Biochips for better cancer therapy
Cancer is the second leading cause of disease-related death in the United States, and may overtake heart disease without aggressive new therapies. One promising area of cancer treatment is photodynamic therapy (PDT), which combines the agents of a photosensitive drug, light, and oxygen to attack cancerous tumors and lesions locally in the targeted region of the body by selective optical illumination.
Research being conducted by Prof. Euisik Yoon’s group aims to dramatically accelerate progress in PDT. And it is being accomplished through a lab-on-a-chip measuring about the size of a quarter. At the heart of this biochip is a 5x5mm testing area that will test the interaction of the drug, light, and oxygen simultaneously, generating results in a fraction of the time of current testing practices.
“In cancer research doctors are always looking for better drugs,” explained Dr. Xia Lou, a postdoctoral fellow in Prof. Yoon’s group. “But there has…
View original post 392 more words
Clinafloxacin from kyorin

Clinafloxacin
7-(3-Aminopyrrolidin-1-yl)-8-chloro-1-cyclopropyl-6-fluoro-4-oxoquinoline-3-carboxylic acid
7-(3-Amino-1-pyrrolidinyl)-8-chloro-1-cyclopropyl-6-fluoro-l,4-dihydro-4-oxo-3-quinolinecarboxylic acid
(±)-7-(3-Amino-1-pyrrolidinyl)-8-chloro-1-cyclopropyl-6-fluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid
105956-99-8 cas no
Clinafloxacin (INN) is a fluoroquinolone antibiotic. Its use is associated with phototoxicity and hypoglycaemia.[1]
Clinafloxacin is a novel quinolone with wide activity against the plethora of microorganisms encountered in intraabdominal infections.
Clinafloxacin is a chlorofluoroquinolone with excellent bioavailability and activity against gram-positive, gram-negative, and anaerobic pathogens . Typical MICs for α-streptococci are 0.06–0.12 µg/mL . MIC90 values for methicillin-resistant Staphylococcus aureus (MRSA) average 1.0 µg/mL. The MIC90 for enterococci is typically 0.5 µg/mL . Both intravenous and oral formulations have been developed . Several studies have demonstrated the efficacy of clinafloxacin monotherapy for serious infections Clinafloxacin was also active in animal models of endocarditis, including endocarditis due to ciprofloxacin-resistant S. aureus infection .


-
A mixture of 8-chloro-1-cyclopropyl-6,7-difluoro-1,4-di- hydro-4-oxo-3-quinolinecarboxylic acid (0.6 g), anhydrous acetonitrile (6 ml), 3-aminopyrrolidine (0.35 g) and DBU (0.31 g) was refluxed for an hour. Then, 3-aminopyrrolidine (0.2 g) was more added and further refluxed for 2 hours. After cooling, the resulting precipitate was collected by filtration, dissolved in water (9 ml) containing sodium hydroxide (0.12 g) and neutralized with acetic acid. The resulting precipitate was collected by filtration and washed with water and acetonitrile successively to give the title compound (0.52 g) as colorless powder, mp 237-238 °C (decompd.).
-
Analysis (%) for C17H17ClFN3O3·H2O, Calcd. (Found): C, 53.20 (52.97); H, 4.99 (4.62); N, 10.95 (10.83).
- Example 28 7-(3-Amino-1-pyrrolidinyl)-8-chloro-1-cyclopropyl-6-fluoro-l,4-dihydro-4-oxo-3-quinolinecarboxylic acid
Example 29 7-(3-Amino-1-pyrrolidinyl)-8-chloro-1-cyclopropyl-6-fluoro-l,4-dihydro-4-oxo-3-quinolinecarboxylic acid hydrochloride
-
To a suspension of 7-(3-amino-1-pyrrolidinyl)-8-chloro-1-cyclopropyl-6-fluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid (100 mg) in ethanol (2 ml) was added 0.2 ml of ethanol solution of hydrogen chloride (7.0 mmol HC1/ml) and then the mixture was concentrated. The resulting residue was recrystallized from methanol to give the title compound (79 mg) as light yellow prisms, mp 263-265 °C (decompd.).
-
Analysis (%) for C17H17ClFN3O3.HCl, Calcd. (Found): C, 50.76 (50.50); H, 4.51 (4.44); N, 10.45 (10.38).
…………………..
J. Med. Chem., 23, 1358 (1980)
-
The compound wherein R3 is hydrogen, namely 3-pyrrolidinemethanamine, has been reported in J. Org. Chem., 26, 4955 (1961).
References
- Rubinstein, E. (2001). “History of quinolones and their side effects.”. Chemotherapy. 47 Suppl 3: 3–8; discussion 44–8.doi:10.1159/000057838. PMID 11549783.
| EP0106489A2 * | Sep 6, 1983 | Apr 25, 1984 | Warner-Lambert Company | Antibacterial agents |
| EP0153163A2 * | Feb 15, 1985 | Aug 28, 1985 | Warner-Lambert Company | 7-Substituted-1-cyclopropyl-6,8-difluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acids; 7-substituted-1-cyclopropyl-1,4-dihydro-6-fluoro-4-oxo-1,8-naphthyridine-3-carboxylic acids; their derivatives; and a process for preparing the compounds |
| BE899399A1 * | Title not available | |||
| GB2057440A * | Title not available |
| Examples of | ||
| reported trade | ||
| names for products | ||
| containing the 6- | ||
| 6-Fluoroquinolin- | fluoroquinolin- | |
| 4(1H)-one | 4(1H)-one | Structure |
| amifloxacin | ||
| balofloxacin | ||
| ciprofloxacin | Cipro®, Ciprobay, & Ciproxin | |
| clinafloxacin | ||
| danofloxacin | Advocin & Advocid | |
| difloxacin | Dicural® & Vetequinon | |
| enrofloxacin | Baytril® | |
| fleroxacin | Megalone | |
| flumequine | Flubactin | |
| garenoxacin | ||
| gatifloxacin | Tequin® & Zymar® | |
| grepafloxacin | Raxar | |
| ibafloxacin | ||
| levofloxacin | Levaquin®, Gatigol, Tavanic, Lebact, Levox, & Cravit | |
| lomefloxacin | Maxaquin® | |
| marbofloxacin | Marbocyl® & Zenequin | |
| moxifloxacin | Avelox® & Vigamox® | |
| nadifloxacin | Acuatin, Nadoxia, & Nadixa | |
| norfloxacin | Noroxin®, Lexinor, Quinabic, & Janacin | |
| ofloxacin | Floxin®, Oxaldin, & Tarivid | |
| orbifloxacin | Orbax® & Victas | |
| pazufloxacin | ||
| pefloxacin | ||
| pradofloxacin | ||
| prulifloxacin | ||
| rufloxacin | Uroflox | |
| sarafloxacin | Floxasol, Saraflox, Sarafin | |
| sitafloxacin | ||
| sparfloxacin | Zagam | |
| temalioxacin | Omniflox | |
| enoxacin | Penetrex & Enroxil | |
| gemifloxacin | Factive | |
| tosufloxacin | ||
| trovafloxacin | Trovan | |
PRULIFLOXACIN by Nippon Shinyaku Co.
![]()
PRULIFLOXACIN
(RS)-6-Fluoro-1-methyl-7-[4-(5-methyl-2-oxo-1,3-dioxolen-4-yl)methyl-1-piperazinyl]-4-oxo-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylic acid
6-Fluoro-1-methyl-7-(4-(5-methyl-2-oxo-1,3-dioxelen-4-yl)methyl-1-piperazinyl)-4-oxo-4H-(1,3)thiazeto(3,2-a)quinoline-3-carboxylic acid
123447-62-1 CAS NO
OPT-99
Launched – 2002 BY NIPPON SHINYAKU
SYNTHESIS…….http://www.drugfuture.com/synth/syndata.aspx?ID=151640
Prulifloxacin is an older synthetic chemotherapeutic antibiotic of the fluoroquinolone drug class[1][2] undergoing clinical trials prior to a possible NDA (New Drug Application) submission to the U.S. Food and Drug Administration (FDA). It is a prodrug which is metabolized in the body to the active compound ulifloxacin.[3][4] It was developed over two decades ago by Nippon Shinyaku Co. and was patented in Japan in 1987 and in the United States in 1989.[5][6]
It has been approved for the treatment of uncomplicated and complicated urinary tract infections, community-acquired respiratory tract infections in Italy and gastroenteritis, including infectious diarrheas, in Japan.[3][7] Prulifloxacin has not been approved for use in the United States.
Prulifloxacin is a novel fluoroquinolone antibiotic that was launched pursuant to a collaboration between Meiji Seika and Nippon Shinyaku in 2002 for the oral treatment of systemic bacterial infections, including acute upper respiratory tract infection, bacterial pneumonia, prostatitis, cholecystitis, bacterial enteritis, internal genital infections, otitis media, sinusitis and others. It is currently marketed in a tablet formulation. A once-daily formulation to be taken over a three-day period is in phase III clinical trials at Optimer Pharmaceuticals to be used in the treatment of bacterial gastroenteritis, including traveler’s diarrhea. The formulation had been in phase II trials at the company for the treatment of urinary tract infections, however, no recent development for this indication have been reported. The drug has also been studied at Optimer for the treatment of community-acquired respiratory tract infections, but recent progress reports for this indication have not been made available.
Prulifloxacin has in vitro activity against a wide range of gram-negative and gram-positive microorganisms. Its antibacterial action results from inhibition of DNA gyrase and topoisomerase IV, both Type II isomerases. DNA gyrase is an essential enzyme that is involved in the replication, transcription, and repair of bacterial DNA. Topoisomerase IV is an enzyme known to play a key role in the partitioning of the chromosomal DNA during bacterial cell division. Together, the Type II topoisomerases remove the positive supercoils that accumulate ahead of a translocating DNA polymerase, allowing DNA replication to continue unhindered by topological strain. Fluoroquinolones may be active against pathogens that are resistant to penicillins, cephalosporins, aminoglycosides, macrolides and tetracyclines, as they possess a distinct mechanism of action from these antibiotics.
Prulifloxacin was discovered by Nippon Shinyaku and codeveloped with Meiji Seika in Japan. Nippon Shinyaku granted Angelini a manufacturing and marketing license for Italy in 1993. Exclusive Korean manufacturing and commercialization rights were acquired by Yuhan from Nippon Shinyaku in March 2003. In June 2004, Optimer was granted exclusive development and commercialization rights to prulifloxacin in the U.S. from Nippon Shinyaku. Finally, Recordati signed a nonexclusive licensing agreement with Angelini for the marketing and sale of prulifloxacin in Spain in October 2004. In March 2009, the product was licensed to Lee’s Pharmaceuticals by Nippon Shinyaku for marketing in China as an oral treatment of bacterial infection. In 2010, prulifloxacin was licensed to Algorithm by Nippon Shinyaku in North Africa and the Middle East for the development and marketing for the treatment of bacterial infections.
History
In 1987 a European Patent (EP 315828) for prulifloxacin (Quisnon ) was issued to the Japanese based pharmaceutical company, Nippon Shinyaku Co., Ltd (Nippon). Ten years after the issuance of the European patent, marketing approval was applied for and granted in Japan (March 1997). Subsequent to being approved by the Japanese authorities in 1997 prulifloxacin (Quisnon) was co-marketed and jointly developed in Japan with Meiji Seika as licensee (Sword).[6]
In more recent times, Angelini ACRAF SpA, under license from Nippon Shinyaku, has fully developed prulifloxacin, for the European market.[8] Angelini is the licensee for the product in Italy. Following its launch in Italy, Angelini launched prulifloxacin in Portugal (January 2007) and it has been stated that further approvals will be sought in other European countries.[9][10]
Prulifloxacin is marketed in Japan and Italy as Quisnon (Nippon Shinyaku); Sword (Meiji); Unidrox (Angelini) and generic as Pruquin.
In 1989 and 1992 United States patents (US 5086049) were issued to Nippon Shinyaku for prulifloxacin. It was not until June 2004, when Optimer Pharmaceuticals acquired exclusive rights to discover, develop and commercialize prulifloxacin (Pruvel) in the U.S. from Nippon Shinyaku Co., Ltd., that there were any attempts to seek FDA approval to market the drug in the United States. Optimer Pharmaceuticals expects to file an NDA (new drug application) for prulifloxacin some time in 2010. As the patent for prulifloxacin has already expired, Optimer Pharmaceuticals has stated that this may have an effect on the commercial prospects of prulifloxacin within the United States market.[11]
Licensed uses
Prulifloxacin has been approved in Italy ,Japan,China,India and Greece (as indicated), for treatment of infections caused by susceptible bacteria, in the following conditions:
- Italy
- Acute uncomplicated lower urinary tract infections (simple cystitis)
- Complicated lower urinary tract infections
- Acute exacerbation of chronic bronchitis
- Japan
- Gastroenteritis, including infectious diarrheas
- Other countries
- Prulifloxacin has not been approved for use in the United States, but may have been approved in other Countries, other than that which is indicated above.
Availability
Prulifloxacin is available as:
- Tablets (250 mg, 450 mg or 600 mg)
In most countries, all formulations require a prescription.
Prulifloxacin is chemically known as 6-fluoro-1-methyl-7-{4-[(5-methyl-2-oxo-1 ,3-dioxol- 4-yl)methyl]piperazin-1-yl}-4-oxo-4H-[1 ,3]-thiazeto-[3,2-a]-quinoline-3-carboxylic acid, and it has the structure as shown below as formula I:
FORMULA I
Prulifloxacin has significant antibacterial activity and has been marketed as a synthetic antibacterial agent.
Prulifloxacin was first disclosed in US 5,086,049. The patent discloses a process for the preparation of prulifloxacin by the condensation of ulifloxacin with a 4-halomethyl-5- methyl-1 ,3-dioxolen-2-one of formula III
wherein X is halo selected form chloro, bromo or iodo, in the presence or absence of an aprotic solvent and a base to obtain prulifloxacin free base which is recrytallised with chloroform-methanol. In an exemplified process, ethyl 6,7-difluoro-1-methyl-4-oxo-4H- (1 ,3)-thiazeto-(3,2-a)-quinoline-3-carboxylate is condensed with piperazine in the presence of dimethyl formamide and purified by column chromatography followed by basic hydrolysis to give ulifloxacin, which is then converted to prulifloxacin.
The above process involves column chromatography. Prulifloxacin prepared by this method has a purity of 60-65% containing impurities in unacceptable levels. Removal of these impurities by usual purification procedures, such as recrystallisation, distillation and washing, is difficult and requires extensive and expensive multiple purification processes. This further decreases the overall yield. A method involving column chromatographic purifications and multiple purifications cannot be used for large-scale operations, thereby making the process commercially non-viable.
European Patent No. 315828 disclosed a variety of quinoline carboxylic acid derivatives and pharmaceutically acceptable salts thereof. These compounds are exhibiting antibacterial activity and useful as remedies for various infectious diseases. Among them prulifloxacin, chemically (+)-6-Fluoro- 1 -methyl-7-[4-(5-methyl-2-oxo-1 ,3-dioxolen-4-ylmethyl)-1 -piperazinyl]-4-oxo-4H- [1 ,3]thiazeto[3,2-a]quinoline-3-carboxylic acid is a fluoroquinolone antibacterial prodrug which shows potent and broad-spectrum antibacterial activity both in vitro and in vivo. Prulifloxacin also showed superior activity against strains of Enterobacteriaceae and Pseudomonas aeruginosa. Prulifloxacin is represented by the following structure:
Processes for the preparation of prulifloxacin and related compounds were disclosed in European Patent No. 315828 and UK Patent Application No. GB 2190376.
In – the preparation of prulifloxacin, 6-fluoro-1-methyl-4-oxo-7-(1- piperazinyl)-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylic acid of formula I:
is a key intermediate. According to the UK Patent Application No. GB 2190376, the compound of the formula I was prepared by the reaction of 3,4-difluroaniline with carbon disulfide and triethylamine to give triethylammonium N-(3,4- difluorophenyl)dithio carbamate, which by reaction with ethyl chloroformate and triethylamine in chloroform is converted into 3,4-difluorophenyl isothiocyanate, followed by reaction with diethyl malonate and KOH in dioxane affords the potassium salt, which is then treated with methoxymethyl chloride in dimethylformamide to give diethyl 1-(3,4-difluorophenylamino)-1- (methoxymethylthio)-rnethylene-rnalonate. The cyclization of the thio compound at 2400C in diphenyl ether affords ethyl 6,7-difluoro-4-hydroxy-2- methoxymethylthioquinoline-3-carboxylate, which by treatment with HCI in ethanol gives ethyl δy-difluoro^-hydroxy^-mercaptoquinoline-S-carboxylate. The cyclization of the mercapto compound with 1,1-dibromoethane by means of potassium carbonate and potassium iodide in hot dimethylformamide yields ethyl 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate, which is condensed with piperazine in dimethylformamide to afford ethyl 6- fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto[3,2-a]quinoline-3- carboxylate, which is then subjected to hydrolysis with potassium hydroxide in hot tert-butanol to give the compound of formula I.
The compound of formula I obtained by the process described in the UK Patent Application No. GB 2190376 is not satisfactory from purity point of view, the reaction between ethyl 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2- a]quinoline-3-carboxylate and piperazine requires longer time about 48 hours to complete, the yield obtained is not satisfactory, and the process also involves column chromatographic purifications. Methods involving column chromatographic purifications cannot be used for large-scale operations, thereby making the process commercially not viable. According to the European Patent No. 315828, prulifloxacin is prepared by reacting 6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto[3,2-a] quinoline-3-carboxylic acid with 4-bromomethyl-5-methyl-1 ,3-dioxolen-2-one in presence of potassium bicarbonate in dimethylformamide. However, a need still remains for an improved and commercially viable process of preparing pure prulifloxacin that will solve the aforesaid problems associated with process described in the prior art and will be suitable for large- scale preparation, in terms of simplicity, purity and yield of the product.
Prulifloxacin is chemically 6-fluoro-l-methyl-7-{4-[(5-methyl-2-oxo-l,3-dioxol-4- yl)methyl]piperazin-l-yl}-4-oxo-4H-[l,3]thiazeto[3,2-α]quinoline-3-carboxylic acid of Formula I having the structure as depicted below:
FORMULA I
Prulifloxacin has significant antibacterial activity and has been marketed as a synthetic antibacterial agent. U.S. Patent No. 5,086,049 provides a process for the preparation of prulifloxacin by reacting 6-fluoro-l-methyl-4-oxo-7-piperazin-l-yl-4H- [l,3]thiazeto[3,2-α]quinoline-3-carboxylic acid of Formula II,
FORMULA II and 4-(bromomethyl)-5-methyl-l,3-dioxol-2-one of Formula III,
FORMULA III using N,N-dimethylformamide as a solvent. 4-(Bromomethyl)-5-methyl-l,3-dioxol-2-one of Formula III is used in excess to one mole of the compound of Formula II. The process provided in U.S. Patent No. 5,086,049 further involves concentrating the reaction mixture, pouring the residue into water and isolating prulifloxacin by filtration. The resulting prulifloxacin is recrystallized from chloroform-methanol.
However, U.S. Patent No. 5,086,049 does not provide any method to remove the unreacted or the excess of 4-(bromomethyl)-5-methyl-l,3-dioxol-2-one of Formula III used as a starting material. The present inventors have observed that it is difficult to obtain prulifloxacin with pharmaceutically acceptable purity by following the process provided in U.S. Patent No. 5,086,049, which is typically contaminated by process related impurities including 4-(bromomethyl)-5-methyl-l,3-dioxol-2-one
A need still remains for an improved and commercially-viable process for preparing pure prulifloxacin that will solve the aforesaid problems associated with the process described in the prior art and that will be suitable for large-scale preparation, in terms of simplicity, purity and yield of the product.
EP1626051 A1 mentions that Type I, Type II and Type III crystals of prulifloxacin are obtained by crystallization from acetonitrile as reported in lyakuhin Kenkyu, Vol. 28 (1), (1997), 1-11. However, the conditions of crystallization from acetonitrile for preparing Type I, Type II and Type III crystals are not disclosed in lyakuhin Kenkyu, Vol. 28 (1), (1997), 1-11. EP1626051A1 further mentions that Type III crystals have been marketed by considering the solubility, absorbability, therapeutic effect and the like of the respective crystal forms.
US 2007/0149540 discloses a crystal of prulifloxacin acetonitrile solvate (Compound B) which is an intermediate for producing preferentially the type III crystal of prulifloxacin. A crystal of Compound B can be preferentially precipitated by controlling the supersaturation concentration in crystallization using acetonitrile as a solvent, subsequently; the type III crystal of Compound A can be produced by performing desolvation of the crystal.
WO 2008/111018 discloses processes for the preparation of Type I, Type II and Type III crystals of prulifloxacin. There is disclosed a process for preparing Type I crystals by controlled cooling over a period of 7 to 9 hours and prolonged drying over 24 hours. The inventors of the present invention have found that Type I and Type III crystals prepared according to the WO 2008/111018 process are unstable and the process is non-reproducible.
WO 2010/0084508 discloses processes for the preparation of Type I, Type II and Type III crystals of prulifloxacin.
WO 2008/059512 discloses a process for the preparation of prulifloxacin using novel intermediates.
WO 2008/111016 discloses a process for the preparation of prulifloxacin having purity of about 99% or above. It would be a significant contribution to the art to provide a crystalline form of prulifloxacin, which is consistent and to provide industrially viable methods of preparation, pharmaceutical formulations, and methods of use thereof.

…………………
SYNTHESIS
http://www.google.com/patents/WO2012001357A1?cl=en
Scheme 1.
Formula I
[PRULIFLOXACIN]
Example 1
Preparation of ethyl-6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-(1 ,3)-thiazeto-[3,2-a]- quinoline-3-carboxylate (formula III)
5,6-difluoro-1-methyl-4-oxo-4H-[1 ,3]-thiazeto-[3,2-a]-quinoline-3-carboxylic acid ethyl ester of formula (II) (100 gms, 0.321 moles) was stirred in 500 ml of DMF at room temperature. Piperazine (76 gms, 0.882 moles) was added at room temperature and stirred for 10 minutes. The temperature was slowly raised to 50-55°C and the reaction mass was stirred at 50-55°C for 5 hours. After completion of the reaction, the reaction mass was cooled to 25-30°C and stirred for 2 hours. The reaction mass was further chilled to 10-15°C and stirred for 2 hours. The precipitated solid was filtered, washed of chilled DMF (2 x 50 ml). The solid was slurry washed with water (300 ml), filtered, washed with water ( 2 x 100 ml) and dried under vacuum at 70-75°C to yield the title compound [90 gms, 74 % yield, 95% HPLC purity].
Example 2
Preparation of 6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]-thiazeto-[3,2-a]- quinoline-3-carboxylic acid (formula IV)
Ethyl-6-fluoro-1 -methyl-4-oxo-7-(1 -piperazinyl)-4H-(1 ,3)-thiazeto-[3,2-a]-quinoline-3- carboxylate (100 gms, 0.265 moles) was stirred in water (600 ml) at 25-30°C. To this potassium hydroxide solution (50 gms of potassium hydroxide flakes is dissolved in 200 ml of water) was added and the reaction mass was heated to 80-85°C. The contents were stirred for 1 hour and after completion of reaction, the reaction mass was cooled to 25-30°C. The pH of the reaction mass was adjusted to 6.5-7.0 using 1:1 aqueous acetic acid solution. The contents were stirred at room temperature for 1 hour. The precipitated solid was filtered, washed with water (2 x 100 ml). The solid was slurried in methanol (300 ml) for 1 hour at 25-30°C, filtered, washed with methanol (2 x 50 ml) and dried under vacuum at 70-75°C to yield the title compound [90 gms, 97% yield, 96% HPLC purity]. Example 3
Preparation of prulifloxacin
To a solution of 4-(chloromethyl)-5-methyl-1,3-dioxol-2-one (55 gms, 0.371 moles) in 50 ml of DMF at 25-30°C, sodium bromide (77 gms, 0.748 moles) was added and the reaction mass was slowly heated to 40-45°C. The contents were stirred at 40-45°C for 2 hours, acetone ( 500 ml) was added at 40-45°C and stirred for 3 hours. The reaction mass was filtered over hyflo, and the bed washed with acetone (100 ml). The solvent was completely distilled off under vacuum below 45°C to yield 4-(bromomethyl)-5- methyl-1 ,3-dioxol-2-one (formula V).
To a solution of 6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]-thiazeto-[3,2-a]- quinoline-3-carboxylic acid of formula IV (100 gms, 0.286 moles) in 4.0 It of acetonitrile, DIPEA (70 ml , 0.402 moles)) was added at room temperature, stirred for 10 minutes. The reaction mass was cooled to 10-15°C and a solution of 4-(bromomethyl)-5-methyl- 1 ,3-dioxol-2-one (formula V) in 500 ml of acetonitrile was slowly added at 10-15°C over a period of 1 hour. The contents were stirred at 25-30°C for 20 hour, filtered over hyflo, and the bed washed with 200 ml of acetonitrile. The solvent was distilled off completely under vacuum below 50°C. Acetonitrile (100 ml) was added at 50°C and the contents were stirred for 30-60 minutes. The reaction mass was slowly chilled to 0-5°C and the precipitated solid was filtered, washed with acetonitrile (25 ml) and dried to yield 65 gms of prulifloxacin. Example 4
Preparation of Type I crystals of prulifloxacin
Prulifloxacin (65 gms) was added to 200 ml of DMF at 25-30°C and heated to 80-85°C for 1 hour. The mixture was then slowly cooled to 25-30°C, stirred for 2 hours, chilled to 0-5°C for 2 hours. The precipitated solid was filtered and dried under vacuum at 70- 75°C to yield Type I crystals of prulifloxacin (55 gms, 99.5 % HPLC purity).
Example 5
Preparation of prulifloxacin
(55 gms, 0.371 moles) of 4-(chloromethyl)-5-methyl-1 ,3-dioxol-2-one is taken in 5.0 ml of DMF at 25-30°C. (77 gms, 0.748 moles) of sodium bromide is added and slowly heated the reaction mass to 40-45°C. The contents are stirred at 40-45°C for 2 hours, 500 ml of acetone is added at 40-45°C and stirred for 3 hours. The reaction mass is clarified over hyflo, and the bed washed with 100 ml of acetone to yield a solution of 4- (bromomethyl)-5-methyl-1 ,3-dioxol-2-one (formula V).
To a solution of 6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]-thiazeto-[3,2-a]- quinoline-3-carboxylic acid of formula IV (100 gms, 0.286 moles) in 3.5 Its of acetone was at room temperature DIPEA (70 ml, 0.402 moles) and stirred for 10 minutes. The reaction mass was cooled to 10-15°C and a solution of 4-(bromomethyl)-5-methyl-1 ,3- dioxol-2-one (formula V) in acetone was slowly added to the reaction mass at 10-15°C over a period of 1 hour. The contents were further stirred at 25-30°C for 20 hour, filtered over hyflo and the bed washed with 200 ml of acetone. The solvent was distilled off completely under vacuum below 50°C. Acetonitrile (100 ml) was added at 50°C and the contents were stirred for 30-60 minutes. The reaction mass was further chilled to 0- 5°C and stirred for 2 hours. The precipitated solid was filteredand dried to yield prulifloxacin.
………………….
http://www.google.com/patents/WO2008059512A1?cl=en
novel process for preparing 6-fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto [3,2-a]quinoline-3-carboxylic acid of formula I:
which comprises: a) reacting the difluoro-quinoline compound of formula
wherein R represents hydrogen atom or alkyl containing 1 to 4 carbon atoms; with boric acid of formula III:
in presence of acetic anhydride and acetic acid to give borane compound of formula IV:
b) reacting the borane compound of formula IV with piperazine of formula V:
HN NH V
to give piperazine compound of formula Vl:
c) treating the compound of formula Vl with an alkaline metal hydroxide, carbonate or bicarbonate to obtain the compound of formula I.
Prulifloxacin and pharmaceutically acceptable acid addition salts of prulifloxacin can be prepared by using the compound of formula I by known methods for example as described in the European Patent No. 315828. Borane compound of the formula IV and Vl are novel and forms part of the invention. Preferably the reaction in step (a) is carried out at about 300C to reflux temperature more preferably at about 800C to reflux temperature and still more preferably at reflux temperature.
Example 1 Step-I:
Acetic anhydride (24 ml) and acetic acid (11 ml) are added to boric acid (3.5 gm) under stirring at 25 – 300C, the contents are heated to reflux and then stirred for 3 hours at reflux. The reaction mass is cooled to 1000C, ethyl 6,7- difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate (20 gm) is added at 1000C, the contents are heated to reflux and then refluxed for 2 hours. The reaction mass is cooled to 25 – 350C, toluene (200 ml) is added under stirring, the reaction mass is cooled to 50C and then stirred for 1 hour at 5 – 100C. Filtered the solid, washed with 20 ml of toluene and then dried to give 25.5 gm of 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate -O3,04/bis/acetato-0/-borone. Step-I I: Acetonitrile (125 ml), dimethylsulfoxide (125 ml) and piperazine (13.8 gm) are added to 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3- carboxylate-03,04/bis/acetato-0/-borone (25.5 gm, obtained in step-l) under stirring at 25 – 350C, the contents are heated to 850C and then stirred for 3 hours at 80 – 850C to form a clear solution. The solution is cooled to 100C and then stirred for 1 hour at 10 – 150C. Filtered the solid, washed with 25 ml of acetonitrile and then dried to give 26 gm of 6-fluoro-1-methyl-4-oxo-7-(1- piperazinyl)-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylate-03,04/bis/acetato-0/- borone. Step-Ill: Water (155 ml), potassium hydroxide (17 gm) are added to 6-fluoro-1- methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate- O3,O4/bis/ acetato-0/-borone (26 gm, obtained in step-ll) under stirring at 25 – 350C, the contents are heated to 650C and then stirred for 4 hours at 60 – 650C. The reaction mass is cooled to 250C, filtered the undesired solid on hi-flow bed and then pH of the resulting filtrate is adjusted to 7 – 7.5 with 50% HCI solution at 25 – 300C. The separated solid is stirred for 1 hour at 25 – 300C, filtered the solid, washed with 35 ml of water and then dried to give 17 gm of 6-fluoro-1- methyl-4-oxo-7-(1 -piperazinyl)-4H-[1 ,3]thiazeto [3,2-a]quinoline-3-carboxylic acid (HPLC Purity: 98.5%). Example 2 Step-I:
Acetic anhydride (12 ml) and acetic acid (5.5 ml) are added to boric acid (1.25 gm) under stirring at 25 – 300C, the contents are heated to reflux and then stirred for 3 hours at reflux. The reaction mass is cooled to 1000C, 6,7-difluoro-1- methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylic acid (10 gm) is added at 1000C, the contents are heated to reflux and then refluxed for 3 hours. The reaction mass is cooled to 500C, toluene (100 ml) is added under stirring at 500C, the resulting mass is cooled to 100C and then stirred for 1 hour at 10 – 150C. Filtered the solid, washed with 20 ml of toluene and then dried to give 10 gm of 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate -O3 , 04/bis/acetato-0/-borone . Step-I I:
Acetonitrile (50 ml), dimethylsulfoxide (50 ml) and piperazine (5.5 gm) are added to 6,7-difluoro-1-methyl-4-oxo-4H-[1 ,3]thiazeto[3,2-a]quinoline-3- carboxylate-03,04/bis/acetato-0/-borone (10 gm, obtained in step-l) under stirring at 25 – 350C, the contents are heated to 850C and then stirred for 3 hours at 80 – 850C to form a clear solution. The solution is cooled to 100C and then stirred for 1 hour at 10 – 150C. Filtered the solid, washed with 10 ml of acetonitrile and then dried to give 10.4 gm of 6-fluoro-1-methyl-4-oxo-7-(1- piperazinyl)-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate-03,04/bis/acetato-0/- borone. Step-Ill :
Water (62 ml), potassium hydroxide (7 gm) are added to 6-fluoro-1-methyl-4- oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto[3,2-a]quinoline-3-carboxylate-03,04/bis/ acetato-OAborone (10.4 gm, obtained in step-ll) under stirring at 25 – 350C, the contents are heated to 650C and then stirred for 4 hours at 60 – 650C. The reaction mass is cooled to 250C, filtered the undesired solid on hi-flow bed and then pH of the resulting filtrate is adjusted to 7 – 7.5 with 50% HCI solution at 25 – 300C. The separated solid is stirred for 30 minutes at 25 – 300C, filtered the solid, washed with 20 ml of water and then dried to give 68 gm of 6-fluoro-1- methyl-4-oxo-7-(1-piperazinyl)-4H-[1 ,3]thiazeto [3,2-a]quinoline-3-carboxylic acid (HPLC Purity: 98.6%). Example 3
Acetonitrile (560 ml) and potassium bicarbonate (8 gm) are added to 6- fluoro-i-methyM-oxo-y-CI-piperazinyO^H-CI .SKhiazetofS^-alquinoline-S- carboxylic acid (14 gm, obtained as per the processes described in examples 1 and 2) under stirring at 25 – 300C, the contents are cooled to 150C and then the solution of 4-bromomethyl-5-methyl-1 ,3-dioxolen-2-one (10 gm) in acetonitrile (140 ml) is added at 15 – 200C for 30 to 45 minutes. The contents are stirred for 25 hours at 25 to 300C, filtered and the resulting filtrate is distilled under vacuum. To the residue added acetonitrile (70 ml), cooled the mass to 200C and then stirred for 1 hour to 1 hour 30 minutes at 20 – 250C. Filtered the solid, washed the solid with 15 ml of chilled acetonitrile and then dried to give 16 gm of prulifloxacin crude (HPLC Purity: 98.8%).
To the prulifloxacin crude (obtained above) added acetonitrile (200 ml) at 25 – 300C, the contents are heated to reflux and then refluxed for 30 minutes. To the reaction mass added activated carbon (5 gm) and refluxed for 15 minutes. The reaction mass is filtered on hi-flo bed, the resulting filtrate is cooled to 200C and then stirred for 3 – 4 hours at 20 – 250C. Filtered the solid, washed with 20 ml of acetonitrile and then dried to give 14 gm of prulifloxacin (HPLC Purity: 99.9%).
…………………
http://www.google.com/patents/WO2008111016A1?cl=en
In a first aspect, a process for the preparation of prulifloxacin is provided, the process comprising: a) reacting a compound of Formula II with a compound of Formula III to obtain prulifloxacin;
FORMULA III
FORMULA II
b) contacting the prulifloxacin obtained in step a) with an acid in a biphasic solvent system, wherein the biphasic solvent system comprises water and a water- immiscible organic solvent; c) separating the aqueous layer from the reaction mixture obtained in step b); d) treating the aqueous layer with a base; and e) isolating prulifloxacin.
The process described in steps b – e above may be carried out with prulifloxacin made from any process however.
The compounds of Formula II and Formula III may be prepared according to the methods provided in U.S. Patent No. 5,086,049.
Example 1: Process for the Preparation of Prulifloxacin:
Step A): A solution of 4-(bromomethyl)-5-methyl-l,3-dioxol-2-one (35.5 g, 0.184 mole) in N,N-dimethylformamide (200 ml) was added dropwise at 0 to 5° C to a stirred solution of 6-fluoro-l-methyl-4-oxo-7-piperazin-l-yl-4H-[l,3]thiazeto[3,2-α]quinoline-3- carboxylic acid (50 g, 0.143 mole and potassium bicarbonate (15.8 g, 0.1578 mole) in N,N-dimethylformamide (200 ml). The resulting mixture was stirred at 25° to 28°C for 3 to 4 hours. After the completion of the reaction, the reaction mixture was poured into water (1250 ml). The solid obtained was filtered, washed with water (100 ml), and subsequently dissolved in a mixture of chloroform: methanol (7:3; 1250 ml). The lower organic layer was separated and water (500 ml) was added to the organic layer. A dilute aqueous solution of hydrochloric acid was added to the biphasic reaction mixture to adjust pΗ to 0.8 to 1.0. The reaction mixture was stirred for 15 minutes, allowed to settle and the upper aqueous layer was separated. The process was repeated twice and the aqueous layers were combined. Activated charcoal (10%) was added to the combined aqueous layer and stirred for 30 minutes, filtered and cooled to 20° to 25° C. The pΗ of the reaction mixture was adjusted to 6.5 to 7.0 by adding an aqueous solution of sodium bicarbonate. The solid obtained was extracted with chloroform (375 ml), stirred for 15 minutes and the organic layer was separated. The aqueous layer was further extracted with a mixture of chloroform: methanol (7:3 ratio; 50 ml). The combined organic layer was distilled under vacuum at 35° to 40° C to recover the solvent up to 125 ml. The reaction mass so obtained was stirred for 3 to 4 hours at 28° to 30° C, filtered and washed with chilled chloroform (50 ml). The wet cake obtained was dried at 45° C for 12 hours to obtain the title compound. Step B): The prulifloxacin (30 g) obtained in Step A) was suspended in a mixture of chloroform: ethanol (10:1, v/v, 585 ml: 58.5 ml) and heated to reflux temperature. Activated carbon (3.9 gm) was added to the partially cleared solution and refluxed for 30 minutes, followed by filtration through Celite bed. The bed was further washed with chloroform: ethanol (10:1, v/v, 585 ml: 58.5 ml). The filtrate so obtained was distilled at atmospheric pressure till to partially remove the solvent. The concentrate so obtained was stirred at about 25° C for 1 hour, and filtered. The solid obtained was washed with chloroform: ethanol (39 ml X 2), dried under vacuum at 45° C for 12 hours to obtain the title compound. Yield: 22 g
HPLC Purity: 99%
………………………….
SEE
Studies on pyridonecarboxylic acids. 1. Synthesis and antibacterial evaluation of 7-substituted-6-halo-4-oxo-4H-[1,3]thiazeto[3, 2-a]quinoline-3-carboxylic acids
J Med Chem 1992, 35(25): 4727
http://pubs.acs.org/doi/pdf/10.1021/jm00103a011

The reaction of 3,4-difluoroaniline (I) with carbon disulfide and triethylamine gives triethylammonium N-(3,4-difluorophenyl)dithiocarbamate (II), which by reaction with ethyl chloroformate and triethylamine in chloroform is converted into 3,4-difluorophenyl isothiocyanate (III). The reaction of (III) with diethyl malonate and KOH in dioxane affords the potassium salt (IV), which is treated with chloromethyl methyl ether in DMF to give the corresponding methoxymethylsulfanyl compound (V). The cyclization of (V) at 240 C in diphenyl ether affords 6,7-difluoro-4-hydroxy-2-(methoxymethylsulfanyl)quinoline-3-carboxylic acid ethyl ester (VI), which by treatment with HCl in ethanol gives the corresponding mercapto compound (VII). The cyclization of (VII) with 1,1-dibromoethane by means of K2CO3 and KI in hot DMF yields 5,6-difluoro-1-methyl-4-oxo-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylic acid ethyl ester (VIII), which is condensed with piperazine (IX) in DMF to afford the corresponding piperazino-derivative (X). The hydrolysis of (X) with KOH in hot tert-butanol gives the corresponding free acid (XI) , which is finally condensed with 4-(bromomethyl)-5-methyl-1,3-dioxol-2-one (XII) by means of KHCO3 in DMF.
………………….

Treatment of 3,4-difluoroaniline (I) with CS2 and Et3N gives triethylammonium dithiocarbamate (II), which reacts with ethyl chloroformate in chloroform to yield (III). Isothiocyanate (III) is converted into the potassium salt (IV) by reaction with diethyl malonate and KOH in dioxane and then transformed into methoxymethyl thioether (VI) by means of reagent (V) and Et3N in toluene. Cyclization of (VI) by heating in diphenyl ether affords quinoline (VII), which then reacts with benzoyl chloride (VIII) in pyridine to furnish (IX). Benzoyloxy derivative (IX) is converted into (X) by means of HCl in EtOH, and its reaction with 1-bromo-2-fluoroethane (XI) and NaHCO3 yields compound (XII). Chlorination of (XII) with SO2Cl2 in hexane provides (XIII), which by simultaneous hydrolysis and intramolecular cyclization by means of Et3N /H2O in THF provides the mixture of isomers (XIV). (+)-(XV) is obtained by HPLC chromatography of (XIV) on a chiral stationary phase. Treatment of (+)-(XV) with 1-methylpiperazine (XVI) in DMF provides ethyl ester (+)-(XVII), which is finally hydrolyzed by means of H2SO4 in H2O.
INTERMEDIATES

154330-67-3
Ethyl 6,7-difluoro-2-ethylmercapto-4-hydroxyquinoline-3-carboxylate

154330-68-4
Ethyl 4-acetoxy-6,7-difluoro-2-(ethylthio)quinoline-3-carboxylate

113046-72-3
Ethyl 6,7-difluoro-1-methyl-4-oxo-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylate

113028-17-4
Ethyl 6-fluoro-1-methyl-4-oxo-7-(1-piprazinyl)-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylate

112984-60-8
6-Fluoro-1-methyl-4-oxo-7-(1-piperazinyl)-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylic acid
REFERENCES
- Nelson, Jennifer M.; Chiller, Tom M.; Powers, John H.; Angulo, Frederick J. (2007). “Food Safety: Fluoroquinolone‐Resistant Campylobacter Species and the Withdrawal of Fluoroquinolones from Use in Poultry: A Public Health Success Story”. Clinical Infectious Diseases 44 (7): 977–80. doi:10.1086/512369. PMID 17342653.
- Kawahara S (1998). “[Chemotherapeutic agents under study]”. Nippon Rinsho (in Japanese) 56 (12): 3096–9. PMID 9883617.
- Fritsche, T. R.; Biedenbach, D. J.; Jones, R. N. (2008). “Antimicrobial Activity of Prulifloxacin Tested against a Worldwide Collection of Gastroenteritis-Producing Pathogens, Including Those Causing Traveler’s Diarrhea”. Antimicrobial Agents and Chemotherapy 53 (3): 1221–4. doi:10.1128/AAC.01260-08. PMC 2650572.PMID 19114678.
- Giannarini, Gianluca; Tascini, Carlo; Selli, Cesare (2009). “Prulifloxacin: clinical studies of a broad-spectrum quinolone agent”. Future Microbiology 4 (1): 13–24.doi:10.2217/17460913.4.1.13. PMID 19207096.
- JP patent 1294680, Kise Masahiro; Kitano Masahiko; Ozaki Masakuni; Kazuno Kenji; Matsuda Masato; Shirahase Ichiro; Segawa Jun, “Quinolinecarboxylic Acid Derivative”, issued November 28, 1989
- Prulifloxacin. Drugfuture.com. Retrieved on 2010-11-03.
- Anonymous (2002). “Prulifloxacin [‘Quisnon’; Nippon Shinyaku] has been approved in Japan”. Inpharma 1 (1362): 22.
- Research and Development Department of Angelini. Angelinipharma.com. Retrieved on 2010-11-03.
- Nippon Shinyaku, Annual Report 2007
- “Prulifloxacin. NAD-441A, NM 441, Quisnon”. Drugs in R&D 3 (6): 426–30. 2002.PMID 12516950.
- Annual Report 2008, p. 34
Segawa,J,Mashiko kitano, Kenji Kazuno et al, Studies on Pyridonecarboxylic acids,1.Sythesis and antibacterial Evaluation of 7-substituted-6-halo-4-oxo-4H-[1,3]thiazeto [3,2-]quionoline- 3-caroboxylic acids[J].J Med Chem. 1992,35(25):4727-4738.
Masato Matsuoka, Jun Segawa, Yoshihiko.et al, Studies on Pyridone Carb oxylic acids. V.A Practial synthesis of Ethyl 6,7–Difuoro-1-methyl-4-oxo-[1,3] Thiazeto [3,2-a]quinoline-3- Caroboxylate a key intermediate for the new tricyclic quinolone, prulifloxacin (NM441) and Versatile new syntheses of the 2-thioquinoline Skeleton[J].J Heterocyclic Chem.1997,34,1773-1779.
|
3-13-1996
|
Sustained release capsule
|
|
|
10-11-1995
|
Method of manufacturing solid dispersion
|
|
|
2-5-1992
|
7(4-(5 METHYL-2-OXO-1,3-DIOXALEN-4-YL)METHYL 1-PIPERZINYL)-4-OXO-4H-(1,3)THIAZETO(3,2-A)QUINOLINE-3-CARBOXYLIC ACIDS
|
|
6-31-2011
|
PHARMACEUTICAL COMPOSITION
|
|
|
2-11-2011
|
PROCESS FOR THE PREPARATION OF PURE PRULIFLOXACIN
|
|
|
8-6-2010
|
PROCESS FOR PREPARATION OF PRULIFLOXACIN USING NOVEL INTERMEDIATES
|
|
|
5-7-2010
|
PROCESS FOR THE PREPARATION OF CRYSTALS OF PRULIFLOXACIN
|
|
|
4-9-2010
|
COMPOSITION COMPRISING AN ANTIBIOTIC AND A CORTICOSTEROID
|
|
|
12-11-2009
|
Compounds and Methods for modulating the Silencing of a Polynucleotide of Interest
|
|
|
8-24-2007
|
PHARMACEUTICAL COMPOSITION
|
|
|
6-29-2007
|
PHARMACEUTICAL COMPOSITION
|
|
|
7-15-2005
|
Pharmaceutical composition
|
|
|
2-6-2004
|
Medicinal composition
|
| WO2008059512A1 | Nov 17, 2006 | May 22, 2008 | Hetero Drugs Ltd | Process for preparation of prulifloxacin using novel intermediates |
| WO2008111016A1 | Mar 14, 2008 | Sep 18, 2008 | Ranbaxy Lab Ltd | Process for the preparation of pure prulifloxacin |
| WO2008111018A2 | Mar 14, 2008 | Sep 18, 2008 | Ranbaxy Lab Ltd | Process for the preparation of crystals of prulifloxacin |
| WO2010084508A2 | Dec 10, 2009 | Jul 29, 2010 | Elder Pharmaceuticals Ltd. | Process for the preparation of type i, type ii and type iii crystalline prulifloxacin |
| EP0315828A1 * | Oct 26, 1988 | May 17, 1989 | Nippon Shinyaku Company, Limited | Quinolinecarboxylic acid derivatives |
| EP1626051A1 | Apr 28, 2004 | Feb 15, 2006 | Nippon Shinyaku Co., Ltd. | Crystals of quinolinecarboxylic acid derivative solvate |
| US5086049 | Apr 8, 1991 | Feb 4, 1992 | Nipponshinyaku Co., Ltd. | 7[4-(5 methyl-2-oxo-1,3-dioxalen-4-yl)methyl 1-piperzinyl]-4-oxo-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylic acids |
| US20070149540 | Apr 28, 2004 | Jun 28, 2007 | Nippon Shinyaky Co., Ltd. | Crystals of quinolinecarboxylic acid derivative solvate |
EXTRA INFO
http://www.google.com/patents/EP2524922A1?cl=en
-
formula 1 is S-(-)-6-fluoro-1-methyl-7-[4-(5-methyl-2-oxo-1,3-dioxolen-4-yl)methyl-1-piperazinyl]-4-oxo-4H-[1,3]thiazet o[3,2-α]quinoline-3-carboxylic acid (levo-prulifloxacin for short); its stereo configuration is S configuration; it has optical property of levorotatory polarized light:
-
S-(-) ulifloxacin (as shown in formula 2 below) as raw material and the compound as shown in the following formula 3 are reacted in organic solvent in the presence of alkaline material. The reaction formula is shown below:
-
S-(-)-ulifloxacin and R-(+)-ulifloxacin are prepared according to the method disclosed in CN101550142A .
-
Japanese scholars Masato Matsuoka et al. have proved the absolute configuration of optically pure prulifloxacin. The study (see the publication Chem. Pharm. Bull. 43(7) 1238-1240 (1995)) verifies that (-)-ulifloxacin is S configuration while (+)-ulifloxacin is the enantiomer of R configuration by applying chemical methods together with single-crystal X-ray diffraction.
-
Accordingly, R-prulifloxacin can be prepared from R-(+)-ulifloxacin and the compound of formula 3 by the method described hereinbefore.
-
[0022]
-
S-prulifloxacin prepared in accordance with the present invention is determined to be laevorotatory by optical rotation measurement, so it is S-(-)-prulifloxacin. R-prulifloxacin prepared in accordance with the present invention is determined to be dextrorotatory by optical rotation measurement, so it is R-(+)-prulifloxacin.
-
The present invention studied the absorption features of S-(-)-prulifloxacin and R-(+)-prulifloxacin on circular polarized light by circular dichroism spectroscopy. The two spectrograms are mirror images of each other, which proves that S-(-)-prulifloxacin and R-(+)-prulifloxacin are enantiomer of each other.
-
Comparing the circular dichroism spectrogram as depicted in figure 4 with the circular dichroism spectrogram of analogue of the similar structure with known absolute configuration as disclosed in the publication Chem. Pharm. Bull. 47(12) 1765-1773 (1999), it is found that (-)-prulifloxacin has similar Cotton effect to the two analogues reported in the publication, ethyl S-(-)-6,7-difluoro-1-methyl-4-oxo-4H-[1,3] thiazeto[3,2-α]quinoline-3-carboxylate and ethyl S-(-)-6, 7-difluoro-1-fluoromethyl-4-oxo-4H-[1,3]thiazeto[3,2-α]quinoline-3-carboxylate; so does (+)-prulifloxacin. The results also verify on the other hand that the absolute configuration of levo-prulifloxacin of the present invention is S type while the absolute configuration of dextro-prulifloxacin is R type.
-
The compound of the present invention and physiologically acceptable acid can be prepared to salts: dissolving or suspending S-(-)-prulifloxacin in solvent such as chloroform, DMF and the like; adding into acid or acid solution (for example, hydrochloric acid or hydrogen chloride-methanol solution and the like) while stirring; precipitating and filtering to obtain solid salt from the solvent solution, or alternatively removing solvent from the salt solution directly by concentration, spray drying and the like to obtain the salt of S-(-)-prulifloxacin. The obtained solid may be further recrystallized.
Example 1 Preparation of (S)-(-)-uliflourxacin
-
105 g of racemic uliflourxacin was dissolved in 1,500 mL of dimethyl sulfoxide. 27 g of D-tartaric acid was dissolved in 405 mL of dimethyl sulfoxide dropwise while stirring. After stirring at room temperature for 20 hours, the precipitate was filtrated. The collected solid was dried under vacuum to obtain 86 g solid, which was recrystallized in dimethyl sulfoxide to obtain 37 g of levoulifloxacin-D-tartrate, with C49.08%, H5.06%, N9.50%, S7.44% shown by elemental analysis (molecular formula: C16H16FN3O3S·1/2C4H6O6·H2O, calculated values: C48.86%, H4.78, N9.50%, S7.25%). Said salt was added into water to obtain a suspension, and the pH value was adjusted to 7-8 with 2% NaOH aqueous solution while stirring. After precipitation, filtration, and drying, 24.5 g of (S)-uliflourxacin was obtained, having a chemical name (S)-(-)-6-fluoro-1-methyl-4-oxo-(1-piperazinyl)-1H,4H-[1,3]thiazeto [3,2-α]quinoline-3-carboxylic acid.
-
Specific rotation [α]20 D= -133° (c=0.5, 0.1 mol/L methanesulfonic acid); 1H-NMR (DMSO-d6) δ2.11 (3H, d, j=6.2 Hz), 2.87 (4H, m), 3.19 (4H, m), 6.40 (1H, q, j=6.2 Hz), 6.89 (1H, d, j=7.4Hz), 7.79 (1H, d, j=13.9Hz), optical purity e.e. 96%.
Example 2 Preparation of (R)-(+)-uliflourxacin
-
105 g of racemic uliflourxacin was dissolved in 1,500 mL of DMSO. 27 g of L-tartaric acid was dissolved in 405 mL dimethyl sulfoxide dropwise while stirring to allow that the solution became turbid and the precipitation occurred. The solution was stirred at room temperature for 20 hours and then filtered. The collected solid was dried under vacuum to obtain 82 g solid which was recrystallized in dimethyl sulfoxide to obtain 34 g of dextrouliflourxacin-L-tartarte. Said salt was added into water to obtain a suspension, and the pH value was adjusted to 7-8 with 2% NaOH aqueous solution while stirring. After filtration and drying, 22 g of (R)-uliflourxacin was obtained, having a chemical name (R)-(+)-6-fluoro-1-methyl-4-oxo-(1-piperazinyl)-1H,4H-[1,3]thiazeto[3,2-a]quinoline -3-carboxylic acid.
-
Specific rotation [α]20 D= +132.4° (c=0.5, 0.1 mol/L methanesulfonic acid), optical purity e.e. 96%.
Example 3 Preparation of S-(-)-prulifloxacin
-
3.49 g (0.01 mol) of S-(-)-uliflourxacin prepared in Example 1, 2.02 g (0.02 mol) of triethylamine and 20 ml of dimethylformamide (hereinafter referred to as DMF) were mixed and stirred. After the solution was cooled to -5∼5 °C, 0.012 mol of 4-bromomethyl-5-methyl-1,3-dioxolen-2-one (hereinafter referred to as DMDO-Br) in DMF (5 ml) solution was added thereinto, followed by stirring at -5∼5 °C for 3 hours. The reaction solution was poured into 100 ml of ice water, stirred for 30 minutes, and then filtered. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 2.9 g of S-(-)-prulifloxacin was obtained, having a chemical name: S-(-)-6-fluoro-1-methyl-7-[4-(5-methyl-2-oxo-1,3-dioxolen-4-yl)methyl-1-piperazinyl ]-4-oxo-4H-[1,3]thiazeto[3,2-α]quinoline-3-carboxylic acid, with a purity of 98% and a yield rate of 63%. Specific rotation [α]20 D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid)
Example 4 Preparation of R-(+)-prulifloxacin
-
R-(+)-prulifloxacin prepared in Example 2 was used as raw material to prepare 2.7 g of target product R-(+)-prulifloxacin in accordance with the method as described in Example 3, with a yield rate of 60.7% and a purity of 98%. Specific rotation [α]20 D= +108° (c=0.5, 0.1 mol/L methanesulfonic acid).
-
Comparing the circular dichroism spectrogram as depicted in Figure 4 with the circular dichroism spectrogram of analogue of the similar structure with known absolute configuration as disclosed in the publication Chem. Pharm. Bull. 47(12) 1765-1773 (1999), it was found that (-)-prulifloxacin has similar Cotton effect with the two analogues reported in the publication, ethyl S-(-)-6,7-difluoro-1-methyl-4-oxo-4H-[1,3] thiazeto[3,2-α]quinoline-3-carboxylate and ethyl S-(-)-6, 7-difluoro-1-fluoromethyl-4-oxo-4H-[1,3]thiazeto[3,2-α]quinoline-3-carboxylate; so does (+)-prulifloxacin. The results also verify on the other hand that the absolute configuration of levo-prulifloxacin of the present invention is S type while the absolute configuration of dextro-prulifloxacin is R type.
Example 5 Preparation of S-(-)-prulifloxacin
-
3.49 g (0.01 mol) of S-(-)-uliflourxacin, 1.2 g (0.012 mol) of anhydrous potassium bicarbonate and 20 ml of dimethylsulfoxide were mixed and stirred. 0.012 mol of DMDO-Br in DMSO (5 mL) solution was added dropwise at -20 °C. Stirring proceeded at -20 °C for 3 hours. The reaction solution was poured into 100 ml of ice water, and the pH value was adjusted to 7 with 20% acetic acid. The solution was filtered after stirring for 30 minutes. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 2.5 g of the target product levo-prulifloxacin was obtained with a purity of 98% and a yield rate of 54%.
Specific rotation [α]20 D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid)
Example 6 Preparation of S-(-)-prulifloxacin
-
3.49 g (0.01 mol) of S-(-)-uliflourxacin, 1.04 g (0.008 mol) of N,N-diisopropylethylamine and 20 mL of N,N-dimethylformamide (DMF) was mixed and stirred, 0.008 mol of DMDO-Br in DMF (5 mL) solution was added thereinto. The solution was heating to 60 °C and reacted for 15 minutes. The reaction solution was poured into 100 ml of ice water, and the pH value was adjusted to 7 with 20% acetic acid. The solution was filtered after stirring for 30 minutes. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 2.0 g of the target product levo-prulifloxacin was obtained with a purity of 98% and a yield rate of 43%.
Specific rotation [α]20 D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid)
Example 7 Preparation of S-(-)-prulifloxacin
-
10 g (0.029 mol) of S-(-)-uliflourxacin, 30 ml of N,N-dimethylacetylamide and 14.7 g (0.145mol) of triethylamine was mixed and cooled to 5~10 °C. 8.5 g (0.03 mol) 4-(p-toluenesulfonic acid-1-methyl ester)-5-methyl-1,3-dioxolen-2-one in 25 ml of N,N-dimethylacetylamide solution was added dropwise while stirring. After addition, the solution was reacted at room temperature for 10 hours. The reaction solution was poured into 200 ml of ice water, and the pH value was adjusted to 7 with 20% acetic acid. The solution was filtered after stirring for 30 minutes. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 7.46 g of the target product levo-prulifloxacin was obtained with a purity of 98% and a yield rate of 57%. Specific rotation [α]20 D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid).
Example 8 Preparation of S-(-)-prulifloxacin
-
3.49 g (0.01 mol) of S-(-)-uliflourxacin, 0.79 g (0.05 mol) of potassium carbonate and 20 ml of dimethylformamide (DMF) was mixed and stirred. 0.012 mol of DMDO-Br in DMF (5ml) solution was added at -10 °C. At the same temperature, the solution was reacted for 2 hours. The reaction solution was poured into 100 ml of ice water, and the pH value was adjusted to 7 with 20% acetic acid. The solution was filtered after stirring for 30 minutes. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 2.2 g of the target product levo-prulifloxacin was obtained with a purity of 98% and a yield rate of 48%. Specific rotation [α]20 D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid).
Example 9 Preparation of S-(-)-prulifloxacin
-
3.49 g (0.01 mol) of S-(-)-uliflourxacin, 0.79 g (0.02 mol) of diisopropylamine and 20 ml of dimethylformamide (DMF) was mixed and stirred. 0.02 mol of DMDO-Br in DMF (5ml) solution was added at 0 °C. At the same temperature, the solution was reacted for 2 hours. The reaction solution was poured into 100 ml of ice water, and the pH value was adjusted to 7 with 20% acetic acid. The solution was filtered after stirring for 30 minutes. The filter residue was washed with water. The solid was collected and dried under vacuum. After recrystallization from acetonitrile, 2.5 g of the target product levo-prulifloxacin was obtained with a purity of 98% and a yield rate of 54%. Specific rotation [α]20D= -108° (c=0.5, 0.1 mol/L methanesulfonic acid).
Example 10 Preparation of R-(+)-prulifloxacin
-
In accordance with the method as described in Example 5, the raw material R-(+)-prulifloxacin was prepared to 2.5 g of the target product R-(+)-prulifloxacin with a purity of 98% and a yield rate of 54%. Specific rotation [α]20 D= +108° (c=0.5, 0.1 mol/L methanesulfonic acid).
Example 11 Preparation of levo-prulifloxacin hydrochloride
-
0.5 g of S-(-)-prulifloxacin was dissolved in 15 mL of chloroform and then 0.5 mL of 33% (v/v) hydrochloric acid- methanol solution was added while stirring. The solution was filtered and the filtration residue was washed with methanol. The collected solid was dried to obtain 450 mg said compound with a yield rate of 83%. The melting point of the product is higher than 220 °C (the sample became darker during the test).
- S-(-)-6-fluoro-1-methyl-7-[4-(5-methyl-2-oxo-1,3-dioxolen-4-yl)methyl-1-piperazinyl] -4-oxo-4H-[1,3]thiazeto[3,2-a]quinoline-3-carboxylic acid hydrochloride
Example 12 Preparation of levo-prulifloxacin mesylate
-
0.5 g of S-(-)-prulifloxacin was dissolved in 15 mL of chloroform and then 0.5 mL of 50% methanesulfonic acid- methanol solution was added while stirring. The solution was filtered and the filtration residue was washed with methanol. The collected yellow solid was dried with calcium chloride under vacuum for 24 hours and further dried with calcium chloride at 80 °C under vacuum for 5 hours to obtain 470 mg said compound with a yield rate of 78%. The melting point of the product is higher than 220 °C (the sample became darker during the test).
- S-(-)-6-fluoro-1-methyl-7-[4-(5-methyl-2-oxo-1,3-dioxolen-4-yl)methyl-1-piperazinyl] -4- oxo-4H-[1,3]thiazeto[3,2-α]quinoline-3-carboxylic acid mesylate
Example 13 Preparation of levo-prulifloxacin hydrochloride
-
0.5 g of S-(-)-prulifloxacin was dissolved in 15 mL of chloroform and then 0.5 mL of 33% (v/v) hydrochloric acid- methanol solution was added while stirring. The solution was dried by evaporation. Methanol was added to the residue and stirred for 10 minutes. The solution was filtered and the filtration residue was washed with methanol. The collected solid was dried to obtain 460 mg said compound with a yield rate of 85%.
-
Fandofloxacin In phase 2 by Dong Wha Pharmaceutical Co Ltd
DW-116; fandofloxacin
164150-99-6 FREE BASE ,
164151-00-2., 164150-85-0
6-fluoro-1-(5-fluoropyridin-2-yl)-7-(4-methylpiperazin-1-yl)-4-oxoquinoline-3-carboxylic acid
6-Fluoro-1-(5-fluoropyridin-2-yl)-7-(4-methylpiperazin-1-yl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid
Dong Wha Pharmaceutical Co Ltd
Molecular Formula: C20H18F2N4O3
Molecular Weight: 400.378726
synthesis………….http://www.drugfuture.com/synth/syndata.aspx?ID=226498
http://www.google.com.mx/patents/WO1995005373A1
synthesis 1

Condensation of ethyl 2,4,5-trifluorobenzoylacetate (I) with triethyl orthoformate in refluxing Ac2O produced the benzoyl ethoxyacrylate (II), which was further condensed with 2-amino-5-fluoropyridine (III) to afford enamine (IV). Cyclization of (IV) in the presence of K2CO3 gave rise to the quinolone (V). The 7-fluoride group of (V) was then displaced by N-methylpiperazine (VI) in cold pyridine to furnish the piperazinyl quinolone (VII). Finally, ester hydrolysis in (VII) under acidic conditions yielded the target compound. In a closely related procedure, ester (V) was hydrolyzed to acid (VIII) using HCl. Subsequent displacement of the 7-fluoride of (VIII) with N-methylpiperazine (VI) provided the desired piperazinyl quinolone.
synthesis 2

Condensation of ethyl 2,4,5-trifluorobenzoylacetate (I) with triethyl orthoformate in refluxing Ac2O produced the benzoyl ethoxyacrylate (II), which was further condensed with 2-amino-5-fluoropyridine (III) to afford enamine (IV). Cyclization of (IV) in the presence of K2CO3 gave rise to the quinolone (V). The 7-fluoride group of (V) was then displaced by N-methylpiperazine (VI) in cold pyridine to furnish the piperazinyl quinolone (VII). Finally, ester hydrolysis in (VII) under acidic conditions yielded the target compound. In a closely related procedure, ester (V) was hydrolyzed to acid (VIII) using HCl. Subsequent displacement of the 7-fluoride of (VIII) with N-methylpiperazine (VI) provided the desired piperazinyl quinolone.
Synthesis, pharmacokinetics, and biological activity of a series of new pyridonecarboxylic acid antibacterial agents bearing a 5-fluoro-2-pyridyl group or a 3-fluoro-4-pyridyl group at N-1
J Heterocycl Chem 1997, 34(3): 1021
|
6-31-2011
|
PHARMACEUTICAL COMPOSITION
|
|
|
8-24-2007
|
PHARMACEUTICAL COMPOSITION
|
|
|
6-29-2007
|
PHARMACEUTICAL COMPOSITION
|
|
|
10-28-2005
|
Identification and use of effectors and allosteric molecules for the alteration of gene expression
|
|
|
7-15-2005
|
Pharmaceutical composition
|
|
|
2-6-2004
|
Medicinal composition
|
|
|
4-20-2000
|
NOVEL QUINOLONE CARBOXYLIC ACID DERIVATIVES
|
|
|
3-6-1996
|
Quinolone carboxylic acid derivatives
|
|
|
2-24-1995
|
NOVEL QUINOLONE CARBOXYLIC ACID DERIVATIVES
|
ELLAGIC ACID A CANCER FIGHTING WONDER
ELLAGIC ACID
2,3,7,8-Tetrahydroxy-chromeno[5,4,3-cde]chromene-5,10-dione
as a very potent CK2 inhibitor

Ellagic acid is a natural phenol antioxidant found in numerous fruits and vegetables. The antiproliferative and antioxidant properties of ellagic acid have spurred preliminary research into the potential health benefits of ellagic acid consumption.
Ellagic acid is the dilactone of hexahydroxydiphenic acid.
Ellagic acid is an antioxidant and an anti-proliferative compound present in fruits, nuts and vegetables. In spite of evidences for anticancer activity in various cancer cell-lines, human cancer cells, the mechanistic role of ellagic acid is not conclusive enough to be recommended for a clinical use. The present review provides information about the chemopreventive role of ellagic acid in oral cancer and proposes molecular basis for ellagic acid’s inhibitory activity against oral cancer. We show that ellagic acid modulates growth of tumor cells through regulation of multiple cell signaling pathways including cell proliferation pathway (cyclin dependent kinase 2, cyclin A2, cyclin B1, cyclin D1, c-myc, PKCα), cell survival/apoptosis pathway (Bcl-XL, Bax, Caspase 9/3, Akt), tumor suppressor pathway (p53, p21), inflaming Metastasis pathways (IL-1 beta, TNF-α, matrix metalloproteinases 9/3, COX-2), angiogenesis pathways (VEGF), cell immortalization (TERT), NF-κβ.
Chinese medicine…Cordyceps ( dong chong xia cao ) 冬蟲草 དབྱར་རྩྭ་དགུན་འབུ་ ………..to treat many diseases related to lungs, kidney, and also used as a natural Viagra.
Ophiocordyceps sinensis (left) growing out of the head of a dead caterpillar
Ophiocordyceps sinensis is a fungus that parasitizes larvae of ghost moths and produces a fruiting body valued as an herbal remedy. The fungus germinates in the living larva, kills and mummifies it, and then the stalk-like fruiting body emerges from the corpse. It is known in English colloquially as caterpillar fungus, or by its more prominent foreign names (see below): yartsa gunbu or yatsa gunbu (Tibetan), or Dōng chóng xià cǎo (Chinese: 冬虫夏草; literally “winter worm, summer grass”). Of the various entomopathogenic fungi, Ophiocordyceps sinensis is one that has been used for at least 2000 years[2] to treat many diseases related to lungs, kidney, and also used as a natural Viagra. This fungus is not yet cultivated commercially,[3] despite the fact that several fermentable strains of Ophiocordyceps sinensis are isolated by Chinese Scientists.[4] Overharvesting and over exploitation have led to the classification of O. sinensis as an endangered species in China.[5] Additional research needs to be carried out in order to understand its morphology and growth habit for conservation and optimum utilization.

The moths in which O. sinensis grows are ambiguously referred to as “ghost moth”, which identifies either a single species or the genus Thitarodes, and the species parasitized by O. sinensis may be one of several Thitarodes that live on the Tibetan Plateau (Tibet, Qinghai, West-Sichuan, SW-Gansu & NW Yunnan), and the Himalayas (India, Nepal, Bhutan).
O. sinensis is known in the West as a medicinal mushroom, and its use has a long history in Traditional Chinese medicine as well as Traditional Tibetan medicine.[6] The hand-collected fungus-caterpillar combination is valued by herbalists and as a status symbol;[7] it is used as an aphrodisiac and treatment for ailments such as fatigue and cancer, although such use is mainly based on traditional Chinese medicine and anecdote. Recent research however seems to indicate a variety of beneficial effects in animal testing, including increased physical endurance through heightened ATP production in rats.[8]
| Cordyceps Sinensis |
| Cordyceps sinensis (Berk.) Sacc. and the usually the larvae are the remains of Hepialus varians |
| tonifies lung yin and kidney yang. For impotence, chronic lower back pain, afraid of cold, over abundance of mucus and tears, chronic cough and wheezing from deficiency, blood in phlegm from consumption due tokidney yang deficiency (shenyangxu). |
Cordyceps ( Dong Chong Cao ) 冬蟲草 Chinese Herbal Articles also known as chong cao, dong chong cao, yarsa gumba (Nepalese name of Tibetan origin), yartsa gunbu (dbyar rtswa dgun ‘bu) Tibetan name 蟲草, 冬蟲草. It belong to the “Ascomycetes or Clavicipitaceae” family.
Cordyceps ( Dong Chong Cao ) 冬蟲草 has a sweet, warm properties. It is use for treating the lung and kidney.

|
1. Improves auto-immune system.
2. Protects kidneys from toxins.
3. Protects kidneys from exhaustion.
4. Protects liver from toxins and treats and prevents cirrhosis of liver.
5. Protect the heart from the damaging effect of ouabain (C29H44O12.8H2O).
6. Anti-arrhythmia.
7. Anti-rejection effect in cornea transplant.
8. Antibiotic effect.
9. Inhibits contraction of smooth muscles.
|

Cordyceps ( Dong Chong Cao ) 冬蟲草 use in large dosages and/or long term usage can be toxic to kidneys.
According to the classics Medical Material, “Ben Cao Bei Yao” 本草備要, the best dong chong xia cao 冬蟲夏草, are produced in Sichuan. Today, most of them are produced in Xizang (Tibet) and Qinghai. Because the sizes the larvae are larger, they fetch higher prices.
According to the classics Medical Material, “Ben Cao Bei Yao” 本草備要, the best dong chong xia cao 冬蟲夏草, are produced in Sichuan. Today, most of them are produced in Xizang (Tibet) and Qinghai. Because the sizes the larvae are larger, they fetch higher prices.
Taxonomic History/ Systematics
Morphological Features
Similar to other Cordyceps]] species, O. sinensis consists of two parts, a fungal endosclerotium (caterpillar) and stroma.[2] The stroma is the upper fungal part and is dark brown or black, but can be a yellow color when fresh and, longer than the caterpillar itself, usually 4–10 cm. It grows singly from the larval head, and is clavate, sublanceolate or fusiform and distinct from the stipe.[9] The stipe is slender, glabrous, and longitudinally furrowed or ridged. The fertile part of the stroma is the head. The head is granular due to the ostioles of the embedded perithecia.[2] The perithecia are ordinally arranged and ovoid [9] The asci are cylindrical or slightly tapering at both ends, and may be straight or curved, with a capitate and hemispheroid apex and may be two to four spored.[2] Similarly, ascospores are hyaline, filiform, multiseptate at a length of 5-12 um and subattenuated on both sides.[9] Perithecial, ascus and ascospore characters in the fruiting bodies are the key identification characteristics of O. sinensis. Ophiocordyceps (Petch) Kobayasi species produce whole ascospores and do not separate into part spores which is different from other Cordyceps species, which produce either immersed or superficial perithecia perpendicular to stromal surface and the ascospores at maturity are disarticulated into part spores.[10] Generally Cordyceps species possess brightly colored and fleshy stromata, but O. sinensis had dark pigments and tough to pliant stromata, a typical characteristic feature of most of the Ophiocordyceps species.[3]
Important developments in Classification
The species was first described scientifically by Miles Berkeley in 1843 as Sphaeria sinensis;[11] Pier Andrea Saccardo transferred the species to the genus Cordyceps in 1878.[12]The scientific name‘s etymology is from the Latin cord “club”, ceps “head”, and sinensis “from China“. The fungus was known as Cordyceps sinensis until 2007, when molecularanalysis was used to emend the classification of the Cordycipitaceae and the Clavicipitaceae, resulting in the naming of a new family Ophiocordycipitaceae and the transfer of several Cordyceps species to Ophiocordyceps.[10] Based on a molecular phylogenetic study, Sung et al. (2007) separated the megagenus Cordyceps into four genera as it was polyphyletic, viz. Cordyceps (40 spp.), Ophiocordyceps (146 spp.), Metacordyceps (6 spp.) and Elaphocordyceps (21 spp.), while the remaining 175 spp. were left in Cordyceps. As a result, C. sinensis was transferred to Ophiocordyceps, hence renamed as O. sinensis.[2]
Common Names[edit]
In Tibetan it is known as དབྱར་རྩྭ་དགུན་འབུ་ (ZYPY: yartsa gunbu, Wylie: dbyar rtswa dgun ‘bu, “summer grass winter bug”), which is the source of the Nepali यार्शागुम्बा, yarshagumba,yarchagumba or yarsagumba. The transliteration in Bhutan is Yartsa Guenboob. It is known as keera jhar, keeda jadi, keeda ghas or ‘ghaas fafoond in Hindi. Its name in Chinese Dōng chóng xià cǎo (冬蟲夏草) means “winter worm, summer grass” (i.e., “worm in the winter, [turns to] plant in the summer”). The Chinese name is a literal translation of the original Tibetan name, which was first recorded in the 15th Century by the Tibetan doctor Zurkhar Namnyi Dorje. In colloquial Tibetan Yartsa gunbu is often shortened to simply “bu” or “yartsa”.
In traditional Chinese medicine, its name is often abbreviated as chong cao (蟲草 “insect plant”), a name that also applies to other Cordyceps species, such as C. militaris. InJapanese, it is known by the Japanese reading of the characters for the Chinese name, tōchūkasō (冬虫夏草).
Strangely, sometimes in Chinese English language texts Cordyceps sinensis is referred to as aweto [Hill H. Art. XXXVI: The Vegetable Caterpillar (Cordiceps robertsii). Transactions and Proceedings of the Royal Society of New Zealand 1868-1961. Vol 34, 1901;396-401], which is the Māori name for Cordyceps robertsii, a species from New Zealand.
The English term “vegetable caterpillar” is a misnomer, as no plant is involved. “Caterpillar fungus” is a preferable term.
Nomenclature of the anamorph
Since the 1980s, 22 species in 13 genera have been attributed to the anamorph of O. sinensis. Of the 22 species, Cephalosporium acreomonium is the zygomycetous species ofUmbelopsis, Chrysosporium sinense has very low similarity in RAPD polymorphism, hence it is not the anamorph. Likewise, Cephalosporium dongchongxiacae, C. sp. sensu,Hirsutella sinensis and H. hepiali and Synnematium sinnense are synonymous and only H. sinensis is only validly published in articles. Cephalosporium sinensis possibly might be synonymous to H. sinensis but there is lack of valid information. Isaria farinose is combined to Paecilomyces farinosus and is not the anamorph. Several species like Isaria sp. Verticella sp. Scydalium sp. Stachybotrys sp. were identified only up to generic level, and thus it is dubious that they are anamorph. Mortierella hepiali is discarded as anamorph as it belongs to Zygomycota. Paecilomyces sinensis and Sporothrix insectorum are discarded based on the molecular evidence. P. lingi appeared only in one article and thus is discarded due to incomplete information. Tolypocladium sinense, P. hepiali, and Scydalium hepiali, have no valid information and thus are not considered as anamorph toOphiocordyceps sinensis. V. sinensis is not considered anamorph as there is no valid published information. Similarly, Metarhizium anisopliae is not considered anamorph as it has widely distributed host range, and is not restricted only in high altitude.[13] Thus Hirsutella sinensis is considered the validly published anamorph of O. sinensis. Cordyceps nepalensis and C. multiaxialis which had similar morphological characteristics to C. sinensis, also had almost identical or identical ITS sequences and its presumed anamorph, H. sinensis. This also confirms H. sinensis to be anamorph of O. sinensis and suggests C. nepalensis and C. multiaxialis are synonyms.[14] Evidence based on microcyclic conidiation from ascospores and molecular studies [2] support H. sinensis as the anamorph of the caterpillar fungus, O. sinensis.
Ecology
The caterpillars prone to infection by O. sinensis generally live 6 inches underground [4] in alpine grass and shrub-lands on the Tibetan Plateau and the Himalayas at an altitude between 3,000 and 5,000 m (9,800 and 16,400 ft). The fungus is reported from the northern range of Nepal, Bhutan, and also from the northern states of India, apart from northern Yunnan, eastern Qinghai, eastern Tibet, western Sichuan, southwestern Gansu provinces.[4] The fungus consumes its host from inside out as they hibernate in alpine meadows. Usually the larvae are more vulnerable after shedding their skin, during late summer. The fungal fruiting body disperses spores which infect the caterpillar. The infected larvae tend to remain vertical to the soil surface with their heads up. The fungus then germinates in the living larva, kills and mummifies it, and then the stalk-like fruiting body emerges from the head and the fungus finally emerges from the soil by early spring.[15] Fifty-seven taxa from seven genera (1 Bipectilus, 1 Endoclita, 1 Gazoryctra, 12 Hepialus, 2Magnificus, 3 Pharmacis, and 37 Thitarodes [3]) are recognized as potential hosts of O. sinensis.
Reproduction Biology
Ophiocordyceps sinensis has both teleomorphic and anamorphic phases. Spending up to five years underground before pupating, the Thitarodes caterpillar is attacked while feeding on roots. It is not certain how the fungus infects the caterpillar; possibly by ingestion of a fungal spore or by the fungus mycelium invading the insect through one of the insect’s breathing pores. The dark brown to black fruiting body (or mushroom) emerges from the ground in spring or early summer, the long, usually columnar fruiting body reaches 5–15 cm above the surface and releases spores.
In late autumn, chemicals on the skin of the caterpillar interact with the fungal spores and release the fungal mycelia, which then infects the caterpillar.[4] After invading a host larva, the fungus ramifies throughout the host and eventually kills it. Gradually the host larvae become rigid due to the production of fungal sclerotia. Fungal sclerotia are multihyphal structures that can remain dormant and then germinate to produce spores. After over-wintering, the fungus ruptures the host body, forming a sexual sporulating structure (a perithecial stroma) from the larval head in summer that is connected to the sclerotia (dead larva) below ground and grows upward to emerge from the soil.[16] The slow growing O. sinensis grows at a comparatively low temperature, i.e., below 21oC. Temperature requirements and growth rates are crucial factors that identify O. sinensis from other similar fungi.[3]
Use in medicine
It is used as a curative to many diseases, anti- aging,[17] hypoglycemic,[18] aphrodisiac and also treatment against cancer. Ophiocordyceps sinensis serves against kidney and lung problems and stimulates the immune system; it is used for treatment of fatigue, night sweating, respiratory disease, hyperglycemia, hyperlipidemia, asthenia after severe illness, arrhythmias and other heart diseases and liver disease.[4]
Traditional Asian medicines
Medicinal use of the caterpillar fungus apparently originated in Tibet and Nepal. So far the oldest known text documenting its use was written in the late fourteen hundreds by the Tibetan doctor Zurkhar Nyamnyi Dorje (Wylie: Zur mkhar mnyam nyid rdo rje)[1439-1475]) in his text: Man ngag bye ba ring bsrel (“Instructions on a Myriad of Medicines”). A translation is available at Winkler.[19]
The first mention of Ophiocordyceps sinensis in traditional Chinese Medicine was in Wang Ang’s 1694 compendium of materia medica, Ben Cao Bei Yao.[20] In the 18th Century it was listed in Wu Yiluo‘s Ben cao cong xin (“New compilation of materia medica”).[21] No sources have been published to uphold widespread claims of “thousands of years of use in Chinese medicine” or use of “chong cao since the 7th Century Tang Dynasty in China”. The ethno-mycological knowledge on caterpillar fungus among the Nepalese people is documented byDevkota(2006) The entire fungus-caterpillar combination is hand-collected for medicinal use.
The fungus is a medicinal mushroom which is highly prized by practitioners of Tibetan medicine, Chinese medicine and traditional Folk medicines, in which it is used as an aphrodisiac and as a treatment for a variety of ailments from fatigue to cancer. In Chinese medicine it is regarded as having an excellent balance of yin and yang as it is apparently both animal and vegetable. Assays have found thatOphiocordyceps species produce many pharmacologically active substances. They are now cultivated on an industrial scale for their medicinal value. However, no one has succeeded so far in growing the larva cum mushroom artificially. The biological process that forms the Ophiocordyceps is still unknown and true cultivation has yet to be realized.[3] All artificial products are derived from mycelia grown on grains or in liquids.
According to Bensky et al. (2004), laboratory-grown C. sinensis mycelia have similar clinical efficacy and less associated toxicity. He notes a toxicity case of constipation, abdominal distension, and decreased peristalsis, two cases of irregular menstruation, and one case report ofamenorrhea following ingestion of tablets or capsules containing C. sinensis. In Chinese medicine C. sinensis is considered sweet and warm, entering the lung and kidney channels; the typical dosage is 3–9 grams.[22]
Research
Cordycepin, a compound isolated from the “Caterpillar fungus”.
Some work has been published in which Ophiocordyceps sinensis has been used to protect the bone marrow and digestive systems ofmice from whole body irradiation.[23] An experiment noted Ophiocordyceps sinensis may protect the liver from damage.[24] An experiment conducted with mice noted the mushroom may have an anti-depressant effect.[25] Researchers have noted that the caterpillar fungus has ahypoglycemic effect and may be beneficial for people with insulin resistance.[26][27][28][29][30] There is also experimental evidence of the supposed energizing effect of the fungus, as it has been shown to increase endurance through heightened ATP production in rats.[8]
A March 2013 study on Cordyceps Sinensis documented the medicinal fungus’ anti-inflammatory properties.[31] Scientists were able to show Cordyceps Sinensis’ ability to suppress interleukin-1b and interleukin-18 secretion by inhibiting both canonical and non-canonical inflammasomes. Inflammasomes have long been associated with auto-inflammatory diseases, such as gout. The study used a specific anamorphic mycelial form of Cordyceps Sinensis known as Hirsutella Sinensis.
Introduction to the Western world
| ) |
The Western world was largely unaware of Ophiocordyceps prior to 1993. The fungus dramatically caught the world’s eye due to the performance of three female Chinese athletes, Wang Junxia, Qu Yunxia, and Zhang Linli. These athletes broke five world records for 1,500, 3,000 and 10,000 meter dashes at the National Games in Beijing, China. The number of new world records set at a single track event attracted much attention and suspicion. Following the races, the women were expected by some to fail drug tests for anabolic steroids. However, the athletes’ tests revealed no illegal substances, and coach Ma Junren told the reporters that the runners were takingOphiocordyceps sinensis and turtle blood at his request. However for the 2000 Sydney Olympics, Ma Junren withdrew some of his athletes at the last minute. It was speculated that a new doping test would have revealed illegal substances, thus half a dozen Chinese field and track athletes were left at home.
Economics and impact
Many shops in downtown Lanzhouadvertise Dōng chóng xià cǎo (冬虫夏草) among other local specialties.
In rural Tibet, yartsa gunbu has become the most important source of cash income. The fungi contributed 40% of the annual cash income to local households and 8.5% to the GDP in 2004. Prices have increased continuously, especially since the late 1990s. In 2008, one kilogram traded for US$3,000 (lowest quality) to over US$18,000 (best quality, largest larvae). The annual production on the Tibetan Plateau was estimated in 2009 at 80–175 tons.[32] The Himalayan Ophiocordyceps production might not exceed a few tons.
In 2004 the value of a kilogram of caterpillars was estimated at about 30,000 to 60,000 Nepali rupees in Nepal, and about Rs 100,000 in India.[33] In 2011 the value of a kilogram of caterpillars was estimated at about 350,000 to 450,000 Nepali rupees in Nepal. A 2012 BBC article indicated that in north Indian villages a single fungus was worth Rs 150 (about £2 or $3), which is more than the daily wage of a manual laborer.[34]
According to Daniel Winkler, the price of Ophiocordyceps sinensis has risen dramatically on the Tibetan Plateau, basically 900% between 1998 and 2008, an annual average of over 20% (after inflation). However, the value of big sized caterpillar fungus has increased more dramatically than smaller size Cordyceps, regarded as lower quality.[20]
| Year | % Price Increase | Price/kg (Yuan) |
|---|---|---|
| 1980s | 1,800 | |
| 1997 | 467% (incl. inflation) | 8,400 |
| 2004 | 429% (incl. inflation) | 36,000 |
| 2005 | 10,000–60,000 | |
| 2013 | 125,000–500,000 |
Because of its high value, inter-village conflicts over access to its grassland habitats has become a headache for the local governing bodies and in several cases people were killed. In November 2011, a court in Nepal convicted 19 villagers over the murder of a group of farmers during a fight over the prized aphrodisiac fungus. Seven farmers were killed in the remote northern district of Manang in June 2009 after going to forage for Yarchagumba. [35]
Its value gave it a role in the Nepalese Civil War, as the Nepalese Maoists and government forces fought for control of the lucrative export trade during the June–July harvest season.[36] Collecting yarchagumba in Nepal had only been legalised in 2001, and now demand is highest in countries such as China, Thailand, Vietnam, Korea and Japan. By 2002, the herb was valued at R 105,000 ($1,435) per kilogram, allowing the government to charge a royalty of R 20,000 ($280) per kilogram.
The search for Ophiocordyceps sinensis is often perceived to pose a threat to the environment of the Tibetan Plateau where it grows. While it has been collected for centuries and is still common in such areas, current collection rates are much higher than in historical times.
Ophiocordyceps producers like to perpetuate the story that unscrupulous harvesters insert twigs into the ascocarps of wild C. sinensis to increase their weight and therefore the price paid. A tiny twig is only used when the ascocarp is broken from the caterpillar, and has nothing to do with artificially increasing weight. Supposedly, at some point in the past, someone inserted lead wires with which to increase weight; however, each year hundreds of millions of specimens are harvested and this appears to have been a one-time occurrence.
Cultivated C. sinensis mycelium is an alternative to wild-harvested C. sinensis, and producers claim it may offer improved consistency. Artificial culture of C. sinensis is typically by growth of pure mycelia in liquid culture (in China) or on grains (in the West). The first time in Vietnam, Prof. Aca. Dr. Dai Duy Ban together with scientists and DAIBIO Company and DAIBIO Great Traditional Medicine Family Clinic discovered the Cordyceps sinensis as Isaria cerambycidae N.SP. to develop Fermentation DAIBIO Cordyceps Sinensis.[37]Ascocarps are not produced through in vitro cultivation.
References
- “Ophiocordyceps sinensis (Berk.) G.H. Sung, J.M. Sung, Hywel-Jones & Spatafora 2007″. MycoBank. International Mycological Association. Retrieved 2011-07-19.
- Shrestha, B., Weimin, Z., Yongjie, Z., & Xingzhong, L. (2010). What is the Chinese caterpillar fungus Ophiocordyceps sinensis (Ophiocordycipitaceae)?. Mycology: An International Journal On Fungal Biology, 1(4), 228-236. doi:10.1080/21501203.2010.536791.
- Hsieh, C., et al., A Systematic Review of the Mysterious Caterpillar Fungus Ophiocordyceps sinensis in Dong-ChongXiaCao and Related Bioactive Ingredients. Vol. 3. 2013. 16-32.
- Zhu JS, Halpem GM, Jones K. 1998. The scientific rediscovery of an ancient Chinese herbal medicince: Cordyceps sinensis. I. J Alt Complem Med 4:289-303.
- Xiao-Liang, W., & Yi-Jian, Y. (2011). Host insect species of Ophiocordyceps sinensis: a review. Zookeys, 12743-59. doi:10.3897/zookeys.127.802
- Halpern, Miller (2002). Medicinal Mushrooms. New York, New York: M. Evans and Company, Inc. pp. 64–65. ISBN 0-87131-981-0
- http://www.npr.org/2011/10/09/141164173/caterpillar-fungus-the-viagra-of-the-himalayas
- ^ Jump up to:a b Rajesh Kumar, P.S. Negi, Bhagwat Singh, Govindasamy Ilavazhagan, Kalpana Bhargava, Niroj Kumar Sethy (2011). “Cordyceps sinensis promotes exercise endurance capacity of rats by activating skeletal muscle metabolic regulators”. Journal of Ethnopharmacology 136: 260–266.
- Sung, G. H., et al. (2007). “A multi-gene phylogeny of Clavicipitaceae (Ascomycota, Fungi): identification of localized incongruence using a combinational bootstrap approach.” Molecular Phylogenetics and Evolution 44(3): 1204-1223.
- Sung GH, Hywel-Jones NL, Sung JM, Luangsa-Ard JJ, Shrestha B, Spatafora JW. (2007). “Phylogenetic classification of Cordyceps and the clavicipitaceous fungi”.Studies in Mycology 57: 5–59. doi:10.3114/sim.2007.57.01. PMC 2104736.PMID 18490993.
- Berkeley MJ. (1843). “On some entomogenous Sphaeriae”. London Journal of Botany 2: 205–11.
- Saccardo PA. (1878). “Enumeratio Pyrenomycetum Hypocreaceorum hucusque cognitorum systemate carpologico dispositorum” (PDF). Michelia (in Latin) 1 (3): 277–325.
- Jiang, Y. Y., & Yao, Y. J. (n.d). Names related to Cordyceps sinensis anamorph. Mycotaxon, 84245-254.
- Liu, Z., Liang, Z., Liu, A., Yao, Y., Hyde, K. D., & Yu, Z. (n.d). Molecular evidence for teleomorph-anamorph connections in Cordyceps based on ITS-5.8S rDNA sequences. Mycological Research, 106(9), 1100-1108.
- Stone, R. (2008). Last Stand for the Body Snatcher of the Himalayas?. Science, (5905), 1182. doi:10.2307/20145300
- Xing, X. K., & Guo, S. X. (2008). The Structure and Histochemistry of Sclerotia of Ophiocordyceps sinensis. Mycologia, (4), 616. doi:10.2307/20444986.
- Ji DB, Ye J, Li CL, Wang YH, Zhao J, Cai SQ (2009) Antiaging effect of Cordyceps sinensis extract. Phytotherapy Research 23 (1): 116-122. Doi: 10.1002/ptr.2576
- Zhang GQ, Huang YD, Bian Y, Wong JH, Ng TB, Wang HX (2006) Hypoglycemic activity of the fungus Cordyceps militaris, Cordyceps sinensis, Tricholoma mongolicum and Omphalia lapidescens in streptozotocin-induced diabetic rats. Applied Microbiology and Biotechnology 72 (6): 1152-1156. Doi: 10.1007/s00253-006-0411-9.
- Winkler D. (2008). “The mushrooming fungi market in Tibet exemplified by Cordyceps sinensis and Tricholoma matsutake“. Journal of the International Association of Tibetan Studies. In: In the Shadow of the Leaping Dragon: Demography, Development, and the Environment in Tibetan Areas (4).
- Winkler D. (2008). “Yartsa Gunbu (Cordyceps sinensis) and the fungal commodification of the rural economy in Tibet AR”. Economic Botany 62 (3): 291–305.doi:10.1007/s12231-008-9038-3.
- Wu Y (1757). “Ben cao cong xin” – “New compilation of materia medica” (in Chinese).
- Jump up^ Bensky D, Gamble A, Clavey S, Stöger E, Bensky L. Lai (2004). Materia Medica: Chinese Herbal Medicine (3rd ed.). Seattle, Washington: Eastland Press. ISBN 978-0-939616-42-8.
- Liu W-C, Wang S-C, Tsai M-L, Chen, M-C, Wang Y-C, Hong J-H, McBride WH, Chiang C-S. (2006). “Protection against radiation-induced bone marrow and intestinal injuries byCordyceps sinensis, a Chinese herbal medicine”. Radiation Research 166 (6): 900–907.doi:10.1667/RR0670.1. PMID 17149981.
- WS, Hsu SL, Chyau CC, Chen KC, Peng RY. (July 2009). “Compound Cordyceps TCM-700C exhibits potent hepatoprotective capability in animal model”. Fitoterapia 81(1): 1–7. doi:10.1016/j.fitote.2009.06.018. PMID 19596425.
- Nishizawa K, Torii K, Kawasaki A, et al. (2007). “Antidepressant-like effect ofCordyceps sinensis in the mouse tail suspension test”. Biological and Pharmaceutical Bulletin 30 (9): 1758–62. doi:10.1248/bpb.30.1758. PMID 17827735.
- Kiho T, Hui J, Yamane A, Ukai S. (1993). “Polysaccharides in fungi. XXXII. Hypoglycemic activity and chemical properties of a polysaccharide from the cultural mycelium of Cordyceps sinensis“. Biological and Pharmaceutical Bulletin 16 (12): 1291–3. doi:10.1248/bpb.16.1291. PMID 8130781.
- Kiho T, Yamane A, Hui J, Usui S, Ukai S. (1996). “Polysaccharides in fungi. XXXVI. Hypoglycemic activity of a polysaccharide (CS-F30) from the cultural mycelium of Cordyceps sinensis and its effect on glucose metabolism in mouse liver”. Biological and Pharmaceutical Bulletin 19 (2): 294–6. doi:10.1248/bpb.19.294. PMID 8850325.
- Zhao CS, Yin WT, Wang JY, et al. (2002). “CordyMax Cs-4 improves glucose metabolism and increases insulin sensitivity in normal rats”. Journal of Alternative and Complementary Medicine 8 (3): 309–14. doi:10.1089/10755530260127998.PMID 12165188.
- Lo HC, Tu ST, Lin KC, Lin SC. (2004). “The anti-hyperglycemic activity of the fruiting body of Cordyceps in diabetic rats induced by nicotinamide and streptozotocin”. Life Sciences 74 (23): 2897–908. doi:10.1016/j.lfs.2003.11.003. PMID 15050427.
- Li SP, Zhang GH, Zeng Q, et al. (2006). “Hypoglycemic activity of polysaccharide, with antioxidation, isolated from cultured Cordyceps mycelia”. Phytomedicine 13 (6): 428–33.doi:10.1016/j.phymed.2005.02.002. PMID 16716913.
- Huang, T. et al. (March 2013). “Hirsutella sinensis mycelium suppresses interleukin-1b and interleukin-18 secretion by inhibiting both canonical and non-canonical inflammasomes.” (PDF). Scientific Report. 3, 1374;.
- Winkler, D. (2009). “Caterpillar Fungus (Ophiocordyceps sinensis) Production and Sustainability on the Tibetan Plateau and in the Himalayas”. Asian Medicine 5 (2): 291. doi:10.1163/157342109X568829.
- Sharma S. (2004). “Trade of Cordyceps sinensis from high altitudes of the Indian Himalaya: Conservation and biotechnological priorities” (PDF). Current Science 86(12): 1614–9.
- Jeffrey, Craig (2012-07-07). “The ‘Viagra’ transforming local economies in India”. BBC News. Retrieved July 9, 2012.
- Staff (14 November 2011) ‘Himalayan viagra’: Six men get life for Nepal murders BBC News Asia, Retrieved 9 July 2012
- Baral N, Heinen JT. (2005). “The Maoist people’s war and conservation in Nepal”.Politics and the Life Sciences 24 (1): 2–11. doi:10.2990/1471-5457(2005)24[2:TMPWAC]2.0.CO;2.
- DAIBIO Cordyceps Sinensis in Vietnam
- Winkler, D. 2005. Yartsa Gunbu – Cordyceps sinensis. Economy, Ecology & Ethno-mycology of a Fungus Endemic to the Tibetan Plateau. In: A.BOESI & F. CARDI (eds.). Wildlife and plants in traditional and modern Tibet: Conceptions, Exploitation and Conservation. Memorie della Società Italiana di Scienze Naturali e del Museo Civico di Storia Naturale di Milano, Vol. 33.1:69–85.
- Zhang Y., Zhang S., Wang M., Bai F. & Liu X. (2010). “High Diversity of the Fungal Community Structure in Naturally-Occurring Ophiocordyceps sinensis“. PLoS ONE 5(12): e15570. doi:10.1371/journal.pone.0015570.
External links
Yartsa Gunbu (Cordyceps sinensis) in Tibet
- Daniel Winkler’s Cordyceps blog
- Nepal’s Nature – The Himalayan Viagra
- Page at Everything2.com
- Image gallery of Cordyceps sinensis
- The first time in Vietnam, Prof.Aca.D.Sc Dai Duy Ban with his scientists discovered Cordyceps Sinensis as Isaria cerambycidae N.SP. and Fermentation Daibio Cordyceps Sinensis by Daibio Great Family Traditional Medicine Clinic Company
- Daibio Cordyceps Sinensis in Vietnam
- An Electronic Monograph of Cordyceps and Related Fungi
- Cordyceps information from Drugs.com
- Cordyceps sinensis (Berk.) Sacc. Medicinal Plant Images Database (School of Chinese Medicine, Hong Kong Baptist University) (English) (traditional Chinese)
- Chinese Caterpiller Fungus Chinese Medicine Specimen Database (School of Chinese Medicine, Hong Kong Baptist University) (English) (traditional Chinese)
- Tibet’s Golden “Worm” August 2012 National Geographic (magazine)
Dandelion, Burdock, and Cancer
burdock
dandelionDandelion root and burdock root are my two most commonly prescribed herbs when chronic conditions require anti-inflammatory, blood purifying alteratives for gentle detoxification. This includes conditions such as arthritis and cancer. I’ve studied literally hundreds of herbs from around the world, and considering cost, availability, palatability (no small matter, as people with chronic disease like cancer need to be able to take their herbs at least three times a day for months) – there are probably no two more simple and powerful anticancer herbs on the planet than dandelion and burdock.*
After prescribing both of these in strong dose clinically for years with great results (patients feel better, or experience slowing or even complete remission of some cancers), I learned that many professional British medical herbalists also use the same two-herb combination for conditions requiring blood, lymphatic and liver detoxification.
http://www.planetherbs.com/michaels-blog/dandelion-burdock-and-cancer.html
Glenmark’s novel molecule ‘GRC 27864’ for chronic inflammatory diseases including pain entering human trials

Glenmark’s novel molecule ‘GRC 27864’ for chronic inflammatory diseases including pain entering human trials
- GRC 27864 is a potent, selective, orally bioavailable inhibitor of mPGES-1
- The molecule has successfully completed pre-clinical and Phase 1 enabling studies. Regulatory submission has been filed for Phase 1 trial (first-in-human)with MHRA, UK
- mPGES-1 inhibitors selectively block the production of PGE2 while sparing other prostanoids of physiological importance
- With this announcement, Glenmark has reaffirmed its position globally in the development of novel pain therapies
Mumbai, India: April 3, 2014: Glenmark Pharmaceuticals today announced that its Novel Chemical Entity (NCE) ‘GRC 27864’ is entering human trials. This NCE program targets Microsomal Prostaglandin E synthase-1 (mPGES-1) as a novel therapeutic target in pain management. Selective mPGES-1 inhibitors are expected to inhibit increased prostaglandin E2 (PGE2) production in the disease state without affecting other prostanoid metabolites and, consequently, may be devoid of the GI(gastrointestinal) and cardiovascular side effects seen with NSAIDs and COX-2 inhibitors, respectively.
Glenmark has completed preclinical studies and Phase 1 enabling GLP studies for its selected lead molecule, GRC 27864 and has filed a Phase 1 application forfirst-in-human trial with the MHRA, UK. The Phase 1 studies are to be initiated soon and are likely to get completed by January 2015. Following this, Glenmark will also be initiating a proof of concept study in patients with acute pain.
ANTHONY CRASTO’S NEW DRUG APPROVALS TOUCHES 2 LAKH VIEWS IN 179 COUNTRIES

ANTHONY CRASTO’S NEW DRUG APPROVALS TOUCHES 2 LAKH VIEWS IN 179 COUNTRIES
DR ANTHONY MELVIN CRASTO Ph.D
WORLDDRUGTRACKER,
OTHERS
- Eurekamoments in Organic Chemistry
- NEW DRUG APPROVALS
- WORLD DRUG TRACKER
- Green Chemistry International
- drug regulatory affairs international
- ORGANIC SPECTROSCOPY INTERNATIONAL
- ORGANIC SYNTHESIS INTERNATIONAL
- ALL ABOUT DRUGS
- ORGANIC CHEMISTRY INTERNATIONAL
- Drug Scaleup and Manufacturing International
SEE ALSO
- Organic Chemistry by Dr Anthony
- Technology Transfer
- Stereochemistry
- Spectroscopy
- Polymorphism
- Reactions in Org Chem
DR ANTHONY MELVIN CRASTO, Worlddrugtracker, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his PhD from ICT ,1991, Mumbai, India, in Organic chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with GLENMARK- GENERICS LTD, Research centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Prior to joining Glenmark, he worked with major multinationals like Hoechst Marion Roussel, now sSanofi, Searle India ltd, now Rpg lifesciences, etc. he is now helping millions, has million hits on google on all organic chemistry websites. His New Drug Approvals, Green Chemistry International, Eurekamoments in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 25 year tenure, good knowledge of IPM, GMP, Regulatory aspects, he has several international drug patents published worldwide . He gas good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, polymorphism etc He suffered a paralytic stroke in dec 2007 and is bound to a wheelchair, this seems to have injected feul in him to help chemists around the world, he is more active than before and is pushing boundaries, he has one lakh connections on all networking sites, He makes himself available to all, contact him on +91 9323115463, amcrasto@gmail.com
Personal Links
- my sites on the net
- DR ANTHONY MELVIN CRASTO
- GOOGLE GROUP ORGANIC PROCESS DEVELOPMENT
- mixxt
- epernicus
- scipeople
- jimdo
- yolasite
- my cv
- slidestaxx
- wordpress blog
- ABOUT ME
- BRANDSITE
- SKILLPAGES
- Academia.edu
- RESEARCHGATE
- DIIGO
- SLIDESHATE
- WIX
- WIX BLOG
- ISSUU
- SCRIBD
- BIZ
- GOOGLE BLOG
- APNACIRCLE
- Eurekamoments in Organic Chemistry
- Organic Chemistry by Dr Anthony
- Green Chemistry International
- Technology Transfer
- Stereochemistry
- Spectroscopy
- Polymorphism
- Reactions in Org Chem
- DR ANTHONY MELVIN CRASTO Ph.D
- Pharmaceuticals
- Medicinal chemistry
- Organic chemistry literature
- Patent related site
- Green chemistry
- Reagents
- R & D
- Molecules
- Heterocyclic chem
- Sourcing
- NEW DRUG APPROVALS
- WORLD DRUG TRACKER
- Green Chemistry International
- drug regulatory affairs international
- ORGANIC SPECTROSCOPY INTERNATIONAL
- ORGANIC SYNTHESIS INTERNATIONAL
- ALL ABOUT DRUGS
- ORGANIC CHEMISTRY INTERNATIONAL
- GOOGLE PLUS
- Drug Scaleup and Manufacturing International
amcrasto@gmail.com
email me if u like my posts
Cenicriviroc in Phase 2 for HIV by Takeda/Tobira
![]()
Cenicriviroc
TAK-652; TBR-652
(-)-(S)-8-[4-(2-Butoxyethoxy)phenyl]-1-isobutyl-N-[4-(1-propyl-1H-imidazol-5-ylmethylsulfinyl)phenyl]-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide
(S)–(−)-8-{4-[2-(Butoxy)ethoxy]phenyl}-1-isobutyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide methanesulfonate
497223-25-3 , Molecular Formula: C41H52N4O4S Molecular Weight: 696.94098
497223-28-6 (mesylate) C41 H52 N4 O4 S . C H4 O3 S, 793.047
Cenicriviroc (TAK-652, TBR-652) is an experimental drug candidate for the treatment of HIV infection.[1] It is being developed by Takeda Pharmaceutical and Tobira Therapeutics.
TBR-652 (formerly TAK-652) is a highly potent and orally active CCR5 antagonist in phase II clinical trials at Takeda for the treatment of HIV infection. Tobira Therapeutics is evaluating the compound in preclinical studies for the treatment of rheumatoid arthritis.
TBR-652 binds CCR5 receptors to interfere with the entry of the HIV-1 virus into macrophages and activated T-cells by inhibiting fusion between viral and cellular membranes. This mechanism of action differs from currently used HIV treatments such as nucleoside reverse transcriptase inhibitors and protease inhibitors.
In 2007, Takeda entered into an agreement with Tobira pursuant to which Tobira obtained exclusive worldwide rights to develop, manufacture and commercialize TBR-652 for the treatment of HIV infection.
Cenicriviroc is an inhibitor of CCR2 and CCR5 receptors,[2] allowing it to function as an entry inhibitor which prevents the virus from entering into a human cell. Inhibition of CCR2 may have an anti-inflammatory effect.
A double-blind, randomized, placebo-controlled clinical study to assess the antiviral activity, safety, and tolerability of cenicriviroc was conducted in 2010. HIV-infected patients taking cenicriviroc had significant reductions in viral load, with the effect persisting up to two weeks after discontinuation of treatment.[3] Additional Phase II clinical trials are underway.[4]
Phase IIb data presented at the 20th Conference on Retroviruses and Opportunistic Infections (CROI) in March 2013 showed similar viral suppression rates of 76% for patients taking 100 mg cenicriviroc, 73% with 200 mg cenicriviroc, and 71% with efavirenz. Non-response rates were higher with cenicriviroc, however, largely due to greater drop-out of patients. A new tablet formulation with lower pill burden may improve adherence. Looking at immune and inflammatory biomarkers, levels of MCP-1 increased and soluble CD14 decreased in the cenicriviroc arms.[5]
Although HIV has been largely rendered a chronic infection, there remains a need for new drugs because of the virus’s propensity to develop resistance to the drugs used to keep it at bay.
Pfizer’s maraviroc was the first drug that acted on the cells to prevent viral entry by antagonising the CCR5 co-receptor. Several others have been investigated and have failed; another that is undergoing clinical trials is Takeda’s cenicriviroc, which has been licensed to Tobira Therapeutics. Unlike maraviroc, the new agent also acts at the CCR2 co-receptor, which is implicated in cardiovascular and metabolic diseases.
In a Phase I double blind, placebo controlled trial designed to study safety, efficacy and pharmacokinetics, treatment-experienced but CCR5 antagonist-naïve patients with HIV-1 were given doses of 25, 50, 75, 100 or 150mg of the drug, or placebo once a day for 10 days.2 The maximum median reductions in HIV-1 RNA values were 0.7, 1.6, 1.8 and 1.7 log10 copies/ml for the respective doses, with a median time to nadir of 10 to 11 days. The effect on CD-4 cell counts was negligible. There was also a significant reduction in levels of monocyte chemotactic protein 1, suggesting that CCR2 was also being blocked. The drug was both generally safe and well tolerated, and no patients withdrew from the trial due to adverse events.
In another Phase I trial, designed to look at pharmacokinetics and pharmacodynamics and carried out in a similar patient population, subjects were given the drug as oral monotherapy for 10 days, again in doses of 25, 50, 75, 100 and 150mg, or placebo.3 The drug was well absorbed into the systemic circulation, and the concentration levels declined slowly, with meant elimination half-lives of one to two days. Potent, dose-dependent reductions in viral load were seen, and again it was generally safe and well tolerated across all levels.
In June 2011, Tobira initiated a multi-centre, double blind, double dummy, 48-week comparative Phase IIb trial in 150 patients with HIV-1 infection. Subjects are being given 100 or 200mg once-daily doses of the drug to evaluate its efficacy, safety and tolerability.
PATENTS
WO 2003014105
WO 2003076411
WO 2005116013
WO 2007144720
WO 2011163389
US 20130079233
WO 2013167743
See also
ancriviroc (formerly known as SCH-C), vicroviroc which has the chemical name (4,6-dimethylprymidine-5-yl){4- [(3S)-4-{(1 R)-2-methoxy-1 -[4-(trifluoromethyl)phenyl]ethyl}-3-methylpiperazin-1 -yl]-4-methylpiperidin-1 – yljmethanone, PRO-140, apliviroc (formerly known as GW-873140, Ono-4128, AK-602), AMD-887, INC- B9471 , CMPD-167 which has the chemical name N-methyl-N-((1R,3S,4S)-3-[4-(3-benzyl-1-ethyl-1H- pyrazol-δ-yOpiperidin-i-ylmethylH-IS-fluorophenyllcyclopent-i-yll-D-valine), methyl1-endo-{8-[(3S)-3- (acetylamino)-3-(3-fluorophenyl)propyl]-8-azabicyclo[3.2.1]oct-3-yl}-2-methyl-4,5,6,7-tetrahydro-1 H- imidazo[4,5-c]pyridine-5-carboxylate, methyl 3-endo-{8-[(3S)-3-(acetamido)-3-(3-fluorophenyl)propyl]-8- azabicyclo[3.2.1]oct-3-yi}-2-methyl-4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridine-5-carboxylate, ethyl 1- endo-{8-[(3S)-3-(acetylamino)-3-(3-fiuorophenyl)propyl]-8-azabicyclo[3.2.1]oct-3-yl}-2-methyl-4,5,6,7- tetrahydro-1 H-imidazo[4,5-c]pyridine-5-carboxylate and N-{(1S)-3-[3-endo-(5-lsobutyryl-2-methyl-4,5,6,7- tetrahydro-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.2.1]oct-8-yl]-1-(3-fluorophenyl)propyl}acetamide) and pharmaceutically acceptable salts, solvates or derivatives of the above. The last four compounds are disclosed in WO 03/084954 and WO 05/033107.


http://pubs.acs.org/doi/full/10.1021/jm0509703

Compound (S)-(−)-5b (TAK-652) also inhibited the replication of six macrophage-tropic (CCR5-using or R5) HIV-1 clinical isolates in peripheral blood mononuclear cells (PBMCs) (mean IC90 = 0.25 nM).
(S)–(−)-8-{4-[2-(Butoxy)ethoxy]phenyl}-1-propyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide ((S)–(−)-5a). The 1 N HCl (160 mL) was added to 1931 (35.68 g, 53.4 mmol), and the mixture was extracted with EtOAc. To the aqueous layer was added 25% aqueous K2CO3 (160 mL), and the mixture was extracted with a mixture of EtOAc and i-PrOH (4:1). The organic layer was washed with brine, dried over MgSO4, and concentrated in vacuo to give (S)-18. To a solution of 16a (18.0 g, 41.1 mmol) and DMF (0.5 mL) in THF (180 mL) was added thionyl chloride (SOCl2) (4.50 mL, 61.7 mmol) at room temperature. After being stirred at room temperature for 1.5 h, the reaction mixture was concentrated in vacuo. A solution of the residue in THF (200 mL) was added dropwise to a mixture of (S)-18 and triethylamine (Et3N) (35.0 mL, 251 mmol) in THF (150 mL) under ice cooling. After being stirred at room temperature for 4 h, water was added to the reaction mixture. The mixture was washed with 10% aqueous AcOH, saturated aqueous NaHCO3, and brine, dried over MgSO4, and concentrated in vacuo. The residue was purified by column chromatography on a NH silica gel (hexane/EtOAc = 1:5 → 1:8 → 1:9) to give 21.14 g (75%) of (S)-(−)-5a as a yellow amorphous powder, [α]D = −132.5° (C = 0.507%, EtOH). 1H NMR (300 MHz, CDCl3) δ 0.87−1.03 (9H, m), 1.34−1.49 (2H, m), 1.50−1.85 (8H, m), 2.55−2.65 (2H, m), 3.15−3.25 (2H, m), 3.52−3.58 (4H, m), 3.75−3.83 (4H. m), 4.02 (1H, d, J = 13.8 Hz), 4.08−4.17 (3H, m), 6.56 (1H, d, J = 1.0 Hz), 6.80 (1H, d, J = 8.8 Hz), 6.96 (2H, d, J = 8.8 Hz), 7.31−7.46 (7H, m), 7.55 (1H, s), 7.76 (2H, d, J = 8.8 Hz), 7.98 (1H, s). Anal. (C40H50N4O4S·0.25H2O) C, H, N.
(S)–(−)-8-{4-[2-(Butoxy)ethoxy]phenyl}-1-isobutyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide methanesulfonate ((S)–(−)-5b). The free base of (S)-(−)-5b was prepared in 80% yield from 16band 19 by a method similar to that described for (S)-(−)-5a. To a solution of the free base of (S)-(−)-5b (64.91 g, 93.1 mmol) in EtOAc (600 mL) was added dropwise a solution of methanesulfonic acid (8.95 g, 93.1 mmol) in EtOAc (160 mL) at room temperature. After being stirred at room temperature for 4 h, the crystals were collected by filtration and washed with EtOAc to give 69.09 g (94%) of (S)-(−)-5b as yellow crystals. The crystals (68.0 g) were purified by recrystallization from 2-butanone to give 58.9 g (85%) of (S)-(−)-5b as yellow crystals, mp 145.5−147.5 °C, [α]D = −191.2° (c = 0.508%, EtOH). 1H NMR (300 MHz, DMSO-d6) δ 0.82−0.97 (12H, m), 1.29−1.39 (2H, m), 1.40−1.55 (4H, m), 1.65−1.85 (2H, m), 2.00−2.25 (1H, m), 2.29 (3H,s), 2.38−2.60 (2H, m), 3.10 (2H, d, J = 7.8 Hz), 3.30−3.60 (4H, m), 3.70 (2H, t, J = 4.8 Hz), 3.98 (2H, t,J = 6.6 Hz), 4.10 (2H, t, J = 4.8 Hz), 4.34 (1H, d, J = 15.0 Hz), 4.68 (1H, d, J = 15.0 Hz), 6.87 (1H, d, J = 8.7 Hz), 6.99 (2H, d, J = 8.7 Hz), 7.16 (1H, s), 7.42−7.60 (8H, m), 7.93 (2H, d, J = 8.7 Hz), 9.05 (1H, s), 10.18 (1H, s). Anal. (C42H56N4O7S2) C, H, N.
…………………
WO 2003014105 OR US20090030032
http://www.google.st/patents/US20090030032?hl=pt-PT&cl=un
EXAMPLE 7 Preparation of Compounds 9 and 10
8-[4-(2-Butoxyethoxy)phenyl]-1-propyl-N-[4-[[[1-propyl-1H-imidazol-5-yl]methyl]sulfinyl]phenyl]-1,2,3,4-tetrahydro-1-benzazocin-5-carboxamide (317 mg) was resolved by using CHIRAKCEL OJ 50 mm ID×500 mL (hexane/ethanol) to give (−)-8-[4-(2-butoxyethoxy)phenyl]-1-propyl-N-[4-[[[1-propylimidazol-5-yl]methyl]sulfinyl]phenyl]-1,2,3,4-tetrahydro-1-benzoazocine-5-carboxamide (142 mg) (Compound 9) and (+)-8-[4-(2-butoxyethoxy)phenyl]-1-propyl-N-[4-[[[1-propylimidazol-5-yl]methyl]sulfinyl]phenyl]-1,2,3,4-tetrahydro-1-benzoazocine-5-carboxamide (143 mg) (Compound 10).
Compound 9
[α]D=−127.4° (C=0.533% in ethanol).
Compound 10
[α]D=+121.0° (C=0.437% in ethanol).
………………………….
WO 2003076411
http://www.google.st/patents/WO2003076411A1?cl=en
http://www.google.st/patents/US20050107606?hl=pt-PT&cl=en

Example 21 (−)-8-[4-(2-Butoxyethoxy)phenyl]-1-isobutyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide
To a solution of 8-[4-(2-butoxyethoxy)phenyl]-1-isobutyl-1,2,3,4-tetrahydro-1-benzazocine-5-carboxylic acid (45 g) in tetrahydrofuran (135 ml) was added N,N-dimethylformamide (230 mg) and added dropwise thionyl chloride (12.45 g) at 10 to 15° C., and the resulting solution was stirred at the same temperature for 40 minutes to prepare an acid chloride.
Separately, to a solution of (−)-4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenylamine in tetrahydrofuran (270 ml) was added pyridine (27.59 g), the resulting mixture was adjusted to 5° C. or lower, and then thereto was added dropwise the acid chloride solution at 5° C. or less, and the resulting mixture was stirred at the same temperature for 2 hours. To the mixture were added water (270 ml) and 20% aqueous citric acid solution (180 ml), tetrahydrofuran was distilled off under reduced pressure and the residue was extracted with ethyl acetate. The extract was sequentially washed with water, saturated sodium bicarbonate solution and water, and then the solvent was distilled off. To the residue was added ethyl acetate (360 ml), added heptane (360 ml) at 40° C. and added seed crystals of (−)-8-[4-(2-butoxyethoxy)phenyl]-1-isobutyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide (10 mg), and the mixture was stirred at 25° C. for 2 hours and stirred at 5° C. for 1 hour. The precipitated crystals were collected by filtration to obtain 63.97 g (yield: 92.1%) of the title compound. Melting point: 120-122° C.
Elemental analysis value: in terms of C41H52N4O4S
Calcd. value: C, 70.66; H, 7.52; N, 8.04.
Analytical value: C, 70.42; H, 7.52; N, 8.01
Industrial Applicability
According to the present invention, an optically active sulfoxide derivative having CCR5 antagonism or an intermediate compound thereof can be prepared without causing side reactions such as racemization and Pummerer rearrangement. In particular, Process 7 is industrially advantageous since it is possible to prepare an optically active Compound (II) by asymmetric oxidization in the presence of an optically active acid.
Example 20 (−)-8-[4-(2-Butoxyethoxy)phenyl]-1-propyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide.methanesulfonate
According to the same method as that described in Example 15, the title compound was produced from 8-[4-(2-butoxyethoxy)phenyl]-1-propyl-1,2,3,4-tetrahydro-1-benzazocine-5-carboxylic acid and (−)-4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenylamine.
1H-NMR (CDCl3, δ, 300 MHz) 0.88-1.01 (9H, m), 1.37-1.42 (2H, m), 1.57-1.80 (8H, m), 2.63 (2H, br), 2.77 (3H, s), 3.27 (2H, br), 3.51-3.57 (4H, m), 3.77-3.86 (4H, m), 3.90-4.05 (1H, m), 4.14 (2H, t, J=4.6 Hz), 4.25 (1H, d, J=14.6 Hz), 6.73 (1H, s), 6.84 (1H, d, J=8.7 Hz), 6.93 (2H, d, J=8.8 Hz), 7.21 (2H, d, J=8.7 Hz), 7.40-7.48 (4H, m), 7.61 (1H, s), 7.89 (2H, d, J=8.7 Hz), 8.65 (1H, s), 9.27 (1H, br)
Elemental analysis value: in terms of C41H54N4O7S2
Calcd. value: C, 63.21; H, 6.99; N, 7.19; S, 8.23.
Analytical value: C, 63.00; H, 7.09; N, 7.41; S, 8.25
Example 15 (−)-8-[4-(2-Butoxyethoxy)phenyl]-1-isobutyl-N-(4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenyl)-1,2,3,4-tetrahydro-1-benzazocine-5-carboxamide.methanesulfonate
8-[4-(2-Butoxyethoxy)phenyl]-1-isobutyl-1,2,3,4-tetrahydro-1-benzazocine-5-carboxylic acid (986 mg) was dissolved in tetrahydrofuran (3 ml) and thereto was added N,N-dimethylformamide (one drop). Subsequently, to the resulting solution was added dropwise oxalyl chloride (0.2 ml, 2.29 mmol) under ice-cooling and the mixture was stirred for 80 minutes under ice-cooling to prepare an acid chloride.
Separately, (−)-4-{[(1-propyl-1H-imidazol-5-yl)methyl]sulfinyl}phenylamine (689 mg) was added to tetrahydrofuran (7 ml) and the resulting solution was cooled to 5° C. To the solution was added dropwise pyridine (0.62 ml) and added dropwise the acid chloride solution at 3 to 5° C., and the mixture was stirred for 2 hours under ice-cooling. To the mixture was added water (20 ml) at 10° C. or lower and the mixture was extracted with ethyl acetate. The organic layer was sequentially washed with water, saturated sodium bicarbonate solution and water, and concentrated under reduced pressure. Thereto was added toluene and the mixture was concentrated under reduced pressure. Thereto was added acetonitrile and the mixture was concentrated under reduced pressure. The residue was dissolved in acetonitrile (7 ml) and acetone (7 ml), thereto was added dropwise methanesulfonic acid (209 mg), and added seed crystals and the mixture was stirred at room temperature for 100 minutes. Subsequently, to the mixture was added acetone-acetonitrile (1:1, 5 ml). After stirring at room temperature overnight, the mixture was stirred for 2.5 hours under ice-cooling. The precipitated crystals were collected by filtration and washed with the ice-cooled acetone (9 ml). The crystals were dried at 40° C. under reduced pressure to obtain 1.51 g (yield: 87%) of the title compound as yellow crystals.
1H-NMR (300 MHz, DMSO-d6, δ): 0.78-0.96 (12H, m), 1.25-1.40 (2H, m), 1.41-1.51 (4H, m), 1.65-1.85 (2H, m), 2.05-2.15 (1H, m), 2.30 (3H, s), 2.35-2.50 (2H, m), 3.05-3.15 (2H, m), 3.30-3.55 (4H, m), 3.65-3.70 (2H, m), 3.90-4.05 (2H, m), 4.05-4.10 (2H, m), 4.30 (1H, d, J=14.73 Hz), 4.65 (1H, d, J=14.73 Hz), 6.85 (1H, d, J=8.97 Hz), 6.97 (1H, d, J=8.79 Hz), 7.17 (1H, s), 7.35-7.75 (6H, m), 7.92 (2H, d, J=8.79 Hz), 9.08 (1H, s), 10.15 (1H, s).
Elemental analysis value: in terms of C41H52N4O4S.CH4SO3
Calcd. value: C, 63.61; H, 7.12; N, 7.06; S, 8.09.
Found value: C, 63.65; H, 7.23; N, 7.05; S, 8.08.
………………………….
References
- Klibanov, Olga M.; Williams, Shannon H.; Iler, Cameron A (2010). “Cenicriviroc, an orally active CCR5 antagonist for the potential treatment of HIV infection”. Current Opinion in Investigational Drugs 11 (8): 940–950. PMID 20721836.
- Baba, Masanori; Takashima, Katsunori; Miyake, Hiroshi; Kanzaki, Naoyuki; Teshima, Koichiro; Wang, Xin; Shiraishi, Mitsuru; Iizawa, Yuji (2005). “TAK-652 inhibits CCR5-mediated human immunodeficiency virus type 1 infection in vitro and has favorable pharmacokinetics in humans”. Antimicrobial Agents and Chemotherapy 49 (11): 4584–4591. doi:10.1128/AAC.49.11.4584-4591.2005. PMC 1280155. PMID 16251299.
- C. Reviriego (2011). Drugs of the Future 36 (7): 511–517. doi:10.1358/dof.2011.36.7.1622066.
- “Tobira Therapeutics Initiates Phase 2b Trial of Cenicriviroc”. The Body. July 5, 2011.
- CROI 2013: CCR5/CCR2 Inhibitor Cenicriviroc Has Both Anti-HIV and Anti-inflammatory Effects. Highleyman, Liz. HIVandHepatitis.com. 7 March 2013.
|
11-26-2012
|
Chemokine receptor antagonists.
|
Journal of medicinal chemistry
|
|
|
6-1-2011
|
Safety, efficacy, and pharmacokinetics of TBR-652, a CCR5/CCR2 antagonist, in HIV-1-infected, treatment-experienced, CCR5 antagonist-naive subjects.
|
Journal of acquired immune deficiency syndromes (1999)
|
|
|
8-1-2010
|
Cenicriviroc, an orally active CCR5 antagonist for the potential treatment of HIV infection.
|
Current opinion in investigational drugs (London, England : 2000)
|
|
|
3-1-2009
|
The relative activity of “function sparing” HIV-1 entry inhibitors on viral entry and CCR5 internalization: is allosteric functional selectivity a valuable therapeutic property?
|
Molecular pharmacology
|
|
|
2-1-2007
|
Isolation and characterization of human immunodeficiency virus type 1 resistant to the small-molecule CCR5 antagonist TAK-652.
|
Antimicrobial agents and chemotherapy
|
|
|
9-10-2006
|
[Progress in AIDS therapy].
|
Nihon Naika Gakkai zasshi. The Journal of the Japanese Society of Internal Medicine
|
|
|
3-23-2006
|
Highly potent and orally active CCR5 antagonists as anti-HIV-1 agents: synthesis and biological activities of 1-benzazocine derivatives containing a sulfoxide moiety.
|
Journal of medicinal chemistry
|
|
|
11-1-2005
|
TAK-652 inhibits CCR5-mediated human immunodeficiency virus type 1 infection in vitro and has favorable pharmacokinetics in humans.
|
Antimicrobial agents and chemotherapy
|
|
|
1-27-2005
|
Stereoselective synthesis of [L-Arg-L/D-3-(2-naphthyl)alanine]-type (E)-alkene dipeptide isosteres and its application to the synthesis and biological evaluation of pseudopeptide analogues of the CXCR4 antagonist FC131.
|
Journal of medicinal chemistry
|
|
|
1-1-2005
|
TAK-652, a novel CCR5 inhibitor, has favourable drug interactions with other antiretrovirals in vitro.
|
Antiviral therapy
|
……………….
Chemical structures of selected small molecule CCR5 inhibitors. A. Maraviroc (MVC, Selzentry), B. Vicriviroc (VCV), C. Cenicriviroc (TBR-652), D. PF-232798.

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO
.....






































































