Home » Articles posted by DR ANTHONY MELVIN CRASTO Ph.D (Page 281)
Author Archives: DR ANTHONY MELVIN CRASTO Ph.D
EMA publishes Guideline Draft on Validation of biotechnology-derived Products
DRUG REGULATORY AFFAIRS INTERNATIONAL
For the EMA, it was necessary to develop an independent guideline on the topic as – despite the existence of harmonised ICH documents – specific aspects of the validation of biotechnology-derived products have been missing. A draft is now available.
EMA publishes Guideline Draft on Validation of biotechnology-derived Products
How to become a QP for Europe
DRUG REGULATORY AFFAIRS INTERNATIONAL
How to become a QP for Europe
Both the ECA and the European QP Association are often contacted by people who would like to become a Qualified Person in a Member State of the European Union or outside the EU to release products for the EU market. Read more.
IDX 18719; IDX 719; Samatasvir For HEPATITIS C in phase 2
-

IDX 18719; IDX 719; SamatasvirN-((1R)-2-((2S)-2-(5-(4-(6-(2-((2S)-1-((2S)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)pyrrolidin-2-yl)-3H-benzimidazol-5-yl)thieno(3,2-b)thiophen-3-yl)phenyl)-1H-imidazol-2-yl)pyrrolidin-1-yl)-2-oxo-1-phenylethyl)carbamate
Carbamic acid, N-((1R)-2-((2S)-2-(5-(4-(6-(2-((2S)-1-((2S)-2-((methoxycarbonyl)amino)-3-methyl-1-oxobutyl)-2-pyrrolidinyl)-1H-benzimidazol-6-yl)thieno(3,2-b)thien-3-yl)phenyl)-1H-imidazol-2-yl)-1-pyrrolidinyl)-2-oxo-1-phenylethyl)-, methyl ester
Carbamic acid, N-[(1R)-2-[(2S)-2-[5-[4-[6-[2-[(2S)-1-[(2S)-2-[(methoxycarbonyl)amino]-3-methyl-1-oxobutyl]-2-pyrrolidinyl]-1H-benzimidazol-6-yl]thieno[3,2-b]thien-3-yl]phenyl]-1H-imidazol-2-yl]-1-pyrrolidinyl]-2-oxo-1-phenylethyl]-, methyl ester
[(5)-l-((5)-2- {6-[5-(4- {(5)-2-[l-((R)-2-methoxycarbonylamino-2-phenyl- acetyl)-pyrrolidin-2-yl]-lH-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-2-yl)-lH- benzoimidazol-2-yl} -pyrrolidine- l-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester
[(S)-1-((S)-2-{6-[6-(4-{(S)-2-[1-((R)-2-methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-3H-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-3-yl]-1H-benzoimidazol-2-yl}-pyrrolidine-1-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester
Carbamic acid, N-((1R)-2-((2S)-2-(5-(4-(6-(2-((2S)-1-((2S)-2-((methoxycarbonyl)amino)-3-methyl-1-oxobutyl)-2-pyrrolidinyl)-1H-benzimidazol-6-yl)thieno(3,2-b)thien-3-yl)phenyl)-1H-imidazol-2-yl)-1-pyrrolidinyl)-2-oxo-1-phenylethyl)-, methyl ester
Methyl N-((1R)-2-((2S)-2-(5-(4-(6-(2-((2S)-1-((2S)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)pyrrolidin-2-yl)-3H-benzimidazol-5-yl)thieno(3,2-b)thiophen-3-yl)phenyl)-1H-imidazol-2-yl)pyrrolidin-1-yl)-2-oxo-1-phenylethyl)carbamate
-
- Idenix Pharmaceuticals, Inc. phase 2
-
CAS Number: 1312547-19-5
-
C47-H48-N8-O6-S2
- 885.0782
- UNII-21P699C5FC
- deleted CAS Registry Numbers: 1458063-70-1
- INhttp://www.google.com/patents/WO2011075615A1?cl=en

A 169 IN http://www.google.com/patents/US20120252721
compd inhttp://www.google.com/patents/WO2014036244A1?cl=en
ANY ERROR amcrasto@gmail.com
samatasvir
Samatasvir is an orally-available pan-genotypic hepatitis C virus (HCV) non-structural protein 5A (NS5A) inhibitor in phase II clinical studies at Idenix for the treatment of treatment-naïve genotype 1-4 HCV-infected patients in combination with simeprevir and ribavirin.
Jun 6, 2013Idenix Pharmaceuticals Announces Samatasvir (IDX719) Poster Presentations at the Asian Pacific Association for the Study of the Liver (APASL) Conference
CAMBRIDGE, Mass., June 6, 2013 (GLOBE NEWSWIRE) — Idenix Pharmaceuticals, Inc. (Nasdaq:IDIX), a biopharmaceutical company engaged in the discovery and development of drugs for the treatment of human viral diseases, today announced three poster presentations featuring clinical and preclinical data for samatasvir (IDX719), Idenix’s once-daily pan-genotypic NS5A inhibitor for the treatment of hepatitis C virus (HCV) infection, at the Asian Pacific Association for the Study of the Liver (APASL) Liver Week 2013, taking place in Singapore, June 6-10, 2013. Idenix recently initiated a phase II clinical trial (HELIX-1) evaluating an all-oral, direct-acting antiviral (DAA) HCV combination regimen of samatasvir and simeprevir (TMC435), a once-daily protease inhibitor jointly developed by Janssen R&D Ireland and Medivir AB.
The following abstracts will be presented in poster sessions during APASL Liver Week 2013 in the Conference Exhibition Hall on Friday, June 7, 2013, 8:30am – 5:30pm SGT:
- Abstract No. 2110: “Pharmacokinetics and Pharmacodynamics of IDX719, a Pan-Genotypic HCV NS5A Inhibitor, in Genotype 1, 2, 3 or 4 HCV-Infected Subjects.”
- Abstract No. 2121: “Hepatitis C Virus NS5A Inhibitor IDX719 Demonstrates Potent, Pan-genotypic Activity in Preclinical and Clinical Studies.”
- Abstract No. 2127: “IDX719, a Pan-genotypic HCV NS5A Replication Complex Inhibitor, Is a Promising Candidate for HCV Combination DAA Treatment.”
ABOUT SAMATASVIR (IDX719)
Samatasvir is an NS5A inhibitor with low picomolar, pan-genotypic antiviral activity in vitro. To date, samatasvir has been safe and well-tolerated after single and multiple doses of up to 150 mg in healthy volunteers for up to 14 days’ duration and up to 100 mg in HCV-infected patients up to 3 days’ duration. There have been no treatment-emergent serious adverse events reported in the program. Samatasvir has demonstrated potent pan-genotypic antiviral activity in HCV-infected patients with mean maximal viral load reductions up to approximately 4.0 log10 IU/mL across HCV genotypes 1-4 in a proof-of-concept, three-day monotherapy study.
The HELIX-1 trial is a 12-week, randomized, double-blind, parallel group study evaluating the safety and tolerability of samatasvir and simeprevir in addition to antiviral activity endpoints, with a target enrollment of 90 treatment-naïve, non-cirrhotic, genotype 1b or 4 HCV-infected patients. The HELIX-1 trial is the first study in HCV-infected patients to commence under a non-exclusive collaboration agreement signed with Janssen in January 2013. A second trial (HELIX-2) of samatasvir, simeprevir and TMC647055, a once-daily non-nucleoside polymerase inhibitor boosted with low-dose ritonavir being developed by Janssen, is expected to initiate in the second half of 2013.
ABOUT HEPATITIS C
Hepatitis C virus is a common blood-borne pathogen infecting three to four million people worldwide annually. The World Health Organization (WHO) estimates that more than 170 million people worldwide are chronically infected with HCV, representing a nearly 5-fold greater prevalence than human immunodeficiency virus.
ABOUT IDENIX
Idenix Pharmaceuticals, Inc., headquartered in Cambridge, Massachusetts, is a biopharmaceutical company engaged in the discovery and development of drugs for the treatment of human viral diseases. Idenix’s current focus is on the treatment of patients with hepatitis C virus (HCV) infection. For further information about Idenix, please refer to www.idenix.com.
-

- ………………………………………

- WO 2014036244
- http://www.google.com/patents/WO2014036244A1?cl=en

-
[(5)-l-((5)-2-{6-[6-(4-{(5)-2-[l-((i?)-2- methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-3H-imidazol-4-yl}-phenyl)- thieno[3,2-¾]thiophen-3-yl]- lH-benzoimidazol-2-yl} -pyrrolidine- 1 -carbonyl)-2-methyl- propyl]-carbamic acid methyl ester (“the Compound”), having the structure of Formula I:
(I) or an isotopic variant thereof, or a pharmaceutically acceptable salt or solvate thereof.
The Compound is a nonstructural protein 5A (NS5A) inhibitor. See U.S. Pat.App. Pub. Nos. US 2011/0150827 and US 2012/0252721, the disclosure of each of which is incorporated herein by reference in its entirety. The Compound is a potent and pan-genotypic inhibitor of HCV replication in vitro, with EC50 values ranging from 2 to 24 pM against HCV genotypes la, lb, 2a, 3a, 4a, and 5a. Id.
The Compound can be prepared according to the methods described in U.S.Pat. App. Pub. No. US 2011/0150827. The Compound can be also synthesized according to other methods apparent to those of skill in the art based upon the teaching herein.

- ………………
- WO 2011075615
- http://www.google.com/patents/WO2011075615A1?cl=en
- Example 36Synthesis of [(5)-l-((5)-2- {6-[5-(4- {(5)-2-[l-((R)-2-methoxycarbonylamino-2-phenyl- acetyl)-pyrrolidin-2-yl]-lH-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-2-yl)-lH- benzoimidazol-2-yl} -pyrrolidine- l-carbonyl)-2-methyl-propyl]-carbamic acid methyl esterA215
A215
1] Compound A215 was synthesized as shown
Scheme 27
[00612] Preparation of (S 2-[6-(5-bromo-thieno[3,2,b]thiophen-2-yl)-lH- benzoimidazol-2-yl] -pyrrolidine- 1-carboxylic acid tert-butyl ester E64. In a round bottom flask were added intermediate 66 (2.42 mmol) and 3,6-dibromo-thieno[3,2-b]thiophene (7.26 mmol). The system was purged and anhydrous dioxane (36 mL) was added. Then, NaHC(¾ 1M (7.26 mmol) and Pdl 18 (0.242 mmol) were added. The reaction mixture was stirred under reflux (110 °C) for 1.5 hrs. The reaction mixture was cooled down to room temperature and DCM was added. The mixture was washed with water and the organic layer dried, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent: DCM to DCM/MeOH 2%) to give intermediate E64 as a yellow foam in 19% yield. MS (ESI, EI+) m/z = 505.8 (MH+).[00613] Preparation of 6-(5-bromo-thieno[3,2,b]thiophen-2-yl)-(S -2-pyrrolidin-2-yl- lH-benzoimidazole, hydrochloride E65. Intermediate E65 was synthesized from
intermediate E64 (0.198 mmol), following the procedure as described for intermediate E47 (without purification) to give intermediate E65 as a yellow solid in quantitative yield. MS (ESI, EI+) m/z = 405.8 (MH+).
[00614] Preparation of ((5)-l- {(5)-2-[6-(5-bromo-thieno[3,2-b]thiophen-2-yl)-lH- benzoimidazol-2-yl]-pyrrolidine-l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester E66. Intermediate E65 (0.198 mmol) was dissolved in anhydrous DCM (5 mL). The intermediate 1 (0.198 mmol) was added, followed by HATU (0.257 mmol) and Et3N (0.792 mmol). The reaction mixture was stirred at room temperature for 45 min. DCM was added and the mixture was washed with water. The organic layer was dried over a2S04, filtered, and concentrated under reduced pressure. The residue was purified by silica gel
chromatography (eluent: DCM to DCM/MeOH 2%) to give intermediate E66 in quantitative yield. MS (ESI, EI+) m/z = 562.7 (MH+).
[00615] Preparation of (S 2-{4-[4-(5-{(5′)-2-[l-((5,)-2-methoxycarbonylamino-3- methyl-butyryl)-pyrrolidin-2-yl]-3H-benzoimidazol-5-yl}-thieno[3,2-b]thiophen-2-yl)- phenyl]-lH-imidazol-2-yl}-pyrrolidine-l-carboxylic acid tert-butyl ester E67. Intermediate E67 was synthesized from intermediate E66 (0.196 mmol), following the procedure as described for the compound Al (1 10° C for 35 min). The residue was purified by silica gel chromatography (eluent: DCM to DCM/MeOH 4%) to give intermediate E67 as a yellow solid in 46% yield. MS (ESI, EI ) m/z = 794.2 (MH ).
[00616] Preparation of{2-methyl-(5)-l-[(5)-2-(6-{5-[4-((5)-2-pyrrolidin-2-yl-lH- imidazol-4-yl)-phenyl]-thieno[3,2-b]thiophen-2-yl}-lH-benzoimidazol-2-yl)-pyrrolidine-l- carbonyl]-propyl}-carbamic acid methyl ester, hydrochloride E68. Intermediate E68 was synthesized from intermediate E67 (0.086 mmol), following the procedure as described for intermediate E47 (without purification) to give intermediate E68 as an orange solid in quantitative yield. MS (ESI, EI+) m/z = 694.14 (MH+).
[00617] Preparation of [(5)-l-((5)-2- {6-[5-(4- {(5)-2-[l-((R)-2-methoxycarbonylamino- 2-phenyl-acetyl)-pyrrolidin-2-yl]-lH-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-2-yl)- lH-benzoimidazol-2-yl} -pyrrolidine- l-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester A215. Compound A215 was synthesized from intermediate E68 (0.086 mmol) following the procedure as described for compound A114 to give compound A215 as a yellow solid in 48% yield. H NMR (DMS0-< 400 MHz) δ (ppm) 0.82 (d, J= 6.70 Hz, 3H), 0.86 (d, J= 6.70 Hz, 3H), 1.82-2.10 (m, 7H), 2.16-2.28 (m, 2H), 3.10-3.16 (m, 1H), 3.52-3.55 (m, 6H), 3.80-3.90 (m, 3H), 4.07 (t, J= 8.38 Hz, 1H), 5.04-5.19 (m, 2H), 5.37-5.53 (m, 1H), 6.91-7.1 (m, 1H), 7.30-7.88 (m, 15H), 11.77-1.95 (m, 1H), 12.29 (brs, 1H); MS (ESI, EI+) m/z = 885.3 (MH+).
- …………….
- WO 201213558

……………..
US 2013071352\
http://www.google.com/patents/US20130071352
Example 36 Synthesis of [(S)-1-((S)-2-{6-[5-(4-{(S)-2-[1-((R)-2-methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-1H-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-2-yl)-1H-benzoimidazol-2-yl}-pyrrolidine-1-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester A215
Compound A215 was synthesized as shown in 27.
Preparation of (S)-2-[6-(5-bromo-thieno[3,2,b]thiophen-2-yl)-1H-benzoimidazol-2-yl]-pyrrolidine-1-carboxylic acid tert-butyl ester E64. In a round bottom flask were added intermediate 66 (2.42 mmol) and 3,6-dibromo-thieno[3,2-b]thiophene (7.26 mmol). The system was purged and anhydrous dioxane (36 mL) was added. Then, NaHCO3 1M (7.26 mmol) and Pd118 (0.242 mmol) were added. The reaction mixture was stirred under reflux (110° C.) for 1.5 hrs. The reaction mixture was cooled down to room temperature and DCM was added. The mixture was washed with water and the organic layer dried, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent: DCM to DCM/MeOH 2%) to give intermediate E64 as a yellow foam in 19% yield. MS (ESI, EI+) m/z=505.8 (MH+).
Preparation of 6-(5-bromo-thieno[3,2,b]thiophen-2-yl)-(S)-2-pyrrolidin-2-yl-1H-benzoimidazole, hydrochloride E65. Intermediate E65 was synthesized from intermediate E64 (0.198 mmol), following the procedure as described for intermediate E47 (without purification) to give intermediate E65 as a yellow solid in quantitative yield. MS (ESI, EI+) m/z=405.8 (MH+).
Preparation of ((S)-1-{(S)-2-[6-(5-bromo-thieno[3,2-b]thiophen-2-yl)-1H-benzoimidazol-2-yl]-pyrrolidine-1-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester E66. Intermediate E65 (0.198 mmol) was dissolved in anhydrous DCM (5 mL). The intermediate 1 (0.198 mmol) was added, followed by HATU (0.257 mmol) and Et3N (0.792 mmol). The reaction mixture was stirred at room temperature for 45 min. DCM was added and the mixture was washed with water. The organic layer was dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent: DCM to DCM/MeOH 2%) to give intermediate E66 in quantitative yield. MS (ESI, EI+) m/z=562.7 (MH+).
Preparation of (S)-2-{4-[4-(5-{(S)-2-[1-((S)-2-methoxycarbonylamino-3-methyl-butyryl)-pyrrolidin-2-yl]-3H-benzoimidazol-5-yl}-thieno[3,2-b]thiophen-2-yl)-phenyl]-1H-imidazol-2-yl}-pyrrolidine-1-carboxylic acid tert-butyl ester E67. Intermediate E67 was synthesized from intermediate E66 (0.196 mmol), following the procedure as described for the compound A1 (110° C. for 35 min). The residue was purified by silica gel chromatography (eluent: DCM to DCM/MeOH 4%) to give intermediate E67 as a yellow solid in 46% yield. MS (ESI, EI+) m/z=794.2 (MH+).
Preparation of{2-methyl-(S)-1-[(S)-2-(6-{5-[4-((S)-2-pyrrolidin-2-yl-1H-imidazol-4-yl)-phenyl]-thieno[3,2-b]thiophen-2-yl}-1H-benzoimidazol-2-yl)-pyrrolidine-1-carbonyl]-propyl}-carbamic acid methyl ester, hydrochloride E68. Intermediate E68 was synthesized from intermediate E67 (0.086 mmol), following the procedure as described for intermediate E47 (without purification) to give intermediate E68 as an orange solid in quantitative yield. MS (ESI, EI+) m/z=694.14 (MH+).
Preparation of [(S)-1-((S)-2-{6-[5-(4-{(S)-2-[1-((R)-2-methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-1H-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-2-yl)-1H-benzoimidazol-2-yl}-pyrrolidine-1-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester A215. Compound A215 was synthesized from intermediate E68 (0.086 mmol) following the procedure as described for compound A114 to give compound A215 as a yellow solid in 48% yield. 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.82 (d, J=6.70 Hz, 3H), 0.86 (d, J=6.70 Hz, 3H), 1.82-2.10 (m, 7H), 2.16-2.28 (m, 2H), 3.10-3.16 (m, 1H), 3.52-3.55 (m, 6H), 3.80-3.90 (m, 3H), 4.07 (t, J=8.38 Hz, 1H), 5.04-5.19 (m, 2H), 5.37-5.53 (m, 1H), 6.91-7.1 (m, 1H), 7.30-7.88 (m, 15H), 11.77-1.95 (m, 1H), 12.29 (brs, 1H); MS (ESI, EI+) m/z=885.3 (MH+).
……………………..

US2012/252721
http://www.google.com/patents/US20120252721
Example 33Synthesis of [(S)-1-((S)-2-{6-[6-(4-{(S)-2-[1-((R)-2-methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-3H-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-3-yl]-1H-benzoimidazol-2-yl}-pyrrolidine-1-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester A169
Compound 169 was synthesized as shown in Scheme 24.
Preparation of (S)-2-{5-[4-(6-bromo-thieno[3,2-b]thiophen-3-yl)-phenyl]-1H-imidazol-2-yl}-pyrrolidine-1-carboxylic acid tert-butyl ester E78. To a mixture of DMF and water (20 mL/2.5 mL) were added Pd(PPh3)4 (0.1 mmol), 3,6-dibromo-thieno[3,2-b]thiophene (1.01 mmol), intermediate 6 (1.1 mmol), and sodium carbonate (4.04 mmol). The reaction mixture was degassed and irradiated for 1 hr at 80° C. Ethyl acetate was added and the organic layer was washed with water. The organic layer was dried over Na2SO4, filtered, and evaporated in vacuo. The residue was purified by silica gel chromatography (eluent: DCM-DCM/MeOH 98/2) to give intermediate E78 as a green gum in 41% yield. MS (ESI, EI+) m/z=532.19-530.31 (MH+).
Preparation of (S)-2-{5-[4-(6-{(S)-2-[1-((S)-2-methoxycarbonylamino-3-methyl-butyryl)-pyrrolidin-2-yl]-3H-benzoimidazol-5-yl}-thieno[3,2-b]thiophen-3-yl)-phenyl]-1H-imidazol-2-yl}-pyrrolidine-1-carboxylic acid tert-butyl ester E79. Compound 78 (0.198 mmol), intermediate 83 (0.228 mmol), and 1,1′-bis(di-tert-BP)ferrocene palladium dichloride (0.03 mmol) were added to a solution of dioxane (4 mL) and 1M NaHCO3 in water (0.594 mmol). The reaction mixture was irradiated at 90° C. for 1 hr. The mixture was diluted in dichloromethane and washed with water. The two layers were separated and the organic layer was concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent: DCM-DCM/MeOH 95/5) to give intermediate E79 as a brown foam in 70% yield. 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.90-0.91 (m, 6H), 1.51 (s, 9H), 1.67-2.40 (m, 10H), 3.07-3.1 (m, 2H), 3.45-3.50 (m, 1H), 3.72 (s, 3H), 3.90 (m, 1H), 4.37 (m, 1H), 5.00-5.01 (m, 1H), 5.45-5.48 (m, 2H), 7.26-8.12 (m, 10H), 10.67 (m, 1H); MS (ESI, EI+) m/z=792.79 (MH−).
Preparation of [(S)-1-((S)-2-{6-[6-(4-{(S)-2-[1-((R)-2-methoxycarbonylamino-2-phenyl-acetyl)-pyrrolidin-2-yl]-3H-imidazol-4-yl}-phenyl)-thieno[3,2-b]thiophen-3-yl]-1H-benzoimidazol-2-yl}-pyrrolidine-1-carbonyl)-2-methyl-propyl]-carbamic acid methyl ester A169. Intermediate E79 (0.132 mmol) was dissolved in methanol (2.6 mL) and 4N HCl in dioxane (2.64 mL) was added. The mixture was stirred 1 hr at room temperature before concentration under reduced pressure. The residue was dissolved in DMF (2.6 mL) and the mixture was cooled down to −100C. TEA (0.924 mmol), intermediate 31 (0.139 mmol), and HATU (0.172 mmol) were added and the mixture was stirred at −100C for 1 hr. Ethyl acetate was added and the mixture was washed with water. The organic layer was dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was filtered on a SCX-2 column and purified by silica gel chromatography (eluent: DCM-DCM/MeOH 97/3) to give compound A169 as a beige solid in 74% yield.
1H NMR (CDCl3, 400 MHz) δ (ppm) 0.89-0.91 (m, 6H), 1.40-2.42 (m, 8H), 3.08-3.24 (m, 3H), 3.67 (m, 3H), 3.71 (m, 4H), 3.88-3.89 (m, 1H), 4.34-4.38 (m, 1H), 5.30-5.32 (m, 1H), 5.42-5.45 (m, 3H), 6.03-6.04 (m, 1H), 7.26-8.14 (m, 16H), 10.65 (m, 1H);
MS (ESI, EI+) m/z=885.8 (MH+).

| US20110150827 * | Dec 17, 2010 | Jun 23, 2011 | Idenix Pharmaceuticals, Inc. | 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors |
| US20120252721 * | Mar 29, 2012 | Oct 4, 2012 | Idenix Pharmaceuticals, Inc. | Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor |
Want to know everything on vir series
click
http://drugsynthesisint.blogspot.in/p/vir-series-hep-c-virus-22.html
AND
http://medcheminternational.blogspot.in/p/vir-series-hep-c-virus.html

Want to know everything on vir series
click
http://drugsynthesisint.blogspot.in/p/vir-series-hep-c-virus-22.html
AND
http://medcheminternational.blogspot.in/p/vir-series-hep-c-virus.html
(4aS,7aR)-1-[5-[(3-Fluorophenyl)ethynyl]pyridin-2-yl]hexahydrocyclopenta[d][1,3]oxazin-2(1H)-one

(4aS,7aR)-1-[5-[(3-Fluorophenyl)ethynyl]pyridin-2-yl]hexahydrocyclopenta[d][1,3]oxazin-2(1H)-one
(4aS,7aR)-l-(5-Phenylethynyl-pyridin-2-yl)-hexahydro-cyclopenta[d] [l,3]oxazin-2-one
336.35
C20 H17 F N2 O2
F. Hoffmann-La Roche Ag, Hoffmann-La Roche Inc.
mgluR5 Positive Allosteric Modulators
Signal Transduction Modulators
http://www.google.com/patents/WO2014056710A1?cl=en
Example 1
(4aS,7aR)-l-(5-Phenylethynyl-pyridin-2-yl)-hexahydro-cyclopenta[d] [l,3]oxazin-2-one
Ste 1 : ((lR,2S)-2-Hydroxymethyl-cyclopentyl)-carbamic acid tert-butyl ester
To a well stirred suspension of 0.94 g (24.7 mmol, 2 equiv.) of L1AIH4 in 30ml of THF at 0°C was added dropwise at 0°C a solution of (lS,2R)-methyl 2-(tert-butoxycarbonylamino)- cyclopentanecarboxylate (CAS: 592503-55-4) (3.0 g, 12.3 mmol) (gas evolution, lightly exo therm). After 15 minutes at 0°C the reaction mixture was allowed to warm up to room temperature and was stirred for 2h. The mixture was cooled to 0°C and water was added dropwise. The precipitated inorganic salts were filtered through Celite and were washed with ethyl acetate. The filtrate was evaporated and the residue was purified by column
chromatography on silica gel eluting with a 0% to 50% ethyl acetate in heptane gradient to yield 1.99 g (75%) of the title compound as a crystalline white solid which was directly used in the next step. Ste 2: (4aS,7aR)-Hexahydro-cyclopenta[d][l,3]oxazin-2-one
To a solution of ((lR,2S)-2-hydroxymethyl-cyclopentyl)-carbamic acid tert-butyl ester (1.6 g, 7.43 mmol) in THF (40 ml) was added potassium tert-butoxide (3.34 g, 29.7 mmol, 4.0 equiv.) at room temperature. After stirring for lh at 60°C the reaction was allowed to warm up to room temperature and after workup with Ethyl acetate/water, drying and concentration in vaccuo, the crude material mixture was adsorbed on silica and chromatographed over a prepacked silica column (50g, 50% to 100% EtOAc in Heptane gradient) to yield 950 mg (91%) of the title compound as a white solid, which was directly used in the next step. -Fluoro-5-phenylethynyl-pyridine
In an 100ml 2-necked round bottomed flask under Argon were dissolved 2-fluoro-5-iodopyridine (5.0 g, 22.4 mmol, 1.0 equiv.) in THF (30 ml). After 5 minutes at room temperature were added bis(triphenylphosphin)palladium(II)chloride (944 mg, 1.35 mmol, 0.06 equiv.), triethylamine (6.81 g, 9.32 ml, 67.3 mmol, 3.0 equiv.), phenyl acetylene (2.75 g, 2.95 ml, 26.9 mmol, 1.2 equiv.) and copper(I)iodide (128 mg, 0.67 mmol, 0.03 equiv.). The brown suspension was cooled with water (exothermic) to room temperature and stirred overnight. Then 200ml of diethylether were added, the mixture was filtered, washed with ether and concentrated in vacuum to yield 5.7g of a brown solid which was adsorbed on silica and was chromatographed in 2 portions over a lOOg prepacked silica column eluting with a 0-10% ethyl acetate in heptane gradient to yield 3.99g (91%) of the title compound as a light brown solid, MS: m/e = 198.1 (M+H+). Step 4: (4aS aR)-l-(5-Phenylethynyl-pyridin-2-yl)-hexahydro-cyclopenta[d][l,3]oxazin-2-one In a 10ml Round bottomed flask were dissolved (4aS,7aR)-hexahydro-cyclopenta[d]- [l,3]oxazin-2-one (80 mg, 0.57 mmol, 1.0 equiv.) and 2-fluoro-5-(phenylethynyl)pyridine (112 mg, 0.57 mmol, 1.0 equiv.) in 2ml of DMF. Sodium hydride (60%> suspension) (29.5 mg, 0.74 mmol, 1.3 equiv.) were added and the brown suspension was stirred at room temperature overnight. The reaction mixture was quenched with water and extracted twice with ethyl acetate. The combined organic phases were dried, filtered and concentrated. The crude material was purified by flash chromatography over a prepacked silica column eluting with 0-50% ethyl acetae in heptane gradient to yield 42.5mg of the title compound as colorless amorphous solid, MS: m/e = 319.1 (M+H+).
Example 2
(4aS,7aR)- 1- [5-(3-Fluorophenylethynyl)-py ridin-2-yl] -hexahydro- cyclopenta[d] [l,3]oxazin-2-one
Step 1 : 2-Fluoro-5-(3-fluoro-phenylethynyl)-pyridine
The title compound was prepared in accordance with the general method of Example 1, step 3 using 3-flurorophenylacetylene instead of phenylacetylene to yield the title compound as a crystalline white solid, MS: m/e = 216.2 (M+H+).
Step 2 : (4aS ,7aR)- 1 – [5 -(3 -Fluorophenylethynyl)-pyridin-2-yl] -hexahydro- cyclopenta[d] [ 1 ,3]oxazin-2-one
The title compound was prepared in accordance with the general method of Example 1, step 4 using (4aS,7aR)-hexahydro-cyclopenta[d]-[l,3]oxazin-2-one (66 mg, 0.47 mmol) (Example 1, step 2) and 2-fluoro-5-((3-fluorophenyl)ethynyl)pyridine (100 mg, 0.47 mmol) to yield 48 mg (31%) of the title compound as a light yellow amorphous solid; MS: m/e = 337.3 (M+H+).
(3S)-3-[[2-Amino-5-(2-methoxybenzyl)-5H-pyrrolo[3,2-d]pyrimidin-4-yl]amino]heptan-1-ol

COMPOUND 80 ABOVE
IN
http://www.google.com/patents/WO2014056953A1?cl=en
(3S)-3-[[2-Amino-5-(2-methoxybenzyl)-5H-pyrrolo[3,2-d]pyrimidin-4-yl]amino]heptan-1-ol
TLR7 Receptor Agonists , Signal Transduction Modulators
Janssen R&D Ireland (INNOVATOR)
383.4873
C21 H29 N5 O2
FDA Approves Epanova (omega-3-carboxylic acids) for the Treatment of Severe Hypertriglyceridemia
FDA Approves Epanova (omega-3-carboxylic acids) for the Treatment of Severe Hypertriglyceridemia
Tuesday, 6 May 2014 — AstraZeneca today announced that the US Food and Drug Administration (FDA) has approved Epanova (omega-3-carboxylic acids) as an adjunct to diet to reduce triglyceride levels in adults with severe hypertriglyceridemia (triglyceride levels greater than or equal to 500 mg/dL).
Epanova is the first FDA approved prescription omega-3 in free fatty acid form. The dosage of Epanova is 2 grams (2 capsules) or 4 grams (4 capsules), making it the first prescription omega-3 to have a dosing option as few as two capsules once a day, with or without food.http://www.drugs.com/newdrugs/fda-approves-epanova-omega-3-carboxylic-acids-severe-hypertriglyceridemia-4038.html?utm_source=ddc&utm_medium=email&utm_campaign=Today%27s+news+summary+-+May+6%2C+2014 AT DRUGS.COM
old cut paste
LONDON, Sept. 18, 2013 – AstraZeneca today announced that the US Food and Drug Administration (FDA) has accepted for review a New Drug Application (NDA) for EpanovaTM, an investigational compound for the treatment for patients with severe hypertriglyceridaemia (triglyceride levels greater than or equal to 500mg/dL). The NDA submission for Epanova was filed by Omthera Pharmaceuticals, now a wholly-owned subsidiary of AstraZeneca, as a 505(b)(1) application in July 2013. The Prescription Drug User Fee Act (PDUFA) goal date for the FDA is 5 May 2014.http://www.pharmalive.com/fda-accepts-astrazeneca-nda-for-epanova
Europe approves Pierre Fabre’s Hemangiol
Regulators in Europe have given the green light to Pierre Fabre’s Hemangiol for the treatment of proliferating infantile haemangioma.
The European Commission has granted the French company authorisation to market Hemangiol, an oral solution of the beta-blocker propranolol specially developed for paediatric use. The thumbs-up follows an approval in the USA in March; it will be launched across the Atlantic as Hemangeol next month.
read at
http://www.pharmatimes.com/Article/14-05-06/Europe_approves_Pierre_Fabre_s_Hemangiol.aspx
by
Kevin Grogan
Kevin Grogan is the World News Online Editor for PharmaTimes and has been writing about the industry since 1997, having previously worked as a journalist on rock/pop music publications, while contributing articles to the likes of The Independent and the Manchester Evening News on football. He specialises in the business developments of the pharmaceutical industry – financial results, mergers and acquisitions, alliances, legal tussles – and has a particular interest in emerging markets and the prospects for biotech.
Novel antioxidant makes old arteries seem young again, study finds
An antioxidant that targets specific cell structures—mitochondria—may be able to reverse some of the negative effects of aging on arteries, reducing the risk of heart disease, according to a new study by the University of Colorado Boulder.
When the research team gave old mice—the equivalent of 70- to 80-year-old humans—water containing an antioxidant known as MitoQ for four weeks, their arteries functioned as well as the arteries of mice with an equivalent human age of just 25 to 35 years.
The researchers believe that MitoQ affects the endothelium, a thin layer of cells that lines our blood vessels. One of the many functions of the endothelium is to help arteries dilate when necessary. As people age, the endothelium is less able to trigger dilation and this leads to a greater susceptibility to cardiovascular disease.
“One of the hallmarks of primary aging is endothelial dysfunction,” said Rachel Gioscia-Ryan, a…
View original post 495 more words
FDA OKs Ibalizumab, a Sterile Biologic for HIV
FDA OKs Ibalizumab, a Sterile Biologic for HIV
// // // // // // // // // // // // // // // // // // // // //

WuXi PharmaTech and TaiMed Biologics announced that the FDA has approved the first batch of the ibalizumab drug substance and sterile drug product, manufactured at WuXi’s biologics facilities, for ongoing treatment of patients under investigator-sponsored INDs. Read more…FULL STORY
FDA OKs Ibalizumab, a Sterile Biologic for HIV
// // // // // // // // // // // // // // // // // // // // //
Tomensides A–D, new antiproliferative phenylpropanoid sucrose esters from Prunus tomentosa leaves…..might be valuable source for new potent anticancer drug candidates.

http://www.sciencedirect.com/science/article/pii/S0960894X14003540
Volume 24, Issue 11, 1 June 2014, Pages 2459–2462
Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, PR China
To search for novel cytotoxic constituents against cancer cells as lead structures for drug development, four new 3-phenylpropanoid-triacetyl sucrose esters, named tomensides A–D (1–4), and three known analogs (5–7) were isolated from the leaves of Prunus tomentosa. Their structures were elucidated by spectroscopic analyses (1D, 2D NMR, CD and HRESIMS). The cytotoxic activities of all isolates against four human cancer cell lines (MCF-7, A549, HeLa and HT-29) were assayed, and the results showed that these isolates displayed stronger inhibitory activities compared with positive control 5-fluorouracil. Tomenside A (1) was the most active compound with IC50 values of 0.11–0.62 μM against the four tested cell lines. The structure–activity relationship (SAR) of the isolates was also discussed. The primary screening results indicated that these 3-phenylpropanoid-triacetyl sucrose esters might be valuable source for new potent anticancer drug candidates.














