New Drug Approvals

Home » Posts tagged 'PHASE 3' (Page 12)

Tag Archives: PHASE 3

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

Blog Stats

  • 4,802,069 hits

Flag and hits

Flag Counter

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

Join 37.9K other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

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

Join 37.9K other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

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

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

EVOGLIPTIN


ChemSpider 2D Image | Evogliptin | C19H26F3N3O3

EVOGLIPTIN
CAS: 1222102-29-5 FREE

HCL……1246960-27-9

tartare.. 1222102 -51-3

Dong-A Pharmaceutical. Co., Ltd동아제약 주식회사
2-Piperazinone, 4-((3R)-3-amino-1-oxo-4-(2,4,5-trifluorophenyl)butyl)-3-((1,1-dimethylethoxy)methyl)-, (3R)-
R)-4-((R)-3-Amino-4-(2,4,5-trifluorophenyl)-butanoyl)-3-(t-butoxymethyl)-piperazin-2-one

4-[3(R)-Amino-4-(2,4,5-trifluorophenyl)butyryl]-3(R)-(tert-butoxymethyl)piperazin-2-one hydrochloride

DA-1229

see…http://www.allfordrugs.com/2015/07/03/evogliptin/

DA-1229 is a dipeptidyl peptidase IV (CD26) inhibitor currently being developed in phase III clinical studies at Dong-A for the treatment of type 2 diabetes.

In 2014, Eurofarma aquired rights for product development and commercialization in Brazil.

Evogliptin Tartrate

All About Drugs (1)

All About Drugs (2)

If above image is not clear then see at…….http://www.allfordrugs.com/2015/07/03/evogliptin/

86…………H. J. Kim, W. Y. Kwak, J. P. Min, J. Y. Lee, T. H. Yoon, H. D. Kim, C. Y. Shin, M. K.
Kim, S. H. Choi, H. S. Kim, E. K. Yang, Y. H. Cheong, Y. N. Chae, K. J. Park, J. M.
Jang, S. J. Choi, M. H. Son, S. H. Kim, M. Yoo and B. J. Lee, Bioorg. Med. Chem. Lett.,
2011, 21 (12), 3809-3812.
[87] …………K. S. Lim, J. Y. Cho, B. H. Kim, J. R. Kim, H. S. Kim, D. K. Kim, S. H. Kim, H. J. Yim,
S. H. Lee, S. G. Shin, I. J. Jang and K. S. Yu, Br. J. Clin. Pharmacol., 2009, 68 (6), 883-
890.

  • Originator Dong-A Pharmaceutical
  • Developer Dong-A ST
  • Class Amides; Antihyperglycaemics; Fluorobenzenes; Piperazines; Small molecules
  • Mechanism of Action CD26 antigen inhibitors
  • Orphan Drug Status No
  • On Fast track No
  • New Molecular Entity Yes
  • Available For Licensing Yes – Type 2 diabetes mellitus

Highest Development Phases

  • Phase III Type 2 diabetes mellitus

Most Recent Events

  • 01 Sep 2014 Phase-I clinical trials in Type-2 diabetes mellitus (In volunteers) in United Kingdom (PO)
  • 31 Jul 2014 Phase-III clinical trials in Type-2 diabetes mellitus in South Korea (PO)
  • 31 Jul 2014 Dong-A ST initiates enrolment in a phase I trial in patients with renal impairment in South Korea (NCT02214693)

Evogliptin Tartrate

…………………………………..

WO 2010114291

http://www.google.co.in/patents/WO2010114291A2?cl=en

Formula 1

Figure PCTKR2010001947-appb-C000001

Korea Patent Publication No. 2008-0094604 the call to the scheme, as indicated by A Ⅰ) of formula (II) beta-compound of formula 3 is already substituted heterocyclic compound having 1-hydroxy-benzotriazole group (HOBT) 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide (EDC) and reacting with a tertiary amine to prepare a compound of formula (4) connected by peptide bonds; Ⅱ) beta comprises the step of reacting under acidic conditions a compound of the formula (4) – a method of manufacturing the heterocyclic compounds of the formula I having an amino group is disclosed.

– Scheme A]

Figure PCTKR2010001947-appb-I000001

(Wherein, PG is a protecting group.)

In this case, the beta of the formula (2) of Scheme A – a compound having an amino group is prepared in addition to the DPP-IV inhibitor International Publication represented by Formula 1 WO03 / 000181, WO03 / 004498, WO03 / 082817, WO04 / 007468, WO04 / 032836, WO05 / 011581, WO06 / 097175, WO07 / 077508, WO07 / 063928, WO08 / 028662 WO08 / it may be used for the production of different DPP-IV inhibitors according 087,560 and can be prepared in a number of ways.

To, the compound of Formula 2 is an example as shown in Scheme J. Med.Chem. 2005; 141, and Synthesis 1997; it can be produced by the known method described in 873.

Figure PCTKR2010001947-appb-I000002

Specifically, (2S) – (+) – 2,5- dihydro-3,6-dimethoxy-2-isopropyl-pyrazine 2,4,5-trifluoro-react with benzyl bromide and acid treatment, and then the amine an ester compound obtained by the protection reaction. Ester compounds are hydrolyzed to re-3- (2,4,5-trifluoro-phenyl) -2-amino-propionic acid tert such as isobutyl chloroformate, triethylamine or diisopropylethylamine to give the amine, and then using diazomethane to form a diazo ketone, and then may be prepared by reaction with silver benzoate. However, the reaction can be performed at low temperature (-78 ℃) or high alpha-amino acid to purchase and use, and may have a risk of problems such as the need to use large diazomethane.

To a different process for preparing a compound of Formula 2 as shown in scheme Tetrahedron: Asymmetry 2006; It is known in 2622; 205 or similarly Bioorganic & Medicinal Chemistry Letters 2007.

Figure PCTKR2010001947-appb-I000003

That is, a 1,1′-carbonyl-2,4,5 which the phenyl trifluoroacetic acid activated using the following imidazole mono-methyl words potassium carbonate is reacted with the beta-keto ester compound is prepared. This produced an enamine ester using ammonium acetate and ammonium solution, the ester compound chloro (1,5-cyclooctadiene) rhodium (I) dimer using a chiral ferrocenyl ligands I the reaction of the high-pressure hydrogen with a chiral primary amine with a beta-amino ester compound after production and can lead to hydrolysis to prepare a compound of formula (2). However, use of expensive metal catalyst has a problem that must be performed in high pressure hydrogenation.

The method for preparing a compound of Formula 2 is disclosed in International Publication No. WO 04/87650.

Figure PCTKR2010001947-appb-I000004

Specifically, 2,4,5-fluorophenyl reagent is oxalyl chloride, the acid activated acid with 2,2-dimethyl-1,3-dioxane-4,6-dione, and after the reaction of methanol and the resulting material at reflux to prepare a corresponding compound. With a selective reducing reagents which enantiomers (S) -BINAP-RuCl 2 and hydrogen through a reaction (S) – producing a compound having coordinated to each other, it again after the decomposition, and the singer O- benzyl hydroxyl amine and the coupling reaction and the intermediate is prepared. To do this, the resulting intermediate tree azodicarboxylate and diisopropyl azodicarboxylate presence ring condensation reaction, treated with an aqueous solution of lithium hydroxide to (R) – while having the formula (II) coordinated to the amine group protected with a benzyl-O- the compound can be produced. However, the method has a problem as a whole to be prepared by the reaction yield to be low and a long processing time to perform the reaction.

Thus, the conventional known method for producing a compound of the general formula (2) has the disadvantage of using expensive reagents, or not suitable for commercial mass-production method by a long synthesis time yield is also low.

In addition, the compound represented by General Formula (3), as described in Korea Patent Publication No. 2008-0094604 call, can be prepared by way of reaction schemes.

Figure PCTKR2010001947-appb-I000005

Specifically, the starting material D- serine methyl ester is substituted by a hydroxy group when reflux again substituted by trityl chloride as methoxy groups converted to the aziridine compound.

[Scheme 3]

Figure PCTKR2010001947-appb-I000008

<Example 3> (R)-4-[(R)-3-아미노-4-(2,4,5-트리플루오로페닐)부타노일]-3-(t-부톡시메틸)피페라진-2-온(화학식 1) Preparation of the hydrochloride

Step 1: t- butyl (R)-4-[(R)-2-(t-부톡시메틸)-3-옥소피페라진-1-일]-4-옥소 – 1-(2,4,5-트리플루오로페닐)부탄-2-일카르바메이트(화학식 Preparation of 4)

2 L flask, prepared in Example 1 (R) -3-t- butoxycarbonyl-4- (2,4,5-trifluoro-phenyl) butanoate acid (Formula 2) 10.0 g of toluene was dissolved in 450 mL of bis (2,2′-benzothiazolyl) disulfide 13.0 g, was cooled and then 10.2 g triphenylphosphine was added to the reaction solution at 0 ℃. While stirring the reaction mixture was added to a solution of 0.8 mL of triethylamine in 20 mL of toluene was stirred at room temperature for 5 hours. The reaction mixture was cooled to 0 ℃ and prepared in Example 2 (R) -3- (t- butoxymethyl) piperazin-2-one (Formula 3) was dissolved in 5.6 g of toluene and 40 mL pyridine a 2.4 mL was added slowly. After 30 minutes the reaction mixture was heated to room temperature and stirred for 1 hour. Saturated sheet to be the aqueous acid solution to a pH of 2.5 and then diluted with ethyl acetate 400 mL. Washed twice with brine and the organic layer was dehydrated with magnesium sulfate and concentrated. The residue was purified by column chromatography to give the title compound 838 mg.

1 H NMR (400 MHz, CDCl 3) δ 7.03 (m, 1H), 6.88 (m, 1H), 5.97 (m, 1H), 5.48 (m, 1H), 4.16 ~ 4.07 (m, 1H), 4.02 ~ 3.91 (m, 1H), 3.74 (m, 2H) 3.37 (m, 2H), 3.24 (m, 1H), 2.92 (m, 2H), 2.80 (m, 1H), 2.59 (m, 2H), 1.34 ( d, 9H), 1.13 (s, 9H)

Step 2: (R) -4 – [(R) -3- amino-4- (2,4,5-trifluoro-phenyl) butane five days] -3- (t- butoxymethyl) piperazin-2- on the production of (I) hydrochloride

Prepared in Step 1 t- butyl (R)-4-[(R)-2-(t-부톡시메틸)-3-옥소피페라진-1-일]-4-옥소-1-(2,4,5-트리플루오로페닐)부탄-2-일카르바메이트 97 mg was dissolved in methanol was added 3 mL 2N- hydrochloric acid / diethyl ether 2 mL was stirred at room temperature for 3 hours. The reaction mixture was concentrated and dried under reduced pressure to give 64 mg of the title compound as a foaming solid.

1 H NMR (400 MHz, CD 3 OD) δ 7.37 (m, 1H), 7.23 (m, 1H), 4.80 (m, 1H), 4.59 ~ 4.40 (m, 1H), 3.93 (m, 1H), 3.90 ~ 3.83 (m, 2H), 3.70 (m, 1H), 3.38 (m, 2H), 3.27 (m, 1H), 3.07 (m, 2H), 2.89 ~ 2.66 (m, 2H), 1.18 (s, 3H ), 1.11 (s, 6H)

Mass (M + 1): 402

<Example 4> (R)-4-[(R)-3-아미노-4-(2,4,5-트리플루오로페닐)부타노일]-3-(t-부톡시메틸)피페라진-2-온(화학식 1) tartaric acid salts

Step 1: (R) -4 – [(R) -3- amino-4- (2,4,5-trifluoro-phenyl) butane five days] -3- (t- butoxymethyl) piperazin-2- Preparation of one (I)

Example 3 to give a compound of formula I in hydrochloride 60 mg 5% sodium hydrogen carbonate in dichloromethane was added to 10 mL of an aqueous solution / 2-propanol (4/1 (v / v)) was added to the mixed solution and extracted two times 10 mL The organic layer was dried under reduced pressure to give 55 mg of the title compound as a solid.

1 H NMR (400 MHz, CD 3 OD) δ 7.27 (m, 1H), 7.14 (m, 1H), 4.56 ~ 4.39 (m, 1H), 3.96 ~ 3.81 (m, 3H), 3.70 (m, 1H) , 3.46 (m, 1H), 3.43 ~ 3.32 (m, 1H), 2.83 ~ 2.65 (m, 3H), 2.58 ~ 2.40 (m, 2H), 1.16 (s, 3H), 1.11 (s, 6H)

Mass (M + 1): 402

Step 2: (R) -4 – [(R) -3- amino-4- (2,4,5-trifluorophenyl) butanoyl] -3- (t- butoxymethyl) piperazin-2- one (I) tartaric acid salt [

Was dissolved 55 mg of the compound of step 1 in 0.56 mL of acetone, L- tartrate 26 mg ethanol / water (9/1 (v / v)) was added slowly to a solution of 0.35 mL was stirred for 30 minutes. Here was added 0.56 mL of 2-propanol was stirred for 10 minutes and re-filtered to give 77 mg of the title compound as a solid.

1 H NMR (400 MHz, CD 3 OD) δ 7.38 (m, 1H), 7.22 (m, 1H), 4.80 (m, 1H), 4.59 ~ 4.40 (m, 1H), 4.40 (s, 2H), 3.93 (m, 1H), 3.90 ~ 3.83 (m, 2H), 3.70 (m, 1H), 3.38 (m, 2H), 3.27 (m, 1H), 3.07 (m, 2H), 2.89 ~ 2.66 (m, 2H ), 1.15 (s, 3H), 1.11 (s, 6H)

Mass (M + 1): 402

………………………………

WO 2010114292

http://www.google.com/patents/WO2010114292A2?cl=en

…………………………………

Discovery of DA-1229: a potent, long acting dipeptidyl peptidase-4 inhibitor for the treatment of type 2 diabetes
Bioorg Med Chem Lett 2011, 21(12): 3809

http://www.sciencedirect.com/science/article/pii/S0960894X11004859

Full-size image (3 K)

A series of β-amino amide containing substituted piperazine-2-one derivatives was synthesized and evaluated as inhibitors of dipeptidyl pepdidase-4 (DPP-4) for the treatment of type 2 diabetes. As results of intensive SAR study of the series, (R)-4-[(R)-3-amino-4-(2,4,5-trifluorophenyl)-butanoyl]-3-(t-butoxymethyl)-piperazin-2-one (DA-1229) displayed potent DPP-4 inhibition pattern in several animal models, was selected for clinical development.

About evogliptin tartrate tablets
Evogliptin tartrate tablets is a dipeptidyl peptidase IV inhibitor, in tablet form. Evogliptin tartrate
tablets is expected to be approved for the treatment of type 2 diabetes mellitus. The Group holds
an exclusive intellectual property licence from Dong-A Pharmaceutical Co. Ltd. to develop
and commercialise evogliptin tartrate tablets in China, including the exclusive right to develop
evogliptin tartrate tablets for manufacturing and sale in the Group’s name. The new drug certificate
to be issued by the CFDA will be approved and registered under the Group’s name.
Evogliptin is a patented new molecular entity in the United States and other international markets.
Evogliptin tartrate tablets is being concurrently developed by Dong-A Pharmaceutical Co. Ltd.
for the Korean market. Based on information released from a multi-centre, phase II, randomised,
double-blind, placebo-controlled, therapeutic exploratory clinical trial conducted in Korea by
Dong-A Pharmaceutical Co. Ltd. to investigate the efficacy and safety of evogliptin, evogliptin
was proven to be effective in significantly lowering blood glucose levels in patients with type
2 diabetes. Data also show that the body weights of patients remain stable over the treatment
period. In addition, evogliptin was proven to be safe and well tolerated with no severe adverse
drug reactions observed during those phase II clinical trials. The Company believes evogliptin
tartrate tablets will help reduce the burden of patients with moderate-to-severe renal impairment
as pharmacokinetic study in animal model and healthy human volunteers showed low renal
elimination.
2
According to the statistics of IMS Health Incorporated, the market size of products for the
treatment of diabetes in China in 2013 was approximately RMB7.8 billion, and grew at a
compound annual growth rate of 23.4% from 2011 to 2013.

 http://www.luye.cn/en/uploads//2014-07/21/_1405936452_zr21xh.pdf

Dong-A ST
SEOUL, SOUTH KOREA
14 April 2015 – 5:45pm
Oh Seung-mock

Dong-A ST has licensed its new diabetes drug Evogliptin to 17 Latin American countries including Mexico, Venezuela, Argentina, Chile, Colombia, Ecuador, Peru, the Dominican Republic, and Uruguay, Jung Jae-wook, Dong-A ST’s PR manager, told Business Korea.

Dong-A ST and Eurofarma, a Brazilian pharmaceutical company, concluded the licensing contract at Dong-A ST’s headquarters on April 13 in Seoul.

Eurofarma will be responsible for Evogliptin’s product development and sales in the 17 Latin American countries, Dong-A ST said. Dong-A ST will receive royalties from Eurofarma, and export the raw material of the medicine.

Dong-A ST has been developing Evogliptin with the support of the Ministry of Health & Welfare of South Korea as an innovative new medicine research project since May 2008. Evogliptin is a DPP-4 remedy based on the inhibition mechanism which is “excellent” at reducing blood sugar, whilst “less likely” to cause weight increases and hypoglycemia, the company said.

Park Chan-il, president of Dong-A ST, said that Dong-A ST will pursue further out-licensing “over the globe,” through continuous investment in research and development.

Maurizio Billi, Eurofarma’s president, wished to expand both companies’ partnership in the innovative new remedy development sector, according to Dong-A ST.

Last July, Dong-A ST and Eurofarma concluded a contract out-licensing Evogliptin to Brazil itself, the company said.

– See more at: http://www.businesskorea.co.kr/article/10115/southern-strategy-dong-st-licenses-new-diabetes-drug-evogliptin-17-latin-american#sthash.liqwFTWU.dpuf

//////////

see gliptins at…..http://drugsynthesisint.blogspot.in/p/gliptin-series.html

Dong-A Pharm. Co., Ltd, Yongin-si, Gyeonggi-do, Republic of Korea.

MASITINIB


Masitinib

Masitinib; 790299-79-5; Masivet; AB1010; AB-1010;

CLASS:Immunomodulator
TARGET:KIT (a stem cell factor, also called c-KIT) receptor as well as select other tyrosine kinases
STATUS FOR MS:Phase III
COMMERCIAL:Under development by AB Science..Ab Science
4-((4-Methylpiperazin-1-yl)methyl)-N-(4-methyl-3-((4-(pyridin-3-yl)-1,3-thiazol-2-yl)amino)phenyl)benzamide
AB 1010
UNII-M59NC4E26P

4-((4-Methylpiperazin-1-yl)methyl)-N-(4-methyl-3-((4-(pyridin-3-yl)-1,3-thiazol-2-yl)amino)phenyl)benzamide

Regulatory and Commercial Status

STATUS FOR MS:Phase III
HIGHEST STATUS ACHIEVED (FOR ANY CONDITION):
Marketing Authorization Application for the treatment of pancreatic cancer has been filed with the European Medicines Agency (16 October 2012)
Marketing Authorization Application for the conditional approval in the treatment of pancreatic cancer has been accepted by the European Medicines Agency (30 October 2012)

Masitinib.png

Masitinib is a tyrosine-kinase inhibitor used in the treatment of mast cell tumors in animals, specifically dogs.[1][2] Since its introduction in November 2008 it has been distributed under the commercial name Masivet. It has been available in Europe since the second part of 2009. In the USA it is distributed under the name Kinavet and has been available for veterinaries since 2011.

Masitinib is being studied for several human conditions including cancers. It is used in Europe to fight orphan diseases.[3]

Mechanism of action

Masitinib inhibits the receptor tyrosine kinase c-Kit which is displayed by various types of tumour.[2] It also inhibits the platelet derived growth factor receptor (PDGFR) and fibroblast growth factor receptor (FGFR).

……………………..

http://www.google.com/patents/US7423055

Compound Synthesis

General: All chemicals used were commercial reagent grade products. Dimethylformamide (DMF), methanol (MeOH) were of anhydrous commercial grade and were used without further purification. Dichloromethane and tetrahydrofuran (THF) were freshly distilled under a stream of argon before use. The progress of the reactions was monitored by thin layer chromatography using precoated silica gel 60F 254, Fluka TLC plates, which were visualized under UV light. Multiplicities in 1H NMR spectra are indicated as singlet (s), broad singlet (br s), doublet (d), triplet (t), quadruplet (q), and multiplet (m) and the NMR spectrum were realized on a 300 MHz Bruker spectrometer.

3-Bromoacetyl-pyridine, HBr Salt

Dibromine (17.2 g, 108 mmol) was added dropwise to a cold (0° C.) solution of 3-acetyl-pyridine (12 g, 99 mmol) in acetic acid containing 33% of HBr (165 mL) under vigourous stirring. The vigorously stirred mixture was warmed to 40° C. for 2 h and then to 75° C. After 2 h at 75° C., the mixture was cooled and diluted with ether (400 mL) to precipitate the product, which was recovered by filtration and washed with ether and acetone to give white crystals (100%). This material may be recrystallised from methanol and ether.

IR (neat): 3108, 2047, 2982, 2559, 1709, 1603, 1221, 1035, 798 cm−1−1H NMR (DMSO-d6) δ=5.09 (s, 2H, CH2Br); 7.88 (m, 1H, pyridyl-H); 8.63 (m, 1H, pyridyl-H); 8.96 (m, 1H, pyridyl-H); 9.29 (m, 1H, pyridyl-H).

Methyl-[4-(1-N-methyl-piperazino)-methyl]-benzoate

To methyl-4-formyl benzoate (4.92 g, 30 mmol) and N-methyl-piperazine (3.6 mL, 32 mmol) in acetonitrile (100 mL) was added dropwise 2.5 mL of trifluoroacetic acid. The reaction mixture was stirred at room temperature for 1 h. After slow addition of sodium cyanoborohydride (2 g, 32 mmol), the solution was left stirring overnight at room temperature. Water (10 mL) was then added to the mixture, which was further acidified with 1N HCl to pH=6-7. The acetonitrile was removed under reduced pressure and the residual aqueous solution was extracted with diethyl ether (4×30 mL). These extracts were discarded. The aqueous phase was then basified (pH>12) by addition of 2.5N aqueous sodium hydroxyde solution. The crude product was extracted with ethyl acetate (4×30 mL). The combined organic layers were dried over MgSO4 and concentrated under reduced pressure to afford a slightly yellow oil which became colorless after purification by Kugelrohr distillation (190° C.) in 68% yield.

IR(neat): 3322, 2944, 2802, 1721, 1612, 1457, 1281, 1122, 1012—1H NMR(CDCl3) δ=2.27 (s, 3H, NCH3); 2.44 (m, 8H, 2×NCH2CH2N); 3.53 (s, 2H, ArCH2N); 3.88 (s, 3H, OCH3); 7.40 (d, 2H, J=8.3 Hz, 2×ArH); 7.91 (d, 2H, J=8.3 Hz, 2×ArH)—3C NMR (CDCl3) δ=45.8 (NCH3); 51.8 (OCH3); 52.9 (2×CH2N); 54.9 (2×CH2N); 62.4 (ArCH2N); 128.7 (2×ArC); 129.3 (2×ArC); 143.7 (ArC); 166.7 (ArCO2CH3)-MS CI (m/z) (%) 249 (M+1, 100%).

2-Methyl-5-tert-butoxycarbonylamino-aniline

A solution of di-tert-butyldicarbonate (70 g, 320 mmol) in methanol (200 mL) was added over 2 h to a cold (−10° C.) solution of 2,4-diaminotoluene (30 g, 245 mmol) and triethylamine (30 mL) in methanol (15 mL). The reaction was followed by thin layer chromatography (hexane/ethyl acetate, 3:1) and stopped after 4 h by adding 50 mL of water. The mixture was concentrated in vacuo and the residue was dissolved in 500 mL of ethyl acetate. This organic phase was washed with water (1×150 mL) and brine (2×150 mL), dried over MgSO4, and concentrated under reduced pressure. The resulting light brown solid was washed with small amounts of diethyl ether to give off-white crystals of 2-methyl-5-tert-butoxycarbonylamino-aniline in 67% yield.

IR (neat): 3359; 3246; 2970; 1719; 1609; 1557; 1173; 1050 cm−11H NMR (CDCl3): δ=1.50 (s, 9H, tBu); 2.10 (s, 3H, ArCH3); 3.61 (br s, 2H, NH2); 6.36 (br s, 1H, NH); 6.51 (dd, 1H, J=7.9 Hz, 2.3 Hz, ArH); 6.92 (d, 1H, J=7.9 Hz, ArH); 6.95 (s, 1H, ArH)—13C NMR (CDCl3) δ=16.6 (ArCH3); 28.3 (C(CH3)3); 80.0 (C(CH3)3); 105.2 (ArC); 108.6 (ArC); 116.9 (ArC); 130.4 (ArC—CH3); 137.2 (ArC—NH); 145.0 (ArC—NH2); 152.8 (COOtBu) MS ESI (m/z) (%): 223 (M+1), 167 (55, 100%).

N-(2-methyl-5-tert-butoxycarbonylamino)phenyl-thiourea

Benzoyl chloride (5.64 g, 80 mmol) was added dropwise to a well-stirred solution of ammonium thiocyanate (3.54 g, 88 mmol) in acetone (50 mL). The mixture was refluxed for 15 min, then, the hydrobromide salt of 2-methyl-5-tert-butoxycarbonylamino-aniline (8.4 g, 80 mmol) was added slowly portionswise. After 1 h, the reaction mixture was poured into ice-water (350 mL) and the bright yellow precipitate was isolated by filtration. This crude solid was then refluxed for 45 min in 70 mL of 2.5 N sodium hydroxide solution. The mixture was cooled down and basified with ammonium hydroxide. The precipitate of crude thiourea was recovered by filtration and dissolved in 150 mL of ethyl acetate. The organic phase was washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The residue was purified by column chromatography (hexane/ethyl acetate, 1:1) to afford 63% of N-(2-methyl-5-tert-butoxycarbonylamino)phenyl-thiourea as a white solid.

IR (neat): 3437, 3292, 3175, 2983, 1724, 1616, 1522, 1161, 1053 cm−1— 1H NMR (DMSO-d6) δ=1.46 (s, 9H, tBu); 2.10 (s, 3H, ArCH3); 3.60 (br s, 2H, NH2); 7.10 (d, 1H, J=8.29 Hz, ArH); 7.25 (d, 1H, J=2.23 Hz, ArH); 7.28 (d, 1H, J=2.63 Hz, ArH); 9.20 (s, 1H, ArNH); 9.31 (s, 1H, ArNH)—13C NMR (DMSO-d6) δ=25.1 (ArCH3); 28.1 (C(CH3)3); 78.9 (C(CH3)3); 16.6 (ArC); 117.5 (ArC); 128.0 (ArC); 130.4 (ArC—CH3); 136.5 (ArC—NH); 137.9 (ArC—NH); 152.7 (COOtBu); 181.4 (C═S)—MS CI(m/z): 282 (M+1, 100%); 248 (33); 226 (55); 182 (99); 148 (133); 93 (188).

2-(2-methyl-5-tert-butoxycarbonylamino)phenyl-4-(3-pyridyl)-thiazole

A mixture of 3-bromoacetyl-pyridine, HBr salt (0.81 g, 2.85 mmol), N-(2-methyl-5-tert-butoxycarbonylamino)phenyl-thiourea (0.8 g, 2.85 mmol) and KHCO3 (˜0.4 g) in ethanol (40 mL) was heated at 75° C. for 20 h. The mixture was cooled, filtered (removal of KHCO3) and evaporated under reduced pressure. The residue was dissolved in CHCl3 (40 mL) and washed with saturated aqueous sodium hydrogen carbonate solution and with water. The organic layer was dried over Na2SO4 and concentrated. Colum chromatographic purification of the residue (hexane/ethyl acetate, 1:1) gave the desired thiazole in 70% yield as an orange solid

IR(neat): 3380, 2985, 2942, 1748, 1447, 1374, 1239, 1047, 938—1H NMR (CDCl3) δ=1.53 (s, 9H, tBu); 2.28 (s, 3H, ArCH3); 6.65 (s, 1H, thiazole-H); 6.89 (s, 1H); 6.99 (dd, 1H, J=8.3 Hz, 2.3 Hz); 7.12 (d, 2H, J=8.3 Hz); 7.35 (dd, 1H, J=2.6 Hz, 4.9 Hz); 8.03 (s, 1H); 8.19 (dt, 1H, J=1.9 Hz, 7.9 Hz); 8.54 (br s, 1H, NH); 9.09 (s, 1H, NH)—13C NMR (CDCl3) δ=18.02 (ArCH3); 29.2 (C(CH3)3); 81.3 (C(CH3)3); 104.2 (thiazole-C); 111.6; 115.2; 123.9; 124.3; 131.4; 132.1; 134.4; 139.5; 148.2; 149.1; 149.3; 153.6; 167.3 (C═O)—MS Cl (m/z) (%): 383 (M+1, 100%); 339 (43); 327 (55); 309 (73); 283 (99); 71 (311).

2-(2-methyl-5-amino)phenyl-4-(3-pyridyl)-thiazole

2-(2-methyl-5-tert-butoxycarbonylamino)phenyl-4-(3-pyridyl)-thiazole (0.40 g, 1.2 mmol) was dissolved in 10 mL of 20% TFA/CH2Cl2. The solution was stirred at rool temperature for 2 h, then it was evaporated under reduced pressure. The residue was dissolved in ethyl acetate. The organic layer was washed with aqueous 1N sodium hydroxide solution, dried over MgSO4, and concentrated to afford 2-(2-methyl-5-amino)phenyl-4-(3-pyridyl)-thiazole as a yellow-orange solid in 95% yield. This crude product was used directly in the next step.

A 2M solution of trimethyl aluminium in toluene (2.75 mL) was added dropwise to a cold (0° C.) solution of 2-(2-methyl-5-amino)phenyl-4-(3-pyridyl)-thiazole (0.42 g, 1.5 mmol) in anhydrous dichloromethane (10 mL) under argon atmosphere. The mixture was warmed to room temperature and stirred at room temperature for 30 min. A solution of methyl-4-(1-N-methyl-piperazino)-methyl benzoate (0.45 g, 1.8 mmol) in anhydrous dichloromethane (1 mL) and added slowly, and the resulting mixture was heated at reflux for 5 h. The mixture was cooled to 0° C. and quenched by dropwise addition of a 4N aqueous sodium hydroxide solution (3 mL). The mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×20 mL) and dried over anhydrous MgSO4. (2-(2-methyl-5-amino)phenyl-4-(3-pyridyl)-thiazole) is obtained in 72% after purification by column chromatography (dichloromethane/methanol, 3:1)

IR (neat): 3318, 2926, 1647, 1610, 1535, 1492, 1282, 1207, 1160, 1011, 843—

1H NMR (CDCl3) δ=2.31 (br s, 6H, ArCH3+NCH3); 2.50 (br s, 8H, 2×NCH2CH2N); 3.56 (s, 2H, ArCH2N); 6.89 (s, 1H, thiazoleH); 7.21-7.38 (m, 4H); 7.45 (m, 2H); 7.85 (d, 2H, J=8.3 Hz); 8.03 (s, 1H); 8.13 (s, 1H); 8.27 (s, 1H); 8.52 (br s, 1H); 9.09 (s, 1H, NH)—

13C NMR (CDCl3) δ 17.8 (ArCH3); 46.2 (NCH3); 53.3 (NCH2); 55.3 (NCH2); 62.8 (ArCH2N); 99.9 (thiazole-C); 112.5; 123.9; 125.2; 127.5; 129.6; 131.6; 133.7; 134.0; 137.6; 139.3; 142.9; 148.8; 149.1; 166.2 (C═O); 166.7 (thiazoleC-NH)—

MS CI (m/z) (%): 499 (M+H, 100%); 455 (43); 430 (68); 401 (97); 374 (124); 309 (189); 283 (215); 235 (263); 121 (377); 99 (399).

………………………

 

http://www.google.com/patents/WO2012136732A1?cl=en

In a preferred embodiment of the above-depicted treatment, the active ingredient masitinib is administered in the form of masitinib mesilate; which is the orally bioavailable mesylate salt of masitinib – CAS 1048007-93-7 (MsOH); C28H30N6OS.CH3SO3H; MW 594.76:

Figure imgf000031_0001

 

http://www.google.com/patents/WO2004014903A1?cl=en

Figure imgf000021_0001

003 : 4-(4-Methyl-piperazin-l-ylmethyl)-N-[3-(4-pyridin-3-yl-thiazol-2-ylamino)- phenyl] -benzamide

4-(4-Methyl-piperazin-l-yl)-N-[4-methyl-3-(4-pyridin-3-yl-thiazol-2-ylmethyl)- phenyl] -benzamide

Figure imgf000053_0001

beige brown powder mp : 128-130°C

1H RMN (DMSO-d6) δ = 2.15 (s, 3H) ; 2.18 (s, 3H) ; 2.35-2.41 (m, 4H) ; 3.18-3.3.24 (m, 4H) ; 6.94 (d, J = 8.9 Hz, 2H) ; 7.09 (d, J = 8.4 Hz, IH) ; 7.28-7.38 (m, 3H) ; 7.81 (d, J = 8.9 Hz, 2H) ; 8.20-8.25 (m, IH) ; 8.40 (dd, J = 1.6 Hz, J = 4.7 , IH) ; 8.48 (d, J = 1.9 Hz, IH) ; 9.07 (d, J = 1.5 Hz, IH) ; 9.35 (s, IH) ; 9.84 (s, IH)

……………

http://www.google.com/patents/WO2008098949A2?cl=en

EXAMPLE 4 N- [4-Methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino)-phenyl] -benzamide derivatives

Method A In a reactor and under low nitrogen pressure, add 4-Methyl-N3-(4-pyridin-3-yl-thiazol- 2-yl)-benzene-l,3-diamine (95 g, 336.45 mmol), dichloromethane (2 L). To this suspension cooled to temperature of 5°C was added dropwise 2M/n-hexane solution of trimethylaluminium (588 mL). The reaction mixture was brought progressively to 15°C, and maintained for 2 h under stirring. 4-(4-Methyl-piperazin-l-ylmethyl)-benzoic acid methyl ester (100 g, 402.71 mmol) in dichloromethane (200 mL) was added for 10 minutes. After 1 h stirring at room temperature, the reaction mixture was heated to reflux for 20 h and cooled to room temperature. This solution was transferred dropwise via a cannula to a reactor containing 2N NaOH (2.1 L) cooled to 5°C. After stirring for 3 h at room temperature, the precipitate was filtered through Celite. The solution was extracted with dichloromethane and the organic layer was washed with water and saturated sodium chloride solution, dried over MgSO4 and concentrated under vacuum. The brown solid obtained was recrystallized from /-Pr2O to give 130.7 g (78%) of a beige powder.

Method B Preparation of the acid chloride

To a mixture of 4-(4-Methyl-piperazin-l-ylmethyl)-benzoic acid dihydrochloride (1.0 eq), dichloromethane (7 vol) and triethylamine (2.15 eq), thionyl chloride (1.2 eq) was added at 18-28°C . The reaction mixture was stirred at 28-32°C for 1 hour. Coupling of acid chloride with amino thiazole To a chilled (0-50C) suspension of 4-Methyl-N3-(4-pyridin-3-yl-thiazol-2-yl)-benzene- 1,3-diamine (0.8 eq) and thiethylamine (2.2 eq) in dichloromethane (3 vol), the acid chloride solution (prepared above) was maintaining the temperature below 5°C. The reaction mixture was warmed to 25-300C and stirred at the same temperature for 1O h. Methanol (2 vol) and water (5 vol) were added to the reaction mixture and stirred. After separating the layers, methanol (2 vol), dihloromethane (5 vol) and sodium hydroxide solution (aqueous, 10%, till pH was 9.5-10.0) were added to the aqueous layer and stirred for 10 minutes. The layers were separated. The organic layer was a washed with water and saturated sodium chloride solution. The organic layer was concentrated and ethanol (2 vol) was added and stirred. The mixture was concentrated. Ethanol was added to the residue and stirred. The product was filtered and dried at 50-550C in a vaccum tray drier. Yield = 65-75%.

Method C

To a solution of 4-methyl-N3-(4-pyridin-3-yl-thiazol-2-yl)-benzene-l,3-diamine (1.0 eq) in DMF (20 vol) were added successively triethylamine (5 eq), 2-chloro-l- methylpyridinium iodide (2 eq) and 4-(4-methyl-piperazin-l-ylmethyl)-benzoic acid (2 eq). The reaction mixture was stirred for 7 h at room temperature. Then, the mixture was diluted in diethyl ether and washed with water and saturated aqueous NaHCO3, dried over Na2SO4 and concentrated. The crude product was purified by column chromatography using an elution of 100% EtOAc to give a yellow solid.

Yield = 51%.

1H NMR (CDCl3) : δ = 9.09 (IH, s, NH); 8.52 (IH, br s); 8.27 (IH, s); 8.13 (IH, s);

8.03 (IH, s); 7.85 (2H, d, J= 8.3Hz); 7.45 (2H, m); 7.21-7.38 (4H, m); 6.89 (IH, s);

3.56 (2H, s); 2.50 (8H, br s); 2.31 (6H, br s).

MS (CI) m/z = 499 (M+H)+.

An additional aspect of the present invention relates to a particular polymorph of the methanesulfonic acid salt of N-[4-Methyl-3-(4-pyridin-3-yl-thiazol-2-ylamino)-phenyl]- benzamide of formula (IX).

Figure imgf000023_0001

(VI)

Hereinafter is described the polymorph form of (IX) which has the most advantageous properties concerning processability, storage and formulation. For example, this form remains, dry at 80% relative humidity and thermodynamically stable at temperatures below 2000C.

The polymorph of this form is characterized by an X-ray diffraction pattern illustrated in FIG.I, comprising characteristic peaks approximately 7.269, 9.120, 11.038, 13.704, 14.481, 15.483, 15.870, 16.718, 17.087, 17.473, 18.224, 19.248, 19.441, 19.940, 20.441, 21.469, 21.750, 22.111, 23.319, 23.763, 24.120, 24.681, 25.754, 26.777, 28.975, 29.609, 30.073 degrees θ, and is also characterized by differential scanning calorimetry (DSC) illustrated in FIG.II, which exhibit a single maximum value at approximately 237.49 ± 0.3 0C. X-ray diffraction pattern is measured using a Bruker AXS (D8 advance). Differential scanning calorimetry (DSC) is measured using a Perking Elmer Precisely (Diamond DSC).

This polymorph form can be obtained by treatement of 4-(4-Methyl-piperazin-l- ylmethyl)-N-[4-methyl-3-(4-pyridin-3-yl-thiazol-2-ylamino)-phenyl]-benzamide with 1.0 to 1.2 equivalent of methanesulfonic acid, at a suitable temperature, preferably between 20-800C.

The reaction is performed in a suitable solvent especially polar solvent such as methanol or ethanol, or ketone such as acetone, or ether such as diethylether or dioxane, or a mixture therof. This invention is explained in example given below which is provided by way of illustration only and therefore should not be construed to limit the scope of the invention. Preparation of the above-mentioned polymorph form of 4-(4-Methyl-piperazin-l- ylmethyl)-N- [4-methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino)-phenyl] -benzamide methanesulfonate .

4-(4-Methyl-piperazin- 1 -ylmethyl)-N- [4-methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino) phenyl] -benzamide (1.0 eq) was dissolved in ethanol (4.5 vol) at 65-700C. Methanesulfonic acid (1.0 eq) was added slowly at the same temperature. The mixture was cooled to 25-300C and maintained for 6 h. The product was filtered and dried in a vacuum tray drier at 55-600C. Yield = 85-90%. Starting melting point Smp = 236°C.

 

NMR PREDICT

CAS NO. 1048007-93-7, methanesulfonic acid,4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide H-NMR spectral analysis

methanesulfonic acid,4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide NMR spectra analysis, Chemical CAS NO. 1048007-93-7 NMR spectral analysis, methanesulfonic acid,4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide H-NMR spectrum

methanesulfonic acid,4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide NMR spectra analysis, Chemical CAS NO. 1048007-93-7 NMR spectral analysis, methanesulfonic acid,4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide C-NMR spectrum

CAS NO. 1048007-93-7, methanesulfonic acid,

4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-yl-1,3-thiazol-2-yl)amino]phenyl]benzamide C-NMR spectral analysisPREDICT

References

  1. Hahn, K.A.; Oglivie, G.; Rusk, T.; Devauchelle, P.; Leblanc, A.; Legendre, A.; Powers, B.; Leventhal, P.S.; Kinet, J.-P.; Palmerini, F.; Dubreuil, P.; Moussy, A.; Hermine, O. (2008). “Masitinib is Safe and Effective for the Treatment of Canine Mast Cell Tumors”. Journal of Veterinary Internal Medicine 22 (6): 1301–1309. doi:10.1111/j.1939-1676.2008.0190.x. ISSN 0891-6640.
  2. Information about Masivet at the European pharmacy agency website
  3. Orphan designation for Masitinib at the European pharmacy agency website
WO2004014903A1 Jul 31, 2003 Feb 19, 2004 Ab Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors
WO2008098949A2 Feb 13, 2008 Aug 21, 2008 Ab Science Process for the synthesis of 2-aminothiazole compounds as kinase inhibitors
EP1525200B1 Jul 31, 2003 Oct 10, 2007 AB Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors
US7423055 Aug 1, 2003 Sep 9, 2008 Ab Science 2-(3-Aminoaryl)amino-4-aryl-thiazoles for the treatment of diseases
US20080207572 * Jul 13, 2006 Aug 28, 2008 Ab Science Use of Dual C-Kit/Fgfr3 Inhibitors for Treating Multiple Myeloma
Masitinib.svg
Systematic (IUPAC) name
4-[(4-Methylpiperazin-1-yl)methyl]-N-(4-methyl-3-{[4-(pyridin-3-yl)-1,3-thiazol-2-yl]amino}phenyl)benzamide
Clinical data
Trade names Masivet, Kinavet
AHFS/Drugs.com International Drug Names
Identifiers
790299-79-5
L01XE22
PubChem CID 10074640
ChemSpider 8250179
ChEMBL CHEMBL1908391
Chemical data
Formula C28H30N6OS
498.64 g/mol
Patent Submitted Granted
2-(3-Aminoaryl)amino-4-aryl-thiazoles for the treatment of diseases [US7423055] 2004-06-10 2008-09-09
2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors [US2005239852] 2005-10-27
Use of C-Kit Inhibitors for Treating Fibrosis [US2007225293] 2007-09-27
Use of Mast Cells Inhibitors for Treating Patients Exposed to Chemical or Biological Weapons [US2007249628] 2007-10-25
Use of c-kit inhibitors for treating type II diabetes [US2007032521] 2007-02-08
Use of tyrosine kinase inhibitors for treating cerebral ischemia [US2007191267] 2007-08-16
Use of C-Kit Inhibitors for Treating Plasmodium Related Diseases [US2008004279] 2008-01-03
Tailored Treatment Suitable for Different Forms of Mastocytosis [US2008025916] 2008-01-31
2-(3-AMINOARYL) AMINO-4-ARYL-THIAZOLES AND THEIR USE AS C-KIT INHIBITORS [US2008255141] 2008-10-16
Use Of C-Kit Inhibitors For Treating Inflammatory Muscle Disorders Including Myositis And Muscular Dystrophy [US2008146585] 2008-06-19
Patent Submitted Granted
Aminothiazole compounds as kinase inhibitors and methods of using the same [US8940894] 2013-05-10 2015-01-27
Aminothiazole compounds as kinase inhibitors and methods of using the same [US8492545] 2012-03-08 2013-07-23
Patent Submitted Granted
Use of Dual C-Kit/Fgfr3 Inhibitors for Treating Multiple Myeloma [US2008207572] 2008-08-28
PROCESS FOR THE SYNTHESIS OF 2-AMINOTHIAZOLE COMPOUNDS AS KINASE INHIBITORS [US8153792] 2010-05-13 2012-04-10
COMBINATION TREATMENT OF SOLID CANCERS WITH ANTIMETABOLITES AND TYROSINE KINASE INHIBITORS [US8227470] 2010-04-15 2012-07-24
Anti-IGF antibodies [US8580254] 2008-06-19 2013-11-12
COMBINATIONS FOR THE TREATMENT OF B-CELL PROLIFERATIVE DISORDERS [US2009047243] 2008-07-17 2009-02-19
TREATMENTS OF B-CELL PROLIFERATIVE DISORDERS [US2009053168] 2008-07-17 2009-02-26
Anti-IGF antibodies [US8318159] 2009-12-11 2012-11-27
SURFACE TOPOGRAPHIES FOR NON-TOXIC BIOADHESION CONTROL [US2010226943] 2009-08-31 2010-09-09
EGFR/NEDD9/TGF-BETA INTERACTOME AND METHODS OF USE THEREOF FOR THE IDENTIFICATION OF AGENTS HAVING EFFICACY IN THE TREATMENT OF HYPERPROLIFERATIVE DISORDERS [US2010239656] 2010-05-10 2010-09-23
ANTI CD37 ANTIBODIES [US2010189722] 2008-08-08 2010-07-29
United States National Library of Medicine

Note: Compound name must be entered under “Substance Identification” and then “Names and Synonyms” selected to view synonyms.

Kocic I, Kowianski P, Rusiecka I, Lietzau G, Mansfield C, Moussy A, Hermine O, Dubreuil P
Naunyn Schmiedebergs Arch Pharmacol. 2014 Oct 26. Epub 2014 Oct 26. PMID: 25344204.Abstract
AB SCIENCE HEADQUARTER
3, Avenue George V
75008 PARIS – FRANCE
Tel. : +33 (0)1 47 20 00 14
Fax. : +33 (0)1 47 20 24 11

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

TAJIKISTAN

Tajikistan – Wikipedia, the free encyclopedia

en.wikipedia.org/wiki/Tajikistan

The territory that now constitutes Tajikistan was previously home to several ancient cultures, including the city of Sarazm of the Neolithic and the Bronze Age, …

Map of tajikistan country.
The nature of Tajikistan. Nurek
Tajikistan. Pamiro-Alay.Zeravshan mountain range. Guzn village. Local people
Dushanbe, Tajikistan
Women carry water canisters near Gargara village, 110km south of Tajikistan’s capital, Dushanbe
Ancient Buddhist ruins, Ajina Teppa, Tajikistan
///////////

S-flurbiprofen (TT-063)


(S)-flurbiprofen.png

Cas 51543-39-6,

MW 244.26,

MF C15 H13 F O2
[1,​1′-​Biphenyl]​-​4-​acetic acid, 2-​fluoro-​α-​methyl-​, (αS)​-
  • [1,1′-Biphenyl]-4-acetic acid, 2-fluoro-α-methyl-, (S)-
  • (+)-(S)-Flurbiprofen
  • (+)-Flurbiprofen
  • (2S)-2-(2-Fluoro-1,1′-biphenyl-4-yl)propanoic acid
  • (2S)-2-(2-Fluoro-4-biphenyl)propanoic acid
  • (S)-Flurbiprofen
  • Dexflurbiprofen
  • Esflurbiprofen
  • S-(+)-Flurbiprofen
  • d-Flurbiprofen

On October 20, 2014, Taisho filed for manufacturing and marketing approval for TT-063 from the Ministry of Health, Labour and Welfare as a new drug candidate that will follow the Type 2 diabetes treatment Lusefi®, which was launched in May 2014. TT-063 is a patch formulation that has been co-developed by Taisho and TOKUHON Corporation with the aim of obtaining an indication for osteoarthritis. In Phase 3 clinical trials comparing TT-063 with therapeutic drugs already on the market, TT-063 has been found to be more effective than the control drugs in patients with osteoarthritis of the knee joint (January 16, 2014 announcement ).

Furthermore, Taisho is also preparing to file for approval from the Ministry of Health, Labour and Welfare for CT-064, an oral formulation of the osteoporosis treatment agent Bonviva launched in August 2013. Taisho has confirmed the effectiveness of CT-064 for osteoporosis patients through Phase 3 clinical trials (September 22, 2014 announcement).


In the central nervous system field, TS-091 transitioned from Phase 1 to Phase 2 in Japan in May 2014. Clinical trials of TS-091 have commenced to confirm the effectiveness of this drug in patients with central disorders of hypersomnolence. In addition, Phase 1 clinical trials of TS-091 have commenced overseas. TS-111 and TS-121 are undergoing Phase 1 clinical trials overseas with the aim of obtaining an indication for depression.
Faced with intensifying competition in new drug discovery, we will jointly implement R&D activities with research institutions outside the Taisho Group, and with companies in Japan and overseas, as we work to enhance our drug development pipeline (lineup of drugs in development). Our goal is to discover many more new drugs, primarily in our priority fields.

Company Taisho Pharmaceutical Holdings Co. Ltd.
Description Topical anti-inflammatory analgesic patch containing S-flurbiprofen
Therapeutic Modality Small molecule
Latest Stage of Development Phase III
Standard Indication Osteoarthritis
Indication Details Treat osteoarthritis (OA) and scapulohumeral periarthritis
Regulatory Designation

Full-size image (93 K)

Scheme 2.

Reagents and conditions: (a) THF, EDC, Et3N; (b) TFA; (c) 0.5 equiv 2,5-dimethoxybenzoquinone, EtOH, 50–80 °C for 3–5 h; (d) 1 equiv naphthoquinone, MeOH, rt, overnight.

http://www.sciencedirect.com/science/article/pii/S0960894X13011773

……………………………………………

http://www.google.com/patents/EP2307335A1?cl=en

2-(6-methoxynaphthalen-2-yl) propanoic acid By way of illustration, chemically, flurbiprofen is 2-(2-fluoro-4-biphenylyl) propionic acid and is described in US Patent No. 3,755,427. NSAIDs, such as flurbiprofen, are usually supplied as a racemate. However, recently there has been renewed interest in the separate enantiomers of flurbiprofen, i.e. S-flurbiprofen and R-flurbiprofen.

Figure imgf000004_0001

R-Flurbιprofen

Figure imgf000004_0002

S-Flurtιprofen

Flurbiprofen is a potent inhibitor of cyclooxygenase (both COX-I and COX-2) in humans and it is understood that the inhibitory effect lies predominantly in the S- enantiomer.

Flurbiprofen is generally produced in the form of a racemic compound. It is known that from the racemic compound, flurbiprofen having a high optical purity can be produced by an optical resolution method using, for example, an optically active amine compound, such as α-phenylethylamine, as an optical resolution agent, as is described in US Patent No. 5,599,969. In addition, whether dealing with racemic, S- or R- 2-aryl propionic acid, there is also a need to make the synthetic process as efficient as possible.

Example 2 – Ibuprofen

Example 2.1 Resolution procedure

Racemic ibuprofen (530g) is dissolved in toluene (1335ml) and methanol (900ml).

The mixture is heated to dissolve the solid. S-1-Phenylethylamine (247g) is dissolved in toluene (200ml) and the solution is added with stirring at 600C over about 3 hours while the temperature is maintained at about 65-700C. The mixture is cooled gradually to 0 to 50C to induce crystallisation and stirred at this temperature for 1 hour. The crystals are filtered off, washed with toluene (600ml) and dried in a Vacuum oven at 550C to form crude S-ibuprofen / S-1-phenylethylamine salt (635g).

Crude S-ibuprofen / S-1-phenylethylamine salt (635g) is stirred with toluene (1930ml) and methanol (800ml) and the mixture is heated to 6O0C to dissolve the solid. The solution is cooled gradually to 0 to 5°C to induce crystallisation. The crystals are filtered off and dried in a vacuum oven at 55°C to form pure S-ibuprofen / S-I- phenylethylamine salt (510g). This recrystallisation of the S-ibuprofen / S-I- phenylethylamine salt may be repeated if necessary to upgrade the enantiomeric purity if required.

Pure S-ibuprofen / S-1-phenylethylamine salt (485g) is mixed with toluene (1700ml) with stirring. Water (300ml) and concentrated hydrochloric acid (17Og) are added and

÷ibe mixture is stirred at 600C. The lower aqueous layer is separated off and the upper organic layer is retained. The hydrochloric acid wash is repeated, then the toluene solution is washed with water. Water (370ml) and 47% sodium hydroxide

Figure imgf000023_0001

(118g) are added and the solution is heated to 600C and allowed to settle. The lower aqueous layer is separated and the upper toluene layer is washed with water. The aqueous phases are combined and heptane (420ml) is added. Hydrochloric acid

(130g) is added and the mixture is heated to 600C, stirred and settled. The organic layer is separated off and washed with water. The solution is cooled to -100C to induce crystallisation and the crystals are separated off by filtration, washed with heptane and dried under vacuum to yield (S)-ibuprofen (28Og) at an enantiomeric purity of over 99%.

Example 2.2 Racemisation procedure

Toluene/methanol mother liquors from the filtration of crude S-ibuprofen / S-I- phenylethylamine salt in the resolution procedure (2400ml, containing an estimated 130g of ibuprofen) is charged into a 3 L 3 necked round bottomed flask and methanol and toluene are distilled out at atmospheric pressure (volume removed approximately 1400 ml). The batch is then cooled to around 60°C and washed twice with hydrochloric acid (20 ml concentrated hydrochloric acid in 200 ml of water), and then twice with water (200 ml). Toluene is charged (80 ml) followed by methanol (200 ml) and caustic soda solution (45Og of 28% w/w solution, 5 molar equivalents). The mixture is heated to reflux for about 6 hours. Solvent is then removed at atmospheric pressure until the vapour temperature reaches approximately 85°C. The mixture is cooled to around 60°C and concentrated hydrochloric acid is charged at about 60 to 70°C until the pH of the mixture is 1 or less. The layers are allowed to separate and the bottom aqueous layer removed. The organic layer is washed with water (200 ml) and then azeotroped to dryness using a Dean and Stark trap. A solution of racemic ibuprofen in toluene remains.

…………………………………………

PATENT

 http://www.google.com/patents/CN104478703A?cl=en

Preparation of R – (+) _ flurbiprofen:

 The racemic flurbiprofen as a starting material, to obtain an intermediate product of formula I as shown and then the ester prepared as shown in Formula II with 5-isosorbide monobenzyl ether, ester hydrolysis after obtained R – (+) – flurbiprofen;

Figure CN104478703AD00061

wherein, in formula I, X is Cl or Br;

(2) by the R – (+) _ flurbiprofen obtained (RS) – flurbiprofen:

 The R _ (+) _ flurbiprofen 200mg, potassium hydroxide 150mg, 0. 5mL water into IOmL reaction flask and heated to 120 ° C and held for 2h, then water was added 15mL, cooled to room temperature, the resulting stirring the mixed solution with 10% hydrochloric acid to pH = 0. 5, extracted with ethyl acetate, combined several layers, washed with water until neutral, the organic solvent is recovered, the resulting residue was added at 60~90 ° C under an appropriate amount of petroleum ether by recrystallization, obtained (RS) – flurbiprofen 100mg, 50% yield.

 (3) Preparation of (S) -⑴- flurbiprofen:

 In 25mL single-necked flask, followed by adding (RS) – flurbiprofen 123mg, Portugal TOA 29. 8mg, isopropanol lmL, the mixture was stirred at reflux until clear, half the amount of the solvent evaporated under reduced pressure except , set the refrigerator overnight. The precipitate was collected by suction filtration as white crystals, after washing a small amount of isopropanol, which was dissolved in water, washed with 10% aqueous sodium hydroxide (10% NaOH mean mass fraction) adjusted pH = 13, the sheet-like precipitate was filtered off Portuguese octylamine white crystals. The resulting filtrate was added dropwise with stirring 10% hydrochloric acid to pH = 1, extracted with ethyl acetate, the organic layer was washed with water to recover the solvent, the resulting residue was purified by an appropriate amount of petroleum ether and recrystallized at 60~90 ° C. The product was collected by filtration, and dried in vacuo to give a white (S) – (+) _ flurbiprofen needle crystal 45. 3mg, 65% yield, mp 102~103 ° C, [α] = + 44 ° (C = 1, methanol), ee value of 92.6% (ee value measurement method: (S) – (+) – flurbiprofen after chiral amine derivatization reagents, by HPLC analysis).

wherein in step (3) is a byproduct eleven R _ (+) _ flurbiprofen, its follow step (1) of racemic reused.

 Step (1) of the specific operation is as follows:

 (la) 1:. Synthesis of 2,6-sorbitol dehydration -D- -5- benzyl ether: 4: 3

 250ml volumetric flask isosorbide 18. 25g (125mmol), lithium hydroxide monohydrate 5. 25g (125mmol) and 60ml of dimethyl sulfoxide (DMSO), heated to 90 ° C, stirred for 30min, constant pressure equalizing dropping funnel was added dropwise benzyl chloride 14. 4ml (125mmol), 90 ° C the reaction 19-20h, reaction mixture was adjusted to pH 1 with 2M hydrochloric acid, extracted with ethyl acetate (50ml * 3), the organic layers combined, washed with water ( 30ml * 2), dried over anhydrous sodium sulfate overnight, filtered and concentrated residue Cheng baby gel column chromatography (petroleum ether: ethyl acetate = 5: 1) to give a cream solid, that is 1: 4: 3: 2,6 Dehydration -D- sorbitol -5- benzyl ether 24. 5g, m.p. 59 ~61 ° C.

 (Ib) · 2- (2- fluoro-4-biphenylyl) propionyl chloride Synthesis

 50ml vial before racemic flurbiprofen was added 2. 44g (IOmmol), anhydrous toluene 20ml, freshly distilled thionyl chloride was added dropwise 0. 8ml (Ilmmol), N, N- dimethylformamide amide (DMF) 2 dropwise, stirred at room temperature 2h, the solvent was distilled off under reduced pressure to give a pale yellow gum, i.e., 2- (2-fluoro-4-biphenylyl) propionyl chloride, it was used directly in the reaction without isolation.

 (lc). R-2- (2- fluoro-4-biphenylyl) propionic acid 5- isosorbide monobenzyl ether ester synthesis

 The (Ib) resulting acid chloride was dissolved in 20ml of dry toluene was added dropwise at room temperature, dimethyl amine 3. 5ml, solid precipitation, stirred for about Ih, ice salt bath, a bath temperature of minus 10-15Ό, stirred at this temperature IOmin so, and then the constant pressure dropping funnel (Ia) 5 isosorbide monobenzyl ether (2. 83g, 12mmol) in toluene, keeping the reaction temperature, stirring 8h. The ice bath was removed and the reaction mixture under reduced pressure to remove the solvent, the residue was extracted with ethyl acetate. The extract was washed with water, dried over anhydrous sodium sulfate overnight, ethyl acetate was removed under reduced pressure, the residue was a white gel, recrystallized from petroleum ether to give a white solid that R-2- (2- fluoro-4-biphenylyl) propionic acid 5- isosorbide monobenzyl ether ester 3. 65g (7. 88mmol), in order to put the racemic flurbiprofen yield based on 78.8%.

(ld) R – Synthesis of flurbiprofen – (+)

 Under ice bath (Ic) obtained R-2- (2- fluoro-4-biphenylyl) propionic acid monobenzyl ether isosorbide 5- ester 2. 3Ig (5mmol) was dissolved in 20ml of acetone / water (1/1) was added Iml hydrochloric acid to adjust pH to 3, stirred for 3-4h, the reaction solution was extracted with ethyl acetate (20ml * 2), sash organic layer was washed with ice (10ml * 2), dried over anhydrous sodium sulfate overnight , filtration, and the filtrate was concentrated, the residue was recrystallized from ether to give white crystals, i.e. L-flurbiprofen 1.02g (4 18mmol.), yield 83.5%, optical purity 93% (HPLC method); input-racemic flurbiprofen dollars, the total yield of 78.8% * 83.5% = 65.8%.

 Step (1) reaction of the formula:

Figure CN104478703AD00071
FLURBIPROFEN RACEMIC
3-Fluoro-4-phenyl-α-methylphenylacetic acid 1
M.p. 110-113°C (lit.3d 111-113.5°C).
1 H NMR (CDCl3, δ ppm) 7.51-7.55 (m, 2H), 7.49-7.37 (m, 4H), 7.21-7.16 (m, 2H), 3.85-3.78 (q, 1H, J = 7.1 Hz, CH), 1.60-1.57 (d, 3H, J = 7.1 Hz, CH3);
13C NMR (CDCl3 δ ppm) 180.4 (COOH), 161.3 & 158.0 (3-Ar-C), 140.9 & 140.8, 135.4, 130.9 & 130.8 (5-Ar-C), 128.9, 128.4, 128.2 & 128.0 (4-Ar-C), 127.7 (4′-Ar-C), 123.7 & 123.7 (6-Ar-C), 115.5 & 115.2 (2-Ar-C), 44.8 (CH), 18.0 (CH3).
 (d) Sagami Chemical Research Center. Jpn. Kokai Tokkyo Koho JP 8216840, 1982 (Chem. Abstr. 1982, 97: 5996s).
Flurbiprofen
 RACEMIC
Flurbiprofen
CAS : 5104-49-4
: 2-Fluoro-a-methyl[1,1¢-biphenyl]-4-acetic acid
Additional Names: 2-(2-fluoro-4-biphenylyl)propionic acid; 3-fluoro-4-phenylhydratropic acid
Manufacturers’ Codes: BTS-18322; U-27182
Trademarks: Adfeed (Lead Chem.); Ansaid (Pfizer); Antadys (Thžamex); Cebutid (Boots); Froben (Boots); Flurofen (Boots); Ocufen (Allergan); Stayban (Boots); Zepolas (Mikasa)
Molecular Formula: C15H13FO2
Molecular Weight: 244.26
Percent Composition: C 73.76%, H 5.36%, F 7.78%, O 13.10%
Literature References: Prepn: FR M5737; Adams et al., US 3755427 (1968, 1973 both to Boots Co., Ltd.). Pharmacology: Chalmers et al., Ann. Rheum. Dis. 31, 319 (1972); ibid. 32, 58 (1973); Glenn et al., Agents Actions 3, 210 (1973); Nishizawa et al.,Thromb. Res. 3, 577 (1973). HPLC determn in urine and plasma: J. M. Hutzler et al., J. Chromatogr. B 749, 119 (2000). Symposium on pharmacokinetics and clinical efficacy in pain management: Am. J. Med. 80, Suppl. 3A, 1-157 (1986).
Properties: Crystals from petr ether, mp 110-111°. Slightly sol in water (pH 7.0); readily sol in most polar solvents.
Melting point: mp 110-111°
Therap-Cat: Anti-inflammatory; analgesic.
 racemic

Flurbiprofen NMR spectra analysis, Chemical CAS NO. 5104-49-4 NMR spectral analysis, Flurbiprofen C-NMR spectrum

Flurbiprofen NMR spectra analysis, Chemical CAS NO. 5104-49-4 NMR spectral analysis, Flurbiprofen H-NMR spectrum

 s form
 (S)-Flurbiprofen NMR spectra analysis, Chemical CAS NO. 51543-39-6 NMR spectral analysis, (S)-Flurbiprofen H-NMR spectrum
 (S)-Flurbiprofen NMR spectra analysis, Chemical CAS NO. 51543-39-6 NMR spectral analysis, (S)-Flurbiprofen C-NMR spectrum
Patent Submitted Granted
Methods to accelerate the isolation of novel cell strains from pluripotent stem cells and cells obtained thereby [US2008070303] 2006-11-21 2008-03-20
Herpes Virus-Based Compositions and Methods of Use in the Prenatal and Perinatal Periods [US2008226601] 2006-06-05 2008-09-18
METHOD OF REDUCING ABETA42 AND TREATING DISEASES [US2008021085] 2007-06-21 2008-01-24
METHODS TO ACCELERATE THE ISOLATION OF NOVEL CELL STRAINS FROM PLURIPOTENT STEM CELLS AND CELLS OBTAINED THEREBY [US2010184033] 2009-07-16 2010-07-22
Pyridyl Amide T-Type Calcium Channel Antagonists [US2011112064] 2011-05-12
PROCESS FOR THE MANUFACTURE OF RACEMIC 2-ARYL-PROPIONIC ACID [US2011172460]
Patent Submitted Granted
Nitroxyderivatives having antinflammatory, analgesic and antithrombotic activity [US6613784] 2003-09-02
Global method for mapping property spaces [US6675136] 2004-01-06
Method of reducing Abeta42 and treating diseases [US2006004086] 2006-01-05
11-Beta-hydroxysteroid dehydrogenase 1 inhibitors useful for the treatment of diabetes, obesity and dyslipidemia [US7179802] 2004-06-03 2007-02-20
11-BETA-HYDROXYSTEROID DEHYDROGENASE 1 INHIBITORS USEFUL FOR THE TREATMENT OF DIABETES, OBESITY AND DYSLIPIDEMIA [US6730690] 2004-03-11 2004-05-04
Process for producing optically active flurbiprofen [US7214820] 2006-06-22 2007-05-08
Pyridyl Amide T-Type Calcium Channel Antagonists [US7875636] 2009-11-05 2011-01-25
METHOD FOR PRODUCING OPTICALLY ACTIVE ESTER AND METHOD FOR PRODUCING OPTICALLY ACTIVE CARBOXYLIC ACID [US8115008] 2010-09-16 2012-02-14
DRUG SUBSTANCE PREPARATIONS, PHARMACEUTICAL COMPOSITIONS AND DOSAGE FORMS [US2010087538] 2010-04-08
(R)-2-(3-Benzoylphenyl)propionic acid salts and pharmaceutical preparations containing them [EP0935961] 1999-08-18 2008-04-02

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Taisho Pharmaceutical Co., Ltd. (大正製薬株式会社 Taishō Seiyaku Kabushiki-gaisha?) (TYO: 4535) is a Japanese pharmaceutical company based in Tokyo.

.

////////////

TIRUPATI, INDIA
Map of tirupati town.
Tirupati
తిరుపతి
City
Clockwise from top: Tirumala Venkateswara Temple, Tirumala ghat road, City skyline and Chandragiri fort

Clockwise from top: Tirumala Venkateswara Temple, Tirumala ghat road, City skyline and Chandragiri fort

Tirupati is located in Andhra Pradesh

Tirupati
Tirupati

Location in Andhra Pradesh, India

Coordinates: 13.65°N 79.42°ECoordinates: 13.65°N 79.42°E
Country India
State Andhra Pradesh
Region Rayalaseema
District Chittoor
Government
 • Member of Parliament Varaprasad Rao Velagapalli
Area
 • City 24 km2 (9 sq mi)
Elevation 161 m (528 ft)
Population (2011)[1]
 • City 287,035
 • Density 12,000/km2 (31,000/sq mi)
 • Metro[2] 459,985
Languages
 • Official Telugu
Time zone IST (UTC+5:30)
PIN 517501
Telephone code +91–877
Vehicle registration AP 03
Website Tirupati Mucnicipal Corporation

.

.
Kapila Theertham in Tirupati



Food Service During Tirumala Tirupati Devastanam’s ‘Srinivasa Kalyanam Utsavam’ at MARG Swarnabhoomi

 

 

Evofosfamide


TH-302.svg

Evofosfamide, HAP-302 , TH-302

Evofosfamide
TH-302.svg
Names
IUPAC name

(1-Methyl-2-nitro-1H-imidazol-5-yl)methyl N,N’-bis(2-bromoethyl)phosphorodiamidate
Other names

TH-302; HAP-302
Identifiers
918633-87-1 Yes
ChemSpider 10157061 Yes
Jmol-3D images Image
PubChem 11984561
Properties
C9H16Br2N5O4P
Molar mass 449.04 g·mol−1
6 to 7 g/l

 

TH-302 is a nitroimidazole-linked prodrug of a brominated derivative of an isophosphoramide mustard previously used in cancer drugs

evofosfamide (first disclosed in WO2007002931), useful for treating cancer.

Threshold Pharmaceuticals and licensee Merck Serono are codeveloping evofosfamide, the lead in a series of topoisomerase II-inhibiting hypoxia-activated prodrugs and a 2-nitroimidazole-triggered bromo analog of ifosfamide, for treating cancer, primarily soft tissue sarcoma and pancreatic cancer (phase 3 clinical, as of April 2015).

In November 2014, the FDA granted Fast Track designation to the drug for the treatment of previously untreated patients with metastatic or locally advanced unresectable soft tissue sarcoma.

 

Evofosfamide (INN,[1] USAN;[2] formerly known as TH-302) is an investigational hypoxia-activated prodrug that is in clinical development for cancer treatment. The prodrug is activated only at very low levels of oxygen (hypoxia). Such levels are common in human solid tumors, a phenomenon known as tumor hypoxia.[3]

Evofosfamide is being evaluated in clinical trials for the treatment of multiple tumor types as a monotherapy and in combination with chemotherapeutic agents and other targeted cancer drugs
Discovered at Threshold, TH-302 is a hypoxia-activated prodrug (HAP) designed to exploit low oxygen levels in hypoxic tumor regions. Therapeutics that specifically target resistant hypoxic zones could provide significant additional antitumor activity and clinical benefit over current chemotherapeutic and radiation therapies.

Evofosfamide (TH-302) was developed by Threshold Pharmaceuticals Inc. (Threshold).[4] The company is located in South San Francisco, CA, USA.

In 2012, Threshold signed a global license and co-development agreement for evofosfamide with Merck KGaA, Darmstadt, Germany, which includes an option for Threshold to co-commercialize eofosfamide in the United States. Threshold is responsible for the development of evofosfamide in the soft tissue sarcoma indication in the United States. In all other cancer indications, Threshold and Merck KGaA are developing evofosfamide together.[5] From 2012 to 2013, Merck KGaA paid 110 million US$ for upfront payment and milestone payments to Threshold. Additionally, Merck KGaA covers 70% of all evofosfamide development expenses.[6]
Discovered at Threshold, TH-302 is a hypoxia-activated prodrug (HAP) designed to exploit low oxygen levels in hypoxic tumor regions. Therapeutics that specifically target resistant hypoxic zones could provide significant additional antitumor activity and clinical benefit over current chemotherapeutic and radiation therapies.

History

Date Event
Jun 2005 Threshold files evofosfamide (TH-302) patent applications in the U.S.[49]
Jun 2006 Threshold files a evofosfamide (TH-302) patent application in the EU and in Japan[50]
Sep 2011 Threshold starts a Phase 3 trial (TH-CR-406) of evofosfamide in combination with doxorubicin in patients with soft tissue sarcoma
Feb 2012 Threshold signs an agreement with Merck KGaA to co-develop evofosfamide
Apr 2012 A Phase 2b trial (TH-CR-404) of evofosfamide in combination with gemcitabine in patients with pancreatic cancer meets primary endpoint

SEE

WO2007002931

http://www.google.com/patents/WO2007002931A2?cl=en

Example 8

Synthesis of Compounds 25, 26 [0380] To a solution of 2-bromoethylammmonium bromide (19.4 g) in DCM (90 mL) at – 1O0C was added a solution OfPOCl3 (2.3 mL) in DCM (4 mL) followed by addition of a solution of TEA (14.1 mL) in DCM (25 mL). The reaction mixture was filtered, the filtrate concentrated to ca. 30% of the original volume and filtered. The residue was washed with DCM (3×25 mL) and the combined DCM portions concentrated to yield a solid to which a mixture of THF (6 mL) and water (8 mL) was added. THF was removed in a rotary evaporator, the resulting solution chilled overnight in a fridge. The precipitate obtained was filtered, washed with water (10 mL) and ether (30 mL), and dryed in vacuo to yield 2.1 g of:

Isophosphoramide mustard

can be synthesized employing the method provided in Example 8, substituting 2- bromoethylammmonium bromide with 2-chloroethylammmonium chloride. Synthesis of Isophosphoramide mustard has been described (see for example Wiessler et al., supra).

The phosphoramidate alkylator toxin:

was transformed into compounds 24 and 25, employing the method provided in Example 6 and the appropriate Trigger-OH.

Example 25

Synthesis of l-N-methyl-2-nitroimidazole-5-carboxylis acid

A suspension of the nitro ester (39.2 g, 196.9 rnmol) in IN NaOH (600 mL) and water (200 mL) was stirred at rt for about 20 h to give a clear light brown solution. The pH of the reaction mixture was adjusted to about 1 by addition of cone. HCl and the reaction mixture extracted with EA (5 x 150 mL). The combined ethyl acetate layers were dried over MgS O4 and concentrated to yield l-N-methyl-2-nitroimidazole-5-carboxylis acid (“nitro acid”) as a light brown solid (32.2 g, 95%). Example 26

Synthesis of l-N-methyl-2-nitroimidazole-5-carboxylis acid

A mixture of the nitro acid (30.82 g, 180.23 mmol) and triethylamine (140 niL, 285 mmol) in anhydrous THF (360 mL) was stirred while the reaction mixture was cooled in a dry ice-acetonitrile bath (temperature < -20 0C). Isobutyl chloroformate (37.8 mL, 288 mmol) was added drop wise to this cooled reaction mixture during a period of 10 min and stirred for 1 h followed by the addition of sodium borohydride (36 g, 947 mmol) and dropwise addition of water during a period of 1 h while maintaining a temperature around or less than O0C. The reaction mixture was warmed up to O0C. The solid was filtered off and washed with THF. The combined THF portions were evaporated to yield l-N-methyl-2- nitroimidazole-5-methanol as an orange solid (25 g) which was recrystallized from ethyl acetate.

……………………………………….

WO-2015051921

EXAMPLE 1

1

N-Formylsarcosine ethyl ester 1 (1 ,85 kg) was dissolved in toluene (3,9 kg) and ethyl formate (3,28 kg) and cooled to 10 °C. A 20 wt-% solution of potassium tert-butoxide (1 ,84 kg) in tetrahydrofuran (7,4 kg) was added and stirring was continued for 3h. The reaction mixture was extracted 2x with a solution of sodium chloride in water (10 wt-%) and the combined water extracts were washed lx with toluene.

Aqueous hydrogen chloride (25% wt-%; 5,62 kg) was added to the aqueous solution, followed by ethylene glycol (2,36 kg). The reaction mixture was heated to 55-60 °C for lh before only the organic solvent residues were distilled off under vacuum.

Aqueous Cyanamide (50 wt-%, 2,16 kg) was then added at 20 °C, followed by sodium acetate (3,04 kg). The resulting reaction mixture was heated to 85-90 °C for 2h and cooled to 0-5 °C before a pH of ~ 8-9 was adjusted via addition of aqueous sodium hydroxide (32% wt-%; 4,1 kg). Compound 3 (1,66 kg; 75%) was isolated after filtration and washing with water.

Ή-NMR (400 MHz, d6-DMSO): δ= 1,24 (3H, t, J= 7,1 Hz); 3,53 (3H, s); 4,16 (2H, q, J= 7,0 Hz) ; 6,15 (s, 2 H); 7,28 (s, 1H).

HPLC (Rt = 7,7 min): 97,9% (a/a).

REFERENCES

 

1

Threshold Pharmaceuticals Form 8-K from 3 Nov 2014

 

DHAKA BANGLADESH

 

.

Steamers and ferries in Sadarghat Port

Kawran Bazar

.

Dry fish sellers at the Karwan Dry Fish Market (Bazar), Dhaka, Bangladesh.

Masitinib


Masitinib

Masitinib; 790299-79-5; Masivet; AB1010; AB-1010;

CLASS:Immunomodulator
TARGET:KIT (a stem cell factor, also called c-KIT) receptor as well as select other tyrosine kinases
STATUS FOR MS:Phase III
COMMERCIAL:Under development by AB Science..Ab Science
4-((4-Methylpiperazin-1-yl)methyl)-N-(4-methyl-3-((4-(pyridin-3-yl)-1,3-thiazol-2-yl)amino)phenyl)benzamide
AB 1010
UNII-M59NC4E26P

4-((4-Methylpiperazin-1-yl)methyl)-N-(4-methyl-3-((4-(pyridin-3-yl)-1,3-thiazol-2-yl)amino)phenyl)benzamide

Regulatory and Commercial Status

STATUS FOR MS:Phase III
HIGHEST STATUS ACHIEVED (FOR ANY CONDITION):
Marketing Authorization Application for the treatment of pancreatic cancer has been filed with the European Medicines Agency (16 October 2012)
Marketing Authorization Application for the conditional approval in the treatment of pancreatic cancer has been accepted by the European Medicines Agency (30 October 2012)

Masitinib.png

Masitinib is a tyrosine-kinase inhibitor used in the treatment of mast cell tumors in animals, specifically dogs.[1][2] Since its introduction in November 2008 it has been distributed under the commercial name Masivet. It has been available in Europe since the second part of 2009. In the USA it is distributed under the name Kinavet and has been available for veterinaries since 2011.

Masitinib is being studied for several human conditions including cancers. It is used in Europe to fight orphan diseases.[3]

Mechanism of action

Masitinib inhibits the receptor tyrosine kinase c-Kit which is displayed by various types of tumour.[2] It also inhibits the platelet derived growth factor receptor (PDGFR) and fibroblast growth factor receptor (FGFR).

 

http://www.google.com/patents/WO2012136732A1?cl=en

In a preferred embodiment of the above-depicted treatment, the active ingredient masitinib is administered in the form of masitinib mesilate; which is the orally bioavailable mesylate salt of masitinib – CAS 1048007-93-7 (MsOH); C28H30N6OS.CH3SO3H; MW 594.76:

Figure imgf000031_0001

 

http://www.google.com/patents/WO2004014903A1?cl=en

Figure imgf000021_0001

003 : 4-(4-Methyl-piperazin-l-ylmethyl)-N-[3-(4-pyridin-3-yl-thiazol-2-ylamino)- phenyl] -benzamide

4-(4-Methyl-piperazin-l-yl)-N-[4-methyl-3-(4-pyridin-3-yl-thiazol-2-ylmethyl)- phenyl] -benzamide

Figure imgf000053_0001

beige brown powder mp : 128-130°C

1H RMN (DMSO-d6) δ = 2.15 (s, 3H) ; 2.18 (s, 3H) ; 2.35-2.41 (m, 4H) ; 3.18-3.3.24 (m, 4H) ; 6.94 (d, J = 8.9 Hz, 2H) ; 7.09 (d, J = 8.4 Hz, IH) ; 7.28-7.38 (m, 3H) ; 7.81 (d, J = 8.9 Hz, 2H) ; 8.20-8.25 (m, IH) ; 8.40 (dd, J = 1.6 Hz, J = 4.7 , IH) ; 8.48 (d, J = 1.9 Hz, IH) ; 9.07 (d, J = 1.5 Hz, IH) ; 9.35 (s, IH) ; 9.84 (s, IH)

……………

http://www.google.com/patents/WO2008098949A2?cl=en

EXAMPLE 4 N- [4-Methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino)-phenyl] -benzamide derivatives

Method A In a reactor and under low nitrogen pressure, add 4-Methyl-N3-(4-pyridin-3-yl-thiazol- 2-yl)-benzene-l,3-diamine (95 g, 336.45 mmol), dichloromethane (2 L). To this suspension cooled to temperature of 5°C was added dropwise 2M/n-hexane solution of trimethylaluminium (588 mL). The reaction mixture was brought progressively to 15°C, and maintained for 2 h under stirring. 4-(4-Methyl-piperazin-l-ylmethyl)-benzoic acid methyl ester (100 g, 402.71 mmol) in dichloromethane (200 mL) was added for 10 minutes. After 1 h stirring at room temperature, the reaction mixture was heated to reflux for 20 h and cooled to room temperature. This solution was transferred dropwise via a cannula to a reactor containing 2N NaOH (2.1 L) cooled to 5°C. After stirring for 3 h at room temperature, the precipitate was filtered through Celite. The solution was extracted with dichloromethane and the organic layer was washed with water and saturated sodium chloride solution, dried over MgSO4 and concentrated under vacuum. The brown solid obtained was recrystallized from /-Pr2O to give 130.7 g (78%) of a beige powder.

Method B Preparation of the acid chloride

To a mixture of 4-(4-Methyl-piperazin-l-ylmethyl)-benzoic acid dihydrochloride (1.0 eq), dichloromethane (7 vol) and triethylamine (2.15 eq), thionyl chloride (1.2 eq) was added at 18-28°C . The reaction mixture was stirred at 28-32°C for 1 hour. Coupling of acid chloride with amino thiazole To a chilled (0-50C) suspension of 4-Methyl-N3-(4-pyridin-3-yl-thiazol-2-yl)-benzene- 1,3-diamine (0.8 eq) and thiethylamine (2.2 eq) in dichloromethane (3 vol), the acid chloride solution (prepared above) was maintaining the temperature below 5°C. The reaction mixture was warmed to 25-300C and stirred at the same temperature for 1O h. Methanol (2 vol) and water (5 vol) were added to the reaction mixture and stirred. After separating the layers, methanol (2 vol), dihloromethane (5 vol) and sodium hydroxide solution (aqueous, 10%, till pH was 9.5-10.0) were added to the aqueous layer and stirred for 10 minutes. The layers were separated. The organic layer was a washed with water and saturated sodium chloride solution. The organic layer was concentrated and ethanol (2 vol) was added and stirred. The mixture was concentrated. Ethanol was added to the residue and stirred. The product was filtered and dried at 50-550C in a vaccum tray drier. Yield = 65-75%.

Method C

To a solution of 4-methyl-N3-(4-pyridin-3-yl-thiazol-2-yl)-benzene-l,3-diamine (1.0 eq) in DMF (20 vol) were added successively triethylamine (5 eq), 2-chloro-l- methylpyridinium iodide (2 eq) and 4-(4-methyl-piperazin-l-ylmethyl)-benzoic acid (2 eq). The reaction mixture was stirred for 7 h at room temperature. Then, the mixture was diluted in diethyl ether and washed with water and saturated aqueous NaHCO3, dried over Na2SO4 and concentrated. The crude product was purified by column chromatography using an elution of 100% EtOAc to give a yellow solid.

Yield = 51%.

1H NMR (CDCl3) : δ = 9.09 (IH, s, NH); 8.52 (IH, br s); 8.27 (IH, s); 8.13 (IH, s);

8.03 (IH, s); 7.85 (2H, d, J= 8.3Hz); 7.45 (2H, m); 7.21-7.38 (4H, m); 6.89 (IH, s);

3.56 (2H, s); 2.50 (8H, br s); 2.31 (6H, br s).

MS (CI) m/z = 499 (M+H)+.

An additional aspect of the present invention relates to a particular polymorph of the methanesulfonic acid salt of N-[4-Methyl-3-(4-pyridin-3-yl-thiazol-2-ylamino)-phenyl]- benzamide of formula (IX).

Figure imgf000023_0001

(VI)

Hereinafter is described the polymorph form of (IX) which has the most advantageous properties concerning processability, storage and formulation. For example, this form remains, dry at 80% relative humidity and thermodynamically stable at temperatures below 2000C.

The polymorph of this form is characterized by an X-ray diffraction pattern illustrated in FIG.I, comprising characteristic peaks approximately 7.269, 9.120, 11.038, 13.704, 14.481, 15.483, 15.870, 16.718, 17.087, 17.473, 18.224, 19.248, 19.441, 19.940, 20.441, 21.469, 21.750, 22.111, 23.319, 23.763, 24.120, 24.681, 25.754, 26.777, 28.975, 29.609, 30.073 degrees θ, and is also characterized by differential scanning calorimetry (DSC) illustrated in FIG.II, which exhibit a single maximum value at approximately 237.49 ± 0.3 0C. X-ray diffraction pattern is measured using a Bruker AXS (D8 advance). Differential scanning calorimetry (DSC) is measured using a Perking Elmer Precisely (Diamond DSC).

This polymorph form can be obtained by treatement of 4-(4-Methyl-piperazin-l- ylmethyl)-N-[4-methyl-3-(4-pyridin-3-yl-thiazol-2-ylamino)-phenyl]-benzamide with 1.0 to 1.2 equivalent of methanesulfonic acid, at a suitable temperature, preferably between 20-800C.

The reaction is performed in a suitable solvent especially polar solvent such as methanol or ethanol, or ketone such as acetone, or ether such as diethylether or dioxane, or a mixture therof. This invention is explained in example given below which is provided by way of illustration only and therefore should not be construed to limit the scope of the invention. Preparation of the above-mentioned polymorph form of 4-(4-Methyl-piperazin-l- ylmethyl)-N- [4-methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino)-phenyl] -benzamide methanesulfonate .

4-(4-Methyl-piperazin- 1 -ylmethyl)-N- [4-methyl-3 -(4-pyridin-3 -yl-thiazol-2-ylamino) phenyl] -benzamide (1.0 eq) was dissolved in ethanol (4.5 vol) at 65-700C. Methanesulfonic acid (1.0 eq) was added slowly at the same temperature. The mixture was cooled to 25-300C and maintained for 6 h. The product was filtered and dried in a vacuum tray drier at 55-600C. Yield = 85-90%. Starting melting point Smp = 236°C.

References

  1. Hahn, K.A.; Oglivie, G.; Rusk, T.; Devauchelle, P.; Leblanc, A.; Legendre, A.; Powers, B.; Leventhal, P.S.; Kinet, J.-P.; Palmerini, F.; Dubreuil, P.; Moussy, A.; Hermine, O. (2008). “Masitinib is Safe and Effective for the Treatment of Canine Mast Cell Tumors”. Journal of Veterinary Internal Medicine 22 (6): 1301–1309. doi:10.1111/j.1939-1676.2008.0190.x. ISSN 0891-6640.
  2.  Information about Masivet at the European pharmacy agency website
  3.  Orphan designation for Masitinib at the European pharmacy agency website
WO2004014903A1 Jul 31, 2003 Feb 19, 2004 Ab Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors
WO2008098949A2 Feb 13, 2008 Aug 21, 2008 Ab Science Process for the synthesis of 2-aminothiazole compounds as kinase inhibitors
EP1525200B1 Jul 31, 2003 Oct 10, 2007 AB Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors
US7423055 Aug 1, 2003 Sep 9, 2008 Ab Science 2-(3-Aminoaryl)amino-4-aryl-thiazoles for the treatment of diseases
US20080207572 * Jul 13, 2006 Aug 28, 2008 Ab Science Use of Dual C-Kit/Fgfr3 Inhibitors for Treating Multiple Myeloma
Masitinib.svg
Systematic (IUPAC) name
4-[(4-Methylpiperazin-1-yl)methyl]-N-(4-methyl-3-{[4-(pyridin-3-yl)-1,3-thiazol-2-yl]amino}phenyl)benzamide
Clinical data
Trade names Masivet, Kinavet
AHFS/Drugs.com International Drug Names
Identifiers
790299-79-5
L01XE22
PubChem CID 10074640
ChemSpider 8250179
ChEMBL CHEMBL1908391
Chemical data
Formula C28H30N6OS
498.64 g/mol
Patent Submitted Granted
2-(3-Aminoaryl)amino-4-aryl-thiazoles for the treatment of diseases [US7423055] 2004-06-10 2008-09-09
2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors [US2005239852] 2005-10-27
Use of C-Kit Inhibitors for Treating Fibrosis [US2007225293] 2007-09-27
Use of Mast Cells Inhibitors for Treating Patients Exposed to Chemical or Biological Weapons [US2007249628] 2007-10-25
Use of c-kit inhibitors for treating type II diabetes [US2007032521] 2007-02-08
Use of tyrosine kinase inhibitors for treating cerebral ischemia [US2007191267] 2007-08-16
Use of C-Kit Inhibitors for Treating Plasmodium Related Diseases [US2008004279] 2008-01-03
Tailored Treatment Suitable for Different Forms of Mastocytosis [US2008025916] 2008-01-31
2-(3-AMINOARYL) AMINO-4-ARYL-THIAZOLES AND THEIR USE AS C-KIT INHIBITORS [US2008255141] 2008-10-16
Use Of C-Kit Inhibitors For Treating Inflammatory Muscle Disorders Including Myositis And Muscular Dystrophy [US2008146585] 2008-06-19
Patent Submitted Granted
Aminothiazole compounds as kinase inhibitors and methods of using the same [US8940894] 2013-05-10 2015-01-27
Aminothiazole compounds as kinase inhibitors and methods of using the same [US8492545] 2012-03-08 2013-07-23
Patent Submitted Granted
Use of Dual C-Kit/Fgfr3 Inhibitors for Treating Multiple Myeloma [US2008207572] 2008-08-28
PROCESS FOR THE SYNTHESIS OF 2-AMINOTHIAZOLE COMPOUNDS AS KINASE INHIBITORS [US8153792] 2010-05-13 2012-04-10
COMBINATION TREATMENT OF SOLID CANCERS WITH ANTIMETABOLITES AND TYROSINE KINASE INHIBITORS [US8227470] 2010-04-15 2012-07-24
Anti-IGF antibodies [US8580254] 2008-06-19 2013-11-12
COMBINATIONS FOR THE TREATMENT OF B-CELL PROLIFERATIVE DISORDERS [US2009047243] 2008-07-17 2009-02-19
TREATMENTS OF B-CELL PROLIFERATIVE DISORDERS [US2009053168] 2008-07-17 2009-02-26
Anti-IGF antibodies [US8318159] 2009-12-11 2012-11-27
SURFACE TOPOGRAPHIES FOR NON-TOXIC BIOADHESION CONTROL [US2010226943] 2009-08-31 2010-09-09
EGFR/NEDD9/TGF-BETA INTERACTOME AND METHODS OF USE THEREOF FOR THE IDENTIFICATION OF AGENTS HAVING EFFICACY IN THE TREATMENT OF HYPERPROLIFERATIVE DISORDERS [US2010239656] 2010-05-10 2010-09-23
ANTI CD37 ANTIBODIES [US2010189722] 2008-08-08 2010-07-29
United States National Library of Medicine

Note: Compound name must be entered under “Substance Identification” and then “Names and Synonyms” selected to view synonyms.

Kocic I, Kowianski P, Rusiecka I, Lietzau G, Mansfield C, Moussy A, Hermine O, Dubreuil P
Naunyn Schmiedebergs Arch Pharmacol. 2014 Oct 26. Epub 2014 Oct 26. PMID: 25344204.Abstract
 AB SCIENCE HEADQUARTER
3, Avenue George V
75008 PARIS – FRANCE
Tel. : +33 (0)1 47 20 00 14
Fax. : +33 (0)1 47 20 24 11

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Brexpiprazole ブレクスピプラゾール


Brexpiprazole structure.svg

Brexpiprazole

ブレクスピプラゾール

OPC-34712, UNII-2J3YBM1K8C, OPC34712,
CAS 913611-97-9,
Molecular Formula:C25H27N3O2S
Molecular Weight:433.56578 g/mol
7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one
7-[4-[4-(1-Benzothiophen-4-yl)piperazin-1-yl]butoxy]quinolin-2(1H)-one
2(1H)​-​Quinolinone, 7-​[4-​(4-​benzo[b]​thien-​4-​yl-​1-​piperazinyl)​butoxy]​-
7- [ 4- ( 4-benzo[b]thiophen-4- yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one
7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one
Otsuka Pharma Co Ltd,

OTSUKA ……………INNOVATOR

NDA is considered filed as of September 9, 2014 (60 days after submission). The PDUFA date is July 11, 2015.

 UPDATE JULY 2015 ON STATUS OF APPROVAL

Approval Status:

Approved July 2015

Specific Treatments:

depression and schizophrenia

Therapeutic Areas

Brexpiprazole (/brɛksˈpɪprəzl/ breks-pip-rə-zohl; also called OPC-34712) is a novel D2 dopamine partial agonist investigational product currently in clinical trials for the treatment of depression, schizophrenia, and attention deficit hyperactivity disorder(ADHD).[1]Although it failed Stage 2 trials for ADHD, it has been designed to provide improved efficacy and tolerability (e.g., lessakathisia, restlessness and/or insomnia) over established adjunctive treatments for major depressive disorder (MDD).[2]

OPC-34712 is an antidepressant and antipsychotic drug candidate awaiting approval in the U.S. for the treatment of schizophrenia and also as adjunctive treatment of major depressive disorder (MDD). The product is in phase III clinical trials for the treatment of agitation associated with Alzheimer’s disease. Phase III clinical trials are also underway for the treatment of post-traumatic stress disorder (PTSD).

brexpiprazole (pre-registration, as of April 2015), which is being developed by Otsuka and Lundbeck, useful for treating schizophrenia, agitation associated with Alzheimer’s disease, major depressive disorder and attention deficit hyperactivity disorder. Family members of the product case, WO2006112464, hold protection in EU states until 2026 and its US equivalent, US7888362, has US154 extension, expiring in 2027. Suzhou Vigonvita Life Sciences appears to be new to patenting and is the first collaborative filing from the three assignees.

Phase II clinical trials are also ongoing for use as adjunctive therapy in adults with attention deficit hyperactivity disorder (ADHD). The compound is being developed by Otsuka Pharmaceutical. In 2011, a codevelopment and commercialization agreement was signed by Lundbeck and Otsuka Pharmaceutical in Latin and North America, Australia and Europe for the treatment of psychiatric disorders.

The drug is being developed by Otsuka, and is considered to be a successor[3] of its top-selling antipsychotic agent aripiprazole(brand names: Abilify, Aripiprex). Otsuka’s US patent on aripiprazole expired on October 20, 2014;[4] however, due to a pediatric extension, a generic will not become available until at least April 20, 2015.[5]

Brexpiprazole (1) , a serotonin–dopamine activity modulator, is an investigational new drug currently in phase-III clinical trials for the treatment of depression, schizophrenia, and attention deficit hyperactivity disorder.(1A) Brexpiprazole is also considered to be a possible successor to the top-selling antipsychotic agent aripiprazole.(2A)

  1. 1A……….Phase II and Phase III Drugs in U.S. Development for Depression, Anxiety, Sleep Disorders, Psychosis & ADHD, 2011. http://www.neurotransmitter.net/newdrugs.html(accessed Jan 27, 2015).

  2. 2A…………FDA accepts new schizophrenia drug filing, 2014.http://www.pharmafile.com/news/194878/fda-accepts-new-schizophrenia-drug-filing(accessed Jan 27, 2015).
    BREXPIPRAZOLE.png
    Brexpiprazole

    In the clinical program, brexpiprazole demonstrated improvement in symptoms in both schizophrenia and as adjunctive therapy in major depressive disorder (MDD)

    July 2015 is the anticipated completion timing of the FDA’s review (based on PDUFA timeline)Otsuka Pharmaceutical Co., Ltd. (Otsuka) and H. Lundbeck A/S (Lundbeck) today announced that the U.S. Food and Drug Administration (FDA) has determined that the New Drug Application (NDA) for brexpiprazole for monotherapy in adult patients with schizophrenia and for adjunctive treatment of major depressive disorder (MDD) in adult patients is sufficiently complete to allow for a substantive review, and the NDA is considered filed as of September 9, 2014 (60 days after submission). The PDUFA date is July 11, 2015.The NDA is supported by seven completed placebo-controlled clinical phase II or III studies in the proposed indications – three studies in schizophrenia and four studies with brexpiprazole as adjunctive therapy in MDD. The dossier included data from more than 6,000 participants of whom more than 5,000 received brexpiprazole.

    Brexpiprazole in adult patients with schizophreniaOne clinical phase II and two clinical phase III placebo-controlled studies have been completed using brexpiprazole in adult patients suffering from schizophrenia. Across the three studies more than 1,700 patients have been randomized.In the first pivotal phase III study randomizing approximately 625 patients, brexpiprazole 2mg/day and 4 mg/day both demonstrated greater improvement of symptoms relative to placebo as measured by change from baseline in the Positive and Negative Syndrome Scale (PANSS) Total Score at week 6 (p<0.05). Results of the key secondary endpoint supported primary results.In the second pivotal phase III study randomizing approximately 650 patients, brexpiprazole 4 mg/day again demonstrated greater improvement of symptoms relative to placebo (p<0.05) in change from baseline in the PANSS Total Score at Week 6. Brexpiprazole 2 mg/day showed numerical improvement (p>0.05) over placebo at Week 6.The results from the clinical phase II studyi were presented at the 24th Annual US Psychiatric and Mental Health Congress in November 2011. The study showed a clinically meaningful improvement from baseline measured by PANSS total score at week 6, although it did not achieve statistical separation from placeboii.In the placebo-controlled phase II and III studies, the rates of discontinuation due to adverse events were 8.1% for patients receiving brexpiprazole compared to 12.7% of patients receiving placebo; the only adverse event that occurred in more than 5% of brexpiprazole patients and more frequently than placebo was akathisia (5.8% vs. 4.5%).
    Brexpiprazole as adjunctive therapy in major depressive disorder (MDD) Four studies have been included in the dossier using brexpiprazole as adjunctive therapy for adult patients suffering from MDD who had demonstrated a consistent, inadequate response to at least two regimens of prior antidepressant treatment. Patients with MDD and an inadequate response to one to three antidepressants were enrolled and received antidepressants for 8 weeks, single blinded, in the two phase III studies. Patients with an inadequate response during this prospective phase were provided antidepressant therapy and randomized adjunctive treatment with either brexpiprazole or placebo for 6 weeks. The primary efficacy endpoint was the change in MADRS (Montgomery–Åsberg Depression Rating Scale) Total Score from baseline at week 6. MADRS is a commonly used scale to assess the range of symptoms in patients with MDD. Across the four studies, more than 3,900 patients entered the prospective phase and more than 1,800 patients were included in the randomized phase of the studies.The first pivotal phase III results were presented in a poster session at the 22nd European Psychiatry Association Congress (EPA) in March 2014. This two-arm phase III study randomized approximately 380 patients and demonstrated an improvement of symptoms with an antidepressant plus 2 mg brexpiprazole that was greater than an antidepressant plus placebo (p<0.001)The second pivotal phase III study was a three-arm study in which approximately 675 patients were randomized to treatment with an antidepressant plus either placebo, 1 mg brexpiprazole or 3 mg brexpiprazole.v Patients in both brexpiprazole treatment groups showed greater improvement in symptoms as measured by the MADRS compared to placebo (1 mg p>0.05, 3 mg p<0.05). Results of the second pivotal phase III study in MDD have not yet been published.

    The first clinical phase IIvi study randomized approximately 425 patients in four arms and was presented at the 164th Annual Meeting of the American Psychiatric Association in May 2011. Patients exhibited greater improvements than adjunctive placebo in MADRS Total score with the 1.5 (±0.5) mg/day dose of brexpiprazole after six weeks of treatment (p

    About brexpiprazole (OPC-34712)Brexpiprazole is a novel investigational psychotropic compound discovered by Otsuka and under co-development with Lundbeck. Brexpiprazole is a serotonin-dopamine activity modulator (SDAM) that acts as a partial agonist at 5-HT1A and dopamine D2 receptors at similar potency, and an antagonist at 5-HT2A and noradrenaline alpha1B/2C receptors.

Partnership with Lundbeck

In November 2011, Otsuka and Lundbeck have announced a global alliance.[6] Lundbeck has given Otsuka an upfront payment of $200 million, and the deal includes development, regulatory and sales payments, for a potential total of $1.8 billion. Specifically for OPC-34712, Lundbeck will obtain 50% of net sales in Europe and Canada and 45% of net sales in the US from Otsuka.

The partnership has been presented by Otsuka to its investors as a good fit for several reasons:[7]

  • Geographic strategy: Otsuka in Japan, Asia, US; Lundbeck in Europe, South America and emerging markets
  • Research strategy: Otsuka has knowledge in antipsychotics, Lundbeck in anti-depressant and anxiolytic.
  • CNS strategy: Otsuka has a robust portfolio in next-generation CNS drugs, while Lundbeck covers a wide range of CNS conditions from Alzheimer’s to schizophrenia.
  • Similar corporate culture

Clinical trials

OPC-34712 is currently in clinical trials for adjunctive treatment of MDD, adjunctive treatment of adult ADHD and schizophrenia.[8]

Major depression

Phase II

The Phase 2 multicenter, double-blind, placebo-controlled study randomized 429 adult MDD patients who exhibited an inadequate response to one to three ADTs in the current episode. The study was designed to assess the efficacy and safety of OPC-34712 as an adjunctive treatment to standard ADT. The ADTs included in the study were desvenlafaxine, escitalopram, fluoxetine, paroxetine, sertraline, and venlafaxine.[9]

Phase III

A new Phase III study is currently in the recruiting stage: “Study of the Safety and Efficacy of Two Fixed Doses of OPC-34712 as Adjunctive Therapy in the Treatment of Adults With Major Depressive Disorder (the Polaris Trial)”.[10] Its goal is “to compare the effect of OPC-34712 to the effect of placebo (an inactive substance) as add on treatment to an assigned FDA approved antidepressant treatment (ADT) in patients with Major Depressive Disorder who demonstrate an incomplete response to a prospective trial of the same assigned FDA approved ADT”. Estimated enrollment is 1250 volunteers.

Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates, and its role in multiple receptors, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (improve depressive symptoms); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as adjuvant treatment of severe depression (MDD) Phase III clinical trial; III clinical trial for the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States Phase II adult ADHD Clinical Trials.

Adult ADHD

Phase II

  • Study of the Safety and Efficacy of OPC-34712 as a Complementary Therapy in the Treatment of Adult Attention Deficit/Hyperactivity Disorder (STEP-A)[11] The company did not move the product to Phase III, and it is presumed this drug failed Phase II trials for the disorder.

Schizophrenia

Phase I

  • Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder[12]

Phase II

  • A Dose-finding Trial of OPC-34712 in Patients With Schizophrenia[13]

Phase III

  • Efficacy Study of OPC-34712 in Adults With Acute Schizophrenia (BEACON)[14]
  • Safety and Tolerability Study of Oral OPC-34712 as Maintenance Treatment in Adults With Schizophrenia (ZENITH)[15]
  • Study of the Effectiveness of Three Different Doses of OPC-34712 in the Treatment of Adults With Acute Schizophrenia (VECTOR)[16]
  • A Long-term Trial of OPC-34712 in Patients With Schizophrenia[17]

Conferences

  • Phase II results were presented at the American Psychiatric Association’s 2011 annual meeting in May 2011.[18]
  • The drug has been presented at the 2nd Congress of Asian College of Neuropsychopharmacology[19] in September 2011.
  • At the US Psychiatric and Mental Health Congress in November 2011 in Vegas, Robert McQuade presented the Phase II Trial results for Schizophrenia[20]

 Pharmacology

Brexpiprazole acts as a partial agonist of the 5-HT1A, D2, and D3 receptors, and as an antagonist of the 5-HT2A, 5-HT2B, 5-HT7, α1A, α1B, α1D, and α2C-adrenergic, and H1receptors.[22] It has negligible affinity for the mACh receptors.[22]

Patents

  • U.S. Patent 8,071,600
  • WIPO PCT/JP2006/317704
  • Canadian patent: 2620688[24]
  • WO 2013162046
  • WO 2013161830
  •  WO 2013162048
  • WO 2013015456
  • JP 2008115172
  • WO 2006112464
  • WO2015054976 NEW
Patent Submitted Granted
PIPERAZINE-SUBSTITUTED BENZOTHIOPHENES FOR TREATMENT OF MENTAL DISORDERS [US2011152286] 2011-06-23
Piperazine-substituted benzothiophenes for treatment of mental disorders [US7888362] 2010-07-15 2011-02-15
Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2006112464A1 in the preparation route see Scheme 1, the difficulty of the route is the first reaction generates byproducts easily separated by column chromatography is not easy to obtain high-purity intermediates, thus affecting the final product Bray prazosin purity and yield.Scheme 1:

Subsequently, Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2013015456A1 in the alternative method of preparing the reaction of this step, see Scheme 2, the route the reagents are more expensive, high-cost, environmentally unfriendly and not suitable for industrial production.

Reaction Scheme 2:

Due to the above production process there is a high cost, and difficult to separate impurities and other shortcomings, it is necessary to find an economical, practical, environmental protection, new routes to improve process stability, reduce costs, improve product quality.

Synthesis

WO 2013015456

IN THIS BELOW PIC WE SEE

click on pics below to view

Synthesis of A

1 BROMO 4 CHLORO BUTANE WAS REACTED WITH 7 HYDROXY 1H QUINOLINE -2-ONE TO GIVE A

7 ( 4 CHLORO BUTOXY)-1H -QUINOLINE-2-ONE, WHICH WILL BE USED FOR COUPLING AT LAST STAGE

1 BROMO 4 CHLORO BUTANE

WP_000310

IN THE BELOW PIC  2,6-Dichlorobenzaldehyde AND RHODANINE WERE REACTED TO GIVE 2,6-dichlorobenzylidenerhodanine.

2,6-Dichlorobenzaldehyde

RHODANINE

NEXT WAS
2,6-dichlorobenzylidenerhodanine, GAVE (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2-propenoic acid.

1H-NMR (DMSO-d6) d
ppm; 7.23-7.67 (4H, m), 3.5-5.7 (1H, br.), 11.7-14.5 (1H, br.).

Next was prepration of K salt

(Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid and  potassium hydroxide gave ((Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid potassium salt).

Next stage

((Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid potassium
salt) GAVE  2-carboxy-4-chlorobenzo[b]thiophene.
Yield: 48.8 g. 1H-NMR (DMSO-d6) d ppm; 7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3
Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

NEXT IS DECARBOXYLATION

A mixture of 2-carboxy-4-chlorobenzo[b]thiophene, 1,3-dimethyl-2-imidazolidinone, and 1,8-
diazabicyclo[5.4.0]-undec-7-ene GAVE  compound. 4-chlorobenzo[b]thiophene.  1H-NMR (DMSO-d6) d ppm; 7.38 (1H, t, J = 8.4
Hz), 7.51 (1H, dd, J = 5.5, 0.8 Hz), 7.48 (1H, dd, J = 7.7, 0.9 Hz), 7.94 (1H, dd, J = 5.5, 0.4
Hz), 8.02 (1H, dt, J = 8.0, 0.9 Hz).

WP_000309

BETTER REPRESENTATION OF ABOVE PIC

CLIPS FROM PATENT

Synthesis of 2,6-dichlorobenzylidenerhodanine

2,6-Dichlorobenzaldehyde (77.0 g) , rhodanine (58.6 g) , and acetic acid (539 ml) were suspended with stirring at room temperature. Anhydrous sodium acetate (116 g) was added to the suspension, and the resulting mixture was heated under reflux for 3 hours. The reaction mixture was cooled to 45°C, and ice water (700 ml) was added. After the mixture was stirred for 0.2 hours, the precipitated crystals were collected by filtration, washed with water, and then dried to obtain 2,6- dichlorobenzylidenerhodanine. Even in non-dried form, this product could be subjected to the subsequent step.

Yield: 125.4 g^- MR (CDC13) 6ppm;7.30-7.44 (3H, m) , 7.70 (1H. s), 9.6 (1H, br.).

Reference Example 3

• Synthesis of (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2-propenoic acid

A suspension of 2,6- dichlorobenzylidenerhodanine (160.4 g) and water (800 ml) was stirred at room temperature, and sodium hydroxide (83.0 g) was added over a period of 1 hour. The resulting mixture was heated with stirring for another 0.5 hours. The reaction mixture was cooled with ice (10°C), and concentrated hydrochloric acid (192 ml) was added. After the mixture was stirred while cooling with ice for 0.5 hours, the precipitated crystals were collected by filtration. The crystals obtained by filtration were washed with water and then dried to obtain an equivalent amount of (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2- propenoic acid.

Yield: 138.9 g l-NMR (DMSO-de) δρρπΐ;7.23-7.67 (4H, m) , 3.5-5.7 (1H, br.), 11.7-14.5 (1H, br.).

Reference Example 4

• Synthesis of 2-carboxy-4-chlorobenzo[b] thiophene

A suspension of (Z)-3-(2,6-dichlorophenyl-2-mercapto-2- propenoic acid (72.4 g) and water (362 ml) was stirred at room temperature. Further, potassium hydroxide (40.8 g) was added, and the mixture was heated under reflux for 4 hours . After the mixture was allowed to cool, the mixture was stirred for 1 hour while cooling with ice. The precipitated crystals ((Z)-3-(2,6- dichlorophenyl-2-mercapto-2-propenoic acid potassium salt) were collected by filtration and washed with cold water. After the crystals were suspended in water, 35% concentrated hydrochloric acid (32 ml) was added (pH = 1), and the mixture was stirred at room temperature for 1 hour. The precipitated crystals were collected by filtration and dried to obtain 2-carboxy-4- chlorobenzo[b] thiophene.

Yield of 48.8 g ^- MRiDMSO-de) 6ppm; 7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3 Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

Reference Example 5

• Synthesis of K salt  4-chlorobenzo[b] thiophen-2-carboxylate

Reference Example 6

· Synthesis of 2-carboxy-4-chlorobenzo[b]thiophene

Sodium 4-chlorobenzo[b] thiophen-2-carboxylate (2.40 g) was dissolved in water (33 ml) at 60°C. Concentrated hydrochloric acid (1.3 ml) was added to the solution at the same temperature, and the resulting mixture was stirred. The precipitated crystals were collected by filtration, washed with water, and then dried to obtain 2-carboxy-4-chlorobenzo[b] thiophene.

Yield: 1.61 g ^- MR (DMS0-d6);7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3 Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

e

d

Elaborate description

IN THIS BELOW PIC WE SEE

Synthesis of 4-(1-piperazinyl)benzo[b]thiophene

4-Chlorobenzo[b]thiophene and xylene , Subsequently, piperazine, sodium tert-butoxide, palladium acetate (II), and 2-dicyclohexylphosphino-2′,6′-di-iso-propoxy-1,1′-biphenyl (RuPhos) …… producing 4-(1-piperazinyl)benzo[b]thiophene.

NEXT IS PREPARATION OF HYDROCHLORIDE

4-(1-piperazinyl)benzo[b]thiophene hydrochloride. 1H-NMR (DMSO-d6) d ppm;
3.30 (4H, br.s), 3.61 (4H, br.s), 6.97 (1H, d, J = 7.8 Hz), 7.32 (1H, br. dd, J = 8.4, 7.8 Hz),
7.53 (1H, d, J = 5.6 Hz), 7.70 (1H, d, J = 8.4 Hz), 7.76 (1H, d, J = 5.6 Hz), 9.37 (1H, br.s).

NEXT  IS REACTION WITH A TO GIVE BREXPIPRAZOLE

1-benzo[b]thiophen-4-yl-piperazine hydrochloride, potassium carbonate
and DMF  and  7-(4-chlorobutoxy)-1H-quinolin-2-one A  FROM PIC 1 and potassium iodide  GAVE BREXPIPRAZOLE, ie 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one.

1H-NMR (DMSO-d6) d ppm; 1.6-1.75 (2H, m), 1.75-1.9 (2H, m), 2.44
(2H, t, J = 7.0 Hz), 2.55-2.70 (4H, m), 3.00-3.15 (4H, m), 4.06 (2H, t, J = 6.3 Hz), 6.30 (1H,
d, J = 9.5 Hz), 6.75-6.85 (2H, m), 6.88 (1H, d, J = 7.5 Hz), 7.27 (1H, dd, J = 8 Hz, 8 Hz),
7.40 (1H, d, J = 5.5 Hz), 7.55 (1H, d, J = 9.5 Hz), 7.61 (1H, d, J = 8 Hz), 7.69 (1H, d, J = 5.5
Hz), 7.80 (1H, d, J = 9.5 Hz), 11.58 (1H, bs).

WP_000308

BETTER REPRESENTATION OF  PIC

Example 2

• Synthesis of 4- (l-piperazinyl)benzo[b]thiophene hydrochloride

4-Chlorobenzo[b] thiophene (5.00 g), piperazine (5.11 g) , palladium acetate (II) (2.7 mg), tri-tert-butylphosphonium

tetraphenylborate (6.2 mg), sodium tert-butoxide (8.548 g), and xylene (70 ml) were stirred at 120 to 130°C for 5 hours. After the reaction mixture was cooled to room temperature, water was added thereto, and the layers were separated. The xylene layer was washed with water, and then with saline. After addition of activated carbon, the mixture was stirred at room temperature for 30 minutes. After filtration of the mixture, concentrated

hydrochloric acid was added to the filtrate, and the resulting mixture was stirred at room temperature for 30 minutes. The precipitated crystals were collected by filtration and dried to obtain 4- ( l-piperazinyl)benzo[b] thiophene hydrochloride.

Yield: 6.94 g !H-NMRiDMSO-de) 6ppm; 3.30 (4H, br.s), 3.61 (4H, br.s), 6.97 (1H, d, J= 7.8 Hz), 7.32 (1H, br.dd, J= 8.4. 7.8 Hz), 7.53 (1H, d, J= 5.6 Hz), 7.70 (1H, d, J= 8.4 Hz), 7.76 (1H, d, J= 5.6 Hz), 9.37 (1H, br.s).

Example 3

• Synthesis of 4- ( 1-piperazinyl)benzo[b] thiophene hydrochloride

4-Chlorobenzo[b] thiophene (10.0 g) and xylene (100 ml) were placed in a reaction vessel. The reaction vessel was

evacuated and then purged with argon. Subsequently, piperazine (15.3 g) , sodium tert-butoxide (17.1 g) , palladium acetate (II) (13.0 mg) , and 2-dicyclohexylphosphino-2′,6′-di-iso-propoxy-1,1′- biphenyl (RuPhos) (69.0 mg) were added. After evacuation and purging with argon, the mixture was refluxed for 2 hours. After the reaction mixture was cooled to about 80°C, water (50 ml) and silica #600H (0.65 g) were added. The mixture was stirred at approximately 60°C for about 10 minutes, and then filtered. After the filtrate was separated into layers, the xylene layer was washed with water. Subsequently, the xylene layer was placed into the reaction vessel again. After addition of water (200 ml) and concentrated hydrochloric acid (8.0 ml) , the mixture was heated with stirring for dissolution. The layers were separated at 75°C or more. After the aqueous layer was collected, toluene (150 ml) and 25% aqueous sodium hydroxide solution (16 ml) were added, and the mixture was stirred. The layers were separated, and the organic layer was collected. The organic layer was washed with water and concentrated with an evaporator. Methanol (150 ml) was added to the concentrated oil to dissolve the oil, thus producing a methanol solution. 2-Propanol (150 ml) and concentrated

hydrochloric acid (7 ml) were placed into another reaction vessel, and the methanol solution was added dropwise over a period of 15 minutes or more. After completion of the dropwise addition, the mixture was cooled and stirred at 10°C or less for about 30 minutes, and then filtered (washed with a mixture of 5 ml of methanol and 5 ml of 2-propanol) . The crystals were collected, and then dried to obtain 4-(l-piperazinyl)benzo[b]thiophene hydrochloride.

Yield: 11.61 g

^-NMRfDMSO-de) oppm;

3.25-3.40 (8H, br.s), 6.96 (1H, d, J = 7.5 Hz), 7.32 (1H, dd, J = 8.0, 7.5 Hz), 7.52 (1H, d, J = 5.5 Hz ) . 7.70 (1H, d, J = 8.0 Hz), 7.75 (1H, d, J = 5.5 Hz), 9.35 (1H, br.s).

Reference Example 9

· Synthesis of 7- ( 4-chlorobutoxy) -lH-quinolin-2-one

After 7-hydroxy-lH-quinolin-2-one (10 g) and DMF (50 ml) were heated to approximately 30°C, an aqueous potassium carbonate solution (potassium carbonate: 8.6 g, water: 10 ml) was added. After the mixture was stirred at 30 to 40°C for about 15 minutes, l-bromo-4-chlorobutane (14.3 ml) was added and stirred at approximately 40°C for 5 hours. Water (100 ml) was added dropwise over a period of 30 minutes or more while the

temperature was maintained at 30°C or more. After the mixture was stirred at approximately 30°C for 30 minutes, stirring was continued at 10°C or less for 1 hour, after which the precipitated crystals were collected by filtration. After methanol (100 ml) was added to the precipitated crystals, the mixture was stirred under reflux to ensure dissolution. This solution was cooled and stirred at 30 to 40°C for 30 minutes and then at 5°C or less for about 1 hour, after which the precipitated crystals were

collected by filtration. The crystals were dried at 60°C to obtain 7- (4-chlorobutoxy) -lH-quinolin-2-one as white powder.

Yield: 12.3 g

^I-NMR (300 MHz; CDC13) oppm; 1.95-2.05 (4H, m) , 3.64 (2H, t, J = 6.0Hz), 4.10 (2H, t. J = 5.5 Hz), 6.56 (1H, d, J = 9.5 Hz), 6.80 (1H. dd, J = 9.0 Hz, 2.5 Hz), 6.84 (1H, d, J = 2.5 Hz), 7.45 (1H, d, J = 9.0 Hz), 7.73 (1H, d, J = 9.5 Hz), 12.45 (1H, brs).

Example 4

· Synthesis of 7- [4- (4-benzo[b]thiophen-4-yl-piperazin-l- yl)butoxy] -lH-quinolin-2-one

After 1-benzo[b] thiophen-4-yl-piperazine hydrochloride (10.6 g), potassium carbonate (5.8 g) , and DMF (50 ml) were stirred at 30 to 40°C for about 30 minutes, 7-(4-chlorobutoxy) -1H- quinolin-2-one (10.0 g) and potassium iodide (6.9 g) were added. The mixture was stirred at 90 to 100°C for 2 hours. While the temperature of the mixture was maintained at 60°C or more, water (150 ml) was added dropwise over a period of 10 minutes or more.

After the mixture was cooled to 10°C or less, the precipitated crystals were collected by filtration, and washed with water and then with ethanol.

After ethanol (325 ml) and acetic acid (25 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. Concentrated hydrochloric acid (3.6 ml) was added at around 70°C, and the mixture was cooled. After confirming the precipitation of crystals, the mixture was heated again and stirred under reflux for 1 hour. After the mixture was cooled to 10°C or less, the precipitated crystals were collected by filtration and washed with ethanol.

After ethanol (191 ml) and water (127 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. After activated carbon (0.89 g) was added, the mixture was stirred under reflux for 30 minutes and then hot filtered. After activated carbon was removed, the mixture was heated again for dissolution. After 25% aqueous sodium hydroxide solution (5.8 ml) was added at approximately 70°C, the mixture was stirred under reflux for 30 minutes, after which water (64 ml) was added at approximately 70°C. After the mixture was stirred at 40°C for 30 minutes, the precipitated crystals were collected by filtration at 40°C or less, then washed with water, and dried to obtain 7- [4-(4-benzo[b]thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one as white crystals.

Yield: 14.30 g ^-NMRfDMSO-de) 6ppm; 1.6-1.75 (2H, m) . 1.75-1.9 (2H, m) , 2.44 (2H, t, J = 7.0 Hz),2.55-2.70 (4H, m) , 3.00-3.15 (4H, m) , 4.06 (2H, t, J = 6.3 Hz), 6.30 (1H, d, J = 9.5 Hz), 6.75-6.85 (2H, m) , 6.88 (1H, d, J = 7.5 Hz), 7.27 (1H, dd, J = 8 Hz, 8 Hz), 7.40 (1H, d, J = 5.5 Hz), 7.55 (1H, d, J = 9.5 Hz), 7.61 (1H, d, J = 8 Hz), 7.69 (1H, d, J = 5.5 Hz), 7.80 (1H, d, J = 9.5 Hz), 11.58 (1H, bs) .

c

  logo

SEE  http://www.molbase.com/en/index.html

IH NMR PREDICT

7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one NMR spectra analysis, Chemical CAS NO. 913611-97-9 NMR spectral analysis, 7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one H-NMR spectrum

  logo

13 C NMR PREDICT

7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one NMR spectra analysis, Chemical CAS NO. 913611-97-9 NMR spectral analysis, 7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one C-NMR spectrum

Patent

Reaction Scheme 3:

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=D842B4D68D66F641E505E9690CF876D0.wapp2nB?docId=WO2015054976&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

 

Wherein, X is halogen, such as fluorine, chlorine, bromine, iodine; R and R 1 as defined above in the definition of the compounds of formula I the same;
Scheme 4:

Wherein, X is fluorine, chlorine, bromine or iodine; R 1 as defined above, with a compound of formula I as defined for the same;
Reaction Scheme 5:
Wherein, X is fluorine, chlorine, bromine or iodine; R 1 is the same as defined in the compounds shown above, and R are as defined for formula I. The present invention also provides processes for preparing key intermediates Bray prazosin/  Brexpiprazole or a salt thereof, the method as shown in Scheme 6:
Scheme 6:
Example 26
7- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone
Preparation of

The product (400mg, 0.83mmol) of Example 25 will be implemented, silver carbonate (46mg, 0.16mmol) was dissolved in DMSO (5mL) and the acetic acid was heated to 120 ℃ overnight. Cooling, water was added, extracted with ethyl acetate, ethyl acetate layer was washed with saturated sodium bicarbonate and brine each wash again, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (80mg, yield 22%).
1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1] + = 436.3.

Example 27
7- [4- (2-carboxy-benzothiophen-4-yl-1-piperazinyl) butoxy] -2 (1H) – quinolinone
Preparation of

A mixture of 2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added sodium hydroxide (29mg, 0.73mmol) and thioglycolic acid (0.025mL, 0.36mmol), 120 ℃ stirred for 16 hours. Cooling, water was added, adjusted with 1N HCl aqueous solution is about pH = 5, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (40mg, yield 46 %).
ESI: [M + 1] + = 478.0.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl)
Example 28 1-

The product of Example 17 (100mg, 0.25mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5 mL) in, 100 ℃ stirred for 10 hours. The reaction solution was poured into ice water, stirred for 10min after filtration, to obtain the target substance (38mg, 50% yield).

1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
Preparation of tert-butyl piperazine-1 – Example 224- (2-carboxy-benzothiophen-4-yl)

Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium methoxide (133mg, 2.45mmol ), and the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (130mg, 58% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – (2-carboxy-benzothiophen-4-yl) Example 234-

Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium hydroxide (99mg, 2.45 mmol), the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (180mg, yield 81%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.

Example 24 7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of

A mixture of 2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added DIPEA (94mg, 0.73mmol) and ethyl mercaptoacetate (0.024mL, 0.22mmol), 110 ℃ stirred for 16 hours. Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (40mg, 46% yield).
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 184- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen, was added at room temperature to DMF (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), DIPEA (342mg, 2.48mmol), the mixture was at 105 ℃ stirred for 18 hours, 1N HCl aqueous solution was added adjust pH = 7, and extracted with methyl tert-butyl ether, the ether layer was washed three times with saturated brine, dried over anhydrous sodium sulfate, the drying agent was filtered off, and concentrated by column chromatography to obtain the target (170mg, yield 71%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.

Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 194- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added the product of Reference Example 1 to ethanol (5mL) inside (200mg, 0.62mmol), ethyl mercaptoacetate (0.081ml, 0.74mmol), sodium hydroxide (100mg, 2.48mmol), the mixture 85 ℃ stirred for 6 hours, and concentrated by column chromatography to obtain the target substance (70mg, 30% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 201-

The product of Example 2 (200mg, 0.55mmol), was dissolved in THF (5mL) was added concentrated hydrochloric acid (0.5mL), 50 ℃ heated 6h.Cooling, methyl tert-butyl ether (5mL), filtered to give the target (130mg, yield 79%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.

Piperazine hydrochloride – (benzothiophen-4-yl) Example 211-

The product of Example 20 (130mg, 0.43mmol) was added to diphenyl ether (3mL) in, 260 ℃ heating 0.5h. Cooling, filtration object (60mg, 55% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.

PAPER

Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00027
Figure

Figure 1. Brexpiprazole (1) and intermediate 18.

Abstract Image

2-Chloro-6-fluorobenzaldehyde was converted to 4-(1-piperazinyl)benzo[b]thiophene dihydrochloride (18), an intermediate in the synthesis of brexpiprazole, via a five-step sequence in 54% overall yield. This procedure requires no expensive catalyst and avoids the side products produced in the coupling step in the reported process. Several kilograms of compound 18 were prepared using this economical and scalable process.

1-(Benzo[b]thiophen-4-yl)piperazine Dihydrochloride (18)

Compound 10 (1.5 kg, 4.71 mol) was dissolved in ………………..DELETED…………………, and then dried to give compound 18 (1.17 kg, 85% yield). HPLC for compound 18 (tR = 6.3 min, identical to authentic sample) 99.8% purity; HPLC method B.
18:
1H NMR (400 MHz, DMSO-d6) δ 11.86 (s, 1H), 9.65 (s, 2H), 7.75 (d, J = 5.5 Hz, 1H), 7.69 (d, J = 8.1 Hz, 1H), 7.53 (d, J = 5.5 Hz, 1H), 7.30 (t, J = 7.9 Hz, 1H), 6.96 (d, J = 7.6 Hz, 1H), 3.30 (s, 8H).
13C NMR (100 MHz, DMSO-d6): δ 146.92, 140.62, 133.40, 126.50, 125.06, 121.91, 117.73, 112.56, 48.52, 43.00.
MS (ESI, eV): m/z = 219.1 [M + H]+.

 ………..

PATENT

http://www.google.com/patents/US20140187782

A 4-(1-piperazinyl)benzo[b]thiophene compound represented by Formula (1):

Figure US20140187782A1-20140703-C00002

is useful for various medicines such as antipsychotic drugs. Moreover, a 4-(1-piperazinyl)benzo[b]thiophene compound represented by Formula (4):

Figure US20140187782A1-20140703-C00003

wherein R1 is a hydrogen atom or a protecting group, is useful as an intermediate for synthesizing the compound represented by Formula (1).

Reference Example 30 and Example 1 of PTL 1 specifically disclose a method for producing a benzo[b]thiophene compound (the reaction scheme shown below). In Reference Example 30, 4-(1-piperazinyl)benzo[b]thiophene is produced by heating under reflux a mixture comprising 14.4 g of 4-bromobenzo[b]thiophene, 29.8 g of anhydrous piperazine, 9.3 g of sodium tert-butoxide, 0.65 g of (R)-(+)-2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (BINAP), 0.63 g of tris(dibenzylideneacetone)dipalladium (0), and 250 ml of toluene (step X).

Figure US20140187782A1-20140703-C00004

However, the reaction of the step X produces a relatively large amount of by-products that can hardly be separated, and the purity of the compound (4a) is thus inevitably reduced. Moreover, although column purification is performed to increase the purity of the compound (4a), it is very difficult to completely remove by-products, even by column chromatography purification. For this reason, there is a demand for the development of a novel method for producing the compound (4a) with high yield and high purity.

Furthermore, by-products contained in the compound (4a) inevitably reduce the purity of the compound (1) in the subsequent step Y. Since the method described in PTL 1 requires purification by column chromatography to obtain the target compound (1) with high purity, the method is not suitable for the industrial process of mass production. In addition, according to the method, incorporation of by-products that can hardly be separated is inevitable, and high-purity products usable as active pharmaceutical ingredients cannot be produced without purification by column chromatography.

CITATION LISTPatent Literature

  • PTL 1: Japanese Unexamined Patent Publication No. 2006-316052 Non Patent Literature
  • NPL 1: Tetrahedron Lett., 2004, 45, 9645

Figure US20140187782A1-20140703-C00020

Figure US20140187782A1-20140703-C00021

Figure US20140187782A1-20140703-C00022

Example 4

Synthesis of 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one

After 1-benzo[b]thiophen-4-yl-piperazine hydrochloride (10.6 g), potassium carbonate (5.8 g), and DMF (50 ml) were stirred at 30 to 40° C. for about 30 minutes, 7-(4-chlorobutoxy)-1H-quinolin-2-one (10.0 g) and potassium iodide (6.9 g) were added. The mixture was stirred at 90 to 100° C. for 2 hours. While the temperature of the mixture was maintained at 60° C. or more, water (150 ml) was added dropwise over a period of 10 minutes or more. After the mixture was cooled to 10° C. or less, the precipitated crystals were collected by filtration, and washed with water and then with ethanol.

After ethanol (325 ml) and acetic acid (25 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. Concentrated hydrochloric acid (3.6 ml) was added at around 70° C., and the mixture was cooled. After confirming the precipitation of crystals, the mixture was heated again and stirred under reflux for 1 hour. After the mixture was cooled to 10° C. or less, the precipitated crystals were collected by filtration and washed with ethanol.

After ethanol (191 ml) and water (127 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. After activated carbon (0.89 g) was added, the mixture was stirred under reflux for 30 minutes and then hot filtered. After activated carbon was removed, the mixture was heated again for dissolution. After 25% aqueous sodium hydroxide solution (5.8 ml) was added at approximately 70° C., the mixture was stirred under reflux for 30 minutes, after which water (64 ml) was added at approximately 70° C. After the mixture was stirred at 40° C. for 30 minutes, the precipitated crystals were collected by filtration at 40° C. or less, then washed with water, and dried to obtain 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one as white crystals.

Yield: 14.30 g

1H-NMR (DMSO-d6) δ ppm;

1.6-1.75 (2H, m), 1.75-1.9 (2H, m), 2.44 (2H, t, J=7.0 Hz), 2.55-2.70 (4H, m), 3.00-3.15 (4H, m), 4.06 (2H, t, J=6.3 Hz), 6.30 (1H, d, J=9.5 Hz), 6.75-6.85 (2H, m), 6.88 (1H, d, J=7.5 Hz), 7.27 (1H, dd, J=8 Hz, 8 Hz), 7.40 (1H, d, J=5.5 Hz), 7.55 (1H, d, J=9.5 Hz), 7.61 (1H, d, J=8 Hz), 7.69 (1H, d, J=5.5 Hz), 7.80 (1H, d, J=9.5 Hz), 11.58 (1H, bs).

………………………

PATENT

http://www.google.com/patents/WO2006112464A1?cl=en

Example 1

Preparation of 7- [4- (4-benzo [b] thiophen-4-yl- piperazin-1-yl) butoxy] -lH-quinolin-2-one

A mixture of 9.0 g of 7- ( 4-chlorobutoxy) -IH- quinolin-2-one, 10 g of 1-benzo [b] thiophene-4-yl- piperazine hydrochloride, 14 g of potassium carbonate, 6 g of sodium iodide and 90 ml of dimethylformamide was stirred for 2 hours at 😯0C. Water was added to the reaction solution and precipitated crystals were separated by filtration. The crystals were dissolved in a mixed solvent of dichloromethane and methanol, dried over magnesium sulfate, and the solvent was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane .-methanol = 100:3). Recrystallized from ethanol, 13.6 g of 7- [4- (4-benzo [b] thiophen-4-yl- piperazin-1-yl) butoxy] -lH-quinolin-2-one in the form of a white powder was obtained.

Melting point 183.5-184.50C

1H-NMR ( DMSO-dg) δppm:

1.6-1.75 (2H, m) , 1.75-1.9(2H, m) , 2.44(2H, t, J=7Hz) , 2.5-2.8(4H, m) , 2.9-3.2(4H, m) , 4.06(2H, t, J=6.5Hz), 6.3O(1H, d, J=9.5Hz), 6.75-6.85 (2H, m) , 6.88(1H, d, J=7.5Hz), 7.27 (IH, dd, J=8Hz, 8Hz), 7.40 (IH, d, J=5.5Hz), 7.55 (IH, d, J=9.5Hz), 7.61(1H, d, J=8Hz) , 7.69(1H, d, J=5.5Hz), 7.8O(1H, d, J=9.5Hz), 11.59(1H, bs) .

……………….

PATENT

Figure imgf000006_0001 7- [ 4- ( 4-benzo[b]thiophen-4- yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one

The dihydrate of the benzothiophene compound represented by Formula (I) or of a salt thereof according to the present invention can be produced from an anhydride of the benzothiophene compound or of a salt thereof.

The benzothiophene compound (in the form of an

anhydride) of Formula (I), from which the dihydrate of the present invention is produced, is a known compound, and can be obtained by the production method disclosed in Example 1 of

JP2006-316052A or according to Reference Examples 1 and 2

Fig. 1 shows the ^-NMR spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 2 shows the X-ray powder diffraction pattern of the dihydrate of the benzothiophene compound represented by

Formula (I) prepared in Example 1.

Fig. 3 shows the infrared absorption spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 4 shows the Raman spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 5 shows the XH- MR spectrum of the benzothiophene compound represented by Formula (I) prepared in Example 2.

Reference Example 1: Synthesis of 7-(4-chlorobutoxy)-lH-quinolin- 2-one Methanol (149 L) , 7-hydroxy-lH-quinolin-2-one (14.87 kg), and potassium hydroxide (6.21 kg) were mixed and stirred. After dissolution, l-bromo-4-chlorobutane (47.46 kg) was further added thereto and the resulting mixture was stirred under reflux for seven hours. Thereafter, the mixture was stirred at 10° C for one hour. The precipitated crystal was centrifuged and washed with methanol (15 L). The wet crystal was collected and placed in a tank. Water (149 L) was added thereto, followed by stirring at room temperature. After centrifugation, the resulting solid was washed with water (30 L). The wet crystal was collected and placed in a tank. After adding methanol (74 L), the mixture was stirred under reflux for one hour, cooled to 10° C, and then stirred. The precipitated crystal was centrifuged and washed with methanol (15 L). The separated crystal was dried at 60° C to obtain 7- (4-chlorobutoxy) -lH-quinolin-2-one (15.07 kg).

Reference Example 2: Synthesis of 7- [ 4- ( 4-benzo[b] thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one

Water (20 L), potassium carbonate (1.84 kg), 1- benzo[b] thiophen-4-yl-piperazine hydrochloride (3.12 kg), and ethanol (8 L) were mixed and stirred at 50° C. 7- ( 4-Chlorobutoxy) – lH-quinolin-2-one (2.80 kg) obtained in Reference Example 1 was added to the mixture and stirred under reflux for nine hours.

After concentrating the solvent (8 L) under ordinary pressure, the mixture was stirred at 90° C for one hour and then cooled to 9° C . The precipitated crystal was centrifuged and then

sequentially washed with water (8 L) and ethanol (6 L). The separated crystal was dried at 60° C to obtain a crude product. The crude product (4.82 kg) and ethanol (96 L) were mixed in a reaction vessel, and acetic acid (4.8 L) was introduced into the reaction vessel. The mixture was stirred under reflux for one hour to dissolve the crude product. After introducing

hydrochloric acid (1.29 kg), the mixture was cooled to 10° C. The mixture was heated again, refluxed for one hour, and cooled to 7° C. The precipitated crystal was centrifuged and washed with ethanol (4.8 L). The separated crystal was dried at 60° C to obtain 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one hydrochloride (5.09 kg). The resulting 7- [4- (4- benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy] -lH-quinolin-2-one hydrochloride (5.00 kg), ethanol (45 L), and water (30 L) were mixed in a reaction vessel. The mixture was stirred under reflux to dissolve the 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-l- yl)butoxy] -lH-quinolin-2-one hydrochloride. Activated carbon (500 g) and water (5 L) were added thereto, and an activated carbon treatment was conducted under reflux for 30 minutes. After performing hot filtration, a solution containing sodium hydroxide (511 g) dissolved in water (1.5 L) was flowed into the reaction vessel while stirring the filtrate under reflux. After stirring under reflux for 30 minutes, water (10 L) was introduced thereto and the mixture was cooled to approximately 40° C. The

precipitated crystal was centrifuged and washed with water (125 L). The separated crystal was dried at 80° C to obtain 7- [4- (4- benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy] – lH-quinolin-2-one (3.76 kg).

Example 1: Preparation of 7- [ 4- ( 4-benzo[b]thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate

The 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-1- yl)butoxy] -lH-quinolin-2-one (3.2 kg) obtained in Reference

Example 2, ethanol (64 L) , water (74 L) , and acetic acid (1.77 kg) were mixed in a reaction vessel to prepare an acidic liquid mixture. The mixture was stirred under reflux to dissolve the 7- [ 4- ( 4-benzo[b] thiophen-4-yl-piperazin-1-yl)butoxy] -1H-quinolin-2- one (reflux temperature: 84° C). After cooling to -5°C, the solution obtained above was introduced, over a period of 30 minutes, into a solution containing 25% sodium hydroxide (5.9 kg) and water (54 L) that was cooled to 0°C, to prepare a liquid mixture with pHlO. After being stirred at 5° C or below for one hour, the mixture was heated to 20 to 30° C and further stirred for-seven hours . The precipitated crystal was filtered and washing with water (320 L) was performed, until alkali in the solid component disappeared (i.e.. until the pH value of the filtrate became 7 ) . The solid component was then air-dried until its weight became constant to obtain a white solid 7-[4-(4- benzofb] thiophen-4-yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate (unground, 3.21 kg).

Fig. 1 shows the XH-NMR spectrum (D SO-d6, TMS) of the dihydrate prepared by the aforesaid method. As shown in Fig. 1, in the ^- MR spectrum (DMSO-d6, TMS) , peaks were observed at 1.64 ppm (tt, J = 7.4 Hz, J = 7.4 Hz, 2H) , 1.80 ppm (tt, J = 7.0 Hz, J = 7.0 Hz, 2H), 2.44 ppm (t, J = 7.5 Hz, 2H) , 2.62 ppm (br, 4H) , 3.06 ppm (br, 4H) , 3.32 ppm (s, 4H + H20) , 4.06 ppm (t, J = 6.5 Hz, 2H), 6.29 ppm (d, J = 9.5 Hz, 1H), 6.80 ppm (d, J = 2.5 Hz, 1H) , 6.80 ppm (dd, J = 2.5 Hz, J = 9.0 Hz, 1H) , 6.88 ppm (d, J = 7.5 Hz, 1H), 7.27 ppm (dd, J = 7.8 Hz, J = 7.8 Hz, 1H) , 7.40 ppm (dd, J = 0.5 Hz, J = 5.5 Hz, 1H), 7.55 ppm (d, J = 9.0 Hz, 1H) , 7.61 ppm (d, J = 8.0 Hz, 1H) , 7.69 ppm (d, J = 5.5 Hz, 1H) , 7.80 ppm (d, J = 9.5 Hz, 1H), and 11.57 ppm (s, 1H) .

The X-ray powder diffraction spectrum of the dihydrate prepared by the aforesaid method was measured using an X-ray diffractometer (D8 ADVANCE, available from Bruker AXS). Fig. 2 shows the X-ray powder diffraction spectrum. As shown in Fig. 2, in the X-ray powder diffraction spectrum, diffraction peaks were observed at 2Θ = 8.1° , 8.9° , 15.1° , 15.6° , and 24.4° . Other than those mentioned above, the diffraction peaks were also observed at 2Θ = 11.6°.. 12.2°, 14.0°, 16.3°, 18.1°, 18.4°, 18.9°, 19.5°, 20.5°, 21.5°, 22.6°, 23.3°, 25.0°, 26.1°, 26.4°, 27.1°. 28.1°, 28.5°, 28.9°, 29.8°, 30.4°, 30.7°, 31.6°, 32.9°, 33.9°, 34.4°, 35.2°, 36.0°, 36.7°, 37.4° , and 38.3°.

The IR (KBr) spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 3 shows the IR (KBr) spectrum. As shown in Fig. 3, in the IR (KBr) spectrum, absorption bands were observed in the vicinity of wavenumbers 3509 cm“1, 2934 cm“1, 2812 cm“1, 1651 cm“1, 1626 cm“1, 1447 cm“1, 1223 cm“1 and 839 cm“1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 4 shows the Raman spectrum. As shown in Fig. 4, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1497 cm“1, 1376 cm“1, 1323 cm“1, 1311 cm“1, 1287 cm“1, 1223 cm“1, and 781 cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1613 cm“1, 1563 cm“1, 1512 cm“1, 1468 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1, 1096 cm“1, 1070 cm“1, 971 cm“1, and 822 cm“1.

The water content of the dihydrate prepared by the aforesaid method was measured using a moisture meter (CA-100, available from Mitsubishi Chemical Analytech Co., Ltd.) by the Karl Fischer method. As a result, the dihydrate had a water content of 7.79% by weight.

Example 2; Preparation of finely ground dihydrate

Dihydrate crystal (2.73 kg) obtained in Example 1 was ground using a jet mill. Here, the air pressure was set at 5 kgf/cm2, and the rotational speed of the feeder was set at 20 rpm. As a result, finely ground 7-[4-(4-benzo[b]thiophen-4-yl- piperazin-1-yl)butoxy] -1H-quinoli -2-one dihydrate (2.61 kg,

95.6%) was obtained.

The dihydrate (finely ground product) thus obtained had a mean particle diameter of 5.5 um. The mean particle diameter was measured using a Microtrack HRA, manufactured by Nikkiso Co., Ltd.

Fig. 5 shows the ^-NMR spectrum (DMSO-d6, TMS) of the dihydrate prepared by the above method. As shown in Fig. 5, in the ^- MR spectrum (DMSO-d6, TMS), peaks were observed at 1.64 ppm (tt, J = 7.3 Hz, J = 7.3 Hz, 2H), 1.80 ppm (tt, J = 6.9 Hz, J = 6.9 Hz, 2H), 2.44 ppm (t, J = 7.3 Hz, 2H) , 2.62 ppm (br, 4H) , 3.06 ppm (br, 4H) , 3.32 ppm (s, 4H + H20) , 4.06 ppm (t, J = 6.5 Hz, 2H), 6.29 ppm (d, J = 9.5 Hz, 1H) , 6.80 ppm (d, J = 2.5 Hz , 1H) , 6.80 ppm (dd, J = 2.3 Hz, J = 9.3 Hz, 1H) , 6.88 ppm (d, J = 7.5 Hz, 1H), 7.27 ppm (dd, J = 8.0 Hz, J = 8.0 Hz, 1H) , 7.40 ppm (d, J = 5.5 Hz, 1H), 7.55 ppm (d, J = 9.5 Hz , 1H) , 7.61 ppm (d, J = 8.0 Hz, 1H), 7.69 ppm (d, J = 5.5 Hz, 1H) , 7.80 ppm (d, J = 9.5

Hz, 1H), and 11.57 ppm (s, 1H) .

The X-ray powder diffraction spectrum of the dihydrate prepared by the aforesaid method was measured in the same manner as in Example 1. Fig. 6 shows the X-ray powder diffraction spectrum. As shown in Fig. 6, in the X-ray powder diffraction spectrum, diffraction peaks were observed at 2Θ = 8.2° , 8.9° ,

15.2° , 15.7° and 24.4° .

Other than those mentioned above, the diffraction peaks were also observed at 2Θ = 6.8°, 12.2°, 14.0°, 14.5″, 17.4°,

18.1°, 18.5°, 19.0°, 19.2°, 19.6°, 20.3°, 20.6°, 21.5°, 22.7°,

23.4°, 25.0°, 26.1°, 27.1°, 28.6°, 29.0°, 30.4°, 34.0°, 34.5°,

35.3° , and 36.7° .

The IR (KBr) spectrum of the dihydrate prepared by the aforesaid method was measured in the same manner as in Example 1.

Fig. 7 shows the IR (KBr) spectrum. As shown in Fig. 7, in the IR

(KBr) spectrum, absorption bands were observed in the vicinity of wavenumbers 3507 cm“1, 2936 cm“1, 2812 cm“1, 1651 cm“1, 1626 cm“1,

1447 cm“1, 1223 cm“1 and 839 cm“1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 8 shows the Raman spectrum.

As shown in Fig. 8, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1496 cm‘1, 1376 cm“1, 1323 cm‘1, 1311 cm“1, 1286 cm“1, 1223 cm“1, and 781cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1614 cm“1, 1563 cm“1, 1512 cm“1, 1467 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1,

1095 cm“1, 1069 cm“1, 971 cm“1, and 822 cm“1.

The water content of the dihydrate prepared by the aforesaid method was measured using a moisture meter (CA-100, available from Mitsubishi Chemical Analytech Co., Ltd.) by the

Karl Fischer method. As a result, the dihydrate had a water content of 6.74% by weight . Example 3 : Preparation of 7- [ 4- ( 4-benzo[b] thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate

7- [ 4- ( 4-Benzo[ ] thiophen-4-yl-piperazin-1-yl)butoxy] – lH-quinolin-2-one (5.0 kg), ethanol (100 L), water (115 L), and DL-lactic acid (2.29 kg) were mixed to prepare an acidic liquid mixture. The liquid mixture was stirred under reflux to dissolve the 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one (reflux temperature: 82° C). After cooling to -5°C, the solution obtained above was introduced, over a period of about 15 minutes, into a solution containing sodium hydroxide (1.48 kg) and water (135 L) that was cooled to 1°C, to prepare a liquid mixture with pHll. After being stirred at approximately 2 to 5° C for three hours, the mixture was heated to 45° C and

further stirred at 45 to 50° C for two hours. The precipitated crystal was filtered and washing with water (200 L) was performed until alkali in the solid component disappeared (i.e., until the pH value of the filtrate became 7). The solid component was further washed with a liquid mixture of ethanol (15 L) and water (20 L). The solid component was then dried at room temperature until its weight became constant to obtain a white solid 7- [4- (4- benzo[b] thiophen-4-yl-piperazin-1-yl)butoxy] -1H-quinolin-2-one dihydrate (unground, 5.11 kg).

The dihydrate thus obtained was the same as that obtained in Example 1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 9 shows the Raman spectrum. As shown in Fig. 9, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1497 cm“1, 1376 cm“1, 1323 cm“1, 1311 cm“1, 1287 cm“1, 1223 cm“1, and 782 cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1614 cm“1, 1563 cm“1, 1512 cm“1, 1468 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1, 1126 cm“1, 1096 cm“1, 1070 cm“1, 972 cm“1, and 822 cm“1.

…………………….

PATENT

http://www.google.com/patents/WO2006112464A1?cl=en

…………………..

PATENT

http://www.google.com/patents/US20110152286

References

  1.  “Phase II and Phase III Drugs in U.S. Development for Depression, Anxiety, Sleep Disorders, Psychosis, & ADHD”. Retrieved 9 February 2012.
  2.  “Otsuka Pharmaceutical Development & Commercialization, Inc.”. Bloomberg Businessweek. Retrieved 10 February 2012.
  3.  “Otsuka HD places top priority on development of OPC-34712.”. Chemical Business Newsbase. January 3, 2011. Retrieved 10 February 2012.
  4. Patent 5006528, Oshiro, Yasuo; Seiji Sato & Nobuyuki Kurahashi, “Carbostyril derivatives”, published October 20, 1989
  5. “Patent and Exclusivity Search Results”. Electronic Orange Book. US Food and Drug Administration. Retrieved 8 December 2008.
  6.  “Lundbeck and Otsuka Pharmaceutical sign historic agreement to deliver innovative medicines targeting psychiatric disorders worldwide”. Lundbeck. Retrieved 10 February2012.
  7.  “Otsuka Holdings Financial Results Presentation Q2 FY2011”. Retrieved 10 February2012.
  8.  “OPC-34712 search results”. Retrieved 10 February 2012.
  9.  “Study of the Safety and Efficacy of OPC-34712 as Adjunctive Therapy in the Treatment of Patients With Major”. Retrieved 15 February 2012.
  10.  “Study of the Safety and Efficacy of Two Fixed Doses of OPC-34712 as Adjunctive Therapy in the Treatment of Adults With Major Depressive Disorder (the Polaris Trial)”. Retrieved 10 February 2012.
  11. ^ Jump up to:a b “Study of the Safety and Efficacy of OPC-34712 as a Complementary Therapy in the Treatment of Adult Attention Deficit/Hyperactivity Disorder (STEP-A)”. Retrieved10 February 2012.
  12.  “Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder”. Retrieved 10 February 2012.
  13.  “A Dose-finding Trial of OPC-34712 in Patients With Schizophrenia”. Retrieved10 February 2012.
  14.  “Efficacy Study of OPC-34712 in Adults With Acute Schizophrenia (BEACON)”. Retrieved 10 February 2012.
  15. Jump up^ “Safety and Tolerability Study of Oral OPC-34712 as Maintenance Treatment in Adults With Schizophrenia (ZENITH)”. Retrieved 10 February 2012.
  16.  “Study of the Effectiveness of Three Different Doses of OPC-34712 in the Treatment of Adults With Acute Schizophrenia (VECTOR)”. Retrieved 10 February 2012.
  17.  “A Long-term Trial of OPC-34712 in Patients With Schizophrenia”. Retrieved10 February 2012.
  18.  “Otsuka Pharmaceutical Co., Ltd. Announces Results from a Phase 2 Study of Investigational Product OPC-34712 as Adjunctive Therapy in Adults with Major Depressive Disorder”. Retrieved 16 February 2012.
  19.  “Preclinical Pharmacology of Brexpiprazole (Opc-34712): A Novel Compound with Dopamine D2 Receptor Partial Agonist Activity”. Retrieved 16 February 2012.
  20.  “2011 U.S. Psych Congress Poster Session Abstracts”. Retrieved 16 February 2012.
  21.  “Otsuka Pharmaceutical reports OPC-34712 Phase 2 trial results in major depressive disorder”. News-Medical.Net. Retrieved 10 February 2012.
  22.  Maeda K, Sugino H, Akazawa H et al. (September 2014). “Brexpiprazole I: in vitro and in vivo characterization of a novel serotonin-dopamine activity modulator”. J. Pharmacol. Exp. Ther. 350 (3): 589–604. doi:10.1124/jpet.114.213793.PMID 24947465.
  23. “Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder”. Retrieved 10 February 2012.
  24.  “Canadian Patents Database 2620688”. Retrieved 16 February 2012.
  25. http://zliming2004.blog.163.com/blog/static/6456372120149963644472/

 ………

JP2006316052A Title not available
US20110152286 * Dec 16, 2010 Jun 23, 2011 Otsuka Pharmaceutical Co., Ltd. Piperazine-substituted benzothiophenes for treatment of mental disorders
 UPDATED
SUZHOU VIGONVITA LIFE SCIENCES CO., LTD. [CN/CN]; 398, Ruoshui Road, Suzhou Industrial Park Suzhou, Jiangsu 215123 (CN).
TOPHARMAN SHANGHAI CO., LTD. [CN/CN]; 1088, Chuansha Road, Pudong Shanghai 201209 (CN).
SHANGHAI INSTITUTE OF MATERIA MEDICA, CHINESE ACADEMY OF SCIENCES [CN/CN]; 555, Zuchongzhi Road, Zhangjiang, Pudong Shanghai 201203 (CN)
(EN)The present invention relates to methods of preparing brexpiprazole, analogs thereof, key intermediates and salts thereof. Specifically, the present invention relates to new methods of preparing brexpiprazole, analogs thereof, key intermediates and salts thereof, and to the key intermediates and salts thereof used in the methods. The methods involve mild reaction conditions, stable intermediates, easy operations, and widely available reagents, thereby allowing for reduced synthesis costs, a shortened production cycle, high yield, and high product quality. The methods are suited for use in large-scale production.
Bray prazosin (Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates that act on more than one receptor, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (symptoms of depression); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as an adjunctive treatment of severe depression (MDD) Phase III clinical trials; III clinical trials the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States II Adult ADHD clinical trials.
Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2006112464A1 in the preparation route Bray prazosin, see Scheme 1, the difficulty of the route is the first step in the reaction by-products easily separated by column chromatography is not easy to obtain high-purity intermediates, thus affecting the final product Bray prazosin purity and yield.
Scheme 1:
Subsequently, Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2013015456A1 in the alternative method of preparing the reaction of this step, see Scheme 2, along the route of the reagents are more expensive, high-cost, environmentally unfriendly and not suitable for industrial production.
Reaction Scheme 2:
Due to the above production process there is a high cost, and difficult to separate impurities and other shortcomings, it is necessary to find an economical, practical, environmental protection, new routes to improve process stability, reduce costs, improve product quality.
DISCLOSURE
In response to these deficiencies, an object of the present invention is to provide a new, simple operation, high yield, low cost, environmentally friendly and suitable for industrial mass production Bray prazosin and the like, key intermediates and preparing a salt thereof.
Another object of the present invention is to provide novel compounds and salts thereof of the manufacturing process.
To achieve the above objects, the present invention provides compounds of formula I, the structure is as follows:
Wherein, R is C1 ~ C6 straight or branched chain alkyl, benzyl; preferably, R is C1 ~ C4 straight or branched chain alkyl group; most preferably, R is methyl, ethyl, t-butyl group;
R 1 is acyl amino-protecting groups (e.g. formyl ( ), an acetyl group, a propionyl group, a benzoyl group, haloacetyl group, phthaloyl) or class alkoxycarbonyl amino-protecting group (e.g. t-butoxycarbonyl , benzyloxycarbonyl, 9-fluorenyl methoxy carbonyl); said haloacetyl group is a fluorinated acetyl, bromoacetyl, chloroacetyl or iodoacetyl group; preferably, R 1 is formyl, acetyl and tert-butoxycarbonyl groups;
The present invention also provides a method for preparing a compound of formula I, the compound of formula II with a thioglycolate as shown in the reaction, the compound, the method shown in Formula I as shown in Scheme 3,
Reaction Scheme 3:
Wherein, X is halogen, such as fluorine, chlorine, bromine, iodine; R and R 1 are as defined above, the compound of formula I as defined the same;
The above reaction in the presence of a base, in particular, is an inorganic base (e.g. sodium hydroxide, potassium hydroxide, strontium hydroxide, lithium hydroxide, barium hydroxide, calcium hydroxide, cesium hydroxide, sodium hydrogen carbonate, potassium bicarbonate, potassium carbonate, sodium carbonate, strontium carbonate, cesium carbonate, sodium sulfide, sodium hydroxide, etc.) or an organic base (e.g. sodium alkoxide, potassium alkoxide, butyl lithium, 1,8-diazabicyclo [5,4 0] undecene -7 (DBU), pyridine, quinoline, 4-dimethylaminopyridine (DMAP) or an organic amine, etc.) performed in the presence of, wherein the sodium alkoxide may be sodium methoxide, sodium ethoxide, propoxy sodium alkoxide, sodium isopropoxide, n-butoxide, sodium tert-butoxide and the like; may be the potassium alkoxide, potassium methoxide, potassium ethoxide, potassium-propanol, potassium isopropoxide, n-butoxide, potassium tert-butoxide , the organic amine may be triethylamine, diethylamine, n-butylamine, tripropylamine, diisopropylamine, diisopropylethylamine, etc., preferably, the base may be an inorganic alkali sodium hydroxide, potassium hydroxide, lithium hydroxide, sodium hydrogencarbonate, potassium hydrogencarbonate, potassium carbonate, sodium carbonate, strontium carbonate, sodium sulfide, sodium hydroxide, or organic bases as sodium methoxide, sodium ethoxide, tert-butoxide potassium, triethylamine, diethylamine, diisopropylamine or diisopropylethylamine;
The above reaction is carried out in a suitable solvent, the solvent is water, C 1 ~ C 5 lower alcohol (such as methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol, tert-butanol, n-amyl alcohol, amyl alcohol, ethylene glycol, propylene glycol, glycerol), N, N- dimethylformamide (DMF), dimethyl sulfoxide (DMSO), tetrahydrofuran (THF), acetonitrile, dioxane, N- methylpyrrolidone, methylene chloride, chloroform, ethylene glycol dimethyl ether, diethylene glycol dimethyl ether or ethylene glycol monomethyl ether, and the like, one or more, preferably, the solvent is water , methanol, ethanol, N, N- dimethylformamide (DMF), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), acetonitrile, dioxane or ethylene glycol dimethyl ether or a species; the reaction time from 1 hour to 24 hours, preferably 2 hours to 12 hours. The reaction temperature is 0 ℃ ~ 150 ℃, preferably room temperature ~ 100 ℃.
To achieve the above object, the present invention also provides a compound of formula III, is structured as follows:
Wherein, R 1 is acyl amino protecting groups (e.g. formyl, acetyl, propionyl, benzoyl, halo acetyl, phthaloyl) or class alkoxycarbonyl amino-protecting group (e.g. t-butoxycarbonyl , benzyloxycarbonyl, 9-fluorenyl methoxycarbonyl), said haloacetyl group is a fluorinated acetyl, bromoacetyl, chloroacetyl or iodoacetyl;
Preferably, R 1 is formyl, acetyl or t-butoxycarbonyl;
The present invention also provides a method of preparing compounds of Formula III are shown, thioglycolic acid compound and reacting compound of formula III as shown in the method shown by the formula II as shown in Scheme 4,
Scheme 4:
Wherein, X is fluorine, chlorine, bromine or iodine; R 1 as defined above, with a compound of formula I as defined the same;
Bray prazosin (Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates that act on more than one receptor, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (symptoms of depression); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as an adjunctive treatment of severe depression (MDD) Phase III clinical trials; III clinical trials the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States II Adult ADHD clinical trials.
Scheme 5:

 

Wherein, X is fluorine, chlorine, bromine or iodine; R 1 are the same as defined in the compounds illustrated and R are as defined above for formula I. The present invention also provides processes for preparing key intermediates Bray prazosin or a salt thereof, the method as shown in Scheme 6:
Scheme 6:
14- (3-chloro-2-carboxaldehyde-phenyl-1 -) – Reference Example Synthesis of piperazine-1-carboxylic acid tert-butyl ester
A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), piperazine-1-carboxylate (646mg, 3.47mmol) was dissolved in N, N- dimethylformamide (5mL), and nitrogen at, at room temperature was added potassium carbonate (2.18g, 15.77mmol), the mixture was stirred for 4 hours at 80 ℃, cooled and filtered, water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, filtered The desiccant was concentrated to give a solid, after with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (750mg, 75% yield).
1 HNMR (400 MHz, CDCl 3 ): δ10.37 (s, 1H), 7.40 (t, 1H), 7.01 (d, 1H), 6.99 (d, 1H), 3.20 (m, 4H), 3.00 (s, 4H), 1.47 (s, 9H) ESI: [M + 1] + = 325.8.
Reference Example 21- carboxylic acid (3-chloro-2-carboxaldehyde-phenyl-1 -) – -4- piperazine

 

A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), 1- formyl piperazine (396mg, 3.47mmol) was dissolved in DMF (5mL), and under nitrogen at room temperature was added potassium carbonate (2.18g, 15.77mmol). The mixture was stirred for 4 hours at 80 ℃, cooled water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) After beating 1h, filtered to give a pale yellow solid (588mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 10.45 (s, 1H), 8.13 (s, 1H), 7.44 (t, 1H), 7.18 (d, 1H), 7.02 (d, 1H), 3.80 (s, 2H), 36.4 (s, 2H), 3.10 (m, 4H) ESI: [M + 1] + = 253.1.
Acetyl-31- (3-chloro-2-carboxaldehyde-phenyl-1 -) – 4- Reference piperazine
A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), 1- acetyl-piperazine (444mg, 3.47mmol) was dissolved in DMF (5mL), and under nitrogen at room temperature was added potassium carbonate (2.18g, 15.77 mmol), the mixture was stirred at 80 ℃ 4 hours, cooled and filtered, water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid, after with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (588mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 10.44 (s, 1H), 7.44 (t, 1H), 7.17 (d, 1H), 7.03 (d, 1H), 3.79 (bs, 4H), 3.10 (m, 4H), 2.18 (s, 3H) ESI: [M + 1] + = 267.1.
Piperazine-1-carboxylic acid tert-butyl ester – 14- (2-ethoxycarbonyl phenyl and thien-4-yl) Example
Under nitrogen, to N, was added the product (1.0g, 3.08mmol) of Reference Example 1 N- dimethylformamide (5mL) at room temperature within, ethyl thioglycolate (388mg, 3.20mmol), potassium carbonate (1.38 g, 10mmol), the mixture was stirred for 4 hours at 80 ℃, cooled and filtered, water (20mL), ethyl acetate (3x5mL) was extracted, dried over anhydrous sodium sulfate, the drying agent filtered, and concentrated to give a solid with petroleum ether (50mL ) after beating 1h, filtered to give a pale yellow solid (900mg, 75% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Example 24- (2-carboxy-benzothiophen-4-yl) – piperazine-1-carboxylate Synthesis of
Of the product (1.0g, 2.5mmol) in Example 1 was dissolved into 1,4-dioxane (5mL), was added 4N aqueous sodium hydroxide solution (1.8mL, 7.2mmol), the mixture was stirred for 3h at 80 ℃, cooled to room temperature, water (5mL) and ethyl acetate (10mL), separated and the aqueous phase with 1N HCl at 0 ℃ pH was adjusted to about 4.0, the resulting solid was filtered, dried to give a pale yellow solid.
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
34- (benzothiophen-4-yl) Example – Synthesis of piperazine-1-carboxylic acid tert-butyl ester
The product of Example 2 (20g, 54mmol) will be implemented, cuprous oxide (1g, 7mmol) was dissolved in quinoline (50mL) inside, heated to 140 ℃ overnight. After cooling and filtration, the filtrate was added water, extracted with ethyl acetate, the organic phase was washed with 1N HCl to slightly acidic, saturated aqueous sodium bicarbonate solution, purified by silica gel column chromatography, the concentrated solid slurried with petroleum ether to give an off-white solid (13g, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 7.57 (d, 1H), 7.41 (s, 2H), 7.27 (t, 1H), 6.88 (d, 1H), 3.66 (m, 4H), 3.01 (m, 4H), 1.50 (s, 9H) ESI: [M + 1] + = 319.1.
44- (benzothiophene-4-yl) Example – Synthesis of piperazine-1-carboxylic acid tert-butyl ester
The product of Example 2 (500mg, 1.35mmol) will be implemented, silver carbonate (40mg, 0.135mmol) and acetic acid (8mg) was dissolved in dimethylsulfoxide (5mL) inside, heated to 120 ℃, the reaction overnight, cooled and filtered, and the filtrate Water was added, extracted with ethyl acetate, and concentrated by column chromatography to obtain the target substance.
1 HNMR (400 MHz, CDCl 3 ): [delta] 7.57 (d, 1H), 7.41 (s, 2H), 7.27 (t, 1H), 6.88 (d, 1H), 3.66 (m, 4H), 3.01 (m, 4H), 1.50 (s, 9H) ESI: [M + 1] + = 319.1.
Piperazine hydrochloride – 51- (benzothiophen-4-yl) Example
At room temperature, the product of Example 3 will be implemented (2g, 6.2mmol) was dissolved in dioxane (6mL) was added 4N HCl / dioxane (6mL), stirred 3h, concentrated to dryness, the residue was beating ethyl acetate, filtered to obtain the target substance (1.3g, 95% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Example 61- formyl-4- (2-ethoxycarbonyl phenyl and thien-4-yl) – piperazine Synthesis
In N 2 protected, at room temperature was added the product of Reference Example 2 to DMF (5mL) inside (1.0g, 3.7mmol), ethyl thioglycolate (410mg, 3.80mmol), potassium carbonate (1.38g, 10mmol), the mixture 80 ℃ stirred for 4 hours. Cooling water was added (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) After beating 1h, filtered to give a pale yellow solid (1.0 g, yield 83%).
1 HNMR (400 MHz, CDCl3): [delta] 8.15 (d, 2H), 7.59 (d, 1H), 7.41 (t, 1H), 6.94 (d, 1H), 4.44 (q, 2H), 3.85 (t, 2H ), 3.68 (t, 2H), 3.21-3.15 (m, 4H), 1.44 (t, 3H) ESI: [M + 1] + = 319.1.

Example 71- formyl-4- (2-carboxy-benzothiophen-4-yl) – piperazine

The product (1.0g, 3.1mmol) of Example 6 was dissolved in methanol (5mL) and water (2mL) the addition of lithium hydroxide (420mg, 10mmol), the mixture was stirred at room temperature for 5h, was added water (5mL) and acetic acid ethyl ester (10mL), extracted, the aqueous phase was collected, the pH was adjusted to about 4.0 at 0 ℃ with 1N HCl solution, the precipitated solid was filtered and dried to give a pale yellow solid (510mg, 56% yield).
ESI: [M-1] = 289.1.
Example 81- formyl (benzothiophen-4-yl) -4 – piperazine
The product (1.0g, 3.4mmol) Example 7 will be implemented, cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, the organic phase was washed with aqueous 1N HCl to slightly acidic, then with saturated aqueous sodium bicarbonate solution, and concentrated by silica gel column chromatography, the resulting solid was slurried with petroleum ether to give white solid (520mg, 62% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.15 (s, 1H), 7.62 (d, 1H), 7.42 (m, 2H), 7.31 (t, 1H), 6.04 (d, 1H), 3.82 (t, 2H), 3.63 (t, 2H), 3.19-3.12 (m, 4H) ESI: [M + 1] + = 247.1.
Example 91- (benzothiophen-4-yl) – piperazine hydrochloride
A mixture of the product of Example 8 (500mg) was dissolved in dioxane (2mL) was added 4N HCl / dioxane (3mL), stirred 3h, concentrated to dryness, slurried with ethyl acetate, filtered to give the target (470mg, yield 90%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Example 10 1-Acetyl-4- (2-ethoxycarbonyl phenyl and thien-4-yl) – piperazine Synthesis
Under the protection of N2, at room temperature was added the product of Reference Example 3 (1.0g, 3.74mmol) to DMF (5mL) inside, ethyl thioglycolate (388mg, 3.20mmol), potassium carbonate (1.38g, 10mmol), the mixture was 80 ℃ stirred for 4 hours, cooled water was added (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (863mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.17 (s, 1H), 7.60 (d, 1H), 7.42 (t, 1H), 7.01 (d, 1H), 4.44 (q, 2H), 3.94 (br, 2H), 3.80 (br, 2H), 3.21 (br, 4H), 2.19 (s, 3H), 1.44 (t, 3H) ESI: [M + 1] + = 333.3.
Example 11 1-Acetyl-(2-carboxy-benzothiophen-4-yl) -4 – piperazine
The product (1.0g, 3.0mmol) from Example 10 was dissolved in methanol (5mL) and water (2mL) the addition of lithium hydroxide (300mg, 7.2mmol), the mixture was stirred at rt for 3h, water was added (5mL) and ethyl acetate ester (10mL), separated and the aqueous phase was collected, the pH adjusted with aqueous 1N HCl at 0 ℃ to about 4.0, and the precipitated solid was filtered, dried to give a pale yellow solid (820mg, yield 90%).
ESI: [M-1] = 303.1.
Example 12 1-Acetyl-4- (benzothiazol-4-yl) – piperazine
The product of Example 11 (1.0g, 3.2mmol), cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, washed with 1N HCl solution was added to a weak acid, a saturated aqueous solution of sodium bicarbonate, silica gel column chromatography, and concentrated to give a solid slurried with petroleum ether to give a white solid (600mg , yield 70%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.95 (s, 1H), 7.65 (d, 1H), 7.41 (t, 1H) 6.95 (d, 1H), 3.69 (q, 4H), 3.10 (t , 2H), 3.02 (t, 2H), 2.06 (s, 3H) ESI: [M + 1] + = 261.1.
EXAMPLE 131- (benzothiophen-4-yl) – piperazine hydrochloride
A mixture of the product of Example 12 (1g, 3.8mmol) was dissolved in dioxane (6mL) was added 4N HCl / dioxane (6mL), stirred 3h, concentrated dry, with ethyl beating, filtered to give the product (870mg, yield 90%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
141- (benzothiophen-4-yl) Example – piperazine

The product of (500mg, 1.38mmol) of Example 2 was dissolved in quinoline (3mL) the addition of cuprous oxide (20mg), the reaction temperature was raised to 140 ℃ After 2h, the reaction continues to heat up to 240 ℃ 3h, cooled to room temperature, filtered , water was added, extracted with ethyl acetate, washed with saturated aqueous sodium bicarbonate, silica gel column chromatography, and concentrated to give the desired product. 1 HNMR (300 MHz, DMSO-d 6 ): [delta] 8.74 (bs, 1H), 7.75 (d, 1H), 7.69 (d, 1H), 7.51 (d, 1H), 7.31 (t, 1H), 6.95 ( d, 1H), 3.24 (m, 8H) ESI: [M + 1] + = 219.2.

Example 15 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of

7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone (300mg, 0.59 mmol) was dissolved in methanol (3mL) and water (1mL) was added lithium hydroxide (76mg, 1.8mmol), stirred at rt for 3h, ethyl acetate was added, the aqueous phase was adjusted with 1N dilute hydrochloric acid to about pH 4.0, using bis dichloromethane: methanol (10: 1) extraction, concentration did a white solid (210mg, 46% yield).

1 HNMR (400 MHz, DMSO-d 6 ): [delta] 10.01 (s, 1H), 7.88 (s, 1H), 7.61 (d, 1H), 7.38 (t, 1H), 7.03 (q, 1H), 6.93 ( d, 1H), 6.48 (m, 2H), 3.92 (m, 4H), 3.35 (s, 4H), 2.84 (s, 4H), 2.77 (s, 2H), 2.62 (s, 2H), 1.72 (m , 4H), ESI: [M-1] = 478.3.

EXAMPLE 167- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of

The product (500mg, 1.04mmol) of Example 15 will be implemented, cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, washed with 1N HCl solution was added until pH = 4.0, dichloromethane: methanol (10: 1) extracted, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (320mg , yield 70%).

1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1]+ = 436.3.

Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 17 4- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added to ethanol (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), potassium carbonate (342mg, 2.48mmol), the mixture was 85 ℃ stirred for 18 hours, concentrated, and column chromatography to obtain the target substance (100mg, 42% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 18 4- (2-ethoxycarbonyl phenyl and thien-4-yl)
Under nitrogen, was added at room temperature to DMF (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), DIPEA (342mg, 2.48mmol), the mixture was at 105 ℃ stirred for 18 hours, 1N HCl solution was added adjust the pH = 7, and extracted with methyl tert-butyl ether, the ether layer was then washed three times with saturated brine, dried over anhydrous sodium sulfate, the drying agent filtered, and concentrated by column chromatography to obtain the target (170mg, yield 71%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 19 4- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added the product of Reference Example 1 to ethanol (5mL) inside (200mg, 0.62mmol), ethyl mercaptoacetate (0.081ml, 0.74mmol), sodium hydroxide (100mg, 2.48mmol), the mixture 85 ℃ stirred for 6 hours, concentrated to column chromatography to obtain the target substance (70mg, 30% yield).

1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 201-
The product of Example 2 (200mg, 0.55mmol), was dissolved in THF (5mL), concentrated hydrochloric acid (0.5mL), 50 ℃ heated 6h. Cooling, methyl tert-butyl ether (5mL), filtered to give the target (130mg, yield 79%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
Piperazine hydrochloride – (benzothiophene-4-yl) Example 211-
The product of Example 20 (130mg, 0.43mmol) was added to diphenyl ether (3mL) in, 260 ℃ heating 0.5h. Cooled and filtered to give the object (60mg, 55% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Preparation of tert-butyl piperazine-1 – Example 224- (2-carboxy-benzothiophen-4-yl)
Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium methoxide (133mg, 2.45mmol ), and the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (130mg, 58% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – (2-carboxy-benzothiophen-4-yl) Example 234-
Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium hydroxide (99mg, 2.45 mmol), the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (180mg, yield 81%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Example 24 7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of
2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added DIPEA (94mg, 0.73mmol) and ethyl mercaptoacetate (0.024mL, 0.22mmol), 110 ℃ stirred for 16 hours.Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (40mg, 46% yield).
1 HNMR (400 MHz, DMSO-d 6 ): δ11.69 (s, 1H), 11.24 (s, 1H), 8.09 (s, 1H), 7.81 (d, 1H), 7.74 (d, 1H), 7.57 ( d, 1H), 7.48 (t, 1H), 7.04 (d, 1H), 6.82 (m, 2H), 6.30 (d, 1H), 4.32 (m, 4H), 4.06 (t, 2H), 3.67-3.16 (m, 8H), 1.96 (m, 2H), 1.84 (m, 2H), 1.32 (t, 3H) ESI: [M + 1] + = 506.4.
Example 25 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of
7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone (100mg, 0.19 mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5mL), 100 ℃ stirred for 10 hours. The reaction mixture was poured into ice water, stirred for 10min after filtration, to obtain the target substance (40mg, 43% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 10.01 (s, 1H), 7.88 (s, 1H), 7.61 (d, 1H), 7.38 (t, 1H), 7.03 (q, 1H), 6.93 ( d, 1H), 6.48 (m, 2H), 3.92 (m, 4H), 3.35 (s, 4H), 2.84 (s, 4H), 2.77 (s, 2H), 2.62 (s, 2H), 1.72 (m , 4H), ESI: [M-1] = 478.3.
Example 26 7- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of
The product (400mg, 0.83mmol) of Example 25 will be implemented, silver carbonate (46mg, 0.16mmol) was dissolved in DMSO (5mL) and the acetic acid was heated to 120 ℃ overnight. Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated aqueous sodium bicarbonate and brine again each wash, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (80mg, yield 22%).
1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1]+ = 436.3.
Example 27 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of
2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added sodium hydroxide (29mg, 0.73mmol) and thioglycolic acid (0.025mL, 0.36mmol), 120 ℃ stirred for 16 hours. Cooling, water was added, adjusted with 1N HCl aqueous solution is about pH = 5, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (40mg, yield 46 %).

ESI: [M + 1] + = 478.0.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 28 1-
The product of Example 17 (100mg, 0.25mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5 mL) in, 100 ℃ stirred for 10 hours. The reaction mixture was poured into ice water, stirred for 10min after suction filtration to give the object (38mg, 50% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
uUpdate july 2015

On July 10, the U.S. Food and Drug Administration approved Rexulti (brexpiprazole) tablets to treat adults with schizophrenia and as an add-on treatment to an antidepressant medication to treat adults with major depressive disorder (MDD).

Schizophrenia is a chronic, severe, and disabling brain disorder affecting about one percent of Americans. Typically, symptoms are first seen in adults younger than 30 years of age and include hearing voices; believing other people are reading their minds or controlling their thoughts; and being suspicious or withdrawn.

MDD, commonly referred to as depression, is also a severe and disabling brain disorder characterized by mood changes and other symptoms that interfere with a person’s ability to work, sleep, study, eat, and enjoy once-pleasurable activities. Episodes of depression often recur throughout a person’s lifetime, although some may experience a single occurrence. Other signs and symptoms of MDD include loss of interest in usual activities; significant change in weight or appetite; insomnia or excessive sleeping (hypersomnia); restlessness/pacing (psychomotor agitation); increased fatigue; feelings of guilt or worthlessness; slowed thinking or impaired concentration; and suicide attempts or thoughts of suicide. Not all people with MDD experience the same symptoms.

“Schizophrenia and major depressive disorder can be disabling and can greatly disrupt day-to-day activities,” said Mitchell Mathis, M.D., director of the Division of Psychiatry Products in the FDA’s Center for Drug Evaluation and Research. “Medications affect everyone differently so it is important to have a variety of treatment options available for patients with mental illnesses.”

The effectiveness of Rexulti in treating schizophrenia was evaluated in 1,310 participants in two 6-week clinical trials. Rexulti was shown to reduce the occurrence of symptoms of schizophrenia compared to placebo (inactive tablet).

The effectiveness of Rexulti as an add-on treatment for MDD was evaluated in two 6-week trials that compared Rexulti plus an antidepressant to placebo plus an antidepressant in 1,046 participants for whom an antidepressant alone did not adequately treat their symptoms. The participants taking Rexulti reported fewer symptoms of depression than those taking the placebo.

Rexulti and other drugs used to treat schizophrenia have a Boxed Warning alerting health care professionals about an increased risk of death associated with the off-label use of these drugs to treat behavioral problems in older people with dementia-related psychosis. No drug in this class is approved to treat patients with dementia-related psychosis.

The Boxed Warning also alerts health care professionals and patients to an increased risk of suicidal thinking and behavior in children, adolescents, and young adults taking antidepressants. Patients should be monitored for worsening and emergence of suicidal thoughts and behaviors. Rexulti must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks.

The most common side effects reported by participants taking Rexulti in clinical trials included weight gain and an inner sense of restlessness, such as feeling the need to move.

Rexulti is manufactured by Tokyo-based Otsuka Pharmaceutical Company Ltd.

 

update………….

4-Chlorobenzo[b]thiophene a key intermediate in brexpiprazole synthesis

Abstract Image
We established an improved synthetic route to 4-chlorobenzo[b]thiophene, a key intermediate in brexpiprazole synthesis, via a practical decarboxylation process in three steps. Thermal analysis demonstrated that the coexistence of the decarboxylated product with DBU should be avoided and that removal of the product outside the reactor was vital. Our process yields the target compound by distillation under reduced pressure and is safe, highly batch efficient, cost-effective, and high yielding. Furthermore, manufacturing on a pilot scale was also accomplished through our approach.

Figure

4-Chlorobenzo[b]thiophene-2-carboxylic Acid (4)

 4 as a white solid
mp 260 °C.
1H NMR (300 MHz, DMSO-d6) δ 7.54 (d, 1H, J = 11.6, 7.7 Hz), 7.56 (dd, 1H, J = 17.8, 7.7 Hz), 8.03 (d, 1H, J = 0.7 Hz), 8.07 (td, 1H, J = 7.6, 0.9 Hz), 13.19 (brs, 1H).

13C NMR (75 MHz, DMSO-d6) δ 122.21, 125.01, 126.84, 127.99, 128.96, 136.50, 136.57, 142.55, 163.10.

Elemental analysis calcd for C: 50.83%, H: 2.37%, found C: 50.84%, H: 2.21%.

2,3,4,6,7,8,9,10-Octahydropyrimido[1,2-a]azepin-1-ium 4-Chlorobenzo[b]thiophene-2-carboxylate 5

5 as a white solid
Mp 182.5 °C.
1H NMR (300 MHz, CDCl3) δ 1.66 (m, 6H), 1.80–1.75 (m, 2H), 2.98–2.94 (m, 2H), 3.45–3.39 (m, 4H), 3.55–3.51 (m, 2H), 7.31–7.20 (m, 2H), 7.69 (dd, 1H, J = 3.9, 0.6 Hz), 7.97 (s, 1H), 13.19 (brs, 1H).

13C NMR (75 MHz, CDCl3) δ 19.51, 24.03, 26.70, 28.88, 31.99, 38.01, 48.36, 53.94, 121.04, 123.00, 125.17, 126.61, 128.98, 138.21, 142.43, 146.85, 165.91, 167.20.

Elemental analysis calcd for C: 59.25%, H: 5.80%, N: 7.68%, found C: 59.10%, H: 5.44%, N: 7.53%.

4-Chlorobenzo[b]thiophene (2)

1H NMR (300 MHz, CDCl3) δ 7.26 (t, 1H, J = 7.8 Hz), 7.36 (dd, 1H, J = 7.8, 0.9 Hz), 7.50 (d, 1H, J = 5.7 Hz), 7.52 (d, 1H, J = 5.7 Hz), 7.76 (d, 1H, J = 7.8 Hz).

13C NMR (75 MHz, CDCl3) δ 121.12, 122.40, 125.02, 127.43, 128.93, 138.06, 141.07.

Elemental analysis calcd for C: 56.98%, H: 2.99%, found C: 56.76%, H: 2.94%.

SEE

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00340

http://pubs.acs.org/doi/suppl/10.1021/acs.oprd.5b00340/suppl_file/op5b00340_si_001.pdf

Safe and Efficient Decarboxylation Process: A Practical Synthetic Route to 4-Chlorobenzo[b]thiophene

Bulk Pharmaceutical Chemicals Department, Second Tokushima Factory, Production Headquarters, Otsuka Pharmaceutical Co., Ltd., 224-18, Hiraishi Ebisuno, Kawauchi-cho, Tokushima 771-0182, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00340

 

Retosiban, GSK221149A


Retosiban structure.svg

Retosiban, GSK221149A

820957-38-8

MW 494.5827, MF C27 H34 N4 O5

Oxytocin antagonist

Threatened pre-term labour

PHASE 3 GSK

UNII-GIE06H28OX, GSK 221149A,  820957-38-8,

(3R,6R)-6-((S)-sec-butyl)-3-(2,3-dihydro-1H-inden-2-yl)-1-((R)-1-(2-methyloxazol-4-yl)-2-morpholino-2-oxoethyl)piperazine-2,5-dione

3(R)-(2,3-Dihydro-1H-inden-2-yl)-1-[1(R)-(2-methyloxazol-4-yl)-2-(4-morpholinyl)-2-oxoethyl]-6(R)-[1(S)-methylpropyl]piperazine-2,5-dione

(3R.6R)-3-(2,3-dihvdro-1 H-inden-2-v0-1 -\( R)-1 -(2-methyl-1 ,3-oxazol-4- yl)-2-(4-morpholinyl)-2-oxoethyll-6-r(1S -1-methylpropyn-2.5- piperazinedione

2,​5-​Piperazinedione, 3-​(2,​3-​dihydro-​1H-​inden-​2-​yl)​-​1-​[(1R)​-​1-​(2-​methyl-​4-​oxazolyl)​-​2-​(4-​morpholinyl)​-​2-​oxoethyl]​-​6-​[(1S)​-​1-​methylpropyl]​-​, (3R,​6R)​-

Morpholine, 4-[(2R)-[(3R,6R)-3-(2,3-dihydro-1H-inden-2-yl)-6-[(1S)-1-methylpropyl]-2,5-dioxo-1-piperazinyl](2-methyl-4-oxazolyl)acetyl]-

Retosiban (GSK-221,149-A)[1][2] is an oral drug which acts as a selective, sub-nanomolar (Ki = 0.65 nM) oxytocin receptor antagonist with >1400-fold selectivity[3] over the related vasopressin receptors and is being developed by GlaxoSmithKline for the treatment of preterm labour.[4][5]

Retosibanis an oxytocin (OT) antagonist in phase III clinical trials at GlaxoSmithKline for the prevention of preterm labor. OT antagonism is widely known to inhibit spontaneous uterine contractions.

Retosiban is a diketopiperazine nonpeptide compound with high potency and selectivity for the OT receptor over vasopressin receptors.

This  candidate has been shown to block oxytocin-induced uterine contractions when administered intravenously and to exhibit oral activity

Preterm labor is a major clinical problem leading to death and disability in newborns and accounts for 10% of all births and causes 70% of all infant mortality and morbidity.(Goldenberg, R. L.; Rouse, D.Prevention of premature birth N. Engl. J. Med. 1998, 339, 313)
Oxytocin (OT) is a potent stimulant of uterine contractions and is responsible for the initiation of labor via the interaction with the OT receptors in the mammalian uterus. OT antagonists have been shown to inhibit uterine contractions and delay preterm delivery. So there is increasing interest in OT antagonists because of their potential application in the prevention of preterm labor.
Although several tocolytics have already been approved in clinical practice, they have harmful maternal or fetal side effects.(Enkin, M.; Kierse, M.; Neilson, J.; Preterm Labour: A Guide to Effective Care in Pregnancy and Childbirth, 3rd ed.; Oxford University Press: Oxford, UK, 2000; pp 211225. )
The first clinically tested OT antagonist atosiban has a much more tolerable side effect profile and has recently been approved for use in Europe.
Atosiban SW.svgATOSIBAN

However, atosiban is a peptide and a mixed OT/vasopressin V1a receptor antagonist that has to be given by iv infusion and is not suitable for long-term maintenance treatment, as it is not orally bioavailable.((a) Bossmar, T.Treatment of preterm labor with the oxytocin and vasopressin antagonist atosiban J. Perinat. Med. 1998, 26, 458– 465

See also,(b) Coomarasamy, A.; Knox, E. M.; Gee, H.; Khan, K. S.Oxytocin antagonists for tocolysis in preterm labour—a systematic review Med. Sci. Monit. 2002, 8, RA268RA273)

Hence there has been considerable interest in overcoming the shortcomings of the peptide OT antagonists by identifying orally active nonpeptide OT antagonists with a higher degree of selectivity toward the vasopressin receptors (V1a, V1b, V2) with good oral bioavailability. Although several templates have been investigated as potential selective OT antagonists, few have achieved the required selectivity for the OT receptor vs the vasopressin receptors combined with the bioavailability and physical chemical properties required for an efficacious oral drug.(Borthwick, A. D.Oral Oxytocin Antagonists J. Med. Chem. 2010, 53, 65256538)
Therefore  the objective was to design a potent, orally active OT antagonist with high levels of selectivity over the vasopressin receptor with good oral bioavailability in humans that would delay labor safely by greater than seven days and with improved infant outcome, as shown by a reduced combined morbidity score.
The most potent of these was the 2,4-difluorophenyl dimethylamide 1, which has good in vitro (pKi = 9.2) and in vivo (IC50 = 227 nM) potency and is 20-fold more potent than atosiban in vitro. Compound 1 also has good pharmacokinetics with bioavailability >50% in both the rat and the dog.
Moreover, it is >500-fold selective over all three human vasopressin receptors (hV1aR, hV2R, and hV1bR) and has an acceptable P450 profile. In addition, it has a satisfactory safety profile in the genotoxicity screens and in the four day oral toxicity test in rats.

RETOSIBAN 106

However, 1 had poor aqueous solubility and high intrinsic clearance in human and cynomolgus monkey liver microsomes, so a compound was required that retained high antagonist potency and excellent pharmacokinetics in animal species seen with 1 but was more soluble and with improved human intrinsic clearance to decrease the risk of low bioavailability in humans.
first approach was to replace the 7-aryl ring with a five-membered heterocycle, which led to the oxazole Retosiban (106) a clinical candidate.(Borthwick, A. D.; Liddle, J.The design of orally bioavailable 2,5 diketopiperazine oxytocin antagonists: from concept to clinical candidate for premature labour Med. Res. Rev. 2011, 31, 576604)
As a backup to 106, an alternative replacement of the 7-aryl ring with a six-membered heterocycle was considered and in this report we describe how we investigated the modification of the 7-aryl ring to the 7(3′-pyridyl) ring and optimized substitution in this ring as well as modifying the isobutyl group to obtain good potency, lower intrinsic clearance in human microsomes, and good pharmacokinetics in animal species.
Barusiban.pngBARUSIBAN

 

L-368,899 structure.pngL-368899

L-371,257 structure.pngL-371257

PAPER

Pyridyl-2,5-diketopiperazines as potent, selective, and orally bioavailable oxytocin antagonists: Synthesis, pharmacokinetics, and in vivo potency
J Med Chem 2012, 55(2): 783

http://pubs.acs.org/doi/abs/10.1021/jm201287w

 PAPER

The discovery of GSK221149A: A potent and selective oxytocin antagonist
Bioorg Med Chem Lett 2008, 18(1): 90

http://www.sciencedirect.com/science/article/pii/S0960894X07013170

Full-size image (4 K)

Full-size image (30 K)

Scheme

Reagents and conditions: (a) triethylamine, MeOH; (b) H2, Pd/C, ethanol/acetic acid; (c) carbonyl diimidazole, CH2Cl2 3 h then acetone/2 N HCl; (d) benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate, dichloromethane 1 h then morpholine.

GSK221149A and other tertiary amides were prepared in four steps via the Ugi reaction as outlined in Scheme . A 2:1 mixture of diastereoisomers 24 was formed with the desirable (R)-diastereoisomer being the minor product. Hydrogenation of crude 24 furnished the cyclised phenol 25, again enriched with the undesirable (S)-diastereoisomer.

Activation of the mixture 25 with carbonyl diimidazole followed by the addition of 2 N HCl promoted epimerisation at the exocyclic position and yielded the acids 26 with the required (R)-diastereoisomer as the major product.

Acid activation with benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate followed by the addition of morpholine and subsequent column chromatography yielded homo-chiral GSK221149A.

 

PATENT

WO 2005000840

http://www.google.co.in/patents/EP1641787A1?cl=en

Example 3

(3R.6R)-3-(2,3-dihvdro-1 H-inden-2-v0-1 -\( R)-1 -(2-methyl-1 ,3-oxazol-4- yl)-2-(4-morpholinyl)-2-oxoethyll-6-r(1S -1-methylpropyn-2.5- piperazinedione ( 2R)-[(benzyloxycarbonyl)amino](2,3-dihydro-1 H-inden-2-yl)ethanoic acid (35.84g, 0.110mol) in a 500mL round bottomed flask was treated with 2,2,2-trifluoroethanol (165mL) followed by methanol (55ml) and triethylamine (11.13g, 15.33mL, 0.110mmol) the slurry was stirred for 3.5hrs until dissolution was observed. The solution was then added to (D)- allo Isoleucine methyl ester hydrochloride (20g, .110mol) in a separate flask. The slurry was stirred until dissolution was observed. 2-methyl-4- formyloxazole (12.24g, 0.110mmol) was then added followed by 2- benzyloxyphenylisocynanide (23.04g, 0.110mmol). The dark brown reaction mixture was then stirred at 20-25°C for 24hrs. The solution was then concentrated to a volume of ca. 130mL by distillation at reduced pressure.

The solution was the diluted with dichloromethane (200mL) and washed with water (2 x 200mL). The organic phase was then diluted with N-methyl pyrrolidinone (460mL) was and the dichloromethane removed by stirring at 40°C under vacuum for 2hrs. Acetic acid 46mL) was then added followed by palladium on carbon catalyst (69. Og of 10% Pd wt, 57% water, Johnson Matthey type 87L) and the mixture hydrogenated under balloon pressure of hydrogen with rapid stirring for 2hrs. The reaction mixture was then filtered, washed through with ethyl acetate (960mL) and washed with 3%w/v aq sodium chloride solution (960mL). The biphasic mixture was filtered and the organic phase separated and washed with 3%w/v aq sodium chloride solution (2 x 960mL). The organic solution was then diluted with ethyl acetate (200mL) and concentrated by distillation at atmospheric pressure by distilling out 385mL of solvent. The concentrated solution at 20-25°C was treated with 1 ,1′-carbonyldiimidazoIe (21.46g, 0.132mol) and stirred at 20-25°C for 1 hr then treated with water (290mL) and stirred rapidly at 20-25°C for 24hr. The mixture was allowed to settle and the ethyl acetate layer separated and discarded. The aqueous phase was washed with ethyl acetate (290mL) and the mixture allowed to settle and the aqueous phase was separated and acidified to pH 1-2 by the addition of concentrated hydrochloric acid (18mL).

The aqueous phase was then extracted into ethyl acetate (290mL and then 145mL). The combined ethyl acetate solution was then concentrated by distillation at atmospheric pressure to a volume of ca. 93mL. This solution was then diluted with tetrahydrofuran (62mL) and treated with triethylamine (11.02g, 15.20mL, 0.109mol) and cooled to -78°C. The solution was then treated with trimethylacetyl chloride (4.81 g, 4.92mL, 39.90mmol) and stirred at – 78°C for 7hr. The reaction mixture was then treated with a solution of morpholine (15.82g, 15.83mL, 0.181 mol) in tetrahydrofuran (23mL) and stirred at -78°C for 1hr 20mins before being allowed to warm to 20-25°C. The solution was then diluted with ethyl acetate (76mL) and washed with saturated aqueous sodium bicarbonate solution (2 x 153mL) followed by water (153mL). The organic solution was then diluted with ethyl acetate (54mL) and distilled down to a volume of 69mL at atmospheric pressure. The solution was then cooled to 20-25°C at which point crystallisation of the title compound occurred. The slurry of was then cooled further to 0°C before the title compound was isolated by filtration and sucked dry. Yield 8.92g.

 SYN WILL BE UPDATED.. ……………KEEP WATCHING

References

  • 1  Liddle J, Allen MJ, Borthwick AD, Brooks DP, Davies DE, Edwards RM, Exall AM, Hamlett C, Irving WR, Mason, AM, McCafferty GP, Nerozzi F, Peace S, Philp J, Pollard D, Pullen MA, Shabbir SS, Sollis SL, Westfall TD, Woollard PM, Wu C, Hickey DM (January 2008). “The discovery of GSK221149A: A potent and selective oxytocin antagonist”. Bioorganic & Medicinal Chemistry Letters 18 (1): 90–94. doi:10.1016/j.bmcl.2007.11.008. PMID 18032036.
  • 2
  • Borthwick, A. D.; Liddle, J. (January 2013). “Retosiban and Epelsiban: Potent and Selective Orally available Oxytocin Antagonists”. In Domling, A. Methods and Principles in Medicinal Chemistry: Protein-Protein Interactions in Drug Discovery. Weinheim: Wiley-VCH. pp. 225–256. ISBN 978-3-527-33107-9.
  • 3
  • McCafferty GP, Pullen MA, Wu C, Edwards RM, Allen M.J, Woollard PM, Borthwick AD, Liddle J, Hickey DM, Brooks DP, Westfall TD (March 2007). “Use of a novel and highly selective oxytocin receptor antagonist to characterize uterine contractions in the rat”. American Journal of Physiology – Regulatory, Integrative and Comparative Physiology 293: R299–R305. doi:10.1152/ajpregu.00057.2007. PMID 17395790.
  • 4
  • USAN Council (2007). “Statement on a Nonproprietary Name Adopted by the USAN Council” (PDF).
  • 5  Borthwick AD, Liddle J (July 2011). “The Design of Orally Bioavailable 2,5-Diketopiperazine Oxytocin Antagonists: From Concept to Clinical Candidate for Premature Labour”. Medicinal Research Reviews 31 (4): 576–604. doi:10.1002/med.20193. PMID 20027670.

…………..

OTHER INFO

http://pubs.acs.org/doi/abs/10.1021/jm201287w

Abstract Image

A six-stage stereoselective synthesis of indanyl-7-(3′-pyridyl)-(3R,6R,7R)-2,5-diketopiperazines oxytocin antagonists from indene is described. SAR studies involving mono- and disubstitution in the 3′-pyridyl ring and variation of the 3-isobutyl group gave potent compounds (pKi > 9.0) with good aqueous solubility. Evaluation of the pharmacokinetic profile in the rat, dog, and cynomolgus monkey of those derivatives with low cynomolgus monkey and human intrinsic clearance gave 2′,6′-dimethyl-3′-pyridyl Rsec-butyl morpholine amide Epelsiban (69), a highly potent oxytocin antagonist (pKi = 9.9) with >31000-fold selectivity over all three human vasopressin receptors hV1aR, hV2R, and hV1bR, with no significant P450 inhibition. Epelsiban has low levels of intrinsic clearance against the microsomes of four species, good bioavailability (55%) and comparable potency to atosiban in the rat, but is 100-fold more potent than the latter in vitro and was negative in the genotoxicity screens with a satisfactory oral safety profile in female rats.

EPELSIBAN

(3R,6R)-3-(2,3-Dihydro-1H-inden-2-yl)-1-[(1R)-1-(2,6-dimethyl-3-pyridinyl)-2-(4-morpholinyl)-2-oxoethyl]-6-[(1S)-1-methylpropyl]-2,5-piperazinedione (69)

69 as a white solid (2.4 g, 45%). Recystallisation from ethyl acetate/hexane (1:3) gave colorless needles (75%) mp 140 °C. 1H NMR (CDCl3) δ 7.49 (d, J = 7.8 Hz, 1H, pyridyl-4H), 7.26–7.15 (m, 4H, indanyl-arylH), 7.10 (d, J =8.1 Hz, 1H, pyridyl-5H), 6.68 (s, 1H, NCHpyridyl), 6.49 (d, J = 2.8 Hz, 1H, lactam-NH), 4.10 (dd, J = 10.1 Hz, 4.0 Hz, 1H, NCHindanyl), 4.01 (d, J = 4.5 Hz, NCHsec-butyl), 3.75–2.71 (m, 13H, 8× morpholinyl-H, indanyl-3H, –1H, –2H), 2.62 and 2.58 (2s, 6H, pyridyl-2Me,-6Me), 1.64–1.52 (m, 1H, CHHMe), 0.98–0.79 (m, 2H, CHHMe, CHMeCH2), 0.70 (t, J = 7.1 Hz, 3H, CH2Me), 0.45 (d, J = 6.8 Hz, 3H, CHMe). LCMS m/z 519 (MH+) single component, gradient 2 (tR 2.70 min). HRMS calcd for C30H38N4O4 (MH+) 519.29658, found 519.29667. HPLC: 100% (tR 10.388 min).
To a warm solution of 69 (2.66 g, 5.1 mmol) in acetone (40 mL) was added a solution of benzene sulfonic acid (0.81 g, 5.1 mmol) in acetone (40 mL), and the resulting solution was heated to boiling and allowed to cool to room temperature during 48 h. The resulting crystals were filtered off, air-dried on the filter pad to give the besylate (3.214 g, 92.6%) as white crystals of 69B mp 179–183 °C. 1H NMR (CD3OD) δ 8.30 (d, 1H, J = 8.1 Hz, pyridyl-4H), 7.84–7.80 (m, 2H, PhSO3ortho-H), 7.78 (d, J = 8.3 Hz, 1H, pyridyl-5H), 7.45–7.38 (m, 3H, PhSO3meta-H, para-H), 7.23–7.09 (m, 4H, indanyl-arylH), 6.08 (broad s, 1H, NCHpyridyl), 4.00 (d, J = 4.6 Hz, 1H, NCHsec-butyl), 3.92 (d, J = 9.9 Hz, 1H, NCHindanyl), 3.78–3.39 and 3.14–2.80 (m, 13H, 8× morpholinyl-H, indanyl-3H, –1H, –2H)), 2.79 and 2.78 (2s, 6H, pyridyl-2Me, -6Me), 1.85–1.74 (m, 1H, CHHMe), 1.59–1.48 (m, 1H, CHHMe), 1.15–1.01 (m, 1H, CHMeCH2), 0.92 (d, J = 6.3 Hz, 3H, CHMe), 0.85 (t, J = 7.3 Hz, 3H, CH2Me). LCMS m/z 519 MH+ single components, tR 2.72 min; circular dichroism (CH3CN) λmax 225.4 nm, dE −15.70, E15086; λmax 276 nm, dE 3.82, E5172. HRMS calcd for C30H38N4O4 (MH+) 519.2971, found 519.2972. Anal. (C30H38N4O4·C6H6O3S·3.0H2O) C, H, N, S.

…………..

Updates

Inline image 1

Inline image 2
Inline image 3

ANTHONY MELVIN CRASTO

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
Retosiban
Retosiban structure.svg
Systematic (IUPAC) name
(3R,6R)-6-[(2S)-butan-2-yl]-3-(2,3-dihydro-1H-inden-2-yl)-1-[(1R)-1-(2-methyl-1,3-oxazol-4-yl)-2-(morpholin-4-yl)-2-oxoethyl]piperazine-2,5-dione
Clinical data
Legal status
  • Non-regulated
Identifiers
CAS number 820957-38-8
ATC code None
PubChem CID 96025669
ChemSpider 23323798
UNII GIE06H28OX
KEGG D08986
Synonyms GSK-221,149-A
Chemical data
Formula C27H34N4O5 
Molecular mass 494.58 g/mol

Bremelanotide, Female Libido Enhancer


Bremelanotide chemical structure.png

Female Libido Enhancer  – Bremelanotide

Bremelanotide is a compound that is currently under investigation for its potential uses in managing reperfusion injury, female sexual dysfunction or hemorrhagic shock. The chemical may also see success in managing modulate inflammation or limiting the effects of ischemia.

N-Acetyl-L-norleucyl-L-alpha-aspartyl-L-histidyl-D-phenylalanyl-L-arginyl-L-tryptophyl-L-lysine (2-7)-lactam

Bremelanotide,  PT 141, CAS NO.: 189691-06-3

Synonym…..N-Acetyl-L-norleucyl-L-alpha-aspartyl-L-histidyl-D-phenylalanyl-L-arginyl-L-tryptophyl-L-lysine (2-7)-lactam, Bremelanotide PT 141
Molecular FormulaC50H68N14O10
Molecular Weight1025.16
Gedeon Richter….licensee
In May 2014, the company planned to file an NDA in the first half of 2016, and at that time, approval was expected in the first half of 2017

Bremelanotide Listeni/ˌbrɛmɨˈlænətd/ (formerly PT-141) is a compound under drug development by Palatin Technologies as a treatment for female sexual dysfunction, hemorrhagic shock and reperfusion injury. It functions by activating the melanocortin receptors MC1R and MC4R, to modulate inflammation and limiting ischemia.[2] It was originally tested for intranasal administration in treating female sexual dysfunction but this application was temporarily discontinued in 2008 after concerns were raised over adverse side effects of increased blood pressure. As of December 2014, Palatin is conducting a human Phase 3 study[3] using a subcutaneous drug delivery system that appears to have little effect on blood pressure.

Palatin, in collaboration with European licensee Gedeon Richter, is developing an sc formulation of the synthetic peptide bremelanotide (PT-141; BMT), a melanocortin MCR-4 agonist and a synthetically modified analog of PT-14, also analogous to alpha-melanocyte-stimulating hormone (alpha-MSH), for the potential treatment of female sexual dysfunction (FSD) including hypoactive sexual desire disorder (HSDD)

The Bremelanotide or PT-141 is a mean that explains the revolution caused by the medical world in a silent but attractive manner in the human health related study. Bremelanotide is the latest arrival from the company called Palatin Technologies which forms the basic treatment for the hemorrhagic shock and reperfusion injury.( In short about the company, the Palatin Technologies is the owner of this research and is located in New Jersey. Hence this medicine is a Jersey based Product. And regarding the product under research, is waiting for the approval from the Food and Drug Association. Once this is done, the company has targeted to reach those customers, whom the Viagra has approached. This has the effect of helping the male patients suffering with an erectile dysfunction syndrome. Also if it gets the approval as a treatment measure for the female sexual dysfunction, then this medicine is expected to bring a relief to the post-menopausal and also supports or provides their sexual happiness and also they are checking regarding thehyposexual desire disorder. This is expected to be a blockbuster, if released. So this medicine is waiting for a confirmation as well as an approval.

In February 2015, a randomized, double-blind, placebo-controlled, open-label extension, phase III trial (NCT02338960; BMT-302, Reconnect Study) was initiated in the US in premenopausal women (expected n = 550) with hypoactive sexual desire disorder to evaluate the efficacy and safety of bremelanotide. At that time, the trial was expected to complete in July 2017

Study – Potential Use Erectile Dysfunction

One study has explored the potential use of bremelanotide as a replacement for natural peptide melanocyte stimulating hormones for the sake of treating erectile dysfunction.

  • The goal of this study was to determine if the effects of bremelanotide stimulating sexual desire that was shown in male rats could be replicated in the brains of female rats. To do this, hormone primed female rats in a control group and a test group that were treated with bremelanotide and known to have consummatory sexual disorders was introduced to a group of male rats and the reactions were measured.
  • Heart racing, hops and darts, pacing and customary sexual behaviors were assessed while the brain was stimulated. The stimulation of specific molecular markers within the brain was examined to determine arousal in the female subjects.
  • Results indicated that the females saw an increase in sexual behavior when bremelanotide was applied to the limbic and hypothalamic regions of their brains. It is suggested that this was because the chemical that stimulated the mPOA terminals, leading to activated dopamine in the brain.

Additional study is necessary to determine the extent of the effects bremelanotide has on the brain and natural stimulating chemicals.

Bremelanotide and Ongoing Research

This is an advanced research involved even now. This functions by activating the Melanocortin, which is a group of peptide hormones which includes the adrenocorticotropic hormone and also the different forms of the melanocyte stimulating hormones. These melanocortins are produced or prepared from the proopiomelanocortin in the pituitary glands. The melanocortin releases or exert their effects by making a bind with the melanocortin and thereby activating it).The Bremelanotide functions by activating the melanocortin receptors and thereby makes a modulation in the inflammation. This is actually produced for making use in treating the sexual dysfunction. Due to certain reasons; the process of researching was kept under hold in recently, since it created some adverse side effects of increased blood pressure. In the chemistry of the preparation of the bremelanotide, the Peptide Melanaton II forms the basic compound. This compound is tested using a sunless tanning agent.

The actual information about the peptide melanaton has the effect of making sexual arousal and speed as well as sudden erections and some other side effects. However, there are several other measures taken to test the property of the same under several other health situations to make a detailed study about the chemical compound structure to make a change in the combination of the chemical structure. This medicine has made a revolution in the field of science of the human structure. When made a deep verification of the compound structure of the chemical study showed the following information. The structural design has an appearance of white colored powder like material, which has an accurate purity of nearly 98%. The actual molecular weight of the compound formed is around 1025.2. This compound has the collective share of Amino acids in the composition, peptide and acetate contents also.

The study of the compound structure PT-141 has an enhanced support of making a recombination that produces a different profile of the same medicine but in a different standard with different properties that may support the human requirement.

Bremelanotide PT-141 is known for its aphrodisiac properties

Development

Bremelanotide was developed from the peptide hormone Melanotan II which underwent testing as a sunless tanningagent. In initial testing, Melanotan II did induce tanning but additionally caused sexual arousal and spontaneous erections as unexpected side effects in nine out of the ten original male volunteer test subjects.[4]

In studies, bremelanotide was shown to induce lordosis in an animal model[5] and was also effective in treating sexual dysfunction in both men (erectile dysfunction or impotence) and women (sexual arousal disorder). Unlike Viagra and other related medications, it does not act upon the vascular system, but directly increases sexual desire via the nervous system.[6]

A Phase III clinical trial was scheduled to begin in the first half of 2007, but was delayed until August 2007. On August 30, Palatin announced that the U.S. Food and Drug Administration had expressed serious concerns regarding therisk/benefit ratio of bremelanotide with regards to the side effect of increased blood pressure. The FDA stated that it would consider alternate uses for bremelanotide, including as a treatment for individuals who do not respond to more established ED treatments. However, On May 13, 2008, Palatin Technologies announced it had “discontinued development of Bremelanotide for the treatment of male and female sexual dysfunction” while concurrently announcing plans to develop it as a treatment for hemorrhagic shock instead.[7] The company additionally announced intentions to focus its attention on another compound, PL-6983, that causes lower blood pressure in animal models.[8]Palatin has since re-initiated Bremelanotide studies for ED and FSD using a subcutaneous delivery method. On August 12, 2009, the company announced that in a double-blind study of 54 volunteers bremelanotide failed to evoke the hypertensive side effects seen with the nasal delivery system used in prior studies, concluding that “variability of uptake” inherent in intranasal administration of the drug resulted in “increases in blood pressure and gastrointestinal events…primarily related to high plasma levels in [only] a subset of patients” and that subcutaneous administration of the drug circumvented the potential for this side effect.[8] Palatin has completed a human Phase 2B study utilizing subcutaneous administration and reported positive results.[9]

Structure

Bremelanotide is a cyclic hepta-peptide lactam analog of alpha-melanocyte-stimulating hormone (alpha-MSH) that activates the melanocortin receptors MC3-R and MC4-R in thecentral nervous system. It has the amino acid sequence Ac-Nle-cyclo[Asp-His-D-Phe-Arg-Trp-Lys]-OH or cyclo-[Nle4, Asp5, D-Phe7, Lys10]alpha-MSH-(4-10). It is a metabolite of Melanotan II that lacks the C-terminal amide function.

Bremelanotide
Bremelanotide chemical structure.png
Systematic (IUPAC) name
(3S,6S,9R,12S,15S,23S)-15-[(N-acetyl-L-norleucyl)amino]-9-benzyl-6-{3-[(diaminomethylidene)amino]propyl}-12-(1H-imidazol-5-ylmethyl)-3-(1H-indol-3-ylmethyl)-2,5,8,11,14,17-hexaoxo-1,4,7,10,13,18-hexaa zacyclotricosane-23-carboxylic acid
Clinical data
Legal status
  • US: Unscheduled
Pharmacokinetic data
Half-life 120 minutes[1]
Identifiers
CAS number 189691-06-3 Yes
ATC code None
PubChem CID 9941379
ChemSpider 8116997 Yes
UNII 6Y24O4F92S Yes
KEGG D06569 
ChEMBL CHEMBL2070241 
Chemical data
Formula C50H68N14O10 
Molecular mass 1025.2 g/mol

Sexual dysfunction, including both penile erectile dysfunction or impotence and female sexual dysfunction, are common medical problems. Significant effort has been devoted over the last twenty or more years to develop methods, devices and compounds for treatment of sexual dysfunction. While more effort has been undertaken for treatment of penile erectile dysfunction, female sexual dysfunction is also an area to which significant research and effort has been devoted.

At present, one commonly used orally administered drug for treatment of sexual dysfunction in the male is Viagra®, a brand of sildenafil, which is a phosphodiesterase 5 inhibitor, increasing the persistence of cyclic guanosine monophosphate and thereby enhancing erectile response. There are several other medical treatment alternatives currently available depending on the nature and cause of the impotence problem. Some men have abnormally low levels of the male hormone testosterone, and treatment with testosterone injections or pills may be beneficial. However, comparatively few impotent men have low testosterone levels. For many forms of erectile dysfunction, treatment may be undertaken with drugs injected directly into the penis, including drugs such as papaverin, prostaglandin E1, phenoxybenzamine or phentolamine. These all work primarily by dilating the arterial blood vessels and decreasing the venous drainage. Urethral inserts, such as with suppositories containing prostaglandin, may also be employed. In addition, a variety of mechanical aids are employed, including constriction devices and penile implants.

A variety of treatments have also been explored for female sexual dysfunction, including use of sildenafil, although the Food and Drug Administration has not specifically approved such use. Testosterone propionate has also been employed to increase or augment female libido.

Melanocortin receptor-specific compounds have been explored for use of treatment of sexual dysfunction. In one report, a cyclic α-melanocyte-stimulating hormone (“α-MSH”) analog, called Melanotan-II, was evaluated for erectogenic properties for treatment of men with psychogenic erectile dysfunction. Wessells H. et al., J Urology 160:389-393 (1998); see also U.S. Pat. No. 5,576,290, issued Nov. 19, 1996 to M. E. Hadley, entitled Compositions and Methods for the Diagnosis and Treatment of Psychogenic Erectile Dysfunction and U.S. Pat. No. 6,051,555, issued Apr. 18, 2000, also to M. E. Hadley, entitled Stimulating Sexual Response in Females. The peptides used in U.S. Pat. Nos. 5,576,290 and 6,051,555 are also described in U.S. Pat. No. 5,674,839, issued Oct. 7, 1997, to V. J. Hruby, M. E. Hadley and F. Al-Obeidi, entitled Cyclic Analogs of AlphaMSH Fragments, and in U.S. Pat. No. 5,714,576, issued Feb. 3, 1998, to V. J. Hruby, M. E. Hadley and F. Al-Obeidi, entitled Linear Analogs of AlphaMSH Fragments. Melanotan-II is a peptide of the following formula:

Figure US06794489-20040921-C00001

Additional related peptides are disclosed in U.S. Pat. Nos. 5,576,290, 5,674,839, 5,714,576 and 6,051,555. These peptides are described as being useful for both the diagnosis and treatment of psychogenic sexual dysfunction in males and females. These peptides are related to the structure of melanocortins.

In use of Melanotan-II, significant erectile responses were observed, with 8 of 10 treated men developing clinically apparent erections, and with a mean duration of tip rigidity greater than 80% for 38 minutes with Melanotan-II compared to 3.0 minutes with a placebo (p=0.0045). The drug was administered by subcutaneous abdominal wall injection, at doses ranging from 0.025 to 0.157 mg/kg body weight. Transient side effects were observed, including nausea, stretching and yawning, and decreased appetite.

The minimum peptide fragment of native α-MSH needed for erectile response is the central tetrapeptide sequence, His6-Phe7-Arg8-Trp9 (SEQ ID NO:1). In general, all melanocortin peptides share the same active core sequence, His-Phe-Arg-Trp (SEQ ID NO:1), including melanotropin neuropeptides and adrenocorticotropin. Five distinct melanocortin receptor subtypes have been identified, called MC1-R through MC5-R, and of these MC3-R and MC4-R are believed to be expressed in the human brain. MC3-R has the highest expression in the arcuate nucleus of the hypothalamus, while MC4-R is more widely expressed in the thalamus, hypothalamus and hippocampus. A central nervous system mechanism for melanocortins in the induction of penile erection has been suggested by experiments demonstrating penile erection resulting from central intracerebroventricular administration of melanocortins in rats. While the mechanism of His-Phe-Arg-Trp (SEQ ID NO:1) induction of erectile response has not been fully elucidated, it has been hypothesized that it involves the central nervous system, and probably binding to MC3-R and/or MC4-R.

Other peptides and constructs have been proposed which are ligands that alter or regulate the activity of one or more melanocortin receptors. For example, International Patent Application No. PCT/US99/09216, entitled Isoquinoline Compound Melanocortin Receptor Ligands and Methods of Using Same, discloses two compounds that induce penile erections in rats. However, these compounds were administered by injection at doses of 1.8 mg/kg and 3.6 mg/kg, respectively, and at least one compound resulted in observable side effects, including yawning and stretching. Other melanocortin receptor-specific compounds with claimed application for treatment of sexual dysfunction are disclosed in International Patent Application No. PCT/US99/13252, entitled Spiropiperidine Derivatives as Melanocortin Receptor Agonists.

Both cyclic and linear α-MSH peptides have been studied; however, the peptides heretofore evaluated have had an amide or —NH2 group at the carboxyl terminus. See, for example, Wessells H. et al., J Urology, cited above; Haskell-Luevano C. et al., J Med Chem 40:2133-39 (1997); Schiöth H. B. et al., Brit J Pharmacol 124:75-82 (1998); Schiöth H. B. et al., Eur J Pharmacol 349:359-66 (1998); Hadley M. E. et al., Pigment Cell Res 9:213-34 (1996); Bednarek M. A. et al., Peptides20:401-09 (1999); U.S. Pat. Nos. 6,054,556, 6,051,555 and 5,576,290; and, International Patent Applications PCT/US99/04111 and PCT/US98/03298. While significant research has been conducted in an effort to determine the optimal structure of α-MSH peptides, including a variety of structure-function, agonist-antagonist, molecular modeling and pharmacophore studies, such studies have relied upon peptides with an art conventional —NH2 group at the carboxyl terminus. Further, it has long been believed that biologically active neuropeptides, including α-MSH peptides, are amidated, with an —NH2 group at the carboxyl terminus, and that such amidation is required both for biological activity and stability. See, for example, Metabolism of Brain Peptides, Ed. G. O’Cuinn, CRC Press, New York, 1995, pp. 1-9 and 99-101.

…………………………………………….

Bioorganic and Medicinal Chemistry Letters, 2005 ,  vol. 15,  4  pg. 1065 – 1068

http://www.sciencedirect.com/science/article/pii/S0960894X04014842

Full-size image (23 K)

Figure 2.

NMR structural analysis on compound 3.

Full-size image (24 K)

Figure 4.

NMR structural analysis of compound 1.

……………………………………………….

US6794489

In a preferred embodiment, the invention provides the peptide

Ac-Nle-cyclo(-Asp-His-D-Phe-Arg-Trp-Lys)-OH  Compound 1

The peptide of Compound 1 has a formula of C50H68N14O10, and a net molecular weight of 1025.18. This peptide may be synthesized by solid-phase means and purified to greater than 96% purity by HPLC, yielding a white powder that is a clear, colorless solution in water. The structure of Compound 1 is:

Figure US06794489-20040921-C00002

In general, the peptide compounds of this invention may be synthesized by solid-phase synthesis and purified according to methods known in the art. Any of a number of well-known procedures utilizing a variety of resins and reagents may be used to prepare the compounds of this invention.

The peptides of this invention may be in the form of any pharmaceutically acceptable salt. Acid addition salts of the compounds of this invention are prepared in a suitable solvent from the peptide and an excess of an acid, such as hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, trifluoroacetic, maleic, succinic or methanesulfonic. The acetate salt form is especially useful. Where the compounds of this invention include an acidic moiety, suitable pharmaceutically acceptable salts may include alkali metal salts, such as sodium or potassium salts, or alkaline earth metal salts, such as calcium or magnesium salts.

The invention provides a pharmaceutical composition that includes a peptide of this invention and a pharmaceutically acceptable carrier. The carrier may be a liquid formulation, and is preferably a buffered, isotonic, aqueous solution. Pharmaceutically acceptable carriers also include excipients, such as diluents, carriers and the like, and additives, such as stabilizing agents, preservatives, solubilizing agents, buffers and the like, as hereafter described.

EXAMPLE 1

Peptide Synthesis

The peptide Ac-Nle-cyclo(-Asp-His-D-Phe-Arg-Trp-Lys)-OH was synthesized by standard solid phase peptide synthesis methods, and is a cyclic heptapeptide melanocortin peptide analog with a free acid at the carboxyl terminus and an acetylated amino group at the amino terminus, with the structure:

Figure US06794489-20040921-C00003

The peptide has a net molecular weight of 1025.18, and is supplied in an acetate salt form. The peptide is a white, odorless amorphous hygroscopic powder, soluble in 0.9% saline, composed of C50H68N14O10. For synthesis, an Fmoc-Lys(R3)-p-alkoxybenzyl alcohol resin was transferred to a solid phase peptide synthesizer reactor with a mechanical stirrer. The R3group, such as 1-(1′-adamantyl)-1-methyl-ethoxycarbonyl (Adpoc), allyloxycarbonyl (Aloc) or 4-methyltrityl (Mtt), was removed and the next Fmoc-protected amino acid (Fmoc-Trp(Boc)-OH) was added to the resin through standard coupling procedures. The Fmoc protective group was removed and the remaining amino acids added individually in the correct sequence, by repeating coupling and deprotection procedures until the amino acid sequence was completed. After completion of coupling with the last Fmoc-amino acid derivative, Fmoc-Nle-OH, and cleavage of the Fmoc protective group, the exposed terminal amino group was acetylated with acetic anhydride and pyridine in N,N-dimethylformamide (DMF). The peptide-resin was dried and the Lys and Asp protective groups cleaved. The Lys and Asp deprotected peptide resin was suspended in a suitable solvent, such as DMF, dichloromethane (DCM) or 1-methyl-2-pyrrolidone (NMP), a suitable cyclic coupling reagent, such as 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TBTU), 2-(7-aza-1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TATU), 2-(2-oxo-1(2H)-pyridyl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TBTU) or N,N′-dicyclohexylcarbodiimide/1-hydroxybenzotriazole (DCCl/HOBt) was added, and coupling initiated by use of a suitable base, such as N,N-diispropylethylamine (DIPEA), sym-collidine or N-methylmorpholine (NMM). After cyclization, the peptide-resin was washed and the peptide cleaved from the resin and any remaining protective groups using trifluoroacetic acid (TFA) in the presence of water and 1,2-ethanedithiol (EDT). The final product was precipitated by adding cold ether and collected by filtration. Final purification was by reversed phase HPLC using a C18 column. The purified peptide was converted to acetate salt by passage through an ion-exchange column.

…………………………………………..

WO2014071339

Compounds of the Invention.

in a preferred embodiment of the present invention, fie rneianocortin receptor agonist is;

Ac-Nie”Cyc/o{-Asp-His–D–Phe-Arg–Trp»Lys)–OH (bremeianotide)

The peptide of bremeianotide has a formula of CsaHesN< C½, and a net mofecufar weight of 1025.18, This peptide may be synthesized by conventional means, including either solid-phase or Squid-phase techniques, and purified to greater than 99% purity by HPLC, yielding a white powder that is a clear, colorless solution in water. The structure of bremeianotide is:

Figure imgf000017_0001

in one embodiment of the invention, bremeianotide is synthesized by solid-phase synthesis and purified according to methods known in the art. Any of a number of well-known procedures utilizing a variety of resins and reagents may be used to prepare bremeianotide.

Bremeianotide may be in the form of any pharmaceutically acceptable salt. Acid addition salts of the compounds of this invention are prepared in a suitable solvent from the peptide and an excess of an acid, such as hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, trifluoroacefie, maieic, citric, tartaric, oxalic, succinic or methanesu!fonic acid. The acetate salt form is especially useful.

in a preferred embodiment, bremelanotide is an acetate salt form, and is formulated in a buffered aqueous solution including giycerin, and prepackaged in a syringe and auto-injector device. In alternative embodiments, bremelanotide is any pharmaceutically acceptable salt form, and is formulated in any pharmaceutically acceptable aqueous solution, the aqueous solution optionally including one or more salts, such as sodium chloride, one or more acids, such as citric acid, and one or more additional ingredients, including cellulose or derivatives thereof, saccharides o

polysaccharides such as dextrose, and any of a wide variety of surfactants, chelating agents and preservatives.

………………………………………….

US20050222014

In yet another embodiment of the present invention, the melanocortin receptor agonist is:
AcNle-cyclo(-AspHisDPheArgTrpLys)-OH PT-141

The peptide of PT-141 has a formula of C50H68N14O10, and a net molecular weight of 1025.18. This peptide may be synthesized by conventional means, including either solid-phase or liquid-phase techniques, and purified to greater than 99% purity by HPLC, yielding a white powder that is a clear, colorless solution in water. The structure of PT-141 is:

Figure US20050222014A1-20051006-C00001

In one embodiment of the invention, PT-141 is synthesized by solid-phase synthesis and purified according to methods known in the art. Any of a number of well-known procedures utilizing a variety of resins and reagents may be used to prepare PT-141.

PT-141 may be in the form of any pharmaceutically acceptable salt. Acid addition salts of the compounds of this invention are prepared in a suitable solvent from the peptide and an excess of an acid, such as hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, trifluoroacetic, maleic, citric, tartaric, oxalic, succinic or methanesulfonic acid. The acetate salt form is especially useful. Where the compounds of this invention include an acidic moiety, suitable pharmaceutically acceptable salts may include alkali metal salts, such as sodium or potassium salts, or alkaline earth metal salts, such as calcium or magnesium salts.

In a preferred embodiment, PT-141 is an acetate salt form, and is formulated in a buffered aqueous solution including glycerin, prepackaged in a metered unit dose intranasal delivery device. In alternative embodiments, PT-141 is any pharmaceutically acceptable salt form, and is formulated in any pharmaceutically acceptable aqueous solution, the aqueous solution optionally including one or more salts, such as sodium chloride, one or more acids, such as citric acid, and one or more additional ingredients, including cellulose or derivatives thereof, saccharides or polysaccharides such as dextrose, and any of a wide variety of surfactants, chelating agents and preservatives. In one preferred embodiment, PT-141 is administered to patients in volumes of 100 μL, with the quantity of PT-141 delivered determined by the concentration thereof. As described hereafter, in one preferred embodiment a metered unit dose contains 7.5 mg of PT-141.

While certain embodiments of the present invention are described primarily in the context of PT-141, it is to be understood that other melanocortin receptor agonists may be employed. For example, the metallopeptide melanocortin receptor agonists disclosed in WO 02/064091, filed on Feb. 13, 2001, and U.S. Ser. No. 10/640,755, filed on Aug. 13, 2003, both entitled Melanocortin Metallopeptides for Treatment of Sexual Dysfunction; and WO 01/13112, filed on Jun. 14, 2000, entitled Melanocortin Metallopeptide Constructs, Combinatorial Libraries and Applications, may be employed. In addition, the peptidomimetic melanocortin receptor agonists disclosed in U.S. Ser. No. 10/776,419, filed on Feb. 10, 2004, entitled Peptidomimetics of Biologically Active Metallopeptides; the pyrrolidine melanocortin receptor agonists disclosed in U.S. Ser. No. 10/766,657, filed on Feb. 10, 2004, entitled Pyrrolidine Melanocortin-Specific Compounds; and the bicyclic melanocortin receptor agonists disclosed in PCT/US04/01505, filed on Jan. 20, 2004, entitled Bicyclic Melanocortin-Specific Compounds, may also be employed. Also particular preferred are the piperazine melanocortin agonists disclosed in PCT/US04/01462, filed on Jan. 20, 2004 and U.S. Ser. No. 10/762,079, filed on Jan. 20, 2004, both entitled piperazine Melanocortin-Specific Compounds; the melanocortin agonists disclosed in WO 03/006620, filed on Jul. 11, 2002, entitled Linear and Cyclic Melanocortin Receptor-Specific Peptides; WO 04/005324, filed on Jul. 9, 2003, entitled Peptide Compositions for Treatment of Sexual Dysfunction; PCT/US00/18217, filed on Jun. 29, 2000 and U.S. Ser. No. 10/040,547, filed on Jan. 4, 2002, entitled Compositions and Methods for Treatment of Sexual Dysfunction; and U.S. Ser. No. 10/638,071, filed on Aug. 8, 2003, entitled Cyclic Peptide Compositions and Methods for Treatment of Sexual Dysfunction. The entire disclosure of each of the foregoing are incorporated here by reference. It is to be understood that the foregoing listing of patent applications disclosing melanocortin receptor agonists is intended to only be exemplary, and that other melanocortin receptor agonists, whether heretofore known or hereafter developed, may similarly be used in the practice of this invention.

…………………….

NMR prediction

H-NMR spectral analysis
bremelanotide NMR spectra analysis, Chemical CAS NO. 189691-06-3 NMR spectral analysis, bremelanotide H-NMR spectrum
13 C NMR PREDICTION
bremelanotide NMR spectra analysis, Chemical CAS NO. 189691-06-3 NMR spectral analysis, bremelanotide C-NMR spectrum
References
  1.  King SH, Mayorov AV, Balse-Srinivasan P, Hruby VJ, Vanderah TW, Wessells H (2007).“Melanocortin receptors, melanotropic peptides and penile erection”. Current Topics in Medicinal Chemistry 7 (11): 1098–1106. doi:10.2174/1568026610707011111.PMC 2694735. PMID 17584130.
  2.  Bremelanotide for Organ Protection and Related Indications, Palatin Technologies fact sheet. Retrieved on 2009-01-18.
  3.  “Palatin Announces Start of Bremelanotide Phase 3 Program For Female Sexual Dysfunction”. PR Newswire. Retrieved 2015-02-17.
  4.  “Tanning drug may find new life as Viagra alternative”. CNN. 1999. Retrieved2007-09-16.
  5. Pfaus JG, Shadiack A, Van Soest T, Tse M, Molinoff P (July 2004). “Selective facilitation of sexual solicitation in the female rat by a melanocortin receptor agonist”. Proc. Natl. Acad. Sci. U.S.A. 101 (27): 10201–4. doi:10.1073/pnas.0400491101. PMC 454387.PMID 15226502.
  6. Vicki Mabrey (2006). “ABC News “The Business of Desire – Love Potion””. ABC News. Retrieved 2009-01-24.
  7.  “Palatin Technologies announces new strategic objectives and reports third quarter 2008 financial results”. Palatin Technologies press release. 2008. Retrieved 2008-08-21.
  8.  “Palatin Technologies Announces New Strategic Objectives”. Retrieved 2008-05-13.
  9.  http://www.palatin.com/news/news.asp?ud=306

External links

PALATIN TECHNOLOGIES, INC.: ‘Bremelanotide in Premenopausal Women With Female Sexual Arousal Disorder and/or Hypoactive Sexual Desire Disorder‘ CLINICALTRIALS.GOV (NCT01382719, [Online] 20 March 2012, page 1 Retrieved from the Internet: <URL:http://clinicaltrials.gov/archive/NCT0 1382719/ 2012-03 20> [retrieved on 2014-02-10]
2 * PALATIN TECHNOLOGIES, INC.: ‘Reports Positive Bremelanotide Study; Improved Safety Profile with Subcutaneous Administration‘ PR NEWSWIRE., [Online] 12 August 2009, Retrieved from the Internet: <URL:http://www.thefreelibrary.com/Palatin +Technolo9ies,+Inc.+Reports+Positive+Bremel anotide+Study%38…-a020561 3302> [retrieved on 2014-02-10]
3 * SAFARINEJAD, MR.: ‘Evaluation of the Safety and Efficacy of Bremelanotide, a Melanocortin Receptor Agonist, in Female Subjects with Arousal Disorder: A Double-Blind Placebo-Controlled, Fixed Dose, Randomized Study”.‘ INTERNATIONAL SOCIETY FOR SEXUAL MEDICINE. vol. 5, 2008, pages 887 – 897
US8455617 Jun 7, 2010 Jun 4, 2013 Astrazeneca Ab Melanocortin receptor-specific peptides
US8455618 Oct 26, 2011 Jun 4, 2013 Astrazeneca Ab Melanocortin receptor-specific peptides
US8487073 Nov 23, 2010 Jul 16, 2013 Palatin Technologies, Inc. Melanocortin receptor-specific peptides for treatment of sexual dysfunction
US8729224 Jun 5, 2013 May 20, 2014 Palatin Technologies, Inc. Melanocortin receptor-specific peptides for treatment of female sexual dysfunction
EP2266567A1 May 26, 2009 Dec 29, 2010 Æterna Zentaris GmbH Use of cetrorelix in combination with PDE V inhibitors for the treatment of sex hormone dependent disorders
EP2266568A1 May 26, 2009 Dec 29, 2010 Æterna Zentaris GmbH Use of LHRH antagonists in combination with PDE V inhibitors for the treatment of sex hormone dependent disorders
WO2013067309A1 Nov 2, 2012 May 10, 2013 Xion Pharmaceutical Corporation Methods and compositions for oral administration of melanocortin receptor agonist compounds
WO2014071339A2 * Nov 5, 2013 May 8, 2014 Palatin Technologies, Inc. Uses of bremelanotide in therapy for female sexual dysfunction
WO2009151714A2 * Mar 24, 2009 Dec 17, 2009 Palatin Technologies, Inc. Therapeutic for treatment of circulatory shock, ischemia, inflammatory disease and related conditions
US6794489 * Jan 4, 2002 Sep 21, 2004 Palatin Technologies, Inc. Peptide sequence ac-nle-cyclo(-asp-his-d-phe-arg-trp-lys)-oh derived from a melanocyte-stimulating hormone (? alpha -msh?) analog, called melanotan-ii
US20050222014 * May 26, 2005 Oct 6, 2005 Palatin Technologies, Inc. Administering phosphodiestarase inhibitors and melanocortin receptor antagonist: synergistic mixture
US20110065652 * Nov 23, 2010 Mar 17, 2011 Palatin Technologies, Inc. Melanocortin Receptor-Specific Peptides for Treatment of Sexual Dysfunction

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

Join me on Linkedin

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

Join me on Facebook FACEBOOK

Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus

 amcrasto@gmail.com

KHAJURAHO INDIA

Khajuraho Group of Monuments is located in India

Khajuraho Group of Monuments
Location of Khajuraho Group of Monuments in India.

Location in Madhya PradeshLocation in Madhya Pradesh

  1. Khajuraho Group of Monuments – Wikipedia, the free …

    en.wikipedia.org/wiki/Khajuraho_Group_of_Monuments

    The Khajuraho Group of Monuments are a group of Hindu and Jain temples in Madhya Pradesh, India. About 620 kilometres (385 mi) southeast of New Delhi, …

Hotel Chandela – A Taj Leisure Hotel

LATUR, MAHARASHTRA, INDIA

http://en.wikipedia.org/wiki/Latur

Latur
लातूर
Lattalur, Ratnapur
City

Latur is located in Maharashtra

Latur
Latur

Location in Maharashtra, India

Coordinates: 18.40°N 76.56°ECoordinates18.40°N 76.56°E
Country  India
State Maharashtra
Region Aurangabad Division
District Latur
Settled Possibly 7th century AD
Government
 • Body Latur Municipal Corporation
 • Mayor Akhtar Shaikh
Area[1]
 • Total 117.78 km2(45.48 sq mi)
Area rank 89
Elevation 515 m (1,690 ft)
Population (2011)
 • Total 382,754
 • Rank 89th
 • Density 3,200/km2(8,400/sq mi)
Demonym Laturkar
Languages
 • Official Marathi
Time zone IST (UTC+5:30)
PIN
  • 413 512
  • 413 531
Telephone code 91-2382
Vehicle registration MH-24
Sex ratio 923.54 /1000 
Literacy 89.67
Distance from Mumbai 497 kilometres (309 mi) E (land)
Distance fromHyderabad 337 kilometres (209 mi) NW (land)
Distance fromAurangabad, Maharashtra 294 kilometres (183 mi) SE (land)
Climate BSh (Köppen)
Precipitation 666 millimetres (26.2 in)
Avg. summer temperature 41 °C (106 °F)
Avg. winter temperature 13 °C (55 °F)
http://www.citypopulation.de/world/Agglomerations.html

Map of latur city

his Is The Famous ‘Ganj-Golai’ As The Central Place Of The Latur City. There Are 16 Roads Connecting To This Place And Seperate Markets i.e. Jewellers …

लातूर जिल्हयातील चित्र संग्रह


LATUR AIRPORT

LATUR AIRPORT

2012 Navratri Mahotsav in Latur

SOS Children’s Village Latur

Latur, India: Carnival Resort

Ausa Near Latur

Chakur near Latur


Vilasrao Deshmukh’s ancestral home at Babhalgaon village in Latur. Machindra Amle

UDGIR: Udgir is one of the most important towns of Latur district. Udgir has a great historical significance. It has witnessed the war between the Marathas …

The city of Latur is located in India’s welathiest state, Maharashtra. Together with many of the surrounding villages, Latur was all but destroyed in the

CDK Inhibitor, MK 7965, DINACICLIB, SCH 727965


Cancers 06 02224 g003 1024
CDK Inhibitor, MK 7965, DINACICLIB, SCH 727965

SCH727965, SCH-727965, CAS 779353-01-4, Tube003, Dinaciclib (USAN/INN), UNII-4V8ECV0NBQ
Molecular Formula:C21H28N6O2
Molecular Weight:396.48602 g/mol

REVIEW…….http://www.mdpi.com/2072-6694/6/4/2224/htm

One of the most popular CDK inhibitor in clinical trials in the recent years was dinaciclib (MK-7965, SCH 727965) (Figure 3), the inhibitor of CDK1, CDK2, CDK5, and CDK9. A Phase I trial on the effect of dinaciclib in combination with aprepitant was performed in patients with advanced malignancies [44]. Aprepitant is used for the prevention of chemotherapy-induced nausea and vomiting, is known as an inhibitor and inducer of CYP3A4, which metabolizes dinaciclib.

Coadministration of dinaciclib with aprepitant resulted in no clinically significant effect on the pharmacokinetics and did not alter the safety profile of dinaciclib. The first Phase I clinical trial on dinaciclib as a single agent was performed on patients with advanced malignancies [68]. Forty-eight patients with various solid tumors were treated and 10 of them achieved prolonged stable disease for at least four treatment cycles. Adverse effects were mild, the most common being nausea, anemia, decreased appetite and fatigue.

A phase II multi-center study of dinaciclib for relapsed and/or refractory AML was performed on 20 patients [69]. Temporary decrease in peripheral blood and/or bone marrow blasts was observed in 60% of patients. Four of 13 (31%) patients with circulating blasts had >50% decrease and 6 (46%) >80% decrease in the absolute blast count within 1–8 days of the first dinaciclib dose. Toxicities included diarrhea, fatigue, transaminitis, and manifestations of tumor lysis syndrome, with one patient who deceased of acute renal failure. Another Phase II study was performed of dinaciclib versus erlotinib in patients with non-small cell lung cancer [70].

Unfortunately, it was found that dinaciclib was not successful as monotherapy in non-small cell lung cancer. Most common toxicities included neutropenia, leukopenia, vomiting, and diarrhea. Yet another Phase II study was performed on dinaciclib versus capecitabine in patients with advanced breast cancer [71]. Dinaciclib treatment demonstrated antitumor activity in two of seven patients with ER-positive and ERBB 2-negative metastatic breast cancer, however efficacy was not superior to capecitabine (p = 0.991).

Toxicities included neutropenia, leukopenia, increase in aspartate aminotransferase, and febrile neutropenia. Phase I nonrandomized dose-escalation trial was performed, where patients with relapsed or refractory chronic lymphocytic leukemia were treated with dinaciclib and rituximab [72]. Four out of six patients achieved stable disease, and one patient achieved complete response. Drug-related adverse events were mostly hematological, digestive and metabolic and no dose-limiting toxicities were observed. Dinaciclib was also moved into Phase III development for refractory chronic lymphocytic leukemia [73]. Phase I/II clinical trial Dinaciclib in patients with relapsed multiple myeloma showed promise as single agent [74]. The overall confirmed response rate was 3 of 27 (11%). Adverse effects included leukopenia, thrombocytopenia, gastrointestinal symptoms, alopecia, and fatigue. –

FOR REF See more at: http://www.mdpi.com/2072-6694/6/4/2224/htm#sthash.amBuLwq1.dpuf

Dinaciclib (SCH-727965) is an experimental drug that inhibits cyclin-dependent kinases (CDKs.[1] It is being evaluated in clinical trials for various cancer indications.[2]

Mechanisms of action

Anti-tumoral action

  • In melanoma
    • The anti-melanoma activity of dinaciclib is dependent on p53 signaling.[5]
  • In osteosarcoma
    • Dinacliclib induces the apoptosis of osteosarcoma cells.[8]
    • Apoptosis of osteosarcoma cultures can be induced by the combination of the cyclin-dependent kinase inhibitor SCH727965 and a heat shock protein 90 inhibitor.[9]
Dinaciclib.svg
Systematic (IUPAC) name
(S)-3-(((3-Ethyl-5-(2-(2-hydroxyethyl)piperidin-1-yl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)pyridine 1-oxide
Clinical data
Legal status
  • Investigational
Identifiers
CAS number 779353-01-4
ATC code ?
PubChem CID 46926350
ChemSpider 25027387
ChEMBL CHEMBL2103840
Synonyms SCH-727965
Chemical data
Formula C21H28N6O2 

Clinical trials

  • Phase 1[10]
  • Phase 2
    • Advanced breast cancer[11]
    • Non-small cell lung cancer (NSCLC)[12]

http://www.google.com.tr/patents/US8076479

One example of these inhibitors is the compound of Formula II.

Figure US08076479-20111213-C00006

The synthesis of the compound of Formula II is described in the ‘878 publication according to Scheme II:
Scheme II:

Step 1—Amidization to Form Substituted Pyrazole

Figure US08076479-20111213-C00007

http://www.google.com.tr/patents/US8076479

Step 2—Formation and Dehalogenation of pyrazolo[1,5a]pyrimidine

Figure US08076479-20111213-C00008

Step 3—Amination (Two Separate, Sequential Reactions)

Figure US08076479-20111213-C00009

As described in the ‘878 publication, Synthetic Scheme II leading to the compound of Formula II has several disadvantages from the standpoint of commercial scale synthesis. In step 1, the starting material (compound “C”) used in the formation of compound “D” is a sticky, viscous oil which is difficult to process (weigh, transfer, and blend). Moreover, step 1, as described in the ‘878 publication, requires isolation and chromatographic purification of compounds C and D prior to carrying out each subsequent derivatization reaction. In addition, as described in the ‘878 publication, the reaction of compound C with malonate diester is carried out using the diester as a solvent. After isolation and purification of the resultant malonate adduct, compound D, ring closure to form diketone compound E is carried out in methanol. In accordance with the procedure described in the ‘878 publication, compound E is isolated and dried, then converted to the corresponding dichloride in N,N-dimethyl aniline by treatment with phosphorous oxychloride (POCl3). The dichloride thus formed was isolated and purified by chromatography prior to the sequential amination reactions. Additionally, the compounds of Formula G and of Formula II require chromatography purification and isolations, as described in the ‘878 publication.

As further described in the ‘878 publication, each of the amination reactions were run separately with isolation and chromatographic purification between amination reactions. Accordingly, the ‘878 publication describes the preparation of the compound of Formula II utilizing a scheme consisting of five separate reaction steps with intervening isolation and purification of the products, each sequential step being carried out in a different solvent system. The overall yield of the compound of Formula II reported for this synthesis, based on starting compound C (Scheme II) is about 20%.

 

Example 1Preparation of Diketone Compound E (Scheme VI) 3-Ethylpyrazolo[1,5-a]pyrimidine-5,7(4H,6H)-dione

Figure US08076479-20111213-C00046

To a 250 ml, three-necked flask equipped with a thermometer, a reflux condenser and mechanical stirrer was charged 3-amino-4-ethylpyrazole oxalate (10 g, 50 mmole), dimethylmalonate (10 ml, 88 mmole), methyl alcohol (80 ml) and sodium methoxide (50 ml, 245 mmole, 25% in methyl alcohol). The batch was heated at reflux for 16 hours then cooled to room temperature. Celite (5 g) and water (60 ml) were added to the batch and agitated for 10 minutes. The batch was filtered to remove the solid residue. The filtrate was pH adjusted to pH˜3 with aqueous HCl (10 ml) to effect precipitation. The precipitate (compound “E”) was filtered and washed with water (40 ml). The wet cake was dried for 18 hours in vacuum oven maintained in the range of oven at 45° C. to 55° C., to give a solid product (84.3%, 7.5 g). C8H9N3O3, Mp: 200-205° C.; NMR in DMSO-d6: 1.05 (t, 3H), 2.23 (q, 2H), 3.26 (bs, 1H), 3.89 (bs, 1H), 7.61 (s, 1H), 11.50(bs, 1H).

Example 2Preparation of Dichloride Compound F (Scheme VI) 5,7-Dichloro-3-Ethylpyrazolo[1,5-a]pyrimidine

Figure US08076479-20111213-C00047

Into a 3-neck flask fitted with an inert gas inlet, a reflux condenser and a mechanical stirring apparatus and containing 83 liters of acetonitrile was placed 3-Ethylpyrazolo[1,5-a]pyrimidine-5,7(4H,6H)-dione (E) prepared as described in Step 1 (11.0 kg, 61.5 mole), N,N-dimethylaniline (8.0 L, 63 mole) and POCl3 (7 kg, 430 mole). With stirring the mixture was brought to reflux and maintained under refluxing conditions for 15 hours. The reaction mixture was sampled periodically to monitor the amount of compound “E” present. After the conversion was complete, the solution was cooled to 15° C. Into the cooled reaction mixture was added water which had been cooled to a temperature of less than 20° C. The product is filtered and washed with 4 aliquots of acetonitrile-water (1:3) which had been cooled to a temperature of 20° C. followed by a wash with 10× water. The wet cake is dried in a vacuum oven maintained at 40° C. for at least 15 hours to yield the compound “F” (86.7%); 1H NMR (CDCl3): 1.32(t, 3H), 2.81 (q, 2H), 6.92 (s, 1H), 8.10 (s, 1H)

mp: 90-95° C.

Example 3Preparation of Compound G (Scheme VI) 5-Chloro-3-Ethyl-N-[(1-oxido-pyridinyl)methyl]pyrazolo-[1,5-a]pyrimidine-5.7(4H,6H)-dion-7-amine

Figure US08076479-20111213-C00048

Into a 3-liter, three-necked flask equipped with a thermometer, a reflux condenser and mechanical stirrer was charged an aliquot of the dichloride compound “F” prepared in Step 2 (150 g, 0.69 mole), potassium phosphate tribasic monohydrate (338.0 g, 1.47 mole), the dihydrochloride salt of N-oxide-pyridin-3-yl-methylamine, compound F1a (142.5 g, 0.72 mole), water (1500 ml) and acetonitrile (300 ml). The batch was heated at reflux for 6 hours. At the end of the refluxing period the batch was cooled to room temperature over 2 hours and then held at room temperature for 4 hours. The resulting precipitate was filtered and washed with water (600 ml). The wet cake was returned to the flask with water (1500 ml) and acetonitrile (300 ml), and heated to reflux. Reflux was maintained for 6 hours additional. At the end of the second reflux period the reaction mixture was cooled to room temperature over a 2 hour period and left to stand at room temperature for 4 hours. The resulting precipitate was filtered and washed with water (600 ml). The wet cake was dried in an air draft oven at 50° C. for 18 hours to give the first amine adduct “G” material (179 g, 84.9%). mp: 187-189C; NMR in CDCl3, 1.26(t, 3H), 2.73(q, 2H), 4.60(d, 2H), 5.87(s, 1H), 6.83(bs, 1H), 7.33(t, 1H), 7.70(d, 1H), 7.84(s, 1H), 8.58(d, 1H), 8.64(d, 1H).

Example 4

Preparation of the Compound of Formula II (Scheme VI) 1-[3-Ethyl-7-[(1-oxido-3-pyridinyl)methyl]amino]pyrazolo[1,5-a]pyrimidin-5-yl]-2(s)-piperidinemethanol

Figure US08076479-20111213-C00049

Into a three-neck flask fitted with a mechanical stirrer and a reflux condenser were placed the first amine adduct prepared in Step 3, compound “G”, (7 kg, 23 mole), amino-alcohol compound G1a (5.6 kg, 43.3 mole), sodium carbonate (3.5 kg, 33.0 mole), 110 ml of water and 1-methyl-2-pyrrolidinone (NMP) (11 L). The reaction mixture was heated to 150° C. for 4 days. After chromatography indicated that the reaction was complete (90-95% substrate consumed), the reaction mixture was cooled to room temperature and quenched by adding water. The mixture was then extracted with ethyl acetate. The batch was dried by distillation of the water azeotrope under atmospheric pressure and concentrated to about 28 L volume. THF was added and the solution was heated to reflux until all the solids dissolve. Ethyl acetate and trietylamine are added to the hot solution. The batch was cooled to ambient and then agitated with the temperature maintained in the range of from 20° C. to 25° C. for 12 hours. The solids were collected by filtration, washed first with ethyl acetate then water, and dried in the filter under vacuum for 24 hours with the temperature maintained at from 40° C. to 50° C., yielding 4.9 kg, 51.3% of the compound of Formula II.

DSC, 168.6° C.; Specific Rotation (10 mg/ml in MeOH, 20° C.), −117.8 °;

1HNMR (400 MHz, DMSO): 8.31 ppm (1H, s), 8.11-8.13 ppm (1H, td, J=5.7 Hz, J=1.4 Hz), 7.97 ppm (1H, t, J=6.7 Hz), 7.68 ppm (1H, s), 7.41 ppm (1H, s), 7.37-7.43 ppm (1H, dd), 5.55 ppm (1H, s), 4.85 ppm (1H, t, J=5.4 Hz), 4.49-4.59 ppm (3H, m), 4.24-4.28 ppm (1H, broad), 3.27-3.46 ppm (2H, m), 2.76-2.83 ppm (1H, t, J=13.0 Hz), 2.45-2.50 ppm (2H, q, J=7.5 Hz), 1.72-1.79 (1H, m), 1.54-1.68 ppm (6H, m), 1.30-1.34 ppm (1H, m), 1.16 ppm (3H, t, J=7.5 Hz)

 

 

References

  1. Parry, D; Guzi, T; Shanahan, F; Davis, N; Prabhavalkar, D; Wiswell, D; Seghezzi, W; Paruch, K; Dwyer, M. P.; Doll, R; Nomeir, A; Windsor, W; Fischmann, T; Wang, Y; Oft, M; Chen, T; Kirschmeier, P; Lees, E. M. (2010). “Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor”. Molecular Cancer Therapeutics 9 (8): 2344–53. doi:10.1158/1535-7163.MCT-10-0324. PMID 20663931. edit
  2. Jump up^ Bose P, Simmons GL, Grant S (2013). “Cyclin-dependent kinase inhibitor therapy for hematologic malignancies”. Expert Opin Investig Drugs 22 (6): 723–38.doi:10.1517/13543784.2013.789859. PMC 4039040. PMID 23647051.
  3.  Martin, M. P.; Olesen, S. H.; Georg, G. I.; Schönbrunn, E (2013). “Cyclin-dependent kinase inhibitor dinaciclib interacts with the acetyl-lysine recognition site of bromodomains”. ACS Chemical Biology 8 (11): 2360–5. doi:10.1021/cb4003283. PMC 3846258. PMID 24007471. edit
  4.  Nguyen, T. K.; Grant, S (2013). “Dinaciclib (SCH727665) inhibits the unfolded protein response (UPR) through a CDK1 and CDK5-dependent mechanism”. Molecular Cancer Therapeutics 13(3): 662–74. doi:10.1158/1535-7163.MCT-13-0714. PMID 24362465. edit
  5. Jump up^ Desai, B. M.; Villanueva, J; Nguyen, T. T.; Lioni, M; Xiao, M; Kong, J; Krepler, C; Vultur, A; Flaherty, K. T.; Nathanson, K. L.; Smalley, K. S.; Herlyn, M (2013). “The anti-melanoma activity of dinaciclib, a cyclin-dependent kinase inhibitor, is dependent on p53 signaling”. PLoS ONE 8 (3): e59588. doi:10.1371/journal.pone.0059588. PMC 3601112. PMID 23527225. edit
  6. Jump up^ Johnson, A. J.; Yeh, Y. Y.; Smith, L. L.; Wagner, A. J.; Hessler, J; Gupta, S; Flynn, J; Jones, J; Zhang, X; Bannerji, R; Grever, M. R.; Byrd, J. C. (2012). “The novel cyclin-dependent kinase inhibitor dinaciclib (SCH727965) promotes apoptosis and abrogates microenvironmental cytokine protection in chronic lymphocytic leukemia cells”. Leukemia 26 (12): 2554–7.doi:10.1038/leu.2012.144. PMC 3645353. PMID 22791353. edit
  7. Jump up^ Feldmann, G; Mishra, A; Bisht, S; Karikari, C; Garrido-Laguna, I; Rasheed, Z; Ottenhof, N. A.; Dadon, T; Alvarez, H; Fendrich, V; Rajeshkumar, N. V.; Matsui, W; Brossart, P; Hidalgo, M; Bannerji, R; Maitra, A; Nelkin, B. D. (2011). “Cyclin-dependent kinase inhibitor Dinaciclib (SCH727965) inhibits pancreatic cancer growth and progression in murine xenograft models”.Cancer biology & therapy 12 (7): 598–609. PMC 3218385. PMID 21768779. edit
  8. Jump up^ Fu, W; Ma, L; Chu, B; Wang, X; Bui, M. M.; Gemmer, J; Altiok, S; Pledger, W. J. (2011). “The cyclin-dependent kinase inhibitor SCH 727965 (dinacliclib) induces the apoptosis of osteosarcoma cells”. Molecular Cancer Therapeutics 10 (6): 1018–27. doi:10.1158/1535-7163.MCT-11-0167. PMID 21490307. edit
  9. Jump up^ Fu, W; Sharma, S. S.; Ma, L; Chu, B; Bui, M. M.; Reed, D; Pledger, W. J. (2013). “Apoptosis of osteosarcoma cultures by the combination of the cyclin-dependent kinase inhibitor SCH727965 and a heat shock protein 90 inhibitor”. Cell Death and Disease 4 (3): e566. doi:10.1038/cddis.2013.101. PMC 3613821. PMID 23538447. edit
  10. Jump up^ Nemunaitis, J. J.; Small, K. A.; Kirschmeier, P; Zhang, D; Zhu, Y; Jou, Y. M.; Statkevich, P; Yao, S. L.; Bannerji, R (2013). “A first-in-human, phase 1, dose-escalation study of dinaciclib, a novel cyclin-dependent kinase inhibitor, administered weekly in subjects with advanced malignancies”. Journal of Translational Medicine 11 (1): 259. doi:10.1186/1479-5876-11-259.PMC 3853718. PMID 24131779. edit
  11. Jump up^ Mita, M; Joy, A. A.; Mita, A; Sankhala, K; Jou, Y. M.; Zhang, D; Statkevich, P; Zhu, Y; Yao, S. L.; Small, K; Bannerji, R; Shapiro, C. L. (2013). “Randomized Phase II Trial of the Cyclin-Dependent Kinase Inhibitor Dinaciclib (MK-7965) Versus Capecitabine in Patients with Advanced Breast Cancer”. Clinical Breast Cancer 14 (3): 169–76. doi:10.1016/j.clbc.2013.10.016.PMID 24393852. edit
  12. Jump up^ Stephenson, J. J.; Nemunaitis, J; Joy, A. A.; Martin, J. C.; Jou, Y. M.; Zhang, D; Statkevich, P; Yao, S. L.; Zhu, Y; Zhou, H; Small, K; Bannerji, R; Edelman, M. J. (2014). “Randomized phase 2 study of the cyclin-dependent kinase inhibitor dinaciclib (MK-7965) versus erlotinib in patients with non-small cell lung cancer”. Lung Cancer 83 (2): 219–23.doi:10.1016/j.lungcan.2013.11.020. PMID 24388167. edit

External links

DINACICLIB

Patent                                                         Submitted                                                                Granted

Process and intermediates for the synthesis of (3-alkyl-5-piperidin-1-yl-3,3a-dihydro-pyrazolo[1,5-a]pyrimidin-7-yl)-amino derivatives and intermediates [US8076479]2008-03-06   GRANT2011-12-13

Process for resolving chiral piperidine alcohol and process for synthesis of pyrazolo[1,5-a] pyrimidine derivatives using same [US7786306]2008-02-28   GRANT2010-08-31

Sequential Administration of Chemotherapeutic Agents for Treatment of Cancer [US2011129456]2011-06-02

TARGETING CDK4 AND CDK6 IN CANCER THERAPY [US2011009353]2011-01-13

Pyrazolopyrimidines as cyclin dependent kinase inhibitors [US2007225270]2007-09-27

PYRAZOLO[1,5-a]PYRIMIDINES [US2007275963]2007-11-29

Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors [US2007281951]2007-12-06

Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors [US2008050384]2008-02-28

Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors [US2007054925]2007-03-08

Entinostat


 

Entinostat.png

Entinostat

Also known as: ms-275, 209783-80-2, SNDX-275, MS 275, MS-27-275, SNDX 275, NSC-706995,
  • BAY 86-5274
  • BAY86-5274
Molecular Formula: C21H20N4O3
Molecular Weight: 376.4085 g/mol
pyridin-3-ylmethyl N-[[4-[(2-aminophenyl)carbamoyl]phenyl]methyl]carbamate
N-(2-aminophenyl)-4-[N-(pyridine-3-yl)-methoxycarbonyl-aminomethyl]- benzamide

CAS  209783-80-2

209784-80-5 (HCl)

Bayer Schering Pharma Aktiengesellschaft

Pyridin-3-ylmethyl N-[[4-[(2-aminophenyl)carbamoyl]phenyl]methyl]carbamate

Entinostat, developed by Syndax Pharmaceuticals, is an oral selective histone deacetylase (HDAC) inhibitor primarily targeting class IHDACs (HDAC1, HDAC2, and HDAC3) . It was later licensed to
Jiangsu Hengrui Medicine Co., Ltd., for development and commercialization in China. In 2024, Entinostat has been approved by the NMPA for use in combination with exemestane to treat advanced breast cancer that is HR-positive and HER2-negative.

News…………http://www.prnewswire.com/news-releases/kyowa-hakko-kirin-and-syndax-announce-an-exclusive-license-agreement-to-develop-and-commercialize-entinostat-in-japan-and-korea-300017491.html

KHK and Syndax partner for breast cancer treatment entinostat in Japan and Korea
Japan-based Kyowa Hakko Kirin (KHK) has signed a license agreement with US-based Syndax Pharmaceuticals for the exclusive rights to develop and commercialise entinostat in Japan and Korea.

TOKYO and WALTHAM, Mass., Jan. 7, 2015 /PRNewswire/ — Kyowa Hakko Kirin Co., Ltd., (Headquarters: Chiyoda-ku, Tokyo; president and CEO: Nobuo Hanai, “Kyowa Hakko Kirin”) and Syndax Pharmaceuticals, Inc., (Waltham, Mass.; president and CEO:Arlene M. Morris, “Syndax”) today jointly announced that the companies have entered into a license agreement for the exclusive rights to develop and commercialize entinostat in Japan and Korea. Entinostat is a Class I selective histone deacetylase (HDAC) inhibitor being developed by Syndax in the United States and Europe in combination with hormone therapy for advanced breast cancer and immune therapy combinations in solid tumors.

 

Entinostat.png

Entinostat, also known as SNDX-275 and MS-275, is a benzamide histone deacetylase inhibitor undergoing clinical trials for treatment of various cancers.[1]

Entinostat inhibits class I HDAC1 and HDAC3 with IC50 of 0.51 μM and 1.7 μM, respectively.[2]

Entinostat (formerly known as MS-275) is a histone deacetylase (HDAC) inhibitor in phase III clincal trials at Syndax in combination with exemestane for the treatment of advanced HR-positive breast cancer.

Entinostat (MS-275) preferentially inhibits HDAC1 (IC50=300nM) over HDAC3 (IC50=8µM) and has no inhibitory activity towards HDAC8 (IC50>100µM). MS-275 induces cyclin-dependent kinase inhibitor 1A (p21/CIP1/WAF1), slowing cell growth, differentiation, and tumor development in vivo. Recent studies suggest that MS-275 may be particularly useful as an antineoplastic agent when combined with other drugs, like adriamycin.

In September 2013, Syndax Pharmaceuticals entered into a licensing, development and commercialization agreement with Eddingpharm in China and other asian countries. In 2013, a Breakthrough Therapy Designation was assigned to the compound for the treatment of locally recurrent or metastatic estrogen receptor-positive (ER+) breast cancer when added to exemestane in postmenopausal women whose disease has progressed following non-steroidal aromatase inhibitor therapy.

Clinical trials

There is an ongoing phase II trial studying the effect of entinostat on Hodgkin’s lymphoma.[3] It is in other phase II trials for advanced breast cancer (in combination with aromatase inhibitors)[4] and for metastatic lung cancer (in combination with erlotinib).[5] As of September 2013, the Food and Drug Administration is working with the industry to design phase III clinical trials. They seek to evaluate the application of Entinostat for the reduction, or prevention of, treatment resistance to aromatase inhibitors in hormone receptor positive breast cancer.[6] Syndax pharmaceuticals currently holds the rights to Entinostat and recently received $26.6 million in funds to advance treatments of resistant cancers using epigenetic tools.[7]

PHASE 3………..SYNDAX, BREAST CANCER

SYN


European Journal of Medicinal Chemistry 291 (2025) 117643

Entinostat, developed by Syndax Pharmaceuticals, is an oral selec
tive histone deacetylase (HDAC) inhibitor primarily targeting class I
HDACs (HDAC1, HDAC2, and HDAC3) [7]. It was later licensed to
Jiangsu Hengrui Medicine Co., Ltd., for development and commercial
ization in China. In 2024, Entinostat has been approved by the NMPA for
use in combination with exemestane to treat advanced breast cancer that
is HR-positive and HER2-negative. This approval is specifically for pa
tients whose disease has progressed following prior endocrine therapy
[8]. Entinostat inhibits HDACs, increasing histone acetylation and
reactivating tumor suppressor genes. This mechanism restores sensi
tivity to endocrine therapy and prevents cancer cell proliferation [9].
The therapeutic agent exerts its effects by modulating the tumor
microenvironment through the suppression of immune regulatory cells,
thereby augmenting the immune response. Its clinical efficacy was
confirmed in the E2112 trial (NCT02115282), a global Phase III study.
When used in combination with exemestane, Entinostat demonstrated
the ability to extend PFS in patients with HR-positive, HER2-negative
breast cancer [10]. The median PFS was significantly extended to 6.32
months, contrasting with the 3.72 months observed in the control
cohort. In terms of safety profile, Entinostat demonstrated favorable
tolerability. The frequently encountered adverse events were primarily
neutropenia, fatigue, and nausea. Severe neutropenia occurred in 43 %
of patients but was manageable with supportive care. Liver function
abnormalities were reported but manageable with dose adjustments
[11].
The synthetic route of Entinostat is shown in Scheme 2 [12].
Enti-001 is first treated with trifluoroacetic anhydride to afford
Enti-002. Reaction of Enti-002 with oxalyl chloride yields the acyl
chloride intermediate, which undergoes condensation with Enti-003 to
form Enti-004. Subsequent alkaline hydrolysis of Enti-004 produces
Enti-005. This compound is activated with CDI followed by reaction
with Enti-006 to generate Enti-007. The synthesis concludes with acidic removal of the Boc protecting group from Enti-007, yielding Entinostat

[8] W. Li, Z. Sun, Mechanism of action for HDAC inhibitors-insights from omics
approaches, Int. J. Mol. Sci. 20 (2019) 1616.
[9] N. Bharathy, N.E. Berlow, E. Wang, J. Abraham, T.P. Settelmeyer, J.E. Hooper, M.
N. Svalina, Z. Bajwa, M.W. Goros, B.S. Hernandez, J.E. Wolff, R. Pal, A.M. Davies,
A. Ashok, D. Bushby, M. Mancini, C. Noakes, N.C. Goodwin, P. Ordentlich, J. Keck,
D.S. Hawkins, E.R. Rudzinski, A. Mansoor, T.J. Perkins, C.R. Vakoc, J.E. Michalek,
C. Keller, Preclinical rationale for entinostat in embryonal rhabdomyosarcoma,
Skelet Muscle 9 (2019) 12.
[10] B. Xu, Q. Zhang, X. Hu, Q. Li, T. Sun, W. Li, Q. Ouyang, J. Wang, Z. Tong, M. Yan,
H. Li, X. Zeng, C. Shan, X. Wang, X. Yan, J. Zhang, Y. Zhang, J. Wang, L. Zhang,
Y. Lin, J. Feng, Q. Chen, J. Huang, L. Zhang, L. Yang, Y. Tian, H. Shang, Entinostat,
a class I selective histone deacetylase inhibitor, plus exemestane for Chinese
patients with hormone receptor-positive advanced breast cancer: a multicenter,
randomized, double-blind, placebo-controlled, phase 3 trial, Acta Pharm. Sin. B 13
(2023) 2250–2258.
[11] E.T. Roussos Torres, W.J. Ho, L. Danilova, J.A. Tandurella, J. Leatherman, C. Rafie,
C. Wang, A. Brufsky, P. LoRusso, V. Chung, Y. Yuan, M. Downs, A. O’Connor, S.
M. Shin, A. Hernandez, E.L. Engle, R. Piekarz, H. Streicher, Z. Talebi, M.A. Rudek,
Q. Zhu, R.A. Anders, A. Cimino-Mathews, E.J. Fertig, E.M. Jaffee, V. Stearns, R.
M. Connolly, Entinostat, nivolumab and ipilimumab for women with advanced
HER2-negative breast cancer: a phase Ib trial, Nat Cancer 5 (2024) 866–879.
[12] T. Suzuki, T. Ando, K. Tsuchiya, T. Nakanishi, A. Saito, S. Yamashita, G. Shiraishi,
E. Tanaka, Preparation of Benzamide Derivatives as Anticancer Agents, 1998
JP10152462

SEE SCHEME AT END

 

Patent

http://www.google.im/patents/WO2010022988A1?cl=en

In EP 0 847 992 A1 (which co-patent is US 6,794,392) benzamide derivatives as medicament for the treatment of malignant tumors, autoimmune diseases, de- rmatological diseases and parasitism are described. In particular, these derivatives are highly effective as anticancer drugs, preferred for the haematological malignancy and solid tumors. The preparation of N-(2-aminophenyl)-4-[N- (pyridine-3-yl)methoxycarbonylaminomethyl]-benzamide is described on page 57, Example 48. The compound is neither purified by chromatography nor purified by treatment with charcoal. The final step of the process comprises the re- crystallization from ethanol.

Said compound has a melting point (mp) of 159 – 160 0C.

The IR spectrum shows the following bands: IR(KBr) cm“1: 3295, 1648, 1541 , 1508, 1457, 1309, 1183, 742.

The data indicate the Polymorph A form.

In EP 0 974 576 B1 a method for the production of monoacylated phenylenediamine derivatives is described. The preparation of N-(2- aminophenyl)-4-[N-(pyridine-3-yl)methoxycarbonylamino-methyl] benzamide is described on pages 12 to 13, Example 6. The final step of the process comprises the purification of the compound via silica gel column chromatography.

Said compound has a melting point (mp) of 159 – 160 0C.

The IR spectrum shows the following bands: IR(KBr) cm‘1: 3295, 1648, 1541 , 1508, 1457, 1309, 1183, 742.

The data indicate the Polymorph A form. In J. Med. Chem. 1999, 42, 3001-3003, the synthesis of new benzamide derivatives and the inhibition of histone deacetylase (HDAC) is described. The process for the production of N-(2-aminophenyl)-4-[N-(pyridine-3-yl) meth- oxycarbonylaminomethyl] benzamide is described. The final step of the process comprises the purification of the compound via silica gel column chromatography (ethyl acetate).

Said compound has a melting point (mp) of 159 – 160 0C.

The IR spectrum shows the following bands: IR(KBr) cm‘1: 3295, 1648, 1541 , 1508, 1457, 1309, 1183, 742.

The data indicate the Polymorph A form.

In WO 01/12193 A1 a pharmaceutical formulation comprising N-(2- aminophenyl)-4-[N-(pyridine-3-yl)methoxycarbonylamino-methyl]benzamide is described.

In WO 01/16106 a formulation comprising N-(2-aminophenyl)-4-[N-(pyridine-3- yl)methoxycarbonylamino-methyl]benzamide, having an increased solubility and an improved oral absorption for benzamide derivatives, and pharmaceutically acceptable salts thereof are described.

In WO 2004/103369 a pharmaceutical composition is described which comprises histone deacetylase inhibitors. That application concerns the combined use of N-(2-aminophenyl)-4-[N-(pyridine-3-yl)methoxycarbonylamino- methyl]benzamide together with different cancer active compounds. In fact that application is a later application, which is based on the above mentioned matter and thus concerns the Polymorph A form. Finally, JP 2001-131130 (11-317580) describes a process for the purification of monoacylphenylenediamine derivatives. In Reference Example 2, the process for the production of crude N-(2-aminophenyl)-4-[N-(pyridine-3-yl) meth-oxycarbonylaminomethyl] benzamide is described. Said compound has a melting point (mp) of 159 – 160 0C,

The IR spectrum shows the following bands: IR(KBr) cm“1: 3295, 1648, 1541 , 1508, 1457, 1309, 1183, 742.

The data indicate the Polymorph A form.

Moreover, Working Example 1 describes the purification of crude N-(2- aminophenyl)-4-[N-(pyridine-3-yl) methoxycarbonylaminomethyl] benzamide in aqueous acid medium together with carbon The final crystallization is done under aqueous conditions at 40-500C.

Following the description to that example it can be seen from the Comparative Examples 1 – 3 that the crude N-(2-aminophenyl)-4-[N-(pyridine-3-yl) meth- oxycarbonylaminomethyl] benzamide is not purified by dissolution under reflux conditions in either ethanol, methanol or acetonithle followed by a recrystalliza- tion at 2°C. As a result, these recrystallisations do not yield any pure compound.

In addition a “purification” of crude N-(2-aminophenyl)-4-[N-(pyridine-3-yl) methoxycarbonylaminomethyl] benzamide in ethanol under reflux conditions to- gether with carbon is dechbed. After filtering off the carbon the compound is re- crystallized at 2°C. The purification effect of this method is very limited. 1 ,1 % of an impurity remain in the N-(2-aminophenyl)-4-[N-(pyridine-3-yl) methoxycarbonylaminomethyl] benzamide. As a result, this procedure does not yield any pure compound.

None of the state of the art documents refer to a polymorph B of N-(2- aminophenyl)-4-[N-(pyridine-3-yl)methoxycarbonylamino-methyl]benzamide and no physicochemical features of said compound are known. Several biological and clinical studies have been done with N-(2-aminophenyl)- 4-[N-(pyridine-3-yl) meth-oxycarbonylaminomethyl] benzamide. For example, Kummar et al., Clin Cancer Res. 13 (18), 2007, pp 5411-5417 describe a phase I trial of N-(2-aminophenyl)-4-[N-(pyridine-3-yl) meth-oxycarbonylaminomethyl] benzamide in refractory solid tumors. The compound was applied orally.

The crude N-(2-aminophenyl)-4-[N-(pyridine-3-yl)methoxycarbonylaminomethyl]- benzamide of step a) can be produced according to the method described in example 6 of EP 0974 576 B1.

PATENT

http://www.google.co.in/patents/EP0974576A2?cl=en

Example 6Synthesis of N-(2-aminophenyl)-4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzamide (an example in which after activation with N,N’-carbonyldiimidazole, an acid was added to carry out reaction)

  • [0082]
    7.78 g (48 mmole) of N,N’-carbonyldiimidazole were added to a 1,3-dimethyl-2-imidazolidinone (50 g) suspension including 11.45 g (40 mmole) of 4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic acid. After stirring at room temperature for 2 hours, 17.30 g (0.16 mole) of 1,2-phenylenediamine were added to the solution. After cooling to 2°C, 9.60 g (0.1 mole) of methanesulfonic acid were added dropwise. After stirring for 2 hours, water was added, and the deposited solid was collected by filtration. Purification was then carried out through silica gel column chromatography to obtain 10.83 g (yield: 72%) of N-(2-aminophenyl)-4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzamide.
    Reaction selectivity based on the result in HPLC
      Retention Time/min. Area %
    Benzoylimidazole as Active Intermediate 4.3 0.00
    Monoacylated Phenylenediamine 4.7 98.91
    Diacylated Phenylenediamine 11.7 1.09

    Analysis data of the product
    mp. 159-160°C
       1H NMR (270MHz, DMSO-d6) δ ppm: 4.28 (2H, d, J=5.9Hz), 4.86 (2H, s), 5.10 (2H, s), 6.60 (1H, t, J=7.3Hz), 6.78 (1H, d, J=7Hz), 6.97 (1H, t, J=7Hz), 7.17 (1H, d, J=8Hz), 7.3-7.5 (3H, m), 7.78 (1H, d, J=8Hz), 7.93 (2H, d, J=8Hz), 8.53 (1H, d, J=3.7Hz), 8.59 (1H, s), 9.61 (1H, s).
       IR (KBr) cm-1: 3295, 1648, 1541, 1508, 1457, 1309, 1183, 742

PATENT

WO 2009076206

http://www.google.com/patents/WO2009076206A1?cl=en

Suzuki et al (Suzuki et al Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives, J Med Chem 1999, 42, (15), 3001-3) discloses benzamide derivatives having histone deacetylase inhibitory activity and methods of making benzamide derivatives having histone deacetylase inhibitory activity. Suzuki et al is hereby incorporated herein by reference in its entirety.

[18] An example of the synthesis method of Suzuki et al to produce MS-275 via a three- step procedure in 50.96% overall yield is outlined in Scheme 3 below.

Scheme 3: Previous Procedure for Synthesis of MS-275 en rt, 4h

(used without purification)

[Overall yield: 0.91 x 0.56 x 100 = 50.96%;

MS-275 [19] In addition to the modest overall yield, the procedure of Suzuki et al has other disadvantages, such as a tedious method for the preparation of an acid chloride using oxalyl chloride and requiring the use of column chromatography for purification.

The synthesis of MS-275 is shown below in Scheme 4 as an example of Applicants invention of a two-step procedure: [37] Scheme 4: Preparation of MS-275

Scheme 4: New Synthesis of MS-275 (4)

Condensation of 3-(hydroxymethyl)pyridine (7) and 4-(aminomethyl)benzoic in the presence of CDI gave 4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic Acid (8) in 91.0% yield. In the previous method of Suzuki et ah, the carboxylic acid derivative 8 was first converted into acyl chloride hydrochloride by treatment of oxalyl chloride in toluene and then reacted with imidazole to form the acylimidazole intermediate. (Suzuki et al., Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives. J Med Chem 1999, 42, (15), 3001-3.). However, Applicants synthesized the imidazolide of intermediate 8 by treatment with CDI at about 55-60 0C in THF. The imidazolide was cooled to ambient and further reacted in situ with 1,2-phenylenediamine in the presence of TFA to afford MS-275

(4).

Experimental Section

[62] iV-(2-Aminophenyl)-4-[iV-(pyridin-3-ylmethoxycarbonyl) aminomethyl] benzamide (4, MS-275).

[63] To a suspension of 4-[N-(Pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic

Acid (5.0 g, 0.017 mol) in THF (100 mL) was added CDI (3.12 g, 0.019 mol), and the mixture stirred for 3 h at 60 0C. After formation of acylimidazole the clear solution was cooled to room temperature (rt). To this was added 1,2-phenylenediamine (15.11 g, 0.14 mmol) and trifluoroacetic acid (1.2 mL, 0.015 mol) and then stirred for 16 h. The reaction mixture was evaporated to remove THF and crude product was stirred in a mixture of hexane and water (2:5, v/v) for 1 h and filtered and dried. The residue was stirred in dichloromethane twice to afford pure MS-275 (4) as off white powder 5.25 g, 80% yield:

mp 159-160 * C; IR (KBr) 3295, 1648, 1541, 1508, 1457, 1309, 1183, 742 cm“1.

1H NMR (DMSO-J6) δ 4.28 (d, 2H, J = 5.9 Hz), 4.86 (s, 2H), 5.10 (s, 2H), 6.60 (t, IH, J = 7.3 Hz), 6.78 (d, IH, J = 7 Hz), 6.97 (t, IH, J= 7 Hz), 7.17 (d, IH, J= 8 Hz), 7.3-7.5(m, 3H), 7.78 (d, IH, J= 8 Hz), 7.93 (d, 2H, J = 8 Hz), 8.53 (d, IH, J = 3.7 Hz), 8.59 (s, IH), 9.61 (s, IH);

HRMS: calcd 376.1560 (C2iH2oN4θ3), found 376.1558. These spectral and analytical data are as previously reported in J Med Chem 1999, 42, (15), 3001-3.

[64] 4-[7V-(Pyridin-3-ylmethoxycarbonyI)aminomethyl] benzoic Acid (8) may be prepared as follows. To a suspension of l, l’-carbonyldiimidazole (CDI, 25.6 g, 158 mmol) in THF (120 mL) was added 3-pyridinemethanol (7, 17.3 g, 158 mmol) in THF (50 mL) at 10 0C, and the mixture stirred for 1 h at rt. The resulting solution was added to a suspension of 4-(aminomethyl)benzoic acid (22.6 g, 158 mmol), DBU (24.3 g, 158 mmol), and triethylamine (22.2 mL, 158 mmol) in THF (250 mL). After stirring for 5 h at rt, the mixture was evaporated to remove THF and then dissolved in water (300 mL). The solution was acidified with HCl (pH 5) to precipitate a white solid which was collected by filtration, washed with water (300 mL) and methanol (50 mL), respectively, and dried to yield pure 8 (41.1 g, 91% yield):

mp 207-208 0 C;

IR (KBr) 3043, 1718, 1568, 1434, 1266, 1 108, 1037, 984, 756 cm4; 1H NMR (DMSO-^6) δ 4.28 (d, 2H, J= 5.9 Hz), 5.10 (s, 2H), 7.3-7.5 (m, 3H), 7.7-8.1 (m, 4H), 8.5-8.7 (m, 2H). These spectral and analytical data are as previously reported in Suzuki et al, J Med Chem 1999, 42, (15), 3001-3.

PAPER

Volume 18, Issue 11, 1 June 2010, Pages 3925–3933

http://www.sciencedirect.com/science/article/pii/S0968089610003378

PAPER

see

Bioorg Med Chem 2008, 16(6): 3352

http://www.sciencedirect.com/science/article/pii/S0968089607010577

PAPER

see

Bioorganic and Medicinal Chemistry Letters, 2004 ,  vol. 14,   1  pg. 283 – 287

http://www.sciencedirect.com/science/article/pii/S0960894X03010539

PAPER

J Med Chem 1999, 42(15): 3001

http://pubs.acs.org/doi/abs/10.1021/jm980565u

N-(2-Aminophenyl)-4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzamide (1, MS-275). To a solution of imidazole (0.63 g, 9.2 mmol) in THF (20 mL) was added 3 (1 g, 2.9 mmol), and the mixture stirred for 1 h at room temperature. After imidazole hydrochloride was removed by filtration, 1,2-phenylenediamine (2.52 g, 23.2 mmol) and trifluoroacetic acid (0.2 mL, 2.6 mmol) were added to the filtrate and stirred for 15 h. The reaction mixture was evaporated to remove THF and partitioned between ethyl acetate (500 mL) and water (400 mL). The organic layer was washed with water and dried and then purified by silica gel column chromatography (ethyl acetate) to give 1 (0.62 g, 56% yield):

mp 159−160 °C;

1H NMR (DMSO-d6) δ 4.28 (d, 2H, J = 5.9 Hz), 4.86 (s, 2H), 5.10 (s, 2H), 6.60 (t, 1H, J = 7.3 Hz), 6.78 (d, 1H, J = 7 Hz), 6.97 (t, 1H, J = 7 Hz), 7.17 (d, 1H, J = 8 Hz), 7.3−7.5(m, 3H), 7.78 (d, 1H, J = 8 Hz), 7.93 (d, 2H, J = 8 Hz), 8.53 (d, 1H, J = 3.7 Hz), 8.59 (s, 1H), 9.61 (s, 1H);

IR (KBr) 3295, 1648, 1541, 1508, 1457, 1309, 1183, 742 cm-1.

Anal. (C21H20N4O3) C, H, N.

………………………………………………………………………..

see

Bulletin of the Korean Chemical Society, 2014 ,  vol. 35,   1  pg. 129 – 134

http://koreascience.or.kr/article/ArticleFullRecord.jsp?cn=JCGMCS_2014_v35n1_129

PAPER

see

ChemMedChem, 2013 ,  vol. 8,   5  pg. 800 – 811

http://onlinelibrary.wiley.com/doi/10.1002/cmdc.201300005/abstract;jsessionid=9D48E064CF53253495185AE2030C67BF.f02t03

PAPER

see

ACS Medicinal Chemistry Letters, 2013 ,  vol. 4,   10  pg. 994 – 999

http://pubs.acs.org/doi/full/10.1021/ml400289e

References

  1. Phase I trial of 5-azacitidine (5AC) and SNDX-275 in advanced lung cancer (NSCLC)
  2. Novel Sulphonylpyrroles as Inhibitors of Hdac S Novel Sulphonylpyrroles
  3. A Phase 2 Multi-Center Study of Entinostat (SNDX-275) in Patient With Relapsed or Refractory Hodgkin’s Lymphoma
  4. A Phase 2, Multicenter Study of the Effect of the Addition of SNDX-275 to Continued Aromatase Inhibitor (AI) Therapy in Postmenopausal Women With ER+ Breast Cancer Whose Disease is Progressing
  5. A Phase 2 Exploratory Study of Erlotinib and SNDX-275 in Patients With Non-small Cell Lung Carcinoma Who Are Progressing on Erlotinib
  6. Breakthrough Designation Granted to Entinostat for Advanced Breast Cancer Silas Inman Published Online: Wednesday, September 11, 2013 http://www.onclive.com/web-exclusives/Breakthrough-Designation-Granted-to-Entinostat-for-Advanced-Breast-Cancer
  7. http://www.syndax.com/assets/130827%20Syndax%20Series%20B%20news%20release.pdf
  8. References:
    1. Saito, A. et al. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA 96 4592-4597 (1999).
    2. Jaboin, J., et al. MS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors. Cancer Res 62 6108-6115 (2002).
    3. Rosato RR, et al. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res 2003; 63: 3637–3645.
 
Cited Patent Filing date Publication date Applicant Title
EP0847992B1 * Sep 30, 1997 Jun 23, 2004 Schering Aktiengesellschaft Benzamide derivatives, useful as cell differentiation inducers
US7244751 * Feb 2, 2004 Jul 17, 2007 Shenzhen Chipscreen Biosciences Ltd. N-(2-amino-5-fluorophenyl)-4-[N-(Pyridn-3-ylacryloyl)aminomethyl]benzamide or other derivatives for treating cancer and psoriasis
 
Reference
1 * MAI A: ‘Histone deacetylation in epigenetics: an attractive target for anticancer therapy‘ MED RES REV. vol. 25, no. 3, May 2005, pages 261 – 309
2 * SUZUKI T ET AL.: ‘Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives‘ J MED CHEM. vol. 42, no. 15, 29 July 1999, pages 3001 – 3003
Names
Preferred IUPAC name(Pyridin-3-yl)methyl ({4-[(2-aminophenyl)carbamoyl]phenyl}methyl)carbamate
Other namesSNDX-275; MS-275
Identifiers
CAS Number209783-80-2 
3D model (JSmol)Interactive image
ChEBICHEBI:132082 
ChEMBLChEMBL27759 
ChemSpider4111 
ECHA InfoCard100.158.999 
IUPHAR/BPS7007
KEGGD09338 
PubChem CID4261
UNII1ZNY4FKK9H 
CompTox Dashboard (EPA)DTXSID0041068 
InChI☒☒
SMILES
Properties
Chemical formulaC21H20N4O3
Molar mass376.4085 g/mol
Pharmacology
ATC codeL01XH05 (WHO)
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).☒verify (what is ?)Infobox references

/////////////////approvals 2024, china 2024, Entinostat, SNDX 275, MS 275, Syndax, BAY 86-5274, BAY86-5274

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……