AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

1R,2S-Methoxamine

 Uncategorized  Comments Off on 1R,2S-Methoxamine
Apr 212016
 

1R,2S-methoxamine, also known as L-erythro-methoxamine

CAS 13699-29-1

Benzenemethanol, α-​[(1S)​-​1-​aminoethyl]​-​2,​5-​dimethoxy-​, (αR)​-
Benzenemethanol, α-(1-aminoethyl)-2,5-dimethoxy-, [R-(R*,S*)]-
(-)-Methoxamine
Molecular Weight, 211.26, C11 H17 N O3

HYDROCHLORIDE

(1R,2S)-isomer HCl salt of 1 -(2,5-dimethoxyphenyl)-2-amino-1 -propanol also called as (1R, 2S)methoxamine hydrochloride

CAS  16122-04-6

Used as a pressor agent, as a vasoconstrictor, as a nasal decongestant, in ophthalmology and also found very effective in the treatment of faecal incontinence.

treatment of relief of fecal incontinence and anal itch (pruritis ani) , particularly for patients who have had a major bowel resection and reanastomosis .

Anal or fecal incontinence is the inability to voluntarily control the passage of feces or gas through the anus. It may occur either as fecal soiling or as rare episodes of incontinence for gas or watery stools. It is a very distressing condition that can result in self-inflicted social isolation and despair.

Conventional treatments for fecal incontinence include drug therapy to improve stool consistency, such as morphine, loperamide and codeine phosphate to reduce gut motility, and laxatives to soften stools and relieve constipation. Biofeedback training is another treatment which involves muscle strengthening exercises to improve anal canal resting pressure, and squeeze pressure, and to teach symmetry of anal canal function. The most common form of treatment however, is surgical repair, such as the creation of a neo-sphincter which involves grafting on muscle from other parts of the anus, or a colostomy. (Gastroenterology in Practice, Summer 1995, pl8- 21; Dig Dis 1990; 8:179-188; and The New England Journal of Medicine, April 1992, pl002-1004) . In mild cases of anal leakage, the patient will often try and plug the anus with a ball of cotton wall.

In Gut, 1991, 32, p.345-346 it was reported that two thirds of patients with idiopathic faecal incontinence had a decreased anal resting pressure resulting from an abnormal internal sphincter function. In many incontinent patients, the internal anal sphincter was found to be abnormally thin, while others had an external anal sphincter defect. It has also been reported that in vi tro contractile response of the internal anal sphincter to noradrenaline is decreased in incontinence, (Br. J. Surg. 1992, vol 79, August, p829-832; Digestive Diseases and Sciences, vol 38, no. 11, Nov. 1993, pl961-1969) . A further discussion of the innervation and control of the internal anal sphincter and drugs which can increase or decrease the normal anal resting pressure, is discussed in the text book Coloproctology and the Pelvic Floor (Butterworths) , second edition, 1992, at chapter 3 p37-53; Automic Control of Internal Anal Sphincter; and Journal of Clinical Investigation 1990, 86: p424-429.

In Surgery 1990; 107: p311-315 sodium valproate was found to be useful in the treatment of minor incontinence after ileoanal anastomosis.

It has now surprisingly been found that fecal incontinence and anal itch can be resolved by treatment with α adrenergic agonists, nitric oxide synthase inhibitors, prostaglandins F, dopamine, morphine, β-blockers such as propranolol, and 5-Hydroxytryptamine (5-HT) .

This is surprising since it was always thought that once an anal sphincter began functioning abnormally, the patient would require major surgery.

In this way the anal leakage is reduced or eliminated without the patient having to undergo major surgery.

Accordingly in a first aspect of the invention there is provided use of a physiologically active agent selected from an α adrenergic agonist, nitric oxide synthase inhibitor, prostaglandin F, dopamine, morphine, β-blockers, and 5- Hydroxytryptamine in the preparation of a medicament for the treatment or prophylaxis of fecal incontinence or anal itch.

The agents of the invention appear to at least partially treat the incontinence by increasing the resting pressure of the internal anal sphincter. Preferred agents are λ adrenergic agonists, nitric oxide synthase inhibitors, and prostaglandins F.

Examples of suitable aλ adrenergic agonists are nor- adrenalin, methoxamine, but particularly preferred is phenylephrine .

Examples of suitable F prostaglandin are dinoprost and carboprost.

Examples of suitable NO synthase inhibitors are

NG-monnoommeetthhyyll–LL–aarrggiinn:ine (L-NMMA) , and NG-nitro-L-arginine methyl ester ( -NAME)

The medicament can contain a single active agent or a combination of any of the above active agents.

Nitric Oxide (NO) synthase inhibitors such as LNMMA have previously been suggested for the therapeutic treatment of septic shock.

The prostaglandins, along with thromboxanes and leukotrienes are all derived from 20 -carbon polyunsaturated fatty acids and are collectively termed eicosanoids. F prostaglandins are derived in vivo from the endoperoxide prostaglandin H2which is in turn derived from leukotrienes. Clinically, F prostaglandins such as dinoprost and carboprost are used as uterine stimulants in the termination of pregnancy, missed abortion or the induction of labour.

Phenylephrine (an αx adrenergic agonist) is used as a mydriatic in ophthalmology, and as a decongestant , for example, in cold and flu remedies.

However there has been no suggestion to the inventors knowledge of using any of these active agents to treat fecal incontinence or anal itch. As used herein “fecal incontinence” includes all types of anal leakage from minor leakage or ‘spotting’ through moderate leakage, to major instances of faecal incontinence, and includes neurogenic, active, urge and passive incontinence.

More particularly the class of incontinent patients who will benefit most from the present invention are those with idiopathic incontinence and those whose incontinence is at least partly due to a weakness of either the internal or external anal sphincter, especially those with a normal or low maximum anal pressure and a structurally intact internal anal sphincter muscle, such as with an abnormally thin sphincter. However patients with minor structural damage such as a fragmented sphincter would still benefit from the invention. Not only incontinent patients with a damaged or abnormal internal sphincter can be treated, but also patients with a damaged or abnormal external sphincter since the increase in the internal anal resting tone induced by the invention will compensate for a poorly functioning external sphincter.

Another class of patients who particularly benefit from the invention are post-surgical patients who have had major bowel resection and reanastomosis . For example patients with ileoanal pouch (restorative proctocolectomy) , coloanal (with or without colonic pouch) anostomosis, lower anterior resection, and colectomy with ileorectal anastomosis.

The damage to the sphincter could be caused by trauma, such as experienced in child birth, surgical operations, or road traffic accidents. Furthermore it is also believed that incontinence caused by primary internal anal degeneration can also be relieved by the invention.

Anal leakage also often leads to pruritis of the anus and therefore by reducing or eliminating the leakage, the pruritis or anal itch is also relieved or prevented. Furthermore, as a result of the increased anal resting pressure, the patient no longer has the discomfort of distended anal sphincter muscles.

Methoxamine contains two chiral carbons and thus exists in four isomeric forms. Of all the isomeric forms, the studies revealed (1R,2S)- isomer to be therapeutically active.

US patent 2359707 describes the process for the synthesis of racemic β-(2,5-dimethoxy phenyl)-P-hydroxy-isopropyl amine in neutral, acid salt and its derivative from 2,5- dimethoxy propiophenone by treatment with methylnitrite in diethyl ether medium to obtain 2,5-dimethoxy-a-isonitrosopropiophenone hydrochloride. It is further reduced with palladium on carbon to yield β-(2,5-dimethoxyphenyl)-p-ketoisopropylamine hydrochloride and then with platinum black to get p-(2,5-dimethoxyphenyl)-β- hydroxyisopropyl amine hydrochloride. The described process for di-methoxamine HC1 is not cost-effective, due to the use of two expensive catalysts (platinum black and palladium carbon), solvent diethyl ether and involves more number of steps. The other drawback being it is racemic mixture and cannot be used directly as drug. The process described did not specify the quality of the product.

STR1

In US patent 3284490 the processes for racemic N-alkyl derivatives of methoxamine are described from dl-methoxamine.

JP 63165348 describes process for production of optically active l-(2,5- dimethoxyphenyl)-2-aminophenol by resolving racemic compound with the use of optically active L-N-acetylleucine as resolving agent. The disadvantages of the process are less yield, low quality and use of expensive naturally occurring amino acid, which prevents from employing this method on commercial scale.

WO 03/055474 A1 discloses mainly, the use of (1R, 2S)-methoxamine in the treatment of faecal incontinence at low doses without local or systemic side effects when used topically. The patent also described the synthesis of (1R, 2S)-methoxamine, from L- alanine, by protecting the amino group using methylchloroformate, converting carboxy
group of the N-protected alanine into an acid chloride insitu followed by reaction with an amine to produce an N-protected (S)-alanine amide and coupling that compound with a brominated 2,5-dimethoxybenzene in the presence of n-butyllithium or a magnesium based reagent to give (S)-amino-l-(2,5-dimethoxy-phenyl)-l-propanone, the amino group of which is protected .The reduction of the N-protected propanone was carried out using dimethylphenylsilane and the protecting group was removed by treatment with potassium hydroxide. Other method adopted in the patent to isolate (1R,2S)methoxamine is by separation of racemic methoxamine using chiral column.
STR1
The prior art suffers with some of the disadvantages like using n-butyllithium, which is pyrophoric, expensive and causes hazards to commercial scale. Also, the separation of racemic Methoxamine using chiral column mentioned in the patent can be considered for
isolating small quantities of the required isomer for analytical purposes but cannot be adopted on commercial scale for production of the drug.

US Patent 5962737 described stereospecific synthesis of the racemic threo isomers of 2- nitro-1 -phenylpropanols by reacting benzaldehyde derivative with nitroalkane in the presence of a tertiary amine and reducing 2-nitro-l-phenylpropanols with lithium aluminium hydride to 2-amino-l-phenylpropanols. Also described is phase transfer resolution of racemic mixtures of 2-amino-l-phenylpropanol and its derivatives into their optically pure isomers by reacting with the mono alkali metal salt of tartaric acid ester in a two phase system of a hydrocarbon and water. The specification further describes optically pure isomer D-threo 2-amino-( 1 -dialkoxy or alkoxy)phenylpropanol by resolution of dl- threo 2-amino-( 1 -dialkoxy or alkoxy)phenylpropanol by using dibenzoyltartaric acid. The synthesis of the product (lS,2S)-threo 2-amino-(l-dialkoxy or alkoxy) phenyl propanol involves the use of expensive and hazardous chemicals like LAH making the process technically and commercially difficult for implementation.

STR1

Paper

Journal of the American Chemical Society (1984), 106(16), 4629-30

http://pubs.acs.org/doi/pdf/10.1021/ja00328a062

STR1

 

PATENT

http://www.google.com/patents/EP2275099A1?cl=en

EXAMPLE 3Synthesis of 1R,2S-Methoxamine(S)-N-Methoxycarbonyl alanine

To a stirred solution of L-alanine (300g, 3.37 mol sodium hydroxide (1N, 1800 cm3) at 0°C in an ice bath was added dropwise, over 2 hours, methyl chloroformate (274 cm3, 3.54 mol). The pH of the solution was maintained at 9 by the addition of sodium hydroxide (5N). The reaction mixture was stirred at 0°C for 3 hours whereupon it was acidified to pH 1 by the addition of phosphoric acid solution (15%) and extracted with diethyl ether (5 x 1000 cm3). The combined organic extracts were dried (MgSO4) and concentrated under reduced pressure to yield the product as a viscous green oil (386 g, 78%). 1H NMR (250 MHz; C2HCl3) 1.48 (3H, d, J7.25, CH3), 3.72 (3 H, s, COCH3), 4.40 (1 H, quintet, J7.25, CH), 5.31 (1 H, bs, NH).

(S)-N-Methoxycarbonyl alaninedimethylamide

To a stirred solution of MeOC-alanine (227 g, 1.54 mol) and dimethylformamide (DMF) (25 cm3) in dry dichlorourethane (DCM) (2000 cm3) at 0°C was added dropwise oxalyl chloride (146 cm3, 1.62 mol) over a period of 2 hours. The solution was stirred at 0°C until the evolution of gasses ceased whereupon a basic solution of dimethylamine (676 g, 7.70 mol) in NaOH (3 N, 2000 cm3) was added. The aqueous layer was extracted with diethyl ether (2 x 500 cm3) and the combined organic layers dried (MgSO4) and concentrated under reduced pressure to give the product as a white crystalline solid which required no further purification (230 g, 86%). 1H NMR (250 MHz; C2HCl3) 1.33 (3 H, d, J6.75, CH3), 2.99 3 H, s, OCH3) 3.08, (3 H, s, OCH3), 3.66 (3 H, s, COCH3), 4.66 (H, quintet, J7.00, CH), 5.75 (1 H, d, J5.75, NH).

(S)-2-[(Methoxycarbonyl)amino]-1-(2,5-dimethoxyphenyl)-1-propanone.

To a THF (1000 cm3) solution of bromo-2,5-dimethoxybenzene (55 g, 0.25 mol) at -20°C under nitrogen was addedn-butyl lithium (100 cm3, 2.5 M in hexanes, 0.25 mol). The mixture was stirred at -20°C for 0.75 hours, whereupon a THF (100 cm3) solution of amide (30 g, 0.17 mol) was added via cannula. The solution was stirred at -20°C for 2 hours and was then allowed to warm to room temperature over 1 hour and quenched by the addition of ammonium chloride solution (700 cm3). The solution was diluted with diethyl ether (1000 cm3) and the organic layer was dried (MgSO4) and concentrated under reduced pressure to give a yellow oil. The product was purified by dry flash chromatography on silica (eluant 4:1 hexane/ethyl acetate then 3:2 hexane/ethyl acetate) to give the product as a white crystalline solid (45 g, 98%). 1H NMR (250 MHz; C2HCl3) 1.36 (3 H, d, J7.0, CH3), 3.70 (3 H, s, COCH3), 3.82 (3 H, s, OCH3), 3.92 (3 H, s, OCH3), 5.43 (1 H, quintet, J 7.3, H-2), 5.80 (1 H, bs, NH), 6.94 (1 H, d, J 9.0, ArH), 7.10 (1 H, dd, J 9.0, 3.3, ArH), 7.32 (1 H, d, J 3.3, ArH).

(1R,2S)-2-[(Methoxycarbonyl)amino]-1-(2,5-dimethoxyphenyl)-1-propanol.

To a stirred solution of ketone i.e. (S)-2-[(methoxycarbonyl)amino]-1-(2,5-dimethoxyphenyl)-1-propanone (20 g, 74.9 mmol) and dimethylphenyl silane (10.7 g, 78.6 mmol) in dry DCM (500 cm3) at 0°C in an ice bath was added dropwise trithioroacetic acid (TFA) (50 cm3). The solution was stirred at 0°C for 1 h and then quenched by the addition of sodium hydroxide (500 cm3, 1 N). The organic layer was dried and concentrated under reduced pressure to give a yellow oil which solidified on standing. This solid was crystallized from ether/hexane to give the product as a white crystalline solid (15.6 g, 75%).1H NMR (250 MHz; C2HCl3) 1.03 (3 H, d, J7.0, CH3), 3.04 (1 H, d, J4.3, OH), 3.68 (3 H, s, COCH3), 3.78 (3 H, s, OCH3), 3.80 (3 H, s, OCH3), 3.94-3.99 (1 H, m, H-2), 5.05-5.15 (2 H, m, H-1 and NH), 6.72-6.85 (2 H, m, ArH) 6.97 (1 H, d, J 2.0, ArH).

(1,R,2S)-Methoxamine.

To a stirred solution of methoxycarbonyl (MeOC) protected alcohol i.e. (1R,2S)-2-[(methoxycarbonyl)amino]-1-(2,5-dimethoxyphenyl)-1-propanol (4.0 g, 14.9 mmol) in methanol (175 cm3) was added a solution of KOH (4.06 g, 72.8 mmol in water (60 cm3). The solution was cooled and acidified with phosphoric acid (15% v/v). The solution was extracted with DCM (2 x 50 cm3) and the aqueous layer basified by the addition of K2CO3. The aqueous layer was extracted with diethyl ether (5 x 50 cm3) and the combined ethereal extracts dried (MgSO4) and concentrated under reduced pressure to give the product as a clear yellow oil (1.9 g, 61%), 1H NMR (250 MHz; C2HCl3) 0.84 (3 H, d, J 7.0, CH3), 3.19-3.22 (1 H, m, H-2), 3.71 (6 H, s, 2 x OCH3), 4.67 (1 H, d, J 5.0, H-1), 6.66-6.72 (2 H, m, ArH), 6.92 (1 H, d, J 2.5, ArH).

(1R, 2S)-Methoxamine hydrochloride.

To an ice cooled solution of (1R,2S)-methoxamine (1.9 g, 9.00 mmol) in anhydrous diethyl ether (30 cm3) was passed a stream of dry HCl gas for 45 mins. The resultant precipitate was filtered by suction, washed with cold diethyl ether and dried under nitrogen to yield the title compound as a white solid. (1.5 g, 68%). 1H NMR (250 MHz; [C2H3]2SO) 0.89 (3 H, d, J 6.8, CH3), 3.37-3.42 (1 H,m,H-2), 3.71 (3 H, s, OCH3), 3.75 (3 H, s, OCH3), 5.12 (1 H, s, H-1), 5.92 (1 H, d, J 4.3, OH), 6.84 (1 H, dd, J 8.8, 3.0, ArH), 6.92-7.00 (2 H, m, ArH); HPLC.

Analytical Method for the Analysis of Methoxamine

The following method was used to analyse methoxamine samples.

Method

  • Column : Cyclobond I RSP 250 x 4.6 mm
    Column temperature : 23°C
    Mobile phase : 0.1% Tetraethylammonium pH 4.1*
    95%v/v
    : Acetonitrile 5%v/v
    Flow rate : 0.6 ml/min
    Solution
    Concentration :
    5 mg/l
    Injection volume : 2.5 µl to 20 µl
    Detection : UV 230 nm
    *Tetraethylammonium acetate pH 4.1 was prepared fresh daily.

 

Example 2 above allows the complete assignment of the methoxamine isomers as shown below:

Figure imgb0005
Figure imgb0006

PATENT

INDIAN 1020/CHE/2011

BY


The Managing Director of Malladi Drugs & Pharmaceuticals, Prashant Malladi (left), with the Chief Executive Officer, V. N. Gopalakrishnan

 

 

V.N Gopalakrishnan

V.N Gopalakrishnan

CEO at Malladi Drugs & Pharmaceuticals Ltd

Prabhakaran Ranganathan

Prabhakaran Ranganathan

Vice President (Operations) at Malladi Drugs and Pharmaceuticals Limited

The present invention further provides an improved process for the preparation of (JS, 2S)-Methoxamine HC1 of formula (6) from (1R, 2S)-methoxamine by treating with acetic anhydride in toluene medium followed by acid hydrolysis and basification to obtain (IS, 2S)-Methoxamine base which is further acidified to form (1S,2S)- Methoxamine HC1 (6).

The present invention further provides an improved process for the preparation of (1R, 2R)-Methoxamine HC1 of formula (5) from its diastereomer (1S, 2R)-methoxamine HC1 of formula (2) by treating with acetic anhydride in toluene medium followed by acid hydrolysis and basification to obtain (1R, 2R)-Methoxamine base which is further acidified to form (1R, 2R)-Methoxamine HC1 (5).

STR1

STR1

The following examples illustrate the invention.

EXAMPLES

Example 1
Preparation of l-(2,5-Dimethoxyphenyl)propan-l-one (8)
Aluminium chloride (127.4 g; 0.955 mol) was added to dichloromethane (420 mL) in a round bottomed flask under nitrogen atmosphere. The reaction mixture was cooled to -5 °C; 1,4-dimethoxybenzene (100 g; 0.724 mol) was added slowly within 15-30 minutes. Propionic chloride (87 g; 0.94 mol) dissolved in dichloromethane (245 mL) was added slowly within 2 hours. The reaction mass was allowed to stir for 2 hours and then was quenched in crushed ice (1 kilo) and HC1 (75 mL) at 0 – 5 °C. Separated the layers and the organic layer was washed with 5% sodium hydroxide solution, dried and concentrated (140 g; colorless liquid); Purity by HPLC : 99.04%

Spectroscopic interpretation

The structure of the product, l-(2,5-Dimethoxyphenyl)propan-l-one was confirmed with the help of the following spectroscopic data.

a) IR (cm-1) (KBr)
Aromatic C-H stretch at 3071, aliphatic C – H stretch at 2938, C = O stretch at 1674, benzenoid bands at 1609 and 1584, C – O stretch at 1223, C – H out of plane bending of tri-substituted benzene ring at 814,719.

b) 1H NMR(CDCb, 300 MHz) (δH)
1.16 (3H, t, -CH2-CH3), 3.0 (2H, q, -CH2-CH3), 3.78 (3H, s, -OCH3), 3.85 (3H, s, -OCH3), 6.83 – 7.72 (3H, m, aromatic protons)

c) 13C NMR (CDCb, 300 MHz) (δC)
8.44 (-CH2-CH3), 37.03 (-CH2-CH3), 55.74 (-OCH3), 56.01 (-OCH3), 113.09 – 153.41 (aromatic carbons), 202.96 (C=O)

d) Mass spectrum (ESI, methanol)
[M+Na]+ at m/z 217 (9), [M+H]+ at m/z 195 (100).

Example 2
Preparation of l-(2,5-Dimethoxyphenyl)-2-nitrosopropan-l-one (9) l-(2,5-Dimethoxyphenyl)propan-l-one (100 g; 0.515 mol) was added to dichloromethane (660 mL) in a round bottomed flask under nitrogen atmosphere. Butylnitrite (46.6 g; 0.52 mol) was slowly added in about 30 minutes at 30 – 35 °C. Diethyl ether (60.2 mL) was added to the reaction mixture and dry HC1 gas was purged for about 4 hours at 30 – 35 °C. The reaction mass was maintained for 12 hours and then concentrated under vacuum The residue obtained (60 g; Pale yellow crystalline powder); Purity by HPLC: 99.81%; mp: 104-107 °C

Spectroscopic interpretation

The structure of the product, l-(2,5-Dimethoxyphenyl)-2-nitrosopropan-l-one was confirmed with the help of the following spectroscopic data

a) IR (cm1) (KBr)
O-H stretch at 3250 (broad), aromatic C-H stretch at 3024, aliphatic C – H stretch at 2934, C = O stretch at 1688, C = N stretch at 1645, benzenoid bands at 1589 and 1504, C-O stretch at 1231, C-H out of plane bending of tri-substituted benzene ring at 745,702.

b) 1H NMR(CDCb, 300 MHz) (δh)
2.07 (3H, s, -C-CH3), 3.72 (3H, s, -OCH3), 3.76 (3H, s, -OCH3), 6.84-6.99 (3H, m, aromatic protons), 8.89 (1H, bs, OH)

c) 13C NMR (CDCb, 300 MHz) (δC)
9.16 (-C-CH3), 55.81 (-OCH3), 56.34 (-OCH3), 113.09 – 153.27 (aromatic carbons), 157.07 (C=N-OH); 193.32 (CO)

d) Mass spectrum (ESI, methanol) [M+H]+ at m/z 224 (100)

Example 3
Preparation of dl-erythro-methoxamine HC1 (10)
Raney nickel (50 g); iso-propyl alcohol (250 mL) were added to the autoclave. l-(2,5- Dimethoxyphenyl)-2-nitrosopropan-1 -one (100 g; 0.448 mol) was added slowly at 50 – 55 °C by simultaneously purging the flask with hydrogen at 2-3 Kilo pressure. When hydrogen consumption ceases, the catalyst was filtered and the filtrate was concentrated. iso-Propyl alcohol (200 mL) was added to the concentrated mass followed by acidification with HC1 to obtaindl-erythro-methoxamine HC1 (70 g; white crystalline solid)

Spectroscopic interpretation
The structure of the product, dl-erythro-methoxaxmne HC1 was confirmed with the help of the following spectroscopic data.

a) IR (cm1) (KBr)
O-H stretch at 3409, aromatic C-H stretch at 3010, aliphatic C – H stretch at 2914, HN-H str. at 2574 and 2467, benzenoid bands at 1615 and 1569, C-N stretch at 1279, C-O stretch at 1216, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 812.

b) 1H NMR (DMSO-d6, 300 MHz) (δH)
1.0 (3H,d, -CH-CH3), 3.74 (3H, s, -OCH3), 3.77 (3H, s, -OCH3), 4.89 (1H, q, -CH-CH3),6.1 (1H, d, -CH-OH), 6.87-7.01 (3H, m, aromatic protons), 8.06 (3H, bs, HN-H) The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6, 300 MHz) (δc)
14.75 (-CH-CH3), 52.12 (-OCH3), 55.70 (-OCH3), 55.70 (-CH-CH3), 67.25 (CH-OH), 111.89 – 153.16 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H)+ at m/z 212 (100), [M-H2O]+ at m/z 194 (56).

Example 4
Preparation of(JR,2S)-Metboxamine HC1 (1) and (1S, 2R)-Methoxamine HC1 (2) dl-erythro-methoxamine HC1 (117g; 0.47 mol) was dissolved in water (350 mL) at 30-35 °C. The clear solution obtained was basified using 50% sodium hydroxide solution. dl-erythro-Methoxaumne (3) was extracted into dichloromethane (150 mL) and concentrated. Mixture of methanol/DMSO (4:1; 1650 mL) was added and the mass was heated to 50 °C. L-(+)-Tartaric acid (71.1g; 0.47mol) was added slowly and the temperature of the mass was further raised to 70 °C for complete dissolution. The mass was cooled to 35 °C and maintained for 48 hours. (IR,2.S)-Methoxamine tartrate complex (80 g) precipitated was filtered. From the filtrate on concentration was obtained (1S,2R)- methoxamine tartrate complex (82 g) (IR,25)-Methoxamine tartrate complex was added to water (250 mL) at 35 °C, basified to 12 – 13 pH with 50% sodium hydroxide solution. Dichloromethane (200 mL) was added and stirred for 30 min. Separated the org layer, dried over sodium sulphate and concentrated completely under vacuum at 45° C. Iso-Propyl alcohol (150 mL) was added, charcaolized and filtered. The clear filtrate was acidified with 20%IPA HC1 to yield (1R, 2S)-Methoxamine HC1 which was filtered and dried (48 g); White crystalline powder; Purity by HPLC : 100%; Chiral purity : 100 %; mp : 172-175 °C; [α]D: -47.94° (c = 2% in MeOH)

Spectroscopic interpretation

The structure of the product, (1R,2S)-Methoxamine HC1 was confirmed with the help of the following spectroscopic data.

a) IR (cm1) (KBr)
O-H stretch at 3300, aromatic C-H stretch at 3065, aliphatic C-H stretch at 2938, HN-H str. at 2693 and 2580, benzenoid bands at 1609 and 1578, C-N stretch at 1277, C-O stretch at 1217, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 818.

b) 1H NMR (DMSO-d6 300 MHz) (δH)
0.91 (3H,d, -CH-CH3), 3.71 (3H, s, -OCH3), 3.75 (3H, s, -OCH3), 5.14 (1H, m, -CH- NH3+), 5.95 (1H, d, -CH-OH), 6.83-7.01 (3H, m, aromatic protons), 8.25 (3H, bs, HN-H) The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6, 300 MHz) (δC)
II. 44 (-CH-CH3), 49.22 (-OCH3), 55.24 (-OCH3), 55.70 (-CH-CH3), 66.49 (CH-OH),

III. 41 – 153.03 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H]+ at m/z 212 (100), [M-H2O]+ at m/z 194 (15).
(IS, 2i?)-Methoxamine tartrate complex was added to water (275 mL) at 35 °C, basified

to 12 – 13 pH with 50% sodium hydroxide solution. Dichloromethane (250 mL) was added and stirred for 30 min. Separated the organic layer, dried over sodium sulphate and concentrated completely under vacuum at 45 °C. Iso-Propyl alcohol (175 mL) was added, charcaolized and filtered. The clear filtrate was acidified with 20%IPA HC1 to yield (1S, 2R)-Methoxamine HC1 which was filtered and dried (51 g) White crystalline powder; Purity by HPLC : 99.99%; Chiral purity . 100 %; mp . 172-175 °C;[α]D : + 47.9° (c = 2% in MeOH)

Spectroscopic interpretation

The structure of the product, (1S, 2R)-Methoxamine HC1 was confirmed with the help of the following spectroscopic data.

a) m (cm1) (KBr)
O-H stretch at 3265, aromatic C-H stretch at 3059, aliphatic C-H stretch at 2997, HN-H str. at 2658 and 2567, benzenoid bands at 1611 and 1587,
C-N stretch at 1294, C-O stretch at 1217, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 818.

b) 1H NMR (DMSO-d6,300 MHz) (δH)
0.91 (3H,d, -CH-CH3), 3.71 (3H, s, -OCH3), 3.75 (3H, s, -OCH3), 5.14 (1H, m, -CH- NH3+), 5.97 (1H, d, -CH-OH), 6.83-7.01 (3H, m, aromatic protons), 8.19 (3H, bs, HN-H) The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6,300 MHz) (δc)

II. 46 (-CH-CH3), 49.18 (-OCH3), 55.23 (-OCH3), 55.68 (-CH-CH3), 66.45 (CH-OH),

III. 42 – 153.02 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H]+ at m/z 212 (100), [M-H2O]+ at m/z 194 (15).

Example 5
Preparation of dl-threo-methoxamine HC1 (11)
dl-erythro-methoxamine HC1 (120g; 0.48 mol) was dissolved in DM water (500 mL) at 30 – 35 °C and cooled to 10 – 15 °C. The clear solution was basified using 50 % sodium hydroxide solution and extracted in dichloromethane (250 mL). The organic layer was separated and concentrated under vacuum. The residue thus obtained was dissolved in toluene (200 mL) and was added slowly to acetic anhydride (120 g; 1.17mol) at 65 – 70 °C. The reaction mass was maintained under stirring and further cooled to 10 – 20 °C. Conc.Sulphuric acid (57.6g; 0.58mol) was added to the reaction mass slowly by maintaining the reaction mass at 10 – 200 C. The reaction mass was heated to 35 – 400 C for 3 hours and concentrated under vacuum at below 80 °C.

The reaction mass was cooled to 10 – 15 °C and was dissolved in DM water (250 mL). The mass was maintained for 3 h at reflux temperature and again cooled to 10 – 15 °C.

The pH was adjusted to 12 – 13 using 50% sodium hydroxide solution and extracted the d/-threo-Methoxamine base in dichloromethane (250 mL). Separated the organic layer and concentrated under vacuum. The concentrated mass was triturated with iso-Propyl alcohol (150 mL); acidified using 20% HC1 in iso-propyl alcohol. Distilled the iso- propyl alcohol completely to the final traces and acetone (300 mL) was added. The material precipitated, crude dl-threo-methoxamine HC1 was filtered. (85 g) Off white powder; Purity by HPLC: 99.4%; mp: 221-223 °C Spectroscopic interpretation

The structure of the product, di-threo-methoxamine HC1 was confirmed with the help of the following spectroscopic data.

a) IR (cm”1) (KBr)
O-H stretch at 3401, aromatic C-H stretch at 3005, aliphatic C-H stretch at 2924, HN-H str. at 2581 and 2490, benzenoid bands at 1609 and 1578, C-N stretch at 1277, C-0 stretch at 1215, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 802.

b) NMR (DMSO-d6,300 MHz) (δH)
1.2 (3H,d, -CH-CHs), 3.72 (3H, s, -OCH3), 3.75 (3H, s, -OCH3), 4.87 (1H, q, -CH-CH3),6.3 (1H, d, -CH-OH), 6.83-6.99 (3H, m, aromatic protons), 8.03 (3H, bs, HN-H) The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6, 300 MHz) (δC)
14.76 (-CH-CH3), 52.15 (-OCH3), 55.89 (-OCH3), 67.34 (CH-OH), 111.96 – 153.21 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H]+ at m/z 212 (100), [M-H2O]+ at m/z 194 (52).

Example 6
Preparation of (1S,2S)- Methoxamine HC1 (6)
(IR, 2S)-Methoxamine HC1 (120 g; 0.48 mol) was dissolved in DM water (500 mL) at 30 -35 °C and cooled to 10 – 15 °C. The clear solution was basified using 50 % sodium hydroxide solution and extracted in dichloromethane (250 mL). The organic layer was separated and concentrated under vacuum. The residue thus obtained was dissolved in toluene (200 mL) and was added slowly to acetic anhydride (120 g; 1.17 mol) at 65 – 70 °C. The reaction mass was maintained under stirring and further cooled to 10 – 20 °C. Conc.sulphuric acid (57.6 g; 0.58 mol) was added to the reaction mass slowly by maintaining the reaction mass at 10 – 20 °C. The reaction mass was heated to 35 – 40 °C for 3 hours and concentrated under vacuum at below 80 °C.

The reaction mass was cooled to 10-15°C and was dissolved in DM water (250 mL). The mass was maintained for 3 h at reflux temperature and again cooled to 10 – 15 °C. The pH was adjusted to 12-13 using 50% sodium hydroxide solution and extracted the (1S, 2S)-Methoxamine base in dichloromethane (250 mL). Separated the organic layer and concentrated under vacuum The concentrated mass was triturated with iso-Propyl alcohol (150 mL); acidified using 20% HC1 in iso-propyl alcohol. Distilled the iso- propyl alcohol completely to the final traces and acetone (300 mL) was added. The material precipitated, crude (IS, 2S)-methoxamine HC1 was filtered. (86 g); White crystalline powder; Purity by HPLC . 99.8%; Chiral purity : 99.7%; mp : 172-175 °C; [α]D: + 30.739° (c = 2% in MeOH)

Spectroscopic interpretation
The structure of the product, (IS, 2S)-methoxamine HC1 was confirmed with the help of the following spectroscopic data.

a) IR (cm1) (KBr)
O-H stretch at 3356, aromatic C-H stretch at 3080, aliphatic C-H stretch at 2999, HN-H str. at 2641 and 2583, benzenoid bands at 1611 and 1506, C-N stretch at 1302, C-O stretch at 1229, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 812.

b) 1H NMR (DMSO-d6 300 MHz) (δH)
1.04 (3H,d, -CH-CH3), 3.72 (3H, s, -OCH3), 3.75 (3H, s, -OCH3), 4.90 (1H, m, -CH- CH3),6.07 (1H, d, -CH-OH), 6.84-7.01 (3H, d, aromatic protons), 8.15 (3H, bs, HN-H)
The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6, 300 MHz) (δC)
14.75 (-CH-CH3), 52.18 (-OCH3), 55.21 (-OCH3), 55.69 (-CH-CH3), 67.32 (CH-OH), 111.38 -153.01 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H]+ at m/z 212 (100), [M-H2O]+ at m/z 194 (48).

Example 7
Preparation of (1R, 2R)-Methoxamine HC1 (5)
(IS, 2R)Methoxamine HC1 (120g; 0.48 mol) was dissolved in DM water (500 mL) at 30 – 35 °C and cooled to 10 – 15 °C. The clear solution was basified using 50 % sodium hydroxide solution and extracted in dichloromethane (250 mL). The organic layer was separated and concentrated under vacuum. The residue thus obtained was dissolved in toluene (200 mL) and was added slowly to acetic anhydride (120 g; 1.17mol) at 65 – 70 °C. The reaction mass was maintained under stirring and further cooled to 10 – 20 °C. Cone.Sulphuric acid (57.6g; 0.58mol) was added to the reaction mass slowly by maintaining the reaction mass at 10 – 20 °C. The reaction mass was heated to 35 – 40 °C for 3 hours and concentrated under vacuum at below 80 °C.

The reaction mass was cooled tol0-15°C and was dissolved in DM water (250 mL). The mass was maintained for 3 h at reflux temperature and again cooled to 10 – 15 °C. The pH was adjusted to 12-13 using 50% sodium hydroxide solution and extracted the (IR, 2i?)-Methoxamine base in dichloromethane (250 mL). Separated the organic layer and concentrated under vacuum. The concentrated mass was triturated with iso-Propyl alcohol (150 mL); acidified using 20% HC1 in iso-propyl alcohol Distilled the iso- propyl alcohol completely to the final traces and acetone (300 mL) was added. The material precipitated, crude (1R, 2R)-methoxamine HC1 was filtered. (90 g) White crystalline powder; Purity by HPLC: 99.1%, Chiral purity. 100%; mp: 172-175 °C;[α]D: -29.04° (c – 2% in MeOH)

Spectroscopic interpretation

The structure of the product, (1R, 2R)methoxamine HC1 was confirmed with the help of the following spectroscopic data.

a) IR (cm1) (KBr)
O-H stretch at 3356, aromatic C-H stretch at 3078, aliphatic C-H stretch at 2999, HN-H str. at 2619 and 2500, benzenoid bands at 1611 and 1508, C-N stretch at 1302, C-O stretch at 1229, C-H out of plane bending of 1,2,4-tri- substituted benzene ring at 812.

b) 1H NMR(DMSO-d6 300 MHz) (δH)
I. 04 (3H,d, -CH-CHa), 3.72 (3H, s, -OCH3), 3.75 (3H, s, -OCH3), 4.90 (1H, m, -CH- CH3),6.07 (1H, d, -CH-OH), 6.83-7.01 (3H, d, aromatic protons), 8.13 (3H, bs, HN-H) The -OH proton appears to have exchanged with the solvent.

c) 13C NMR (DMSO-d6 300 MHz) (δe)
II. 41 (-CH-CH3), 52.16 (-OCH3), 55.22 (-OCH3), 55.70 (-CH-CH3), 67.32 (CH-OH), III. 39-153.15 (aromatic carbons)

d) Mass spectrum (ESI, methanol)
[M+H]+ at m/z 212 (100), [M-H2O]+ at m/z 194 (44).

 

 

PATENT

http://www.google.com/patents/US8491931

(1,R,2S)-Methoxamine

To a stirred solution of methoxycarbonyl (MeOC) protected alcohol i.e. (1R,2S)-2-[(methoxycarbonyl)amino]-1-(2,5-dimethoxyphenyl)-1-propanol (4.0 g, 14.9 mmol) in methanol (175 cm3) was added a solution of KOH (4.06 g, 72.8 mmol in water (60 cm3). The solution was cooled and acidified with phosphoric acid (15% v/v). The solution was extracted with DCM (2×50 cm3) and the aqueous layer basified by the addition of K2CO3. The aqueous layer was extracted with diethyl ether (5×50 cm3) and the combined ethereal extracts dried (MgSO4) and concentrated under reduced pressure to give the product as a clear yellow oil (1.9 g, 61%), 1H NMR (250 MHz; C2HCl3) 0.84 (3H, d, J 7.0, CH3), 3.19-3.22 (1H, m, H-2), 3.71 (6H, s, 2×OCH3), 4.67 (1H, d, J 5.0, H-1), 6.66-6.72 (2H, m, ArH), 6.92 (1H, d, J 2.5, ArH).

(1R,2S)-Methoxamine hydrochloride

To an ice cooled solution of (1R,2S)-methoxamine (1.9 g, 9.00 mmol) in anhydrous diethyl ether (30 cm3) was passed a stream of dry HCl gas for 45 mins. The resultant precipitate was filtered by suction, washed with cold diethyl ether and dried under nitrogen to yield the title compound as a white solid. (1.5 g, 68%). 1H NMR (250 MHz; [C2H3]2SO) 0.89 (3H, d, J 6.8, CH3), 3.37-3.42 (1H,M,H-2), 3.71 (3H, s, OCH3), 3.75 (3H, s, OCH3), 5.12 (1H, s, H-1), 5.92 (1H, d, J 4.3, OH), 6.84 (1H, dd, J 8.8, 3.0, ArH), 6.92-7.00 (2H, m, ArH); HPLC.

//1R,2S-methoxamine

 

RACEMIC

Methoxamine
Title: Methoxamine
CAS Registry Number: 390-28-3
CAS Name: a-(1-Aminoethyl)-2,5-dimethoxybenzenemethanol
Additional Names: a-(1-aminoethyl)-2,5-dimethoxybenzyl alcohol; 2-amino-1-(2,5-dimethoxyphenyl)-1-propanol; b-hydroxy-b-(2,5-dimethoxyphenyl)isopropylamine; b-(2,5-dimethoxyphenyl)-b-hydroxyisopropylamine; 2,5-dimethoxynorephedrine
Molecular Formula: C11H17NO3
Molecular Weight: 211.26
Percent Composition: C 62.54%, H 8.11%, N 6.63%, O 22.72%
Literature References: a1-Adrenergic agonist. Prepn: Baltzly et al., US 2359707 (1944 to Burroughs Wellcome). Metabolism: A. Klutch, M. Bordun, J. Med. Chem. 10, 860 (1967). Clinical pharmacology: N. T. Smith, C. Whitcher, Anesthesiology 28, 735 (1967); P. D. Snashall et al., Clin. Sci. Mol. Med. 54, 283 (1978). HPLC determn in plasma: I. A. Al-Meshal et al., J. Liq. Chromatogr. 12, 1589 (1989). Therapeutic use: P. M. C. Wright et al., Anesth. Analg. 75, 56 (1992); L. Cabanes et al., N. Engl. J. Med. 326, 1661 (1992). Comprehensive description: A. M. Al-Obaid, M. M. El-Domiaty, Anal. Profiles Drug Subs. 20, 399-431 (1991).
Derivative Type: Hydrochloride
CAS Registry Number: 61-16-5
Trademarks: Vasoxine (Burroughs Wellcome); Vasoxyl (Burroughs Wellcome); Vasylox (Burroughs Wellcome)
Molecular Formula: C11H17NO3.HCl
Molecular Weight: 247.72
Percent Composition: C 53.33%, H 7.32%, N 5.65%, O 19.38%, Cl 14.31%
Properties: Crystals, mp 212-216°. pKa (25°C) 9.2. Very sol in water: One gram dissolves in 2.5 ml water, in 12 ml ethanol. Practically insol in ether, benzene, chloroform. pH of a 2% aq soln between 4.5 and 5.5.
Melting point: mp 212-216°
pKa: pKa (25°C) 9.2
Therap-Cat: Antihypotensive.
Keywords: a-Adrenergic Agonist; Antihypotensive.
Share

GMP Oversight of Medicines Manufacturers in the European Union

 regulatory  Comments Off on GMP Oversight of Medicines Manufacturers in the European Union
Apr 212016
 

 

 

A System of Equivalent Member States, a Coordinating Agency and a Centralized Institution

The regulatory system for supervision of pharmaceutical manufacturers and GMP inspection in the European Union is one of the most advanced in the world. Due to the globalization of pharmaceutical manufacture, it also affects industry, regulators and patients outside the European Union. This system, however, is often poorly understood beyond the EU borders.

What follows is an explanation of the EU system in order to increase awareness and facilitate cooperation on GMP between European Union regulators and those outside the European Union.

The European Union

The European Union includes 28 Member States located in Europe, which are: Austria, Belgium, Bulgaria, Croatia, Cyprus, Czech Republic, Denmark, Estonia, Finland, France, Germany, Greece, Hungary, Ireland, Italy, Latvia, Lithuania, Luxemburg, Malta, Netherlands, Poland, Portugal, Romania, Slovakia, Slovenia, Spain, Sweden, and United Kingdom. The EU total population is about 500 million people.

The European Union operates through a system of supranational independent institutions and intergovernmental negotiated decisions by its Member States. It is a legal entity and can negotiate international agreements on behalf of its Member States. The European Parliament, the Council of the European Union and the European Commission are the three main EU institutions. They produce through the “Ordinary Legislative Procedure” (formerly “co-decision”) the policies and laws that apply throughout the European Union.

The European Union has developed a single market through a standardized system of laws that apply in all its Member States. The same rules and harmonized procedures apply to all the 28 Member States regarding the authorization of medicines and the supervision of safety of medicines.

The EU Regulatory System for Medicines

The EU has developed a regulatory system based on a network of decentralized National Competent Authorities (NCAs) in the Member States, supported and coordinated by a centralized agency, theEuropean Medicines Agency (EMA).

The European Commission’s role is multifaceted and focuses on the following:

  • Right of initiative: To propose new or amending legislation for the pharmaceutical sector
  • Implementation: To adopt implementing measures as well as to ensure and monitor the correct application of EU law
  • Risk management: To grant EU-wide marketing authorizations for centralized products or maximum residue limits on the basis of a scientific opinion of the EMA
  • Supervisory authority: To oversee the activities of the EMA in compliance with the mandate of the EMA, EU law and the EU policy objectives
  • Global outreach: To ensure appropriate collaboration with relevant international partners and to promote the EU system globally

The EMA was created in 1995 to coordinate the existing scientific resources in the EU Member States and is an interface for cooperation and coordination of Member States’ activities with respect to medicinal products. EMA scientific decisions are made through its scientific committees, whose members are chosen on the bases of their scientific expertise and are appointed by the Member States. One of the main roles of EMA is to mobilize scientific resources in the Member States, so that many of its scientific activities are carried out through a large network of scientific experts made available by the Member States.

The system for Marketing Authorisation (MA) of medicines, including the referral procedure, is an example of how the European Commission, the EMA and the Member States cooperate. The EU national, decentralized and mutual recognition MA procedures coexist with the centralized procedure (Table 1).

Table 1 - EMA GMP

The referral procedure is an EU binding mechanism that ensures that the same measures are applied to products subject to national, decentralized and mutual recognition MA procedures. This procedure may be notably invoked when the conditions of authorizations need to be reviewed in the light of quality, safety and efficacy data (Union Interest Referral), when Member States have adopted different decisions regarding products that are authorized in at least two Member States (Divergent Decision Referral) or in the absence of agreement among Member States in the course of the mutual recognition or decentralized authorization procedures (Mutual Recognition and Decentralised Referral). This mechanism involves an opinion from the appropriate EMA committee and results in a decision of the European Commission that is binding for all Member States.

In order to provide for the same level of access to critical medicines to all the patients in the Union, the centralized procedure is mandatory for orphan products, biotechnological products, advanced-therapy products (gene therapy, somatic cell therapy and tissue engineering) and products intended for the treatment of critical therapeutic classes (HIV or AIDS, cancer, diabetes neurodegenerative diseases, auto-immune and other immune dysfunctions, and viral diseases). Veterinary medicines for use as growth or yield enhancers are also in the mandatory scope of the centralized procedure.

A fundamental aspect is that the legislation applicable to pharmaceuticals in the European Union is the same irrespective of the Member State or authorization route of the product, as it is developed at Union level. The same applies to the guidelines in use by assessors and inspectors for the assessment of MA applications and inspections, which are developed by EMA, in cooperation with Member States, through its scientific committees and working groups.

Clinical trials of Investigational Medicinal Products (IMPs) require authorization by each NCA and a favorable opinion by an ethics committee in which the clinical trial takes place and is granted in the form of a Clinical Trial Authorisation (CTA). The assessment for a CTA takes into account the holding of an appropriate authorization for each EU site of manufacture or importation.

The EU System for GMP Supervision of Manufacturers and Inspection

Any manufacturer, no matter where it is located, must comply with GMP if they are to supply products to the EU. There is a single system for GMP supervision of manufacturers which is valid throughout all the EU Member States; this includes authorized medicinal products for human or veterinary use placed on the market and IMPs used in clinical trials. The system is based on two main pillars, the authorization/registration of operators in the supply chain and inspection of those operators to ensure compliance with legal requirements, including compliance with GMP and the requirements in the MA or CTA.

Manufacturers and Importers of Medicinal Products*

Manufacturers and importers of medicinal products located in the EU need to be authorized to carry out their activities. This obligation also applies to manufacturers and importers of products only intended for export and IMPs. The competent authorities of each Member State are responsible for granting the authorizations for these activities occurring within their respective territory.

A condition for grant of a manufacturing or import authorization is that the manufacturers must comply with EU GMP. GMP principles and guidelines are set out in two Directives, one for medicines for human use and the other for medicines for veterinary use. More detailed guidelines have been developed through the work of the GMP and GDP Inspectors Working Group (GMDP IWG) and the European Commission and included in the EU GMP guide, published on the European Commission website.

Inspection of Manufacturers and Importers of Medicinal Products

Manufacturers and importers of medicinal products located in the European Union or manufacturers located in a third country are regularly inspected by an EU competent authority for compliance with EU GMP. The outcome of these inspections must be accepted by all other EU authorities. After every inspection a GMP certificate (positive outcome) or noncompliance report (negative outcome) must be issued by the inspecting authority and entered in the EudraGMDP database, which is accessible by regulators in other countries. Most of this information is also available to the general public.

Inspections of manufacturers are typically requested in order to grant or maintain a manufacturing or import authorization (EU sites) or in the context of assessment, approval and maintenance of an MA (typically sites outside the EU) or CTA. For example, EMA may request that an EU competent authority undertake a preapproval GMP inspection of a site included in a MA application through the Centralised procedure or that an EU competent authority undertake periodic repeated postauthorization surveillance inspections of sites named in centralized MAs, in order to verify ongoing compliance with GMP and that the requirements of the MA are being met.

According to EU legislation, the interval for repeated GMP inspection should be based on risk. As a result, a procedure outlining a risk-based model to frequency of inspections is included in theCompilation of European Union Procedures on Inspections and Exchange of Information.

Manufacturers and Importers of Active Substance**

Manufacturers, importers and distributors of active substance located in the European Union are required to comply with GMP and must be registered to the National Competent Authority of the Member State where they are located.

For active substances manufactured outside the EU and imported, each batch needs to be accompanied by a written confirmation issued by the competent authority of the country where it is produced, confirming, among other things, that GMP at least equivalent to that in place in the European Union has been applied to its manufacture. The competent authority of the exporting country also needs to confirm that any GMP noncompliance arising at the manufacturing site would be communicated to the European Union. The receipt of this noncompliance information is via the EMA.

The requirement for the written confirmation can only be waived if the third country is included by the European Commission, after assessment, in a list of countries with an equivalent system of supervision and inspection or, exceptionally, in order to ensure availability of medicines in the EU market, if a GMP certificate for the site has been issued by an EU competent authority after inspection.

The requirement for written confirmation, introduced from July 2013 by Directive 2011/62/EU (the so called Falsified Medicines Directive), requires that authorities outside of the EU take responsibility for active substances manufactured in their territory, if exported to the EU. This requirement caused some debate before its implementation since there were concerns on its potential to cause shortages in the EU, if the exporting authorities were not willing or able to provide the written confirmations, which turned out not to be the case.

The increased dialogue and mutual understanding between the EU and the authorities of exporting countries was instrumental to ensure a smooth implementation of this requirement. It is a good example of the importance of regulatory cooperation in the current globalized manufacturing and supply environment to the benefit of all.

Inspection of Active Substance Manufacturers

The EU legislation places the responsibility for using active substances manufactured in compliance with GMP on the medicinal product manufacturer or the importer (in case the medicinal product is manufactured outside the European Union). The holder of the manufacturing authorization (medicinal product manufacturer in the European Union or EU importer) must verify the registration status of the manufacturer of the active substance and verify compliance by the manufacturer of active substance with GMP, by conducting audits at the manufacturing site. The holder of the manufacturing authorization shall verify compliance directly or they may use a third party acting under a contract.

Inspections of active substance manufacturers are carried out by EU competent authorities following a risk-based approach, or if there is suspicion of noncompliance.

Furthermore, every application for an MA must include a confirmation that the holder of the manufacturing authorization has verified compliance of the manufacturer of the active substance with principles and guidelines of GMP. The confirmation shall contain a reference to the date of the audit and a declaration by the Qualified Person that the outcome of the audit confirms that the manufacturing complies with GMP principles and guidelines.

 

Inspections of active substance manufacturers may also be organised by the European Directorate for the Quality of Medicines & Healthcare (EDQM) of the Council of Europe, on behalf of the EU. The Council of Europe has 47 members including all EU Member States and it has close cooperation with the EU. EDQM is responsible for developing and maintaining the European Pharmacopoeia.

EDQM issues Certificates of Suitability with the monographs of the European Pharmacopoeia (CEP) that can replace most of the data normally expected in EU MA dossiers for the active substance. In order to issue and maintain these certificates, EDQM runs its own inspection program of active substance manufacturers. Most of the inspections organised by the EDQM are carried out by inspectors from EU inspectorates.

 

The Supervisory Authority

As inspections are carried out by inspectorates of Member States, in order to avoid duplication it is necessary to identify the Member State responsible for supervision and inspection of any manufacturing sites involved in production of active substances and medicines for the EU market. This is achieved through the identification of one or more Supervisory Authority (SA); the SA is the NCA in the EU responsible for the GMP supervision of the site, including granting the manufacturing or import authorization and GMP inspection.

If the manufacturing site is in the EU, the SA is the NCA of the Member State where the site is located. In cases where the manufacturing site is outside the EU, the SA is the NCA of the Member State in which the importer of the product(s) is located. Where products from a manufacturing site located in a country outside the EU are imported in more than one Member State, there may be more than one SA, which cooperate in the supervision of the manufacturing site.

The Qualified Person & Batch Certification Prior to Release

An important feature of the supervision system in place in Europe is the role of Qualified Person (QP). In order to obtain an authorization, EU manufacturers and importers must have at their disposal the services of at least one Qualified Person. The Qualified Person must take responsibility for securing that each batch of medicinal product, manufactured or imported, has been manufactured in accordance with EU GMP, and must certify compliance with GMP and with the relevant MA(s). A batch may only be released by a manufacturer or importer for distribution in the EU after certification by the QP. Member States are empowered to take administrative and disciplinary measures against QPs if they have failed to fulfil their obligations.

Furthermore, imported batches need to undergo a full retest in the EU to ensure the quality of the product in accordance with the MA specification. There-testing requirement is waived if there is an operational Mutual Recognition Agreement in place between the EU and the exporting country.

Consequences of Noncompliance with EU GMP

The discovery of serious GMP noncompliance may have implications not only for the Member State which carries out the inspection but also other, possibly all, Member States as well as international authorities should the active substance or product be supplied to them. A mechanism that ensures a coordinated approach for protection of public and/or animal health is taken throughout the European Union has been developed and is published in the Compilation of European Union Procedures. The objective of this procedure is to achieve a coordinated and harmonized assessment and proportionate supervisory actions to balance the protection of patients and minimize supply disruptions whilst ensuring maximum efficiency and avoiding full parallel reviews on a national level across the European Union.

European legislation provides that manufacturer and import authorizations may be suspended or not granted as a result of noncompliance with GMP. Also, existing MAs for the products affected can be varied (e.g., to delete a certain manufacturing site), not granted or revoked. Urgent measures include prohibition of manufacture, importation or supply, and/or withdrawal of all, or of specific batches from the market.

EudraGMDP

EudraGMDP is a publicly accessible Union database which is a repository of, among other things, manufacturing and import authorizations, GMP certificates and non-compliance reports. After every GMP inspection carried out by an EU competent authority, a GMP certificate (positive outcome) or a noncompliance report (negative outcome) is issued by the inspecting authority and entered in the EudraGMDP database.

The database includes a planning module (only accessible to the relevant regulators) for coordination of inspections planned by EU authorities in countries outside the European Union. Data are entered into the planning module in order to facilitate exchange of information between competent authorities and reduce duplication and ensure the best use of inspectional resources. EMA and EU authorities recognize the global nature of modern pharmaceutical supply chains and the need for close collaboration and cooperation with regulatory authorities outside the European Union and therefore work is ongoing to extend the use of the EudraGMDP database planning module to include exchange of information on inspections planned by authorities outside the European Union.

Overview of Inspection Activities

The chart below shows a summary of the inspections carried out by EEA competent authorities in 2014. Domestic inspections are inspections carried out by EEA competent authorities within the EEA territory. Foreign inspections are inspections carried out by EEA competent authorities outside the EEA. The data are extracted from EudraGMDP.

 

Ensuring and Maintaining Equivalence among Member States Inspectorates

In order to ensure the functioning of the EU system for GMP supervision of manufacturers and inspections described above, it is necessary to ensure that all the National inspectorates in the Member States are equivalent as regards the level of supervision they are able to provide. A number of measures are put in place to ensure that this is the case, summarized below.

Legislation

The pharmaceutical legislation is developed at EU level, mainly in the form of Regulations and Directives. Both are applicable to all the Member States, the difference being that Regulations are directly applicable to the entire EU territory while Directives have to be transposed into national legislation, in a timeframe established in the Directive itself, usually 18 months.

The EU legal framework for medicinal products is intended to ensure a high level of public health protection and to promote the functioning of the EU internal market. The system is also designed to encourage innovation. It is a large body of legislation that ensures extensive harmonization within the European Union, including GMP and inspections. The pharmaceutical legislation is published in the Official Journal of the European Union.

The EU GMP guide

A single GMP guide is in use in the European Union. The guide is referenced in the EU legislation (Directives 2001/83/EC for human products, 2001/82/EC for veterinary products and in clinical trial legislation) and has long since replaced any previously existing national GMP guide. The EU GMP guide provides the standards and requirements used by EU inspectors for any GMP inspections, both in or outside of the European Union.

The guide is subdivided into tree parts and 19 annexes dealing with specific types of manufacture. Part 1 is the GMP for finished products, Part 2 GMP for active substances and Part 3 includes GMP-related documents. The EU GMP guide is harmonized with the PIC/S GMP guidelines on an ongoing basis. EU GMP Part 2 reflects the EU’s agreement to the ICH Q7 guidelines and forms the basis of the detailed guidelines.

 

The Compilation of European Union Procedures on Inspections and Exchange of Information

The Compilation of European Union Procedures on Inspections and Exchange of Information (CoUPs) is a collection of procedures for GMP and Good Distribution Practice (GDP) inspectorates, applicable to all the inspectorates in the European Union. It provides a tool to facilitate cooperation between EU Member States and a means to achieve harmonization. The CoUP covers, among other things, the basis for national procedures that form part of the national inspectorates’ quality systems, how quality defects and noncompliance are handled and how GMP and GDP inspections are carried out and reported.

The contents of the CoUP are constantly updated, developed and agreed, under the coordination of the EMA, by representatives of the Inspectorates of each Member State, including those supervising the manufacture and import of veterinary medicinal products only. Once agreed, they are adopted by the European Commission and then published on its behalf by the EMA.

Common Union formats for manufacturing and import authorizations, GMP certificates and for statements of non-compliance with GMP have been agreed and published in the compilation and implemented by EU competent authorities in order to enhance communication, collaboration and co-operation between authorities. This common format enables Member States to enter manufacturing, importing and distribution authorizations in the Union database, EudraGMDP.

The GMP/GDP Inspectors Working Group

The GMP/GDP Inspectors Working Group (GMDP IWG) is a group of senior inspectors appointed by all the EEA competent authorities which meets at EMA premises four times a year. It is chaired by EMA and a European Commission representative attends the meetings, as well as observers from the European EDQM, accession countries (countries which have applied to be part of the EU but have not joined yet) and MRA partners. Representatives from other international authorities can be invited on a case-by-case basis.

The group is a forum for harmonization and discussion of common issues which are taken by the inspectors back to their NCA for implementation. Any new or amended text of the EU GMP guide is developed by this group, with the European Commission responsible for the final adoption. The GMDP IWG also maintains the CoUP and oversees, on behalf of the Heads of Medicines Agencies (HMAs) the Joint Audit Programme.

Training

The GMDP IWG organises training for EEA inspectors and inspectors from accession countries, aimed at raising the technical capability of the inspectors, ensuring common understanding of issues related to GMP and harmonization. In addition, EMA has signed a partnership agreement with PIC/S on cooperation on training for GMP inspectors, which recognizes the role that PIC/S plays in this area and avoids duplication of effort.

Ensuring Equivalence before Joining the EU

Becoming a member of the European Union is a complex procedure and there are strict conditions for EU membership to ensure that new members are admitted only when they are fully able to take on the obligations of membership, including compliance with all the EU’s standards and rules. For the purpose of accession negotiations, these are divided into 35 different policy fields(chapters).

For acceding to the EU, a candidate country must implement the EU rules and regulations in all areas. The length of the membership negotiations can vary and depends on the time needed to complete the necessary reforms and the alignment with EU law. The candidates are supported financially, administratively and technically during this preaccession period.

In order to ensure that new Member States joining the European Union have reached the same level as the other members before the date of accession, a number of measures are put in place. These include:

  • The European Commission checks compliance with the EU legislation (including pharmaceutical legislation)
  • Through the TAIEX program, financed by the European Commission, technical support may be provided
  • Accession countries are invited as observers to EU meetings (including the GMDP IWG)
  • Specific training on EU procedures is organized

Auditing Member States

Auditing is an important part of the measures put in place in order to oversee the equivalence of Member States. There are a number of contexts in which Member States NCAs and/or inspectorates can be audited.

The Joint Audit Program (JAP) of the EU NCAs’ GMP inspectorates is an internal audit program under the Heads of Medicines Agencies (HMA) and is run on behalf of HMA by the GMDP IWG. JAP aims at achieving and maintaining equivalence between Member States’ national inspectorates responsible for GMP. It was established in October 2000 and is an important part of the quality system adopted by all GMP inspectorates in the EU.

JAP auditors are senior GMP inspectors, further qualified for auditing inspectorates through specific training. A list of qualified JAP auditors is maintained by the Compliance Group, which is a subgroup of the GMDP IWG. JAP auditors also provide technical advice and support to accession countries before they become EU Member States.

EU inspectorates are audited through the JAP onsite, at intervals established through a risk-based approach (typically every five to six years). Mutual Recognition Agreement and other international partners are invited on a case-by-case basis to join JAP audits of EU Member States inspectorates as observers.

Audits are also organized in the framework of the Pharmaceutical Inspection Convention and Pharmaceutical Inspection Co-operation Scheme (jointly referred to as PIC/S) and Mutual Recognition Agreement (MRA) (see International Cooperation Activities below). Since most of the EU authorities and all MRA partners are member of PIC/S, synergies between the various audit schemes are used in order to avoid duplication.

BEMA Audits

The Benchmarking of European Medicines Agencies (BEMA) is an internal EU program managed by the Heads of Medicines Agencies, based on assessment of the systems and processes in individual agencies against a set of indicators in four main areas:

  • Management systems
  • Assessment of marketing authorization applications
  • Pharmacovigilance (drug safety) activities
  • Inspection services

The assessment identifies strengths and best practices in agencies and any opportunity for improvement. The program has concluded its third cycle in 2015.

International Cooperation Activities

The European Union and its Member States are involved in several bilateral and multilateral cooperation activities with international partners in the GMP area. The main advantage is that international cooperation allows, by relying on information received from trusted international authorities, to reallocate foreign inspections towards areas more at risk. It thus optimizes available inspection resources.

PIC/S

The Pharmaceutical Inspection Convention and Pharmaceutical Inspection Co-operation Scheme (jointly referred to as PIC/S) aims at harmonizing inspection procedures worldwide by developing common standards in the field of GMP and by providing training opportunities to inspectors. It also aims at facilitating cooperation and networking between competent authorities, regional and international organisations, thus increasing mutual confidence. Most EU Member States are members of PIC/S while EMA is participating in PIC/S activities as a partner organization.

Mutual Recognition Agreements

Mutual Recognition Agreements (MRAs) are official agreements on the mutual recognition of assessment of conformity of regulated products which are negotiated and signed at EU level. MRAs concluded by the European Union include pharmaceuticals and cover GMP. Consequently, inspection results carried out by MRA partners in their territory are recognized by EU Member States and vice versa and retesting upon importation into the European Union is not needed in the QP batch certification process. The MRA scope can cover both human and veterinary products, finished products, active substances and Investigational Medicinal Products, but there are differences in scope between the various MRAs.

Currently, the European Union has operational MRAs in place with Australia, Canada, Japan, New Zealand and Switzerland. The EU also has in place an Agreement on Conformity Assessment and Acceptance of industrial products (ACAA), which includes GMP, with Israel. An ACAA is a specific type of MRA; the main practical difference is that in the ACAA case results of inspections carried out outside the territory of the agreement partners are mutually recognized as well, in addition to inspections carried out in the partners’ territory. An MRA between the European Union and the United States was signed in 1999; at the time of this writing it is operational only toward rapid alerts.

International Coalition of Medicines Regulatory Authorities

The European Commission, EMA and some EU Member States (France, Germany, Ireland, Italy, Spain and UK) participate to the activities of the International Coalition of Medicines Regulatory Authorities (ICMRA). ICMRA is a recent initiative started by Heads of Medicines Agencies worldwide, which aims at providing global strategic coordination and direction on areas that are common to many regulatory authorities’ missions worldwide, and which builds on existing arrangements such as those of PIC/S. The ICMRA has the objective to establish synergies and to foster global cooperation among regulators and GMP is one of the ICMRA main areas of interest.

Other International Cooperation Activities

In addition to MRAs, the European Union is involved in several less formalized cooperation schemes on GMP with international partners and/or in areas not covered by an MRA.

The API international cooperation project has as main objectives the sharing of information on inspection planning, policy and inspection reports and joint inspections on manufacturers located outside the participating countries. It includes the following participants: the EMA and all EU member States, the European EDQM, the U.S. FDA, the Australian Therapeutic Goods Administration (TGA) and WHO.

Several bilateral pilots and programs between EMA and FDA were also developed during the last ten years with the view to increase collaboration on domestic and third country GMP inspections.

This less formal form of cooperation in the last years has allowed the building of confidence among cooperating countries and regions, mainly through joint inspections and exchange of information, and is opening new possibilities of mutual reliance on inspection results. In this perspective, it is worth noting that the European Union has identified the recognition of GMP inspections carried out in the European Union and the United States and in third countries as a main objective for the pharmaceutical sector in the context of the negotiations of the Transatlantic Trade and Investment Partnership (TTIP).

 

Disclaimer: The views expressed in this article are those of the authors and may not be understood or quoted as being made on behalf of or reflecting the position of the Agencies or Institutions with which the authors are affiliated.

Notes

*The term “Medicinal Product” in the European Union approximately corresponds to the term “Drug Product” in the United States. Sometimes the term “Finished Product” is used instead.

**The term “Active Substance” in the European Union corresponds to drug “Drug Substance” in the United States.

Tags: EMA , Europe , inspections , GMP , EC , European Commission , European regulations , PIC/S , GMP regulation

Share

GMP/GDP: When will I be inspected by the Authorities?

 regulatory  Comments Off on GMP/GDP: When will I be inspected by the Authorities?
Apr 212016
 

 

 

Various competent authorities are performing inspections. But who is subject to such an inspection?

http://www.gmp-compliance.org/enews_05297_GMP-GDP-When-will-I-be-inspected-by-the-Authorities_15352,15356,15274,15432,Z-QAMPP_n.html

GMP Inspections are carried out at Manufacturer Licence Holders

A manufacturer of medicinal products must meet Good Manufacturing Practice (GMP) standards. These standards are defined in various laws and regulations. In the EU the compliance with these regulations is checked and assessed by the national competent authorities. The overall goal is to have medicinal products of consistent high quality that meet the requirements of the marketing authorisation (MA) or product specification.

If a company supplies product to the USA, the U.S. Food and Drug Administration (FDA) might inspect the site assuring that drugs, medical devices, certain active pharmaceutical ingredients (APIs) and biological products manufactured in foreign countries and intended for U.S. distribution are in compliance with the applicable U.S. law and regulations.

GDP Inspections are carried out at Wholesale Dealer Licence Holders

Good Distribution Practice (GDP) requires that medicines are obtained from the licensed supply chain and are consistently stored, transported and handled under suitable conditions, as required by the MA or product specification. Many of the actors in the supply chain must implement GDP but are not under supervision. The competent authority for GDP will normally not carry out GDP inspections at transport companies (shipping companies) or at airport hubs.

You will also be inspected when you apply for a manufacturer or wholesaler dealer licence and then periodically, normally based on risk assessments. Overseas manufacturing sites are also inspected when medicinal products or certain APIs are imported to the EU.

Types of inspection

Inspections under a risk-based compliance programme

It is the aim of the competent authorities and inspectorates to prioritise regular inspections based on risk assessments. These inspections are generally announced in advance.

GMP inspections may sometimes be carried out with other inspections, such as with GDP, Good Clinical Practice (GCP) or Good Pharmacovigilance Practice (GPvP).

Product-related GMP inspections

Inspectorates may conduct product-related GMP inspections when assessing an application for a marketing authorisation. This inspection checks if the manufacturer complies with GMP. FDA may also carry out these pre-approval inspections. These inspections are generally announced in advance.

Product-related inspections can also be requested by the European Medicines Agency (EMA) for example by the Committee for Human Medicinal products (CHMp) during the pre-application of a centralised marketing authorisation application or the Co-ordination group for Mutual Recognition and Decentralised Procedures – human (CMDh). EMA uses inspectors from EU member states to ensure compliance with GMP principles.

Triggered or For Cause Inspections

Competent Authorities may inspect you if they are informed about possible GMP or GDP breaches for example by a whistle blower, the press/ media or another regulatory authority.

Here, only little or no notification of these inspections is given in advance.

 

 

///GMP inspections, Manufacturer Licence Holders

Share

Plecanatide, 普卡那肽 , ليكاناتيد ,плеканатид

 NDA, Uncategorized  Comments Off on Plecanatide, 普卡那肽 , ليكاناتيد ,плеканатид
Apr 202016
 

 

STR1

PLECANATIDE;  UNII-7IK8Z952OK;  (3-Glutamic acid(D>E))human uroguanylin (UGN); 467426-54-6;

Molecular Formula: C65H104N18O26S4
Molecular Weight: 1681.88626 g/mol
Share

ORVEPITANT

 phase 2  Comments Off on ORVEPITANT
Apr 202016
 

Molecular Formula: C31H35F7N4O2
Molecular Weight: 628.624022 g/mol

CAS 579475-18-6

Orvepitant (GW823296)

(2R,4S)-4-[(8aS)-6-oxo-1,3,4,7,8,8a-hexahydropyrrolo[1,2-a]pyrazin-2-yl]-N-[(1R)-1-[3,5-bis(trifluoromethyl)phenyl]ethyl]-2-(4-fluoro-2-methylphenyl)-N-methylpiperidine-1-carboxamide

Orvepitant maleate

 

MALEATE

CAS [579475-24-4] MALEATE

MF C31H35F7N4O2.C4H4O4
MW 744.70

https://clinicaltrials.gov/ct2/show/NCT01000493

  • Phase IICough; Pruritus
  • DiscontinuedAnxiety disorders; Major depressive disorder; Post-traumatic stress disorders

Most Recent Events

  • 19 Dec 2015NeRRe Therapeutics terminates a phase II trial in Pruritus in Italy and the United Kingdom (EudraCT2013-002763-25)
  • 16 Dec 2013No development reported – Phase-II for Post-traumatic stress disorder in USA (PO)
  • 16 Dec 2013No development reported – Phase-II for Major depressive disorder in Canada (PO)
Company NeRRe Therapeutics Ltd.
Description Neurokinin 1 (NK1) receptor antagonist
Molecular Target Neurokinin 1 (NK1) substance P receptor (TACR1)
Mechanism of Action Neurokinin-1 (NK-1) (Substance P) receptor antagonist
Therapeutic Modality Small molecule
Latest Stage of Development Phase II
Standard Indication Itch
Indication Details Treat intense pruritus (itch) associated with epidermal growth factor receptor inhibitor (EGFRi) anticancer therapies

Start of Phase II study of neurokinin-1 receptor antagonist orvepitant for intense pruritus induced by epidermal growth factor receptor inhibitors

First Clinical Trial for NeRRe Therapeutics

Stevenage, UK, 23 January 2014.

NeRRe Therapeutics Ltd, which is focused on the development of neurokinin (NK) receptor antagonists for a range of indications, is pleased to announce the start of a Phase II study of the novel NK-1 receptor antagonist orvepitant. The proof-of-concept study, results of which are expected in 2015, is investigating orvepitant’s effectiveness as a treatment for the intense pruritus (itch) associated with epidermal growth factor receptor inhibitor (EGFRi) anticancer therapies. The itch intensity experienced by patients can be so severe that their EGFRi dose must be reduced or the treatment withdrawn; also pruritus along with rash has a significant effect on quality of life1.

The RELIEVE-1 trial is a randomised, double-blind, placebo-controlled study to evaluate the safety, tolerability and efficacy of two daily dose levels of oral orvepitant on EGFRi-induced intense pruritus in oncology subjects. Its primary endpoint is the difference between orvepitant and placebo in reducing the intensity of pruritus over 4 weeks, as measured on a subject-recorded numerical rating scale. RELIEVE-1 is being undertaken in 15 clinical sites in Italy, with Dr Bruno Vincenzi from Università Campus Bio-Medico di Roma as lead investigator. Dr Vincenzi and his colleagues at the centre have pioneered the use NK-1 antagonists as anti-pruritics in this setting2. Chemistry, manufacturing and control support for RELIEVE-1 is being provided by Aptuit (Verona) Srl, with clinical operations assistance from the CRO Cromsource.

Dermatologic adverse events such as pruritus are a common feature of targeted anti-cancer therapies, with incidence of this symptom induced by EGFRia drugs in clinical trials ranging from 14.6% to 54.9% depending on the specific agent3. Open-label studies in patients suffering from refractory chronic pruritus have indicated that NK-1 receptor antagonism can provide rapid and highly effective relief as well as significantly improving quality of life.2,4,5,6

 

Dr Mike Trower, Co-founder & Chief Operating Officer of NeRRe Therapeutics said: 

‘We are very pleased to announce the start of RELIEVE-1, NeRRe’s first clinical trial, in this important area of unmet medical need. There is a strong rationale and a growing body of clinical evidence supporting the potential of orvepitant as an anti-pruritic for this devastating symptom commonly associated with EGFRis. Given its known effects on mood and sleep, orvepitant may also provide additional benefits for patient well-being.’

 

Dr Emiliangelo Ratti, NeRRe Therapeutics Co-founder added:

The intense pruritus induced by EGFRis can lead to significant suffering and poor quality of life, and we believe that a treatment for this troubling side effect would be welcomed by cancer patients and supportive care doctors alike. A successful study of orvepitant in this indication would provide further evidence of the broad therapeutic potential of the NK-1 receptor antagonist mechanism which NeRRe is exploiting in its pipeline.’

–ENDS–

a This includes monoclonal antibodies that target the extracellular domain of EFGR, small molecule tyrosine kinase (TK) inhibitors, and small molecule dual TK inhibitors.

 

About NeRRe Therapeutics

NeRRe Therapeutics was formed in December 2012 and is focussed on the development of a portfolio of NK receptor antagonists acquired from GlaxoSmithKline (GSK), which have therapeutic potential in a broad range of indications. NeRRe Therapeutics was co-founded by Drs Emiliangelo Ratti and Mike Trower, both of whom are both former senior leaders of neurosciences drug discovery at GSK with intimate knowledge of the transferred assets and the neurokinin receptor system field. In 2012 NeRRe Therapeutics raised £11.5 million ($18.4 million) in Series A financing from two leading European financial institutions, Novo A/S (www.novo.dk/ventures) and Advent Life Sciences (www.adventventures.com), who are represented by Dr Martin Edwards (Chairman) and Dr Kaasim Mahmood respectively on the company’s Board.

NeRRe (www.nerretherapeutics.com) is based at the state-of-the-art Stevenage Bioscience Catalyst (www.stevenagecatalyst.com), the UK’s first open innovation bioscience campus.

 

About Orvepitant

Orvepitant is a ‘novel generation’ brain penetrant, selective and potent, small molecule NK-1 receptor antagonist7 that features high receptor occupancy and full and long lasting (≥24hrs) central NK-1 receptor occupancy8. It has previously completed extensive safety and toxicology studies to support its clinical development; and it has already demonstrated a positive antidepressant effect in a Phase II clinical study together with beneficial effects on sleep8.

PATENT

http://www.google.com/patents/EP2297152A1?cl=en

NK1 antagonist compound orvepitant maleate, pharmaceutical formulations comprising this crystalline form, its use in therapy and processes for preparing the same. Background of the invention

WO03/066635 describes a number of diazabicycle derivatives having NK1 activity, including the 2-(R)-(4-Fluoro-2-methyl-phenyl)-4-(S)-((8aS)-6-oxo-hexahydro- pyrrolo[1 ,2-a]-pyrazin-2-yl)-piperidine-1-carboxylic acid [1-(R)-(3,5-bis-trifluoromethyl- phenyl)-ethyl]-methylamide (otherwise known as orvepitant).

The structure of the 2-(R)-(4-Fluoro-2-methyl-phenyl)-4-(S)-((8aS)-6-oxo-hexahydro- pyrrolo[1 ,2-a]-pyrazin-2-yl)-piperidine-1-carboxylic acid [1-(R)-(3,5-bis-trifluoromethyl- phenyl)-ethyl]-methylamide (otherwise known as orvepitant) is shown in formula (I) below:

Figure imgf000002_0001

Hereinafter any reference to orvepitant refers to the compound of formula (I).

Orvepitant may also be known as: CAS Index name

1-Piperidinecarboxamide, Λ/-[(1R)-1-[3,5-bis(trifluoromethyl)phenyl]ethyl]-2-(4-fluoro-

2-methylphenyl)-4-[(8aS)-hexahydro-6-oxopyrrolo[1 ,2-a]pyrazin-2(1 /-/)-yl]-Λ/-methyl-,

(2RAS) and IUPAC name :

(2R,4S)-Λ/-{(1 R)-1-[3,5-bis(trifluoromethyl)phenyl]ethyl}-2-(4-fluoro-2-methylphenyl)-

Λ/-methyl-4-[(8aS)-6-oxohexahydropyrrolo[1 ,2-a]pyrazin-2(1 /-/)-yl]-1- piperidinecarboxamide. A preferred salt of this compound is its hydrochloride salt which is otherwise known as orvepitant hydrochloride.

A further preferred salt of this compound is its maleate salt which is otherwise known as orvepitant maleate.

Particularly Example 1 1 C of WO03/066635 describes the synthesis of orvepitant maleate using substantially the same experimental conditions described in the Example 1 in the present patent application.

We have now found that orvepitant maleate can be obtained in a new crystalline form. In particular, we have discovered a form of orvepitant maleate which is anhydrous and crystalline and which surprisingly has particularly good pharmaceutical properties. This is particularly stable and essentially non hygroscopic. It also has good storage properties and can be readily formulated into pharmaceutical compositions such as tablets and capsules.

Example 1 : preparation of orvepitant maleate (Form 2) {(1 R)-1 -[3,5-bis(trifluoromethyl)phenyl]ethyl}methylamine – (2R)-2-hydroxybutanedioic acid (1.8 kg) was added to ethyl acetate (5.4 litres) and 15% w/w sodium carbonate solution (5.4 litres) and was stirred until all solids had dissolved. The organic phase was separated and was washed with water (5.4 litres). Fresh ethyl acetate (6.7 litres) was added and the solution was distilled to 5.4 litres under reduced pressure.

The solution was diluted with ethyl acetate (3.6 litres). The reactor was purged with carbon dioxide and a continuous steady stream of carbon dioxide was maintained. Triethylamine (810 ml) was added over 30 minutes and was rinsed in with ethyl acetate (250 ml). The reaction mixture was stirred for 30 minutes. Chlorotrimethylsilane (850 ml) was added over 30 minutes with cooling to keep the temperature between 17°C and 23°C and was rinsed in with ethyl acetate (250 ml). The reaction mixture was stirred for 30 minutes. Pyridine (720 ml) was added and was rinsed in with ethyl acetate (250 ml). Thionyl chloride (480 ml) was added over 10 minutes and then a rinse of ethyl acetate (500 ml). The reaction mixture was stirred at 200C for 16 hours under a carbon dioxide atmosphere.

28% w/w Racemic malic acid solution (5.3 litres) was added and the mixture was stirred for 15 minutes. The organic phase was separated, diluted with ethyl acetate (1.5 litres) and was washed with water (2 x 2.7 litres) and 20% w/w dibasic potassium phosphate solution (5.6 litres). The solution was distilled under reduced pressure to a total volume of 2.5 litres. Ethyl acetate (5 litres) was added and the solution was redistilled to 3 litres to give a solution of {(1 R)-1-[3,5- bis(trifluoromethyl)phenyl]ethyl}methylcarbamic chloride.

(2R)-2-(4-fluoro-2-methylphenyl)-4-piperidinone – (2S)-hydroxy(phenyl)ethanoic acid (1.2 kg) was added to 15% w/w sodium carbonate solution (4.8 litres) and ethyl acetate (4.8 litres) and the mixture was stirred until solids dissolved. The organic phase was separated and was washed with 20% w/w sodium chloride solution (4 litres). Fresh ethyl acetate (4.8 litres) was added and the solution of (2R)-2-(4-fluoro- 2-methylphenyl)-4-piperidinone was distilled under reduced pressure to a volume of 3 litres. The solution of (2R)-2-(4-fluoro-2-methylphenyl)-4-piperidinone was charged to the solution of {(1 R)-1-[3,5-bis(trifluoromethyl)phenyl]ethyl}methylcarbamic chloride followed by an ethyl acetate (300 ml) rinse. Triethylamine (857 g) was added followed by ethyl acetate (300 ml) and the mixture was boiled at reflux for 18 hours. The slurry was cooled to 200C and N-acetylpiperazine (240 g) was added. The reaction mixture was stirred for 30 minutes at 200C and was then charged with 28% w/w racemic malic acid solution (3.6 litres). The organic phase was separated and was washed with 20% w/w sodium chloride solution (4.8 litres). Ethyl acetate (4.8 litres) was added and the solution of (2R)-N-{(1 R)-1-[3,5- bis(trifluoromethyl)phenyl]ethyl}-2-(4-fluoro-2-methylphenyl)-N-methyl-4-oxo-1- piperidinecarboxamide was distilled under reduced pressure distillation to a total volume of 3 litres.

(8aS)-hexahydropyrrolo[1 ,2-a]pyrazin-6(2H)-one – (2S)-(acetyloxy)(phenyl)ethanoic acid (1.5 kg) was added to acetonitrile (11.4 litres) and triethylamine (450 g) was added. An acetonitrile (250 ml) rinse was added and the slurry was stirred at 200C for 30 min. Sodium triacetoxyborohydride (900 g) was added and the reaction was cooled to 100C. Formic acid (396 ml) was added to the mixture over 30 min, maintaining the temperature below 15°C. An acetonitrile (250 ml) rinse was added and the reaction was warmed to 200C. The solution of (2R)-N-{(1 R)-1-[3,5- bis(trifluoromethyl)phenyl]ethyl}-2-(4-fluoro-2-methylphenyl)-N-methyl-4-oxo-1- piperidinecarboxamide in ethyl acetate was added to the reaction mixture and was rinsed in with acetonitrile (1 litre). The reaction was stirred for 16 hours at 200C.

The slurry was distilled to 5 litres under reduced pressure. The mixture was diluted with ethyl acetate (10 litres) and was washed with 13% w/w ammonia solution (2 x 4 litres), and 10% w/w sodium chloride solution (4 litres). The organic solution was distilled to 5 litres under reduced pressure. The solution was diluted with IPA (8 litres) and was distilled under reduced pressure to 5 litres. Further IPA (8 litres) was added and the solution was again distilled to 5 litres.

A solution of maleic acid (248.5 g) in IPA (2.5 litres) was added. The mixture was then seeded with orvepitant maleate A (1 g) and the mixture was aged for 1 hour. Iso-octane (10 litres) was added over 30 min. and the mixture further aged for 1 hour. The slurry was cooled to 7°C and was further aged for 90 minutes. The solid formed was filtered and washed with a 1 :1 mixture of IPA/iso-octane (2 x 3 litres). The resulting solid was dried at 40°C under reduced pressure to give the title compound (1.095kg, 44%). NMR (CD3OD) δ (ppm) 1.52-1.53 (d, 3H), 1.68-1.78 (m, 1 H), 1.82-1.91 (q, 1 H), 1.95- 2.05 (m, 1 H), 2.16-2.37 (m, 3H), 2.38-2.50 (m, 2H), 2.44 (s, 3H), 2.81-2.87 (t, 1 H),

2.83 (s, 3H), 2.90-2.99 (m, 2H), 3.1 1-3.18 (dt, 1 H), 3.48-3.60 (m, 3H), 3.66-3.69 (d, 1 H), 3.89-3.96 (m, 1 H), 4.15-4.19 (dd, 1 H), 4.33-4.36 (dd , 1 H), 5.40-5.45 (q, 1 H), 6.26 (s, 2H), 6.76-6.81 (dt, 1 H), 6.85-6.88 (dd, 1 H), 7.27-7.31 (dd, 1 H), 7.70 (s, 2H), 7.88 (s, 1 H). (M+H)+ Calcd for C3iH35F7N4O 629, found 629.

References:

  1. Rosen AC et al. Am J Clin Dermatol. (2013), 14(4):327-33
  2. Santini D et al. Lancet Oncol. (2012), 13(10):1020-4
  3. Ensslin CJ et al. J Am Acad Dermatol. (2013), 69(5):708-20
  4. Duval A, Dubertret L. N Engl J Med. (2009), 1;361(14):1415-6
  5. Ständer S et al. PLoS One. (2010), 5(6):e10968
  6. Torres T et al. J Am Acad Dermatol. (2012), 66(1):e14-5
  7. Di Fabio R et al. Bioorg Med Chem. (2013), 21(21):6264-73
  8. Ratti E et al. J Psychopharmacol. (2013), 27(5):424-34
Patent ID Date Patent Title
US2015238486 2015-08-27 NOVEL USES
US2014128395 2014-05-08 Novel Method
US2011166150 2011-07-07 Anhydrous Crystal Form Of Ovrepitant Maleate
US2010317666 2010-12-16 Composition Comprising An NK-1 Receptor Antagonist And An SSRI For The Treatment Of Tinnitus And Hearing Loss
US2010152446 2010-06-17 Piperidine Derivatives
US2010105688 2010-04-29 PHARMACEUTICAL COMPOSITIONS COMPRISING 3,5-DIAMINO-6-(2,3-DICHLOPHENYL)-1,2,4-TRIAZINE OR R(-)-2,4-DIAMINO-5-(2,3-DICHLOROPHENYL)-6-FLUOROMETHYL PYRIMIDINE AND AN NK1
US7652012 2010-01-26 2-(R)-(4-fluoro-2-methyl-phenyl)-4-(S)-((8aS)-6-oxo-hexahydro-pyrrolo[1,2-a]-pyrazin-2-yl)-piperidine-1-carboxylic acid [1-(R)-3,5-bis-trifluoromethyl-phenyl)-ethyl]-methylamide maleate and pharmaceutical compositions thereof
US2009326032 2009-12-31 PHARMACEUTICAL COMPOSITIONS COMPRISING NK1 RECEPTOR ANTAGONISTS AND SODIUM CHANNEL BLOCKERS
US2009318530 2009-12-24 PHARMACEUTICAL COMPOSITIONS COMPRISING NK1 RECEPTOR ANTAGONISTS AND SODIUM CHANNEL BLOCKERS
US7189713 2007-03-13 Piperidine derivatives
Patent ID Date Patent Title
US7189713 2007-03-13 Piperidine derivatives
US2006287325 2006-12-21 Combinations of paroxetine and 2-(r)-(4-fluoro-2-methyl-phenyl)-4-(s)-((8as)-6-oxo-hexahydro-pyrrolo’1,2-a!-pyrazin-2-yl)-piperidine-1-carboxylicacid’1-(r)-(3,5-bis-trifluoromethyl-phenyl)-
US6384099 2002-05-07 Method for curing polymeric materials, such as those used in dentistry, and for tailoring the post-cure properties of polymeric materials through the use of light source power modulation
US6282013 2001-08-28 System for curing polymeric materials, such as those used in dentistry, and for tailoring the post-cure properties of polymeric materials through the use of light source power modulation
US6008264 1999-12-28 Method for curing polymeric materials, such as those used in dentistry, and for tailoring the post-cure properties of polymeric materials through the use of light source power modulation

REFERENCES

1: Di Fabio R, Alvaro G, Braggio S, Carletti R, Gerrard PA, Griffante C, Marchioro C, Pozzan A, Melotto S, Poffe A, Piccoli L, Ratti E, Tranquillini E, Trower M, Spada S, Corsi M. Identification, biological characterization and pharmacophoric analysis of a new potent and selective NK1 receptor antagonist clinical candidate. Bioorg Med Chem. 2013 Nov 1;21(21):6264-73. doi: 10.1016/j.bmc.2013.09.001. Epub 2013 Sep 11. PubMed PMID: 24075145.

2: Ratti E, Bettica P, Alexander R, Archer G, Carpenter D, Evoniuk G, Gomeni R, Lawson E, Lopez M, Millns H, Rabiner EA, Trist D, Trower M, Zamuner S, Krishnan R, Fava M. Full central neurokinin-1 receptor blockade is required for efficacy in depression: evidence from orvepitant clinical studies. J Psychopharmacol. 2013 May;27(5):424-34. doi: 10.1177/0269881113480990. Epub 2013 Mar 28. PubMed PMID: 23539641.

///////Orvepitant, GW823296, PHASE 2, Neurokinin 1 (NK1) receptor antagonist

C[C@@H](N(C)C(=O)N1CC[C@@H](C[C@@H]1c1ccc(F)cc1C)N1CCN2[C@@H](CCC2=O)C1)c1cc(cc(c1)C(F)(F)F)C(F)(F)F

CC1=C(C=CC(=C1)F)C2CC(CCN2C(=O)N(C)C(C)C3=CC(=CC(=C3)C(F)(F)F)C(F)(F)F)N4CCN5C(C4)CCC5=O

Share

Enasidenib (AG-221)

 Uncategorized  Comments Off on Enasidenib (AG-221)
Apr 202016
 

img

Enasidenib.png

Enasidenib (AG-221)

1446502-11-9
Chemical Formula: C19H17F6N7O
Exact Mass: 473.13988

AG-221; AG 221; AG221; CC-90007; CC 90007; CC90007; Enasidenib

IUPAC/Chemical Name: 2-methyl-1-((4-(6-(trifluoromethyl)pyridin-2-yl)-6-((2-(trifluoromethyl)pyridin-4-yl)amino)-1,3,5-triazin-2-yl)amino)propan-2-ol

2-methyl-1-(4-(6-(trifluoromethyl)pyridin-2-yl)-6-(2-(trifluoromethyl)pyridin-4-ylamino)-1,3,5-triazin-2-ylamino)propan-2-ol

Agios Pharmaceuticals, Inc. innovator

Enasidenib, aslo known as AG-221 and CC-90007, is a potent and selective IDH2 inhibitor with potential anticancer activity (IDH2 = Isocitrate dehydrogenase 2). The mutations of IDH2 present in certain cancer cells result in a new ability of the enzyme to catalyze the NAPH-dependent reduction of α-ketoglutarate to R(-)-2-hydroxyglutarate (2HG). The production of 2HG is believed to contribute to the formation and progression of cancer . The inhibition of mutant IDH2 and its neoactivity is therefore a potential therapeutic treatment for cancer

AG-221 is an orally available, selective, potent inhibitor of the mutated IDH2 protein, making it a highly targeted investigational medicine for the potential treatment of patients with cancers that harbor an IDH2 mutation. AG-221 has received orphan drug and fast track designations from the U.S. FDA. In September 2013, Agios initiated a Phase 1 multicenter, open-label, dose escalation clinical trial of AG-221 designed to assess the safety and tolerability of AG-221 in advanced hematologic malignancies. In October 2014, Agios initiated four expansion cohorts as part of the ongoing Phase 1 study and expanded its development program with the initiation of a Phase 1/2 study of AG-221 in advanced solid tumors. For the detailed information of AG-221, the solubility of AG-221 in water, the solubility of AG-221 in DMSO, the solubility of AG-221 in PBS buffer, the animal experiment (test) of AG-221, the cell expriment (test) of AG-221, the in vivo, in vitro and clinical trial test of AG-221, the EC50, IC50,and affinity,of AG-221, For the detailed information of AG-221, the solubility of AG-221 in water, the solubility of AG-221 in DMSO, the solubility of AG-221 in PBS buffer, the animal experiment (test) of AG-221, the cell expriment (test) of AG-221, the in vivo, in vitro and clinical trial test of AG-221, the EC50, IC50,and affinity,of AG-221,

Agios Announces New Data from Ongoing Phase 1 Dose Escalation and Expansion Trial of AG-221 Showing Durable Clinical Activity in Patients with Advanced Hematologic Malignancies

IDH2-Mutant Inhibitor Shows Durable Responses of More than 15 Months in Patients with Advanced Acute Myeloid Leukemia (AML) and Other Blood Cancers

Proof-of-Concept Demonstrated in Myelodysplastic Syndrome (MDS) and Untreated AML

125-Patient Expansion Cohort and Global Registration-Enabling Program Remain on Track

Company to Host Conference Call and Webcast Today

CAMBRIDGE, Mass. & VIENNA–(BUSINESS WIRE)–Jun. 12, 2015– Agios Pharmaceuticals, Inc. (Nasdaq:AGIO), a leader in the fields of cancer metabolism and rare genetic disorders of metabolism, today announced new data from the dose-escalation phase and expansion cohorts from the ongoing Phase 1 study evaluating single agent AG-221, a first-in-class, oral, selective, potent inhibitor of mutant isocitrate dehydrogenase-2 (IDH2), in advanced hematologic malignancies. The data will be presented at the 20th Congress of the European Hematology Association (EHA) taking place June 11-14, 2015 in Vienna.

Data as of May 1, 2015 from 177 patients (104 in dose escalation and 73 from the first four expansion cohorts) with advanced hematologic malignancies treated with single agent AG-221 showed durable clinical activity and a favorable safety profile. More than half of the 177 patients remain on treatment. The study had an overall response rate of 40 percent (63 of 158 response-evaluable patients, using the criteria below) and a complete remission rate of 16 percent (26 of 158 response-evaluable patients). Patients responding to AG-221 continue to show durable clinical activity on treatment for more than 15 months, with an estimated 76 percent of responders staying on treatment for six months or longer. The overall safety profile observed was consistent with previously reported data with more than 100 additional patients treated as of the last analysis.

This new data reflects responses in the evaluable population, which includes all patients with a pre-AG-221 screening assessment and day 28 or later response assessment or an earlier discontinuation for any reason. Patients with a screening assessment who were still on treatment, but had not reached the day 28 disease assessment, were excluded.

“The clinical profile of AG-221 continues to be impressive from the perspectives of response rate, durability, safety and unique mechanism of action,” said Courtney DiNardo, M.D., lead investigator and assistant professor, leukemia atUniversity of Texas MD Anderson Cancer Center. “Additionally, it is encouraging to see early proof-of-concept in myelodysplastic syndrome (MDS) and untreated acute myeloid leukemia (AML) given the need for more effective therapies for these patients.”

“As the data from the AG-221 study continue to mature, we are compiling a robust dataset to quickly move this program into global registration studies later this year in collaboration with Celgene,” said Chris Bowden, M.D., chief medical officer of Agios. “We are excited about the speed of enrollment we’ve seen to date in our four expansion cohorts and are on track to enroll our recently announced fifth expansion cohort of 125 patients with relapsed and/or refractory AML. With this progress, we are executing on our strategy to combine speed and breadth to reach people with hematologic malignancies in urgent need of better treatments.”

About the Ongoing Phase 1 Trial for AG-221 in Advanced Hematologic Malignancies

AG-221 is currently being evaluated in an ongoing Phase 1 trial that includes a dose-escalation phase and four expansion cohorts of 25 patients each, evaluating patients with relapsed or refractory AML who are 60 years of age and older and transplant ineligible; relapsed or refractory AML patients under age 60; untreated AML patients who decline standard of care chemotherapy; and patients with other IDH2-mutant positive hematologic malignancies. Data reported here are from patients receiving AG-221 administered from 60 mg to 450 mg total daily doses in the dose escalation arm and 100 mg once daily in the first four expansion arms, as of May 1, 2015. The median age of these patients is 69 (ranging from 22-90). Treatment with AG-221 showed substantial reduction in the plasma levels of the oncometabolite 2-hydroxglutarate (2HG) to the level observed in healthy volunteers.

Safety Data

A safety analysis was conducted for all 177 treated patients as of May 1, 2015.

  • The majority of adverse events reported by investigators were mild to moderate, with the most common being nausea, fatigue, increased blood bilirubin and diarrhea.
  • The majority of serious adverse events (SAE) were disease related; SAEs possibly related to study drug were reported in 27 patients.
  • A maximum tolerated dose (MTD) has not been reached.
  • The all-cause 30-day mortality rate was 4.5 percent.

Efficacy Data

Sixty-three out of 158 response-evaluable patients achieved investigator-assessed objective responses for an overall response rate of 40 percent as of May 1, 2015.

  • Of the 63 patients who achieved an objective response, there were 26 (16 percent) complete remissions (CR), three CRs with incomplete platelet recovery (CRp), 14 marrow CRs (mCR), two CRs with incomplete hematologic recovery (CRi) and 18 partial remissions (PR).
  • Of the 111 patients with relapsed or refractory AML, 46 (41 percent) achieved an objective response, including 20 (18 percent) CRs, one CRp, 16 PRs, eight mCRs and one CRi.
  • Of the 22 patients with AML that had not been treated, seven achieved an objective response, including three CRs, two PRs, one mCR and one CRi.
  • Of the 14 patients with myelodysplastic syndrome (MDS), seven achieved an objective response, including two CRs, one CRp and four mCRs.
  • Responses were durable, with duration on study drug more than 15 months and ongoing. As of the analysis date, an estimated 88 percent of responses lasted three months or longer, and 76 percent of responses lasted six months or longer.

Upcoming Milestones for AG-221

Agios studies in IDH2-mutated solid and hematologic tumors are ongoing or planned for 2015 to further support development of AG-221.

  • Continue to enroll patients in the fifth expansion cohort of 125 patients with IDH2 mutant-positive AML who are in second or later relapse, refractory to second-line induction or re-induction treatment, or have relapsed after allogeneic transplantation.
  • Initiate combination trials to evaluate AG-221 as a potential frontline treatment for patients with AML and a broad range of hematologic malignancies in the second half of 2015.
  • Initiate a global Phase 3 registration-enabling study in relapsed/refractory AML patients that harbor an IDH2 mutation in the second half of 2015.
  • Continue dose escalation in the Phase 1/2 trial in patients with advanced solid tumors, including glioma and angioimmunoblastic T-cell lymphoma (AITL) that carry an IDH2 mutation in 2015.

Conference Call Information

Agios will host a conference call and webcast from the congress to review the data on Friday, June 12, 2015, beginning at 8:00 a.m. ET (2:00 p.m. CEST). To participate in the conference call, please dial (877) 377-7098 (domestic) or (631) 291-4547 (international) and refer to conference ID 53010830. The webcast will be accessible live or in archived form under “Events & Presentations” in the Investors and Media section of the company’s website at www.agios.com.

About Agios/Celgene Collaboration

AG-221, the IDH1-mutant inhibitor AG-120 and the pan-IDH mutant inhibitor AG-881 are part of Agios’ global strategic collaboration with Celgene Corporation. Under the terms of the collaboration, Celgene has worldwide development and commercialization rights for AG-221. Agios continues to conduct clinical development activities within the AG-221 development program and is eligible to receive up to $120 million in payments on achievement of certain milestones and royalties on net sales. For AG-120, Agios retains U.S. development and commercialization rights. Celgene has an exclusive license outside the United States. Celgene is eligible to receive royalties on net sales in the U.S. Agios is eligible to receive royalties on net sales outside the U.S. and up to $120 million in payments on achievement of certain milestones. For AG-881, the companies have a joint worldwide development and 50/50 profit share collaboration, and Agios is eligible to receive regulatory milestone payments of up to $70 million.

About IDH Mutations and Cancer

IDH1 and IDH2 are two metabolic enzymes that are mutated in a wide range of hematologic and solid tumor malignancies, including AML. Normally, IDH enzymes help to break down nutrients and generate energy for cells. When mutated, IDH increases production of an oncometabolite 2-hydroxyglutarate (2HG) that alters the cells’ epigenetic programming, thereby promoting cancer. 2HG has been found to be elevated in several tumor types. Agios believes that inhibition of the mutated IDH proteins may lead to clinical benefit for the subset of cancer patients whose tumors carry them.

About Acute Myelogenous Leukemia (AML)

AML, a cancer of blood and bone marrow characterized by rapid disease progression, is the most common acute leukemia affecting adults. Undifferentiated blast cells proliferate in the bone marrow rather than mature into normal blood cells. AML incidence significantly increases with age, and according to the American Cancer Society, the median age of onset is 66. Less than 10 percent of U.S. AML patients are eligible for bone marrow transplant, and the vast majority of patients do not respond to chemotherapy and progress to relapsed/refractory AML. The five-year survival rate for AML is approximately 20 to 25 percent. IDH2 mutations are present in about 9 to 13 percent of AML cases.

About Myelodysplastic Syndrome (MDS)

MDS comprises a diverse group of bone marrow disorders in which immature blood cells in the bone marrow do not mature or become healthy blood cells. The National Cancer Institute estimates that more than 10,000 people are diagnosed with MDS in the United States each year. Failure of the bone marrow to produce mature healthy cells is a gradual process, and reduced blood cell and/or reduced platelet counts may be accompanied by the loss of the body’s ability to fight infections and control bleeding. For roughly 30 percent of the patients diagnosed with MDS, this bone marrow failure will progress to AML. Chemotherapy and supportive blood products are used to treat MDS.

About Agios Pharmaceuticals, Inc.

Agios Pharmaceuticals is focused on discovering and developing novel investigational medicines to treat cancer and rare genetic disorders of metabolism through scientific leadership in the field of cellular metabolism. In addition to an active research and discovery pipeline across both therapeutic areas, Agios has multiple first-in-class investigational medicines in clinical and/or preclinical development. All Agios programs focus on genetically identified patient populations, leveraging our knowledge of metabolism, biology and genomics. For more information, please visit the company’s website at agios.com.

clips

AG-221, Inhibitor Of IDH2 Mutants

 

09338-scitech1-Agioscxd
COMBATTING CANCER
Agios’s AG-221 team. Front row (from left): Erin Artin, Kate Yen, Fang Wang, Hua Yang, and Lee Silverman. Back row (from left): Michael Su, Stefan Gross, Sam Agresta, Jeremy Travins, Yue Chen, and Lenny Dang.
Credit: Kevin Graham/Agios

The enzyme isocitrate dehydrogenase (IDH) is probably most famous for its role in the central cellular metabolic pathway, the Krebs cycle. The enzyme catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate. One subtype of the enzyme, IDH1, is found in cells’ cytoplasm, and another, IDH2, is found in their mitochondria.

 

Print
AG-221
Company: Agios Pharmaceuticals
Target: IDH2

People with certain mutations in IDH end up making R-2-hydroxyglutarate (2-HG) instead of α-ketoglutarate. 2-HG is known to make cancer cells flourish. In fact, IDH mutations have been implicated in about 70% of brain cancers and have also been identified in solid tumors and blood cancers, such as acute myeloid leukemia.

Jeremy M. Travins of Agios Pharmaceuticals spoke about how scientists at the company found compounds based on substituted triazines that can cut down on 2-HG production by inhibiting a dimer of mutant IDH2. Using structure-activity relationships and a crystal structure of a lead compound bound to the mutant IDH2 dimer, they managed to develop a clinical candidate: AG-221. It turns out that AG-221 doesn’t bind to the active site of mutant IDH2. Rather, the compound binds to the spot where the two enzymes meet in the dimer.

Hitting this position in just the right way is tricky, Travins explained. Hydrogen-bonding interactions from the triazine and the two amino groups that flank it are critical.

The compound is in Phase I clinical trials, Travins said, and it’s been shown to lower 2-HG levels to those seen in people without cancer. What’s more, he noted, the drug candidate has few side effects, giving patients a higher quality of life than standard chemotherapeutic agents do.

Patent

http://www.google.com/patents/US20130190287

Compound 409—2-methyl-1-(4-(6-(trifluoromethyl)pyridin-2-yl)-6-(2-(trifluoromethyl)pyridin-4-ylamino)-1,3,5-triazin-2-ylamino)propan-2-ol

Figure US20130190287A1-20130725-C00709

1H NMR (METHANOL-d4) δ 8.62-8.68 (m, 2H), 847-8.50 (m, 1H), 8.18-8.21 (m, 1H), 7.96-7.98 (m, 1H), 7.82-7.84 (m, 1H), 3.56-3.63 (d, J=28 Hz, 2H), 1.30 (s, 6H). LC-MS: m/z 474.3 (M+H)+.

 

Patent ID Date Patent Title
US2013190287 2013-07-25 THERAPEUTICALLY ACTIVE COMPOUNDS AND THEIR METHODS OF USE

REFERENCES

1: Caino MC, Altieri DC. Molecular Pathways: Mitochondrial Reprogramming in Tumor Progression and Therapy. Clin Cancer Res. 2016 Feb 1;22(3):540-5. doi: 10.1158/1078-0432.CCR-15-0460. Epub 2015 Dec 9. PubMed PMID: 26660517; PubMed Central PMCID: PMC4738153.

2: Stein EM. IDH2 inhibition in AML: Finally progress? Best Pract Res Clin Haematol. 2015 Jun-Sep;28(2-3):112-5. doi: 10.1016/j.beha.2015.10.016. Epub 2015 Oct 19. Review. PubMed PMID: 26590767.

3: Rowe JM. Reasons for optimism in the therapy of acute leukemia. Best Pract Res Clin Haematol. 2015 Jun-Sep;28(2-3):69-72. doi: 10.1016/j.beha.2015.10.002. Epub 2015 Oct 22. Review. PubMed PMID: 26590761.

4: Stein EM. Molecular Pathways: IDH2 Mutations-Co-opting Cellular Metabolism for Malignant Transformation. Clin Cancer Res. 2016 Jan 1;22(1):16-9. doi: 10.1158/1078-0432.CCR-15-0362. Epub 2015 Nov 9. PubMed PMID: 26553750.

5: Kiyoi H. Overview: A New Era of Cancer Genome in Myeloid Malignancies. Oncology. 2015;89 Suppl 1:1-3. doi: 10.1159/000431054. Epub 2015 Nov 10. Review. PubMed PMID: 26551625.

6: Tomita A. [Progress in molecularly targeted therapies for acute myeloid leukemia]. Rinsho Ketsueki. 2015 Feb;56(2):130-8. doi: 10.11406/rinketsu.56.130. Japanese. PubMed PMID: 25765792.

/////////Enasidenib, AG-221,

CC(O)(C)CNC1=NC(C2=NC(C(F)(F)F)=CC=C2)=NC(NC3=CC(C(F)(F)F)=NC=C3)=N1

Share

Regulatory Approval Pathways: EU vs US

 regulatory, Uncategorized  Comments Off on Regulatory Approval Pathways: EU vs US
Apr 202016
 

Regulatory Approval Pathways: EU vs US

 

Drug Authorization Procedures in the EU 

Sponsors have several options when seeking market approval for a new drug in Europe: a national authorization procedure, a decentralized procedure, a mutual recognition procedure and a centralized procedure. Depending on a product’s eligibility, each of these authorization routes offers different advantages and disadvantages to the sponsor, and these should be considered when setting up the market strategy of a product.

National Procedure

This procedure is used whenever a company wants to commercialize a product in only one EU Member State.

The National procedure is specific to each country. That is, each country within the EU has its own procedures for authorizing a marketing application for a new drug. Sponsors can find information regarding the requirements and procedure of each country on the websites of the regulatory agencies.

CREDIT….https://www.pda.org/pda-letter-portal/home/full-article/gmp-oversight-of-medicines-manufacturers-in-the-european-union

ADVANTAGES of National Procedure

There are some advantages in submitting a MAA through this procedure. First, it allows the sponsor to choose which country the company will submit to first. This is especially advantageous when the sponsor can’t afford to go through the centralized or decentralized procedure, due to lack of resources of distribution infrastructure for example. Choosing the country that the sponsor is most familiar with in regards to its regulation can also be an important factor.  The national authorization procedure also allows the sponsor to, further down the line, get his drug approved through the mutual recognition procedure, seeing as one country already approved its drug. Overall, this procedure is less resource heavy than the others, and thus it is the cheapest and safest alternative for a sponsor.

DISADVANTAGES of National Procedure

The disadvantages are obvious, seeing as this procedure only allows the sponsor to commercialize in one single market, cutting potential revenue streams it could have by bringing the drug to more markets.

Centralized procedure

The centralized procedure is a Europe wide authorization procedure, conducted by EMA’s Committee for Human Medicinal Products (CHMP), an organization which has representatives of all Member states, EEA members, patient organizations and health professionals.

When a sponsor applies for drug approval through the Centralized Procedure, two member states are first selected, a rapporteur and a co-rapporteur. These two member states will be responsible for the creation of an evaluation report that will be assessed by the CHMP.  First, a draft report is prepared and sent to the committee for review. The committee prepares a set of questions to send to the sponsor. After receiving a response, further discussions continue and a final evaluation report is arranged, containing a positive or negative opinion. This whole process can take up to 210 days. After the report is completed, it is sent to the European Commission in less than 15 days. The European Commission has the final say on the matter, granting the MA or not after evaluation of the CHMP’s report. The EC’s decision is applicable to all Member States of the European Union and EEA states – Iceland, Norway e Liechtenstein. After approval from the EC, the MA is valid for five years.

The centralized procedure, when it was introduced by Regulation (EEC) no 2309/93, followed the footsteps first established by Directive 87/22/EEC with its concertation procedure , and it was first made obligatory to products made from Recombinant DNA technology, controlled gene expression and monoclonal antibodies.

Afterwards, Regulation (EC) No 726/2004 extended the scope of the procedure to include orphan medicinal products and new active substances for the treatment of acquired immune deficiency syndrome (HIV), cancer, neurodegenerative disorder or diabetes. It went into force in 20th November 2005.

Recital 8 and Point 3 of the Annex to Regulation (EC) No 726/2004 also established that, starting 20 May 2008, the centralized procedure would be obligatory for drug products containing new active substances for the treatment of autoimmune diseases and other immune dysfunctions and viral diseases.

Lastly, regulation EC No 1394/2007 made the procedure compulsory for Advanced Therapy Medicinal products, like gene therapy, tissue engineered and somatic cell therapy products.

Article 3(2) of Regulation (EC) No 726/2004 defines the optional scope of the centralized procedure. It states that the procedure can be followed optionally by medicines that contain a new active substance, or if the applicant shows that the therapeutic entity provides a significant therapeutic, scientific or technical innovation, and it would be in the best interest of public health if it was approved at a community level.

ADVANTAGES of Centralized Procedure

Products authorized through the centralized procedure are granted marketing authorizations that cover all EU member states and the EEA, a big, 500 million user market where the sponsor can potentially recoup the losses from drug development. The drug will be commercialized in all countries with a single, unique brand name.

The convenience of the centralized procedure is however accompanied by fees that are significantly higher than the national procedure’s.

DISADVANTAGES of Centralized Procedure

Also, it is also a very risky, all or nothing procedure. If the CHMP refuses an application, the drug is barred from sale in every EU country, whereas if the sponsor tried another authorization procedure, there was the possibility of getting approval in at least one country. Since the sponsor can’t choose the rapporteur countries like he can in other procedures, this also leaves him at a disadvantage.

Mutual Recognition Procedure

This procedure requires the drug to be already approved in a MS.

This procedure is based upon the principle that a marketing authorization and the evaluation in one Member State (the so-called reference Member State) ought to be recognized by the competent authorities of the other Member States (the so-called concerned Member States), that is, if a Member State concedes a national MA to a drug, other Member States can recognize the evaluation conducted by it and grant a MA for the drug themselves.

It’s also noteworthy to point out that both a Member State and the Sponsor can trigger the Mutual Recognition Procedure.

After the first marketing authorization in the Community is granted, the marketing authorization holder may request one or more Member State(s) to recognize an authorization approved by the reference Member State, by submitting an application in accordance with Article 28 of Directive 2001/83/EC.

Within 90 days of receipt of a valid application, the reference Member State will provide the assessment report together with the approved summary of product characteristics, labeling and package leaflet to the concerned Member States and to the marketing authorization holder.

Within 90 days of the receipt of these documents, the concerned Member States shall recognize the decision of the reference Member State and the approved summary of product characteristics, package leaflet and labeling by granting a MA.

If any country refuses to grant a MA by safety reasons, the matter will be taken to The Co-ordination Group for Mutual Recognition and Decentralized Procedures, which will attempt to make all member states reach a consensus in 60 days. If it fails, the request will be taken to the CHMP and treated like a centralized procedure.

Decentralized procedure

The decentralized procedure works in a similar way as the mutual recognition one, except here the medicinal product in question has not yet received a marketing authorization in any Member State at the time of application. Like the MRP, a reference member state is chosen, which will evaluate the MAA. The remaining member states then proceed to give their opinion on the evaluation. If all concerned member states agree on the evaluation by the reference member state, the drug will be approved and allowed for sale in those countries. If a member state disagrees, the Co-ordination Group for Mutual Recognition and Decentralized Procedures will, like in the MRP, play a referee role.

ADVANTAGES and DISADVANTAGES of MRP & Decentralized Procedure

Both the MRP and the decentralized procedure carry a set of advantages and disadvantages that sponsors ought to know before setting their product market strategy. Both of them allow a sponsor to avoid the need to go through different national procedures in each country. Moreover, they aren’t as risky as the centralized procedure, and, in the case of the MRP, the sponsor can choose the reference member state that will conduct the evaluation of the drug product (by first attaining a MA in that country). In both these procedures, fees have to be paid to all Member states who participate in the process, and, unlike the centralized procedure, the sponsor may have to attribute a different name for its drug product in different Member States., which may hurt brand awareness.

The MRP often sees disagreements between member states, holding up the procedure and causing delays. In these occasions, a lengthy dispute solving mechanism has to be employed, costing both time and money to the sponsor

The decentralized procedure avoids some of the potential disputes between member states by engaging each of the member states the applicant wishes to apply to at the time the first marketing authorization is made. Disputes are this less common in the decentralized procedure than in the MRP. Lastly, the decentralized procedure is faster than the MRP.  The first can take up to 210 days to complete its two steps. The MRP, on the other hand, a national MA is first needed, which can take up to 210 days, alongside the update period of the MA license before the MRP procedure starts proper, which can take more 180 days. The take home message is that there is no one-size fits all in regards to drug authorization procedures. Each one of the four available has different advantages and disadvantages, which have to be carefully weighed out by the sponsor.

Drug Approval Process for the US

http://www.jpsr.pharmainfo.in/Documents/Volumes/vol5issue06/jpsr05061302.pdf

Types of Applications Submitted to the US FDA for New Medicines/Treatments

Investigational New Drug (IND) – Federal law requires that a drug be the subject of an approved marketing application before it is transported or distributed across state lines.

New Drug Application (NDA) – When the sponsor of a new drug believes that enough evidence on the drug’s safety and effectiveness has been obtained to meet FDA’s   requirements for marketing approval, the sponsor submits a new drug application (NDA) to FDA. The application must contain data from specific technical viewpoints for review, including chemistry, pharmacology, medical, biopharmaceutics, and statistics. If the NDA is approved, the product may be marketed in the United States.

Biologic License Application (BLA) – Biological products are approved for marketing     under   the provisions of the Public Health Service Act. The Act requires a firm who manufactures a    biologic for sale in interstate commerce to hold a license for the product. A biologics license   application is a submission that contains specific information on the manufacturing processes,  chemistry, pharmacology, clinical pharmacology and the medical effects of the biologic product. If the information provided meets FDA requirements, the application is approved and a license is issued allowing the firm to market the product.

US Drug Approval Process

If an IND drug survives the clinical trials (phase 1-3), an NDA is submitted to the FDA. An NDA contains all the preclinical and clinical information obtained during the testing phase. The application contains information on the chemical makeup and manufacturing process, pharmacology and toxicity of the compound, human pharmacokinetics, results of the clinical trials, and proposed labeling. An NDA can include experience with the medication from outside the United States as well as external studies related to the drug.

After receiving an NDA, the FDA completes an independent review and makes its recommendations. The Prescription Drug User Fee Act of 1992 (PDUFA) was designed to help shorten the review time. This act allowed the agency to collect user fees from pharmaceutical companies as financial support to enhance the review process. The 1992 Prescription Drug User Fee Act (PDUFA) established a two-tiered system – Standard Review and Priority Review.

Standard Review is applied to a drug that offers at most, only minor improvement over existing marketed therapies. The 2002 amendments to PDUFA set a 10 month goal for a standard review.

Priority Review designation is given to drugs that offer major advances in treatment, or provide a treatment where none existed. The goal for completing a Priority Review is six months.

If during the review the FDA staff feels there is a need for additional information or corrections, they will make a written request to the applicant. During the review process it is not unusual for the FDA to interact with the applicant staff.

The following four FDA programs are intended to facilitate and expedite development and review of new drugs to address unmet medical need in the treatment of a serious or life-threatening3 condition: fast track designation, breakthrough therapy designation, accelerated approval, and priority review designation.

Drug development in the fast lane: FDA approaches to expedited approval.

Fast track designation applies to the drug (either alone or in combination with other drugs) and the specific use for which it is being studied. The term drugrefers to the combination of two or more drugs if the combination is the subject of the fast track designation or request. Where appropriate, FDA may grant designation to the development of a new use of an approved drug.

  1. Serious Condition
  2. Demonstrating the Potential to Address Unmet Medical Need

The type of information needed to demonstrate the potential of a drug to address an unmet medical need will depend on the stage of drug development at which fast track designation is requested. Early in development, evidence of activity in a nonclinical model, a mechanistic rationale, or pharmacologic data could be used to demonstrate such potential. Later in development, available clinical data should demonstrate the potential to address an unmet medical need.

BREAKTHROUGH Therapy Designation

Section 506(a) of the FD&C Act provides for designation of a drug as a breakthrough therapy “. . . if the drug is intended, alone or in combination with 1 or more other drugs, to treat a serious or life-threatening disease or condition and preliminary clinical evidence indicates that the drug may demonstrate substantial improvement over existing therapies on 1 or more clinically significant endpoints, such as substantial treatment effects observed early in clinical development.” It is important to recognize that the standard for breakthrough therapy designation is not the same as the standard for drug approval. The clinical evidence needed to support breakthrough designation is preliminary. In contrast, as is the case for all drugs, FDA will review the full data submitted to support approval of drugs designated as breakthrough therapies to determine whether the drugs are safe and effective for their intended use before they are approved for marketing.

ACCELERATED APPROVAL

The accelerated approval provisions of FDASIA in section 506(c) of the FD&C Act provide that FDA may grant accelerated approval to:

. . . a product for a serious or life-threatening disease or condition . . . upon a determination that the product has an effect on a surrogate endpoint that is reasonably likely to predict clinical benefit, or on a clinical endpoint that can be measured earlier than irreversible morbidity or mortality, that is reasonably likely to predict an effect on irreversible morbidity or mortality or other clinical benefit, taking into account the severity, rarity, or prevalence of the condition and the availability or lack of alternative treatments.

For drugs granted accelerated approval, post marketing confirmatory trials have been required to verify and describe the anticipated effect on IMM or other clinical benefit

Post marketing surveillance is important, because even the most well-designed phase 3 studies might not uncover every problem that could become apparent once a product is widely used. Furthermore, the new product might be more widely used by groups that might not have been well studied in the clinical trials, such as elderly patients. A crucial element in this process is that physicians report any untoward complications. The FDA has set up a medical reporting program called Medwatch to track serious adverse events (1-800-FDA-1088). The manufacturer must report adverse drug reactions at quarterly intervals for the first 3 years after approval, including a special report for any serious and unexpected adverse reactions

Regulatory Links for the US FDA Guidances

Guidance for Industry -Expedited Programs for Serious Conditions – Drugs and Biologics, May 2014

http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm

Good Review Practice: Refuse to File, available on the Internet at http://www.fda.gov/downloads/aboutfda/centersoffices/officeofmedicalproductsandtobacco/cder/manualofpoliciesprocedures/ucm370948.htm and CBER SOPP 8404, Refusal to File Procedures for Biologic License Applications (August 27, 2007), available on the Internet athttp://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/ProceduresSOPPs/ucm073474.htm.

Regulatory Links for the EU:

Directive 2001/20/EC of the European Parliament and of the Council of 4 April2001 on the approximation of the laws, regulations and administrative provisions of the MS relating to the implementation of good clinical practice in the conduct of clinical trials on medicinal products for human use. http://eur-lex.europa.eu/LexUriServ/LexUriServ.douri=OJ:L:2001:121:0034:0044:en:PDF

Detailed guidance on the request to the competent authorities for authorization of a clinical trial on a medicinal product for human use, the notification of substantial amendments and the declaration of the end of the trial (CT-1) (2010/C 82/01) http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:C:2010:082:0001:0019:

EFPIA: Status of the implementation of the European Union Clinical Trials

Directive at member state level, Circular N° 12.784 , June 2008

Klingmann I et al. Impact on Clinical Research of European Legislation. Final report, February 2009http://www.efgcp.be/downloads/icrel_docs/Final_report_ICREL.pdf

Assessment of the functioning of the “Clinical Trials Directive” 2001/20/EC, Public Consultation Paper, ENTR/F/2/SF D(2009) 32674http://ec.europa.eu/enterprise/sectors/pharmaceuticals/files/clinicaltrials/docs/2009_ 10_09_public-consultation-paper.pdf

Report of the multidisciplinary workshop on “A single CTA in multinational clinical trials – dream or option?”, Brussels, Belgium, 7 July 2009http://www.efgcp.be/Conference_details.asp?id=265&L1=10&L2=2&TimeRef=2

Clinical Trials Facilitation Groups, Guidance document for a VoluntaryHarmonization Procedure (VHP) for the assessment of multinational Clinical Trial Applications, Version 2 ; Doc.ref.: CTFG/VHP/2010/Rev1, March 2010 http://www.hma.eu/uploads/media/VHP_version_2_March_2010.pdf

European Commission Enterprise Directorate-General. Detailed guidance on the application format and documentation to be submitted in an application for an Ethics Committee opinion on the clinical trial on medicinal products for human use (ENTR/CT2), Revision 1, February 2006http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/vol-10/12_ec_guideline_200 60216.pdf

The EFGCP Report on The Procedure for the Ethical Review of Protocols forClinical Research Projects in Europe, Update April 2010http://www.efgcp.be/EFGCPReports.asp?L1=5&L2=1

European Commission-European Medicines Agency Conference on the Operation of the Clinical Trials Directive (Directive 2001/20/EC) and Perspectives for the Future, Report on the Conference held on 3 October 2007 at the EMEA, London, Doc. ref.: EMEA/565466/2007http://www.eortc.be/services/doc/EUCTD/EC-EMEA_report_CT_20071003.pdf

Assessment of the functioning of the “Clinical Trials Directive” 2001/20/EC,Summary of responses to the public consultation paper, SANCO/C/8/SF/dn D(2010) 380240http://ec.europa.eu/enterprise/sectors/pharmaceuticals/files/clinicaltrials/2010_03_30_summary_responses.pdf

Directive 2001/83/EC of the European Parliament and of the Council of 6 November 2001 on the Community Code relating to Medicinal Products for Human Use, as amendedhttp://ec.europa.eu/enterprise/pharmaceuticals/eudralex/vol-1/dir_2001_83/dir_2001 _83_de.pdf

Responses to the Public consultation paper “Assessment of the functioning of the ‘Clinical Trials Directive’ 2001/20/EC”, March 2010http://ec.europa.eu/enterprise/sectors/pharmaceuticals/human-use/clinicaltrials/ developments/responses_2010-02_en.htm

Regulation (EC) No 1394/2007 of the European Parliament and of the Council of 13 November 2007 on advanced therapy medicinal products and amending Directive 2001/83/EC and Regulation (EC) No 726/2004 http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2007:324:0121:0137:

Commission Directive 2005/28/EC of 8 April 2005 laying down principles and detailed guidelines for good clinical practice as regards investigational medicinal products for human use, as well as the requirements for authorization of the manufacturing or importation of such products http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2005:091:0013:0019:

European Commission, Impact Assessment, 2010 Roadmaps “Legislative proposal on a Regulation/Directive amending the Clinical Trials Directive 2001/20/EC”, Version 2, 23/03/2010http://ec.europa.eu/governance/impact/planned_ia/docs/47_sanco_clinical_trials_directive_en.pdf

 

//////////Regulatory Approval Pathways,  EU vs US

Share

PF 14

 Uncategorized  Comments Off on PF 14
Apr 202016
 

str1

SCHEMBL15754248.png

PF 14

Molecular Formula: C14H14N4O2S
Molecular Weight: 302.35156 g/mol

6-[(4R)-4-methyl-1,1-dioxo-1,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile

1612755-71-1  CAS

The androgen receptor (“AR”) is a ligand-activated transcriptional regulatory protein that mediates induction of male sexual development and function through its activity with endogenous androgens. Androgenic steroids play an important role in many physiologic processes, including the development and maintenance of male sexual characteristics such as muscle and bone mass, prostate growth, spermatogenesis, and the male hair pattern. The endogenous steroidal androgens include testosterone and dihydrotestosterone (“DHT”). Steroidal ligands which bind the AR and act as androgens (e.g. testosterone enanthate) or as antiandrogens (e.g. cyproterone acetate) have been known for many years and are used clinically.

PATENT

WO 2015173684

http://www.google.com/patents/WO2015173684A1?cl=en

The androgen receptor (“AR”) is a ligand-activated transcriptional regulatory protein that mediates induction of male sexual development and function through its activity with endogenous androgens. Androgenic steroids play an important role in many physiologic processes, including the development and maintenance of male sexual characteristics such as muscle and bone mass, prostate growth,

spermatogenesis, and the male hair pattern. The endogenous steroidal androgens include testosterone and dihydrotestosterone (“DHT”). Steroidal ligands which bind the AR and act as androgens (e.g. testosterone enanthate) or as antiandrogens (e.g.

cyproterone acetate) have been known for many years and are used clinically.

6-[(4f?)-4-Methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile (Formula I), in its free base form, has the chemical formula C14H14N4SO2 and the following structural formula:

Formula I

Synthesis of 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile is disclosed in co-pending international patent application,

PCT/IB2013/060381 , filed 25th November 2013, and published as WO 2014/087298 on 12th June 2014, assigned to the assignee of the present invention and which is incorporated herein by reference in its entirety. 6-[(4f?)-4-Methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile is known to be active as a selective androgen receptor modulator (SARM) and, as such, is useful for treating and/or preventing a variety of hormone-related conditions, for example, conditions associated with androgen decline, such as, inter alia, anaemia; anorexia; arthritis; bone disease; musculoskeletal impairment; cachexia; frailty; age-related functional decline in the elderly; growth hormone deficiency; hematopoietic disorders; hormone replacement; loss of muscle strength and/or function; muscular dystrophies; muscle loss following surgery; muscular atrophy; neurodegenerative disease; neuromuscular disease;

obesity; osteoporosis; and, muscle wasting.

Identification of new solid forms of a known pharmaceutical active ingredient provide a means of optimising either the physicochemical, stability, manufacturability and/or bioperformance characteristics of the active pharmaceutical ingredient without modifying its chemical structure. Based on a chemical structure, one cannot predict with any degree of certainty whether a compound will crystallise, under what conditions it will crystallise, or the solid state structure of any of those crystalline forms. The specific solid form chosen for drug development can have dramatic influence on the properties of the drug product. The selection of a suitable solid form is partially dictated by yield, rate and quantity of the crystalline structure. In addition, hygroscopicity, stability, solubility and the process profile of the solid form such as compressibility, powder flow and density are important considerations.

As such, there is a need to identify solid forms of 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazi

Example 1

Procedure:

Into a 2L 3-neck round bottom flask equipped with a mechanical stirrer, reflux condenser and thermocouple with heating mantle was placed 2-methyltetrahydrofuran (2-MeTHF) (10 mL/g; 8.15 moles; 817 ml_; 702 g) followed by racemic-2,2′-bis(diphenylphosphino)-1 ,1 ‘-binaphthyl (BINAP) (0.04 equiv (molar); 14.0 mmol; 8.74 g) and bis(dibenzylideneacetone)palladium (Pd2(dba)3) (0.04 equiv (molar); 14.0 mmol;

8.07 g). The mixture was degassed by pulling vacuum and refilling with nitrogen three times then heated to 75 °C for 15 minutes and cooled to ambient temperature. In a separate flask, (S)-3-amino-2-methylpropan-1-ol (1.60 equiv; 561 mmol; 50.0 g, prepared using literature methods, for example as disclosed in EP-A-0,089, 139 published on 21st September 1983) was dissolved in 2-methyltetrahydrofuran (5 ml_/g;

4.08 moles; 409 ml_; 351 g) and degassed by pulling vacuum and refilling with nitrogen three times. Into the pot containing the catalyst was added 6-(bromoisoquinoline-1- carbonitrile) (1.00 equiv; 351 mmol; 81.75 g) and cesium carbonate (1.6 equiv (molar); 561 mmol; 185 g) in single portions followed by the solution of the aminoalcohol via addition funnel. The reaction mixture was again degassed by pulling vacuum and refilling with nitrogen three times. The reaction was heated to 70 °C for 3 hours. The reaction was cooled to ambient temperature and filtered through a pad of Celite. The contents of the flask were rinsed out with three 100 mL portions of 2-methyltetrahydrofuran. The filtrate was transferred into a 2L round bottom flask equipped with a thermocouple and mechanical stirrer under nitrogen. Silica Gel (Silicylate SiliaMet® Thiol) (0.4 g/g-pure-LR; 544 mmol; 32.7 g) was charged and the flask was stirred at 40 °C overnight. The following morning, the reaction was cooled to < 30 °C and filtered again through Celite. The pad was washed with 100ml_ of 2-methyltetrahydrofuran (or until no yellow color persisted in the filtrate). The filtrate was placed into a 3L round bottom flask equipped with a magnetic stir bar, distillation head (with condenser and receiving flask), and thermocouple. The mixture was heated to 60 °C and placed under vacuum (-450-500 mbar) to distil out 1.3 L total of 2-methyltetrahydrofuran. 500 mL of toluene was added to precipitate the desired product. The heating mantle was removed and the reaction was allowed to reach ambient temperature. The mixture was stirred for 1 hour at ambient temperature and then the solids were collected by vacuum filtration on a sintered glass funnel. The cake was dried overnight on the funnel under vacuum. The following morning, the solids were transferred into an amber bottle and weighed (71.9 g; 298 mmol). The product was used in the next step without further purification.

Example 2

Procedure:

In a 1 L reactor equipped with a temperature probe and overhead stirring was added the product of Example 1 (20.0 g; 1.00 equiv; 82.9 mmol) and 2-methyltetrahydrofuran (2-MeTHF) (30 mL/g-pure-LR; 5.98 moles; 600 mL; 515 g). The reaction mixture was

gently warmed to 40°C to achieve partial solubility. The reaction was cooled to 0°C. Once the reaction reached 0°C methanesulfonyl chloride (MsCI) (1.4 equiv (molar); 1 16 mmol; 8.98 mL; 13.3 g) was added in a single portion followed immediately by triethylamine (TEA) (1.4 equiv (molar); 116 mmol; 16.2 mL; 11.7 g) dropwise via syringe over a period of 15 minutes. The reaction mixture was further stirred for 30 min at 0°C and then warmed to 23°C for 60 minutes. The product (26.47 g; 1.00 equiv; 82.88 mmol; 26.47 g; 100% assumed yield) was then used without purification for the sulfonylation reaction.

Example 3

t-BuOH, 2-MeTHF

o 0 °C to 23 °C o

CI-S-N=C=0 CI-S-NHBoc

0 O

Procedure:

To a solution of t-butyl alcohol (t-BuOH) (1 equiv (molar); 116 mmol; 1 1.0 mL; 8.60 g) in 2-methyltetrahydrofuran (2-MeTHF) (1 M; 1.16 moles; 116 mL; 99.6 g) at 0°C was added chlorosulfonyl isocyanate (116 mmol; 1.00 equiv; 10.1 mL; 16.4 g) dropwise. The homogeneous solution was stirred for 30 minutes at ambient temperature and then used directly in the sulfonylation reaction.

Example 4

Sulfonylation Reaction Procedure:

A previously prepared solution of the product of Example 3 (1.4 equiv (molar); 1 16 mmol; 116 g) in 2-methyltetrahydrofuran was added to a suspension of the product of Example 2 (1.00 equiv; 82.89 mmol; 26.5 g) at 0°C. The mixture was warmed to ambient temperature over 30 minutes. HPLC analysis revealed the reaction was complete. The reaction was quenched with a 10% sodium carbonate solution (2 equiv

(molar); 165 mmol; 101 mL; 1 17 g) and water (to dissolve salts) (5 L/kg; 7.35 moles; 132 mL; 132 g). The top organic layer was removed and passed through a plug of Carbon (Darco G60) (0.5 g/g) on a filter. A significant improvement in color (dark orange to yellow) was observed. The solution was concentrated to 10 total volumes and used in the next step without purification.

Example 5

Procedure:

A solution of the product of Example 4 (1.OOequiv; 82.9 mmol; 41.3 g) in 2-methyltetrahydrofuran (2-MeTHF) (10ml_/g; 4.12 moles; 413 mL; 355 g) was placed into a 1 L reactor equipped with an overhead stirrer and temperature probe. Next, potassium carbonate (K2CO3) (325 mesh) (6 equiv (molar); 497 mmol; 69.4 g) and water (0.0 L/100-g-bulk-LR; 459 mmol; 8.26 mL; 8.26 g) were added and the mixture heated to 40°C (jacket temperature) and stirred overnight. The reaction was cooled to ambient temperature and water (4L/kg-pure-LR; 9.17 moles; 165 mL; 165 g]) was added. The biphasic reaction was stirred for 1 hour at 23 °C. The aqueous layer was extracted and removed. The organic layer was passed through a plug of Carbon (Darco G60) (0.5 g/g-pure-LR; 20.7g) in a disposable filter. The 2-methyltetrahydrofuran solution was switched to a 10 volume solution of toluene via a constant strip-and-replace distillation to no more than 1 % 2-methyltetrahydrofuran. The toluene solution of the reaction product (1.00 equiv; 82.9 mmol; 33.4 g; 100% assumed yield) was used as-is in the next step without further purification.

Example 6

Procedure:

To a 1 L reactor under nitrogen and equipped with overhead stirring and a temperature probe was added the product of Example 5 (1.00 equiv; 78.7 mmol; 33.4 g) as a solution in toluene (10 mL/g-pure-LR; 3.00 moles; 317 ml_; 276 g). Next, trifluoroacetic acid (TFA) (10 equiv (molar); 787 mmol; 59.5 ml_; 89.8 g) was added to the reaction over a period of 1 hour keeping the internal temperature below 30°C. The dark red mixture was stirred for 1 hour. The reaction was quenched at 23 °C by the addition of sodium carbonate (5 equiv (molar); 394 mmol; 240 ml_; 278 g). The reaction was quenched slowly, over a period of 1 hour to form the TFA salt of the product. Once the charge was complete, the mixture was cooled to 0°C, held for 1 hour and filtered. The next morning, the solid product (6-[(4R)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile in its free base form) was weighed (0.89 equiv; 70.0 mmol; 21.2 g; 89.0% yield) and used in the next step without further purification.

Example 7

Crystalline 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile free base (Form (1)) was prepared as follows.

In a 1 L 3-neck round bottom flask was added 6-[(4R)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile free base (1.00 equiv; 70.0 mmol; 21.2 g) a magnetic stir bar and acetone (40ml_/g; 1 1.5 moles; 847 ml_; 669 g). The mixture was heated to reflux (approximately 57°C) and stirred for 1 hour. The mixture was concentrated by atmospheric distillation (heating mantle set at 65°C) and 40ml_ of acetone was collected into a graduated cylinder. Next, water (25 mL/g; 29.4 moles; 530 ml_; 530 g) was charged over a period of one hour. The mixture was stirred at ambient temperature for 60min before being cooled to 0°C at 1 °C /min for 1 hour. The solids were collected by filtration in a disposable funnel. Crystalline 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile (Form (1), 0.88 equiv; 61.9 mmol; 18.7 g; 88.3% yield) was dried under vacuum overnight at 40 °C. Typical purity after crystallization is 98%.

PATENT

US 20140155390

http://www.google.com/patents/US20140155390

PATENT

WO 2015181676

http://www.google.com/patents/WO2015181676A1?cl=en

xample 9

6-[(3S)-3-methyl-1 , 1 -dioxido-1 ,2,5-thiadiazolidin-2-yl1naphthalene-1 -carbonitrile

(stereochemistry is arbitrarily assigned)

LCMS m/z = 286.0 (M – H). 1 H NMR (400 MHz, cf6-DMSO): δ 1 .31 (d, J = 6.2 Hz, 3H), 3.13 – 3.25 (m, 1 H), 3.71 (dt, J = 12.5, 6.8 Hz, 1 H), 4.49 – 4.62 (m, 1 H), 7.62 – 7.70 (m, 1 H), 7.75 – 7.83 (m, 2H), 7.99 (t, J = 7.8 Hz, 1 H), 8.07 (d, J = 6.6 Hz, 1 H), 8.14 (d, J = 8.9 Hz, 1 H), 8.28 (d, J = 8.4 Hz, 1 H). Chiral HPLC purity: 99.1 % (retention time 17.12 minutes)

Step 1. Synthesis of aminoester (#D1). Thionylchlride (8.5 ml_, 1 16.5 mmol) was added to the solution of amino acid (4.0 g, 38.8 mmol) in MeOH (170 ml_) at 0 °C, and the reaction mixture was stirred for 6 h at room temperature. The reaction was monitored by TLC, and after disappearance of the starting material it was cooled to room temperature and solid NaHC03 was added. The reaction mixture was filtered, concentrated in vacuo and the resulting residue was triturated with diethyl ether to obtain crude #D1 (4 g, 90%) as a white solid. Rf: 0.4 (f-BuOH: AcOH: H20 (4:0.5:0.5)).

GCMS m/z = 1 17.1 (M). 1H NMR (400 MHz, cf6-DMSO): δ 1.17 (d, J = 6.8 Hz, 3H), 2.83 – 2.88 (m, 2H), 3.03 – 3.05 (m, 1 H), 3.65 (s, 3H), 8.02 – 8.30 (br s, 3H).

Step 2. Synthesis of aminoalcohol (#D2). #D1 (2.0 g, 13.0 mmol) was added

portionwise to a suspension of LiAIH4 (1.4 g, 39.2 mmol) in THF (75 ml_) under nitrogen atmosphere at 0 °C. The reaction mixture was stirred for 30 minutes and then allowed to stir at room temperature for another 30 minutes. The reaction mixture was refluxed for 2 h, and then it was cooled to -10 °C and quenched carefully with ice cold water (1.4 ml_). 10% NaOH solution (2.8 ml_) and ice cold water (4.2 ml_) were added, and the mixture was stirred for 15 minutes. It was filtered, and the filtrate washed with EtOAc (3 x 100 ml_), dried over anhydrous Na2S04 and concentrated under vacuum to obtain #D2 (1.2 g, 86%) as a pale yellow liquid. Rf: 0.2 (20% MeOH in DCM).

1H NMR (400 MHz, cf6-DMSO): δ 0.78 (d, J = 6.8 Hz, 3H), 1.46 – 1.54 (m, 1 H), 2.41 -2.45 (m, 2H), 2.50 – 2.54 (m, 1 H), 3.22 – 3.34 (m, 4H).

Step 3. Synthesis of coupling product (#D3). K3P04 (6.1 g, 28.8 mmol), BINAP (0.44 g, 0.72 mmol) and Pd2(dba)3 (0.32.0 g, 0.36 mmol) was added to the degassed

suspension of 6-bromo-1 -cyanoisoquinoline #A3 (1.7 g, 7.2 mmol), #D2 (1.2 g, 14.5 mmol) in DMSO at room temperature. The reaction mixture was heated at 105 °C for 2 h. The reaction was cooled to room temperature, water (500 ml_) followed by EtOAc (100 ml_) were added, and the mixture was stirred for 10 minutes. The biphasic mixture was filtered through a Celite™ pad and washed with EtOAc (100 ml_). The organic layer was separated, and the aqueous layer was extracted with EtOAc (3 x 100 ml_). The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure to get a crude material. This was purified by column chromatography on 100 – 200 mesh silica gel, using 50 – 70% EtOAc in petroleum ether as the eluent to obtain #D3 (0.5 g, 48.5%) as a yellow solid. Rf: 0.4 (60% EtOAC in petroleum ether).

LCMS m/z = 242.0 (M + H). 1 H NMR (400 MHz, cf6-DMSO): δ 0.97 (d, J = 6.4 Hz, 3H), 1.87 – 1.99 (m, 1 H), 2.92 – 2.99 (m, 1 H), 3.20 – 3.27 (m, 1 H), 3.38 – 3.42 (m, 2H), 4.59 (t, J = 5.2 Hz, 1 H), 6.77 (d, J = 2.0, 1 H), 7.01 (t, J = 5.6 Hz, 1 H), 7.34 (dd, J = 9.2 Hz, J = 2.0 Hz, 1 H), 7.73 (d, J = 6.0 Hz, 1 H), 7.88 (d, J = 8.8 Hz, 1 H), 8.312 (d, J = 6.0 Hz, 1 H).

Step 4. Methanesulfonated coupling product (#D4). Triethylamine (0.44 mL, 3.1 mmol) was added to a solution of #D3 (0.50 g, 2.0 mmol) in DCM at 0 °C.

Methanesulfonylchloride (0.25 mL, 3.1 mmol) was added over 10 minutes, and the reaction mixture was stirred for 1 h at room temperature. After disappearance of the starting material by TLC, it was diluted with DCM and washed with water. The organic layer was separated, dried over Na2S04, concentrated under reduced pressure to obtain crude #D4 (0.6 g, crude) as yellow solid. This was used for next step without any purification. Rf: 0.6 (50% EtOAc in petroleum ether).

LCMS m/z = 320.0 (M + H). 1 H NMR (400 MHz, CDCI3): δ 1.17 (d, J = 6.8 Hz, 3H), 2.32 – 2.37 (m, 1 H), 3.06 (s, 3H), 3.26 – 3.41 (m, 2H), 4.16 – 4.20 (m, 1 H), 4.33 – 4.37 (m, 1 H), 4.75 (br s, 1 H), 6.70 (d, J = 2.4, 1 H), 7.09 (dd, J = 9.2 Hz, 2.4 Hz, 1 H), 7.57 (d, J = 6.0 Hz, 1 H), 8.05 (d, J = 9.2 Hz, 1 H), 8.39 (d, J = 5.6 Hz, 1 H).

Step 5. Cyclized and uncyclized intermediates (#D5, #D6). Chlorosulfonylisocyanate (1.2 mL, 13.1 mmol) was added dropwise to a solution f-BuOH (1.4 mL, 13.1 mmol) in toluene (4.0 mL) at -5 °C. The reaction mixture was stirred at room temperature for 20 minutes, and then THF (1 mL) was added to the resulting suspension to obtain clear solution. In another flask, DIPEA (2.3 mL, 13.1 mmol) was added to a solution of #D4 (0.6 g, crude 2.6 mmol) in dry THF (3 mL). The above prepared reagent (CIS02NH-Soc) was added to this reaction mixture dropwise at room temperature over a period of 20 minutes. The resulting reaction mixture was then stirred for 16 h at room temperature. The mixture was diluted with EtOAc (100 mL) and washed with water (100 mL). The aqueous layer was washed with EtOAc (2 x 100 mL), combined all the organic layers, dried over Na2S04, concentrated under reduced pressure to obtain the crude product (LCMS shows desired #D6 and uncyclized #D5. This crude was purified by column chromatography on 100 – 200 mesh silica gel, using 10 – 30% EtOAc in petroleum ether as an eluent to obtain desired #D6 (0.35 g, 47.8%), and uncyclized #D5 (0.22 g, crude).

The uncyclized #D5 (0.22 g, crude) was dissolved in THF (1 mL) and DIPEA (0.6 mL) was added to the solution. The reaction mixture was stirred for another 12 h at room temperature. After which time, it was diluted with EtOAc (100 mL) and washed with water (100 mL). The aqueous layer was washed with EtOAc (2 x 100 mL), combined all the organic layers, dried over Na2S04, concentrated under reduced pressure to obtain crude product. This crude was purified by column chromatography on 100 – 200 mesh silica gel, using 10 – 30% EtOAc in petroleum ether as an eluent to obtain desired #D6 (1 .1 g, 13.2%). Total amount of #D6 was (0.5 g, 60% for two steps, 82% LCMS purity). Rf: 0.8 (60% EtOAc in petroleum ether).

LCMS m/z = 403.1 (M + H). 1 H NMR (400 MHz, CDCI3): δ 1 .04 (d, J = 6.8 Hz, 3H), 1 .50 (s, 9H), 2.38 – 2.48 (m, 1 H), 3.65 – 3.82 (m, 2H), 3.92 – 4.02 (m, 1 H), 4.30 – 4.38 (m, 1 H), 7.79 – 7.81 (m, 1 H), 7.86 – 7.88 (m, 2H), 8.34 – 8.37 (d, J = 9.2 Hz, 1 H), 8.67 (d, J = 6.0 Hz, 1 H).

Step 6. Racemate #D7 and final products (#10, #11 ). TFA (5 mL) was added to a solution of #D6 (0.15 g, 0.37 mmol) in DCM (100 mL) at 0 °C. The reaction mixture was stirred for 1 h at 0 °C. The solution was neutralized with saturated aqueous NaHC03 solution at 0 °C. The mixture was diluted with water, extracted with DCM (3 x 100 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure to obtain racemic #D7 (0.10 mg, 73%).

LCMS m/z = 303.0 (M + H). Rf: 0.3 (60% EtOAc in petroleum ether).

Enantiomeric separation: #D7 was submitted for chiral separation to obtain final compounds #10 (0.015 mg) and #11 (0.016 mg).

Column: CHIRALPAK IA, 4.6 χ 250 mm, 5 m; Mobile phase: n-Hexane/ /-PrOH/DCM (60%/15%/15%); Flow rate: 0.8 mL/min.

Example 10

6-[(4R)-4-methyl-1 , 1 -dioxido-1 ,2,6-thiadiazinan-2-yl1isoquinoline-1 -carbonitrile (#10; R = (R)-CH3)

LCMS m/z = 303.0 (M + 1 ). 1 H NMR (400 MHz, cf6-DMSO): δ 0.98 (d, J = 6.4 Hz, 3H), 2.22 – 2.26 (m, 1 H), 3.16 – 3.22 (m, 1 H), 3.34 – 3.39 (m, 1 H), 3.59 – 3.65 (m, 1 H), 3.77 – 3.81 (m, 1 H), 7.75 – 7.79 (m, 1 H, disappeared in D20 exchange), 7.95 (dd, J = 8.8 Hz, J = 2.0 Hz, 1 H), 8.06 (d, J = 1 .6 Hz, 1 H), 8.23 – 8.27 (m, 2H), 8.703 (d, J = 5.2 Hz, 1 H). Rf: 0.3 (60% EtOAc in petroleum ether). Chiral HPLC purity: 98.2% (retention time 1 1 .43 minutes).

Patent ID Date Patent Title
US2014155390 2014-06-05 NOVEL SELECTIVE ANDROGEN RECEPTOR MODULATORS

//////////pf 14,

C[C@H]3CN(c1cc2ccnc(C#N)c2cc1)S(=O)(=O)NC3

Share

ND 0126

 Uncategorized  Comments Off on ND 0126
Apr 192016
 

SCHEMBL3808941.png

Figure imgf000102_0003

ND 0126

CAS 1240322-54-6

Molecular Formula: C29H25F3N6O3
Molecular Weight: 562.54241 g/mol

methyl 5-[[2-methyl-5-[[3-(4-methylimidazol-1-yl)-5-(trifluoromethyl)benzoyl]amino]phenyl]methylamino]-1H-pyrrolo[2,3-b]pyridine-2-carboxylate

5-{2-Methyl-5-[3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzoylamino]-benzylamino}-1H-pyrrolo[2,3-b]pyridine-2-carboxylic Acid Methyl Ester

Oribase Pharma

Nova Decision, Azasynth

Potent dual ABL​/SRC inhibitors based on a 7-​azaindole core with the aim of developing compds. that demonstrate a wider activity on selected oncogenic kinases.  Multi-​Targeted Kinase Inhibitors (MTKIs) were then derived, focusing on kinases involved in both angiogenesis and tumorigenesis processes.

Dysfunction/deregulation of protein kinases (PK) is the cause of a large number of pathologies including oncological, immunological, neurological, metabolic and infectious diseases. This has generated considerable interest in the development of small molecules and biological kinase inhibitors for the treatment of these disorders.

Numerous PK are particularly deregulated during the process of tumorigenesis. Consequently protein kinases are attractive targets for anticancer drugs, including small molecule inhibitors that usually act to block the binding of ATP or substrate to the catalytic domain of the tyrosine kinase and monoclonal antibodies that specifically target receptor tyrosine kinases (RTK) and their ligands. In solid malignancies, it is unusual for a single kinase abnormality to be the sole cause of disease and it is unlikely that tumors are dependent on only one abnormally activated signaling pathway. Instead multiple signaling pathways are dysregulated. Furthermore, even single molecular abnormalities may have multiple downstream effects. Multi targeted therapy using a single molecule (MTKI = “Multi-Targeted Kinase Inhibitors”) which targets several signaling pathways simultaneously, is more effective than single targeted therapy. Single targeted therapies have shown activity for only a few indications and most solid tumors show deregulation of multiple signaling pathways. For example, the combination of a vascular endothelial growth factor receptor (VEGFR) inhibitor and platelet derived growth factor receptor (PDGFR) inhibitor results in a cumulative antitumor efficacy (Potapova et al, Mol Cancer Ther 5, 1280-1289, 2006).

Tumors are not built up solely of tumor cells. An important part consists of connective tissue or stroma, made up of stromal cells and extracellular matrix, which is produced by these cells. Examples of stromal cells are fibroblasts, endothelial cells and macrophages. Stromal cells also play an important role in the carcinogenesis, where they are characterized by upregulation or induction of growth factors and their receptors, adhesion molecules, cytokines, chemokines and proteolytic enzymes (Hofmeister et al., Immunotherapy 57, 1-17, 2007; Raman et al, Cancer Letters 256, 137-165, 2007; Fox et al, The Lancet Oncology 2, 278-289, 2001) The receptor associated tyrosine kinase VEGFR on endothelial and tumor cells play a central role in the promotion of cancer by their involvement in angiogenesis (Cebe-Suarez et al, Cell Mol Life Sci 63, 601-615, 2006). In addition, the growth factors TGF-β, PDGF and FGF2 secreted by cancer cells transform normal fibroblasts into tumor associated fibroblasts, which make their receptors a suitable target for inhibition by kinase inhibitors (Raman et al, 2007).

Moreover, increasing evidence suggests a link between the EGF receptor (EGFR) and HER2 pathways and VEGF-dependent angiogenesis and preclinical studies have shown both direct and indirect angiogenic effects of EGFR signaling (Pennell and Lynch, The Oncologist 14, 399-411, 2009). Upregulation of tumor pro -angiogenic factors and EGFR- independent tumor-induced angiogenesis have been suggested as a potential mechanism by which tumor cells might overcome EGFR inhibition. The major signaling pathways regulated by EGFR activation are the PI3K, MAPK and Stat pathways that lead to increased cell proliferation, angiogenesis, inhibition of apoptosis and cell cycle progression. EGFR is overexpressed in a wide variety of solid tumors, such as lung, breast, colorectal and cancers of the head and neck (Cook and Figg, CA Cancer J Clin 60, 222-243 2010). Furthermore, higher expression of EGFR has been shown to be associated with metastasis, decreased survival and poor prognosis.

c-Src, a membrane-associated non receptor tyrosine kinase, is involved in a number of important signal transduction pathways and has pleiotropic effects on cellular function. c-Src integrates and regulates signaling from multiple transmembrane receptor-associated tyrosine kinases, such as the EGFR, PDGFR, IGF1R, VEGFR, HER2. Together, these actions modulate cell survival, proliferation, differentiation, angiogenesis, cell motility, adhesion, and invasion (Brunton and Frame, Curr Opin Pharmacol 8, 427-432, 2008). Overexpression of the protein c-Src as well as the increase in its activity were observed in several types of cancers including colorectal, gastrointestinal (hepatic, pancreatic, gastric and oesophageal), breast, ovarian and lung (Yeatman, Nat Rev Cancer 4, 470-480, 2004).

The activation in EGFR or KRAS in cancers leads to a greatly enhanced level of Ras- dependent Raf activation. Hence, elimination of Raf function is predicted to be an effective treatment for the numerous cancers initiated with EGFR and KRAS lesions (Khazak et al, Expert Opin. Ther. Targets 11, 1587-1609, 2007). Besides activation of Raf signaling in tumors, a number of studies implicate the activation of the Ras-Raf-MAPK signaling pathway as a critical step in vasculo genesis and angiogenesis. Such activation is induced by growth factor receptors such as VEGFR2, FGFR2 and thus inhibition of Raf activation represents a legitimate target for modulation of tumor angiogenesis and vascularization.

Although VEGFR, PDGFR, EGFR, c-Src and Raf are important targets on both tumor cells and tumor stroma cells, other kinases such as FGFR only function in stromal cells and other oncogenes often only function in tumor cells.

Protein kinases are fundamental components of diverse signaling pathways, including immune cells. Their essential functions have made them effective therapeutic targets. Initially, the expectation was that a high degree of selectivity would be critical; however, with time, the use of “multikinase” inhibitors has expanded. Moreover, the spectrum of diseases in which kinase inhibitors are used has also expanded to include not only malignancies but also immune-mediated diseases / inflammatory diseases. The first step in signaling by multi-chain immune recognition receptors is mediated initially by Src family protein tyrosine kinases. MTKI targeting kinases involved in immune function are potential drugs for autoimmune diseases such as rheumatoid arthritis, psoriasis and inflammatory bowel diseases (Kontzias et al. , F 1000 Medicine Reports 4, 2012)

Protein kinases mentioned previously are also key components of many other physiological and pathological mechanisms such as neurodegeneration and neuroprotection (Chico et al, Nature Reviews Drug Discovery 8, 892-909, 2009), atherosclerosis, osteoporosis and bone resorption, macular degeneration, pathologic fibrosis, Cystogenesis (human autosomal dominant polycystic kidney disease…).

In WO2010/092489 and related patents/patent applications, we identified several compounds which exhibited interesting properties for such applications. However, we have discovered that some of these compounds could be enhanced in their properties by selectively working on particular regions of their structures. However, the mechanism of action of these structures on kinases was not precisely elucidated at the time of WO2010/092489’s filing and thus it was unexpectedly that we found the high activities of the structures disclosed in the present application. The subject matter of the present invention is to offer novel multi-targeted kinase inhibitors, having an original backbone, which can be used therapeutically in the treatment of pathologies associated with deregulation of protein kinases including tumorigenesis, human immune disorders, inflammatory diseases, thrombotic diseases, neurodegenerative diseases, bone diseases, macular degeneration, fibrosis, cystogenesis. The inhibitors of the present invention can be used in particular for the treatment of numerous cancers and more particularly in the case of liquid tumors such hematological cancers (leukemias) or solid tumors including but not limited to squamous cell cancer, small- cell lung cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, glial cell tumors such as glioblastoma and neurofibromatosis, cervical cancer, ovarian cancer, liver cancer, bladder cancer, breast cancer, melanoma, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, renal cancer, prostate cancer, vulval cancer, thyroid cancer, sarcomas, astrocytomas, and various types of hyperproliferative diseases.

 

 

Abstract Image

Efforts were made to improve a series of potent dual ABL/SRC inhibitors based on a 7-azaindole core with the aim of developing compounds that demonstrate a wider activity on selected oncogenic kinases. Multi-targeted kinase inhibitors (MTKIs) were then derived, focusing on kinases involved in both angiogenesis and tumorigenesis processes. Antiproliferative activity studies using different cellular models led to the discovery of a lead candidate (6z) that combined both antiangiogenic and antitumoral effects. The activity of 6z was assessed against a panel of kinases and cell lines including solid cancers and leukemia cell models to explore its potential therapeutic applications. With its potency and selectivity for oncogenic kinases, 6z was revealed to be a focused MTKI that should have a bright future in fighting a wide range of cancers.

 

5-{2-Methyl-5-[3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzoylamino]-benzylamino}-1H-pyrrolo[2,3-b]pyridine-2-carboxylic Acid Methyl Ester (6z)

The reaction was carried out as described in general procedure A using 4a (170 mg, 0.63 mmol), 3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzoic acid 5z (200 mg, 0.63 mmol), HATU (735 mg, 1.93 mmol), DIEA (0.56 mL, 3.22 mmol), and anhydrous DMF (16 mL). Purification by flash chromatography on silica gel (EtOAc/EtOH, 100/0 to 90/10) yielded 6z (108 mg, 30%).
1H NMR (300 MHz, DMSO-d6, δ) 12.05 (s, 1H), 10.41 (s, 1H), 8.42–8.34 (m, 2H), 8.20 (s, 1H), 8.16–8.04 (m, 2H), 7.670–7.62 (m, 3H), 7.22 (d, J = 8.2 Hz, 1H), 6.97 (d, J = 2.3 Hz, 1H), 6.90 (d, J = 1.9 Hz, 1H), 6.11 (t, J = 5.0 Hz, 1H), 4.25 (d, J = 5.0 Hz, 2H), 3.83 (s, 3H), 2.34 (s, 3H), 2.17 (s, 3H). MS (ESI) m/z 563.2 [M + H]+ and 561.2 [M – H].

Rational Design, Synthesis, and Biological Evaluation of 7-Azaindole Derivatives as Potent Focused Multi-Targeted Kinase Inhibitors

OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, Montpellier 34189 CEDEX 4,France
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.6b00087
Publication Date (Web): March 24, 2016
Copyright © 2016 American Chemical Society
*E-mail: ayasri@oribase-pharma.com. Phone: (+33) 467 727 670.
PATENT
WO 2010092489

https://www.google.com/patents/WO2010092489A1?cl=en

Example 91: Preparation of methyl 5-(5-(3-(trifluoromethγl)-5~(4-methyl-1 H-imidazol-1 – yl)benzamido)-2-methγlbenzylamino)-1H-pyrrolo[2,3-blpyridine-2-carboχylate (ND0126)

Step 1 : preparation of methyl 5-(3-(trifluoromethyl)-5-(4-methyl-1 H-imidazol-1 – yl)benzamido)-2-methylbenzoate

Figure imgf000102_0001

The compound is obtained using the procedures of example 88 (step 4) replacing the 4-((3-(dimethylamino)pyrrolidin-1-yl)methyl)-3-(trifluoromethyl)-benzoic acid

(Shakespeare W. C, WO2007133562) by the 3-(trifluoromethyI)-5-(4-methyl-1H- imidazol-1-yl)benzoic acid.

Step 2: preparation of 3-(tπϊluoromethyl)-N-(3-formyl-4-methylphenyl)-5-(4- methyl-1H-imidazol-1-yl)benzamide

Figure imgf000102_0002

The compound is obtained by using the procedures of examples 83 (steps 1 and 2) replacing the methyl 5-(4-((4-methylpiperazin-1-yl)methyl)benzamido)-2- methylbenzoate with the methyl 5-(3-(trifluorometny))-5-(4-metbyl-1H-imidazol-1- yl)benzamido)-2-methylbenzoate.

Step 3: preparation of methyl 5-(5-(3-(trifluoromethyl)-5-(4-methyl-1 H-imidazol- 1-yl)benzamido)-2-methylbenzylamino)-1H-pyrrolo[2,3-bJpyridine-2-carboxylate (ND0126)

Figure imgf000102_0003

The composed is obtained according to example 83 (step 3) replacing N-(3-formyl-4- methylphenyl)-4-((4-methylpiperazin~1-yl)methyl)-benzamide with the 3- (trifluoromethyl)-N-(3-formyl-4-methylphenyl)-5-(4-methyl-1 H-imidazol-1-yl)benzamide.

 

PATENT

WO 2014102376

str1

 

REFERENCES

WO2005063747A1 * Dec 23, 2004 Jul 14, 2005 Pfizer Italia S.R.L. PYRROLO[2,3-b] PYRIDINE DERIVATIVES ACTIVE AS KINASE INHIBITORS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITION COMPRISING THEM
WO2008028617A1 * Sep 4, 2007 Mar 13, 2008 F. Hoffmann-La Roche Ag Heteroaryl derivatives as protein kinase inhibitors
WO2008124849A2 * Apr 10, 2008 Oct 16, 2008 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
WO2008144253A1 * May 9, 2008 Nov 27, 2008 Irm Llc Protein kinase inhibitors and methods for using thereof
WO2014102376A1 * Dec 30, 2013 Jul 3, 2014 Oribase Pharma Protein kinase inhibitors
WO2014102377A1 * Dec 30, 2013 Jul 3, 2014 Oribase Pharma Azaindole derivatives as multi kinase inhibitors
WO2014102378A1 * Dec 30, 2013 Jul 3, 2014 Oribase Pharma Azaindole derivatives as inhibitors of protein kinases
US20150353540 * Dec 30, 2013 Dec 10, 2015 Oribase Pharma Azaindole derivatives as inhibitors of protein kinases
US2011312959 2011-12-22 Derivatives of Azaindoles as Inhibitors of Protein Kinases ABL and SRC

///////ND 0126, 1240322-54-6, PRECLINICAL

O=C(OC)c1cc2cc(cnc2n1)NCc3cc(ccc3C)NC(=O)c4cc(cc(c4)n5cc(C)nc5)C(F)(F)F

CC1=C(C=C(C=C1)NC(=O)C2=CC(=CC(=C2)N3C=C(N=C3)C)C(F)(F)F)CNC4=CN=C5C(=C4)C=C(N5)C(=O)OC

 

Share

PF-06260414

 Uncategorized  Comments Off on PF-06260414
Apr 192016
 

img

PF-06260414
CAS: 1612755-71-1
Chemical Formula: C14H14N4O2S
Exact Mass: 302.0837

PF-06260414; PF 06260414; PF06260414; PF6260414; PF-6260414; PF 6260414.

IUPAC/Chemical Name: (R)-6-(4-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)isoquinoline-1-carbonitrile

  • 6-[(4R)-4-Methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile

https://clinicaltrials.gov/ct2/show/NCT02070939

  • 28 Jul 2015Discontinued – Phase-I for Cachexia in USA (PO)
  • 27 Apr 2015Pfizer terminates a phase I trial (In volunteers) in USA (NCT02393807)
  • 26 Mar 2015Pfizer plans a phase I pharmacokinetic trial for Healthy volunteers in USA (NCT02393807)
Company Pfizer Inc.
Description Selective androgen receptor modulator
Molecular Target Androgen receptor
Mechanism of Action
Therapeutic Modality
Latest Stage of Development Phase I
Standard Indication Cachexia
Indication Details Treat cachexia

PF-06260414 is a selective androgen receptor modulator, or SARM, which is developed to treat muscle weakening. Testosterone’s anabolic properties help develop muscle mass, and its androgenic activity is associated with reproduction. Improving muscle mass would improve quality of life and may even prolong survival in certain patient populations.

PATENT

WO 2015173684

http://www.google.com/patents/WO2015173684A1?cl=en

The androgen receptor (“AR”) is a ligand-activated transcriptional regulatory protein that mediates induction of male sexual development and function through its activity with endogenous androgens. Androgenic steroids play an important role in many physiologic processes, including the development and maintenance of male sexual characteristics such as muscle and bone mass, prostate growth,

spermatogenesis, and the male hair pattern. The endogenous steroidal androgens include testosterone and dihydrotestosterone (“DHT”). Steroidal ligands which bind the AR and act as androgens (e.g. testosterone enanthate) or as antiandrogens (e.g.

cyproterone acetate) have been known for many years and are used clinically.

6-[(4f?)-4-Methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile (Formula I), in its free base form, has the chemical formula C14H14N4SO2 and the following structural formula:

Formula I

Synthesis of 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile is disclosed in co-pending international patent application,

PCT/IB2013/060381 , filed 25th November 2013, and published as WO 2014/087298 on 12th June 2014, assigned to the assignee of the present invention and which is incorporated herein by reference in its entirety. 6-[(4f?)-4-Methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile is known to be active as a selective androgen receptor modulator (SARM) and, as such, is useful for treating and/or preventing a variety of hormone-related conditions, for example, conditions associated with androgen decline, such as, inter alia, anaemia; anorexia; arthritis; bone disease; musculoskeletal impairment; cachexia; frailty; age-related functional decline in the elderly; growth hormone deficiency; hematopoietic disorders; hormone replacement; loss of muscle strength and/or function; muscular dystrophies; muscle loss following surgery; muscular atrophy; neurodegenerative disease; neuromuscular disease;

obesity; osteoporosis; and, muscle wasting.

Identification of new solid forms of a known pharmaceutical active ingredient provide a means of optimising either the physicochemical, stability, manufacturability and/or bioperformance characteristics of the active pharmaceutical ingredient without modifying its chemical structure. Based on a chemical structure, one cannot predict with any degree of certainty whether a compound will crystallise, under what conditions it will crystallise, or the solid state structure of any of those crystalline forms. The specific solid form chosen for drug development can have dramatic influence on the properties of the drug product. The selection of a suitable solid form is partially dictated by yield, rate and quantity of the crystalline structure. In addition, hygroscopicity, stability, solubility and the process profile of the solid form such as compressibility, powder flow and density are important considerations.

The general reaction schemes provided herein illustrate the preparation of 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile (Formula I).

Example 1

Procedure:

Into a 2L 3-neck round bottom flask equipped with a mechanical stirrer, reflux condenser and thermocouple with heating mantle was placed 2-methyltetrahydrofuran (2-MeTHF) (10 mL/g; 8.15 moles; 817 ml_; 702 g) followed by racemic-2,2′-bis(diphenylphosphino)-1 ,1 ‘-binaphthyl (BINAP) (0.04 equiv (molar); 14.0 mmol; 8.74 g) and bis(dibenzylideneacetone)palladium (Pd2(dba)3) (0.04 equiv (molar); 14.0 mmol;

8.07 g). The mixture was degassed by pulling vacuum and refilling with nitrogen three times then heated to 75 °C for 15 minutes and cooled to ambient temperature. In a separate flask, (S)-3-amino-2-methylpropan-1-ol (1.60 equiv; 561 mmol; 50.0 g, prepared using literature methods, for example as disclosed in EP-A-0,089, 139 published on 21st September 1983) was dissolved in 2-methyltetrahydrofuran (5 ml_/g;

4.08 moles; 409 ml_; 351 g) and degassed by pulling vacuum and refilling with nitrogen three times. Into the pot containing the catalyst was added 6-(bromoisoquinoline-1- carbonitrile) (1.00 equiv; 351 mmol; 81.75 g) and cesium carbonate (1.6 equiv (molar); 561 mmol; 185 g) in single portions followed by the solution of the aminoalcohol via addition funnel. The reaction mixture was again degassed by pulling vacuum and refilling with nitrogen three times. The reaction was heated to 70 °C for 3 hours. The reaction was cooled to ambient temperature and filtered through a pad of Celite. The contents of the flask were rinsed out with three 100 mL portions of 2-methyltetrahydrofuran. The filtrate was transferred into a 2L round bottom flask equipped with a thermocouple and mechanical stirrer under nitrogen. Silica Gel (Silicylate SiliaMet® Thiol) (0.4 g/g-pure-LR; 544 mmol; 32.7 g) was charged and the flask was stirred at 40 °C overnight. The following morning, the reaction was cooled to < 30 °C and filtered again through Celite. The pad was washed with 100ml_ of 2-methyltetrahydrofuran (or until no yellow color persisted in the filtrate). The filtrate was placed into a 3L round bottom flask equipped with a magnetic stir bar, distillation head (with condenser and receiving flask), and thermocouple. The mixture was heated to 60 °C and placed under vacuum (-450-500 mbar) to distil out 1.3 L total of 2-methyltetrahydrofuran. 500 mL of toluene was added to precipitate the desired product. The heating mantle was removed and the reaction was allowed to reach ambient temperature. The mixture was stirred for 1 hour at ambient temperature and then the solids were collected by vacuum filtration on a sintered glass funnel. The cake was dried overnight on the funnel under vacuum. The following morning, the solids were transferred into an amber bottle and weighed (71.9 g; 298 mmol). The product was used in the next step without further purification.

Example 2

Procedure:

In a 1 L reactor equipped with a temperature probe and overhead stirring was added the product of Example 1 (20.0 g; 1.00 equiv; 82.9 mmol) and 2-methyltetrahydrofuran (2-MeTHF) (30 mL/g-pure-LR; 5.98 moles; 600 mL; 515 g). The reaction mixture was

gently warmed to 40°C to achieve partial solubility. The reaction was cooled to 0°C. Once the reaction reached 0°C methanesulfonyl chloride (MsCI) (1.4 equiv (molar); 1 16 mmol; 8.98 mL; 13.3 g) was added in a single portion followed immediately by triethylamine (TEA) (1.4 equiv (molar); 116 mmol; 16.2 mL; 11.7 g) dropwise via syringe over a period of 15 minutes. The reaction mixture was further stirred for 30 min at 0°C and then warmed to 23°C for 60 minutes. The product (26.47 g; 1.00 equiv; 82.88 mmol; 26.47 g; 100% assumed yield) was then used without purification for the sulfonylation reaction.

Example 3

t-BuOH, 2-MeTHF

o 0 °C to 23 °C o

CI-S-N=C=0 CI-S-NHBoc

0 O

Procedure:

To a solution of t-butyl alcohol (t-BuOH) (1 equiv (molar); 116 mmol; 1 1.0 mL; 8.60 g) in 2-methyltetrahydrofuran (2-MeTHF) (1 M; 1.16 moles; 116 mL; 99.6 g) at 0°C was added chlorosulfonyl isocyanate (116 mmol; 1.00 equiv; 10.1 mL; 16.4 g) dropwise. The homogeneous solution was stirred for 30 minutes at ambient temperature and then used directly in the sulfonylation reaction.

Example 4

Sulfonylation Reaction Procedure:

A previously prepared solution of the product of Example 3 (1.4 equiv (molar); 1 16 mmol; 116 g) in 2-methyltetrahydrofuran was added to a suspension of the product of Example 2 (1.00 equiv; 82.89 mmol; 26.5 g) at 0°C. The mixture was warmed to ambient temperature over 30 minutes. HPLC analysis revealed the reaction was complete. The reaction was quenched with a 10% sodium carbonate solution (2 equiv

(molar); 165 mmol; 101 mL; 1 17 g) and water (to dissolve salts) (5 L/kg; 7.35 moles; 132 mL; 132 g). The top organic layer was removed and passed through a plug of Carbon (Darco G60) (0.5 g/g) on a filter. A significant improvement in color (dark orange to yellow) was observed. The solution was concentrated to 10 total volumes and used in the next step without purification.

Example 5

Procedure:

A solution of the product of Example 4 (1.OOequiv; 82.9 mmol; 41.3 g) in 2-methyltetrahydrofuran (2-MeTHF) (10ml_/g; 4.12 moles; 413 mL; 355 g) was placed into a 1 L reactor equipped with an overhead stirrer and temperature probe. Next, potassium carbonate (K2CO3) (325 mesh) (6 equiv (molar); 497 mmol; 69.4 g) and water (0.0 L/100-g-bulk-LR; 459 mmol; 8.26 mL; 8.26 g) were added and the mixture heated to 40°C (jacket temperature) and stirred overnight. The reaction was cooled to ambient temperature and water (4L/kg-pure-LR; 9.17 moles; 165 mL; 165 g]) was added. The biphasic reaction was stirred for 1 hour at 23 °C. The aqueous layer was extracted and removed. The organic layer was passed through a plug of Carbon (Darco G60) (0.5 g/g-pure-LR; 20.7g) in a disposable filter. The 2-methyltetrahydrofuran solution was switched to a 10 volume solution of toluene via a constant strip-and-replace distillation to no more than 1 % 2-methyltetrahydrofuran. The toluene solution of the reaction product (1.00 equiv; 82.9 mmol; 33.4 g; 100% assumed yield) was used as-is in the next step without further purification.

Example 6

Procedure:

To a 1 L reactor under nitrogen and equipped with overhead stirring and a temperature probe was added the product of Example 5 (1.00 equiv; 78.7 mmol; 33.4 g) as a solution in toluene (10 mL/g-pure-LR; 3.00 moles; 317 ml_; 276 g). Next, trifluoroacetic acid (TFA) (10 equiv (molar); 787 mmol; 59.5 ml_; 89.8 g) was added to the reaction over a period of 1 hour keeping the internal temperature below 30°C. The dark red mixture was stirred for 1 hour. The reaction was quenched at 23 °C by the addition of sodium carbonate (5 equiv (molar); 394 mmol; 240 ml_; 278 g). The reaction was quenched slowly, over a period of 1 hour to form the TFA salt of the product. Once the charge was complete, the mixture was cooled to 0°C, held for 1 hour and filtered. The next morning, the solid product (6-[(4R)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile in its free base form) was weighed (0.89 equiv; 70.0 mmol; 21.2 g; 89.0% yield) and used in the next step without further purification.

Example 7

Crystalline 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile free base (Form (1)) was prepared as follows.

In a 1 L 3-neck round bottom flask was added 6-[(4R)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile free base (1.00 equiv; 70.0 mmol; 21.2 g) a magnetic stir bar and acetone (40ml_/g; 1 1.5 moles; 847 ml_; 669 g). The mixture was heated to reflux (approximately 57°C) and stirred for 1 hour. The mixture was concentrated by atmospheric distillation (heating mantle set at 65°C) and 40ml_ of acetone was collected into a graduated cylinder. Next, water (25 mL/g; 29.4 moles; 530 ml_; 530 g) was charged over a period of one hour. The mixture was stirred at ambient temperature for 60min before being cooled to 0°C at 1 °C /min for 1 hour. The solids were collected by filtration in a disposable funnel. Crystalline 6-[(4f?)-4-methyl-1 , 1-dioxido-1 ,2,6-thiadiazinan-2-yl]isoquinoline-1-carbonitrile (Form (1), 0.88 equiv; 61.9 mmol; 18.7 g; 88.3% yield) was dried under vacuum overnight at 40 °C. Typical purity after crystallization is 98%.

PATENT

US 20140155390

Figure US20140155390A1-20140605-C00007

Figure US20140155390A1-20140605-C00008

Step 1. Synthesis of 6-bromoisoquinoline (#A1). A mixture of 4-bromobenzaldehyde (300.0 g, 1620.0 mmol) and amino acetaldehyde dimethyl acetal (170.4 g, 1620 mmol) in anhydrous toluene (1.5 L) was refluxed under a Dean-Stark condenser for 12 h. The solution was concentrated under vacuum. The residue was dissolved in anhydrous THF and cooled to —10° C. Ethyl chloroformate (193.3 mL, 1782 mmol) was added and stirred for 10 min at −10° C., and then allowed to warm to room temperature. Subsequently trimethyl phosphite (249.6 mL, 1782.0 mmol) was added dropwise to the reaction mixture and stirred for 10 h at room temperature. The solvent was evaporated under vacuum and the residue was dissolved in anhydrous DCM (1.5 L) and stirred for 30 minutes. The reaction mixture was cooled to 0° C., and titanium tetrachloride (1.2 L, 6480 mmol) was added dropwise. The reaction mixture was stirred at 40° C. for 6 days. The reaction mixture was poured into ice and pH was adjusted to 8-9 with aqueous 6N NaOH solution. The suspension was extracted three times with EtOAc. The organic layer was extracted with 3 M HCl. The acidic aqueous solution was adjusted to pH to 7-8 with 3N NaOH solutions and extracted two times with EtOAc. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide the product. Crude compound was dissolved in minimum amount of DCM and mixed with pentane to get compound #A1 as light brown solid. Yield: 90 g (35%). Rf: 0.6 (30% EtOAc in petroleum ether).

 

LCMS m/z=209 (M+1). 1H NMR (400 MHz, d6-DMSO): δ 7.82 (m, 2H), 8.11 (d, J=8.8 Hz, 2H), 8.30 (br s, 1H), 8.56 (d, J=6.0 Hz, 1H), 9.35 (s, 1H).

Step 2. Synthesis of 6-bromoisoquinoline 2-oxide (#A2). m-Chloroperoxybenzoic acid (120.0 g, 720.0 mmol) was added to a solution of #A1 (90.0 g, 480.0 mmol) in DCM (500 mL) at room temperature, and the reaction mixture was stirred for 16 h. 1N NaOH was added to the stirred reaction mixture to adjust the pH to 7-8. The layers were separated and the aqueous layer was extracted with DCM. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to render crude product. The solid product was triturated with the mixture of n-pentane and ethanol (8:2) to get the #A2 as white solid. Yield: 65 g (60%). Rf: 0.2 (EtOAc).

LCMS m/z=225 (M+1). 1H NMR (400 MHz, d6-DMSO): δ 7.83 (m, 2H), 7.91 (d, J=6.8 Hz, 1H), 8.21 (dd, J=8.0, 1.2 Hz, 1H), 8.26 (br s, 1H), 8.97 (s, 1H).

 

Step 3. Synthesis of 6-bromoisoquinoline-1-carbonitrile (#A3). Trimethylsilyl cyanide (52.0 mL, 580.0 mmol) was added dropwise to the stirred solution of #A2 (65.0 g, 290.0 mmol) and DBU (50.0 mL, 348.0 mmol) in THF (500 mL) at room temperature over a period of 15 minutes. The reaction mixture was stirred at room temperature for 1 h. Water was added to the reaction mixture, and the solution was extracted with DCM. The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to give crude product. The product was purified by column chromatography using silica gel (100-200 mesh) with 0-4% EtOAc in petroleum ether as an eluent to give #A3 as white solid. Yield: 41 g (61%). Rf: 0.6 (30% EtOAc in petroleum ether).

LCMS m/z=233 (M+1). 1H NMR (400 MHz, d6-DMSO): δ 8.07 (dd, J=11.2, 2.0 Hz, 1H), 8.21 (m, 2H), 8.55 (br s, 1H), 8.77 (d, J=7.6 Hz, 1H).

A General Procedure to Prepare Intermediates of #A4, #A5, #A6 and #1, #2, #3, #4, #6, #7.

Step 4. A solution of #A3 (1 eq.) in toluene (50 mL) was degassed by bubbling with argon gas for 15 min and then Pd2dba3 (0.03 eq.), BINAP (0.06 eq.) and Cs2CO3(3 eq.) were added to the solution followed by the addition aminoalcohol (2 eq.). The mixture was heated at 100° C. under argon atmosphere for 3 h. Reaction mixture was cooled to room temperature, diluted with EtOAC and washed with water and brine. The organic layer was dried over Na2SO4 and concentrated to get crude product. The crude compounds were purified by silica gel (100-200 mesh) column chromatography by using 0-5% MeOH in DCM. Yields: 25-45%.

Step 5. MsCl (1 eq.) was added dropwise to a solution of #A4 (1 eq.) and Et3N (2 eq.) in DCM (10 mL) at 0° C. and was stirred at room temperature for 3 h. The reaction mixture was diluted with DCM, washed with water and brine. The organic layer was dried over Na2SO4 and concentrated. Crude products were used in next step without further purification.

Step 6. t-Butanol (2 eq.) was slowly added to a solution of chloro sulfonyl isocyanate (2 eq.) in toluene (1 mL/1 mmol) at 0° C. The reaction mixture was stirred at room temperature for 45 min. This solution (t-butyl chlorosulfonylcarbamate) was then added to a solution of #A5 (1 eq.) and DIPEA (4 eq.) in THF and stirred at room temperature for 12 h. Reaction mixture was diluted with water and extracted with EtOAc. Organic layer was washed with water, brine, then dried over anhydrous Na2SO4 and concentrated. Crude products were purified by silica gel (100-200 mesh) column chromatography using 0-40% EtOAc in petroleum ether.

Step 7. TFA was added to a solution of #A6 (1 eq.) in DCM (8 mL) at 0° C. and stirred at room temperature for 2 h. Reaction mixture was concentrated, diluted with water, neutralized with sat. aq. NaHCO3 soln. then extracted with DCM. The organic layer was washed with water and dried over Na2SO4 then concentrated. The crude products were purified by triturating with DCM and pentane to provide the compound. In the case of racemic materials, the enantiomers were separated by chiral preparative HPLC.

Column: CHIRALPAK IA, 4.6 mm×250, 5 μm; Mobile phase: n-Hexane: EtOH (65:35) (For X3: 35:65; For X2: 70:30); Flow rate: 1 mL/min; Eluent: EtOH.

EXAMPLE 16-[(3S)-3-methyl-1,1-dioxido-1,2,5-thiadiazolidin-2-yl]isocluinoline-1-carbonitrile (#1; R═CH3)

LCMS m/z=289.1 (M+1). 1H NMR (400 MHz, d6-DMSO): δ 1.37 (d, J=6.3 Hz, 3H), 3.27 (m, 1H), 3.74 (m, 1H), 4.63 (m, 1H), 7.17 (d, J=5.7 Hz, 1H), 7.72 (m, 1H), 7.89 (dd, J=10.7, 2.1 Hz, 1H), 8.26 (m, 2H), 8.62 (d, J=5.7 Hz, 1H).

PATENT

example 9

6 – [(3S) -3-methyl-1, 1 -dioxido-1, 2,5-thiadiazolidin-2-carbonitrile 1-yl1naphthalene

(Stereochemistry is arbitrarily Assigned)

LCMS m / z = 286.0 (M – H). 1 H NMR (400 MHz, cf 6 -DMSO): δ 1 .31 (d, J = 6.2 Hz, 3H), 3.13 – 3.25 (m, 1H), 3.71 (dt, J = 12.5, 6.8 Hz, 1H), 4.49 – 4.62 (m, 1H), 7.62 – 7.70 (m, 1H), 7.75 – 7.83 (m, 2H), 7.99 (t, J = 7.8 Hz, 1H), 8.07 (d, J = 6.6 Hz, 1H), 8.14 (d, J = 8.9 Hz, 1H), 8.28 (d, J = 8.4 Hz, 1H). Chiral HPLC purity: 99.1% (retention time 17.12 minutes)

Step 1. Synthesis of amino ester (# D1). Thionylchlride (8.5 mL, 1 16.5 mmol) Was added to the solution of amino acid (4.0 g, 38.8 mmol) in MeOH (170 mL) at 0 ° C, and the reaction mixture Was Stirred for 6 h at room temperature. The reaction Was monitored by TLC, and after-disappearance of the starting material It was cooled to room temperature and solid NaHC0 3 Was added. The reaction mixture Was filtered, concentrated in vacuo and the resulting and residue Was triturated with diethyl ether to crude obtenir # D1 (4 g, 90%) as a white solid. R f : 0.4 (f-BuOH: AcOH: H 2 0 (4: 0.5: 0.5)).

GCMS m / z 1 17.1 (M +). 1 H NMR (400 MHz, cf 6 -DMSO): δ 1.17 (d, J = 6.8Hz, 3H), 2.83 – 2.88 (m, 2H), 3.03 – 3.05 ( m, 1H), 3.65 (s, 3H), 8.02 – 8.30 (br s, 3H).

Step 2. Synthesis of aminoalcohol (# D2). # D1 (2.0 g, 13.0 mmol) Was added

portionwise to a suspension of LiAlH 4 (1.4 g, 39.2 mmol) in THF (75 mL) under nitrogen atmosphere at 0 ° C. The reaction mixture Was Stirred for 30 minutes and allowed to stir Then at room temperature for Reviews another 30 minutes. The reaction mixture Was Refluxed for 2 h, And Then It was cooled to -10 ° C and quenched with ice cold water Carefully (1.4 mL). 10% NaOH solution (2.8 mL) and ice cold water (4.2 mL) Were added, and the mixture Was Stirred for 15 minutes. It was filtered, and the filtrate washed with EtOAc (3 x 100 mL), dried over anhydrous Na 2 S0 4 and Concentrated under vacuum to obtenir # D2 (1.2 g, 86%) as a pale yellow liquid. R f: 0.2 (20% MeOH in DCM).

1 H NMR (400 MHz, cf 6 -DMSO): δ 0.78 (d, J = 6.8Hz, 3H), 1.46 – 1.54 (m, 1H), 2.41 -2.45 (m, 2H), 2.50 – 2.54 (m , 1H), 3.22 – 3.34 (m, 4H).

Step 3. Synthesis of coupling product (# D3). K 3 P0 4 (6.1 g, 28.8 mmol), BINAP (0.44 g, 0.72 mmol) and Pd 2 (dba) 3 (0.32.0 g, 0.36 mmol) Was added to the degassed

suspension of 6-bromo-1 -cyanoisoquinoline # A3 (1.7 g, 7.2 mmol), # D2 (1.2 g, 14.5 mmol) in DMSO at room temperature. The reaction mixture Was heated at 105 ° C for 2 h. The reaction Was cooled to room temperature, water (500 mL) Followed by EtOAc (100 mL) Were added, and the mixture Was Stirred for 10 minutes. The biphasic mixture Was filtered through a Celite ™ pad and washed with EtOAc (100 mL). The organic layer Was separated, and the aqueous layer Was Extracted with EtOAc (3 x 100 mL). The combined organic layers Were dried over anhydrous Na 2 S0 4 , concentrated under Reduced pressure to get a crude material. Reviews This was purified by column chromatography on 100-200 mesh silica gel, using 50-70% EtOAc in petroleum ether as the eluent to obtenir # D3 (0.5 g, 48.5%) as a yellow solid. R f : 0.4 (60% EtOAc in petroleum ether).

LCMS m / z = 242.0 (M + H). 1 H NMR (400 MHz, cf 6 -DMSO): δ 0.97 (d, J = 6.4Hz, 3H), 1.87 – 1.99 (m, 1H), 2.92 – 2.99 (m, 1H), 3.20 – 3.27 (m, 1H), 3.38 – 3.42 (m, 2H), 4.59 (t, J = 5.2 Hz, 1H), 6.77 (d, J = 2.0, 1H ), 7.01 (t, J = 5.6 Hz, 1H), 7.34 (dd, J = 9.2 Hz, J = 2.0 Hz, 1H), 7.73 (d, J = 6.0 Hz, 1H), 7.88 (d, J = 8.8 Hz, 1H), 8.312 (d, J = 6.0 Hz, 1H).

Step 4. Methanesulfonated coupling product (# D4). Triethylamine (0.44 mL, 3.1 mmol) Was added to a solution of # D3 (0.50 g, 2.0 mmol) in DCM at 0 ° C.

Methanesulfonylchloride (0.25 mL, 3.1 mmol) Was added over 10 minutes, and the reaction mixture Was Stirred for 1 h at room temperature. After disappearance of the starting material by TLC, It was diluted with DCM and washed with water. The organic layer Was separated, dried over Na 2 S0 4 , concentrated under pressure to obtenir Reduced crude # D4 (0.6 g, crude) as yellow solid. Reviews This was used for next step Without Any purification. R f : 0.6 (50% EtOAc in petroleum ether).

LCMS m / z = 320.0 (M + H). 1 H NMR (400 MHz, CDCl 3 ): δ 1.17 (d, J = 6.8Hz, 3H), 2.32 – 2.37 (m, 1H), 3.06 (s, 3H), 3.26 – 3.41 (m, 2H), 4.16 – 4.20 (m, 1H), 4.33 – 4.37 (m, 1H), 4.75 (br s, 1H), 6.70 (d, J = 2.4, 1 H), 7.09 (dd, J = 9.2 Hz, 2.4 Hz, 1H), 7.57 (d, J = 6.0 Hz, 1H), 8.05 (d, J = 9.2 Hz, 1H), 8.39 (d, J = 5.6 Hz, 1H).

Step 5. cyclized and uncyclized intermediates (# D5, D6 #). Chlorosulfonyl isocyanate (1.2 mL, 13.1 mmol) Was added dropwise to a solution of f-BuOH (1.4 mL, 13.1 mmol) in toluene (4.0 mL) at -5 ° C. The reaction mixture Was Stirred at room temperature for 20 minutes, And Then THF (1 mL) Was added to the resulting suspension to obtenir clear solution. In Reviews another flask, DIPEA (2.3 mL, 13.1 mmol) Was added to a solution of # D4 (0.6 g, 2.6 mmol crude) in dry THF (3 mL). The Above Prepared reagent (CIS0 2 NH-Soc) Was added to this reaction mixture dropwise at room temperature over a period of 20 minutes. The resulting and reaction mixture Was Then Stirred for 16 h at room temperature. The mixture Was diluted with EtOAc (100 mL) and washed with water (100 mL). The aqueous layer Was washed with EtOAc (2 x 100 mL), combined all the organic layers, dried over Na 2 S0 4 , concentrated under Reduced pressure to obtenir the crude product (LCMS shows Desired # D6 and uncyclized # D5. This crude Was purified by column chromatography on 100-200 mesh silica gel, using 10-30% EtOAc in petroleum ether as an eluent to obtenir Desired # D6 (0.35 g, 47.8%), and uncyclized # D5 (0.22 g, crude).

The uncyclized # D5 (0.22 g, crude) Was Dissolved in THF (1 mL) and DIPEA (0.6 ml) Was added to the solution. The reaction mixture Was Stirred Reviews another for 12 h at room temperature. After qui time, It was diluted with EtOAc (100 mL) and washed with water (100 mL). The aqueous layer Was washed with EtOAc (2 x 100 mL), combined all the organic layers, dried over Na 2 S0 4 , concentrated under pressure to obtenir Reduced crude product. Was this crude purified by column chromatography on 100-200 mesh silica gel, using 10-30% EtOAc in petroleum ether as an eluent to obtenir Desired # D6 (1 .1 g, 13.2%). Total amount of # D6 Was (0.5 g, 60% for two steps, 82% purity LCMS). R f : 0.8 (60% EtOAc in petroleum ether).

LCMS m / z = 403.1 (M + H). 1 H NMR (400 MHz, CDCl3): δ 1 .04 (d, J = 6.8 Hz, 3H), 1 .50 (s, 9H), 2.38 – 2.48 ( m, 1H), 3.65 – 3.82 (m, 2H), 3.92 – 4.02 (m, 1H), 4.30 – 4.38 (m, 1H), 7.79 – 7.81 (m, 1H), 7.86 – 7.88 (m , 2H), 8.34 – 8.37 (d, J = 9.2 Hz, 1H), 8.67 (d, J = 6.0 Hz, 1H).

Step 6. Racemate # D7 and final products (# 10, # 11). TFA (5 mL) Was added to a solution of # D6 (0.15 g, 0.37 mmol) in DCM (100 mL) at 0 ° C. The reaction mixture Was Stirred for 1 h at 0 ° C. The solution Was Neutralized with saturated aqueous NaHC03 solution at 0 ° C. The mixture Was diluted with water, Extracted with DCM (3 x 100 mL). The combined organic layers Were dried over anhydrous Na 2 S0 4 and Concentrated under pressure Reduced to obtenir racemic # D7 (0.10 mg, 73%).

LCMS m / z = 303.0 (M + H). R f : 0.3 (60% EtOAc in petroleum ether).

Enantiomeric separation: # D7 Was Submitted for chiral separation to obtenir final compounds # 10 (0.015 mg) and # 11 (0.016 mg).

Column: CHIRALPAK IA, 4.6 χ 250 mm, 5 m; Mobile phase: n-Hexane / / -PrOH / DCM (60% / 15% / 15%); Flow rate: 0.8 mL / min.

example 10

6 – [(4R) -4-methyl-1, 1 -dioxido-1, 2,6-thiadiazinan-2-yl1isoquinoline-1-carbonitrile (# 10; R = (R) -CH 3 )

LCMS m / z = 303.0 (M + 1). 1 H NMR (400 MHz, cf 6 -DMSO): δ 0.98 (d, J = 6.4Hz, 3H), 2.22 – 2.26 (m, 1H), 3.16 – 3.22 (m, 1H), 3.34 – 3.39 (m, 1H), 3.59 – 3.65 (m, 1H), 3.77 – 3.81 (m, 1H), 7.75 – 7.79 (m, 1H, Disappeared in D20 exchange), 7.95 (dd, J = 8.8 Hz, J = 2.0 Hz, 1H), 8.06 (d, J = 1 .6 Hz, 1H), 8.23 – 8.27 (m, 2H), 8703 (d, J = 5.2 Hz, 1H). R f : 0.3 (60% EtOAc in petroleum ether). Chiral HPLC purity: 98.2% (retention time on January 1 .43 minutes).

CLIP

PF-06260414, A Treatment For Muscle Diseases

 

 

Print
PF-06260414
Company: Pfizer
Target: Androgen receptors
Disease: Muscular dystrophy, atrophy, sarcopenia
09338-scitech1-CheklerPf
Chekler

There aren’t many options when it comes to treating weakening muscles caused either by a disease such as muscular dystrophy or atrophy or by sarcopenia, the natural muscle weakening that comes with age. Doctors’ primary option is to give patients testosterone—a hormone with serious unwanted side effects on reproductive organs, the liver, and kidneys.

 

09338-scitech1-MorrisPf
Morris
Credit: Pfizer
09338-scitech1-OwensPf
Owens

Pfizer’s Eugene Chekler spoke about PF-06260414, a selective androgen receptor modulator, or SARM, the company developed to treat muscle weakening. The idea, Chekler told C&EN, was to develop a nonsteroidal small molecule that would target androgen receptors but wouldn’t have any of testosterone’s negative side effects.

09338-scitech1-GilbertPf
Gilbert

Testosterone’s anabolic properties help develop muscle mass, and its androgenic activity is associated with reproduction. To discover their SARM, Pfizer’s scientists used a novel screening strategy in which they decoupled anabolic and androgenic properties in vitro, Chekler said. Compounds that performed well in the muscle assay but had little effect in an assay that predicts androgenic response were developed further.

PF-06260414’s key pharmacophore is an isoquinoline with a pendant cyano group. The molecule also features a cyclic sulfuric diamide. It has completed Phase I clinical trials. “The market potential for this kind of treatment is huge,” Chekler said. “Improving muscle mass would improve quality of life and may even prolong survival in certain patient populations.”

Many answers from a first in human (FIH) study: Safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of PF-06260414 in healthy Western and Japanese males
Annu Meet Am Soc Clin Pharmacol Ther (ASCPT) (March 8-12, San Diego) 2016, Abst PI-021

/////////////////////PF-06260414

N#CC1=NC=CC2=C1C=CC(N(C[C@H](C)CN3)S3(=O)=O)=C2

Share
Follow

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

Join other followers: