AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

MELOGLIPTIN

 diabetes, phase 2, Uncategorized  Comments Off on MELOGLIPTIN
Jul 032015
 

 

GRC 8200; 868771-57-7, EMD-675992

4-fluoro-1-[2-[[(1R,3S)-3-(1,2,4-triazol-1-ylmethyl)cyclopentyl]amino]acetyl]pyrrolidine-2-carbonitrile

4(S)-Fluoro-1-[2-[(1R,3S)-3-(1H-1,2,4-triazol-1-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile

GRC-8200, a dipeptidyl peptidase IV inhibitor (DPP-IV), is currently undergoing phase II clinical trials at Glenmark Pharmaceuticals and Merck KGaA for the treatment of type 2 diabetes. In 2006, the compound was licensed by Glenmark Pharmaceuticals to Merck KGaA in Europe, Japan and N. America for the treatment of type 2 diabetes, however, these rights were reaquired by Glenmark in 2008.

.

ALTERNATE……….

 

 

 

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

Share
Jul 032015
 

 

ChemSpider 2D Image | Evogliptin | C19H26F3N3O3

 

EVOGLIPTIN
CAS: 1222102-29-5 FREE

HCL……

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

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

 

 

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

 

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

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

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

 

Share
Jul 032015
 

ChemSpider 2D Image | gosogliptin | C17H24F2N6O

GOSOGLIPTIN

CAS 869490-23-3 FREE BASE

DIHYDROCHLORIDE..869490-47-1

GOSOGLIPTIN; UNII-GI718UO477;  PF-00734200; PF-734200;

(3,3-difluoropyrrolidin-1-yl)-[(2S,4S)-4-(4-pyrimidin-2-ylpiperazin-1-yl)pyrrolidin-2-yl]methanone

Molecular Formula: C17H24F2N6O
Molecular Weight: 366.408866 g/mol
Company Pfizer Inc.
Description Dipeptidyl peptidase-4 (DPP-4) inhibitor
Molecular Target Dipeptidyl peptidase-4 (DPP-4) (CD26) 
Mechanism of Action Dipeptidyl peptidase-4 (DPP-4) inhibitor
Latest Stage of Development Phase II
Standard Indication Diabetes
Indication Details Treat Type II diabetes

Type 2 diabetes mellitus is a chronic disorder characterized by hyperglycemia coupled with a gradual decline in insulin sensitivity and insulin secretion. The incretin hormone glucagon-like peptide-1 (GLP-1), which is released post-prandially from the L-cells of the intestine, stimulates the release of insulin from pancreatic β-cells. However, GLP-1 is rapidly degraded in vivo by peptidases, including dipeptidyl peptidase IV (DPP-4), which is a widely distributed serine protease that specifically cleaves N-terminal dipeptides from polypeptides with proline or alanine at the penultimate position.

In vivo administration of DPP-4 inhibitors to human subjects results in higher circulating concentrations of endogenous GLP-1 and subsequent decrease in plasma glucose. Long term treatment with a DPP-4 inhibitor leads to a reduction in circulating HbA1c (glycosylated hemoglobin). DPP-4 inhibition also offers the potential to improve the insulin producing function of the pancreas through either β-cell preservation or regeneration. Therefore, DPP-4 inhibition has emerged as a promising new treatment of Type 2 diabetes

PF-734200 is a potent, selective, orally active dipeptidyl peptidase IV inhibitor. It had been in phase II clinical development at Pfizer for the treatment of type 2 diabetes; however, in 2010 the company discontinued these trials. In 2012, the product was licensed to SatRx, a spin-off of the ChemRar High Tech Center, by Pfizer on an exclusive worldwide basis (with the exception of China) for the development and commercialization as monotherapy or in combination with other therapies for the treatment of type 2 diabetes. SatRx is conducting phase II clinical trials for the treatment of type 2 diabetes.

GOSOGLIPTIN.png

……………………….

PAPER

New synthetic route to a dipeptidyl peptidase-4 inhibitor
Org Process Res Dev 2012, 16(3): 409

http://pubs.acs.org/doi/abs/10.1021/op200309z

Abstract Image

A new synthetic route to a dipeptidyl peptidase-4 (DPP4) inhibitor was developed and demonstrated on a multigram scale. This approach takes advantage of the cheap and readily available Boc-trans-4-hydroxy-l-proline methyl ester as starting material which was derivatized through an SN2 reaction. Several leaving groups were studied, and the nosylate group showed superiority over other derivatives. Formation of an amide using the most costly starting material, 3,3-difluoropyrrolidine, was performed late in the synthesis to minimize its economical impact on the overall cost of the API.

(3,3-Difluoropyrrolidin-1-yl)-(2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)methanone.FREE BASE

Mp 149 °C (decomp).

[α]d = −31.1 (T = 24 °C, c = 1, CHCl3). Specific rotation of product 4 prepared using the initial route: [α]d = −31.5 (T = 24 °C, c = 1, CHCl3). 

1H NMR (400 MHz; CDCl3) δ 8.30 (d, J = 4 Hz, 2H), 6.48 (t, J = 4 Hz, 1H), 3.95–3.6 (m, 9H), 3.25–2.85 (m, 4H), 2.6–2.25 (m, 7H), 1.75–1.6 (m, 1H). 

13C NMR (100 MHz; CDCl3) δ 172.28; 161.55; 157.70; 127.22 (t, 1J C–F = 248 Hz), 126.22 (t, 1J C–F = 246 Hz), 109.95; 66.54; 58.87; 57.99; 52.71 (t, 2 J C–F = 32 Hz); 52.00; 50.41; 43.03; 34.46, 34.37, 34.25; 19F NMR (377 MHz, CDCl3) δ −102.1 (m, 2F).

IR (neat): 2951w, 2864w, 2799w, 2759w, 1630s, 1585vs, 1547m, 1449m, 1172m, 1254m, 1129m, 982w, 923m, 796m, 638w.

HRMS (ES, N2) Calcd for C17H24F2N6O: 367.20524, found: 367.20592.

……………………….

PAPER

(3,3-difluoro-pyrrolidin-1-yl)-((2S,4S)-(4-(4-pyrimidin-2-yl-piperazin-1-yl)-pyrrolidin-2-yl)-methanone: A potent, selective, orally active dipeptidyl peptidase IV inhibitor
Bioorg Med Chem Lett 2009, 19(7): 1991

 http://www.sciencedirect.com/science/article/pii/S0960894X09001966?np=y

  • Pfizer Global Research & Development, Groton/New London Laboratories, Pfizer Inc, Groton, CT 06340, United States

A series of 4-substituted proline amides was evaluated as inhibitors of dipeptidyl pepdidase IV for the treatment of type 2 diabetes. (3,3-Difluoro-pyrrolidin-1-yl)-[(2S,4S)-(4-(4-pyrimidin-2-yl-piperazin-1-yl)-pyrrolidin-2-yl]-methanone (5) emerged as a potent (IC50 = 13 nM) and selective compound, with high oral bioavailability in preclinical species.

Full-size image (4 K)

SEE………….https://docs.google.com/viewer?url=http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2FMiamiMultiMediaURL%2F1-s2.0-S0960894X09001966%2F1-s2.0-S0960894X09001966-mmc1.doc%2F271398%2Fhtml%2FS0960894X09001966%2Fce1f70bd989d6d4b79b40c26570693d2%2Fmmc1.doc

………………….

PATENT

WO 2005116014

http://www.google.co.in/patents/WO2005116014A1?cl=en

Example 113 (3.3-Difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)-methanone

 

Figure imgf000030_0001

Step 1 – (S)-2-(3.3-Difluoro-pyrrolidine-1-carbonyl)-4-oxo-pyrrolidine-1 -carboxylic acid tert-butyl ester

(S)-4-Oxo-pyrrolidine-1 ,2-dicarboxylic acid 1-tert-butyl ester (6.6 kg, 1.0 equivalent) was charged to a reactor, followed by addition of dichloromethane (15 volumes). The reaction mixture was cooled to 0°C. Triethylamine (4.82 liters, 1.2 equiv) was added over 30 minutes. The mixture turned from suspension to a clear solution at the end of triethylamine addition. The mixture was held at 0°C to 5°C for 10 minutes. Pivaloyl chloride (3.65 kg, 1.05 equivalents) was added slowly while keeping the reaction temperature at 0°C to 5°C. The reaction mixture turned back to aslurry. The reaction mixture was sampled for completion by HPLC (using diethylamine to derivatize) after held for 1 hour at 0°C to 5°C.

3,3-Difluoro- pyrrolidine hydrochloride (4.13 kg, 1.0 equivalent) was charged to the above mixture over 10 minutes at – 10°C to 0°C. Triethylamine (4.0 liters, 1.0 equiv) was introduced slowly over 70 minutes at -10°C to 0°C. Upon completion of triethylamine addition, the mixture was stirred for 1h at 0 to 5°C. The reaction was complete by HPLC assay (-1% starting material). The reaction was quenched with water (10 volumes) at 0°C to 5 °C. The mixture was heated to 20°C to 25 °C. The layers were separated, and the organic layer was washed with 0.5 M HCI (5 volumes). The organic layer was again washed with combined 5% NaHC03 (2 volumes) and half saturated brine solution (1.64 M, 3 volumes). The organic solution was concentrated atmospherically to a low stirrable volume (approximately 20 liters). Ethyl acetate (12.6 volumes, 82.8 liters) was added, the solution was concentrated atmospherically to -6 volumes. The mixture was held at 60°C to 65 °C for 2 hours and cooled to room temperature over 3 hours. The mixture was held at 20°C to 25 °C for 8 hours. Heptane (8 volumes) was added, and the mixture was granulated for a minimum of 2 hours. The solid was filtered, rinsed with 2:1 heptane/ethyl acetate (1 volume), and dried in a tray dryer at 25°C to 35°C for a minimum of 12 h. Yield: 7.26 kg, 79%. HPLC purity: 99.7%. The mother liquor (86 liters) was concentrated to 12 liters under partial vacuum at 65°C to 70°C. The mixture was cooled to 60°C to 65 °C. Ethyl acetate (4.0 liters) was added slowly over 15 minutes. The mixture was cooled to 20°C to 25 °C over 2 hours and was held at that temperature for at least 2 hours. The solid was filtered and rinsed with heptane/ethyl acetate (3:1 v/v, 1.7 liters). Drying in a tray dryer for 12 hours at 35°C to 45 °C yielded 435 grams of product. HPLC purity: 96.4%.

Step 2 – (2S.4S)-2-(3.3-Dif luoro-pyrrolidine-1 -carbonyl)-4-(4-pyrimidin-2-yl-piperazin-1 -yl)-pyrrolidine-1 – carboxylic acid tert-butyl ester A reactor was charged with THF (20 volumes), 2-piperazin-1-yl-pyrimidine (2.17 kg, 1.05 equivalents) and the product from Step 1 (4.00 kg, 1.0 equivalent). The mixture was held at 20°C to 25°C until all material was dissolved over 30 minutes. Acetic acid (0.792 kg, 1.05 equivalents) as added. The mixture was stirred for 1 hour during which the reaction mixture turned to cloudy. The reaction mixture was refluxed for 30 minutes and then concentrated at 60°C to 70°C until a steady temperature of 66.9°C was observed in the overheads indicating complete removal of water from the system. More THF was added as necessary. At the end, THF was added to bring the total volume in the reactor to 15 volumes of the limit reagent. The reaction mixture was cooled to -3°C to 7°C and sampled for complete formation of imine by HPLC (using sodium triacetoxyborohydride to reduce imine). Sodium triacetoxyborohydride (5.33 kg, 2.0 equivalents) was added portion-wise to the suspension at -5°C to 15°C. The reaction mixture was heated to 20°C to 25°C and held for 12 hours. HPLC results confirmed the reaction was complete by 99.8%. Sodium bicarbonate aqueous solution (10% w/w, 10 volumes) was added. The slurry was concentrated to remove 10 volumes of THF under partial vacuum at 30°C to 60°C. Ethyl acetate (10 volumes) was added to the suspension after it cooled to 20°C to 25CC. The organic phase was separated and the aqueous phase was checked by HPLC. It contained less than 2% of the product. The organic phase was washed with water (5 volumes), saturated brine solution (5 volumes) and concentrated to a small volume (2 volumes) under partial vacuum at 45°C to 50°C. To the slurry was added heptane (10 volumes) at 45°C to 50°C over 30 minutes. The mixture was cooled to 20°C to 25°C and granulated for 2 hours. Solid was collected by filtration, rinsed with heptane (2 volumes). Drying in a tray dryer for 12 hours at 35°C to 45°C yield 5.35 kg (91.3%) of the product. Step 3 – (3.3-Dif luoro-pyrrolidin-1 -yl)-f(2S.4S)-4-(4-pyrimidin-2-yl-piperazin-1 -yl)-pyrrolidin-2-yll- methanone Water (19 liters, 2 volumes) was charged to a reactor followed by the product from Step 2 (9.57 kg,

1.0 equivalent). To the slurry was added concentrated HCI (37 wt% in water, 19.1 liters, 2 volumes) slowly at 20°C to 30°C over 4 hours. The slurry went into solution after 12 liters of HCI was added. After the addition completion, the reaction was complete by HPLC assay. The reaction mixture was cooled to 5°C to 15°C. To the mixture was added 50% NaOH aqueous solution slowly with agitation to pH 10 to pH 11. The pH was monitored with a pH meter closely during the neutralization. The total volume of 50% NaOH added was 12.45 liters. The mixture was warmed to 20°C to 25°C and extracted with ethyl acetate twice (115 liters, 12 volumes and 57 liters, 6 volumes, respectively). The sample from aqueous layer after second extraction was analyzed by HPLC and showed only 1% of the product in that aqueous solution.

The organic layers were combined and treated with magnesium sulfate (5 kg) for 1 hour. The mixture was filtered. The filter cake was rinsed with ethyl acetate (10 liters). The filtrate was charged back to the reactor via a 0.2 micron in-line filter for speck free operation. (The following operations were performed under speck free conditions.) The solution was concentrated to 20 liters (2 volumes) under partial vacuum at 50°C to 60°C. The mixture was cooled to 20°C to 25°C over 30 minutes. Upon cooling to room temperature, crystallization occurred. The mixture was held for 30 minutes. Hexanes (20 liters, 2 volumes) was added slowly over 1 hour. The mixture was granulated for 2 hours. The solid product was collected by filtration and rinsed with hexanes/ethyl acetate (10 liters, 1 :1 v/v). The filter was blown dry with nitrogen for a minimum of 2 hours. The product was dried in a tray dryer at 44°C for 12 hours.

Yield: 5.7 kg, 75.9%.

m.p. 156°C. MS m/z 367 (MH+).

Figure imgf000030_0001FREE BASE

1H NMR (400 MHz, D20): δ 8.15 (d, 2H, J = 5.0 Hz, CH of pyrimidine), 6.55 (t, 1 H, J = 4.8 Hz, CH of pyrimidine), 3.87-3.81 (dd, 1 H, H2b of proline, rotomeric), 3.78-3.50 (m, 4H, N-CH2 of pyrrolidide), 3.55-3.40 (m, 4H, N-CH2 of piperazine), 2.97 (dd, 1 H, J = 10.2, 6.6 Hz, H5a of proline), 2.85-2.75 (m, 1 H, H4b of proline), 2.69 (dd, 1 H, J = 10.0, 9.1 Hz, H5b of proline), 2.55-2.20 (m, 7H, overlapping N-CH2 of piperazine, CH2 of pyrrolidide and H3b of proline), 1.47-1.38 (m, 1 H, H3a of proline).

Alternatively, the dihydrochloride salt of the titled compound was prepared according to the method of Example 1.

………………

US 2005/0256310

http://www.google.com/patents/US20050256310

Figure

 

This approach begins with Nt-Boc-4-oxo-l-proline (1) that undergoes a mixed anhydride activation with pivaloyl chloride at 0 °C, followed by amidation with 3,3-difluoropyrrolidine to yield the intermediate 2. Reductive amination with 1-(2-pyrimidyl)piperazine using sodium triacetoxyborohydride in THF/AcOH provided the desired stereoisomer 3 in high yield and selectivity, the undesired diastereomer being completely removed by crystallization. Deprotection of 3 with 6 N HCl, followed by neutralization with 50% NaOH and extraction provided PF-734200 (4) in good yield.

EXAMPLE 113 (3,3-Difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)-methanone

 

Figure US20050256310A1-20051117-C00011

 

Step 1—(S)-2-(3,3-Difluoro-pyrrolidine-1-carbonyl)-4-oxo-pyrrolidine-1-carboxylic acid tert-butyl

(S)-4-Oxo-pyrrolidine-1,2-dicarboxylic acid 1-tert-butyl ester (6.6 kg, 1.0 equivalent) was charged to a reactor, followed by addition of dichloromethane (15 volumes). The reaction mixture was cooled to 0° C. Triethylamine (4.82 liters, 1.2 equiv) was added over 30 minutes. The mixture turned from suspension to a clear solution at the end of triethylamine addition. The mixture was held at 0° C. to 5° C. for 10 minutes. Pivaloyl chloride (3.65 kg, 1.05 equivalents) was added slowly while keeping the reaction temperature at 0° C. to 5° C. The reaction mixture turned back to a slurry. The reaction mixture was sampled for completion by HPLC (using diethylamine to derivatize) after held for 1 hour at 0° C. to 5° C. 3,3-Difluoro-pyrrolidine hydrochloride (4.13 kg, 1.0 equivalent) was charged to the above mixture over 10 minutes at −10° C. to 0° C. Triethylamine (4.0 liters, 1.0 equiv) was introduced slowly over 70 minutes at −10° C. to 0° C. Upon completion of triethylamine addition, the mixture was stirred for 1 h at 0 to 5° C. The reaction was complete by HPLC assay (˜1% starting material). The reaction was quenched with water (10 volumes) at 0° C. to 5 ° C. The mixture was heated to 20° C. to 25 ° C. The layers were separated, organic layer was washed with 0.5 M HCl (5 volumes). The organic layer was again washed with combined 5% NaHCO(2 volumes) and half saturated brine solution (1.64 M, 3 volumes). The organic solution was concentrated atmospherically to a low stirrable volume (approximately 20 liters). Ethyl acetate (12.6 volumes, 82.8 liters) was added, the solution was concentrated atmospherically to ˜6 volumes. The mixture was held at 60° C. to 65° C. for 2 hours and cooled to room temperature over 3 hours. The mixture was held at 20° C. to 25 ° C. for 8 hours. Heptane (8 volumes) was added, and the mixture was granulated for a minimum of 2 hours. The solid was filtered, rinsed with 2:1 heptane/ethyl acetate (1 volume), and dried in a tray dryer at 25° C. to 35° C. for a minimum of 12 h. Yield: 7.26 kg, 79%. HPLC purity: 99.7%. The mother liquor (86 liters) was concentrated to 12 liters under partial vacuum at 65° C. to 70° C. The mixture was cooled to 60° C. to 65° C. Ethyl acetate (4.0 liters) was added slowly over 15 minutes. The mixture was cooled to 20° C. to 25° C. over 2 hours and was held at that temperature for at least 2 hours. The solid was filtered and rinsed with heptane/ethyl acetate (3:1 v/v, 1.7 liters). Drying in a tray dryer for 12 hours at 35° C. to 45° C. yielded 435 grams of product. HPLC purity: 96.4%.

Step 2—(2S,4S)-2-(3,3-Difluoro-pyrrolidine-1-carbonyl)-4-(4-pyrimidin-2-yl-piperazin-1-yl)-pyrrolidine-1-carboxylic acid tert-butyl ester

A reactor was charged with THF (20 volumes), 2-piperazin-1-yl-pyrimidine (2.17 kg, 1.05 equivalents) and the product from Step 1 (4.00 kg, 1.0 equivalent). The mixture was held at 20° C. to 25° C. until all material was dissolved over 30 minutes. Acetic acid (0.792 kg, 1.05 equivalents) as added. The mixture was stirred for 1 hour during which the reaction mixture turned to cloudy. The reaction mixture was refluxed for 30 minutes and then concentrated at 60° C. to 70° C. until a steady temperature of 66.9° C. was observed in the overheads indicating complete removal of water from the system. More THF was added as necessary. At the end, THF was added to bring the total volume in the reactor to 15 volumes of the limit reagent. The reaction mixture was cooled to −3° C. to 7° C. and sampled for complete formation of imine by HPLC (using sodium triacetoxyborohydride to reduce imine). Sodium triacetoxyborohydride (5.33 kg, 2.0 equivalents) was added portion-wise to the suspension at −5° C. to 15° C. The reaction mixture was heated to 20° C. to 25° C. and held for 12 hours. HPLC results confirmed the reaction was complete by 99.8%. Sodium bicarbonate aqueous solution (10% w/w, 10 volumes) was added. The slurry was concentrated to remove 10 volumes of THF under partial vacuum at 30° C. to 60° C. Ethyl acetate (10 volumes) was added to the suspension after it cooled to 20° C. to 25° C. The organic phase was separated and the aqueous phase was checked by HPLC. It contained less than 2% of the product. The organic phase was washed with water (5 volumes), saturated brine solution (5 volumes) and concentrated to a small volume (2 volumes) under partial vacuum at 45° C. to 50° C. To the slurry was added heptane (10 volumes) at 45° C. to 50° C. over 30 minutes. The mixture was cooled to 20° C. to 25° C. and granulated for 2 hours. Solid was collected by filtration, rinsed with heptane (2 volumes). Drying in a tray dryer for 12 hours at 35° C. to 45° C. yield 5.35 kg (91.3%) of the product.

Step 3—(3,3-Difluoro-pyrrolidin-1-yl)-[(2S,4S)-4-(4-pyrimidin-2-yl-piperazin-1-yl)-pyrrolidin-2-yl]-methanone

Water (19 liters, 2 volumes) was charged to a reactor followed by the product from Step 2 (9.57 kg, 1.0 equivalent). To the slurry was added concentrated HCl (37 wt % in water, 19.1 liters, 2 volumes) slowly at 20° C. to 30° C. over 4 hours. The slurry went into solution after 12 liters of HCl was added. After the addition completion, the reaction was complete by HPLC assay. The reaction mixture was cooled to 5° C. to 15° C. To the mixture was added 50% NaOH aqueous solution slowly with agitation to pH 10 to pH 11. The pH was monitored with a pH meter closely during the neutralization. The total volume of 50% NaOH added was 12.45 liters. The mixture was warmed to 20° C. to 25° C. and extracted with ethyl acetate twice (115 liters, 12 volumes and 57 liters, 6 volumes, respectively). The sample from aqueous layer after second extraction was analyzed by HPLC and showed only 1% of the product in that aqueous solution. The organic layers were combined and treated with magnesium sulfate (5 kg) for 1 hour. The mixture was filtered. The filter cake was rinsed with ethyl acetate (10 liters). The filtrate was charged back to the reactor via a 0.2 micron in-line filter for speck free operation. (The following operations were performed under speck free conditions.) The solution was concentrated to 20 liters (2 volumes) under partial vacuum at 50° C. to 60° C. The mixture was cooled to 20° C. to 25° C. over 30 minutes. Upon cooling to room temperature, crystallization occurred. The mixture was held for 30 minutes. Hexanes (20 liters, 2 volumes) was added slowly over 1 hour. The mixture was granulated for 2 hours. The solid product was collected by filtration and rinsed with hexanes/ethyl acetate (10 liters, 1:1 v/v). The filter was blown dry with nitrogen for a minimum of 2 hours. The product was dried in a tray dryer at 44° C. for 12 hours.

Yield: 5.7 kg, 75.9%. m.p. 156° C. MS m/z 367 (MH+).

1H NMR (400 MHz, D2O): δ 8.15 (d, 2H, J=5.0 Hz, CH of pyrimidine), 6.55 (t, 1H, J=4.8 Hz, CH of pyrimidine), 3.87-3.81 (dd, 1H, H2b of proline, rotomeric), 3.78-3.50 (m, 4H, N—CHof pyrrolidide), 3.55-3.40 (m, 4H, N—CHof piperazine), 2.97 (dd, 1H, J=10.2, 6.6 Hz, H5a of proline), 2.85-2.75 (m, 1H, H4b of proline), 2.69 (dd, 1H, J=10.0, 9.1 Hz, H5b of proline), 2.55-2.20 (m, 7H, overlapping N—CHof piperazine, CHof pyrrolidide and H3b of proline), 1.47-1.38 (m, 1H, H3a of proline).

Alternatively, the dihydrochloride salt of the titled compound was prepared according to the method of Example 1.

……………..

PAPER

Full-size image (21 K)

Scheme 1.

Reagents and conditions: (a) 3,3-difluoropyrrolidine hydrochloride, EDC, HOBt, TEA, DCM, rt; (b) NaBH4, MeOH, (c) (1) trifluoromethane-sulphonyl chloride, DIPEA, DCM; (2) 2-(1-piperazinyl)pyrimidine, DCM, −10 °C; (d) 4 N HCl in dioxane, rt; (e) 2-(1-piperazinyl)pyrimidine, NaBH(OAc)3, AcOH, DCE; (f) R1R2NH hydrochloride, EDC, HOBt TEA, DCM, 0–rt; (g) N-heterocyclic piperazine, NaBH(OAc)3, AcOH, DCE.

……………………….

 

Patent Submitted Granted
Therapeutic compounds [US7291618] 2005-11-17 2007-11-06
(2S,4S)-4-(piperazin-1-yl)pyrrolidine-2-methanone derivatives [US7465732] 2007-05-03 2008-12-16
THERAPEUTIC COMPOUNDS [US2007161664] 2007-07-12
Therapeutic compounds [US2006079498] 2006-04-13

 

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

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

Share
Jul 032015
 

Teneligliptin.svg

TENELIGLIPTIN

Teneligliptin; 760937-92-6; UNII-28ZHI4CF9C; Teneligliptin (INN); 28ZHI4CF9C
MF C22H30N6OS
MW 426.5782 g/mol

Teneligliptin (INN; trade name Tenelia) is a pharmaceutical drug for the treatment of type 2 diabetes mellitus. It is approved for use in Japan.[1] It belongs to the class of anti-diabetic drugs known as dipeptidyl peptidase-4 inhibitors or “gliptins”.[2] {(2S,4S)-4-[4-(3-Methyl-1-phenyl-1H-pyrazol-5-yl)-1-piperazinyl]-2-pyrrolidinyl}(1,3-thiazolidin-3-yl)methanone

Teneligliptin was launched in Japan in 2012 by Mitsubishi Pharma and Daiichi Sankyo for the treatment of type 2 diabetes mellitus. In 2013, the indication was partially changed to include it as a combination therapy with existing oral hypoglycemic agents, such as biganides, alpha-glucosidaseinhibitors, rapid-acting insulin secretagogues, and insulin preparations, as well as sulfonylureas and thiazolidines that had been approved for the combination.

In 2014, the product was registered in KR for the treatment of type 2 diabetes mellitus.
In 2013, Mitsubishi Tanabe Pharma filed for approval in Japan for use of the compound as combination therapy for the treatment of diabetes type 2.

CAS  760937-92-6

Teneligliptin.png

3-{(2S,4S)-4-[4-(3-methyl-l -phenyl- 1 H- pyrazol-5-yl)- l-piperazinyl]-2-pyrrolidinylcarbonyl}-l , 3-thiazolidine is represented structurally by a compound of formula (I):

 

Figure imgf000003_0001

Teneligliptin (CAS 760937-92-6) is a novel, potent and long-lasting dipeptidyl peptidase-4 inhibitor in treatment of type 2 diabetes. Dipeptidyl-peptidase-4 (DPP- 4) inhibitor has been demonstrated to improve glycemic control, in particular postparandial hyperglycemic control.

Despite of their common mechanism of action, DPP-4 inhibitors show marked structural heterogeneity. DPP-4 inhibitors may be classified into peptidomimetic (i.e. sitagliptin, vildagliptin, saxagliptin, and anagliptin) and non-peptidomimetic (i.e. alogliptin and linagliptin) subtypes.

Teneligliptin, is chemically known as a 3- {((2S,4S)-4-(4-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl)pyrrolidin-2-yl 25 carbonyl}thiazolidine hemipentahydrobromide hydrate and is peptidomimetic with the molecular formula of C22H30N6OS.2½HBr.xH2O and molecular weight of 642.88 g/mol for hemipentahydrobromide. The hydrate can be from mono to dihydrate.

U.S. Patent No. 7,074,794 B2 (the US ‘794) discloses teneligliptin as L-proline derivative and its pharmaceutically acceptable salts which exhibits a Dipeptidyl 5 peptidase IV (DPP-IV) inhibitory activity, which is useful for the treatment or prophylaxis of diabetes, obesity, HIV infection, cancer metastasis, dermopathy, prostatic hyperplasia, periodontitis, autoimmune diseases and the like.

The example-222 of the US ‘794 discloses the process for the preparation of teneligliptin as trihydrochloride salt U.S. Patent No. 8,003,790 B2 (the US ‘790) discloses salts of proline derivative, solvate thereof and production method thereof. In particular, the US ‘790 discloses 2.0 hydrochloride or 2.5 hydrochloride; 2.0 hydrobromide or 2.5 hydrobromide, and hydrates thereof teneligliptin.

The US ‘790 B2 further discloses different salts 15 of teneligliptin which are incorporated herein as reference in their entirety U.S. PG-Pub. No. 2011/0282058 A1 discloses salts of 3-{((2S,4S)-4-(4-(3-methyl- 1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl)pyrrolidin-2-ylcarbonyl}thiazolidine with mono-, di- and tri-basic acids or a solvate thereof. 20 International (PCT) publication No. WO 2012/165547 A1 discloses a process for preparation of teneligliptin and pharmaceutically acceptable salts thereof.

International (PCT) publication No. WO 2007/127635 A2 (the WO ‘635 A2) discloses a process for the preparation of diketo-piperazine and piperidine 25 derivatives. In particular, the WO ‘635 A2 discloses the process for preparation of 4-oxo-2-(thiazolidine-3-carbonyl)-pyrrolidine-1-carboxylic acid tert-butyl ester [herein compound (III)] by reacting piperazine with aryl halide.

International (PCT) publication No. WO 2012/099915 A1 (the WO ‘915 A1) 5 discloses the process for the preparation of deuterated thiazolidine derivatives. The WO ‘915 A1 also discloses the process for the preparation of 1-(3-methyl-1- phenyl-1H-pyrazol-5-yl)piperazine herein compound (V) by condensation of 5- chloro-3-methyl-1-phenyl-1H-pyrazole with piperazine.

Bioorganic & Medicinal Chemistry, 20(19), 5705-5719 (2012) discloses the process for the preparation of 1-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazine herein compound (V) by deprotection of Boc-protected 1-(3-methyl-1-phenyl-1Hpyrazol-5-yl)piperazine with triflouroacetic acid.

U.S. Patent Nos. 7,807,676 B2 and 7,807,671 B2 discloses a process for the preparation of 1-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazine by condensation of 5-chloro-3-methyl-1-phenyl-1H-pyrazole with piperazine in presence of n-BuLi in tetrahydrofuran. Bioorganic & Medicinal Chemistry, 14(11), 3662-3671 (2006),

Bioorganic & Medicinal Chemistry, 20(16), 5033-5041 (2012) and U.S. Patent Nos. 7,807,676 B2 and 7,807,671 B2 discloses a process for the preparation of (2S,4R)-tert-butyl 4-hydroxy-2-(thiazolidine-3-carbonyl)pyrrolidine-1-carboxylate by reacting (2S,4R)-1-(tert-butoxycarbonyl)-4-hydroxypyrrolidine-2-carboxylic acid with 25 thiazolidine in presence of HOBT and EDC.HCl in dimethylformamide solvent.

Bioorganic & Medicinal Chemistry, 15(2), 641-655 (2007) discloses a process for the preparation of (2S,4R)-tert-butyl 4-hydroxy-2-(thiazolidine-3- carbonyl)pyrrolidine-1-carboxylate by treating (2S,4S)-tert-butyl 4-[[(1,1-dimethylethyl)dimethylsilyl]oxy]-2-(3-thiazolidinylcarbonyl)pyrrolidine-1- carboxylate with tetrabutylammonium fluoride in tetrahydrofuran.

Bioorganic & Medicinal Chemistry, 20(19), 5705-5719 (2012) discloses the 5 process for the preparation of herein compound (II) after by reacting 1-(3-methyl- 1-phenyl-1H-pyrazol-5-yl)piperazine herein compound (V) with (2S,4R)-tert-butyl 4-hydroxy-2-(thiazolidine-3-carbonyl)pyrrolidine-1-carboxylate in presence of sodium triacetoxyborohydride. There is provided different alternative processes for the preparation of teneligliptin and intermediates thereof.

Bioorganic & Medicinal Chemistry, 20(19), 5705-5719 (2012) also discloses the process for the preparation of 4-[4-(5-methyl-2-phenyl-2H-pyrazol-3-yl)-piperazin- 1-yl]-2-(thiazolidine-3-carbonyl)pyrrolidine-1-carboxylic acid tert-butyl ester [herein compound (II)] after by reacting 1-(3-methyl-1-phenyl-1H-pyrazol-5- 15 yl)piperazine [herein compound (V)] with (2S,4S)-tert-butyl 4-[[(1,1- dimethylethyl)dimethylsilyl]oxy]-2-(3-thiazolidinylcarbonyl)pyrrolidine-1- carboxylate in presence of trifluoromethylsulfonic anhydride and diisopropylethylamine. 3 – [[(2S, 4S) -4- [4- (3- methyl-1-phenyl–1H- pyrazol-5-yl) -1-piperazinyl ] -2-pyrrolidinyl] carbamoyl] thiazolidine, having the formula below, is a very novel DPP-4 inhibitor potential.

Figure CN104177295AD00031

World Patent Application No. W02012099915 for Ge Lieting discloses a process for the preparation route is as follows:

Figure CN104177295AD00032

Journal B10rganic & Medicinal Chemistry, 2012, 20, 5705-5719 also discloses a preparation method for Ge Lieting, the route is as follows:

Figure CN104177295AD00041

[0009] 1- (3-methyl-1-phenyl-5-pyrazolyl) piperazine, was prepared for the Ge Lieting key intermediate. Journals B10rganic & Medicinal Chemistry, 2012,20,5705-5719 reported the preparation of the intermediates prepared route is as follows:

Figure CN104177295AD00042

[0011] The preparative route after the N-Boc-N- acetoacetyl piperazine phenylhydrazine and methanesulfonic acid in an ethanol solution of the reaction at room temperature 14h, concentrated under reduced pressure after addition of pyridine.Was added phosphorus oxychloride in pyridine, 20h post treatment reaction at room temperature the reaction system. The compound obtained above was then added trifluoroacetic acid was dissolved in methylene chloride after, after treatment at room temperature for 1.5h to give 1- (3-methyl-1-phenyl-5-pyrazolyl) piperazine.

The reaction process requires mesylate mesylate flammable, easy-absorbent deliquescence, and has a strong corrosive and irritating, easy to cause the body burns; phosphorus oxychloride, a highly toxic substance, water violent hair in the air smoke, hydrolyzed into phosphoric acid and hydrogen chloride, is very unstable, to operate a lot of trouble; trifluoroacetic acid is highly corrosive and irritant, can cause the body burns; low yield of the reaction (10%). Seeking a simple operation, high reaction yield, low cost and suitable for industrial production production process 1- (3-methyl-1-phenyl-5-pyrazolyl) piperazine has a very important role in the field of medicine.

…………………………………….

ten 1

ten 2 ten 3

ten 4

ten 1

ten 2

 

ten 4

 

since the capture is staggered, compd 165 is not clear in above pic see below

 

ten 3

 

 

…………

 

…………………….

CN104177295

reaction scheme in   http://www.google.com/patents/CN104177295A?cl=en

Figure CN104177295AD00043

Description: LR as Lawesson reagent (Lawesson Reagent), is a sulfur oxygen exchange reagent. The present invention provides a method for preparing key intermediates Ge Lieting method, comprising the steps of: (I) N-Boc-N- acetoacetyl piperazine Lawesson’s reagent in the presence of an organic solvent, with a phenylhydrazine of the formula occurs ⑴ reaction shown:

Figure CN104177295AD00051

(2) the step (1) The product was dissolved in an organic solvent, the following formula (II) in concentrated hydrochloric acid to deprotected shown:

Figure CN104177295AD00052
格列汀 refers to 1- (3-methyl-1-phenyl-5-pyrazolyl) piperazine
……………………………..

Volume 20, Issue 19, 1 October 2012, Pages 5705–5719

Full-size image (24 K)
…………………………………..

 

………………………..

http://www.google.co.in/patents/WO2015019238A1?cl=en

Example 5: Preparation of {(2^,.4^)-4-r4-(3-methyl-l-phenyl-lH-pyrazol-5-yl)piperazin- 1 -vHpyrrolidin-2-yl } ( 1.3 -thiazolidin-3 -vDmethanone hemipentahydrobromide hydrate (Formula II)

Activated carbon (10 g) was added to a solution of the residue (obtained in Example 4) in isopropyl alcohol (1000 mL) at 30°C to 35°C. The reaction mixture was filtered through a Hyflo® bed. The filtrate was heated to a temperature of 70°C to 75°C. Hydrobromic acid (48%; 168 g) was slowly added to the filtrate at 70°C to 75°C over a period of 10 minutes to 15 minutes. The reaction mixture was stirred for 2.5 hours at 70°C to 77°C. The progress of the reaction was monitored by HPLC. After completion of the reaction, the reaction mixture was cooled to a temperature of 20°C to 25 °C, and stirred at the same temperature for 60 minutes. The reaction mixture was filtered to obtain a solid. The solid obtained was washed with isopropyl alcohol (2 x 200 mL), and dried at 50°C under reduced pressure for 15 hours to obtain crude {(25*,45)-4-[4-(3-methyl-l-phenyl-lH- pyrazol-5 -yl)piperazin- 1 -yl]pyrrolidin-2-yl} ( 1 ,3 -thiazolidin-3 -yl)methanone

hemipentahydrobromide hydrate.

Yield: 90%

Example 6: Purification of {(2^’.4^)-4-r4-(3-methyl-l-phenyl-lH-pyrazol-5-yl)piperazin- 1 -yllpyrrolidin-2-yl } ( 1.3 -thiazolidin-3 -vDmethanone hemipentahydrobromide hydrate (Formula II)

A reaction mixture containing {(2S,4S)-4-[4-(3-methyl-l-phenyl-lH-pyrazol-5- yl)piperazin- 1 -yl]pyrrolidin-2-yl } ( 1 ,3 -thiazolidin-3 -yl)methanone

hemipentahydrobromide hydrate (100 g; prepared according to the process of Example 5) in ethanol (700 mL) was heated at 70°C to 75°C to obtain a solution. The solution was filtered at the same temperature. The filtrate was allowed to cool to a temperature of 65 °C to 68°C, and deionized water (10 mL) was added at the same temperature. The solution was cooled to a temperature of 55°C to 60°C, and stirred at the same temperature for 2 hours. The solution was further cooled to a temperature of 20°C to 25 °C, and stirred at the same temperature for 60 minutes to obtain a solid. The solid was filtered, washed with ethanol (100 mL), and dried at 45°C to 50°C under reduced pressure for 18 hours to 20 hours to obtain pure {(2S,4S)-4-[4-(3-methyl-l-phenyl-lH-pyrazol-5-yl)piperazin-l- yl]pyrrolidin-2-yl } ( 1 ,3 -thiazolidin-3 -yl)methanone hemipentahydrobromide hydrate .

Yield: 90%

HPLC Purity: 99.93%

WO2012099915A1 * 18 Jan 2012 26 Jul 2012 Hongwen Zhu Thiazolidine derivatives and their therapeutic use
WO2012165547A1 * 31 May 2012 6 Dec 2012 Mitsubishi Tanabe Pharma Corporation Method for manufacturing pyrazole derivative
WO2014041560A2 * 28 Aug 2013 20 Mar 2014 Glenmark Pharmaceuticals Limited; Glenmark Generics Limited Process for the preparation of teneligliptin
US7074794 10 Aug 2001 11 Jul 2006 Mitsubishi Pharma Corporation Proline derivatives and the use thereof as drugs
US8003790 17 Feb 2006 23 Aug 2011 Mitsubishi Tanabe Pharma Corporation Salt of proline derivative, solvate thereof, and production method thereof
US20050256310 * 12 May 2005 17 Nov 2005 Pfizer Inc Therapeutic compounds
EP1854795A1 * 17 Feb 2006 14 Nov 2007 Mitsubishi Pharma Corporation Salt of proline derivative, solvate thereof, and production method thereof
EP1894567A1 * 2 Jun 2006 5 Mar 2008 Mitsubishi Tanabe Pharma Corporation Concomitant pharmaceutical agents and use thereof
US20040106655 * 10 Aug 2001 3 Jun 2004 Hiroshi Kitajima Proline derivatives and the use thereof as drugs
 Patent Filing date Publication date Applicant Title
WO2015019238A1 * 28 Jul 2014 12 Feb 2015 Ranbaxy Laboratories Limited Process for the preparation of n-protected (5s)-5-(1,3-thiazolidin-3-ylcarbonyl)pyrrolidin-3-one
Patent Submitted Granted
Proline derivatives and use thereof as drugs [US7060722] 2005-11-03 2006-06-13
Proline derivatives and the use thereof as drugs [US7074794] 2004-06-03 2006-07-11
Proline derivatives and use thereof as drugs [US2006173056] 2006-08-03
SALT OF PROLINE DERIVATIVE, SOLVATE THEREOF, AND PRODUCTION METHOD THEREOF [US8003790] 2009-08-27 2011-08-23
METHOD OF TREATING ABNORMAL LIPID METABOLISM [US2010305139] 2010-12-02
COMBINED USE OF DIPEPTIDYL PEPTIDASE 4 INHIBITOR AND SWEETENER [US2010113382] 2010-05-06
CONCOMITANT PHARMACEUTICAL AGENTS AND USE THEREOF [US2009082256] 2009-03-26
PROPHYLACTIC/THERAPEUTIC AGENT FOR ABNORMALITIES OF SUGAR/LIPID METABOLISM [US2009088442] 2009-04-02
SALT OF PROLINE DERIVATIVE, SOLVATE THEREOF, AND PRODUCTION METHOD THEREOF [US2011282058] 2011-11-17
  1.  Joanne Bronson, Amelia Black, T. G. Murali Dhar, Bruce A. Ellsworth, and J. Robert Merritt. “Teneligliptin (Antidiabetic)”. Annual Reports in Medicinal Chemistry 48: 523–524. doi:10.1016/b978-0-12-417150-3.00028-4
  2.  Kishimoto, M (2013). “Teneligliptin: A DPP-4 inhibitor for the treatment of type 2 diabetes”Diabetes, metabolic syndrome and obesity : targets and therapy 6: 187–95. doi:10.2147/DMSO.S35682PMC 3650886PMID 23671395.

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

 

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

http://organicsynthesisinternational.blogspot.in/p/gliptin-series-22.html

 

Share

trans-Cinnamamide , (2E)-3-Phenyl-2-propenamide

 Uncategorized  Comments Off on trans-Cinnamamide , (2E)-3-Phenyl-2-propenamide
Jun 272015
 

trans-Cinnamamide/(2E)-3-Phenyl-2-propenamide

195
Name trans-Cinnamamide
Synonyms (2E)-3-Phenyl-2-propenamide
Name in Chemical Abstracts 2-Propenamide, 3-phenyl-, (2E)-
CAS No 22031-64-7
EINECS No
Molecular formula C9H9NO
Molecular mass 147.18
SMILES code NC(=O)/C=C/c1ccccc1

 

1H NMR

 

1H NMR

1H-NMR: trans-Cinnamamide
250 MHz, DMSO-d6
delta [ppm] mult. atoms assignment
6.61 d (JAB= 15.9) 1 H C=CH-CO (2-H)
7.13 broad s 1 H NH
7.2-7.6 m 7 H CH (arom.) + NH + -CH=C (3-H)
7.42 d (JAB= 15.9) 1 H -CH=C (3-H)
6.53 d 1 H C=CH-CO (2-H, cinnamic acid)
7.82 d 1 H -CH=C (3-H, cinnamic acid)
2.5 s DMSO
3.33 s O-CH3 (tBu-OMe)
1.19 s C-CH3 (tBu-OMe)

 

13C-NMR

13C NMR

13C-NMR: trans-Cinnamamide
250 MHz, DMSO-d6
delta [ppm] assignment
122.31 C2 (=CH-)
127.52 CH arom.
128.90 CH arom.
129.42 C4 (arom.)
134.86 C quart. arom.
139.16 C3 (-CH=C)
166.68 C1 (-C(=O)NH2)
38.5-40.5 DMSO-d6

 

IR

IR

IR: trans-Cinnamamide
[KBr, T%, cm-1]
[cm-1] assignment
3375, 3175 N-H valence
3084 aliph. C-H valence, =C-H
1665 C=O valence, carboxamide
1610 alkene C=C valence
1580, 1495 arom. C=C valence

 

Chromatogram

crude product chromatogram

HPLC: crude product
column Phenomenex Luna C18; particle diameter 3 µm, L= 150 mm, ID= 4.6 mm
column temperature 25 °C
injection 5 µL
mobile phase 5% MeCN / H2O (0.0059% CF3COOH), gradient to 95% MeCN / H2O (40 min), 10 min isocratic
flow 1.0 mL/min
detector (UV 220 nm) percent concentration calculated from relative peak area


pure product chromatogram

HPLC: pure product
column Phenomenex Luna C18, particle diameter 3 µm, Länge 150 mm, Innendurchmesser 4.6 mm
column temperature 25 °C
injection 5 µL
mobile phase 5% MeCN/H2O (0.0059% CF3COOH), gradient to 95% MeCN/H2O (40 min), 10 min isocratic
flow 1.0 mL/min
detector (UV 220 nm) percent concentration calculated from relative peak area

 

 

 

 

Name trans-Cinnamamide
Synonyms (2E)-3-Phenyl-2-propenamide
Name in Chemical Abstracts 2-Propenamide, 3-phenyl-, (2E)-
CAS No 22031-64-7
EINECS No
Molecular formula C9H9NO
Molecular mass 147.18
SMILES code NC(=O)/C=C/c1ccccc1
trans-Cinnamoyl chloride
NH3
reacts to
trans-Cinnamamide + Hydrogen chloride

 

 

IR

IR

IR: trans-Cinnamamide
[KBr, T%, cm-1]
[cm-1] assignment
3375, 3175 N-H valence
3084 aliph. C-H valence, =C-H
1665 C=O valence, carboxamide
1610 alkene C=C valence
1580, 1495 arom. C=C valence

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

 

Denpasar, bali, indonesia

  1. Denpasar – Wikipedia, the free encyclopedia

    https://en.wikipedia.org/wiki/Denpasar

    Denpasar (Indonesian: Kota Denpasar, Indonesian pronunciation: [dənˈpasar]) is the capital and the most populous city of the Indonesian province of Bali.

    Etymology – ‎History – ‎Geography – ‎Demography
    .
    Denpasar market, Denpasar is the capital of Bali Province. The main street of Denpasar is Gajah Mada street where is the main shopping center, .
    A very giant Ogoh-ogoh at a cross junction in Denpasar
    A view from the Kumbasari Market

    Airport of BaliPT (PERSERO) ANGKASA PURA I CABANG BANDARA NGURAH RAIGEDUNG WISTI SABHA LANTAI 3 BANDARA NGURAH RAIDENPASAR, BALI 80362

    Bali Airport (Ngurah Rai) Denpasar – Indonesia

    Bali Ngurah Rai International Airport, also known as Denpasar International Airport, is located in southern Bali, 13 km south of Denpasar. It is Indonesia’s third-busiest international airport.

    Bali Airport - Denpasar

    Bali Airport Check-in Counters

    Bali Airport Terminal Interior

    Bali Airport

    Bajak Laut Nasi Tempong & Seafood, Renon, Bali

    Sporting the growth of Denpasar residents’ likes for Nasi Tempong, Bajak Laut Nasi Tempong & Seafood sets on a different kind of path by combining Nasi Tempong with the other well-known Bali’s best: Seafood.

    Sets in the cozy neighborhood of Renon, Denpasar, Bajak Laut is the newest addition of restaurant openings in this area. From the down-to-earth food courts selling Ayam Goreng, Chinese food and Sup Kepala Ikan, to the more luxurious XO Suki & Cuisine, Ayucious, or the more established Bendega, Ikan Bakar Cianjur, and Hanamasa, Bajak Laut further marks Renon as a leading Denpasar’s culinary destination.

    Though opened really close to the market leader Nasi Tempong Indra, that with its aggressive market expansion in 2012 opens two new branches around Renon area alone, Bajak Laut however has what Indra has not: various choices of seafood comprising of fishes, shellfish, crabs, and shrimps. Therefore market wise, Bajak Laut is aiming at a slightly different crowds: those who loves the spicy Nasi Tempong, and those who loves Seafood; especially those too tired to go through all the traffic madness at Simpang Siur to reach Jimbaran.

    (Or believes it’s too touristy.)

    As the champion of this premise, Bajak Laut offers “Kepiting Asap ala Bajak Laut”, which are crabs cooked in sweet and savory rubs, then grilled inside banana leaf wraps to enhance its aroma. The result is a treat not only delicious to the taste but also to the sight.

    Ingredients used for the rub is dominated with daun salam, or Indonesian bay leaves. For those familiar with gepuk; fried beef first marinated in spices and brown sugar, Kepiting Asap ala Bajak Laut has an almost identical seasoning.

    One portion of Kepiting Asap ala Bajak Laut consisting of two crabs weighing total of 5 ons (500 grams). At 120K they’re good for two, while the 7 ons one costs 150K. For the 5 ons portion, the crab size is a bit small, hence eating them requires quite an effort.

    Nasi Tempong is a good example how a food originated from outside Bali could becomes a local hit. Originated from Banyuwangi, Nasi Tempong managed to get quite followers due to its main character of super spicy sambal. Nasi Tempong usually served as a package consisting of white rice, steamed vegetables, tahu, tempe, anemic salted fish, and super spicy sambal, and a main dish of either fried chicken, or other kind of proteins.

    While I’m not a die-hard spicy food fans, I found their Nasi Tempong quite palatable, especially since the stewed vegetables plus sambal that’s the core of every Nasi Tempong dish, is like a staple food in Western Java where I grew up.

    Service is polite and attentive, though we found it’s a bit hard to get attention from the waiters, except from the one stand by the front door. Beside of the seafood, the Nasi Tempong variations are sold around 12K to 45K,

    Starting December 2013 but don’t know for how long, Bajak Laut Nasi Tempong & Seafood offers a special discount for their set menu, at 150K for 4 people, and 250K for 6 people.

    One annoying condition that we have to face as well, that even though the premise is fully airconed, people are allowed to smoke! And here in Denpasar, Bali, sadly it’s the common case with many eating premises, and Bajak Laut is no exception. Therefore while their crab is quite delicious, until Bajak Laut separates its smoking and non-smoking section it poses health hazard to your youngsters. (byms)

    Bajak Laut Nasi Tempong & Seafood
    Jl. Cok Agung Tresna No.23, Renon, Denpasar, Bali
    (0361) 7984007

    //////////

 

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

 

/////////

Share

11-Chloro-1-undecene

 Uncategorized  Comments Off on 11-Chloro-1-undecene
Jun 272015
 

11-Chloro-1-undecene

125
Name 11-Chloro-1-undecene
Synonyms
Name in Chemical Abstracts 1-Undecene, 11-chloro-
CAS No 872-17-3
EINECS No
Molecular formula C11H21Cl
Molecular mass 188.74
SMILES code ClCCCCCCCCCC=C

 

 

 

10-Undecen-1-ol
SOCl2
reacts to
11-Chloro-1-undecene + Hydrochloric acid + Sulfur dioxide

1H-NMR

1H NMR

1H-NMR: crude product
300 MHz, CDCl3
delta [ppm] mult. atoms assignment
1.1-1.5 m 12 H CH2
1.75 tt 2 H 2-H
2.02 dt 2 H 9-H
3.51 t 2 H 1-H
4.95 2xdd 2 H 11-H
5.80 m 1 H 10-H


1H NMR

1H-NMR: 11-Chloro-1-undecene
300 MHz, CDCl3
delta [ppm] mult. atoms assignment
1.1-1.5 m 12 H CH2
1.75 tt 2 H 2-H
2.02 dt 2 H 9-H
3.51 t 2 H 1-H
4.95 2xdd 2 H 11-H
5.80 m 1 H 10-H

 

13C-NMR

13C NMR

13C-NMR: crude product
75.5 MHz, CDCl3
delta [ppm] assignment
32.7 C2
33.9 C9
45.0 C1
114.1 C11
139.1 C10
76.5-77.5 CDCl3


13C NMR

13C-NMR: 11-Chloro-1-undecene
75.5 MHz, CDCl3
delta [ppm] assignment
32.7 C2
33.9 C9
45.0 C1
114.1 C11
139.1 C10
76.5-77.5 CDCl3

 

IR

IR

IR: 11-Chloro-1-undecene
[Film, T%, cm-1]
[cm-1] assignment
3077 aliph. C-H valence, H2C=C
2927, 2855 aliph. C-H valence
993, 910 deform. C-H, H2C=C
723 C-Cl valence

 

Operating scheme

Operating scheme

 

 

 

 

Chromatogram

crude product chromatogram

GC: crude product
column DB-WAX, L=30 m, d=0.33 mm, film=0.25 µm
inlet on column injection, 0.2 µL
carrier gas H2, 40 cm/s
oven 90°C (5 min), 10°C/min –> 240°C (30 min)
detector FID, 270°C
integration percent concentration calculated from relative peak area


pure product chromatogram

GC: pure product
column DB-WAX, L=30 m, d=0.33 mm, film=0.25 µm
inlet on column injection, 0.2 µL
carrier gas H2, 40 cm/s
oven 90°C (5 min), 10°C/min –> 240°C (30 min)
detector FID, 270°C
integration percent concentration calculated from relative peak area

8 must-see places in Southeast Asia for great views: bucket list 2015

8 must-see places in Southeast Asia for great views: bucket list 2015
Southeast Asia is more than food and culture; here are 8 eye-candy places with magnificent views for a highly memorable trip!

Not just a melting pot of cultures, religions, history and food, Southeast Asia offers many picturesque spots that your eyes will thank you for. Whether it’s enjoying a sunset from a mountain top or just taking in the bucolic sights of Mother Nature’s hand-sculpted terrains, you’ll attest that these 8 suggestions offer some pretty unique charms that take your breath away.

 

1. Inle Lake, Myanmar

Myanmar has become a hotspot for the intrepid traveller and opens up plenty of opportunities to lap up many of its natural scenic wonders. If you’re heading there, a must-see place is Inle Lake renowned for its vast body of water where one can spot fishing communities and homes built on stilts.

Not only is the lake famous for its photogenic quality, you can hire guides to visit fish farms and shop at handicraft stores. Inle Lake’s picturesque charm comes from watching leg-rowing fisherman haul their catch during sunset. Find cheap flights to the capital Naypyidaw and best time to travel there is between November and February.

Read more: Top 10 things to do in Myanmar

Be mesmerised by the scenic Inle Lake in Myanmar

 

 

2. Tiger’s Nest Monastery, Bhutan

Perched some 3,000 metres above sea level and build in 1692, Bhutan’s Taktsang monastery, or more popularly known as Tiger’s Nest, is a must-see when you come to this nation steeped in Buddhist history. Getting there is not for the faint-hearted as one has to traipse through a hilly, rocky and undulating path to reach the peak.

Do hire guides to reach the apex successfully, and you’ll be rewarded by 360-views of sylvan mountain tops. Spring time from March to May is the best time to visit Bhutan and Drukair Royal Airlines of Bhutan flies theredirect.

Read more: 5 tips on tipping when travelling in Southeast Asia

Take in the lofty, airy views of Tiger’s Nest in Bhutan

 

 

3. Mount Kinabalu, Sabah, Malaysia

Recognised as one of the tallest peaks in Southeast Asia, Mount Kinabalu is a trekker’s dream come true. Getting to the summit takes about two days to accomplish. There is a 4-km climb to Laban Rata lodge where you can rest and replenish on sustenance. The next day is a 2-km climb to Low’s Peak.

The trek may be arduous but with lush rainforest terrain, there’s always something new at every corner to keep you distracted. About a kilometer away from the peak, the terrain changes to rock, stone and pebbles complemented by vegetation normally found in cooler climes. To catch the sunrise on the second day, it’s advisable to depart at 2am but remember to bring extra clothing as the mercury will drop to 2 degrees Celsius.

Read more: 5 fun extreme sports in Singapore for the adventure seekers

The scenic misty peaks of Mt. Kinabalu also offer spots for picturesque photos

 

 

4. Palawan Island, Luzon, Philippines

Recently coined by Huffington Post as “The Most Beautiful Island In the World” while Conde Nast Traveler’sReader Choice Awards named it “The Top Island in the World”, Palawan island is quite the magnificent sight. With its beautiful azure waters infused with emerald hues, it’s also hard to refute such claims.

Dotting the waters are jungled-filled islands, each with a distinctive hill rising above the ocean. Just by half-hour domestic flight from Manila airport, once you soar above Palawan’s oceanic landscape, you’ll feel like you’ve reached Shangri-la. Whether it’s island-hopping or sea kayaking, fun-filled times are never in short supply.

Read more: 12 best beaches in Asia Pacific

The beaches of Palawan have powdery white sand!

 

 

5. Penang National Park, Malaysia

Penang is truly a foodie paradise but many people are flocking there for other reasons, one being its attractive natural environment. Located just west of mainland Malaysia, a flight from Singapore is slightly over an hour.

With plenty of diverse lifestyle choices and entertainment options, Penang also has its idyllic charms. Aside from its UNESCO-designated George Town, the Penang National Park located on the North-Western side of the island rewards one with rich rainforests, a diverse ecosystem and some 1,381 hectares of wetlands to indulge trekking fanatics and eco-photographers.

Read more: Best cruises from Singapore

Unique flora and fauna found at Penang National Park makes for picture perfect memories too

 

 

6. Tanah Lot Temple, Bali, Indonesia

Bali is never in short supply of mysticism and wonder. A two-hour flight out of Singapore is all it takes to enjoy a short vacation. And of course, visiting its picturesque sea temple on the west coast of Bali, Tanah Lot, promises many Kodak moments. A simple traipse during low-tide rewards a sight to behold too – a Hindu shrine ensconced among lush trees perched on a rock is postcard-worthy from any angle. Framed by crashing waves, Tanah Lot Temple brims with a dab of fable and mysticism that makes it a must-see when visiting Bali!

Read more: Top 5 places to go diving in Southeast Asia

Tanah Lot in Bali offers scenic views of splendid structures amidst crashing waves

 

 

7. Angkor Wat, Siem ReapCambodia

Angkor Wat became even more famous, thanks to the Tomb Raider movie starring Angelina Jolie. Founded in the 12th Century, it is also the 7th Wonder of the World. This Khmer temple’s architecture will seize the gaze of any first-time visitor. At the centre of this city, within a moat, is a towering stupa that provides sylvan views of its 3.6km, vine-covered outer wall. Just 5.5km north of Siem Reap, the Angkor Archaeological Park is a must-see for travellers with a penchant for history and artefacts.

Read more: Top 10 most romantic places in Asia (part 2)

Angkor Wat’s lush views are both captivating and mysterious

 

 

8. Halong Bay, Hanoi, Vietnam

Halong Bay, which means “Bay of Descending Dragons”, is a unique karst topography carved out by Mother Nature. The UNESCO World Heritage site offers views of vertical formations which are rich in dense vegetation. A boat cruise meandering through any of the 1,969 islets is both tranquil and insightful. Avoid the monsoons from June to September and from January to March, but visit the high seasons to enjoy sunny skies that won’t put a damper on your exploration plans of the natural outlying islets. After a three-hour fight to Hanoi from Singapore, take a five-hour road trip via mini bus to the port; it costs around USD 6 (SGD 7.50) and can be arranged upon arrival.

Read more: Top 10 most romantic places in Asia (part 1)

Halong Bay in Vietnam promises oceanic vistas

 

 

All these places will astound you in a multi-sensory way. Whichever activity you decide to experience at these destinations, you’ll agree that many good memories await.

 

 

  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

///////////

Share

Sun Kim ……Quality-by-Design Evangelist

 Uncategorized  Comments Off on Sun Kim ……Quality-by-Design Evangelist
Jun 252015
 

Sun Kim

Sun Kim

QbDWorks.com – Quality by Design for Pharma, Biotech, Medical Devices

Dr. Sun K. Kim is a Quality-by-Design Evangelist, transforming how Product Development is executed in the Biologics, Pharmaceutical and Medical Devices industry. In addition, he teaches at Keio University and Stanford University. His current focus of research is Quality-by-Design, Agile Development of Drugs and Therapeutics.

He received his MS and PHD in Mechanical Engineering at Stanford University. Sun was recently a Professor at Keio University in Japan. Prior to Silicon Valley days, he served in the Korean Army and worked at BMW in Munich, Germany

  • Sun Kim – Google+

    https://plus.google.com/104532120968165429422

    Sun Kim. Worked at QbDWorks. Attended Stanford University. Lives in San Francisco, CA. 2,886 views … QbD Risk Assessment – Quality by Design. 1.

    • Sun Kim – YouTube

      https://www.youtube.com/channel/UCoWqD615csn0MUEXDW2O2Qg

      QbDWorks share what works and does not when implementing Quality-by-Design in the Biotech, Biologics, Pharmaceutical and Medical Devices Industry.

      • Sun Kim | Facebook

        https://www.facebook.com/kimsunkist?fref=nf

        • Sun Kim is on Facebook. Join Facebook to connect with Sun Kim and others you may know. Facebook gives people the power to share and makes the world …

 

Sun Kim

Experience

Quality by Design – Founder

QbDWorks

 – Present (2 years 6 months)http://QbdWorks.com

Founder of QbDWorks.com

Quality by Design for Biotech, Pharmaceutical and Medical Devices – Quality by Design Tools and Case Studies

Lecturer

Stanford University

 – Present (10 years)Stanford, CA

Teach Design for Manufacturing, Robust Design, Design of Experiments

Master Black Belt in Quality-by-Design, Lean Six Sigma, Sr. Manager

Bayer HealthCare

 –  (2 years 7 months)Berkeley, CA

Sr. Manager, Master Black Belt in Quality-by-Design, Design for Lean Six Sigma,
Leading Business Process Management

Design for Excellence Evangelist

Abbott

 –  (1 year 9 months)

Master Black Belt (Lean Six Sigma), Project Management Professional, Scrum Master

Assistant Professor of Graduate School of Systems Design and Management Assistant

Keio University

 –  (3 years 1 month)

http://www.sdm.keio.ac.jp/en/faculty/kim_s.html

Lecture and advise graduate-level, professional students on system and product design, design thinking, creative brainstorming methodologies, prototyping, project management and business development. Solicited 15 industry project partners. Generated $35,000/year after developing non-degree curriculum for professionals. Co-Investigator of research projects of $50,000: Indoor Location-based Services Technology for Mobile Devices. Consults manufacturing companies (Hitachi, Toshiba) on growth strategies for Service Business Innovation. Others include developing cost simulation tool of product design based on injection molding, design for manufacturing and healthcare delivery systems. Began as a lecturer in Feb. 2008 to co-develop a project-based design curriculum, Active Learning Program Sequence (ALPS), educating over 100 graduate students every year.

Invited as an Assistant Professor in June, 2009.

Research, Teaching Assistant

Stanford University

 –  (3 years 10 months)

Lectured, coached and managed over 40 multi-disciplinary teams on Design for Manufacturing projects from Biomedical device (Medtronic, Maquet, St. Jude Medical, etc.) and automotive companies (Toyota, Nissan, GM, etc.). Served as the main research associate of Toshiba Corporation Six-Sigma Consulting Inc., developing systems design and manufacturing programs for Toshiba employees. Innovative projects were mobile personal-assistant IT system and agile transportation infrastructure.

Industry-sponsored Projects

Stanford University

 –  (4 years 10 months)

Maquet Cardiovascular: Coached and led a 3 member team in redesigning the crimping process of Hemashield Grafts, resulting in cost reduction of $75,000 and operators’ medical costs from injuries.

Satiety (Bariatric Surgery Device for Obesity Treatment) Design for Manufacturing Project: Coached a 3 member team in redesigning the packaging and supply chain for the Toga System, resulting in supply chain efficiency of 50% improvement by applying Lean and Errorproofing (Poka Yoke) Techniques.

Medtronic Vascular: Coached a 4 member team in redesigning the manufacturing line of a stent-graft, resulting in 73% reduction of lead time and increase in reliabilty and performance. Observed over 5 vascular and general surgery cases. http://www.youtube.com/watch?v=RkA2TyCsV0A

St. Jude Medical: Led a 4 member team in developing a 7-year supply chain strategy for new service centers of ICD/pacemaker programmers in Europe, Asia, N. and S. America and Oceania. The recommendation consists of an optimized cost model from net present value analysis and AHP location decision modeling that will save $461 million over 7 years and increase customer service rate compared to the existing service centers.

Nissan Motors: Led a 4 member team to construct a 20 year technology / business roadmap of Nissan Fuel Cell powertrain / vehicles which projects $ 4.8 billion revenue. Created a fuel cell vehicle concept design by applying Design for manufacturing tools including market research, manufacturability, and profitability analysis.

Zimmer Orthopedics: Implemented, with five team members, a FEA (finite element analysis) simulation tool with ABAQUS which assists in the development of treating knee osteoarthritis, based on MRI and gait data. http://www.youtube.com/watch?v=47QOdiauHwE

General Motors: Achieved potential cost reduction of $450 per vehicle and reduced 50kg of car weight by replacing wires with conductive coatings and RFID applications with a team of four members.

Design for Six Sigma Research Fellow

Toshiba

 –  (3 years 7 months)

Developed Design for Six Sigma Curriculum for Toshiba Corp.
Trained engineers, managers in systems design methodologies.
Coached Six Sigma projects.

Medical Device Design Innovation Program Developer

Johnson & Johnson

 –  (3 months)

Developed an innovation-incubation program to design/develop next generation product/technology with physicians and multidisciplinary design teams.

Design, Manufacturing Consultant

NeoGuide Systems

 –  (6 months)

Performed Robust Design, Design of Experiment, Developed manufacturing tooling, testing protocols and automated stations.

Reliability Research Assistant

Stanford Linear Accelerator Center

 –  (7 months)

Developed a reliability decision analysis tool for the LINAC System which will save $83 million per year on the $8 billion International Linear Collider Project. Enhanced reliability of the Accelerator system up to 20% and had increased throughput by linking Failure Mode and Effect Analysis of the Tuner to the evaluation tool.

Optimization/ Structural Analysis Intern

Samsung Electronics

 –  (1 month)

Improved impact-worthiness (20%) by optimizing design parameters of cell phone cases after performing structural analysis.

Design, Manufacturing Engineer

BMW

 –  (8 months)

Applied for 1 patent individually and created 3 design proposals as a team in 3 months. Saved $2,760 per month by implementing knowledge database management system. Resolved 3 process problems during 2 weeks of manufacturing
rotation program in Munich assembly plant with the manufacturing engineers.

US – ROK Army Radiology Tech, Company Leader, Manager

Republic of Korea Army

 –  (2 years 3 months)

Served 20,000 US Army patients as a Radiology Technician, tasks including diagnostic x-ray imaging, upper GI, etc. Managed 12 multi-national soldiers and a radiology department. Was awarded as “the accident-free company.” Increased the availability of the radiology department, which takes care of 20,000 patients, up to 130% by building a forecast schedule planning system.

Education

Stanford University

Stanford University

Ph.D, Mechanical Engineering

Focus in Systems (Product, Service, Business) Design, Design Thinking, Design for Manufacturing and Six Sigma

Activities and Societies: Design SocietyASMEIEEEINCOSEACM

Stanford University

Stanford University

MS, Mechanical Engineering

Focus in Biomedical Device Design, Reliability Engineering, Operations Research

Activities and Societies: KOSEF Academic Fellow

Publications

A New Project-Based Curriculum of Design Thinking with Systems Engineering Techniques

International Journal of System of Systems Engineering

2013

Agile Project Management for Root Cause Analysis Projects

International Conference on Engineering Design

2013

A New Project-Based Curriculum of Design Thinking with Systems Engineering Techniques

Council of Engineering Systems Universities

2012

Evaluation of Design for Service Innovation Curriculum: Validation Framework and Preliminary Results

nternational Journal of Services Technology and Management

2011

A Validation Regarding Effectiveness of Scenario Graph

ASME International Design Engineering Technical Conferences

2011

Wants Chain Analysis: Human-centered Method for Analyzing and Designing Social Systems

International Conference on Engineering Design

2011

Scenario-based Amorphous Design (SAD) Framework for a Location-based Services Technology

Mobile Human Computer Interaction

2010

Transforming Seamless Positioning Technology into a Business using a Systems Design Approach—Scenario-based Amorphous Design

IEEE- International Systems Conference

2010

Design for Service Innovation: A Methodology for Designing Service as a Business for Manufacturing Companies

International Journal of Services Technology and Management

2010

Preliminary Validation of Scenario-based Design for Amorphous Systems

International Conference on Systems Engineering

2010

Tools for Project-based Active Learning of Amorphous Systems Design: Scenario Prototyping and Cross Team Peer Evaluation

ASME International Design Engineering Technical Conferences

2009

Active Learning Project Sequence: Capstone Experience for Multi-disciplinary System Design and Management Education

International Conference on Engineering Design

2009

Demystifying Ambiguity in The Design of Amorphous Systems

International Conference on Systems Engineering

2009

Scenario-based Design for Amorphous Systems

ASME International Mechanical Engineering Congress and Exposition

2008

Analysis and Design Methodology for Recognizing Opportunities and Difficulties for Product-based Services

Information Processing Society of Japan (IPSJ) Journal

2007

Scenario Graph: Discovering new business opportunities and Failure Modes,”

ASME International Design Engineering Technical Conferences

2007

Analysis and Design Methodology for Product-based Services

Annual Conference of the Japanese Society for Artificial Intelligence

2007

Analysis and Design Methodology for Recognizing Opportunities and Difficulties for Product-based Services

PICMET

2007

CUT PASTE FROM
KIM SUN SPEAKS
Sun Kim

About QbDWorks…http://qbdworks.com/about/

Are you a Scientist in the Pharmaceutical, Biopharmaceutical or Medical Devices industries?

Then you are probably asking:

  • Does Quality-by-Design actually work?
  • Or is it just another program like Lean or Six Sigma?
  • How do I implement QbD successfully?
  • How do I persuade my management?
  • What is the first step?

As a QbD practitioner, I had the same questions and am trying to answer them as I test different elements of QbD.

Through our members’ successes and failures in QbD, you can save time by not having to repeat them yourself. There are many lessons learned and knowledge that you can share with your QbD team.

Who are You?

Sun Kim

My name is Sun Kim. I currently practice Quality-by-Design, transforming how Product Development is executed in Biopharmaceutical, Pharmaceutical, Biologics, and Medical Device industries.

In addition, I teach at Stanford University. My focus of research is Lean Quality by Design.

I received my MS and PHD in Mechanical Engineering at Stanford University and was recently an Asst.  Professor at Keio University in Japan. Prior to Silicon Valley days, I served in the Korean Army and worked at BMW AG in Munich, Germany, where my lifelong pursuit of “Product Development Methodology” began.

Why is an Engineer working in the Bio/pharmaceutical Industry?

Thanks for asking! When working at BMW, as a member of the elite “KREATIV” (Creative) team, my goal was to develop the best technologies and products for the automotive industry. However our approach was somewhat adhoc and heuristics-based. I knew there was a better way.

So I set my heart to learn how best products are developed across all industries. This led me to my PhD research at Stanford University.

Little did I know this would turn out to be more than just a graduate program. On top of the typical coursework, research and publishing, my schedule was packed with hands-on consulting/research projects with GE Healthcare, GE Aviations, Toyota, Nissan, Toshiba, Medtronic, Johnson & Johnson, Startup’s etc.

After contributing to the product development approaches for the Academia, Fortune 500 and Startup’s, I knew my heart was always with Health Care. So I returned to the benches and trenches.

For the last 10 years, I have been working in the Biopharmaceutical, Pharmaceutical and Medical Devices Industry, to make Quality by Design a reality.

So please join me in this Quality by Design journey.

Sign up to connect to the community and receive updates.

Together, we can change how drugs and therapies are developed for our patients and families.

Let’s Connect!

If you’d like to connect, please invite me:

www.linkedin.com/in/kimsunkist/

Previously I worked (full time or consulting) with:

Company Logos

The views expressed on this website are personal opinions and in no way reflect the position of any organization.

  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

FREEMONT CALFORNIA
Map of fremont ca
LAKE ELIZABETH

  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

////
Share

Gatifloxacin

 Uncategorized  Comments Off on Gatifloxacin
Jun 182015
 
Gatifloxacin.svg
GATIFLOXACIN
BMS-206584, CG-5501, AM-1155, Zymar, Bonoq, Gatiflo, AM-1155
(±)-1-Cyclopropyl-6-fluoro-8-methoxy-7-(3-methyl-1-piperazinyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid
Gatifloxacin sold under the brand names GatifloTequin and Zymar, is an antibiotic of the fourth-generation fluoroquinolonefamily,[1] that like other members of that family, inhibits the bacterial enzymes DNA gyrase and topoisomerase IVBristol-Myers Squibb introduced Gatifloxacin in 1999 under the proprietary name Tequin for the treatment of respiratory tract infections, having licensed the medication from Kyorin Pharmaceutical Company of Japan. Allergan produces it in eye-drop formulation under the names Zymar and Zymaxid. In many countries, gatifloxacin is also available as tablets and in various aqueous solutions forintravenous therapy.
Originally developed at Kyorin, gatifloxacin was first licensed to Gruenenthal in Europe, and that company still maintains rights to the oral and injectable formulations of the product. In October 1996, Kyorin licensed gatifloxacin to BMS, granting the company development and marketing rights in the U.S., Canada, Australia, Mexico, Brazil and certain other markets. In 2006, rights to the compound were returned by BMS. Subsequently, Senju and Kyorin signed a licensing agreement regarding the development of ethical eye drops containing the fluoroquinolone. In April 2000, Sumitomo Dainippon Pharma agreed to comarket the oral formulation in Japan. In August of that year, Allergan in-licensed gatifloxacin from Kyorin, gaining development and commercialization rights to the drug in all territories except Japan, Korea, China and Taiwan. The India-based Lupin Pharmaceuticals signed an agreement in June 2004 with Allergan to promote the ophthalmic solution of gatifloxacin in the pediatric specialty area in the U.S. PediaMed Pharmaceuticals also holds rights to the drug. In 2009, Kyorin licensed the drug candidate to Senju in China.
Gatifloxacin is the common name for (±)-1-cyclopropyl-6-fluoro-1,4-dihydro-8-methoxy-7-(3-methyl-1-piperazinyl)-4-oxo-3-quinolinecarboxylic acid (1), one of the most important broad-spectrum antibacterial agents and a member of the fourth-generation fluoroquinolone family.(1)Fluoroquinolones inhibit the enzyme DNA gyrase (topoisomerase II), which is responsible for the supercoiling of the DNA double helix, preventing the replication and repair of bacterial DNA and RNA.(2) Gatifloxacin (1) reached the market in 1999 under the brand name Tequin for the treatment of respiratory tract infections. The drug is available as tablets and aqueous solutions for intravenous therapy as well as eye drop formulation (Zymar).
To date, there are several processes described for the preparation of gatifloxacin, which can be grouped into two main categories: direct substitution of the 7-position fluorine atom of 1-cyclopropyl-6,7-difluoro-1,4-dihydro-8-methoxy-4-oxo-3-quinolinecarboxylic acid (2) by 2-methylpiperazine (Scheme 1),(3-5) and through boron chelate-type intermediates to overcome the diminished reactivity induced by the 8-methoxy group, which uses as starting material the ethyl ester derivative 3 (Scheme 2).(6-9)
SCHEME1
Figure
SCHEME2
Figure
  1. 1.
    Mather, R.; Karenchak, L. M.; Romanowski, E. G.; Kowalski, R. P. Am. J. Ophthalmol.2002, 133 ( 4) 463

  2. 2.
    Corey, E. J.; Czakó, B.; Kürti, L. Molecules and Medicine; Wiley: NJ, 2007; p 135.

  3. 3.
    Masuzawa, K.; Suzue, S.; Hirai, K.; Ishizaki, T. 8-Alkoxyquinolonecarboxylic acid and salts thereof excellent in the selective toxicity and process of preparing the same EP 0 230 295 A3, 1987.

  4. 4.
    Niddam-Hildesheim, V.; Dolitzky, B.-Z.; Pilarsky, G.; Steribaum, G. Synthesis of Gatifloxacin WO 2004/069825 A1, 2004.

  5. 5.
    Ruzic, M; Relic, M; Tomsic, Z; Mirtek, M. Process for the preparation of Gatifloxacin and regeneration of degradation products WO 2006/004561 A1, 2006.

  6. 6.
    Iwata, M.; Kimura, T.; Fujiwara, Y.; Katsube, T. Quinoline-3-carboxylic acid derivatives, their preparation and use EP 0 241 206 A2, 1987.

  7. 7.
    Sanchez, J. P.; Gogliotti, R. D.; Domagala, J. M.; Garcheck, S. J.; Huband, M. D.; Sesnie,J. A.; Cohen, M. A.; Shapiro, M. A. J. Med. Chem. 1995, 38, 4478

  8. 8.
    Satyanarayana, C.; Ramanjaneyulu, G. S.; Kumar, I. V. S. Novel crystalline forms of Gatifloxacin WO 2005/009970 A1 2005.

  9. 9.
    Takagi, N.; Fubasami, H.; Matsukobo, H.; (6,7-Substituted-8-alkoxy-1-cyclopropyl-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid-O3,O4)bis(acyloxy-O)borates and the salts thereof, and methods for their manufacture EP 0 464 823 A1, 1991.

………………………….

WO 2005009970

http://www.google.com/patents/WO2005009970A1?cl=en

preparation of Gatifloxacin hemihydrate from Ethyl-1- Cyclopropyl-6, 7-difluoro-8-methoxy-4-oxo-l, 4-dihydro-3-quinoline carboxylate through boron difluoride chelate. Ethyl-1-cyclopropyl- 6, 7-difluoro-8-methoxy-4-oxo-l, 4-dihydro-3-quinoline carboxylate is reacted with aqueous hydrofluoroboric acid followed by condensation with 2-methyl piperazine in polar organic solvent resulting in an intermediate l-Cyclopropyl-7- (3-methyl piperazin-1- yl). -6-fluoro-8-methoxy-4-oxo-l, 4-dihydro-3-quinoline carboxylic acid boron difluoride chelate. This intermediate may be further hydrolyzed to yield Gatifloxacin. Gatifloxacin so obtained may needs purification to yield high purity product. However to obtain directly high purity Gatifloxacin it is desirable to isolate the intermediate by cooling to low temperatures . Treating with an alcohol or mixture of alcohols purifies this intermediate. The purified condensed chelate in aqueous ethanol on hydrolysis with triethylamine followed by crystallization in ethanol gives Gatifloxacin hemihydrate with high purity.

STAGE – I:

 

Figure imgf000006_0001

Ethyl l-cyclopropyl-6,7-difluoro-8-met oxy l-Cycloproρyl-6, 7-difluoro-8-methoxy -4-oxo-l, -dihydro-3-quinoline -4-oxo-l, 4-dihydro-3-quinoline carboxylate carboxylic acid boron difluoride chelate

STAGE – II :

 

Figure imgf000007_0001

l-Cycloprop l-7- ( 3-methylpiperazin-l-yl.

Figure imgf000007_0002

6-fluoro~8-methoxy-4-oxo-l , 4-dihydro-3- carboxylicacid borondifluoride chelate quinoline carboxylicacid borondifluoride chelate

STAGE -III :

 

Figure imgf000007_0003

l-Cyclopropyl-7- (3- ethylpiperaz.in-l-yl . GATIFLOXACIN

-6-fluoro-8-methoxy-4-oxo-l , 4-dihydro-3- quinoline carboxylicacid borondifluoride chelate

Example-I: Preparation of Gatifloxacin • with isolation of intermediate (boron difluoride chelate derivative)

Stage-1: Preparation of l-cyclopropyl-6, 7-di luoro-8-methoxy-4-oxo- 1, 4-dihydro-3-quinoline carboxylic acid boron difluoride chelate. Ethyl-l-cyclopropyl-6, 7-difluoro-8-methoxy-4-oxo-l, -dihydro-3- quinόline carboxylate (100g)is suspended in ,40%aq..hydrofluoroboric acid -(1000 ml). Temperature of • the reaction mass is raised and maintained at 95°C to 100°C for 5hrs followed by cooling to 30°C – 35°C. Water (400 ml) is added and maintained at 25°C – 30°C for 2hrs . Product is filtered, washed with water (500 ml) and dried at 40°C – 45°C to constant weight. Dry weight of the product: 101.6 g (Yield: 95.8 %)

Stage-2: Preparation of 1- Cyclopropyl-7- (3-methylpiperazin-l-yl) – 6-fluoro-8-methoxy-4-oxo-l, -dihydro-3-quinoline carboxylic acid boron difluoride chelate

100 g of Boron difluoride chelate derivative prepared as above in stage-1 is suspended in acetonitrile (800 ml) , to that 2-methyl piperazine (44.0 g, 1.5 mole equiv.) is added and mixed for 15 min to obtain a clear solution. The reaction mass is maintained at 30°C – 35°C for 12 hrs followed by cooling to -10°C to -5°C. The reaction mass is maintained at -10°C to -5°C for 1 hr. The product is filtered and dried at 45°C – 50°C to constant weight. Dry weight of the product: 116.0 g (Yield: 93.9 %) .

The condensed chelate (100 g) prepared as above is suspended in methanol (1500 ml), maintained at 40°C – 45°C for 30 min. The reaction mass is gradually cooled, maintained for 1 hr at -5°C to 0°C. The product is filtered, washed with methanol (50 ml) and dried at 45°C – 50°C to constant weight. Dry weight of the product: 80.0 g (Yield: 80.0 %)

Stage -3: Preparation of Gatifloxacin (Crude)

The pure condensed chelate (100.0 g) prepared as above in stage-2 is suspended in 20% aq. ethanol (1000 ml) , the temperature is raised and maintained at 75°C to 80°C for 2 hrs. The reaction mass is cooled, filtered to remove insolubles, distilled under vacuum to remove solvent. Fresh ethanol (200 ml) is added and solvent is removed under vacuum at temperature below 50°C. Ethanol (200 ml) is added to the residue and gradually cooled to -10°C to -5°C. The reaction mass is mixed at -10°C to -5°C for 1 hr and then filtered. The wet cake is washed with ethanol (25 ml) and dried at 45°C – 50°C to constant weight.

The dry weight of the Gatifloxacin is 83.3 g (Yield: 91.7 %)

Stage- 4: Purification of crude Gatifloxacin

Crude Gatifloxacin (100.0 g) prepared as above in stage-3 is suspended in methanol (4000 ml), the temperature is raised and maintained at 60°C to 65°C for 20 min. to get a clear solution. Activated carbon (5 g) is added, maintained for 30 min and the solution is filtered. The filtrate is concentrated to one third of its original volume under vacuum at temperature below 40°C. The reaction mass is gradually cooled and maintained at -10°C to -5°C for 2 hrs. The product is filtered, washed with methanol (50 ml) and dried at 45°C – 50°C to constant weight. The dry weight of the pure Gatifloxacin is 76.0 g (Yield: 76.0 %)

Example-II: Preparation of Gatifloxacin without isolation of intermediate (boron difluoride chelate derivative)

Stage-1: Preparation of l-cyclopropyl-6, 7-difluoro-8-methoxy-4- oxo-1, 4-dihydro-3-quinoline carboxylic acid boron difluoride chelate.

Ethyll-cyclopropyl-6, 7-difluoro-8-methoxy-4-oxo-l, 4-dihydro-3- quinoline carboxylate (lOOg) is suspended in 40% aq. hydrofluoroboric acid (1000 ml) . Temperature of the reaction mass is raised and maintained at 95°C to 100°C for 5 hrs followed by cooling to 30°C – 35°C. 400 ml DM water is added, maintained at 25°C – 30°C for 2hrs . The product is filtered, washed with DM water (500 ml) and dried at 40°C – 45°C to constant weight. The dry wt is 102.5 g (Yield: 96.6 %)

Stage – 2: Preparation of Gatifloxacin (Crude)

The boron difluoride chelate derivative (100 g) prepared as above in stage-1 is suspended in acetonitrile (800 ml) , 2-methyl piperazine (44 g, 1.5 mole equiv.) is added and mixed for 15 min to obtain a clear solution. The reaction mass is maintained at 30°C – 35°C for 12 hrs. Removed the solvent by vacuum distillation. 20% Aq. ethanol (1000 ml) is added, raised the temperature and maintained at 75°C to 80°C for 2 hrs. The reaction mass is cooled, filtered to remove insolubles. The filtrate is distilled under vacuum to remove solvent completely. Fresh ethanol (250 ml) is added and distilled under vacuum at temperature below 50°C. Fresh Ethanol (250 ml) is added to the residue and gradually cooled to -10°C to -5°C. The reaction mass is maintained at -10°C to -5°C for 1 hr and filtered. The wet cake is washed with ethanol (30 ml) and dried at 45°C – 50°C to constant weight.

The dry weight of the Gatifloxacin is 73.5 g (Yield: 65.4 %)

Stage -3: Purification of crude Gatifloxacin

Crude Gatifloxacin (80.0 g) prepared as above in stage-2 is suspended in methanol (2000 ml) , the temperature is raised and maintained at 60°C to 65°C for 20 min. to get a clear solution. The reaction mixture is filtered. The filtrate is gradually cooled and maintained at -10°C to -5°C for 2 hrs. The product is filtered, washed with methanol (50 ml) and dried at 45°C – 50°C to constant weight.

The dry weight of the pure Gatifloxacin is 56.0 g (Yield: 70.0 %)

……………………….

WO 2005047260

http://www.google.co.in/patents/WO2005047260A1?cl=en

Gatifloxacin is the international common name of l-cyclopropyl-6-fluoro-l, 4-dihydro-8-methoxy- 1- (3-methyl-l-piperazinyl) -4-oxo-3-guinolin-carboxylic acid of formula (I) , with application in medicine and known for its antibiotic activity:

 

Figure imgf000002_0001

European patent application EP-A-230295 discloses a process for obtaining gatifloxacin that consists on the reaction of compound (II) with 2-

 

Figure imgf000002_0002

In this process the gatifloxacin is isolated in the form of a hemihydrate after a laborious process of column chromatography and recrystallisation in methanol, which contributes towards making the final yield lower than 20% by weight. Moreover, in said process an undesired by-product is formed, resulting from demethylation at position 8 of the ring. European patent application EP-A-241206 discloses a process for preparing gatifloxacin, whose final steps are as follows:

 

Figure imgf000003_0001

(III) H ft N Me H DMSO

Gatifloxacin (I)

Figure imgf000003_0002

(IV) This process uses the intermediate compound (III) , which has been prepared and isolated in a separate operation, while the intermediate compound (IV) is also isolated before proceeding to its conversion into gatifloxacin by treatment with ethanol in the presence of triethylamine. The overall yield from these three steps is lower than 40%. These disadvantages — a synthesis involving several steps, low yields, and the need to isolate the intermediate products — hinder the production of gatifloxacin on an industrial scale. There is therefore a need to provide a process for preparing gatifloxacin with a good chemical yield, without the need to isolate the intermediate compounds and that substantially avoids demethylation in position 8 of the ring. The processes termed in English “one pot” are characterised in that the synthesis is carried out in the same reaction vessel, without isolating the intermediate compounds, and by means of successive addition of the reacting compounds. The authors of the present invention have discovered a simplified process for preparing gatifloxacin which does not require isolation of the intermediate compounds .

 

Example 1: Preparing gatifloxacin from compound (II) 10 g (0.0339 moles, 1 equivalent) of compound

(II) is placed in a flask, 30 ml of acetonitryl (3 volumes) is added and this is heated to a temperature of 76-80° C.

Figure imgf000004_0001

Once reflux has been attained, and being the temperature maintained, 3.28 g (0.0203 moles, 0.6 equivalents) of hexamethyldisilazane (HMDS) is added with a compensated adding funnel. Once addition is completed, the reaction is maintained with stirring for 1 hour at a temperature of 76-80° C. Once this period has elapsed, the reaction mixture is cooled to a temperature ranging between 0 and 15° C, and 5.78 g (0.0407 moles, 1.2 equivalents) of boron trifluoride ethyletherate is added while keeping the temperature below 15° C. Once addition is completed, the temperature is allowed to rise to 15- 25° C and it is kept under these conditions for approximately 2 hours. The pH of the mixture is then adjusted to an approximate value of 9 with triethylamine (approximately 2 ml) . To the resulting suspension is added a solution of 10.19 g (0.1017 moles, 3 equivalents) of 2-methylpiperazine in 28 ml of acetonitryl, while maintaining the temperature between 15 and 25° C. The resulting amber solution is kept with stirring under these conditions for approximately 3 hours . Once the reaction has been completed, the solution is distilled at low pressure until a stirrable paste is obtained. At this point 50 ml of methanol is added, the resulting suspension is raised to a temperature of 63-67° C and is kept under these conditions for approximately 5 hours . Once the reaction has been completed, the mixture is cooled to a temperature of 25-35° C in a water bath, and then at a temperature of 0-5° C in a water/ice bath for a further 1 hour. The resulting precipitate is filtered, washed with cold methanol (2 x 10 ml) and dried at 40° C in a vacuum oven to constant weight. 10.70 g of crude gatifloxacin is obtained, having a water content of 2.95% by weight. The yield of the process is 81.8%.

The crude product is crystallised in methanol by dissolving 20 g of crude gatifloxacin in 1 1 of methanol (50 volumes) at a temperature of 63-67° C. Once all the product has been dissolved, the solution is left to cool to a temperature of 30-40° C, and then to a temperature of 0-5° C in a water/ice bath, maintaining it under these conditions for 1 hour. The resulting suspension is filtered and the solid retained is washed with 20 ml (1 volume) of cold methanol. The solid obtained is dried at 40° C in a vacuum oven to provide 18.65 g of gatifloxacin with a water content of 2.36% by weight.

The overall yield from the compound (II) is 77.7%, with a purity exceeding 99.8% as determined by HPLC chromatography. The content of by-product resulting from demethylation in position 8 of the ring is lower than 0.1% as determined by HPLC chromatography.

Gatifloxacin ball-and-stick.png
Systematic (IUPAC) name
1-cyclopropyl-6-fluoro- 8-methoxy-7-(3-methylpiperazin-1-yl)- 4-oxo-quinoline-3-carboxylic acid
Clinical data
Trade names Zymar
AHFS/Drugs.com monograph
MedlinePlus a605012
  • ℞ (Prescription only)
Oral (discontinued),
Intravenous(discontinued)
ophthalmic
Pharmacokinetic data
Protein binding 20%
Half-life 7 to 14 hours
Identifiers
112811-59-3 Yes
J01MA16 S01AE06
PubChem CID: 5379
DrugBank DB01044 Yes
ChemSpider 5186 Yes
UNII 81485Y3A9A Yes
KEGG D08011 Yes
ChEBI CHEBI:5280 Yes
ChEMBL CHEMBL31 Yes
NIAID ChemDB 044913
Chemical data
Formula C19H22FN3O4
375.394 g/mol

PAPER

Abstract Image

An improved process to obtain gatifloxacin (1) through use of boron chelate intermediates has been developed. The methodology involves an initial activation step which accelerates the formation of the first chelate under low-temperature conditions and prevents demethylation of the starting material. To increase the overall yield and to avoid the isolation and manipulation of the resulting intermediates, the process has been designed to be carried out in one pot. As a result, we present here an easy, scaleable and substantially impurity-free process to obtain gatifloxacin (1) in high yield.

A High-Throughput Impurity-Free Process for Gatifloxacin

Department of Research & Development, Química Sintética S.A., c/ Dulcinea s/n, 28805 Alcalá de Henares, and Department of Organic Chemistry, University of Alcalá, 28871 Madrid, Alcalá de Henares, Spain
Org. Process Res. Dev., 2008, 12 (5), pp 900–903
DOI: 10.1021/op800042a
gatifloxacin (1) as white crystals. Yield 32.3 kg, (93%); purity by HPLC 99.87%; Assay by HPLC 100.8%; mp 167−168 °C(18) (Lit. (J. Med. Chem. 1995, 38, 4478)159−162 °C).
18

DSC analysis showed two endothermic peaks at 166.2 °C (T onset = 164.3 °C) and 190.0 °C (T onset = 188.2 °C) and an exothermic one at 168.1 °C. The shape of this DSC curve is characteristic of a monotropic transition between crystalline forms

Water content by Karl Fischer 3.0%(19) MS m/z 376 (M+ + H);
19

Although there are several hydrates described for gatifloxacin such as, among others, the hemimydrate, sesquihydrate, and pentahydrate(Raghavan, K. S.; Ranadive, S. A.;Gougoutas, J. Z.; Dimarco, J. D.; Parker, W. L.; Dovich, M.; Neuman, A.Gatifloxacin pentahydrate. WO 2002/22126 A1, 2002) , the Gatifloxacin obtained by the present procedure does not seem to form a stoichometric hydrate, but instead it retains moisture.

Thus, the product is usually obtained with a Karl-Fischer value below 1% after drying, but it can absorb moisture until a final content of about 3%. This water content can vary between 2.0% and 3.5%, depending on the relative humidity of the environment. DSC analysis revealed a broad endothermic signal with minimum at 76 °C, while TGA analysis showed that the product loses all the water below 80 °C.

No loss of weight is registered when the product melts, and the weight is constant until the decomposition of the material at about 200 °C. On the basis of these results, it can be said that the water content of the gatifloxacin obtained by the present process is retained moisture instead of water belonging to the lattice. The shape of the derivative of the weight curve at the beginning of the analysis shows that the sample has already lost part of the moisture when the register starts. This is probably due to the sample starting to lose weight when makes contact with the dry atmosphere of the TGA oven that could explain the different values obtained for water content of the analyzed sample by TGA (1.90%) and Karl-Fischer (2.64%) methods.

 1H NMR (DMSO-d6) δ 0.97 (d, J = 6.1 Hz, 3H), 1.04 (m, 2H), 1.15 (m, 2H), 2.75−2.94 (m, 4H) 3.14 (m, 1H), 3.30 (m, 2H), 3.74 (s, 3H), 4.15 (m, 1H), 7.70 (d, JH−F = 12.2 Hz, 1H), 8.67 (s, 1H). 
13C NMR (DMSO-d6) δ 8.40, 8.42, 18.66, 40.28, 45.46, 50.17, 50.29 (d, JC−F = 3.44 Hz), 57.36 (d, JC−F = 3.74 Hz), 62.15, 106.0 (d, JC−F = 22.7 Hz), 106.04, 120.05 (d, JC−F = 8.6 Hz), 133.6 (d, JC−F = 1.1 Hz), 138.9 (d, JC−F = 11.9 Hz), 145.2 (d, JC−F = 5.87 Hz), 149.88, 155.06 (d, JC−F = 249.2 Hz), 165.56, 175.56 (d, JC−F = 3.3 Hz).
 19F NMR (DMSO-d6) δ −120.4 (d, J = 12.2 Hz).
Anal. Calcd for C19H22N3O4F + 3.0% H2O; C, 58.95; H, 6.07; N, 10.85. Found: C, 58.90; H, 5.82; N, 10.90.

Side-effects and removal from the market

Canadian study published in the New England Journal of Medicine in March 2006 claims Tequin can have significant side effectsincluding dysglycemia.[2] An editorial by Dr. Jerry Gurwitz in the same issue called for the Food and Drug Administration (FDA) to consider giving Tequin a black box warning.[3] This editorial followed distribution of a letter dated February 15 by Bristol-Myers Squibb to health care providers indicating action taken with the FDA to strengthen warnings for the medication.[4] Subsequently it was reported on May 1, 2006 that Bristol-Myers Squibb would stop manufacture of Tequin, end sales of the drug after existing stockpiles were exhausted, and return all rights to Kyorin.[5]

Union Health and Family Welfare Ministry of India on 18 March 2011 banned the manufacture, sale and distribution of Gatifloxacin as it caused certain adverse side effects[6]

Contraindications

Diabetes[7]

Availability

Gatifloxacin is currently available only in the US and Canada as an ophthalmic solution.

In China it is sold in tablet as well as in eye drop formulations.

Ophthalmic anti-infectives are generally well tolerated. The concentration of the drug observed following oral administration of 400 mg gatifloxacin systemically is approximately 800 times higher than that of the 0.5% Gatifloxacin eye drop. Given as an eye drop, Gatifloxacin Ophthalmic Solution 0.3% & 0.5% cause very low systemic exposures. Therefore, the systemic exposures resulting from the gatifloxacin ophthalmic solution are not likely to pose any risk for systemic toxicities.

  • The reaction of 1-bromo-2,4,5-trifluoro-3-methoxybenzene (I) with CuCN and N-methyl-2-pyrrolidone at 150 C gives 2,4,5-trifluoro-3-methoxybenzonitrile (II), which by treatment with concentrated H2SO4 yields the benzamide (III) The hydrolysis of (III) with H2SO4 -. water at 110 C affords 2,4,5-trifluoro-2-methoxybenzoic acid (IV), which by reaction with SOCl2 is converted into the acyl chloride (V). The condensation of (V) with diethyl malonate by means of magnesium ethoxide in toluene affords diethyl 2- (2,4,5-trifluoro-3-methoxybenzoyl) malonate (VI), which by treatment with p-toluenesulfonic acid in refluxing water gives ethyl 2- (2,4,5-trifluoro-3-methoxybenzoyl) acetate (VII). The condensation of (VII) with triethyl orthoformate in refluxing acetic anhydride yields 3-ethoxy -2- (2,4,5-trifluoro-3-methoxybenzoyl) acrylic acid ethyl ester (VIII), which is treated with cyclopropylamine (IX) to afford the corresponding cyclopropylamino derivative (X). The cyclization of (X) by means of NaF in refluxing DMF gives 1-cyclopropyl-6,7-difluoro-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylic acid ethyl ester (XI), which is hydrolyzed with H2SO4 in acetic acid to yield the corresponding free acid (XII). Finally, this compound is condensed with 2-methylpiperazine (XIII) in hot DMSO.

 

Gatifloxacin
Title: Gatifloxacin
CAS Registry Number: 112811-59-3
CAS Name: 1-Cyclopropyl-6-fluoro-1,4-dihydro-8-methoxy-7-(3-methyl-1-piperazinyl)-4-oxo-3-quinolinecarboxylic acid
Trademarks: Tequin (BMS); Zymar (Allergan)
Molecular Formula: C19H22FN3O4
Molecular Weight: 375.39
Percent Composition: C 60.79%, H 5.91%, F 5.06%, N 11.19%, O 17.05%
Literature References: Fluorinated quinolone antibacterial. Prepn: K. Masuzawa et al., EP 230295eidem, US 4980470 (1987, 1990 both to Kyorin); J. P. Sanchez et al., J. Med. Chem. 38, 4478 (1995); of the sesquihydrate: T. Matsumoto et al., US5880283 (1999 to Kyorin). In vitro antibacterial activity: A. Bauernfeind, J. Antimicrob. Chemother. 40, 639 (1997); H. Fukuda et al., Antimicrob. Agents Chemother. 42, 1917 (1998). Clinical pharmacokinetics: M. Nakashima et al., ibid. 39, 2635 (1995). Clinical study in urinary tract infection: H. Nito, 10th Mediterranean Congr. Chemother. 1996, 327; in respiratory tract infection: S. Sethi, Expert Opin. Pharmacother. 4, 1847 (2003).
Properties: Pale yellow prisms from methanol as hemihydrate, mp 162°.
Melting point: mp 162°
 
Derivative Type: Sesquihydrate
CAS Registry Number: 180200-66-2
Manufacturers’ Codes: AM-1155
Molecular Formula: C19H22FN3O4.1½H2O
Molecular Weight: 384.40
Percent Composition: C 59.37%, H 6.03%, F 4.94%, N 10.93%, O 18.73%
Therap-Cat: Antibacterial.
Keywords: Antibacterial (Synthetic); Quinolones and Analogs

References

  1.  Burka JM, Bower KS, Vanroekel RC, Stutzman RD, Kuzmowych CP, Howard RS (July 2005). “The effect of fourth-generation fluoroquinolones gatifloxacin and moxifloxacin on epithelial healing following photorefractive keratectomy”Am. J. Ophthalmol. 140 (1): 83–7. doi:10.1016/j.ajo.2005.02.037.PMID 15953577.
  2.  Park-Wyllie, Laura Y.; David N. Juurlink; Alexander Kopp; Baiju R. Shah; Therese A. Stukel; Carmine Stumpo; Linda Dresser; Donald E. Low; Muhammad M. Mamdani (March 2006).“Outpatient Gatifloxacin Therapy and Dysglycemia in Older Adults”The New England Journal of Medicine 354 (13): 1352–1361. doi:10.1056/NEJMoa055191PMID 16510739. Retrieved 2006-05-01. Note: publication date 30 March; available on-line 1 March
  3.  Gurwitz, Jerry H. (March 2006). “Serious Adverse Drug Effects — Seeing the Trees through the Forest”The New England Journal of Medicine 354 (13): 1413–1415.doi:10.1056/NEJMe068051PMID 16510740. Retrieved2006-05-01.
  4.  Lewis-Hall, Freda (February 15, 2006). “Dear Healthcare Provider:” (PDF). Bristol-Myers Squibb. Retrieved May 1, 2006.
  5.  Schmid, Randolph E. (May 1, 2006). “Drug Company Taking Tequin Off Market”Associated Press. Archived from the original on November 25, 2007. Retrieved 2006-05-01.[dead link]
  6.  “Two drugs banned”The Hindu (Chennai, India). 19 March 2011.
  7.  Peggy Peck (2 May 2006). “Bristol-Myers Squibb Hangs No Sale Sign on Tequin”. Med Page Today. Retrieved 24 February2009.

 

EP0610958A2 * 20 Jul 1989 17 Aug 1994 Ube Industries, Ltd. Intermediates in the preparation of 4-oxoquinoline-3-carboxylic acid derivatives
ES2077490A1 * Title not available
Citing Patent Filing date Publication date Applicant Title
WO2008126384A1 31 Mar 2008 23 Oct 2008 Daiichi Sankyo Co Ltd Method for producing quinolone carboxylic acid derivative
CN101659654B 28 Aug 2008 6 Nov 2013 四川科伦药物研究有限公司 2-Methylpiperazine fluoroquinolone compound and preparation method and application thereof
CN102351843A * 18 Aug 2011 15 Feb 2012 张家口市格瑞高新技术有限公司 Synthesis method of 2-methyl piperazine lomefloxacin
EP1832587A1 * 2 Mar 2007 12 Sep 2007 Quimica Sintetica, S.A. Method for preparing moxifloxacin and moxifloxacin hydrochloride
US7365201 2 Mar 2006 29 Apr 2008 Apotex Pharmachem Inc. Process for the preparation of the boron difluoride chelate of quinolone-3-carboxylic acid
US7875722 30 Sep 2009 25 Jan 2011 Daiichi Sankyo Company, Limited Method for producing quinolone carboxylic acid derivative
EP0464823A1 * Jul 4, 1991 Jan 8, 1992 Kyorin Pharmaceutical Co., Ltd. (6,7-Substituted-8-alkoxy-1-cyclopropyl-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid-O3,O4)bis(acyloxy-O)borates and the salts thereof, and methods for their manufacture
US4997943 * Mar 31, 1987 Mar 5, 1991 Sankyo Company Limited Quinoline-3-carboxylic acid derivatives
Citing Patent Filing date Publication date Applicant Title
CN101659654B Aug 28, 2008 Nov 6, 2013 四川科伦药物研究有限公司 2-Methylpiperazine fluoroquinolone compound and preparation method and application thereof
CN102351843A * Aug 18, 2011 Feb 15, 2012 张家口市格瑞高新技术有限公司 Synthesis method of 2-methyl piperazine lomefloxacin
* Cited by examiner

 

TAKE A TOUR

 

 

TAKE A TOUR

 

Amritsar, punjab, India

  1. Amritsar – Wikipedia, the free encyclopedia

    https://en.wikipedia.org/?title=Amritsar

    Amritsar is one of the largest cities of the Punjab state in India. The city origin lies in the village of Tung, and was named after the lake founded by the fourth Sikh  …

    Map of amritsar
    Punjab
    GOLDEN TEMPLE
    .
    Tandoori chicken at Surjit Food Plaza. amritsar

    Bullet marks on the walls of the park premises

    The Jallianwalla Bagh in 1919, months after the massacre

    Mealtime at the Golden Temple Amritsar

     

    Golden Temple – Harmandir Sahib: Free food for everyone

    Sri Guru Ram Dass Jee International Airport in Amritsar

    Amritsar – Wagah Border – Street food stall | Explore bernic… |

    Charles W. BartlettAmritsar (The Lake by the Golden Temple) 1920

    tandoori chicken

    • Golden Temple

    • Maharaja Ranjit Singh’s Ram Bagh Gardens

    • Golden Temple

    • Durgiana Temple

    • The holy water

    • Jallianwala Bagh

    • Jallianwala Bagh

    • The holy water

    • Golden Temple

    • Golden Temple

    • Sikh Gurdwara

    • The holy water

    Night view of the Harmandir Sahib

    Night view of the Harmandir Sahib
    ///////////
Share

NMR Structure Elucidation of Small Organic Molecules and Natural Products: Choosing ADEQUATE vs HMBC

 Uncategorized  Comments Off on NMR Structure Elucidation of Small Organic Molecules and Natural Products: Choosing ADEQUATE vs HMBC
Jun 092015
 
Abstract Image

Long-range heteronuclear shift correlation methods have served as the cornerstone of modern structure elucidation protocols for several decades. The 1H–13C HMBC experiment provides a versatile and relatively sensitive means of establishing predominantly 3JCHconnectivity with the occasional 2JCH or 4JCH correlation being observed. The two-bond and four-bond outliers must be identified specifically to avoid spectral and/or structural misassignment. Despite the versatility and extensive applications of the HMBC experiment, it can still fail to elucidate structures of molecules that are highly proton-deficient, e.g., those that fall under the so-called “Crews rule”. In such cases, recourse to the ADEQUATE experiments should be considered. Thus, a study was undertaken to facilitate better investigator understanding of situations where it might be beneficial to apply 1,1- or 1,n-ADEQUATE to proton-rich or proton-deficient molecules. Equipped with a better understanding of when a given experiment might be more likely to provide the necessary correlation data, investigators can make better decisions on when it might be advisible to employ one experiment over the other. Strychnine (1) and cervinomycin A2 (2) were employed as model compounds to represent proton-rich and proton-deficient classes of molecules, respectively. DFT methods were employed to calculate the relevant nJCHheteronuclear proton–carbon and nJCC homonuclear carbon–carbon coupling constants for this study.

NMR Structure Elucidation of Small Organic Molecules and Natural Products: Choosing ADEQUATE vs HMBC

† Discovery and Preclinical Sciences, Process and Analytical Chemistry, NMR Structure Elucidation, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
‡ Discovery and Preclinical Sciences, Process and Analytical Chemistry, NMR Structure Elucidation, Merck Research Laboratories, Rahway, New Jersey 07065, United States
J. Nat. Prod., 2014, 77 (8), pp 1942–1947
DOI: 10.1021/np500445s
*Tel: 908-740-3990. Fax: 908-740-4042. E-mail: alexei.buevich@merck.com.
more from my collection
Using HMBC and ADEQUATE NMR Data To Define and Differentiate Long-Range Coupling Pathways: Is the Crews Rule Obsolete?
It is well known that as molecules become progressively more proton-deficient, structure elucidation becomes correspondingly more challenging. When the ratio of 1H to 13C and the sum of other heavy atoms falls below 2, an axiom that has been dubbed the “Crews rule” comes into play. The general premise of the Crews rule is that highly proton-deficient molecules may have structures that are difficult, and in some cases impossible, to elucidate using conventional suites of NMR experiments that include proton and carbon reference spectra, COSY, multiplicity-edited HSQC, and HMBC (both 1H–13C and 1H–15N). However, with access to modern cryogenic probes and microcyroprobes, experiments that have been less commonly utilized in the past and new experiments such as inverted 1JCC 1,n-ADEQUATE are feasible with modest sized samples. In this light, it may well be time to consider revising the Crews rule. The complex, highly proton-deficient alkaloid staurosporine (1) is used as a model proton-deficient compound for this investigation to highlight the combination of inverted 1JCC 1,n-ADEQUATE with 1.7 mm cryoprobe technology.

Using HMBC and ADEQUATE NMR Data To Define and Differentiate Long-Range Coupling Pathways: Is the Crews Rule Obsolete?

Gary E Martin
† Discovery and Preclinical Sciences, Process and Analytical Chemistry, Structural Elucidation Group, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
‡ Discovery and Preclinical Sciences, Process and Analytical Chemistry, Structural Elucidation Group, Merck Research Laboratories, Rahway, New Jersey 07065, United States
§ Discovery and Preclinical Sciences, Process and Analytical Chemistry, Structural Elucidation Group, Merck Research Laboratories, Summit, New Jersey 07901, United States
J. Nat. Prod., 2013, 76 (11), pp 2088–2093
DOI: 10.1021/np400562u
Publication Date (Web): November 6, 2013
Copyright © 2013 The American Chemical Society and American Society of Pharmacognosy
*Phone: 908-473-5398. Fax: 908-473-6559. E-mail: gary.martin2@merck.com.
TAKE A TOUR
Austral Islands
Map of australs
Tubuai is in the Austral Archipelago. These island chains are spread out over an area the size of Europe with 120 islands in all, 25 of which that are …
////////
Share

Juliana Aristéia de Lima

 SYNTHESIS, Uncategorized  Comments Off on Juliana Aristéia de Lima
Jun 062015
 

.

Juliana ARISTÉIA DE LIMA,

PhD in Chemistry (31, Brazil)

Juliana Aristéia de Lima holds a Ph.D. in chemistry and is currently conducting research at the State University of Campinas, located in the state of São Paulo in Southeast Brazil. She works on the development of biodegradable polymers blends (biopolymers).

State University of Campinas, Brazil

 

LINKS

http://www.researchgate.net/profile/Juliana_De_Lima2

 

 

Research focus: Sustainable management in the chemical industry

Juliana Aristéia de Lima holds a Ph.D. in chemistry and is currently conducting research at the State University of Campinas, located in the state of São Paulo in Southeast Brazil. She works on the development of biodegradable polymers blends (biopolymers). Polymers are ubiquitous in modern everyday life, most notably in the form of plastics. Because of that, it is essential for the future that they don’t constitute a waste problem in the way they often have in the past, but instead degrade in the way natural materials like paper or food would.

With her research, Juliana Aristéia de Lima addresses an important topic in the area of sustainable resource management. In the future, the Brazilian researcher also hopes to work on conductive ionic liquids, which could serve as solvents for preparation of polymer membranes. She is aspiring to a postdoctoral research position in Germany and wants to make new contacts with German experts in industry and academia for that purpose.

Universidade Estadual de Campinas

 

Map of unicamp

.

.

.

 

Take a tour

SOLOMON ISLANDS

HONIARA

Image result for solomon island

Malaita, Solomon Islands …

 

 

 

.

http://graphics8.nytimes.com/images/2010/10/12/science/12saw_street/12saw_street-articleInline.jpg

.

Gizo, on Ghizo Island, is the capital of the Solomon Islands’ far-flung Western Province, a paradise of coral cays, atolls, lagoons and volcanic islands east of Papua New Guinea where, on a rainy day in late July, crowds flocked to the local netball court for the opening of the inaugural Akuila Talasasa Arts Festival.

 

Motorised canoes lined up in Gizo Harbour near the daily marketplace. Picture: David May

Motorised canoes lined up in Gizo Harbour near the daily marketplace.

 

 

 

 

.

Gizo Hotel, the best accommodation on Ghizo Island. Picture: David May

Vona Vona Lagoon and the beach at Zipolo Habu Resort on Lola Island. Picture: David May

Water views from Zipolo Habu Resort on Lola Island. Picture: David May

/////////

Share
Follow

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

Join other followers: