DR ANTHONY MELVIN CRASTO,WorldDrugTracker, helping millions, A 90 % paralysed man in action for you, I am suffering from transverse mylitis and bound to a wheel chair, With death on the horizon, this will not stop me, Only God and death can..........
DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai), INDIA, worlddrugtracker, 29Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.8 Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution
n, सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये।औकात बस इतनी देना,कि औरों का भला हो जाये।...........P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Thursday 3 December 2015

BETRIXABAN



N-(5-chloropyridin-2-yl)-2-([4-(N,N-dimethylcarbamimidoyl)benzoyl]amino)-5-methoxybenzamide

Betrixaban:PRT-54021, PRT-021, MK-4448, PRT-054021

N- (5- chloro-2-pyridyl) -2 – [[4 – [(dimethylamino) methyl] benzoyl] amino] -5 – methoxy – benzamide
CAS 330942-05-7
MW 451.91, C23H22ClN5O3
Venous Thromboembolism (VTE)
Millennium INNOVATOR
Takeda Pharmaceutical Co Ltd

Lee’s Pharmaceutical Holdings (Hong Kong) Ltd; Portola Pharmaceuticals Inc…DEVELOPERS

PHASE 3  for Venous Thromboembolism (VTE)


Patents CN1391555A, CN102336702A, CN101595092A, CN102762538A

Portola Pharmaceuticals, under license from Takeda (formerly known as Millennium Pharmaceuticals), is developing betrixaban (was reported to be in phase III in November 2015), for treating venous thrombosis
In October 2015, betrixaban was granted Fast Track designation by the FDA for extended-duration prevention of VTE or blood clots in acute medically ill patients

Betrixaban (INN, codenamed PRT-054,021) is an anticoagulant drug which acts as a direct factor Xa inhibitor.[1] It is potent, orally active and highly selective for factor Xa, being selected from a group of similar compounds for its low hERG affinity.[2] Betrixaban has undergone human clinical trials for prevention of embolism after knee surgery,[3] and prevention of stroke following atrial fibrillation,[4] with promising results.[5] Betrixaban is currently being studied in a human clinical trial for extended duration thromboprophylaxis to prevent venous thromboembolism in acute medically ill patients.[6] Joint development with Portola was discontinued in 2011 by Merck.[7] Betrixaban is now being developed by Portola Pharmaceuticals.


Long-acting, oral, direct Factor Xa Inhibitor

Description

Betrixaban is an oral small molecule anticoagulant that directly inhibits the activity of Factor Xa, an important validated target in the blood coagulation pathway.

Key Characteristics

Betrixaban has been specifically designed for chronic, once-a-day treatment. It has a half-life that supports true, once-daily dosing and a low peak-to-trough drug concentration ratio that minimizes anticoagulant variability. Betrixaban is primarily eliminated unchanged in the bile and has been studied in patients with all degrees of renal function, including those with severe renal impairment (excluding dialysis patients). Betrixaban is minimally metabolized through the Cytochrome 450 enzyme system, which may result in low potential for CYP-related drug interactions. Betrixaban is reversible with PRT4445, a universal Factor Xa inhibitor antidote that Portola is developing as a companion product.

Potential Indications

Treatment or prevention of life-threatening blood clots (venous thromboembolism; VTE) in acute medically ill patients.

Clinical Development

ClinicalTrials.gov Identifier:
NCT01583218
COMPLETION-August 2014
http://clinicaltrials.gov/ct2/show/NCT01583218
APEX Study
Portola has initiated its pivotal Phase 3 APEX Study to demonstrate the safety and efficacy of betrixaban for extended duration venous thromboembolism (VTE) prophylaxis for up to 35 days in acute medically ill patients with restricted mobility and certain risk factors. This randomized, double-blind, active-controlled, multicenter, multinational study will compare a once-daily dose of 80 mg of betrixaban for a total of 35 days (including both in the hospital and after discharge) with in-hospital administration of 40 mg of enoxaparin once daily for 6 to 14 days followed by placebo. The global study is expected to enroll approximately 6,850 patients at more than 400 study sites throughout the world. The primary objective of the trial is to demonstrate the superiority of betrixaban as compared to the current standard of care in the reduction of VTE-related events at 35 days while maintaining a favorable benefit to risk profile.
The APEX study is adequately powered to show a clinically relevant benefit with a p-value of less than 0.01 on the primary endpoint of total asymptomatic proximal DVT (as detected by ultrasound), symptomatic DVT (proximal or distal), non-fatal pulmonary embolism and VTE-related death. The first patient was enrolled in March 2012.
The safety and tolerability of betrixaban for stroke prevention was evaluated in 508 patients with atrial fibrillation in the Phase 2 EXPLORE-Xa dose-ranging study. Results were presented during a late-breaking session at the American College of Cardiology’s 59th Annual Scientific Session in March 2010. The data showed that a once-daily dose of oral betrixaban, given to patients with non-valvular atrial fibrillation or atrial flutter and at least one risk factor for stroke, reduced the incidence of major and clinically relevant non-major bleeds compared to dose-adjusted warfarin. In August 2010, additional pharmacodynamic data from a pre-specified analysis of EXPLORE-Xa showed a concentration dependent relationship and provided further evidence for the anticoagulant activity of betrixaban across all three doses studied in the clinical trial. The additional pharmacodynamic analysis provides information for dose selection for Phase 3 evaluation of betrixaban.
In 2007, positive top-line results from EXPERT were published in The Journal of Thrombosis and Haemostasis. This randomized, multi-center, Phase 2 in-hospital efficacy and safety study of the prevention of VTE compared betrixaban with enoxaparin in 215 patients undergoing knee replacement surgery.
Portola Pharmaceuticals
Betrixaban (INN, codenamed PRT-054,021) is an anticoagulant drug which acts as a direct factor Xa inhibitor.[1] It is potent, orally active and highly selective for factor Xa, being selected from a group of similar compounds for its low hERG affinity.[2] Betrixaban has undergone human clinical trials for prevention of embolism after knee surgery,[3] and prevention of stroke following atrial fibrillation,[4] with promising results.[5]
b1
b2

Patent Document CN1391555A first discloses a preparation method (see Scheme 1):
Figure CN104693114AD00042

CN101595092A  (See Scheme 2).
Figure CN104693114AD00051

Patent Document CN102762538A  (see Scheme 3).
[0013]
Figure CN104693114AD00061


CN104693114
Machine translated from chinese please bear with names
http://www.google.com/patents/CN104693114A?cl=en
Figure CN104693114AD00071

Preparation Example 1 shell song in Spanish
Figure CN104693114AD00111
  Under stirring, temperature 15 ~ 20 ° C, was added dropwise 2mol / L tetrahydrofuran solution of isopropylmagnesium chloride (available commercially available) 308ml (0 • 615mol, 5eq) to 2mol / L dimethylamine THF Solution (commercially available can) 339ml (0.677mol, 5. 5eq) to give dimethylamine reaction solution.
  Under stirring, temperature 15 ~ 20 ° C, the compound of formula II 50. 0g (0 123mol, leq.) Was mixed with 500ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition continued at 25 The reaction was stirred for ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 700ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water; the filter cake was mixed by stirring with 500ml of acetone, the pH adjusted with triethylamine to 7-8; filtered; the cake at 40 ~ 45 ° C and dried under reduced pressure to give Pui Spanish song 45. 5g. . Yield: 82 0%; HPLC purity: 98.9%, of which 0.05% dechlorinated impurities VIII, IX desmethyl impurities were not detected.
Take the above Tony Qu Spanish 45.0g, at about 70 ° C under stirring dissolved in N, N- dimethylacetamide 180ml, a toluene solution of 360ml; cooling crystallization, filtration, the filter cake washed with an appropriate amount of acetone at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish HPLC purity 99.7%.
(+) LC-MS: m / z = 452 ([M + H] +). Che NMR (400MHz, DMS0-d6) S:…. 2 96 (s, 6H), 3 83 (s, 3H), 7. 06-7 09 (dd, 1H), 7. 55-7 59 ( m, 3H), 7. 80-7. 83 (dd, 1H), 8. 21-8. 23 (d, 1H), 8. 27-8. 30 (d, 2H), 8. 37-8. 40 (d, 1H), 8. 41-8. 43 (d, 1H), 10. 54 (br., 2H).
Preparation Example 2 Tony Qu Spanish maleate
  Under stirring, temperature 0 ~ 5 ° C, dropping 2mol isopropyl magnesium chloride in tetrahydrofuran / L (available commercial available) 105ml (0 • 21mol, 8. 4eq) twenty methylamine hydrochloride 8. 91g (0 • llmol, 4. 4eq) in tetrahydrofuran 60ml of the suspension, the reaction solution obtained dimethylamine.
  Under stirring, temperature 0 ~ 5 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, and then dropping the above reaction liquid dimethylamine; After the addition continued 10 The reaction was stirred for ~ 15 ° c, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 10 ~ 15 ° C, the reaction solution was added to an aqueous solution of 45g and 100ml dubbed maleic acid solution; the organic solvent was evaporated under reduced pressure and concentrated, filtered concentrate precipitated solid cake was washed with the right amount of water washing. Cake at 40 ~ 45 ° C and dried under reduced pressure to give Tony Qu Spanish maleate 12.lg. . Yield: 85 4%; HPLC purity: 98.6%, which is 0.03% dechlorinated impurities VIII, IX desmethyl impurities were not detected.
Take the above shellfish Spanish song maleate 10. 0g, at about 70 ° C under stirring dissolved in a mixed solvent of ethanol 50ml and 25ml of water, dropping water 150ml; cooling crystallization, filtration, the filter cake at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish maleate HPLC purity 99.9%.
: HNMR (400MHz, DMS〇-d6) 8: 3. 25 (s, 3H), 3. 32 (s, 3H), 3. 87 (s, 3H), 6. 02 (s, 2H) , 7. 19-7. 21 (dd, 1H), 7. 44-7. 45 (1H), 7. 75-7. 77 (d, 2H), 7. 97-9. 98 (d, 2H) , 8. 08-8. 13 (m, 3H), 8. 44-8. 45 (d, 1H), 9. 01 (br., 1H), 9. 37 (br., 1H), 11.04 (s , 1H), 11. 13 (s, 1H).
Preparation Example 3 Tony Spanish song of [0075] Example
  Under stirring, temperature 25 ~ 30 ° C, isopropylmagnesium chloride in tetrahydrofuran was added dropwise a solution of 2mol / L (available commercially available) 81ml (0 • 161mol, 7eq) to 2mol / L dimethylamine THF Solution (commercially available can) 121ml (0 • 242mol, 10. 5eq) to give dimethylamine reaction solution.
Under stirring, temperature 25 ~ 30 ° C, the hydrochloride salt of a compound of formula II 10. 0g (0 023mol, leq.) Was mixed with 100ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition was complete The reaction continued stirring at 25 ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 210ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water; the filter cake with 90ml acetone was stirred and mixed, the pH adjusted with triethylamine to 7-8; filtration; cake was 45 ~ 50 ° C and dried under reduced pressure to give Pui Qu Spanish 8. 35g. Yield: 80.5%. HPLC purity: 98.7%, which is 0.03% dechlorinated impurities VIII, IX desmethyl impurities were not detected.
Preparation Example 4 shellfish Spanish song hydrochloride
  Under stirring, temperature 15 ~ 20 ° C, dropping lmol / n-amyl magnesium bromide tetrahydrofuran solution (which can be commercialized available) 75ml (0 • 075mol, 3eq) to 2mol / L of dimethyl L amine in tetrahydrofuran (commercially available can) 56ml (0 • 113mol, 4. 5eq) to give dimethylamine reaction solution.
Under stirring, temperature 15 ~ 20 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition continued at 25 The reaction was stirred for ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 100ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water. Cake at 40 ~ 45 ° C and dried under reduced pressure to give Tony Qu Spanish hydrochloride 10.lg, yield:. 82 9%; HPLC purity: 99.0%, which is 0.02% dechlorination impurity VIII, from A impurities IX was not detected.
  Take the above shellfish Spanish song hydrochloride 10. 0g, at about 70 ° C under stirring dissolved in N, N- dimethylacetamide 40ml, a toluene solution of 80ml; cooling crystallization, filtration, cake at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish hydrochloride HPLC purity 99.8%.
Preparation 5 shellfish Spanish song of [0082] Example
Under stirring, temperature 0 ~ 5 ° C, dropping lmol / diethyl zinc toluene solution of L (available commercially oriented) 50ml (0. 050mol, 2eq) to 2mol / L dimethylamine tetrahydrofuran (commercially available can) 28ml (0. 055mol, 2. 2eq) to give dimethylamine reaction solution.
  Under stirring, temperature 0 ~ 5 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, and then dropping the above reaction liquid dimethylamine; After dropping 5 continues The reaction was stirred for ~ 10 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, in the next 5 ~ 10 ° C, the reaction mixture was added to about 2mol L dilute hydrochloric acid solution 70ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake was washed successively with a suitable amount of water; the filter cake with acetone l〇〇ml mixing, the pH adjusted with triethylamine to 7-8; filtered; the cake at 40 ~ 45 ° C under reduced pressed and dried to give Tony Qu Spanish 9. 03g. . Yield: 80 1%; HPLC purity: 99.0%, which is 0.02% dechlorinated impurities VIII, IX desmethyl impurities were not detected.
  Preparation of compounds of Formula II Preparation Example 1
Methoxy-2-nitro – (5-chloro-pyridin-2-yl) -5 – benzamide (compound V) Preparation of [0086] (1) N-
Figure CN104693114AD00131
  with stirring at room temperature, 5-methoxy-2-nitrobenzoic acid (Compound VI, can be commercially available) 250g (1. 27mol, leq) and 2-amino-5-chloropyridine (Compound VII .) 163g (l 27mol, leq) was suspended in 1700ml of acetonitrile, pyridine 301g (3 81mol, 3eq), and then phosphorus oxychloride was added dropwise 231g (l 52mol, 1 2eq);… After stirring for 1 hour the reaction 3500ml water quenching crystallization; the filter cake was washed with water 1700mlX2; dried under reduced pressure to obtain compound V349g.
  (2) 2-Amino -N- (5- chloro – pyridin-2-yl) -5-methoxy – benzamide (compound IV) is prepared
Figure CN104693114AD00132
  with stirring at room temperature, the N- (5- chloro – pyridin-2-yl) -5-methoxy-2-nitro – benzamide (Compound V) 300g (0 • 977mol, 1.Oeq) 3000ml was dissolved in acetic acid, and iron powder was added portionwise 546g (9 77mol, 10eq.); After the addition of iron stirring was continued for 3 hours, and then ethyl acetate and water 6000ml 3000ml, liquid separation; the aqueous phase was separated 3000mlX2 extracted with ethyl acetate; combined organic phases were washed with water, saturated aqueous sodium bicarbonate, saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give compound IV244g.
(3) N- (5- chloro – pyridin-2-yl) -2- (4-cyano – benzoyl – amino) -5-methoxy – benzamide (compound II) is prepared
Figure CN104693114AD00141
at 10 ~ 20 ° C, a solution of a compound of formula IV 200g (0 • 72mol, 1.Oeq) and triethylamine 109g (1. 08mol, 1. 5eq) 2000ml dissolved in tetrahydrofuran, to which was added dropwise to cyano benzoyl chloride (compound III, commercially available technology) 130g (0 79mol, 1.leq.) and tetrahydrofuran solution dubbed 1000ml, HPLC monitoring progress of the reaction; after the reaction was filtered, the filter cake washed with an appropriate amount of ethanol, dried under reduced pressure to obtain compound II263g. HPLC purity: 98.7%.
  (+) LC-MS: m / z = 407 ([M + H] +). Insect NMR (400MHz, DMS0-d6) S:… 3 85 (s, 3H), 7 16-7 .19 (dd, 1H), 7. 39-7 41 (d, 1H), 7. 93- 7. 96 (d, 2H), 8. 02-8. 04 (m, 4H), 8. 13-8. 14 (d, 2H), 8. 42-8. 43 (d, 1H), 11. 06 (br. 2H).
Example 2 Preparation of the hydrochloride salt of the compound of formula II
  at 10 ~ 20 ° C, a solution of a compound of formula IV 40. 0g (0 • 14mol, 1.Oeq) was dissolved in 400ml of tetrahydrofuran, a solution of cyanobenzoyl chloride (Compound III, can be commercialized available) 24 8g (0 15mol, 1.leq) and tetrahydrofuran solution 200ml dubbed, HPLC monitoring progress of the reaction;.. After the reaction was filtered, the filter cake washed with ethanol and after an appropriate amount, and dried under reduced pressure to obtain a compound of formula II hydrochloride . HPLC purity: 99.5%.

 WO 2015176591

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015176591&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText
Example 1: Preparation of Spanish Preparation and Form A half-L- malic acid shellfish song
At 55 ~ 60 ℃, the shellfish song Spanish 6.0g (13.3mmol), L- malic acid 1.1g (8.0mmol) was dissolved in tetrahydrofuran 70mL / water 7mL mixed solvent acetone was added with stirring 60mL, cooled to room temperature, Crystallization. Precipitated solid was filtered, and the resulting solid at 40 ~ 45 ℃ vacuum dried to give half L- malic acid shellfish Spanish song.
1H NMR(400MHz,MeOD)δ:2.355-2.419(dd,0.5H),2.735-2.781(dd,0.5H),3.226(s,6H),3.907(s,3H),4.302-4.326(dd,0.5H),7.195-7.224(dd,1H),7.448-7.455(d,1H),7.744-7.764(d,2H),7.821-7.849(dd,1H),8.145-8.165(d,2H),8.196-8.219(d,1H),8.238-8.261(d,1H),8.323-8.329(d,1H)。
Above 1 H-NMR results, δ: 3.907 (s, 3H) attributed to shellfish Spanish song molecule methyl CH 3 , 4.302-4.326 (dd, 0.5H) attributed to L- malic acid molecule methine CH , you can determine the song title product in shellfish Spanish and L- malic acid molar ratio of 2: 1.

PATENT

http://www.google.com.na/patents/EP2101760A2?cl=en

Example 2
Preparation of the compound of Formula II

a. Gram scale preparation A slurry of the compound of Formula F (455 g, 1.0 eq.) in THF (4.67 kg,

10.3 parts) was prepared and adjusted to <10 0C. Lithium dimethyl amide was prepared as follows :hexyllithium (2.3 N/hexane, 2.45 L, 5.5 eq.) was added to dimethylamine solution (2 N/THF, 2.8 L, 5.5 eq.) maintaining <10 0C. The lithium dimethyl amide solution was charged into the slurry containing the compound of Formula F keeping the pot temperature of <10 0C. The reaction progress was monitored by in-process HPLC which confirmed that the amount of Formula F was <1.0 A%. A buffer solution of NaHCO3 (490 g, 1.1 parts, 5.7 eq.) and Na2CO3 (490 g, 1.1 parts, 4.5 eq.) in deionized water (6.6 kg, 14.51 parts) was prepared, and the above reaction mixture was transferred to this aqueous solution maintaining < 5 0C. The product precipitated out and the resulting slurry was adjusted to 20 0C over a period of 12 hr. The solid was filtered, and the resulting wet cake was washed with 3.5 kg (7.7 parts) of deionized water. The solid was filtered off using a coarse frit glass bench filter, and rinsed forwarded with cold (0-5 0C) absolute ethanol (628 g, 1.4 parts). The product was dried at 30-35 0C. Dry product was obtained in 458 g (73% yield). b. Kilogram scale preparation A slurry of the compound of Formula F (31.5 kg, 1.0 eq.) in THF (251 kg,
8.0 parts) was prepared in a 780 L Hastelloy reactor (Reactor A) and adjusted to 0 0C (-3 to 3 0C). 2 M Dimethylamine in THF (161.0 kg, 5.0 eq.) and THF (63 kg, 2 parts) were charged into a 1900 L GLMS reactor (Reactor B) and adjusted to 0 0C (-3 to 3 0C) with maximum agitation. Hexyllithium (2.3 M, 97.2 kg, 4.5 eq.) was slowly charged to Reactor B while maintaining a max temperature of 10 0C. The pump and lines were rinsed forward to Reactor B with THF (3.2 kg). The Reactor B contents were adjusted to 0 0C (-3 to 3 0C), then transferred to Reactor A while keeping Reactor A temperature < 10 0C. The Reactor B pump and lines were rinsed forward with THF (31.4 kg, 1.0 part). The Reactor A contents were adjusted to 0 0C (-3 to 3 0C), and agitated at this temperature until the reaction was complete as verified by HPLC (1-2 hrs). After about 1 hr of agitation, in-process HPLC analysis indicated that 0 A% starting material remained (in-process criteria: max 1 A%). Reactor A contents were adjusted to -5 0C (-8 to -3 0C). In-process cleaning of Reactor B with water was performed. Two previously prepared aqueous solutions (NaHCO3 (35.0 kg, 1.1 parts) in water (236 kg, 7.5 parts), and Na2CO3 (35.0 kg 1.1 parts) in water (236 kg, 7.5 parts))were charged to Reactor B and adjusted to -3 0C (0 to 6 0C). Reactor A contents were transferred to Reactor B through an insulated line, maintaining the temperature of Reactor B at -8 0C to a maximum of 5 0C. The Reactor A pump and lines were rinsed forward with cold [-5 0C (-8 to -3 0C)] THF (31.4 kg, 1.0 part). Reactor B contents were adjusted to 22 0C (19-25 0C) and agitated for ca. 3 hrs. Slurry formation was visually confirmed, and Reactor B contents were filtered onto a 30″ centrifuge fitted with a filter cloth. The Reactor B pump and lines were rinsed forward onto the 30″ centrifuge fitted with a filter cloth with drinking water (63 kg, 2 parts). The wet filter cake (66.5 kg) was transferred back to Reactor B and submitted to a slurry wash in drinking water (1005 kg, 32 parts) at 22 0C (19-25) 0C for ca. 1 hr. The product was filtered onto the 30″ centrifuge (after in-process cleaning and fitting with a filter cloth), and the Reactor B lines and pump were rinsed forward with drinking water (63 kg, 2 parts). The water rinse was sampled for test by TDS, which was found to be 0.46%. The Reactor B pump, lines and wet filter cake were further rinsed with cold [0 0C (-3 to 3 0C)] ethanol (44 kg, 1.39 parts). The wet filter cake was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 35 0C. In-process LOD was 0% after ca. 24 hrs of drying, and the product was discharged (24.8 kg) in 76.7% yield. HPLC showed 98 % purity, with dechlorinated impurity at 1.14 %. Example 3
Preparation of the compound of Formula F Step 1. Synthesis of 2-nitro-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (C)

5-Methoxy-2-nitrobenzoic acid (A) (25.0 kg, 1.0 eq.), 2-amino-5- chloropyridine (B) (16.3 kg, 1.0 eq.), and acetonitrile (87.5 kg, 3.5 parts) were charged to a 380 L GLMS reactor. The reaction mixture was adjusted to 22 0C (19-25 0C) and anhydrous pyridine (30.0 kg, 3.0 eq.) was added. The pump and lines were rinsed forward with acetonitrile (22.5 kg, 0.9 parts), and the reactor contents were adjusted to a temperature of 19-22 0C. Phosphorous oxychloride (23.3 kg, 1.20 eq.) was charged to the contents of the reactor via a metering pump, while maintaining a temperature of 25 0C (22-28 0C). The metering pump and lines were rinsed forward with acetonitrile (12.5 kg, 0.5 parts), while keeping the temperature at 25 0C (22-28 0C). The reaction mixture normally turned from a slurry to a clear solution after the addition of about 1/3 of the POCI3. At the end of the addition, it became turbid. After complete addition, the reaction mixture was agitated at 25 0C (22-28 0C) for ca. 1 hr, at which time HPLC analysis confirmed reaction completion. The solution was cooled to 15 0C (12-18 0C) and drinking water (156.3 kg, 6.25 parts) was charged slowly while keeping reaction temperature of between 12 and 30 0C. The reaction mixture was then adjusted to 22 0C (19-25 0C) and agitated for ca. 5 hrs until exotherm ceased. Formation of a slurry was visually confirmed and the contents of the reactor were filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were washed forward onto the pressure nutsche with two portions of drinking water (62.5 kg, 2.5 parts each). The filtrate had a pH value of 7. The product (41.8 kg) was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 50 0C. After ca. 12 hrs, in-process LOD analysis indicated a solvent content of 0.72%. The dry product (C) was discharged (34.4 kg) with 88.2% yield and 99.1 % purity by HPLC. Step 2. Synthesis of 2-amino-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (D)

To a 780 L Hastelloy reactor, compound C (33 kg, 1.0 eq.), 5% platinum carbon (sulfided, 0.33 kg, 0.010 parts) and dichloromethane (578 kg, 17.5 parts) were charged. Agitation was started and reactor contents were adjusted to 22 0C (19-25 0C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 28 0C (25-31 0C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 28 0C (25 to 31 0C; maximum 31 0C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned celite pad (0.2-0.5 kg celite conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and celite bed were rinsed forward with two portions of dichloromethane (83 kg, 2.5 parts each). The filtrate was transferred to and concentrated in a 570 L GLMS reactor under a atmospheric pressure to ca. 132 L (4 parts volume). Ethanol (69 kg, 2.1 parts) was charged and concentration continued under atmospheric pressure to ca. 99 L (3 parts volume). In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg, 2.1 parts) was charged again and concentration continued again to ca. 99 L (3 parts volume). In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 0C (0 to 6 0C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 0C (0-6 0C)] ethanol (26 kg, 0.8 parts). The wet filter cake (36.6 kg) was dried under vacuum at 40-50 0C with a maximum temperature of water bath (to heat dryer jacket) of 50 0C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (D) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %. Step 3. Synthesis of N-(5-chloro-pyridin-2-yl)-2-(4-cyano-benzoyl-amino)-5-methoxy- benzamide Hydrochloride (F)
To a 780 L Hastelloy reactor, was charged 4-cyanobenzoyl chloride (E)
(17.2 kg, 1.1 eq.) and THF (92 kg, 3.5 parts). Reactor contents were agitated at 22 0C (19- 25 0C) until all of the solids had dissolved. The resulting solution was transferred to a lower receiver and the reactor was rinsed forward with THF (26 kg, 1 part). Compound D (26.4 kg, 1 eq.), THF (396 kg, 15 parts) and pyridine (2.90 kg, 0.4 eq.) were charged to a clean reactor. The pump and lines were rinsed forward with THF (34 kg, 1.3 parts). Via a metering pump, the 4-cyanobenzoyl chloride/THF solution was charged to the reactor, keeping the temperature at < 30 0C and rinsing forward with THF (ca. 10 kg). The resulting yellow-colored slurry was agitated at 22 0C (19-25 0C) for ca 2 hrs. In-process HPLC taken after 2 hrs showed a compound of Formula D content of 0%, indicating completion of the reaction. The slurry was filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, lines and wet cake were rinsed with three portions of ethanol (ca. 15 kg each). The wet filter cake was discharged (65.4 kg) and transferred back to the reactor for slurry wash in ethanol (317 kg, 12 parts) at 22 0C (19-25 0C) for ca. 1 hr. The slurry was filtered onto the pressure nutsche and the reactor, pump, lines, and wet filter cake were rinsed with two portions of ethanol (ca. 15 kg each) and two portions of THF (ca. 15 kg each). The wet filter cake was dried under vacuum with a maximum temperature of warm glycol bath (to heat the dryer jacket) of 40 0C. After 14.5 hrs of drying, LOD was 0.75%. The dried material was milled (screen 0.125″) to give 31.8 kg of product, which was dried under vacuum for another 10.5 hrs. LOD after drying was 1.8%, and the product was discharged (31.5 kg) in 74.8% yield (expected 60-90%). HPLC showed 100 % purity.

PATENT

http://www.google.com/patents/WO2011084519A1?cl=en
U.S. Patent No. 6,376,515 B2 discloses a class of benzamide based compounds as specific factor Xa inhibitors. In particular, U.S. Patent No. 6,376,515 B2 describes a compound identified as Example 206, which is also disclosed in U.S. Patent No. 6,835,739 B2 as Example 206 and herein identified as betrixaban, which has the chemical formula of Formula I:



Scheme 1
Example 1: Preparation of betrixaban
[0113] Dimethylformamide (13L) and hydrochloride (18 mL) were charged into a reactor. Compound B (1 kg) was added followed by Compound A (0.88 kg).
Compound A is commercially available or, just as with Compound B may be prepared using the methods described in Examples 4 and 5. The reaction mixture was cooled between 0 °C and -10 °C. EDC (0.752 kg) was added while maintaining the temperature between -10 °C and 0 °C. The reaction mixture was stirred until the content of
Compound B is below 0.10% area by HPLC. The reaction mixture was stirred until betrixaban started to crystallize. Acetone (26 L) was then added during a period of at least 1 hr while the temperature was maintained at between -10 °C and 0 °C. The suspension was then stirred for additional 2 hrs at a temperature of between 0 °C and 10 °C. The suspension was filtered and washed with cold acetone to give a wet product betrixaban. Example 2: Preparation of a maleate salt of betrixaban
[0114] The wet betrixaban obtained above was reacted with maleic acid (0.52 x weight of maleic acid/weight of dry betrixaban) in ethanol (22.4 x volume of
liquid/weight of dry betrixaban (v/w)) and purified water (5.7 x v/w) to form a betrixaban maleate salt. The solution of the betrixaban maleate salt was filtered and concentrated under vacuum until a final volume of 5.7 x v/w. Water (2 x v/w) was then added and the mixture was back concentrated until the same volume. The procedure of adding water and distil until a final volume of 5.7 x v/w was carried out until the molar ratio between the content of ethanol and the content of betrixaban maleate salt in the mixture was lower than, or equal to, 6. Betrixaban maleate salt crystallized during the removal of ethanol. The suspension was cooled to a temperature between 19 °C and 25 °C and stirred for not less than 2 hours at this temperature range. Betrixaban maleate salt was isolated by filtration, washed with water and dried under vacuum at a maximum temperature of 40 °C until the content of water was lower than, or equal to, 0.5 % w/w by Karl-Fisher. The purity of the maleate salt was determined to be greater than 99 % by HPLC. The betrixaban maleate isolated was in a crystalline form A which was concluded based on IR, DSC and XRPD results obtained, see Figures 3-5, respectively. The major peaks of XRPD pattern of crystalline form A are also listed in Table 2. Table 2: Betrixaban Form A XRPD Peak °2-Theta (2Θ0)
Example 3: Synthesis of 2-nitro-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (C)
D E C
[0115] 5-Methoxy-2-nitrobenzoic acid (D) (25.0 kg, 1.0 eq.), 2-amino-5- chloropyridine (E) (16.3 kg, 1.0 eq.), and acetonitrile (87.5 kg) were charged to a 380 L glass-lined reactor. The reaction mixture was adjusted to 22 °C (19-25 °C) and anhydrous pyridine (30.0 kg, 3.0 eq.) was added. The pump and lines were rinsed forward with acetonitrile (22.5 kg), and the reactor contents were adjusted to a temperature of 19-22 °C. Phosphorous oxychloride (23.3 kg, 1.20 eq.) was charged to the contents of the reactor via a metering pump, while maintaining a temperature of 25 °C (22-28 °C). The metering pump and lines were rinsed forward with acetonitrile (12.5 kg), while keeping the temperature at 25 °C (22-28 °C). The reaction mixture normally turned from a slurry to a clear solution after the addition of about 1/3 of the POCI3. At the end of the addition, it became turbid. After complete addition, the reaction mixture was agitated at 25 °C (22- 28 °C) for ca. 1 hr, at which time HPLC analysis confirmed reaction completion. The solution was cooled to 15 °C (12-18 °C) and water (156.3 kg) was charged slowly while keeping reaction temperature of between 12 and 30 °C. The reaction mixture was then adjusted to 22 °C (19-25 °C) and agitated for ca. 5 hrs until exotherm ceased. Formation of a slurry was visually confirmed and the contents of the reactor were filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were washed forward onto the pressure nutsche with two portions of water (62.5 kg). The filtrate had a pH value of 7. The product (41.8 kg) was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. After ca. 12 hrs, in-process LOD analysis indicated a solvent content of 0.72%. The dry product (C) was discharged (34.4 kg) with 88.2% yield and 99.1 % purity by HPLC.
Exam le 4. Synthesis of 2-amino-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide
Process A
[0116] To a 780 L Hastelloy reactor, Compound C (33 kg, 1.0 eq.), 5%> platinum carbon (sulfided, 0.33 kg) and dichloromethane (578 kg) were charged. Agitation was started and reactor contents were adjusted to 22 °C (19-25 °C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 28 °C (25-31 °C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 28 °C (25 to 31 °C; maximum 31 °C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned Celite™ (diatomaceous earth; Celite Co., Santa Barbara, Ca.) pad (0.2-0.5 kg Celite™ conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and Celite™ bed were rinsed forward with two portions of dichloromethane (83 kg). The filtrate was transferred to and concentrated in a 570 L glass-lined reactor under an atmospheric pressure to ca. 132 L. Ethanol (69 kg) was charged and concentration continued under atmospheric pressure to ca. 99 L. In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg) was charged again and concentration continued again to ca. 99 L. In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 °C (0 to 6 °C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 °C (0-6 °C)] ethanol (26 kg. The wet filter cake (36.6 kg) was dried under vacuum at 40-50 °C with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (B) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %.
Process B
[0117] To a 780 L Hastelloy reactor, Compound C (33 kg, 1.0 eq.), 5%> platinum carbon (sulfided, 0.33 kg) and dichloromethane (578 kg) were charged. Agitation was started and reactor contents were adjusted to 22 °C (19-25 °C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 26 °C (21 to 31 °C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 26 °C (21 to 31 °C; maximum 31 °C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned Celite™ pad (0.2-0.5 kg Celite™ conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and Celite™ bed were rinsed forward with two portions of dichloromethane (83 kg). The filtrate was transferred to and concentrated in a 570 L glass-lined reactor under vacuum and a maximum temperature of 45 °C to ca. 132 L. Ethanol (69 kg) was charged and concentration continued under vacuum and a maximum temperature of 45 °C to ca. 132 L. In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg) was charged again and concentration continued again to ca. 132 L. In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 °C (0 to 6 °C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 °C (0-6 °C)] ethanol (26 kg. The wet filter cake (36.6 kg) was dried under vacuum at 40-50 °C with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (B) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %.
Example 5. Synthesis of 4-(N,N-dimethylcarbamimidoyl)benzoic acid (A)
Process A
Step 1: Amidine Formation
[0118] To a tetrahydrofuran solution of 2M dimethylamine, 2.3M hexane solution of hexyllithium was slowly added over a period of at least three (3) hours while maintaining the temperature at between -8°C and -12°C. This solution was added to the tetrahydrofuran solution of ethyl-4-cyanobenzoate (F) while maintaining the temperature between -8°C and -12°C. The completion of the reaction was confirmed by HPLC, and the solution temperature was adjusted to between -8°C and 3°C. The reaction mixture was slowly added to the cold solution of aqueous sodium bicarbonate solution and the desired ethyl-4-(N,N-dimethylcarbamimidoyl)benzoate (G) was extracted with ethyl acetate. The ethyl acetate layer was dried, filtered and evaporated under vacuum to afford ethyl-4-(N,N-dimethylcarbamimidoyl)benzoate (G) as a white solid.
Step 2: Hydrolysis of ester
[0119] To a THF solution of ethyl -4(N,N-dimethylcarbamimidoyl)benzoate (G) was added an aqueous solution of lithium hydroxide (2 eq.) and the reaction mixture was stirred for 6 hr. The completion of the reaction was confirmed by HPLC. To the reaction mixture was added water, followed by extraction with ethyl acetate. The aqueous layer was acidified with 6N HCI to pH between 3-4 at which point the desired 4-(N,N- dimethylcarbamimidoyl)benzoic acid precipitated as the white solid. The white solid isolated was washed with hexane to afford 4-(N,N-dimethylcarbamimidoyl)benzoic acid as an hydrochloride salt (A).
Process B:
Step 1: Ester Formation
[0120] To a methanolic solution of 4-cyanobenzoic acid was added concentrated sulfuric acid and refluxed the reaction for at least 12 hours. The completion of the reaction was confirmed by HPLC. The solution was cooled and the solvent was evaporated. To the residue was added ethyl acetate followed by washing with 10 % sodium hydroxide solution. The ethyl acetate layer was dried, filtered and evaporated to give desired 4-methyl cyanobenzoate as a white solid.
Step 2: Dimethylamidine formation
[0121] A stream of HCI (gas) was bubbled through a 0 °C solution of 4-methyl cyanobenzoate (1 mmol) in 50 mL of ethanol until saturation. The mixture was stirred at room temperature overnight and evaporated to afford compound P. The resulting residue was treated with dimethylamine hydrochloride (0.15 eq.) in 20 mL ethanol at reflux temperature for 4 hours. The solvent was removed at reduced pressure and the residue was washed with hexane to afford desired product Q as a light yellow solid.
Step 3: Ester hydrolysis
[0122] To a THF solution of ethyl-4(N,N-dimethylcarbamimidoyl)benzoate (Q) was added an aqueous solution of lithium hydroxide (2 eq.) and the reaction mixture was stirred for 6 hours. The completion of the reaction was confirmed by HPLC. To the reaction mixture was added water, followed by extraction with ethyl acetate. The aqueous layer was acidified with 6N HC1 to pH between 3-4 at which point the desired 4- (N,N-dimethylcarbamimidoyl)benzoic acid precipitated as the white solid. The white solid isolated was washed with hexane to afford 4-(N,N-dimethylcarbamimidoyl)benzoic acid as an hydrochloride salt (A).
Example 6: Preparation of betrixaban, free base
[0123] To 100 mL round bottom flask, was added compound B (2.0 g, obtained as in Example 4), compound A (1.98 g, obtained as in example 5), 20 mL N,N- dimethylacetamide. The reaction mixture was stirred briefly so as to dissolve most of the solid, then con. HC1 (36 microliters) was added. To this thin slurry add EDC HCl (1.8 g total, Aldrich) in 3 portions, 0.6 g each, 20 min apart. The reaction mixture was stirred for 1.5 hours for complete reaction. [0124] To this reaction was added 2.3 g sodium carbonate solution in 10 mL water while the batch was cooled with water bath to keep the batch temperature 22-30 °C. Vigorous agitation was required to keep the batch well mixed. Then 10 mL water was added. The batch was stirred at 22-25 °C for 30 min. After a slurry was formed, 20 mL more water was added. The batch was stirred at 22 °C for 1 hour. The batch was filtered and the wet cake was washed with 3×5 mL water, then 5 mL acetone. The cake was dried on the funnel by suction. The weight of the dry cake is 2.95 g -2.92 g which is the crude betrixaban. To purify the crude betrixaban obtained, 1.0 g of the crude solid was mixed with 4 mL Ν,Ν-dimethylacetamide and heated to 70 °C for 30 min. Then add 8 mL toluene was added and the mixture was heated for 30 min, then cooled to 22 °C over 1 h, then cooled to 0 °C, aged at 0 °C for 2 hours, filtered, washed with 2×1 mL toluene. The cake was dried on the funnel by suction to obtain 0.88 g pure betrixaban (I).

WO2012031017A1 * Aug 31, 2011 Mar 8, 2012 Merck Sharp & Dohme Corp. CRYSTALLINE FORMS OF A FACTOR Xa INHIBITOR
WO2013033370A1 * Aug 30, 2012 Mar 7, 2013 Portola Pharmaceuticals, Inc. Prevention and treatment of thrombosis in medically ill patients
US8946269 Aug 31, 2011 Feb 3, 2015 Portola Pharmaceuticals, Inc. Crystalline forms of a factor Xa inhibitor
WO2004083174A2 * Mar 17, 2004 Sep 30, 2004 Timur Gangor Sulfonyl-amidino containing and tetrahydropyrimidino containing compounds as factor xa inhibitors
WO2008057972A1 Nov 1, 2007 May 15, 2008 Millennium Pharm Inc Methods of synthesizing pharmaceutical salts of a factor xa inhibitor
US6376515 Feb 28, 2001 Apr 23, 2002 Cor Therapeutics, Inc. Benzamides and related inhibitors of factor Xa
US6835739 Oct 15, 2003 Dec 28, 2004 Millennium Pharmaceuticals, Inc. Benzamides and related inhibitors of factor Xa
US6844367 Sep 15, 2000 Jan 18, 2005 Millennium Pharmaceuticals, Inc. Benzamides and related inhibitors of factor Xa

US61287680

References



  1. Eriksson BI, Quinlan DJ, Weitz JI (2009). “Comparative pharmacodynamics and pharmacokinetics of oral direct thrombin and factor xa inhibitors in development”. Clinical Pharmacokinetics48 (1): 1–22. PMID19071881.
  2. Zhang P, Huang W, Wang L, Bao L, Jia ZJ, Bauer SM, Goldman EA, Probst GD, Song Y, Su T, Fan J, Wu Y, Li W, Woolfrey J, Sinha U, Wong PW, Edwards ST, Arfsten AE, Clizbe LA, Kanter J, Pandey A, Park G, Hutchaleelaha A, Lambing JL, Hollenbach SJ, Scarborough RM, Zhu BY (April 2009). “Discovery of betrixaban (PRT054021), N-(5-chloropyridin-2-yl)-2-(4-(N,N-dimethylcarbamimidoyl)benzamido)-5-methoxybenzamide, a highly potent, selective, and orally efficacious factor Xa inhibitor”. Bioorganic & Medicinal Chemistry Letters19 (8): 2179–85. doi:10.1016/j.bmcl.2009.02.111. PMID19297154.
  3. Turpie AG, Bauer KA, Davidson BL, Fisher WD, Gent M, Huo MH, Sinha U, Gretler DD (January 2009). “A randomized evaluation of betrixaban, an oral factor Xa inhibitor, for prevention of thromboembolic events after total knee replacement (EXPERT)”. Thrombosis and Haemostasis101 (1): 68–76. PMID19132191.
  4. Piccini, J. P.; Lopes, R. D.; Mahaffey, K. W. (2010). “Oral factor Xa inhibitors for the prevention of stroke in atrial fibrillation”. Current Opinion in Cardiology25 (4): 312. doi:10.1097/HCO.0b013e32833a524f. PMID20520539edit
  5. Sobieraj-Teague, M.; O’donnell, M.; Eikelboom, J. (2009). “New Anticoagulants for Atrial Fibrillation”. Seminars in Thrombosis and Hemostasis35 (5): 515–24. doi:10.1055/s-0029-1234147. PMID19739042edit
Husten, Harry. “Merck Abandons Development of Factor Xa Inhibitor Betrixaban”. CardioBrief. Retrieved 11 April 2014.
Betrixaban
Betrixaban.svg
Systematic (IUPAC) name
N-(5-chloropyridin-2-yl)-2-([4-(N,N-dimethylcarbamimidoyl)benzoyl]amino)-5-methoxybenzamide
Clinical data
Legal status
  • Development terminated
Identifiers
CAS Number 330942-05-7 
ATC code None
PubChem CID: 10275777
ChemSpider 18981107 Yes
UNII 74RWP7W0J9 Yes
ChEMBL CHEMBL512351 Yes
Chemical data
Formula C23H22ClN5O3
Molecular mass 451.905 g/mol

/////////////CN(C)C(=N)C1=CC=C(C=C1)C(=O)NC2=C(C=C(C=C2)OC)C(=O)NC3=NC=C(C=C3)Cl


See...........http://newdrugapprovals.org/2013/03/05/phase-3-portola-pharma-betrixaban-long-acting-oral-direct-factor-xa-inhibitor/

Share this:

DRL 17822 from Reddy US Therapeutics/Dr Reddy's

Figure imgf000125_0002

Figure imgf000076_0001Figure imgf000226_0001
cas 920493-71-6 and 898911-09-6

reddys1
DRL 17822
MW 603.6045, MFC30 H31 F6 N7
Cas 898911-09-6, 1454689-50-9
3-([[3,5-Bis(trifluoromethyl)benzyl](2-methyl-2H-tetrazol-5-yl)amino]methyl)-N,N-bis(cyclopropylmethyl)-8-methylquinolin-2-amine
3-​Quinolinemethanamine​, 2-​[bis(cyclopropylmeth​yl)​amino]​-​N-​[[3,​5-​bis(trifluoromethyl)​phenyl]​methyl]​-​8-​methyl-​N-​(2-​methyl-​2H-​tetrazol-​5-​yl)​-
3-(((3,5-bis(trifluoromethyl)benzyl)(2- methyl-2H-tetrazol-5-yl)amino)methyl)-N,N-bis(cyclopropylmethyl)-8- methylquinolin-2-amine
(3-{ [3,5-bis-trifluoromethyl-benzyl)-(2-methyl-2H-tetrazoIe-5-yl)- amino]-methyl}-8-methyl-quinolin-2-yl)-bis-cyclopropylmethyl-amine
Reddy US Therapeutics   (Innovator)
Dr. Reddy's Laboratories Ltd.



r1

Treatment of Atherosclerosis Therapy Lipoprotein Disorders,
CETP inhibitor (dyslipidemia/atherosclerosis/cardiovascular diseases), Dr Reddy's
Selective inhibitor of cholesteryl ester transfer protein (CETP)
  • 30 Jun 2012Dr Reddy's Laboratories completes a phase II trial in Hypercholesterolaemia in Italy, Poland and Ukraine (NCT01388816)
  • 09 Mar 2012Dr Reddy's Laboratories completes enrolment in its phase II trial for Hypercholesterolaemia in Italy, Poland, and Ukraine (NCT01388816)
  • 02 Sep 2011Phase-II clinical trials in Hypercholesterolaemia in Ukraine (PO)
CLINICAL TRIALS.....Type II Hyperlipidemia PHASE 2............https://clinicaltrials.gov/ct2/show/NCT01388816
Cardiovascular disease is a leading cause of death worldwide. Among cardiovascular disorders, coronary heart disease (CHD) caused by atherosclerosis is the most common cause of morbidity and mortality. Prevention, stabilization and regression of atherosclerotic plaques may have a major impact on reducing the risk of acute coronary events.
LDL-C lowering agents, primarily the statins, are the current mainstay in the pharmacologic management of dyslipidemia. However even with stain use, residual CHD risk from dyslipidemia remains. Epidemiologic and observational studies have shown that HDL-C is also a strong independent predictor of CHD, suggesting that raising HDL-C levels might afford clinical benefit in the reduction of cardiovascular risk.
Presently only niacin is approved by the FDA for HDL-C elevation and can raise HDL-C levels by 20-30%. However its use can be limited by a high incidence of flushing and, less commonly, by elevation of blood glucose and potential hepatic toxicity.
Cholesteryl ester transfer protein (CETP) inhibitors are being explored for their ability to elevate HDL-C. A small molecule CETP inhibitor, torcetrapib, has been demonstrated to elevate HDL-C by 60-100%. However, a large clinical trial (ILLUMINATE) where it increased HDL-C by a mean of 72% compared to baseline was halted as it failed to show benefit. Post-hoc analysis of this study implicated an off-target increase in blood pressure as potentially counteracting any anti-atherosclerotic benefits. Post-hoc subgroup analysis showed that patients in the highest HDL-C quartile had a 57% reduction in the risk of cardiovascular events.
Increased blood pressure appears to be specifically related to torcetrapib as two other small molecule CETP inhibitors, anacetrapib and dalcetrapib, have not shown this in clinical trials and have been well tolerated. DRL-17822 has also not shown elevation of blood pressure in either animals or in normal volunteers.
This study will investigate the efficacy and tolerability of DRL-17822 as dyslipidemia monotherapy in patients with Type II hyperlipidemia.

Hyperlipidemia or an elevation in serum lipids is associated with an increase incidence of cardiovascular disease and atherosclerosis. Primary hyperlipidemia is a term used to describe a defect in lipoprotein metabolism. The lipoproteins commonly affected are low density lipoprotein (LDL) cholesterol, which transports mainly cholesterol, and very low density lipoprotein-cholesterol (VLDL-cholesterol), which transports mainly triglycerides (TG). Most subjects with hyperlipidemia have a defect in LDL metabolism, characterized by raised cholesterol, LDL-C levels, with or without raised triglyceride levels; such subjects are termed hypercholesterolemic (Fredrickson Type II). Familial hypercholesterolemia (FH) is caused by any one of a number of genetically-determined defects in the LDL receptor, which is important for the entry of cholesterol into cells. The condition is characterized by a reduced number of functional LDL receptors, and is therefore associated with raised serum LDL-C levels due to an increase in LDL.

It is reasonably known in the art that the likelihood of cardiovascular disease can be decreased, if the serum lipids, and in particular LDL-C, can be reduced. It is further known that the progression of atherosclerosis can be retarded or the regression of atherosclerosis can be induced if serum lipids can be lowered. In such cases, individuals diagnosed with hyperlipidemia or hypercholesteremia should consider lipid-lowering therapy to retard the progression or induce the regression of atherosclerosis for purposes of reducing their risk of cardiovascular disease, and in particular coronary artery disease.
Cholesteryl ester-transfer protein (CETP) is an important player in metabolism of lipoproteins, such as, for example, a high density lipoprotein (HDL). CETP is a 70 kDa plasma glycoprotein that is physically associated with HDL particles. It facilitates the transport of cholesteryl ester from HDL to apolipoprotein B-containing lipoproteins. This transfer is accompanied by transfer of triglycerides in the opposite direction. Thus, a decrease in CETP activity can result in an increase in the level of HDL cholesterol and a decrease in the level of very low density lipoprotein (VLDL) and low density lipoprotein (LDL). CETP can therefore simultaneously affect the concentrations of pro-atherogenic (for example, LDL) and anti-atherogenic (for example, HDL) lipoproteins.
Several CETP inhibitors are currently in various clinical phases of development for treating various aforementioned disorders. In spite of having various advantages, CETP inhibitors are proven to be difficult to formulate for oral administration. CETP inhibitors are of a highly lipophilic nature and have extremely low solubility in water. Due to their poor solubility, bioavailability of conventional oral compositions is very poor. The lipophilic nature of CETP inhibitors not only leads to low solubility but also tends to poor wettability, further reducing their tendency to be absorbed from the gastrointestinal tract. In addition to the low solubility, CETP inhibitors also tend to have significant, "food effect", where a significant difference in rate and amount of drug absorption is observed when the drug is administered with or without a meal. This "food effect", often complicates the dosing regimen and may require high dosing to achieve the desired therapeutic effect, resulting in potentially unwanted side effects.
Several attempts have been made to improve the solubility of CETP inhibitors, but have generally ended up with limited success. At the outset, most methods aimed at enhancing aqueous concentration and bioavailability of low-solubility drugs only offer moderate improvements. References describing improving the dissolution of poorly soluble drugs include: U.S. Patent Nos. 5,456,923, 5,993,858, 6,057,289, 6,096,338, 6,267,985, 6,280,770, 6,436,430, 6,451,339, 6,531,139, 6,555,558, 6,638,522, 6,962,931 and 7,374,779.
PATENT
WO 2014128564
https://www.google.co.in/patents/WO2014128564A2?cl=en

WO-2014076568
http://www.google.com/patents/WO2014076568A2?cl=en
EXAMPLES
In the following Examples 1-17, various compositions in accordance with the present application were prepared comprising 3-(((3,5-bis(trifluoromethyl)benzyl)(2- methyl-2H-tetrazol-5-yl)amino)methyl)-N,N-bis(cyclopropylmethyl)-8- methylquinolin-2-amine as the CETP inhibitor.:
EXAMPLE 1 :
Figure imgf000066_0001
1. 3-(((3,5-bis(trifluoromethyl)benzyl)(2-methyl-2H-tetrazol-5-yl)amino)methyl)- N,N-bis(cyclopropylmethyl)-8-methylquinolin-2-amineand hydroxypropyl methyl cellulose acetate succinate were mixed together in given solvent mixture to form clear solution.
2. To the solution of step I, Polyoxyl 35 castor oil and talc were added to form a homogenous suspension.
3. The suspension of step 2 was sprayed over inert sugar spheres and dried.
4. The drug layered spheres of step 3 were coated with dispersion made from given seal layer ingredients.
5. The coated spheres of step 4 were formulated further as capsule dosage form.

PATENT
WO 2013046045
https://www.google.co.in/patents/WO2013046045A1?cl=en
PATENT
WO 2013024358

PATENT
WO 2007075194
https://www.google.co.in/patents/WO2007075194A1?cl=en
Syntheis construction
Figure imgf000073_0001Figure imgf000073_0002Figure imgf000074_0001
Figure imgf000075_0001Figure imgf000075_0002
reddys1
Example 1
Synthesis of (3-{[3,5-bis trifluoromethyl-benzyl )-(2-cyclopropyImethyI-2H- tetrazole -5-yl)-amino]-methyl-}-8-methyI-quinolme-2-yl)-bis- cyclopropylmethyl-amine Step (i): Synthesis of 2~chloro-8-methyl-quinoline-3-carbaldehyde
Figure imgf000073_0001
DMF (1.22 g, 16.7 mmol) was taken in a flask equipped with a drying tube and POCl3 (7.32 g, 46.7 mmol) was added dropwise with stirring at 0° C. To this solution, TV-o-Tolyl acetamide (1.00 g, 6.7 mmol) was added and the solution was refluxed for 6 h at 90° C. The excess POCl3 was distilled off, water was added to the residue and this was stirred at room temperature for 10 min. The solid was filtered and dried under vacuum..This crude compound was purified over silica gel (100-200 mesh) using 6% ethyl acetate and petroleum ether to give the product as a yellowish solid (yield: 78%). 1H NMR (CDCl3, 200 MHz): δ 10.5 (s, IH)5 8.71 (s, IH), 7.83- 7.79 (m, IH), 7.74- 7.70 (m, IH), 7.56-7.49 (m, IH), 2.79 (s, 3H); m/z (EI-MS): 206 (M+, 100%). Step (ϋ): Synthesis of 2-(bis(cyclopropylmethyl)amino)-8-methylquinoline-3- carbaldehyde:
Figure imgf000073_0002
2-Chloro-8-methyl-quinoline-3-carbaldehyde (.115 g, 0.559 mmol), and potassium carbonate (0.231 g, 1.67 mmol) were put in a 25 mL two necked RB flask. To this, 3 mL of DMF was added followed by dropwise addition of bis- cyclopropylmethyl amine (0.083 g, 0.67 mmol). The reaction mixture was refluxed for 2 h and was cooled to RT. It was then poured on crushed ice (10 mL) and extracted with EtOAc (3 x 10 mL). The organic layer was washed with brine and dried over sodium sulphate. The solvent was evaporated under vacuum to give a yellow colored oil (0.081 g, 50%).
1H NMR (CDCl3, 400 MHz): δ 10.5 (s, IH), 8.71 (s, IH), 7.83- 7.79 (m, IH),
7.74-7.70 (m, IH), 7.56-7.49 (m, IH), 3.55-3.47 (m, 4H), 2.79 (s, 3H), 1.73-1.72
(m, 2H), 1.70-1.46 (m, 4H), 1.20-1.11 (m, 4H); m/z (ES-MS ): 295 (M+H-I5
100%); IR (neat, cm"1): 3385, 2948, 1691.
Step (iii): Synthesis of 3-((3,5-bis(trifluoromethyl)benzylamino)methyl)-N,N- bis(cyclopropylmethyl)-8-methylquinolin-2-amine
Figure imgf000074_0001
2-(Bis(cyclopropylmethyl)amino)-8-methylquinoline-3-carbaldehyde (0.081 g, 0.39 mmol), 3,5-bis-trifluoromethylbenzylamine (0.096 g, 0.39 mmol) and acetic acid (0.047 g, 0.78 mmol) were put in a 25 mL RB flask. To this, 2 rnL of methanol was added and stirred at RT for 15 min. Sodium cyanoborohydri.de (0.075 g, 0.77 mmol) was added portionwise and stirring was continued at RT for another 1 h. Methanol was removed from the reaction mixture under vacuum, water was added to this crude and was extracted with ethyl acetate (3 x 50 mL). The organic layer was washed with saturated NaHCO3 solution, brine and dried over sodium sulphate. The solvent was evaporated and the crude residue was purified by column chromatography over silica gel (100-200 mesh) eluting with 4% ethyl acetate in petroleum ether to give the title amine (0.142 g, yield: 99%). 1R NMR (CDCl3, 400 MHz): δ 7.89-7.86 (m, IH), 7.80 (m, IH), 7.75-7.74 (m, IH), 7.60-7.40 (m, 3H), 7.30-7.26 (m,lH), 4.12 (s, 2H), 3.88 (s, 2H), 3.24-3.22 (m, 4H), 2.72 (s, 3H), 0.99-0.92 (m, 2H), 0.44-0.35 (m, 4H), 0.11-0.05 (m, 4H); m/z (EI-MS ): 522 (M++l, 100%); IR (neat, cm"1): 3357, 2929, 2851.
Step (iv): Synthesis of N-(3,5-bis(trifluoromethyl)benzyl)-N-((2- (bis(cyclopropylmethyl)amino)-8-methylqumolin-3-yl)methyl)cyanamide
Figure imgf000075_0001
To a solution of 3-((3,5~bis(trifluoromethyl)benzylamino)methyl)-N,N- bis(cyclopropylmethyl)-8-methylquinolin-2-amine (0.176 g , 0.33 mmol ), obtained in step (iii) , in MeOH (4 mL) under N2 atmosphere was added sodium bicarbonate (0.056 g, 0.67 mmol ) followed by the addition of cyanogen bromide (0.063 g, 0.60 mmol). The reaction mixture was stirred at RT for 2 h. The solvent was removed under vacuum to give the crude residue which was dissolved in water, extracted with ethyl acetate and dried over sodium sulphate. The solvent was evaporated and concentrated in vacuo to afford N-(3,5-bis(trifluoromethyl)benzyl)- N-((2-(bis(cyclopropylmethyl)amino)-8-methylquinolin-3-yl)methyl)cyanamide (0.118 g, 64%).
1H NMR (CDCl3, 400 MHz ): δ 8.07 (s, IH) , 7.82 (s, IH), 7.70 (s, 2H), 7.56-7.55 (m, IH), 7.50-7.49 (m, IH), 4.49 (s, 2H), 4.23 (s, 2H), 3.17 -3.15 (m, 4H), 2.71 (s, 3H), 0.097-0.085 (m, 2H), 0.405-0.401 (m, 4H), 0.385-0.381 (m, 4H); m/z (ES- MS): 547 (M++l, 100%); IR(KBr ,Cm"1 ) : 2273, 1280.
Step (v): Synthesis of (3-{[(3,5-bistrifluoromethyl-benzyl)-(2H-tetrazol-5-yl)- amino]-methyl}-8-methyl-quinolin-2-yl)-bis-cyclopropylmethyl-amine
Figure imgf000075_0002
7V-(3,5-Bis(tiifluoromethyl)benzyl)-N-((2-(bis(cyclopropylmethyl)amino)- 8-methylqumolin-3-yl)methyl)cyanamide (0.118 g, 0.216 mmol), sodium azide (0.70 g 1.08 mmol) and ammonium chloride (.058 g, 1.08 mmol) were put in a RB flask under N2atmosphere. To this reaction mixture, DMF (2 mL) was added and was refluxed for 1 h. The reaction mixture was cooled to RT and ice was added to this and extracted with ethylacetate (3x10 mL). The combined organic layer was washed with brine, dried over sodium sulphate and then concentrated under vacuum to afford of (3-{[(3,5-bistrifluoromethyl-benzyl)-(2H-tetrazol-5-yl)- amino]-methyl}-8-methyl-quinolin-2-yl)-bis-cyclopropylmethyl-amine as a yellow solid (0.125 g, 99%).
1H NMR (CDCl3, 400 MHz ): δ 7.99 (s, IH) , 7.79 -7.74 (m, 4H ), 7.41-7.40 (m,
IH ), 7.33-7.31 (m, IH), 4.99 (s, 2H), 4.80 (s, 2H), 3.68 (s, 4H), 2.16 (s, IH) 1.56-
1.06 (m, HH); m/z (ES-MS): 578 (M++l, 100%); IR (KBr , cm"1) 3680 , 2922 ,
1660 , 1616.
METHYLATION SHOULD GIVE THE PRODUCT
Scheme 1
Figure imgf000046_0001
Figure imgf000046_0002
 
 
PATENT
WO 2006073973
http://www.google.co.in/patents/WO2006073973A2?cl=en
Example 47
Synthesis of [2-(bis-cycIopropylmethyI-amino)-8-methyl-quinolin-3-ylmethyI]-(3,5- bis-trifluoromethyl-benzyl)-carbamic acid methyl ester
Step (i): Synthesis of bis-cyclopropylmethyl-amine
(i) a. Synthesis of cyclopropanecarboxylic acid cyclopropylmethyl-amide:
Figure imgf000212_0002
Cyclopropyl carboxylic acid (1.0 g, 11.63 mmol) was added to a 50 mL two neck round bottom flask, along with DCM (25 mL). This mixture was cooled to 0° C, EDCI (4.15 g, 13.95 mmol) was added portionwise to the mixture with stirring under nitrogen atmosphere, and the temperature was maintained for 0.5 h. After this time, hydroxybenzotriazole (1.88 g, 13.95 mmol) was added to the 0° C mixture which was stirred for 10 min, then triethylamine (1.7 g, 11.63 mmol) was added, and stirring of the mixture was continued at the same temperature for another 0.5 h. Then, cyclopropylmethylamine (0.825 g, 11.63 mmol) was added, and the reaction was allowed to reach RT, and stirring was continued overnight. The solvent was then removed in vacuo, and the crude residue was purified by passing through a column over 60-120 silica gel, eluting with dichloromethane, to afford the title compound (1.6 g), yield: 87%. 1H NMR (CDCl3, 200 MHz): d 5.75 (br s, NH, D2O exchangeable), 3.17-3.16 (m, 2H), 1.00-0.80 (m, 4H), 0.77-0.67 (m, 2H), 0.56-0.43 (m, 2H), 0.24-0.16 (m, 2H) m/z (CI-MS): 139 (M+, 100%) (i) b. Synthesis of bis-cyclopropylmethyl-amine
Figure imgf000213_0001
To a suspension of lithium aluminum hydride (1.3 g, 9.35mmol) in 10 mL dry ether, a solution of N-cyclopentenoyl-ethylamine (1.7 g, 13.3 mmol) in dry ether (10 mL) was added under a nitrogen atmosphere. This reaction was stirred at RT for 8 h and the reaction mixture was then quenched with saturated sodium sulfate solution, filtered, and the precipitate was washed with diethyl ether. The filtrate was concentrated to afford the title amine (0.8 g), yield: 69%.
1H NMR (CDCl3, 200 MHz): d 5.75 (br s, NH, D2O exchangeable), 3.16-3.09 (m, 2H), 2.50-2.4 (m, 2H), 0.56-0.43 (m, 4H), 0.24-0.21 (m, 3H), 0.21-0.13 (m, 3H) m/z (ES-MS): 139 (M^+14, 100%)
Step (ii): Synthesis of [2-(bis-cyclopropylmethyl-amino)-8-methyl-quinolin-3-ylmethyl]- (S^-bis-trifluoromethyl-benzy^-carbamicacid methyl ester
Figure imgf000214_0001
The title compound was synthesized by using the same procedure as in Example 35, except using o-tolyl acetanilide in step (i) instead of acetanilide and bis- cyclopropylmethyl amine in step (iii), which yielded the desired product as a light yellow, viscous liquid (0.05 g), yield:40%, of purity 98.8% (HPLC: Symmetry Shield RP8, [0.01M KH2PO4: CH3CN], 217 nM, Rt12.719 min).
1H NMR (CDCl3, 400 MHz): d 7.7 (s, IH), 7.68-7.44 (m, 3H), 7.27-7.24 (m, 2H), 4.78- 4.65 (m, 2H), 4.47-4.4 (m, 2H), 3.8 (s, 3H), 3.16-3.14 (d, J=7Hz, 2H), 2.7 (s, 3H), 1.55 (s, 3H), 1.01-0.9(m, IH), 0.38-0.34 (m, 4H), 0.07-0.05 (m, 4H); m/z (CI-MS): 579 (M+, 100%)
Example 57
Synthesis of (3-{ [3,5-bis-trifluoromethyl-benzyl)-(2-methyl-2H-tetrazoIe-5-yl)- amino]-methyl}-8-methyl-quinolin-2-yl)-bis-cyclopropylmethyl-amine
Figure imgf000226_0001
The title compound was prepared as an oil by following the same synthetic procedures as in Example 52, except using {3-[(3,5-bis-trifluoromethyl-benzylamino)- methyl]-8-methyl-quinolin-2-yl}-bis-cyclopropylmethyl-amine in step (i) instead of {3- [(3,5-bis-trifluoromethyl-benzylamino)-methyl]-quinolin-2-yl}-cyclopentylmethyl-ethyl- amine (0.07 g), yield: 52%.
Purity: 95.53% (HPLC: Symmetry Shield RP8, [0.01M KH2PO4: CH3CN], 217 nM, Rt 9.538 min).
IR (neat, cm4) 3079, 2925, 1582;
1H NMR (CDCl3, 400 MHz): d 7.82 (s, IH), 7.69-7.67 (m, 2H), 7.44-7.41 (m, IH), 7.23- 7.2 (m, 3H), 4.91 (s, 2H), 4.65 (s, 2H), 4.21 (s, 3H), 3.29 -3.19 (m, 4H)5 2.71 (s, 3H), 1.01-1.00 (m, 2H), 0.99-0.83 (m, 2H), 0.39-0.34 (m, 3H), 0.08-0.07 (m, 3H). m/z (ES-MS): 604 (M++!, 100%)

Dr. Reddy’s announces start of Phase II study with the CETP inhibitor, DRL-17822 in dyslipidemia patients
Hyderabad, India, September 02, 2011: Dr Reddy’s Laboratories (NYSE: RDY) announced the initiation of dosing with DRL-17822 in patients with diagnosis of type II dyslipidaemia. DRL-17822, is a selective, orally bioavailable inhibitor of cholesteryl ester transfer protein (CETP), for the treatment and/or prevention of dyslipidaemia, atherosclerosis and associated cardiovascular disease.
The current study is being conducted under a CTA in a number of countries in Europe. The objective of the study is to evaluate the efficacy and safety of DRL-17822 in patients with Type-II dyslipidemia. This is a randomized, double blind, placebo controlled, parallel group study in 160 subjects. The primary outcome measure is to assess the elevation in HDL cholesterol and reduction in LDL cholesterol from baseline to end of treatment compared to placebo. Three doses (50, 150 & 300 mg) of DRL-17822 given once daily for 4 weeks will be evaluated during this study.
Three human Phase I studies with DRL-17822 had already been conducted in Europe, where DRL-17822 was shown to be safe and well tolerated. In these studies, the proof of mechanism had been demonstrated by dose-dependent inhibition of plasma CETP activity as well as by significant increase in HDL cholesterol & decrease in LDL cholesterol levels.
Cardiovascular disease is a leading cause of death among men and women worldwide. Among cardiovascular disorders, coronary heart disease (CHD), caused by atherosclerosis is the most common cause of morbidity and mortality. Stabilization and/or regression of atherosclerotic plaques may have a major impact on reducing the risk of acute coronary events. Low-density lipoprotein cholesterol lowering agents, primarily the statins, are the current mainstay in the pharmacological management of dyslipidaemia. However, significant residual cardiovascular risk remains despite use of statins.
Epidemiological and observational studies demonstrate that reduced high density lipoprotein cholesterol levels are a strong, independent predictor of CHD, suggesting that raising HDL cholesterol levels might afford clinical benefit in the reduction of cardiovascular risk. One approach to raise HDL level has been inhibition of CETP activity. Currently it is believed that, raising HDL cholesterol and lowering LDL cholesterol through CETP inhibition would lead to a significant benefit in terms of CHD risk reduction.
Dr. K. Anji Reddy, Founder Chairman, Dr. Reddy’s Laboratories added, “We are committed to delivering products of differentiated value in this area of high global clinical unmet need. We are excited to continue to advance our CETP program and look forward to the data from our Phase II study. This class of therapy could transform the treatment of CHD and DRL 17822 is in a position to be one of the front-running products in the class”.
Disclaimer
This press release includes forward-looking statements, as defined in the U.S. Private Securities Litigation Reform Act of 1995. We have based these forward-looking statements on our current expectations and projections about future events. Such statements involve known and unknown risks, uncertainties and other factors that may cause actual results to differ materially. Such factors include, but are not limited to, changes in local and global economic conditions, our ability to successfully implement our strategy, the market acceptance of and demand for our products, our growth and expansion, technological change and our exposure to market risks. By their nature, these expectations and projections are only estimates and could be materially different from actual results in the future.
About Dr. Reddy’s
Dr. Reddy’s Laboratories Ltd. (NYSE: RDY) is an integrated global pharmaceutical company, committed to providing affordable and innovative medicines for healthier lives. Through its three businesses - Pharmaceutical Services and Active Ingredients, Global Generics and Proprietary Products – Dr. Reddy’s offers a portfolio of products and services including APIs, custom pharmaceutical services, generics, biosimilars, differentiated formulations and NCEs. Therapeutic focus is on gastro-intestinal, cardiovascular, diabetology, oncology, pain management, anti-infective and pediatrics. Major markets include India, USA, Russia and CIS, Germany, UK, Venezuela, S. Africa, Romania, and New Zealand. For more information, log on to: www.drreddys.com

For more information please contact:
Investors and Financial Analysts:
Kedar Upadhye at kedaru@drreddys.com / +91-40-66834297
Raghavender R at raghavenderr@drreddys.com / +91-40-49002135
Milan Kalawadia (USA) at mkalawadia@drreddys.com / +1 908-203-4931
Media:
S Rajan at rajans@drreddys.com / +91-40- 49002445
WO2005011634A1 *Jul 21, 2004Feb 10, 2005William John CuratoloDosage forms providing controlled release of cholesteryl ester transfer protein inhibitors and immediate release of hmg-coa reductase inhibitors
WO2006073973A2 *Dec 28, 2005Jul 13, 2006Reddy Us Therapeutics IncNovel benzylamine derivatives as cetp inhibitors
WO2006129167A1 *May 22, 2006Dec 7, 2006Pfizer Prod IncPHARMACEUTICAL COMPOSITIONS OF CHOLESTERYL ESTER TRANSFER PROTEIN INHIBITORS AND HMG-CoA REDUCTASE INHIBITORS
WO2007128568A1 *May 8, 2007Nov 15, 2007Novartis AgBicyclic derivatives as cetp inhibitors
EP0298666A2 *Jul 1, 1988Jan 11, 1989American Home Products CorporationSpray dried ibuprofen compositions
EP1741424A2 *Jul 27, 1998Jan 10, 2007Pfizer Products Inc.Solid pharmaceutical dispersions with enhanced bioavailabilty
US5456923Dec 23, 1993Oct 10, 1995Nippon Shinyaku Company, LimitedMethod of manufacturing solid dispersion
US5474989Mar 1, 1994Dec 12, 1995Kurita Water Industries, Ltd.Drug composition
US5985326May 30, 1996Nov 16, 1999Icos CorporationMethod of producing a solid dispersion of a poorly water soluble drug
US6350786Sep 7, 1999Feb 26, 2002Hoffmann-La Roche Inc.Stable complexes of poorly soluble compounds in ionic polymers
US6548555Jan 31, 2000Apr 15, 2003Pfizer IncBasic drug compositions with enhanced bioavailability
US6638522Dec 10, 1999Oct 28, 2003Pharmasolutions, Inc.Microemulsion concentrate composition of cyclosporin
US6730679Mar 20, 1997May 4, 2004Smithkline Beecham CorporationPharmaceutical formulations
US7008640Jul 16, 2001Mar 7, 2006Yamanouchi Pharmaceutical Co., Ltd.Pharmaceutical composition for oral use with improved absorption
US7034013Mar 19, 2002Apr 25, 2006Cydex, Inc.Formulations containing propofol and a sulfoalkyl ether cyclodextrin
US7037528Jun 5, 2001May 2, 2006Baxter International Inc.Microprecipitation method for preparing submicron suspensions
US7078057Dec 19, 2000Jul 18, 2006Kerkhof Nicholas JProcess for producing nanometer particles by fluid bed spray-drying
US7081255Aug 14, 2002Jul 25, 2006Janssen Pharmaceutica, N.V.Antifungal compositions with improved bioavailability
US8030359Feb 9, 2007Oct 4, 2011Merck Sharp & Dohme Corp.Polymer formulations of CETP inhibitors
US20060178514Dec 28, 2005Aug 10, 2006Anima BaruahNovel benzylamine derivatives as CETP inhibitors
Reference
1*EINFAL T ET AL: "Methods of amorphization and investigation of the amorphous state", ACTA PHARMACEUTICA 20130901 CROATIAN PHARMACEUTICAL SOCIETY DEU, vol. 63, no. 3, 1 September 2013 (2013-09-01), pages 305-334, XP002721717, ISSN: 1330-0075
2*KAI TOSHIYA ET AL: "Oral absorption improvement of poorly soluble drug using solid dispersion technique", CHEMICAL AND PHARMACEUTICAL BULLETIN (TOKYO), vol. 44, no. 3, 1996, pages 568-571, XP002721716, ISSN: 0009-2363
3*KIM TAE-WAN ET AL: "Characterization of dual layered pellets for sustained release of poorly water-soluble drug", CHEMICAL & PHARMACEUTICAL BULLETIN (TOKYO), vol. 55, no. 7, July 2007 (2007-07), pages 975-979, XP002721715, ISSN: 0009-2363
4*KIM TAE-WAN ET AL: "Modified release of coated sugar spheres using drug-containing polymeric dispersions.", ARCHIVES OF PHARMACAL RESEARCH JAN 2007, vol. 30, no. 1, January 2007 (2007-01), pages 124-130, XP002721714, ISSN: 0253-6269
Citing PatentFiling datePublication dateApplicantTitle
WO2014128564A2 *Feb 21, 2014Aug 28, 2014Dr. Reddy's Laboratories Ltd.Pharmaceutical compositions of cetp inhibitors
SEE
http://circ.ahajournals.org/cgi/content/meeting_abstract/122/21_MeetingAbstracts/A13981
 
////////
Cn1nc(nn1)N(Cc2cc5cccc(C)c5nc2N(CC3CC3)CC4CC4)Cc6cc(cc(c6)C(F)(F)F)C(F)(F)F

Lefucoxib (乐福昔布)


C3
Lefucoxib (乐福昔布)
5-(3,4-dimethyl-phenyl)-1-methanesulfonyl-3-trifluoromethol-pyrazole
1 [4- (methylsulfonyl) phenyl] -3-trifluoromethyl-5- (3,4-dimethylphenyl) - pyrazole
CAS 849048-84-6
394.411, C19 H17 F3 N2 O2 S
IND FILED
Prostaglandin G/H Synthase 2 (PTGS2; COX-2) Inhibitors
A COX-2 inhibitor potentially for the treatment of rheumatoid arthritis.
cyclooxygenase-2 (COX-2) inhibitor
National Center of Biomedical Analysis
Example 1
1 [4- (methylsulfonyl) phenyl] -3-trifluoromethyl-5- (3,4-dimethylphenyl) - pyrazole (I1)
1- (3,4- two toluene-yl) -4,4,4-trifluoro-methyl - D-1,3-dione (IV1) of sodium metal was weighed 2.3g (0.1mol) was added 50ml of anhydrous toluene to prepare a sodium sand. After cooling, ethanol was added dropwise 12ml, and then heated at 60 ℃, complete reaction of sodium metal. After cooling to room temperature, was added 3,4-dimethylphenyl ethanone 23.8g (0.1mol) and trifluoroacetic ethyl acetate 20ml (0.2mol), reacted at 100 ℃ 5 hours. Toluene was distilled off under reduced pressure, a 10% aqueous hydrochloric acid was added, the pH was adjusted to 2-3, extracted with ethyl acetate, washed with water, dried over anhydrous MgSO4, ethyl acetate was distilled off under reduced pressure. Then under reduced pressure, distillation, collecting fractions 105-107 ℃ / 0.7mmHg, was 14.6g, 60% yield.
1- [4- (methylsulfonyl) phenyl] -3-trifluoromethyl-5- (3,4-dimethylphenyl) - pyrazole (I1) take the above-prepared substituted (IV1) 2.38g (0.01mol ), 15ml of ethanol, then added p-methanesulfonyl phenyl hydrazine salt alkoxide 2.3g (0.01ml). Was refluxed for 15 hours. Place the refrigerator overnight, the crystals were collected by filtration, recrystallized from ethanol, mp 129-31 ℃, to give 3.1 g.
Elemental analysis: C19H17F3N2O2S Calculated: C, 57.86; H, 4.34; N, 7.10 Found: C, 57.97; H, 4.29; N, 7.20MS (m / z): 395 (M + 1)
C4

CN101497585BJan 31, 2008Jan 12, 2011中国科学院理化技术研究所Method for photocatalytic synthesis of 1,3,5-trisubstituted-2-pyrazole derivative
//////////