New Drug Approvals

Home » 2014 » April (Page 2)

Monthly Archives: April 2014

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

Blog Stats

  • 4,804,028 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

GKT-137831 a NOX1 and NOX4 inhibitor from GenKyoTex being developed for diabetic nephropathy


2D chemical structure of 1218942-37-0

 

 

GTK 137831

1218942-37-0

Genkyotex Sa INNOVATOR

1H-​Pyrazolo[4,​3-​c]​pyridine-​3,​6(2H,​5H)​-​dione, 2-​(2-​chlorophenyl)​-​4-​[3-​(dimethylamino)​phenyl]​-​5-​methyl-

 

C21 H19 Cl N4 O2

  • 2-(2-Chlorophenyl)-4-(3-(dimethylamino)phenyl)-5-methyl-1H-pyrazolo(4,3-c)pyridine-3,6(2H,5H)-dione
  • 394.8601 mw
  • in phase 2
  • UNII-45II35329V

drug recently advancing to phase II trials is GKT-137831, a NOX1 and NOX4 inhibitor from GenKyoTex being developed for diabetic nephropathy, the leading cause of chronic kidney disease in the US and Europe.

 

GKT137831 is a selective NOX1/4 inhibitor in Phase II clinical development for the treatment of diabetic nephropathy, one of the complications of diabetes. It is a potent, NOX specific, small molecule with good oral availability.

Data from the Phase 1 programme to assess safety and exposure to single and multiple oral doses of GKT137831 was presented at the ASN Kidney week in San Diego in 2012. More than 100 subjects have been exposed to GKT137831 and the drug was well tolerated with no serious adverse events. In summer 2013, the FDA approved the IND to allow commencement of the Ph2 PoC trial of GKT137831 in diabetic nephropathy. Subsequently, approvals have been received from the competent authorities in Australia, Canada, Germany, Czech Republic and Poland. Enrollment to this study is ongoing and data is expected in H1 2015.

GKT137831 has been found to be effective in a range of preclinical disease models. This work has been conducted by leading academic collaborators in disease models of diabetic nephropathy, atherosclerosis, idiopathic pulmonary fibrosis, liver fibrosis and angiogenesis. GKT137831 has therefore, the potential to treat a wide range of important and poorly managed diseases

PATENT

WO 2010035221

http://www.google.com/patents/WO2010035221A1?cl=en

Scheme 1

Figure imgf000055_0001

R18 = Me Pr, iPr, Bu

G /NH Toluen

Il

Figure imgf000055_0002

G1 as described above G1 = H (Ib) (Ia) VIII

 

Scheme 2

Figure imgf000057_0001

R18 = Me, Et, Pr, iPr, Bu

Toluene

G^

Figure imgf000057_0002

G1 as described above G1 = H (Ib) (Ia) VIII

Scheme 3

 

Figure imgf000059_0001

IV R19 = Me, Et, XII

R18 = Me, Et, Pr, iPr, Bu

 

Figure imgf000059_0003
Figure imgf000059_0002

G1 = H, G3 = CH2NR20R21 (Ia) XIV XIII

 

Figure imgf000059_0004

G1, G3 as described above (Ib)

Figure imgf000105_0001

Genkyotex’s GKT137831 Found to Reverse Fibrosis and Improve Survival in a Model of Persistent Lung Fibrosis

Genkyotex, the leading developer of selective NOX enzyme inhibitors, announced today the publication of data showing that GKT137831, a first in class NOX1 and 4 inhibitor, was able to reverse lung fibrosis associated with aging in a new model of idiopathic pulmonary fibrosis. Collaborators led by Professor Victor Thannickal at the University of Alabama at Birmingham published the results in the April 9, 2014 issue of Science Translational Medecine. Genkyotex is investigating GKT137831 in a Phase II trial in patients with diabetic nephropathy, another progressive fibrotic disease.

23 MARCH 2014

Keystone Symposia Conference 2014

March 26th, 2014. Today, Dr. Philippe Wiesel, CMO at Genkoytex presented preclinical data showing the beneficial effect of NOX1/4 inhibitor for the treatment of NASH (Non-Alcoholic Steatohepatitis)

Genkyotex held a breakfast meeting on the 28th on the role of NADPH oxidases in fibrosis

The presentations can be downloaded here

11 NOVEMBER 2013

Genkyotex NOX Inhibitor GKT137831 Successfully Shown to Halt Diabetic Kidney Disease

Genkyotex, the leading developer of selective NOX enzyme inhibitors, announced today that data from a group of academic collaborators demonstrated that NOX4 is an important driver of kidney injury in diabetes and that its novel, first in class NOX 1 and 4 inhibitor, GKT137831, has the potential to prevent or delay the development of diabetic nephropathy. Data were presented at the American Society of Nephrology’s Kidney Week 2013 in Atlanta and have been accepted for publication in the Journal of the American Society of Nephrology (JASN).

08 NOVEMBER 2013

Genkyotex attended the American Society of Nephrology Annual Meeting during Kidney week in Atlanta GA.

November 7th to 10th, 2013. Genkyotex attended the American Society of Nephrology Annual Meeting during Kidney week in Atlanta, GA. Ursula Ney, CEO, Philippe Wiesel, CMO, and the clinical team attended. Presentations from the Ancillary meeting held on 8th November can be found here.

05 NOVEMBER 2013

Genkyotex Initiates Multinational Phase II Study with First in Class NOX Inhibitor GKT137831 in Diabetic Nephropathy Patients

Genkyotex, the leading developer of selective NOX enzyme inhibitors, announced today the initiation of a multinational Phase II clinical study of GKT137831 in patients with diabetic nephropathy. GKT137831 is a first in class inhibitor targeting NOX1 and NOX4 enzymes, both of which play a key role in the development of diabetic complications and chronic kidney disease in particular. In phase I studies in more than 100 subjects, GKT137831 was found to be safe and well tolerated when administered orally once and twice daily.

21 OCTOBER 2013

Genkyotex Collaborators Elucidate Role of NOX4 in Osteoporosis

Genkyotex, the leading developer of NOX enzyme inhibitors, announced today that a group of collaborators have discovered a link between the enzyme NOX4 and development of osteoporosis. These results, published online in the Journal of Clinical Investigationdoi:10.1172/JCI67603), indicate that inhibitors of NOX4, such as GKT137831 developed by Genkyotex could lead to a novel way of treating patients with osteoporosis. GKT137831, the first in class NOX1 and 4 inhibitor, has shown favorable safety and pharmacokinetic profiles in Phase I studies, and following a recently FDA approved IND will enter a Phase II trial in patients with diabetic nephropathy.

08 SEPTEMBER 2013

Genkyotex Receives FDA IND Approval for Phase II Clinical Study with First in Class NOX Inhibitor GKT137831

Genkyotex, the leading developer of NOX enzyme inhibitors, announced today that the U.S. Food and Drug Administration has approved the company’s Investigational New Drug (IND) application to begin a Phase II clinical study of GKT137831 in patients with diabetic nephropathy. GKT137831 is a first in class inhibitor targeting NOX1 and NOX4 enzymes. Enrollment of patients into the multinational Phase II study is expected to begin during Q4, 2013.

07 MAY 2013

Genkyotex Collaborators Discover Role of NOX in Development of Atherosclerosis in Diabetic Mice

Genkyotex, the leading developer of NOX inhibitors to treat oxygen-radical mediated diseases, announced today that its collaborators at the Baker IDI Heart & Diabetes Research Institute, Melbourne (Australia) and Maastricht University (The Netherlands) have elucidated the role of NOX1 in causing atherosclerosis in diabetic mice. The researchers found that NOX1 produces toxic amounts of oxygen radicals in the wall of blood vessels, which along with other inflammatory chemicals led to atherosclerotic plaque development. The researchers also demonstrated that Genkyotex’s selective NOX1 and 4 inhibitor, GKT137831, was able to dramatically reduce development of atherosclerosis. The research and accompanying editorial from Dr. David G. Harrison from Vanderbilt University was published in May 7th issue ofCirculation.

17 DECEMBER 2012

Genkyotex Issued U.S. Patent Covering Parent NOX Inhibitor Chemical Series

Genkyotex, the leading developer of NOX inhibitors to treat oxygen-radical mediated diseases, today announced that the United States Patent and Trademark Office (USPTO) has issued a Notice of Allowance for U.S. Patent Application No. 12/532,336, titled “pyrazolo pyridine derivatives as NADPH oxidase inhibitors”.

02 NOVEMBER 2012

Genkyotex’s NOX Inhibitor GKT137831 Phase I Data Presented at Kidney Week 2012

Genkyotex, the leading developer of NOX inhibitors to treat oxygen-radical mediated diseases, announced today that Phase I studies have demonstrated excellent safety and tolerability following single and multiple oral doses of GKT137831, the first in class NOX 1 and 4 inhibitor. In addition, GKT137831 demonstrated a favourable pharmacokinetic profile in these subjects.

15 OCTOBER 2012

Genkyotex’s First in Class NOX Inhibitor GKT137831 to be Presented at Kidney Week

Genkyotex will present data from single and multiple dose Phase I studies with the NOX 1 and 4 inhibitor, GKT137831, at Kidney Week 2012 (San Diego, October 30 – November 4). The Phase I data will be presented on Friday, November 2, 2012, 10.00 AM -12.00 PM (PosterBoard# FR-PO831; Abstract# 2279).

08 AUGUST 2012

Genkyotex’s Lead NOX Inhibitor GKT137831 Demonstrates Activity in Models of Liver Fibrosis

Genkyotex, with collaborator Professor David Brenner, M.D., Dean, School of Medicine, University of California San Diego, has published data online in Hepatology regarding its lead (NOX) inhibitor, GKT137831, in models of liver fibrosis, a scarring process associated with chronic liver disease that can lead to loss of liver function. The data demonstrates the specificity of GKT137831 and its ability to attenuate development of fibrosis in the liver and production of reactive oxygen species (ROS) in two models of disease, as well as inhibiting messenger RNA expression of fibrotic and NOX genes.

09 JULY 2012

Genkyotex closes CHF25 million (USD26 million) extension to its Series C financing.

Investors in the Series C round, including Eclosion, Edmond de Rothschild Investment Partners, Vesalius Biocapital Partners, MP Healthcare Venture, all participated in the financing extension. The proceeds will be used to advance clinical development of Genkyotex’s lead compound, the NOX1/4 inhibitor GKT137831, through Phase II development for the treatment of diabetic nephropathy.

22 JUNE 2012

Genkyotex Announces Successful Phase Ia Data with First in Class NOX Inhibitor GKT137831

Diabetic Nephropathy First Target Indication for NOX1/4 Inhibitor

31 OCTOBER 2011

GenKyoTex Starts Phase I Trial with First in Class NOX inhibitor GKT137831

GenKyoTex, the leading developer of NOX inhibitors to treat oxygen-radical mediated diseases, announced today that a Phase I study has been initiated with GKT137831, a first in class dual inhibitor of NOX1 and NOX4 enzymes.

GenKyoTex raises CHF 18 million in a Series C Venture Financing to develop NOX enzyme inhibitors.

Appoints New Management Team & Board

02 DECEMBER 2010

GKT137831 granted Orphan Drug status for Idiopathic Pulmonary Fibrosis by the EC (EMEA)

Genkyotex announced today that its lead clinical candidate GKT137831 has been granted the orphan drug status by the European Commission for the treatment of idiopathic pulmonoary fibrosis.

27 SEPTEMBER 2010

FDA granting Genkyotex Orphan Drug Designation of GKT137831 for IPF

Genkyotex announced today having received a letter from FDA dated of 21st September 2010, granting Genkyotex Orphan Drug Designation of GKT137831 for the treatment of Idiopathic Pulmonary Fibrosis (IPF).

Medical Mushrooms – The Future of Cancer Treatment?


Lyranara.me's avatarLyra Nara Blog

Cancer rates are on the rise worldwide, which means that in coming generations more and more people will have their lives turned inside out with a diagnosis, and with having to turn their attention to battling this new plague. The psychological effects of having your world turned on its so quickly can be devastating, and often put people in a depressed, anxious and negative emotional state.

With so many types of cancers affecting people these days, there is no such thing as a single cure for cancer, because each type is different and will respond to different remedies. Finding the miracle cure often requires an intense search, deviation from standard doctor’s recommendations, a huge investment of time and money, and tremendous amount of hope, belief and faith. Not everything works for every cancer, but, some things consistently aid in the struggle with all cancers, like the right diet…

View original post 1,125 more words

Binimetinib in phase 3 for for the treatment of metastatic or unresectable cutaneous melanoma with NRAS mutations and in combination with LGX-818 in adult patients with BRAF V600


Figure imgf000024_0001

 

 

Binimetinib

5-[(4-bromo-2-fluorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-1-methyl-1H-benzimidazole-6-carboxamide
5-(4-Bromo-2-fluorophenylamino)-4-fluoro-1-methyl-1H-benzimidazole-6-carbohydroxamic acid 2-hydroxyethyl ester
6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide
606143-89-9  CAS
tyrosine kinase inhibitor, antineoplastic

Array BioPharma Inc;PHASE 3 Cancer, ovary (serous)

Novartis PHASE 3 Melanoma

AGARRY-162
ARRY-438162
MEK-162

 

MEK-1 protein kinase inhibitor; MEK-2 protein kinase inhibitor

Liver injury; Melanoma; Noonan syndrome; Ovary tumor; Solid tumor

Growth factor-mediated proliferative signals are transmitted from the extracellular environment to the nucleus through several pathways, including the RAS/RAF/ MEK pathway. The RAS/RAF/MEK kinase signal transduction pathway is activated through initial extracellular binding and stimulation of tyrosine receptor kinases (RTKs) by their respective cognate ligands. Upon autophosphorylation of specific tyrosine residues in the cytosolic domain of RTKs, the Grb2-Sos complex translocates to the plasma membrane, and converts the inactive RAS’GDP to active RAS’GTP. The interaction between the Grb2 docking protein and the activated kinases or the phosphorylated receptor associated proteins is mediated by the Src Homology (SH2) domain of the signaling protein that recognizes specific phosphotyrosine sequences. RAS undergoes a conformational change upon guanosine 5 ‘-triphosphate (GTP) binding and causes the recruitment of RAF- 1 to the cytoplasmic membrane where it is phosphorylated by several kinases and simultaneous disphosphorylated at key residues by protein phosphatase-2B. Activated RAF phosphorylates the mitogen- activated protein kinase kinase (MEK) on two serine residues in the activation loop, which results in the activation of this protein kinase. MEK then phosphorylates and activates extracellular signal-regulated kinase (ERK), allowing its translocation to the nucleus where it phosphorylates transcriptional factors permitting the expression of a variety of genes.

The RAS/RAF/MEK signal transduction pathway is deregulated, often through mutations that result in ectopic protein activation, in roughly 1/3 of human cancers. This deregulation in turn results in a wide array of cellular changes that are integral to the etiology and maintenance of a cancerous phenotype including, but not limited to, the promotion of proliferation and evasion of apoptosis (Dhillon et al., Oncogene, 2007, 26: 3279-3290).

Accordingly, the development of small molecule inhibitors of key members of the RAS/ RAF/ MEK signal transduction pathway has been the subject of intense effort within the pharmaceutical industry and oncology community.

MEK is a major protein in the RAS/ RAF/ MEK pathway, which signals toward cell proliferation and survival, and frequently activated in tumors that have mutations in the RAS or RAF oncogenes or in growth receptor tyrosine kinases. MEK is a key player in the RAS/RAF/MEK pathway as it is downstream of RAS and RAF. Despite being only rarely mutated in cancer (Murugan et al., Cell Cycle, 2009, 8: 2122-2124; Sasaki et al., J. Thorac. Oncol., 2010, 5: 597-600), inhibitors of the MEK1 and MEK2 proteins have also been targeted for small molecule inhibition owing to their central position within the RAS/ RAF/ MEK signal transduction pathway signaling cascade (Fremin and Meloche, J. Hematol.

Oncol., 2010, 3:8). Recently a potent MEK inhibitor failed to demonstrate efficacy in clinical trials in patients with advanced non-small cell lung cancer (Haura et al., Clin. Cancer Res., 2010, 16: 2450-2457). The reason for failure in this trial is not clear.

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (hereinafter, “Compound A”) is a benzimidazole compound that is a known potent and selective inhibitor of the MEK1 and MEK2 proteins, and useful in the treatment of hyperproliferative diseases, particularly cancer, in mammals. For example, in a recently published Phase I study of 28 patients suffering from unresectable, locally advanced or metastatic biliary cancer and who had received < 1 prior systemic therapy, oral Compound A treatment (60 mg twice daily) resulted in 1 complete regression, 1 partial regression and 11 stable disease diagnoses after at least 6 weeks of treatment (Finn et al., J. Clin. Oncol. 30, 2012 (Supplement 4, 2012 Gastrointestinal Cancers Symposium, Abstract No. 220). Compound A has also been demonstrated to be effective in the treatment of patients with either BRAFV600 or NRAS-mutant melanoma (Ascierto et al., J. Clin. Oncol. 30, 2012 (Supplement, 2012 ASCO Annual Meeting, Abstract No. 8511).

The compound, as well as a process for its preparation, is disclosed in PCT Pub. No. WO 03/077914

 

MEK-162, a potent, orally active MEK1/2 inhibitor, is in phase III clinical trials at Array BioPharma and licensee Novartis for the treatment of metastatic or unresectable cutaneous melanoma with NRAS mutations and in combination with LGX-818 in adult patients with BRAF V600. Phase III studies are also under way at Array BioPharma for the treatment of low grade serous carcinomas of the ovary, fallopian tube or primary peritoneum following at least one prior platinum-based chemotherapy regimen and no more than three lines of prior chemotherapy regimens. Novartis and Array BioPharma are also conducting phase II clinical studies for the treatment of locally advanced and unresectable or metastatic malignant cutaneous melanoma, harboring BRAFV600E mutations; in BRAF mutated melanoma in combination with AMG-479 and for the treatment of Noonan’s syndrome, and in non-small cell lung cancer harboring KRAS or EGFR mutation and in combination with erlotinib. MEK-162 is being evaluated in phase I/II as first line treatment of advanced biliary tract carcinoma and for the treatment of adult patients with mutant or wild-type RAS metastatic colorectal cancer. The product is in early clinical trials at Array Biopharma for the treatment of biliary cancer.

According to Array, MEK-162 may also provide broad therapeutic benefits in the treatment of chronic degenerative diseases. However, a phase II trial for the treatment of stable rheumatoid arthritis (RA) did not meet its primary endpoint. Based on these data, the company focused development of MEK-162 solely in oncology.

In 2010, MEK-162 was licensed to Novartis by Array BioPharma for worldwide development. In 2013, orphan drug designation was assigned in Japan for the treatment of malignant melanoma with NRAS or BRAF V600 mutation.

WO-2014063024 DEALS WITH Preparation, crystalline forms, and formulations comprising binimetinib. Binimetinib is a MEK-1/2 inhibitor originally claimed in WO03077914, which Array and Novartis are developing for the treatment of cancer, including melanoma, low-grade serous ovarian cancer, and other solid tumors, as well as Noonan syndrome hypertrophic cardiomyopathy and hepatic impairment. See also WO2014018725 for the most recent filing on the agent

 

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

WO 03/077914

http://www.google.com/patents/WO2003077914A1?cl=en

 

Schemes 1-4.

Scheme 1

 

Scheme la

Scheme 2

Scheme 3

 

17 18

Scheme 4

25

Scheme 5

General synthetic methods which may be referred to for preparing some of the compounds of the present invention are provided in PCT published application number WO 00/42022 (published July 20, 2000). The foregoing patent application is incorporated herein by reference in its entirety.

 similar ie chloro instead of fluoro

Example 52

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide (lOcc) Step A: 3-Chloro-2,4-difluoro-5-nitro-benzoic acid 2a

3-Chloro-2,4-difluoro-benzoic acid la (3.00 g, 15.6 mmol) is added to a stirred solution of concentrated H2SO4 (16 mL) and fuming nitric acid (0.85 mL, 20.3 mmol). After 3 hours a precipitate forms. The yellow slurry is poured onto ice water (100 mL). The aqueous mixture is extracted with diethyl ether (3x). The organic extracts are dried (Na2SO4) and concentrated under reduced pressure to give 3.50 g (95%) of clean desired product as a pale yellow solid.

Step B: 4-Amino-3-chloro-2-fluoro-5-nitro-benzoic acid 3a

Ammonium hydroxide solution (6.88 g, -30% in water, 58.9 mmol) is added to a solution of 3-chloro-2,4-difluoro-5-nitro-benzoic acid 2a (3.5 g, 14.7 mmol) in water (16 mL) at 0 °C with stirring. Upon completion of the ammonium hydroxide addition the reaction mixture is warmed to room temperature. After 5 hours the reaction mixture is cooled to 0 °C and concentrated HCl is carefully added until the pH of the reaction mixture is near zero. The solid is collected by filtration and washed with water and diethyl ether. The solids are transferred to a round bottom flask as a solution in MeOH and EtOAc and concentrated under reduced pressure to give 2.96 g of a yellow solid. The filtrate is partitioned between diethyl ether and water and the organic layer is washed with brine. The combined organic extracts are dried (Na2SO ) and concentrated under reduced pressure to give 0.65 g of product. Recovered a total of 3.61 g (104%) of pure desired product, that is carried forward without further purification.

Step C: 4~Amino-3-chloro-2-fluoro-5-nitro-benzoic acid methyl ester 4a

To a stirred solution of 4-amino-3-chloro-2-fluoro-5-nitro-benzoic acid 3a (3.61 g, 15.4 mmol) in THF (30 mL) and MeOH (10 mL), TMS diazomethane (9.23 mL, 2.0 M solution in hexanes, 18.5 mmol) is added. After completion of reaction, the reaction mixture is concentrated via rotary evaporation with acetic acid in the trap. The recovered oily solid is triturated with diethyl ether to provide 1.51 g of a yellow solid. The filtrate is concentrated and triturated with diethyl ether to give an additional 0.69 g of yellow solid. A total of 2.20 g (57%) of pure desired product is recovered.

Step D: 4-Amino-3-chloro-5-nitro-2-phenylamino-benzoic acid methyl ester 5c

4-Amino-3-chloro-2-fluoro-5-nitro-benzoic acid methyl ester 4a (2.20 g, 8.84 mmol) is suspended in MeOH (9.4 mL) and aniline (3.22 mL, 35.4 mmol) is added. The reaction mixture is heated to reflux with stirring under a nitrogen atmosphere. After 19 hours, the reaction is complete. Distilled water (3.22 mL) is added to the reaction mixture and refluxing is continued for one hour. The reaction mixture is cooled to 0 °C in an ice bath for 20 minutes. The reaction mixture is filtered and washed with 3:10 distilled water/MeOH (65 mL total) and then with MeOH. The solid is dissolved with CH2C12 and concentrated under reduced pressure to give 2.40 g (84%) of pure desired product. MS APCI (-) m/z 320.3 (M-l) detected.

Step E: 4, 5-Diamino-3-chloro-2-phenylamino-benzoic acid methyl ester 6b

4-Amino-3-chloro-5-nitro-2-phenylamino-benzoic acid methyl ester 5c (0.50 g, 1.55 mmol) is dissolved into 2:1 EtOH/MeOH (15.5 mL). Saturated aqueous NH4C1 (15 mL), Zn powder (1.02 g, 15.6 mmol), and THF (10 mL) are added. After stirring for 20 hours, the reaction mixture is diluted with CH C12/THF and water. The organic layer is washed with water (3x). The combined organic extracts are dried (Na2SO4) and concentrated under reduced pressure. The solids are triturated with ether to give 0.32 g (70%) clean desired product. Step F: 7-Chloro-6-phenylamino-3H-benzoimidazole-5-carboxylic acid methyl ester 7c

4,5-Diamino-3-chloro-2-phenylamino-benzoic acid methyl ester 6b (0.32 g, 1.09 mmol) and formamidine acetate (72 mg, 1.64 mmol) in EtOH (36 mL) are heated, with stirring, to 80 °C. After 44 hours, the reaction mixture is cooled to room temperature and diluted with EtOAc and washed with water (3x), saturated NaHCO3, and brine. The combined organic extracts are dried (Na2SO4) and concentrated under reduced pressure to give 0.33 g (99%) clean desired product as a solid. MS APCI (+) m/z 302.3 (M+l) detected.

Step G: 6-(4-Bromo-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester 8g

7-Chloro-6-phenylamino-3H-benzoimidazole-5-carboxylic acid methyl ester 7c (0.327 g, 1.08 mmol) is dissolved into DMF (16 mL) and NBS (0.193 g, 1.08 mmol) is added. After one hour, the reaction mixture is quenched by the addition of saturated aqueous NaHSO3. The reaction mixture is then partitioned between EtOAc/THF and water. The organic layer is washed with water and brine. The combined organic extracts are dried (Na2SO ) and concentrated under reduced pressure. The recovered solid is triturated with ether to give 0.225 g (54%) pure desired product. MS ESI (+) m/z 382, 384 (M+, Br pattern) detected.

Step H: 6-(4-Bromo-2-chloro-phenylamino)- 7 -chloro-3H-benzoimidazole-5 -carboxylic acid methyl ester lOdd 6-(4-Bromo-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester 8g (0.225 g, 0.591 mmol) is dissolved in DMF (2 mL) and NCS (79 mg, 0.591 mmol) is added. After the NCS is in solution concentrated HCl (0.005 mL, 0.059 mmol) is added. After 2 hours, sodium bicarbonate, water and NaHSO3 are added to the reaction mixture. Solids are filtered and washed with water and ether to give 0.141 g (57%) of clean desired product as a tan solid. MS APCI (-) m/z 414, 416 (M-, Br pattern) detected.

Step I: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid methyl ester lOee

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester lOdd (0.141 g, 0.34 mmol), potassium carbonate (0.141 g, 1.02 mmol), and iodomethane (0.063 mL, 1.02 mmol) are dissolved in dimethylformamide (3 mL). After 20 hours, the reaction mixture is diluted with EtOAc and washed with water (3x), potassium carbonate, and brine. The organic layer is dried (Na2SO4) and concentrated to a brown oil. The N3 and Nl alkylated regioisomers are separated by flash chromatography (EtOAc). The recovery of the N3 alkylated regioisomer is 20.4 mg (28%). MS ESI (+) m/z 428, 430 (M+, Br pattern) detected.

Step J: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid 10 ff

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid methyl ester lOee (21 mg, 0.048 mmol) is dissolved into 2:1 THF/water (1.2 mL) and NaOH (0.190 mL, 1.0 M aqueous solution, 0.190 mmol) is added. After stirring for 4 hours the reaction is diluted with water and acidified to pH 2 by addition of 1.0 M HCl. The mixture is then extracted with 3:1 EtOAc/THF (3x), dried (Na2SO ) and concentrated to give quantitative yield of desired prodcut as a white solid. MS APCI (+) m/z 414, 416 (M+, Br pattern) detected.

Step K: 6-(4-Bromo-2’chloro-phenylamino)- 7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-vinyloxy-ethoxy) -amide lOgg

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid lOff (32 mg, 0.077 mmol), O-(2-vinyloxy-ethyl)-hydroxylamine (0.010 mL, 0.092 mmol), HOBt (13 mg, 0.093 mmol), triethylamine (0.011 mL, 0.077 mmol), and EDCI (19 mg, 0.10 mmol) are dissolved into dimethylformamide (1.0 mL) and allowed to stir under a nitrogen atmosphere at room temperature for 24 hours. The reaction mixture is diluted with EtOAc, washed with water (3x), 10% potassium carbonate (2x), saturated ammonium chloride, brine, dried (Na2SO4), and concentrated under reduced pressure to give 39 mg of 85% pure material. MS APCI (-) m/z 497, 501 (M-, Br pattern) detected.

Step L: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide lOcc

Hydrochloric acid (0.78 mL, 1.0 M aqueous solution, 0.78 mmol) is added to a suspension of 6-(4-bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H- benzoimidazole-5-carboxylic acid lOgg (2-vinyloxy-ethoxy)-amide (39 mg, 0.078 mmol) in MeOH (1 mL). After one hour, the reaction mixture is neutralized to pH 7 and concentrated under reduced pressure. The solids are dissolved in EtOAc, washed with brine, dried (Na SO4), and concentrated under reduced pressure. Flash chromatography (20:1 CH2Cl2/MeOH) provides 9 mg (23%) of pure product: MS APCI (+) m/z 473, 475 (M+, Br pattern) detected; 1H NMR (400 MHz, CDC13) δ 8.30 (s, IH), 8.08 (s, IH), 7.57

(d, IH), 7.15 (dd, IH), 6.21 (d, IH), 3.97 (s, 3H) 3.86 (m, 2H), 3.57 (m, 2H).

 

actual is below

Example 18

The following compounds are prepared by methods similar to those described in

Example 10 by using methyl ester 8d and the appropriate alkylating agent (Step A) and

the appropriate hydroxylamine (Step C):

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

WO2014063024

http://patentscope.wipo.int/search/en/detail.jsf;jsessionid=E10680BCA177F821C7FEFA1AFC44A438.wapp2nA?docId=WO2014063024&recNum=6&maxRec=53841&office=&prevFilter=%26fq%3DICF_M%3A%22C07D%22&sortOption=Pub+Date+Desc&queryString=&tab=PCTDescription

COMPD A

Example 1. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-

 

Compound 1 Compound 3

 

In an inertized (N2) reaction vessel at internal temperature 20°C and under exclusion of humidity and air, Compound 1 (1.0 eq.) and Compound 2 (1.2 eq.) are reacted in the presence of cesium carbonate (2.4 eq.), tris(dibenzylidenaceton) dipalladium(O) (0.035 eq.) and Xantphos (0.07 eq.) in a mixture of toluene and 1 ,4-dioxane at internal temperature of 99°C. After 8 hours, the mixture is cooled to internal temperature of 60°C.

Subsequently, dimethylformamide (DMF), filter aid (CEFOK) and activated charcoal (EKNS) are added, and the mixture is stirred and cooled to internal temperature of 35 °C. The solids are filtered off and washed with a mixture of dimethylformamide and toluene. To the filtrate, which contains the product Compound 3, is introduced at internal temperature of

25 °C hydrogen chloride gas (CLC) whereupon the HQ salt of Compound 3 crystallizes. The palladium residue mainly remains in solution. After warming to 60 °C and cooling to 0°C, the solids are filtered using a centrifuge and are washed with a mixture of toluene and dimethylformamide.

The damp Compound 3 HC1 salt is charged to a reactor (equipped with pH probe) together with dimethylformamide and is heated to 60°C. By adding a 4 wt% of aqueous tripotassium phosphate solution, the pH is adjusted to a pH range of 6.8-7.6 (with a target of pH 7.2) while Compound 3 crystallizes as free base. After cooling to 22°C and stirring, the solids are filtered using a centrifuge and are washed with drinking water. The moist solids are dried at 50 °C under vacuum to give dry, crude Compound 3.

In order to remove residual palladium, dry, crude Compound 3 is dissolved in dimethylformamide at internal temperature of 60°C and stirred together with Smopex-234 (commercially available from Johnson Matthey) and activated charcoal for 90 minutes. The solids are filtered off at internal temperature of 60°C and are washed with

dimethylformamide. To the filtrate are added drinking water and Compound 3 seed crystals. More drinking water is added while Compound 3 crystallizes. After cooling to internal temperature of 20 °C, the solids are filtered using a centrifuge and are washed with a mixture of deionized water and dimethylformamide and with deionized water. The moist solids are dried at 50°C under vacuum, providing 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid methyl ester (Compound 3).

 

Example 2. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide

A. “One-pot” Synthesis

 

Compound 3 Intermediate 1

t-Bu-O. /\ ^ H2

(Compound 4)

 

Compound 5

In an inertized reaction vessel at internal temperature 20-25 °C under nitrogen, 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid methyl ester (Compound 3, 1.0 eq.) is added to a mixture of DMF and THF. To this slurry, a solution of potassium trimethylsilanolate (1.05 eq.) in THF is added to the mixture at internal temperature of 25 °C over a period of about 40 minutes, and the resulting mixture is stirred for about 1 hour, providing a potassium salt solution of Intermediate 1. A THF/methanol mixture is then sequentially distilled off from the mixture at 85-120°C during about 2 hours.

The potassium salt solution is then added to a suspension of CDI (1.25 eq.) and imidazole hydrochloride (1.40 eq.) in THF at internal temperature of 25 °C over a period of about 1 hour. The resulting mixture is then stirred for approximately 1 hour at 50°C, and the following imidazolide intermediate

 

 

The imidazolide intermediate is not further isolated.

Subsequently, 1.2 eq. of 0-(2-tert-butoxyethyl)hydroxylamine (Compound 4, CAS No. 1023742-13-3, available from suppliers such as Huhu Technology, Inc.®) is added over a period of about 30 minutes at 50°C and stirred for 1.5 hours. Demineralized water is then added at 50°C, producing a precipitate. After cooling to 20°C and stirring for about 3-16 hours, the slurry is filtered off, washed with THF/ demineralized water (1 :2) in 2 portions and with demineralized water in three portions, and dried at 50°C / <70 mbar for about 17 hours, providing 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) as monohydrate.

 

B. A synthesis method with isolation of the intermediate of step a) from the reaction mixture of step a) prior to the reaction of step b)

Alternatively, 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5 -carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) can be made by the synthesis method as shown below. Compound 3, which is a methyl ester, is first converted to a carboxylic acid, which is then isolated by a crystallization to form Compound

6. Compound 6 is then coupled with Compound 4 to form Compound 5 as monohydrate.

The crystallization step in this method removes starting materials such as Compound 1, process impurities, and the dba ligand from the prior catalyst before the coupling reaction with Compound 4, and at the same time maintains the overall yield of the synthesis.

 

 

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-memy acid In an inertized (N2) reaction vessel at internal temperature of 60°C, Compound 3 (1.0 eq.) is dissolved in DMF and stirred with a fiber, which is sold under the trademark

SMOPEX 234, and activated charcoal for the removal of palladium to not more than 100 ppm. The fiber and activated charcoal are removed by filtration at 60°C and washed with DMF.

The filtrate (containing Compound 3) is transferred to a second inertized (N2) reaction vessel and cooled to an internal temperature of 30°C. A thin suspension can form at this point of time. 30% sodium hydroxide (1.1 eq.) and water (for rinsing) are added, and the resulting reaction mixture is vigorously stirred for 3 hours at an internal temperature of 30 °C. The methyl ester is saponified. Conversion is checked by an IPC (HPLC). As soon as the IPC criterion is met, a filter aid, which is sold under the trademark HYFLO, is added. The mixture is stirred for 15 minutes and then filtered at 30°C via a plate filter and polish filter to a third reaction inertized (N2) vessel.

An aqueous HC1 solution 7.5 % is added to the clear filtrate in the third vessel at an internal temperature of 30 °C until a pH value of 8 is reached. Then the solution is seeded at an internal temperature of 30°C with Compound 6, and an aqueous HC1 solution 7.5 % is added under vigorous stirring until a pH value of pH 2.8 is reached. The product gradually crystalizes. The suspension is cooled over 60 min to an internal temperature of 25 °C and

water is added. The suspension is stirred for at least 4 hours at an internal temperature of 25°C.

The resulting solid is collected by centrifugation or filtration. The filter cake is first washed with DMF/water 1 :1 (w/w) and then with water, discharged and dried in a vacuum at 50°C. The water content is controlled by IPC. The crystalline product Compound 6 is discharged as soon as the IPC criterion is met.

 

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid- (2-tert-butoxyethoxy) – amide

An inertized (N2) reaction vessel is charged with Compound 6 (1.0 eq.), DMF, and

THF at room temperature. The suspension is heated to 25 °C under stirring with flow of nitrogen. After CDI (1.13 eq.) is added, the suspension can get thinner and slight evolution of gases can be observed. After the suspension finally becomes a solution, it is then monitored by IPC (HPLC).

As soon as the IPC (HPLC) criterion is met, the reaction mixture is heated to 50°C over 20 minutes and imidazole hydrochloride (0.3 eq.) is added, forming a solution of

Intermediate 2.

To the solution of Intermediate 2, Compound 4 (1.3 eq.) is added over 60 minutes at internal temperature of 50°C under stirring at a speed of 300 rpm with flow of nitrogen. As soon as the IPC (HPLC) criterion is met, the mixture is cooled to 20-25 °C over 30 minutes. The mixture is then stored at ambient temperature overnight under nitrogen without stirring. DMF is added to the mixture followed by heating it to 50 °C over 30 minutes. Complete conversion of Intermediate 2 to Compound 5 is confirmed by IPC (HPLC).

Water is added to the mixture at internal temperature of 50 °C over 20 minutes. Then the solution is seeded with Compound 5. After stirring at 50 °C for 60 minutes, more water is added to the suspension at 50 °C over 90 minutes. After vigorous stirring, the suspension is cooled to 20 °C over 2 hours and filtered. The filter cake is washed twice with THF/water (v/v: 1 :2) at 20 °C, and twice with water at 20 °C. Finally, the filter cake is dried at 50 °C under vacuum to provide 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) as monohydrate.

 

Example 3. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A)

Compound 5 Compound A

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) monohydrate is added in 3 portions to a premixed solution of Acetonitrile and excess Phosphoric acid (85 % aqueous solution) at internal temperature 20-25 °C. After stirring for about 15 minutes, the suspension is heated to internal temperature 50-53 °C. The suspension is maintained at this temperature for 6 hours, cooled to internal temperature 20-25 °C. The mixture is then heated to internal temperature 35-37°C and diluted with Ethanol- Water (3 :1 v/v). EKNS and CEFOK are added, the reaction mixture is stirred approximately 15 minutes and filtered over a funnel coated with CEFOK. The filtrate is cooled to approximately 30°C. 3 N aqueous potassium hydroxide (ΚΟΗ) is added to the cooled filtrate over a period of 90 minutes until a pH- value of about 8.1 is reached. The suspension is heated to internal temperature 60-63 °C, stirred at this temperature for a period of about 2 hours, cooled to 20-23 °C over a period of about 45 minutes, filtered over a funnel, and dried at 50°C pressure <100 mbar over a period of about 17 hours, providing 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A) as a white powder.

 

Example 4. Preparation of Crystallized 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A) In a dry vessel at room temperature, Compound A is added to a premixed solvent solution of methanol/THF/water (35/35/30 w/w). The suspension is heated to internal temperature 53-55°C, and the resulting solution is hot filtered by deep and membrane filtration (via a paper filter and PTFE membrane) at internal temperature 53-56°C. The clear solution is stirred and cooled to 47-48°C, and the seed crystals suspension (i.e., seed crystals of crystallized Compound A in water, 10% m/m) is added (0.2 to 0.5% of crystallized Compound A expected yield mass). After about 20 minutes, water is slowly added within 25 hours (33.3% within 15 hours and 66.6% within 10 hours with at least 10 minute stirring after addition of water) to obtain a final ratio of methanol THF/water (20/20/60 w/w). After the water is added, the suspension is cooled down to internal temperature 3-5 °C within 10 hours and stirred for 0.5 hours. The white suspension is filtered over a sinter glass nutsche (75 ml, diameter = 6 cm, pore 3) suction filter and washed once with ice cold methanol/THF/water (15/15/70 w/w at 2-4 °C), and two times with ice cold water (2-4 °C). Drying takes place in a vacuum oven dryer at 20°C for 10 hours, and then at 40°C for 10 hours, and then at 60°C for at least 12 hours with pressure < lOmbar, providing crystallized Compound A.

Example 5. Pharmaceutical Composition

Crystallized Compound A is formulated as indicated in Table 1 :

Table 1

 

* The weight of the drug substance is taken with reference to the dried substance (100%) on the basis of assayed value. The difference in weight is adjusted by the amount of lactose monohydrate.

** The Opadry II is combined with the sterile water to make a 12% w/w Opadry II (85F) film coat suspension, which is then sprayed onto the core tablet.

*** Removed during processing

 

Upon mixing of the tablet core components, the pharmaceutical composition is converted into a tablet form by direct compression. The formed tablet may be further coated with the tablet coating provided above.

 

Revaprazan hydrochloride


Molecular formula: C22H23FN4 =362.5.

Yuhan Corporation

Reversible H+/K+-ATPase Inhibitors

CAS: 199463-33-7FREE BASE  . CAS 178307-42-1

UNII code: 5P184180P5.

SB-641257A
YH-1885

N-(4-Fluorophenyl)-4,5-dimethyl-6-(1-methyl-1,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidin-2-amine hydrochloride
N-[4,5-Dimethyl-6-(1-methyl-1,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidin-2-yl]-N-(4-fluorophenyl)amine hydrochloride

5,6-dimethyl-2-(4-fluorophenyl¬ amino)-4-(1-methyl-l,2,3,4-tetrahvdroisoquinolin-2-yl)- pyrimidine hydrochloride

 

INTRO

Revaprazan hydrochloride, a reversible proton pump inhibitor with long-lasting acid-suppressive effects, was first launched in Korea in 2005 by Yuhan for the treatment of duodenal ulcer, gastric ulcer and gastritis. The compound is also undergoing phase II clinical studies for the treatment of of Non-erosive Reflux Disease (NERD).

Discovered by Yuhan, revaprazan hydrochloride was licensed to GlaxoSmithKline (GSK) in 2000 for worldwide development and commercialization except in South and North Korea.

Revaprazan, whose chemical name is 5,6-dimethyl-2-(4-fluorophenylamino)-4-(1-methyl-1,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine, is represented by the following Formula 1. Revaprazan can be used in a form of an acid addition salt, including e.g., HCl salt (see International Publication No. WO1996/05177, WO1997/042186, and WO1998/018784).

Formula 1

 

Revaprazan or its salt is reversibly bound to a H+/K+ exchange site of a proton pump (H+/K+ ATPase) existing in a gastric parietal cell so that secretion of H+ into the gastric lumen is competitively inhibited. Revaprazan or its salt is also bound to a specific site of H+/K+ ATPase, thereby inhibiting transport of H+ and suppressing an acid secretion to the gastric lumen, which results in increasing the intragastric pH. Unlike irreversible proton pump inhibitors, e.g., omeprazole, revaprazan or its salt is not dependent upon acid activation of a drug in a stomach or secretion status of a proton pump. Therefore, based on the mechanism different from irreversible proton pump inhibitors, such as omeprazole, revaprazan or its salt is classified into an acid pump antagonist (APA).

Revaprazan has very low water-solubility, i.e. less than 0.2 mg/mL, and even lower solubility in a buffer solution having pH 1 to 12. And also, revaprazan has very low intrinsic dissolution rate, i.e., about 0.0086 mg/min/cm2. Due to such a low solubility and intrinsic dissolution rate, its dissolution in the gastrointestinal tract is also very low. Therefore, when revaprazan is orally administered, its absorption rate is relatively low. Revaprazan also has strong adhesion and agglutination properties, and thus, when revaprazan is formulated into a capsule or a tablet, it may be stuck to a punch or a die, thereby showing low formulation processability. In order to address these problems, WO 2008/078922 has disclosed a pharmaceutical composition for oral administration comprising a solid dispersion in which revaprazan particles are surface-modified with a water-soluble polymer, a water-soluble saccharide, a surfactant, or a mixture thereof.

WO-2014060908   patent………. Improved process for preparation of revaprazan hydrochloride comprising reacting 1-methyl-1,2,3,4-tetrahydroisoquinoline with 4-halide-2-(4-flurophenylamino)-5,6-dimethylpyrimidine in the presence of a transfer catalyst (eg tetra butyl ammonium bromide), solvent (eg methyl isobutyl ketone) and treated with HCl. Also claims purification and crystallization of the API. Appears to be the first filing from the Lupin on this API. Family members of the product case, WO9605177 (assigned to Yuhan Corp,    Yuhan Corporation   ), expire in August 2015

Chlorination of 5,6-dimethyl-2,4-dihydroxypyrimidine (VIII) using phosphorus oxychloride in the presence of N,N-dimethylaniline provided dichloropyrimidine (IX). The 4-chloro group of (IX) was then selectively displaced with tetrahydroisoquinoline (IV) to afford adduct (X). The title compound was then obtained by condensation of the 2-chloropyrimidine (X) with 4-fluoroaniline (XI), followed by conversion to the corresponding hydrochloride salt

 

In a different method, amine (I) was alkylated with 2-bromoethanol (V) to give the N-(hydroxyethyl) amine (VI), which was further converted to bromo amine (VII) by treatment with concentrated HBr. Friedel-Crafts cyclization of (VII) upon heating in the presence of AlCl3 furnished tetrahydroisoquinoline (IV).

………….

 

The intermediate tetrahydroisoquinoline (IV) has been prepared by two synthetic strategies. Condensation of alpha-methyl benzylamine (I) with alpha-chloro-alpha-(methylsulfanyl)acetyl chloride (II) in the presence of SnCl2 furnished the tetrahydroisoquinolinone (III). Reductive cleavage of the methylsulfanyl group of (III) employing Raney-Ni, followed by lactam reduction, provided intermediate (IV).

 

In a different method, cetylation of phenethylamine (XVI) with acetyl chloride (XVII) by means of Et3N in dichloromethane provides N-(2-phenylethyl)acetamide (XVIII), which is cyclized with hot polyphosphoric acid to afford 1-methyl-3,4-dihydroisoquinoline (XIX). Finally, compound (XIX) is reduced with sodium borohydride in EtOH.

 

In an alternative procedure, 4-fluoroaniline (XI) was condensed with cyanamide under acidic conditions to afford the fluorophenyl guanidine (XII). Cyclization of guanidine (XII) with ethyl 2-methylacetoacetate (XIII) in hot DMF produced pyrimidine (XIV). After chlorination of (XIV) with POCl3, the resultant chloropyrimidine (XV) was condensed with tetrahydroisoquinoline (IV) in the presence of either KOAc or Et3N to furnish the title diaminopyrimidine.
///////////////////

WO1996005177

http://www.google.com/patents/WO1996005177A1?cl=en

Example 15: Synthesis of 5,6-dimethyl-2-(4-fluorophenyl¬ amino)-4-(1-methyl-l,2,3,4-tetrahvdroisoquinolin-2-yl)- pyrimidine hydrochloride

After 4-fluoroaniline(l.0ml, lOmmol) waε added to a mixture εolution of 5,6-dimethyl-4-(1-methyl-l,2,3,4- tetrahydroiεoquinolin-2-yl )-2-chloropyrimidine( 1.4g, 4.8mmol) and dimethylformamide(10ml) , 1.32g of the title compound waε obtained in accordance with the εame procedure as in Step 2 of Example 1. Yield: 69% M.P.: 205-208°C 1H-NMR(DMSO-d6) : δ 1.58(d, 3H), 2.17(s, 3H), 2.36(s, 3H), 2.89(bd, IH), 3.08(m, IH), 3.59(m, IH), 4.19(bd, IH), 5.38(q, IH), 7.34(m, 6H), 7.60(m, 2H), 10.40(s, IH). Example 16: Synthesis of (R)-5,6-dimethyl-2-(4-fluorophenyl¬ amino)-4-(1-methyl-l,2,3,4-tetrahydroisoquinolin-2-yl)- pyrimidine hydrochloride

After 4-fluoroaniline(lml, lOmmol) was added to a mixture solution of (R)-5,6-dimethyl-4-(1-methyl-l,2,3,4- tetrahydroiεoquinolin-2-yl )-2-chloropyrimidine(1.4g, 4.8mmol) and dimethylformamide(10ml) , 1.20g of the titled compound waε obtained in accordance with the εame procedure as in Step 2 of Example 1. Yield: 62.7% M.P.: 205-207°C

1H-NMR(DMSO-d6) : δ 1.58(d, 3H), 2.17(s, 3H), 2.36(s, 3H), 2.89(bd, IH), 3.08(m, IH), 3.59(m, IH), 4.19(bd, IH), 5.38(q, IH), 7.34(m, 6H), 7.60(m, 2H), 10.40(s, IH) .

,,,,,,,,,,,,,,,,,,,,,,,,

 http://www.google.com/patents/EP0900214A1?cl=en

5,6-Dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4-tetra- hydroisoquinolin-2-yl)pyrimidine of the above formula (I) inhibits gastric acid secretion by means of a reversible proton-pump inhibiting effect and, therefore, can be used as an anti -ulcer agent. This compound was developed by the inventors of the present invention, who then applied for patents for the compound and/or its method of preparation in Korea and other countries (see International Publication No. WO 96/05177).

According to the method disclosed in the above patent application, 5,6-dimethyl-2-(4-f luoropheny lamino) -4- ( 1 -methyl- 1, 2,3,4 -tetrahydroisoq uinolin-2-yl)pyrimidine is prepared according to the following reaction scheme A:

Reaction scheme A

 

 

Since the starting material of the above reaction scheme has two reactive sites (i.e., the two CI atoms), the first reaction inevitably produces a side product, which reduces the yield of the desired compound.

The present inventors have long labored to develop a novel method for preparing 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine of formula (I) without producing side products. As a result, we have discovered that the desired compound of formula (I) can be efficiently prepared without side products by reacting the pyrimidine derivative represented by formula (LI-A) with l-methyl-l,2,3,4-tetrahydroisoquinoline represented by formula (III) and, thus, have completed the present invention.

DISCLOSURE OF THE INVENTION

The present invention relates to a novel process for preparation of 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4-tetrahydroiso- quinolin-2-yl)pyrimidine represented by formula (I) and its acid addition salts.

More specifically, the present invention relates to a process for preparation of 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4- tetrahydroisoquinolin-2-yl)pyrimidine represented by formula (I),

 

and its acid addition salts wherein a pyrimidine derivative represented by the following formula (II-A),

 

in which Hal represents a halogen, is reacted with 1 -methyl- 1,2,3,4 – tetrahydroisoquinoline represented by formula (HI),

 

In addition, the present invention relates to a process for preparation of the pyrimidine derivative of formula (II-A) and the compound of formula (HI). Further, the present invention relates to a novel intermediate compound represented by the following formula (LI), which includes the pyrimidine derivative represented by formula (II-A),

 

in which R represents hydroxy or a halogen.

BEST MODE FOR CARRYING OUT THE INVENTION

According to the present invention, the compound of formula (I) can be prepared by reacting the compound of formula (LI-A) with 1 -methyl -1,2,3,4-tetrahydroisoquinoline of formula (IT), as depicted in the following reaction scheme 1:

Reaction scheme 1

 

ω

Since the starting compound of the reaction scheme 1 (i.e., the compound of formula (II-A)) contains a single reactive site (i.e., Hal), this reaction scheme does not produce any side product and, thus, optimizes the yield of the compound of formula (I), the desired product.

The present invention is described in more detail below. Although the 4-halogeno-2-(4-fluorophenylamino)-5,6-dimethyl- pyrimidine represented by formula (II-A) can be reacted according to the present invention with an equivalent amount of 1 -methyl- 1,2,3,4-tetra- hydroisoquinoline represented by formula (HI), it is preferable to conduct the reaction using an excess, rather than an equivalent amount, of the latter. Since the latter is a liquid under reaction conditions, the unreacted l-methyl-l,2,3,4-tetrahydroisoquinoline can be readily removed after the reaction has gone to completion.

Preparation of 5.6-dimethyl-2-(4-fluorophenylaminn)-4-(l-methyl- lr2.3.4-tetrahvdroisoαuinolin-2-yl)pyrimidine and its hydrochloride

In Examples 14 to 20, inclusive, l-methyl-l,2,3,4-tetrahydroiso- quinoline prepared according to the method disclosed in International 0 Publication No. WO 94/14795 was used as the reactant.

Example 14

2.65g(27 mmole) of potassium acetate and 4.0g(26.9 mmole) of 5 1-methyl -1,2,3,4-tetrahydroisoquinoline were added to 85ml of n-hexanol and then warmed to 80 °C. 6.17g(24.5 mmole) of 4-chloro-2-(4-fluoro- pheny lamino) -5,6 -dimethy Ipyrimidine was added thereto and then reacted at 140 °C for 28 hours under refluxing to prepare 5,6-dimethyl-2-(4-fluo- rophenylamino)-4-(l-methyl-l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. 0

The reaction solution was cooled to room temperature, diluted with 20ml of acetone and then added dropwise to 120ml of water with stirring. After it had been stirred for 2 hours, the resulting solid product was filtered, washed with 30ml of water, dissolved in 150ml of dichloromethane 5 and then washed successively with 20ml of 4N-HC1, 20ml of water and then 20ml of 4N-sodium hydroxide solution. The dichloromethane layer was dehydrated with anhydrous magnesium sulfate, concentrated under reduced pressure, and then diluted with 100ml of ethanol. To this reaction solution was added 30g of cone, hydrochloric acid, and the 0 mixture thereby obtained was stirred for 5 hours. The resulting solid product was filtered, washed with 20ml of ethanol and then dried to obtain 6.1g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l- methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)ρyrimidine hydrochloride.

S5 Yield : 62.4% m.p. : 255*0

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 211), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

Examnle 15

8.12g(11.2ml, 80.3 mmole) of triethylamine, 30ml of n-butanol and 6.58g(44.1 mmole) of 1-methyl- 1,2,3,4-tetrahydroisoquinoline were added to 40ml of ethylene glycol. 10.1g(40.1 mmole) of 4-chloro-2-(4-fluoro- phenylamino)-5,6-dimethylpyrimidine was added thereto and then reacted at 130 °C for 30 hours under refluxing to prepare 5,6-dimethyl-2-(4- f luorophenylamino )- 4 -( 1 – methyl – 1 ,2,3,4 – tetrahydroisoquinolin- 2 – yl ) -pyri – midine. This product was treated according to the procedure detailed in Example 14 to obtain 14.7g of purified 5,6-dimethyl-2-(4-f luorophenyl – amino)-4-(l-methyl-l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine hydro¬ chloride.

Yield : 91% m.p. : 256*0

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

III), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

F/xatnple 1β

45ml of triethylamine, 50ml of n-butanol and 32g(217 mmole) of l-methyl-l,2,3,4-tetrahydroisoquinoline were added to 150ml of ethylene glycol. 51.3g(203.8 mmole) of 4-chloro-2-(4-f luorophenylamino) -5,6- dimethy Ipyrimidine was added thereto and then reacted at 135 °C for 28 hours under refluxing to prepare 5,6-dimethyl-2-(4-fluorophenylamino)- 4- (1-methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 14 to obtain 66g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2, 3,4-tetrahydroiso-quinolin-2-yl)pyrimidine hydrochloride.

Yield : 81.1% > m.p. : 256*0

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 311), 2.84(m,

III), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH) 0

 

 

75ml of triethylamine and 65g(442 mmole) of 1-methyl- 1,2,3,4- tetrahydroisoquinoline were added to 100ml of 1,2 -propylene glycol. 5 100.9g(0.40 mmole) of 4-chloro-2-(4-fluorophenylamino)-5,6-dime- thy Ipyrimidine was added thereto and then reacted at 120 °C for 64 hours under refluxing to prepare 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l- methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 14 to obtain 91g 0 of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4- tetrahy droi soquinolin – 2 – y 1 )py rimidine hydrochloride.

Yield : 57.1% m.ρ. : 258°C 5 NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

0 Fvample 18

720ml of triethylamine and 695g(4.72 mmole) of l-methyl-1,2,3,4- tetrahydroisoquinoline were added to 2100ml of 1,2-propylene glycol.

1179g(4.68 mmole) of 4-chloro-2-(4-f luorophenylamino) -5,6-dimethyl-

35 pyrimidine was added thereto and the mixture thereby obtained was reacted at 130°C for 58 hours to prepare 5,6-dimethyl-2-(4-fluorophenyl- amino ) – 4 – ( 1 – methyl – 1 ,2,3,4 – tetrahydroisoquinolin – 2 – y 1 )pyrimidine . Thi s product was treated according to the procedure detailed in Example 14 to obtain 1250g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l- methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine hydrochloride.

Yield : 66.9% m.p. : 258*0

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m, IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH),

5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

Example 19

110ml of n-butanol, 240ml of triethylamine and 236g(1.60 mmole) of 1-methyl- 1,2,3,4-tetrahydroisoquinoline were added to 600ml of ethylene glycol. 400g(1.59 mmole) of 4-chloro-2-(4-fluorophenyl- amino)-5,6-dimethylpyrimidine was added thereto and then reacted at 140 °C for 48 hours to prepare 5,6-dimethyl-2-(4-fluorophenylamino)-4- ( 1 -methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 14 to obtain 485g of purified 5,6-dimethyl-2- (4-f luorophenylamino) -4- (1 -methyl – l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine hydrochloride.

Yield : 76.5% m.p. : 257 °C

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33

(s, IH), 13.43(bs, IH)

Example 20

240ml of triethylamine and 9.7g(65.8 mmole) of 1-methyl- 1,2,3,4- tetrahydroisoquinoline were added to 25ml of 1,2-propylene glycol. Then, 15g(51 mmole) of 4-bromo-2-(4-fluorophenylamino)-5,6- dimethyl- pyrimidine was added thereto and the mixture thereby obtained was reacted at 110°C for 28 hours. The resulting product was treated according to the procedure detailed in Example 14 to obtain 15.86g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4-tetra- hydroisoquinolin-yDpyrimidine hydrochloride.

Yield : 78% m.p. : 257 °C

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 311), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 211), 10.33 (s, IH), 13.43(bs, IH)

Example 21

8.12g(11.2ml, 80.3 mmole) of triethylamine, 30ml of n-butanol and 6.58g(44.1 mmole) of 1-methyl- 1,2,3,4-tetrahydroisoquinoline as prepared in Example 5 were added to 40ml of ethylene glycol. 10.1g(40.1 mmole) of 4-chloro-2-(4-fluorophenylamino)-5,6-dimethylpyrimidine was added thereto and then reacted at 130 °C for 30 hours under refluxing to prepare 5,6-dimethyl-2-(4-fluorophenylamino)-4-( 1-methyl- 1, 2,3,4 -tetra- hydroisoquinolin-2-yl)pyrimidine.

The reaction solution was cooled to room temperature, diluted with 30ml of acetone and then added dropwise to 200ml of water with stirring. After it had been stirred for 2 hours, the resulting solid product was filtered, washed with 60ml of water, dissolved in 250ml of dichloromethane and washed successively first with 35ml of 4N-HC1, 35ml of water and then with 40ml of 4N- sodium hydroxide solution. The dichloromethane layer was dehydrated with anhydrous magnesium sulfate, concentrated under reduced pressure, and then diluted with 200ml of ethanol. To this reaction solution was added 45g of concentrated hydrochloric acid, and the mixture was stirred for 5 hours. The resulting solid product was filtered, washed with 30ml of ethanol and then dried to obtain 9.82g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4-tetra- hydroisoquinolin-2-yl)pyrimidine hydrochloride.

Yield : 66.53% m.p. : 255 °C

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

 

Example 22

75ml of triethylamine and 65g(442 mmole) of l-methyl-1,2,3,4- tetrahydroisoquinoline as prepared in Example 7 were added to 100ml of 1,2-propylene glycol. 100.9g(0.40 mmole) of 4-chloro-2-(4-fluoro- phenylamino)-5,6-dimethylpyrimidine was added thereto and then reacted at 120*0 for 64 hours to prepare 5,6-dimethyl-2-(4-fluorophenylamino)- 4- (1-methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 21 to obtain 95.1g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine hydrochloride.

Yield : 59.67% m.p. : 258 °C

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21 (s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

Example 23

14ml of triethylamine and 9.7g(65.8 mmole) of 1-methyl- 1,2,3,4- tetrahydroisoquinoline as prepared in Example 7 were added to 25ml of 1,2-propylene glycol. 15g(51 mmole) of 4-bromo-2-(4-fluorophenyl- amino) -5,6 -dimethy Ipyrimidine was added thereto and then reacted at 120 °C for 28 hours to prepare 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l- methyl-l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 21 to obtain 14.9g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4- tetr ahy droisoquinolin – 2 – y 1 ) pyrimidine hydrochloride.

Yield : 73.28% m.p. : 257*0 NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

Example 24

8.12g(11.2ml, 80.3 mmole) of triethylamine, 30ml of n-butanol and 6.58g(44.1 mmole) of (R)-(+)-l-methyl-l,2,3,4-tetxahydroisoquinoline as prepared in Example 9 were added to 40ml of ethylene glycol. 10. Ig (40.1 mmole) of 4-chloro-2-(4-f luorophenylamino) -5,6-dimetlιylpyrimidine was added thereto and then reacted at 130 °C for 30 hours under refluxing to prepare 5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl-l,2,3,4- tetrahy droisoquinolin- 2 – y 1 ) pyrimidine.

The reaction solution was cooled to room temperature, diluted with

30ml of acetone and then added dropwise to 200ml of water with stirring. After it had been stirred for 2 hours, the resulting solid product was filtered, washed with 60ml of water, dissolved in 250ml of dichloromethane arid then washed successively with 35ml of 4N-HC1, 35ml of water and then 40ml of 4N-sodium hydroxide solution. The dichloromethane layer was dehydrated with anhydrous magnesium sulfate, concentrated under reduced pressure, and then diluted with 200ml of ethanol. To this reaction solution was added 45g of cone, hydrochloric acid, and the resulting mixture was stirred for 5 hours. The resulting solid product was filtered, washed with 30ml of ethanol and then dried to obtain 9.21g of purified (R)-(+)-5,6-dimethyl-2-(4-fluorophenylamino)-4-(l- methyl- l,2,3,4-tetrahydroisoquinolin-2-yl)pyrimidine hydrochloride.

Yield : 62.4% m.p. : 255 °C

[ a h20 : +250° (c=l, in CHC13)

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33 (s, IH), 13.43(bs, IH)

Example 25

23ml of triethylamine and 16g(108.5 mmole) of (R)-(+)-l-methyl- 1,2,3,4-tetrahydroisoquinoline as prepared in Example 10 were added to 75ml of ethylene glycol. 25.7g(101.8 mmole) of 4-chloro-2-(4-fluoro- phenylamino)-5,6-dimethylpyrimidine was added thereto and the mixture thereby obtained was reacted at 135 °C for 28 hours under refluxing to prepare (R)-(+)-5,6-dimethyl-2-(4-fluorophenylamino)-4-(l-methyl- 1,2,3, 4-tetrahydroisoquinolin-2-yl)pyrimidine. This product was treated according to the procedure detailed in Example 24 to obtain 33g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4-( 1-methyl- 1,2,3,4-tetra- hydroisoquinolin-2-yl)-pyrimidine hydrochloride.

Yield : 81.1% m.p. : 257 °C

I a h20 ■ +250° (c=l, in CHCI3)

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m,

IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33

(s, IH), 13.43(bs, IH)

Example 2β

14ml of triethylamine and 9.7g(65.8 mmole) of (R)-(+)-l-methyl- 1,2,3,4-tetrahydroisoquinoline as prepared in Example 10 were added to 25ml of 1,2-propylene glycol. 15g(51 mmole) of 4-bromo-2-(4-fluoro- phenylamino)-5,6-dimethylpyrimidine was added thereto and the mixture thereby obtained was reacted at 120 °C for 28 hours. The reaction product was thentreated according to the procedure detailed in Example 24 to obtain 16.2g of purified 5,6-dimethyl-2-(4-fluorophenylamino)-4- ( 1 – methyl – 1 ,2,3,4 – tetrahy droisoquinolin – 2 – y 1 )py rimidine hydrochloride.

Yield : 79.97% m.p. : 257 °C

[ a h20 : +250° (c=l, in CHC13)

NMR(CDC13, ppm) : 1.58(d, 3H), 2.21(s, 3H), 2.38(s, 3H), 2.84(m, IH), 3.12(m, IH), 3.61(m, 2H), 4.23(m, IH), 5.38(q, IH), 7.25(m, 6H), 7.61(m, 2H), 10.33(s, IH), 13.43(bs, IH)……….SDEE PATENT

………………………………..

http://www.google.com/patents/CN102863423A?cl=en

the formula I 5,6 _ ni-2 – (4 – fluorophenyl amino) -4 – (1 – methyl-1, 2,3,4 tetrahydro-isoquinolin-2 _ – yl) pyrimidine and its hydrochloride salt is a kind of reversible proton pump inhibitors ー having novel and unique mechanism of action of potassium competitive acid pump inhibitor to acid inhibition stronger, faster onset smaller side, the Short-term treatment of gastritis, duodenal ulcer, gastric ulcer and gastroesophageal reflux has certain advantages.

[0003]

Figure CN102863423AD00031

[0004] CN95194599. 8 for the first time disclosed the compound and its preparation method, CN97194367. 2 discloses the compound to another ー preparation methods.

[0005] CN95194599. 8 discloses prepared as follows:

[0006]

[0007]

Figure CN102863423AD00032

[0008] 2,4 – chloro-5 ,6 ni – ni methylpyrimidine with two chlorine in the preparation, to be equivalent to twice the substrate in the reaction of phosphorus oxychloride; the two chlorine atoms, or two a reactive centers, and I-methyl-1, 2,3,4 – tetrahydroisoquinoline reaction inch, will inevitably produce structurally similar by-products affecting the yield of the reaction, while the reaction Refined product difficult.

[0009] The CN97194367. 2 Public Preparation: [0010]

Figure CN102863423AD00041

[0011] 1_ methyl-1, 3,4 – four oxyiso Thrill Lynn is by I-methyl -3,4-_ ■ oxyiso obtained by the reduction of noise Lynn, there is not sufficient to restore the problem, raw materials and products of similar structure, easy separation and purification. Prepared by this method to obtain an I-methyl-1, 2,3,4 – tetrahydroisoquinoline is often close to tan a brown liquid, and I-methyl-1, 2,3,4 – tetrahydro- isoquinoline is ー secondary amines, placed in contact with air at room temperature, long time ー easy oxidative deterioration, become darker in color, is not conducive to storage.

More particularly, the present invention relates to formula I is 5,6 _ ni-2 – (4 – fluorophenyl amino) -4 – (1 – methyl-1 ,2,3,4 – Four Hydrogen isoquinolin-2 – yl) pyrimidine and its hydrochloride salt thereof. In the method, represented by formula III 5,6 – ni methyl -2 – (4 – fluorophenyl amino) pyrimidine represented by formula II and I-methyl-3 ,4 – ni isoquinoline hydrogen, to give a quaternary ammonium salt represented by formula IV.

[0014]

Figure CN102863423AD00051

  Then borohydride reduction obtained with high purity 5,6 – ni methyl -2 – (4 – fluorophenyl-amino) -4 – α-methyl-1, 2,3,4 – tetrahydro- isoquinolin-2 – yl) pyrimidine and its hydrochloride.

  reaction scheme is as follows:

 

Figure CN102863423AD00052

Example I

  I-methyl-3 ,4 – ni hydrogen isoquinoline 20g, 5,6 – ni methyl -2 – (4 – fluorophenyl-amino) pyrimidine 28.8g, in 200mL of dry toluene, loading a stirrer, a thermometer, a reflux condenser, drying tube reaction flask under nitrogen, heated to reflux, began to produce turbidity, the reaction 30h. Filtration, washed with anhydrous ether ko. Solid rapidly dissolved with anhydrous ko alcohol 200mL, graded by adding NaBH44. 8g, plus complete, continue stirring at room temperature lh. The solvent was distilled off under reduced pressure, the residue was added water, 200mL, concentrated aqueous ammonia was adjusted to pH> 10, ni chloride extract was washed with water, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness, and then dissolved in acetone, which leads to dry HCl gas, a solid precipitated. Filtered and the solid was dissolved with anhydrous alcohol ko, active carbon, filtered, frozen crystallization to give a white powder 29. 2g, yield 63.9%.

Example 2

  I-methyl-3 ,4 – ni hydrogen isoquinoline 15g, 5,6 – ni methyl -2 – (4 – fluorophenyl amino) Li Jie secret 21. 6g, in 150mL of dry toluene , flask equipped with stirrer, thermometer, reflux condenser, drying tube reaction flask under nitrogen, heated to reflux, began to produce turbidity, the reaction 24h. Filtration, washed with anhydrous ether ko. Solid rapidly dissolved with anhydrous ko alcohol 150mL, graded by adding NaBH43. 6g, plus complete, continue stirring at room temperature lh. The solvent was distilled off under reduced pressure, the residue was added water, 150mL, concentrated aqueous ammonia was adjusted to pH> 10, ni chloride extract was washed with water, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness, and then dissolved in acetone, which leads to dry HCl gas, a solid precipitated. Filtered and the solid was dissolved with anhydrous alcohol ko, active carbon, filtered, frozen crystallization to give a white powder 21. 9g, yield 64. I%.

W. LI ET AL.: ‘Preparation and in vitro/in vivo evaluation of ravaprazan hydrochloride nanosuspension‘ INTERNATIONAL JOURNAL OF PHARMACEUTICS vol. 408, 02 February 2011, pages 157 – 162
WO1996005177A1 * Aug 10, 1995 Feb 22, 1996 Jeong Seok Chae Novel pyrimidine derivatives and processes for the preparation thereof
WO2007064128A1 * Nov 28, 2006 Jun 7, 2007 Ki-Baik Hahm Composition for preventing or treating damages of the mucosa in the gastrointestinal tracts
WO2008078922A1 * Dec 21, 2007 Jul 3, 2008 Yuhan Corp Revaprazan-containing solid dispersion and process for the preparation thereof

 

Kundalini meditation: an amazing experience with therapeutic effect


Vertex Pharmaceuticals: Another FDA Orphan Drug Designation For Cystic Fibrosis


Orphan Druganaut Blog's avatarOrphan Druganaut Blog

On April 24th , Vertex Pharmaceuticals’ investigational drug VX-661 ((R)-1-(2,2-difluorobenzo [d][1,3] dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl) cyclopropanecarboxamide) receives FDA Orphan Drug Designation (ODD) for the treatment of Cystic Fibrosis (CF) :
FDA Orphan Drug Designation Database Record
Generic Name:(R)-1-(2,2-difluorobenzo [d][1,3] dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl) cyclopropanecarboxamide
Trade Name:n/a
Date Designated:04-24-2014
Orphan Designation:Treatment of cystic fibrosis
Orphan Designation Status:Designated
FDA Orphan Approval Status:Not FDA Approved for Orphan Indication
Sponsor: Vertex Pharmaceuticals Inc. 50 Northern Avenue Boston, MA 02210-1862 The sponsor address listed is the last reported by the sponsor to OOPD.

.

VX-661 is a Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) corrector. VX-661 is being studied in combination with Kalydeco (Ivacaftor) for patients who have the F508del mutation. VX-661 is currently recruiting participants for a Phase II clinical trial to evaluate the safety and efficacy of VX-661 in combination with Kalydeco in subjects with CF who are homozygous (have 2 copies) for the F508del CFTR…

View original post 173 more words

Radiation therapy to treat uterine cancer linked with increased risk of bladder cancer later in life


Lyranara.me's avatarLyra Nara Blog

Radiation therapy used to treat uterine cancer may increase a patient’s risk of developing bladder cancer. That is the conclusion of a recent study published in BJU International. The findings indicate the importance of monitoring patients for potential signs of bladder cancer to ensure early diagnosis and treatment.

In the United States, uterine cancer is the fourth most common cancer in women, with an estimated 49,560 women diagnosed in 2013. In addition to surgery, 38 percent ofpatients undergo pelvic radiation therapy to decrease uterine cancer recurrence. Studies have found that women treated with radiation therapy for uterine cancer, like men who received radiation therapy for prostate cancer, have an increased risk of developing bladder cancer later in life.

To investigate the issue, Guan Wu, MD, PhD, of the University of Rochester Medical Center, and his colleagues analyzed the records of 56,681 patients diagnosed with uterine cancer as their…

View original post 176 more words

ANTHONYFLOZIN………Find one if you can in this review


find here

http://medcheminternational.blogspot.in/p/flozin-series.html

1 TOFOGLIFLOZIN
2 SERGLIFLOZIN
3 DAPAGLIFLOZIN
4 IPRAGLIFLOZIN
5 EMPAGLIFLOZIN
6 LUSEOGLIFLOZIN
7 REMOGLIFLOZIN
8 ERTUGLIFLOZIN
9 SOTAGLIFLOZON

DR ANTHONY

BLOGS………

ALL ABOUT DRUGS,

WORLD DRUG TRACKER,

MEDICINAL CHEM INTERNATIONAL,

DRUG SYN INTERNATIONAL

SCALEUP OF DRUGS,

MEDICINAL CHEM INTERNATIONAL,

DRUG SYN INTERNATIONAL,

SCALEUP OF DRUGS, 

EUREKAMOMENTS

***/*

Fibroblasts could offer alternative to heart transplants


Lyranara.me's avatarLyra Nara Blog

Fibroblasts could offer alternative to heart transplants

Cardiac fibroblasts

Fibroblasts, cells long thought to be boring and irrelevant, could offer an alternative to heart transplants for patients with heart disease.  Researcher Dr Milena Furtado, and her team from the Australian Regenerative Medicine Institute (ARMI) at Monash University, found the heart cell fibroblast is a close relative to a cardiomyocyte, the cell responsible for a healthy beating heart.

In research published today in Circulation Research, Dr Furtado has found that cardiac fibroblasts are unique cells due to their genetic program, and will aid in the development of cell therapies for congenital heart disease and heart failure.

“Heart disease is still one of the major killers in our society and so far no effective therapeutic options are available. Our laboratory aims to understand how the various cell types present in a heart can improve the outcome of heart failure,’ Dr Furtado said.

“Fibroblasts were thought to act as…

View original post 199 more words

ALK Inhibitor CEP-28122


(1S,2S,3R,4R)-3-[5-Chloro-2-(S)-1-methoxy-7-morpholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]bicycle[2.2.1]hept-5-ene-2-carboxylic Acid Amide
(1S,2S,3R,4R)-3-((5-chloro-2-(((S)-1-methoxy-7-morpholino-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)amino)pyrimidin-4-yl)amino)bicyclo[2.2.1]hept-5-ene-2-carboxamide.
(1S,2S,3R,4R)-3-[5-Chloro-2-(S)-1-methoxy-7-morpholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]bicycle[2.2.1]hept-5-ene-2-carboxylic Acid Amide Methanesulfonic Acid Hydrochloride Salt
(CEP-28122)
 (l S,2S,3R,4R)-3-[5-Chloro-2-((R)-l-methoxy-7-morpholin-4-yl-6,7,8,9- tetrahydro-5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]- bicyclo[2.2.1]hept-5-ene-2-carboxylic acid amide

CAS:  1022958-60-6

Chemical Formula: C28H35ClN6O3

Molecular Weight: 539.06890

Elemental Analysis: C, 62.39; H, 6.54; Cl, 6.58; N, 15.59; O, 8.90

CEP-28122 is a Highly Potent and Selective Orally Active Inhibitor of Anaplastic Lymphoma Kinase with Antitumor Activity in Experimental Models of Human Cancers. (source: Mol Cancer Ther; 11(3); 670-9.)
CEP-28122 is used as an orally efficacious inhibitor of (ALK), analplastic lymphoma kinase, in the treatment of cancer.
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) member of the insulin receptor superfamily identified as part of the NPM–ALK fusion gene in anaplastic large cell lymphoma (ALCL) with a t(2;5) chromosomal translocation.(1) ALK, when fused with NPM, is constitutively activated and shown to be involved in proliferation and survival of a variety of human cancers.(2) The aberrant signaling of ALK resulting from rearrangements or mutations/gene amplification leads to an “oncogenic addiction” which can be targeted with kinase inhibitors.(3)
 Crizotinib is the first ALK inhibitor to be approved and has shown a clinical impact with patients that are highly refractory.(4) The need for novel ALK inhibitors to impact emergence of resistance mechanisms as well as to provide improved kinase selectivity profiles is of great importance. CEP-28122 is a selective, potent ALK inhibitor, demonstrating robust antitumor efficacy in tumor xenograft mouse models, which advanced into preclinical development.(5)
 It is a complex small molecule comprised of three core subunits, two of which contain one or more chiral centers

Various ALK inhibitors have been reported, such as indazoloisoquinolines (WO 2005/009389), thiazole amides and oxazole amides (WO 2005/097765), pyrrolopyrimidines (WO 2005080393), and pyrimidinediamines (WO 2005/016894).

WO 2008/051547 discloses fused bicyclic derivatives of 2,4-diaminopyrimidine as ALK and c-Met inhibitors. The lead drug candidate disclosed in the ‘547 application is CEP-28122, a potent ALK inhibitor with oral efficacy against SUP-M2 and Karpas-299 ALK-dependent tumors in mouse xenograft models. CEP-28122 progressed to IND- enabling studies until its development was terminated due to the unexpected occurrence of severe lung toxicity in CEP-28122-treated monke s.

Figure imgf000003_0001

CEP-28122

…………….caution
closest or analogues please check …………reader caution

Example 1047: (lS,2S,3R,4R)-3-[5-Chloro-2-(3-methoxy-7-moφholin-4-yl-6,7,8,9- tetrahydro-5H-benzocyclohepten-2-ylarnino)-pyrimidin-4-ylamino]- bicyclo[2.2.1]hept-5-ene-2-carboxylic acid amide (Single Diasteromer A) 1047a) (2-Hydroxymethyl-4-methoxy-phenyl)-methanol To a stirred suspension of Lithium tetrahydroaluminate (16.6 g, 0.436 mol) in Tetrahydrofuran (300 mL, 4 mol) at 0 °C under nitrogen was added dropwise a solution of 4-Methoxy-phthalic acid dimethyl ester (24.46 g, 0.1091 mol) in Tetrahydrofuran (100 mL, 1 mol). The reaction was stirred at 0 °C for 1 h then warmed to room temperature overnight. HPLC indicated no starting material present. Reaction was recooled at 0 °C and quenched with addition of water (125 mL) carefully dropwise, 1 N NaOH (100 mL) and water (125 mL). Evolution of gas was observed upon initial quenching with water. A white solid precipitated out of solution (aluminum salts). Following complete quenching of the reaction mixture, the aluminum salts were removed by filtration. The filtrate was diluted with ethyl acetate, washed with water, dried over magnesium sulfate, filtered and concentrated in vacuo to provide 17.80 grams (97%) of (2-Hydroxvmethyl-4-methoxy- phenyl)-methanol as a colorless oil.

1047b) 1 ,2-Bis-bromomethyl-4-methoxy-benzene

Using the procedure outlined in J. Am. Chem. Soc. 1994, 116, 10593 – 10600, (2-

Hydroxymethyl-4-methoxy-phenyl)-methanol (17.80 g, 0.1058 mol) was dissolved in

Chloroform (200 mL, 2 mol) and the reaction was treated with Phosphorus tribromide (60.2 g, 0.222 mol) dropwise over 6 hours. After stirring overnight at room temperature, the mixture was cooled at 0 °C and was treated with 50 mL of water. The reaction mixture was poured over saturated sodium bicarbonate, and organics were extracted with dichloromethane. Combined organics were dried over sodium sulfate, filtered and reduced en vacuo. The product, 16.0 grams (51%), was used without further purification.

1047c) 2-Methoxy-7-oxo-6,7,8,9-tetrahydro-5H-benzocycloh eptene-6,8-dicarboxylic acid diethyl ester

From an adapted procedure in Helvetic Chimica Acta, 2001, 84, 2051-2063, to a stirred solution of Tetra-n-butyl ammonium iodide (12.1 g, 0.0326 mol) in 0.6 M of Sodium bicarbonate in Water (300 mL) and Methylene chloride (130 mL, 2.1 mol) was added a solution of 1 ,2-Bis-bromomethyl-4-methoxy-benzene (16.00 g, 0.05442 mol) and 3- Oxopentanedioic acid, diethyl ester (14.31 g, 0.07075 mol) in Methylene chloride (40 mL, 0.6 mol). The solution was stirred vigorously at room temperature for -20 h. Saturated ammonium chloride solution was added to the reaction mixture. The product was extracted with ethyl acetate (3 X 100 mL). The ethyl acetate extracts were washed with water and brine, then dried over magnesium sulfate, filtered and concentrated in vacuo to a yellow oil. The oil was triturated with ether and a precipitate crashed out of solution and was removed by filtration (tetrabutyl ammonium salts). The filtrate was concentrated to an oil (20.0 grams, 100%) that was carried on to the next step without further purification. 1047d) 2-Methoxy-5,6,8,9-tetrahydro-benzocyclohepten-7-one

2-Methoxy-7-oxo-6,7,8,9-tetrahydro-5H-benzocycloheptene-6,8-dicarboxylic acid diethyl ester (18.2 g, 0.0544 mol) was dissolved in ethanol and the solution was treated with Potassium hydroxide (24.4 g, 0.435 mol) in Water (14O g, 7.6 mol). The reaction was then refluxed until HPLC showed consumption of starting material (~5 hours). The reaction was then acidified with IN HCl and the product was extracted with dichloromethane.

Organic extracts were dried over sodium sulfate, filtered and reduced. The crude mixture was filtered through a plug of silica rinsing with dichloromethane before purification. The crude mixture was purified by Isco flash column chromatography (Hexane/Ethyl Acetate). Combined fractions were reduced en vacuo to afford 6.0 grams (58%) of 2-Methoxy- 5,6,8,9-tetrahydro-benzocyclohepten-7-one.

1047e) 2-Methoxy-3-nitro-5,6,8,9-tetrahydro-benzocyclohepten-7-one and 2-Methoxy-l- nitro-5,6,8,9-tetrahydro-benzocyclohepten-7-one 2-Methoxy-5,6,8,9-tetrahydro-benzocyclohepten-7-one (6.00 g, 0.0315 mol) was dissolved in Acetonitrile (280 mL, 5.4 mol) and was added to a mixture of Trifiuoroacetic anhydride (13.4 mL, 0.0946 mol) in Acetonitrile at 0 °C. Potassium nitrate (3.19 g, 0.0315 mol) was then added and the reaction was allowed to warm to room temperature. When HPLC showed consumption of starting material, the mixture was poured over saturated sodium bicarbonate, and organics were extracted with ethyl acetate/dichloromethane. Combined organics were dried over sodium sulfate, filtered and reduced en vacuo. The crude mixture was purified by Isco flash column chromatography (Hexane/Ethyl Acetate). The gradient run was 0% EA-50% EA. Combined fractions were reduced en vacuo to afford 3.62 (49%) of 2-Methoxy-3-nitro-5,6,8,9-tetrahydro-benzocyclohepten-7-one and 1.80 grams (25%). 1047f) 4-(2-Methoxy-3-nitro-6,7,8,9-tetrahydro-5H-benzocyclohepten-7-yl)-morpholine 2-Methoxy-3-nitro-5,6,8,9-tetrahydro-benzocyclohepten-7-one (4.94 g, 0.0210 mol) in Methylene chloride (100 mL, 2 mol) was treated with Morpholine (18.30 g, 0.2100 mol) and then Acetic acid (12.61 g, 0.2100 mol). Two mass equivalents of powdered 4A molecular sieves were added and the mixture was heated to reflux and was allowed to stir for 4 hours. The solution was then cooled to room temp and Sodium triacetoxyborohydride (8.90 g, 0.0420 mol) was added. The reaction was then allowed to proceed until HPLC showed consumption of starting material. The reaction mixture was poured over saturated sodium bicarbonate, and organics were extracted with ethyl acetate/dichloromethane. Combined organics were dried over sodium sulfate, filtered and reduced en vacuo. The crude mixture was purified by Isco flash column chromatography (DCM/MeOH). Combined fractions were reduced en vacuo to afford 5.41 grams (84%) of 4-(2-Methoxy-3-nitro-6,7,8,9-tetrahydro-5H-benzocyclohepten-7-yl)-moφholine. 4-(2- Methoxy-l-nitro-6,7,8,9-tetrahydro-5H-benzocyclohepten-7-yl)-morpholine was made in an analogous manner using the same conditions described above. 1047g) 3-Methoxy-7-moφholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamine 4-(2-Methoxy-3-nitro-6,7,8,9-tetrahydro-5H-benzocyclohepten-7-yl)-moφholine (5.40 g, 0.0176 mol) was dissolved in Ethanol (100 mL, 2 mol) and the reaction mixture was carefully added to 10% Palladium on Carbon (0.750 g) under nitrogen in a Parr vessel. The reaction was then placed on a Parr shaker until uptake of hydrogen had ceased (~5 hours). Catalyst was filtered and the filtrate was reduced en vacuo to afford 4.10 grams (84%) of 3-Methoxy-7-moφholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2- ylamine. 2-Methoxy-7-moφholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-l – ylamine was made in an analogous fashion. The following intermediates were made in an analogous fashion as above utilizing the appropriate amine precursors: N*7*-(2,2-Difluoro-ethyl)-3-methoxy-6,7,8,9-tetrahydro- 5H-benzocycloheptene-2,7-diamine, 3-Methoxy-N*7*-(2-methoxy-ethyl)-6,7,8,9- tetrahydro-5H-benzocycloheptene-2,7-diamine, N*7*-(2,2-Difluoro-ethyl)-2-methoxy- 6,7,8,9-tetrahydro-5H-benzocycloheptene-l ,7-diamine, 2-(2-Amino-3-methoxy-6,7,8,9- tetrahydro-5H-benzocyclohepten-7-ylamino)-ethanol and 3-Methoxy-7-(4-methyl- piperazin-l-yl)-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamine.

1047h) (lS,2S,3R,4R)-3-[5-Chloro-2-(3-methoxy-7-moφholin-4-yl-6,7,8,9-tetrahydro- 5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]-bicyclo[2.2.1 ]hept-5-ene-2- carboxylic acid amide (Single Diasteromer A)

3-Methoxy-7-morpholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamine (880.0 mg, 0.003184 mol), (l S,2S,3R,4R)-3-(2,5-Dichloro-pyrimidin-4-ylamino)- bicyclo[2.2.1]hept-5-ene-2-carboxylic acid amide (952 mg, 0.00318 mol) and 4M of Hydrogen Chloride in 1 ,4-Dioxane (2 mL) were dissolved in 2-Methoxyethanol (30.0 mL, 0.380 mol) and the reaction was heated at 100 °C until HPLC showed consumption of starting material. The reaction mixture was poured over saturated sodium bicarbonate, and organics were extracted with ethyl acetate/dichloromethane. Combined organics were dried over sodium sulfate, filtered and reduced en vacuo. The crude residue was isolated and purified by Gilson prep HPLC as the first peak to elute to afford the desired product as a TFA salt. The TFA salt was taken up in dichloromethane and was poured over saturated sodium bicarbonate, and organics were extracted with ethyl acetate/dichloromethane. Combined organics were dried over sodium sulfate, filtered and reduced en vacuo to afford 439 mg (26%) of (lS,2S,3R,4R)-3-[5-Chloro-2-(3-methoxy-7-morpholin-4-yl- 6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]- bicyclo[2.2.1]hept-5-ene-2-carboxylic acid amide (Single Diasteromer A). LC/MS (ESI): 539.22. 1H NMR (400 MHz, DMSO, d6) δ 9.60 (m, IH), 8.12 (s, IH), 7.90 (s, IH), 7.79 (m, IH), 7.39 (s, IH), 6.98 (s, IH), 6.36 (m, IH), 6.16 (m, IH), 4.00 (m, 3H), 3.83 (s, 3H), 3.30 (m, 5H), 2.74 – 2.90 (m, 6H), 2.39 (m, 3H), 1.94 (d, IH, J = 4.80 Hz), 1.44 (m, 3H), 1.28 (m, IH), 1.04 (s, IH), 0.74 (s, IH).

………………………
PA[PER
Org. Process Res. Dev.201216 (1), pp 148–155
DOI: 10.1021/op200313v
Abstract Image
Evolution of the process strategies to prepare CEP-28122, an anaplastic lymphoma kinase (ALK) inhibitor, is presented. The initial medicinal chemistry route, used for the preparation of key supplies for biological screening, is reviewed. In addition, the process research and development of the final optimized process for manufacture of preclinical and clinical supplies is discussed. Details regarding a blocking group strategy for selective nitration; discovery of a one-pot transfer hydrogenation to effect a reductive amination, nitro group reduction, and dehalogenation; an enzymatic resolution of a critical intermediate; and the discovery of a novel, stable, in situ generated mixed mesylate hydrochloride salt of the API are disclosed.
(1S,2S,3R,4R)-3-[5-Chloro-2-(S)-1-methoxy-7-morpholin-4-yl-6,7,8,9-tetrahydro-5H-benzocyclohepten-2-ylamino)-pyrimidin-4-ylamino]bicycle[2.2.1]hept-5-ene-2-carboxylic Acid Amide Methanesulfonic Acid Hydrochloride Salt (CEP-28122)

 The solids were then dried to constant weight (50 mmHg, 50 °C), yielding 2.94 kg (4.37 mol, 87.1%) of CEP-28122 monomesylate/monohydrochloride with 97.4 A% chemical purity and 97% ee.
1H NMR (400 MHz, DMSO-d6) δ 10.6 (s, b, 1H), 9.93 (s, b, 1H), 9.58 (s, b, 1H), 8.36 (s, 1H), 8.06 (s, 1H), 7.86 (d, J = 7.28 Hz, 1H), 7.48 (s, 1H), 7.09 (d, J = 8.36 Hz, 1H), 6.39 (dd, J = 2.88, 5.56 Hz, 1H), 6.23 (dd, J = 2.92, 5.52 Hz, 1H), 3.93 (m, 6H), 3.69 (s, 3H), 3.69 (s, b, 1H), 3.58 (m, 2H), 3.29 (m, b, 4H), 3.17 (m, 2H), 2.94 (m, 3H), 2.77 (t, J = 12.04 Hz, 1H), 2.53 (d, J = 8.00 Hz, 2H), 2.49 (d, b, J = 13.68 Hz, 2H), 2.34 (s, 3H), 1.96 (d, J = 8.80 Hz, 1H), 1.46 (m, b, 3H), 1.01 (d, J = 6.24 Hz, 1H).
Anal. Calcd for C29H40N6O6SCl2 (671.64): C, 51.86; H, 6.00; N, 12.51; Cl, 10.56. Found: C, 51.75; H, 6.07; N, 12.37; Cl, 10.57. Heavy metals <20 ppm.
ref……………
  1. MorrisS. W.; KirsteinM. N.; ValentineM. B.; DittmerK. G.; ShapiroD. N.; SaltmanD. L.; LookA. T. Science 19942631281– 1284
  2. GrandeE.; BolosM.; ArriolaE. Mol. Cancer Ther. 201110 ( 4569– 571

  3. ShawA. T.; SolomonB. Clin. Cancer Res. 2011172081– 2086

  4. MosseY. P.; WoodA.; MarisJ. M. Clin. Cancer Res. 2009155608– 5614

    Gingrich, D. et al: J. Med Chem, 55, 4580 (2012);…