DR ANTHONY MELVIN CRASTO,WorldDrugTracker, helping millions, A 90 % paralysed man in action for you, I am suffering from transverse mylitis and bound to a wheel chair, With death on the horizon, this will not stop me, Only God and death can..........
DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai), INDIA, worlddrugtracker, 29Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.8 Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution
n, सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये।औकात बस इतनी देना,कि औरों का भला हो जाये।...........P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Tuesday 27 October 2015

LINAGLIPTIN


linagliptin
C25H28N8O2
CAS : 668270-12-0
Molecular Weight: 472.54
Purity: > 98%
(R)-8-(3-aminopiperidin-1-yl)-7-(but-2-ynyl)-3-methyl-1-((4-methylquinazolin-2-yl)methyl)-1H-purine-2,6(3H,7H)-dione
8-(3R)-3-aminopiperidinyl)-7-butyn-2-yl-3-methyl-1-(4-methylquinazolin-2-ylmethyl)-3,7-dihydropurine-2,6-dione
Solubility: Up to 25 mM in DMSO
Synonyms: BI-1356, BI1356, Linagliptin, Tradjenta, Trajenta
BI-1356 (Linagliptin) is a highly potent and selective dipeptidyl peptidase 4 (DPP-4) inhibitor (IC50 = 1 nM) for treatment of type II diabetes. [1] BI-1356 can increase incretin levels (GLP-1 and GIP), which increases insulin secretion and inhibits glucagon release, decreases gastric emptying, and decreases blood glucose levels. BI-1356 shows 10,000-fold more selectivity for DPP-4 against other protease/peptidases, including DPP-8, DPP-9, trypsin, plasmin, and thrombin, It is a DPP-4 inhibitor developed by Boehringer Ingelheim for the treatment of type II diabetes.
Linagliptin is a highly potent, selective DPP-4 inhibitor with IC50 of 1 nM.
“This study provides much-needed data on glucose-lowering treatment of elderly people with Type 2 Diabetes, inadequately controlled with common anti-hyperglycaemic agents”
Data published in The Lancet showed that elderly people with Type 2 Diabetes (T2D) treated for 24 weeks with the dipeptidyl peptidase-4 (DPP-4) inhibitor linagliptin, marketed by Boehringer Ingelheim and Eli Lilly and Company, experienced significant reductions in blood glucose levels (HbA1c) compared with those receiving placebo. In addition, the overall safety and tolerability profile of linagliptin was similar to placebo, with no significant difference in hypoglycaemia
http://www.news-medical.net/news/20130817/Study-Type-2-diabetic-patients-treated-with-DPP-4-linagliptin-experience-reductions-in-blood-glucose-levels.aspx

INTRODUCTION

Linagliptin (BI-1356, trade names Tradjenta and Trajenta) is a DPP-4 inhibitor developed by Boehringer Ingelheim for treatment of type II diabetes.
Linagliptin (once-daily) was approved by the US FDA on 2 May 2011 for treatment of type II diabetes.[1] It is being marketed by Boehringer Ingelheim and Lilly.
  • Linagliptin, namely 8-(3R)-3-aminopiperidinyl)-7-butyn-2-yl-3-methyl-1-(4-methylquinazolin-2-ylmethyl)-3,7-dihydropurine-2,6-dione, of formula (A), is a long acting inhibitor of dipeptidylpeptidase-IV (DPP-IV) activity, at present under development for the treatment of type II diabetes mellitus.
    Figure imgb0001
  • The synthesis of Linagliptin is reported in US 7,407,955 , according to the scheme below, where 8-bromo xanthine of formula (B) is condensed with 3-(R)-Boc-aminopiperidine of formula (C) to obtain a compound of formula (D), which is converted to Linagliptin (A) by deprotection of the amine function
    Figure imgb0002
  • Optically active 3-aminopiperidine protected as the tert-butylcarbamate (Boc), compound (C), although commercially available, is very expensive and difficult to prepare; moreover in this process impurities are very difficult to remove, particularly on an industrial scale, in particular because of the Boc protective group. For this reason,US 2009/0192314 discloses a novel process for the preparation of Linagliptin (A) which makes use of a 3-(R)-aminopiperidine protected as a phthalimide of formula (E).
    Figure imgb0003
  • Accordingly, a compound of formula (E) can be prepared starting from 3-aminopyridine by hydrogenation, reaction with phthalic anhydride, resolution through diastereoisomeric salts using expensive D-tartaric acid, and then cleavage of the tartrate salt.
  • This intermediate is, however, still expensive and its use in the substitution reaction of the bromine derivative of formula (B) is still poorly efficient, as it takes place under drastic reaction conditions.
  • As it can be noted, these processes make use of drastic reaction conditions, or expensive, difficult to prepare starting materials, thus negatively affecting costs. There is therefore the need for an alternative synthetic route to provide Linagliptin or a salt thereof with high enantiomeric and chemical purity, from low cost starting materials.
US '955 is schematically represented in scheme
Figure imgf000002_0002
U.S. Patent No. 7,820,815 ("US '815) discloses a process for preparation of Linagliptin wherein it is prepared by deprotecting 1 -[(4-methyl-quinazolin-2-yl)methyl]-3- methyl-7-(2-butyn-1 -yl)-8-(3-(R)-phthalimidopiperidin-1 -yl)-xanthine of formula Ilia in presence of ethanolamine. The 1 -[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2- butyn-1 -yl)-8-(3-(R)phthalimidopiperidin-1 -yl)-xanthine is prepared by condensing 1 -[(4- l methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromo xanthine of formula III with (R)-3-phthalimidopiperidine of formula I la. The process disclosed in US '815 is schematically represented in scheme-ll.
Figure imgf000003_0001
Scherre
PCT Publications WO 2004/018468 and WO 2006/048427 describe synthesis of Linagliptin. Crystalline forms of Linagliptin, Forms A, B, C, D, and E are described in the PCT Publication No. WO 2007/128721. According to WO 2007/128721, Linagliptin prepared according to Publication No.
WO 2004/018468 is present in ambient temperature as a mixture of two enantiotropic polymorphs. The temperature at which the two polymorphs transform into one another is 25±15° C. The pure high temperature form (polymorph A), can be obtained by heating the mixture to temperatures>40° C. The low temperature form (polymorph B) is obtained by cooling to temperatures<10° C.”.
According to WO 2007/128721, the transition point between forms A and B is at room temperature, such that they exist as a polymorphic mixture. In addition, WO 2007/128721 teaches that form D “is obtained if polymorph C is heated to a temperature of 30-100° C. or dried at this temperature”. Since the procedure to obtain form C according to this application includes drying at 70° C., the dried form C is expected to be obtained in admixture with form D.
WO 2007/128721 teaches that Form E is obtained only at high temperatures (after melting of form D at 150±3° C.), and therefore is not relevant industrially.

 PATENT

Figure imgb0010
Figure imgb0008Figure imgb0009
Figure imgb0007
Figure imgb0006
Figure imgb0005
Figure imgb0004
Example 1: Preparation of a compound of formula (II) with X=OEt
    • The bromoxanthine of formula (B) prepared according to US 7,407, 955 (28.2 g, NMR title 90%, 56.0 mmols) and L-(+)-tartrate salt of (R)-ethylnipecotate (22.4 g, 72.8 mmols) are suspended in 50 mL of 1-methyl-2-pyrrolidone. The suspension is heated at 100° under stirring and, maintaining such temperature, diisopropylethylamine (38.3 ml, 224 mmols) is slowly dropwise added. The suspension is moderately refluxed for 2 hours. The mixture is cooled to 30°C and 400 mL of are dropwise added under vigorous stirring. The suspension is stirred for 30 minutes, then filtered off and the solid is washed with 100 mL of water. 27 g of solid product are obtained after drying with a 90% yield.
    • 1H-NMR (300 MHz, CDCl3), δ 8.02 (d, 1H), 7.87 (d, 1H), 7.76 (t, 1H), 7.51 (t, 1H), 5.55 (s, 2H), 4.90 (s, 2H), 4.25 - 4.10 (m, 2H), 3.82 (dd, 1H), 3.65 - 3.51 (m, 4H), 3.33 (dd, 1H), 3.15 (m, 1H), 2.88 - 2.72 (m, 4H), 2.08 (m, 1H), 1.92 - 1.73 (m, 6H), 1.27 (t, 3H).
Example 2: Preparation of a compound of formula (II) with X=OH
    • The compound of formula (II) having X = OEt, prepared according to Example 1 (27 g, 51 mmols), is suspended in 270 mL of MeOH and 4.1 g of NaOH scales and 13.7 mL of water are added under stirring. The reaction mixture is maintained under stirring for 2 hours at reflux temperature and then cooled to 40°C and diluted with 400 ml of water.
    • [0080]
      The mixture is then acidified by adding 6.6 mL of acetic acid and the solid is filtered off and washed with water and dried under vacuum at 50°C, obtaining 21 g of product, with a yield of 82%.
    • 1H-NMR (300 MHz, DMSO-d6), δ 8.11 (d, 1H), 7.85 (t, 1H), 7.80 (d, 1H), 7.62 (t, 1H), 5.30 (s, 2H), 4.87 (s, 2H), 3.79 (dd, 1H), 3.57 (m, 1H), 3.38 (s, 3H), 3.33 (dd, 1H), 3.10 (m, 1H), 2.85 (s, 3H), 2.62 (m, 1H), 1.95 (m, 1H), 1.78 - 1.60 (m, 6H).
Example 3: Preparation of a compound of formula (IV) with R = OCH(CH3)2
    • The compound of formula (II) with X=OH prepared according to Example 2 (0.5 g; 1 mmol), 5 ml of isopropanol and trietylamine (0.17 ml, 1.2 mmols) are mixed under stirring. 0.3 g of diphenylphosphorylazide (DPPA) are added in a sole portion. The mixture is heated at reflux temperature for 2 hours under stirring. The mixture is then cooled to room temperature and the solid is filtered off and washed with 2 ml of isopropyl alcohol. The solid is dried under vacuum at 50°C obtaining 0.4 g of product with a yield of 72%.
    • 1H-NMR (300 MHz, DMSO-d6), δ 8.12 (d, 1H), 7.85 (t, 1H), 7.80 (d, 1H), 7.63 (t, 1H), 5.28 (s, 2H), 4.85 (s, 2H), 4.75 (ep, 1H), 4.27 (d, 1H), 3.78-3.55 (m, 4H), 3.35 (s, 3H), 2.85 (s, 3H), 1.85 - 1.60 (m, 6H). 1.42 (m, 1H), 1.02 (d, 6H).
Example 4: Preparation of Linagliptin
    • The carbamate of formula (IV), prepared according to Example 3 (400 mg, 0.72 mmols), is dissolved in 5 ml of 32% HCl in water. The reaction mixture is maintained under stirring at 65-70°C for 7 hours and then cooled to room temperature. The pH of the solution is brought to about 8-9 by treatment with 30% NaOH in water and the obtained suspension is stirred for 10 minutes and then filtered off. The solid is dissolved in 10 ml of AcOEt, the solution is filtered and the filtrate is evaporated under reduced pressure. 250 mg of Linagliptin are obtained with a yield of 73%.
Example 5: Preparation of a compound of formula (IV) with R = S(CH2)11CH3
    • The compound of formula (II) with X =OH, prepared according to Example 2 (3.0 g, 6 mmols), 30 ml of acetonitrile and triethylamine (1.09 ml, 7.8 mmols) are mixed together. Subsequently, 1.55 ml (7.2 mmols) of diphenylphosphorylazide (DPPA) are added. The reaction mixture is heated at reflux temperature for 1 hour under stirring and then cooled to 60°C and treated with dodecanethiol (1.87 ml, 7.8 mmols). The mixture is maintained under stirring at the same temperature for 30 minutes and then cooled to 25°C. The formed solid is filtered off and washed with 10 ml of acetonitrile. The solid is dried under vacuum at 60°C obtaining 3.5 g of product with a yield of 85%.
    • 1H-NMR (300 MHz, DMSO-d6), δ 8.21 (d, 1H), 7.88 (t, 1H), 7.83 (d, 1H), 7.64 (t, 1H), 5.30 (s, 2H), 4.86 (s, 2H), 3.85 (m, 1H), 3.70 (d, 1H), 3.56 (d, 1H), 3.38 (s, 3H), 3.10-2.87 (m, 3H), 2.85 (s, 3H), 2.74 (t, 2H), 1.90-1.60 (m, 3H), 1.74 (s, 3H), 1.60-1.40 (m, 2H), 1.38-1.10 (m, 18H), 0.82 (t, 3H).
Example 6: Preparation of Linagliptin
    • The thiocarbamate of formula (IV) (10 g, 14,3 mmols), prepared according to Example 5, is dissolved in 100 mL of N-methylpyrrolidone (NMP) and treated with a 30% NaOH solution (7.6 g, 57.0 mmols). The reaction mixture is stirred for 3 hours and then diluted with water and acidified by adding concentrated H2SO4. The mixture is extracted with hexane and brought to pH 9.5 by adding 30% NaOH and repeatedly extracted with dichloromethane. The dichloromethane phases are collected and washed with water and then dried over Na2SO4, filtered and concentrated under reduced pressure. The so obtained oily residue is then dissolved in methyl tert-butyl ether (MTBE) and the mixture is maintained under stirring for 2 hours, then cooled to 0-5°C and the so obtained solid is filtered off, washed with MTBE and dried under vacuum at 50°C till constant weight. 4.2 g of Linagliptin with a yield of 63% are obtained.
Example 7: Preparation of a compound of formula (IV) with R=C7H5N2S (2-mercaptobenzoimidazole)
    • The compound of formula (II) with X =OH, prepared according to Example 2 (2.0 g, 4 mmols), 20 ml of acetonitrile and triethylamine (0.8 ml, 5.6 mmols) are mixed together. Subsequently, 1.43 g (5.2 mmols) of diphenylphosphorylazide (DPPA) are added. The reaction mixture is heated at reflux temperature for 1 hour under stirring and then cooled to 60°C and treated with 2-marcaptobenzimidazole (0.8 g, 5.2 mmols). The mixture is maintained under stirring at the same temperature for 30 minutes, then cooled to 25°C and evaporated under reduced pressure with Rotavapor®. The residue is treated with 50 ml of dichloromethane (CH2Cl2) and washed with 2X20 ml of 5% NaOH. The organic phase is dried over Na2SO4, filtered and concentrated under reduced pressure and the residue is triturated with 30 ml of MTBE. The so obtained solid is filtered off, dried under vacuum at 60°C till constant weight obtaining 2.5 g of light brown powder.
Example 8: Preparation of Linagliptin
  • Starting from the compound of formula (IV) as obtained in example 7 and following the procedure of example 6, product Linagliptin is obtained.

PAPER

Org. Biomol. Chem., 2015,13, 7624-7627
DOI: 10.1039/C5OB01111F
http://pubs.rsc.org/en/content/articlelanding/2015/ob/c5ob01111f#!divAbstract
By employing a rhodium–Duanphos complex as the catalyst, β-alkyl (Z)-N-acetyldehydroamino esters were smoothly hydrogenated in a highly efficient and enantioselective way. Excellent enantioselectivities together with excellent yields were achieved for a series of substrates. An efficient approach for the synthesis of the intermediate of the orally administered anti-diabetic drugs Alogliptin and Linagliptin in the DPP-4 inhibitor class was also developed.

Graphical abstract: Highly enantioselective synthesis of non-natural aliphatic α-amino acids via asymmetric hydrogenation


Mechanism of action

Linagliptin is an inhibitor of DPP-4, an enzyme that degrades the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). Both GLP-1 and GIP increase insulin biosynthesis and secretion from pancreatic beta cells in the presence of normal and elevated blood glucose levels. GLP-1 also reduces glucagon secretion from pancreatic alpha cells, resulting in a reduction in hepatic glucose output. Thus, linagliptin stimulates the release of insulin in a glucose-dependent manner and decreases the levels of glucagon in the circulation.


PAPER

http://www.gosalute.it/linagliptin-nuovi-dati-presentati-allada-sugli-eventi-cardiovascolari-e-sulla-sicurezza-ed-efficacia-nei-pazienti-affetti-da-diabete-di-tipo-2-con-insufficienza-renale-da-moderata-a-grave/

PATENT

http://www.google.com/patents/WO2013098775A1?cl=en
In one aspect, the application provides a process for preparation of Linagliptin comprising reacting (R)-piperidine-3-amine of formula II or an acid addition salt thereof with 1 -[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine of formula III in the presence of a suitable base in an inert organic solvent.
Figure imgf000004_0001
In another aspect, the application provides Linagliptin or a pharmaceutically acceptable salt thereof, having less than about 0.15 area % of potential process related impurities viz., regio-impurity of the formula la, bromo-impurity of the formula lb and S- isomer as measured by HPLC.
Figure imgf000004_0002
L nag pt n S- somer
Example 1 : Preparation of Linagliptin
a) Preparation of 3-methyl-7-(2-butyn-l-yl)-8-bromo-xanthine (compound of formula IV)
3-Methyl-8-bromo-xanthine (30 gm) and N,N-dimethylformamide (170 ml_) were charged into a 1000 ml_ round bottomed flask equipped with a mechanical stirrer. Diisopropylethylamine (DIPEA, 1 5.9 gm) and 1 -bromo-2-butyne (16.2 gm) were added at 30°C. The reaction mixture was heated to 85 °C and maintained the temperature for 4 hours. The reaction mixture was cooled to 30°C and pre cooled water (300 ml_) was added. The solid formed was collected by filtration and washed with pre cooled water (150 ml_) and diethyl ether (30 ml_). The solid was dried in oven under vacuum at 50°C to get 30.9 gm of the title compound.
(b) Preparation of 1 -[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8- bromoxanthine (compound of formula III) 3-Methyl-7-(2-butyn-l-yl)-8-bromo-xanthine (10 gm) and Ν,Ν-dimethylacetamide (150 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (9.3 gm) and 2-(chloromethyl)-4- methylquinazoline (6.8 gm) were added to the reaction mixture at room temperature. The reaction mixture was heated to 90 °C and maintained the temperature for 8 hours. The reaction mixture was cooled to 30°C and water (450 mL) was added and the mixture was stirred for 1 hour at 30°C. The solid formed was collected by filtration and washed with water (150 mL). The wet cake was charged into 500 mL round bottomed flask and toluene (220 mL) was added and the mixture was heated to reflux temperature and maintained for 1 hour. The mixture was cooled to 10°C and maintained for 2 hours. The solid was collected by filtration and washed with toluene (50 mL). The solid was dried in oven under vacuum at 80°C to get 10.8 gm of the title compound. Purity by HPLC: 99.59%
(c) Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (5 gm) and Ν,Ν-dimethylformamide (DMF, 50 mL) were charged into a 500 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (4.57 gm) and (R)-piperidine-3-amine dihydrochloride (2.86 gm) were added to the reaction mixture at room temperature. The reaction mixture was heated to 80 °C and maintained at that temperature for 8 hours. The reaction mixture was cooled to room temperature and DMF was evaporated under vacuum, then dichloromethane (DCM, 50 mL) was added, and stirred for 15 minutes. The reaction mixture was filtered to separate out the non- dissolved material and the non-dissolved material was washed with 15 mL of dichloromethane. The dichloromethane was evaporated under vacuum to give 4 gm of crude Linagliptin.
Example 2: One pot process for preparation of Linagliptin
3-Methyl-8-bromo-xanthine (5 gm) and Ν,Ν-dimethylformamide (DMF, 28.5 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Diisopropylethylamine (DIPEA, 2.6 gm) and 1 -bromo-2-butyne (2.7 gm) were added at 30 °C. The reaction mixture was heated to 85 °C and maintained at this temperature for 4 hours. The reaction mixture is cooled to 30°C and Ν,Ν-dimethylformamide (DMF, 100 ml_) was added. Potassium carbonate (4.4 gm) and 2-(chloromethyl)-4- methylquinazoline (4.2 gm) were added to the reaction mixture at room temperature. The reaction mixture was heated to 85 °C and maintained at this temperature for 4 hours. The reaction mixture was cooled to 30°C and Ν,Ν-dimethylformamide (DMF, 90 ml_) was added. Potassium carbonate (8.3 gm) and (R)-piperidine-3-amine dihydrochloride (5.2 gm) were added to the reaction mixture at room temperature. The reaction mixture was heated to 80 °C and maintained at this temperature for 8 hours. The reaction mixture was cooled to 30 °C and DMF was evaporated under vacuum. Dichloromethane (DCM, 30 ml_) was added and stirred for 15 minutes. The reaction mixture was filtered to separate out the undissolved material and the undissolved material was washed with dichloromethane (30 ml_). The dichloromethane was evaporated under vacuum and 10% acetic acid (100 ml_) was added. The resulted solution was stirred for 30 minutes and washed with dichloromethane (25 ml_x3). The pH of the aqueous layer was adjusted to 8.5 with 10% aqueous sodium bicarbonate solution. The aqueous layer was extracted with dichloromethane (25 ml_x2) and the dichloromethane was evaporated under vacuum to get 1 .2 gm of Linagliptin.
Example 3: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (20 gm) and methyl isobutyl ketone (MIBK 200 ml_) were charged into a 1000 ml_ round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (18.3 gm) and (R)-piperidine-3-amine dihydrochloride (1 1 .5 gm) were added to the reaction mixture at 30°C. The reaction mixture was heated to 95°C and maintained at that temperature for 8 hours. The reaction mixture was cooled to 30°C and filtered and washed with MIBK (40 ml_). The filtrate was charged into another flask and added 10% aqueous acetic acid solution and stirred for one hour at room temperature. The aqueous layer was separated and washed with 60 ml_ of dichloromethane. The aqueous layer was charged into another flask and 200 ml_ of dichloromethane and 100 ml_ of aqueous sodium hydroxide solution was added drop-wise at 30 °C. The mixture was stirred for one hour at 30 °C and the organic layer was separated and the aqueous layer was extracted with 100 ml of dichloromethane. Combined the organic layers and evaporated under vacuum at below 45°C. Isopropyl alcohol (100 mL) was added to the residue and stirred for 3 hours at room temperature. Filtered the compound and washed with isopropyl alcohol (20 mL) and dried the compound at below 60 °C under vacuum to give 17.6 gm of Linagliptin. PXRD pattern: Fig. 2, Purity: 99.0%
Example 4: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (20 gm) and methyl isobutyl ketone (MIBK 200 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (18.3 gm) and (R)-piperidine-3-amine (1 1 .5 gm) were added to the reaction mixture at room temperature. The reaction mixture was heated to 95 °C and maintained at that temperature for 8 hours. The reaction mixture was cooled to room temperature and filtered and washed with MIBK (40 mL). The filtrate was charged into another flask and added 10% aqueous acetic acid solution and stirred for one hour at room temperature. The aqueous layer was separated and washed with 60 mL of dichloromethane. The aqueous layer was charged into another flask and 200 mL of dichloromethane and 100 mL of aqueous sodium hydroxide solution (16 gm of sodium hydroxide in 100 mL of water) was added drop-wise at room temperature. The mixture was stirred for one hour at room temperature and the organic layer was separated and the aqueous layer was extracted with 100 ml of dichloromethane. Combined the organic layers and evaporated under vacuum at below 45 °C. Hexane (100 mL) was added to the residue and stirred for 3 hours at 30 °C. Filtered the compound and washed with Hexane (40 mL) and dried the compound at below 60°C under vacuum to give 17.6 gm of Linagliptin. PXRD pattern: Fig. 2, Purity: 98.92%
Example 5: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (20 gm) and methyl isobutyl ketone (MIBK 200 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (18.3 gm) and (R)-piperidine-3-amine (1 1 .5 gm) were added to the reaction mixture at 30°C. The reaction mixture was heated to 95°C and maintained at that temperature for 8 hours. The reaction mixture was cooled to 30°C and filtered and washed with MIBK (40 mL). The filtrate was charged into another flask and added 10% aqueous acetic acid solution and stirred for one hour at 30 °C. The aqueous layer was separated and washed with 60 mL of dichloromethane. The aqueous layer was charged into another flask and 200 mL of dichloromethane and 100 mL of aqueous sodium hydroxide solution (16 gm of sodium hydroxide in 100 mL of water) was added drop-wise at 30°C. The mixture was stirred for one hour at 30 °C and the organic layer was separated and the aqueous layer was extracted with 100 ml of dichloromethane. Combined the organic layers and evaporated under vacuum at below 45 °C. Toluene (100 mL) was added to the residue and stirred for 3 hours at 30 °C. Filtered the compound and washed with Toluene (40 mL) and dried the compound at below 60 °C under vacuum to give 16.8 gm of Linagliptin. Purity: 98.91 %, PXRD pattern: Fig. 2.
Example 6: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (20 gm) and methyl isobutyl ketone (MIBK 200 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (18.3 gm) and (R)-piperidine-3-amine (1 1 .5 gm) were added to the reaction mixture at 30°C. The reaction mixture was heated to 95 °C and maintained at that temperature for 8 hours. The reaction mixture was cooled to 30°C and filtered and washed with MIBK (40 mL). The filtrate was charged into another flask and added 10% aqueous acetic acid solution and stirred for one hour at 30 °C. The aqueous layer was separated and washed with 60 mL of dichloromethane. The aqueous layer was charged into another flask and 200 mL of dichloromethane and 100 mL of aqueous sodium hydroxide solution (16 gm of sodium hydroxide in 100 mL of water) was added drop-wise at room temperature (pH is > 10). The mixture was stirred for one hour 30 °C and the organic layer was separated and the aqueous layer was extracted with 100 ml of dichloromethane. Combined the organic layers and evaporated under vacuum at below 45 °C. Ethyl acetate (100 mL) was added to the residue and stirred for 3 hours at 30 °C. Filtered the compound and washed with ethyl acetate (40 mL) and dried the compound at below 60 °C under vacuum to give 17.6 gm of Linagliptin. PXRD pattern: Fig. 2, Purity: 98.72%
Example 7: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (4 gm) and methyl isobutyl ketone (MIBK 100 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (3.7 gm) and (R)-piperidine-3-amine dibenzoyl-D-tartrate (6.1 gm) were added to the reaction mixture at 26°C. The reaction mixture was heated to 100°C and maintained at that temperature for 6 hours. The reaction mixture was cooled to 30 °C and filtered, and the salt was washed with MIBK (8 mL). The filtrate was charged into another flask and added slowly 10% aqueous acetic acid solution (40 mL) and stirred for one hour at 26°C. The aqueous layer was separated and washed with 12 mL of dichloromethane. The aqueous layer was charged into another flask and 40 mL of dichloromethane and 20 mL of 16 % aqueous sodium hydroxide solution was added drop-wise at 26°C. The mixture was stirred for one hour at 26 °C and the organic layer was separated and the aqueous layer was extracted with 20 ml of dichloromethane. Combined the organic layers and evaporated under vacuum at below 45 °C. Isopropyl alcohol (8 mL) was added to the residue and evaporated under vacuum at below 45 °C. Isopropyl alcohol (16 mL) was added to the residue and stirred for 2 hours at 2Q°C. Filtered the compound and washed with isopropyl alcohol (4 mL) and dried the compound at 60 °C under vacuum to give 3.2 gm of Linagliptin. PXRD pattern: Fig. 2, Chemical Purity: 98.68%, Chiral Purity: 99.82%, S-isomer content: 0.12%, Regio impurity: 0.57%, Bromo impurity: 0.28%
Example 8: Preparation of Linagliptin
1 -[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1 -yl)-8-bromoxanthine (20 gm) and methyl isobutyl ketone (MIBK 200 mL) were charged into a 1000 mL round bottomed flask equipped with a mechanical stirrer. Potassium carbonate (18.3 gm) and (R)-piperidine-3-amine dihydrochloride (8.4 gm) were added to the reaction mixture at 26°C. The reaction mixture was heated to '\ 00 °C and maintained at that temperature for 4 hours. The reaction mixture was cooled to 30 °C and filtered and washed with MIBK (40 mL). The filtrate was charged into another flask and added 200 mL of 10% aqueous acetic acid solution and stirred for 30 minutes at 28 °C. The aqueous layer was separated and washed with 60 mL of dichloromethane. The aqueous layer was charged into another flask and 200 mL of dichloromethane and 100 mL of aqueous sodium hydroxide solution (16 gm of sodium hydroxide in 100 mL of water) were added drop- wise at 28°C (pH is > 10). The mixture was stirred for one hour at 28°C and the organic layer was separated and the aqueous layer was extracted with 100 ml of dichloromethane. Combined the organic layers and divided into 5 equal parts.
Part 1 : The organic layer was distilled off completely under vacuum at 45 °C. Methanol (8 mL) was added to the residue and distilled off completely under vacuum at 45°C. Methanol (16 mL) was added to the residue stirred for 30 minutes at 28 °C and 48 mL of MTBE was added over a period of 30 minutes to the resulted solution at 27°C and stirred for 1 hour. Filtered the compound and washed with 8 mL of MTBE and dried the compound at 65 °C under vacuum to give 3.0 gm of Linagliptin. PXRD pattern: Fig. 3. Chemical Purity: 99.46%, Regio impurity: 0.37%, Bromo impurity: 0.03%
Part 2: The organic layer was distilled off completely under vacuum at 45 °C. Methanol (8 mL) was added to the residue and distilled off completely under vacuum at 45°C. Methanol (24 mL) was added to the residue stirred for 30 minutes at 28 °C and the resulted solution was cooled to 5°C and stirred for 1 hour. Filtered the compound and washed with 5 mL of chilled methanol and dried the compound at 65°C under vacuum to give 3.0 gm of Linagliptin. PXRD pattern: Fig. 3. Chemical Purity: 99.41 %, Regio impurity: 0.38%, Bromo impurity: 0.03%
Part 3: The organic layer was distilled off completely under vacuum at 45 °C. Methanol (8 mL) was added to the residue and distilled off completely under vacuum at 45°C. Methanol (20 mL) was added to the residue stirred for 30 minutes at 28 °C and 20 mL of MTBE was added over a period of 30 minutes to the resulted solution at 27°C and stirred for 1 hour. Filtered the compound and washed with 8 mL of MTBE and dried the compound at 65 °C under vacuum to give 2.8 gm of Linagliptin. PXRD pattern: Fig. 3. Chemical Purity: 99.47%, Regio impurity: 0.36%, Bromo impurity: 0.03%.
Part 4: The organic layer was distilled off completely under vacuum at 45 °C. Isopropyl alcohol (8 mL) was added to the residue and distilled off completely under vacuum at 45 °C. Methanol (16 mL) was added to the residue stirred for 30 minutes at 28 °C and 16 mL of isopropyl alcohol was added over a period of 30 minutes to the resulted solution at 27°C and stirred for 1 hour. Filtered the compound and washed with 4 mL of isopropyl alcohol and dried the compound at 65 °C under vacuum to give 2.9 gm of Linagliptin. PXRD pattern: Fig. 1 .
Chemical Purity: 99.44%, Regio impurity: 0.38%, Bromo impurity: 0.02%.
Part 5: The organic layer was distilled off completely under vacuum at 45 °C. Ethyl acetate (8 mL) was added to the residue and distilled off completely under vacuum at 45 °C. Ethyl acetate (16 mL) was added to the residue stirred for 30 minutes at 28°C and 16 mL of methanol was added over a period of 30 minutes to the resulted solution at 27°C and stirred for 1 hour. Filtered the compound and washed with 4 mL of ethyl acetate and dried the compound at 65 °C under vacuum to give 0.7 gm of Linagliptin. PXRD pattern: Fig. 2.
Chemical Purity: 99.57%, Regio impurity: 0.29%, Bromo impurity: 0.02%
Example 9: Purification of Linagliptin
Linagliptin (3.5 gm) was dissolved in 10% aqueous acetic acid and stirred for 15 minutes. Dichloromethane (50 mL) was added to the solution and stirred for 30 minutes. The aqueous layer was separated and the pH of this layer was adjusted to 8.5 using 10% aqueous sodium bicarbonate solution. The aqueous layer was extracted with dichloromethane (50 mLx2). The dichloromethane was evaporated under vacuum to give 3 gm of Linagliptin.
Example 10: Purification of Linagliptin
Linagliptin (31 gm) and methanol (124 mL) were charged into 500 mL round bottomed flask and the solution was heated to 40 °C and stirred for 60 minutes. Charcoal (3 gm) was added to the clear solution and stirred for 30 minutes. The solution was filtered through Hy-flow and the Hy-flow bed was washed with methanol (30 mL). Filtrate was charged into 1000 mL round bottomed flask and methyl tertiary butyl ether was added drop-wise to the solution and stirred for 2 hours at 30 °C. The precipitate so formed was filtered and the wet cake was washed with methyl tertiary butyl ether (30 mL) to get 25.6 gm of pure Linagliptin. PXRD pattern: Fig. 3. Chemical Purity: 99.57%, Chiral purity: 99.73%, Regio impurity: 0.10%, Bromo impurity: 0.1 %
Example 1 1 : Purification of Linagliptin
Linagliptin (4 gm) and methanol (24 mL) were charged into 100 mL round bottomed flask and the solution is heated to 50 °C and stirred for 60 minutes. Methyl tertiary butyl ether (MTBE, 80mL) was charged into 500 mL round bottomed flask and the methanol solution containing linagliptin was added drop-wise at 27 °C and stirred for 2 hours at same temperature. The precipitate formed was filtered and the wet cake was washed with methyl tertiary butyl ether (8 mL) to get 2.6 gm of pure Linagliptin. PXRD pattern: Fig. 2, Bromo impurity content: 0.04%.
Example 12: Purification of Linagliptin
a) Preparation of linagliptin-(D)-tartrate
Linagliptin (10 gm) and methanol (300 mL) were charged into 1000 mL round bottomed flask and (D)-tartaric acid solution (3.3 gm of (D)-tartaric acid in 100 mL of methanol) was added at 26 °C. The solution was heated to 65 °C and stirred for 60 minutes. The solution was cooled to 28 °C and stirred for 2 hours at 27 °C. The precipitate formed was filtered and the wet cake was washed with methanol (20 mL) and the solid was dried under vacuum at 55°C to get 8.3 gm of Linagliptin-(D)-tartrate. PXRD pattern: Fig. 4. Chemical Purity: 99.72%, Chiral purity: 99.89%, Regio impurity: 0.08%, Bromo impurity: 0.05%, S-isomer: 0.1 1%.
b) Isolation of pure Linagliptin
Linagliptin-(D)-tartrate (8 gm) and water (100 mL) were charged into 1000 mL round bottomed flask and stirred for 30 minutes at 26 °C. Dichloromethane (80 mL) was added to the solution and cooled to 5°C. Aqueous sodium hydroxide solution (0.6 gm of NaOH is added to 20 mL of water) was added to the mixture at 5°C and maintained for 1 hour. Layers were separated and aqueous layer was extracted with dichloromethane (20 mL). Combined both organic layers and dried over sodium sulphate and distilled off the organic layer under vacuum at 45 °C. Hexane (20 mL) was added to the crude and stirred for 1 hour at 26°C. The precipitate was filtered and washed with 4 mL of hexane and dried the compound at 60°C under vacuum to give 6 gm of pure Linagliptin. PXRD pattern: Fig. 2, Chemical Purity: 99.67%, Chiral purity: 99.85%, (S)-isomer content: 0.1 5%, Regio impurity: 0.09%, Bromo impurity: 0.07%.

PATENT
http://www.google.com/patents/US20130123282
      Example 34Preparation of (R)-8-(3-amino-piperidin-1-yl)-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione (Form-XXII): A. 3-Methyl-7-(2-butyne-1-yl)-8-bromoxanthine
    • [0181]
      8-Bromo-3-methylxanthine was reacted with 1-bromo-2-butyne in the presence of base in a mixture of N-methyl pyrrolidone and toluene mixture. The reaction mixture was heated overnight. The reaction completion was determined, and the mixture was then cooled to ambient temperature. A solid precipitate formed on cooling precipitation. The product, 3-Methyl-7-(2-butyne-1-yl)-8-bromoxanthine, having greater than 95% purity was isolated by filtration and washed with toluene.
Example 35Preparation of 8-bromo-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione
    • [0182]
      3-Methyl-7-(2-butine-1-yl)-8-bromoxanthine was reacted with 2-(chloromethyl)-4-methylquinazoline in the presence of base under phase transfer catalyst using a N-methyl pyrrolidone/toluene mixture as the reaction solvent. The reaction mixture was heated overnight. When the reaction was complete, the reaction mixture was cooled to ambient temperature. A solid precipitate formed and was separated by filtration and washed with toluene and then with water to provide the product, 8-bromo-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione having more than 97% purity.
Example 36Preparation of (R)-8-(3-Amino-piperidin-1-yl)-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione (Form-XXII)
  • [0183]
    (R)-3-N-tert-Butoxycarbonylaminopiperidine was reacted with 8-bromo-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione in the presence of base. The reaction mixture was heated overnight. When the reaction was complete, the reaction mixture was cooled to ambient temperature. The cooled reaction mixture was washed several times with water and separated. The resulting 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-[(R)-3-(tert-butoxycarbonylamino)-piperidin-1-yl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purine organic solution was greater than 95%. Purified by HPLC. An excess of aqueous HCl solution was added to the obtained 1-[(4-methylquinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-[(R)-3-(tert-butoxycarbonylamino)-piperidin-1-yl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purine organic solution. The resulting mixture was stirred under heating until complete conversion was observed. Aqueous base was added to the reaction. The resulting mixture was stirred and separated. The organic phase was washed with aqueous base and separated. A non-polar or moderately polar solvent was added to the resulting organic phase. The mixture was partially concentrated to achieve precipitation, and the concentrated mixture was cooled and filtered to provide the wet crude product. The crude product was re-crystallized from alcohol, filtered and dried in vacuum oven with heating to afford dry solid Form-XXII of (R)-8-(3-amino-piperidin-1-yl)-7-(but-2-ynyl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione having more than 98% purity.

Clinical trials

Results in 2010 from a Phase III clinical trial of linagliptin showed that the drug can effectively reduce blood sugar.[2]




Scheme:
. J. Med Chem 2009, 52, 6433..
J. Med Chem 2007, 50, 6450...

References

  • H. Spreitzer (September 1, 2008). "Neue Wirkstoffe - BI-1356". Österreichische Apothekerzeitung (in German) (18/2008): 918.
  • Wang, Y, Serradell, N, Rosa, E, Castaner, R (2008). "BI-1356". Drugs of the Future 33 (6): 473–477. doi:10.1358/dof.2008.033.06.1215244.
  1. ^ "FDA Approves Type 2 Diabetes Drug from Boehringer Ingelheim and Lilly". 3 May 2011.
  2. "Four Phase III Trials Confirm Benefits of BI’s Oral, Once-Daily Type 2 Diabetes Therapy". Genetic Engineering & Biotechnology News. 28 June 2010.
CN101735218A *Dec 17, 2009Jun 16, 2010廖国超Piperidine carbamic acid ester derivative and application thereof
US7407955Aug 12, 2003Aug 5, 2008Boehringer Ingelheim Pharma Gmbh & Co., Kg8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US20040097510 *Aug 12, 2003May 20, 2004Boehringer Ingelheim Pharma Gmbh & Co. Kg8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US20090192314Mar 30, 2009Jul 30, 2009Boehringer Ingelheim International GmbhProcess for the preparation of chiral 8-(3-aminopiperidin-1yl)-xanthines
WO2005085246A1 *Feb 12, 2005Sep 15, 2005Boehringer Ingelheim Int8-[3-amino-piperidin-1-yl]-xanthine, the production thereof and the use in the form of a dpp inhibitor
Reference
1CHIRALITY vol. 7, 1995, pages 90 - 95
2*JEAN L ET AL: "A convenient route to 1-benzyl 3-aminopyrrolidine and 3-aminopiperidine", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 42, no. 33, 13 August 2001 (2001-08-13), pages 5645-5649, XP004295831, ISSN: 0040-4039, DOI: DOI:10.1016/S0040-4039(01)00985-6
Citing PatentFiling datePublication dateApplicantTitle
WO2014033746A2 *Aug 6, 2013Mar 6, 2014Glenmark Pharmaceuticals Limited; Glenmark Generics LimitedProcess for the preparation of dipeptidylpeptidase inhibitors
WO2014059938A1 *Oct 17, 2013Apr 24, 20142Y-Chem, Ltd.Method for preparing important intermediate of linagliptin
WO2014097314A1 *Dec 16, 2013Jun 26, 2014Mylan Laboratories LtdAn improved process for the preparation of linagliptin
WO2010072776A1 *Dec 22, 2009Jul 1, 2010Boehringer Ingelheim International GmbhSalt forms of organic compound
CN101784270A *Aug 15, 2008Jul 21, 2010贝林格尔.英格海姆国际有限公司Pharmaceutical composition comprising a glucopyranosyl-substituted benzene derivative
CN102127080A *Nov 2, 2005Jul 20, 2011贝林格尔.英格海姆国际有限公司Method for producing chiral 8-(3-amino-piperidin-1-yl)-xanthines
Citing PatentFiling datePublication dateApplicantTitle
WO2015067539A1 *Oct 31, 2014May 14, 2015Chemelectiva S.R.L.Process and intermediates for the preparation of linagliptin
WO2015087240A1Dec 9, 2014Jun 18, 2015Ranbaxy Laboratories LimitedProcess for the preparation of linagliptin and an intermediate thereof
WO2015107533A1 *Sep 1, 2014Jul 23, 2015Harman Finochem LimitedA process for preparation of 1h-purine-2,6-dione, 8-[(3r)-3-amino-1-piperidinyl]-7 (2-butyn-1-yl)-3,7-dihydro-3-methyl-1-[(4-methyl-2quinazolinyl) methyl] and its pharmaceutically acceptable salts


Eckhardt M, et al. 8-(3-(R)-aminopiperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydropurine-2,6-dione (BI 1356), a highly potent, selective, long-acting, and orally bioavailable DPP-4 inhibitor for the treatment of type 2 diabetes. J Med Chem. 2007; 50(26):6450-3. Pubmed ID: 18052023
2.Thomas L, et al. (R)-8-(3-amino-piperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione (BI 1356), a novel xanthine-based dipeptidyl peptidase 4 inhibitor, has a superior potency and longer duration of action compared with other dipeptidyl peptidase-4 inhibitors. J Pharmacol Exp Ther. 2008; 325(1):175-82. Pubmed ID: 18223196
Linagliptin.png
//////////BI-1356, BI1356, Linagliptin, Tradjenta, Trajenta, DPP-IV, DPP-4 inhibitor

IPI 504, Retaspamycin, Retaspimycin


IPI 504, Retaspamycin, Retaspimycin
CAS 857402-63-2
Cas 857402-23-4 ( Retaspimycin); 857402-63-2 ( Retaspimycin  HCl).
MF C31H45N3O8 BASE
MW: 587.32067 BASE
[(3R,5S,6R,7S,8E,10S,11S,12Z,14E)-6,20,22-trihydroxy-5,11-dimethoxy-3,7,9,15-tetramethyl-16-oxo-21-(prop-2-enylamino)-17-azabicyclo[16.3.1]docosa-1(22),8,12,14,18,20-hexaen-10-yl] carbamate;hydrochloride
17-Allylamino-17-demethoxygeldanamycin Hydroquinone Hydrochloride
  1. UNII-928Q33Q049
  2. SEE.........http://www.biotechduediligence.com/retaspamycin-hcl-ipi-504.html
Retaspimycin hydrochloride; 8,21-didehydro-17-demethoxy-18,21-dideoxo-18,21-dihydroxy-17-(2-propenylamino)-geldanamycin monohydrochloride
Application:A novel, water-soluble, potent inhibitor of heat-shock protein 90 (Hsp90)
Molecular Weight:624.17 ..........HCl salt
Molecular Formula:C31H46ClN3O8..........HCl salt

Introduction

IPI-504 is a novel, water-soluble, potent inhibitor of heat-shock protein 90 (Hsp90).
Orphan drug designation was assigned to the compound by the FDA for the treatment of gastrointestinal stromal cancer (GIST).

Retaspimycin Hydrochloride is the hydrochloride salt of a small-molecule inhibitor of heat shock protein 90 (HSP90) with antiproliferative and antineoplastic activities. Retaspimycinbinds to and inhibits the cytosolic chaperone functions of HSP90, which maintains the stability and functional shape of many oncogenic signaling proteins and may be overexpressed or overactive in tumor cells. Retaspimycin-mediated inhibition of HSP90 promotes the proteasomal degradation of oncogenic signaling proteins in susceptible tumor cell populations, which may result in the induction of apoptosis.
Phase I study of Retaspimycin: A phase 1 study of IPI-504 (retaspimycin hydrochloride) administered intravenously twice weekly for 2 weeks at 22.5, 45, 90, 150, 225, 300 or 400 mg/m(2) followed by 10 days off-treatment was conducted to determine the safety and maximum tolerated dose (MTD) of IPI-504 in patients with relapsed or relapsed/refractory multiple myeloma (MM). Anti-tumor activity and pharmacokinetics were also evaluated. Eighteen patients (mean age 60.5 years; median 9 prior therapies) were enrolled. No dose-limiting toxicities (DLTs) were reported for IPI-504 doses up to 400 mg/m(2).
The most common treatment-related adverse event was grade 1 infusion site pain (four patients). All other treatment-related events were assessed as grade 1 or 2 in severity. The area under the curve (AUC) increased with increasing dose, and the mean half-life was approximately 2-4 h for IPI-504 and its metabolites. Four patients had stable disease, demonstrating modest single-agent activity in relapsed or relapsed/refractory MM.  (source: Leuk Lymphoma. 2011 Dec;52(12):2308-15.)

Figure Hsp90 protein partners and clients destabilized by Hsp90 inhibition (Jackson et al., 2004).
In a different approach, Infinity Pharmaceuticals has developed IPI504 (retaspimycin or 17-AAG hydroquinone, Figure 4) (Adams et al., 2005; Sydor et al., 2006), a new GA analogue, in which the quinone moiety was replaced by a dihydroquinone one. Indeed, the preclinical data suggested that the hepatotoxicity of 17-AAG was attributable to the ansamycin benzoquinone moiety, prone to nucleophilic attack.
Furthermore, it was recently reported that the hydroquinone form binds Hsp90 with more efficiency than the corresponding quinone form (Maroney et al., 2006). In biological conditions, the hydroquinone form can interconvert with GA, depending on redox equilibrium existing in cell. It has been recently proposed, that NQ01 (NAD(P)H: quinone oxidoreductase) can produce the active hydroquinone from the quinone form of IPI504 (Chiosis, 2006).
However, Infinity Pharmaceuticals showed that if the overexpression of NQ01 increased the level of hydroquinone and cell sensitivity to IPI504, it has no significant effect on its growth inhibitory activity. These results suggest that NQ01 is not a determinant of IPI504 activity in vivo (Douglas et al., 2009).
Figure 4: GA, 17-AAG, 17-DMAG and IPI504.
IPI-504.png

PATENT

http://www.google.com/patents/EP2321645A1?cl=en
Geldanamycin (IUPAC name ([18S-(4E,5Z,8R*.9R*.10E,12R*.13S*,14R*,l6S*)]- 9- [(aminocarbonyl)oxy]- 13- hydroxy- 8,14,19- trimetoxy- 4,10,12,16- tetramethyl- 2- azabicyclo[16.3.1.]docosa- 4,6.10,18,21- pentan- 3.20,22trion) is a benzoquinone ansamycin antibiotic which may be produced by the bacterium Streptorayces hygroscopicus. Geldanamycin binds specifically to HSP90 (Heat Shock Protein 90) and alters its function.
While Hsp90 generally stabilizes folding of proteins and, in particular in tumor cells, folding of overexpressed/mutant proteins such as v-Src. Bcr-Abl and p53. the Hsp90 inhibitor Geldanamycin induces degradation of such proteins.
The respectiv e formula of geldanamycin is given herein below:
Figure imgf000022_0001
E\en though geldanamycin is a potent antitumor agent, the use of geldanamycin also shows some negathe side-effects (e.g. hepatotoxicity) which led to the dev elopment of geldanamycin analogues/derivatives, in particular analogues/deriv atives containing a derivatisation at the 17 position. Without being bound by theory , modification at the 17 position of geldanamycin may lead to decreases hepatotoxicity.
Accordingly geldanamycin analogues/derivatives which are modified at the 17 position, such as 17-AAG (17-N-Allylamino-17-demethoxygeldanamycin), are preferred in context of the present invention. Also preferred herein are geldanamycin derivatives to be used in accordance with the present invention which are water-soluble or which can be dissoh ed in water completely (at least 90 %. more preferably 95 %. 96 %. 97 %, 98 % and most preferably 99 %). 17-AAG ([QS.5S,6RJS$EΛ0R,l \SΛ2E,14E)-2\- (allylamino)-6-hydroxy-5.11-diraethoxy-
3.7.9,15-tetramethyl-16.20.22-trioxo-17-azabicyclo[16.3.1]docosa-8,12.14,18,21-pentaen-10- yl] carbamate) is. as mentioned above a preferred derivative of geldanamycin. 17- AAG is commercially available under the trade name "Tanespimycin" (also known as KOS-953) for example from Kosan Biosciences Incorporated (Acquired by Bristol-Myers Squibb Company). Tanespimycin is presently studied in phase II clinical trials for multiple myeloma and breast cancer and is usually administered intravenously.
The respective formula of 17- AAG is given herein below:
Figure imgf000023_0001
Preferred geldanamycin-derh ative (HSP90 inhibitor) to be used in context of the present invention are IPI-504 (also known as retaspiimcin or Mcdi-561 : lnfinin Pharmaceuticals (Medlmmunc/ Astra Zeneca)). Clinical trials on the use of IPI-504 (which is usually administered intravenously) in the treatment of non-small cell lung cancer (NSCLC) and breast cancer are performed. Also alvespimycin hy drochloride (Kosan Biosciences Incorporated Acquired By : Bristol-Myers Squibb Company) is a highly potent, water-soluble and orally acti\e derivative of geldanamycin preferably used in context of the present invention.
Figure imgf000024_0001
IPI-504


PATENT

WO 2005063714
http://www.google.co.ug/patents/WO2005063714A1?cl=en
Example 24
Preparation of Air-stable Hydroquinone Derivatives of the Geldanamycin Family of Molecules
,
Figure imgf000118_0001
17-Allylamino-17-Demethoxygeldanamycin (10.0 g, 17.1 mmol) in ethyl acetate
(200 mL) was stirred vigorously with a freshly prepared solution of 10% aqueous sodium hydrosulfite (200 mL) for 2 h at ambient temperature. The color changed from dark purple to bright yellow, indicating a complete reaction. The layers were separated and the organic phase was dried with magnesium sulfate (15 g). The drying agent was rinsed with ethyl acetate (50 mL). The combined filtrate was acidified with 1.5 M hydrogen chloride in ethyl acetate (12 mL) to pH 2 over 20 min. The resulting slurry was stirred for 1.5 h at ambient temperature. The solids were isolated by filtration, rinsed with ethyl acetate (50 mL) and dried at 40 °C, 1 mm Hg, for 16 h to afford 9.9 g (91%) of off-white solid. Crude hydroquinone hydrochloride (2.5 g) was added to a stirred solution of 5% 0.01 N aq. hydrochloric acid in methanol (5 mL). The resulting solution was clarified by filtration then diluted with acetone (70 mL). Solids appeared after 2-3 min. The resulting slurry was stirred for 3 h at ambient temperature, then for 1 h at 0-5 °C. The solids were isolated by filtration, rinsed with acetone (15 mL) and dried

PAPER

J. Med. Chem., 2006, 49 (15), pp 4606–4615
DOI: 10.1021/jm0603116
Abstract Image
17-Allylamino-17-demethoxygeldanamycin (17-AAG)1 is a semisynthetic inhibitor of the 90 kDa heat shock protein (Hsp90) currently in clinical trials for the treatment of cancer. However, 17-AAG faces challenging formulation issues due to its poor solubility. Here we report the synthesis and evaluation of a highly soluble hydroquinone hydrochloride derivative of 17-AAG, 1a (IPI-504), and several of the physiological metabolites. These compounds show comparable binding affinity to human Hsp90 and its endoplasmic reticulum (ER) homologue, the 94 kDa glucose regulated protein (Grp94). Furthermore, the compounds inhibit the growth of the human cancer cell lines SKBR3 and SKOV3, which overexpress Hsp90 client protein Her2, and cause down-regulation of Her2 as well as induction of Hsp70 consistent with Hsp90 inhibition. There is a clear correlation between the measured binding affinity of the compounds and their cellular activities. Upon the basis of its potent activity against Hsp90 and a significant improvement in solubility, 1a is currently under evaluation in Phase I clinical trials for cancer.
17-Allylamino-17-demethoxygeldanamycin Hydroquinone Hydrochloride Ia
17-AAG hydroquinone hydrochloride (1a) as an off-white solid (11 g, 18 mmol, 80% yield). HPLC purity:  99.6%;
IR (neat):  3175, 2972, 1728, 1651, 1581, 1546, 1456, 1392, 1316, 1224, 1099, 1036 cm-1;
1H NMR (CDCl3:d6-DMSO, 6:1, 400 MHz): 
δ 10.20 (1H, br), 9.62 (2H, br), 8.53 (1H, s), 8.47 (1H, s), 7.74 (1H, s), 6.72 (1H, d, J= 11.6 Hz), 6.28 (1H, dd, J = 11.6, 11.2 Hz), 5.73 (1H, dddd, J = 17.2, 10.0, 3.2, 2.4 Hz), 5.53 (1H, d, J = 10.4 Hz), 5.49 (1H, dd, J = 10.8, 10.0 Hz), 5.32 (2H, s), 5.04 (1H, d, J = 4.8 Hz), 5.02 (1H, d, J = 16.0 Hz), 4.81 (1H, s), 4.07 (1H, d, J = 9.6 Hz), 3.67 (2H, d, J = 6.4 Hz), 3.31 (1H, d,J = 8.8 Hz), 3.07 (3H, s), 3.07−3.04 (1H, m), 2.99 (3H, s), 2.64 (1H, m), 2.52−2.49 (1H, m), 1.76 (3H, s), 1.61−1.39 (3H, m), 0.78 (3H, d, J = 6.4 Hz), 0.64 (3H, d, J = 7.2 Hz);
13C NMR (CDCl3:d6-DMSO, 6:1, 100 MHz):  δ 167.3, 155.8, 143.3, 136.3, 135.0, 134.2, 132.9, 132.1, 128.8, 127.6, 125.9, 125.3, 123.7, 123.0, 115.1, 104.5, 80.9, 80.7, 80.1, 72.5, 56.2, 56.2, 52.4, 34.6, 33.2, 31.1, 27.2, 21.6, 12.1, 12.1, 11.7;
HRMS calculated for C31H45N3O8 (M+ + H):  588.3285, Found 588.3273.

POSTER

Synthesis and biological evaluation of IPI-504, an aqueous soluble analog of 17-AAG and potent inhibitor of Hsp90

MEDI 210

James R. Porter, jporter@ipi.com, Jie Ge, Emmanuel Normant, Janid Ali, Marlene S. Dembski, Yun Gao, Asimina T. Georges, Louis Grenier, Roger Pak, Jon Patterson, Jens R. Sydor, Jim Wright, Julian Adams, and Jeffrey K. Tong.
 
Infinity Pharmaceuticals, Inc, 780 Memorial Drive, Cambridge, MA 02139
IPI-504 is the hydroquinone hydrochloride salt of 17-allylamino-17-demethoxy-geldanamycin (17-AAG), an Hsp90 inhibitor that is currently in clinical trials for the treatment of cancer.
IPI-504 demonstrates high aqueous solubility (>200 mg/mL). Interestingly, in vitro and in vivo IPI-504 interconverts with 17-AAG and exists in a pH and enzyme-mediated redox equilibrium. This occurs due to oxidation of the hydroquinone (IPI-504) to the quinone (17-AAG) at physiological pH and the reduction of 17-AAG by quinone reductases such as NQO1 to IPI-504.
Here we report the design and synthesis of the stabilized hydroquinone IPI-504 and its inhibitory effect against Hsp90 and Grp94. Although IPI-504 was originally designed to be a soluble prodrug of 17-AAG, the hydroquinone is more potent than the quinone in the biochemical Hsp90 binding assay.
Various hydroquinone analogs have been prepared to investigate the structure activity relationship of hydroquinone binding to Hsp90. Hydroquinone and quinone forms of 17-AAG metabolites show comparable binding affinities for Hsp90 and in cancer cell lines, hydroquinone analogs elicit specific responses consistent with Hsp90 inhibition.
The desirable pharmacological properties as well as in vitro and in vivo activity of our lead compound, IPI-504, has led to the initiation of Phase I clinical trials in multiple myeloma.
 http://oasys2.confex.com/acs/231nm/techprogram/P945016.HTM


References

Synthesis and biological evaluation of IPI-504, an aqueous soluble analog of 17-AAG and potent inhibitor of Hsp90
231st Am Chem Soc (ACS) Natl Meet (March 26-30, Atlanta) 2006, Abst MEDI 210
Design, synthesis, and biological evaluation of hydroquinone derivatives of 17-amino-17-demethoxygeldanamycin as potent, water-soluble inhibitors of Hsp90
J Med Chem 2006, 49(15): 4606
http://www.biotechduediligence.com/retaspamycin-hcl-ipi-504.html
///////////////////Hsp90, IPI-504, infinity pharma, Retaspamycin, Retaspimycin

Monday 26 October 2015

IPI 926, Saridegib, Patidegib

Saridegib3Dan.gif
Saridegib.svg
IPI 926, Saridegib, Patidegib
C29H48N2O3S
Exact Mass: 504.33856
1037210-93-7
2D chemical structure of 1169829-40-6
  • Patidegib hydrochloride
  • Saridegib hydrochloride
    • C29-H48-N2-O3-S.Cl-H
    • 541.2361
http://chem.sis.nlm.nih.gov/chemidplus/rn/1169829-40-6
Methanesulfonamide, N-((2S,3R,3'R,3aS,4'aR,6S,6'aR,6'bS,7aR,12'aS,12'bS)-2',3',3a,4,4',4'a,5,5',6,6',6'a,6'b,7,7',7a,8',10',12',12'a,12'b-eicosahydro-3,6,11',12'b-tetramethylspiro(furo(3,2-b)pyridine-2(3H),9'(1'H)-naphth(2,1-a)azulen)-3'-yl)-, hydrochloride (1:1)
 CAS 1169829-40-6 HCL
Saridegib also known as IPI-926 is an experimental drug candidate undergoing clinical trials for the treatment of various types of cancer, including hard to treat hematologic malignancies such as myelofibrosis and ligand-dependant tumors such as chondrosarcoma.[1] IPI-926 exhibits its pharmacological effect by inhibition of the G protein-coupled receptor smoothened, a component of the hedgehog signaling pathway.[2]
Chemically, it is a semi-synthetic derivative of the alkaloid cyclopamine. The process begins with cyclopamine extracted from harvested Veratrum californicum which is taken through a series of alterations resulting in an analogue of the natural product cyclopamine, making IPI-926 the only compound in development/testing that is not fully synthetic.[2]
ChemSpider 2D Image | N-[(2S,3R,3'R,3aR,4a'R,6S,6a'R,6b'S,7aR,12a'S,12b'S)-3,6,11',12b'-Tetramethyl-2',3',3a,4,4',4a',5,5',6,6',6a',6b',7,7',7a,8',10',12',12a',12b'-icosahydro-1'H,3H-spiro[furo[3,2-b]pyridine-2,9'-naphtho[ 2,1-a]azulen]-3'-yl]methanesulfonamide | C29H48N2O3S
Saridegib is a member of a class of anti-cancer compounds known as hedgehog inhibitors (Hhi). Most of these compounds affect thehedgehog signaling pathway via inhibition of smoothened (Smo), a key component of the pathway. Depending on when a Hh inhibiting compound is approved by the U.S. Food and Drug Administration (FDA), there may be a perceived need for one to be differentiated over another for marketing purposes, which could lead to different nomenclature (e.g., a Hhi or an agonist of Smo).
This marketing technique is more of a differentiation strategy than a scientific property of these compounds, as the mechanism of action (MOA) in the end is inhibition of the Hh pathway, targeting cancer stem cells. However, as these new compounds are further studied, identification of differences in a compound's MOA, could lead to hypotheses regarding the stage at which Smo is inhibited, where along the pathway the compound binds, or specific binding properties of a compound.
If these hypotheses are proven, claims could be made regarding a specific compound's MOA and how it affects efficacy, safety, combinability with other cancer treatments, etc. Scientific data in support of such hypotheses have not been published to date.
SARIDEGIB

N-[(3R,3'R,3'aS,4aR,6'S,6aR,6bS,7'aR,9S,12aS,12bS)-3',6',11,12b-tetramethylspiro[1,2,3,4,4a,5,6,6a,6b,7,8,10,12,12a-tetradecahydronaphtho[2,1-a]azulene-9,2'-3a,4,5,6,7,7a-hexahydro-3H-furo[3,2-b]pyridine]-3-yl]methanesulfonamide
There are currently no drugs in the Hhi class FDA approved, however IPI-926 and GDC-0449 are the 2 leading compounds in the class. IPI-926, GDC-0449, and LDE-225 are the only compounds that have generic names passed by the United States Adopted Name (USAN) council (Infinity IPI-926/saridegib, Genentech GDC-0449/vismodegib, and Novartis LDE-225/erismodegib). Although Infinity is further along in chondrosarcoma, myelofibrosis, and AML, Roche/Genentech recently submitted an NDA for GDC-0449 for the treatment of adults with advanced basal cell carcinoma (BCC) when surgery is no longer an option, and the FDA has accepted and has filed the NDA, giving it priority review status. Thus it appears that Roche/Genentech will be the first Hhi to market with GDC-0449, if approved, for the treatment of advanced BCC, with Infinity second to market with IPI-926 for treatment in chondrosarcoma. It appears Infinity will not pursue an indication for BCC and focus on cancers with high unmet needs.[1][3][4][5][6]
Other Hhi-class compounds not as far along in development as IPI-926 and GDC-0449 include:[7]
  • Novartis' LDE-225 (USAN generic name erismodegib)
  • Exelixis/Bristol-Myers Squibb's BMS-833923 (XL139)
  • Millennium Pharmaceuticals's TAK-441
  • Pfizer's PF-04449913

 

Fig 1. Chemical structure comparison between IPI-926 and cyclopamine
IPI-926 is currently developed by Infinity Pharmaceuticals, Inc. Malignant activation of the Hedgehog pathway is implicated in multiple cancer settings and Infinity's development strategy is designed to enable IPI-926 to target a broad range of critical oncology targets - from the tumor cell to the cancer microenvironment. This broadly applicable, targeted approach represents an innovative method for fighting cancer and has potential in treating a range of cancers, including pancreatic cancer, small cell lung cancer, ovarian cancer, bladder cancer, medulloblastoma, basal cell carcinoma, and certain hematological malignancies.
The hedgehog pathway inhibitor IPI-926 has been in clinical investigation for basal cell carcinoma, chondrosarcoma, and pancreatic cancer. In the final step of the synthesis of IPI-926  the drug substance (DS) is isolated as the hydrochloride salt of the 2-propanol (2-PrOH) solvate
Abstract Image
A design of experiments (DoE) approach was taken to optimize purity and reaction yield of the final debenzylation and hydrochloride salt formation of IPI-926. The study involved a careful dissection of the different process steps to enable an independent investigation of these steps while ensuring that process streams were representative. The results enabled a streamlined process from the final chemical transformation to the salting and isolation and led to the elimination of variability in the process as well as a robust control of impurities. The optimized process was applied to production and demonstrated on the kilogram scale.

A Design of Experiments Approach to a Robust Final Deprotection and Reactive Crystallization of IPI-926, A Novel Hedgehog Pathway Inhibitor

Infinity Pharmaceuticals, 784 Memorial Drive, Cambridge, Massachusetts 02139, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00214
The product was dried at a jacket temperature of 45 °C until an LOD <2.30% (w/w) was achieved. Yield: 11.5 kg (73% from compound 1, correcting for the seed). HPLC purity: 99.9% area (compound 2 content: 0.08% w/w). Assay: 83.7% w/w (as-is), 99.1% w/w (anhydrous, solvent-free). Moisture content: 1.6% w/w. Chlorine content: 5.72% w/w. Residual solvents: acetone (720 ppm); acetonitrile (<41 ppm); 2-MeTHF (none detected); 2-propanol (81 147 ppm); toluene (<90 ppm). Residual metals: palladium (0 ppm); platinum (0 ppm); ruthenium (0 ppm). Additional data for the IPI-926 free base:
1H NMR (400 MHz, CDCl3) 6.90 (br s, 1H), 3.31 (dt, J = 10.6, 3.8 Hz, 1H), 3.20 (br s, 1H), 3.10 (dd, J = 13.7, 4.5 Hz, 1H), 2.91 (s, 3H), 2.62 (dd,J = 9.9, 7.6 Hz, 1H), 2.33 (br d, J = 14.5 Hz, 1H), 2.27–2.15 (m, 1H), 2.10 (dd, J = 14.5, 6.9 Hz, 1H), 1.99–1.17 (m, 28H), 1.05 (q, J = 11.6 Hz, 1H), 0.93 (d, J = 7.4 Hz, 3H), 0.88 (d, J = 6.6 Hz, 3H), 0.86 (s, 3H) ppm.
13C NMR (100 MHz, CDCl3) 140.47, 124.53, 82.48, 76.97, 63.73, 54.08, 53.87, 50.12, 49.98, 47.19, 44.73, 42.27, 42.10, 40.24, 37.55, 37.44, 36.04, 34.44, 31.87, 31.33, 30.46, 29.79, 28.37, 27.94, 26.26, 24.19, 22.70, 18.92, 10.19 ppm;
MS: m/z = 505.29 [M + H]+.
PAPER
Tremblay, M. R.; Lescarbeau, A.; Grogan, M. J.; Tan, E.; Lin, G.; Austad, B. C.; Yu, L.-C.;Behnke, M. L.; Nair, S. J.; Hagel, M.; White, K.; Conley, J.; Manna, J. D.; Alvarez-Diez, T. M.; Hoyt, J.; Woodward, C. N.; Sydor, J. R.; Pink, M.; MacDougall, J.; Campbell, M. J.;Cushing, J.; Ferguson, J.; Curtis, M. S.; McGovern, K.; Read, M. A.; Palombella, V. J.;Adams, J.; Castro, A. C. J. Med. Chem. 2009, 52, 44004418, DOI: 10.1021/jm900305z
J. Med. Chem., 2009, 52 (14), pp 4400–4418
DOI: 10.1021/jm900305z
Abstract Image
Recent evidence suggests that blocking aberrant hedgehog pathway signaling may be a promising therapeutic strategy for the treatment of several types of cancer. Cyclopamine, a plant Veratrum alkaloid, is a natural product antagonist of the hedgehog pathway. In a previous report, a seven-membered D-ring semisynthetic analogue of cyclopamine, IPI-269609 (2), was shown to have greater acid stability and better aqueous solubility compared to cyclopamine. Further modifications of the A-ring system generated three series of analogues with improved potency and/or solubility. Lead compounds from each series were characterized in vitro and evaluated in vivo for biological activity and pharmacokinetic properties. These studies led to the discovery of IPI-926 (compound 28), a novel semisynthetic cyclopamine analogue with substantially improved pharmaceutical properties and potency and a favorable pharmacokinetic profile relative to cyclopamine and compound2. As a result, complete tumor regression was observed in a Hh-dependent medulloblastoma allograft model after daily oral administration of 40 mg/kg of compound 28.
28 (4.06 g, 8.05 mmol, 95% for two steps). NMR δH (400 MHz, CDCl3) 6.90 (br s, 1H), 3.31 (dt, J = 10.6, 3.8 Hz, 1H), 3.20 (br s, 1H), 3.10 (dd, J = 13.7, 4.5 Hz, 1H), 2.91 (s, 3H), 2.62 (dd, J = 9.9, 7.6 Hz, 1H), 2.33 (br d, J = 14.5 Hz, 1H), 2.27−2.15 (m, 1H), 2.10 (dd, J = 14.5, 6.9 Hz, 1H), 1.99−1.17 (m, 28H), 1.05 (q, J = 11.6 Hz, 1H), 0.93 (d, J = 7.4 Hz, 3H), 0.88 (d, J = 6.6 Hz, 3H), 0.86 (s, 3H); NMR δC (100 MHz, CDCl3) 140.47, 124.53, 82.48, 76.97, 63.73, 54.08, 53.87, 50.12, 49.98, 47.19, 44.73, 42.27, 42.10, 40.24, 37.55, 37.44, 36.04, 34.44, 31.87, 31.33, 30.46, 29.79, 28.37, 27.94, 26.26, 24.19, 22.70, 18.92, 10.19; m/z = 505.29 [M + H]+; HPLC 99.1 a/a % at 215 nm.
sari 13c sari mass sari1h nmr

Click on images for clear view.................

 

 

 

Paper
Abstract Image
A design of experiments (DoE) approach was taken to optimize purity and reaction yield of the final debenzylation and hydrochloride salt formation of IPI-926. The study involved a careful dissection of the different process steps to enable an independent investigation of these steps while ensuring that process streams were representative. The results enabled a streamlined process from the final chemical transformation to the salting and isolation and led to the elimination of variability in the process as well as a robust control of impurities. The optimized process was applied to production and demonstrated on the kilogram scale.
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00214..........http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00214
 IPI-926 free base:
1H NMR (400 MHz, CDCl3) 6.90 (br s, 1H), 3.31 (dt, J = 10.6, 3.8 Hz, 1H), 3.20 (br s, 1H), 3.10 (dd, J = 13.7, 4.5 Hz, 1H), 2.91 (s, 3H), 2.62 (dd,J = 9.9, 7.6 Hz, 1H), 2.33 (br d, J = 14.5 Hz, 1H), 2.27–2.15 (m, 1H), 2.10 (dd, J = 14.5, 6.9 Hz, 1H), 1.99–1.17 (m, 28H), 1.05 (q, J = 11.6 Hz, 1H), 0.93 (d, J = 7.4 Hz, 3H), 0.88 (d, J = 6.6 Hz, 3H), 0.86 (s, 3H) ppm.
13C NMR (100 MHz, CDCl3) 140.47, 124.53, 82.48, 76.97, 63.73, 54.08, 53.87, 50.12, 49.98, 47.19, 44.73, 42.27, 42.10, 40.24, 37.55, 37.44, 36.04, 34.44, 31.87, 31.33, 30.46, 29.79, 28.37, 27.94, 26.26, 24.19, 22.70, 18.92, 10.19 ppm;
MS: m/z = 505.29 [M + H]+.

References

  1.  "Pipeline: IPI-926". Infinity Pharmaceuticals.
  2.  Tremblay, MR; Lescarbeau, A; Grogan, MJ; Tan, E; Lin, G; Austad, BC; Yu, LC; Behnke, ML et al. (2009). "Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926)". Journal of Medical Chemistry 52 (14): 4400–18. doi:10.1021/jm900305z. PMID 19522463.
  3.  "Pipeline". Infinity Pharmaceuticals.
  4.  "Genentech Pipeline". Genentech.
  5.  "USAN Stem List" (PDF). AMA.
  6.  "Names under consideration". AMA.
  7.  "Search results for Hh clinical trials". United National Institute of Health's ClinicalTrials.gov.
  8. 1. Tremblay MR, Lescarbeau A, Grogan MJ, Tan E, Lin G, Austad BC, Yu LC, Behnke ML, Nair SJ, Hagel M et al.. (2009)
    Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926).
    J. Med. Chem.52 (14): 4400-18.
Saridegib
Saridegib.svg
Saridegib3Dan.gif
Names
IUPAC name
N-((2S,3R,3aS,3′R,4a′R,6S,6a′R,6b′S,7aR,12a&prmie;S,12b′S)-3,6,11′,12b′-tetramethyl-2′,3a,3′,4,4′,4a′,5,5&prmie;,6,6′,6a′,6b′,7,7a,7′,8′,10′,12′,12a′,12b′-icosahydro-1′H,3H-spiro[furo[3,2-b]pyridine-2,9'-naphtho[2,1-a]azulen]-3'-yl)methanesulfonamide
Other names
saridegib
Identifiers
1037210-93-7 Yes
ChEMBLChEMBL538867
ChemSpider26353073
8198
Jmol-3D imagesImage
PubChem25027363
UNIIJT96FPU35X Yes
Properties
C29H48N2O3S
Molar mass504.77 g·mol−1
Pharmacology
Legal status
  • Investigational
/////Saridegib, IPI-926

Sunday 18 October 2015

Synthesis of a fluorinated Ezetimibe analogue

f eze nmr
Synthesis of a fluorinated Ezetimibe analogue using radical allylation of [small alpha]-bromo-[small alpha]-fluoro-[small beta]-lactam
New J. Chem., 2015, Advance Article
DOI: 10.1039/C5NJ01969A, Paper
Atsushi Tarui, Ayumi Tanaka, Masakazu Ueo, Kazuyuki Sato, Masaaki Omote, Akira Ando
 
*Corresponding authors
aFaculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Japan
E-mail: aando@pharm.setsunan.ac.jp
A facile and efficient synthesis of a fluorinated Ezetimibe analogue was achieved by radical allylation, Wacker oxidation, and nucleophilic arylation of [small alpha]-bromo-[small alpha]-fluoro-[small beta]-lactam
The synthesis of an α-fluoro-β-lactam-containing Ezetimibe analogue was accomplished starting from α-bromo-α-fluoro-β-lactam which was readily prepared from ethyl dibromofluoroacetate. A facile and efficient method for the introduction of the C3 alkyl side chain was realized via radical allylation. The diastereoselective allylation of α-bromo-α-fluoro-β-lactam was successfully applied to construct the relative configuration of the β-lactam nucleus between C3 and C4. Further modification of the allyl side chain gave the 3′-(4-fluorophenyl)-3′-hydroxypropyl group through Wacker oxidation and nucleophilic arylation.
http://pubs.rsc.org/en/Content/ArticleLanding/2015/NJ/C5NJ01969A?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FNJ+%28RSC+-+New+J.+Chem.+latest+articles%29#!divAbstract