New Drug Approvals
Follow New Drug Approvals on WordPress.com

FLAGS AND HITS

Flag Counter
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO

Archives

Categories

Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Researchgate

Anthony Melvin Crasto Dr.

  Join me on Facebook FACEBOOK   ...................................................................Join me on twitter Follow amcrasto on Twitter     ..................................................................Join me on google plus Googleplus

MYSELF

DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai) , INDIA 36Yrs Exp. in the feld of Organic Chemistry,Working for AFRICURE PHARMA as ADVISOR earlier with GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.Million hits on google, NO ADVERTISEMENTS , ACADEMIC , NON COMMERCIAL SITE, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution, ........amcrasto@gmail.com..........+91 9323115463, Skype amcrasto64 View Anthony Melvin Crasto Ph.D's profile on LinkedIn Anthony Melvin Crasto Dr.

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 →

Recent Posts

GNE-272


SCHEMBL17794706.png

GNE-272

(S)-1-(3-((2-fluoro-4-(1-methyl-1H-pyrazol-4- yl)phenyl)amino)-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1H-pyrazolo[4,3-c]pyridin- 5(4H)-yl)ethanone

1-[3-[2-fluoro-4-(1-methylpyrazol-4-yl)anilino]-1-[(3S)-oxolan-3-yl]-6,7-dihydro-4H-pyrazolo[4,3-c]pyridin-5-yl]ethanone

CAS 1936428-93-1

Molecular Formula: C22H25FN6O2
Molecular Weight: 424.471303 g/mol

GENENTECH, INC. [US/US]; 1 DNA Way South San Francisco, California 94080-4990 (US).
CONSTELLATION PHARMACEUTICALS, INC. [US/US]; 215 First Street Suite 200 Cambridge, Massachusetts 02142 (US)

ROMERO, F. Anthony; (US).
MAGNUSON, Steven; (US).
PASTOR, Richard; (US).
TSUI, Vickie Hsiao-Wei; (US).
MURRAY, Jeremy; (US).
CRAWFORD, Terry; (US).
ALBRECHT, Brian, K.; (US).
COTE, Alexandre; (US).
TAYLOR, Alexander, M.; (US).
LAI, Kwong Wah; (CN).
CHEN, Kevin, X.; (CN).
BRONNER, Sarah; (US).
ADLER, Marc; (US).
EGEN, Jackson; (US).
LIAO, Jiangpeng; (CN).
WANG, Fei; (CN).
CYR, Patrick; (US).
ZHU, Bing-Yan; (US).
KAUDER, Steven; (US)

Chromatin is a complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended) forms. The major components of chromatin are DNA and proteins. Histones are the chief protein components of chromatin, acting as spools around which DNA winds. The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control expression and DNA replication. The chromatin structure is controlled by a series of post-translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the “histone tails” which extend beyond the core nucleosome structure. Histone tails tend to be free for protein-protein interaction and are also the portion of the histone most prone to post-translational modification. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, and SUMOylation. These epigenetic marks are written and erased by specific enzymes that place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription.

Of all classes of proteins, histones are amongst the most susceptible to post-translational modification. Histone modifications are dynamic, as they can be added or removed in response to specific stimuli, and these modifications direct both structural changes to chromatin and alterations in gene transcription. Distinct classes of enzymes, namely histone acetyltransferases (HATs) and histone deacetylases (HDACs), acetylate or de-acetylate specific histone lysine residues (Struhl K., Genes Dev., 1989, 12, 5, 599-606).

Bromodomains, which are approximately 1 10 amino acids long, are found in a large number of chromatin-associated proteins and have been identified in approximately 70 human proteins, often adjacent to other protein motifs (Jeanmougin F., et al., Trends Biochem. Sc , 1997, 22, 5, 151-153; and Tamkun J.W., et al., Cell, 1992, 7, 3, 561-572).

Interactions between bromodomains and modified histones may be an important mechanism underlying chromatin structural changes and gene regulation. Bromodomain-containing proteins have been implicated in disease processes including cancer, inflammation and viral replication. See, e.g., Prinjha et al,, Trends Pharm. Sci., 33(3):146-153 (2012) and Muller et al , Expert Rev. , 13 (29): 1 -20 (September 201 1 ).

Cell-type specificity and proper tissue functionality requires the tight control of distinct transcriptional programs that are intimately influenced by their environment.

Alterations to this transcriptional homeostasis are directly associated with numerous disease states, most notably cancer, immuno-inflammation, neurological disorders, and metabolic diseases. Bromodomains reside within key chromatin modifying complexes that serve to control distinctive disease-associated transcriptional pathways. This is highlighted by the observation that mutations in bromodomain-containing proteins are linked to cancer, as well as immune and neurologic dysfunction. Hence, the selective inhibition of bromodomains across a specific family, such as the selective inhibition of a bromodomain of CBP/EP300, creates varied opportunities as novel therapeutic agents in human dysfunction.

There is a need for treatments for cancer, immunological disorders, and other

CBP/EP300 bromodomain related diseases.

PATENT

WO-2016086200

Scheme 1

Scheme 2

Scheme 3

Scheme 4

General procedure for Intermediates A & B

Intermediate A

Intermediate

General procedure for Intermediates F & G

Intermediate F

Intermediate G

Step 1:

(R)-tetrahydrofuran-3-yI methanesulfonate

To a solution of (^)-tetrahydrofuran-3-ol (25 g, 253.7 mmol) in DCM (250 mL) at 0 °C was added triethylamine (86 g, 851.2 mmol) and mesyl chloride (39 g, 340.48 mmol) dropwise. The mixture was stirred at room temperature for 12 h. The reaction was quenched with water (100 mL) and extracted with DCM (100 mL x 2). The combined organic layers were dried over anhydrous Na2S04, filtered and concentrated in vacuo to give the title compound (47 g, 99%) as a brown oil. Ή NMR (400 MHz, CDC13) δ 5.35 – 5.27 (m, 1H), 4.05 – 3.83 (m, 4H), 3.04 (s, 3 H), 2.28 – 2.20 (m, 2 H).

Step 2:

(S)-tert-butyl 3-bromo-l-(tetrahydrofuran-3-yI)-6,7-dihydro-li/-pyrazolo[43- c] pyridine-5(4H)-carboxylate

To a solution of tert-butyl 3-bromo-6,7-dihydro-lH-pyrazolo[4,3-c]pyridine-5(4H)-carboxylate (Intermediate A, 24.8 g, 82 mmol) in DMF (200 mL) was added Cs2C03 (79 g, 246 mmol) and (/?)-tetrahydrofuran-3-yl methanesulfonate (17.4 g, 98 mmol). The mixture was heated to 80 °C for 12 h. After cooling the reaction to room temperature, the mixture was concentrated in vacuo. The crude residue was purified by silica gel chromatography

(petroleum ether / EtOAc = from 10 : 1 to 3 : 1) to give the title compound (Intermediate F, 50 g, 71 %) as a yellow oil. Ή NMR (400 MHz, DMSO-i ) δ 4.97 – 4.78 (m, 1H), 4.13 (s, 2H), 3.98 – 3.86 (m, 2H), 3.81 – 3.67 (m, 2H), 3.56 (t, J= 5.6 Hz, 2H), 2.68 (t, J= 5.6 Hz, 2H), 2.33 – 2.08 (m, 2H), 1.38 (s, 9H).

Step 3:

(5)-l-(3-bromo-l-(tetrahydrofuran-3-yl)-6,7-dihydro-lH-pyrazoIo[4,3-c]pyridin-5(4//)- yl)ethanone

To a solution of (S)-tert-buty\ 3-bromo- 1 -(tetrahydrofuran-3-yl)-6,7-dihydro-lH-pyrazolo [4,3 -c]pyridine-5(4H)-carboxy late (29 g, 78 mmol) in DCM (300 mL) was added trifluroacetic acid (70 mL) dropwise. The mixture was stirred at room temperature for 2 h. The solvent was concentrated in vacuo and the crude residue was re -dissolved in DMF (100 mL). The mixture was cooled to 0 °C before triethylamine (30 g, 156 mmol) and acetic anhydride (8.7 g, 86 mmol) were added dropwise. The mixture was stirred at room temperature for an additional 2 h. The reaction was quenched with water (200 mL) at 0 °C and extracted with EtOAc (150 mL x 3). The combined organic layers were dried over anhydrous Na2S0 , filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (DCM / MeOH = 30 : 1) to give the title compound (Intermediate G, 21.3 g, 87%) as a white solid. lH NMR (400 MHz, CDC13) δ 4.78 – 4.67 (m, 1H), 4.45 -4.29 (m, 2H), 4.15 – 4.06 (m, 2H), 3.96 – 3.92 (m, 2H), 3.88 – 3.70 (m, 2H), 2.71 – 2.67 (m, 2H), 2.38 – 2.34 (m, 2H), 2.16 (s, 3H).

PATENT

US-20160158207

Example 300 1-[3-[2-fluoro-4-(1-methylpyrazol-4- yl)anilino]-1-[(3S)-tetrahydrofuran-3- yl]-6,7-dihydro-4H-pyrazolo[4,3- c]pyridin-5-yl]ethanone
1H NMR (400 MHz, DMSO- d6) δ 8.03 (s, 1H), 7.83-7.68 (m, 3H), 7.36-7.33 (m, 1H), 7.32-7.21 (m, 1H), 4.88- 4.84 (m, 1H), 4.40-4.33 (m, 2H), 4.03-3.99 (m, 2H), 3.84- 3.67 (m, 7H), 2.79-2.64 (m, 2H), 2.26-2.21 (m, 2H), 2.08-2.05 (m, 3H) 425

General Procedure for Intermediates F & G


Step 1

(R)-tetrahydrofuran-3-yl methanesulfonate


      To a solution of (R)-tetrahydrofuran-3-ol (25 g, 253.7 mmol) in DCM (250 mL) at 0° C. was added triethylamine (86 g, 851.2 mmol) and mesyl chloride (39 g, 340.48 mmol) dropwise. The mixture was stirred at room temperature for 12 h. The reaction was quenched with water (100 mL) and extracted with DCM (100 mL×2). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give the title compound (47 g, 99%) as a brown oil. 1H NMR (400 MHz, CDCl3) δ 5.35-5.27 (m, 1H), 4.05-3.83 (m, 4H), 3.04 (s, 3H), 2.28-2.20 (m, 2H).

Step 2

(S)-tert-butyl 3-bromo-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1H-pyrazolo[4,3-c]pyridine-5(4H)-carboxylate


      To a solution of tert-butyl 3-bromo-6,7-dihydro-1H-pyrazolo[4,3-c]pyridine-5(4H)-carboxylate (Intermediate A, 24.8 g, 82 mmol) in DMF (200 mL) was added Cs2CO3 (79 g, 246 mmol) and (R)-tetrahydrofuran-3-yl methanesulfonate (17.4 g, 98 mmol). The mixture was heated to 80° C. for 12 h. After cooling the reaction to room temperature, the mixture was concentrated in vacuo. The crude residue was purified by silica gel chromatography (petroleum ether/EtOAc=from 10:1 to 3:1) to give the title compound (Intermediate F, 50 g, 71%) as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ 4.97-4.78 (m, 1H), 4.13 (s, 2H), 3.98-3.86 (m, 2H), 3.81-3.67 (m, 2H), 3.56 (t, J=5.6 Hz, 2H), 2.68 (t, J=5.6 Hz, 2H), 2.33-2.08 (m, 2H), 1.38 (s, 9H).

Step 3

(S)-1-(3-bromo-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1H-pyrazolo[4,3-c]pyridin-5(4H)-yl)ethanone


      To a solution of (S)-tert-butyl 3-bromo-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1H-pyrazolo[4,3-c]pyridine-5(4H)-carboxylate (29 g, 78 mmol) in DCM (300 mL) was added trifluroacetic acid (70 mL) dropwise. The mixture was stirred at room temperature for 2 h. The solvent was concentrated in vacuo and the crude residue was re-dissolved in DMF (100 mL). The mixture was cooled to 0° C. before triethylamine (30 g, 156 mmol) and acetic anhydride (8.7 g, 86 mmol) were added dropwise. The mixture was stirred at room temperature for an additional 2 h. The reaction was quenched with water (200 mL) at 0° C. and extracted with EtOAc (150 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (DCM/MeOH=30:1) to give the title compound (Intermediate G, 21.3 g, 87%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 4.78-4.67 (m, 1H), 4.45-4.29 (m, 2H), 4.15-4.06 (m, 2H), 3.96-3.92 (m, 2H), 3.88-3.70 (m, 2H), 2.71-2.67 (m, 2H), 2.38-2.34 (m, 2H), 2.16 (s, 3H).

OTHER ISOMER

Example 299 1-[3-[2-fluoro-4-(1-methylpyrazol-4- yl)anilino]-1-[(3R)-tetrahydrofuran-3- yl]-6,7-dihydro-4H-pyrazolo[4,3- c]pyridin-5-yl]ethanone
1H NMR (400 MHz, DMSO- d6) δ 8.03 (s, 1H), 7.83-7.67 (m, 3H), 7.39-7.34 (m, 1H), 7.26-7.21 (m, 1H), 4.87- 4.77 (m, 1H), 4.41-4.34 (m, 2H), 4.02-3.97 (m, 2H), 3.83 (s, 3H), 3.81-3.67 (m, 4H), 2.77-2.66 (m, 2H), 2.26- 2.22 (m, 2H), 2.08-2.05 (m, 3H) 425

PAPER

Abstract Image

The single bromodomain of the closely related transcriptional regulators CBP/EP300 is a target of much recent interest in cancer and immune system regulation. A co-crystal structure of a ligand-efficient screening hit and the CBP bromodomain guided initial design targeting the LPF shelf, ZA loop, and acetylated lysine binding regions. Structure–activity relationship studies allowed us to identify a more potent analogue. Optimization of permeability and microsomal stability and subsequent improvement of mouse hepatocyte stability afforded 59 (GNE-272, TR-FRET IC50 = 0.02 μM, BRET IC50 = 0.41 μM, BRD4(1) IC50 = 13 μM) that retained the best balance of cell potency, selectivity, and in vivo PK. Compound 59 showed a marked antiproliferative effect in hematologic cancer cell lines and modulates MYC expression in vivo that corresponds with antitumor activity in an AML tumor model.

Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of CBP/EP300

Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, People’s Republic of China
§ Constellation Pharmaceuticals, Inc. 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
J. Med. Chem., Article ASAP
*Phone: +1-650-467-6384. E-mail: romero.frank@gene.com.

UNDESIRED R ISOMER

In a similar procedure to59, the title compound was prepared from (S)-tetrahydrofuran-3-yl
methanesulfonate and purified by Prep-TLC (DCM / MeOH = 15 : 1) to give the title
compound as a light yellow solid.

1H NMR (400 MHz, CDCl3) δ 7.76–7. 42 (m,1H), 7.68 (s, 1H), 7.53 (s, 1H), 7.20–7.12 (m, 2H), 5.86–5.77 (m, 1H), 4.79–4.69 (m, 1H),4.47–4.29 (m, 2H), 4.25–4.08 (m, 2H), 4.06–3.72 (m, 4H), 3.99 (s, 3H), 2.76–2.65 (m, 2H),
2.49–2.28 (m, 2H), 2.25–2.12 (m, 3H).

13C NMR (100 MHz, CDCl3) δ 169.81, 169.36,151.71 (d, J = 238.9 Hz), 145.51, 144.64, 137.83, 136.32, 135.89, 126.35, 121.41, 116.44 (d,J = 26.0 Hz), 111.88, 103.09 (d, J = 24.0 Hz), 71.94, 68.10, 57.65, 43.24, 42.24, 39.02, 37.83,32.49, 22.01.

LCMS M/Z (M+H) 425.

[α]27D +8.8 (c 0.78, CHCl3, 99% ee).

DESIRED S ISOMER

(S)-1-(3-((2-fluoro-4-(1-methyl-1H-pyrazol-4- yl)phenyl)amino)-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1H-pyrazolo[4,3-c]pyridin- 5(4H)-yl)ethanone

aReagents and conditions: (a) 4-bromo-2-fluoro-1-isothiocyanato-benzene, KOtBu, THF, rt (b) CH3I, 40 °C, 51%; (c) hydrazine monohydrate, EtOH, 85 °C; 96%; (d) 1-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole, dioxane / water, Na2CO3, Pd(dppf)Cl2, 100 °C, 63%; (e) (R)-tetrahydrofuran-3-yl methanesulfonate, Cs2CO3, DMF, 90 oC, 42%.

The crude residue was purified by silica gel chromatography (DCM / MeOH = 100:1) to give (S)-1-(3-((2-fluoro-4-(1- methyl-1H-pyrazol-4-yl)phenyl)amino)-1-(tetrahydrofuran-3-yl)-6,7-dihydro-1Hpyrazolo[4,3-c]pyridin-5(4H)-yl)ethanone as a light yellow solid.

1H NMR (400 MHz, CDCl3) δ 7.76–7.72 (m, 1H), 7.68 (s, 1H), 7.53 (s, 1H), 7.20–7.12 (m, 2H), 5.86–5.77 (m, 1H), 4.79–4.69 (m, 1H), 4.47–4.29 (m, 2H), 4.25–4.08 (m, 2H), 4.06– 3.72 (m, 4H), 3.99 (s, 3H), 2.76–2.65 (m, 2H), 2.49–2.28 (m, 2H), 2.25–2.12 (m, 3H).

13C NMR (100 MHz, CDCl3) δ 169.8, 169.4, 151.7 (d, J = 238.9 Hz), 145.5, 144.64, 137.83, 136.3, 135.9, 126.4, 121.4, 116.4 (d, J = 26.0 Hz), 111.9, 103.1 (d, J = 24.0 Hz), 71.9, 68.1, 57.7, 43.2, 42.2, 39.0, 37.8, 32.5, 22.0.

LCMS m/z (M+H) 425.

[α]27 D -11.0 (c 1.0, CHCl3, 99% ee).

HRMS m/z 425.2093 (M + H+ , C22H25FN6O2, requires 425.2057).

//////////GNE-272, Genentech, CBP, EP300, cancer, immune system regulation,  1936428-93-1

[H][C@@]1(CCOC1)N1N=C(NC2=C(F)C=C(C=C2)C2=CN(C)N=C2)C2=C1CCN(C2)C(C)=O

Identifying “green chemistry” industrialisation barriers through case-studies


DR ANTHONY MELVIN CRASTO Ph.D's avatarGreen Chemistry International

Nitesh Mehta

Nitesh Mehta

Convenor of Industrial Green Chemistry World and Founder – Director of Newreka Green Synth Technologies Pvt Ltd

nitesh.mehta@newreka.co.in

Identifying “green chemistry” industrialisation barriers through case-studies
– Mr. Nitesh Mehta, Founder Director, Newreka Green Synth Technologies Pvt. Ltd., India

View presentation

Image result for waitThe presentation will load below


///////green chemistry, industrialisation barriers,  case-studies, Nitesh Mehta, Founder Director, Newreka Green Synth Technologies Pvt Ltd, India

View original post

Consumption of a bioactive compound from Neem plant could significantly suppress development of prostate cancer


(From left to right) Principal Investigator Associate Professor Gautam Sethi and NUS PhD candidate Ms Zhang Jingwen from the Department of Pharmacology at the NUS Yong Loo Lin School of Medicine led a research which found that a bioactive compound from the neem plant could significantly suppress development of prostate cancer.

Credit: National University of Singapore

Image result for SINGAPORE ANIMATED FLAG

Image result for National University of Singapore

 

Image result for nimbolideImage result for nimbolide

Date:September 29, 2016Source:National University of SingaporeSummary:Oral administration of nimbolide, over 12 weeks shows reduction of prostate tumor size by up to 70 per cent and decrease in tumor metastasis by up to 50 per cent, report investigators.

Nimbolide.png

Nimbolide; NSC309909; NSC 309909; Methyl[8-(furan-3-yl)-2a,5a,6a,7-tetramethyl-2,5-dioxo-2a,5a,6,6a,8,9,9a,10a,10b,10c-decahydro-2h,5h-cyclopenta[d]naphtho[2,3-b:1,8-b’c’]difuran-6-yl]acetate; CCRIS 5723;

CAS 25990-37-8;
Molecular Formula: C27H30O7
Molecular Weight: 466.5229 g/mol

Oral administration of nimbolide, over 12 weeks shows reduction of prostate tumor size by up to 70 per cent and decrease in tumor metastasis by up to 50 per cent

A team of international researchers led by Associate Professor Gautam Sethi from the Department of Pharmacology at the Yong Loo Lin School of Medicine at the National University of Singapore (NUS) has found that nimbolide, a bioactive terpenoid compound derived from Azadirachta indica or more commonly known as the neem plant, could reduce the size of prostate tumor by up to 70 per cent and suppress its spread or metastasis by half.

Prostate cancer is one of the most commonly diagnosed cancers worldwide. However, currently available therapies for metastatic prostate cancer are only marginally effective. Hence, there is a need for more novel treatment alternatives and options.

“Although the diverse anti-cancer effects of nimbolide have been reported in different cancer types, its potential effects on prostate cancer initiation and progression have not been demonstrated in scientific studies. In this research, we have demonstrated that nimbolide can inhibit tumor cell viability — a cellular process that directly affects the ability of a cell to proliferate, grow, divide, or repair damaged cell components — and induce programmed cell death in prostate cancer cells,” said Assoc Prof Sethi.

Image result for National University of Singapore

Nimbolide: promising effects on prostate cancer

Cell invasion and migration are key steps during tumor metastasis. The NUS-led study revealed that nimbolide can significantly suppress cell invasion and migration of prostate cancer cells, suggesting its ability to reduce tumor metastasis.

The researchers observed that upon the 12 weeks of administering nimbolide, the size of prostate cancer tumor was reduced by as much as 70 per cent and its metastasis decreased by about 50 per cent, without exhibiting any significant adverse effects.

“This is possible because a direct target of nimbolide in prostate cancer is glutathione reductase, an enzyme which is responsible for maintaining the antioxidant system that regulates the STAT3 gene in the body. The activation of the STAT3 gene has been reported to contribute to prostate tumor growth and metastasis,” explained Assoc Prof Sethi. “We have found that nimbolide can substantially inhibit STAT3 activation and thereby abrogating the growth and metastasis of prostate tumor,” he added.

The findings of the study were published in the April 2016 issue of the scientific journal Antioxidants & Redox Signaling. This work was carried out in collaboration with Professor Goh Boon Cher of Cancer Science Institute of Singapore at NUS, Professor Hui Kam Man of National Cancer Centre Singapore and Professor Ahn Kwang Seok of Kyung Hee University.

Neem — The medicinal plant

The neem plant belongs to the mahogany tree family that is originally native to India and the Indian sub-continent. It has been part of traditional Asian medicine for centuries and is typically used in Indian Ayurvedic medicine. Today, neem leaves and bark have been incorporated into many personal care products such as soaps, toothpaste, skincare and even dietary supplements.

Image result for National University of Singapore

Future Research

The team is looking to embark on a genome-wide screening or to perform a large-scale study of proteins to analyse the side-effects and determine other potential molecular targets of nimbolide. They are also keen to investigate the efficacy of combinatory regimen of nimbolide and approved drugs such as docetaxel and enzalutamide for future prostate cancer therapy.



Journal Reference:

  1. Jingwen Zhang, Kwang Seok Ahn, Chulwon Kim, Muthu K. Shanmugam, Kodappully Sivaraman Siveen, Frank Arfuso, Ramar Perumal Samym, Amudha Deivasigamanim, Lina Hsiu Kim Lim, Lingzhi Wang, Boon Cher Goh, Alan Prem Kumar, Kam Man Hui, Gautam Sethi. Nimbolide-Induced Oxidative Stress Abrogates STAT3 Signaling Cascade and Inhibits Tumor Growth in Transgenic Adenocarcinoma of Mouse Prostate Model. Antioxidants & Redox Signaling, 2016; 24 (11): 575 DOI:10.1089/ars.2015.6418

Image result for nimbolide

A PAPER

Image result for nimbolide

NIMBOLIDE 1

http://pubs.rsc.org/en/content/articlelanding/2015/ra/c5ra16071e#!divAbstract

Nimbolide (1): Pale yellow crystals; C27H30O7;

FT-IR (KBr, υmax, cm -1): 2978, 1778, 1730, 1672, 1433, 1296, 1238, 1192, 1153, 1069, 951, 827, 750;

1H NMR (500 MHz, CDCl3) δH: 7.32 (t, J = 1.5 Hz, 1H), 7.28 (d, J = 9.5 Hz, 1H), 7.22 (s, 1H), 6.25 (m, 1H), 5.93 (d, J = 10.0 Hz, 1H), 5.53 (m, 1H), 4.62 (dd, J = 3.67 Hz, 12 .5 Hz, 1H), 4.27 (d, J = 3.5 Hz, 1H), 3.67 (d, J = 9.0 Hz, 1H), 3.54 (s, 3H), 3.25 (dd, J = 5.0 Hz, 16.25 Hz, 1H), 3.19 (d, J = 12.5 Hz, 1H), 2.73 (t, J = 5.5 Hz, 1H), 2.38 (dd, J = 5.5 Hz, 16.25 Hz, 1H), 2.22 (dd, J = 6.5 Hz, 12.0 Hz, 1H), 2.10 (m, 1H), 1.70 (s, 3H), 1.47 (s, 3H), 1.37 (s, 3H), 1.22 (s, 3H);

13C NMR (125 MHz, CDCl3) δC: 200.8 (CO), 175.0 (COO), 173.0 (COO), 149.6 (CH), 144.8 (C), 143.2 (CH), 138.9 (CH), 136.4 (C), 131.0 (CH), 126.5 (C), 110.3 (CH), 88.5 (CH), 82.9 (CH), 73.4 (CH), 51.8 (OCH3), 50.3 (C), 49.5 (CH), 47.7 (CH), 45.3 (C), 43.7 (C), 41.2 (CH2), 41.1 (CH), 32.1 (CH2), 18.5 (CH3), 17.2 (CH3), 15.2 (CH3), 12.9 (CH3);

HR-MS (m/z): 467.20795 [(M+H)+ ].

Content Page No 1 1H NMR spectrum of nimbolide S1 2 13C NMR spectrum of nimbolide S2 3 Mass spectrum of nimbolide

Dr Gautam Sethi

phcgs@nus.edu.sg
Tel.: (65)6516 3267
Fax: (65)6873 7690

Academic Qualifications
BSc. Chem. (Hons) 1998 Banaras Hindu University, Varanasi, India.
MSc. Biochemistry 2000 Banaras Hindu University, Varanasi, India.
Ph.D. Biotechnology 2004 Banaras Hindu University, Varanasi, India.
Appointments to Date
Assistant
Professor
2008-date Department of Pharmacology, National University of Singapore, Singapore
Postdoctoral Fellow 2004-2007 Department of Experimental Therapeutics,
The University of Texas.
MD Anderson Cancer Center, Houston TX USA.
Senior Research Fellow 2002-2004 (CSIR-NET) at School of Biotechnology,
Banaras Hindu University, Varanasi, India.
Junior Research Fellow 2000-2002 (CSIR-NET) at School of Biotechnology, Banaras Hindu University, Varanasi, India.
Honours and Awards
2007 Ramalingaswamy fellowship from Department of Biotechnology, Government of India for outstanding research contributions in the field of Cancer Biology.
2002 Senior Research Fellowship award, Council of Scientific and Industrial Research, New Delhi, India.
2000 Junior Research Fellowship award, Council of Scientific and Industrial Research, New Delhi, India.
Research Interests
Selected Publications
Reviews and Book Chapters

 

Image result for SINGAPORE ANIMATED FLAG

/////////NIMBOLIDE, CANCER, NEEM, PROSTRATE, National University of Singapore, Gautam Sethi

CC1=C2C(CC1C3=COC=C3)OC4C2(C(C5(C6C4OC(=O)C6(C=CC5=O)C)C)CC(=O)OC)C

Green Solvent – A sustainable option – Dr. Denis Prat, SANOFI, France


DR ANTHONY MELVIN CRASTO Ph.D's avatarGreen Chemistry International

Prof. Andrea Larson

Dr. Denis Prat

Head of Process Safety & Automated Chemistry Chemistry & Biotechnology Development, SANOFI, France

Green Solvent – A sustainable option
– Dr. Denis Prat, Head of Process Safety & Environment, Chemistry & Biochemistry, SANOFI, France

View presentation

/////////Green Solvent, sustainable option, Denis Prat, SANOFI, France

View original post

Novel Autotaxin Inhibitors for the Treatment of Osteoarthritis Pain from Lilly Research Laboratories


SCHEMBL15875396.png

str1Figure imgf000023_0002

2-(2-(1H-1,2,3-triazol-5-yl)ethoxy)-1-(2-((2,3-dihydro-1H-inden-2-yl)amino)-5,7-dihydro-6Hpyrrolo[3,4-d]pyrimidin-6-yl)ethan-1-one

l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

CAS 1619971-30-0

1-[2-(2,3-dihydro-1H-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]-2-[2-(1H-1,2,3-triazol-4-yl)ethoxy]ethanone;
Molecular Formula: C21H23N7O2
Molecular Weight: 405.45302 g/mol

US2014200231

Scheme A

Scheme B

Scheme C

VI

Scheme E

Autotaxin is an enzyme reported to be the source of lysophosphatidic acid (LPA) which up-regulates pain-related proteins through one if its cognate receptors, LPAi. LPA is an intracellular lipid mediator which influences a multiplicity of biological and biochemical processes. Targeted inhibition of autotaxin-mediated LPA biosynthesis may provide a novel mechanism to prevent nerve injury-induced neuropathic pain.

Compounds that inhibit autotaxin are desired to offer a potential treatment option for patients in need of treatment for pain.

Pain associated with osteoarthritis (OA) is reported to be the primary symptom leading to lower extremity disability in OA patients. Over 20 million Americans have been diagnosed with OA, the most common of the arthropathies. The currently approved treatments for OA pain may be invasive, lose efficacy with long term use, and may not be appropriate for treating all patients. Additional treatment options for patients suffering from pain associated with OA are desired. Compounds that inhibit autotaxin represent another possible treatment option for patients with pain associated with OA.

U.S. Patent 7,524,852 (‘852) discloses substituted bicyclic pyrimidine derivatives as anti-inflammatory agents.

PCT/US2011/048477 discloses indole compounds as autotoxin inhibitors.

There is a need for novel compounds that provide autotaxin inhibition. The present invention provides novel compounds which are autotaxin inhibitors. The present invention provides certain novel compounds that inhibit the production of LPA.

Autotaxin inhibitor compounds are desired to provide treatments for autotaxin mediated conditions, such as pain and pain associated with OA.

PAPER

Abstract Image

In an effort to develop a novel therapeutic agent aimed at addressing the unmet need of patients with osteoarthritis pain, we set out to develop an inhibitor for autotaxin with excellent potency and physical properties to allow for the clinical investigation of autotaxin-induced nociceptive and neuropathic pain. An initial hit identification campaign led to an aminopyrimidine series with an autotaxin IC50 of 500 nM. X-ray crystallography enabled the optimization to a lead compound that demonstrated favorable potency (IC50 = 2 nM), PK properties, and a robust PK/PD relationship.

Image result for Lilly Research Laboratories

Novel Autotaxin Inhibitors for the Treatment of Osteoarthritis Pain: Lead Optimization via Structure-Based Drug Design

Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, Indiana 46285, United States
ACS Med. Chem. Lett., 2016, 7 (9), pp 857–861
DOI: 10.1021/acsmedchemlett.6b00207
*E-mail: jonessp@lilly.com. Tel: +1-317-277-5543.

http://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.6b00207

Spencer Jones

Spencer Jones

Senior Research Scientist at Eli Lilly and Company

2-(2-(1H-1,2,3-triazol-5-yl)ethoxy)-1-(2-((2,3-dihydro-1H-inden-2-yl)amino)-5,7-dihydro-6Hpyrrolo[3,4-d]pyrimidin-6-yl)ethan-1-one (9)

………… Purified the resulting residue by silica gel chromatography (gradient elution: 0-9% methanol in ethyl acetate ) to give the title compound……..

1H NMR (400 MHz, CDCl3): 60:40 mixure of rotamers * indicates minor rotamer δ 8.18 (bs, 0.6H), *8.13 (bs, 0.4H), 7.49 (s, 1H), 7.21-7.09 (m, 4 H), 5.70-5.50 (m, 1H), 4.87-4.78 (m, 1H), 4.75 (s, 1.2H), *4.67 (s, 0.8H), 4.64 (s, 1.2H) *4.53 (s, 0.8H), *4.30 (s, 0.8H), 4.28 (s, 1.2H), 3.93 (t, J = 5.6 Hz, 2H), 3.43 (dd, J = 16.2, 7.1 Hz, 2H), 3.10 (t, J = 5.6 Hz, 2H), 2.89 (dd, J = 16.2, 4.9 Hz, 2H).

13C NMR (400 MHz, CDCl3): * indicates minor δ *169.3, 16 169.2, 167.0, *166.8, *162.4, 162.2, 152.8, *152.3, 141.1, 137.8, 130.9, 126.7, 124.9, 115.9, 69.8, 69.3, *69.0, 52.7, *52.5, 51.2, 49.0, *47.9, 40.1, 24.7.

LC/MS (ESI+ ): (m/z) 406 (C21H24N7O2 = (M+1)+ ).

PATENT

WO-2014110000-A1

Example 2

Synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000023_0002

Stir a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid

(20.22 g; 70.90 mmol), N-(2,3-dihydro- lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (27.99 g; 81.54 mmol) and triethylamine (98.83 mL; 709.03 mmol) in dimethylformamide (404.40 mL) at 0°C. Add a solution of 1-propanephosphonic acid cyclic anhydride (50% solution in DMF; 51.89 mL; 81.54 mmol) over 30 minutes, and stir the mixture at room temperature for 18 hours.

Concentrate the reaction mixture under reduced pressure to give a residue. Add water (200 mL) and extract the mixture with ethyl acetate (4 x 250 mL) and

dichloromethane (4 x 250 mL). Wash the combined organic layers with saturated aqueous sodium bicarbonate (2 x 100 mL) and brine (100 mL), then dry over anhydrous sodium sulfate. Filter the mixture and concentrate the solution under reduced pressure to give a red solid (25.70 g) that is slurried in ethyl acetate/methanol (9: 1 mixture; 200 mL) for 2 hours at room temperature. Filter the resulting solid and wash with cold ethyl acetate (50 mL) to give a solid (ca.18.2 g) that is re-slurried in ethyl acetate (200 mL) at reflux for 1 hour. On cooling to room temperature, stir the mixture for 1 hour and filter the resulting light pink solid.

Slurry the light pink solid in water/methanol (1 : 1 mixture; 200 mL) and heat the mixture at 50°C for 30 minutes. Add ammonium hydroxide solution (32% ; 50 mL) and continue to heat the mixture at 50°C for 30 minutes. Upon cooling to room temperature, add additional ammonium hydroxide solution (32% ; 50 mL) and continue stirring for 1 hour at room temperature. Filter the resulting light gray solid, dry and slurry again in ethyl acetate (200 mL) for 1 hour to afford a light gray solid that is filtered, washed with ethyl acetate (25 mL), and dried to give the title compound (12.42 g; 43%) as a gray solid. MS (m/z): 406 (M+l).

PATENT

US-20140200231-A1

https://www.google.com/patents/US20140200231

Scheme E

Figure imgf000014_0001

Preparation 7

Synthesis of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid.

Figure imgf000018_0001

Pressurize 1 atmosphere of hydrogen (g) to a flask containing [2-(l-benzyl-lH- l,2,3-triazol-5-yl)ethoxy]acetic acid (10.1 g; 1.00 equiv; 38.66 mmoles) and palladium (II) chloride (3 g; 16.92 mmoles; 3.00 g) in isopropyl alcohol (300 mL) and water (60 mL). Maintain the flask under a hydrogen atmosphere for 3 h, then filter through Celite™ and concentrate. Add toluene (2×50 mL) and concentrate to afford the title compound (7.96 g, 100%). ]H NMR (d6-DMSO): 2.86 (t, / = 7 Hz, 2 H), 3.65 (t, / = 7 Hz, 2 H), 3.98 (s, 2 H), 7,77 (s, 1 H), 13.4 – 13.6 (br s, 2 H).

Example 1

Synthesis of l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin- 6(5H)-yl]-2-[2-(lH-l,2,3-triazol-4- l)ethoxy]ethanone.

Figure imgf000018_0002

Add N-indan-2-yl-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin-2-amine (4.2 g, 15.8 mmol) to a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid (2.7 g, 15.8 mmol), 1-hydroxybenzotriazole (3.20 g, 23.7 mmol), and dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (5.44 g, 28.4 mmol) in dichloromethane (40 mL) at 25 °C. Add triethylamine (4.40 mL, 31.6 mmol) to the reaction mixture and stir for 16 h. Wash with water (2 x 50 mL) and concentrate the organic layer. Purify by silica gel column chromatography, eluting with ethyl acetate/methanol, to give the title compound (4.0 g, 60%) as a solid. MS (m/z): 420 (M + Η). Preparation 8

Synthesis of 2-chloro-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8-dihydropyrido[4,3- d]pyrimidin-6(5H)-yl]ethanone.

Figure imgf000019_0001

To N-indan-2-yl-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin-2-amine (11.0 g, 41.3 mmol) and triethylamine (7.48 mL, 53.7 mmol) in dichloromethane (200 mL), add 2- chloroacetyl chloride (3.61 mL, 5.13 g, 45.4 mmol) dropwise over five minutes at 23 °C. Stir for 30 minutes and pour the reaction mixture into 1 : 1 50% saturated aqueous sodium bicarbonate: dichloromethane (75 mL). Separate the organic layer from the aqueous layer and further extract the aqueous layer with dichloromethane (2 x 25 mL). Combine the organic extracts and dry over anhydrous sodium sulfate, filter, and concentrate. Dissolve the residue in chloroform (10 mL) and purify via silica gel column chromatography (gradient elution: 25% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (9.75 g, 69%). ]H NMR (CDC13, * = minor amide rotamer) δ 2.77* (t, 2H), 2.84 (dd, 2H), 2.87 (t, 2H), 3.35 (dd, 2H), 3.76 (t, 2H), 3.85* (t, 2H), 4.12 (s, 2H), 4.52* (s, 2H), 4.57 (s, 2H), 4.72-4.82 (m, IH), 5.48-5.64 (m, IH), 7.12-7.21 (m, 4H), 8.03-8.10 (m, IH).

Preparation 9

Synthesis of 2-(but-3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8- dihydropyrido[4,3-d]p rimidin-6(5H)-yl]ethanone.

Figure imgf000019_0002

To sodium hydride (60 wt% in mineral oil, 1.58 g, 39.6 mmol) in tetrahydrofuran (50 mL) at 23 °C, add 3-butyn-l-ol (7.93 g, 8.59 mL, 113.2 mmol) dropwise, then stir at 23 °C for 20 minutes. Add this solution to 2-chloro-l-[2-(2,3-dihydro-lH-inden-2- ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin-6(5H)-yl]ethanone (9.70 g, 28.3 mmol) in tetrahydrofuran (150 mL) at 23 °C and stir for one hour. Pour the reaction mixture into 50% saturated aqueous sodium bicarbonate solution. Separate the organic layer and further extract the aqueous layer with ethyl ether (x 2) and ethyl acetate (x 2). Combine the organic extracts and wash with brine, then dry over anhydrous sodium sulfate, filter, and concentrate. Purify the resulting crude product by silica gel column chromatography (gradient elution: 20% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (8.16 g, 77%). MS (m/z): 377 (M + 1).

Example la

Alternative synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-7,8-dihydropyrido[4,3- d]pyrimidin-6(5H)-yl]-2-[2-(lH- l,2,3-triazol-4- l)ethoxy]ethanone.

Figure imgf000020_0001

Sparge a solution of 2-(but-3-yn- l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2- ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin-6(5H)-yl]ethanone (8.15 g, 21.7 mmol) and L-ascorbic acid sodium salt (8.58 g, 43.3 mmol) in dimethylformamide (60 mL) and water (60 mL) with nitrogen for ten minutes, then evacuate and backfill with nitrogen three times. Add copper (II) sulfate pentahydrate (1.08 g, 4.33 mmol) and heat to 90 °C, then add azidotrimethylsilane (23.1 mL, 20.0 g, 173 mmol) dropwise and stir for one hour. Cool reaction mixture to 23 °C and pour into water (50 mL). Extract this mixture with ethyl acetate (4 x 50 mL). Combine the organic extracts and wash with saturated aqueous sodium chloride, dry over anhydrous sodium sulfate, filter, and concentrate.

Purify the resulting crude product by silica gel column chromatography (gradient elution: 0 to 10% methanol in ethyl acetate) to give the title compound (3.60 g, 40%). MS (m/z): 420 (M + 1). Preparation 10

Synthesis of tert-butyl-2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidine-6-carboxylate.

Figure imgf000021_0001

Charge 450 rriL (2.58 mol) of N-ethyl-N-isopropylpropan-2-amine into a 15 °C solution of tert-butyl 2-chloro-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidine-6-carboxylate (220 g, 860.37 mmol) and 2,3-dihydro-lH-inden-2-amine (137.7 g, 1.03 mol) in 1- methylpyrrolidin-2-one (3.6 L). Heat the resulting mixture to 80 °C for 16 h, then cool to 30 °C and transfer the resulting mixture into 5 L of water at 25 °C. Filter the resulting solid and rinse the filter cake with water (2 x 300 rriL). Reslurry the solid in ethyl acetate (350 iriL) for 45 min at 15 °C. Filter the slurry, rinsing with 15 °C ethyl acetate ( 2 x 250 rriL), and dry to give the title compound (226 g, 75%) as an off-white solid. ‘H NMR (d6-DMSO) 1.45 (s, 9 H), 2.87 (dd, /= 7.2, 15.8 Hz, 2 H), 3.24 (dd, /= 7.2, 15.8 Hz, 2 H), 4.36 (d, 10.4 Hz, 2 H), 4.44 (d, /= 12.8 Hz, 2 H), 4.60 (m, 1 H), 7.14 (m, 2 H), 7.20 (m, 2 H), 7.55 (d, /= 6.8 Hz, 1 H), 8.27 (d, /= 7.2 Hz, 1 H).

Preparation 11

Synthesis of N-(2,3-dihydro-lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2- amine dihydrochloride hydrate.

Figure imgf000021_0002

Charge 670 rriL of 5 M hydrochloric acid (3.35 mol) to a solution of tert-butyl 2-

(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H pyrrolo[3,4-d]pyrimidine-6- carboxylate (226 g, 641.25 mmol) in tetrahydrofuran (2.0 L) at 17 °C, maintaining the internal temperature below 26 °C during the addition. Heat the resulting solution to 50 °C for 16 h, cool to 25 °C and dilute with 500 rriL of water and 500 mL of tert- butylmethylether. Separate the resulting layers and extract with tert-butylmethylether (3 x 1 L). Concentrate the water phase down to a reaction volume of ca. 200 mL, and filter the resulting slurry. Rinse the cake with tert-butylmethylether (2 x 200 mL) and dry to give the title product (177 g, 80%) as a light brown solid. MS (m/z): 253.2 (M-2HC1- H20+1).

Preparation 12

Syntheis of tert-butyl 2-but-3-ynox acetate.

Figure imgf000022_0001

Stir a mixture of but-3-yn-l-ol (6.00 g; 85.60 mmol), tetrabutylammonium sulfate (2.07 g; 8.54 mmol) and sodium hydroxide (40% wt/wt; 150 mL) in dichloromethane (150 mL) at 0°C. Add tert-butyl bromoacetate (19.34 mL; 128.40 mmol) dropwise and stir the mixture for 2.5 hours at room temperature. Dilute the reaction mixture with dichloromethane (200 mL) and water (100 mL), separate the layers, and further extract the aqueous layer with dichloromethane (2 x 100 mL). Wash the combined organic layers with brine (100 mL), dry over anhydrous sodium sulfate, and concentrate to afford the crude title compound as a brown oil (11.93 g). Purify the oil by silica gel column chromatography, eluting with hexane: ethyl acetate (0% to 10% mixtures) to give the title compound (11.35 g; 72%) as a colorless oil. ]H NMR (CDCI3) δ 1.48 (s, 9H), 2.00 (m, 1H), 2.52 (m, 2H), 3.67 (m, 2H), 4.01 (bs, 2H).

Preparation 13

Synthesis of tert-butyl 2-[2-(lH-triazol-5- l)ethoxy]acetate.

Figure imgf000022_0002

Stir tert-Butyl 2-but-3-ynoxyacetate (11.34 g; 61.55 mmol) and copper(I)iodide (584 mg; 3.07 mmol) in a mixture of dimethylformamide (56.70 mL) and methanol (11.34 mL) at 0°C. Add azido(trimethyl)silane (12.33 mL; 86.47 mmol) dropwise and heat the mixture at 90°C for 18 hours.

In a second batch, stir tert-butyl 2-but-3-ynoxyacetate (4.38 g; 23.77 mmol) and copper(I)iodide (226 mg; 1.19 mmol) in a mixture of dimethylformamide (22 mL) and methanol (6 mL) at 0°C. Add azido(trimethyl)silane (4.8 mL; 33.66 mmol) dropwise and the mixture heated at 90°C for 18 hours.

Upon cooling to room temperature, combine the crude products from both batches and concentrate the mixture to afford a greenish residue. Purify the crude product by filtration through a plug of silica eluting with dichloromethane: ethyl acetate (75% to 100% mixtures) to afford the title compound (14.15 g, 73%) as a colorless oil. MS (m/z): 228.15 (M+l).

Preparation 14

Synthesis of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid.

Figure imgf000023_0001

Stir a mixture of ieri-butyl 2-[2-(lH-triazol-5-yl)ethoxy]acetate (14.15 g; 62.26 mmol) and trifluoroacetic acid (70.75 mL, 935.69 mmol) in dichloromethane (70.75 mL) for 2 hours at room temperature. Concentrate the reaction mixture under reduced pressure to provide the title compound containing additional trifluoroacetic acid (20.22 g, >100%) as a brown solid. MS (m/z): 172.05 (M+l).

Example 2

Synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000023_0002

Stir a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid

(20.22 g; 70.90 mmol), N-(2,3-dihydro- lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (27.99 g; 81.54 mmol) and triethylamine (98.83 mL; 709.03 mmol) in dimethylformamide (404.40 mL) at 0°C. Add a solution of 1-propanephosphonic acid cyclic anhydride (50% solution in DMF; 51.89 mL; 81.54 mmol) over 30 minutes, and stir the mixture at room temperature for 18 hours.

Concentrate the reaction mixture under reduced pressure to give a residue. Add water (200 mL) and extract the mixture with ethyl acetate (4 x 250 mL) and

dichloromethane (4 x 250 mL). Wash the combined organic layers with saturated aqueous sodium bicarbonate (2 x 100 mL) and brine (100 mL), then dry over anhydrous sodium sulfate. Filter the mixture and concentrate the solution under reduced pressure to give a red solid (25.70 g) that is slurried in ethyl acetate/methanol (9: 1 mixture; 200 mL) for 2 hours at room temperature. Filter the resulting solid and wash with cold ethyl acetate (50 mL) to give a solid (ca.18.2 g) that is re-slurried in ethyl acetate (200 mL) at reflux for 1 hour. On cooling to room temperature, stir the mixture for 1 hour and filter the resulting light pink solid.

Slurry the light pink solid in water/methanol (1 : 1 mixture; 200 mL) and heat the mixture at 50°C for 30 minutes. Add ammonium hydroxide solution (32% ; 50 mL) and continue to heat the mixture at 50°C for 30 minutes. Upon cooling to room temperature, add additional ammonium hydroxide solution (32% ; 50 mL) and continue stirring for 1 hour at room temperature. Filter the resulting light gray solid, dry and slurry again in ethyl acetate (200 mL) for 1 hour to afford a light gray solid that is filtered, washed with ethyl acetate (25 mL), and dried to give the title compound (12.42 g; 43%) as a gray solid. MS (m/z): 406 (M+l).

Preparation 15

Synthesis of 2-chloro- l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl]ethanone.

Figure imgf000024_0001

Stir a suspension of N-(2,3-dihydro-lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (14.4 g, 41.9 mmol) and triethylamine (14.3 g, 19.7 mL, 141.4 mmol) in dichloromethane (200 mL) at 23 °C for 10 minutes, then cool to -30 °C. Add 2-chloroacetyl chloride (5.49 g, 3.86 mL, 48.6 mmol) over two minutes and warm to 23 °C over 10 minutes. Add methanol (5 mL) and remove the solvent in vacuo. Slurry the crude reaction mixture in methanol (30 mL), add 50 g silica gel and remove solvent in vacuo. Load the resulting residue onto a loading column and purify via silica gel column chromatography (gradient elution: 50% ethyl acetate in hexanes to ethyl acetate to 10% methanol in ethyl acetate) to give the title compound (11.5 g, 84%). MS (m/z): 329(M+1).

Preparation 16

Synthesis of 2-(but-3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro- 6H-pyrrolo[3,4-d]pyrimidin-6-yl]ethanone.

Figure imgf000025_0001

To sodium hydride (60 wt% in mineral oil, 2.06 g, 51.4 mmol) in tetrahydrofuran (86 mL) at 0 °C, add 3-butyn-l-ol (4.64 g, 5.03 mL, 64.3 mmol), then stir at 23 °C for 15 minutes. Add this solution to 2-chloro-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7- dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]ethanone (8.45 g, 25.7 mmol) in

tetrahydrofuran (86 mL) at 0 °C and stir for five minutes. Pour reaction mixture into 50% saturated aqueous sodium bicarbonate solution. Separate the organic layer and further extract the aqueous layer with ethyl ether and ethyl acetate (2 x 50 mL each). Combine the organic extracts and wash with brine, then dry over anhydrous sodium sulfate, filter, and concentrate. Combine the crude product with the crude product from a second reaction (run reaction under identical conditions and stoichiometry employing 2-chloro- 1- [2-(indan-2-ylamino)-5,7-dihydropyrrolo[3,4-d]pyrimidin-6-yl]ethanone (3.0 g, 9.1 mmol)) and purify by silica gel column chromatography (gradient elution: 25% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (2.90 g, 23%). MS

(m/z): 363(M+1). Example 2a

Alternative synthesis of l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro- pyrrolo[3,4-d]pyrimidin-6-yl]-2-[2-(lH-l,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000026_0001

Add dimethylformamide (27 mL) and water (27 mL) to a flask containing 2-(but- 3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]ethanone (2.90 g, 8.00 mmol). Add copper (II) sulfate pentahydrate (400 mg, 1.60 mmol) and L-ascorbic acid sodium salt (3.17 g, 16.0 mmol). Evacuate flask and backfill with nitrogen (x 2), then add azidotrimethylsilane (7.37 g, 8.53 mL, 64.0 mmol) and heat the reaction to 90 °C for 70 minutes. Cool the reaction mixture to 23 °C and remove all solvent in vacuo. Suspend the residue in methanol/dichloromethane and then add silica gel and remove solvent in vacuo. Load this material onto a loading column and purify via silica gel column chromatography (gradient elution: 0-9% methanol in ethyl acetate) to give the title compound (980 mg, 30%). MS (m/z):

406(M+1).

/////////Autotaxin LPA osteoarthritis tool molecule, lily, Spencer Jones, PRECLINICAL

N1(Cc2cnc(nc2C1)NC3Cc4ccccc4C3)C(=O)COCCc5cnnn5

Identification of an Orally Efficacious GPR40/FFAR1 Receptor Agonist from Zydus Cadila


Indian flag
str1
(S)-3-(4-((3-((isopropyl(thiophen-3- ylmethyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoic acid
str1
Calcium (S)-3-(4-((3-((isopropyl(thiophen-3-yl methyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoate
Calcium (S)-3-(4-((3-((isopropyl(thiophen-3-yl methyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoate
 

The compounds of theese type lower blood glucose, regulate peripheral satiety, lower or modulate triglyceride levels and/or cholesterol levels and/or low-density lipoproteins (LDL) and raises the high-density l ipoproteins (HDL) plasma levels and hence are useful in combating different medical conditions, where such lowering (and raising) is beneficial. Thus, it could be used in the treatment and/or prophylaxis of obesity, hyperlipidemia, hypercholesteremia, hypertension, atherosclerotic disease events, vascular restenosis, diabetes and many other related conditions.

The compounds of are useful to prevent or reduce the risk of developing atherosclerosis, which leads to diseases and conditions such as arteriosclerotic cardiovascular diseases, stroke, coronary heart diseases, cerebrovascular diseases, peripheral vessel diseases and related disorders. -These compounds  are useful for the treatment and/or prophylaxis of metabolic disorders loosely defined as Syndrome X. The characteristic features of Syndrome X include initial insulin resistance followed by hyperinsulinemia, dyslipidemia and impaired glucose tolerance. The glucose intolerance can lead to non-insulin dependent diabetes mel litus (N I DDM, Type 2 diabetes), which is characterized by hyperglycemia, which if not controlled may lead to diabetic complications or metabolic disorders caused by insulin resistance. Diabetes is no longer considered to be associated only with glucose metabol ism, but it affects anatomical and physiological parameters, the intensity of which vary depending upon stages/duration and severity of the diabetic state. The compounds of this invention are also useful in prevention, halting or slowing progression or reducing the risk of the above mentioned disorders along with the resulting secondary diseases such as cardiovascular diseases, l ike arteriosclerosis, atherosclerosis; diabetic retinopathy, diabetic neuropathy and renal disease including diabetic nephropathy, glomerulonephritis, glomerular sclerosis, nephrotic syndrome, hypertensive nephrosclerosis and end stage renal diseases, like microalbuminuria and albuminuria, which may be result of hyperglycemia or hyperinsulinemia.

Diabetes mellitus is a serious disease affl icting over 1 00 mi l lion people worldwide. In the United States, there are more than 12 mill ion diabetics, with 600,000 new cases diagnosed each year.

Diabetes mellitus is a diagnostic term for a group of disorders characterized by abnormal glucose homeostasis resulting in elevated blood sugar. There are many- types of diabetes, but the two most common are Type 1 (also referred to as insulin- dependent diabetes mellitus or IDDM) and Type II (also referred to as non- insulin-dependent diabetes mellitus or NIDDM).

The etiology of the different types of diabetes is not the same; however, everyone with diabetes has two things in common: overproduction of glucose by the liver and little or no ability to move glucose out of the blood, into the cells where it becomes the body’s primary fuel.

People who do not have diabetes rely on insulin, a hormone made in the pancreas, to move glucose from the blood into the cells of the body. However, people who have diabetes either don’t produce insulin or can’t efficiently use the insulin they produce; therefore, they can’t move glucose into their cells. Glucose accumulates in the blood creating a condition called hyperglycemia, and over time, can cause serious health problems.

Diabetes is a syndrome with interrelated metabolic, vascular, and neuropathic components. The metabolic syndrome, generally characterized by hyperglycemia, comprises alterations in carbohydrate, fat and protein metabolism caused by absent or markedly reduced insulin secretion and/or ineffective insulin action. The vascular syndrome consists of abnormalities in the blood vessels leading to cardiovascular, retinal and renal complications. Abnormal ities in the peripheral and autonomic nervous systems are also part of the diabetic syndrome.

About 5% to 10% of the people who have diabetes have IDDM. These individuals don’t produce insulin and therefore must inject insulin to keep their blood glucose levels normal . IDDM is characterized by low or undetectable levels of endogenous insulin production caused by destruction of the insulin-producing β cells of the pancreas, the characteristic that most readily distinguishes IDDM from NIDDM. IDDM, once termed juvenile-onset diabetes, strikes young and older adults alike.

Approximately 90 to 95% of people with diabetes have Type II (or NIDDM). NIDDM subjects produce insulin, but the cells in their bodies are insulin resistant: the cells don’t respond properly to the hormone, so glucose accumulates i n their blood. NIDDM is characterized by a relative disparity between endogenous insulin production and insulin requirements, leading to elevated blood glucose levels. In contrast to IDDM, there is always some endogenous insulin production in NIDDM; many NIDDM patients have normal or even elevated blood insul in levels, whi le other NIDDM patients have inadequate insul in production ( otwein, R. et al. N. Engl. J. Med. 308, 65-71 ( 1983)). Most people diagnosed with NIDDM are age 30 or older, and half of all new cases are age 55 and older. Compared with whites and Asians, NIDDM is more common among Native Americans, African-Americans, Latinos, and Hispanics. In addition, the onset can be insidious or even clinically non-apparent, making diagnosis difficult.

The primary pathogenic lesion on NIDDM has remained elusive. Many have suggested that primary insulin resistance of the peripheral tissues is the initial event. Genetic epidemiological studies have supported this view. Similarly, insulin secretion abnormalities have been argued as the primary defect in NIDDM. It is l ikely that both phenomena are important contributors to the disease process (Rimoin, D. L., et. al. Emery and Rimoin’s Principles and Practice of Medical Genetics 3rd Ed. 1 : 1401 – 1402 ( 1996)).

Many people with NIDDM have sedentary lifestyles and are obese; they weigh approximately 20% more than the recommended weight for their height and build. Furthermore, obesity is characterized by hyperinsul inemia and insul in resistance, a feature shared with NIDDM, hypertension and atherosclerosis.

The G-protein -coupled receptor GPR 40 functions as a receptor for long-chain free fatty acids (FFAs) in the body and as such is impl icated in a large number of metabolic conditions in the body. For example it has been alleged that a GPR 40 agonist promotes insulin secretion whilst a GPR 40 antagonist inhibits insulin secretion and so depending upon the circumstances the agonist and antagonist may be useful as therapeutic agents for the number of insul in related conditions such as type 2 diabetes, obesity, impaired glucose tolerance, insul in resistance, neurodegenerative diseases and the like.

There is increasing evidences that lipids can also serve as extracel lular l igands for a specific class of receptors and thus act as “nutritional sensors” (Nolan CJ et al. J. Clinic. Invest., 2006, 1 1 6, 1 802- 1 812The free fatty acids can regulate cell function. Free fatty acids have demonstrated as ligands for orphan G protein-coupled receptors (GPCRs) and have been proposed to play a critical role in physiological glucose homeostasis.

GPR40, GPR 120, GPR41 and GPR43 exemplify a growing number of GPCRs that have been shown to be activated by free fatty acids. GPR40 and GPR 120 are activated by medium to long-chain free fatty acids whereas GPR 41 and GPR 43 are activated by short-chain fatty acid (Brown AJ et al, 2003).

GPR 40 is highly expressed on pancreatic β-cells, and enhances glucose- stimulated insulin secretion {Nature, 2003, 422, 1 73- 1 76, J. Bio. Chem. 2003, 278, 1 1303- 1 13 1 1 , Biochem. Biophys. Res. Commun. 2003, 301, 406-4 10).

Free fatty acids regulate insulin secretion from pancreatic β cells through GPR40 is reported {Lett, to Nature 2003, 422, 1 73- 1 76).

GlaxoSmith line Research and Development, US published an article in Bioorg. Med. Chem. Lett. 2006, 16, 1840- 1 845 titled Synthesis and activity of small molecule GPR40 agonists. (Does this describe GW9508?)Another article titled Pharmacological regulation of insul in secretion in ΜΓΝ6 cells through the fatty – acid receptor GPR40: Identification of agonist and antagonist small molecules is reported in

Br. J. Pharmacol. 2006, 148, 619-928 from GlaxoSmithKl i ne. USA (Does this describe GW9508?) ‘

GW 9508.

Solid phase synthesis and SAR of small molecule agonists for the. GPR 40 receptor is published in Bioorg. Med. Chem. Lett. 2007, 16, 1 840- 1 845 by Glaxo Smith line Res. 8c Dev. USA, including those with the following structures.

Johnson & Johnson Pharmaceutical Research and development , USA published

Synthesis and Biological Evaluation of 3-Aryl-3-(4-phenoxy)-propanoic acid as a Novel Series of G-protein -coupled receptor 40 agonists J. Med. Chem. 2007,

76, 2807-2817)

National Institutes of Health, Bethesda, Maryland publ ished “Bidirectional Iterative Approach to the Structural Delineation of the Functional Chemo print in GPR 40 for agonist Recognition (J. Med. Chem. 2007. 50, 298 1 -2990).

Discov roglucinols of the following formula

as a new class of GPR40 (FFAR 1 ) agonists has been publ ished by Piramal Li fe Sciences, Ltd. in Bioorg. Med. Chem. Lett. 2008, 1 8, 6357-6361

Synthesis and SAR of 1 ,2,3,4-tctrahydroisoquinoline- l -ones as novel G-protein coupled receptor40(GPR40) antagonists of the following formula has been published in Bioorg. Med. Chem. Lett. 2009, 79, 2400-2403 by Pfizer

Piramal Life Sciences Ltd. published “Progress in the discovery and development of small molecule modulators of G-protei n coupled receptor 40(GPR40/FFA 1 /FFAR1 ), an emerging target for type 2 diabetes” in Exp. Opin. Therapeutic Patents 2009, 19(2), 237 -264.

There was a report published in Zhonggno Bingli Shengli ^Zazhi 2009, 25(7), 1376- 1380 from Sun Yat. Sen University, Guangzhou, which mentions the role GPR 40 on lipoapoptosis.

A novel class of antagonists for the FFA’s receptor GPR 40 was published in Biochem. Biophy. Res. Commun. 2009 390, 557-563.

N41 (DC260126)

Merck Res. Laboratories published “Discovery of 5-aryloxy-2,4-thiazolidinediones as potent GPR40 agonists” having the following formula in Bioorg. Med. Chem. Lett. 2010 20, 1298- 1 301

Discovery of TA -875, a potent, selective, and oral ly bioavai lable G PR 40 agonist is reported by Takeda Pharmaceutical Ltd. ACS Med. Chem. Lett. 2010,

7(6), 290-294

In another report from University of Southern Denmark” Structure -Activity of Dihydrocinnamic acids and discovery of potent FFA l (GPR40) agonist TUG-469″ is reported in ACS Me -349.

The free fatty acid 1 receptor (FFAR 1 or GPR40), which is highly expressed on pancreatic β-cells and amplifies glucose-stimulated insul in secretion, has emerged as an attractive target for the treatment of type 2 diabetes (ACS Med. Chem. Lett. 2010, 1 (6), 290-294).

G-protein coupled receptor (GPR40) expression and its regulation in human pancreatic islets: The role of type 2 diabetes and fatty acids is reported in Nutrition Metabolism & Cardiovascular diseases 2010, 2(9( 1 ), 22-25

Ranbaxy reported “Identification of Berberine as a novel agonist of fatty acid receptor GPR40” in Phytother Res. 2010, 24, 1260-63.

The following substituted 3-(4-aryloxyaryI)-propanoic acids as GPR40 agonists are reported by Merck Res. Lab. in Bioorg. ed. Chem. Lett. 201 1 , 21, 3390-3394

4 EC50=0.970 μΜ 5. EC50=2.484 μΜ

CoMSIA study on substituted aryl alkanoic acid analogs as GPR 40 agonists is reported Chem. Bio. Drug. Des. 201 1 , 77, 361 -372

Takeda further published “Design, Synthesis and biological activity of potential and orally available G-protein coupled receptor 40 agonists” in J. Med. Chem. 201 1 , 54(5), 1365- 1 378.

Amgen disclosed a potent oral ly bioavai lable GPR 40 agonist AMG-837 in Bioorg. Med. Chem. Lett.

Discovery of phenylpropanoic acid derivatives containing polar functional ities as Potent and orally bioavailable G protein-coupled receptor 40 Agonist for the treatment of type 2 Diabetes is reported in J. Med. Chem. 2012, 55, 3756-3776 by Takeda.

Discovery of AM- 1638: A potent and orally bioavailable GPR40/FFA 1 full agonist is reported in ACS Med. Chem. Lett. 2012, 3(9), 726-730.

 

Ranjit Desai

Ranjit Desai

Sr Vice President. Head-Chemistry
Zydus Research Centre, Ahmedabad · Chemistry

Sameer Agarwal

Sameer Agarwal

Cadila Healthcare Ltd., India

Sameer Agarwal has obtained Master’s in Chemistry from IIT, Delhi and was awarded DAAD (German Govt. Scholarship) fellowship to purse research project at Karlsruhe University, Germany. He has received PhD degree from Technical University, Dresden, Germany in the field of Synthetic and bio-organic chemistry under direction of Prof. Dr. Hans-Joachim Knölker, FRSC, a well-known scientist of present times for his contribution towards Alkaloid Chemistry. He worked as Research Scientist (Post-Doc), JADO Technologies, (collaboration with Max Planck Institute (MPI) of Molecular Cell Biology and Genetics and Chemsitry Department, Technical University), Germany. He then decided to return to his home country and working with Zydus Research Centre, Cadila Healthcare Ltd., Ahmedabad as Principal Scientist / Group Leader in the area of basic drug discovery and his research interest includes discovery of cardio metabolic, anti-inflammatory and oncology drugs. He has large number of publications in international journals and patents and is a reviewer of many prestigious journals including American Chemical Society.

Paper

Identification of an Orally Efficacious GPR40/ FFAR1 Receptor Agonist

ArticleinACS Medicinal Chemistry Letters · September 2016
DOI: 10.1021/acsmedchemlett.6b00331
Abstract Image

GPR40/FFAR1 is a G protein-coupled receptor predominantly expressed in pancreatic β-cells and activated by long-chain free fatty acids, mediating enhancement of glucose-stimulated insulin secretion. A novel series of substituted 3-(4-aryloxyaryl)propanoic acid derivatives were prepared and evaluated for their activities as GPR40 agonists, leading to the identification of compound 5, which is highly potent in in vitro assays and exhibits robust glucose lowering effects during an oral glucose tolerance test in nSTZ Wistar rat model of diabetes (ED50 = 0.8 mg/kg; ED90 = 3.1 mg/kg) with excellent pharmacokinetic profile, and devoid of cytochromes P450 isoform inhibitory activity

Synthesis of compound 5 is depicted in Scheme 1a.

The reductive amination1 of commercially available 3-thiophene-aldehyde (3) and isopropyl amine using sodium triacetoxyborohydride resulted in secondary amine intermediate 4. Compound 4 on further reductive amination under similar conditions with aldehyde intermediate, (S)-3-(4-((3-formylbenzyl)oxy)phenyl)hex-4-ynoic acid (8), afforded 2d in high yields. The aldehyde intermediate, 8 was obtained from (S)-3-(4-hydroxyphenyl)hex-4-ynoic acid (6) as shown in Scheme 1b. Acid 6 was synthesized via 5-step reported procedure using commercially available 4-hydroxybenzaldehyde and Meldrum’s acid.2 Resolution of racemic acid 6 was accomplished via diastereomeric salt formation with (1S,2R)-1-amino-2-indanol followed by salt break with aqueous acid to furnish compound 6. Treatment of 6 with of 40% aqueous tetrabutylphosphonium hydroxide (nBu4POH) in THF, followed by addition of 3-formyl benzyl bromide (7), afforded aldehyde intermediate 8. Compound 2d was further converted to its corresponding calcium salt (5) in two-step sequence with excellent chemical purity.

Scheme 1a. Synthesis of Compounds 2d and 5. Reagent and Conditions: (a) CH(CH3)2NH2, NaB(OAc)3H, CH3COOH, dry THF, 0 ᵒC to r.t., 16 h; (b) Comp 8, NaB(OAc)3H, CH3COOH, dry THF, 0 ᵒC to r.t., 16 h; (c) NaOH, MeCN/H2O, r.t., 3 h; (d) CaCl2, MeOH/H2O, r.t., 16 h.

BASE

(S)-3-(4-((3-((isopropyl(thiophen-3- ylmethyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoic acid (1.557 g, 3.34 mmol, 43.0 % yield) as wax solid.

1H NMR (400 MHz, DMSO-d6): δ = 12.35 (br s, 1H), 7.44 (q, J = 3.2 Hz, 2H), 7.32 – 7.24 (m, 6H), 7.04 (d, J = 4.8 Hz, 1H), 6.94 (d, J = 8.4 Hz, 2H), 5.06 (s, 2H), 3.93 (d, J = 2.4 Hz, 1H), 3.51 (d, J = 8.8 Hz, 4H), 2.84 (sept, J = 6.4 Hz, 1H), 2.57 (d, J = 8 Hz, 2H), 1.77 (d, J = 2.4 Hz, 3H), 1.01 (d, J = 6.4 Hz, 6H);

13C NMR and DEPT: DMSO-d6, 100MHz):- δ = 172.35 (C), 157.63 (C), 142.13 (C), 141.44 (C), 137.42 (C), 133.93 (C), 128.73 (CH), 128.64 (CH), 128.43 (CH), 127.99 (CH), 127.73 (CH), 126.28 (CH), 122.21 (CH), 115.10 (CH), 81.16 (C), 78.52 (C), 69.69 (CH2), 52.90 (CH2), 48.64 (CH), 48.49 (CH2), 43.44 (CH2), 33.15 (CH), 17.92 (CH3), 3.66 (CH3);

MS (EI): m/z (%) = 462.35 (100) (M+H) + ;

IR (KBr): ν = 3433, 2960, 2918, 2810, 1712, 1608, 1510, 1383, 1240, 1174, 1109, 1018 cm-1 .

CA SALT

calcium (S)-3-(4-((3-((isopropyl(thiophen-3-yl methyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoate (1.51 g, 1.536 mmol, 46% yield) as white powder. mp: 124.5 o C;

1H NMR (400 MHz, DMSO-d6): δ = 7.43 – 7.42 (m, 2H), 7.28 – 7.24 (m, 6H), 7.04 (d, J = 4.4 Hz, 1H), 6.89 (d, J = 8.4 Hz, 2H), 5.02 (s, 2H), 4.02 (s, 1H), 3.50 (d, J = 7.2 Hz, 4H), 2.84 – 2.77 (sept, J = 6.4 Hz, 1H), 2.43 (dd, J1 = 6.8 Hz, J2 = 7.2 Hz, 1H), 2.28 (dd, J1 = 6.8 Hz, J2 = 7.2 Hz, 1H), 1.73 (s, 3H), 0.99 (d, J = 6.4 Hz, 6H);

13C NMR and DEPT (100 MHz, DMSO-d6): δ = 177.78 (C), 157.23 (C), 142.11 (C), 141.4 (C), 137.46 (C), 135.81 (C), 128.83 (CH), 128.62 (CH), 128.40 (CH), 127.94 (CH), 127.69 (CH), 126.26 (CH), 122.18 (CH), 114.77 (CH), 83.18 (C), 77.32 (C), 69.66 (CH2), 52.89 (CH2), 48.59 (CH), 48.48 (CH2), 46.86 (CH2), 33.52 (CH), 17.88 (CH3), 3.78 (CH3);

MS (EI): m/z (%) = 462.05 (100) (M+H)+ ;

ESI-Q-TOF-MS: m/z [M+H]+ calcd for [C28H31NO3S + H]+ : 462.6280; found: 462.4988;

IR (KBr): ν = 3435, 2960, 2918, 2868, 2818, 1608, 1550, 1508, 1440, 1383, 1359, 1240 cm-1 ;

HPLC (% Purity) = 99.38%; Calcium Content (C56H60CaN2O6S2) Calcd.: 4.17%. Found: 3.99%.

 COMPD Ca salt

Calcium (S)-3-(4-((3-((isopropyl(thiophen-3-yl methyl)amino)methyl)benzyl)oxy)phenyl)hex-4-ynoate

Identification of an Orally Efficacious GPR40/FFAR1 Receptor Agonist

Zydus Research Centre, Cadila Healthcare Ltd., Sarkhej-Bavla N.H. No. 8 A, Moraiya, Ahmedabad-382 210, India
ACS Med. Chem. Lett., Article ASAP
DOI: 10.1021/acsmedchemlett.6b00331
*(S.A.) E-mail: sameeragarwal@zyduscadila.com or sameer_ag@yahoo.com., *(R.C.D.) E-mail: ranjitdesai@zyduscadila.com. Fax:+91-2717-665355. Tel: +91-2717-665555.
Ranjit Desai

Sr Vice President, Head Chemistry

Zydus Cadila

2012 – Present (4 years)Zydus Research Centre, Ahmedabad, India

Pankaj Patel, chairman and MD, Cadila Healthcare Ltd
Dr. Mukul Jain

Senior Vice President at Zydus Research Centre

Prashant Deshmukh

Prashant Deshmukh

Research Officer at Zydus Cadila

Dr. Poonam Giri

Dr. Poonam Giri

Principal Scientist at Zydus Research Centre

Bhadresh Rami

Bhadresh Rami

Debdutta Bandyopadhyay

Debdutta Bandyopadhyay

Senior General manager at Zydus Research Centre

Suresh Giri

Suresh Giri

Research Scientist

 References
1. Abdel-Magid, A. F.; Carson, K. G.; Harris, B. D.; Maryanoff, C. A.; Shah, R. D. Reductive Amination of Aldehydes and Ketones with Sodium Triacetoxyborohydride. Studies on Direct and Indirect Reductive Amination Procedures. J. Org. Chem., 1996, 61 (11), 3849–3862.
2. Walker, S. D.; Borths, C. J.; DiVirgilio, E.; Huang, L.; Liu, P.; Morrison, H.; Sugi, K.; Tanaka, M.; Woo, J. C. S.; Faul, M. M. Development of a Scalable Synthesis of a GPR40 Receptor Agonist. Org. Process Res. Dev. 2011, 15, 570–580.
3. Desai, R. C., Agarwal, S. Novel Heterocyclic Compounds, Pharmaceutical Compositions and Uses Thereof. Indian Pat. Appl. 2025/MUM/2015, 25 May 2015.
4. Cheng, Z., Garvin, D., Paguio, A., Stecha, P., Wood, K., & Fan, F. Luciferase Reporter Assay System for Deciphering GPCR Pathways. Current Chemical Genomics, 2010, 4, 84–91. http://doi.org/10.2174/1875397301004010084
5. Arkin, M. R., Connor, P. R., Emkey, R., et al. FLIPR™ Assays for GPCR and Ion Channel Targets. 2012 May 1 [Updated 2012 Oct 1]. In: Sittampalam, G. S., Coussens, N. P., Nelson, H., et al., editors. Assay Guidance Manual [Internet]. Bethesda (MD): Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2004. Available from: http://www.ncbi.nlm.nih.gov/books/NBK92012/
6. Garbison, K. E., Heinz, B. A., Lajiness, M. E. IP-3/IP-1 Assays. 2012 May 1. In: Sittampalam, G. S., Coussens, N. P., Nelson, H., et al., editors. Assay Guidance Manual [Internet]. Bethesda (MD): Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2004. Available from: http://www.ncbi.nlm.nih.gov/books/NBK92004/
7. Milić, A., Mihaljević, V.B., Ralić, J. et al. A comparison of in vitro ADME properties and pharmacokinetics of azithromycin and selected 15-membered ring macrolides in rodents. Eur J Drug Metab Pharmacokinet, 2014, 39, 263. doi:10.1007/s13318-013-0155-8
8. Bell, R. H.; Hye, R. J. Animal models of diabetes mellitus: physiology and pathology. J. Surg. Res. 1983, 35, 433-460.
9. Shafrir, E. Animal models of non insulin dependent diabetes. Diabetes Metab Rev. 1992, 8, 179- 208.

 

Paper
Development of a Scalable Synthesis of a GPR40 Receptor Agonist
Chemical Process Research and Development, Amgen Inc., Thousand Oaks, California 91320, United States
Org. Process Res. Dev., 2011, 15 (3), pp 570–580
*Tel: 805-313-5152. Fax: 805-375-4532. E-mail: walkers@amgen.com.
Abstract Image

Early process development and salt selection for AMG 837, a novel GPR40 receptor agonist, is described. The synthetic route to AMG 837 involved the convergent synthesis and coupling of two key fragments, (S)-3-(4-hydroxyphenyl)hex-4-ynoic acid (1) and 3-(bromomethyl)-4′-(trifluoromethyl)biphenyl (2). The chiral β-alkynyl acid 1 was prepared in 35% overall yield via classical resolution of the corresponding racemic acid (±)-1. An efficient and scalable synthesis of (±)-1 was achieved via a telescoped sequence of reactions including the conjugate alkynylation of an in situ protected Meldrum’s acid derived acceptor prepared from 3. The biaryl bromide 2 was prepared in 86% yield via a 2-step Suzuki−Miyaura coupling−bromination sequence. Chemoselective phenol alkylation mediated by tetrabutylphosphonium hydroxide allowed direct coupling of 1 and 2 to afford AMG 837. Due to the poor physiochemical stability of the free acid form of the drug substance, a sodium salt form was selected for early development, and a more stable, crystalline hemicalcium salt dihydrate form was subsequently developed. Overall, the original 12-step synthesis of AMG 837 was replaced by a robust 9-step route affording the target in 25% yield.

Image result for AMG 837
CAS [1291087-14-3] AMG 837
 Image result for AMG 837
“Enantioselective Synthesis of a GPR40 Agonist AMG 837 via Catalytic Asymmetric Conjugate Addition of Terminal Alkyne to α,β-Unsaturated Thioamide” Yazaki, R.; Kumagai, N.; Shibasaki, M. Org. Lett. 2011, 13, 952.   highlighted by Synfacts 2011, 6, 586.
NMR

/////////fatty acids, FFAR1 GPR40, GPR40 agonist, insulin secretion, type 2 diabetes, GPR40/FFAR1 Receptor Agonist, ZYDUS CADILA
c1(ccc(cc1)OCc2cc(ccc2)CN(Cc3ccsc3)C(C)C)[C@H](CC(=O)O[Ca]OC(C[C@@H](c4ccc(cc4)OCc5cc(ccc5)CN(Cc6ccsc6)C(C)C)C#CC)=O)C#CC
c1(ccc(cc1)OCc2cc(ccc2)CN(Cc3ccsc3)C(C)C)[C@H](CC(=O)O)C#CC

Photoinduced Conversion of Antimelanoma Agent Dabrafenib to a Novel Fluorescent BRAFV600E Inhibitor


Abstract Image

str1

N-(5-amino-2-tert-butyl)-11-fluorbenzol[f]thiazol-[4,5-h]-quinazolin-10-yl)-2,6-difluorbenzolsulfonamide = Dabrafenib_photo (2)

C23H18F3N5O2S2 (Mr = 517.09)

Solution of 5 mg (9.6 μmol) dabrafenib in 2 ml THF was irradiated at 365 nm with 5.4 W for 2 min. This procedure was repeated 18 times at room temperature. The reaction batches were combined. The total initial weight of dabrafenib was 101 mg (190 μmol). The solvent was removed under reduced pressure and the residue was purified by the flash chromatography (SiO2 reversed phase, MeOH/water gradient 50:50 to 100:0) to give compound 2 as a yellowish solid (36.2 mg, 70.0 μmol, yield: 37%).

1H-NMR (DMSO-d6 , 300 MHz): δ = 1.52 (s, 9 H, H-8), 7.28 (m, 2 H, NH2), 7.28 (ddd, 5 J = 0.4 Hz, 4 J = 1.7 Hz, 3 J = 8.5 Hz, 3 J = 8.9 Hz, 2 H, H-18), 7.59 (dd, 3 J = 7.4 Hz, 3 J = 7.8 Hz, 1 H, H-13), 7.71 (tt, 4 J = 6.1 Hz, 3 J = 8.5 Hz, 1 H, H-19), 8.56 (dd, 4 J = 0.9 Hz, 3 J = 9.3 Hz, 1 H, H-14), 9.79 (s, 1 H, H-2), 11.01 (s, 1 H, NH) ppm.

13C-NMR (DMSO-d6 , 300 MHz): δ = 30.4 (s, C-8), 38.3 (s, C-7), 110.9 (d, 4 JCF = 1.6 Hz, C-3), 113.4 (dd, 2 JCF = 22.7 Hz, 2 JCH = 3.5 Hz, C-18), 114.6 (d, 3 JCF = 10.3 Hz, C-9), 117.4 (d, 2 JCF = 16.1 Hz, C-16), 117.6 (dd, 4 JCF = 0.54 Hz, 2 JCH = 4.4 Hz, C-13), 120.8 (d, 2 JCF = 12.3 Hz, C-10), 125.4 (s, C-13), 129.3 (d, 3 JCF = 3.9 Hz, C-15), 130.6 (s, C-5), 135.9 (tt, 3 JCF = 10.9 Hz, 2 JCH = 3.3 Hz, C-19), 148.8 (dd, 2 JCF = 0.54 Hz, 2 JCH = 7.2 Hz, C-12), 149.2 (s, C-4), 150.1 (s, C-11), 157.1 160.5 (dd, 3 JFF = 257.3 Hz, 2 JCF = 3.61 Hz, C-4), 157.9 (s, C-2), 162.1 (s, C-1), 184.0 (s, C-6) ppm.

15N-HMBC (DMSO-d6 , 300 MHz): δ = 9.79/-119.60, 11.01/-268.37 ppm. 19F-NMR (DMSO-d6 , 300 MHz): δ = -121.03 (s, 1 F, F-11), -107.18 (m, 2 F, F-17) ppm.

HRMS (EI, 205 °C, THF): m/z = 517.0849 [M]+ .

LC-MS (ESI, 70 eV, MeOH): tR = 9.3 min; m/z (%) = 518.1 (100) [M+H]+

IR (ATR):  ̃ = 3490 (N-H), 3176 (arom. C-H), 2926 (C-H3), 1696 (N=N), 1613 (N-H), 1587, 1522, 1488, 1469 (arom. C=C), 1342 (sulfonamide), 1277, 1240, 1174 (C-F) cm-1 .

Photoinduced Conversion of Antimelanoma Agent Dabrafenib to a Novel Fluorescent BRAFV600E Inhibitor

Institute of Pharmacy, University of Kiel, Gutenbergstr. 76, D-24118 Kiel, Germany
ACS Med. Chem. Lett., Article ASAP
DOI: 10.1021/acsmedchemlett.6b00340
Publication Date (Web): September 20, 2016
Copyright © 2016 American Chemical Society
*E-mail: cpeifer@pharmazie.uni-kiel.de. Tel: +49-431-880-1137.

ACS Editors’ Choice – This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.

Abstract

Dabrafenib (Tafinlar) was approved in 2013 by the FDA as a selective single agent treatment for patients with BRAFV600E mutation-positive advanced melanoma. One year later, a combination of dabrafenib and trametinib was used for treatment of BRAFV600E/K mutant metastatic melanoma. In the present study, we report on hitherto not described photosensitivity of dabrafenib both in organic and aqueous media. The half-lives for dabrafenib degradation were determined. Moreover, we revealed photoinduced chemical conversion of dabrafenib to its planar fluorescent derivative dabrafenib_photo 2. This novel compound could be isolated and biologically characterized in vitro. Both enzymatic and cellular assays proved that 2 is still a potent BRAFV600E inhibitor. The intracellular formation of 2 from dabrafenib upon ultraviolet irradiation is shown. The herein presented findings should be taken in account when handling dabrafenib both in preclinical research and in clinical applications.

////////Photoinduced Conversion, Antimelanoma Agent,  Dabrafenib, Novel Fluorescent BRAFV600E Inhibitor, BRAFV600E; Dabrafenib, fluorescent probe kinase inhibitor photoinduced conversion

Synthesis of 4-Heteroaryl–Quinazoline Derivatives as Potential Anti-breast Cancer Agents


Image result for Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt

Banner

Figure 1.

Figure 2.

Ethyl 2-[(6,7-dimethoxyquinazolin-4-yl)amino]-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxylate (15b)

Yield: 76%; mp: 254–256°C; IR (cm−1): 3200 (NH), 2974, 2854 (CH-aliphatic), 1656 (C=O); 1H NMR (DMSO-d6) δ ppm 1.03 (t, 3H, CH3CH2, J = 7.2 Hz), 1.21 (t, 2H, CH2, J = 6.9 Hz), 2.88 (t, 2H, CH2, J = 6.9 Hz), 3.40 (q, 2H, CH2, J = 6.9 Hz), 3.88 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.96 (m, 4H, 2 CH2), 4.24 (q, 2H, CH3CH2, J = 7.2 Hz), 7.40 (s, 1H, Ar-H), 7.62 (s, 1H, Ar-H), 8.99 (s, 1H, Ar-H), 12.00 (s, 1H, NH, D2O exchangeable); Anal. Calcd for C22H25N3O4S: C, 61.81; H, 5.89; N, 9.83. Found: C, 61.93; H, 5.96; N, 9.98.

General procedure for the synthesis of compounds 15a,15b

A mixture of 4-chloro-6,7-dimethoxyquinazoline (1) (0.22 g, 1 mmol) and ethyl 2-amino-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxylate (14a) or ethyl 2-amino-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxylate (14b) (1 mmol) in isopropanol (15 mL) was heated under reflux for 10 h. The reaction was cooled, and the solid formed was filtered, dried, and crystallized from isopropanol.

Synthesis of 4-Heteroaryl–Quinazoline Derivatives as Potential Anti-breast Cancer Agents

A. E. Kassab, E. M. Gedawy, H. B. El-Nassan+

+Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt

E-mail: hala_bakr@hotmail.com

str1

Asmaa Elsayed Abd Ellatief Kassab( A. E. Kassab)

4-Heteroaryl or heteroalkyl–quinazoline derivatives were prepared as dual epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitors. The new compounds were tested for their dual enzyme inhibition as well as their cytotoxic activity on MCF7 cell line. The results indicated that almost all the compounds showed moderate dual inhibition of both enzymes. Compound 3 (methyl piperidine-4-carboxylate derivative) showed the highest inhibitory activity against both enzymes with IC50 97.6 and 64.0 µM against EGFR and VEGFR-2 kinases, respectively. Most of the test compounds showed potent to moderate antitumor activity on MCF7 cell line. Five compounds (3, 9c, 11, 13, and 15b) showed potent cytotoxic activity with IC50values between 10 and 17 µM.

Scheme 4.

Scheme 4.

Scheme 3.

Scheme 3.

Scheme 2.

Scheme 2.

Scheme 1.

Scheme 1.

Map of cairo university

Image result for UNIVERSITY OF CAIRO

CAIRO UNIVERSITY

LIBRARY.CAIRO UNIV


DOWNTOWN CAIRO

Image result for Faculty of Pharmacy, Cairo University

Image result for Faculty of Pharmacy, Cairo University


Dean of faculty of pharmacy, Cairo University, Dr. Azza Agha during the opening of the first international day at Faculty of Pharmacy.

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

Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK

Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus

 amcrasto@gmail.com

//////////4-Heteroaryl–Quinazoline Derivatives,  Anti-breast Cancer Agents

VT 1129, QUILSECONAZOLE


str1

VT 1129 BENZENE SULFONATE

CAS 1809323-18-9

Image result for VT1129

str1

VT 1129

QUILSECONAZOLE

1340593-70-5 CAS

MF C22 H14 F7 N5 O2, MW 513.37
2-Pyridineethanol, α-(2,4-difluorophenyl)-β,β-difluoro-α-(1H-tetrazol-1-ylmethyl)-5-[4-(trifluoromethoxy)phenyl]-, (αR)-
R ISOMER
ROTATION +
  • Originator Viamet Pharmaceuticals
  • Class Antifungals; Small molecules
  • Mechanism of Action 14-alpha demethylase inhibitors
  • Orphan Drug Status Yes – Cryptococcosis
  • On Fast track Cryptococcosis
  • Phase I Cryptococcosis
  • Most Recent Events

    • 01 Jun 2016 VT 1129 receives Fast Track designation for Cryptococcosis [PO] (In volunteers) in USA
    • 30 May 2016 Viamet Pharmaceuticals plans a phase II trial for Cryptococcal meningitis in USA (Viamet Pharmaceuticals pipeline; May 2016)
    • 27 May 2016 Phase-I clinical trials in Cryptococcosis (In volunteers) in USA (PO) before May 2016 (Viamet Pharmaceuticals pipeline; May 2016)

Image result for Viamet Pharmaceuticals Holdings LLC

William J. Hoekstra, Stephen William Rafferty,Robert J. Schotzinger
Applicant Viamet Pharmaceuticals, Inc.

Image result for VT1129

Viamet, in collaboration with Therapeutics for Rare and Neglected diseases, is investigating VT-1129, a small-molecule lanosterol demethylase inhibitor, developed using the company’s Metallophile technology, for treating fungal infections, including Cryptococcus neoformans meningitis.

VT-1129 is a novel oral agent that we are developing for the treatment of cryptococcal meningitis, a life-threatening fungal infection of the brain and the spinal cord that occurs most frequently in patients with HIV infection, transplant recipients and oncology patients. Without treatment, the disease is almost always fatal.

VT-1129VT-1129 has shown high potency and selectivity in in vitro studies and is an orally administered inhibitor of fungal CYP51, ametalloenzyme important in fungal cell wall synthesis. In preclinical studies, VT-1129 has demonstrated substantial potency against Cryptococcus species, the fungal pathogens that cause cryptoccocal meningitis, and has also been shown to accumulate to high concentrations within the central nervous system, the primary site of infection.

In in vitro studies, VT-1129 was significantly more potent against Cryptococcus isolates than fluconazole, which is commonly used for maintenance therapy of cryptococcal meningitis in the United States and as a primary therapy in the developing world. Oral VT-1129 has also been studied in a preclinical model of cryptococcal meningitis, where it was compared to fluconazole.  At the conclusion of the study, there was no detectable evidence of Cryptococcus in the brain tissue of the high dose VT-1129 treated groups, in contrast to those groups treated with fluconazole. To our knowledge, this ability to reduce the Cryptococcus pathogen in the central nervous system to undetectable levels in this preclinical model is unique to VT-1129.

Opportunity

An estimated 3,400 hospitalizations related to cryptococcal meningitis occur annually in the United States and the FDA has granted orphan drug designation to VT-1129 for the treatment of this life-threatening disease. In addition, the FDA has granted Qualified Infectious Disease Product designation to VT-1129 for the treatment of Cryptococcus infections, which further underscores the unmet medical need. In developing regions such as Africa, cryptococcal meningitis is a major public health problem, with approximately one million cases and mortality rates estimated to be as high as 55-70%.

Current Status

VT-1129 has received orphan drug and Fast Track designations for the treatment of cryptococcal meningitis and has been designated a Qualified Infectious Disease Product (QIDP) by the U.S. Fod and Drug Administration.  We are currently conducting a Phase 1 single-ascending dose study of VT-1129 in healthy volunteers.

str1 str2

Conclusions

• VT-1129 has robust activity against Cryptococcus isolates with elevated fluconazole MICs and may be a viable option in persons infected with such strains.

• A Phase 1 study of VT-1129 in healthy volunteers is scheduled to begin by the end of 2015. Phase 2 trials in persons with cryptococcal meningitis are targeted to begin by the end of 2016.

Image result for VT 1129

Living organisms have developed tightly regulated processes that specifically import metals, transport them to intracellular storage sites and ultimately transport them to sites of use. One of the most important functions of metals such as zinc and iron in biological systems is to enable the activity of metalloenzymes. Metalloenzymes are enzymes that incorporate metal ions into the enzyme active site and utilize the metal as a part of the catalytic process. More than one-third of all characterized enzymes are metalloenzymes.

The function of metalloenzymes is highly dependent on the presence of the metal ion in the active site of the enzyme. It is well recognized that agents which bind to and inactivate the active site metal ion dramatically decrease the activity of the enzyme. Nature employs this same strategy to decrease the activity of certain metalloenzymes during periods in which the enzymatic activity is undesirable. For example, the protein TIMP (tissue inhibitor of metalloproteases) binds to the zinc ion in the active site of various matrix metalloprotease enzymes and thereby arrests the enzymatic activity. The pharmaceutical industry has used the same strategy in the design of therapeutic agents. For example, the azole antifungal agents fluconazole and voriconazole contain a l-(l,2,4-triazole) group that binds to the heme iron present in the active site of the target enzyme lanosterol demethylase and thereby inactivates the enzyme.

In the design of clinically safe and effective metalloenzyme inhibitors, use of the most appropriate metal-binding group for the particular target and clinical indication is critical. If a weakly binding metal-binding group is utilized, potency may be suboptimal. On the other

hand, if a very tightly binding metal-binding group is utilized, selectivity for the target enzyme versus related metalloenzymes may be suboptimal. The lack of optimal selectivity can be a cause for clinical toxicity due to unintended inhibition of these off-target metalloenzymes. One example of such clinical toxicity is the unintended inhibition of human drug metabolizing enzymes such as CYP2C9, CYP2C19 and CYP3A4 by the currently- available azole antifungal agents such as fluconazole and voriconazole. It is believed that this off-target inhibition is caused primarily by the indiscriminate binding of the currently utilized l-(l,2,4-triazole) to iron in the active site of CYP2C9, CYP2C19 and CYP3A4. Another example of this is the joint pain that has been observed in many clinical trials of matrix metalloproteinase inhibitors. This toxicity is considered to be related to inhibition of off-target metalloenzymes due to indiscriminate binding of the hydroxamic acid group to zinc in the off-target active sites.

Therefore, the search for metal-binding groups that can achieve a better balance of potency and selectivity remains an important goal and would be significant in the realization of therapeutic agents and methods to address currently unmet needs in treating and preventing diseases, disorders and symptoms thereof. Similarly, methods of synthesizing such therapeutic agents on the laboratory and, ultimately, commercial scale is needed. Addition of metal-based nucleophiles (Zn, Zr, Ce, Ti, Mg, Mn, Li) to azole-methyl substituted ketones have been effected in the synthesis of voriconazole (M. Butters, Org. Process Res. Dev.2001, 5, 28-36). The nucleophile in these examples was an ethyl-pyrimidine substrate. Similarly, optically active azole-methyl epoxide has been prepared as precursor electrophile toward the synthesis of ravuconazole (A. Tsuruoka, Chem. Pharm. Bull.1998, 46, 623-630). Despite this, the development of methodology with improved efficiency and selectivity is desirable.

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2011133875

Scheme 1

EXAMPLE 7

2-(2, 4-Difluorophenyl)-l, l-difluoro-3-(lH-tetrazol-l-yl)-l-(5-(4- (trifluoromethoxy) phenyl) pyridin-2-yl) propan-2-ol (7)

To a stirred solution of bromo epoxide C (0.5 g, 1.38 mmol) in THF (30 mL) and water (14 mL) were added 4-(trifluoromethoxy) phenylboronic acid (0.22 g, 1.1 mmol), Na2C03 (0.32 g, 3.1 mmol) and Pd(dppf)2Cl2 (0.28 g, 0.34 mmol) at RT under inert atmosphere. After purged with argon for a period of 30 min, the reaction mixture was heated to 75°C and stirring was continued for 4 h. Progress of the reaction was monitored by TLC. The reaction mixture was cooled to RT and filtered through a pad of celite. The filtrate was concentrated under reduced pressure; obtained residue was dissolved in ethyl acetate (30 mL). The organic layer was washed with water, brine and dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the coupled product (0.45 g, 1.0 mmol, 73%) as solid. 1H NMR (200 MHz, CDC13): δ 8.87 (s, 1 H), 7.90 (dd, / = 8.2, 2.2 Hz, 1 H), 7.66-7.54 (m, 3 H), 7.49-7.34 (m, 3 H), 6.90-6.70 (m, 2 H), 3.49 (d, / = 5.0 Hz, 1 H), 3.02-2.95 (m, 1 H). Mass: m/z 444 [M++l].

To a stirred solution of the coupled product (0.45 g, 1.0 mmol) in DMF (10 mL) was added K2C03 (70 mg, 0.5 mmol) followed by IH-tetrazole (70 mg, 1.0 mmol) at RT under inert atmosphere. The reaction mixture was stirred for 4 h at 80 °C. The volatiles were removed under reduced pressure and obtained residue was dissolved in water (15 mL) and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were washed with water, brine and dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford 7 (0.19 g, 0.37 mmol, 36 %) as white solid. 1H NMR (500 MHz, CDC13): δ 8.76 (s, 1 H), 8.70 (s, 1 H), 7.97 (dd, / = 8.0, 2.0 Hz, 1 H), 7.68 (d, / = 8.5 Hz, 1 H), 7.60-7.56 (m, 3 H), 7.43-7.36 (m, 3 H), 6.80-6.76 (m, 1 H), 6.70-6.67 (m, 1 H), 5.57 (d, / = 14.5 Hz, 1 H), 5.17 (d, / = 14.5 Hz, 1 H). HPLC: 98.3%. Mass: m/z 513.9 [M++l].

Chiral preparative HPLC of enantiomers:

The enantiomers of 7 (17.8 g, 34.6 mmol) were separated by normal-phase preparative high performance liquid chromatography (Chiralpak AD-H, 250 x 21.2 mm, 5μ; using (A) n-hexane – (B) IPA (A:B : 70:30) as a mobile phase; Flow rate: 15 mL/min) to obtain 7(+) (6.0 g) and 7(-) (5.8 g).

Analytical data for 7 (+):

HPLC: 99.8%.

Chiral HPLC: Rt = 9.88 min (Chiralpak AD-H, 250 x 4.6mm, 5μ; mobile phase (A) n-Hexane (B) IPA (7/3): A: B (70:30); flow Rate: 1.00 mL/min)

Optical rotation [a]D25: + 19° (C = 0.1 % in MeOH).

Patent

WO2015143137,

https://patentscope.wipo.int/search/ko/detail.jsf;jsessionid=61AAA66F887FDBB9CFC3F752AFF04016.wapp2nC?docId=WO2015143137&recNum=303&office=&queryString=&prevFilter=%26fq%3DICF_M%3A%22C07D%22&sortOption=%EA%B3%B5%EA%B0%9C%EC%9D%BC(%EB%82%B4%EB%A6%BC%EC%B0%A8%EC%88%9C)&maxRec=58609

Examples

The present invention will now be demonstrated using specific examples that are not to be construed as limiting.

General Experimental Procedures

Definitions of variables in the structures in schemes herein are commensurate with those of corresponding positions in the formulae delineated herein.

Synthesis of 1 or la

A process to prepare enantiopure compound 1 or la is disclosed. Syntheses of 1 or la may be accomplished using the example syntheses that are shown below (Schemes 1-9). The preparation of precursor ketone 8 is performed starting with reaction of dibromo-pyridine 2-Br with ethyl 2-bromo-difluoroacetate to produce ester 3-Br. This ester is reacted with tetrazole reagent 4 via Claisen reaction to furnish 5-Br. Decarboxylation of 5-Br via a two-step process produces compound 6-Br. Suzukin coupling of 6-Br with boronate 7 furnishes 8.

Scheme 1. Synthesis of ketone 8

Ketone 8 may be prepared in an analogous fashion as described in Scheme 1 starting from corresponding substituted 2-bromo-pyridines, which can be prepared using according to synthetic transformations known in the art and contained in the references cited herein (Scheme 2).

Scheme 2. Synthesis of ketone 8

= halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, – 0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, – 0(S02)-aryl, or -0(S02)-substituted aryl.

Compounds 6 or 8 may be reacted with a series of metallated derivatives of 2,4-difluoro-bromobenzene and chiral catalysts/reagents (e.g. BINOL) to effect enantiofacial-selective addition to the carbonyl group of 6 or 8 (Scheme 3). These additions can be performed on 6 or 8 to furnish 9 (or 9a, the enantiomer of 9, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof), respectively.

Scheme 3. Synthesis of 1 or la

R-i = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, -0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, -0(S02)-aryl, or -0(S02)-substituted aryl.

Alternatively, ketone 8 can be synthesized from aldehyde 10 (Scheme 4). Aldehyde 10 is coupled with 7 to produce 11. Compound 11 is then converted to 12 via treatment with diethylaminosulfurtrifluoride (DAST).

Scheme 4. Alternate synthesis of ketone 8

Scheme 5 outlines the synthesis of precursor ketone 15-Br. The ketone is prepared by conversion of 2-Br to 3-Br as described above. Next, ester 3-Br is converted to 15-Br by treatment via lithiation of 2,4-difluoro-bromobenzene.

Scheme 5. Synthesis of ketone 15-Br

Ketone 15 may be prepared in an analogous fashion as described for 15-Br in Scheme 5 starting from corresponding substituted 2-bromo-pyridines, which can be prepared using according to synthetic transformations known in the art and contained in the references cited herein (Scheme 6).

Scheme 6. Synthesis of ketone 15

F = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, – 0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, – 0(S02)-aryl, or -0(S02)-substituted aryl.

Ketone 15 may be used to prepare 9 (or 9a, the enantiomer of 9, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof) by the following three-step process (Scheme 7). In the presence of a chiral catalyst/reagent (e.g. proline derivatives), base-treated nitromethane is added to 15 or 16 to furnish 17 (or 17a, the enantiomer of 17, or mixtures thereof) or 18 (or 18a, the enantiomer of 18, or mixtures thereof), respectively. Reduction of 17 (or 17a, the enantiomer of 17, or mixtures thereof) or 18 (or 18a, the enantiomer of 18, or mixtures thereof) (e.g. lithium aluminum hydride) produces 19 (or 19a, the enantiomer of 19, or mixtures thereof) or 20 (or 20a, the enantiomer of 20, or mixtures thereof). Annulation of 19 (or 19a, the enantiomer of 19, or mixtures thereof) or 20 (or 20a, the enantiomer of 20, or mixtures thereof) by treatment with sodium azide/triethylorthoformate furnishes tetrazoles 9 (or 9a, the enantiomer of 9, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof). Suzuki coupling of 9 (or 9a, the enantiomer of 9, or mixtures thereof) with 4-trifluoromethoxyphenyl-boronic acid produces 1 (or la, the enantiomer of 1, or mixtures thereof).

Scheme 7. Asymmetric Henry reaction

R-ι = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, 0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted a 0(S02)-aryl, or -0(S02)-substituted aryl.

Ketone 21 may be employed to prepare optically-active epoxides via Horner-Emmons reaction of a difluoromethyl substrate to produce 22 or 22a. Ketones related to 21 have been prepared (M. Butters, Org. Process Res. Dev. 2001, 5, 28-36). Nucleophilic addition of metalated 5-(4-trifluoromethoxy)phenyl-2-pyridine (M = metal) to epoxide 22 or 22a may furnish compound

1 or la.

Scheme 8. Enantioselective epoxidation strategy

Ketone 15 or 16 may be used to prepare 9 (or 9a, the enantiomer of 9, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof) by an alternative three-step process to Scheme 7 (Scheme 9). In the presence of a chiral catalyst/reagent, trimethylsilyl-cyanide is added to 15 or 16 to furnish 23 (or 23a, the enantiomer of 23, or mixtures thereof) or 24 (or 24a, the enantiomer of 24, or mixtures thereof), respectively (S.M. Dankwardt, Tetrahedron Lett. 1998, 39, 4971-4974). Reduction of 23 (or 23a, the enantiomer of 23, or mixtures thereof) or 24 (or 24a, the enantiomer of 24, or mixtures thereof) (e.g. lithium aluminum hydride) produces 19 (or 19a, the enantiomer of 19, or mixtures thereof) or 20 (or 20a, the enantiomer of 20, or mixtures thereof). Annulation of 19 (or 19a, the enantiomer of 19, or mixtures thereof) or 20 (or 20a, the enantiomer of 20, or mixtures thereof) by treatment with sodium azide/triethylorthoformate furnishes tetrazoles 9 (or 9a, the enantiomer of 9, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof). Suzuki coupling of 9 (or 9a, the enantiomer of 9, or mixtures thereof) with 4-trifluoromethoxyphenyl-boronic acid produces 1 (or la, the enantiomer of 1, or mixtures thereof).

Scheme 9. Asymmetric cyanohydrin strategy

R’ = H or trimethylsilyl

Suzuki

R-i = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, -0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, -0(S02)-aryl, or -0(S02)-substituted aryl.

1

2-(2, 4-Difluorophenyl)-l, l-difluoro-3-(lH-tetrazol-l-yl)-l-(5-(4-(trifluoromethoxy) phenyl) pyridin-2-yl) propan-2-ol (1 or la)

White powder: *H NMR (500 MHz, CDC13): δ 8.76 (s, 1 H), 8.70 (s, 1 H), 7.97 (dd, J = 8.0, 2.0 Hz, 1 H), 7.68 (d, / = 8.5 Hz, 1 H), 7.60-7.56 (m, 3 H), 7.43-7.36 (m, 3 H), 6.80-6.76 (m, 1 H), 6.70-6.67 (m, 1 H), 5.57 (d, J = 14.5 Hz, 1 H), 5.17 (d, J = 14.5 Hz, 1 H). HPLC: 98.3%. Mass: m/z 513.9 [M++l]. HPLC: 99.8%. Optical rotation [a]D25: + 19° (C = 0.1 % in MeOH).

INTERMEDIATE 3-Br Ri = Br)

To a clean and dry 100 L jacketed reactor was added copper powder (1375 g, 2.05 equiv, 10 micron, sphereoidal, SAFC Cat # 326453) and DMSO (17.5 L, 7 vol). Next, ethyl bromodifluoroacetate (2.25 kg, 1.05 equiv, Apollo lot # 102956) was added and the resulting slurry stirred at 20-25 °C for 1-2 hours. Then 2,5-dibromopyridine (2-Br, 2.5 kg, 1.0 equiv, Alfa Aesar lot # F14P38) was added to the batch and the mixture was immediately heated (using the glycol jacket) to 35 °C. After 70 hours at 35 °C, the mixture was sampled for CG/MS analysis. A sample of the reaction slurry was diluted with 1/1 CH3CN/water, filtered (0.45 micron), and the filtrate analyzed directly. Ideally, the reaction is deemed complete if <5% (AUC) of 2,5-dibromopyridine remains. In this particular batch, 10% (AUC) of 2,5-dibromopyridine remained. However due to the already lengthy reaction time, we felt that prolonging the batch would not help the conversion any further. The reaction was then deemed complete and diluted with EtOAc (35 L). The reaction mixture was stirred at 20-35 °C for 1 hour and then the solids (copper salts) were removed by filtration through a pad of Celite. The residual solids inside the reactor were rinsed forward using EtOAc (2 x 10 L) and then this was filtered through the Celite. The filter cake was washed with additional EtOAc (3 x 10 L) and the EtOAc filtrates were combined. A buffer solution was prepared by dissolving NH4CI (10 kg) in DI water (100 L), followed by the addition of aqueous 28% NH4OH (2.0 L) to reach pH = 9. Then the combined EtOAc filtrates were added slowly to a pre-cooled (0 to 15 °C) solution of NH4C1 and NH4OH (35 L, pH = 9) buffer while maintaining T<30 °C. The mixture was then stirred for 15-30 minutes and the phases were allowed to separate. The aqueous layer (blue in color) was removed and the organic layer was washed with the buffer solution until no blue color was discernable in the aqueous layer. This experiment required 3 x 17.5 L washes. The organic layer was then washed with a 1/1 mixture of Brine (12.5 L) and the pH = 9 NH4C1 buffer solution (12.5 L), dried over MgS04, filtered, and concentrated to dryness. This provided crude compound 3-Br [2.29 kg, 77% yield, 88% (AUC) by GC/MS] as a yellow oil. The major impurity present in crude 3-Br was unreacted 2,5-dibromopyridine [10% (AUC) by GC/MS]. ‘ll NMR (CDC13) was consistent with previous lots of crude compound 3-Br. Crude compound 3-Br was then combined with similar purity lots and purified by column chromatography (5/95 EtO Ac/heptane on S1O2 gel).

INTERMEDIATE 15-Br (R, = Br)

To a clean and dry 72 L round bottom flask was added l-bromo-2,4-difluorobenzene (1586 g,

1.15 equiv, Oakwood lot # H4460) and MTBE (20 L, 12.6 vol). This solution was cooled to -70 to -75 °C and treated with n-BuLi (3286 mL, 1.15 equiv, 2.5 M in hexanes, SAFC lot # 32799MJ), added as rapidly as possible while maintaining -75 to -55 °C. This addition typically required 35-45 minutes to complete. (NOTE: If the n-BuLi is added slowly, an white slurry will form and this typically gives poor results). After stirring at -70 to -65 °C for 45 minutes, a solution of compound 3-Br (2000 g, 1.0 equiv, AMRI lot # 15CL049A) in MTBE (3 vol) was added rapidly (20-30 min) by addition funnel to the aryl lithium solution while maintaining -75 to -55 °C. After stirring for 30-60 minutes at -75 to -55 °C, the reaction was analyzed by GC/MS and showed only trace (0.5% AUC) l-bromo-2,4-difluorobenzene present. The reaction was slowly quenched with aqueous 2 M HC1 (3.6 L) and allowed to warm to room temperature. The mixture was adjusted to pH = 6.5 to 8.5 using NaHCC>3 (4 L), and the organic layer was separated. The MTBE layer was washed with brine (5% NaCl in water, 4 L), dried over MgS04, filtered, and concentrated. In order to convert the intermediate hemi-acetal to 4-Br, the crude mixture was heated inside the 20 L rotovap flask at 60-65 °C for 3 hours (under vacuum), at this point all the hemi-acetal was converted to the desired ketone 4 by !Η NMR (CDC13). This provided crude compound 4-Br [2.36 kg, 75% (AUC) by HPLC] as a brown oil that solidified upon standing. This material can then be used “as-is” in the next step without further purification.

Image result for VT1129

PATENT FOR VT1161    SIMILAR TO VT 1129

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016149486&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Synthesis of 1 or la

EXAMPLE 1

Preparation of Compound 1 X-Hydrate

Compound 1 and its preparation are described in the art, including in US Patent 8,236,962 (incorporated by reference herein). Compound 1 can then be partitioned between ethanol and water to afford Compound 1 X-hydrate.

EXAMPLE 2

Compound 1 Anhydrous Form Recrystallization

Compound 1 X-hydrate (29.1 g, 28.0 g contained 1) was suspended in 2-propanol (150 ml) and heated to 56 °C. The solution was filtered through a 0.45 μιη Nylon membrane with 2-propanol rinses. The combined filtrate was concentrated to 96.5 g of a light amber solution. The solution was transferred to a 1-L flask equipped with overhead stirring, thermocouple and addition funnel, using 2-propanol (30 ml total) to complete the transfer. The combined solution contained about 116 ml 2-propanol.

The solution was heated to 50 °C and n-heptane (234 ml) was added over 22 minutes. The resulting hazy mixture was seeded with 1 anhydrous form. After about 1 hour a good

suspension had formed. Additional n-heptane (230 ml) was added over 48 minutes. Some granular material separated but most of the suspension was a finely divided pale beige solid. After about ½ hour at 50 °C the suspension was cooled at 10 °C/h to room temperature and stirred overnight. The product was collected at 22 °C on a vacuum filter and washed with 1:4 (v/v) 2-PrOH/ n-heptane (2 x 50 ml). After drying on the filter for 1-2 hours the weight of product was 25.5 g. The material was homogenized in a stainless steel blender to pulverize and blend the more granular solid component. The resulting pale beige powder (25.37 g) was dried in a vacuum oven at 50 °C. The dry weight was 25.34 g. The residual 2-propanol and n- heptane were estimated at <0.05 wt% each by 1H NMR analysis. The yield was 90.5% after correcting the X-hydrate for solvent and water content. Residual Pd was 21 ppm. The water content was 209 ppm by KF titration. The melting point was 100.7 °C by DSC analysis.

Table 1: Data for the isolated and dried Compound 1 – X-hydrate and anhydrous forms

M.P. by DSC; Pd by ICP; Purity by the API HPLC method; Chiral purity by HPLC; water content by KF titration; residual solvent estimated from :H NMR.

Table 2: Characterisation Data for Compounds 1 (X-hydrate) and 1 (anhydrous)

Needle like crystals Needle like crystals and agglomerates

PLM

particle size >100μιη particle size range from 5μπι-100μιη

0.59%w/w water uptake at 90%RH. 0.14%w/w water uptake at 90%RH.

GVS

No sample hysteresis No sample hysteresis

XRPD

No form change after GVS experiment No form change after GVS experiment post GVS

KF 2.4%w/w H20 Not obtained

<0.001mg/ml <0.001mg/ml

Solubility

pH of saturated solution = 8.6 pH of saturated solution = 8.7

Spectral Pattern 1 Spectral Pattern 2

Charcteristic bands/ cm“1: Charcteristic bands/ cm 1:

FT-IR 3499, 3378, 3213, 3172 3162

1612, 1598, 1588, 1522, 1502 1610, 1518, 1501 931, 903, 875, 855, 828, 816 927, 858, 841, 829, 812

The structure solution of Compound 1 anhydrous form was obtained by direct methods, full-matrix least-squares refinement on F 2 with weighting w‘1 = <52{F02) + (0.0474P)2 + (0.3258P), where P = (F02+2F 2)/3, anisotropic displacement parameters, empirical absorption correction using spherical harmonics, implemented in SCALE3 ABSPACK scaling algorithm. Final wR2

= {∑[w(F02-Fc2)2]/∑[w(F02)2]m} = 0.0877 for all data, conventional Ri = 0.0343 on F values of 8390 reflections with F0 > 4a( F0), S = 1.051 for all data and 675 parameters. Final Δ/a (max) 0.001, A/a(mean), 0.000. Final difference map between +0.311 and -0.344 e A“3.

Below shows a view of two molecules of Compound 1 in the asymmetric unit of the anhydrous form showing the numbering scheme employed. Anisotropic atomic displacement ellipsoids for the non-hydrogen atoms are shown at the 50% probability level. Hydrogen atoms are displayed with an arbitrarily small radius. The absolute configuration of the molecules has been determined to be R.

EXAMPLE 3

Compound 1 Ethanol Solvate Recrystallization

Compound 1 X-hydrate (50 mg) was suspended in -40 volumes of 15% H20/EtOH. The suspension was then placed in an incubation chamber for maturation. The maturation protocol involved treating the suspension to a two-temperature cycle of 50 °C/ ambient temperature at 8 hours per cycle for 3 days with constant agitation. After maturation, the suspension was cooled in a fridge at 4°C for up to 2 days to encourage the formation of crystals. Then, the solvent was removed at RT and the sample was vacuum dried at 30°C -35°C for up to 1 day. Suitable crystals formed on cooling were harvested and characterized.

Table 4: Single Crystal Structure of 1 Ethanol solvate

Molecular formula C25H22F7N5O3

The structure solution of Compound 1 ethanol solvate was obtained by direct methods, full-matrix least-squares refinement on F 2 with weighting w‘1 = σ2^2) + (0.0450P)2 + (0.5000P), where P = (F02+2F 2)/3, anisotropic displacement parameters, empirical absorption correction using spherical harmonics, implemented in SCALE3 ABSPACK scaling algorithm. Final wR2 = {∑[w(F02-F 2)2]/∑[w(F02)2]m} = 0.0777 for all data, conventional Ri = 0.0272 on F values of 4591 reflections with F0 > 4σ( F0), S = 1.006 for all data and 370 parameters. Final Δ/σ (max) 0.000, A/a(mean), 0.000. Final difference map between +0.217 and -0.199 e A“3.

Below shows a view of the asymmetric unit of the ethanol solvate from the crystal structure showing the numbering scheme employed. Anisotropic atomic displacement ellipsoids for the non-hydrogen atoms are shown at the 50% probability level. Hydrogen atoms are displayed with an arbitrarily small radius. The asymmetric unit shows stoichiometry of 1 : 1 for solvent of crystallisation to Compound 1.

EXAMPLE 4

Compound 1 1.5 Hydrate Recrystallization

Compound 1 X-hydrate (50 mg) was suspended in -40 volumes of 15% Η20/ΙΡΑ. The suspension was then placed in an incubation chamber for maturation. The maturation protocol involved treating the suspension to a two-temperature cycle of 50 °C/ ambient temperature at 8 hours per cycle for 3 days with constant agitation. After maturation, the suspension was cooled in a fridge at 4°C for up to 2 days to encourage the formation of crystals. Then, the solvent was removed at RT and the sample was vacuum dried at 30°C -35°C for up to 1 day. Suitable crystals formed on cooling were harvested and characterized.

Table 5: Single Crystal Structure of 1 1.5 Hydrate

The structure solution of Compound 1 1.5 hydrate was obtained by direct methods, full-matrix least-squares refinement on F 2 with weighting w‘1 = ^(F 2) + (0.1269P)2 + (0.0000P), where P = (F02+2F 2)/3, anisotropic displacement parameters, empirical absorption correction using spherical harmonics, implemented in SCALE3 ABSPACK scaling algorithm. Final wR2 = {∑[w(F 2-F 2)2]/∑[w(F 2)2] m} = 0.1574 for all data, conventional Ri = 0.0668 on F values of 2106 reflections with F0 > 4σ( F0), S = 1.106 for all data and 361 parameters. Final Δ/σ (max) 0.000, A/a(mean), 0.000. Final difference map between +0.439 and -0.598 e A“3.

Below shows a view of the asymmetric unit of the 1.5 hydrate from the crystal structure showing the numbering scheme employed. Anisotropic atomic displacement ellipsoids for the non-hydrogen atoms are shown at the 50% probability level. Hydrogen atoms are displayed with an arbitrarily small radius. The asymmetric unit shows stoichiometry of 1.5: 1 for water to Compound 1.

EXAMPLE 5

Human Pharmacokinetic Comparison of Compound 1 X-Hydrate and Compound 1 Anhydrous Form

Table 6 compares human multiple-dose pharmacokinetic (PK) parameters between dosing with Compound 1 X-hydrate and Compound 1 Anhydrous form. Compound 1 X-hydrate was dosed at 600 mg twice daily (bid) for three days followed by dosing at 300 mg once daily (qd) for 10 days. Compound 1 Anhydrous form was dosed at 300 mg qd for 14 days. Despite the higher initial dosing amount and frequency (i.e., 600 mg bid) of Compound 1 X-hydrate, Compound 1 Anhydrous form surprisingly displayed higher maximal concentration (Cmax) and higher area-under-the-curve (AUC) than Compound 1 X-hydrate.

Table 6. Comparison of Multiple Dose PK between Compound 1 X-Hydrate and Compound 1

Anhydrous Polymorph

Further characterization of the various polymorph forms of compound 1 are detailed in the accompanying figures.

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015143154

Examples

General Experimental Procedures

Definitions of variables in the structures in schemes herein are commensurate with those of corresponding positions in the formulae delineated herein.

Synthesis of 1 or la

la

A process to prepare enantiopure compound 1 or la is disclosed. Syntheses of lor la may be accomplished using the example syntheses that are shown below (Schemes 1-4). The preparation of precursor ketone 3-Br is performed starting with reaction of 2,5-dibromo-pyridine with ethyl 2-bromo-difluoroacetate to produce ester 2-Br. This ester is reacted with morpholine to furnish morpholine amide 2b-Br, followed by arylation to provide ketone 3-Br.

Scheme 1. Synthesis of ketone 3-Br

Ketone 3 may be prepared in an analogous fashion as described in Scheme 1 starting from corresponding substituted 2-bromo-pyridines, which can be prepared using according to synthetic transformations known in the art and contained in the references cited herein (Scheme 2).

Scheme 2. Synthesis of ketone 3

R1 = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)-substituted aryl, -0(C=0)-0-alkyl, – 0(C=0)-0-substituted alkyl, -0(C=0)-0-aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, 0(S02)-aryl, or -0(S02)-substituted aryl.

Alternatively, compound 1 (or la, the enantiomer of 1, or mixtures thereof) can be prepared according to Scheme 3 utilizing amino-alcohols ±4b or ±1-6. Epoxides 4 and 5 can be prepared by reacting ketones 3 and 1-4 with trimethylsulfoxonium iodide (TMSI) in the presence of a base (e.g., potassium i-butoxide) in a suitable solvent or a mixture of solvents (e.g., DMSO or THF). Also, as indicated in Scheme 3, any of pyridine compounds, 3, 4, ±4b, 4b, or 6, can be converted to the corresponding 4-CF3O-PI1 analogs (e.g., 1-4, 5, ±1-6, 1-6*, or 1 or the corresponding enantiomers, or mixtures thereof) by cross-coupling with (4-trifluoromethoxyphenyl)boronic acid (or the corresponding alkyl boronates or pinnacol boronates or the like), in a suitable solvent system (e.g., an organic-aqueous solvent mixture), in the presence of a transition metal catalyst (e.g., (dppf)PdCl2; dppf = 1,1′-(diphenylphosphino)ferrocene), and in the presence of a base (e.g., KHCO3, K2CO3, CS2CO3, or Na2CC>3, or the like). Epoxides 4 and 5 can then be converted into amino-alcohols ±4b and ±1-6 through ammonia-mediated epoxide opening using ammonia in a suitable solvent (e.g., MeOH, EtOH, or water). Racemic amino-alcohols ±4b and ±1-6 can then be enantio-enriched by exposure to a chiral acid (e.g., tartaric acid, di-benzoyltartaric acid, or di-p-toluoyltartaric acid or the like) in a suitable solvent (e.g., acetonitrile, isopropanol, EtOH, or mixtures thereof, or a mixture of any of these with water or MeOH; preferably acetonitrile or a mixture of acetonitrile and MeOH, such as 90:10, 85: 15, or 80:20 mixture) to afford compounds 4b (or 4c, the enantiomer of 4b, or mixtures thereof) or 1-6* (or 1-7*, the enantiomer of 1-6*, or mixtures thereof). Subsequent treatment with TMS-azide in the presence of trimethylorthoformate and sodium acetate in acetic acid would yield compounds 20 (or 20a, the enantiomer of 20, or mixtures thereof) or 1 (or la, the enantiomer of 1, or mixtures thereof) (US 4,426,531).

Scheme 3. Synthesis of 1 or la via TMSI Epoxidation Method

R-ι = halo, -0(C=0)-alkyl, -0(C=0)-substituted alkyl, -0(C=0)-aryl, -0(C=0)- substituted aryl, -0(C=0)-0-alkyl, -0(C=0)-0-substituted alkyl, -0(C=0)-0- aryl, -0(C=0)-0-substituted aryl, -0(S02)-alkyl, -0(S02)-substituted alkyl, – 0(S02)-aryl, or -0(S02)-substituted aryl.

Compound 1 (or la, the enantiomer of 1, or mixtures thereof) prepared by any of the methods presented herein can be converted to a sulfonic salt of formula IX (or IXa, the enantiomer of

IX, or mixtures thereof), as shown in Scheme 4. This can be accomplished by a) combining compound 1 (or la, the enantiomer of 1, or mixtures thereof), a crystallization solvent or crystallization solvent mixture (e.g., EtOAc, iPrOAc, EtOH, MeOH, or acetonitrile, or o

Z-S-OH

combinations thereof), and a sulfonic acid o (e.g., Z = Ph, p-tolyl, Me, or Et), b) diluting the mixture with an appropriate crystallization co-solvent or crystallization co-solvent mixture (e.g., pentane, methyl i-butylether, hexane, heptane, or toluene, or combinations thereof), and c) filtering the mixture to obtain a sulfonic acid salt of formula IX (or IXa, the enantiomer of IX, or mixtures thereof).

Scheme 4. Synthesis of a Sulfonic Acid Salt of Compound 1 or la

EXAMPLE 1: Preparation of l-(2,4-difluorophenyl)-2,2-difluoro-2-(5-(4- (trifluoromethoxy)phenyl)pyridin-2-yl)ethanone (1-4).

la. ethyl 2-(5-bromopyridin-2-yl)-2,2-difluoroacetate (2)

2-Br
Typical Procedure for Preparing 2-Br

Copper ( 45μιη, 149g, 0.198moles, 2.5 equiv) was placed into a 3L, 3-neck round bottom flask equipped with a condenser, thermocouple, and an overhead stirrer. DMSO (890 mL, 4.7 vol. based on ethyl 2-bromo-2,2-difluoroacetate) and 14mL of concentrated sulfuric acid was added and the mixture stirred for 30 minutes. The mixture self-heated to about 31°C during the stir time. After cooling the contents to 23°C, 2,5-dibromopyridine 1 (277g, 1.17 moles, 1.5 eq) was added to the reaction mixture. The temperature of the contents decreased to 16°C during a 10 minute stir time. 2-bromo-2,2-difluoroacetate (190 g, 0.936 moles, 1.0 eq) was added in one portion and the mixture stirred for 10 min. The flask contents were warmed to 35°C and the internal temperature was maintained between 35-38° for 18 h. In-process HPLC showed 72% desired 2-Br. The warm reaction mixture was filtered through filter paper and the collected solids washed with 300mL of 35°C DMSO. The solids were then washed with 450mL of n-heptane and 450mL of MTBE. The collected filtrate was cooled to about 10°C and was slowly added 900mL of a cold 20% aqueous NH4C1 solution, maintaining an internal temperature of <16°C during the addition. After stirring for 15 minutes, the layers were settled and separated. The aqueous layer was extracted 2 X 450mL of a 1: 1 MTBE: n-heptane mixture. The combined organic layers were washed 2 X 450mL of aqueous 20% NH4CI and with 200mL of aqueous 20% NaCl. The organic layer was dried with 50g MgS04 and the solvent removed to yield 2-Br as a dark oil. Weight of oil = 183g ( 70% yield by weight) HPLC purity ( by area %) = 85%. *H NMR (400 MHz, d6-DMSO) : 58.86 (m, 1H), 8.35 ( dd, J= 8.4, 2.3Hz, 1H), 7.84 (dd, J= 8.3, 0.6Hz, 1H), 4.34 ( q, J= 7.1Hz, 2H), 1.23 ( t, J= 7.1Hz, 3H). MS m/z 280 ( M+H+), 282 (M+2+H+).

lb. 2-(5-bromopyridin-2-yl)-2,2-difluoro-l-morpholinoethanone (2b-Br)

Table 2 illustrates the effects of the relative proportions of each of the reagents and reactants, and the effect of varying the solvent had on the overall performance of the transformation as measured by the overall yield and purity of the reaction.

Table 2. Process Development for the Preparation of compound 2b-Br

Note: All reactions were conducted at 22- 25°C

Typical Procedure for Converting 2-Br to 2b-Br

Crude ester 2-Br (183g, 0.65moles) was dissolved in 1.5L of n-heptane and transferred to a 5L 3-neck round bottom flask equipped with a condenser, an overhead stirrer and a thermocouple. Morpholine ( 248g, 2.85 moles, 4.4 equiv.) was charged to the flask and the mixture warmed to 60°C and stirred for 16 hours. In-process HPLC showed <1 % of ester 2-Br. The reaction mixture was cooled to 22-25 °C and 1.5L of MTBE was added with continued cooling of the mixture to 4°C and slowly added 700mL of a 30%, by weight, aqueous citric acid solution. The temperature of the reaction mixture was kept < 15°C during the addition. The reaction was stirred at about 14°C for one hour and then the layers were separated. The organic layer was washed with 400mL of 30%, by weight, aqueous citric acid solution and then with 400mL of aqueous 9% NaHC03. The solvent was slowly removed until 565g of the reaction mixture

remained. This mixture was stirred with overhead stirring for about 16 hours. The slurry was filtered and the solids washed with 250mL of n-heptane. Weight of 2b-Br = 133g. HPLC purity (by area %) 98%.

This is a 44% overall yield from 2,5-dibromopyridine.

*H NMR (400 MHz, d6-DMSO): 58.86 (d, J= 2.3Hz, 1H), 8.34 (dd, J= 8.5, 2.3Hz, 1H), 7.81 (dd, J = 8.5, 0.5Hz, 1H), 3.63-3.54 ( m, 4H), 3.44-3.39 (m, 2H), 3.34-3.30 ( m, 2H). MS m/z 321 (M+H+), 323 (M+2+H+).

lc. 2-(5-bromopyridin-2-yl)-l-(2,4-difluorophenyl)-2,2-difluoroethanone (3-Br)

Process Development

Table 3 illustrates the effects of the relative proportions of each of the reagents and reactants, and the effect of varying the temperature had on the overall performance of the transformation as measured by the overall yield and purity of the reaction.

Table 3. Process Development for the Preparation of bromo-pyridine 3-Br

Typical Procedure for Converting 2b-Br to 3-Br

Grignard formation:

Magnesium turnings (13.63 g, 0.56 moles) were charged to a 3-neck round bottom flask equipped with a condenser, thermocouple, addition funnel, and a stir bar. 540 mL of anhydrous tetrahydrofuran was added followed by l-Bromo-2,4-difluorobenzene (16.3 mL, 0.144 moles). The contents were stirred at 22-25°C and allowed to self -heat to 44°C. 1- Bromo-2,4-difluorobenzene ( 47mL, 0.416 moles) was added to the reaction mixture at a rate that maintained the internal temperature between 40-44°C during the addition. Once the addition was complete, the mixture was stirred for 2 hours and allowed to cool to about 25° during the stir time.

This mixture was held at 22-25°C and used within 3-4 hours after the addition of l-bromo-2,4-difluorobenzene was completed.

Coupling Reaction

Compound 2b-Br (120 g, 0.0374 moles) was charged to a 3-neck round bottom flask equipped with a condenser, thermocouple, and an overhead stirrer. 600 mL of anhydrous

tetrahydrofuran was added. The flask contents were stirred at 22°C until a clear solution was obtained. The solution was cooled to 0-5°C. The previously prepared solution of the Grignard reagent was then added slowly while maintaining the reaction temperature at 0-2°C. Reaction progress was monitored by HPLC. In-process check after 45 minutes showed <1% amide 2b-Br remaining. 2 N aqueous HC1 (600 mL, 3 vol) was added slowly maintaining the temperature below 18°C during the addition. The reaction was stirred for 30 minutes and the layers were separated. The aqueous layer was extracted with 240mL MTBE. The combined organic layers were washed with 240mL of aqueous 9% NaHCC>3 and 240mL of aqueous 20% NaCl. The organic layer was dried over 28g of MgS04 and removed the solvent to yield 3-Br (137g) as an amber oil.

HPLC purity ( by area %) = -90%; *H NMR (400 MHz, d6-DMSO) : 58.80 (d, J= 2.2Hz, 1H), 8.41 ( dd, J= 8.3, 2.3Hz, 1H), 8.00 (m, 2H), 7.45 ( m, 1H), 7.30 ( m, 1H). MS m/z 348 (M+H+), 350 (M+2+H+).

Id. l-(2,4-difluorophenyl)-2,2-difluoro-2-(5-(4-(trifluoromethoxy)phenyl)pyridin-2-yl)ethanone (1-4)

Typical Procedure for Converting 3-Br to 1-4

Into a 250 mL reactor were charged THF (45 mL), water (9.8 mL), bromo-pyridine 3-Br (6.0 g, 17.2 mmoles), 4-(trifluoromethoxy)phenylboronic acid (3.57 g, 17.3 mmoles), and Na2CC>3 (4.55 g, 42.9 mmoles). The stirred mixture was purged with nitrogen for 15 min. The catalyst (Pd(dppf)Cl2 as a CH2C12 adduct, 0.72 g, 0.88 mmoles) was added, and the reaction mixture was heated to 65 °C and held for 2.5 h. The heat was shut off and the reaction mixture was allowed to cool to 20-25 °C and stir overnight. HPLC analysis showed -90% ketone 1-4/hydrate and no unreacted bromo-pyridine 3-Br. MTBE (45 mL) and DI H20 (20 mL) were added, and the quenched reaction was stirred for 45 min. The mixture was passed through a plug of Celite (3 g) to remove solids and was rinsed with MTBE (25 mL). The filtrate was transferred to a separatory funnel, and the aqueous layer drained. The organic layer was washed with 20% brine (25 mL). and split into two portions. Both were concentrated by rotovap to give oils (7.05 g and 1.84 g, 8.89 g total, >100% yield, HPLC purity -90%). The larger aliquot was used to generate hetone 1-4 as is. The smaller aliquot was dissolved in DCM (3.7 g, 2 parts) and placed on a pad of Si02 (5.5 g, 3 parts). The flask was rinsed with DCM (1.8 g), and the rinse added to the pad. The pad was eluted with DCM (90 mL), and the collected filtrate concentrated to give an oil (1.52 g). To this was added heptanes (6 g, 4 parts) and the mixture stirred. The oil crystallized, resulting in a slurry. The slurry was stirred at 20-25 °C overnight. The solid was isolated by vacuum filtration, and the cake washed with heptanes (-1.5 mL). The cake was dried in the vacuum oven (40-45 °C) with a N2 sweep. 0.92 g of ketone 1-4 was obtained, 60.1% yield (corrected for aliquot size), HPLC purity = 99.9%.

TMSI Epoxidation Method

3d. 2-((2-(2,4-difluorophenyl)oxiran-2-yl)difluoromethyl)-5-(4-(trifluoromethoxy)phenyl)pyridine (5)

Typical Procedure for Converting 1-4 to 5

i-BuOK (2.22 g, 19.9 mmoles), TMSI (4.41 g, 20.0 mmoles), and THF (58.5 mL) were charged to a reaction flask, and the cloudy mixture was stirred. DMSO (35.2 mL) was added, and the clearing mixture was stirred at 20-25°C for 30 min before being cooled to 1-2°C.

Ketone 1-4 (crude, 5.85 g, 13.6 mmoles) was dissolved in THF (7.8 mL), and the 1-4 solution was added to the TMSI mixture over 12.75 min, maintaining the temperature between 1.5 and 2.0°C. The reaction was held at 0-2°C. After 1 h a sample was taken for HPLC analysis, which showed 77.6% epoxide 5, and no unreacted ketone 1-4. The reaction was quenched by the slow addition of 1 N HC1 (17.6 mL), keeping the temperature below 5°C. After 5 min 8% NaHCC>3 (11.8 mL) was added slowly below 5°C to afford a pH of 8. The reaction mixture was transferred to a separatory funnel, and the layers were separated. The aqueous layer was extracted with MTBE (78 mL), and the combined organic layers were washed with 20% NaCl (2 x 20 mL). After concentration, 7.36 g of a dark oil was obtained. HPLC of the crude oil shows it contained 75% epoxide 5. The oil was dissolved in DCM (14.7 g, 2 parts) and the solution placed on a pad of Si02 (22 g, 3 parts). The flask was rinsed with DCM (7.4 g, 1 part) and the rinse placed on the pad. The pad was eluted with DCM (350 mL) to give an amber filtrate. The filtrate was concentrated by rotovap, and when space in the flask allowed, heptane (100 mL) was added. The mixture was concentrated until 39.4 g remained in the flask, causing solid to form. The suspension was stirred for 70 min at 20-25°C. Solid was isolated by vacuum filtration, and the cake washed with heptane (10 mL) and pulled dry on the funnel. After drying in a vacuum oven (40-45 °C) with a N2 sweep, 3.33 g solid was obtained, 55.1% yield from bromo-pyridine 3, HPLC purity = 99.8%.

3e. 3-amino-2-(2,4-difluorophenyl)-l,l-difluoro-l-(5-(4-(trifluoromethoxy)phenyl)pyridin-2-yl)propan-2-ol (±1-6)

Process Development

Table 8 illustrates the effects of the relative proportions of each of the reagents and reactants, the effect of varying the solvent, and the effect of varying the temperature had on the overall performance of the transformation as measured by the overall yield and purity of the reaction. Table 8. Process Development for the Preparation of ±1-6

Typical Procedure for Converting 5 to +1-6

Epoxide 5 (2.17 g, 4.89 mmoles) was combined in a glass pressure tube with methanol (48 mL) and aqueous ammonia (19.5 mL). The tube was sealed and placed in an oil bath held at 54°C, with stirring. After 15 h the tube was removed from the bath, cooled, and the reaction sampled for HPLC, which showed 93.6% amino-alcohol ±1-6 and 6.0% di-adducts. To the reaction were added MTBE (48 mL) and 20% NaCl (20 mL). The layers were separated and the aqueous layer extracted with MTBE (20 mL). The combined organic layers were washed with H20 (20 mL) and transferred to a rotovap flask. Heptane (20 mL) was added, and the solution was concentrated until 16.9 g remained in the flask. An H20 layer appeared in the flask, and was pipetted out, leaving 12.8 g. Compound 1-6 seed was added, and the crystallizing mixture was stirred at 20-25 °C overnight. The flask was cooled in an ice bath for 2 h prior to filtration, and the isolated solid was washed with cold heptane (5 mL), and pulled dry on the funnel. After drying in a vacuum oven (40-45°C) for several hours 1.37 g of amino-alcohol ±1-6 was obtained, 60.8% yield, HPLC purity = 98.0%.

3f . 3-amino-2-(2,4-difluorophenyl)- 1 , 1-difluoro- 1 -(5-(4-(trifluoromethoxy)phenyl)pyridin-2- yl)propan-2-ol (1-6* or 1-7*)

Process Development

Table 9 illustrates the initial screen performed surveying various chiral acid/solvent combinations. All entries in Table 9 were generated using 0.1 mmoles of amino-alcohol ±1-6, 1 equivalent of the chiral acid, and 1ml of solvent.

Table 9. Resolution of ±1-6 (Initial Screen)

Since the best results from Table 9 were generated using tartaric acid and di-p-toluoyltartaric acid, Table 10 captures the results from a focused screen using these two chiral acids and various solvent combinations. All entries in Table 10 were performed with 0.2 mmoles of amino-alcohol ±1-6, 87 volumes of solvent, and each entry was exposed to heating at 51 °C for lh, cooled to RT, and stirred at RT for 24h.

Table 10. Resolution of ±1-6 (Focused Screen)

Each of the three entries using di-p-toluoyltartaric acid in Table 10 resulted in higher levels of enantio-enrichment when compared to tartaric acid. As such, efforts to further optimize the enantio-enrichment were focusing on conditions using di-p-toluoyltartaric acid (Table 11).

Ό.6 equivalents used

ee sense was opposite from the other entries in the table (i.e., enantiomer of 1-6*)

Typical Procedure for Converting +1-6 to 1-6* or 1-7*

(This experimental procedure describes resolution of ±1-6, but conditions used for DPPTA resolution of 1-6 or 1-7 are essentially the same.)

Amino-alcohol ±1-6 (7.0 g, 15 mmoles) was dissolved in a mixture of acetonitrile (84 mL) and methanol (21 mL). (D)-DPTTA (5.89 g, 15 mmoles) was added, and the reaction was warmed to 50°C and held for 2.5 h. The heat was then removed and the suspension was allowed to cool and stir at 20-25 °C for 65 h. The suspension was cooled in an ice bath and stirred for an additional 2 h. Solid was isolated by vacuum filtration, and the cake was washed with cold 8:2 ACN/MeOH (35 mL). After drying at 50°C, 5.18 g of 1-6* or l-7*/DPPTA salt was isolated, HPLC purity = 99.0, ee = 74.

The 1-6* or l-7*/DPPTA salt (5.18 g) was combined with 8:2 ACN/MeOH (68 mL) and the suspension was heated to 50°C and held for 20 min. After cooling to 20-25 °C the mixture was stirred for 16 h. Solids were isolated by vacuum filtration, and the cake washed with cold 8:2 ACN/MeOH (30 mL), and pulled dry on the funnel. 2.82 g of 1-6* or l-7*/DPPTA salt was obtained, 44.4% yield (from crude ±1-6), ee = 97.5. The resulting solids were freebased to provide 1-6* or 1-7* with the same achiral and chiral purity as the DPPTA salt.

EXAMPLE 4: Preparation of 2-(2.4-difluorophenyl -l.l-difluoro-3-(lH-tetrazol-l-yl -l-(5-(4-(trifluoromethoxy)phenyl)pyridin-2-yl)propan-2-ol (1 or la).

The procedure used to generate compound 1 or la is as described in US 4,426,531. Table 13 illustrates the efficient and quantitative nature of this procedure as performed on amino- alcohol 1-6* or 1-7* produced from both the TMS-cyanohydrin method and the TMSI- epoxidation method.

Table 13. Formation of Compound 1 or la

EXAMPLE 5: 2-(2.4-difluorophenyl -l.l-difluoro-3-(lH-tetrazol-l-yl -l-(5-(4- (trifluoromethoxy)phenyl)pyridin-2-yl)propan-2-ol benzenesulfonate (1 or la-BSA).

Typical Procedure for Converting 1 or la to 1 or la-BSA

46.6 g of compound 1 or la was dissolved in ethylacetate (360ml). The solution was filtered through a glass microfiber filter and placed in a 2 L reaction flask equipped with an overhead stirrer, condenser, and a J-Kem thermocouple. Pharma-grade benzenesulfonic acid (BSA, 14.39g, leq) was dissolved in ethyl acetate (100ml). The BSA solution was filtered through a glass microfiber filter and added to the stirred 1 or la solution in one portion. The mixture was warmed to 60-65 °C; precipitation of the 1 or la/BSA salt occurred during the warm up period. The slurry was held for 60 minutes at 60-65 °C. The suspension was allowed to slowly cool to 22 °C and was stirred at 20-25 °C for 16 hours. n-Heptane (920ml) was charged in one portion and the suspension was stirred at 22 °C for an additional 90 minutes. The slurry was filtered and the collected solids washed with n-heptane (250ml). The isolated solids were placed in a vacuum oven at 50 °C for 16 hours. 52.26g (86% yield) of 1 or la

benzenesulfonate was obtained.

*H NMR (400 MHz, DMSO-d6 + D20): 89.16 (s, 1H), 8.95 (d, J = 2.1 Hz, 1H), 8.26 (dd, J = 8.2, 2.3 Hz, 1H), 7.96-7.89 (m, 2H), 7.66-7.61 (m, 2H), 7.59 (dd, J = 8.3, 0.4 Hz, 1H), 7.53 (br d, J = 8.0 Hz, 2H), 7.38-7.15 (m, 5H), 6.90 (dt, J = 8.3, 2.5 Hz, 1H), 5.69 (d, J = 14.8 Hz, 1H), 5.15 (d, J = 15.2 Hz, 1H).

Further results are in Table 14.

Table 14. Formation of 1 or la-BSA

( ) (%ee) Yield Purity (%) ee

97.9 95.9 84% 98.2 97.1

Figures 1-2 contain the analytical data for 1 or la-BSA prepared by the TMSI-epoxidation process.

EXAMPLE 6: 5-bromo-2-((2-(2,4-difluorophenyl)oxiran-2-yl)difluoromethyl)pyridine -Br).

Typical Procedure for Converting 3-Br to 4-Br

KOtBu ( 41.7g, 0.372moles, 1.05 equiv) and trimethylsulfoxonium iodide ( 85.7g,

0.389moles, 1.1 equiv) were charged to a 3L 3-neck round bottom flask equipped with an overhead stirrer, a thermocouple and an addition funnel. 1.2L of anhydrous THF and 740mL of DMSO were added to the flask and stirred at 22-25 °C for 70 minutes. The contents were cooled to 0°C. Crude ketone 3 was dissolved in 250mL of anhydrous THF and slowly added the ketone 3-Br solution to the reaction mixture over 20 minutes while maintaining a reaction temperature at < 3°C during the addition and stirred at 0°C for one hour. In-process HPLC showed <1% ketone 3-Br remaining. 200mL of IN HC1 was slowly added maintaining a reaction temperature of < 6°C during the addition. After stirring for 30 minutes the layers were separated and the aqueous layer was extracted with 375mL of MTBE. The combined organic layers were washed with 375mL of aqueous 9% NaHCC>3 and with 375mL of aqueous 20% NaCl. The solvent was removed to yield 4-Br as a brown waxy solid.

Weight of crude epoxide 4-Br = 124.6g; *H NMR (400 MHz, d6-DMSO) : 58.82 (d, J= 2.3Hz, 1H), 8.21 ( dd, J= 8.3, 2.3Hz, 1H), 7.50 (dd, J= 8.3, 0.5Hz, 1H), 7.41 ( m, 1H), 7.25 ( m, 1H), 7.10 (m,lH), 3.40 ( d, J= 4.5Hz, 1H), 3.14 ( m, 1H). MS m/z 362 (M+H+), 364 (M+2+H+).

EXAMPLE 7: 3-amino-l-(5-bromopyridin-2-yl)-2-(2,4-difluorophenyl)-l,l-difluoropropan-2-ol (4b-Br).

Typical Procedure for Converting 4-Br to 4b-Br

Crude epoxide 4-Br ( 54.4g, 0.15moles) was placed into a Schott autoclave bottle equipped with a stir bar. 550mL of MeOH was added to the bottle and stirred for 90 minutes at 22-25 °C. Concentrated NH4OH ( 550mL, 7.98 moles, 53 equiv) was added to the epoxide 4-Br

solution. The bottle was sealed and placed in an oil bath at 55 °C. The mixture was stirred at 55°C for 17 hours. The bottle was removed from the oil bath and cooled to 22-25°C. In-process HPLC showed <1% epoxide 4-Br remaining. The solvent was removed via rotary evaporation until 362g ( 37%) of the reaction mass remained. 500mL of MTBE was added and cooled the mixture to 8°C. 500mL of 6N HCl was slowly added maintaining the reaction temperature between 8 – 12°C during the addition. After stirring for 10 minutes, the layers were separated. The MTBE layer was extracted with 350mL of 6N HCl. The combined aqueous layers were washed with 250mL MTBE and 2 X 250mL heptane. MTBE, 250mL, was added to the aqueous layer and the mixture was cooled to 2°C. 344g of KOH was dissolved in 500mL of water. The KOH solution was slowly added to the reaction mixture over one hour while maintaining the temperature at <19°C. After stirring for 15 minutes, the layers were separated. The aqueous layer was extracted with 250mL MTBE. The combined organic layers were washed with 250mL of aqueous 20% NaCl and the solvent was removed to yield ±4b-Br as a dark oil. Weight of crude amino alcohol ±4b-Br = 46.0g. HPLC purity ( by area %) = 92%; *H NMR (400 MHz, d6-DMSO) : 58.67 (d, J= 2.2Hz, 1H), 8.15 ( dd, J= 8.6, 2.4Hz, 1H), 7.46 (m, 1H), 7.40 ( dd, J= 8.5, 0.7Hz, 1H), 7.10 ( m, 1H), 7.00 (m,lH), 3.37 (dd, J= 13.7, 2.1Hz, 1H), 3.23 ( dd, J= 13.7, 2.7, 1H). MS m/z 379 (M+H+), 381 (M+2+H+).

EXAMPLE 8: 3-amino-l-(5-bromopyridin-2-yl -2-(2.4-difluorophenyl -l.l-difluoropropan-2-ol (4b-Br or 4c-Br).

Typical Procedure for Converting 4-Br to 4b-Br or 4c-Br

Crude amino alcohol ±4b-Br ( 42.4, O. llmoles) was dissolved in 425mL of 8:2 IPA: CH3CN. The solution was charged to a 1L 3-neck round bottom flask equipped with a condenser, overhead stirrer and a thermocouple. Charged di-p-toluoyl-L-tartaric acid ( 21.6g, 0.056moles, 0.5 equiv) to the flask and warmed the contents to 52°C. The reaction mixture was stirred at 52°C for 5 hours, cooled to 22-25°C and stirred for 12 hours. The slurry was cooled to 5-10°C and stirred for 90 minutes. The mixture was filtered and collected solids washed with 80mL of cold CH3CN. The solids were dried in a vacuum oven 45-50°C. Weight of amino alcohol/ DPTTA salt = 17.4g

Chemical purity by HPLC ( area %) = 98.5%; Chiral HPLC= 98.0% ee.

13.60g of the amino alcohol/ DPTTA salt was placed into a 250mL flask with a stir bar and to this was added lOOmL of MTBE and lOOmL of 10% aqueous K2CO3solution. The reaction was stirred until complete dissolution was observed. The layers were separated and the aqueous layer was extracted with 50mL of MTBE. The combined MTBE layers were washed with 50mL of 20% aqueous NaCl and the solvent removed to yield 8.84 (98%) of 4b-Br or 4c-Br as a light yellow oil.

EXAMPLE 9: 3-amino-2-(2,4-difluorophenyl)-l J-difluoro-l-(5-(4-(trifluoromethoxy)phenyl)pyridin-2-yl)propan-2-ol (1-6* or 1-7*).

Typical Procedure for Converting 4b-Br or 4c-Br to 1-6* or 1-7*

Amino alcohol 4b-Br or 4c-Br (8.84g, 0.023moles, 1 equiv) was dissolved in 73mL of n-propanol. The solution was transferred to a 250mL 3-neck round bottom flask equipped with a condenser, thermocouple, stir bar and septum. 17mL of water was added and stirred at 22-25°C for 5 minutes. To the reaction was added K2CO3 ( 9.67g, 0.07moles, 3 equiv), 4-(trifluoromethoxy)phenylboronic acid ( 5.76g, 0.028moles, 1.2 equiv.) and Pd(dppf)Cl2 as a CH2Cl2 adduct ( 0.38g, 0.47mmoles, 0.02 equiv) to the flask. After the mixture was purged with nitrogen for 10 minutes, the reaction was then warmed to 85-87°C and stirred at 85-87°C for 16 hours. HPLC analysis showed < 1% of the amino alcohol 4b-Br or 4c-Br remaining. The mixture was cooled to 22-25 °C, then 115mL of MTBE and 115mL of water were added and stirred for 30 minutes. The layers were separated and the organic layer was washed with 2 X 60mL of 20% aqueous NaCl. The solvent was removed to yield 12.96g ( 121% yield) of 1-6* or 1-7* as a crude dark oil. It should be noted that the oil contains residual solvent, Pd and boronic acid impurity.

‘ll NMR (400 MHz, d6-DMSO) : 58.90 (d, J= 2.2Hz, 1H), 8.22 ( dd, J= 8.3, 2.3Hz, 1H), 7.91 (m, 2H), 7.54 ( m, 4H), 7.14 ( m, 1H), 7.02 (m,lH), 3.41 (m, 1H), 3.27 ( dd, J= 14.0, 2.7, 1H). MS m/z 461 (M+H+)

CLIP

Med. Chem. Commun., 2016,7, 1285-1306

DOI: 10.1039/C6MD00222F

Fungal infections directly affect millions of people each year. In addition to the invasive fungal infections of humans, the plants and animals that comprise our primary food source are also susceptible to diseases caused by these eukaryotic microbes. The need for antifungals, not only for our medical needs, but also for use in agriculture and livestock causes a high demand for novel antimycotics. Herein, we provide an overview of the most commonly used antifungals in medicine and agriculture. We also present a summary of the recent progress (from 2010–2016) in the discovery/development of new agents against fungal strains of medical/agricultural relevance, as well as information related to their biological activity, their mode(s) of action, and their mechanism(s) of resistance.

 

Graphical abstract: A complex game of hide and seek: the search for new antifungals
CLIP
Design and optimization of highly-selective fungal CYP51 inhibitors
  • Viamet Pharmaceuticals Inc., Durham, NC 27703, USA

Image for figure Scheme 1

able 3.Antifungal activity of difluoromethyl-pyridyl-benzenes

Antifungal activity of difluoromethyl-pyridyl-benzenes
Compound R C. albicans MICa T. rubrum MICa CYP3A4 IC50b Selectivity indexc
7a Cl ⩽0.001 0.004 36 9000
7b CF3 ⩽0.001 0.002 54 27,000
7c

VT 1129

OCF3 ⩽0.001 ⩽0.001 79 >79,000
7d

VT 1161

OCH2CF3 ⩽0.001 ⩽0.001 65 >65,000
Itraconazole 0.016 0.062 0.07 1.1
aMinimum concentration that achieved 50% inhibition of fungal growth; MIC units in μg/mL.5
bInhibition of CYP3A4 measured in microsomes obtained from pooled human hepatocytes, IC50 units in μM.8
cIn vitro selectivity calculated as CYP3A4 IC50/T. rubrum MIC.
(R)-(+)-Enantiomers (7a7d) were isolated from racemates using chiral chromatography.
16 Hoekstra, W.J.; Schotzinger, R.J.; Rafferty, S.W. U.S. Patent 8,236,962 issued Aug. 7, 2012.

update………….

QUILSECONAZOLE, VT 1129, New Patent, WO, 2017049080, Viamet

str1 Figure imgf000002_0001

<p>Formula (I)</p> <p>Crizotinib is a potent small-molecule inhibitor of c-Met/HGFR (hepatocyte growth factor receptor) kinase and ALK (anaplastic lymphoma kinase) activity. Enantiomerically pure compound of formula I was first disclosed in US Patent No. 7,858,643. Additionally, the racemate of compound of formula I was disclosed in U.S. patent application 2006/0128724, both of these references discloses similar methods for the synthesis of Compound of Formula I.</p> <p>Conventionally, the compounds of formula I are prepared by reacting Bis(pinacolato)diboron with protected 5-bromo-3-[l-(2,6-dichloro-3-fluoro-phenyl)-ethoxy]-pyridin-2-ylamine in the presence of Pd catalyst. The obtained product after deprotection is reacted with N- protected 4-(4-bromo-pyrazol-l-yl)-piperidine in the presence of Pd Catalyst. The obtained product is filtered through celite pad and purified by Column Chromatography. The final product of formula I was obtained by deprotection of the purified compound by using HCl/dioxane. US Patent No. 7,858,643 provides enantiomerically pure aminoheteroaryl compounds, particularly aminopyridines and aminopyrazines, having protein tyrosine kinase activity. More particularly, US 7,858,643 describes process for the preparation of 3-[(lR)-l-(2,6- dichloro-3-fluorophenyl)ethoxy]-5-(l-piperidin-4-ylpyrazol-4-yl)pyridin-2-amine. The Scheme is summarized below in Scheme- 1 :</p>

<p>Scheme-1</p> <p>wherein, “Boc” means tert-butoxycarbonyl; and a) (Boc)<sub>2</sub>, DMF, Dimethylaminopyridine b) Pd(dppf)Cl<sub>2</sub>, KOAc, Dichloromethane; c) HC1, Dioxane, Dichloromethane; d) Pd(PPh<sub>3</sub>)<sub>2</sub>Cl<sub>2</sub>, Na<sub>2</sub>C0<sub>3</sub>, DME/H<sub>2</sub>0; e) 4M HCl/Dioxane, Dichloromethane</p> <p>A similar process has been disclosed in the U.S. patent application 2006/0128724 for the preparation of Crizotinib. J. Jean Cui et. al. in J. Med. Chem. 2011, 54, 6342-6363, also provides a similar process for the preparation of Crizotinib and its derivatives.</p> <p>However, above mentioned synthetic process requires stringent operational conditions such as filtration at several steps through celite pad. Also column chromatography is required at various steps which is not only tedious but also results in significant yield loss. Another disadvantage of above process involves extensive use of palladium catalysts, hence metal scavengers are required to remove palladium content from the desired product at various steps which makes this process inefficient for commercial scale.</p> <p>Yet another disadvantage of above process is the cost of Bis(pinacolato)diboron. This reagent is used in excess in the reaction mixture resulting in considerable cost, especially during large-scale syntheses.</p> <p>US Patent No. 7,825,137 also discloses a process for the preparation of Crizotinib where Boc protected 4-(4-iodo-pyrazol-l-yl)-piperidine is first reacted with Bis(pinacolato)diboron in the presence of Pd catalyst. The reaction mixture is filtered through a bed of celite and the obtained filtrate is concentrated and purified by silica gel chromatography to give to form tert-butyl-4-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazol-l-yl]piperidine-l- carboxylate. To this compound, 5-bromo-3-[l-(2,6-dichloro-3-fluoro-phenyl)-ethoxy]- pyridin-2-ylamine is added in the presence of a Pd catalyst. The reaction mixture is stirred for 16h at 87°C. The reaction mixture is filtered through celite pad and the concentrated filtrate is purified on silica gel column to obtain (4-{6-amino-5-[(R)-l-(2,6-dichloro-3-fluoro- phenyl)-ethoxy]-pyri- din-3-yl}-pyrazol-l-yl)-piperidine-l-carboxylic acid tert-butyl ester of 95% purity. To the solution of resulting compound in dichloromethane 4N HCl/Dioxane is added and thereby getting the reaction suspension is filtered in Buchner funnel lined with filter paper. The obtained solid is dissolved in HPLC water and pH is adjusted to 10 with the addition of Na<sub>2</sub>C0<sub>3</sub> Compound is extracted using dichloroform and is purified on a silica gel column by eluting with CH<sub>2</sub>Cl<sub>2</sub> MeOH/NEt<sub>3</sub> system to obtain Crizotinib. The scheme is summarized below in scheme 2:</p>

<p>Formula (i) Formula (ii)</p>

<p>Formula (iii) Formula (ii) ula (iv)</p>

<p>Formula (v) Formula (I)</p> <p>Scheme-2</p> <p><span style=”color:#ff0000;”>Preparation of Crizotinib:</span></p> <p>To a stirred solution of Tert-butyl 4-(4-{ 6-amino-5-[(li?)-l-(2,6-dichloro-3- fluorophenyl)ethoxy]pyridin-3 -yl } – lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate (material obtained in Example 3) (l.Og, 0.00181 moles) in dichloromethane (-13 ml) at 0°C was added 4.0 M dioxane HQ (6.7 ml, 0.0272 moles). Reaction mixture was stirred at room temperature for 4h. After the completion of reaction monitored by TLC, solid was filtered and washed with dichloromethane (10 ml). The obtained solid was dissolved in water (20 ml); aqueous layer was extracted with dichloromethane (10×2). The pH of aqueous layer was adjusted to 9-10 with Na<sub>2</sub>C03 and compound was extracted with dichloromethane (10 x 3), combined organic layers were washed with water (20 ml), evaporated under vacuum to get solid product. The solid was stirred with ether (10 ml), filtered off, washed well with ether, dried under vacuum to get <span style=”color:#ff0000;”>Crizotinib.</span></p> <p>Yield: 0.45g (55 %)</p> <p>HPLC Purity: 99.35 %</p> <p><span style=”color:#ff0000;”>1HNMR (400 MHz, CDC1<sub>3</sub>) δ: 7.76 (d, J = 1.6 Hz, 1H), 7.56 (s, 1H), 7.49 (s, 1H), 7.30 (dd, J = 9.2 Hz), 7.0 (m, 1H), 6.86 (d, J = 1.6 Hz, 1H), 6.09 ( q, J= 6.8 Hz, 1H), 4.75 (brs, 1H), 4.19 (m, 1H), 3.25 (m, 2H), 2.76 (m, 2H), 2.16 (m, 2H), 1.92 (m, 2H), 1.85 (d, J= 6.8 Hz, 3H), 1.67 (brs, 1H)</span></p> <p>…………………………</p> <p><a href=”http://www.sciencedirect.com/science/article/pii/S0040403914000872″>http://www.sciencedirect.com/science/article/pii/S0040403914000872</a></p&gt;

Abstract

A novel approach for the synthesis of Crizotinib (1) is described. In addition, new efficient procedures have been developed for the preparation of (S)-1-(2,6-dichloro-3-fluorophenyl)ethanol (2) and tert-butyl 4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazol-1-yl)piperidine-1-carboxylate (4), the key intermediates required for the synthesis of Crizotinib.

<hr id=”absgraphicalab0051″ class=”artHeader” />

Graphical abstract

textboxdefaultfig

“>

Full-size image (16 K)
…………………
http://www.sciencedirect.com/science/article/pii/S0040403911021745

Abstract

4-(4-Iodo-1H-pyrazol-1-yl)piperidine is a key intermediate in the synthesis of Crizotinib. We report a robust three-step synthesis that has successfully delivered multi-kilogram quantities of the key intermediate. The process includes nucleophilic aromatic substitution of 4-chloropyridine with pyrazole, followed by hydrogenation of the pyridine moiety and subsequent iodination of the pyrazole which all required optimization to ensure successful scale-up.

<hr id=”absgraphical1″ class=”artHeader” />

Graphical abstract

textboxdefaultfig

“>

Full-size image (6 K)

</div> </div> </dt> </dl> </div> </div> </div> <p>……………………</p>

Org. Process Res. Dev., 2011, 15 (5), pp 1018–1026
DOI: 10.1021/op200131n
Abstract Image

<p class=”articleBody_abstractText”>A robust six-step process for the synthesis of crizotinib, a novel c-Met/ALK inhibitor currently in phase III clinical trials, has been developed and used to deliver over 100 kg of API. The process includes a Mitsunobu reaction, a chemoselective reduction of an arylnitro group, and a Suzuki coupling, all of which required optimization to ensure successful scale-up. Conducting the Mitsunobu reaction in toluene and then crystallizing the product from ethanol efficiently purged the reaction byproduct. A chemoselective arylnitro reduction and subsequent bromination reaction afforded the key intermediate <b>6</b>. A highly selective Suzuki reaction between <b>6</b> and pinacol boronate <b>8</b>, followed by Boc deprotection, completed the synthesis of crizotinib <b>1</b>.</p> </div> <p><span id=”d43162769e1806″ class=”title2″>3-[(1<i>R</i>)-1-(2,6-Dichloro-3-fluorophenyl)ethoxy]-5-[1-(piperidin-4-yl)-1<i>H</i>-pyrazol-4-yl]pyridin-2-amine <b>1</b></span></p> <p><span style=”color:#ff0000;”> <i>crizotinib</i><b>1</b> (20.7 kg, 80%) as a white solid. </span></p> <p><span style=”color:#ff0000;”>Mp 192 °C;</span></p> <p><span style=”color:#ff0000;”><sup>1</sup>H NMR (400 MHz, CDCl<sub>3</sub>) δ: 7.78 (d, <i>J</i> = 1.8 Hz, 1H), 7.58 (s, 1H), 7.52 (s, 1H), 7.31 (dd, <i>J</i> = 9.0, 4.9 Hz, 1H), 7.06 (m, 1H), 6.89 (d, <i>J</i> = 1.7 Hz, 1H), 6.09 (q, 1H), 4.79 (br s, 2H), 4.21 (m, 1H), 3.26 (m, 2H), 2.78 (m, 2H), 2.17 (m, 2H), 1.90 (m, 2H), 1.87 (d, <i>J</i> = 6.7 Hz, 3H), 1.63 (br s, 1H).</span></p> <p><span style=”color:#ff0000;”> <sup>13</sup>C NMR (100.6 MHz, CDCl<sub>3</sub>) δ: 157.5 (d, <i>J</i> = 250.7 Hz), 148.9, 139.8, 137.0, 135.7, 135.6, 129.9, 129.0 (d, <i>J</i> = 3.7 Hz), 122.4, 122.1 (d, <i>J</i> = 19.0 Hz), 119.9, 119.3, 116.7 (d, <i>J</i> = 23.3 Hz), 115.0, 72.4, 59.9, 45.7, 34.0, 18.9.</span></p> <p><span style=”color:#ff0000;”> LC-MS: found <i>m</i>/<i>z</i> 450.0, 451.0, 452.0, 453.0, 454.0, 455.0. </span></p> <p><span style=”color:#ff0000;”>Anal. Calcd for C<sub>21</sub>H<sub>22</sub>Cl<sub>2</sub>FN<sub>5</sub>O: C, 56.01; H, 4.92; N, 15.55. Found: C, 56.08; H, 4.94; N, 15.80.</span></p>

Cui, J. J.; Botrous, I.; Shen, H.; Tran-Dube, M. B.; Nambu, M. D.; Kung, P.-P.; Funk, L. A.; Jia, L.; Meng, J. J.; Pairish, M. A.; McTigue, M.; Grodsky, N.; Ryan, K.; Alton, G.; Yamazaki, S.; Zou, H.; Christensen, J. G.; Mroczkowski, B.Abstracts of Papers; 235th ACS National Meeting, New Orleans, LA, United States, April 6–10, 2008.

</div>

Cui, J. J.; Funk, L. A.; Jia, L.; Kung, P.-P.; Meng, J. J.; Nambu, M. D.; Pairish, M. A.; Shen, H.; Tran-Dube, M. B. U.S. Pat. Appl. U. S. 2006/0046991 A1, 2006.

textboxdefaultfig

“>

textboxdefaultfig

“>

WO2010048131A1 * Oct 20, 2009 Apr 29, 2010 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
WO2011042389A2 * Oct 4, 2010 Apr 14, 2011 Bayer Cropscience Ag Phenylpyri(mi)dinylazoles
US7825137 Nov 23, 2006 Nov 2, 2010 Pfizer Inc. Method of treating abnormal cell growth
US7858643 Aug 26, 2005 Dec 28, 2010 Agouron Pharmaceuticals, Inc. Crizotinib, a c-Met protein kinase inhibitor anticancer agent; 3-[(R)-1-(2,6-dichloro-3-fluoro-phenyl)-ethoxy]-5-(1-piperidin-4-yl-1H-pyrazol-4-yl)-pyridin-2-ylamine is crizotinib
US20060128724 Aug 26, 2005 Jun 15, 2006 Agouron Pharmaceuticals, Inc. Pyrazole-substituted aminoheteroaryl compounds as protein kinase inhibitors
1 J. JEAN CUI J. MED. CHEM. vol. 54, 2011, pages 6342 – 6363
2 ORG. PROCESS RES. DEV. vol. 15, 2011, pages 1018 – 1026
3 * PIETER D. DE KONING ET AL: “Fit-for-Purpose Development of the Enabling Route to Crizotinib (PF-02341066)“, ORGANIC PROCESS RESEARCH & DEVELOPMENT, vol. 15, no. 5, 16 September 2011 (2011-09-16), pages 1018-1026, XP055078841, ISSN: 1083-6160, DOI: 10.1021/op200131n

 

str1

 

VT 1129 BENZENE SULFONATE

CAS 1809323-18-9

Image result for VT1129

VT 1129

1340593-70-5 CAS
MF C22 H14 F7 N5 O2, MW 513.37
2-Pyridineethanol, α-(2,4-difluorophenyl)-β,β-difluoro-α-(1H-tetrazol-1-ylmethyl)-5-[4-(trifluoromethoxy)phenyl]-, (αR)-
R ISOMER
ROTATION +

QUILSECONAZOLE, VT-1129

Viamet, in collaboration with Therapeutics for Rare and Neglected diseases, is investigating quilseconazole benzenesulfonate (VT-1129), a small-molecule lanosterol demethylase (CYP51) inhibitor, developed using the company’s Metallophile technology, for treating fungal infections, including Cryptococcus neoformans meningitis.

WO-2017049080

 

 

////////VT 1129,  VIAMET, WO 2016149486,  Viamet Pharmaceuticals,  Antifungals,  Small molecules,  14-alpha demethylase inhibitors, Orphan Drug Status, Cryptococcosis, On Fast track, PHASE 1, VT-1129, QUILSECONAZOLE

O[C@@](Cn1cnnn1)(c2ccc(F)cc2F)C(F)(F)c3ccc(cn3)c4ccc(OC(F)(F)F)cc4

WO 2016147197, DAPAGLIFLOZIN, NEW PATENT, HARMAN FINOCHEM LIMITED


Image result for HARMAN FINOCHEM LIMITED

Dapagliflozin structure.svg

WO 2016147197, DAPAGLIFLOZIN, NEW PATENT, HARMAN FINOCHEM LIMITED

LINK>>> (WO2016147197) A NOVEL PROCESS FOR PREPARING (2S,3R,4R,5S,6R)-2-[4-CHLORO-3-(4-ETHOXYBENZYL)PHENY 1] -6-(HY DROXY METHYL)TETRAHYDRO-2H-PY RAN-3,4,5-TRIOL AND ITS AMORPHOUS FORM

HARMAN FINOCHEM LIMITED [IN/IN]; 107, Vinay Bhavya Complex 159-A, C.S.T. Road Kalina, Mumbai 400098 Maharashtra (IN)

Image result for HARMAN FINOCHEM LIMITED

KADAM, Vijay Trimbak; (IN).
SAIKRISHNA; (IN).
CHOUDHARE, Tukaram Sarjerao; (IN).
MINHAS, Harpreet Singh; (IN).
MINHAS, Gurpreet Singh; (IN)

Image result for HARMAN FINOCHEM LIMITED

CHAIRMAN

HARPREET SINGH MINHAS

HARPREET SINGH MINHAS

Owner, HARMAN FINOCHEM LIMITED

Image result for HARMAN FINOCHEM LIMITED

(2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol is sodium dependent glucose transporter (SGLT) which is currently under investigation for the treatment of type-2 diabetes. (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol is marketed under the tradename Farxiga or Forxiga.

(2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol is also known as D-glucitol, l,5-anhydro-l-C-[4-chloro-3-[(4ethoxyphenyl)methyl]phenyl]-, (I S). (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3, 4,5 -triol is a white to off-white powder with a molecular formula of C2iH25C106 and a molecular weight of 408.87

Formula-I

US 6,515,117 B2 discloses (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol and its pharmaceutically acceptable salts. US 6,515,117 B2 also describes process for preparation of (2S,3R,4R,5S,6R)-2-[4- chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol which comprises reaction of 5-bromo-2-chloro-4′-ethoxydiphenylmethane with 2,3,4,6-tetra-O-trimethylsilyl- -D-glucolactone in presence of THF/Toluene, methansulfonic acid to yield o-methylglucoside product which further reacts with Et3SiH, BF3Et20 in presence of MDC and acetonitrile to yield yellow solidified foam which is dissolved in MDC, pyridine and followed by acetylation with acetic anhydride, DMAP to yield tetra acetylated- β-C-glucoside as a white solid which is further deprotected with LiOH H20 in presence of THF/MeOH/H20 to get (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol.

The drawback of said prior art is having multiple process steps which makes the process very lengthy and tedious. Moreover the process discloses use of hazardous chemicals like pyridine which is not applicable to industry.

Process for preparation of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenylJ-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol is disclosed in US 7,375,213 B2 and J.Med.Chem.2008, 51, 1145-1149. The preparation process is depicted in Scheme-I.

Scheme-1

Prior art US’213 describes reaction of 2-chloro-5-bromo-4′-ethoxy-diphenylmethane with 2,3,4,6-tetra-O-trimethylsilyl-D-gluconolactone, n-BuLi in presence of THF and Heptane. After basification with TEA, the oily residue of methyl- l-C-(2-chloro-4′- ethoxy-diphenylmethan-3-yl)-a-D-glucopyranose obtained as solid compound after workup. This compound reacts with acetic anhydride in presence of THF, DIPEA and DMAP to get oily residue of methyl-2,3,4,6 tetra-0-acetyl-l-C-(2-chloro-4′-ethoxydiphenylmethan-3-yl)-a-D-glucopyranose which further undergoes reduction reaction in presence of acetonitirle, t riethylsilane, boron trifluoride etherate to yield 2,3,4,6-tetra-0-acetyl-l-C-(2-chloro-4′-ethoxydi henylmethan-3-yl)-β-D-glucopyranose which is further deprotected by reacting with LiOH monohydrate in presence of THF/MeOH/H20 to get (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol.

The said prior art describes multiple, time consuming process steps which involves getting the intermediate products as oily residue at various stages of the process, which is difficult to purify and handle for further process step. More over the workup involves multiple evaporation of product which may result in decomposition. Another drawback of the process is that the process describes n-BuLi reaction with two pot reaction. It is very difficult to transfer the material from one reactor to second reactor at -78 °C at industrial level with highly moisture sensitive reaction mass. This makes process uneconomical, cumbersome and commercially not viable. Further when practically the said method followed, a-Isomer of the final product is formed in the range of 6-8% along ith Des-bromo impurity formed in the range of 7-9 %, which increases after addition of n-butyllithium and kept the mass for overnight reaction. Moreover lactone ring cleavage is also observed in the range of 3-4% after addition of Methanesulphonic Acid/Methanol and maintained overnight for reaction completion, the removal of which is difficult from the final product.

WO 2008002824 A 1 discloses crystalline forms of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol comprising (S)-propylene glycol (PG), (R)-PG, EtOH, ethylene glycol (EG), 1 :2 L-proline, 1 : 1 L-proline, 1 : 1 L-proline hemihydrate, 1 : 1 L-phenylalanine and its preparation process.

In the light of the above drawbacks, it is necessitated to provide economical, robust, safe and commercially viable process for preparing (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol.

Accordingly, it is an objective of the present invention to provide a commercially viable process for the preparation of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxyb.enzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol, prepared via riovel intermediates which gives higher yield and purity and facilitates easy recovery of the final compound. The purification process does not involve any costly technique/equipment, however, carried out with solvents which are industrially feasible. More over the present invention discloses the n-BuLi insitu reaction that makes the present invention cost-effective over the teachings of prior art.

Image result for HARMAN FINOCHEM LIMITED

Image result for HARMAN FINOCHEM LIMITED

Scheme-II

Formula-Ill Formula-IV

Formula-V where R1= allyl, prop-2-ynyl,isopropyl

Scheme-Ill

where R = allyl, prop-2-ynyl

Scheme-IV

Scheme-V

Examples:

Example-1: Preparation of 3,4,5-Tris-trimethylsiIanyloxy-6-trimethylsiIanyloxymethyl-tetrahydro-pyran-2-one

To 750 cc of dry THF added 1.12 mole 3,4,5-Trihydroxy-6-hydroxymethyl-tetrahydro-pyran-2-one at ambient temperature and stirred for 20 min. To the reaction mass added 9.0 mole N-Methyl morpholine and stirred for another 30.0 min at ambient temperature. Reaction mass was cooled to -5 °C to 0 °C and stirred for 30.0 min. Added 18.0 mole Trimethyl sillyl chloride at the temp -5 °C to 0 °C and stirred for 30.0 min. Temperature was raised to 25 °C to 30 °C and maintained for 18-20hrs. After reaction complies by GC, the reaction mass was cooled to -5 deg to 0 deg. Added Sat.Sodium bicarbonate solution to obtain the pH 7-8 and stirred for 1 hr at 0 °C. Added 500 cc toluene and stirred for lhr. Reaction mass was settled down for 30.0 min and layers were separated. To the Aqueous layer added 250 cc of toluene and stirred for 30.0 min. Layers separated and both the organic layers mixed and back washed with sat.brine solution. Organic layer was distilled under reduced pressure at a temperature of about 40 – 48 deg. Unload the oily mass . Purity: 92-96 %

Example-2: Preparation of 2-Allyloxy-2-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyI-tetrahydro-pyran-3,4,5-triol

To the mixture of 10 cc THF and 10 cc Toluene added 0.138 mole 4-(5-bromo-2-chlorobenzyl)phenyl ethyl ether at ambient temperature and stirred for 15 min. Cooled to -70 to -80°C in dry ice /acetone bath and stirred for 15 min. Added a solution of 0.014 mole n-Butyl lithium (1.9M in hexanes) at -70 to -80°C. and stirred for lhr. Added solution of 3, 4, 5-Tris-trimethylsilanyloxy-6-trimethylsilanyloxymethyl-tetrahydro-pyran-2-one in 5 cc of Toluene at -70 to -80°C and stirred for 2 to 3hrs. After the compliance of the reaction, reaction mass was quenched with Methane sulphonic acid and Allyl alcohol mixture at -70 to -80°C. Temperature was raised to ambient temperature and stirred overnight. Reaction mass was quenched with 30 cc sat.sodiumbicarbonate solution to bring the pH neutral to alkaline and stirred for 30.0 min. Layers separated and aqueous layer was extracted with 10 cc of Toluene. Organic layer was combined and washed with 30cc water and 50 cc sat. brine solution. Organic layer was distilled under reduced pressure to recover toluene. Solid compound was dissolved in 50cc of toluene and quenched in n-Hexane to obtain 83 % the compound as crystalline solid.

HPLC purity: 88 – 91 %

I R data:

Anomeric C-0 stretching: 1242 cm“1

Allylic C- O stretching: 1 177 cm“1

Allylic C- H stretching: 3010 – 3120 cm“1

Aromatic C- CI stretching: 820 cm“1

Lactones O – H stretching: 3240 – 3380 cm“1

Lactones C – 0 stretching: 1045 – 1092 cm“1

Aromatic C=C stretching: 1510 , 1548 , 1603 , 1703 cm“1

Alkane C – H stretching: 2877,2866, 2956, 2958, 2962 cm“1

Aromatic C – H stretching: 3050 – 3090 cm“1

Dip-Mass

(M+Na) 487.19 m/z

(M+K) 503.17 m/z

Example 3: Preparation of 2-prop-2ynyl-2-[4-Chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyl-tetrahydro-pyran-3,4,5-triol

To the mixture of 10 cc THF and 10 cc Toluene added 0.138 mole 4-(5-bromo-2-chlorobenzyl)phenyl ethyl ether at ambient temperature and stirred for 15 min. Cooled to -70 to -80°C in dry ice /acetone bath and stirred for 15 min. Added a solution of 0.014 mole n-Butyl lithium (1.9M in hexanes) at -70 to -80°C. and stirred for lhr. Added solution of 3, 4, 5-Tris-trimethylsilanyloxy-6-trimethylsilanyloxymethyl-tetrahydro-pyran-2-one in 5 cc of Toluene at -70 to -80°C and stirred for 2 to 3hrs. After the compliance of the reaction, the reaction mass was quenched with Methane sulphonic acid and propargyl alcohol mixture at -70 to -80°C. Temperature was raised to ambient temperature and stirred overnight. Reaction mass was quenched with 30 cc sat.sodiumbicarbonate solution to bring the pH neutral to alkaline. Reaction mass stirred for 30.0 min. Layers separated and aqueous layer was extracted with 10 cc of Toluene. Organic layer were combined and washed with 30cc water and 50 cc sat. brine solution. Organic layer was distilled under reduced pressure to recover toluene. Solid compound dissolved in 50cc of toluene and quenched in n-Hexane to obtain 75 – 80 % the compound as crystalline solid.

HPLC purity: 88 – 93 %

IR data:

Anomeric C-0 stretching: 1242 cm“1

Propargyl ~c CH stretching: 2125 cm“1

Propargyl C- H stretching : 3010 – 3120 cm“1

Aromatic C- CI stretching: 820 cm“1

Lactones O – H stretching: 3240 – 3380 cm“1

Lactones C – 0 stretching: 1045 – 1092 cm“1

Aromatic C=C stretching: 1510 , 1548 , 1603 , 1703 cm“1

Alkane C – H stretching: 2877, 2866,2956,2958,2962 cm“1

Aromatic C – H stretching: 3050 – 3090 cm“1

Dip-Mass

(M+Na) 485.25 m/z

(M+K) 501.25 m/z

Example-4: Preparation of 2-[4-Chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyI-tetrahydro-pyran-3,4,5-trioI

To the mixture of 20 cc (1 : 1 MDC + ACN) added 0.1 1 mole 2-Allyloxy-2-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyl-tetrahydro-pyran-3,4,5-triol under argon atmosphere, and stirred the reaction mass for 30.0 min. Cooled the reaction mass to -40 to -55°C in a dry ice/acetone bath under argon atmosphere. Charged 3 mole Triethylsilane at -40 to -55°C and stirred the reaction mass for 30.0 min at -50 to -55°C. Slowly added Borontrifloride in diethyl ether solution at -40 to -55°C and stirred the reaction mass for 2 hrs. Quenched the reaction mass with 50 cc sat. sodium bicarbonate solution at -40 to -55°C . and stirred the reaction mass for 30.0 min. Slowly raised the temperature to 25 to 30°C. Settled down the reaction mass and separated the layers, extracted the aqueous layer with 100 cc of MDC. Combined the organic layer and wash with 500 cc water. Washed the organic layer with 500 cc of sat. Brine solution. Distilled out the MDC under reduced pressure below 40°C. to get 85 %the light yellow solid.

HPLC purity: 92-95 %

Example 5: Preparation of 2-[4-Chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyl-tetrahydro-pyran-3,4,5-triol

To the mixture of 20 cc (1 :1 MDC + ACN) added 0.11 mole 2-prop-2-ynyl-2-[4-Chloro-3-(4-ethoxy-benzyl)-phenyl]-6-hydroxymethyl-tetrahydro-pyran-3,4,5-triol under argon

atmosphere. Stirred the reaction mass for 30.0 min. Cooled the reaction mass to -40 to -55°C in a dry ice/acetone bath under argon atmosphere. Charged 3 mole Triethylsilane at -40 to -55°C and stirred the reaction mass for 30.0 min at -50 to -55°C. Slowly added Borontrifloride in diethyl ether solution at -40 to -55°C and stirred the reaction mass for 2 hrs. Quenched the reaction mass with 50 cc sat. sodium bicarbonate solution at -40 to -55°C and Stirred the reaction mass for 30.0 min. Slowly raised the temperature to 25 to 30°C. Settled down the reaction mass and separated the layers, extracted the aqueous layer with 100 cc of MDC. Combined the organic layer and washed with 500 cc water. Washed the organic layer with 500 cc of sat. Brine solution. Distilled out the MDC under reduced pressure below 40°C. to get 85%the light yellow solid.

HPLC purity: 90%

Example 6: Preparation of amorphous form of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol

To the solid obtained from example 4 charged 500cc of n-heptane and stirred for ½hrs at ambient temperature. Heated the reaction mass to 55-60°C and stirred it for 2-3 hrs.; cooled to room temperature and maintained for 4-5 hrs. Filtered the solid and washed the, cake with 100 cc n-heptane. Dried at 40-45°C under vacuum to get 85% amorphous form of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol.

HPLC purity: 91-93%

Example 7: Preparation of amorphous form of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol

To the solid obtained from example 5 charged 500cc of n-heptane and stirred for ½ hrs at ambient temperature. Heated the reaction mass to 55-60°C and stirred it for 2-3 hrs., cooled to room temperature and maintained for 4-5 hrs. Filtered the solid and washed the cake with 100 cc n-heptane. Dried at 40-45 °C under vacuum to get 85-88% amorphous form of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6- (hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol.

HPLC purity: 89-91%

Example 8: Preparation of L-proline – (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyI]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol co crystal

To the 10 cc of Ethyl acetate charged 1.0 mole (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol under argon atmosphere at ambient temperature and stirred for 30.0 min to get clear solution. Slowly heated the reaction mass to 60 – 65°C and stirred for 1 hr. Slowly added L-proline at 60 -65°C and maintained for 1 hr. Slowly added 15 cc n-Heptane to the reaction mass at 60 -65°C and stirred the mass for 2.5 hrs. Cooled the mass to ambient temperature for 3-4 hrs and maintained for 5 hrs. Filtered the mass under argon atmosphere. Washed the cake with 10 cc n-Heptane. Dried the cake at 50-55°C under reduced pressure to get 92% titled compound.

HPLC purity: 99%

Example 9: Preparation of L-proline – (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triolco crystal

To the 10 cc of acetone charged 1.0 mole (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol under argon atmosphere at ambient temperature and stirred for 30.0 min to get clear solution. Slowly heated the reaction mass to 60 – 65°C and stirred for 1 hr. Slowly added proline at 60 -65°C and maintained for 1 hr. Slowly added 15 cc n-Heptane to the reaction mass at 60 -65°C and stirred the mass for 2.5 hrs. Cooled the mass to ambient temperature for 3-4 hrs and maintained for 5 hrs. Filtered the mass under argon atmosphere. Washed the cake with 10 cc n-Heptane. Dried the cake at 50-55°C under reduced pressure to get 93-95% titled compound.

HPLC purity: 98-99%

Example 10: Preparation of amorphous form of (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol

To the 15 cc ethyl acetate added (2S,3R,4R,5S,6R)-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol at ambient temperature and stirred for 30.0 min. Slowly added 5- 8 cc sat. sodium bicarbonate solution at ambient temperature and stirred for 1.5 hr to get the clear solution. Settled down and separated layers. Extracted the aqueous layer with 25 cc ethyl acetate.

Combined the organic layers and washed the ethyl acetate layer with 50 cc sat. Sodium chloride solution. Distilled out ethyl acetate under reduced pressure at 40 – 45°C to get fluffy solid. Charged 50 cc n-Heptane and stirred for 5 hrs to get 70-78% the title compound as Amorphous soild.

HPLC purity: 99.8-99.95 %

Example 11: Preparation of 2-chloro -4′- ethoxydiphenylmethane (impurity)

To the 20 cc THF and 20 cc Toluene added 0.25 mole 2-ehloro-5-bromo-4′- ethoxydiphenylmethane under argon atmosphere. Cooled the reaction mass to – 78° C. Slowly added n-Butyl lithium (1.9 M in hexane) at – 78° C and stirred for 30 min. Slowly added 20 % Ammonium chloride solution to the reaction mass. Brought the reaction mass to ambient temperature and stirred for 30 min. Settled and separated layers. Extracted the aqueous layer with 50 cc toluene. Washed the combined organic layer with 500 cc brine solution. Distilled out the toluene and charged heptanes, stirred for 2 – 3 hrs at ambient temperature. Filtered the product and dried the product at 45 – 50°C under reduced pressure to get 93 % titled compound.

Mass: (m+1) 247 m/z found 247.1 1

HPLC purity: 96.33 %

SHENDRA AURANGABAD, MAHARASHTRA, INDIA

Image result for HARMAN FINOCHEM LIMITED

Bhupinder Singh Manhas

 

Google's 18th Birthday

////////WO 2016147197, DAPAGLIFLOZIN, NEW PATENT, HARMAN FINOCHEM LIMITED

Follow New Drug Approvals on WordPress.com

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

Join 37.9K other subscribers

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

DISCLAIMER

I , Dr A.M.Crasto is writing this blog to share the knowledge/views, after reading Scientific Journals/Articles/News Articles/Wikipedia. My views/comments are based on the results /conclusions by the authors(researchers). I do mention either the link or reference of the article(s) in my blog and hope those interested can read for details. I am briefly summarising the remarks or conclusions of the authors (researchers). If one believe that their intellectual property right /copyright is infringed by any content on this blog, please contact or leave message at below email address amcrasto@gmail.com. It will be removed ASAP