New Drug Approvals

Home » 2014 » February (Page 6)

Monthly Archives: February 2014

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,811,312 hits

Flag and hits

Flag Counter

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

FDA Breakthrough Therapy Designation: 32 And Counting


Orphan Druganaut Blog's avatarOrphan Druganaut Blog

On February 3rd, GlaxoSmithKline (GSK) announces that Promacta (US)/Revolade (Europe) (Eltrombopag) receives the coveted FDA Breakthrough Therapy Designation (BTD) for cytopenias in patients with Severe Aplastic Anemia (SAA), who have had insufficient response to Immunosuppressive Therapy (IST). The drug is not approved or licensed anywhere in the world for this indication.

SAA is a rare disorder where the bone marrow fails to make enough new blood cells. There are currently no therapies approved for this indication. About forty percent (40%) of patients who do not respond to initial IST die within 5 years of diagnosis.

Regulatory Actions

•   Receives FDA ODD in November 2013 for Aplastic Anemia

•   Receives FDA BTD in February 2014 for Aplastic Anemia

•   Receives FDA ODD in May 2008 & FDA approval in November 2008 for Idiopathic Thrombocytopenia Purpura.

This is the 32nd BTD that is announced by a sponsor company since…

View original post 150 more words

SUMATRIPTAN …Avanir files new drug application for migraine drug


SUMATRIPTAN, GR-43175

1-[3-(2-dimethylaminoethyl)-1H-indol-5-yl]- N-methyl-methanesulfonamide

3-[2-(Dimethylamino)ethyl]-N-methyl-1H-indole-5-methanesulfonamide

Formula C14H21N3O2S 
Mol. mass 295.402 g/mol
CAS number 103628-46-2 
 
Melting point: mp 169-171°
Therap-Cat: Antimigraine.
Keywords: Antimigraine; Serotonin Receptor Agonist.
NDA 020626,GSK, IMITREX, 1997

Avanir Pharmaceuticals has filed a new drug application (NDA) with the US Food and Drug Administration (FDA) for approval of its new breath-powered investigational drug-device combination product, ‘AVP-825’, for the acute treatment of migraines.  click on title  Avanir files new drug application for migraine drug 

Sumatriptan moleculeSUMATRIPTAN

SUMATRIPTAN SUCCINATE

CAS Registry Number:
103628-48-4 ((1:1) salt), 103628-47-3 ((2:1) salt), 103628-46-2 (free base)
GlaxoSmithKline (Originator), Atrix (Formulation), Nastech (Formulation), NovaDel Pharma (Formulation)
Manufacturers’ Codes: GR-43175C
Trademarks: Imigran (GSK); Imitrex (GSK); Imiject (GSK)
Molecular Formula: C14H21N3O2S.C4H6O4
Molecular Weight: 413.49
Percent Composition: C 52.28%, H 6.58%, N 10.16%, O 23.22%, S 7.75%
Properties: mp 165-166°.
Melting point: mp 165-166°
Launched-1991, Acute Attacks of Migraine, Treatment of, Analgesic and Anesthetic Drugs, Antimigraine Drugs, 5-HT1B Agonists, 5-HT1D Agonists

AVP-825 is an investigational drug-device combination product consisting of low-dose sumatriptan powder delivered intranasally utilizing a novel Breath Powered delivery technology. If approved, AVP-825 would be the first and only fast-acting, dry-powder intranasal form of sumatriptan for the treatment of migraine.

The Breath Powered delivery technology is activated by user’s breath to propel medications deep into the nasal cavity where absorption is more efficient and consistent than through most other routes. A user exhales into the device, automatically closing the soft palate and sealing off the nasal cavity completely. Through a sealing nosepiece placed into the nostril, the exhaled breath carries medication from the device directly into one side of the nose. Narrow nasal passages are gently expanded and medication is dispersed deep into the nasal cavity reaching areas where it can be rapidly absorbed. As the medication is delivered, the air flows around to the opposite side of the nasal cavity and exits through the other nostril. Closure of the soft palate helps prevent swallowing or inhalation of sumatriptan powder into the lungs.

Canada 2469019 APPROVED 2005-09-13 EXP 2022-12-04
United States 6135979                  1997-03-21        2017-03-21
United States 5705520                  1994-12-10        2011-12-10
Canada 2098302                  2001-10-16        2011-12-10
Patent No PatentExpiry use code
5307953 Dec 2, 2012  
5307953*PED Jun 2, 2013  
5554639 Sep 10, 2013 U-232…METHOD OF TREATING MIGRAINE
5554639*PED Mar 10, 2014

Sumatriptan is a synthetic drug belonging to the triptan class, used for the treatment of migraine headaches. Structurally, it is an analog of the naturally occurring neuro-active alkaloids dimethyltryptamine (DMT), bufotenine, and 5-methoxy-dimethyltryptamine, with an N-methyl sulfonamidomethyl- group at position C-5 on the indole ring.[1]

Sumatriptan is produced and marketed by various drug manufacturers with many different trade names such as Sumatriptan, Imitrex, Treximet, Imigran, Imigran recovery.

Large doses of sumatriptan can cause sulfhemoglobinemia, a rare condition in which the blood changes from red to greenish-black, due to the integration of sulfur into the hemoglobin molecule.[2] If sumatriptan is discontinued, the condition reverses within a few weeks.

Serious cardiac events, including some that have been fatal, have occurred following the use of sumatriptan injection or tablets. Events reported have included coronary artery vasospasm, transient myocardial ischemia, myocardial infarctionventricular tachycardia, and ventricular fibrillation.

The most common side-effects[3] reported by at least 2% of patients in controlled trials of sumatriptan (25, 50, and 100 mg tablets) for migraine are atypical sensations (paresthesias and warm/cold sensations) reported by 4% in the placebo group and 5–6% in the sumatriptan groups, pain and other pressure sensations (including chest pain) reported by 4% in the placebo group and 6–8% in the sumatriptan groups, neurological events (vertigo) reported by less than 1% in the placebo group and less than 1% to 2% in the sumatriptan groups. Malaise/fatigue occurred in less than 1% of the placebo group and 2–3% of the sumatriptan groups. Sleep disturbance occurred in less than 1% in the placebo group to 2% in the sumatriptan group.

SUMATRIPTAN

Sumatriptan is structurally similar to serotonin (5HT), and is a 5-HT (types 5-HT1D and 5-HT1B[4]agonist. The specific receptor subtypes it activates are present on the cranial arteries and veins. Acting as an agonist at these receptors, sumatriptan reduces the vascular inflammation associated with migraines.

The specific receptor subtype it activates is present in the cranial and basilar arteries. Activation of these receptors causes vasoconstriction of those dilated arteries. Sumatriptan is also shown to decrease the activity of the trigeminal nerve, which, it is presumed, accounts for sumatriptan’s efficacy in treating cluster headaches. The injectable form of the drug has been shown to abort a cluster headache within fifteen minutes in 96% of cases.[5]

 

Sumatriptan is administered in several forms; tablets, subcutaneous injection, and nasal spray. Oral administration (as succinate) suffers from poorbioavailability, partly due to presystemic metabolism—some of it gets broken down in the stomach and bloodstream before it reaches the target arteries. A new rapid-release tablet formulation has the same bioavailability, but the maximum concentration is achieved on average 10–15 minutes earlier. When injected, sumatriptan is faster-acting (usually within 10 minutes), but the effect lasts for a shorter time. Sumatriptan is metabolised primarily by monoamine oxidase A into an indole acetic acid analogue, part of which is further conjugated with glucuronic acid. These metabolites are excreted in the urine and bile. Only about 3% of the active drug may be recovered unchanged.

There is no simple, direct relationship between sumatriptan concentration (pharmacokinetics) per se in the blood and its anti-migraine effect (pharmacodynamics). This paradox has, to some extent, been resolved by comparing the rates of absorption of the various sumatriptan formulations, rather than the absolute amounts of drug that they deliver.[6][7]

 

Sumatriptan was the first clinically available triptan (in 1991). In the United States, it is available only by medical prescription. However, it can be bought over the counter in the UK and Sweden in 50 mg dosage. Several dosage forms for sumatriptan have been approved, including tablets, solution for injection, and nasal inhalers.

On April 15, 2008, the US FDA approved Treximet, a combination of sumatriptan and naproxen, an NSAID.[8] This combination has shown a benefit over either medicine used separately.[9]

In July 2009, the US FDA approved a single-use jet injector formulation of sumatriptan. The device delivers a subcutaneous injection of 6 mg sumatriptan, without the use of a needle.Autoinjectors with needles have been previously available in Europe and North America for several years.[10]

Phase III studies with a iontophoretic transdermal patch (Zelrix/Zecuity) started in July 2008.[11] This patch uses low voltage controlled by a pre-programmed microchip to deliver a single dose of sumatriptan through the skin within 30 minutes.[12][13]Zecuity was approved by the US FDA in January 2013.[14]

 

Sumatriptan vials 100 5509

On November 6, 2008, Par Pharmaceutical announced that it would begin shipping generic versions of sumatriptan injection (sumatriptan succinate injection) 4 mg and 6 mg starter kits and 4 mg and 6 mg pre-filled syringe cartridges to the trade immediately. In addition, Par anticipates launching the 6 mg vials early in 2009.[15]

Mylan Laboratories Inc., Ranbaxy, Sandoz, Dr. Reddy’s Pharmaceuticals and other companies have received FDA approval for generic versions of Imitrex tablets in 25-, 50-, and 100-milligram doses since 2009. The drug is available in U.S. and European markets, since Glaxo’s patent protections have expired in those jurisdictions. However, sales of a generic delivered via nasal spray are still restricted in the United States.

See also Sumavel DosePro (above).[10]

Chemistry

hydrogenation of nitrile with pd/c in presence of dimethyl amine

…………………

Sumatriptan synth.png

U.S. Patent 4,785,016

The diazotation of 4-amino-N-methylbenzenemethanesulfonamide (I) with NaNO2-HCl followed by reduction with SnCl2 gives the 4-hydrazino compound (II), which is condensed with (phenylthio)acetaldehyde (III) in ethanol yielding the ethylideneamino compound (IV). The cyclization of (IV) with HCl in ethanol affords N-methyl-3-(phenylthio)-1H-indole-5-methansulfonamide (V), which is desulfurized with RaNi in refluxing ethanol-water to give N-methyl-1H-indole-5-methanesulfonamide (VI). The reaction of (VI) with oxalyl chloride and dimethylamine yields the oxalyl derivative (VII), which is finally reduced with LiAlH4 in refluxing THF.

The condensation of hydrazine (II) with 4,4-dimethoxy-N,N-dimethylbutylamine (VIII) by means of HCl in water gives the butylidenehydrazino compound (IX), which is cyclized with polyphosphate ester (PPE) in CHCl3.

……………………

Beilstein J. Org. Chem. 2011, 7, 442–495.

http://www.beilstein-journals.org/bjoc/single/articleFullText.htm?publicId=1860-5397-7-57#S9

ref are below article

Indoles

The neuroamine transmitter serotonin contains an indole ring, so it is not surprising that indoles are a recurring theme in many drugs affecting central nervous system (CNS) function including antidepressants, antipsychotics, anxiolytics and antimigraine drugs, as well as psychedelic agents. Indole is also one of the best represented heterocyclic motifs present in the top selling pharmaceuticals, being found in eight of the top 200 drugs, with five of these belonging to the triptan family of antimigraine treatments. The classical Fischer indole synthesis is usually reported as one of the first choice routes to prepare these scaffolds. Drugs such as GSK’s serotonin receptor modulators sumatriptan (49, Imitrex) and zolmitriptan (50, Zomig) use the Fischer indole synthesis at a late stage in order to form the desired compound albeit in only low to moderate yields (Scheme 9).

[1860-5397-7-57-i9]
Scheme 9: Key steps in the syntheses of sumatriptan and zolmitriptan.

However, in sumatriptan the indole product resulting from the Fischer synthesis can still react further which leads to the formation of by-products and significantly reduced yields. One way to minimise this was to protect the nitrogen of the sulfonamide group prior to indole formation [11]. This leads not only to an increased yield in the indole forming step (to 50%) but also facilitates chromatographic purification. The dimethylamino group can be present from the beginning of the synthesis or can be introduced via displacement of chloride or reduction of a cyano moiety. Alternatively, the dimethyl ethylene amine side chain can be introduced in position 3 via a Friedel–Crafts-type acylation. The resulting acid chloride is transformed in situ to the corresponding amide which on reduction with lithium aluminium hydride affords sumatriptan (Scheme 10) [12].

[1860-5397-7-57-i10]
Scheme 10: Introduction of the N,N-dimethylaminoethyl side chain.

In the standard Fischer indole synthesis a hydrazine, which is most commonly derived from the corresponding diazonium salt, is reacted with a suitable carbonyl compound. Alternatively, the Japp–Klingemann reaction can be used to directly couple the diazonium salt with a β-ketoester to obtain a hydrazone which can then undergo indole ring formation (Scheme 11) [13].

[1860-5397-7-57-i11]
Scheme 11: Japp–Klingemann reaction in the synthesis of sumatriptan.

As can be seen from Scheme 11 the indole 59 prepared via the Japp–Klingemann reaction is substituted at position 2 by an ester group which prevents reaction with electrophiles, thereby reducing the amount of undesired by-products. A simple sequence of hydrolysis and decarboxylation then affords sumatriptan [14].

All the reported methods for the synthesis of sumatriptan begin with the sulfonamide group already present on the aromatic ring and several routes are possible to introduce this functional group. The scalable route to the sulfonamides inevitably involves the preparation of the sulfonyl chloride intermediate which is then trapped with the desired amine. The sulfonyl chloride can also be prepared from the corresponding hemithioacetal 61 by treatment with NCS in wet acetic acid (Scheme 12). This efficient oxidation produces only methanol and formaldehyde as by-products [15].

[1860-5397-7-57-i12]
Scheme 12: Synthesis of the intermediate sulfonyl chlorides 62 and 63.
  1. 11. Pete, B.; Bitter, I.; Szántay, C., Jr.; Schön, I.; Töke, L. Heterocycles 1998, 48, 1139–1149. doi:10.3987/COM-97-8087
  2. 12…Oxford, A. W. Indole Derivative. U.S. Patent 5,037,845, Aug 6, 1991.
  3. 13…Japp, F. R.; Klingemann, F. Chem. Ber. 1887, 20, 2942–2944. doi:10.1002/cber.188702002165
  4. Pete, B.; Bitter, I.; Harsányi, K.; Töke, L. Heterocycles 2000, 53, 665–673. doi:10.3987/COM-99-8815
  5. Kim, D.-W.; Ko, Y. K.; Kim, S. H. Synthesis 1992, 12, 1203–1204. doi:10.1055/s-1992-26333
    [15

 

References for full article

 
  1.  The presence of the sulfonamide group in the molecule does not make sumatriptan a “sulfa drug”, since it does not have any anti-microbial properties.
  2.  “Patient bleeds dark green blood”BBC News. 8 June 2007. Retrieved 6 March 2010.
  3.  Tablets
  4.  Razzaque Z, Heald MA, Pickard JD, et al. (1999). “Vasoconstriction in human isolated middle meningeal arteries: determining the contribution of 5-HT1B- and 5-HT1F-receptor activation”.Br J Clin Pharmacol 47 (1): 75–82. doi:10.1046/j.1365-2125.1999.00851.xPMC 2014192.PMID 10073743.
  5.  Treatment of acute cluster headache with sumatriptan. The Sumatriptan Cluster Headache Study Group. N Engl J Med 1991;325:322-6.
  6.  Fox, A. W. (2004). “Onset of effect of 5-HT1B/1D agonists: a model with pharmacokinetic validation”. Headache 44 (2): 142–147. doi:10.1111/j.1526-4610.2004.04030.x.PMID 14756852edit
  7.  Freidank-Mueschenborn, E.; Fox, A. (2005). “Resolution of concentration-response differences in onset of effect between subcutaneous and oral sumatriptan”. Headache 45 (6): 632–637. doi:10.1111/j.1526-4610.2005.05129a.xPMID 15953294edit
  8.  GSK press release – Treximet (sumatriptan and naproxen sodium) tablets approved by FDA for acute treatment of migraine
  9.  Brandes JL, Kudrow D, Stark SR, et al. (April 2007). “Sumatriptan-naproxen for acute treatment of migraine: a randomized trial”JAMA 297 (13): 1443–54.doi:10.1001/jama.297.13.1443PMID 17405970.
  10.  Brandes, J.; Cady, R.; Freitag, F.; Smith, T.; Chandler, P.; Fox, A.; Linn, L.; Farr, S. (2009). “Needle-free subcutaneous sumatriptan (Sumavel DosePro): bioequivalence and ease of use.”. Headache 49 (10): 1435–1444. doi:10.1111/j.1526-4610.2009.01530.x.PMID 19849720edit
  11.  ClinicalTrials.gov NCT00724815 The Efficacy and Tolerability of NP101 Patch in the Treatment of Acute Migraine (NP101-007)
  12.  SmartRelief -electronically assisted drug delivery (iontophoresis)
  13.  Pierce, M; Marbury, T; O’Neill, C; Siegel, S; Du, W; Sebree, T (2009). “Zelrix: a novel transdermal formulation of sumatriptan”. Headache 49 (6): 817–25. doi:10.1111/j.1526-4610.2009.01437.xPMID 19438727.
  14.  Zecuity Approved by the FDA for the Acute Treatment of Migraine
  15.  “PAR PHARMACEUTICAL BEGINS SHIPMENT OF SUMATRIPTAN INJECTION”Par Pharmaceutical. 2008-11-06. Retrieved 2008-11-25.
  16. Serotonin 5HT1-receptor agonist. Prepn: M. D. Dowle, I. H. Coates, DE 3320521eidem, US 4816470; A. W. Oxford, GB 2162522 (1983, 1989, 1986 all to Glaxo).
  17. Receptor binding studies: P. P. A. Humphrey et al., Br. J. Pharmacol.94, 1123 (1988); P. Schoeffter, D. Hoyer, Arch. Pharmacol. 340, 135 (1989).
  18. LC-MS determn in plasma: J. Oxford, M. S. Lant, J. Chromatogr. 496, 137 (1989).
  19. Clinical evaluations in migraine: A. Doenicke et al., Lancet 1, 1309 (1988);
  20. Subcutaneous Sumatriptan International Study Group, N. Engl. J. Med. 325, 316 (1991); in acute cluster headache: Sumatriptan Cluster Headache Study Group, ibid. 322.
  21. Review of pharmacology and clinical experience: S. J. Peroutka, Headache 30 (Suppl. 2), 554-560 (1990).
  22. Drugs Fut 1989,14(1),35
     
1-4-2012
Noncardiotoxic pharmaceutical compounds
7-9-2010
NON-MUCOADHESIVE FILM DOSAGE FORMS
1-22-2010
Fixed Combination Dosage Forms for the Treatment of Migraine
12-11-2009
ACTIVE AGENT DELIVERY SYSTEMS AND METHODS FOR PROTECTING AND ADMINISTERING ACTIVE AGENTS
10-9-2009
PHARMACEUTICAL COMPOSITIONS COMPRISING A TRIPTAN AND A NONSTEROIDAL ANTI-INFLAMMATORY DRUG
10-9-2009
ACTIVE AGENT DELIVERY SYSTEMS AND METHODS FOR PROTECTING AND ADMINISTERING ACTIVE AGENTS
5-7-2009
Patient controlled drug delivery device
3-20-2009
DEUTERIUM-ENRICHED SUMATRIPTAN
3-13-2009
Rapid dissolution of combination products
2-19-2009
A METHOD OF IDENTIFYING MODULATORS OF CELL SURFACE MEMBRANE RECEPTORS USEFUL IN THE TREATMENT OF DISEASE
4-8-1992
PREPARATION OF INDOLE DERIVATIVES
1-10-1992
PHARMACEUTICAL PREPARATIONS
10-32-1991
SYSTEM AND METHOD FOR DETERMINING THREE-DIMENSIONAL STRUCTURES OF PROTEINS
8-7-1991
Indole derivative
7-4-1990
Pharmaceutical formulations
8-8-1984
Fuel and water homogenizer

Avanir Pharmaceuticals, Inc. is a biopharmaceutical company focused on bringing innovative medicines to patients with central nervous system disorders of high unmet medical need. As part of our commitment, we have extensively invested in our pipeline and are dedicated to advancing medicines that can substantially improve the lives of patients and their loved ones. For more information about Avanir, please visit http://www.avanir.com.

AVANIR® is a trademark or registered trademark of Avanir Pharmaceuticals, Inc. in the United States and other countries. All other trademarks are the property of their respective owners.

Avanir Pharmaceuticals, Inc. licensed exclusive rights for the development and commercialization of AVP-825, a novel Breath Powered intranasal system containing a low-dose sumatriptan powder from OptiNose Inc. of Yardley, PA.

IMITREX Tablets contain sumatriptan succinate, a selective 5-HT1B/1D receptor agonist. Sumatriptan succinate is chemically designated as 3-[2-(dimethylamino)ethyl]-N-methyl-indole- 5-methanesulfonamide succinate (1:1), and it has the following structure:

IMITREX Tablets contain sumatriptan succinate, a selective 5-HT1B/1Dreceptor agonist. Sumatriptan succinate is chemically designated as 3-[2-(dimethylamino)ethyl]-N-methyl-indole- 5-methanesulfonamide succinate (1:1), and it has the following structure:

IMITREX (sumatriptan succinate) Structural Formula Illustration

The empirical formula is C14H21N3O2S•C4H6O4, representing a molecular weight of 413.5. Sumatriptan succinate is a white to off-white powder that is readily soluble in water and in saline.

Each IMITREX Tablet for oral administration contains 35, 70, or 140 mg of sumatriptan succinate equivalent to 25, 50, or 100 mg of sumatriptan, respectively. Each tablet also contains the inactive ingredients croscarmellose sodium, dibasic calcium phosphate, magnesium stearate, microcrystalline cellulose, and sodium bicarbonate. Each 100-mg tablet also contains hypromellose, iron oxide, titanium dioxide, and triacetin.

Topiroxostat 托匹司他 for gout and hyperuricemia


 

str1

 

Figure JPOXMLDOC01-appb-C000001

Topiroxostat

托匹司他

FUJI YAKUHIN  ……..INNOVATOR

Approved in japan PMDA JUNE 28 2013

Xanthine oxidase inhibitor

FOR GOUT AND HYPERURICEMIA

Launched – 2013, Fuji YakuhinSanwa, Topiloric  Uriadec

IUPAC Name: 4-(5-pyridin-4-yl-1H-1,2,4-triazol-3-yl)pyridine-2-carbonitrile

CAS Registry Number: 577778-58-6

4 – [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazol-3 – yl] pyridine-2 – carbonitrile (1)

5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

3-(3-cyano-4-pyridyl)-5-(4-pyridyl)-1,2,4-triazole
Synonyms: 4-(5-PYRIDIN-4-YL-1H-1,2,4-TRIAZOL-3-YL)PYRIDINE-2-CARBONITRILE,

AC1NRB9T, Topiroxostat (JAN/INN),  DB01685, D09786, FYX-051
SK-0910

4-[5-PYRIDIN-4-YL-1H-[1,2,4]TRIAZOL-3-YL]-PYRIDINE-2-CARBONITRILE,

C13H8N6 MF,248.2482 MW

TOPIROXOSTAT

托匹司他

A xanthine oxidase inhibitor used to treat gout and hyperuricemia.

PATENT EXP 3/12/22, US /EU/CN

 

str1

FYX-051, TOPIROXOSTAT is a xanthine oxidase inhibitor. This agent was approved in Japan by Fuji Yakuhin and Sanwa for the treatment of gout and hyperuricemia in 2013 and launched at the same year. In 2009, the compound was licensed to Sanwa by Fuji Yakuhin in Japan for the codevelopment and commercialization of gout.

The number of patients with hyperuricemia in Japan is reported to be 1.25 million and the number suffering from asymptomatic hyperuricemia is estimated to reach several millions. Hyperuricemia is becoming a popular disease.

Presently, hyperuricemia and gout due to hyperuricemia are treated by improving the living environment and administering various drug therapies for each period when an attack of gout is predicted to occur (presymptomatic period), when an attack of gout occurs, or when an attack of gout subsides. That is, preventive therapy is conducted in the presymptomatic period by administering colchicines as well as controlling the daily living environment. When an attack occurs, drug therapy using non-steroidal or steroidal anti-inflammatory agents is mainly conducted. After the attack subsides, patients are given guidance to improve their lifestyle. When improvement is judged insufficient, an assessment is made as to whether hyperuricemia is caused by reduced excretion of uric acid or by increased production of uric acid followed by treatment with drugs, which exhibit a uricosuric effect, such as probenecid and benzbromarone, those which inhibit resorption of uric acid, such as sulfinpyrazone, those which improve acidurea conditions, such as citrates, and xanthine oxidase inhibitors which inhibit production of uric acid, such as allopurinol. Colchicine is said to be able to prevent about 90% of attacks through inhibiting chemotaxis and phagocytosis of leukocytes, such as neutrophils, if administration thereof has been completed within a few hours before the attack. Since colchicine has various adverse effects, however, the use thereof is limited to the minimum and it is therefore difficult to timely administer it.

Accordingly, drug therapies are mainly adopted, but only allopurinol is available for the treatment of a disease caused by increased production of uric acid. However, a metabolite of allopurinol, oxypurinol, tends to accumulate and may cause calculi formation. Furthermore, this drug has been reported to induce adverse events such as rash, a decreased renal function and hepatitis, and it is not easy to administer.

Examples of compounds having xanthine oxidase inhibiting activity that can be used for treating gout caused by increased production of uric acid and that are effective for hyperuricemia and gout due to hyperuricemia have been described in J. Medicinal Chemistry, 1975, Vol. 18, No. 9, pp. 895–900, Japanese Patent Publication No. 49-46622 and Japanese Patent Publication No. 50-24315, which disclose some 1,3,5-substituted or 3,5-substituted 1,2,4-triazole compounds.

4 – [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazol-3 – yl] pyridine-2 – carbonitrile (1) has a xanthine oxidase inhibitory activity and serum uric acid level known as the agent that reduces (Patent Document 1).

Figure JPOXMLDOC01-appb-C000001

The method for producing the compound (1), for example, 2 by Reissert Henze reaction isonicotinic acid methyl N-oxide – is a cyano isonicotinate, and the hydrazide which is then, 4 – this condensed cyanopyridine After obtaining a hydrazide of isonicotinic acid N-oxide (Patent Document 1, Example 12) and method, a cyano group after introduction, 4 by Reissert Henze reaction – method of condensing a cyano pyridine is known (Patent Document 1, Example 39).Further, 4 – as a starting material cyano-N-oxide, a triazole ring after construction (Patent Document 3), Reissert Henze unprotected or (Patent Document 2) to protect the ring condensed with isonicotinic acid hydrazide method of obtaining the compound (1) by introducing a cyano group by the reaction have also been reported.

The crystalline polymorph, yet the same molecule with the same chemical composition, the molecular arrangement in the crystal are different, and are different crystalline states. The pharmaceutical compounds having crystal polymorphism such the differences in physicochemical properties, affect pharmacological activity, solubility, bioavailability, stability and the like are known.Therefore, when the crystal polymorphism is present in a pharmaceutically useful compound, producing compounds of the crystalline form highly useful from polymorphs thereof is desirable.

WO 2003/064410 discloses WO 2005/009991 discloses Japanese Patent Publication No. 2005-41802

However, 4 of the above Patent Document – no description about the presence of crystalline polymorph on carbonitrile – pyridine-2-[yl 5 – (pyridin-4 – yl)-1H-1, 2,4 – – -3 triazol] It has not been, to these manufacturing methods, it is disclosed a method for the purpose of improving the chemical purity and yield, there is no description of the crystallographic plane.

Method of producing topiroxostat, useful for preventing or treating gout; and its intermediates. Picks up from WO2012060308, claiming the use of this topiroxostat for treating renal dysfunction. Along with the concurrently published WO2014017515, claiming crystalline Forms I and II of this compound, which, Fuji Yakuhin, in collaboration with Sanwa Kagaku, has developed and launched for the treatment of gout and hyperuricemia.WO-2014017516

Crystalline Forms I and II of topiroxostat, useful for preventing or treating gout. Along with the concurrently published WO2014017516, claiming a method of producing this compound. Picks up from WO2012060308, claiming a method of treating renal dysfunction using topiroxostat, which Fuji Yakuhin, in collaboration with Sanwa Kagaku, has developed and launched for the treatment of gout and hyperuricemia.WO-2014017515

novel 1,2,4-triazole compounds having an optionally substituted 2-cyanopyridin-4-yl group at 3-position and an optionally substituted aromatic group at 5-position inhibit a xanthine oxidase and are useful for treatment of gout and hyperuricemia, and have previously filed a patent application (Patent Document 1). The compounds can be prepared according to a method shown by the following reaction scheme:

  • Figure imgb0001
    wherein TMS represents trimethylsilyl group and Ar represents an aromatic group
    Although this method can achieve the object in a small-scale production, there were such problems that the process for production of a substituted or unsubstituted 2-cyanoisonicotinic acid hydrazide is complicated, and a reaction solvent must be selected in compliance with the physical property of the product compound in each step, and isolation of a product is required in each step. Furthermore, the overall yield is not sufficiently high, and therefore there is a problem in the production on an industrial scale.
    Patent Document 1: JP-A-2002-017825
    • A compound represented by formula (1) which is a starting material may be prepared by a method described in, for example, JP-A-47-7120, JP-A-61-152661A, JP-A-62-149673, JP-A-2002-528447, or European Patent Application No. 559363 specification. However, it is preferable to prepare compound (1) according to the following reaction scheme:
    • Figure imgb0004

 

SYNTHESIS

 

 

 

 

PATENT

EP1650204A1

    Example 2
      Preparation of 5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole p-toluenesulfonate

    • To the toluene solution obtained in Example 1 (2) was added 2-propanol (700 mL), and the mixture was stirred. To the resulting solution was added p-toluenesulfonic acid monohydrate (151.16 g) and the resulting mixture was stirred for 8 hours at an internal temperature of 80°C. The mixture was brought to room temperature, and the precipitated crystals were taken out and washed with 2-propanol (210 mL×2). The white crystals were dried under reduced pressure at 60°C for 15 hours to give 106.0 g of the captioned compound as white crystals. Subsequently, 90.0 g of the crystals was suspended in a mixture of 2-butanol (49 mL) and water (491 mL) and heated to an internal temperature of 80°C for 1 hour. The internal temperature was brought to room temperature, and the crystals were filtered and washed with a mixture of 2-butanol and water (1:10) (270 mL×3). The resulting crystals were dried under reduced pressure at 60°C for 15 hours to give 75.7 g of the captioned compound in a high purity.
    • 1H―NMR(DMSO-d6)δppm:2.29(s,3H), 7.11 (m,2H), 7.48 (dd, 2H, J=6.48, 1.62Hz) , 8.32-8.35(m, 3H) , 8.57(dd, 1H, J=1.62, 0.81Hz) , 8.94-8.98(m, 3H)

Example 3

Preparation of 5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

  • To the white crystals (50.5g) obtained in Example 2 was added 2-propanol (937.5 mL) and water (312.5 mL), and the resulting mixture was heated and dissolved at an internal temperature of 80°C. Immediately thereafter, the solution was filtered and the filtrate was cooled to an internal temperature of 20°C. To the resulting suspension was added dropwise 0.52 mol/l of an aqueous sodium hydrogen carbonate solution (250 mL), and the mixture was stirred at room temperature for 2 hours. Then the crystals were filtered and washed with water (150 mL×3) and 2-butanol (150 mL×2). The crystals were dried under reduced pressure at 80°C for 15 hours to give 29.4 g of the captioned compound as pale yellow crystals.
  • 1H―NMR(DMSO-d6)δppm:8.02(dd, 2H, J=4.59, 1.62Hz),8.32(dd, 1H, J=5.13, 1.62Hz), 8.55(dd, 1H, J=1.62, 1.08Hz), 8.80(dd, 2H, J=4.59, 1.62Hz), 8.93 (dd, 1H, J=5.13, 1.08Hz)

 

SYNTHESIS

US7074816

Example 12

5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

1) Production of methyl isonicotinate N-oxide

13.9 g of isonicotinic acid N-oxide was added to 209 ml of methylene chloride, 29.7 g of 1-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline was further added thereto, and the mixture was stirred under argon atmosphere at room temperature for one hour. 32.1 g of methanol was added to this mixture, which was stirred at room temperature for 17 hours. After the solvent was evaporated under reduced pressure, the residue was subjected to silica gel column chromatography. Chloroform-acetone (3:1) was used as an eluent to yield 11.1 g of a white powder.

1H-NMR (CDCl3) δppm: 3.95 (3H, s), 7.88 (2H, d, J=7.25 Hz), 8.22 (2H, J=7.25 Hz)

2) Production of Methyl 2-cyanoisonicotinate

11.1 g of the crystal obtained in 1) was dissolved in 170 ml of acetonitrile, 14.6 g of triethylamine and 21.5 g of trimethylsilylnitrile were added thereto, and the mixture was refluxed under argon atmosphere for 16 hours. After the solvent was evaporated under reduced pressure, the residue was subjected to silica gel column chromatography. Chloroform-acetone (95:5) was used as an eluent to yield 8.44 g of a pale yellow powder.

1H-NMR (CDCl3) δppm: 4.01 (3H, s), 8.08 (1H, d, J=5.45 Hz), 8.24 (1H, s), 8.90 (1H, d, J=5.45 Hz)

3) Production of 2-cyanoisonicotinic acid hydrazide

8.44 g of the crystal obtained in 2) was added to 85 ml of methanol, 1.84 g of hydrazine was further added thereto, and the mixture was stirred under argon temperature for 2 hours. After the solvent was evaporated under reduced pressure, chloroform was added to the residue, which was stirred at room temperature for one hour. The precipitated crystal was filtered, washed with chloroform and dried with a vacuum pump to yield 4.15 g of a pale yellow powder.

1H-NMR (DMSO-d6) δppm: 4.72 (2H, s), 8.05 (1H, d, J=5.12 Hz), 8.31 (1H, s),8.90 (1H, d, J=5.12 Hz), 10.23 (1H, s)

4) Production of the Object Compound

2.67 g of 4-cyanopyridine was dissolved in 40 ml of methanol, 0.83 g of sodium methoxide was added thereto, and the mixture was stirred at room temperature for one hour. Then 4.15 g of the crystal obtained in 3) was added and the mixture was refluxed for 37 hours. After the reaction completed, the precipitated solid was filtered, washed with methanol and dried with a vacuum pump to yield 3.66 g of the object compound as a yellow powder.

1H-NMR (DMSO-d6) δppm: 8.01 (2H, dd, J=4.54, 1.57 Hz), 8.31 (1H, dd, J=5.11, 1.65 Hz), 8.53 (1H, dd, J=1.65, 0.50 Hz), 8.80 (2H, dd, J=4.54, 1.57 Hz), 8.93 (1H, dd, J=5.11, 0.50 Hz)

Example 39

5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

1) Production of isonicotinic acid (N-2-tert-butoxycarbonyl)hydrazide-1-oxide

585 ml of methylene chloride was added to 39.0 g of isonicotinic acid N-oxide, and after 34.0 g of triethylamine was further added thereto, the mixture was cooled under argon atmosphere to −15° C. 33.5 g of ethyl chlorocarbonate in 117 ml of methylene chloride was added dropwise to this mixture, which was stirred at a temperature from −5 to −10° C. for one hour. Then 44.4 g of tert-butyl ester of carbamic acid in 117 ml of methylene chloride was added dropwise to this mixture and it was allowed to slowly rise to room temperature while it was stirred. The precipitated solid was filtered after 15 hours, washed with methylene chloride, and dried with a vacuum pump to yield 49.7 g of white crystal.

1H-NMR (DMSO-d6) δppm: 1.42 (9H, s), 7.82 (2H, d, J=7.09 Hz), 8.33 (2H, d, J=7.09 Hz), 9.02 (1H, s), 10.44 (1H, s)

Production of 2-cyanoisonicotinic acid hydrazine 1½ P-Toluenesulfonic acid salt

228 ml of dioxane was added to 30.4 g of the crystal obtained in 1), and after 13.1 g of trimethylsilyl cyanide and 38.8 g of N,N-dimethylcarbamoyl chloride were further added thereto, the mixture was stirred under argon atmosphere at 60° C. for 5 hours. After the solvent was evaporated under reduced pressure, the residue was dissolved in ethyl acetate and subsequently washed with 1.5 M sodium carbonate aqueous solution and a saturated saline solution and dried over magnesium sulfate. After the magnesium sulfate was filtered off, the solvent was evaporated under reduced pressure. Ethyl acetate was added to the residue, 68.5 g of p-toluenesulfonic acid monohydrate was added thereto, and the mixture was stirred at room temperature for 22 hours. The precipitated crystal was filtered, washed with ethyl acetate, and dried with a vacuum pump to yield 40.3 g of white crystal 2).

1H-NMR (DMSO-d6) δppm: 2.28 (4.5H, s), 7.12 (3H, dd, J=7.92 & 0.66 Hz), 7.48 (3H, dd, J=7.92 & 0.66 Hz), 8.10 (1H, dd, J=5.11 & 1.81 Hz), 8.39 (1H, dd, J=1.81 & 0.33 Hz), 8.99 (1H, dd, J=5.11 & 0.33 Hz)

3) Production of 5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

9.98 g of 4-cyanopyridine was dissolved in 250 ml of methanol, and after 7.77 g of sodium methoxide was added thereto, the mixture was stirred at room temperature for one hour. Then 40.3 g of the crystal obtained in 2) was added and the mixture was refluxed for 24 hours. After the reaction completed, the precipitated crystal was filtered, washed with methanol, and dried with a vacuum pump to yield 16.3 g of yellow crystal.

1H-NMR (DMSO-d6) δppm: 8.01 (2H, dd, J=4.54 & 1.57 Hz), 8.31 (1H, dd, J=5.11 & 1.65 Hz), 8.53 (1H, dd, J=1.65 & 0.50 Hz), 8.80 (2H, dd, J=4.54 & 1.57 Hz), 8.93 (1H, dd, J=5.11 & 0.50 Hz)

4) Production of 5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole

45 ml of ethanol and 15 ml of 1-methyl-2-pyrrolidone were added to 3.0 g of the crystal obtained in 3), and the mixture was heated and stirred at 80° C. for 19 hours. The crystal was filtered, subsequently washed with a mixture of ethanol and 1-methyl-2-pyrrolidone (3:1) and ethanol, and dried with a vacuum pump to yield 2.71 g of yellow crystal.

5) Production of 5-(2-cyano-4-pyridyl)-3-(4-pyridyl)-1,2,4-triazole p-toluenesulfonic acid salt

5 ml of ethanol and 30 ml of water were added to 2.48 g of the crystal obtained in 4), and after 3.8 g of p-toluenesulfonic acid monohydrate was further added thereto, the mixture was stirred at room temperature for 5 hours. The precipitated crystal was filtered, subsequently washed with a mixture of ethanol and water (1:6), water and then ethanol, and dried with a vacuum pump to yield 3.5 g of white crystal.

1H-NMR (DMSO-d6) δppm: 2.28 (3H, s), 7.12 (2H, dd, J=7.75 & 0.50 Hz), 7.48 (2H, dd, J=7.75 & 0.50 Hz), 8.33 (1H, dd, J=5.12 & 1.65 Hz), 8.45 (2H, d, J=6.11 Hz), 8.57 (1H, dd, J=1.65 & 0.66 Hz), 8.96˜9.02 (3H, m)

6) Production of the object compound

17 ml of ethanol and 17 ml of water were added to 3.36 g of the crystal obtained in 5), and the mixture was stirred at room temperature for 30 minutes. A solution of sodium carbonate (0.74 g of sodium carbonate in 17 ml of water) was further added, and the mixture was stirred at room temperature for 2 hours. The precipitated crystal was filtered, subsequently washed with water and ethanol, and dried with a vacuum pump to yield 1.89 g of the object compound as a pale yellow crystal.

 

2D image of a chemical structureTOPIROXOSTAT

SYNTHESIS

WO2014017516A1

Figure JPOXMLDOC01-appb-C000020

(First step)
The first step, 4 – is a step of obtaining a compound (3) is reacted in the presence of an alkali metal alkoxide, cyano-N-oxide and (2), and isonicotinic acid hydrazide.

4 used in this reaction – isonicotinic acid hydrazide and (2) a cyano-N-oxide is a known compound both, I can be prepared by known means.
The alkali metal alkoxide is used, 6 alkoxide alkali metal C 1-C are preferred, sodium methylate, sodium ethylate and the like can be given as specific examples. The reaction is preferably carried out in a solvent, as the solvent, alcohol solvents such as methanol, ethanol and the like are preferable.

The reaction is preferably first in a solvent, is treated with an alkali metal alkoxide compound (2) and then to react the isonicotinic acid hydrazide. First, heated to reflux under cooling, at 80 ℃ from 15 ℃ preferably, 30 minutes and 12 hours in general, the reaction temperature in the reaction with an alkali metal alkoxide (2) with the compound is reacted 1-4 hours, preferably about. Under the temperature conditions, using an excess amount or one equivalent of 30 minutes to 12 hours usually, reaction with isonicotinic acid hydrazide Subsequent to reaction for 1 to 5 hours, preferably.

Example 1:

Synthesis 4 oxide (3) – – – (4 – pyridin-carbonyl) -4 – N “pyridine hydrazide imide -1 was suspended in 40mL of methanol cyanopyridine-N-oxide and (2) 5.00g, sodium was added to methylate 22.4mg, and the mixture was stirred for 2 hours under 40 ℃ nitrogen atmosphere. was cooled to room temperature. reaction solution was stirred for 4 hours at 40 ℃ was added isonicotinic acid hydrazide 5.71g at the same temperature, precipitated The filtrated crystals were, washed with methanol 15mL, and dried 15 hours at 80 ℃, N “- to give (3) 9.60g oxide – (4 – pyridin) -4 – pyridine-hydrazide imide -1.
1 H-NMR (DMSO-d 6) δ (ppm): 6.98 (br, 2H), 7.81 (d, 2H, J = 5.77Hz), 7.85 (d, 2H, J = 7 .09 Hz), 8.29 (d, 2H, J = 7.09Hz), 8.73 (d, 2H, J = 5.77Hz), 10.37 (br, 1H)
MS m / z: 256 [M-H] 

(Second step)
The second step is a step of obtaining compound (4) by cyanation agent cyano compound (3).

As the cyanation agent used, trialkyl cyanide alkali metal cyanide, sodium cyanide, potassium cyanide and the like, zinc cyanide, trimethylsilyl cyanide and the like.

The cyanation reaction is preferably, for example, be carried out (Heterocycles, Vol.22, No.5, 1994) by Reissert Henze reaction. This reaction, for example, to give compound (4) by an organic solvent in the compound (3), and after activation with carbamoyl halide, and reacting the cyano agent. The alkylcarbamoyl halide used in the carbamoylation is a first step in Reissert Henze reaction, 6 alkylcarbamoyl halide di C 1-C dimethylcarbamoyl chloride, and di-propyl carbamoyl chloride can be used, preferably, dimethylcarbamoyl is chloride. The solvent used in this reaction, N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, tetrahydrofuran and acetonitrile can be used, however, N, N-dimethylformamide is preferred. Further, 15 ~ 60 ℃, more preferably 30 ~ 50 ℃ reaction temperature. The reaction time is preferably 1 to 24 hours, more preferably 1 to 3 hours. As the cyanation agent used in the cyanation reaction followed, cyano agents above can be used, sodium cyanide, potassium cyanide, zinc cyanide, and trimethylsilyl cyanide, and more preferably, it is sodium cyanide . -20 ~ 60 ℃ is preferred, more preferably -10 ~ 40 ℃, reaction temperature is 1-4 hours.

Is a novel compound (4) The compound obtained in this second step, it is useful as an intermediate for the production of compound (1). If through Compound (4) can be synthesized in good yield and easily without the need for purification in the second step is also possible, and can be produced (1) Compound industrially efficiently compound (4).

Synthetic N “hydrazide (4) – (4 – pyridine carbonyl) -4 – pyridine carboxylic acid N’-(carboxylic imidoyloxy – 2 – – cyano-4)

Example 2

4 pyridine hydrazide imide -1 – oxide ( was suspended in N, N-dimethylformamide 48mL and 3) 10.0g, under nitrogen atmosphere, followed by stirring for 1 hour was added dimethylcarbamoyl chloride 9.20g at 40 ℃. was added sodium cyanide 2.48g at the same temperature, After cooling to 5 ℃ below. reaction mixture was stirred for 1 hour, the crystals were collected by filtration. precipitate was successively added dropwise a 5% aqueous sodium bicarbonate solution 100mL, and 100mL water, and washed with water 100mL, at 80 ℃ for 15 h and dried under reduced pressure to give 4 – hydrazide (4) 9.28g of pyridine-carboxylic acid N’-(carboxylic imide yl – 2 – cyano-4).
1 H-NMR (DMSO-d 6) δ (ppm): 7.15 (br, 2H), 7.82 (d, 2H, J = 5.61Hz), 8.14 (d, 1H, J = 5 .11 Hz), 8.37 (s, 1H), 8.75 (d, 2H, J = 5.61Hz), 8.86 (d, 1H, J = 5.11Hz), 10.47 (br, 1H )
MS m / z: 265 [M-H] 

Figure JPOXMLDOC01-appb-C000019

(Third step)
The third step is a step of obtaining a compound (1) by the presence of an acid catalyst, the cyclization reaction of the compound (4).

As the acid, organic phosphoric acid, p-toluenesulfonic acid, such as hydrochloric acid, inorganic acids can be used, inorganic acids, phosphoric acid is particularly preferable. As the reaction solvent, water, 2 – butanol, 2 – mixed solvent of alcohol and water or alcohol, propanol, ethanol and the like can be used, but water and 2 – I was mixed 5:1 to 10:1 butanol solvent. The reaction temperature and time, 60 ~ 100 ℃, preferably 2 to 12 hours at 70 ~ 90 ℃, I want to 8-10 hours, preferably.

Intermediates and compounds of the present invention the method (1) can be isolated and purified from the washed reaction mixture, recrystallization, by means of various conventional chromatography.

Example 3:

4 – [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazol-3 – yl] pyridine-2 – carbonitrile 4 Synthesis of (1) – pyridine-carboxylic acid N’- (2 – cyano-4 – carboxylic imide yl) water 82mL, 2 hydrazide (4) 9.25g – butanol was added 8.2mL, phosphate 4.00g, was stirred for 8 h at 80 ℃. After cooling to room temperature, the reaction mixture was precipitated crystals were collected by filtration, water: 2 – were washed with a mixed solution of 92.5mL butanol = 10:1. The 13 h and dried under reduced pressure at 80 ℃ crystals obtained 4 – [5 – (pyridin-4 – yl) – 1 H-1, 2,4 – triazol-3 – yl] pyridine-2 – carbonitrile (1 I got a) 7.89g.

Topiroxostat


1 H-NMR (DMSO-d 6) δ (ppm): 8.02 (dd, 2H, J = 4.59,1.62 Hz), 8.32 (dd, 1H, J = 5.13,1. 62Hz), 8.55 (dd, 1H, J = 1.62,1.08 Hz), 8.80 (dd, 2H, J = 4.59,1.62 Hz), 8.93 (dd, 1H, 5 .13,1.08 Hz)
MS m / z: 247 [M-H] 

PATENT

WO2014017515A1

Synthetic water-carbonitrile p-toluenesulfonate – pyridine Example 1: 4 – [yl 5 – (pyridin-4 – yl)-1H-1, 2,4 – – -3 triazol]: 2 – butanol = was added monohydrate 6.62g p-toluenesulfonic acid in a mixed solution of 55mL of 10:1, 4 at 80 ℃ – [5 – (pyridin-4 – yl)-1H-1, 2,4 – yl] pyridine-2 – – triazol-3 was added carbonitrile 7.85g, and the mixture was stirred at the same temperature for 1 hour. After cooling to room temperature, the reaction mixture, and the precipitated crystals were collected by filtration, and water: 2 – were washed with a mixed solution of 40mL of butanol = 10:1. The dried under reduced pressure for 10 hours at 80 ℃ crystals obtained 4 – [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazol-3 – yl] pyridine-2 – carbonitrile p-toluene I got a sulfonate 12.6g.
1 H-NMR (DMSO-d 6) δ (ppm): 2.29 (s, 3H), 7.11 (m, 2H), 7.48 (dd, 2H, J = 6.48,1.62 Hz ) ,8.32-8 .35 (m, 3H), 8.57 (dd, 1H, J = 1.62,0.81 Hz) ,8.94-8 .98 (m, 3H)

– [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazole and potassium carbonate 8.22g, 4 in a mixed solution of 80mL of ethanol = 9:1: preparation water of crystal form I: Example 2 I was dissolved carbonitrile p-toluenesulfonate 10.0g – -3 – yl] pyridine-2. After stirring for 5 hours plus 15mL 6M hydrochloric acid at 20 ℃, was the precipitated crystals were collected by filtration, and washed with water 100mL. The 23 h and dried under reduced pressure at 80 ℃, 4 – to obtain carbonitrile 5.78g – pyridin-2 [yl 5 – (pyridin-4 – yl)-1H-1, 2,4 – – -3 triazole. Having a DSC as shown in FIG 4 and the powder X-ray diffraction pattern shown in FIG 1, the resulting crystals were type-I crystals.
1 H-NMR (DMSO-d 6) δ (ppm): 8.02 (dd, 2H, J = 4.59,1.62 Hz), 8.32 (dd, 1H, J = 5.13,1. 62Hz), 8.55 (dd, 1H, J = 1.62,1.08 Hz), 8.80 (dd, 2H, J = 4.59,1.62 Hz), 8.93 (dd, 1H, 5 .13,1.08 Hz)
Melting point: 327 ℃

N, N carbonitrile 40.0g – preparation of 4 Form II – [5 – (pyridin-4 – yl)-1H-1, 2,4 – yl – triazol-3]-2: Example 3 – dimethylformamide was added 300mL, and stirred for 25 min at 150 ℃. After cooling to room temperature the solution, and the precipitated crystals were collected by filtration, and washed twice with water 200mL, 4 and dried under reduced pressure overnight at 80 ℃ the crystal – [5 – (pyridin-4 – yl)-1H-1 , 2,4 – I got carbonitrile 30.4g – yl] pyridine-2 – triazole-3. Having a DSC as shown in FIG 5 and powder X-ray diffraction pattern shown in FIG 2, the resulting crystals were type II crystals.
1 H-NMR (DMSO-d 6) δ (ppm): 8.02 (dd, 2H, J = 4.59,1.62 Hz), 8.32 (dd, 1H, J = 5.13,1. 62Hz), 8.55 (dd, 1H, J = 1.62,1.08 Hz), 8.80 (dd, 2H, J = 4.59,1.62 Hz), 8.93 (dd, 1H, 5 .13,1.08 Hz)
Melting point: 327 ℃

The 25 ℃, about 2g carbonitrile, – preparation of the hydrate 4 – [5 – (pyridin-4 – yl)-1H-1, 2,4 – triazol-3 – yl] pyridine-2: Example 4 I was stored for 14 days under conditions of relative humidity 97%. Having a DSC as shown in FIG 7 and the powder X-ray diffraction pattern shown in FIG 3, the obtained crystal was a hydrate.
1 H-NMR (DMSO-d 6) δ (ppm): 8.02 (dd, 2H, J = 4.59,1.62 Hz), 8.32 (dd, 1H, J = 5.13,1. 62Hz), 8.55 (dd, 1H, J = 1.62,1.08 Hz), 8.80 (dd, 2H, J = 4.59,1.62 Hz), 8.93 (dd, 1H, 5 .13,1.08 Hz)
Melting point: 327 ℃

Test Example: solubility test Type I crystal by crystal form, II-type crystal, and water solubility of the hydrate was calculated by absorbance measurement method, a saturated solution concentration of each sample. I Figure 8 shows the results.Whereas the 6.2μg/mL water solubility of crystalline Form I, II type crystal 4.2μg/mL, hydrate was 1.9μg/mL.
From Figure 8, the water solubility of Form II and Form I crystals is good, water-soluble type I crystal is particularly good.

 

NMR

BMCL Volume 19, Issue 21, 1 November 2009, Pages 6225–6229

http://www.sciencedirect.com/science/article/pii/S0960894X09012372?np=y

view compd 39 and ignore rest

Full-size image (3 K)TOPIROXOSTAT, FYX O51

view compd 39 and ignore rest

SUPP INFO…….https://docs.google.com/viewer?url=http://www.sciencedirect.com/science/MiamiMultiMediaURL/1-s2.0-S0960894X09012372/1-s2.0-S0960894X09012372-mmc1.doc/271398/FULL/S0960894X09012372/50d911fe734c16dfb94912d481cb466a/mmc1.doc

1 * Baldwin, J.J., J. Med. Chem.; 1975; 18(9); 895-900, especially p. 898, lines 3-5.
2 * Geldard, J.F. et al., J. Org. Chem.; 1965; 30(1); 318-319, especially p. 319, starting line 33.
3 * Lever, A.B.P., Inorg. Chem; 1990; 29; 1271-1285, especially p. 1275, line 18 and 19.

Nucleosides, Nucleotides and Nucleic Acids, 2008 ,  vol. 27,  6-7  pg. 888 – 893

Inoue, Tsutomu; Sato, Takahiro; Ashizawa, Naoki; Iwanaga, Takashi; Matsumoto, Koji; Nagata, Osamu; Nakamura, Hiroshi
Bioorganic and Medicinal Chemistry Letters, 2009 ,  vol. 19,   21  pg. 6225 – 6229

WO 2012060308

WO 2007148835

WO 2005009991

WO2003064410A1 * Dec 3, 2002 Aug 7, 2003 Naoki Ashizawa Novel 1,2,4-triazole compound
US3882134 * May 21, 1973 May 6, 1975 Merck & Co Inc 1-Substituted-3,5-dipyridyl-1,2,4-triazoles
US3947577 * Jan 8, 1975 Mar 30, 1976 Merck & Co., Inc. Anti-hyperuricemia composition
US3984558 * Nov 29, 1974 Oct 5, 1976 Merck & Co., Inc. 1,3,5-Trisubstituted-1,2,4-triazole compounds used as bronchodilators
US4011218 * Dec 3, 1974 Mar 8, 1977 Merck & Co., Inc. 1,2,4-triazoles
US4104393 * Sep 2, 1977 Aug 1, 1978 Merck & Co., Inc. 1,3,5-Trisubstituted-1,2,4-triazole compounds
US5571897 * Dec 5, 1991 Nov 5, 1996 Wallac Oy Luminescent lanthanide chelates
Publication Number Publication Date IPCR Assignee/Applicant Structure hits Tools
1.

US-9199970-B2

2015-12-01
4-[5-(pyridin-4-yl)-1H-1,2,4-triazol-3-yl]pyridine-2-carbonitrile crystalline polymorph and production method therefor
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
2.

US-20150322006-A1

2015-11-12
PHENYLTHIOACETATE COMPOUNDS, COMPOSITIONS AND METHODS OF USE
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
3.

US-20150309021-A1

2015-10-29
Advanced Drug Development and Manufacturing
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
4.

US-20150291543-A1

2015-10-15
2-ARYL SELENAZOLE COMPOUND AND PHARMACEUTICAL COMPOSITION THEREOF
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
5.

EP-2927219-A1

2015-10-07
2-ARYL SELENAZOLE COMPOUND AND PHARMACEUTICAL COMPOSITION THEREOF
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
6.

US-20150274680-A1

2015-10-01
THERAPEUTIC OR PROPHYLACTIC AGENT FOR TUMOR LYSIS SYNDROME
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
7.

EP-2913053-A1

2015-09-02
THERAPEUTIC OR PROPHYLACTIC AGENT FOR TUMOR LYSIS SYNDROME
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
8.

EP-2511844-B1

2015-08-12
X-ray microscope
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
9.

EP-2712861-B1

2015-07-29
Thioacetate compounds, compositions and methods of use
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
10.

US-20150203490-A1

2015-07-23
RING-FUSED COMPOUND
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
11.

US-20150191463-A1

2015-07-09
DIFLUOROMETHYLENE COMPOUND
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
12.

US-20150166510-A1

2015-06-18
METHOD FOR PRODUCING 4-[5-(PYRIDIN-4-YL)-1H-1,2,4-TRIAZOL-3-YL]PYRIDINE-2-CARBONITRILE, AND INTERMEDIATE THEREOF
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
13.

EP-2878594-A1

2015-06-03
DIFLUOROMETHYLENE COMPOUND
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
14.

EP-2878598-A1

2015-06-03
4-[5-(PYRIDINE-4-YL)-1H-1,2,4-TRIAZOLE-3-YL]PYRIDINE-2-CARBONITRILE CRYSTALLINE POLYMORPH AND PRODUCTION METHOD THEREFOR
E
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
15.

EP-2878595-A1

2015-06-03
METHOD OF PRODUCING 4-[5-(PYRIDIN-4-YL)-1H-1,2,4-TRIAZOLE-3-YL]PYRIDIN-2-CARBONITRILE, AND INTERMEDIARY THEREOF
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
16.

US-20150126558-A1

2015-05-07
4-[5-(PYRIDIN-4-YL)-1H-1,2,4-TRIAZOL-3-YL]PYRIDINE-2-CARBONITRILE CRYSTALLINE POLYMORPH AND PRODUCTION METHOD THEREFOR
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
17.

US-8987473-B2

2015-03-24
Ring-fused compound
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
18.

EP-2842948-A1

2015-03-04
Novel compounds and compositions and methods of use
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
19.

EP-2776028-A1

2014-09-17
3,4-DI-SUBSTITUTED PYRIDINE COMPOUND, METHODS OF USING AND COMPOSITIONS COMPRISING THE SAME
EN
N#CC1=CC(=CC=N1)C1=NNC(=N1)C1=CC=NC=C1
20.

US-20140256748-A1

2014-09-11
3,4-DI-SUBSTITUTED PYRIDINE COMPOUND, METHODS OF USING AND COMPOSITIONS COMPRISING THE SAME

 

Topiroxostat
Topiroxostat.svg
Systematic (IUPAC) name
4-[5-(4-Pyridinyl)-1H-1,2,4-triazol-3-yl]-2-pyridinecarbonitrile
Clinical data
Trade names Topiloric, Uriadec
Legal status
  • Approved in Japan
Identifiers
CAS Number 577778-58-6
ATC code None
PubChem CID: 5288320
ChemSpider 4450517
Chemical data
Formula C13H8N6
Molecular mass 248.24 g/mol

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

C1=CN=CC=C1C2=NC(=NN2)C3=CC(=NC=C3)C#N

Pamicogrel KB 3022 for Coagulation Disorders Therapy


Pamicogrel
Pamicogrel
CAS  101001-34-7
D01090, TO-192, KBT-3022, KB-3022, Paminate, UNII-398FD8EDAL
2-[4,5-Bis(4-methoxyphenyl)-2-thiazolyl]-1H-pyrrole-1-acetic acid ethyl ester
Ethyl 2-[4,5-bis(4-methoxyphenyl)thiazol-2-yl]-pyrrole-1-acetate
ethyl 2-[4,5-bis(p-methoxyphenyl)-2-thiazolyl]pyrrole-1-acetate
Manufacturers’ Codes: KB-3022; KBT-3022
Kanebo (Originator), Torii (Codevelopment)
Antiplatelet Therapy, Coagulation Disorders Therapy, HEMATOLOGIC DRUGS, Cyclooxygenase Inhibitors
Molecular Formula: C25H24N2O4S
Molecular Weight: 448.53
Percent Composition: C 66.94%, H 5.39%, N 6.25%, O 14.27%, S 7.15%
Properties: Crystals from ligroin, mp 132.5-135.5°.
Melting point: mp 132.5-135.5°
Toxicity data: LD50 orally in male mice: >3000 mg/kg (Yoshino)
Therap-Cat: Antithrombotic.
.PAMICOGREL

Pamicogrel (CAS NO.: 101001-34-7), with its systematic name of 1H-Pyrrole-1-acetic acid, 2-(4,5-bis(4-methoxyphenyl)-2-thiazolyl)-, ethyl ester, could be produced through many synthetic methods.

Following is one of the synthesis routes:
alpha-Bromo-4,4-dimethoxidesoxybenzoin (I) is cyclized with pyrrole-2-carbothioamide (II) in hot acetonitrile to produce 4,5-bis(4-methoxyphenyl)-2-(pyrrol-2-yl)thiazole (III), which is then condensed with ethyl bromoacetate (IV) in the prsence of NaOH and tetrabutylammonium bromide in refluxing dichloromethane – water.

The cyclization of alpha-bromo-4,4′-dimethoxydesoxybenzoin (I) with pyrrole-2-carbothioamide (II) in hot acetonitrile gives 4,5-bis(4-methoxyphenyl)-2-(pyrrol-2-yl)thiazole (III), which is then condensed with ethyl bromoacetate (IV) by means of NaOH and tetrabutylammonium bromide in refluxing dichloromethane – water
EP 0159677; JP 1985222481; JP 1986033186; JP 1986200985; US 4659726
pamicogrel
SYNTHESIS
    Reaction Scheme-I:

  • Figure imgb0005
    Figure imgb0006

    wherein R is as defined above, and Y is a halogen such as chlorine, bromine or iodine, or p-toluenesulfonyloxy group.

  • The process of the above reaction scheme-I can be carried out by reacting a compound (II) and an equimolar or excess amount of a compound (III) in the presence of a base or a phase transfer catalyst. In case of using a base such as metallic potassium, metallic sodium, potassium tert-butoxide etc.; the reaction is carried out in a solvent of tetrahydrofuran or dimethoxyethane at a temperature of from room temperature to a boiling point of the solvent for 1 to 24 hours. In case of using a phase transfer catalyst such as a quaternary ammonium salt (e.g. tetra-n-butylammonium bromide, methyltrioctylammonium chloride, etc.), the reaction is carried out in two phases of benzene or dichloromethane and 50 % aqueous sodium hydroxide or 60 % aqeuous potassium hydroxide at a temperature of from 0°C to a boiling point of the solvent for one minute to 24 hours.
    Reaction Scheme-II:

  • Figure imgb0007

    wherein X is a halogen such as bromine or chlorine.

  • The above process can be carried out by reacting a compound (IV) and an equimolar amount of a compound (V) in a solvent such as acetonitrile, dimethylformamide (DMF), dimethyl sulfoxide (DMSO), or an alcohol (e.g. ethanol) at a temperature of from 50°C to a boling point of the solvent for 10 minutes to 4 hours.
    Reaction Scheme-III:

  • Figure imgb0008
    Figure imgb0009
    Reaction Scheme-IV:

  • Figure imgb0010

    wherein R1 is as defined above.

  • The process can be carried out by converting a compound (VII) into an oxime (VIII) by a conventional oxime forming reaction, heating the oxime (VIII) in acetic anhydride to obtain a compound (IX), and treating the compound (IX) with hydrogen sulfide, that is, by blowing hydrogen sulfide gas into a reaction system containing the compound (IX) in a solvent such as DMF, DMSO or pyridine in the presence of 0.5 to 5 equimolar amount of a base such as a tertiary amine (e.g. triethylamine) at a temperature of from 0° to 40°C for 3 to 24 hours
    Reaction Scheme-V:

  • Figure imgb0011
    Reference Example 6
    4,5-Bis(4-methoxyphenyl)-2-(pyrrol-2-yl)thiazole [compound of the formula (II)]:
  • Pyrrole-2-carbothioamide (cf. J. Org. Chem., 38, 667, 1973) (1.51 g, 12 mmole) and α-bromo-4,4′-dimethoxy- deoxybenzoin (cf. Aust. J. Chem., 8, 385. 1955) (4.02 g, 12 mmole) are dissolved in acetonitrile (120 ml). The mixture is stirred at 60°C for 50 minutes. After the reaction, the reaction mixture is distilled under reduced pressure to remove the solvent. To the resulting residue are added chloroform and aqueous solution of sodium carbonate, and the mixture is shaken. The chloroform layer is taken, and the aqueous layer is further extracted with chloroform. The chloroform layers are combined, dried over anhydrous magnesium sulfate, and distilled under reduced pressure to remove the solvent. The residue is recrystallized from ligroin to give 4,5-bis(4-methoxyphenyl)-2-(pyrrol-2-yl)-thiazole (3.74 g, yield: 86 %).
  • M.p. 131.5 – 134.0°C
  • NMR (CDCl3, δ ppm): 3.7 (6H) , 6.1 (1H, dd) , 6.5-6.9 (6H), 7.1-7.5 (4H), 9.4-9.8 (lH).
  • Elementary analysis for C21H18N2O2S:

    Figure imgb0016
    Example 14

  • Ethyl 2-[4,5-bis(4-methoxyphenyl)thiazol-2-yl]-pyrrole-1-acetate (compound of the formula (I) wherein R1 = -CH2-COOC2H5):

    • 4,5-Bis(4-methoxyphenyl)-2-(pyrrol-2-yl)thiazole obtained in the same manner as described in Reference Example 6 (3.62 g, 10 mmole), ethyl bromoacetate (1.67 g, 10 mmole), and tetra-n-butylammonium bromide (0.32 g, 1 mmole) are refluxed with vigorous stirring in two phases of dichloromethane (40 ml) and 50 % aqueous sodium hydroxide (40 ml) at room temperature for 2 minutes. To the mixture are added water and dichloromethane under ice-cooling, and the mixture is shaken. The dichloromethane layer is taken, and the aqueous layer is further extracted with dichloromethane. The dichloromethane layers are combined, dried over anhydrous magnesium sulfate, and distilled under reduced pressure to remove the solvent. The residue is recrystallized from ligroin to give ethyl 2-[4,5-bis(4-methoxyphenyl)thiazol-2-yl]pyrrole-1-acetate (3.64 g, yield: 81 %).
  • M.p. 132.5 – 135.5°C
  • NMR (CDCl3, δ ppm): 1.2 (3H, t), 3.8 (6H), 4.15 (2H, q), 5.25 (2H, s), 6.25 (1H, dd), 6.7-6.95 (6H), 7.2-7.55 (4H).
  • Elementary analysis for C25H24N2O4S:

    Figure imgb0026
Literature References:
Cyclooxygenase inhibitor. Prepn: K. Yoshino et al., EP 159677eidem, US 4659726 (1985, 1987 both to Kanebo).
Drugs Fut1991,16,(2):105
Chemical and Pharmaceutical Bulletin, 1992 ,  vol. 40,  11  pg. 3048 – 3051
HPLC determn in plasma and urine: Y. Nakada et al., Chem. Pharm. Bull. 38, 1093 (1990).
Pharmacokinetics: Y. Nakada et al., Yakuzaigaku 53, 210 (1993), C.A. 120, 315100 (1993).
Activity as antithrombotic: K. Yokoto et al., Jpn. J. Pharmacol. 68, 201 (1995); as platelet aggregation inhibitor: K. Yokoto et al., J. Pharm. Pharmacol. 47, 768 (1995).
Evaluation of cerebral protective effects: N. Yamamoto et al., Jpn. J. Pharmacol. 69, 421 (1995); eidem, Eur. J. Pharmacol. 297, 225 (1996).

Determination of the antiplatelet agent. KB-3022, and its metabolite by high-performance liquid chromatography.Nakada Y, Ikuta Y, Kawashima T, Awata N.Chem Pharm Bull (Tokyo). 1990 Apr;38(4):1093-5.

pamicogrel
PATENTS
EP0037274A1 * 30 Mar 1981 7 Oct 1981 Eli Lilly And Company Substituted triaryl thiazole compounds
EP0077024A2 * 7 Oct 1982 20 Apr 1983 Schering Aktiengesellschaft Imidazole derivatives, process for their preparation and pharmaceutical products containing them
US4168315 * 28 Sep 1977 18 Sep 1979 The Upjohn Company Dianisyl thiazole compound, compositions and method of antithrombotic treatment

TAKEDA PHARMACEUTICALS 武田薬品工業株式会社 ON THE RISE


Tadataka Yamada, M.D., Chief Medical & Scientific Officer of Takeda

TAKEDA US CHICAGO OFFICE

TAKEDA PIPELINE SEE LINKS BELOW

1    https://www.takeda.com/investor-information/annual/files/ar2013_10_en.pdf

2. http://www.takeda.com/research/files/pipeline_20131031_en.pdf

3  http://www.takeda.com/research/pipeline/

PDF PDF 1.0MB 18P

Takeda’s top executives had frequently pointed to TAK-875 as one of their best shots at coming up with an important new approach to treating diabetes. The drug is designed to spur insulin secretion in the pancreas and Takeda had confidently projected an approval in Japan in 2015 with a follow-up approval in the big U.S. market a year or two later.

The termination of the high-profile program caused some anxiety among investors. Takeda’s shares plunged 8% on the loss as analysts wondered how the pharma company could counter the loss of Actos, a $3.7 billion drug that accounted for about a quarter of its revenue in 2011.

Takeda won an approval on a trio of DPP-4 diabetes drugs–Nesina (alogliptin) and two combos with alogliptin, dubbed Oseni and Kazano–at the beginning of the year. But Takeda suffered some big delays in gaining acceptance, a common fate in this field, where regulators are particularly cautious about new drugs. And Merck  had already solidified its lead in the DPP-4 market with Januvia whileOnglyza trailed closely behind it. Takeda had hoped that a combination of TAK-875 and Januvia could help regain some lost market territory–but that dream has clearly vanished as well.

January 27, 2014
January 22, 2014
January 17, 2014
January 14, 2014
January 10, 2014

2013

December 27, 2013
December 25, 2013
December 25, 2013
December 24, 2013
December 20, 2013
December 20, 2013
December 19, 2013
December 10, 2013
December 10, 2013
December 10, 2013
December 10, 2013
December 9, 2013
December 5, 2013
December 4, 2013
November 30, 2013
November 21, 2013
November 19, 2013
November 14, 2013
November 12, 2013
November 12, 2013
October 21, 2013
October 7, 2013
October 2, 2013
October 1, 2013
September 26, 2013
September 26, 2013
September 24, 2013
September 20, 2013
September 20, 2013
September 13, 2013
September 13, 2013
September 5, 2013
September 2, 2013
August 27, 2013
August 27, 2013
August 27, 2013
August 22, 2013
August 13, 2013
August 1, 2013
July 31, 2013
July 31, 2013
July 31, 2013
July 30, 2013
July 29, 2013
July 26, 2013
July 19, 2013
July 19, 2013
July 19, 2013
July 18, 2013
July 10, 2013
July 1, 2013

CLIPPED

Takeda isn’t quite in the top 10 among global drugmakers, but the company boasts the 7th-largest pipeline in the industry, according to its presentation at the conference. Yamada noted that 31% of the pipeline assets are in late-stage trials. Millennium is leading development of three late-stage contenders, TAK-700 for prostate cancer, MLN9708 for multiple myeloma and MLN0002 for ulcerative colitis andCrohn’s disease.

In an effort to revive its diabetes franchise, Takeda is in the final stage of development for a first-of-a-kind GPR40 agonist called TAK-875, designed to provide glucose-dependent insulin secretion.

With a rich late-stage pipeline at Takeda, Yamada wants the company to focus on growing its ranks of earlier-stage drug candidates. To do this the company has landed a variety of deals, including the purchase of Intellikine for $310 million to acquire anti-cancer drugs and more recently the acquisition of Envoy Therapeutics last year for $140 million.

Takeda has formed a New Frontier Science group to scout out the hottest research in academia and elsewhere and form collaborations with scientists behind those innovations. At the J.P. Morgan conference, Yamada said, he was attending many meetings with members of the biotech community.

Takeda Pharmaceutical Company Limited (武田薬品工業株式会社 Takeda Yakuhin Kōgyō Kabushiki-gaisha?) is the largest pharmaceutical company in Japan and Asia and a top 15 pharmaceutical company. The company has over 30,000 employees worldwide and achieved $16.2 billion USD in revenue during the 2012 fiscal year.[1] The company is focused on metabolic disordersgastroenterologyneurologyinflammation, as well asoncology through its independent subsidiary, Millennium: The Takeda Oncology Company.[2] Its headquarters is located in Chuo-kuOsaka, and it has an office in NihonbashiChuoTokyo.[3][4] In January 2012, Fortune Magazine ranked the Takeda Oncology Company as one the 100 best companies to work for in the United States.

Takeda Pharmaceuticals was founded on June 12, 1781 and was incorporated on January 29, 1925.

Takeda’s Japanese logo

In 1977, Takeda first entered the U.S. pharmaceutical market by developing a joint venture with Abbott Laboratories called TAP Pharmaceuticals.[5]Through TAP Pharmaceuticals, Takeda and Abbott launched the blockbusters Lupron (leuprolide) in 1985 and Prevacid (lansoprazole) in 1995.

One of the firm’s mainstay drugs is Actos, a compound in the thiazolidinedione class of drugs used in the treatment of type 2 diabetes. Launched in 1999, Actos has become the best-selling diabetes drug in the world with $4 billion USD in sales during the 2008 fiscal year.[6]

In February 2005, Takeda announced its acquisition of San Diego, California-based Syrrx, a company specializing in high-throughput X-ray crystallography, for $270 million.[7]

In February 2008, Takeda acquired the Japanese operations of Amgen and rights to a dozen of the California biotechnology company’s pipeline candidates for the Japanese market.[8]

In March 2008, Takeda and Abbott Laboratories announced plans to conclude their 30-year old joint venture, TAP Pharmaceuticals, that had over $3 billion in sales in its final year. The split resulted in Abbott acquiring U.S. rights to Lupron and the drug’s support staff. On the other hand, Takeda received rights to Prevacid and TAP’s pipeline candidates. The move also increased Takeda’s headcount by 3,000 employees.[9]

In April 2008, Takeda announced that it was acquiring Millennium Pharmaceuticals of CambridgeMassachusetts, a company specializing in cancerdrug research, for $8.8 billion. The acquisition brought in Velcade, a drug indicated for hematological malignancies, as well as a portfolio of pipeline candidates in the oncology, inflammation, and cardiovascular therapeutic areas. Millennium now operates as an independent subsidiary, serving as the global center of excellence in oncology under its new name: “Millennium: The Takeda Oncology Company.” [10]

In May 2008, the company licensed non-exclusively the RNAi technology platform developed by Alnylam Pharmaceuticals, creating a potentially long-term partnership between the companies.[11]

On May 19, 2011, Takeda Pharmaceutical and Nycomed announced that Takeda will acquire Nycomed for  9.6 billion. The acquisition was completed by September 30, 2011.[12]

On April 11, 2012, Takeda Pharmaceutical and URL Pharma announced that Takeda will acquire URL Pharma for $800 million. The acquisition is expected to be completed within 60 days.

On 25 May 2012, Takeda announced the purchase of Brazilian pharmaceutical company Multilab by R$ 540 million.[13]

Takeda Midosuji Building, headquarters of Takeda Pharmaceutical Company, inChuo-ku, Osaka, Japan

Takeda operates two primary bases in Japan in Osaka and Tokyo. Its United States subsidiary is based in Deerfield, Illinois, and all Global Operations outside of Japan and U.S. are based in Opfikon (Zurich), Switzerland. The company maintains research & development sites in Osaka and Tsukuba, Japan; San Diego andSan Francisco, United States; Cambridge, United Kingdom; and Singapore.[14]

The company has manufacturing facilities in Japan, China, Indonesia, Italy, and Ireland.[15] Following the Nycomed acquisition, the Takeda manufacturing sites have been extended with facilities in Argentina,Austria,Belgium,Brazil,Denmark, Estonia,Germany,Mexico,Norway and Poland. Takeda has overseas marketing presences in the U.S., UK, France, Italy, Germany, Austria, Switzerland, Spain, China, Taiwan, Philippines, Thailand, Indonesia, and Singapore. It has recently[when?] announced its first foray into Canada, Portugal, Spain, Mexico, and Ireland.[15]

AT INDONESIA

Products

Some of the key products that Takeda produces on behalf of partners include:[16]

AT UK

References

  1.  “Financial Results for Fiscal 2012” (PDF). Takeda Pharmaceutical Company Limited. May 9, 2013. Retrieved June 13, 2013.
  2. “Takeda Initiates Cardiovascular Outcomes Trial for Alogliptin, An Investigational Treatment for Type 2 Diabetes”. Newsblaze.com. 2009-08-28. Retrieved 2010-09-18.
  3.  “FAQ.” Takeda Pharmaceutical Company. Retrieved on February 2, 2011. “Q : Where is Takeda located? A : The Head Office is located in Osaka, Japan, and the Tokyo Head Office is located in Tokyo, Japan.”
  4.  “Overview.” Takeda Pharmaceutical Company. Retrieved on February 2, 2011. “Headquarters Head Office 1-1, Doshomachi 4-chome, Chuo-ku, Osaka 540-8645” and “Tokyo Head Office 12-10, Nihonbashi 2-chome, Chuo-ku, Tokyo 103-8668”
  5.  “TAP Pharmaceutical Products, Inc.: Private Company Information – BusinessWeek”. Investing.businessweek.com. 2008-04-30. Retrieved 2010-09-18.
  6. Decker, Susan (2009-07-06). “Takeda Sues Torrent to Stop Generic Copy of Actos Diabetes Pill”. Bloomberg. Retrieved 2010-09-18.
  7.  Somers, Terri (2005-02-08). “Japanese drug giant taking over Syrrx here | The San Diego Union-Tribune”. Signonsandiego.com. Retrieved 2010-09-18.
  8.  “Takeda, Amgen in exclusive tie-up for Japanese market”. MarketWatch. 2008-02-04. Retrieved 2010-09-18.
  9.  Marrazzo, Amanda (2008-05-15). “Featured Articles From The Chicago Tribune”. Archives.chicagotribune.com. Retrieved 2010-09-18.
  10.  “MILLENNIUM: The Takeda Oncology Company | About Millennium | Our History”. Mlnm.com. Retrieved 2010-09-18.
  11.  staff (2008-06-15). “Takeda Signs On as Alnylam’s Asian Partner for $150M Upfront”. Genetic Engineering & Biotechnology News (print) (Mary Ann Liebert, Inc.). p. 14.
  12.  http://www.takeda.com/press/article_43116.html
  13.  Hirschler, Ben (May 25, 2012). “Farmacêutica Takeda comprará Multilab por até R$ 540 mi”Grupo Abril (in portuguese). Exame. Retrieved January 27, 2013.
  14.  “Locations | Worldwide | Takeda Pharmaceutical Company Limited”. Takedaism.com. Retrieved 2010-09-18.
  15.  “By Business | Worldwide | Takeda Pharmaceutical Company Limited”. Takedaism.com. Retrieved 2010-09-18.
  16.  “Annual Reports | Investor Information | Takeda Pharmaceutical Company Limited”. Takeda.com. Retrieved 2010-09-18.

Takeda Pharmaceutical Company Limited
Takeda-pharma 200x200.jpg
Native name 武田薬品工業株式会社
Type Public KK
Traded as
Industry Pharmaceuticals
Founded Doshomachi, Osaka, Japan (June 12, 1781)
Headquarters 1-1, Doshomachi Yonchome,Chuo-ku, Osaka, Japan
Key people Yasuchika Hasegawa (President & CEO)
Revenue Increase ¥1,557,267 million (FY2012)[* 1]
Operating income Decrease ¥122,505 million (FY 2012)[* 1]
Net income Increase ¥131,244 million (FY 2012)[* 1]
Total assets Increase ¥3,955,599 million (FY 2012)[* 1]
Total equity Increase ¥2,223,359 million (FY 2012)[* 1]
Employees 30,481 (2012)
Website takeda.com (Global website)
References:

  1. “Financial Results for Fiscal 2012” (PDF). Takeda Pharmaceutical Company Limited. May 9, 2013. Retrieved June 13, 2013.

CMC CENTRE

The Chemistry, Manufacturing and Controls (CMC) Center is a global organization responsible for overall R&D activities ranging from chemical information on development candidates to the processes leading to “manufacturing” of pharmaceutical products.

The main sites are located in Osaka and consist of the following laboratories: the Chemical Development Laboratories in charge of R&D for developing the manufacturing methods of active pharmaceutical ingredients and the manufacturing of drug substances for clinical samples; the Pharmaceutical Technology R&D Laboratories in charge of R&D for dosage forms, manufacturing and packaging, as well as manufacturing of clinical samples; and the Analytical Development Laboratories in charge of R&D for the development of analytical methods and stability studies of clinical samples. In addition, Hikari Bio-Manufacturing Technology Laboratories is located in Hikari (Yamaguchi) and this is where antibody drug substances are manufactured.

As for overseas sites, the Cambridge Biologics CMC Group (Massachusetts) and the Chicago Pharmaceutical Science Group (Illinois) are located in the USA, while the CMC Center Europe is mainly located in Roskilde, Denmark. All research and development activities at Takeda are promoted with the cooperation of these sites.

List of Publications of Takeda Research Laboratories

PDF PDF 0.4MB 34P
PDF PDF 0.5MB 26P
PDF PDF 0.5MB 16P
PDF PDF 0.5MB 16P
PDF PDF 0.1MB 17P
PDF PDF 0.1MB 15P
PDF PDF 0.1MB 17P

Trelagliptin succinate (SYR-472) for the treatment of type 2 diabetes.


Trelagliptin succinate (SYR-472)

2-[[6-[(3R)-3-aminopiperidin-1-yl]-3-methyl-2, 4-dioxopyrimidin-1-yl]methyl]-4-fluorobenzonitrile; butanedioic acid

2-[6-[3(R)-Aminopiperidin-1-yl]-3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidin-1-ylmethyl]-4-fluorobenzonitrile

2- [ [6- [ (3R) -3-amino-l-piperidinyl] -3, 4-dihydro-3- methyl-2, 4-dioxo-l (2H) -pyrimidinyl]methyl] -4-fluorobenzonitrile

succinic acid salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile

Sponsor/Developer: Takeda Pharmaceuticals and Furiex Pharmaceuticals

Mechanism of action: DPP-4 inhibitor

865759-25-7 cas FREE BASE

1029877-94-8  succinate

  • SYR 111472 succinate
  • SYR 472
  • Syr-472
  • Syr111472 succinate
  • Trelagliptin succinate
  • UNII-4118932Z90

Trelagliptin-succinate M. Wt: 475.47
Trelagliptin-succinate Formula: C22H26FN5O6

SYR-472 is an oral dipeptidyl peptidase IV inhibitor originated by Takeda. It is in phase III clinical trials for the treatment of type 2 diabetes.

  • Diabetes affects 25.8 million people of all ages, or roughly 8.3 percent of the U.S. population.
  • The World Health Organization predicts that there will be 366 million people worldwide affected by diabetes by the year 2030.
  • The advent of trelagliptin succinate, a unique once weekly medication for patients with type 2 Diabetes is now the focus of clinical trials and exciting research and development.
  • Phase III clinical trials of trelagliptin succinate commenced in September 2011, and are estimated to be complete by the second half of 2013.
TRELAGLIPTIN (SYR-472)
Trelagliptin is a novel DPP-4 inhibitor that is being developed by Takeda. In contrast to alogliplitin, which is once a day, trelagliptin is a once-weekly oral agent which should provide patients with a convenient therapeutic alternative and has the potential to improve compliance. Takeda has commenced Phase III trials of trelagliptin in Japan for the treatment of Type 2 diabetes.

Indication (Phase): Japan—Once-weekly oral treatment for type 2 diabetes (Phase III; study expected to be completed in second half of 2013)


trelagliptin succinate

Compound I, A, TRELAGLIPTIN which has the formula:

Figure US20080227798A1-20080918-C00002

is a DPP-IV inhibitor that is described in U.S. patent application Ser. No. 11/080,992 filed Mar. 15, 2005 (see Compound 34). Its dosing, administration and biological activities are described in U.S. patent application Ser. No. 11/531,671 filed Sep. 13, 2006. U.S. patent application Ser. No. 11/080,992 and Ser. No. 11/531,671 are incorporated herein by reference in their entirety.

Dipeptidyl peptidase IV (IUBMB Enzyme Nomenclature EC.3.4.14.5) (referred herein as “DPP-IV”) is a type II membrane protein and a non-classical serine aminodipeptidase that removes Xaa-Pro dipeptides from the amino terminus (N-terminus) of polypeptides and proteins. DPP-IV is constitutively expressed on epithelial and endothelial cells of a variety of different tissues (e.g., intestine, liver, lung, kidney and placenta), and is also found in body fluids. DPP-IV is also expressed on circulating T-lymphocytes and has been shown to be synonymous with the cell-surface antigen, CD-26. DPP-IV has been implicated in a number of human disease states, including, but are not limit to, diabetes, particularly type II diabetes mellitus, diabetic dislipidemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose (IFG), metabolic acidosis, ketosis, appetite regulation and obesity; autoimmune diseases such as inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis; AIDS; and cancers.

DPP-IV inhibitors are believed to be useful agents for the prevention, delay of progression, and/or treatment of conditions mediated by DPP-IV.

Compound (A) or a salt thereof has been reported as an inhibitor of dipeptidyl peptidase (DPP-IV) , which is an enzyme that decomposes glucagon-like peptide-1 (GLP-1) , a hormone increasing insulin secretion (patent document 1) .

In addition, a method including administering 1 – 250 mg of compound (A) or a salt thereof to a patient once per week (patent documents 2, 3), crystal polymorphs of compound (A) (patent documents 4, 5) , and a preparation of compound (A)

(patent documents 6, 7) have also been reported. Compound (A) and a salt thereof are recommended for oral administration in view of the easiness of self-administration, and a tablet, particularly a tablet in the dosage form for administration once per week, is desired. [0006]

The dosage form of once per week is expected to improve drug compliance of patients, whereas it requires supply of compound (A) or a salt thereof to patients in a high dose as compared to, for example, the dosage form of once per day. Since a solid preparation containing compound (A) or a salt thereof in a high dose increases its size, it may conversely degrade the drug compliance for patients, particularly infants and elderly patients having difficulty in swallowing

……………………..

SYNTHESIS

US20090275750

Compound 34 IS TRELAGLIPTIN

Figure US20090275750A1-20091105-C00078

4-Fluoro-2-methylbenzonitrile (31).

A mixture of 2-bromo-5-fluorotoluene (3.5 g, 18.5 mmol) and CuCN (2 g, 22 mmol) in DMF (100 mL) was refluxed for 24 hours. The reaction was diluted with water and extracted with hexane. The organics were dried over MgSOand the solvent removed to give product 31 (yield 60%). 1H-NMR (400 MHz, CDCl3): δ 7.60 (dd, J=5.6, 8.8 Hz, 1H), 6.93-7.06 (m, 2H), 2.55 (s, 3H).

2-Bromomethyl-4-fluorobenzonitrile (32).

A mixture of 4-fluoro-2-methylbenzonitrile (2 g, 14.8 mmol), NBS (2.64 g, 15 mmol) and AIBN (100 mg) in CClwas refluxed under nitrogen for 2 hours. The reaction was cooled to room temperature. The solid was removed by filtration. The organic solution was concentrated to give crude product as an oil, which was used in the next step without further purification. 1H-NMR (400 MHz, CDCl3): δ 7.68 (dd, J=5.2, 8.4 Hz, 1H), 7.28 (dd, J=2.4, 8.8 Hz, 1H), 7.12 (m, 1H), 4.6 (s, 2H).

Alternatively, 32 was made as follows.

4-Fluoro-2-methylbenzonitrile (1 kg) in DCE (2 L) was treated with AIBN (122 g) and heated to 75° C. A suspension of DBH (353 g) in DCE (500 mL) was added at 75° C. portionwise over 20 minutes. This operation was repeated 5 more times over 2.5 hours. The mixture was then stirred for one additional hour and optionally monitored for completion by, for example, measuring the amount of residual benzonitrile using HPLC. Additional AIBN (e.g., 12.5 g) was optionally added to move the reaction toward completion. Heating was stopped and the mixture was allowed to cool overnight. N,N-diisopropylethylamine (1.3 L) was added (at <10° C. over 1.5 hours) and then diethyl phosphite (1.9 L) was added (at <20° C. over 30 min). The mixture was then stirred for 30 minutes or until completion. The mixture was then washed with 1% sodium metabisulfite solution (5 L) and purified with water (5 L). The organic phase was concentrated under vacuum to afford 32 as a dark brown oil (3328 g), which was used without further purification (purity was 97% (AUC)).

2-(6-Chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile (33).

A mixture of crude 3-methyl-6-chlorouracil (0.6 g, 3.8 mmol), 2-bromomethyl-4-fluorobenzonitrile (0.86 g, 4 mmol) and K2CO(0.5 g, 4 mmol) in DMSO (10 mL) was stirred at 60° C. for 2 hours. The reaction was diluted with water and extracted with EtOAc. The organics were dried over MgSOand the solvent removed. The residue was purified by column chromatography. 0.66 g of the product was obtained (yield: 60%). 1H-NMR (400 MHz, CDCl3): δ 7.73 (dd, J=7.2, 8.4 Hz, 1H), 7.26 (d, J=4.0 Hz, 1H), 7.11-7.17 (m, 1H), 6.94 (dd, J=2.0, 9.0 Hz, 1H), 6.034 (s, 2H), 3.39 (s, 3H). MS (ES) [m+H] calc’d for C13H9ClFN3O2, 293.68; found 293.68.

Alternatively, 33 was made as follows.

To a solution of 6-chloro-3-methyluracil (750 g) and N,N-diisopropylethylamine (998 mL) in NMP (3 L) was added (at <30° C. over 25 min) a solution of 32 (2963 g crude material containing 1300 g of 32 in 3 L of toluene). The mixture was then heated at 60° C. for 2 hours or until completion (as determined, for example, by HPLC). Heating was then stopped and the mixture was allowed to cool overnight. Purified water (3.8 L) was added, and the resultant slurry was stirred at ambient temperature for 1 hour and at <5° C. for one hour. The mixture was then filtered under vacuum and the wet cake was washed with IPA (2×2.25 L). The material was then dried in a vacuum oven at 40±5° C. for 16 or more hours to afford 33 as a tan solid (>85% yield; purity was >99% (AUC)).

TFAsalt OF TRELAGLIPTIN

2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile (34).

2-(6-Chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile (300 mg, 1.0 mmol), (R)-3-amino-piperidine dihydrochloride (266 mg, 1.5 mmol) and sodium bicarbonate (500 mg, 5.4 mmol) were stirred in a sealed tube in EtOH (3 mL) at 100° C. for 2 hrs. The final compound was obtained as TFA salt after HPLC purification. 1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, 1H), 7.16-7.27 (m, 2H), 5.46 (s, 1H), 5.17-5.34 (ABq, 2H, J 35.2, 15.6 Hz), 3.33-3.47 (m, 2H), 3.22 (s, 3H), 2.98-3.08 (m, 1H), 2.67-2.92 (m, 2H), 2.07-2.17 (m, 1H), 1.82-1.92 (m, 1H), 1.51-1.79 (m, 2H). MS (ES) [m+H] calc’d for C18H20FN5O2, 357.38; found, 357.38.

FREE BASE NOF TRELAGLIPTIN

Alternatively, the free base of 34 was prepared as follows. A mixture of 33 (1212 g), IPA (10.8 L), (R)-3-amino-piperidine dihydrochloride (785 g), purified water (78 mL) and potassium carbonate (2.5 kg, powder, 325 mesh) was heated at 60° C. until completion (e.g., for >20 hours) as determined, for example, by HPLC. Acetonitrile (3.6 L) was then added at 60° C. and the mixture was allowed to cool to <25° C. The resultant slurry was filtered under vacuum and the filter cake was washed with acetonitrile (2×3.6 L). The filtrate was concentrated at 45° C. under vacuum (for >3 hours) to afford 2.6 kg of the free base of 34.

HCL salt OF TRELAGLIPTIN

The HCl salt of 34 was prepared from the TFA salt as follows. The TFA salt (34) was suspended in DCM, and then washed with saturated Na2CO3. The organic layer was dried and removed in vacuo. The residue was dissolved in acetonitrile and HCl in dioxane (1.5 eq.) was added at 0° C. The HCl salt was obtained after removing the solvent. 1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, 1H), 7.12-7.26 (m, 2H), 5.47 (s, 1H), 5.21-5.32 (ABq, 2H, J=32.0, 16.0 Hz), 3.35-3.5 (m, 2H), 3.22 (s, 3H), 3.01-3.1 (m, 1H), 2.69-2.93 (m, 2H), 2.07-2.17 (m, 1H), 1.83-1.93 (m, 1H), 1.55-1.80 (m, 2H). MS (ES) [m+H] calc’d for C18H20FN5O2, 357.38; found, 357.38.

Alternatively, the HCl salt was prepared from the free base as follows. To a solution of free base in CH2Cl(12 L) was added (at <35° C. over 18 minutes) 2 M hydrochloric acid (3.1 L). The slurry was stirred for 1 hour and then filtered. The wet cake was washed with CH2Cl(3.6 L) and then THF (4.8 L). The wet cake was then slurried in THF (4.8 L) for one hour and then filtered. The filter cake was again washed with THF (4.8 L). The material was then dried in a vacuum oven at 50° C. (with a nitrogen bleed) until a constant weight (e.g., >26 hours) to afford 34 as the HCl salt as a white solid (1423 g, >85% yield).

Succinate salt OF TRELAGLIPTIN

Figure US20080227798A1-20080918-C00001

The succinate salt of 34 was prepared from the HCl salt as follows. To a mixture of the HCl salt of 34 (1414 g), CH2Cl(7 L) and purified water (14 L) was added 50% NaOH solution (212 mL) until the pH of the mixture was >12. The biphasic mixture was stirred for 30 min and the organic layer was separated. The aqueous layer was extracted with CH2Cl(5.7 L) and the combined organic layers were washed with purified water (6 L). The organic layer was then passed through an in-line filter and concentrated under vacuum at 30° C. over three hours to afford the free base as an off-white solid. The free base was slurried in prefiltered THF (15 L) and prefiltered IPA (5.5 L). The mixture was then heated at 60° C. until complete dissolution of the free base was observed. A prefiltered solution of succinic acid (446 g) in THF (7 L) was added (over 23 min) while maintaining the mixture temperature at >57° C. After stirring at 60° C. for 15 min, the heat was turned off, the material was allowed to cool, and the slurry was stirred for 12 hours at 25±5° C. The material was filtered under vacuum and the wet cake was washed with prefiltered IPA (2×4.2 L). The material was then dried in a vacuum oven at 70±5° C. (with a nitrogen bleed) for >80 hours to afford the succinate salt of 34 as a white solid (1546 g, >90% yield).

The product was also converted to a variety of corresponding acid addition salts. Specifically, the benzonitrile product (approximately 10 mg) in a solution of MeOH (1 mL) was treated with various acids (1.05 equivalents). The solutions were allowed to stand for three days open to the air. If a precipitate formed, the mixture was filtered and the salt dried. If no solid formed, the mixture was concentrated in vacuo and the residue isolated. In this way, salts of 34 were prepared from the following acids: benzoic, p-toluenesulfonic, succinic, R-(−)-Mandelic and benzenesulfonic. The succinate was found to be crystalline as determined by x-ray powder diffraction analysis.

 Methanesulfonate salt 

In addition, the methanesulfonate salt was prepared as follows. A 10.5 g aliquot of the benzonitrile product was mixed with 400 mL of isopropylacetate. The slurry was heated to 75° C. and filtered through #3 Whatman filter paper. The solution was heated back to 75° C. and a 1M solution of methanesulfonic acid (30.84 mL) was added slowly over 10 minutes while stirring. The suspension was cooled to room temperature at a rate of about 20° C./hr. After 1 hr at room temperature, the solid was filtered and dried in an oven overnight to obtain the methanesulfonate salt.

…………………………

FORMULATION

WO2008114800A2

COMPD A IS TRELAGLIPTIN

Examples (Comparative Example IA)

Succinate of compound (A) (26.6 mg) was weighed in a glass bottle and used as Comparative Example IA. (Comparative Example 2A)

The succinate of compound (A) and microcrystalline cellulose were uniformly mixed in a mortar at a ratio of 1:10, and the mixture (226.6 mg) was weighed in a glass bottle and used as Comparative Example 2A. (Comparative Example 3A)

The succinate of compound (A) and corn starch were uniformly mixed in a mortar at a ratio of 1:5, and the mixture (126.6 mg) was weighed in a glass bottle and used as Comparative Example 3A. (Example IA) Succinate of compound (A) , mannitol and corn starch according to the formulation of Table IA were uniformly mixed in a fluid bed granulator (LAB-I, POWREX CORPORATION) , and the mixture was granulated by spraying an aqueous solution of dissolved hypromellose 2910, and dried therein. The obtained granules were passed through a sieve -(16M) to give milled granules. To the milled granules were added croscarmellose sodium, microcrystalline cellulose and magnesium stearate, and they were mixed in a bag to give granules for tableting. The granules were punched by a rotary tableting machine (Correct 19K, Kikusui Seisakusho, Ltd.) with a 6.5 mmφ punch to give a plain tablet weighting 121 mg. On the other hand, titanium oxide, yellow ferric oxide and talc were dispersed in a hypromellose 2910 aqueous solution to prepare a film coating liquid. The aforementioned coating liquid was sprayed onto the above-mentioned plain tablet in a film coating machine (Hicoater HCP-75, Freund Corporation), to give 2500 film- coated tablets containing 3.125 mg of compound (A) (free form) per tablet. Table IA

Figure imgf000028_0001

………………………..

POLYMORPHS AND SYNTHESIS

WO2008067465A1

FORM A

Form A may be prepared by crystallization from the various solvents and under the various crystallization conditions used during the polymorph screen (e.g., fast and slow evaporation, cooling of saturated solutions, slurries, and solvent/antisolvent additions). Tables B and C of Example 3 summarize the procedures by which Form A was prepared. For example, Form A was obtained by room temperature slurry of an excess amount of Compound I in acetone, acetonitrile, dichloromethane, 1,4-dioxane, diethyl ether, hexane, methanol, isopropanol, water, ethylacetate, tetrahydrofuran, toluene, or other like solvents on a rotating wheel for approximately 5 or 7 days. The solids were collected by vacuum filtration, and air dried in the hood. Also, Form A was precipitated from a methanol solution of Compound I by slow evaporation (SE).

[0091] Form A was characterized by XRPD, TGA, hot stage microscopy, IR, Raman spectroscopy, solution 1H-NMR, and solid state 13C-NMR.

[0092] Figure 1 shows a characteristic XRPD spectrum (CuKa, λ=1.5418A) of Form A. The XRPD pattern confirmed that Form A was crystalline. Major X-Ray diffraction lines expressed in °2Θ and their relative intensities are summarized in Table 1.

Table 1. Characteristic XRPD Peaks (CuKa) of Form A

Figure imgf000018_0001
Figure imgf000019_0001

Characterization Data of Form A of Compound I

Figure imgf000064_0001

8. Amorphous Form

[0137] The Amorphous Form of Compound I was prepared by lyophilization of an aqueous solution of Compound I (Example 10). The residue material was characterized by XRPD and the resulting XRPD spectrum displayed in Figure 26. The XRPD spectrum shows a broad halo with no specific peaks present, which confirms that the material is amorphous. The material was further characterized by TGA, DSC, hot stage microscopy, and moisture sorption analysis.

Table A. Approximate Solubilities of Compound I

 Compound I having the formula
Figure imgf000076_0002
Figure imgf000052_0001
Figure imgf000053_0001
POLYMORPH SCREEN

Crystallization Experiments of Compound I from Solvents

Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001

a) FE = fast evaporation; SE = slow evaporation; RT = room temperature; SC = slow cool;CC = crash cool, MB = moisture sorption/desorption analysis b) qty = quantity; PO = preferred orientation

…………………………

SYNTHESIS

WO2008033851A2

EXAMPLES

1. Preparation of 2-[6-(3-Amino-piperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydro- 2H-pyrimidin-l-ylmethyl]-4-fluoro-benzonitrile and pharmaceutically acceptable salts

Figure imgf000039_0001
Figure imgf000039_0002

4-Fluoro-2-methylbenzonitrile (3)

[0166] A mixture of 2-bromo-5fluorotoluene ( 2) (3.5 g, 18.5 mmol) and CuCN (2 g, 22 mmol) in DMF (100 mL) was re fluxed for 24 hours. The reaction was diluted with water and extracted with hexane. The organics were dried over MgSO4 and the solvent removed to give product 3 (yield 60%). 1H-NMR (400 MHz, CDCl3): δ 7.60 (dd, J=5.6, 8.8 Hz, IH), 6.93-7.06 (m, 2H), 2.55 (s, 3H). 2-Bromomethyl-4-fluorobenzonitrile (4)

[0167] A mixture of 4-fluoro-2-methylbenzonitrile (3) (2 g, 14.8 mmol), NBS (2.64 g, 15 mmol) and AIBN (100 mg) in CCl4 was refluxed under nitrogen for 2 hours. The reaction was cooled to room temperature. The solid was removed by filtration. The organic solution was concentrated to give crude product as an oil, which was used in the next step without further purification.1H-NMR (400 MHz, CDCl3): δ 7.68 (dd, J= 5.2, 8.4 Hz, IH), 7.28 (dd, J= 2.4, 8.8 Hz, IH), 7.12 (m, IH), 4.6 (s, 2H).

2-(6-Chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-l-ylmethyl)-4-fluoro- benzonitrile (6)

[0168] A mixture of crude 3-methyl-6-chlorouracil (5) (0.6 g, 3.8 mmol), 2- Bromomethyl-4-fluorobenzonitrile (0.86 g, 4 mmol) and K2CO3 (0.5 g, 4 mmol) in DMSO

(10 mL) was stirred at 60 C for 2 hours. The reaction was diluted with water and extracted with EtOAc. The organics were dried over MgSO4 and the solvent removed. The residue was purified by column chromatography. 0.66 g of the product was obtained (yield: 60%). 1H-NMR (400 MHz, CDCl3): δ 7.73 (dd, 1=12, 8.4Hz, IH), 7.26 (d, J- 4.0Hz, IH), 7.11-7.17 (m, IH), 6.94 (dd, J=2.0, 9.0 Hz, IH), 6.034 (s, 2H), 3.39 (s, 3H). MS (ES) [m+H] calc’d for Ci3H9ClFN3O2, 293.68; found 293.68.

2-[6-(3-Amino-piperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-l- ylmethyl]-4-fluoro-benzonitrile, TFA salt (1) (TFA salt of Compound I)

Figure imgf000040_0001

[0169] 2-(6-Chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-l-ylmethyl)-4- fluoro-benzonitrile (5) (300 mg, 1.0 mmol), (i?)-3-amino-piperidine dihydrochloride (266 mg, 1.5 mmol) and sodium bicarbonate (500 mg, 5.4 mmol) were stirred in a sealed tube in EtOH (3 mL) at 100 0C for 2 hrs. The final compound was obtained as a TFA salt after HPLC purification. 1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, IH), 7.16-7.27 (m, 2H), 5.46 (s, IH), 5.17-5.34 (ABq, 2H, J = 35.2, 15.6 Hz), 3.33-3.47 (m, 2H), 3.22 (s, 3H), 2.98-3.08 (m, IH), 2.67-2.92 (m, 2H), 2.07-2.17 (m, IH), 1.82-1.92 (m, IH), 1.51-1.79 (m, 2H). MS (ES) [m+H] calc’d for Ci8H20FN5O2, 357.38; found, 357.38.

2-[6-(3-Amino-piperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-l- ylmethyl]-4-fluoro-benzonitrile, HCl salt

Figure imgf000041_0001

[0170] The TFA salt of Compound I was suspended in DCM, and then washed with saturated Na2CO3. The organic layer was dried and removed in vacuo. The residue was dissolved in acetonitrile and HCl in dioxane (1.5 eq.) was added at 0 C. The HCl salt was obtained after removing the solvent. 1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, IH), 7.12-7.26 (m, 2H), 5.47 (s, IH), 5.21-5.32 (ABq, 2H, J = 32.0, 16.0 Hz), 3.35-3.5 (m, 2H), 3.22 (s, 3H), 3.01-3.1 (m, IH), 2.69-2.93 (m, 2H), 2.07-2.17 (m, IH), 1.83-1.93 (m, IH), 1.55-1.80 (m, 2H). MS (ES) [m+H] calc’d for Ci8H20FN5O2, 357.38; found, 357.38.

General procedure for the preparation of salts of Compound I.

[0171] The benzonitrile product may be isolated as the free base if desired, but preferably, the product may be further converted to a corresponding acid addition salt. Specifically, the benzonitrile product (approximately 10 mg) in a solution of MeOH (1 mL) was treated with various acids (1.05 equivalents). The solutions were allowed to stand for three days open to the air. If a precipitate formed, the mixture was filtered and the salt dried. If no solid formed, the mixture was concentrated in vacuo and the residue isolated. In this way, salts of Compound I were prepared from the following acids: benzoic, p-toluenesulfonic, succinic, R-(-)-Mandelic and benzenesulfonic. [0172] The isolation and/or purification steps of the intermediate compounds in the above described process may optionally be avoided if the intermediates from the reaction mixture are obtained as relatively pure compounds and the by-products or impurities of the reaction mixture do not interfere with the subsequent reaction steps. Where feasible, one or more isolation steps may be eliminated to provide shorter processing times, and the elimination of further processing may also afford higher overall reaction yields.

…………………..

TABLET

US20070060530

2. Exemplary formulations comprising succinate salt of 2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile

Provided are examples of tablet formulations that may be used to administer succinate salt of 2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile (Succinate salt of Compound I) according to the present invention. It is noted that the formulations provided herein may be varied as is known in the art.

The exemplary tablet formulations are as follows:

12.5 mg of Compound I (weight of free base form) per tablet
Core Tablet Formulation
(1) 2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4- 17.0 mg
dioxo-3,4-dihydro-2H-pyrimidin-1-
ylmethyl]-4-fluoro-benzonitrile (succinate salt)
(2) Lactose Monohydrate, NF, Ph, Eur 224.6 mg
(FOREMOST 316 FAST FLO)
(3) Microcrystalline Cellulose, NF, Ph, Eur 120.1 mg
(AVICEL PH 102)
(4) Croscarmellose Sodium, NF, Ph, Eur 32.0 mg
(AC-DO-SOL)
(5) Colloidal Silicon Dioxide, NF, Ph, Eur 3.2 mg
(CAB-O-SIL M-5P)
(6) Magnesium Stearate, NF, Ph, Eur 3.2 mg
(MALLINCKRODT, Non-bovine Hyqual)
TOTAL 400.0 mg
(per tablet)

…………..

US20080227798 AND US20120197018

POLYMORPHS AND SYNTHESIS

EXAMPLES Example 1 Preparation of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile succinate (Compound I)

Figure US20080227798A1-20080918-C00004

Compound I may be prepared by the follow synthetic route (Scheme 1)

Figure US20080227798A1-20080918-C00005

A. Preparation of 4-fluoro-2-methylbenzonitrile (Compound B)

Figure US20080227798A1-20080918-C00006

Compound B was prepared by refluxing a mixture of 2-bromo-5-fluoro-toluene (Compound A) (3.5 g, 18.5 mmol) and CuCN (2 g, 22 mmol) in DMF (100 mL) for 24 hours. The reaction was diluted with water and extracted with hexane. The organics were dried over MgSOand the solvent removed to give product B (yield 60%). 1H-NMR (400 MHz, CDCl3): δ 7.60 (dd, J=5.6, 8.8 Hz, 1H), 6.93-7.06 (m, 2H), 2.55 (s, 3H).

B. Preparation of 2-bromomethyl-4-fluorobenzonitrile (Compound C)

Figure US20080227798A1-20080918-C00007

Compound C was prepared by refluxing a mixture of 4-fluoro-2-methylbenzonitrile (Compound B) (2 g, 14.8 mmol), N-bromosuccinimide (NBS) (2.64 g, 15 mmol) and azo-bis-isobutyronitrile (AIBN) (100 mg) in CClunder nitrogen for 2 hours. The reaction was cooled to room temperature. The solid was removed by filtration. The organic solution was concentrated to give the crude product the form of an oil, which was used in the next step without further purification. 1H-NMR (400 MHz, CDCl3): δ 7.68 (dd, J=5.2, 8.4 Hz, 1H), 7.28 (dd, J=2.4, 8.8 Hz, 1H), 7.12 (m, 1H), 4.6 (s, 2H).

C. Preparation of 2-(6-chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile (Compound D)

Figure US20080227798A1-20080918-C00008

Compound E was prepared by stirring a mixture of crude 3-methyl-6-chlorouracil D (0.6 g, 3.8 mmol), 2-bromomethyl-4-fluorobenzonitrile (0.86 g, 4 mmol) and K2CO(0.5 g, 4 mmol) in DMSO (10 mL) at 60° C. for 2 hours. The reaction was diluted with water and extracted with EtOAc. The organics were dried over MgSOand the solvent removed. The residue was purified by column chromatography. 0.66 g of the product was obtained (yield: 60%). 1H-NMR (400 MHz, CDCl3): δ 7.73 (dd, J=7.2, 8.4 Hz, 1H), 7.26 (d, J=4.0 Hz, 1H), 7.11-7.17 (m, 1H), 6.94 (dd, J=2.0, 9.0 Hz, 1H), 6.034 (s, 2H), 3.39 (s, 3H). MS (ES) [m+H] calc’d for C13H9ClFN3O2, 293.68; found 293.68.

D. Preparation of 2-(6-chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile (Compound F)

Figure US20080227798A1-20080918-C00009

Compound F was prepared by mixing and stirring 2-(6-chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile (Compound E) (300 mg, 1.0 mmol), (R)-3-amino-piperidine dihydrochloride (266 mg, 1.5 mmol) and sodium bicarbonate (500 mg, 5.4 mmol) in a sealed tube in EtOH (3 mL) at 100° C. for 2 hrs. The final compound was obtained as trifluoroacetate (TFA) salt after HPLC purification. 1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, 1H), 7.16-7.27 (m, 2H), 5.46 (s, 1H), 5.17-5.34 (ABq, 2H, J=35.2, 15.6 Hz), 3.33-3.47 (m, 2H), 3.22 (s, 3H), 2.98-3.08 (m, 1H), 2.67-2.92 (m, 2H), 2.07-2.17 (m, 1H), 1.82-1.92 (m, 1H), 1.51-1.79 (m, 2H). MS (ES) [m+H] calc’d for C18H20FN5O2, 357.38; found, 357.38.

E. Preparation of Compound I: the succinic acid salt of 2-(6-Chloro-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl)-4-fluoro-benzonitrile

Figure US20080227798A1-20080918-C00010

The TFA salt prepared in the above step (Example 1, Step D) was suspended in DCM, and then washed with saturated Na2CO3. The organic layer was dried and removed in vacuo. The benzonitrile product (approximately 10 mg) was dissolved in MeOH (1 mL) and to which succinic acid in THF (1.05 equivalents) was added. The solutions were allowed to stand for three days open to the air. If a precipitate formed, the solid was collected by filtration. If no solid formed, the mixture was concentrated in vacuo, and the succinate salt was obtained after removing the solvent.

SUCCINATE SALT OF TRELAGLIPTIN

1H-NMR (400 MHz, CD3OD): δ. 7.77-7.84 (m, 1H), 7.12-7.26 (m, 2H), 5.47 (s, 1H), 5.21-5.32 (ABq, 2H, J=32.0, 16.0 Hz), 3.35-3.5 (m, 2H), 3.22 (s, 3H), 3.01-3.1 (m, 1H), 2.69-2.93 (m, 2H), 2.07-2.17 (m, 1H), 1.83-1.93 (m, 1H), 1.55-1.80 (m, 2H). MS (ES) [m+H] calc’d for C18H20FN5O2, 357.38; found, 357.38.

Compound I such prepared was found to be crystalline as determined by x-ray powder diffraction analysis (FIG. 1). The crystal material was designated Form A.

……………

patents

1. US 2013172377

2. WO 2011013639

3. WO 2009099172

4.WO 2009099171

5. WO 2008114807

6.WO 2008114800

7. WO 2008033851

8. WO 2007074884

9WO 2007035629

patent document 1: US2005/0261271

patent document 2: US2007/0060530

patent document 3: US2008/0287476

patent document 4: US2008/0227798

patent document 5: US2008/0280931

patent document 6: WO2008/114800

patent document 7: WO2011/013639

US7906523 * Oct 30, 2007 Mar 15, 2011 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8084605 * Nov 29, 2007 Dec 27, 2011 Kelly Ron C Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
US8188275 * Oct 30, 2007 May 29, 2012 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US8222411 * Sep 15, 2006 Jul 17, 2012 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US20090275750 * Sep 15, 2006 Nov 5, 2009 Jun Feng Dipeptidyl peptidase inhibitors
WO2013183784A1 Jun 4, 2013 Dec 12, 2013 Takeda Pharmaceutical Company Limited Solid preparation
US20080227798 * Nov 29, 2007 Sep 18, 2008 Kelly Ron C Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2h-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
US20120197018 * Feb 15, 2012 Aug 2, 2012 Kelly Ron C Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2h-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
WO2007033265A1 * Sep 13, 2006 Mar 22, 2007 Takeda Pharmaceutical Dipeptidyl peptidase inhibitors for treating diabetis
WO2007033266A2 * Sep 13, 2006 Mar 22, 2007 Takeda Pharmaceutical Dipeptidyl peptidase inhibitors for treating diabetis
WO2007033350A1 * Sep 13, 2006 Mar 22, 2007 Takeda Pharmaceutical Dipeptidyl peptidase inhibitors for treating diabetes
EP1586571A1 * Dec 21, 2004 Oct 19, 2005 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors

13 NMR TRELAGLIPTIN SUCCINATE

1H NMR TRELAGLIPTIN SUCCINATE

Rare Diseases: Newborn Screening Saves Lives Reauthorization Act


Malaria drug combo could help prevent pregnancy complications in lupus patients — ScienceDaily


Reversing Alzheimer’s II


Worry on the brain: Researchers find new area linked to anxiety — ScienceDaily