New Drug Approvals

Home » Posts tagged 'PHASE 1' (Page 9)

Tag Archives: PHASE 1

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

Blog Stats

  • 4,802,379 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

SEMAPIMOD


Semapimod cs.svg

Semapimod Mesylate

CPSI-2364,  AXD-455,  CN-1493, CNI 1493

CAS No. 352513-83-8(Semapimod base)

Cas 164301-51-3   4x HCl

 CAS 872830-80-3 (Semapimod mesylate)

MW 1129

CROHNS DISEASE, PHASE 1

N,N’-bis[3,5-bis[(E)-N-(diaminomethylideneamino)-C-methylcarbonimidoyl]phenyl]decanediamide

Decanediamide, N,N’-bis[3,5-bis[1-[(aminoiminomethyl)hydrazono]ethyl]phenyl]-, methanesulfonate

N,N’-Bis(3,5-bis(1-(carbamimidoylhydrazono)ethyl)phenyl)decanediamide

 

A nitric oxide synthesis inhibitor and a p38 MAPK inhibitor potentially for the treatment of Crohn’s disease.

 Semapimod, a small molecule known to inhibit proinflammatory cytokine activity, was studied to determine the optimal dose necessary to achieve a response in patients with moderate to severe Crohn’s disease (CD).

Crohn’s disease (CD) is a chronic inflammatory disease involving the upper and lower gastrointestinal tract and characterized by abdominal pain, weight loss, gastrointestinal bleeding and formation of fistulas between loops of bowel and from the bowel to the skin or other organs. Current therapy for active Crohn’s disease consists of symptomatic treatment, nutritional therapy, salicylates and immunosuppressants or surgical management.

Tumor necrosis factor a (TNF-a) plays a central role in the initiation and amplification of the granulomatous inflammatory reaction seen in CD (van Deventer, 1997). Increased TNF-a is present in gut mucosa as well as in stool of patients with active CD (Braegger et al, 1992). CNI-1493 is a synthetic guanylhydrazone compound that is an inhibitor of TNF-a synthesis. A monoclonal antibody to TNF, infliximab, is now approved for treatment of CD, but not all patients respond and many who do respond eventually become refractory to this treatment as well.

CNI-1493 is a synthetic compound which blocks the production of several inflammatory cytokines, including TNF. Because it blocks production of multiple inflammatory mediators, it may be more active than products targeted to a specific cytokine. In addition, as it is not a biologic, it should not cause hypersensitivity reactions or induce formation of antibodies.

The purpose of this trial is to determine if CNI-1493 is safe and effective in treating patients with moderate to severe Crohn’s Disease in a placebo controlled setting………https://clinicaltrials.gov/ct2/show/NCT00038766

Semapimod (INN), formerly known as CNI-1493, is an investigational new drug which has anti-inflammatory,[1] anti-cytokine,[2] immunomodulatory,[3] antiviral[4] and antimalarial[5] properties.

History

Semapimod was developed at the former Picower Institute for Medical Research, and is now licensed to Cytokine PharmaSciences. In 2000, Cytokine PharmaSciences licensed anti-infective applications of semapimod to Axxima Pharmaceuticals, but Axxima became insolvent in Dec. 2004 and its assets were acquired by GPC Biotech, which has recently merged into Agennix AG[1]. Although the disposition of Axxima’s partial rights to semapimod was not specified in these merger announcements, Cytokine PharmaSciences does not currently list any licensees for semapimod on its website.

Mechanism of action

Semapimod was first developed to inhibit nitric oxide synthesis by inflammatory macrophages, via inhibition of the uptake of arginine which macrophages require for nitric oxide synthesis.[1] Subsequently it was found that suppression of nitric oxide synthesis occurred even at semapimod concentrations 10-fold less than required for inhibition of arginine uptake, suggesting that this molecule was a more general inhibitor of inflammatory responses.[2] Further work revealed that semapimod suppressed the translation efficiency of tumor necrosis factor production.[6] Specifically, semapimod was found to be an inhibitor of p38 MAP kinase activation.[7] Surprisingly, however, the primary mode of action in vivo is now thought to be via stimulation of the vagus nerve, thereby down-regulating inflammatory pathways via the recently discovered cholinergic anti-inflammatory pathway.[8][9]

Pharmacology and clinical trials

In a preclinical study in rats, semapimod was found to suppress cytokine-storm induction by the anticancer cytokine interleukin-2 (IL-2) without decreasing its anticancer properties, allow larger doses of IL-2 to be administered.[10] A subsequent phase I trial in humans failed to show an increase in the tolerated dose of IL-2, although indications of pharmacological activity as an inhibitor of tumor necrosis factor production were observed.[11]

In a preliminary clinical trial of semapimod in patients with moderate to severe Crohn’s disease, positive clinical changes were observed, including endoscopic improvement, positive responses in some patients not responding to infliximab, healing of fistulae, and indications for tapering of steroids; no significant adverse effects were observed.[12]

In a small clinical trial against post-ERCP pancreatitis, significant suppression was not observed, although investigators observed a significant reduction of the incidence of hyperamylasemia and the levels of post-ERCP amylase.[13]

In the clinical trials above, semapimod tetrahydrochloride was administered by intravenous injection. This route has drawbacks such as dose-limiting phlebitis.[2] Recently Cytokine PharmaSciences has announced the development of novel salt forms of semapimod which are said to be orally absorbable; a phase I clinical trial of one of these salt forms, CPSI-2364, has been completed, and a phase II trial is planned for 2010.[3][4]

Chemistry

Semapimod is synthesized by reacting 3,5-diacetylaniline[14] with sebacoyl chloride in the presence of pyridine, followed by reaction of the resulting tetraketone with aminoguanidine hydrochloride.[1]

PATENT

 

  • N,N′-bis(3,5-diacetylphenyl) decanediamide tetrakis (amidinohydrazone) tetrahydrochloride (CNI-1493), which has the following structural formula:

 

SYNTHESIS

The reaction of decanedioyl dichloride (I) with 3,5-diacetylaniline (II) by means of pyridine in dichloromethane gives the corresponding diamide (III), which is condensed with aminoguanidine (IV) in refluxing aqueous ethanol to afford the target tetrakis amidinohydrazone.  EP 0746312; EP 1160240; US 5599984; WO 9519767

http://www.google.com/patents/EP0746312A1?cl=en

References

 

 

 

Semapimod.png

 

Patent Submitted Granted
NORMALIZATION OF CULTURE OF CORNEAL ENDOTHELIAL CELLS [US2015044178] 2012-12-27 2015-02-12
Patent Submitted Granted
Neural tourniquet [US2005282906] 2005-12-22
Guanylhydrazone Salts, Compositions, Processes of Making, and Methods of Using [US2008262090] 2008-10-23
Protective role of semapimod in necrotizing enterocolitis [US7795314] 2007-12-06 2010-09-14
METHOD OF TREATING ILEUS BY PHARMACOLOGICAL ACTIVATION OF CHOLINERGIC RECEPTORS [US2011112128] 2011-05-12
Method of treating ileus by pharmacological activation of cholinergic receptors [US2007213350] 2007-09-13
Pharmaceutically active aromatic guanylhydrazones [US2005171176] 2005-08-04
Guanylhydrazone salts, compositions, processes of making and methods of using [US7244765] 2006-01-19 2007-07-17
GUANYLHYDRAZONE SALTS, COMPOSITIONS, PROCESSES OF MAKING, AND METHODS OF USING [US8034840] 2008-06-19 2011-10-11
METHOD FOR TREATING GLIOBLASTOMAS AND OTHER TUMORS [US2014323576] 2014-03-14 2014-10-30
Methods of treatment of fatty liver disease by pharmacological activation of cholinergic pathways [US8865641] 2012-06-14 2014-10-21
Semapimod
Semapimod cs.svg
Semapimod sf.gif
Systematic (IUPAC) name
N,N’-bis[3,5-bis[N-(diaminomethylideneamino)-C-methylcarbonimidoyl]phenyl] decanediamide tetrahydrochloride
Identifiers
CAS Number 164301-51-3 Yes
352513-83-8 (base)
ATC code None
PubChem CID: 5745214
UNII 9SGW2H1K8P Yes
ChEMBL CHEMBL2107779
Chemical data
Formula C34H56Cl4N18O2
Molecular mass 890.73984 g/mol

see………http://worlddrugtracker.blogspot.in/2015/12/semapimod.html

/////////Semapimod Mesylate,  CPSI-2364,  AXD-455,  CN-149, PHASE 1, FERRING, CNI 1493

CC(=NN=C(N)N)C1=CC(=CC(=C1)NC(=O)CCCCCCCCC(=O)NC2=CC(=CC(=C2)C(=NN=C(N)N)C)C(=NN=C(N)N)C)C(=NN=C(N)N)C

Zidebactam, WCK 5107 in PHASE 1 FROM WOCKHARDT


Figure imgf000036_0001

2D chemical structure of 1436861-97-0

Zidebactam,  WCK 5107

Wockhardt Limited

Useful for treating bacterial infections

CAS 1436861-97-0, UNII: YPM97423DB, Wockhardt Biopharm

Molecular Formula, C13-H21-N5-O7-S
Molecular Weight, 391.4029

Disclosed in PCT International Patent Application No. PCT/IB2012/054290D

  • 01 Aug 2015 Phase-I clinical trials in Bacterial infections (In volunteers, Combination therapy) in USA (IV) (NCT02532140)

trans- sulphuric acid mono-[2-(N’-[(R)-piperidin-3-carbonyl]-hydrazinocarbonyl)-7-oxo-l,6-diaza-bicyclo[3.2.1]oct-6-yl] ester

(2S, 5R)-sulphuric acid mono-[2-(N’-[(R)-piperidin-3-carbonyl]-hydrazinocarbonyl)-7-oxo-l,6-diaza-bicyclo[3.2.1]oct-6-yl] ester

(1R,2S,5R)-l,6-Diazabicyclo [3.2.1] octane-2-carboxylic acid, 7-oxo-6-(sulfooxy)-, 2-[2-[(3R)-3-piperidinylcarbonyl]hydrazide]

trans- sulphuric acid mono-[2-(N’-[(R)-piperidin-3-carbonyl]-hydrazinocarbonyl)-7-oxo-l,6-diaza-bicyclo[3.2.1]oct-6-yl] ester

(2S, 5R)-sulphuric acid mono-[2-(N’-[(R)-piperidin-3-carbonyl]-hydrazinocarbonyl)-7-oxo-l,6-diaza-bicyclo[3.2.1]oct-6-yl] ester

(lR,2S,5R)-l,6-Diazabicyclo [3.2.1] octane-2-carboxylic acid, 7-oxo-6-(sulfooxy)-, 2-[2-[(3R)-3 -piperidinylcarbonyl] hydrazide]

1,6-Diazabicyclo(3.2.1)octane-2-carboxylic acid, 7-oxo-6-(sulfooxy)-, 2-(2-((3R)-3-piperidinylcarbonyl)hydrazide), (1R,2S,5R)-


Zidebactam potassium
  cas is  1706777-49-2

 

Zidebactam sodium ………..below
2D chemical structure of 1706777-46-9UNII-NHY7N0Y9DG.png

Cas 1706777-46-9

Sodium;[(2S,5R)-7-oxo-2-[[[(3R)-piperidine-3-carbonyl]amino]carbamoyl]-1,6-diazabicyclo[3.2.1]octan-6-yl] sulfate

UNII-NHY7N0Y9DG; NHY7N0Y9DG; Zidebactam sodium; Zidebactam sodium, (-)-; 1,6-Diazabicyclo(3.2.1)octane-2-carboxylic acid, 7-oxo-6-(sulfooxy)-, 2-(2-((3R)-3-piperidinylcarbonyl)hydrazide), sodium salt (1:1), (1R,2S,5R)-; 1706777-46-9;

Molecular Formula: C13H20N5NaO7S
Molecular Weight: 413.381969 g/mol

 

 

In September 2015, the drug was reported to be in phase I clinical trial.One of the family members US09132133, claims a combination of sulbactam and WCK-5107.

Bacterial infections continue to remain one of the major causes contributing towards human diseases. One of the key challenges in treatment of bacterial infections is the ability of bacteria to develop resistance to one or more antibacterial agents over time. Examples of such bacteria that have developed resistance to typical antibacterial agents include: Penicillin-resistant Streptococcus pneumoniae, Vancomycin-resistant Enterococci, and Methicillin-resistant Staphylococcus aureus. The problem of emerging drug-resistance in bacteria is often tackled by switching to newer antibacterial agents, which can be more expensive and sometimes more toxic. Additionally, this may not be a permanent solution as the bacteria often develop resistance to the newer antibacterial agents as well in due course. In general, bacteria are particularly efficient in developing resistance, because of their ability to multiply very rapidly and pass on the resistance genes as they replicate.

Treatment of infections caused by resistant bacteria remains a key challenge for the clinician community. One example of such challenging pathogen is Acinetobacter baumannii (A. baumannii), which continues to be an increasingly important and demanding species in healthcare settings. The multidrug resistant nature of this pathogen and its unpredictable susceptibility patterns make empirical and therapeutic decisions more difficult. A. baumannii is associated with infections such as pneumonia, bacteremia, wound infections, urinary tract infections and meningitis.

Therefore, there is a need for development of newer ways to treat infections that are becoming resistant to known therapies and methods. Surprisingly, it has been found that a compositions comprising cefepime and certain nitrogen containing bicyclic compounds (disclosed in PCT/IB2012/054290) exhibit unexpectedly synergistic antibacterial activity, even against highly resistant bacterial strains.

 

 

http://chem.sis.nlm.nih.gov/chemidplus/structure/1436861-97-0?maxscale=30&width=300&height=300

PATENT

http://www.google.com/patents/WO2013030733A1?cl=en

Figure imgf000022_0001

Scheme-1

Figure imgf000023_0001

function with Boc group)

o ormua –

Scheme-2

 

Example-2 trans-sulfuric acid mono-r2-(N,-r(R)-piperidin-3-carbonyll-hvdrazinocarbonyl)-7-oxo-l,6- diaza-bicyclo Γ3.2.11 oct-6-νΠ ester

Figure imgf000036_0001

Step-1: Preparation of trans-3-[N’-(6-benzyloxy-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2- carbonyl)-hydrazinocarbonyl]-(R)-piperidin-l-carboxylic acid tert-butyl ester:

By using the procedure described in Step-1 of Example- 1 above, and by using trans-6- benzyloxy-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carboxylic acid (25 gm, 0.084 mol), N,N- dimethyl formamide (625 ml), EDC hydrochloride (24 gm, 0.126 mol), HOBt (16.96 gm, 0.126 mol), (R)-N-tert-butoxycarbonyl-piperidin-3-carboxylic acid hydrazide (21.40 gm , 0.088 mol) to provide the title compound in 17.0 gm quantity, 41% yield as a white solid.

Analysis: MS (ES+) CzsHasNsOe = 502.1 (M+l);

I^NMR (CDCI3) = 8.40 (br s, IH), 7.34-7.44 (m, 5H), 5.05 (d, IH), 4.90 (d, IH), 4.00 (br d, IH), 3.82 (br s, IH), 3.30 (br s, IH), 3.16-3.21 (m, IH), 3.06 (br d, IH), 2.42 (br s, IH), 2.29-2.34 (m, IH), 1.18-2.02 (m, 4H), 1.60-1.75 (m, 4H), 1.45-1.55 (m, 2H),1.44 (s, 9H).

Step-2: Preparation of trans-3-[N’-(6-hydroxy-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2- carbonyl)-hydrazinocarbonyl]-(R)-piperidin-l-carboxylic acid tert-butyl ester:

By using the procedure described in Step-2 of Example- 1 above, and by using trans-3- [N ‘ -(6-benzyloxy-7-oxo- 1 ,6-diaza-bicyclo [3.2.1 ]octane-2-carbonyl)-hydrazinocarbonyl] -(R)- piperidin-l-carboxylic acid tert-butyl ester (16.5 gm , 0.033 mol), methanol (170 ml) and 10% palladium on carbon (3.5 gm) to provide the title compound in 13.5 gm quantity as a pale pink solid and it was used for the next reaction immediately.

Analysis: MS (ES+) CiglfeNsOe = 411.1 (M+l);

Step-3: Preparation of tetrabutylammonium salt of trans-3-[N’-(6-sulfooxy-7-oxo-l,6-diaza- bicyclo [3.2.1] octane-2-carbonyl)-hydrazinocarbonyl] -(R)-piperidin- 1 -carboxylic acid tert- butyl ester:

By using the procedure described in Step-3 of Example- 1 above, and by using trans-3- [N’-(6-hydroxy-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carbonyl)-hydrazinocarbonyl]-(R)- piperidin-1 -carboxylic acid tert-butyl ester (13.5 gm , 0.033 mol), pyridine (70 ml) and pyridine sulfur trioxide complex (26.11 gm, 0.164 mol), 0.5 N aqueous potassium dihydrogen phosphate solution (400 ml) and tetrabutylammonium sulphate (9.74 gm, 0.033 mol) to provide the title compound in 25 gm quantity as a yellowish solid, in quantitative yield.

Analysis: MS (ES-)

Figure imgf000037_0001

as a salt = 490.0 (M-l) as a free sulfonic acid;

Step-4: trans-sulfuric acid mono-[2-(N’-[(R)-piperidin-3-carbonyl]-hydrazinocarbonyl)-7- oxo-l,6-diaza-bicyclo[3.2.1]oct-6-yl]ester:

By using the procedure described in Step-4 of Example- 1 above, and by using tetrabutylammonium salt of trans-3-[N’-(6-sulfooxy-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2- carbonyl)-hydrazinocarbonyl]-(R)-piperidin-l-carboxylic acid tert-butyl ester (24 gm , 0.032 mmol), dichloromethane (60 ml) and trifluoroacetic acid (60 ml) to provide the title compound in 10 gm quantity as a white solid, in 79% yield.

Analysis: MS (ES-)= C13H21N5O7S = 390.2 (M-l) as a free sulfonic acid;

HXNMR (DMSO-d6) = 9.97 (d, 2H), 8.32 (br s, 2H), 4.00 (br s, IH), 3.81 (d, IH), 3.10-3.22 (m, 3H), 2.97-3.02 (m, 2H), 2.86-2.91 (m, IH), 2.65-2.66 (m, IH), 1.97-2.03 (m, IH), 1.57-1.88 (m, 7H).

-32.6°, (c 0.5, water).

PATENT

http://www.google.com/patents/WO2015059643A1?cl=en

Both, cefepime and a compound of Formula (I) may be present in the composition in their free forms or in the form of their pharmaceutically acceptable derivatives (such as salts, pro-drugs, metabolites, esters, ethers, hydrates, polymorphs, solvates, complexes, or adducts).

Individual amounts of a compound of Formula (I) or a stereoisomer or a pharmaceutically acceptable derivative thereof, and cefepime or pharmaceutically acceptable derivative thereof in the composition may vary depending on clinical requirements. In some embodiments, a compound of Formula (I) or a stereoisomer or a pharmaceutically acceptable derivative thereof in the composition is present in an amount from about 0.01 gram to about 10 gram. In some other embodiments, cefepime or a pharmaceutically acceptable derivative thereof in the composition is present in an amount from about 0.01 gram to about 10 gram.

 

PATENT

http://www.google.com/patents/WO2015063653A1?cl=en

PATENT

WO 2015110885

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015110885

Formula (I)

(a) hydrogenolysis of a compound of Formula (II) to obtain a compound of Formula (III);

convertin a compound of Formula (III) to a compound of Formula (IV);

Example 1

Synthesis of (25, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I):

Step-1: Preparation of (25, 5R)-6-hydroxy-7-oxo-2-[((3R)-iV-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (III):

(25, 5i?)-6-benzyloxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazino-carbonyl] -l,6-diazabicyclo[3.2.1]octane (II) (130 g, 0.259 mol) was dissolved in methanol (1040 ml) to obtain a clear solution. To this solution, was added 10% palladium on carbon (13 g, 0.26 mol). The suspension was stirred under 230-250 psi hydrogen atmosphere at temperature of about 30 °C for about 2 hour. The catalyst was filtered over celite bed and catalyst containing bed was washed with additional methanol (400 ml). The methanolic solution was re-filtered through fresh celite bed and washed with methanol (100 ml). The filtrate was concentrated under vacuum at temperature of about 30°C to obtain the off white solid as product. The so obtained solid was stirred with cyclohexane (750 ml). The solid was then filtered and washed with cyclohexane (320 ml) and dried under suction to obtain 107 g of (25, 5i?)-6-hydroxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo [3.2.1]octane (III).

Analysis:

Mass: 412.4 (M+l); for Molecular Formula of C18H29N5O6 and Molecular Weight of 411.5; and

Purity as determined by HPLC: 98.02%.

Step-2: Preparation of tetrabutylammonium salt of (25, 5R)-6-sulfooxy-7-oxo-2-[((3R)-iV-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo[3.2.1] octane (IV):

A solution of (25, 5i?)-6-hydroxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (III) (106 g, 0.26 mol) in dichloromethane was charged with triethyl amine (110 ml, 0.78 mol) under stirring. To this clear solution was added pyridine sulfur trioxide complex (82.5 g, 0.53 mol) under nitrogen atmosphere and stirred at temperature of about 30°C for about 2 hour. The reaction mixture was diluted with 0.5 N aqueous potassium dihydrogen phosphate solution (2100 ml) followed by ethyl acetate (2100 ml). The turbid solution was stirred for 15 minute and then the layers were separated. The aqueous layer was washed with dichloromethane (530 ml) and then with ethyl acetate (1060 ml). Tetrabutyl ammonium sulfate (79 g, 0.23 mol) was added to the separated aqueous layer and stirred for 12 hour. The extraction of the product was done using dichloromethane as solvent (1150 ml x 2). The organic layer was dried over sodium sulfate and then evaporated under vacuum at temperature below 40°C to furnish 108 g of tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo

[3.2.1] octane (IV).

Analysis:

Mass: 490.3 (M-l) as free sulfonic acid; for Molecular Formula of Ci8H28N509S.N(C4H9)4 and Molecular weight of 733.0; and

Purity as determined by HPLC: 86.50 %.

Step-3: Preparation of (25, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I):

Tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo[3.2.1]octane (IV) (88 g, 0.12 mol) was dissolved in dichloromethane (225 ml). The reaction mass was cooled to about -10°C and to this trifluoroacetic acid (225 ml) was added slowly. The reaction mixture was stirred for 1 hour at temperature of about -10°C. The solvent was removed under high vacuum at about 30°C. The residue (280 g) was stirred with diethyl ether (1320 ml) for 1 hour. The precipitated solid was filtered and the cake was washed with fresh diethyl ether (440 ml). This process was repeated with fresh diethyl ether (1320 ml + 440 ml). The obtained white solid was dried at temperature of about 30°C and suspended in acetone (1320 ml). The pH of the suspension was adjusted to 6.5-7.0 using 10% solution of sodium 2-ethyl hexanoate in acetone. The resulting suspension was filtered under suction and the wet cake was washed with acetone (440 ml) to provide the crude solid. The solid was further dried under vacuum at 40°C to yield 40 g of (25, 5i?)-7-oxo-6-sulphooxy-2-[((3i?)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I).

Analysis:

Mass: 392.2 (M+l); for Molecular formula of C13H21N5O7S and Molecular Weight of 391.4;

Purity as determined by HPLC: 92.87%; and

Melting point as determined by DSC: 274°C.

Example 2

Synthesis of Pure (25, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I):

Step-1: Preparation of (25, 5R)-6-hydroxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (III):

The procedure for the synthesis of (25, 5i?)-6-hydroxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (III) is same as given in Step- 1 of Example 1.

Step-2: Preparation of tetrabutylammonium salt of (25, 5R)-6-sulfooxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo[3.2.1] octane (IV):

A solution of (25, 5i?)-6-hydroxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (III) (106 g, 0.26 mol) in dichloromethane was charged with triethylamine (110 ml, 0.78 mol) under stirring to provide a clear solution. To this clear solution was added pyridine sulfur trioxide complex (82.5 g, 0.53 mol) under nitrogen atmosphere and stirred at temperature of about 30 °C for 2 hours. The reaction mixture was diluted with 0.5 N aqueous potassium dihydrogen phosphate solution (2100 ml) followed by ethyl acetate (2100 ml). The turbid solution was stirred for 15 minutes and then the layers were separated. The aqueous layer was washed with dichloromethane (530 ml) and then with ethyl acetate (1060 ml) respectively. Tetrabutyl ammonium sulfate (79 g, 0.23 mol) was added to the separated aqueous layer and stirred for 12 hours. The extraction of the product was done using dichloromethane as solvent (1150 ml x 2). Aliquot of the organic layer was dried over sodium sulfate for purity check. Considering the purity of the product as obtained above, silica gel (530 g) was added to the dichloromethane layer and stirred for 1 hour. This was filtered and again silica was taken in dichloromethane (3200 ml) and stirred for 45 minutes and filtered. Combined dichloromethane layer was filtered through the celite bed again and washed with additional 200 ml dichloromethane. The solvent was removed to obtain 88 g of tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-!, 6-diaza-bicyclo[3.2.1]octane (IV) as white foam.

Analysis:

Mass: 490.3 (M-l) as a free sulfonic acid; for Molecular Formula of Ci8H28N509S.N(C4H9)4 and Molecular Weight of 733.0; and

Purity as determined by HPLC: 98.34%.

Step-3: Preparation of (25, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I):

The above obtained tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo[3.2.1]octane (IV) having purity of more than 98% (88 g, 0.12 mol) was dissolved in dichloromethane (225 ml). The reaction mass was cooled to temperature of about -10°C and to this trifluoroacetic acid (225 ml) was added slowly. The reaction mixture was stirred for 1 hour at about -10°C. The solvent was removed under high vacuum at temperature of about 30°C. The residue (280 g) was stirred with diethyl ether (1320 ml) for 1 hour. The precipitated solid was filtered and the cake was washed with fresh diethyl ether (440 ml). This process was repeated with fresh diethyl ether (1320 ml + 440 ml). The obtained white solid was dried at about 30°C and suspended in acetone (1320 ml). The pH of the suspension was adjusted to 6.5-7.0 using 10% solution of sodium 2-ethyl hexanoate in acetone. The resulting suspension was filtered under suction and the wet cake was washed with acetone (440 ml) to provide the crude solid. The solid was further dried under vacuum at 40°C to yield 40 g of (25, 5i?)-7-oxo-6-sulphooxy-2-[((3i?)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I).

Analysis:

Mass: 392.2 (M+l); for Molecular Formula of C13H21N5O7S and Molecular Weight of 391.4; and

Purity as determined by HPLC: 98.7%.

Recovery of tetrabutylammonium salt of (25, 5R)-6-sulfooxy-7-oxo-2-[((3R)-iV-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1] octane (IV):

The silica recovered from the Step-2 was stirred with dichloromethane containing 2%

methanol (2000 ml) for one hour. Silica was filtered, washed with additional same composition of solvents (500 ml). Combined dichloromethane was filtered through the celite bed and washed with same composition of solvents (200 ml), evaporated to afford 1 1 g of tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l , 6-diaza-bicyclo[3.2.1] octane (IV) as off white solid.

Repeating Step-3 with the above obtained tetrabutylammonium salt of (25, 5R)-6-sulfooxy-7-oxo-2- [((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl] – 1 , 6-diaza-bicyclo [3.2.1] octane (IV) produced additional 7 g of compound of Formula (I).

Analysis:

Mass: 392.2 (M+l); for Molecular Formula of CnH^NsOvS and Molecular Weight of 391.4;

Purity as determined by HPLC: 98.7%; and

Assay as determined by HPLC: 104% against reference standard of compound of Formula (I).

Example 3

Preparation of amorphous form of (25, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl] – 1, 6-diaza-bicyclo[3.2. l]octane (I) :

Tetrabutylammonium salt of (25, 5i?)-6-sulfooxy-7-oxo-2-[((3i?)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l, 6-diaza-bicyclo[3.2.1]octane (IV) (60 g, 0.081 mol), obtained in Step-2 of Example-2 was dissolved in dichloromethane (150 ml, 2.5 volume) to obtain a clear solution. Reaction mass was cooled to about -10°C and to it trifluoroacetic acid (150 ml) was slowly added. The reaction mixture was stirred for 1 hour at about – 10°C. The solvent was removed under high vacuum at about 30°C. Diethyl ether (600 ml x 3) was added to the residue ( 184 g) and stirred for 15 minute every time. The solvent was decanted off and the residue was washed with acetonitrile (600 ml x 3). This process was also repeated with dichloromethane (600 ml x 3). The off white solid was

isolated and dried under high vacuum at about 35 °C for 3 hour to obtain 33 g of amorphous form of (25, 5i?)-7-oxo-6-sulphooxy-2-[((3i?)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I). The XRD is shown in Figure 1.

Analysis:

Mass: 392.2 (M+l); for Molecular Formula of C13H21N5O7S and Molecular Weight of 391.4;

HPLC purity: 92.26%; and

Melting point as determined by DSC: 210°C (loss of moisture below 100°C).

Example 4

Preparation of crystalline form of (25, 5R)-7-oxo-6-sulpho-oxy-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I):

The (25, 5i?)-7-oxo-6-sulphooxy-2-[((3i?)-piperidine-3-carbonyl)-hydrazino carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I) obtained as white solid (40 g) in Step-3 of Example 2 was dissolved in demineralised water (40 ml) to obtain a clear solution. To this isopropyl alcohol (280 ml) was added under stirring at room temperature. The obtained turbid solution became sticky initially then slowly started to convert into white solid, stirring continued for about 17 hours at temperature of about 30°C. The precipitated solid was filtered and washed with water: isopropyl alcohol mixture (20 ml: 140 ml). White solid was dried under high vacuum at temperature of about 45 °C for 5 hours to get 34 g of crystalline form of (25, 5i?)-7-oxo-6-sulphooxy-2-[((3i?)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1] octane (I).

Analysis:

Mass: 392.2 (M+l) for Molecular Formula of C13H21N5O7S and Molecular Weight of 391.4;

Purity as determined by HPLC: 98.7%;

Assay as determined by HPLC: 104% against reference standard of compound of Formula (I); and

Melting point as determined by DSC: 278°C (9% loss of moisture at 143-152°C).

X-ray powder diffraction pattern comprising a peak selected from the group consisting of 10.31 (± 0.2), 10.59 (± 0.2), 12.56 (± 0.2), 13.84 (± 0.2), 15.65 (± 0.2), 18.19 (± 0.2), 18.51(± 0.2), 20.38 (± 0.2), 20.65 (± 0.2), 24.30 (± 0.2), 24.85 (± 0.2) and 25.47 (± 0.2) degrees 2 theta.

 

PATENT

WO 2014135931

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2014135931

Scheme 1.

Formula (I)

 

 

preparation of a compound of Formula (I), comprising:

Formula (I)

(a) reacting a compound of Formula (II) with a compound of Formula (III) to obtain a compound of Formula (IV);

Formula (II) Formula (III)

Formula (IV)

(b) hydrogenolysis of a compound of Formula (IV) to obtain a compound of Formula

X. Formula (V)

(c) sulfonating a compound of Formula (V) to obtain a compound of Formula (VI); and

Formula (VI)

(d) converting a compound of Formula (VI) into a compound of Formula (I).

 

Example -1

Preparation of (R)-N-Boc-piperidine-3-carboxylic acid hydrazide (II):

Step-1: Preparation of (R)-Ethyl-N-Boc-piperidine-3-carboxylate (VIII)

To a solution of (R)-N-Boc-piperidine-3-carboxylic acid (1 kg. 4.36 mol) in N,N-dimethylacetamide (3 L) was charged potassium carbonate (0.664 kg, 4.80 mol) under mechanical stirring and the resulting suspension was stirred for 30 minutes at room temperature. To the reaction mass, ethyl iodide (0.75 kg, 4.80 mol) was charged via addition funnel and the reaction mass was stirred for 15 minutes at room temperature followed by at 50°C for 1 hour. The reaction was monitored using TLC (ethyl acetate: hexane 1:1). After the reaction was complete, the reaction mass was allowed to cool to room temperature and diluted with ethyl acetate (5 L). The suspension was filtered under suction and the wet cake was washed with ethyl acetate (5 L). The filtrate was stirred with 5% w/v sodium thio sulfate (15 L) and layers were separated. The aqueous layer was re-extracted with additional ethyl acetate (5 L). The combined organic layer was washed with water (5 L) and dried over sodium sulfate. The organic layer was evaporated under vacuum to provide semi-solid which solidifies upon standing as (R)-ethyl-N-Boc-piperidine-3-carboxylate in 1.1 kg quantity in 99.5% yield.

Analysis:

NMR: (CDC13): 4.63 (q, 2H), 3.90 (d, 1H), 2.87-2.95 (m, 2H), 2.73 (td, 1H), 2.32-2.39 (m, 1H), 1.66-2.01 (m, 2H), 1.52-1.68 (m, 2H), 1.39 (s, 9H), 1.19 (t, 3H).

Mass: (M+l): 258.1 for C13H23N04;

Step-2: Preparation of (R)-N-Boc-piperidine-3-carboxylic acid hydrazide (II):

(R)-N-Boc-ethyl-piperidine-3-carboxylate (1.1 kg, 4.28 mol) was liquefied by warming and transferred to a round bottom flask (10 L), to this was charged hydrazine hydrate (0.470 kg, 9.41 mol) and stirring was started. The reaction mixture was stirred at about 120°C to 125°C for 5 hours. As the TLC showed (Chloroform: methanol 9:1) completion of reaction, the reaction mixture was cooled to room temperature and diluted with water (5.5 L) followed by dichloromethane (11 L) and was stirred for 20 minutes. The layers were separated and aqueous layer was extracted with additional dichloro methane (5.5 L). Combined organic layer was washed with water (2.75 L). The organic layer was dried over sodium sulfate and evaporated under vacuum to provide a thick gel which upon stirring and seeding in the presence of cyclohexane (5.5 L) provided white solid. The suspension was filtered and wet cake was washed with fresh cyclohexane (0.5 L). The cake was dried at 35°C under vacuum to provide (R)-N-Boc-piperidine-3-carboxylic acid hydrazide as a white solid in 0.90 kg quantity in 87% yield.

Analysis

NMR: (CDC13): 7.42 (br s, 1H), 3.92 (d, 1H), 3.88 (s, 2H), 3.54-3.65 (br s, 1H), 3.17 (br t, 1H), 2.98 (br s, 1H), 2.22-2.32 (br s, 1H), 1.82-1.90 (br m, 2H), 1.76 (s, 1H), 1.60-1.70 (m, 1H), 1.45 (s, 9H).

Mass (M+l): 244.1 for C11H21N303.

Specific rotation: [ ]25D = -53.5° (c 0.5, Methanol).

HPLC purity: 99%

Example 2

Preparation of (2S, 5R)-7-oxo-6-sulphooxy-2-[((3R)-piperidine-3-carbonyl)- hydrazinocarbonyl] -l,6-diaza-bicyclo[3.2.1]octane (I):

Step-1: Preparation of (2S, 5R)- 6-benzyloxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl] – 1 ,6-diaza-bicyclo [3.2.1 ] octane(IV) :

Sodium (2S, 5R)-7-oxo-6-benzyloxy-l,6-diaza-bicyclo[3.2.1]octane-2-carboxylate (III, 200 gm, 0.67 mol; prepared using a method disclosed in Indian Patent Application No 699/MUM/2013) was dissolved in water (2.8 L) to obtain a clear solution under stirring at room temperature. To the clear solution was added successively, (R)-N-Boc-piperidine-3-carboxylic acid hydrazide (171 gm, 0.70 mol), EDC hydrochloride (193 gm, 1.01 mol), and HOBt (90.6 gm, 0.67 mol) followed by water (0.56 L) under stirring at 35°C. The reaction mixture was stirred at 35°C for 20 hours. As maximum precipitation was reached, TLC (acetone: hexane 35:65) showed completion of reaction. The suspension was filtered under

suction and the wet cake was washed with additional water (2 L). The wet cake was suspended in warm water (10 L) and stirred for 5 hours. It was filtered under suction and dried under vacuum at 45°C to furnish (2S, 5R)-6-benzyloxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (IV) as a white powder in 270 gm quantity in 87% yield.

Analysis

NMR: (CDC13): 8.40 (br s, 1H), 7.34-7.44 (m, 5H), 5.05 (d, 1H), 4.90 (d, 1H), 4.00 (br d, 1H), 3.82 (br s, 1H), 3.30 (br s, 1H), 3.16-3.21 (m, 1H), 3.06 (br d, 1H), 2.42 (br s, 1H), 2.29-2.34 (m, 1H), 1.18-2.02 (m, 4H), 1.60-1.75 (m, 4H), 1.45-1.55 (m, 2H),1.44 (s, 9H).

Mass: (M+l) = 502.1 for C25H35N506

HPLC purity: 98.4%

Step-2: Preparation of (2S, 5R)-6-hydroxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2. l]octane (V):

(2S,5R)-6-benzyloxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazino-carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (153 gm, 0.305 mol) was dissolved in methanol (1.23 L) to obtain a clear solution. To this solution, was added 10% Pd-C (15.3 gm, 50% wet) catalyst. The suspension was stirred for 3 hours under 100 psi hydrogen atmosphere at 35°C. As reaction showed completion on TLC (TLC system methanol: chloroform 10:90), the catalyst was filtered through celite under suction. The catalyst was washed with additional methanol (600 ml). The filtrate was evaporated under vacuum below 40°C to provide a crude residue. The residue was stirred with cyclohexane (1.23 L) for 1 hour. The solid was filtered at suction and the wet cake was washed with additional cyclohexane (0.25 L) to furnish (2S, 5R)-6-hydroxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2.1]octane (V) in 125 gm quantity as a solid in quantitative yield. The product being unstable was used immediately for the next reaction.

Analysis:

NMR: (CDC13): 9.0 (br s, 2H), 4.01 (br d, 2H), 3.80 (br s, 1H), 3.74 (br s, 1H), 3.48 (s, 1H), 3.13-3.26 (m, 3H), 2.96 (br s, 1H), 2.47 (br s, 1H), 2.28-2.32 ( br dd, 1H), 2.08 (br s, 1H), 1.90-2.0 (m, 3H),1.65-1.80 (m, 3H) 1.44 (s, 9H).

Mass: (M-l): 410.3 for C18H29N506

HPLC purity: 96.34%

Step-3: Preparation of Tetrabutyl ammonium salt of (2S, 5R)-6-sulfooxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazinocarbonyl]- 1 ,6-diaza-bicyclo[3.2.1 ] octane (VI) :

A solution of (2S, 5R)-6-hydroxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazino carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (113 gm, 0.274 mol), in dichloromethane (1.13 L) was charged with triethylamine (77 ml, 0.548 mol) under stirring to provide a clear solution. To the clear solution, was added pyridine sulfur trioxide complex (57 gm, 0.356 mol) under stirring at 35°C. The reaction mixture was stirred for 3 hours. The reaction mixture was worked up by adding 0.5 M aqueous potassium dihydrogen phosphate (1.13 L) followed by ethyl acetate (2.26 L) and the biphasic mixture was stirred for 15 minutes at 35°C. Layers were separated. Aqueous layer was re-extracted with dichloromethane ethyl acetate mixture (1:2 v/v, 2.26 L twice). Layers were separated. To the aqueous layer, was added solid tetrabutyl ammonium hydrogen sulfate (84 gm, 0.247 mol) and stirring was continued for 3 hours at room temperature. Dichloromethane (1.13 L) was added to the reaction mixture. Layers were separated. The aqueous layer was re-extracted with additional dichloromethane (0.565 L). Layers were separated. To the combined organic layer was added silica gel (226 gm) and the suspension was stirred for 1 hour. Suspension was filtered and silica gel was washed with dichloromethane (1 L). The combined filtrate was evaporated under vacuum to provide solid mass. To the solid mass was added cyclohexane (0.9 L) and stirred till complete solidification occurred (about 1 to 2 hours). The suspension was filtered under suction and the wet cake was dried under vacuum below 40°C to furnish tetrabutyl ammonium salt of (2S, 5R)-6-sulfooxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazino carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (VI) as a white solid in 122 gm quantity in 60% yield.

Analysis

NMR: (CDC13): 8.50 (br s, 2H), 4.32 (br s, 1H), 3.97 (d, 2H), 3.15-3.37 (m, 12H), 2.43 (br s, 1H), 2.33 (d, 1H), 2.10-2.2 (br m, 1H), 1.84-1.95 (m, 3H), 1.60-1.73 (m, 13H), 1.39-1.48 (m, 19H), 0.98 (t, 12H).

Mass: (M-l): 490.4 as a free sulfonic acid for C18H28N509S.N(C4H9)4;

HPLC purity: 96.3%

Step-4: Synthesis of (2S, 5R)-6-sulfooxy-7-oxo-2-[((3R)-piperidine-3-carbonyl)-hydrazinocarbonyl]-l,6-diaza-bicyclo[3.2. l]octane (I):

Tetra-butyl ammonium salt of (2S, 5R)-6-sulfooxy-7-oxo-2-[((3R)-N-Boc-piperidine-3-carbonyl)-hydrazino carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (113 gm, 0.154 mol) was dissolved in dichloromethane (280 ml) and to the clear solution was slowly added trifluoroacetic acid (280 ml) between 0 to 5°C. The reaction mixture was stirred between 0 to 5°C for 1 hour. The solvent and excess trifluoroacetic acid was evaporated under vacuum below 40°C to approximately 1/3 of it’s original volume to provide pale yellow oily residue. The oily residue was stirred with diethyl ether (2.25 L) for 1 hour to provide a suspension. The precipitate was filtered under suction and transferred to a round bottom flask, to it was added diethyl ether (1.1 L) under stirring. The suspension was stirred for 30 minutes and filtered under suction to provide a solid. The solid was charged in a round bottom flask and to it was added acetone (1.130 L). The pH of suspension was adjusted to 4.5 to 5.5 by adding 10% solution of sodium-2-ethyl hexanoate in acetone carefully. The resulting suspension was filtered under suction and the wet cake was washed with acetone (550 ml) to provide a crude solid. The obtained solid was dried under vacuum below 40°C to furnish 65 gm of a crude mass. The crude mass was dissolved in water (65 ml) under stirring and to the clear solution was added isopropyl alcohol (455 ml). The suspension was stirred for 24 hours and filtered under suction. The wet cake was washed with isopropyl alcohol (225 ml) and dried under vacuum below 40°C to provide a crystalline (2S, 5R)-6-sulfooxy-7-oxo-2-[((3R)-piperidine-3-carbonyl)-hydrazino carbonyl]-l,6-diaza-bicyclo[3.2.1]octane (I) free from impurities in 48 gm quantity in 80% yield.

Analysis:

NMR: (DMSO-d6) = 9.97 (d, 2H), 8.32 (br s, 2H), 4.00 (br s, IH), 3.81 (d, IH), 3.10-3.22 (m, 3H), 2.97-3.02 (m, 2H), 2.86-2.91 (m, IH), 2.65-2.66 (m, IH), 1.97-2.03 (m, IH), 1.57-1.88 (m, 7H).

Mass: (M-l): 390.3 for C13H21N507S

HPLC purity: 95.78%

Specific rotation: [(X]25D: – 32.6° (c 0.5, water)

X-ray powder diffraction pattern comprising peak at (2 Theta Values): 10.28 (+ 0.2), 10.57 (± 0.2), 12.53 (± 0.2), 13.82 (± 0.2), 15.62 (± 0.2), 18.16 (± 0.2), 18.49 (± 0.2), 20.35 (+ 0.2), 20.64 (± 0.2), 21.33 (+ 0.2), 22.99 (+ 0.2), 23.18 (+ 0.2), 24.27 (± 0.2), 24.81 (+ 0.2), 25.45 (± 0.2), 29.85 (+ 0.2), 30.45 (± 0.2), 32.39 (+ 0.2), 36.84 (± 0.2).

REFERENCES

Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of WCK-5107 Alone and in Combination With Cefepime (NCT02532140)  https://clinicaltrials.gov/show/NCT02532140
ClinicalTrials.gov Web Site 2015, September 01, To evaluate the safety,tolerability and pharmacokinetics of single intravenous doses of WCK 5107 alone and in combination with cefepime in healthy adult human subjects.

WO2013030733A1 * Aug 24, 2012 Mar 7, 2013 Wockhardt Limited 1,6- diazabicyclo [3,2,1] octan-7-one derivatives and their use in the treatment of bacterial infections
WO2014135931A1 * Oct 12, 2013 Sep 12, 2014 Wockhardt Limited A process for preparation of (2s, 5r)-7-oxo-6-sulphooxy-2-[((3r)-piperidine-3-carbonyl)-hydrazino carbonyl]-1,6-diaza-bicyclo [3.2.1]- octane
IB2012054290W Title not available

Mr Habil Khorakiwala, Chairman, Wockhardt Ltd.

///////see………http://apisynthesisint.blogspot.in/2015/11/wck-5107-in-phase-1-from-wockhardt.html

SEE BACTAM SERIES…………..http://apisynthesisint.blogspot.in/p/bactam-series.html

C1C[C@H](CNC1)C(=O)NNC(=O)[C@@H]2CC[C@@H]3C[N@]2C(=O)N3OS(=O)(=O)O

or

O=C(NNC(=O)[C@@H]2CC[C@@H]1CN2C(=O)N1OS(=O)(=O)O)[C@@H]3CCCNC3

C1CC(CNC1)C(=O)NNC(=O)C2CCC3CN2C(=O)N3OS(=O)(=O)[O-].[Na+]

SCYX 7158


SCYX-7158

[4-fluoro-N-(1-hydroxy-3,3-dimethyl-1,3-dihydro-benzo[c]oxaborol-6-yl-2-trifluoromethyl benzamide]

4-Fluoro-N-(1-hydroxy-3,3-diméthyl-1,3-dihydro-2,1-benzoxaborol-6-yl)-2-(trifluorométhyl)benzamide
Benzamide, N-(1,3-dihydro-1-hydroxy-3,3-dimethyl-2,1-benzoxaborol-6-yl)-4-fluoro-2-(trifluoromethyl)-
4-fluoro-N-(l-hydroxy-3,3-dimethyl-l,3-dihydro- benzo[c][l,2]oxaborol-6-yl-2-trifluoromethyl benzamide
4-fluoro-N-(l-hydroxy-3,3-dimethyl-l,3-dihydro- benzo[c][l,2]oxaborol-6-yl)-2-trifluoromethyl benzamide
SCYX-7158
1266084-51-8
UNII-2IOR2OO3GW
AN 5568
PHASE 1..Anacor Pharmaceuticals Drugs for Neglected Diseases Initiative, Trypanosomiasis, African (Sleeping sickness)
SEE……Future Medicinal Chemistry (2011), 3(10), 1259-1278.
  • C17H14BF4NO3
  • Average mass 367.103 Da

 

Human African trypanosomiasis (HAT) is an important public health problem in sub-Saharan Africa, affecting hundreds of thousands of individuals. An urgent need exists for the discovery and development of new, safe, and effective drugs to treat HAT, as existing therapies suffer from poor safety profiles, difficult treatment regimens, limited effectiveness, and a high cost of goods. We have discovered and optimized a novel class of small-molecule boron-containing compounds, benzoxaboroles, to identify SCYX-7158 as an effective, safe and orally active treatment for HAT.

The presence of a boron atom in the heterocyclic core structure has been found essential for trypanocidal activity of orally active series of benzoxaborole-6-carboxamides in murine models of human African trypanosomiasis. SCYX-7158  has been identified as an effective, safe and orally active treatment for human African trypanoso-miasis to enter preclinical studies, with expected progression to phase 1 clinical trials in 2011 ………http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3764666/

21. Jacobs RT, Plattner JJ, Nare B, Wring SA, Chen D, Freund Y, et al. Benzoxaboroles: a new class of potential drugs for human African trypanosomiasis. Future Med Chem. 2011;3:1259–1278. [PubMed]
22. Jacobs RT, Nare B, Wring SA, Orr MD, Chen D, Sligar JM, et al. SCYX-7158, an orally-active benzoxaborole for the treatment of stage 2 human African trypanosomiasis. PLoS Negl Trop Dis. 2011;5:e1151. [PMC free article] 

Figure 1. Chemical structures of compounds.

 

A drug discovery project employing integrated biological screening, medicinal chemistry and pharmacokinetic characterization identified SCYX-7158 as an optimized analog, as it is active in vitro against relevant strains of Trypanosoma brucei, including T. b. rhodesiense and T. b. gambiense, is efficacious in both stage 1 and stage 2 murine HAT models and has physicochemical and in vitro absorption, distribution, metabolism, elimination and toxicology (ADMET) properties consistent with the compound being orally available, metabolically stable and CNS permeable.

In a murine stage 2 study,SCYX-7158 is effective orally at doses as low as 12.5 mg/kg (QD×7 days). In vivo pharmacokinetic characterization of SCYX-7158 demonstrates that the compound is highly bioavailable in rodents and non-human primates, has low intravenous plasma clearance and has a 24-h elimination half-life and a volume of distribution that indicate good tissue distribution.

Most importantly, in rodents brain exposure of SCYX-7158 is high, with Cmax >10 µg/mL and AUC0–24 hr >100 µg*h/mL following a 25 mg/kg oral dose. Furthermore, SCYX-7158 readily distributes into cerebrospinal fluid to achieve therapeutically relevant concentrations in this compartment.

 

Medicinal Chemistry Synthesis of SCYX-7158  SCHEME1

While the original route was eff ective for producing multi-gram quantities of the API, it was not amenable to scale-up. The route started with 2, a relatively expensive aryl boronic acid. This was protected as borocan 3 and halogen-lithium exchange followed by reaction with acetone and subsequent deprotection provided the oxaborole 4. This protection/alkylation/deprotection sequence added two steps to the overall synthesis and the metalation was not reliable. However, the biggest concern in the sequence was nitration of 4 to give 5. This was accomplished by adding a concentrated solution of 4 to cold fuming nitric acid. Besides the signifi cant safety considerations, the reaction did not scale well. Reduction of the nitro group to give aniline 6 was followed by amide formation to provide 1. While this end game was effi cient, the material produced was dark in color. The colored impurities were not removed by crystallization of 1 and furthermore a mixture of two polymorphs was formed under the original conditions.

 

 

Scheme 2 – Process Chemistry Synthesis of SCYX-7158

The process chemistry route to SCYX-7158 is shown in Scheme 2. When considering alternative routes to 1, the readily available and inexpensive methyl 2-bromobenzoate (8) was identifi ed as an attractive starting point. Gratifyingly, treatment of 8 with methylmagnesium bromide aff orded 2-bromocumyl alcohol (9) in high yield using simple operating conditions. Lithiumhalogen exchange followed by reaction with triisopropyl borate and acidic work-up provided benzoxaborole 4, along with cumyl alcohol (10). While this conversion was not completely atom-effi cient, it was easily scalable and several strategies are available to suppress the by-product in the future.

With benzoxaborole 4 in hand, attention turned to the introduction of a nitrogen-linked amide at the C(6) position. This was accomplished using the same nitration/reduction/acylation strategy used in Scheme 1. Yet signifi cant changes to the chemistry were required for safety and reliability reasons. The fi rst task was introduction of the nitrogen. Nitration was demonstrated using acetic anhydride/nitric acid. However, due to slow rates of nitration and potential for accumulation of a reactive intermediate, alternative conditions had to be identifi ed. These limitations were overcome by use of trifl uoroacetic anhydride/nitric acid, which provided a more reactive nitrating intermediate, thus improving the rate of nitration and aff ording a process in which nitric acid was slowly added until 4 was consumed. Full safety assessment of the nitration reaction, including extensive calorimetry studies, demonstrated the safety of this reaction. This process was used to prepare kilogram quantities of 5.

Following reduction of nitrobenzoxaborole 5 to aniline 6 under standard catalytic hydrogenation conditions, acylation with 7 provided the fi nal drug candidate in high chemical yield. Two challenges remained which needed to be addressed through further optimization of the process. The fi rst challenge was color and purity of the API, which derived from a highly colored impurity generated in the nitration reaction which carried through to fi nal product and was not removed by crystallization. The second challenge was to consistently obtain a single polymorph of the API. Both challenges were addressed by isolation of crystalline isopropyl boronate 11 which rejected colored impurities, followed by regeneration of 1 through addition of water and azeotropic removal of isopropanol. This crystallization provided the API as a single polymorph. The API was isolated in good yield, very high purity and was white in color.

 

PATENT

https://www.google.co.in/patents/WO2011019616A1?cl=en

N-(3,3-Dimethyl-l-phenyl-2,3-dihvdro-lH-benzotblborol-6-yl)-4-fluoro-2- trifluoromethylbenzatnide

HNO3

Figure imgf000104_0001
Figure imgf000104_0002This is not the compd, see precursor

To a suspension of 2-bromophenylboronic acid (75.Og, 373.4 mmol) in toluene (525 niL) was added JV-butyldiethanolamine (64.ImL, 392.1 mmol, 1.05 equiv.) via a syringe. The mixture was heated at 50 0C for two hours. After cooling to room temperature, the toluene was evaporated under reduced pressure and the remaining clear colorless oil was treated with heptanes (500 mL). The heptanes mixture was then sonicated for 5 min and the resulting suspension was allowed to stand at room temperature overnight. The solid that precipitated was collected by filtration, washed with heptanes, and dried in a vacuum oven overnight to yield 2-(2′- bromophenyl)-6-butyl[l,3,6,2]dioxazaborocan as a white solid. Data: 1H NMR (400 MHz, CHLOROFORM-^) δ ppm 0.86 (t, J=7.4 Hz, 3 H) 1.14 – 1.25 (m, 2 H) 1.51 – 1.62 (m, 2 H) 2.61 – 2.70 (m, 2 H) 3.01 – 3.11 (m, 2 H) 3.26 – 3.37 (m, 2 H) 4.09 – 4.26 (m, 4 H) 7.10 (td, J=7.6, 2.0 Hz, 1 H) 7.24 (td, J=7.3, 1.1 Hz, 1 H) 7.51 (d, J=7.9 Hz, 1 H) 7.81 (dd, J=IA, 1.9 Hz, 1 H). Amount obtained, 123.7 g (98.6% yield).

To a solution of 2-(2′-bromophenyl)-6-butyl[l,3,6,2]dioxazaborocan (30.0g, 89.2 mmol) in THF (740 mL) at -78 0C was added /?-BuLi (42.8 mL, 2.5M in hexane, 107.0 mmol, 1.2 equiv.) dropwise via a syringe over a period of 10 min while maintaining reaction temperature at -78 0C. After the addition the reaction solution was stirred for 20 min at -78 0C before acetone (7.5 mL, 124.8 mmol, 1.4 equiv.) was added dropwise via a syringe over a period of 10 min while maintaining the reaction temperature at -78 0C. The resulting mixture was allowed to stir for 20 min at -78 0C then warm to room temperature gradually. Once the reaction vessel reached room temperature, 6N HCl solution (150 mL) was added and the mixture was stirred for an additional 30 min. The mixture was extracted with EtOAc (3X). The EtOAc extracts were dried over Na2SO4, filtered and concentrated under reduced pressure. The light yellow oil was then subjected to flash chromatography (Isco Companion, 8Og SiO2 cartridge, solid loaded SiO2, neat heptanes to 20:80 EtOAc gradient at 60 ml/min for 90 min). 3,3-Dimethyl-3H-benzo[c][l,2]oxaborol-l-ol was recovered as clear colorless oil. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.44 (s, 6 H) 7.31 (d, J=Ll Hz, 1 H) 7.38 – 7.47 (m, 2 H) 7.66 (d, J=7.2 Hz, 1 H) 8.99 (s, 1 H). Amount obtained: 9.4O g (65.2 % yield).

To 60 mL fuming HNO3 at -45 0C was slowly added a solution of 3,3- dimethyl-3H-benzo[c][l,2]oxaborol-l-ol (9.4 g, 58.0 mmol) in 11.9 mL nitrobenzene via a syringe while maintaining the reaction temperature between -40 to -45 0C. Once the addition was complete the resulting solution was allowed to stir at -45 ° C for an additional 45 min before poured into crushed ice. The ice mixture was allowed to melt and the aqueous solution was extracted with DCM (3X). The combined DCM extracts were dried over Na2SO4 then evaporated. The crude oil remaining was mixed with one liter 1 : 1 DCM/heptanes. The volume of the solution was reduced under reduced pressure by half and the resulting solution was allowed to stand overnight in a -20 0C freezer. The precipitate formed was filtered out, washed with heptanes and vacuum dried to give 3,3-dimethyl-6-nitro-3H-benzo[c][1.2]oxaborol-l-ol as a white solid. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.46 (s, 6 H) 7.69 (d, J=8.4 Hz, 1 H) 8.28 (dd, J=8.4, 2.3 Hz, 1 H) 8.48 (d, J=2.2 Hz, 1 H) 9.41 (br. s., 1 H). Amount obtained: 7.31 g (60.4 % yield).

To a solution of 3,3-dimethyl-6-nitro-3H-benzo[c][l .2]oxaborol-l-ol (6.98 g, 33.3 mol) in THF ( 277 mL) was added 6N HC1( 16.6 mL, 100.2 mmol, 3.0 equiv.). The vessel was vacuum/N2 purged three times and 5% Pd/C (3.5 g) was added. The mixture was again vacuum/N2 purged three times then vacuum purged again. H2 was then introduced from a balloon and the reaction was allowed to stir at room

temperature over night. The reaction solution was filtered through a short pad of celite and the filtrate was evaporated to yield 6-amino-3, 3 -dimethyl -3H- benzo[c][l,2]oxaborol-l-ol HCl salt as a dark brown foamy solid. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.36 (s, 6 H) 4.94 (s, 2 H) 6.66 (dd, J=8.1, 2.2 Hz, 1 H) 6.79 (d, J=2.0 Hz, 1 H) 7.01 (d, J=8.1 Hz, 1 H) 8.72 (s, 1 H). Amount obtained: 8.29 g (100% yield).

To a solution of 6-amino-3, 3 -dimethyl -3H-benzo[c][l,2]oxaborol-l-ol HCl salt (8.29 g, 33.3 mmol) in DCM (170 mL) was added Et3N (11.6 mL, 83.2 mmol, 2.5 equiv.). The mixture was cooled to 0 0C and 2-trifluoromethyl-4- fluorobenzoyl chloride (6.1 mL, 39.9 mmol, 1.2 equiv.) was added slowly via a syringe. The resulting solution was allowed to warm to room temperature gradually and stir for 2 hours. The reaction solution was diluted with DCM, washed with IN HCl, H2O, brine and then dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give an off- white solid. The solid was recrystallized from DCM/heptanes to give 4-fluoro-N-(l-hydroxy-3,3-dimethyl-l,3-dihydro- benzo[c][l,2]oxaborol-6-yl-2-trifluoromethyl benzamide as a white solid. LCMS (M/Z) : 368 (M+H); 1H NMR (DMSO-d6) δ: 10.58 (s, IH), 9.11 (s, IH), 8.02 (d, J = 1.7 Hz, IH), 7.75 – 7.83 (m, 2H), 7.60 – 7.71 (m, 2H), 7.38 (d, J = 8.2 Hz, IH), 1.44 (s, 6H). Amount obtained: 11.7 g (96% yield)………IS SCYX 7158

BELOW NOT SCYX 7158

The title compound was prepared using a similar procedure to that of N-(I- phenyl- 1 ,3 -dihydrobenzo [c] [ 1 ,2]oxaborol-6-yl)-2-trifluoromethylbenzamide with phenyl magnesium bromide replacing p-to IyI magnesium bromide and 4-fluoro-iV-(l- hydroxy-3,3-dimethyl-2,3-dihydro-lH-benzo[b]borol-6-yl)-2-trifluoromethyl benzamide replacing N-(I -hydroxy-1 ,3-dihydrobenzo[c] [ 1 ,2]oxaborol-6-yl)-2- trifiuoromethylbenzamide. Data: LCMS m/e: 428 (M+H); 1H NMR (400 MHz, DMSO-J6) δ ppm 1.59 (s, 6 H) 7.46 – 7.62 (m, 4 H) 7.71 (td, J=8.5, 2.7 Hz,l H) 7.77 – 7.90 (m, 3 H) 8.00 – 8.09 (m, 2 H) 8.39 (d, J=2.0 Hz, 1 H) 10.66 (s, 1 H). 10 N-fl-p-Tolyl-lJ-dihydro-benzofcIflJIoxaborol-ό-vD-benzatnide

 

PATENT

82 4-Fluow-N-(l-hydwxy-3,3-dimethyl-l,3-dihydw-benzofcIfl,2Ioxabowl-6- yl-2-trifluoromethyl benzamide

Figure imgf000136_0001

To a suspension of 2-bromophenylboronic acid (10. Og, 49.7 mmol) in toluene (70 niL) was added N-butyldiethanolamine (8.5 mL, 52.2 mmol, 1.05 equiv.) via a syringe. The mixture was heated at 50 0C for two hours. After cooling to room temperature, the toluene was evaporated under reduced pressure and the remaining clear colorless crude oil was treated with heptanes (~ 500 mL). The heptanes mixture was then sonicated ~ 5 min and the resulting suspension was allowed to stand at room temperature overnight. The solid that precipitated was collected by filtration, washed with heptanes, and dried in a vacuum oven overnight to yield a white solid as the titled compound. 1U NMR (400 MHz, CHLOROFORM-J) δ ppm 0.86 (t, J=7.4 Hz, 3 H) 1.14 – 1.25 (m, 2 H) 1.51 – 1.62 (m, 2 H) 2.61 – 2.70 (m, 2 H) 3.01 – 3.11 (m, 2 H) 3.26 – 3.37 (m, 2 H) 4.09 – 4.26 (m, 4 H) 7.10 (td, J=7.6, 2.0 Hz, 1 H) 7.24 (td, J=7.3, 1.1 Hz, 1 H) 7.51 (d, J=7.9 Hz, 1 H) 7.81 (dd, J=IA, 1.9 Hz, 1 H). Amount obtained, 16.0 g, (98 % yield).

To a solution of 2-(2′-bromophenyl)-6-butyl[l,3,6,2]dioxazaborocan (3.0g, 9.2 mmol) in THF (76 mL) at -78 0C was added /?-BuLi (4.4 mL, 2.5M in hexane, 11.0 mmol, 1.2 equiv.) dropwise via a syringe over a period of 10 min while maintaining reaction temperature at -78 0C. After the addition the reaction solution was stirred 20 min at -78 0C before acetone (946 μL, 12.8 mmol, 1.4 equiv.) was added dropwise via a syringe over a period of 10 min while maintaining the reaction temperature at -78 0C. The resulting mixture was allowed to stir for 20 min at -78 0C then warm to room temperature gradually. Once the reaction vessel reached room temperature, 6M HCl solution (30 mL) was added and the mixture was stirred for 30 min. The mixture was extracted with EtOAc (3X). The EtOAc extracts were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude slightly yellow in color residual oil remaining was then subjected to flash chromatography (Isco Companion, 8Og SiO2 cartridge, solid loaded SiO2, neat heptane to 20:80 EtOAc gradient at 60 ml/min for 90 min). The product was recovered as clear colorless oil. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.44 (s, 6 H) 7.31 (d, J=Ll Hz, 1 H) 7.38 – 7.47 (m, 2 H) 7.66 (d, J=7.2 Hz, 1 H) 8.99 (s, 1 H). Amount obtained: 1.76 g (61%).

To 14.2 ml fuming HNO3 at -45 0C was added a solution of 3,3-dimethyl- 3H-benzo[c][l,2]oxaborol-l-ol (2.28 g, 14.1 mmol) in 3.0 ml nitrobenzene slowly via a syringe while maintaining the reaction temperature between -40 to -45 0C. Once the addition was complete the resulting solution was allowed to stir at -45 ° C for an additional 45 min before poured into crushed ice (600 g). The ice mixture was allowed to melt and the aqueous solution was extracted with dichloromethane. The combined dichloromethane extracts were dried over Na2SO4 then evaporated. The crude oil remaining was mixed with one liter 1 : 1 DCM:heptane. The volume of the solution was reduced on a rotovap by half and the resulting solution was allowed to stand overnight in a -20 0C freezer overnight. The precipitate formed was filtered out, washed with heptanes and vacuum dried to give the titled compound as a white solid. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.46 (s, 6 H) 7.69 (d, J=8.4 Hz, 1 H) 8.28 (dd, J=8.4, 2.3 Hz, 1 H) 8.48 (d, J=2.2 Hz, 1 H) 9.41 (br. s., 1 H). Amount obtained: 2.01 g (68%).

To a solution of 3,3-dimethyl-6-nitro-3H-benzo[c][1.2]oxaborol-l-ol (790 mg, 3.8 mmol) in THF ( 20 mL) was added HOAc (1.7 mL, 30 mmol). The vessel was vacuum/N2 purged three times and 5% Pd/C (200 mg) was added. The mixture was again vacuum/N2 purged three times then vacuum purged again. H2 was then introduced from a balloon and the reaction was allowed to stir for 2.5 hours. The reaction solution was filtered through a short pad of celite and the filtrate was evaporated to yield the title compound as a dark brown foamy solid. 1H NMR (400 MHz, DMSO-J6) δ ppm 1.36 (s, 6 H) 4.94 (s, 2 H) 6.66 (dd, J=8.1, 2.2 Hz, 1 H) 6.79 (d, J=2.0 Hz, 1 H) 7.01 (d, J=8.1 Hz, 1 H) 8.72 (s, 1 H). Amount obtained: 670 mg (89%). [0382] To a solution of 6-amino-3, 3 -dimethyl -3H-benzo[c][l,2]oxaborol-l-ol acetate salt (100 mg, 0.42 mmol) in DCM (2 niL) was added Et3N ( 117.3 μL, 0.84 mmol). The mixture was cooled to 0 0C and the 2-trifluoromethyl-4-fluorobenzoyl chloride (70.0 μL, 0.46 mmol) was added slowly via a syringe. The resulting solution was allowed to warm to room temperature gradually and stir for 2 hours. The reaction solution was diluted with DCM, washed with IN HCl, H2O and then dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure and the crude material was subjected to flash chromatography (Isco Companion, 4 g SiO2 cartridge, SiO2 solid load, neat heptanes to neat EtOAc gradient over 45 min, flow rate = 18 ml/min). The title compound was recovered as a white foam. LCMS (M/Z) : 368 (M+H); 1H NMR (DMSO-d6) δ: 10.58 (s, IH), 9.11 (s, IH), 8.02 (d, J = 1.7 Hz, IH), 7.75 – 7.83 (m, 2H), 7.60 – 7.71 (m, 2H), 7.38 (d, J = 8.2 Hz, IH), 1.44 (s, 6H). Amount obtained: 144.6 mg (93% yield).

Alternate Synthesis

Figure imgf000138_0001

82e

82b

A 500 mL round-bottomed-flask equipped with a magnetic stir bar and ice- H2O bath was charged with 82a (18.4g, 85.5 mmol) and anhydrous THF (200 mL). MeMgCl (68 mL, 3.0M in 2-methylTHF) was added dropwise through an additional funnel. The mixture was allowed to warm to rt. gradually and stirred overnight. After cooling back to 0 0C, the white milky suspension was carefully treated with HCl (3M) until the upper layer turned clear with white precipitate at the bottom of the flask (pH = 6). The upper clear solution was decanted into a separatory funnel. The precipitate was rinsed with methyl tert-butyl ether (MTBE) (100 mL) 3 times. Combined MTBE with the clear solution and the mixture was washed with H2O (100 mL) 3 times, brine (100 niL), dried over MgSO4, filtered and concentrated under reduced pressure to give 82b as a light yellow oil (20.2g, 100%).

82c

A 50 mL round-bottomed-flask equipped with a magnetic stir bar and ice- H2O bath was charged with 82b (860 mg, 4.0 mmol) and anhydrous THF (20 mL). MeMgBr (1.3 mL, 2.0 M in THF) was slowly added via a syringe. The mixture was stirred at 0 0C for 10 minute and the ice bath was replaced with a dry ice-acetone bath at -40 0C. BuLi (1.9 mL, 2.5 M in hexanes) was added dropwise via a syringe. The resulting mixture was stirred at -40 0C for another 2h before B(O-ipr)3 (1.4 mL, 4.8 mmol) was added dropwise. The mixture was allowed to warm up to rt gradually and stirred overnight. After carefully quenched the reaction with H2O (1 mL), HCl (3M, 10 mL) was added and the mixture was stirred at rt for Ih. The mixture was extracted with EtOAc (20 mL) 3 times. Combined extracts was washed with H2O (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated under reduced pressure to give a clear oil. The oil solidified overnight to give 82c as a pale yellow waxy solid (544mg, 82.4%).

82d

A 3 L round-bottomed-flask equipped with a mechanical stirrer, thermocouple and ice bath was charged with 82c (86.2 g of 58 wt%, 309 mmol) and trifluoroacetic acid (259 mL). Trifluoroacetic anhydride (129 mL, 926 mmol) was added in one portion. An exotherm of 18 0C was observed. The solution was again cooled to 0 0C and 90% nitric acid (18.0 mL, 386 mmol) was added via syringe pump over 2 h. After the addition was complete, the solution was aged for 1 h. Water (1.75 L) was added. Note: Initially the quench is quite exothermic. Add the water in 5 mL aliquots until the exotherm subsides. The resulting suspension was stirred for 16 h while warming to rt. The solids were collected on a frit, rinsed with water (2 x 500 mL), and air dried to constant weight to provide 50.3 g of crude 82d as a free-flowing orange solid. Note: the crude 82d can be carried forward without recrystallization. The solid was charged to a IL three-necked round-bottomed-flask equipped with a nitrogen inlet adapter, thermocouple, heating mantle and mechanical stirrer.

Isopropylacetate (IPAc, 75 mL) was added and the resulting slurry was warmed to 75 0C and heptanes (250 mL) was added over 15 min while maintaining an internal temp of > 65 0C. The slurry was allowed to cool to rt over night. The solids were collected on a frit and rinsed with 10% IP Ac/heptanes (100 mL) and then heptanes (100 rnL). The product was air dried to constant weight to provide a tan solid (31.7 g, 58%).

82e

A 500 mL round-bottomed-flask equipped with a magnetic stir bar, thermocouple and septum was charged with 82d (29.7 g, 192 mmol) and THF (150 mL, anhydrous stabilizer free). The vessel was inerted by cycling vacuum the nitrogen three times and 5% Pd/C (6.0 g, 50% wet, Degussa type NO/W) was added. The vessel was again inerted by cycling vacuum then nitrogen three times. A hydrogen filled balloon was attached via needle and the atmosphere was changed by cycling vacuum the hydrogen three times. The slurry was stirred vigorously for 16 h. The atmosphere was changed again to nitrogen by cycling vacuum then nitrogen three times. The mixture was filtered through a 1″ pad of celite and the cake was rinsed with THF (50 mL). Concentration in vacuo provided a light tan powder (26.82 g). In a 500 mL round bottomed-flask, the solids were slurried in IPAc (50 mL) and warmed in an 80 0C water bath. Heptanes (150 mL) were added over 10 min. The resulting slurry was allowed to cool to rt and stir for 16 h. The solids were collected on a frit, rinsed with heptanes (50 mL) and air dried to provide an off- white solid (24.39 g, 96%).

4-Fluoro-N-(l-hvdroxy-3,3-dimethyl-l,3-dihvdro-benzofcJfl,2Joxaborol-6-yl-2- triβuoromethyl benzatnide

A lL three-necked round-bottomed-flask equipped with a nitrogen inlet adapter, mechanical stirrer and thermocouple was charged with 82e (15.7g, 88.4 mmol), THF (160 mL, anhydrous, stabilizer free) and K2CO3 (14.7g, 106 mmol). The suspension was stirred at rt and 4-fluoro-2-(trifluoromethyl)benzoyl chloride (22.Og, 97.3 mmol) was added over 10 min. The resulting suspension was aged for 24 h at rt. Water (80 mL) and isopropyl acetate (160 mL) were added and the phases were partitioned. The organic phase was further extracted with water (80 mL) and then brine (50 mL). The organic phase was dried over MgSO4 (20 g) and concentrated in vacuo to provide a tan solid (34.26 g). The solid was dissolved with acetone (195 mL) and transferred to a mechanically stirred IL round-bottomed-flask. Distilled water (113 mL) was added in one portion and the mixture was stirred for 30 min to produce a seed bed and then additional distilled water (60 mL) was added over 30 min. The suspension was stirred at rt overnight and the solids were collected on a frit. The cake was rinsed with 1 : 1 acetone/water (100 rnL) and air dried to constant weight to provide an off-white solid (30.5 g, 94%).

Alternate Synthesis

HNO3 CF3CO2H (CF3CO)2O

Figure imgf000141_0001

to RT

Figure imgf000141_0002
Figure imgf000141_0003

1-1

A 72 L round-bottomed-flask was equipped with a cold bath, mechanical stirrer, nitrogen inlet adaptor, oxygen sensor, thermowell and 2 L dropping funnel. The flask was charged with methyl 2-bromobenzoate (2513 g, 11.7 mol) and the system was flushed with nitrogen to <0.1% O2. THF (18L, anhydrous, inhibitor free) was added and the cold bath was charged with ice and acetone. When the internal temp reached -4 0C, MeMgBr (11.6 L of a 3M solution in ether, 34.8 mol) was added via dropping funnel over 3 h. The internal temp was maintained below 15 0C throughout. At the end of addition, the cold bath was drained and the reaction was aged overnight at ambient temperature. The bath was again charged with ice and acetone and the suspension cooled to below 15 0C. HPLC indicated incomplete conversion (92:8 product, starting ester), so additional MeMgBr (2.3L of a 3M solution in ether) was added. After Ih, HPLC showed the conversion to be >99: 1. The reaction was quenched by slow addition of IN HCl (42 L) keeping the internal temp below 15 0C throughout. At the end of the quench, the pH was adjusted to 6 with IN HCl. The mixture was extracted with MTBE (10 L then 2x5L). The combined organic phases were dried over MgSO4, filtered and concentrated via rotary evaporation to provide 2482 g of 2-(2-bromophenyl)-propan-2-ol as a pale yellow oil. 1H NMR (CHLOPvOFORM-d) δ: 7.62 – 7.67 (m, IH), 7.53 – 7.58 (m, IH), 7.24 – 7.30 (m, IH), 7.03 – 7.10 (m, IH), 1.70 – 1.75 (m, 6H). 1-2

A 72L round-bottomed-flask was equipped with a mechanical stirrer, O2 sensor, thermowell, 2L dropping funnel, N2inlet adaptor, and cold bath. The vessel was inerted to 0.01% O2 and charged with THF (27L, anhydrous, inhibitor free). The resulting solution was cooled to -70 0C using dry ice and acetone and n-BuLi (8.2 L of a 2.5M solution in heptane, 20.5 mol) was added over Ih. 2-(2-Bromophenyl)- propan-2-ol (1994 g, 9.27 mol) was dissolved in THF (9L) and the solution was added to the BuLi via dropping funnel over 2h, keeping the internal temp below -70 0C. The resulting thin yellow suspension was aged for 30 min then B(OiPr)3 (244 Ig, 13.0 mol) was added rapidly via addition funnel. The cold bath was drained and the misture was allowed to warm to room temperature while aging over night. HPLC analysis shows an 81 :19 ratio of desired product: 2-phenyl-2-propanol. The mixture was cooled to -10 0C and 2N HCl (9.3 L) was added via dropping funnel over 30 min, keeping the reaction mixture below 10 0C. After 3 h, the pH was adjusted to 4 with additional HCl. The reaction mixture was extracted with MTBE (2 x 4L). The combined organic phases were concentrated to provide 2028 g of a heavy oil. The oil was dissolved in MTBE (14L) and extracted with IN NaOH (4.6, then 5, then 4L). The aqueous phases were combined and acidified with 2N HCl (6.8 L) to a pH of 4-5. The mixture was extracted with MTBE (5L). The organic phase was dried over MgSO4 (282 g) and concentrated to provide 1450 g (ca 60 wt%) of 3,3-dimethyl-3H- benzo[c][l,2]oxaborol-l-ol as a waxy white solid. LC/MS: m/z 163 (M+H)+; 1H NMR (DMSO-de) δ: 8.96 (br. s., IH), 7.62 (d, J = 7.2 Hz, IH), 7.33 – 7.45 (m, 2H), 7.25 – 7.30 (m, IH), 1.40 (s, 6H).

1-3

A 22 L round-bottomed-flask equipped with a mechanical stirrer, thermocouple, 2 L dropping funnel and cold bath was charged with 3,3-dimethyl-3H- benzo[c][l,2]oxaborol-l-ol (508 g, 300 g contained, 1.85 mol) and trifluoroacetic acid (1.54 L). The solution was cooled to 5 0C. Trifluoroacetic anhydride (722 mL, 5.56 mol, 3.00 eq) was added via dropping funnel over 15 min. After aging at 0 – 3 0C for 30 min, nitric acid (90% fuming, 108 mL, 2.31 mol, 1.5 eq) was added dropwise over 2h 50 min keeping the internal temp below 5 0C. After aging for 1 h, icewater (10.4L) was added over 50 min maintaining the reaction temp below 15 0C to provide a slurry. The slurry was aged at 0 0C overnight to provide an orange suspension. The solids were collected on a frit, rinsed with cold water (5L) and air dried under a stream of air to constant weight (ca 24h) to provide 364 g of 3,3- dimethyl-6-nitro-3H-benzo[c][l,2]oxaborol-l-ol as a 92.4 wt% pure solid (88%). LC/MS : m/z 208 (M+H)+; 1H NMR (DMSO-d6) δ: 8.52 (d, J = 2.2 Hz, IH), 8.32 (dd, J = 8.4, 2.2 Hz, IH), 7.74 (d, J = 8.4 Hz, IH), 1.50 (s, 6H)

1-4

A 2 gallon stirred pressure vessel was charged with 3,3-dimethyl-6-nitro- 3H-benzo[c][l,2]oxaborol-l-ol (966 g, 812 g corrected, 3.92 mol), 5% Pd/C (193 g, 50% wet, Degussa type 101 NO/W) and THF (4.83 L, inhibitor free). The vessel was sealed, the atmosphere was changed to H2 (5 psi) and the reaction was fun for 16 h. An exotherm to 30 0C was observed over about 30 min. The vessel was purged with N2, and completion of reaction was determined by HPLC. The reaction was vacuum filtered through a pad of celite (very slow filtration) and the filter cake was rinsed with THF (2L). The filtrate was concentrated via rotary evaporation to provide 982 g of a dark brown solid. This was transferred to a 22L round-bottomed-flask and warmed to 80 0C in iPAc (1.83 L) to provide a dark brown slurry. The slurry was cooled to 60 0C and heptanes (5.49L) were added over 2 h. The slurry was allowed to age with stirring over night while cooling to room temperature. The solids were collected on a frit, rinsed with heptanes (4L) and air dried to provide a dark brown solid (747 g).

The solids (747 g) were transferred to a 22L rbf and slurried in iPAc (3 L) at 70 0C. The batch was allowed to cool to 40 0C and heptanes (3L) were added over 5 h. The slurry was aged at room temperature over night and the solids were collected on a frit, rinsed with 1 : 1 iP Ac/heptanes (2L) then heptanes (IL) and air dried to provide 554 g of 6-amino-3,3-dimethyl-3H-benzo[c][l,2]oxaborol-l-ol as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.36 (s, 6 H) 4.94 (s, 2 H) 6.66 (dd, J=8.1, 2.2 Hz, 1 H) 6.79 (d, J=2.0 Hz, 1 H) 7.01 (d, J=8.1 Hz, 1 H) 8.72 (s, 1 H). 4-Fluow-N-(l-hvdwxy-3,3-dimethyl-l,3-dihvdw-benzofcJfl,2Joxabowl-6-yl)-2- triβuoromethyl benzatnide

A 22L four-necked round-bottomed-flask equipped with a nitrogen inlet adapter, mechanical stirrer and thermocouple was charged with 6-amino-3,3- dimethyl-3H-benzo[c][l,2]oxaborol-l-ol (554g, 3.13 mol), THF (5.5 L, anhydrous, stabilizer free) and K2CO3 (865 g, 6.26 mol). The suspension was stirred at room temperature for 30 min and 4-fluoro-2-(trifluoromethyl)benzoyl chloride (780 g, 3.44 mol) was added over 30 min. The resulting suspension was aged for 24 h at room temperature. HPLC showed unreacted 6-amino-3,3-dimethyl-3H-benzo[c][l,2] oxaborol-1-ol so an additional 42 niL of the acid chloride was added. After 30 min, water (2.8 L) and isopropyl acetate (5.5 L) were added and the phases were partitioned. The organic phase was further extracted with water (2.8 L) and then brine (2.8 L). The organic phase was dried over MgSO4 and concentrated in vacuo to provide a tan solid. The solid was dissolved with acetone (3.0 L) and transferred to a mechanically stirred 5OL round-bottomed-flask. Distilled water (2.0 L) was added in one portion and the mixture was stirred for 30 min to produce a seed bed and then additional water (1.0 L) was added over 30 min. The suspension was stirred at room temperature overnight and the solids were collected on a frit. The cake was rinsed with 1 : 1 acetone/water (1.0 L) and air dried to constant weight to provide 4-fluoro-N- ( 1 -hydroxy-3 ,3 -dimethyl- 1 ,3 -dihydro-benzo [c] [ 1 ,2]oxaborol-6-yl)-2-trifluoromethyl benzamide as a dark tan solid (1.3 kg).

Recrystallization of4-Fluoro-N-(l-hydroxy-3,3-dimethyl-l,3-dihvdro- benzotcl t 1,21 oxaborol-6-yl)-2-trifluoromethyl benzamide

A 22 L round-bottomed-flask was charged with the dark tan crude 4- fluoro-N-(l -hydroxy-3, 3 -dimethyl- 1 ,3-dihydro-benzo[c] [ 1 ,2]oxaborol-6-yl)-2- trifluoromethyl benzamide (1.3 kg), acetone (8L) and Darco G-60 (55 g, 400 mesh) and water (5.3L). The resulting suspension was stirred for 15 min, filtered through a pad of celite (ca 500 g) to provide a brown solution. The celite pad was washed with 60% acetone/water (8L). The combined filtrate and rinse were transferred to a 50 L round-bottomed-flask and water (2L) was added. The solution was seeded (5 g) to initiate crystallization and additional water (2.2 L) was added slowly via addition funnel. After aging at room temperature overnight, the solids were collected and the filter cake was rinsed with 30% acetone/water (4L). The solids were air dried for 24 h then dried in a room temperature vacuum oven for 5 days to constant weight to provide 969 g (72% recovery) of 4-fluoro-N-(l-hydroxy-3,3-dimethyl-l,3-dihydro- benzo[c][l,2]oxaborol-6-yl)-2-trifluoromethyl benzamide as a light tan solid.

LC/MS: m/z 368 (M+H)+;

1H NMR (400 MHz, DMSO-d6) δ ppm 1.44 (s, 5 H) 1.49 (s, 2 H) 7.39 (d, J=8.2 Hz, 1 H) 7.61 – 7.76 (m, 2 H) 7.77 – 7.84 (m, 2 H) 7.86 – 7.90 (m, 0 H) 8.03 (d, J=I.7 Hz, 1 H) 9.09 (s, 1 H) 10.58 (s, 1 H).

 

POTASSIUM SALT

Formation of potassium salt

Figure imgf000145_0001

To a 50OmL three-neck flask fitted with a mechanical stirrer was charged KOH (1.51 g, 26.9 mmol, 1.0 eq.). Under a nitrogen atmosphere, anhydrous acetone (140 mL) and H2O (2.5 mL, 5 eq.) were added via syringe. A solution of 4-fluoro-N- (l-hydroxy-3,3-dimethyl-l,3-dihydro-benzo[c][l,2]oxaborol-6-yl-2-trifluoromethyl benzamide (10.0 g, 27.2 mmol, 1.0 eq.) in anhydrous acetone (60 mL) was added to the flask with vigorous stirring. The resulting clear solution was stirred at room temperature. The potassium salt precipitated from the solution over ca. 4 hours to afford a thick suspension. The precipitate was collected by filtration, washed with acetone (200 mL) and dried in a vacuum oven overnight to afford a white solid (10.6g, 91.9% yield). 1H NMR (methanol-d4) δ: 7.70 – 7.76 (m, IH), 7.53 – 7.60 (m, 2H), 7.47 – 7.53 (m, IH), 7.33 – 7.36 (m, IH), 7.01 – 7.06 (m, IH), 1.46 (s, 6H); M.P. (range) 197 – 200 0C; Elemental analysis: Theory: C 48.25%, H 3.57%, N 3.31%, K 9.24%; Found: C 48.70%, H 3.41%, N 3.25%, K 9.19%.

REFERENCES

http://journals.plos.org/plosntds/article?id=10.1371/journal.pntd.0001151

 

  • touguia J, Costa J (1999) Therapy of human African trypanosomiasis: current situation. Mem Inst Oswaldo Cruz 94: 221–224
  • Barrett MP, Boykin DW, Brun R, Tidwell RR (2007) Human African trypanosomiasis: pharmacological re-engagement with a neglected disease. Br J Pharmacol 152: 1155–1171.
  1. 1986. Epidemiology and control of African trypanosomiasis. Report of a WHO expert committee. World Health Organization. Geneva, Switzerland. Technical Report Series, No. 739. 126 pp.
  2. Benzoxaboroles: a new class of potential drugs for human African trypanosomiasis. Robert T Jacobs, Jacob J Plattner, Bakela Nare, Stephen A Wring, Daitao Chen, Yvonne Freund, Eric G Gaukel, Matthew D Orr, Joe B Perales, Matthew Jenks, Robert A Noe, Jessica M Sligar, Yong-Kang Zhang, Cyrus J Bacchi, Nigel Yarlett, and Robert Don. Future Medicinal Chemistry. August 2011. Vol. 3, No. 10. Pages 1259-1278.

http://www.swisstph.ch/fileadmin/user_upload/Pdfs/Events/2010_09_Jacobs.pdf  ……….POWERPOINT

Lead optimization investigation of oxaboroles for the treatment of human African trypanosomiasis
238th Am Chem Soc (ACS) Natl Meet (August 16-20, Washington) 2009, Abst MEDI 345

LINK

https://www.acsmedchem.org/ama/orig/abstracts/mediabstractf2009.pdf

Robert Jacobs, bob.jacobs@scynexis.com

Daitao Chen1 , Matt Orr1 , Jessica Sligar1 , Matt. Jenks1 , Andy Noe1 , Bakela Nare2 , Luke T. Mercer2 , Tana S. Bowling2 , Cindy Rewerts1 , Stephen Wring1 , Cyrus Bacchi3 , Nigel Yarllet3 , Charles Ding4 , Yvonne Freund5 , Kurt Jarnagin5 , Jacobs Plattner5 , and Robert Don6 . (1) Scynexis Inc, Duhram, NC 27713, (2) SCYNEXIS, Inc, Research Triangle Park, NC 27709-2878, (3) Pace University, New York, NY, (4) Anacor Pharmaceuticals, Inc, Palo Alto, CA, (5) Anacor Pharmaceuticals, Inc, (6) Drugs for Neglected Diseases initiative, Geneva, Switzerland

 

 

 

///////////SCYX-7158

Pevonedistat


Figure

Millennium Pharmaceuticals, Inc. INNOVATOR

Millennium Pharmaceuticals, Inc., a subsidiary of Takeda Pharmaceutical Company Limited,

MLN4924, MLN 4924-003, TAK-924

905579-51-3 BASE

1160295-21-5 HcL

A potent and selective inhibitor of NAE. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. The ubiquitin-proteasome pathway mediates the destruction of unwanted proteins.

(((1S,2S,4R)-4-{4-[(S)-2,3-Dihydro-1H-inden-1-ylamino]-7H-pyrrolo[2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl)methyl sulfamate hydrochloride) (pevonedistat), a novel NEDD8-activating enzyme (NAE) inhibitor, has demonstrated in vitro cytotoxic activity against a variety of human malignancies and is currently being developed by Takeda Pharmaceuticals Company Limited as a clinical candidate for the treatment of cancer

In 2011, orphan drug designation was assigned to MLN-4924 for the treatment of MDS and for the treatment of acute myelogenous leukemia.

PHASE 1…….CANCER SOLID TUMOR

………………….

PATENT

http://www.google.com/patents/US20120330013

preparing a compound represented by the following formula 1 by reacting the compound of formula 11 with TFA (step 9):

Figure US20120330013A1-20121227-C00001
Figure US20120330013A1-20121227-C00002

The retrosynthetic analysis of MLN4924 (1), as the final desired nucleoside, is shown in the following.

Figure US20120330013A1-20121227-C00003

MLN 4924 (1) can be synthesized by condensing cyclic sulfate 3 as the glycosyl donor with a purine base. The glycosyl donor 3 can be produced from diol 4, which in turn can be obtained from cyclopentanone 5 via a stereoselective reduction and a regioselective cleavage of the isopropylidene moiety. The cyclopentanone 5 can be synthesized from cyclopentenone 6 by stereoselective reduction. The intermediate cyclopentenone 6 can be easily derived from D-ribose according to our previously published procedure (Jeong, L. S. et al., J. Org. Chem. 2004, 69, 2634-2636).

The synthetic route for the glycosyl donor 3 is shown in the following scheme 1.

Figure US20120330013A1-20121227-C00004

Example 1 Preparation of MLN4924 Step 1: Preparation of 6-(tert-butyl-diphenyl-silanyloxymethyl)-2,2-dimethyl-tetrahydro-cyclopenta[1,3]dioxol-4-one (Compound 5)

Figure US20120330013A1-20121227-C00006

To a suspension of the compound 6 (20.0 g, 47.1 mmol) in methanol (400 ml) was added 10% palladium on activated carbon (1.0 g), and the mixture was stirred at room temperature overnight under H2 atmosphere. After filtration of the reaction mixture, the solvent was removed and the residue was dissolved in methylene chloride and then filtered through short pad silica gel. Then, the solvent was evaporated to give the compound 5 (20.1 g, 100%) as a colorless syrup.

[α]20 D −28.32 (c 1.49, MeOH); HR-MS (ESI): m/z calcd for C25H32NaO4Si [M+Na]+ 447.1968, Found 447.1956; 1H NMR (400 MHz, CDCl3) δ 7.69 (m, 4H), 7.40 (m, 6H), 4.84 (t, J=4.4 Hz, 1H), 4.22 (dd, J=1.2, 4.8 Hz, 1H), 3.96 (dd, J=8.0, 10.0 Hz, 1H), 3.82 (dd, J=6.8, 10.0 Hz, 1H), 2.37 (m, 1H), 2.30 (ddd, J=1.2, 8.4, and 18.4 Hz, 1H), 2.20 (ddd, J=1.2, 12.0, and 18.4 Hz, 1H), 1.37 (s, 3H), 1.35 (s, 3H), 1.06 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 112.6, 80.5, 77.6, 77.2, 76.9, 63.6, 38.1, 36.9, 27.1, 27.02, 27.01, 25.3, 19.5; Anal. Calcd for C25H32O4Si: C, 70.72; H, 7.60. Found: C, 70.79; H, 7.75.

Step 2: Preparation of 6-(tert-butyl-diphenyl-silanyloxymethyl)-2,2-dimethyl-tetrahydro-cyclopenta[1,3]dioxol-4-ol (Compound 7)

Figure US20120330013A1-20121227-C00007

To a suspension of the compound 5 (20.1 g, 47.1 mmol) in methanol (500 ml) were added sodium borohydride (2.17 g, 57.4 mmol) and cerium (III) chloride heptahydrate (21.3 g, 57.2 mmol) at 0° C., and the mixture was stirred at room temperature for 30 min. After the solvent was removed, the residue was partitioned between ethyl acetate and water. The organic layer was then washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate=5/1) to give the compound 7 (20.86 g, 98%) as a colorless syrup.

[α]20 D +34.55 (c 0.55, MeOH); HR-MS (ESI): m/z calcd for C25H34NaO4Si [M+Na]+: 449.2124; Found: 449.2110; 1H NMR (400 MHz, CDCl3) δ 7.69 (m, 4H), 7.39 (m, 6H), 4.62 (t, J=5.6 Hz, 1H), 4.44 (t, J=5.6 Hz, 1H), 3.89 (dd, J=6.0, 7.6 Hz, 1H), 3.84 (m, 1H), 3.68 (dd, J=6.4, 10.0 Hz, 1H), 1.91 (m, 2H), 1.26 (m, 1H), 1.42 (s, 3H), 1.33 (s, 3H), 1.05 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 135.9, 135.8, 134.2, 134.1, 129.8, 129.7, 127.8, 127.7, 110.6, 79.4, 78.9, 77.6, 77.2, 76.9, 72.5, 62.9, 41.6, 33.4, 27.0, 25.9, 27.0, 25.9, 24.4, 19.5; Anal. Calcd for C25H34O4Si: C, 70.38; H, 8.03. Found: C, 70.41; H, 8.08.

Step 3: Preparation of 3-tert-butoxy-4-(tert-butyl-diphenyl-silanyloxymethyl)-cyclopentane-1,2-diol (Compound 4)

Figure US20120330013A1-20121227-C00008

To a solution of the compound 7 (20.86 g, 47.12 mmol) in methylene chloride was added trimethylaluminum (2.0 M in toluene, 132.1 ml) at 0° C., and the mixture was stirred at room temperature for 2 days. The mixture was cooled to 0° C., slowly quenched with an aqueous saturated ammonium chloride solution, filtered, and evaporated. The residue was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate=2/1) to give the compound 4 (13.42 g, 62%) as a colorless syrup.

[α]20 D +3.30 (c 0.55, MeOH); HR-MS (ESI): m/z calcd for C26H38NaO4Si [M+Na]+: 465.2437; Found: 465.2423; 1H NMR (400 MHz, CDCl3) δ 7.70 (m, 4H), 7.41 (m, 6H), 4.05 (dd, J=4.4, 7.2 Hz, 1H), 3.93 (m, 1H), 3.72 (m, 2H), 3.59 (dd, J=3.6, 12.0 Hz, 2H), 2.70 (d, J=20.8 Hz, 1H), 2.10 (m, 2H), 1.60 (m, 1H), 1.20 (s, 9H), 1.06 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 135.9, 133.5, 130.0, 129.9, 127.9, 127.9, 77.6, 77.2, 76.9, 74.9, 73.8, 72.7, 72.1, 63.3, 42.1, 34.0, 28.5, 27.0, 19.4; Anal. Calcd for C26H38O4Si: C, 70.55; H, 8.65. Found: C, 70.61; H, 8.70.

Step 4: Preparation of (4-tert-butoxy-2,2-dioxo-tetrahydro-2-yl-6-cyclopenta[1,3,2]-dioxathiol-5-ylmethoxy)-tert-butyl-diphenyl-silane (Compound 3)

Figure US20120330013A1-20121227-C00009

To a solution of the compound 4 (13.42 g, 30.3 mmol) in methylene chloride were added triethyl amine (14.5 ml, 101.0 mmol) and thionyl chloride (3.7 ml, 47.4 mmol) at 0° C., and the reaction mixture was stirred at 0° C. for 10 minutes. The reaction mixture was partitioned between methylene chloride and water. The organic layer was washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate=6/1) to give the cyclic sulfite (14.37 g, 97%) as a white foam.

[α]20 D +20.00 (c 0.05, MeOH); HR-MS (ESI): m/z calcd for C26H36NaO5SSi [M+Na]+: 511.1950; Found: 511.1929; 1H NMR (400 MHz, CDCl3) δ 7.64 (m, 4H), 7.40 (m, 6H), 5.23 (m, 1H), 5.04 (dd, J=4.4, 6.0 Hz, 1H), 4.01 (t, J=4.8 Hz, 1H), 3.68 (dd, J=3.6, 10.4 Hz, 1H), 3.56 (dd, J=8.0, 10.4 Hz, 1H), 2.07 (m, 2H), 1.96 (m, 1H), 1.14 (s, 9H), 1.05 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 135.8, 135.7, 133.9, 133.8, 129.9, 129.9, 127.9, 127.8, 85.7, 83.2, 77.6, 77.2, 76.9, 75.0, 71.1, 62.7, 44.7, 31.4, 28.5, 27.1, 19.4; Anal. Calcd for C26H36O5SSi: C, 63.90; H, 7.42; S, 6.56. Found: C, 63.94; H, 7.45; S, 6.61.

To a solution of the cyclic sulfite obtained above (14.37 g, 29.4 mmol) in the mixture of carbon tetrachloride, acetonitrile and water (1:1:1.5, 210 ml) were added sodium metaperiodate (18.56 g, 56.4 mmol) and ruthenium chloride (1.72 g, 8.25 mmol), and the reaction mixture was stirred at room temperature for 10 minutes. The reaction mixture was partitioned between methylene chloride and water. The organic layer was washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate=4/1) to give the compound 3 (13.36 g, 90%) as a white solid.

mp 101-104° C.; [α]20 D −80.00 (c 0.05, MeOH); HR-MS (ESI): m/z calcd for C26H36NaO6SSi [M+Na]+: 527.1900; Found: 527.1881; 1H NMR (400 MHz, CDCl3) δ 7.64 (m, 4H), 7.41 (m, 6H), 5.13 (m, 1H), 4.83 (dd, J=4.4, 6.8 Hz, 1H), 4.13 (t, J=4.0 Hz, 1H), 3.92 (dd, J=6.4, 10.4 Hz, 1H), 3.69 (dd, J=5.2, 10.4 Hz, 1H), 2.11 (m, 2H), 2.02 (m, 1H), 1.15 (s, 9H), 1.05 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 135.7, 135.0, 133.8, 133.7, 130.0, 128.0, 127.9, 83.5, 82.2, 77.6, 77.2, 76.9, 75.4, 70.4, 70.4, 62.2, 43.9, 31.3, 28.2, 27.1, 26.8, 19.4; Anal. Calcd for C26H36O6SSi: C, 61.87; H, 7.19; S, 6.35. Found: C, 61.91; H, 7.14; S, 6.30.

Step 5: Preparation of 2-tert-butoxy-3-(tert-butyl-diphenyl-silanyloxymethyl)-5-[4-(indan-1-ylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-cyclopentanol (Compound 8)

Figure US20120330013A1-20121227-C00010

A suspension of N6-indanyl-7-deazaadenine (8.80 g, 35.2 mmol), sodium hydride (1.38 g, 45.7 mmol) and 18-crown-6 (9.11 g, 45.7 mmol) in THF (200 ml) was stirred at 80° C. To the reaction mixture was added a solution for the compound 3 (13.36 g, 26.5 mmol) in THF (150 ml), and the stirring was continued at 80° C. overnight. The reaction mixture was cooled down to 0° C., and conc. HCl was added slowly until pH reaches 1-2. Then the reaction mixture was further stirred at 80° C. for 2 hours. After neutralized with saturated aqueous NaHCO3 solution, the reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate=2/1) to give the compound 8 (11.62 g, 65%) as a white foam.

UV (CH2Cl2) λmax 272.5 nm; [α]20 D −8.89 (c 0.45, MeOH); HR-MS (ESI): m/z calcd for C41H51N4O3Si [M+H]+: 675.3730; Found: 675.3717; 1H NMR (400 MHz, CDCl3) δ 8.38 (s, 1H), 7.70 (m, 4H), 7.41 (m, 6H), 6.92 (d, J=3.6 Hz, 1H), 6.29 (d, J=3.2 Hz, 1H), 5.91 (dd, J=7.6, 14.8 Hz, 1H), 5.14 (br d, J=6.8 Hz, 1H), 4.77 (m, 1H), 4.36 (t, J=6.0 Hz, 1H), 4.22 (dd, J=5.2, 10.8 Hz, 1H), 3.84 (dd, J=5.6, 10.4 Hz, 1H), 3.73 (dd, J=8.4, 10.4 Hz, 1H), 3.37 (d, J=5.6 Hz, 1H), 3.06 (m, 1H), 2.95 (m, 1H), 2.75 (m, 1H), 2.75 (m, 1H), 2.58 (m, 1H), 2.38 (m, 1H), 2.15 (m, 1H), 1.98 (m, 1H), 1.65 (s, 1H), 1.55 (s, 1H), 1.16 (s, 9H), 1.07 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 156.4, 151.8, 150.3, 144.1, 143.8, 135.9, 134.0, 129.9, 128.2, 127.9, 127.9, 127.0, 125.1, 124.4, 123.3, 103.8, 97.4, 77.8, 77.6, 77.2, 76.9, 74.9, 72.4, 63.5, 62.1, 56.3, 43.9, 34.9, 30.5, 30.5, 28.5, 27.2, 19.5; Anal. Calcd for C41H50N4O3Si: C, 72.96; H, 7.47; N, 8.30. Found: C, 73.01; H, 7.45; N, 8.36.

Step 6: Preparation of {7-[3-tert-butoxy-4-(tert-butyl-diphenyl-silanyloxymethyl)-cyclopentyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}-indan-1-yl-amine (Compound 9)

Figure US20120330013A1-20121227-C00011

To a solution of the compound 8 (11.62 g, 17.2 mmol) in methylene chloride (300 ml) were added N,N-dimethylaminopyridine (5.64 g, 51.6 mmol) and phenyl chlorothionocarbonate (4.3 ml, 34.4 mmol), and the reaction mixture was stirred at room temperature overnight. After the solvent was removed, the residue was purified by silica gel column chromatography (hexane/ethyl acetate=6/1) to give the thiocarbonate (13.82 g, 99%) as a white foam.

UV (MeOH) λmax 271.50 nm; [α]20 D +10.00 (c 0.15, MeOH); HR-MS (ESI): m/z calcd for C48H55N4O4SSi [M+H]+: 811.3713; Found: 811.3687; 1H NMR (400 MHz, CDCl3) δ 8.36 (s, 1H), 7.61 (dd, J=1.6, 7.6 Hz, 4H), 7.34 (m, 5H), 7.26 (m, 4H), 7.18 (m, 6H), 6.86 (s, 1H), 6.25 (d, J=3.2 Hz, 1H), 6.00 (dd, J=3.2, 8.4 Hz, 1H), 5.83 (d, J=6.8 Hz, 1H), 5.19 (m, 1H), 5.07 (br s, 1H), 4.48 (t, J=3.6 Hz, 1H), 3.82 (dd, J=7.2, 10.4 Hz, 1H), 3.52 (dd, J=7.2, 10.0 Hz, 1H), 2.99 (m, 1H), 2.88 (m, 2H), 2.69 (m, 2H), 2.18 (dd, J=11.2, 13.6 Hz, 1H), 1.94 (m, 2H), 1.12 (s, 9H), 0.98 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 194.9, 153.5, 152.1, 143.9, 135.9, 135.8, 134.1, 129.9, 129.6, 128.3, 127.9, 127.0, 126.7, 125.1, 124.6, 123.2, 122.0, 87.9, 77.6, 77.2, 76.9, 74.6, 70.4, 63.5, 57.3, 42.8, 35.0, 30.7, 30.5, 29.9, 28.7, 27.1, 19.4; Anal. Calcd for C48H54N4O4SSi: C, 71.08; H, 6.71; N, 6.91; S, 3.95. Found: C, 71.14; H, 6.75; N, 6.95; S, 4.01.

To a solution of the thiocarbonate obtained above (13.82 g, 17.0 mmol) in toluene (200 ml) were added tri-n-butyltinhydride (9.4 ml, 34.1 mmol) and 2,2′-azo-bis-isobutyronitrile (4.32 g, 26.3 mmol), and the reaction mixture was stirred at 110° C. for 1 hour. After the mixture was cooled down, the solvent was removed. The resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate=3/1) to give the compound 9 (9.21 g, 82%) as a white foam.

UV (MeOH) λmax 272.50 nm; [α]20 D −10.00 (c 0.20, MeOH); HR-MS (ESI): m/z calcd for C41H51N4O2Si [M+H]+: 659.3781; Found: 659.3757; 1H NMR (400 MHz, CDCl3) δ 8.41 (s, 1H), 7.69 (m, 4H), 7.41 (m, 6H), 7.29 (m, 2H), 7.23 (m, 2H), 6.92 (d, J=3.6 Hz, 1H), 6.31 (d, J=3.6 Hz, 1H), 5.90 (dd, J=7.2, 14.8 Hz, 1H), 5.38 (m, 1H), 5.15 (br s, 1H), 4.33 (dd, J=5.2, 8.4 Hz, 1H), 3.88 (dd, J=6.4, 10.0 Hz, 1H), 3.68 (dd, J=7.2, 10.4 Hz, 1H), 3.05 (m, 1H), 2.96 (dd, J=7.6, 15.6 Hz, 1H), 2.76 (m, 1H), 2.45 (d, J=5.2 Hz, 1H), 2.29 (m, 2H), 2.06 (m, 1H), 1.95 (m, 2H), 1.55 (s, 1H), 1.13 (s, 9H), 1.06 (s, 9H);13C NMR (100 MHz, CDCl3) δ 156.3, 151.9, 144.1, 143.9, 135.9, 135.8, 134.3, 129.8, 128.2, 127.8, 127.0, 125.1, 124.6, 121.8, 77.6, 77.2, 76.7, 73.5, 72.2, 63.6, 56.4, 52.8, 46.8, 42.8, 34.9, 34.5, 30.5, 28.6, 27.2, 28.7, 19.4; Anal. Calcd for C41H50N4O2Si: C, 74.73; H, 7.65; N, 8.30. Found: C, 74.79; H, 7.61; N, 8.25.

Step 7: Preparation of 2-tert-butoxy-4-[4-(indan-1-ylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-cyclopentanol (Compound 10)

Figure US20120330013A1-20121227-C00012

To a solution of the compound 9 (9.21 g, 13.97 mmol) in the mixture of THF and pyridine (1:1, 160 ml) was added dropwise pyridine hydrofluoride (18.42 ml, 190.0 mmol) at 0° C., and the reaction mixture was stirred at room temperature for 1 hour. The mixture was neutralized with saturated aqueous NaHCO3 solution and partitioned between ethyl acetate and water. The organic layer was washed with brine, dried with anhydrous MgSO4, filtered, and evaporated. Then, the residue was purified by silica gel column chromatography (hexane/ethyl acetate=1/3) to give the compound 10 (5.63 g, 99%) as a white foam.

UV (MeOH) λmax 273.00 nm; [α]20 D −6.36 (c 1.10, MeOH); HR-MS (ESI): m/z calcd for C25H33N4O2 [M+H]+: 421.2604; Found: 421.2599; 1H NMR (400 MHz, CDCl3) δ 8.34 (s, 1H), 7.30 (d, J=7.6 Hz, 1H), 7.22 (d, J=7.2 Hz, 2H), 7.15 (t, J=6.8 Hz, 1H), 6.88 (d, J=3.2 Hz, 1H), 6.23 (d, J=3.6 Hz, 1H), 5.83 (dd, J=7.2, 15.2 Hz, 1H), 5.28 (m, 1H), 5.06 (m, 1H), 4.47 (dd, J=5.6, 10.4 Hz, 1H), 3.78 (m, 1H), 3.70 (m, 1H), 3.24 (t, J=5.2 Hz, 1H), 2.98 (m, 1H), 2.87 (m, 1H), 2.68 (m, 1H), 2.46 (m, 1H), 2.37 (m, 2H), 1.93 (m, 2H), 1.18 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 156.2, 151.8, 147.9, 143.9, 143.9, 128.3, 126.9, 125.1, 124.5, 121.9, 97.7, 77.6, 77.2, 76.9, 75.5, 74.9, 63.4, 56.4, 53.8, 44.2, 42.2, 34.9, 33.2, 30.5, 28.6; Anal. Calcd for C25H32N4O2: C, 71.40; H, 7.67; N, 13.32. Found: C, 71.46; H, 7.60; N, 13.35.

Step 8: Preparation of sulfamic acid 2-tert-butoxy-4-[4-(indan-1-ylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-cyclopentylmethyl ester (Compound 11)

Figure US20120330013A1-20121227-C00013

Preparation of 2.0 M solution of chlorosulfonamide in acetonitrile: Formic acid (14.15 ml, 166.0 mmol) was added dropwise to chlorosulfonyl isocyanate (32.0 ml, 162.5 mmol) under nitrogen atmosphere at 0° C. When the addition was completed, the mixture was solidified. To the mixture was added acetonitrile (61.3 ml), and the resulting solution was left to stand under nitrogen source at room temperature overnight.

To a solution of the compound 10 (5.63 g, 13.83 mmol) and triethyl amine (9.7 ml, 0.74 mmol) in acetonitrile (278 ml) was added 2.0 M solution of chlorosulfonamide in acetonitrile (13.83 ml, 27.76 mmol) at 0° C., and the reaction mixture was stirred at room temperature for 45 minutes. Additional 2.0 M chlorosulfonamide solution in acetonitrile (13.83 ml, 27.76 mmol) was added and the mixture was stirred at room temperature for 15 minutes. The reaction was quenched with methanol, and the solvent was removed. The residue was purified by silica gel column chromatography (methylene chloride/methanol=20/1) to give the compound 11 (6.37 g, 92%) as a white foam.

UV (MeOH) λmax 273.00 nm; [α]20 D −18.00 (c 0.50, MeOH); HR-MS (ESI): m/z calcd for C25H34N5O4S [M+H]+: 500.2332; Found: 500.2331; 1H NMR (400 MHz, CDCl3) δ 8.38 (s, 1H), 7.36 (d, J=7.2 Hz, 1H), 7.29 (d, J=7.2 Hz, 1H), 7.22 (m, 2H), 6.95 (d, J=3.6 Hz, 1H), 6.31 (d, J=3.2 Hz, 1H), 5.89 (d, J=6.4 Hz, 1H), 5.10 (s, 2H), 4.41 (m, 2H), 4.26 (m, 1H), 3.05 (m, 1H), 2.94 (m, 1H), 2.76 (m, 2H), 2.27 (m, 3H), 2.06 (m, 1H), 1.97 (m, 1H), 1.76 (br s, 1H); 13C NMR (100 MHz, CDCl3) δ 156.4, 151.9, 149.9, 143.9, 143.8, 128.3, 126.9, 125.1, 124.5, 121.9, 121.9, 103.5, 97.9, 77.4, 77.2, 76.9, 74.3, 71.9, 71.3, 56.4, 53.1, 49.0, 42.3, 34.9, 34.3, 30.5, 28.6; Anal. Calcd for C25H33N5O4S: C, 60.10; H, 6.66; N, 14.02; S, 6.42. Found: C, 60.15; H, 6.71; N, 13.98; S, 6.39.

Step 9: Preparation of sulfamic acid 2-hydroxy-4-[4-(indan-1-ylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-cyclopentylmethyl ester (Compound 1)

Figure US20120330013A1-20121227-C00014

A solution of the compound 11 (6.37 g, 12.72 mmol) in 70% trifluoroacetic acid (149.24 ml) was stirred at room temperature for 2 hours. The solvent was removed and the residue was purified by silica gel column chromatography (hexane/ethylene acetate=1/2) to give the compound 1 (5.08 g, 90%) as a white foam.  BASE

UV (MeOH) λmax 279.50 nm; [α]20 D −6.41 (c 2.34, MeOH);

HR-MS (ESI): m/z calcd for C21H26N5O4S [M+H]+: 444.1705; Found: 444.1706;

1H NMR (400 MHz, CD3OD) δ 8.17 (d, J=1.6 Hz, 1H), 7.25 (m, 2H), 7.18 (m, 2H), 6.64 (d, J=3.6 Hz, 1H), 5.86 (t, J=7.6 Hz, 1H), 5.46 (m, 1H), 4.49 (d, J=2.8 Hz, 1H), 3.07 (m, 1H), 2.92 (m, 1H), 2.80 (m, 1H), 2.64 (m, 1H), 2.35 (m, 1H), 2.25 (m, 2H), 2.03 (m, 2H);

13C NMR (100 MHz, CD3OD) δ 152.1, 145.3, 144.6, 128.8, 127.6, 125.7, 125.2, 122.6, 100.5, 73.1, 70.9, 56.9, 54.0, 44.8, 43.6, 34.9, 34.6, 31.1;

Anal. Calcd for C21H25N5O4S: C, 56.87; H, 5.68; N, 15.79; S, 7.23. Found: C, 56.91; H, 5.73; N, 15.82; S, 7.26.

…………………….

http://www.google.com/patents/WO2010132110A1?cl=en

((lS,2S,4R)-4-{4-[(lS)-2,3-dihydro-lH-inden-l-ylamino]-7H-pyrrolo[2,3-d]pyrimidin-7-yl }-2-hydroxycyclopentyl)methyl sulfamate (//) is described in Intl. App. Pub. No. WO 07/092213, U.S. App. Pub. No. 2007/0191293, and U.S. App. Pub. No. 2009/0036678. The potassium salt of ((lS,2S,4R)-4-{4-[( 1 S)-2,3-dihydro- 1 H-inden- 1 -ylamino]-7H-pyrrolo[2,3-d]pyrimidin-7-yl } -2-hydroxycyclopentyl)methyl sulfamate is disclosed in Intl. App. Pub. No. WO 07/092213 and U.S. App. Pub. No. 2007/0191293.

(H)

((lS,2S,4R)-4-{4-[(lS)-2,3-dihydro-lH- inden-l-ylamino]-7H-pyπOlo[2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl)methyl sulfamate (/):

Figure imgf000002_0001

Step 3: Synthesis of ((lS,2S.4R)-4-(4-r(lS)-2,3-dihydro-lH-inden-l-ylaminol-7H-pyrrolor2.3-dlpyrimidin-7-yl}-2-hvdroxycvclopentyl)methyl sulfamate hydrochloride Form 1

[0158] A reactor was charged with ((lS,2S,4R)-4-{4-[(lS)-2,3-dihydro-lH-inden-l-ylarnino]-7H-pyrrolo[2,3-d]pyrimidin-7-yl }-2-hydroxycyclopentyl)methyl sulfamate (13.4 Kg, 30.2 mol) and 200-proof ethanol (106.2 Kg). The mixture was heated to reflux to afford a clear solution. The mixture was cooled to 50 ± 5 0C and passed through a cartridge filter. 200 proof ethanol (8.9 Kg) was used to rinse the filter. 1.27M hydrogen chloride in ethanol (10.2 Kg) was added via a cartridge filter at a rate to maintain a temperature of 50 ± 5 0C. The mixture was then seeded with Form 1 (67 g). Further 1.27M HCl (10.2 Kg) was added via a cartridge filter at a rate to maintain a temperature of 50 ± 5 0C. The mixture was then stirred at 50 ± 5 0C for about 3 hours. The mixture was then cooled to 20 ± 5 0C over about 3 hours and then stirred for about 2.5 hours. The solid product was then isolated by filtration and washed with 200-proof ethanol (I x 20.4 Kg and 1 x 21.2 Kg). The solids were dried by aspiration on the filter until no supernatant was seen to be collected, and then further dried under reduced pressure at <30 0C to afford the title compound (12.2 Kg) as a white solid determined to be Form 1 by XRPD. IH NMR (300MHz, DMSO, δ): 9.83 (s, IH), 8.34 (s, IH), 7.62 (s, IH), 7.44 (s, 2H), 7.30 (m, 3H), 7.22 (t, IH), 7.07 (s, IH), 5.86 (dd, IH), 5.42 (m, IH), 4.32 (m, IH), 4.21 (dd, IH), 4.02 (dd, IH), 3.04 (m, IH), 2.88 (m, IH), 2.67 (m, 2H), 2.15 (m, 2H), 2.08 (m, 2H), 1.94 (m, IH). XRPD data for Form 1 is shown in FIGURE 1 and Table 1; DSC data is shown in FIGURE 2, and TGA data for Form 1 is shown in FIGURE 3.

…………..

http://www.google.com/patents/WO2007092213A2?cl=en

Example 70: Diastereoisomeric mixture of (lS/2R/4R)-4-{4-[(lS)-2/3-dihydro-lH-inden-l- ylaimnol-ZH-pyrrolop^-dlpyxirnidin-Z-ylJ^-hydroxycyclopentyl s ulf amate and (lRf2S/4S)-4-{4-[(lS)-2,3-dihydro-lH-inden-l-ylaminol-7H-pyrrolo[2,3d]- pyrimidin-7-yl}-2-hydroxycyclopentyl sulfamate (Compounds 1-77 and 1-78)

Figure imgf000141_0001

Step a: Cyclopent-3-en-l-yl methanesulfonate

[0335] 3-Cydopentene-l-ol (0.500 g, 5.94 mmol) was stirred in DCM (95 mL).

Pyridine (2.40 mL), N,N-dimethylaminopyridine (0.10 g, 1.00 mmol) and methanesulfonyl chloride (0.690 mL, 8.92 mmol) were added, and the reaction mixture was stirred at 350C for 4 h. N,N-Dimethylarrιinopyridirιe (0.14 g, 1.2 mmol) and methanesulfonyl chloride (0.69 mL, 8.92 mmol) were added, and the reaction was stirred overnight. TLC indicated complete conversion. The reaction mixture was cooled and concentrated. The residue was purified by silica gel chromatography, eluting with DCM, to afford the title compound as a clear oil (0.660 g, 68%).

Step b: 7-Cyclopent-3-en-l-yl-N-r(lSV2,3-dihydro-lH-inden-l-yn-7H-pyrrolor2,3-rfl- pyrmτidin-4-arnine

[0336] N-[(lS)-2,3-DihydrcHlH-mden-l-yl]-7H-pyrrolo[2/3-d]p3αimidin-4-amine (1.32 g, 5.29 mmol) was azeotroped with toluene and placed under high vacuum for 30 min. N,N-Dimethylformamide (17.7 mL) was added, followed by cesium carbonate (1.99 g, 6.10 mmol). The mixture was stirred at 700C for 10 min. Cyclopent-3-en-l-yl methanesulfonate (0.660 g, 4.07 mmol) in N,N-dimethylformarnide (12.6 mL) was added dropwise. The reaction mixture was heated to 1100C for 1 h. The reaction mixture was cooled, quenched with brine and diluted with H2O. The aqueous layer was extracted with EtOAc (3x), washed with H2O and brine, dried (Na2SO4), filtered, and concentrated. The residue -was purified by via silica gel chromatography, eluting with a gradient of 0 to 5% MeOH in DCM followed by 25 to 50% EtOAc in hexanes, to afford the title compound as a pale brown solid (0.684 g, 53%). LC/MS: R1 = 1.38 min, ES+ 317 (FA standard). Step c: (lR,2S,45)-4-{4-r(lS)-2,3-dihydro-lH-inden-l-ylaininol-7H-pyrrolof2.3- rf1pyrimidin-7-yl}cyclopentane-l,2-diol

[0337] 7-Cyclopent-3-en-l-yl-N-[(lS)-2^-dihyrdo-lH-inden-l-yl]-7H-pyrrolo[2,3- d]pyτimidin-4-amine (0.312 g, 0.986 mmol) was stirred in tert-butyl alcohol (4.9 mL) and H2O (4.9 mL). AD-mix-α (Sigma- Aldrich, 1.4 g) was added, and the suspension was stirred at rt overnight. TLC indicated complete conversion. The reaction was quenched with sodium sulfite (1.48 g, 11.7 mmol), and the mixture was stirred for 5 h. The reaction mixture was diluted with EtOAc and H2O, and the aqueous layer was extracted with EtOAc (2x). The organic layer was dried (Na2SO4), filtered, and concentrated. The residue was purified via silica gel chromatography, eluting with EtOAc, to afford the title compound as a white solid (0.190 g, 55%).

Step d: Diastereoisomeric mixture of (lS,2R,4R)-4-{4-r(15)-23-dihydro-lH-inden-l- ylarninoi^jH-pyrrolofΣ^dlpyrirnidin-y-yll-l-hydroxycyclopentyl sulfamate and (lR,2S,4S)-4-{4-iαSV2,3-dihydro-lH-inden-l-ylarninol-7H-pyrrolor2,3- rf1pyrimidm-7-yl)-2-hydroxycyclopenryl sulfamate (Compounds 1-77 and 1-78)

[0338] (lR,2S,4S)-4-{4-[(lS)-2,3-Dihydro-lH-inden-l-ylarnino]-7H-pyrrolo[2/3- d]pyrimidin-7-ylJcyclopentane-l,2-diol (0.080 g, 0.23 mmol) was azeotroped with toluene and then was dissolved in anhydrous acetonitrile (2.3 mL). Pyridine (0.0369 mL, 0.458 mmol) was added. The reaction mixture was cooled to 00C, and a 2N solution of chlorosulfonamide in acetonitrile (0.144 mL) was added dropwise. The reaction was stirred for 1 h, and then additional 2N chlorosulfonamide in acetonitrile (0.028 mL) was added. After 30 min, additional 2N chlorosulfonamide in acetonitrile (0.0342 mL) was added, and the reaction mixture was stirred for 2 h. The reaction was quenched with methanol, and the mixture was concentrated in vacuo. The residue was purified by preparative thin layer chromatography using DCM:AcCN:MeOH (50:45:5). The relevant band was cut, washed with acetone, filtered, and concentrated to give a mixture of diastereomers as a white solid. (11 mg, 11%). 1H NMR (CDCl3, 400 NMR, δ): 8.36-8.27 (m, IH); 7.38-7.09 (m, 5H); 6.90-6.80 (m, IH); 6.36- 6.20 (m, IH); 5.95-5.76 (m, IH); 5.51-5.22 (m, 2H); 4.83-4.68 (m, IH); 3.87-3.72 (m, IH); 3.12- 2.83 (m, 2H); 2.75-2.53 (m, IH); 2.50-2.14 (m, 2H); 2.08-1.79 (m, 2H) ppm. LC/MS: R, = 1.16 min, ES+ 430 (FA standard).

…………

WO 2012061551

http://www.google.im/patents/WO2012061551A1?cl=en

The compound ((lS,2S,4R)-4-(4-((lS)-2,3-dihydro-lH-inden-l-ylamino)-7H-pyrrolo[2,3-d]- pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulfamate:

Figure imgf000002_0001

also known as MLN4924, is an inhibitor of NEDD8-activating enzyme (NAE). Inhibition of NAE has been shown to induce cancer cell death and inhibit the growth of tumors in xenograft models. See, e.g., T.A. Soucy et al., Nature, 2009, 458, 732-737; T.A. Soucy ei al., Clin. Cancer Res., 2009, 15 (12), 3912-3916; and J.E. Brownell et al., Mol. Cell., 2010, 37 (1), 102-111, each of which is hereby incorporated by reference herein in its entirety. MLN4924, pharmaceutical compositions of MLN4924, processes for its synthesis, and polymorphic forms have been described previously. See, e.g., US Patent Appl. Nos. 11/700,614 (Publ. No. 2007/0191293), 12/221,399 (Publ. No. 2009/0036678) and 12/779,331 (Publ. No. 2011/0021544),

……………

Org. Process Res. Dev., Article ASAP
Abstract Image

A practical synthesis of a novel NEDD8-activating enzyme (NAE) inhibitor pevonedistat (MLN4924) is described. Key steps include an enantioselective synthesis of an amino-diol cyclopentane intermediate containing three chiral centers and a novel, regioselective sulfamoylation using N-(tert-butoxycarbonyl)-N-[(triethylenediammonium)sulfonyl]azanide. The linear process, involving six solid isolations, has been carried out in multiple cGMP productions on 15–30 kg scale to produce pevonedistat in 98% (a/a) chemical purity and 25% overall yield.

Figure

Figure

((1S,2S,4R)-4-(4-(((S)-2,3-Dihydro-1H-inden-1-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl Sulfamate (1)

 The reaction yielded 1 (0.285 kg, 58.5%, 93.0% a/a) as an off-white solid.
HPLC retention time of 1   BASE(Method C): 22.6 min;
1H NMR (400 MHz, DMSO) δ 8.19 (s, 1H), 7.77 (d, J = 8.4 Hz, 1H), 7.45 (s, 2H), 7.31–7.26 (m, 2H), 7.22 (t, J = 6.6 Hz, 2H), 7.15 (t, J = 7.2 Hz, 1H), 6.66 (d, J = 3.5 Hz, 1H), 5.92 (q, J = 8.0 Hz, 1H), 5.39 (qd, J = 8.8, 5.7 Hz, 1H), 4.95 (d, J = 3.9 Hz, 1H), 4.42–4.31 (m, 1H), 4.25 (dd, J = 9.7, 7.0 Hz, 1H), 4.07 (dd, J = 9.6, 8.0 Hz, 1H), 3.01 (ddd, J = 15.7, 8.7, 3.0 Hz, 1H), 2.95–2.81 (m, 1H), 2.81–2.65 (m, 1H), 2.58–2.49 (m, 1H), 2.31–1.86 (m, 5H);
13C NMR (100 MHz, DMSO) δ 155.91, 151.18, 149.02, 144.66, 142.98, 127.30, 126.28, 124.49, 124.11, 121.68, 102.83, 98.86, 70.82, 69.37, 54.48, 52.15, 42.58, 42.25, 33.50, 33.26, 29.72;
m/z: 444.4 (M + H)+;
mp: 164–166 °C.

((1S,2S,4R)-4-(4-(((S)-2,3-Dihydro-1H-inden-1-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl Sulfamate·Hydrochloride (Pevonedistat)

Pevonedistat (14.0 g, 92.5%, 99.0% a/a) as a white solid.
HPLC retention time of pevonedistat (Method C): 22.6 min;
1H NMR (400 MHz, DMSO) δ 9.70 (s, 1H), 8.39 (s, 1H), 7.63 (s, 1H), 7.45 (s, 2H), 7.41–7.20 (m, 4H), 7.04 (s, 1H), 5.78 (s, 1H), 5.44 (s, 1H), 4.42–4.28 (m, 1H), 4.24 (dd, J = 9.7, 6.9 Hz, 1H), 4.05 (dd, J = 9.6, 8.0 Hz, 1H), 3.18–2.99 (m, 1H), 2.91 (dt, J = 15.6, 7.7 Hz, 1H), 2.81–2.57 (m, 2H), 2.24–1.86 (m, 6H).
13C NMR (100 MHz, DMSO) δ 149.12, 145.71, 143.23, 142.11, 141.30, 128.28, 126.64, 124.97, 124.82, 124.49, 102.57, 101.74, 70.67, 69.22, 57.38, 53.14, 42.52, 42.40, 33.57, 32.56, 29.80;
m/z: 444.4 (M + H)+;
mp: 155–157 °C.
Figure
……………..
J. Org. Chem., 2011, 76 (9), pp 3557–3561
DOI: 10.1021/jo2001897
Abstract Image

MLN4924 (1), which is in clinical trials as an anticancer agent, was stereoselectively synthesized from d-ribose via a route involving stereoselective reduction, regioselective cleavage of an isopropylidene moiety, and selective displacement of a cyclic sulfate moiety as key steps.

Sulfamic Acid 2-Hydroxy-4-[4-(indan-1-ylamino)pyrrolo[2,3-d]pyrimidin-7-yl]cyclopentylmethyl Ester (1)  BASE

purified by silica gel column chromatography (hexane/ethyl acetate = 1/2) to give 1 (5.08 g, 90%) as a white foam:
UV (MeOH) λmax 279.50 nm;
[α]20D −6.41 (c 2.34, MeOH);
HR-MS (ESI) m/z calcd for C21H26N5O4S [M + H]+ 444.1705, found 444.1706;
1H NMR (400 MHz, CD3OD) δ 8.17 (d, J = 1.6 Hz, 1H), 7.25 (m, 2H), 7.18 (m, 2H), 6.64 (d, J = 3.6 Hz, 1H), 5.86 (t, J = 7.6 Hz, 1H), 5.46 (m, 1H), 4.49 (d, J = 2.8 Hz, 1H), 3.07 (m, 1H), 2.92 (m, 1H), 2.80 (m, 1H), 2.64 (m, 1H), 2.35 (m, 1H), 2.25 (m, 2H), 2.03 (m, 2H);
13C NMR (100 MHz, CD3OD) δ 152.1, 145.3, 144.6, 128.8, 127.6, 125.7, 125.2, 122.6, 100.5, 73.1, 70.9, 56.9, 54.0, 44.8, 43.6, 34.9, 34.6, 31.1. Anal. Calcd for C21H25N5O4S: C, 56.87; H, 5.68; N, 15.79; S, 7.23. Found: C, 56.91; H, 5.73; N, 15.82; S, 7.26.
MLN1 MLN2 MLN3
NMR FROM CHEMIETEK
NMR
WO2012061551A1 * Nov 3, 2011 May 10, 2012 Millennium Pharmaceuticals, Inc. Administration of nedd8-activating enzyme inhibitor
WO2013028832A2 * Aug 23, 2012 Feb 28, 2013 Millennium Pharmaceuticals, Inc. Inhibitors of nedd8-activating enzyme
WO2013028832A3 * Aug 23, 2012 May 2, 2013 Millennium Pharmaceuticals, Inc. Inhibitors of nedd8-activating enzyme
US8809356 Aug 23, 2012 Aug 19, 2014 Millennium Pharmaceuticals, Inc. Inhibitors of NEDD8-activating enzyme

1H NMR PREDICT

1H NMR G 1HNMR

13 C NMR

13CNMR G 13CNMR

//////////Pevonedistat, MLN4924, Millennium Pharmaceuticals, TAKEDA, TAK-924 , PHASE 1, orphan drug designation

Tazemetostat


Tazemetostat.svg

Tazemetostat

Current developer:  Epizyme, Inc., Cambridge, MA 02139.

EPZ-6438 (Tazemetostat)
CAS: 1403254-99-8

HBR 1467052-75-0

タゼメトスタット臭化水素酸塩

Current developer:  Epizyme, Inc., Cambridge, MA 02139.

EPZ-6438 (Tazemetostat)
CAS: 1403254-99-8

HBR

Chemical Formula: C34H44N4O4
Exact Mass: 572.33626

USFDA APPROVED 23/1/2020 AS HBR SALT, TAZVERIK, EPIZYME

N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[1,1′-biphenyl]-3-carboxamide
SIMLES: O=C(C1=CC(C2=CC=C(CN3CCOCC3)C=C2)=CC(N(CC)C4CCOCC4)=C1C)NCC5=C(C)C=C(C)NC5=O

(1,1′-Biphenyl)-3-carboxamide, N-((1,2-dihydro-4,6-dimethyl-2-oxo-3-pyridinyl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(4-morpholinylmethyl)-

N-((4,6-Dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(oxan-4-yl)amino)-4-methyl-4′-((morpholin-4-yl)methyl)(1,1′-biphenyl)-3-carboxamide

UNII-Q40W93WPE1

Tazemetostat, sold under the brand name Tazverik, is a medication used for the treatment of adults and adolescents aged 16 years and older with metastatic (when cancer cells spread to other parts of the body) or locally advanced (when cancer has grown outside the organ it started in, but has not yet spread to distant parts of the body) epithelioid sarcoma not eligible for complete resection (surgically removing all of a tissue, structure, or organ).[1]

Tazemetostat is a cancer drug that acts as a potent selective EZH2 inhibitor.[2]

Tazemetostat blocks activity of the EZH2 methyltransferase, which may help keep the cancer cells from growing.[1] Most cases of epithelioid sarcoma begin in the soft tissue under the skin of an extremity, though it can start in other areas of the body.[1] Surgical removal is considered the main treatment when the cancer is localized to one area of the body.[1] Chemotherapy or radiation may also be given.[1] However, there is a high likelihood for local and regional spread of the disease even with treatment and approximately 50% of patients have metastatic disease at the time of diagnosis.[1] Metastatic disease is considered life-threatening to the patient.[1]

The most common side effects are pain, fatigue, nausea, decreased appetite, vomiting and constipation.[1] People taking tazemetostat are at increased risk of developing secondary malignancies including: T-cell lymphoblastic lymphoma (a type of blood cancer that affects the lymphatic system usually found in the lymph nodes), myelodysplastic syndrome (a disorder resulting from poorly formed or dysfunctional blood cells) and acute myeloid leukemia (a cancer of the blood and bone marrow).[1]

According to the NCI Drug Dictionary, “tazemetostat is an orally available, small molecule selective and S-adenosyl methionine (SAM) competitive inhibitor of histone methyl transferase EZH2, with potential antineoplastic activity. Upon oral administration, tazemetostat selectively inhibits the activity of both wild-type and mutated forms of EZH2. Inhibition of EZH2 specifically prevents the methylation of histone H3 lysine 27 (H3K27). This decrease in histone methylation alters gene expression patterns associated with cancer pathways and results in decreased tumor cell proliferation in EZH2 mutated cancer cells. EZH2, which belongs to the class of histone methyltransferases (HMTs), is overexpressed or mutated in a variety of cancer cells and plays a key role in tumor cell proliferation.”[3]

History

The U.S. Food and Drug Administration (FDA) approved tazemetostat in January 2020,[1] based on the results of a clinical trial (NCT02601950) enrolling 62 subjects with metastatic or locally advanced epithelioid sarcoma.[1][4] During the clinical trial, subjects received 800 milligrams (mg) of tazemetostat twice a day until the disease progressed or the subject reached an unacceptable level of toxicity.[1][4] Tumor response assessments were performed every eight weeks during the clinical trial.[1] The trial measured how many subjects experienced complete or partial shrinkage (by a certain amount) of their tumors during treatment (overall response rate).[1] The overall response rate was 15%, with 1.6% of subjects having a complete response and 13% having a partial response.[1] Of the nine subjects that had a response, six (67%) subjects had a response lasting six months or longer.[1]

The trial was conducted at 22 sites in France, United Kingdom, Taiwan, Italy, Canada, Belgium, and the United States.[4]

The FDA granted the application for tazemetostat accelerated approval and orphan drug designation.[1] The FDA granted the approval of Tazverik to Epizyme Inc.[1]

PATENT

PRODUCT PAT

US 8410088 EXP 21/1/2034

WO 2012142504

US 9090562 EXP 13/4/32

SEE  Proceedings of the National Academy of Sciences of the United States of America (2013), 110(19), 7922-7927, S7922/1-S7922/5….http://www.pnas.org/content/110/19/7922.abstract

http://www.epizyme.com/wp-content/uploads/2014/11/Ribrag-ENA-FINAL.pdf

2D chemical structure of 1403254-99-8

Tazemetostat, also known as EPZ-6438,  is a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity. EPZ-6438 induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells.

Treatment of xenograft-bearing mice with EPZ-6438 leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation.

These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers. EPZ-6438 is currently in clinical trials.

Epizyme, Inc., Eisai R&D Management Co.Ltd.

Epizyme is developing tazemetostat, a lead from several small molecule EZH2 inhibitors, for treating cancer (phase 1 clinical, as of April 2015). Japanese licensee Eisai was developing the program for the potential oral treatment of cancers, including non-Hodgkin’s lymphoma; however, in March 2015, Epizyme regained worldwide, ex-Japan, rights to the program.

It appeared that Eisai was planning to investigate the program in Japan .

WO-2015057859 From, Eisai Research Institute; Epizyme Inc, indicates Novel crystalline polymorphic form C of tazemetostat, useful for treating an EZH2-mediated cancer, including non-Hodgkin’s lymphoma and breast cancer.

see WO2013155317, claiming novel hydrobromide salt of tazemetostat.

PREDICT

TAZ 1H NMR

TAZ 13

TAZ 13 2

………………………………….

PATENT

WO 2012142504

http://www.google.com/patents/WO2012142504A1?cl=en

Example 44: Synthesis of N-((4,6-dimethyl-2-oxo-l ,2-dihydropyridin-3- yl)methyl)-5-(ethyl (tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(moφholinomethyl)-[l , – biphenyl]-3-carboxamide

Compound 44

[Step 1 : Synthesis of 5-brom -2-methyl-3-nitrobenzoic acid

To stirred solution of 2-methyl-3-nitrobenzoic acid ( 100 g, 552 mmol) in cone. H2S04 (400 mL), 1 ,3-dibromo-5,5-dimethyl-2,4-imidazolidinedione (88 g, 308 mmol) was added in a portion wise manner at room temperature and the reaction mixture was then stirred at room temperature for 5 h. The reaction mixture was poured onto ice cold water, the precipitated solid was filtered off, washed with water and dried under vacuum to afford the desired compound as a solid ( 140 g, 98%). The isolated compound was taken directly into the next step. Ή NMR (DMSO-4$, 400 MHz) δ 8.31 (s, 1 H), 8.17 (s, 1 H), 2.43 (s, 3H).

Step 2: Synthesis of methyl -bromo-2-methyl-3-nitrobenzoate

To a stirred solution of 5-bromo-2-methyl-3-nitrobenzoic acid (285 g, 1 105 mmol) in DMF (2.8L) at room temperature was added sodium carbonate (468 g, 4415 mmol) followed by addition of methyl iodide (626.6 g, 4415 mmol). The resulting reaction mixture was heated at 60 °C for 8 h. After completion (monitored by TLC), the reaction mixture was filtered (to remove sodium carbonate) and washed with ethyl acetate ( 1 L X 3). The combined filtrate was washed with water (3L X 5) and the aqueous phase was back extracted with ethyl acetate (1L X 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound as a solid (290g, 97% yield). The isolated compound was taken directly into the next step. Ή NMR (CDC13, 400 MHz) δ 8.17 (s, 1H), 7.91 (s, 1H), 3.96 (s, 3H), 2.59 (s, 3H).

Step 3: Synthesis of methyl 3-amino-5-bromo-2-methylbenzoate

To a stirred solution of methyl 5-bromo-2-methyl-3-nitrobenzoate (290 g,

1058 mmol) in ethanol (1 .5L) was added aqueous ammonium chloride (283 g, 5290 mmol dissolved in 1.5L water). The resulting mixture was stirred at 80°C to which iron powder (472 g, 8451 mmol) was added in a portion wise manner. The resulting reaction mixture was heated at 80 °C for 12 h. Upon completion as determined by TLC, the reaction mixture was hot filtered over celite® and the celite bed was washed with methanol (5L) followed by washing with 30% MeOH in DCM (5L). The combined filtrate was concentrated in-vacuo, the residue obtained was diluted with aqueous sodium bicarbonate solution (2L) and extracted with ethyl acetate (5L X 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound as a solid (220 g, 85%). The compound was taken directly into the next step. Ή NMR (CDC13, 400 MHz) δ 7.37 (s, 1 H), 6.92 (s, 1 H), 3.94 (s, 3H), 3.80 (bs, 2H), 2.31 (s, 3H).

Step 4: Synthesis of methyl 5-bromo-2-methyl-3-((tetrahydro-2H-pyran-4-yl) amino) benzoate

To a stirred solution of methyl 3-amino-5-bromo-2-methylbenzoate (15 g, 61 .5 mmol) and dihydro-2H-pyran-4(3)-one (9.2 g, 92 mmol) in dichloroethane (300 mL) was added acetic acid (22 g, 369 mmol) and the reaction mixture stirred at room temperature for 15 minutes, then the reaction mixture was cooled to 0°C and sodium triacetoxyborohydnde (39 g, 184 mmol) was added. The reaction mixture was stirred overnight at room temperature. Upon completion of the reaction as determined by TLC, aqueous sodium bicarbonate solution was added to the reaction mixture until a pH of 7-8 was obtained. The organic phase was separated and the aqueous phase was extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude compound was purified by column chromatography (100-200 mesh silica gel) eluting with ethyl acetate: hexane to afford the desired compound as a solid ( 14 g, 69%). ‘H NMR (DMSO-<fc, 400 MHz) δ 7.01 (s, 1 H), 6.98 (s, 1 H), 5.00 (d, 1 H, J=7.6 Hz), 3.84-3.87 (m, 2H), 3.79 (s, 31 1), 3.54-3.56 (mf 1 H), 3.43 (L 21 1, J 12 Hz), 2.14 (s. 31 1). 1 . 1 – 1 .84 (m: 211). 1 .47- 1 .55 (m, 2H).

Step 5: Synthesis of methyl 5-bromo-3-(ethyl (tetrahydro-2H-pyran-4-yl) amino)-2- methylbenzoate

To a stirred solution of methyl 5-bromo-2-methyl-3-((tetrahydro-2H-pyran-4-yl) amino) benzoate (14 g, 42.7 mmol) in dichloroethane (150 mL) was added acetaldehyde (3.75 g, 85.2 mmol) and acetic acid ( 15.3 g, 256 mmol). The resulting reaction mixture was stirred at room temperature for 15 minutes. The mixture was cooled to 0 °C and sodium

triacetoxyborohydnde (27 g, 128 mmol) was added. The reaction mixture was stirred at room temperature for 3 hours. Upon completion of the reaction as determined by TLC, aqueous sodium bicarbonate solution was added to the reaction mixture until a pH 7-8 was obtained, the organic phase was separated and the aqueous phase was extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude compound was purified by column chromatography (100- 200 mesh silica gel) eluting with ethyl acetate: hexane to afford the desired compound as a viscous liquid (14 g, 93%). Ή NMR (DMSO-cfo 400 MHz) δ 7.62 (s, 1 H), 7.52 (s, 1 H), 3.80 (bs, 5H), 3.31 (t, 2H), 2.97-3.05 (m, 2H), 2.87-2.96 (m, 1 H), 2.38 (s, 3H), 1.52-1.61 (m, 2H), 1 .37-1.50 (m, 2H), 0.87 (t, 3H, J=6.8 Hz).

Step 6: Synthesis of 5-bromo-N-((4, 6-dimethyl-2-oxo-l , 2-dihydropyridin-3-yl) methyl)-3 -(ethyl (tetrahydro-2H-pyra -4-yl) amino)-2-methylbenzamide

To a stirred solution of 5-bromo-3-(ethyl (tetrahydro-2H-pyran-4-yl) amino)-2- methylbenzoate (14 g, 39.4 mmol) in ethanol ( 100 mL) was added aqueous NaOH (2.36 g, 59.2 mmol in 25mL water) and the resulting mixture was stirred at 60 °C for 1 h. Upon completion of the reaction as determined by TLC, the solvent was removed under reduced pressure and the residue obtained was acidified with IN HC1 until a pH 7 was obtained and then aqueous citric acid solution was added until a pH 5-6 was obtained. The aqueous layer was extracted with 10% MeOH in DCM (200mL X 3), the combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the respective acid (14 g, 100%).

The above acid (14 g, 40.9 mmol) was then dissolved in DMSO (70 mL) and 3- (amino methyl)-4, 6-dimethylpyridin-2( l H)-one ( 12.4 g, 81 .9 mmol) was added to it. The reaction mixture was stirred at room temperature for 15 minutes, then PYBOP (31.9 g, 61.4 mmol) was added and stirring was continued for overnight at room temperature. Upon completion of the reaction as determined by TLC, the reaction mixture was poured onto ice- cold water (700 mL), stirred for 30 minutes and the precipitated solid was collected by filtration, washed with water (500 mL) and air dried. The solid obtained was stirred with acetonitrile (75mL X 2), filtered and air dried. The solid obtained was again stirred with 5% MeOH in DCM ( l OOmL), filtered and dried completely under vacuum to afford the title compound as a solid ( 14 g, 74 %). Ή NMR (DMSO- 6, 400 MHz) δ 1 1.47 (s, 1 H), 8.23 (t, 1 H), 7.30 (s, 1 H), 7.08 (s, 1 H), 5.85 (s, 1 H), 4.23 (d, 2H, J=4.4 Hz), 3.81 (d, 2H, J=l 0.4 Hz), 3.20-3.26 (m, 2H), 3.00-3.07 (m, I H), 2.91 -2.96 (m, 2H), 2.18 (s, 3H), 2.14 (s, 3H), 2.10 (s, 3H), 1.58-1.60 (m, 2H), 1.45-1.50 (m, 2H), 0.78 (t, 3H, J=6.8 Hz).

Step 7: Synthesis of N-((4, 6-dimethyl-2-oxo-l , 2-dihydropyridin-3-yl) methyl)-5- (ethyl (tetrahydro-2H-pyran-4-yl) amino)-4-methyl-4′-(morpholinomethyl)-[l , l ‘-biphenyl]-3- carboxamide

Figure imgf000226_0001 TITLE COMPD

To a stirred solution of 5-bromo-N-((4, 6-dimethyl-2-oxo-l , 2-dihydropyridin-3-yl) methyl)-3-(ethyl (tetrahydro-2H-pyran-4-yl) amino)-2-methylbenzamide (14 g, 29.5 mmol) in dioxane/ water mixture (70 mL/ 14 mL) was added 4-(4-(4, 4, 5, 5-tetramethyl- l , 3, 2- dioxaborolan-2-yl) benzyl) morpholine (13.4 g, 44.2 mmol) followed by addition of Na2C03 (1 1 .2 g, 106.1 mmol). The solution was purged with argon for 15 minutes and then Pd (PPh3)4 (3.40 g, 2.94 mmol) was added and the solution was again purged with argon for a further 10 min. The reaction mixture was heated at 100°C for 4 h. After completion (monitored by TLC), the reaction mixture was diluted with water and extracted with 10% MeOH/DCM.

The combined organic layers were dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure. The crude compound was purified by column chromatography (100- 200 mesh silica gel) eluting with methanol: DCM to the title compound as a solid (12 g, 71 %).

Analytical Data: LCMS: 573.35 (M + 1 )+; HPLC: 99.5% (@ 254 nm) (R,;3.999; Method: Column: YMC ODS-A 1 50 mm x 4.6 mm x 5 μ; Mobile Phase: A; 0.05% TFA in water/ B; 0.05% TFA in acetonitrile; Inj. Vol : 10 μΐ, Col. Temp.: 30 °C; Flow rate: 1 .4 mL/min.;

Gradient: 5% B to 95% B in 8 min, Hold for 1 .5 min, 9.51 -12 min 5% B);

Ή NMR (DMSO-i 6, 400 MHz) 5 1 1 .46 (s, I H), 8. 19 (t, 1 H), 7.57 (d, 2H, J=7.2 Hz), 7.36-7.39 (m, 3H), 7.21 (s, I H), 5.85 (s, I H), 4.28 (d, 2H, J=2.8 Hz), 3.82 (d, 2H, J=9.6 Hz), 3.57 (bs, 4H), 3.48 (s, 2H), 3.24 (t, 2H, J=10.8Hz), 3.07-3.09 (m, 2H), 3.01 (m, I H), 2.36 (m, 4H), 2.24 (s, 3H), 2.20 (s, 3H), 2.10 (s, 3H), 1 .64-1 .67 (m, 2H), 1 .51 – 1 .53 (m, 2H), 0.83 (t, 3H, J=6.4 Hz).

TRIHYDROCHLORIDE

Step 8: Synthesis of N-((4,6-dimethyl-2-oxo-l ,2-dihydropyridin-3-yl)methyl)-5- (ethyl (tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[ 1 , 1 ‘-biphenyl]-3- carboxamide trihydrochloride

N-((4, 6-dimethyl-2-oxo-l , 2-dihydropyridin-3-yl) methyl)-5-(ethyl (tetrahydro- 21 l-pyran-4-yl) amino)-4-methyI-4′-(niorpholinornethyl)-[ 1 , 1 ‘-biphenyl]-3-carboxamide ( 12 g, 21.0 mmol) was dissolved in methanolic HC1 (200 mL) and stirred at room temperature for 3 h. After three hours of stirring, the reaction mixture was concentrated under reduced pressure. The solid obtained was stirred with ether ( l OOmL X 2) to afford the desired salt as a solid ( 1 1 g, 77 %).

Analytical Data of the tri-HCl salt: LCMS: 573.40 (M + 1 )+; HPLC: 99.1 % (@ 254 nm) (R,;3.961 ; Method: Column: YMC ODS-A 150 mm x 4.6 mm x 5 μ; Mobile Phase: A; 0.05% TFA in water/ B; 0.05% TFA in acetonitrile; Inj. Vol: 10 pL, Col. Temp.: 30 °C; Flow rate: 1.4 mL/min.; Gradient: 5% B to 95% B in 8 min, Hold for 1.5 min, 9.51 -12 min 5% B);

Ή NMR (D20 400 MHz) δ 7.92 (bs, I H,) 7.80 (s, I H), 7.77 (d, 2H, J=8 Hz), 7.63 (s, I H), 7.61 (s, I H), 6.30 (s, I H), 4.48 (s, 2H), 4.42 (s, 2H), 4.09-4.1 1 (m, 4H), 3.95-3.97 (m, 2H), 3.77 (t, 3H, J=10.4 Hz), 3.44-3.47 (m, 3H), 3.24-3.32 (m, 3H), 2.42 (s, 3H), 2.35 (s, 3H), 2.26 (s, 3H), 2.01 (m, 2H), 1 .76 (m, 2H), 1 .04 (t, 3H, J=6.8 Hz).

…………………………………………

PATENT

WO2013155317

 http://www.google.com/patents/WO2013155317A1?cl=en

N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)-5-(ethyl (tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[l,l’- biphenyl] -3-carboxamide hydrobromide:

N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-5-(ethyl

(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[l,l’-biphenyl]-3- carboxamide hydrobromide:

As used herein, “Compound I” refers to N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)-5-(ethyl (tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[l,l’- biphenyl]-3-carboxamide. The hydrobromide of Compound I can be used to inhibit the histone methyltransferase activity of EZH2, either in a subject or in vitro. The hydrobromide of Compound I can also be used to treat cancer in a subject in need thereof.

Scheme 1

……………………………………..Compound I                                       Compound I – HBr

HPLC

HPLC was conducted on an Agilent 1200 HPLC quaternary pump, low pressure mixing, with an in-line degasser. Analytical method conditions: 8 μΐ^ sample (20 mg of ER-581982-06 diluted with 50 mL of a methanol to provide approximately 0.4 mg/mL solution) was injected onto a Agilent Zorbax Eclipse XDB-C18 (4.6 x 150 mm, 3.5 um), Chromatography conditions: mobile phase A, water with 5mM ammonium formate; mobile phase B, 5 mM ammonium formate in 50/45/5 acetonitrile/methanol/water; flow rate, 1.5 ml/min.; gradient: isocratic at 10% B from 0 to 3 min; linear increase to 70% B from 3 to 7 min; isocratic at 70% B from 7 to 12 min; linear increase to 100% B from 12 to 15 min isocratic at 100% B from 15 to 20 min;

column temperature, 35 °C; detection, UV 230 nm. Approximate retention time of Compound I = 10.7 min.

Synthesis of Polymorph A

5-bromo-2-methyl-3-nitrobenzoic acid stirred solution of 2-methyl-3-nitrobenzoic acid (100 g, 552 mmol) in cone. H2S04 (400 mL), l,3-dibromo-5,5-dimethyl-2,4- imidazolidinedione (88 g, 308 mmol) was added in a portion wise manner at room temperature and the reaction mixture was then stirred at room temperature for 5 h. The reaction mixture was poured onto ice cold water, the precipitated solid was filtered off, washed with water and dried under vacuum to afford the desired compound as a solid (140 g, 98%). The isolated compound was taken directly into the next step. 1H NMR (DMSO-J6, 400 MHz) δ 8.31 (s, 1H), 8.17 (s, 1H), 2.43 (s, 3H).

Methyl 5-bromo-2-methyl-3-nitrobenzoate To a stirred solution of 5-bromo-2- methyl-3-nitrobenzoic acid (285 g, 1105 mmol) in DMF (2.8L) at room temperature was added sodium carbonate (468 g, 4415 mmol) followed by addition of methyl iodide (626.6 g, 4415 mmol). The resulting reaction mixture was heated at 60 °C for 8 h. After completion (monitored by TLC), the reaction mixture was filtered (to remove sodium carbonate) and washed with ethyl acetate (1L X 3). The combined filtrate was washed with water (3L X 5) and the aqueous phase was back extracted with ethyl acetate (1L X 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound as a solid (290g, 97% yield). The isolated compound was taken directly into the next step. 1H NMR (CDC13, 400 MHz) δ 8.17 (s, 1H), 7.91 (s, 1H), 3.96 (s, 3H), 2.59 (s, 3H).

Methyl 3-amino-5-bromo-2-methylbenzoate (1) To a stirred solution of methyl 5- bromo-2-methyl-3-nitrobenzoate (290 g, 1058 mmol) in ethanol (1.5L) was added aqueous ammonium chloride (283 g, 5290 mmol dissolved in 1.5L water). The resulting mixture was stirred at 80°C to which iron powder (472 g, 8451 mmol) was added in a portion wise manner. The resulting reaction mixture was heated at 80 °C for 12 h. Upon completion as determined by TLC, the reaction mixture was hot filtered over celite® and the celite bed was washed with methanol (5L) followed by washing with 30% MeOH in DCM (5L). The combined filtrate was concentrated in- vacuo, the residue obtained was diluted with aqueous sodium bicarbonate solution (2L) and extracted with ethyl acetate (5L X 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound as a solid (220 g, 85%). The compound was taken directly into the next step. 1H

NMR (CDCI3, 400 MHz) δ 7.37 (s, 1H), 6.92 (s, 1H), 3.94 (s, 3H), 3.80 (bs, 2H), 2.31 (s, 3H).

Methyl 5-bromo-2-methyl-3-((tetrahydro-2H-pyran-4-yl) amino) benzoate (2) A reactor was charged with methyl 3-amino-5-bromo-2-methylbenzoate (455.8 g, 1.87 mol), 1,2- Dichloroethane (4.56 L), and acetic acid (535 ml, 9.34 mol). To the mixture were added dihydro-2H-pyran-4(3H)-one (280 g, 2.80 mol) and sodium triacetoxyborohydride (594 g, 2.80 mol) maintaining the internal temperature below 40 °C. The mixture was stirred at 25 °C for 2.5 h and then the reaction was quenched with a solution of sodium hydroxide (448 g, 11.20 mol) in water (5.61 L). After stirring for 20 minutes at ambient temperature, the organic layer was separated and the aqueous layer was extracted with ethyl acetate (3.65 L). The organic layers were combined, washed with brine (1.5 L), and concentrated under vacuum.

The residue was treated with ethyl acetate (1.8 L) and heated to 65-70 °C. The mixture was stirred at 65-70 °C for 15 minutes to give a clear solution and then treated with n-heptane (7.3 L) maintaining the temperature between 60-70 °C. Once the heptane was completely added to the solution, the mixture was held at 65-70 °C for 15 minutes and then allowed to cool to 18- 22 °C over 3 h. The resulting suspension was stirred at 18-22 °C for 4 h, cooled to 0-5 °C over 1 h, and held at 0-5 °C for 2 h. The precipitate was filtered, washed twice with n-heptane (1.4 L), and dried under vacuum to give the title compound (540 g, 88%). The XRPD pattern of this compound is shown in Figure 17.

Methyl 5-bromo-3-(ethyl (tetrahydro-2H-pyran-4-yl) amino)-2-methylbenzoate (3)

To a stirred solution of methyl 5-bromo-2-methyl-3-((tetrahydro-2H-pyran-4-yl) amino) benzoate (14 g, 42.7 mmol) in dichloroethane (150 mL) was added acetaldehyde (3.75 g, 85.2 mmol) and acetic acid (15.3 g, 256 mmol). The resulting reaction mixture was stirred at room temperature for 15 minutes. The mixture was cooled to 0 °C and sodium triacetoxyborohydride (27 g, 128 mmol) was added. The reaction mixture was stirred at room temperature for 3 hours. Upon completion of the reaction as determined by TLC, aqueous sodium bicarbonate solution was added to the reaction mixture until a pH 7-8 was obtained, the organic phase was separated and the aqueous phase was extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude compound was purified by column chromatography (100-200 mesh silica gel) eluting with ethyl acetate: hexane to afford the desired compound as a viscous liquid (14 g, 93%). 1H NMR DMSO-d6, 400 MHz) δ 7.62 (s, 1H), 7.52 (s, 1H), 3.80 (bs, 5H), 3.31 (t, 2H), 2.97-3.05 (m, 2H), 2.87-2.96 (m, 1H), 2.38 (s, 3H), 1.52-1.61 (m, 2H), 1.37-1.50 (m, 2H), 0.87 (t, 3H, J=6.8 Hz).

Methyl 5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-

[l,l’-biphenyl]-3-carboxylate (4): A mixture of methyl 5-bromo-3-(ethyl(tetrahydro-2H-pyran- 4-yl)amino)-2-methylbenzoate (580 g, 1.63 mol), 4-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)benzyl)morpholine (592 g, 1.95 mol), 1,4-dioxane (3.86 L), sodium carbonate (618 g, 5.83 mol), and water (771 ml) was degassed by bubbling nitrogen through the mixture at 20 °C for 20 minutes and treated with tetrakis(triphenylphosphine)palladium(0) (14.11 g, 12.21 mmol). The resulting mixture was degassed for an additional 20 minutes and then heated to 87-89 °C for 17 h. After cooling to 20 °C, the mixture was diluted with ethyl acetate (5.80 L) and a solution of (R)-2-Amino-3-mercaptopropionic acid (232 g) in water (2.320 L). After stirring for 1 h at 20 °C, the organic layer was separated and washed again with a solution of (R)-2-Amino-3- mercaptopropionic acid (232 g) in water (2.320 L). The aqueous layers were combined and extracted with ethyl acetate (5.80 L). The organic layers were combined, washed with a solution of sodium hydroxide (93 g) in water (2.32 L), and concentrated under vacuum at 35 °C to give the title compound as an orange oil (1.21 kg, 164% yield).

5-(Ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[l,l’- biphenyl]-3-carboxylic acid (5): Methyl 5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′- (morpholinomethyl)-[l,l’-biphenyl]-3-carboxylate (69.0 g, 152.5 mmol) (based on the theoretical yield from the previous step) was suspended in ethanol (380 mL) and treated with a solution of sodium hydroxide (24.84 g, 621.0 mmol) in water (207 mL). The mixture was stirred at 40°C for 18 h. After cooling to 0-5 °C, the mixture was neutralized to pH 6.5 with 1 N hydrochloric acid (580 mL) maintaining the temperature below 25 °C. Then, the mixture was extracted twice with a mixture of dichloromethane (690 mL) and methanol (69.0 mL). The organic layers were combined and concentrated under vacuum to give a crude product as a yellow solid (127g).

The crude product was dissolved in 2-methyltetrahydrofuran (656 mL) at 70 °C and then treated with IPA (828 mL). The mixture was allowed to cool to rt over 3-4 h and then stirred overnight at rt. The precipitate was filtered, washed twice with IPA (207 mL), and dried under vacuum to give the title compound as an off white solid (53.54 g, 80%). The XRPD pattern of this compound is shown in Figure 9.

N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H- pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[l,l’-biphenyl]-3-carboxamide

(Compound I): A mixture of 5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′- (morpholinomethyl)-[l,l’-biphenyl]-3-carboxylic acid (540 g, 1.23 mol) and 3-(aminomethyl)- 4,6-dimethyl-dihydro-pyridin-2(lH)-one hydrochloride (279 g, 1.48 mol) was suspended in DMSO (2.70 L) and treated with triethylamine (223 ml, 1.60 mol). The mixture was stirred at 25 °C for 30 min and treated with EDC-HC1 (354 g, 1.85 mol) and HOBT hydrate (283 g, 1.85 mol). The reaction mixture was stirred at rt for 16 h. After addition of triethylamine (292 ml, 2.09 mol), the mixture was cooled to 15 °C, diluted with water (10.1 L) maintaining the temperature below 30 °C, and stirred at 19-25 °C for 4 h. The resulting precipitate was filtered, washed twice with water (2.70 L), and dried under vacuum to give a crude product (695 g, wt-wt analysis = 78%).

For the further purification of the product, recrystallization was conducted. A crude product (20.00 g, 34.92 mmol) was suspended in a mixture of ethanol (190 ml) and water (10.00 ml) and heated to 75°C until a clear solution was obtained. The solution was allowed to cool to rt overnight. The precipitate was filtered, washed twice with a mixture of ethanol (30.0 ml) and water (30.0 ml), and dried under vacuum at 35 °C to give the title compound as an off white solid (14.0 g, 70% recovery from the crude and 90% yield based on wt-wt assay).

4-((3′-(((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)carbamoyl)-5′- (ethyl(tetrahydro-2H-pyran-4-yl)amino)-4′-methyl-[l,l’-biphenyl]-4-yl)methyl)morpholin- 4-ium bromide (Polymorph A): A crude N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)am

biphenyl]-3-carboxamide (595 g, 464 g based on wt-wt assay, 810.3 mmol) was suspended in ethanol (3.33 L). After heating to 70 °C, the mixture was treated with 48% aqueous HBr (97 ml, 850.8 mmol) and stirred at 70 °C for 30 min. The resulting orange-red solution was treated with ethyl acetate (3.33 L) maintaining the temperature above 60 °C. The mixture was slowly cooled to rt over 18 h. The mixture was cooled to 0 °C over 1 h and stirred at that temperature for 5.5 h. The resulting precipitate was filtered, washed twice with ethyl acetate (1.39 L), and dried under vacuum to give the title compound as an off white solid (515 g, 97% yield).

Recrystallization of Polymorph A: 4-((3′-(((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)carbamoyl)-5′-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4′-methyl-[l,l’-biphenyl]-4- yl)methyl)morpholin-4-ium bromide (0.50 g, 0.77 mmol; 95.6% pure by HPLC) was suspended in ethanol (3.0 mL) and heated to 80 °C until a clear solution was obtained. To the solution was added MTBE (5.0 mL) slowly. The resulting solution was allowed to cool to 18-22 °C over 3 h and stirred at 18-22 °C for 15 h. The precipitate was filtered, washed twice with MTBE (2 mL) and dried under vacuum to give 0.45 g of the title compound (89% recovery, 96.6% pure by HPLC).

Compound I is protonated at the nitrogen of the morpholino substituent, providing a monohydrobromide of Compound I having the following structure:

This particular monohydrobromide can be referred to as “4-((3′-(((4,6-dimethyl-2-oxo- l,2-dihydropyridin-3-yl)methyl)carbamoyl)-5′-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4′- methyl-[l, -biphenyl]-4-yl)methyl)morpholin-4-ium bromide.” Figure 11 depicts the X-ray crystal structure of this particular salt form.

…………………………………………………………..

see

WO-2015057859

Eisai Research Institute; Epizyme Inc

Novel crystalline polymorphic form C of tazemetostat, useful for treating an EZH2-mediated cancer, including non-Hodgkin’s lymphoma and breast cancer.

…………………

Synthesis

Trade Names

Country Trade Name Vendor Annotation
USA Tazverik Epizyme, 2020

Formulations

  • oral tabs. and suspension

References

    • Knutson, S. K. et al., Proc. Natl. Acad. Sci USA, (2013) 110(19), 7922-7927.
    • WO 2012 142504 (Epizyme/Eisai Co; 18.10.2012; appl. 13.4.2012; USA-prior. 13.4.2011).
    • WO 2013 155317 (Epizyme/Eisai Co; 17.10.2013; appl. 11.4.2013).
    • WO 2015 057859 (Epizyme/Eisai Co; 23.4.2015; appl. 15.10.2014; USA-prior. 16.10.2013).
  • EZH2 inhibitors for treating lymphona:

    • WO 2015 195848 (Epizyme; 23.12.2015; appl. 17.6.2015; USA-prior. 17.6.2014).

////////

PAPER

RSC Advances (2015), 5(33), 25967-25978

http://pubs.rsc.org/en/content/articlelanding/2015/ra/c5ra02365c#!divAbstract

RSC Adv., 2015,5, 25967-25978,

DOI: 10.1039/C5RA02365C

The histone lysine methyltransferase EZH2 has been implicated as a key component in cancer aggressiveness, metastasis and poor prognosis. This study discovered a new class of hexahydroisoquinolin derivatives as EZH2 inhibitors. A structure–activity relationship study showed that the steric hindrance was important to the activity for EZH2. A preliminary optimization study led to the discovery of several potent compounds with low nanomolar to sub-nanomolar potency for EZH2. Biological evaluation indicated that SKLB1049 was a highly potent with improved solubility compared to EPZ6438, SAM-competitive, and cell-active EZH2 inhibitor that decreased global H3K27me3 in SU-DHL-6 and Pfeiffer lymphoma cells in a concentration- and time-dependent manner. Further study indicated that SKLB1049 caused cell arrest in G0/G1 phase. These compounds would be useful as chemical tools to further explore the biology of EZH2 and provided us with a start point to develop new EZH2 inhibitors.

Graphical abstract: Design, synthesis and biological evaluation of novel 1-methyl-3-oxo-2,3,5,6,7,8-hexahydroisoquinolins as potential EZH2 inhibitors

In vitro protocol:

Proc Natl Acad Sci U S A. 2013 May 7;110(19):7922-7.

In vivo protocol:

Proc Natl Acad Sci U S A. 2013 May 7;110(19):7922-7.

References

1: Knutson SK, Warholic NM, Johnston LD, Klaus CR, Wigle TJ, Iwanowicz D, Littlefield BA, Porter-Scott M, Smith JJ, Moyer MP, Copeland RA, Pollock RM, Kuntz KW, Raimondi A, Keilhack H. Synergistic Anti-Tumor Activity of EZH2 Inhibitors and Glucocorticoid Receptor Agonists in Models of Germinal Center Non-Hodgkin Lymphomas. PLoS One. 2014 Dec 10;9(12):e111840. doi: 10.1371/journal.pone.0111840. eCollection 2014. PubMed PMID: 25493630; PubMed  Central PMCID: PMC4262195.

2: Knutson SK, Kawano S, Minoshima Y, Warholic NM, Huang KC, Xiao Y, Kadowaki T,  Uesugi M, Kuznetsov G, Kumar N, Wigle TJ, Klaus CR, Allain CJ, Raimondi A, Waters NJ, Smith JJ, Porter-Scott M, Chesworth R, Moyer MP, Copeland RA, Richon VM, Uenaka T, Pollock RM, Kuntz KW, Yokoi A, Keilhack H. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol Cancer Ther. 2014 Apr;13(4):842-54. doi: 10.1158/1535-7163.MCT-13-0773. Epub 2014 Feb 21. PubMed PMID: 24563539

3. Inhibitors of human histone methyltransferase EZH2, and methods of use thereof for treating cancer. By Kuntz, Kevin W.; Knutson, Sarah K.; Wigle, Timothy James Nelson . From U.S. Pat. Appl. Publ. (2013), US 20130040906 A1 20130214.

4. Aryl-or heteroaryl-substituted benzamide compounds as anticancer agents and their preparation By Kuntz, Kevin Wayne; Chesworth, Richard; Duncan, Kenneth William; Keilhack, Heike; Warholic, Natalie; Klaus, Christine; Zheng, Wanjun; Seki, Masashi; Shirotori, Syuji; Kawano, Satoshi From PCT Int. Appl. (2012), WO 2012142504 A1 20121018.

5: Knutson SK, Warholic NM, Wigle TJ, Klaus CR, Allain CJ, Raimondi A, Porter Scott M, Chesworth R, Moyer MP, Copeland RA, Richon VM, Pollock RM, Kuntz KW, Keilhack H. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A. 2013 May 7;110(19):7922-7. doi: 10.1073/pnas.1303800110. Epub 2013 Apr 25. PubMed PMID: 23620515; PubMed Central PMCID: PMC3651445.

WO2013155317A1 * Apr 11, 2013 Oct 17, 2013 Epizyme, Inc. Salt form of a human hi stone methyltransf erase ezh2 inhibitor
WO2013155464A1 * Apr 12, 2013 Oct 17, 2013 Epizyme, Inc. Combination therapy for treating cancer
WO2014049488A1 * Sep 16, 2013 Apr 3, 2014 Pfizer Inc. Benzamide and heterobenzamide compounds
WO2014062732A1 * Oct 15, 2013 Apr 24, 2014 Epizyme, Inc. Substituted benzene compounds
WO2014062733A2 * Oct 15, 2013 Apr 24, 2014 Epizyme, Inc. Substituted benzene compounds
WO2014172044A1 * Mar 14, 2014 Oct 23, 2014 Epizyme, Inc. Substituted benzene compounds
WO2015004618A1 * Jul 9, 2014 Jan 15, 2015 Glaxosmithkline Intellectual Property (No.2) Limited Enhancer of zeste homolog 2 inhibitors
WO2015010049A1 * Jul 18, 2014 Jan 22, 2015 Epizyme, Inc. Substituted benzene compounds
WO2015010078A2 Jul 18, 2014 Jan 22, 2015 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
WO2011140325A1 * May 5, 2011 Nov 10, 2011 Glaxosmithkline Llc Indazoles
WO2012142504A1 * Apr 13, 2012 Oct 18, 2012 Eisai Co., Ltd. Aryl-or heteroaryl-substituted benzene compounds
WO2014062720A2 * Oct 15, 2013 Apr 24, 2014 Epizyme, Inc. Methods of treating cancer
WO2011140324A1 * May 5, 2011 Nov 10, 2011 Glaxosmithkline Llc Indoles
WO2011140325A1 * May 5, 2011 Nov 10, 2011 Glaxosmithkline Llc Indazoles
WO2012005805A1 * May 5, 2011 Jan 12, 2012 Glaxosmithkline Llc Azaindazoles
US4522811 Jul 8, 1982 Jun 11, 1985 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5763263 Jul 24, 1996 Jun 9, 1998 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
US7563589 May 27, 2005 Jul 21, 2009 The University Of North Carolina At Chapel Hill Including EED, EZH2 and SUZ12 wherein the reconstituted complex has histone methyltransferase (HMTase) activity for lysine 27 of histone H3 (H3-K27); cancer

References

  1. Jump up to:a b c d e f g h i j k l m n o p q r “FDA approves first treatment option specifically for patients with epithelioid sarcoma, a rare soft tissue cancer”U.S. Food and Drug Administration (FDA) (Press release). 23 January 2020. Retrieved 23 January 2020.  This article incorporates text from this source, which is in the public domain.
  2. ^ Lue JK, Amengual JE (October 2018). “Emerging EZH2 Inhibitors and Their Application in Lymphoma”. Curr Hematol Malig Rep13 (5): 369–382. doi:10.1007/s11899-018-0466-6PMID 30112706S2CID 52010283.
  3. ^ “Tazemetostat”NCI Drug Dictionary. National Cancer Institute.
  4. Jump up to:a b c “Drug Trials Snapshots: Tazverik”U.S. Food and Drug Administration (FDA). 23 January 2020. Retrieved 22 February 2020.  This article incorporates text from this source, which is in the public domain.

External links

Tazemetostat
Tazemetostat.svg
Clinical data
Trade names Tazverik
Other names EPZ-6438
AHFS/Drugs.com Monograph
MedlinePlus a620018
License data
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C34H44N4O4
Molar mass 572.750 g·mol−1
3D model (JSmol)
About  EPZ-­‐6438  Epizyme  is developing  EPZ-­‐6438,  a  small  molecule  inhibitor  of  EZH2  created  with  our
proprietary  product  platform,  for  the  treatment  of  non-­‐Hodgkin  lymphoma  patients and  patients  with  INI1-­‐deficient  solid  tumors.  In  many  human  cancers,  misregulated  EZH2  enzyme  activity  results  in  misregulation  of  genes  that  control  cell  proliferation—without  these  control  mechanisms,  cancer  cells  are  free  to  grow
About  Epizyme,  Inc.
Epizyme,  Inc.  is  a  clinical  stage  biopharmaceutical  company  creating  novel  epigenetic therapeutics  for  cancer  patients.
Epizyme  has  built  a  proprietary  product  platform  that  the  company  uses  to  create  small  molecule  inhibitors  of  a  96 member  class  of  enzymes  known  as  histone  methyltransferases,  or  HMTs.  HMTs  are  part  of  the  system  of  gene  regulation,  referred
to  as  epigenetics,  that  controls  gene  expression.  Genetic  alterations  can  result  in  changes to
the  activity  of  HMTs,  making  them  oncogenic  (cancer -­‐causing).  By  focusing  on  the  genetic  drivers  of  cancers,  Epizyme’s  targeted  science  seeks  to  match  the  right  medicines  with  the  right  patients.

Epizyme®, Inc.
400 Technology Square, 4th Floor
Cambridge, MA 02139

Phone: (617) 229-5872
Fax: (617) 349-0707
contact@Epizyme.com

Victoria Richon, vice president of biological sciences, Epizyme Inc.

Jason Rhodes (left) has been appointed to president of Epizyme Inc.,

100 Technology Square

Central Square – The square – Cambridge, MA, United States
/////////TAZVERIK, EPIZYME, FDA 2020, APPROVALS 2020, Tazemetostat, EPZ-6438, EPZ 6438

K 912, NC 6300, Epirubicin nano


Epirubicin.png

PHASE 1 JAPAN SOLID TUMOURS

DNA/RNA Synthesis Inhibitor

WITH Nano Carrier Co.,Ltdhttp://pdf.irpocket.com/C4571/qnwX/eFou/vG1J.pdf

KOWA COMPANY LTD

CAS FREE FORM. 56420-45-2

Smiles

NC-6300, an epirubicin-incorporating micelle, extends the antitumor effect and reduces the cardiotoxicity of epirubicin.

Epirubicin is widely used to treat various human tumors. However, it is difficult to achieve a sufficient antitumor effect because of dosage limitation to prevent cardiotoxicity. We hypothesized that epirubicin-incorporating micelle would reduce cardiotoxicity and improve the antitumor effect. NC-6300 comprises epirubicin covalently bound to PEG polyaspartate block copolymer through an acid-labile hydrazone bond. The conjugate forms a micellar structure of 40-80 nm in diameter in an aqueous milieu. NC-6300 (10, 15 mg/kg) and epirubicin (10 mg/kg) were given i.v. three times to mice bearing s.c. or liver xenograft of human hepatocellular carcinoma Hep3B cells. Cardiotoxicity was evaluated by echocardiography in C57BL/6 mice that were given NC-6300 (10 mg/kg) or epirubicin (10 mg/kg) in nine doses over 12 weeks. NC-6300 showed a significantly potent antitumor effect against Hep3B s.c. tumors compared with epirubicin. Moreover, NC-6300 also produced a significantly longer survival rate than epirubicin against the liver orthotopic tumor of Hep3B. With respect to cardiotoxicity, epirubicin-treated mice showed significant deteriorations in fractional shortening and ejection fraction. In contrast, cardiac functions of NC-6300 treated mice were no less well maintained than in control mice. This study warrants a clinical evaluation of NC-6300 in patients with hepatocellular carcinoma or other cancers.

K-912(NC-6300)の概要 K-912(NC-6300)は、世界的に幅広く使用されているアントラサイクリン系の抗が ん剤の一つであるエピルビシンを内包したミセル化ナノ粒子製剤で、その特性により、 エピルビシンの有する心毒性の軽減が期待できます。さらに、pH 応答性システムを採 用することで、腫瘍細胞内でのエピルビシンの放出量を高め、既存のエピルビシンに比 べより強力な抗腫瘍効果が期待できます。

Epirubicin is an anthracycline drug used for chemotherapy. It can be used in combination with other medications to treat breast cancer in patients who have had surgery to remove the tumor. It is marketed by Pfizer under the trade name Ellence in the US andPharmorubicin or Epirubicin Ebewe elsewhere.

Similarly to other anthracyclines, epirubicin acts by intercalating DNA strands. Intercalation results in complex formation which inhibits DNA and RNA synthesis. It also triggers DNA cleavage by topoisomerase II, resulting in mechanisms that lead to cell death. Binding to cell membranes and plasma proteins may be involved in the compound’s cytotoxic effects. Epirubicin also generates free radicalsthat cause cell and DNA damage.

Epirubicin is favoured over doxorubicin, the most popular anthracycline, in some chemotherapy regimens as it appears to cause fewer side-effects. Epirubicin has a different spatial orientation of the hydroxyl group at the 4′ carbon of the sugar – it has the opposite chirality – which may account for its faster elimination and reduced toxicity. Epirubicin is primarily used against breast and ovarian cancer, gastric cancer, lung cancer and lymphomas.

Development history

The first trial of epirubicin in humans was published in 1980.[1] Upjohn applied for approval by the U.S. Food and Drug Administration(FDA) in node-positive breast cancer in 1984, but was turned down because of lack of data.[2] It appears to have been licensed for use in Europe from around this time however.[3] In 1999 Pharmacia (who had by then merged with Upjohn) received FDA approval for the use of epirubicin as a component of adjuvant therapy in node-positive patients.

Patent protection for epirubicin expired in August 2007.

References

  1.  Bonfante, V; Bonadonna, G; Villani, F; Martini, A (1980). “Preliminary clinical experience with 4-epidoxorubicin in advanced human neoplasia”. Recent results in cancer research 74: 192–9. PMID 6934564. PM6934564.
  2.  “On Target”.
  3.  According to the proprietary database iddb.com

External links

1H NMR PREDICT

Epirubicin NMR spectra analysis, Chemical CAS NO. 56420-45-2 NMR spectral analysis, Epirubicin H-NMR spectrum

 

 

13C NMR PREDICT

Epirubicin NMR spectra analysis, Chemical CAS NO. 56420-45-2 NMR spectral analysis, Epirubicin C-NMR spectrum

 

COSY

 

COSY NMR prediction EPI

 

 

1H NMR

 

1H  NMR prediction EPI

 

 

 

1H  NMR prediction EPI 2

 

 

 

Epirubicin
Epirubicin.png
Epirubicin ball-and-stick.png
Systematic (IUPAC) name
(8R,10S)-10-((2S,4S,5R,6S)-4-amino-5-hydroxy-6-methyltetrahydro-2H-pyran-2-yl)-6,8,11-trihydroxy-8-(2-hydroxyacetyl)-1-methoxy-7,8,9,10-tetrahydrotetracene-5,12-dione
Clinical data
Trade names Ellence
AHFS/Drugs.com monograph
MedlinePlus a603003
  • ℞-only (U.S.), POM (UK)
Intravenous
Pharmacokinetic data
Bioavailability NA
Protein binding 77%
Metabolism Hepatic glucuronidationand oxidation
Excretion Biliary and renal
Identifiers
56420-45-2 Yes
L01DB03
PubChem CID 41867
DrugBank DB00445 Yes
ChemSpider 38201 Yes
UNII 3Z8479ZZ5X Yes
KEGG D07901 Yes
ChEBI CHEBI:47898 Yes
ChEMBL CHEMBL417 Yes
Chemical data
Formula C27H29NO11
543.519 g/mol

 

 

KOWA COMPANY LTD

Nano Carrier Co

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

GSK 923295, a CENP-E Inhibitor


GSK-923295A

1088965-37-0

Synonym: GSK-923295; GSK 923295; GSK923295.

CENP-E Inhibitor

IUPAC/Chemical name: 

3-Chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide

3-​chloro-​N-​[(1S)​-​2-​[[2-​(dimethylamino)​acetyl]​amino]​-​1-​[[4-​[8-​[(1S)​-​1-​hydroxyethyl]​imidazo[1,​2-​a]​pyridin-​2-​yl]​phenyl]​methyl]​ethyl]​-​4-​(1-​methylethoxy)​- Benzamide,

3-Chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide

3-Chloro-N-[(1S)-2-[(N,N-dimethylglycyl)amino]-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

3-Chloro-N-[1-(N,N-dimethylglycinamido)-3-[4-[8-[1(S)-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl]phenyl]propan-2(S)-yl]-4-isopropoxybenzamide

C32H38ClN5O4
Exact Mass: 591.26123
Molecular Weight: 592.12822
Elemental Analysis: C, 64.91; H, 6.47; Cl, 5.99; N, 11.83; O, 10.81

Kinesin-like protein KIF11 inhibitor; Centromere protein E inhibitor

GSK-923295 is a novel antimitotic inhibitor of centromere-associated protein E (CENP-E) with potential anticancer activity. GSK923295A demonstrated significant antitumor activity against solid tumor models, inducing CRs in Ewing sarcoma, rhabdoid, and rhabdomyosarcoma xenografts.

GSK-923295, a small-molecule inhibitor of centromere associated protein (CENP), is in early clinical development at Cytokinetics for the treatment of refractory cancer. No recent development has been reported for early clinical research which had been ongoing at GlaxoSmithKline.

Clinical study showed that GSK923295  had dose-proportional pharmacokinetics and a low number of grade 3 or 4 adverse events. The observed incidence of myelosuppression and neuropathy was low. Further investigations may provide a more complete understanding of the potential for GSK923295 as an antiproliferative agent.

GSK923295 is a first-in-class, specific allosteric inhibitor of CENP-E kinesin motor ATPase with Ki of 3.2 nM, and less potent to mutant I182 and T183. Phase 1.

The compound potently inhibits CENP-E ATPase activity and exerts broad-spectrum antiproliferative activity against cancer cells and xenografts. GSK-923295 has demonstrated a broad spectrum of activity against a range of human tumor xenografts grown in nude mice, including models of colon, breast, ovarian, lung and other tumors.

Cytokinetics was developing GSK-923295, the lead from a series of small-molecule mitotic kinesin spindle protein inhibitors, for treating cancer including advanced solid tumors. However, since October 2014, the program was no longer listed on the Cytokinetics’ website

In 2001, a strategic alliance was established between Cytokinetics and GlaxoSmithKline to discover, develop and commercialize novel small-molecule therapeutics targeting mitotic kinesins for applications in the treatment of cancer and other diseases.

WP_000314

…………………….

PATENT

US8772507

http://www.google.com/patents/US8772507

1,1-Dimethylethyl [(1S)-2-(4-bromophenyl)-1-(hydroxymethyl)ethyl]carbamate

To a solution of 4-bromo-N-{[(1,1-dimethylethyl)oxy]carbonyl}-L-phenylalanine (72.6 mmol), in anhydrous diethyl ether (550 mL) at 0° C. was added slowly lithium aluminum hydride, 95% (108.9 mmol). The resulting solution was stirred for an additional 2 h at 0° C. The reaction was then carefully quenched with a saturated aqueous solution of sodium bicarbonate (73 mL) which stirred at RT for half an hour. Lithium aluminium salts crashed out of solution and were removed by filtration. The filtrate was concentrated and vacuum pumped for 24 h to afford the title product as a white solid (97%). ESMS [M+H]+: 331.2.

1,1-Dimethylethyl {(1S)-2-(4-bromophenyl)-1-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]ethyl}carbamate

To a solution of 1,1-dimethylethyl [(1S)-2-(4-bromophenyl)-1-(hydroxymethyl)ethyl]carbamate (70.6 mmol), tripheylphosphine (84.7 mmol), and phthalimide (84.7 mmol) in anhydrous tetrahydrofuran (550 mL) at 0° C. was added dropwise diisopropyl azodicarboxylate (84.7 mmol) over 10 minutes. The reaction continued to stir allowing to warm to RT over 5 h. The reaction was then concentrated in vacuo and product was triturated out of solution using ethyl acetate (500 mL). The precipitate was filtered, washed with ethyl acetate (3×100 mL), and dried to afford the title product as a white solid (57%). ESMS [M+H]+: 460.4.

1,1-Dimethylethyl {(1S)-2-[4-(bromoacetyl)phenyl]-1-[(1,3-d oxo-1,3-dihydro-21′-isoindol-2-yl)methyl]ethyl}carbamate

A solution of 1,1-dimethylethyl {(1S)-2-(4-bromophenyl)-1-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]ethyl}carbamate (21.7 mmol), 1-ethoxyvinyltri-n-butylin (43.5 mmol), and trans-dichlorobis(triphenylphosphine)palladium(II) (5 mol %) were stirred in anhydrous dioxane (300 mL) at 100° C. for 3 h. The reaction was then concentrated in vacuo and redissolved in a solution of tetrahydrofuran and water (3:1, 400 mL). The mixture was treated with N-bromosuccinimide (108.8 mmol) and stirred at RT for half an hour. The reaction solution was then concentrated to dryness and redissolved in ethyl acetate (150 mL). Precipate formed upon addition of hexanes (350 mL) and was filtered and dried to afford the title product as yellow solid (71%). ESMS [M+H]+: 502.4.

1,1-Dimethylethyl [(1S)-2-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]carbamate

A mixture of 1,1-dimethylethyl{(1S)-2-{4-(bromoacetyl)phenyl]-1-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]ethyl}carbamate (1.90 g, 3.79 mmol), 1-(2-amino-3-pyridinyl)ethanol (0.523 g, 3.79 mmol), and solid sodium bicarbonate (0.398 g, 4.72 mmol) in isopropanol (24 mL) was refluxed for 3.0 h. The mixture was concentrated in vacuo and the residue dissolved in ethyl acetate, washed with water and saturated sodium chloride, dried (Na2SO4), and concentrated to give the title compound (1.79 g, 87%) as a light pink solid. MS (ES+) m/e 541 [M+H]+.

3-Chloro-N-[(1S)-2-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

A mixture of 1,1-dimethylethyl [(1S)-2-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]carbamate (1.79 g, 3.31 mmol) and 4 M HCl in 1,4-dioxane (20 mL, 80 mmol) was stirred at room temperature for 45 minutes. The reaction was concentrated to dryness and redissolved in DMF (30 mL). To this solution was added N,N-diisopropylethylamine (2.14 g, 16.55 mmol) and pentafluorophenyl 3-chloro-4 [(1-methylethyl)oxy]benzoate (1.36 g, 3.31 mmol). The mixture was stirred overnight at room temperature, diluted with water, and extracted into ethyl acetate. The extracts were washed with water, dried (Na2SO4), and concentrated in vacuo to give the title compound (2.10 g, 100%) as a tan solid. MS (ES+) m/e 637 [M+H]+.

N-[(1S)-2-Amino-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide

A mixture of 3-chloro-N-[(1S)-2-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide (2.10 g, 3.30 mmol) and hydrazine monohydrate (0.83 g, 16.5 mmol) in ethanol (30 mL) was heated at 57° C. overnight. The reaction was cooled, diluted with ethanol, filtered, and concentrated to give the title compound (1.67 g, 100%) as a pale yellow powder. MS (ES+) m/e 507 [M+H]+.

3-Chloro-N-[(1S)-2-[(N,N-dimethylglycyl)amino]-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

A mixture of N-[(1S)-2-amino-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide (0.912 g, 1.80 mmol), EDCI (0.69 g, 3.6 mmol), N,N-diisopropylethylamine (0.466 g, 3.6 mmol), and N,N-dimethylglycine (0.372 g, 3.6 mmol) in methylene chloride (17 mL) was stirred overnight at room temperature. The reaction was diluted with water, washed with brine, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography on silica gel (8%-10% MeOH:CH2Cl2) to give the title compound (0.515 g, 48%) as a pale yellow solid. MS (ES+) ink 592 [M+H]+.

………………….

WO2005107762

https://www.google.im/patents/WO2005107762A2

Example 1

cheme E:

ide

NaHCOj, IPA 100 ‘C

1 , 1 -Dimethylethyl [( 1 S)-2-(4-bromophenyl)- 1 -(hydroxymethyl)ethyl]carbamate:

To a solution of 4-bromo-N-{[(l ,1 -dimethylethyl)oxy] carbonyl }-L- phenylalanine (72.6 mmol), in anhydrous diethyl ether (550 mL) at 0 °C was added slowly lithium aluminum hydride, 95% (108.9 mmol). The resulting solution was stiπed for an additional 2 h at 0 °C, The reaction was then carefully quenched with a saturated aqueous solution of sodium bicarbonate (73 mL) which stiπed at RT for half an hour. Lithium aluminium salts crashed out of solution which were removed by filtration. The filtrate was concentrated and vacuum pumped for 24 h to afford the title product as a white solid (97%).

ESMS [M+H]+: 331.2.

1,1 -Dimethylethyl {(lS)-2-(4-bromophenyl)-l-[(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)methyl]ethyl}carbamate:

To a solution of 1 ,1 -dimethylethyl [(lS)-2-(4-bromophenyl)-l –

(hydroxymethyl)ethyl]carbamate (70.6 mmol), tripheylphosphine (84.7 mmol), and phthalimide (84.7 mmol) in anhydrous tetrahydrofuran (550 mL) at 0 °C was added dropwise diisopropyl azodi carboxyl ate (84.7 mmol) over 10 minutes. The reaction continued to stir allowing to wai to RT over 5h, The reaction was then concentrated in vacuo and product was tritarated out of solution usingl acetate (500 mL). The precipitate was filtered, washed with ethyl acetate (3 x 100 mL), and dried to afford the title product as a white solid (57%).

ESMS [M+H]+: 460.4.

1 ,1 -Dimethylethyl {(15)-2-[4-(bromoacetyl)phenyl]-l -[(l,3-dioxo-l ,3-dihydro-2H-isoindol- 2-yl)methyl]ethyl}carbamate:

A solution of 1,1 -dimethyl ethyl {(lS)-2-(4-bromophenyl)-l-[(l,3-dioxo-l,3- dihydro-2H-isoindol-2-yl)methyl]ethyl}carbamate (21.7 mmol), 1-ethoxyvinyltri-n-butylin (43.5 mmol), and /ra/?s–dichlorobis(triphenylphospine)palladιum(II) (5 mol%) were stiπed in anhydrous dioxane (300 mL) at 100 °C for 3h. The reaction was then concentrated in vacuo and redissolved in a solution of tetrahydrofuran and water (3:1, 400mL) and treated with N- bromosuccinimide (108.8 mmol) and stined at RT for half an hour. The reaction solution was then concentrated to dryness and redissolved in ethyl acetate (150 mL) and precipate formed upon addition of hexanes (350 mL). The precipitate was filtered and dried to afford the title product as yellow solid (71%). ESMS [M+Η]+: 502.4. l,l-Dimethylethyl [(lS)-2-(l ,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)-l-({4-[8-(l- hydroxyethyl)imidazo[l,2-β]pyridin-2-yl]phenyl}methyl)ethyl]carbamate:

A mixture of l!l-dimethylethyl{(lS)-2-{4-(biOinoacetyl)phenyl]-l-[(l,3- dioxo-l ,3-dihydro-2H-isoindol-2-yl)methyl]ethyl}carbamate (1.90 g, 3.79 mmol), l-(2- amino-3-pyτidinyl)ethanol (0.523 g, 3.79 mmol), and solid sodium bicarbonate (0.398 g, 4,72 mmol) in isopropanol (24 mL) was refluxed for 3.0 h. and concentrated in vacuo. The residue was dissolved in ethyl acetate, washed with water and saturated sodium chloride, dried (Na2S04), and concentrated to give the title compound (1.79 g, S7%) as a light pink solid. MS(ES+) m/e 541 [M+Η]+.

3-Chloro-N-[(lS)-2-(l,3-dioxo-l ,3-dihydro-2H-isoindol-2-yl)-l-({4-[8-(l- hydroxyethyl)imidazo[l,2-Λ]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(l – methylethyl)oxy]benzamide:

A mixture of 1,1 -dimethylethyl [(15)-2-(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)-l-({4-[8-(l-hydroxyethyl)imidazo[l,2-fl]pyridin-2-yl]phenyl}methyl)ethyl]carbamate (1.79 g, 3.31 mmol) and 4M ΗC1 in 1,4-dioxane (20 mL, 80 mmol) was stirred at room temperature for 45 minutes. The reaction was concentrated to dryness ,redissolved in DMF (30 mL), and to this solution was added N,N-diisopropylethylamine (2.14 g, 16,55 mmol) and pentafluorophenyl 3-chloro-4 [(l-methylethyl)oxy]benzoate (1.36 g, 3.31 mmol). The mixture was stirred overnight at room temperature, diluted with water, and extracted into ethyl acetate. The extracts were washed with water, dried (Na SO ), and concentrated in vacuo to give the title compound (2.10 g, 100%) as a tan solid. MS(ES+) m/e 637 [M+H]+.

N-[(lS)-2-Amino-l-({4-[8-(l-hydroxyethyl)imidazo[l,2-α]p>tidin-2- yl]phenyl}methyl)eth)’l]-3-chloro-4-[(l-methylethyl)oxy]benzamide:

A mixture of 3-chloro-N-[(lS)-2-(l,3-dioxo-l ,3-dihydro-2N-isoindol-2-yl)-l-

({4-[8-(l -hydiOxyethyl)imidazo[l,2-β]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(l- methylethyl)oxy]benzamide (2.10 g, 3.30 mmol) and hydrazine monohydrate (0.83 g, 16.5 mmol) in ethanol (30 mL) was heated at 57°C ovemight. The reaction was cooled, diluted with ethanol, filtered, and concentrated to give the title compound(1.67 g, 100%) as a pale yellow powder. MS(ES+) m/e 507 [M+H]+.

3-Chloro-N-[(15)-2-[(7VN-dimethylglycyl)amino]-l-({4-[8-(l-hydroxyethyl)imidazo[l ,2- «]pyitdin-2-yl]phenyl}methyl)ethyl]-4-[(l-methylethyl)oxy]benzamide:

A mixture ofN-[(lS)-2-amino-l-({4-[S-(l-hydroxyethyl)imidazo[l,2- α]pyridin-2-yl]phenyl)methyl)ethyl]-3-chloro-4-[(l-methylethyl)oxy]benzamide (0.912 g, 1 ,80 mmol), EDCI (0.69 g, 3,6 mmol), NN-diisopropylethylamine (0.466 g, 3,6 mmol), and N,N-dimethylglycine (0.372 g, 3.6 mmol) in methylene chloride (17 mL) was stirred overnight at room temperature. The reaction was diluted with water, washed with brine, dried (Νa2S0 ), and concentrated. The residue was purified by flash chromatography on silica gel (8%-10% MeOH:CH2Cl2) to give the title compound ( 0.515 g, 48%) as a pale yellow solid. MS(ES+) m/e 592 [M+H]+.

SEE

WO2008 / 138561

………………..

Organic Process Research & Development (2010), 14(5), 1254-1263

Org. Process Res. Dev., 2010, 14 (5), pp 1254–1263
DOI: 10.1021/op100186c

http://pubs.acs.org/doi/abs/10.1021/op100186c

Abstract Image

The discovery and development of an efficient manufacturing route to the CENP-E inhibitor 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}−4-[(1-methylethyl)oxy]benzamide (GSK923295A) is described. The existing route to GSK923295A was expensive, nonrobust, used nonideal reagents, and consistently struggled to deliver the API needed for clinical studies. The new synthesis commences from the readily available l-phenylalaninol, which is smoothly converted through to GSK923295A using key Friedel−Crafts acylation as well as selective acylation chemistries. Downstream chemistry to GSK923295A is both high yielding and robust, and the resulting process has been demonstrated first on the kilo scale and subsequently in the pilot plant where 55 kg was successfully prepared. The resulting process is simple, uses cheaper raw materials, is greener in that it avoids using aluminum, tin, and bromination chemistries, and obviates the need for chromatographic purification. Also discussed are the route derived impurities, how they were unambiguously prepared to confirm structure and processing amendments to control their formation, and enhancements to the new process to facilitate future processing.

1H NMR (400 MHz, CD3OD) δH 1.34 (6H, d, J = 6.0, (CH3)2), 1.59 (3H, d, J = 7.0, CH3CH), 2.21 (6H, s, N(CH3)2), 2.87−3.01 (4H, m, CH2Ph and CH2N(CH3)2), 3.49 (2H, m, CH2NPhthal), 4.50 (1H, m, CHNH), 4.70 (1H, m, (CH3)2CHO)), 5.49 (1H, q, J = 7.0, CHOH), 6.88 (1H, t, J = 7.0, H-j), 7.08 (1H, d, J = 7.5, H-b), 7.33−7.37 (3H, m, H-k and H-d), 7.63 (1H, dd, J = 7.5 and 2.0, H-c), 7.78 (1H, s, H-a), 7.83 (2H, d, J = 7.0, H-e), 8.09 (1H, m, H-h), 8.27 (1H, d, J = 8.0, H-i);

13C NMR (100 MHz, CD3OD) δC 22.2, 24.1, 39.3, 43.8, 46.1, 53.0, 63.7, 66.2, 73.0, 110.4, 113.8, 115.3, 121.2, 124.5, 126.1, 127.5, 128.4, 128.5, 130.6, 130.7, 133.3, 136.0, 139.4, 145.1, 146.1, 157.6, 168.5 and 173.6;

HRMS (ESI+) m/z calculated for [M+H]+ C32H39N5O4Cl 592.2691, found 592.2684.

…………………….

PREDICTIONS

http://orgspectroscopyint.blogspot.in/2015/03/gsk-923295.html

gsk 923295 chemspider

Predict 13C carbon NMR spectra (1)

1H NMR PREDICT

Predict 1H proton NMR spectra

gsk 923295 chemspider 1

see http://orgspectroscopyint.blogspot.in/2015/03/gsk-923295.html

ACS Medicinal Chemistry Letters (2010), 1(1), 30-34

http://pubs.acs.org/doi/abs/10.1021/ml900018m

Abstract Image

Inhibition of mitotic kinesins represents a novel approach for the discovery of a new generation of anti-mitotic cancer chemotherapeutics. We report here the discovery of the first potent and selective inhibitor of centromere-associated protein E (CENP-E) 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide (GSK923295; 1), starting from a high-throughput screening hit, 3-chloro-4-isopropoxybenzoic acid 2. Compound 1 has demonstrated broad antitumor activity in vivo and is currently in human clinical trials.

SEE

WO-2015037460

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=F8D2DAAA427F9EBAB6B7CE67A7EE0772.wapp1nC?docId=WO2015037460&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

Method for producing optically active 3-(biphenyl-4-yl)-2-[(t-butoxycarbonyl)amino]propan-1-ol

Process for preparing optically active 3-(biphenyl-4-yl)-2-[(t-butoxycarbonyl)amino]propan-1-ol, useful as an intermediate in the synthesis of pharmaceuticals described in WO2005107762 and WO2008138561 (such as GSK-923295 and tubulysin derivatives respectively). Appears to be a new area of interest to the assignee.

…………..

WO2010118207

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2010118207&recNum=278&docAn=US2010030350&queryString=%28SYK%29%2520&maxRec=1655

References

1: Mayes PA, Degenhardt YY, Wood A, Toporovskya Y, Diskin SJ, Haglund E, Moy C, Wooster R, Maris JM. Mitogen-activated protein kinase (MEK/ERK) inhibition sensitizes cancer cells to centromere-associated protein E inhibition. Int J Cancer. 2013 Feb 1;132(3):E149-57. doi: 10.1002/ijc.27781. Epub 2012 Sep 28. PubMed PMID: 22948716.

2: Chung V, Heath EI, Schelman WR, Johnson BM, Kirby LC, Lynch KM, Botbyl JD, Lampkin TA, Holen KD. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein E (CENP-E), in patients with refractory cancer. Cancer Chemother Pharmacol. 2012 Mar;69(3):733-41. doi: 10.1007/s00280-011-1756-z. Epub 2011 Oct 22. PubMed PMID: 22020315.

3: Lock RB, Carol H, Morton CL, Keir ST, Reynolds CP, Kang MH, Maris JM, Wozniak AW, Gorlick R, Kolb EA, Houghton PJ, Smith MA. Initial testing of the CENP-E inhibitor GSK923295A by the pediatric preclinical testing program. Pediatr Blood Cancer. 2012 Jun;58(6):916-23. doi: 10.1002/pbc.23176. Epub 2011 May 16. PubMed PMID: 21584937; PubMed Central PMCID: PMC3163687.

4: Balamuth NJ, Wood A, Wang Q, Jagannathan J, Mayes P, Zhang Z, Chen Z, Rappaport E, Courtright J, Pawel B, Weber B, Wooster R, Sekyere EO, Marshall GM, Maris JM. Serial transcriptome analysis and cross-species integration identifies centromere-associated protein E as a novel neuroblastoma target. Cancer Res. 2010 Apr 1;70(7):2749-58. doi: 10.1158/0008-5472.CAN-09-3844. Epub 2010 Mar 16. PubMed PMID: 20233875; PubMed Central PMCID: PMC2848992.

5: Wood KW, Lad L, Luo L, Qian X, Knight SD, Nevins N, Brejc K, Sutton D, Gilmartin AG, Chua PR, Desai R, Schauer SP, McNulty DE, Annan RS, Belmont LD, Garcia C, Lee Y, Diamond MA, Faucette LF, Giardiniere M, Zhang S, Sun CM, Vidal JD, Lichtsteiner S, Cornwell WD, Greshock JD, Wooster RF, Finer JT, Copeland RA, Huang PS, Morgans DJ Jr, Dhanak D, Bergnes G, Sakowicz R, Jackson JR. Antitumor activity of an allosteric inhibitor of centromere-associated protein-E. Proc Natl Acad Sci U S A. 2010 Mar 30;107(13):5839-44. doi: 10.1073/pnas.0915068107. Epub 2010 Feb 18. PubMed PMID: 20167803; PubMed Central PMCID: PMC2851928.

GSK 2636771


 

 

 

 

 

 

Company: GlaxoSmithKline
Meant to treat: tumors with loss-of-function in the tumor suppressor protein PTEN (phosphatase and tensin homolog)- 2nd most inactivated tumor suppressor after p53- cancers where this is often the case include prostate and endometrial
Mode of action: inhibitor of phosphoinositide 3-kinase-beta (PI3K-beta). Several lines of evidence suggest that proliferation in certain PTEN-deficient tumor cell lines is driven primarily by PI3K-beta.
Medicinal chemistry tidbits: The GSK team seemed boxed in because in 3 out of 4 animals used in preclinical testing, promising drug candidates had high clearance. It turned out that a carbonyl group that they thought was critical for interacting with the back pocket of the PI3K-beta enzyme wasn’t so critical after all. When they realized they could replace the carbonyl with a variety of functional groups, GSK2636771 eventually emerged. GSK2636771B (shown) is the tris salt of GSK2636771.
Status in the pipeline: Phase I clinical trials……….http://cenblog.org/the-haystack/2012/03/liveblogging-first-time-disclosures-from-acssandiego/

CARMEN

Posted By on Mar 24, 2012

Phone: 202-872-4502

Fax: 202-872-8727 or -6381

 

 

1372540-25-4

1H-​Benzimidazole-​4-​carboxylic acid, 2-​methyl-​1-​[[2-​methyl-​3-​(trifluoromethyl)​phenyl]​methyl]​-​6-​(4-​morpholinyl)​-

2-Methyl-1-[[2-methyl-3-(trifluoromethyl)phenyl]methyl]-6-(4-morpholinyl)-1H-benzimidazole-4-carboxylic acid

GSK2636771 is a potent, orally bioavailable, PI3Kβ-selective inhibitor, sensitive to PTEN null cell lines.

Formula:C22H22F3N3O3
M.Wt:433.43

WO 2014158467

http://www.google.com/patents/WO2014158467A1?cl=en

According to another embodiment, the invention relates to a method of re- sensitizing BRAF inhibitor resistant melanoma brain metastases comprising the administration of a therapeutically effective amount of

(i) a compound of formula (I)

or a pharmaceutically acceptable salt thereof;

……………………………………………………………….

http://www.google.co.in/patents/WO2014108837A1?cl=en

 

A combination comprising:
(i) a compound of Structure (I):
I
or a pharmaceutically acceptable salt thereof;
………………………………………………
SYNTHESIS
 GSK 2636771
………………………………………………
Example 26
Preparation of methyl 2-methyl-6-(4-morpholinyl)-l-(l-naphthalenylmethyl)-lH- benzimidazole-4-carboxylate a) 3-amino-5-chloro-2-nitrobenzoic acid

Under nitrogen, to a solution of t-BuOK (156.8 g) and Cu(OAc)2 (3.6 g) in DMF (1.2 L) was added a solution of 5-chloro-2-nitrobenzoic acid (40.0 g) and MeONH2 HCl (33.2 g) in DMF (300 mL) at 0° C. After 3h the reaction was quenched by addition of H20 (2.5 L) and acidified with 10% HC1 solution to pH= 1.The mixture was extracted with EA (2 L x 2) and the combined organic layers were then washed with brine, dried over anhydrous Na2S04, filtered and concentrated in- vacuo to afford the crude product as a yellow solid (43.2g, yield 100%). 1H NMR (300 MHz, CDC13): δ ppm 6.88 (s, 1H, J= 2.4Hz), 6.91 (d, 1H, J= 2.4Hz), 8.08 (br s, 2H); LC-MS: m/e = 217 [M+l]+. b) methyl 3-amino-5-chloro-2-nitrobenzoate

A mixture of 3-amino-5-chloro-2-nitrobenzoic acid (43.2 g) and HATU (2-(lH-7- Azabenzotriazol-l-yl)~l,l,3,3-tetramethyl uronium hexafluorophosphate Methanaminium, commercially available) (76 g) in MeOH (81 mL), Et3N (83 mL) and THF (300 mL) was stirred at room temperature for 3h. When TLC showed no starting material, the solvent was removed in-vacuo and the residue was then diluted with EtOAc (2 L). It was then washed with brine (1 L><3) and dried over anhydrous Na2S04, filtered and concentrated in-vacuo. The residue was then purified by silica gel chromatography eluted with EtOAc : petroleum ether = 1 : 8 to afford the desired product as a yellow solid (29.5 g, yield 64%). 1H NMR (300 MHz, CDC13): δ ppm 3.90 (s, 3H, s), 5.85 (br s, 2H), 6.80 (d, 1H, J = 2.4 Hz), 6.90 (d, 1H, J = 2.4 Hz); LC-MS: m/e = 231 [M+l]+ . c) methyl 3-amino-5-(4-morpholinyl)-2-nitrobenzoate

A mixture of combined batches of methyl 3-amino-5-chloro-2-nitrobenzoate (39 g), morpholine (29.5 g) and K2C03 (47 g) was stirred in DMF (200ml) at 110 0 C for 5 h. The mixture was cooled to room temperature and poured into water (1 L). It was extracted with EtOAc (500 mL x 3). The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated in-vacuo to afford the desired product as a yellow solid (22 g, yield 46%). 1H NMR (300 MHz, CDC13): δ ppm 3.31 (t, 4H, J= 4.8 Hz), 3.82 (t, 4H, J= 4.8 Hz), 3.89 (s, 3H), 6.03 (d, 1H, J= 2.4 Hz), 6.34 (d, 1H, J= 2.4 Hz); LC- MS: m/e = 282 [M+l]+ . d) methyl 2-methyl-5-(4-morpholinyl)-lH-benzimidazole-7-carboxylate

To a solution of methyl 3-amino-5-(4-morpholinyl)-2-nitrobenzoate (22 g) stirring at reflux in HOAc (400 mL) was added iron powder in portions (13 g). After the addition, the mixture was stirred at reflux for 5 h. It was cooled to room temperature and the solvent was removed in- vacuo. The residue was neutralized with aqueous Na2C03 solution (1 L). It was extracted with EtOAc (500 mL x3). The combined organic layers were then concentrated in-vacuo and the residue was purified by silica gel chromatography eluted with MeOH : DCM = 1 : 30 to afford the desired product as a solid (16.6 g, yield 77%).

1H NMR (300 MHz, CDC13): δ ppm 2.67 (s, 3H), 3.17 (t, 4H, J= 4.8 Hz), 3.90 (t, 4H, J= 4.8 Hz), 3.98 (s, 3H), 7.44 (d, IH, J= 1.8 Hz), 7.54 (d, IH, J= 1.8 Hz);

LC-MS: m/e = 276 [M+l]+ .

Example 30

Preparation of methyl 2-methyl-l- {r2-methyl-3-(trifluoromethyl)phenyl1methyl|-6-(4- morpholinyl)- 1 H-benzimidazole-4-carboxylate

A solution of methyl 2-methyl-5-(4-morpholinyl)-lH-benzimidazole-7-carboxylate prepared as described in Example 26

Figure imgf000072_0001 methyl 2-methyl-5-(4-morpholinyl)-lH-benzimidazole-7-carboxylate

, step d (500mg, 1.8 mmol), l-(bromomethyl)-2-methyl-3- (trifluoromethyl)benzene (483 mg, 1.9 mmol)

l-(bromomethyl)-2-methyl-3- (trifluoromethyl)benzene

and K2C03 (497 mg, 3.6 mmol) in DMF (50 mL) was stirred at 80° C for 3 h. The reaction mixture was cooled to rt and poured into water (50 mL), extracted with EtOAc (30 mL x 3). The combined organic layers were washed with brine, dried over Na2S04 and concentrated. The resulting residue was purified by silica gel chromatography eluted with DCM : MeOH = 50 : 1 to give the crude product IE METHYL ESTER (230 mg, yield 29%), as a white solid.

1H NMR (300 MHz, DMSO-d6): δ ppm 2.39 (s, 3H), 2.54 (s, 3H), 3.08 (t, 4H, J=4.8 Hz), 3.72 (t, 4H, J=4.8 Hz), 3.89 (s, 3H), 5.57 (s, 2H), 6.27 (d, IH, J=7.5 Hz), 7.22 (t, IH, J=7.5 Hz), 7.27 (d, IH, J=2.4 Hz), 7.38 (d, IH, J=2.4 Hz) 7.60 (d, IH, J=7.5 Hz);

LC-MS: m/e = 448 [M+l]+

Example 31

Preparation of 2-methyl- 1 – { [2-methyl-3-(trifluoromethyl)phenyllmethyl| -6-(4-morpholiny0- 1 H-benzimidazole-4-carboxylic acidAn aqueous solution of 2 N LiOH (1.2 mL) was added to a solution of methyl 2-methyl- 1- {[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-lH-benzimidazole-4- carboxylate, prepared as described in Example 30 (180 mg, 0.4 mmol) in THF (10 mL) and stirred at 50° C for 1 h. When TLC showed no starting material remaining, the mixture was cooled to rt and THF was removed under reduced pressure. The pH of the mixture was acidified to pH 3. The suspension was filtered and the filtrate was collected, and washed with water (lOmL) to give the product as a white solid (152 mg, yield 88%).

1H NMR (300 MHz,DMSO-d6):

δ ppm 2.46 (s, 3H), 2.54 (s, 3H), 3.10 (t, 4H, J=4.8 Hz), 3.73 (t, 4H, J=4.8 Hz), 5.63 (s, 2H), 6.37 (d, IH, J=7.8 Hz), 7.26 (t, IH, J=7.8 Hz), 7.35 (d, IH, J=2.4 Hz), 7.44 (d, IH, J=2.4 Hz), 7.62 (d, IH, J=7.8 Hz);

LC-MS: m/e = 434 [M+l]

 

 

WO2010006225A1 * 10 Jul 2009 14 Jan 2010 Novartis Ag Combination of (a) a phosphoinositide 3-kinase inhibitor and (b) a modulator of ras/raf/mek pathway
WO2011038380A2 * 28 Sep 2010 31 Mar 2011 Glaxosmithkline Llc Combination
WO2012061683A2 * 4 Nov 2011 10 May 2012 Glaxosmithkline Llc Methods for treating cancer
US20120088767 * 3 Oct 2011 12 Apr 2012 Junya Qu Benzimidazole derivatives as pi3 kinase inhibitors
O2013019620A2 * Jul 27, 2012 Feb 7, 2013 Glaxosmithkline Llc Method of treating cancer using combination of braf inhibitor, mek inhibitor, and anti-ctla-4 antibody
US20120202822 * Oct 12, 2010 Aug 9, 2012 Kurtis Earl Bachman Combination

 

 

CARMEN DRAHL

Links

Carmen Drahl (@carmendrahl) | Twitter

www.linkedin.com/in/carmendrahl/en

http://www.ddn-news.com/

http://cenblog.org/the-safety-zone/

Carmen Drahl – Google+

Carmen Drahl

 

Award-winning science communicator and social media power user based in Washington, DC.

Specialties: interviewing, science writing, social media, Twitter, Storify, YouTube, public speaking, hosting, video production, iPhone videography, non-linear video editing, blogging (WordPress and Blogger), HTML website coding

Education

Princeton University

Ph.D., Chemistry

2002 – 2007

Ph.D. with Erik J. Sorensen
She was on a team that completed the first total synthesis of abyssomicin C, a molecule found in small quantities in nature that showed hints of promise as a potential antibiotic. I constructed molecular probes from abyssomicin for proteomics studies of its biological activity.

M.A. with George L. McLendon
worked toward developing a drug conjugate as a potential treatment for cancer. I synthesized a photosensitizer dye-peptide conjugate for targeting the cell death pathway called apoptosis.

image

At a reception before the Alumni Day luncheon, President Tilghman (third from left) congratulated the winners of the University’s highest awards for students: (from left) Pyne Prize winners Lester Mackey and Alisha Holland; and Jacobus Fellowship recipients Sarah Pourciau, Egemen Kolemen and Carmen Drahl. Unable to attend the event was Jacobus Fellowship winner William Slauter. (photo: Denise Applewhite

 

 

B.A., Chemistry

1998 – 2002

Graduated summa cum laude with specialized honors in chemistry. Honors thesis entitled “Structural, kinetic, and mechanistic studies: the protein tyrosine phosphatases CD45 and PTP1B”

Activities and Societies: Phi Beta Kappa

Carmen Drahl, Class of 2002,

 

Experience

Science Journalist

Freelance

January 2014 – Present Washington D.C. Metro Area

Multimedia science journalist – I deliver clean products on time. Experience in reporting on chemistry, food science, history of science, drug development, science education.

Senior Editor, Chemical & Engineering News

American Chemical Society

August 2007 – December 2014 (7 years 5 months)Washington D.C. Metro Area

Reporting:
Cover the science of chemistry for C&EN, the American Chemical Society’s weekly magazine, circulation 160,000. Track new research findings daily, particularly in forensic science, drug discovery, organic chemistry, and food science.

Video:
Doubled circulation to C&EN’s YouTube channel in 2013. Scripted, narrated, edited footage.
Managed a core team of 4 and collaborated with other reporters to produce 30 videos, some reproduced in The Atlantic, Scientific American, Eater National, The Daily Mail.

Incepted, scripted, and co-hosted “Speaking of Chemistry”, a monthly web show that summarizes top chemistry news for the busy scientist.

Social Media:
Developed magazine-wide best practices for YouTube videos and Twitter. Ran staff workshops about Storify, Slashdot, and Reddit.

Hosting/Public Speaking:
Topics include communicating chemistry simply, transitioning from a Ph.D. to careers in science communication. Moderated discussions on chemophobia, social media usage in the chemical sciences. On-camera co-host for web newscasts produced by ACS.

Innovation:
With C&EN art and web teams, developed first-for-the-magazine features, including a 90th anniversary commemorative timeline poster, a pullout guide to top conference speakers, interactive quizzes and database searches.

Carmen Drahl, senior editor of Chemical and Engineering News, used her Ph.D. in chemistry as a springboard into the career she envisioned for herself. Here she shares some advice that helped her make the decision.

Carmen Drahl made the transition to a writing career while earning a Ph.D. in chemistry at Princeton University. Born and raised in New Jersey, she now lives in Washington, D.C., and reports for Chemical and Engineering News (C&EN). At C&EN she has written about how new medications get their names, explained the science behind a controversial hair-straightening product, and covered the scientific firestorm sparked by an alleged arsenic life form. Her work has been featured on SiriusXM’s Doctor Radio, Radio New Zealand’s This Way Up, and elsewhere. Her coverage has also been recognized by MIT’s Knight Science Journalism Tracker.

(Open)1 honor or award
Scientific Cocktails: Award-winning video

Scientific Cocktails: Award-winning video

Speaking of Chemistry: All About Tinsel

Speaking of Chemistry: All About Tinsel

Carmen Drahl

Twitter Maven

World Central Kitchen

March 2013 – August 2014 (1 year 6 months)Washington D.C. Metro Area

I was the “voice of Twitter” for World Central Kitchen, the humanitarian organization founded by renowned Chef José Andrés. Doubled followers to Twitter account in 2013, developed Twitter strategy for projects and events. Edited Annual Report, press releases and other communication materials. Volunteered in person at outreach events.

Contributing Editor, AWIS Magazine

Association of Women in Science

December 2005 – August 2007 (1 year 9 months)

sHE reported and wrote profiles of prominent women scientists in a range of fields (molecular biology, physics, geoscience) for the Research Advances column in AWIS Magazine.

Writer, various publications

Princeton University

April 2005 – May 2007 (2 years 2 months)

She reported and wrote news for the Princeton University News Office’s Research Notes, and wrote news and features for the Princeton University Chemistry Department’s Industrial Affiliates Program Newsletter and Chemistry Alumni Newsletter.

Honors & Awards

Eddie Digital Award- Best Video (B-to-B)

FOLIO Magazine

December 2014

Porter Ogden Jacobus Fellowship

Princeton University

February 2007

NSF Graduate Research Fellowship

National Science Foundation

2002

Volunteer Experience & Causes

Board Member

Princeton Alumni Weekly Magazine

October 2013

Advisory Committee

American Institute of Physics News and Media Services

October 2013

Member, Graduate Alumni Leadership Council

Princeton University

2009 – 2012 (3 years)

INTERVIEW

Continuing with the tradition from last two years, I will occasionally post interviews with some of the participants of the ScienceOnline2010 conference that was held in the Research Triangle Park, NC back in January. See all the interviews in this series here. You can check out previous years’ interviews as well: 2008 and 2009.

Today, I asked Carmen Drahl, Associate Editor for Science/Technology/Education at Chemical & Engineering News (find her as @carmendrahl on Twitter) to answer a few questions.

Welcome to A Blog Around The Clock. Would you, please, tell my readers a little bit more about yourself? Where are you coming from (both geographically and philosophically)? What is your (scientific) background?

i-b183f89fe33d3d9f0b308a6cb30d9b5b-Carmen Drahl pic1.JPGIt’s a pleasure and a privilege to be interviewed, Bora.

Good conversations make me happy. School was fun for me (well, maybe not grad school) and that’s evolved into a desire to always be learning something new. I enjoy doing nothing as much as I enjoy doing things. On Mondays, if I’m not too busy, I take hip-hop dance classes.

My hometown is Hackettstown, New Jersey. M&M’s are made there. I got a bachelor’s in chemistry from Drew University and a Ph.D. in chemistry at Princeton. Scientifically my expertise hovers somewhere around the interface between organic chemistry and biochemistry. A short while after defending my dissertation, I moved to Washington DC to write for Chemical & Engineering News, and that’s where I’ve been for almost three years now.

When and how did you first discover science blogs?

Scandal led me to science blogs. Seriously. In March 2006 I was still an organic chemistry grad student. Everyone in my lab was buzzing about a set of retractions in the Journal of the American Chemical Society (disclosure: today I work for the American Chemical Society, which publishes JACS). A rising young organic chemistry star retracted the papers because work by one of his graduate students couldn’t be reproduced. It was a big deal and became an even bigger deal as the inevitable rumors (salacious and otherwise) surfaced. The blogosphere had the details first. So that’s where Google pointed me and the other members of my lab when we searched for more information. I learned about the awesome (but sadly now defunct) blogs Tenderbutton and The Endless Frontier, by Dylan Stiles and Paul Bracher, both chemistry grad students like me. I also discovered the solid mix of chemistry and pharma at Derek Lowe’s In the Pipeline, which is still the first blog I visit every day.

Tell us a little more about your career trajectory so far: interesting projects past and present?

i-b7bd4d4568d9689c2daf400303c886c3-Carmen Drahl pic2.JPGBy the time I discovered science blogs I knew my career goals were changing. I’d already been lucky enough to audit a science writing course at Princeton taught by Mike Lemonick from TIME, and thought that maybe science writing was a good choice for me. After reading chemistry blogs for a while I realized “Hey, I can do this!” and started my own blog, She Blinded Me with Science, in July 2006. It was the typical grad student blog, a mix of posts about papers I liked and life in the lab.

At C&E News I’ve contributed to its C&ENtral Science blog, which premiered in spring 2008. I’ve experimented with a few different kinds of posts- observations and on-the-street interviews when I run into something chemistry-related in DC, in-depth posts from meetings, and video demos of iPod apps. One of my favorite things to do is toy with new audio/video/etc technology for the blog.

What is taking up the most of your time and passion these days? What are your goals?

In March I just started a new era in my web existence- I’m becoming a pharma blogger. I’m the science voice at The Haystack, C&E News’s new pharma blog and one of seven new blogs the magazine launched last month. My co-blogger is the talented Lisa Jarvis, who’s written about the business side of pharma for ten years and who brings a solid science background to the table as well. I kicked us off by liveblogging/livetweeting a popular session at the American Chemical Society’s meeting in San Francisco where drug companies reveal for the first time the chemical structures of potential new drugs being tested in clinical trials. The whole thing synced to FriendFeed as well. Folks followed the talks from all three venues, which was great. I hope I can continue doing that sort of thing in the future.

For this August, I’m co-organizing a mini-symposium at the American Chemical Society meeting in Boston about the chem/pharma blogosphere and its impact on research and communication. I’m in the process of inviting speakers right now. It’s my first time doing anything like this and part of me is petrified that no one will show up. Tips on organizing a conference session and how not to stress when doing so are welcome!

More broadly, I’d love to get more chemistry bloggers to connect with the community that attends ScienceOnline. I don’t ever want to become that old (or not-so-old) person who is clueless about them-thar newfangled whosiwhatsits that the kids are using nowadays.

What aspect of science communication and/or particular use of the Web in science interests you the most?

A few things come to mind, actually. I’d like to think that the web has made grad school a helluva lot less isolating for science grad students. You have the virtual journal clubs like Totally Synthetic, posts like SciCurious’s letter to a grad student, etc.

As a journalist the web’s capacity to equalize fascinates me. I’m extremely lucky to have a staff gig as a science writer without having gone to journalism school or landed a media fellowhip and it’s weird to think that my old blog might’ve helped my visibility. I didn’t know Ed Yong’s story until Scio10 but I think he’s a highly talented example of how the web can open doors.

The web’s equalizing power goes to readers of science content as well as writers, of course. In the ideal situation a reader can give a writer instant feedback and you can get a real conversation going, something that was much harder with the snail-paced system of letters to the editor and reader surveys. Not that the conversation is always civil. Most of C&EN’s readers have a decent amount of scientific training, but the debate that rages whenever we run an editorial about climate change is as intense as any I’ve seen.

In cases like that I don’t know that the web gives people a good representation of what the consensus is. For folks who don’t have scientific training, how do you ensure that people don’t just go to the content that already confirms their pre-existing beliefs about autism or global warming? John Timmer touched on this more eloquently in his interview with you, and I agree with him that I don’t think we have an answer yet. Though on a slightly different note, I will mention that I’ve been enjoying the New York Times’s recent attempts to recapture the spontaneity of flipping through the newspaper in online browsing, like the Times Skimmer for Google Chrome.

What are some of your favourite science blogs? Have you discovered any cool science blogs by the participants at the Conference?

In addition to the blogs I’ve already mentioned I enjoy Carbon-Based Curiosities, Wired Science, Chemistry Blog, and Terra Sigillata, to name a few of the 50 or so blogs on my feed reader.

I discovered scads of new blogs at Scio10 but I’ll focus on the one that’s become required reading for me these days: Obesity Panacea. I’d covered obesity drug development for C&EN but I’d never met Travis Saunders and Peter Janiszewski or heard of their blog until the conference.

What was the best aspect of ScienceOnline2010 for you? Is there anything that happened at this Conference – a session, something someone said or did or wrote – that will change the way you think about science communication, or something that you will take with you to your job, blog-reading and blog-writing?

Dave Mungeris my hero – his blogging 102 session was packed with practical tips that I brought back to C&EN for incorporating into our blogs, such as the use of the Disqus plugin for catching conversations on social networks, getting smart about using stats and surveys, etc. Some of that’s already happened, and some of the ideas are still in the works.

I came for the nuts-and-bolts blogging tips but I stayed for the conversations, especially the ones at the bar after the official program was done for the night. And the icing on the cake was seeing folks I’d worked with but never met, like Cameron Neylon and you, Bora, and catching up with people I hadn’t seen in months, like Jean-Claude Bradley, Aaron Rowe, Jennifer Ouellette and Nancy Shute.

It was so nice to meet you in person and thank you for the interview. I hope to see you again next January.

GSK 2256294


Figure imgf000077_0001

GSK 2256294

GSK 2256294A

CAS  1142090-23-0

MF C21H24F3N7O
MW 447.46

Antiasthmatics, soluble epoxide hydrolase inhibitor

Chronic obstructive pulmonary disease COPD …PHASE 1

(1R,3S)- Cyclohexanecarboxamide, N-[[4-cyano-2-(trifluoromethyl)phenyl]methyl]-3-[[4-methyl-6-(methylamino)-1,3,5-triazin-2-yl]amino]-,

cis-N-[[4-Cyano-2-(trifluoromethyl)phenyl]methyl]-3-[[4-methyl-6-(methylamino)-1,3,5-triazin-2-yl]amino]cyclohexanecarboxamide

(1R,3S)-N-(4-cyano-2-(trifluoromethyl)benzyl)-3-(4-methyl-6-(methylamino)-1,3,5-triazin-2-ylamino)cyclohexanecarboxamide

cis-N-((4-Cyano-2-(trifluoromethyl)phenyl)methyl)-3-((4-methyl-6-(methylamino)-1,3,5-triazin-2-yl)amino)cyclohexanecarboxamide

Cyclohexanecarboxamide, N-((4-cyano-2-(trifluoromethyl)phenyl)methyl)-3-((4-methyl-6-(methylamino)-1,3,5-triazin-2-yl)amino)-, (1R,3S)-rel-

  • Originator GlaxoSmithKline
  • Class Antiasthmatics
  • Mechanism of Action Epoxide hydrolase inhibitors

GSK 2256294 is a soluble epoxide hydrolase inhibitor in phase I clinical trials at GlaxoSmithKline for the oral treatment of patients with chronic obstructive pulmonary disease (COPD).

GSK2256294A is a potent, reversible, tight binding inhibitor of isolated recombinant human sEH (IC50 value 27 pM), and displays potent inhibition against the rat (IC50 = 61 pM) and murine (IC50 = 189 pM) orthologs of sEH. GSK2256294A also displays potent cellular inhibition (IC50 = 0.66 nM) of sEH in a cell line transfected with the human sEH enzyme.The selectivity of the compound has been demonstrated by testing against a large panel of enzymes, receptors and ion channels, including the phosphatase activity of EPHX2.

  • 01 Jan 2015GlaxoSmithKline initiates enrolment in a phase I trial in Healthy volunteers in USA (NCT02262689)
  • 09 Oct 2014GlaxoSmithKline plans a phase I trial in Healthy volunteers in USA (NCT02262689)
  • 01 May 2014GlaxoSmithKline completes a phase I pharmacokinetics trial for Chronic obstructive pulmonary disease (in the elderly, in volunteers) in USA (NCT02006537)

 

PATENT

http://www.google.com/patents/WO2009049157A1?cl=en

Step 1:

4- (bromomethyl) -3- (trifluoromethyl) benzonitrile

 

Figure CN101896065BD00313

 A mixture of 4-methyl-3- (trifluoromethyl) benzonitrile (10g, 54mmOl) was dissolved in 200mL of carbon tetrachloride, and acid imide with N- desert shot glass (10.5g, 59mmol) and peroxybenzoate (benzoyl peroxide) (1.3g, 0.54mmol) processing. The reaction mixture was heated to reflux temperature and stirred for one week. SOmL water was then added, and the layers separated. The aqueous layer with methylene chloride (2X50mL) and extracted. The organic layers were washed with water (2X50mL), dried over magnesium sulfate, and concentrated to give 4- (bromomethyl) -3- (trifluoromethyl) benzonitrile (14g, 53mm0l), as a yellow oil which was used without further purification for the subsequent steps.

Step 2:

4- (aminomethyl) -3- (trifluoromethyl) benzonitrile

 

Figure CN101896065BD00314

 4- (bromomethyl) -3- (trifluoromethyl) benzonitrile (14g) was dissolved in 500mL of 5M methanol solution of ammonia, and the mixture was stirred at room temperature for 24 hours. The solvent was removed in vacuo to give a yellow solid, which was dissolved in IM HCl and extracted with diethyl ether (3X30mL). Then, with IM NaOH and the aqueous layer was adjusted to pH 9-10 and extracted with dichloromethane (3X80mL). Thus obtained 4- (aminomethyl) -3- (trifluoromethyl) benzonitrile (4.7g, 23mm0l, 43%), as a yellow solid. MS (ES) m / e 201 [M + H] + “1H NMR (400MHz, DMS0-D6) δ 8.2 (s, 1H), 8.15 (d, 1H), 8.0 (d, 1H), 3.9 (s, 2H).

Step 1:

4-chloro -N, 6- dimethyl-1,3,5-triazin-2-amine

 

Figure CN101896065BD00411

 Intermediate 13 (500mg, 3.07mmol) was added 25-30% methylamine (300uL, 3.07mmol) in aqueous CH3CN / H20 (15mL) in a solution. The mixture was cooled to (TC, with the pH adjusted to 9_10.pH IMNaOH maintained at 9-10 for 0.5 hours. The reaction progress was monitored by LCMS, the mixture was used in the subsequent step without any treatment.

Intermediate 19

 3 – {[methyl (methyl-amino) triazin-2-yl 4 -1,3,5_ -6-] amino} cyclohexanecarboxylic acid was prepared

 

Figure CN101896065BD00303

 To 2,4-dichloro-6-methyl-1,3,5-triazine (2.291g, 13.97mmol) and methylamine (6.98ml, 13.97mmol) was added dropwise IN NaOH, to maintain the pH of 10. The reaction mixture was stirred for 30 minutes. Subsequently, a solution of 3-amino-cyclohexane – carboxylic acid (2.0g, 13.97mmol), was added dropwise to maintain a pH of 10 INNaOH. The reaction mixture was heated to 70 ° C overnight. Cooling the reaction mixture was directly purified by preparative HPLC. MS (ES +): m / e266.2 [M + H] +. 1H NMR (400MHz, DMS0-D6) δ 9.0_8.5 (bm, 2Η), 3.9 (bs, 1Η), 2.9 (m, 2Η), 2.3 (s, 3Η), 2.2 (s, 3Η), 1.9- 1.7 (bm, 4Η), 1.4-1.1 (bm, 4Η).

Step 2:

3 – {[4_-methyl-6- (methylamino) _1,3,5_ triazine _2_ yl] amino} cyclohexanecarboxylic acid

 

Figure CN101896065BD00412

 in (TC, 4-chloro -N, 6- dimethyl-1,3,5-triazin-2-amine mixture (485mg, 3.07mmol) was added 3-amino-cyclohexyl burning acid (527mg, 3.68mmol). The mixture was allowed to warm to room temperature .pH maintained between 9 to 10 for 3 hours. The mixture was concentrated and the product was purified by HPLC to afford 0.6g (2.26mmol, 74% yield) of the desired product, as a white solid .MS (ES +): m / e 266.2 [M + H] + “

 

 

Example 74

(cis)-N-{[4-cyano-2-(trifluoromethyl)phenyl]methyl}-3-{[4-methyl-6-(methylamino)-1 ,3,5- triazin-2-yl]amino}cyclohexanecarboxamide

Figure imgf000077_0001

To a solution of 3-{[4-methyl-6-(methylamino)-1 ,3,5-triazin-2- yl]amino}cyclohexanecarboxylic acid (0.100 g, 0.264 mmol) in N,N-Dimethylformamide (DMF) (4 ml) was added 4-(aminomethyl)-3-(trifluoromethyl)benzonitrile (0.053 g, 0.264 mmol) followed by diisopropylethylamine (0.101 ml, 0.580 mmol) and 1 H-1 ,2,3- benzotriazol-1-yloxy-tris(dirnethylamino)-phosphonium hexafluorophosphate (BOP reagent, 0.128 g, 0.290 mmol). The reaction was stirred at room temperature for 4 hours and then purified by preparative HPLC to provide (cis)-N-{[4-cyano-2- (trifluoromethyl)phenyl]methyl}-3-{[4-methyl-6-(methylamino)-1 ,3,5-triazin-2- yl]amino}cyclohexanecarboxamide (83 mg, 0.148 mmol, 56 %). MS (ES) m/e 448

[M+H]+. 1H NMR (400 MHz, DMSO-D6) D 7.8 (bs, 1 H), 7.3 (bs, 1 H), 7.2 (m, 1 H), 6.9 (m, 1 H), 3.8 (bs, 2H), 3.3 (bm, 1 H), 2.2 (bm, 4H), 1.8 – 1.5 (bm, 4H), 1.3 – 1.1 (bm, 4H), 0.8 – 0.5 (bm, 4H)

PATENT

http://www.google.com/patents/CN101896065B?cl=en

(cis) -N – {[4- cyano-2- (trifluoromethyl) phenyl] methyl} -3 – {[4_-methyl-6- (methylamino) _1,3, .5- triazin-2-yl] amino} cyclohexanecarboxamide

Figure CN101896065BC00051

 

Example 74

(cis) -N- {[4- cyano-2- (trifluoromethyl) phenyl] methyl} -3- {[4_ methyl _6_ (methylamino) -1,3 , 5-triazin-2-yl] amino} cyclohexanecarboxamide

 

Figure CN101896065BD00571

 To 3 – {[4_-methyl-6- (methylamino) -l, 3,5- triazin-2-yl] amino} cyclohexanecarboxylic acid (0.1OOg,

0.264mmol) in N, N- dimethylformamide (DMF) (4ml) was added 4- (aminomethyl) -3- (trifluoromethyl) benzonitrile (0.053g, 0.264mmol), followed by the addition of diisopropylethylamine (0.1Olml, 0.580mmol) and 1H-1,2,

3- benzotriazol-1-yloxy – tris (dimethylamino) _ scale hexafluorophosphate (Β0Ρ reagent, 0.128g, 0.290mmol). The reaction mixture was stirred at room temperature for 4 hours, and then purified by preparative HPLC to afford (cis) -N- {[4- cyano-2- (trifluoromethyl) phenyl] methyl} -3 – {[4_ methyl-6- (methylamino) -1,3,5_ triazin-2-yl] amino} cyclohexane carboxamide (83mg, 0.148mmol, 56%) “MS (ES) m / e 448 [ M + H] +. 1H NMR (400MHz, DMS0-D6) δ 7.8 (bs, 1H), 7.3 (bs, 1H), 7.2 (m, 1H), 6.9 (m, 1H), 3.8 (bs, 2H) , 3.3 (bm, 1H), 2.2 (bm, 4H),

1.8-1.5 (bm, 4H), 1.3-1.1 (bm, 4H), 0.8-0.5 (bm, 4H).

SMILES  Cc1nc(nc(n1)N[C@H]2CCC[C@H](C2)C(=O)NCc3ccc(cc3C(F)(F)F)C#N)NC

P.L. Podolin et al. In vitro and in vivo characterization of a novel soluble epoxide hydrolase inhibitor. Prostaglandins Other Lipid Mediat. 2013, 104-105, 25-31.
L.A. Morgan et al. Soluble epoxide hydrolase inhibition does not prevent cardiac remodeling and dysfunction after aortic constriction in rats and mice. J. Cardiovasc. Pharmacol. 2013, 61, 291-301. 

GSK 2126458, Omipalisib, PI3K/mTOR inhibitor


GSK 2126458

CAS 1086062-66-9

OMipalisib;GSK2126458;GSK-2126458;GSK2126458 (GSK458);GSK212;

2,4-Difluoro-N-[2-methoxy-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl]benzenesulfonamide;

2,4-Difluoro-N-[2-Methoxy-5-[4-(pyridazin-4-yl)quinolin-6-yl]pyridin-3-yl]benzenesulfonaMide

2,4-Difluoro-N-[2-methoxy-5-[4-(4-pyridazinyl)quinolin-6-yl]pyridin-3-yl]benzenesulfonamide

phosphoinositide 3 kinase inhibitor

idiopathic pulmonary fibrosis

PHASE 1

MW 505.49598

MF C25H17F2N5O3S

GSK…….http://www.gsk.com/media/280387/product-pipeline-2014.pdf

Omipalisib (GSK2126458): Omipalisib, also known as GSK2126458, is a small-molecule pyridylsulfonamide inhibitor of phosphatidylinositol 3-kinase (PI3K) with potential antineoplastic activity. PI3K inhibitor GSK2126458 binds to and inhibits PI3K in the PI3K/mTOR signaling pathway, which may trigger the translocation of cytosolic Bax to the mitochondrial outer membrane, increasing mitochondrial membrane permeability and inducing apoptotic cell death. Bax is a member of the proapoptotic Bcl2 family of proteins. PI3K, often overexpressed in cancer cells, plays a crucial role in tumor cell regulation and survival.

GlaxoSmithKline (GSK) is developing omipalisib (GSK-2126458), a phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) inhibitor as well as mTOR complex 1 and 2 inhibitor, for the potential oral treatment of cancer and idiopathic pulmonary fibrosis

MEDKOO

Certificate of Analysis:

View current batch of CoA

QC data:

View NMR, View HPLC, View MS

GSK2126458 is a highly potent PI3K and mTOR inhibitor. In vivo, GSK2126458 showed anti-tumor activity in both pharmacodynamic and tumor growth efficacy models. GSK2126458 reduced the phosphorylated AKT, p70S6K contents in a dose and time dependent way. The IC50 of GSK2126458 is 2 nM for pAKT in the HCC1954 breast carcinoma cell line. In various human tumor cells, GSK2126458 had a width of inhibitory activity for potent cell growth and induced cell death. Notably, GSK2126458 acted mainly by not induction of apoptosis but cell cycle arrest, particularly in G1-phase

GlaxoSmithKline (GSK) is developing omipalisib (GSK-2126458), a phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) inhibitor as well as mTOR complex 1 and 2 inhibitor, for the potential oral treatment of cancer and idiopathic pulmonary fibrosis

GSK-2126458 is a phosphatidylinositol 3-Kinase (PI3K) inhibitor in early clinical development for the oral treatment of solid tumors and for the oral treatment of lymphoma. Early clinical studies are ongoing for the treatment of idiopathic pulmonary fibrosis. The compound is being developed b GlaxoSmithKline.

In August 2009, a phase I trial began for solid tumors and lymphoma . In April 2012, phase Ib co-clinical trials in advanced prostate cancer (PC) were underway . In March 2013, a phase I trial was initiated in the UK in patients with idiopathic pulmonary fibrosis

In April 2014, a phase I, open-label, multicenter, dose-escalation study (study number P3K113794) and safety data were presented at the 105th AACR meeting in San Diego, CA. Advanced solid tumor patients (n = 69) received oral continuous GSK-2126458 or intermittent GSK-2126458 bid  + trametinib. For GSK-2126458 and trametinib, the MTD in QD cohort was 2 and 1 mg, respectively, and also 1 and 1.5 mg, respectively

PAPER 

Discovery of GSK2126458, a highly potent inhibitor of PI3K and the mammalian target of rampamycin
ACS Med Chem Lett 2010, 1(1): 39

 

Abstract Image

Phosphoinositide 3-kinase α (PI3Kα) is a critical regulator of cell growth and transformation, and its signaling pathway is the most commonly mutated pathway in human cancers. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of PI3K/AKT pathway inhibition. 2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide (GSK2126458, 1) has been identified as a highly potent, orally bioavailable inhibitor of PI3Kα and mTOR with in vivo activity in both pharmacodynamic and tumor growth efficacy models. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.

 ……………….. 

synthesis

omalipisib

 

Figure imgf000151_0002

Figure imgf000145_0002

………………..

PATENT

WO 2008144463

http://www.google.co.in/patents/WO2008144463A1?cl=en

Example 345

2,4-difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3- pyridinyl } benzenesulf onamide

a) 6-bromo-4-(4-pyridazinyl)quinoline

Dissolved 6-bromo-4-iodoquinoline (17.43 g, 52.2 mmol), 4- (tributylstannanyl)pyridazine (19.27 g, 52.2 mmol), and PdC12(dppf)-CH2C12 (2.132 g, 2.61 mmol) in 1,4-dioxane (200 mL) and heated to 105 °C. After 3 h, added more palladium catalyst and heated for 6 h. Concentrated and dissolved in methylene chloride/methanol. Purified by column chromatography (combiflash) with 2% MeOH/EtOAc to 5% MeOH/EtOAc to give the crude title compound. Trituration with EtOAc furnished 6-bromo-4-(4-pyridazinyl)quinoline (5.8 g, 20.27 mmol, 38.8 % yield). MS(ES)+ m/e 285.9, 287.9 [M+H]+.

b) 2,4-difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3- pyridinyl } benzenesulf onamide A slurry of 6-bromo-4-(4-pyridazinyl)quinoline (4.8 g, 16.78 mmol), bis(pinacolato)diboron (4.69 g, 18.45 mmol) , PdC12(dppf)-CH2C12 (530 mg, 0.649 mmol) and potassium acetate (3.29 g, 33.6 mmol) in anhydrous 1,4-dioxane (120 ml) was heated at 100 °C for 3 h. The complete disappearance of the starting bromide was observed by LCMS. The reaction was then treated with N-[5-bromo-2- (methyloxy)-3-pyridinyl]-2,4-difluorobenzenesulfonamide (6.68 g, 17.61 mmol) and another portion of PdC12(dppf)-CH2C12 (550 mg, 0.673 mmol), then heated at 110 °C for 16 h. The reaction was allowed to cool to room temperature, filtered, and concentrated. Purification of the residue by chromatography (Analogix; 5% MeOH / 5% CH2C12 / 90% EtOAC) gave 6.5 g (76%) desired product. MS(ES)+ m/e 505.9 [M+H]+.

 

INTERMEDIATES:

Intermediate 1  Similar but not same

Scheme A:

Conditions: a) Tributyl(vinyl)tin, Pd(PPh3)4, dioxane, reflux; b) OsO4, NaIO4, 2,6- lutidine, r-BuOH, dioxane, H2O, rt; c) (4-pyridyl)boronic acid, Pd(PPh3)4, 2 M K2CO35 DMF, 100 DC.

4-(4-pyridinyl)-6-quinolinecarbaldehydeSimilar but not same

a) 4-chloro-6-ethenylquinoline

A mixture of 6-bromo-4-chloroquinoline (6.52 g, 26.88 mmol; see J. Med. Chem., H 268 (1978) ), tributyl(vinyl)tin (8.95 g, 28.22 mmol), and tetrakistriphenylphospbine palladium (0) (0.62 g, 0.54 mmol) in 1,4-dioxane (150 mL) was refluxed for 2.0 h, cooled to room temperature, and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (0-4% MeOH:CH2Cl2) to give the title compound (5.1 g) as a pale yellow solid. MS (ES)+ m/e 190 [M+H]+. This material was used directly in the next step.

b) 4-chloro-6-quinolinecarbaldehyde

A mixture of 4-chloro-6-ethenylquinoline (5.1 g, 26.88 mmol), 2,6-lutidine

(5.76 g, 53.75 mmol), sodium (meta) periodate (22.99 g, 107.51 mmol), and osmium tetroxide (5.48 g of a 2.5% solution in tert-butanol, 0.538 mmol) in l,4-dioxane:H2θ (350 mL of 3: 1 mixture) was stirred for 3.5 h at room temperature and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (CH2Cb) to give the title compound (4.26 g, 83% for 2 steps) as a pale yellow solid. MS (ES)+ m/e 192 [M+H]+.

c) 4-(4-pyridmyl)-6-qumolinecarbaldehyde

A mixture of 4-chloro-6-quinolinecarbaldehyde (3.24 g, 16.92 mmol), A- pyridylboronic acid (3.12 g, 25.38 mmol), tetrakistriphenylphosphine palladium (0) (0.978 g, 0.846 mmol), and 2M aqueous K2CO3 (7.02 g, 50.76 mmol, 25.4 mis of 2M solution) in DMF (100 mL) was heated at 100 °C for 3.0 h and cooled to room temperature. The mixture was filtered through Celite and the Celite was washed with EtOAc. The filtrate was transferred to a separatory funnel, washed with water and saturated NaCl, dried (Na2SO4), filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (5% MeOH:CH2Cl2) to give the title compound (2.03 g, 51%) as a tan solid. MS (ES)+ m/e 235 [M+H]+.

Intermediate 2

Preparation of 2-amino-5 -bromo-N,N-dimethyl-3 -pyridinesulfonamideSimilar but not same

a) 2-ammo-5-bromo-3-pyridinesulfonyl chloride

To a cooled (0 °C) solution of chlorosulfonic acid (58 mL) under vigorous stirring was added 5-bromo-2-pyridinamine (86.7 mmol) portionwise. The reaction mixture was then heated at reflux for 3 hrs. Upon cooling to room temperature, the reaction mixture was poured over ice (-100 g) with vigorous stirring. The resulting yellow precipitate was collected by suction filtration, washing with cold water and petroleum ether to provide the title compound as an orange-yellow solid (18.1 g, 77% yield). MS(ES)+ m/e 272.8 [M+H]+.

* Other sulfonyl chlorides can be prepared using this procedure by varying the choice of substituted aryl or heteroaryl.

b) 2-amino-5-bromo-N,N-dimethyl-3-pyridinesulfonamide

To a cold (0 DC) suspension of 2-amino-5-bromo-3-pyridinesulfonyl chloride (92.1 mmol) in dry 1,4-dioxane (92 mL) was added pyridine (101.3 mmol) followed by a 2M solution of dimethylamine in THF (101.3 mmol). The reaction was allowed to warm to rt for 2 h, heated to 50 DC for 1 h, then cooled to rt. After standing for 2 h, the precipitate was collected by filtration and rinsed with a minimal amount of cold water. Drying the precipitate to constant weight under high vacuum provided 14.1 g (55%) of the title compound as a white solid. MS(ES)+ m/e 279.8, 282.0 [M+H]+.

 

Intermediate 3

Preparation of 2-amino-N,N-dimethyl-5-(4,4,5,5-tetramethyl-l,3.2-dioxaborolan-2- yl)-3 -pyridinesulfonamideSimilar but not same

c) To a solution of 2-amino-5-bromo-N,N-dimethyl-3 -pyridinesulfonamide (7.14 mmol) in 1,4-dioxane (35 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi-l,3,2- dioxaborolane (7.86 mmol), potassium acetate (28.56 mmol) and [1,1 ‘- bis(diphenylphosphmo)-ferrocene] dichloropalladium(II) dichloromethane complex (1 :1) (0.571 mmol). The reaction mixture was stirred at 100 °C for 18 h. The reaction was concentrated in vacuo, re-dissolved in ethyl acetate (50 mL) and purified on silica using 60% ethyl acetate/hexanes to yield the title compound as a tan solid (86 %). IH ΝMR (400 MHz, DMSOd6) δ ppm 8.41 (d, 1 H, J =1.52), 7.92 (d, 1 H, J = 1.77), 2.68 (s, 6 H), 1.28 (s, 12 H).

* Other boronate or boronic acids can be prepared using this procedure by varying the choice of aryl or heteroaryl bromide. Scheme 17:

Conditions: a) NaO(Rl), (Rl)OH, O 0C to room temperature; b) SnCl2-2H2O, ethyl acetate, reflux; c) (R2)SO2C1, pyridine, O 0C to room temperature.

Intermediate 4

Preparation of N-r5-bromo-2-(methyloxy)-3-pyridinyll-2,4- difluorobenzenesulfonamide

Figure imgf000151_0002N-[5-bromo-2-(methyloxy)-3-pyridinyl]-2,4- difluorobenzenesulfonamide

a) 5-bromo-2-(methyloxy)-3-nitropyridine

To a cooled (0 °C) solution of 5-bromo-2-chloro-3-nitropyridine (50 g, 211 mmol) in methanol (200 mL) was added dropwise over 10 minutes 20% sodium methoxide (50 mL, 211 mmol) solution. The reaction, which quickly became heterogeneous, was allowed to warm to ambient temperature and stirred for 16 h. The reaction was filtered and the precipitate diluted with water (200 mL) and stirred for 1 h. The solids were filtered, washed with water (3 x 100 mL) and dried in a vac oven (40 °C) to give 5-bromo-2-(methyloxy)-3-nitropyridine (36 g, 154 mmol, 73.4 % yield) as a pale yellow powder. The original filtrate was concentrated in vacuo and diluted with water (150 mL). Saturated ammonium chloride (25 mL) was added and the mixture stirred for 1 h. The solids were filtered, washed with water, and dried in a vac oven (40 °C) to give a second crop of 5-bromo-2-(methyloxy)-3- nitropyridine (9 g, 38.6 mmol, 18.34 % yield). Total yield = 90%. MS(ES)+ m/e 232.8, 234.7 [M+H]+.

b) 5-bromo-2-(methyloxy)-3-pyridinamine

To a solution of 5-bromo-2-(methyloxy)-3-nitropyridine (45 g, 193 mmol) in ethyl acetate (1 L) was added tin(II) chloride dihydrate (174 g, 772 mmol). The reaction mixture was heated at reflux for 4 h. LC/MS indicated some starting material remained, so added 20 mol% tin (II) chloride dihydrate and continued to heat at reflux. After 2 h, the reaction was allowed to cool to ambient temperature and concentrated in vacuo. The residue was treated with 2 N sodium hydroxide and the mixture stirred for 1 h. The mixture was then with methylene chloride (1 L), filtered through Celite, and washed with methylene chloride (500 mL). The layers were separated and the organics dried over magnesium sulfate and concentrated to give 5-bromo-2-(methyloxy)-3-pyridinamine (23 g, 113 mmol, 58.7 % yield). The product was used crude in subsequent reactions. MS(ES)+ m/e 201.9, 203.9 [M+H]+.

c) N-[5-bromo-2-(methyloxy)-3-pyridinyl]-2,4-difluorobenzenesulfonamide

Figure imgf000151_0002

To a cooled (0 °C) solution of 5-bromo-2-(methyloxy)-3-pyridinamine (20.3 g, 100 mmol) in pyridine (200 mL) was added slowly 2,4-difluorobenzenesulfonyl chloride (21.3 g, 100 mmol) over 15 min (reaction became heterogeneous). The ice bath was removed and the reaction was stirred at ambient temperature for 16 h, at which time the reaction was diluted with water (500 mL) and the solids filtered off and washed with copious amounts of water. The precipitate was dried in a vacuum oven at 50 °C to give N-[5-bromo-2-(methyloxy)-3-pyridinyl]-2,4- difluorobenzenesulfonamide (12 g, 31.6 mmol, 31.7 % yield) MS(ES)+ m/e 379.0, 380.9 [M+H]+.

 

 

References

1. Knight et al., ACS Med. Chem. Lett. 2010, 1, 39-43.
2. Hardwick et al., Mol. Cancer Ther. 2009, 8(12), Supplement I, Abstract C63.
3. Greger et al., Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol. Cancer Ther. 2012, 11(4), 909-920.

1: Zhang Y, Xue D, Wang X, Lu M, Gao B, Qiao X. Screening of kinase inhibitors targeting BRAF for regulating autophagy based on kinase pathways. Mol Med Rep. 2014 Jan;9(1):83-90. doi: 10.3892/mmr.2013.1781. Epub 2013 Nov 7. PubMed PMID: 24213221.

2: Villanueva J, Infante JR, Krepler C, Reyes-Uribe P, Samanta M, Chen HY, Li B, Swoboda RK, Wilson M, Vultur A, Fukunaba-Kalabis M, Wubbenhorst B, Chen TY, Liu Q, Sproesser K, DeMarini DJ, Gilmer TM, Martin AM, Marmorstein R, Schultz DC, Speicher DW, Karakousis GC, Xu W, Amaravadi RK, Xu X, Schuchter LM, Herlyn M, Nathanson KL. Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma. Cell Rep. 2013 Sep 26;4(6):1090-9. doi: 10.1016/j.celrep.2013.08.023. Epub 2013 Sep 19. PubMed PMID: 24055054; PubMed Central PMCID: PMC3956616.

3: Kim HG, Tan L, Weisberg EL, Liu F, Canning P, Choi HG, Ezell SA, Wu H, Zhao Z, Wang J, Mandinova A, Griffin JD, Bullock AN, Liu Q, Lee SW, Gray NS. Discovery of a potent and selective DDR1 receptor tyrosine kinase inhibitor. ACS Chem Biol. 2013 Oct 18;8(10):2145-50. doi: 10.1021/cb400430t. Epub 2013 Aug 13. PubMed PMID: 23899692; PubMed Central PMCID: PMC3800496.

4: Khalili JS, Yu X, Wang J, Hayes BC, Davies MA, Lizee G, Esmaeli B, Woodman SE. Combination small molecule MEK and PI3K inhibition enhances uveal melanoma cell death in a mutant GNAQ- and GNA11-dependent manner. Clin Cancer Res. 2012 Aug 15;18(16):4345-55. doi: 10.1158/1078-0432.CCR-11-3227. Epub 2012 Jun 25. PubMed PMID: 22733540; PubMed Central PMCID: PMC3935730.

5: Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, Dickerson SH, Laquerre SG, Liu L, Gilmer TM. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol Cancer Ther. 2012 Apr;11(4):909-20. doi: 10.1158/1535-7163.MCT-11-0989. Epub 2012 Mar 2. PubMed PMID: 22389471.

6: Wang M, Gao M, Miller KD, Sledge GW, Zheng QH. [11C]GSK2126458 and [18F]GSK2126458, the first radiosynthesis of new potential PET agents for imaging of PI3K and mTOR in cancers. Bioorg Med Chem Lett. 2012 Feb 15;22(4):1569-74. doi: 10.1016/j.bmcl.2011.12.136. Epub 2012 Jan 10. PubMed PMID: 22297110.

7: Schenone S, Brullo C, Musumeci F, Radi M, Botta M. ATP-competitive inhibitors of mTOR: an update. Curr Med Chem. 2011;18(20):2995-3014. Review. PubMed PMID: 21651476.

8: Leung E, Kim JE, Rewcastle GW, Finlay GJ, Baguley BC. Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells. Cancer Biol Ther. 2011 Jun 1;11(11):938-46. Epub 2011 Jun 1. PubMed PMID: 21464613; PubMed Central PMCID: PMC3127046.