Quantcast
Channel: DR ANTHONY MELVIN CRASTO Ph.D – New Drug Approvals
Viewing all 1640 articles
Browse latest View live

Wockhardt, WO 2007023507, N-[[3-[3,5-difluoro-4-[4-(tetrazol-2-yl)piperidin-1-yl]phenyl]-2-oxo-1,3-oxazolidin-5-yl]methyl]acetamide

$
0
0

 

wck 4086.1wck 4086

 

Cas 928156-95-0,

Acetamide, N-​[[(5S)​-​3-​[3,​5-​difluoro-​4-​[4-​(2H-​tetrazol-​2-​yl)​-​1-​piperidinyl]​phenyl]​-​2-​oxo-​5-​oxazolidinyl]​methyl]​-

C18H21F2N7O3
Molecular Weight: 421.401246 g/mol

N-[[3-[3,5-difluoro-4-[4-(tetrazol-2-yl)piperidin-1-yl]phenyl]-2-oxo-1,3-oxazolidin-5-yl]methyl]acetamide

Example- 14 and 15

(S)-N- { 3- [4-(4-(2H-tetrazol-2-yl)-piperidin- 1 -yl)-3 , 5-difluorophenyl] -2-oxo-oxazolidin-

5-ylmethyl }-acetamide and

(S)-N- { 3- [4-(4-(l H-tetrazol- 1 -yl)-piperidin- 1 -yl)-3 , 5-difluorophenyl] -2-oxo-oxazolidin-

5-ylmethyl }-acetamide

Figure imgf000080_0001

and

Figure imgf000080_0002

A mixture of (S)-N-{3-[4-methanesulphonyloxy piperidin-l-yl)-3,5-difluorophenyl]-2- oxo-oxazolidin-5-ylmethyl}-acetamide (1.12 mM), tetrazole (1.68 mM), and K2CO3 (1.68 mM) in DMF (6 ml) was heated for 22 hrs at 850C. The resulting mixture was poured into ice-water mixture, stirred for 30 min. And the separated solid was purified by column chromatography to obtain two isomeric products in 18% and 12% yields respectively. Isomer A: M.P. 234-2370C; MS(M+1)- 422 ; M.F. C18H21F2N7O3 Isomer B: M.P. 214-2170C; MS(M+1)- 422 ; M.F. C18H2JF2N7O3

 

WOCKHARDT LIMITED [IN/IN]; D-4, MIDC Area, Chikalthana, Aurangabad 431006 (IN)

 

Our New Drug Discovery team has developed a number of lead molecules, mainly in the area of anti-infectives; these are currently at various stages of development.

Of these molecules, the most advanced of the New Chemical Entities (NCE) is WCK 771, which has commenced Phase II human clinical trials.

WCK 771 is a broad-spectrum antibiotic, which has proven effective in treating diverse staphylococcal infections like MRSA and VISA.

Other lead molecules at various stages of pre-clinical trials are: WCK 2349, WCK 4873 and WCK 4086.

http://www.wockhardt.com/how-we-touch-lives/new-drug-discover.aspx

 

 

///////


Filed under: Uncategorized Tagged: wck, WO 2007023507, Wockhardt

TR 700, TR 701FA, Tedizolid phosphate

$
0
0
Figure US08426389-20130423-C00003

“TR-700”

5R)-3-{3-Fluoro-4-[6-(2-methyl-2H-1,2,3,4-tetrazol-5-yl)-pyridin-3-yl]-phenyl}-5-hydroxymethyl-1,3-oxazolidin-2-one

Trius Therapeutics, Inc.

US Patent Publication No. 20070155798, which is hereby incorporated by reference in its entirety, recently disclosed a series of potently anti-bacterial oxazolidinones including

Figure US08426389-20130423-C00001

wherein R═H, PO(OH)2, and PO(ONa)2.

(R)-3-(4-(2-(2-methyltetrazol-5-yl)pyridin-5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate, CAS 856867-55-5

Image for unlabelled figure

DISODIUM SALT

CAS 856867-39-5

  • C17 H16 F N6 O6 P . 2 Na
  • 2-​Oxazolidinone, 3-​[3-​fluoro-​4-​[6-​(2-​methyl-​2H-​tetrazol-​5-​yl)​-​3-​pyridinyl]​phenyl]​-​5-​[(phosphonooxy)​methyl]​-​, sodium salt (1:2)​, (5R)​-
    • DA 7218, Tedizolid phosphate disodium salt

In addition, improved methods of making the free acid are disclosed in U.S. patent application Ser. No. 12/577,089, which is assigned to Trius Therapeutics, Inc., and which is incorporated herein by reference

crystalline (R)-3-(4-(2-(2-methyltetrazol-5-yl)pyridin-5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate 1 (R═PO(OH)2), was more stable and non-hygroscopic than the salt forms that were tested. In addition, unlike typical crystallizations, where the crystallization conditions, such as the solvent and temperature conditions, determine the particular crystalline form, the same crystalline form of 1 (R═PO(OH)2) was produced using many solvent and crystallization conditions. Therefore, this crystalline form was very stable, was made reproducibly, and ideal for commercial production because it reduced the chances that other polymorphs would form contaminating impurities during production. However, in all preliminary testing, the free acid crystallized as fine particles, making filtering and processing difficult.

To overcome difficulties in filtering and processing crystalline (R)-3-(4-(2-(2-methyltetrazol-5-yl)pyridin-5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate 1 (R═PO(OH)2), processes described herein result in significantly reduced filtering time, avoid more toxic solvents, and significantly increased ease of preparing dosage forms such as tablets. It has been found that implementing various processes can control the particle size distribution of the resulting material, which is useful for making the crystalline form, and for commercial production and pharmaceutical use. Surprisingly, the process for increasing the particle size reduces the amount of the dimer impurity, in comparison to the process for making the free acid disclosed in U.S. patent application Ser. No. 12/577,089. Thus, various methods of making and using the crystalline form are also provided.

In addition, by using methods of making the free acid disclosed in U.S. patent application Ser. No. 12/577,089, which is assigned to the same assignee as in the present application, and by using the crystallization methods described herein, a crystalline free acid having at least 96% purity by weight may be formed that comprises a compound having the following formula:

Figure US08426389-20130423-C00002

(hereinafter “the chloro impurity”), i.e., (R)-5-(chloromethyl)-3-(3-fluoro-4-(6-(2-methyl-2H-tetrazol-5-yl)pyridin-3-yl)phenyl)oxazolidin-2-one in an amount less than 1%.

Similarly, by using methods of making the free acid disclosed in U.S. patent application Ser. No. 12/577,089, which is assigned to the same assignee as in the present application, and by using the crystallization methods described herein, a crystalline free acid having at least 96% purity by weight may be formed that comprises a compound having the following formula:

Figure US08426389-20130423-C00003

(hereinafter “TR-700”), i.e., 5R)-3-{3-Fluoro-4-[6-(2-methyl-2H-1,2,3,4-tetrazol-5-yl)-pyridin-3-yl]-phenyl}-5-hydroxymethyl-1,3-oxazolidin-2-one, in an amount less than 1%.

The crystalline free acid may have one or more of the attributes described herein.

In some aspects, a purified crystalline (R)-3-(4-(2-(2-methyltetrazol-5-yl)-pyridin-5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate, i.e., the free acid, has a purity of at least about 96% by weight. In some embodiments, the crystalline free acid has a median volume diameter of at least about 1.0 μm.

BRIEF DESCRIPTION OF THE DRAWINGS……http://www.google.com/patents/US8426389

FIG. 1 the FT-Raman spectrum of crystalline 1 (R═PO(OH)2).

FIG. 2 shows the X-ray powder pattern of crystalline 1 (R═PO(OH)2).

http://www.google.com/patents/US8426389

FIG. 3 shows the differential scanning calorimetry (DSC) thermogram of crystalline 1 (R═PO(OH)2).

http://www.google.com/patents/US8426389

FIG. 4 shows the 1H NMR spectrum of 1 (R═PO(OH)2).

FIG. 5 depicts the TG-FTIR diagram of crystalline 1 (R═PO(OH)2).

http://www.google.com/patents/US8426389

FIG. 6 is a diagram showing the dynamic vapor sorption (DVS) behavior of crystalline 1 (R═PO(OH)2).

FIG. 7 is a manufacturing process schematic for 1 (R═PO(OH)2) (TR-701 FA) in a tablet dosage form.

FIG. 8 is a manufacturing process schematic for 1 (R═PO(OH)2) (TR-701 FA) Compounding Solution for Lyophilization.

FIG. 9 is a manufacturing process schematic for 1 (R═PO(OH)2) (TR-701 FA) for Injection, 200 mg/vial: sterile filtering, filling, and lyophilization.

FIG. 10 is a representative particle size distribution of crystalline free acid without regard to controlling particle size distribution as also described herein.

FIG. 11 is a representative particle size distribution of crystalline free acid made using laboratory processes to control particle size described herein.

FIG. 12 is a representative particle size distribution of crystalline free acid made using scaled up manufacturing processes to control particle size described herein.

 

These impurities include

Figure US08426389-20130423-C00004

i.e., 5R)-3-{3-Fluoro-4-[6-(2-methyl-2H-1,2,3,4-tetrazol-5-yl)-pyridin-3-yl]-phenyl}-5-hydroxymethyl-1,3-oxazolidin-2-one (“TR-700”) and/or

Figure US08426389-20130423-C00005

i.e., (R)-5-(chloromethyl)-3-(3-fluoro-4-(6-(2-methyl-2H-tetrazol-5-yl)pyridin-3-yl)phenyl)oxazolidin-2-one (“chloro impurity”).

 

Cited Patent Filing date Publication date Applicant Title
US4128654 Feb 10, 1978 Dec 5, 1978 E. I. Du Pont De Nemours And Company 5-Halomethyl-3-phenyl-2-oxazolidinones
US4250318 Aug 9, 1978 Feb 10, 1981 Delalande S.A. Novel 5-hydroxymethyl oxazolidinones, the method of preparing them and their application in therapeutics
US4340606 Oct 23, 1980 Jul 20, 1982 E. I. Du Pont De Nemours And Company 3-(p-Alkylsulfonylphenyl)oxazolidinone derivatives as antibacterial agents
US4461773 Jan 5, 1984 Jul 24, 1984 E. I. Dupont De Nemours And Company P-Oxooxazolidinylbenzene compounds as antibacterial agents
US4476136 Feb 24, 1982 Oct 9, 1984 Delalande S.A. Aminomethyl-5 oxazolidinic derivatives and therapeutic use thereof
US4948801 Jul 29, 1988 Aug 14, 1990 E. I. Du Pont De Nemours And Company Aminomethyloxooxazolidinyl arylbenzene derivatives useful as antibacterial agents
US5523403 May 22, 1995 Jun 4, 1996 The Upjohn Company Tropone-substituted phenyloxazolidinone antibacterial agents
US5565571 Apr 28, 1994 Oct 15, 1996 The Upjohn Company Substituted aryl- and heteroaryl-phenyloxazolidinones
US5652238 Sep 27, 1994 Jul 29, 1997 Pharmacia & Upjohn Company Esters of substituted-hydroxyacetyl piperazine phenyl oxazolidinones
US5688792 Aug 16, 1994 Nov 18, 1997 Pharmacia & Upjohn Company Substituted oxazine and thiazine oxazolidinone antimicrobials
US6365751 Apr 17, 2001 Apr 2, 2002 Zeneca Ltd. Antibiotic oxazolidinone derivatives
US6627646 * Jul 17, 2001 Sep 30, 2003 Sepracor Inc. Norastemizole polymorphs
US6689779 May 18, 2001 Feb 10, 2004 Dong A Pharm. Co., Ltd. Oxazolidinone derivatives and a process for the preparation thereof
US7129259 Dec 1, 2004 Oct 31, 2006 Rib-X Pharmaceuticals, Inc. Halogenated biaryl heterocyclic compounds and methods of making and using the same
US7141583 Apr 23, 2001 Nov 28, 2006 Astrazeneca Ab Oxazolidinone derivatives with antibiotic activity
US7144911 Dec 24, 2003 Dec 5, 2006 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
US7202257 Jul 6, 2004 Apr 10, 2007 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
US7396847 Sep 9, 2002 Jul 8, 2008 Astrazeneca Ab Oxazolidinone and/or isoxazoline as antibacterial agents
US7462633 Jun 29, 2004 Dec 9, 2008 Merck & Co., Inc. Cyclopropyl group substituted oxazolidinone antibiotics and derivatives thereof
US7473699 Feb 25, 2003 Jan 6, 2009 Astrazeneca Ab 3-cyclyl-5-(nitrogen-containing 5-membered ring)methyl-oxazolidinone derivatives and their use as antibacterial agents
US7498350 Nov 24, 2003 Mar 3, 2009 Astrazeneca Ab Oxazolidinones as antibacterial agents
US7816379 Dec 17, 2004 Oct 19, 2010 Dong-A Pharm. Co., Ltd. Oxazolidinone derivatives
US20020115669 Aug 29, 2001 Aug 22, 2002 Wiedeman Paul E. Oxazolidinone chemotherapeutic agents
US20030166620 May 18, 2001 Sep 4, 2003 Jae-Gul Lee Novel oxazolidinone derivatives and a process for the preparation thereof
US20040180906 Dec 24, 2003 Sep 16, 2004 Flynn Daniel L Anti-inflammatory medicaments
US20050038092 Jun 29, 2004 Feb 17, 2005 Yasumichi Fukuda Cyclopropyl group substituted oxazolidinone antibiotics and derivatives thereof
US20050107435 Sep 9, 2002 May 19, 2005 Gravestock Michael B. Oxazolidinone and/or isoxazoline as antibacterial agents
US20050288286 Jul 6, 2004 Dec 29, 2005 Flynn Daniel L Anti-inflammatory medicaments
US20060116386 Nov 24, 2003 Jun 1, 2006 Astrazeneca Ab Oxazolidinones as antibacterial agents
US20060116400 Nov 24, 2003 Jun 1, 2006 Astrazeneca Ab Oxazolidinone and/or isoxazoline derivatives as antibacterial agents
US20060270637 Feb 24, 2004 Nov 30, 2006 Astrazeneca Ab Hydroxymethyl substituted dihydroisoxazole derivatives useful as antibiotic agents
US20070155798 Dec 17, 2004 Jul 5, 2007 Dong-A Pharm. Co., Ltd. Novel oxazolidinone derivatives
US20070185132 Jun 29, 2004 Aug 9, 2007 Yasumichi Fukuda Cyclopropyl group substituted oxazolidinone antibiotics and derivatives thereo
US20070191336 Dec 23, 2004 Aug 16, 2007 Flynn Daniel L Anti-inflammatory medicaments
US20070203187 Jan 22, 2007 Aug 30, 2007 Merck & Co., Inc. Cyclopropyl group substituted oxazolidinone antibiotics and derivatives thereof
US20070208062 May 24, 2005 Sep 6, 2007 Astrazeneca Ab 3-(4-(2-dihydroisoxazol-3-ylpyridin-5-yl)phenyl)-5-triazol-1-ylmethyloxazolidin-2-one derivatives as mao inhibitors for the treatment of bacterial infections
US20080021012 May 24, 2005 Jan 24, 2008 Astrazeneca Ab 3-[4-{6-Substituted Alkanoyl Pyridin-3-Yl}-3-Phenyl]-5-(1H-1,2,3-Triazol-1-Ylmethyl)-1,3-Oxazolidin-2-Ones As Antibacterial Agents
US20080021071 May 24, 2005 Jan 24, 2008 Astrazeneca Ab 3-{4-(Pyridin-3-Yl) Phenyl}-5-(1H-1,2,3-Triazol-1-Ylmethyl)-1,3-Oxazolidin-2-Ones as Antibacterial Agents
US20080064689 May 24, 2004 Mar 13, 2008 Astrazeneca Ab 3-[4-(6-Pyridin-3-Yl)-3-Phenyl] -5-(1H-1,2,3-Triazol-1-Ylmethyl)-1,3-Oxazolidin-2-Ones as Antibacterial Agents
US20090018123 Jun 19, 2006 Jan 15, 2009 Milind D Sindkhedkar Oxazolidinones Bearing Antimicrobial Activity Composition and Methods of Preparation
US20090192197 Jul 30, 2009 Dong-A Pharm. Co., Ltd. Novel oxazolidinone derivatives
US20100093669 Oct 9, 2009 Apr 15, 2010 Trius Therapeutics Methods for preparing oxazolidinones and compositions containing them
US20100227839 Sep 9, 2010 Trius Therapeutics Crystalline form of r)-3-(4-(2-(2-methyltetrazol-5-yl)pyridin- 5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate
AU2004299413A1 Title not available
AU2009200606A1 Title not available
CA2549062A1 Dec 17, 2004 Jun 30, 2005 Dong-A Pharm. Co., Ltd. Novel oxazolidinone derivatives
CN101982468A Dec 17, 2004 Mar 2, 2011 东亚制药株式会社 Novel oxazolidinone derivatives and pharmaceutical compositions comprising the derivatives
EP0312000A1 Oct 12, 1988 Apr 19, 1989 The Du Pont Merck Pharmaceutical Company Aminomethyl oxooxazolidinyl aroylbenzene derivatives useful as antibacterial agents
EP0352781A2 Jul 27, 1989 Jan 31, 1990 The Du Pont Merck Pharmaceutical Company Aminomethyloxooxazolidinyl arylbenzene derivatives useful as antibacterial agents
EP1699784A1 Dec 17, 2004 Sep 13, 2006 Dong-A Pharmaceutical Co., Ltd. Novel oxazolidinone derivatives
EP2305657A2 Dec 17, 2004 Apr 6, 2011 Dong-A Pharmaceutical Co., Ltd. Oxazolidinone derivatives
EP2435051A1 May 27, 2010 Apr 4, 2012 Trius Therapeutics Oxazolidinone containing dimer compounds, compositions and methods to make and use
IN236862A1 Title not available
JPS5799576A Title not available
KR20110071107A Title not available
NZ547928A Title not available
NZ575842A Title not available
WO1993009103A1 Oct 5, 1992 May 13, 1993 Upjohn Co Substituted aryl- and heteroarylphenyloxazolidinones useful as antibacterial agents
WO1993023384A1 Apr 21, 1993 Nov 25, 1993 Upjohn Co Oxazolidinones containing a substituted diazine moiety and their use as antimicrobials
WO1995007271A1 Aug 16, 1994 Mar 16, 1995 Michael R Barbachyn Substituted oxazine and thiazine oxazolidinone antimicrobials
WO1995014684A1 Sep 27, 1994 Jun 1, 1995 Michel R Barbachyn Esters of substituted-hydroxyacetyl piperazine phenyl oxazolidinones
WO2001094342A1 May 18, 2001 Dec 13, 2001 Cho Jong Hwan Novel oxazolidinone derivatives and a process for the preparation thereof
WO2002081470A1 Apr 3, 2002 Oct 17, 2002 Astrazeneca Ab Oxazolidinones containing a sulfonimid group as antibiotics
WO2003022824A1 Sep 9, 2002 Mar 20, 2003 Astrazeneca Ab Oxazolidinone and/or isoxazoline as antibacterial agents
WO2003035648A1 Oct 23, 2002 May 1, 2003 Astrazeneca Ab Aryl substituted oxazolidinones with antibacterial activity
WO2003047358A1 Dec 2, 2002 Jun 12, 2003 Vaughan Leslie Crow Cheese flavour ingredient and method of its production
WO2003072575A1 Feb 25, 2003 Sep 4, 2003 Astrazeneca Ab 3-cyclyl-5-(nitrogen-containing 5-membered ring) methyl-oxazolidinone derivatives and their use as antibacterial agents
WO2003072576A2 Feb 25, 2003 Sep 4, 2003 Astrazeneca Ab Oxazolidinone derivatives, processes for their preparation, and pharmaceutical compositions containing them
WO2004048350A2 Nov 24, 2003 Jun 10, 2004 Astrazeneca Ab Oxazolidinones as antibacterial agents
WO2004083205A1 Mar 16, 2004 Sep 30, 2004 Astrazeneca Ab Antibacterial 1, 3- oxazolidin -2- one derivatives
WO2005005398A2 Jun 29, 2004 Jan 20, 2005 Yasumichi Fukuda Cyclopropyl group substituted oxazolidinone antibiotics and derivatives thereof
WO2005051933A1 Nov 23, 2004 Jun 9, 2005 Vijay Kumar Kaul An improved process for the synthesis of 4-(4-benzyloxy-carbonylamino-2-fluorophenyl)-piperazine-1-carboxylic acid tert-butyl ester, a key intermediate for oxazolidinone antimicrobials and compounds prepared thereby
WO2005058886A1 Dec 17, 2004 Jun 30, 2005 Dong A Pharm Co Ltd Novel oxazolidinone derivatives
WO2005116017A1 May 24, 2005 Dec 8, 2005 Astrazeneca Ab Process for the preparation of aryl substituted oxazolidinones as intermediates for antibacterial agents
WO2006038100A1 Oct 6, 2005 Apr 13, 2006 Ranbaxy Lab Ltd Oxazolidinone derivatives as antimicrobials
WO2007023507A2 Jun 19, 2006 Mar 1, 2007 Milind D Sindkhedkar Oxazolidinones bearing antimicrobial activity composition and methods of preparation
WO2007138381A2 Oct 13, 2006 Dec 6, 2007 Delorme Daniel Phosphonated oxazolidinones and uses thereof for the prevention and treatment of bone and joint infections
WO2010042887A2 Oct 9, 2009 Apr 15, 2010 Trius Therapeutics Methods for preparing oxazolidinones and compositions containing them
WO2010091131A1 Feb 3, 2010 Aug 12, 2010 Trius Therapeutics Crystalline form of r)-3-(4-(2-(2-methyltetrazol-5-yl)pyridin-5-yl)-3-fluorophenyl)-5-hydroxymethyl oxazolidin-2-one dihydrogen phosphate
WO2010138649A1 May 27, 2010 Dec 2, 2010 Trius Therapeutics, Inc. Oxazolidinone containing dimer compounds, compositions and methods to make and use

Filed under: Uncategorized Tagged: Tedizolid phosphate, TR 700, TR-701, TR-701 FA

Genotoxic impurities: the new ICH M7 addendum to calculation of compound-specific acceptable intakes

$
0
0

Originally posted on DRUG REGULATORY AFFAIRS INTERNATIONAL:

Genotoxic impurities: the new ICH M7 addendum to calculation of compound-specific acceptable intakes

The draft for a guideline ICH M7(R1) published recently supplements the ICH-M7 guideline published last year. Read more about the calculation of compound-specific acceptable intakes of genotoxic impurities.

The final document of the ICH-Guideline M7 was published in June 2014. It describes the procedure for evaluating the genotoxic potential of impurities in medicinal products (see also our news Final ICH M7 Guideline on Genotoxic Impurities published dated 23 July 2014).

An important approach to the risk characterisation of impurities is the TTC concept (TTC = threshold of toxicological concern). According to this approach the exposure to a mutagenic impurity having the concentration of 1.5 µg per adult person per day is considered to be associated with a negligible risk. It can be used as default evaluation approach to most pharmaceuticals for long-term treatment (> 10 years)…

View original 200 more words


Filed under: Uncategorized

New FDA Requirements for the Development of Herbal Medicinal Products

$
0
0

Originally posted on DRUG REGULATORY AFFAIRS INTERNATIONAL:

The previous FDA guideline for herbal medicinal products from 2004 is supposed to be replaced by a new version. In August 2015, the FDA has presented the draft of the revised guideline. Find out more about the FDA Guideline Botanical Drug Development.

http://www.gmp-compliance.org/enews_05045_New-FDA-Requirements-for-the-Development-of-Herbal-Medicinal-Products_9397,Z-RAM_n.html

In August 2015, the FDA has published a draft of the guideline “Botanical Drug Development”. This guideline addresses issues arising from the particular nature of herbal medicinal products. After its finalization it is supposed to replace the previous guideline from June 2004.

The general approach in the development of herbal medicinal products remained unchanged since 2004. But due to the better understanding of herbal medicinal products and the experience gained during the review of the approval documents for herbals (NDAs/New Drug Applications and INDs/Investigational New Drug Applications), specific recommendations could be adjusted. Still, new sections will be supplemented to better address the late development phase.

The…

View original 34 more words


Filed under: Uncategorized

Efficacy and Safety of Olive in the Management of Hyperglycemia

$
0
0

 

Postprandial hyperglycemia indicates the abnormality in glucose turnover leading to the onset of type 2 diabetes. Therefore, correction of postprandial hyperglycemia is crucial in the early stage of diabetes therapy. One of the most effective strategies to control postprandial hyperglycemia is medication combined with intake restriction and an exercise program. However, along with the prevalence of chronic diseases with multi-pathogenic factor, drugs with single chemical composition are usually not effective. In this view, phytotherapy has a promising future in the management of diabetes, considered to have less side effects as compared to synthetic drugs.

The World Health Organization estimates that in developing countries about 80% of the population now still depend on herbal treatment. Olive (Olea europea) (OE) has been used in traditional remedies in Europe and Mediterranean countries as a food and medicine for over 5,000 years especially for the prevention and treatment of chronic diseases such as hypertension, atherosclerosis , cancer and diabetes. In addition, olive is considered as the most important component of the Mediterranean diet with many health benefits.

Several experimental studies have demonstrated the beneficial effect of OE on diabetes. This effect has been demonstrated in the animal models such as streptozotocin-induced diabetic rats, alloxaninduced diabetic rats and obese diabetic sand rats fed a hypercaloric diet. In these models olive extracts have been shown to exhibit a significant reduction on both blood glucose and insulin levels. Few randomized clinical trials have demonstrated the beneficial effect of olive and one study has shown that the subjects treated with olive leaf extract exhibited significantly lower Glycated hemoglobin (HbA1c) and fasting plasma insulin levels.

Another study performed in recent onset type 2 diabetic patients has revealed that OE leaves exhibited antidiabetic activity when it added as a mixture of extract of leaves of Juglans regia, Urtica dioica and Atriplex halimus. The underlying mechanism seems to be the improvement of glucose uptake and no side effect was reported while extracts from OE have been found to exhibit cytotoxic effects only at concentrations higher than 500 μg/ mL in cells from the liver hepatocellular carcinoma cell line (HepG2) and cells from the rat L6 muscle cell line. As far as the phytochemical analysis is concerned, it is now well-established that major fatty acid constituents and minor phenolic components in olives and olive oil exert important health benefits particularly for cardiovascular diseases, metabolic syndrome and inflammatory conditions.

Hydroxytyrosol and oleuropein are considered as major polyphenolic compounds in olive leaf. Oleuropeoside, a phenylethanoid isolated from OE demonstrated a significant hypoglycemic activity in alloxan-induced diabetes and the hypoglycemic activity of this compound may result from both the increased peripheral uptake of glucose and potentiation of glucose-induced insulin secretion. In addition, Maslinic acid (MA), a natural triterpene from OE with hypoglycemic activity is a wellknown inhibitor of glycogen phosphorylase in diabetic rats without affecting hematological, histopathologic and biochemical variables, thus suggesting a sufficient margin of safety for its putative use as a nutraceutical. More recently a study has showed that MA exerts antidiabetic effects by increasing glycogen content and inhibiting glycogen phosphorylase activity in HepG2 cells.

Furthermore, MA was shown to induce the phosphorylation level of insulin-receptor β-subunit, protein kinase B (Akt) and glycogen synthase kinase-3β. MA treatment of mice fed with a high-fat diet reduced the model-associated adiposity, mRNA expression of proinflammatory cytokines and then insulin resistance, and increased the accumulated hepatic glycogen.

Finally, a recent clinical study has revealed that supplementation with olive leaf polyphenols significantly improved insulin sensitivity and pancreatic β-cell secretory capacity in overweight middle-aged men at risk of developing the metabolic syndrome. In conclusion, OE has been and continue to represent a natural source of phytocompounds eliciting a beneficial effect in human health especially in the management of hyperglycemia [115].

 

 

 

 

 

 

 

 

 

Prof. Mohamed Eddouks

Dean, Polydisciplinary Faculty of Errachidia

Moulay Ismail University, Morocco

Professor of Physiology/Pharmacology
Email: Mohamed.eddouks@laposte.net
Qualifications
1997  Ph.D., University of Sidi Mohammed Ben Abdellah, Fez
1994  Postdoctoral, University of Montreal, Montreal
1993  Ph.D., University of Liège, Belgium
1990  M.Sc., University Paris 6, France

RESEARCH EXPERIENCE

  • Oct 1995–present, Professor
    Université Moulay Ismail · Department of Biology · Physiology and endcorine Pharmacology
    Morocco · Errachidia, Meknès-Tafilalet
    -Professor (2001 until now) -Vice Dean of Scientific Research and Cooperation Faculty of Sciences and Techniques Errachidia (2005-2008 -Dean Polydisciplinary faculty of Errachidia (2008-2012)
Publications (Selected)
  1. Eddouks M, Chattopadhyay D, Zeggwagh NA.Animal models as tools to investigate antidiabetic and anti-inflammatory plants.Evid Based Complement Alternat Med. 2012;2012:142087.
  2. Zeggwagh NA, Michel JB, Eddouks M.Vascular Effects of Aqueous Extract of Chamaemelum nobile: In Vitro Pharmacological Studies in Rats.Clin Exp Hypertens. 2012.
  3. Oufni L, Taj S, Manaut B, Eddouks M. 2011.Transfer of uranium and thorium from soil to different parts of medicinal plants using SSNTD. Journal of Radioanalytical and Nuclear Chemistry, 287; 403-411.
  4. Zeggwagh NA, Moufid A, Michel JB, Eddouks M. Hypotensive effect of Chamaemelum nobile aqueous extract in spontaneously hypertensive rats.Clin Exp Hypertens. 2009.31(5):440-50.
  5. Zeggwagh NA, Farid O, Michel JB, Eddouks M. Cardiovascular effect of Artemisia herba alba aqueous extract in spontaneously hypertensive rats.Methods Find Exp Clin Pharmacol. 2008. 30(5):375-81.
  6. Eddouks M, Maghrani M, Louedec L, Haloui M, Michel JB.Antihypertensive activity of the aqueous extract of Retama raetam Forssk. leaves in spontaneously hypertensive rats.J Herb Pharmacother. 2007;7(2):65-77.
  7. Zeggwagh, N-A., Eddouks, M . Anti-hyperglycaemic and hypolipidemic effects of Ocimum basilicum aqueous extract in diabetic rats. American Journal of Pharmacology and Toxicology. 2(3): 123-129, 2007.
  8. Lemhadri, A., Burcelin, R., Eddouks, M. Chamaemelum nobile L. aqueous extract represses endogenous glucose production and improves insulin sensitivity in streptozotocin-induced diabetic mice. American Journal of Pharmacology and Toxicology. 2(3): 116-122, 2007.
  9. Lemhadri, A., Eddouks, M., Burcelin, R. Anti-hyperglycaemic and anti-obesity effects of Capparis spinosa and Chamaemelum nobile aqueous extracts in HFD mice. American Journal of Pharmacology and Toxicology. 2(3): 106-110, 2007.
  10. Zeggwagh, N.A., Michel, J.B, and Eddouks, M. Acute Hypotensive and Diuretic Activities of Chamaemelum nobile Aqueous Extract in Normal Rats. American Journal of Pharmacology and Toxicology. 2(3): 140-145, 2007.
  11. Zeggwagh, N-A., Michel, JB., Eddouks, M . Cardiovascular effect of Capapris spinosa aqueous extract in rats Part II: Furosemide-like effect of Capparis spinosa aqueous extract in normal rats. 2(3): 130-134, 2007.
  12. Zeggwagh, N-A., Michel, JB., Eddouks, M . Cardiovascular effect of Capparis spinosa aqueous extract. Part III: Antihypertensive effect in spontaneously hypertensive rats. American Journal of Pharmacology and Toxicology. 2(3): 111-115, 2007.
  13. Zeggwagh, N-A., Eddouks, M .Michel, JB. Cardiovascular effect of Capparis spinosa aqueous extract. Part VI: in vitro vasorelaxant effect.American Journal of Pharmacology and Toxicology. 2(3): 135-139, 2007.
  14. Eddouks, M., Ouahidi, M.L., Farid, O., Moufid, A., Lemhadri, A. The use of medicinal plants in the treatment of diabetes in Morocco. Phytothérapie. 2007, 5, no4, pp.194-203.
  15. Eddouks M; Khalidi A; Zeggwagh N.-A; Pharmacological approach of plants traditionally used in treating hypertension in Morocco. Phytothérapie. 2009, 7, no2, pp. 122-127.
  16. Zeggwagh NA, Ouahidi ML, Lemhadri A, Eddouks M. 2006. Study of hypoglycaemic and hypolipidemic effects of Inula viscosa L. aqueous extract in normal and diabetic rats. Journal ofEthnopharmacology. 24; 108(2): 223-7.
  17. Lemhadri A, Hajji L, Michel JB, Eddouks M. Cholesterol and triglycerides lowering activities of caraway fruits in normal and streptozotocin diabetic rats. Journal ofEthnopharmacology 2006 19; 106(3):321-6.
  18. Eddouks, M., Maghrani, M, Michel, J-B.Antihypertensive action of Lepidium sativum in SHR rats. In Press. Journal of Herbal Pharmacotherapy.Eddouks, M., Michel, J-B., Mghrani, M. Effect of Lepidium sativum L. On renal glucose reabsorption and urinary TGF B levels in diabetic rats. Phytotherapy Research. 2008 ;22(1):1-5.
  19. Eddouks M, Maghrani M, Michel JB.2005.Hypoglycaemic effect of Triticum repens P. Beauv. in normal and diabetic rats. Journal of Ethnopharmacology. 2005 ; 102(2):228-32.
  20. Eddouks, M. 2005. Les plantes anti-diabétiques. Phytothérapie Européenne. 28, 8-12.
  21. Zhang J, Onakpoya IJ, Posadzki P, Eddouks M. The safety of herbal medicine: from prejudice to evidence. Evid Based Complement Alternat Med. 2015;2015:316706.
  22. Yakubu MT, Sunmonu TO, Lewu FB, Ashafa AO, Olorunniji FJ, Eddouks M. Efficacy and safety of medicinal plants used in the management of diabetes mellitus. Evid Based Complement Alternat Med. 2014; 2014: 793035.
  23. Eddouks M, Chattopadhyay D, De Feo V, Cho WC. Medicinal plants in the prevention and treatment of chronic diseases 2013. Evid Based Complement Alternat Med. 2014;2014:180981.
  24. Eddouks M, Bidi A, El Bouhali B, Hajji L, Zeggwagh NA. Antidiabetic plants improving insulin sensitivity. J Pharm Pharmacol. 2014 Sep;66(9):1197-214.

 

 

Efficacy and Safety of Olive in the Management of Hyperglycemia

Mohamed Eddouks

Eddouks M*

Faculty of Sciences and Techniques Errachidia, Moulay Ismail university, BP 21, Errachidia, 52000, Morocco

MOHAMED EDDOUKS

Professor
Faculty of Sciences and Techniques Errachidia
Moulay Ismail University
Morocco

Dr. Mohamed Eddouks is currently working as a professor at Moulay ismail university, morocco. He worked as assistant professor at faculty of sciences and techniques errachidia (1995) and as head of the department of biology at faculty of sciences and techniques errachidia (2003). He completed his PhD degree in Physiology and Pharmacology from University of Liege, Belgium and Sidi Mohammed Ben Abdellah University. He published many articles in international journals.

 

Eddouks M
Faculty of Sciences and Techniques Errachidia
Moulay Ismail university, BP 21
Errachidia, 52000, Morocco
Tel: +212535574497
Fax: +212535574485
E-mail: mohamed.eddouks@laposte.net

Citation: Eddouks M (2015) Efficacy and Safety of Olive in the Management of Hyperglycemia. Pharmaceut Reg Affairs 4:e145. doi:10.4172/2167-7689.1000e145

 

Er Rachidi; Errachidia

………..

Morocco

////////

 

 


Filed under: ARAB MEDICINE, Uncategorized Tagged: ARAB, Efficacy and Safety, Errachidia, Faculty of Sciences and Techniques, Hyperglycemia, Management, MOROCCO, Moulay Ismail university, Olive

Defibrotide

$
0
0

Defibrotide is the sodium salt of a mixture of single-stranded oligodeoxyribonucleotides derived from porcine mucosal DNA. It has been shown to have antithrombotic, anti-inflammatory and anti-ischemic properties (but without associated significant systemic anticoagulant effects). It is marketed under the brand names Dasovas (FM), Noravid, and Prociclide in a variety of countries, but is currently not approved in the USA. The manufacturer is Gentium.

Defibrotide is used to treat or prevent a failure of normal blood flow (occlusive venous disease, OVD) in the liver of patients who have had bone marrow transplants or received certain drugs such as oral estrogens, mercaptopurine, and many others.

In 2012, an IND was filed in Japan seeking approval of the compound for the treatment of veno-occlusive disease.

Polydeoxyribonucleotides from bovine lung or other mamalian organs with molecular weight between 15,000 and 30,000 Da

CAS 83712-60-1

 

Defibrotide (Defitelio, Gentium)[1] is a deoxyribonucleic acid derivative (single-stranded) derived from cow lung or porcine mucosa. It is an anticoagulant with a multiple mode of action (see below).

It has been used with antithrombin III.[2]

Jazz Pharmaceuticals plc announced that the FDA has accepted for filing with Priority Review its recently submitted New Drug Application (NDA) for defibrotide. AS ON OCT 2015

Defibrotide is an investigational agent proposed for the treatment of patients with hepatic veno-occlusive disease (VOD), also known as sinusoidal obstruction syndrome (SOS), with evidence of multi-organ dysfunction (MOD) following hematopoietic stem-cell transplantation (HSCT).

Priority Review status is designated for drugs that may offer major advances in treatment or provide a treatment where no adequate therapy exists. Based on timelines established by the Prescription Drug User Fee Act (PDUFA), FDA review of the NDA is expected to be completed by March 31, 2016.

“The FDA’s acceptance for filing and Priority Review status of the NDA for defibrotide is an important milestone for Jazz and reflects our commitment to bringing meaningful medicines to patients who have significant unmet needs,” said Karen Smith, M.D., Ph.D., Global Head of Research and Development and Chief Medical Officer of Jazz Pharmaceuticals. “We look forward to continuing to work closely with the FDA to obtain approval for defibrotide for patients with hepatic VOD with evidence of MOD in the U.S. as quickly as possible, as there are no other approved therapies for treating this rare, often fatal complication of HSCT.”

The NDA includes safety and efficacy data from three clinical studies of defibrotide for the treatment of hepatic VOD with MOD following HSCT, as well as a retrospective review of registry data from the Center for International Blood and Marrow Transplant Research. The safety database includes over 900 patients exposed to defibrotide in the clinical development program for the treatment of hepatic VOD.

The compound was originally developed under a collaboration between Sanofi and Gentium. In December 2001, Gentium entered into a license and supply agreement with Sigma-Tau Pharmaceuticals, pursuant to which the latter gained exclusive rights to distribute, market and sell the product for the treatment of VOD in the U.S. This agreement was expanded in 2005 to include all of North America, Central America and South America.

Defibrotide was granted orphan drug designations from the FDA in July 1985, May 2003 and January 2007 for the treatment of thrombotic thrombocytopenic purpura (TTP), for the treatment of VOD and for the prevention of VOD, respectively. Orphan drug was also received in the E.U. for the prevention and treatment of hepatic veno-occlusive disease (VOD) in 2004 and for the prevention of graft versus host disease (GvHD) in 2013.

Pharmacokinetics

Defibrotide is available as an oral, intravenous, and intramuscular formulation. Its oral bioavailability is in the range of 58-70% of theparenteral forms. T1/2 alpha is in the range of minutes while T1/2 beta is in the range of hours in studies with oral radiolabelleddefibrotide. These data suggest that defibrotide, in spite of its macromolecular nature, is absorbed well after oral administration. Due to the drug’s short half-life, it is necessary to give the daily dose divided in 2 to 4 doses (see below).

Mode of action

The drug appears to prevent the formation of blood clots and to help dissolve blood clots by increasing levels of prostaglandin I2, E2, and prostacyclin, altering platelet activity, increasing tissue plasminogen activator (tPA-)function, and decreasing activity of tissue plasminogen activator inhibitor. Prostaglandin I2 relaxes the smooth muscle of blood vessels and prevents platelets from adhering to each other. Prostaglandin E2 at certain concentrations also inhibits platelet aggregation. Moreover, the drug provides additional beneficial anti-inflammatory and antiischemic activities as recent studies have shown. It is yet unclear, if the latter effects can be utilized clinically (e.g., treatment of ischemic stroke).

Unlike heparin and warfarin, defibrotide appears to have a relatively mild anticoagulant activity, which may be beneficial in the treatment of patients at high risk of bleeding complications. Nevertheless, patients with known bleeding disorders (e.g., hemophilia A) or recent abnormal bleedings should be treated cautiously and under close medical supervision.

The drug was marketed under the brand names Dasovas (FM), Noravid, and Prociclide in a variety of countries. It is currently not approved in the USA. The manufacturer is Gentium.

Usual indications

Defibrotide is used to treat or prevent a failure of normal blood flow (Veno-occlusive disease, VOD) in the liver of patients having had bone marrow transplants or received certain drugs such as oral estrogens, mercaptopurine, and many others. Without intensive treatment, VOD is often a fatal condition, leading to multiorgan failure. It has repeatedly been reported that defibrotide was able to resolve the condition completely and was well tolerated.

Other indications are: peripheral obliterative arterial disease, thrombophlebitis, and Raynaud’s phenomenon. In very high doses, defibrotide is useful as treatment of acute myocardial infarction. The drug may also be used for the pre- and postoperative prophylaxis of deep venous thrombosis and can replace the heparin use during hemodialytic treatments.

It has been investigated for use in treatment of chronic venous insufficiency.[3]

Potential indications in the future

Other recent preclinical studies have demonstrated that defibrotide used in conjunction with Granulocyte Colony-Stimulating Factor (rhG-CSF) significantly increases the number of Peripheral Blood Progenitor Cells (Stem cells). The benefit of this increase in stem cells may be crucial for a variety of clinical indications, including graft engineering procedures and gene therapy programs. This would expand the clinical usefulness of defibrotide to a complete distinct area.

Very recently (since early 2006) combination therapy trials (phase I/II) with defibrotide plus melphalan, prednisone, and thalidomide in patients with multiple myeloma have been conducted. The addition of defibrotide is expected to decrease the myelosuppressive toxicity of melphalan. However, is too early for any definitive results at that stage.

Cautions and contraindications

  • The efficacy of the drug has been reported to be poorer in patients with diabetes mellitus.
  • Pregnancy: The drug should not be used during pregnancy, because adequate and well controlled human studies do not exist.
  • Lactation: No human data is available. In order to avoid damage to the newborn, the nursing mother should discontinue either the drug or breastfeeding, taking into account the importance of treatment to the mother.
  • Known Bleeding Disorders or Bleeding Tendencies having occurred recently: Defibrotide should be used cautiously. Before initiation of treatment, the usual coagulation values should be obtained as baseline and regularly controlled under treatment. The patient should be observed regularly regarding local or systemic bleeding events.

Side-effects

Increased bleeding and bruising tendency, irritation at the injection site, nausea, vomiting, heartburn, low blood pressure. Serious allergic reactions have not been observed so far.

Drug interactions

Use of heparin with defibrotide may increase the aPTT, reflecting reduced ability of the body to form a clot. Nothing is known about the concomitant application of other anticoagulants than heparin and dextran containing plasma-expanders, but it can be anticipated that the risk of serious bleeding will be increased considerably.

References

  1.  “Jazz Pharma Acquiring Gentium for $1B”. Gen. Eng. Biotechnol. News (paper) 34 (2). January 15, 2014. p. 10.
  2.  Haussmann U, Fischer J, Eber S, Scherer F, Seger R, Gungor T (June 2006). “Hepatic veno-occlusive disease in pediatric stem cell transplantation: impact of pre-emptive antithrombin III replacement and combined antithrombin III/defibrotide therapy”. Haematologica 91 (6): 795–800. PMID 16769582.
  3.  Coccheri S, Andreozzi GM, D’Addato M, Gensini GF (June 2004). “Effects of defibrotide in patients with chronic deep insufficiency. The PROVEDIS study”. Int Angiol 23 (2): 100–7.PMID 15507885.

External links

 

 

Defibrotide
Clinical data
AHFS/Drugs.com International Drug Names
Pregnancy
category
  • X
Legal status
  • Rx only (where available)
Routes of
administration
oral, i.m., i.v.
Pharmacokinetic data
Bioavailability 58 – 70% orally (i.v. and i.m. = 100%)
Biological half-life t1/2-alpha = minutes; t1/2-beta = a few hours
Identifiers
CAS Registry Number 83712-60-1 Yes
ATC code B01AX01
DrugBank DB04932 Yes
UNII 438HCF2X0M Yes
KEGG D07423 Yes

///////////


Filed under: NDA, Priority review, Uncategorized Tagged: Defibrotide, NDA, Priority review

SILDENAFIL

$
0
0

File:Sildenafil.svg

SILDENAFIL

The chemical name of sildenafil is 5-[2-ethoxy-5-(4-methylpiperazin-1-ylsulfonyl)phenyl]-1- methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one and its formula is C22H30N6O4S. The melting point of sildenafil is 189-190oC. Its solubility is 3.5 mg/mL in water.

The 1H NMR data of sildenafil is given below. The abbreviations used are s for singlet, d for doublet, t for triplet and q for quartet. The chemical shifts are given in ppm (parts per million) and are followed by the number of Hydrogens the peaks account for:

1H NMR data:
peak (ppm) integration multiplicity
0.94 3H t
1.32 3H t
2.15 3H s
2.35 4H broad s
2.76 2H t
2.88 4H broad s
4.14 3H s
4.18 2H q
7.36 1H d
7.80 2H multiplet
12.16 1H broad s

Sildenafil, sold as Viagra and other trade names, is a medication used to treat erectile dysfunction and pulmonary arterial hypertension.[1] Its effectiveness for treating sexual dysfunction in women has not been demonstrated.[1]

Common side effects include headaches and heart burn, as well as flushed skin. Caution is advised in those who have cardiovascular disease. Rare but serious side effects include prolonged erections, which can lead to damage to the penis, and sudden-onset hearing loss. Sildenafil should not be taken by people who take nitrates such as nitroglycerin, as this may result in a severe and potentially fatal drop in blood pressure.[1]

It acts by inhibiting cGMP-specific phosphodiesterase type 5 (PDE5), an enzyme that promotes degradation of cGMP, which regulates blood flow in the penis.

It was originally discovered by Pfizer scientists Andrew Bell, David Brown, and Nicholas Terrett.[2][3] Since becoming available in 1998, sildenafil has been a common treatment for erectile dysfunction; its primary competitors are tadalafil (Cialis) and vardenafil (Levitra).

EP0463756A,US6469012,WO2008074512A1

Chemical synthesis

Dunn PJ (2005). “Synthesis of Commercial Phosphodiesterase(V) Inhibitors”. Org Process Res Dev 2005 (1): 88–97. doi:10.1021/op040019c.

The preparation steps for synthesis of sildenafil are:[40]

  1. Methylation of 3-propylpyrazole-5-carboxylic acid ethyl ester with hot dimethyl sulfate
  2. Hydrolysis with aqueous NaOH to free acid
  3. Nitration with oleum/fuming nitric acid
  4. Carboxamide formation with refluxing thionyl chloride/NH4OH
  5. Reduction of nitro group to amino
  6. Acylation with 2-ethoxybenzoyl chloride
  7. Cyclization
  8. Sulfonation to the chlorosulfonyl derivative
  9. Condensation with 1-methylpiperazine.


The synthesis of sildenafil citrate was first reported in the Bioorganic & Medicinal Chemistry Letters, Vol 6, pp. 1819, 1824, 1996. The reaction scheme is reproduced below. Sildenafil was reported in this journal as “a potent and selective inhibitor of type 5 PDE with utility for the treatment of male erectile dysfunction”.

he first step of the synthesis is the reaction of a diketoester (1) and hydrazine to give the pyrazole ring. The regioselective N-methylation of the pyrazole and hydrolysis gives a carboxylic acid (3). Compound (3) is then reacted with HNO3 and H2SO4 to give a nitrated product.
This is then followed by a carboxamide formation and the reduction of the nitro group. The compound (4) is then acylated under basic conditions and this produces the pyrazolopyrimidinone (6). (6) is then chlorosulphonylated selectively on the 5′-position of the phenyl ring. This can then couple with an amine to give sildenafil (7).
The yield of each step is given on the reaction scheme.

This is the original synthesis which was reported in the literature when the molecule was first synthesised. A variant of the synthesis was published but the changes it involved only consisted in the change of a few reactants, and no major changes were reported. This synthesis appeared in the January 1999 issue of Chemistry in Britain. This journal only reported the original discovery synthesis and said that the synthesis used commercially had not been published.

The drug is commercially manufactured by an alternative route. The reaction scheme is described in the patent which was published on 17 decembre 1997. However, the synthesis used in the commercial manufacture could be different to this. The patent was filed by the Pfizer Research and Development Company. The scheme is reproduced below.

The synthesis was described in a lot of detail, including the solvents that were the best to use, however, these details have not been reproduced here. These and further details about the synthesis can be found on the original patent document.

The reaction pathway is explained in more detail below.
Compound 2 can be prepared by the chlorosulphonation of 2-ethoxybenzoic acid (1). The conversion of compound 2 to compound 4 is achieved by N-sulphonation of 1-methylpiperazine and may be conducted in a one or two step procedure. Coupling of compound 4 with compound 6 can be achieved by any of the known amide bond-forming reactions. The aminopyrazole (6) is obtainable by the conventional reduction of the corresponding nitropyrazole (5). The resulting solution of compound 6 may be used directly after filtration in the coupling reaction with compound 4.
The cyclisation of compound 7 to give sildenafil has been achieved in yields up to 95%. Thus the overall yield of sildenafil based on compound 1 as a starting material, depending on whether the one or two step sulphonylation procedure is used can be as high as 51.7% or 47.8% respectively. This compares favourably with the first synthesis in which the overall yield is 27.6%.
The cyclisation of compound 7 to sildenafil can be conducted under neutral or acidic conditions. Under neutral conditions, compound 7 is heated, optionally in the presence of a solvent and/or optionally in the presence of a dehydrating agent and/or mechanical water removal system. Under acidic conditions, the reaction is carried out with a prolic acid or Lewis acid optionally in the presence of a solvent.

The reagents employed in the reactions can vary, but the following are among the ones recommended by the submitters of the patent:
The first step is the chlorosulphonylation of 2-ethoxybenzoic acid. This can be achieved by reacting 1 equivalent mole of thionyl chloride with 4 equivalent mole of chlorosulphonic acid. Addition of 1-methylpiperazine to an aqueous suspension of compound 2 is a suitable reaction to obtain compound 4 in one step. The carboxylic function of compound 4 can be activated using a 5% excess of N,N’-carbonyldiimidazole in ethyl acetate. This intermediate can then be reacted with imidazolide and compound 6. Compound 6 is obtainable by reduction of the corresponding nitropyrazole 5 for example by using palladium catalysed hydrogenation in ethyl acetate. Compound 7 is then cyclised to complete the reaction scheme and give sildenafil.
Information about the synthesis used to manufatcure Viagra was not available, and the two presented above are only the ones which were published. It is not surprising that the commercial manufacture of the drug is by a pathway that is not published.

………………………………………………..
SYNTHESIS

EP2024369

SCHEME2
Figure imgf000007_0001

Example 1
Preparation of 2- hydroxy-5-(4 methyl)-l-piperazinyl sulphonyl) benzoic acid Step-1: Preparation of 5-Chlorosulfonyl-2-hydroxy benzoic acid
To the chilled chlorosulfonic acid (1012 g), salicylic acid (200 g) was added at 0-50C over a period of 1 hour 40 min. The temperature of the reaction mixture was maintained at 20-250C for 2 hrs. Then thionyl chloride (172.4 g) was added over a period of 15 min and maintained for 12 hrs. The product formed was poured onto ice and maintained for lhr. The product was filtered and washed with DM water to get 5-Chlorosulfonyl-2- hydroxy benzoic acid.
Step-2: Preparation of 2-hydroxy-5-(4-methyI)-l-piperazinylsulphonyl)benzoic acid
5-Chlorosulfonyl-2-hydroxy benzoic acid (40Og) obtained in step 1 was dissolved in acetone (1200 ml) and cooled to 5-100C. To this clear solution N-methyl piperazine (254 g) was added and maintained for 2 hrs. The product formed was filtered, washed with water and purified in methanol to get 308 g of the titled compound.
NMR Data:
1H-NMR (300 MHz in DMSO-d6): δ 2.78 (3H, s), 3.17 (8H, brs), 6.85(1H, d, J = 8.7),
7.52 – 7.56 (IH, dd, J=8.7, 2.7), 7.95 (IH, d, J = 2.7)
13C-NMR (75 MHz in DMSO-d6): δ 41.98, 43.36, 51.60, 117.58, 118.33, 119.46,
130.28, 132.01, 167.63, 170.35.
Melting point: 268-2720C
Purity by HPLC: 99.4% Example 2
Preparation of 4-[2-hydroxy-5-(4-methyI-l-piperazinyIsulphonyl)benzamido]-l- methyl-3-n-propyl-lH-pyrazole-5-carboxamide
2-Hydroxy-5-(4-methyl-l-piperazinylsulphonyl)benzoic acid (10Og) was dissolved in dichloromethane (500 ml) and triethylamine (50 ml) followed by distillation to get residual mass. The residual mass was dissolved in dichloromethane (1500ml) followed by the addition of 1,3-dicyclohexylcarbodiimide (75.6 g) and 1-hydroxybenzotriazole (45g). The reaction mixture was stirred at 27-280C and then 4-amino-l-methyl-3-n-propyl- pyrazole-5-carboxamide (60.6 g) was added. The reaction mixture was heated to reflux temperature and maintained for 3 hours. Filtered the undissolved material at hot and washed the cake with dichloromethane (200ml). The filtrate was distilled out completely to get residue. Dissloved the residue in methanol (300ml) at 4O0C and then cooled the mass to 27-280C and stirred overnight. Further, cooled the mass to 5-70C and stirred for lhr. Filtered the product and washed the cake with chilled methanol (100ml) and dried to get 130 g of title compound.
NMR Data:
1H-NMR (300 MHz in DMSO-d6): δ 0.87 (3H, t, J = 7.5), 1.53-1.60 (2H, m), 2.39- 2.46(5H, m), 2.72 (4H, brs), 2.96 (4H, brs), 3.17 (3H, s), 3.91 (3H, s), 6.93 (H, d, J = 8.7), 7.57-7.61 (H, dd, J=8.7 & 2.1), NH2-(2H, brs, J =7.69 & 7.72), 8.15 (IH, d, J=2.1) 11.5 (OH, br).
13C-NMR (75 MHz in DMSO-(I6): 613.80, 21.37, 27.45, 44.05, 44.75, 48.60, 52.87, 116.37, 118.06, 119.67, 120.03, 130.64, 132.17, 132.38, 146.16, 160.83, 166.33, 166.89.
Purity by HPLC: 97.5%
Example 3
Preparation of 5-[2-hydroxy-5-(4-methylpiperazinyl-l-yl-sulphonyl)phenyl]-l- methyl -3-n- propyl-l,6-dihydro-7H-pyrazolo-[4,3-d]pyrimidin-7-one Sodium hydroxide (34 g) was added into diethylene glycol (780ml) and then heated to 110-1150C. 4-[2-hydroxy-5-(4-methyl-l-piperazinylsulphonyl)benzamido)-l-methyl-3-n- propyl-lH-pyrazole-5-carboxamide (130 g) obtained from example 2 was added to the above reaction mixture. The reaction mixture was maintained at 125-13O0C for 4-6 hrs. The reaction mixture was cooled to room temperature and then DM water (1300ml) was added slowly over 20min at 250C and maintained at this temperature for 1 hour. Filtered the mass and filtrate pH was adjusted to 6.5-7.5 with dilute hydrochloric acid at room temperature and stirred at room temperature for 2-3hrs. Product was filtered and slurried the cake with excess DM Water followed by purification in methanol to get 91 g of titled compound.
NMR Data:
1H-NMR (300 MHz in DMSO-d6): δ 0.96 (3H, t, J=7.2), 1.71-1.83 (2H, m), 2.41 (3H, s), 2.78-2.83 (6H, m), 2.99 (4H, brs), 4.15 (3H3 s), 6.93 (IH, d, J=8.7), 7.54-7.57 (IH, dd, J=8.7, 2.1), 8.47 (lH, d, J=2.1).
13C-NMR (75MHz in DMSO-d6): 513.84, 21.52, 27.20, 37.80, 43.94, 44.72,- 52.80, 115.97, 119.82, 120.19, 124.48, 128.71, 131.13, 136.46, 143.82, 151.26, 154.05, 167.24.
Purity by HPLC: 97.8%
Example 4
Preparation of 5-[2-ethoxycarbonyloxy-5-(4-methylpiperazin-l-yl-sulfonyI)phenyl]- l-methyI-3n-propyI-l,6-dihydro-7H-pyrazolo-[4,3-d]pyrimidin-7-one
5-[2-hydroxy-5-(4-methylpiperazinyl-l -yl-sulphonyl)phenyl]- 1 -methyl-3 -n- propyl- 1 ,6- dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (90 g) obtained from example 3 was dissolved in dichloromethane (360 ml) and added triethyl amine (41 ml) at room temperature and stirred for 10 min. The reaction mixture was cooled to 0-50C and followed by the addition of ethyl chloro formate (24ml) over 30 min under nitrogen atmosphere. The temperature of the reaction was raised slowly to 28-3O0C and maintained for 24 hrs. The reaction mixture was cooled to 0-50C and kept it for 1 hr. The product formed was filtered, washed with dichloromethane, dried and purified from methanol (270ml) to obtain 81 g of the title compound.
NMR Data:
1H-NMR (300 MHz in DMSO-d6): δ 0.92 (3H, t, J=7.2), 1.17 (3H, t, J=7.2), 1.68-1.75 (2H, m), 2.16 (3H, s), 3.99 (4H, br), 2.73 (2H, t, J=7.0), 4.12-4.19 (2H, t, J=6.9), 4.15 (3H, s), 7.71 (IH, d, J = 8.7), 7.93-7.97 (IH, dd, J=8.7 & 2.1), 8.01 (IH, d, J=2.0)
13C-NMR (75 MHz in DMSO-d6): 513.47, 13.80, 21.57, 27.03, 37.90, 45.72, 53.49, 65.12, 124.51, 127.65, 130.14, 130.61, 132.82, 137.30, 144.96, 146.51, 151.38, 151.66, 154.36.
Purity by HPLC: 98.6%
Example 5
Preparation of 5-[2-ethoxy-5-(4-methyl piperazine-l-ylsulfonyl)phenyl]-l-methyl-3- n-propyl-1 ,6-dihydro-7H-pyrazolo [4,3-d] pyrimidin-7-one (Sildenafil base)
5-[2-Ethoxycarbonyloxy-5-(4-methylpiperazin-l-yl-sulfonyl)phenyl]-l-methyl-3-n- propyl-l,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (50g) was dissolved in ethanol (750ml) in an autoclave and then added dicyclohexylcarbodimide (29.8g). The reaction temperature was raised to HO0C with internal pressure of 1.8-4.0 kg/cm and maintained for 6 hours followed by cooling to room temperature. The solvent was distilled off to get the crude Sildenafil base. The base thus obtained was dissolved in dichloromethane (380ml), filtered and filtrate was distilled out completely to get solid material, which is again dissolved in a mixture dichloromethane and isopropyl ether. The crude obtained was recrystallized from ethanol (260ml) to obtain 17.4gm of pure Sildenafil base.
Purity by HPLC: 99.77% Example 6
Preparation of 5-[2-Ethoxy-5-(4-methylpiperazine-l-yI-sulfonyl)phenyl]-l-methyl- 3-n-propyl-l,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one citrate (Sildenafil Citrate)
Sildenafil base (50 g) was dissolved in acetone (850 ml) at 550C and then slowly added citric acid solution (20 g in 100 ml acetone) over 45 min and maintain the reaction mixture for about 30 min. The reaction mixture was cooled, filtered and dried to get 65 g of Sildenafil citrate.
Purity by HPLC: 99.85%

Chemical synthesis

…………………………………………………………………………………
SYNTHESIS


……………………………………………


…………………………………………………………………

SYNTHESIS

…………………………………………………………………………..


……………………………
PRECURSORS

…………………………………..
SYNTHESIS


Patents

European Union

Pfizer’s patent on sildenafil citrate expired in some member countries of the EU, Austria, Denmark, France, Germany, Ireland, Italy, The Netherlands, Spain, Sweden, the United Kingdom and Switzerland on 21 June 2013.[53][54][55] A UK patent held by Pfizer on the use of PDE5 inhibitors (see below) as treatment of impotence was invalidated in 2000 because of obviousness; this decision was upheld on appeal in 2002.[56][57]

United States

In 1992, Pfizer filed a patent covering the substance sildenafil and its use to treat cardiovascular diseases.[58] This patent was published in 1993 and expired in 2012. In 1994, Pfizer filed a patent covering the use of sildenafil to treat erectile dysfunction.[59] This patent was published in 2002 and will expire in 2019. Teva sued to have the latter patent invalidated, but Pfizer prevailed in an August 2011 federal district court case.[60]

The patent on Revatio (indicated for pulmonary arterial hypertension rather than erectile dysfunction) expired in late 2012. Generic versions of this low-dose form of sildenafil have been available in the U.S. from a number of manufacturers, including Greenstone, Mylan, and Watson, since early 2013.[61] No legal barrier exists to doctors prescribing this form of sildenafil “off label” for erectile dysfunction, although the dosage typically required for treating ED requires patients to take multiple pills.

Canada

In Canada, Pfizer’s patent 2,324,324 for Revatio (sildenafil used to treat pulmonary hypertension) was found invalid by the Federal Court in June 2010, on an application by Ratiopharm Inc.[62][63]

On November 8, 2012, the Supreme Court of Canada ruled that Pfizer’s patent 2,163,446 on Viagra was invalid from the beginning because the company did not provide full disclosure in its application. The decision, Teva Canada Ltd. v. Pfizer Canada Inc., pointed to section 27(3)(b) of The Patent Act which requires that disclosure must include sufficient information “to enable any person skilled in the art or science to which it pertains” to produce it. It added further: “As a matter of policy and sound statutory interpretation, patentees cannot be allowed to ‘game’ the system in this way. This, in my view, is the key issue in this appeal.”[64]

Teva Canada launched Novo-Sildenafil, a generic version of Viagra, on the day the Supreme Court of Canada released its decision.[65][66][67] To remain competitive, Pfizer then reduced the price of Viagra in Canada.[68] However, on November 9, 2012, Pfizer filed a motion for a re-hearing of the appeal in the Supreme Court of Canada,[69] on the grounds that the court accidentally exceeded its jurisdiction by voiding the patent.[70] Finally, on April 22, 2013, the Supreme Court of Canada invalidated Pfizer’s patent altogether.[71]

India

Manufacture and sale of sildenafil citrate drugs known as “generic viagra” is common in India, where Pfizer’s patent claim does not apply. Trade names include Kamagra (Ajanta Pharma), Silagra (Cipla), Edegra (Sun Pharmaceutical), Penegra (Zydus Cadila), and Zenegra (Alkem Laboratories).

China

Manufacture and sale of sildenafil citrate drugs is common in China, where Pfizer’s patent claim is not widely enforced.

Other countries

Egypt approved Viagra for sale in 2002, but soon afterwards allowed local companies to produce generic versions of the drug, citing the interests of poor people who would not be able to afford Pfizer’s price.[72]

Pfizer’s patent on sildenafil citrate expired in Brazil in 2010.[73]

References

External link

Official Viagra Website

Sildenafil
Sildenafil.svg
Sildenafil-from-xtal-3D-balls.png
Systematic (IUPAC) name
1-[4-ethoxy-3-(6,7-dihydro-1-methyl-
7-oxo-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-5-yl)
phenylsulfonyl]-4-methylpiperazine
Clinical data
Trade names Viagra, Revatio, others
AHFS/Drugs.com monograph
MedlinePlus a699015
Licence data EMA:Link, US FDA:link
Pregnancy
category
  • US: B (No risk in non-human studies)
Legal status
Routes of
administration
Oral, IV
Pharmacokinetic data
Bioavailability 40%
Metabolism Hepatic (mostly CYP3A4, also CYP2C9)
Biological half-life 3 to 4 hours
Excretion Fecal (80%) and renal (around 13%)
Identifiers
CAS Registry Number 139755-83-2 Yes
ATC code G04BE03
PubChem CID: 5281023
DrugBank DB00203 Yes
ChemSpider 56586 Yes
UNII 3M7OB98Y7H Yes
KEGG D08514 Yes
ChEBI CHEBI:58987 Yes
ChEMBL CHEMBL1737 
PDB ligand ID VIA (PDBe, RCSB PDB)
Chemical data
Formula C22H30N6O4S
Molecular mass base: 474.6 g/mol

///////


Filed under: Uncategorized Tagged: SILDENAFIL

Merestinib

$
0
0
ChemSpider 2D Image | merestinib | C30H22F2N6O3
1206799-15-6[RN]
3-Pyridinecarboxamide, N-[3-fluoro-4-[[1-methyl-6-(1H-pyrazol-4-yl)-1H-indazol-5-yl]oxy]phenyl]-1-(4-fluorophenyl)-1,2-dihydro-6-methyl-2-oxo-[ACD/Index Name]
LY2801653
LY-2801653
Merestinib[USAN]

1206799-15-6 (Merestinib)

Chemical Formula: C30H22F2N6O3
Exact Mass: 552.17215

N-(3-fluoro-4-((1-methyl-6-(1H-pyrazol-4-yl)-1H-indazol-5-yl)oxy)phenyl)-1-(4-fluorophenyl)-6-methyl-2-oxo-1,2-dihydropyridine-3-carboxamide

  • OriginatorEli Lilly
  • ClassAmides; Antineoplastics; Dihydropyridines; Pyrazoles; Small molecules
  • Mechanism of ActionMKNK1 protein inhibitors; MKNK2 protein inhibitors; Proto oncogene protein c met inhibitors; ROS1-protein-inhibitors
  • 29 Jun 2015Immunocore in collaboration with Eli Lilly plans a phase Ib/II trial for Uveal Melanoma (Metastatic disease, Combination therapy)
  • 18 Jun 2015Eli Lilly completes a phase I bioavailability trial in healthy volunteers in USA (NCT02370485)
  • 01 Feb 2015Eli Lilly initiates enrolment in a phase I bioavailability trial in healthy volunteers in USA (NCT02370485)
Company Eli Lilly and Co.
Description C-Met inhibitor
Molecular Target c-Met receptor tyrosine kinase (c-MET) (MET) (HGFR) (c-Met proto-oncogene)
Mechanism of Action c-Met receptor tyrosine kinase inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Phase II
Standard Indication Cancer (unspecified)
Indication Details Treat advanced cancer
LY2801653, also known as Merestinib,  is an orally available, small molecule inhibitor of the proto-oncogene c-Met (mesenchymal-epithelial transition, also known as hepatocyte growth factor receptor [HGFR]) with potential antineoplastic activity. c-Met inhibitor LY2801653 selectively binds to c-Met, thereby inhibiting c-Met phosphorylation and disrupting c-Met signal transduction pathways. This may induce cell death in tumor cells overexpressing c-Met protein or expressing constitutively activated c-Met protein. This agent has potent anti-tumor efficacy in mono- and combination therapy in a broad range of cancers. c-Met, a receptor tyrosine kinase overexpressed or mutated in many tumor cell types, plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis.

 LY2801653 was identified and developed as a novel, potent, and orally active small molecule inhibitor of human c-Met. It demonstrated dose dependent inhibition of c-Met phosphorylation in xenograft tumors with a long lasting PD effect.  LY2801653 displayed potent anti-tumor efficacy in a number of non small cell lung, renal, pancreatic, and breast tumor models. Examination of c-Met expression in these tumors by immunohistochemistry (IHC) revealed a good correlation between response and c-Met expression in the tumor tissue.  LY2801653 treatment led to increase in functional vessel areas, and decrease in tumor hypoxia. Enhanced anti-tumor efficacy was achieved when Erlotinib was combined with LY2801653. . (source: http://cancerres.aacrjournals.org/cgi/content/meeting_abstract/70/8_MeetingAbstracts/3611).

   

References

1: Yan SB, Peek VL, Ajamie R, Buchanan SG, Graff JR, Heidler SA, Hui YH, Huss KL, Konicek BW, Manro JR, Shih C, Stewart JA, Stewart TR, Stout SL, Uhlik MT, Um SL,  Wang Y, Wu W, Yan L, Yang WJ, Zhong B, Walgren RA. LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and  other oncoproteins, and displays anti-tumor activities in mouse xenograft models. Invest New Drugs. 2012 Dec 29. [Epub ahead of print] PubMed PMID: 23275061.

 WATCH OUT SYNTHESIS WILL BE UPDATED…………
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00240
Abstract Image

An NH4Cl-catalyzed ethoxy ethyl deprotection was developed for the synthesis of merestinib, a MET inhibitor. Alternative reactor technologies using temperatures above the solvent boiling point are combined with this mild catalyst to promote the deprotection reaction. The reaction is optimized for flow and has been used to synthesize over 100 kg of the target compound. The generality of the reaction conditions is also demonstrated with other compounds and protecting groups.

////////


Filed under: Uncategorized

Acceptability of Draft Labeling to Support ANDA Approval Guidance for Industry

$
0
0

Ritalin-SR-20mg-1000x1000.jpg

Acceptability of Draft Labeling to Support ANDA Approval Guidance for Industry

INTRODUCTION This guidance provides recommendations and information related to the submission of proposed labeling with abbreviated new drug applications (ANDAs) under section 505(j)(2)(A)(v) of the Federal Food, Drug, and Cosmetic Act (the Act) and FDA’s implementing regulations (21 CFR 314.94(a)(8)). This guidance is intended to assist applicants submitting ANDAs under section 505(j) of the Act to the Office of Generic Drugs (OGD) in the Center for Drug Evaluation and Research (CDER). It explains FDA’s interpretation of the regulatory provision related to the submission of copies of applicants’ proposed labeling in ANDAs and clarifies that OGD will accept draft labeling and does not require the submission of final printed labeling (FPL) in order to approve an ANDA. FDA is implementing this guidance without prior public comment because the Agency has determined that prior public participation is not feasible or appropriate (see 21 CFR 10.115(g)(2) and (g)(3)). FDA made this determination because this guidance presents a less burdensome policy that is consistent with the public health. In general, FDA’s guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.

DISCUSSION OGD is issuing this guidance to provide regulated industry and other interested persons with our current thinking on the requirement that ANDA applicants submit copies of proposed labeling in their applications. Specifically, OGD is clarifying whether submission of FPL as opposed to draft labeling is required in order for OGD to approve an ANDA…………http://www.fda.gov/ucm/groups/fdagov-public/@fdagov-drugs-gen/documents/document/ucm465628.pdf

Acceptability of Draft Labeling to Support Abbreviated New Drug Application Approval; Guidance for Industry

//////


Filed under: Regulatory, Uncategorized Tagged: Acceptability of Draft Labeling, anda, Approval, Guidance for Industry, Support

Synthesis of a fluorinated Ezetimibe analogue

$
0
0

f eze nmr

Synthesis of a fluorinated Ezetimibe analogue using radical allylation of [small alpha]-bromo-[small alpha]-fluoro-[small beta]-lactam

New J. Chem., 2015, Advance Article
DOI: 10.1039/C5NJ01969A, Paper
Atsushi Tarui, Ayumi Tanaka, Masakazu Ueo, Kazuyuki Sato, Masaaki Omote, Akira Ando
*Corresponding authors
aFaculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Japan
E-mail: aando@pharm.setsunan.ac.jp

A facile and efficient synthesis of a fluorinated Ezetimibe analogue was achieved by radical allylation, Wacker oxidation, and nucleophilic arylation of [small alpha]-bromo-[small alpha]-fluoro-[small beta]-lactam

The synthesis of an α-fluoro-β-lactam-containing Ezetimibe analogue was accomplished starting from α-bromo-α-fluoro-β-lactam which was readily prepared from ethyl dibromofluoroacetate. A facile and efficient method for the introduction of the C3 alkyl side chain was realized via radical allylation. The diastereoselective allylation of α-bromo-α-fluoro-β-lactam was successfully applied to construct the relative configuration of the β-lactam nucleus between C3 and C4. Further modification of the allyl side chain gave the 3′-(4-fluorophenyl)-3′-hydroxypropyl group through Wacker oxidation and nucleophilic arylation.

http://pubs.rsc.org/en/Content/ArticleLanding/2015/NJ/C5NJ01969A?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FNJ+%28RSC+-+New+J.+Chem.+latest+articles%29#!divAbstract

 


Filed under: Uncategorized Tagged: ezetimibe, fluorinated Ezetimibe

Altiratinib

$
0
0

Altiratinib
DCC-2701; DP-5164
CAS :1345847-93-9
N-[4-({2-[(cyclopropylcarbonyl)amino]pyridin-4-yl}oxy)-2,5-difluorophenyl]-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide

N-(4-((2-(cyclopropanecarboxamido)pyridin-4-yl)oxy)-2,5-difluorophenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide

Mechanism of Action:MET/TIE2/VEGFR2/TRK (A,B,C) kinase inhibitor
Indication:invasive solid tumors. The FDA has granted altiratinib Orphan Drug Designation for glioblastoma multiforme (GBM)
Development Stage:Phase I
Developer:Deciphera Pharmaceuticals, Llc

Altiratinib, also known as DCC-270, DP-5164, is an oral, selective and  highly potent inhibitor of MET, TIE2, VEGFR2 and TRK kinases with potential anticancer activity. DCC-2701 effectively reduces tumor burden in vivo and blocks c-MET pTyr(1349)-mediated signaling, cell growth and migration as compared with a HGF antagonist in vitro. Importantly, DCC-2701’s anti-proliferative activity was dependent on c-MET activation induced by stromal human fibroblasts and to a lesser extent exogenous HGF. DCC-2701 may be superior to HGF antagonists that are in clinical trials and that pTyr(1349) levels might be a good indicator of c-MET activation and likely response to targeted therapy as a result of signals from the microenvironment.  Inhibition of MET kinase blocks a key mechanism in tumor cells that causes cancer invasiveness and metastasis. (Oncogene. 2015 Jan 8;34(2):144-53.)

DCC-2701 is an angiogenesis inhibitor acting on Tie2 receptor, HGF receptor and VEGFR-2. The product is being evaluated in phase I clinical studies at Deciphera for the oral treatment of solid tumors.

Altiratinib(DCC-2701) is a novel c-MET/TIE-2/VEGFR inhibitor; effectively reduce tumor burden in vivo and block c-MET pTyr(1349)-mediated signaling, cell growth and migration as compared with a HGF antagonist in vitro.

Altiratinib

SYNTHESIS

CLICK ON IMAGE TO ENLARGE

OR SEE

http://apisynthesisint.blogspot.in/2015/10/altiratinib-dcc-2701-dp-5164.html

IM5

IM3

IM2

IM4

PATENT

https://www.google.co.in/patents/WO2011137342A1?cl=en

Figure imgf000025_0001

Scheme 1

Figure imgf000030_0002
BEAWARE THIS IS NOT THE COMPD

Scheme 11

INTERMEDIATES

Scheme 9

[00108] A non-limiting example of Scheme 9 is illustrated below for the synthesis of 36, a specific example of 26 wherein X is F, Y is CI, and Zl , Z2, and Z3 are CH (Scheme 10). Addition of l ,2,4-trifluoro-5-nitrobenzene (33) to a solution of 2-chloropyridin-4-ol (34) and sodium hydride in DMF at 0 °C yields the nitro intermediate 35. The nitro moiety of 35 is subsequently reduced at RT in the presence of zinc dust and ammonium chloride in solution of me hanol and THF to yield amine 36.

Scheme 10

[00109] A non-limiting example of Scheme 7 is illustrated in Scheme 11, beginning with intermediate 36, prepared in Scheme 10. Thus, 36 readily reacts with acid chloride 13 (see Scheme 3) in the presence of triethylamine to yield chloro-pyridine 37. Chloro-pyridine 37 is then converted to 38, a specific example of 1 wherein Rl is F, X is F, Zl , Z2, and Z3 are CH and R3 is -C(0)CH3, upon treatment with acetamide (an example of R3-NH2 27 where R3 is acetyl) and cesium carbonate in the presence of a catalytic amount of palladium acetate and xantphos.

 NOTE 38 IS NOT THE FINAL PRODUCT

REFERENCES

Kwon Y, Smith BD, Zhou Y, Kaufman MD, Godwin AK. Effective inhibition of c-MET-mediated signaling, growth and migration of ovarian cancer cells is influenced by the ovarian tissue microenvironment. Oncogene. 2015 Jan 8;34(2):144-53. doi: 10.1038/onc.2013.539. Epub 2013 Dec 23. PubMed PMID: 24362531; PubMed Central PMCID: PMC4067476.

////Altiratinib, DCC-2701, DP-5164, Phase I, Deciphera Pharmaceuticals


Filed under: PHASE1 Tagged: Altiratinib, DCC-2701, Deciphera Pharmaceuticals, DP-5164, Phase I

Sparsentan, PS433540, RE-021

$
0
0

  Figure imgf000137_0001

Sparsentan(PS433540,RE-021)

  • C32H40N4O5S
  • Average mass592.749

4′-((2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl)-N-(4,5-dimethylisoxazol-3-yl)-2′-(ethoxymethyl)-[1,1′-biphenyl]-2-sulfonamide 

4′-[(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methvn-N-(3,4- dimethyl-5-isoxazolyl)-2′-ethoxymethyl [ 1 , l’-biphenyll -2-sulfonamide

Sparsentan
PS433540; RE-021, formerly known as DARA
CAS :254740-64-2
4-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(4,5- dimethylisoxazol-3-yl)-2-(ethoxymethyl)biphenyl-2-sulfonamide
Mechanism of Action:acting as both an Endothelin Receptor Antagonist (ERA) and Angiotensin Receptor Blocker (ARB).
Indication: Focal Segmental Glomerulosclerosis (FSGS).Focal Segmental Glomerulosclerosis (FSGS) is a rare and severe nephropathy which affects approximately 50,000 patients in the United States. Most cases of FSGS are pediatric.
Development Stage: Phase II
Developer:Retrophin, Inc

  • OriginatorBristol-Myers Squibb
  • DeveloperRetrophin
  • ClassAntihypertensives; Isoxazoles; Small molecules; Spiro compounds; Sulfonamides
  • Mechanism of ActionAngiotensin type 1 receptor antagonists; Endothelin A receptor antagonists
  • Orphan Drug Status Yes – Focal segmental glomerulosclerosis
    • 09 Jan 2015 Sparsentan receives Orphan Drug status for Focal segmental glomerulosclerosis in USA
    • 31 Dec 2013 Phase-II/III clinical trials in Focal segmental glomerulosclerosis in USA (PO)
    • 07 May 2012I nvestigation in Focal segmental glomerulosclerosis in USA (PO)

Sparsentan is an investigational therapeutic agent which acts as both a selective endothelin receptor antagonist and an angiotensin receptor blocker. Retrophin is conducting the Phase 2 DUET trial of Sparsentan for the treatment of FSGS, a rare and severe nephropathy that is a leading cause of end-stage renal disease. There are currently no therapies approved for the treatment of FSGS in the United States. Ligand licensed worldwide rights of Sparsentan (RE-021) to Retrophin in 2012 .The Food and Drug Administration (FDA) has granted orphan drug designation for Retrophins sparsentan for the treatment of focal segmental glomerulosclerosis (FSGS) in January 2015.

In 2006, the drug candidate was licensed to Pharmacopeia by Bristol-Myers Squibb for worldwide development and commercialization. In 2012, a license was obtained by Retrophin from Ligand. In 2015, Orphan Drug Designation was assigned by the FDA for the treatment of focal segmental glomerulosclerosis.

Sparsentan, also known as RE-021, BMS346567, PS433540 and DARA-a, is a Dual angiotensin II and endothelin A receptor antagonist. Retrophin intends to develop RE-021 for orphan indications of severe kidney diseases including Focal Segmental Glomerulosclerosis (FSGS) as well as conduct proof-of-concept studies in resistant hypertension and diabetic nephropathy. RE-021, with its unique dual blockade of angiotensin and endothelin receptors, is expected to provide meaningful clinical benefits in mitigating proteinuria in indications where there are no approved therapies

PATENT

WO 2000001389

https://www.google.co.in/patents/WO2000001389A1?cl=en

Figure imgf000030_0001

 

Figure imgf000033_0001

Example 41

4′- [(2-Butyl-4-oxo- 1.3-diazaspiro [4.4! non- l-en-3-yl)methyll -N-(3.4- dimethyl-5-isoxazolyl)-2′-hydroxymethyl[l, l’-biphenyl! -2-sulfonamide

Figure imgf000136_0001

A. 4′-[(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methyll-N-(3.4- dimethyl-5-isoxazolyl)-N-[(2-trimethylsilylethoxy)methyl]-2′- hydroxym ethyl [1, l’-biphenyl] -2-sulfonamide P14 (243 mg, 0.41 mmol) was used to alkylate 2-butyl-4-oxo-l,3- diazaspiro[4.4]non-l-ene hydrochloride according to General Method 4. 41A (100 mg, 35% yield) was isolated as a slightly yellow oil after silica gel chromatography using 1:1 hexanes/ethyl acetate as eluant. B. 4′- [(2-Butyl-4-oxo- 1 ,3-diazaspiro [4.41 non- l-en-3-yl)methvn -N-0.4- dimethyl-5-isoxazolyl)-2′-hydroxymethyl[l,l’-biphenyn-2- sulfonamide

Deprotection of 41A (100 mg, 0.14 mmol) according to General Method 8 (ethanol) gave the title compound as white solid in 46% yield following silica gel chromatography (96:4 methanol/chloroform eluant):

MS m/e 565 (ESI+ mode); HPLC retention time 3.21 min (Method A);

HPLC purity >98%.

Example 42

4′-[(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methvn-N-(3,4- dimethyl-5-isoxazolyl)-2′-ethoxymethyl [ 1 , l’-biphenyll -2-sulfonamide

Figure imgf000137_0001

A. 4′- [(2-Butyl-4-oxo- 1 ,3-diazaspiro [4.41 non- l-en-3-yl)methyll -N-(3 ,4- dimethyl-5-isoxazolyl)-N-[(2-methoxyethoxy)methyll-2′- hvdroxym ethyl [1 , l’-biphenyl] -2-sulfonamide

Triethylsilane (6 ml) and TFA (6 ml) were added to a solution of 5F (960 mg, 1.5 mmol) in 15 ml dichloromethane at RT. The mixture was stirred at RT for 2 h and was then concentrated. The residue was taken up in ethyl acetate and was washed successively with aqueous sodium bicarbonate, water, and brine. The organic layer was dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel using 100:2 dichloromethane/methanol to afford 42A (740 mg, 77%) as a colorless gum. Rf=0.13, silica gel, 100:5 dichloromethane/methanol. B. 4′- [(2-Butyl-4-oxo- 1.3-diazaspiro [4.41 non- l-en-3-yl)methyll -N-(3.4- dimethyl-5-isoxazolyl)-N-r(2-methoxyethoxy)methyll-2′- ethoxymethyl[l.l’-biphenyll-2-sulfonamide A mixture of 42A (100 mg, 0.15 mmol), iodoethane (960 mg, 6.1 mmol) and silver (I) oxide (180 mg, 0.77 mmol) in 0.7 ml DMF was heated at 40 ° C for 16 h.. Additional iodoethane (190 mg, 1.2 mmol) and silver (I) oxide (71 mg, 0.31 mmol) were added and the reaction mixture was heated at 40 ° C for an additional 4 h. The mixture was diluted with 1:4 hexanes/ethylacetate and was then washed with water and brine. The organic layer was dried over sodium sulfate and was then concentrated. The residue was chromatographed on silica gel using 200:3 dichloromethane/methanol as eluant to afford 42B (51mg, 49%) as a colorless gum. Rf=0.35, silica gel, 100:5 dichloromethane/methanol.

C. 4,-[(2-Butyl-4-oxo-1.3-diazaspirof4.41non-l-en-3-yl)methyll-N-(3.4- dimethyl-5-isoxazolyl )-2′-ethoxym ethyl [ 1. l’-biphenyll -2-sulfonamide

42B (51 mg) was deprotected according to General Method 7 to afford the title compound in 80% yield following preparative reverse-phase HPLC purification: white solid; m.p. 74-80 ° C (amorphous); IH NMR (CDCL, )δ0.87(tr, J=7Hz, 3H), 0.99(tr, J=7Hz, 3H), 1.32(m, 2H), 1.59(m, 2H), 1.75-2.02(m, 11H), 2.16(s, 3H), 2.35(m, 2H), 3.38 (m, 2H), 4.23(m, 2H), 4.73(s, 2H), 7.11-7.85 (m, 7H); MS m/e 593 (ESI+ mode); HPLC retention time 18.22 min. (Method E); HPLC purity >97%.

PATENT

WO 2001044239

http://www.google.co.in/patents/WO2001044239A2?cl=en

……………………

Dual angiotensin II and endothelin A receptor antagonists: Synthesis of 2′-substituted N-3-isoxazolyl biphenylsulfonamides with improved potency and pharmacokinetics
J Med Chem 2005, 48(1): 171

J. Med. Chem., 2002, 45 (18), pp 3829–3835
DOI: 10.1021/jm020138n
Abstract Image BMS 248360 A DIFFERENT COMPD

The ETA receptor antagonist (2) (N-(3,4-dimethyl-5-isoxazolyl)-4‘-(2-oxazolyl)-[1,1‘-biphenyl]-2-sulfonamide, BMS-193884) shares the same biphenyl core as a large number of AT1 receptor antagonists, including irbesartan (3). Thus, it was hypothesized that merging the structural elements of 2 with those of the biphenyl AT1 antagonists (e.g., irbesartan) would yield a compound with dual activity for both receptors. This strategy led to the design, synthesis, and discovery of (15) (4‘-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(3,4-dimethyl-5-isoxazolyl)-2‘-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-[1,1‘-biphenyl]-2-sulfonamide, BMS-248360) as a potent and orally active dual antagonist of both AT1 and ETAreceptors. Compound 15 represents a new approach to treating hypertension.

Figure

Scheme 2 a  DIFFERENT COMPD

a (a) DIBAL, toluene; (b) NaBH4, MeOH; (c) (Ph)3P, CBr4, THF (51% from 9); (d) compound 7, NaH, DMF; (e) 1 N HCl; (f) compound 4, (Ph3P)4Pd, aqueous Na2CO3, EtOH/toluene; (g) 6 N aqueous HCl/EtOH (60% from 10); (h) 13, sodium triacetoxy borohydride, AcOH, (i) diisopropylcarbodiimide, CH2Cl2 (31% from 12).

……….

WO 2010135350

http://www.google.com/patents/WO2010135350A2?cl=en

Compound 1 :

Figure imgf000003_0001

Scheme IV

Figure imgf000013_0003

Scheme V

Figure imgf000015_0001

Formula IV 1

Scheme VII

Figure imgf000016_0001

Formula Vl

Figure imgf000016_0002

A solution of 2-(2,4-dimethylphenyl)benzenesulfonic acid (Compound 12) (0.5 g, 1.9 mmol) in 50 mL of anhydrous acetonitrile was prepared and transferred to a round-bottom flask. After flushing with nitrogen gas, N-bromosuccinimide (0.75 g, 4.2 mmol) was added followed by 50 mg (0.2 mmol) of benzoyl peroxide. The solution was heated at reflux for 3 hours. The solvent was removed in-vacuo and the resulting syrup purified by silica gel chromatography (1 :1 hexanes/EtOAc) to yield Compound 13 as a white solid. 1H NMR (500 MHz, CD3CN) 8.12 (d, J = 7.5 Hz, IH), 7.92 (t, J = 7.5 Hz, IH), 7.78 (d, J= 7.5 Hz, IH), 7.74-7.71 (m, 2H), 7.68-7.65 (m, 2H), 5.12 (s, 2H), 4.70 (s, 2H). Example 4 2-(4-Bromomethyl-2-ethoxymethylphenyl)benzenesulfonic acid (Compound 14)

Figure imgf000019_0001

A solution of 20 mg (0.058 mmol) of (l-bromomethylbenzo[3,4- d])benzo[l,2-f]-2-oxa-l,l-dioxo-l-thiocycloheptane (Compound 13) in ethanol was stirred at elevated temperature until the starting material was consumed to give crude product (compound 14) that was used directly in the next step without isolation or purification.

Example 5

2-(4-((2-Butyl-4-oxo-l,3-diazaspiro[4.4]non-l-en-3-yl)methyl>2- ethoxymethylphenyl)benzenesulfonic acid (Compound 15)

Figure imgf000019_0002

To the above ethanol solution of crude 2-(4-bromomethyl-2- ethoxymethylphenyl)benzenesulfonic acid (Compound 14) described in Example 4 was added approximately 25 mL of anhydrous DMF. The ethanol was removed from the system under reduced pressure. Approximately 15 mg (0.065 mmol) of 2-butyl-l,3- diazaspiro[4.4]non-l-en-4-one (compound 7 in Scheme IV) was added followed by 300 μL of a IM solution of lithium bis-trimethylsilylamide in THF. The solution was allowed to stir at room temperature for 3 hours. The solvents were removed under reduced pressure and the remaining residue purified by preparative RP-HPLC employing a Cl 8 column and gradient elution (H2O:MeCN) affording the title compound as a white solid; [M+H]+ calcd for C27H34N2O5S 499.21, found, 499.31 ; 1H NMR (500 MHz, CD3CN) 8.04 (t, J= 5.5 Hz, IH), 7.44-7.10 (m, 2H), 7.28 (s, IH), 7.22 (d, J= 8.0 Hz, 2H), 7.08- 7.04 (m, 2H), 4.74 (br s, 2H), 4.32 (d, J= 13.0 Hz IH), 4.13 (d, J= 13.0 Hz IH), 3.40- 3.31 (m, 2H), 2.66 (t, J= 8 Hz, 2H), 2.18-2.13 (m, 5H), 1.96-1.90 (m, 2H obscured by solvent), 1.48 (m, 2H), 1.27 (s, J= 7 Hz, 2H), 1.16 (t, J= 7 Hz, 3H), 0.78 (t, J= 7.5 Hz, 3H).

Example 6

2-(4-((2-Butyl-4-oxo-l,3-diazaspiro[4.4]non-l-en-3-yl)methyl>2- ethoxymethylphenyl)benzenesulfonyl chloride (Compound 16)

Figure imgf000020_0001

To a solution of DMF (155 μL, 2 mmol, 2 equiv.) in dichloromethane (5 mL) at 0 0C was added dropwise oxalyl chloride (175 μL, 2 mmol, 2 equiv.) followed by a dichloromethane (5 mL) solution of 2-(4-((2-butyl-4-oxo-l,3-diazaspiro[4.4]non-l- en-3-yl)methyl)-2-ethoxymethylphenyl)benzenesulfonic acid (Compound 15) (0.50 g, 1.0 mmol). The resulting mixture was stirred at 0 0C for ~2 hours, diluted with additional dichloromethane (25 mL), washed with saturated sodium bicarbonate solution (10 mL), water (10 mL), and brine (10 mL), dried over sodium sulfate, and then concentrated to give crude sulfonyl chloride (compound 16) that was used without purification.

Example 7

N-(3,4-Dimethyl-5-isoxazolyl)-2-(4-(2-butyl-4-oxo-l,3-diazospiro[4.4]non-l-en- 3yl)methyl-2-ethoxymethylphenyl)phenylsulfonamide (Compound 1)

Figure imgf000021_0001

[0062] To a solution of 5-amino-3,4-dimethylisoxazole (60 mg, 0.54 mmol) in THF at -60 °C was added dropwise potassium tert-butoxide (1 mL of 1 M solution) followed by a solution of crude 2-(4-((2-butyl-4-oxo-l,3-diazaspiro[4.4]non-l-en-3- yl)methyl)-2-ethoxymethylphenyl)benzenesulfonyl chloride (Compound 16) (0.28 g, 0.54 mmol) in THF (4 mL). The resulting mixture was stirred at about -60 °C for 1 hour, allowed to warm to room temperature overnight, and then quenched with IN HCl solution to about pH 4. Standard workup of extraction with ethyl acetate, washing with water, drying, and concentration provided the final compounds as a white solid. 1H NMR (400 MHz, CDCl3) 8.03 (dd, J = 8.0 and 1.2, IH), 7.60 (td, J = 7.5 and 1.5, IH), 7.50 (td, J = 7.7 and 1.5, IH), 7.36 (s, IH), 7.28 (d, J= 2.1, 1 H), 7.25 (dd, J = 7.5 and 1.2, IH), 7.09 (dd, J= 7.9 and 1.6, IH), 6.61 (bs, IH), 4.77 (AB quartet, J= 15.5 and 8.1, 2H), 4.18 (AB quartet, J= 12.0 and 35, 2H), 3.45-3.32 (m, 2H), 2.39 (t, J= 7.5, 2H), 2.26 (s, 3H), 2.02- 1.84 (m, 8H), 1.82 (s, 3H), 1.63 (quint, J = 7.5, 2H), 1.37 (sextet, J = 7.3, 2H), 1.07 (t, J = 7.0, 3H), and 0.90 (t J= 7.3, 3H).

Example 8 l-Bromo-2-ethoxymethyl-4-hydroxymethylbenzene (Compound 17)

Figure imgf000021_0002

To a solution of ethyl 4-bromo-3-ethoxymethylbenzoate (9.4 g, 33 mmol) in toluene (56 mL) at about -10 0C was added 51 g of a 20% diisobutylaluminum hydride solution in toluene (ca. 70 mmol). The reaction was stirred at the same temperature for about 30 minutes until the reduction was completed, and then quenched with icy 5% NaOH solution to keep the temperature below about 10 °C. Organic phase of the resulting mixture was separated and the aqueous phase was extracted with toluene. The combined organic phase was concentrated in vacuo to a final volume of ~60 mL toluene solution of l-bromo-2-ethoxymethyl-4-hydroxymethylbenzene (Compound 17) that was used in next step without purification.

Example 9 l-Bromo-2-ethoxymethyl-4-methanesulfonyloxymethylbenzene (Compound 18)

Figure imgf000022_0001

To a solution of 1 -bromo-2-ethoxymethyl-4-hydroxymethylbenzene (Compound 17) (8.4 g, 33 mmol) in toluene (60 mL) prepared in Example 8 at about -10 °C was added methanesulfonyl chloride (7.9 g, 68 mmol). The reaction was stirred at the same temperature for about 30 minutes until the reduction was completed, and then quenched with icy water to keep the temperature at about 0 °C. The organic layer was separated and washed again with icy water to provide a crude product solution of 1 – bromo-2-ethoxymethyl-4-methanesulfonyloxymethylbenzene (Compound 18) that was used without purification.

Example 10

1 -Bromo-4-((2-butyl-4-oxo- 1 ,3 -diazaspiro [4.4]non- 1 -en-3 -yl)methy l)-2- ethoxymethylbenzene bisoxalic acid salt (Compound 19)

Figure imgf000022_0002

To the crude solution of 1 -bromo-2-ethoxymethyl-4- methanesulfonyloxymethylbenzene (Compound 18) (1 1 g, 33 mmol) in toluene (80 mL) prepared in Example 9 was added a 75% solution of methyltributylammonium chloride in water (0.47 mL). The resulting mixture was added to a solution of 2-butyl-4-oxo-l,3- diazaspiro[4.4]non-l-ene (compound 7 in Scheme VI) (7.5 g, 32 mmol) in dichloromethane (33 mL) pretreated with a 10 M NaOH solution (23 mL). The reaction mixture was stirred at room temperature for 2 hours until compound 18 was not longer detectable by HPLC analysis and then was quenched with water (40 mL). After stirring about 10 minutes, the organic layer was separated and aqueous layer was extracted with toluene. The combined organic phase was washed with water and concentrated to a small volume. Filtration through a silica gel pad using ethyl acetate as solvent followed by concentration yielded 1 -bromo-4-((2-buty 1-4-oxo- 1 ,3 -diazaspiro [4.4]non- 1 -en-3 – yl)methyl)-2-ethoxymethylbenzene as a crude oil product.

The crude oil was dissolved in ethyl acetate (22 mL) and warmed to around 50 °C. Anhydrous oxalic acid (4.6 g) was added to the warm solution at once and the resulting mixture was stirred until a solution was obtained. The mixture was cooled gradually and the bisoxalic acid salt (compound 19) was crystallized. Filtration and drying provided pure product (compound 19) in 50-60% yield from ethyl 4-bromo-3- ethoxymethylbenzoate in 3 steps. 1H NMR (400 MHz, CDCl3) 12.32 (bs, 4H), 7.58 (d, J = 7.8, IH), 7.36 (s, IH), 7.12 (d, J= 7.8, IH), 4.90 (s, 2H), 4.56 (s, 2H), 3.68 (q, J= 7.5, 2H), 2.87-2.77 (m, 2H), 2.40-1.95 (m, 8H), 1.62-1.53 (m, 2H), 1.38-1.28 (m, 4H), and 1.82 (t, J= 7.5, 3H).

Example 11

N-(3,4-Dimethyl-5-isoxazolyl)-2-(4-(2-butyl-4-oxo-l,3-diazospiro[4.4]non-l-en- 3yl)methyl-2-ethoxymethylphenyl)phenylsulfonamide (Compound 1)

Figure imgf000023_0001

To a suspension of l-bromo-4-((2-butyl-4-oxo-l,3-diazaspiro[4.4]non- l-en-3-yl)methyl)-2-ethoxymethylbenzene bisoxalic acid salt (Compound 19) (5.0 g, 8.3 mmol) in toluene (20 niL) under nitrogen was added water (30 mL) and pH was adjusted to 8-9 by addition of a 2 M NaOH solution at room temperature. The organic phase was separated and mixed with 2-(N-(3,4-dimethyl-5-isoxazolyl)-N- methoxymethylamino)sulfonylphenylboronic acid pinacol ester (Scheme VII, Formula IX, where R8is methoxymethyl and M = boronic acid pinacol ester) (3.6 g, 8.5 mmol), bis(dibenzylideneacetone)palladium(0) (Pd(dba)2) (0.12 g), and a standard phosphine ligand. After a 2 M sodium carbonate solution was added, the reaction mixture was warmed to 70 0C and stirred until the reaction was complete by HPLC analysis. The reaction was cooled to room temperature and quenched with water, and then separated in phases. The organic phase was treated with activated carbon, filtered through a pad of silica gel, and was concentrated to afford a crude mixture.

The crude reaction mixture was dissolved in ethanol (40 mL) after palladium catalyst was removed and was treated with 6 M HCl solution (ca. 40 mL). The mixture was warmed to 75-80 °C and stirred for about 2 hours until the reaction was completed by HPLC analysis. After the mixture was cooled to room temperature, the pH of the mixture was adjusted to 8 by addition of 10 M NaOH solution. The mixture was stirred for 2 more hours and the pH was adjusted to 6 by adding 2 M HCl and the crystal seeds. Filtration of the crystalline solid followed by drying provided N-(3,4-dimethyl-5- isoxazolyl)-2-(4-(2-butyl-4-oxo-l,3-diazospiro[4.4]non-l-en-3yl)methyl-2- ethoxymethylphenyl)phenylsulfonamide (Compound 1) as a white solid.1H NMR (400 MHz, CDCIa) 8.03 (dd, J= 8.0 and 1.2, IH), 7.60 (td, J = 7.5 and 1.5, IH), 7.50 (td, J = 7.7 and 1.5, IH), 7.36 (s, IH), 7.28 (d, J= 2.1, 1 H), 7.25 (dd, J = 7.5 and 1.2, IH), 7.09 (dd, J= 7.9 and 1.6, IH), 6.61 (bs, IH), 4.77 (AB quartet, J= 15.5 and 8.1, 2H), 4.18 (AB quartet, J= 12.0 and 35, 2H), 3.45-3.32 (m, 2H), 2.39 (t, J= 7.5, 2H), 2.26 (s, 3H), 2.02- 1.84 (m, 8H), 1.82 (s, 3H), 1.63 (quint, J= 7.5, 2H), 1.37 (sextet, J= 7.3, 2H), 1.07 (t, J = 7.0, 3H), and 0.90 (t J= 7.3, 3H).

 

 

US20040002493 * Aug 20, 2001 Jan 1, 2004 Kousuke Tani Benzoic acid derivatives and pharmaceutical agents comprising the same as active ingredient
US20070054806 * Sep 6, 2006 Mar 8, 2007 Bayer Cropscience Gmbh Novel sulfonamide-comprising solid formulations
US20070054807 * Sep 8, 2006 Mar 8, 2007 Bayer Cropscience Gmbh Storage-stable formulations of sulfonamides

.//////////////Sparsentan, PS433540, RE-021, Bristol-Myers Squibb, ORPHAN DRUG, Retrophin

O=S(C1=CC=CC=C1C2=CC=C(CN3C(CCCC)=NC4(CCCC4)C3=O)C=C2COCC)(NC5=NOC(C)=C5C)=O


Filed under: Phase2 drugs, Uncategorized Tagged: Bristol-Myers Squibb, Orphan Drug, phase 2, PS433540, RE-021, Retrophin, Sparsentan

BMS-248360, A NEW SARTAN ON HORIZON

$
0
0

BMS-248360.pngFigure imgf000095_0001

2-[4-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2-[(3,3-dimethyl-2-oxopyrrolidin-1-yl)methyl]phenyl]-N-(3,4-dimethyl-1,2-oxazol-5-yl)benzenesulfonamide

4‘-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(3,4-dimethyl-5-isoxazolyl)-2‘-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-[1,1‘-biphenyl]-2-sulfonamide,

4′- . (2-Butyl-4-oxo- 1 ,3-diazaspiro [4.41 non-l-en-3-yl)methyll -N-C3.4- dimethyl-5-isoxazolyl)-2,-[(3.3-dimethyl-2-oxo-l- pyrrolidinvDmethyll [1.1 ‘-biphenyl] -2-sulfonamide

4-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N(3,4-dimethyl-5-isoxazolyl)-2-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-[1,1-biphenyl]-2-sulfonamide

BMS-248360

PRECLINICAL …..treating hypertension

Bristol Myers Squibb Co, INNOVATOR

Hypertension remains one of the largest unmet medical needs in the 21st century, especially when one considers that hypertension is the portent of future debilitating cardiovascular disease. While many drugs are available for treating the disease, approximately one-third of the hypertensive population is still not adequately treated. Of the more recent avenues explored for treating hypertension, disruption of the effects of either angiotensin II (AII) or endothelin-1 (ET-1) has shown promise. These endogenous vasoactive peptides are among the most potent vasoconstrictors and cell proliferative factors identified to date. AII is the effector molecule of the renin−angiotensin system (RAS), and a large number of AII receptor (AT1) antagonists, including irbesartan , have been developed for treating hypertension

SYNTHESIS

picked from…….http://www.drugfuture.com/synth/syndata.aspx?ID=324487

EP 1094816; JP 2002519380; US 2002143024; WO 0001389

The intermediate biphenyl aldehyde (XI) is prepared by two related methods. 4-Bromo-3-methylbenzonitrile (I) is oxidized to aldehyde (II) via radical bromination with N-bromosuccinimide/benzoyl peroxide, followed by treatment with trimethylamine N-oxide. Suzuki coupling of aryl bromide (II) with the pinacol boronate (III) affords biphenyl (IV). After protection of the aldehyde moiety of (IV) as the corresponding ethylene ketal (V), its cyano group is reduced to aldehyde (VI) employing DIBAL in THF. Subsequent reduction of (VI) with NaBH4 leads to alcohol (VII), which is further converted into the benzyl bromide (VIII) by means of CBr4/PPh3. Bromide (VIII) is condensed with the spiro imidazolone (IX) in the presence of NaH, to produce (X). Then acidic hydrolysis of the ethylene ketal and SEM groups of (X) gives rise to the intermediate aldehyde (XI)

NEXT

Alternatively, reduction of 4-bromo-3-formylbenzonitrile ethylene ketal (XII) by means of DIBAL leads to aldehyde (XIII), which is further reduced to alcohol (XIV) with NaBH4. After bromination of (XIV) with CBr4/PPh3, the resultant benzyl bromide (XV) is condensed with the spiro imidazolone (IX), yielding (XVI). Then, acidic ketal hydrolysis in (XVI) furnishes aldehyde (XVII). Suzuki coupling between aryl bromide (XVII) and boronic acid (XVIII) gives biphenyl (XIX). The SEM group of (XIX) is then removed under acidic conditions to provide (XI)

Reductive amination of the biphenyl aldehyde (XI) with 4-amino-2,2-dimethylbutanoic acid (XX) in the presence of NaBH(OAc)3 produces aminoacid (XXI). This is finally cyclized to the corresponding lactam by treatment with DIC

Coupling of 2-bromobenzenesulfonyl chloride (I) with 5-amino-3,4-dimethylisoxazole (II) affords sulfonamide (III), which is further protected as the N-methoxyethoxymethyl derivative (IV) employing MEM-chloride in DMF. Lithiation of bromosulfonamide (IV), followed by treatment with trimethyl borate and acidic work up leads to the boronic acid intermediate (V). This is then subjected to Suzuki coupling with 4-bromo-3-methylbenzaldehyde (VI) to yield the biphenyl adduct (VII). After reduction of aldehyde (VII) to the benzylic alcohol (VIII) with NaBH4, reaction with methanesulfonyl chloride and diisopropylethylamine gives rise to the mesylate (IX) (1-3).

Mesylate (IX) is condensed with ethyl 2-propyl-4-ethylimidazole-5-carboxylate (X) yielding (XI). Simultaneous ester group hydrolysis and MEM group deprotection under acidic conditions gives rise to the imidazolecarboxylic acid (XII). This is finally coupled with methylamine via activation with CDI to produce the desired N-methyl carboxamide (1-3).

Reductive amination of the biphenyl aldehyde (XI) with 4-amino-2,2-dimethylbutanoic acid (XX) in the presence of NaBH(OAc)3 produces aminoacid (XXI). This is finally cyclized to the corresponding lactam by treatment with DIC

PAPER

J. Med. Chem., 2002, 45 (18), pp 3829–3835
DOI: 10.1021/jm020138n
Abstract Image BMS 248360

The ETA receptor antagonist (2) (N-(3,4-dimethyl-5-isoxazolyl)-4‘-(2-oxazolyl)-[1,1‘-biphenyl]-2-sulfonamide, BMS-193884) shares the same biphenyl core as a large number of AT1 receptor antagonists, including irbesartan (3). Thus, it was hypothesized that merging the structural elements of 2 with those of the biphenyl AT1 antagonists (e.g., irbesartan) would yield a compound with dual activity for both receptors. This strategy led to the design, synthesis, and discovery of (15) (4‘-[(2-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(3,4-dimethyl-5-isoxazolyl)-2‘-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-[1,1‘-biphenyl]-2-sulfonamide, BMS-248360) as a potent and orally active dual antagonist of both AT1 and ETAreceptors. Compound 15 represents a new approach to treating hypertension.

Figure

Scheme 2 a  

a (a) DIBAL, toluene; (b) NaBH4, MeOH; (c) (Ph)3P, CBr4, THF (51% from 9); (d) compound 7, NaH, DMF; (e) 1 N HCl; (f) compound 4, (Ph3P)4Pd, aqueous Na2CO3, EtOH/toluene; (g) 6 N aqueous HCl/EtOH (60% from 10); (h) 13, sodium triacetoxy borohydride, AcOH, (i) diisopropylcarbodiimide, CH2Cl2 (31% from 12).

15 as a white solid (40 mg, 31%): 

mp 104−110 °C;

1H NMR (CDCl3) δ 0.90 (t, J = 7.0 Hz, 3H), 1.08 (s, 3H), 1.14 (s, 3H), 1.36 (m, 2H), 1.61 (m, 2H), 1.75−2.06 (m, 13H), 2.17 (s, 3H), 2.39 (m, 2H), 4.18 (m, 2H), 4.71 (m, 2H), 7.02−7.93 (m, 7H);

13CNMR (CDCl3 ) δ 7.82, 11.91, 14.79, 23.36, 25.50, 25.61, 27.11, 28.81, 29.88, 35.33, 38.42, 41.48, 44.59, 46.24, 46.47, 109.29, 125.15, 125.76, 129.68, 130.58, 131.76, 133.20, 134.07, 137.15, 138.27, 139.11, 139.57, 155.81, 162.68, 162.91, 181.25, 187.83.

Anal. (C36H45N5O5S) C, H, N, S.

……………………………

PATENT

US 2002143024

http://www.google.com/patents/US20020143024

Figure US20020143024A1-20021003-C00070Zhang, H.-Y. et al., Tetrahedron, 1994, 50, 11339-11362.

Figure US20020143024A1-20021003-C00069

N-(3,4-Dimethyl-5-iso-xazolyl)-2′-formyl-4′-(hydroxy-methyl)-N-[[2-(tri-methylsilyl)ethoxy]- methyl][1,1′- biphenyl]-2- sulfonamide

Example 3 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-[1,1′-biphenyl]-2-sulfonamide

[0414]

Figure US20020143024A1-20021003-C00097

Example 3 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-[1,1′-biphenyl]-2-sulfonamide

Figure US20020143024A1-20021003-C00097

A. 4′-Cyano-2′-(1,3-dioxolan-2-yl)-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl)[1,1′-biphenyl]-2-sulfonamide

A mixture of 2B (1.28 g, 2.73 mmol), ethylene glycol (1.69 g, 27.3 mmol) and p-toluenesulfonic acid (38 mg) in toluene (30 mL) was heated at 130° C. for 5 h, while a Dean-Stark water separator was used. After cooling, the mixture was diluted with EtOAc. The organic liquid was separated and washed with H2O and brine, dried and concentrated. The residue was chromatographed on silica gel using 5:4 hexane/EtOAc to afford 3A (1.1 g, 79%) as a colorless gum: Rf=0.57, silica gel, 1:2 hexane/EtOAc.

B. 2′-(1,3-Dioxolan-2-yl)-4′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl)[1,1′-biphenyl]-2-sulfonamide

 To 3A (1.1 g, 2.14 mmol) in THF (21 mL) at 0° C. was added DIBAL-H (1M in CH2Cl2, 4.28 mL 4.28 mmol) dropwise. The reaction was stirred at RT overnight. MeOH (20 mL) was added and the reaction was stirred for 5 min. The mixture was poured into cold 0.1 N HCl solution (150 mL), shaken for 5 min, and then extracted with 3:1 EtOAc/hexane. The combined organic extracts were washed with H2O and brine, dried and concentrated. The residue was chromatographed on silica gel using 3:4 hexane/EtOAc to afford 3B (710 mg, 64%) as a colorless gum: Rf=0.45, silica gel, 2:3 hexane/EtOAc.

 C. 2′-(1,3-Dioxolan-2-yl)-4′-hydroxymethyl-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl) [1,1′-biphenyl]-2-sulfonamide

 3B (710 mg, 1.4 mmol) was subjected to sodium borohydride reduction according to General Method 11 to afford 3C, which was used for the next reaction step without further purification.

 D. 4′-Bromomethyl-2′-(1,3-dioxolan-2-yl)-N-(3,4′-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl) [1,1′-biphenyl]-2-sulfonamide

3C was treated with carbon tetrabromide and triphenylphosphine according to General Method 2. The crude residue was chromatographed on silica gel using 3:2 hexane/EtOAc to afford 3D (750 mg, 94%) as a colorless gum: Rf=0.74, silica gel, 1:2 hexane/EtOAc.

 E. 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-(1,3-dioxolan-2-yl)-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl)[1,1′-biphenyl]-2-sulfonamide

 3D (750 mg, 1.3 mmol) was treated with 2-n-butyl-1,3-diazaspiro[4.4]non-1-en-4-one hydrochloride (387 mg, 1.68 mmol) according to General Method 4. The crude residue was chromatographed on silica gel using 100:1.7 CH2Cl2/MeOH to afford 3E as a gum (830 mg, 93%): Rf=0.40, silica gel, 100:5 CH2Cl2/MeOH.

F. 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-[1,1′-biphenyl]-2-sulfonamide

3E (830 mg, 1.20 mmol) was subjected to deprotection according to General Method 7. The crude residue was chromatographed on silica gel using 100:1.5 and then 100:4 CH2Cl2 /MeOH to afford the title compound as a gum (480 mg, 72%): Rf=0.16, silica gel, 100:5 CH2Cl2/MeOH.

Example 4 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-N-(3,4-dimethyl-5-isoxazolyl)-2′-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl][1,1′-biphenyl]-2-sulfonamide

Figure US20020143024A1-20021003-C00098

 To 3F (110 mg, 0.20 mmol) in CH2Cl2 (4 mL) was added 4-amino-2,2-dimethylbutanoic acid hydrochloride (98 mg, 0.59 mmol) [Scheinmann, et al., J. Chem. Research (S), 414-415 (1993)] and 3 Å molecular sieves, followed by glacial acetic acid (35 mg, 0.59 mmol) and then sodium acetate (48 mg, 0.59 mmol). The mixture was stirred for 8 minutes, and NaB(AcO)3H (124 mg, 0.59 mmol) was then added. The reaction mixture was stirred at RT for 2 h, diluted with EtOAc and filtered through celite. The filtrate was washed with H2O and brine, dried and concentrated. This material was dissolved in CH2Cl2 (6 mL) and 1,3-diisopropylcarbodiimide (32 mg, 0.25 mmol) was added. The reaction mixture was stirred at RT for 2 h and diluted with CH2Cl2, washed with H2O and brine, dried and concentrated. The residue was purified by preparative HPLC to provide the title compound as a white solid (40 mg, 31%, for two steps): mp 104-110° C. Analysis calculated for C36H45N5O5S.0.8 H2O: Calc’d: C, 64.13; H, 6.97; N, 10.39; S, 4,75. Found: C, 64.18; H, 6.60; N, 10.23; S, 4.50.

Example 5 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-[1,1′-biphenyl]-2-sulfonamide (Alternative Preparation for 3F)

 A. 2-[(2′-Bromo-5′-formyl)phenyl)]-1,3-dioxolane

DIBAL-H (1.0 M solution in toluene, 445 mL, 445 mmol, 1.1 eq) was added over 30 minutes to a solution of 2-[(2′-bromo-5′-cyano)phenyl)]-1,3-dioxolane (103 g, 404 mmol, 1.0 eq) [Zhang, H.-Y. et al., Tetrahedron, 50, 11339-11362 (1994)] in toluene (2.0 L) at −78° C. The solution was allowed to warm to 0° C. After 1 hour, a solution of Rochelle’s salt (125 g) in water (200 mL) was added, and the mixture was allowed to warm to room temperature and was stirred vigorously for 16 h. The organic layer was concentrated and the residue partitioned between ethyl acetate (1 L) and 1 N hydrochloric acid (800 mL). The organic layer was washed with saturated aqueous sodium bicarbonate (800 mL), dried over sodium sulfate, and then concentrated to give 70.5 g of crude 5A as a yellow solid, which was used without further purification.

 B. 2-[(2′-Bromo-5′-hydroxymethyl)phenyl)]-1,3-dioxolane

Sodium borohydride (3.66 g, 96.7 mmol, 0.5 eq) was added to a solution of crude 5A (49.7 g, approximately 193 mmol, 1.0 eq) in absolute ethanol (1300 mL) at 0° C. After 2 hours, a solution of 10% aqueous sodium dihydrogen phosphate (50 mL) was added and the mixture was stirred and allowed to warm to room temperature. The mixture was concentrated, then partitioned between ethyl acetate (800 mL) and saturated aqueous sodium bicarbonate (500 mL). The organic layer was dried over sodium sulfate and concentrated to give 49.0 g of crude 5B as a yellow oil, which was used without further purification.

 C. 2-[(2′-Bromo-5′-bromomethyl)phenyl)]-1,3-dioxolane

Triphenylphosphine (52.7 g, 199 mmol, 1.05 eq) was added in portions over 15 minutes to a solution of crude 5B (49.0 g, approximately 189 mmol, 1.0 eq) and carbon tetrabromide (69.0 g, 208 mmol, 1.1 eq) in THF at 0° C. After 2 hours, saturated aqueous sodium bicarbonate solution (20 mL) was added, and the mixture was allowed to warm to room temperature and was then concentrated. Ether (500 mL) was added, and the resulting mixture was filtered. The filtrate was dried over magnesium sulfate and concentrated. The residue was chromatographed on silica gel (8:1 hexanes/ethyl acetate as eluant) to give 5C as a white solid (31.1 g, 51% yield from 2-[(2′-bromo-5′-cyano)phenyl)]-1,3-dioxolane).

 D. 2-(1,3-Dioxolan-2-yl)-4-[(2-n-butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]bromobenzene

[0436] Sodium hydride (60% dispersion in mineral oil, 9.65 g, 241 mmol, 2.5 eq) was added in portions over 15 minutes to a mixture of 2-n-butyl-1,3-diazaspiro[4.4]non-1-en-4-one hydrochloride (18.7 g, 96.5 mmol, 1.0 eq) in DMF (400 mL) at 0° C. The mixture was stirred and allowed to warm to room temperature over 15 minutes. To this mixture was added via canula a solution of 5C (31.1 g, 96.5 mmol, 1.0 eq) in DMF (100 mL). After 14 hours, the mixture was concentrated in vacuo and partitioned between ethyl acetate (500 mL) and 10% aqueous sodium dihydrogen phosphate (300 mL). The organic layer was dried over sodium sulfate and concentrated to give crude 5D as an orange oil (42.7 g), which was used without further purification.

E. 4-[(2-n-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2-formyl-bromobenzene

 A solution of crude 5D (6.0 g, approximately 13.6 mmol, 1.0 eq) in THF (180 mL) and 1N hydrochloric acid (30 mL) was heated at 65° C. for 1.5 hours. The mixture was cooled and then treated with saturated aqueous sodium carbonate solution (75 mL) and ethyl acetate (200 mL). The organic layer was removed and dried over sodium sulfate, concentrated, and then further dried azeotropically with toluene to give 5E as a crude yellow oil (8.2 g) which contained a small amount of toluene. This material was used without further purification.

F. 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl)[1,1′-biphenyl]-2-sulfonamide

Palladium catalyzed Suzuki coupling of 5E and [2-[[(3,4-dimethyl-5-isoxazolyl)[(2-methoxyethoxy)methyl]amino]sulfonyl]phenyl]boronic acid was performed according to General Method 1 to yield 5F in 60% yield.

 G. 4′-[(2-Butyl-4-oxo-1,3-diazaspiro[4.4]non-1-en-3-yl)methyl]-2′-formyl-N-(3,4-dimethyl-5-isoxazolyl)-[1,1′-biphenyl]-2-sulfonamide

 Deprotection of 5F according to General Method 7 provided the title compound (5G=3F) in 73% yield: Rf=0.2 (silica gel using CH2Cl2/MeOH [100:5]).

PATENT

EP 1237888; WO 0144239

Example 3 4′-r(2-Butyl-4-oxo-1.3-diazaspiror4.41non-l-en-3-yl)methvn-2′-formyl-N-

(3, 4-dimethyl-5-isoxazolyl)-[ 1,1 ‘-biphenyl] -2-sulfonamide

Figure imgf000093_0001

A. 4′-Cvano-2>-(1.3-dioxolan-2-yl)-N-(3.4-dimethyl-5-isoxazolyl)-N-(2- methoxyethoxymethyl) [1.1 ‘-biphenyl] -2-sulfonamide

A mixture of 2B (1.28 g, 2.73 mmol), ethylene glycol (1.69 g, 27.3 mmol) and p-toluenesulfonic acid (38 mg) in toluene (30 mL) was heated at 130°C for 5 h, while a Dean-Stark water separator was used. After cooling, the mixture was diluted with EtOAc. The organic liquid was separated and washed with H2O and brine, dried and concentrated. The residue was chromatographed on silica gel using 5:4 hexane/EtOAc to afford 3A (1.1 g, 79%) as a colorless gum: R^0.57, silica gel, 1:2 hexane EtOAc.

B. 2,-(1.3-Dioxolan-2-yl)-4′-formyl-N-(3.4-dimethyl-5-isoxazolyl)-N-(2- methoxyethoxymethyl) [1 , l’-biphenyl] -2-sulfonamide To 3A (1.1 g, 2.14 mmol) in THF (21 mL) at 0°C was added DIBAL- H (IM in CH2C12, 4.28 mL 4.28 mmol) dropwise. The reaction was stirred at RT overnight. MeOH (20 mL) was added and the reaction was stirred for 5 min. The mixture was poured into cold 0.1 N HCI solution (150 mL), shaken for 5 min, and then extracted with 3:1 EtOAc/hexane. The combined organic extracts were washed with H2O and brine, dried and concentrated. The residue was chromatographed on silica gel using 3:4 hexane/EtOAc to afford 3B (710 mg, 64%) as a colorless gum: R^O.45, silica gel, 2:3 hexane/EtOAc. C. 2′-(1.3-Dioxolan-2-yl)-4′-hvdroxymethyl-N-(3.4-dimethyl-5- isoxazolyl)-N-(2-methoxyethoxymethyl) [1.1 ‘-biphenyl] -2- sulfonamide

3B (710 mg, 1.4 mmol) was subjected to sodium borohydride reduction according to General Method 11 to afford 3C, which was used for the next reaction step without further purification.

D. 4l-Bromomethyl-2,-(1.3-dioxolan-2-yl)-N-(3.4-dimethyl-5-isoxazolyl)- N-(2-methoxyethoxymethyl) [1 , l’-biphenyl] -2-sulfonamide 3C was treated with carbon tetrabromide and triphenylphosphine according to General Method 2. The crude residue was chromatographed on silica gel using 3:2 hexane/EtOAc to afford 3D (750 mg, 94%) as a colorless gum: R^0.74, silica gel, 1:2 hexane/EtOAc.

E. 4′-[(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methvn- 2,-(1.3- dioxolan-2-yl)-N-(3.4-dimethyl-5-isoxazolyl)-N-(2- methoxyethoxymethyl) [ 1. l’-biphenyll -2-sulfonamide 3D (750 mg, 1.3 mmol) was treated with 2-re-butyl-l,3- diazaspiro[4.4]non-l-en-4-one hydrochloride (387 mg, 1.68 mmol) according to General Method 4. The crude residue was chromatographed on silica gel using 100:1.7 CH2CL/MeOH to afford 3E as a gum (830 mg, 93%): R^O.40, silica gel, 100:5 CH2Cl2/MeOH.

F. 4′-r(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methyl1-2,– formyl-N-(3.4-dimethyl-5-isoxazolyl)-[l.l’-biphenyl1-2-sulfonamide

3E (830 mg, 1.20 mmol) was subjected to deprotection according to General Method 7. The crude residue was chromatographed on silica gel using 100:1.5 and then 100:4 CH2C12 /MeOH to afford the title compound as a gum (480 mg, 72%): R^O.16, silica gel, 100:5 CH.Cl MeOH.

Example 4

4′- . (2-Butyl-4-oxo- 1 ,3-diazaspiro [4.41 non-l-en-3-yl)methyll -N-C3.4- dimethyl-5-isoxazolyl)-2,-[(3.3-dimethyl-2-oxo-l- pyrrolidinvDmethyll [1.1 ‘-biphenyl] -2-sulfonamide

Figure imgf000095_0001

To 3F (110 mg, 0.20 mmol) in CH2C12 (4 mL) was added 4-amino- 2,2-dimethylbutanoic acid hydrochloride (98 mg, 0.59 mmol) [Scheinmann, et al., J. Chem. Research (S), 414-415 (1993)] and 3A molecular sieves, followed by glacial acetic acid (35 mg, 0.59 mmol) and then sodium acetate (48 mg, 0.59 mmol). The mixture was stirred for 8 minutes, and NaB(AcO)3H (124 mg, 0.59 mmol) was then added. The reaction mixture was stirred at RT for 2 h, diluted with EtOAc and filtered through celite. The filtrate was washed with H2O and brine, dried and concentrated. This material was dissolved in CH2C12 (6 mL) and 1,3-diisopropylcarbodiimide (32 mg, 0.25 mmol) was added. The reaction mixture was stirred at RT for 2 h and diluted with CH2C12, washed with H2O and brine, dried and concentrated. The residue was purified by preparative HPLC to provide the title compound as a white solid (40 mg, 31%, for two steps): mp 104- 110°C. Analysis calculated for C36H45N5O5S • 0.8 H2O: Calc’d: C, 64.13; H, 6.97; N, 10.39; S, 4,75. Found: C, 64.18; H, 6.60; N, 10.23; S, 4.50.

Example 5

4′-[(2-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methyl1-2,-formyl-N-

(3,4-dimethyl-5-isoxazolyl)-[l,l’-biphenyl]-2-sulfonamide (Alternative

Preparation for 3F)

A. 2-[(2′-Bromo-5′-formyl)phenyl)1-1.3-dioxolane

DIBAL-H (1.0 M solution in toluene, 445 mL, 445 mmol, 1.1 eq) was added over 30 minutes to a solution of 2-[(2′-bromo-5′-cyano)phenyl)]-l,3- dioxolane (103 g, 404 mmol, 1.0 eq) [Zhang, H.-Y. et al., Tetrahedron, 50, 11339-11362 (1994)] in toluene (2.0 L) at -78 °C. The solution was allowed to warm to 0 °C. After 1 hour, a solution of Rochelle’s salt (125 g) in water (200 mL) was added, and the mixture was allowed to warm to room temperature and was stirred vigorously for 16 h. The organic layer was concentrated and the residue partitioned between ethyl acetate (1 L) and 1 N hydrochloric acid (800 mL). The organic layer was washed with saturated aqueous sodium bicarbonate (800 mL), dried over sodium sulfate, and then concentrated to give 70.5 g of crude 5A as a yellow solid, which was used without further purification.

B. 2-[(2′-Bromo-5′-hvdroxymethyl)phenyl)l-1.3-dioxolane

Sodium borohydride (3.66 g, 96.7 mmol, 0.5 eq) was added to a solution of crude 5A (49.7 g, approximately 193 mmol, 1.0 eq) in absolute ethanol (1300 mL) at 0 °C. After 2 hours, a solution of 10% aqueous sodium dihydrogen phosphate (50 mL) was added and the mixture was stirred and allowed to warm to room temperature. The mixture was concentrated, then partitioned between ethyl acetate (800 mL) and saturated aqueous sodium bicarbonate (500 mL). The organic layer was dried over sodium sulfate and concentrated to give 49.0 g of crude 5B as a yellow oil, which was used without further purification. C. 2-[(2′-Bromo-5′-bromomethyl)phenyl)]-l,3-dioxolane Triphenylphosphine (52.7 g, 199 mmol, 1.05 eq) was added in portions over 15 minutes to a solution of crude 5B (49.0 g, approximately 189 mmol, 1.0 eq) and carbon tetrabromide (69.0 g, 208 mmol, 1.1 eq) in THF at 0 °C. After 2 hours, saturated aqueous sodium bicarbonate solution (20 mL) was added, and the mixture was allowed to warm to room temperature and was then concentrated. Ether (500 mL) was added, and the resulting mixture was filtered. The filtrate was dried over magnesium sulfate and concentrated. The residue was chromatographed on silica gel (8:1 hexanes/ethyl acetate as eluant) to give 5C as a white solid (31.1 g, 51% yield from 2-[(2′-bromo-5′-cyano)phenyl)]-l,3-dioxolane).

D. 2-( 1 ,3-Dioxolan-2-yl)-4- [ (2-re-butyl-4-oxo- 1 ,3-diazaspiro [4.4] non- 1- en-3-yl)methyl] bromobenzene Sodium hydride (60% dispersion in mineral oil, 9.65 g, 241 mmol,

2.5 eq) was added in portions over 15 minutes to a mixture of 2-rc-butyl- l,3-diazaspiro[4.4]non-l-en-4-one hydrochloride (18.7 g, 96.5 mmol, 1.0 eq) in DMF (400 mL) at 0°C. The mixture was stirred and allowed to warm to room temperature over 15 minutes. To this mixture was added via canula a solution of 5C (31.1 g, 96.5 mmol, 1.0 eq) in DMF (100 mL). After 14 hours, the mixture was concentrated in vacuo and partitioned between ethyl acetate (500 mL) and 10% aqueous sodium dihydrogen phosphate (300 mL). The organic layer was dried over sodium sulfate and concentrated to give crude 5D as an orange oil (42.7 g), which was used without further purification.

E. 4-[(2-n-Butyl-4-oxo-1.3-diazaspiro[4.41non-l-en-3-yl)methyl1-2- formyl-bromobenzene

A solution of crude 5D (6.0 g, approximately 13.6 mmol, 1.0 eq) in THF (180 mL) and IN hydrochloric acid (30 mL) was heated at 65°C for 1.5 hours. The mixture was cooled and then treated with saturated aqueous sodium carbonate solution (75 mL) and ethyl acetate (200 mL). The organic layer was removed and dried over sodium sulfate, concentrated, and then further dried azeotropically with toluene to give 5E as a crude yellow oil (8.2 g) which contained a small amount of toluene. This material was used without further purification.

F. 4′-.(2-Butyl-4-oxo-1.3-diazaspiro■4.41non-l-en-3-yl)methyl1-2,– formyl-N-(3,4-dimethyl-5-isoxazolyl)-N-(2-methoxyethoxymethyl) f 1.1 ‘-biphenyl] -2-sulfonamide Palladium catalyzed Suzuki coupling of 5E and [2-[[(3,4-dimethyl-5- isoxazolyl) [(2-methoxyethoxy)methyl] amino] sulfonyl] phenyl]boronic acid was performed according to General Method 1 to yield 5F in 60% yield.

G. 4’-[ 2-Butyl-4-oxo-1.3-diazaspiro[4■41non-l-en-3-yl)methvn-2,– formyl-N-(3 ,4-dimethyl-5-isoxazolyl)- fi .1 ‘-biphenyl] -2-sulfonamide

Deprotection of 5F according to General Method 7 provided the title compound (5G = 3F) in 73% yield: R^0.2 (silica gel using CH2ClJ eOH [100:5]).

Patent Submitted Granted
Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists [US6638937] 2002-10-03 2003-10-28
Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists [US6835741] 2004-06-03 2004-12-28
Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists [US6852745] 2004-07-01 2005-02-08

///////////BMS-248360, Preclinical, SARTAN, BMS, HYPERTENTION

CCCCC1=NC2(CCCC2)C(=O)N1CC3=CC(=C(C=C3)C4=CC=CC=C4S(=O)(=O)NC5=C(C(=NO5)C)C)CN6CCC(C6=O)(C)C


Filed under: Preclinical drugs Tagged: BMS-248360, HYPERTENTION, preclinical, SARTAN

Ravidasvir, PPI-668, BI 238630

$
0
0

CAS # 1303533-81-4, Ravidasvir dihydrochloride

Ravidasvir dihydrochloride

C42H50N8O6.2(HCl), 835.83

CAS 1303533-81-4

Phase II/IIIHepatitis C

Ravidasvir
PPI-668 free base; BI 238630;
CAS:1242087-93-9

C42H50N8O6, 762.38
Chemical Name:methyl N-[(1S)-1-({(2S)-2-[5-(6-{2-[(2S)-1-{(2S)-2-[(methoxycarbonyl)amino]- 3- methylbutanoyl}pyrrolidin-2-yl]-1H-imidazol-4-yl}naphthalen-2-yl) -1H- benzimidazol- 2-yl]pyrrolidin-1-yl}carbonyl)-2-methylpropyl]carbamate
Mechanism of Action:NS5A Inhibitor
Indication: hepatitis C
Development Stage: Phase II
Developer:Presidio Pharmaceuticals, Inc

  • OriginatorXTL Biopharmaceuticals
  • Developer Pharco Corporation; Presidio Pharmaceuticals
  • Class Antivirals; Benzimidazoles; Carbamates; Naphthalenes; Pyrrolidines; Small molecules
  • Mechanism of Action Hepatitis C virus NS 5 protein inhibitors; Hepatitis C virus replication inhibitors
  • 31 Aug 2015 Ascletis plans to initiate the phase II EVEREST trial for Hepatitis C (Combination therapy; Treatment-naive) in Taiwan
  • 31 Aug 2015 Taiwan Food and Drug Administration approves Clinical Trial Application to initiate a phase II trial for interferon free regimen comprising danoprevir and ravidasvir in Hepatitis C
  • 24 Jun 2015 Efficacy data from a phase IIa trial in Hepatitis C released by Ascletis

r12

Ravidasvir [Methyl N-[(1S)-1-({(2S)-2-[5-(6-{2-[(2S)-1-{(2S)-2-[(methoxycarbonyl)amino]- 3- methylbutanoyl}pyrrolidin-2-yl]-1H-imidazol-4-yl}naphthalen-2-yl) -1H- benzimidazol- 2-yl]pyrrolidin-1-yl}carbonyl)-2-methylpropyl]carbamate] is an Nonstructural protein 5A (NS5A) inhibitor. It is an antiviral agent that is being developed as a potential treatment for hepatitis C virus infection.

PPI-668, a non-structural 5A (NS5A) protein of hepatitis C virus (HCV) inhibitor, is in phase II clinical studies at Presidio Pharmaceuticals for the treatment of chronic genotype 1 hepatitis C virus infection.

Ravidasvir has 50% inhibitory concentrations (EC50s) values of 0.02-1.3 nM in replicon assays for HCV genotypes 1-7 (gt1-gt7).

Ravidasvir was developed by Presidio Pharmaceuticals Inc, later Ascletis licensed it. Ravidasvir is in Phase II clinical trials proving interferon (IFN)-free regimen to treat chronic hepatitis C (CHC). Ascletis is now the first Chinese company to file clinical trial applications in China for an IFN-free regimen.
In 2014, Ascletis acquired rights for development and commercialization in Greater China and Pharco in Egypt for the treatment of hepatitis C.

Hepatitis C virus infection is a major health problem worldwide and no vaccine has yet been developed against this virus. The standard therapy of pegylated-interferon and ribavirin induces serious side effects and provides viral eradication in less than 50% of patients. Combination therapy of HCV including ribavirin and interferon are currently is the approved therapy for HCV. Unfortunately, such combination therapy also produces side effects and is often poorly tolerated, resulting in major clinical challenges in a significant proportion of patients. The combination of direct acting agents can also result in drug-drug interactions. To date, no HCV therapy has been approved which is interferon free. There is therefore a need for new combination therapies which have reduced side effects, and interferon free, have a reduced emergence of resistance, reduced treatment periods and/or and enhanced cure rates.

Nonstructural protein 5A (NS5A) is a zinc-binding and proline-rich hydrophilic phosphoprotein that plays a key role in Hepatitis C virus RNA replication.

A number of direct-acting antiviral agents (DAAs) are under development for the treatment of chronic HCV infection. These agents block viral production by directly inhibiting one of several steps of the HCV lifecycle. several viral proteins involved in the HCV lifecycle, such as the non-structural (NS)3/4A serine protease, the NS5B RNA-dependent RNA polymerase (RdRp), and the NS5A protein, have been targeted for drug development. Two NS3/4A protease inhibitors already approved for clinical use, numerous other protease inhibitors are being developed as well as inhibitors of viral replication, including nucleoside/nucleotide analogue inhibitors of HCV RdRp, non-nucleoside inhibitors of RdRp, cyclophilin inhibitors, and NS5A inhibitors.

Inhibition of NS5A at picomolar concentrations has been associated with significant reductions in HCV RNA levels in cell culture-based models, which makes these agents among the most potent antiviral molecules yet developed.


Activity:

This NS5A inhibitor has been shown to possess high efficacy against HCV genotype 1, with up to 3.7 log10 mean HCV RNA reductions, in a Phase Ib clinical trial. Activity was demonstrated against variants harbouring the L31M substitution. In an added genotype-2/3 cohort, the first 2 patients achieved mean 3.0 log10 RNA level reductions [1].

Results from the Phase IIa study involving a combination therapy with Faldaprevir and Deleobuvir plus Ravidasvir came with positive news where the said combination cured 92 percent of those with genotype 1a of hepatitis C virus (HCV) when given with ribavirin.  The results presented at the 49th annual meeting of the European Association for the Study of the Liver (EASL) in London [2, 3].

The 36 study participants were randomly dived into three even cohorts of 12 each: The first received 600 mg of Deleobuvir twice a day as well as once-daily doses of Faldaprevir (120 mg), Ravidasvir and Ribavirin. The second group received the same regimen except the Faldaprevir dose was 400 mg. The third group took the regimen with the higher dose of Faldaprevir, but without Ribavirin. All participants were treated for 12 weeks with follow up for next 24 weeks.

Ninety-two percent of the first and second cohorts (11 out of 12 in both cases) achieved a sustained virologic response 12 weeks after completing therapy (SVR12, considered a cure). In the end, 14 participants were required for the third cohort, because one was incarcerated early on during treatment and another experienced viral rebound at week eight as a result of not adhering to the treatment regimen. Of the other 12 participants, eight, or two-thirds, have achieved an SVR12, while one more participant stopped taking the therapy at week eight but has since achieved an SVR8.

PATENT

WO 2011054834

http://www.google.co.in/patents/WO2011054834A1?cl=en

Scheme 1

Figure imgf000018_0001

GOING TO PRODUCT USING STRUCTURES FROM PATENT

Figure imgf000031_0002 IIa

Figure imgf000032_0001  IIIa   one of side chain

DO NOT MISS OUT synthesis of XIIIa or XIII’a, this is needed in one of side chain

Figure imgf000034_0004L-boc-prolinol

Figure imgf000035_0001Z-boc-prolinal

Figure imgf000035_0002XXIV

Figure imgf000036_0001XIIIa

or

Figure imgf000036_0002

Figure imgf000038_0002XVIb

Figure imgf000043_0001

Figure imgf000045_0001

MY CONSTRUCTION of 3

R1

Figure imgf000052_0001

Compound 3 was prepared following the procedure reported for the synthesis of compound 1 using intermediate XVIIIb instead of intermediate XVIIIa. see my construction below

R1

Compound 3. BASE

1H NMR (400 MHz, DMSO-d6) δ ppm 8.34 (2 H, s), 8.21 (1 H, s), 8.19

(1 H, d, J=8.69 Hz), 8.06 – 8.11 (2 H, m), 8.00 (1 H, dd, J=8.88, 1.61 Hz), 7.88 – 7.96

(2 H, m), 7.86 (1 H, d, J=8.48 Hz), 7.32 (1 H, d, J=8.48 Hz), 7.34 (1 H, d, J=8.53 Hz), 5.27 (1 H, dd, J=8.17, 5.33 Hz), 5.17 (1 H, t, J=7.00 Hz), 4.15 (2 H, t, J=7.95 Hz), 3.84

– 3.96 (4 H, m), 3.56 (6 H, s), 2.38 – 2.47 (2 H, m), 1.95 – 2.30 (8 H, m), 0.86 (3 H, d,

J=6.70 Hz), 0.85 (3 H, d, J=6.70 Hz), 0.81 (6 H, d, J=6.63 Hz).

[a] 2°= -148.98 0 (c 0.3336 w/v %, MeOH)

Alternative preparation of compound 3 and the corresponding HC1 salt

Figure imgf000052_0001

N-methoxycarbonyl-L- Valine (3.09 g, 17.7 mmol, 2.1 equiv) was dissolved in dichloro- methane (300 mL). Triethylamine (11.7 mL, 84.1 mmol, 10 equiv) and (l-cyano-2- ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino-carbenium hexafluoro- phosphate were added (7.57 g, 17.7 mmol, 2.1 eq). The reaction mixture was stirred at room temperature for 5 minutes, after which XVIIIb was added (5 g, 8.41 mmol in case x.HCl equals 4 HC1). Stirring was continued for 30 minutes. HC1 in iPrOH (6N) was added to the mixture (until pH = 2), and the resulting mixture was stirred for 5 minutes. The solution was then washed with saturated aqueous sodium carbonate (2 x 200 mL) and once with brine (200 mL). The organic layer was separated, dried on magnesium sulphate and filtrated. After removal of the solvent in vacuum, the obtained residue was further dried in vacuum to afford an orange powder (6.84 g)

The powder was purified by silica gel column chromatography using gradient elution with 0 to 10 % MeOH (7N NH3) in dichloromethane, resulting in compound 3 (2.81 g) as a foam.

Compound 3 was dissolved in iPrOH (40 mL) and HC1 (6N in iPrOH, 10 mL) was added. The volatiles were removed in vacuum. Then, iPrOH (30 mL) was added and the mixture was heated at reflux. The solution was cooled to room temperature and stirred at room temperature for 4 days. tBuOMe (100 mL) was added to the solution, resulting in white precipitation, which was filtered, washed immediately with tBuOMe (3 x 10 mL) under nitrogen atmosphere and dried under vacuum at 40°C. The residue was mixed with acetonitrile and evaporated to dryness (2x). The residue was stirred in acetonitrile (150 mL) and the mixture was sonicated for 10 minutes. The precipitate was filtered under nitrogen atmosphere, washed twice with acetonitrile (50 mL) and dried in vacuum at 40°C, resulting in a slightly yellow powder (4 g).

HCL salt of compound 3:

[a] *° = -110.02 ° (589 nm, 20 °C, c 0.429 w/v%, MeOH)

1H NMR (600 MHz, DIMETHYLFORMAMIDE- y, 280K) δ ppm 0.86 (d, J=6.6 Hz, 6 H), 0.95 (d, J=7.0 Hz, 6 H), 2.03 – 2.20 (m, 2 H), 2.26 – 2.37 (m, 3 H), 2.39 – 2.61 (m, 5 H), 3.61 – 3.63 (m, 6 H), 3.93 – 4.01 (m, 2 H), 4.23 – 4.32 (m, 2 H), 4.32 – 4.39 (m, 2 H), 5.49 (t, J=7.5 Hz, 1 H), 5.52 (dd, J=8.3, 5.3 Hz, 1 H), 7.22 (d, J=8.8 Hz, 1 H), 7.27 (d, J=8.8 Hz, 1 H), 7.98 (d, J=8.6 Hz, 1 H), 8.01 (dd, J=8.6, 1.1 Hz, 1 H), 8.03 (dd, J=8.8, 1.8 Hz, 1 H), 8.09 (d, J=8.8 Hz, 1 H), 8.19 (d, J=8.8 Hz, 1 H), 8.22 (dd, J=8.4, 1.8 Hz, 1 H), 8.25 (s, 1 H), 8.32 (s, 1 H), 8.41 (s, 1 H), 8.88 (s, 1 H).

Anal. Calcd for C42H5oN806 . 2 HCl . 4 H20: C 55.56, H 6.66 , N 12.34. Found: C 55.00, H 6.60, N 12.30

Going reverse…………………..

Intermediate XVIIIb

2.8 preparation of intermediate XVIIIb (A=

Figure imgf000044_0002

To a solution of XVIIb (960 mg, 1.48 mmol) in CH2C12 (25mL) was added HCI (5-6 M in isopropanol, 5 mL). The mixture was stirred at room temperature overnight. The solvent was evaporated, the obtained solid was dried in vacuum and used as such in the next step. 2.8a Alternative preparation of intermediate XVIIIb (A=

Figure imgf000045_0001

XVIIb (19.52 g, 30.1 mmol, 1.00 equiv.) was dissolved in dichloromethane (200 mL) and HCI in isopropanol (5-6 N, 300 mL) was added. The reaction mixture was stirred for 1 hour at room temperature. tBuOMe (1000 mL) was added to the suspension and the slurry was stirred at roomtemperature for 30 minutes. The filtered solid was rinced with tBuOMe (2x 100 mL) and dried under vacuum overnight to afford XVIIIb as a powder (15.2 g). 1H NMR (400 MHz, MeOD-d4) δ ppm 2.15 – 2.37 (m, 2 H), 2.37 – 2.52 (m, 2 H), 2.52 – 2.69 (m, 2 H), 2.69 – 2.88 (m, 2 H), 3.56 – 3.71 (m, 4 H), 5.19 – 5.41 (m, 2 H), 7.90 – 8.02 (m, 3 H), 8.05 (dd, J= 8.6, 1.6 Hz, 1 H), 8.10 – 8.25 (m, 4 H), 8.30 (d, J=1.4 Hz, 1 H), 8.47 (d, J=1.2 Hz, 1 H)

INTERMEDIATE XVIIb

2.7 reparation of intermediate XVIIb (A= PG= Boc)

Figure imgf000043_0001

To boronic ester XVIb (1.22 g, 2.26 mmol), bromide Xllla (1072 mg, 3.39 mmol), sodium bicarbonate (380 mg, 4.52 mmol), Pd(dppf)Cl2 (166 mg, 0.226 mmol) in toluene (50 mL), was added water (1 mL). The resulting mixture was heated at reflux overnight. The reaction mixture was filtered, evaporated to dryness and purified by column chromatography by gradient elution with heptane to ethyl acetate. The collected fractions containing the product were pooled and the volatiles were removed under reduced pressure. The residue (960 mg, 65 %) was used as such in the next reaction.

2.7a Alternative preparation of intermediate XVIIb (A= . PG= Boc)

Figure imgf000043_0002

XVIb (10 g, 18.5 mmol), Xlll’a (8.76 g, 24 mmol), NaHC03 (9.32 g, 111 mmol) and Pd(dppf)Cl2 (lg) were stirred in dioxane/water (140 mL, 6/1) under argon. The mixture was heated to 85 °C for 15 hours. Brine (100 mL ) was added and the mixture was extracted with CH2CI2, after drying on MgSC^, filtration and evaporation of the solvent, the residue was purified by column chromotography by gradient elution with CH2CI2 to EtOAc to afford XVIIb (7 g, 58 %).

Figure imgf000044_0001

To a stirred, deoxygenated solution of Vlllb (20.0 g, 45.2 mmol, 1.00 equiv.), Ilia (20.6 g, 49.7 mmol, 1.1 equiv.) and sodium bicarbonate (11.4 g, 136 mmol, 3.0 equiv.) in 1 ,4-dioxane/water (500 mL, 5: 1) under nitrogen, was added l.,.r-Bis(diphenyi~ phosphmo)ferrocene-paiIadium(]I)dichloride dichJoromethane complex (2.50 g, 4.52 mmol, 0.1 equiv.). The mixture was heated at 80°C under argon for 15 hours and cooled to room temperature. The reaction mixture was diluted with dichloromethane (500 mL) and washed with brine (2 x 150 mL) dried on magnesium sulphate; filtered and evaporated to dryness to afford a dark brown foam (43 g). The foam was purified using silicagel column chromatography (gradient elution with 0-6% MeOH in CH2CI2) to afford XVIIb (19.52 g, 65%) as an off-white powder.

INTERMEDIATE XVIb

Figure imgf000038_0001

Bromide XVb (1890 mg, 3.83 mmol), 4,4,4\4\5,5,5\5*-octamethyl-2,2′-bis(l,3,2- dioxaborolane) (2437 mg, 9.59 mmol), KF (390 mg; 6.71 mmol) and (dppf)PdCl2 (281 mg, 0.384 mmol) were dissolved in toluene (50 mL) and heated 3 days at reflux.

The solids were removed by filtration over dicalite and the filtrate was evaporated to dryness on silica. The residue was purified by column chromatography using a heptane to ethylacetate gradient. The fractions containing the product were pooled and the solvent was removed under reduced pressure. The residue (1.22 g, 59 %) was used as such in the next reaction

Figure imgf000038_0002

Under nitrogen, Ilia (25 g, 60.5 mmol), 6-bromonaphthalen-2-yl trifluoromethane- sulfonate (20 g, 56.7 mmol), K3P04 (36.65 g, 173 mmol) and (PPh3)4Pd (717 mg, 0.62 mmol) were stirred in THF (60 mL) and water (15 mL) with the heating mantle at 85 °C (reflux) for 2 hours. CH2CI2 (50 mL) was added and the water layer was separated. The organic layer was dried on MgS04 and after filtration, the filtrate was concentrated resulting in a sticky solid. The residue was purified by column

chromatography (petroleum ether/Ethyl acetate 15/1 to 1/1) to afford XVb (20 g;

40.6 mmol). Compound XVb (1 g, 2.0 mmol), potassium acetate (0.5 g, 5.0 mmol), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bis(l,3,2-dioxaborolane) (1.29 g, 5.0 mmol), and Pd(dppf)Cl2 (0. lg) were stirred in DMF (15 mL) under argon. The mixture was heated at 60°C for 5 hours. After cooling, CH2CI2 (50 mL) was added and the mixture was washed with saturated NaHC03. The water layer was separated and extracted with CH2CI2. The organic layers were combined and dried on MgSC^. After filtration the solvent was removed and the product was purified by column chromatography (gradient elution with petroleum ether/ethyl acetate 10/1 to 1/1) to give of XVIb (0.7 g,1.3 mmol, 65 %) as light yellow solid.

INTERMEDIATE XVb

Figure imgf000037_0001

2,6-Dibromonaphthalene (6.92 g, 24.2 mmol), boronic ester Ilia (2 g, 4.84 mmol), NaHC03 (813 mg, 9.68 mmol), (dppf)PdCl2(710 mg, 0.968 mmol) were dissolved in toluene (75 mL). Water (1 mL) was added and the mixture was heated for 7 hours at reflux. The solids were removed by filtration over dicalite and the filtrate was evaporated to dryness on silica. The residue was purified by column chromatography by gradient elution with heptane to ethylacetate. The appropriate fractions were pooled and the solvent was removed under reduced pressure. The residue (1.89 g, 79 %) was used as such in the next step.

1.2 Preparation of intermediate IIIa (PG= Boc)

Figure imgf000032_0001 IIIa

To a mixture of Ila (200 g, 546 mmol), potassium acetate (160.8 g, 1.64 mol) and 4,4,4*,4*,5,5,5*,5*-octamethyl-2,2,-bis(l,3,2-dioxaborolane) (416 g, 1.64 mol) in DMF (3L) was added Pd(dppf)Cl2 (20 g) under nitrogen gas. The reaction mixture was stirred at 85°C for 15 hours. The mixture was diluted with ethyl acetate, washed with water and brine, dried over magnesium sulfate, the solids removed by filtration, and the solvents of the filtrate were removed under reduced pressure. The residue was purified by silica column chromatography (petroleum ether : ethyl acetate 10: 1 to 2: 1) to afford 125 g of Ilia as a white solid (contains 15% of boronic acid).

INT IIa

1.1 preparation of intermediate Ila (PG= Boc; X= Br)

Figure imgf000031_0002

Ma

To a solution of Boc-Z-Proline (2669 mg, 12.4 mmol) in pyridine/DMF (30 mL, 1/1) was added di(lH-imidazol-l-yl)ketone (2205 mg, 13.6 mmol). The mixture was stirred at 45°C for 2 hours. 4-bromobenzene-l,2-diamine (2319 mg, 12.4 mmol) was added and the mixture was stirred at ambient temperature overnight. The solvent was removed and the residue heated in acetic acid (15 mL) at 100°C for 30 minutes. After

concentration of the residue, the mixture was partitioned between ethyl acetate and a saturated sodium bicarbonate solution. The organic phase was separated and washed with water, after drying over Na2SC”4, the mixture was filtrated and the filtrate was concentrated in vacuum. The obtained residue was purified by flash chromatography using CH2Cl2/EtOAc 90/10 to 50/50, resulting in compound Ila (3.146 g, 69 %).

DO NOT MISS OUT synthesis of XIIIa or XIII’a, this is needed in one of side chain

2.1 preparation of L-boc-prolinol

Figure imgf000034_0004

Borane-methyl sulfide complex (180 mL, 1.80 mol) was added dropwise to a solution of N-Boc- L-Proline (300 g, 1.39 mol) in anhydrous THF (3.0 L) which was cooled to 0°C. When gas evolution ceased, the ice bath was removed and the solution was stirred at 10°C for 18 hours. Thin layer chromatography (TLC) showed that no starting material remained and that the desired product was formed. The solution was cooled to 0°C and methanol (2.4 L) was slowly added. The solvents were removed under reduced pressure. The residue was reconstituted in dichloromethane (1 L), washed with

NaHC03 (500 mL, saturated, aqueous) and brine (500 mL), dried over MgS04, the solids were removed via filtration, and the solvents of the filtrate were removed under reduced pressure to afford a white solid, 260 g (93%), used in the next step without further purification.

2.2 preparation of Z-boc-prolinal

Figure imgf000035_0001

To a solution of Z-boc-prolinol (100 g, 500 mmol) in CH2CI2 (1.5 L) at 0°C were added successively, under vigorous stirring, 2,2,6,6-tetramethylpiperidine-l-oxyl (TEMPO; 1.56 g, 10 mmol) and NaBr (5.14 g, 50 mmol). To the resulting mixture was added dropwise a solution of NaHC03 (6.3 g, 75 mmol) and 6% NaCIO in active chlorine (750 mL, 750 mmol) at 0°C over a period of 1 hour. TLC showed no starting material remained and that the desired product was formed. The mixture was rapidly extracted with dichloromethane (2 x 1.5 L). The organic layers were combined, washed with NaHS04 (10%, 1 L) and KI (4%, 200 mL), then with Na2S203 (10%, 1 L) and brine (1.5 L), dried over MgS04, the solids were removed via filtration, and the solvents evaporated to afford a yellow oil, Z-boc-prolinal, (89 g, 92%>), used in the next step without further purification.

2.3 preparation of intermediate XXIV

Figure imgf000035_0002

ammonia

XXIV

Aqueous ammonia (25~28%>, 200 mL) was added dropwise to a solution of L-boc- prolinal (89 g, 0.44 mol) and glyoxal (183 mL of 40% in water) in methanol (1 L). The reaction mixture was sealed and reacted at 10°C. After 16 hours, additional glyoxal (20 mL) and aqueous ammonia (20 mL) were added and reacted for an additional 6 hours. The solvents were removed under reduced pressure, and the crude was reconstituted in ethyl acetate (1.0 L), washed with water and brine, dried over MgSC^, the solids were removed via filtration and the solvents were removed under reduced pressure. The crude was purified by column chromatography (silica gel, dichloromethane to methanol/dichloromethane 1 :70) to obtain 73 g (70%) intermediate XXIV as a white solid.

1H NMR: (CD3OD 400 MHz) δ 6.95 (s, 2H), 4.82-4.94 (m, 1H), 3.60-3.70 (m, 1H), 3.41-3.50 (m, 1H), 2.20-2.39 (m, 1H), 1.91-2.03 (m, 3H), 1.47 (s, 3H), 1.25 (s, 6H)

2.4 preparation of intermediate XHIa (PG= Boc)

Figure imgf000036_0001

XXIV Xllla

N-Bromosuccinimide (47.2 g, 0.26 mol) was added portion wise over 1 hour to a cooled (ice-ethanol bath, -10 °C) solution of XXIV (63.0 g, 0.26 mol) in CH2C12 (1.5 L) and stirred at similar temperature for 2 hours. The reaction mixture was concentrated in vacuum and the residue was purification by preparatory HPLC to provide 25.3 g (30%) of Xllla as a pale yellow solid.

1H NMR: CD3OD 400Mhz

δ 6.99-7.03 (s,lH), 4.77-4.90 (m, 1H), 3.61-3.68 (m, 1H), 3.42-3.50 (m, 1H), 2.20-2.39 (m, 1H), 1.89-2.05 (m, 3H), 1.47 (s, 3H), 1.27 (s, 6H).

2.4a preparation of intermediate XHI’a (PG= Boc)

Figure imgf000036_0002

To a solution of iodine (43.3 g, 170.5 mmol, 2 eq) in chloroform (210 mL) in a round bottomed flask (1L) a suspension of XXIV (20 g, 84.3 mmol) in an aqueous NaOH solution (2M, 210 mL) was added. The mixture was stirred at room temperature for 15 hours. To the resulting reaction mixture was added a saturated aqueous Na2S2C”3 solution (100 mL) and the organic layer was separated. The aqueous layer was extracted with chloroform (4x 150 mL). The organic layers were combined, washed with water and dried on magnesium sulphate. The solids were filtered and the solution was evaporated to dryness to afford diiodide (38.61 g, 89 %).

The above obtained intermediate diiodide (2.24 g, 4.58 mmol) and sodium sulfite (4.82 g, 38 mmol) were placed in a round bottomed flask (100 mL) and suspended in 30% EtOH/water (80 mL). The resulting mixture was refluxed for 40 hours. The solvent was removed and after addition of H20 (20 mL), the mixture was stirred at room temperature overnight. The solids were filtered, washed with water and dried in a vacuum oven to afford compound XHI’a (1.024 g, 61 %).

1H NMR (400 MHz, DMSO-d6) δ ppm 1.16 and 1.38 (2x br. s., 9 H), 1.68 – 2.02 (m, 3 H), 2.02 – 2.27 (m, 1 H), 3.18 – 3.38 (m, 1 H), 3.38 – 3.59 (m, 1 H), 4.53 – 4.88 (m, 1 H), 6.81 (m, -0.1 H), 7.05 – 7.28 (m, -0.9 H), 11.90 – 12.20 (m, -0.9 H), 12.22 – 12.40 (m, -0.1 H)

PATENT

WO 2011149856

http://www.google.co.in/patents/WO2011149856A1?cl=en

1st scheme

Figure imgf000107_0001

IN ABOVE SCHEME CONVERSION OF f to g N-methoxycarbonyl-L-Val-OH is used,

USE R =H IN LAST STEP TO GET RAVIDASVIR

EXAMPLE 1 – Synthesis of compounds of Formula lie

Scheme 1-1 describes preparation of target molecules and their analogs with symmetrical and non-symmetrical functionalized ends.

[0341] Step a. To a solution of 2-bromonaphthane a (62.0 g, 300 mmol) in DCM (1 L) was added A1C13 (44.0 g, 330 mmol) and 2-chloroacetyl chloride (34.0 g, 330 mmol) at 0 °C. The reaction mixture was stirred at 0 °C for 1 h and then H20 added (500 mL) and extracted. The organic layer was washed with H20, dried over anhydrous Na2S04, evaporated under reduced pressure to give 80 g crude product, which was purified by re-crystallization from 10% EtOAc- hexane (v/v) to yield b (28 g, 36% yield) as a white solid: JH NMR (500 MHz, CDC13) δ 8.44 (s, 1H), 8.07 (s, 1H), 8.04 (d, J= 11.0 Hz, 1H), 7.84 (d, J= 8.5 Hz, 2H), 7.66 (d, J= 8.5 Hz, 1H), 4.81 (s, 2H) ppm; LCMS (ESI) m/z 282.9 (M + H)+.

Step b. To a solution of b (28.0 g, 100 mmol) in DCM (500 mL) was added N-Boc- L-Pro-OH (24.7 g, 115 mmol) and Et3N (70.0 mL, 500 mmol) and the mixture was stirred at rt for 2 h. The mixture was concentrated under reduced pressure to afford crude c which was used for the next step without further purification. LC-MS (ESI) m/z 462.1 (M + H)+.

Step c. To a solution of c (46.0 g, 100 mmol) in toluene (500 mL) was added

NH4OAc (77 g, 1.0 mol) and the mixture was stirred at 110 °C overnight, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (petroleum ether/EtOAc l :l(v/v)) to afford d (30 g, 68% yield) as a yellow solid: LC-MS (ESI) m/z 442 A (M + H)+.

Step d. To a solution of d (10.0 g, 23.0 mmol) in anhydrous DME (200 mL) and equal molar of boronate e was added PPh3 (1.2 g, 4.6 mmol), Pd(PPh3)4 (1.6 g, 2.3 mmol), and 2.0 M Na2C03 solution. The mixture was refluxed under argon overnight. The organic solvent was removed under reduced pressure and the residue was treated with H20, extracted with EtOAc (2 x 200 mL). The combined organic phase was dried, filtered, and concentrated in vacuo to give a residue, which was purified by silica gel column chromatography (petroleum

ether/EtOAc 3: l(v/v)) to afford f (10 g, 96% yield) as a yellow solid. LC-MS (ESI): m/z 709.3 (M+H)+.

Step e. To a stirred solution of f (150 mg, 0.29 mmol) in dioxane (3 mL) was added 4.0 N HCl in dioxane (3 mL) dropwise. The mixture was stirred at rt for 4 h, and then

concentrated to yield a yellowish solid (134 mg), which was used directly for the next step. The residue (134 mg, 0.290 mmol) was suspended in THF (5 mL) and DIPEA (0.32 mL) was added and followed by addition of N-methoxycarbonyl-L-Val-OH (151 mg, 0.860 mmol). After stirring for 15 min, HATU (328 mg, 0.860 mmol) was added and the mixture was stirred at rt for another 2 h and then concentrated. The residue was purified by prep-HPLC to obtain g (40 mg, 19% yield).

2nd scheme

Figure imgf000110_0001

SCHEME SIMILAR UPTO PENULTIMATE STEP

Note 9 is not final product pl ignore it

Step a. Referring to Scheme 1-2, to a solution of compound 3 (2.0 g, 4.5 mmol) in dioxane (25 mL) was added 4.0 N HCl in dioxane (25 mL). After stirring at rt for 4 h, the reaction mixture was concentrated and the residue was dried in vacuo to give a yellowish solid (2.1 g), which was used directly for the next step without further purification.

[0347] Step b. To the residue of step a (4.5mmol) was added DMF (25 mL), followed by adding HATU (2.1 g, 5.4 mmol), DIPEA (3.7 mL, 22.5 mmol) and N-methyl carbamate-L-valine (945 mg, 5.4 mmol). After stirring at rt for 15 min, the reaction mixture was added slowly to H20 (400 mL). A white solid precipitated was filtered and dried to give compound 6 (2.2 g, 98% yield). LC-MS (ESI): m/z 499.1 (M+H)+.

[0348] Step c. To a mixture of compound 6 (800 mg, 1.6 mmol), compound 7 (718 mg, 1.6 mmol), and NaHC03 (480 mg, 5.7 mmol) in 1 ,2-dimethoxyethane (15mL) and H20 (5mL) was added Pd(dppf)Cl2 (59 mg, 0.08 mmol). After stirring at 80°C overnight under an atmosphere of N2, the reaction mixture was concentrated. The residue was partitioned between 20%

methanol/CHCl3 (100 mL) and H20 (100 mL). The organic phase was separated and the aqueous phase was extracted with 20% methanol/CHCl3 (100 mL) again. The combined organic phase was consequently washed with brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (Petroleum

ether/EtOAc=15: l(v/v)) to give compound 8 (1.0 g, 85% yield) as a yellow solid. LC-MS (ESI): m/z 732.4 (M+H)+.

Step d. To a solution of compound 8 (200 mg, 0.27 mmol) in dioxane (3.0 mL) was added 4 N HCl in dioxane (3.0 mL). After stirring at rt for 2 h, the reaction mixture was concentrated and the residue was dried in vacuo to give an HCl salt in quantitative yield, which was used directly for the next step without further purification…………..CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

CAUTION SIMILAR BUT NOT SAME……..Step e. To a solution of the salt (0.27 mmol) in DMF (5.0 mL) was added DIPEA (0.47mL, 2.7 mmol), followed by adding N,N-dimethyl-D-phenyl glycine (59 mg, 0.33 mmol) and HATU (125 mg, 0.33 mmol). After stirring at rt for lh, the reaction mixture was partitioned between H20 and DCM. The organic phase was washed successively with H20 and brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by prep-HPLC to give compound 9……..CAUTION SIMILAR BUT NOT SAME. LC-MS (ESI): m/z 793.4 (M+H)+.

3rd scheme

Figure imgf000112_0001

SCHEME SIMILAR UPTO PENULTIMATE STEP

15 NOT THE COMPD PL IGNORE IT IF YOU NEED RAVIDASVIR

Step a. To a mixture of compound 3 (3.2 g, 7.2 mmol), bis(pinacolato)diboron (3.86 g, 15.2 mmol), and KOAc (1.85g, 18.8mmol) in 1,4-dioxane (100 mL) was added Pd(dppf)Cl2 (440 mg, 0.6 mmol). After stirring at 80 °C for 3 h under an atmosphere of N2, the reaction mixture was concentrated. The residue was purified with silica gel column chromatography (Petroleum ether/EtOAc=2/l(v/v)) to give compound 11 (2.8 g, 80% yield) as a white solid. LC- MS (ESI): m/z 490.3 (M+H)+.

[0352] Step b. To a mixture of compound 11 (626 mg, 1.27 mmol), compound 12 (570 mg, 1.27 mmol), and NaHC03 (420 mg, 4.99 mmol) in 1, 2-dimethoxyethane (30 mL) and H20 (10 mL) was added Pd(dppf)Cl2 (139 mg, 0.19 mmol). After stirring at 80°C overnight under an atmosphere of N2, the reaction mixture was concentrated. The residue was partitioned between 20% methanol/CHCl3 (100 mL) and H20 (100 mL). The aqueous phase was extracted with 20% methanol/CHCl3 (100 mL) again. The combined organic phase was consequently washed with brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc=2/l(v/v)) to give compound 13 (635 mg, 68% yield) as a yellow solid. LC-MS (ESI): m/z 732.4 (M+H)+.

Step c. To a solution of compound 13 (200 mg, 0.27 mmol) in dioxane (3.0 mL) was added 4 N HC1 in dioxane (3.0 mL). After stirring at rt for 2 h, the reaction mixture was concentrated and the residue was dried in vacuo to yield the HC1 salt of compound 14 in quantitative yield, which was used directly for the next step without further purification…..CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

CAUTION SIMILAR BUT NOT SAME………Step d. To a solution of the salt (0.27 mmol) in DMF (5.0 mL) was added DIPEA (0.47 mL, 2.7 mmol), followed by adding N,N-dimethyl-D-phenyl glycine (59 mg, 0.33 mmol) and HATU (125 mg, 0.33 mmol). After stirring at rt for lh, the reaction mixture was partitioned between H20 and DCM. The organic phase was consequently washed with H20 and brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by prep-HPLC to give compound 15..CAUTION SIMILAR BUT NOT SAME. LC-MS (ESI): m/z 793.4 (M+H)+.

4 th scheme

Figure imgf000114_0001

SCHEME SIMILAR UPTO PENULTIMATE STEP

5 NOT THE COMPD,  PL IGNORE IT IF YOU NEED RAVIDASVIR

4 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

scheme ……..CAUTION SIMILAR BUT NOT SAME

EXAMPLE 2 – Synthesis of compounds of Formula Hie

Step a. Referring to Scheme 2-1, to a mixture of compound 1 (5.05 g, 13.8 mmol), bis(pinacolato)diboron (7.1 g, 27.9 mmol), and KOAc (3.2 g, 32.5 mmol) in 1,4-dioxane (100 mL) was added Pd(dppf)Cl2 (400 mg, 0.5 mmol). After stirring at 80 °C for 3 h under an atmosphere of N2, the reaction mixture was concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc=2/l(v/v)) to give compound 2 (3.0 g, 53% yield) as a gray solid. LC-MS (ESI): m/z 414.2 (M+H)+.

Step b. To a mixture of compound 2 (522 mg, 1.26 mmol), compound 3 (500 mg, 1.13 mmol), and NaHC03 (333 mg, 3.96 mmol) in 1, 2-dimethoxyethane (30 mL) and H20 (10 mL) was added Pd(dppf)Cl2 (74 mg, 0.1 mmol). After stirring at 80°C overnight under an atmosphere of N2, the reaction mixture was concentrated. The residue was partitioned between 20% methanol/CHCl3 (100 mL) and H20 (100 mL). The organic phase was separated and the aqueous phase was extracted with 20% methanol/CHCl3 (100 mL) again. The combined organic phase was consequently washed with brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (DCM/MeOH=50:l (v/v)) to give compound 4 (450 mg, 55% yield) as a yellow solid. LC-MS (ESI): m/z 649.3 (M+H)+.

Step c. To a stirred solution of compound 4 (160 mg, 0.25 mmol) in dioxane (2.0 mL) was added 4N HCl in dioxane (2.0 mL). After stirring at rt for 3h, the reaction mixture was concentrated and the residue was dried in vacuo to give an HCl salt in quantitative yield, which was used directly for the next step without further purification.4 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

SCHEME SIMILAR UPTO PENULTIMATE STEP

5 NOT THE COMPD,  PL IGNORE IT IF YOU NEED RAVIDASVIR

scheme ……..CAUTION SIMILAR BUT NOT SAME

5 th scheme

Figure imgf000116_0001

SCHEME SIMILAR UPTO PENULTIMATE STEP

18NOT THE COMPD,  PL IGNORE IT IF YOU NEED RAVIDASVIR

17 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

scheme ……..CAUTION SIMILAR BUT NOT SAME

Step a. Referring to Scheme 2-2, to a mixture of compound 2 (1.16 g, 2.32 mmol), compound 6 (1.40 g, 3.39 mmol), and NaHC03 (823 mg, 9.8 mmol) in 1, 2-dimethoxyethane (30 mL) and H20 (10 mL) was added Pd(dppf)Cl2 (103 mg, 0.14 mmol). After stirring at 80 °C over night under an atmosphere of N2, the reaction mixture was concentrated. The residue was partitioned between 20% methanol/CHCl3 (150 mL) and H20 (150 mL). The aqueous phase was extracted with 20% methanol/CHCl3 (150 mL) again. The combined organic phase was consequently washed with brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/acetone=1.5/l (v/v)) to give compound 16 (1.32g, 80% yield) as a yellow solid. LC-MS (ESI): m/z 706.4 (M + H)+.

tep b. To a solution of compound 16 (200 mg, 0.28 mmol) in dioxane (3.0 mL) was added 4 N HC1 in dioxane (3.0 mL). After stirring at rt for 2 h, the reaction mixture was concentrated and the residue was dried in vacuo to give the HC1 salt of compound 17 in quantitative yield, which was used directly for the next step…….17 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

6 th scheme

 

Figure imgf000118_0001scheme 2-3

SCHEME SIMILAR UPTO PENULTIMATE STEP

22NOT THE COMPD,  PL IGNORE IT IF YOU NEED RAVIDASVIR

21 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

scheme ……..CAUTION SIMILAR BUT NOT SAME

Scheme 2-3

Step a. Referring to Scheme 2-3, to a solution of compound 1 (4.0 g, 10.9 mmol) in dioxane (40 mL) was added 4 N HC1 in dioxane (40 mL). After stirring at rt overnight, the reaction mixture was concentrated. The residue was washed with DCM, filtered, and dried in vacuo to afford a hydrochloride salt in quantitative yield, which was used for the next step without further purification.

Step b. To a solution of the salt (10.9 mmol) in DMF (30 mL) was added DIPEA (5.8 mL, 33.0 mmol), followed by adding N-methoxycarbonyl-L-valine (2.1 g, 12.1 mmol) and HATU (4.6 g, 12.1 mmol). After stirring at rt for lh, the reaction mixture was partitioned between H20 and DCM. The organic phase was consequently washed with H20 and brine, dried with anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (DCM/Petroleum ether=4/l (v/v)) to give compound 19 (3.0 g, 65% yield). LC- MS (ESI): m/z 423.1 (M+H)+.

Step c. To a mixture of compound 11 (800 mg, 1.9 mmol), compound 19 (700 mg, 1.7 mmol), and NaHC03 (561 mg, 6.6 mmol) in 1, 2-dimethoxyethane (60 mL) and H20 (20 mL) was added Pd(dppf)Cl2 (183 mg, 0.25 mmol). After stirring at 80 °C overnight under an atmosphere of N2, the reaction mixture was concentrated. The residue was then partitioned between 20% methanol/CHCl3 (100 mL) and H20 (100 mL). The aqueous phase was extracted with 20% methanol/CHCl3(100 mL) again. The combined organic phase was consequently washed with brine, dried with Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc=2/l(v/v)) to give compound 20 (600 mg, 52% yield) as a yellow solid. LC-MS (ESI): m/z 706.4 (M+H)+.

Step d. To a solution of compound 20 (200 mg, 0.28 mmol) in dioxane (3.0 mL) was added 4N HC1 in dioxane (3.0 mL). After stirring at rt for 2h, the reaction mixture was concentrated and the residue was dried in vacuo to yield the HC1 salt of compound 21 in quantitative yield, which was used directly for the next step without further purification.

21 CAN BE USED AS PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

7 th scheme

 

Figure imgf000148_0001

Scheme 6-2

SCHEME SIMILAR UPTO n-2 STEP in above scheme

84, 85 NOT THE COMPD,  PL IGNORE IT IF YOU NEED RAVIDASVIR

83 CAN BE USED AS early PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

scheme ……..CAUTION SIMILAR BUT NOT SAME

Step a. Referring to Scheme 6-2, a solution of compound 78 (50.0 g, 0.30 mol) in THF (500 mL) and H20 (500 mL) was added K2C03 (83 g, 0.60 mol) and (Boc)20 (73. Og, 0.330 mol). After stirring at rt overnight, the reaction mixture was concentrated and the residue was extracted with EtOAc (250 mL x 3). The extracts were combined, washed with brine, and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 78 (62 g), which was used for the next step without further purification. LC-MS (ESI) m/z 230.1 (M + H)+.

[0453] Step b. To a solution of compound 78 (60.0 g, 260 mmol) in EtOH (1 L) was slowly added NaBH4 (50.0 g, 1.30 mol) at rt. After stirring at rt overnight, the reaction was quenched by adding acetone (10 mL). The resulting mixture was concentrated and the residue was diluted with EtOAc (500 mL). The mixture was washed with brined and dried in vacuo. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 1/1 (v/v)) to give compound 79 (42.0 g, 80% yield) as a white solid. LC-MS (ESI) m/z 202 A (M + H)+.

[0454] Step c. To a solution of compound 79 (30.0 g, 150 mmol) and DMSO (35.0 g, 450 mmol) in DCM (1 L) was added oxalyl chloride (28.0 g, 220 mmol) at -78 °C. After stirring at – 78 °C for 4 h, the reaction mixture was added Et3N (60.0 g, 600 mol) and the resulting mixture was stirred for another 1 h at -78 °C. Subsequently, the reaction was quenched by adding H20. The organic layer was separated and the aqueous layer was extracted with DCM (200mL x 2). The extracts were combined, washed with brine, and dried with Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 80 (22.0 g) as a colorless oil, which was used immediately without further purification. LC-MS (ESI) m/z 200.1 (M + H)+.

[0455] Step d. A mixture of compound 80 (7.7 g, 38.5 mmol), 6-bromopyridine-2,3-diamine (8.0 g, 42.8 mmol) (PCT Intl. Appl. WO 2008021851) , and iodine (1.08 g, 4.28 mmol) in AcOH (30 mL) was stirred at rt overnight. The reaction mixture was neutralized by adding saturated aqueous NaHC03. The resulting mixture was extracted with EtOAc (200 mL x 3). The extracts were combined, washed with brine, and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography (DCM/MeOH = 80/1 (v/v)) to give compound 81 (7.8 g, 55% yield). LC-MS (ESI) m/z 367.1 (M + H)+.

[0456] Step e. A mixture of compound 82 (10.0 g, 20.1 mmol), bis(pinacolato)diboron (7.65 g, 30.1 mmol), potassium acetate (6.89 g, 70.3 mmol), and Pd(dppf)Cl2-CH2Cl2 (886 mg, 1.0 mmol) in 1,4-dioxane (200 mL) was stirred at 80 °C for 3 h under an atmosphere of N2. The reaction mixture was filtered through CELITE™ 545 and the filtered cake was washed with EtOAc (200 mL x 3). The filtrate was washed with brine and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography

(DCM/MeOH = 50/1 (v/v)) to give compound 83 (9.8 g, 89% yield) as a white solid: LC-MS (ESI) m/z 547.3 (M + H)+.83 CAN BE USED AS early PRECURSOR FOR RAVIDASVIR UPTO THIS POINT

PATENT

CN 102796084

http://www.google.com/patents/CN102796084A?cl=en

Step One: Formula (2) compounds strokes trichloride catalyst (AlCl3), chloroacetyl chloride (2-chloroacetylchloride) at room temperature to obtain a compound of formula (3),

Figure CN102796084AD00072

(3);

  wherein the reaction temperature is room temperature, the solvent is methylene chloride. Material I (i.e., formula (2) compound) and chloroacetyl chloride (2-chloroacetyl chloride) was slowly added, higher yields can be obtained. (3) The compound was recrystallized from ether to obtain.

  In the present embodiment, the 20.5 g of formula (2) compound (0. Imol) and 26.2 g AlCl3 (0.2mol) was added to 200ml of dichloromethane, cooled to room temperature, stirring speed slowly was added 13.4 g of chloroacetyl chloride (I. 2mol), within three hours after the addition and then mixed by stirring maintained at room temperature for 3 hours. Was slowly added 50 ml of ice water, the precipitate was collected by filtration. The filter cake was washed with 10 ml of water and 10 ml petroleum ether (twice). The filtrate and the organic layer together with 50 ml of dichloromethane and extracted twice with 50 ml brine and then paint extraction solution, the extract was dried over magnesium sulfate, the solution was removed, the solid with 100 ml of diethyl ether and recrystallized to afford 20g (71% yield compounds) of formula (3).

Step II: Formula (3) with a compound of formula (4) compound under acidic conditions and chloroform (CCl3H) heating the reaction, and the reaction system reached reflux to give a compound of formula (5),

Figure CN102796084AD00073

(5);

[0042] wherein, the formula (3) with a compound of formula (4) compound in acetonitrile (chloroform (CCl3H), the reaction system must be reached reflux, and must be reacted under acidic conditions to give the compound of formula (5). [0043] In this embodiment, the compound (3) (0. Imol) 28. 2 克 formula and the compound (4) (0. Imol) 21. 5 克 style with 3 g of trifluoroacetic acid was added to 200 ml of chloroform, in was stirred at reflux under nitrogen for 17 hours. After cooling to room temperature, spin-dry, to give 46. I g of a yellow solid of formula (5) compound (99% yield).

  Step three: (5) the compound obtained in toluene (toluene) and ammonium acetate (NH4OAc) reflux (6) of

Thereof,

Figure CN102796084AD00081

Compound  of formula (5) is ammonium acetate with toluene under reflux conditions for ring closure.

In the present embodiment, the compound (0. Imol) and 10 g of ammonium acetate (NH4OAc) was added 46. I g of formula (5) to IJ 200ml of toluene, heated under reflux for 3 hours with stirring. Was slowly added 50 ml of ice water, filtered, washed with 100 ml of toluene and extracted twice with 50 ml brine and then paint extraction solution, the extract was dried over magnesium sulfate, the solution was removed, the solid with 100 ml of diethyl ether and recrystallized to afford 40g (89% compound yield) of the formula (6).

Step Four: (6) compound in the catalyst and the associated button pinacolato ester (Bis (pinacolato) diboron) reacting a compound of formula (7),

Figure CN102796084AD00082

  wherein, Pd (dppf) 2Cl2 can be replaced by another of a palladium catalyst, a palladium catalyst with the other, the same effect.

  In the present embodiment, 44 g of the compound of formula (6) (0. Imol) and 3 g Pd (dppf) 2C12,25. 4 克 United pinacolato ester (0. Imol) and 8.4 g of sodium bicarbonate (0. Imol) was added to a 200 ml I. 4- dioxane, stirred at reflux for 24 hours. Diatomaceous earth filtration, spin dry. Spin-dry 100 ml of ethyl acetate dissolved. Anhydrous magnesium sulfate and spin dry. Recrystallization from ether to yield 40 g (82% yield) of a yellow solid of formula (7) compound.

Step Five: formula (7) under palladium catalyst compound and the compound (8) obtained by reacting the compound of formula (9),

Figure CN102796084AD00083

  wherein, Pd (dppf) 2Cl2 can be replaced by another of a palladium catalyst, a palladium catalyst with the other, the same effect.

  In the present embodiment, 48.9 g of the compound of formula (7) (0. Imol) and 3 g Pd compound (8) (0. Imol) (dppf) 2C12,41. 3 and 8 克 style. 4 g of sodium hydrogen carbonate (0. Imol) was added to a 200 ml I. 4- dioxane, stirred at reflux for 24 hours. Diatomaceous earth filtration, spin dry. Spin-dry 100 ml of ethyl acetate dissolved. Anhydrous magnesium sulfate and spin dry. Recrystallized from ether to give compound 55 g (85% yield) of a yellow solid of formula (9).

[0056] Step Six: formula (9) compound deprotected under acidic conditions to give a compound of formula (10),

[0057]

Figure CN102796084AD00091

  In the present embodiment, the 64.8 grams of formula (9) compound (0. Imol) was added to 100 ml I. 4_ dioxane was stirred, 100 ml of 5M / L of I under nitrogen 4- dioxane solution of hydrochloric acid. Spin-dry for 24 hours later, get 52. I g of pale yellow solid formula (10) compound (99% yield).

Step 7: Formula (10) with a compound (11) in a condensing agent is 2- (7-azo BTA) -N, N, N ‘, N’- tetramethyluronium hexafluorophosphate phosphate (HATU) under condensation reaction conditions to give the final product compound C0S-101, i.e. the compound of formula (I):

Figure CN102796084AD00092

In the present embodiment, the compound of formula 52. I g of (10) (0. Imol) was added to a 200 ml N, N- dimethylformamide (DMF) cooled to 0 ° with stirring, in a nitrogen atmosphere was added 20.2 g of triethylamine (0. 2mol) 0 After 10 minutes of stirring, was added 19 g of formula (11) compound (0. Ilmol) was added followed by 26 g HATU (0. 2mol), stirred at room temperature for 32 hours . Was slowly added 50 ml of ice water, the precipitate was collected by filtration. The filter cake was washed with 10 ml of water and 50 ml dichloromethane twice. Together with the filtrate and the organic layer was extracted 2 times 50 ml of dichloromethane, and then washed with 50 ml brine solution, the extract was dried over magnesium sulfate, the solution was removed, solid was recrystallized from 100 ml of ethanol, to give 50g (66% yield) The pale yellow compound C0S-101.

  In summary this compound on C0S-101 non-structural protein 5A inhibitor, or a pharmaceutically acceptable salt thereof, the treatment of hepatitis C active substance. A compound of formula (3) Friedel-Crafts reaction occurs directly from 2-bromo-naphthalene chloride and chlorine. A compound of formula (3) with a compound of formula (4) condensing a compound of formula (5). The compound of formula (5) self-condensation of a compound of formula (6). Of formula (6) is reacted with boronic acid pinacol ester linking reaction of the compound of formula (7). A compound of formula (7) with a compound of formula (8) coupling reaction of a compound of formula (9). Off compound under acidic conditions (9) protect the compound of formula (10) and formula (10) compound condensation of the final product C0S-101, method of operation of the invention is simple, mild conditions, process maturity, yield and high purity suitable for industrial production.

PATENT

WO 2013123092

http://www.google.com/patents/WO2013123092A1?cl=en

Figure imgf000003_0001

Scheme 3

Figure imgf000025_0001

3-3 2HCI salt

Step 1. Referring to Scheme 3, compounds l-5a (1.3 kg , 1.0 eq.), 2-2a (975.0 g, 1.0 eq.), NaHCOs (860.0 g, 3.80 eq.), Pd(dppf)Cl2 (121.7 g, 0.05 eq.), purified water (5.2 L, 4.0 volume) and 1 ,2-dimethoxy ethane (DME) (24.7 L, 19.0 volume) were charged into a 50.0 L 4-necked round bottom flask under argon atmosphere. After being degassed using argon for a period of 30 min, the reaction mass was slowly heated to ~ 80 °C and stirred at this temperature for 12 – 14 hrs. HPLC analysis indicated that > 97% of compound 2-2a was consumed. Next, the reaction mass was concentrated to completely remove DME under vacuum (600 mmHg) at 40 – 45 °C and the residue was diluted with 20% (v/v) MeOH in DCM (13.0 L , 10 volume) and purified water (13.0 L, 10.0 volume) with stirring. The organic layer was separated and the aqueous layer was extracted with 20% (v/v) MeOH in DCM (6.5 L x 2, 10.0 volume). The combined organic extracts were washed twice with water (6.5 L x 2, 10.0 volume) and once with saturated brine (6.5 L, 5.0 volume) and dried over anhydrous Na2S04. The solvent was removed under vacuum (600 mmHg) and the residue was purified by flash column chromatography using silica gel with hexanes/EtOAc as eluent to give compound 3-1 (1.0 kg, 63% yield) as off white solid with a purity of > 98.0%> determined by HPLC analysis. LC-MS (ESI): m/z 649.3 [M + H]+. 1H NMR (400 MHz, d6– DMSO): δ 12.26 – 12.36 (m, 1H), 11.88 – 11.95 (m, 1H), 8.23 (s, 1H), 8.11 (s, 1H), 7.91 (m, 3H), 7.85 – 7.87 (m, 2H), 7.51 – 7.81 (m, 3H), 4.78 -4.99 (m, 2H), 3.55 – 3.59 (m, 2H), 3.35 – 3.44 (m, 2H), 2.30 – 2.47 (m, 2H), 1.85 – 2.01 (m, 6H), 1.39, 1.14, 1.04 (s, s, s, 18H) ppm. Alternatively, compound 3-1 can be obtained following the same procedure and using compounds l-4a and 2-3a instead of compounds l-5a and 2-2a as the Suzuki coupling components.

Step 2. Compound 3-1 (1.0 kg, 1.0 eq.) and IPA (7.0 L, 7.0 volume) were charged into a 20.0 L four-necked RB flask under nitrogen atm. The reaction mass was cooled to 18 – 20°C and 3.0 N HC1 in isopropyl alcohol (7.0 L, 7.0 volume) was added over a period of 90 – 120 min under nitrogen atmosphere. After stirring at 25 – 30 °C for 10 – 12 hrs under nitrogen atmosphere, HPLC analysis indicated that > 98%> compound 3-1 was consumed. Next, the reaction mass was concentrated to remove IPA under vacuum at 40 – 45 °C. The semi solid obtained was added to acetone (2.0 L, 2.0 volume) with stirring and the resulting suspension was filtered under nitrogen atmosphere. The solid was washed with acetone (2.0 L, 2.0 volume) and dried in a vacuum tray drier at 40 – 45 °C for 10 hrs to give compound 3- 2 (860 g, 94%o yield) as pale yellow solid with a purity of > 98.0%> determined by HPLC analysis. LC-MS (ESI): m/z 449.2 [M + H]+. 1H NMR (400 MHz, -DMSO): δ 10.49 – 10.59 (m, 2H), 10.10 and 9.75 (m, m, 2H), 8.60 (s, 1H), 8.31 (s, 2H), 8.15 (m, 1H), 8.13 – 8.15 (m, 2H), 7.96 – 8.09 (m, 2H), 7.82 (s, 2H), 5.08 (m, 2H), 3.39 – 3.53 (m, 4H), 2.47 – 2.54 (m, 3H), 2.37 (m, 1H), 2.14 – 2.21 (m, 2H), 2.08 (m, 2H) ppm.

Step 3. Compound 3-2 (2.2 kg, 1.0 eq.) was added to a four necked round bottom flask charged with DMF (4.4 L, 20.0 volume) under a nitrogen atmosphere. After stirring for 15 min, the mixture was added N-Moc-L-Valine (226.2 g, 3.52 eq.) in one lot at 25 – 30 °C. Next, the mixture was cooled to -20 to -15 °C, followed by adding HATU (372.9 g, 2.0 eq.) portion wise over 30 min. After stirring for 10 min, a solution of DIPEA (238.9 g, 5.0 eq.) in DMF (1.1 L, 5.0 volume) was added over 45 min. Subsequently, the reaction mass was warmed to 25 – 30 °C with stirring. After stirring for 1 hr, HPLC analysis indicated that > 99%) of compound 3-2 was consumed. The reaction mixture was poured into water (38.0 L) and the mixture was extracted with DCM (10.0 L x 3, 45.0 volume). The combined organic extracts were washed with water (10.0 L x 3, 45.0 volume) and saturated brine (10 L, 45.0 volume) and dried over anhydrous Na2S04. The solvent was removed at 40 – 45 °C under vacuum (600 mmHg) and the residue was purified by column chromatography on silica gel using DCM and MeOH as the eluent to give compound 3-3 (1.52 kg, 47% yield) as off white solid with a purity of > 97.0% determined by HPLC analysis. LC-MS (ESI): m/z 763.4 [M + H]+. 1H NMR (400 MHz, -DMSO): δ 8.60 (s, 1H), 8.29 (s, 1H), 8.20 (s, 1H), 8.09 – 8.14 (m, 2H), 7.99 – 8.05 (m, 2H), 7.86 – 7.95 (m, 3H), 7.20-7.21 (m, 2H), 5.24 – 5.33 (m, 2H), 4.06 – 4.18 (m, 4H), 3.83 (m, 2H), 3.53 (m, 6H), 2.26 – 2.55 (m, 10H), 0.85 (m, 6H), 0.78 (m, 6H) ppm. The transformation of 3-2 to 3-3 (Compound I) can be achieved via a range of conditions. One of these conditions is described below.

A reactor was charged with N-Moc-V aline (37.15 g, 0.211 mol), acetonitrile (750 mL) and DIPEA (22.5 g). The reaction mixture was agitated for 10 min and HOBT (35.3 g 0.361 mole) and EDCI (42.4 g, 0.221 mole) were added while keeping temperature < 2 °C. The reaction mixture was agitated for 30 min and DIPEA (22.5 g) and compound 3-2 (48.0 g, 0.092 mole) was added slowly to reactor over 30 min to keep temperature < 3 °C. The reaction mixture was agitated 4 hrs at 20 – 25 °C, and sample was submitted for reaction completion analysis by HPLC (IPC specification: < 1.0% area 3-2 remaining). At the completion of reaction as indicated by HPLC analysis, isopropyl acetate (750 mL) was added to the reactor and stirred for 10 min. The organic layer (product layer) was washed with brine (300 mL x 2) and 2% NaOH (200 mL). The organic solution was filtered through a silica gel pad to remove insoluble material. The silica gel pad was washed with isopropyl acetate and concentrated under vacuum (400 mm/Hg) to a minimum volume. The crude product was purified by column chromatography on silica gel using ethyl acetate and methanol as eluent to give compound 3-3 (38.0 g, 65%> yield) with purity of > 95 %>. LC-MS (ESI): m/z 763.4 [M + H]+.

Step 4. Compound 3-3 (132.0 g, 1.0 eq.) and ethanol (324.0 mL, 2.0 volume) were charged into a 10 L four-necked round bottom flask under nitrogen atmosphere. After stirring for 15 min, the suspension was cooled to 5 – 10 °C, to it was added 2.0 N HC1 in ethanol (190 mL, 1.5 volume) over 30 min. The resulting solution was allowed to warm to 25 – 30 °C. Acetone (3.96 L, 30.0 volume) was added over 90 min in to cause the slow precipitation. Next, the suspension was warmed to 60 °C and another batch of acetone (3.96 L, 30.0 volume) was added over 90 min. The temperature was maintained at 55 – 60 °C for 1 hr, and then allowed to cool to 25 – 30 °C. After stirring at 25 – 30 °C for 8 – 10 hrs, the mixture was filtered. The solid was washed with acetone (660.0 mL, 5.0 volume) and dried in a vacuum tray drier at 50 – 55 °C for 16 hrs to give the di-HCl salt of compound 3-3

(compound I) (101 g, 71% yield) as pale yellow solid with a purity of > 96.6% determined by HPLC analysis.

Preparation of N-Moc-L-Valine

N-Moc-L-Valine is available for purchase but can also be made. Moc-L-Valine was prepared by dissolving 1.0 eq of L-valine hydrochloride in 2-methyltetrahydrofuran (2- MeTHF) /water containing sodium hydroxide and sodium carbonate, and then treating with 1.0 eq of methyl chloroformate at 0 – 5°C for 6 hr. The reaction mixture was diluted with 2- MeTHF, acidified with HC1, and the organic layer was washed with water. The 2-MeTHF solution is concentrated and the compound is precipitated with n-heptane. The solid was rinsed with 2-MeTHF/ n-heptane and dried in vacuo to give N-Moc-L-Valine in 68% yield. Crystallization of Compound I to Yield Form A

Compound I Salt Formation and Crystallization, Example 1

Ethanol (3.19 L, 1.0 volume, 200 proof) was charged to the 230-L glass lined reactor under nitrogen atmosphere. Free base form of compound 3-3 (3.19 kg, 4.18 mol) was added to the flask with stirring, stir continued for an additional 20 to 30 min. To the thick solution of 3-3 in ethanol was added slowly 2.6 N HC1 in ethanol (3.19 L, 1.0 volume) to the above mass at 20 – 25 °C under nitrogen atmosphere. The entire mass was stirred for 20 min at rt, and then heated to 45 – 50 °C. Acetone (128.0 L, 40.0 volume) was added to the above reaction mass at 45 – 50 °C over a period of 3-4 hrs before it was cooled to ~25 °C and stirred for ~15 hrs. The precipitated solid was collected by filtration and washed with acetone (6.4 L x 2, 4.0 volume), suck dried for 1 hr and further dried in vacuum tray drier at 40 – 45 °C for 12 hrs. Yield: 2.5 kg (71.0% yield), purity by HPLC: 97.70%, XRPD: amorphous.

Isopropyl alcohol (7.5 L, 3.0 volume) was charged to a 50.0 L glass reactor protected under a nitrogen atmosphere. The amorphous di-HCl salt of 3-3 (2.5 kg) was added to the above reactor with stirring. The entire mass was heated to 60 – 65 °C to give a clear solution. Stir continued at 65 ± 2 °C for ~15 hrs, solid formation started during this time. The heating temperature was lowered to ~50 °C over a period of 3 hrs, methyl tertiary butyl ether (12.5 L, 5.0 volume) was added to the above mass slowly over a period of ~3 hrs with gentle agitation. The above reaction mass was further cooled to 25 – 30 °C over 2 – 3 hrs. The solid was collected by filtration, washed with 10.0% isopropyl alcohol in methyl tertiary butyl ether (6.25 L, 2.5 volume), suck dried for 1 hr and further dried in a tray drier at 45 – 50 °C under vacuum (600 mm/Hg) for 70 – 80 hrs. Yield: 2.13 kg (85.0% recovery, 61.0% yield based on the input of compound free base 3-3), purity by HPLC: 97.9%.

FIG. 1 : 1H NMR (500 MHz, -DMSO): δ 15.6 (bs, 2H), 14.7 (bs, 2H), 8.58 (s, 1H), 8.35 (s, 1H), 8.25 (s, 1H), 8.18 (d, J= 8.7 Hz, 1H), 8.13 (s, 1H), 8.06 (d, J= 8.6 Hz, 1H), 8.04 (s, 1H), 8.00 (s, 1H), 7.98 (d, J= 8.7 Hz, 1H), 7.91 (d, J= 8.6 Hz, 1H), 7.36 (d, J = 8.6 Hz, 1H), 7.33 (d, J= 8.6 Hz, 2H), 5.31 (m, 1H), 5.26 (m, 1H), 4.16 (d, J= 7.7 Hz, 1H), 4.04 (m, 2H), 3.87 (m, 2H), 3.55 (s, 6H), 2.42 (m, 2H), 2.22-2.26 (m, 4H), 2.07-2.14 (m, 4H), 0.86 (d, J= 2.6 Hz, 3H), 0.84 (d, J= 2.6 Hz, 3H), 0.78 (d, J= 2.2 Hz, 3H), 0.77 (d, J= 2.2 Hz, 3H), 3.06 (s, OMe of MTBE), 1.09 (s, t-Bu of MTBE), 1.03 (d, 2Me of IP A) ppm.

FIG. 2: 13C NMR (500 MHz, /-DMSO): δ 171.6, 171.5, 157.4, 156.1, 150.0, 138.2, 138.0, 133.5, 132.5, 131.3, 129.8, 129.4, 128.0, 127.0, 126.4, 125.6, 125.3, 124.4, 124.2, 115.8, 115.0, 112.5, 58.37, 58.26, 54.03, 53.34, 52.00 (2 carbons), 47.71 (2 carbons), 31.52, 31.47, 29.42 (2 carbons), 25.94, 25.44, 20.13, 20.07, 18.37, 18.36 ppm.

FIG. 3: FT-IR (KBr pellet): 3379.0, 2963.4, 2602.1, 1728.4, 1600.0, 1523.4, 1439.7, 1420.6, 1233.2, 1193.4, 1100.9, 1027.3 cm“1.

Elemental Analysis: Anal. Calcd for C42H52C12N806: C, 60.35; H, 6.27; N, 13.41; CI, 8.48. Found C, 58.63; H, 6.42; N, 12.65, CI, 8.2.

FIG. 1 is a representative 1H NMR spectrum of Compound I Form A.

FIG. 2 is a representative 13C NMR spectrum of Compound I Form A.

FIG. 3 is a representative FT-IR spectrum of Compound I Form A.

References:
1. Lalezari, J. P.; et. al. PPI-668, a potent new pan-genotypic HCV NS5A inhibitor: phase 1 efficacy and safety. Hepatology 2012, 56, 1065A-1066A.

  1. ClinicalTrials.govA Study of the Efficacy and Safety of PPI-668 (NS5A Inhibitor) Plus Sofosbuvir, With or Without Ribavirin, in Patients With Chronic Hepatitis C Genotype-4. NCT02371408(retrieved on 24-03-2015)
    3. ClinicalTrials.gov Study of PPI-668, BI 207127 and Faldaprevir, With and Without Ribavirin, in the Treatment of Chronic Hepatitis C. NCT01859962 (retrieved on 15-09-2015)
    4. Lalezari, J.; et. al. High rate of sustained virologic response in patients with hcv genotype-1a infection: a phase 2 trial of faldaprevir, deleobuvir and ppi-668, with and without ribavirin. EASL-The International Liver Congress 2014 49th Annual Meeting of the European  Association for the Study of the Liver London, United Kingdom  April 9-13 (article here)
US20070185175 * 27 Jul 2006 9 Aug 2007 Bristol-Myers Squibb Company Benzothiazole and azabenzothiazole compounds useful as kinase inhibitors
US20080050336 * 8 Aug 2007 28 Feb 2008 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
WO2012087976A2 * 19 Dec 2011 28 Jun 2012 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
WO2013123092A1 * 13 Feb 2013 22 Aug 2013 Presidio Pharmaceuticals, Inc. Solid forms comprising inhibitors of hcv ns5a, compositions thereof, and uses therewith
WO2013158776A1 * 17 Apr 2013 24 Oct 2013 Gilead Sciences, Inc. Compounds and methods for antiviral treatment
US8765731 16 Nov 2012 1 Jul 2014 Vertex Pharmaceuticals Incorporated Benzimidazole analogues for the treatment or prevention of flavivirus infections
US8779156 24 Sep 2012 15 Jul 2014 Vertex Pharmaceuticals Incorporated Analogues for the treatment or prevention of flavivirus infections
US8809330 1 Nov 2013 19 Aug 2014 Gilead Sciences, Inc. Pyrazolo[1,5-A]pyrimidines for antiviral treatment
US8946238 20 Dec 2012 3 Feb 2015 Gilead Sciences, Inc. Pyrazolo[1,5-A]pyrimidines as antiviral agents
US8980878 17 Apr 2013 17 Mar 2015 Gilead Sciences, Inc. Compounds and methods for antiviral treatment
US20110274648 * 4 Nov 2010 10 Nov 2011 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors

////////////Phase III, Hepatitis C, RAVIDASVIR, PPI-668,  BI 238630


Filed under: Phase3 drugs Tagged: BI 238630, hepatitis C, PHASE 3, Phase III, PPI-668, RAVIDASVIR

Daprodustat, GSK1278863

$
0
0

ChemSpider 2D Image | daprodustat | C19H27N3O6

Figure imgf000039_0001

Daprodustat, GSK1278863

960539-70-2

GSK1278863; GSK 1278863; GSK-1278863; Daprodustat

C19H27N3O6
Exact Mass: 393.18999

(1,3-dicyclohexyl-2,4,6-trioxohexahydropyrimidine-5-carbonyl)glycine

N-[(l,3-dicyclohexyl-6-hydroxy-2,4-dioxo-l,2,3,4- tetrahydro-5-pyrimidinyl)carbonyl]glycine

2-(1,3-dicyclohexyl-2,4,6-triohexahydropyrimidine-5-carboxamide acetic acid
Mechanism of Action: HIF-prolyl hydroxylase inhibitor
Indication: anemia, diabetic wounds, and reduction of ischemic complications
Development Stage: Phase II
Developer:GlaxoSmithKline

UNII:JVR38ZM64B

Daprodustat , also known as GSK1278863, is a novel HIF-prolyl hydroxylase inhibitor. Hypoxia inducible factor (HIF) stabilization by HIF-prolyl hydroxylase (PHD) inhibitors may improve ischemic conditions such as peripheral artery disease (PAD). Short-term treatment with a novel HIF-prolyl hydroxylase inhibitor (GSK1278863) failed to improve measures of performance in subjects with claudication-limited peripheral artery disease

  • OriginatorGlaxoSmithKline
  • ClassAntianaemics; Pyrimidines; Small molecules
  • Mechanism of ActionErythropoiesis stimulants; Prolyl hydroxylase inhibitors
  • Phase II Anaemia; Perioperative ischaemia
  • Phase I Diabetic foot ulcer; Tendon injuries
  • DiscontinuedPeripheral arterial disorders

Most Recent Events

  • 27 Jul 2015No recent reports of development identified – Phase-II for Anaemia in India and New Zealand (PO)
  • 27 Jul 2015Daprodustat is still in phase II trials for Anaemia in the USA, Australia, Canada, Czech Republic, Denmark, France, Germany, Hungary, Japan, Poland, Russia, Spain, South Korea, and United Kingdom
  • 01 Jun 2015GlaxoSmithKline completes a phase I trial in Tendon injuries (In volunteers) in USA (PO) (NCT02231190)
WHO ATC code: B03 (Antianemic Preparations)

C (Cardiovascular System)

C01 (Cardiac Therapy)

D03 (Preparations for Treatment of Wounds and Ulcers)

M09A-X (Other drugs for disorders of the musculo-skeletal system)

EPhMRA code: B3 (Anti-Anaemic Preparations)

C1 (Cardiac Therapy)

C6A (Other Cardiovascular Products)

D3A (Wound Healing Agents)

M5X (All Other Musculoskeletal Products)

WO 2007150011

https://www.google.com.ar/patents/WO2007150011A2

Illustrated Methods of preparation

Scheme 1

Figure imgf000023_0001

a) 1. NaH, THF, rt 2. R1NCO, 60 0C; b) 1. NaH, THF or dioxane, rt 2. R4NCX, heat; c) H2NCH2CO2H, DBU, EtOH, 1600C, microwave.

Scheme 2

Figure imgf000023_0002

a) R1NH2, CH2Cl2 or R1NH2-HCl, base, CH2Cl2; b) CH2(C(O)Cl)2, CH2Cl2, reflux or CH2(CO2Et)2, NaOEt, MeO(CH2)2OH, reflux or 1. EtO2CCH2COCl, CHCl3, 70 0C 2.

DBU, CHCl3, 70 0C; c) 1. YCNCH2CO2Et,, EtPr’2N, CHCl3 or CH2Cl2 2. aq NaOH, EtOH, rt. Scheme 3 (for R1 = R4)

a) CDI,

Figure imgf000024_0001

DMF, 70 0C or , EtOAc, rt

Scheme 4

Figure imgf000024_0002

a) OCNCH2CO2Et, EtPr’2N, CHCl3 or CH2Cl2; b) 1. R1HaI, Na/K2CO3, DMF or DMA, 100 0C or R1HaI, pol-BEMP, DMF, 120 0C, microwave 2. aq NaOH, MeOH or EtOH, rt.

Scheme 5

Figure imgf000024_0003

a) 1. CH2(CO2H)2, THF, O 0C – rt 2. EtOH, reflux; b) 1. OCNCH2CO2Et, EtPr’2N, CH2Cl2 2. aq NaOH, EtOH, rt.

Scheme 6

Figure imgf000024_0004

a) 1. Phthalimide, DIAD, PPh3, THF 2. (NH2)2, EtOH, reflux.

Scheme 7

Figure imgf000025_0001

a) Ac2O, AcOH, 130 0C.

Example 18

Figure imgf000039_0001

N-T(1 ,3-Dicvclohexyl-6-hydroxy-2,4-dioxo- 1 ,2,3,4-tetrahvdro-5-pyrimidinyl)carbonyl1grycine Method 1

18.1a) h3-Dicvclohexyl-2A6(lH,3H,5H)-pyrimidinetrione. Dicyclohexylurea (3.0 g, 13.39 mmoles) was stirred in chloroform (80 mL) and treated with a solution of malonyl dichloride (1.3 mL, 13.39 mmoles) in chloroform (20 mL), added dropwise under argon. The mixture was heated at 500C for 4 hours, wasahed with 1 molar hydrochloric acid and evaporated onto silica gel. Flash chromatography (10-30% ethyl acetate in hexane) to give the title compound (2.13 g, 55%). 1Η NMR (400 MHz, OMSO-d6) δ ppm 4.46 (tt, J=12.13, 3.54 Hz, 2 H), 3.69 (s, 2 H), 2.15 (qd, J=12.46, 3.28 Hz, 4 H), 1.77 (d, J=13.14 Hz, 4 H), 1.59 (t, J=12.76 Hz, 6 H), 1.26 (q, J=12.97 Hz, 4 H), 1.04 – 1.16 (m, 2 H)

18.1b) N-r(1.3-Dicvclohexyl-6-hvdroxy-2.4-dioxo-1.2.3.4-tetrahvdro-5- pyrimidinvDcarbonyll glycine. Ethyl isocyanatoacetate (802 uL, 7.15 mmoles) was added to a mixture of l,3-dicyclohexyl-2,4,6(lH,3H,5H)-pyrimidinetrione (2.1 g, 7.15 mmoles) and diisopropylethylamine (2.47 mL, 14.3 mmoles) in dichloromethane (100 mL) and stirred overnight. The reaction mixture was washed with 1 molar hydrochloric acid (x2) and evaporated. The residue was dissolved in ethanol (10 mL) and treated with 1.0 molar sodium hydroxide (5 mL). The mixture was stirred for 72 hours, acidified and extracted into ethyl acetate. Some ester remained, therefore the solution was evaporated and ther residue was dissolved in 1 molar soldium hydroxide solution with warming and strred for 2 hours. The mixture was acidified with IM HCl and extracted with ethyl acetate (x2). The combined extracts were washed with 1 molar hydrochloric acid , dried and evaporated to a solid which was slurried in a mixture of diethyl ether and hexane, collected, washed with the same solvent mixture and dried to give the title compound (1.86 g, 66%). IH NMR (400 MHz, DMSO-^6) δ ppm 13.07 (br. s., 1 H), 10.19 (t, J=5.31 Hz, 1 H), 4.63 (t, J=10.99 Hz, 2 H), 4.12 (d, J=5.56 Hz, 2 H), 2.27 (q, J=I 1.71 Hz, 4 H), 1.79 (d, J=12.88 Hz, 4 H), 1.50 – 1.69 (m, 6 H), 1.28 (q, J=12.97 Hz, 4 H), 1.12 (q, J=12.72 Hz, 2 H)

Method 2

18.2a) 1.3-Dicvclohexyl-2.4.6πH.3H.5H)-pyrimidinetrione. A solution of N5N- dicyclohexylcarbodiimide (254 g; 1.23 mol.) in anhydrous TΗF (700 mL) was added dropwise to a cold (0 0C) solution of malonic acid (64.1 g; 0.616 mol.) in anhydrous TΗF (300 mL) over a period of- 30 minutes. The mixture was stirred and allowed to warm to room temperature over 2 h. (After 1 h, the mixture became very thick with precipitate so further anhydrous TΗF (500 mL) was added to facilitate agitation.). The mixture was filtered and the filtrate evaporated to afford a yellow solid which was immediately slurried in ethanol (1 L) and heated to reflux temperature. The mixture was then allowed to cool to room temperature then filtered and the solid washed with cold ethanol (250 mL) to afford the title compound (129.4 g; 72%) as a colorless solid. 1Η NMR (400 MHz, DMSO-(Z6) δ ppm 1.03 – 1.18 (m, 2 H) 1.18 – 1.34 (m, 4 H) 1.59 (t, J=13.14 Hz, 6 H) 1.76 (d, J=12.88 Hz, 4 H) 2.04 – 2.24 (m, 4 H) 3.69 (s, 2 H) 4.35 – 4.54 (m, 2 H).

18.2b) Ethyl N-[(l .3-dicvclohexyl-6-hvdroxy-2.4-dioxo- 1.2.3.4-tetrahydro-5- pyrimidinyPcarbonyll glycinate. A solution of l,3-dicyclohexyl-2,4,6(lH,3H,5H)-pyrimidinetrione (120.0 g; 0.41 mol.) and diisopropylethylamine (105.8 g; 0.82 mol.) in dichloromethane (1 L) was stirred and treated dropwise with a solution of ethyl isocyanatoacetate (53.0 g; 0.41 mol.) in dichloromethane (500 mL) and the mixture was then stirred at room temperature overnight. The mixture was then treated dropwise with 6M aq. hydrochloric acid (500 mL) and the separated organic layer was dried and evaporated. The resulting solid was slurried in hexanes (500 mL) and heated to reflux temperature. The mixture was then allowed to cool and filtered to afford ethyl N- [(1 ,3-dicyclohexyl-6-hydroxy-2,4-dioxo- 1 ,2,3,4-tetrahydro-5-pyrimidinyl)carbonyl]glycinate (159.1 g; 92%) as a cream powder. IH NMR (400 MHz, CHLOROFORM-,/) δ ppm 1.24 (s, 2 H) 1.37 (s, 7 H) 1.52 – 1.76 (m, 6 H) 1.78 – 1.94 (m, 4 H) 2.25 – 2.48 (m, 4 H) 4.17 (d, J=5.81 Hz, 2 H) 4.28 (q, J=7.24 Hz, 2 H) 4.74 (s, 2 H) 10.37 (t, J=4.67 Hz, 1 H). 18.2c)

N-rπ^-Dicyclohexyl-ό-hydroxy^^-dioxo-l^J^-tetralivdro-S- pyrimidinyDcarbonyll glycine. A stirred suspension of ethyl Ν-[(l,3-dicyclohexyl-6-hydroxy-2,4- dioxo-l,2,3,4-tetrahydro-5-pyrimidinyl)carbonyl]glycinate (159.0 g; 0.377 mol.) in ethanol (1.5 L) was treated dropwise with 6M aq. Sodium hydroxide (250 mL) and stirred at room temperature for 3 h. The solution was then acidified by the dropwise addition of 6M aq. hydrochloric acid (300 mL), diluted with water (IL) and then filtered. The crude solid was slurried in water (2 L) then stirred vigorously and heated at 35 0C for 1 h and filtered and dried. The solid material (~ 138 g) was then crystallized from glacial acetic acid (1.5 L) (with hot filtration to remove a small amount of insoluble material). The solid, which crystallized upon cooling, was collected and washed with cold glacial acetic acid (3 x 100 mL) to afford N-[(l,3-dicyclohexyl-6-hydroxy-2,4-dioxo-l,2,3,4- tetrahydro-5-pyrimidinyl)carbonyl]glycine (116.2 g; 78%) as a colorless solid.

IH NMR (400 MHz, DMSO-(Z6) δ ppm 1.11 (d, J=12.88 Hz, 2 H) 1.27 (q, J=12.80 Hz, 4 H) 1.62 (s, 6 H) 1.70 – 1.90 (m, J=12.88 Hz, 4 H) 2.11 – 2.44 (m, 4 H) 4.11 (d, J=5.81 Hz, 2 H) 4.45 – 4.77 (m, 2 H) 10.19 (t, J=5.81 Hz, 1 H) 13.08 (s, 1 H).

………….

SEE

http://www.google.com/patents/WO2014132100A1?cl=en

 

 

 

///////////////Daprodustat, GSK1278863, GlaxoSmithKline , PHASE 2


Filed under: Phase2 drugs, Uncategorized Tagged: Daprodustat, GlaxoSmithKline, GSK1278863, phase 2

Pharma Regulations for Generic Drug Products in India and US: Case Studies and Future Prospectives

$
0
0

Dr. Suryakanta Swain

Introduction

The Indian pharmaceutical industry has come a long way from being non-existent before independence to a prominent provider of medicines and health care products in the current decade. The Indian pharmaceutical industry at present is the global leader of growing pharmaceutical manufacturing companies, providing wide range capabilities in the complex field of technology and drug manufacturing. Indian pharma market growing at a rapid pace currently providing Indian pharmaceutical industry third rank all over the world in terms of volume and fourteen ranks, according to market value [1]. The major strength of currently growing Indian pharmaceutical sector is its capability to manufacture wide range of simple analgesic pills to complicated antibiotics, cardiac compounds with peer quality and efficacy and altogether exporting them to developed world. The industry bulk profit comes from exporting generics and API to the developed market mainly US followed by UK, Germany, Brazil etc. The total share of generics accounts in export is 58% providing the major boost, the Indian commerce ministry has set an ambitious export target of $ 25 billion by 2013-14, which can be achieved only by major contribution from generics market [2]. The Indian generics market is growing day by day with Indian pharmaceutical companies seeking more Abbreviated New Drug Application approvals (ANDAs) in US in major segments such as cardiovascular, antibiotics and other groups. The major force for the development of generics market in US came in the form of enacting the Drug Price Competition and Patent Restoration Act of 1984, public law 98-417 better known as “The Hatch- Waxman Act” which created opportunities for developing and marketing generics or better called as abbreviated new drug applications for 180 days. Under ANDAs a pharmaceutical manufacturer can develop and market low price generic version of previously approved innovator drugs, thus providing the same product to patient in pregnable price with safety and efficacy. A generic or biosimilar drug product is one that is comparable to an innovators drug product in dosage form, strength and route of administration, quality, performance characteristics and intended use. All approved products, both innovator and generics, are enlisted in FDA’s orange book. Generic drug application are termed as “abbreviated” because they are generally not required to include preclinical (animal) and clinical (human) data to establish safety and efficacy instead, generics applicant must demonstrate that there product is bioequivalent (i.e., performs in similar manner to innovator products). India has its unique position all over the world generics market, providing drugs at low cost to the developed world, this is because of its rigid and flexible pharma regulations, patent act which is updated from time to time, thus Indian generics market is playing a major role in growth of Indian economy as it provides a major share in export, mainly exporting generics to US, therefore a proper set of rules and regulations is required in future for producing generics and exporting them, so that Indian pharmaceutical sector and economy maintains its growth and becomes leaders globally.

A Generic Drugs Must:
▪ Contain the same active ingredients as the innovator drug
▪ Be identical in strength, dosage form, and route of administration
▪ Have the same use indications
▪ Be bioequivalent (as a marker for therapeutic interchangeability)
▪ Meet the same batch requirements for identity, strength, purity and quality
▪ Be manufactured under the same strict standards of GMP required for innovator  products
Table 1: Regulatory requirements for generic drugs.

Pharma Regulations for Generic Product in India and US

Generics have an important role to play in public health as they are well known to medical community and usually more affordable due to competition. They are formulated when patent and other exclusivity rights expire. The key for generic medicines is their therapeutic interchangeability with originator products. To ensure the therapeutic efficacy generic products must be pharmaceutically interchangeable (contain the same amount of active ingredient and have the same dosage form) and bioequivalent to the originator product. Bioequivalence is usually established using comparative in-vivo pharmacokinetic studies with originator products. The detailed description how it is carried out is described in respective WHO document and national regulatory guidelines. Well resourced regulatory authorities require that a generic medicine must meet certain regulatory criteria [3,4]. The major regulatory requirement for generic drug is presented in Table 1. For applying the ANDA’s in US, application is submitted under any of the below subsections of 505(j) of Federal act, it is important to comply with rule and regulations of US because it’s the major export destination for Indian generics manufacturers [5], the various application which can be applied for ANDAs in US is depicted in Table 2. The ANDAs review process is most important for developing generics, the review by FDA and CDER is done for generic applicant to compare its therapeutic bioequivalent with brand drugs after its approval for equivalency generic version of drug can be marketed (Figure 1). The review for equivalency is done by taking into account the bioavailability of product with branded drug, its microbiology, chemistry and labeling of product, this are current regulation to follow for generic approvals given by respective FDA.

Subsection of 505(J) Products Type
Paragraph I For the products for which no patent information is available in the orange book.
Paragraph II Used for the products for which all the applicable patents are expired
Paragraph III Used for the products for which the some or all the applicable patents are valid and the applicant confirms that the product will not be placed in the market till such patents are expired.
Paragraph IV Used for the products for which some or all the applicable patents are valid and applicant try to file the product which does not infringe those patents or applicant invalidates the granted patents. On successful outcome the generic applicant enjoys the six month exclusivity in the market.
Table 2: Different types of ANDA applications in US.

 

Figure 1: Explain the ANDA reviews process for development of generic drugs.

Future Generic Products in India and US

It is seen that there is an upward swing in the generic market. It has reached 100 billion dollars in the past and is estimated to be three times higher than the overall growth of drugs. The current trend exhibits that blockbuster drugs are scheduled to lose their patent protection, opening the doors to cheaper generic drugs between 2013 and 2015 with the total market value in billions. It is expected that the percentage of generic drugs in the US market will rise from 14 to 21. This growth will enhance the export prospect of India and it will be doubled every year. It will be due to increase in the number of low cost workers and degree of innovation. Recent success in track record in design operation of high tech manufacturing, testing, quality control, research, clinical testing and biotechnology also contribute to this higher growth. Indian pharmaceutical industries those who have USFDA (United States Food and Drug Administration) affiliations and approval of ANDA (Abbreviated New Drug Applications) will stand benefited. Now India’s global share in the field of generic market is stipulated at 35% which is very high [6,7]. Table 3 describes list of various drugs going to get off-patent in 2015. To make the situation more favorable the Indian government has also introduced scheme of providing generic drugs to patient in hospitals with various Jan-aushadhi Kendra (Facilitation Centre). Thus future prospects of generics in India and US are very high as they are the next big thing in health care scenario. Consistent with prior research, MEPs (Market Exclusivity Periods) for drugs experiencing initial generic entry in 2011-2012 was 12.6 years for New Molecular Entities (NMEs) with sales greater than $100 million in the year prior to generic entry, and 12.9 years for all NMEs. Further research may reveal variation by type of NME, whether defined by molecule type or other classification. Generic competition has intensified over the past 10-15 years, and the MEP has become an even more important indicator of the economics of brand-name drugs. The MEP is critical to manufacturers’ ability to earn profits on brand-name drugs to fund future research and development activities, and brand-name drug shares rapidly drop following initial generic entry. Over 80% of brandname drugs experiencing initial generic entry in 2012 had faced at least one Paragraph IV patent challenge from a generic manufacturer, up from only 9% for drugs experiencing initial generic entry in 1995. These challenges are filed relatively early in the brand drug life cycle, on average within 7 years of brand launch. Developments for the generic pharmaceutical industry are encouraging as more brand-name drugs come off patent and payers push for cost cuts in health care. In addition, due to increasing FDA budget and staffing should begin to cut the backlog of branded and generic drug applications and increase the ability of the FDA to inspect facilities here and overseas as generic biologics get to market in the next few years [8].

Sl. No. Name of the Drug Category Patent Holder Expiry Year
1 Eletriptan Migraine Merck August, 2013
2 Teriparatide Hormone Eli Lilly July, 2013
3 ImatinibMesylate Oncology Novartis May, 2013
4 Insulin Lispro Diabetes Eli Lilly May, 2013
5 Linezolid Antibiotic Pharmacia Nov, 2014
6 Transtuzumab Biopharm Gentech Oct, 2014
7 Posaconazole Antibiotic Shering Aug, 2014
8 Omalizumb Respiratory Roche Feb, 2015
9 Telethromycin Antibiotic Aventis April, 2015
10 Alemtuzumb Oncology Millennium Dec, 2015
Table 3: List of some important drugs going to be off-patents

Upcoming Challenges for Indian Generics Manufacturers in Global Market

The generic drug companies in India have broad technological and diversified market capabilities. As more and more patents expire, the generic portion of the pharmaceutical market is expected to continue to have increased sales. The scientific capability for manufacturing and supplying generic drugs of these companies will give them an edge over others and make them major players in the international generics market. Fortunately India has the best subject skills to galvanize foreign investors. The encouraging scenario of basic research and drug discovery will also support the changed dynamics. But their future sustainable growth depends on sustaining in competitive markets of developed world. The major challenges for generic manufactures are strengthening the existing regulatory system especially for enabling more detailed and universal classification of drugs and chemicals between branded generic and generics. High R&D cost and investment in research is also a major stumbling block in this direction [9].

Generic  Name Generic Manufacturer Brand Name Approval Date Year Of Exclusivity Rights Expire
Pioglitazone Hydrochloride and Glimepiride Tablets, 30 Mg/2 Mg and 30 Mg/4 Mg Takeda Pharms USA Duetact Tablets Jul 28, 2006 2027
Fexofenadine Hydrochloride Orally Disintegrating Capsules or  oral: 60 Mg Sanofi Aventis USA Children’s Allegra Allergy & Allegra Hives Jul 25, 1996 May 26, 2014
Mupirocin Calcium, Cream; topical Glaxosmithkline Bactroban Cream Dec 11, 1997 Oct 29, 2013
Doxorubicin Hydrochloride Liposome Injectable, Liposomal; Injection 2 mg/ml Janssen Res And Dev Doxil Liposome Injection Nov 17, 1995 May 17, 2014
Zoledronic Acid Injection, 4 Mg (Base)/5 Ml; Packaged In Single-Dose Vials Novartis Zometa Injection Jun 17, 2011 2031
Esomeprazole sodium – injectable;intravenous (Eq 20mg base/vial) Astrazeneca Nexium I.V. For Injection March 31, 2005 2025
Amlodipine besylate; hydrochlorothiazide; valsartan – tablet; oral (5Mg;12.5Mg;160Mg) Novartis ExforgeTablets April 30, 2009 2029
Table 4: Describes list of various new ANDAs approval in the year 2013.

Amendments in the Pharma Regulations for Generic Products

The Hatch-Waxman Act enacted 1984 is a landmark act. It allows generic drugs to enter the market without repeating expensive clinical trials required for their branded drugs. The legislation is meant for balancing the world of generic and branded drug industries. It provides accessibility to lower-cost generic drugs while still encouraging innovation and development of new drugs. Nevertheless, the legislation created unintended legal barriers that have slowed the entry of generic drugs into the market due to significant legal loopholes. The generic drug companies are allowed to market the drug after the patent and certain exclusivities expire. It has led to the prolific growth of generic drugs in the market. Thus some changes are required so that the loopholes can be filled and the regulation can be strengthened and selling of low cost drugs can be achieved. The change in rule related to alleged abuse of the 30-months stay provision is to be taken care were the ANDA applicant informs the original patent holder about the generic version filing, where they have 45 days to file a patent infringement suit against the generic applicant. If an infringement suit is filed within the 45-days period, FDA approval to market the generic version is automatically postponed for 30 months. These stays are extremely advantageous to innovating companies, because they provide over 2 years of additional market sales. Company takes profit by utilizing this route and delays the entry of generic drug in market; many steps have been taken by amending act of Greater Access to Affordable Pharmaceuticals Act passed in 2003 by American government. Extending the extensions by alleged abuse of the 30-month stay provision is done by many companies that holds patent, the companies are able to further delay the market entry of generic drugs is through multiple patent listings in the Orange Book, which is the FDA’s official listing of all the approved products. There are instances in which brand-name companies listed related patents in the Orange Book after an ANDA had already been filed by a generic manufacturer. The effect of these “later-listings” is that the generic applicant is then required to re-certify that the laterlisted patent is also invalid or not infringed and notify the patent holder of the re-certification. Thus more delay occurs in generic drug to reach market [10]( Figure 2).

Recent Cases and Incidents of Generic Products Regulation in India and US

The future prospects of generic product regulation in India and US are of great importance as they will decide the direction of growth of Indian Pharmaceutical Industries. Based on the recent cases and incidents that have occurred in India and US related to the generic product utilization, the new crucial roles will be implemented. The list of a few recent cases and incidents that happened in connection with generics in India & US are discussed in detail below (Figure 3).

Figure 2: Schematic overview for the benefits of Hatch-Waxman Act.

 

Figure 3: Steps for the launching of generic drugs

The Karen L. Bartlett case

In December-2004, Physician of Karen L. Bartlett was prescribed Clinoril, the brand-name version of the Non-Steroidal Anti- Inflammatory Drug (NSAID) sulindac, for shoulder pain of Karen L. Bartlett. Her pharmacist dispensed a generic form of sulindac manufactured by petitioner Mutual Pharmaceutical. Karen L. Bartlett soon developed an acute case of toxic epidermal necrolysis. She is severely disfigured, has physical disabilities, and is nearly blind. At the time of the prescription, sulindacs label did not specifically refer to toxic epidermal necrolysis. By 2005, however, the FDA had recommended changing all NSAID labeling to contain a more explicit toxic epidermal necrolysis warning. Respondent sued Mutualin New Hampshire state court. A jury found Mutual liable on respondent’s design-defect claim and awarded her over $21 million. The First Circuit gets ratified. As relevant, it found that neither the FDCA nor the FDA’s regulations pre-empted respondent’s design-defect claim. It distinguished PLIVA, Inc. v. Mensing, 564 U.S in which the Court held that failure-to-warn claims against generic manufacturers are pre-empted by the FDCA’s prohibition on changes to generic drug labels by arguing that generic manufacturers facing design-defect claims could comply with both federal and state law simply by choosing not to make the drug at all. This case is being closely watched by pharmaceutical companies, federal regulators and others, the Supreme Court will decide on whether Mutual can be held responsible for Ms. Bartlett’s injuries. The outcome is likely to further clarify the legal recourse for patients who take generic drugs, which now account for 80 percent of all prescriptions in the US. The verdict on both the sides will be playing a crucial role in drafting future pharma regulations as if the court agrees with Mutual and rules that generic companies cannot be sued for defective products, trial lawyers warn that patients will be left with very few options if they are injured by a generic drug whereas manufacturers of generic drugs and other business groups have said that if the court sides with Ms. Bartlett, the decisions of individual juries could trump the authority of federal agencies like the Food and Drug Administration and potentially lead drug makers to remove valuable medicines from the market. Thus this case will be important for the future of generics drug market in US and India [11,12].

 

Pay to delay pharmaceutical case

The question of whether the manufacturer of a branded drug can pay another drug manufacturer to keep a generic version of the drug off the market was heard by the United States Supreme Court on 25th March, 2013. The court will decide whether “pay-to-delay” or reverse settlements arrangements, in which the manufacturer of a branded medication pays another company to keep a generic version off the market, are legal or not, the outcome of the case is very important because it will decide for how many patients pay for medications. Federal Trade Commission challenges the payments. It sees these arrangements as collusion, design to stop competition in the market place and is meant for violation of antitrust laws of the nations. The drug makers, in contrast, see the settlements as a routine way of settling a legal dispute, with each side getting something it wants. The Hatch- Waxman Act 1984 has some loophole. Payments are made possible by using these loopholes. Certain amendments are made in the last decade to encourage generic manufacturers to challenge patents held by branded manufacturers before they are set to expire. Typically, the generic manufacturer files for FDA approval to market a generic version of a branded medication that is still under patent protection, and the branded manufacturer sues the generic manufacturer for patent infringement. An increasing number of such cases end in “payto- delay” agreements according to which the generic manufacturer agrees to hold off on introducing the generic version in exchange for payment from the branded manufacturer. The case in point is Androgen (testosterone gel), produced by Solvay Pharmaceuticals whose patent is set to expire in 2020. The bone of contention between Actavis (formerly Watson Pharmaceuticals) and Solvay Pharmaceuticals was Andro Gel. Actavis filed for FDA approval to market a generic version of Andro Gel in 2003, and Solvay sued. In 2006, the FDA approved the generic version for marketing of Actavis, but the suit remained status quo. Later in 2006, the companies came to a settlement according to which Solvay would pay Actavis $20 to $30 million per year in exchange for help with marketing and an agreement to keep its generic version of Andro Gel off the market until 2015. The FTC (Federal Trade Commission) contends that the drug companies colluded to maintain Solvay’s monopoly on Andro Gel because, without the settlement, the generic version would have become available in 2006. A federal district court dismissed the FTC’s argument in this case, but another district court in a similar case decided the opposite way, so it is now up to the Supreme Court to decide and decision is expected. Moreover the best verdict according to many experienced federal judges that supreme court should not generalize the law, where as it should be implemented on case to case basis, thus this case should be great importance for Pharma regulators to draw guidelines for future regulations of generics in India and US and it will be important for patients to decide whether they will opt for cheaper or expensive medicines [13,14].

The Ranbaxy saga case

The criminal fraud that Ranbaxy has done with US FDA has let down many but it’s the fellow generics drug maker of India that will face the heat, this will be a very important incident which will decide fate of generics drug market of India in US and its regulation. Ranbaxy pharmaceutical of India is charged with producing low quality generic drugs in US and manipulating data’s required for filing NDA and ANDA approvals in US, thus cheating their counter parts in many ways to be first in the race of producing generic version. Ranbaxy pleaded guilty to seven federal criminal counts of selling adulterated drugs with intent to defraud, failing to report that its drugs did not meet specifications, and making intentionally false statements to the government. Ranbaxy agreed to pay $500 million in fines, forfeitures, and penalties-the most ever levied against a generic-drug company. The company, now majority owned by Japanese drug maker Daiichi Sankyo, sells its products in more than 150 countries and has 14,600 employees. It also came to light that even Ranbaxy scientist adulterated there generic testing drug with branded drugs for manipulating bioequivalence study.

Thus these serious allegations on one of the top India pharmaceutical company could be a major setback for generic manufactures and Indian Pharma regulator as they have failed to, therefore some strict regulations could be implemented by US FDA in future for Indian generics producers which could be a serious issue as it will lead to effect the generics drug market in India. Thus this will be the major factor which will decide the fate of future regulation of generics in India and US [15,16].

Miscellaneous cases and incidents

The study discusses the case of Swiss drug maker Novartis plea overruled recently by the Supreme Court was an attempt to win patent protection for its cancer drug Glivec. This was a serious blow to Western pharmaceutical firms who are increasingly focusing on India to drive sales and it also affects Indian and US generic market. Glivec (ß-polymorphic form of imatinib mesylate) is indicated for treatment of certain blood and stomach cancers. The Supreme Court decision implies that a clutch of Indian companies, including Cipla, Ranbaxy and Natco, could continue marketing generic versions of the drug at a fraction of the cost of Novartis’ product. While Novartis’ Glivec costs over one lakh a month, local companies sell versions of the drug at roughly ten thousand a month. Supreme Court’s ruling states that the drug has failed in “both the tests of invention and patentability” under Indian law. On the other hand, Glivec is widely recognized as one of the most important medical discoveries in decades, but it lost the battle on innovative quality grounds. The verdict can be interpreted as a battle between research and innovation on one side and public health and affordability on the other. It is true that the prospect of producing cheaper generic versions of lifesaving drugs in the country, thus sale of generics will increase and generic market will be boosted up. Thus the case study suggests that the future of generics in India is bright and this case will be a benchmark for it. The well documented Novartis case in the ‘Glivec’ matter has brought the Indian patent system into sharp focus, whereas Indian regulatory authority should reform new rules for granting patent so that bigger MNCs should be attracted to India in future for better business [1820].

Recent patents

With expiration of patent branded drugs are applied for generics version, some of the new ANDAs approval in year 2013 [17] are described briefly in Table 4.

CONCLUSION

In situations where demand for medicines exceeds supply, and cost effective drug in demand with minimum expenditure, generic drug are best choice fulfilling this demand. The current and future prospective of generics in India and US is very bright as Indian government looking towards generic drugs for providing better health care to public. Indian pharmaceutical industries grow rapidly all over the world and one of largest generic exporter in world where as, US being the major destination for export. Thus, the proper validated regulation is required for manufacturing generic drugs in India and US which requires proper symbiotic relation between India and US. Some amendments are warranted in Hatch Waxman Act 1984 for developing generic drug in better way, where as re-election of Barack Obama in US provides positive increase in generic market as his government extending health care insurance for additional 30 million Americans in the health care ambit, creating increased demand for generics.

REFERENCES

Pharma Regulations for Generic Drug Products in India and US: Case Studies and Future Prospectives

Suryakanta Swain*, Ankita Dey, Chinam Niranjan Patra and Muddana Eswara Bhanoji Rao

Roland Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Berhampur, Odisha, India

Suryakanta Swain
Assistant Professor
Roland Institute of Pharmaceutical Sciences
Department of Pharmaceutics
P.O.: Khodasingi, Berhampur-7600 10, Odisha, India
Tel: 91-943-803-8643; 909-037-4275
E-mail: swain_suryakant@yahoo.co.in

Citation: Swain S, Dey A, Patra CN, Bhanoji Rao ME (2014) Pharma Regulations for Generic Drug Products in India and US: Case Studies and Future Prospectives. Pharmaceut Reg Affairs 3:119. doi: 10.4172/2167-7689.1000119

Suryakanta Swain
Asst. Professor-cum-Placement Officer
Department of Pharmaceutics at Roland Institute of Pharmaceutical Sciences

Biography
Dr. Suryakanta Swain was born on 8th June 1980 in Debendrapur, Balasore, Odisha (INDIA). After completing his B. Pharm with 79.37% from Berhampur University, Odisha, India and join in to M. Pharm (Pharmaceutics) by qualifying GATE and N.I.P.E.R with All India entrance examinations with C.G.P.A 8.89 from Biju Patnaik University of Technology, Rourkela, Odisha, India. He is completed his Ph.D in Pharmacy from Berhampur University on 09.12.2013. He started his career as a Research Trainee Executive in Formulation Research & Development in Medley Pharmaceuticals Pvt. Ltd, Daman, India. Presently he is working as Asst. Professor-cum-Placement Officer in Department of Pharmaceutics at Roland Institute of Pharmaceutical Sciences, Berhampur, Odisha, India. So far he has published thirty articles of reputed national & international journals with high indexing or impact factor. He has edited one book, authored four books & one book chapter an international level. He has filled One Indian patent. He has permanent Editor, Advisary, Editorial board members and reviewers in more than 15 national & international journals.
Research Interest
Mucoadhesive DDS, Transdermal DDS, Liposomal DDS, Selfemulsifying DDS, Micro and Nanoparticulate DDS, Gastro-Intestinal DDS, Colon Specific DDS and Controlled DDS.
Publications
Solid Lipid Nanoparticle: An Overview
Suryakanta Swain and Sitty Manohar Babu
Editorial: Pharmaceut Reg Affairs 2015, 4: e154
doi: 10.4172/2167-7689.1000e154
Pharmaceutical Impurities and Degradation Products: An Overview
Prafulla Kumar Sahu, Suryakanta Swain and Manohar Babu S
Editorial: Pharmaceut Reg Affairs 2015, 4: e146
doi: 10.4172/2167-7689.1000e146
Impact of Pharmacovigilance in Healthcare System: Regulatory Perspective
Suryakanta Swain and Chinam Niranjan Patra
Editorial: Pharmaceut Reg Affairs 2014, 3: e143
doi: 10.4172/2167-7689.1000e143
Bio-Relevant and Bioequivalence Studies: An Overview
Suryakanta Swain and Nerella Nagadivya
Editorial: Pharmaceut Reg Affairs 2014, 3: e140
doi: 10.4172/2167-7689.1000e140

Roland Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Berhampur, Odisha, India

 

/////////Abbreviated new drug application approvals, Cases and incidents, Pharma regulations, Recent patents, Roland Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Berhampur, Odisha, India


Filed under: Regulatory Tagged: Abbreviated new drug application approvals, Berhampur, Cases and incidents, Department of Pharmaceutics, INDIA, Odisha, Pharma regulations, Recent patents, Roland Institute of Pharmaceutical Sciences

Cutting Edge of Pharmaceutical Nanotechnology

$
0
0

Nanoscience is the engineering of functional systems at the molecular scale. This covers both current work and concepts that are more advanced. In its original sense, nanotechnology refers to the projected ability to construct items from the bottom up, using techniques and tools being developed today to make complete, high performance products. Some researches and findings in the field of Nanoscience are selected and expended here: “Fabrication of Novel Poly (ethylene terephthalate)/TiO2 Nanofibers by Electrospinning and their Photocatalytic Activity” reports on functional nanocomposites PET/TiO2 nanofibers membranes prepared via simple electrospinning and hydrothermal processing, involving preparation of titania precursor sol solution, electrospinning the homogeneous mixture of PET solution and sol solution, and in-situ growth of nanoscale TiO2 within PET nanofibers in hot water.

“Oxidation of glyoxal to glyoxalic acid by Prepared Nano-Au/C catalysts” describes that Nano-Au/C catalysts were obtained by loading the gold nanoparticles which were prepared by photochemical reduction method to the activated carbon, and were used for the catalytic oxidation reaction of glyoxal into glyoxylic acid.

“Preparation of the Al-CNT (Carbon Nanotubes) Compound Material by High Energy Milling” using high energy ball milling (HEM), researched the technology of preparation of Al-CNT compound material.

“Theoretical Prediction of Tensile Behavior of Single-Walled Carbon Nanotubes” establishes a link between molecular and continuum mechanics based on the Morse potential function.

In the paper “Research on the stress-relaxation characteristics of cancer cells based on Atomic Force Microscope”, the AFM indentation experiments are carried out on two different transferring characteristic cancer cells (Anip-937 and AGZY-83a) under physiological conditions using the expansion of atomic force microscope (AFM) indentation and the improvement of Hertz model.

“Application of Nanoscale Zero-valent Iron (nZVI) to Enhance Microbial Reductive Dechlorination of TCE: A Feasibility Study” evaluates the feasibility of nanoscale zero-valent iron (nZVI) application to enhance microbial reductive dechlorination of trichloroethylene (TCE).

“Hydrothermal Processing-Assisted Synthesis of Nanocrystalline YFeO3 and its Visible-Light Photocatalytic Activity” finds that the single phase YFeO3 can be obtained through the calcination of hydrothermally processed YFeO3 precursors at 800°C, and the resulting product has a spherical shape and uniform size distribution.

“Preparation and exothermic characterization of HTPB-coated aluminum nano-powders prepared by laser-induction hybrid heating” calculates the temperature distribution of aluminum with the heating time and the distance from the crucible centre based on the ANSYS software.

“Application Thinking of Nanotechnology in Acupuncture” discusses the application of nanotechnology methods for the researches on meridians of Chinese medicine, acupoint catgut embedding therapy (ACET) and therapeutic mechanism in acupuncture field.

“The Research of Conjunction Calculated Relationships between Proteins with Gold Nanoparticles” researches the conjunction calculated relationship between proteins and gold nanoparticles.

“Engineered nanoparticles as precise drug delivery systems”- Nanoparticles, an evolvement of nanotechnology, are increasingly considered as a potential candidate to carry therapeutic agents safely into a targeted compartment in an organ, particular tissue or cell.

“Dendrimers: emerging polymers for drug-delivery systems”, the unique properties associated with these dendrimers such as uniform size, high degree of branching, water solubility, multivalency, welldefined molecular weight and available internal cavities make them attractive for biological and drug-delivery applications.

“Strategies for in vivo siRNA delivery in cancer”- As a research tool, siRNA has proven to be highly effective in silencing specific genes and modulating intracellular signaling pathways.

“Rapid delivery of drug carriers propelled and navigated by catalytic nanoshuttles”- nanoshuttles’ navigation ability is illustrated by the transport of the drug carriers through a microchannel from the pick-up to the release microwell. Such ability of nanomotors to rapidly deliver drug-loaded polymeric particles and liposomes to their target destination represents a novel approach towards transporting drug carriers in a target-specific manner.

“Multigram-scale fabrication of monodisperse conducting polymer and magnetic carbon nanoparticles” is an emerging tool for cutting edge nanotechnology approach.

Cutting Edge of Pharmaceutical Nanotechnology

Suryakanta Swain*

Suryakanta Swain
Roland Institute of Pharmaceutical Sciences
Department of Pharmaceutics
Khodasinghi, Berhampur-760 010 (Ganjam)
Odisha, India
Email: swain_suryakant@yahoo.co.in

Roland Institute of Pharmaceutical Sciences, Department of Pharmaceutics, IndiaCitation: Swain S (2012) Cutting Edge of Pharmaceutical Nanotechnology. Pharmaceut Reg Affairs 1:e110. doi: 10.4172/2167-7689.1000e110

http://www.omicsgroup.org/journals/cutting-edge-of-pharmaceutical-nanotechnology-2167-7689.1000e110.php?aid=8206

/////////////Cutting Edge,  Pharmaceutical Nanotechnology


Filed under: Nanotechnology, SYNTHESIS Tagged: Cutting Edge, Pharmaceutical Nanotechnology

Israeli scientists turn Nano science fiction into fact

$
0
0

 

Find out how Israeli scientists are manipulating the tiniest parts of matter to make life better for millions.

Think of a tiny robot transporting drugs to a cancer cell in your body. An artificial retina to restore lost sight. Self-cleaning windows and bullet-proof fabrics.

It’s all possible today with nanotechnology from Israel.

Tune into ISRAEL21c’s TLV1 radio show for a fascinating discussion of how Israeli scientists are turning science fiction into fact. Guests include Nava Swersky Sofer, founder and co-chair of NanoIsrael; Prof. Uriel Levy, head of the Nanotechnology Institute at the Hebrew University of Jerusalem; and Prof. Uri Sivan, one of the Technion’s leading nanotechnology experts……….http://www.israel21c.org/israeli-scientists-turn-science-fiction-into-fact-audio/

INNI

http://www.nanoisrael.org/

NanoIsrael 2016

About the INNI mission

President Shimon Peres holding nanotechnology priz

The INNI Board of Directors is appointed by The Chief Scientist in the Ministry of Economy.  The INNI BoD operates out of The MAGNET Program at the Office of the Chief Scientist.

The mission of INNI — the Israel National Nanotechnology Initiative is to make nanotechnology the next wave of successful industry in Israel by creating an engine for global leadership.

A primary task for INNI is to promote fruitful collaboration between Israeli and global nanotechnology stakeholders, particularly for projects that lead to continuing success in academia and industry.
To achieve this task, INNI activities include:
  • Establishing a national policy of resources for nanotechnology, with the aim of faster commercialization.
  • Long-range nanotechnology programs for scientific research and technology development in academia and industry, and promoting development of world-class infrastructure in Israel to support them.
  • Leading in the creation of projects that promote agreed national priorities; allocate their budgets and review development progress.
  • Actively seeking funding resources from public and private sources in order to implement the selected projects.
  • Promoting development of innovative local nanotechnology industries which will strongly impact Israeli economic growth and benefit investors.
  • Encouraging Academia and Industry cooperation with public access to a national database of Israel’s nanotechnology researchers and industry.  Effective access to information about Israel’s researchers and companies accelerates cooperation on R&D projects and on innovative new products. Israel’s nanotechnology National Database may be accessed here or from the link in the INNI website upper navigation menu.
Key to development of nanotechnology-based industry in Israel is promotion of academia–industry collaboration. INNI in collaboration with d&a Visual Insights has created a unique graphic mapping engine of Israel’s nanotechnology academia and industry. It is an extensive database of companies and researchers which is accessed with an integrated text and graphical mapping search engine. It is the ideal starting point to locate researchers and companies with the unique knowledge and skill sets for cooperation in research projects and the transfer of technology for innovative products and establishing new nanotechnology enterprises.

Sivan Uri .

Room 611, Lidow Building

Physics

Russell Berrie Nanotechnology InstituteTechnion - Israel Institute of Technology

Nano Area: Nano Electronics, Nano Materials & Nano Particles, Nanobiotechnology & Nanomedicine

Phone: +972-4-8293452

Fax: +972-4-8292418

Email: phsivan@tx.technion.ac.il

Personal website

Main

Ph.D.: Tel Aviv University 1988
M.Sc.: Physics, Tel Aviv University 1984
B.Sc.: Physics and Mathematics, Tel Aviv University 1982

Main Nano Field:

Selection of antibodies and peptides against electronic materials, electrical control over bioreactions, bioassembly of electronic devices.

Bertoldo Badler Chair in Physics

Former director of the Russell Berrie Nanotechnology Institute

Head of Ben and Esther Rosenbloom Center of Excellence in Nanoelectronics by Biotechnology

……………………………..
Uriel Levy
Prof. Uriel Levy named outstanding young researcher at the Hebrew University

 Prof. Uriel Levy of the Hebrew University of Jerusalem has received the Hebrew University President’s Prize as the Outstanding Young Researcher for 2010-11. The prize is awarded in memory of Prof. Yoram Ben-Porath, former president and rector of the Hebrew University.Hebrew University President Prof. Menahem Ben-Sasson said that the prize was being awarded to Prof. Levy “for his impressive list of scientific articles, for his creativity, and for his groundbreaking innovations.”

Prof. Levy is a member of the applied physics department at the Benin School of Computer Science and Engineering and is a renowned researcher in nanophotonics He is a member of the Harvey M. Kruger Family Center for Nanoscience and Nanotechnology at the Hebrew University.

A graduate of the Technion in physics and materials engineering, he subsequently earned a Ph.D. in electro-optics at Tel Aviv University in 2002. He then was awarded a Rothschild Fellowship for post-doctoral work at the University of California, San Diego, which he completed in 2006.

Prof. Levy has published until now 55 scientific articles and has had a number of his research discoveries patented.

Downloadable File: PresidentsPrize2010.doc

The NanoOpto group is affiliated with the Applied Physics Department at the Hebrew University of Jerusalem, Israel. Our research is mainly focused on Silicon Photonics, Polarization Optics, Plasmonics and Opto-Fluidics.

Our group  host SPP7 in Jerusalem from 31 of may till the 5 of June 2015:

Uriel Levy at ulevy@cc.huji.ac.il

Research highlights:

Silicon Photonics
In this work we study the optimization of interleaved Mach-Zehnder silicon carrier depletion electro-optic modulator. Following the simulation results we demonstrate a phase shifter with the lowest figure of merit (modulation efficiency multiplied by the loss per unit length) 6.7V-dB. This result was achieved by reducing the junction width to 200 nm along the phase-shifter and optimizing the doping levels of the PN junction for operation in nearly fully depleted mode. The demonstrated low FOM is the result of both low VπL of ~0.78 Vcm (at reverse bias of 1V), and low free carrier loss (~6.6 dB/cm for zero bias). Our simulation results indicate that additional improvement in performance may be achieved by further reducing the junction width followed by increasing the doping levels. (read more)

Light vapor interactions on a chip
Alkali vapours, such as rubidium, are being used extensively in many important fields of research. Recently, there is a growing effort towards miniaturizing traditional centimetre-size vapour cells. Owing to the significant reduction in device dimensions, light– matter interactions are greatly enhanced, enabling new functionalities due to the low power threshold needed for nonlinear interactions. Here, we construct an efficient and flexible platform for tailored light–vapour interactions on a chip, and demonstrate efficient interaction of the electromagnetic guided mode with absorption saturation at powers in the nanowatt regime. (read more)

Active Silicon Plasmonics
In this work, we experimentally demonstrate an on-chip nanoscale silicon surface-plasmon Schottky photodetector based on internal photoemission process and operating at telecom wavelengths. The responsivity of the nanodetector to be 0.25 and 13.3mA/W for incident optical wavelengths of 1.55 and 1.31 μm, respectively. The presented device can be integrated with other nanophotonic and nanoplasmonic structures for the realization of monolithic opto-electronic circuitry on-chip. (read more)

Plasmonics
Planar plasmonic devices are becoming attractive for myriad applications. Mitigating the challenges of using plasmonics in on-chip configurations requires precise control over the properties of plasmonic modes, in particular their shape and size. Here we achieve this goal by demonstrating a planar plasmonic graded index lens focusing surface plasmons propagating along the device. Focusing and divergence of surface plasmons is demonstrated experimentally. The demonstrated approach can be used for manipulating the propagation of surface plasmons, e.g. for beam steering, splitting, cloaking, mode matching and beam shaping applications (read more)

Metamaterials
The interaction of an incident plane wave with a metamaterial periodic structure consisting of alternating layers of positive and negative refractive index with average zero refractive index is studied. We show that the existence of very narrow resonance peaks for which giant absorption – 50% at layer thickness of 1% of the incident wavelength – is exhibited. Maximum absorption is obtained at a specific layer thickness satisfying the critical coupling condition. This phenomenon is explained by the Rayleigh anomaly and excitation of Fabry Perot modes. (read more)

Plasmonics
Great hopes rest on surface plasmon polaritons’ (SPPs) potential to bring new functionalities and applications into various branches of optics. In this work, we demonstrate a pin cushion structure capable of coupling light from free space into SPPs, split them based on the polarization content of the illuminating beam of light, and focus them into small spots. We also show that for a circularly or randomly polarized light, four focal spots will be generated at the center of each quarter circle comprising the pin cushion device. Furthermore, following the relation between the relative intensity of the obtained four focal spots and the relative position of the illuminating beam with respect to the structure, we propose and demonstrate the potential use of our structure as a miniaturized plasmonic version of the well-known four quadrant detector. (read more)

Silicon Photonics
We demonstrate a nanoscale mode selector supporting the propagation of the first antisymmetric mode of a silicon waveguide. The mode selector is based on embedding a short section of PhC into the waveguide. On the basis of the difference in k-vector distribution between orthogonal waveguide modes, the PhC can be designed to have a band gap for the fundamental mode, while allowing the transmission of the first antisymmetric mode. The device was tested by directly measuring the modal content before and after the PhC section using a near field scanning optical microscope. Extinction ratio was estimated to be ~23 dB. Finally, we provide numerical simulations demonstrating strong coupling of the antisymmetric mode to metallic nanotips. On the basis of the results, we believe that the mode selector may become an important building block in the realization of on chip nanofocusing devices. (read more)

Plasmonics
We experimentally demonstrate the focusing of surface plasmon polaritons by a plasmonic lens illuminated with radially polarized light . The field distribution is characterized by near-field scanning optical microscope. A sharp focal spot corresponding to a zero-order Bessel function is observed. For comparison, the plasmonic lens is also measured with linearly polarized light illumination, resulting in two separated lobes. Finally, we verify that the focal spot maintains its width along the optical axis of the plasmonic lens. The results demonstrate the advantage of using radially polarized light for nanofocusing applications involving surface plasmon polaritons. (read more)
Hebrew University of Jerusalem
Map of hebrew university of jerusalem
////////

Filed under: Nanotechnology Tagged: ISRAEL, Nanotechnology

The new APIC Guidance on Handling of Insoluble Matter and Foreign Particles in the Manufacture of Active Pharmaceutical Ingredients

$
0
0

The new APIC Guidance on Handling of Insoluble Matter and Foreign Particles in the Manufacture of Active Pharmaceutical Ingredients

The occurrence of foreign particles in the manufacture of active pharmaceutical ingredients is always undesirable. For the responsible QA departments it involves an increased effort as concerns the search for the root causes and for CAPA measures. A new APIC Guidance offers concrete recommendations for the GMP compliant handling of foreign particles in APIs, intermediates and raw materials.

Foreign particles in APIs or medicinal preparations are undesirable and sometimes lead to a recall of the batches concerned. Depending on the type of particles their presence in active pharmaceutical ingredients may be harmless; in many cases they are inevitable. In any case the manufacturer must find an adequate way how to handle those impurities visible to the human eye. The search for a guideline or another official document in the relevant regulations is in vain. Visible particles or fibres are only mentioned in the USP chapter <790>, in chapter 2.9.20 of the European Pharmacopoeia as well as in the United States Food, Drug and Cosmetic Act (FD&C Act).

In order to remedy this lack of guidance or recommendations a group of experts within APIC has drawn up a guidance on the handling of foreign particles. This “Guidance on Handling of insoluble Matter and Foreign Particles in APIs” describes in detail

  • the types of particles which can often occur during the manufacture of APIs, API intermediates and raw materials (including packaging materials),
  • suitable measures to minimize the presence of particles or to remove them,
  • how to determine them analytically
  • how to identify the source and to carry out subsequent CAPA measures and an adequate risk management.

This APIC guidance offers valuable assistance for all API manufacturers that are confronted with the problem of the occurrence of foreign particles in their products, intermediates or raw materials. The implementation of the very concrete and practicable recommendations in this guidance offers also valuable supporting arguments for GMP inspections or audits and can help to avoid unpleasant surprises.

http://www.gmp-compliance.org/enews_05022_The-new-APIC-Guidance-on-Handling-of-Insoluble-Matter-and-Foreign-Particles-in-the-Manufacture-of-Active-Pharmaceutical-Ingredients_9300,S-WKS_n.html

 

 

 

///////APIC Guidance,   Handling of Insoluble Matter and Foreign Particles, Manufacture, Active Pharmaceutical Ingredients


Filed under: Regulatory Tagged: active pharmaceutical ingredients, APIC Guidance, Handling of Insoluble Matter and Foreign Particles, MANUFACTURE

Finally published: new Annex 16 on QP Certification and Batch Release

$
0
0

 

Finally published: new Annex 16 on QP Certification and Batch Release

The European Commission finally has published the new EU-GMP Guideline Annex 16 “Certification by a Qualified Person and Batch Release“.

The European Commission has published the final version of the revised EU-GMP Guideline Annex 16 “Certification by a Qualified Person and Batch Release”. Deadline for coming into operation is 15 April 2016.

As one important topic, it has been pointed out that the major task of a Qualified Person (QP) is the certification of a batch for its release. In this context, the QP must personally ensure the responsibilities listed in chapter 1.6 are fulfilled.  In chapter 1.7 a lot of additional responsibilities are listed which need to be secured by the QP. The work can be delegated and the QP can rely on the respective Quality Management Systems. However “the QP should have on-going assurance that this reliance is well founded” (1.7). Amongst these twenty-one tasks are for example:

  • Starting materials comply and the supply chain is secured, including GMP assessments by third parties
  • The necessary audits have been performed and the audit reports are available
  • Manufacturing and testing performance are compliant with the MA
  • Manufacturing and testing processes are validated
  • Changes have been evaluated and investigations completed

It is important to mention in this context that “the ultimate responsibility for the performance of an authorised medicinal product over its lifetime; its safety, quality and efficacy lies with the marketing authorisation holder (MAH). However “the QP is responsible for ensuring that each individual batch has been manufactured and checked in compliance with laws in force (…), in accordance with the requirements of the marketing authorisation (MA) and with Good Manufacturing Practice (GMP)” (see General Principles).

In the case that the QP has to rely on the correct functioning of the quality management system of other sites, the QP “should ensure that a written final assessment and approval of third party audit reports has been made”. The QP should also “be aware of the outcome of an audit with critical impact on the product quality before certifying the relevant batches.”

Another important section clarifies the role of the QP when it comes to deviations, implementing main features of the EMA Position Paper on QP Discretion (which was issued in February 2006 and updated January 2008). Chapter 3 of the draft describes the “handling of unexpected deviations”. A batch with an unexpected deviation from details contained within the Marketing Authorisation and/or GMP may be certified if a risk assessment is performed, evaluating a “potential impact of the deviation on quality, safety or efficacy of the batch(es) concerned and conclusion that the impact is negligible.” Depending on the outcome of the investigation and the root cause, the submission of a variation to the MA for the continued manufacture of the product might be required.

During the consultation phase, stakeholders expressed their concerns regarding the sampling of imported products. Now the new annex is clear on this: “Samples may either be taken after arrival in the EU, or be taken at the manufacturing site in the third country in accordance with a technically justified approach which is documented within the company’s quality system. (…) Any samples taken outside the EU should be shipped under equivalent transport conditions as the batch that they represent.”

The new annex is rather short on other importation requirements. These requirements will probably be defined in the new Annex 21

http://www.gmp-compliance.org/enews_05058_Finally-published-new-Annex-16-on-QP-Certification-and-Batch-Release_9336,15099,15138,Z-QAMPP_n.html



 

 

 

 

.////////////published, new Annex 16, QP Certification and Batch Release


Filed under: Regulatory Tagged: new Annex 16, published, QP Certification and Batch Release
Viewing all 1640 articles
Browse latest View live