AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

EMA reviews Medicines manufactured at U.S. Company

 regulatory  Comments Off on EMA reviews Medicines manufactured at U.S. Company
Jul 142016
 

Following the issuance of two Non-Compliance Reports for two sites of the US based company, EMA has started a review of medicines manufactured by Pharmaceutics International Inc., USA.

The European Medicines Agency (EMA) has started a review of medicines manufactured by Pharmaceutics International Inc., USA. This follows the issuance of two Non-Compliance Reports for two sites of the US based company after an inspection in February 2016 conducted by the MHRA (the medicines regulatory agency in the United Kingdom) which highlighted several shortcomings in relation to good manufacturing practice (GMP).

Pharmaceutics International Inc. manufactures the centrally authorised medicine Ammonaps (sodium phenylbutyrate) and is also the registered manufacturing site for some other medicines that have been authorised through national procedures in the European Union (EU).

This inspection which was a follow-up to an inspection in June 2015 aimed to assess whether corrective measures agreed previously had been appropriately implemented. It found that shortcomings remained, which included insufficient measures to reduce the risk that traces of one medicine could be transferred to another (cross-contamination), as well as problems with the way data were generated and checked and deficiencies in the systems for ensuring medicines’ quality (quality assurance).

EMA’s Committee for Medicinal Products for Human Use (CHMP) will now review the impact of the inspection findings on the products’ overall benefits and risks and make a recommendation as to whether any changes are needed to their marketing authorisations.

There is no evidence that patients have been put at risk by this issue. However, as a precautionary measure, medicines from this site will no longer be supplied to the EU unless they are considered to be ‘critical’ to public health. Criticality will be assessed by national medicines regulatory agencies for their territories, taking into account alternatives and any impact of shortages on patients. In case where a medicine manufactured at this site is considered not critical in a member state it will no longer be supplied in this member state and any medicine remaining on the market will be recalled.

Source: EMA Press Release

Pharmaceutics International Inc., USA

/////////// EMA,  Medicines,  manufactured, U.S. Company, Pharmaceutics International Inc., USA

Share

Cipla to invest in South Africa’s first biosimilars production facility

 INDIA, MONOCLONAL ANTIBODIES  Comments Off on Cipla to invest in South Africa’s first biosimilars production facility
Jul 142016
 

Cipla to invest in South Africa’s first biosimilars production facility
Indian-based pharmaceutical and biotechnology company Cipla will invest more than R1.3bn ($19.34m) in the first advanced biotech manufacturing facility in South Africa for the production of biosimilars.

Indian-based pharmaceutical and biotechnology company Cipla will invest more than R1.3bn ($19.34m) in the first advanced biotech manufacturing facility in South Africa for the production of biosimilars.

The investment will be carried out by South African subsidiary Cipla BioTec…………………cont

read at

http://www.pharmaceutical-technology.com/news/newscipla-invest-south-africas-first-biosimilars-production-facility-4945516?WT.mc_id=DN_News

Cipla Managing director and global CEO Subhanu Saxena

 

Dr Y.K. Hamied,

Department of Trade and Industries Special Economic Zone of Dube Tradeport, DURBAN, SOUTHAFRICA

 

///Cipla, South Africa, biosimilars,  production facility, Dube Tradeport, Cipla BioTec Pvt Ltd, Durban, SOUTHAFRICA

Share

Besifloxacin hydrochloride (Besivance)

 Uncategorized  Comments Off on Besifloxacin hydrochloride (Besivance)
Jul 132016
 

Besifloxacin.png

Besifloxacin

SS 734, BOL 303224A, ISV-403

MW 430.301, MF C19H21ClFN3O3

141388-76-3 CAS

7-[(3R)-3-aminoazepan-1-yl]-8-chloro-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid

(R)-(+)-7-(3-amino-2,3,4,5,6,7-hexahydro-1H-azepin-1-yl)-1,4-dihydro-4-oxoquinoline-3-carboxylic acid

(R) -7- (3- amino-hexahydro-azepin -1H- mushroom-1-yl) -8-chloro-1-cyclopropylmethyl -6-fluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid

Synthesis of the molecule (R)-(+)-7-(3-amino-2,3,4,5,6,7-hexahydro-1H-azepin-1-yl)-1,4-dihydro-4-oxoquinoline-3-carboxylic acid is disclosed in U.S. Pat. No. 5,447,926,

Besifloxacin is a fourth generation fluoroquinolone-type opthalmic antibiotic for the treatment of bacterial conjunctivitis. FDA approved on May 28, 2009. by Bausch & Lomb, for the treatment of non-viral bacterial conjunctivitis

Besifloxacin, (+)-7-[(3R)-3-aminohexahydro-1H-azepin-1-yl]-8-chloro-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid hydrochloride, developed by SS Pharmaceutical (SSP) Co.Ltd. was a fourth-generation fluoroquinolone antibiotic . Besifloxacin hydrochloride eye drop was used to treat bacterial conjunctivitis caused by aerobic and facultative Gram-positive microorganisms and aerobic and facultative Gram-negative microorganisms

Besifloxacin.png

Besifloxacin (INN/USAN) is a fourth-generation fluoroquinolone antibiotic. The marketed compound is besifloxacin hydrochloride. It was developed by SSP Co. Ltd., Japan, and designated SS734. SSP licensed U.S. and European rights to SS734 for ophthalmic useto InSite Vision Incorporated (OTCBB: INSV) in 2000. InSite Vision developed an eye drop formulation (ISV-403) and conducted preliminary clinical trials before selling the product and all rights to Bausch & Lomb in 2003.[1]

The eye drop was approved by the United States Food and Drug Administration (FDA) on May 29, 2009 and marketed under the trade name Besivance.[2]

Name Dosage Strength Route Labeller Marketing Start Marketing End
Besivance suspension 6 mg/mL ophthalmic Bausch & Lomb Incorporated 2009-05-28 Not applicable Us
Besivance suspension 0.6 % ophthalmic Bausch & Lomb Inc 2010-01-27 Not applicable Canada
Besivance suspension 6 mg/mL ophthalmic Physicians Total Care, Inc. 2011-07-13 Not applicable Us

405165-61-9 CAS

Besifloxacin Hydrochloride

Besifloxacin hydrochloride is a fourth-generation fluoroquinolone antibiotic.
IC50 Value:
Target: Antibacterial
Besifloxacin has been found to inhibit production of pro-inflammatory cytokines in vitro. Besifloxacin is a novel 8-chloro-fluoroquinolone agent with potent, bactericidal activity against prevalent and drug-resistant pathogens.besifloxacin is the most potent agent tested against gram-positive pathogens and anaerobes and is generally equivalent to comparator fluoroquinolones in activity against most gram-negative pathogens. Besifloxacin demonstrates potent, broad-spectrum activity, which is particularly notable against gram-positive and gram-negative isolates that are resistant to other fluoroquinolones and classes of antibacterial agents.

Clinical Information of Besifloxacin Hydrochloride

Product Name Sponsor Only Condition Start Date End Date Phase Last Change Date
Besifloxacin Hydrochloride Bucci Laser Vision Institute Bacterial infection 31-MAY-11 31-DEC-11 Phase 4 05-JUN-13
Bucci Laser Vision Institute 31-MAY-11 31-DEC-11 Phase 4 03-JUN-13
Innovative Medical Services 30-SEP-10 31-OCT-12 Phase 4 11-SEP-13
Ophthalmology Consultants, Ltd Cataract 30-SEP-10 28-FEB-11 Phase 4 11-SEP-13
University of Louisville Blepharitis 31-AUG-11 31-OCT-11 Phase 4 01-DEC-11

Pharmacodynamics

Besifloxacin is a fluoroquinolone that has a broad spectrum in vitro activity against a wide range of Gram-positive and Gram-negativeocular pathogens: e.g., Corynebacterium pseudodiphtheriticum, Moraxella lacunata, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus hominis, Streptococcus mitis, Streptococcus oralis, Streptococcus pneumoniae and Streptococcus salivarius. Besifloxacin has been found to inhibit production of pro-inflammatory cytokines in vitro.[3] The mechanism of action of besifloxacin involves inhibition of two enzymes which are essential for the synthesis and replication of bacterial DNA: the bacterialDNA gyrase and topoisomerase IV.

Medical Use

Besifloxacin is indicated in the treatment of bacterial conjunctivitis caused by sensitive germs,[4] as well as in the prevention of infectious complications in patients undergoing laser therapy for the treatment of cataracts.[5][6]

Adverse Effects

During the treatment, the most frequently reported ocular adverse reaction was the appearance of conjunctival redness (approximately 2% of patients). Other possible adverse reactions, reported in subjects treated with besifloxacin were: eye pain, itching of the eye, blurred vision, swelling of the eye or eyelid.

MORE SYNTHESIS COMING, WATCH THIS SPACE…………………..

 

STR1

 

 

 

STR1

STR1

 

 

 

STR1

 

 

 

PATENT

WO 2010111116

https://www.google.com/patents/WO2010111116A1?cl=en

 

PATENT

CN 104592196

https://www.google.com/patents/CN104592196A?cl=en

STR1

The method comprises performing condensation reaction of 1-​cyclopropyl-​6,​7-​dichloro-​1,​4-​dihydro-​4-​oxy-​3-​quinoline carboxylic acid with (R)​-​3-​aminohexahydroazepine in the presence of org. base in org. solvent I at 45°C-​solvent b.p. temp. under refluxing, washing with acid, vacuum concg. to obtain (R)​-​7-​(3-​amino-​hexahydro-​1H-​azepine-​1-​yl)​-​1-​cyclopropyl-​6-​fluoro-​1,​4-​dihydro-​4-​oxy-​3-​quinoline carboxylic acid, dissolving in 5-​10 fold org. solvent II, reacting with thionyl chloride at 0-​40°C, and vacuum concg. to obtain (R) -7- (3- amino-hexahydro-azepin -1H- mushroom-1-yl) -8-chloro-1-cyclopropylmethyl -6-fluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid hydrochloride

Preparation method of the present invention provides hydrochloride Besifloxacin, comprising the steps of:

(1), in three _6 flask of 1-cyclopropyl, 6,7-difluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid 10g of acetonitrile added 100mL, was added (R ) -3-amino-hexahydro-aza mushroom 4.73g and 7.2mL of triethylamine was heated at reflux for 5h TLC plate detection point, the reaction was complete spin dry plus 100mL dissolved in chloroform and then 200mL 1M hydrochloric acid and washed twice with saturated brine The organic phase to pH 4-6, the organic phase was poured into the jar and dried to obtain the single (R) -7- (3- amino-hexahydro-azepin -1H- leather-yl) cyclopropyl-6 -1_ fluoro-1,4-dihydro-4-oxo-3-quinoline-carboxylic acid in chloroform solution; spin-dried to give (R) -7- (3- amino-hexahydro-azepin -1H- leather-yl) cyclopropyl-6-fluoro-1,4-dihydro-4-oxo-quinoline-3-carboxylic acid.

(2), obtained in the previous step (R) -7- (3_ atmosphere atmosphere -1H- gas hybrid group six leather-1-yl) cyclopropyl-6-fluoro-1,4 _1_ dihydro-4-oxo-3-quinolinecarboxylic acid in chloroform solution was cooled to 0 ° C, was slowly added dropwise under constant stirring 18mL S0C12, temperature does not exceed 5 ° C added, the mixture was stirred at 0 ° C after 2h l to room temperature, TLC detection, after completion of the reaction was evaporated to dryness to column chromatography to give (R) -7- (3- amino-hexahydro-azepin -1H- mushroom-1-yl) -8-chloro-1-cyclopropylmethyl -6-fluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid hydrochloride 5. 12g.

 

PATENT

US 20110144329

https://www.google.com/patents/US20110144329

EXAMPLE 1Preparation of Besifloxacin Free Base Solid

Besifloxacin free base was prepared from besifloxacin hydrochloride addition salt.

An amount of about 5 g of besifloxacin HCl (HCl addition salt of besifloxacin made, for example, by the method of U.S. Pat. No. 5,447,926; which is incorporated herein by reference in its entirety) was added to about 750 ml of water. The besifloxacin HCl was allowed to dissolve in said water. Twenty milliliters of 1N NaOH solution were added slowly to the besifloxacin aqueous solution while stirring (final pH 10.2). Besifloxacin free base started to precipitate. Eight milliliters of 1N HCl solution were added slowly while stirring (final pH of 9.7). The resulting mixture was allowed to mix for 2 hours while besifloxacin free base continued to precipitate. At the end of 2 hours, the precipitated besifloxacin free base was filtered through a Millipore type RA 1.2 μm filter. The besifloxacin free base thus collected was dried in a vacuum oven at room temperature. 4.35 g of besifloxacin free base was recovered.

FIG. 1 shows a UV absorption spectrum of besifloxacin free base starting material of Example 1.

FIG. 3 shows an IR spectrum of free base starting material of Example 1.

PATENT

https://www.google.com/patents/CN103044397A?cl=en

Figure CN103044397AD00041

Example 6 (R) -7_ (3- amino-hexahydro–1H- diazepan-1-yl) -8_ chloro-1-cyclopropyl-6-fluoro-1,4- Hydrogen oxo – quinoline-3-carboxylic acid (Besifloxacin). [0021] The reaction vessel was added chloroform (50ml) as a reaction solvent, in the case of a solid material was added with stirring (III) (3. 59g, O. Olmol), until the intermediate (III) is completely dissolved, was added dropwise under ice- chlorosulfonic acid, stirred for I hour under ice-cooling, gradually warmed to room temperature, stirred for 6 hours, and then reacted at reflux temperature for 6 hours. After completion of the reaction by TLC, the reaction solution was cooled to 0 ° C, white solid was precipitated, filtered, washed with a small amount of dichloromethane to give a crude product besifloxacin (3. 65g, 93. 01%). [0022] Example 7 (R) -7_ (3- amino-hexahydro–1H- diazepan-1-yl) -8_ chloro-1-cyclopropyl-6-fluoro-1,4- Hydrogen oxo – quinoline-3-carboxylic acid (Besifloxacin). [0023] The reaction vessel was added chloroform (50ml) as a reaction solvent, in the case of a solid material was added with stirring (III) (3. 59g, 0. Olmol), until the intermediate (III) is completely dissolved, was added dropwise under ice- chlorosulfonic acid was stirred for I hour under ice-cooling, gradually warmed to room temperature, stirred for 6 hours, and then reacted at reflux temperature for 12 hours. After completion of the reaction by TLC, the reaction solution was cooled to 0 ° C, the precipitated white solid was filtered , washed with a little dichloromethane to give Besifloxacin crude (3. 05g, 77. 22%).

PAPER

Molbank 2013, 2013(2), M801; doi:10.3390/M801
Short Note
(R)-7-(Azepan-3-ylamino)-8-chloro-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic Acid Hydrochloride
Supplementary File 3:Support Information (PDF, 340 KB)
Download PDF [188 KB, 27 May 2013; original version 22 May 2013]
R&D Center, Jiangsu Yabang Pharmaceutical Group, Changzhou 213200, China
In this paper (R)-7-(azepan-3-ylamino)-8-chloro-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid hydrochloride 1was isolated and identified as the N-substituted regioisomer of besifloxacin, which has been synthesized from the reaction of 8-chloro-1-cyclopropyl-6,7-difluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid 3 with (R)-tert-butyl 3-aminoazepane-1-carboxylate 2in acetonitrile as solvent in 37% yield. The chemical structure of compound 1 was established on the basis of 1H-NMR, 13C-NMR, mass spectrometry data and elemental analysis

REGIOMER OF BESIFLOXACIN

 

Besifloxacin.pngBESIFLOXACIN

 

STR1

STR1

References

  1.  “InSite Vision Reaches Agreement to Sell ISV-403 to Bausch & Lomb” (Press release). InSite Vision. 2003-12-19. Retrieved 2009-08-15.
  2.  “Bausch & Lomb Receives FDA Approval of Besivance, New Topical Ophthalmic Antibacterial for the Treatment of Bacterial Conjunctivitis (“Pink Eye”)” (Press release). Bausch & Lomb. 2009-05-29. Retrieved 2009-05-29.
  3.  Zhang JZ, Ward KW (January 2008). “Besifloxacin, a novel fluoroquinolone antimicrobial agent, exhibits potent inhibition of pro-inflammatory cytokines in human THP-1 monocytes”. J. Antimicrob. Chemother. 61 (1): 111–6. doi:10.1093/jac/dkm398. PMID 17965029.
  4.  Malhotra R, Ackerman S, Gearinger LS, Morris TW, Allaire C (December 2013). “The safety of besifloxacin ophthalmic suspension 0.6 % used three times daily for 7 days in the treatment of bacterial conjunctivitis”. Drugs in R&D 13 (4): 243–52. doi:10.1007/s40268-013-0029-1. PMC 3851703. PMID 24142473. Retrieved 2015-01-06.
  5.  Majmudar PA, Clinch TE (May 2014). “Safety of besifloxacin ophthalmic suspension 0.6% in cataract and LASIK surgery patients”. Cornea33 (5): 457–62. doi:10.1097/ICO.0000000000000098. PMC 4195578. PMID 24637269. Retrieved 2015-01-06.
  6.  Nielsen SA, McDonald MB, Majmudar PA (2013). “Safety of besifloxacin ophthalmic suspension 0.6% in refractive surgery: a retrospective chart review of post-LASIK patients”. Clinical Ophthalmology (Auckland, N.Z.) 7: 149–56. doi:10.2147/OPTH.S38279. PMC 3552478. PMID 23355771. Retrieved 2015-01-06.

 

CLIPS

Besifloxacin hydrochloride (Besivance) Besifloxacin is a fourth-generation fluoroquinolone antibiotic which is marketed as besifloxacin hydrochloride. It was originally developed by the Japanese firm SSP Co. Ltd and designated SS734. SSP then licensed U.S. and European rights of SS734 for ophthalmic use to InSite Vision, Inc., in 2000, who then developed an eye drop formulation (ISV-403) and conducted preliminary clinical trials before selling the product and all rights to Bausch & Lomb in 2003.

The eye drop was approved by the United States Food and Drug Administration (FDA) on May 29, 2009 and marketed under the trade name Besivance.24a

Besifloxacin has been found to inhibit production of pro-inflammatory cytokines in vitro. The synthesis of besifloxacin commences with commercially available ethyl 3-(3-chloro-2,4,5-trifluorophenyl)-3-oxopropanoate (13, Scheme3).24b

Condensation of this ketoester with triethyl orthoformate resulted in a mixture of vinylogous esters 14. Substitution with cyclopropanamine converts 14 to the vinylogous amide 15 as an unreported distribution of cis- and trans-isomers. This mixture was treated with base at elevated temperature to give 16.

Presumably, the trans-isomer isomerizes to the cis-isomer, which subsequently undergoes an intramolecular nucleophilic aromatic substitution with concomitant saponification to construct quinolone acid 16.

Quinolone 16 is then subjected to another nucleophilic substitution involving readily available iminoazepine 17 and the displacement reaction proceeds regioselectively to furnish the atomic framework of besifloxacin (18).

Acidic methanolysis of 18 at elevated temperature gave besiflozacin (III).

str1

24. (a) Bertino, J. S.; Zhang, J.-Z. Expert Opin. Pharmacother. 2009, 10, 2545; (b) Harms, A. E.; Arul, R.; Soni, A. K. U.S. 2009561283 A1, 2009.

US5447926 * Sep 16, 1994 Sep 5, 1995 Ss Pharmaceutical Co., Ltd. Quinolone carboxylic acid derivatives
Citing Patent Filing date Publication date Applicant Title
CN104458945A * Nov 27, 2014 Mar 25, 2015 广东东阳光药业有限公司 Separation and measurement method of besifloxacin hydrochloride and isomer of besifloxacin hydrochloride
CN102659761A * Apr 27, 2012 Sep 12, 2012 常州亚邦制药有限公司 Method for preparing besifloxacin hydrochloride
US5385900 * Nov 8, 1993 Jan 31, 1995 Ss Pharmaceutical Co., Ltd. Quinoline carboxylic acid derivatives
Reference
1 * 黄山等: “克林沙星的 2, 4, 5-三氟苯甲酸路线合成“, 《中国医药工业杂志》, vol. 31, no. 8, 31 December 2000 (2000-12-31)
Citing Patent Filing date Publication date Applicant Title
CN103709100A * Dec 31, 2013 Apr 9, 2014 南京工业大学 Preparation method of 8-chloroquinolone derivatives
Besifloxacin
Besifloxacin.png
Besifloxacin-3D-balls.png
Systematic (IUPAC) name
7-[(3R)-3-Aminoazepam-1-yl]-8-chloro-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid
Clinical data
Trade names Besivance
AHFS/Drugs.com Monograph
MedlinePlus a610011
License data
Routes of
administration
Ophthalmic
Legal status
Legal status
Identifiers
CAS Number 141388-76-3
ATC code S01AE08 (WHO)
PubChem CID 10178705
ChemSpider 8354210
UNII BFE2NBZ7NX Yes
ChEMBL CHEMBL1201760
Chemical data
Formula C19H21ClFN3O3
Molar mass 393.84 g·mol−1
Patent Number Pediatric Extension Approved Expires (estimated)
US5,447,926 No 1995-09-05 2012-09-05 Us
US5447926 No 1996-04-13 2016-04-13 Us
US6,685,958 No 2004-02-03 2021-06-20 Us
US6,699,492 No 2004-03-02 2019-03-31 Us
US6685958 No 2001-06-29 2021-06-29 Us
US6699492 No 1999-03-31 2019-03-31 Us
US8415342 No 2010-11-07 2030-11-07 Us
US8481526 No 2011-01-09 2031-01-09 Us
US8604020 No 2010-03-12 2030-03-12 Us
US8937062 No 2009-11-13 2029-11-13 Us

 

  1. O’Brien TP: Besifloxacin ophthalmic suspension, 0.6%: a novel topical fluoroquinolone for bacterial conjunctivitis. Adv Ther. 2012 Jun;29(6):473-90. doi: 10.1007/s12325-012-0027-7. Epub 2012 Jun 20. [PubMed:22729919 ]
  2. Proksch JW, Granvil CP, Siou-Mermet R, Comstock TL, Paterno MR, Ward KW: Ocular pharmacokinetics of besifloxacin following topical administration to rabbits, monkeys, and humans. J Ocul Pharmacol Ther. 2009 Aug;25(4):335-44. doi: 10.1089/jop.2008.0116. [PubMed:19492955 ]
  3. Besifloxacin Hydrochloride

    [1]. Wang Z, Wang S, Zhu F, Chen Z, Yu L, Zeng S. Determination of enantiomeric impurity in besifloxacin hydrochloride by chiral high-performance liquid chromatography with precolumn derivatization. Chirality. 2012 Jul;24(7):526-31. doi: 10.1002/chir.22042.
    Abstract
    Besifloxacin hydrochloride is a novel chiral broad-spectrum fluoroquinolone developed for the treatment of bacterial conjunctivitis. R-besifloxacin hydrochloride is used in clinics as a consequence of its higher antibacterial activity. To establish an enantiomeric impurity determination method, some chiral stationary phases (CSPs) were screened. Besifloxacin enantiomers can be separated to a certain extent on Chiral CD-Ph (Shiseido Co., Ltd., Japan), Chiral AGP, and Crownpak CR (+) (Daicel Chemical IND., Ltd., Japan). However, the selectivity and sensitivity were both unsatisfactory on these three CSPs. Therefore, Chiral AGP, Chiral CD-Ph, and Crownpak CR (+) were not used in the enantiomeric impurity determination of besifloxacin hydrochloride. The separation of enantiomers of besifloxacin was further performed using a precolumn derivatization chiral high-performance liquid chromatography method. 2,3,4,6-Tetra-O-acetyl-beta-D-glucopyranosyl isothiocyanate was used as the derivatization reagent. Besifloxacin enantiomer derivates were well separated on a C(18) column (250 × 4.6 mm, 5 μm) with a mobile phase that consisted of methanol-KH(2)PO(4) buffer solution (20 mM; pH 3.0) (50:50, v/v). Selectivity, sensitivity, linearity, accuracy, precision, stability, and robustness of this method were all satisfied with the method validation requirement. The method was suitable for the quality control of enantiomeric impurity in besifloxacin hydrochloride.

    [2]. Hussar DA. New drugs: golimumab, besifloxacin hydrochloride, and artemether/lumefantrine. J Am Pharm Assoc (2003). 2009 Jul-Aug;49(4):570-4.

    [3]. Nafziger AN, Bertino JS Jr. Besifloxacin ophthalmic suspension for bacterial conjunctivitis. Drugs Today (Barc). 2009 Aug;45(8):577-88.
    Abstract
    Besifloxacin hydrochloride ophthalmic suspension 0.6% (Besivance) is a recently approved fluoroquinolone for the topical treatment of bacterial conjunctivitis. The drug is rapidly bactericidal against common bacterial pathogens causing conjunctivitis, i.e., coagulase-negative Staphylococcus, Streptococcus pneumoniae, Staphylococcus aureus and Haemophilus influenzae as well as against other less common organisms. In addition to being a potent agent against Gram-positive and Gram-negative pathogens including those resistant to other fluoroquinolones, besifloxacin has balanced DNA gyrase and topoisomerase IV activity, which should slow the development of resistance. Topical administration achieves high sustained concentrations in human tears and good ocular tissue penetration in animals while demonstrating an excellent safety profile. Besifloxacin’s pharmacokinetic and pharmacodynamic characteristics meet the criteria for successful eradication of many Gram-positive and Gram-negative bacteria while demonstrating minimal systemic exposure. The biochemical properties, achievement of target pharmacokinetic/pharmacodynamic goals and the restriction of besifloxacin to topical ophthalmic use should result in slower development of bacterial resistance, making besifloxacin a new, appealing option for empiric therapy in acute bacterial conjunctivitis.

    [4]. Proksch JW, Ward KW. Ocular pharmacokinetics/pharmacodynamics of besifloxacin, moxifloxacin, and gatifloxacin following topical administration to pigmented rabbits. J Ocul Pharmacol Ther. 2010 Oct;26(5):449-58.
    Abstract
    PURPOSE: The purpose of this investigation was to evaluate the ocular pharmacokinetic/pharmacodynamic (PK/PD) relationship for besifloxacin, moxifloxacin, and gatifloxacin using rabbit ocular PK data, along with in vitro minimum inhibitory concentration (MIC90) values against methicillin- and ciprofloxacin-resistant Staphylococcus aureus (MRSA-CR) and Staphylococcus epidermidis (MRSE-CR).METHODS: Rabbits received a topical instillation of Besivance? (besifloxacin ophthalmic suspension, 0.6%), Vigamox (moxifloxacin hydrochloride ophthalmic solution, 0.5% as base), or Zymar (gatifloxacin ophthalmic solution, 0.3%), and ocular tissues and plasma were collected from 4 animals/treatment/collection time at 8 predetermined time intervals during the 24h after dosing. Ocular levels of each agent were measured by LC/MS/MS, and PK parameters (Cmax, Tmax, and AUC????) were determined. AUC????/MIC?? ratios were calculated for tears, conjunctiva, cornea, and aqueous humor using previously reported MIC??values for MRSA-CR and MRSE-CR.RESULTS: All of the fluoroquinolones tested demonstrated rapid penetration into ocular tissues after a single instillation. Besifloxacin demonstrated the highest exposure in tear fluid, while exposure in conjunctiva was comparable for all 3 compounds. Peak concentrations of all fluoroquinolones in aqueous humor were at or below ~1g/mL. In comparison with their MIC??values against MRSE-CR and MRSA-CR, besifloxacin achieved an AUC????/MIC?? ratio of ~800 in tears, compared with values of ≤10 for moxifloxacin and gatifloxacin. In cornea, conjunctiva, and aqueous humor, the AUC????/MIC?? ratios were <10 for all compounds. However, in these tissues AUC????/MIC?? ratios for besifloxacin were 1.5- to 38-fold higher than moxifloxacin and gatifloxacin….

    [5]. Comstock TL, Paterno MR, Usner DW, Pichichero ME. Efficacy and safety of besifloxacin ophthalmic suspension 0.6% in children and adolescents with bacterial conjunctivitis: a post hoc, subgroup analysis of three randomized, double-masked, parallel-group, multicenter clinical trials. Paediatr Drugs. 2010 Apr 1;12(2):105-12. doi: 10.2165/11534380-000000000-00000.
    Abstract
    BACKGROUND: Acute conjunctivitis is the most frequent eye disorder seen by primary care physicians and one that often affects children. Besifloxacin is a new topical fluoroquinolone, the first chlorofluoroquinolone, for the treatment of bacterial conjunctivitis.OBJECTIVE: To examine the efficacy and safety of besifloxacin ophthalmic suspension 0.6% in patients aged 1-17 years with bacterial conjunctivitis.METHODS: This was a post hoc analysis of a subgroup of pediatric patients aged 1-17 years who had participated in three previously reported, randomized, double-masked, parallel-group, multicenter, clinical trials evaluating the safety and efficacy of besifloxacin in the treatment of bacterial conjunctivitis. The studies were conducted in a community setting (clinical centers). All three clinical trials included children (aged > or = 1 year) with a clinical diagnosis of bacterial conjunctivitis in at least one eye, based on the presence at baseline of grade 1 or greater purulent conjunctival discharge and conjunctival injection, and pin-hole visual acuity of at least 20/200 in both eyes for verbal patients. Two trials were vehicle controlled; the third trial was comparator controlled (moxifloxacin hydrochloride ophthalmic solution 0.5% as base). In all studies, besifloxacin ophthalmic suspension 0.6% was administered as one drop in the affected eye(s) three times daily, at approximately 6-hourly intervals, for 5 days. The main outcome measures were clinical resolution and microbial eradication at visit 2 (day 4 +/- 1 in one study; day 5 +/- 1 in the other two studies) and visit 3 (day 8 or 9). Data from the two vehicle-controlled studies were combined for the assessments to provide greater statistical power.RESULTS: This analysis included 815 pediatric patients aged 1-17 years (447 with culture-confirmed bacterial conjunctivitis). Clinical resolution was significantly greater (p < 0.05) in the besifloxacin group than in the vehicle group at both visit 2 (53.7% vs 41.3%) and visit 3 (88.1% vs 73.0%). Similarly, microbial eradication was significantly higher with besifloxacin than with vehicle at visit 2 (85.8% vs 56.3%) and visit 3 (82.8% vs 68.3%). No significant differences in clinical resolution and microbial eradication were noted between besifloxacin and moxifloxacin. Besifloxacin was well tolerated, with similar incidences of adverse events in the besifloxacin, vehicle, and moxifloxacin groups.CONCLUSION: Besifloxacin ophthalmic suspension 0.6% was shown to be safe and effective for the treatment of bacterial conjunctivitis in children and adolescents aged 1-17 years.

///////Besifloxacin hydrochloride, Besivance, Besifloxacin, SS734, 141388-76-3, 405165-61-9, BOL 303224A, ISV-403, Bausch & Lomb, treatment of non-viral bacterial conjunctivitis

Fc1c(c(Cl)c2c(c1)C(=O)C(\C(=O)O)=C/N2C3CC3)N4CCCC[C@@H](N)C4

Share

Virtue Insight invites you to the 9th Biosimilars Congregation 2016, 22nd Sept 2016, The Lalit Hotel, Mumbai, India

 CONFERENCE  Comments Off on Virtue Insight invites you to the 9th Biosimilars Congregation 2016, 22nd Sept 2016, The Lalit Hotel, Mumbai, India
Jul 132016
 

STR1

9th Biosimilars Congregation 2016 during 22nd September 2016 Mumbai,  INDIA

22nd September 2016, The Lalit Hotel, Mumbai, India

Link http://www.virtueinsight.com/pharma/9th-Biosimilars-Congregation-2016/

ANINDITA DAS

ANINDITA DAS

Dr. Reddy’s Laboratories

MANISH VERMA

MANISH VERMA

Sanofi

DEBOLINA PARTAP

DEBOLINA PARTAP

Wockhardt

PARIKSHIT CHAUDHARI

PARIKSHIT CHAUDHARI

Nestle

HANMANT BARKATE

HANMANT BARKATE

Intas Pharmaceuticals

CONFERENCE INTRODUCTION:-

Virtue Insight invites you to the 9th Biosimilars Congregation 2016 during 22nd September 2016 Mumbai – India. This 9th Biosimilars Congregation 2016 brings together scientists, researchers and CROs from around the world. Biologics are a highly effective class of medicines that are based on naturally occurring proteins and produced using living cells. Current concepts of drugs and biologics, Unique considerations for biologics, Early clinical development essentials, Cancer therapeutics, Comparability for Biologics, Biosimilar approvals are the points of discussion in this session.

9th Biosimilars Congregation 2016 conference will bring together top pharmaceutical, biotechnology and regulatory representatives under one roof that will address the key issues of the industry. Hence, this global event will look at the multiple facets of biosimilars, ranging from the evolving regulatory landscapes, biosimilar guidelines to the legal and economic aspects and current challenges in biosimilar development. This biosimilar conference will focus on multiple aspects of biosimilar product development to successfully deliver safe, biosimilar products to the market place. By attending this biologics conference you will gain a comprehensive outlook on the key issues surrounding biosimilars. This event will provide an important platform for biosimilars stakeholders to discuss and share best practices in expediting development in Biosimilars 2016 and onwards.

 

KEY THEMES DISCUSSED IN THIS CONFERENCE:-

  • Global biopharma policy & market trends
  • The future of biosimilars in India
  • The evolving biosimilar sector: Trends and Implications
  • Complex biological models in biosimilar development
  • Viable extrapolation strategies for biosimilars with special emphasis on TNF antagonists
  • Effectively communicate the value of biosimilar products to raise confidence in the class
  • Improving characterization of biosimilars with technology
  • Create a robust patient services and reimbursement support program for biosimilar products
  • The future of the biosimilar industry – mapping the evolution and development of the biosimilar sector
  • Forecasting the financial growth of biosimilars in the global healthcare marketplace
  • Planning of a biosimilar development project – what to consider from the very beginning
  • Clinical development strategies for biosimilars
  • Gaining Better Market Access: Is Commercial CMO Acceptable for the Biosimilars Market?
  • Regulatory Updates and Development
  • Be part of a major networking opportunity

 

WHO WILL YOU MEET:-

CSOs, CMOs, Vice Presidents, Presidents, Heads, Directors, Team Leaders, and Senior Scientists from the following roles:

  • Biopharmaceuticals/ Biotherapeutics
  • Follow on Biologics/Follow on Proteins
  • Biologics/Biotechnology/ Biogenerics
  • Legal Affairs
  • Intellectual Property
  • Health Economics
  • Pricing and Reimbursement
  • Clinical Immunology
  • Principal Scientist
  • Chief Scientific Officer
  • Process Control and Analytical Technologies
  • Analytical Characterisation
  • Regulatory Compliance
  • Pharmacovigilance
  • Drug Safety & Risk Management
  • Quality Affairs/ Quality Control
  • New Product Development
  • Process Science
  • Portfolio Management
  • Research & Development
  • Business Development
  • Business Operations
  • Scientific Affairs
  • Commercial Affairs

 

WHY SHOULD YOU ATTEND?

Get more from the event, with a broader scope bringing the whole communications value chain together? Enjoy and make the best out of ourdedicated networking drinks time, meet the leading international vendors showcasing the products of tomorrow in the co-located exhibition.Expand your knowledge of the latest business models and strategies in the high-level conference. Whether you are on the branded or generic side, you cannot afford to miss this opportunity to benchmark your tactics and strategies against the industry leaders who will be the first to traverse the pathway. Devise an immediate action plan for your biosimilar prosecution and litigation strategies in light of the barriers to entry, research and development costs, and regulatory hurdles, which are balanced against an enormous potential for increased profit margin

Fen Castro

Fen Castro

Head – Productions

Virtue Insight

Tel (India) –       + 91 44 64614333

Mobile (India) –  + 91 9003 26 0693

Tel (UK) –          + 44 2036120886

Fen Castro fen@virtuefirms.com
Cc: Siddhaarth siddhaarth@virtueinsight.co.in

Link http://www.virtueinsight.com/pharma/9th-Biosimilars-Congregation-2016/

 

//////

Share

Stereoselective synthesis of tricyclic compounds by intramolecular palladium-catalyzed addition of aryl iodides to carbonyl groups

 SYNTHESIS  Comments Off on Stereoselective synthesis of tricyclic compounds by intramolecular palladium-catalyzed addition of aryl iodides to carbonyl groups
Jul 122016
 

1860-5397-12-118.

 

Stereoselective synthesis of tricyclic compounds by intramolecular palladium-catalyzed addition of aryl iodides to carbonyl groups

Freie Universität Berlin, Institut für Chemie und Biochemie, Takustrasse 3, D-14195 Berlin, Germany
Email of corresponding author Corresponding author email     hreissig@chemie.fu-berlin.de
This article is part of the Thematic Series “Organometallic chemistry” and is dedicated to the memory of Professor Peter Hofmann.
Guest Editor: B. F. Straub
Beilstein J. Org. Chem. 2016, 12, 1236–1242.
doi:10.3762/bjoc.12.118

Jakub Saadi, Christoph Bentz, Kai Redies, Dieter Lentz, Reinhold Zimmer, Hans-Ulrich ReissigEmail of corresponding author

Beilstein J. Org. Chem. 2016, 12, 1236–1242. published 16 Jun 2016

Abstract

Starting from γ-ketoesters with an o-iodobenzyl group we studied a palladium-catalyzed cyclization process that stereoselectively led to bi- and tricyclic compounds in moderate to excellent yields. Four X-ray crystal structure analyses unequivocally defined the structure of crucial cyclization products. The relative configuration of the precursor compounds is essentially transferred to that of the products and the formed hydroxy group in the newly generated cyclohexane ring is consistently in trans-arrangement with respect to the methoxycarbonyl group. A transition-state model is proposed to explain the observed stereochemical outcome. This palladium-catalyzed Barbier-type reaction requires a reduction of palladium(II) back to palladium(0) which is apparently achieved by the present triethylamine.

For our systematic studies on samarium diiodide promoted cyclizations leading to benzannulated medium-sized rings [1-4] we required starting materials such as alkenyl-substituted compounds B (Scheme 1). Obvious precursors for B are aryl iodides A that smoothly undergo palladium-catalyzed coupling reactions to provide the desired products. However, in one case [A: R1–R2 = (CH2)4] typical Heck reaction conditions employing styrene as olefin component not only led to the desired styrene derivative B but mainly to the cyclized product C. If the reaction was performed without the olefin it provided only the tertiary alcohol C in reasonable yield [5]. Similar C–C bond forming reactions of aryl halides that involve an insertion of the intermediate aryl palladium species into a carbonyl group are relatively rare (see discussion below). Therefore this serendipitous discovery led us to investigate the reaction in more detail.

[1860-5397-12-118-i1]
Scheme 1: Planned Heck reaction of A to compound B and serendipitous discovery of the palladium-catalyzed cyclization to products C.

Conclusion

We have found new examples of intramolecular palladium-catalyzed nucleophilic additions of aryl iodides to alkyl ketones. These additions proceed in the presence of only 2–5 mol % Pd(PPh3)4 and afford bi- and tricyclic compounds with excellent stereoselectivity and in moderate to very good efficacy. The low mass balance observed in several cases may be due to subsequent reactions such as simple de-iodination of the precursor compounds or elimination of water in the products. However, in general none of these byproducts has been isolated. For compound 2 the bulky isopropyl group slows down the addition to the carbonyl group and an enolate arylation was observed instead as major reaction pathway. Although the scope of the discovered aryl iodide addition to carbonyl groups may be limited it is attractive since only low catalyst loadings are required and interesting products are formed with high stereoselectivity.

STR1

Methyl (2RS,4SR)-4-hydroxy-4-methyl-1,2,3,4-tetrahydronaphthalene-2-carboxylate

Methyl (2RS,4SR)-4-hydroxy-4-methyl-1,2,3,4-tetrahydronaphthalene-2-carboxylate (7): According to the GP1: compound 1 (108 mg, 0.31 mmol), Pd(PPh3)4 (7 mg, 6 µmol), NEt3 (102 mg, 1.01 mmol), DMF (1.5 mL), 110 °C, 3 d. Column chromatography (silica gel, hexanes/ethyl acetate 4:1 to 1:1) provided 24 mg (35%) of 7 as colorless oil.

1H NMR (CDCl3, 700 MHz): δ = 1.66 (s, 3 H, Me), 1.83 (sbr, 1 H, OH), 1.92 (dd, J = 13.7, 12.7 Hz, 1 H, 3-H), 2.33 (ddd, J = 13.7, 2.8, 2.1 Hz, 1 H, 3-H), 2.93-2.96 (m, 1 H, 1-H), 3.04 (ddd, J = 12.7, 4.5, 2.8 Hz, 1 H, 2-H), 3.09 (dddd, J = 15.4, 4.5, 2.1, 0.5 Hz, 1 H, 1-H), 3.74 (s, 3 H, CO2Me), 7.14 (d, J = 7.7 Hz, 1 H, Ar), 7.22 (td, J = 7.4, 1.4 Hz, 1 H, Ar), 7.25 (t, J ≈ 7.4 Hz, 1 H, Ar), 7.55 (dd, J = 7.7, 1.4 Hz, 1 H, Ar) ppm.

13C NMR (CDCl3, 176 MHz): δ = 30.0 (q, Me), 32.7 (t, C-1), 36.3 (d, C-2), 41.6 (t, C-3), 69.7 (s, C-4), 126.0, 127.0, 127.8, 129.2, 134.6, 140.4 (4 d, 2 s, Ar), 51.8, 175.7 (q, s, CO2Me) ppm.

IR (neat): ν̃= 3470 (O-H), 3060, 3020 (=C-H), 2950, 2850 (C-H), 1730 (C=O) cm-1 .

HRMS (ESI-TOF): C13H16O3 Na+ calcd.: 243.0992; found: 243.0984. EA: C13H16O3 (220.3) calcd. (%): C 70.89, H 7.32; found (%): C 70.71, H 7.15.

 

 

/////////1,2-addition,  aryl iodides,  ketones,  nucleophilic addition,  palladium catalysis,

Share

Multicomponent reactions: A simple and efficient route to heterocyclic phosphonates

 SYNTHESIS  Comments Off on Multicomponent reactions: A simple and efficient route to heterocyclic phosphonates
Jul 122016
 

1860-5397-12-121.

Multicomponent reactions: A simple and efficient route to heterocyclic phosphonates

Mohammad Haji

Beilstein J. Org. Chem. 2016, 12, 1269–1301. published 21 Jun 2016

Chemistry Department, Science and Research Branch, Islamic Azad University, Tehran, Iran
Email of corresponding author Corresponding author email     mh_1395@yahoo.com
Associate Editor: T. J. J. Müller
Beilstein J. Org. Chem. 2016, 12, 1269–1301.

doi:10.3762/bjoc.12.121

Abstract

Multicomponent reactions (MCRs) are one of the most important processes for the preparation of highly functionalized organic compounds in modern synthetic chemistry. As shown in this review, they play an important role in organophosphorus chemistry where phosphorus reagents are used as substrates for the synthesis of a wide range of phosphorylated heterocycles. In this article, an overview about multicomponent reactions used for the synthesis of heterocyclic compounds bearing a phosphonate group on the ring is given.

http://www.beilstein-journals.org/bjoc/single/articleFullText.htm?publicId=1860-5397-12-121&vt=f&tpn=0&bpn=home

Conclusion

In this article the use of different multicomponent reactions (MCRs) for the synthesis of heterocyclic phosphonates has been reviewed. This review demonstrates the synthetic potential of multicomponent reactions for the construction of phosphono-substituted heterocyclic rings. The Kabachnik–Fields reaction can be considered the starting point of multicomponent synthesis of this class of compounds. However, the major advancements in this interesting field have been achieved in recent years. More than 75% of the cited literature in this review has been published within the last six years, of which more than three quarters dealt with the synthesis of new heterocyclic phosphonates from non-heterocyclic phosphorus reagents. The remaining works reported the phosphorylation of parent heterocyclic systems. It is worth mentioning, that most of the cited publications focused on the synthesis of five and six-membered rings and only four articles described the synthesis of three and seven-membered heterocycles. Additionally, the majority of the reported syntheses were devoted to the development of new methodologies including the use of advanced catalytic systems, alternative solvents and microwave irradiation. Thus, the development of novel MCR based on phosphorous reagents would allow the synthesis of macrocyclic and medium or large-sized heterocyclic systems, substances which are currently underrepresented in the literature. Further, the design of new biocompatible scaffolds such as β-lactams and peptidomimetics possessing phosphonate groups by MCR-based strategies would significantly extend the synthetic potential of MCRs towards heterocyclic phosphonates

//////////multicomponent reactions,  organophosphorus chemistry,  phosphorus reagents,  phosphorylated heterocycles

Share

OSILODROSTAT for Treatment of Cushing’s Syndrome

 Phase 3 drug  Comments Off on OSILODROSTAT for Treatment of Cushing’s Syndrome
Jul 122016
 

ChemSpider 2D Image | osilodrostat | C13H10FN3

OSILODROSTAT

LCI 699, LCI 699NX

Novartis Ag INNOVATOR

UNII-5YL4IQ1078, CAS 928134-65-0

Benzonitrile, 4-[(5R)-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl]-3-fluoro-
4-[(5R)-6,7-Dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl]-3-fluorobenzonitrile
(R)-4-(6,7-Dihydro-5H-pyrrolo[l,2-c]imidazol-5-yl)-3-fluoro- benzonitrile
  • Molecular FormulaC13H10FN3
  • Average mass227.237 Da
  • Originator Novartis
  • Class Antihypertensives; Fluorobenzenes; Imidazoles; Nitriles; Pyridines; Small molecules
  • Mechanism of Action Aldosterone synthase inhibitors
  • Phase III Cushing syndrome
  • Phase I Liver disorders
  • Discontinued Heart failure; Hypertension; Solid tumours

Most Recent Events

  • 27 Feb 2016 Novartis plans the phase III LINC-4 trial for Cushing’s syndrome in Greece, Thailand, Poland, Turkey, Russia, Brazil, Belgium, Spain, Denmark, Switzerland and USA (PO) (NCT02697734)
  • 12 Jun 2015 Novartis plans a phase II trial for Cushing syndrome in Japan (NCT02468193)
  • 01 Apr 2015 Phase-I clinical trials in Liver disorders in USA (PO)

 

Osilodrostat phosphate
CAS: 1315449-72-9

MF, C13-H10-F-N3.H3-O4-P

MW, 325.2347

  • LCI 699AZA

An orally active aldosterone-synthase inhibitor.

for Treatment of Cushing’s Syndrome

4-((5R)-6,7-Dihydro-5H-pyrrolo(1,2-c)imidazol-5-yl)-3-fluorobenzonitrile dihydrogen phosphate

Aromatase inhibitor; Cytochrome P450 11B1 inhibitor

MORE SYNTHESIS COMING, WATCH THIS SPACE…………………..

 

 

SYNTHESIS

STR1

ACS Medicinal Chemistry Letters, 4(12), 1203-1207; 2013

REMIND ME,  amcrasto@gmail.com, +919323115463

Osilodrostat, as modulators of 11-β-hydroxylase, useful for treating a disorder ameliorated 11-β-hydroxylase inhibition eg Cushing’s disease, hypertension, congestive heart failure, metabolic syndrome, liver diseases, cerebrovascular diseases, migraine headaches, osteoporosis or prostate cancer.

Novartis is developing osilodrostat, an inhibitor of aldosterone synthase and aromatase, for treating Cushing’s disease. In July 2016, osilodrostat was reported to be in phase 3 clinical development.

The somatostatin analog pasireotide and the 11β-hydroxylase inhibitor osilodrostat (LCI699) reduce cortisol levels by distinct mechanisms of action. There exists a scientific rationale to investigate the clinical efficacy of these two agents in combination. This manuscript reports the results of a toxicology study in rats, evaluating different doses of osilodrostat and pasireotide alone and in combination. Sixty male and 60 female rats were randomized into single-sex groups to receive daily doses of pasireotide (0.3mg/kg/day, subcutaneously), osilodrostat (20mg/kg/day, orally), osilodrostat/pasireotide in combination (low dose, 1.5/0.03mg/kg/day; mid-dose, 5/0.1mg/kg/day; or high dose, 20/0.3mg/kg/day), or vehicle for 13weeks. Mean body-weight gains from baseline to Week 13 were significantly lower in the pasireotide-alone and combined-treatment groups compared to controls, and were significantly higher in female rats receiving osilodrostat monotherapy. Osilodrostat and pasireotide monotherapies were associated with significant changes in the histology and mean weights of the pituitary and adrenal glands, liver, and ovary/oviduct. Osilodrostat alone was associated with adrenocortical hypertrophy and hepatocellular hypertrophy. In combination, osilodrostat/pasireotide did not exacerbate any target organ changes and ameliorated the liver and adrenal gland changes observed with monotherapy. Cmax and AUC0-24h of osilodrostat and pasireotide increased in an approximately dose-proportional manner. In conclusion, the pasireotide and osilodrostat combination did not exacerbate changes in target organ weight or toxicity compared with either monotherapy, and had an acceptable safety profile; addition of pasireotide to the osilodrostat regimen may attenuate potential adrenal gland hyperactivation and hepatocellular hypertrophy, which are potential side effects of osilodrostat monotherapy.

The somatostatin analog pasireotide and the 11β-hydroxylase inhibitor osilodrostat (LCI699) reduce cortisol levels by distinct mechanisms of action. There exists a scientific rationale to investigate the clinical efficacy of these two agents in combination. This manuscript reports the results of a toxicology study in rats, evaluating different doses of osilodrostat and pasireotide alone and in combination. Sixty male and 60 female rats were randomized into single-sex groups to receive daily doses of pasireotide (0.3 mg/kg/day, subcutaneously), osilodrostat (20 mg/kg/day, orally), osilodrostat/pasireotide in combination (low dose, 1.5/0.03 mg/kg/day; mid-dose, 5/0.1 mg/kg/day; or high dose, 20/0.3 mg/kg/day), or vehicle for 13 weeks. Mean body-weight gains from baseline to Week 13 were significantly lower in the pasireotide-alone and combined-treatment groups compared to controls, and were significantly higher in female rats receiving osilodrostat monotherapy. Osilodrostat and pasireotide monotherapies were associated with significant changes in the histology and mean weights of the pituitary and adrenal glands, liver, and ovary/oviduct. Osilodrostat alone was associated with adrenocortical hypertrophy and hepatocellular hypertrophy. In combination, osilodrostat/pasireotide did not exacerbate any target organ changes and ameliorated the liver and adrenal gland changes observed with monotherapy. Cmax and AUC0–24h of osilodrostat and pasireotide increased in an approximately dose-proportional manner.

In conclusion, the pasireotide and osilodrostat combination did not exacerbate changes in target organ weight or toxicity compared with either monotherapy, and had an acceptable safety profile; addition of pasireotide to the osilodrostat regimen may attenuate potential adrenal gland hyperactivation and hepatocellular hypertrophy, which are potential side effects of osilodrostat monotherapy.

The somatostatin class is a known class of small peptides comprising the naturally occurring somatostatin- 14 and analogues having somatostatin related activity, e.g. as disclosed by A.S. Dutta in Small Peptides, Vol.19, Elsevier (1993). By “somatostatin analogue” as used herein is meant any straight-chain or cyclic polypeptide having a structure based on that of the naturally occurring somatostatin- 14 wherein one or more amino acid units have been omitted and/or replaced by one or more other amino radical(s) and/or wherein one or more functional groups have been replaced by one or more other functional groups and/or one or more groups have been replaced by one or several other isosteric groups. In general, the term covers all modified derivatives of the native somatostatin- 14 which exhibit a somatostatin related activity, e.g. they bind to at least one of the five somatostatin receptor (SSTR), preferably in the nMolar range. Commonly known somatostatin analogs are octreotide, vapreotide, lanreotide, pasireotide.

Pasireotide, having the chemical structure as follow:

Figure imgf000002_0001

Pasireotide is called cyclo[{4-(NH2-C2H4-NH-CO-0-)Pro}-Phg-DTrp-Lys-Tyr(4-Bzl)- Phe], wherein Phg means -HN-CH(C6H5)-CO- and Bzl means benzyl, in free form, in salt or complex form or in protected form.

Cushing’s syndrome is a hormone disorder caused by high levels of Cortisol in the blood. This can be caused by taking glucocorticoid drugs, or by tumors that produce Cortisol or adrenocorticotropic hormone (ACTH) or CRH. Cushing’s disease refers to one specific cause of the syndrome: a tumor (adenoma) in the pituitary gland that produces large amounts of ACTH, which elevates Cortisol. It is the most common cause of Cushing’s syndrome, responsible for 70% of cases excluding glucocorticoid related cases. The significant decrease of Cortisol levels in Cushing’s disease patients on pasireotide support its potential use as a targeted treatment for Cushing’s disease (Colao et al. N Engl J Med 2012;366:32^12).

Compound A is potent inhibitor of the rate-limiting enzyme 1 1-beta-hydroxylase, the last step in the synthesis of Cortisol. WO 201 1/088188 suggests the potential use of compound A in treating a disease or disorder characterised by increased stress hormone levels and/or decreased androgen hormone levels, including the potential use of compound A in treating heart failure, cachexia, acute coronary syndrome, chronic stress syndrome, Cushing’s syndrome or metabolic syndrome.

Compound A, also called (R)-4-(6,7-Dihydro-5H-pyrrolo[l,2-c]imidazol-5-yl)-3-fluoro- benzonitrile, has formula (II).

Figure imgf000003_0001

Compound A can be synthesized or produced and characterized by methods as described in WO2007/024945.

PRODUCT PATENT

WO2007024945, hold protection in the EU states until August 2026, and expire in the US in March 2029 with US154 extension

PAPER

ACS Medicinal Chemistry Letters (2013), 4(12), 1203-1207.

http://pubs.acs.org/doi/abs/10.1021/ml400324c?source=chemport&journalCode=amclct

Discovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 Inhibitors

Novartis Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
Novartis Pharmaceuticals Corporation, East Hanover, New Jersey 07936, United States
ACS Med. Chem. Lett., 2013, 4 (12), pp 1203–1207
DOI: 10.1021/ml400324c
*(E.L.M.) Tel: 617-871-7586. Fax: 617-871-7045. E-mail: erik.meredith@novartis.com.
Abstract Image

Aldosterone is a key signaling component of the renin-angiotensin-aldosterone system and as such has been shown to contribute to cardiovascular pathology such as hypertension and heart failure. Aldosterone synthase (CYP11B2) is responsible for the final three steps of aldosterone synthesis and thus is a viable therapeutic target. A series of imidazole derived inhibitors, including clinical candidate 7n, have been identified through design and structure–activity relationship studies both in vitro and in vivo. Compound 7n was also found to be a potent inhibitor of 11β-hydroxylase (CYP11B1), which is responsible for cortisol production. Inhibition of CYP11B1 is being evaluated in the clinic for potential treatment of hypercortisol diseases such as Cushing’s syndrome.

PATENT

WO-2016109361

silodrostat (LCI699; 4-[(5R)-6,7-dihydro-5H-pyrrolo[l,2-c]imidazol-5-yl]-3-fluoro-benzonitrile; CAS# 928134-65-0). Osilodrostat is a Ι Ι-β-hydroxylase inhibitor.

Osilodrostat is currently under investigation for the treatment of Cushing’s disease, primary aldosteronism, and hypertension. Osilodrostat has also shown promise in treating drug-resistant hypertension, essential hypertension, hypokalemia, hypertension, congestive heart failure, acute heart failure, heart failure, cachexia, acute coronary syndrome, chronic stress syndrome, Cushing’s syndrome, metabolic syndrome, hypercortisolemia, atrial fibrillation, renal failure, chronic renal failure, restenosis, sleep apnea, atherosclerosis, syndrome X, obesity, nephropathy, post-myocardial infarction, coronary heary disease, increased formation of collagen, cardiac or myocardiac fibrosis and/or remodeling following hypertension and endothelial dysfunction, Conn’s disease, cardiovascular diseases, renal dysfunction, liver diseases, cerebrovascular diseases, vascular diseases, retinopathy, neuropathy, insulinopathy, edema, endothelial dysfunction, baroreceptor dysfunction, migraine headaches, arrythmia, diastolic dysfunction, diastolic heart failure, impaired diastolic filling, systolic dysfunction, ischemia, hypertrophic cardiomyopathy, sudden cardia death, impaired arterial compliance, myocardial necrotic lesions, vascular damage, myocardial infarction, left ventricular hypertrophy, decreased ej ection fraction, cardiac lesions, vascular wall hypertrophy, endothelial thickening, fibrinoid, necrosis of coronary arteries, ectopic ACTH syndrome, change in adrenocortical mass, primary pigmented nodular adrenocortical disease (PPNAD), Carney complex (CNC), anorexia nervosa, chronic alcoholic poisoning, nicotine withdrawal syndrome, cocaine withdrawal syndrome, posttraumatic stress syndrome, cognitive impairment after a stroke or cortisol-induced mineral corticoid excess, ventricular arrythmia, estrogen-dependent disorders, gynecomastia, osteoporosis, prostate cancer, endometriosis, uterine fibroids, dysfunctional uterine bleeding, endometrial hyperplasia, polycyctic ovarian disease, infertility, fibrocystic breast disease, breast cancer, and fibrocystic mastopathy. WO 2013109514; WO 2007024945; and WO 2011064376.

Osilodrostat

Osilodrostat is likely subject to extensive CYP45o-mediated oxidative metabolism. These, as well as other metabolic transformations, occur in part through polymorphically-expressed enzymes, exacerbating interpatient variability. Additionally, some metabolites of osilodrostat derivatives may have undesirable side effects. In order to overcome its short half-life, the drug likely must be taken several times per day, which increases the probability of patient incompliance and discontinuance. Adverse effects associated with osilodrostat include fatigue, nausea, diarrhea, headache, hypokalemia, muscle spasms, vomiting, abdominal discomfort, abdominal pain, arthralgia, arthropod bite, dizziness, increased lipase, and pruritis.

Scheme I

 

EXAMPLE 1

(R)-4-(6,7-dihvdro-5H-pyrrolo[l,2-elimidazol-5-yl)-3-fluorobenzonitrile

(osilodrostat)

[00144] 4-(bromomethyl)-3-fluorobenzonitrile: 3-Fluoro-4-methylbenzonitrile (40 g, 296 mmol), NBS (63.2 g, 356 mmol) and benzoyl peroxide (3.6 g, 14.8 mmol) were taken up in carbon tetrachloride (490 mL) and refiuxed for 16 h. The mixture was allowed to cool to room temperature and filtered. The filtrate was concentrated and purified via flash column chromatography (0-5% EtOAc/hexanes) to give 4-(bromomethyl)-3-fluorobenzonitrile (35.4 g, 56%).

[00145] 2-(l-trityl-lH-imidazol-4-yl)acetic acid: Trityl chloride (40 g, 143.88 mmol, 1.2 equiv) was added to a suspension of (lH-imidazol-4-yl) acetic acid hydrochloride (20 g, 123.02 mmol, 1.0 equiv) in pyridine (200 mL). This was stirred at 50 °C for 16 h. Then the mixture was cooled and concentrated under vacuum and the crude product was purified by recrystallization from ethyl acetate (1000 ml) to afford 42 g (90%) of 2-[l-(triphenylmethyl)-lH-imidazol-4-yl] acetic acid as an off-white solid. LCMS (ESI): m/z = 369.2 [M+H]+

Step 2

2 step 2

2-( 1 -trityl- lH-imidazol-4-yl)ethanol : 2-(l-Trityl-lH-imidazol-4-yl) acetic acid (42 g, 114.00 mmol, 1.0 equiv) was suspended in THF (420 mL) and cooled to 0 °C. To this was added BH3 (1M in THF, 228.28 mL, 2.0 equiv). The clear solution obtained was stirred at 0 °C for 60 min, then warmed to room temperature until LCMS indicated completion of the reaction. The solution was cooled again to 0 °C and quenched carefully with water (300 mL). The resulting solution was extracted with ethyl acetate (3 x 100 mL) and the organic layers combined and dried over anhydrous Na2S04 and evaporated to give a sticky residue which was taken up in ethanolamine (800 mL) and heated to 90 °C for 2 h. The reaction was transferred to a separatory funnel, diluted with EtOAc (1 L) and washed with water (3 x 600 mL). The organic phase was dried over anhydrous Na2S04 and evaporated afford 35 g (87%) of 2-[l-(triphenylmethyl)-lH-imidazol-4-yl]ethanol as a white solid, which was used in the next step without further purification. LCMS (ESI) : m/z = 355.1 [M+H]+.

Step 3

3 step 3 4

4-(2-(tert-butyldimethylsilyloxy)ethyl)-l-trityl-lH-imidazole: 2-(l-Trityl-lH-imidazol-4-yl) ethanol (35 g, 98.75 mmol, 1.00 equiv) was dissolved in DCM (210 mL). To this was added imidazole (19.95 g, 293.05 mmol, 3.00 equiv) and tert-butyldimethylsilylchloride (22.40 g, 149.27 mmol, 1.50 equiv). The mixture was stirred at room temperature until LCMS indicated completion of the reaction. Then the resulting solution was diluted with 500 mL of DCM. The resulting mixture was washed with water (3 x 300 mL). The residue was purified by a silica gel column, eluted with ethyl

acetate/petroleum ether (1 :4) to afford 40 g (77%) of 4-[2-[(tert-butyldimethylsilyl)oxy]ethyl]-l-(triphenylmethyl)-lH-imidazole as a white solid. LCMS (ESI) : m/z = 469.1 [M+H]+.

Step 4

4-((5-(2-(tert-butyldimethylsilyloxy )ethylVlH-iniidazol-l -vnmethylV3-fluorobenzonitrile: 4-(2-((tert-Butyldimethylsilanyl)oxy)ethyl)-l rityl-lH-irnidazole (40 g, 85.34 mmol, 1.00 equiv) and 4-(Bromomethyl)-3-fluorobenzonitrile (27.38 g, 127.92 mmol, 1.50 equiv) obtained as a product of step 0, were dissolved in MeCN (480 mL) and DCM (80 mL), and stirred at room temperature for 48 h. Et2NH (80 mL) and MeOH (480 mL) were then added and the solution was warmed 80 °C for 3 h. The solution was evaporated to dryness and the residue was purified via flash column chromatography (EtOAc/hexanes 1 :5 to EtOAc) to afford 4-((5-(2-((tert-Butyldimethylsilanyl)oxy)ethyl)-lH-imidazol-l -yl)methyl)-3-fluorobenzonitrile (15 g, 50%). ¾ NMR (400 MHz, CDCh) δ: 7.67 (s, 1H), 7.43 (m, 2H), 6.98 (s, 1H), 6.88-6.79 (m, 1H), 5.34 (s, 2H), 3.79 (t, J= 8.0 Hz, 2H), 2.67 (t, J = 8.0 Hz, 2H), 0.88 (s, 9H), 0.02 (s, 6H). LCMS (ESI) : m/z = 360.1 [M+H]+.

Step 5

5 6

Methyl 2-(5-(2-(tert-butyldimethylsilyloxy)ethyl)-lH-imidazol-l -yl)-2-(4-cvano-2-fluorophenvDacetate: 4-((5-(2-((tert-Butyldimethylsilanyl)oxy)ethyl)-lH-imidazol-l -yl)methyl)-3-fluorobenzonitrile (15 g, 41.72 mmol, 1.00 equiv) was dissolved in anhydrous THF (150 mL) and stirred at -78 °C, then a THF solution of LiHMDS (75 mL, 1.80 equiv, 1.0 M) was added dropwise over 15 min. After 30 min, methyl cyanoformate (4.3 g, 45.50 mmol, 1.10 equiv) was added dropwise over 10 min and the solution was stirred at -78 °C for 2 h. The excess LiHMDS was quenched with aqueous saturated NH4CI and the mixture was allowed to warm to room temperature. The mixture was then diluted with EtOAc and washed

with aqueous saturated NH4CI (200 mL). The organic layers was dried over anhydrous Na2S04 and evaporated. The crude residue was purified via flash column chromatography (EtOAc/PE 3: 10 to EtOAc) to give methyl 2-(5-(2-((tert-butyldimethylsilanyl)oxy)ethyl)-lH-imidazol-l-yl)-2-(4-cyano-2-fluorophenyl) acetate (15 g, 86%) as a light yellow solid.

¾ NMR (400 MHz, CDCL3) δ: 7.66 (s, 1H), 7.54-7.43 (m, 2H), 7.15 (t, J= 8.0 Hz 1H), 6.93 (s, 1H), 6.47 (s, 1H), 3.88-3.74 (m, 5H), 2.81-2.62 (m, 2H), 0.89 (s, 9H), 0.05 (s, 6H) . LCMS (ESI) : m/z = 418.2 [M+H]+.

Step 6

Methyl 2-(4-cvano-2-fluorophenyl)-2-(5-(2-hvdroxyethyl)-lH-imidazol-l-yl) acetate: Methyl 2-(5-(2-((tert-butyldimethylsilanyl)oxy)ethyl)-lH-imidazol-l-yl)-2-(4-cyano-2-fiuorophenyl)acetate (15 g, 35.92 mmol, 1.00 equiv) was added to a solution of HCl in 1,4-dioxane (89 mL, 4.0 M, 359.2 mmol) at 0 °C and the mixture was allowed to warm to room temperature and stirred for 2 h. The solution was concentrated to dryness to give the crude alcohol, methyl 2-(4-cyano-2-fluorophenyl )-2-(5-(2 -hydroxy ethyl)-lH-imidazol-l-yl)acetate (10 g, 92%), which was used without further purification. LCMS: m/z = 304.0 [M+H]+.

Step 7

7 8

Methyl 2-(4-cvano-2-fluorophenyl)-2-(5-(2-(methylsulfonyloxy)ethyl)-lH-imidazol-l-yl) acetate: The crude methyl 2-(4-cyano-2-fluorophenyl )-2-(5-(2-hydroxyethyl)-lH-imidazol-l-yl)acetate (10 g, 32.97 mmol, 1.00 equiv) was dissolved in DCM (200 mL) and stirred at 0 °C, then Et3N (20 g, 197.65 mmol, 6.00 equiv) and

methanesulfonyl chloride (4.52 g, 39.67 mmol, 1.20 equiv) were added. After completion of the reaction, the solution was diluted with DCM and washed with aqueous saturated

NaHCC . The organic layer was dried over anhydrous Na2S04, filtered and evaporated to give the crude methyl 2-(4-cyano-2-fluorophenyl)-2-(5-(2-((methylsulfonyl)oxy)ethyl)-lH-imidazol-l-yl)acetate (11.43 g, 91%), which was used in the next step without further purification. LCMS (ESI) : m/z = 382.0 [M+H]+.

Step 8

Methyl 5-(4-cvano-2-fluorophenyl)-6.7-dihvdro-5H-pyrrolo[1.2-elimidazole-5-carboxylate: The crude methyl 2-(4-cyano-2 -fluorophenyl )-2-(5-(2- ((methylsulfonyl)oxy)ethyl)-lH-imidazol-l-yl)acetate (11.43 g, 29.97 mmol, 1.00 equiv) was dissolved in MeCN (550 mL) and then K2CO3 (12.44 g, 90.01 mmol, 3.00 equiv), Nal (13.50 g, 90.00 mmol, 3.00 equiv) and Et3N (9.09 g, 89.83 mmol, 3.00 equiv) were added. The reaction was stirred at 80 °C for 42 h. The mixture was filtered. The solids were washed with DCM. The filtrate was concentrated and purified by flash column chromatography (EtOAc) to give methyl 5-(4-cyano-2-fluorophenyl)-6,7-dihydro-5H-pyrrolo[l,2-c]imidazole-5-carboxylate (4.2 g, 49% in 3 steps).

[00153] ¾ NMR (400 MHz, CDCb) δ: 7.61 (s, 1H), 7.47-7.47 (m, 2H), 6.88 (s, 1H), 6.79-6.75 (m, 1H), 4.17-4.12 (m, 1H), 3.87 (s, 3H), 3.78-3.70 (m, 1H), 3.08-3.02 (m, 1H), 2.84-2.71 (m, 2H). LCMS (ESI) : m/z = 286.0 [M+H]+.

Step 9

10

4-(6.7-dihvdro-5H-pyrrolo[1.2-elimidazol-5-yl)-3-fluorobenzonitrile: To a 40-mL sealed tube, was placed methyl 5-(4-cyano-2-fluorophenyl)-5H,6H,7H-pyrrolo[l,2-c]imidazole-5-carboxylate (1 g, 3.51 mmol, 1.00 equiv), DMSO (10 mL), water (5 mL). The final reaction mixture was irradiated with microwave radiation for 40 min at 140 °C. The resulting solution was diluted with 100 mL of EtOAc. The resulting mixture was washed with (3 x 20 mL) brine, dried over anhydrous Na2S04, filtered and concentrated. The residue was purified by a silica gel column, eluted with ethyl acetate/petroleum ether (4: 1) to afford 420 mg (44%) of 5-(4-cyano-2-fluorophenyl)-5H,6H,7H-pyrrolo[l,2-c]irnidazole-5-carboxylic acid as a light yellow solid.

¾ NMR (400 MHz, CDCL3) δ: 7.55-7.28 (m, 3H), 6.90-6.85 (m, 2H), 5.74-5.71 (m, 1H), 3.25-3.15 (m, 1H), 3.02-2.92 (m, 2H), 2.58-2.50 (m, 1H). LCMS (ESI) : m/z = 228.2 [M+H]+.

Step 10

10

(R)-4-(6 -dihvdro-5H-pyrrolo[1.2-elirnidazol-5-yl)-3-fluorobenzonitrile:

Resolution of the enantiomers of the title compound (300 mg) was performed by chiral HPLC: Column, Chiralpak IA2, 2*25cm, 20um; mobile phase, Phase A: Hex (50%, 0.1% DEA), Phase B: EtOH (50%) ; Detector, UV 254/220 nm to afford the (S)-enantiomer (RT = 17 min) and the (R)-enantiomer (97.6 mg, desired compound) (RT = 21 min).

 ¾ NMR (400 MHz, DMSO-<4) δ: 7.98-7.95 (m, 1H), 7.70-7.69 (m, 1H), 7.50 (s, 1H), 6.87 (t, J= 8.0 Hz, 1H), 6.70 (s, 1H), 5.79-5.76 (m, 1H), 3.15-3.06 (m, 1H), 2.92-2.74 (m, 2H), 2.48-2.43 (m, 1H). LCMS (ESI) : m/z = 228.1 [M+H]+.

 

PATENT

WO2013/153129

https://www.google.com/patents/WO2013153129A1?cl=en

 

PATENT

WO2007/024945

http://www.google.co.in/patents/WO2007024945A1?cl=en

 

PATENT

 EP 2815749

Aspect (iii) of the present invention relates to phosphate salt or nitrate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile according to Formula (III)

Figure imgb0004

abbreviated as ‘{drug3}’. In particular, the present invention relates to crystalline form of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3a}’; to crystalline Form A of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3b}’; to crystalline Form B of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3c}’; to crystalline Form C of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3d}’; to crystalline Form D of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3e}’; to crystalline Form E of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3f}’; to crystalline Form F of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3g}’; to crystalline Form G of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3h}’; to crystalline Form H of phosphate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3i}’; and to crystalline form of nitrate salt of 4-(R)-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl)-3-fluoro-benzonitrile, abbreviated as ‘{drug3j}’. {drug3a}, {drug3b}, {drug3c}, {drug3d}, {drug3e}, {drug3f}, {drug3g}, {drug3h}, {drug3i}, and {drug3j} are specific forms falling within the definition of {drug3}. Aspect (iii) of the invention is separate from aspects (i), (ii), (iv), (v), (vi), (vii), and (viii) of the invention. Thus, all embodiments of {drug3a}, {drug3b}, {drug3c}, {drug3d}, {drug3e}, {drug3f}, {drug3g}, {drug3h}, {drug3i}, and {drug3j}, respectively, are only related to {drug3}, but neither to {drug1}, nor to {drug2}, nor to {drug4}, nor to {drug5}, nor to {drug6}, nor to {drug7}, nor to {drug8}.

 

PAPER

Osilodrostat (LCI699), a potent 11β-hydroxylase inhibitor, administered in combination with the multireceptor-targeted somatostatin analog pasireotide: A 13-week study in rats

  • a Preclinical Safety, Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
  • b Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
  • c Novartis Oncology Development, Basel, Switzerland

doi:10.1016/j.taap.2015.05.004http://www.sciencedirect.com/science/article/pii/S0041008X15001684

CLIPS

STR1

 

STR1

WO2011088188A1 * Jan 13, 2011 Jul 21, 2011 Novartis Ag Use of an adrenal hormone-modifying agent
Reference
1 * BOSCARO M ET AL: “Treatment of Pituitary-Dependent Cushing’s Disease with the Multireceptor Ligand Somatostatin Analog Pasireotide (SOM230): A Multicenter, Phase II Trial“, JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM, vol. 94, no. 1, January 2009 (2009-01), pages 115-122, XP002698507, ISSN: 0021-972X

 

REFERENCES

1: Guelho D, Grossman AB. Emerging drugs for Cushing’s disease. Expert Opin Emerg Drugs. 2015 Sep;20(3):463-78. doi: 10.1517/14728214.2015.1047762. Epub 2015 Jun 2. PubMed PMID: 26021183.

2: Li L, Vashisht K, Boisclair J, Li W, Lin TH, Schmid HA, Kluwe W, Schoenfeld H, Hoffmann P. Osilodrostat (LCI699), a potent 11β-hydroxylase inhibitor, administered in combination with the multireceptor-targeted somatostatin analog pasireotide: A 13-week study in rats. Toxicol Appl Pharmacol. 2015 Aug 1;286(3):224-33. doi: 10.1016/j.taap.2015.05.004. Epub 2015 May 14. PubMed PMID: 25981165.

3: Papillon JP, Adams CM, Hu QY, Lou C, Singh AK, Zhang C, Carvalho J, Rajan S, Amaral A, Beil ME, Fu F, Gangl E, Hu CW, Jeng AY, LaSala D, Liang G, Logman M, Maniara WM, Rigel DF, Smith SA, Ksander GM. Structure-Activity Relationships, Pharmacokinetics, and in Vivo Activity of CYP11B2 and CYP11B1 Inhibitors. J Med Chem. 2015 Jun 11;58(11):4749-70. doi: 10.1021/acs.jmedchem.5b00407. Epub 2015 May 21. PubMed PMID: 25953419.

4: Fleseriu M. Medical treatment of Cushing disease: new targets, new hope. Endocrinol Metab Clin North Am. 2015 Mar;44(1):51-70. doi: 10.1016/j.ecl.2014.10.006. Epub 2014 Nov 4. Review. PubMed PMID: 25732642.

5: Wang HZ, Tian JB, Yang KH. Efficacy and safety of LCI699 for hypertension: a meta-analysis of randomized controlled trials and systematic review. Eur Rev Med Pharmacol Sci. 2015;19(2):296-304. Review. PubMed PMID: 25683946.

6: Daniel E, Newell-Price JD. Therapy of endocrine disease: steroidogenesis enzyme inhibitors in Cushing’s syndrome. Eur J Endocrinol. 2015 Jun;172(6):R263-80. doi: 10.1530/EJE-14-1014. Epub 2015 Jan 30. Review. PubMed PMID: 25637072.

7: Fleseriu M, Petersenn S. Medical therapy for Cushing’s disease: adrenal steroidogenesis inhibitors and glucocorticoid receptor blockers. Pituitary. 2015 Apr;18(2):245-52. doi: 10.1007/s11102-014-0627-0. PubMed PMID: 25560275.

8: Ménard J, Rigel DF, Watson C, Jeng AY, Fu F, Beil M, Liu J, Chen W, Hu CW, Leung-Chu J, LaSala D, Liang G, Rebello S, Zhang Y, Dole WP. Aldosterone synthase inhibition: cardiorenal protection in animal disease models and translation of hormonal effects to human subjects. J Transl Med. 2014 Dec 10;12:340. doi: 10.1186/s12967-014-0340-9. PubMed PMID: 25491597; PubMed Central PMCID: PMC4301837.

9: Oki Y. Medical management of functioning pituitary adenoma: an update. Neurol Med Chir (Tokyo). 2014;54(12):958-65. Epub 2014 Nov 29. PubMed PMID: 25446388.

10: Cai TQ, Stribling S, Tong X, Xu L, Wisniewski T, Fontenot JA, Struthers M, Akinsanya KO. Rhesus monkey model for concurrent analyses of in vivo selectivity, pharmacokinetics and pharmacodynamics of aldosterone synthase inhibitors. J Pharmacol Toxicol Methods. 2015 Jan-Feb;71:137-46. doi: 10.1016/j.vascn.2014.09.011. Epub 2014 Oct 7. PubMed PMID: 25304940.

11: Lother A, Moser M, Bode C, Feldman RD, Hein L. Mineralocorticoids in the heart and vasculature: new insights for old hormones. Annu Rev Pharmacol Toxicol. 2015;55:289-312. doi: 10.1146/annurev-pharmtox-010814-124302. Epub 2014 Sep 10. Review. PubMed PMID: 25251996.

12: Cuevas-Ramos D, Fleseriu M. Treatment of Cushing’s disease: a mechanistic update. J Endocrinol. 2014 Nov;223(2):R19-39. doi: 10.1530/JOE-14-0300. Epub 2014 Aug 18. Review. PubMed PMID: 25134660.

13: Yin L, Hu Q, Emmerich J, Lo MM, Metzger E, Ali A, Hartmann RW. Novel pyridyl- or isoquinolinyl-substituted indolines and indoles as potent and selective aldosterone synthase inhibitors. J Med Chem. 2014 Jun 26;57(12):5179-89. doi: 10.1021/jm500140c. Epub 2014 Jun 5. PubMed PMID: 24899257.

14: Li W, Luo S, Rebello S, Flarakos J, Tse FL. A semi-automated LC-MS/MS method for the determination of LCI699, a steroid 11β-hydroxylase inhibitor, in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2014 Jun 1;960:182-93. doi: 10.1016/j.jchromb.2014.04.012. Epub 2014 Apr 30. PubMed PMID: 24814004.

15: Trainer PJ. Next generation medical therapy for Cushing’s syndrome–can we measure a benefit? J Clin Endocrinol Metab. 2014 Apr;99(4):1157-60. doi: 10.1210/jc.2014-1054. PubMed PMID: 24702012.

16: Bertagna X, Pivonello R, Fleseriu M, Zhang Y, Robinson P, Taylor A, Watson CE, Maldonado M, Hamrahian AH, Boscaro M, Biller BM. LCI699, a potent 11β-hydroxylase inhibitor, normalizes urinary cortisol in patients with Cushing’s disease: results from a multicenter, proof-of-concept study. J Clin Endocrinol Metab. 2014 Apr;99(4):1375-83. doi: 10.1210/jc.2013-2117. Epub 2013 Dec 11. PubMed PMID: 24423285.

17: Oki Y. Medical management of functioning pituitary adenoma: an update. Neurol Med Chir (Tokyo). 2014;54 Suppl 3:958-65. PubMed PMID: 26236804.

18: Schumacher CD, Steele RE, Brunner HR. Aldosterone synthase inhibition for the treatment of hypertension and the derived mechanistic requirements for a new therapeutic strategy. J Hypertens. 2013 Oct;31(10):2085-93. doi: 10.1097/HJH.0b013e328363570c. PubMed PMID: 24107737; PubMed Central PMCID: PMC3771574.

19: Brown NJ. Contribution of aldosterone to cardiovascular and renal inflammation and fibrosis. Nat Rev Nephrol. 2013 Aug;9(8):459-69. doi: 10.1038/nrneph.2013.110. Epub 2013 Jun 18. Review. PubMed PMID: 23774812; PubMed Central PMCID: PMC3922409.

20: van der Pas R, de Herder WW, Hofland LJ, Feelders RA. Recent developments in drug therapy for Cushing’s disease. Drugs. 2013 Jun;73(9):907-18. doi: 10.1007/s40265-013-0067-6. Review. PubMed PMID: 23737437.

///////OSILODROSTAT, Novartis ,  osilodrostat, an inhibitor of aldosterone synthase and aromatase, treating Cushing’s disease,  July 2016, phase 3 clinical development, LCI 699, 928134-65-0, 1315449-72-9, PHASE 3, LCI 699NX, LCI 699AZA, CYP11B1 CYP11B2

c1cc(c(cc1C#N)F)[C@H]2CCc3n2cnc3.OP(=O)(O)O

N#CC1=CC=C([C@H]2CCC3=CN=CN32)C(F)=C1

Share

TEZACAFTOR, VX 661 for treatment of cystic fibrosis disease.

 Phase 3 drug  Comments Off on TEZACAFTOR, VX 661 for treatment of cystic fibrosis disease.
Jul 112016
 

VX-661.png

ChemSpider 2D Image | Tezacaftor | C26H27F3N2O6

img

2D chemical structure of 1152311-62-0

TEZACAFTOR, VX 661

CAS : 1152311-62-0;

  • Molecular FormulaC26H27F3N2O6
  • Average mass520.498 Da

l-(2,2-difluoro-l,3-benzodioxol-5-yl)-N-[l-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(2-hydroxy-l,l-dimethylethyl)-lH-indol-5-yl]-cyclopropanecarboxamide).

(R)-1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide

Cyclopropanecarboxamide, 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-[1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(2-hydroxy-1,1-dimethylethyl)-1H-indol-5-yl]-

1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-[1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)indol-5-yl]cyclopropane-1-carboxamide

Cyclopropanecarboxamide, 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-(1-((2R)-2,3-dihydroxypropyl)-6-fluoro-2-(2-hydroxy-1,1-dimethylethyl)-1H-indol-5-yl)-

1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-(1-((2R)-2,3-dihydroxypropyl)-6-fluoro-2-(2-hydroxy-1,1-dimethylethyl)-1H-indol-5-yl)cyclopropanecarboxamide

Vertex (INNOVATOR)

 

UNII: 8RW88Y506K

In July 2016, this combination was reported to be in phase 3 clinical development.

Tezacaftor, also known asVX-661, is CFTR modulator. VX-661 is potentially useful for treatment of cystic fibrosis disease. Cystic fibrosis (CF) is a genetic disease caused by defects in the CF transmembrane regulator (CFTR) gene, which encodes an epithelial chloride channel. The most common mutation, Δ508CFTR, produces a protein that is misfolded and does not reach the cell membrane. VX-661 can correct trafficking of Δ508CFTR and partially restore chloride channel activity. VX-661 is currently under Phase III clinical trial.

VX-661 is an orally available deltaF508-CFTR corrector in phase III clinical trials at Vertex for the treatment of cystic fibrosis in patients homozygous to the F508del-CFTR mutation

Novel deuterated analogs of a cyclopropanecarboxamide ie tezacaftor (VX-661), as modulators of cystic fibrosis transmembrane conductance regulator (CFTR) proteins, useful for treating a CFTR-mediated disorder eg cystic fibrosis.

VX-661 (CAS #: 1152311-62-0; l-(2,2-difluoro-l,3-benzodioxol-5-yl)-N-[l-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(2-hydroxy-l,l-dimethylethyl)-lH-indol-5-yl]-cyclopropanecarboxamide). VX-661 is a cystic fibrosis transmembrane conductance regulator modulator. VX-661 is currently under investigation for the treatment of cystic fibrosis. VX-661 has also shown promise in treating sarcoglycanopathies, Brody’s disease, cathecolaminergic polymorphic ventricular tachycardia, limb girdle muscular dystrophy, asthma, smoke induced chronic obstructive pulmonary disorder, chronic bronchitis, rhinosinusitis, constipation, pancreatitis, pancreatic insufficiency, male infertility caused by congenital bilateral absence of the vas deferens (CBAVD), mild pulmonary disease, idiopathic pancreatitis, allergic bronchopulmonary aspergillosis (ABPA), liver disease, hereditary emphysema, hereditary hemochromatosis, coagulation-fibrinolysis deficiencies, such as protein C deficiency, type 1 hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, type 1 chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, mucopolysaccharidoses, Sandhof/Tay-Sachs, Crigler-Najjar type II, polyendocrinopathy/hyperinsulinemia, diabetes mellitus, Laron dwarfism, myeloperoxidase deficiency, primary hypoparathyroidism, melanoma, glycanosis CDG type 1, congenital hyperthyroidism, osteogenesis imperfecta, hereditary hypofibrinogenemia, ACT deficiency, diabetes insipidus (DI), neurohypophyseal DI, nephrogenic DI, Charcot-Marie tooth syndrome, Pelizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, progressive supranuclear palsy, Pick’s disease, polyglutamine neurological disorders such as Huntington’s, spinocerebellar ataxia type I, spinal and bulbar muscular atrophy, dentatombral pallidoluysian, and myotonic dystrophy, as well as spongifiorm encephalopathies, such as hereditary Creutzfeldt- Jakob disease (due to prion protein processing defect), Fabry disease, Gerstrnarm-Straussler-Scheinker syndrome, chronic obstructive pulmonary disorder, dry-eye disease, or Sjogren’s disease, osteoporosis, osteopenia, bone healing and bone growth (including bone repair, bone regeneration, reducing bone resorption and increasing bone deposition), Gorham’s Syndrome, chloride channelopathies such as myotonia congenita (Thomson and Becker forms), Bartter’s

syndrome type III, Dent’s disease, hyperekplexia, epilepsy, lysosomal storage disease, Angelman syndrome, and primary ciliary dyskinesia (PCD), a term for inherited disorders of the structure and/or function of cilia, including PCD with situs inversus (also known as Kartagener syndrome), PCD without situs inversus, and ciliary aplasia. WO 2014086687; WO2013185112.

VX-661

VX-661 is likely subject to extensive CYP45o-mediated oxidative metabolism. These, as well as other metabolic transformations, occur in part through polymorphically-expressed enzymes, exacerbating interpatient variability. Additionally, some metabolites of VX-661 may have undesirable side effects. In order to overcome its short half-life, the drug likely must be taken several times per day, which increases the probability of patient incompliance and discontinuance.Deuterium Kinetic Isotope Effect

PATENT

WO 2016109362

 

Scheme I

EXAMPLE 1

(R)-l-(2,2-difluorobenzo[dl[l,31dioxol-5-vn-N-(l-q,3-dihvdroxypropyn-6-fluoro-2-(l- hvdroxy-2-methylpropan-2-yl)-lH-indol-5-yl)cvclopropanecarboxamide

(VX-661)

Methyl 2.2-difluorobenzo[dl [1.31dioxole-5-carboxylate: To a 200 mL pressure tank reactor (10 atm. in CO), was placed 5-bromo-2,2-difluoro-2H-l,3-benzodioxole (20.0 g, 84.4 mmol, 1.00 equiv), methanol (40 mL), triethylamine (42.6 g, 5.00 equiv.), Pd2(dba)3 (1.74 g, 1.69 mmol, 0.02 equiv), Pd(dppf)Cl2 (1.4 g, 1.69 mmol, 0.02 equiv.). The resulting solution was stirred at 85 °C under an atmosphere of CO overnight and the reaction progress was monitored by GCMS. The reaction mixture was cooled. The solids were filtered out. The organic phase was concentrated under vacuum to afford 17.5 g of methyl 2,2-difluoro-2H-l,3-benzodioxole-5-carboxylate as a crude solid, which was used directly in the next step. Step 2

2 step 2 3

(2.2-difluorobenzo[dl [ 1.31 dioxol-5 -vDmethanol : To a 500mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen were placed methyl 2,2-difluoro-2H-l,3-benzodioxole-5-carboxylate (17.5 g, 81.01 mmol, 1.00 equiv.), tetrahydrofuran (200 mL). This was followed by the addition of L1AIH4 (6.81 mg, 162.02 mmol, 2.00 equiv.) at 0 °C. The resulting solution was stirred for 1 h at 25 °C and monitored by GCMS. The reaction mixture was cooled to 0 °C until GCMS indicated the completion of the reaction. The pH value of the solution was adjusted to 8 with sodium hydroxide (1 mol/L). The solids were filtered out. The organic layer combined and concentrated under vacuum to afford 13.25 g (87%) of (2,2-difluoro-2H-l,3-benzodioxol-5-yl)methanol as yellow oil.

Step 3

step 3

5-(chloromethyl)-2.2-difluorobenzo[diri.31dioxole: (2.2-difluoro-2H-1.3-benzodioxol-5-yl)methanol (13.25 g, 70.4 mmol, 1.00 equiv.) was dissolved in DCM (200 mL). Thionyl chloride (10.02 g, 1.20 equiv.) was added to this solution. The resulting mixture was stirred at room temperature for 4 hours and then concentrated under vacuum. The residue was then diluted with DCM (500 mL) and washed with 2 x 200 mL of sodium bicarbonate and 1 x 200 mL of brine. The mixture was dried over anhydrous sodium sulfate, filtered and evaporated to afford 12.36 g (85%) of 5-(chloromethyl)-2,2-difluoro-2H-l ,3-benzodioxole as yellow oil.

Step 4

step 4 5

[00160] 2-(2.2-difluorobenzordi ri .31dioxol-5-yl)acetonitrile: 5-(chloromethyl)-2,2-difluoro-2H-l,3-benzodioxole (12.36 g, 60 mmol, 1.00 equiv.) was dissolved in DMSO (120 mL). This was followed by the addition of NaCN (4.41 g, 1.50 equiv.) with the inert temperature below 40 °C. The resulting solution was stirred for 2 hours at room temperature. The reaction progress was monitored by GCMS. The reaction was then quenched by the addition of 300 mL of water/ice. The resulting solution was extracted with 3 x 100 mL of ethyl acetate. The organic layers combined and washed with 3 x 100 mL brine dried over anhydrous sodium sulfate and concentrated under vacuum to afford 10.84 g (92%) of 2-(2,2-difluoro-2H-l ,3-benzodioxol-5-yl)acetonitrile as brown oil.

Step 5

l -(2.2-difluoro-2H-1.3-benzodioxol-5-yl)cvclopropane-l -carbonitrile: To a 100 mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, were placed 2-(2,2-difluoro-2H-l ,3-benzodioxol-5-yl)acetonitrile (10.84 g, 55 mmol, 1.00 equiv.),

NaOH (50%) in water), 1 -bromo-2-chloroethane (11.92g, 82.5 mmol, 1.50 equiv.), BmNBr

(361 mg, 1.1 mmol, 0.02 equiv.). The resulting solution was stirred for 48 h at 70 °C. The reaction progress was monitored by GCMS. The reaction mixture was cooled. The resulting solution was extracted with 3 x 200 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 1 x 200 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum to afford 10.12g of 1 -(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carbonitrile as brown oil.

Step 6

[00162] l-(2.2-difluoro-2H-1.3-benzodioxol-5-yl)cvclopropane-l-carboxylic acid: To a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carbonitrile (10.12 g, 45.38 mmol, 1.00 equiv), 6 N NaOH (61 mL) and EtOH (60 mL). The resulting solution was stirred for 3 h at 100 °C. The reaction mixture was cooled and the pH value of the solution was adjusted to 2 with hydrogen chloride (1 mol/L) until LCMS indicated the completion of the reaction. The solids were collected by filtration to afford 9.68 g (88%) of l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carboxylic acid as a light yellow solid.

Step 7

[00163] l-(2.2-difluoro-2H-1.3-benzodioxol-5-yl)cvclopropane-l-carbonyl chloride; To a solution of l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carboxylic acid (687 mg, 2.84 mmol, 1.00 equiv.) in toluene (5 mL) was added thionyl chloride (1.67 g, 5.00 equiv.). The resulting solution was stirred for 3h at 65 °C. The reaction mixture was cooled and concentrated under vacuum to afford 738 mg (99%) of l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carbonyl chloride as a yellow solid.

Step 8

9 STEP 8 10

2-methyl-4-(trimethylsilyl)but-3-vn-2-ol: To a solution of ethynyltrimethylsilane (20 g, 203.63 mmol, 1.00 equiv) in THF (100 mL) was added n-BuLi (81 mL, 2.5M in THF)

dropwise with stirring at -78 °C. Then the resulting mixture was warmed to 0 °C for 1 h with stirring and then cooled to -78 °C. Propan-2-one (11.6 g, 199.73 mmol, 1.00 equiv.) was added dropwise with the inert temperature below -78 °C. The resulting solution was stirred at -78 °C for 3 h. The reaction was then quenched by the addition of 100 mL of water and extracted with 3 x 100 mL of MTBE. The combined organic layers was dried over anhydrous sodium sulfate and concentrated under vacuum to afford 28 g (90%) of 2-methyl-4-(trimethylsilyl)but-3-yn-2-ol as an off-white solid. ¾ NMR (400 MHz, CDCh) δ: 1.50 (s, 6H), 1.16-1.14 (m, 9H).

Step 9

step 9

10

(3-chloro-3-methylbut-l-vnvntrimethylsilane: To a lOOmL round-bottom flask, was placed 2-methyl-4-(trimethylsilyl) but-3-yn-2-ol (14 g, 89.57 mmol, 1.00 equiv.), cone. HC1 (60 mL, 6.00 equiv.). The resulting solution was stirred for 16 h at 0 °C. The resulting solution was extracted with 3 x 100 mL of hexane. The combined organic layers was dried over anhydrous sodium sulfate and concentrated under vacuum to afford 8 g (51%) of (3-chloro-3-methylbut-l-yn-l-yl)trimethylsilane as light yellow oil. ¾ NMR (400 MHz, CDCh) δ: 1.84 (s, 6H), 1.18-1.16 (m, 9H).

Step 10

step 10

11 12

(4-(benzyloxy)-3.3-dimethylbut-l-vnyl)trimethylsilane: Magnesium turnings (1.32 g, 1.20 equiv) were charged to a 250-mL 3-necked round-bottom flask and then suspended in THF (50 mL). The resulting mixture was cooled to 0 °C and maintained with an inert atmosphere of nitrogen. (3-chloro-3-methylbut-l-yn-l-yl)trimethylsilane (8 g, 45.78 mmol, 1.00 equiv.) was dissolved in THF (50 mL) and then added dropwise to this mixture with the inert temperature between 33-37 °C. The resulting solution was stirred at room temperature for an addition 1 h before BnOCH2Cl (6.45 g, 41.33 mmol, 0.90 equiv.) was added dropwise with the temperature below 10 °C. Then the resulting solution was stirred for 16 h at room temperature. The reaction was then quenched by the addition of 50 mL of water and extracted with 3 x 100 mL of hexane. The combined organic layers was dried over

anhydrous sodium sulfate and concentrated under vacuum to afford 10 g (84%) of [4-(benzyloxy)-3,3-dimethylbut-l-yn-l-yl]trimethylsilane as light yellow oil. ¾ NMR (400 MHz, CDCh) δ: 7.37-7.35 (m, 5H), 4.62 (s, 2H), 3.34 (s, 2H), 1.24 (s, 6H), 0.17-0.14 (m, 9H).

Step 11

((2.2-dimethylbut-3-vnyloxy)methyl)benzene: To a solution of [4-(benzyloxy)-3,3-dimethylbut-l-yn-l-yl]trimethylsilane (10 g, 38.40 mmol, 1.00 equiv) in methanol (100 mL) was added potassium hydroxide (2.53 g, 38.33 mmol, 1.30 equiv). The resulting solution was stirred for 16 h at room temperature. The resulting solution was diluted with 200 mL of water and extracted with 3 x 100 mL of hexane. The organic layers combined and washed with 1 x 100 mL of water and then dried over anhydrous sodium sulfate and concentrated under vacuum to afford 5 g (69%) of [[(2,2-dimethylbut-3-yn-l-yl)oxy]methyl]benzene as light yellow oil. ¾ NMR (300 MHz, D20) δ: 7.41-7.28 (m, 5H) , 4.62 (s, 2H), 3.34 (s, 2H), 2.14 (s, 1H), 1.32-1.23 (m, 9H).

Step 12

14 15

methyl 2.2-difluorobenzo[d1[1.31dioxole-5-carboxylate: To a solution of 3-fluoro-4-nitroaniline (6.5 g, 41.64 mmol, 1.00 equiv) in chloroform (25 mL) and AcOH (80 mL) was added Bn (6.58 g, 41.17 mmol, 1.00 equiv.) dropwise with stirring at 0 °C in 20 min. The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 150 mL of water/ice. The pH value of the solution was adjusted to 9 with sodium hydroxide (10 %). The resulting solution was extracted with 3 x 50 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 1 x 50 mL of water and 2 x 50 mL of brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was re-crystallized from PE/EA (10: 1) to afford 6 g (61%) of 2-bromo-5-fluoro-4-nitroaniline as a yellow solid.

Step 13

(R)-l-(benzyloxy)-3-(2-bromo-5-fluoro-4-nitrophenylamino)propan-2-ol: 2-bromo-5-fluoro-4-nitroaniline (6.00 g, 25.56 mmol, 1.00 equiv.), Zn(C104)2 (1.90 g, 5.1 mmol, 0.20 equiv.), 4A Molecular Sieves (3 g), toluene (60 mL) was stirred at room temperature for 2 h and maintain with an inert atmosphere of N2 until (2R)-2-[(benzyloxy)methyl]oxirane (1.37 g, 8.34 mmol, 2.00 equiv.) was added. Then the resulting mixture was stirred for 15 h at 85 °C. The reaction progress was monitored by LCMS. The solids were filtered out and the resulting solution was diluted with 20 mL of ethyl acetate. The resulting mixture was washed with 2 x 20 mL of Sat. NH4CI and 1 x 20 mL of brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by a silica gel column, eluted with ethyl acetate/petroleum ether (1 :5) to afford 7.5 g (70%) of N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-bromo-5-fluoro-4-nitroaniline as a yellow solid.

Step 14

[00170] (R)-l-(4-amino-2-bromo-5-fluorophenylamino)-3-(benzyloxy)propan-2-ol: To a 250-mL round-bottom flask, was placed N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-bromo-5-fluoro-4-nitroaniline (7.5 g, 18.84 mmol, 1.00 equiv.), ethanol (80 mL), water (16 mL), NH4CI (10 g, 189 mmol, 10.00 equiv.), Zn (6.11 g, 18.84 mmol, 5.00 equiv.). The resulting solution was stirred for 4 h at 85 °C. The solids were filtered out and the resulting solution was concentrated under vacuum and diluted with 200 mL of ethyl acetate. The resulting mixture was washed with 1 x 50 mL of water and 2 x 50 mL of brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by a silica gel column, eluted with ethyl acetate/petroleum ether (1 :3) to afford 4.16 g (60%) of l-N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-bromo-5-fluorobenzene-l ,4-diamine as light yellow oil.

Step 15

TsO

(R)-4-(3-(benzyloxy)-2-hvdroxypropylamino)-5-bromo-2-fluorobenzenaminium 4-methylbenzenesulfonate: l-N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-bromo-5-fluorobenzene-l ,4-diamine (2 g, 5.42 mmol, 1.00 equiv.) was dissolved in dichloromethane (40 mL) followed by the addition of TsOH (1 g, 5.81 mmol, 1.10 equiv.). The resulting mixture was stirred for 16 h at room temperature and then concentrated under vacuum to afford 2.8 g (95%) of 4-[[(2R)-3-(benzyloxy)-2-hydroxypropyl]amino]-5-bromo-2-fluoroanilinium 4-methylbenzene-l -sulfonate as an off-white solid.

Step 16

(R)-l-(4-amino-2-(4-(benzyloxy)-3.3-dimethylbut-l-vnyl)-5-fluorophenylamino)-3-(benzyloxy)propan-2-ol: To a 100-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 4-[[(2R)-3-(benzyloxy)-2-hydroxypropyl]amino]-5-bromo-2-fluoroanilinium 4-methylbenzene-l -sulfonate (2.9 g, 5.36 mmol, 1.00 equiv.), [[(2,2-dimethylbut-3-yn-l-yl)oxy]methyl]benzene (1.2 g, 6.37 mmol, 1.20 equiv.), Pd(OAc)2 (48 mg, 0.21 mmol, 0.04 equiv.), dppb (138 mg, 0.32 mmol, 0.06 equiv.), potassium carbonate (2.2 g, 15.92 mmol, 3.00 equiv.) and MeCN (50 mL). The resulting solution was stirred for 16 h at 80 °C. The solids were filtered out and the resulting mixture was concentrated under vacuum until LCMS indicated the completion of the reaction. The residue was purified by a silica gel column, eluted with ethyl acetate/petroleum ether (1 :4) to afford 2.2 g (86%) of l-N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-[4-(benzyloxy)-3,3-dimethylbut-l-yn-l-yl]-5-fluorobenzene-l ,4-diamine as a light brown solid.

Step 17

l-(2.2-difluoro-2H-1.3-benzodioxol-5-yl)cvclopropane-l-carboxylic acid: To a 40-mL vial purged and maintained with an inert atmosphere of nitrogen, was placed 1-N-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-[4-(benzyloxy)-3,3-dimethylbut-l-yn-l-yl]-5-fluorobenzene-l,4-diamine (1 g, 2.1 mmol, 1.00 equiv.), MeCN (10 mL), Pd(MeCN)2Cl2 (82 mg, 0.32 mmol, 0.15 equiv.). The resulting solution was stirred for 12 h at 85 °C. The reaction progress was monitored by LCMS. The resulting mixture was concentrated under vacuum to afford 900 mg (crude) of (2R)-l-[5-amino-2-[l-(benzyloxy)-2-methylpropan-2-yl]-6-fluoro-lH-indol-l-yl]-3-(benzyloxy)propan-2-ol as a brown solid, which was used for next step without further purification.

Step 18

(R)-N-(l-(3-(benzyloxy)-2-hvdroxypropyl)-2-(l-(benzyloxy)-2-methylpropan-2-yl)-6-fluoro- lH-indol-5-yl)- 1 -(2.2-difluorobenzo[dl [ 1.31 dioxol-5-vDcvclopropanecarboxamide: To a 40 mL vial purged and maintained with an inert atmosphere of nitrogen, was placed (2R)-l-[5-amino-2-[l-(benzyloxy)-2-methylpropan-2-yl]-6-fluoro-lH-indol-l-yl]-3-(benzyloxy)propan-2-ol (800 mg, 1.68 mmol, 1.00 equiv.), dichloromethane (20 mL), TEA (508 mg, 5.04 mmol, 3.00 equiv.). l-(2,2-difiuoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carbonyl chloride (524 mg, 2 mmol, 1.20 equiv.) was added to this mixture at 0 °C. The resulting solution was stirred for 2 h at 25 °C. The reaction progress was monitored by LCMS. The resulting solution was diluted with 20 mL of DCM and washed with 3 xlO mL of brine. The combined organic layers was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by a silica gel column, eluted with ethyl acetate/petroleum ether (1:5) to afford 400 mg (30%) of N-[l-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-[l-(benzyloxy)-2-methylpropan-2-yl]-6-fluoro-lH-indol-5-yl]-l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carboxamide as a light yellow solid.

Step 19

(R)-l-(2,2-difluorobenzo[d] [l,3]dioxol-5-yl)-N-(l-(2,3-dihydroxypropyl)-6-fluoro-2-(l-hydroxy-2-methylpropan-2-yl)-lH-indol-5-yl)cyclopropanecarboxamide: To a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of H2, were placed N-[l-[(2R)-3-(benzyloxy)-2-hydroxypropyl]-2-[l-(benzyloxy)-2-methylpropan-2-yl]-6-fluoro-lH-indol-5-yl]-l-(2,2-difluoro-2H-l,3-benzodioxol-5-yl)cyclopropane-l-carboxamide (400 mg, 0.77 mmol, 1.00 equiv.) dry Pd/C (300 mg) and MeOH (5 Ml, 6M HC1). The resulting mixture was stirred at room temperature for 2 h until LCMS indicated the completion of the reaction. The solids were filtered out and the resulting mixture was concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD Column 19 x 150 mm, 5um; mobile phase and Gradient, Phase A: Waters (0.1%FA ), Phase B: ACN; Detector, UV 254 nm to afford 126.1 mg (42.4%) of (R)-l-(2,2-difluorobenzo[d] [l,3]dioxol-5-yl)-N-(l-(2,3-dihydroxypropyl)-6-fluoro-2-(l-hydroxy-2-methylpropan-2-yl)-lH-indol-5-yl)cyclopropanecarboxamide as a light yellow solid.

¾ NMR (400 MHz, OMSO-de) δ: 8.32 (s, 1H), 7.54 (s, 1H), 7.41-7.38 (m, 2H), 7.34-7.31 (m, 2H), 6.22 (s, 1H), 5.03-5.02 (m, 1H), 4.93-4.90 (m, 1H), 4.77-4.75 (m, 1H), 4.42-4.39 (m, 1H), 4.14-4.08 (m, 1H), 3.91 (brs, 1H) , 3.64-3.57 (m, 2H), 3.47-3.40 (m, 2H), 1.48-1.46 (m, 2H), 1.36-1.32 (m, 6H), 1.14-1.12 (m, 2H).

LCMS: m/z = 521.2[M+H]+.

PATENT

WO 2015160787

https://www.google.com/patents/WO2015160787A1?cl=en

PATENT

WO 2014014841

https://www.google.com/patents/WO2014014841A1?cl=en

All tautomeric forms of the Compound 1 are included herein. For example, Compound 1 may exist as tautomers, both of which are included herein:

Figure imgf000026_0001

Methods of Preparing Compound 1 Amorphous Form and Compound 1 Form A

Compound 1 is the starting point and in one embodiment can be prepared by coupling an acid chloride moiety with an amine moiety according to Schemes 1-4.

Scheme 1. Synthesis of the acid chloride moiety.

Figure imgf000037_0001

Toluene, H20, 70 °C

Figure imgf000037_0002

Bu4NBr

1. NaOH

2. HC1

Figure imgf000037_0003

Scheme 2. Synthesis of acid chloride moiety – alternative synthesis.

Figure imgf000038_0001

1. NaCN

2. H20

Figure imgf000038_0002

SOC1,

Figure imgf000038_0003

Scheme 3. Synthesis of the amine moiety.

Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000039_0003

Scheme 4. Formation of Compound 1.

Figure imgf000040_0001

Compound 1

Methods of Preparing Compound 1 Amorphous Form

Starting from Compound 1 , or even a crystalline form of Compound 1 , Compound 1 Amorphous Form may be prepared by rotary evaporation or by spray dry methods.

Dissolving Compound 1 in an appropriate solvent like methanol and rotary evaporating the methanol to leave a foam produces Compound 1 Amorphous Form. In some embodiments, a warm water bath is used to expedite the evaporation.

Compound 1 Amorphous Form may also be prepared from Compound 1 using spray dry methods. Spray drying is a process that converts a liquid feed to a dried particulate form. Optionally, a secondary drying process such as fluidized bed drying or vacuum drying, may be used to reduce residual solvents to pharmaceutically acceptable levels. Typically, spray drying involves contacting a highly dispersed liquid suspension or solution, and a sufficient volume of hot air to produce evaporation and drying of the liquid droplets. The preparation to be spray dried can be any solution, coarse suspension, slurry, colloidal dispersion, or paste that may be atomized using the selected spray drying apparatus. In a standard procedure, the preparation is sprayed into a current of warm filtered air that evaporates the solvent and conveys the dried product to a collector (e.g. a cyclone). The spent air is then exhausted with the solvent, or alternatively the spent air is sent to a condenser to capture and potentially recycle the solvent. Commercially available types of apparatus may be used to conduct the spray drying. For example, commercial spray dryers are manufactured by Buchi Ltd. And Niro (e.g., the PSD line of spray driers manufactured by Niro) (see, US 2004/0105820; US 2003/0144257).

Spray drying typically employs solid loads of material from about 3% to about 30% by weight, (i.e., drug and excipients), for example about 4% to about 20% by weight, preferably at least about 10%. In general, the upper limit of solid loads is governed by the viscosity of (e.g., the ability to pump) the resulting solution and the solubility of the components in the solution. Generally, the viscosity of the solution can determine the size of the particle in the resulting powder product.

Techniques and methods for spray drying may be found in Perry’s Chemical

Engineering Handbook, 6th Ed., R. H. Perry, D. W. Green & J. O. Maloney, eds.), McGraw-Hill book co. (1984); and Marshall “Atomization and Spray-Drying” 50, Chem. Eng. Prog. Monogr. Series 2 (1954). In general, the spray drying is conducted with an inlet temperature of from about 60 °C to about 200 °C, for example, from about 95 °C to about 185 °C, from about 110 °C to about 182 °C, from about 96 °C to about 180 °C, e.g., about 145 °C. The spray drying is generally conducted with an outlet temperature of from about 30 °C to about 90 °C, for example from about 40 °C to about 80 °C, about 45 °C to about 80 °C e.g., about 75 °C. The atomization flow rate is generally from about 4 kg h to about 12 kg/h, for example, from about 4.3 kg/h to about 10.5 kg h, e.g., about 6 kg/h or about 10.5 kg/h. The feed flow rate is generally from about 3 kg/h to about 10 kg/h, for example, from about 3.5 kg/h to about 9.0 kg/h, e.g., about 8 kg/h or about 7.1 kg/h. The atomization ratio is generally from about 0.3 to 1.7, e.g., from about 0.5 to 1.5, e.g., about 0.8 or about 1.5.

Removal of the solvent may require a subsequent drying step, such as tray drying, fluid bed drying (e.g., from about room temperature to about 100 °C), vacuum drying, microwave drying, rotary drum drying or biconical vacuum drying (e.g., from about room temperature to about 200 °C).

Synthesis of Compound 1

Acid Chloride Moiety

Synthesis of (2,2-difluoro-l,3-benzodioxol-5-yl)-l-ethylacetate-acetonitrile

Figure imgf000083_0001

ouene, 2 , CN

A reactor was purged with nitrogen and charged with 900 mL of toluene. The solvent was degassed via nitrogen sparge for no less than 16 h. To the reactor was then charged Na3P04 (155.7 g, 949.5 mmol), followed by bis(dibenzylideneacetone) palladium (0) (7.28 g, 12.66 mmol). A 10% w/w solution of tert-butylphosphine in hexanes (51.23 g, 25.32 mmol) was charged over 10 min at 23 °C from a nitrogen purged addition funnel. The mixture was allowed to stir for 50 min, at which time 5-bromo-2,2-difluoro-l,3-benzodioxole (75 g, 316.5 mmol) was added over 1 min. After stirring for an additional 50 min, the mixture was charged with ethyl cyanoacetate (71.6 g, 633.0 mmol) over 5 min followed by water (4.5 mL) in one portion. The mixture was heated to 70 °C over 40 min and analyzed by HPLC every 1 – 2 h for the percent conversion of the reactant to the product. After complete conversion was observed (typically 100% conversion after 5 – 8 h), the mixture was cooled to 20 – 25 °C and filtered through a celite pad. The celite pad was rinsed with toluene (2 X 450 mL) and the combined organics were concentrated to 300 mL under vacuum at 60 – 65 °C. The concentrate was charged with 225mL DMSO and concentrated under vacuum at 70 – 80 °C until active distillation of the solvent ceased. The solution was cooled to 20 – 25 °C and diluted to 900 mL with DMSO in preparation for Step 2. Ή NMR (500 MHz, CDC13) δ 7.16 – 7.10 (m, 2H), 7.03 (d, J = 8.2 Hz, 1H), 4.63 (s, 1H), 4.19 (m, 2H), 1.23 (t, J= 7.1 Hz, 3H).

Synthesis of (2,2-difluoro-l^-benzodioxol-5-yl)-acetonitrile.

Figure imgf000084_0001

[00311] The DMSO solution of (2,2-difluoro-l,3-benzodioxol-5-yl)-l-ethylacetate-acetonitrile from above was charged with 3 N HCl (617.3 mL, 1.85 mol) over 20 min while maintaining an internal temperature < 40 °C. The mixture was then heated to 75°C over 1 h and analyzed by HPLC every 1 – 2 h for % conversion. When a conversion of > 99% was observed (typically after 5 – 6 h), the reaction was cooled to 20 – 25 °C and extracted with MTBE (2 X 525 mL), with sufficient time to allow for complete phase separation during the extractions. The combined organic extracts were washed with 5% NaCl (2 X 375 mL). The solution was then transferred to equipment appropriate for a 1.5 – 2.5 Torr vacuum distillation that was equipped with a cooled receiver flask. The solution was concentrated under vacuum at < 60°C to remove the solvents. (2,2-Difluoro-l,3-benzodioxol-5-yl)-acetonitrile was then distilled from the resulting oil at 125 – 130 °C (oven temperature) and 1.5 – 2.0 Torr. (2,2-Difluoro-l,3- benzodioxol-5-yl)-acetonitrile was isolated as a clear oil in 66% yield from 5-bromo-2,2- difluoro-l,3-benzodioxole (2 steps) and with an HPLC purity of 91.5% AUC (corresponds to a w/w assay of 95%). Ή NMR (500 MHz, DMSO) 6 7.44 (br s, 1H), 7.43 (d, J= 8.4 Hz, 1H), 7.22 (dd, J= 8.2, 1.8 Hz, 1H), 4.07 (s, 2H).  Synthesis of (2,2-difluoro- l,3-benzodioxol-5-yl)-cycIopropanecarbonitrUe.

Figure imgf000085_0001

MTBE

A stock solution of 50% w/w NaOH was degassed via nitrogen sparge for no less than 16 h. An appropriate amount of MTBE was similarly degassed for several hours. To a reactor purged with nitrogen was charged degassed MTBE (143 mL) followed by (2,2-difluoro-l,3- benzodioxol-5-yl)-acetonitrile (40.95 g, 207.7 mmol) and tetrabutylammonium bromide (2.25 g, 10.38 mmol). The volume of the mixture was noted and the mixture was degassed via nitrogen sparge for 30 min. Enough degassed MTBE is charged to return the mixture to the original volume prior to degassing. To the stirring mixture at 23.0 °C was charged degassed 50% w/w NaOH (143 mL) over 10 min followed by l-bromo-2-chloroethane (44.7 g, 311.6 mmol) over 30 min. The reaction was analyzed by HPLC in 1 h intervals for % conversion. Before sampling, stirring was stopped and the phases allowed to separate. The top organic phase was sampled for analysis. When a % conversion > 99 % was observed (typically after 2.5 – 3 h), the reaction mixture was cooled to 10 °C and was charged with water (461 mL) at such a rate as to maintain a temperature < 25 °C. The temperature was adjusted to 20 – 25 °C and the phases separated. Note: sufficient time should be allowed for complete phase separation. The aqueous phase was extracted with MTBE (123 mL), and the combined organic phase was washed with 1 N HC1 (163mL) and 5% NaCl (163 mL). The solution of (2,2-difluoro- 1,3 -benzodioxol-5-yl)- cyclopropanecarbonitrile in MTBE was concentrated to 164 mL under vacuum at 40 – 50 °C. The solution was charged with ethanol (256 mL) and again concentrated to 164 mL under vacuum at 50 – 60 °C. Ethanol (256 mL) was charged and the mixture concentrated to 164 mL under vacuum at 50 – 60 °C. The resulting mixture was cooled to 20 – 25 °C and diluted with ethanol to 266 mL in preparation for the next step. lH NMR (500 MHz, DMSO) 6 7.43 (d, J= 8.4 Hz, 1H), 7.40 (d, J= 1.9 Hz, 1H), 7.30 (dd, J= 8.4, 1.9 Hz, 1H), 1.75 (m, 2H), 1.53 (m, 2H). [00314] Synthesis of l-(2,2-difluoro-l,3-benzodioxol-5-yl)-cyclopropanecarboxylic acid.

Figure imgf000086_0001

The solution of (2,2-difluoro-l ,3-benzodioxol-5-yl)-cyclopropanecarbonitrile in ethanol from the previous step was charged with 6 N NaOH (277 mL) over 20 min and heated to an internal temperature of 77 – 78 °C over 45 min. The reaction progress was monitored by HPLC after 16 h. Note: the consumption of both (2,2-difluoro-l,3-benzodioxol-5-yl)- cyclopropanecarbonitrile and the primary amide resulting from partial hydrolysis of (2,2-difluoro- l,3-benzodioxol-5-yl)-cyclopropanecarbonitrile were monitored. When a % conversion > 99 % was observed (typically 100% conversion after 16 h), the reaction mixture was cooled to 25 °C and charged with ethanol (41 mL) and DCM (164 mL). The solution was cooled to 10 °C and charged with 6 N HC1 (290 mL) at such a rate as to maintain a temperature < 25 °C. After warming to 20 – 25 °C, the phases were allowed to separate. The bottom organic phase was collected and the top aqueous phase was back extracted with DCM (164 mL). Note: the aqueous phase was somewhat cloudy before and after the extraction due to a high concentration of inorganic salts. The organics were combined and concentrated under vacuum to 164 mL. Toluene (328 mL) was charged and the mixture condensed to 164 mL at 70 – 75 °C. The mixture was cooled to 45 °C, charged with MTBE (364 mL) and stirred at 60 °C for 20 min. The solution was cooled to 25 °C and polish filtered to remove residual inorganic salts. MTBE (123 mL) was used to rinse the reactor and the collected solids. The combined organics were transferred to a clean reactor in preparation for the next step.

Isolation of l-(2,2-difluoro-l,3-benzodioxol-5-yl)-cyclopropanecar boxy lie acid.

Figure imgf000086_0002

The solution of l-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)-cyclopropanecarboxylic acid from the previous step is concentrated under vacuum to 164 mL, charged with toluene (328 mL) and concentrated to 164 mL at 70 – 75 °C. The mixture was then heated to 100 – 105 °C to give a homogeneous solution. After stirring at that temperature for 30 min, the solution was cooled to 5 °C over 2 hours and maintained at 5 °C for 3 hours. The mixture was then filtered and the reactor and collected solid washed with cold 1 :1 toluene/n-heptane (2 X 123 mL). The material was dried under vacuum at 55 °C for 17 hours to provide l-(2,2-difluoro-l,3-benzodioxol-5-yl)- cyclopropanecarboxylic acid as an off-white crystalline solid. l-(2,2-difluoro-l,3-benzodioxol- 5-yl)-cyclopropanecarboxylic acid was isolated in 79% yield from (2,2-difluoro-l,3- benzodioxol-5-yl)-acetonitrile (3 steps including isolation) and with an HPLC purity of 99.0% AUC. ESI-MS m/z calc. 242.04, found 241.58 (M+l)+; Ή NMR (500 MHz, DMSO) δ 12.40 (s, 1H), 7.40 (d, J= 1.6 Hz, 1H), 7.30 (d, J= 8.3 Hz, 1H), 7.17 (dd, J= 8.3, 1.7 Hz, 1H), 1.46 (m, 2H), 1.17 (m, 2H).

Alternative Synthesis of the Acid Chloride Moiety [00319] Synthesis of (2,2-ditluoro-l,3-benzodioxol-5-yl)-methanol.

1. Vitride (2 equiv)

PhCH3 (10 vol)

Figure imgf000087_0001

[00320] Commercially available 2,2-difluoro-l,3-benzodioxole-5-carboxylic acid (1.0 eq) is slurried in toluene (10 vol). Vitride® (2 eq) is added via addition funnel at a rate to maintain the temperature at 15-25 °C. At the end of addition the temperature is increased to 40 °C for 2 h then 10% (w/w) aq. NaOH (4.0 eq) is carefully added via addition funnel maintaining the temperature at 40-50 °C. After stirring for an additional 30 minutes, the layers are allowed to separate at 40 °C. The organic phase is cooled to 20 °C then washed with water (2 x 1.5 vol), dried (Na2SO4), filtered, and concentrated to afford crude (2,2-difluoro-l,3-benzodioxol-5-yl)-methanol that is used directly in the next step.

Synthesis of 5-chloromethyl-2,2-difluoro-l,3-benzodioxole.

1. SOCl2 (1.5 equiv)

DMAP (0.01 equiv)

Figure imgf000087_0002

(2,2-difluoro- 1 ,3-benzodioxol-5-yl)-methanol ( 1.0 eq) is dissolved in MTBE (5 vol). A catalytic amount of DMAP (1 mol %) is added and S0C12 (1.2 eq) is added via addition funnel. The S0C12 is added at a rate to maintain the temperature in the reactor at 15-25 °C. The temperature is increased to 30 °C for 1 hour then cooled to 20 °C then water (4 vol) is added via addition funnel maintaining the temperature at less than 30 °C. After stirring for an additional 30 minutes, the layers are allowed to separate. The organic layer is stirred and 10% (w/v) aq. NaOH (4.4 vol) is added. After stirring for 15 to 20 minutes, the layers are allowed to separate. The organic phase is then dried (Na2SO_ , filtered, and concentrated to afford crude 5-chloromethyl- 2,2-difluoro-l,3-benzodioxole that is used directly in the next step.

Synthesis of (2,2-difluoro-l,3-benzodioxol-5-yl)-acetonitrile.

Figure imgf000088_0001

A solution of 5-chloromethyl-2,2-difluoro- 1 ,3-benzodioxole ( 1 eq) in DMSO ( 1.25 vol) is added to a slurry of NaCN (1.4 eq) in DMSO (3 vol) maintaining the temperature between 30-40 °C. The mixture is stirred for 1 hour then water (6 vol) is added followed by MTBE (4 vol). After stirring for 30 min, the layers are separated. The aqueous layer is extracted with MTBE (1.8 vol). The combined organic layers are washed with water (1,8 vol), dried (Na2S04), filtered, and concentrated to afford crude (2,2-difluoro-l,3-benzodioxol-5-yl)-acetonitrile (95%) that is used directly in the next step.

The remaining steps are the same as described above for the synthesis of the acid moiety.

Amine Moiety

Synthesis of 2-bromo-5-fluoro-4-ntroaniline.

Figure imgf000088_0002
A flask was charged with 3-fluoro-4-nitroaniline (1.0 equiv) followed by ethyl acetate (10 vol) and stirred to dissolve all solids. N-Bromosuccinimide (1.0 equiv) was added as a portion-wise as to maintain internal temperature of 22 °C. At the end of the reaction, the reaction mixture was concentrated in vacuo on a rotavap. The residue was slurried in distilled water (5 vol) to dissolve and remove succinimide. (The succinimide can also be removed by water workup procedure.) The water was decanted and the solid was slurried in 2-propanol (5 vol) overnight. The resulting slurry was filtered and the wetcake was washed with 2-propanol, dried in vacuum oven at 50 °C overnight with N2 bleed until constant weight was achieved. A yellowish tan solid was isolated (50% yield, 97.5% AUC). Other impurities were a bromo-regioisomer (1.4% AUC) and a di- bromo adduct (1.1% AUC). Ή NMR (500 MHz, DMSO) δ 8.19 (1 H, d, J= 8.1 Hz), 7.06 (br. s, 2 H), 6.64 (d, 1 H, J= 14.3 Hz).

Synthesis of benzyIglycoIated-4-ammonium-2-bromo-5-fluoroaniline tosylate salt.

1) l ^OBn

cat. Zn(C104)2-2H20 ®

Figure imgf000089_0001

DCM

A thoroughly dried flask under N2 was charged with the following: Activated powdered 4A molecular sieves (50 wt% based on 2-bromo-5-fluoro-4-nitroaniline), 2-Bromo-5- fluoro-4-nitroaniline (1.0 equiv), zinc perchlorate dihydrate (20 mol%), and toluene (8 vol). The mixture was stirred at room temperature for NMT 30 min. Lastly, (R)-benzyl glycidyl ether (2.0 equiv) in toluene (2 vol) was added in a steady stream. The reaction was heated to 80 °C (internal temperature) and stirred for approximately 7 hours or until 2-Bromo-5-fluoro-4-nitroaniline was <5%AUC.

The reaction was cooled to room temperature and Celite (50 wt%) was added, followed by ethyl acetate (10 vol). The resulting mixture was filtered to remove Celite and sieves and washed with ethyl acetate (2 vol). The filtrate was washed with ammonium chloride solution (4 vol, 20% w/v). The organic layer was washed with sodium bicarbonate solution (4 vol x 2.5% w/v). The organic layer was concentrated in vacuo on a rotovap. The resulting slurry was dissolved in isopropyl acetate (10 vol) and this solution was transferred to a Buchi hydrogenator.

The hydrogenator was charged with 5wt% Pt(S)/C (1.5 mol%) and the mixture was stirred under N2 at 30 °C (internal temperature). The reaction was flushed with N2 followed by hydrogen. The hydrogenator pressure was adjusted to 1 Bar of hydrogen and the mixture was stirred rapidly (>1200 rpm). At the end of the reaction, the catalyst was filtered through a pad of Celite and washed with dichloromethane (10 vol). The filtrate was concentrated in vacuo. Any remaining isopropyl acetate was chased with dichloromethane (2 vol) and concentrated on a rotavap to dryness.

The resulting residue was dissolved in dichloromethane (10 vol). jP-Toluenesulfonic acid monohydrate (1.2 equiv) was added and stirred overnight. The product was filtered and washed with dichloromethane (2 vol) and suction dried. The wetcake was transferred to drying trays and into a vacuum oven and dried at 45 °C with N2 bleed until constant weight was achieved. Benzylglycolated-4-ammonium-2-bromo-5-fluoroaniline tosylate salt was isolated as an off-white solid.

Chiral purity was determined to be >97%ee.

[00334] Synthesis of (3-Chloro-3-methylbut-l-ynyl)trimethylsilane.

Figure imgf000090_0001

[00335] Propargyl alcohol (1.0 equiv) was charged to a vessel. Aqueous hydrochloric acid (37%, 3.75 vol) was added and stirring begun. During dissolution of the solid alcohol, a modest endotherm (5-6 °C) is observed. The resulting mixture was stirred overnight (16 h), slowly becoming dark red. A 30 L jacketed vessel is charged with water (5 vol) which is then cooled to 10 °C. The reaction mixture is transferred slowly into the water by vacuum, maintaining the internal temperature of the mixture below 25 °C. Hexanes (3 vol) is added and the resulting mixture is stirred for 0.5 h. The phases were settled and the aqueous phase (pH < 1) was drained off and discarded. The organic phase was concentrated in vacuo using a rotary evaporator, furnishing the product as red oil. [00336] Synthesis of (4-(Benzyloxy)-3,3-dimethylbut-l-yttyl)trimethylsiIane.

Figure imgf000091_0001

[00337] Method A

[00338] All equivalent and volume descriptors in this part are based on a 250g reaction.

Magnesium turnings (69.5 g, 2.86 mol, 2.0 equiv) were charged to a 3 L 4-neck reactor and stirred with a magnetic stirrer under nitrogen for 0.5 h. The reactor was immersed in an ice- water bath. A solution of the propargyl chloride (250 g, 1.43 mol, 1.0 equiv) in THF (1.8 L, 7.2 vol) was added slowly to the reactor, with stirring, until an initial exotherm (-10 °C) was observed. The Grignard reagent formation was confirmed by IPC usingΉ-NMR spectroscopy. Once the exotherm subsided, the remainder of the solution was added slowly, maintaining the batch temperature <15 °C. The addition required ~3.5 h. The resulting dark green mixture was decanted into a 2 L capped bottle.

[00339] All equivalent and volume descriptors in this part are based on a 500g reaction. A 22 L reactor was charged with a solution of benzyl chloromethyl ether (95%, 375 g, 2.31 mol, 0.8 equiv) in THF (1.5 L, 3 vol). The reactor was cooled in an ice-water bath. Two Grignard reagent batches prepared as described above were combined and then added slowly to the benzyl chloromethyl ether solution via an addition funnel, maintaining the batch temperature below 25 °C. The addition required 1.5 h. The reaction mixture was stirred overnight (16 h).

[00340] All equivalent and volume descriptors in this part are based on a 1 kg reaction. A solution of 15%» ammonium chloride was prepared in a 30 L jacketed reactor (1.5 kg in 8.5 kg of water, 10 vol). The solution was cooled to 5 °C. Two Grignard reaction mixtures prepared as described above were combined and then transferred into the ammonium chloride solution via a header vessel. An exotherm was observed in this quench, which was carried out at a rate such as to keep the internal temperature below 25 °C. Once the transfer was complete, the vessel jacket temperature was set to 25 °C. Hexanes (8 L, 8 vol) was added and the mixture was stirred for 0.5 h. After settling the phases, the aqueous phase (pH 9) was drained off and discarded. The remaining organic phase was washed with water (2 L, 2 vol). The organic phase was concentrated in vacuo using a 22 L rotary evaporator, providing the crude product as an orange oil.

[00341] Method B

[00342] Magnesium turnings (106 g, 4.35 mol, 1.0 eq) were charged to a 22 L reactor and then suspended in THF (760 mL, 1 vol). The vessel was cooled in an ice-water bath such that the batch temperature reached 2 °C. A solution of the propargyl chloride (760 g, 4.35 mol, 1.0 equiv) in THF (4.5 L, 6 vol) was added slowly to the reactor. After 100 mL was added, the addition was stopped and the mixture stirred until a 13 °C exotherm was observed, indicating the Grignard reagent initiation. Once the exotherm subsided, another 500 mL of the propargyl chloride solution was added slowly, maintaining the batch temperature <20 °C. The Grignard reagent formation was confirmed by IPC using Ή-NMR spectroscopy. The remainder of the propargyl chloride solution was added slowly, maintaining the batch temperature <20 °C. The addition required -1.5 h. The resulting dark green solution was stirred for 0.5 h. The Grignard reagent formation was confirmed by IPC using Ή-NMR spectroscopy. Neat benzyl

chloromethyl ether was charged to the reactor addition funnel and then added dropwise into the reactor, maintaining the batch temperature below 25 °C. The addition required 1.0 h. The reaction mixture was stirred overnight. The aqueous work-up and concentration was carried out using the same procedure and relative amounts of materials as in Method A to give the product as an orange oil.

[00343] Syntheisis of 4-Benzyloxy-3,3-dimethylbut-l-yne.

Figure imgf000092_0001

2 steps

[00344] A 30 L jacketed reactor was charged with methanol (6 vol) which was then cooled to 5 °C. Potassium hydroxide (85%, 1.3 equiv) was added to the reactor. A 15-20 °C exotherm was observed as the potassium hydroxide dissolved. The jacket temperature was set to 25 °C. A solution of 4-benzyloxy-3,3-dimethyl-l-trimethylsilylbut-l-yne (1.0 equiv) in methanol (2 vol) was added and the resulting mixture was stirred until reaction completion, as monitored by HPLC. Typical reaction time at 25 °C is 3-4 h. The reaction mixture is diluted with water (8 vol) and then stirred for 0.5 h. Hexanes (6 vol) was added and the resulting mixture was stirred for 0.5 h. The phases were allowed to settle and then the aqueous phase (pH 10-11) was drained off and discarded. The organic phase was washed with a solution of KOH (85%, 0.4 equiv) in water (8 vol) followed by water (8 vol). The organic phase was then concentrated down using a rotary evaporator, yielding the title material as a yellow-orange oil. Typical purity of this material is in the 80% range with primarily a single impurity present. Ή NMR (400 MHz, C6D6) δ 7.28 (d, 2 H, J = 7.4 Hz), 7.18 (t, 2 H, J= 7.2 Hz), 7.10 (d, 1H, J= 7.2 Hz), 4.35 (s, 2 H), 3.24 (s, 2 H), 1.91 (s, 1 H), 1.25 (s, 6 H).

[00345] Synthesis of N-benzylglycolated-5-amino-2-(2-benzyloxy-l,l-dimethylethyl)-6- fluoroindole.

[00346] Method A

[00347] Synthesis of Benzylglycolated 4-Amino-2-(4-benzyloxy-3,3-dimethyIbut- l-ynyl)-5- fluoroaniline.

Figure imgf000093_0001

[00348] Benzylglycolated 4-ammonium-2-bromo-5-flouroaniline tosylate salt was freebased by stirring the solid in EtOAc (5 vol) and saturated NaHCC>3 solution (5 vol) until clear organic layer was achieved. The resulting layers were separated and the organic layer was washed with saturated NaHC03 solution (5 vol) followed by brine and concentrated in vacuo to obtain benzylglocolated 4-ammonium-2-bromo-5-flouroaniline tosylate salt as an oil.

[00349] Then, a flask was charged with benzylglycolated 4-ammonium-2-bromo-5- flouroaniline tosylate salt (freebase, 1.0 equiv), Pd(OAc) (4.0 mol%), dppb (6.0 mol%) and powdered K2CO3 (3.0 equiv) and stirred with acetonitrile (6 vol) at room temperature. The resulting reaction mixture was degassed for approximately 30 min by bubbling in N2 with vent. Then 4-benzyloxy-3,3-dimethylbut-l-yne (1.1 equiv) dissolved in acetonitrile (2 vol) was added in a fast stream and heated to 80 °C and stirred until complete consumption of 4-ammonium-2- bromo-5-flouroaniline tosylate salt was achieved. The reaction slurry was cooled to room temperature and filtered through a pad of Celite and washed with acetonitrile (2 vol). Filtrate was concentrated in vacuo and the residue was redissolved in EtOAc (6 vol). The organic layer was washed twice with NH4CI solution (20% w/v, 4 vol) and brine (6 vol). The resulting organic layer was concentrated to yield brown oil and used as is in the next reaction.

[00350] Synthesis of N-benzylglycolated-5-amino-2-(2-benzyloxy-l,l-dimethylethyl)-6- fluoroindole.

Figure imgf000094_0001

[00351] Crude oil of benzylglycolated 4-amino-2-(4-benzyloxy-3,3-dimethylbut-l-ynyl)-5- fluoroaniline was dissolved in acetonitrile (6 vol) and added (MeCN)2PdCl2 (15 mol%) at room temperature. The resulting mixture was degassed using N2 with vent for approximately 30 min. Then the reaction mixture was stirred at 80 °C under N2 blanket overnight. The reaction mixture was cooled to room temperature and filtered through a pad of Celite and washed the cake with acetonitrile (1 vol). The resulting filtrate was concentrated in vacuo and redissolved in EtOAc (5 vol). Deloxane-II THP (5 wt% based on the theoretical yield of N-benzylglycolated-5-amino-2- (2-benzyloxy-l,l-dimethylethyl)-6-fluoroindole) was added and stirred at room temperature overnight. The mixture was then filtered through a pad of silica (2.5 inch depth, 6 inch diameter filter) and washed with EtOAc (4 vol). The filtrate was concentrated down to a dark brown residue, and used as is in the next reaction.

[00352] Repurification of crude N-benzylglycolated-5-amino-2-(2-benzyloxy- 1,1- dimethylethyl)-6-fluoroindole:

[00353] The crude N-benzylglycolated-5-amino-2-(2-benzyloxy- 1 , l-dimethylethyl)-6- fluoroindole was dissolved in dichloromethane (~1.5 vol) and filtered through a pad of silica initially using 30% EtOAc/heptane where impurities were discarded. Then the silica pad was washed with 50% EtO Ac/heptane to isolate N-benzylglycolated-5-amino-2-(2-benzyloxy-l,l- dimethylethyl)-6-fluoroindole until faint color was observed in the filtrate. This filtrate was concentrated in vacuo to afford brown oil which crystallized on standing at room temperature. Ή NMR (400 MHz, DMSO) 6 7.38-7.34 (m, 4 H), 7.32-7.23 (m, 6 H), 7.21 (d, 1 H, J= 12.8 Hz), 6.77 (d, 1H, J= 9.0 Hz), 6.06 (s, 1 H), 5.13 (d, 1H, J = 4.9 Hz), 4.54 (s, 2 H), 4.46 (br. s, 2 H), 4.45 (s, 2 H), 4.33 (d, 1 H, J= 12.4 Hz), 4.09-4.04 (m, 2 H), 3.63 (d, 1H, J= 9.2 Hz), 3.56 (d, 1H, J= 9.2 Hz), 3.49 (dd, 1H, J= 9.8, 4.4 Hz), 3.43 (dd, 1H, J= 9.8, 5.7 Hz), 1.40 (s, 6 H).

[00354] Synthesis of N-benzyIglycolated-5-amino-2-(2-benzyIoxy-l,l-diniethylethyl)-6- fluoroindole.

[00355] Method B

Figure imgf000095_0001

2. (MeCN)2PdCl2

MeCN, 80 <€

3. Silica gel filtration

[00356] Palladium acetate (33 g, 0.04 eq), dppb (94 g, 0.06 eq), and potassium carbonate (1.5 kg, 3.0 eq) are charged to a reactor. The free based oil benzylglocolated 4-ammonium-2-bromo- 5-flouroaniline (1.5 kg, 1.0 eq) was dissolved in acetonitrile (8.2 L, 4.1 vol) and then added to the reactor. The mixture was sparged with nitrogen gas for NLT 1 h. A solution of 4-benzyloxy- 3,3-dimethylbut-l-yne (70%), 1.1 kg, 1.05 eq) in acetonitrile was added to the mixture which was then sparged with nitrogen gas for NLT 1 h. The mixture was heated to 80 °C and then stirred overnight. IPC by HPLC is carried out and the reaction is determined to be complete after 16 h. The mixture was cooled to ambient temperature and then filtered through a pad of Celite (228 g). The reactor and Celite pad were washed with acetonitrile (2 x 2 L, 2 vol). The combined phases are concentrated on a 22 L rotary evaporator until 8 L of solvent have been collected, leaving the crude product in 7 L (3.5 vol) of acetonitrile. [00357] 5 s-acetonitriledichloropalladium ( 144 g, 0.15 eq) was charged to the reactor. The crude solution was transferred back into the reactor and the roto-vap bulb was washed with acetonitrile (4 L, 2 vol). The combined solutions were sparged with nitrogen gas for NLT 1 h. The reaction mixture was heated to 80 °C for NLT 16 h. In process control by HPLC shows complete consumption of starting material. The reaction mixture was filtered through Celite (300 g). The reactor and filter cake were washed with acetonitrile (3 L, 1.5 vol). The combined filtrates were concentrated to an oil by rotary evaporation. The oil was dissolved in ethyl acetate (8.8 L, 4.4 vol). The solution was washed with 20% ammonium chloride (5 L, 2.5 vol) followed by 5% brine (5 L, 2.5 vol). Silica gel (3.5 kg, 1.8 wt. eq.) of silica gel was added to the organic phase, which was stirred overnight. Deloxan THP II metal scavenger (358 g) and heptane (17.6 L) were added and the resulting mixture was stirred for NLT 3 h. The mixture was filtered through a sintered glass funnel. The filter cake was washed with 30% ethyl acetate in heptane (25 L). The combined filtrates were concentrated under reduced pressure to give N- benzylglycolated-5-amino-2-(2-benzyloxy-l,l-dimethylethyl)-6-fluoroindole as a brown paste ( 1.4 kgl.Svnthesis of Compound 1

[00358] Synthesis of benzyl protected Compound 1.

Figure imgf000096_0001
Figure imgf000096_0002
Figure imgf000096_0003

[00359] 1 -(2,2-difluoro- 1 ,3 -benzodioxol-5-yl)-cyclopropanecarboxylic acid (1.3 equiv) was slurried in toluene (2.5 vol, based on l-(2,2-difluoro-l,3-benzodioxol-5-yi)- cyclopropanecarboxylic acid) and the mixture was heated to 60 °C. SOCl2 (1.7 equiv) was added via addition runnel. The resulting mixture was stirred for 2 hr. The toluene and the excess

SOCI2 were distilled off using rotavop. Additional toluene (2.5 vol, based on l-(2,2-difluoro- l,3-benzodioxol-5-yl)-cyclopropanecarboxylic acid) was added and distilled again. The crude acid chloride was dissolved in dichloromethane (2 vol) and added via addition funnel to a mixture of N-benzylglycolated-5-amino-2-(2-benzyloxy-l,l-dimethylethyl)-6-fluoroindole (1.0 equiv), and triethylamine (2.0 equiv) in dichloromethane (7 vol) while maintaining 0-3 °C (internal temperature). The resulting mixture was stirred at 0 °C for 4 hrs and then warmed to room temperature overnight. Distilled water (5 vol) was added to the reaction mixture and stirred for NLT 30 min and the layers were separated. The organic phase was washed with 20 wt% K2CO3 (4 vol x 2) followed by a brine wash (4 vol) and concentrated to afford crude benzyl protected Compound 1 as a thick brown oil, which was purified further using silica pad filtration.

[00360] Silica gel pad filtration: Crude benzyl protected Compound 1 was dissolved in ethyl acetate (3 vol) in the presence of activated carbon Darco-G (10wt%, based on theoretical yield of benzyl protected Compound 1) and stirred at room temperature overnight. To this mixture was added heptane (3 vol) and filtered through a pad of silica gel (2x weight of crude benzyl protected Compound 1). The silica pad was washed with ethyl acetate/heptane (1:1, 6 vol) or until little color was detected in the filtrate. The filtrate was concentrated in vacuo to afford benzyl protected Compound 1 as viscous reddish brown oil, and used directly in the next step.

[00361] Repurification: Benzyl protected Compound 1 was redissolved in dichloromethane (1 vol, based on theoretical yield of benzyl protected Compound 1) and loaded onto a silica gel pad (2x weight of crude benzyl protected Compound 1). The silica pad was washed with

dichloromethane (2 vol, based on theoretical yield of benzyl protected Compound 1) and the filtrate was discarded. The silica pad was washed with 30% ethyl acetate/heptane (5 vol) and the filtrate was concentrated in vacuo to afford benzyl protected Compound 1 as viscous reddish orange oil, and used directly in the next step. [00362] Synthesis of Compound 1.

Figure imgf000098_0001

OBn 4 steps

Figure imgf000098_0002

[00363] Method A

[00364] A 20 L autoclave was flushed three times with nitrogen gas and then charged with palladium on carbon (Evonik E 101 NN/W, 5% Pd, 60% wet, 200 g, 0.075 mol, 0.04 equiv). The autoclave was then flushed with nitrogen three times. A solution of crude benzyl protected Compound 1 (1.3 kg, ~ 1.9 mol) in THF (8 L, 6 vol) was added to the autoclave via suction. The vessel was capped and then flushed three times with nitrogen gas. With gentle stirring, the vessel was flushed three times with hydrogen gas, evacuating to atmosphere by diluting with nitrogen. The autoclave was pressurized to 3 Bar with hydrogen and the agitation rate was increased to 800 rpm. Rapid hydrogen uptake was observed (dissolution). Once uptake subsided, the vessel was heated to 50 °C.

[00365] For safety purposes, the thermostat was shut off at the end of every work-day. The vessel was pressurized to 4 Bar with hydrogen and then isolated from the hydrogen tank.

[00366] After 2 full days of reaction, more Pd / C (60 g, 0.023 mol, 0.01 equiv) was added to the mixture. This was done by flushing three times with nitrogen gas and then adding the catalyst through the solids addition port. Resuming the reaction was done as before. After 4 full days, the reaction was deemed complete by HPLC by the disappearance of not only the starting material but also of the peak corresponding to a mono-benzylated intermediate. [00367] The reaction mixture was filtered through a Celite pad. The vessel and filter cake were washed with THF (2 L, 1.5 vol). The Celite pad was then wetted with water and the cake discarded appropriately. The combined filtrate and THF wash were concentrated using a rotary evaporator yielding the crude product as a black oil, 1 kg.

[00368] The equivalents and volumes in the following purification are based on 1 kg of crude material. The crude black oil was dissolved in 1 :1 ethyl acetate-heptane. The mixture was charged to a pad of silica gel (1.5 kg, 1.5 wt. equiv) in a fritted funnel that had been saturated with 1 :1 ethyl acetate-heptane. The silica pad was flushed first with 1 :1 ethyl acetate-heptane (6 L, 6 vol) and then with pure ethyl acetate (14 L, 14 vol). The eluent was collected in 4 fractions which were analyzed by HPLC.

[00369] The equivalents and volumes in the following purification are based on 0.6 kg of crude material. Fraction 3 was concentrated by rotary evaporation to give a brown foam (600 g) and then redissolved in MTBE (1.8 L, 3 vol). The dark brown solution was stirred overnight at ambient temperature, during which time, crystallization occurred. Heptane (55 mL, 0.1 vol) was added and the mixture was stirred overnight. The mixture was filtered using a Buchner funnel and the filter cake was washed with 3:1 MTBE-heptane (900 mL, 1.5 vol). The filter cake was air-dried for 1 h and then vacuum dried at ambient temperature for 16 h, furnishing 253 g of Compound 1 as an off-white solid.

[00370] The equivalents and volumes for the following purification are based on 1.4 kg of crude material. Fractions 2 and 3 from the above silica gel filtration as well as material from a previous reaction were combined and concentrated to give 1.4 kg of a black oil. The mixture was resubmitted to the silica gel filtration (1.5 kg of silica gel, eluted with 3.5 L, 2.3 vol of 1 :1 ethyl acetate-heptane then 9 L, 6 vol of pure ethyl acetate) described above, which upon concentration gave a tan foamy solid (390 g).

[00371] The equivalents and volumes for the following purification are based on 390 g of crude material. The tan solid was insoluble in MTBE, so was dissolved in methanol (1.2 L, 3 vol). Using a 4 L Morton reactor equipped with a long-path distillation head, the mixture was distilled down to 2 vol. MTBE (1.2 L, 3 vol) was added and the mixture was distilled back down to 2 vol. A second portion of MTBE (1.6 L, 4 vol) was added and the mixture was distilled back down to 2 vol. A third portion of MTBE (1.2 L, 3 vol) was added and the mixture was distilled back down to 3 vol. Analysis of the distillate by GC revealed it to consist of -6% methanol. The thermostat was set to 48 °C (below the boiling temp of the MTBE-methanol azeotrope, which is 52 °C). The mixture was cooled to 20 °C over 2 h, during which time a relatively fast crystallization occurred. After stirring the mixture for 2 h, heptane (20 mL, 0.05 vol) was added and the mixture was stirred overnight (16 h). The mixture was filtered using a Buchner funnel and the filter cake was washed with 3:1 MTBE-heptane (800 mL, 2 vol). The filter cake was air- dried for 1 h and then vacuum dried at ambient temperature for 16 h, furnishing 130 g of Compound 1 as an off-white solid.

[00372] Method B

[00373] Benzyl protected Compound 1 was dissolved in THF (3 vol) and then stripped to dryness to remove any residual solvent. Benzyl protected Compound 1 was redissolved in THF (4 vol) and added to the hydrogenator containing 5 wt% Pd/C (2.5 mol%, 60% wet, Degussa E5 El 01 N /W). The internal temperature of the reaction was adjusted to 50 °C, and flushed with N2 (x5) followed by hydrogen (x3). The hydrogenator pressure was adjusted to 3 Bar of hydrogen and the mixture was stirred rapidly (>1100 rpm). At the end of the reaction, the catalyst was filtered through a pad of Celite and washed with THF (1 vol). The filtrate was concentrated in vacuo to obtain a brown foamy residue. The resulting residue was dissolved in MTBE (5 vol) and 0.5N HC1 solution (2 vol) and distilled water (1 vol) were added. The mixture was stirred for NLT 30 min and the resulting layers were separated. The organic phase was washed with 10wt% K2CO3 solution (2 vol x2) followed by a brine wash. The organic layer was added to a flask containing silica gel (25 wt%), Deloxan-THP II (5wt%, 75% wet), and

Na2S04 and stirred overnight. The resulting mixture was filtered through a pad of Celite and washed with 10%THF/MTBE (3 vol). The filtrate was concentrated in vacuo to afford crude Compound 1 as pale tan foam.

[00374] Compound 1 recovery from the mother liquor: Option A.

[00375] Silica gel pad filtration: The mother liquor was concentrated in vacuo to obtain a brown foam, dissolved in dichloromethane (2 vol), and filtered through a pad of silica (3x weight of the crude Compound 1). The silica pad was washed with ethyl acetate/heptane (1 :1, 13 vol) and the filtrate was discarded. The silica pad was washed with 10% THF/ethyl acetate (10 vol) and the filtrate was coiicentraied in vacuo to afford Compound 1 as pale tan foam. The above crystallization procedure was followed to isolate the remaining Compound 1.

{00376] Compound 1 recovery from the mother liquor: Option B,

[00377] Silica gel column chromatography: After chromatography on silica gel (50% ethyl acetate/hexaties to 100% ethyl acetate), the desired compound was isolated as pale tan foam. The above crystallization procedure was followed to isolate the remaining Compound 1.

{003781 Additional Recrystaliization of Compound 1

[ 0379j Solid Compound 1 (135 kg) was suspended in IPA (5.4 L, 4 vol) and then heated to 82 °C. Upon complete dissolution (visual), heptane (540 mL, 0.4 vol) was added slowly. The mixture was cooled to 58 °C The mixture was then cooled slowly to 51 °C, during which time crystallization occurs. The heat source was shut down and the recrystalfeation mixture was allowed to cool naturally overnight. The mixture was filtered using a benchtop Buclmer funnel and the filter cake was washed with IPA (2.7 L, 2 vol). The filler cake was dried in the tunnel under air flow for 8 h and then was oven-dried in vacuo at 45-50 °C overnight to give 1.02 kg of recrystallized Compound 1 ,

100380] Compound 1 may also be prepared by one of several synthetic routes disclosed in US published patent application U S20090131 92, incorporated herein by reference.

{003811 Table 6 below recites analytical data for Compound 1.

Table 6.

Figure imgf000101_0001

 Synthesis of Compound 1 Amorphous Form [00383] Spray-Dried Method

[00384] 9.95g of Hydroxypropylmethylcellulose acetate succinate HG grade (HPMCAS-HG) was weighed into a 500 ml beaker, along with 50 mg of sodium lauryl sulfate (SLS). MeOH (200 ml) was mixed with the solid. The material was allowed to stir for 4 h. To insure maximum dissolution, after 2 h of stirring the solution was sonicated for 5 mins, then allowed to continue stirring for the remaining 2 h. A very fin suspension of HPMCAS remained in solution. However, visual observation determined that no gummy portions remained on the walls of the vessel or stuck to the bottom after tilting the vessel.

[00385] Compound 1 (1 Og) was poured into the 500 ml beaker, and the system was allowed to continue stirring. The solution was spray dried using the following parameters:

Formulation Description: Compound 1 Form A/HPMCAS/SLS (50/49.5/0.5)

Buchi Mini Spray Dryer

T inlet (setpoint) 145 °C

T outlet (start) 75 °C

T outlet (end) 55 °C

Nitrogen Pressure 75 psi

Aspirator 100 %

Pump 35 %

Rotometer 40 mm

Filter Pressure 65 mbar

Condenser Temp -3 °C

Run Time l h

REFERENCES

1: Veit G, Avramescu RG, Perdomo D, Phuan PW, Bagdany M, Apaja PM, Borot F, Szollosi D, Wu YS, Finkbeiner WE, Hegedus T, Verkman AS, Lukacs GL. Some gating potentiators, including VX-770, diminish ΔF508-CFTR functional expression. Sci Transl Med. 2014 Jul 23;6(246):246ra97. doi: 10.1126/scitranslmed.3008889. PubMed PMID: 25101887.

2: Pettit RS, Fellner C. CFTR Modulators for the Treatment of Cystic Fibrosis. P T. 2014 Jul;39(7):500-11. PubMed PMID: 25083129; PubMed Central PMCID: PMC4103577.

3: Norman P. Novel picolinamide-based cystic fibrosis transmembrane regulator modulators: evaluation of WO2013038373, WO2013038376, WO2013038381, WO2013038386 and WO2013038390. Expert Opin Ther Pat. 2014 Jul;24(7):829-37. doi: 10.1517/13543776.2014.876412. Epub 2014 Jan 7. PubMed PMID: 24392786.

//////TEZACAFTOR, VX 661, PHASE 3, 1152311-62-0, UNII: 8RW88Y506K,  deltaF508-CFTR corrector, Vertex,  treatment of cystic fibrosis in patients homozygous to the F508del-CFTR mutation

CC(C)(CO)C1=CC2=CC(=C(C=C2N1CC(CO)O)F)NC(=O)C3(CC3)C4=CC5=C(C=C4)OC(O5)(F)F

CC(C)(CO)c1cc2cc(c(cc2n1C[C@H](CO)O)F)NC(=O)C3(CC3)c4ccc5c(c4)OC(O5)(F)F

Share

Nickel-Catalyzed Decarbonylative Suzuki–Miyaura Coupling of Amides To Generate Biaryls

 PROCESS, spectroscopy, SYNTHESIS  Comments Off on Nickel-Catalyzed Decarbonylative Suzuki–Miyaura Coupling of Amides To Generate Biaryls
Jul 112016
 

Thumbnail image of graphical abstract

Shi et al. have reported a nickel-catalyzed decarbonylative Suzuki–Miyaura reaction which uses an N-aroylpiperidine-2,6-dione as the coupling partner for the boronic acid ( Angew. Chem., Int. Ed. 2016, 55, 6959−6963).
The method is attractive from the point of view of the stability of N-aroylpyrrolidine-2,5-diones toward storage and manipulation and the flexibility they add to the chemist’s toolbox, given their preparation from a different group of precursors to aryl halides or triflates.
Notably, the reaction uses an air-stable and inexpensive nickel catalyst, and the reactions tolerate the presence of water. While a standard reaction temperature of 150 °C is quoted, the use of temperatures as low as 80 °C also seem to be possible. Coupling efficiency is reported to be adversely affected when the aromatic rings of both of the coupling partners bear electron-donating substituents.
Ortho substituents on the aromatic rings seem to be beneficial as they facilitate decarbonylation as part of the cross-coupling. Oxidative addition into the N–C(aroyl) bond of the amide is proposed as initiating the catalytic cycle and is possible on account of a reduction in the resonance stabilization of the N-aroyl functionality versus a conventional aromatic amide.

Suzuki–Miyaura Coupling

Synthesis of Biaryls through Nickel-Catalyzed Suzuki–Miyaura Coupling of Amides by Carbon–Nitrogen Bond Cleavage (pages 6959–6963)Shicheng Shi, Guangrong Meng and Prof. Dr. Michal Szostak

Version of Record online: 21 APR 2016 | DOI: 10.1002/anie.201601914

Thumbnail image of graphical abstract

Breaking and making: The first nickel-catalyzed Suzuki–Miyaura coupling of amides for the synthesis of biaryl compounds through N−C amide bond cleavage is reported. The reaction tolerates a wide range of sensitive and electronically diverse substituents on both coupling partners.

STR1

STR1

1H NMR (500 MHz, CDCl3) δ 7.70 (s, 4 H), 7.61 (d, J = 7.3 Hz, 2 H), 7.48 (t, J = 7.6 Hz, 2 H), 7.42 (t, J = 7.3 Hz, 1 H).

 

STR1

13C NMR (125 MHz, CDCl3) δ 144.87, 139.92, 129.48 (q, J F = 32.5 Hz), 129.13, 128.32, 127.56, 127.42, 125.83 (q, J F = 3.8 Hz), 124.46 (q, J F = 270.0 Hz).

 

STR1

19F NMR (471 MHz, CDCl3) δ -62.39.

//////Nickel-Catalyzed,  Decarbonylative Suzuki–Miyaura Coupling,  Amides, Biaryls

Share

Febuxostat

 Uncategorized  Comments Off on Febuxostat
Jul 102016
 
Febuxostat

Febuxostat

Febuxostat; 144060-53-7; Uloric; Adenuric; Tei 6720; 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylic acid;
Molecular Formula: C16H16N2O3S
Molecular Weight: 316.37484 g/mol

2-[3-cyano-4-(2-methylpropoxy)phenyl]-4-methyl-1,3-thiazole-5-carboxylic acid

Febuxostat is a thiazole derivative and inhibitor of XANTHINE OXIDASE that is used for the treatment of HYPERURICEMIA in patients with chronic GOUT.

CAS 144060-53-7

  • 2-[3-Cyano-4-(2-methylpropoxy)phenyl]-4-methyl-5-thiazolecarboxylic acid
  • 2-(3-Cyano-4-isobutyloxyphenyl)-4-methyl-5-thiazolecarboxylic acid
  • FBX
  • Febugood
  • Feburic
  • Febutaz
  • TMX 67
  • Zurig

Febuxostat.png

Febuxostat (INN; trade names Adenuric in Europe and New Zealand, Uloric in the US, Goturic in Latin America, Feburic in Japan) is a drug that inhibits xanthine oxidase, thus reducing production of uric acid in the body. It is used in the treatment of chronicgout and hyperuricemia.

Febuxostat was discovered by scientists at the Japanese pharmaceutical company Teijin in 1998. Teijin partnered the drug with TAP Pharmaceuticals in the US and Ipsen in Europe. Ipsen obtained marketing approval for febuxostat from the European Medicines Agency in April 2008, Takeda obtained FDA approval in February 2009, and Teijin obtained approval from the Japanese “Pharmaceuticals and Medical Devices Agency” in 2011.

Medical uses

Febuxostat is used to treat chronic gout and hyperuricemia.[2] National Institute for Health and Clinical Excellence concluded that febuxostat is more effective than standard doses of allopurinol, but not more effective than higher doses of allopurinol.[2]

Uloric 40 mg tablet

Febuxostat is in the US pregnancy category C; there are no adequate and well-controlled studies in pregnant women.[3]

Side effects

The adverse effects associated with febuxostat therapy include nausea, diarrhea, arthralgia, headache, increased hepatic serum enzyme levels and rash.[3][4]

Drug interactions

Febuxostat is contraindicated with concomitant use of theophylline and chemotherapeutic agents, namely azathioprine and 6-mercaptopurine, because it could increase blood plasma concentrations of these drugs, and therefore their toxicity.[3][5]

Mechanism of action

Febuxostat is a non-purine-selective inhibitor of xanthine oxidase.[3] It works by non-competitively blocking the molybdenum pterincenter which is the active site on xanthine oxidase. Xanthine oxidase is needed to successively oxidize both hypoxanthine andxanthine to uric acid. Hence, febuxostat inhibits xanthine oxidase, therefore reducing production of uric acid. Febuxostat inhibits both oxidized as well as reduced form of xanthine oxidase because of which febuxostat cannot be easily displaced from the molybdenum pterin site.[4]

History

Febuxostat was discovered by scientists at the Japanese pharmaceutical company Teijin in 1998.[6] Teijin partnered the drug withTAP Pharmaceuticals in the US and Ipsen in Europe.[7][8][9]

Ipsen obtained marketing approval for febuxostat from the European Medicines Agency in April 2008,[10] Takeda obtained FDA approval in February 2009,[11][12] and Teijin obtained approval from the Japanese authorities in 2011.[13] Ipsen exclusively licensed its European rights to Menarini in 2009.[14] Teijin partnered with Astellas for distribution in China and southeast Asia.[15][16]

Society and culture

Cost

In the UK, NICE has found that febuxostat has a higher cost/benefit ratio than allopurinol and on that basis recommended febuxostat as a second-line drug for people who cannot use allopurinol.[2]

Trade names

Febuxostat is marketed as Adenuric in Europe and New Zealand, Uloric in the US, Goturic and Goutex in Latin America, Feburic in Japan, and is generic in several countries and is available by many names in those countries.[1]

Febuxostat (Formula I) is an inhibitor of xanthine oxidase, which was discovered by the Japanese company Teijin Pharma Ltd and it is indicated for use in the treatment of hyperuricemia and chronic gout. Its chemical name is 2-(3-cyano-4-isobutoxyphenyl)-4-methyl- l,3-thiazole-5-carboxylic acid. It is marketed under the brand names Adenuric in Europe, Feburic in Japan and Uloric in USA and Canada.

In EP0513379B1 Febuxostat is prepared from 4-hydroxy-3-nitrobenzaldehyde, according to the following scheme.

This particular process suffers from major drawbacks. Not only it is very long, including seven steps from the starting material to the final product, but, most importantly, it employs the use of cyanides, which are extremely toxic reagents. Cyanide salts are likely to generate hydrocyanide, which sets a high amount of risk in an industrial scale process.

In Japanese patent JP06345724A(JP2706037B) the intermediate ethyl ester of Febuxostat is prepared from p-cyano-nitrobenzene, in three steps. Febuxostat may, then, be prepared by alkaline hydrolysis, according to prior art.

MeCSNH,

The use of extremely toxic potassium cyanide makes this process unsuitable for manufacturing purposes.

Route A

In Japanese patent JP3202607B Febuxostat ethyl ester is prepared, according to the above scheme, through two similar routes. Route A uses flash column chromatography for the purification of the hydroxylamine reaction product, while Route B suffers from low yield and the use of chlorinated solvents for recrystallization. In addition, the reaction solvent is, in both cases, formic acid which causes severe skin burns and eye damage to humans. Formic acid is also corrosive towards metal-based materials of construction (MOC), like stainless steel and nickel alloys, limiting the options, essentially, to glass reactors or vessels. The drawbacks of using this solvent are also related to the high volumes of formic acid required per batch, which hinder the waste treatment.

In CN101723915B focus is made to the improvement of the hydroxylamine reaction. Formic acid is replaced with dimethylformamide (DMF) and other solvents. However, according to widely used organic chemistry textbooks, such as March’s Advanced Organic Chemistry, pi 287, 6th edition, M. B. Smith and J. March, ISBN 0-471-72091-7, the mechanism of the reaction involves the formation of an oxime, upon the action of hydroxylamine, which further dehydrates to form a nitrile, with the aid of a suitable reagent, for example formic acid, or acetic anhydride. In the absence of such a reagent, it is expected that the reaction will, at least, not lead to completion, thereby leading to low yields and undesired impurity levels, namely the intermediate oxime. Such impurities, arising from the reactions of the process and which exhibit similar structure of the desired product, are often difficult to remove with common industrial techniques, e.g. crystallization.

In WO2010142653A1 the intermediate Febuxostat ethyl ester is prepared from 4-cyanophenol, through a five-step process. Febuxostat can be prepared from its respective ethyl ester via alkaline hydrolysis, as in the previous case.

OH

 

 

1: patents US5614520 febuxostat synthetic process:

Figure CN104418823AD00031

 

2: Patent JP1994329647 febuxostat synthesis

Figure CN104418823AD00032
Figure CN104418823AD00041

PATENT

https://www.google.com/patents/CN102936230A?cl=en

Gout occurs because the body produces too much uric acid and renal clearance capacity decreased, uric acid accumulation in the body, leading to urate crystals deposited in the joints and organs. Therefore, it means the treatment of gout usually taken to be: to promote uric acid excretion and suppression of uric acid, and the use of appropriate measures to improve symptoms. Uric acid formation and purine metabolism, the final step in the purine metabolism, hypoxanthine generation xanthine xanthine oxidoreductase (XOR) effect, further generate uric acid, inhibit the activity of the enzyme can effectively reduce uric acid production. Febuxostat is currently the world’s newly developed XOR inhibitors, which act by highly selective to the oxidase, reduce uric acid synthesis, reduce uric acid levels, so as to effectively treat the disease ventilation.

Compared with the traditional treatment of gout drug allopurinol, febuxostat has obvious advantages: (1) allopurinol reduced the XOR only inhibit rather than febuxostat of oxidized and reduced form are XOR significant inhibition, thus reducing the role of uric acid, which is more powerful and lasting; (2) Since allopurinol is a purine analogue, the inevitable result of the purine and other activity related to the impact of pyridine metabolism. So allopurinol treatment should be repeated large doses of the drug to maintain a high level. Which also brought serious or even fatal adverse reactions due to drug accumulation due.Instead of febuxostat non-purine XOR inhibitors, so it has better security.

Document TMX-67. Drugs Fut2001, 26, I, 32, and EP0513379, US5614520, W09209279, public

The detailed preparation febuxostat. Using 3-nitro-4-hydroxybenzaldehyde as the starting material is first reacted with hydroxylamine hydrochloride, to give 3-nitro-4-hydroxybenzonitrile. In effect then HCl, reaction with thioacetamide to give 3-nitro-4-hydroxy-thiobenzamide. Closed loop then reacted with 2-chloro ethyl acetoacetate to give 2- (3_ nitro-4-hydroxyphenyl) methyl-5-thiazolyl -4_ carboxylic acid ethyl ester. Followed by potassium carbonate effect, isobutane is reacted with bromo, to give 2- (3_ nitro-4-isobutyloxyphenyl) -4-methyl-5-carboxylic acid ethyl ester. Under the catalytic action of palladium on carbon, hydrogen reduction to give 2- (3-amino-4-isobutyloxyphenyl) -4-methyl-5-thiazole carboxylic acid ethyl ester. Followed by diazotization with sodium nitrite occur, was added cuprous cyanide and potassium cyanide, to give 2- (3-cyano-4-isobutyloxyphenyl) -4-methyl-5-thiazolecarboxylic acid ethyl ester. Finally, under the effect of the hydrolysis of sodium hydroxide, to give the product 2- (3-cyano-4-isobutyloxyphenyl) -4-methyl – thiazole-5-carboxylic acid, to obtain febuxostat.The process route is as follows:

Figure CN102936230AD00041

This route in the preparation of febuxostat, there are many disadvantages: raw 3-nitro-4-hydroxybenzaldehyde in the country is difficult to buy; requires the use of palladium-carbon catalytic hydrogenation reaction under the factory equipment higher requirements, there is a certain danger; the cyano preparation, the need to use sodium nitrite diazotization, could easily lead to corrosion of equipment; the cyano preparation, the need to use toxic cyanide copper, potassium cyanide, pollution, higher risk.

Document JP1994329647, JP1998045733, US3518279 reported another synthesis of febuxostat

Methods. From 4-hydroxy-thiobenzamide as a starting material, and the cyclization reaction to give ethyl 2-bromo-acetyl occurred

2- (4_ hydroxyphenyl) -4_ methyl-5-carboxylic acid ethyl ester in polyphosphoric acid effect, HMTA (hexamethylene tetramine) reacts with 2- (3_ aldehyde – 4-hydroxyphenyl) methyl-5-thiazolyl -4_ carboxylic acid ethyl ester. Then two cases: the first case, the effect of potassium carbonate, is reacted with isobutane to give bromo-2- (4-isobutyloxyphenyl 3_ aldehyde) -4_-methyl-5- thiazole carboxylic acid ethyl ester, and then reacted with hydroxylamine hydrochloride to give 2- (3_-cyano-4-isobutyloxyphenyl) -4_-methyl-5-thiazole carboxylic acid ethyl ester; second case is the first with hydroxylamine hydrochloride to give 2- (3_ cyano-4-hydroxyphenyl) methyl-5-thiazolecarboxylic -4_ carboxylic acid ethyl ester, and then under the effect of potassium carbonate, and reacted with isobutane to give bromo-2- (3 _-cyano-4-isobutyloxyphenyl) -4-methyl-5-carboxylic acid ethyl ester.

Finally, under the effect of the hydrolysis of sodium hydroxide, to give the product 2- (3_-cyano-4-isobutyloxyphenyl) -4_ methyl – thiazole-5-carboxylic acid, i.e., to obtain febuxostat . The process route is as follows:

Figure CN102936230AD00051

This synthesis route febuxostat process, since the introduction of aldehyde HMTA in PPA (polyphosphoric acid) effect. So there are a lot of phosphorus wastewater, serious environmental pollution, but also because PPA has great viscosity, and therefore difficult to stir the production, operation is extremely inconvenient.

Document Heterocyclesl998, 47,2,857 JP1994345724 also reported the synthesis method of febuxostat, using p-nitrophenyl-carbonitrile as a starting material in the reaction with potassium cyanide in DMSO solvent, and then the carbonate lower potassium catalyzed reaction of isobutane and brominated 1,3-cyano-4-diisobutoxybenzene ether. By reaction with thioacetamide to afford

3-cyano-4-isobutyloxyphenyl thiobenzamide. Under heating, and 2-chloro ethyl acetoacetate, ring closure reaction occurs to give 2- (3-cyano-4-isobutyloxyphenyl) -4-methyl-5-carboxylic acid ethyl ester, and finally hydrolysis under the effect of sodium hydroxide, to give the product 2- (3-cyano-4-isobutyloxyphenyl) -4-methyl – thiazole-5-carboxylic acid, to obtain febuxostat.

The present invention febuxostat new technology system, comprising the steps of:

(1) 2-hydroxy-5-cyano – NaSH reacted with benzaldehyde to give 4-hydroxy-3- aldehyde thiobenzamide;

Figure CN102936230AD00061

(2) the step (I) to give 4-hydroxy-3-aldehyde thiobenzamide reaction with ethyl 2-halo-acetyl, closed

Ring to give 2- (3-aldehyde-4-hydroxyphenyl) -4-methyl-5-ethoxycarbonyl thiazole;

Figure CN102936230AD00062

X is a halogen, preferably Cl or Br;

(3) the step (2) to give 2- (3-aldehyde-4-hydroxyphenyl) -4-methyl-5-ethoxycarbonyl thiazole with hydroxylamine in formic acid in the reaction solution to give 2- (3- cyano-4-hydroxyphenyl) -4-methyl-5-ethoxycarbonyl thiazole;

Figure CN102936230AD00063

(4) The step (3) to give 2- (3-cyano-4-hydroxyphenyl) -4-methyl-5-ethoxycarbonyl thiazole isobutane with halo effect in potassium carbonate, to give 2- (3-aldehyde-4-isobutyloxyphenyl) -4-methyl-5-ethoxycarbonyl thiazole;

(5) in step (4) to give 2- (3-aldehyde-4-isobutyloxyphenyl) -4-methyl-5-ethoxycarbonyl-thiazol-off hydrolyzable ester group, to obtain a non-Tendon Disposition Tanzania.

[0011] Scheme of the method is as follows:

Figure CN102936230AD00071

X is halogen, may be Cl, Br;

Preparation 5 febuxostat Example

To a 500ml reaction flask was added 200ml of absolute ethanol, the product of Step 4 was added with stirring (60g, O. 174mol),

5% sodium hydroxide was added 100ml. Stirring heated to 40 degrees, until it is completely dissolved. 40 degrees heat, reaction 4h. The reaction by TLC tracking. After completion of the reaction, the reaction solution was added 10% hydrochloric acid to adjust the pH to 3, the precipitated solid was filtered. And dried to give a pale yellow solid. Dried over anhydrous recrystallized from methanol to give 31. 2g of white crystals, yield 56.7%.

 TLC monitoring of the reaction. Eluent: petroleum ether / ethyl acetate = 3: 1 Melting point:. 201 · 7 ~202 30C (literature value 201 ~202 ° C)

1H-NMR δ:. 1 01 (m, 6H), 2.06 (m, lH), 2.57 (m, 3 H), 3.96 (d, 2H), 7.30 (d, lH), 8.13 (m, 1H), 8. 19 (d, 1H);

MS (m / z):. 316 O (M +)

Infrared detection: 3550-3400cm_1; 2961, 2933,2874; 2227cm_1; 1680U604U511cm_1; 1425cm_1; 1296U283CHT1;

Elemental analysis for C, Η, N, S purified product actual measurement of the content of C, H, N, S content: C:. 60 57%, H:. 5 32%, N:. 8 86%, S: 10. 16%; theoretical value: In C16H16N203S calculated C: 60 74%, H: 510%, N: 885%, S: 1014%..

 

CLIP

 

Facile OnePot Transformation of Arenes into Aromatic Nitriles …

onlinelibrary.wiley.com

Facile OnePot Transformation of Arenes into Aromatic Nitriles under MetalCyanideFree Conditions

 

Patent

Clip

synthesis  describes synthesis of febuxostat (I) from 4-hydroxybenzonitrile (II) in six stages. The synthesis shown is a short, concise route and does not require use of poisonous reagents such as KCN (14). Compound II was converted to 4-hydroxybenzothioamide (III) with 85% yield using NaHS in the presence of hydrated magnesium chloride as Lewis acid. Intermediate III, on cyclization with ethyl-2-chloroacetoacetate, gave thiazole ester (IV) with quantitative yield. In these two stages, the source of potential impurities was identified as an ortho isomer (i.e., 2-hydroxybenzonitrile), which can lead to Impurity VIII and subsequently to Impurity IX . Impurities VIII and IX can be controlled in starting material II with appropriate specification.

 

Figure 2
Figure 2: Impurities identified during the various stages of synthesis of febuxostat.

The ortho formylation of hydroxyl compound IV by using Duff condition (hexamine/TFA) gave aldehyde V (15). The major impurity identified in this reaction was dialdehyde X. Although we have used only 1.0 equivalence of hexamine with respect to Compound IV, the dialdehyde X impurity was formed to a 5-10% ratio in only 2.5 h. It is, therefore, impossible to get rid of this impurity during the reaction, and only effective recrystallization will eliminate it. Impurity X was minimized (≤ 2%) by recrystallization using IPA/H2O (3:5) to get aldehyde V with 50% yield and & #8805; 97% HPLC purity.

Aldehyde V, on alkylation with isobutyl bromide in the presence of potassium carbonate base, gave compound VI with 90% yield. In this stage, Impurities XI and XII were alkylations of carryover Compound IV and dialdehyde, respectively. Two more isomeric impurities n-butyl-aldehyde XIII and 1-methyl propyl-aldehyde XIV were also identified in this stage. Both isomeric impurities can be controlled with appropriate specification for isobutyl bromide. The reaction of Compound VI with hydroxylamine hydrochloride and sodium formate in formic acid at reflux temperature gave Compound VII with 85% yield. Impurities XIII and XIV will also carry forward to impurities n-butyl-nitrile XV and 1-methyl propyl-nitrile XVI, respectively.

In the final step, Compound VII was hydrolyzed using sodium hydroxide in a MeOH:THF:H2O (1:1:1) solvent combination to yield febuxostat (85%). During saponification, methyl ester Impurity XVII was identified via trans-esterification. Its hydrolysis was comparatively slower than its ethyl isomer VII. One way to avoid Impurity XVII is to replace methanol with ethanol. Carryover impurities XI, XV, and XVI were also hydrolyzed to their respective acid derivatives impurities XVIII, XIX, and XX. However, the acid derivatives of impurities X and XII were unexpectedly absent as impurities. It is believed that, because they were present in low concentrations during workup, they were eliminated in the mother liquor. Two additional impurities, amide XXI and diacid XXII, formed by the side reaction of the febuxostat nitrile group with sodium hydroxide, were identified during saponification. The amide XXI and diacid XXII impurities can be controlled by using appropriate equivalence of sodium hydroxide and controlled reaction time. Febuxostat, on acetone recrystallization and seed Crystal A at 45°C, gave pure febuxostat with 75% yield.

http://www.pharmtech.com/investigation-various-impurities-febuxostat

References

  1.  Drugs.com Drugs.com international names for febuxostat Page accessed June 25, 2015
  2.  Febuxostat for the management of hyperuricaemia in people with gout (TA164) Chapter 4. Consideration of the evidence
  3.  Uloric label Updated February, 2009.
  4.  Love BL, Barrons R, Veverka A, Snider KM (2010). “Urate-lowering therapy for gout: focus on febuxostat”. Pharmacotherapy 30 (6): 594–608. doi:10.1592/phco.30.6.594.PMID 20500048.
  5.  Ashraf Mozayani; Lionel Raymon (2011). Handbook of Drug Interactions: A Clinical and Forensic Guide. Springer Science+Business Media.
  6. Teijin Febuxostat Story Page accessed June 25, 2015
  7.  Tomlinson B. Febuxostat (Teijin/Ipsen/TAP). Curr Opin Investig Drugs. 2005 Nov;6(11):1168-78. PMID 16312139
  8.  Bruce Japsen for the Chicago Tribune. August 17, 2006. FDA puts gout treatment on hold
  9.  Note: TAP Pharmaceuticals was a joint venture between Abbott Laboratories and Takedathat was dissolved in 2008 per this press release: Takeda, Abbott Announce Plans to Conclude TAP Joint Venture
  10.  “Adenuric (febuxostat) receives marketing authorisation in the European Union” (PDF). Retrieved 2008-05-28.
  11.  “Uloric Approved for Gout”. U.S. News and World Report. Retrieved 2009-02-16.
  12.  Teijin and Takeda. February 14, 2009 Press release: ULORIC® (TMX-67, febuxostat) Receives FDA Approval for the Chronic Management of Hyperuricemia in Patients with Gout
  13.  Teijin. January 21, 2011 Press release: TMX-67 (febuxostat) Approved in Japan
  14.  Genetic Engineering News. October 2009. Menarini to Market Takeda/Ipsen Gout Therapy in 41 European Countries
  15.  First Word Pharma. April 1st, 2010 Teijin Pharma and Astellas Pharma enter into agreement for marketing rights of TMX-67 in China and Hong Kong
  16.  Research Views. Aug 11 2011 Teijin Pharma Enters Into Distribution Agreement With Astellas Pharma For Febuxostat

Febuxostat is an inhibitor of xanthine oxidase, and was developed by Teijin pharma. This compound is known as a new drug that is effective against gout and hyperuricemia, and it has been 40 years since the last time a drug of this kind of drug was developed.

Febuxostat has therefore gained a lot of popularity and it has already been accepted as a drug in Europe, USA, Korea and Japan. The synthesis of this molecule have been reported in patents by Teijin pharma as shown below.[1,2]

 

2014-04-20_05-03-25

Recently, Itami group was reported the rapoid synthesis of febxostat by using Ni-catalyzed direct coupling of azoles and arylhalides[3]

References

Sorbera, L.A.; Castaner, J.; Rabasseda, X.; Revel, L.; TMX-67. Drugs Fut 2001, 26, 1, 32

[1] Hasegawa, M.; A facile one-pot synthesis of 4-alkoxy-1,3-benzenedicarbonitrile. Heterocycles 1998, 47, 2, 857. [2] Hasegawa, M.;  Hasegawa, M.; Komoriya, K. (Teijin Ltd.); Cyano cpds. and their preparation method. JP 1994345724 . [3] “Nickel-Catalyzed Biaryl Coupling of Heteroarenes and Aryl Halides/Triflates”

Canivet, J.; Yamaguchi, J.; Ban, I.; Itami, K. Org. Lett. 2009, 11, 1733-1736. DOI: 10.1021/ol9001587

ol-2009-001587_0001

Ni-based catalytic systems for the arylation of heteroarenes with aryl halides and triflates have been established. Ni(OAc)2/bipy is a general catalyst for aryl bromides/iodides, and Ni(OAc)2/dppf is effective for aryl chlorides/triflates. Thiazole, benzothiazole, oxazole, benzoxazole, and benzimidazole are applicable as heteroarene coupling partners. A rapid synthesis of febuxostat, a drug for gout and hyperuricemia, is also demonstrated.

A CLIP

A final example of a thiazole containing drug is given in the novel xanthine oxidase inhibitor febuxostat (359, Uloric) which was approved by the FDA in 2009. This inhibitor works by blocking xanthine oxidase in a non-competitive fashion. Consequently, the amount of the oxidation product uric acid is reduced. Thus it is an efficient treatment for hyperuricemia in gout. In order to prepare febuxostat first a synthesis of the noncommercial 4-isobutoxy-1,3-dicyanobenzene building block (363), has to be conducted. An elegant way of achieving this was shown through the reaction of 4-nitrocyanobenzene (360) with potassium cyanide in dry DMSO followed by quenching with isobutyl bromide under basic conditions (Scheme 70). It is suggested that a Meisenheimer-complex intermediate 361 is initially formed, which after rearomatisation, undergoes nucleophilic aromatic substitution of the nitro group by the DMSO solvent [107]. Upon hydrolysis and O-alkylation the desired 4-isobutoxy-1,3-dicyanobenzene (363) is obtained in good overall yield. Subsequently, the less hindered nitrile is converted to the corresponding thioamide 365 in an intriguing reaction using thioacetamide (364). The thiazole ring is then formed by condensation with chloroacetoacetate 366 followed by ester hydrolysis (Scheme 70).

STR1

107 Hasegawa, M. Heterocycles 1998, 47, 857–864. doi:10.3987/COM-97-S(N)89

Paper | Special issue | Vol 47, No. 2, 1998, pp.857-864

DOI: 10.3987/COM-97-S(N)89
A Facile One-Pot Synthesis of 4-Alkoxy-1,3-benzenedicarbonitrile

Masaichi Hasegawa

*Teijin Institute, Bio-Medical Research, Asahigaoka 4-3-2, Hino, Tokyo 191, Japan

Abstract

2-(3-Cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxlic acid (TEI-6720) was prepared. The introduction of cyano group to 4-nitrobenzonitrile with KCN in dry DMSO followed by quenching with alkyl halide afforded the key intermediates, 4-alkoky-1,3-benzenedicarbonitriles, in good yield. The reaction was completed in dry DMSO, while no reaction occurred in dry DMF. This observation can be suggested by the participation of DMSO in the reaction.

PDF (208KB)

http://www.beilstein-journals.org/bjoc/single/articleFullText.htm?publicId=1860-5397-7-57#S70

 

 

A CLIP

Synthesis and characterization of process-related impurities of an anti-hyperuricemia drug-Febuxostat

Venkateswara Rao Vallu,$ Krunal Girishbhai Desai, Sandip Dhaya Patil, Rajendra Agarwal, Pratap Reddy Padi and Mahesh Reddy Ghanta

*Process Research Laboratory-I, Research & Development Centre, Macleods Pharmaceuticals Ltd, G-2, Mahakali Caves Road, Shantinagar, Andheri (East), Mumbai, Maharastra, India

$Department of Chemistry, Pacific University, Pacific Hills, Airport Road, Pratap Nagar Extension, Debari, Udaipur, Rajasthan, India _____________________________________________________________________

Der Pharma Chemica, 2014, 6(3):300-311 (http://derpharmachemica.com/archive.html)

http://derpharmachemica.com/vol6-iss3/DPC-2014-6-3-300-311.pdf

Synthesis of 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylic acid (1) [10] A solution of 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylic acid (1 tech grade, 5.0 g, 0.015 mol.) in methanol (50.0 mL) was heated the reaction mass at 60-65°C till clear solution was obtained. Water (50.0 mL) was added drop wise into reaction mass with in 30.0 min. at 60-65°C. Resultant white crystalline solid was filtrated, Mahesh Reddy Ghanta et al Der Pharma Chemica, 2014, 6 (3):300-311 _____________________________________________________________________________ 302 www.scholarsresearchlibrary.com washed with water (10.0 mL) and dried in vacuum tray drier at 50-55°C under vacuum to give

2-(3-cyano-4- isobutoxyphenyl)-4-methylthiazole-5-carboxylic acid (1). Yield: 95.0 % (4.75 g)

mp 239°C. Purity by HPLC: 99.74 % (10.2 min. retention time),

Anal. Calcd for C16H16N2O3S: C, 60.74; H, 5.10; N, 8.85. Found: C, 60.70; H, 5.11; N, 8.87 %;

IR (KBr) υmax (in cm−1): 3834.61, 3742.03, 3680.30, 3556.85, 3456.55, 2962.76, 2877.89, 2661.85, 2546.12, 2353.23, 2229.79, 2168.06, 2029.18, 1921.16, 1790.00, 1674.27, 1604.83, 1512.24, 1427.37, 1381.08, 1280.78, 1172.76, 1118.75, 1010.73, 918.15, 833.28, 771.55, 725.26, 648.10, 524.66, 462.93; 1H NMR (300 MHz, CDCl3 or DMSO-d6) δH (in ppm): 1.00-1.02 (d, 6H, (CH3)2-CH-), 2.49-2.50 (m, 1H, (CH3)2-CH-), 3.97-3.99 (d, 2H, -CH-CH2−), 7.33–8.25 (d, dd, 3H, Ar-H), 2.64 (s, 3H, -CH3), 13.39 (s, 1H, -COOH);

13C NMR (300 MHz, DMSO–d6) δC (in ppm) (Positiona ): 166.3 (l), 162.9 (p), 162.2 (n), 159.6 (e), 133.1 (g), 131.6 (i), 125.5 (m), 123.0 (h), 115.5 (k), 114.0 (f), 101.7 (j), 75.2 (d), 27.7 (b), 18.8 (a, c), 17.1 (o);

MS m/z (%) (70 eV): m/z =317.0 (100.0 %) [M+1], 318.0 (16.0 %) [M+2], 403.0 (63.0 %), 512.0 (47.0 %), 482.0 (46.0 %), 405.0 (27.0 %), 468.0 (25.0 %), 570.0 (24.0 %).

STR1

 

 

PATENT

WO 2012066561

As per the present invention, hydroxylamine hydrochloride is added to compound of Formula-Ill in presence of a polar aprotic solvent like DMSO, DMA, ACN or DMF. To this reaction mixture acetyl halides or sulfonyl chlorides are added and temperature raised to 70 -80 °C. Acetyl halides are selected from acetyl bromide or acetyl chloride. Sulfonyl chlorides are selected from methane sulfonyl chloride or para toluene sulfonyl chloride. To this reaction mixture a base selected from alkali metal carbonates like potassium carbonate or sodium carbonate, preferably potassium carbonate and alkyl halide selected from isobutyl bromide is successively added. The reaction mass is washed with water and compound of Formula-II is isolated. In one embodiment the present invention provides, process for the preparation of Febuxostat comprising the steps of:

a) reacting the compound of Formula-III(a) with hydroxylamine hydrochloride in presence of organic solvent;

Figure imgf000008_0001

Formula-III(a)

b) adding acyl halides or sulfonyl chlorides to the reaction mixture;

c) optionally isolating compound of Formula- IV (a)

Figure imgf000008_0002

Formula-IV(a)

d) reacting with isobutyl bromide in presence of base;

e) isolating the compound of Formula-II(a); and

Figure imgf000008_0003

FormuIa-II(a)

f) hydrolyzing the compound of Formula-II(a) to get Febuxostat.

The following examples are provided to illustrate the process of the present invention. They, are however, not intended to limiting the scope of the present invention in any way and several variants of these examples would be evident to person ordinarily skilled in the art. Experimental procedure:

Example – 1: Preparation of Ethyl-2-(3-cyano-4-isobutoxy phenyl)-4-methyI thiozole -5-carboxylate

A mixture of 10. Og of Ethyl -2-(3-formyl-4-hydroxy phenyl)-4-methyl thiozole -5- carboxylate and 2.85 g of hydroxylamine hydrochloride were stirred for 30 minutes in 40 g of Dimethyl sulfoxide. To this reaction mixture 3.3 grams of acetyl chloride was added and stirred at 70 -80°C for 2-3 hours. Reaction mass was cooled to room temperature and to this 19 g of potassium carbonate and 19 g of isobutyl bromide was added successively. The reaction mass was stirred for 5 hours at 70-80°C. Reaction mass was diluted with 200 ml of purified water. The reaction mass was filtered and washed with purified water to give 10.0 g of Ethyl-2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxyltae (yield 84.0%)

Example – 2: Preparation of Ethyl-2-(3-cyano-4-hydroxyphenyl)-4-methyl thiozole – 5-carboxylate

A mixture of 10. Og of Ethyl-2-(3-formyl-4-hydroxy phenyl)-4-methyl thiozole -5- carboxylate and 2.85 g of hydroxylamine hydrochloride were stirred for 30 minutes in 30 g of Dimethylformamide. To this reaction mixture 3.3 grams of acetyl chloride was added and stirred at 90°C for 2-3 hours. Reaction mass was cooled to room temperature and diluted with 100 ml of water and stir for 2 hours. The reaction mass was filtered and washed with purified water to give 10.0 g of Ethyl-2-(3-cyano-4-hydroxy phenyl)-4- methyl thiozole -5-carboxyltae (yield 99.0%).

Example – 3: Preparation of Ethyl 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxylate

A mixture of 10. Og of Ethyl-2-(3-cyano-4-hydroxy phenyl)-4-methyl thiozole -5- carboxylate, 30 g of NMP, 9.6 g of potassium carbonate and 7.2 g of isobutyl bromide were stirred for 3 hours at 90°C. Reaction mass was diluted with 100 ml of purified water. The reaction mass was filtered and washed with purified water and ethanol to give 10.5 g of Ethyl-2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxyltae (yield 88.0%). Example – 4: Preparation of 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5- carboxylic acid

A mixture of 10. Og of Ethyl-2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5- carboxyltae, 2.0g of sodium hydroxide was heated at 45-60°C in 75 ml of aqueous methanol for 1 hour. Reaction mass was cooled to ambient temperature and pH adjusted to 2.0 to 2.5 with dilute hydrochloric acid and precipitated crystal was collected by filtration to give 8.8g of 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxylic acid (yield 95.8%).

Example – 5-13: Preparation of 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole – 5-carboxylic acid

The above compound was prepared by following the procedure as disclosed in Example- 4, using the below listed solvents instead of aqueous methanol.

Figure imgf000010_0001

Example – 14: Preparation of pure 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxylic acid

10.0 g of 2-(3-cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxylic acid was dissolved in 100 ml of ethanol at reflux temperature. After dissolution reaction mass was cooled and precipitated crystal was collected by filtration to give 9.6 g of pure 2-(3- cyano-4-isobutoxy phenyl)-4-methyl thiozole -5-carboxylic acid (yield 96%).

PATENT

KR 201603732

PATENT

WO 2015018507

https://www.google.com/patents/WO2015018507A2?cl=en

EXPERIMENTAL

of compound of formula lib

Dissolve 14.14g of ethyl 2-(3-formyl-4-hydroxyphenyl)-4-methylthiazole-5-carboxylate (Formula III) in 55 ml dimethylformamide, at ambient temperature. Add 40g of potassium carbonate, along with 15.9 ml isobutyl bromide. Heat the reaction to 75-80 °C and stir for 4 hours. Cool to 25-30 °C, while 165 ml process water is added. Further cool to 0-5 °C and stir for 30 minutes at this temperature. Filter off the precipitated solid and wash the filter cake with 55 ml process water. The wet cake is dried under vacuum at 40 °C for 7 hours, to furnish 16.43 g of ethyl 2-(3-formyl-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate (Formula lib).

of compound of formula Illb

In a 25 mL round-bottomed flask charge under stirring at 25-30 °C, 1.0 g (2.88 mmol) of ethyl 2-(3-formyl-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate in 3.0 mL dimethylformamide. Add 34 mg (0.19 mmol) copper acetate under stirring at 25-30 °C. Flush with oxygen (02) and add 0.66 ml (34.92 mmol) 25% aqueous ammonia. Flush again with 02. Heat the reaction mixture to 80-82 °C overnight. Check the progress of the reaction by TLC (cyclohexanerethyl acetate 3:1). Cool reaction mass to 25-30 °C. Add 25mL ethyl acetate and 25mL brine at the reaction mass, separate organic layer and extract aqueous layer twice with 25mL ethyl acetate. Combine organic layers, dry over anhydrous sodium sulfate, filter off and concentrate till dry. The residue is purified with column chromatography (cyclohexane:ethyl acetate 9:1). afforded 0.754g of ethyl 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate (Formula Illb) Yield: 75.4%.

EXAMPLE 3: Preparation of compound of formula Illb

In a 25 mL round-bottomed flask charge under stirring at 25-30 °C, 0.17 g (0.49 mmol) of ethyl 2-(3-formyl-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate in 2.5 mL tetrahydrofuran. Add 2.9 mL (153.43 mmol) 25% aqueous ammonia, under stirring at 25-30 °C. Add 137 mg (0.54 mmol) iodine (I2) to the reaction mass, stir the reaction mixture at 25-30 °C for 15-30 min. Check the progress of the reaction by TLC (cyclohexane: ethyl acetate 3:1). Starting material is consumed. Add 2.5 mL 5% w/v aqueous sodium thiosulfate Na2S203 and 15mL ethyl acetate at the reaction mass, separate organic layer and extract twice aqueous layer with 15mL ethyl acetate. Combine organic layers, dry over anhydrous sodium sulfate, filter off and concentrate till dry. 0.158g of ethyl 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate (Formula Illb) are collected.

EXAMPLE 4: Preparation of Febuxostat

In a 100 ml 2-neck round-bottomed flask charge 2.407g of ethyl-2-(3-cyano-4-isobutoxyphenyl)-4-methylhiazole-carboxylate in 20ml tetrahydrofuran under stirring, at 25-35 °C, 0.748g of sodium hydroxide and heat reaction mass to 60-65 °C for approximately 8 hrs. Check the progress of the reaction by TLC (cyclohexane:ethyl acetate 3:1). Cool reaction mass to 0-5 °C and add 50 ml process water keeping temperature within 0-5 °C. Adjust pH to 1-2 with 4.5 ml 6 N hydrochloric acid, keeping temperature within 0-5 °C. Warm up reaction mass to 25-30 °C and stir reaction mass at the above temperature for 15 min. Filter off the precipitated solid through Buchner funnel under reduced pressure, spray wash with 2 ml process water and suck dry for 20-30 min. Transfer the crude solid in a 50 ml round-bottomed flask, charge 12 ml process water and 12 ml acetone at 25-30°C. Heat the reaction mass to 50-60 °C for 60 min. Cool down reaction mass to 0-5 °C and stir for 60 min at the above temperature. Filter off the precipitated solid though Buchner funnel under reduced pressure, spray wash with 2 ml of a 1 : 1 mixture of acetone and process water and suck dry for 30-45 min. Dry under vacuum at 60 °C. 1.821g of (compound I) Febuxostat are collected, Purity: 82.6%, Yield: 0.62w/w.

on of compound of formula Ilia

In a 50 mL round-bottomed flask charge under stirring 0.5g (1.72 mmol) of ethyl 2-(3-formyl-4-hydroxyphenyl)-4-methylthiazole-5-carboxylate in 8.6 mL THF, at 25-30 °C. Add 10.3 mL (544.94 mmol) 25% aqueous ammonia, under stirring at 25-30 °C. Add 480 mg (1.89 mmol) iodine (I2) to the reaction mass, stir the reaction mixture at 25-30 °C for 15-30 min. Check the progress of the reaction by TLC (cyclohexane: ethyl acetate 1 :1). Starting material is consumed. Add 8.6 mL 5% w/v aqueous thiosulfate and 40 mL ethyl acetate at the reaction mass, separate organic layer and extract aqueous layer twice with 40 mL ethyl acetate. Combine organic layers, dry over anhydrous sodium sulfate, filter off and concentrate to dryness. Purification of the residue with column chromatography (cyclohexane: ethyl acetate 3: 1) afforded 0.213 g of ethyl 2-(3-cyano-4-hydroxyphenyl)-4-methylthiazole-5-carboxylate (Formula Ilia). Yield : 42.6%.

EXAMPLE 6: Preparation of compound Illb

Dissolve 2.2 g of ethyl 2-(3-cyano-4-hydroxyphenyl)-4-methylthiazole-5-carboxylate (Formula VI) in 7 ml dimethylformamide and to this mixture add 6.6 g of potassium carbonate and 3.14 g of isobutyl bromide. Stir the reaction at 75 °C for 15 hours and then cool to 40 °C. Add 15 ml process water and cool to 0-5 °C. Filter the precipitated solid off and wash with 15 ml process water, which, after drying, affords 2.28 g of ethyl 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylate (Formula Illb).

EXAMPLE 7: Preparation of compound I (Febuxostat)

In a 100 ml 2-neck round-bottomed flask charge 2.131 g of ethyl-2(3-cyano-4-isobutoxyphenyl)-4-methylhiazole-carboxylate, 64 ml methanol and 2.5 ml process water are added under stirring at 25-35 °C. Add 1.718 g potassium carbonate and heat reaction mass to reflux for approximately 2-3 hrs. Check the progress of the reaction by TLC (cyclohexane: ethyl acetate 3:1). Cool reaction mass to 20-25 °C. Concentrate solvent at below 40 °C. To the residue add 43 ml process water, 21 ml ethyl acetate and stir for 30 min at 25-35 °C. Separate layers and transfer aqueous layer in a 100 ml round-bottomed flask. Adjust pH to 2.3-2.7 with 25 ml 1 N hydrochloric acid, at 25-35 °C. Warm up reaction mass to 40 °C and stir reaction mass at this temperature for 60-90 min. Cool down reaction mass to 25-35 °C. Filter off the precipitated solid through Buchner funnel under reduced pressure, spray wash with 5 ml process water and suck dry for 30-45 min. Dry under vacuum at 60 °C. 1.708g of (compound I) Febuxostat are collected, Purity: 86.7%, Yield: 0.69w/w.

EXAMPLE 8: Preparation of Febuxostat crystalline form III

In a 250 mL round-bottomedflask charge under stirring at 25-30 °C 10 g of crude 2-(3-cyano-4-isobutoxyphenyl)-4-methylthiazole-5-carboxylic acid (Febuxostat) in 200 mL ethyl acetate. Heat reaction mass to reflux and stir for 30 min. Cool reaction mass to 25-30°C. Warm again reaction mass and partially distill off solvent from the reaction mass at temperature below 40 °C under reduced pressure. Cool reaction mass to 25-30°C. Filter off the precipitated solid through Buchner funnel under reduced pressure and spray wash with 10 mL ethyl acetate. Dry under vacuum at 60°C. 8.5 g of Febuxostat are collected. Yield: 85 % w/w. XRPD of crystalline compound is in accordance with the one reported in Chinese patent CN101412700B.

PATENT

CN 104418823

https://www.google.com/patents/CN104418823A?cl=zh

Figure CN104418823AD00042

PATENT

CN 103588723

https://www.google.com/patents/CN103588723A?cl=zh

Chinese patent CN1275126 described by the Japanese company Teijin invention relates febuxostat Form A, B, C, D, G, and six kinds of amorphous and crystalline preparation method, reported in the literature Form A relatively stable . The method used is a solvent of methanol and water, patent phase diagram (Figure 7 Zone I) can be obtained in anhydrous crystalline Form A (hereinafter referred to as “Form A”), the mixing process by a temperature and the formation of methanol and water to determine the composition of the solvent, and the need to add a certain amount of Form a as a seed crystal to induce precipitation of crystals to control crystallization conditions are very harsh, operable range is very small, easy to form methanol solvate, hydrate or stable crystalline type C, to obtain reproducible single crystal type a low, it is difficult to achieve industrial production, and no mention of the preparation of Form a yield and purity in this patent.

[0011]

[0012] Chinese patent CN102267957A invention discloses a method for preparing febuxostat Form A, the solvent is preferably acetone, dissolved into 25 ~ 40 ° C was allowed to stand, when there began to crystallize when stirred for 20 to 40 minutes, then placed in -15 ~ 0 ° C to continue the crystallization of 8 to 10 hours. The crystallization process need to well below zero, when industrial mass production, resulting in high production costs, is not conducive to industrial production, the process yield up to 95.4%.

[0013] Chinese patent CN101139325 of Example 7 discloses the preparation of Form A with acetone method, although the process is simple, but the yield is low, only 50%.

[0014] Although the Chinese patent CN101684108A isopropyl alcohol as a solvent is disclosed a method for preparing crystalline form, the crystalline form of preparation is used to cool and heat a phased manner was allowed to stand, the crystallization temperature, long crystallization time, about 30 hours, the yield is low, and its products are not crystalline Form A.

[0015] In addition, Chinese patent CN101525319A, CN101805310, CN101926795A, CN101926794, W02012020272A2 are disclosed ethanol as a solvent or aqueous ethanol as a solvent preparation methods, and its products are crystalline ethanol solvate.

[0016] World Patent W02011139886A2 discloses the use of a mixed solvent of alcohol, and its products are not obtained polymorph A0

PAPER

Letters in Organic Chemistry (2015), 12(3), 217-221

Synthesis of the Major Metabolites of Febuxostat

Author(s): Xiao Long Li, Rui Qiu, Wei Li Wan, Xu Cheng, Li Hai and Yong Wu

Affiliation: Key Laboratory of Drug Targeting of Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.

Graphical Abstract:

 

Abstract:

Total synthesis of three Febuxostat metabolites, named 67M-1, 67M-2, and 67M-4,is described in this article. Through condensation of the key intermediate compound A with different side chains, and then oxidation and hydrolysis, we obtained three target compounds with an overall yield of 19.5%-28.0%.

VOLUME: 12
ISSUE: 3
Page: [217 – 221]
Pages: 5
DOI: 10.2174/1570178612666150108000805http://www.eurekaselect.com/127479/article

 

 

ULORIC (febuxostat) is a xanthine oxidase inhibitor. The active ingredient in ULORIC is 2-[3-cyano-4-(2-methylpropoxy) phenyl]-4-methylthiazole-5-carboxylic acid, with a molecular weight of 316.38. The empirical formula is C16H16N2O3S.

The chemical structure is:

ULORIC (febuxostat) Structural Formula Illustration

Febuxostat is a non-hygroscopic, white crystalline powder that is freely soluble in dimethylformamide; soluble in dimethylsulfoxide; sparingly soluble in ethanol; slightly soluble in methanol and acetonitrile; and practically insoluble in water. The melting range is 205°C to 208°C.

LORIC tablets for oral use contain the active ingredient, febuxostat, and are available in two dosage strengths, 40 mg and 80 mg. Inactive ingredients include lactose monohydrate, microcrystalline cellulose, hydroxypropyl cellulose, sodium croscarmellose, silicon dioxide and magnesium stearate. ULORIC tablets are coated with Opadry II, green.

CN1642546A * Mar 28, 2003 Jul 20, 2005 Teijin Ltd. Containing a single crystalline solid preparation
CN102471295A * Jul 14, 2010 May 23, 2012 Teijin Pharma Ltd. The method of manufacturing the poor solvent additive method of 2- (3-cyano-4-isobutyl-phenyl) -4-methyl-5-carboxylic acid crystalline polymorph of
EP2502920A1 * Mar 25, 2011 Sep 26, 2012 Sandoz Ag Crystallization process of Febuxostat from A
JP2011020950A * Title not available
WO2015018507A3 * Jul 30, 2014 Oct 22, 2015 Pharmathen S.A. A novel process for the preparation of febuxostat
CN103304512A * Jun 4, 2013 Sep 18, 2013 华南理工大学 Preparation method for febuxostat
WO2011031409A1 * Aug 12, 2010 Mar 17, 2011 Teva Pharmaceutical Industries Ltd. Processes for preparing febuxostat
JP2834971B2 Title not available
JP3202607B2 Title not available
JPH1045733A * Title not available
US5614520 Jan 30, 1995 Mar 25, 1997 Teijin Limited 2-arylthiazole derivatives and pharmaceutical composition thereof
CN102229581A * Nov 15, 2010 Nov 2, 2011 邹巧根 Preparation method for febuxostat intermediate
JPH1045733A * Title not available
CN101497589A * Feb 26, 2009 Aug 5, 2009 沈阳药科大学 Method for synthesizing 2-(3-cyano-4- isobutoxy phenyl)-4-methyl-carboxylate
CN101863854A * Jun 29, 2010 Oct 20, 2010 沈阳药科大学 Synthesis method of 2-(3-cyan-4-isobutoxy) phenyl-4-methyl-5-thiazole formic acid
JP2706037B2 * Title not available
Reference
1 * HASEGAWA, M. ET AL.: ‘A facile one-pot synthesis of 4-alkoxy-1,3-benzenedicarbonitrile‘ HETEROCYCLES vol. 47, no. 2, 1998, pages 857 – 864
Citing Patent Filing date Publication date Applicant Title
WO2012131590A1 * Mar 28, 2012 Oct 4, 2012 Sandoz Ag An improved process for preparation of febuxostat and its polymorphic crystalline form c thereof
WO2014009817A1 * Mar 19, 2013 Jan 16, 2014 Alembic Pharmaceuticals Limited Pharmaceutical composition of febuxostat
WO2014057461A1 Oct 10, 2013 Apr 17, 2014 Ranbaxy Laboratories Limited Process for the preparation of crystalline form g of febuxostat
Febuxostat
Febuxostat.svg
Systematic (IUPAC) name
2-(3-cyano-4-isobutoxyphenyl)-4-methyl-
1,3-thiazole-5-carboxylic acid
Clinical data
Trade names Uloric, Adenuric, Atenurix, Feburic, Goturic, Goutex. Generic in several countries.[1]
AHFS/Drugs.com Monograph
MedlinePlus a609020
License data
Pregnancy
category
  • US: C (Risk not ruled out)
Routes of
administration
Oral
Legal status
Legal status
Pharmacokinetic data
Bioavailability ~49% absorbed
Protein binding ~99% to albumin
Metabolism via CYP1A2, 2C8, 2C9,UGT1A1, 1A3, 1A9, 2B7
Biological half-life ~5-8 hours
Excretion Urine (~49% mostly as metabolites, 3% as unchanged drug); feces (~45% mostly as metabolites, 12% as unchanged drug)
Identifiers
CAS Number 144060-53-7 
ATC code M04AA03 (WHO)
PubChem CID 134018
IUPHAR/BPS 6817
DrugBank DB04854 Yes
ChemSpider 118173 Yes
UNII 101V0R1N2E Yes
KEGG D01206 Yes
ChEMBL CHEMBL1164729 Yes
Chemical data
Formula C16H16N2O3S
Molar mass 316.374 g/mol

/////////Febuxostat, 144060-53-7, Uloric, Adenuric,  Tei 6720,  thiazole derivative, inhibitor of XANTHINE OXIDASE,  treatment of HYPERURICEMIA, chronic GOUT, FBX, Febugood, Feburic, Febutaz, TMX 67, Zurig

CC1=C(SC(=N1)C2=CC(=C(C=C2)OCC(C)C)C#N)C(=O)O

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: