AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

P7435 from Piramal Enterprises Mumbai, India

 phase 1  Comments Off on P7435 from Piramal Enterprises Mumbai, India
Apr 052016
 

str1

str1

P7435

Piramal Enterprises Mumbai, India

P-7435; P7435-DGAT1, P7435, P 7435

CAS 1210756-48-1,
C22 H19 F N4 O4 S
L-​Valine, N-​[[3-​[4-​[(6-​fluoro-​2-​benzothiazolyl)​amino]​phenyl]​-​5-​isoxazolyl]​carbonyl]​-
Molecular Weight, 454.47

GDAT1 inhibitor

  • Phase IDiabetes mellitus; Lipid metabolism disorders
  • ClassAntihyperglycaemics; Antihyperlipidaemics; Small molecules
  • Mechanism of ActionDiacylglycerol O acyltransferase inhibitors
Company Piramal Enterprises Ltd.
Description Diacylglycerol O-acyltransferase-1 (DGAT1) inhibitor
Molecular Target Diacylglycerol O-acyltransferase-1 (DGAT1)
Mechanism of Action Diacylglycerol O-acyltransferase-1 (DGAT1) inhibitor
Therapeutic Modality
Latest Stage of Development Phase I
Standard Indication Metabolic (unspecified)
Indication Details Treat metabolic disorders

https://clinicaltrials.gov/ct2/show/NCT01910571

https://clinicaltrials.gov/ct2/show/NCT01764425

  • 24 Nov 2014Piramal Enterprises completes a phase I trial in healthy, overweight or obese subjects in USA (NCT01910571)
  • 17 Jun 2014Adverse events and pharmacokinetics data from a phase I trial in healthy male volunteers presented at the 74th Annual Scientific Sessions of the American Diabetes Association (ADA-2014)
  • 17 Jun 2014Pharmacodynamics data from preclinical studies in Dyslipidaemia and obesity presented at the 74th Annual Scientific Sessions of the American Diabetes Association (ADA-2014)

Chairman Ajay Piramal

Swati Piramal-The Vice Chairperson of Piramal Enterprises Ltd

Nandini Piramal, Executive Director, Piramal Enterprises

Piramal Enterprises gets US FDA approval for P7435 IND

http://www.pharmabiz.com/NewsDetails.aspx?aid=76992&sid=2

Our Bureau, Mumbai
Tuesday, August 06, 2013, 12:25 Hrs  [IST]

Piramal Enterprises Ltd has received US Food and Drug Administration (FDA) approval for its Investigational New Drug (IND) P7435. This is a novel, potent and highly selective, oral diacylglycerolacyltransferase 1 (DGAT1) inhibitor.

P7435 has been developed by the NCE Research Division of PEL for the management of metabolic disorders such as lipid abnormalities and diabetes. It is well-established that increased lipid levels’ (including triglycerides) is one of the major risk factors for cardiovascular disease (CVD). It has been reported by the World Health Organisation, that CVD, is the number one cause of deaths globally, representing approximately 30 per cent of all deaths. Currently, there is a significant medical need for effective and safe drugs for the management of lipid abnormalities and metabolic disorders.

P7435 has demonstrated its lipid lowering potential in various preclinical studies by showing significant reduction in triglyceride levels, glucose and insulin levels,and decrease in food intake and body weight gain -factors which are associated with lipid abnormalities and metabolic disorders.

PEL has established the safety and tolerability of P7435 in a phase I trial recently completed in India. This extension trial in the US will further evaluate the safety and efficacy of P7435 in a larger population.

Dr Swati Piramal, vice chairperson, Piramal Enterprises, said, “The NCE Research division of PEL continues its ambitious diabetes/metabolic disorders programme to discover and develop NCEs to fight against diseases like diabetes and lipid disorders. With P7435 we are looking at addressing a serious need for effective and well-tolerated drugs that treat lipid disorders, which are commonly associated with diabetes and CVDs. Expansion of this trial will allow testing this NCE in a wider population,which is critical to the development of this drug and will provide therapeutic solutions not just to India but also to the rest of the world.”

The NCE Research division of Piramal Enterprises focuses on the discovery and development of innovative small molecule medicines to improve the lives of patients suffering from cancer, metabolic disorders and inflammatory conditions. The key elements of its strategy include capitalizing on Piramal’s strengths, in particular the India advantage, and leveraging external partnerships to achieve high levels of R&D productivity. Piramal’s state-of-the-art Research Centre in Mumbai has comprehensive capabilities spanning target identification all the way through clinical development. Its robust pipeline, including 8 compounds in clinical development, bears testimony to its innovative and rigorous drug discovery process.

PAPER

European Journal of Medicinal Chemistry (2012), 54, 324-342

http://www.sciencedirect.com/science/article/pii/S0223523412003133

PATENT

WO 2010023609

http://www.google.co.in/patents/WO2010023609A1?cl=en

/////////Piramal Enterprises,  Mumbai, India, P-7435, P7435-DGAT1, P7435, P 7435, GDAT1 inhibitor

O=C(O)[C@@H](NC(=O)c1cc(no1)c2ccc(cc2)Nc3nc4ccc(F)cc4s3)C(C)C

Share

Novartis Molecule for functionally liver selective glucokinase activators for the treatment of type 2 diabetes

 Uncategorized  Comments Off on Novartis Molecule for functionally liver selective glucokinase activators for the treatment of type 2 diabetes
Apr 052016
 

STR3

Figure US07750020-20100706-C00023

2 (R)-3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

(3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide)

cas 866772-52-3

Novartis Ag

NVP-LBX192

LBX-192

54 Discovery and Evaluation of NVP-LBX192, a Liver Targeted Glucokinase Activator

Thursday, October 8, 2009: 10:30 AM
Nathan Hale North (Hilton Third Floor)
Gregory R. Bebernitz, PhD , Global Discovery Chemistry, Novartis Institute for Biomedical Research, Cambridge, MA
Glucokinase (GK) activators are currently under investigation by a number of pharmaceutical companies with only a few reaching clinical evaluation.  A GK activator has the promise of potentially affecting both the beta-cell of the pancreas, by improving glucose sensitive insulin secretion, as well as the liver, by reducing uncontrolled glucose output and restoring post prandial glucose uptake and storage as glycogen.  We will describe our efforts to generate liver selective GK activators which culminated in the discovery of NVP-LBX192 (3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide).  This compound activated the GK enzyme in vitro at low nM concentrations and significantly reduced glucose levels during an oral glucose tolerance test in normal as well as diabetic mice.

https://acs.confex.com/acs/nerm09/webprogram/Paper75087.html

Molecular Formula: C26H33N5O4S2
Molecular Weight: 543.70132 g/mol

Sulfonamide-Thiazolpyridine Derivatives,  Glucokinase Activators, Treatment Of Type 2 Diabetes

2009 52 (19) 6142 – 6152
Investigation of functionally liver selective glucokinase activators for the treatment of type 2 diabetes
Journal of Medicinal Chemistry
Bebernitz GR, Beaulieu V, Dale BA, Deacon R, Duttaroy A, Gao JP, Grondine MS, Gupta RC, Kakmak M, Kavana M, Kirman LC, Liang JS, Maniara WM, Munshi S, Nadkarni SS, Schuster HF, Stams T, Denny IS, Taslimi PM, Vash B, Caplan SL

2010 240th (August 22) Medi-198
Glucokinase activators with improved physicochemicalproperties and off target effects
American Chemical Society National Meeting and Exposition
Kirman LC, Schuster HF, Grondine MS et al

2010 240th (August 22) Medi-197
Investigation of functionally liver selective glucokinase activators
American Chemical Society National Meeting and Exposition
Schuster HF, Kirman LC, Bebernitz GC et al

PATENT

http://www.google.com/patents/US7750020

EXAMPLE 1 3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

A. Phenylacetic Acid Ethyl Ester

A solution of phenylacetic acid (50 g, 0.36 mol) in ethanol (150 mL) is treated with catalytic amount of sulfuric acid (4 mL). The reaction mixture is refluxed for 4 h. The reaction is then concentrated in vacuo. The residue is dissolved in diethyl ether (300 mL) and washed with saturated aqueous sodium bicarbonate solution (2×50 mL) and water (1×100 mL). The organic layer dried over sodium sulfate filtered and concentrated in vacuo to give phenylacetic acid ethyl ester as a colorless oil: 1H NMR (400 MHz, CDCl3) δ 1.2 (t, J=7.2, 3H), 3.6 (s, 2H), 4.1 (q, J=7.2, 2H), 7.3 (m, 5H); MS 165 [M+1]+.

B. (4-Chlorosulfonyl-phenyl)-acetic acid ethyl ester

To a cooled chlorosulfonic acid (83.83 g, 48 mL, 0.71 mol) under nitrogen is added the title A compound, phenylacetic acid ethyl ester (59 g, 0.35 mol) over a period of 1 h. Reaction temperature is brought to RT (28° C.), then heated to 70° C., maintaining it at this temperature for 1 h while stirring. Reaction is cooled to RT and poured over saturated aqueous sodium chloride solution (200 mL) followed by extraction with DCM (2×200 mL). The organic layer is washed with water (5×100 mL), followed by saturated aqueous sodium chloride solution (1×150 mL). The organic layer dried over sodium sulfate, filtered and concentrated in vacuo to give crude (4-chlorosulfonyl-phenyl)acetic acid ethyl ester. Further column chromatography over silica gel (60-120 mesh), using 100% hexane afforded pure (4-chlorosulfonyl-phenyl)-acetic acid ethyl ester as a colorless oil.

C. [4-(4-Methyl-piperazine-1-sulfonyl)-phenyl]-acetic acid ethyl ester

A solution of N-methylpiperazine (9.23 g, 10.21 ml, 0.092 mol), DIEA (13 g, 17.4 mL, 0.10 mol) and DCM 80 mL is cooled to 0° C., and to this is added a solution of the title B compound, (4-chlorosulfonyl-phenyl)-acetic acid ethyl ester (22 g, 0.083 mol) in 50 mL of DCM within 30 min. Reaction mixture stirred at 0° C. for 2 h, and the reaction mixture is washed with water (100 mL), followed by 0.1 N aqueous hydrochloric acid solution (1×200 mL). The organic layer dried over sodium sulfate, filtered and concentrated under vacuo to give crude [4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-acetic acid ethyl ester. Column chromatography over silicagel (60-120 mesh), using ethyl acetate afforded pure [4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-acetic acid ethyl ester as white crystalline solid: 1H NMR (400 MHz, CDCl3) δ 1.3 (t, J=7.4, 3H), 2.3 (s, 3H), 2.5 (m, 4H), 3.0 (br s, 4H), 3.7 (s, 2H), 4.2 (q, J=7.4, 2H), 7.4 (d, J=8.3, 2H), 7.7 (d, J=7.3, 2H); MS 327 [M+1]+.

D. 3-Cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid ethyl ester

A solution of the title C compound, [4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-acetic acid ethyl ester (15 g, 0.046 mol) in a mixture of THF (60 mL) and DMTP (10 mL) is cooled to −78° C. under nitrogen. The resulting solution is stirred at −78° C. for 45 min and to this is added LDA (25.6 mL, 6.40 g, 0.059 mol, 25% solution in THF/Hexane). A solution of iodomethylcyclopentane (11.60 g, 0.055 mol) in a mixture of DMTP (12 mL) and THF (20 mL) is added over a period of 15 min at −78° C. and reaction mixture stirred at −78° C. for 3 h further, followed by stirring at 25° C. for 12 h. The reaction mixture is then quenched by the dropwise addition of saturated aqueous ammonium chloride solution (50 mL) and is concentrated in vacuo. The residue is diluted with water (50 mL) and extracted with ethyl acetate (3×100 mL). The organic solution is washed with a saturated aqueous sodium chloride (2×150 mL), dried over sodium sulfate, filtered and concentrated in vacuo. Column chromatography over silica gel (60-120 mesh), using 50% ethyl acetate in hexane as an eluent to afford 3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid ethyl ester as a white solid: 1H NMR (400 MHz, CDCl3) δ 0.9-2.1 (m, 11H), 1.2 (t, J=7.1, 3H), 2.3 (s, 3H), 2.5 (br s, 4H), 3.0 (br s, 4H), 3.6 (m, 1H), 4.1 (q, J=7.1, 2H), 7.5 (d, J=8.3, 2H), 7.7 (d, J=8.3, 2H); MS 409 [M+1]+.

E. 3-Cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid

A solution of the title D compound, 3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid ethyl ester (14 g, 0.034 mol) in methanol:water (30 mL:10 mL) and sodium hydroxide (4.11 g, 0.10 mol) is stirred at 60° C. for 8 h in an oil bath. The methanol is then removed in vacuo at 45-50° C. The residue is diluted with water (25 mL) and extracted with ether (1×40 mL). The aqueous layer is acidified to pH 5 with 3 N aqueous hydrochloric acid solution. The precipitated solid is collected by vacuum filtration, washed with water (20 mL), followed by isopropyl alcohol (20 mL). Finally, solid cake is washed with 100 mL of hexane and dried under vacuum at 40° C. for 6 h to give 3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid as a white solid: 1H NMR (400 MHz, CDCl3) δ 1.1-2.0 (m, 11H), 2.4 (s, 3H), 2.7 (br s, 4H), 3.1 (br s, 4H), 3.6 (m, 1H), 7.5 (d, J=8.3, 2H), 7.6 (d, J=8.3, 2H); MS 381 [M+l]+.

F. 5-Methoxy-thiazolo[5,4-b]pyridin-2-ylamine

A solution of 6-methoxy-pyridin-3-ylamine (5.0 g, 0.0403 mol) in 10 mL of acetic acid is added slowly to a solution of potassium thiocyanate (20 g, 0.205 mol) in 100 mL of acetic acid at 0° C. followed by a solution of bromine (2.5 mL, 0.0488 mol) in 5 mL of acetic acid. The reaction is stirred for 2 h at 0° C. and then allowed to warm to RT. The resulting solid is collected by filtration and washed with acetic acid, then partitioned between ethyl acetate and saturated aqueous sodium bicarbonate. The insoluble material is removed by filtration and the organic layer is evaporated and dried to afford 5-methoxy-thiazolo[5,4-b]pyridin-2-ylamine as a tan solid.

G. 3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

A solution of the title E compound, 3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid (5 g, 0.013 mol) in DCM (250 mL) is cooled to 0° C. and then charged HOBt hydrate (2.66 g, 0.019 mol), followed by EDCI hydrochloride (6 g, 0.031 mol). The reaction mixture is stirred at 0° C. for 5 h. After that the solution of the title F compound, 5-methoxy-thiazolo[5,4-b]pyridin-2-ylamine (2.36 g, 0.013 mol) and D1EA (8 mL, 0.046 mol) in a mixture of DCM (60 mL) and DMF (20 mL) is added dropwise over 30 min. Reaction temperature is maintained at 0° C. for 3 h, then at RT (28° C.) for 3 days. Reaction is diluted with (60 mL) of water and the organic layer is separated and washed with saturated sodium bicarbonate solution (2×50 mL) followed by water washing (2×50 mL) and saturated sodium chloride aqueous solution (1×150 mL). Finally the organic layer is dried over sodium sulfate, filtered, and evaporated under vacuo. The crude product is purified using column chromatography over silica gel (60-120 mesh), using 40% ethyl acetate in hexane as an eluent to afford 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide as a white solid: 1H NMR (400 MHz, CDCl3) δ 0.9-2.1 (m, 11H), 2.2 (s, 3H), 2.5 (br s, 4H), 3.1 (br s, 4H), 3.7 (m, 1H), 4.0 (s, 3H), 6.8 (d, J=8.8, 1H), 7.5 (d, J=8.3, 2H), 7.7 (d, J=8.3, 2H), 7.8 (d, J=8.8, 1H), 8.6 (s, 1H); MS 617 [M+1]+.

H. 3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide dihydrochloride

The title G compound, 3-cyclopentyl-2-(4-methyl piperazinyl sulfonyl)phenyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)propionamide (2.8 g, 0.0051 mol) is added to a cooled solution of 10% hydrochloric acid in isopropanol (3.75 mL). The reaction mixture is stirred at 0° C. for 1 h and then at RT for 2 h. The solid is separated, triturated with 10 mL of isopropanol and collected by vacuum filtration and washed with 50 mL of hexane. The solid is dried at 70° C. for 48 h to afford 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide dihydrochloride as an off white solid.

EXAMPLE 2 (R)-3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

The title compound is obtained analogously to Example 1 by employing the following additional resolution step:

The racemic title E compound of Example 1,3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid (10 g, 0.026 mol) in 1,4-dioxane (500 mL) is treated in a three necked 1 liter flask, equipped with heating mantle, water condenser, calcium chloride guard tube and mechanical stirrer with 3.18 g (0.026 mol) of (R)-(+)-1-phenylethylamine. This reaction mixture is then refluxed at 100° C. for 1 h. The clear reaction solution is cooled to RT (27° C.) and stirred for 10 h. The crystallized salt is collected by filtration under vacuum, washed with 5 mL of hexane and dried under vacuum to afford salt A.

The salt A is dissolved in 1,4-dioxane (500 mL) and heated at 100° C. for 1 h. The clear reaction solution is cooled to RT (27° C.) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 50 mL of hexane, and dried under vacuum to afford salt B.

The salt B is dissolved in 1,4-dioxane (290 mL) and heated at 100° C. for 1 h. The clear reaction solution is cooled to RT (27° C.) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 30 mL of hexane, and dried under vacuum to afford salt C.

The salt C is dissolved in 1,4-dioxane (100 mL) and heated at 100° C. for 1 h. The clear reaction solution is cooled to RT (27° C.) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 30 ml of hexane, and dried under vacuum to afford salt D.

The salt D is treated with aqueous hydrochloric acid solution (20 mL, 1 mL of concentrated hydrochloric acid diluted with 100 mL of water) and stirred for 5 min. The white solid precipitates out and is collected by vacuum filtration, washed with 10 mL of cold water, 5 mL of isopropanol and 20 mL of hexane, and dried under vacuum to yield the hydrochloride salt of (R)-(−)-3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid, salt E.

The salt E is neutralized by stirring with aqueous sodium bicarbonate solution (10 mL, 1 g of sodium bicarbonate dissolved in 120 mL of water) for 5 min. The precipitated solid is collected by filtration, washed with 10 mL of cold water, 100 mL of hexane, and dried to afford (R)-(−)-3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid: m.p. 202.2-203.4° C.

Alternatively, the title compound may be obtained by the resolution of the racemic title compound of Example 1 using the following preparative chiral HPLC method:

  • Column: Chiralcel OD-R (250×20 mm) Diacel make, Japan;
  • Solvent A: water:methanol:acetonitrile (10:80:10 v/v/v);
  • Solvent B: water:methanol:acetonitrile (05:90:05 v/v/v);
  • Using gradient elution: gradient program (time, min/% B): 0/0, 20/0, 50/100, 55/0, 70/0;
  • Flow rate: 6.0 mL/min; and
  • Detection: by UV at 305 nm.

EXAMPLE 3 (S)-3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

The title compound is prepared analogously to Example 2.

J MED CHEM 2009, 52, 6142-52

Investigation of Functionally Liver Selective Glucokinase Activators for the Treatment of Type 2 Diabetes

Novartis Institutes for BioMedical Research, Inc., 100 Technology Square, Cambridge, Massachusetts 02139
Torrent Research Centre, Village Bhat, Gujarat, India
J. Med. Chem., 2009, 52 (19), pp 6142–6152
DOI: 10.1021/jm900839k

http://pubs.acs.org/doi/abs/10.1021/jm900839k

Abstract Image

Type 2 diabetes is a polygenic disease which afflicts nearly 200 million people worldwide and is expected to increase to near epidemic levels over the next 10−15 years. Glucokinase (GK) activators are currently under investigation by a number of pharmaceutical companies with only a few reaching early clinical evaluation. A GK activator has the promise of potentially affecting both the β-cells of the pancreas, by improving glucose sensitive insulin secretion, as well as the liver, by reducing uncontrolled glucose output and restoring post-prandial glucose uptake and storage as glycogen. Herein, we report our efforts on a sulfonamide chemotype with the aim to generate liver selective GK activators which culminated in the discovery of 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide (17c). This compound activated the GK enzyme (αKa = 39 nM) in vitro at low nanomolar concentrations and significantly reduced glucose levels during an oral glucose tolerance test in normal mice.

STR3

STR3

PATENT

EP-1735322-B1

Example 2(R)-3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionamide

Image loading...

The title compound is obtained analogously to Example 1 by employing the following additional resolution step:

The racemic title E compound of Example 1, 3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid (10 g, 0.026 mol) in 1,4-dioxane (500 mL) is treated in a three necked 1 liter flask, equipped with heating mantle, water condenser, calcium chloride guard tube and mechanical stirrer with 3.18 g (0.026 mol) of (R)-(+)-1-phenylethylamine. This reaction mixture is then refluxed at 100°C for 1 h. The clear reaction solution is cooled to RT (27°C) and stirred for 10 h. The crystallized salt is collected by filtration under vacuum, washed with 5 mL of hexane and dried under vacuum to afford salt A.

The salt A is dissolved in 1,4-dioxane (500 mL) and heated at 100°C for 1 h. The clear reaction solution is cooled to RT (27°C) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 50 mL of hexane, and dried under vacuum to afford salt B.

The salt B is dissolved in 1,4-dioxane (290 mL) and heated at 100°C for 1 h. The clear reaction solution is cooled to RT (27°C) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 30 mL of hexane, and dried under vacuum to afford salt C.

The salt C is dissolved in 1,4-dioxane (100 mL) and heated at 100°C for 1 h. The clear reaction solution is cooled to RT (27°C) and stirred for 10 h. The crystallized product is collected by filtration under vacuum, washed with 30ml of hexane, and dried under vacuum to afford salt D.

The salt D is treated with aqueous hydrochloric acid solution (20 mL, 1 mL of concentrated hydrochloric acid diluted with 100 mL of water) and stirred for 5 min. The white solid precipitates out and is collected by vacuum filtration, washed with 10 mL of cold water, 5 mL of isopropanol and 20 mL of hexane, and dried under vacuum to yield the hydrochloride salt of (R)-(-)-3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid, salt E.

The salt E is neutralized by stirring with aqueous sodium bicarbonate solution (10 mL, 1 g of sodium bicarbonate dissolved in 120 mL of water) for 5 min. The precipitated solid is collected by filtration, washed with 10 mL of cold water, 100 mL of hexane, and dried to afford (R)-(-)-3-cyclopentyl-2-[4-(4-methyl-piperazine-1-sulfonyl)-phenyl]-propionic acid: m.p. 202.2-203.4°C.

Alternatively, the title compound may be obtained by the resolution of the racemic title compound of Example 1 using the following preparative chiral HPLC method:

  • Column: Chiralcel OD-R (250 x 20 mm) Diacel make, Japan;
  • Solvent A: water:methanol:acetonitrile (10:80:10 v/v/v);
  • Solvent B: water:methanol:acetonitrile (05:90:05 v/v/v);
  • Using gradient elution: gradient program (time, min / %B): 0/0, 20/0, 50/100, 55/0, 70/0;
  • Flow rate: 6.0 mL/min; and
  • Detection: by UV at 305 nm.

REFERENCES

US 7750020

WO-2005095418-A1

US-20080103167-A1

1 to 2 of 2
Patent ID Date Patent Title
US2015218151 2015-08-06 NOVEL PHENYLACETAMIDE COMPOUND AND PHARMACEUTICAL CONTAINING SAME
US7750020 2010-07-06 Sulfonamide-Thiazolpyridine Derivatives As Glucokinase Activators Useful The Treatment Of Type 2 Diabetes

///NOVARTIS, DIABETES, Sulfonamide-Thiazolpyridine Derivatives,  Glucokinase Activators, Treatment Of Type 2 Diabetes, 866772-52-3, Novartis Molecule, functionally liver selective glucokinase activators, treatment of type 2 diabetes , NVP-LBX192, LBX-192

c1(sc2nc(ccc2n1)OC)NC(C(c3ccc(cc3)S(=O)(=O)N4CCN(CC4)C)CC5CCCC5)=O

Share

Dr. Reddy’s Laboratories CEO G V Prasad has been recognized as one of India’s top 5 most valuable CEOs

 Uncategorized  Comments Off on Dr. Reddy’s Laboratories CEO G V Prasad has been recognized as one of India’s top 5 most valuable CEOs
Apr 052016
 

 

reddyDr. Reddy's Laboratories

CEO G V Prasad has been recognized as one of India’s top 5 most valuable CEOs Read more: bit.ly/CEOsRanking

http://businessworld.in/article/How-We-Ranked-The-CEOs/31-03-2016-92402/

Share

CFG 920, Novartis Scientists team up with Researchers at Aurigene, Bangalore, India,

 phase 2, Uncategorized  Comments Off on CFG 920, Novartis Scientists team up with Researchers at Aurigene, Bangalore, India,
Apr 052016
 

str1

CFG920,

Inhibitor Of Prostate Cancer With Fewer Cardiac Side Effects

Cas 1260006-20-9

Novartis
Target: CYP17/CYP11B2
Disease: Castration-resistant prostate cancer

MF C14H13ClN4O
MW: 288.0778

Elemental Analysis: C, 58.24; H, 4.54; Cl, 12.28; N, 19.40; O, 5.54

Steroid 17-alpha-hydroxylase inhibitors

CFG920 is a CYP17 inhibitor, is also an orally available inhibitor of the steroid 17-alpha-hydroxylase/C17,20 lyase (CYP17A1 or CYP17), with potential antiandrogen and antineoplastic activities. Upon oral administration, CYP17 inhibitor CFG920 inhibits the enzymatic activity of CYP17A1 in both the testes and adrenal glands, thereby inhibiting androgen production. This may decrease androgen-dependent growth signaling and may inhibit cell proliferation of androgen-dependent tumor cells.

https://clinicaltrials.gov/ct2/show/NCT01647789
NCT01647789: A Study of Oral CFG920 in Patients With Castration Resistant Prostate Cancer2012 

  • 09 Nov 2015Adverse events, efficacy and pharmacokinetics data from the phase I part of a phase I/II trial in Prostate cancer (Metastatic disease) presented at the 27th AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics (AACR-NCI-EORTC-2015)
  • 29 Jan 2013Phase-I clinical trials in Prostate cancer in Spain (PO)
  • 10 Dec 2012Phase-I clinical trials in Prostate cancer in Canada (PO)

In August 2015, preclinical data were presented at the 250th ACS meeting in Boston, MA. In monkeys, treatment with CFG-920 (3 mg/kg, po) showed good bioavailability with F value of 93%, Tmax of 0.5 h, Cmax of 1382 nM.dn and AUC of 2364 nM.h, while CFG-920 (10 mg/kg, po) showed F value of 183%, Cmax of 1179 nM.dn and Tmax of 1.04 h

 

str1

Bethany Halford on Twitter: “CFG920 – @Novartis CMOS for …

twitter.com

Bethany Halford on Twitter: “CFG920 – @Novartis CMOS for castration resistant prostate cancer #ACSBoston MEDI 1st disclosures http://t.co/XJJ3tCvpUk”

Novartis is developing CFG-920 (structure shown), an oral CYP17 inhibitor, for the potential treatment of metastatic castration-resistant prostate cancer. In March 2013, a phase I/II trial was initiated and at that time, the study was expected to complete in January 2015; in August 2015, clinical data were presented

2015 250th (August 19) Abs MEDI 341
Discovery of CFG920, a dual CYP17/CYP11B2 inhibitor, for the treatment of castration resistant prostate cancer
American Chemical Society National Meeting and Exposition
Christoph Gaul, Prakash Mistry, Henrik Moebitz, Mark Perrone, Bjoern Gruenenfelder, Nelson Guerreiro, Wolfgang Hackl, Peter Wessels, Estelle Berger, Mark Bock, Saumitra Sengupta, Venkateshwar Rao, Murali Ramachandra, Thomas Antony, Kishore Narayanan, Samiulla Dodheri, Aravind Basavaraju, Shekar Chelur

09338-scitech1-NovartisAcxd

CHEMISTRY COLLABORATORS
Novartis-Aurigene team: (from left) Brahma Reddy V, Thomas Antony, Murali Ramachandra, Venkateshwar Rao G, Wesley Roy Balasubramanian, Kishore Narayanan, Samiulla DS, Aravind AB, and Shekar Chelur. Not pictured: Björn Grünenfelder, Saumitra Sengupta, Nelson Guerreiro, Andrea Gerken, Mark Perrone, Mark Bock, Wolfgang Hackl, Henrik Möbitz, Peter Wessels, Christoph Gaul, Prakash Mistry, and Estelle Marrer.
Credit: Aurigene

Preclinical and clinical studies were performed to evaluate the efficacy of CFG-920, a dual cytochrome P450 (CYP)17 and CYP11B2 dual inhibitor, for the potential treatment of castration resistant prostate cancer. CFG-920 showed potent activity against human CYP17 and CYP11B2 enzymes with IC50 values of 0.023 and 0.034 microM, respectively. In monkeys, treatment with CFG-920 (3 mg/kg, po) showed good bioavailability (93%), Tmax of 0.5 h, Cmax of 1382 nM.dn and AUC of 2364 nM.h, while CFG-920 (10 mg/kg, po) showed F value of 183%, Cmax of 1179 nM.dn and Tmax of 1.04 h. In a phase I, first-in-man study, patients received continuous po dosing of CFG-920 (50 mg, bid) plus prednisone (5 mg) in 28-day cycles. At the time of presentation, CFG-920 was under phase II development.

 

Print
CFG920

WO 2010149755

09338-scitech1-Novartisgrocxd
Novartis team: (clockwise from left) Wolfgang Hackl, Henrik Möbitz, Peter Wessels, Christoph Gaul, Prakash Mistry, and Estelle Marrer., Credit: Novartis

Prostate cancer is the most commonly occurring cancer in men. Doctors often treat the metastatic stage of the disease by depriving the patient of sex hormones via chemical or surgical castration. But if it progresses far enough, the cancer can survive this therapy, transforming into the castration-resistant form. “Once the cancer becomes castration-resistant, the prognosis is poor,” said Novartis’s Christoph Gaul.

In recent years, CYP17, a bifunctional 17α-hydroxylase/17,20-lyase cytochrome P450 enzyme, has emerged as a target for treating castration-resistant prostate cancer. The enzyme catalyzes the biosynthesis of sex hormones, including testosterone, and blocking it can starve prostate cancer of the androgens it needs to thrive.

Johnson & Johnson’s CYP17 inhibitor, abiraterone acetate (Zytiga), a steroid that binds irreversibly to CYP17, was approved by the Food & Drug Administration in 2011. But Novartis scientists thought they could make a better CYP17 inhibitor, Gaul told C&EN. They teamed up with researchers at Aurigene, in Bangalore, India, and came up with their clinical candidate, CFG920.

Unlike abiraterone, CFG920 isn’t a steroid, and it inhibits CYP17 reversibly. It also reversibly inhibits another cytochrome P450 enzyme, CYP11B2, which is involved in the synthesis of the mineralocorticoids, hormones that regulate cardiac function.

Treating prostate cancer patients by lowering their androgen levels turns out to have negative cardiac side effects: Patients’ lipid metabolism is thrown off and their mineralocorticoid levels jump, leading to increases in blood pressure. Those changes can be stressful for the heart. “If prostate cancer patients don’t die because of the cancer, a lot of times they die because of cardiac disease,” Gaul said.

Because CFG920 also keeps mineralocorticoid levels in check, Novartis is hoping the drug candidate will ameliorate some of the cardiac side effects of inhibiting CYP17. The compound is currently in Phase I clinical trials.

PATENT

WO 2010149755

https://www.google.co.in/patents/WO2010149755A1?cl=en

Example 58

Prύpιn”ation ofI'(2’ChIoroψ}ri(ibi-^’\l)’3’f4’metMψ}τUin’3’yl)-imiJazoliJin’2’θne (5HA)-

Figure imgf000079_0001

Using the same reaction conditions as in Example 14. 1-(4-methyl-pyridin-3-yl)- itnida/olidin-2-onc ().-.!.4b: 600 mg. 3.3898 mmol) uas reacted with 2-chloro-4-iodo- py.idine (974 mg.4.067 mmol). 1 , 4-dioxane (60 mL). copper iodide (65 mg, 0.3398 mmol), /r<w.v-1.2-diamino cycK)hexane (0.12 ml,, 1.0169 mmol) and potassium phosphate (2.15 g, 10.1694 mmol) to afford 810 mg of the product (83% yield).

1H NMR (C1DCI3. 300 Mi l/): 6 8.5-8.4 (m. 211). 8.3 (d. IH), 7.6-7.5 (m, 2H). 7.2 (S. 111). 4.1-3.9 (ni. 4H), 2.35 <s. 3H)

LCVIS puιϊt>: 90.8%. nι-7 – 289.1 (M M)

HPl C: 97.14%

REFERENCES

1: Gomez L, Kovac JR, Lamb DJ. CYP17A1 inhibitors in castration-resistant prostate cancer. Steroids. 2015 Mar;95:80-7. doi: 10.1016/j.steroids.2014.12.021. Epub 2015 Jan 3. Review. PubMed PMID: 25560485; PubMed Central PMCID: PMC4323677.

2: Yin L, Hu Q, Hartmann RW. Recent progress in pharmaceutical therapies for castration-resistant prostate cancer. Int J Mol Sci. 2013 Jul 4;14(7):13958-78. doi: 10.3390/ijms140713958. Review. PubMed PMID: 23880851; PubMed Central PMCID: PMC3742227.

///////CFG-920,  CYP17 inhibitor (prostate cancer), Novartis, CFG 920, Novartis scientists,   team up , researchers ,  Aurigene, Bangalore, India,

Share

GDC-0919; NLG-919; RG-6078

 Uncategorized  Comments Off on GDC-0919; NLG-919; RG-6078
Apr 052016
 

img
MF C18H22N2O
MW: 282.17321

GDC-0919; NLG-919; RG-6078, GDC0919; GDC-0919; GDC 0919; NLG919; NLG 919; NLG-919; RG6078; RG-6078; RG 6078.

 1-cyclohexyl-2-(5H-imidazo[5,1-a]isoindol-5-yl)ethanol
CAS No.1402836-58-1

GDC-0919, also known as NLG919 and RG6078, is an orally available inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), with potential immunomodulating and antineoplastic activities. Upon administration, NLG919 targets and binds to IDO1, a cytosolic enzyme responsible for the oxidation of the essential amino acid tryptophan into kynurenine. By inhibiting IDO1 and decreasing kynurenine in tumor cells, this agent increases tryptophan levels, restores the proliferation and activation of various immune cells, including dendritic cells (DCs), natural killer (NK) cells, T-lymphocytes, and causes a reduction in tumor-associated regulatory T-cells (Tregs). Activation of the immune system, which is suppressed in many cancers, may induce a cytotoxic T-lymphocyte (CTL) response against the IDO1-expressing tumor cells

  • Originator Lankenau Institute for Medical Research
  • Developer Genentech; NewLink Genetics Corporation
  • Class Antineoplastics; Small molecules
  • Mechanism of Action Immunomodulators; Indoleamine-pyrrole 2,3-dioxygenase inhibitors

Phase I Solid tumours

Patent ID Date Patent Title
US2015210769 2015-07-30 ANTIBODY MOLECULES TO PD-1 AND USES THEREOF
US2014066625 2014-03-06 Fused Imidazole Derivatives Useful as IDO Inhibitors
  • 27 Sep 2015 Pharmacokinetics results from a phase-I clinical trial in Solid tumours presented at the European Cancer Congress 2015 (ECC-2015)
  • 27 Sep 2015 Positive efficacy and safety results from a phase-I clinical trial in Solid tumours presented at the European Cancer Congress 2015 (ECC-2015)
  • 31 Jul 2015 Phase-I clinical trials in Solid tumours (Combination therapy, Late-stage disease, Second-line therapy or greater) in USA (PO) (NCT02471846)

 

PATENT

http://www.google.com/patents/WO2012142237A1?cl=en

str1

PATENT

US-20160002249-A1 / 2016-01-07

Fused Imidazole Derivatives Useful as IDO Inhibitors

1304Image loading...1-cyclohexyl-2-(5H-imidazo[5,1- a]isoindol-5-yl)ethanol79 1H NMR (a mixture of diastereomers) 1.10-1.37 (m, 6H), 1.66-1.80 (m, 5H), 2.05 (m, 2H), 2.15 (m, 1H), 3.72 (m, 1H), 5.36 and 5.46 (two m, 1H), 7.16 (s, 1H), 7.25 (m, 1H), 7.34 (m, 1H), 7.43 (d, 1H, J = 7.6 Hz), 7.54 (d, 1H, J = 7.6 Hz), 7.80 (s, 1H)

 

 

 

WO2011056652A1 * Oct 27, 2010 May 12, 2011 Newlink Genetics Imidazole derivatives as ido inhibitors
WO2012142237A1 * Apr 12, 2012 Oct 18, 2012 Newlink Geneticks Corporation Fused imidazole derivatives useful as ido inhibitors
WO2014159248A1 Mar 10, 2014 Oct 2, 2014 Newlink Genetics Corporation Tricyclic compounds as inhibitors of immunosuppression mediated by tryptophan metabolization
US8722720 Oct 27, 2010 May 13, 2014 Newlink Genetics Corporation Imidazole derivatives as IDO inhibitors
US9260434 Oct 14, 2013 Feb 16, 2016 Newlink Genetics Corporation Fused imidazole derivatives useful as IDO inhibitors
US20140066625 * Oct 14, 2013 Mar 6, 2014 Newlink Genetics Corporation Fused Imidazole Derivatives Useful as IDO Inhibitors
US20160002249 * Jul 8, 2015 Jan 7, 2016 Newlink Genetics Corporation Fused Imidazole Derivatives Useful as IDO Inhibitors

REFERENCES

Nature Reviews Drug Discovery14,373(2015)doi:10.1038/nrd4658

http://www.ncbi.nlm.nih.gov/pubmed/21517759

http://www.roche.com/irp150128-annex.pdf

/////CRD1152, CRD 1152, CRD-1152, Curadev,  Research Collaboration, Licensing Agreement, Develop,  Cancer Immunotherapeutic, IDO1 and TDO inhibitors

img

OC(C1CCCCC1)CC(C2=C3C=CC=C2)N4C3=CN=C4

 

 

 

 

 

/////GDC-0919; NLG-919; RG-6078

Share

CRD 1152, CURADEV PHARMA PRIVATE LTD

 cancer, Uncategorized  Comments Off on CRD 1152, CURADEV PHARMA PRIVATE LTD
Apr 052016
 

Several candidates……One is …..CRD1152

ONE OF THEM IS CRD 1152

Kynurenine pathway regulators (solid tumors)

Compound 2

CAS1638121-21-7

US159738837

N3-(3-Chloro-4- fluorophenyl) furo[2,3- c]pyridine-2,3- diamine

COMPD 190

CAS 1638118-99-6

US159738837

COMPD248

US159738837

7-Chloro-N3- (3-chloro-4- fluorophenyl) furo[2,3- c]pyridine-2,3- diamine,  166

DMSO-d6: δ 7.87 (d, J = 5.1 Hz, 1H), 7.25 (s, 2H), 7.16-7.10 (m, 2H), 6.88 (d, J = 5.1 Hz, 1H), 6.59 (dd, J′ = 6.2 Hz, J″ = 2.6 Hz, 1H), 6.48 (dt, J′ = 8.8 Hz, J″ = 6.7 Hz, J′′′ = 3.4 Hz, 1H) M + H] 312

US159738837

OR

N3-(3,4- difluorophenyl)- 7-(pyridin-4- yl)furo[2,3- c]pyridine-2,3- diamine, 184

CD3CN: δ 8.72 (s, 2H), 8.26 (s, 3H), 7.07-7.03 (m, 2H), 6.47-6.40 (m, 2H), 5.74 (s, 1H), 5.55 (s, 2H) M + H] 339

US159738837

OR

COMPD73

CAS 1638117-85-7

US159738837

Several candidates………..CRD1152

67

66

Company Curadev Pharma Pvt. Ltd.
Description Small molecule dual indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO1; IDO) inhibitor
Molecular Target Indoleamine 2,3-dioxygenase (INDO) (IDO) ; Tryptophan 2,3-dioxygenase (TDO2) (TDO)
Mechanism of Action Indoleamine 2,3-dioxygenase (INDO) inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Preclinical
Standard Indication Cancer (unspecified)
Indication Details Treat cancer
Regulatory Designation
Partner Roche

Hoffmann-La Roche partners with Curadev Pharma Ltd. for IDO1 and TDO inhibitors (April 20, 2015)

Curadev Pharma Pvt Ltd., founded in 2010 and headquartered in New Delhi, announced that it has entered into a research collaboration and exclusive license agreement with Roche for the development and commercialization of IDO1 and TDO inhibitors to treat cancer. The agreement covers the development of CRD1152, the lead preclinical immune tolerance inhibitor and a research collaboration with Roche’s research and early development organization to further explore the IDO and TDO pathways.

IDO1 (indoleamine-2,3-dioxygenase-1) and TDO (tryptophan-2,3-dioxygenase) are enzymes that mediate cancer-induced immune suppression. This mechanism is exploited by tumor cells as well as certain type of immune cells, limiting the anti-tumor immune response. Dual inhibition of the IDO1 and TDO pathways promises to maintain the immune response, prevent local tumor immune escape and potentially avoid resistance to other immunotherapies when used in combination, and could lead to new treatment options for cancer patients. Curadev’s preclinical lead-compound, a small-molecule that shows potent inhibition of the two rate-limiting enzymes in the tryptophan to kynurenine metabolic pathways, has the potential for mono therapy as well as combination with Roche’s broad oncology pipeline and portfolio.

Under the terms of agreement, which includes a research collaboration with Roche’s research and early development organization, Curadev will receive an upfront payment of $25 million and will be eligible to receive up to $530 million in milestone payments, as well as escalating royalties potentially reaching double digits for the first product from the collaboration developed and commercialized by Roche. Curadev is also eligible for milestones and royalties on any additional products resulting from the research collaboration.

Curadev Announces Research Collaboration and Licensing Agreement to Develop Cancer Immunotherapeutic

Curadev’s dual IDO and TDO immune tolerance inhibitor – a novel approach in cancer immunotherapy

Apr 20, 2015, 06:30 ET from Curadev

NEW DELHI, India, April 20, 2015 /PRNewswire/ —

Curadev Pharma Private Ltd. today announced that it has entered into a research collaboration and exclusive license agreement with Roche for the development and commercialization of IDO1 and TDO inhibitors. The agreement covers the development of the lead preclinical immune tolerance inhibitor and a research collaboration with Roche’s research and early development organization to further explore the IDO and TDO pathways.

IDO1 (indoleamine-2, 3-dioxygenase-1) and TDO (tryptophan-2, 3-dioxygenase) are enzymes that mediate cancer-induced immune suppression. This mechanism is exploited by tumor cells as well as certain type of immune cells, limiting the anti-tumor immune response.

Dual inhibition of the IDO1 and TDO pathways promises to maintain the immune response, prevent local tumor immune escape and potentially avoid resistance to other immunotherapies when used in combination, and could lead to new treatment options for cancer patients. Curadev’s preclinical lead-compound, a small-molecule that shows potent inhibition of the two rate-limiting enzymes in the tryptophan – to kynurenine metabolic pathways, has the potential for mono therapy as well as combination with Roche’s broad oncology pipeline and portfolio.

“We are very excited to be working with the global leader in oncology with their unrivalled expertise in clinical development,” said Arjun Surya, PhD, Chief Scientific Officer, Curadev. “The collaboration acknowledges our focused research efforts on patient-critical drug targets that have yielded a drug candidate that could make a significant difference in the development of novel treatments for patients suffering from cancer.”

Under the terms of agreement, which includes a research collaboration with Roche’s research and early development organization to further extend Curadev’s findings, Curadev will receive an upfront payment of $25 million and will be eligible to receive up to $530 million in milestone payments based on achievement of certain predetermined events and sales levels as well as escalating royalties potentially reaching double digits for the first product from the collaboration developed and commercialized by Roche. Curadev would also be eligible for milestones and royalties on any additional products resulting from the research collaboration. Roche will fund future research, development, manufacturing and commercialization costs and will also provide additional research funding to Curadev for support of the research collaboration.

About Curadev

Headquartered in New Delhi, India, Curadev Pharma Private Limited was founded in 2010 by a team of professionals from the pharmaceutical and biotech sectors with the mission to improve human health and enhance the quality of human life by accelerating the discovery and delivery of new drugs. Curadev focuses on the creation and out-licensing of pre-IND assets and IND packages for drug development.

For further information:

Curadev Partnering

Manish Tandon – VP and Chief Financial Officer, manish@curadev.in

PATENT

US20160046596) INHIBITORS OF THE KYNURENINE PATHWAY

https://patentscope.wipo.int/search/en/detail.jsf?docId=US159738837&recNum=2&maxRec=17&office=&prevFilter=&sortOption=Pub+Date+Desc&queryString=FP%3A%28curadev%29&tab=PCTDescription

Monali Banerjee
Sandip Middya
Ritesh Shrivastava
Sushil Raina
Arjun Surya
Dharmendra B. Yadav
Veejendra K. Yadav
Kamal Kishore Kapoor
Aranapakam Venkatesan
Roger A. Smith
Scott K. Thompson

ONE ………….Example 2

Synthesis of N3-(3-Chloro-4-fluoro-phenyl)-furo[2,3-c]pyridine-2,3-diamine (Compound 2)


Step 1: 3-Methoxymethoxy-pyridine


      To a stirred solution of 3-hydroxypyridine (60 g, 662.9 mmol) in THF:DMF (120:280 mL) at 0° C. was added t-BuOK (81.8 gm, 729.28 mmol) portion-wise. After stirring the reaction mixture for 15 min, methoxymethyl chloride (52 mL, 696.13 mmol) was added to it at 0° C. and the resulting mixture was stirred for 1 hr at 25° C. Reaction mixture was diluted with water and extracted with ethyl acetate (4×500 mL). The organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure to afford 100 g crude which was purified by column chromatography using silica (100-200 mesh) and 10% EtOAc-hexane as eluent to afford 3-methoxymethoxy-pyridine (54 g) as pale brown liquid. LCMS: 140 (M+H).

Step 2: 3-Methoxymethoxy-pyridine-4-carbaldehyde


      To a stirred solution of 3-methoxymethoxypyridine (2 g, 14.3885 mmol) in anhydrous THF (40 mL) was added TMEDA (1.83 g, 15.82 mmol) at 25° C. The reaction mixture was cooled to −78° C., n-BuLi (7.3 mL, 15.82 mmol, 2.17 M in hexane) was added dropwise manner maintaining the temperature −78° C. After stirring for 2 hr at −78° C., DMF (1.52 g, 20.86 mmol) was added to it and stirred for 2 hr at 25° C. Reaction mixture was cooled to −40° C. and saturated ammonium chloride solution was added drop wise. The reaction mass was extracted with ethyl acetate (250 mL×2), EtOAc part was washed with water followed by brine, dried over sodium sulfate and concentrated under reduced pressure to afford 3 g of crude product which was passed through a pad of silica (100-200 mesh) using 10% EtOAc-hexane as eluent to afford 1.6 g of 3-methoxymethoxy-pyridine-4-carbaldehyde as pale yellow liquid. GC-MS: 167 (m/z).

Step 3: 3-Hydroxy-pyridine-4-carbaldehyde


      To a stirred solution of 3-methoxymethoxypyridine-4-carbaldehyde (11 g, 65.83 mmol) in THF (50 mL) was added 3N HCl (100 mL) and stirred at 60° C. for 1 hr. The reaction mixture was cooled under ice bath and pH was adjusted to 7 with solid K2CO3. Resulting mixture was extracted with EtOAc (250 mL×5). The organic layer was dried over sodium sulfate, concentrated under reduced pressure to afford 15 g of crude which was purified by column chromatography using silica gel (100-200 mesh) and 23% EtOAc/hexane as eluent to afford 4 g of 3-hydroxy-pyridine-4-carbaldehyde as pale yellow solid. GC-MS: 123 (m/z), 1H-NMR (DMSO-d6, 400 MHz): δ 11.04 (bs, 1H), 10.37 (s, 1H), 8.46 (s, 1H), 8.20 (d, 1H, J=4.88 Hz), 7.46 (d, 1H, J=4.88 Hz). GC-FID: 99.51%.

Step 4: 4-{[3-Chloro-4-fluoro-phenylimino]-methyl}-pyridin-3-ol


      3-Hydroxypyridine-4-carbaldehyde (3 g, 24.39 mmol) was taken in mixed solvent (TFE (20 mL):MeCN (20 mL)) and 4-fluoro-3-chloroaniline (3.55 g, 24.39 mmol) was added to it at 25° C. The resulting mixture was stirred at this temperature for 1 hr. The reaction mass was concentrated and purified by triturating with n-pentane to afford 6 g of 4-{[3-chloro-4-fluoro-phenylimino]-methyl}-pyridin-3-ol). LCMS: 251.2 (M+H).

Step 5: N3-(3-Chloro-4-fluoro-phenyl)-furo[2,3-c]pyridine-2,3-diamine


      To a stirred solution of 4-{[3-chloro-4-fluoro-phenylimino]-methyl}-pyridin-3-ol (6 g, 24 mmol) in mixed solvent [DCM (10 mL):TFE (10 mL)] was added TMSCN (10.5 mL, 84 mmol) at 25° C. The reaction mixture was stirred 3 hr at 25° C., concentrated, and the crude material was triturated with n-pentane to provide 4.9 g (73% yield) of N3-(3-chloro-4-fluoro-phenyl)-furo[2,3-c]pyridine-2,3-diamine as pale pink solid. LCMS: 278 (M+H), HPLC: 98.65%, 1H-NMR (DMSO-d6, 400 MHz): δ 8.41 (s, 1H), 8.06 (d, 1H, J=5.08 Hz), 7.14-7.10 (m, 2H), 6.91 (s, 2H), 6.86 (d, 1H, J=5.08 Hz), 6.56-6.54 (m, 1H), 6.48-6.45 (m, 1H).

 

 

Monali Banerjee – Director, R&D

Ms. Banerjee has more than 10 years of research experience, during which she has held positions of increasing responsibility. Her past organizations include TCG Lifesciences (Chembiotek) and Sphaera Pharma. Ms. Banerjee is a versatile scientist with a deep understanding of the fundamental issues that underlie various aspects of drug discovery. At Curadev, she has been responsible for target selection, patent analysis, pharmacophore design, assay development, ADME/PK and in vivo and in vitro pharmacology. Ms. Banerjee holds a Masters in Biochemistry and a Bachelors in Chemistry both from Kolkata University.

writeup

The essential amino acid Tryptophan (Trp) is catabolized through the kynurenine (KYN) pathway. The initial rate-limiting step in the kynurenine pathway is performed by heme-containing oxidoreductase enzymes, including tryptophan 2,3-dioxygenase (TDO), indoleamine 2,3-dioxygenase-1 (IDO1), and indoleamine 2,3-dioxygenase-2 (IDO2). IDO1 and IDO2 share very limited homology with TDO at the amino acid level and, despite having different molecular structures, each enzyme has the same biochemical activity in that they each catalyze tryptophan to form N-formylkynurenine. IDO1, IDO2, and/or TDO activity alter local tryptophan concentrations, and the build-up of kynurenine pathway metabolites due to the activity of these enzymes can lead to numerous conditions associated with immune suppression.
      IDO1 and TDO are implicated in the maintenance of immunosuppressive conditions associated with the persistence of tumor resistance, chronic infection, HIV infection, malaria, schizophrenia, depression as well as in the normal phenomenon of increased immunological tolerance to prevent fetal rejection in utero. Therapeutic agents that inhibit IDO1, IDO2, and TDO activity can be used to modulate regulatory T cells and activate cytotoxic T cells in immunosuppressive conditions associated with cancer and viral infection (e.g. HIV-AIDS, HCV). The local immunosuppressive properties of the kynurenine pathway and specifically IDO1 and TDO have been implicated in cancer. A large proportion of primary cancer cells have been shown to overexpress IDO1. In addition, TDO has recently been implicated in human brain tumors.
      The earliest experiments had proposed an anti-microbial role for IDO1, and suggested that localized depletion of tryptophan by IDO1 led to microbial death (Yoshida et al., Proc. Natl. Acad. Sci. USA, 1978, 75(8):3998-4000). Subsequent research led to the discovery of a more complex role for IDO1 in immune suppression, best exemplified in the case of maternal tolerance towards the allogeneic fetus where IDO1 plays an immunosuppressive role in preventing fetal rejection from the uterus. Pregnant mice dosed with a specific IDO1 inhibitor rapidly reject allogeneic fetuses through induction of T cells (Munn et al., Science, 1998, 281(5380): 1191-3). Studies since then have established IDO1 as a regulator of certain disorders of the immune system and have discovered that it plays a role in the ability of transplanted tissues to survive in new hosts (Radu et al., Plast. Reconstr. Surg., 2007 June, 119(7):2023-8). It is believed that increased IDO1 activity resulting in elevated kynurenine pathway metabolites causes peripheral and ultimately, systemic immune tolerance. In-vitro studies suggest that the proliferation and function of lymphocytes are exquisitely sensitive to kynurenines (Fallarino et al., Cell Death and Differentiation, 2002, 9(10):1069-1077). The expression of IDO1 by activated dendritic cells suppresses immune response by mechanisms that include inducing cell cycle arrest in T lymphocytes, down regulation of the T lymphocyte cell receptor (TCR) and activation of regulatory T cells (T-regs) (Terness et al., J. Exp. Med., 2002, 196(4):447-457; Fallarino et al., J. Immunol., 2006, 176(11):6752-6761).
      IDO1 is induced chronically by HIV infection and in turn increases regulatory T cells leading to immunosuppression in patients (Sci. Transl. Med., 2010; 2). It has been recently shown that IDO1 inhibition can enhance the level of virus specific T cells and concomitantly reduce the number of virus infected macrophages in a mouse model of HIV (Potula et al., 2005, Blood, 106(7):2382-2390). IDO1 activity has also been implicated in other parasitic infections. Elevated activity of IDO1 in mouse malaria models has also been shown to be abolished by in vivo IDO1 inhibition (Tetsutani K., et al., Parasitology. 2007 7:923-30.
      More recently, numerous reports published by a number of different groups have focused on the ability of tumors to create a tolerogenic environment suitable for survival, growth and metastasis by activating IDO1 (Prendergast, Nature, 2011, 478(7368):192-4). Studies of tumor resistance have shown that cells expressing IDO1 can increase the number of regulatory T cells and suppress cytotoxic T cell responses thus allowing immune escape and promoting tumor tolerance.
      Kynurenine pathway and IDO1 are also believed to play a role in maternal tolerance and immunosuppressive process to prevent fetal rejection in utero (Munn et al., Science, 1998, 281(5380):1191-1193). Pregnant mice dosed with a specific IDO1 inhibitor rapidly reject allogeneic fetuses through suppression of T cells activity (Munn et al., Science, 1998, 281(5380):1191-1193). Studies since then have established IDO1 as a regulator of immune-mediated disorders and suggest that it plays a role in the ability of transplanted tissues to survive in new hosts (Radu et al., Plast. Reconstr. Surg., 2007 June, 119(7):2023-8).
      The local immunosuppressive properties of the kynurenine pathway and specifically IDO1 and TDO have been implicated in cancer. A large proportion of primary cancer cells overexpress IDO1 and/or TDO (Pilotte et al., Proc. Natl. Acad. Sci. USA, 2012, Vol. 109(7):2497-2502). Several studies have focused on the ability of tumors to create a tolerogenic environment suitable for survival, growth and metastasis by activating IDO1 (Prendergast, Nature, 2011, 478:192-4). Increase in the number of T-regs and suppression of cytotoxic T cell responses associated with dysregulation of the Kynurenine pathway by overexpression of IDO1 and/or TDO appears to result in tumor resistance and promote tumor tolerance.
      Data from both clinical and animal studies suggest that inhibiting IDO1 and/or TDO activity could be beneficial for cancer patients and may slow or prevent tumor metastases (Muller et al., Nature Medicine, 2005, 11(3):312-319; Brody et al., Cell Cycle, 2009, 8(12):1930-1934; Witkiewicz et al., Journal of the American College of Surgeons, 2008, 206:849-854; Pilotte et al., Proc. Natl. Acad. Sci. USA, 2012, Vol. 109(7):2497-2502). Genetic ablation of the IDO1 gene in mice (IDO1−/−) resulted in decreased incidence of DMBA-induced premalignant skin papillomas (Muller et al., PNAS, 2008, 105(44):17073-17078). Silencing of IDO1 expression by siRNA or a pharmacological IDO1 inhibitor 1-methyl tryptophan enhanced tumor-specific killing (Clin. Cancer Res., 2009, 15(2). In addition, inhibiting IDO1 in tumor-bearing hosts improved the outcome of conventional chemotherapy at reduced doses (Clin. Cancer Res., 2009, 15(2)). Clinically, the pronounced expression of IDO1 found in several human tumor types has been correlated with negative prognosis and poor survival rate (Zou, Nature Rev. Cancer, 2005, 5:263-274; Zamanakou et al., Immunol. Lett. 2007, 111(2):69-75). Serum from cancer patients has higher kynurenine/tryptophan ratio, a higher number of circulating T-regs, and increased effector T cell apoptosis when compared to serum from healthy volunteers (Suzuki et al., Lung Cancer, 2010, 67:361-365). Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase has been studied by Pilotte et al. (Pilotte et al., Proc. Natl. Acad. Sci. USA, 2012, Vol. 109(7):2497-2502). Thus, decreasing the rate of kynurenine production by inhibiting IDO1 and/or TDO may be beneficial to cancer patients.
      IDO1 and IDO2 are implicated in inflammatory diseases. IDO1 knock-out mice don’t manifest spontaneous disorders of classical inflammation and existing known small molecule inhibitors of IDO do not elicit generalized inflammatory reactions (Prendergast et al. Curr Med Chem. 2011; 18(15):2257-62). Rather, IDO impairment alleviates disease severity in models of skin cancers promoted by chronic inflammation, inflammation-associated arthritis and allergic airway disease. Moreover, IDO2 is a critical mediator of autoantibody production and inflammatory pathogenesis in autoimmune arthritis. IDO2 knock-out mice have reduced joint inflammation compared to wild-type mice due to decreased pathogenic autoantibodies and Ab-secreting cells (Merlo et al. J. Immunol. (2014) vol. 192(5) 2082-2090). Thus, inhibitors of IDO1 and IDO2 are useful in the treatment of arthritis and other inflammatory diseases.
      Kynurenine pathway dysregulation and IDO1 and TDO play an important role in the brain tumors and are implicated in inflammatory response in several neurodegenerative disorders including multiple sclerosis, Parkinson’s disease, Alzheimer’s disease, stroke, amyotrophic lateral schlerosis, dementia (Kim et al., J. Clin. Invest, 2012, 122(8):2940-2954; Gold et al., J. Neuroinflammation, 2011, 8:17; Parkinson’s Disease, 2011, Volume 2011). Immunosuppression induced by IDO1 activity and the Kynurenine metabolites in the brain may be treated with inhibitors of IDO1 and/or TDO. For example, circulating T-reg levels were found to be decreased in patient with glioblastoma treated with anti-viral agent inhibitors of IDO1 (Soderlund, et al., J. Neuroinflammation, 2010, 7:44).
      Several studies have found Kynurenine pathway metabolites to be neuroactive and neurotoxic. Neurotoxic kynurenine metabolites are known to increase in the spinal cord of rats with experimental allergic encephalomyelitis (Chiarugi et al., Neuroscience, 2001, 102(3):687-95). The neurotoxic effects of Kynurenine metabolities is exacerbated by increased plasma glucose levels. Additionally, changes in the relative or absolute concentrations of the kynurenines have been found in several neurodegenerative disorders, such as Alzheimer’s disease, Huntington’s disease and Parkinson’s disease, stroke and epilepsy (Németh et al., Central Nervous System Agents in Medicinal Chemistry, 2007, 7:45-56; Wu et al. 2013; PLoS One; 8(4)).
      Neuropsychiatric diseases and mood disorders such as depression and schizophrenia are also said to have IDO1 and Kynurenine dysregulation. Tryptophan depletion and deficiency of neurotransmitter 5-hydroxytryptamine (5-HT) leads to depression and anxiety. Increased IDO1 activity decreases the synthesis of 5-HT by reducing the amount of Tryptophan availability for 5-HT synthesis by increasing Tryp catabolism via the kynurenine pathway (Plangar et al. (2012) Neuropsychopharmacol Hung 2012; 14(4): 239-244). Increased IDO1 activity and levels of both kynurenine and kynurenic acid have been found in the brains of deceased schizophrenics (Linderholm et al., Schizophrenia Bulletin (2012) 38: 426-432)). Thus, inhibition of IDO1, IDO1, and TDO may also be an important treatment strategy for patients with neurological or neuropsychiatric disease or disorders such as depression and schizophrenia as well as insomnia.
      Kynurenine pathway dysregulation and IDO1 and/or TDO activity also correlate with cardiovascular risk factors, and kynurenines and IDO1 are markers for Atherosclerosis and other cardiovascular heart diseases such as coronary artery disease (Platten et al., Science, 2005, 310(5749):850-5, Wirlietner et al. Eur J Clin Invest. 2003 July; 33(7):550-4) in addition to kidney disease. The kynurenines are associated with oxidative stress, inflammation and the prevalence of cardiovascular disease in patients with end-stage renal disease (Pawlak et al., Atherosclerosis, 2009, (204)1:309-314). Studies show that kynurenine pathway metabolites are associated with endothelial dysfunction markers in the patients with chronic kidney disease (Pawlak et al., Advances in Medical Sciences, 2010, 55(2):196-203).

///////CRD1152, CRD-1152, CRD 1152, CURADEV PHARMA PRIVATE LTD, ROCHE, IDO1 and TDO inhibitors, COLLABORATION, CANCER, indoleamine-2,3-dioxygenase-1, Hoffmann-La Roche, kynurenine pathway regulators, solid tumors

Share

AUNP-12 from Aurigene Discovery Technologies Limited

 Uncategorized  Comments Off on AUNP-12 from Aurigene Discovery Technologies Limited
Apr 042016
 

 

 

AUNP-12

AUR-012; Aurigene-012; NP-12, Aurigene; PD-1 inhibitor peptide (cancer), Aurigene; PD-1 inhibitor peptide (cancer), Aurigene/ Pierre Fabre; W-014A

 

Company Aurigene Discovery Technologies Ltd.
Description A programmed cell death 1 (PDCD1; PD-1; CD279) peptide antagonist
Molecular Target Programmed cell death 1 (PD-1) (PDCD1) (CD279)
Mechanism of Action Programmed cell death 1 (PD-1) antagonist
Therapeutic Modality Peptide
Latest Stage of Development Preclinical
Standard Indication Cancer (unspecified)
Indication Details Treat cancer
Regulatory Designation
Partner Laboratoires Pierre Fabre S.A.

Aurigene Discovery Technologies Limited

INNOVATOR

 

 

  • Programmed Cell Death 1 or PD-1 (also referred to as PDCD1) is a 50 to 55 kD type I membrane glycoprotein (Shinohara T et al, Genomics, 1994, Vol. 23, No. 3, pp. 704-706). PD-1 is a receptor of the CD28 superfamily that negatively regulates T cell antigen receptor signalling by interacting with the specific ligands and is suggested to play a role in the maintenance of self tolerance.
  • PD-1 peptide relates to almost every aspect of immune responses including autoimmunity, tumour immunity, infectious immunity, transplantation immunity, allergy and immunological privilege.
  • The PD-1 protein’s structure comprise of—

      • an extracellular IgV domain followed by
      • a transmembrane region and
      • an intracellular tail
  • The intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which suggests that PD-1 negatively regulates TCR signals. Also, PD-1 is expressed on the surface of activated T cells, B cells, and macrophages, (Y. Agata et al., Int Immunol 8, 765, May 1996) suggesting that compared to CTLA-4 ((Cytotoxic T-Lymphocyte Antigen 4, also known as CD152 (Cluster of differentiation 152) is a protein that also plays an important regulatory role in the immune system), PD-1 more broadly negatively regulates immune responses.
  • PD-1 has two ligands, PD-L1 (Programmed Death Ligand for PDCD1L1 or B7-H1) (Freeman G J et al, Journal of Experimental Medicine, 2000, Vol. 19, No. 7, pp. 1027-1034) and PD-L2 (Programmed Death Ligand 2 or PDCD1L2 or B7-DC) (Latchman Y et al, Nature Immunology, 2001, Vol. 2, No. 3, pp. 261-267), which are members of the B7 family. PD-L1 is known to be expressed not only in immune cells, but also in certain kinds of tumour cell lines (such as monocytic leukaemia-derived cell lines, mast cell tumour-derived cell lines, hematoma-derived cell lines, neuroblastoma-derived cell lines, and various mammary tumour-derived cell lines) and in cancer cells derived from diverse human cancer tissues (Latchman Y et al, Nature Immunology, 2001, Vol. 2, No. 3, pp. 261-267) and on almost all murine tumour cell lines, including PA1 myeloma, P815 mastocytoma, and B16 melanoma upon treatment with IFN-γ (Y. Iwai et al., Proc Natl Acad Sci USA 99, 12293, Sep. 17, 2002 and C. Blank et al., Cancer Res 64, 1140, February, 2004). Similarly PD-L2 expression is more restricted and is expressed mainly by dendritic cells and a few tumour cell lines. PD-L2 expression has been verified in Hodgkin’s lymphoma cell lines and others. There is a hypothesis that some of the cancer or tumour cells take advantage from interaction between PD-1 and PD-L1 or PD-L2, for suppressing or intercepting T-cell immune responses to their own (Iwai Y et al, Proceedings of the National Academy of Science of the United States of America, 2002, Vol. 99, No. 19, pp. 12293-12297).
  • Tumour cells and virus (including HCV and HIV) infected cells are known to express the ligand for PD-1 (to create Immunosuppression) in order to escape immune surveillance by host T cells. It has been reported that the PD-1 gene is one of genes responsible for autoimmune diseases like systemic lupus erythematosis (Prokunina et al, Nature Genetics, 2002, Vol. 32, No. 4, 666-669). It has also been indicated that PD-1 serves as a regulatory factor for the onset of autoimmune diseases, particularly for peripheral self-tolerance, on the ground that PD-1-deficient mice develop lupus autoimmune diseases, such as glomerulonephritis and arthritis (Nishimura H et al, International Immunology, 1998, Vol. 10, No. 10, pp. 1563-1572; Nishimura H et al, Immunity, 1999, Vol. 11, No. 2, pp. 141-151), and dilated cardiomyopathy-like disease (Nishimura H et al, Science, 2001, Vol. 291, No. 5502, pp. 319-332).
  • Hence, in one approach, blocking the interaction of PD-1 with its ligand (PD-L1, PD-L2 or both) may provide an effective way for specific tumour and viral immunotherapy.
  • Wood et al in U.S. Pat. No. 6,808,710 discloses method for down modulating an immune response comprising contacting an immune cell expressing PD-1 with an antibody that binds to PD-1, in multivalent form, such that a negative signal is transduced via PD-1 to thereby down modulate the immune response. Such an antibody may be a cross-linked antibody to PD-1 or an immobilized antibody to PD-1.
  • Freeman et al in U.S. Pat. No. 6,936,704 and its divisional patent U.S. Pat. No. 7,038,013 discloses isolated nucleic acids molecules, designated B7-4 nucleic acid molecules, which encode novel B7-4 polypeptides, isolated B7-4 proteins, fusion proteins, antigenic peptides and anti-B7-4 antibodies, which co-stimulates T cell proliferation in vitro when the polypeptide is present on a first surface and an antigen or a polyclonal activator that transmits an activating signal via the T-cell receptor is present on a second, different surface.
  • There are some reports regarding substances inhibiting immunosuppressive activity of PD-1, or interaction between PD-1 and PD-L1 or PD-L2, as well as the uses thereof. A PD-1 inhibitory antibody or the concept of a PD-1 inhibitory peptide is reported in WO 01/14557, WO 2004/004771, and WO 2004/056875. On the other hand, a PD-L1 inhibitory antibody or a PD-L1 inhibitory peptide is reported in WO 02/079499, WO 03/042402, WO 2002/086083, and WO 2001/039722. A PD-L2 inhibitory antibody or a PD-L2 inhibitory peptide is reported in WO 03/042402 and WO 02/00730.
  • WO2007005874 describes isolated human monoclonal antibodies that specifically bind to PD-L1 with high affinity. The disclosure provides methods for treating various diseases including cancer using anti-PD-L1 antibodies.
  • US2009/0305950 describes multimers, particularly tetramers of an extracellular domain of PD-1 or PD-L1. The application describes therapeutic peptides.
  • Further, the specification mentions that peptides can be used therapeutically to treat disease, e.g., by altering co-stimulation in a patient. An isolated B7-4 or PD-1 protein, or a portion or fragment thereof (or a nucleic acid molecule encoding such a polypeptide), can be used as an immunogen to generate antibodies that bind B7-4 or PD-1 using standard techniques for polyclonal and monoclonal antibody preparation. A full-length B7-4 or PD-1 protein can be used, or alternatively, the invention provides antigenic peptide fragments of B7-4 or PD-1 for use as immunogens. The antigenic peptide of B7-4 or PD-1 comprises at least 8 amino acid residues and encompasses an epitope of B7-4 or PD-1 such that an antibody raised against the peptide forms a specific immune complex with B7-4 or PD-1. Preferably, the antigenic peptide comprises at least 10 amino acid residues, more preferably at least 15 amino acid residues, even more preferably at least amino acid residues, and most preferably at least 30 amino acid residues.
  • Freeman et al in U.S. Pat. No. 7,432,059 appears to disclose and claim methods of identifying compounds that up modulate T cell activation in the presence of a PD-1-mediated signal. Diagnostic and treatment methods utilizing compositions of the invention are also provided in the patent.
  • Further, Freeman et al in U.S. Pat. No. 7,709,214 appears to cover methods for up regulating an immune response with agents that inhibit the interactions between PD-L2 and PD-1.
  • Despite existence of many disclosures as discussed above, however, a significant unmet medical need still exists due to the lack of effective peptides or modified peptides as therapeutic agents as alternatives in the therapeutic area. It is known that synthetic peptides offer certain advantages over antibodies such as ease of production with newer technologies, better purity and lack of contamination by cellular materials, low immunogenicity, improved potency and specificity. Peptides may be more stable and offer better storage properties than antibodies. Moreover, often peptides possess better tissue penetration in comparison with antibodies, which could result in better efficacy. Peptides can also offer definite advantages over small molecule therapeutics counterparts such as lesser degree of toxicity and lower probability of drug-drug interaction.
  • The present invention therefore may provide the solution for this unmet medical need by offering novel synthetic peptide and its derivatives which are based on the PD1 ectodomain.

09338-scitech1-NovartisAcxd
Aurigene team: (from left) Brahma Reddy V, Thomas Antony, Murali Ramachandra, Venkateshwar Rao G, Wesley Roy Balasubramanian, Kishore Narayanan, Samiulla DS, Aravind AB, and Shekar Chelur

 

 

Patent

http://www.google.com/patents/US20110318373

8. SNTSESFK(SNTSESF)FRVTQLAPKAQIKE-NH2 (SEQ ID NO: 49)

 

Example 2 Synthesis of

Synthesis of Linear Fragment—Fmoc-FRVTQLAPKAQIKE

  • Desiccated CLEAR-Amide resin ((100-200 mesh) 0.4 mmol/g, 0.5 g) was distributed in 2 polyethylene vessels equipped with a polypropylene filter. The linear peptide synthesis on solid phase were carried out automatically, using Symphony parallel synthesizer (PTI) using the synthesis programs mentioned in the table below. Swelling, C-terminal amino acid [Fmoc-Glu(OtBu)-OH] attachment and capping of the peptidyl resin was carried out as per the protocol in Table I. Subsequent amino acid coupling was carried out as mentioned in Table II. The amino acids used in the synthesis were Fmoc Phe-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Val-OH, Fmoc-Thr(OtBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Pro-OH, Fmoc-Ile-OH. After the completion of Fmoc-Phe-OH coupling the resin was taken out form peptide synthesiser and manual coupling was carried out as follows
  • Fmoc-Phe-OH peptidyl resin from automated synthesiser was pooled in to a glass vessel with frit. The Fmoc group of the peptidyl resin was deprotected by treating it twice with 20% (v/v) piperidine/DMF solution for 5 and 15 min (10 m L). The resin was washed with DMF (6×15 m L), DCM (6×15 m L) and DMF (6×15 m L). Kaiser test on peptide resin aliquot upon completion of Fmoc-deprotection was positive. Fmoc-Lys (Fmoc)-OH (0.48 g; 4 equiv. 0.8 m mol) in dry DMF was added to the deprotected resin and coupling was initiated with DIC (0.15 m L; 5 equiv, 1 m mol) and HOBT (0.08 g; 5 equiv, 0.6 m mol) in DMF. The concentration of each reactant in the reaction mixture was approximately 0.4 M. The mixture was rotated on a rotor at room temperature for 3 h. Resin was filtered and washed with DMF (6×15 mL), DCM (6×15 mL) and DMF (6×15 mL). Kaiser test on peptide resin aliquot upon completion of coupling was negative. The Fmoc group on the peptidyl resin is deprotected by treating it twice with 20% (v/v) piperidine/DMF solution for 5 and 15 min (15 mL). The resin was washed with DMF (6×15 mL), DCM (6×15 mL) and DMF (6×15 mL). Kaiser test on peptide resin aliquot upon completion of Fmoc-deprotection was positive. After the deprotection of Fmoc group on Fmoc-Lys(Fmoc)-attached peptidyl resin the peptide chain growth was carried out from both the free amino terminus suing 8 equivalent excess of amino acid (1.6 m mol, 8 equivalent excess of HOBt (0.22 g, 1.6 m mol) and 10 equivalent excess of DIC (0.32 m L, 2 m mol) relative to resin loading. The coupling was carried out at room temperature for 3 h. The amino acids coupled to the peptidyl resin were; Fmoc-Phe-OH (0.62 g; 8 equiv, 1.6 m mol), Fmoc-Ser (OtBu)-OH (0.62 g; 8 equiv, 1.6 m mol), Fmoc-Glu (OtBu)-OH (0.68 g; 8 equiv, 1.6 m mol), Fmoc-Ser (OtBu)-OH (0.62 g; 8 equiv, 1.6 m mol), Fmoc-Thr (OtBu)-OH (0.64 g; 8 equiv, 1.6 m mol), Fmoc-Asn (Trt)-OH (0.95 g; 8 equiv, 1.6 m mol) and N-terminus amino acids as Boc-Ser (OtBu)-OH (0.41 g; 8 equiv, 1.6 m mol) The peptidyl resin was cleaved as mentioned in procedure for cleavage using cleavage cocktail A to yield (565 mg), 70% yield. The crude material was purified by preparative HPLC on Zorbax Eclipse XDB-C18 column (9.4 mm×250 mm, 5 μm) with buffer A: 0.1% TFA/Water, buffer B: Acetonitrile. The peptide was eluted by gradient elution 0-5 min=5-10% buffer B, 10-20 min=29% buffer B with a flow rate of 7 mL/min. HPLC: (method 1): RT-12 min (96%); LCMS Calculated Mass: 3261.62, Observed Mass: 1631.6 [M/2+H]+; 1088 [M/3+H]+); 816.2[M/4+H]+;

STRUCTURE , READER DISCRETION IS NEEDED

aunf12

N2,N6-Bis(L-seryl-L-asparaginyl-L-threonyl-L-seryl-L-alpha-glutamyl-L-seryl-L-phenylalanyl)-L-lysyl-L-phenylalanyl-L-arginyl-L-valyl-L-threonyl-L-glutaminyl-L-leucyl-L-alanyl-L-prolyl-L-lysyl-L-alanyl-L-glutaminyl-L-isoleucyl-L-lysyl-L-alpha-glutamine

C142 H226 N40 O48, 3261.553

 CAS 1353563-85-5,
L-​α-​Glutamine, N2,​N6– ​bis(L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L- ​seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

aunf12

aunf12

SEE ALSO

CAS 1353564-61-0,
L-​α-​Glutamine, N2,​N6– ​bis(D-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L- ​seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-
 CAS 1353563-91-3
D-​α-​Glutamine, N2,​N6– ​bis(D-​seryl-​D-​asparaginyl-​D-​threonyl-​D-​seryl-​D-​α-​glutamyl-​D- ​seryl-​D-​phenylalanyl)​-​D-​lysyl-​D-​phenylalanyl-​D-​arginyl-​D-​ valyl-​D-​threonyl-​D-​glutaminyl-​D-​leucyl-​D-​alanyl-​D-​prolyl-​D-​ lysyl-​D-​alanyl-​D-​glutaminyl-​D-​isoleucyl-​D-​lysyl-

US 2015087581

Compound 8 (SEQ ID NO: 49) SNTSESFK(SNTSESF)FRVTQLAPKAQIKE-NH2Image loading...

Example 2Synthesis of Sequence Shown in SEQ ID NO: 49

Image loading...

Synthesis of Linear Fragment—Fmoc-FRVTQLAPKAQIKE

Desiccated CLEAR-Amide resin ((100-200 mesh) 0.4 mmol/g, 0.5 g) was distributed in 2 polyethylene vessels equipped with a polypropylene filter. The linear peptide synthesis on solid phase were carried out automatically, using Symphony parallel synthesizer (PTI) using the synthesis programs mentioned in the table below. Swelling, C-terminal amino acid [Fmoc-Glu(OtBu)-OH] attachment and capping of the peptidyl resin was carried out as per the protocol in Table I. Subsequent amino acid coupling was carried out as mentioned in Table II. The amino acids used in the synthesis were Fmoc Phe-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Val-OH, Fmoc-Thr(OtBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Pro-OH, Fmoc-Ile-OH. After the completion of Fmoc-Phe-OH coupling the resin was taken out form peptide synthesiser and manual coupling was carried out as follows.

Fmoc-Phe-OH peptidyl resin from automated synthesiser was pooled in to a glass vessel with frit. The Fmoc group of the peptidyl resin was deprotected by treating it twice with 20% (v/v) piperidine/DMF solution for 5 and 15 min (10 mL). The resin was washed with DMF (6×15 mL), DCM (6×15 mL) and DMF (6×15 mL). Kaiser test on peptide resin aliquot upon completion of Fmoc-deprotection was positive.

Fmoc-Lys (Fmoc)-OH (0.48 g; 4 equiv. 0.8 mmol) in dry DMF was added to the deprotected resin and coupling was initiated with DIC (0.15 mL; 5 equiv, 1 mmol) and HOBT (0.08 g; 5 equiv, 0.6 mmol) in DMF. The concentration of each reactant in the reaction mixture was approximately 0.4 M. The mixture was rotated on a rotor at room temperature for 3 h. Resin was filtered and washed with DMF (6×15 mL), DCM (6×15 mL) and DMF (6×15 mL). Kaiser test on peptide resin aliquot upon completion of coupling was negative. The Fmoc group on the peptidyl resin is deprotected by treating it twice with 20% (v/v) piperidine/DMF solution for 5 and 15 min (15 mL). The resin was washed with DMF (6×15 mL), DCM (6×15 mL) and DMF (6×15 mL). Kaiser test on peptide resin aliquot upon completion of Fmoc-deprotection was positive.

After the deprotection of Fmoc group on Fmoc-Lys(Fmoc)-attached peptidyl resin the peptide chain growth was carried out from both the free amino terminus suing 8 equivalent excess of amino acid (1.6 mmol, 8 equivalent excess of HOBt (0.22 g, 1.6 mmol) and 10 equivalent excess of DIC (0.32 mL, 2 mmol) relative to resin loading. The coupling was carried out at room temperature for 3 h. The amino acids coupled to the peptidyl resin were; Fmoc-Phe-OH (0.62 g; 8 equiv, 1.6 mmol), Fmoc-Ser (OtBu)-OH (0.62 g; 8 equiv, 1.6 mmol), Fmoc-Glu (OtBu)-OH (0.68 g; 8 equiv, 1.6 mmol), Fmoc-Ser (OtBu)-OH (0.62 g; 8 equiv, 1.6 mmol), Fmoc-Thr (OtBu)-OH (0.64 g; 8 equiv, 1.6 mmol), Fmoc-Asn (Trt)-OH (0.95 g; 8 equiv, 1.6 m mol) and N-terminus amino acids as Boc-Ser (OtBu)-OH (0.41 g; 8 equiv, 1.6 mmol) The peptidyl resin was cleaved as mentioned in procedure for cleavage using cleavage cocktail A to yield (565 mg), 70% yield. The crude material was purified by preparative HPLC on Zorbax Eclipse XDB-C18 column (9.4 mm×250 mm, 5 μm) with buffer A: 0.1% TFA/Water, buffer B:Acetonitrile. The peptide was eluted by gradient elution 0-5 min=5-10% buffer B, 10-20 min=29% buffer B with a flow rate of 7 mL/min. HPLC: (method 1): RT—12 min (96%); LCMS Calculated Mass: 3261.62, Observed Mass: 1631.6 [M/2+H]+; 1088 [M/3+H]+;); 816.2[M/4+H]+.

SMILES

O=C(N[C@@H](CCCCNC(=O)[C@H](Cc1ccccc1)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(=O)O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CO)N)[C@@H](C)O)C(=O)N[C@@H](Cc2ccccc2)C(=O)N[C@@H](CCCNC(=N)N)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N3CCC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(=O)O)C(N)=O)[C@H](Cc4ccccc4)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(=O)O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CO)N)[C@@H](C)O

NEXT………..

CAS 1353564-65-4
C142 H226 N40 O48
L-​α-​Glutamine, L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​seryl-​L-​phenylalanyl-​N6– ​(L-​seryl-​D-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​ seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-
Molecular Weight, 3261.55

aunf12

NEXT……….

CAS 1353564-31-4, C142 H226 N40 O48
L-​α-​Glutamine, L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​seryl-​L-​phenylalanyl-​N6– ​(D-​seryl-​D-​asparaginyl-​D-​threonyl-​D-​seryl-​D-​α-​glutamyl-​D-​ seryl-​D-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-
USE ALL YOUR DISCRETION……………

Clips

Aurigene and Pierre Fabre Pharmaceuticals Announce a Licensing Agreement for a New Cancer Therapeutic in Immuno-oncology: AUNP12, an Immune Checkpoint Modulator Targeting the PD-1 Pathway

Pierre Fabre are thus reinforcing their oncology portfolio which already enjoys a combination of chemotherapies, monoclonal antibodies and immuno-conjugates assets at various development phases

Feb 13, 2014, 03:14 ET from Aurigene and Pierre Fabre Pharmaceuticals

CASTRES, France and BANGALORE, India, February 13, 2014 /PRNewswire/ —

Pierre Fabre, the third largest French pharmaceutical company, and Aurigene, a leading biotech company based in India, today announced that the two companies have entered into a collaborative license, development and commercialization agreement granting Pierre Fabre global Worldwide rights (excluding India) to a new immune checkpoint modulator, AUNP-12.

AUNP-12 offers a breakthrough mechanism of action in the PD-1 pathway compared to other molecules currently in development in the highly promising immune therapy cancer space. AUNP-12 is the only peptide therapeutic in this pathway and could offer more effective and safer combination opportunities with emerging and established treatment regimens.  AUNP-12 will be in development for numerous cancer indications.

Under the terms of this agreement, Aurigene will receive an upfront payment from Pierre Fabre. Aurigene will also receive additional milestone payments based upon the continued development, regulatory progresses and commercialization of AUNP-12.

“We are pleased that Pierre Fabre see the PD-1 program as a strategic asset in their portfolio. Overall, the deal structure, in line with the financial terms that have been seen in this space, demonstrate the importance that Pierre Fabre attach to the program,” said CSN Murthy, CEO, Aurigene.

“The plans that Pierre Fabre have detailed for the development of this differentiated asset highlight the long-term opportunities for this novel cancer therapeutic,” added Murali Ramachandra, Sr VP, Research, Aurigene.

“This agreement, in the field of oncology, is fully consistent with our vision to build Pierre Fabre’s future in prescription drugs, from a combination of cutting-edge internal R&D capabilities and license partnerships with innovative biotech companies like Aurigene,” stated Bertrand Parmentier, CEO, Pierre Fabre.

“With this deal, Pierre-Fabre Pharmaceuticals are reinforcing their portfolio of oncology assets and capitalizing on their proven capabilities in developing biological compounds such as monoclonal antibodies and immuno-conjugates. We have been impressed by the science at Aurigene and encouraged by the differentiated profile reported for AUNP-12,” added Frédéric Duchesne, President, Pierre Fabre Pharmaceuticals.

About immuno-oncology

Immuno-oncology is an emerging field in cancer therapy, where the body’s own immune system is harnessed to fight against cancer. This approach of targeting cancer through immune response has had a breakthrough when robust and sustained responses were obtained only upon blocking the immune checkpoint targets (such as PD-1 and CTLA4). Recent successes in clinical trials performed with such therapies suggest that immunotherapy should be considered alongside surgery, chemotherapy, radiotherapy and targeted therapy as the fifth cornerstone of cancer treatment.

PD-1 (Programmed cell Death 1) is a receptor that negatively regulates T-cell activation by interacting with specific ligands PD-L1 and PD-L2. Tumor cells express these ligands and thereby escape from the action of T-cells.

About AUNP-12

AUNP-12  is a branched 29-amino acid peptide sequence engineered from the PD-L1/ L2 binding domain of PD-1 It blocks the PD-1/PD-L1, PD-1/PD-L2 and PD-L1/CD80 pathways. AUNP-12 is highly effective in antagonizing PD-1 signaling, with desirable in vivo exposure upon subcutaneous dosing. It inhibits tumor growth and metastasis in preclinical models of cancer and is well tolerated with no overt toxicity at any of the tested doses.

About Aurigene

Aurigene is a biotech focused on development of innovative small molecule and peptide therapeutics for Oncology and Inflammation; key focus areas for Aurigene are Immuno-oncology, Epigenetics and the Th17 pathway. Aurigene’s PD-1 program is the first of several peptide-based immune checkpoint programs that are at different stages of Discovery.

Aurigene has partnered with several big pharma and mid-pharma companies in the US and Europe, and has delivered multiple clinical compounds through these partnerships. With over 500 scientists, Aurigene has collaborated with 6 of the top 10 pharma companies.

Aurigene’s pre-clinical pipeline includes (1) Selective and pan-BET Bromodomain inhibitors (2) RoR gamma reverse agonists (3) EZH2 inhibitors (4) NAMPT inhibitors and (5) Several immune check point peptide inhibitor programs.

For more information:  http://aurigene.com/

About Pierre Fabre:

Pierre Fabre is a privately-owned health care company created in 1961 by Mr Pierre Fabre. It is the second largest French independent pharmaceutical group with 2013 sales amounting to about €2 billion (yet to be audited) across 140 countries. The company is structured around two divisions: Pharmaceuticals (Prescription drugs, OTC, Oral care) and Dermo-cosmetics. Prescription drugs are organized around four main franchises: oncology, dermatology, women’s health and neuropsychiatry. Pierre Fabre employs some 10 000 people worldwide, including 1 300 in R&D. The company allocates about 20% of its pharmaceuticals sales to R&D and relies on more than 25 years of experience in the discovery, development and global commercialization of innovative drugs in oncology. Pierre Fabre has a long commitment to oncology and immunology with major R&D centers in France: the Pierre Fabre immunology Centre (CIPF) in Saint Julien en Genevois and the Pierre Fabre Research Institute (IRPF) located on the Toulouse-Oncopole campus  which has been officially recognized as a National Center of Excellence for cancer research since 2012.

 

REFERENCES

http://www.differding.com/data/AUNP_12_A_novel_peptide_therapeutic_targeting_PD_1_immune_checkpoint_pathway_for_cancer_immunotherapy.pdf

http://slideplayer.com/slide/5760496/

P. Sasikumar, R. Shrimali, S. Adurthi, R. Ramachandra, L. Satyam, A. Dhudashiya, D. Samiulla, K. B. Sunilkumar and M. Ramachandra, “A novel peptide therapeutic targeting PD1 immune checkpoint with equipotent antagonism of both ligands and a potential for better management of immune-related adverse events,” Journal for ImmunoTherapy of Cancer, vol. 1, no. Suppl 1,  O24, 2013.

P. G. N. Sasikumar, M. Ramachandra, S. K. Vadlamani, K. R. Vemula, L. K. Satyam, K. Subbarao, K. R. Shrimali and S. Kandepudu (Aurigene Discovery Technologies Ltd, Bangalore, India), “Immunosuppression modulating compounds”, US Patent application US 2011/0318373, 29 Dec 2011.

P. G. Sasikumar, L. K. Satyam, R. K. Shrimali, K. Subbarao, R. Ramachandra, S. Vadlamani, A. Reddy, A. Kumar, A. Srinivas, S. Reddy, S. Gopinath, D. S. Samiulla and M. Ramachandra, “Demonstration of anti-tumor efficacy in multiple preclinical cancer models using a novel peptide inhibitor (Aurigene-012) of the PD1 signaling pathway,” Cancer Research, vol. 72, no. 8 Suppl. 1, Abstract 2850, 2012.

P. G. N. Sasikumar, M. Ramachandra, S. K. Vadlamani, K. R. Shrimali and K. Subbarao, “Therapeutic compounds for immunomodulation” (Aurigene Discovery Technologies Ltd, Bangalore, India), PCT Patent Application WO 2012/168944, 13 Dec 2012.

P. G. N. Sasikumar and M. Ramachandra, “Immunomodulating cyclic compounds from the BC loop of human PD1” (Aurigene Discovery Technologies Ltd, Bangalore, India), PCT Patent Application WO/2013/144704, 3 Oct 2013.

P. G. N. Sasikumar, M. Ramachandra and S. S. S. Naremaddepalli, “Peptidomimetic compounds as immunomodulators” (Aurigene Discovery Technologies Ltd, Bangalore, India), US Patent Application US 2013/0237580, 12 Sep 2013.

A. H. Sharpe, M. J. Butte and S. Oyama (Harvard College), “Modulators of immunoinhibitory receptor PD-1, and methods of use thereof”, PCT Patent Application WO/2011/082400, 7 Jul 2011.

M. Cordingley, “Battle of PD-1 blockade is on”, February 7, 2014 : http://discoveryview.ca/battle-of-pd-1-blockade-is-on/ [Accessed 25 February 2014].

Mr. CSN Murthy

Chief Executive Officer, Aurigene Discovery Technologies Ltd.

Mr. CSN Murthy began his career with ICICI Ventures, India’s first Venture Capital fund. He was subsequently a management consultant to the Pharma and Chemical sectors. Later, he worked in the Business Development and General Management functions in Pharmaceutical companies, including as the Chief Operating Officer of Gland Pharma Ltd. CSN holds a Bachelors degree in Chemical Engineering from the Indian Institute of Technology (IIT), Madras and an MBA from the Indian Institute of Management (IIM), Bangalore.


Dr.Thomas Antony

Associate Research Director, Aurigene Discovery Technologies Ltd.

Dr.Thomas Antony did his Ph.D in Biophysical Chemistry from University of Delhi and had his postdoctoral training at Jawaharlal Nehru University- Delhi, The University of Medicine and Dentistry of New Jersey- USA, and Max Planck Institute for Biophysical Chemistry- Germany. He is the recipient of many research fellowships, including Max Planck Fellowship and Humboldt Research Fellowship.  He has more than 20 years of research experience. Dr.Thomas has published 24 research papers and he is the co-author of three international patents. His core area of expertise is in assay development and screening. At Aurigene, Dr.Thomas leads the Biochemistry and Structural Biology Divisions.  He was the coordinator of Aurigene-University of Malaya collaboration programs.


Dr. Kavitha Nellore

Associate Research Director, Aurigene Discovery Technologies Ltd.

Dr. Kavitha Nellore obtained her PhD in Bioengineering from Pennsylvania State University, USA.  During this time, she was a fellow of the Huck’s Institute of Life Sciences specializing in Biomolecular Transport Dynamics. She has been at Aurigene for more than a decade, and is currently leading a group of cell biologists at both Bangalore and Kuala Lumpur. At Aurigene, she leads multiple drug discovery programs in the therapeutic areas of inflammation, oncology and immuno-oncology. She plays a key role in target selection as well as validation efforts to add to Aurigene’s pipeline. Kavitha also played a key role in coordinating the Aurigene-University of Malaya collaboration.

 

/////////AUNP-12,  Aurigene,  Pierre Fabre Pharmaceuticals, Licensing Agreement,  New Cancer Therapeutic,  Immuno-oncology, AUNP 12, Immune Checkpoint Modulator Targeting the PD-1 Pathway, PEPTIDES

FEW MORE ACADEMIC COMPDS FROM PATENT, REDER DISCRETION NEEDED

C142 H225 N39 O49

L-​Glutamic acid, N2,​N6- ​bis(L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L- ​seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

3262.54, Sequence Length: 29, 22, 7

multichain; modified (modifications unspecified)

SNTSESFK FRVTQ LAPKAQIKE,  1353564-66-5

SNTSESF

C142 H225 N39 O49

L-​Glutamic acid, N2,​N6– ​bis(L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L- ​seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

3262.54

NEXT……………………

SNTSESFK FRVTQ LAPKAQI KE

SNTSESF

CAS  1353564-64-3

C142 H226 N40 O48

L-​α-​Glutamine, L-​seryl-​D-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​seryl-​L-​phenylalanyl-​N6- ​(L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​ seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

MW 3261.55, Sequence Length: 29, 22, 7

multichain; modified

smiles

O=C(N[C@@H](CCCCNC(=O)[C@H](Cc1ccccc1)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(=O)O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](N)CO)[C@@H](C)O)C(=O)N[C@@H](Cc2ccccc2)C(=O)N[C@@H](CCCNC(=N)N)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N3CCC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(=O)O)C(N)=O)[C@H](Cc4ccccc4)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(=O)O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@@H](CC(N)=O)NC(=O)[C@@H](N)CO)[C@@H](C)O
NEXT……………..

CAS  1353564-60-9

C142 H226 N40 O48

L-​α-​Glutamine, D-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​seryl-​L-​phenylalanyl-​N6- ​(L-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L-​ seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

3261.55

Sequence Length: 29, 22, 7multichain; modified

SNTSESFKFR VTQLAPKAQI KE

NEXT…………………….

. CAS  1353564-61-0

C142 H226 N40 O48

L-​α-​Glutamine, N2,​N6- ​bis(D-​seryl-​L-​asparaginyl-​L-​threonyl-​L-​seryl-​L-​α-​glutamyl-​L- ​seryl-​L-​phenylalanyl)​-​L-​lysyl-​L-​phenylalanyl-​L-​arginyl-​L-​ valyl-​L-​threonyl-​L-​glutaminyl-​L-​leucyl-​L-​alanyl-​L-​prolyl-​L-​ lysyl-​L-​alanyl-​L-​glutaminyl-​L-​isoleucyl-​L-​lysyl-

3261.55

Sequence Length: 29, 22, 7multichain; modified

SNTSESFK FRVTQ LAPKAQI KE
SNTSESF

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

Share

DS-1040, Activated thrombin activatable fibrinolysis (TAFIa) inhibitor

 phase 2, Uncategorized  Comments Off on DS-1040, Activated thrombin activatable fibrinolysis (TAFIa) inhibitor
Apr 012016
 

str1

DS-1040

Daiichi Sankyo Co Ltd

Ischemic stroke

(2S)-5-amino-2-[[1-(4-methylcyclohexyl)imidazol-4-yl]methyl]pentanoic acid

1H-​Imidazole-​4-​propanoic acid, α-​(3-​aminopropyl)​-​1-​(trans-​4-​methylcyclohexyl)​-​, (αS)​-

(2S)-5-amino-2-{[1-(trans-4-methylcyclohexyl)-1H-imidazol-4-yl]methyl}pentanoic acid

free form cas 1335138-62-9

1:1 TOSYLATE 1335138-89-0

1335138-90-3  1:1:1 TOSYLATE HYDRATE

phase 2, Ischemic stroke

Molecular Formula: C16H27N3O2
Molecular Weight: 293.40448 g/mol

TAFIa inhibitors, useful for treating myocardial infarction, angina, pulmonary hypertension and deep vein thrombosis.

In March 2016, DS-1040 was reported to be in phase 2 C clinical development, and the study was expected to complete in June 2017.

https://clinicaltrials.gov/ct2/show/NCT02560688

  • 01 Feb 2016Daiichi Sankyo initiates a phase I trial in Healthy volunteers in United Kingdom (NCT02647307)
  • 09 Jan 2016Daiichi Sankyo plans a phase I trial in Healthy volunteers in United Kingdom (NCT02647307)
  • 29 Sep 2015Daiichi Sankyo plans a drug-interaction phase I trial in Healthy volunteers in United Kingdom (IV) (NCT02560688)

SCHEMBL14631441.png

SYNTHESIS

DS 1010 1

 

COMPLETE SYNTHESIS

 

DS 1010

 

 

WO201111506

WO2013039202

WO 2016043254

PATENT

DS 1010 1

 

COMPLETE SYN……….

 

DS 1010

WO2016043253

The optical purity of the obtained compound was measured by the following HPLC analysis conditions.
(2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans -4- methylcyclohexyl)-lH-imidazol-4-yl] methyl} valeric acid (S)-2-amino 1-propanol salt (A1 step, A2 step, A3 step), (2S) -5 – [ (tert- butoxycarbonyl) amino] -2 – {[1- (trans -4- methylcyclohexyl)-lH-imidazole 4-yl] methyl} optical purity measurement conditions valerate (A4 step):
column: CHIRAL AGP 4.6mmI. D. × 250mm (5μm),
mobile phase: methanol / 10mM phosphate buffer solution (pH7.0) = 95/5,
temperature: 40 ℃,
flow rate: 0.5mL / min,
detection method: UV at 220nm,
retention time: R body: 5.9 minutes, S body: 7.3 minutes.

(2S)-5-amino-2 – Optical purity measurement conditions {[1- (trans-4- methylcyclohexyl)-lH-imidazol-4-yl] methyl} valerate p- toluenesulfonate (A5 Step) :
column: CHIRLCEL OZ-H 4.6mmI. D. × 250mm (5μm),
mobile phase: hexane / ethanol / methanol / isopropanol / trifluoroacetic acid / triethylamine = 860/100/20/2/2
temperature: 30 ℃
flow rate: 1.0mL / min
detection method: UV at 220nm
retention time: R body: 16.1 minutes, S body: 13.0 minutes  (example  1) (1-1) 5 – [(Tert- butoxycarbonyl) amino] -2-methoxy-carbonyl) valeric acid morpholine salt

 

[Of 11]

 

 In methanol (400mL) solution of di -tert- butyl (100.0g) and 3-chloro-propylamine hydrochloride (71.5g), was added dropwise triethylamine (51.0g) at 0 ℃, at the same temperature It was stirred for 16 hours. To the reaction solution was added toluene (400 mL) and water (400 mL), then were separated, and the organic layer was washed with water. Toluene 400mL was added to the organic layer, was concentrated under reduced pressure to 300 mL, N, N-dimethylacetamide (210 mL) was added and concentrated in vacuo to 300 mL. Potassium carbonate solution (126.66g), tetrabutylammonium bromide (44.32g), was added dimethyl malonate (90.82g) and N, N-dimethylacetamide (100 mL), stirred for 20 hours at 55 ° C. did. Toluene (400 mL) and water (700 mL) was added to the reaction mixture, after separation, The organic layer was washed with water, with 1M aqueous sodium hydroxide and water, and concentrated under reduced pressure to 150 mL. This solution methanol (1870mL) and 1M sodium hydroxide solution (430.8mL) in addition to, and the mixture was stirred for 27.5 hours at 0 ℃. Concentrated hydrochloric acid to the reaction solution (2.5 mL) was added, the pH was adjusted to 7-9, and concentrated in vacuo to 375 mL. After addition of ethyl acetate (500mL) to the reaction solution, concentrated hydrochloric acid (35.1mL) was added, the pH was adjusted to 2.2-2.5, and the layers were separated. The aqueous layer was extracted with ethyl acetate (500 mL), after mixing the organic layer under reduced pressure, and prepared by dehydration condensation of ethyl acetate (250 mL) solution. The resulting solution of ethyl acetate (500 mL) and morpholine (37.5 g) was added to and stirred overnight. The precipitated crystals were filtered, washed with ethyl acetate, and dried under reduced pressure, to give the title compound (136.1g, 81.9% yield).

1 H-NMR (DMSO-d- . 6 ) [delta]: 6.79 (1H, t, J = 5.5 Hz), 3.61 (4H, t, J = 4.9 Hz), 3.58 The (3H, s) , 3.14 (1H, t, J = 7.8Hz), 2.90-2.80 (6H, m), 1.74-1.59 (2H, m), 1.37 (9H, s) , 1.34-1.25 (2H, m).

 

(1-2) [1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methanol

 

[Of 12]

 

 N, and stirred for 4 h methanol (56 mL) solution at 5 ~ 10 ℃ of N- dimethylformamide dimethyl acetal (77.4 g) and ethyl isocyanoacetate (70.0g).The reaction solution was cooled to 0 ℃, water (5.3mL) and trans-4- methylcyclohexyl amine (105.1g) was added, and the mixture was stirred for 24 hours at 60 ~ 65 ℃. The reaction was cooled to room temperature, toluene (420 mL), supplemented with 10% brine (280 mL) and concentrated hydrochloric acid (68 mL), After separation, the organic layer was washed with 10% brine (140 mL). Organic layer to 10% sodium chloride solution (280mL) and concentrated hydrochloric acid were added for liquid separation after (78.4g), was added to separate liquid further 10% saline solution into the organic layer (210mL) and concentrated hydrochloric acid (31.3g). After dissolving sodium chloride (70.0 g) in aqueous layer, adding toluene (420 mL) and 50% aqueous sodium hydroxide (85 mL), after separation, toluene (350 mL) the organic layer was added, under reduced pressure, dehydration concentrated was prepared in toluene (420 mL) solution was. The solution was cooled to 0 ℃, dropped the hydrogenated bis (2-methoxyethoxy) aluminum sodium (70% toluene solution) (207.4g), and the mixture was stirred at room temperature for 1 hour. The reaction was cooled to 0 ° C., was added dropwise 12.5% ​​aqueous sodium hydroxide solution (700 mL), stirred for 1 hour at room temperature. After the solution was separated and the organic layer was washed successively with 12.5% ​​aqueous solution of sodium hydroxide (700mL) and 20% sodium chloride solution (140mL), toluene in the organic layer (140mL), 1- butanol (14mL), water ( 280mL) and was added to aliquots of concentrated hydrochloric acid (48mL). It was further added to liquid separation with water (140 mL) and concentrated hydrochloric acid (2 mL) to the organic layer. Met The aqueous layer was stirred in for 1 hour activated carbon (10.5 g), activated charcoal was filtered off, the activated carbon was washed with water (210 mL). Matches the filtrate and washings, sodium chloride (140 g), toluene was added (980 mL) and 50% aqueous sodium hydroxide (42 mL), After separation, under reduced pressure and the organic layer was dried concentrated toluene (210 mL) It was prepared in solution. The solution was stirred 30 minutes at 50-55 ° C., cooled to room temperature, it was added dropwise heptane (560 mL), and stirred at the same temperature for 3 hours. The precipitated crystals were filtered to give after washing with toluene / heptane (1/4) mixture solution, the title compound was dried under reduced pressure (77.2 g, 64.2% yield).

 

 1 H-NMR (CDCl 3 ) [delta]: 7.49 (1H, s), 6.91 (1H, s), 4.58 (2H, s), 3.83 (1H, tt, J = 12.0 , 3.9Hz), 2.10-2.07 (2H, m), 1.87-1.84 (2H, m), 1.70-1.61 (2H, m), 1.48-1 .42 (1H, m), 1.15-1.06 (2H, m), 0.95 (3H, d, J = 6.5Hz).

(1-3) (2E) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1-trans-4- methylcyclohexyl]-lH-imidazol-4-yl} methylidene} methyl valerate

 

[Of 13]

 

 (1-2) The compound obtained in (50.0 g) in toluene (350 mL) and acetic acid (150 mL) was dissolved in a mixed solution, 2,2,6,6-tetramethylpiperidine -N- oxyl at 30 ° C. It was added (966mg) and ortho-periodic acid (16.9g), and the mixture was stirred for 1 hour at 30-35 ℃. The reaction mixture was added 10% aqueous sodium bisulfite solution (150 mL), after stirring for 30 minutes at room temperature, toluene was added (400 mL), and concentrated in vacuo to 300 mL. The solution further by the addition of toluene (400 mL), after concentration under reduced pressure again to 300 mL, was added toluene (500 mL), water (200 mL) and 50% aqueous sodium hydroxide (118 mL). Were separated, the organic layer was washed with 20% brine (150 mL), addition of toluene (200 mL), under reduced pressure and dehydrated concentrated prepared in toluene (400 mL) solution. The compound obtained in the solution (1-1) (116.5g), N, N- dimethylformamide (175 mL) and acetic acid (4.2 mL) was added, under reduced pressure, and dried for 8 hours under reflux. The reaction was cooled to room temperature, adding toluene (400 mL), washed once with 3 times with 5% aqueous sodium bicarbonate solution (400 mL) and 10% brine (250 mL), under reduced pressure and the organic layer was dried concentrated toluene It was prepared (900 mL) solution. This solution was added activated charcoal (15 g) at 35 ~ 40 ° C., after stirring for 30 minutes at the same temperature, filtered and the activated carbon was washed with toluene. Meet the filtrate and washings, after which was concentrated under reduced pressure until 250mL, it was added dropwise heptane (500mL) at room temperature. After stirring for 1.5 hours at the same temperature, then cooled to 0 ℃, and the mixture was stirred for 1 hour. The precipitated crystals were filtered to give after washing with toluene / heptane (1/2) mixture solution, the title compound was dried under reduced pressure (85.0 g, 81.5% yield).

 

 1 H-NMR (CDCl 3 ) [delta]: 7.59 (1H, s), 7.47 (1H, s), 7.15 (1H, s), 7.08 (1H, brs), 3.92- 3.87 (1H, m), 3.78 (3H, s), 3.16-3.12 (2H, m), 2.96 (2H, t, J = 7.5Hz), 2.14- 2.11 (2H, m), 1.90-1.87 (2H, m), 1.77-1.65 (5H, m), 1.47 (9H, s), 1.17-1. 10 (2H, m), 0.96 (3H, d, J = 6.5Hz).

 

 (1-4) (2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl)-lH-imidazol-4-yl] methyl} valerate (S ) -2-amino-1-propanol salt (A1 process, A2 process, A3 process)

 

[Of 14]

 

 The compound obtained in (1-3) (40.0g), (R) -2,2′- bis (di-3,5-xylyl) -1,1′-binaphthyl (507.4Mg) and dichloro (p- cymene) ruthenium (II) (dimer) and (211.4mg), were dissolved in degassed 2,2,2 trifluoroethanol (400 mL), hydrogen under pressure (400-450kPa) , and the mixture was stirred for 24 hours at 60 ℃. The reaction was cooled to room temperature, after nitrogen substitution, and then concentrated under reduced pressure to 60 mL.Tetrahydrofuran (200 mL) was added, was concentrated under reduced pressure to 120 mL, of tetrahydrofuran was added (200 mL).

 

 To the resulting solution was added water (160mL), cooled to 0 ℃, was added a 50% aqueous solution of sodium hydroxide (24.0mL). After stirring the reaction mixture at room temperature for 26 hours, and the addition of 50% sodium hydroxide solution (8.00mL), and the mixture was stirred for a further 4 hours. The reaction mixture under ice-cooling was added dropwise concentrated hydrochloric acid (28 mL), activated carbon was added (2.0 g) was stirred at room temperature for 10 minutes. The active carbon was filtered off, washed with tetrahydrofuran / water (2/1) mixed solvent (180 mL), sodium chloride (40 g) was separated by adding and re-extract the aqueous layer with tetrahydrofuran (400 mL). The organic layer was matched, and concentrated in vacuo to 200 mL. After addition of toluene (400 mL) to this solution, under reduced pressure and dehydrated concentrated prepared in toluene (200 mL) solution.

 

 After adding tetrahydrofuran (400 mL) to the resulting solution was added (S) -2- amino-1-propanol (8.2 g) at room temperature and stirred for 3 hours. The solution was cooled to 0 ℃, and was filtered after stirring for 1.5 hours, it was precipitated crystals. The crystals were washed with tetrahydrofuran and dried under reduced pressure to give the title compound (45.4g, 98.2% yield, optical purity: ee 97.5%) was obtained.

 

 1 H-NMR (CD 3 OD) [delta]: 7.57 (1H, s), 6.94 (1H, s), 3.98-3.85 (1H, yd), 3.69-3.64 ( 1H, m), 3.47-3.42 (1H , m), 3.29-3.23 (1H, m), 3.01 (2H, t, J = 6.5Hz), 2.84 ( 1H, dd, J = 14.6,8.4Hz) , 2.55 (1H, dd, J = 14.6,6.2Hz), 2.52-2.45 (1H, m), 2.03 (2H, d, J = 12.7Hz ), 1.83 (2H, d, J = 13.3Hz), 1.71 (2H, q, J = 12.5Hz), 1.60-1.44 ( 5H, m), 1.41 (9H , s), 1.23-1.20 (3H, m), 1.18-1.09 (2H, m), 0.94 (3H, d, J = 6.8Hz).

 

 (1-5) (2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl)-lH-imidazol-4-yl] methyl} valerate (A4 process)

 

[Of 15]

 

 (1-4) The compound obtained in (40.0 g) in tetrahydrofuran (400 mL) and dissolved in a mixed solvent of water (160 mL), concentrated hydrochloric acid (7.3 mL) and added separation of sodium chloride (40 g) and washed 3 times with the organic layer 20% (w / w) brine (160 mL). The organic layer under reduced pressure, dehydrated concentrated prepared in toluene (320 mL) solution was dissolved after addition of tetrahydrofuran (80 mL) was warmed precipitated 83 ° C. crystal. After stirring overnight and cooled to room temperature, and stirred for a further 3 hours at 0 ℃, and filtered the precipitated crystals. After washing the crystals with toluene / tetrahydrofuran (4/1) mixed solution, and dried under reduced pressure to give the title compound (30.9g, 92.1% yield, optical purity: 97.4% ee) was obtained.

 

 1 H-NMR (CDCl 3 ) [delta]: 7.59 (1H, s), 6.73 (1H, s), 4.67 (1H, brs), 3.85-3.80 (1H, yd), 3.12-3.08 (2H, m), 2.88 (1H, dd, J = 15.2,8.8Hz), 2.79 (1H, dd, J = 15.2,3.6Hz) , 2.70-2.64 (1H, m), 2.13-2.06 (2H, m), 1.90-1.82 (2H, m), 1.79-1.52 (5H, m), 1.49-1.44 (2H, m ), 1.43 (9H, s), 1.15-1.05 (2H, m), 0.95 (3H, d, J = 6. 5Hz).

 

 (1-6) (2S) -5- amino -2 – {[1- (trans-4- methylcyclohexyl)-lH-imidazol-4-yl] methyl} valerate p- toluenesulfonate (A5 Step)
[Of 16]

 

 In tetrahydrofuran (100 mL), was dissolved the compound obtained in (1-5) (25.0 g) and p- toluenesulfonic acid monohydrate (13.3 g), activated charcoal (1 to this solution. 25 g) was added and stirred for 1 hour at 20 ~ 30 ℃. The charcoal was filtered and washed with tetrahydrofuran (50 mL).It matches the filtrate and washings, p- toluenesulfonic acid monohydrate (13.3 g) and water (7.5 mL) and the mixture was heated under reflux for 6 hours. The reaction was cooled to room temperature, it was added triethylamine (7.7 g), at room temperature and stirred overnight. To the reaction solution was added dropwise tetrahydrofuran (350 mL), after stirring for 3 hours at room temperature and filtered the precipitated crystal. After washing with tetrahydrofuran / water (50/1) mixed solution, and dried under reduced pressure to give the title compound (27.7g, 93.5% yield, optical purity: 98.4% ee) was obtained.

 

 1 H-NMR (CD 3 OD) [delta]: 8.18 (1H, s), 7.70 (2H, d-, J = 8.1 Hz), 7.22 (2H, d-, J = 7.5 Hz), 7.16 (1H, s), 4.06 (1H, tt, J = 12.0,3.9Hz), 2.94-2.86 (3H, m), 2.69 (1H, dd, J = 14.6,5.8Hz), 2.62-2.59 (1H, m), 2.36 (3H, s), 2.08-2.05 (2H, m), 1.86-1 .83 (2H, m), 1.76-1.46 (7H, m), 1.18-1.11 (2H, m), 0.94 (3H, d, J = 6.5Hz).

 

 (Example
2) (2-1) (2S) -5 – [(tert-butoxycarbonyl) amino] -2 – {[1- (trans -4- methylcyclohexyl)-lH-imidazol-4-yl] methyl } methyl valerate
[Of 17]

 

 It was asymmetrically reduced using a number of catalysts. The reaction conversion and the optical purity of the obtained title compound was determined by the following HPLC analysis conditions.

 

 Reaction conversion rate measurement:
Column: Waters XBridge C18 4.6mmI. D. × 150mm (3.5μm),
mobile phase: (A) 10mM aqueous ammonium acetate solution, (B)
acetonitrile, Gradient conditions: B: conc. ; 20% (0-5 minutes), 20-90% (5-20 minutes), 90% (20-24 minutes),
temperature: 40 ℃,
flow rate: 1.0mL / min,
detection method: UV at 215nm
retention time: raw material: 21.1 minutes, the product: 19.1 minutes,
(peak area of peak area + product of raw materials) peak area / of the reaction conversion rate = product.

 

 Optical purity measurement conditions:
column: CHIRALPAK IA 4.6mmI. D. × 250mm (5μm),
mobile phase: ethanol / hexane = 20/80
Temperature: 35 ℃,
flow rate: 1.0mL / min,
detection method: UV at 210nm,
retention time: R body: 6.8 minutes, S body: 7.8 minutes.

 

PATENT

Daiichi Sankyo Company,Limited, 第一三共株式会社

WO2011115064…..

http://www.google.co.in/patents/WO2011115064A1?cl=en

 

[Reference Example 1] 5 – [(tert- butoxycarbonyl) amino] -2- (diethoxyphosphoryl) valeric acid tert- butyl

Figure JPOXMLDOC01-appb-C000058

Diethylphosphonoacetate tert- butyl (20.0g) was dissolved in tetrahydrofuran (500mL), sodium hydride (63%, 3.32g) was added at 0 ℃, 15 min at 0 ℃, and stirred for 1 hour at room temperature . (3-bromopropyl) tetrahydrofuran carbamic acid tert- butyl (20.0g) (20mL) was slowly at room temperature, and the mixture was stirred at room temperature for 18 hours. A saturated aqueous solution of ammonium chloride was added to the reaction solution, the organic matter was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered to give the solvent was distilled off under reduced pressure the crude product. This silica gel column chromatography and purified (eluent hexane / ethyl acetate = 1/1-ethyl acetate) to give the title compound (26.6g).
1 H-NMR (CDCl 3) δ: 1.31-1.36 (6H, m), 1.44 (9H, m), 1.48 (9H, m), 1.51-1.59 (2H, m), 1.78-2.00 (2H, m) , 2.83 (1H, ddd, J = 22.9, 10.7, 4.4 Hz), 3.06-3.18 (2H, m), 4.10-4.18 (4H, m), 4.58 (1H, br).

[Reference Example 2] 5 – [(tert- butoxycarbonyl) amino] -2- (1H- imidazol-4-ylmethyl) valeric acid tert- butyl

Figure JPOXMLDOC01-appb-C000059

In acetonitrile (100mL) solution of the compound obtained in Reference Example 1 (8.35g), at room temperature 1,8-diazabicyclo [5.4.0] undec-7-ene (4.58mL) and lithium chloride (1 .30g) and we were added. The suspension was added with 1-trityl–1H- imidazole-4-carbaldehyde (6.90g) was stirred at room temperature overnight, under vacuum, and the solvent was evaporated. After the solution separated by adding ethyl acetate and 10% citric acid aqueous solution, an organic layer, saturated brine, and then washed with a saturated aqueous sodium bicarbonate solution and brine. Dried over anhydrous sodium sulfate, (2E) -5 – [(tert- butoxycarbonyl) amino] -2 – [(1-trityl–1H- imidazol-4-yl) methylene] valeric acid tert- butyl and (2Z) -5 – obtain [(1-trityl–1H- imidazol-4-yl) methylene] valeric acid tert- butyl mixture (11.3g) – [(tert- butoxycarbonyl) amino] -2. The mixture was suspended in methanol (500mL), 10% palladium-carbon catalyst (water content, 4g) was added and stirred for 3 days at room temperature under hydrogen atmosphere. The catalyst was removed by filtration, and the filtrate was concentrated under reduced pressure. Silica gel chromatography gave (eluting solvent: methylene chloride / methanol = 9/1) the title compound (5.60g).
1 H-NMR (CDCl 3) δ: 1.41 (9H, s), 1.44 (9H, s), 1.48-1.57 (3H, m), 1.57-1.66 (1H, m), 2.58-2.68 (1H, m) , 2.73 (1H, dd, J = 14.7, 5.3 Hz), 2.89 (1H, dd, J = 14.7, 8.4 Hz), 3.02-3.19 (2H, m), 4.67 (1H, br s), 6.79 (1H, s), 7.54 (1H, s).

[Reference Example 3] 5 – [(tert- butoxycarbonyl) amino] -2- (methoxycarbonyl) valeric acid

Figure JPOXMLDOC01-appb-C000060

Sodium methoxide in dimethyl malonate (102mL) – methanol (28%, 90.4mL) was added at room temperature and stirred at 60 ℃ 30 minutes. After cooling the white suspension solution to room temperature, (3-bromopropyl) was added carbamic acid tert- butyl (106g) in one portion and stirred at room temperature for 12 hours. Water was added to the reaction solution and the organics extracted with diethyl ether. The organic layer was successively washed with 1 N sodium hydroxide aqueous solution and saturated brine, dried over anhydrous sodium sulfate, filtered and the solvent was distilled off under reduced pressure {3 – [(tert- butoxycarbonyl) amino] propyl} malonic I got acid dimethyl of crude product. The resulting ester (94g) was dissolved in methanol (100mL), water lithium hydroxide monohydrate (13.6g) (300mL) – was added to methanol (300mL) solution at 0 ℃, 15 h stirring at room temperature It was. The methanol was distilled off under reduced pressure and the organics were extracted with ethyl acetate. 2N hydrochloric acid (160mL) was added to the aqueous layer was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered to give the solvent was distilled off under reduced pressure the crude product. This silica gel column chromatography: – is purified (eluent methylene chloride methylene chloride / methanol = 10/1) to give the title compound (69.1g).
1 H-NMR (CDCl 3) δ: 1.44 (9H, m), 1.50-1.60 (2H, m), 1.86-2.01 (2H, m), 3.07-3.20 (2H, m), 3.43 (1H, m) , 3.77 (3H, s), 4.64 (1H, br).

[Reference Example 4] 1- (trans-4- methylcyclohexyl) -1H- imidazole-4-carbaldehyde [Step 1] 1- (trans-4- methylcyclohexyl) -1H- imidazole-4-carboxylic acid ethyl

Figure JPOXMLDOC01-appb-C000061

Was dissolved in 3- (dimethylamino) -2-isocyanoethyl ethyl acrylic acid (Liebigs Annalen der Chemie, 1979 years 1444 pages) (1.52g) and the trans-4- methyl cyclohexylamine (3.07g), 70 ℃ in it was stirred for 4 hours. A saturated aqueous solution of ammonium chloride was added to the reaction solution, the organic matter was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and filtered to give the solvent was distilled off under reduced pressure the crude product. This silica gel column chromatography and purified (eluent hexane / ethyl acetate = 2 / 1-1 / 2) to give the title compound (1.90g).
1 H-NMR (CDCl 3) δ: 0.96 (3H, d, J = 6.6 Hz), 1.13 (2H, m), 1.39 (3H, d, J = 7.0 Hz), 1.47 (1H, m), 1.68 ( 2H, m), 1.88 (2H, m), 2.12 (2H, m), 3.91 (1H, tt, J = 12.1, 3.9 Hz), 4.36 (2H, q, J = 7.0 Hz), 7.54 (1H, s ), 7.66 (1H, s).

[Step 2] [1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methanol

Figure JPOXMLDOC01-appb-C000062

Lithium aluminum hydride (92%, 0.31g) it was suspended in tetrahydrofuran (6mL). The compound obtained in Step 1 of this reference example (1.50g) was dissolved in tetrahydrofuran (6mL), it was slowly added dropwise to the suspension at 0 ℃.0 After stirring for 30 min at ℃, the reaction solution was diluted with diethyl ether, it was added a saturated aqueous solution of sodium sulfate. After stirring for 1 hour at room temperature, the resulting inorganic salt was removed by filtration through Celite. The filtrate to give the crude product was concentrated under reduced pressure. Mixed solvent of this from hexane and ethyl acetate: water (5 1), to give the title compound (1.09g).
1 H-NMR (CDCl 3) δ: 0.95 (3H, d, J = 6.6 Hz), 1.04-1.17 (2H, m), 1.44 (1H, m), 1.59-1.73 (2H, m), 1.81-1.89 (2H, m), 2.04-2.13 (2H, m), 2.78 (1H, br), 3.84 (1H, tt, J = 12.1, 3.9 Hz), 4.59 (2H, s), 6.91 (1H, s), 7.49 (1H, s).

[Step 3] 1- (trans-4- methylcyclohexyl) -1H- imidazole-4-carbaldehyde

Figure JPOXMLDOC01-appb-C000063

The compound obtained in Step 2 of this reference example (1.04g) was dissolved in toluene (10mL). Aqueous solution of sodium hydrogen carbonate (1.35g) (5mL), iodine (2.72g) and 2,2,6,6-tetramethyl-1-sequential piperidinyloxy (84mg) was added and stirred for 2 hours at room temperature It was. The reaction solution was added saturated aqueous sodium thiosulfate solution and the organics were extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and filtered to give the solvent was distilled off under reduced pressure the crude product. This silica gel column chromatography and purified (eluent hexane / ethyl acetate = 1 / 1-1 / 2) to give the title compound (0.900g).
1 H-NMR (CDCl 3) δ: 0.97 (3H, d, J = 6.8 Hz), 1.09-1.19 (2H, m), 1.48 (1H, m), 1.65-1.75 (2H, m), 1.87-1.93 (2H, m), 2.11-2.18 (2H, m), 3.95 (1H, tt, J = 12.2, 3.9 Hz), 7.62 (1H, s), 7.68 (1H, s), 9.87 (1H, s).

 

[Example 15] (2R) -5- amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methyl} valeric acid and (2S) -5- amino-2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} valeric acid [Step 1] 5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans 4-methylcyclohexyl) -1H- imidazole-4-yl] methyl} methyl valerate

Figure JPOXMLDOC01-appb-C000124

The compound obtained in Reference Example 4 (300mg) and the compound obtained in Reference Example 3 (860mg) was suspended in cyclohexane (10mL). Piperidine (0.154mL) and cyclohexane propionic acid (0.116mL) and (10mL) solution was added, and the mixture was heated under reflux for 48 hours. After cooling, aqueous potassium carbonate solution was added to the reaction solution, and the organic matter was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure. The obtained crude product was dissolved in ethanol (12mL), 10% palladium-carbon catalyst (water, 250mg) was added and atmospheric pressure hydrogen atmosphere at room temperature for 4 hours and stirred at 60 ℃ 2.5 hours. After Celite filtration, to give the crude product and the filtrate was concentrated under reduced pressure. This silica gel column chromatography and purified (eluent hexane / ethyl acetate = 2 / 1-1 / 3) to give the title compound (562mg).
1 H-NMR (CDCl 3) δ: 0.94 (3H, d, J = 6.6 Hz), 1.02-1.15 (2H, m), 1.34-1.69 (7H, m), 1.43 (9H, s), 1.80-1.87 (2H, m), 1.99-2.09 (2H, m), 2.69 (1H, dd, J = 13.7, 6.3 Hz), 2.79 (1H, m), 2.88 (1H, dd, J = 13.7, 7.4 Hz), 3.03-3.13 (2H, m), 3.63 (3H, s), 3.79 (1H, tt, J = 12.1, 3.9 Hz), 4.76 (1H, br), 6.67 (1H, s), 7.47 (1H, s) .

[Step 2] (2R) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} methyl valerate and ( 2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} methyl valerate

Figure JPOXMLDOC01-appb-C000125

The compound obtained in Step 1 of this Example (40mg) was dissolved in hexane (1.5mL) and ethanol (0.5mL), using CHIRALPAK IA semi-preparative column (2.0cm × 25.0cm) It was optically resolved by high performance liquid chromatography. Flow rate: 15mL / min, elution solvent: hexane / ethanol = 75/25, detection wavelength: 220nm.

The solvent of the divided solution was evaporated under reduced pressure to give both enantiomers each (15mg). Both enantiomers were confirmed to be optically pure by analytical HPLC. Column: CHIRALPAK IA (0.46cm × 25.0cm), flow rate: 1mL / min, elution solvent: hexane / ethanol = 80/20 <v / v>, detection wavelength: 220nm, retention time: (2R) -5- [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} methyl valerate (7.2 min), (2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} methyl valerate (11.2 min).

[Step 3] (2R) -5- amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methyl} valerate

Figure JPOXMLDOC01-appb-C000126

Obtained in Step 2 of this Example (2R) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl } the methyl valerate (15.0mg) was added to 5 N hydrochloric acid (2mL), and the mixture was heated under reflux for 4 hours. After cooling, the solvent it was evaporated under reduced pressure. The resulting crude hydrochloride salt was dissolved in methanol, was added DOWEX50WX8-200. After the resin was washed with water and eluted with 4% aqueous ammonia. The eluate was concentrated, the crude product was washed with acetone to give the title compound (2.2mg).

[Step 4] (2S) -5- amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methyl} valerate

Figure JPOXMLDOC01-appb-C000127

Obtained in Step 2 of this Example (2S) -5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl } the methyl valerate (15.0mg) was added to 5 N hydrochloric acid (2mL), and the mixture was heated under reflux for 4 hours. After cooling, the solvent it was evaporated under reduced pressure. The resulting crude hydrochloride salt was dissolved in methanol, was added DOWEX50WX8-200 (200mg). After the resin was washed with water, ammonia water (4%, 80mL) and eluted with. The eluate was concentrated, the crude product was washed with acetone to give the title compound (1.8mg).

[Example 16] 5-amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazole-4-yl] methyl} valeric acid benzyl hydrochloride [Step 1] 5 – [(tert- butoxycarbonyl) amino] -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} valerate

Figure JPOXMLDOC01-appb-C000128

The compound obtained in step 1 of Example 15 (7.00g) was dissolved in a mixed solvent consisting of tetrahydrofuran (70mL) and water (14mL), lithium hydroxide monohydrate and (1.26g) at room temperature The mixture was stirred overnight.The reaction solution 2 N hydrochloric acid (8.6mL) was added to neutralize, followed by distilling off the solvent under reduced pressure. The resulting residue was dried with anhydrous sodium sulfate added methylene chloride was to give the crude product was distilled off the solvent under reduced pressure the title compound. This it was used in the next reaction.
MS (ESI) m / z 394 [M + H] +.

[Example 40] (2S) -5- Amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} valerate · p- toluenesulfonate, anhydrous

Figure JPOXMLDOC01-appb-C000196

The compound obtained in Step 4 of Example 15 (2.04g) was suspended stirring in tetrahydrofuran (15mL), p- toluenesulfonate monohydrate (1.32g) was added, at room temperature for 1 day the mixture was stirred. The precipitated crystals were collected by vacuum filtration to obtain dried in one day like the title compound (3.01g).
1 H-NMR (CD 3 OD) δ: 0.95 (3H, d, J = 6.5 Hz), 1.11-1.21 (2H, m), 1.43-1.79 (7H, m), 1.83-1.89 (2H, m), 2.05-2.10 (2H, m), 2.37 (3H, s), 2.57-2.64 (1H, m), 2.70 (1H, dd, J = 14.5, 5.5 Hz), 2.85-2.95 (3H, m), 4.07 ( 1H, tt, J = 11.7, 3.9 Hz), 7.18 (1H, s), 7.23 (2H, d, J = 7.8 Hz), 7.70 (2H, d, J = 8.2 Hz), 8.22 (1H, s).
Elemental analysis: C 16 H 27 N 3 O 2 · C 7 H 8 O 3 S,
Theoretical value: C; 59.33, H; 7.58, N; 9.02, O; 17.18, S; 6.89,
Measured value: C; 59.09, H; 7.53, N; 8.92, O; 17.22, S; 6.78.
———————————-.

[Example 41] (2S) -5- Amino -2 – {[1- (trans-4- methylcyclohexyl) -1H- imidazol-4-yl] methyl} valerate · p- toluenesulfonate & 1 Water hydrate

Figure JPOXMLDOC01-appb-C000197

The obtained compound (101.6mg) in 6% water-containing tetrahydrofuran (600μL) was added in Example 40, was dissolved by heating at 60 ℃. Was allowed to stand at room temperature for 1 day, it was collected by filtration and the precipitated crystals were obtained by dried for one day wind the title compound (79.3mg).
Elemental analysis: C 16 H 27 N 3 O 2 · C 7 H 8 O 3 S · 1H 2 O,
Theoretical value: C; 57.12, H; 7.71, N; 8.69, O; 19.85, S; 6.63,
Measured value: C; 56.90, H; 7.69, N; 8.67, O; 19.81, S; 6.42.

References

Study to Assess the Safety, Pharmacokinetics, and Pharmacodynamics of DS-1040b in Subjects With Acute Ischemic Stroke (NCT02586233

Phase I Study to Evaluate the Safety and Tolerability of DS-1040b Intravenous Infusion With Clopidogrel in Healthy Subjects (NCT02560688)

Study of the Effects of Ethnicity on the Pharmacokinetics, Pharmacodynamics and Safety of DS-1040b (NCT02647307)

Edo, N.; Noguchi, K.; Ito, Y.; Morishima, Y.; Yamaguchi, K.
Hemorrhagic risk assessment of DS-1040 in a cerebral ischemia/reperfusion model of rats with hypertension and hyperglycemia
41st Int Stroke Conf (February 17-19, Los Angeles) 2016, Abst TP283

Noguchi, K.; Edo, N.; Ito, Y.; Morishima, Y.; Yamaguchi, K.
Improvement of cerebral blood flow with DS-1040 in a rat thromboembolic stroke model
41st Int Stroke Conf (February 17-19, Los Angeles) 2016, Abst TP271

Lapchak, P.A.; Boitano, P.D.; Noguchi, K.
DS-1040 an inhibitor of the activated thrombin activatable fibrinolysis inhibitor improves behavior in embolized rabbits
41st Int Stroke Conf (February 17-19, Los Angeles) 2016, Abst WP282 

A first-in-human, single ascending dose study of DS-1040, an inhibitor of the activated form of thrombinactivatable fibrinolysis inhibitor (TAFIa), in healthy subjects
25th Congr Int Soc Thromb Haemost (ISTH) (June 20-25, Toronto) 2015, Abst PO621-MON

Dow, J.; Puri, A.; McPhillips, P.; Orihashi, Y.; Dishy, V.; Zhou, J.
A drug-drug interaction study of DS-1040 and aspirin in healthy subjects
25th Congr Int Soc Thromb Haemost (ISTH) (June 20-25, Toronto) 2015, Abst PO603-TUE

Noguchi, K.; Edo, N.; Ito, Y.; Yamaguchi, K.
Effect of DS-1040 on endogenous fibrinolysis and impact on bleeding time in rats
25th Congr Int Soc Thromb Haemost (ISTH) (June 20-25, Toronto) 2015, Abst AS145

Noguchi, K.; Edo, N.; Ito, Y.; Maejima, T.; Yamaguchi, K.
DS-1040: A novel selective inhibitor of activated form of thrombin-activatable fibrinolysis inhibitor
25th Congr Int Soc Thromb Haemost (ISTH) (June 20-25, Toronto) 2015, Abst PO203-MON

DS1040b/Aspirin Drug/Drug Interaction Study (NCT02071004)
ClinicalTrials.gov Web Site 2014, February 26

Patent ID Date Patent Title
US2014178349 2014-06-26 Cycloalkyl-Substituted Imidazole Derivative
US8609710 2013-12-17 Cycloalkyl-substituted imidazole derivative

//////DS-1040, DS 1040, phase 2, Daiichi Sankyo Co Ltd, Ischemic stroke

O=C(O)[C@@H](CCCN)Cc1cn(cn1)[C@@H]2CC[C@@H](C)CC2

O=S(=O)(O)c1ccc(C)cc1.O=C(O)[C@@H](CCCN)Cc1cn(cn1)[C@@H]2CC[C@@H](C)CC2

Share

QP Education and Qualification – What is needed?

 regulatory  Comments Off on QP Education and Qualification – What is needed?
Apr 012016
 

 

We are frequently asked about the educational requirements in order to become a Qualified Person in Europe. Comprehensive educational modules are offered, especially in the UK. These training courses contain different topics like pharmaceutical law, Microbiology, Quality Management etc and require the trainee to take part in multiple courses over an extended period. But is this needed to become a QP in Europe?

Read more about QP education and qualification.

see…………http://www.gmp-compliance.org/enews_05211_QP-Education-and-Qualification—What-is-needed_15432,15354,15367,S-QSB_n.html

 

We are frequently asked about the educational requirements in order to become a Qualified Person in Europe. Comprehensive educational modules are offered, especially in the UK. These training courses contain different topics like pharmaceutical law, Microbiology, Quality Management etc and require the trainee to take part in multiple courses over an extended period. But is this needed to become a QP in Europe?

The answer comes in two parts.

First: If you are located in the UK then those training courses and modules might be useful in order to prepare for a QP exam (but they are not mandatory).

Second: In all other countries there is no need to go through all these modules. The European legislation does not contain any requirement for specific additional educational programmes.

In order to become a Qualified Person in Europe the requirements are laid down in the EU Directive 2001/83:

1) A qualified person shall be in possession of a diploma, certificate or other evidence of formal qualifications awarded on completion of a university course of study, or a course recognized as equivalent by the Member State concerned, extending over a period of at least four years of theoretical and practical study in one of the following scientific disciplines: pharmacy, medicine, veterinary medicine, chemistry, pharmaceutical chemistry and technology, biology. Further special cases are explained in more detail in Article 48 of the EU Directive.
2) The qualified person shall have acquired practical experience over at least two years, in one or more undertakings which are authorized to manufacture medicinal products, in the activities of qualitative analysis of medicinal products, of quantitative analysis of active substances and of the testing and checking necessary to ensure the quality of medicinal products. The duration of practical experience may be reduced by one year where a university course lasts for at least five years and by a year and a half where the course lasts for at least six years.

Each EU Member State has implemented the above mentioned EU Directive into national law. There are slight differences in each EU Member State, but with the exception of the UK all other EU Member States do not require a defined exam by an official body in order to be named as a QP on the manufacturing license of a pharmaceutical company.

However, it is strongly recommended that Qualified Persons receive initial and ongoing education in Good Manufacturing Practice (GMP). The ECA Academy for example offers a wide range of GMP training courses which are helpful for QPs. But QPs and those who want to become a QP are free to select those topics which are of interest and are helpful to perform the daily work as a QP rather than a standardized scheme. This means that the recommended training course should be different for a QP working in aseptic manufacturing compared to a QP in solid dosage form manufacturing, herbal drug manufacturing or in biopharmaceutical manufacturing. There is no “one fits all approach”.

You may want to read the article The role of the Qualified Person in pharmaceutical legislation to learn more.

 

////////// Qualified Person in Europe, pharmaceutical law, Microbiology, Quality Management, QP education, qualification,

Share

EDQM adopts revised monograph for WFI allowing non-destillation techniques

 regulatory  Comments Off on EDQM adopts revised monograph for WFI allowing non-destillation techniques
Apr 012016
 

 

In a press release the EDQM has announced that the new monograph draft on Water for Injection (169) had been adopted. Read on to learn more about the production of WFI with membrane systems.

http://www.gmp-compliance.org/enews_05274_EDQM-adopts-revised-monograph-for-WFI-allowing-non-destillation-techniques_15254,15160,15090,15267,Z-PEM_n.html

 

In a press release, the European Pharmacopeia Commission has announced that the revised monograph on Water for Injection (WFI) had been adopted.

According to the revised monograph, it will be allowed in Europe in future to produce WFI with a purification method equivalent to distillation like e.g. reverse osmosis coupled with appropriate techniques. Moreover, the EDQM declares that a notice to the respective supervisory authorities will be required when a “non-distillation” technology is used for the production of WFI. Besides, the EDQM points out that it is not only a matter of equivalence of a specification but rather the robustness of the purification of WFI. Therefore, Annex 1, which is currently under revision, will also include requirements with regard to the production of WFI. The new Annex 1 will be available when the revised monograph becomes applicable.

With the modification of this monograph, harmonisation with the US Pharmacopeia and the Japanese Pharmacopeia goes one step further. In both countries, non-distillation technologies for the production of WFI are already allowed.

The revised monograph Water for Injections (169) will be published in Ph.Eur. Supplement 9.1 and apply as of April 2017. For further information please see the EDQM’s press release.

 

//////production, WFI with membrane systems, EDQM, new monograph draft, Water for Injection (169)

Share
Follow

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

Join other followers: