AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Worlddrugtracker, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his PhD from ICT ,1991, Mumbai, India, in Organic chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA as ADVISOR earlier GLENMARK LS Research centre as consultant,Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Prior to joining Glenmark, he worked with major multinationals like Hoechst Marion Roussel, now sSanofi, Searle India ltd, now Rpg lifesciences, etc. he is now helping millions, has million hits on google on all organic chemistry websites. His New Drug Approvals, Green Chemistry International, Eurekamoments in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 year tenure, good knowledge of IPM, GMP, Regulatory aspects, he has several international drug patents published worldwide . He gas good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, polymorphism etc He suffered a paralytic stroke in dec 2007 and is bound to a wheelchair, this seems to have injected feul in him to help chemists around the world, he is more active than before and is pushing boundaries, he has one lakh connections on all networking sites, He makes himself available to all, contact him on +91 9323115463, amcrasto@gmail.com

Tezosentan disodium for pulmonary hypertension

 Uncategorized  Comments Off on Tezosentan disodium for pulmonary hypertension
Jan 242014
 

TEZOSENTAN

180384-57-0  CAS OF FREE ACID

N-[6-(2-Hydroxyethoxy)-5-(2-methoxyphenoxy)-2-[2-(2H-tetrazol-5-yl)pyridin-4-yl]pyrimidin-4-yl]-5-propan-2-ylpyridine-2-sulfonamide

5-isopropyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5- (2-methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-ylamide

Formula C27H27N9O6S 
Mol. mass 605.624

…………………………………………………………………………….

 

Tezosentan disodium, Ro-61-0612, Veletri

5-isopropyl-pyridine-2-sulfonic acid [6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide sodium salt (1:2)

180384-58-1 of disodium salt, 180384-57-0 (free acid)

MF C27-H25-N9-O6-S.2-Na
MW 649.5975
Roche (Originator), Actelion (Licensee), Genentech (Codevelopment)
CARDIOVASCULAR DRUGS, Heart Failure Therapy, Renal Failure, Agents for, RENAL-UROLOGIC DRUGS, Treatment of Renal Diseases, Endothelin ETA Receptor Antagonists, Endothelin ETB Receptor Antagonists
Phase III

TEZOSENTAN

Tezosentan is a non-selective ETA and ETB receptor antagonist.[1] It acts as a vasodilator and was designed as a therapy for patients with acuteheart failure. Recent studies have shown however, that tezosentan does not improve dyspnea or reduce the risk of fatal or nonfatal cardiovascular events.[2]

Pulmonary disease (COPD), which may possibly be associated with pulmonary hypertension, as well as allergic and non-allergic rhinitis, provided that treatment with endothelin from a therapeutic standpoint is not contraindicated.

Tezosentan disodium is an endothelin ETB receptor antagonist in phase II clinical development for the treatment of stable, chronic pulmonary arterial hypertension. The drug was previously being evaluated for heart failure, but trials in that indication have been discontinued. The compound is being developed by Actelion.

………………………………..

SYNTHESIS

 

………………………………..

SYNTHESIS

 

Reaction of 4-cyano-pyridine (I) with Na in methanol followed by treatment with ammonium chloride provides 4-amidino-pyridine hydrochloride (II), which is then converted into 5-(2-methoxyphenoxy)-2-(pyridin-4-yl)-pyrimidine-4,6-diol (IV) by condensation with diethyl malonate derivative (III) by means of Na in MeOH. By heating compound (IV) with phosphorus oxychloride (POCl3), 4,6-dichloro-5-(2-methoxyphenoxy)-2-pyridin-4-yl)pyrimidine (V) is obtained, which in turn is oxidized with peracetic acid in refluxing acetonitrile to afford N-oxide derivative (VI). Condensation of (VI) with 5-isopropylpyridine-2-sulfonamide potassium (VII) furnishes 5-isopropylpyridine-2-sulfonic acid 6-chloro-5-(2-methoxyphenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-yl amide (VIII), which is then dissolved in dimethoxyethane and subjected to reaction with Na in hot ethylene glycol (IX) to provide N-[6-(2-hydroxyethoxy)-5-(2-methoxyphenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-yl]-5-isopropylpyridine-2-sulfonamide (X). Refluxing of (X) with trimethylsilylcyanide and Et3N in acetonitrile yields cyano derivative (XI), which is then converted into the tetrazole derivative (XII) by reaction with sodium azide and NH4Cl in DMF at 70 C. Finally, the disodium salt of tezosentan is obtained by treatment of (XII) with Na/MeOH in THF. refEP 0799209; JP 1998509182; WO 9619459

…………………………………..

SYNTHESIS PROCEDURE as in    EP0979822A1

Examples

        Example 1

      • 1360 ml of formamide were added to 136 g (437 mmol) of 5-(2-methoxy-phenoxy)-2-pyridine-4-yl-pyrimidine-4,6-diole. Then, at a temperature of 0°C, 11.7 ml (219 mmol) of concentrated sulfuric acid and thereafter 36.5 g (130 mmol) of iron(II)sulfate heptahydrate were added to the suspension. After that, 89 ml (874 mmol) of 30% hydrogen peroxide were added dropwise within 1 hr at a temperature of 0°C to 5°C. The viscous yellow-brownish suspension was stirred at 0°C for 1.5 hr. Subsequently, a solution of 83 g (437 mmol) of sodium pyrosulfite in 680 ml of de-ionized water was added dropwise to the reaction mixture within 30 min. at 0°C to 5°C and the reaction mixture was stirred at 0°C to 5°C for 30 min. The suspension was then filtered under reduced pressure. The filtrate was first washed with 1750 ml of de-ionized water and thereafter with 700 ml of ethanol. Then the solid was dried at 80°C, 2000 Pa for 16 hr. There were obtained 132.4 g (91% of theory) of 4-[4,6-dihydroxy-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carboxylic acid amide with a HPLC purity of 91.4% (w/w).
      • Preparation of starting material:
        • a) 53.1 g of 4-cyano-pyridine (98%) are added all at once to a solution of 1.15 g of sodium in 200 ml of abs. MeOH. After 6 hr 29.5 g of NH4Cl are added while stirring vigorously. The mixture is stirred at room temperature overnight. 600 ml of ether are added thereto, whereupon the precipitate is filtered off under suction and thereafter dried at 50°C under reduced pressure. There is thus obtained 4-amidino-pyridine hydrochloride (decomposition point 245-247°C).
        • b) 112.9 g of diethyl (2-methoxyphenoxy)malonate are added dropwise within 30 min. to a solution of 27.60 g of sodium in 400 ml of MeOH. Thereafter, 74.86 g of the amidine hydrochloride obtained in a) are added all at once. The mixture is stirred at room temperature overnight and evaporated at 50°C under reduced pressure. The residue is treated with 500 ml of ether and filtered off under suction. The filter cake is dissolved in 1000 ml of H2O and treated little by little with 50 ml of CH3COOH. The precipitate is filtered off under suction, washed with 400 ml of H2O and dried at 80°C under reduced pressure. There is thus obtained 5-(2-methoxy-phenoxy)-2-(pyridine-4-yl)-pyrimidine-4,6-diole (or tautomer), melting point above 250°C.

Example 2

    • Within 20 min. 61 ml (633 mmol) of POCl3 were added dropwise to 34 ml (200 mmol) of diisopropyl ethylamine at 5°C to 10°C followed by stirring at 5°C to 10°C for 15 min. Then 23.5 g (66 mmol) of 4-[4,6-dihydroxy-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carboxylic acid amide were added in four portions under cooling followed by stirring at 90°C for 25 hr. The reaction mixture was cooled down to 20°C and transferred to a new flask together with 50 ml of dichloromethane. Volatile components (i.e. excess of POCl3) was removed by evaporation from 20°C to 70°C followed by re-distillation with 100 ml of toluene. After adding 250 ml of dichloromethane to the residue (88 g of a black oil) the solution was heated to 35°C to 40°C and 80 ml of de-ionized water were added dropwise within 30 min. whereby the pH was kept constant by the subsequent addition of 28% NaOH solution (60 ml) within 5 to 6 hr. The mixture was stirred at 35°C to 40°C for 30 min. followed by removal of dichloromethane by distillation. The resulting suspension was allowed to cool down to 20°C and was stirred for additional 2 hr. The solid was filtered off under suction, washed with 500 ml of water and dried at 70°C, 2000 Pa for 16 hr. There were obtained 21.3 g (86% of theory) of 4-[4,6-dichloro-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carbonitrile with a HPLC purity of 94.3% (w/w).

 

      Example 4

    • 8.95 g (24 mmol) of 4-[4,6-dichloro-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carbonitrile were suspended in 100 ml of acetone. At a temperature of 20°C, 5.04 g (25 mmol) of 5-isopropyl-pyridine-2-sulfonamide, 1 ml of de-ionized water, 10.6 g (77 mmol) of potassium carbonate and 135 mg (1.2 mmol) 1,4-diazobicyclo[2.2.2]octane were added. The mixture was stirred at 40°C for 20 hr. Thereafter, another 240 mg (1.2 mmol) of 5-isopropyl-pyridine-2-sulfonamide and 80 mg (0.7 mmol) of 1,4-diazobicyclo[2.2.2]octane were added. The reaction mixture was stirred for 24 hr at 40°C followed by cooling to 20°C. Then 50 ml of de-ionized water and 45 ml of 3 N aqueous hydrochloric acid were added slowly until pH = 1. The acetone was removed by distillation and the resulting suspension was stirred at 20°C for 1.5 hr. The solid was filtered off under suction, washed first with 100 ml of de-ionized water and thereafter with 50 ml of t-butylmethylether. Then the solid was dried at 70°C, 2000 Pa for 20 hr. There were obtained 13.2 g (102% of theory) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-2-(2-cyano-pyridine-4-yl)-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide with a HPLC purity of 87.8% (w/w).

 

      Example 6

    • 122 g (233 mmol) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-2-(2-cyano-pyridine-4-yl)-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide was suspended in 450 ml of N,N-dimethyl formamide and the mixture was cooled down to 15°C. At this temperature, 35 ml of hydrazine hydrate were added dropwise within 1 hr. The resulting solution was stirred at 15°C to 20°C for 16 hr and thereafter diluted with 600 ml of de-ionized water. Then 50 ml of glacial acetic acid were added dropwise at 0°C to 5°C until pH = 5.5. 600 g of ice were added and the suspension was stirred for 1 hr. The solid was filtered off under suction, washed with 3000 ml of water and dried at 40°C, 2000 Pa for 24 hr. There were obtained 126 g (97% of theory) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-2-[2-(hydrazino-imino-methyl)-pyridine-4-yl]-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide with a HPLC purity of 91.8% (w/w).

 

      Example 8

    • 20 g (35 mmol) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-2-[2-(hydrazino-imino-methyl)-pyridine-4-yl]-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide were added to 160 ml of N,N-dimethyl formamide. The solution was kept at 15°C to 20°C and 23 ml of 6 N aqueous hydrochloric acid were added, followed by addition of a solution containing 4.8 g (7 mmol) of sodium nitrite in 20 ml de-ionized water within 10 min. The mixture was stirred at 20°C for 1 hr, then 140 ml of de-ionized water were added and the suspension was stirred at 0°C for 1 hr. The solid was filtered, firstly washed with 80 ml of de-ionized water and thereafter with 80 ml of t-butylmethylether. Then the solid was dried at 70°C and 2000 Pa for 16 hr. The crude product (23.4 g) was taken up with 117 ml of tetrahydrofuran for 1 hr. After filtration at 0°C the crystallized product was washed with 25 ml of t-butylmethylether and was then dried at 70°C, 2000 Pa for 16 hr. There were obtained 17.3 g (84% of theory) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide with a HPLC purity of 91.1% (w/w).

 

    Example 10

  • 6.2 g of sodium hydroxide were added to 15 g (26 mmol) of 5-isopropyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amid and 75 ml of ethylene glycol. The mixture was heated to 85°C for 5 hr. Then 55 ml of de-ionized water were added and thereafter 55 ml of 3 N hydrochloric acid were added dropwise. The mixture was allowed to cool down to 20°C and was stirred for 1 hr. The solid was filtered off and dried at 70°C, 2000 Pa for 18 hr. There were obtained 16.2 g (103%) of 5-isopropyl-pyridine-2-sulfonic acid 16-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide with a HPLC purity of 92% (w/w). 80 ml of dioxane and 80 ml of ethanol were added to this solid. At a temperature of 60°C, gaseous ammonia was introduced into the liquid until pH = 9 to 10. The resulting suspension was allowed to cool down to 20°C and was stirred at 20°C for 20 hr and thereafter at 0°C for 2.5 hr. Then the solid was filtered off and dried at 70°C, 2000 Pa for 18 hr. There were obtained 14.2 g of mono ammonium salt with a HPLC purity of 96.2% (w/w). The solid was heated (reflux) in 70 ml of methanol, cooled down slowly to 20°C and stirred at 20°C for 19 hr and thereafter at 0°C for 2 hr. Then the solid was filtered off and dried at 70°C, 2000 Pa for 19 hr. There were obtained 11.5 g (66% of theory) of 5-isopropyl-pyridine-2-sulfonic acid [6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide sodium salt (1:2) with a HPLC purity of 98.6% (w/w).

 

 

 

Reaction of 2-chloro-5-ispropylpyridine (VII) with thiourea (A) in aqueous HCl gives 5-isopropyl- pyridine-2-thiol (VIII), which is chlorinated with chlorine in acetic acid to yield 5-isopropylpyridine-2-sulfochloride (IX). This compound is converted into 5-isopropylpyridine-2-sulfonamide potassium salt (X).

…………………………

synthesis

WO1996019459A1

. Example 1

a) 200 ml of dimethoxyethane and 1 10.9 g of 4-[4-(4-tert- butyl-phenyl-sulphonylamino)-6-chloro-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide are added all at once to a solution of 23.80 g of sodium in 660 ml of ethylene glycol. The solution is heated at 90°C for 20 hours while stirring, thereafter cooled, poured into 2500 ml of H2O and thereafter treated with CH3COOH to pH 5. The mixture is extracted three times with EtOAc, the organic phase is washed with H2O, dried with Na2Sθ4 and evaporated under reduced pressure. The residue is recrystall- ized from CH3CN and thereafter twice from a mixture of acetone and CH3CN. There is thus obtained 4-[4-(4-tert-butyl-phenyl- sulphonylamino)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide.

Preparation of the starting material:

b) 53.1 g of 4-cyano-pyridine (98%) are added all at once to a solution of 1.15 g of sodium in 200 ml of abs. MeOH. After

6 hours 29.5 g of NH4CI are added while stirring vigorously. The mixture is stirred at room temperature overnight. 600 ml of ether are added thereto, whereupon the precipitate is filtered off under suction and thereafter dried at 50°C under reduced pressure. There is thus obtained 4-amidino-pyridine hydro- chloride (decomposition point 245-247°C).

c) 1 12.9 g of diethyl (2-methoxyphenoxy)malonate are added dropwise within 30 minutes to a solution of 27.60 g of sodium in 400 ml of MeOH. Thereafter, 74.86 g of the amidine hydro- chloride obtained in b) are added all at once. The mixture is stirred at room temperature overnight and evaporated at 50°C under reduced pressure. The residue is treated with 500 ml of ether and filtered off under suction. The filter cake is dissolved in 1000 ml of H2O and treated little by little with 50 ml of CH3COOH. The precipitate is filtered off under suction, washed with 400 ml of H2O and dried at 80°C under reduced pressure. There is thus obtained 5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)- pyrimidine-4,6-diol (or tautomer), melting point above 250°C.

d) A suspension of 1 54.6 g of 5-(2-methoxy-phenoxy)-2- (pyridin-4-yl)-pyrimidine-4,6-diol (or tautomer) in 280 ml of POCI3 is heated at 120°C in an oil bath for 24 hours while stirring vigorously. The reaction mixture changes gradually into a dark brown liquid which is evaporated under reduced pressure and thereafter taken up three times with 500 ml of toluene and evaporated. The residue is dissolved in 1000 ml of CH2CI2, treated with ice and H2O and thereafter adjusted with 3N NaOH until the aqueous phase has pH 8. The organic phase is separated and the aqueous phase is extracted twice with CH2CI2. The combined CH2CI2 extracts are dried with MgSθ4, evaporated to half of the volume, treated with 1000 ml of acetone and the CH2CI2 remaining is distilled off at normal pressure. After standing in a refrigerator for 2 hours the crystals are filtered off under suction and dried at 50°C overnight. There is thus obtained 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)- pyrimidine, melting point 1 78-1 80°C.

e) A solution of 1 7.4 g of 4,6-dichloro-5-(2-methoxy- phenoxy)-2-pyridin-4-yl)-pyrimidine in 100 ml of CH3CN is boiled at reflux for 3 hours with 1 5 ml of a 32% peracetic acid solution, thereafter cooled and stored in a refrigerator overnight. The crystals are filtered off under suction and dried at 50°C under reduced pressure. There is thus obtained 4-[4,6-dichloro- 5-(2-methoxy-phenoxy)-pyrimidin-2-yl]-pyridine 1 -oxide, melting point 189-1 90°C.

f) A solution of 36.4 g of 4-[4,6-dichloro-5-(2-methoxy- phenoxy)-pyrimidin-2-yl]-pyridine 1 -oxide and 52.8 g of p-tert- butylphenyl-sulphonamide potassium in 1 50 ml of abs. DMF is stirred at room temperature for 24 hours. Thereafter, it is poured into a mixture of 1 500 ml of H2O and 1000 ml of ether while stirring mechanically, whereby a precipitate forms. The suspension is adjusted to pH 5 with CH3COOH, suction filtered, the crystals are washed with cold water and thereafter with ether and dried at 50°C. There is thus obtained 4-[4-(4-tert- butyl-phenylsulphonylamino)-6-chloro-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide as a colourless material of melting point 247-249°C.

Example 2

A solution of 78.45 g of 4-[4-(4-tert-butyl-phenyl- sulphonylamino)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide, 122.5 g of trimethylsilyl cyanide, 127.8 g of triethylamine and 1200 ml of CH3CN is boiled at reflux for 20 hours and thereafter evaporated under reduced pressure. The oily residue is taken up in 1000 ml of EtOAc and the solution is washed with CH3COOH:H2θ 9:1 and then with H2O. The EtOAc extracts are dried with Na2SO4. After evaporation of the solvent the residue is taken up in a mixture of CH3CN and CF3COOH (20:1 ), whereby a crystalline precipitate separates. There is thus obtained 4-tert-butyl-N-[2-(2-cyano-pyridin-4- yl)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-pyrimidin-4- yl]-benzenesulphonamide of melting point 176-1 79°C.

Example 3 for analogy only compd is different

A suspension of 50.0 g of 4-tert-butyl-N-[2-(2-cyano- pyridin-4-yl)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-4-yl]-benzenesulphonamide, 46.33 g of NH4CI and 56.47 g of NaN3 in 1600 ml of DMF is heated to 70°C for 24 hours while stirring vigorously. The majority of the solvent is distilled off under reduced pressure, the residue is dissolved in H2O, the solution is extracted four times at pH 6.5 with ether, thereafter treated with CH3COOH to pH = 4.5 and extracted with EtOAc. After working up there is obtained a residue which is treated with ether and filtered off under suction therefrom. There is thus obtained 4-tert-butyl-N-[6-(2-hydroxy-ethoxy)-5-(2- methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-yl]-benzenesulphonamide, melting point 225-227°C.

Example 30 final product

In analogy to Example 3, from 5-isopropyl-pyridine-2- sulphonic acid 2-(2-cyano-pyridin-4-yl)-6-(2-hydroxy-ethoxy)- 5-(2-methoxy-phenoxy)-pyrimidin-4-ylamide there is obtained 5-isopropyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5- (2-methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-ylamide (tezosantan free base) as a white substance of melting point 1 98- 200°C from acetonitrile.

The corresponding disodium salt (tezosantan di sodium salt) is obtained as a white powder from this product using sodium methylate in analogy to Example 5

Example 5 for analogy only, compd is different

A solution of 47.8 g of 2-[6-(4-tert-butyl-phenylsulphonyl- amino)-5-(2-methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin- 4-yl)-pyrimidin-4-yloxy]-ethyl pyridin-2-ylcarbamate in 500 ml of abs. THF is treated dropwise with a cold solution of 2.8 g of sodium in 50 ml of methanol, whereby there forms gradually a solid precipitate which, after stirring at room temperature for 1 hour, is filtered off under suction, dried under greatly reduced pressure at 35°C for 3 days and thereafter at 50°C for 2 days. There is thus obtained the bis-sodium salt, decomposition point above 250°C.

References

  1.  Urbanowicz, W; Sogni, P, Moreau, R, Tazi, K A, Barriere, E, Poirel, O, Martin, A, Guimont, M C, Cazals-Hatem, D, Lebrec, D (2004). “Tezosentan, an endothelin receptor antagonist, limits liver injury in endotoxin challenged cirrhotic rats”Gut (BMJ Publishing Group Ltd & British Society of Gastroenterology) 53 (12): 1844–1849. doi:10.1136/gut.2003.036517PMC 1774327PMID 15542526.
  2.  “Tezosentan does not appear to improve symptoms for patients with acute heart failure”Medical Studies/Trials. news-medical.net. 7 Nov 2007. Retrieved 2007-11-24.

EP0979822A1

4 US2003/100507 A1

5 Drugs Fut 2003,28(8),754

6 WO 1996019459……

7 EP 0897914

8 WO 2011163085

9 WO 2004082637

10 WO 2002074034

11…

4-8-2004
Discovery, modeling, and human pharmacokinetics of N-(2-acetyl-4,6-dimethylphenyl)-3-(3,4-dimethylisoxazol-5-ylsulfamoyl)thiophene-2-carboxamide (TBC3711), a second generation, ETA selective, and orally bioavailable endothelin antagonist.
Journal of medicinal chemistry

12  ..

7-1-1999
RO 610612                            .
Drugs in R&D

13….


3-27-2003
Aqueous pharmaceutical composition comprising Tezosentan
8-16-2000
Carbamoylation process
6-30-2000
METHODS AND COMPOSITIONS FOR TREATMENT OF CELL PROLIFERATIVE DISORDERS METHODS AND COMPOSITIONS FOR TREATMENT OF CELL PROLIFERATIVE DISORDERS
5-17-2000
Methods and compositions for treatment of cell proliferative disorders
Share

VORINOSTAT

 Uncategorized  Comments Off on VORINOSTAT
Jan 242014
 

Vorinostat

Zolinza, SAHA, suberoylanilide hydroxamic acid, Suberanilohydroxamic acid, N-hydroxy-N’-phenyloctanediamide

US patent 5369108, PDT PATENT

For the treatment of cutaneous manifestations in patients with cutaneous T-cell lymphoma who have progressive, persistent or recurrent disease on or following two systemic therapies. Inhibits histone deacetylase I & 3. 

  • CCRIS 8456
  • HSDB 7930
  • M344
  • N-Hydroxy-N’-phenyloctanediamide
  • SAHA
  • SAHA cpd
  • Suberanilohydroxamic acid
  • suberoylanilide hydroxamic acid
  • UNII-58IFB293JI
N-hydroxy-N‘-phenyl-octanediamide
Trade names Zolinza, 100 MG, CAPSULE, ORAL
   ZOLINZA (VORINOSTAT) [Merck Sharp & Dohme Corp.]
MedlinePlus a607050
Licence data US FDA:link
   LAUNCHED 2006 MERCKhttp://www.accessdata.fda.gov/drugsatfda_docs/label/2011/021991s002lbl.pdf
Legal status -only (US)
Routes Oral
Pharmacokinetic data
Protein binding 71%
Metabolism Hepatic glucuronidation andoxidation
CYP system not involved
Half-life 2 hours
Excretion Renal (negligible)
Identifiers
CAS number 149647-78-9 
ATC code L01XX38
 
Chemical data
Formula C14H20N2O3 
Mol. mass 264.32 g/mol

CLINICAL TRIALS..http://clinicaltrials.gov/search/intervention=Vorinostat

 

Vorinostat (rINN) also known as suberanilohydroxamic acid (suberoyl+anilide+hydroxamic acid abbreviated as SAHA) is a member of a larger class of compounds that inhibit histone deacetylases (HDAC). Histone deacetylase inhibitors (HDI) have a broad spectrum of epigenetic activities.

Vorinostat is marketed under the name Zolinza for the treatment of cutaneous T cell lymphoma (CTCL) when the disease persists, gets worse, or comes back during or after treatment with other medicines.[1] The compound was developed by Columbia University chemist, Ronald Breslow.

VORINOSTAT

Vorinostat was the first histone deacetylase inhibitor[2] approved by the U.S. Food and Drug Administration (FDA) for the treatment of CTCL on October 6, 2006. It is manufactured by Patheon, Inc., in MississaugaOntarioCanada, for Merck & Co., Inc.White House Station, New Jersey.[3]

ZOLINZA contains vorinostat, which is described chemically as N-hydroxy-N’-phenyloctanediamide. The empirical formula is C14H20N2O3. The molecular weight is 264.32 and the structural formula is:

 

 

ZOLINZA® (vorinostat) Structural Formula Illustration

 

Vorinostat is a white to light orange powder. It is very slightly soluble in water, slightly soluble in ethanol, isopropanol and acetone, freely soluble in dimethyl sulfoxide and insoluble in methylene chloride. It has no chiral centers and is non-hygroscopic. The differential scanning calorimetry ranged from 161.7 (endotherm) to 163.9°C. The pH of saturated water solutions of vorinostat drug substance was 6.6. The pKa of vorinostat was determined to be 9.2.

Each 100 mg ZOLINZA capsule for oral administration contains 100 mg vorinostat and the following inactive ingredients: microcrystalline cellulose, sodium croscarmellose and magnesium stearate. The capsule shell excipients are titanium dioxide, gelatin and sodium lauryl sulfate.

Vorinostat has been shown to bind to the active site of histone deacetylases and act as a chelator for Zinc ions also found in the active site of histone deacetylases [4] Vorinostat’s inhibition of histone deacetylases results in the accumulation of acetylated histones and acetylated proteins, including transcription factors crucial for the expression of genes needed to induce cell differentiation. [4]
SAHA inhibits class I and class II HDACs at nanomolar concentrations and arrests cell growth in a wide variety of transformed cells in culture at 2.5-5.0 µM. This compound efficiently suppressed MES-SA cell growth at a low dosage (3 µM) already after 24 hours treatment. Decrease of cell survival was even more pronounced after prolonged treatment and reached 9% and 2% after 48 and 72 hours of treatment, respectively. Colony forming capability of MES-SA cells treated with 3 µM vorinostat for 24 and 48 hours was significantly diminished and blocked after 72 hours.

Vorinostat has also been used to treat Sézary syndrome, another type of lymphoma closely related to CTCL.[5]

A recent study suggested that vorinostat also possesses some activity against recurrent glioblastoma multiforme, resulting in a median overall survival of 5.7 months (compared to 4 – 4.4 months in earlier studies).[6] Further brain tumor trials are planned in which vorinostat will be combined with other drugs.

Including vorinostat in treatment of advanced non-small-cell lung cancer (NSCLC) showed improved response rates and increased median progression free survival and overall survival (although the survival improvements were not significant at the P=0.05 level).[7]

It has given encouraging results in a phase II trial for myelodysplastic syndromes in combination with Idarubicin and Cytarabine.[8]

Vorinostat is an interesting target for scientists interested in eradicating HIV from infected persons.[9] Vorinostat was recently shown to have both in vitro and in vivo effects against latently HIV infected T-cells.[10][11]

Vorinostat, represented by structural formula (I) and chemically named as N-hydroxy-N’- phenyl-octanediamide or suberoylanilide hydroxamic acid (SAElA), is a member of a larger class of compounds that inhibit histone deacetylases (HDAC). Histone deacetylase inhibitors (HDI) have a broad spectrum of epigenetic activities and vorinostat is marketed, under the brand name Zolinza®, for the treatment of a type of skin cancer called cutaneous T-cell lymphoma (CTCL). Vorinostat is approved to be used when the disease persists, gets worse, or comes back during or after treatment with other medicines. Vorinostat has also been used to treat Sέzary’s disease and, in addition, possesses some activity against recurrent glioblastoma multiforme.

 

Figure imgf000002_0001

Vorinostat was first described in US patent 5369108, wherein four different synthetic routes for the preparation of vorinostat are disclosed (Schemes 1 to 4).

The single step process illustrated in Scheme 1 involves coupling of the diacid chloride of suberic acid with aniline and hydiOxylamine hydrochloride. However, the yield of this reaction is only 15-30%.

Figure imgf000003_0001

Scheme 1

The multistep process illustrated in Scheme 2 begins with the monomethyl ester of suberic acid, which undergoes conversion to the corresponding acid chloride. Further coupling with aniline gives the methyl ester of suberanilic acid. Hydrolysis of the ester and further coupling with benzyl protected hydroxylamine gives benzyl protected vorinostat which on deprotection gives vorinostat.

HO. (CH2J6 OMe . ,OOMM e

O O

Figure imgf000003_0002
Figure imgf000003_0003
Figure imgf000003_0004

Scheme 2

In addition to the disadvantage of being a five-step process with overall yields reported as 35-65%, this process suffers from further disadvantages such as the use of the expensive monomethyl ester of suberic acid.

Figure imgf000004_0001

Scheme 3

The two step process illustrated in Scheme 3 involves coupling of the diacid chloride of suberic acid with aniline and O-benzyl hydroxylamine and then deprotection. However, the overall yield of this reaction is only 20-35%.

 

Figure imgf000004_0002

Scheme 4

The process illustrated in Scheme 4 is similar to that illustrated in Scheme 3, with the exception that O-trimethylsilyl hydroxylamine was used instead of O-benzyl hydroxylamine. The overall yield of this reaction is reported as 20-33%.

Another process for the preparation of vorinostat has been reported in J. Med. Chem.,

1995, vol. 38(8), pages 1411-1413. The reported process, illustrated in Scheme 5, begins with the conversion of suberic acid to suberanilic acid by a high temperature melt reaction.

Suberanilic acid is further converted to the corresponding methyl ester using Dowex resin and the methyl ester of suberanilic acid thus formed is converted to vorinostat by treatment with hydroxylamine hydrochloride. However, this process employs high temperatures (1900C) in the preparation of vorinostat which adds to the inefficiency and high processing costs on commercial scale. The high temperatures also increase the likelihood of impurities being formed during manufacture and safety concerns. The overall yield reported was a poor 35%.

 

Figure imgf000005_0001

MeOH, Dowex, 22 hours

Figure imgf000005_0002

 

Figure imgf000005_0003

Scheme 5

Another process for the preparation of vorinostat has been reported in OPPI Briefs, 2001, vol. 33(4), pages 391-394. The reported process, illustrated in Scheme 6, involves conversion of suberic acid to suberic anhydride, which on treatment with aniline gives suberanilic acid. Coupling of this suberanilic acid with ethyl chloroformate gives a mixed anhydride which upon treatment with hydroxylamine gives vorinostat in an overall yield of 58%. In the first step, there is competition between the formation of suberic anhydride and the linear anhydride and consequently isolation of pure suberic anhydride from the reaction mixture is very difficult. This process step is also hindered by the formation of process impurities and competitive reactions. In the second step, there is formation of dianilide by reaction of two moles of aniline with the linear anhydride. In the third step, suberanilic acid is an inconvenient by-product as the suberanilic acid is converted to a mixed anhydride with ethyl chloroformate, which is highly unstable and is converted back into suberanilic acid. Consequently, it is very difficult to obtain pure vorinostat from the reaction mixture. Although the reported yield was claimed to be 58%, when repeated a yield of only 38% was obtained.

 

Figure imgf000006_0001

Scheme 6

A further process for the preparation of vorinostat has been reported in J. Med. Chem., 2005, vol. 48(15), pages 5047-5051. The reported process, illustrated in Scheme 7, involves conversion of monomethyl suberate to monomethyl suberanilic acid, followed by coupling with hydroxylamine hydrochloride to afford vorinostat in an overall yield of 79%. However, the process uses the expensive monomethyl ester of suberic acid as starting material.

HOBt, DCC, DMF, RT, 4 hours

Figure imgf000006_0002
Figure imgf000006_0003

 

Figure imgf000006_0004
Processes for the preparation of vorinostat, and its form 1 crystalline polymorph, have been disclosed in patent applications US 2004/0122101 and WO 2006/127319. However, the disclosed processes, comprising the preparation of vorinostat from suberic acid, are a cumbersome three step process comprising the sequential steps of amidation of suberic acid with aniline, esterification of the mono-amide product with methanol, and finally reaction with hydroxylamine hydrochloride and sodium methoxide to afford vorinostat. This process is not very convenient as it involves elevated temperatures, lengthy reaction times and has a low overall yield of around 23%. In addition, the intermediate products and final product are not very pure and require exhaustive purification steps.

…………………….

VORINOSTAT

http://www.google.com/patents/EP2349985A2

A preferred embodiment of the first aspect of the present invention is illustrated in Scheme

 

Figure imgf000016_0001

suberic acid subefanilic acid      NH2OHHCl, CDI

Figure imgf000016_0002

suberoylanilide hydroxamic acid (T)

Scheme 8

Optionally, an activating agent can be used in step (a) and/ or step (b) to afford products with high yields and purity. Preferably, the activating agent is selected from cyanuric chloride, cyanuric fluoride, catecholborane, or a mixture thereof. The activating agent is preferably used in combination with the coupling agent. A preferred embodiment of the process according to the first aspect of the present invention comprises the following steps:

(i) taking a mixture of THF, CDI and DCC;

(ii) adding suberic acid; (iii) adding aniline in THF to the solution from step (ii);

(iv) stirring at 25-30°C;

(v) filtering off the solid dicyclohexyl urea formed in the reaction;

(vi) concentrating the filtrate in vacuo;

(vii) adding a solution of KOH in water; (vϋi) filtering off the solid by-product;

(ix) heating the filtrate;

(x) adding aq. HCl;

(xi) isolating suberanilic acid;

(xii) mixing the suberanilic acid and CDI in DMF; (xiii) adding hydroxylamine hydrochloride as solid to the mixture from step (xii);

(xiv) isolating vorinostat from the mixture obtained in step (xiii);

(xv) adding acetonitrile and aq. ammonia to the vorinostat from step (xiv);

(xvi) heating the mixture;

(xvii) cooling the mixture to 20-27°C; and (xvϋi) isolating pure vorinostat from the mixture obtained in step (xvii).

Preferably, by utilising the same organic solvent in steps (a) and (b), pure vorinostat can be obtained without isolation of any synthetic intermediate^).

A preferred embodiment of the second aspect of the present invention is illustrated in Scheme 9.

Figure imgf000018_0001

suberic acid N-hydtoxy-7-carboxy-heptanamide

Figure imgf000018_0002

Example 1

Stage 1 : Conversion of suberic acid to suberanilic acid

A mixture of CDI (0.5eq) and DCC (0.8eq) in THF (15 vol) was stirred for 1 hour at 25- 3O0C. Suberic acid (leq) and aniline (leq) in THF (1 vol) was added and the mixture stirred for a further 16-20 hours. The solid by-product was removed by filtration and the filtrate was concentrated in vacuo at 5O0C. The solid residue obtained was treated with a solution of KOH (2eq) in water (10 vol) and stirred for 30 minutes at 25-300C and any solid byproduct formed was removed by filtration. The filtrate obtained was heated at 6O0C for 3-4 hours and cooled to 200C before addition of an aqueous solution of HCl (17.5%, 3 vol). The mixture was stirred for 30 minutes and the solid filtered, washed with water (2×5 vol) and dried under vacuum at 60-650C. Molar Yield = 60-65% Purity by HPLC = 99.5%

Stage 2: Conversion of suberanilic acid to crude vorinostat The suberanilic acid (leq) obtained in stage 1 was dissolved in DMF (5 vol) and CDI (2eq) was added at 25-3O0C and maintained for 30 minutes under stirring. Hydroxylamine hydrochloride (4eq) was added and stirring continued for 30 minutes. Water (25 vol) was then added and the mixture stirred for 2 hours. The precipitated solid was filtered, washed with water (2×5 vol) and dried under vacuum at 500C. Molar Yield = 70-75% Purity by HPLC = 99% Stage 3: Purification of crude vorinostat

Aqueous ammonia (2.5 vol) was added to the crude vorinostat (leq) in acetonitrile (15 vol) at 25-30°C. The mixture was then maintained at 55-60°C for 1 hour before being cooled to 20-25°C and being stirred for a further hour. The resulting solid was filtered, washed with acetonitrile (2×0.5 vol) and dried under vacuum at 45-5O0C for 5 hours. Molar Yield = 55-60% Purity by HPLC > 99.8%

Example 2

Stage 1 : Conversion of suberic acid to crude vorinostat

A mixture of CDI (0.5eq) and DCC (0.8eq) in THF (15 vol) was stirred for 1 hour at 25- 30°C. Suberic acid (leq) and hydroxylamine (leq) in THF (1 vol) was added and the mixture stirred for a further 1 hour. Then CDI (0.5eq), DCC (0.8eq) and aniline (leq) were added to the mixture and the mixture was stirred for a further 16-20 hours. The solid byproduct was removed by filtration and the filtrate was concentrated in vacuo at 50°C to obtain crude vorinostat. Molar Yield = 55-60% Purity by HPLC > 95.8%

Stage 2: Purification of crude vorinostat

Aqueous ammonia (2.5 vol) was added to the crude vorinostat (leq) in acetonitrile (15 vol) at 25-3O0C. The mixture was then maintained at 55-600C for 1 hour before being cooled to 20-250C and being stirred for a further hour. The resulting solid was filtered, washed with acetonitrile (2×0.5 vol) and dried under vacuum at 45-500C for 5 hours. Molar Yield = 35-40% Purity by HPLC > 99.8%

…………………………………….

SYNTHESIS

/WO2009098515A1

Scheme V. – –

 

Figure imgf000012_0001

Vorinostat

Suberic acid (l.Oeq) was dissolved in tetrahydrofuran (15vol) and the clear solution was chilled to 0-5°C. Methyl chloro formate (l.leq) and triethylamine (1.1 eq) were added to the solution at the same temperature and the mixture was stirred for 15 minutes. The triethylamine.HCl salt formed was filtered off, then aniline (leq) was added to the reaction mixture at 0-50C and stirring was continued for 15 minutes. Methyl chloroformate (l.leq) and triethylamine (l.leq) were added to the clear solution and stirring was continued for a further 15 minutes at 0-5°C. This chilled reaction mixture was added to a freshly prepared hydroxylamine solution in methanol (*see below) chilled to 0-5°C and stirred for 15 minutes at 0-5°C. The solvent was removed under vacuum at 40°C and the residue obtained was taken in methylene dichloride and the organic solution was washed with water and dried over anhydrous sodium sulfate. Methylene dichloride was removed under vacuum at 40°C and acetonitrile was added to the residue. This mixture was stirred for 15 minutes before the solid was filtered under vacuum and dried under vacuum at 60°C to afford the product as a white solid. Molar yield = 35-41%; HPLC purity = 99.90%.

VORINOSTAT

1H-NMR (DMSO-d6): 1.27 (m, 4H, 2 x -CH2-), 1.53 (m, 4H, 2 x -CH2-), 1.94 (t, J = 7.3 Hz, 2H, -CH2-), 2.29 (t, J = 7.4 Hz, 2H, -CH2-), 7.03 (t, J = 7.35 Hz, IH, aromatic para position), 7.27 (t, J = 7.90 Hz, 2H, aromatic meta position), 7.58 (t, J = 7.65 Hz, 2H, aromatic ortho position), 8.66 (s, IH, -OH, D2O exchangeable), 9.85 (s, IH, amide -NH-, D2O exchangeable), 10.33 (s, IH, -NH-OH, D2O exchangeable).

13C-NMR (DMSO-d6): 25.04 (2C, 2 x -CH2-), 28.43 (2C, 2 x -CH2-), 32.24 (1C, -CH2-), 36.34 (1C, -CH2-), 119.01 (2C, Ar-C), 122.96 (1C, Ar-C), 128.68 (2C, Ar-C), 139.24 (1C, Ar- C, =CNH-), 169.23 (1C, -CO-), 171.50 (1C, -CO-).

*Preparation of hydroxylamine solution:

Potassium hydroxide (l.leq) was added to methanol (8vol) and the solution was chilled to 0-5°C. Similarly hydroxylamine hydrochloride (l.leq) was added to methanol (8vol) and chilled to 0-5°C. The chilled amine solution was added to the chilled alkali solution and stirred for 15 minutes at 0-50C. The white potassium chloride salt was filtered off and the filtrate was used as such.

……………………………………………………..
POLYMORPHS
The present invention is directed to a Form I polymorph of SAHA characterized by an X-ray diffraction pattern substantially similar to that set forth in FIG. 13A. SAHA Form I is also characterized by an X-ray diffraction pattern including characteristic peaks at about at about 9.0, 9.4, 17.5, 19.4, 20.0, 24.0, 24.4, 24.8, 25.0, 28.0, and 43.3 degrees 2θ. SAHA Form I is further characterized by an X-ray diffraction pattern including characteristic peaks at about 9.0, 9.4, 17.5, 19.4, 20.0, 24.0, 24.4, 24.8, 25.0, 28.0, 43.3 degrees 20, and lacking at least one peak at about <8.7, 10.0-10.2, 13.4-14.0, 15.0-15.2, 17.5-19.0, 20.1-20.3, 21.1-21.3, 22.0-22.22, 22.7-23.0, 25.0-25.5, 26.0-26.2, and 27.4-27.6 degrees 2θ.
………………………………………………………

SPECTRAL DATA AND SYNTHESIS

Journal of Medicinal Chemistry, 2011 ,  vol. 54,  13  pg. 4694 – 4720

http://pubs.acs.org/doi/full/10.1021/jm2003552

 http://pubs.acs.org/doi/suppl/10.1021/jm2003552/suppl_file/jm2003552_si_001.pdf

for structures see above link

Suberoylanilide hydroxamic acid (26, SAHA, vorinostat).

Suberic acid monomethyl ester (23) (15.09 g, 80.2 mmol) and DMF (0.10 mL) in anhydrous
DCM (300 mL) was added SOCl2 (34.6 mL, 0.481 mol), and the reaction mixture was refluxed for 3
h. The mixture was then concentrated. Toluene (300 mL) was added to the residue and evaporated
to afford crude acid chloride 24. Crude 24 was dissolved in DCM (240 mL), and followed by
addition of aniline (7.3 mL, 80.2 mmol) and Et3N (16.9 mL, 0.120 mol). The reaction mixture was
stirred for 90 min at room temp. The course of reaction was monitored by TLC (30% EtOAc in
hexanes) and LC–MS. DCM was removed, and ethyl acetate (500 mL) was added to dissolve the
residue. The organic layer was washed with aqueous NaHCO3 (500 mL × 2), 1 N HCl (400 mL × 2),
water, dried (Na2SO4), and evaporated to dryness under reduced pressure. The residue was purified
by vacuum liquid chromatography (silica, 20% EtOAc in hexanes) to afford compound 25as white crystalline solids (20.15 g, 96 %). NaOMe in MeOH solution (5.4 M, 106 mL, 0.573 mol) was added to a solution of compound 25 (10.05 g, 38.2 mmol) and NH2OH·HCl (26.54 g, 0.382 mol) in

dry MeOH (375 mL). The reaction mixture was stirred for 40 min at room temp. The reaction was
quenched by adding of 1 N HCl to pH 7–8. MeOH was removed under reduced pressure and water
(1 L) was added to the residue. The precipitated solid was filtered and washed with water (300 mL)
and EtOAc (150 mL) to afford crude 26 which was further purified by recrystallization. MeOH (200
mL) was added to crude 26 (5 g) and warmed to dissolve all solids. The MeOH solution was filtered,

and deionized water (400 mL) was added to the filtrate, the resulting solution was placed at 4 oC
overnight. Crystals obtained were filtered and washed with deionized water (100 mL) to afford pure
26 (vorinostat, SAHA) as off-white crystals. Overall yield: 80–85% from compound 23. Compound
26,

LC–MS m/z 265.1 ([M + H]+).

1H NMR (DMSO-d6)  10.35 (1H, s), 9.86 (1H, s), 8.68 (1H, s),
7.58 (2H, d, J = 7.6 Hz), 7.28 (2H, t, J = 7.5 Hz), 7.02 (1H, t, J = 7.4 Hz), 2.29 (2H, t, J = 7.4 Hz),
1.94 (2H, t, J = 7.4 Hz), 1.57 (2H, m), 1.49 (2H, m), 1.33 – 1.20 (2H, m); 13C NMR (DMSO-d6) 
171.2, 169.1, 139.3, 128.6, 122.9, 119.0, 36.3, 32.2, 28.4, 28.3, 25.0. Anal. (C10H20N2O3) C, H, N.

 

………………………………………………………….

References

  1.  “ZOLINZA, Merck’s Investigational Medicine for Advanced Cutaneous T-Cell Lymphoma (CTCL), To Receive Priority Review from U.S. Food and Drug Administration” (Press release). Merck & Co. June 7, 2006. Retrieved 2006-10-06.
  2.  HDAC Inhibitors Base (vorinostat)
  3.  “FDA Approves New Drug for Skin Cancer, Zolinza” (Press release). Food and Drug Administration. October 6, 2006. Retrieved 2006-10-06.
  4.  Richon, Victoria. “Cancer biology: mechanism of antitumour action of vorinostat (suberoylanilide hydroxamic acid), a novel histone deacetylase inhibitor”. British Journal of Cancer. Retrieved 3 May 2012.
  5.  Cuneo A, Castoldi. “Mycosis fungoides/Sezary’s syndrome”. Retrieved 2008-02-15.
  6.  “Vorinostat shows anti-cancer activity in recurrent gliomas” (Press release). Mayo Clinic. June 3, 2007. Retrieved 2007-06-03.
  7.  http://www.rtmagazine.com/reuters_article.asp?id=20091209clin013.html Dec 2009. URL dead Jan 2012
  8.  “Zolinza, Idarubicin, Cytarabine Combination Yields High Response Rates In MDS Patients (ASH 2011)”.
  9.  “Study of the Effect of Vorinostat on HIV RNA Expression in the Resting CD4+ T Cells of HIV+ Pts on Stable ART”ClinicalTrials.gov. 2011-03-21.
  10.  Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, Margolis DM (2009). “Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid.”AIDS Res Hum Retroviruses 25 (2): 207–12. doi:10.1089/aid.2008.0191PMC 2853863PMID 19239360.
  11.  Contreras X, Schweneker M, Chen CS, McCune JM, Deeks SG, Martin J et al. (2009). “Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells.”J Biol Chem 284 (11): 6782–9.doi:10.1074/jbc.M807898200PMC 2652322PMID 19136668.
  12. Vorinostat bound to proteins in the PDB
  13. J. Med. Chem.,1995, vol. 38(8), pages 1411-1413.
  14. A new simple and high-yield synthesis of suberoylanilide hydroxamic acid and its inhibitory effect alone or in combination with retinoids on proliferation of human prostate cancer cells
    J Med Chem 2005, 48(15): 5047
  15. A new facile and expeditious synthesis of N-hydroxy-N’-phenyloctanediamide, a potent inducer of terminal cytodifferentiation
    Org Prep Proced Int 2001, 33(4): 391
  16. US patent 5369108, PDT PATENT
  17. WO2007/22408………
  18. WO 1993007148
  19. CN 102344392
United States 7456219     APPROVAL    2006-11-14 EXPIRY 2026-11-14
United States 6087367                        1994-10-04             2011-10-04
Canada 2120619                        2006-11-21             2012-10-05
Patent Patent Expiry pat use code
7399787 Feb 9, 2025 U-892
7456219 Mar 11, 2027
7652069 Mar 4, 2023
7732490 Mar 4, 2023 U-892
7851509 Feb 21, 2024 U-892
8067472 Mar 4, 2023 U-892
8093295 May 16, 2026
8101663 Mar 4, 2023 U-892
RE38506 Nov 29, 2013

U 892 =TREATMENT OF CUTANEOUS MANIFESTATIONS IN PATIENTS WTIH CUTANEOUS T-CELL LYMPHOMA (CTCL)

Exclusivity Code Exclusivity_Date
ODE Oct 6, 2013

 

WO2009098515A1 * Feb 6, 2009 Aug 13, 2009 Generics Uk Ltd Novel process for the preparation of vorinostat

Marks, P.A., Breslow, R. Dimethyl sulfoxide to vorinostat: Development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotech 25(1) 84-90 (2007). DOI: 10.1038/nbt1272
Takashi Kumagai, et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (Vorinostat, SAHA) profoundly inhibits the growth of human pancreatic cancer cells. International Journal of Cancer. 2007 Aug 1;121(3):656-65. DOI: 10.1002/ijc.22558
Hrzenjak A, et al. Histone deacetylase inhibitor vorinostat suppresses the growth of uterine sarcomas in vitro and in vivo. Mol Cancer. 2010 Mar 4;9:49. DOI: 10.1186/1476-4598-9-49

………………………………………………………………………………………

US7148257 Aug 26, 2003 Dec 12, 2006 Merck Hdac Research, Llc Methods of treating mesothelioma with suberoylanilide hydroxamic acid
US7375137 Mar 28, 2006 May 20, 2008 Merck Hdac Research, Llc Methods of treating cancer with HDAC inhibitors
US7399787 Jul 9, 2003 Jul 15, 2008 Merck Hdac Research, Llc Methods of treating cancer with HDAC inhibitors
US7456219 Jun 19, 2003 Nov 25, 2008 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US7652069 Oct 30, 2007 Jan 26, 2010 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US7732490 Sep 11, 2007 Jun 8, 2010 Merck Hdac Research, Llc Methods of treating cancer
US7847122 Mar 18, 2008 Dec 7, 2010 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US7851509 Mar 18, 2008 Dec 14, 2010 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US7879865 Nov 18, 2005 Feb 1, 2011 Sloan-Kettering Institute For Cancer Research Treatment of cancer of the brain using histone deacetylase inhibitors
US7998957 Feb 6, 2008 Aug 16, 2011 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicylcoheptenes, their preparation and use
US8058268 Jul 29, 2009 Nov 15, 2011 Lixte Biotechnology, Inc. Neuroprotective agents for the prevention and treatment of neurodegenerative diseases
US8067472 Apr 23, 2010 Nov 29, 2011 Merck Hdac Research, Llc Methods of treating Hodgkin’s and non-Hodgkin’s lymphoma
US8088951 Nov 30, 2007 Jan 3, 2012 Massachusetts Institute Of Technology Epigenetic mechanisms re-establish access to long-term memory after neuronal loss
US8093295 May 16, 2006 Jan 10, 2012 Merck Sharp & Dohme Corp. Formulations of suberoylanilide hydroxamic acid and methods for producing the same
US8101663 Dec 7, 2009 Jan 24, 2012 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US8143445 Oct 1, 2008 Mar 27, 2012 Lixte Biotechnology, Inc. HDAC inhibitors
US8227473 Jul 17, 2009 Jul 24, 2012 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
US8288440 * Jan 13, 2010 Oct 16, 2012 Merck Sharp & Dohme Corp. Formulations of suberoylanilide hydroxamic acid and methods for producing same
US8329719 Aug 1, 2011 Dec 11, 2012 Lixte Biotechnology, Inc. Neuroprotective agents for the prevention and treatment of neurodegenerative diseases
US8426444 Jun 30, 2011 Apr 23, 2013 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
US8450372 * Jan 13, 2010 May 28, 2013 Merck Sharp & Dohme Corp. Formulations of suberoylanilide hydroxamic acid and methods for producing same
US8455688 Mar 21, 2012 Jun 4, 2013 Lixte Biotechnology, Inc. HDAC inhibitors
US8541458 Jun 11, 2012 Sep 24, 2013 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
US8563615 Nov 1, 2010 Oct 22, 2013 Massachusetts Institute Of Technology Use of CI-994 and dinaline for the treatment of memory/cognition and anxiety disorders
US20100112046 * Jan 13, 2010 May 6, 2010 Jeannie Chow Wong Formulations of suberoylanilide hydroxamic acid and methods for producing same
US20100113829 * Jan 13, 2010 May 6, 2010 Cote Aaron S Formulations of suberoylanilide hydroxamic acid and methods for producing same
US20100119596 * Jan 13, 2010 May 13, 2010 Jeannie Chow Wong Formulations of suberoylanilide hydroxamic acid and methods for producing same
US20110263712 * Oct 14, 2009 Oct 27, 2011 Generics (Uk) Limited Process for the preparation of vorinostat
US20110313044 * Jun 16, 2011 Dec 22, 2011 Urquima S.A. Polymorphs of Suberoylanilide Hydroxamic Acid
EP2079304A1 * Sep 24, 2007 Jul 22, 2009 Merck &amp; Co., Inc. Amine base salts of saha and polymorphs thereof
EP2229941A1 * May 16, 2006 Sep 22, 2010 Merck Sharp & Dohme Corp. Formulations of suberoylanilide hydroxamic acid and methods for producing same
EP2292221A2 * May 16, 2006 Mar 9, 2011 Merck Sharp & Dohme Corp. Formulations of suberoylanilide hydroxamic acid and methods for producing same
WO2006127319A2 * May 16, 2006 Nov 30, 2006 Merck & Co Inc Formulations of suberoylanilide hydroxamic acid and methods for producing same
WO2006127321A2 * May 16, 2006 Nov 30, 2006 Merck & Co Inc Formulations of suberoylanilide hydroxamic acid and methods for producing same
WO2008039421A2 * Sep 24, 2007 Apr 3, 2008 Arlene E Mckeown Pharmaceutical compositions of hdac inhibitors and chelatable metal compounds, and metal-hdac inhibitor chelate complexes
WO2008042146A1 * Sep 24, 2007 Apr 10, 2008 Arlene E Mckeown Amine base salts of saha and polymorphs thereof
WO2008097654A1 * Feb 8, 2008 Aug 14, 2008 Nancie M Archin Methods of using saha for treating hiv infection
WO2009020565A1 * Aug 1, 2008 Feb 12, 2009 Lixte Biotechnology Inc Use of phosphatases to treat neuroblastomas and medulloblastomas
WO2010061220A2 * Nov 25, 2009 Jun 3, 2010 Generics [Uk] Limited Novel processes and pure polymorphs

 

EXTRAS

MS-275 (Entinostat)CI-994 (Tacedinaline)BML-210M344MGCD0103 (Mocetinostat)PXD101 (Belinostat)LBH-589 (Panobinostat)Tubastatin AScriptaidNSC 3852NCH 51HNHABML-281CBHASalermidePimelic DiphenylamideITF2357 (Givinostat)PCI-24781APHA Compound 8DroxinostatSB939.

Share

Panobinostat

 orphan status, Phase 3 drug, Uncategorized  Comments Off on Panobinostat
Jan 232014
 

 

Panobinostat

HDAC inhibitors, orphan drug

cas 404950-80-7 

2E)-N-hydroxy-3-[4-({[2-(2-methyl-1H-indol-3-yl)ethyl]amino}methyl)phenyl]acrylamide

N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide (alternatively, N-hydroxy-3-(4-{[2-(2-methyl-1H-indol-3-yl)-ethylamino]-methyl}-phenyl)-acrylamide)

Molecular Formula: C21H23N3O2   Molecular Weight: 349.42622

  • Faridak
  • LBH 589
  • LBH589
  • Panobinostat
  • UNII-9647FM7Y3Z

A hydroxamic acid analog histone deacetylase inhibitor from Novartis.

NOVARTIS, innovator

Histone deacetylase inhibitors

Is currently being examined in cutaneous T-cell lymphoma, CML and breast cancer.

clinical trials click here  phase 3

DRUG SUBSTANCE–LACTATE AS IN  http://www.google.com/patents/US7989639  SEE EG 31

Panobinostat (LBH-589) is an experimental drug developed by Novartis for the treatment of various cancers. It is a hydroxamic acid[1] and acts as a non-selective histone deacetylase inhibitor (HDAC inhibitor).[2]

panobinostat

Panobinostat is a cinnamic hydroxamic acid analogue with potential antineoplastic activity. Panobinostat selectively inhibits histone deacetylase (HDAC), inducing hyperacetylation of core histone proteins, which may result in modulation of cell cycle protein expression, cell cycle arrest in the G2/M phase and apoptosis. In addition, this agent appears to modulate the expression of angiogenesis-related genes, such as hypoxia-inducible factor-1alpha (HIF-1a) and vascular endothelial growth factor (VEGF), thus impairing endothelial cell chemotaxis and invasion. HDAC is an enzyme that deacetylates chromatin histone proteins. Check for

As of August 2012, it is being tested against Hodgkin’s Lymphomacutaneous T cell lymphoma (CTCL)[3] and other types of malignant disease in Phase III clinical trials, against myelodysplastic syndromesbreast cancer and prostate cancer in Phase II trials, and against chronic myelomonocytic leukemia (CMML) in a Phase I trial.[4][5]

Panobinostat is a histone deacetylase (HDAC) inhibitor which was filed for approval in the U.S. in 2010 for the oral treatment of relapsed/refractory classical Hodgkin’s lymphoma in adult patients. The company is conducting phase II/III clinical trials for the oral treatment of multiple myeloma, chronic myeloid leukemia and myelodysplasia. Phase II trials are also in progress for the treatment of primary myelofibrosis, post-polycythemia Vera, post-essential thrombocytopenia, Waldenstrom’s macroglobulinemia, recurrent glioblastoma (GBM) and for the treatment of pancreatic cancer progressing on gemcitabine therapy. Additional trials are under way for the treatment of hematological neoplasms, prostate cancer, colorectal cancer, renal cell carcinoma, non-small cell lung cancer (NSCLC), malignant mesothelioma, acute lymphoblastic leukemia, acute myeloid leukemia, head and neck cancer and gastrointestinal neuroendocrine tumors. Early clinical studies are also ongoing for the treatment of HER2 positive metastatic breast cancer. Additionally, phase II clinical trials are ongoing at Novartis as well as Neurological Surgery for the treatment of recurrent malignant gliomas as are phase I/II initiated for the treatment of acute graft versus host disease. The National Cancer Institute had been conducting early clinical trials for the treatment of metastatic hepatocellular carcinoma; however, these trials were terminated due to observed dose-limiting toxicity. In 2009, Novartis terminated its program to develop panobinostat for the treatment of cutaneous T-cell lymphoma. A program for the treatment of small cell lung cancer was terminated in 2012. Phase I clinical trials are ongoing for the treatment of metastatic and/or malignant melanoma and for the treatment of sickle cell anemia. The University of Virginia is conducting phase I clinical trials for the treatment of newly diagnosed and recurrent chordoma in combination with imatinib. Novartis is evaluating panobinostat for its potential to re-activate HIV transcription in latently infected CD4+ T-cells among HIV-infected patients on stable antiretroviral therapy.

Mechanistic evaluations revealed that panobinostat-mediated tumor suppression involved blocking cell-cycle progression and gene transcription induced by the interleukin IL-2 promoter, accompanied by an upregulation of p21, p53 and p57, and subsequent cell death resulted from the stimulation of caspase-dependent and -independent apoptotic pathways and an increase in the mitochondrial outer membrane permeability. In 2007, the compound received orphan drug designation in the U.S. for the treatment of cutaneous T-cell lymphoma and in 2009 and 2010, orphan drug designation was received in the U.S. and the E.U., respectively, for the treatment of Hodgkin’s lymphoma. This designation was also assigned in 2012 in the U.S. and the E.U. for the treatment of multiple myeloma.

Cardiovascular disease is the leading cause of morbidity and mortality in the western world and during the last decades it has also become a rapidly increasing problem in developing countries. An estimated 80 million American adults (one in three) have one or more expressions of cardiovascular disease (CVD) such as hypertension, coronary heart disease, heart failure, or stroke. Mortality data show that CVD was the underlying cause of death in 35% of all deaths in 2005 in the United States, with the majority related to myocardial infarction, stroke, or complications thereof. The vast majority of patients suffering acute cardiovascular events have prior exposure to at least one major risk factor such as cigarette smoking, abnormal blood lipid levels, hypertension, diabetes, abdominal obesity, and low-grade inflammation.

Pathophysiologically, the major events of myocardial infarction and ischemic stroke are caused by a sudden arrest of nutritive blood supply due to a blood clot formation within the lumen of the arterial blood vessel. In most cases, formation of the thrombus is precipitated by rupture of a vulnerable atherosclerotic plaque, which exposes chemical agents that activate platelets and the plasma coagulation system. The activated platelets form a platelet plug that is armed by coagulation-generated fibrin to form a biood clot that expands within the vessel lumen until it obstructs or blocks blood flow, which results in hypoxic tissue damage (so-called infarction). Thus, thrombotic cardiovascular events occur as a result of two distinct processes, i.e. a slowly progressing long-term vascular atherosclerosis of the vessel wall, on the one hand, and a sudden acute clot formation that rapidly causes flow arrest, on the other. This invention solely relates to the latter process.

Recently, inflammation has been recognized as an important risk factor for thrombotic events. Vascular inflammation is a characteristic feature of the atherosclerotic vessel wall, and inflammatory activity is a strong determinant of the susceptibility of the atherosclerotic plaque to rupture and initiate intravascular clotting. Also, autoimmune conditions with systemic inflammation, such as rheumatoid arthritis, systemic lupus erythematosus and different forms of vasculitides, markedly increase the risk of myocardial infarction and stroke.

Traditional approaches to prevent and treat cardiovascular events are either targeted 1) to slow down the progression of the underlying atherosclerotic process, 2) to prevent clot formation in case of a plaque rupture, or 3) to direct removal of an acute thrombotic flow obstruction. In brief, antiatherosclerotic treatment aims at modulating the impact of general risk factors and includes dietary recommendations, weight loss, physical exercise, smoking cessation, cholesterol- and blood pressure treatment etc. Prevention of clot formation mainly relies on the use of antiplatelet drugs that inhibit platelet activation and/or aggregation, but also in some cases includes thromboembolic prevention with oral anticoagulants such as warfarin. Post-hoc treatment of acute atherothrombotic events requires either direct pharmacological lysis of the clot by thrombolytic agents such as recombinant tissue-type plasminogen activator or percutaneous mechanical dilation of the obstructed vessel.

Despite the fact that multiple-target antiatherosclerotic therapy and clot prevention by antiplatelet agents have lowered the incidence of myocardial infarction and ischemic stroke, such events still remain a major population health problem. This shows that in patients with cardiovascular risk factors these prophylactic measures are insufficient to completely prevent the occurrence of atherothrombotic events.

Likewise, thrombotic conditions on the venous side of the circulation, as well as embolic complications thereof such as pulmonary embolism, still cause substantial morbidity and mortality. Venous thrombosis has a different clinical presentation and the relative importance of platelet activation versus plasma coagulation are somewhat different with an preponderance for the latter in venous thrombosis, However, despite these differences, the major underlying mechanisms that cause thrombotic vessel occlusions are similar to those operating on the arterial circulation. Although unrelated to atherosclerosis as such, the risk of venous thrombosis is related to general cardiovascular risk factors such as inflammation and metabolic aberrations.

Panobinostat can be synthesized as follows: Reduction of 2-methylindole-3-glyoxylamide (I) with LiAlH4 affords 2-methyltryptamine (II). 4-Formylcinnamic acid (III) is esterified with methanolic HCl, and the resulting aldehyde ester (IV) is reductively aminated with 2-methyltryptamine (II) in the presence of NaBH3CN (1) or NaBH4 (2) to give (V). The title hydroxamic acid is then obtained by treatment of ester (V) with aqueous hydroxylamine under basic conditions.

Panobinostat is currently being used in a Phase I/II clinical trial that aims at curing AIDS in patients on highly active antiretroviral therapy (HAART). In this technique panobinostat is used to drive the HI virus’s DNA out of the patient’s DNA, in the expectation that the patient’s immune system in combination with HAART will destroy it.[6][7]

panobinostat

Panobinostat has been found to synergistically act with sirolimus to kill pancreatic cancer cells in the laboratory in a Mayo Clinic study. In the study, investigators found that this combination destroyed up to 65 percent of cultured pancreatic tumor cells. The finding is significant because the three cell lines studied were all resistant to the effects of chemotherapy – as are many pancreatic tumors.[8]

Panobinostat has also been found to significantly increase in vitro the survival of motor neuron (SMN) protein levels in cells of patients suffering fromspinal muscular atrophy.[9]

Panobinostat was able to selectively target triple negative breast cancer (TNBC) cells by inducing hyperacetylation and cell cycle arrest at the G2-M DNA damage checkpoint; partially reversing the morphological changes characteristic of breast cancer cells.[10]

Panobinostat, along with other HDAC inhibitors, is also being studied for potential to induce virus HIV-1 expression in latently infected cells and disrupt latency. These resting cells are not recognized by the immune system as harboring the virus and do not respond to antiretroviral drugs.[11]

Panobinostat inhibits multiple histone deacetylase enzymes, a mechanism leading to apoptosis of malignant cells via multiple pathways.[1]

The compound N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide (alternatively, N-hydroxy-3-(4-{[2-(2-methyl-1H-indol-3-yl)-ethylamino]-methyl}-phenyl)-acrylamide) has the formula

 

Figure US07989639-20110802-C00001

 

as described in WO 02/22577. Valuable pharmacological properties are attributed to this compound; thus, it can be used, for example, as a histone deacetylase inhibitor useful in therapy for diseases which respond to inhibition of histone deacetylase activity. WO 02/22577 does not disclose any specific salts or salt hydrates or solvates of N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide.

The compounds described above are often used in the form of a pharmaceutically acceptable salt. Pharmaceutically acceptable salts include, when appropriate, pharmaceutically acceptable base addition salts and acid addition salts, for example, metal salts, such as alkali and alkaline earth metal salts, ammonium salts, organic amine addition salts, and amino acid addition salts, and sulfonate salts. Acid addition salts include inorganic acid addition salts such as hydrochloride, sulfate and phosphate, and organic acid addition salts such as alkyl sulfonate, arylsulfonate, acetate, maleate, fumarate, tartrate, citrate and lactate. Examples of metal salts are alkali metal salts, such as lithium salt, sodium salt and potassium salt, alkaline earth metal salts such as magnesium salt and calcium salt, aluminum salt, and zinc salt. Examples of ammonium salts are ammonium salt and tetramethylammonium salt. Examples of organic amine addition salts are salts with morpholine and piperidine. Examples of amino acid addition salts are salts with glycine, phenylalanine, glutamic acid and lysine. Sulfonate salts include mesylate, tosylate and benzene sulfonic acid salts.

……………………………..

GENERAL METHOD OF SYNTHESIS

ADD YOUR METHYL AT RIGHT PLACE

WO2002022577A2

 

As is evident to those skilled in the art, the many of the deacetylase inhibitor compounds of the present invention contain asymmetric carbon atoms. It should be understood, therefore, that the individual stereoisomers are contemplated as being included within the scope of this invention.

The hydroxamate compounds of the present invention can be produced by known organic synthesis methods. For example, the hydroxamate compounds can be produced by reacting methyl 4-formyl cinnamate with tryptamine and then converting the reactant to the hydroxamate compounds. As an example, methyl 4-formyl cinnamate 2, is prepared by acid catalyzed esterification of 4-formylcinnamic acid 3 (Bull. Chem. Soc. Jpn. 1995; 68:2355-2362). An alternate preparation of methyl 4-formyl cinnamate 2 is by a Pd- catalyzed coupling of methyl acrylate 4 with 4-bromobenzaldehyde 5.

CHO

 

Figure imgf000020_0001

Additional starting materials can be prepared from 4-carboxybenzaldehyde 6, and an exemplary method is illustrated for the preparation of aldehyde 9, shown below. The carboxylic acid in 4-carboxybenzaldehyde 6 can be protected as a silyl ester (e.g., the t- butyldimethylsilyl ester) by treatment with a silyl chloride (e.g., f-butyldimethylsilyl chloride) and a base (e.g. triethylamine) in an appropriate solvent (e.g., dichloromethane). The resulting silyl ester 7 can undergo an olefination reaction (e.g., a Horner-Emmons olefination) with a phosphonate ester (e.g., triethyl 2-phosphonopropionate) in the presence of a base (e.g., sodium hydride) in an appropriate solvent (e.g., tetrahydrofuran (THF)). Treatment of the resulting diester with acid (e.g., aqueous hydrochloric acid) results in the hydrolysis of the silyl ester providing acid 8. Selective reduction of the carboxylic acid of 8 using, for example, borane-dimethylsuflide complex in a solvent (e.g., THF) provides an intermediate alcohol. This intermediate alcohol could be oxidized to aldehyde 9 by a number of known methods, including, but not limited to, Swern oxidation, Dess-Martin periodinane oxidation, Moffatt oxidation and the like.

 

Figure imgf000020_0002

The aldehyde starting materials 2 or 9 can be reductively aminated to provide secondary or tertiary amines. This is illustrated by the reaction of methyl 4-formyl cinnamate 2 with tryptamine 10 using sodium triacetoxyborohydride (NaBH(OAc)3) as the reducing agent in dichloroethane (DCE) as solvent to provide amine 11. Other reducing agents can be used, e.g., sodium borohydride (NaBH ) and sodium cyanoborohydride (NaBH3CN), in other solvents or solvent mixtures in the presence or absence of acid catalysts (e.g., acetic acid and trifluoroacetic acid). Amine 11 can be converted directly to hydroxamic acid 12 by treatment with 50% aqueous hydroxylamine in a suitable solvent (e.g., THF in the presence of a base, e.g., NaOH). Other methods of hydroxamate formation are known and include reaction of an ester with hydroxylamine hydrochloride and a base (e.g., sodium hydroxide or sodium methoxide) in a suitable solvent or solvent mixture (e.g., methanol, ethanol or methanol/THF).

 

Figure imgf000021_0001

 

NOTE ….METHYL SUBSTITUENT ON 10 WILL GIVE YOU PANOBINOSTAT

……………………………….

Journal of Medicinal Chemistry, 2011 ,  vol. 54,  13  pg. 4694 – 4720

(E)-N-Hydroxy-3-(4-{[2-(2-methyl-1H-indol-3-yl)-ethylamino]-methyl}-phenyl)-acrylamide
lactate

(34, panobinostat, LBH589)

http://pubs.acs.org/doi/full/10.1021/jm2003552

 http://pubs.acs.org/doi/suppl/10.1021/jm2003552/suppl_file/jm2003552_si_001.pdf

for str see above link

α-methyl-β-(β-bromoethyl)indole (29) was made according to method reported by Grandberg et al.(2. Grandberg, I. I.; Kost, A. N.; Terent’ev, A. P. Reactions of hydrazine derivatives. XVII. New synthesis of α-methyltryptophol. Zhurnal Obshchei Khimii 1957, 27, 3342–3345. )

The bromide 29 was converted to amine 30 by using similar method used by Sletzinger et al.(3. Sletzinger, M.; Ruyle, W. V.; Waiter, A. G. (Merck & Co., Inc.). Preparation of tryptamine
derivatives. U.S. Patent US 2,995,566, Aug 8, 1961.)

To a 500 mL flask, crude 2-methyltryptamine 30 (HPLC purity 75%, 1.74 g, 7.29 mmol) and 3-(4-
formyl-phenyl)-acrylic acid methyl ester 31 (HPLC purity 84%, 1.65 g, 7.28 mmol) were added,
followed by DCM (100 mL) and MeOH (30 mL). The clear solution was stirred at room temp for 30
min, then NaBH3CN (0.439 g, 6.99 mmol) was added in small portions. The reaction mixture was
stirred at room temp overnight. After removal of the solvents, the residue was diluted with DCM and
added saturated NaHCO3 aqueous solution, extracted with DCM twice. The DCM layer was dried
and concentrated, and the resulting residue was purified by flash chromatography (silica, 0–10%
MeOH in DCM) to afford 33 as orange solid (1.52 g, 60%). LC–MS m/z 349.2 ([M + H]+). 33 was
converted to hydroxamic acid 34 according to procedure D (Experimental Section), and the freebase
34 was treated with 1 equiv of lactic acid in MeOH–water (7:3) to form lactic acid salt which was
further recrystallized in MeOH–EtOAc to afford the lactic acid salt of 34as pale yellow solid. LC–MS m/z 350.2 ([M + H − lactate]+).

= DELTA

1H NMR (DMSO-d6)  10.72 (s, 1H, NH), 7.54 (d, J = 8.0 Hz, 2H), 7.44 (d, J = 16 Hz, 1H), 7.43 (d, J = 7.8 Hz, 2H), 7.38 (d, J = 7.6 Hz, 1H), 7.22 (d, J = 7.8 Hz, 1H), 6.97 (td, J = 7.8 Hz, 1H), 7.44 (d, J = 15.8 Hz, 1H), 7.22 (t, J = 7.8 Hz, 2H), 7.08 (d, J = 7.8Hz, 2H), 7.01 (t, J = 7.4, 0.9 Hz, 1H), 6.91 (td, J = 7.4, 0.9 Hz, 1H), 6.47 (d, J = 15.2 Hz, 1H), 3.94(q, J = 6.8 Hz, 1H, lactate CH), 3.92 (s, 2H), 2.88 and 2.81 (m, each, 4H, AB system, CH2CH2),2.31 (s, 3H), 1.21 (d, J = 6.8 Hz, 3H).;

13C NMR (DMSO-d6)  176.7 (lactate C=O), 162.7, 139.0,
137.9, 135.2, 134.0, 132.1, 129.1, 128.1, 127.4, 119.9, 119.0, 118.1, 117.2, 110.4, 107.0, 66.0, 51.3,
48.5, 22.9, 20.7, 11.2.

…………………………………………..

PANOBINOSTAT DRUG SUBSTANCE SYNTHESIS AND DATA

http://www.google.com/patents/US7989639

Figure US07989639-20110802-C00002

 

A flow diagram for the synthesis of LBH589 lactate is provided in FIG. A. A nomenclature reference index of the intermediates is provided below in the Nomenclature Reference Index:

 

Nomenclature reference index
Compound Chemical name
1 4-Bromo-benzaldehyde
2 Methyl acrylate
3 (2E)-3-(formylphenyl)-2-propenoic acid, methyl ester
4 3-[4-[[[2-(2-Methyl-1H-indol-3-
yl)ethyl]amino]methyl]phenyl]-2-
propenoic acid, methyl ester, monohydrochloride
5 (2E)-N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-
yl)ethyl]amino]methyl]phenyl]-2-propenamide
6 2-hydroxypropanoic acid, compd. with 2(E)-N-
hydroxy-3-[4-[[[2-(2-methyl-1H-
indol-3-yl)ethyl]amino]methyl]phenyl]-2-propenamide
Z3a 2-Methyl-1H-indole-3-ethanamine
Z3b 5-Chloro-2-pentanone
Z3c Phenylhydrazine

The manufacture of LBH589 lactate (6) drug substance is via a convergent synthesis; the point of convergence is the condensation of indole-amine Z3a with aldehyde 3.

The synthesis of indole-amine Z3a involves reaction of 5-chloro-2 pentanone (Z3b) with phenylhydrazine (Z3c) in ethanol at reflux (variation of Fischer indole synthesis).

Product isolation is by an extractive work-up followed by crystallization. Preparation of aldehyde 3 is by palladium catalyzed vinylation (Heck-type reaction; Pd(OAc)2/P(o-Tol)3/Bu3N in refluxing CH3CN) of 4-bromo-benzyladehyde (1) with methyl acrylate (2) with product isolation via precipitation from dilute HCl solution. Intermediates Z3a and 3 are then condensed to an imine intermediate, which is reduced using sodium borohydride in methanol below 0° C. (reductive amination). The product indole-ester 4, isolated by precipitation from dilute HCl, is recrystallized from methanol/water, if necessary. The indole ester 4 is converted to crude LBH589 free base 5 via reaction with hydroxylamine and sodium hydroxide in water/methanol below 0° C. The crude LBH589 free base 5 is then purified by recrystallization from hot ethanol/water, if necessary. LBH589 free base 5 is treated with 85% aqueous racemic lactic acid and water at ambient temperature. After seeding, the mixture is heated to approximately 65° C., stirred at this temperature and slowly cooled to 45-50° C. The resulting slurry is filtered and washed with water and dried to afford LBH589 lactate (6).

If necessary the LBH589 lactate 6 may be recrystallised once again from water in the presence of 30 mol % racemic lactic acid. Finally the LBH589 lactate is delumped to give the drug substance. If a rework of the LBH589 lactate drug substance 6 is required, the LBH589 lactate salt is treated with sodium hydroxide in ethanol/water to liberate the LBH589 free base 5 followed by lactate salt formation and delumping as described above.

All starting materials, reagents and solvents used in the synthesis of LBH589 lactate are tested according to internal specifications or are purchased from established suppliers against a certificate of analysis.

 

EXAMPLE 7 Formation of Monohydrate Lactate Salt

About 40 to 50 mg of N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide free base was suspended in 1 ml of a solvent as listed in Table 7. A stoichiometric amount of lactic acid was subsequently added to the suspension. The mixture was stirred at ambient temperature and when a clear solution formed, stirring continued at 4° C. Solids were collected by filtration and analyzed by XRPD, TGA and 1H-NMR.

 

TABLE 7
LOD, %
Physical Crystallinity (Tdesolvation)
Solvent T, ° C. Appear. and Form Tdecomposit. 1H-NMR
IPA 4 FFP excellent 4.3 (79.3)
HA 156.3
Acetone 4 FFP excellent 4.5 (77.8) 4.18 (Hbz)
HA 149.5

 

The salt forming reaction in isopropyl alcohol and acetone at 4° C. produced a stoichiometric (1:1) lactate salt, a monohydrate. The salt is crystalline, begins to dehydrate above 77° C., and decomposes above 150° C.

EXAMPLE 18 Formation of Anhydrous Lactate Salt

DL-lactic acid (4.0 g, 85% solution in water, corresponding to 3.4 g pure DL-lactic acid) is diluted with water (27.2 g), and the solution is heated to 90° C. (inner temperature) for 15 hours. The solution is allowed to cool down to room temperature and is used as lactic acid solution for the following salt formation step.

N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide free base (10.0 g) is placed in a 4-necked reaction flask with mechanical stirrer. Demineralized water (110.5 g) is added, and the suspension is heated to 65° C. (inner temperature) within 30 minutes. The DL-lactic acid solution is added to this suspension during 30 min at 65° C. During the addition of the lactate salt solution, the suspension converted into a solution. The addition funnel is rinsed with demineralized water (9.1 g), and the solution is stirred at 65° C. for an additional 30 minutes. The solution is cooled down to 45° C. (inner temperature) and seed crystals (10 mg N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate monohydrate) are added at this temperature. The suspension is cooled down to 33° C. and is stirred for additional 20 hours at this temperature. The suspension is re-heated to 65° C., stirred for 1 hour at this temperature and is cooled to 33° C. within 1 hour. After additional stirring for 3 hours at 33° C., the product is isolated by filtration, and the filter cake is washed with demineralized water (2×20 g). The wet filter-cake is dried in vacuo at 50° C. to obtain the anhydrous N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt as a crystalline product. The product is identical to the monohydrate salt (form HA) in HPLC and in 1H-NMR, with the exception of the integrals of water signals in the 1H-NMR spectra.

In additional salt formation experiments carried out according to the procedure described above, the product solution was filtered at 65° C. before cooling to 45° C., seeding and crystallization. In all cases, form A (anhydrate form) was obtained as product.

EXAMPLE 19 Formation of Anhydrous Lactate Salt

DL-lactic acid (2.0 g, 85% solution in water, corresponding to 1.7 g pure DL-lactic acid) is diluted with water (13.6 g), and the solution is heated to 90° C. (inner temperature) for 15 hours. The solution was allowed to cool down to room temperature and is used as lactic acid solution for the following salt formation step.

N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide free base (5.0 g) is placed in a 4-necked reaction flask with mechanical stirrer. Demineralized water (54.85 g) is added, and the suspension is heated to 48° C. (inner temperature) within 30 minutes. The DL-lactic acid solution is added to this suspension during 30 minutes at 48° C. A solution is formed. Seed crystals are added (as a suspension of 5 mg N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt, anhydrate form A, in 0.25 g of water) and stirring is continued for 2 additional hours at 48° C. The temperature is raised to 65° C. (inner temperature) within 30 minutes, and the suspension is stirred for additional 2.5 hours at this temperature. Then the temperature is cooled down to 48° C. within 2 hours, and stirring is continued at this temperature for additional 22 hours. The product is isolated by filtration and the filter cake is washed with demineralized water (2×10 g). The wet filter-cake is dried in vacuo at 50° C. to obtain anhydrous N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt (form A) as a crystalline product.

EXAMPLE 20 Conversion of Monohydrate Lactate Salt to Anhydrous Lactate Salt

DL-lactic acid (0.59 g, 85% solution in water, corresponding to 0.5 g pure DL-lactic acid) is diluted with water (4.1 g), and the solution is heated to 90° C. (inner temperature) for 15 hours. The solution is allowed to cool down to room temperature and is used as lactic acid solution for the following salt formation step.

10 g of N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt monohydrate is placed in a 4-necked reaction flask. Water (110.9 g) is added, followed by the addition of the lactic acid solution. The addition funnel of the lactic acid is rinsed with water (15.65 g). The suspension is heated to 82° C. (inner temperature) to obtain a solution. The solution is stirred for 15 minutes at 82° C. and is hot filtered into another reaction flask to obtain a clear solution. The temperature is cooled down to 50° C., and seed crystals are added (as a suspension of 10 mg N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt, anhydrate form, in 0.5 g of water). The temperature is cooled down to 33° C. and stirring is continued for additional 19 hours at this temperature. The formed suspension is heated again to 65° C. (inner temperature) within 45 minutes, stirred at 65° C. for 1 hour and cooled down to 33° C. within 1 hour. After stirring at 33° C. for additional 3 hours, the product is isolated by filtration and the wet filter cake is washed with water (50 g). The product is dried in vacuo at 50° C. to obtain crystalline anhydrous N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl) ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt (form A).

EXAMPLE 21 Formation of Anhydrous Lactate Salt

DL-lactic acid (8.0 g, 85% solution in water, corresponding to 6.8 g pure DL-lactic acid) was diluted with water (54.4 g), and the solution was heated to 90° C. (inner temperature) for 15 hours. The solution was allowed to cool down to room temperature and was used as lactic acid solution for the following salt formation step.

N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide (20 g) is placed in a 1 L glass reactor, and ethanol/water (209.4 g of a 1:1 w/w mixture) is added. The light yellow suspension is heated to 60° C. (inner temperature) within 30 minutes, and the lactic acid solution is added during 30 minutes at this temperature. The addition funnel is rinsed with water (10 g). The solution is cooled to 38° C. within 2 hours, and seed crystals (20 mg of N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt, anhydrate form) are added at 38° C. After stirring at 38° C. for additional 2 hours, the mixture is cooled down to 25° C. within 6 hours. Cooling is continued from 25° C. to 10° C. within 5 hours, from 10° C. to 5° C. within 4 hours and from 5° C. to 2° C. within 1 hour. The suspension is stirred for additional 2 hours at 2° C., and the product is isolated by filtration. The wet filter cake is washed with water (2×30 g), and the product is dried in vacuo at 45° C. to obtain crystalline anhydrous N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide lactate salt (form A).

EXAMPLE 28 Formation of Lactate Monohydrate Salt

3.67 g (10 mmol) of the free base monohydrate (N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl) ethyl]amino]methyl]phenyl]-2E-2-propenamide) and 75 ml of acetone were charged in a 250 ml 3-neck flask equipped with a magnetic stirrer and an addition funnel. To the stirred suspension were added dropwise 10 ml of 1 M lactic acid in water (10 mmol) dissolved in 20 ml acetone, affording a clear solution. Stirring continued at ambient and a white solid precipitated out after approximately 1 hour. The mixture was cooled in an ice bath and stirred for an additional hour. The white solid was recovered by filtration and washed once with cold acetone (15 ml). It was subsequently dried under vacuum to yield 3.94 g of the lactate monohydrate salt of N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide (86.2%).

 

References

  1. Revill, P; Mealy, N; Serradell, N; Bolos, J; Rosa, E (2007). “Panobinostat”Drugs of the Future 32 (4): 315. doi:10.1358/dof.2007.032.04.1094476ISSN 0377-8282.
  2.  Table 3: Select epigenetic inhibitors in various stages of development from Mack, G. S. (2010). “To selectivity and beyond”. Nature Biotechnology 28 (12): 1259–1266.doi:10.1038/nbt.1724PMID 21139608edit
  3.  ClinicalTrials.gov NCT00425555 Study of Oral LBH589 in Adult Patients With Refractory Cutaneous T-Cell Lymphoma
  4.  ClinicalTrials.gov: LBH-589
  5.  Prince, HM; M Bishton (2009). “Panobinostat (LBH589): a novel pan-deacetylase inhibitor with activity in T cell lymphoma”Hematology Meeting Reports (Parkville, Australia: Peter MacCallum Cancer Centre and University of Melbourne) 3 (1): 33–38.
  6.  Simons, J (27 April 2013). “Scientists on brink of HIV cure”. The Telegraph.
  7.  ClinicalTrials.gov NCT01680094 Safety and Effect of The HDAC Inhibitor Panobinostat on HIV-1 Expression in Patients on Suppressive HAART (CLEAR)
  8.  Mayo Clinic Researchers Formulate Treatment Combination Lethal To Pancreatic Cancer Cells
  9.  Garbes, L; Riessland, M; Hölker, I; Heller, R; Hauke, J; Tränkle, Ch; Coras, R; Blümcke, I; Hahnen, E; Wirth, B (2009). “LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate”Human Molecular Genetics 18 (19): 3645–3658. doi:10.1093/hmg/ddp313.PMID 19584083.
  10.  Tate, CR; Rhodes, LV; Segar, HC; Driver, JL; Pounder, FN; Burow, ME; and Collins-Burow, BM (2012). “Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat”Breast Cancer Research 14 (3).
  11.  TA Rasmussen, et al. Comparison of HDAC inhibitors in clinical development: Effect on HIV production in latently infected cells and T-cell activation. Human Vaccines & Immunotherapeutics 9:5, 1-9, May 2013.
  12. Drugs of the Future 32(4): 315-322 (2007)
  13. WO 2002022577…
  14. WO 2007146718
  15. WO 2013110280
  16. WO 2010009285
  17. WO 2010009280
  18. WO 2005013958
  19. WO 2004103358
  20. WO 2003048774…
  21. Journal of Medicinal Chemistry, 2011 ,  vol. 54,  13  pg. 4694 – 4720
  22. 11-26-2012
    Selective histone deacetylase 6 inhibitors bearing substituted urea linkers inhibit melanoma cell growth.
    Journal of medicinal chemistry
  23. 7-14-2011
    Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
    Journal of medicinal chemistry
  24. 4-28-2011
    Discovery, synthesis, and pharmacological evaluation of spiropiperidine hydroxamic acid based derivatives as structurally novel histone deacetylase (HDAC) inhibitors.
    Journal of medicinal chemistry
  25. 4-23-2009
    Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum.
    Journal of medicinal chemistry
  26. 1-1-2005
    The American Society of Hematology–46th Annual Meeting and Exposition. HDAC, Flt and farnesyl transferase inhibitors.
    IDrugs : the investigational drugs journal
  27. 8-3-2011
    PROCESS FOR MAKING SALTS OF N-HYDROXY-3-[4-[[[2-(2-METHYL-1H-INDOL-3-YL)ETHYL]AMINO]METHYL]PHENYL]-2E-2-PROPENAMIDE
    11-12-2010
    SALTS OF N-HYDROXY-3-[4-[[[2-(2-METHYL-1H-INDOL-3-YL)ETHYL]AMINO]METHYL]PHENYL]-2E-2-PROPENAMIDE
    7-16-2010
    Use of HDAC Inhibitors for the Treatment of Bone Destruction
    6-25-2010
    USE OF HDAC INHIBITORS FOR THE TREATMENT OF MYELOMA
    6-4-2010
    USE OF HDAC INHIBITORS FOR THE TREATMENT OF GASTROINTESTINAL CANCERS
    12-11-2009
    PROCESS FOR MAKING N-HYDROXY-3-[4-[[[2-(2-METHYL-1H-INDOL-3-YL)ETHYL]AMINO]METHYL]PHENYL]-2E-2-PROPENAMIDE AND STARTING MATERIALS THEREFOR
    11-13-2009
    USE OF HDAC INHIBITORS FOR THE TREATMENT OF LYMPHOMAS
    10-23-2009
    Combination of a) N–4-(3-pyridyl)-2-pyrimidine-amine and b) a histone deacetylase inhibitor for the treatment of leukemia
    8-7-2009
    SALTS OF N-HYDROXY-3-[4-[[[2-(2-METHYL-1H-INDOL-3-YL)ETHYL]AMINO]METHYL]PHENYL]-2E-2-PROPENAMIDE
    1-9-2009
    Method of Use of Deacetylase Inhibitors
12-26-2008
Combination of Histone Deacetylase Inhibitors and Radiation
9-12-2008
Use of Hdac Inhibitors for the Treatment of Myeloma
7-25-2008
DEACETYLASE INHIBITORS
8-25-2006
Deacetylase inhibitors
6-28-2006
Deacetylase inhibitors
5-12-2006
Combination of a) n-{5-[4-(4-methyl-piperazino-methyl)-benzoylamido]2-methylphenyl}-4- (3-pyridyl)-2-pyrimidine-amine and b) a histone deacetylase inhibitor for the treatment of leukemia
12-22-2004
Deacetylase inhibitors
4-23-2003
Deacetylase inhibitors
GB776693A Title not available
GB891413A Title not available
GB2185020A Title not available
WO2002022577A2 Aug 30, 2001 Mar 21, 2002 Kenneth Walter Bair Hydroxamate derivatives useful as deacetylase inhibitors
WO2003016307A1 Aug 6, 2002 Aug 19, 1993 Jolie Anne Bastian β3 ADRENERGIC AGONISTS
WO2003039599A1 Nov 5, 2002 May 15, 2003 Ying-Nan Pan Chen Cyclooxygenase-2 inhibitor/histone deacetylase inhibitor combination
WO2005105740A2 Apr 26, 2005 Nov 10, 2005 Serguei Fine Preparation of tegaserod and tegaserod maleate
WO2006021397A1 Aug 22, 2005 Mar 2, 2006 Recordati Ireland Ltd Lercanidipine salts

…………………………………..

extras

5. Mocetinostat (MGCD0103), including pharmaceutically acceptable salts thereof. Balasubramanian et al., Cancer Letters 280: 211-221 (2009).
Mocetinostat, has the following chemical structure and name:

 

Figure US20130266649A1-20131010-C00007
,………………………………

Vorinostat, including pharmaceutically acceptable salts thereof. Marks et al., Nature Biotechnology 25, 84 to 90 (2007); Stenger, Community Oncology 4, 384-386 (2007).
Vorinostat has the following chemical structure and name:

 

Figure US20130266649A1-20131010-C00003
………………………

Belinostat (PXD-101 , PX-105684)

(2E)-3-[3-(anilinosulfonyl)phenyl]-N-hydroxyacrylamide

Figure imgf000014_0001

……………………………………………….

Dacinostat (LAQ-824, NVP-LAQ824,)

((E)-N-hydroxy-3-[4-[[2-hydroxyethyl-[2-(1 H-indol-3-yl)ethyl]amino]methyl]phenyl]prop-2-enamide

 

Figure imgf000014_0002
…………………………………………

Entinostat (MS-275, SNDX-275, MS-27-275)

4-(2-aminophenylcarbamoyl)benzylcarbamate

Figure imgf000015_0001
………………….

(a) The HDAC inhibitor Vorinostat™ or a salt, hydrate, or solvate thereof.

Figure imgf000270_0001

Vorinostat………………..

 

(b) The HDAC inhibitor Givinostat or a salt, hydrate, or solvate thereof.

Figure imgf000270_0002

Givinostat or a salt, hydrate, or solvate thereof.

……………………………………………

…………………………..
Share

BELINOSTAT, FAST TRACK, ORPHAN DRUG, A hydroxamate-type inhibitor of histone deacetylase.

 phase 2, Uncategorized  Comments Off on BELINOSTAT, FAST TRACK, ORPHAN DRUG, A hydroxamate-type inhibitor of histone deacetylase.
Jan 232014
 

File:Belinostat.svg

 

Belinostat (PXD101)

PHASE 2, FAST TRACK FDA , ORPHAN STATUS

  • PDX101
  • PX 105684
  • PXD-101
  • PXD101
  • UNII-F4H96P17NZ

Belinostat (PXD101) is a novel HDAC inhibitor with IC50 of 27 nM, with activity demonstrated in cisplatin-resistant tumors.

CLINICAL TRIALS…http://clinicaltrials.gov/search/intervention=Belinostat+OR+PXD101

Belinostat inhibits the growth of tumor cells (A2780, HCT116, HT29, WIL, CALU-3, MCF7, PC3 and HS852) with IC50 from 0.2-0.66 μM. PD101 shows low activity in A2780/cp70 and 2780AD cells. Belinostat inhibits bladder cancer cell growth, especially in 5637 cells, which shows accumulation of G0-G1 phase, decrease in S phase, and increase in G2-M phase. Belinostat also shows enhanced tubulin acetylation in ovarian cancer cell lines. A recent study shows that Belinostat activates protein kinase A in a TGF-β signaling-dependent mechanism and decreases survivin mRNA.

MW 318.07
MF C15H14N2O4S

414864-00-9  cas no

866323-14-0

(2E)-N-hydroxy-3-[3-(phenylsulfamoyl)phenyl]acrylamide

A novel HDAC inhibitor

…………………………

BELINOSTAT

Belinostat (PXD101) is experimental drug candidate under development byTopoTarget for the treatment of hematological malignancies and solid tumors. It is a histone deacetylase inhibitor.[1]

A hydroxamate-type inhibitor of histone deacetylase.

NCI: A novel hydroxamic acid-type histone deacetylase (HDAC) inhibitor with antineoplastic activity. Belinostat targets HDAC enzymes, thereby inhibiting tumor cell proliferation, inducing apoptosis, promoting cellular differentiation, and inhibiting angiogenesis. This agent may sensitize drug-resistant tumor cells to other antineoplastic agents, possibly through a mechanism involving the down-regulation of thymidylate synthase

In 2007 preliminary results were released from the Phase II clinical trial of intravenous belinostat in combination with carboplatin and paclitaxel for relapsedovarian cancer.[2] Final results in late 2009 of a phase II trial for T cell lymphomawere encouraging.[3] Belinostat has been granted orphan drug and fast trackdesignation by the FDA.[4]

 

The study of inhibitors of histone deacetylases indicates that these enzymes play an important role in cell proliferation and differentiation. The inhibitor Trichostatin A (TSA) (Yoshida et al., 1990a) causes cell cycle arrest at both G1 and G2 phases (Yoshida and Beppu, 1988), reverts the transformed phenotype of different cell lines, and induces differentiation of Friend leukaemia cells and others (Yoshida et al., 1990b). TSA (and SAHA) have been reported to inhibit cell growth, induce terminal differentiation, and prevent the formation of tumours in mice (Finnin et al., 1999).

Trichostatin A (TSA)

 

Figure imgf000005_0001

Suberoylanilide Hydroxamic Acid (SAHA)

 

Figure imgf000005_0002

Cell cycle arrest by TSA correlates with an increased expression of gelsolin (Hoshikawa et al., 1994), an actin regulatory protein that is down regulated in malignant breast cancer (Mielnicki et al., 1999). Similar effects on cell cycle and differentiation have been observed with a number of deacetylase inhibitors (Kim et al., 1999). Trichostatin A has also been reported to be useful in the treatment of fibrosis, e.g., liver fibrosis and liver cirrhosis. See, e.g., Geerts et al., 1998.

Recently, certain compounds that induce differentiation have been reported to inhibit histone deacetylases. Several experimental antitumour compounds, such as trichostatin A (TSA), trapoxin, suberoylanilide hydroxamic acid (SAHA), and phenylbutyrate have been reported to act, at least in part, by inhibiting histone deacetylase (see, e.g., Yoshida et al., 1990; Richon et al., 1998; Kijima et al., 1993). Additionally, diallyl sulfide and related molecules (see, e.g., Lea et al., 1999), oxamflatin (see, e.g., Kim et al., 1999), MS-27-275, a synthetic benzamide derivative (see, e.g., Saito et al., 1999; Suzuki et al., 1999; note that MS-27-275 was later re-named as MS-275), butyrate derivatives (see, e.g., Lea and Tulsyan, 1995), FR901228 (see, e.g., Nokajima et al., 1998), depudecin (see, e.g., Kwon et al., 1998), and m-carboxycinnamic acid bishydroxamide (see, e.g., Richon et al., 1998) have been reported to inhibit histone deacetylases. In vitro, some of these compounds are reported to inhibit the growth of fibroblast cells by causing cell cycle arrest in the G1 and G2 phases, and can lead to the terminal differentiation and loss of transforming potential of a variety of transformed cell lines (see, e.g., Richon et al, 1996; Kim et al., 1999; Yoshida et al., 1995; Yoshida & Beppu, 1988). In vivo, phenybutyrate is reported to be effective in the treatment of acute promyelocytic leukemia in conjunction with retinoic acid (see, e.g., Warrell et al., 1998). SAHA is reported to be effective in preventing the formation of mammary tumours in rats, and lung tumours in mice (see, e.g., Desai et al., 1999).

The clear involvement of HDACs in the control of cell proliferation and differentiation suggest that aberrant HDAC activity may play a role in cancer. The most direct demonstration that deacetylases contribute to cancer development comes from the analysis of different acute promyelocytic leukaemias (APL). In most APL patients, a translocation of chromosomes 15 and 17 (t(15;17)) results in the expression of a fusion protein containing the N-terminal portion of PML gene product linked to most of RARσ (retinoic acid receptor). In some cases, a different translocation (t(11 ;17)) causes the fusion between the zinc finger protein PLZF and RARα. In the absence of ligand, the wild type RARα represses target genes by tethering HDAC repressor complexes to the promoter DNA. During normal hematopoiesis, retinoic acid (RA) binds RARα and displaces the repressor complex, allowing expression of genes implicated in myeloid differentiation. The RARα fusion proteins occurring in APL patients are no longer responsive to physiological levels of RA and they interfere with the expression of the RA- inducible genes that promote myeloid differentiation. This results in a clonal expansion of promyelocytic cells and development of leukaemia. In vitro experiments have shown that TSA is capable of restoring RA-responsiveness to the fusion RARα proteins and of allowing myeloid differentiation. These results establish a link between HDACs and oncogenesis and suggest that HDACs are potential targets for pharmaceutical intervention in APL patients. (See, for example, Kitamura et al., 2000; David et al., 1998; Lin et al., 1998).

BELINOSTAT

Furthermore, different lines of evidence suggest that HDACs may be important therapeutic targets in other types of cancer. Cell lines derived from many different cancers (prostate, coloreetal, breast, neuronal, hepatic) are induced to differentiate by HDAC inhibitors (Yoshida and Horinouchi, 1999). A number of HDAC inhibitors have been studied in animal models of cancer. They reduce tumour growth and prolong the lifespan of mice bearing different types of transplanted tumours, including melanoma, leukaemia, colon, lung and gastric carcinomas, etc. (Ueda et al., 1994; Kim et al., 1999).

Psoriasis is a common chronic disfiguring skin disease which is characterised by well-demarcated, red, hardened scaly plaques: these may be limited or widespread. The prevalence rate of psoriasis is approximately 2%, i.e., 12.5 million sufferers in the triad countries (US/Europe/Japan). While the disease is rarely fatal, it clearly has serious detrimental effects upon the quality of life of the patient: this is further compounded by the lack of effective therapies. Present treatments are either ineffective, cosmetically unacceptable, or possess undesired side effects. There is therefore a large unmet clinical need for effective and safe drugs for this condition. Psoriasis is a disease of complex etiology. Whilst there is clearly a genetic component, with a number of gene loci being involved, there are also undefined environmental triggers. Whatever the ultimate cause of psoriasis, at the cellular level, it is characterised by local T-cell mediated inflammation, by keratinocyte hyperproliferation, and by localised angiogenesis. These are all processes in which histone deacetylases have been implicated (see, e.g., Saunders et al., 1999; Bernhard et al, 1999; Takahashi et al, 1996; Kim et al , 2001 ). Therefore HDAC inhibitors may be of use in therapy for psoriasis. Candidate drugs may be screened, for example, using proliferation assays with T-cells and/or keratinocytes.

 ………………………………………………………………………..

 

PXD101/Belinostat®

(E)-N-hydroxy-3-(3-phenylsulfamoyl-phenyl)-acrylamide, also known as PXD101 and Belinostat®, shown below, is a well known histone deacetylate (HDAC) inhibitor. It is being developed for treatment of a range of disorders mediated by HDAC, including proliferative conditions (such as cancer and psoriasis), malaria, etc.

Figure US20100286279A1-20101111-C00001

PXD101 was first described in WO 02/30879 A2. That document describes a multi-step method of synthesis which may conveniently be illustrated by the following scheme.

Figure US20100286279A1-20101111-C00002
Figure US20100286279A1-20101111-C00003

…………………………………..

GENERAL SYNTHESIS

WO2002030879A2

IGNORE 10

Figure imgf000060_0002

ENTRY 45 IS BELINOSTAT

Scheme 1

 

Figure imgf000101_0001

By using amines instead of aniline, the corresponding products may be obtained. The use of aniline, 4-methoxyaniline, 4-methylaniline, 4-bromoaniline, 4-chloroaniline, 4-benzylamine, and 4-phenethyamine, among others, is described in the Examples below.

In another method, a suitable amino acid (e.g., ω-amino acid) having a protected carboxylic acid (e.g., as an ester) and an unprotected amino group is reacted with a sulfonyl chloride compound (e.g., RSO2CI) to give the corresponding sulfonamide having a protected carboxylic acid. The protected carboxylic acid is then deprotected using base to give the free carboxylic acid, which is then reacted with, for example, hydroxylamine 2-chlorotrityl resin followed by acid (e.g., trifluoroacetic acid), to give the desired carbamic acid.

One example of this approach is illustrated below, in Scheme 2, wherein the reaction conditions are as follows: (i) RSO2CI, pyridine, DCM, room temperature, 12 hours; (ii) 1 M LiOH or 1 M NaOH, dioxane, room temperature, 3-48 hours; (iii) hydroxylamine 2-chlorotrityl resin, HOAt, HATU, DIPEA, DCM, room temperature, 16 hours; and (iv) TFA/DCM (5:95, v/v), room temperature, 1.5 hours.

Scheme 2

 

Figure imgf000102_0001

Additional methods for the synthesis of compounds of the present invention are illustrated below and are exemplified in the examples below.

Scheme 3A

 

Figure imgf000102_0002

Scheme 3B

 

Figure imgf000103_0001

Scheme 4

 

Figure imgf000104_0001
Figure imgf000105_0001

 

 

Scheme 8

 

Figure imgf000108_0002

Scheme 9

 

Figure imgf000109_0001

……………………………………………………………………..

SYNTHESIS

WO2002030879A2

Example 1

3-Formylbenzenesulfonic acid, sodium salt (1)

 

Figure imgf000123_0001

Oleum (5 ml) was placed in a reaction vessel and benzaldehyde (2.00 g, 18.84 mmol) was slowly added not exceeding the temperature of the reaction mixture more than 30°C. The obtained solution was stirred at 40°C for ten hours and at ambient temperature overnight. The reaction mixture was poured into ice and extracted with ethyl acetate. The aqueous phase was treated with CaC03 until the evolution of C02 ceased (pH~6-7), then the precipitated CaSO4was filtered off and washed with water. The filtrate was treated with Na2CO3 until the pH of the reaction medium increased to pH 8, obtained CaCO3 was filtered off and water solution was evaporated in vacuum. The residue was washed with methanol, the washings were evaporated and the residue was dried in desiccator over P2Oβ affording the title compound (2.00 g, 51%). 1H NMR (D20), δ: 7.56-8.40 (4H, m); 10.04 ppm (1 H, s).

Example 2 3-(3-Sulfophenyl)acrylic acid methyl ester, sodium salt (2)

 

Figure imgf000124_0001

Sodium salt of 3-formylbenzenesulfonic acid (1) (1.00 g, 4.80 mmol), potassium carbonate (1.32 g, 9.56 mmol), trimethyl phosphonoacetate (1.05 g, 5.77 mmol) and water (2 ml) were stirred at ambient temperature for 30 min., precipitated solid was filtered and washed with methanol. The filtrate was evaporated and the title compound (2) was obtained as a white solid (0.70 g, 55%). 1H NMR (DMSO- dβl HMDSO), δ: 3.68 (3H, s); 6.51 (1 H, d, J=16.0 Hz); 7.30-7.88 (5H, m).

Example 3 3-(3-Chlorosulfonylphenyl)acrylic acid methyl ester (3)

 

Figure imgf000124_0002

To the sodium salt of 3-(3-sulfophenyl)acrylic acid methyl ester (2) (0.670 g, 2.53 mmol) benzene (2 ml), thionyl chloride (1.508 g, 0.9 ml, 12.67 mmol) and 3 drops of dimethylformamide were added and the resultant suspension was stirred at reflux for one hour. The reaction mixture was evaporated, the residue was dissolved in benzene (3 ml), filtered and the filtrate was evaporated to give the title compound (0.6’40 g, 97%).

Example 4 3-(3-Phenylsulfamoylphenyl)acrylic acid methyl ester (4a)

 

Figure imgf000125_0001

A solution of 3-(3-chlorosulfonylphenyl)acrylic acid methyl ester (3) (0.640 g, 2.45 mmol) in dichloromethane (2 ml) was added to a mixture of aniline (0.465 g, 4.99 mmol) and pyridine (1 ml), and the resultant solution was stirred at 50°C for one hour. The reaction mixture was evaporated and the residue was partitioned between ethyl acetate and 10% HCI. The organic layer was washed successively with water, saturated NaCl, and dried (Na2S0 ). The solvent was removed and the residue was chromatographed on silica gel with chloroform-ethyl acetate (7:1 , v/v) as eluent. The obtained product was washed with diethyl ether to give the title compound (0.226 g, 29%). 1H NMR (CDCI3, HMDSO), δ: 3.72 (3H, s); 6.34 (1H, d, J=16.0 Hz); 6.68 (1 H, br s); 6.92-7.89 (10H, m).

Example 5 3-(3-Phenylsulfamoylphenyl)acrylic acid (5a)

 

Figure imgf000125_0002

3-(3-Phenylsulfamoylphenyl)acrylic acid methyl ester (4a) (0.220 g, 0.69 mmol) was dissolved in methanol (3 ml), 1N NaOH (2.08 ml, 2.08 mmol) was added and the resultant solution was stirred at ambient temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The aqueous layer was acidified with 10% HCI and stirred for 30 min. The precipitated solid was filtered, washed with water and dried in desiccator over P2Os to give the title compound as a white solid (0.173 g, 82%). Example 6 3-(3-Phenylsulfamoylphenyl)acryloyl chloride (6a)

 

Figure imgf000126_0001

To a suspension of 3-(3-phenylsulfamoylphenyl)acrylic acid (5a) (0.173 g, 0.57 mmol) in dichloromethane (2.3 ml) oxalyl chloride (0.17 ml, 1.95 mmol) and one drop of dimethylformamide were added. The reaction mixture was stirred at 40°C for one hour and concentrated under reduced pressure to give crude title compound (0.185 g).

Example 7

N-Hydroxy-3-(3-phenylsulfamoylphenyl)acrylamide (7a) (PX105684) BELINOSTAT

 

Figure imgf000126_0002

To a suspension of hydroxylamine hydrochloride (0.200 g, 2.87 mmol) in tetrahydrofuran (3.5 ml) a saturated NaHCOβ solution (2.5 ml) was added and the resultant mixture was stirred at ambient temperature for 10 min. To the reaction mixture a 3-(3-phenylsulfamoylphenyl)acryloyl chloride (6a) (0.185 g) solution in tetrahydrofuran (2.3 ml) was added and stirred at ambient temperature for one hour. The reaction mixture was partitioned between ethyl acetate and 2N HCI. The organic layer was washed successively with water and saturated NaCl, the solvent was removed and the residue was washed with acetonitrile and diethyl ether.

The title compound was obtained as a white solid (0.066 g, 36%), m.p. 172°C. BELINOSTAT

1H NMR (DMSO-d6, HMDSO), δ: 6.49 (1 H, d, J=16.0 Hz); 7.18-8.05 (10H, m); 9.16 (1 H, br s); 10.34 (1 H, s); 10.85 ppm (1 H, br s).

HPLC analysis on Symmetry C18column: impurities 4% (column size 3.9×150 mm; mobile phase acetonitrile – 0.1 M phosphate buffer (pH 2.5), 40:60; sample concentration 1 mg/ml; flow rate 0.8 ml/ min; detector UV 220 nm).

Anal. Calcd for C154N204S, %: C 56.59, H 4.43, N 8.80. Found, %: C 56.28, H 4.44, N 8.56.

……………………………………………………………………….

SYNTHESIS

US20100286279

Figure US20100286279A1-20101111-C00034

 

 

…………………………………………………….

SYNTHESIS AND SPECTRAL DATA

Journal of Medicinal Chemistry, 2011 ,  vol. 54,  13  pg. 4694 – 4720

(E)-N-Hydroxy-3-(3-phenylsulfamoyl-phenyl)-acrylamide (28, belinostat, PXD101).

http://pubs.acs.org/doi/full/10.1021/jm2003552

 http://pubs.acs.org/doi/suppl/10.1021/jm2003552/suppl_file/jm2003552_si_001.pdf

The methyl ester (27) (8.0 g) was prepared according to reported synthetic route,

(Watkins, C. J.; Romero-Martin, M.-R.; Moore, K. G.; Ritchie, J.; Finn, P. W.; Kalvinsh, I.;
Loza, E.; Dikvoska, K.; Gailite, V.; Vorona, M.; Piskunova, I.; Starchenkov, I.; Harris, C. J.;
Duffy, J. E. S. Carbamic acid compounds comprising a sulfonamide linkage as HDAC
inhibitors. PCT Int. Appl. WO200230879A2, April 18, 2002.)
but using procedure D (Experimental Section) or method described for 26 to convert the methyl ester to crude
hydroxamic acid which was further purified by chromatography (silica, MeOH/DCM = 1:10) to
afford 28 (PXD101) as off-white or pale yellow powder (2.5 g, 31%).

LC–MS m/z 319.0 ([M +H]+).

1H NMR (DMSO-d6)  12–9 (very broad, 2H), 7.90 (s, 1H), 7.76 (d, J = 7.7 Hz, 1H), 7.70 (d, J

= 7.8 Hz, 1H), 7.56 (t, J = 7.8 Hz, 1H), 7.44 (d, J = 15.8 Hz, 1H), 7.22 (t, J = 7.8 Hz, 2H), 7.08 (d,
J = 7.8 Hz, 2H), 7.01 (t, J = 7.3 Hz, 1H), 6.50 (d, J = 15.8 Hz, 1H);

13C NMR (DMSO-d6)  162.1,
140.6, 138.0, 136.5, 135.9, 131.8, 130.0, 129.2, 127.1, 124.8, 124.1, 121.3, 120.4.

Anal.
(C15H14N2O4S) C, H, N

………………………………………………..

SYNTHESIS

WO2009040517A2

PXDIOI / Belinostat®

(E)-N-hydroxy-3-(3-phenylsulfamoyl-phenyl)-acrylamide, also known as PXD101 and Belinostat®, shown below, is a well known histone deacetylate (HDAC) inhibitor. It is being developed for treatment of a range of disorders mediated by HDAC, including proliferative conditions (such as cancer and psoriasis), malaria, etc.

 

Figure imgf000003_0001

PXD101 was first described in WO 02/30879 A2. That document describes a multi-step method of synthesis which may conveniently be illustrated by the following scheme.

Scheme 1

Not isolated

Figure imgf000003_0002

ed on (A)

on (D)

Figure imgf000003_0003

d on (H)

Figure imgf000004_0001

There is a need for alternative methods for the synthesis of PXD101 and related compounds for example, methods which are simpler and/or employ fewer steps and/or permit higher yields and/or higher purity product.

Scheme 5

 

Figure imgf000052_0001

DMAP, toluene

Figure imgf000052_0003
Figure imgf000052_0002

 

Figure imgf000052_0004

Synthesis 1 3-Bromo-N-phenyl-benzenesulfonamide (3)

 

Figure imgf000052_0005

To a 30 gallon (-136 L) reactor was charged aniline (2) (4.01 kg; 93.13 g/mol; 43 mol), toluene (25 L), and 4-(dimethylamino)pyridine (DMAP) (12 g), and the mixture was heated to 50-600C. 3-Bromobenzenesulfonyl chloride (1) (5 kg; 255.52 g/mol; 19.6 mol) was charged into the reactor over 30 minutes at 50-600C and progress of the reaction was monitored by HPLC. After 19 hours, toluene (5 L) was added due to losses overnight through the vent line and the reaction was deemed to be complete with no compound (1) being detected by HPLC. The reaction mixture was diluted with toluene (10 L) and then quenched with 2 M aqueous hydrochloric acid (20 L). The organic and aqueous layers were separated, the aqueous layer was discarded, and the organic layer was washed with water (20 L), and then 5% (w/w) sodium bicarbonate solution (20 L), while maintaining the batch temperature at 45-55°C. The batch was then used in the next synthesis.

Synthesis 2 (E)-3-(3-Phenylsulfamoyl-phenyl)-acrylic acid ethyl ester (5)

 

Figure imgf000053_0001

To the batch containing 3-bromo-N-phenyl-benzenesulfonamide (3) (the treated organic layer obtained in the previous synthesis) was added triethylamine (2.97 kg; 101.19 g/mol; 29.4 mol), tri(o-tolyl)phosphine (119 g; 304.37 g/mol; 0.4 mol), and palladium (II) acetate (44 g; 224.51 g/mol; 0.2 mol), and the resulting mixture was degassed four times with a vacuum/nitrogen purge at 45-55°C. Catalytic palladium (0) was formed in situ. The batch was then heated to 80-900C and ethyl acrylate (4) (2.16 kg; 100.12 g/mol; 21.6 mol) was slowly added over 2.75 hours. The batch was sampled after a further 2 hours and was deemed to be complete with no compound (3) being detected by HPLC. The batch was cooled to 45-55°C and for convenience was left at this temperature overnight.

The batch was then reduced in volume under vacuum to 20-25 L, at a batch temperature of 45-55°C, and ethyl acetate (20 L) was added. The batch was filtered and the residue washed with ethyl acetate (3.5 L). The residue was discarded and the filtrates were sent to a 100 gallon (-454 L) reactor, which had been pre-heated to 600C. The 30 gallon (-136 L) reactor was then cleaned to remove any residual Pd, while the batch in the 100 gallon (-454 L) reactor was washed with 2 M aqueous hydrochloric acid and water at 45-55°C. Once the washes were complete and the 30 gallon (-136 L) reactor was clean, the batch was transferred from the 100 gallon (-454 L) reactor back to the 30 gallon (-136 L) reactor and the solvent was swapped under vacuum from ethyl acetate/toluene to toluene while maintaining a batch temperature of 45-55°C (the volume was reduced to 20-25 L). At this point, the batch had precipitated and heptanes (10 L) were added to re-dissolve it. The batch was then cooled to 0-100C and held at this temperature over the weekend in order to precipitate the product. The batch was filtered and the residue was washed with heptanes (5 L). A sample of the wet-cake was taken for Pd analysis. The Pd content of the crude product (5) was determined to be 12.9 ppm.

The wet-cake was then charged back into the 30 gallon (-136 L) reactor along with ethyl acetate (50 L) and heated to 40-500C in order to obtain a solution. A sparkler filter loaded with 12 impregnated Darco G60® carbon pads was then connected to the reactor and the solution was pumped around in a loop through the sparkler filter. After 1 hour, a sample was taken and evaporated to dryness and analysed for Pd content. The amount of Pd was found to be 1.4 ppm. A second sample was taken after 2 hours and evaporated to dryness and analysed for Pd content. The amount of Pd had been reduced to 0.6 ppm. The batch was blown back into the reactor and held at 40-500C overnight before the solvent was swapped under vacuum from ethyl acetate to toluene while maintaining a batch temperature of 45-55°C (the volume was reduced to 20-25 L). At this point, the batch had precipitated and heptanes (10 L) were added to re-dissolve it and the batch was cooled to 0-100C and held at this temperature overnight in order to precipitate the product. The batch was filtered and the residue was washed with heptanes (5 L). The filtrate was discarded and the residue was dried at 45-55°C under vacuum for 25 hours. A first lot of the title compound (5) was obtained as an off-white solid (4.48 kg, 69% overall yield from 3-bromobenzenesulfonyl chloride (1)) with a Pd content of 0.4 ppm and a purity of 99.22% (AUC) by HPLC.

Synthesis 3 (E)-3-(3-Phenylsulfamoyl-phenyl)-acrvlic acid (6)

 

Figure imgf000054_0001

To the 30 gallon (-136 L) reactor was charged the (E)-3-(3-phenylsulfamoyl-phenyl)- acrylic acid ethyl ester (5) (4.48 kg; 331.39 g/mol; 13.5 mol) along with 2 M aqueous sodium hydroxide (17.76 L; -35 mol). The mixture was heated to 40-50°C and held at this temperature for 2 hours before sampling, at which point the reaction was deemed to be complete with no compound (5) being detected by HPLC. The batch was adjusted to pH 2.2 using 1 M aqueous hydrochloric acid while maintaining the batch temperature between 40-500C. The product had precipitated and the batch was cooled to 20-300C and held at this temperature for 1 hour before filtering and washing the cake with water (8.9 L). The filtrate was discarded. The batch was allowed to condition on the filter overnight before being charged back into the reactor and slurried in water (44.4 L) at 40-500C for 2 hours. The batch was cooled to 15-20°C, held for 1 hour, and then filtered and the residue washed with water (8.9 L). The filtrate was discarded. The crude title compound (6) was transferred to an oven for drying at 45-55°C under vacuum with a slight nitrogen bleed for 5 days (this was done for convenience) to give a white solid (3.93 kg, 97% yield). The moisture content of the crude material was measured using Karl Fischer (KF) titration and found to be <0.1% (w/w). To the 30 gallon (-136 L) reactor was charged the crude compound (6) along with acetonitrile (47.2 L). The batch was heated to reflux (about 80°C) and held at reflux for 2 hours before cooling to 0-10°C and holding at this temperature overnight in order to precipitate the product. The batch was filtered and the residue was washed with cold acetonitrile (7.9 L). The filtrate was discarded and the residue was dried under vacuum at 45-55°C for 21.5 hours. The title compound (6) was obtained as a fluffy white solid (3.37 kg, 84% yield with respect to compound (5)) with a purity of 99.89% (AUC) by HPLC.

Synthesis 4 (E)-N-Hvdroxy-3-(3-phenylsulfamoyl-phenyl)-acrylamide (PXD101) BELINOSTAT

 

Figure imgf000055_0001

To the 30 gallon (-136 L) reactor was charged (E)-3-(3-phenylsulfamoyl-phenyl)-acrylic acid (6) (3.37 kg; 303.34 g/mol; 11.1 mol) and a pre-mixed solution of 1 ,8-diazabicyclo[5.4.0]undec-7-ene (DBU) in isopropyl acetate (IPAc) (27 g in 30 L; 152.24 g/mol; 0.18 mol). The slurry was stirred and thionyl chloride (SOCI2) (960 mL; density ~1.631 g/mL; 118.97 g/mol; -13 mol) was added to the reaction mixture and the batch was stirred at 20-300C overnight. After 18.5 hours, the batch was sampled and deemed to be complete with no compound (6) being detected by HPLC. The resulting solution was transferred to a 100 L Schott reactor for temporary storage while the

30 gallon (-136 L) reactor was rinsed with isopropyl acetate (IPAc) and water. Deionized water (28.9 L) was then added to the 30 gallon (-136 L) reactor followed by 50% (w/w) hydroxylamine (6.57 L; -1.078 g/mL; 33.03 g/mol; -214 mol) and another charge of deionized water (1.66 L) to rinse the lines free of hydroxylamine to make a 10% (w/w) hydroxylamine solution. Tetrahydrofuran (THF) (6.64 L) was then charged to the

30 gallon (-136 L) reactor and the mixture was stirred and cooled to 0-100C. The acid chloride solution (from the 100 L Schott reactor) was then slowly charged into the hydroxylamine solution over 1 hour maintaining a batch temperature of 0-10°C during the addition. The batch was then allowed to warm to 20-300C. The aqueous layer was separated and discarded. The organic layer was then reduced in volume under vacuum while maintaining a batch temperature of less than 300C. The intention was to distill out 10-13 L of solvent, but this level was overshot. A larger volume of isopropyl acetate (IPAc) (16.6 L) was added and about 6 L of solvent was distilled out. The batch had precipitated and heptanes (24.9 L) were added and the batch was held at 20-30°C overnight. The batch was filtered and the residue was washed with heptanes (6.64 L). The filtrate was discarded and the residue was dried at 45-55°C under vacuum with a slight nitrogen bleed over the weekend. The title compound (PXD101) was obtained as a light orange solid (3.11 kg, 89% yield with respect to compound (6)) with a purity of 99.25% (AUC) by HPLC.

The title compound (PXD101) (1.2 kg, 3.77 mol) was dissolved in 8 volumes of 1:1 (EtOH/water) at 600C. Sodium bicarbonate (15.8 g, 5 mol%) was added to the solution. Water (HPLC grade) was then added at a rate of 65 mL/min while keeping the internal temperature >57°C. After water (6.6 L) had been added, crystals started to form and the water addition was stopped. The reaction mixture was then cooled at a rate of 10°C/90 min to a temperature of 0-10cC and then stirred at ambient temperature overnight. The crystals were then filtered and collected. The filter cake was washed by slurrying in water (2 x 1.2 L) and then dried in an oven at 45°C for 60 hours with a slight nitrogen bleed. 1.048 kg (87% recovery) of a light orange solid was recovered. Microscopy and XRPD data showed a conglomerate of irregularly shaped birefringant crystalline particles. The compound was found to contain 0.02% water.

As discussed above: the yield of compound (5) with respect to compound (1) was 69%. the yield of compound (6) with respect to compound (5) was 84%. the yield of PXD101 with respect to compound (6) was 89%.

……………….

FORMULATION

WO2006120456A1

Formulation Studies

These studies demonstrate a substantial enhancement of HDACi solubility (on the order of a 500-fold increase for PXD-101) using one or more of: cyclodextrin, arginine, and meglumine. The resulting compositions are stable and can be diluted to the desired target concentration without the risk of precipitation. Furthermore, the compositions have a pH that, while higher than ideal, is acceptable for use.

 

Figure imgf000047_0001

UV Absorbance

The ultraviolet (UV absorbance E\ value for PXD-101 was determined by plotting a calibration curve of PXD-101 concentration in 50:50 methanol/water at the λmax for the material, 269 nm. Using this method, the E1i value was determined as 715.7.

Methanol/water was selected as the subsequent diluting medium for solubility studies rather than neat methanol (or other organic solvent) to reduce the risk of precipitation of the cyclodextrin.

Solubility in Demineralised Water

The solubility of PXD-101 was determined to be 0.14 mg/mL for demineralised water. Solubility Enhancement with Cvclodextrins

Saturated samples of PXD-101 were prepared in aqueous solutions of two natural cyclodextrins (α-CD and γ-CD) and hydroxypropyl derivatives of the α, β and Y cyclodextrins (HP-α-CD, HP-β-CD and HP-γ-CD). All experiments were completed with cyclodextrin concentrations of 250 mg/mL, except for α-CD, where the solubility of the cyclodextrin was not sufficient to achieve this concentration. The data are summarised in the following table. HP-β-CD offers the best solubility enhancement for PXD-101.

 

Figure imgf000048_0001

Phase Solubility Determination of HP-β-CD

The phase solubility diagram for HP-β-CD was prepared for concentrations of cyclodextrin between 50 and 500 mg/mL (5-50% w/v). The calculated saturated solubilities of the complexed HDACi were plotted against the concentration of cyclodextrin. See Figure 1.

………………………..

  1.  Plumb, Jane A.; Finn, Paul W.; Williams, Robert J.; Bandara, Morwenna J.; Romero, M. Rosario; Watkins, Claire J.; La Thangue, Nicholas B.; Brown, Robert (2003). “Pharmacodynamic Response and Inhibition of Growth of Human Tumor Xenografts by the Novel Histone Deacetylase Inhibitor PXD101”. Molecular Cancer Therapeutics 2 (8): 721–728. PMID 12939461.
  2.  “CuraGen Corporation (CRGN) and TopoTarget A/S Announce Presentation of Belinostat Clinical Trial Results at AACR-NCI-EORTC International Conference”. October 2007.
  3. Final Results of a Phase II Trial of Belinostat (PXD101) in Patients with Recurrent or Refractory Peripheral or Cutaneous T-Cell Lymphoma, December 2009
  4.  “Spectrum adds to cancer pipeline with $350M deal.”. February 2010.
  5. Helvetica Chimica Acta, 2005 ,  vol. 88,  7  PG. 1630 – 1657, MP 172
  6. WO2009/40517 A2, ….
  7. WO2006/120456 A1, …..
  8. Synthetic Communications, 2010 ,  vol. 40,  17  PG. 2520 – 2524, MP 172
  9. Journal of Medicinal Chemistry, 2011 ,  vol. 54,   13  PG. 4694 – 4720, NMR IN SUP INFO

 

11-7-2008
Histone Deacetylase (Hdac) Inhibitors (Pxd101) for the Treatment of Cancer Alone or in Combination With Chemotherapeutic Agent
9-19-2008
CYCLOOXYGENASE-2 INHIBITOR/HISTONE DEACETYLASE INHIBITOR COMBINATION
9-5-2008
Combination Therapies Using Hdac Inhibitors
8-15-2008
Pharmaceutical Formulations Of Hdac Inhibitors
8-6-2008
Carbamic acid compounds comprising a sulfonamide linkage as HDAC inhibitors
7-23-2008
Cyclooxygenase-2 inhibitor/histone deacetylase inhibitor combination
2-28-2007
Carbamic acid compounds comprising a sulfonamide linkage as HDAC inhibitors
5-20-2005
Carbamic acid compounds comprising a sulfonamide linkage as HDAC inhibitors
5-4-2005
Carbamic acid compounds comprising a sulfonamide linkage as hdac inhibitors

 

WO2002030879A2 Sep 27, 2001 Apr 18, 2002 Prolifix Ltd Carbamic acid compounds comprising asulfonamide linkage as hdac inhibitors
7-6-2011
HYDROXAMIC ACID DERIVATIVES AS INHIBITORS OF HDAC ENZYMATIC ACTIVITY
4-20-2011
Combined Use of Prame Inhibitors and Hdac Inhibitors
3-32-2011
5-LIPOXYGENASE INHIBITORS
1-7-2011
Methods of Treatment Employing Prolonged Continuous Infusion of Belinostat
11-12-2010
Methods of Synthesis of Certain Hydroxamic Acid Compounds
7-30-2010
Methods for identifying patients who will respond well to cancer treatment
1-15-2010
HDAC INHIBITORS
12-18-2009
COMBINATION OF ORGANIC COMPOUNDS
7-31-2009
CYCLOOXYGENASE-2 INHIBITOR/HISTONE DEACETYLASE INHIBITOR COMBINATION
7-8-2009
CARBAMIC ACID COMPOUNDS COMPRISING A SULFONAMIDE LINKAGE AS HDAC INHIBITORS
WO1998038859A1 * Mar 4, 1998 Sep 11, 1998 Thomas E Barta Sulfonyl divalent aryl or heteroaryl hydroxamic acid compounds
WO1999024399A1 * Nov 12, 1998 May 20, 1999 Darwin Discovery Ltd Hydroxamic and carboxylic acid derivatives having mmp and tnf inhibitory activity
WO2000056704A1 * Mar 22, 2000 Sep 28, 2000 Duncan Batty Hydroxamic and carboxylic acid derivatives
WO2000069819A1 * May 12, 2000 Nov 23, 2000 Thomas E Barta Hydroxamic acid derivatives as matrix metalloprotease inhibitors
WO2001038322A1 * Nov 22, 2000 May 31, 2001 Methylgene Inc Inhibitors of histone deacetylase
EP0570594A1 * Dec 7, 1992 Nov 24, 1993 SHIONOGI &amp; CO., LTD. Hydroxamic acid derivative based on aromatic sulfonamide
EP0931788A2 * Dec 16, 1998 Jul 28, 1999 Pfizer Inc. Metalloprotease inhibitors
GB2312674A * Title not available

 

WO2002030879A2 Sep 27, 2001 Apr 18, 2002 Prolifix Ltd Carbamic acid compounds comprising a sulfonamide linkage as hdac inhibitors
WO2005063806A1 Dec 30, 2003 Jul 14, 2005 Council Scient Ind Res Arginine hydrochloride enhances chaperone-like activity of alpha crystallin
US4642316 May 20, 1985 Feb 10, 1987 Warner-Lambert Company Parenteral phenytoin preparations

 

WO2008090585A2 * Jan 25, 2008 Jul 31, 2008 Univ Roma Soluble forms of inclusion complexes of histone deacetylase inhibitors and cyclodextrins, their preparation processes and uses in the pharmaceutical field
WO2009109861A1 * Mar 6, 2009 Sep 11, 2009 Topotarget A/S Methods of treatment employing prolonged continuous infusion of belinostat
WO2010048332A2 * Oct 21, 2009 Apr 29, 2010 Acucela, Inc. Compounds for treating ophthalmic diseases and disorders
WO2011064663A1 Nov 24, 2010 Jun 3, 2011 Festuccia, Claudio Combination treatment employing belinostat and bicalutamide
US20110003777 * Mar 6, 2009 Jan 6, 2011 Topotarget A/S Methods of Treatment Employing Prolonged Continuous Infusion of Belinostat

………………………..

SPECTRUM

Tiny Biotech With Three Cancer Drugs Is More Alluring Takeover Bet Now
Forbes
The drug is one of Spectrum’s two drugs undergoing phase 3 clinical trials. Allergan paid Spectrum $41.5 million and will make additional payments of up to $304 million based on achieving certain milestones. So far, Raj Shrotriya, Spectrum’s chairman, 

http://www.forbes.com/sites/genemarcial/2013/07/14/tiny-biotech-with-three-cancer-drugs-is-more-alluring-takeover-bet-now/

……………………………..

Copenhagen, December 10, 2013
Topotarget announces the submission of a New Drug Application (NDA) for belinostat for the treatment of relapsed or refractory (R/R) peripheral T-cell lymphoma (PTCL) to the US Food and Drug Administration (FDA). The NDA has been filed for Accelerated Approval with a request for Priority Review. Response from the FDA regarding acceptance to file is expected within 60 days from the FDA receipt date.
read all this here
…………………….
Share
Jan 222014
 

Clazosentan

IUPAC Name: N-[6-(2-hydroxyethoxy)-5-(2-methoxyphenoxy)-2-[2-(2H-tetrazol-5-yl)
pyridin-4-yl]pyrimidin-4-yl]-5-methylpyridine-2-sulfonamide

5-methyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2- methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-ylamide

5-methyl-pyridine-2-sulfonic acid [6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide

VASODILATOR, Endothelin -1 – receptor antagonist

Clazosentan (Ro61-1790, AXV-034343)

  • AXV 034
  • AXV 034343
  • AXV-034343
  • AXV-343434
  • Clazosentan
  • Ro 61-1790
  • Ro-61-1790
  • UNII-3DRR0X4728
  • VML 588
  • VML-588

180384-56-9  cas no

CLINICAL TRIALS…http://clinicaltrials.gov/search/intervention=CLAZOSENTAN  in phase 3

Formula: C25H23N9O6S
Molecular Weight: 577.5718

Endothelin type-A receptor antagonist for the treatment of vasospasm in subarachnoid hemorrhage

Selective endothelin receptor antagonist (Pivlaz)

Acteliion…… innovator

Clazosentan is a drug with orphan drug status , which since 2007, currently in Phase III clinical trials CONSCIOUS-2 ( Clazosentan to O vercome N euro logical i SC Hemia and I nfarct O cc U rring after S ubarachnoid hemorrage) is located. It is for  treatment of vasospasm after subarachnoid hemorrhage are used (SAH).

Clazosentan is used by the Swiss pharmaceutical company Actelion developed. Medicinally, the disodium salt is used.Clazosentan to come under the name Pivlaz on the market.

The endothelin -1 – receptor is one of the strongest known vasoconstrictors . Clazosentan is an E -1 – receptor antagonist , for the treatment of vasospasm after subarachnoid hemorrhage is under development. After subarachnoid hemorrhage , an irritation of theblood vessels to a vasospasm and the associated to a reduced supply of brain tissue with oxygen lead. A possible consequence may be a Ischemic stroke be. Clazosentan acts this vasoconstriction contrary.

The plasma half-life of 6-10  min .

Actelion has initiated a comprehensive global phase IIb/III development program for clazosentan sodium (formerly Ro-61-1790, VML-588, …

CLAZOSENTAN

CLAZOSENTAN

CLAZOSENTAN DI-NA SALT is discontinued

Clazosentan, shown below, is a well known endothelin receptor antagonist.

 

Figure US20090069352A1-20090312-C00001

 

Since clazosentan is a known and useful pharmaceutical, it is desirable to discover novel derivatives thereof. Clazosentan is described in European Patent No. 0,799,209

IT IS DESCRIBED IN US6103902

US6004965 * Dec 8, 1995 Dec 21, 1999 Hoffmann-La Roche Inc. Sulfonamides
WO1996019459A1 * Dec 8, 1995 Jun 27, 1996 Volker Breu Novel sulfonamides

 

8-13-1998
Pyrrolidine-3-carboxylic acids as endothelin antagonists. 3. Discovery of a potent, 2-nonaryl, highly selective ETA antagonist (A-216546).
Journal of medicinal chemistry

 

7-23-2009
In silico prediction of volume of distribution in human using linear and nonlinear models on a 669 compound data set.
Journal of medicinal chemistry

………………………………………………………..

SYNTHESIS

EP0799209B1

EXAMPLE 15

a) In analogy to Example 1a), from 5-methyl-pyridine-2-sulphonic acid 6-chloro-5-(2-methoxy-phenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-ylamide there is obtained 5-methyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-ylamide, melting point 188-190

b) In analogy to Example 2, from 5-methyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-ylamide there is obtained 5-methyl-pyridine-2-sulphonic acid 2-(2-cyano-pyridin-4-yl)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-pyrimidin-4-ylamide.

……………………………………………..

SYNTHESIS

 

<br />
Clazosentan<br />
pk_prod_list.xml_prod_list_card_pr?p_tsearch=A&p_id=239030<br />

 

4-Cyanopyridine (I) is reacted with ammonium chloride in methanolic NaOMe to afford the amidine (II), which is cyclized with diethyl (2-methoxyphenoxy)malonate (III) producing the dihydroxypyrimidine (IV). Chlorination of (IV) in hot POCl3, followed by oxidation of the obtained dichloropyrimidine (V) with peracetic acid leads to the pyridine N-oxide (VI). Subsequent condensation of the dichloropyrimidine derivative (VI) with the potassium salt of 5-methylpyridine-2-sulfonamide (VII) yields the sulfonamido pyrimidine (VIII). The remaining chloride group of (VIII) is then displaced with the sodium alkoxide of ethylene glycol in hot DME to furnish the hydroxyethyl ether (IX). Treatment of the pyridine N-oxide (IX) with cyanotrimethylsilane and Et3N in refluxing acetonitrile gives rise to the 2-cyanopyridine (X), which is finally converted to the title tetrazole derivative by treatment with sodium azide in the presence of NH4Cl in DMF (1).

………………………………………………………….

SYNTHESIS OF DISODIUM SALT OF CLAZOSENTAN

US6103902

Figure 00050001

DESCRIBED IN WO 9619459.

Figure 00050002

Figure 00060001

  • III is reacted with a compound of formula V
    Figure 00060002
  • The reaction type is known in the art and may be performed under basic conditions for example in the presence of a coupling agent, e.g. 1,4-diazobicyclo[2.2.2]octane, together with potassium carbonate in acetone.

EXAMPLE 1

1360 ml of formamide were added to 136 g (437 mmol) of 5-(2-methoxy-phenoxy)-2-pyridine-4-yl-pyrimidine-4,6-diole. Then, at a temperature of 0 acid and thereafter 36.5 g (130 mmol) of iron(II)sulfate heptahydrate were added to the suspension. After that, 89 ml (874 mmol) of 30% hydrogen peroxide were added dropwise within 1 hr at a temperature of 0 to 5 0 sodium pyrosulfite in 680 ml of de-ionized water was added dropwise to the reaction mixture within 30 min. at 0 reaction mixture was stirred at 0 The suspension was then filtered under reduced pressure. The filtrate was first washed with 1750 ml of de-ionized water and thereafter with 700 ml of ethanol. Then the solid was dried at 80 There were obtained 132.4 g (91% of theory) of 4-[4,6-dihydroxy -5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carboxylic acid amide with a HPLC purity of 91.4% (w/w).

Preparation of Starting Material

a) 53.1 g of 4-cyano-pyridine (98%) are added all at once to a solution of 1.15 g of sodium in 200 ml of abs. MeOH. After 6 hr 29.5 g of NH.sub.4 Cl are added while stirring vigorously. The mixture is stirred at room temperature overnight. 600 ml of ether are added thereto, whereupon the precipitate is filtered off under suction and thereafter dried at 50 4-amidino-pyridine hydrochloride (decomposition point 245-247

b) 112.9 g of diethyl (2-methoxyphenoxy)malonate are added dropwise within 30 min. to a solution of 27.60 g of sodium in 400 ml of MeOH. Thereafter, 74.86 g of the amidine hydrochloride obtained in a) are added all at once. The mixture is stirred at room temperature overnight and evaporated at 50 of ether and filtered off under suction. The filter cake is dissolved in 1000 ml of H.sub.2 O and treated little by little with 50 ml of CH.sub.3 COOH. The precipitate is filtered off under suction, washed with 400 ml of H.sub.2 O and dried at 80 obtained 5-(2-methoxy-phenoxy)-2-(pyridine-4-yl)-pyrimidine-4,6-diole (or tautomer), melting point above 250

EXAMPLE 2

Within 20 min. 61 ml (633 mmol) of POCl.sub.3 were added dropwise to 34 ml (200 mmol) of diisopropyl ethylamine at 5 followed by stirring at 5 23.5 g (66 mmol) of 4-[4,6-dihydroxy-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carboxylic acid amide were added in four portions under cooling followed by stirring at 90 to 20 dichloromethane. Volatile components (i.e. excess of POCl.sub.3) was removed by evaporation from 20 re-distillation with 100 ml of toluene. After adding 250 ml of dichloromethane to the residue (88 g of a black oil) the solution was heated to 35 were added dropwise within 30 min. whereby the pH was kept constant by the subsequent addition of 28% NaOH solution (60 ml) within 5 to 6 hr. The mixture was stirred at 35 by removal of dichloromethane by distillation. The resulting suspension was allowed to cool down to 20 hr. The solid was filtered off under suction, washed with 500 ml of water and dried at 70 (86% of theory) of 4-[4,6-dichloro-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carbonitrile with a HPLC purity of 94.3% (w/w).

EXAMPLE 3

12.5 g (33.5 mmol) of 4-[4,6-dichloro-5-(2-methoxy-phenoxy)-pyrimidine-2-yl]-pyridine-2-carbonitrile and 6.06 g (35 mmol) of 5-methyl-pyridine-2-sulfonamide were added to 130 ml of acetone. 15 g of potassium carbonate and 190 mg (1.6 mmol) of 1,4-diazobicyclo[2.2.2]octane were added and the suspension was stirred at 40 de-ionized water were added followed by dropwise addition of 50 ml of 3 N hydrochloric acid (pH of the solution=1). Acetone was removed by evaporation and the suspension was stirred for 1 hr. The solid was filtered and washed with 100 ml of water. The residue was heated (reflux) in 100 ml of methanol for 1 hr followed by cooling to 20 solid was filtered and dried at 80 were obtained 16.0 g (93% of theory) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-2-(2-cyano-pyridine-4-yl)-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide with a HPLC purity of 90.3% (w/w).

 

EXAMPLE 5

20 g (39 mmol) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-2-(2-cyano-pyridine-4-yl)-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide were suspended in 100 ml of N,N-dimethyl formamide and 7.6 ml (156 mmol) of hydrazine hydrate were added within 15 min. The reaction mixture was allowed to warm up slowly to 20 temperature of 15 followed by slow addition of 10.5 ml acetic acid (until pH=5.4). The resulting suspension was stirred for 2 hr at 20 additional 2 hr 0 firstly washed with 200 ml of de-ionized water and thereafter with 100 ml of t-butylmethylether. The residue was dried at 40 18 hr. There were obtained 21.7 g (102% of theory) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-2-[2-(hydrazino-imino-methyl)-pyridine-4-yl]-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide with a HPLC purity of 81.4% (w/w).

 

EXAMPLE 7

20 g (37 mmol) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-2-[2-(hydrazino-imino-methyl)-pyridine-4-yl]-5-(2-methoxy-phenoxy)-pyrimidine-4-yl]-amide were added to 160 ml of N,N-dimethyl formamide. To this solution was added dropwise 23 ml of 6 N aqueous hydrochloric acid at a temperature of 15 mmol) of sodium nitrite in 20 ml de-ionized water was added slowly. The reaction mixture was allowed to warm up to 20 for 1.5 hr. Then 160 ml of de-ionized water were added and the suspension was stirred for 1 hr. The solid was filtered off under suction, washed with 100 ml of de-ionized water and dried at 50 hr. There were obtained 18.9 g (92% of theory) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide with a HPLC purity of 89.6% (w/w).

 

EXAMPLE 9

15 g (27 mmol) of 5-methyl-pyridine-2-sulfonic acid [6-chloro-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amide were suspended in 75 ml of ethylene glycol and 6.5 g (163 mmol) of sodium hydroxide were added. The reaction mixture was heated to 85 thereafter 55 ml of 3 N aqueous hydrochloric acid were added dropwise. The suspension was stirred at 20 off under suction, washed with 150 ml of de-ionized water and dried at 70 in 50 ml of N,N-dimethyl formamide and 40 ml of dioxane at 70 Gaseous ammonia was introduced into this solution until pH=9. The resulting suspension was allowed to cool down slowly. The suspension was stirred at 0 washed with 25 ml of dioxane and thereafter with 25 ml of ethanol. Then the solid was dried at 50 ammonium salt (10.4 g, 17.5 mmol) was suspended in 50 ml of methanol and thereafter 6.5 ml (35 mmol) of a 5.4 N sodium methylate solution were added. The solution was heated (reflux) for 3 hr, cooled down slowly to 20 filtering, washed with 10 ml of ice-cold methanol and dried at 70 C., 2000 Pa for 17 hr. There were obtained 6.9 g (41% of theory) of 5-methyl-pyridine-2-sulfonic acid [6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2-[2-(1H-tetrazole-5-yl)-pyridine-4-yl]-pyrimidine-4-yl]-amidesodium salt (1:2) with a HPLC purity of 98.2% (w/w).

 

This application claims benefit to EP 98114978.4 filed Aug. 10, 1998.

 SIMILAR SYNTHESIS OF TEZOSENTAN  AND INTERMEDIATES… AN EXPERT WILL PICK UP NAMES AND INTERMEDIATES… just change the isopropyl gp in vii to methyl

Reaction of 4-cyano-pyridine (I) with Na in methanol followed by treatment with ammonium chloride provides 4-amidino-pyridine hydrochloride (II), which is then converted into 5-(2-methoxyphenoxy)-2-(pyridin-4-yl)-pyrimidine-4,6-diol (IV) by condensation with diethyl malonate derivative (III) by means of Na in MeOH. By heating compound (IV) with phosphorus oxychloride (POCl3), 4,6-dichloro-5-(2-methoxyphenoxy)-2-pyridin-4-yl)pyrimidine (V) is obtained, which in turn is oxidized with peracetic acid in refluxing acetonitrile to afford N-oxide derivative (VI). Condensation of (VI) with 5-isopropylpyridine-2-sulfonamide potassium (VII) furnishes 5-isopropylpyridine-2-sulfonic acid 6-chloro-5-(2-methoxyphenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-yl amide (VIII), which is then dissolved in dimethoxyethane and subjected to reaction with Na in hot ethylene glycol (IX) to provide N-[6-(2-hydroxyethoxy)-5-(2-methoxyphenoxy)-2-(1-oxy-pyridin-4-yl)-pyrimidin-4-yl]-5-isopropylpyridine-2-sulfonamide (X). Refluxing of (X) with trimethylsilylcyanide and Et3N in acetonitrile yields cyano derivative (XI), which is then converted into the tetrazole derivative (XII) by reaction with sodium azide and NH4Cl in DMF at 70 C. Finally, the disodium salt of tezosentan is obtained by treatment of (XII) with Na/MeOH in THF.

………………………………..

SYNTHESIS

WO1996019459A1

Example 29

In analogy to Example 3, from 5-methyl-pyridine-2- sulphonic acid 2-(2-cyano-pyridin-4-yl)-6-(2-hydroxy-ethoxy)- 5-(2-methoxy-phenoxy)-pyrimidin-4-ylamide there is obtained 5-methyl-pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2- methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-ylamide as a white substance of melting point 239- 241 °C from CH3CN

Exgmple, 1 5

a) In analogy to Example l a), from 5-methyl-pyridine-2- sulphonic acid 6-chloro-5-(2-methoxy-phenoxy)-2-(1 -oxy- pyridin-4-yl)-pyrimidin-4-ylamide there is obtained 5-methyl- pyridine-2-sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2-methoxy- phenoxy)-2-(l -oxy-pyridin-4-yl)-pyrimidin-4-ylamide, melting point 188-190°C (from acetonitrile).

b) In analogy to Example 2, from 5-methyl-pyridine-2- sulphonic acid 6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-2- (1 -oxy-pyridin-4-yl)-pyrimidin-4-ylamide there is obtained 5- methyl-pyridine-2-sulphonic acid 2-(2-cyano-pyridin-4-yl)-6- (2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-pyrimidin-4- ylamide

Example 1

a) 200 ml of dimethoxyethane and 1 10.9 g of 4-[4-(4-tert- butyl-phenyl-sulphonylamino)-6-chloro-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide are added all at once to a solution of 23.80 g of sodium in 660 ml of ethylene glycol. The solution is heated at 90°C for 20 hours while stirring, thereafter cooled, poured into 2500 ml of H2O and thereafter treated with CH3COOH to pH 5. The mixture is extracted three times with EtOAc, the organic phase is washed with H2O, dried with Na2Sθ4 and evaporated under reduced pressure. The residue is recrystall- ized from CH3CN and thereafter twice from a mixture of acetone and CH3CN. There is thus obtained 4-[4-(4-tert-butyl-phenyl- sulphonylamino)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide.

Preparation of the starting material:

b) 53.1 g of 4-cyano-pyridine (98%) are added all at once to a solution of 1.15 g of sodium in 200 ml of abs. MeOH. After

6 hours 29.5 g of NH4CI are added while stirring vigorously. The mixture is stirred at room temperature overnight. 600 ml of ether are added thereto, whereupon the precipitate is filtered off under suction and thereafter dried at 50°C under reduced pressure. There is thus obtained 4-amidino-pyridine hydro- chloride (decomposition point 245-247°C).

c) 1 12.9 g of diethyl (2-methoxyphenoxy)malonate are added dropwise within 30 minutes to a solution of 27.60 g of sodium in 400 ml of MeOH. Thereafter, 74.86 g of the amidine hydro- chloride obtained in b) are added all at once. The mixture is stirred at room temperature overnight and evaporated at 50°C under reduced pressure. The residue is treated with 500 ml of ether and filtered off under suction. The filter cake is dissolved in 1000 ml of H2O and treated little by little with 50 ml of CH3COOH. The precipitate is filtered off under suction, washed with 400 ml of H2O and dried at 80°C under reduced pressure. There is thus obtained 5-(2-methoxy-phenoxy)-2-(pyridin-4-yl)- pyrimidine-4,6-diol (or tautomer), melting point above 250°C.

d) A suspension of 1 54.6 g of 5-(2-methoxy-phenoxy)-2- (pyridin-4-yl)-pyrimidine-4,6-diol (or tautomer) in 280 ml of POCI3 is heated at 120°C in an oil bath for 24 hours while stirring vigorously. The reaction mixture changes gradually into a dark brown liquid which is evaporated under reduced pressure and thereafter taken up three times with 500 ml of toluene and evaporated. The residue is dissolved in 1000 ml of CH2CI2, treated with ice and H2O and thereafter adjusted with 3N NaOH until the aqueous phase has pH 8. The organic phase is separated and the aqueous phase is extracted twice with CH2CI2. The combined CH2CI2 extracts are dried with MgSθ4, evaporated to half of the volume, treated with 1000 ml of acetone and the CH2CI2 remaining is distilled off at normal pressure. After standing in a refrigerator for 2 hours the crystals are filtered off under suction and dried at 50°C overnight. There is thus obtained 4,6-dichloro-5-(2-methoxy-phenoxy)-2-pyridin-4-yl)- pyrimidine, melting point 1 78-1 80°C.

e) A solution of 1 7.4 g of 4,6-dichloro-5-(2-methoxy- phenoxy)-2-pyridin-4-yl)-pyrimidine in 100 ml of CH3CN is boiled at reflux for 3 hours with 1 5 ml of a 32% peracetic acid solution, thereafter cooled and stored in a refrigerator overnight. The crystals are filtered off under suction and dried at 50°C under reduced pressure. There is thus obtained 4-[4,6-dichloro- 5-(2-methoxy-phenoxy)-pyrimidin-2-yl]-pyridine 1 -oxide, melting point 189-1 90°C.

 

for analogy

f) A solution of 36.4 g of 4-[4,6-dichloro-5-(2-methoxy- phenoxy)-pyrimidin-2-yl]-pyridine 1 -oxide and 52.8 g of p-tert- butylphenyl-sulphonamide potassium in 1 50 ml of abs. DMF is stirred at room temperature for 24 hours. Thereafter, it is poured into a mixture of 1 500 ml of H2O and 1000 ml of ether while stirring mechanically, whereby a precipitate forms. The suspension is adjusted to pH 5 with CH3COOH, suction filtered, the crystals are washed with cold water and thereafter with ether and dried at 50°C. There is thus obtained 4-[4-(4-tert- butyl-phenylsulphonylamino)-6-chloro-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide as a colourless material of melting point 247-249°C.
Example 2

A solution of 78.45 g of 4-[4-(4-tert-butyl-phenyl- sulphonylamino)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-2-yl]-pyridine 1 -oxide, 122.5 g of trimethylsilyl cyanide, 127.8 g of triethylamine and 1200 ml of CH3CN is boiled at reflux for 20 hours and thereafter evaporated under reduced pressure. The oily residue is taken up in 1000 ml of EtOAc and the solution is washed with CH3COOH:H2θ 9:1 and then with H2O. The EtOAc extracts are dried with Na2SO4. After evaporation of the solvent the residue is taken up in a mixture of CH3CN and CF3COOH (20:1 ), whereby a crystalline precipitate separates. There is thus obtained 4-tert-butyl-N-[2-(2-cyano-pyridin-4- yl)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)-pyrimidin-4- yl]-benzenesulphonamide of melting point 176-1 79°C.

Example 3

A suspension of 50.0 g of 4-tert-butyl-N-[2-(2-cyano- pyridin-4-yl)-6-(2-hydroxy-ethoxy)-5-(2-methoxy-phenoxy)- pyrimidin-4-yl]-benzenesulphonamide, 46.33 g of NH4CI and 56.47 g of NaN3 in 1600 ml of DMF is heated to 70°C for 24 hours while stirring vigorously. The majority of the solvent is distilled off under reduced pressure, the residue is dissolved in H2O, the solution is extracted four times at pH 6.5 with ether, thereafter treated with CH3COOH to pH = 4.5 and extracted with EtOAc. After working up there is obtained a residue which is treated with ether and filtered off under suction therefrom. There is thus obtained 4-tert-butyl-N-[6-(2-hydroxy-ethoxy)-5-(2- methoxy-phenoxy)-2-(2-1 H-tetrazol-5-yl-pyridin-4-yl)- pyrimidin-4-yl]-benzenesulphonamide, melting point 225-227°C.

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

EXTRA INFO

Bosentan (Ro-470203), Atransentan (ABT627), Tezosentan (Ro-610612), Sitaxsentan (TBC-11251), Darusentan (LU-135252), Clazosentan (Ro61-1790, AXV-034343), ZD-4054, Ambrisentan (LU-208075), TAK-044, Avosentan (SPP301), and BQ-123 (Ihara et al Life Sci 1992, 50(4):247-55).

Antagonists of Endothelin type A receptor ETA
Name Structure
BQ-123
Figure US20120202744A1-20120809-C00001
Bosentan
Figure US20120202744A1-20120809-C00002
Atrasentan
Figure US20120202744A1-20120809-C00003
Tezosentan
Figure US20120202744A1-20120809-C00004
Sitaxsentan
Figure US20120202744A1-20120809-C00005
Darusentan
Figure US20120202744A1-20120809-C00006
Clazosentan
Figure US20120202744A1-20120809-C00007
ZD-4054 (Zibotentan)
Figure US20120202744A1-20120809-C00008
Ambrisentan
Figure US20120202744A1-20120809-C00009
Tak-044
Figure US20120202744A1-20120809-C00010
Avosentan
Figure US20120202744A1-20120809-C00011

 

 

Share

ARAB MEDICINE- REVIEW

 Arab medicine review  Comments Off on ARAB MEDICINE- REVIEW
Jan 212014
 

ARAB MEDICINE- REVIEW

In the history of medicine, Islamic medicineArabic medicineGreco-Arabic and Greco-Islamic refer to medicine developed in the Islamic Golden Age, and written in Arabic, the lingua franca of Islamic civilization. The emergence of Islamic medicine came about through the interactions of the indigenous Arab tradition with foreign influences.Translation of earlier texts was a fundamental building block in the formation of Islamic medicine and the tradition that has been passed down.

Latin translations of Arabic medical works had a significant influence on the development of medicine in the high Middle Ages and early Renaissance, as did Arabic texts which translated the medical works of earlier cultures.

In the early Islamic and Mack’s period (661–750 AD), Muslims believed that Allah provided a treatment for every illness.Around the ninth century, the Islamic medical community began to develop and utilize a system of medicine based on scientific analysis. The importance of the health sciences to society was emphasized, and the early Muslim medical community strived to find ways to care for the health of the human body. Medieval Islam developed hospitals, expanded the practice of surgery.

Important medical thinkers and physicians of Islam were Al-Razi and Ibn Sina. Their knowledge on medicine was recorded in books that were influential in medical schools throughout Muslim history, and Ibn Sina in particular (under his Latinized name Avicenna) was also influential on the physicians of later medieval Europe. Throughout the medieval Islamic world, medicine was included under the umbrella of natural philosophy, due to the continued influence of the Hippocratic Corpus and the ideas of Aristotle and Galen. The Hippocratic Corpus was a collection of medical treatises attributed to the famous Greek physician Hippocrates of Cos (although it was actually composed by different generations of authors). The Corpus included a number of treatises which greatly influenced medieval Islamic medical literature

The first encyclopedia of medicine in Arabic language] was Persian scientist Ali ibn Sahl Rabban al-Tabari‘s Firdous al-Hikmah(“Paradise of Wisdom”), written in seven parts, c. 860. Al-Tabari, a pioneer in the field of child development, emphasized strong ties between psychology and medicine, and the need for psychotherapy and counseling in the therapeutic treatment of patients. His encyclopedia also discussed the influence of Sushruta and Chanakya on medicine, including psychotherapy

Medical contributions made by Medieval Islam not only involved the development and expansion of the human anatomy, but also included the use of plants as a type of remedy or medicine. Medieval Islamic physicians used natural substances as a source of medicinal drugs—including Papaver somniferum Linnaeus, poppy, and Cannabis sativa Linnaeus, hemp. In pre-Islamic Arabia, neither poppy nor hemp was known. Hemp was introduced into the Islamic countries in the ninth century from India through Persia and Greek culture and medical literature. Dioscorides, who according to the Arabs is the greatest botanist of antiquity, recommended hemp’s seeds to “quench geniture” and its juice for earaches.[27] Beginning in 800 and lasting for over two centuries, poppy use was restricted to the therapeutic realm. However, the dosages often exceeded medical need and was used repeatedly despite what was originally recommended. Poppy was prescribed by Yuhanna b. Masawayh to relieve pain from attacks of gallbladder stones, for fevers,indigestion, eye, head and tooth aches, pleurisy, and to induce sleep. Although poppy had medicinal benefits, Ali al-Tabari explained that the extract of poppy leaves was lethal, and that the extracts and opium should be considered poisons

The way early Arab medicine developed should be contrasted to how medicine evolved in Christianity up until the Renaissance. While Christian Rome and Byzantium inherited the rich Graeco-Roman medical legacy of thinkers like Hippocrates and Galen, after the fall of Rome in 476, Dark Age Europe increasingly tended towards a fatalistic view of suffering and disease, further tempered by superstition about curses and God’s punishment for man’s sins sent down in the form of disease and affliction.

Many historians point to the explicit tradition of fact-based, scientific medicine as articulated by the Prophet himself (pbuh). First, the concept of ‘sinful’ mankind seems not as strong in Islam as in early Christianity. Disease was seen by Arabs and other Muslims as one more problem to be solved, not a curse from God or a trial to be endured so one would be assured of entering Paradise.

Consider these statements on health and medicine attributed to the Prophet (pbuh):

“There is no disease that Allah has created, except that He also has created its treatment.”

“Make use of medical treatment, for Allah has not made a disease without appointing a remedy for it, with the exception of one disease, namely old age.”

The Prophet (pbuh) was also credited with articulating several specific medical treatments, including the use of honey, cupping, and cauterisation. He spoke about the contagious nature of leprosy, sexually transmitted disease, and the animal disease known as the mange. But most importantly, whereas other societies usually stigmatised and feared the sick and afflicted, at best isolating them and at worst leaving them somewhere to die, the Prophet (pbuh) and early Islam had a very compassionate and forgiving view of the sick.

As in other fields, the earliest Arab-Muslim medical efforts were devoted to translating the medical wisdom of older civilisations, beginning in the late 700s in Baghdad with the works of the Roman physician Galen as well as advanced medical writings from Persia, including the great pre-Islamic medical centre at Gundishapur.

Gundishapur is credited with having developed the first truly modern hospital, where patients actually went to be healed and cured, rather than prayed over as they suffered a slow and inevitable death as in Dark Age Europe.

The first major Arab-Muslim healer was the chemist Al Razi, who turned to medicine at about age 30, perhaps to find cures for his injuries suffered during alchemical experiments, especially eye ailments. His first inspiration was the Roman physician Galen.

Galen had pushed Roman medical knowledge as far as it could go in that time, undertaking innumerable vivisections of live animals to see how their organs functioned, as well as dissections of human cadavers.

Al Razi was especially troubled by Galen’s theory of the humours, which just didn’t hold up to examination. There seemed a lot more going on inside the human body than those four humours. And so he would write around 865:

“I prayed to God to direct and lead me to the truth in writing this book. It grieves me to oppose and criticise the man Galen from whose sea of knowledge I have drawn much. Indeed, he is the Master and I am the disciple. Although this reverence and appreciation will and should not prevent me from doubting, as I did, what is erroneous in his theories. I imagine and feel deeply in my heart that Galen has chosen me to undertake this task, and if he were alive, he would have congratulated me on what I am doing. I say this because Galen’s aim was to seek and find the truth and bring light out of darkness. I wish indeed he were alive to read what I have published.”

Al Razi would write as many as 184 papers and articles on subjects ranging from his doubts about Galen to the first known distinction between smallpox and measles, the discovery of allergic asthma, the discovery of fever as the body’s defence mechanism, medical ethics, using opium as a treatment for depression, the first medical handbook for common people, and paediatrics.

Al Razi would also theorise about the connection of the soul and state of mind to the physical health of the body, suggesting that someone with mental and emotional disturbances would be more vulnerable to infection and chronic ailments.

Al Razi’s medical insights would be translated into Latin several centuries after his death. By the late 1200s, mediaeval Europeans were beginning to stir out of their long Dark Age sleep and for a century were captivated by the writings of Al Razi – who by then had been given the Latin name Rhazes.

About eight decades after Al Razi, a brilliant healer named Al Zahrawi laid the foundation of modern surgery while working in the Umayyad imperial compound outside Cordoba.

Because all records were destroyed in the civil wars that marked the end of the Umayyad reign in Spain, hardly any facts about Al Zahrawi’s personal life remain. What does survive is his 30-chapter Kitab al Tasrif, a compendium of this man’s medical knowledge and genius. A century and a half after his death, it would be translated into Latin and have even more impact than the work of Rhazes. Al Zahrawi’s Latin name was Albucasis.

His discoveries would continue to resonate into the 21st century, first for his invention of about 200 medical instruments, many of which are still in use – such as the obstetrical forceps, scalpel, surgical needle, surgical retractor, specula, and the use of catgut for internal suturing. But he was also exceptional for innovating surgical procedures like mastectomies, orthodontia, repairing fractures, and using ligature for suturing arteries instead of cauterising them.

Another Muslim healer would follow in the Arabic tradition and even eclipse the great Al Zahrawi, this one a Persian working exclusively in Persia. This man was Ibn Sina. Europe and the Arab world would come to know him as Avicenna, the Prince of Medicine, and the single most important influence on Islamic and Western medicine for about 500 years.

Ibn Sina was consummately gifted. He is reputed to have memorised the Qur’an by age 10, Aristotle’s Metaphysics several years later (he claimed to have read it 40 times), and had become a practising physician by age 16.

Ibn Sina’s greatest motivation was his burning intellectual curiosity for the world, and the world beyond, not social status or financial security. By the age of 20, he had turned down his ruler’s offer to become court physician, preferring only the right to study as much as he wanted in the ruler’s royal library.

A political upheaval overthrew the ruler and Ibn Sina began a long life of wandering Persia in search of a secure patron who would allow him to indulge in his medical and scientific research. Unfortunately, political instability and Ibn Sina’s harshly arrogant manner meant he was constantly changing jobs.

But despite his unending struggle, he was able to gradually systemise Islamic understanding of the medical sciences in such a way that not only was the Arab and Islamic world forever indebted, so also was Europe and the West.

Although Ibn Sina is credited with writing as many as 450 papers and books in a dozen fields, the work that continued to resonate most powerfully was his Canon of Medicine written around 1025, a 14-volume work that was for 500 years Europe’s most influential medical source book. The Canon was a combination both of the collected medical wisdom of other writers as well as his own observations and research. Although it provided a window into forgotten Greek medicine, its greatest value was in the modernistic approach it took to a field riddled with false theory and ignorance.

It could be argued that Ibn Sina was the first to formally explain the experimental method in medicine, the spread of contagious diseases, the use of quarantine, clinical trials, psychiatry, and psychotherapy. He also seems to be the first to show that tuberculosis was a contagious disease, as well as to identify diabetes.

According to some sources, the Canon was the first documented explanation of modern medical methods like the randomised clinical trial, and the first modern set of comprehensive rules for testing new drugs.

His deeper research into the mind-body connection, and the mental or spiritual source of physical ailments, was built on the first intuitive work of men like Al Razi. But Ibn Sina went further, beginning the first documented forays into what we today would call psychotherapy, 900 years before Sigmund Freud.

One account says that a young man had come to him with a condition that looked very much like consumption. He was literally wasting away. But Ibn Sina could find no signs of a cancer or other disease that would indicate some physical explanation.

He conducted a series of interviews or conversations with the young man. As Ibn Sina spoke certain key words and phrases, he was also checking the man’s pulse and found it became elevated around certain terms. Thus it gradually emerged that the patient was in love with a woman back in his home village. For whatever reason he had never expressed this to her, and the unfulfilled desire was sapping him of his energy.

Ibn Sina gradually concluded that the source of the young man’s physical condition was his unexpressed love. He suggested that the patient go to the object of his affections and profess his love to her. The young man did this, the girl agreed to marry him, and the patient swiftly recovered his vitality.

As far as we know this was the earliest documented account of the use of word association in psychoanalysis, which modern medicine credits to Carl Jung 900 years later.

While medical thinkers like Al Razi, Al Zahrawi and Ibn Sina are closely tied to their innovations through their writings, many of the great breakthroughs of Arab medicine were collective undertakings and are difficult to identify with any single author or inventor.

This is particularly true with key Arab-Muslim institutions like the modern insane asylum, the public hospital, free medical care, and the pharmacy. The modern hospital itself was not an Arab invention, but Arabs and their partners made it a public institution and spread it around the world.

Isolated healing temples and places for the sick had existed in many older cultures including around the Mediterranean and across Asia. But with few exceptions they were unable to offer real cures in the modern sense. Often their method was a mixture of magic or religion with means of making one feel better, if only briefly.

But in 6th century pre-Islamic Persia, a true hospital called a bimaristan or ‘sick place’ was built in the city of Gundishapur, complete with surgery, pharmacy, and outpatient treatments. This came to the attention of the Arabs, in particular Caliphs Harun Al Rashid and his half-Persian son Al Mamun, and they set about replicating these institutions across their realm.

Harun invited a doctor from the bimaristan in Gundishapur to open the first bimaristan in Baghdad. Al Razi was later commissioned with overseeing the Audidi Hospital in Baghdad, in the mid 800s. He applied his evolving understanding of sanitation and infection to find the best location possible. He hung raw meat in various parts of the city to see comparative rates of decay, and where the meat lasted longest, there he put the hospital.

Audidi had more than two dozen doctors including surgeons, eye specialists, and physiologists.

By the year 1000, Baghdad alone would number five public hospitals when there were none in all of Europe. Hospitals would also be found in Cairo, Damascus, Aleppo, North Africa, and Al Andalus. These centres would offer surgery, outpatient clinics, mental wards, convalescent centres, and even nursing homes.

One of the greatest hospitals would be Al Mansuri in Cairo, which was reported to have as many as 8,000 beds and annual revenues of one million dirhams. Al Mansuri was a true public hospital because it was charged with offering treatment to anyone, rich or poor, including the indigent who could not pay at all.

The Arab establishment of humane mental wards and insane asylums was especially futuristic and important. The Arab world, in line with the teachings of the Prophet (pbuh) and others, never stigmatised the mentally afflicted, seeing mental illness as one more disease that might be cured. Europe and the West did not develop a modern non-judgmental view of mental illness until the 19th and 20th centuries.

Arab pharmacies were another important invention. Although other cultures offered various potions and herbs for sale, it was rare to find cures that really worked. People were just as inclined to faith healing and magic as to ‘healing’ substances, because they were all equally ineffective. But the evolution of modern evidence-based pharmacology under thinkers like Al Razi, Al Kindi and Ibn Sina created a new class of substances that really worked.

Arab pharmacies were known as saydala, and the first one seems to have been at Harun al Rashid’s hospital in Baghdad built in the late 700s. Within half a century saydala were spreading throughout the caliphate. These remedies were often fabricated right on the spot at in-house laboratories. More importantly, they were overseen by government inspectors to make sure they were pure, not out of date, measured in verified scales, and correctly identified.

Al Razi would even introduce the concept of generic drugs for the poor, while Al Kindi would also seek to identify cheaper alternative treatments for those who could not afford expensive drugs.

The same kind of modern pharmacies selling remedies that really worked would only begin to appear in Italy in about the 12th century, fuelled largely by the growing trade between Arabs and Europeans.

READ A GREAT ARTICLE AT

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1297506/

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1475945/

Aqrabadhin of Al-Kindi. Translated by Martin Levey. Madison: The University of Wisconsin Press, 1966.

Kamal, Hassan. Encyclopedia of Islamic Medicine. Cairo: General Egyptian Book Organization, 1975.

Levey, Martin. Early Arabic Pharmacology. Leiden, Netherlands: E. J. Brill, 1973.

Savage-Smith, Emilie. Islamic Culture and the Medical Arts. Bethesda, Md.: National Library of Medicine, 1994.

Siddiqi, Muhammad Zubayr. Studies in Arabic and Persian Medical Literature. Calcutta: Calcutta University Press, 1959.

Usama, Ibn Shuraik. Sunna Abu-Dawud, Book 28, No. 3846 (part of the hadith, a narrative record of the sayings of Mohammed and his companions).

Share

Oncolytic Drugs …Preparation of (4-{4-[({3-tert-butyl-1-[3-(hydroxymethyl)phenyl]-1H-pyrazol-5-yl}carbamoyl)amino]-3-fluorophenoxy}-N-methylpyridine-2-carboxamide)

 cancer  Comments Off on Oncolytic Drugs …Preparation of (4-{4-[({3-tert-butyl-1-[3-(hydroxymethyl)phenyl]-1H-pyrazol-5-yl}carbamoyl)amino]-3-fluorophenoxy}-N-methylpyridine-2-carboxamide)
Jan 212014
 


Patents–  EP2111401B1

1036712-77-2 cas NO

see also WO 2008079968 BAYER

VEGFR-2 (FLK-1/KDR) Inhibitors
Bcr-Abl Kinase Inhibitors
HGFR (MET; c-Met) Inhibitors 

Inhibitors of protein kinases, such as wild-type and mutations of Bcr-Abl, Flk1, c-Met, expected to be useful for the treatment of hyperproliferative and/or angiogenesis disorders such as cancer. A representative compound suppressed Flk-1, c-Met and wild type and T135I mutant Bcr-Abl enzymes with IC50 values below 1 mcM. Compound also inhibited the proliferation of K562 (IC50 = 1.58 nM) and BAF3 cells expressing wild-type and T315I, E255K, M351T and Y253F mutant Brc-Abl enzymes (IC50 = 3.84, 34.1, 503, 811 and 564 nM, respectively).


Example 1HYDROXY METHYL PHENYL PYRAZOLYL UREA (4-{4-[({3-tert-Butyl-1-[3-(hydroxymethyl)phenyl]-1H-pyrazol-5-yl}carbamoyl)amino]-3-fluorophenoxy}-N-methylpyridine-2-carboxamide)

    • Figure imgb0001

HYDROXY METHYL PHENYL PYRAZOLYL UREAStep 1. Preparation of ethyl 3-(5-amino-3-tert-butyl-1H-pyrazol-1-yl)benzoate

    • Figure imgb0002
    • Sulfuric acid (concentrated, 15.7 mL, 295.7 mmol) was carefully added drop-wise to cold EtOH (600 mL) with stirring. To this, 3-hydrazinobenzoic acid (45 g, 295.7 mmol) and 4,4-dimethyl-3-oxopentanenitrile (40.7 g, 325.3 mmol) were added and then the mixture was heated at 90°C for 48 h. Most of the solvent was evaporated at reduced pressure, and the residual mixture was diluted with ethyl acetate. The resulting mixture was washed with ice cold 2M NaOH followed by brine, and dried (Na2SO4). The solution was filtered through a bed of silica gel, washing with more ethyl acetate. Evaporation of ethyl acetate and treatment of the residue with dichloromethane/hexanes gave the product as an off-white crystalline solid (61 g, 71%). MS mlz 288.2 (M+H)+; calcd. mass 287. Retention time (LC-MS): 2.99 min. 1H-NMR (DMSO-d6): δ 8.16 (m 1H); 7.88 (m, 2H); 7.60 (t, 1H); 5.40 (s, 1H); 5.32 (s, 2H); 4.36 (q, 2H); 1.34 (t, 3H); 1.21 (s, 9H).

Step 2. Preparation of ethyl 3-{3-tert-butyl-5-[(phenoxycarbonyl)amino]-1H-pyrazol-1-yl}-benzoate

    • Figure imgb0003
    • To a mixture of ethyl 3-(5-amino-3-tert-butyl-1H-pyrazol-1-yl)benzoate (60 g, 208.8 mmol) and K2CO3 (86.6 g, 626.4 mmol) in THF (1400 mL) was added phenyl chloroformate (98.1 g, 626.4 mmol). The reaction was stirred at room temperature overnight. The solid was removed by filtration and most of the solvent was evaporated under reduced pressure. The residual mixture was dissolved in EtOAc and washed with brine, then water. The organic layer was then dried and concentrated. The crude product was purified by recrystallization from CH2Cl2/hexanes to give the desired product as a white powder (78.5 g, 92%). MS m/z 408.1 (M+H)+; calcd. mass 407. Retention time (LC-MS): 3.92 min. 1H-NMR (DMSO-d6): δ 10.19 (s, broad, 1H); 8.11 (m 1H); 7.97 (d, J = 7.6 Hz, 1H); 7.86 (m, 1H); 7.71 (t, 1H); 7.38 (m, 2H); 7.24 (m, 1H); 7.08 (m, 1H); 6.40 (s, 1H); 4.38 (q, 2H); 1.32 (t, 3H); 1.29 (s, 9H).

Step 3. Preparation of ethyl 3-(3-tert-butyl-5-{[(2-fluoro-4-{[2-(methylcarbamoyl)pyridin-4-yl]-oxy}phenyl)carbamoyl]amino}-1H-pyrazol-1-yl)benzoate

    • Figure imgb0004
    • A solution of ethyl 3-{3-tert-butyl-5-[(phenoxycarbonyl)amino]-1H-pyrazol-1-yl}benzoate (9.36 g, 22.0 mmol), 4-(4-amino-3-fluorophenoxy)-N-methylpyridine-2-carboxamide (5.0 g, 19.1 mmol; prepared as described in Dumas et al., PCT Int. Appl. WO 2004078748 (2004 )) and triethyl amine (3.87 g, 38.3 mmol) in anhydrous THF (100 mL) was stirred at room temperature overnight. The crude product was purified by column chromatography (CH2Cl2 plus 1% to 3% of 2M NH3 in MeOH), followed by recrystallization from EtOAc/hexanes to give the desired product as an off-white crystalline solid (6.32 g, 57%). MS m/z 575.1 (M+H)+; calcd. mass 574. Retention time (LC-MS): 3.75 min.1H-NMR (DMSO-d6): δ 8.97 (m, 1H); 8.89 (m, 1H); 8.80 (m, 1H); 8.52 (d, J = 5.6 Hz, 1H); 8.16 (t, 1H); 8.06 (m, 1H); 7.99 (m, 1H); 7.85 (m, 1H); 7.71 (t, 1H); 7.39 (m, 1H); 7.33 (m, 1H); 7.17 (m, 1H); 7.06 (m, 1H); 6.42 (s, 1H); 4.36 (q, 2H); 2.78 (d, J = 5.2 Hz, 3H); 1.31 (m, 12H).

Step 4. Preparation of (4-{4-[({3-tert-butyl-1-[3-(hydroxymethyl)phenyl]-1H-pyrazol-5-yl}carbamoyl)amino]-3-fluorophenoxy}-N-methylpyridine-2-carboxamide)

  • Figure imgb0005
  • To a well-stirred cooled solution of 4-(4-{3-[5-tert-butyl-2-(3-ethoxycarbonyl-phenyl)-2H-pyrazol-3-yl]-ureido}-3-fluoro-phenoxy)-pyridine-2-carboxylic acid methylamide (56 mg, 0.1 mmol) in ethanol (10 mL), NaBH4 (50 mg) was added in portions. After 14 h, ice water (10 mL) was carefully added to the reaction mixture. Then, most of the ethanol was evaporated under reduced pressure. The residual mixture was treated with saturated aqueous ammonium chloride solution (10 mL) and extracted three times with dichloromethane (50, 25, and 25 mL). The combined dichloromethane extract was dried (sodium sulfate) and the solvent was evaporated. The crude product was purified by preparative thin layer chromatography on silica gel using 3-5% 2M ammonia in methanol in dichloromethane as the eluent to yield the desired product as a white powder (31 mg, 58%).
    For a larger scale synthesis, the following similar procedure was followed: To a solution of ethyl 3-(3-tert-butyl-5-{[(2-fluoro-4-{[2-(methylcarbamoyl)pyridin-4-yl]oxy}phenyl)carbamoyl]-amino}-1H-pyrazol-1-yl)benzoate (11.2 g, 19.5 mmol) in EtOH was added NaBH4 stepwise as a solid. The reaction was then stirred at room temperature overnight, and then quenched by gradual addition of aqueous NH4Cl. The mixture was diluted with EtOAc, washed with aq. NH4Cl, followed by brine. The organic layer was then dried and concentrated. The crude product was then purified by column chromatography on silica gel (CH2Cl2 plus 1 to 5% of 2M NH3 in MeOH), followed by recrystallization from dichloromethane/hexanes to give the desired product as a white crystalline solid (8.0 g, 77%). Mp 160 ºC; after further recrystallization, desired product was obtained with mp 196 ºC.
  •  MS m/z 533.3 (M+H)+; calcd. mass 532. Retention time (LC-MS): 3.13 min.
  •  1H-NMR (DMSO-d6): δ 9.02 (s, broad, 1H); 8.87 (s, 1H); 8.81 (m, 1H); 8.52 (d, J= 5.2 Hz, 1H); 8.21 (t, 1H); 7.51 (m, 2H); 7.39 (m, 3H); 7.32 (m, 1H); 7.17 (m, 1H); 7.06 (m, 1H); 6.40 (s, 1H); 5.36 (t, 1H); 4.59 (d, J = 5.6 Hz, 2H); 2.78 (d, J = 4.8 Hz, 3H); 1.27 (s, 9H).
  • Elemental Analysis: C 62.92%; H 5.43%; N 15.70%; calcd. C 63.15%; H 5.49%; N 15.78%.
Share

JAPAN…..Takeda wins Japanese OK for Adcetris

 japan  Comments Off on JAPAN…..Takeda wins Japanese OK for Adcetris
Jan 202014
 

Takeda wins Japanese OK for Adcetris

Takeda’s Adcetris (brentuximab vedotin) has been given the regulatory go-head in Japan to treat malignant lymphoma.

More specifically, the Japanese Ministry of Health, Labour and Welfare (MHLW) has issued a greenlight for its use in patients with CD30 Positive Relapsed or Refractory Hodgkin Lymphoma (HL) or Relapsed or Refractory Anaplastic Large Cell Lymphoma (ALCL).

Read more at: http://www.pharmatimes.com/Article/14-01-18/Takeda_wins_Japanese_OK_for_Adcetris.aspx#ixzz2qwGAi5Qo

Share

Valspodar, PSC-833

 Phase 3 drug, Uncategorized  Comments Off on Valspodar, PSC-833
Jan 202014
 

PSC833(Valspodar)

Valspodar, SDZ-PSC-833, PSC-833, Amdray

P-Glycoprotein (MDR-1; ABCB1) Inhibitors , Multidrug Resistance Modulators

Valspodar is a cyclosporine derivative and a P-glycoprotein inhibitor currently in phase III clinical trials at the National Cancer Institute (NCI) in combination with chemotherapy for the treatment of leukemia. The drug was also being developed in combination with chemotherapy for the treatment of various other types of cancers, however, no recent developments on these trials have been reported.

P-glycoprotein is an ABC-transporter protein that has been implicated in conferring multidrug resistance to tumor cells. In previous trials, valspodar was associated with greater disease-free and overall survival in younger patients (45 years or below), and was shown to significantly increase the cellular uptake of daunorubicin in leukemic blast cells in vivo. However, in a phase III trial examining the drug candidate’s effects on AML in patients at least 60 years of age, valspodar was associated with excessive mortality and complete remission rates were higher in groups not treated with the compound.

Nonimmunosuppressive cyclosporin analog which is a potent multidrug resistance modifier; 7-10 fold more potent than cyclosporin A; a potent P glycoprotein inhibitor; MW 1215.

M.Wt: 1214.62
Formula: C63H111N11O12

CAS : 121584-18-7

IUPAC/Chemical name: 

(3S,6S,9S,12R,15S,18S,21S,24S,30S,33S)-6,9,18,24-tetraisobutyl-3,21,30-triisopropyl-1,4,7,10,12,15,19,25,28-nonamethyl-33-((R,E)-2-methylhex-4-enoyl)-1,4,7,10,13,16,19,22,25,28,31-undecaazacyclotritriacontan-2,5,8,11,14,17,20,23,26,29,32-undecaone

6 – [(2S, 4R, 6E)-4-Methyl-2-(methylamino)-3-oxo-6-octenoic acid]-7-L-valine-cyclosporin A; Cyclo [[(2S, 4R, 6E) -4-methyl-2-(methylamino)-3-oxo-6-octenoyl]-L-valyl-N-methylglycyl-N-methyl-L-leucyl-L-valyl-N-methyl-L-leucyl-L- alanyl-D-alanyl-N-methyl-L-leucyl-Nm

[3′-oxo-4-butenyl-4-methyl-Thr1]-[Val2]-cyclosporine

Novartis (Originator), National Cancer Institute (Codevelopment)
Modulators of the Therapeutic Activity of Antineoplastic Agents, Multidrug Resistance Modulators, ONCOLYTIC DRUGS, P-Glycoprotein (MDR-1) Inhibitors
Phase III

Clinical trials

http://clinicaltrials.gov/search/intervention=psc+833

Synonyms

  • 3′-Keto-bmt(1)-val(2)-cyclosporin A
  • Amdray
  • Psc 833
  • PSC-833
  • PSC833
  • SDZ PSC 833
  • Sdz-psc-833
  • UNII-Q7ZP55KF3X
  • Valspodar

Valspodar or PSC833 is an experimental cancer treatment and chemosensitizer drug.[1] It is a derivative of ciclosporin D.

Its primary use is that of a p-glycoprotein inhibitor. Previous studies in animal models have found it to be effective at preventing cancer cell resistance to chemotherapeutics, but these findings did not translate to clinical success.[2]
Valspodar, also known as PSC-833 is an analogue of cyclosporin-A. Valspodar inhibits p-glycoprotein, the multidrug resistance efflux pump, thereby restoring the retention and activity of some drugs in some drug-resistant tumor cells. This agent also induces caspase-mediated apoptosis.
PSC-833 is a non-immunosuppressive cyclosporin derivative that potently and specifically inhibits P-gp.  In vitro experiments indicate that PSC-833interacts directly with P-gp with high affinity and probably interferes with the ATPase activity of P-gp. Studies in multidrug resistant tumor models confirm P-gp as the in vivo target of PSC-833 and demonstrate the ability of PSC-833 to reverse MDR leukemias and solid tumors in mice. Presently,PSC-833 is being evaluated in the clinic.

Valspodar can cause nerve damage.[1]

Valspodar

Synthesis By oxidation of cyclosporin D (I) with N-chlorosuccinimide and dimethylsulfide in toluene (1) Scheme 1 Description alpha (20, D) -..?. 255.1 (c 0.5, CHCl3) Manufacturer Sandoz Pharmaceuticals Corp (US).. . References 1 Bollinger, P., B flounder sterli, JJ, Borel, J.-F., Krieger, M., Payne, TG, Traber, RP, Wenger, R. (Sandoz AG; Sandoz Patent GmbH; Sandoz Erfindungen VmbH ). Cyclosporins and their use as pharmaceuticals.

AU 8817679, EP 296122, JP 89045396. AU 8817679; EP 0296122; JP 1989045396; JP 1996048696; US 5525590

……………………………..

 

  • The cyclosporins comprise a class of structurally distinctive, cyclic, poly-N-methylated undecapeptides, generally possessing pharmacological, in particular immunosuppressive, anti-­inflammatory and/or anti-parasitic activity, each to a greater or lesser degree. The first of the cyclosproins to be isolated was the naturally occurring fungal metabolite Ciclosporin or Cyclo­sporine, also known as cyclosporin A and now commercially available under the Registered Trade Mark SANDIMMUN®. Ciclosporin is the cyclosporin of formula A

    Figure imgb0001

    wherein -MeBmt- represents the N-methyl-(4R)-4-but-2E-­en-1-yl-4-methyl-(L)threonyl residue of formula B

    Figure imgb0002

    in which -x-y- is trans -CH=CH- and the positive 2′, 3′ and 4′ have the configuration S, R and R respectively.

  • Since the original discovery of Ciclosporin, a wide variety of naturally occurring cyclosporins have been isolated and identified and many further non-natural cyclosporins have been prepared by total- or semi-synthetic means or by the application of modified culture techniques. The class comprised by the cyclosporins is thus now substantial and includes, for example, the naturally occurring cyclosporins A through Z [c.f. Traber et al. 1, Helv. Chim. Acta, 60, 1247-1255 (1977); Traber et al. 2, Helv. Chim. Acta, 65, 1655-1667 (1982); Kobel et al., Europ. J. Applied Microbiology and Biotechnology 14, 273-240 (1982); and von Wartburg et al. Progress in Allergy, 38, 28-45 (1986)], as well as various non-natural cyclosporin derivatives and artificial or synthetic cyclosporins including the dihydro- and iso-cyclosporins [in which the moiety -x-y- of the -MeBmt- residue (Formula B above) is saturated to give -x-y- = -CH₂-CH₂- / the linkage of the residue -MeBmt- to the residue at the 11-position of the cyclosporin molecule (Formula A above) is via the 3′-O-atom rather than the α-N-atom]; derivatised cyclosporins (e.g. in which the 3′-O-atom of the -MeBmt- residue is acylated or a further substituent is introduced at the α-carbon atom of the sarcosyl residue at the 3-position); cyclosporins in which the -MeBmt- residue is present in isomeric form (e.g. in which the configuration across positions 6′ and 7′ of the -MeBmt- residue is cis rather than trans); and cyclosporins wherein variant amino acids are incorporated at specific positions within the peptide sequence employing e.g. the total synthetic method for the production of cyclosporins developed by R. Wenger – see e.g. Traber et al. 1, Traber et al. 2 and Kobel et al. loc. cit.; U.S. Patents Nos 4 108 985, 4 210 581, 4 220 641, 4 288 431, 4 554 351 and 4 396 542; European Patent Publications Nos. 0 034 567 and 0 056 782; International Patent Publication No. WO 86/02080; Wenger 1, Transpl. Proc. 15, Suppl. 1:2230 (1983); Wenger 2, Angew. Chem. Int. Ed., 24, 77 (1985); and Wenger 3, Progress in the Chemistry of Organic Natural Products 50, 123 (1986).
  • The class comprised by the cyclosporins is thus now very large indeed and includes, for example [Thr]²-, [Val]²-, [Nva]²- and [Nva]²-[Nva]⁵-Ciclosporin (also known as cyclosporins C, D, G and M respectively), [3-O-acetyl-MeBmt]¹-Ciclosporin (also known as cyclosporin A acetate), [Dihydro-MeBmt]¹-[Val]²-Ciclosporin (also known as dihydro-cyclosporin D), [Iso-MeBmt]¹-[Nva]²-Ciclosporin (also known as isocyclosporin G), [(D)Ser]⁸-Ciclosporin, [MeIle]¹¹-Ciclosporin, [(D)MeVal]¹¹-Ciclosporin (also known as cyclosporin H), [MeAla]⁶-Ciclosporin, [(D)Pro]³-Ciclosporin and so on.
  • [In accordance with conventional nomenclature for cyclosporins, these are defined throughout the present specification and claims by reference to the structure of Ciclosporin (i.e. Cyclosporin A). This is done by first indicating the amino acid residues present which differ from those present in Ciclosporin (e.g. “[(D)Pro]³” to indicate that the cyclosporin in question has a -(D)Pro- rather than -Sar- residue at the 3-position) and then applying the term “Ciclosporin” to characterise remaining residues which are identical to those present in Ciclosporin.
  • The residue -MeBmt- at position 1 in Ciclosporin was unknown before the discovery of the cyclosporins. This residue and variants or modifications of it, e.g. as described below, are thus generally characteristic of the cyclosporins. In general, variants or alternatives to [MeBmt]¹ are defined by reference to the -MeBmt- structure. Thus for dihydrocyclosporins in which the moiety -x-y- (see formula B above) is reduced to -CH₂-CH₂-, the residue at the 1-position is defined as “-dihydro-MeBmt-“. Where the configuration across the moiety -x-y- is cis rather than trans, the resulting residue is defined as “-cis-MeBmt-“.
  • Where portions of the -MeBmt- residue are deleted, this is indicated by defining the position of the deletion, employing the qualifier “des” to indicate deletion, and then defining the group or atom omitted, prior to the determinant “-MeBmt-“, “-dihydro-MeBmt-“, “-cis-MeBmt-” etc.. Thus “-N-desmethyl-MeBmt-“, “-3′-desoxy-MeBmt-“, and “-3′-desoxy-4′-desmethyl-MeBmt-” are the residues of Formula B¹, B² and B³ respectively:

    Figure imgb0003

    B¹ – X = CH₃, Y = OH, Z = H.
    B² – X = CH₃, Y = H, Z = CH₃.
    B³ – X = H, Y = H, Z = CH₃.

  • Where positions or groups, e.g. in -MeBmt-, are substituted this is represented in conventional manner by defining the position and nature of the substitution. Thus -3′-O-acetyl-MeBmt- is the resi­due of formula B in which the 3′-OH group is acetylated (3′-O­-COCH₃). Where substituents of groups, in e.g. -MeBmt-, are replaced, this is done by i) indicating the position of the re­placed group by “des-terminology” as described above and ii) de­fining the replacing group. Thus -7′-desmethyl-7′-phenyl-MeBmt- is the residue of formula B above in which the terminal (8′) methyl group is replaced by phenyl. 3′-Desoxy-3′-oxo-MeBmt- is the resi­due of formula B above in which the 3′-OH group is replaced by =O.
  • In addition, amino acid residues referred to by abbreviation, e.g. -Ala-, -MeVal-, -αAbu- etc… are, in accordance with conventional practice, to be understood as having the (L)-configuration unless otherwise indicated, e.g. as in the case of “-(D)Ala-“. Residue abbreviations preceded by “Me” as in the case of “-MeLeu-“, represent α-N-methylated residues. Individual residues of the cyclosporin molecule are numbered, as in the art, clockwise and starting with the residue -MeBmt-, -dihydro-MeBmt- etc. … in position 1. The same numerical sequence is employed throughout the present specification and claims.]
  • [0010]
    Because of their unique pharmaceutical potential, the cyclosporins have attracted very considerable attention, not only in medical and academic circles, but also in the lay press. Cyclo­sporin itself is now commonly employed in the prevention of rejection following allogenic organ, e.g. heart, heart-lung, kidney and bone-marrow transplant, as well as, more recently, in the treatment of various auto-immune and related diseases and conditions. Extensive work has also been performed to investigate potential utility in the treatment of various parasitic diseases and infections, for example coccidiomycosis, malaria and schistosomiasis. Reports of investigative work into the potential utility of the very many other known cyclosporins in these or related indications now abound in the literature.

 

………………………………

References

  1.  Wilkes, Gail; Ades, Terri B. (2004). Consumers Guide to Cancer Drugs. Jones & Bartlett Learning. p. 226. ISBN 9780763722548. Retrieved 29 May 2013.
  2.  Tao, Jian’guo; Sotomayor, Eduardo. (2012). Hematologic Cancers: From Molecular Pathobiology to Targeted Therapeutics. Springer. p. 335. ISBN 9789400750289.
  3. PSC-833Drugs Fut 1995, 20(10): 1010
  4. US 5525590
  5. Synthesis of [S-[1-14C]Val(7)]VALSPODAR application of (+)/(-)-[13,14Cn]BABS and (+)/(-)-[13,14Cn]DPMGBS, part 4J Label Compd Radiopharm 2000, 43(3): 205
  6. WO 2006013094
  7. WO 2005013947
  8. WO 2002098418
  9. WO 1999017757
  10. Pharmaceutical Research, 2001 ,  vol. 18,  2  pg. 183 – 190
  11. US2003/158097 A1
  12. Valspodar; EP-B1 0 296 122:
  13. WO 94/07858
Share

Glenmark conferred with Best Biotech New Molecular Entity Patent award

 companies, drugs  Comments Off on Glenmark conferred with Best Biotech New Molecular Entity Patent award
Jan 162014
 

GLENMARK PHARMA

IDMA best biotech NEW MOLECULAR ENTITY patent award to Glenmark

YEAR 2012-2013 YEAR in Mumbai India

PATENT  US 8236315

GLENMARK PHARMACEUTICALS, S.A., SWITZERLAND

INVENTORS

Elias LazaridesCatherine WoodsXiaomin FanSamuel HouHarald MottlStanislas BleinMartin BertschingerALSO PUBLISHED ASCA2712221A1CN101932606A,EP2245069A1US20090232804,WO2009093138A1

Publication number US8236315 B2
Publication type Grant
Application number US 12/358,682
Publication date 7 Aug 2012
Filing date 23 Jan 2009
Priority date 23 Jan 2008

USPTOUSPTO AssignmentEspacenetUS 8236315

The present disclosure relates generally to humanized antibodies or binding fragments thereof specific for human von Willebrand factor (vWF), methods for their preparation and use, including methods for treating vWF mediated diseases or disorders. The humanized antibodies or binding fragments thereof specific for human vWF may comprise complementarity determining regions (CDRs) from a non-human antibody (e.g., mouse CDRs) and human framework regions.

The present disclosure provides a humanized antibody or binding fragment thereof specific for vWF that comprises a heavy chain variable region sequence as set forth in SEQ ID NO: 19 and a light chain variable region sequence as set forth in SEQ ID NO: 28 ……….. CONT

MR GLEN SALDANHA

MD , CEO GLENMARK

INDIAN DRUG MANUFACTURERS’ ASSOCIATION   (IDMA)

102-B Poonam Chambers, Dr A B Road, Worli, Mumbai 400 018, INDIA
Tel : +91 – 22 – 24944625 / 24974308. Fax : ++91 – 22 – 24957023
email: ppr@idmaindia.com website : www.idma-assn.org

Share
Follow

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

Join other followers: