New Drug Approvals

Home » Posts tagged 'fda 2021' (Page 4)

Tag Archives: fda 2021

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

Blog Stats

  • 4,804,099 hits

Flag and hits

Flag Counter

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

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

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

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

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

DR ANTHONY MELVIN CRASTO Ph.D

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

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Odevixibat


img

Odevixibat.png

Odevixibat

A-4250, AR-H 064974

CAS 501692-44-0

BUTANOIC ACID, 2-(((2R)-2-((2-((3,3-DIBUTYL-2,3,4,5-TETRAHYDRO-7-(METHYLTHIO)-1,1-DIOXIDO-5-PHENYL-1,2,5-BENZOTHIADIAZEPIN-8-YL)OXY)ACETYL)AMINO)-2-(4-HYDROXYPHENYL)ACETYL)AMINO)-, (2S)-

(2S)-2-[[(2R)-2-[[2-[(3,3-dibutyl-7-methylsulfanyl-1,1-dioxo-5-phenyl-2,4-dihydro-1λ6,2,5-benzothiadiazepin-8-yl)oxy]acetyl]amino]-2-(4-hydroxyphenyl)acetyl]amino]butanoic acid

Molecular Formula C37H48N4O8S2
Molecular Weight 740.929
        • UPDATE 7/20/2021FDA APPROVED, To treat pruritus,

      Bylvay

    • New Drug Application (NDA): 215498
      Company: ALBIREO PHARMA INC
  • Orphan Drug Status Yes – Primary biliary cirrhosis; Biliary atresia; Intrahepatic cholestasis; Alagille syndrome
  • New Molecular Entity Yes
  • Phase III Biliary atresia; Intrahepatic cholestasis
  • Phase II Alagille syndrome; Cholestasis; Primary biliary cirrhosis
  • No development reported Non-alcoholic steatohepatitis
  • 22 Jul 2020 Albireo initiates an expanded-access programme for Intrahepatic cholestasis in USA, Canada, Australia and Europe
  • 14 Jul 2020 Phase-III clinical trials in Biliary atresia (In infants, In neonates) in Belgium (PO) after July 2020 (EudraCT2019-003807-37)
  • 14 Jul 2020 Phase-III clinical trials in Biliary atresia (In infants, In neonates) in Germany, France, United Kingdom, Hungary (PO) (EudraCT2019-003807-37)

UPDATE Bylvay, FDA APPROVED2021/7/20 AND EMA 2021/7/16

Odevixibat, sold under the trade name Bylvay, is a medication for the treatment of progressive familial intrahepatic cholestasis (PFIC).[1]

The most common side effects include diarrhea, abdominal pain, hemorrhagic diarrhea, soft feces, and hepatomegaly (enlarged liver).[1]

Odevixibat is a reversible, potent, selective inhibitor of the ileal bile acid transporter (IBAT).[1][2]

In May 2021, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) recommended granting a marketing authorization in the European Union for odevixibat for the treatment of PFIC in people aged six months or older.[1][3]

A-4250 (odevixibat) is a selective inhibitor of the ileal bile acid transporter (IBAT) that acts locally in the gut. Ileum absorbs glyco-and taurine-conjugated forms of the bile salts. IBAT is the first step in absorption at the brush-border membrane. A-4250 works by decreasing the re-absorption of bile acids from the small intestine to the liver, whichreduces the toxic levels of bile acids during the progression of the disease. It exhibits therapeutic intervention by checking the transport of bile acids. Studies show that A-4250 has the potential to decrease the damage in the liver cells and the development of fibrosis/cirrhosis of the liver known to occur in progressive familial intrahepatic cholestasis. A-4250 is a designated orphan drug in the USA for October 2012. A-4250 is a designated orphan drug in the EU for October 2016. A-4250 was awarded PRIME status for PFIC by EMA in October 2016. A-4250 is in phase II clinical trials by Albireo for the treatment of primary biliary cirrhosis (PBC) and cholestatic pruritus. In an open label Phase 2 study in children with cholestatic liver disease and pruritus, odevixibat showed reductions in serum bile acids and pruritus in most patients and exhibited a favorable overall tolerability profile.

str1

albireo_logo_nav.svg

Odevixibat is a highly potent, non-systemic ileal bile acid transport inhibitor (IBATi) that has has minimal systemic exposure and acts locally in the small intestine. Albireo is developing odevixibat to treat rare pediatric cholestatic liver diseases, including progressive familial intrahepatic cholestasisbiliary atresia and Alagille syndrome.

With normal function, approximately 95 percent of bile acids released from the liver into the bile ducts to aid in liver function are recirculated to the liver via the IBAT in a process called enterohepatic circulation. In people with cholestatic liver diseases, the bile flow is interrupted, resulting in elevated levels of toxic bile acids accumulating in the liver and serum. Accordingly, a product capable of inhibiting the IBAT could lead to a reduction in bile acids returning to the liver and may represent a promising approach for treating cholestatic liver diseases.

The randomized, double-blind, placebo-controlled, global multicenter PEDFIC 1 Phase 3 clinical trial of odevixibat in 62 patients, ages 6 months to 15.9 years, with PFIC type 1 or type 2 met its two primary endpoints demonstrating that odevixibat reduced serum bile acids (sBAs) (p=0.003) and improved pruritus (p=0.004), and was well tolerated with a low single digit diarrhea rate. These topline data substantiate the potential for odevixibat to be first drug for PFIC patients. The Company intends to complete regulatory filings in the EU and U.S. no later than early 2021, in anticipation of regulatory approval, issuance of a rare pediatric disease priority review voucher and launch in the second half of 2021.

Odevixibat is being evaluated in the ongoing PEDFIC 2 open-label trial (NCT03659916) designed to assess long-term safety and durability of response in a cohort of patients rolled over from PEDFIC 1 and a second cohort of PFIC patients who are not eligible for PEDFIC 1.

Odevixibat is also currently being evaluated in a second Phase 3 clinical trial, BOLD (NCT04336722), in patients with biliary atresia. BOLD, the largest prospective intervention trial ever conducted in biliary atresia, is a double-blind, randomized, placebo-controlled trial which will enroll approximately 200 patients at up to 75 sites globally to evaluate the efficacy and safety of odevixibat in children with biliary atresia who have undergone a Kasai procedure before age three months. The company also anticipates initiating a pivotal trial of odevixibat for Alagille syndrome by the end of 2020.

For more information about the PEDFIC 2 or BOLD studies, please visit ClinicalTrials.gov or contact medinfo@albireopharma.com.

The odevixibat PFIC program, or elements of it, have received fast track, rare pediatric disease and orphan drug designations in the United States. In addition, the FDA has granted orphan drug designation to odevixibat for the treatment of Alagille syndrome, biliary atresia and primary biliary cholangitis. The EMA has granted odevixibat orphan designation, as well as access to the PRIority MEdicines (PRIME) scheme for the treatment of PFIC. Its Paediatric Committee has agreed to Albireo’s odevixibat Pediatric Investigation Plan for PFIC. EMA has also granted orphan designation to odevixibat for the treatment of biliary atresia, Alagille syndrome and primary biliary cholangitis.

PATENT

https://patents.google.com/patent/US9694018B1/en

Example 5

1,1-Dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(N—{(R)-α-[N—((S)-1-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine, Mw. 740.94.

This compound is prepared as described in Example 29 of WO3022286.

PATENT

https://patents.google.com/patent/WO2003022286A1/sv

Example 29

1,1-Dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(N-((R)-α-[N-((S)- 1-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine

A solution of 1,1-dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-[N-((R)-α-carboxy-4-hydroxybenzyl)carbamoylmethoxy]-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine (Example 18; 0.075 g, 0.114 mmol), butanoic acid, 2-amino-, 1,1-dimethylethyl ester, hydrochloride, (2S)-(0.031 g, 0.160 mmol) and Ν-methylmorpholine (0.050 ml, 0.457 mmol) in DMF (4 ml) was stirred at RT for 10 min, after which TBTU (0.048 g, 0.149 mmol) was added. After 1h, the conversion to the ester was complete. M/z: 797.4. The solution was diluted with toluene and then concentrated. The residue was dissolved in a mixture of DCM (5 ml) and TFA (2 ml) and the mixture was stirred for 7h. The solvent was removed under reduced pressure. The residue was purified by preparative HPLC using a gradient of 20-60% MeCΝ in 0.1M ammonium acetate buffer as eluent. The title compound was obtained in 0.056 g (66 %) as a white solid. ΝMR (400 MHz, DMSO-d6): 0.70 (3H, t), 0.70-0.80 (6H, m), 0.85-1.75 (14H, m), 2.10 (3H, s), 3.80 (2H, brs), 4.00-4.15 (1H, m), 4.65 (1H, d(AB)), 4.70 (1H, d(AB)), 5.50 (1H, d), 6.60 (1H, s), 6.65-7.40 (11H, m), 8.35 (1H, d), 8.50 (1H, d) 9.40 (1H, brs).

PATENT

https://patents.google.com/patent/US20140323412A1/en

PATENT

https://patents.google.com/patent/WO2013063526A1/e

PATENT

https://patents.google.com/patent/WO2019245448A1/en

The compound l,l-dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(A/-{(R)-a-[A/-((S)-l-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-l,2,5-benzothiadiazepine (odevixibat; also known as A4250) is disclosed in WO 03/022286. The structure of odevixibat is shown below.

Figure imgf000002_0001

As an inhibitor of the ileal bile acid transporter (IBAT) mechanism, odevixibat inhibits the natural reabsorption of bile acids from the ileum into the hepatic portal circulation. Bile acids that are not reabsorbed from the ileum are instead excreted into the faeces. The overall removal of bile acids from the enterohepatic circulation leads to a decrease in the level of bile acids in serum and the liver. Odevixibat, or a pharmaceutically acceptable salt thereof, is therefore useful in the treatment or prevention of diseases such as dyslipidemia, constipation, diabetes and liver diseases, and especially liver diseases that are associated with elevated bile acid levels.

According to the experimental section of WO 03/022286, the last step in the preparation of odevixibat involves the hydrolysis of a tert-butyl ester under acidic conditions. The crude compound was obtained by evaporation of the solvent under reduced pressure followed by purification of the residue by preparative HPLC (Example 29). No crystalline material was identified.

Amorphous materials may contain high levels of residual solvents, which is highly undesirable for materials that should be used as pharmaceuticals. Also, because of their lower chemical and physical stability, as compared with crystalline material, amorphous materials may display faster

decomposition and may spontaneously form crystals with a variable degree of crystallinity. This may result in unreproducible solubility rates and difficulties in storing and handling the material. In pharmaceutical preparations, the active pharmaceutical ingredient (API) is for that reason preferably used in a highly crystalline state. Thus, there is a need for crystal modifications of odevixibat having improved properties with respect to stability, bulk handling and solubility. In particular, it is an object of the present invention to provide a stable crystal modification of odevixibat that does not contain high levels of residual solvents, that has improved chemical stability and can be obtained in high levels of crystallinity.

Example 1

Preparation of crystal modification 1

Absolute alcohol (100.42 kg) and crude odevixibat (18.16 kg) were charged to a 250-L GLR with stirring under nitrogen atmosphere. Purified water (12.71 kg) was added and the reaction mass was stirred under nitrogen atmosphere at 25 ± 5 °C for 15 minutes. Stirring was continued at 25 ± 5 °C for 3 to 60 minutes, until a clear solution had formed. The solution was filtered through a 5.0 m SS cartridge filter, followed by a 0.2 m PP cartridge filter and then transferred to a clean reactor.

Purified water (63.56 kg) was added slowly over a period of 2 to 3 hours at 25 ± 5 °C, and the solution was seeded with crystal modification 1 of odevixibat. The solution was stirred at 25 ± 5 °C for 12 hours. During this time, the solution turned turbid. The precipitated solids were filtered through centrifuge and the material was spin dried for 30 minutes. The material was thereafter vacuum dried in a Nutsche filter for 12 hours. The material was then dried in a vacuum tray drier at 25 ± 5 °C under vacuum (550 mm Hg) for 10 hours and then at 30 ± 5 °C under vacuum (550 mm Hg) for 16 hours. The material was isolated as an off-white crystalline solid. The isolated crystalline material was milled and stored in LDPE bags.

An overhydrated sample was analyzed with XRPD and the diffractogram is shown in Figure 2.

Another sample was dried at 50 °C in vacuum and thereafter analysed with XRPD. The diffractogram of the dried sample is shown in Figure 1.

The diffractograms for the drying of the sample are shown in Figures 3 and 4 for 2Q ranges 5 – 13 ° and 18 – 25 °, respectively (overhydrated sample at the bottom and dry sample at the top).

References

  1. Jump up to:a b c d “First treatment for rare liver disease”European Medicines Agency (EMA) (Press release). 21 May 2021. Retrieved 21 May 2021. Text was copied from this source which is © European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
  2. ^ “Odevixibat”Albireo Pharma. Retrieved 21 May 2021.
  3. ^ “Bylvay: Pending EC decision”European Medicines Agency (EMA). 19 May 2021. Retrieved 21 May 2021.

External links

  • “Odevixibat”Drug Information Portal. U.S. National Library of Medicine.

ClinicalTrials.gov

CTID Title Phase Status Date
NCT04336722 Efficacy and Safety of Odevixibat in Children With Biliary Atresia Who Have Undergone a Kasai HPE (BOLD) Phase 3 Recruiting 2020-09-02
NCT04483531 Odevixibat for the Treatment of Progressive Familial Intrahepatic Cholestasis Available 2020-08-25
NCT03566238 This Study Will Investigate the Efficacy and Safety of A4250 in Children With PFIC 1 or 2 Phase 3 Active, not recruiting 2020-03-05
NCT03659916 Long Term Safety & Efficacy Study Evaluating The Effect of A4250 in Children With PFIC Phase 3 Recruiting 2020-01-21
NCT03608319 Study of A4250 in Healthy Volunteers Under Fasting, Fed and Sprinkled Conditions Phase 1 Completed 2018-09-19
CTID Title Phase Status Date
NCT02630875 A4250, an IBAT Inhibitor in Pediatric Cholestasis Phase 2 Completed 2018-03-29
NCT02360852 IBAT Inhibitor A4250 for Cholestatic Pruritus Phase 2 Terminated 2017-02-23
NCT02963077 A Safety and Pharmakokinetic Study of A4250 Alone or in Combination With A3384 Phase 1 Completed 2016-11-16

EU Clinical Trials Register

EudraCT Title Phase Status Date
2019-003807-37 A Double-Blind, Randomized, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Odevixibat (A4250) in Children with Biliary Atresia Who Have Undergone a Kasai Hepatoportoenterostomy (BOLD) Phase 3 Ongoing 2020-07-29
2015-001157-32 An Exploratory Phase II Study to demonstrate the Safety and Efficacy of A4250 Phase 2 Completed 2015-05-13
2014-004070-42 An Exploratory, Phase IIa Cross-Over Study to Demonstrate the Efficacy Phase 2 Ongoing 2014-12-09
2017-002325-38 An Open-label Extension Study to Evaluate Long-term Efficacy and Safety of A4250 in Children with Progressive Familial Intrahepatic Cholestasis Types 1 and 2 (PEDFIC 2) Phase 3 Ongoing
2017-002338-21 A Double-Blind, Randomized, Placebo-Controlled, Phase 3 Study to Demonstrate Efficacy and Safety of A4250 in Children with Progressive Familial Intrahepatic Cholestasis Types 1 and 2 (PEDFIC 1) Phase 3 Ongoing, Completed

.

Odevixibat
Odevixibat structure.png
Clinical data
Trade names Bylvay
Routes of
administration
By mouth
ATC code
  • None
Identifiers
CAS Number
  • 501692-44-0
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C37H48N4O8S2
Molar mass 740.93 g·mol−1
3D model (JSmol)

////////////odevixibat, Orphan Drug Status, phase 3, Albireo, A-4250, A 4250, AR-H 064974

CCCCC1(CN(C2=CC(=C(C=C2S(=O)(=O)N1)OCC(=O)NC(C3=CC=C(C=C3)O)C(=O)NC(CC)C(=O)O)SC)C4=CC=CC=C4)CCCC

publicationnumber
US-2020046635-A1
US-2020046636-A1 US-2020046757-A1 US-2020046758-A1 US-2020002299-A1 WO-2019245448-A1 WO-2019245449-A1 US-2019046451-A1 US-2019070217-A1
US-10441605-B2
US-2017224720-A1
US-2017224721-A1
US-2018264029-A1
US-2018360869-A1
US-2018360870-A1
US-2018360871-A1
WO-2017133517-A1
US-2017143738-A1
US-2017143783-A1
EP-2968230-A2
EP-2968262-A1
US-2014271734-A1
US-2014275090-A1
WO-2014144485-A1
WO-2014144485-A9
WO-2014144650-A2
EP-2770990-A1
EP-2771003-A1
EP-2771003-B1
EP-3266457-A1
EP-3278796-A1
US-10512657-B2
US-2013108573-A1
US-2013109671-A1
US-2013338093-A1
US-2014243281-A1
US-2014323412-A1
US-2016310518-A1
US-2017368085-A1
US-2019169217-A1
US-2020069715-A1
WO-2013063512-A1
WO-2013063526-A1
EP-2739286-A2
WO-2013020108-A2
EP-2637646-B1
EP-2637668-B1
EP-3023102-A1
EP-3023102-B1
EP-3400944-A1
US-10000528-B2
US-10011633-B2
US-10093697-B2
US-10221212-B2
US-2012114588-A1
US-2013225511-A1
US-2013236541-A1
US-2015031636-A1
US-2015031637-A1
US-2016193277-A1
US-2016194353-A1
US-2017182059-A1
US-2017182115-A1
US-2018022776-A1
US-2018030088-A1
US-2018030089-A1
US-2018162904-A1
US-2018362577-A1
US-9688720-B2
US-9694018-B1
US-10555950-B2
US-2016220577-A1
US-9339480-B2
WO-2008039829-A2
US-2009069285-A1
US-7842684-B2
WO-2007051995-A2
EP-1896408-A1
EP-1896409-A1
EP-1896457-A1
US-2010048529-A1
US-2010048530-A1
US-2010137273-A1
US-2010152156-A1
US-2010168039-A1
US-2010168075-A1
US-7893048-B2
US-7906502-B2
WO-2006137792-A1
WO-2006137794-A1
WO-2006137795-A1
US-2010216759-A1
US-7863265-B2
US-2008194494-A1
US-2009186834-A1
WO-2006102674-A2
US-2009005321-A1
EP-1831151-A1
US-2008114064-A1
WO-2006065214-A1
EP-1699759-A1
US-2007142304-A1
US-2008064676-A1
US-2010099657-A2
US-7871998-B2
WO-2005061452-A1
EP-1638922-A1
EP-1638926-A1
EP-1638930-A1
EP-1675820-A2
EP-1676833-A1
US-2005148656-A1
US-2006142389-A1
US-2006178432-A1
US-2006194879-A1
US-2006258866-A1
US-2007099928-A1
US-2007099997-A1
US-2007244198-A1
US-7309720-B2
WO-2004110984-A1
WO-2004113270-A2
WO-2004113276-A1
WO-2004113283-A1
EP-1610770-A1
EP-1610770-B1
EP-1894564-A2
US-2006199797-A1
US-7514421-B2
WO-2004089350-A1
EP-1572626-A1
US-2005131068-A1
WO-2004056748-A1
EP-1539120-A1
US-2006083790-A1
WO-2004006899-A1
EP-1521742-A1
US-2005239766-A1
US-7470678-B2
WO-2004005247-A1
EP-1517679-A1
EP-1517679-B1
EP-1517883-A1
EP-1517883-B1
EP-1517883-B8
US-2005222261-A1
US-2005256198-A1
US-2005267149-A1
US-7351858-B2
US-7355069-B2
US-7521461-B2
WO-2004000294-A1
WO-2004000790-A1
EP-1478368-A1
US-2005124557-A1
WO-03061663-A1
EP-1458672-A1
EP-1458672-B1
EP-1458673-A1
EP-1458673-B1
EP-1458677-A1
EP-1458677-B1
US-2005113362-A1
US-2005171204-A1
US-2005215630-A1
US-2005282822-A1
US-7256307-B2
US-7276539-B2
US-7488844-B2
US-7514471-B2
WO-03051821-A1
WO-03051822-A1
WO-03051826-A1
EP-1427423-B1
EP-1427423-B9
US-2005038009-A1
US-7132416-B2

Viloxazine, ヴィロキサジン;


Viloxazine structure.svg

ChemSpider 2D Image | Viloxazine | C13H19NO3

Viloxazine

  • Molecular FormulaC13H19NO3
  • Average mass237.295 Da

update FDA APPROVED 2021/4/2, Qelbree, Viloxazine hydrochloride

Formula
C13H19NO3. HCl
CAS
35604-67-2
Mol weight
273.7558
 
2-[(2-Ethoxyphenoxy)methyl]morpholine
 
256-281-7 [EINECS]
3489
46817-91-8 free [RN], Hcl 35604-67-2
5I5Y2789ZF
Emovit [Wiki]
Morpholine, 2-((2-ethoxyphenoxy)methyl)-
Morpholine, 2-[(2-ethoxyphenoxy)methyl]-
UNII:5I5Y2789ZF
Viloxazine hydrochloride.png
Viloxazine hydrochloride OQW30I1332 35604-67-2

Polymorph

FORM A , B US226136693US2011032013

NEW DRUG APPROVALS

ONE TIME

$10.00

Viloxazine (trade names VivalanEmovitVivarint and Vicilan) is a morpholine derivative and is a selective norepinephrine reuptake inhibitor (NRI). It was used as an antidepressant in some European countries, and produced a stimulant effect that is similar to the amphetamines, except without any signs of dependence. It was discovered and brought to market in 1976 by Imperial Chemical Industries and was withdrawn from the market in the early 2000s for business reasons.

Image result for viloxazine synthesis

Clip

https://www.sciencedirect.com/science/article/pii/S0040402015302659

Image result for viloxazine synthesis

Patent

US 20180265482

 Viloxazine ((R,S)-2-[(2-ethoxyphenoxy)methyl]morpholine]) is a bicyclic morpholine derivative, assigned CAS No. 46817-91-8 (CAS No. 35604-67-2 for the HCl salt). It is characterized by the formula C 1319NO 3, with a molecular mass of 237.295 g/mol. Viloxazine has two stereoisomers, (S)-(−)- and (R)-(+)-isomer, which have the following chemical structures:
 (MOL) (CDX)
      Viloxazine is known to have several desirable pharmacologic uses, including treatment of depression, nocturnal enuresis, narcolepsy, sleep disorders, and alcoholism, among others. In vivo, viloxazine acts as a selective norepinephrine reuptake inhibitor (“NRI”).
      Between the two stereoisomers, the (S)-(−)-isomer is known to be five times as pharmacologically active as the (R)-(+)-isomer. See, e.g., “Optical Isomers of 2-(2-ethoxyphenoxymethyl)tetrahydro-1,4 oxazine (viloxazine) and Related Compounds” (Journal of Medicinal Chemistry, Jan. 9, 1976, 19(8); 1074) in which it is disclosed that optical isomers of 2-(2-ethoxyphenoxymethyl)tetrahydro-1,4-oxazine (viloxazine) and 2-(3-methoxyphenoxymethyl)tetrahydro-1,4-oxazine were prepared and absolute configurations assigned. The synthesis of optical isomers of viloxazine analogs of known configuration was accomplished by resolution of the intermediate 4-benzyl-2-(p-toluenesulfonyloxymethyl)tetrahydro-1,4-oxazine isomers.
      Some unsatisfactory methods of synthesizing viloxazine are known in the art. For example, as disclosed in U.S. Pat. No. 3,714,161, viloxazine is prepared by reacting ethoxyphenol with epichlorohydrin to afford the epoxide intermediate 1-(2-ethoxyphenoxy)-2,3-epoxypropane. This epoxide intermediate is then treated with benzylamine followed with chloroacetyl chloride. The resulting morpholinone is then reduced by lithium aluminum hydride and then by Pd/C-catalyzed hydrogenation to yield viloxazine free base.
      Yet another unsatisfactory synthesis of viloxazine is disclosed in U.S. Pat. No. 3,712,890, which describes a process to prepare viloxazine HCl, wherein the epoxide intermediate, 1-(2-ethoxyphenoxy)-2,3-epoxypropane, is reacted with 2-aminoethyl hydrogen sulfate in ethanol in the presence of sodium hydroxide to form viloxazine free base. The product is extracted with diethyl ether from the aqueous solution obtained by evaporating the solvent in the reaction mixture then adding water to the residue. The ethereal extract is dried over a drying agent and the solvent is removed. Viloxazine HCl salt is finally obtained by dissolving the previous residue in isopropanol, concentrated aqueous HCl, and ethyl acetate followed by filtration.
      The foregoing methods of synthesizing viloxazine suffer from a number of deficiencies, such as low reaction yield and unacceptably large amount of impurities in the resulting product. Effective elimination or removal of impurities, especially those impurities possessing genotoxicity or other toxicities, is critical to render safe pharmaceutical products. For example, certain reagents traditionally utilized in viloxazine HCl preparation, such as epichlorohydrin and 2-aminoethyl hydrogen sulfate, present a special problem due to their toxicity. There is a need for effective methods to remove or limit harmful impurities down to a level that is appropriate and safe according to contemporary sound medical standards and judgment. Accordingly, a continuing and unmet need exists for new and improved methods of manufacturing viloxazine and its various salts to yield adequate quantities of pharmacologically desirable API with predictable and reliable control of impurities.
     Polymorph control is also an important aspect of producing APIs and their associated salts that are used in pharmaceutical products. However, no polymorphs of viloxazine HCl have previously been disclosed. A need therefore exists for new polymorphic forms of viloxazine that have improved pharmacological properties.

PATENT

WO 2011130194

US2011032013

For the sake of convenience and without putting any limitations thereof, the methods of manufacture of viloxazine have been separated into several steps, each step being disclosed herein in a multiplicity of non-limiting embodiments. These steps comprise Step 1, during which 2-ethoxyphenol and epichlorhydrin are reacted to produce l-(2-ethoxyphenoxy)-2,3-epoxypropane (Epoxide 1); Step 2, during which l-(2-ethoxyphenoxy)-2,3-epoxypropane (Epoxide 1) is converted into viloxazine base which is further converted into viloxazine salt, and Step 3, during which viloxazine salt is purified/recrystallized, and various polymorphic forms of viloxazine salt are prepared.

The above-mentioned steps will be considered below in more details.

[0031] The process of the Step 1 may be advantageously carried out in the presence of a phase-transfer catalyst to afford near quantitative yield of l-(2-ethoxyphenoxy)-2,3-epoxypropane. Alternatively, the process may make use of a Finkelstein catalyst described in more details below. Additionally, the reaction may take place without the use of the catalyst.

 FIG. 1, depicted below, schematically illustrates the preparation of l-(2-ethoxyphenoxy)-2,3-epoxypropane (“Epoxide 1”) in accordance with Step I of an exemplary synthesis of viloxazine:

STEP I:

Epoxide 1

In one embodiment of the Step 1, the preparation of l-(2-ethoxyphenoxy)-2,3-epoxypropane (epoxide 1) can be effected by the use of a phase transfer catalyst in the presence of a solid or liquid base with a solution of a corresponding phenol and epichlorohydrin in one or more solvents (Fig. 1). The phase transfer catalyst can be selected from ammonium salts, such as benzyltriethylammonium salts, benzyltrimethylammonium salts, and tetrabutylammonium salts, phosphonium salts, guanidinium salts, crown ether, polyethylene glycol, polyethylene glycol ether, or polyethylene glycol ester, or other phase transfer catalysts know in the art. The solid or liquid base can be a carbonate such as alkali carbonate, NaOH, KOH, LiOH, LiOH/LiCl, amines such as mono-, di- or tri-substituted amines (such as diethylamine, triethylamine, dibutylamine, tributylamine), DMAP, or other appropriate base. The solvents used in the solution of a corresponding phenol and epichlorohydrin include but are not limited to ethers such as methyl t-butyl ether, ketones, non-substituted or substituted aromatic solvents (xylene), halo-substituted hydrocarbons (e.g. CH2C12, CHC13), THF, DMF, dioxanes, non-substituted and substituted pyridines, acetonitrile, pyrrolidones, nitromethane , or other appropriate solvent. Additional catalyst, such as, for example, Finkelstein catalyst, can also be used in the process of this embodiment. This reaction preferably takes place at an elevated temperature. In one variation of the embodiment, the temperature is above 50°C. In another variation, epichlorohydrin, potassium carbonate, and a phase transfer catalyst are mixed with a solution of 2-ethoxyphenol in a solvent at an elevated temperature, such as 50 – 60°C. After the reaction is complete, the reaction mixture can be washed with water, followed by work-up procedures known in the art. Variations of this embodiment of the invention are further disclosed in Examples 1-8.

[0033] In one variation of the above embodiment of the Step 1 , Epoxide 1 is prepared by reacting 2-ethoxyphenol and epichlorohydrin in a solvent in the presence of two different catalysts, and a base in a solid state. The first catalyst is a phase transfer catalyst as described above; the second catalyst is a Finkelstein reaction catalyst. Without putting any limitation

hereon, metal iodide and metal bromide salts, such as potassium iodide, may be used as an example of a Finkelstein catalyst. The phase transfer catalyst and a solvent may be selected from any phase transfer catalysts and solvents known in the art. Potassium carbonate may be used as a non-limiting example of a solid base. Using the solid base in a powdered form may be highly beneficial due to the greatly enhanced interface and limiting the side reactions. This variation of the embodiment is further illustrated by Example 9. In another variation of the embodiment, liquid base such as triethylamine can be used to replace the solid base.

[0034] In a different embodiment of Step 1 , 2-ethoxyphenol and epichlorohydrin are reacted in a solvent-free system that comprises a solid or liquid base, a phase transfer catalyst as listed above and a Finkelstein catalyst.

[0035] FIG. 2, depicted below, schematically illustrates the preparation of l-(2-ethoxyphenoxy)-2,3-epoxypropane (“Epoxide 1”) in accordance with the Step I of another exemplary synthesis of viloxazine ( biphasic):

STEP I (alternative embodiment):

In this embodiment of Step 1, illustrated in Fig. 2, Epoxide 1 can be prepared by reacting epichlorohydrin with 2-ethoxyphenol in the presence of a catalytic amount of a phase transfer catalyst without the use of solvents at elevated temperatures in a two-stage process to afford near quantitative yield of l-(2-ethoxyphenoxy)-2,3-epoxypropane with very few side products. This embodiment of the invention is further illustrated by a non-limiting Example 12. The phase transfer catalyst for this embodiment can be selected from ammonium salts such as benzyltriethylammonium salts, benzyltrimethylammonium salts, tetrabutylammonium salts, etc; phosphonium salts, guanidinium salts, crown ether, polyethylene glycol, polyethylene glycol ether, or polyethylene glycol ester, or other phase transfer catalysts know in the art. The first stage of the process of this embodiment may take place without a solvent in a presence of a large excess of epichlorohydrin. This stage is followed by a de-chlorination stage, before or after

removal of excess epichlorohydrin, using a base and a solvent. The reaction produces l-(2-ethoxyphenoxy)-2,3-epoxypropane in high yield. Example of the bases used herein include but are not limited to NaOH, KOH, LiOH, LiOH/LiCl, K2C03, Na2C03, amines such as mono-, di-or tri-substituted amines (such as diethylamine, triethylamine, dibutylamine, tributylamine etc.), DMAP. In one variation of this embodiment of Step 1, the phase transfer catalyst may be used only at the de-chlorination stage of the process. The de-chlorination stage can be carried out in a biphasic system or in a single phase system. For a biphasic system, it can be an organic-aqueous liquid biphasic system, or a liquid-solid biphasic system. Solvents that are useful for the process include but are not limited to non-substituted and substituted aromatic solvents (e.g. toluene, benzene, chlorobenzene, dimethylbenzene, xylene), halo-substituted hydrocarbons (e.g. CH2C12, CHC13), THF, dioxanes, DMF, DMSO, non-substituted and substituted pyridines, ketones, pyrrolidones, ethers, acetonitrile, nitromethane. As mentioned above, this process takes place at the elevated temperature. In one variation of the embodiment, the temperature is above 60°C. In another variation, 2-ethoxyphenol and epichlorohydrin are heated to 60 – 90°C for a period of time in the presence of phase transfer catalyst. Excess of epichlorohydrin is removed and the residue is dissolved in a solvent such as toluene or benzene treated with an aqueous base solution, such as NaOH, KOH, LiOH, LiOH/LiCl. In yet another variation of the embodiment, the residue after epichlorohydrin removal can be dissolved in one or more of the said solvent and treated with a base (solid or liquid but not an aqueous solution) and optionally a second phase transfer catalyst, optionally at elevated temperatures.

[0036] In yet another embodiment of Step 1 , Epoxide 1 can also be prepared by using a catalyst for a so-called Finkelstein reaction in the presence of a Finkelstein catalyst but without the need to use a phase transfer catalyst. Finkelstein catalysts useful herein include metal iodide salts and metal bromide salts, among others. In one variation of this embodiment, 2-ethoxyphenol and epichlorohydrin are dissolved in a polar aprotic solvent such as DMF, and a catalytic amount of an iodide such as potassium iodide and a base, as solid or liquid, are used. Preferably, the base is used as a solid, such as potassium carbonate powder. This embodiment is further illustrated by the Example 11.

[0037] In the alternative embodiment of Step 1 , Epoxide 1 can also be prepared by a different method that comprises reacting epichlorohydrin and the corresponding phenol in the presence of a base at a temperature lower than the ambient temperature, especially when a base solution is used, and without the use of a phase transfer catalyst. This embodiment is illustrated by the Example 10.

[0038] A very high, almost quantitative, yield of 1 -(2-ethoxyphenoxy)-2,3-epoxypropane can be obtained through realizing the above-described embodiments of Step 1 , with less impurities generated in Epoxide 1.

[0039] Epoxide 1 , produced in Step 1 as described above, is used to prepare viloxazine base (viloxazine), which is further converted into viloxazine salt through the processes of Step 2.

[0040] FIG. 3, depicted below, schematically illustrates the preparation of viloxazine

(“Step Ila”) and the preparation of viloxazine hydrochloride (“Step lib”), as well as their purification (“Step III”) in accordance with another example embodiment hereof:

STEP Ila:

Hydrogen Sulfate

STEP lib:

Step III:

Conversion

Viloxazine free base ► Viloxazine salt

Wash/ raction

Recrystallization

Purified viloxazine salt

In the embodiment of Step 2, illustrated in Fig. 3, the preparation of viloxazine base is achieved by reacting the Epoxide 1 intermediate prepared in Step 1 and aminoethyl hydrogen sulfate in presence of a large excess of a base as illustrated by the Examples 5-7 and 14. The base may be present as a solid or in a solution. Preferably, the molar ratio of the base to Epoxide 1 is more than 10. More preferably the ratio is more than 12. Even more preferably, the ratio is between 15 and 40. It was unexpectedly discovered that the use of a higher ratio of a base results in a faster reaction, less impurities, and lower reaction temperature.

[0041] Further advantages may be offered by a specific variation of this embodiment, wherein the base is added to the reaction mixture in several separate steps. For example, a third of the base is added to the reaction mixture, and the mixture is stirred for a period of time. Then the rest of the base is added followed by additional stirring. Alternatively, half of the base is added initially followed by the second half after some period of time, or the base is added in three different parts separated by periods of time. The bases used herein include but are not limited to NaOH, KOH, LiOH, LiOH/LiCl, K2C03, Na2C03, amines such as mono-, di- or tri-substituted amines (such as diethylamine, triethylamine, dibutylamine, tributylamine), DMAP, and combinations thereof. . In one embodiment of the invention, the base is KOH. In another embodiment, the base is NaOH. In a further embodiment, the base is K2C03 powder. In yet further embodiment, the base is triethylamine. This embodiment is illustrated further by

Examples 13,15 and 16.

[0042] In another exemplary embodiment of Step 2, viloxazine is produced by cyclization of novel intermediate compound “Diol 1 ,” which is made from Epoxide 1 and N-benzyl-aminoethanol. This method allows one to drastically reduce the use of potentially toxic materials in the manufacturing process, completely eliminating some of them such as aminoethyl hydrogen sulfate. The first stage of the reaction results in the formation of an intermediate of Formula 3 (Diol 1), which is a new, previously unidentified compound.

[0043] Formula 3

Diol 1

FIG. 4, depicted below, schematically illustrates the preparation of viloxazine and its salts via “Diol 1” in accordance with another exemplary embodiment hereof (Bn = benzyl, Et = ethyl):

Viloxazine HCI

As illustrated in Fig. 4, Diol 1 is turned into N-benzyl viloxazine by cyclization. Removal of the benzyl protective group yields viloxazine base. Similarly, FIG. 5, depicted below, schematically illustrates the cyclization of Diol 1, as well as some side-reactions thereof.

Uses

Viloxazine hydrochloride was used in some European countries for the treatment of clinical depression.[4][5]

Side effects

Side effects included nausea, vomiting, insomnia, loss of appetite, increased erythrocyte sedimentation, EKG and EEG anomalies, epigastric pain, diarrhea, constipationvertigoorthostatic hypotensionedema of the lower extremities, dysarthriatremor, psychomotor agitation, mental confusion, inappropriate secretion of antidiuretic hormone, increased transaminasesseizure, (there were three cases worldwide, and most animal studies (and clinical trials that included epilepsy patients) indicated the presence of anticonvulsant properties, so was not completely contraindicated in epilepsy,[6]) and increased libido.[7]

Drug interactions

Viloxazine increased plasma levels of phenytoin by an average of 37%.[8] It also was known to significantly increase plasma levels of theophylline and decrease its clearance from the body,[9] sometimes resulting in accidental overdose of theophylline.[10]

Mechanism of action

Viloxazine, like imipramine, inhibited norepinephrine reuptake in the hearts of rats and mice; unlike imipramine, it did not block reuptake of norepinephrine in either the medullae or the hypothalami of rats. As for serotonin, while its reuptake inhibition was comparable to that of desipramine (i.e., very weak), viloxazine did potentiate serotonin-mediated brain functions in a manner similar to amitriptyline and imipramine, which are relatively potent inhibitors of serotonin reuptake.[11] Unlike any of the other drugs tested, it did not exhibit any anticholinergic effects.[11]

It was also found to up-regulate GABAB receptors in the frontal cortex of rats.[12]

Chemical properties

It is a racemic compound with two stereoisomers, the (S)-(–)-isomer being five times as pharmacologically active as the (R)-(+)-isomer.[13]

History

Viloxazine was discovered by scientists at Imperial Chemical Industries when they recognized that some beta blockers inhibited serotonin reuptake inhibitor activity in the brain at high doses. To improve the ability of their compounds to cross the blood brain barrier, they changed the ethanolamine side chain of beta blockers to a morpholine ring, leading to the synthesis of viloxazine.[14]:610[15]:9 The drug was first marketed in 1976.[16] It was never approved by the FDA,[5] but the FDA granted it an orphan designation (but not approval) for cataplexy and narcolepsy in 1984.[17] It was withdrawn from markets worldwide in 2002 for business reasons.[14][18]

As of 2015, Supernus Pharmaceuticals was developing formulations of viloxazine as a treatment for ADHD and major depressive disorder under the names SPN-809 and SPN-812.[19][20]

Research

Viloxazine has undergone two randomized controlled trials for nocturnal enuresis (bedwetting) in children, both of those times versus imipramine.[21][22] By 1990, it was seen as a less cardiotoxic alternative to imipramine, and to be especially effective in heavy sleepers.[23]

In narcolepsy, viloxazine has been shown to suppress auxiliary symptoms such as cataplexy and also abnormal sleep-onset REM[24] without really improving daytime somnolence.[25]

In a cross-over trial (56 participants) viloxazine significantly reduced EDS and cataplexy.[18]

Viloxazine has also been studied for the treatment of alcoholism, with some success.[26]

While viloxazine may have been effective in clinical depression, it did relatively poorly in a double-blind randomized controlled trial versus amisulpride in the treatment of dysthymia.[27]

It is also under investigation as a treatment for attention deficit hyperactivity disorder.[28]

REFERNCES

  1. ^ Bouchard JM, Strub N, Nil R (October 1997). “Citalopram and viloxazine in the treatment of depression by means of slow drop infusion. A double-blind comparative trial”. Journal of Affective Disorders46 (1): 51–8. doi:10.1016/S0165-0327(97)00078-5PMID 9387086.
  2. ^ Case DE, Reeves PR (February 1975). “The disposition and metabolism of I.C.I. 58,834 (viloxazine) in humans”. Xenobiotica5 (2): 113–29. doi:10.3109/00498257509056097PMID 1154799.
  3. ^ “SID 180462– PubChem Substance Summary”. Retrieved 5 November 2005.
  4. ^ Pinder, RM; Brogden, RN; Speight, ™; Avery, GS (June 1977). “Viloxazine: a review of its pharmacological properties and therapeutic efficacy in depressive illness”. Drugs13 (6): 401–21. doi:10.2165/00003495-197713060-00001PMID 324751.
  5. Jump up to:a b Dahmen, MM, Lincoln, J, and Preskorn, S. NARI Antidepressants, pp 816-822 in Encyclopedia of Psychopharmacology, Ed. Ian P. Stolerman. Springer-Verlag Berlin Heidelberg, 2010. ISBN 9783540687061
  6. ^ Edwards JG, Glen-Bott M (September 1984). “Does viloxazine have epileptogenic properties?”Journal of Neurology, Neurosurgery, and Psychiatry47 (9): 960–4. doi:10.1136/jnnp.47.9.960PMC 1027998PMID 6434699.
  7. ^ Chebili S, Abaoub A, Mezouane B, Le Goff JF (1998). “Antidepressants and sexual stimulation: the correlation” [Antidepressants and sexual stimulation: the correlation]. L’Encéphale (in French). 24 (3): 180–4. PMID 9696909.
  8. ^ Pisani F, Fazio A, Artesi C, et al. (February 1992). “Elevation of plasma phenytoin by viloxazine in epileptic patients: a clinically significant drug interaction”Journal of Neurology, Neurosurgery, and Psychiatry55 (2): 126–7. doi:10.1136/jnnp.55.2.126PMC 488975PMID 1538217.
  9. ^ Perault MC, Griesemann E, Bouquet S, Lavoisy J, Vandel B (September 1989). “A study of the interaction of viloxazine with theophylline”. Therapeutic Drug Monitoring11 (5): 520–2. doi:10.1097/00007691-198909000-00005PMID 2815226.
  10. ^ Laaban JP, Dupeyron JP, Lafay M, Sofeir M, Rochemaure J, Fabiani P (1986). “Theophylline intoxication following viloxazine induced decrease in clearance”. European Journal of Clinical Pharmacology30 (3): 351–3. doi:10.1007/BF00541543PMID 3732375.
  11. Jump up to:a b Lippman W, Pugsley TA (August 1976). “Effects of viloxazine, an antidepressant agent, on biogenic amine uptake mechanisms and related activities”. Canadian Journal of Physiology and Pharmacology54 (4): 494–509. doi:10.1139/y76-069PMID 974878.
  12. ^ Lloyd KG, Thuret F, Pilc A (October 1985). “Upregulation of gamma-aminobutyric acid (GABA) B binding sites in rat frontal cortex: a common action of repeated administration of different classes of antidepressants and electroshock”The Journal of Pharmacology and Experimental Therapeutics235 (1): 191–9. PMID 2995646.
  13. ^ Danchev ND, Rozhanets VV, Zhmurenko LA, Glozman OM, Zagorevskiĭ VA (May 1984). “Behavioral and radioreceptor analysis of viloxazine stereoisomers” [Behavioral and radioreceptor analysis of viloxazine stereoisomers]. Biulleten’ Eksperimental’noĭ Biologii i Meditsiny (in Russian). 97 (5): 576–8. PMID 6326891.
  14. Jump up to:a b Williams DA. Antidepressants. Chapter 18 in Foye’s Principles of Medicinal Chemistry, Eds. Lemke TL and Williams DA. Lippincott Williams & Wilkins, 2012. ISBN 9781609133450
  15. ^ Wermuth, CG. Analogs as a Means of Discovering New Drugs. Chapter 1 in Analogue-based Drug Discovery. Eds.IUPAC, Fischer, J., and Ganellin CR. John Wiley & Sons, 2006. ISBN 9783527607495
  16. ^ Olivier B, Soudijn W, van Wijngaarden I. Serotonin, dopamine and norepinephrine transporters in the central nervous system and their inhibitors. Prog Drug Res. 2000;54:59-119. PMID 10857386
  17. ^ FDA. Orphan Drug Designations and Approvals: Viloxazine Page accessed August 1, 2-15
  18. Jump up to:a b Vignatelli L, D’Alessandro R, Candelise L. Antidepressant drugs for narcolepsy. Cochrane Database Syst Rev. 2008 Jan 23;(1):CD003724. Review. PMID 18254030
  19. ^ Bloomberg Supernus profile Page accessed August 1, 2015
  20. ^ Supernus. Psychiatry portfolio Page accessed August 1, 2015
  21. ^ Attenburrow AA, Stanley TV, Holland RP (January 1984). “Nocturnal enuresis: a study”. The Practitioner228 (1387): 99–102. PMID 6364124.
  22. ^ ^ Yurdakök M, Kinik E, Güvenç H, Bedük Y (1987). “Viloxazine versus imipramine in the treatment of enuresis”. The Turkish Journal of Pediatrics29 (4): 227–30. PMID 3332732.
  23. ^ Libert MH (1990). “The use of viloxazine in the treatment of primary enuresis” [The use of viloxazine in the treatment of primary enuresis]. Acta Urologica Belgica (in French). 58 (1): 117–22. PMID 2371930.
  24. ^ Guilleminault C, Mancuso J, Salva MA, et al. (1986). “Viloxazine hydrochloride in narcolepsy: a preliminary report”. Sleep9 (1 Pt 2): 275–9. PMID 3704453.
  25. ^ Mitler MM, Hajdukovic R, Erman M, Koziol JA (January 1990). “Narcolepsy”Journal of Clinical Neurophysiology7 (1): 93–118. doi:10.1097/00004691-199001000-00008PMC 2254143PMID 1968069.
  26. ^ Altamura AC, Mauri MC, Girardi T, Panetta B (1990). “Alcoholism and depression: a placebo controlled study with viloxazine”. International Journal of Clinical Pharmacology Research10 (5): 293–8. PMID 2079386.
  27. ^ León CA, Vigoya J, Conde S, Campo G, Castrillón E, León A (March 1994). “Comparison of the effect of amisulpride and viloxazine in the treatment of dysthymia” [Comparison of the effect of amisulpride and viloxazine in the treatment of dysthymia]. Acta Psiquiátrica Y Psicológica de América Latina (in Spanish). 40 (1): 41–9. PMID 8053353.
  28. ^ Mattingly, GW; Anderson, RH (December 2016). “Optimizing outcomes in ADHD treatment: from clinical targets to novel delivery systems”. CNS Spectrums21 (S1): 45–59. doi:10.1017/S1092852916000808PMID 28044946.
Patent ID Title Submitted Date Granted Date 
US2004229942Analeptic and antidepressant combinations2004-05-122004-11-18
US2004242698Analeptic and antidepressant combinations2004-05-122004-12-02
US2002192302Transdermal and topical administration of antidepressant drugs using basic enhancers2002-06-192002-12-19
US2018105877GENETIC POLYMORPHISMS ASSOCIATED WITH DEPRESSION2017-05-19 
US2013244990GENETIC POLYMORPHISMS ASSOCIATED WITH DEPRESSION2012-11-302013-09-19
Patent ID Title Submitted Date Granted Date 
US2009197849TRANSDERMAL AND TOPICAL ADMINISTRATION OF DRUGS USING BASIC PERMEATION ENHANCERS2008-09-032009-08-06
US2006150989Method of diagnosing, treating and educating individuals with and/or about depression2005-01-122006-07-13
US8153159Modafinil modified release pharmaceutical compositions2004-09-172012-04-10
US2004229940Analeptic and antidepressant combinations2004-05-122004-11-18
US2004229941Analeptic and antidepressant combinations2004-05-122004-11-18
Patent ID Title Submitted Date Granted Date 
US9434703METHODS FOR PRODUCING VILOXAZINE SALTS AND NOVEL POLYMORPHS THEREOF2014-12-192015-05-07
US2012220962TRANSDERMAL AND TOPICAL ADMINISTRATION OF VITAMINS USING BASIC PERMEATION ENHANCERS2012-05-102012-08-30
US9358204FORMULATIONS OF VILOXAZINE2013-02-072013-08-08
US2012289515Combination therapy for cognitive distortions, resisting relapse of unipolar non-psychotic depression2011-05-252012-11-15
US2009317453TRANSDERMAL AND TOPICAL ADMINISTRATION OF DRUGS USING BASIC PERMEATION ENHANCERS2008-12-232009-12-24
Patent ID Title Submitted Date Granted Date 
US2006013834Room temperature stable aqueous liquid pharmaceutical composition2005-02-252006-01-19
US2004220262Transdermal and topical administration of drugs using basic permeation enhancers2004-06-032004-11-04
US2003124176Transdermal and topical administration of drugs using basic permeation enhancers2002-06-212003-07-03
US6713089Quick release pharmaceutical compositions of drug substances2001-07-102004-03-30
WO0015195QUICK RELEASE PHARMACEUTICAL COMPOSITIONS OF DRUG SUBSTANCES2000-03-23 
Patent ID Title Submitted Date Granted Date 
US2014315720POLYSACCHARIDE ESTER MICROSPHERES AND METHODS AND ARTICLES RELATING THERETO2014-04-042014-10-23
US2014113826POLYSACCHARIDE ESTER MICROSPHERES AND METHODS AND ARTICLES RELATING THERETO2013-10-232014-04-24
US2014052264Porous, Stabilized Craniomaxillofacial Implants and Methods and Kits Relating Thereto2013-08-122014-02-20
US2014348936GASTRORETENTIVE CONTROLLED RELEASE VEHICLES THAT INCLUDE ETHYLENE COPOLYMERS, ETHYL CELLULOSES, AND/OR THERMOPLASTIC POLYURETHANES2012-12-172014-11-27
US2016304475METHODS FOR PRODUCING VILOXAZINE SALTS AND NOVEL POLYMORPHS THEREOF2016-06-24 
Patent ID Title Submitted Date Granted Date 
US2017319698GASTRORETENTIVE GEL FORMULATIONS2015-10-26 
US8231899Quick release pharmaceutical compositions of drug substances2004-01-132012-07-31
US9662338FORMULATIONS OF VILOXAZINE2016-06-032016-09-29
US9603853FORMULATIONS OF VILOXAZINE2016-05-182016-09-08
US2015306230DRUG DELIVERY VEHICLES COMPRISING CELLULOSE DERIVATIVES, STARCH DERIVATIVES, AND COMBINATIONS THEREOF2014-06-122015-10-29
Patent ID Title Submitted Date Granted Date 
US3959273Morpholine derivatives1976-05-25 
US8802596Multi-functional ionic liquid compositions for overcoming polymorphism and imparting improved properties for active pharmaceutical, biological, nutritional, and energetic ingredients2012-06-072014-08-12
US6060516N1-propargylhydrazines, N2-propargylhydrazines and their analogs for the treatment of depression, anxiety and neurodegeneration2000-05-09 
US6479491Disubstituted morpholine, oxazepine or thiazepine derivatives, their preparation and their use as dopamine d4 receptor antagonists 2002-11-12
US2017258801FORMULATIONS OF VILOXAZINE2017-05-25 
Patent ID Title Submitted Date Granted Date 
US6034117Methods of treating and diagnosing sleep disordered breathing and means for carrying out the method2000-03-07 
EP0923370ACETYL CHOLINE ESTERASE INHIBITORS FOR TREATING AND DIAGNOSING SLEEP DISORDERED BREATHING1999-06-232005-11-16
US2014328884CONTROLLED RELEASE VEHICLES HAVING DESIRED VOID VOLUME ARCHITECTURES2012-12-172014-11-06
US9096743PROCESS FOR FORMING FILMS, FIBERS, AND BEADS FROM CHITINOUS BIOMASS2010-06-012012-05-10
US2003104041Transdermal and topical administration of drugs using basic permeation enhancers2002-06-202003-06-05
Patent ID Title Submitted Date Granted Date 
WO9807710DISUBSTITUTED MORPHOLINE, OXAZEPINE OR THIAZEPINE DERIVATIVES, THEIR PREPARATION AND THEIR USE AS DOPAMINE D4 RECEPTOR ANTAGONISTS1998-02-26 
US2014051771Biomedical Devices Comprising Molded Polyethylene Components2013-08-122014-02-20
US9278134DUAL FUNCTIONING IONIC LIQUIDS AND SALTS THEREOF2009-12-292012-02-23
US8232265Multi-functional ionic liquid compositions for overcoming polymorphism and imparting improved properties for active pharmaceutical, biological, nutritional, and energetic ingredients2007-04-262012-07-31
US2005074487Transdermal and topical administration of drugs using basic permeation enhancers2004-06-072005-04-07
Patent ID Title Submitted Date Granted Date 
US9199918SMALL MOLECULE INHIBITORS OF AGBL22012-02-142013-12-12
US9403783METHODS FOR PRODUCING VILOXAZINE SALTS AND NOVEL POLYMORPHS THEREOF2011-10-13 
US7973043Combination therapy for depression, prevention of suicide, and various medical and psychiatric conditions2003-07-252011-07-05
US6207662Disubstituted morpholine, oxazepine or thiazepine derivatives, their preparation and their use as dopamine D4 receptor antagonists 2001-03-27
EP0920423DISUBSTITUTED MORPHOLINE, OXAZEPINE OR THIAZEPINE DERIVATIVES, THEIR PREPARATION AND THEIR USE AS DOPAMINE D4 RECEPTOR ANTAGONISTS1999-06-092005-01-26
Clinical data
Routes of
administration
By mouthintravenous infusion[1]
ATC codeN06AX09 (WHO)
Legal status
Legal statusIn general: uncontrolled
Pharmacokinetic data
Elimination half-life2–5 hours
ExcretionRenal[2]
Identifiers
IUPAC name[show]
CAS Number46817-91-8  35604-67-2 (HCl salt)
PubChem CID5666
ChemSpider5464 
UNII5I5Y2789ZF
KEGGD08673 
ChEMBLChEMBL306700 
ECHA InfoCard100.051.148 
Chemical and physical data
FormulaC13H19NO3
Molar mass237.295 g/mol g·mol−1
3D model (JSmol)Interactive image
ChiralityRacemic mixture
SMILES[hide]O(c1ccccc1OCC)CC2OCCNC2
InChI[hide]InChI=1S/C13H19NO3/c1-2-15-12-5-3-4-6-13(12)17-10-11-9-14-7-8-16-11/h3-6,11,14H,2,7-10H2,1H3 Key:YWPHCCPCQOJSGZ-UHFFFAOYSA-N 

/////////////////Viloxazine, ヴィロキサジン , Emovit, Vivalan, Emovit, Vivarint, Vicilan

ABL 001, Asciminib


img

Image result for ABL001 / Asciminib

ABL001 / Asciminib

Cas 1492952-76-7
Chemical Formula: C20H18ClF2N5O3
Molecular Weight: 449.8428
Elemental Analysis: C, 53.40; H, 4.03; Cl, 7.88; F, 8.45; N, 15.57; O, 10.67

N-[4-[Chloro(difluoro)methoxy]phenyl]-6-[(3R)-3-hydroxypyrrolidin-1-yl]-5-(1H-pyrazol-5-yl)pyridine-3-carboxamide

3-Pyridinecarboxamide, N-[4-(chlorodifluoromethoxy)phenyl]-6-[(3R)-3-hydroxy-1-pyrrolidinyl]-5-(1H-pyrazol-3-yl)-

PHASE 3, Chronic Myeloid Leukemia, NOVARTIS

UPDATE FDA APPROVED 10/29/2021,

Scemblix

To treat Philadelphia chromosome-positive chronic myeloid leukemia with disease that meets certain criteria

Asciminib, sold under the brand name Scemblix, is a medication used to treat Philadelphia chromosome-positive chronic myeloid leukemia (Ph+ CML).[1][2][3] Asciminib is a protein kinase inhibitor.[1]

The most common adverse reactions include upper respiratory tract infections, musculoskeletal pain, fatigue, nausea, rash, and diarrhea.[2]

Asciminib was approved for medical use in the United States in October 2021.[1][4][5]

The U.S. Food and Drug Administration (FDA) granted the application for asciminib priority reviewfast trackorphan drug, and breakthrough therapy designations.[2][6][7]

Asciminib is an orally bioavailable, allosteric Bcr-Abl tyrosine kinase inhibitor with potential antineoplastic activity. Designed to overcome resistance, ABL001 binds to the Abl portion of the Bcr-Abl fusion protein at a location that is distinct from the ATP-binding domain. This binding results in the inhibition of Bcr-Abl-mediated proliferation and enhanced apoptosis of Philadelphia chromosome-positive (Ph+) hematological malignancies. The Bcr-Abl fusion protein tyrosine kinase is an abnormal enzyme produced by leukemia cells that contain the Philadelphia chromosome.

ABL001 has been used in trials studying the health services research of Chronic Myelogenous Leukemia and Philadelphia Chromosome-positive Acute Lymphoblastic Leukemia.
  • Originator Novartis
  • Developer Novartis; Novartis Oncology
  • Class Antineoplastics; Pyrazoles; Pyrrolidines; Small molecules
  • Mechanism of Action Bcr-abl tyrosine kinase inhibitors

Highest Development Phases

  • Phase III Chronic myeloid leukaemia
  • No development reported Precursor cell lymphoblastic leukaemia-lymphoma

Most Recent Events

  • 04 Nov 2017 No recent reports of development identified for phase-I development in Acute-lymphoblastic-leukaemia(Second-line therapy or greater) in Australia (PO)
  • 04 Nov 2017 No recent reports of development identified for phase-I development in Acute-lymphoblastic-leukaemia(Second-line therapy or greater) in France (PO)
  • 04 Nov 2017 No recent reports of development identified for phase-I development in Acute-lymphoblastic-leukaemia(Second-line therapy or greater) in Germany (PO)
  • The tyrosine kinase activity of the ABLl protein is normally tightly regulated, with the N-terminal cap region of the SH3 domain playing an important role. One regulatory mechanism involves the N-terminal cap glycine-2 residue being myristoylated and then interacting with a myristate binding site within the SHI catalytic domain. A hallmark of chronic myeloid leukemia (CML) is the Philadelphia chromosome (Ph), formed by the t(9,22) reciprocal chromosome translocation in a haematopoietic stem cell. This chromosome carries the BCR-ABL1 oncogene which encodes the chimeric BCR-ABL1 protein, that lacks the N-terminal cap and has a constitutively active tyrosine kinase domain.Although drugs that inhibit the tyrosine kinase activity of BCR-ABL1 via an ATP-competitive mechanism, such as Gleevec® / Glivec® (imatinib), Tasigna® (nilotinib) and Sprycel® (dasatinib), are effective in the treatment of CML, some patients relapse due to the emergence of drug-resistant clones, in which mutations in the SHI domain compromise inhibitor binding. Although Tasigna® and Sprycel® maintain efficacy towards many Gleevec-resistant mutant forms of BCR-ABLl, the mutation in which the threonine-315 residue is replaced by an isoleucine (T315I) remains insensitive to all three drugs and can result in CML patients developing resistance to therapy. Therefore, inhibiting BCR-ABLl mutations, such as T315I, remains an unmet medical need. In addition to CML, BCR-ABLl fusion proteins are causative in a percentage of acute lymphocytic leukemias, and drugs targeting ABL kinase activity also have utility in this indication.Agents targeting the myristoyl binding site (so-called allosteric inhibitors) have potential for the treatment of BCR-ABLl disorders (J. Zhang, F. J. Adrian, W. Jahnke, S. W. Cowan- Jacob, A. G. Li, R. E. Iacob4, T. Sim, J. Powers, C. Dierks, F. Sun, G.-R. Guo, Q. Ding, B. Okram, Y. Choi, A. Wojciechowski, X. Deng, G. Liu, G. Fendrich, A. Strauss, N. Vajpai, S. Grzesiek, T. Tuntland, Y. Liu, B. Bursulaya, M. Azam, P. W. Manley, J. R. Engen, G. Q. Daley, M. Warmuth., N. S. Gray. Targeting BCR-ABL by combining allosteric with ATP -binding-site inhibitors. Nature 2010;463:501-6). To prevent the emergence of drug resistance from ATP inhibitor and/or allosteric inhibitor use, a combination treatment using both types of inhibitor can be developed for the treatment of BCR-ABLl related disorders. In particular, the need exists for small molecules, or combinations thereof, that inhibit the activity of BCR-ABLl and BCR-ABLl mutations via the ATP binding site, the myristoyl binding site or a combination of both sites.Further, inhibitors of ABL 1 kinase activity have the potential to be used as therapies for the treatment of metastatic invasive carcinomas and viral infections such as pox and Ebola viruses.The compounds from the present invention also have the potential to treat or prevent diseases or disorders associated with abnormally activated kinase activity of wild-type ABL1, including non-malignant diseases or disorders, such as CNS diseases in particular neurodegenerative diseases (for example Alzheimer’s, Parkinson’s diseases), motoneuroneuron diseases (amyotophic lateral sclerosis), muscular dystrophies, autoimmune and inflammatory diseases (diabetes and pulmonary fibrosis), viral infections, prion diseases.

Asciminib is an allosteric inhibitor of BCR-ABL kinase in phase III clinical development at Novartis for the treatment of patients with chronic myelogenous leukemia (CML) in chronic phase who have been previously treated with ATP-binding site tyrosine kinase inhibitors. Early clinical trials are also under way in patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) and as first-line threapy of CML.

PATENT

WO2013171639

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013171639&recNum=141&docAn=IB2013053768&queryString=EN_ALL:nmr%20AND%20PA:novartis&maxRec=3644

To illustrate tautomerism with the following specific examples, (R)-N-(4- (chlorodifluoromethoxy)phenyl)-6-(3-hydroxypyrrolidin-l-yl)-5-(lH-pyrazol-5-yl)nicotinamide

(right structure, below) is a tautomer of (R)-N-(4-(chlorodifluoromethoxy)phenyl)-6-(3-hydroxypyrrolidin-l-yl)-5-(lH-pyrazol-3-yl)nicotinamide (left structure, below) and vice versa:

[0045] Where the plural form (e.g. compounds, salts) is used, this includes the singular

Example 9

(R)-N-(4-(Chlorodifluoromethoxy)phenyl)-6-(3-hvdroxypyrrolidin-l-yl)-5-(lH-pyrazol-5- vDnicotinamide

[00365] A mixture of (R)-5-Bromo-N-(4-(chlorodifluoromethoxy)phenyl)-6-(3-hydroxypyrrolidin-l-yl)nicotinamide (Stage 9.2, 100 mg, 0.216 mmol) and 5-(4 ,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- IH-pyrazole (215 mg, 0.663 mmol), Pd(PPh3)2Cl2 (17 mg, 0.024 mmol), Na2C03 (115 mg, 1.081 mmol), DME (917 μί), water (262 μΕ) and EtOH (131 μί) in a MW vial was sealed, evacuated / purged 3 times with argon and subjected to MW irradiation at 125°C for 20 min. The RM was diluted with 2 mL

of DME, stirred with Si-Thiol (Silicycle 1.44 mmol/g, 90 mg, 0.130 mmol) for 3 h. The mixture was centrifuged and the supernatant was filtered through a 0.45 μηι PTFE filter and the solvent was evaporated off under reduced pressure. The crude product was purified by flash

chromatography (RediSep® Silica gel column, 12 g, cyclohexane / EtOAc from 40% to 100% EtOAc) to afford the protected intermediate as a colorless oil. Ethylene diamine (96 μί, 1.428 mmol) and TBAF 1 M in THF (1.428 mL, 1.428 mmol) were then added and the RM was stirred at 80-85°C for 5 days. The solvent was evaporated off under reduced pressure and the residue was dissolved in EtOAc (40 mL), washed 3 times with sat. aq. NaHCC and brine, dried over Na2S04 and The solvent was evaporated off under reduced pressure to give a residue which was purified by preparative SFC (Column DEAP, from 25% to 30% in 6 min) to yield the title compound as a white solid.

[00366] Alternatively, Example 9 was prepared by adding TFA (168 mL, 2182 mmol) to a solution of N-(4-(chlorodifluoromethoxy)phenyl)-6-((R)-3-hydroxypyrrolidin-l-yl)-5-(l-(tetrahydro-2H-pyran-2-yl)-lH-pyrazol-5-yl)nicotinamide (Stage 9.1, 31.3 g, 54.6 mmol) in DCM (600 mL). The mixture was stirred at RT for 2.5 h. The solvent was evaporated off under reduced pressure and the residue was dissolved in EtOAc (1.5 L),washed with a sat. solution of NaHC03 (3 x 500 mL) and brine (500 mL), dried over Na2S04 and the solvent was evaporated off under reduced pressure to give a residue which was suspended in DCM (300 mL), stirred at RT for 15 min, filtered, washed with DCM (200 mL), dried and purified by chromatography (Silica gel, 1 kg, DCM / MeOH 95:5). The residue was dissolved in MeOH (500 mL) and treated with Si-Thiol (Biotage, 5.0 g , 6.5 mmol) for 16 h at 25°C. The resin was filtered off, the solvent was evaporated off under reduced pressure and the residue was crystallized from MeCN to afford the title compound as a white crystalline solid.

[00367] Alternatively, Example 9 was prepared by the dropwise addition of aqueous HC1

(7.7 mL of 6M) to a solution of N-(4-(chlorodifluoromethoxy)phenyl)-6-((R)-3-hydroxypyrrolidin- 1 -yl)-5-( 1 -(tetrahydro-2H-pyran-2-yl)- 1 H-pyrazol-5-yl)nicotinamide (Stage 9.1, 3.8 g, 7.12 mmol) in MeOH (20 mL) and THF (10 mL) with cooling (below 35°C). The mixture was stirred at 22°C for 2 h and then added to cooled (10°C) 1.2 M NaOH (22 mL).

Throughout the addition the temperature was kept below 30°C and pH was kept in the range of 9-10. The RM was then stirred for 30 min at 30°C. The solvent was evaporated off under reduced pressure, until the desired compound precipitated. The precipitate was filtered and dried to give the title compound as a yellow solid.

[00368] Analytical data for Example 9: HPLC (Condition 5) tR = 5.54 min, HPLC Chiral

(CHIRALCEL® OD-H, 250 x 4.6 mm, eluent : n-heptane/EtOH/MeOH (85: 10:5), 1 mL/min, UV 210 nm) tR = 10.17 min, UPLC-MS (condition 3) tR = 0.93 min, m/z = 450.3 [M+H]+, m/z = 494.1 [M+formic acid-H]XH-NMR (400 MHz, DMSO-d6) δ ppm 1.65 – 1.76 (m, 1 H) 1.76 – 1.87 (m, 1 H) 2.93 (d, J=l 1.73 Hz, 1 H) 3.19 – 3.29 (m, 2 H) 3.35 – 3.51 (m, 1 H) 4.10 – 4.25 (m, 1 H) 4.89 (br. s, 1 H) 6.41 (br. s, 1 H) 7.33 (d, J=8.50 Hz, 2 H) 7.57/7.83 (br. s, 1 H) 7.90 (d, J=8.50 Hz, 2 H) 8.07 (br. s, 1 H) 8.77 (br. s, 1 H) 10.23 (s, 1 H) 12.97/13.15 (br. s, 1 H).

[00369] Stage 9.1 : N-(4-(Chlorodifluoromethoxy)phenyl)-6-((R)-3-hydroxypyrrolidin- 1 -yl)-5-( 1 -(tetrahydro-2H-pyran-2- l)- 1 H-pyrazol-5-yl)nicotinamide

[00370] l-(Tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (29.6 g, 102 mmol), K3P04 (51.6 g, 236 mmol) and Pd(PPh3)4 (4.55 g, 3.93 mmol) were added to a suspension of (R)-5-bromo-N-(4-(chlorodifluoromethoxy)phenyl)-6-(3-hydroxypyrrolidin-l-yl)nicotinamide (Stage 9.2, 36.4 g, 79 mmol) in toluene (360 mL) under an argon atmosphere and the mixture was stirred at 110°C for 4 h. The RM was poured into brine (500 mL) and extracted with EtOAc (2 x 1 L). The combined extracts were washed with brine (500 mL), dried over Na2S04, and the solvent was evaporated off under reduced pressure to give a residue which was purified by chromatography (Silica gel column, 1.5 kg, DCM / MeOH 95:5) to afford a dark yellow foam, that was dissolved in MeOH / DCM (1 L of 3: l) and treated with Si-Thiol (Biotage, 35 g , 45.5 mmol) for 17 h at 30°C. The resin was filtered off, and solvent was evaporated off under reduced pressure, until the desired compound crystallized. The product was filtered washed with MeOH and dried to afford the title compound.

[00371] Alternatively, Stage 9.1 was prepared by adding 4-(chlorodifluoromethoxy)aniline

(16.6 g, 84.9 mmol), NMM (21.7 g, 212.1 mmol), hydroxybenzotriazole hydrate (HOBt H20, 11.9 g, 77.77 mmol) and l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDCIHCl, 20.9 g, 109.0 mmol) to a solution of 6-((R)-3-hydroxypyrrolidin-l-yl)-5-(l-(tetrahydro-2H-pyran-2-yl)-lH-pyrazol-5-yl)nicotinic acid (Stage 9.4, 29.83 g, 70.7 mmol) in THF (271 mL). The mixture was stirred for 1.5 h at 25°C and then at 65°C for 16 h. After cooling the RM to 35 °C, further EDCIHCl (13.3 g, 69.4 mmol) was added and the RM was stirred for 1.5 h at 35°C then again at 65°C for 16 h. After cooling the RM to 35°C, water (150 mL) was added, the THF was removed under reduced pressure, EtOAc (180 mL) was added and the mixture was stirred for at 35 °C fori h. The two layers were separated and the aq. phase was then extracted with EtOAc (60 mL). The combined organic layers were washed with water (90 mL), brine (90 mL). The solvent was evaporated off under reduced pressure to give a brown solid which was purified by column chromatography (Silica gel, DCM / MeOH 40: 1 to 20: 1) to afford the title compound as a yellow solid.

[00372] Analytical data for Stage 9.1: HPLC (Condition 5) tR = 6.12 min, UPLC-MS

(Condition 3) tR = 1.06 min, m/z = 533.2 [M+H]+XH-NMR (400 MHz, DMSO-d6) δ ppm 1.36 -2.02 (m, 7 H) 2.23 – 2.38 (m, 1 H) 3.08 – 3.29 (m, 2 H) 3.32 – 3.52 (m, 2 H) 3.73 – 3.93 (m, 1 H) 4.13 – 4.25 (m, 1 H) 4.80 – 4.90 (m, 1 H) 4.95 – 5.17 (m, 1 H) 6.33 – 6.50 (m, 1 H) 7.33 (d, J=8.99 Hz, 2 H) 7.61 (d, J=1.56 Hz, 1 H) 7.86 (d, J=8.99 Hz, 2 H) 7.97 – 8.11 (m, 1 H) 8.82 (s, 1 H) 10.13 – 10.25 (m, 1 H).

[00373] Stage 9.2: (R)-5-Bromo-N-(4-(chlorodifluoromethoxy)phenyl)-6-(3-hydroxypyrrolidin- 1 -yl)nicotinamide

[00374] (R)-Pyrrolidin-3-ol (9.55 g, 109.6 mmol) and DIPEA (35.1 ml, 201.3 mmol) were added to a suspension of 5-bromo-6-chloro-N-(4-(chlorodifluoromethoxy)phenyl)nicotinamide (Stage 9.3, 37.7 g, 91.5 mmol) in iPrOH (65 mL) and stirred at 140°C for 1 h. EtOAc (700 mL) was added and the solution was washed IN HC1 (2 x 200 mL), sat. NaHCC (200 mL) and brine (2 x 200 mL), dried over Na2S04, and the solution was concentrated under reduced pressure until crystallization commenced. n-Heptane (1 L) were added and the mixture was stirred at RT for 30 min, filtered and washed with ΪΡΓ20 (500 mL) to afford the title compound as a white crystalline solid. HPLC (Condition 5) tR = 6.68 min, UPLC-MS (Condition 3) tR = 1.10 min, m/z =

462.2/464.2 [M+H]+XH-NMR (400 MHz, DMSO-d6) δ ppm 1.78 – 2.01 (m, 2 H) 3.55 (d, J=l 1.34 Hz, 1 H) 3.66 – 3.75 (m, 1 H) 3.79 – 3.93 (m, 2 H) 4.34 (br. s, 1 H) 4.98 (d, =3.13 Hz, 1 H) 7.32 (d, J=8.99 Hz, 2 H) 7.84 (d, J=8.99 Hz, 2 H) 8.33 (d, J=1.96 Hz, 1 H) 8.66 (d, J=1.96 Hz, 1 H) 10.21 (s, 1 H).

[00375] Stage 9.3: 5-Bromo-6-chloro-N- 4-(chlorodifluoromethoxy)phenyl)nicotinamide

[00376] DMF (2.55 mL, 33.0 mmol) and SOCl2 (24.08 ml, 330 mmol) were added to a suspension of 5-bromo-6-chloro-nicotinic acid (26 g, 110 mmol) in toluene (220 mL) and the RM was stirred at 80°C for 1 h. The solvent was evaporated off under reduced pressure and the residue was dissolved in THF (220 mL) and cooled to -16°C. DIPEA (38.4 mL, 220 mmol) was added, followed by dropwise addition of a solution of 4-(chlorodifluoromethoxy)aniline (22.35 g, 115 mmol) in THF (220 mL) over 15 min. The suspension was stirred for 1 h at RT. The solvent was evaporated off under reduced pressure and the residue was dissolved in TBME (700 mL), washed with IN HC1 (2 x 200 mL), sat. NaHC03 (200 mL) and brine (2 x 200 mL), dried over Na2S04, and the solvent was evaporated off under reduced pressure to give the product which was crystallized from EtOAc – n-heptane to afford the title compound as a white crystalline solid. HPLC (Condition 5) tR = 7.77 min, UPLC-MS (Condition 3) tR = 1.24 min, m/z =

409.1/411.1/413.1 [M+H]+XH-NMR (400 MHz, DMSO-d6) δ ppm 7.38 (d, =8.99 Hz, 2 H) 7.85 (d, =8.99 Hz, 2 H) 8.72 (br. s, 1 H) 8.92 (br. s, 1 H) 10.68 (s, 1 H).

[00377] Stage 9.4: 6-((R)-3-Hydroxypyrrolidin-l-yl)-5-(l-(tetrahydro-2H-pyran-2-yl)-lH-pyrazol-5-yl)nicotinic acid

[00378] Aq. NaOH (180 niL of 2.6 M) was added to a solution of methyl 6-((R)-3-hydroxypyrrolidin- 1 -yl)-5-(l -(tetrahydro-2H-pyran-2-yl)- 1 H-pyrazol-5-yl)nicotinate (Stage 9.5, 11 lg, 299 mmol) in MeOH (270 mL) and the RM was stirred at RT for 14 h. The MeOH was evaporated off under reduced pressure and the aq. residue was treated with brine (90 mL), extracted with MeTHF twice (540 mL + 360 mL) and the combined organic layers were washed with water (90 mL). MeTHF was added to the combined aq. layers, the biphasic mixture was cooled to 0 °C and acidified (pH = 4-4.5) with aq. HC1 solution (18%) and extracted with

MeTHF. The combined organic extracts were washed with brine and the solvent was evaporated off under reduced pressure to give a residue which was recrystallized from a EtOAc / TBME (1 : 1) to afford the title compound as a white solid. HPLC (Condition 7) tR = 4.74 min, LC-MS

(Condition 8) tR = 3.37 min, m/z = 359.0 [M+H]+XH-NMR (400 MHz, DMSO-d6) δ ppm 1.44 (br. s, 2 H), 1.51 (d, J=11.54 Hz, 2 H), 1.64 – 1.86 (m, 4 H), 1.90 (br. s, 1 H), 2.31 (d, J=9.29 Hz, 1 H), 2.77 (br. s, 1 H), 3.10 (br. s, 1 H), 3.21 (d, J=8.78 Hz, 2 H), 3.27 – 3.51 (m, 4 H), 3.87 (d, J=11.54 Hz, 1 H), 4.16 (br. s, 1 H), 4.75 – 4.93 (m, 1 H), 5.04 (br. s, 1 H), 6.35 (d, J=17.32 Hz, 1 H), 7.51 – 7.64 (m, 1 H), 7.64 – 7.82 (m, 1 H), 8.67 (d, J=2.26 Hz, 1 H), 12.58 (br. s, 1 H).

[00379] Stage 9.5: Methyl 6-((R)-3-hydroxypyrrolidin-l-yl)-5-(l-(tetrahydro-2H-pyran-2-yl)- 1 H-pyrazol-5-yl)nicotinate

[00380] A mixture of (R)-methyl 5-bromo-6-(3-hydroxypyrrolidin-l-yl)nicotinate (Stage

9.6, 90 g, 299 mmol), l-(tetrahydro-2H-pyran-2-yl)-lH-pyrazole-5-boronic acid pinacol ester (103.9 g, 373.6 mmol), K3P04 (126.9 g, 597.7 mmol), Pd(PPh3)2Cl2 (6.29 g, 8.97 mmol) in toluene (900 mL) was stirred at 92°C and for 16 h. After cooling the mixture to RT, the solution was washed with water (450 mL), 5% NaHCC solution (430 mL) and the solvent was evaporated off under reduced pressure to give a residue which was used without further purifications in the next step. HPLC (Condition 7) tR = 6.929 min, LC-MS (Condition 8) tR = 4.30 min, m/z = 373.0 [M+H ; XH-NMR (400 MHz, DMSO-d6) δ ppm 1.19 – 1.28 (m, 1 H), 1.35 – 1.63 (m, 4 H), 1.63 -1.86 (m, 3 H), 1.89 (br. s, 1 H), 2.12 – 2.39 (m, 1 H), 3.11 (br. s, 1 H), 3.18 – 3.48 (m, 4 H), 3.78 (s, 4 H), 3.88 (d, J=11.54 Hz, 1 H), 4.08 – 4.24 (m, 1 H), 4.86 (dd, J=18.20, 2.89 Hz, 1 H), 5.02 (d, J=8.28 Hz, 1 H), 6.39 (br. s, 1 H), 7.58 (d, J=1.25 Hz, 1 H), 7.78 (br. s, 1 H), 8.69 (t, J=2.01 Hz, 1 H).

[00381] Stage 9.6: (R)-methyl 5-bromo-6-(3-hydroxypyrrolidin-l-yl)nicotinate

[00382] DIPEA (105.3 g, 142.2 mL, 814.4 mmol) was added to a solution of methyl-5-bromo-6-chroronicotinate (85 g, 339.5 mmol) and (R)-pyrrolidin-3-ol (54.2 g, 441.2 mmol) in isopropyl acetate and the RM was stirred at 70°C for 14 h . The solvent was evaporated off under reduced pressure to give a the residue which was dissolved in toluene (850 mL), washed with water (127 mL) and brine (127 mL)and concentrated under reduced pressure until precipitation commenced. n-Heptane (340 mL) was slowly added to the stirred mixture at 22 °C, which was then cooled to 0 °C and the product was filtered, washed with a toluene / n-heptane mixture

(1 : 1.5) and dried to give the title compound as a yellow solid. HPLC (Condition 7) tR = 8.54 min, LC-MS (Condition 8) tR = 4.62 min, m/z = 300.9/302.9 [M+H]+XH-NMR (400 MHz, DMSO-d6) δ ρριη 1.77 – 1.99 (m, 2 H), 3.57 (d, J=11.54 Hz, 1 H), 3.72 (ddd, J=l 1.11, 7.97, 3.26 Hz, 1 H), 3.78 (s, 3 H), 3.81 -3.90 (m, 2 H), 4.26 – 4.39 (m, 1 H), 4.99 (br. s, 1 H), 8.11 (d, J=2.01 Hz, 1 H), 8.56 (d, J=1.76 Hz, 1 H).

PAPER

  • By Wylie, Andrew A.; Schoepfer, Joseph; Jahnke, Wolfgang; Cowan-Jacob, Sandra W.; Loo, Alice; Furet, Pascal; Marzinzik, Andreas L.; Pelle, Xavier; Donovan, Jerry; Zhu, Wenjing; et al
  • From Nature (London, United Kingdom) (2017), 543(7647), 733-737.

By Wylie, Andrew A. et alFrom Nature (London, United Kingdom), 543(7647), 733-737; 2017

PAPER

  • By Molica, Matteo; Massaro, Fulvio; Breccia, Massimo
  • From Expert Opinion on Pharmacotherapy (2017), 18(1), 57-65.

PATENT

US 20170216289

PAPER

  • By El Rashedy, Ahmed A.; Olotu, Fisayo A.; Soliman, Mahmoud E. S.
  • From Chemistry & Biodiversity (2018), 15(3), n/a.
wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

 
Patent ID

 

Patent Title

 

Submitted Date

 

Granted Date

 

US2016108123 ANTIBODY MOLECULES TO PD-L1 AND USES THEREOF
2015-10-13
2016-04-21
US2014343086 COMPOUNDS AND COMPOSITIONS FOR INHIBITING THE ACTIVITY OF ABL1, ABL2 AND BCR-ABL1
2014-07-31
2014-11-20
US8829195 Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
2013-05-13
2014-09-09
Asciminib
Asciminib.svg
Clinical data
Trade names Scemblix
Other names ABL001
Routes of
administration
By mouth
Drug class Tyrosine kinase inhibitor
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
PDB ligand
Chemical and physical data
Formula C20H18ClF2N5O3
Molar mass 449.84 g·mol−1
3D model (JSmol)

References

  1. Jump up to:a b c d “Scemblix- asciminib tablet, film coated”DailyMed. Retrieved 4 November 2021.
  2. Jump up to:a b c “FDA approves asciminib for Philadelphia chromosome-positive chronic myeloid leukemia”U.S. Food and Drug Administration (FDA) (Press release). 29 October 2021. Retrieved 4 November 2021. Public Domain This article incorporates text from this source, which is in the public domain.
  3. ^ Breccia M, Colafigli G, Scalzulli E, Martelli M (August 2021). “Asciminib: an investigational agent for the treatment of chronic myeloid leukemia”. Expert Opinion on Investigational Drugs30 (8): 803–811. doi:10.1080/13543784.2021.1941863PMID 34130563.
  4. ^ “Scemblix: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 29 October 2021.
  5. ^ “FDA approves Novartis Scemblix (asciminib), with novel mechanism of action for the treatment of chronic myeloid leukemia”Novartis (Press release). Retrieved 29 October 2021.
  6. ^ “Asciminib Orphan Drug Designations and Approvals”U.S. Food and Drug Administration (FDA). 27 February 2017. Retrieved 29 October 2021.
  7. ^ “Novartis receives FDA Breakthrough Therapy designations for investigational STAMP inhibitor asciminib (ABL001) in chronic myeloid leukemia”Novartis (Press release). 8 February 2020. Retrieved 29 October 2021.

External links

  • “Asciminib”Drug Information Portal. U.S. National Library of Medicine.
  • Clinical trial number NCT02081378 for “A Phase I Study of Oral ABL001 in Patients With CML or Ph+ ALL” at ClinicalTrials.gov
  • Clinical trial number NCT03106779 for “Study of Efficacy of CML-CP Patients Treated With ABL001 Versus Bosutinib, Previously Treated With 2 or More TKIs” at ClinicalTrials.gov

 

////////////////ABL001, Asciminib, ABL 001, ABL-001, PHASE 3, Chronic Myeloid Leukemia,  NOVARTIS

 O=C(NC1=CC=C(OC(F)(Cl)F)C=C1)C2=CN=C(N3C[C@H](O)CC3)C(C4=CC=NN4)=C2

wdt-7

NEW DRUG APPROVALS

one time

$10.00

Atogepant, атогепант , أتوجيبانت , 阿托吉泮 ,


imgChemSpider 2D Image | atogepant | C29H23F6N5O3Atogepant.pngImage result for AtogepantImage result for AtogepantFigure imgf000011_0002

Atogepant

  • Molecular FormulaC29H23F6N5O3
  • Average mass603.515 Da

AGN 241689; MK 8031

(3S)-N-[(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]-2-oxospiro[1H-pyrrolo[2,3-b]pyridine-3,6′-5,7-dihydrocyclopenta[b]pyridine]-3′-carboxamide

Spiro[6H-cyclopenta[b]pyridine-6,3′-[3H]pyrrolo[2,3-b]pyridine]-3-carboxamide, 1′,2′,5,7-tetrahydro-N-[(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)-3-piperidinyl]-2′-ox o-, (6S)-[ACD/Index Name]
атогепант [Russian] [INN]
أتوجيبانت [Arabic] [INN]
阿托吉泮 [Chinese] [INN]
(6S)-N-[(3S,5S,6R)-6-Methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)-3-piperidinyl]-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxamide [ACD/IUPAC Name]
10510
1374248-81-3 [RN]
7CRV8RR151
Atogepant; UNII-7CRV8RR151; 7CRV8RR151; AGN-241689; MK-8031; 1374248-81-3

 Spiro(6H-cyclopenta(b)pyridine-6,3′-(3H)pyrrolo(2,3-b)pyridine)-3-carboxamide, 1′,2′,5,7-tetrahydro-N-((3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)-3-piperidinyl)-2′-oxo-, (3’S)-

Oral prevention of episodic migraine in adult patients.
 
Innovator – Allergan Phase 3
Allergan announced positive results from Phase 2b/3 clinical trial in Jun 2018 evaluating the efficacy, safety, and tolerability of orally administered Atogepant,  
Being CGRP antagonist, is more efficacious than any other preventative treatment on the market
  • Originator Merck AG
  • Developer Allergan
  • Class Antimigraines; Monoclonal antibodies; Piperidines; Pyridines; Pyrroles; Small molecules; Spiro compounds
  • Mechanism of Action Calcitonin gene-related peptide antagonists

Highest Development Phases

  • Phase II/III Migraine

Most Recent Events

  • 11 Jun 2018 Efficacy and adverse events data from a phase IIb/III trial in Migraine released by Allergan
  • 23 Apr 2018 Allergan completes a phase II/III trial for Migraine (Prevention) in USA (PO) (NCT02848326)
  • 14 Sep 2017 Chemical structure information added

UPDATE………..

FDA APPROVED 2021, 28/9/21, Qulipta

The product was discovered by Merck and, in August 2015, it was licensed to Allergan for worldwide development and marketing.

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

  •  

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter 

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

Synthesis

US20160130273

Figure US20160130273A1-20160512-C00031

 Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000061_0002
 
PATENT
WO 2007133491
PATENT
PRODUCT PATENT
WO 2012064910

INTERMEDIATE 1

Figure imgf000041_0002
Figure imgf000042_0001

carboxylic acid

The title compound can be prepared by either Method I or Method II as described below.

Method I:

Step A: (6S)-3-Iodo-5 J-dihyc ospiro cyclopentar¾1pyrid e-6 ‘-py

one

A solution of sodium nitrite (36.1 g, 523 mmol) in water (20 mL) was added dropwise over 5 min to a solution of (6S -3-amino-5,7-dihydros iro[cyclopenta[ί)]pyridi e-6,3,– pyrrolo[2,3-0]pyridin]-2′(rH)-one (prepared according to the procedures described in

WO2008/020902, 66.0 g, 262 mmol) and -toluenesulfonic acid (149 g, 785 mmol) in acetonitrile (650 mL) at 23 °C. After stirring for 30 min, a solution of potassium iodide (109 g, 654 mmol) in water (20 mL) was added over 5 min. The resulting mixture was stirred at 23 °C for 40 min, then diluted with water (1 L) and basified by the addition of solid NaOH (33.0 g, 824 mmol) with stirring. Iodine by-product was reduced by the addition of 10% aqueous sodium thio sulfate solution and stirring for an additional 30 min. The solids were collected by filtration, washed with water, and dried under nitrogen atmosphere to give the title compound, which was used without further purification. MS: mlz = 363.9 (M + 1).

Step B: Methyl (65V2′-oxo-lΛ2 5J-tetrahydrospiroicvclopenta[6]p ridine-6.3′-pyrlΌlo[2. – 6]py ridine] – 3 -car boxy late

A solution of (65)-3-iodo~5 ,7-dihydrospiro[cyclopenta[&]pyridine-6,3′- pyrrolo[2,3-&]pyridin]-2′(rH)-one (51.0 g, 140 mmol), sodium acetate (23.0 g, 281 mmol) and dichloro l,l’~bis(diphenylphosphino)ferrocene palladium(II) dichloromethane adduct (2.9 g, 3.5 mmol) in MeOH (560 mL) was pressurized to 120 psi of CO at 23 °C and then heated at 80 °C for 12 h with stirring. The reaction mixture was diluted with water (1 L), and the precipitate collected by filtration, washed with water, and dried under nitrogen atmosphere to give the title compound, which was used without further purification. MS: mlz = 296.1 (M + 1).

Figure imgf000042_0002

3 -carboxylic acid

A mixture of methyl (6S)-2′-oxo-r,2′,5,7-tetrahydrospiro[cyclopenta[i)]pyridine- 6,3′-pyrrolo[2,3-&]pyridine]-3-carboxylate (30.0 g, 102 mmol) and aqueous 6 N sodium hydroxide solution (50.8 mL, 305 mmol) in MeOH (920 mL) was heated at reflux for 1 h. The mixture was allowed to cool to 23 °C before it was acidified to pH ~6 with aqueous 1 N hydrochloric acid solution, resulting in a black precipitate which was removed by filtration. The filtrate was concentrated under reduced pressure to a volume of ~100 mL and then partitioned between water (500 mL) and 2-methyltetrahydrofuran (2- eTHF, 250 mL). The aqueous layer was extracted with 2-MeTHF (5 χ 250 mL), and the combined organic layers were dried over sodium sulfate and concentrated to provide the title compound. MS: mlz ~ 282.0 (M + 1).

Method II:

Step A: Dimethyl 5-bromopyridine-2,3-dicarboxylate

Concentrated sulfuric acid (1 L, 18.7 mol) was added slowly over 10 min to a . suspension of pyridine-2,3-dicarboxylic acid (5.00 kg, 29.9 mol) in methanol (50 L), dissolving the suspension. The resulting mixture was heated at reflux for 48 h then cooled to 40 °C.

Bromine (8.0 kg, 50 mol) was added slowly over 2 h in 1-kg portions, keeping the temperature below 55 °C. The reaction mixture was then heated at 55 °C for 24 h, cooled to 50 °C and additional Br2 (4.0 kg, 25 mol) was added slowly over 1 h in 1-kg portions, keeping temperature below 55 °C. The reaction mixture was heated at 55 °C for 24 h, concentrated to a minimum volume (internal temp -30 °C, solution may occasionally foam), then diluted with isopropyl acetate (50 L) and washed with a saturated aqueous sodium sulfite solution (3 x 20 L) (final extract is ~pH 8) followed by water (20 L). The organic layer was concentrated to

approximately 15 L then diluted with heptane (40 L). The resulting slurry was stirred for 24 h at 23 °C. The solids were filtered, washed with heptane (10 L), and dried to give the title compound. Step B: (5-Bromopyridine-23-diyl)dimcthanol

Sodium borohydride (15.9g, 420 mmol) was added portionwise over 30 min to a solution of dimethyl 5-bromopyridine-2,3-dicarboxylate (20 g, 73 mmol) in ethanol (460 mL) precooled to 0 °C. A solution of calcium chloride (23.3 g, 209 mmol) in 150 mL was added slowly at 0 °C, and the reaction mixture was warmed to 23 °C and stirred overnight. Excess sodium borohydride was quenched by slow addition of aqueous 2 N HCl solution (230 mL, 460 mmol), followed by a stirring at 23 °C for 2 h. The mixture was concentrated to dryness.

Saturated aqueous sodium bicarbonate solution was added to the residue until a pH of approximately 7 was reached. The aqueous mixture was extracted with 2-methyltetrahydrofuran (4 x 200 mL). The combined organic layers were dried over sodium sulfate then treated with a solution of 4 N HC1 in dioxane (25 mL, 100 mmol). The resulting solid was filtered, washed with 2-methyltetrahydrofuran, and dried to give the title compound as a hydrochloride salt. MS: m!z = 218.1 (M + 1). Step C: (5-Bromopyridine-2,3-diyI)dimethanediyl dimethanesulfonate

A slurry of (5-bromopyridine-2,3-diyl)dimethanol hydrochloride (12.9g, 59.2 mmol) in tetrahydrofuran (400 mL) at 0 °C was treated with triethylamine (37.1 mL, 266 mmol). To the resulting mixture was added portionwise methanesulfonic anhydride (30.9 g, 177 mmol), keeping temperature below 5 °C. The reaction mixture was stirred at 0 °C for 1 h, then partitioned between saturated aqueous sodium bicarbonate solution (500 mL) and ethyl acetate (500 mL). The organic layer was washed saturated aqueous sodium bicarbonate solution, dried over magnesium sulfate, and concentrated to give the title compound. MS: m/z – 376.0 (M + 1).

Step D: 3-Bromo-r-{[2-(trimethylsilyl)ethoxy]methyl}-5,7- dihyjirpspiro [cyclop

(5-Bromopyridine-2,3-diyI)dimethanediyl dimethanesulfonate (17.0 g, 45.4 mmol) was added to a mixture of l-{[2-(trimetliylsilyl)ethoxy]methyl}-l}3-dihydro-2H- pyrrolo[2,3-&]pyridin-2-one (prepared according to the procedures described in

WO2008/020902, 14.0 g, 53.0 mmol) and cesium carbonate (49.0 g, 150 mmol) in ethanol (500 mL) 23 °C, and the resulting mixture was stirred for 20 h. The reaction mixture was

concentrated then partitioned between ethyl acetate (500 mL) and water (500 mL). The organic layer was dried over magnesium sulfate and concentrated. The residue was purified via silica gel chromatography (heptane initially, grading to 100% EtOAc) to give the title compound. MS: m/z = 448.1 (M + 1).

Step E: Methyl (6<Sf)-2′-oxo-r-{r2-(trimethylsilyl ethoxylmethyli-r,2′,5 J- tetrahydrospiro [cy clopenta[6] pyridine-6 ,3 ‘-pyrrolo [2, 3 -b]py ridinel -3 -carboxy late

A mixture of 3-bromo-r-{[2-(trimethylsilyl)ethoxy]methyl}-5,7- dihydrospiro[cyclopenta[¾]pyridine-6,3′-pyrrolo[2,3-¾pyridin]-2′(rH)-one (22.0 g, 49.3 mmol), PdCl2(dppf)»CH Cl2 (2.012g, 2.46 mmol), and sodium acetate (8.1g, 99 mmol) in in methanol (150 mL) was pressurized to 300 psi of carbon monoxide and then heated at 85 °C for 72 h. The reaction mixture was allowed to cool then concentrated. The residue was purified via silica gel chromatography (heptane initially, grading to 100% EtOAc) to give the title compound as a racemic mixture. MS: m/z – 426.1 (M +1). Resolution of the enantiomers by supercritical fluid chromatography (SFC) using a ChiralPak AD-H column and eluting with 40% ethanol in C02 (0.05% diethylamine as modifier) provided the title compound as the second enantiomer to elute.

Figure imgf000045_0001

A solution of methyl (65)-2′-oxo- -{[2-(trimethylsilyl)ethoxy]methyl}-r!2′f5,7- tetrahydrospiro[cyclopenta[&]pyridine-6,3′-pyrrolo[2,3-&]pyridine]-3-carboxylate (238 g, 559 mmol) in methanol (2 L) was saturated with HCI gas, allowing temperature to increase to 55 °C. The reaction mixture was cooled to 23 °C, stirred for 20 h, then concentrated. Aqueous 10 N sodium hydroxide (400 mL, 4 mol) was added to a solution of the residue in methanol (2 L), and the resulting mixture was heated at reflux for 2 h. The solution was cooled to 23 °C and the pH was adjusted to 3 with concentrated HCI. The resulting solid was filtered, washed with water then heptane, and dried to give the title compound. MS: m!z = 282.2 (M + 1).

INTERMEDIATE 15

Figure imgf000066_0001
Figure imgf000066_0002

hydrochloride

Step A: (5SSR & 5j?,6y)-6-Methvi-l-r2.2.2-trifluoroethvn-5-(2,3.6-trifluorophenvnpiperidin-2- one

Essentially following the procedures described in Intermediate 14, but using 2,3,6-trifluorophenylboronic acid in place of 2,3,5-trifluorophenylboronic acid, the title compound was obtained. MS: m/z = 326.0 (M + 1).

Step B: GS.5S.6R & 3i?,5J?.6 ‘ -3-Azido-6-methyl-i-r2.2.2 rifluoroethyl)-5-(2.3.6- trifluorophenyl)piperidin-2-one

To a stirred solution of lithium 6w(trimethylsilyl)amide (1.0 M in THF, 4.80 mL,

4.80 mmol) in THF (20 mL) at -78 °C was added a cold (-78 °C) solution of (5S,6R & 5i?,6,S)-6- methyl-l-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one (1.30 g, 4,00 mmol) in THF (10 mL) dropwise, keeping the internal temperature of the reaction mixture below -65 °C. The resulting mixture was stirred at -78 °C for 30 min, then a cold (-78 °C) solution of 2,4,6- triisopropylbenzenesulfonyl azide (Harmon et l. (1973) J Org. Chem. 38, 11-16) (1.61 g, 5.20 mmol) in THF (10 mL) was added dropwise, keeping the internal temperature of the reaction mixture below -65 °C. The reaction mixture was stirred at -78 °C for 30 min, then AcOH (1.05 mL, 18.4 mmol) was added. The resulting mixture was allowed to warm slowly to ambient temperature and was poured into saturated aqueous sodium bicarbonate (50 mL) and the mixture was extracted with EtOAc (2 χ 75 mL). The combined organic layers were washed with brine, then dried over sodium sulfate, filtered, and concentrated to dryness in vacuo. The crude product was purified by silica gel chromatography, eluting with a gradient of hexanes:EtOAc – 100:0 to 20:80, to give the diastereomeric azide products (3R,5Sf6R & 3S, ;5i?,65)-3-azido-6- methyl-l-(2,2,2-trifluoroethyl)-5-(2f3,5-trifluorophenyl)piperidin-2-one, which eluted second, and the title compound, which eluted first. MS: mlz = 367.1 (M+ 1).

Step C: ferf-Butyl [(3&5^6^ν6^Φν1-2-οχο-1-(2.2,2-ΐπΑηοΓθ£υΐν1 -5-ί2.3,6- trifluorophenyl)piperidin-3-yl|carbamate

To a solution of ( S,5S,6R & 3JR,5if,6S)-3-azido-6-methyl-l-(2,2,2- trifiuoroethyl)-5-(2,3,5-trifluorophenyl)piperidin-2-one (280 mg, 0.764 mmol) and di-tert-butyl dicarbonate (217 mg, 0.994 mmol) in EtOH (5 mL) was added 10% palladium on carbon (25 mg, 0.024 mmol) and the resulting mixture was stirred vigorously under an atmosphere of hydrogen (ca. 1 atm) for 1 h. The reaction mixture was filtered through a pad of Celite® washing with EtOH, and the filtrate was concentrated in vacuo to give a crude solid. The crude product was purified by silica gel chromatography, eluting with a gradient of hexanes:EtOAc – 100:0 to 30:70, to give the racemic title compound. Separation of the enantiomers was achieved by SFC on a ChiralTech IC column, eluting with C02:MeOH:CH CN – 90:6.6:3.3, to give tert- butyl [(3i?,5i?,65)-6-methyl-2-oxo-l-(2J2,2-trifluoroemyl)-5-(2,3J6-tri¾orophenyl)piperidin-3- yl]carbamate as the first major peak, and fert-butyl [(3Sf5S,6R)-6-methyl-2-oxo-l -(2,2,2- trifluoroethyl)-5-(2,3,6-trifiuorophenyl)piperidin-3-yl]carbamate, the title compound, as the second major peak. MS: mlz = 463.2 (M + Na).

Step D: (3&5^6i?)-3-Amino-6-methyi-l-(2,2.2-trifluoroethyl)-5-(2,3,6- trifluorophenyl)piperidin-2-one hydrochloride

A solution of tert-butyl [(35′,55′,6ii)-6-methyl-2-oxo-l-(2J2,2-trifluoroethyl)-5-

(2s3,6-trifluorophenyl)piperidin-3-yl]carbamate (122 mg, 0.277 mmol) in EtOAc (10 mL) was saturated with HCl (g) and aged for 30 min. The resulting mixture was concentrated in vacuo to give the title compound. MS: mlz = 341.1 (M + 1); lH NM (500 MHz, CD3OD) δ 7.33 (qd, 1H, J- 9.3, 4.9 Hz), 7.05 (tdd, 1H, J= 9.8, 3.7, 2.2 Hz), 4.78 (dq, 1H, J= 15.4, 9.3 Hz), 4.22 (dd, 1H, J = 12.2, 6.6 Hz ), 4.06 (ddd, 1H, J- 13.3, 4.5, 2.7 Hz ), 3.97 (m, 1H), 3.73 (dq, 1H, J = 15.4, 8.8 Hz), 2.91 (qt, 1H, J- 12.7, 3.1 Hz), 2.36 (ddd, 1H, J= 12.7, 6.4, 2.0 Hz), 1.22 (d, 3H, J = 6.6 Hz).

EXAMPLE 4

Figure imgf000075_0001

f6SyN-[f3£5£6iO-6-Methyl-2-QXO-i-(2,2,,2-trifl^yl]-2′-oxo-l\2 5J~tetrahydrospiro[cyciopen^

carboxamide dihvdrochloride

To a stirred mixture of (6>$)-2′-οχο-Γ,2′,5,7- tetrahydrospirotcyclopenta[6]pyridine-6,3′-pyrroio[2,3-6]pyridine]-3-carboxylic acid (described in Intermediate 1) (264 mg, 0.939 mmol), (35′,5S’36J?)-3-amino-6-methyl-l-(2,2,2-trifluoroethyl)- 5-(2f3s6-trifluorophenyl)piperidin-2-one hydrochloride (described in Intermediate 15) (295 mg, 0.782 mmol), HOBT (144 mg, 0.939 mmol), and EDC (180 mg, 0.939 mmol) in DMF (8 mL) was added 7V,N-diisopropylethylamine (0.34 mL, 1.96 mmol), and the resulting mixture was stirred at ambient temperature for 3 h. The reaction mixture was then poured into saturated aqueous sodium bicarbonate (30 mL) and extracted with EtOAc (2 χ 40 mL). The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by silica gel chromatography, eluting with a gradient of

CH2Cl2:MeOH:NH40H – 100:0:0 to 90:10:0.1, to give the product, which was treated with HC1 in EtOAc at 0 °C to afford the title compound. HRMS: m/z = 604.1783 (M + 1), calculated m/z = 604.1778 for C29H24F6N5O3. iH NMR (500 MHz, CD3OD) δ 9.09 (s, 1H), 8.69 (s, 1H), 8.18 (dd, 1H, J = 5.9, 1.5 Hz), 7.89 (dd5 1H, J= 7.3, 1.5 Hz), 7.30 (m, 1H), 7.23 (dd, 1H, J= 7.3, 5.9 Hz), 7.03 (m, 1H), 4.78 (m, 1H), 4.61 (dd, 1H, J = 11.5, 6.6 Hz), 4.05 (dd, 1H, J= 13.8, 2.8 Hz), 3.96 (m, 1H), 3.84 (d, 1H, J= 18.6 Hz), 3.76 (d, 1H, J = 18.6 Hz), 3.73 (d, 1H, J= 17.3 Hz), (m, 1H), 3.61 (d, 1H, J = 17.3 Hz), 3.22 (m, 1H), 2.38 (m, 1H), 1.34 (d, 3H, J= 6.6 Hz).

 
POLYMORPHS
US 20160130273
Monohydrate, trihydrate, and carboxamide L-tartaric acid cocrystal;
 
  • Schemes 1 to 15 described below.
  • [0122]
    Scheme 1 illustrates a route to 3-aminopiperidinone intermediates of type 1.5 which may be used to prepare compounds of the present invention. Aryl acetone 1.1 can be alkylated using the iodoalanine derivative 1.2 under basic conditions to provide keto ester 1.3.
  • [0123]
    Reductive amination followed by cyclization and epimerization provides primarily cis-substituted lactam 1.4 as a racemic mixture. Chiral resolution using normal-phase liquid chromatography, for example, and removal of the Boc protecting group with HCl in EtOAc furnishes 3-aminopiperidinone 1.5 as a hydrochloride salt.
  • [0000]
    Figure US20160130273A1-20160512-C00020
  • [0124]
    An alternative sequence to 3-aminopiperidinone intermediates of type 1.5 is shown in Scheme 2. Reductive amination of keto ester 1.3 with ammonia followed by epimerization provides 2.1 as a mostly cis-substituted racemic mixture. Chiral resolution of the enantiomers provides 2.2. N-Alkylation with LiHMDS as base, for example, and an alkyl halide or epoxide affords 1.4. Removal of the Boc protecting group with HCl then affords 1.5 as a hydrochloride salt.
  • [0000]
    Figure US20160130273A1-20160512-C00021
  • [0125]
    A third method to 3-aminopiperidinone intermediates of type 1.5 is shown in Scheme 3. N-Alkylation of 5-bromo-6-methylpyridin-2(1H)-one (3.1) using cesium carbonate as base and an alkyl halide followed by nitration provides 3.2. Palladium-catalyzed cross-coupling with an aryl boronic acid then affords 3.3. Hydrogenation using platinum oxide under acidic conditions and chiral resolution of the mostly cis-substituted racemic product mixture provides 1.5 as a single enantiomer.
  • [0000]
    Figure US20160130273A1-20160512-C00022
  • [0126]
    A synthetic route to 3-aminopiperidinone intermediates of type 4.4 is shown in Scheme 4. Aryl acetonitrile 4.1 can be alkylated using the iodoalanine derivative 1.2 under basic conditions to provide cyano ester 4.2. Reductive cyclization using hydrogen and palladium hydroxide on carbon or Raney nickel, epimerization, and chiral resolution affords cis lactam 4.3 as a single enantiomer. N-Alkylation and removal of the Boc protecting group then provides 4.4 as a hydrochloride salt.
  • [0000]
    Figure US20160130273A1-20160512-C00023
  • [0127]
    Scheme 5 illustrates an alternative route to 3-aminopiperidinone intermediates of type 4.4. The arylacetonitrile 5.1 may be condensed with acrylate 5.2 at elevated temperature to give the 4-cyanobutanoate ester 5.3. Hydrogenation of nitrile 5.3 using Raney nickel catalyst and an ethanolic solution of ammonia affords the corresponding amine product, which typically cyclizes in situ to provide piperidinone 5.4. N-Alkylation of lactam 5.4 may be accomplished by a variety of methods known to those skilled in the art of organic synthesis, the exact choice of conditions being influenced by the nature of the alkylating agent, R1X. Electrophilic azidation of the resulting substituted lactam 5.5 can be accomplished using similar methodology to that described by Evans and coworkers (Evans et al. (1990) J. Am. Chem. Soc. 112, 4011-4030) to provide the azide 5.6 as a mixture of diastereoisomers, which can be separated by chromatography. The desired cis diastereomer of azide 5.6 may be reduced by catalytic hydrogenation in the presence of di-tert-butyl dicarbonate to give the corresponding Boc-protected amine 5.7, and separation of the enantiomers using chiral HPLC or SFC leads to the (3S,5S)-isomer 5.8. Finally, standard deprotection affords the desired 3-aminopiperidinone intermediate 4.4 as a hydrochloride salt.
  • [0000]
    Figure US20160130273A1-20160512-C00024
  • [0128]
    Another approach to 3-aminopiperidinone intermediates of interest, which is particularly useful for preparing 3-amino-6-methyl-5-arylpiperidin-2-ones such as 1.5, is outlined in Scheme 6. The pyridin-2(1H)-one 3.1 may be converted to the N-substituted pyridinone 6.1 by treatment with a suitable electrophile (R1X) under basic conditions. Pyridinone 6.1 can then be subjected to Suzuki-Miyaura coupling with the boronic acid 6.2, and the resulting 5-arylpyridinone 6.3 may be hydrogenated using, for example, platinum(IV) oxide catalyst to afford the corresponding 5-arylpiperidinone 6.4, which is usually obtained as predominantly the cis isomer. Further elaboration of piperidinone 6.4 may be achieved using analogous methodology to that described in Scheme 5. Specifically, electrophilic azidation followed by one-pot reduction and Boc protection leads to carbamate 6.6, and the desired enantiomer may be obtained using chiral chromatography. In some cases, the desired diastereomer of azide 6.5 may be isolated as a racemic mixture of the (3S,5S,6R)- and (3R,5R,6S)-isomers following silica gel chromatography of the crude product, and this mixture may be elaborated as outlined in Scheme 6. In other cases, it may be advantageous to take a mixture of diastereomers of azide 6.5 forward to the corresponding carbamate 6.6. The mixture of carbamate 6.6 diastereomers may be epimerized under basic conditions, such as potassium carbonate in EtOH, to afford a mixture that is significantly enriched in the desired (3S,5S,6R)- and (3R,5R,6S)-isomers, further purification may be employed to obtain the enantiomer of interest as outlined herein.
  • [0000]
    Figure US20160130273A1-20160512-C00025
    Figure US20160130273A1-20160512-C00026
  • [0129]
    A synthetic route to the azaoxindole pyridine acid intermediate 7.4 is shown in Scheme 7. Diazotization of aminopyridine 7.1, whose preparation is described in WO 2008/020902, followed by treatment with potassium iodide in the presence of NaNOprovides iodide 7.2. Palladium-catalyzed carbonylation in methanol then affords ester 7.3, which may be saponified with sodium hydroxide to furnish 7.4.
  • [0000]
    Figure US20160130273A1-20160512-C00027
  • [0130]
    An alternative synthesis of the azaoxindole pyridine acid intermediate 7.4 is shown in Scheme 8. Esterification of diacid 8.1 followed by bromination provides 8.2. Reduction with sodium borohydride then furnishes diol 8.3. Alkylation of the protected azaoxindole 8.4 with the bis-mesylate produced from 8.3 affords the spirocycle 8.5. Palladium-catalyzed carbonylation in methanol followed by chiral resolution gives ester 8.6 as a single enantiomer. Removal of the SEM protecting group under acidic conditions and hydrolysis of the ester using sodium hydroxide then provides 7.4.
  • [0000]
    Figure US20160130273A1-20160512-C00028
  • [0131]
    A synthetic route to diazaoxindole carboxylic acid intermediate 9.7 is shown in Scheme 9. Esterification of acid 9.1 is followed by vinylation under palladium catalysis to afford divinyl pyridine 9.2. Ozonolysis with a borohydride reductive workup then yields diol 9.3. After mesylation and treatment with sodium choride, the resulting dichloro intermediate 9.4 can be alkylated with oxindole 9.5 under basic conditions to give spirocycle 9.6, following chiral resolution of the enantiomers. Dechlorination under buffered hydrogenation conditions and acidic deprotection affords acid 9.7.
  • [0000]
    Figure US20160130273A1-20160512-C00029
  • [0132]
    Useful derivatives of the intermediates described herein may be prepared using well-precedented methodology. One such example is illustrated in Scheme 10, in which the azaoxindole intermediate 7.4 is converted to the corresponding nitrile derivative 10.2, which may be used to prepare compounds of the present invention. Bromination of 7.4 with N-bromosuccinimide in boron trifluoride dihydrate provides the bromo derivative 10.1, which may be converted to the desired nitrile 10.2 using zinc cyanide and a palladium catalyst as shown.
  • [0000]
    Figure US20160130273A1-20160512-C00030
  • [0133]
    A synthetic route to the azaoxindole indane acid intermediate 11.17 is shown in Scheme 11. Esterification of diacid 11.1 followed by hydrogenation using palladium on carbon as a catalyst provides aniline 11.2. Dibenzylation under basic conditions with heat affords 11.3, and reduction of the diester with LiAlHfurnishes diol 11.4. Chlorination with thionyl chloride provides benzyl chloride 11.5. Palladium-catalyzed amination of bromide 11.6 with tert-butylamine gives 11.7. Sequential treatment with n-hexyllithium and methyl chloroformate (2×) affords azaoxindole ester 11.8. Alkylation with the benzylchloride 11.5 under basic conditions in the presence of the cinchonidine-derived catalyst 11.12 (prepared via the alkylation of cinchonidine 11.10 with benzyl bromide 11.11) affords spirocycle 11.13. Deprotection of the azaoxindole using methanesulfonic acid with heat and debenzylation under standard hydrogenation conditions provides aniline 11.14. Diazotization followed by treatment with potassium iodide provides iodide 11.15. Palladium-catalyzed carbonylation in methanol then affords ester 11.16, which may be saponified with sodium hydroxide to furnish 11.17.
  • [0000]
    Figure US20160130273A1-20160512-C00031
  • [0134]
    An alternative synthesis of the azaoxindole pyridine acid intermediate 11.17 is shown in Scheme 12. Alkylation of the azaoxindole ester 11.8 with dibenzyl bromide 12.1 followed by chiral resolution of the enantiomers provides ester 12.2. Sequential deprotection of the azaoxindole using methanesulfonic acid with heat and hydrolysis of the ester provides 11.17.
  • [0000]
    Figure US20160130273A1-20160512-C00032
  • [0135]
    A synthetic route to the diazaoxindole carboxylic acid intermediate 13.4 is shown in Scheme 13. Alkylation of dibromide 12.1 with oxindole 9.5 under basic conditions and subsequent chiral resolution affords spirocycle 13.2. Dechlorination under buffered hydrogenation conditions and ester hydrolysis then affords acid 13.4.
  • [0000]
    Figure US20160130273A1-20160512-C00033
  • [0136]
    Useful derivatives of the intermediates described herein may be prepared using well-precedented methodology. One such example is illustrated in Scheme 14, in which the azaoxindole intermediate 11.17 is converted to the corresponding nitrile derivative 14.2, which may be used to prepare compounds of the present invention. Treatment of 11.17 with bromine in acetic acid provides the bromo derivative 14.1, which may be converted to the desired nitrile 14.2 using zinc cyanide and a palladium catalyst as shown.
  • [0000]
    Figure US20160130273A1-20160512-C00034
  • [0137]
    Scheme 15 illustrates conditions that can be used for the coupling of 3-aminopiperidinone intermediates, such as 15.1, and carboxylic acid intermediate 15.2, to produce, in this instance, amides 15.3. These standard coupling conditions are representative of the methods used to prepare the compounds of the present invention.
  • [0000]
    Figure US20160130273A1-20160512-C00035
  • [0138]
    The previous methods for synthesizing the lactam intermediate suffered from one or more drawbacks: racemic mixture was separated by chiral-HPLC, separation of diasteromixture by crystallization and/or use of costly PtO2. The process of the instant invention utilizes a transaminase induced dynamic kinetic resolution providing high diastereoselectivity at positions C5 and C6. N-mono-trifluoroethylation was discovered and developed. Cis and trans isomer at the alpha position of the amine was successfully controlled by crystallization in the presence of arylaldehyde derivatives. Overall, synthetic steps are shorter, practical and efficient and yield is dramatically improved.
      • Example 1 Isopropyl 2-(tert-butoxycarbonylamino)-3-(methylsulfonyloxy)propanoate (2)

     

    • [0139]
      Figure US20160130273A1-20160512-C00036
    • [0140]
      To a solution of N-tert-butyl-L-serine isopropyl ester 1 (12 g, 48.5 mmol)* and methanesulfonyl chloride (4.0 ml) in dichloromethane (100 mL), triethylamine (7.2 ml) was added slowly under an ice bath. The reaction mixture was stirred at room temperature for 1 h, then 1 N HCl (40 mL) was added with stirring. The organic layer was separated, washed with 1 N HCl (40 ml) and brine (40 ml), dried over MgSO4, and concentrated in vacuo to give 2 (14.5 g, 91.9%) as a solid. 1H NMR (CDCl3, 500 MHz): δ 5.45 (s, broad, 1H), 5.13 (m, 1H), 4.62-4.47 (m, 3H), 3.04 (s, 3H), 1.48 (s, 9H), 1.31 (d, J=6.4 Hz, 6H); 13C NMR (CDCl3, 100 MHz): δ 168.0, 135.1, 80.6, 70.5, 69.1, 53.3, 37.4, 28.3, 21.7, 21.6; HRMS m/z calcd. for C12H23NO7S 348.1087 (M+Na). found 348.1097
    • [0000]
      * preparation of 1 was reported in J. Med. Chem., 2010, 53, 6825-6837 6825

Isopropyl 2-(tert-butoxycarbonylamino)-3-iodopropanoate (3)

    • [0141]
      Figure US20160130273A1-20160512-C00037
    • [0142]
      To a solution of 2 (392 g) in acetone (3.14 L), sodium iodide (542 g) was added. The reaction temperature went up to 29° C. from 17° C. The reaction mixture was maintained at room temperature over weekend. The mixture was filtrated and washed with MTBE. The filtrate and washings were combined and concentrated. The residue was treated with MTBE and water with a small amount of sodium thiosulfate. The organic layer was washed with water and concentrated to an oil. The oil was charged slowly into a mixture of water (2 L) and DMF (300 ml) with a small amount of seed at 5° C. The crystals were filtered and dried to give 3 (400 g, 93% yield).

Isopropyl 4-(4-bromophenyl)-2-(tert-butoxycarbonylamino)-5-oxohexanoate (5) and isopropyl 4-phenyl-2-(tert-butoxycarbonylamino)-5-oxohexanoate (6)

    • [0143]
      Figure US20160130273A1-20160512-C00038
    • [0144]
      To a solution of 4 (51.7 g, 243 mmol) in DMF (850 ml) was added 3 (88 g, 246 mmol). The resulting solution was cooled to 5° C. and Cs2CO(240 g) was added in one portion. The suspension was warmed to 15° C. and stirred at this temperature for 2.5 h. Additional Cs2CO(25 g) was charged and the mixture was stirred for additional 8 h or until HPLC analysis indicated the conversion was greater than 95%. The batch was then slowly quenched into a mixture of 2N HCl (850 mL) and MTBE (900 mL) at 5-20° C. Organic layer was separated and aqueous layer extracted with MTBE (400 mL). Combined organic layers were washed with 5% NaHCO3solution (400 mL) twice. The resulting solution containing desired product 5 (90% LC purity) was concentrated under vacuum. The residue was dissolved in isopropanol (1 L). To the solution was added K2CO(25 g), potassium formate (34 g) and 10% Pd/C (20 g). The mixture was warmed up to 60° C. and stirred for 2 h. The mixture was filtered after cooling to room temperature. The HPLC analysis of the filtrate indicated that the solution contained 6 (54.7 g, 95 wt %, 62% yield). The crude product was used directly in the next step without further purification. The compound 6 is a mixture of two pair of diastereomers 6-1 and 6-2, partially separable by flash chromatography on silica gel with ethyl acetate and heptane as a eluant (1:10). 6-1: 1H NMR (CDCl3, 500 MHz): δ 7.35 (m, 2H), 7.30 (m, 1H), 7.20 (m, 2H), 5.17 (br, 1H), 4.95 (m, 1H), 4.76 (br, 1H), 3.73 (m, 1H), 2.70 (br, 1H), 2.07 (s, 1H), 1.45 (s, 9H), 1.29 (d, J=6.6 Hz, 3H), 1.28 (d, J=6.6 Hz, 3H); 6-2: 1H NMR (CDCl3, 500 MHz): δ 5.12 (m, 1H), 4.70 (m, 1H), 3.27 (m, 1H), 2.80 (m 1H), 2.34 (s, 3H), 1.50 (s, 9H), 1.26 (d, J=6.6 Hz, 3H), 1.25 (d, J=6.6 Hz, 3H); HRMS m/z: cacld. for 6-1: C20H29NO386.1938 (M+Na). found 386.1947.

Isopropyl 2-((tert-butoxycarbonyl)amino)acrylate (7)

    • [0145]
      Figure US20160130273A1-20160512-C00039
    • [0146]
      To a solution of 1 (10.05 g, 40.6 mmol) in DMF (100 mL) was added MsCl (4.12 mL, 52.8 mmol) under ice-cooling. Triethylamine (14.16 mL, 102.0 mmol) was then added dropwise via an addition funnel over 30 min, while maintaining the reaction temperature between 0-5° C. When the addition was complete, the cooling bath was removed and the yellow heterogeneous reaction mixture was aged at room temperature under N2for overnight. The reaction mixture was diluted with ice cold water (1 L) and MTBE (1 L). The layers were separated and the aqueous layer was back-extracted with MTBE (500 mL). The organic layers were combined and washed with 1M citric acid (750 mL), water (1 L) and then 10% aqueous NaCl (1 L). The organic solution contained 7 (8.652 g, 93% yield). Solvent was switched to DMSO at <40° C. and use solution directly in next step.

Isopropyl 4-phenyl-2-(tert-butoxycarbonylamino)-5-oxohexanoate (6)

    • [0147]
      Figure US20160130273A1-20160512-C00040
    • [0000]
      Compound 6 was prepared from 7 in DMSO in the presence of 0.5 equiv. Cs2COwith 1.05 equiv. of phenylacetone at room temperature in 79% yield.

tert-Butyl(5S,6R)-6-methyl-2-oxo-5-phenylpiperidin-3-ylcarbamate (8)

    • [0148]
      Figure US20160130273A1-20160512-C00041
    • [0149]
      To a 5 L RBF with overhead stirring, a temperature control, a pH probe and a base addition line, was added sodiumtetraborate decahydrate (26.7 g) and DI water (1.4 L). After all solids were dissolved, isopropylamine (82.8 g) was added. The pH of the buffer was adjusted to pH 10.5 using 6 N HCl. The buffer was cooled to room temperature. Then, pyridoxal-5-phosphate (2.8 g) and SEQ ID NO: 1 (70 g) were added and slowly dissolved at room temperature.
    • [0150]
      An oil (197.9 g, containing 70.7 wt % keto ester 6 (140 g, 0.385 mol) were dissolved in DMSO (1.4 L). The solution was added to the flask over 5-10 min and the reaction was heated to 55° C. The pH was adjusted to 10.5 according to a handheld pH meter and controlled overnight with an automated pH controller using 8 M aqueous isopropylamine. The reaction was aged for 24 h.
    • [0151]
      After confirmation of >95A % conversion by HPLC, the reaction was extracted by first adding a mixture of iPA:IPAc (3:4, 2.8 L) and stirring for 20 min. The phases were separated and the aqueous layer was back extracted with a mixture of iPA:IPAc (2:8, 2.8 L). The phases were separated, the organic layers were combined and washed with DI water (0.5 L). The HPLC based assay yield in the organic layer was 8 (114.6 g) with >60:1 dr at the positions C5 and C6. The ratio of stereoisomers at position C2 was ˜1:1. The extract was concentrated and dissolved in CH2Cl2. The organic solution was washed with water then saturated aqueous NaCl, concentrated and crystallized from MTBE/n-hexane (2:3). The crystal was filtered at room temperature and washed with MTBE/n-hexane (2:3) and dried to afford a cis and trans mixture (˜1:1.2) of the lactam 8 (99.6 g, 80.0%) as crystals.
    • [0000]
      cis: trans (˜1:1.2) mixture but NMR integration was reported as 1:1 (for proton number counts) Mp 87-90.9° C.; 1H NMR (CDCl3, 400 MHz): δ 7.40-7.20 (m, 8H, cis and trans), 7.16-7.12 (m, 2H, cis and trans); 6.56 (broad s, 1H, trans), 6.35 (broad s, 1H, cis), 5.57 (broad d, J=4.6 Hz, 1H, cis), 5.34 (broad d, J=5.7 Hz, 1H, trans), 4.33-4.15 (m, 2H, cis and trans), 3.93 (m, 1H, trans), 3.81 (m, 1H, cis), 3.41 (dt, J=11.8, 5.0 Hz, 1H, cis), 3.29 (dt, J=8.0, 4.4 Hz, 1H, trans), 2.74 (m, 1H, cis), 2.57 (m, 1H, trans), 2.23 (ddd, J=13.5, 8.0, 4.4 Hz, trans), 2.07 (q, J=11.8 Hz, 1H, cis), 1.46 (s, 9H, cis), 1.42 (s, 9H, trans), 1.05 (d, J=6.9 Hz, 3H, trans), 0.89 (d, J=6.9 Hz, 3H, cis); 13C NMR (CDCl3, 100 MHz): δ 171.5(cis), 171.4(trans), 156.0(cis or trans), 155.93 (cis or trans), 140.8 (cis), 139.9 (trans), 128.8 (trans), 128.7 (cis), 128.6 (trans), 128.1 (cis), 127.2(trans), 127.1(cis), 79.9(trans), 79.91(cis), 52.4 (trans), 51.8 (broad, cis), 51.7 (cis), 49.0 (broad, trans), 42.1 (cis), 41.9 (trans), 32.4 (broad, trans), 30.1 (cis), 28.5(cis or trans), 28.53(cis or trans), 18.3 (cis), 18.1 (broad, trans); HRMS m/z cacld. for C17H24N2O3327.1679 (M+Na). found 327.1696

tert-Butyl(5S,6R)-6-methyl-2-oxo-5-phenyl-1-(2,2,2-trifluoroethyl)piperidine-3-ylcarbamate (9) and tert-butyl(5S,6R)-6-methyl-2-oxo-5-phenyl-1-(2,2,2-trifluoroethyl)piperidine-3-yl(2,2,2-trifluoroethyl)carbamate (10)

    • [0152]
      Figure US20160130273A1-20160512-C00042
    • [0153]
      To the solution of 8 (480 g, 1.58 mol) in anhydrous THF (3.8 L) was added lithium tert-amoxide solution in heptane (512 mL, 3.1 M, 1.58 mol) over about 15 min while maintaining the reaction temperature between 15 and 20° C. The resulting solution was then cooled to a temperature between 0 and 2° C. 2,2,2-Trifluoroethyl trifluoromethanesulfonate (368 g, 1.58 mol) was added over 15 min while maintaining the reaction temperature between 0 and 3° C. The solution was agitated at 0° C. for 15 min. DMPU (300 ml) was charged to the mixture through an additional funnel over 30 min while maintaining the reaction temperature between 0 and 3° C. The resulting solution was agitated at 0° C. for 2.5 h. Another 2,2,2-trifluoroethyl trifluoromethanesulfonate (182 g, 0.79 mol) was added to the mixture over 10 min followed by another 3.1 M lithium tert-amoxide solution (104 mL) while maintaining the reaction temperature between 0 and 3° C. The batch was agitated for another 2.5 h at 0° C. The mixture was quenched into a mixture of heptane (4.8 L), water (3.4 L) and 2N HCl solution (280 mL) below 15° C. The phases were separated. The aqueous phase was extracted with heptane (4 L). The combined organic phase was washed with water (2 L). The solution was concentrated to a volume of about 1 L under vacuum between 25 and 50° C. The crude material was passed through a short silica gel plug with heptane/ethyl acetate. The resulting solution was concentrated under vacuum until distillation stopped at a temperature below 50° C., dissolved in IPAc (2 L) and used for the next processing step. The assay yield of 9 for both cis and trans isomers was 85% in the ratio of ˜8 to 1.
    • [0154]
      Analytically pure cis and trans isomers of 9 were isolated by chromatography on silica gel with ethyl acetate and heptane as eluant. 9 (cis): 1H NMR (CDCl3, 500 MHz): δ 7.30 (m, 5H), 5.75 (s, broad, 1H), 4.35 (m, 1H), 4.15 (m, 1H), 3.80 (m, 1H), 3.50 (m, 1H), 3.17 (m, 1H), 2.45 (m, 2H), 1.45 (s, 9H), 0.93 (d, J=6.7 Hz, 3H); 13C NMR (CDCl3, 100 MHz): δ 170.3, 155.9, 140.0, 128.6, 127.6, 127.1, 124.6 (q, J=279 Hz), 79.7, 58.7, 52.2, 45.3 (q, J=33.7 Hz), 41.9, 28.3, 27.4, 13.4; HRMS: m/z calcd for C19H25F3N2O387.1890 (M+H). found: 387.1899. 9 (trans): 1H NMR (CDCl3, 500 MHz): δ 7.40 (m, 2H), 7.30 (m, 3H), 5.55 (br, 1H), 4.53 (br, 1H), 4.45 (m, 1H), 3.78 (m 2H), 3.45 (m, 1H), 3.0 (m, 1H), 2.12 (m, 1H), 1.46 (s, 9H), 1.12 (d, J=7.0 Hz, 3H); 13C NMR (CDCl3, 100 MHz): δ 170.2, 155.9, 139.6, 128.7, 127.9, 127.4, 124.3 (q, J=279 Hz), 80.0, 59.6, 49.1, 46.9 (q, J=34.0 Hz), 42.1, 28.3, 25.3, 13.4; HRMS: m/z calcd for C19H25F3N2O3387.1890 (M+H). found 387.1901.

(3S,5S,6R)-6-Methyl-2-oxo-5-phenyl-1-(2,2,2-trifluoroethyl)piperidine-3-aminium 4-nitrobenzoate (11)

    • [0155]
      Figure US20160130273A1-20160512-C00043
    • [0156]
      To a solution of the crude 9 obtained from above experiment (10 g assay, 25.9 mmol) in iPAC (8 ml) was added p-toluenesulfonic acid monohydrate (6.7 g, 35.2 mmol) and the mixture was stirred at 50-60° C. for 3 hr until the reaction was completed (>99%). The solution was cooled to 15-20° C., and washed with 10% aqueous K2COfollowed by water. The aqueous layers were re-extracted with iPAc (5 ml). The organic layers were combined and heated to 55-60° C. 4-Nitrobenzoic acid (3.9 g, 23.2 mmol) was slowly added in 20 min. The mixture was slowly cooled to room temperature. 5-Nitro-2-hydroxylbenzaldehyde (50 mg) was added and the batch was agitated for at least 12 h. The mixture was filtrated and washed with MeCN to give 11 as crystals. Optionally, a slurry in MeCN was carried out for further purification of 11. The isolated yield was 90%. Mp 205-208° C.; 1H NMR (DMSO-d6, 400 MHz): δ 8.21 (dd, J=9.0, 2.1 Hz, 2H), 8.08 (dd, J=9.0, 2.1 Hz, 2H), 7.37 (t, J=7.4 Hz, 2H), 7.28 (t, J=7.4 Hz, 1H), 7.24 (d, J=7.4 Hz, 2H), 4.65 (ddd, J=15.1, 9.7, 7.7 Hz, 1H), 3.72-3.98 (m, 3H), 3.57 (m, 1H), 2.46 (q, J=12.6 Hz, 1H), 2.25 (m, 1H), 0.90 (d, J=6.4 Hz, 3H); 19F NMR (DMSO-d6, 376 MHz): δ −69 (s); 13C NMR (DMSO-d6, 100 MHz): δ 168.7, 167.3, 148.3, 143.8, 140.1, 130.1, 128.6, 127.4, 127.0, 124.9 (q, J=280.9 Hz), 122.8, 58.7, 49.8, 44.5 (q, J=32.7 Hz), 40.6, 25.3, 13.2.

(5S,6R)-3-Amino-6-methyl-5-phenyl-1-(2,2,2-trifluoroethyl)piperidin-2-one (12)

    • [0157]
      Figure US20160130273A1-20160512-C00044
    • [0158]
      To a mixture of 8 (20.0 g, 65.7 mmol) and Na2S2O(0.52 g, 3.3 mmol) in THF (200 mL) was added tert-BuOLi (6.8 g, 85 mmol) at 16° C. The mixture was stirred at 16° C. for 15 min followed by addition of trifluoroethyl trifluoromethansulfonate (20.6 g, 89 mmol) in one portion. The resulting mixture was stirred for 18 h at 16° C. The reaction mixture was then quenched by addition of toluene (70 mL) followed by 0.5N HCl solution (50 mL). The aqueous layer was separated and extracted with toluene (20 mL). The combined organic layer contained 87% of 9, 6% of 10 and 6% of 8 by HPLC and yield for the desired product 9 was 87%. The organic layer was then stirred with 3N HCl solution (80 ml) and tetrabutylammoniium bromide (0.8 g) for about 3 h until HPLC analysis indicated selective removal of the Boc group in the unreacted 8 was completed. The aqueous layer was removed. The organic layer containing 9 and 10 was then concentrated under vacuum at 60° C. to remove most of solvent. The residue was dissolved in MTBE (60 mL), and 5N HCl solution (65 mL) was added. The diphasic solution was agitated vigorously at 50° C. for about 5 h until the deprotection of 9 was completed while 10 was mainly intact. After addition of heptane (30 mL) to the mixture, the organic layer was separated at 45° C. The aqueous layer was diluted with water (60 mL) and resulting aqueous and washed with heptane (30 mL) at 45° C. The aqueous solution was then mixed with MTBE (100 mL) and basified with 10 N NaOH solution until the pH of the mixture was about 10. The organic layer was separated and the aqueous layer was back-extracted with MTBE (60 mL). The combined organic layers were washed with brine (60 mL). The resulting organic solution was suitable for next reaction. The solution was contained 12 (15.6 g, 83% from 8) with 97% LC purity as a mixture of two diastereomers (cis and trans) in 4 to 1 ratio.

(3S,5S,6R)-6-Methyl-2-oxo-5-phenyl-1-(2,2,2-trifluoroethyl)piperidin-3-aminium 4-methylbenzoate (13)

    • [0159]
      Figure US20160130273A1-20160512-C00045
    • [0160]
      To a suspension of 4-methylbenzoic acid (6.8 g, 49.9 mmol) and 3,5-dichlorosalicylaldehyde (93 mg, 0.49 mmol) in MTBE (40 mL) was added a solution of 12 (13.9 g, 48.5 mmol) in MTBE (about 150 mL) over 1 h at 50° C. The resulting suspension was agitated for about 3 h at 50° C. The solids were collected by filtration after cooling to −5° C. over 1 h. The cake was washed with MTBE (50 mL). The solids were dried in a vacuum oven to give 13 (17.6 g, 86%) as crystals with 99.5% LC purity and 99.6% de. 1H NMR (DMSO-d6, 400 MHz): δ 7.85 (d, J=8.1 Hz, 2H), 7.40 (m, 2H), 7.25 (m, 5H), 6.0 (br, 3H), 4.65 (m, 1H), 3.65-3.80 (m, 2H), 3.45-3.65 (m, 2H), 2.35 (s, 3H), 2.30 (m, 1H), 2.15 (m, 1H), 0.88 (d, J=6.5 Hz, 3H); 13C NMR (DMSO-d6, 100 MHz): δ 172.4, 168.5, 142.1, 141.1, 130.9, 129.7, 129.2, 129.0, 128.0, 125.5 (q, J=279 Hz), 59.1, 51.6, 45.1 (q, J=32 Hz), 41.6, 28.0, 21.5, 13.9.

(S)—N-((3S,5S,6R)-6-Methyl-2-oxo-5-phenyl-1-(2,2,2-trifluoroethyl)piperidine-3-yl)-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxamide trihydrate (15)

    • [0161]
      Figure US20160130273A1-20160512-C00046
    • [0162]
      To a suspension of 11 (465 g, 96% wt, 0.99 mol) in iPAc (4.6 L) was added 5% aqueous K3PO(4.6 L). The mixture was stirred for 5 min. The organic layer was separated and washed with 5% aqueous K3PO(4.6 L) twice and concentrated in vacuo and dissolved in acetonitrile (1.8 L).
    • [0163]
      To another flask was added 14 (303 g, 91.4 wt %), acetonitrile (1.8 L) and water (1.8 L) followed by 10 N NaOH (99 mL). The resulting solution was stirred for 5 min at room temperature and the chiral amine solution made above was charged to the mixture and the container was rinsed with acetonitrile (900 mL). HOBT hydrate (164 g) was charged followed by EDC hydrochloride (283 g). The mixture was agitated at room temperature for 2.5 h. To the mixture was added iPAc (4.6 L) and organic layer was separated, washed with 5% aqueous NaHCO(2.3 L) followed by a mixture of 15% aqueous citric acid (3.2 L) and saturated aqueous NaCl (1.2 L). The resulting organic layer was finally washed with 5% aqueous NaHCO(2.3 L). The organic solution was concentrated below 50° C. and dissolved in methanol (2.3 L). The solution was slowly added to a mixture of water (6 L) and methanol (600 mL) with ˜2 g of seed crystal. And the resulting suspension was stirred overnight at room temperature. Crystals were filtered, rinsed with water/methanol (4 L, 10:1), and dried under nitrogen flow at room temperature to provide 15 (576 g, 97% yield) as trihydrate.
    • [0164]
      1H NMR (500 MHz, CDCl3): δ 10.15 (br s, 1H), 8.91 (br s, 1H), 8.21 (d, J=6.0 Hz, 1H), 8.16 (dd, J=5.3, 1.5 Hz, 1H), 8.01 (br s, 1H), 7.39-7.33 (m, 2H), 7.31-7.25 (m, 1H), 7.22-7.20 (m, 2H), 7.17 (dd, J=7.4, 1.6 Hz, 1H), 6.88 (dd, J=7.4, 5.3 Hz, 1H), 4.94 (dq, J=9.3, 7.6 Hz, 1H), 4.45-4.37 (m, 1H), 3.94-3.87 (m, 1H), 3.72 (d, J=17.2 Hz, 1H), 3.63-3.56 (m, 2H), 3.38-3.26 (m, 1H), 3.24 (d, J=17.3 Hz, 1H), 3.13 (d, J=16.5 Hz, 1H), 2.78 (q, J=12.5 Hz, 1H), 2.62-2.56 (m, 1H), 1.11 (d, J=6.5 Hz, 3H); 13C NMR (126 MHz, CD3CN): δ 181.42, 170.63, 166.73, 166.63, 156.90, 148.55, 148.08, 141.74, 135.77, 132.08, 131.09, 130.08, 129.66, 129.56, 128.78, 128.07, 126.25 (q, J=280.1 Hz), 119.41, 60.14, 53.07, 52.00, 46.41 (q, J=33.3 Hz), 45.18, 42.80, 41.72, 27.79, 13.46; HRMS m/z: calcd for C29H26F3N5O550.2061 (M+H). found 550.2059.

Alternative Procedure for 15

    • [0165]
      Figure US20160130273A1-20160512-C00047
    • [0166]
      To a suspension of 13 (10 g, 98 wt %, 23.2 mmol) in MTBE (70 mL) was added 0.6 N HCl (42 mL). The organic layer was separated and extracted with another 0.6 N HCl (8 mL). The combined aqueous solution was washed with MTBE (10 mL×3). To the resulting aqueous solution was added acetonitrile (35 mL) and 14 (6.66 g, 99 wt %). To the resulting suspension was neutralized with 29% NaOH solution to pH 6. HOPO (0.26 g) was added followed by EDC hydrochloride (5.34 g). The mixture was stirred at room temperature for 6-12 h until the conversion was complete (>99%). Ethanol (30 ml) was added and the mixture was heated to 35° C. The resulting solution was added over 2 h to another three neck flask containing ethanol (10 mL), water (30 mL) and 15 seeds (0.4 g). Simultaneously, water (70 mL) was also added to the mixture. The suspension was then cooled to 5° C. over 30 min and filtered. The cake was washed with a mixture of ethanol/water (1:3, 40 mL). The cake was dried in a vacuum oven at 40° C. to give 15 trihydrate (13.7 g, 95%) as crystals.

Example 2 N-Methoxy-N-methyl-2-(2,3,6-trifluorophenyl)acetamide (17)

    • [0167]
      Figure US20160130273A1-20160512-C00048
    • [0168]
      To a solution of DMF (58.1 mL, 750 mmol) in iPAc (951 mL) was added POCl(55.9 mL, 600 mmol) under ice-cooling. After aged for 1 h under ice-bath, acid 16 (95 g, 500 mmol) was added under ice-cooling. The solution was stirred under ice-cooling for 30 min. The solution was added over 30 min into a solution of K2CO(254 g, 1.835 mol) and NHMe(OMe)HCl (73.2 g, 750 mmol) in water (951 mL) below 8° C. After aged for 30 min below 8° C., the organic layer was separated, washed with water (500 mL) twice and sat. NaCl aq (100 mL) once, and concentrated in vacuo to afford 17 as an oil (117.9 g, 97.7 wt %, 99% yield). 1H NMR (CDCl3, 400 MHz); δ 7.05 (m, 1H), 6.82 (m, 1H), 3.86 (s, 2H), 3.76 (s, 3H), 3.22 (s, 3H); 19F NMR (CDCl3, 376.6 MHz); δ −120.4 (dd, J=15.1, 2.7 Hz), −137.9 (dd, J=20.8, 2.7 Hz), −143.5 (dd, J=20.8, 15.1 Hz); 13C NMR (CDCl3, 100 MHz); δ 169.4, 156.9 (ddd, J=244, 6.2, 2.7 Hz), 149.3 (ddd, J=249, 14.4, 8.4 Hz), 147.1 (ddd, J=244, 13.1, 3.5 Hz), 115.5 (ddd, J=19.4, 9.9, 1.5 Hz), 133.4 (dd, J=22.3, 16.4 Hz), 110.2 (ddd, J=24.8, 6.7, 4.1 Hz), 32.4 (broad), 26.6 (m); HRMS m/z calcd for C10H10F3NO234.0736 (M+H). found 234.0746.

1-(2,3,6-Trifluorophenyl)propan-2-one (18)

    • [0169]
      Figure US20160130273A1-20160512-C00049
    • [0170]
      A mixture of CeCl(438 g, 1779 mmol) and THF (12 L) was heated at 40° C. for about 2 h then cooled to 5° C. Methylmagensium chloride in THF (3 M, 3.4 L) was charged at 5-9° C. and then it was warmed up to 16° C. and held for 1 h. The suspension was re-cooled to −10 to −15° C. A solution of 17 (1.19 kg) in THF (2.4 L) was charged into the suspension over 15 min. After confirmation of completion of the reaction, the reaction mixture was transferred to a cold solution of hydrochloric acid (2 N, 8.4 L) and MTBE (5 L) in 5-10° C. The aqueous phase was separated and the organic layer was washed with aqueous 5% K2CO(6 L) and then 10% aqueous NaCl (5 L). The organic layer was dried over Na2SO4, concentrated to give crude 18 (917 g, >99 wt %) in 95% yield. The crude 18 was used in the next step without further purification. Analytically pure 18 was obtained by silica gel column.
    • [0171]
      1H NMR (CDCl3, 400 MHz); δ 7.07 (m, 1H), 6.84 (m, 1H), 3.82 (s, 2H), 2.28 (s, 3H); 19F NMR (CDCl3, 376.6 MHz); δ −120.3 (dd, J=15.3, 2.5 Hz), −137.8 (dd, J=21.2, 2.5 Hz), −143.0 (dd, J=20.2, 15.3 Hz); 13C NMR (CDCl3, 100 MHz); δ 202.2, 156.5 (ddd, J=244, 6.3, 2.9 Hz), 148.9 (ddd, J=249, 14.4, 8.6 Hz), 147.0 (ddd, J=244, 13.1, 3.5 Hz), 115.7 (ddd, J=19.4, 10.5, 1.2 Hz), 112.8 (dd, J=22.7, 17.0 Hz), 110.3 (ddd, J=24.8, 6.7, 4.1 Hz), 37.2 (d, J=1.2 Hz), 29.3.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

    • [0172]
      Figure US20160130273A1-20160512-C00050
    • [0173]
      To a solution of 18 (195 g, 1.03 mol) in MTBE (1.8 L) was added zinc bromide (67 g, 0.30 mol) followed by 2 (390 g, 1.2 mol). tert-BuOLi (290 g, 3.6 mol) was then added in several portions while maintaining the reaction temperature below 40° C. The resulting mixture was stirred at 35° C. for 24 h and quenched into a mixture of 2 N HCl (5.6 L) and heptane (5 L) at 0° C. The organic layer was separated and washed with 5% aqueous NaHCO(5 L) twice. The resulting organic solution was concentrated under vacuum. The residue was dissolved in heptane (2 L) and the solution was concentrated again under vacuum. The resulting oil was dissolved in DMSO (2.5 L) and the solution was used in the next step without further purification. HPLC analysis indicated that the solution contained the desired product 19 (290 g, 67% yield) as the major component along with 5% of starting material 18. The analytically pure product 19 as one pair of diastereomers was isolated by chromatography on silica gel with ethyl acetate and heptane mixture as an eluant. HRMS: m/z calcd for C20H26F3NO418.1836 (M+H). found 418.1849.

tert-Butyl((5S,6R)-6-methyl-2-oxo-5-(2,3,6-trifluorophenyl)piperidin-3-yl)carbamate (20)

    • [0174]
      Figure US20160130273A1-20160512-C00051
    • [0175]
      To a 0.5 L cylindrical Sixfors reactor with an overhead stirring, a temperature control, a pH probe and a base addition line, was added sodiumtetraborate decahydrate (3.12 g) and DI water (163 mL). After all solids were dissolved, isopropylamine (9.63 g) was added. The pH of the buffer was adjusted to pH 10.5 using 6 N HCl. The buffer was cooled to room temperature. Then, pyridoxal-5-phosphate (0.33 g) and SEQ ID NO: 1 (8.15 g) were added and slowly dissolved at room temperature.
    • [0176]
      Crude keto ester 19 (23.6 g, 69 wt %, 16.3 g assay, 39 mmol) was dissolved in DMSO (163 mL) and the solution was added to the reactor over 5-10 min. Then the reaction was heated to 55° C. The pH was adjusted to 10.5 according to a handheld pH meter and controlled overnight with an automated pH controller using 8 M aqueous isopropylamine. The reaction was aged for 27.5 hours.
    • [0177]
      After confirmation of >95A % conversion by HPLC, the reaction was extracted by first adding a mixture of iPA: iPAc (3:4, 350 mL) and stirring for 20 min. The phases were separated and the aqueous layer was back extracted with a mixture of iPA: iPAc (2:8, 350 mL). The phases were separated. The organic layers were combined and washed with DI water (90 mL). The HPLC based assay yield in the organic layer was 20 (9.86 g, 70.5% assay yield) with >60:1 dr at the positions C5 and C6.

tert-Butyl((3S,5S,6R)-6-methyl-2-oxo-5-(2,3,6-trifluorophenyl)piperidin-3-yl)carbamate (21)

    • [0178]
      Figure US20160130273A1-20160512-C00052
    • [0179]
      A solution of crude cis and trans mixture 20 in a mixture of iPAc and iPA (1.83 wt %, 9.9 kg; 181 g assay as a mixture) was concentrated in vacuo and dissolved in 2-Me-THF (3.6 L). To the solution was added tert-BuOK (66.6 g, 0.594 mol) at room temperature. The suspension was stirred at room temperature for 2 h. The mixture was poured into water (3.5 L) and the organic layer was separated, washed with 15 wt % of aqueous NaCl (3.5 L), dried over Na2SO4, and concentrated to dryness. The residue was suspended with iPAc (275 mL) and heptane (900 mL) at 60° C. The suspension was slowly cooled down to 1° C. The solid was filtered and rinsed with iPAc and heptane (1:3), dried to afford 21 (166 g, 93 wt %; 85%) as crystals. Mp 176-179° C.; 1H NMR (CDCl3, 500 MHz): δ 7.06 (m, 1H), 6.84 (m, 1H), 5.83 (broad s, 1H), 5.58 (broad s, 1H), 4.22 (m, 1H), 3.88-3.79 (m, 2H), 2.77 (m, 1H), 2.25 (m, 1H), 1.46 (s, 9H), 1.08 (d, J=6.4 Hz, 3H); 19F NMR (CDCl3, 376 MHz): δ −117 (d, J=14 Hz), −135 (d, J=20 Hz), −142 (dd, J=20, 14 Hz); 13C NMR (CDCl3, 100 MHz): δ 171.1, 156.6 (ddd, J=245, 6.4, 2.8 Hz), 155.8, 149.3 (ddd, J=248, 14.4, 8.8 Hz), 147.4 (ddd, J=245, 14.2, 3.8 Hz), 118.0 (dd, J=19.3, 14.5 Hz), 115.9 (dd, J=19.2, 10.4 Hz), 111.0 (ddd, J=26.4, 6.0, 4.3 Hz), 79.8, 51.4, 49.5, 34.1, 29.3, 28.3, 18.0; HRMS: m/z calcd for C17H21F3N2O381.1396 (M+Na). found 381.1410.

tert-Butyl((5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl)carbamate (22)

    • [0180]
      Figure US20160130273A1-20160512-C00053
    • [0181]
      To a solution of 21 (10 g, 87% purity, 24.3 mmol) in THF (70 ml) was added tert-BuOLi (2.5 g, 31.2 mmol) at 5° C. in one portion. The solution was cooled to between 0 and 5° C. and trifluoroethyl trifluoromethanesulfonate (10.0 g, 43 mmol) was added in one portion. DMPU (7 mL) was added slowly over 15 min while maintaining the the reaction temperature below 5° C. After the mixture was stirred at 0° C. for 3 h, additional tert-BuOLi (0.9 g, 11.2 mmol) was added. The mixture was aged for an additional 90 min. The mixture was quenched with 0.2 N HCl (70 ml), followed by addition of heptane (80 ml). The organic layer was separated and aqueous layer extracted with heptane (30 ml). The combined organic layers were washed with 15% aqueous citric acid (50 mL) and 5% aqueous NaHCO3(50 mL). The solution was concentrated under vacuum at 40° C. and the resulting oil was dissolved in iPAc (30 mL). The solution was used directly in the next step without further purification. The HPLC analysis indicated that the solution contained 22 (9.8 g, 92% as cis and trans mixture in a ratio of 6.5 to 1) along with 4% of starting material 21 and 8% of a N,N′-alkylated compound. Analytically pure 22 (cis isomer) was isolated by chromatography on silica gel with ethyl acetate and heptane as an eluant. 1H NMR (CDCl3, 500 MHz): δ 7.15 (m, 1H), 6.85 (m, 1H), 5.45 (broad, s, 1H), 4.90 (m, H), 4.20 (m, 1H), 3.92 (m, 2H), 3.28 (m, 1H), 2.70 (m, 2H), 1.48 (s, 9H), 1.20 (d, J=5.9 Hz, 3H); 13C NMR (CDCl3, 100 MHz): δ 170.2, 156.9 (ddd, J=245, 6.3, 2.7 Hz), 156.0, 149.6 (ddd, J=251, 14.8, 8.8 Hz), 147.6 (ddd, J=246, 13.9, 3.6 Hz), 124.5 (q, J=281 Hz), 117.6 (dd, J=19.2, 3.7 Hz), 116.4 (dd, J=19.1, 10.4 Hz), 111.4 (ddd, J=25.8, 6.4, 4.1 Hz), 56.6, 52.8, 45.3 (q, J=34.2 Hz), 35.2, 28.7, 28.3 (br t, J=4 Hz), 14.6; HRMS: m/z calcd for C19H22F6N2O(M+H): 441.1607. found 441.1617.

(3S,5S,6R)-6-Methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-aminium (S)-2-acetamido-3-phenylpropanoate (23)

    • [0182]
      Figure US20160130273A1-20160512-C00054
    • [0183]
      iPAc solution of 22 (529 g assayed, 1.2 mol), obtained from previous step, was diluted to 6 L with iPAc, p-toluenesulfonic acid monohydride (343 g, 1.8 mol) was added and the solution was heated to 55° C. After 4 h, the reaction completed (>99% conversion). Aqueous K2CO(530 g in 3 L of water) was charged into the solution after cooled to 15-25° C. The aqueous layer was separated and was back-extracted with iPAc (2 L). The iPAc solutions were combined and the total volume was adjusted to 10 L by adding iPAc. The solution was heated to 50-60° C. About 20 g of N-acetyl L-phenylalanine was added and the solution was agitated for 15 min or until solids precipitated out. The remaining N-acetyl L-phenylalanine (total 250 g, 1.2 mol) was charged slowly and 2-hydroxy-5-nitrobenzaldehyde (2 g) was charged. The suspension was agitated for 12 h at 20° C. and then cooled to 0° C. for 3 h. The suspension was filtrated, washed with iPAc three times and dried to give 23 (583 g, 89% yield) as crystals. Mp 188-190° C.; 1H NMR (DMSO-d6, 400 MHz): δ 7.96 (d, J=8.0 Hz, 1H), 7.48 (m, 1H), 7.15-7.25 (m, 6H), 4.65 (ddd, J=19.4, 15.3, 9.6 Hz, 1H), 4.33 (ddd, J=8.7, 8.4, 4.9 Hz, 1H), 3.70-3.87 (m, 3H), 3.57 (dd, J=11.5, 6.6 Hz, 1H), 3.04 (dd, J=13.7, 4.9 Hz, 1H), 2.82 (dd, J=13.7, 8.9 Hz, 1H), 2.59 (m, 1H), 2.24 (m, 1H), 2.95 (s, 3H), 1.10 (d, J=6.4 Hz, 1H); 19F NMR (DMSO-d6, 376 MHz): δ −69 (s), −118 (d, J=15 Hz), −137 (d, J=21 Hz), −142 (dd, J=21, 15 Hz); 13C NMR (DMSO-d6, 100 MHz): δ 173.6, 171.1, 168.7, 156.3 (ddd, J=243.5, 7.0, 3.1 Hz), 148.7 (ddd, J=249, 14.4, 9.1 Hz), 146.8 (ddd, J=245, 13.7, 3.1 Hz), 138.5, 129.2, 128.0, 126.1, 124.9 (q, J=280.9 Hz), 117.4.0 (dd, J=19.3, 13.8 Hz), 116.7 (dd, J=19.3, 10.6 Hz), 111.8 (ddd, J=26.0, 6.7, 3.6 Hz), 56.6, 54.3, 51.2, 44.3 (q, J=32.5 Hz), 37.2, 34.8, 26.9 (br t, J=4 Hz), 22.5, 14.1.

(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-aminium 2,2-diphenylacetate (25)

    • [0184]
      Figure US20160130273A1-20160512-C00055
    • [0185]
      To a mixture of crude material containing (5S,6R)-3-amino-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one (24, 2.00 g, 5.88 mmol), prepared according to the same method as the previous example, and 3,5-dichloro-2-hydroxybenzaldehyde (0.011 g, 0.059 mmol) in isopropyl acetate (15.0 ml) at 55-60° C. under nitrogen was slowly added a solution of diphenylacetic acid (1.26 g, 5.88 mmol) in THF (10.0 ml) over 2 h. Upon completion of acid addition, a thick salt suspension was agitated at 55-60° C. for another 18 h and then was allowed to cool to ambient temperature. The salt was filtered and washed with isopropyl acetate. After drying at 60° C. in a vacuum oven with nitrogen purge for 8 hours, 25 (2.97 g, 91.4%) was obtained as crystals. 1H NMR (500 MHz, DMSO-d6): δ 7.48 (qd, J=9.4, 4.9 Hz, 1H), 7.32 (d, J=7.7 Hz, 4H), 7.25-7.26 (m, 4H), 7.19-7.17 (m, 3H), 6.79 (br, 3H), 4.95 (s, 1H), 4.67 (dq, J=15.3, 9.7 Hz, 1H), 3.81-3.79 (m, 3H), 3.62 (dd, J=11.6, 6.5 Hz, 1H), 2.66-2.62 (m, 1H), 2.25 (dd, J=12.9, 6.4 Hz, 1H), 1.11 (d, J=6.5 Hz, 3H); 13C NMR (100 MHz, DMSO-d6): δ 174.4, 171.8, 156.9 (ddd, J=244, 7.0, 2.5 Hz), 149.1 (ddd, J=249, 14.4, 8.5 Hz), 147.2 (ddd, J=246, 13.9, 3.2 Hz), 141.4, 129.0, 128.5, 126.7, 125.5 (q, J=281 Hz), 118.0 (dd, J=19.8, 13.8 Hz), 117.1 (dd, J=19.2, 10.6 Hz), 112.3 (ddd, J=26.1, 6.7, 3.3 Hz), 58.5, 57.1, 51.7, 44.8 (q, J=32.7 Hz), 35.3, 27.5 (br t, J=4.6 Hz), 14.5.

(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-aminium 1H-indole-2-carboxylate (26)

    • [0186]
      Figure US20160130273A1-20160512-C00056
    • [0187]
      To a mixture of crude material containing 24 (2.00 g, 5.88 mmol) and 3,5-dichloro-2-hydroxybenzaldehyde (0.011 g, 0.059 mmol) in isopropyl acetate (15.0 ml) at 55-60° C. under nitrogen was slowly added a solution of 1H-indole-2-carboxylic acid (0.96 g, 5.88 mmol) in THF (10.0 ml) over 2 hours. Upon completion of acid addition, a thick salt suspension was agitated at 55-60° C. for another 18 h and then was allowed to cool to ambient temperature. The salt was filtered and washed with isopropyl acetate. After drying at 60° C. in a vacuum oven with nitrogen purge for 8 h, 26 (2.33 g, 79.0%) was isolated as crystals. 1H NMR (500 MHz, DMSO): δ 11.40 (s, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.45 (br, 3H), 7.47 (ddd, J=14.8, 10.1, 8.3 Hz, 1H), 7.41-7.40 (m, 1H), 7.16-7.14 (m, 2H), 6.98-6.97 (m, 1H), 6.87 (s, 1H), 4.69 (dq, J=15.3, 9.6 Hz, 1H), 3.84-3.81 (m, 4H), 2.76-2.71 (m, 1H), 2.34 (dd, J=12.7, 6.3 Hz, 1H), 1.13 (d, J=6.5 Hz, 3H); 13C NMR (100 MHz, DMSO-d6): δ 170.9, 164.8, 156.8 (ddd, J=244, 7.0, 2.5 Hz), 149.1 (ddd, J=249, 14.4, 8.5 Hz), 147.2 (ddd, J=246, 13.9, 3.2 Hz), 137.0, 133.5, 127.8, 125.4 (q, J=282 Hz), 123.3, 121.8, 119.7, 117.8 (dd, J=19.8, 13.8 Hz), 117.2 (dd, J=19.2, 10.6 Hz), 112.7, 112.3 (ddd, J=26.1, 6.7, 3.3 Hz), 105.1, 57.1, 51.3, 44.8 (q, J=32.7 Hz), 35.2, 26.9, 14.5.

N-((3S,5S,6R)-6-Methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl)-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxamide monohydrate (28)

    • [0188]
      Figure US20160130273A1-20160512-C00057
    • [0189]
      To a suspension of 23 (5.0 g, 9.1 mmol) in isopropyl acetate (50 mL) was added 5% aqueous K3PO(50 mL). The mixture was stirred for 5 min. The organic layer was separated and washed with aqueous K3PO(50 mL). Solvent removed under vacuum and resulting oil (27) was dissolved in acetonitrile (20 mL). To another flask was added 14 (2.57 g), acetonitrile (40 mL), water (20 mL) and NaOH solution (10N, 0.9 mL). The solution of 27 in acetonitrile was charged to the mixture followed by HOBT monohydrate (1.5 g) and EDC hydrochloride (2.6 g). The mixture was agitated at room temperature for 4 h and HPLC analysis indicated a complete conversion. The reaction mixture was stirred with isopropyl acetate (60 mL) and the aqueous layer was removed. The organic layer was washed with 5% aqueous NaHCO(40 mL) followed by a mixture of 15% aqueous citric acid (40 mL) and saturated aqueous NaCl (10 mL). The resulting organic layer was finally washed with 5% aqueous NaHCO(40 mL). The solvent was removed under vacuum and the residue was dissolved in methanol (20 mL). The methanol solution was slowly charged into a mixture of water (50 mL) and methanol (5 mL) over 30 min with good agitation, followed by addition of water (50 mL) over 30 min. The suspension was stirred over night at room temperature. The mixture was filtered and crystals were dried in a vacuum oven for 5 h at 50° C. to give 28 (5.4 g, 95%) as monohydrate. 1H NMR (500 MHz, CD3OD): δ 8.88 (t, J=1.2 Hz, 1H), 8.15 (t, J=1.2 Hz, 1H), 8.09 (dd, J=5.3, 1.5 Hz, 1H), 7.36 (dd, J=7.4, 1.5 Hz, 1H), 7.28 (qd, J=9.3, 4.7 Hz, 1H), 7.01 (tdd, J=9.7, 3.6, 1.9 Hz, 1H), 6.96 (dd, J=7.4, 5.3 Hz, 1H), 4.80 (dq, J=15.2, 9.2 Hz, 1H), 4.56 (dd, J=11.7, 6.8 Hz, 1H), 4.03 (ddd, J=13.6, 4.2, 2.6 Hz, 1H), 3.97-3.90 (m, 1H), 3.68 (dq, J=15.3, 8.8 Hz, 1H), 3.59 (t, J=16.2 Hz, 2H), 3.35 (d, J=4.4 Hz, 1H), 3.32 (d, J=3.5 Hz, 1H), 3.21 (qt, J=12.7, 3.1 Hz, 1H), 2.38-2.32 (m, 1H), 1.34 (d, J=6.5 Hz, 3H); 13C NMR (126 MHz, CD3OD): δ 182.79, 171.48, 168.03, 166.71, 159.37 (ddd, J=244.1, 6.5, 2.1 Hz), 157.43, 150.88 (ddd, J=249.4, 14.4, 8.7 Hz), 148.96 (ddd, J=243.8, 13.7, 3.1 Hz), 148.67, 148.15, 136.84, 133.43, 131.63, 130.83, 130.48, 126.41 (q, J=280.0 Hz), 119.85, 118.89 (dd, J=19.0, 13.5 Hz), 117.77 (dd, J=19.8, 10.8 Hz), 112.80 (ddd, J=26.5, 6.5, 4.2 Hz), 58.86, 53.67, 52.87, 46.56 (q, J=33.3 Hz), 45.18, 42.06, 36.95, 27.76 (t, J=4.8 Hz), 14.11.

Example 3 3-Hydroxy-3-(2,3,6-trifluorophenyl)butan-2-one (30)

    • [0190]
      Figure US20160130273A1-20160512-C00058
    • [0191]
      To a solution of 1,2,4-trifluorobenzene (29, 49.00 g, 371 mmol) and diisopropylamine (4.23 mL, 29.7 mmol) in THF (750 mL) at −70° C. was slowly added 2.5 M of n-BuLi (156.0 ml, 390 mmol) to maintain temperature between −45 to −40° C. The batch was agitated for 30 min. To another flask, a solution of 2,3-butadione (37.7 mL, 427 mmol) in THF (150 mL) was prepared and cooled to −70° C. The previously prepared lithium trifluorobenzene solution was transferred to the second flask between −70 to −45° C. The reaction was agitated for 1 hour at −55 to −45 and then quenched by adding AcOH (25.7 mL, 445 mmol) and then water (150 mL). After warmed to room temperature, the aqueous layer was separated. The aqueous solution was extracted with MTBE (200 mL×1) and the combined organic layers were washed with brine (100 mL×1). The organic layer was concentrated at 25-35° C. The residue was flashed with heptane (100 mL×1) and concentrated to dryness and give 30 (87.94 g, 90.2 wt %, 98% yield, and >99% HPLC purity) as an oil. 1H NMR (CDCl3, 400 MHz): δ 7.16 (m, 1H), 6.86 (m, 1H), 6.88 (s, 1H), 4.59 (s, 1H), 2.22 (s, 3H), 1.84 (dd, J=4.0, 2.8 Hz, 3H); 19F NMR (CDCl3, 376.6 MHz): δ −114.6 (dd, J=14.5, 1.4 Hz), −133.6 (d, J=19.9 Hz), −141.3 (dd, J=19.9, 14.5 Hz); 13C NMR (CDCl3, 100 MHz): δ 207.4, 156.4 (ddd, J=247, 6.2, 2.9 Hz), 149.4 (ddd, J=253, 15.0, 9.0 Hz), 147.5 (ddd, J=245, 14.4, 3.3 Hz), 119.4 (dd, J=17.3, 11.7 Hz), 117.0 (ddd, J=19.3, 11.1, 1.4 Hz), 116.6 (ddd, J=26.6, 6.5, 4.1 Hz), 77.9, 25.0 (dd, J=6.5, 4.9 Hz), 23.3.

3-(2,3,6-Trifluorophenyl)but-3-en-2-one (31)

    • [0192]
      Figure US20160130273A1-20160512-C00059
    • [0193]
      The hydroxy ketone 30 (7.69 g, 35.2 mmol) and 95% H2SO(26.2 mL, 492.8 mmol) were pumped at 2.3 and 9.2 mL/min respectively into the flow reactor. The temperature on mixing was controlled at 22-25° C. by placing the reactor in a water bath (21° C.). The effluent was quenched into a a mixture of cold water (106 g) and heptane/IPAc (1:1, 92 mL) in a jacketed reactor cooled at 0° C.; the internal temperature of the quench solution was ˜7° C. during the reaction. The layers in the quench reactor were separated and the organic layer was washed with 10% NaH2PO4/Na2HPO(1:1, 50 mL). The pH of the final wash was 5-6. Solka flock (3.85 g, 50 wt %) was added to the organic solution. The resulting slurry was concentrated and solvent-switched to heptanes at 25-30° C. The mixture was filtered, rinsed with heptanes (50 mL×1). The combined filtrates were concentrated under vacuum to give 31 as an light yellow oil (6.86 g, 90 wt %, 87% yield), which solidified in a freezer. 1H NMR (CDCl3, 400 MHz): δ 7.13 (m, 1H), 6.86 (m, 1H), 6.60 (s, 1H), 6.15 (s, 1H), 2.46 (s, 3H); 19F NMR (CDCl3, 376.6 MHz): δ −117.7 (dd, J=15.0, 1.4 Hz), −135.4 (dd, J=21.4, 1.4 Hz), −42.7 (dd, J=21.4, 15.0 Hz); 13C NMR (CDCl3, 100 MHz): δ 196.3, 155.3 (ddd, J=245, 5.1, 2.9 Hz), 147.9 (ddd, J=250, 14.5, 7.8 Hz), 147.0 (ddd, J=245, 13.4, 3.7 Hz), 137.5 (d, J=1.3 Hz), 131.7, 116.6 (ddd, J=19.9, 9.7, 1.2 Hz), 116.2 (dd, J=22.6, 16.5 Hz), 110.6 (ddd, J=24.8, 6.5, 4.1 Hz), 25.8.

Alternative Synthesis of 3-(2,3,6-trifluorophenyl)but-3-en-2-one (31)

    • [0194]
      Figure US20160130273A1-20160512-C00060
    • [0195]
      A solution of 18 (3.5 g, 18.6 mmol), acetic acid (0.34 ml, 5.58 mmol), piperidine (0.37 ml, 3.72 mmol), formaldehyde (6.0 g, 37% aqueous solution) in MeCN (20 mL) was heated over weekend. The conversion was about 60%. Reaction was heated to 70° C. overnight. The mixture was concentrated and extracted with MTBE and HCl (0.5N). The organic layer was washed with aqueous K2CO(0.5N) and water, in turns. The organic layer was concentrated. The product was isolated by chromatography column (hexane and EtOAc), yielding 31 (2.29 g, 61.5%).

Isopropyl 2-((diphenylmethylene)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (32)

    • [0196]
      Figure US20160130273A1-20160512-C00061
    • [0197]
      Diphenylidene isopropyl glycinate (2.0 g, 7.0 mmol) and 31 (1.4 g, 7.0 mmole) were dissolved in THF (10 ml). The solution was cooled to −10° C. tert-BuOLi (0.56 g, 7.0 mmole) was charged into the solution in several portions. The reaction was warmed up to room temperature slowly and stirred overnight. After quenched by addition of aqueous NH4Cl, the solvents were removed by distillation under vacuum. The residue was subjected to silica chromatography column eluted by hexane and EtOAc yielding 32 (3.0 g, 89%) as an oil, which was directly used in the next step.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

    • [0198]
      Figure US20160130273A1-20160512-C00062
    • [0199]
      Compound 32 (100 mg, 0.21 mmol) was dissolved in THF (2 ml) and the solution was cooled to −10° C. Hydrochloric acid (2N, 1 ml) was added and stirred until all starting material disappeared by TLC. The pH of the reaction was adjusted (pH.>10) by addition of aqueous K2CO3. Boc2O (68 mg, 0.31 mmole) was added into the mixture and stirred overnight. The reaction was completed checked by TLC and the product was identical to the one prepared from the iodo coupling route.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

    • [0200]
      Figure US20160130273A1-20160512-C00063
    • [0201]
      To a 100 mL round bottom was charged 2-methyl THF (43.7 mL) and diisopropyl amine (4.92 mL, 34.2 mmol) and the solution was cooled to −70° C. n-BuLi (13.08 mL, 32.7 mmol) was charged dropwise during which the temperature was controlled below −45° C. The mixture was stirred at −45° C. for 0.5 h. N-Boc-glycine ester (3.58 g) was added dropwise keeping temperature between −45 to −40° C. and aged at the same temperature for 1 h.
    • [0202]
      The solution of 31 (2.91 g, 14.5 mmol) in 2-methyl THF (2.9 mL) was then added dropwise in the same manner at −45 to −40° C. After a 0.5-1 h age, LC analysis showed nearly complete reaction. The reaction was quenched by addition of HOAc (3.83 mL) and the mixture was warmed to −10° C. and water (11.6 mL, 4 vol) was charged at <20° C. The phase was separated, and the organic layer was washed with 16% NaCl aqueous solution (11.6 mL). Assay desired product 19 as a mixture of diastereomers in the organic solution was 5.40 g (89% yield). The organic layer was concentrated to give crude product 19, which was directly used in the next step reaction. For characterization purposes, a small sample was purified by flash chromatography (silica gel, EtOAc/hexanes=1:10) to give two diastereomers 19A and 19B. 19A as a colorless oil, 1H NMR (CD3CN, 400 MHz) δ: 7.29 (m, 1H), 7.02 (m, 1H), 5.58 (d, J=6.1 Hz, 1H), 4.91 (m, 1H), 4.19-4.05 (m, 2H), 2.79 (m, 1H), 2.05 (s, 3H), 1.84 (m, 1H), 1.41 (s, 9H), 1.23 (d, J=6.7 Hz, 3H), 1.22 (d, J=6.7 Hz, 3H); 13C NMR (CD3CN, 100 MHz) δ: 204.7, 172.4, 158.6 (ddd, J=244, 6, 3 Hz), 156.3, 149.8 (ddd, J=248, 15, 9 Hz), 148.5 (ddd, J=242, 14, 3 Hz), 118.3 (dd, J=21, 16 Hz), 117.7 (ddd, J=19, 10, 2 Hz), 112.6 (ddd, J=26, 7, 4 Hz), 80.2, 70.0, 53.5, 46.0, 32.0, 28.5, 22.0, 21.9. 19B as colorless crystals, MP 91.5-92.0° C., 1H NMR (CD3CN, 400 MHz) δ: 7.31 (m, 1H), 7.03 (m, 1H), 5.61 (d, J=8.2 Hz, 1H), 4.95 (m, 1H), 4.19 (dd, J=10.2, 5.1 Hz, 1H), 3.72 (m, 1H), 2.45-2.29 (m, 2H), 2.09 (s, 3H), 1.41 (s, 9H), 1.21 (d, J=6.3 Hz, 3H), 1.20 (d, J=6.3 Hz, 3H); 13C NMR (CD3CN, 100 MHz) δ: 205.0, 172.8, 157.9 (ddd, J=244, 7, 3 Hz), 156.5, 150.3 (ddd, J=248, 149, 9 Hz), 148.5 (ddd, J=242, 13, 4 Hz), 117.9 (dd, J=19, 10 Hz), 115.9 (dd, J=21, 15 Hz), 111.5 (ddd, J=25, 8, 4 Hz), 80.1, 69.9, 52.9, 46.5, 31.1, 28.5, 22.0, 21.9.

Example 4 N-Methoxy-N-methyl-2-(o-tolyl)acetamide (34)

    • [0203]
      Figure US20160130273A1-20160512-C00064
    • [0204]
      To a solution of NHMe(OMe).HCl (203 g, 2.1 mol) in THF (1 L), H2O (400 mL) and TEA (263 g, 2.2 mol) was added 33 (200 g, 1.3 mol) and CDI (243 g, 1.5 mol) at 0-10° C. The reaction mixture was stirred at 0-10° C. for 5 h. After HPLC showed that the reaction was complete, the mixture was filtered through celite and the filtrate was partitioned with water and EtOAc. The organic solution was dried over Na2SOand concentrated. The crude residual was further purified by flash chromatography on silica gel (5-10% EtOAc/PE) to give 34 (200 g, 78% yield). 1H NMR (CDCl3, 400 MHz): δ 7.17-7.13 (m, 4H), 3.75 (m, 2H), 3.66 (d, 3H), 3.11 (s, 3H), 2.20 (s, 3H), 1.63-1.55 (m, 1H); MS (ESI) m/e [M+H]+: 194.1.

1-(o-Tolyl)propan-2-one (35)

    • [0205]
      Figure US20160130273A1-20160512-C00065
    • [0206]
      A solution of CeCl(114.4 g, 0.45 mol) in THF (4 L) was degassed for 1 h and heated to 45-50° C. for 5 h. When the solution was cooled to −10˜−5° C., MeMgCl (193.2 g, 2.6 mol) in THF was added and the mixture was stirred for 1 h at −10˜−5° C. After amide 34 (256 g, 1.3 mol) was charged into the reaction mixture at −10˜−5° C., the mixture was stirred for 5 h at 10-20° C. After the reaction was complete monitored by LCMS, the mixture was quenched by 1M HCl, and then partitioned with water and EtOAc. The organic phase was dried over Na2SOand concentrated. The crude residual was further purified by flash chromatography on silica gel (2-10% EtOAc/PE) to give 35 (157 g, 80% yield). 1H NMR (CDCl3, 400 MHz): δ 7.1-6.91 (d, 4H), 3.55 (s, 3H), 2.25 (s, 3H), 2.05 (s, 3H); MS (ESI) m/e [M+H]+: 149.05.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(o-tolyl)hexanoate (36)

  • [0207]
    Figure US20160130273A1-20160512-C00066
  • [0208]
    To a solution of 2 (181.2 g, 0.557 mol) in THF (1 L) was added TEA (84.6 g, 0.836 mol) in portions at 15-20° C. The mixture was stirred for 30 h. After the reaction was complete, the solution was concentrated to give crude 7. To a solution of 35 (82.5 g, 0.557 mol) and Cs2CO(91 g, 0.279 mol) in DMSO (1 L) was added slowly crude 7 in DMSO (500 mL) over 30 min at 15-20° C. The mixture was stirred for 1 h. After the reaction was complete, the mixture was partitioned with water and MTBE (5 L), and extracted with MTBE twice. The combined organic layer was dried over Na2SOand concentrated. The crude residual was further purified by flash chromatography on silica gel (5-10% EtOAc/PE) to give 36 (138 g, 65% yield). 1H NMR (DMSO-d6, 400 MHz): δ 7.14-7.09 (m, 3H), 7.10-6.91 (d, 1H), 4.93-4.89 (m, 1H), 4.05-3.98 (s, 3H), 2.39-2.37 (d, 3H), 1.98-1.92 (d, 3H), 1.20-1.19 (m, 9H), 1.18-1.15 (m, 6H); MS (ESI) m/e [M+H]+: 364.2
      • (S)-1′-(tert-Butyl)-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxylic acid (59)

     

    • [0249]
      Figure US20160130273A1-20160512-C00088
    • [0250]
      A mixture of 58 (5.0 g, 14.5 mmol), K2CO(5.01 g, 36.2 mmol), Pd(OAc)2(33 mg, 0.145 mmol), 1,3-bis(dicyclohexylphosphino)propane (DCPP, 127 mg, 0.290 mmol) and water (0.522 mL, 29.0 mmol) in NMP (32 mL) was heated at 120° C. under 30 psi of CO for 24 h. After cooling to room temperature, the resulting slurry was diluted with water (100 mL). The pH was slowly adjusted to 3-4 with 2 N HCl. The slurry was aged at room temperature for 1 h, filtered, rinsed with water (40 to 50 mL), dried under oven at 60° C. to give 59 (4.64 g, 95%) as a solid. 1H NMR (DMSO-d6, 500 MHz): δ 8.90 (s, 1H), 8.19 (d, J=5.2 Hz, 1H), 7.54 (d, J=7.3 Hz, 1H,), 6.99 (dd, J=7.3, 5.2 Hz, 1H), 3.33 (m, 4H), 1.72 (s, 9H); 13C NMR (DMSO-d6, 125 MHz): δ 180.16, 167.44, 166.97, 158.07, 149.76, 146.61, 135.39, 133.09, 130.36, 128.81, 125.48, 118.44, 58.19, 51.12, 44.56, 41.24, 28.91.

(S)-2′-Oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxylic acid (14)

  • [0251]
    Figure US20160130273A1-20160512-C00089
  • [0252]
    To 59 (4 g, 97% wt) was charged 37% HCl (40 to 44 mL). The slurry was heated at 94° C. for up to 48 h, cooled down to room temperature. The solvent was partially removed by reducing pressure to about total 2 vol (˜4 mL water remained). The residue was diluted with water (20 mL) followed by adjusting pH to 2.6 with NaOH (3.5 N, 4.5 mL). The thick slurry was aged for 1 to 2 h, filtered, rinsed with water (2×8 mL), followed by water/acetone (1:1, 8 mL). The wet cake was dried to give compound 14 (3.1 g, 98% wt, 94%) as crystals. 1H NMR (DMSO-d6, 500 MHz): δ 13.31 (br, 1H), 11.14 (s, 1H), 8.91 (s, 1H), 8.11 (m, 2H), 7.49 (dd, J=7.3, 1.3 Hz, 1H), 6.93 (dd, J=7.3, 5.3 Hz, 1H), 3.36 (m, 4H); 13C NMR (DMSO-d6, 125 MHz): δ 181.06, 167.36, 166.95, 156.80, 149.79, 147.32, 135.37, 133.19, 130.73, 128.88, 125.50, 118.46, 51.78, 44.12, 40.70.
PATENT
 
WO 2013169348
 

2′-oxo-l\2 5,7-tetrahydrospiro[cyclopenta[¾]pyridine-6,3′-pyrrolo[2,3-¾]pyridine]-3-carboxamide monohydrate (28)

To a suspension of 23 (5.0 g, 9.1 mmol) in isopropyl acetate (50 mL) was added 5% aqueous K3PO4 (50 mL). The mixture was stirred for 5 min. The organic layer was separated and washed with aqueous K3PO4 (50 mL). Solvent removed under vacuum and resulting oil (27) was dissolved in acetonitrile (20 mL). To another flask was added 14 (2.57 g), acetonitrile (40 mL), water (20 mL) and NaOH solution (10N, 0.9 mL). The solution of 27 in acetonitrile was

charged to the mixture followed by HOBT monohydrate (1.5 g) and EDC hydrochloride (2.6 g). The mixture was agitated at room temperature for 4 h and HPLC analysis indicated a complete conversion. The reaction mixture was stirred with isopropyl acetate (60 mL) and the aqueous layer was removed. The organic layer was washed with 5% aquoues NaHC03 (40 mL) followed by a mixture of 15% aqueous citric acid (40 mL) and saturated aqueous NaCl (10 mL). The resulting organic layer was finally washed with 5% aquous NaHC03 (40 mL). The solvent was removed under vacuum and the residue was dissolved in methanol (20 mL). The methanol solution was slowly charged into a mixture of water (50 mL) and methanol (5 mL) over 30 min with good agitation, followed by addition of water (50 mL) over 30 min. The suspension was stirred over night at room temperature. The mixture was filtered and crystals were dried in a vacuum oven for 5 h at 50 °C to give 28 (5.4 g, 95%) as monohydrate. Ή NMR (500 MHz, CD3OD): δ 8.88 (t, J= 1.2 Hz, 1 H), 8.15 (t, J = 1.2 Hz, 1 H), 8.09 (dd, J= 5.3, 1.5 Hz, 1 H), 7.36 (dd, J= 7.4, 1.5 Hz, 1 H), 7.28 (qd, J= 9.3, 4.7 Hz, 1 H), 7.01 (tdd, J= 9.7, 3.6, 1.9 Hz, 1 H), 6.96 (dd, J= 7.4, 5.3 Hz, 1 H), 4.80 (dq, J= 15.2, 9.2 Hz, 1 H), 4.56 (dd, J= 11.7, 6.8 Hz, 1 H), 4.03 (ddd, J= 13.6, 4.2, 2.6 Hz, 1 H), 3.97-3.90 (m, 1 H), 3.68 (dq, J= 15.3, 8.8 Hz, 1 H), 3.59 (t, J= 16.2 Hz, 2 H), 3.35 (d, J= 4.4 Hz, 1 H), 3.32 (d, J= 3.5 Hz, 1 H), 3.21 (qt, J= 12.7, 3.1 Hz, 1 H), 2.38-2.32 (m, 1 H), 1.34 (d, J= 6.5 Hz, 3 H); 13C NMR (126 MHz, CD3OD): δ 182.79, 171.48, 168.03, 166.71, 159.37 (ddd, J= 244.1, 6.5, 2.1 Hz), 157.43, 150.88 (ddd, J = 249.4, 14.4, 8.7 Hz), 148.96 (ddd, J= 243.8, 13.7, 3.1 Hz), 148.67, 148.15, 136.84, 133.43, 131.63, 130.83, 130.48, 126.41 (q, J = 280.0 Hz), 119.85, 118.89 (dd, J= 19.0, 13.5 Hz), 117.77 (dd, J= 19.8, 10.8 Hz), 112.80 (ddd, J= 26.5, 6.5, 4.2 Hz), 58.86, 53.67, 52.87, 46.56 (q, J = 33.3 Hz), 45.18, 42.06, 36.95, 27.76 (t, J= 4.8 Hz), 14.11.

PATENT
 
 

This invention relates to a process for making piperidinone carboxamide indane and azainane derivatives, which are CGRP receptor antagonists useful for the treatment of migraine. This class of compounds is described in U.S. Patent Application Nos. 13/293,166 filed November 10, 2011, 13/293,177 filed November 10, 2011 and 13/293,186 filed November 10, 2011, and PCT International Application Nos. PCT/US11/60081 filed November 10, 2011 and PCT/US 11/60083 filed November 10, 2011.

CGRP (Calcitonin Gene -Related Peptide) is a naturally occurring 37-amino acid peptide that is generated by tissue-specific alternate processing of calcitonin messenger RNA and is widely distributed in the central and peripheral nervous system. CGRP is localized

predominantly in sensory afferent and central neurons and mediates several biological actions, including vasodilation. CGRP is expressed in alpha- and beta-forms that vary by one and three amino acids in the rat and human, respectively. CGRP-alpha and CGRP -beta display similar biological properties. When released from the cell, CGRP initiates its biological responses by binding to specific cell surface receptors that are predominantly coupled to the activation of adenylyl cyclase. CGRP receptors have been identified and pharmacologically evaluated in several tissues and cells, including those of brain, cardiovascular, endothelial, and smooth muscle origin.

Based on pharmacological properties, these receptors are divided into at least two subtypes, denoted CGRPi and CGRP2- Human a-CGRP-(8-37), a fragment of CGRP that lacks seven N-terminal amino acid residues, is a selective antagonist of CGRPi, whereas the linear analogue of CGRP, diacetoamido methyl cysteine CGRP ([Cys(ACM)2,7]CGRP), is a selective agonist of CGRP2- CGRP is a potent neuromodulator that has been implicated in the pathology of cerebrovascular disorders such as migraine and cluster headache. In clinical studies, elevated levels of CGRP in the jugular vein were found to occur during migraine attacks (Goadsby et al, Ann. Neurol, 1990, 28, 183-187), salivary levels of CGRP are elevated in migraine subjects between attacks (Bellamy et al., Headache, 2006, 46, 24-33), and CGRP itself has been shown to trigger migrainous headache (Lassen et al., Cephalalgia, 2002, 22, 54-61). In clinical trials, the CGRP antagonist BIBN4096BS has been shown to be effective in treating acute attacks of migraine (Olesen et al, New Engl. J. Med., 2004, 350, 1104-1110) and was able to prevent headache induced by CGRP infusion in a control group (Petersen et al., Clin. Pharmacol. Ther., 2005, 77, 202-213).

CGRP -mediated activation of the trigeminovascular system may play a key role in migraine pathogenesis. Additionally, CGRP activates receptors on the smooth muscle of intracranial vessels, leading to increased vasodilation, which is thought to contribute to headache pain during migraine attacks (Lance, Headache Pathogenesis: Monoamines, Neuropeptides, Purines and Nitric Oxide, Lippincott-Raven Publishers, 1997, 3-9). The middle meningeal artery, the principle artery in the dura mater, is innervated by sensory fibers from the trigeminal ganglion which contain several neuropeptides, including CGRP. Trigeminal ganglion stimulation in the cat resulted in increased levels of CGRP, and in humans, activation of the trigeminal system caused facial flushing and increased levels of CGRP in the external jugular vein (Goadsby et al., Ann. Neurol., 1988, 23, 193-196). Electrical stimulation of the dura mater in rats increased the diameter of the middle meningeal artery, an effect that was blocked by prior administration of CGRP(8-37), a peptide CGRP antagonist (Williamson et al., Cephalalgia, 1997, 17, 525-531). Trigeminal ganglion stimulation increased facial blood flow in the rat, which was inhibited by CGRP(8-37) (Escott et al, Brain Res. 1995, 669, 93-99). Electrical stimulation of the trigeminal ganglion in marmoset produced an increase in facial blood flow that could be blocked by the non-peptide CGRP antagonist BIBN4096BS (Doods et al, Br. J.

Pharmacol., 2000, 129, 420-423). Thus the vascular effects of CGRP may be attenuated, prevented or reversed by a CGRP antagonist.

CGRP -mediated vasodilation of rat middle meningeal artery was shown to sensitize neurons of the trigeminal nucleus caudalis (Williamson et al., The CGRP Family:

Calcitonin Gene -Related Peptide (CGRP), Amylin, and Adrenomedullin, Landes Bioscience, 2000, 245-247). Similarly, distention of dural blood vessels during migraine headache may sensitize trigeminal neurons. Some of the associated symptoms of migraine, including extracranial pain and facial allodynia, may be the result of sensitized trigeminal neurons (Burstein et al, Ann. Neurol. 2000, 47, 614-624). A CGRP antagonist may be beneficial in attenuating, preventing or reversing the effects of neuronal sensitization.

The ability of the compounds to act as CGRP antagonists makes them useful pharmacological agents for disorders that involve CGRP in humans and animals, but particularly in humans. Such disorders include migraine and cluster headache (Doods, Curr Opin Inves Drugs, 2001, 2 (9), 1261-1268; Edvinsson et al, Cephalalgia, 1994, 14, 320-327); chronic tension type headache (Ashina et al, Neurology, 2000, 14, 1335-1340); pain (Yu et al, Eur. J. Pharm., 1998, 347, 275-282); chronic pain (Hulsebosch et al, Pain, 2000, 86, 163-175);

neurogenic inflammation and inflammatory pain (Holzer, Neurosci., 1988, 24, 739-768; Delay- Goyet et al, Acta Physiol. Scanda. 1992, 146, 537-538; Salmon et al, Nature Neurosci., 2001, 4(4), 357-358); eye pain (May et al. Cephalalgia, 2002, 22, 195-196), tooth pain (Awawdeh et al, Int. Endocrin. J., 2002, 35, 30-36), non-insulin dependent diabetes mellitus (Molina et al, Diabetes, 1990, 39, 260-265); vascular disorders; inflammation (Zhang et al, Pain, 2001, 89, 265), arthritis, bronchial hyperreactivity, asthma, (Foster et al, Ann. NY Acad. Sci., 1992, 657, 397-404; Schini et al, Am. J. Physiol, 1994, 267, H2483-H2490; Zheng et al, J. Virol, 1993, 67, 5786-5791); shock, sepsis (Beer et al, Crit. Care Med., 2002, 30 (8), 1794-1798); opiate withdrawal syndrome (Salmon et al, Nature Neurosci., 2001, 4(4), 357-358); morphine tolerance (Menard et al, J. Neurosci., 1996, 16 (7), 2342-2351); hot flashes in men and women (Chen et al, Lancet, 1993, 342, 49; Spetz et al, J. Urology, 2001, 166, 1720-1723); allergic dermatitis (Wallengren, Contact Dermatitis, 2000, 43 (3), 137-143); psoriasis; encephalitis, brain trauma, ischaemia, stroke, epilepsy, and neurodegenerative diseases (Rohrenbeck et al, Neurobiol. of Disease 1999, 6, 15-34); skin diseases (Geppetti and Holzer, Eds., Neurogenic Inflammation, 1996, CRC Press, Boca Raton, FL), neurogenic cutaneous redness, skin rosaceousness and erythema; tinnitus (Herzog et al, J. Membrane Biology, 2002, 189(3), 225); inflammatory bowel disease, irritable bowel syndrome, (Hoffman et al. Scandinavian Journal of Gastroenterology, 2002, 37(4) 414-422) and cystitis. Of particular importance is the acute or prophylactic treatment of headache, including migraine and cluster headache.

The present invention describes a novel process for making piperidinone carboxamide indane and azainane derivatives, which are CGRP receptor antagonists, having less steps and improved yields as compared to previous synthetic methods for making these compounds.

Another embodiment of the invention encompasses crystalline monohydrate free base of the compound having the structure

Figure imgf000011_0002

and having the following chemical name: (S)-N-((3S,5S,6R)-6-mQthyl-2-oxo-l -(2,2,2- trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl)-2′-oxo- ,2′,5,7- tetrahydrospiro [cyclopenta[b]pyridine-6,3 ‘-pyrrolo [2,3 -b]pyridine] -3 -carboxamide monohydrate

EXAMPLE 2 acetamide (17)

K2C03, water

Figure imgf000055_0002

To a solution of DMF (58.1 mL, 750 mmol) in iPAc (951 mL) was added POCl3 (55.9 mL, 600 mmol) under ice-cooling. After aged for 1 h under ice-bath, acid 16 (95 g, 500 mmol) was added under ice-cooling. The solution was stirred under ice-cooling for 30 min. The solution was added over 30 min into a solution of K2CO3 (254 g, 1.835 mol) and

NHMe(OMe)HCl (73.2 g, 750 mmol) in water (951 mL) below 8 °C. After aged for 30 min below 8 °C, the organic layer was separated, washed with water (500 mL) twice and sat. NaCl aq (100 mL) once, and concentrated in vacuo to afford 17 as an oil (117.9 g, 97.7 wt%, 99% yield). ‘H NMR (CDCI3, 400 MHz); δ 7.05 (m, 1H), 6.82 (m, 1H), 3.86 (s, 2H), 3.76 (s, 3H), 3.22 (s,

3H); 19F NMR (CDCI3, 376.6 MHz); δ -120.4 (dd, J= 15.1, 2.7 Hz), -137.9 (dd, J= 20.8, 2.7 Hz), -143.5 (dd, J= 20.8, 15.1 Hz); 13C NMR (CDC13, 100 MHz); δ 169.4, 156.9 (ddd, J= 244, 6.2, 2.7 Hz), 149.3 (ddd, J= 249, 14.4, 8.4 Hz), 147.1 (ddd, J= 244, 13.1, 3.5 Hz), 115.5 (ddd, J = 19.4, 9.9, 1.5 Hz), 133.4 (dd, J= 22.3, 16.4 Hz), 110.2 (ddd, J= 24.8, 6.7, 4.1 Hz), 32.4 (broad), 26.6 (m); HRMS m/z calcd for C10H10F3NO2 234.0736 (M+H); found 234.0746 l-(2,3,6-Trifluorophenyl)propan-2-one (18)

Figure imgf000056_0001

A mixture of CeCl3 (438 g, 1779 mmol) and THF (12 L) was heated at 40 °C for about 2 h then cooled to 5 °C. Methylmagensium chloride in THF (3 M, 3.4 L) was charged at 5- 9 °C and then it was warmed up to 16 °C and held for 1 h. The suspension was re-cooled to -10 to -15 °C. A solution of 17 (1.19 kg) in THF (2.4 L) was charged into the suspension over 15 min. After confirmation of completion of the reaction, the reaction mixture was transferred to a cold solution of hydrochloric acid (2 N, 8.4 L) and MTBE (5 L) in 5-10°C. The aqueous phase was separated and the organic layer was washed with aqueous 5%> K2CO3 (6 L) and then 10%> aqueous NaCl (5 L). The organic layer was dried over Na2S04, concentrated to give crude 18 (917g, >99wt%>) in 95% yield. The crude 18 was used in the next step without further purification. Analytically pure 18 was obtained by silica gel column.

!H NMR (CDCI3, 400 MHz); δ 7.07 (m, 1H), 6.84 (m, 1H), 3.82 (s, 2H), 2.28 (s, 3H); 19F NMR (CDCI3, 376.6 MHz); δ -120.3 (dd, J= 15.3, 2.5 Hz), -137.8 (dd, J= 21.2, 2.5 Hz), -143.0 (dd, J = 20.2, 15.3 Hz); 13C NMR (CDCI3, 100 MHz); δ 202.2, 156.5 (ddd, J= 244, 6.3, 2.9 Hz), 148.9 (ddd, J= 249, 14.4, 8.6 Hz), 147.0 (ddd, J = 244, 13.1, 3.5 Hz), 115.7 (ddd, J = 19.4, 10.5, 1.2 Hz), 112.8 (dd, J= 22.7, 17.0 Hz), 110.3 (ddd, J = 24.8, 6.7, 4.1 Hz), 37.2 (d, J=1.2 Hz), 29.3. Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

Figure imgf000057_0001

To a solution of 18 (195 g, 1.03 mol) in MTBE (1.8 L) was added zinc bromide (67 g, 0.30 mol) followed by 2 (390 g, 1.2 mol). tert-BuOLi (290 g, 3.6 mol) was then added in several portions while maintaining the reaction temperature below 40 °C. The resulting mixture was stirred at 35 °C for 24 h and quenched into a mixture of 2 N HC1 (5.6 L) and heptane (5 L) at 0 °C. The organic layer was separated and washed with 5% aqueous NaHC03 (5 L) twice. The resulting organic solution was concentrated under vacuum. The residue was dissolved in heptane (2 L) and the solution was concentrated again under vacuum. The resulting oil was dissolved in DMSO (2.5 L) and the solution was used in the next step without further purification. HPLC analysis indicated that the solution contained the desired product 19 (290 g, 67% yield) as the major component along with 5% of starting material 18. The analytically pure product 19 as one pair of diastereomers was isolated by chromatography on silica gel with ethyl acetate and heptane mixture as an eluant. HRMS: m/z calcd for C2oH26F3N05 418.1836 (M+H); found 418.1849. tert- Butyl ((55,,6i?)-6-methyl-2-oxo-5-(2,3,6-trifluorophenyl)piperidin-3-yl)carbamate (20)

Figure imgf000057_0002

To a 0.5 L cylindrical Sixfors reactor with an overhead stirring, a temperature control, a pH probe and a base addition line, was added sodiumtetraborate decahydrate (3.12 g) and DI water (163 mL). After all solids were dissolved, isopropylamine (9.63 g) was added. The pH of the buffer was adjusted to pH 10.5 using 6 N HC1. The buffer was cooled to room temperature. Then, pyridoxal-5 -phosphate (0.33 g) and SEQ ID NO: 1 (8.15 g) were added and slowly dissolved at room temperature. Crude keto ester 19 (23.6 g, 69 wt%, 16.3 g assay, 39 mmol) was dissolved in DMSO (163 mL) and the solution was added to the reactor over 5-10 min. Then the reaction was heated to 55 °C. The pH was adjusted to 10.5 according to a handheld pH meter and controlled overnight with an automated pH controller using 8 M aqueous isopropylamine. The reaction was aged for 27.5 hours.

After confirmation of >95A% conversion by HPLC, the reaction was extracted by first adding a mixture of iPA: iPAc (3:4, 350 mL) and stirring for 20 min. The phases were separated and the aqueous layer was back extracted with a mixture of iPA: iPAc (2:8, 350 mL). The phases were separated. The organic layers were combined and washed with DI water (90 mL). The HPLC based assay yield in the organic layer was 20 (9.86 g, 70.5 % assay yield) with >60:1 dr at the positions C5 and C6. tert- utyl ((35′,55′,6i?)-6-methyl-2-oxo-5-(2,3,6-trifiuorophenyl)piperidin-3-yl)carbamate (21)

Figure imgf000058_0001

A solution of crude cis and trans mixture 20 in a mixture of iPAc and iPA (1.83 wt%, 9.9 kg; 181 g assay as a mixture) was concentrated in vacuo and dissolved in 2-Me-THF (3.6 L). To the solution was added tert-BuOK (66.6 g, 0.594 mol) at room temperature. The suspension was stirred at room temperature for 2 h. The mixture was poured into water (3.5 L) and the organic layer was separated, washed with 15 wt% of aqueous NaCl (3.5 L), dried over Na2S04, and concentrated to dryness. The residue was suspended with iPAc (275 mL) and heptane (900 mL) at 60 °C. The suspension was slowly cooled down to 1 °C. The solid was filtered and rinsed with iPAc and heptane (1 :3), dried to afford 21 (166 g, 93 wt%; 85 %) as crystals. Mp 176-179 °C; 1H NMR (CDC13, 500 MHz): δ 7.06 (m, 1H), 6.84 (m, 1H), 5.83 (broad s, 1H), 5.58 (broad s, 1H), 4.22 (m, 1H), 3.88-3.79 (m, 2H), 2.77 (m, 1H), 2.25 (m, 1H), 1.46 (s, 9H), 1.08 (d, J= 6.4 Hz, 3H); 19F NMR (CDCI3, 376 MHz): δ -117 (d, J= 14 Hz), -135 (d, J= 20 Hz), -142 (dd, J= 20, 14 Hz); 13C NMR (CDC13, 100 MHz): δ 171.1, 156.6 (ddd, J = 245, 6.4, 2.8 Hz), 155.8, 149.3 (ddd, J= 248, 14.4, 8.8 Hz), 147.4 (ddd, J= 245, 14.2, 3.8 Hz), 118.0 (dd, J= 19.3, 14.5 Hz), 115.9 (dd, J= 19.2, 10.4 Hz), 111.0 (ddd, J = 26.4, 6.0, 4.3 Hz), 79.8, 51.4, 49.5, 34.1, 29.3, 28.3, 18.0; HRMS: m/z calcd for Ci7H2iF3N203 381.1396 (M+ Na); found 381.1410. tert-Butyl ((55′,6i?)-6-methyl-2-oxo-l-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3- yl)carbamate (22)

Figure imgf000059_0001

To a solution of 21 (10 g, 87% purity, 24.3 mmol) in THF (70 ml) was added tert- BuOLi (2.5 g, 31.2 mmol) at 5 °C in one portion. The solution was cooled to between 0 and 5 °C and trifluoroethyl trifluoromethanesulfonate (10.0 g, 43 mmol) was added in one portion. DMPU (7 mL) was added slowly over 15 min while maintaining the the reaction temperature below 5 °C. After the mixture was stirred at 0 °C for 3 h, additional tert-BuOLi (0.9 g, 11.2 mmol) was added. The mixture was aged for an additional 90 min. The mixture was quenched with 0.2 N HC1 (70 ml), followed by addition of heptane (80 ml). The organic layer was separated and aqueous layer extracted with heptane (30 ml). The combined organic layers were washed with 15%) aquoeus citric acid (50 mL) and 5% aqueous NaHC03 (50 mL). The solution was concentrated under vacuum at 40 °C and the resulting oil was dissolved in iPAc (30 mL). The solution was used directly in the next step without further purification. The HPLC analysis indicated that the solution contained 22 (9.8 g, 92% as cis and trans mixture in a ratio of 6.5 to 1) along with 4% of starting material 21 and 8% of a N,N’-alkylated compound. Analytically pure 22 (cis isomer) was isolated by chromatography on silica gel with ethyl acetate and heptane as an eluant. 1H NMR (CDC13, 500 MHz): δ 7.15 (m, 1H), 6.85 (m, 1H), 5.45 (broad, s, 1H), 4.90 (m, H), 4.20 (m, 1H), 3.92 (m, 2H), 3.28 (m, 1H), 2.70 (m, 2H), 1.48 (s, 9H), 1.20 (d, J= 5.9 Hz, 3H); 13C NMR (CDC13, 100 MHz): δ 170.2, 156.9 (ddd, J= 245, 6.3,2.7 Hz), 156.0, 149.6 (ddd, J= 251, 14.8, 8.8 Hz), 147.6 (ddd, J= 246, 13.9,3.6 Hz), 124.5 (q, J= 281 Hz), 117.6 (dd, J = 19.2, 3.7 Hz), 116.4 (dd, J= 19.1, 10.4 Hz), 111.4 (ddd, J= 25.8, 6.4,4.1Hz), 56.6, 52.8, 45.3 (q, J= 34.2 Hz), 35.2, 28.7, 28.3 (br t, J= 4 Hz), 14.6; HRMS: m/z calcd for Ci9H22F6N203 (M+H): 441.1607; found 441.1617. (35′,55′,6i?)-6-Methyl-2-oxo-l-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperi (S)-2-acetamido-3 -phenylpropanoate (23)

Figure imgf000060_0001

iPAc solution of 22 (529 g assayed, 1.2 mol), obtained from previous step, was diluted to 6 L with iPAc, /?-toluenesulfonic acid monohydride (343 g, 1.8 mol) was added and the solution was heated to 55 °C. After 4 h, the reaction completed (>99% conversion). Aqueous K2CO3 (530 g in 3 L of water) was charged into the solution after cooled to 15-25 °C. The aqueous layer was separated and was back-extracted with iPAc (2 L). The iPAc solutions were combined and the total volume was adjudted to 10 L by adding iPAc. The solution was heated to 50-60 °C. About 20 g of N-acetyl L-phenylalanine was added and the solution was agitated for 15 min or until solids precipitated out. The remaining N-acetyl L-phenylalanine (total 250 g, 1.2 mol) was charged slowly and 2-hydroxy-5-nitrobenzaldehyde (2 g) was charged. The suspension was agitated for 12 h at 20 °C and then cooled to 0 °C for 3 h. The suspension was filtrated, washed with iPAc three times and dried to give 23 (583g, 89% yield) as crystals. Mp 188 – 190 °C; 1H NMR (DMSO-de, 400 MHz): δ 7.96 (d, J= 8.0 Hz, 1H) , 7.48 (m, 1H), 7.15-7.25 (m, 6H), 4.65 (ddd, J= 19.4, 15.3, 9.6 Hz, 1H), 4.33 (ddd, J= 8.7, 8.4, 4.9 Hz, 1H), 3.70-3.87 (m, 3H), 3.57 (dd, J= 11.5, 6.6 Hz, 1H), 3.04 (dd, J= 13.7, 4.9 Hz, 1H), 2.82 (dd, J= 13.7, 8.9 Hz,lH), 2.59 (m, 1H), 2.24 (m, 1H), 2.95 (s, 3H), 1.10 (d, J= 6.4 Hz, 1H); 19F NMR (DMSO-d6, 376 MHz): δ -69 (s) , -118 (d, J= 15 Hz), -137 (d, J = 21 Hz), -142 (dd, J= 21, 15 Hz); 13C NMR (DMSO-d6, 100 MHz): δ 173.6, 171,. l, 168.7, 156.3 (ddd, J= 243.5, 7.0, 3.1 Hz), 148.7 (ddd, J= 249, 14.4, 9.1 Hz), 146.8 (ddd, J = 245, 13.7, 3.1 Hz), 138.5, 129.2, 128.0, 126.1, 124.9 (q, J= 280.9 Hz), 117.4.0 (dd, J= 19.3, 13.8 Hz), 116.7 (dd, J= 19.3, 10.6 Hz), 111.8 (ddd, J= 26.0, 6.7, 3.6 Hz), 56.6, 54.3, 51,2, 44.3 (q, J= 32.5 Hz), 37.2, 34.8, 26.9 (br t, J= 4 Hz), 22.5, 14.1.

(35′,55′,6i?)-6-methyl-2-oxo-l-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3- aminium 2,2-diphenylacetate (25)

Figure imgf000061_0001

To a mixture of crude material containing (55′,6i?)-3-amino-6-methyl-l -(2,2,2- trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one (24, 2.00 g, 5.88 mmol), prepared according to the same method as the previous example, and 3,5-dichloro-2-hydroxybenzaldehyde (0.011 g, 0.059 mmol) in isopropyl acetate (15.0 ml) at 55-60 °C under nitrogen was slowly added a solution of diphenylacetic acid (1.26 g, 5.88 mmol) in THF (10.0 ml) over 2 h. Upon completion of acid addition, a thick salt suspension was agitated at 55-60 °C for another 18 h and then was allowed to cool to ambient temperature. The salt was filtered and washed with isopropyl acetate. After drying at 60 °C in a vacuum oven with nitrogen purge for 8 hours, 25 (2.97 g, 91.4%) was obtained as crystals. 1H NMR (500 MHz, DMSO-d6): δ 7.48 (qd, J= 9.4, 4.9 Hz, 1 H), 7.32 (d, J= 7.7 Hz, 4 H), 7.25-7.26 (m, 4 H), 7.19-7.17 (m, 3 H), 6.79 (br, 3H), 4.95 (s, 1 H), 4.67 (dq, J= 15.3, 9.7 Hz, 1 H), 3.81-3.79 (m, 3 H), 3.62 (dd, J= 11.6, 6.5 Hz, 1 H), 2.66-2.62 (m, 1 H), 2.25 (dd, J= 12.9, 6.4 Hz, 1 H), 1.11 (d, J= 6.5 Hz, 3 H); 13C NMR (100 MHz, DMSO-de): δ 174.4, 171.8, 156.9 (ddd, J= 244, 7.0, 2.5 Hz), 149.1 (ddd, J= 249, 14.4, 8.5 Hz), 147.2 (ddd, J= 246, 13.9, 3.2 Hz), 141.4, 129.0, 128.5, 126.7, 125.5 (q, J= 281 Hz), 118.0 (dd, J= 19.8, 13.8 Hz), 117.1 (dd, J= 19.2, 10.6 Hz), 112.3 (ddd, J= 26.1, 6.7, 3.3 Hz), 58.5, 57.1, 51.7, 44.8 (q, J= 32.7 Hz), 35.3, 27.5 (br t, J= 4.6 Hz), 14.5.

(35′,55′,6i?)-6-methyl-2-oxo-l-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-amM lH-indole-2-carboxylate (26)

Figure imgf000061_0002

To a mixture of crude material containing 24 (2.00 g, 5.88 mmol) and 3,5-dichloro-2- hydroxybenzaldehyde (0.011 g, 0.059 mmol) in isopropyl acetate (15.0 ml) at 55-60 °C under nitrogen was slowly added a solution of lH-indole-2-carboxylic acid (0.96 g, 5.88 mmol) in THF (10.0 ml) over 2 hours. Upon completion of acid addition, a thick salt suspension was agitated at 55-60 °C for another 18 h and then was allowed to cool to ambient temperature. The salt was filtered and washed with isopropyl acetate. After drying at 60 °C in a vacuum oven with nitrogen purge for 8 h, 26 (2.33 g, 79.0%) was isolated as crystals. 1H NMR (500 MHz, DMSO): δ 11.40 (s, 1 H), 7.56 (d, J= 8.0 Hz, 1 H), 7.45 (br, 3 H), 7.47 (ddd, J= 14.8, 10.1, 8.3 Hz, 1 H), 7.41- 7.40 (m, 1 H), 7.16-7.14 (m, 2 H), 6.98-6.97 (m, 1 H), 6.87 (s, 1 H), 4.69 (dq, J= 15.3, 9.6 Hz, 1 H), 3.84-3.81 (m, 4 H), 2.76-2.71 (m, 1 H), 2.34 (dd, J= 12.7, 6.3 Hz, 1 H), 1.13 (d, J= 6.5 Hz, 3 H); 13C NMR (100 MHz, DMSO-d6): δ 170.9, 164.8, 156.8 (ddd, J= 244, 7.0, 2.5 Hz), 149.1 (ddd, J= 249, 14.4, 8.5 Hz), 147.2 (ddd, J = 246, 13.9, 3.2 Hz), 137.0, 133.5, 127.8, 125.4 (q, J = 282 Hz), 123.3, 121.8, 119.7, 117.8 (dd, J= 19.8, 13.8 Hz), 117.2 (dd, J= 19.2, 10.6 Hz), 112.7, 112.3 (ddd, J= 26.1, 6.7, 3.3 Hz), 105.1, 57.1, 51.3, 44.8 (q, J= 32.7 Hz), 35.2, 26.9, 14.5.

Figure imgf000062_0001

2′-oxo-l\2 5,7-tetrahydrospiro[cyclopenta[¾]pyridine-6,3′-pyrrolo[2,3-¾]pyridine]-3- carboxamide monohydrate (28)

Figure imgf000062_0002

To a suspension of 23 (5.0 g, 9.1 mmol) in isopropyl acetate (50 mL) was added 5% aqueous K3PO4 (50 mL). The mixture was stirred for 5 min. The organic layer was separated and washed with aqueous K3PO4 (50 mL). Solvent removed under vacuum and resulting oil (27) was dissolved in acetonitrile (20 mL). To another flask was added 14 (2.57 g), acetonitrile (40 mL), water (20 mL) and NaOH solution (10N, 0.9 mL). The solution of 27 in acetonitrile was charged to the mixture followed by HOBT monohydrate (1.5 g) and EDC hydrochloride (2.6 g). The mixture was agitated at room temperature for 4 h and HPLC analysis indicated a complete conversion. The reaction mixture was stirred with isopropyl acetate (60 mL) and the aqueous layer was removed. The organic layer was washed with 5% aquoues NaHC03 (40 mL) followed by a mixture of 15% aqueous citric acid (40 mL) and saturated aqueous NaCl (10 mL). The resulting organic layer was finally washed with 5% aquous NaHC03 (40 mL). The solvent was removed under vacuum and the residue was dissolved in methanol (20 mL). The methanol solution was slowly charged into a mixture of water (50 mL) and methanol (5 mL) over 30 min with good agitation, followed by addition of water (50 mL) over 30 min. The suspension was stirred over night at room temperature. The mixture was filtered and crystals were dried in a vacuum oven for 5 h at 50 °C to give 28 (5.4 g, 95%) as monohydrate. Ή NMR (500 MHz, CD3OD): δ 8.88 (t, J= 1.2 Hz, 1 H), 8.15 (t, J = 1.2 Hz, 1 H), 8.09 (dd, J= 5.3, 1.5 Hz, 1 H), 7.36 (dd, J= 7.4, 1.5 Hz, 1 H), 7.28 (qd, J= 9.3, 4.7 Hz, 1 H), 7.01 (tdd, J= 9.7, 3.6, 1.9 Hz, 1 H), 6.96 (dd, J= 7.4, 5.3 Hz, 1 H), 4.80 (dq, J= 15.2, 9.2 Hz, 1 H), 4.56 (dd, J= 11.7, 6.8 Hz, 1 H), 4.03 (ddd, J= 13.6, 4.2, 2.6 Hz, 1 H), 3.97-3.90 (m, 1 H), 3.68 (dq, J= 15.3, 8.8 Hz, 1 H), 3.59 (t, J= 16.2 Hz, 2 H), 3.35 (d, J= 4.4 Hz, 1 H), 3.32 (d, J= 3.5 Hz, 1 H), 3.21 (qt, J= 12.7, 3.1 Hz, 1 H), 2.38-2.32 (m, 1 H), 1.34 (d, J= 6.5 Hz, 3 H); 13C NMR (126 MHz, CD3OD): δ 182.79, 171.48, 168.03, 166.71, 159.37 (ddd, J= 244.1, 6.5, 2.1 Hz), 157.43, 150.88 (ddd, J = 249.4, 14.4, 8.7 Hz), 148.96 (ddd, J= 243.8, 13.7, 3.1 Hz), 148.67, 148.15, 136.84, 133.43, 131.63, 130.83, 130.48, 126.41 (q, J = 280.0 Hz), 119.85, 118.89 (dd, J= 19.0, 13.5 Hz), 117.77 (dd, J= 19.8, 10.8 Hz), 112.80 (ddd, J= 26.5, 6.5, 4.2 Hz), 58.86, 53.67, 52.87, 46.56 (q, J = 33.3 Hz), 45.18, 42.06, 36.95, 27.76 (t, J= 4.8 Hz), 14.11.

EXAMPLE 3

3-Hydroxy-3-(2,3,6-trifluorophenyl)butan-2-one (30)

Figure imgf000063_0001

To a solution of 1,2,4-trifluorobenzene (29, 49.00 g, 371 mmol) and diisopropylamine (4.23 mL, 29.7 mmol) in THF (750 mL) at -70 °C was slowly added 2.5 M of ft-BuLi (156.0 ml, 390 mmol) to maintain temperature between -45 to -40 °C. The batch was agitated for 30 min. To another flask, a solution of 2,3-butadione (37.7 mL, 427 mmol) in THF (150 mL) was prepared and cooled to -70 °C. The previously prepared lithium trifluorobenzene solution was transferred to the second flask between -70 to -45 °C. The reaction was agitated for 1 hour at -55 to -45 and then quenched by adding AcOH (25.7 mL, 445 mmol) and then water (150 mL). After warmed to room temperature, the aqueous layer was seperated. The aqueous solution was extracted with MTBE (200 mL x 1) and the combined organic layers were washed with brine (100 mL x 1). The organic layer was concentrated at 25-35 °C. The residue was flashed with heptane (100 mL x 1) and concentrated to dryness and give 30 (87.94 g, 90.2 wt%, 98% yield, and >99% HPLC purity) as an oil. H NMR (CDCI3, 400 MHz): δ 7.16 (m, 1H), 6.86 (m, 1H), 6.88 (s, 1H), 4.59 (s, 1H), 2.22 (s, 3H), 1.84 (dd, J= 4.0, 2.8 Hz, 3H); 19F NMR (CDCI3, 376.6 MHz): δ -114.6 (dd, J= 14.5, 1.4 Hz), -133.6 (d, J= 19.9 Hz), -141.3 (dd, J =

19.9, 14.5 Hz); 13C NMR (CDCI3, 100 MHz): δ 207.4, 156.4 (ddd, J= 247, 6.2, 2.9 Hz), 149.4 (ddd, J= 253, 15.0, 9.0 Hz), 147.5 (ddd, J= 245, 14.4, 3.3 Hz), 119.4 (dd, J=17.3, 11.7 Hz), 117.0 (ddd, J=19.3, 11.1, 1.4 Hz), 116.6 (ddd, J= 26.6, 6.5, 4.1 Hz), 77.9, 25.0 (dd, J= 6.5, 4.9 Hz), 23.3. -(2,3,6-Trifluorophenyl)but-3-en-2-one (31)

Figure imgf000064_0001

The hydroxy ketone 30 (7.69 g, 35.2 mmol) and 95% H2S04 (26.2 mL, 492.8 mmol) were pumped at 2.3 and 9.2 mL/min respectively into the flow reactor. The temperature on mixing was controlled at 22-25 °C by placing the reactor in a water bath (21 °C). The effluent was quenched into a a mixture of cold water ( 106 g) and heptane/IP Ac ( 1 : 1 , 92 mL) in a j acketed reactor cooled at 0 °C; the internal temperature of the quench solution was ~ 7 °C during the reaction. The layers in the quench reactor were separated and the organic layer was washed with 10% NaH2P04/Na2HP04 (1 :1, 50 mL). The pH of the final wash was 5-6. Solka flock (3.85 g, 50 wt%>) was added to the organic solution. The resulting slurry was concentrated and solvent- switched to heptanes at 25-30 °C. The mixture was filtered, rinsed with heptanes (50 mL x 1). The combined filtrates were concentrated under vacuum to give 31 as an light yellow oil (6.86 g, 90 wt%, 87% yield), which solidified in a freezer. *H NMR (CDC13, 400 MHz): δ 7.13 (m, 1H), 6.86 (m, 1H), 6.60 (s, 1H), 6.15 (s, 1H), 2.46 (s, 3H); 19F NMR (CDC13, 376.6 MHz): δ -117.7 (dd, J= 15.0, 1.4 Hz), -135.4 (dd, J= 21.4, 1.4 Hz), -42.7 (dd, J= 21.4, 15.0 Hz); 13C NMR (CDCls, 100 MHz): δ 196.3, 155.3 (ddd, J= 245, 5.1, 2.9 Hz), 147.9 (ddd, J= 250, 14.5, 7.8 Hz), 147.0 (ddd, J = 245, 13.4, 3.7 Hz), 137.5 (d, J=1.3 Hz), 131.7, 116.6 (ddd, J= 19.9, 9.7, 1.2 Hz), 116.2 (dd, J= 22.6, 16.5 Hz), 110.6 (ddd, J= 24.8, 6.5, 4.1 Hz), 25.8.

Alternative synthesis of 3-(2,3,6-trifluorophenyl)but-3-en-2-one (31)

Figure imgf000065_0001

A solution of 18 (3.5 g, 18.6 mmol), acetic acid (0.34 ml, 5.58 mmol), piperidine (0.37 ml, 3.72 mmol), formaldehyde (6.0 g, 37%> aqueous solution) in MeCN (20 mL) was heated over weekend. The conversion was about 60%. Reaction was heated to 70 °C overnight. The mixtrure was concentrated and extracted with MTBE and HC1 (0.5N). The organic layer was washed with aqueous K2CO3 (0.5N) and water, in turns. The organic layer was concentrated. The product was isolated by chromatography column (hexane and EtOAc), yielding 31 (2.29 g, 61.5%).

Isopropyl 2-((diphenylmethylene)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (32)

Figure imgf000065_0002

Diphenylidene isopropyl glycinate (2.0 g, 7.0 mmol) and 31 (1.4 g, 7.0 mmole) were dissolved in THF (10 ml). The solution was cooled to -10 °C. tert- uOLi (0.56 g, 7.0 mmole) was charged into the solution in several portions. The reaction was warmed up to room temperature slowly and stirred overnight. After quenched by addition of aqueous NH4CI, the solvents were removed by distillation under vacuum. The residue was subjected to silica chromatography column eluted by hexane and EtOAc yielding 32 (3.0 g, 89 %) as an oil, which was directly used in the next step.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

Figure imgf000066_0001

Compound 32 (100 mg, 0.21 mmol) was dissolved in THF (2 ml) and the solution was cooled to -10 °C. Hydrochloric acid (2N, 1 ml) was added and stirred until all starting material disappeared by TLC. The pH of the reaction was adjusted (pH.>10) by addition of aqueous K2CO3. Boc20 (68 mg, 0.31 mmole) was added into the mixture and stirred overnight. The reaction was completed checked by TLC and the product was identical to the one prepared from the iodo coupling route.

Isopropyl 2-((tert-butoxycarbonyl)amino)-5-oxo-4-(2,3,6-trifluorophenyl)hexanoate (19)

Figure imgf000066_0002

To a 100 mL round bottom was charged 2-methyl THF (43.7 mL) and diisopropyl amine (4.92 mL, 34.2 mmol) and the solution was cooled to -70 °C. n-BuLi (13.08 mL, 32.7 mmol) was charged dropwise during which the temperature was controlled below -45 °C. The mixture was stirred at -45 °C for 0.5 h. N-Boc-glycine ester (3.58 g) was added dropwise keeping temperature between -45 to -40 °C and aged at the same temperature for 1 h.

The solution of 31 (2.91 g, 14.5 mmol) in 2-methyl THF (2.9 mL) was then added dropwise in the same manner at -45 to -40 °C. After a 0.5-1 h age, LC analysis showed nearly complete reaction. The reaction was quenched by addition of HO Ac (3.83 mL) and the mixture was warmed to -10 °C and water (1 1.6 mL, 4 vol) was charged at < 20 °C. The phase was separated, and the organic layer was washed with 16% NaCl aqueous solution (11.6 mL). Assay desired product 19 as a mixture of diastereomers in the organic solution was 5.40 g (89% yield). The organic layer was concentrated to give crude product 19, which was directly used in the next step reaction. For characterization purposes, a small sample was purified by flash chromatography (silica gel, EtOAc/hexanes = 1 : 10) to give two diastereomers 19A and 19B. 19A as a colorless oil, 1H NMR (CD3CN, 400 MHz) δ: 7.29 (m, 1 H), 7.02 (m, 1 H), 5.58 (d, J = 6.1 Hz, 1 H), 4.91 (m, 1 H), 4.19-4.05 (m, 2 H), 2.79 (m, 1 H), 2.05 (s, 3 H), 1.84 (m, 1 H), 1.41 (s, 9 H), 1.23 (d, J = 6.7 Hz, 3 H), 1.22 (d, J = 6.7 Hz, 3 H); 13C NMR (CD3CN, 100 MHz) δ: 204.7, 172.4, 158.6 (ddd, J = 244, 6, 3 Hz), 156.3, 149.8 (ddd, J = 248, 15, 9 Hz), 148.5 (ddd, J = 242, 14, 3 Hz), 118.3 (dd, J = 21, 16 Hz), 117.7 (ddd, J = 19, 10, 2 Hz), 112.6 (ddd, J = 26, 7, 4 Hz), 80.2, 70.0, 53.5, 46.0, 32.0, 28.5, 22.0, 21.9. 19B as colorless crystals, MP 91.5-92.0 °C, 1H NMR (CD3CN, 400 MHz) δ: 7.31 (m, 1 H), 7.03 (m, 1 H), 5.61 (d, J = 8.2 Hz, 1 H), 4.95 (m, 1 H), 4.19 (dd, J = 10.2, 5.1 Hz, 1 H), 3.72 (m, 1 H), 2.45-2.29 (m, 2 H), 2.09 (s, 3 H), 1.41 (s, 9 H), 1.21 (d, J = 6.3 Hz, 3 H), 1.20 (d, J = 6.3 Hz, 3 H); 13C NMR (CD3CN, 100 MHz) δ: 205.0, 172.8, 157.9 (ddd, J= 244, 7, 3 Hz), 156.5, 150.3 (ddd, J= 248, 149, 9 Hz), 148.5 (ddd, J = 242, 13, 4 Hz), 117.9 (dd, J = 19, 10 Hz), 115.9 (dd, J = 21, 15 Hz), 111.5 (ddd, J = 25, 8, 4 Hz), 80.1, 69.9, 52.9, 46.5, 31.1, 28.5, 22.0, 21.9.

 

PATENT

https://encrypted.google.com/patents/US20120122911

[0000]

Figure US20120122911A1-20120517-C00039

(3S,5S,6R)-3-Amino-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one hydrochlorideStep A: (5S,6R & 5R,6S)-6-Methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one

Essentially following the procedures described in Intermediate 14, but using 2,3,6-trifluorophenylboronic acid in place of 2,3,5-trifluorophenylboronic acid, the title compound was obtained. MS: m/z=326.0 (M+1).

Step B: (3S,5S,6R & 3R,5R,6S)-3-Azido-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one

To a stirred solution of lithium bis(trimethylsilyl)amide (1.0 M in THF, 4.80 mL, 4.80 mmol) in THF (20 mL) at −78° C. was added a cold (−78° C.) solution of (5S,6R & 5R,6S)-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one (1.30 g, 4.00 mmol) in THF (10 mL) dropwise, keeping the internal temperature of the reaction mixture below −65° C. The resulting mixture was stirred at −78° C. for 30 min, then a cold (−78° C.) solution of 2,4,6-triisopropylbenzenesulfonyl azide (Harmon et al. (1973) J. Org. Chem. 38, 11-16) (1.61 g, 5.20 mmol) in THF (10 mL) was added dropwise, keeping the internal temperature of the reaction mixture below −65° C. The reaction mixture was stirred at −78° C. for 30 min, then AcOH (1.05 mL, 18.4 mmol) was added. The resulting mixture was allowed to warm slowly to ambient temperature and was poured into saturated aqueous sodium bicarbonate (50 mL) and the mixture was extracted with EtOAc (2×75 mL). The combined organic layers were washed with brine, then dried over sodium sulfate, filtered, and concentrated to dryness in vacuo. The crude product was purified by silica gel chromatography, eluting with a gradient of hexanes:EtOAc—100:0 to 20:80, to give the diastereomeric azide products (3R,5S,6R & 3S,5R,6S)-3-azido-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,5-trifluorophenyl)piperidin-2-one, which eluted second, and the title compound, which eluted first. MS: m/z=367.1 (M+1).

Step C: tent-Butyl [(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]carbamate

To a solution of (3S,5S,6R & 3R,5R,6S)-3-azido-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,5-trifluorophenyl)piperidin-2-one (280 mg, 0.764 mmol) and di-tert-butyl dicarbonate (217 mg, 0.994 mmol) in EtOH (5 mL) was added 10% palladium on carbon (25 mg, 0.024 mmol) and the resulting mixture was stirred vigorously under an atmosphere of hydrogen (ca. 1 atm) for 1 h. The reaction mixture was filtered through a pad of Celite®, washing with EtOH, and the filtrate was concentrated in vacuo to give a crude solid. The crude product was purified by silica gel chromatography, eluting with a gradient of hexanes:EtOAc—100:0 to 30:70, to give the racemic title compound. Separation of the enantiomers was achieved by SFC on a ChiralTech IC column, eluting with CO2:MeOH:CH3CN—90:6.6:3.3, to give tert-butyl [(3R,5R,6S)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]carbamate as the first major peak, and tert-butyl [(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]carbamate, the title compound, as the second major peak. MS: m/z=463.2 (M+Na).

Step D: (3S,5S,6R)-3-Amino-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one hydrochloride

A solution of tert-butyl [(3S,5S,6R)-6-methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]carbamate (122 mg, 0.277 mmol) in EtOAc (10 mL) was saturated with HCl (g) and aged for 30 min. The resulting mixture was concentrated in vacuo to give the title compound. MS: m/z=341.1 (M+1); 1H NMR (500 MHz, CD3OD) δ 7.33 (qd, 1H, J=9.3, 4.9 Hz), 7.05 (tdd, 1H, J=9.8, 3.7, 2.2 Hz), 4.78 (dq, 1H, J=15.4, 9.3 Hz), 4.22 (dd, 1H, J=12.2, 6.6 Hz), 4.06 (ddd, 1H, J=13.3, 4.5, 2.7 Hz), 3.97 (m, 1H), 3.73 (dq, 1H, J=15.4, 8.8 Hz), 2.91 (qt, 1H, J=12.7, 3.1 Hz), 2.36 (ddd, 1H, J=12.7, 6.4, 2.0 Hz), 1.22 (d, 3H, J=6.6 Hz).

Example 4

Figure US20120122911A1-20120517-C00047

(6S)—N-[(3S,5S,6R)-6-Methyl-2-oxo-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-3-yl]-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxamide dihydrochloride

To a stirred mixture of (6S)-2′-oxo-1′,2′,5,7-tetrahydrospiro[cyclopenta[b]pyridine-6,3′-pyrrolo[2,3-b]pyridine]-3-carboxylic acid (described in Intermediate 1) (264 mg, 0.939 mmol), (3S,5S,6R)-3-amino-6-methyl-1-(2,2,2-trifluoroethyl)-5-(2,3,6-trifluorophenyl)piperidin-2-one hydrochloride (described in Intermediate 15) (295 mg, 0.782 mmol), HOBT (144 mg, 0.939 mmol), and EDC (180 mg, 0.939 mmol) in DMF (8 mL) was added N,N-diisopropylethylamine (0.34 mL, 1.96 mmol), and the resulting mixture was stirred at ambient temperature for 3 h. The reaction mixture was then poured into saturated aqueous sodium bicarbonate (30 mL) and extracted with EtOAc (2×40 mL). The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by silica gel chromatography, eluting with a gradient of CH2Cl2:MeOH:NH4OH—100:0:0 to 90:10:0.1, to give the product, which was treated with HCl in EtOAc at 0° C. to afford the title compound. HRMS: m/z=604.1783 (M+1), calculated m/z=604.1778 for C29H24F6N5O31H NMR (500 MHz, CD3OD) δ 9.09 (s, 1H), 8.69 (s, 1H), 8.18 (dd, 1H, J=5.9, 1.5 Hz), 7.89 (dd, 1H, J=7.3, 1.5 Hz), 7.30 (m, 1H), 7.23 (dd, 1H, J=7.3, 5.9 Hz), 7.03 (m, 1H), 4.78 (m, 1H), 4.61 (dd, 1H, J=11.5, 6.6 Hz), 4.05 (dd, 1H, J=13.8, 2.8 Hz), 3.96 (m, 1H), 3.84 (d, 1H, J=18.6 Hz), 3.76 (d, 1H, J=18.6 Hz), 3.73 (d, 1H, J=17.3 Hz), 3.72 (m, 1H), 3.61 (d, 1H, J=17.3 Hz), 3.22 (m, 1H), 2.38 (m, 1H), 1.34 (d, 3H, J=6.6 Hz).

 

Publication numberPriority datePublication dateAssigneeTitle
US9487523B22012-03-142016-11-08Merck Sharp & Dohme Corp.Process for making CGRP receptor antagonists
Family To Family Citations
CA2865941A12012-03-142013-09-19Merck Sharp & Dohme Corp.Bis-quarternary cinchona alkaloid salts as asymmetric phase transfer catalysts
CN105037210A *2015-05-272015-11-11江苏大学Alpha,beta-dehydrogenated-alpha-amino acid synthesis method
GB201519194D02015-10-302015-12-16Heptares Therapeutics LtdCGRP receptor antagonists
GB201519195D02015-10-302015-12-16Heptares Therapeutics LtdCGRP Receptor Antagonists
GB201519196D02015-10-302015-12-16Heptares Therapeutics LtdCGRP Receptor Antagonists
PATENT 
Cited Patent Filing date Publication date Applicant Title
US7390798 * Feb 9, 2005 Jun 24, 2008 Merck & Co., Inc. Carboxamide spirolactam CGRP receptor antagonists
US20090054408 * Sep 6, 2005 Feb 26, 2009 Bell Ian M Monocyclic anilide spirolactam cgrp receptor antagonists
US20100160334 * Mar 5, 2010 Jun 24, 2010 Bell Ian M Tricyclic anilide spirolactam cgrp receptor antagonists
US20100179166 * Jun 2, 2008 Jul 15, 2010 Ian Bell Carboxamide heterocyclic cgrp receptor antagonists
US20120122899 * Nov 10, 2011 May 17, 2012 Merck Sharp & Dohme Corp. Piperidinone carboxamide azaindane cgrp receptor antagonists
US20120122900 * Nov 10, 2011 May 17, 2012 Merck Sharp & Dohme Corp. Piperidinone carboxamide azaindane cgrp receptor antagonists
US20120122911 * Nov 10, 2011 May 17, 2012 Merck Sharp & Dohme Corp. Piperidinone carboxamide azaindane cgrp receptor antagonists
Reference
1 * See also references of EP2849568A4
Citing Patent Filing date Publication date Applicant Title
CN105037210A * May 27, 2015 Nov 11, 2015 江苏大学 Alpha,beta-dehydrogenated-alpha-amino acid synthesis method
US9688660 Oct 28, 2016 Jun 27, 2017 Heptares Therapeutics Limited CGRP receptor antagonists
US9802935 Oct 28, 2016 Oct 31, 2017 Heptares Therapeutics Limited CGRP receptor antagonists
US9808457 Oct 28, 2016 Nov 7, 2017 Heptares Therapeutics Limited CGRP receptor antagonists
 
Patent ID

 

Patent Title

 

Submitted Date

 

Granted Date

 

US2016346214 TABLET FORMULATION FOR CGRP ACTIVE COMPOUNDS
2015-01-30
 
US9850246 Process for Making CGRP Receptor Antagonists
2015-09-15
2016-05-12
US9499545 PIPERIDINONE CARBOXAMIDE AZAINDANE CGRP RECEPTOR ANTAGONISTS
2014-09-12
2015-01-01
US9487523 PROCESS FOR MAKING CGRP RECEPTOR ANTAGONISTS
2013-09-19
2015-02-05
US9174989 Process for making CGRP receptor antagonists
2013-03-12
2015-11-03
 
Patent ID

 

Patent Title

 

Submitted Date

 

Granted Date

 

US8481556 Piperidinone carboxamide azaindane CGRP receptor antagonists
2011-11-10
2013-07-09
US8754096 Piperidinone carboxamide azaindane CGRP receptor antagonists
2011-11-10
2014-06-17
US8912210 Piperidinone carboxamide azaindane CGRP receptor antagonists
2011-11-10
2014-12-16
US2017027925 PIPERIDINONE CARBOXAMIDE AZAINDANE CGRP RECEPTOR ANTAGONISTS
2016-10-14
 
US2016346198 NOVEL DISINTEGRATION SYSTEMS FOR PHARMACEUTICAL DOSAGE FORMS
2015-02-04
 

////////////////Atogepant, атогепант أتوجيبانت 阿托吉泮 , PHASE 3, MERCK, ALLERGAN, 

CC1C(CC(C(=O)N1CC(F)(F)F)NC(=O)C2=CC3=C(CC4(C3)C5=C(NC4=O)N=CC=C5)N=C2)C6=C(C=CC(=C6F)F)F

wdt-2

NEW DRUG APPROVALS

ONE TIME

$10.00

TRILACICLIB, G1T28


ChemSpider 2D Image | Trilaciclib | C24H30N8OTrilaciclib.png

Trilaciclib

update 2021/2/12 US FDA APPROVED COSELA

  • Molecular FormulaC24H30N8O
  • Average mass446.548 Da
  • G1T 28
CAS 1374743-00-6
2′-{[5-(4-Methyl-1-piperazinyl)-2-pyridinyl]amino}-7′,8′-dihydro-6’H-spiro[cyclohexane-1,9′-pyrazino[1′,2′:1,5]pyrrolo[2,3-d]pyrimidin]-6′-one
G1T28, SHR 6390
Spiro[cyclohexane-1,9′(6’H)-pyrazino[1′,2′:1,5]pyrrolo[2,3-d]pyrimidin]-6′-one, 7′,8′-dihydro-2′-[[5-(4-methyl-1-piperazinyl)-2-pyridinyl]amino]-
  • 7′,8′-Dihydro-2′-[[5-(4-methyl-1-piperazinyl)-2-pyridinyl]amino]spiro[cyclohexane-1,9′(6’H)-pyrazino[1′,2′:1,5]pyrrolo[2,3-d]pyrimidin]-6′-one
  • 2′-[[5-(4-Methylpiperazin-1-yl)pyridin-2-yl]amino}-7′,8′-dihydro-6’H-spiro[cyclohexane-1,9′-pyrazino[1′,2′:1,5]pyrrolo[2,3-d]pyrimidin]-6′-one
UNII:U6072DO9XG

Reduction of Chemotherapy-Induced Myelosuppression

Trilaciclib dihydrochloride
1977495-97-8

2D chemical structure of 1977495-97-8

In phase II clinical development as a chemoprotectant at G1 Therapeutics for first- or second-line treatment in patients with metastatic triple negative breast cancer, in combination with gemcitabine and carboplatin

logo

PATENT, WO 2014144326Compound 89 (also referred to as Compound T)

WO2014144847A3
Inventors Norman E. SharplessJay Copeland StrumJohn Emerson BisiPatrick Joseph RobertsFrancis Xavier Tavares
Applicant G1 Therapeutics, Inc.
Norman Sharpless
Norman Sharpless official photo.jpg
Born Norman Edward Sharpless
September 20, 1966 (age 51)
Greensboro, North Carolina
Nationality American
Other names Ned Sharpless
Occupation Director, Lineberger Comprehensive Cancer Center Founder, G1 Therapeutics ($GTHX)
Notable work Wellcome Distinguished Professor, American Society of Clinical Investigation Member, Association of American Cancer Institute board of directors,

NCI Director Dr. Norman E. SharplessPinterest

NCI Director Dr. Norman E. Sharpless, Credit: National Institutes of Health

Norman E. “Ned” Sharpless, M.D., was officially sworn in as the 15th director of the National Cancer Institute (NCI) on October 17, 2017. Prior to his appointment, Dr. Sharpless served as the director of the University of North Carolina (UNC) Lineberger Comprehensive Cancer Center, a position he held since January 2014.

Dr. Sharpless was a Morehead Scholar at UNC–Chapel Hill and received his undergraduate degree in mathematics. He went on to pursue his medical degree from the UNC School of Medicine, graduating with honors and distinction in 1993. He then completed his internal medicine residency at the Massachusetts General Hospital and a hematology/oncology fellowship at Dana-Farber/Partners Cancer Care, both of Harvard Medical School in Boston.

After 2 years on the faculty at Harvard Medical School, he joined the faculty of the UNC School of Medicine in the Departments of Medicine and Genetics in 2002. He became the Wellcome Professor of Cancer Research at UNC in 2012.

Dr. Sharpless is a member of the Association of American Physicians as well as the American Society for Clinical Investigation (ASCI), the nation’s oldest honor society for physician–scientists, and served on the ASCI council from 2011 to 2014. Dr. Sharpless was an associate editor of Aging Cell and deputy editor of the Journal of Clinical Investigation. He has authored more than 150 original scientific papers, reviews, and book chapters, and is an inventor on 10 patents. He cofounded two clinical-stage biotechnology companies: G1 Therapeutics and HealthSpan Diagnostics.

In addition to serving as director of NCI, Dr. Sharpless continues his research in understanding the biology of the aging process that promotes the conversion of normal self-renewing cells into dysfunctional cancer cells. Dr. Sharpless has made seminal contributions to the understanding of the relationship between aging and cancer, and in the preclinical development of novel therapeutics for melanoma, lung cancer, and breast cancer.

Record ID Title Status Phase
NCT03041311 CarboplatinEtoposide, and Atezolizumab With or Without Trilaciclib (G1T28), a CDK 4/6 Inhibitor, in Extensive Stage Small Cell Lung Cancer (SCLC) Recruiting 2
NCT02978716 Trilaciclib (G1T28), a CDK 4/6 Inhibitor, in Combination With Gemcitabineand Carboplatin in Metastatic Triple Negative Breast Cancer (mTNBC) Recruiting 2
NCT02514447 Trilaciclib (G1T28), a CDK 4/6 Inhibitor, in Patients With Previously Treated Extensive Stage SCLC Receiving Topotecan Chemotherapy Recruiting 2
NCT02499770 Trilaciclib (G1T28), a CDK 4/6 Inhibitor, in Combination With Etoposide and Carboplatin in Extensive Stage Small Cell Lung Cancer (SCLC) Active, not recruiting 2

Synthesis

WO  2016040858

Trilaciclib (G1T28)

Trilaciclib is a potential first-in-class short-acting CDK4/6 inhibitor in development to preserve hematopoietic stem cells and enhance immune system function during chemotherapy. Trilaciclib is administered intravenously prior to chemotherapy and has the potential to significantly improve treatment outcomes.

G1 is currently evaluating trilaciclib in four Phase 2 clinical trials: three studies in patients with small-cell lung cancer (SCLC), and one study in patients with triple-negative breast cancer (TNBC). Preliminary data from the SCLC trials were presented at the American Society of Clinical Oncology 2017 Annual Meeting and at the 2016 World Conference on Lung Cancer.

Data from a Phase 1 trial in healthy volunteers were presented at the American Society of Clinical Oncology 2015 Annual Meeting and published in Science Translational Medicine. Trilacicilib has been extensively studied in animals; these preclinical data have been presented at several scientific meetings and published in Molecular Cancer Therapeutics, Science Translational Medicine, and Cancer Discovery.

Trilaciclib is a small molecule, competitive inhibitor of cyclin dependent kinases 4 and 6 (CDK4/6), with potential antineoplastic and chemoprotective activities. Upon intravenous administration, trilaciclib binds to and inhibits the activity of CDK4/6, thereby blocking the phosphorylation of the retinoblastoma protein (Rb) in early G1. This prevents G1/S phase transition, causes cell cycle arrest in the G1 phase, induces apoptosis, and inhibits the proliferation of CDK4/6-overexpressing tumor cells. In patients with CDK4/6-independent tumor cells, G1T28 may protect against multi-lineage chemotherapy-induced myelosuppression (CIM) by transiently and reversibly inducing G1 cell cycle arrest in hematopoietic stem and progenitor cells (HSPCs) and preventing transition to the S phase. This protects all hematopoietic lineages, including red blood cells, platelets, neutrophils and lymphocytes, from the DNA-damaging effects of certain chemotherapeutics and preserves the function of the bone marrow and the immune system. CDKs are serine/threonine kinases involved in the regulation of the cell cycle and may be overexpressed in certain cancer cell types. HSPCs are dependent upon CDK4/6 for proliferation.

Trilaciclib (G1T28) is a CDK4/6 inhibitor in phase II clinical development as a chemoprotectant at G1 Therapeutics for first- or second-line treatment in patients with metastatic triple negative breast cancer, in combination with gemcitabine and carboplatin. Also, phase II trials are ongoing in newly diagnosed, treatment-naive small-cell lung cancer patients, in combination with carboplatin, etoposide, and atezolizumab and phase I trials in previously treated small-cell lung cancer patients, in combination with topotecan.

U.S. Patent Nos. 8,822,683; 8,598,197; 8,598,186, 8,691,830, 8,829,102, 8,822,683, 9, 102,682, 9,499,564, 9,481,591, and 9,260,442, filed by Tavares and Strum and assigned to Gl Therapeutics describe a class of N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amine cyclin dependent kinase inhibitors including those of the formula with variables as defined therein):

U.S. Patent Nos. 9,464,092, 9,487,530, and 9,527,857 which are also assigned to Gl Therapeutics describe the use of the above pyrimidine-based agents in the treatment of cancer.

These patents provide a general synthesis of the compounds that is based on a coupling reaction of a fused chloropyrimidine with a heteroaryl amine to form the central disubstituted amine. Such coupling reactions are sometimes referred to as Buchwald coupling (see WO Ί56 paragraph 127; reference WO 2010/020675). The lactam of the fused chloropyrimidine, for example, a 2-chloro-spirocyclo-pyrrolo[2,3-d]pyrimidine-one such as Intermediate K as shown below can be prepared by dehydration of the corresponding carboxylic acid. The reported process to prepare intermediate IK requires seven steps.


(Intermediate IK; page 60, paragraph 215 of WO Ί56)

WO 2013/148748 (U.S. S.N. 61/617,657) entitled “Lactam Kinase Inhibitors” filed by Tavares, and also assigned to Gl Therapeutics likewise describes the synthesis of N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amines via the coupling reaction of a fused chloropyrimidine with a heteroaryl amine to form the central disubstituted amine.

WO 2013/163239 (U.S. S.N. 61/638,491) “Synthesis of Lactams” describes a method for the synthesis of this class of compounds with the variation that in the lactam preparation step, a carboxylic acid can be cyclized with a protected amine in the presence of a strong acid and a dehydrating agent, which can be together in one moiety as a strong acid anhydride. The purported improvement is that cyclization can occur without losing the protecting group on the amine before cyclization. The typical leaving group is “tBOC” (t-butoxycarbonyl). The application teaches (page 2 of WO 2013/163239) that the strong acid is, for example, trifluoroacetic acid anhydride, tribromoacetic acid anhydride, trichloroacetic acid anhydride or mixed anhydrides. An additional step may be necessary to take off the N-protecting group. The dehydrating agent can be a carbodiimide-based compound such as DCC (Ν,Ν-dicyclohexylcarbodiimide), EDC (l-ethyl-3-(3-dimethylaminopropyl)carbodiimide, or DIC (Ν,Ν-diisopropylcarbodiimide). DCC and DIC are in the same class of reagents-carbodiimides. DIC is sometimes considered better because it is a liquid at room temperature, which facilitates reactions.

WO 2015/061407 filed by Tavares and licensed to Gl Therapeutics also describes the synthesis of these compounds via the coupling of a fused chloropyrimidine with a heteroaryl amine to form the central disubstituted amine. WO ‘407 focuses on the lactam production step and in particular describes that the fused lactams of these compounds can be prepared by treating the carboxylic acid with an acid and a dehydrating agent in a manner that a leaving group on the amine is not removed during the amide-forming ring closing step.

Other publications that describe compounds of this general class include the following. WO 2014/144326 filed by Strum et al. and assigned to Gl Therapeutics describes compounds and methods for protection of normal cells during chemotherapy using pyrimidine based CDK4/6 inhibitors. WO 2014/144596 filed by Strum et al. and assigned to Gl Therapeutics describes compounds and methods for protection of hematopoietic stem and progenitor cells against ionizing radiation using pyrimidine based CDK4/6 inhibitors. WO 2014/144847 filed by Strum et al. and assigned to Gl Therapeutics describes HSPC-sparing treatments of abnormal cellular proliferation using pyrimidine based CDK4/6 inhibitors. WO2014/144740 filed by Strum et al. and assigned to Gl Therapeutics describes highly active anti -neoplastic and anti-proliferative pyrimidine based CDK 4/6 inhibitors. WO 2015/161285 filed by Strum et al. and assigned to Gl Therapeutics describes tricyclic pyrimidine based CDK inhibitors for use in radioprotection. WO 2015/161287 filed by Strum et al. and assigned to Gl Therapeutics describes analogous tricyclic pyrimidine based CDK inhibitors for the protection of cells during chemotherapy. WO 2015/161283 filed by Strum et al. and assigned to Gl Therapeutics describes analogous tricyclic pyrimidine based CDK inhibitors for use in HSPC-sparing treatments of RB-positive abnormal cellular proliferation. WO 2015/161288 filed by Strum et al. and assigned to Gl Therapeutics describes analogous tricyclic pyrimidine based CDK inhibitors for use as anti -neoplastic and anti-proliferative agents. WO 2016/040858 filed by Strum et al. and assigned to Gl Therapeutics describes the use of combinations of pyrimidine based CDK4/6 inhibitors with other anti-neoplastic agents. WO 2016/040848 filed by Strum et al. and assigned to Gl Therapeutics describes compounds and methods for treating certain Rb-negative cancers with CDK4/6 inhibitors and topoisomerase inhibitors.

Other biologically active fused spirolactams and their syntheses are described, for example, in the following publications. Griffith, D. A., et al. (2013). “Spirolactam-Based Acetyl-CoA Carboxylase Inhibitors: Toward Improved Metabolic Stability of a Chromanone Lead Structure.” Journal of Medicinal Chemistry 56(17): 7110-7119, describes metabolically stable spirolactams wherein the lactam resides on the fused ring for the inhibition of acetyl-CoA carboxylase. WO 2013/169574 filed by Bell et al. describes aliphatic spirolactams as CGRP receptor antagonists wherein the lactam resides on the spiro ring. WO 2007/061677 filed by Bell et al. describes aryl spirolactams as CGRP receptor antagonists wherein the lactam resides on the spiro ring. WO 2008/073251 filed by Bell et al. describes constrained spirolactam compounds wherein the lactam resides on the spiro ring as CGRP receptor antagonists. WO 2006/031606 filed by Bell et al. describes carboxamide spirolactam compounds wherein the spirolactam resides on the spiro ring as CGRP receptor antagonists. WO 2006/031610, WO 2006/031491, and WO 2006/029153 filed by Bell et al. describe anilide spirolactam compounds wherein the spirolactam resides on the spiro ring; WO 2008/109464 filed by Bhunai et al. describes spirolactam compounds wherein the lactam resides on the spiro ring which is optionally further fused.

Given the therapeutic activity of selected N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amines, it would be useful to have additional methods for their preparation. It would also be useful to have new intermediates that can be used to prepare this class of compounds.

PATENT

WO 2014144596

PATENT

WO 2014144326

Compound 89 (also referred to as Compound T)

WO2014144847A3
Inventors Norman E. SharplessJay Copeland StrumJohn Emerson BisiPatrick Joseph RobertsFrancis Xavier Tavares
Applicant G1 Therapeutics, Inc.

EXAMPLES

Intermediates B, E, K, L, 1A, IF and 1CA were synthesized according to US 8,598,186 entitled CDK Inhibitors to Tavares, F.X. and Strum, J.C..

The patents WO 2013/148748 entitled Lactam Kinase Inhibitors to Tavares, F.X., WO 2013/163239 entitled Synthesis of Lactams to Tavares, F.X., and US 8,598,186 entitled CDK Inhibitors to Tavares, F.X. and Strum, J.C. are incorporated by reference herein in their entirety. Example 1

Synthesis of tert-butyl N- [2- [(5-bromo-2-chloro-pyrimidin-4yl)amino] ethyl] carbamate, Compound 1

Figure imgf000106_0001

To a solution of 5-bromo-2,4-dichloropyrimidine (3.2 g, 0.0135 mol) in ethanol (80 mL) was added Hunig’s base (3.0 mL) followed by the addition of a solution of N-(tert- butoxycarbonyl)-l,2-diaminoethane (2.5 g, 0.0156 mole) in ethanol (20 mL). The contents were stirred overnight for 20 hrs. The solvent was evaporated under vacuum. Ethyl acetate (200 mL) and water (100 mL) were added and the layers separated. The organic layer was dried with magnesium sulfate and then concentrated under vacuum. Column chromatography on silica gel using hexane/ethyl acetate (0- 60%) afforded tert-butyl N-[2-[(5-bromo-2-chloro-pyrimidin-4- yl)amino]ethyl]carbamate. 1HNMR (d6-DMSO) δ ppm 8.21 (s, 1H), 7.62 (brs, 1H), 7.27 (brs, 1H), 3.39 (m, 2H), 3.12 (m, 2H), 1.34 (s, 9H). LCMS (ESI) 351 (M + H).

Example 2

Synthesis of tert-butyl N-[2-[[2-chloro-5-(3,3-diethoxyprop-l-ynyl)pyrimidin-4- yl] amino] ethyl] carbamate, Compound 2

Figure imgf000106_0002

To tert-butyl N-[2-[(5-bromo-2-chloro-pyrimidin-4-yl)amino]ethyl]carbamate (1.265 g, 6 mmol) in THF (10 mL) was added the acetal (0.778 mL, 5.43 mmol), Pd(dppf)CH2Cl2 (148 g), and triethylamine (0.757 mL, 5.43 mmol). The contents were degassed and then purged with nitrogen. To this was then added Cul (29 mg). The reaction mixture was heated at reflux for 48 hrs. After cooling, the contents were filtered over CELITE™ and concentrated. Column chromatography of the resulting residue using hexane/ethyl acetate (0- 30%) afforded tert-butyl N- [2- [ [2-chloro-5 -(3 ,3 -diethoxyprop- 1 -ynyl)pyrimidin-4-yl]amino] ethyl] carbamate. 1HNMR (d6-DMSO) δ ppm 8.18 (s, 1H), 7.63 (brs, 1H), 7.40 (brs, 1H), 5.55 (s, 1H), 3.70 (m, 2H), 3.60 (m, 2H), 3.42 (m, 2H), 3.15 (m, 2H), 1.19 – 1.16 (m, 15H). LCMS (ESI) 399 (M + H).

Example 3

Synthesis of tert-butyl N-[2-[2-chloro-6-(diethoxymethyl)pyrrolo[2,3-d]pyrimidin-7- yl] ethyl] carbamate, Compound 3

Figure imgf000107_0001

To a solution of the coupled product (2.1 g, 0.00526 mole) in THF (30 mL) was added TBAF solid (7.0 g). The contents were heated to and maintained at 65 degrees for 2 hrs. Concentration followed by column chromatography using ethyl acetate/hexane (0-50%) afforded tert-butyl N-[2-[2-chloro-6-(diethoxymethyl)pyrrolo[2,3-d]pyrimidin-7-yl]ethyl]carbamate as a pale brown liquid (1.1 g). 1FiNMR (d6-DMSO) δ ppm 8.88 (s, 1H), 6.95 (brs, 1H), 6.69 (s, 1H), 5.79 (s, 1H), 4.29 (m, 2H), 3.59 (m, 4H), 3.34 (m, 1H), 3.18 (m, 1H), 1.19 (m, 9H), 1.17 (m, 6H). LCMS (ESI) 399 (M + H).

Example 4

Synthesis of tert-buty\ N-[2-(2-chloro-6-formyl-pyrrolo [2,3-d] pyrimidin-7- yl)ethyl] carbamate, Compound 4

Figure imgf000108_0001

To the acetal (900 mg) from the preceeding step was added AcOH (8.0 mL) and water

(1.0 mL). The reaction was stirred at room temperature for 16 hrs. Cone, and column chromatography over silica gel using ethyl acetate/hexanes (0- 60%) afforded tert-butyl N-[2-(2- chloro-6-formyl-pyrrolo[2,3-d]pyrimidin-7-yl)ethyl]carbamate as a foam (0.510 g). 1HNMR (d6-DMSO) δ ppm 9.98 (s, 1H), 9.18 (s, 1H), 7.66 (s, 1H), 6.80 (brs, 1H), 4.52 (m, 2H), 4.36 (m, 2H), 1.14 (s, 9H). LCMS (ESI) 325 (M + H).

Example 5

Synthesis of 7- [2-(teri-butoxycarbonylamino)ethyl] -2-chloro-pyrrolo [2,3-d] pyrimidine-6- carboxylic acid, Compound 5

Figure imgf000108_0002

To the aldehyde (0.940 g) from the preceeding step in DMF (4 mL) was added oxone (1.95 g, 1.1 eq). The contents were stirred at room temp for 7 hrs. Silica gel column chromatography using hexane/ethyl acetate (0- 100%) afforded l-\2-(tert- butoxycarbonylamino)ethyl]-2-chloro-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid (0.545 g). 1HNMR (d6-DMSO) δ ppm 9.11 (s, 1H), 7.39 (s, 1H), 4.38 (m, 2H), 4.15 (m, 2H), 1.48 (m, 9H). LCMS (ESI) 341(M + H).

Example 6

Synthesis of methyl 7-[2-(teri-butoxycarbonylamino)ethyl]-2-chloro-pyrrolo[2,3- d]pyrimidine-6-carboxylate, Compound 6

Figure imgf000109_0001

To a solution of 2-chloro-7-propyl-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid (0.545 g, 0.00156 mole) from the preceeding step in toluene (3.5 mL) and MeOH (1 mL) was added TMS- diazomethane (1.2 mL). After stirring overnight at room temperature, the excess of TMS- diazomethane was quenched with acetic acid (3 mL) and the reaction was concentrated under vacuum. The residue was purified by silica gel column chromatography with hexane/ethyl acetate (0- 70%) to afford methyl 7-[2-(tert-butoxycarbonylamino)ethyl]-2-chloro-pyrrolo[2,3- d]pyrimidine-6-carboxylate as an off white solid (0.52 g). 1HNMR (d6-DMSO) δ ppm 9.10 (s, 1H), 7.45 (s, 1H), 6.81 (brs, 1H) 4.60 (m, 2H), 3.91 (s, 3H), 3.29 (m, 2H), 1.18 (m, 9H) LCMS (ESI) 355 (M + H).

Example 7

Synthesis of Chloro tricyclic amide, Compound 7

Figure imgf000109_0002

To methyl 7- [2-(tert-butoxycarbonylamino)ethyl] -2-chloro-pyrrolo [2,3 -d]pyrimidine-6- carboxylate (0.50 g, 0.0014 mole) from the preceeding step in dichloromethane (2.0 mL) was added TFA (0.830 mL). The contents were stirred at room temperature for 1 hr. Concentration under vacuum afforded the crude amino ester which was suspended in toluene (5 mL) and Hunig’s base (0.5 mL). The contents were heated at reflux for 2 hrs. Concentration followed by silica gel column chromatography using hexane/ethyl acetate (0- 50%) afforded the desired chloro tricyclic amide (0.260 g). 1HNMR (d6-DMSO) δ ppm 9.08 (s, 1H), 8.48 (brs, 1H), 7.21 (s, 1H) 4.33 (m, 2H), 3.64 (m, 2H). LCMS (ESI) 223 (M + H).

Example 8

Synthesis of chloro-N-methyltricyclic amide, Compound 8

Figure imgf000110_0001

To a solution of the chloro tricycliclactam, Compound 7, (185 mg, 0.00083 mole) in DMF (2.0 mL) was added sodium hydride (55% dispersion in oil, 52 mg). After stirring for 15 mins, methyl iodide (62 μί, 1.2 eq). The contents were stirred at room temperature for 30 mins. After the addition of methanol (5 mL), sat NaHCOs was added followed by the addition of ethyl acetate. Separation of the organic layer followed by drying with magnesium sulfate and concentration under vacuum afforded the N-methylated amide in quantitative yield. 1FiNMR (d6-DMSO) δ ppm 9.05 (s, 1H), 7.17 (s, 1H) 4.38 (m, 2H), 3.80 (m, 2H), 3.05 (s, 3H). LCMS (ESI) 237 (M + H). Example 9

Synthesis of l-methyl-4-(6-nitro-3-pyridyl)piperazine, Compound 9

Figure imgf000110_0002

To 5-bromo-2-nitropyridine (4.93 g, 24.3 mmole) in DMF (20 mL) was added N- methylpiperazine (2.96 g, 1.1 eq) followed by the addition of DIPEA (4.65 mL, 26.7 mmole). The contents were heated at 90 degrees for 24 hrs. After addition of ethyl acetate (200 mL), water (100 mL) was added and the layers separated. Drying followed by concentration afforded the crude product which was purified by silica gel column chromatography using (0-10%) DCM/Methanol. 1HNMR (d6-DMSO) δ ppm 8.26 (s, 1H), 8.15 (1H, d, J = 9.3 Hz), 7.49 (1H, d, J = 9.4 Hz), 3.50 (m, 4H), 2.49 (m, 4H), 2.22 (s, 3H).

Example 10

Synthesis of 5-(4-methylpiperazin-l-yl)pyridin-2-amine, Compound 10

Figure imgf000111_0001

To l-methyl-4-(6-nitro-3-pyridyl)piperazine (3.4 g) in ethyl acetate (100 mL) and ethanol (100 mL) was added 10%> Pd/C (400 mg) and then the reaction was stirred under hydrogen (10 psi) overnight. After filtration through CELITE™, the solvents were evaporated and the crude product was purified by silica gel column chromatography using DCM/ 7N ammonia in MeOH (0- 5%) to afford 5-(4-methylpiperazin-l-yl)pyridin-2-amine (2.2 g). 1HNMR (d6-DMSO) δ ppm 7.56 (1H, d, J = 3 Hz), 7.13 (1H, m), 6.36 (1H, d, J = 8.8 Hz), 5.33 (brs, 2H), 2.88 (m, 4H), 2.47 (m, 4H), 2.16 (s, 3H).

Example 11

Synthesis of tert-butyl 4-(6-amino-3-pyridyl)piperazine-l-carboxylate, Compound 11

Figure imgf000111_0002

This compound was prepared as described in WO 2010/020675 Al .

Synthesis of Compound 89 (also referred to as Compound T)

Figure imgf000169_0002

Compound 89 was synthesized in a similar manner to that described for compound 78 and was converted to an HCl salt. 1HNMR (600 MHz, DMSO-d6) δ ppm 1.47 (br. s., 6 H) 1.72 (br. s., 2 H) 1.92 (br. s., 2 H) 2.77 (br. s., 3 H) 3.18 (br. s., 2 H) 3.46 (br. s., 2 H) 3.63 (br. s., 2 H) 3.66 (d, J=6.15 Hz, 2 H) 3.80 (br. s., 2 H) 7.25 (s, 1 H) 7.63 (br. s., 2 H) 7.94 (br. s., 1 H) 8.10 (br. s., 1 H) 8.39 (br. s., 1 H) 9.08 (br. s., 1 H) 11.59 (br. s., 1 H). LCMS (ESI) 447 (M + H)

PATENT

WO 2014144740

PATENT

WO 2016040858

Preparation of Active Compounds

Syntheses

The disclosed compounds can be made by the following general schemes:

Scheme 1

In Scheme 1, Ref-1 is WO 2010/020675 Al; Ref-2 is White, J. D.; et al. J. Org. Chem. 1995, 60, 3600; and Ref-3 Presser, A. and Hufher, A. Monatshefte fir Chemie 2004, 135, 1015.

Scheme 2

In Scheme 2, Ref-1 is WO 2010/020675 Al; Ref-4 is WO 2005/040166 Al; and Ref-5 is Schoenauer, K and Zbiral, E. Tetrahedron Letters 1983, 24, 573.

92

93 

3) Pd/C/H2 

Scheme 6

Scheme 7

NHfOH

Scheme 8

In Scheme 8, Ref-1 is WO 2010/020675 Al; Ref-2 is WO 2005/040166 Al; and Ref-3 is Schoenauer, K and Zbiral, E. Tetrahedron Letters 1983, 24, 573.

Alternatively, the lactam can be generated by reacting the carboxylic acid with a protected amine in the presence of a strong acid and a dehydrating agent, which can be together in one moiety as a strong acid anhydride. Examples of strong acid anhydrides include, but are not limited to, trifluoroacetic acid anhydride, tribromoacetic acid anhydride, trichloroacetic acid anhydride, or mixed anhydrides. The dehydrating agent can be a carbodiimide based compound such as but not limited to DCC (Ν,Ν-dicyclohexylcarbodiimide), EDC (l-ethyl-3-(3-

dimethylaminopropyl)carbodiimide or DIC (Ν,Ν-diisopropylcarbodiimide). An additional step may be necessary to take off the N-protecting group and the methodologies are known to those skilled in the art.

Alternatively, the halogen moiety bonded to the pyrimidine ring can be substituted with any leaving group that can be displaced by a primary amine, for example to create an intermediate for a final product such as Br, I, F, SMe, SO2Me, SOalkyl, SO2alkyl. See, for Exmaple PCT /US2013/037878 to Tavares.

Other amine intermediates and final amine compounds can be synthesized by those skilled in the art. It will be appreciated that the chemistry can employ reagents that comprise reactive functionalities that can be protected and de-protected and will be known to those skilled in the art at the time of the invention. See for example, Greene, T.W. and Wuts, P.G.M., Greene’s Protective Groups in Organic Synthesis, 4th edition, John Wiley and Sons.

Scheme 9

CDK4/6 Inhibitors of the present invention can be synthesized according to the generalized Scheme 9. Specific synthesis and characterization of the Substituted 2-aminopyrmidines can be found in, for instance, WO2012/061156.

Compounds T, Q, GG, and U were prepared as above and were characterized by mass spectrometry and NMR as shown below:

Compound T

1H NMR (600 MHz, DMSO- d6) ppm 1.47 (br. s., 6 H) 1.72 (br. s., 2 H) 1.92 (br. s., 2 H) 2.77 (br. s., 3 H) 3.18 (br. s., 2 H) 3.46 (br. s., 2 H) 3.63 (br. s., 2 H) 3.66 (d, J=6.15 Hz, 2 H) 3.80 (br. s., 2 H) 7.25 (s, 1 H) 7.63 (br. s., 2 H) 7.94 (br. s., 1 H) 8.10 (br. s., 1 H) 8.39 (br. s., 1 H) 9.08 (br. s., 1 H) 11.59 (br. s., 1 H). LCMS ESI (M + H) 447.

PATENT

WO-2018005865

Synthesis of N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amines. The application appears to be particularly focused on methods for the preparation of trilaciclib and an analog of it. Trilaciclib is the company’s lead CDK4/6 inhibitor presently in phase II trials against small-cell lung cancer and triple negative breast cancer. Interestingly, the company is working on a second CDK4/6 inhibitor, G1T38 , which is in a phase II trial against breast cancer.

GENERAL METHODS

The structure of starting materials, intermediates, and final products was confirmed by standard analytical techniques, including NMR spectroscopy and mass spectrometry. Unless otherwise noted, reagents and solvents were used as received from commercial suppliers. Proton nuclear magnetic resonance spectra were obtained on a Bruker AVANCE 500 at 500 MHz in DMSO-dis. HPLC analyses were performed on a Waters HPLC using the below HPLC method.

HPLC Method

Column: Atlantis T3 (150 χ 4.6, 3 μιη)

Column Temperature: 40°C

Flow Rate: 1 mL/min

Detection: UV @ 275 nm

Analysis Time: 36 min

Mobile Phase A: Water (with 0.1% Trifluoroacetic Acid)

Mobile Phase B : Acetonitrile (with 0.1% Trifluoroacetic Acid)

Sample preparation: dissolve PC sample, wet or dry solid (~1 mg of active compound) in acetonitrile/water (1/1) to achieve complete dissolution.

HPLC Method Gradient

Example 1. General Routes of Synthesis

Scheme 1-1 : Starting from an appropriately substituted halo pyrimidine, compounds of the present invention can be prepared. In Step 1 the appropriately substituted halo pyrimidine is subjected to l,4-diazaspiro[5.5]undecan-3-one in the presence of base and heat to afford a substituted spirolactam. In Step 2 the appropriately substituted spirolactam is protected with a group selected from R2. In Step 3 the protected spirolactam is cyclized in the presence of base to afford a fused spirolactam. The fused spirolactam can be optionally oxidized to a sulfoxide or sulfone after Step 3, Step 4, Step 5, or Step 6. Oxidation prior to Step 3 results in undesired byproducts. In Step 4 the hydroxyl group of the fused spirolactam is converted to a leaving group.

In Step 5 the leaving group is dehydrated to afford a compound of Formula IV. In Step 6 the compound of Formula IV is optionally deprotected.

Scheme 1-2: Starting from an appropriately substituted halo pyrimidine compounds of the present invention can be prepared. In Step 1 the appropriately substituted halo pyrimidine is subjected to l,4-diazaspiro[5.5]undecan-3-one in the presence of base and heat to afford a substituted spirolactam. In Step 2 the appropriately substituted spirolactam is protected with a group selected from R2. In Step 3 the protected spirolactam is cyclized in the presence of base to afford a fused spirolactam of Formula IV. The fused spirolactam can be optionally oxidized to a sulfoxide or sulfone after Step 3 or Step 4. Oxidation prior to Step 3 results in undesired byproducts. In Step 4 the compound of Formula IV is optionally deprotected.

Scheme 1-3 : Starting from an appropriately substituted alkyl glycinate, compounds of the present invention can be prepared. In Step 1 the appropriately substituted alkyl glycinate is subjected to cyclohexanone and TMSCN in the presence of base to afford a cyano species. In Step 2 the appropriately substituted cyanospecies is reduced and subsequently cyclized to afford a compound of Formula I.

Scheme 1-4

Scheme 1-4: Starting from an appropriately substituted l-(aminomethyl)cyclohexan-l-amine, compounds of the present invention can be prepared. In Step 1 the appropriately substituted l-(aminomethyl)cyclohexan-l -amine is reductively aminated with an aldehyde. In Step 2 the appropriately substituted cyclohexane amine is optionally deprotected (i.e.: the group selected from R2 if not H is optionally replaced by H). In Step 3 the cyclohexane amine is cyclized to afford a compound of Formula I. In Step 4 the compound of Formula I is optionally protected.

1-5

Conversion

Scheme 1-5: Starting from an appropriately substituted halo pyrimidine, compounds of the present invention can be prepared. In Step 1 the appropriately substituted halo pyrimidine is subjected to l,4-diazaspiro[5.5]undecan-3-one in the presence of base and heat to afford a

substituted spirolactam. In Step 2 the protected spirolactam is cyclized in the presence of base to afford a fused spirolactam. The fused spirolactam can be optionally oxidized to a sulfoxide or sulfone after Step 2, Step 3, Step 4, or Step 5. Oxidation prior to Step 2 results in undesired byproducts. In Step 3 the hydroxyl group of the fused spirolactam is converted to a leaving group. In Step 4 the leaving group is dehydrated to afford a compound of Formula IV. In Step 5 the compound of Formula IV is optionally deprotected.

S

Scheme 1-6: Starting from an appropriately substituted halo pyrimidine compounds of the present invention can be prepared. In Step 1 the appropriately substituted halo pyrimidine is subjected to l,4-diazaspiro[5.5]undecan-3-one in the presence of base and heat to afford a substituted spirolactam. In Step 2 the protected spirolactam is cyclized in the presence of base to afford a fused spirolactam of Formula IV. The fused spirolactam can be optionally oxidized to a sulfoxide or sulfone after Step 2 or Step 3. Oxidation prior to Step 2 results in undesired byproducts. In Step 3 the compound of Formula IV is optionally deprotected.

Scheme 1-7: Starting from compound of Formula IV a CDK4/6 inhibitor can be prepared. In Step 1 a heteroaryl amine is subjected to a base and a compound of Formula IV is added slowly under chilled conditions to afford a nucleophilic substitution reaction. The compound of Formula IV can previously be prepared as described in the schemes herein.

Example 2. Representative Routes of Synthesis

Scheme 2-1

quant, yield 2 steps

isolated

70% yield 2 steps 75% yield 95% yield

isolated isolated isolated

Scheme 2-1 : An ester route is one embodiment, of the present invention. Ideally, the best synthesis scheme would afford crystalline intermediates to provide material of consistent purity without column chromatography, and high yielding steps while using safe and cost effective reagents when possible.

The first step in the ester route is a SNAr nucleophilic substitution of CI group in commercially available ester 3 using spirolactam 4. Due to low reactivity of 4, a reaction temperature of 85-95 °C was required. Because of the temperature requirements, DIPEA and dimethylacetamide were selected as the base and solvent, respectively. The reaction follows second-order kinetics and usually stalls after -85% conversion. Therefore, the reaction was typically stopped after 60 hours by first cooling it to room temperature at which point solid formation was observed. The mixture was then partitioned between MTBE and water and product was filtered with excellent purity with -53% yield of the desired product 5. The obtained

compound 5 was protected with a Boc group using Boc anhydride and DMAP as the catalyst and dichloromethane as the solvent. The intermediate 6 was obtained in a quantitative yield. Due to the semi-solid nature of compound 6, the material was taken to the next step without further purification. The Dieckmann condensation was initially performed with strong bases such as LiHMDS and tBuOK. A similar result to the aldehyde route (Scheme 2-2) was obtained: a partial deprotection of Boc group was observed that required column chromatography. However, the best results were obtained when DBU was used as base and THF as solvent. The reaction outcome was complete, clean conversion of 6 to 7. Moreover, the product crystallized from the reaction mixture upon seeding, and a quantitative yield was obtained for the two steps.

The hydroxyl group of 7 was removed via a two-step procedure. First, compound 7 was converted completely into triflate 8 using triflic anhydride and triethylamine in dichloromethane. The reaction was found to proceed well at 0°C. Due to the potential instability of the triflate intermediate, it was not isolated. It was immediately taken to the next step and reduced with triethylsilane and palladium tetrakis to afford the product 9 after ethyl acetate crystallization in -70% yield. The Boc group of 9 was removed using trifluoroacetic acid in dichloromethane to afford 10. Intermediate 10 was converted into the final sulfone 11 using Oxone™ in acetonitrile/water solvent system.

The obtained sulfone 11 was use-tested in the coupling step and was found to perform well. In conclusion, the route to sulfone 11 was developed which eliminated the use of column chromatography with good to excellent yields on all steps.

Scheme 2-2


Molecular Weight: 421 

Scheme 2-2: The first step of Scheme 2-2 consistently afforded product 13 contaminated with one major impurity found in substantial amount. Thorough evaluation of the reaction impurity profile by LC-MS and 2D MR was performed, which showed the impurity was structurally the condensation of two aldehyde 12 molecules and one molecule of lactam 4. Therefore, column chromatography was required to purify compound 13, which consistently resulted in a modest 30% yield. A solvent screen revealed that sec-butanol, amyl alcohol, dioxane, and tert-butanol can all be used in the reaction but a similar conversion was observed in each case. However, tert-butanol provided the cleanest reaction profile, so it was selected as a solvent for the reaction. Assessing the impact of varying the stoichiometric ratio of 4 and 12 on the reaction outcome was also investigated. The reaction was performed with 4 equivalents of amine 4 in an attempt to disrupt the 2: 1 aldehyde/amine composition of the impurity. The result was only a marginal increase in product 13 formation. The temperature impact on the reaction outcome was evaluated next. The coupling of aldehyde 12 and 4 was investigated at two different temperatures: 50 °C and 40 °C with 1 : 1 ratio of aldehyde/amine. Reactions were checked at 2 and 4 hours and then every 12 hours. The reaction progress was slow at 50°C and was accompanied by growth of other impurities. The reaction at 40°C was much cleaner; however the conversion was lower in the same time period. The mode of addition of the reagents was investigated as well at 80°C with a slow addition (over 6 hours) of either aldehyde 12 or amine 4 to the reaction mixture. The product distribution did not change and an about 1 to 1 ratio was observed between product and impurity when amine 4 was added slowly to the reaction mixture containing aldehyde 12 and

DIPEA at reflux. The product distribution did change when aldehyde 12 was added slowly to the mixture of amine 4 and DIPEA. However, the major product of the reaction was the undesired impurity. Other organic bases were tried as well as different ratios of DIPEA. No product was observed when potassium carbonate was used as a base. The results of the experiments are presented in Table 1 below.

Table 1

Compound 13 was successfully formed in three cases: triethylamine, 2,6-lutidine and DIPEA, with the DIPEA result being the best. The use of Boc protected spirolactam 4 had no effect on the impurity formation as well. Its utilization was speculated to be beneficial in performing the coupling step together with the following step, preparation of compound 14.

The major impurity formed during Step 1 of Scheme 2-2 is:

Chemical Formula:€2)Η(¾ 62ί>2

Molecular Weight: 527.4903

The second step (Boc protection of the free lactam) proceeded well using DMAP as a catalyst in dichloromethane at room temperature. The product 14 is a thick oil, and, therefore, cannot be purified by crystallization. The Boc protected intermediate 14 was cyclized successfully into the desired pentacyclic structure 10 upon treatment with a strong base such as LiHMDS or tBuOK. Surprisingly, the Boc group was partially removed during the reaction. The level of deprotection was independent from the internal reaction temperature and was positively correlated with excess of base used. Therefore the mixture of the desired product 10 and 10-Boc compound was treated with acid to completely deprotect Boc group. The conversion of methyl sulfide into the final sulfone 11 was carried out with Oxone™. Initially a mixture of methanol and water was used for the reaction. As the result, a partial displacement of sulfone by methoxy group was detected. The methanol was replaced with acetonitrile and the sulfone displacement was eliminated.

In summary, the ester route (Scheme 2-1) is preferred because:

1. Formation of the impurity during the first step of Scheme 2-2 was unavoidable and resulted in yields of < 35%.

2. Column purification was required to isolate intermediate 14.

3. The aldehyde starting material was not commercially available and required two synthetic steps from the corresponding ester.

Scheme 2-3 : Starting with cyclohexanone, compounds of the present invention can be prepared. In Step 1 the methyl glycinate is subjected to cyclohexanone and TMSCN in the presence of tri ethyl amine in DCM to afford 15. In Step 2 15 hydrogenated with hydrogen gas in the presence of catalytic platinum oxide and subsequently undergoes an intramolecular cyclization to afford compound 16 which is used in the schemes above.

Scheme 2-4: Starting with compound 17, compounds of the present invention can be prepared. In Step 1 compound 17 is subjected to ethyl 2-oxoacetate in the presence platinum on carbon and hydrogen gas to afford compound 18. In Step 2 compound 18 is Boc-deprotected with hydrochloric acid. In Step 3 compound 18 is cyclized to afford compound 16 which is used in the schemes above.

Scheme 2-5

11 19

Scheme 2-5: Starting from compound 11 the CDK 4/6 inhibitor 19 can be prepared. In Step 1 5-(4-methylpiperazin-l-yl)pyridin-2-amine is subjected to LiHMDS and compound 11 is added slowly under chilled conditions to afford a nucleophilic substitution reaction and compound 19. Compound 11 can be prepared as described in the schemes herein.

Scheme 2-6: Starting from compound 11 the CDK 4/6 inhibitor 20 can be prepared. In Step 1 5-(4-isopropylpiperazin-l-yl)pyridin-2-amine is subjected to LiHMDS and compound 11 is added slowly under chilled conditions to afford a nucleophilic substitution reaction and compound 20. Compound 11 can be prepared as described in the schemes herein.

Preparation of Compound 5:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet, and reflux condenser was charged with ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate 3 (49.2 g, 0.21 mol, 1.00 equiv.), spirolactam 4 (39.2 g, 0.23 mol, 1.10 equiv.), DIPEA (54.7 g, 0.42 mol, 2.00 equiv.), and DMAc (147.6 mL, 3 vol). The batch was heated to 90-95 °C, and after 60 h, IPC confirmed -14% (AUC) of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate remained. The batch was cooled to RT, and precipitate formation was observed. The suspension was diluted with MTBE (100 mL, 2 vol) and water (442 mL, 9 vol) and stirred for 2 h at RT. The product was isolated by vacuum filtration and washed with MTBE (49 mL, 1 vol). The solid cake was conditioned for 1 h and dried under vacuum at 40 °C for 16 h to afford compound 5 [41.0 g, 53% yield] as an off-white solid with a purity of >99% AUC. ¾ MR (CDCh): δ 8.76 (d, J = 2.0 Hz, 1H), 6.51-6.29 (br, 1H), 4.33 (q, J = 7.0 Hz, 2H), 3.78 (s, 2H), 3.58 (s, 2H), 2.92 (s, 2H), 2.53 (s, 3H), 1.63-1.37 (m, 12H). LCMS (ESI, m/z = 365.3 [M+H]).

Preparation of Compound 6:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with 5 [41.0 g, 0.11 mol, 1.00 equiv.], Boc-anhydride (36.8 g, 0.17 mol, 1.50 equiv.), DMAP (1.37 g, 0.01 mol, 0.10 equiv.), and dichloromethane (287 mL, 7 vol). The batch was stirred for 3 h at RT. IPC confirmed no starting material remained (AUC). The batch was concentrated into a residue under reduced pressure and taken to the next step (a quantitative yield is assumed for this step). An aliquot (200 mg) was purified by column chromatography (heptanes/ethyl acetate 0 to 100%) to afford compound 6. 1H MR (CDCh): δ 8.64 (s, 1H), 4.31 (q, J = 7.0 Hz, 2H), 4.07 (s, 2H), 3.83 (S, 2H), 3.15 (m, 2H), 2.56 (s, 3H), 172 (m, 3H), 1.59 (m, 15H), 1.42 (t, J= 7.0 Hz, 3H). LCMS (ESI, m/z = 465.2 [M+H]).

Preparation of Compound 7:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with compound 6 [residue from a previous step, quantitative yield assumed, 52.2 g, 0.11 mol, 1.00 equiv.], and THF (261 mL, 5 vol). The batch was cooled to 0°C and 1,8-diazabicyclo[5.4.0]un-dec-7-ene (17.1 g, 0.11 mmol, 1.00 equiv.) was added keeping the internal temperature in 0-10°C range. After the addition was complete, the cooling bath was removed and the reaction mixture was allowed to warm up to RT and after 2 h, IPC confirmed no starting material remained. The batch was seeded with the product (1.0 g) and was cooled to 0°C. The slurry was stirred at 0°C for 2 h. The product was isolated by vacuum filtration and washed with cold (0°C) THF (50 mL, 1 vol). The solid cake was conditioned for 1 h and dried under vacuum at 40°C for 16 h to afford 7 [47 g, quantitative yield] as a light orange solid with a purity of >99% AUC. The color of the product changed into yellow once the batch was exposed to air for an extended period of time (~ 1 day). Material was isolated with substantial amount DBU, according to proton NMR. However, it did not interfere with the next step. 1H MR (CDCh): δ 8.71 (s, 1H), 4.03 (s, 2H), 2.57 (s, 3H), 1.85 (m, 10H), 1.51 (s, 9H). LCMS (ESI, m/z = 419.2 [M+H]).

Preparation of Compound 8:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with 7 [40.8 g, 0.10 mol, 1.00 equiv.], triethylamine (31.5 g, 0.31 mol, 3.20 equiv.), and dichloromethane (408 mL, 10 vol). The batch was purged with N2 for 15 min and was cooled to 0°C. Triflic anhydride (44.0 g, 0.16 mol, 1.60 equiv.) was added keeping the

internal temperature in 0-10°C range. The batch was stirred at 0°C and after 3 h, IPC confirmed -7.0% (AUC) of 7 remained. [It was speculated that the product was hydrolyzing back into starting material during the analysis.] Once the reaction was deemed complete, the batch was transferred to a 1 L, separatory funnel and was washed with 50% saturated sodium bicarbonate (200 mL, 5 vol). [It was prepared by mixing saturated sodium bicarbonate (100 mL) with water (100 mL)).] The aqueous layer was separated and was extracted with DCM (2×40 mL, 1 vol). The organic layers were combined and concentrated into a residue under reduced pressure and taken to the next step. LCMS (ESI, m/z = 551.6 [M+H]).

Preparation of Compound 9:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with compound 8 [residue from a previous step, quantitative yield assumed, 53.7 g, 0.10 mol, 1.00 equiv.], and THF (110 mL, 2 vol). The solvent was removed under vacuum distillation and the procedure was repeated two times. The flask was charged with triethylsilane (22.7 g, 0.20 mol, 2.00 equiv.), and DMF (268 mL, 5 vol). The batch was degassed by five cycles of evacuation, followed by backfilling with nitrogen. The flask was charged with tetrakis(triphenylphosphine)palladium(0) (11.3 g, 0.01 mol, 0.1 equiv.). The batch was heated to 45-50°C, and after 14 h, IPC confirmed no starting material remained. The batch was transferred to a 500 mL, separatory funnel while still warm. The reaction was partitioned between water (5 vol) and ethyl acetate (5 vol). The aqueous layer was extracted with ethyl acetate (3 x3 vol). The organic layers were combined and concentrated down to 2 volumes. The precipitate was filtered and washed with ethyl acetate (2x 1 vol). The solid cake was conditioned for 1 h and dried under vacuum at 40°C for 16 h to afford 9 [27.5 g, 70% yield] as a yellow solid with a purity of -98% AUC. Proton NMR showed some triphenylphosphine oxide present. ¾ NMR (DMSO-i¾):5 9.01 (s, 1H), 7.40 (s, 1H), 4.30 (s, 2H), 2.58 (m, 2H), 2.58 (s, 3H), 1.81 (m, 5H), 1.51 (s, 9H). LCMS (ESI, m/z = 403.4 [M+H]).

Preparation of Compound 10 from the Scheme 2-1 route:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged 9 (12.8 g, 31.8 mmol, 1.00 equiv.) and dichloromethane (64 mL, 5 vol). Trifluoroacetic acid (18.2 g, 159 mmol, 5.00 equiv.) was added over 20 min and the solution was stirred for 2 h at RT. IPC confirmed reaction was complete. The batch was transferred to a 500 mL, separatory funnel and washed with saturated sodium bicarbonate (200 mL). The aqueous layer was extracted with dichlorom ethane (3 x3 vol). The organic layers were combined and concentrated down to 1 volume. The precipitate was filtered and conditioned for 1 h and dried under vacuum at 40 °C for 16 h to afford 9 [6.72 g, 70% yield] as an off-white solid with a purity of 99.1% AUC. ¾ NMR (DMSO-dis): δ 8.95 (s, 1H), 8.32 (s, 1H), 7.15 (s, 1H), 3.68 (d, J = 1.0 Hz, 2H), 2.86 (m, 2H), 2.57 (s, 3H), 1.92 (m, 2H), 1.73 (m, 3H), 1.39 (m, 3H). LCMS, ESI, m/z = 303.2 [M+H]).

Preparation of Compound 10 from Scheme 2-2 route:

A 50 mL, three-neck flask equipped with a magnetic stirring bar, thermocouple, N2 inlet was charged 14 (680 mg, 1.62 mmol, 1.00 equiv.) and THF (6.8 mL, 10 vol). A I M solution of potassium tert-butoxide (3.2 mL, 3.24 mmol, 2.00 equiv.) in THF was added over 10 min and the solution was stirred for 2 h at RT. IPC confirmed reaction was complete. The batch was acidified with 4 N hydrogen chloride solution in dioxane (2.4 mL, 9.72 mmol, 6.00 equiv.) and stirred for additional 1 h. The batch was transferred to a 500 mL, separatory funnel and washed with saturated sodium bicarbonate (100 mL). The aqueous layer was extracted with ethyl acetate (3 x20 vol). The organic layers were combined and concentrated down to 3volumes and product precipitated. The precipitate was filtered and conditioned for 1 h and dried under vacuum at 40 °C for 16 h to afford 9 [489 mg, quantitative yield] as an off-white solid.

Preparation of Compound 11 :

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with 10 (9.00 g, 29.8 mmol, 1.00 equiv.), and acetonitrile (180 mL, 20 vol). A solution of Oxone™ (45.9 g, 0.15 mol, 5.00 equiv.) in water (180 mL, 20 vol) was added to the batch over 20 min. The batch was stirred for 2 h and IPC confirmed the reaction was complete. The batch was concentrated down to ½ of the original volume and was extracted with dichloromethane DCM (4x 10 vol). The organic layers were combined; polish filtered and concentrated down to -10 vol of DCM. The product was slowly crystallized out by addition of heptanes (-30 vol). The mixture was cooled to 0°C and the product was filtered and dried under vacuum at 40 °C for 16 h to afford 11 [9.45 g, 95% yield] as an off-white solid with a purity of >99% AUC. ¾ NMR (CDCb): 5 9.24 (s, 1H), 7.78 (s, 1H), 7.46 (s, 1H), 3.89 (d, J= 2.0 Hz, 2H), 3.43 (s, 3H), 2.98 (m, 2H), 2.10 (m, 2H), 1.86 (m, 3H), 1.50 (m, 3H). LCMS (ESI, m/z = 335.2 [M+H]).

Preparation of Compound 13:

A 250 mL, single-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet, and reflux condenser was charged with 4-chloro-2-(methylthio)pyrimidine-5-carbaldehyde (2.00 g, 10.6 mmol, 1.00 equiv.), spirolactam 4 (1.96 g, 11.7 mmol, 1.10 equiv.), DIPEA (2.74 g, 21.2 mmol, 2.00 equiv.), and fert-butanol (20 mL, 10 vol). The batch was heated to 80-85 °C, and after 24 h, IPC confirmed no aldehyde 12 remained. The batch was cool to RT and concentrated into a residue, which was loaded on silica gel column. The product was eluted with ethyl acetate/heptanes (0% to 100%). The product containing fractions were pulled out and concentrated to afford 13 [0.98 g, 29% yield] as an off-white solid.

Preparation of Compound 14:

A 500 mL, three-neck flask equipped with a mechanical overhead stirrer, thermocouple, N2 inlet was charged with 13 [0.98 g, 3.00 mmol, 1.00 equiv.], Boc-anhydride (4.90 g, 21.5 mmol, 7.00 equiv.), DMAP (36 mg, 0.30 mmol, 0.10 equiv.), and dichloromethane (7 mL, 7 vol). The batch was stirred for 3 h at RT. IPC confirmed no starting material remained. The batch was cool to RT and concentrated into a residue, which was loaded on silica gel column. The product was eluted with ethyl acetate/heptanes (0% to 100%). The product containing fractions were pulled out and concentrated to afford 14 [0.98 g, 29% yield] as an off-white solid.

Preparation of Compound 15:

To a suspension of methyl glycinate (500 g, 3.98 mol, 1 eq) in DCM (10 L) was added

TEA dropwise at rt under nitrogen atmosphere, followed by the addition of cyclohexanone (781 g, 7.96 mol, 2 eq) dropwise over 15 min. To the resulting mixture was added TMSCN (591 g, 5.97 mol, 1.5 eq) dropwise over 1 hour while maintaining the internal reaction temperature below 35

°C. After stirred at rt for 2 hrs, the suspension became a clear solution. The progress of the reaction was monitored by H- MR.

When the methyl glycinate was consumed completely as indicated by H-NMR analysis, the reaction was quenched by water (5 L). The layers were separated. The aqueous layer was extracted with DCM (1 L). The combined organic phase was washed with water (5 L X 2) and

dried over Na2S04 (1.5 Kg). After filtration and concentration, 1.24 Kg of crude 15 was obtained as oil.

The crude 15 was dissolved in IPA (4 L). The solution was treated with HC1/IPA solution (4.4 mol/L, 1.1L) at RT. A large amount of solid was precipitated during the addition. The resulting suspension was stirred for 2 hrs. The solid product was collected by vacuum filtration and rinsed with MTBE (800 mL). 819 g of pure 15 was obtained as a white solid. The yield was 88.4%. ¾- MR (300 MHz, CD3OD) 4.20 (s, 2H), 3.88 (s, 3H), 2.30-2.40 (d, J = 12 Hz, 2H), 1.95-2.02 (d, J = 12 Hz, 2H), 1.55-1.85 (m, 5H), 1.20-1.40 (m, 1H).

Preparation of Compound 16:

To a solution of 15 (10 g, 43 mmol) in MeOH (100 mL) was added methanolic hydrochloride solution (2 .44 mol/L, 35.3 mL, 2 eq) and Pt02 (0.5 g, 5 wt %). The reaction suspension was stirred with hydrogen bubble at 40 °C for 6 hours. H- MR analysis showed consumption of 15. To the reaction mixture was added K2CO3 (15 g, 108 mmol, 2.5 eq) and the mixture was stirred for 3 hrs. The suspension was filtered and the filtrate was concentrated to dryness. The residual oil was diluted with DCM (100 mL) and resulting suspension was stirred for 3 hrs. After filtration, the filtrate was concentrated to provide crude 16 (6.6 g) as an oil. The crude 16 was diluted with EtOAc/hexane (1 : 1, 18 mL) at rt for 2 hrs. After filtration, 16 (4 g) was isolated. The obtained 16 was dissolved in DCM (16.7 mL) and hexane (100 mL) was added dropwise to precipitate the product. After further stirred for 1 h, 2.8 g of the pure 16 was isolated as a white solid. The yield was 39%. HPLC purity was 98.3%; MS (ESI): 169.2 (MH+); 1 H-NMR (300 MHz, D2O) 3.23 (s, 3H), 3.07 (s, 3H), 1.37-1.49 (m, 10H).

Preparation of compound 19:

5-(4-methylpiperazin-l-yl)pyridin-2-amine (803.1 g; 3.0 equivalents based on sulfone 11) was charged to a 22 L flask. The flask was blanketed with N2 and anhydrous THF added (12.4 kg). The resulting black-purple solution was cooled in an ice bath to < 5°C. 1M LiHMDS (4.7 L; 1.2 equivalents based on sulfone 11) was added via an addition funnel in three equal additions to keep the temperature below 10°C. Upon the completion of the addition, the reaction mixture was warmed to 16°C. The sulfone 11 (455.1 g; 1.00 equivalents) was added in five additions. Reaction proceeded until HPLC analysis of an IPC sample indicated less than 3% of sulfone 11 remained.

To quench the reaction, the contents of the 22L flask were transferred to a 100 L flask containing water. After stirring for 30 minutes at <30°C, the crude product was collected by filtration, washed with water and dried to afford 19 (387 g, 99.1% purity, 63.7% yield).

Preparation of compound 20:

5-(4-isopropylpiperazin-l-yl)pyridin-2-amine (1976.2 g; 3.0 equivalents based on sulfone 11) was charged to a 50 L flask. The flask was blanketed with N2 and anhydrous THF added (10.7 kg). The resulting black-purple solution was cooled in an ice bath to < 5°C. 1M LiHMDS (9.6 kg; 3.6 equivalents based on sulfone) was added via an addition funnel at a rate to keep the temperature below 10°C. Upon the completion of the addition, the reaction mixture was warmed to 16°C over 120 minutes by removing the ice bath. The sulfone (1000 g; 1.00 mol) was added in five additions. The reaction proceeded until HPLC analysis of an IPC sample indicated less than 1% of sulfone 11 remained. After completion of the reaction, ammonium chloride was added to the reaction mixture. The mixture stirred at < 32°C for at least 30 minutes and the solids collected by filtration to afford 20 (900 g, 99.1% purity, 64.2% yield).

Alternate Route to Spirolactam via cyclohexanone:

Scheme 2-7

26

In one embodiment the spirolactam is made via the synthetic scheme above. By reducing the nitrile group before addition of the glycinate group the reaction sequence proceeds in higher yield. The chemistry used in Step 1 is described in the literature (J. Org. Chem. 2005, 70,8027-8034), and was performed on a kilogram scale. The chemistry to convert Compound 24 into the

spirolactam was also demonstrated on kilogram scale. The Boc protection of Compound 23, is carried out at -70°C in order to limit formation of the di-Boc protected product. Experimental details of a 200 g pilot run are described below.

Step 1

200 g of cyclohexanone 21 was converted to 22 using Ti(Oi-Pr)4 /TMSCN/NH3. After work-up, 213 g of 22 was obtained. The H- MR was clean. The yield was 84%. The titanium salts were removed by vacuum filtration. In one embodiment, the titanium salts are removed by centrifugation or Celite filtration.

Step 2

190 g of 22 was mixed with LAH (2 eq) in MTBE for 30 minutes at 45°C. After work-up, 148 g of crude 23 was obtained.

Step 3

136 g of the crude 23 from step 2 was converted to 24 with 0.9 eq of B0C2O at -70°C. The reaction was completed and worked up. After concentration, 188 g of 24 was obtained. The yield was 86%. The H-NMR and C-NMR spectra confirmed that the compound was pure.

Step 4

188 g of 24 was subjected to methyl 2-bromoacetate and K2CO3 in acetonitrile to afford 25. 247 g of crude 25 was obtained.

Step 5

247 g of 25 was subjected to TFA in DCE heated to reflux to afford 26. After work-up, 112 g of 6 as TFA salt was obtained. H- MR was clean.

Step 6

26 27

Compound 26 was stirred in EtOH in the presence at room temperature overnight to afford 27. In one embodiment DCM is used as the solvent instead of EtOH.

Example 3. Purge of residual palladium from Step 5 Scheme 2-1:

Since palladium was used in Step 5 of Scheme 2-1, the levels of residual Pd present in the subsequent synthetic steps was determined. Table 2 below and Figure 3 show the palladium levels in the isolated solids.

Table 2

The material after Step 5 was isolated containing 1.47% (14700 ppm) of residual palladium. This data represents the highest level of palladium in the worst case scenario. The workup conditions of the latter steps purged nearly all of the palladium and the final product, 19 bis HC1 salt, contained 14 ppm of Pd, which is below the standard 20 ppm guidline. The Pd levels will likely be even lower once the catal st loading is optimized in Step 5.

19

The process developed in this route was a significant improvement over the one used for the first generation synthesis. Overall, the scheme consists of seven steps with five isolations, all by crystallization. No silica column chromatography is employed in the synthesis, which makes the process highly scalable. The process workup conditions can successfully purge the 1.47% of residual palladium after step 5 of Scheme 2-1.

Patent ID

Patent Title

Submitted Date

Granted Date

US8829012 CDK inhibitors
2014-01-23
2014-09-09
US8598197 CDK inhibitors
2013-04-24
2013-12-03
US8598186 CDK inhibitors
2013-04-24
2013-12-03
US8691830 CDK inhibitors
2013-04-24
2014-04-08
US2014274896 Transient Protection of Hematopoietic Stem and Progenitor Cells Against Ionizing Radiation
2014-03-14
2014-09-18
Patent ID

Patent Title

Submitted Date

Granted Date

US2015297607 Tricyclic Lactams for Use in the Protection of Normal Cells During Chemotherapy
2015-04-17
2015-10-22
US2015297608 Tricyclic Lactams for Use as Anti-Neoplastic and Anti-Proliferative Agents
2015-04-17
2015-10-22
US9487530 Transient Protection of Normal Cells During Chemotherapy
2014-03-14
2014-09-18
US2017057971 CDK Inhibitors
2016-11-10
US2017037051 TRANSIENT PROTECTION OF NORMAL CELLS DURING CHEMOTHERAPY
2016-10-07
Patent ID

Patent Title

Submitted Date

Granted Date

US2017100405 HSPC-Sparing Treatments for RB-Positive Abnormal Cellular Proliferation
2016-12-21
US2017065597 Transient Protection of Normal Cells During Chemotherapy
2016-11-03
US2016310499 Highly Active Anti-Neoplastic and Anti-Proliferative Agents
2016-07-01
US2016220569 CDK4/6 Inhibitor Dosage Formulations For The Protection Of Hematopoietic Stem And Progenitor Cells During Chemotherapy
2016-02-03
2016-08-04
US2015297606 Tricyclic Lactams for Use in the Protection of Hematopoietic Stem and Progenitor Cells Against Ionizing Radiation
2015-04-17
2015-10-22
Patent ID

Patent Title

Submitted Date

Granted Date

US9717735 Tricyclic Lactams for Use in HSPC-Sparing Treatments for RB-Positive Abnormal Cellular Proliferation
2015-04-17
2015-10-22
US9527857 HSPC-Sparing Treatments for RB-Positive Abnormal Cellular Proliferation
2014-03-14
2014-09-18
US2014271460 Highly Active Anti-Neoplastic and Anti-Proliferative Agents
2014-03-14
2014-09-18
US2017182043 Anti-Neoplastic Combinations and Dosing Regimens using CDK4/6 Inhibitor Compounds to Treat RB-Positive Tumors
2017-03-13
US2017246171 Treatment Of RB-Negative Tumors Using Topoisomerase Inhibitors In Combination With Cyclin Dependent Kinase 4/6 Inhibitors
2017-03-13

///////////////TRILACICLIB, G1T28, G1T 28, SHR 6390, PHASE 2, G1 Therapeutics, Inc.

CN1CCN(CC1)C2=CN=C(C=C2)NC3=NC=C4C=C5C(=O)NCC6(N5C4=N3)CCCCC6

Maralixibat Chloride, ماراليكسيبات كلوريد , 氯马昔巴特 , Мараликсибата хлорид


STR1

2D chemical structure of 228113-66-4

Maralixibat chloride

Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

SHP625, Maralixibat chloride, Molecular Formula C40-H56-N3-O4-S.Cl, Molecular Weight, 710.4184

4-Aza-1-azoniabicyclo(2.2.2)octane, 1-((4-((4-((4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl)-, chloride (1:1)

1-[4-({4-[(4R,5R)-3,3-Dibutyl-7-(dimethylamino)-4-hydroxy-1,1-dioxido-2,3,4,5-tetrahydro-1-benzothiepin-5-yl]phenoxy}methyl)benzyl]-4-aza-1-azoniabicyclo[2.2.2]octane chloride

4-Aza-1-azoniabicyclo[2.2.2]octane, 1-[[4-[[4-[(4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-, chloride

(4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt

(4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride

Cas: 228113-66-4, Free form 716313-53-0
UNII: V78M04F0XC, LUM 001, Lopixibat chloride, Treatment of Cholestatic Liver Diseases

update, FDA APPROVED 2021, 2021/9/29, Livmaril AS CHLORIDE CAS 228113-66-4

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

  •  

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter 

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter, Less «
Applicant G.D. Searle & Co.

 

Several drawings of Maralixibat chloride

STR1

ChemSpider 2D Image | maralixibat chloride | C40H56ClN3O4S

STR1Figure imgf000053_0001

It is well established that agents which inhibit the 20 transport of bile acids across the ileum can also cause a decrease in the level of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255- 25287, discusses biochemistry, physiology, and known active agents affecting bile acids and cholesterol.

A class of ileal bile acid transport-inhibiting compounds which was recently discovered to be useful for influencing the level of blood serum cholesterol is 30 tetrahydrobenzothiepine-l,l-dioxides (THBDO compounds). (U.S. Patent Application No. 08/816,065)

Some classes of compounds show enhanced potency as pharmaceutical therapeutics after they have been enantiomerically-enriched (see, for example, Richard B. Silverman, The Organic Chemistry of Drug Design and Drug Action, Academic Press, 1992, pp. 76-82) . Therefore, THBDO compounds that have been enantiomerically-enriched are of particular interest.

A class of chemistry useful as intermediates in the preparation of racemic THBDO compounds is tetrahydrobenzothiepine-1-oxides (THBO compounds) . THBDO compounds and THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring. A method of preparing enantiomerically-enriched samples of another phenyl/seven-member fused ring system, the benzothiazepines, is described by Higashikawa (JP 59144777) , where racemic benzothiazepine derivatives are optically resolved on a chromatographic column containing chiral crown ethers as a stationary phase. Although optical resolution is achieved, the Higashikawa method is limited to producing only small quantities of the enantiomerically-enriched benzothiazepine derivatives. Giordano (CA 2068231) reports the cyclization of (2S, 3S) -aminophenylthiopropionates in the presence of a phosphonic acid to produce (2S, 3S) -benzothiazepin-4-ones . However, that preparation is constrained by the need to use enantiomerically-enriched starting materials rather than racemic starting materials. In addition, the Giordano method controls the stereochemistry of the seven-member ring of the benzothiazepin-4-one only at the 2- and 3 -positions. The 4- and 5-positions of the seven-member ring of the benzothiazepin-4-one are not asymmetric centers, and the stereochemistry at these sites therefore cannot be controlled by the Giordano method. A method by which enantiomerically-enriched 1,5- benzothiazepin-3-hydroxy-4 (5H) -one compounds have been produced is through the asymmetric reduction of 1,5- benzothiazepin-3,4 (2H, 5H) -dione compounds, reported by Yamada, et al . (J. Org. Chem. 1996, 61 (24), 8586-8590). The product is obtained by treating the racemic 1,5- benzothiazepin-3,4 (2H, 5H) -dione with the reaction product of an optically active alpha-amino acid and a reducing agent, for example sodium borohydride. Although a product with high optical purity was achieved, the method is limited by the use of a relatively expensive chemical reduction step.

The microbial reduction of racemic 1, 5-benzothiazepin- 3 , 4 (2H, 5H) -dione compounds to produce enantiomerically- enriched 1, 5-benzothiazepin-3-hydroxy-4 (5H) -one compounds is reported by Patel et al . , U.S. Patent 5,559,017. This method is limited by the inherent problems of maintaining a viable and pure bacterial culture of the appropriate species and variety. In addition, that method is limited in scale, producing only microgram quantities of the desired product. Until now, there have been no reported processes for preparing enantiomerically-enriched THBDO compounds or enantiomerically-enriched THBO compounds. Furthermore, there have been no reported processes for controlling the stereochemistry at the 4- and 5-positions of the seven- member rings of THBDO compounds or THBO compounds

FDA Grants Breakthrough Designation to Shire’s Rare GI Therapies

Tue, 06/14/2016

Shire announced that the U.S. Food and Drug Administration (FDA) has granted Breakthrough Therapy Designation for two investigational products for rare diseases: SHP621 (budesonide oral suspension, or BOS) for eosinophilic esophagitis (EoE), and SHP625 (maralixibat) for progressive familial intrahepatic cholestasis type 2 (PFIC2).

“Receiving Breakthrough Therapy Designation on two pipeline products this past week reflects the potential of our strong and innovative pipeline of more than 60 programs,” said Flemming Ornskov, M.D., MPH, and CEO, Shire. “Shire is committed to bringing innovation to the rare and specialty areas we focus on. We persevere to see compounds through the many stages of development through their challenges and successes, and always keep patients with unmet needs top of mind.”

EoE is a serious, chronic and rare disease that stems from an elevated number of eosinophils, a type of white blood cell, that infiltrate the walls of the esophagus. EoE is characterized by an inflammation of the esophagus that may lead to difficulty swallowing (dysphagia). The diagnosed prevalence of EoE ranges from approximately 15-55 cases per 100,000 persons, with high-end estimates reported by studies in Western regions.

PFIC refers to a group of autosomal-recessive liver disorders of childhood that disrupt bile formation and present with cholestasis. The symptoms of PFIC include severe itching of the skin (pruritus), and jaundice. PFIC is estimated to affect 1 in 50,000 to 1 in 100,000 births. PFIC2 is the most common type of PFIC, accounting for around half of cases.

According to the FDA, Breakthrough Therapy Designation is granted to a therapy that is intended to treat a serious or life-threatening disease or condition and preliminary clinical evidence indicates that the drug may demonstrate substantial improvement on one or more clinically significant endpoints over current standard of care. Under the designation, the FDA provides intensive guidance, organizational commitment involving senior managers, and eligibility for rolling and priority review of the application; this process helps ensure patients have access to therapies as soon as possible, pending approval. Breakthrough Therapy Designation does not guarantee that FDA will ultimately approve BOS for EoE or maralixibat for PFIC2, and the timing of any such approval is uncertain.

“On behalf of patients in the United States with EoE and PFIC2, we are so pleased that the FDA has granted Breakthrough Therapy Designation to BOS and maralixibat,” said Philip J. Vickers, Ph.D., Head of R&D, Shire. “We look forward to working with the agency to continue their development and, pending FDA approval, deliver these therapeutic options to the patients who need them most.”

Source: Shire

Patent

WO 2003022804

It is well established that agents which inhibit the transport of bile acids across the tissue of the ileum can also cause a decrease in the levels of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255-287 discusses biochemistry, physiology, and known active agents surrounding bile acids and cholesterol. Bile acids are actively transported across the tissue of the ileum by an apical sodium co-dependent bile acid transporter (ASBT), alternatively known as an ileal bile acid transporter (IBAT).
A class of ASBT-inhibiting compounds that was recently discovered to be useful for influencing the level of blood serum cholesterol comprises tetrahydrobenzothiepine oxides (THBO compounds, PCT Patent Application No. WO 96/08484). Further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 97/33882.
Additional THBO compounds useful as ASBT inhibitors are described in U.S. Patent No. 5,994,391. Still further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 99/64409. Included in the THBO class are tetrahydrobenzo-thiepine-l -oxides and tetrahydrobenzothiepine- 1,1 -dioxides. THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring.

Published methods for the preparation of THBO compounds include the synthesis through an aromatic sulfone aldehyde intermediate. For example l-(2,2-dibutyl-3-oxopropylsulfonyl)-2-((4-methoxyphenyl)methyl)benzene (29) was cyclized with potassium t-butoxide to form tetrahydrobenzothiepine- 1,1 -dioxide (svn-24) as shown in Eq. 1.

Compound 29 was prepared by reacting 2-chloro-5-nitrobenzoic acid chloride with anisole in the presence of aluminum trichloride to produce a chlorobenzophenone compound; the chlorobenzophenone compound was reduced in the presence of trifluoromethanesulfonic acid and triethylsilane to produce a chlorodiphenylmethane compound; the
chlorodiphenylmethane compound was treated with lithium sulfide and 2,2-dibutyl-3-(methanesulfonato)propanal to produce l-(2,2-dibutyl-3-oxopropylthio)-2-((4-methoxyphenyl)methyl)-4-dimethylaminobenzene (40); and 40 was oxidized with m-chloroperbenzoic acid to produce 29. The first step of that method of preparing compound 29 requires the use of a corrosive and reactive carboxylic acid chloride that was prepared by the reaction of the corresponding carboxylic acid with phosphorus pentachloride.
Phosphorus pentachloride readily hydrolyzes to produce volatile and hazardous hydrogen chloride. The reaction of 2,2-dibutyl-3-(methanesulfonato)propanal with the lithium sulfide and the chlorodiphenylmethane compound required the intermediacy of a cyclic tin compound to make the of 2,2-dibutyl-3-(methanesulfonato)propanal. The tin compound is expensive and creates a toxic waste stream.
In WO 97/33882 compound syn-24 was dealkylated using boron tribromide to produce the phenol compound 28. Boron tribromide is a corrosive and hazardous material that generates hydrogen bromide gas and requires special handling. Upon hydrolysis, boron tribromide also produces borate salts that are costly and time-consuming to separate and dispose of.

An alternative method of preparing THBO compounds was described in WO
97/33882, wherein a 1,3-propanediol was reacted with thionyl chloride to form a cyclic sulfite compound. The cyclic sulfite compound was oxidized to produce a cyclic sulfate compound. The cyclic sulfate was condensed with a 2-methylthiophenol that had been deprotonated with sodium hydride. The product of the condensation was a (2-methylphenyl) (3′-hydroxypropyl)thioether compound. The thioether compound was oxidized to form an thioether aldehyde compound. The thioether aldehyde compound was further oxidized to form an aldehyde sulfone compound which in turn was cyclized in the presence of potassium t-butoxide to form a 4-hydroxytetrahydrobenzothiepine 1,1 -dioxide compound. This cyclic sulfate route to THBO compounds requires an expensive catalyst. Additionally it requires the use of SOCI2, which in turn requires special equipment to handle.
PCT Patent Application No. WO 97/33882 describes a method by which the phenol compound 28 was reacted at its phenol hydroxyl group to attach a variety of functional groups to the molecule, such as a quaternary ammonium group. For example, (4R,5R)-28 was reacted with l,4-bis(chloromethyl)benzene (?,??’-dichloro-p-xylene) to produce the chloromethyl benzyl- ether (4R,5R)-27. Compound (4R,5R)-27 was treated with diazabicyclo[2.2.2]octane (DABCO) to produce (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4-hydroxy-l , 1 -dioxido-1 -benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza-l-azomabicyclo[2.2.2]octane chloride (41). This method suffers from low yields because of a propensity for two molecules of compound (4R,5R)-28 to react with one molecule of l,4-bis(chloromethyl)benzene to form a bis(benzothiepine) adduct. Once the bis-adduct forms, the reactive chloromethyl group of compound (4R,5R)-27 is not available to react with an amine to form the quaternary ammonium product.

A method of preparing enantiomerically enriched tetrahydrobenzothiepine oxides is described in PCT Patent Application No. WO 99/32478. In that method, an aryl-3- hydroxypropylsulfide compound was oxidized with an asymmetric oxidizing agent, for example (lR (->(8,9-dichloro-10-camphorsulfonyl)oxaziridine, to yield a chiral aryl-3-hydroxypropylsulfoxide. Reaction of the aryl-3-hydroxypropylsulfoxide with an oxidizing agent such as sulfur trioxide pyridine complex yielded an aryl-3-propanalsulfoxide. The aryl- 3-propanalsulfoxide was cyclized with a base such as potassium t-butoxide to
enantioselectively produce a tetrahydrobenzothiepine- 1 -oxide. The tetrahydrobenzothiepine- 1 -oxide was further oxidized to produce a tetrahydrobenzothiepine- 1 , 1 -dioxide. Although this method could produce tetrahydrobenzothiepine- 1,1 -dioxide compounds of high enantiomeric purity, it requires the use of an expensive asymmetric oxidizing agent.
Some 5-amidobenzothiepine compounds and methods to make them are described in

PCT Patent Application Number WO 92/18462.
In Svnlett. 9, 943-944(1995) 2-bromophenyl 3-benzoyloxy-l-buten-4-yl sulfone was treated with tributyl tin hydride and AIBN to produce 3-benzoyloxytetrahydrobenzothiepine-1,1 -dioxide.
In addition to forming the desired ASBT inhibitors, it is also desirable to form such

ASBT inhibitors of higher purity and having lower levels of residual solvent impurities. This is especially so with respect to ASBT inhibitors having a positively charged substituent, for example, the compounds designated as 41 (supra) and 60 (infra).
It is further desirable to provide methods for making such high purity ASBT inhibitors.

Example 11.

Preparation of (4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride,
41


41

Ste l. Preparation of (4R.5R1-26.


( 4R, 5R) -26
A 1000 mL 4 neck jacketed Ace reactor flask was fitted with a mechanical stirrer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask was purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). To this was added 4.2 grams of 50% sodium hydroxide. The mixture was heated to 50°C and stiπed for 15 minutes. To the flask was added 8.3 grams of 55 dissolved in 10 mL of DMAC, all at once. The temperature was held at 50°C for 24 hrs. To the flask was added 250 mL of toluene followed by 125 mL of dilution water. The mixture was stiπed for 15 minutes and the layers were then allowed to separate at 50°C. The flask was then charged with 125 mL of saturated sodium chloride solution and stiπed 15 minutes. Layers separated cleanly in 30 seconds at 50°C. Approximately half of the solvent was distilled off under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.


( 4R, 5R) -27
Toluene was charged back to the reaction mixture of Step 1 and the mixture was cooled to 35°C. To the mixture was then added 7.0 grams of thionyl chloride over 5 minutes. The reaction was exothermic and reached 39°C. The reaction turned cloudy on first addition of thionyl chloride, partially cleared then finally remained cloudy. The mixture was stirred for 0.5 hr and was then washed with 0.25N NaOH. The mixture appeared to form a small amount of solids that diminished on stirring, and the layers cleanly separated. The solvent was distilled to a minimum stir volume under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 was charged with 350 mL of methyl ethyl ketone (MEK) followed by 10.5 mL water and 6.4 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 10 mL of MEK. The mixture was heated to reflux, and HPLC showed <0.5% of (4R,5R)-27. The reaction remained homogenous initially then crystallized at the completion of the reaction. An additional 5.3 mL of water was charged to the flask to redissolve product. Approximately 160 mL of solvent was then distilled off at atmospheric pressure. The mixture started to form crystals after 70 mL of solvent was distilled. Water separated out of distillate indicating a ternary azeotrope between toluene, water and methyl ethyl ketone (MEK). The mixture was then cooled to 25°C. The solids were filtered and washed with 150 mL MEK, and let dry under vacuum at 60°C. Isolated 29.8.0 g of off-white crystalline 4 Example 11a.
Alternate Preparation of (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 100 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 4.02 grams of 50% sodium hydroxide. The mixture is stiπed for 30 minutes. To the flask is added 8.7 grams of 55 dissolved in 12.5 mL of DMAC, all at once. The charge vessel is washed with 12.5 mL DMAC and the wash is added to the reactor. The reactor is stiπed for 3 hours. To the reactor is added 0.19 mL of 49.4% aq. NaOH and the mixture is stirred for 2 hours. To the mixture is added 0.9 g DABCO dissolved in 12.5 mL DMAC. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of dilution water. The mixture is stiπed for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stiπed 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (225 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 30°C. To the mixture is then added 6.7 grams of thionyl chloride over 30 to 45 minutes. The temperature is maintained below 35°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is stiπed for 0.5 hr and is then charged with 156.6 mL of 4% NaOH wash over a 30 minute period. The addition of the wash is stopped when the pH of the mixture reaches’ 8.0 to 10.0. The bottom aqueous layer is removed at 30°C and any rag layer is retained with the organic layer. To the mixture is charged 175 mL of saturated NaCl wash with agitation. The layers are separated at 30°C and the bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 325 mL of methyl ethyl ketone (MEK) and 13 mL water. Next, the reactor is charged 6.2 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 25 mL of MEK. The mixture is heated to reflux and held for 30 minutes. Approximately 10% of solvent volume is then distilled off. The mixture starts to form crystals during distillation. The mixture is then cooled to 20°C for 1 hour. The off-white crystalline 41 (Form U) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example lib.
Alternate Preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 7.11 grams of 30% sodium hydroxide over a period of 15 to 30 minutes with agitation. The mixture is stiπed for 30 minutes. To the flask is added 9.5 grams of solid 55. The reactor is stiπed for 3 hours. To the mixture is added 1.2 g of solid DABCO. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of water. The mixture is stirred for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained with the organic layer. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stirred 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. The flask is then charged with 225 mL of water and stirred 15 minutes. The layers are allowed to separate at 50°C. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (112.5 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 25°C. To the mixture is then added 7.3 grams of thionyl chloride over 15 to 45 minutes. The temperature of the mixture is maintained above 20°C and below 40°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is then charged with 179.5 mL of 4% NaOH wash over a 30 minute period. The mixture is maintained above 20°C and below 40°C during this time. The addition of the wash is stopped when the pH of the mixture reaches 8.0 to 10.0. The mixture is then allowed to separate at 40°C for at least one hour.

The bottom aqueous layer is removed and any rag layer is retained with the organic layer. To the mixture is charged 200 mL of dilution water. The mixture is stiπed for 15 minutes and then allowed to separate at 40°C for at least one hour. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-2 .

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 350 mL of methyl ethyl ketone (MEK) and 7 mL water. The mixture is stiπed for 15 minutes and the temperature of the mixture is adjusted to 25°C. Next, the reactor is charged with 6.7 grams of solid
diazabicyclo[2.2.2]octane (DABCO). The mixture is maintained at 25°C for three to four hours. It is then heated to 65°C and maintained at that temperature for 30 minutes. The mixture is then cooled to 25°C for 1 hour. The off-white crystalline 41 (Form II) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example 12.
Alternate preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form I of 41

(4R,5R)-27 (2.82 kg dry basis, 4.7 mol) was dissolved in MTBE (9.4 L). The solution of (4R,5R)-22 was passed through a 0.2 mm filter cartridge into the feeding vessel. The flask and was rinsed with MTBE (2 x 2.5 L). The obtained solution as passed through the cartridge filter and added to the solution of (4R,5R)-2 in the feeding vessel. DABCO
(diazabicyclo[2.2.2]octane, 0.784 kg, 7.0 mol) was dissolved in MeOH (14.2 L). The DABCO solution was passed through the filter cartridge into the 100 L nitrogen-flushed reactor. The Pyrex bottle and the cartridge filter were rinsed with MeOH (7.5 L) and the solution was added to the reactor. The (4R,5R)-22 solution was added from the feeding vessel into the reactor at 37°C over a period of 10 min, while stirring. Methanol (6.5 L) was added to the Pyrex bottle and via the cartridge filter added to the feeding vessel to rinse the remaining (4R,5R)-2 into the reactor. The reaction mixture was brought to 50-60°C over 10-20 min and stiπed at that temperature for about 1 h. The mixture was cooled to 20-25°C over a period of 1 h. To the reaction mixture, methyl t-butyl ether (MTBE) (42 L) was added over a period of 1 h and stiπed for a minimum of 1 h at 20 – 25°C. The suspension was filtered through a Buchner funnel. The reactor and the filter cake were washed with MTBE (2 x 14 L). The solids were dried on a rotary evaporator in a 20 L flask at 400 – 12 mbar, 40°C, for 22 h. A white crystalline solid was obtained. The yield of 4 . (Form I) was 3.08 kg (2.97 kg dry, 93.8 %) and the purity 99.7 area % (HPLC; Kromasil C 4, 250 x 4.6 mm column; 0.05% TFA in H2O/0.05% TFA in ACN gradient, UV detection at 215 nm).

Example 12a.
Conversion of Form I of Compound 41 into Form II of Compound 41.

To 10.0 grams of Form I of 4 . in a 400 mL jacketed reactor is added 140 mL of MEK. The reactor is stirred (358 φm) for 10 minutes at 23 °C for 10 minutes and the stirring rate is then changed to 178 φm. The suspension is heated to reflux over 1 hour using a programmed temperature ramp (0.95°C/minute) using batch temperature control (cascade mode). The delta Tmaχ is set to 5°C. The mixture is held at reflux for 1 hour. The mixture is cooled to

25°C. After 3 hours at 25°C, a sample of the mixture is collected by filtration. Filtration is rapid (seconds) and the filtrate is clear and colorless. The white solid is dried in a vacuum oven (80°C, 25 in. Hg) to give a white solid. The remainder of the suspension is stirred at 25°C for 18 hours. The mixture is filtered and the cake starts to shrink as the mother liquor reaches the top of the cake. The filtration is stopped and the reactor is rinsed with 14 mL of MEK. The reactor stirrer speed is increased from 100 to 300 φm to rinse the reactor. The rinse is added to the filter and the solid is dried with a rapid air flow for 5 minutes. The solid is dried in a vacuum oven at 25 in. Hg for 84 hours to give Form II of 4

PATENT

WO 2014144650

Scheme 3:

PAPER

Journal of Medicinal Chemistry (2005), 48(18), 5853-5868

Discovery of Potent, Nonsystemic Apical Sodium-Codependent Bile Acid Transporter Inhibitors (Part 2)

Department of Discovery Chemistry and Department of Cardiovascular Disease, Pharmacia, 700 Chesterfield Parkway W, Chesterfield, Missouri 63017, Office of Science and Technology, Chemical Science Division, Pharmacia, 800 Lindbergh Boulevard, Creve Coeur, Missouri 63167, Department of Pharmaceutical Sciences, Pharmacia, Skokie, Illinois, and Department of Chemistry, University of Missouri, St. Louis, Missouri
J. Med. Chem., 2005, 48 (18), pp 5853–5868
DOI: 10.1021/jm0402162
 

http://pubs.acs.org/doi/abs/10.1021/jm0402162

 

Abstract

Abstract Image

In the preceding paper several compounds were reported as potent apical sodium-codependent bile acid transporter (ASBT) inhibitors. Since the primary site for active bile acid reabsorption is via ASBT, which is localized on the luminal surface of the distal ileum, we reasoned that a nonsystemic inhibitor would be desirable to minimize or eliminate potential systemic side effects of an absorbed drug. To ensure bioequivalency and product stability, it was also essential that we identify a nonhygroscopic inhibitor in its most stable crystalline form. A series of benzothiepines were prepared to refine the structure−activity relationship of the substituted phenyl ring at the 5-position of benzothiepine ring and to identify potent, crystalline, nonhygroscopic, and efficacious ASBT inhibitors with low systemic exposure.

compd R IC50 (nM)b hygroscp I wt gain (%)c hygroscp II % wt gain (%)d crystallinitye
74 OCH2C6H4(p)CH2(N+)DB 0.28 1.59 2.1 yes

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt (74). Following a similar procedure as in General Method B, the title compound 74 was prepared from the corresponding chloromethyl benzyl ether and DABCO as a white solid, mp 223−230 °C (dec); 1H NMR (CDCl3) δ 0.89 (m, 6H), 1.27−1.52 (br m, 10H), 1.63 (m, 1H), 2.20 (m, 1H), 2.81 (s, 6H), 3.06 (ABq, JAB = 15.1 Hz, J = 43.3 Hz, 2H), 3.16 (s, 6H), 3.76 (s, 6H), 4.11 (d, J = 7.7 Hz, 1H), 5.09 (s, 2H), 5.14 (s, 2H), 5.48 (s, 1H), 5.96 (s, 1H), 6.49 (d, J = 8.9 Hz, 1H), 6.99 (d, J = 8.0 Hz, 2H), 7.26 (m, 1H), 7.44 (d, J = 8.0 Hz, 2H), 7.52 (d, J = 7.4 Hz, 2H), 7.68 (d, J = 7.4 Hz, 2H), 7.87 (d, J = 8.9 Hz, 1H). HRMS calcd for C40H56N3O4S:  674.3992; found, 674.4005. Anal. Calcd for C40H56N3O4S:  ‘ C, 67.62; H, 7.95; N, 5.92; S, 4.51. Found:  C, 67.48; H, 8.32; N, 5.85; S, 4.60.

a All compounds were prepared using method B in Scheme 3.b Taurocholate is transported across the baby hamster kidney cell membrane.c % weight gain in a 25 °C, 57% humidity chamber for 2 weeks.d % weight gain in a 40 °C, 80% humidity chamber for 2 weeks.e Crystallinity as determined by X-ray powder diffraction analysis.f (N+)DB is a DABCO terminal group with the quaternary ammonium attached to the linke

ANY ERROR EMAIL amcrasto@gmail.com, +919323115463

PATENT

https://www.google.com/patents/WO1999032478A1?cl=en

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter,
Applicant G.D. Searle & Co.

Example 10. Preparation of enantiomerically-enriched (4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane chloride ( (4R,5R) -XXVII) ♦

Figure imgf000053_0001

( (4R,5R) -XXVII) * = chiral center

Step 1. Preparation of 4-flUoro-2- ( (4- methoxyphenyl) methyl) -phenol To a stirred solution of 23.66 g of 95% sodium hydride (0.94 mol) in 600 mL of dry toluene was added 100.0 g of 4- fluorophenol (0.89 mol) at 0°C. The mixture was stirred at 90°C for 1 hour until gas evolution stopped. The mixture was cooled down to room temperature and a solution of 139.71 g of 3 -methoxybenzyl chloride (0.89 mol) in 400 mL of dry toluene was added. After refluxing for 24 hours, the mixture was cooled to room temperature and quenched with 500 mL of water. The organic layer was separated, dried over MgS04, and concentrated under high vacuum. The remaining starting materials were removed by distillation. The crude dark red oil was filtered through a layer of 1 L of silica gel with neat hexane to yield 53.00 g (25.6%) of the product as a pink solid: *H NMR (CDC13) d 3.79 (s, 3H) , 3.90 (s, 2H) , 4.58 (s, IH) , 6.70-6.74 (m, IH) , 6.79-6.88 (m, 4H) , 7.11-7.16 (m, 2H) .

Step 2. Preparation of 4-fluoro-2- ( (4- methoxyphenyl) methyl) -thiophenol

Step 2a. Preparation of thiocarbamate To a stirred solution of 50.00 g (215.30 mmol) of 4- fluoro-2- ( ( -methoxyphenyl) methyl) -phenol in 500 mL of dry DMF was added 11.20 g of 60% sodium hydride dispersion in mineral oil (279.90 mmol) at 2°C. The mixture was allowed to warm to room temperature and 26.61 g of dimethylthiocarbamoyl chloride (215.30 mmol) was added. The reaction mixture was stirred at room temperature overnight. The mixture was quenched with 100 mL of water in an ice bath. The solution was extracted with 500 mL of diethyl ether. The ether solution was washed with 500 mL of water and 500 mL of brine. The ether solution was dried over MgS04 and stripped to dryness. The crude product was filtered through a plug of 500 mL silica gel using 5% ethyl acetate/hexane to yield 48.00 g (69.8%) of the product as a pale white solid: XH NMR (CDC13) d 3.21 (s, 3H) , 3.46 (s, 3H) , 3.80 (s, 3H) , 3.82 (s, 2H) , 6.78-6.86 (m, 3H) , 6.90- 7.00 (m, 2H) , 7.09 (d, J = 8.7 Hz, 2H) .

Step 2b. Rearrangement and hydrolysis of thiocarbamate to 4-fluoro-2- ( (4 -methoxyphenyl) methyl) -thiophenol A stirred solution of 48.00 g (150.29 mmol) of thiocarbamate (obtained from Step 2a) in 200 mL of diphenyl ether was refluxed at 270°C overnight. The solution was cooled down to room temperature and filtered through 1 L of silica gel with 2 L of hexane to remove phenyl ether. The rearrangement product was washed with 5% ethyl acetate/hexane to give 46.00 g (95.8%) of the product as a pale yellow solid: XH NMR (CDC13) d 3.02 (s, 3H) , 3.10 (s, 3H) , 3.80 (s, 3H) , 4.07 (s, 2H) , 6.82-6.86 (m, 3H) , 6.93 (dt, J = 8.4 Hz, 2.7 Hz, IH) , 7.08 (d, J = 8.7 Hz, 2H) , 7.49 (dd, J = 6.0 Hz, 8.7 Hz, IH) . To a solution of 46.00 g (144.02 mmol) of the rearrangement product (above) in 200 mL of methanol and 200 mL of THF was added 17.28 g of NaOH (432.06 mmol) . The mixture was refluxed under nitrogen overnight . The solvents were evaporated off and 200 mL of water was added. The aqueous solution was washed with 200 mL of diethyl ether twice and placed in an ice bath. The aqueous mixture was acidified to pH 6 with concentrated HCl solution. The solution was extracted with 300 mL of diethyl ether twice. The ether layers were combined, dried over MgS04 and stripped to dryness to afford 27.00 g (75.5%) of the product as a brown oil: XH NMR (CDC13) d 3.24 (s, IH) , 3.80 (s, 3H) , 3.99 (s, 2H) , 6.81-6.87 (m, 4H) , 7.09 (d, J = 8.7 Hz, 2H) , 7.27- 7.33 (m, IH) .

Step 3. Preparation of dibutyl cyclic sulfate

Step 3a. Preparation of 2 , 2-dibutyl-l, 3-propanediol . To a stirred solution of di-butyl-diethylmalonate (Aldrich) (150g, 0.55 mol in dry THF (700ml) in an acetone/dry ice bath was added LAH (1 M THF) 662 ml (1.2 eq. , 0.66 mol) dropwise maintaining the temperature between -20 to 0°C. The reaction was stirred at RT overnight. The reaction was cooled to -20°C and 40 ml of water, and 80 mL of 10% NaOH and 80 ml of water were added dropwise. The resulting suspension was filtered. The filtrate was dried over sodium sulphate and concentrated in vacuo to give diol 598.4 g (yield 95%) as an oil. MS spectra and proton and carbon NMR spectra were consistent with the product.

Step 3b. Preparation of dibutyl cyclic sulfite

A solution of 2 , 2-dibutyl-l, 3-propanediol (103 g, 0.548 0 mol, obtained from Step 3a) and triethylamine (221 g, 2.19 mol) in anhydrous methylene chloride (500 ml) was stirred at 0°C under nitrogen. To the mixture, thionyl chloride (97.8* g, 0.82 mol) was added dropwise and within 5 min the solution turned yellow and then black when the addition was 5 completed within half an hour. The reaction mixture was stirred for 3 hrs. at 0°C. GC showed that there was no starting material left. The mixture was washed with ice water twice then with brine twice . The organic phase was dried over magnesium sulfate and concentrated under vacuum 0 to give 128 g (100%) of the dibutyl cyclic sulfite as a black oil. Mass spectrum (MS) was consistent with the product .

Step 3c. Oxidation of dibutyl cyclic sulfite to 5 dibutyl cyclic sulfate

To a solution of the dibutyl cyclic sulfite (127.5 g , 0.54 mol, obtained from Step 3b) in 600 ml acetonitrile and 500 ml of water cooled in an ice bath under nitrogen was added ruthenium (III) chloride (1 g) and sodium periodate 0 (233 g, 1.08 mol) . The reaction was stirred overnight and the color of the solution turned black. GC showed that there was no starting material left. The mixture was extracted with 300 ml of ether and the ether extract was washed three times with brine. The organic phase was dried over magnesium sulfate and passed through celite. The filtrate was 5 concentrated under vacuum and to give 133 g (97.8%) of the dibutyl cyclic sulfate as an oil. Proton and carbon NMR and MS were consistent with the product.

Step 4. Preparation of aryl-3-hydroxypropylsulfide

10 To a stirred solution of 27.00 g (108.73 mmol) of 4- fluoro-2- ( (4-methoxyphenyl) methyl) thiophenol (obtained from Step 2) in 270 mL of diglyme was added 4.35 g of 60% sodium-, hydride dispersion in mineral oil (108.73 mmol) at 0°C. After gas evolution ceased, 29.94 g (119.60 mmol) of the

15 dibutyl cyclic sulfate (obtained from Step 3c) was added at 0°C and stirred for 10 minutes. The mixture was allowed to warm up to room temperature and stirred overnight. The solvent was evaporated and 200 mL of water was added. The solution was washed with 200 mL of diethyl ether and added

2025 mL of concentrated sulfuric acid to make a 2.0 M solution that was refluxed overnight. The solution was extracted with ethyl acetate and the organic solution was dried over MgS04 and concentrated in vacuo. The crude aryl-3 – hydroxypropylsulfide was purified by silica gel

25 chromatography (Waters Prep 500) using 8% ethyl acetate/hexane to yield 33.00 g (72.5%) of the product as a light brown oil: E NMR (CDC13) d 0.90 (t, J = 7.1 Hz, 6H) , 1.14-1.34 (m, 12H) , 2.82 (s, 2H) , 3.48 (s, 2H) , 3.79 (s, 3H) , 4.10 (s, 2H) , 6.77-6.92 (m, 4H) , 7.09 (d, J = 8.7 Hz,

302H) , 7.41 (dd, J = 8.7 Hz, 5.7 Hz, IH) . Step 5. Preparation of enantiomerically-enriched aryl-3 – hydroxypropylsulfoxide

To a stirred solution of 20.00 g (47.78 mmol) of aryl- 53 -hydroxypropylsulfide (obtained from Step 4) in 1 L of methylene chloride was added 31.50 g of 96% (12?) – ( -) – (8 , 8- dichloro-10-camphor-sulfonyl) oxaziridine (100.34 mmol, Aldrich) at 2°C. After all the oxaziridine dissolved the mixture was placed into a -30 °C freezer for 72 hours. The

10 solvent was evaporated and the crude solid was washed with 1 L of hexane. The white solid was filtered off and the hexane solution was concentrated in vacuo. The crude oil was purified on a silica gel column (Waters Prep 500) using 15% ethyl acetate/hexane to afford 19.00 g (95%) of the

15 enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide as a colorless oil: lH NMR (CDC13) d 0.82-0.98 (m, 6H) , 1.16-1.32 (m, 12H) , 2.29 (d, J – 13.8 Hz, IH) , 2.77 (d, J = 13.5 Hz, IH) , 3.45 (d, J = 12.3 Hz, IH) , 3.69 (d, J = 12.3 Hz, IH) , 3.79 (s, 3H) , 4.02 (q, J = 15.6 Hz, IH) , 6.83-6.93 (m, 3H) ,

207.00 (d, J = 8.1 Hz, 2H) , 7.18-7.23 (m, IH) , 7.99-8.04 (m, IH) . Enantiomeric excess was determined by chiral HPLC on a (2?,2?) -Whelk-0 column using 5% ethanol/hexane as the eluent. It showed to be 78% e.e. with the first eluting peak as the major product.

25

Step 6. Preparation of enantiomerically-enriched aryl-3- propanalsulfoxide

To a stirred solution of 13.27 g of triethylamine (131.16 mmol, Aldrich) in 200 mL dimethyl sulfoxide were

30 added 19.00 g (43.72 mmol) of enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide (obtained from Step 5) and 20.96 g of sulfur trioxide-pyridine (131.16 mmol, Aldrich) at room temperature. After the mixture was stirred at room temperature for 48 hours, 500 mL of water was added to the mixture and stirred vigorously. The mixture was then 5 extracted with 500 mL of ethyl acetate twice. The ethyl acetate layer was separated, dried over MgS04, and concentrated in vacuo. The crude oil was filtered through 500 mL of silica gel using 15% ethyl acetate/hexane to give 17.30 g (91%) of the enantiomerically-enriched aryl-3-

10 propanalsulfoxide as a light orange oil: lE NMR (CDC13) d 0.85-0.95 (m, 6H) , 1.11-1.17 (m, 4H) , 1.21-1.39 (m, 4H) , 1.59-1.76 (m, 4H) , 1.89-1.99 (m, IH) , 2.57 (d, J = 14.1 Hz, IH) , 2.91 (d, J = 13.8 Hz, IH) , 3.79 (s, 3H) , 3.97 (d, J = 15.9 Hz, IH) , 4,12 (d, J = 15.9 Hz, IH) , 6.84-6.89 (m, 3H) ,

157.03 (d, J = 8.4 Hz, 2H) , 7.19 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 8.02 (dd, J = 8.7 Hz, 5.7 Hz, IH) , 9.49 (s, IH) .

Step 7. Preparation of the enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R, 5R)

20 To a stirred solution of 17.30 g (39.99 mmol) of enantiomerically-enriched aryl-3 -propanalsulfoxide (obtained from Step 6) in 300 mL of dry THF at -15°C was added 48 mL of 1.0 M potassium t-butoxide in THF (1.2 equivalents) under nitrogen. The solution was stirred at -15°C for 4 hours.

25 The solution was then quenched with 100 mL of water and neutralized with 4 mL of concentrated HCl solution at 0°C. The THF layer was separated, dried over MgS04, and concentrated in vacuo. The enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R,5R) was purified by

30 silica gel chromatography (Waters Prep 500) using 15% ethyl acetate/hexane to give 13.44 g (77.7%) of the product as a white solid: ‘H NMR (CDC13) d 0.87-0.97 (m, 6H) , 1.16-1.32 (m, 4H) , 1.34-1.48 (m, 4H) , 1.50-1.69 (m, 4H) , 1.86-1.96 (m, IH) , 2.88 (d, J = 13.0 Hz, IH) , 3.00 (d, J = 13.0 Hz, IH) , 3.85 (s, 3H) , 4.00 (s, IH) , 4.48 (s, IH) , 6.52 (dd, J = 9.9 5Hz, 2.4 Hz, IH) , 6.94 (d, J = 9 Hz, 2H) , 7.13 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 7.38 (d, J = 8.7 Hz, 2H) , 7.82 (dd, J = 8.7 Hz, 5.7 Hz, IH) .

Step 8. Preparation of enantiomerically-enriched

10 tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R)

To a stirred solution of 13.44 g (31.07 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (obtained from Step 7) in 150 mL of methylene chloride was added 9.46 g of 68% m-chloroperoxybenzoic acid (37.28 mmol,

15 Sigma) at 0 °C. After stirring at 0 °C for 2 hours, the mixture was allowed to warm up to room temperature and stirred for 4 hours. 50 mL of saturated Na2S03 was added into the mixture and stirred for 30 minutes. The solution was then neutralized with 50 mL of saturated NaHC03 solution.

20 The methylene chloride layer was separated, dried over MgS04, and concentrated in vacuo to give 13.00 g (97.5%) of the enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (4R,5R) as a light yellow solid: ‘H NMR (CDC13) d 0.89-0.95 (m, 6H) , 1.09-1.42 (m, 12H) , 2.16-2.26 (m, IH) ,

253.14 (q, J = 15.6 Hz, IH) , 3.87 (s, 3H) , 4.18 (s, IH) , 5.48 (s, IH) , 6.54 (dd, J = 10.2 Hz, 2.4 Hz, IH) , 6.96-7.07 (m, 3H) , 7.40 (d, J = 8.1 Hz, 2H) , 8.11 (dd, J = 8.6 Hz, 5.9 Hz, IH) .

30 Step 9. Preparation of enantiomerically-enriched 7-

(dimethylamino) tetrahydrobenzothiepine-1 , 1-dioxide (4R.5R) – To a solution of 13.00 g (28.98 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (obtained from Step 8) in 73 mL of dimethylamine (2.0 M in THF, 146 mmol) in a Parr Reactor was added ca . 20 5 mL of neat dimethylamine . The mixture was sealed and stirred at 110 °C overnight, and cooled to ambient temperature. The excess dimethylamine was evaporated. The crude oil was dissolved in 200 mL of ethyl acetate and washed with 100 mL of water, dried over MgS04 and

10 concentrated in vacuo. Purification on a silica gel column (Waters Prep 500) using 20% ethyl acetate/hexane gave 12.43 g (90.5%) of the enantiomerically- enriched 7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R) as a colorless solid: *H NMR (CDC13) d 0.87-0.93 (m, 6H) ,

151.10-1.68 (m, 12H) , 2.17-2.25 (m, IH) , 2.81 (s, 6H) , 2.99 (d, J = 15.3 Hz, IH) , 3.15 (d, J = 15.3 Hz, IH) , 3.84 (s, 3H) , 4.11 (d, J = 7.5 Hz, IH) , 5.49 (s, IH) , 5.99 (d, J = 2.4 Hz, IH) , 6.51 (dd, J = 8.7 Hz, 2.4 Hz, IH) , 6.94 (d, J = 8.7 Hz, 2H) , 7.42 (d, J = 8.4 Hz, 2H) , 7.90 (d, J = 8.7 Hz,

20 IH) . The product was determined to have 78% e.e. by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. Recrystallization of this solid from ethyl acetate/hexane gave 1.70 g of the racemic product. The remaining solution was concentrated and recrystallized to

25 give 9.8 g of colorless solid. Enantiomeric excess of this solid was determined by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. It showed to have 96% e.e with the first eluting peak as the major product.

30 Step 10: Demethylation of 5- (4 ‘ -methoxyphenyl) -7-

(dimethylamino) tetrahydrobenzothiepine-1.1-dioxide (4R, 5R) To a solution of 47 g (99 mmol) of enantiomeric- enriched (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (obtained from Step 9) in 500 mL of methylene chloride at -10 °C was added dropwise a solution of boron tribromide (297 mL, 1M in methylene chloride, 297 mmol), and the resulting solution was stirred cold (-5 °C to 0 °C) for 1 hour or until the reaction was complete. The reaction was cooled in an acetone-dry ice bath at -10 °C, and slowly quenched with 300 mL of water. The mixture was warmed to 10 °C, and further diluted with 300 mL of saturated sodium bicarbonate solution to neutralize the mixture. The aqueous layer was separated and extracted with 300 mL of methylene chloride, and the combined extracts were washed with 200 mL of water, brine, dried over MgS04 and concentrated in vacuo. The residue was dissolved in 500 mL of ethyl acetate and stirred with 50 mL of glacial acetic acid for 30 minutes at ambient temperature. The mixture was washed twice with 200 mL of water, 200 mL of brine, dried over MgS04 and concentrated in vacuo to give the crude 4-hydroxyphenyl intermediate. The solid residue was recrystallized from methylene chloride to give 37.5 g (82%) of the desired (4R, 5R) -5- (4′ – hydoxyphenyl) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1- dioxide as a white solid: *H NMR (CDC13) d 0.84-0.97 (m, 6H) , 1.1-1.5 (m, 10H) , 1.57-1.72 (m, IH) , 2.14-2.28 (m, IH) , 2.83 (s, 6H) , 3.00 (d, J = 15.3 Hz, IH) , 3.16 (d, J – 15.3 Hz, IH) , 4.11 (s, 2H) , 5.48 (s, IH) , 6.02 (d, J – 2.4 Hz, IH) , 6.55 (dd, J = 9, 2.4 Hz, IH) , 6.88 (d, 8 , 7 Hz , 2H) , 7.38 (d, J – 8.7 Hz, 2H) , 7.91 (d, J = 9 Hz, 2H) .

Step 11: Preparation of enantiomerically-enriched chlorobenzyl intermediate Treat a solution of enantiomerically-enriched (4R,5R)- 5- (4′ -hydoxypheny1) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (5.0 g, 10.9 mmol, obtained from Step 10) in acetone (100 mL) at 25 °C under N2 with powdered 5 K2C03 (2.3 g, 16.3 mmol, 1.5 eq.) and a, a’ -dichloro-p-xylene (6.7 g, 38.1 mmol, 3.5 eq.) . Stir the resulting solution at 65 °C for about 48 hours. Cool the reaction mixture to 25 °C and concentrate to 1/5 of original volume. Dissolve the residue in EtOAc (150 mL) and wash with water (2 x 150 mL) .

10 Extract the aqueous layer with EtOAc (2 x 150 mL) and wash the combined organic extracts with saturated aqueous NaCI (2 x 150 mL. Dry the combined extracts with MgS04 and concentrate in vacuo to provide the crude product . Purification by flash chromatography (5.4 x 45 cm silica,

1525-40% EtOAc/hexane) will afford the enantiomerically- enriched chlorobenzyl intermediate .

Step 12: Preparation of enantiomerically-enriched (4R.5R)- 1- r [4- [ [4- [3 , 3-Dibutyl-7- (dimethylamino) -2,3 , 4 , 5-tetrahvdro-

204 -hydroxy-1.1-dioxido-1-benzothiepin-5- yl] phenoxy] methyll phenyl! methyl] -4-aza-l- azoniabicyclo f2.2.2] octane chloride (XXVII)

Treat a solution of the enantiomerically-enriched chlorobenzyl intermediate (4.6 g, 7.7 mmol, obtained from

25 above in Step 11) in acetonitrile (100 mL) at 25 °C under N2 with diazabicyclo [2.2.2] -octane (DABCO, 0.95 g, 8.5 mmol, 1.1 eq.) and stir at 35 °C for 2 hours. Collect the precipitated solid and wash with CH3CN. Recrystallization from CH3OH/Et20 will give the desired title compound (XXVII) .

ANY ERROR,  EMAIL amcrasto@gmail.com, +919323115463

///////////FDA, Breakthrough Designation,  Shire, Rare GI Therapies, SHP625, maralixibat, progressive familial intrahepatic , Maralixibat chloride, 228113-66-4, UNII: V78M04F0XC, LUM 001, Lopixibat chloride, cholestasis type 2 (PFIC2), Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

CCCCC1(CS(=O)(=O)c2ccc(cc2[C@H]([C@H]1O)c3ccc(cc3)OCc4ccc(cc4)C[N+]56CCN(CC5)CC6)N(C)C)CCCC.[Cl-]

wdt-1

NEW DRUG APPROVALS

ONE TIME

$10.00

Ponesimod


Ponesimod.svg

Ponesimod

Phase III

MW 460.97, C23 H25 Cl N2 O4 S

A sphingosine-1-phosphate receptor 1 (S1P1) agonist potentially for the treatment of multiple sclerosis.

  • (2Z,5Z)-5-[[3-Chloro-4-[(2R)-2,3-dihydroxypropoxy]phenyl]methylene]-3-(2-methylphenyl)-2-(propylimino)-4-thiazolidinone
  • 5-[3-Chloro-4-[((2R)-2,3-dihydroxypropyl)oxy]benz-(Z)-ylidene]-2-((Z)-propylimino)-3-(o-tolyl)thiazolidin-4-one
  • ACT 128800

ACT-128800; RG-3477; R-3477

CAS No. 854107-55-4

update 18/3/21 FDA APPROVEDAS PONVORY

SYNTHESIS

STR1

Ponesimod

str1

str1

NMR CDCL3 FROM NET

STR1

STR1

STR1

STR1

STR1

SEE……http://www.slideserve.com/truda/discovery-of-the-novel-orally-active-s1p-1-receptor-agonist-act-128800-ponesimod

Ponesimod (INN, codenamed ACT-128800) is an experimental drug for the treatment of multiple sclerosis (MS) and psoriasis. It is being developed by Actelion.

The first oral treatment for relapsing multiple sclerosis, the nonselective sphingosine-1-phosphate receptor (S1PR) modulator fingolimod, led to identification of a pivotal role of sphingosine-1-phosphate and one of its five known receptors, S1P1R, in regulation of lymphocyte trafficking in multiple sclerosis. Modulation of S1P3R, initially thought to cause some of fingolimod’s side effects, prompted the search for novel compounds with high selectivity for S1P1R. Ponesimod is an orally active, selective S1P1R modulator that causes dose-dependent sequestration of lymphocytes in lymphoid organs. In contrast to the long half-life/slow elimination of fingolimod, ponesimod is eliminated within 1 week of discontinuation and its pharmacological effects are rapidly reversible. Clinical data in multiple sclerosis have shown a dose-dependent therapeutic effect of ponesimod and defined 20 mg as a daily dose with desired efficacy, and acceptable safety and tolerability. Phase II clinical data have also shown therapeutic efficacy of ponesimod in psoriasis. These findings have increased our understanding of psoriasis pathogenesis and suggest clinical utility of S1P1R modulation for treatment of various immune-mediated disorders. A gradual dose titration regimen was found to minimize the cardiac effects associated with initiation of ponesimod treatment. Selectivity for S1P1R, rapid onset and reversibility of pharmacological effects, and an optimized titration regimen differentiate ponesimod from fingolimod, and may lead to better safety and tolerability. Ponesimod is currently in phase III clinical development to assess efficacy and safety in relapsing multiple sclerosis. A phase II study is also ongoing to investigate the potential utility of ponesimod in chronic graft versus host disease.http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4707431/

Biology and pharmacology of sphingosine-1-phosphate receptor 1

The past decades have witnessed major advances in the treatment of autoimmune and chronic inflammatory diseases. A plethora of novel therapies targeting specific molecules involved in the inflammatory or immune system activation cascades have become available. These have significantly increased our understanding of disease pathogenesis and improved the management of immune-mediated disorders. However, most of the targeted therapies are biological drugs which need to be injected, are eliminated slowly (e.g. over several weeks) and can lose efficacy or tolerability due to their potential immunogenicity. In an attempt to overcome these hurdles, pharmaceutical research has made considerable efforts to develop novel oral targeted therapies for autoimmune and chronic inflammatory diseases.

Sphingosine-1-phosphate receptor 1 (S1P1R) is one of five known G protein-coupled receptors with nanomolar affinity for the lysophospholipid sphingosine-1-phosphate (S1P), which is generated through physiologic metabolism of the cell membrane constituent sphingomyelin by all cells [Brinkmann, 2007]. S1P receptors, including S1P1R, are widely expressed in many tissues [Chun et al. 2010]. S1P1R expression on lymphocytes controls their egress from thymus and secondary lymphoid organs [Cyster and Schwab, 2012]. Lymphocyte egress requires a gradient of S1P concentration, which is established by a high S1P concentration in blood and lymph compared with a low concentration in the interstitial fluid of lymphoid organs [Grigorova et al. 2009].

Synthetic S1P1 receptor modulators disrupt the interaction of the physiologic S1P ligand with S1P1R by promoting initial activation followed by sustained internalization and desensitization of S1P1R [Hla and Brinkmann, 2011; Pinschewer et al. 2011]. Experiments conducted in animal models of transplant rejection, multiple sclerosis, lupus erythematosus, arthritis and inflammatory bowel disease with the first-generation, nonselective S1P receptor modulator, fingolimod, have demonstrated the potential efficacy of this mode of action across several immune-mediated chronic inflammatory conditions [Brinkmann, 2007]. Fingolimod is a structural analog of sphingosine that is phosphorylated in the body by a sphingosine kinase to generate the bioactive form of the drug, fingolimod phosphate, which binds to multiple S1P receptors [Brinkmann, 2007]. Clinical trials in multiple sclerosis (MS) have confirmed the efficacy of fingolimod in relapsing MS, but not in primary progressive disease, and led to the approval of the first oral medication for the treatment of relapsing forms of MS in 2010 [Kappos et al. 2010].

The mechanism of action of fingolimod has increased our understanding of MS pathogenesis. T and B cells, but not natural killer (NK) cells, express functional S1P1R and are affected by fingolimod [Cyster and Schwab, 2012]. Furthermore, S1P1R is differentially expressed and regulated in functionally distinct subsets of lymphocytes and fingolimod has been shown to predominantly affect naïve T cells and central memory T cells (TCM) while sparing effector memory T cells (TEM), and terminally differentiated effector T cells (TE) in patients with relapsing MS [Mehling et al. 2008, 2011]. This has raised the possibility that, at least in MS, retention of TCM cells, which include pro-inflammatory T helper 17 (Th17) cells, by fingolimod may prevent their accumulation in the cerebrospinal fluid (CSF) and subsequent differentiation to TE cells in the central nervous system (CNS) [Hla and Brinkmann, 2011]. The effects of S1P1R modulation on B cells are less well defined. Recent data from patients with relapsing MS have shown predominant reduction of memory B cells and recently activated memory B cells (CD38int-high) in peripheral blood after treatment with fingolimod [Claes et al. 2014; Nakamura et al. 2014]. As memory B cells are implicated in the pathogenesis of MS and other autoimmune diseases, these observations suggest another potential mechanism underlying the therapeutic effects of S1P1R modulators.

Astrocytes, microglia, oligodendrocytes and neurons express various S1P receptors including S1P1R, S1P3R and S1P5R. Fingolimod has been shown to penetrate the CNS tissues and in vitro studies have shown activation of astrocytes and oligodendrocytes by fingolimod [Foster et al. 2007]. Conditional deletion of S1P1R on neural cells in mice reduced the severity of experimental autoimmune encephalomyelitis (EAE) and reductions in the clinical scores were paralleled by decreased demyelination, axonal loss and astrogliosis [Choi et al. 2011]. Unfortunately, there was no beneficial effect in a recently completed, large study of fingolimod in patients with primary progressive MS [Lublin et al. 2015], suggesting that the direct effect on CNS cells alone may not be sufficient. Taken together, these data suggest the possibility of a direct beneficial effect of S1P1R modulation in the brain of patients with relapsing MS [Dev et al. 2008]; however, its contribution to efficacy relative to the immunological effects remains unclear.

Initial studies in rodents suggested that modulation of S1P3R on cardiac myocytes by fingolimod was associated with a reduction of heart rate (HR) by activation of G-protein-coupled inwardly rectifying potassium channels (GIRK) that regulate pacemaker frequency, and the shape and duration of action potentials [Koyrakh et al. 2005; Camm et al. 2014]. Modulation of S1P2R and S1P3R on myofibroblasts by fingolimod was also shown to stimulate extracellular matrix synthesis [Sobel et al. 2013]. Modulation of these receptors on vascular smooth muscle cells appeared to be associated with vasoconstriction, leading to the slight increase in blood pressure observed with fingolimod treatment [Salomone et al. 2003; Watterson et al. 2005; Hu et al. 2006; Lorenz et al. 2007; Kappos et al. 2010]. These observations raised the possibility that some side effects associated with fingolimod treatment could be avoided by more selective S1P1R modulators, thus triggering the search for novel compounds.

Currently, there are several selective S1P1R modulators in clinical development [Gonzalez-Cabrera et al.2014; Subei and Cohen, 2015]. Here we review data and the development status of ponesimod, a selective S1P1R modulator developed by Actelion Pharmaceuticals Ltd.http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4707431/

Ponesimod, a selective, rapidly reversible, orally active, sphingosine-1-phosphate receptor modulator

Ponesimod (ACT-128800 (Z,Z)-5-[3-chloro-4-(2R)-2,3-dihydroxy-propoxy)-benzylidene]-2-propylimino-3-o-tolylthiazolidin-4-one) is a selective, rapidly reversible, orally active, S1P1R modulator. Ponesimod emerged from the discovery of a novel class of S1P1R agonists based on the 2-imino-thiazolidin-4-one scaffold (Figure 1) [Bolli et al. 2010]. Ponesimod activates S1P1R with high potency [half maximal effective concentration (EC50) of 5.7 nM] and selectivity. Relative to the potency of S1P, the potency of ponesimod is 4.4 higher for S1P1R and 150-fold lower for S1P3R, resulting in an approximately 650-fold higher S1P1R selectivity compared with the natural ligand.

Figure 1.

Chemical structure of ponesimod, C23H25N2O4CIS (molecular weight 460.98).http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4707431/

Clinical trials

In a 2009–2011 Phase II clinical trial including 464 MS patients, ponesimod treatment resulted in fewer new active brain lesions thanplacebo, measured during the course of 24 weeks.[3][4]

In a 2010–2012 Phase II clinical trial including 326 patients with psoriasis, 46 or 48% of patients (depending on dosage) had a reduction of at least 75% Psoriasis Area and Severity Index (PASI) score compared to placebo in 16 weeks.[3][5]

SEE https://clinicaltrials.gov/ct2/show/NCT02425644

Adverse effects

Common adverse effects in studies were temporary bradycardia (slow heartbeat), usually at the beginning of the treatment,dyspnoea (breathing difficulties), and increased liver enzymes (without symptoms). No significant increase of infections was observed under ponesimod therapy.[3] QT prolongation is detectable but was considered to be too low to be of clinical importance in a study.[6]

Mechanism of action

Like fingolimod, which is already approved for the treatment of MS, ponesimod blocks the sphingosine-1-phosphate receptor. This mechanism prevents lymphocytes (a type of white blood cells) from leaving lymph nodes.[3] Ponesimod is selective for subtype 1 of this receptor, S1P1.[7]

PAPER

Bolli, Martin H.; Journal of Medicinal Chemistry 2010, V53(10), P4198-4211 CAPLUS

2-Imino-thiazolidin-4-one Derivatives as Potent, Orally Active S1P1Receptor Agonists

Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
J. Med. Chem., 2010, 53 (10), pp 4198–4211
DOI: 10.1021/jm100181s
Publication Date (Web): May 06, 2010
Copyright © 2010 American Chemical Society
*To whom correspondence should be addressed. Phone: + 41 61 565 65 70. Fax: + 41 61 565 65 00. E-mail:martin.bolli@actelion.com.
Abstract Image

Sphingosine-1-phosphate (S1P) is a widespread lysophospholipid which displays a wealth of biological effects. Extracellular S1P conveys its activity through five specific G-protein coupled receptors numbered S1P1 through S1P5. Agonists of the S1P1 receptor block the egress of T-lymphocytes from thymus and lymphoid organs and hold promise for the oral treatment of autoimmune disorders. Here, we report on the discovery and detailed structure−activity relationships of a novel class of S1P1 receptor agonists based on the 2-imino-thiazolidin-4-one scaffold. Compound 8bo (ACT-128800) emerged from this series and is a potent, selective, and orally active S1P1 receptor agonist selected for clinical development. In the rat, maximal reduction of circulating lymphocytes was reached at a dose of 3 mg/kg. The duration of lymphocyte sequestration was dose dependent. At a dose of 100 mg/kg, the effect on lymphocyte counts was fully reversible within less than 36 h. Pharmacokinetic investigation of8bo in beagle dogs suggests that the compound is suitable for once daily dosing in humans.

(Z,Z)-5-[3-Chloro-4-((2R)-2,3-dihydroxy-propoxy)-benzylidene]-2-propylimino-3-o-tolyl-thiazolidin-4-one (8bo)

…………..DELETED…………… column chromatography on silica gel eluting with heptane:ethyl acetate 1:4 to give the title compound (1.34 g, 37%) as a pale-yellow foam.
1H NMR (CDCl3): δ 0.94 (t, J = 7.3 Hz, 3 H), 1.58−1.70 (m, 2 H), 2.21 (s, 3 H), 3.32−3.48 (m, 2 H), 3.82−3.95 (m, 3 H), 4.12−4.27 (m, 4 H), 7.07 (d, J = 8.8 Hz, 1 H), 7.21 (d, J = 7.0 Hz, 1 H), 7.31−7.39 (m, 3 H), 7.49 (dd, J = 8.5, 2.0 Hz, 1 H), 7.64 (d, J= 2.0 Hz, 1 H), 7.69 (s, 1 H).
13C NMR (CDCl3): δ 11.83, 17.68, 23.74, 55.42, 63.46, 69.85, 70.78, 133.48, 120.75, 123.71, 127.05, 128.25, 128.60, 129.43, 130.06, 131.13, 131.50, 134.42, 136.19, 146.98, 154.75, 166.12. LC-MS (ES+): tR 0.96 min. m/z: 461 (M + H).
HPLC (ChiralPak AD-H, 4.6 mm × 250 mm, 0.8 mL/min, 70% hexane in ethanol): tR 11.8 min. Anal. (C23H25N2O4SCl): C, H, N, O, S, Cl.

PATENT

WO 2014027330

https://www.google.com/patents/WO2014027330A1?cl=3Den

The present invention relates inter alia to a new process for the preparation of (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one (hereinafter also referred to as the “COMPOUND” or “compound (2)”), especially in crystalline form C which form is described in WO 2010/046835. The preparation of COMPOUND and its activity as immunosuppressive agent is described in WO 2005/054215. Furthermore, WO 2008/062376 describes a new process for the preparation of (2Z,5Z)-5-(3-chloro-4-hydroxy-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one which can be used as an intermediate in the preparation of COMPOUND.

Example 1 a) below describes such a process of preparing (2Z,5Z)-5-(3-chloro-4-hydroxy-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one according to WO 2008/062376. According to WO 2008/062376 the obtained (2Z,5Z)-5-(3-chloro-4-hydroxy-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one can then be transformed into COMPOUND by using standard methods for the alkylation of phenols. Such an alkylation is described in Example 1 b) below. Unfortunately, this process leads to the impurity (2Z,5Z)-5-(3-chloro-4-((1 ,3-dihydroxypropan-2-yl)oxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one which is present in about 2% w/w in the crude product (see Table 1 ) and up to 6 recrystallisations are necessary in order to get this impurity below 0.4% w/w (see Tables 1 and 2) which is the specified limit based on its toxicological qualification.

the obtained (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde (1 ) with 2-[(Z)-propylimino]-3-o-tolyl-thiazolidin-4-one to form (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one (2):


.

The reaction of (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde (1 ) with 2-[(Z)-propylimino]-3-o-tolyl-thiazolidin-4-one can be performed under conditions which are typical for a Knoevenagel condensation. Such conditions are described in the literature for example in Jones, G., Knoevenagel Condensation in Organic Reaction, Wiley: New York, 1967, Vol. 15, p 204; or Prout, F. S., Abdel-Latif, A. A., Kamal, M. R., J. Chem. Eng. Data, 2012, 57, 1881-1886.

2-[(Z)-Propylimino]-3-o-tolyl-thiazolidin-4-one can be prepared as described in WO 2008/062376, preferably without the isolation and/or purification of intermediates such as the thiourea intermediate that occurs after reacting o-tolyl-iso-thiocyanate with n-propylamine. Preferably 2-[(Z)-propylimino]-3-o-tolyl-thiazolidin-4-one obtained according to WO 2008/062376 is also not isolated and/or purified before performing the Knoevenagel condensation, i.e. before reacting 2-[(Z)-propylimino]-3-o-tolyl-thiazolidin-4-one with (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde (1 ), i.e. in a preferred embodiment compound (2) is prepared in a one-pot procedure analogous to that described in WO 2008/062376.

Example 1 : (2Z,5Z)-5-(3-Chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one

a) Preparation of (2Z,5Z)-5-(3-chloro-4-hydroxy-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one:

Acetic acid solution: To acetic acid (149.2 mL) are added sodium acetate (1 1 .1 1 g, 2.00 eq.) and 3-chloro-4-hydroxybenzaldehyde (10.60 g, 1.00 eq.) at 20 °C. The mixture is stirred at 20 °C until complete dissolution (2 to 3 h).

n-Propylamine (4.04 g, 1.00 eq.) is added to a solution of o-tolyl-iso-thiocyanate (10 g, 1.00 eq.) in dichloromethane (100 mL) at 20 °C. The resulting pale yellow solution is agitated for 40 min at 20 °C before IPC (conversion specification≥ 99.0 %). The reaction is cooled to -2 °C. Bromoacetyl bromide (13.53 g, 1.00 eq.) is added and the resulting solution is stirred for 15 min at -2 °C. Pyridine (10.92 g, 2.05 eq.) is then added slowly at -2 °C. The intensive yellow reaction mixture is stirred for 15 min at -2 °C before IPC (conversion specification≥ 93.0 %). 70 mL of dichloromethane are distilled off under atmospheric pressure and jacket temperature of 60 °C. The temperature is adjusted to 42 °C and the acetic acid solution is added to the reaction mixture. The resulting solution is heated to 58 °C and stirred at this temperature for 15 h before IPC (conversion specification≥ 95 %). 25 mL of solvents are distilled off under vacuum 900 – 500 mbars and jacket temperature of 80 °C. The temperature is adjusted to 60 °C and water (80.1 mL) is added to the reaction mixture over 1 h. The resulting yellow suspension is stirred at 60 °C for 30 min. The suspension is cooled to 20 °C over 1 h and stirred at this temperature for 30 min.

The product is filtered and washed with a mixture of acetic acid (30 mL) and water (16 mL) and with water (50 mL) at 20 °C. The product is dried under vacuum at 50 °C for 40 h to afford a pale yellow solid; yield 25.93 g (78 %).

b) Preparation of crude (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one:

To a suspension of (2Z,5Z)-5-(3-chloro-4-hydroxy-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one (10.00 g, 1.00 eq.) in ethanol (47.2 mL) is added (R)-3-chloro-1 ,2-

propanediol (3.37 g, 1.18 eq.) at 20 °C. Potassium tert-butoxide (3.39 g, 1.13 eq.) is added in portions at 20 °C. The resulting fine suspension is stirred at 20 °C for 25 min before being heated to reflux (88 °C). The reaction mixture is stirred at this temperature for 24 h before IPC (conversion specification≥ 96.0 %). After cooling down to 60 °C, acetonitrile (28.6 mL) and water (74.9 mL) are added. The resulting clear solution is cooled from 60 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.010 g, 0.001 eq.; crystalline form C can be prepared as described in WO 2010/046835) are added at 50 °C. The suspension is heated from 0 °C to 50 °C, cooled to 0 °C over 6 h and stirred at this temperature for 12 h.

The product is filtered and washed with a mixture of acetonitrile (23.4 mL) and water (23.4 mL) at 0 °C. The product is dried under vacuum at 45 °C for 24 h to afford a pale yellow solid; yield 1 1.91 g (84 %).

c) Purification of (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one:

Recrystallisation I: The crude (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one (10 g) is dissolved in acetonitrile (30 mL) at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed with acetonitrile at -10 °C (2 x 12.8 mL).

Recrystallisation II: The wet product is dissolved in acetonitrile (27.0 mL) at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed with acetonitrile at -10 °C (2 x 1 1.3 mL).

Recrystallisation III: The wet product is dissolved in acetonitrile (24.3 mL) at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4- one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed with acetonitrile at -10 °C (2 x 10.1 mL).

Recrystallisation IV: The wet product is dissolved in acetonitrile (21.9 mL) at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed with acetonitrile at -10 °C (2 x 9.1 mL).

Recrystallisation V: The wet product is dissolved in acetonitrile (19.7 mL) at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h. During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed with acetonitrile at -10 °C (2 x 8.2 mL).

Recrystallisation VI: The wet product is dissolved in acetonitrile (23.9 mL) at 70 °C. Water (20 mL) is added at 70 °C. The reaction mixture is cooled from 70 °C to 0 °C over 2 h.

During the cooling ramp, (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2- (propylimino)-3-(o-tolyl)thiazolidin-4-one seeds of crystalline form C (0.0075 g, 0.00075 eq.) are added at 50 °C. The suspension is heated up to 52 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h. The product is filtered and washed twice with a mixture of acetonitrile (4.5 mL) and water (4.5 mL) at -10 °C.

The product is dried under vacuum at 45 °C for 24 h to afford a pale yellow solid; yield: 7.0 g (70 %).

Example 2: (R)-3-Chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde

Potassium tert-butoxide (1 18 g, 1.20 eq.) is added to n-propanol (963 mL) followed by 3-chloro-4-hydroxybenzaldehyde (137 g, 1.00 eq.). To the mixture is added (R)-3-chloro-1 ,2-propanediol (126 g, 1.30 eq.). The suspension is heated to 90 °C and stirred at this temperature for 17 h. Solvent (500 mL) is distilled off at 120 °C external temperature and reduced pressure. Water is added (1.1 L) and solvent (500 mL) is removed by distillation. The turbid solution is cooled to 20 °C. After stirring for one hour a white suspension is obtained. Water (500 mL) is added and the suspension is cooled to 10 °C. The suspension is filtered and the resulting filter cake is washed with water (500 mL). The product is dried at 50 °C and reduced pressure to yield 149 g of a white solid (73%), which is (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde in crystalline form A.

Example 3: (R)-3-Chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde

Potassium tert-butoxide (8.60 g, 1.20 eq.) is added to n-propanol (70 mL) below 15 °C, the temperature is allowed to rise. After the addition the temperature is corrected again to below 15 °C before addition of 3-chloro-4-hydroxybenzaldehyde (10 g, 1 .00 eq.). The suspension is heated to 40 °C and stirred for 30 min. (R)-3-Chloro-1 ,2-propanediol (9.18 g, 1.30 eq.) is added at 40 °C. The resulting suspension is heated to 60 °C and stirred at this temperature for 15 h then heated to 94 °C till meeting the IPC-specification (specification conversion≥ 90.0 %). The mixture is cooled to 30 °C and n-propanol is partially distilled off (-50 mL are distilled off) under reduced pressure and a maximum temperature of 50 °C, the jacket temperature is not allowed to raise above 60 °C.

Water (81 mL) is added and a second distillation is performed under the same conditions (24 mL are distilled off). The mixture is heated till homogeneous (maximum 54 °C) and then cooled to 24 °C. At 24 °C the mixture is seeded with crystalline (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde of form A (0.013 g, 0.00085 eq.). How to obtain the crystalline seeds is described in Examples 2 and 5. The reaction mixture is cooled to 0 °C over 7.5 h.

The product is filtered and washed with water (2 x 35 mL) and once with methyl tert-butyl ether (20 mL) at 5 °C. The product is dried under vacuum at 40 °C for 20 h to afford an off-white solid; yield: 10.6 g (72 %), which is (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde in crystalline form A.

Example 4: (2Z,5Z)-5-(3-Chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)- 3-(o-tolyl)thiazolidin-4-one

a) Preparation of crude (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one:

n-Propylamine (5.23 g, 1.32 eq.) is added to a solution of o-tolyl-iso-thiocyanate (10 g, 1.00 eq.) in dichloromethane (100 mL) at 20 °C. The resulting pale yellow solution is agitated for 15 min at 20 °C before IPC (conversion specification≥ 99.0 %). The reaction is cooled to -2 °C. Bromoacetyl bromide (14.88 g, 1.10 eq.) is added and the resulting solution is stirred for 15 min at -2 °C. Pyridine (10.92 g, 2.05 eq.) is then added slowly at -2 °C. The intensive yellow reaction mixture is stirred for 15 min at -2 °C before IPC (conversion specification≥ 93.0 %). Dichloromethane is partially distilled off (66 mL are distilled off) under atmospheric pressure and jacket temperature of 60 °C. Ethanol (1 1 1.4 mL), sodium acetate (12.75 g, 2.30 eq.) and (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde from Example 3 (14.38 g, 0.93 eq.) are added. The remaining dichloromethane and a part of ethanol are distilled off (49.50 mL are distilled off) under atmospheric pressure and jacket temperature up to 85 °C. The reaction mixture (orange suspension) is stirred for 3 – 5 h under reflux (78 °C) before IPC (conversion specification≥ 97.0 %).

Water (88.83 mL) is added and the temperature adjusted to 40 °C before seeding with micronized (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one in crystalline form C (0.075 g, 0.0024 eq.). The reaction mixture is cooled to 0 °C over 5 h, heated up to 40 °C, cooled to 0 °C over 6 h and stirred at this temperature for 2 h.

The product is filtered and washed with a 1 :1 ethanohwater mixture (2 x 48 mL) at 0 °C. The product is dried under vacuum at 45 °C for 10 h to afford a pale yellow solid; yield: 24.71 g (86 %).

b) Purification of (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one:

The crude (2Z,5Z)-5-(3-chloro-4-((R)-2,3-dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one (10 g) is dissolved in ethanol (40 mL) at 70 °C. The temperature is adjusted at 50 °C for seeding with micronised (2Z,5Z)-5-(3-chloro-4-((R)-2,3- dihydroxypropoxy)benzylidene)-2-(propylimino)-3-(o-tolyl)thiazolidin-4-one in crystalline form C (0.016 g, 0.0016 eq.). The reaction mixture is cooled from 50 °C to 0 °C over 4 h, heated up to 50 °C, cooled to 0 °C over 6 h and agitated at this temperature for 2 h.

The product is filtered and washed with ethanol at 0 °C (2 x 12.8 mL). The product is dried under vacuum at 45 °C for 10 h to afford a pale yellow solid; yield: 9.2 g (92 %).

Example 5: Preparation of crystalline seeds of (R)-3-chloro-4-(2,3-dihydroxypropoxy)- benzaldehyde

10 mg of (R)-3-chloro-4-(2,3-dihydroxypropoxy)-benzaldehyde of at least 99.5% purity by 1 H-NMR assay is dissolved in a 4 mL vial by adding 1 mL of pure ethanol (puriss p. a.). The solvent is allowed to evaporate through a small hole in the cap (approx. 2 mm of diameter) of the vial until complete dryness. The white solid residue is crystalline (R)-3-chloro-4-(2,3- dihydroxypropoxy)-benzaldehyde in crystalline form A. Alternatively, methanol or methylisobutylketone (both in puriss p. a. quality) is used. This procedure is repeated until sufficient seeds are made available.

PATENT

WO 2005054215

SEE https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2005054215

WO2005054215A1 Nov 16, 2004 Jun 16, 2005 Actelion Pharmaceuticals Ltd 5-(benz- (z) -ylidene) -thiazolidin-4-one derivatives as immunosuppressant agents
WO2008062376A2 Nov 22, 2007 May 29, 2008 Actelion Pharmaceuticals Ltd New process for the preparation of 2-imino-thiazolidin-4-one derivatives
WO2010046835A1 Oct 19, 2009 Apr 29, 2010 Actelion Pharmaceuticals Ltd Crystalline forms of (r) -5- [3-chloro-4- ( 2, 3-dihydroxy-propoxy) -benz [z] ylidene] -2- ( [z] -propylimino) -3-0-tolyl-thiazolidin-4-one
Reference
1 * BOLLI, M.H. ET AL.: “2-Imino-thiazolidin-4-one Derivatives as Potent, Orally Active S1P1 Receptor Agonists“, JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, no. 10, 2010, pages 4198-4211, XP55090073, ISSN: 0022-2623, DOI: 10.1021/jm100181s

References

  1. “Multiple-dose tolerability, pharmacokinetics, and pharmacodynamics of ponesimod, an S1P1 receptor modulator: Favorable impact of dose up-titration”. The Journal of Clinical Pharmacology 54: 179–88. Feb 2014. doi:10.1002/jcph.244. PMID 24408162.
  2.  “Mass balance, pharmacokinetics and metabolism of the selective S1P1 receptor modulator ponesimod in humans”. Xenobiotica 45: 139–49. Feb 2015. doi:10.3109/00498254.2014.955832. PMID 25188442.
  3. H. Spreitzer (29 September 2014). “Neue Wirkstoffe – Ponesimod”. Österreichische Apothekerzeitung (in German) (20/2014): 42.
  4.  “Oral ponesimod in relapsing-remitting multiple sclerosis: a randomised phase II trial”. Journal of Neurology, Neurosurgery 85: 1198–208. Nov 2014. doi:10.1136/jnnp-2013-307282. PMC 4215282. PMID 24659797.
  5.  “Oral ponesimod in patients with chronic plaque psoriasis: a randomised, double-blind, placebo-controlled phase 2 trial”. The Lancet 384: 2036–45. Dec 2014. doi:10.1016/S0140-6736(14)60803-5. PMID 25127208.
  6. “Effect of Ponesimod, a selective S1P1 Receptor Modulator, on the QT Interval in Healthy Subjects”. Basic 116: 429–37. May 2015.doi:10.1111/bcpt.12336. PMID 25287214.
  7.  “Ponesimod”. Actelion. Retrieved 31 October 2014.

ABOUT PONESIMOD

Ponesimod is a potent orally active, selective sphingosine-1-phosphate receptor 1 (S1P1) immunomodulator.

Ponesimod prevents lymphocytes from leaving lymph nodes, thereby reducing circulating blood lymphocyte counts and preventing infiltration of lymphocytes into target tissues. The lymphocyte count reduction is rapid, dose-dependent, sustained upon continued dosing, and quickly reversible upon discontinuation. Initial data suggest that ponesimod does not cause lymphotoxicity by destroying/depleting lymphocytes or interfering with their cellular function. Other blood cells e.g. cells of the innate immune system are largely unaffected. Ponesimod is therefore considered a promising new oral agent for the treatment of a variety of autoimmune disorders.

CURRENT STATUS

OPTIMUM (Oral Ponesimod versus Teriflunomide In relapsing MUltiple sclerosis) is a Phase III multi-center, randomized, double-blind, parallel-group, active-controlled superiority study to compare the efficacy and safety of ponesimod to teriflunomide in patients with relapsing multiple sclerosis (RMS). The study aims to determine whether ponesimod is more efficacious than teriflunomide in reducing relapses. The study is expected to enroll approximately 1’100 patients, randomized in 2 groups in a 1:1 ratio to receive ponesimod 20 mg/day or teriflunomide 14 mg/day, and is expected to last a little over 3 years. An additional study to further characterize the utility and differentiation of ponesimod in multiple sclerosis is being discussed with Health Authorities.

Ponesimod is also evaluated in a Phase II open-label, single-arm, intra-subject dose-escalation study to investigate the biological activity, safety, tolerability, and pharmacokinetics of ponesimod in patients suffering from moderate or severe chronic graft versus host disease (GvHD)inadequately responding to first- or second-line therapy. The study will also investigate the clinical response to ponesimod treatment in these patients. Approximately 30 patients will be enrolled to receive ponesimod in escalating doses of 5, 10, and 20 mg/day over the course of 24 weeks. The study is being conducted at approximately 10 sites in the US and is expected to last approximately 18 months.

AVAILABLE CLINICAL DATA

The decision to move into Phase III development was based on the Phase IIb dose-finding study with ponesimod in patients with relapsing-remitting multiple sclerosis. A total of 464 patients were randomized into this study and the efficacy, safety and tolerability of three ponesimod doses (10, 20, and 40 mg/day) versus placebo, administered once daily for 24 weeks.

The primary endpoint of this study was defined as the cumulative number of new gadolinium-enhancing lesions on T1-weighted magnetic resonance imaging (MRI) scans at weeks 12, 16, 20, and 24 after study drug initiation. A key secondary endpoint of this study was the annualized relapse rate over 24 weeks of treatment. Patients who completed 24 weeks of treatment were offered the opportunity to enter into an extension study. This ongoing trial is investigating the long-term safety, tolerability, and efficacy of 10 and 20 mg/day of ponesimod in patients with relapsing-remitting multiple sclerosis, in a double-blind fashion. The study continues to provide extensive safety and efficacy information for ponesimod in this indication, with some patients treated for more than 6 years.

The safety database from all studies with ponesimod now comprises more than 1,300 patients and healthy volunteers.

MILESTONES

2015 – Phase III program in multiple sclerosis initiated
2011 – Phase IIb dose-finding study in multiple sclerosis successfully completed
2006 – Entry-into-man
2004 – Preclinical development initiated

KEY SCIENTIFIC LITERATURE

Olsson T et al. J Neurol Neurosurg Psychiatr. 2014 Nov;85(11):1198-208. doi: 10.1136/jnnp-2013-307282. Epub 2014 Mar 21

Freedman M.S, et al. Multiple Sclerosis Journal, 2012; 18 (4 suppl): 420 (P923).

Fernández Ó, et al. Multiple Sclerosis Journal, 2012; 18 (4 suppl): 417 (P919).

Piali L, Froidevaux S, Hess P, et al. J Pharmacol Exp Ther 337(2):547-56, 2011

Bolli MH, Abele S, Binkert C, et al. J Med Chem. 53(10):4198-211, 2010

Kappos L et al. N Engl J Med. 362(5):387-401, 2010

Ponesimod
Ponesimod.svg
Ponesimod ball-and-stick model.png
Systematic (IUPAC) name
(2Z,5Z)-5-{3-Chloro-4-[(2R)-2,3-dihydroxypropoxy]benzylidene}-3-(2-methylphenyl)-2-(propylimino)-1,3-thiazolidin-4-one
Clinical data
Routes of
administration
Oral
Legal status
Legal status
  • Investigational
Pharmacokinetic data
Metabolism 2 main metabolites
Biological half-life 31–34 hrs[1]
Excretion Feces (57–80%, 26% unchanged), urine (10–18%)[2]
Identifiers
CAS Number 854107-55-4
ATC code none
PubChem CID 11363176
ChemSpider 9538103
ChEMBL CHEMBL1096146
Synonyms ACT-128800
Chemical data
Formula C23H25ClN2O4S
Molar mass 460.974 g/mol

////Ponesimod, Phase III , A sphingosine-1-phosphate receptor 1, S1P1 agonist, multiple sclerosis.  ACT-128800; RG-3477; R-3477, autoimmune disease, lymphocyte migration, multiple sclerosis, psoriasis, transplantation

CCC/N=C\1/N(C(=O)/C(=C/C2=CC(=C(C=C2)OC[C@@H](CO)O)Cl)/S1)C3=CC=CC=C3C

Finerenone, BAY 94-8862


Finerenone

Finerenone; UNII-DE2O63YV8R; BAY 94-8862; DE2O63YV8R; 1050477-31-0

update FDA approved, 7/9/2021, Kerendia, To reduce the risk of kidney and heart complications in chronic kidney disease associated with type 2 diabetes

C21H22N4O3
MW 378.42438 g/mol

(4s)-4-(4-cyano-2-methoxyphenyl)-5-ethoxy-2,8-dimethyl-1,4-dihydro-1-6-naphthyridine-3-carbox-amide

Bayer Corp

Bayer Healthcare Ag,

Mineralocorticoid receptor antagonist

phase III in January 2016, for treating diabetic kidney disease and chronic heart failure in patients with worsening chronic cardiac insufficiency

Used as mineralocorticoid receptor antagonist for treating heart failure and diabetic nephropathy.

SYNTHESIS

str1

Finerenone (INN, USAN) (developmental code name BAY-94-8862) is a non-steroidal antimineralocorticoid that is in phase IIIclinical trials for the treatment of chronic heart failure as of October 2015. It has less relative affinity to other steroid hormone receptors than currently available antimineralocorticoids such as eplerenone and spironolactone, which should result in fewer adverse effects like gynaecomastia, impotence, and low sex drive.[1][2]

Pharmacology

Finerenone blocks mineralocorticoid receptors, which makes it a potassium-sparing diuretic.

This table compares inhibitory (blocking) concentrations (IC50, unit: nM) of three antimineralocorticoids. Mineralocorticoid receptor inhibition is responsible for the desired action of the drugs, whereas inhibition of the other receptors potentially leads to side effects. Lower values mean stronger inhibition.[1]

Spironolactone Eplerenone Finerenone
Mineralocorticoid receptor 24 990 18
Glucocorticoid receptor 2400 22,000 >10,000
Androgen receptor 77 21,200 >10,000
Progesterone receptor 740 31,200 >10,000

The above-listed drugs have insignificant affinity for the estrogen receptor.

Chemistry

Unlike currently marketed antimineralocorticoids, finerenone is not a steroid but a dihydropyridine derivative.

Research

The drug is also being investigated in early trials for the treatment of diabetic nephropathy.[3]

 PAPER

Discovery of BAY 94-8862: A Nonsteroidal Antagonist of the Mineralocorticoid Receptor for the Treatment of Cardiorenal Diseases

  1. Dr. Lars Bärfacker1,*,
  2. Dr. Alexander Kuhl1,
  3. Prof. Dr. Alexander Hillisch1,
  4. Dr. Rolf Grosser1,
  5. Dr. Santiago Figueroa-Pérez1,
  6. Dr. Heike Heckroth1,
  7. Adam Nitsche1,
  8. Dr. Jens-Kerim Ergüden1,
  9. Dr. Heike Gielen-Haertwig1,
  10. Dr. Karl-Heinz Schlemmer2,
  11. Prof. Dr. Joachim Mittendorf1,
  12. Dr. Holger Paulsen1,
  13. Dr. Johannes Platzek3 and
  14. Dr. Peter Kolkhof4

Article first published online: 12 JUL 2012

DOI: 10.1002/cmdc.201200081

ChemMedChem

ChemMedChem

Volume 7, Issue 8, pages 1385–1403, August 2012

Abstract

Aldosterone is a hormone that exerts manifold deleterious effects on the kidneys, blood vessels, and heart which can lead to pathophysiological consequences. Inhibition of the mineralocorticoid receptor (MR) is a proven therapeutic concept for the management of associated diseases. Use of the currently marketed MR antagonists spironolactone and eplerenone is restricted, however, due to a lack of selectivity in spironolactone and the lower potency and efficacy of eplerenone. Several pharmaceutical companies have implemented programs to identify drugs that overcome the known liabilities of steroidal MR antagonists. Herein we disclose an extended SAR exploration starting from cyano-1,4-dihydropyridines that were identified by high-throughput screening. Our efforts led to the identification of a dihydronaphthyridine, BAY 94-8862, which is a potent, selective, and orally available nonsteroidal MR antagonist currently under investigation in a clinical phase II trial.

str1

str1

PATENT

WO2008104306,

http://www.google.co.in/patents/WO2008104306A2?cl=en

Bayer Healthcare Ag,

Lars Baerfacker, BELOW

Peter Kolkhof, BELOW

Karl-Heinz Schlemmer, Rolf Grosser, Adam Nitsche,Martina Klein, Klaus Muenter, Barbara Albrecht-Kuepper, Elke Hartmann,

EXAMPLES

Example 1

4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2-methyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxamide

Figure imgf000066_0001

100 mg (ca. 0:24 mmol) of the compound from Example 23A are initially charged in 3 ml DMF. Is 2.94 mg Then (0.024 mmol) of 4-N, N-dimethylaminopyridine and 340 ul of ammonia (28 wt .-% – solution in water, 2:41 mmol) and 3 h at 100 0 C temperature. After cooling, the crude product is purified directly by preparative HPLC (eluent: acetonitrile / water with 0.1% formic acid, gradient 20:80 → 95: 5). There are 32 mg (37% d. Th.) The title connection receive.

LC-MS (Method 3): R, = 1:57 min; MS (EIPOS): m / z = 365 [M + H] +

1 H-NMR (300 MHz, DMSOd6): δ = 1:07 (t, 3H), 2.13 (s, 3H), 3.83 (s, 3H), 4:04 (m, 2H), 5:36 (s, IH), 6:42 (d, IH), 6.66 (br. s, 2H), 7.18 (d, IH), 7.29 (dd, IH), 7:38 (d, IH), 7.67 (d, IH), 8.80 (s, IH).

Example 2

4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,7-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxamide

Figure imgf000067_0001

640 mg (1.69 mmol) of the compound from Example 27A are initially charged in 30 ml of ethyl acetate, 342 mg (2.11 mmol) l, r-carbonyldiimidazole and then stirred overnight at room temperature. A TLC check (silica gel; mobile phase: cyclohexane / ethyl acetate 1: 1 or dichloromethane / methanol 9: 1) shows complete conversion. The volatile components are removed on a rotary evaporator and the residue taken up in 20 ml DMF. Subsequently, 2.36 ml of ammonia (28 wt .-% – solution in water, 16.87 mmol) was added and the reaction mixture for 8 hours at 50 0 C temperature. The solvent is distilled off under reduced pressure and the residue purified by preparative HPLC (eluent: acetonitrile / water with 0.1% formic acid, gradient 20:80 -> 95: 5). This gives 368 mg (58% d. Th.) Of the title compound.

LC-MS (method 7): R t = 1.91 min; MS (EIPOS): m / z = 379 [M + H] +

1 H-NMR (300 MHz, DMSO-d 6): δ = 1:05 (t, 3H), 2.13 (s, 3H), 2.19 (s, 3H), 3.84 (s, 3H), 4:02 (q, 2H) , 5:32 (s, IH), 6.25 (s, IH), 6.62 (br. s, 2H), 7.16 (d, IH), 7.28 (dd, IH), 7:37 (d, IH), 8.71 (s, IH ).

Example 3

e ‘f 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,7-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox- amide [(-) – enantiomer and (+) – enantiomer \

Figure imgf000068_0001

The racemate of Example 2 can be separated on a preparative scale by chiral HPLC into its enantiomers [column: Chiralpak IA, 250 mm x 20 mm; Eluent: methyl tert-butyl ether / methanol 85: 15 (v / v); Flow: 15 ml / min; Temperature: 30 0 C; UV detection: 220 Dm].

(-) – Enantiomer:

HPLC: R, = 5.28 min, ee> 98% [column: Chiralpak IA, 250 mm x 4.6 mm; Eluent: methyl tert-butyl ether / methanol 80:20 (v / v); Flow: 1 ml / min; Temperature: 25 0 C; UV detection: 220 nm];

specific optical rotation (chloroform, nm 589, 19.8 ° C, c = 0.50500 g / 100 ml): -239.3 °.

A single crystal X-ray structural analysis revealed a ^ -configuration at C * for this enantiomer – atom.

(+) – Enantiomer:

HPLC: R = 4:50 min, ee> 99% [column: Chiralpak IA, 250 mm x 4.6 mm; Eluent: methyl tert-butyl ether / methanol 80:20 (v / v); Flow: 1 ml / min; Temperature: 25 ° C; UV detection: 220 nm];

specific optical rotation (chloroform, nm 589, 20 0 C, c = 0.51000 g / 100 ml): + 222.7 °.

Example 4

4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxamide

Figure imgf000069_0001

1:46 g (3.84 mmol) of the compound from Example 3oA are introduced into 50 ml of ethyl acetate, 777 mg (4.79 mmol) l, r-carbonyldiimidazole and then stirred overnight at room temperature. A TLC check (silica gel; eluent: ethyl acetate) shows complete conversion. The volatile components are removed on a rotary evaporator and the residue taken up in 20 ml DMF.Then 10.74 ml of ammonia (28 wt% solution in water, 76.8 mmol) was added and the reaction mixture heated for 30 minutes at 100 0 C. The solvent is distilled off under reduced pressure and the residue purified by preparative HPLC (eluent: acetonitrile / water with 0.1% formic acid, gradient 20:80 -> 95: 5). After concentrating the product fractions, the residue in 40 ml of dichloromethane / methanol (1: 1 v / v) and treated with 100 ml of ethyl acetate. The solvent is concentrated to a volume of about 20 ml, whereupon the product crystallized. The precipitate is filtered off and washed with a little diethyl ether.After drying at 40 0 C in a vacuum oven obtained 1:40 g (96%. Th.) The title connection.

LC-MS (Method 3): R, = 1.64 min; MS (EIPOS): m / z = 379 [M + H] +

1 H-NMR (300 MHz, DMSOd6): δ = 1:05 (t, 3H), 2.12 (s, 3H), 2.18 (s, 3H), 3.82 (s, 3H), 3.99-4.07 (m, 2H) , 5:37 (s, IH), 6.60-6.84 (m, 2H), 7.14 (d, IH), 7.28 (dd, IH), 7:37 (d, IH), 7:55 (s, IH), 7.69 (s, IH ).

Example 5

e “M- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox- amide [(-) – enantiomer and (+ ) enantiomer]

Figure imgf000070_0001

The racemate of Example 4 can be separated on a preparative scale by chiral HPLC into its enantiomers [column: 680 mm x 40 mm; Silica gel phase based on the chiral selector poly (N-methacryloyl-D-leucine dicyclopropylmethylamide; eluent: ethyl acetate; temperature: 24 ° C; flow: 80 ml / min; UV detection: 260 nm].

(-) – Enantiomer:

HPLC: R = 2:48 min, ee = 99.6% [column: 250 mm x 4.6 mm; Silica gel phase based on the chiral selector poly (N-methacryloyl-D-leucine dicyclopropylmethylamide; eluent: ethyl acetate; temperature: 24 ° C; flow: 2 ml / min; UV detection: 260 nm];

specific optical rotation (chloroform, nm 589, 19.7 ° C, c = 0.38600 g / 100 ml): -148.8 °.

A single crystal X-ray structure analysis showed this enantiomer S configuration at C * – atom.

(+) – Enantiomer:

HPLC: R = 4:04 min, ee = 99.3% [column: 250 mm x 4.6 mm; Silica gel phase based on the chiral selector poly (N-methacryloyl-D-leucine dicyclopropylmethylamide; eluent: ethyl acetate; temperature: 24 ° C; flow: 2 ml / min; UV detection: 260 nm];

specific optical rotation (chloroform, nm 589, 19.8 ° C, c = 0.36300 g / 100 ml): + 153.0 °.

PATENT

WO 2016016287

The present invention relates to a novel and improved process for preparing 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-1, 4-dihydro- 1, 6-naphthyridine-3-carbox- amide of formula (I)

as well as the preparation and use of crystalline modification I of (4S) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-1, 4-dihydro- 1, 6-naphthyridine-3- carbox-amide of formula (I).

The compound of formula (I) acts as a non-steroidal mineralocorticoid receptor antagonist and can be used as agents for the prophylaxis and / or treatment of cardiovascular and renal diseases such as heart failure and diabetic nephropathy.

The compound of formula (I) and their preparation process are described in WO 2008/104306 and ChemMedChem 2012 7, described in 1385, in both publications a detailed discussion of research synthesis is disclosed. A disadvantage of the synthesis described there is the fact that this synthesis is not suitable for another large-scale process, since many steps in very high dilution, at very high reagent surpluses and thus run on a relatively low overall yield. Furthermore, many chromatographic cleanings are necessary, which are usually very expensive and require a high consumption of solvents, are costly and which should therefore be avoided if possible.Some stages can not be realized due to safety and procedural difficulties.

There is therefore a need for an industrially viable synthesis, reproducible in high overall yield, low production costs and high purity provides the compound of formula (I) and complies with all regulatory requirements in order to supply the clinical trials on drug and for subsequent regulatory submission to be used.

With the present invention a very efficient synthesis has been found, which allows to meet the above requirements.

In the publication ChemMedChem 2012 7, in which the research synthesis of the compound of formula (I) disclosed in 1385, the compound of formula (I), starting from vanillin prepared in 10 steps with an overall yield of 3.76% of theory , The compound of formula (I) was obtained by evaporation of the chromatography fractions as an amorphous solid, a defined process Kristalhsations- the stage for polymorphism-setting has not been described.

The following Scheme 1 shows the known process for preparing the compound of formula (I).

(II) (HI) (IV)

(V) (VI)

(XIII) (I)

Scheme 1: synthesis research of the compound of formula (I)

There are used 3 chromatographic purifications, and a chiral chromatography step to separate the enantiomers of the racemate of formula (XIII). The steps run partially in very high dilution and using very large amounts of reagent.

Thus, in particular the sequence of the preparation of the nitrile aldehyde intermediate (VI), which occupies a central role in the synthesis of atom not economically acceptable.

Furthermore, not to apply this process to an industrial scale, since [=> (IV) (III)] and excesses of acrylic acid tert-butyl ester are used for a very expensive reagents such as trifluoromethanesulfonic anhydride. When upscaling the Heck reaction (IV) => (V) formed in the boiler, a plastic similar residue resulting from the polymerization of acrylic acid tert.butyl ester used in excess. This is not acceptable in the technical implementation, there is a risk that there may be a Rührerbruch and it would lead to strong to remove residues in the agitators.

The subsequent cleavage of the double bond with sodium and the highly toxic osmium tetroxide is to be avoided since there is a delay of reaction and thereby caused to a strongly exothermic and connected with that comes a runaway under the test conditions described.

Scheme 2 illustrates the new process of the invention that the compound of formula (I) in 9 levels in 27.7% d. Th. Total yield without a chromatographic

Purification of intermediates supplies.

Scheme 2: According to the Invention for preparing the compound of formula (I).

Examples

example 1

Methyl 4-bromo-2-methoxybenzoate (XV)

3.06 kg (22.12 mol) potassium carbonate are placed in 1 acetone 3.6 and heated to reflux. To this suspension is metered in 1.2 kg of 4-bromo-2-hydroxybenzoic acid (5.53 mol) suspended in 7.8 1 of acetone and rinsed with 0.6 1 acetone. The mixture is heated for one hour under reflux (vigorous evolution of gas!). is boiled for 2.65 kg (21.01 mol) Dimethylsufat over 4 hours then metered. 2.5 hours then is stirred under reflux. The solvent is distilled off to a large extent (up to the stirrability) and returns to 12 1 toluene, then the remaining acetone is distilled off at 110 ° C. There are about 3 1 distillate distilled, these are supplemented by the addition of a further 3 1 toluene to approach. Allow to cool to 20 ° C and are 10.8 1 water were added and agitated vigorously. The organic phase is separated and the aqueous phase extracted again with 6.1 1 of toluene. The combined organic phases are washed with 3 1 of saturated sodium chloride solution, and the toluene phase is concentrated to about 4 first A quantitative analysis by evaporating a subset results converted a yield 1.306 kg (96.4% of theory). The solution is used directly in the next stage.

HPLC method A: RT about 11.9 min.

MS (EIPOS): m / z = 245 [M + H] +

H NMR (400 MHz, CD 2 C1 2 ): δ = 3.84 (s, 3H), 3.90 (s, 3H), 7:12 to 7:20 (m, 2H), 7.62 (d, 1H).

example 2

4-bromo-2-methoxybenzaldehyde (XVI)

It puts 1.936 kg (6.22 mol) 65% Red- Al solution in toluene with 1.25 1 of toluene at -5 ° C before. To this solution was dosed 0.66 kg (6.59 mol) of 1-methylpiperazine and rinsed with 150 ml of toluene, the temperature keeps you here from -7 to -5 ° C.. It is allowed for 30 minutes at 0 ° C. for. This solution is then dosed to a solution of 1.261 kg (5.147 mol) of methyl 4-bromo-2-methoxybenzoate (XV), dissolved in 4 1 of toluene, the temperature is maintained at – 8-0 ° C. Rinse twice with 0.7 1 of toluene and stirred for 1.5 hours at 0 ° C to. For working up, dosed to a 0 ° C cold aqueous sulfuric acid (12.5 1 water + 1.4 kg of conc. Sulfuric acid). The temperature should rise to a maximum of 10 ° C (slow dosage). The pH is, if necessary, by addition of further sulfuric acid to a pH of the first The organic phase is separated and extracted the aqueous phase with 7.6 1 of toluene. The combined organic phases are washed with 5.1 1 of water and then substantially concentrated and the residue taken up with 10 1 DMF. The mixture is concentrated again to about 5 1 volume. A quantitative analysis by evaporating a subset results converted a yield 1.041 kg (94.1% of theory). The solution is used directly in the next stage.

HPLC method A: RT approximately 12.1 min.

MS (EIPOS): m / z = 162 [M + H] +

X H-NMR (CDCl, 400MHz): δ = 3.93 (3H, s), 7.17 (2H, m), 7.68 (1H, d), 10:40 (1H, s)

example 3

4-formyl-3-methoxybenzonitrile (VI)

719 g (3.34 mol) of 4-bromo-2-methoxybenzaldehyde (XVI) as a solution in 4.5 1 of DMF with 313 g (0.74 mol) of potassium hexacyanoferrate (K4 [Fe (CN) 6]) and 354 g submitted (3.34 mol) of sodium carbonate and a further 1.2 1 of DMF and 3.8 g (0.017 mol) of palladium acetate. It is stirred for 3 hours at 120 ° C. Allow to cool to 20 ° C and are 5.7 1 water to approach. It is extracted with 17 1 ethyl acetate, and the aqueous phase is washed again with 17 1 of ethyl acetate to. The organic phases are combined and substantially concentrated with 5 1 of isopropanol was added and concentrated to about 2 1st The mixture is heated to boiling and dripping 2 1 of water.Allow to cool to 50 ° C and are again added 2 1 water. It is cooled to 3 ° C and stirred for one hour at this temperature. The product is filtered and washed with water (2 times 1.2 1) washed. It is dried at 40 ° C under vacuum.

Yield: 469 g (87% of theory.) Of a beige solid.

HPLC method A: RT about 8.3 min.

MS (EIPOS): m / z = 162 [M + H] +

1H-NMR (300 MHz, DMSO-d6): δ = 3.98 (s, 3H), 7:53 (d, 1H), 7.80 (s, 1H), 7.81 (d, 1H), 10:37 (s, 1H).

example 4

2-cyanoethyl 4- (4-cyano-2-methoxyphenyl) -2,8-dimethyl-5-oxo-l, 4,5,6-tett ^

din-3-carboxylate (X)

option A

1.035 kg (6.422 mol) of 4-formyl-3-methoxybenzonitrile (VI), 1.246 kg (8.028 mol) of 2-Cyanefhyl 3-oxobutanoate, 54.6 g (0.642 mol) of piperidine and 38.5 g (0.642 mol) of glacial acetic acid are heated under reflux on a water in 10 1 dichloromethane 6.5 hours. Allow to cool to room temperature and the organic phase was washed 2 times with 5 1 water. Subsequently, the dichloromethane phase is concentrated under atmospheric pressure and the still stirrable residue with 15.47 kg of 2-butanol and 0.717 kg (5.78 mol) of 4-amino-5-methylpyridone added. The residual dichloromethane is distilled off until an internal temperature of 98 ° C is reached. Then, 20 hours, heated under reflux. It is cooled to 0 ° C, can be 4 hours at this temperature is stirred and filtered off the product. It is dried at 40 ° C under vacuum to the carrier gas.

Yield: 2.049 kg (87.6% of theory based on 4-amino-5-methylpyridone, since this component is used in deficiency) of a slightly yellowish colored solid.

HPLC method A: RT about 9.7 min.

MS (EIPOS): m / z = 405 [M + H] +

Ή-NMR (300 MHz, DMSO-d 6 ): δ = 2:03 (s, 3H), 2:35 (s, 3H), 2.80 (m, 2H), 3.74 (s, 3H), 4:04 (m, 1H), 4.11 (m, 1H), 5.20 (s, 1H), 6.95 (s, 1H), 7.23 (dd, 1H), 7:28 to 7:33 (m, 2H), 8.18 (s, 1H), 10.76 (s, 1H) ,

variant B

1.344 kg (8.34 mol) of 4-formyl-3-methoxy-benzonitrile (VI), 71 g (0.834 mol) piperidine and 50.1 g (0.834 mol) of glacial acetic acid are introduced into 6 1 of isopropanol at 30 ° C within 3 hours, a solution of 1.747 kg (11.26 mol) of 2-cyanoethyl 3-oxobutanoate metered in 670 ml of isopropanol. Stirring an hour after at 30 ° C. It is cooled to 0-3 ° C and stirred at 0.5 hours. the product is filtered off and washed 2 times with 450 ml of cold isopropanol to. For yield determination is under vacuum at 50 ° C. (2.413 kg, 97% of theory..); but it is usually due to the high yield continued to work directly with the isopropanol-moist product. For this, the product is taken up with 29 1 of isopropanol and 1.277 kg (7.92

mol) of 4-amino-5-methylpyridone added, followed by 24 internal temperature under about 1.4 bar overpressure in the closed vessel is heated at 100 ° C h. It is cooled by a ramp within 5 h at 0 ° C. stirred for 3 hours at 0 ° C. It is filtered off and washed with 2.1 1 of cold isopropanol. It is dried under vacuum at 60 ° C.

Yield: 2.819 kg (88% of theory based on 4-amino-5-methylpyridone, since this component is used in deficiency) of a slightly yellowish colored solid.

HPLC method A: RT about 9.7 min.

MS (EIPOS): m / z = 405 [M + H] +

Ή-NMR (300 MHz, DMSO-d 6 ): δ = 2:03 (s, 3H), 2:35 (s, 3H), 2.80 (m, 2H), 3.74 (s, 3H), 4:04 (m, 1H), 4.11 (m, 1H), 5.20 (s, 1H), 6.95 (s, 1H), 7.23 (dd, 1H), 7:28 to 7:33 (m, 2H), 8.18 (s, 1H), 10.76 (s, 1H) ,

example 5

2- cyanoethyl-4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylate (XI)

2.142 kg (5.3 mol) of 2-cyanoethyl 4- (4-cyano-2-methoxyphenyl) -2,8-dimefhyl-5-oxo-l, 4,5,6-tetrahydro-l, 6-naphthyridin-3 carboxylate (X) and 4.70 kg (29 mol) of triethyl orthoacetate are dissolved in 12.15 1 of dimethylacetamide and 157.5 grams of concentrated sulfuric acid was added. The mixture is heated for 1.5 hours at 115 ° C and then cooled to 50 ° C. At 50 ° C are added dropwise to 30 minutes 12.15 1 water. After complete addition the Titelbelbindung (XI) is treated with 10 g seeded and further added dropwise to 12.15 1 of water over 30 minutes at 50 ° C. It is cooled to 0 ° C (ramp, 2 hours) and stirred for 2 hours at 0 ° C to. The product is filtered, washed 2 times each with 7.7 1 of water and dried in vacuo at 50 ° C.

Yield: 2114.2 g (92.2% of theory) of a slightly yellowish colored solid.

HPLC Method B: RT 10,2 min.

MS (EIPOS): m / z = 433 [M + H] +

X H-NMR (300 MHz, DMSO-d 6 ): δ = 1.11 (t, 3H), 2.16 (s, 3H), 2:42 (s, 3H), 2.78 (m, 2H), 3.77 (s, 3H) , 4:01 to 4:13 (m, 4H), 5:37 (s, 1H), 7.25 (d, 1H), 7:28 to 7:33 (m, 2H), 7.60 (s, 1H), 8:35 (s, 1H).

Alternatively, the reaction in NMP (l-methyl-2-pyrrolidone) may be carried out

2- cyanoethyl-4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylate (XI)

2.142 kg (5.3 mol) of 2-cyanoethyl 4- (4-cyano-2-methoxyphenyl) -2,8-dimethyl-5-oxo-l, 4,5,6-tetrahydro-l, 6-naphthyridin-3 carboxylate (X) and 2.35 kg (14.5 mol) of triethyl orthoacetate are in 3.21 kg NMP (l-methyl-2-pyrrolidone) and dissolved 157.5 g of concentrated sulfuric acid was added. The mixture is heated for 1.5 hours at 115 ° C and then cooled to 50 ° C. At 50 ° C are added dropwise to 30 minutes 2.2 1 water. After complete addition the Titelbelbindung (XI) is treated with 10 g seeded and dropped further 4.4 1 of water over 30 minutes at 50 ° C. It is cooled to 0 ° C (ramp, 2 hours) and stirred for 2 hours at 0 ° C to. The product is filtered off, washed 2 times each with 4 1 of water and dried under vacuum at 50 ° C.

Yield: 2180.7 g (95.1% of theory) of a slightly yellowish colored solid.

HPLC Method B: RT 10,2 min.

example 6

4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-1, 4-dihydro- 1, 6-naphthyridine-3-carboxylic acid IXM

2.00 kg (4.624 mol) of 2-cyanoethyl 4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylate (XI ) are dissolved in a mixture of 12 1 THF and 6 1 of water and cooled to 0 ° C. To this solution, a sodium hydroxide solution is added in drops within 15 minutes at 0 ° C (prepared from 0.82 kg 45% aqueous. NaOH (9.248 mol) and 4.23 1 of water and stirred for 1.5 hours at 0 ° C to . The mixture is extracted 2 times with each 4.8 1 methyl tert-butyl and once with 4.8 1 of ethyl acetate. The aqueous solution is at 0 ° C with dilute hydrochloric acid (prepared from 0.371 kg 37% HCl and 1.51 1 water ) adjusted to pH 7. the mixture is allowed to warm to 20 ° C and adding an aqueous solution of 2.05 kg of ammonium chloride in 5.54 1 water. the mixture is stirred 1 hour at 20 ° C, the product filtered and 2 times with each each 1.5 1 water and washed once with 4 1 acetonitrile. It is dried at 40 ° C under vacuum to the carrier gas.

Yield: 1736.9 g (99% of theory..) Of an almost colorless powder (very slight yellow tinge).

HPLC Method C: RT: about 6.8 min.

MS (EIPOS): m / z = 380 [M + H]

X H-NMR (300 MHz, DMSO-d 6 ): δ = 1.14 (t, 3H), 2.14 (s, 3H), 2:37 (s, 3H), 3.73 (s, 3H), 4:04 (m, 2H) , 5:33 (s, 1H), 7.26 (m, 2H), 7:32 (s, 1H), 7:57 (s, 1H), 8.16 (s, 1H), 11:43 (br. s, 1H).

Alternative workup with toluene for extraction:

4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylic-isäure (XII)

2.00 kg (4.624 mol) of 2-cyanoethyl 4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylate (XI ) are dissolved in a mixture of 12 1 THF and 6 1 of water and cooled to 0 ° C. To this solution, a sodium hydroxide solution is added in drops within 15 minutes at 0 ° C (prepared from 0.82 kg 45% aqueous. NaOH (9.248 mol) and 4.23 1 of water and stirred for 1.5 hours at 0 ° C to . Add 5 L of toluene and 381.3 g Natiumacetat added and stirred vigorously. Allow to settle the phases and the organic phase is separated. the aqueous phase is adjusted with 10% hydrochloric acid to pH 6.9 (at about pH 9.5 is inoculated with 10 g of the title compound of). After completion of the precipitation of the product for one hour at 0 ° C is stirred and then filtered and washed twice with 4 1 of water and twice with 153 ml of toluene. the mixture is dried at 40 ° C under vacuum to carrier gas (nitrogen, 200 mbar. yield:.. 1719.5 g (98% of theory) of an almost colorless powder (very slight yellow tinge).

HPLC Method C: RT: about 6.8 min).

example 7

4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-1, 4-dihydro- 1, 6-naphthyridine-3-carboxamide

1.60 kg (4.22 mol) of 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carboxylic-isäure ( XII) and 958 g (5.91 mol) of 1,1-carbodiimidazole be presented in 8 1 of THF and at 20 ° C 51 g (0.417 mol) of DMAP was added. Stirring for one hour at 20 ° C (gas evolution!) And then heated 2.5 hours 50 ° C. are added to this solution 2.973 kg (18.42 mol) of hexamethyldisilazane and boil for 22 hours under reflux. Man admits further 1.8 1 THF and cooled to 5 ° C. A mixture is prepared from 1.17 1 of THF and 835 g of water is metered in over 3 hours, so that the temperature is between 5 and 20 ° C remains. Then boiled for one hour under reflux, then cooled via a ramp (3 hours) at 0 ° C. and stirred for one hour at this temperature. The product is filtered off and washed 2 times with 2.4 1 THF and twice with 3.2 1 water. It is dried under vacuum at 70 ° C under a carrier gas.

Yield: 1.501 kg (. 94% of theory) of an almost colorless powder (very slight yellow tinge).

HPLC Method B: RT about 6.7 min.

MS (EIPOS): m / z = 379 [M + H]

Ή-NMR (300 MHz, DMSO-d 6 ): δ = 1:05 (t, 3H), 2.12 (s, 3H), 2.18 (s, 3H), 3.82 (s, 3H), 3.99-4.07 (m, 2H ), 5:37 (s, 1H), 6.60-6.84 (m, 2H), 7.14 (d, 1H), 7.28 (dd, 1H), 7:37 (d, 1H), 7:55 (s, 1H), 7.69 (s, 1H).

example 8

(4S) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy

carbox-amide (I) as a solution in acetonitrile / Methariol 40:60

Enantiomeric separation on a SMB unit

As a feed solution a solution corresponding to a concentration is used consisting of 50 g racemic 4- (4-cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridin-3 -carbox-amide (XIII) dissolved in 1 liter of a mixture of methanol / acetonitrile 60:40.

There is a SMB unit on a stationary phase: 20 chromatographed μιη Chiralpak AS-V. The pressure is 30 bar, as the eluent a mixture of methanol / acetonitrile 60:40 is used.

9.00 kg of 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox-amide (XII) are dissolved in 180 1 a mixture dissolved consisting of methanol / acetonitrile 60:40 and chromatographed by SMB. After concentrating the product-containing fractions, 69.68 liters of a 6.2% solution (corresponding to 4.32 kg (4S) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl- 1, 4-dihydro- 1, 6-naphthyridine-3-carbox-amide (I) as a solution in acetonitrile / methanol 40:60).

Yield: 4.32 kg (48% of theory.) Dissolved in 69.68 liters of acetonitrile / methanol 40:60 as a colorless fraction.

Enantiomeric purity:> 98.5% ee (HPLC, method D)

A sample is concentrated in vacuum to give: MS (EIPOS): m / z = 379 [M + H] +

Ή-NMR (300 MHz, DMSO-d 6 ): δ = 1:05 (t, 3H), 2.12 (s, 3H), 2.18 (s, 3H), 3.82 (s, 3H), 3.99-4.07 (m, 2H ), 5:37 (s, 1H), 6.60-6.84 (m, 2H), 7.14 (d, 1H), 7.28 (dd, 1H), 7:37 (d, 1H), 7:55 (s, 1H), 7.69 (s, 1H).

example 9

(4S) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox-amide (I)

Crystallization and Polymorphism setting

64.52 liters of a 6.2% solution of Example 8 in a mixture Acetonitiril / methanol 40:60 (equal 4.00 kg of compound 1) (1.2 .mu.m) via a filter cartridge and then concentrated at 250 mbar applicable so that the solution is still stirrable. It added 48 1 of ethanol denatured with toluene and distilled again at 250 mbar to stirrability from (Umdestillation on ethanol). They gave an additional 48 1 of ethanol denatured with toluene and then distilled at atmospheric pressure to a total volume of about 14 1 from (jacket temperature 98 ° C). The mixture was cooled via a ramp (4 hours) to 0 ° C, stirred for 2 hours at 0 ° C and filtered by the product from. It was washed twice with 4 1 of cold ethanol and then dried in vacuo at 50 ° C.

Yield: 3.64 kg (91% of theory.) Of a colorless, crystalline powder

Enantiomeric purity: “99% ee (HPLC method D); Retention times / RRT: (4S) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox-amide (1) ca. 11 min. RRT: 1.00; (4R) – 4- (4-Cyano-2-methoxyphenyl) -5-ethoxy-2,8-dimethyl-l, 4-dihydro-l, 6-naphthyridine-3-carbox-amide (I) is about 9 min ,RRT: 0.82

Purity:> 99.8% (HPLC method B) RT: about 6.7 min.

Content: 99.9% (against an external standard)

specific rotation (chloroform, 589 nm, 19.7 ° C, c = 0.38600 g / 100 ml): – 148.8 °.

MS (EIPOS): m / z = 379 [M + H] +

Ή-NMR (300 MHz, DMSO-d 6 ): δ = 1:05 (t, 3H), 2.12 (s, 3H), 2.18 (s, 3H), 3.82 (s, 3H), 3.99-4.07 (m, 2H ), 5:37 (s, 1H), 6.60-6.84 (m, 2H), 7.14 (d, 1H), 7.28 (dd, 1H), 7:37 (d, 1H), 7:55 (s, 1H), 7.69 (s, 1H).

Melting point: 252 ° C (compound of formula (I) in crystalline form of modification I)

Physico-chemical characterization of compound of formula (I) in crystalline form of modification I

Compound of formula (I) melts in crystalline form of modification I at 252 ° C, ΔΗ = 95 -113 Jg 1 (heating rate 20 K min 1 , Figure 1).

A depression of the melting point was observed as a function of the heating rate.

The melting point decreases at a lower heating rate (eg 2 K min “1 ) because decomposition occurs. There were no other phase transitions. A mass loss of about 0.1% was observed up to a temperature of 175 ° C.

References

  1.  Schubert-Zsilavecz, M, Wurglics, M, Neue Arzneimittel Herbst 2015 (German)
  2.  Pitt, B; Anker, S. D.; Böhm, M; Gheorghiade, M; Køber, L; Krum, H; Maggioni, A. P.; Ponikowski, P; Voors, A. A.; Zannad, F; Nowack, C; Kim, S. Y.; Pieper, A; Kimmeskamp-Kirschbaum, N; Filippatos, G (2015). “Rationale and design of MinerAlocorticoid Receptor antagonist Tolerability Study-Heart Failure (ARTS-HF): A randomized study of finerenone vs. Eplerenone in patients who have worsening chronic heart failure with diabetes and/or chronic kidney disease”. European Journal of Heart Failure 17 (2): 224–32.doi:10.1002/ejhf.218. PMID 25678098.
  3.  Bakris, G. L.; Agarwal, R; Chan, J. C.; Cooper, M. E.; Gansevoort, R. T.; Haller, H; Remuzzi, G; Rossing, P; Schmieder, R. E.; Nowack, C; Kolkhof, P; Joseph, A; Pieper, A; Kimmeskamp-Kirschbaum, N; Ruilope, L. M.; Mineralocorticoid Receptor Antagonist Tolerability Study–Diabetic Nephropathy (ARTS-DN) Study Group (2015). “Effect of Finerenone on Albuminuria in Patients with Diabetic Nephropathy: A Randomized Clinical Trial”. JAMA 314 (9): 884–94. doi:10.1001/jama.2015.10081. PMID 26325557.
Finerenone.svg
Systematic (IUPAC) name
(4S)-4-(4-Cyano-2-methoxyphenyl)-5-ethoxy-2,8-dimethyl-1,4-dihydro-1,6-naphthyridine-3-carboxamide
Clinical data
Legal status
  • Investigational
Routes of
administration
Oral
Identifiers
CAS Number 1050477-31-0
ATC code None
PubChem CID 60150535
ChemSpider 28669387
UNII DE2O63YV8R
KEGG D10633
ChEMBL CHEMBL2181927
Synonyms BAY 94-8862
Chemical data
Formula C21H22N4O3
Molar mass 378.42 g/mol

SEE………http://apisynthesisint.blogspot.in/2016/02/finerenone-bay-94-8862.html

////Finerenone , BAYER, PHASE 3, BAY 94-8862

CCOC1=NC=C(C2=C1C(C(=C(N2)C)C(=O)N)C3=C(C=C(C=C3)C#N)OC)C

Fexinidazole Hoe-239


Fexinidazole.svg

Fexinidazole, Hoe-239

1-Methyl-2-{[4-(methylsulfanyl)phenoxy]methyl}-5-nitro-1H-imidazole

cas59729-37-2 
Molecular formula C12H13N3O3S
Molar mass 279.31 g mol−1

Hoechst Aktiengesellschaft

Sanofi (Originator)
University of Dundee
Drugs for Neglected Diseases Initiative

Winkelmann, E.; Raether, W.
Chemotherapeutically active nitro compounds. 4,5-nitroimidazoles. Part III
Arzneim-Forsch Drug Res 1978, 28(5): 739

US 4042705, DE 2531303,

UPDATE 7/16/2021 FDA APPROVES

To treat human African trypanosomiasis caused by the parasite Trypanosoma brucei gambiense

600 MG TABLET ORAL, DRUGS FOR NEGLECTED DISEASES INITIATIVE

US FDA approves fexinidazole as the first all-oral treatment for sleeping sickness

POSTED ON JULY 19

The US Food and Drug Administration (FDA) has approved fexinidazole as the first all-oral treatment for both stages of the Trypanosoma brucei gambiense form of sleeping sickness (Human African trypanosomiasis) in patients 6 years of age and older and weighing at least 20 kg.
Fexinidazole was developed as part of an innovative partnership between the non-profit research and development organization Drugs for Neglected Diseases initiative (DNDi), which conducted the pivotal clinical trials for this treatment, in partnership with the National Sleeping Sickness Programs of the Democratic Republic of Congo (DRC) and Central African Republic (CAR), and Sanofi.

Sleeping sickness is a parasitic disease transmitted by the bite of an infected tse-tse fly. It affects mostly populations living in remote rural areas of sub-Saharan Africa, where about 65 million people are at risk of infection. Left untreated, sleeping sickness is almost always fatal. Through Sanofi’s collaboration the number of sleeping sickness cases reported to the WHO has been reduced by ~97% between 2001 and 2020. DNDiSanofi and partners are deeply committed to ensuring access to fexinidazole in all sleeping sickness-endemic countries.

Current treatment options for the disease are effective, but burdensome for patients and health workers due to the need for infusion or injection, requiring hospitalization, especially challenging for people living in remote areas.

“Having a simple, all-oral treatment for sleeping sickness is a dream come true for frontline clinicians,” said Dr Bernard Pécoul, DNDi Executive Director. “We are proud of this latest milestone in our long-term partnership with Sanofi, developed in close collaboration with researchers in countries hard-hit by sleeping sickness.”

Fexinidazole is indicated as a 10-day once-a-day treatment for Trypanosoma brucei gambiense sleeping sickness, the most common form of the disease found in West and Central Africa. Fexinidazole is the first all-oral treatment that works both for the first stage of the disease, as well as the second stage of the disease in which the parasites have crossed the blood-brain barrier, causing patients to suffer from neuropsychiatric symptoms.

This FDA approval is a key milestone in Sanofi’s long-term commitment to fight sleeping sickness, started 20 years ago alongside the WHO through an ambitious partnership to combat Neglected Tropical Diseases” said Luc Kuykens, Senior Vice President, Sanofi Global Health unit. “Following the positive scientific opinion granted by the European Medicines Agency end 2018, the FDA approval is an important step to revitalize efforts to support the sustainable elimination of the disease”.

As a result of FDA approval, a Tropical Disease Priority Review Voucher (PRV) has been awarded to DNDi. The FDA Tropical Disease PRV Program was established in 2007 to incentivize development of new treatments for neglected tropical diseases, including sleeping sickness. Any benefits from the PRV will be shared between Sanofi and DNDi, which will enable continued investments in innovating for and ensuring access to new health tools for sleeping sickness and other neglected diseases. Sanofi commits to continue to provide the drug free-of-charge to the World Health Organization for distribution to affected countries, as part of a long-term collaboration with WHO.

About Sleeping sickness
Sleeping sickness, or human African trypanosomiasis (HAT), is usually fatal without treatment. Transmitted by the bite of an infected tse-tse fly, following a period with nonspecific symptoms, it evolves to cause neuropsychiatric symptoms, including abnormal behaviour, and a debilitating disruption of sleep patterns that have given this neglected disease its name. About 65 million people in sub-Saharan Africa are at moderate to very high risk of infection.

About DNDi
The Drugs for Neglected Diseases initiative (DNDi) is a collaborative, patient needs-driven, not-for-profit research and development (R&D) organization that develops safe, effective, and affordable treatments for sleeping sickness, leishmaniasis, Chagas disease, filarial infections, mycetoma, paediatric HIV, hepatitis C, and covid-19. Since its inception in 2003, DNDi has delivered eight new treatments, including nifurtimox-eflornithine combination therapy (NECT) for late-stage sleeping sickness, and fexinidazole, the first all-oral drug for sleeping sickness.

Fexinidazole is an antiparasitic agent.[1] It has activity against Trypanosoma cruziTritrichomonas foetusTrichomonas vaginalis,Entamoeba histolytica,[1] Trypanosoma brucei,[2] and Leishmania donovani.[3] The biologically relevant active metabolites in vivo are the sulfoxide and sulfone [3][4]

Fexinidazole was discovered by the German pharmaceutical company Hoechst AG, but its development as a pharmaceutical was halted in the 1980s.[5] Fexinidazole is now being studied through a collaboration between Sanofi and the Drugs for Neglected Diseases Initiative for the treatment of Chagas disease and human African trypanosomiasis (sleeping sickness).[6][7] Fexinidazole is the first drug candidate for the treatment of advanced-stage sleeping sickness in thirty years.[8]

Fexinidazole is currently in phase II/III clinical development at Drugs for Neglected Diseases Initiative for the oral treatment of African trypanosomiasis (sleeping sickness). In May 2009, Sanofi (formerly known as sanofi-aventis) licensed the drug candidate to Drugs for Neglected Diseases Initiative for the development, manufacturing and distribution as a treatment of human African trypanosomiasis. Once approved, the companies plan to make the drug available on a nonprofit basis.

Fexinidazole was originally developed by a German pharmaceutical company called Hoechst, now part of Sanofi; however, its development was abandoned in the 1980s when the company gave up its tropical disease programs. Fexinidazole is one of a class of drugs known as azoles, like fluconazole, that work against fungi and may work against cancer.

  • Onset of trypanosomiasis is caused by Trypanosoma protozoa and it is said that every year 200,000 to 300,000 of new patients of African sleeping sickness fall sick. At present the number of patients of African sleeping sickness cannot be confirmed due to the low reliability of the investigative data. According to the WHO, at least 150,000 people died of African sleeping sickness in 1996 and it is said that its aftereffect remains in not less than 100,000 people. Beyond that, enormous is the damage to domestic animals caused by a disease called as nagana, and several hundred thousands of cattle which are to be protein sources for people die every year. Further, in the area of about 10,000,000 km2of savanna equal to the United States of America, cattle-breeding is impossible due to Trypanosoma. Thus, African sleeping sickness remarkably damages the health and the economical development of African people, and this is the reason why the WHO adopts the trypanosomiasis as one of the infectious diseases that should be controlled.
  • African sleeping sickness is a protozoal infectious disease by Trypanosoma transmitted through tsetse flies and the protozoa appear in the blood stream in about 10 days after infection. In the initial period of infection the protozoa multiply in the blood stream and give fever, physical weakness, headache, a pain of muscles and joints and a feeling of itching to proceed. On entering the chromic period, the central nerve is affected to show symptoms such as mental confusion and systemic convulsion, and finally the patients lapse into lethargy and are led to death.
  • The trypanosomiasis of domestic animals has Trypanosoma brucei brucei, Trypanosoma evansi, Trypanosoma congolense and Trypanosoma vivax as pathogens and is a communicable disease which affects domestic animals such as horses, cattle, pigs and dogs and, in addition, mice, guinea pigs, rabbits and the like. Particularly, the loss of cattle and horses is greatest and almost fetal, and they are led to anemia, edema, weakening and the like and fall dead in one month after infection.
  • In treating trypanosomiasis, pentamidine, melarsoprol, eflornithine and the like are used and there was a feeling in the 1960s that its eradication might be possible. However, these drugs are old and are gradually losing their efficacy. Particularly, the resistance to melarsoprol of an arsenic agent causes a big problem and the situation is so dire that patients with no efficacy only await death and the development of novel antitrypanosoma agents are strongly desired.
  • Trypanosoma mainly lives in the blood stream of the human body. This bloodstream energy metabolism depends on the glycolytic pathway localized in the organelle characteristic of the protozoa which is called as glycosome and the so-called oxidative phosphorylation does not function. However, in order to efficiently drive this glycolytic pathway, the produced NADH has to be reoxidized, and the glycerol-3-phosphate oxidation system of mitochondria plays an important role in this reoxidation. The terminal oxidase of this oxidation system functions as a quinol oxidase having a reduced ubiquinone as an electron donor and has properties greatly different from those of cytochrome oxidase of an aerobic respiration system which the host has. Particularly, a remarkable point is that the terminal oxidase of the oxidation system is non-sensitive to the cyanide which quickly inhibits the cytochrome oxidase of the host. Then, many researchers centered around Western countries have tried to develop drugs targeting this cyanide resistant oxidase but effective drugs having a selective toxicity have not been obtained.
  • Under these circumstances the present inventors et al. found that isoprenoid based physiologically active substances of ascochlorin, ascofuranone and derivatives thereof, particularly ascofuranone specifically inhibits the glycerol-3-phosphate oxidation system of trypanosome at a very low concentration of the order of nM and filed a patent application (Japanese Patent Publication A No. : H09-165332). They also clarified that acofuranone exhibits a very strong multiplication inhibition effect in the copresence of glycerin (Molecular and Biochemical Parasitology, 81: 127-136, 1996).
    In consideration of practical use of ascofuranone, it was found essential to discover agents which replace glycerin and exhibit an effect of the combined use in a small amount, and by using an alkaloid compound having an indole skeleton existing in a plant of the family Simaroubaceae together with ascofuranone, the prolongation of life and recovery effect in African seeping sickness was found and a patent application was filed (Japanese Patent Application No.: 2003-24643, Japanese Patent Publication A No.: 2004-23601).

Method for the preparation of fexinidazole, useful for the treatment of parasitic diseases, visceral leishmaniasis, chagas disease and human African trypanosomiasis. Family members of the product patent, WO2005037759, are expected to expire from October 2024. This to be the first application from Drugs for Neglected Diseases Initiative (DNDi) on this API. DNDi in collaboration with Sanofi, the Swiss Tropical & Public Health Institute and the University of Dundee, is developing fexinidazole, an antiparasitic agent, for treating human African trypanosomiasis (HAT) and visceral Leishmaniasis (VL). By June 2013, phase I clinical studies had been completed and at that time, DNDi was planning to initiate a phase II proof-of-concept study in VL patients in early 2013.

fexinidazole[inn], 59729-37-2, 1-Methyl-2-((4-(methylthio)phenoxy)methyl)-5-nitro-1H-imidazole, Fexinidazol, Fexinidazolum

Chemotherapeutically active nitro compounds. 4,5-Nitroimidazoles. Part III
Synthesis
By condensation of 4 – (methylmercapto) phenol (II) with 1-mehtyl-2-chloromethyl-5-nitroimidazole (I) by means of K2CO3 in DMF (1,2) Description:. Crystals, mp 116 C. References: 1) Raether, W., Winkelman, E.; Chemotherapeutically active nitro compounds 4,5-Nitroimidazoles Part III Arzneim-Forsch 1978, 28 (5):. 739 2) Winkelmann,… E., Raether, W. (Hoechst AG); DE 2531303.
Winkelman, E.; Raether, W.;… Chemotherapeutically active nitro compounds 4,5-Nitroimidazoles Part III Arzneim-Forsch 1978, 28, 5, 739
Arzneim-Forsch1978, 28, (5): 739

………………..

http://www.google.com/patents/EP1681280A1?cl=en

…………..

US 4042705

http://www.google.co.in/patents/US4042705

…………

new patent june 2014

WO-2014079497

Process for preparing fexinidazole – comprising the reaction of 1-methyl-2-hydroxymethyl-5-nitro-imidazole with methanesulfonyl chloride, followed by reaction with 4-methylmercapto-phenol, and further manipulative steps.

1-Methyl-2-hydroxymethyl-5-nitro-imidazole is (I) and 1-methyl-2-(4-methylmercapto-phenyloxymethyl)-5-nitro-imidazole (fexinidazole) is (II) (claim 1, page 12).

The synthesis of (II) via intermediate (I) is described (example 1, pages 6-8).

A process for preparing fexinidazole comprising the reaction of 1-methyl-2-hydroxymethyl-5-nitro-imidazole with methanesulfonyl chloride in the presence of a suspension of powdered alkaline carbonate (eg potassium carbonate) in an anhydrous organic solvent (eg acetone), followed by reaction with 4-methylmercapto-phenol, removal of hydrochloride salt, and isolation and purification is claimed. Also claimed is their use for treating parasitic diseases, visceral leishmaniasis, chagas disease, and human African trypanosomiasis. Fexinidazole is known to be an antiparasitic agent.

2-1-1983
The activity of fexinidazole (HOE 239) against experimental infections with Trypanosoma cruzi, trichomonads and Entamoeba histolytica.
Annals of tropical medicine and parasitology
1-1-1983
The use of the 2 substituted 5-nitroimidazole, Fexinidazole (Hoe 239) in the treatment of chronic T. brucei infections in mice.
Zeitschrift für Parasitenkunde (Berlin, Germany)
5-1-2011
1-Aryl-4-nitro-1H-imidazoles, a new promising series for the treatment of human African trypanosomiasis.
European journal of medicinal chemistry
2-1-2011
Compounds containing 2-substituted imidazole ring for treatment against human African trypanosomiasis.
Bioorganic & medicinal chemistry letters
1-1-2011
Trypanocidal activity of nitroaromatic prodrugs: current treatments and future perspectives.
Current topics in medicinal chemistry
12-1-2010
Potential new drugs for human African trypanosomiasis: some progress at last.
Current opinion in infectious diseases
7-1-2010
Cross-resistance to nitro drugs and implications for treatment of human African trypanosomiasis.
Antimicrobial agents and chemotherapy
1-1-2010
Fexinidazole–a new oral nitroimidazole drug candidate entering clinical development for the treatment of sleeping sickness.
PLoS neglected tropical diseases
1-1-1999
[Use of megazol for the treatment of trypanosomiasis].
Médecine tropicale : revue du Corps de santé colonial
11-1-1998
A method to assess invasion and intracellular replication of Trypanosoma cruzi based on differential uracil incorporation.
Journal of immunological methods
10-1-1996
Topical chemotherapy for experimental murine African CNS-trypanosomiasis: the successful use of the arsenical, melarsoprol, combined with the 5-nitroimidazoles, fexinidazole or MK-436.
Tropical medicine & international health : TM & IH
6-1-1991
Chemotherapy of CNS-trypanosomiasis: the combined use of the arsenicals and nitro-compounds.
11-15-2013
Targeting the human parasite Leishmania donovani: discovery of a new promising anti-infectious pharmacophore in 3-nitroimidazo[1,2-a]pyridine series.
Bioorganic & medicinal chemistry
10-1-2013
The R enantiomer of the antitubercular drug PA-824 as a potential oral treatment for visceral Leishmaniasis.
Antimicrobial agents and chemotherapy
2-1-2013
Assessing the essentiality of Leishmania donovani nitroreductase and its role in nitro drug activation.
Antimicrobial agents and chemotherapy
9-1-2012
Genotoxicity profile of fexinidazole–a drug candidate in clinical development for human African trypanomiasis (sleeping sickness).
Mutagenesis
7-15-2012
Discovery of nitroheterocycles active against African trypanosomes. In vitro screening and preliminary SAR studies.
Bioorganic & medicinal chemistry letters
2-1-2012
The anti-trypanosome drug fexinidazole shows potential for treating visceral leishmaniasis.
Science translational medicine
1-1-2012
Fexinidazole: a potential new drug candidate for Chagas disease.
PLoS neglected tropical diseases
1-1-2012
Management of trypanosomiasis and leishmaniasis.
British medical bulletin
12-1-2011
Antitrypanosomal activity of fexinidazole, a new oral nitroimidazole drug candidate for treatment of sleeping sickness.
Antimicrobial agents and chemotherapy
6-1-2011
Development of novel drugs for human African trypanosomiasis.
Future microbiology
US3682951 * 2 Nov 1970 8 Aug 1972 Searle & Co 1-{8 {62 -(1-adamantyloxy)halophenethyl{9 {0 imidazoles and congeners
US3714179 * 8 Sep 1970 30 Jan 1973 Searle & Co 1-alkyl-2-furfurylthioimidazoles and congeners
US3796704 * 16 Aug 1971 12 Mar 1974 Bayer Ag Phenyl-imidazolylalkanyl derivatives
US3828065 * 11 Dec 1972 6 Aug 1974 Searle & Co 2-methyl-5-nitro-1-(2-phenylthioethyl)imidazoles
US3842097 * 22 Jan 1973 15 Oct 1974 Searle & Co 2-(phenoxyalkylthio)imidazoles and congeners
US3910925 * 24 May 1974 7 Oct 1975 Searle & Co {8 2-(2-Methyl-5-nitro-1-imidazolyl)ethyl{9 benzo(b)pyridyloxy ethers
US3922277 * 14 Nov 1974 25 Nov 1975 Hoechst Ag (1-Alkyl-5-nitro-imidazolyl-2-alkyl)-pyridyl compounds
DE2124103A1 * 14 May 1971 25 Nov 1971 Title not available

References

  1.  Raether, W; Seidenath, H (1983). “The activity of fexinidazole (HOE 239) against experimental infections with Trypanosoma cruzi, trichomonads and Entamoeba histolytica”. Annals of Tropical Medicine and Parasitology 77 (1): 13–26. PMID 6411009.
  2.  Jennings, FW; Urquhart, GM (1983). “The use of the 2 substituted 5-nitroimidazole, Fexinidazole (Hoe 239) in the treatment of chronic T. brucei infections in mice”. Zeitschrift für Parasitenkunde 69 (5): 577–581. doi:10.1007/bf00926669PMID 6636983.
  3.  Wyllie, S; Patterson, S; Stojanovski, FRC; Norval, S; Kime, R; Read, RD; Fairlamb, AH (2012). “The anti-trypanosome drug fexinidazole shows potential for treating visceral leishmaniasis”Science Translational Medicine 4 (119): 119re1.doi:10.1126/scitranslmed.3003326PMC 3457684PMID 22301556.
  4.  Sokolova, AY; Wyllie, S; Patterson, S; Oza, SL; Read, RD; Fairlamb, AH (2010). “Cross-resistance to nitro drugs and implications for treatment of human African trypanosomiasis”. Antimicrobial Agents and Chemotherapy 54 (7): 2893–900. doi:10.1128/AAC.00332-10.PMID 20439607.
  5.  “Jump-Start on Slow Trek to Treatment for a Disease”New York Times. January 8, 2008.
  6.  “Fexinidazole Progresses into Clinical Development”. DNDi Newsletter. November 2009.
  7.  “Sanofi-aventis and DNDi enter into a Collaboration Agreement on a New Drug for Sleeping Sickness, Fexinidazole”. DNDi. May 18, 2009.
  8.  Torreele, E; Bourdin Trunz, B; Tweats, D; Kaiser, M; Brun, R; Mazué, G; Bray, MA; Pécoul, B (2010). “Fexinidazole–a new oral nitroimidazole drug candidate entering clinical development for the treatment of sleeping sickness”. In Boelaert, Marleen. PLOS Neglected Tropical Diseases 4 (12): e923. doi:10.1371/journal.pntd.0000923PMC 3006138PMID 21200426.