, 3H), 2.96-2.93 (m, 2H), 2.95 (s, 3H), 2.89-2.79 (m, 3H), 2.52 (s, 3H), 2.14-2.06 (m, 3H), 1.96-1.90 (m, 1H), 1.84-1.78 (m, 1H), 1.69-1.62 (m, 1H ).

References

  1. “First market approval in Japan for LIXIANA (Edoxaban)”. Press Release. Daiichi Sankyo Europe GmbH. 2011-04-22.
  2. O’Riordan, Michael (9 January 2015). “FDA Approves Edoxaban for Stroke Prevention in AF and DVT/PE Prevention”. Medscape. Retrieved 10 January 2015.
  3. http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/206316lbl.pdf
  4. lexicomp.com
  5. Savaysa (edoxaban) [prescribing information]. Parsippany, NJ: Daiichi Sankyo; January 2015.
  6. http://www.drugs.com/cons/edoxaban.html
  7. Yoshiyuki, I., et al. “Biochemical and pharmalogical profile of darexaban, an oral direct Xa inhibitor.” European Journal of Pharmacology (2011): 49-55
  8. Katsung, B., S. Masters and A. Trevor. Basic and Clinical Pharmacology 11th Edition. United States of America: McGraw-Hill, 2009
  9. Turpie AG (January 2008). “New oral anticoagulants in atrial fibrillation”. European Heart Journal 29 (2): 155–65. doi:10.1093/eurheartj/ehm575. PMID 18096568.

Edoxaban, a factor Xa inhibitor, is supplied as edoxaban tosylate monohydrate. The chemical name is N-(5-Chloropyridin-2-yl)-N’-[(1S,2R,4S)-4-(N,N-dimethylcarbamoyl)-2-(5-methyl- 4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyridine-2-carboxamido)cyclohexyl] oxamide mono (4- methylbenzenesulfonate) monohydrate. Edoxaban tosylate monohydrate has the empirical formula C24H30ClN7O4S•C7H8O3S•H2O representing a molecular weight of 738.27. The chemical structure of edoxaban tosylate monohydrate is:

SAVAYSA (edoxaban) Structural Formula Illustration

It is a white to pale yellowish-white crystalline powder. The solubility of edoxaban tosylate (pKa 6.7) decreases with increasing pH. It is slightly soluble in water, pH 3 to 5 buffer, very slightly soluble at pH 6 to 7; and practically insoluble at pH 8 to 9.

SAVAYSA is available for oral administration as a 60 mg, 30 mg, or 15 mg round shaped, film-coated tablet, debossed with product identification markings. Each 60 mg tablet contains 80.82 mg edoxaban tosylate monohydrate equivalent to 60 mg of edoxaban. Each 30 mg tablet contains 40.41 mg edoxaban tosylate monohydrate equivalent to 30 mg of edoxaban. Each 15 tablet contains 20.20 mg edoxaban tosylate monohydrate equivalent to 15 mg of edoxaban.

The inactive ingredients are: mannitol, pregelatinized starch, crospovidone, hydroxypropyl cellulose, magnesium stearate, talc, and carnauba wax. The color coatings contain hypromellose, titanium dioxide, talc, polyethylene glycol 8000, iron oxide yellow (60 mg tablets and 15 mg tablets), and iron oxide red (30 mg tablets and 15 mg tablets).

Edoxaban
Edoxaban.svg
Systematic (IUPAC) name
N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide
Clinical data
Trade names Lixiana, Savaysa
AHFS/Drugs.com Monograph
Pregnancy
category
  • US: C (Risk not ruled out)
Legal status
Routes of
administration
Oral
Pharmacokinetic data
Bioavailability 62%; Tmax 1–2 hours
Protein binding 55%
Metabolism Minimal hepatic
Biological half-life 10–14 hours
Excretion 50% renal; <50% bile
Identifiers
CAS Registry Number 912273-65-5 Yes
ATC code None
PubChem CID: 25022378
IUPHAR/BPS 7575
ChemSpider 8456212 
UNII NDU3J18APO 
KEGG D09710 
ChEBI CHEBI:85973 Yes
Chemical data
Formula C24H30ClN7O4S
Molecular mass 548.056 g/mol

/////////

SEE ABAN SERIES AT…………http://organicsynthesisinternational.blogspot.in/p/aban-series.html

 

 


Leave a comment

This site uses Akismet to reduce spam. Learn how your comment data is processed.

DR ANTHONY CRASTO

Follow New Drug Approvals on WordPress.com

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,841 other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Personal Links

View Full Profile →

bloglovin

Follow my blog with Bloglovin The title of your home page You could put your verification ID in a comment Or, in its own meta tag Or, as one of your keywords Your content is here. The verification ID will NOT be detected if you put it here.