New Drug Approvals

Home » Phase2 drugs (Page 18)

Category Archives: Phase2 drugs

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

Blog Stats

  • 4,799,973 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

NAVEGLITAZAR (LY519818)

NAVEGLITAZAR

2(S)-Methoxy-3-[4-[3-(4-phenoxyphenoxy)propoxy]phenyl]propionic acid

476436-68-7

C25 H26 O6, 422.4703

  • CCRIS 9448
  • LY 519818
  • LY 9818
  • LY519818
  • LY9818
  • Naveglitazar
  • UNII-Y995M7GM0G

http://clinicaltrials.gov/search/intervention=NAVEGLITAZAR

Naveglitazar, a peroxisome proliferator-activated receptor (PPAR) modulator, had been in phase II clinical trials for the once-daily oral treatment of type 2 diabetes, however, no recent development for this indication has been reported. The compound was originally discovered through an ongoing research collaboration between Lilly and Ligand, but, in 2006, Lilly discontinued the development program.

Naveglitazar [LY519818; benzenepropanoic acid, alpha-methoxy-4-[3-(4-phenoxyphenoxy)propoxy], (alpha-S)-] is a nonthiozolidinedione peroxisome proliferator-activated receptor alpha-gamma dual, gamma-dominant agonist that has shown glucose-lowering potential in animal models and in the clinic.

Studies have been conducted to characterize the disposition, metabolism, and excretion of naveglitazar in mice, rats, and monkeys after oral and/or i.v. bolus administration.

………………………………

2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.

Martín JA, Brooks DA, Prieto L, González R, Torrado A, Rojo I, López de Uralde B, Lamas C, Ferritto R, Dolores Martín-Ortega M, Agejas J, Parra F, Rizzo JR, Rhodes GA, Robey RL, Alt CA, Wendel SR, Zhang TY, Reifel-Miller A, Montrose-Rafizadeh C, Brozinick JT, Hawkins E, Misener EA, Briere DA, Ardecky R, Fraser JD, Warshawsky AM.

Bioorg Med Chem Lett. 2005 Jan 3;15(1):51-5.

………………………………………..

http://www.google.im/patents/US20050020684?cl=un

EXAMPLE 153

′2-Methoxy-3-{3-[3-(4-phenoxy-phenoxy)-propoxy]-phenyl}-propionic acid

Figure US20050020684A1-20050127-C00299

The title compound was prepared from 3-(3-Hydroxy-phenyl)-2-methoxy-propionic acid methyl ester from Example 152, Step D with 4-(3-bromopropoxy)1-phenoxybenzene in a manner analogous as in Example 152, Step E. MS (ES) for C25H26O6[M+NH4]+: 440.2, [M+Na]+: 445.2. 1H-NMR (CDCl3, 200.15 MHz): 7.33-7.17 (m, 3H), 7.07-6.78 (m, 10H), 4.15 (dt, 4H, J=1.9, 6.2), 4.03 (dd, 1H, J=7.3, 4.3), 3.40 (s, 3H), 3.13 (dd, 1H, J=14.2, 4.6), 2.98 (dd, 1H, J=14.0, 7.5), 2.25 (qui, 2H, J=5.9)ppm.

Celgene oral Crohn’s drug GED-0301, Mongersen impresses in Phase II

Celgene签署$26亿协议获克罗恩病反义药物GED-0301

Nogra制药,Celgene

Nogra Pharma Limited

公司寡核苷酸(Oligonucleotides)

克罗恩病(Crohn’s disease)

Shares in Celgene Corp have risen steadily following promising mid-stage data of its closely-watched Crohn’s disease drug mongersen.

Company Nogra Pharma Ltd.
Description Antisense oligonucleotide targeting SMAD family member 7 (MADH7; SMAD7)
Molecular Target SMAD family member 7 (MADH7) (SMAD7)
Mechanism of Action
Therapeutic Modality Nucleic acid: Linear RNA: Antisense
Latest Stage of Development Phase II
Standard Indication Crohn’s disease
Indication Details Treat moderate to severe Crohn’s disease
Regulatory Designation
Partner

Celgene Corp.

Mongersen (GED-0301) from Celgene Corp. (NASDAQ:CELG) produced clinical remission rates as high as 65.1% in a Phase II trial in 166 patients with moderate to severe Crohn’s disease, according to an abstract published in advance of the United European Gastroenterology’s meeting in Vienna.

In the trial, 55% of patients receiving 40 mg/day of mongersen and 65.1% of those receiving 160 mg/day achieved clinical remission compared with 9.5% of placebo patients (p<0.0001 for both). A cohort receiving 10 mg/day achieved a clinical remission rate of 12.2%, which was not significantly better than placebo.

The study’s primary outcomes were clinical remission, defined by a CDAI score less than 150 at day 15 and maintained for more than two weeks, and safety. Mongersen was well-tolerated, and toxicities associated with systemically active antisense therapies were not observed.

The study’s secondary endpoint is clinical response, defined as a CDAI score reduction of 100 points at day 28. Those rates were dose-dependent: 36.6%, 57.5% and 72.1% for the low, medium and high doses compared with 16.7% for placebo.

Celgene said it plans to start Phase III testing of mongersen shortly. The company paid $710 million up front to obtain exclusive, worldwide rights to the antisense oligonucleotide targeting SMAD family member 7 (MADH7; SMAD7) from Nogra Pharma Ltd. (Dublin, Ireland) in April. Nogra is eligible for $1.9 billion in milestones, plus tiered single-digit royalties.

 GED-0301, an antisense oligonucleotide targeting the SMAD7 gene, is in phase II clinical trials at Nogra Pharma for the oral treatment of moderate to severe Crohn’s disease.

生物技术公司新基(Celgene)从爱尔兰制药商Nogra制药手中获得了一种处于后期临床开发的克罗恩病(Crohn’s disease)药物GED-0301。GED-0301是一种口服反义药物,靶向于Smad7信使RNA(mRNA),该药开发用于中度至重度克罗恩病 的治疗。反义药物是一种合成的核酸拷贝,旨在结合导致疾病的基因的mRNA,关闭基因的表达;口服;【Celgene签署$26亿协议获克罗恩病反义药物 GED-0301】http://www.hfoom.com/product/20140425/8311.html

 

Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract suffered by approximately one million patients in the United States. The two most common forms of IBD are Crohn’s disease (CD) and ulcerative colitis (UC). Although CD can affect the entire gastrointestinal tract, it primarily affects the ilieum (the distal or lower portion of the small intestine) and the large intestine. UC primarily affects the colon and the rectum. Current treatment for both CD and UC include aminosalicylates (e.g., 5- aminosalicylic acid, sulfasalazine and mesalamine), antibiotics (e.g., ciprofloxacin and metronidazole), corticosteroids (e.g., budesonide or prednisone), immunosuppressants (e.g., azathioprine or methotrexate) and tumor necrosis factor (TNF) antagonists (e.g., infliximab (Remicade®)). Patient response to these therapies varies with disease severity and it can vary over cycles of active inflammation and remission. Moreover, many of the current therapies for IBD are associated with undesirable side effects.

Although the etiologies of CD and UC are unknown, both are considered inflammatory diseases of the intestinal mucosa. Recent studies have demonstrated that TGF-β 1 acts as a potent immunoregulator able to control mucosal intestinal inflammation. TGF-βΙ binds a heterodimeric transmembrane serine/threonine kinase receptor containing two subunits, TGF-βΙ Rl and TGF-βΙ R2. Upon ligand binding, the TGF-βΙ Rl receptor is phosphorylated by the constitutively active TGF-βΙ R2 receptor and signal is propagated to the nucleus by proteins belonging to the SMAD family. Activated TGF-β Ι Rl directly phosphorylates SMAD2 and SMAD3 proteins, which then interact with SMAD4. The complex of SMAD2/SMAD3/SMAD4 translocates to the nucleus and modulates the transcription of certain genes.

Additional studies have demonstrated that another SMAD protein, SMAD7, also plays a role in inflammation. SMAD7, an intracellular protein, has been shown to interfere with binding of SMAD2/SMAD3 to the TGF-βΙ Rl preventing phosphorylation and activation of these proteins. Further, increased expression of SMAD7 protein is associated with an inhibition of TGF-βΙ mediated-signaling. Mucosal samples from IBD patients are characterized by high levels of SMAD7 and reduced levels of phosphorylated-SMAD3 indicating that TGF-βΙ -mediated signaling is compromised in these patients.

Recent studies have focused on SMAD7 as a target for treating patients suffering from IBD.

Such therapies include anti-SMAD7 antisense therapies. As such, there is a need for methods based on predictive biomarkers that can be used to identify patients that are likely (or unlikely) to respond to treatment with anti- SMAD7 therapies.

GTCGCCCCTTCTCCCCGCAGC

GED-0301, Mongersen

Phosphorothioate antisense oligonucleotide targeting human mothers against decapentaplegic homolog 7 (SMAD7) gene, whose sequence is 5′-GTCGCCCCTTCTCCCCGCAGC-3′, wherein ‘C’ at postions 3 and 16 is 5-methyl 2′-deoxycytidine 5′-monophosphate

WO 2004087920

http://www.google.com/patents/WO2004087920A1?cl=en

…………………………

WO 2013037970

http://www.google.com/patents/WO2013037970A1?cl=en

…………………

WO 2013158868

http://www.google.com/patents/WO2013158868A1?cl=en

……………………………………………

WO 2014140333

http://www.google.com/patents/WO2014140333A1?cl=en

5*-GTCGCCCCTTCTCCCCGCAGC-3* (SEQ ID NO: 3).

 

Reference
1 BADARU, A.; PIHOKER, C.: ‘Type 2 diabetes in childhood: clinical characteristics and role of beta-cell autoimmunity‘ CURR. DIAB. REP. vol. 12, 2012, pages 75 – 81
2 * BHAT ET AL: “Antisense inhibition of 11betahydroxysteroid dehydrogenase type 1 improves diabetes in a novel cortisone-induced diabetic KK mouse model“, BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 365, no. 4, 20 November 2007 (2007-11-20), pages 740-745, XP022384861, ISSN: 0006-291X, DOI: 10.1016/J.BBRC.2007.11.032
3 * GUTIERREZ-AGUILAR ET AL: “Minor contribution of SMAD7 and KLF10 variants to genetic susceptibility of type 2 diabetes“, DIABETES & METABOLISM, PARIS, AMSTERDAM, NL, vol. 33, no. 5, 10 October 2007 (2007-10-10), pages 372-378, XP022327080, ISSN: 1262-3636, DOI: 10.1016/J.DIABET.2007.06.002
4 * H. Y. CHEN ET AL: “The Protective Role of Smad7 in Diabetic Kidney Disease: Mechanism and Therapeutic Potential“, DIABETES, vol. 60, no. 2, 27 October 2010 (2010-10-27), pages 590-601, XP55071874, ISSN: 0012-1797, DOI: 10.2337/db10-0403
5 HONG, S. ET AL.: ‘Smad7 sensitizes tumor necrosis factor induced apoptosis through the inhibition of antiapoptotic gene expression by suppressing activation of the nuclear factor-kappaB pathway‘ CANCER RES. vol. 67, 2007, pages 9577 – 9583
6 HOOK, S. M. ET AL.: ‘Smad2: A candidate gene for the murine autoimmune diabetes locus Idd21.1‘ 1. CLIN. ENDOCRINOL. METAB. vol. 96, 2011, pages E2072 – E2077
7 KAWAMOTO, K. ET AL.: ‘Transforming growth factor beta 1 (TGF-?1) and rapamycin synergize to effectively suppress human T cell responses via upregulation of FoxP3+ Tregs‘ TRANSPL. IMMUNOL. vol. 23, 2010, pages 28 – 33
8 LI, M. O.; FLAVELL, R. A.: ‘TGF-beta: a master of all T cell trades‘ CELL vol. 134, 2008, pages 392 – 404
9 * LIANG Y ET AL: “Reduction in Glucagon Receptor Expression by an Antisense Oligonucleotide Ameliorates Diabetic Syndrome in db/db Mice“, DIABETES, AMERICAN DIABETES ASSOCIATION, US, vol. 53, February 2004 (2004-02), pages 410-417, XP002995165, ISSN: 0012-1797, DOI: 10.2337/DIABETES.53.2.410
10 * LU ZHU ET AL: “Unraveling the biological functions of Smad7 with mouse models“, CELL & BIOSCIENCE, BIOMED CENTRAL LTD, LONDON, UK, vol. 1, no. 1, 28 December 2011 (2011-12-28), page 44, XP021132085, ISSN: 2045-3701, DOI: 10.1186/2045-3701-1-44
11 LUO, X. ET AL.: ‘Systemic transforming growth factor-?1 gene therapy induces Foxp3+ regulatory cells, restores self-tolerance, and facilitates regeneration of beta cell function in overtly diabetic nonobese diabetic mice‘ TRANSPLANTATION vol. 79, 2005, pages 1091 – 1096
12 MARGOLLES-CLARK, E. ET AL.: ‘Small molecule costimulatory blockade: organic dye inhibitors of the CD40-CD154 interaction‘ J. MOL. MED. vol. 87, 2009, pages 1133 – 1143
13 * MIZOBUCHI TERUAKI ET AL: “Differential expression of Smad7 transcripts identifies the CD4+CD45RChigh regulatory T cells that mediate type V collagen-induced tolerance to lung allografts“, THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 171, no. 3, 1 August 2003 (2003-08-01), pages 1140-1147, XP002430371, ISSN: 0022-1767
14 MONTELEONE, G. ET AL.: ‘A failure of transforming growth factor-?1 negative regulation maintains sustained NF-KB activation in gut inflammation‘ J. BIOL. CHEM. vol. 279, 2004, pages 3925 – 3932
15 MORITANI, M. ET AL.: ‘Abrogation of autoimmune diabetes in nonobese diabetic mice and protection against effector lymphocytes by transgenic paracrine TGF-?1‘ J. CLIN. INVEST. vol. 102, 1998, pages 499 – 506
16 * NORA G. SMART ET AL: “Conditional Expression of Smad7 in Pancreatic [beta] Cells Disrupts TGF-[beta] Signaling and Induces Reversible Diabetes Mellitus“, CELL, vol. 88, no. 2, 31 January 2006 (2006-01-31), page 561, XP55071875, ISSN: 0092-8674, DOI: 10.1371/journal.pbio.0040039
17 OLIVIERI, A. ET AL.: ‘Serum transforming growth factor ?1 during diabetes development in non-obese diabetic mice and humans‘ CLIN. EXP. IMMUNOL. vol. 162, 2010, pages 407 – 414
18 Remington’s Pharmaceutical Sciences‘, 1990, MACK PUBLISHING COMPANY
19 ROEP, B. O. ET AL.: ‘Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes‘ NAT. REV. IMMUNOL. vol. 4, 2004, pages 989 – 997
20 * S. M. HOOK ET AL: “Smad2: A Candidate Gene for the Murine Autoimmune Diabetes Locus Idd21.1“, JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM, vol. 96, no. 12, 5 October 2011 (2011-10-05), pages E2072-E2077, XP55071877, ISSN: 0021-972X, DOI: 10.1210/jc.2011-0463
21 SHODA, L. K. ET AL.: ‘A comprehensive review of interventions in the NOD mouse and implications for translation‘ IMMUNITY vol. 23, 2005, pages 115 – 126
22 SMART, N. G. ET AL.: ‘Conditional expression of Smad7 in pancreatic beta cells disrupts TGF-beta signaling and induces reversible diabetes mellitus‘ PLOS BIOL. vol. 4, 2006, page E39
23 WALLBERG, M. ET AL.: ‘An islet-specific pulse of TGF-? abrogates CTL function and promotes ? cell survival independent of Foxp3+ T cells‘ J. IMMUNOL. vol. 186, 2011, pages 2543 – 2551
24 YAN, X.; CHEN, Y. G.: ‘Smad7: not only a regulator, but also a cross-talk mediator of TGF-beta signalling‘ BIOCHEM. J. vol. 434, 2011, pages 1 – 10
WO2003037368A2 * Oct 31, 2002 May 8, 2003 Andreas Steinbrecher Smad7 inhibitors for the treatment of cns diseases
WO2009129544A1 * Apr 20, 2009 Oct 22, 2009 Baxter International Inc. Microsphere-based composition for preventing and/or reversing new-onset autoimmune diabetes
WO2010054826A1 Nov 13, 2009 May 20, 2010 Giuliani International Limited Antisense compositions and methods of making and using same

Vibegron ビベグロン

Chemical structure for Vibegron (USAN)

 

Vibegron, MK-4618, KRP 114V

update FDA APPROVED 12/23/2020, GEMTESA, To treat overactive bladder

UNII-M5TSE03W5U; M5TSE03W5U; D10433
Molecular Formula: C26H28N4O3   Molecular Weight: 444.52552
phase 2 for the treatment of overactive bladder
 (6S)-N-[4-([(2S,5R)-5-[(R)-Hydroxy(phenyl)methyl]pyrrolidin-2-yl]methyl)phenyl]-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-6-carboxamide
(6S)-N-[4-[[(2S,5R)-5-[(R)-hydroxy(phenyl)methyl]pyrrolidin-2-yl]methyl]phenyl]-4-oxo-7,8-dihydro-6H-pyrrolo[1,2-a]pyrimidine-6-carboxamide

Target-based Actions Beta 3 adrenoceptor agonist
Indications Overactive bladder; Urinary incontinence

UPDATE 2018/9/21  pmda Beova JAPAN 2018Kyorin Pharmaceutical, under license from Merck, is developing vibegron (phase II, September 2014) for the treating of overactive bladder. In July 2014, Merck has granted to Kyorin an exclusive license to develop, manufacture and commercialize vibegron in Japan.

MK-4618 is being developed in phase II clinical trials at Merck & Co. for the treatment of overactive bladder. The company had been developing the compound for the treatment of endocrine disorders and hypertension; however, recent progress reports are not available at present.

In 2014, Merck licensed the product to Kyorin for development and commercialization in Japan.

The function of the lower urinary tract is to store and periodically release urine. This requires the orchestration of storage and micturition reflexes which involve a variety of afferent and efferent neural pathways, leading to modulation of central and peripheral neuroeffector mechanisms, and resultant coordinated regulation of sympathetic and parasympathetic components of the autonomic nervous system as well as somatic motor pathways. These proximally regulate the contractile state of bladder (detrusor) and urethral smooth muscle, and urethral sphincter striated muscle.

β Adrenergic receptors (βAR) are present in detrusor smooth muscle of various species, including human, rat, guinea pig, rabbit, ferret, dog, cat, pig and non-human primate. However, pharmacological studies indicate there are marked species differences in the receptor subtypes mediating relaxation of the isolated detrusor; β1AR predominate in cats and guinea pig, β2AR predominate in rabbit, and β3AR contribute or predominate in dog, rat, ferret, pig, cynomolgus and human detrusor. Expression of βAR subtypes in the human and rat detrusor has been examined by a variety of techniques, and the presence of β3AR was confirmed using in situ hybridization and/or reverse transcription-polymerase chain reaction (RT-PCR). Real time quantitative PCR analyses of β1AR, β2AR and β3AR mRNAs in bladder tissue from patients undergoing radical cystectomy revealed a preponderance of β3AR mRNA (97%, cf 1.5% for β1AR mRNA and 1.4% for β2AR mRNA). Moreover, β3AR mRNA expression was equivalent in control and obstructed human bladders. These data suggest that bladder outlet obstruction does not result in downregulation of β3AR, or in alteration of β3AR-mediated detrusor relaxation. β3AR responsiveness also has been compared in bladder strips obtained during cystectomy or enterocystoplasty from patients judged to have normal bladder function, and from patients with detrusor hyporeflexia or hyperreflexia. No differences in the extent or potency of β3AR agonist mediated relaxation were observed, consistent with the concept that the β3AR activation is an effective way of relaxing the detrusor in normal and pathogenic states.

Functional evidence in support of an important role for the β3AR in urine storage emanates from studies in vivo. Following intravenous administration to rats, the rodent selective β3AR agonist CL316243 reduces bladder pressure and in cystomeric studies increases bladder capacity leading to prolongation of micturition interval without increasing residual urine volume.

Overactive bladder is characterized by the symptoms of urinary urgency, with or without urgency urinary incontinence, usually associated with frequency and nocturia. The prevalence of OAB in the United States and Europe has been estimated at 16 to 17% in both women and men over the age of 18 years. Overactive bladder is most often classified as idiopathic, but can also be secondary to neurological condition, bladder outlet obstruction, and other causes. From a pathophysiologic perspective, the overactive bladder symptom complex, especially when associated with urge incontinence, is suggestive of detrusor overactivity. Urgency with or without incontinence has been shown to negatively impact both social and medical well-being, and represents a significant burden in terms of annual direct and indirect healthcare expenditures. Importantly, current medical therapy for urgency (with or without incontinence) is suboptimal, as many patients either do not demonstrate an adequate response to current treatments, and/or are unable to tolerate current treatments (for example, dry mouth associated with anticholinergic therapy). Therefore, there is need for new, well-tolerated therapies that effectively treat urinary frequency, urgency and incontinence, either as monotherapy or in combination with available therapies. Agents that relax bladder smooth muscle, such as β3AR agonists, are expected to be effective for treating such urinary disorders.

PATENT

http://www.google.com/patents/WO2013062881A1?cl=en

Figure imgf000013_0001

EXAMPLE 3

To a three neck flask equipped with a N2 inlet, a thermo couple probe was charged pyrrolidine i-11 (10.0 g), sodium salt i-12 (7.87 g), followed by IPA (40 mL) and water (24 mL). 5 N HC1 (14.9 mL) was then slowly added over a period of 20 min to adjust pH = 3.3- 3.5, maintaining the batch temperature below 35 °C. Solid EDC hydrochloride (7.47 g) was charged in portions over 30 min. The reaction mixture was aged at RT for additional 0.5 – 1 h, aqueous ammonia (14%) was added dropwise to pH ~8.6. The batch was seeded and aged for additional 1 h to form a slurry bed. The rest aqueous ammonia (14%, 53.2 ml total) was added dropwise over 6 h. The resulting thick slurry was aged 2-3 h before filtration. The wet-cake was displacement washed with 30% IPA (30 mL), followed by 15% IPA (2 x 20mL) and water (2 X 20mL). The cake was suction dried under N2 overnight to afford 14.3 g of compound of Formula (I)-

1H NMR (DMSO) δ 10.40 (s, NH), 7.92 (d, J = 6.8, 1H), 7.50 (m, 2H), 7.32 (m, 2H), 7.29 (m, 2H), 7.21 (m, 1H), 7.16 (m, 2H), 6.24 (d, J = 6.8, 1H), 5.13 (dd, J = 9.6, 3.1, 1H), 5.08 (br s, OH), 4.22 (d, J = 7.2, 1H), 3.19 (p, J = 7.0, 1H), 3.16-3.01 (m, 3H), 2.65 (m, 1H), 2.59-2.49 (m, 2H), 2.45 (br s, NH), 2.16 (ddt, J = 13.0, 9.6, 3.1, 1H), 1.58 (m, 1H), 1.39 (m, 1H), 1.31-1.24 (m, 2H).

13C NMR (DMSO) δ 167.52, 165.85, 159.83, 154.56, 144.19, 136.48, 135.66, 129.16, 127.71, 126.78, 126.62, 119.07, 112.00, 76.71, 64.34, 61.05, 59.60, 42.22, 31.26, 30.12, 27.09, 23.82.

HPLC method – For monitoring conversion

Column: XBridge C18 cm 15 cm x 4.6 mm, 3.5 μιη particle size;

Column Temp. : 35 °C; Flow rate: 1.5 mL/min; Detection: 220 nm;

Mobile phase: A. 5 mM Na2B407.10 H20 B: Acetonitrile

Gradient:

HPLC method – For level of amide epimer detection

Column: Chiralpak AD-H 5 μηι, 250 mm x 4.6 mm.

Column Temp: 35 °C; Flow rate: 1.0 mL/min; Detection: 250 nm;

Mobile phase: Isocratic 30% Ethanol in hexanes + 0.1% isobutylamine

PATENT

WO 2009124167

http://www.google.com/patents/WO2009124167A1?cl=en

EXAMPLE 103

(6y)-N-r4-({(25′. 5R)-5-r(R)-hvdroxy(phenvnmethyl1pyrrolidin-2-yl}methvnphenyl1-4-oxo- 4,6J,8-tetrahydropyiτolori,2-α1pyrimidine-6-carboxamide

ter?-butyl(2R. 55f)-2-rCR)-hvdroxy(phenvnmethyl1-5-r4-({r(65f)-4-oxo-4.6.7.8-

tetrahydropyrrolof 1.2-alpyrimidin-6- yl]carbonyl} amino)benzyl]pyrrolidine- 1 – carboxylate

To a solution of i-13a (21.4 g, 55.9 mmol) in N,N-dimethylformamide (100 ml) at O0C was added [(65)-4-oxo-4,6,7,8-tetrahydropyrrolo[l,2-α]pyrimidine-6-carboxylic acid (11.1 g, 61.5 mmol), followed by 1 -hydroxybenzotriazole (i-44, 7.55 g, 55.9 mmol), N-(3- dimethylaminopropyl)-Nl-ethylcarbodiimide hydrochloride (16.1 g, 84.0 mmol) and N,N- diisopropylethylamine (29.2 ml, 168 mmol). The reaction mixture was stirred from O0C to ambient temperature for 2 h. Water (600 ml) was added and it was extracted with dichloromethane (600 ml x 2). The combined organic layers were dried over Na2SO4. After removal of the volatiles, the residue was purified by using a Biotage Horizon® system (0-5% then 5% methanol with 10% ammonia/dichloromethane mixture) to afford the title compound which contained 8% of the minor diastereomer. It was further purified by supercritical fluid chromatography (chiral AS column, 40% methanol) to afford the title compound as a pale yellow solid (22.0 g, 72%). 1H NMR (CDCl3): δ 9.61 (s, IH), 7.93 (d, J = 6.6 Hz, IH), 7.49 (d, J = 8.4 Hz, 2H), 7.35-7.28 (m, 5H), 7.13 (d, J = 8.5 Hz, 2H), 6.40 (d, J = 6.7 Hz, IH), 5.36 (d, J = 8.6 Hz, IH), 4.38 (m, IH), 4.12-4.04 (m, 2H), 3.46 (m,lH), 3.15-3.06 (m, 2H), 2.91 (dd, J = 13.1, 9.0 Hz, IH), 2.55 (m, IH), 2.38 (m, IH), 1.71-1.49 (m, 13H). LC-MS 567.4 (M+23).

(6S)-N-\4-( U2S. 5R)-5-r(R)-hvdroxy(phenyl)methyl1pyrrolidin-2-

yl}methyl)phenyl1-4-oxo-4,6J,8-tetrahvdropyrrolori,2-α1pyrimidine-6- carboxamide

To a solution of the intermediate from Step A (2.50 g, 4.59 mmol) in dichloromethane (40 ml) was added trifluoroacetic acid (15 ml). The reaction mixture was stirred at ambient temperature for 1.5 h. After removal of the volatiles, saturated NaHCCh was added to make the PH value to 8-9. The mixture was then extracted with dichloromethane. The combined organic layers were dried over Na2SO4. After concentration, crystallization from methanol/acetonitrile afforded the title compound as a white solid (1.23g, 60%). 1H NMR (DMSO-Cl6): δ 10.40 (s, IH), 7.91 (d, J = 6.7 Hz, IH), 7.49 (d, J = 8.3 Hz, 2H), 7.32-7.26 (m, 4H), 7.21 (m, IH), 7.15 (d, J = 8.4 Hz, 2H), 6.23 (d, J = 6.7 Hz, IH), 5.11 (dd, J = 9.6, 2.9 Hz, IH), 5.10 (br, IH), 4.21 (d, J = 7.1 Hz, IH), 3.20-3.00 (m, 4H), 2.66-2.51 (m, 3H), 2.16 (m, IH), 1.57 (m, IH), 1.38 (m, IH), 1.29-1.23 (m, 2H). LC-MS 445.3 (M+l).

Using the Biological Assays described above, the human β3 functional activity of Example 103 was determined to be between 11 to 100 nM.

PATENT

CHECK STRUCTURE…………….CAUTION

http://www.google.com/patents/US8247415

Figure US08247415-20120821-C00547

Figure US08247415-20120821-C00015

CAUTION…………….

Example 103(6S)-N-[4-({(2S,5R)-5-[(R)-hydroxy(phenyl)methyl]pyrrolidin-2-yl}methyl)phenyl]-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-α]pyrimidine-6-carboxamide

Step A: tert-butyl(2R,5S)-2-[(R)-hydroxy(phenyl)methyl]-5-[4-({[(6S)-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-α]pyrimidin-6-yl]carbonyl}amino)benzyl]pyrrolidine-1-carboxylate

To a solution of i-13a (21.4 g, 55.9 mmol) in N,N-dimethylformamide (100 ml) at 0° C. was added [(6S)-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-α]pyrimidine-6-carboxylic acid (11.1 g, 61.5 mmol), followed by 1-hydroxybenzotriazole (i-44, 7.55 g, 55.9 mmol), N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (16.1 g, 84.0 mmol) and N,N-diisopropylethylamine (29.2 ml, 168 mmol). The reaction mixture was stirred from 0° C. to ambient temperature for 2 h. Water (600 ml) was added and it was extracted with dichloromethane (600 ml×2). The combined organic layers were dried over Na2SO4. After removal of the volatiles, the residue was purified by using a Biotage Horizon® system (0-5% then 5% methanol with 10% ammonia/dichloromethane mixture) to afford the title compound which contained 8% of the minor diastereomer. It was further purified by supercritical fluid chromatography (chiral AS column, 40% methanol) to afford the title compound as a pale yellow solid (22.0 g, 72%). 1H NMR (CDCl3): δ 9.61 (s, 1H), 7.93 (d, J=6.6 Hz, 1H), 7.49 (d, J=8.4 Hz, 2H), 7.35-7.28 (m, 5H), 7.13 (d, J=8.5 Hz, 2H), 6.40 (d, J=6.7 Hz, 1H), 5.36 (d, J=8.6 Hz, 1H), 4.38 (m, 1H), 4.12-4.04 (m, 2H), 3.46 (m, 1H), 3.15-3.06 (m, 2H), 2.91 (dd, J=13.1, 9.0 Hz, 1H), 2.55 (m, 1H), 2.38 (m, 1H), 1.71-1.49 (m, 13H). LC-MS 567.4 (M+23).

Step B: (6S)-N-[4-({(2S,5R)-5-[(R)-hydroxy(phenyl)methyl]pyrrolidin-2-yl}methyl)phenyl]-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-α]pyrimidine-6-carboxamide

To a solution of the intermediate from Step A (2.50 g, 4.59 mmol) in dichloromethane (40 ml) was added trifluoroacetic acid (15 ml). The reaction mixture was stirred at ambient temperature for 1.5 h. After removal of the volatiles, saturated NaHCO3 was added to make the PH value to 8-9. The mixture was then extracted with dichloromethane. The combined organic layers were dried over Na2SO4. After concentration, crystallization from methanol/acetonitrile afforded the title compound as a white solid (1.23 g, 60%). 1H NMR (DMSO-d6): δ 10.40 (s, 1H), 7.91 (d, J=6.7 Hz, 1H), 7.49 (d, J=8.3 Hz, 2H), 7.32-7.26 (m, 4H), 7.21 (m, 1H), 7.15 (d, J=8.4 Hz, 2H), 6.23 (d, J=6.7 Hz, 1H), 5.11 (dd, J=9.6, 2.9 Hz, 1H), 5.10 (br, 1H), 4.21 (d, J=7.1 Hz, 1H), 3.20-3.00 (m, 4H), 2.66-2.51 (m, 3H), 2.16 (m, 1H), 1.57 (m, 1H), 1.38 (m, 1H), 1.29-1.23 (m, 2H). LC-MS 445.3 (M+1).

Using the Biological Assays described above, the human β3 functional activity of Example 103 was determined to be between 11 to 100 nM.

PATENT

WO2014150639

http://patentscope.wipo.int/search/en/detail.jsf?docId=WO2014150639&recNum=4&docAn=US2014023858&queryString=EN_ALL:nmr%20AND%20PA:merck&maxRec=11148

Step 6. Preparation of Compound 1-7 from Compound 1-6 and Compound A-2

To a three neck flask equipped with a N2 inlet, a thermo couple probe was charged pyrrolidine hemihydrate 1-6 (10.3 g), sodium salt A-2 (7.87 g), followed by IPA (40 mL) and water (24 mL). 5 N HC1 (14.9 mL) was then slowly added over a period of 20 minutes to adjust pH = 3.3-3.5, maintaining the batch temperature below 35°C. Solid EDC hydrochloride (7.47 g) was charged in portions over 30 minutes. The reaction mixture was aged at RT for additional 0.5 – 1 hour, aqueous ammonia (14%) was added dropwise to pH -8.6. The batch was seeded and aged for additional 1 hour to form a slurry bed. The rest aqueous ammonia (14%, 53.2 ml total) was added dropwise over 6 hours. The resulting thick slurry was aged 2-3 hours before filtration. The wet-cake was displacement washed with 30% IPA (30 mL), followed by 15% IPA (2 x 20mL) and water (2 X 20mL). The cake was suction dried under N2 overnight to afford 14.3 g of compound 1-7.

1H NMR (DMSO) δ 10.40 (s, NH), 7.92 (d, J = 6.8, 1H), 7.50 (m, 2H), 7.32 (m, 2H), 7.29 (m, 2H), 7.21 (m, 1H), 7.16 (m, 2H), 6.24 (d, J = 6.8, 1H), 5.13 (dd, J = 9.6, 3.1, 1H), 5.08 (br s, OH), 4.22 (d, J = 7.2, 1H), 3.19 (p, J = 7.0, 1H), 3.16-3.01 (m, 3H), 2.65 (m, 1H), 2.59-2.49 (m, 2H), 2.45 (br s, NH), 2.16 (ddt, J = 13.0, 9.6, 3.1, 1H), 1.58 (m, 1H), 1.39 (m, 1H), 1.31-1.24 (m, 2H).

13C NMR (DMSO) δ 167.52, 165.85, 159.83, 154.56, 144.19, 136.48, 135.66, 129.16, 127.71, 126.78, 126.62, 119.07, 112.00, 76.71, 64.34, 61.05, 59.60, 42.22, 31.26, 30.12, 27.09, 23.82.

The crystalline freebase anhydrous form I of Compound 1-7 can be characterized by XRPD by

PATENT

WO-2014150633
Merck Sharp & Dohme Corp
Process for preparing stable immobilized ketoreductase comprises bonding of recombinant ketoreductase to the resin in a solvent. Useful for synthesis of vibegron intermediates. For a concurrent filling see WO2014150639, claiming the method for immobilization of ketoreductase. Picks up from WO2013062881, claiming the non enzymatic synthesis of vibegron and intermediates.

PAPER

Discovery of Vibegron: A Potent and Selective β3 Adrenergic Receptor Agonist for the Treatment of Overactive Bladder

Merck Research Laboratories, 2015 Galloping Hill Road, PO Box 539, Kenilworth, New Jersey 07033, United States
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.5b01372
Publication Date (Web): December 27, 2015
Copyright © 2015 American Chemical Society
*Telephone: (908) 740-0287. E-mail scott.edmondson@merck.com.

http://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.5b01372

http://pubs.acs.org/doi/suppl/10.1021/acs.jmedchem.5b01372/suppl_file/jm5b01372_si_001.pdf

Abstract Image

The discovery of vibegron, a potent and selective human β3-AR agonist for the treatment of overactive bladder (OAB), is described. An early-generation clinical β3-AR agonist MK-0634 (3) exhibited efficacy in humans for the treatment of OAB, but development was discontinued due to unacceptable structure-based toxicity in preclinical species. Optimization of a series of second-generation pyrrolidine-derived β3-AR agonists included reducing the risk for phospholipidosis, the risk of formation of disproportionate human metabolites, and the risk of formation of high levels of circulating metabolites in preclinical species. These efforts resulted in the discovery of vibegron, which possesses improved druglike properties and an overall superior preclinical profile compared to MK-0634. Structure–activity relationships leading to the discovery of vibegron and a summary of its preclinical profile are described.

Reference
1 H.P. Kaiser, et al., “Catalytic Hydrogenation of Pyrroles at Atmospheric Pressure“, J. Org. Chem., vol. 49, No. 22, p. 4203-4209 (1984).
A study of the efficacy and safety of MK-4618 in patients with overactive bladder (OAB) (MK-4618-008 EXT1) (NCT01314872)
ClinicalTrials.gov Web Site 2011, April 28
WO2011043942A1 * Sep 27, 2010 Apr 14, 2011 Merck Sharp & Dohme Corp. Combination therapy using a beta 3 adrenergic receptor agonist and an antimuscarinic agent
US20090253705 * Apr 2, 2009 Oct 8, 2009 Richard Berger Hydroxymethyl pyrrolidines as beta 3 adrenergic receptor agonists
US20110028481 * Apr 2, 2009 Feb 3, 2011 Richard Berger Hydroxymethyl pyrrolidines as beta 3 adrenergic receptor agonists
 
Citing Patent Filing date Publication date Applicant Title
US8642661 Aug 2, 2011 Feb 4, 2014 Altherx, Inc. Pharmaceutical combinations of beta-3 adrenergic receptor agonists and muscarinic receptor antagonists
US8653260 Jun 20, 2012 Feb 18, 2014 Merck Sharp & Dohme Corp. Hydroxymethyl pyrrolidines as beta 3 adrenergic receptor agonists
US20120202819 * Sep 27, 2010 Aug 9, 2012 Merck Sharp & Dohme Corporation Combination therapy using a beta 3 adrenergic receptor agonists and an antimuscarinic agent
US20020028835 Jul 12, 2001 Mar 7, 2002 Baihua Hu Cyclic amine phenyl beta-3 adrenergic receptor agonists
US20070185136 Feb 2, 2007 Aug 9, 2007 Sanofi-Aventis Sulphonamide derivatives, their preparation and their therapeutic application
US20110028481 Apr 2, 2009 Feb 3, 2011 Richard Berger Hydroxymethyl pyrrolidines as beta 3 adrenergic receptor agonists
WO2003072572A1 Feb 17, 2003 Sep 4, 2003 Jennifer Anne Lafontaine Beta3-adrenergic receptor agonists
8-22-2012
Hydroxymethyl pyrrolidines as [beta]3 adrenergic receptor agonists

////////////C1CC(NC1CC2=CC=C(C=C2)NC(=O)C3CCC4=NC=CC(=O)N34)C(C5=CC=CC=C5)O

INCB-039110, Janus kinase-1 (JAK-1) inhibitor……..for the treatment of rheumatoid arthritis, myelofibrosis, rheumatoid arthritis and plaque psoriasis.

Figure imgf000005_0001 INCB-39110,

CAS 1334298-90-6

INCB-039110, Jak1 tyrosine kinase inhibitor

3-​Azetidineacetonitril​e, 1-​[1-​[[3-​fluoro-​2-​(trifluoromethyl)​-​4-​pyridinyl]​carbonyl]​-​4-​piperidinyl]​-​3-​[4-​(7H-​pyrrolo[2,​3-​d]​pyrimidin-​4-​yl)​-​1H-​pyrazol-​1-​yl]​-

 C26H23F4N9O (MW, 553.51)

{ l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile

2-(3-(4-(7H-pyrrolo[2,3-( Jpyrimidin-4-yl)-lH- pyrazol- 1 -yl)- 1 -( 1 -(3 -fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin- 3-yl)acetonitrile

2-(3-(4-(7H- Pyrrolo[2,3 -i/]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)- 1 -(1 -(3 -fluoro-2- (trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipate MAY BE THE DRUG… HAS CAS 1334302-63-4

Figure imgf000005_0001Adipic acidADIPATE OF INCB-39110

ALSO/OR

 

Figure US20130060026A1-20130307-C00027

3-​Azetidineacetonitril​e, 1-​[1-​(3-​fluorobenzoyl)​-​4-​methyl-​4-​piperidinyl]​-​3-​[4-​(7H-​pyrrolo[2,​3-​d]​pyrimidin-​4-​yl)​-​1H-​pyrazol-​1-​yl]​-​, 2,​2,​2-​trifluoroacetateMAY BE THE DRUG ????…  HAS CAS  1334300-52-5

US 2011/0224190 is the pdt patent

 

 

Incyte Corporation

 

Clinical trials

 

IN PHASE 2 for the treatment of rheumatoid arthritis, myelofibrosis, rheumatoid arthritis and plaque psoriasis.

SEE

http://clinicaltrials.gov/show/NCT01633372

 

 

Jak2 tyrosine kinase inhibitor; Jak1 tyrosine kinase inhibitor

Breast tumor; Chronic obstructive pulmonary disease; Crohns disease; Inflammatory bowel disease; Influenza virus infection; Insulin dependent diabetes; Liver tumor; Multiple sclerosis; Prostate tumor; Rheumatoid arthritis; SARS coronavirus infection

Used for treating cancers (eg prostate cancer, hepatic cancer and pancreatic cancer) and autoimmune diseases. Follows on from WO2013036611, claiming the process for preparing the same JAK inhibitor. Incyte is developing INCB-39110 (phase II, September 2014), for the oral treatment of myelofibrosis, hematological neoplasm and non-small cell lung cancer.

INCB-039110 is a Jak1 inhibitor in phase II clinical studies at Incyte for the treatment of rheumatoid arthritis, myelofibrosis, rheumatoid arthritis and plaque psoriasis. The company is also conducting a phase I clinical study for the treatment of advanced or metastatic solid tumors.

Protein kinases (PKs) regulate divINCB-039110 is a Jak1 inhibitor in phase II clinical studies at Incyte for the treatment of rheumatoid arthritis, myelofibrosis, rheumatoid arthritis and plaque psoriasis. The company is also conducting a phase I clinical study for the treatment of advanced or metastatic solid tumors.erse biological processes including cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair, and regeneration, among others. Protein kinases also play specialized roles in a host of human diseases including cancer. Cytokines, low-molecular weight polypeptides or glycoproteins, regulate many pathways involved in the host

inflammatory response to sepsis. Cytokines influence cell differentiation,

proliferation and activation, and can modulate both pro-inflammatory and antiinflammatory responses to allow the host to react appropriately to pathogens.

Signaling of a wide range of cytokines involves the Janus kinase family (JAKs) of protein tyrosine kinases and Signal Transducers and Activators of Transcription

(STATs). There are four known mammalian JAKs: JAK1 (Janus kinase-1), JAK2, JAK3 (also known as Janus kinase, leukocyte; JAKL; and L-JAK), and TYK2

(protein-tyros ine kinase 2).

Cytokine-stimulated immune and inflammatory responses contribute to pathogenesis of diseases: pathologies such as severe combined immunodeficiency (SCID) arise from suppression of the immune system, while a hyperactive or inappropriate immune/inflammatory response contributes to the pathology of autoimmune diseases (e.g., asthma, systemic lupus erythematosus, thyroiditis, 20443-0253WO1 (INCY0124-WO1) PATENT myocarditis), and illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000) Arthritis Res 2(1): 16-32).

Deficiencies in expression of JAKs are associated with many disease states. For example, Jakl-/- mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373-83). Jak2-/- mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive

erythropoiesis.

The JAK/STAT pathway, and in particular all four JAKs, are believed to play a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract. Multiple cytokines that signal through JAKs have been linked to inflammatory diseases/conditions of the upper respiratory tract, such as those affecting the nose and sinuses (e.g., rhinitis and sinusitis) whether classically allergic reactions or not. The JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of the eye and chronic allergic responses.

Activation of JAK/STAT in cancers may occur by cytokine stimulation (e.g. IL-6 or GM-CSF) or by a reduction in the endogenous suppressors of JAK signaling such as SOCS (suppressor or cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V., and Kovarik, J., Neoplasm. 49:349-355, 2002).

Activation of STAT signaling, as well as other pathways downstream of JAKs (e.g., Akt), has been correlated with poor prognosis in many cancer types (Bowman, T., et al. Oncogene 19:2474-2488, 2000). Elevated levels of circulating cytokines that signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be beneficial to cancer patients for reasons that extend beyond potential anti-tumor activity.

JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorders, e.g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387- 397). Inhibition of the JAK2V617F kinase decreases proliferation of hematopoietic cells, suggesting JAK2 as a potential target for pharmacologic inhibition in patients with PV, ET, and MMM. 20443-0253WO1 (INCY0124-WO1) PATENT

Inhibition of the JAKs may benefit patients suffering from skin immune disorders such as psoriasis, and skin sensitization. The maintenance of psoriasis is believed to depend on a number of inflammatory cytokines in addition to various chemokines and growth factors (JCI, 1 13 : 1664-1675), many of which signal through JAKs (Adv Pharmacol. 2000;47: 113-74).

JAKl plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL-6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (Fonesca, J.E. et al, Autoimmunity

Reviews, 8:538-42, 2009). Because IL-6 signals, at least in part, through JAKl, antagonizing IL-6 directly or indirectly through JAKl inhibition is expected to provide clinical benefit (Guschin, D., N., et al Embo J 14: 1421, 1995; Smolen, J. S., et al. Lancet 371 :987, 2008). Moreover, in some cancers JAKl is mutated resulting in constitutive undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S A.106:9414-8, 2009; Flex E., et al.J Exp Med. 205:751-8, 2008). In other autoimmune diseases and cancers elevated systemic levels of inflammatory cytokines that activate JAKl may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAKl inhibition. Selective inhibitors of JAKl may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases.

Selective inhibitors of JAKl, relative to other JAK kinases, may have multiple therapeutic advantages over less selective inhibitors. With respect to selectivity against JAK2, a number of important cytokines and growth factors signal through JAK2 including, for example, erythropoietin (Epo) and thrombopoietin (Tpo)

(Parganas E, et al. Cell. 93:385-95, 1998). Epo is a key growth factor for red blood cells production; hence a paucity of Epo-dependent signaling can result in reduced numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034-45, 2006). Tpo, another example of a JAK2-dependent growth factor, plays a central role in controlling the proliferation and maturation of megakaryocytes – the cells from which platelets are produced (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced Tpo signaling would decrease megakaryocyte numbers (megakaryocytopenia) and lower circulating platelet counts (thrombocytopenia). This can result in undesirable 20443-0253WO1 (INCY0124-WO1) PATENT and/or uncontrollable bleeding. Reduced inhibition of other JAKs, such as JAK3 and Tyk2, may also be desirable as humans lacking functional version of these kinases have been shown to suffer from numerous maladies such as severe-combined immunodeficiency or hyperimmunoglobulin E syndrome (Minegishi, Y, et al.

Immunity 25:745-55, 2006; Macchi P, et al. Nature. 377:65-8, 1995). Therefore a JAK1 inhibitor with reduced affinity for other JAKs would have significant

advantages over a less-selective inhibitor with respect to reduced side effects involving immune suppression, anemia and thrombocytopenia.

……………………….

http://www.google.com/patents/US20110224190

 

EXAMPLESThe example compounds below containing one or more chiral centers were obtained in enantiomerically pure form or as scalemic mixtures, unless otherwise specified.Unless otherwise indicated, the example compounds were purified by preparativeHPLC using acidic conditions (method A) and were obtained as a TFA salt or using basic conditions (method B) and were obtained as a free base.Method A:Column: Waters Sun Fire C18, 5 μm particle size, 30×100 mm;
Mobile phase: water (0.1% TFA)/acetonitrile
Flow rate: 60 mL/min
Gradient: 5 min or 12 min from 5% acetonitrile/95% water to 100% acetonitrileMethod B:Column: Waters X Bridge C18, 5 μm particle size, 30×100 mm;
Mobile phase: water (0.15% NH4OH)/acetonitrileMethod C:Column: C18 column, 5 μm OBD
Mobile phase: water+0.05% NH4OH (A), CH3CN+0.05% NH4OH (B)Gradient: 5% B to 100% B in 15 minFlow rate: 60 mL/minExample 1
{1-{1-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile

Step A: tert-Butyl 3-Oxoazetidine-1-carboxylate

To a mixture of tert-butyl 3-hydroxyazetidine-1-carboxylate (10.0 g, 57.7 mmol), dimethyl sulfoxide (24.0 mL, 338 mmol), triethylamine (40 mL, 300 mmol) and methylene chloride (2.0 mL) was added sulfur trioxide-pyridine complex (40 g, 200 mmol) portionwise at 0° C. The mixture was stirred for 3 hours, quenched with brine, and extracted with methylene chloride. The combined extracts were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column (0-6% ethyl acetate (EtOAc) in hexanes) to give tert-butyl 3-oxoazetidine-1-carboxylate (5.1 g, 52% yield).

Step B: tert-Butyl 3-(Cyanomethylene)azetidine-1-carboxylate

An oven-dried 1 L 4-neck round bottom flask fitted with stir bar, septa, nitrogen inlet, 250 ml addition funnel and thermocouple was charged with sodium hydride (5.6 g, 0.14 mol) and tetrahydrofuran (THF) (140 mL) under a nitrogen atmosphere. The mixture was chilled to 3° C., and then charged with diethyl cyanomethylphosphonate (22.4 mL, 0.138 mol) dropwise via a syringe over 20 minutes. The solution became a light yellow slurry. The reaction was then stirred for 75 minutes while warming to 18.2° C. A solution of tert-butyl 3-oxoazetidine-1-carboxylate (20 g, 0.1 mol) in tetrahydrofuran (280 mL) was prepared in an oven-dried round bottom, charged to the addition funnel via canula, then added to the reaction mixture dropwise over 25 minutes. The reaction solution became red in color. The reaction was allowed to stir overnight. The reaction was checked after 24 hours by TLC (70% hexane/EtOAc) and found to be complete. The reaction was diluted with 200 mL of 20% brine and 250 mL of EtOAc. The solution was partitioned and the aqueous phase was extracted with 250 mL of EtOAc. The combined organic phase was dried over MgSO4 and filtered, evaporated under reduced pressure, and purified by flash chromatography (0% to 20% EtOAc/hexanes, 150 g flash column) to give the desired product, tert-butyl 3-(cyanomethylene)azetidine-1-carboxylate (15 g, 66.1% yield).

Step C: 4-Chloro-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine

To a suspension of sodium hydride (36.141 g, 903.62 mmol) in N,N-dimethylacetamide (118 mL) at −5° C. (ice/salt bath) was added a dark solution of 4-chloropyrrolo[2,3-d]pyrimidine (119.37 g, 777.30 mmol) in N,N-dimethylacetamide (237 mL) slowly. The flask and addition funnel were rinsed with N,N-dimethylacetamide (30 mL). A large amount of gas was evolved immediately. The mixture became a slightly cloudy orange mixture. The mixture was stirred at 0° C. for 60 min to give a light brown turbid mixture. To the mixture was slowly added [2-(trimethylsilyl)ethoxy]methyl chloride (152.40 g, 914.11 mmol) and the reaction was stirred at 0° C. for 1 h. The reaction was quenched by addition of 12 mL of H2O slowly. More water (120 mL) was added followed by methyl tert-butyl ether (MTBE) (120 mL). The mixture was stirred for 10 min. The organic layer was separated. The aqueous layer was extracted with another portion of MTBE (120 mL). The organic extracts were combined, washed with brine (120 mL×2) and concentrated under reduced pressure to give the crude product 4-chloro-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine as a dark oil. Yield: 85.07 g (97%); LC-MS: 284.1 (M+H)+. It was carried to the next reaction without purification.

Step D: 4-(1H-Pyrazol-4-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine

A 1000 mL round bottom flask was charged with 4-chloro-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (10.00 g, 35.23 mmol), 1-butanol (25.0 mL), 1-(1-ethoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (15.66 g, 52.85 mmol), water (25.0 mL) and potassium carbonate (12.17 g, 88.08 mmol). This solution was degased 4 times, filling with nitrogen each time. To the solution was added tetrakis(triphenylphosphine)palladium(0) (4.071 g, 3.523 mmol). The solution was degased 4 times, filling with nitrogen each time. The mixture was stirred overnight at 100° C. After being cooled to room temperature, the mixture was filtered through a bed of celite and the celite was rinsed with ethyl acetate (42 mL). The filtrate was combined, and the organic layer was separated. The aqueous layer was extracted with ethyl acetate. The organic extracts were combined and concentrated under vacuum with a bath temperature of 30-70° C. to give the final compound 4-(1H-pyrazol-4-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine. Yield: 78%. LC-MS: 316.2 (M+H)+.

Step E: tert-Butyl 3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidine-1-carboxylate

A 2 L round bottom flask fitted with overhead stirring, septa and nitrogen inlet was charged with tert-butyl 3-(cyanomethylene)azetidine-1-carboxylate (9.17 g, 0.0472 mol), 4-(1H-pyrazol-4-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (14.9 g, 0.0472 mol) and acetonitrile (300 mL). The resulting solution was heterogeneous. To the solution was added 1,8-diazabicyclo[5.4.0]undec-7-ene (8.48 mL, 0.0567 mol) portionwise via syringe over 3 min at room temperature. The solution slowly became homogeneous and yellow in color. The reaction was allowed to stir at room temperature for 3 h. The reaction was complete by HPLC and LC/MS and was concentrated by rotary evaporation to remove acetonitrile (˜150 mL). EtOAc (100 mL) was added followed by 100 ml of 20% brine. The two phases were partitioned. The aqueous phase was extracted with 150 mL of EtOAC. The combine organic phases were dried over MgSO4, filtered and concentrated to yield an orange oil. Purification by flash chromatography (150 grams silica, 60% EtOAc/hexanes, loaded with CH2Cl2) yielded the title compound tert-butyl 3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidine-1-carboxylate as a yellow oil (21.1 g, 88% yield). LC-MS: [M+H]+=510.3.

Step F: {3-[4-(7-{[2-(Trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile dihydrochloride

To a solution of tert-butyl 3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidine-1-carboxylate (2 g, 3.9 mmol) in 10 mL of THF was added 10 mL of 4 N HCl in dioxane. The solution was stirred at room temperature for 1 hour and concentrated in vacuo to provide 1.9 g (99%) of the title compound as a white powder solid, which was used for the next reaction without purification. LC-MS: [M+H]+=410.3.

Step G: tert-Butyl 4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-1-yl}piperidine-1-carboxylate

Into the solution of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile dihydrochloride (2.6 g, 6.3 mmol), tert-butyl 4-oxo-1-piperidinecarboxylate (1.3 g, 6.3 mmol) in THF (30 mL) were added N,N-diisopropylethylamine (4.4 mL, 25 mmol) and sodium triacetoxyborohydride (2.2 g, 10 mmol). The mixture was stirred at room temperature overnight. After adding 20 mL of brine, the solution was extracted with EtOAc. The extract was dried over anhydrous Na2SO4 and concentrated. The residue was purified by combiflash column eluting with 30-80% EtOAc in hexanes to give the desired product, tert-butyl 4-{3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-1-yl}piperidine-1-carboxylate. Yield: 3.2 g (86%); LC-MS: [M+H]+=593.3.

Step H: {1-Piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile trihydrochloride

To a solution of tert-butyl 4-{3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-1-yl}piperidine-1-carboxylate (3.2 g, 5.4 mmol) in 10 mL of THF was added 10 mL of 4 N HCl in dioxane. The reaction mixture was stirred at room temperature for 2 hours. Removing solvents under reduced pressure yielded 3.25 g (100%) of {1-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile trihydrochloride as a white powder solid, which was used directly in the next reaction. LC-MS: [M+H]+=493.3. 1H NMR (400 MHz, DMSO-d6): δ 9.42 (s 1H), 9.21 (s, 1H), 8.89 (s, 1H), 8.69 (s, 1H), 7.97 (s, 1H), 7.39 (d, 1H), 5.68 (s, 2H), 4.96 (d, 2H), 4.56 (m, 2H), 4.02-3.63 (m, 2H), 3.55 (s, 2H), 3.53 (t, 2H), 3.49-3.31 (3, 3H), 2.81 (m, 2H), 2.12 (d, 2H), 1.79 (m, 2H), 0.83 (t, 2H), −0.10 (s, 9H).

Step I: {1-{1-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile

A mixture of {1-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile trihydrochloride (1.22 g, 2.03 mmol), 3-fluoro-2-(trifluoromethyl)isonicotinic acid (460 mg, 2.2 mmol), benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate (1.07 g, 2.42 mmol), and triethylamine (2.0 mL, 14 mmol) in dimethylformamide (DMF) (20.0 mL) was stirred at room temperature overnight. LS-MS showed the reaction was complete. EtOAc (60 mL) and saturated NaHCO3 aqueous solution (60 mL) were added to the reaction mixture. After stirring at room temperature for 10 minutes, the organic phase was separated and the aqueous layer was extracted with EtOAc three times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. Purification by flash chromatography provided the desired product {1-{1-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile. LC-MS: 684.3 (M+H)+.

Step J: {1-{1-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile

Into a solution of {1-{1-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile (56 mg, 0.1 mmol) in methylene chloride (1.5 mL) was added trifluoroacetic acid (1.5 mL). The mixture was stirred at room temperature for 2 hours. After removing the solvents in vacuum, the residue was dissolved in a methanol solution containing 20% ethylenediamine. After being stirred at room temperature for 1 hour, the solution was purified by HPLC (method B) to give the title compound. LC-MS: 554.3 (M+H)+; 1H NMR (400 MHz, CDCl3): 9.71 (s, 1H), 8.82 (s, 1H), 8.55 (d, J=4.6 Hz, 1H), 8.39 (s, 1H), 8.30 (s, 1H), 7.52 (t, J=4.6 Hz, 1H), 7.39 (dd, J1=3.4 Hz, J2=1.5 Hz, 1H), 6.77 (dd, J1=3.6 Hz, J2=0.7 Hz, 1H), 4.18 (m, 1H), 3.75 (m, 2H), 3.63 (dd, J1=7.8 Hz, J2=3.7 Hz, 2H), 3.45 (m, 2H), 3.38 (s, 2H), 3.11 (m, 1H), 2.57 (m, 1H), 1.72 (m, 1H), 1.60 (m, 1H), 1.48 (m, 1H), 1.40 (m, 1H).

 

………………………..

http://www.google.com/patents/US20130060026

Example 1Synthesis of 4-(1H-pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (5)

Step 1. 4-Chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (3)

To a flask equipped with a nitrogen inlet, an addition funnel, a thermowell, and the mechanical stirrer was added 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (1, 600 g, 3.91 mol) and N,N-dimethylacetimide (DMAC, 9.6 L) at room temperature. The mixture was cooled to 0-5° C. in an ice/brine bath before solid sodium hydride (NaH, 60 wt %, 174 g, 4.35 mol, 1.1 equiv) was added in portions at 0-5° C. The reaction mixture turned into a dark solution after 15 minutes. Trimethylsilylethoxymethyl chloride (2, SEM-Cl, 763 mL, 4.31 mol, 1.1 equiv) was then added slowly via an addition funnel at a rate that the internal reaction temperature did not exceed 5° C. The reaction mixture was then stirred at 0-5° C. for 30 minutes. When the reaction was deemed complete determined by TLC and HPLC, the reaction mixture was quenched by water (1 L). The mixture was then diluted with water (12 L) and methyl tert-butyl ether (MTBE) (8 L). The two layers were separated and the aqueous layer was extracted with MTBE (8 L). The combined organic layers were washed with water (2×4 L) and brine (4 L) and solvent switched to 1-butanol. The solution of crude product (3) in 1-butanol was used in the subsequent Suzuki coupling reaction without further purification. Alternatively, the organic solution of the crude product (3) in MTBE was dried over sodium sulfate (Na2SO4). The solvents were removed under reduced pressure. The residue was then dissolved in heptane (2 L), filtered and loaded onto a silica gel (SiO2, 3.5 Kg) column eluting with heptane (6 L), 95% heptane/ethyl acetate (12 L), 90% heptane/ethyl acetate (10 L), and finally 80% heptane/ethyl acetate (10 L). The fractions containing the pure desired product were combined and concentrated under reduced pressure to give 4-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (3, 987 g, 1109.8 g theoretical, 88.9% yield) as a pale yellow oil which partially solidified to an oily solid on standing at room temperature. For 3: 1H NMR (DMSO-d6, 300 MHz) δ 8.67 (s, 1H), 7.87 (d, 1H, J=3.8 Hz), 6.71 (d, 1H, J=3.6 Hz), 5.63 (s, 2H), 3.50 (t, 2H, J=7.9 Hz), 0.80 (t, 2H, J=8.1 Hz), 1.24 (s, 9H) ppm; 13C NMR (DMSO-d6, 100 MHz) δ 151.3, 150.8, 150.7, 131.5, 116.9, 99.3, 72.9, 65.8, 17.1, −1.48 ppm; C12H18ClN3OSi (MW 283.83), LCMS (EI) m/e 284/286 (M++H).

Step 2. 4-(1H-Pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (5)

To a reactor equipped with the overhead stirrer, a condenser, a thermowell, and a nitrogen inlet was charged water (H2O, 9.0 L), solid potassium carbonate (K2CO3, 4461 g, 32.28 mol, 2.42 equiv), 4-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (3, 3597 g, 12.67 mol), 1-(1-ethoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (4, 3550 g, 13.34 mol, 1.05 equiv), and 1-butanol (27 L) at room temperature. The resulting reaction mixture was degassed three timed backfilling with nitrogen each time before being treated with tetrakis(triphenylphosphine)palladium(0) (Pd(PPh3)4, 46 g, 0.040 mol, 0.003 equiv) at room temperature. The resulting reaction mixture was heated to gentle reflux (about 90° C.) for 1-4 hours. When the reaction was deemed complete determined by HPLC, the reaction mixture was gradually cooled down to room temperature before being filtered through a Celite bed. The Celite bed was washed with ethyl acetate (2×2 L) before the filtrates and washing solution were combined. The two layers were separated, and the aqueous layer was extracted with ethyl acetate (12 L). The combined organic layers were concentrated under reduced pressure to remove solvents, and the crude 4-(1-(1-ethoxyethyl)-1H-pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (6) was directly charged back to the reactor with tetrahydrofuran (THF, 4.2 L) for the subsequent acid-promoted de-protection reaction without further purification.

To a suspension of crude 4-(1-(1-ethoxyethyl)-1H-pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (6), made as described above, in tetrahydrofuran (THF, 4.2 L) in the reactor was charged water (H2O, 20.8 L), and a 10% aqueous HCl solution (16.2 L, 45.89 mol, 3.44 equiv) at room temperature. The resulting reaction mixture was stirred at 16-30° C. for 2-5 hours. When the reaction was deemed complete by HPLC analysis, the reaction mixture was treated with a 30% aqueous sodium hydroxide (NaOH) solution (4 L, 50.42 mol, 3.78 equiv) at room temperature. The resulting reaction mixture was stirred at room temperature for 1-2 hours. The solids were collected by filtration and washed with water (2×5 L). The wet cake was charged back to the reactor with acetonitrile (21.6 L), and resulting suspension was heated to gentle reflux for 1-2 hours. The clear solution was then gradually cooled down to room temperature with stirring, and solids were precipitated out from the solution with cooling. The mixture was stirred at room temperature for an additional 1-2 hours. The solids were collected by filtration, washed with acetonitrile (2×3.5 L), and dried in oven under reduced pressure at 45-55° C. to constant weight to afford 4-(1H-pyrazol-4-yl)-7-(2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (5, 3281.7 g, 3996.8 g theoretical, 82.1% yield) as white crystalline solids (99.5 area % by HPLC). For 5: 1H NMR (DMSO-d6, 400 MHz) δ 13.41 (br. s, 1H), 8.74 (s, 1H), 8.67 (br. s, 1H), 8.35 (br. s, 1H), 7.72 (d, 1H, J=3.7 Hz), 7.10 (d, 1H, J=3.7 Hz), 5.61 (s, 2H), 3.51 (t, 2H, J=8.2 Hz), 0.81 (t, 2H, J=8.2 Hz), 0.13 (s, 9H) ppm; C15H21N5OSi (MW, 315.45), LCMS (EI) m/e 316 (M++H).

Example 2tert-Butyl 3-(cyanomethylene)azetidine-1-carboxylate (13)

Step 1. 1-Benzhydrylazetidin-3-ol hydrochloride (9)

A solution of diphenylmethanamine (7, 2737 g, 15.0 mol, 1.04 equiv) in methanol (MeOH, 6 L) was treated with 2-(chloromethyl)oxirane (8, 1330 g, 14.5 mol) from an addition funnel at room temperature. During the initial addition a slight endotherm was noticed. The resulting reaction mixture was stirred at room temperature for 3 days before being warmed to reflux for an additional 3 days. When TLC showed that the reaction was deemed complete, the reaction mixture was first cooled down to room temperature and then to 0-5° C. in an ice bath. The solids were collected by filtration and washed with acetone (4 L) to give the first crop of the crude desired product (9, 1516 g). The filtrate was concentrated under reduced pressure and the resulting semisolid was diluted with acetone (1 L). This solid was then collected by filtration to give the second crop of the crude desired product (9, 221 g). The crude product, 1-benzhydrylazetidin-3-ol hydrochloride (9, 1737 g, 3998.7 g theoretical, 43.4% yield), was found to be sufficiently pure to be used in the subsequent reaction without further purification. For 9: 1H NMR (DMSO-d6, 300 MHz), δ 12.28 (br. d, 1H), 7.7 (m, 5H), 7.49 (m, 5H), 6.38 (d, 1H), 4.72 (br. s, 1H), 4.46 (m, 1H), 4.12 (m, 2H), 3.85 (m, 2H) ppm; C16H18ClNO (free base of 9, C16K7NO MW, 239.31), LCMS (EI) m/e 240 (M++H).

Step 2. tert-Butyl 3-hydroxyazetidine-1-carboxylate (10)

A suspension of 1-benzhydrylazetidin-3-ol hydrochloride (9, 625 g, 2.27 mol) in a 10% solution of aqueous sodium carbonate (Na2CO3, 5 L) and dichloromethane (CH2Cl2, 5 L) was stirred at room temperature until all solids were dissolved. The two layers were separated, and the aqueous layer was extracted with dichloromethane (CH2Cl2, 2 L). The combined organics extracts were dried over sodium sulfate (Na2SO4) and concentrated under reduced pressure. This resulting crude free base of 9 was then dissolved in THF (6 L) and the solution was placed into a large Parr bomb. Di-tert-butyl dicarbonate (BOC2O, 545 g, 2.5 mol, 1.1 equiv) and 20% palladium (Pd) on carbon (125 g, 50% wet) were added to the Parr bomb. The vessel was charged to 30 psi with hydrogen gas (H2) and stirred under steady hydrogen atmosphere (vessel was recharged three times to maintain the pressure at 30 psi) at room temperature for 18 h. When HPLC showed that the reaction was complete (when no more hydrogen was taken up), the reaction mixture was filtered through a Celite pad and the Celite pad was washed with THF (4 L). The filtrates were concentrated under reduced pressure to remove the solvent and the residue was loaded onto a Biotage 150 column with a minimum amount of dichloromethane (CH2Cl2). The column was eluted with 20-50% ethyl acetate in heptane and the fractions containing the pure desired product (10) were collected and combined. The solvents were removed under reduced pressure to afford tert-butyl 3-hydroxyazetidine-1-carboxylate (10, 357 g, 393.2 g theoretical, 90.8% yield) as colorless oil, which solidified upon standing at room temperature in vacuum. For 10: 1HNMR (CDCl3, 300 MHz), δ 4.56 (m 1H), 4.13 (m, 2H), 3.81 (m, 2H), 1.43 (s, 9H) ppm.

Step 3. tert-Butyl 3-oxoazetidine-1-carboxylate (11)

A solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (10, 50 g, 289 mmol) in ethyl acetate (400 mL) was cooled to 0° C. The resulting solution was then treated with solid TEMPO (0.5 g, 3.2 mmol, 0.011 equiv) and a solution of potassium bromide (KBr, 3.9 g, 33.2 mmol, 0.115 equiv) in water (60 mL) at 0-5° C. While keeping the reaction temperature between 0-5° C. a solution of saturated aqueous sodium bicarbonate (NaHCO3, 450 mL) and an aqueous sodium hypochlorite solution (NaClO, 10-13% available chlorine, 450 mL) were added. Once the solution of sodium hypochlorite was added, the color of the reaction mixture was changed immediately. When additional amount of sodium hypochlorite solution was added, the color of the reaction mixture was gradually faded. When TLC showed that all of the starting material was consumed, the color of the reaction mixture was no longer changed. The reaction mixture was then diluted with ethyl acetate (EtOAc, 500 mL) and two layers were separated. The organic layer was washed with water (500 mL) and the saturated aqueous sodium chloride solution (500 mL) and dried over sodium sulfate (Na2SO4). The solvent was then removed under reduced pressure to give the crude product, tert-butyl 3-oxoazetidine-1-carboxylate (11, 48 g, 49.47 g theoretical, 97% yield), which was found to be sufficiently pure and was used directly in the subsequent reaction without further purification. For crude 11: 1HNMR (CDCl3, 300 MHz), δ 4.65 (s, 4H), 1.42 (s, 9H) ppm.

Step 4. tert-Butyl 3-(cyanomethylene)azetidine-1-carboxylate (13)

Diethyl cyanomethyl phosphate (12, 745 g, 4.20 mol, 1.20 equiv) and anhydrous tetrahydrofuran (THF, 9 L) was added to a four-neck flask equipped with a thermowell, an addition funnel and the nitrogen protection tube at room temperature. The solution was cooled with an ice-methanol bath to −14° C. and a 1.0 M solution of potassium tert-butoxide (t-BuOK) in anhydrous tetrahydrofuran (THF, 3.85 L, 3.85 mol, 1.1 equiv) was added over 20 minutes keeping the reaction temperature below −5° C. The resulting reaction mixture was stirred for 3 hours at −10° C. and a solution of 1-tert-butoxycarbonyl-3-azetidinone (11, 600 g, 3.50 mol) in anhydrous tetrahydrofuran (THF, 2 L) was added over 2 h keeping the internal temperature below −5° C. The reaction mixture was stirred at −5 to −10° C. over 1 hour and then slowly warmed up to room temperature and stirred at room temperature for overnight. The reaction mixture was then diluted with water (4.5 L) and saturated aqueous sodium chloride solution (NaCl, 4.5 L) and extracted with ethyl acetate (EtOAc, 2×9 L). The combined organic layers were washed with brine (6 L) and dried over anhydrous sodium sulfate (Na2SO4). The organic solvent was removed under reduced pressure and the residue was diluted with dichloromethane (CH2Cl2, 4 L) before being absorbed onto silica gel (SiO2, 1.5 Kg). The crude product, which was absorbed on silica gel, was purified by flash column chromatography (SiO2, 3.5 Kg, 0-25% EtOAc/hexanes gradient elution) to afford tert-butyl 3-(cyanomethylene)azetidine-1-carboxylate (13, 414.7 g, 679.8 g theoretical, 61% yield) as white solid. For 13: 1H NMR (CDCl3, 300 MHz), δ 5.40 (m, 1H), 4.70 (m, 2H), 4.61 (m, 2H), 1.46 (s, 9H) ppm; C10H14N2O2 (MW, 194.23), LCMS (EI) m/e 217 (M′+Na).

Example 3(3-Fluoro-2-(trifluoromethyl)pyridin-4-yl)(1,4-dioxa-8-azaspiro[4,5]decan-8-yl)methanone (17)

Step 1. 1,4-Dioxa-8-azaspiro[4.5]decane (15)

To a 30 L reactor equipped with a mechanic stirrer, an addition funnel and a septum was charged sodium hydroxide (NaOH, 1.4 kg, 35 mol) and water (7 L, 3.13 kg, 17.43 mol). To the solution thus obtained was added 1,4-dioxa-8-azaspiro[4.5]decane hydrochloric acid (14, 3.13 kg, 17.43 mol). The mixture was stirred at 25° C. for 30 minutes. Then the solution was saturated with sodium chloride (1.3 kg) and extracted with 2-methyl-tetrahydrofuran (3×7 L). The combined organic layer was dried with anhydrous sodium sulfate (1.3 kg), filtered and concentrated under reduced pressure (70 mmHg) at 50° C. The yellow oil thus obtained was distilled under reduced pressure (80 mmHg, bp: 115° C. to 120° C.) to give compound 15 (2.34 kg, 16.36 mol, 93.8%) as a clear oil, which was used directly in the subsequent coupling reaction.

Step 2. (3-Fluoro-2-(trifluoromethyl)pyridin-4-yl)(1,4-dioxa-8-azaspiro[4,5]decan-8-yl)methanone (17)

To a dried 100 L reactor equipped with a mechanic stirrer, an addition funnel, a thermometer and a vacuum outlet were placed 3-fluoro-2-(trifluoromethyl)isonicotinic acid (16, 3.0 kg, 14.35 mol), benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate (BOP reagent, 7.6 kg, 17.2 mol, 1.20 equiv) in dimethylformamide (DMF, 18 L). To the resulting solution was added 1,4-dioxa-8-azaspiro[4.5]decane (15, 2.34 kg, 16.36 mol, 1.14 equiv) with stirring over 20 minutes. Triethylamine (Et3N, 4 L, 28.67 mol, 2.00 equiv) was then added over 1 hour. The temperature was kept between 5° C. and 10° C. during the additions. The dark brown solution thus obtained was stirred for 12 hours at 20° C. and then chilled to 10° C. With vigorous stirring, 18 L of saturated sodium bicarbonate solution and 36 L of water were sequentially added and the temperature was kept under 15° C. The precipitation (filter cake) thus obtained was collected by filtration. The aqueous phase was then saturated with 12 kg of solid sodium chloride and extracted with EtOAc (2×18 L). The combined organic layer was washed with saturated sodium bicarbonate solution (18 L), and water (2×18 L) in sequence. The filter cake from the previous filtration was dissolved back in the organic phase. The dark brown solution thus obtained was washed twice with 18 L of water each and then concentrated under reduced pressure (40-50° C., 30 mm Hg) to give 5.0 kg of the crude product as viscous brown oil. The crude product 17 obtained above was dissolved in EtOH (8.15 L) at 50° C. Water (16.3 L) was added over 30 minutes. The brown solution was seeded, cooled to 20° C. over 3 hours with stirring and stirred at 20° C. for 12 h. The precipitate formed was filtered, washed with a mixture of EtOH and water (EtOH:H2O=1:20, 2 L) and dried under reduced pressure (50 mmHg) at 60° C. for 24 hours to afford (3-fluoro-2-(trifluoromethyl)pyridin-4-yl)(1,4-dioxa-8-azaspiro[4,5]decan-8-yl)methanone (17, 3.98 kg, 11.92 mol, 83.1%) as a white powder. For 17: 1H NMR (300 MHz, (CD3)2SO) δ 8.64 (d, 3JHH=4.68 Hz, 1H, NCH in pyridine), 7.92 (dd, 3JHH=4.68 Hz, 4JHF=4.68 Hz, 1H, NCCH in pyridine), 3.87-3.91 (m, 4H, OCH2CH2O), 3.70 (br s, 2H, one of NCH2 in piperidine rine, one of another NCH2 in piperidine ring, both in axial position), 3.26 (t, 3JHH=5.86 Hz, 2H, one of NCH2 in piperidine rine, one of another NCH2 in piperidine ring, both in equatorial position), 1.67 (d, 3JHH=5.86 Hz, 2H, one of NCCH2 in piperidine ring, one of another NCCH2 in piperidine ring, both in equatorial position), 1.58 (br s, 2H, one of NCCH2 in piperidine ring, one of another NCCH2 in piperidine ring, both in axial position) ppm; 13C NMR (75 MHz, (CD3)2SO) δ 161.03 (N—C═O), 151.16 (d, 1JCF=266.03 Hz, C—F), 146.85 (d, 4JCF=4.32 Hz, NCH in pyridine), 135.24 (d, 2JCF=11.51 Hz, C—C═O), 135.02 (quartet, 2JCF=34.57 Hz, NCCF3), 128.24 (d, 4JCF=7.48 Hz, NCCH in pyridine), 119.43 (d×quartet, 1JCF=274.38 Hz, 3JCF=4.89 Hz, CF3), 106.74 (OCO), 64.60 (OCCO), 45.34 (NC in piperidine ring), 39.62 (NC in piperidine ring), 34.79 (NCC in piperidine ring), 34.10 (NCC in piperidine ring) ppm; 19F NMR (282 MHz, (CD3)2SO) δ-64.69 (d, 4JFF=15.85 Hz, F3C), −129.26 (d×quartet, 4JFF=15.85 Hz, 4JFH=3.96 Hz, FC) ppm; C14H14F4N2O3 (MW, 334.27), LCMS (EI) m/e 335.1 (M++H).

Example 4(3-Fluoro-2-(trifluoromethyl)pyridin-4-yl) (1,4-dioxa-8-azaspiro[4,5]decan-8-yl)methanone (18)

In a 5 L 4-necked round bottom flask equipped with a mechanical stirrer, a thermocouple, an addition funnel and a nitrogen inlet was placed (3-fluoro-2-(trifluoromethyl)pyridin-4-yl)(1,4-dioxa-8-azaspiro[4,5]decan-8-yl)methanone (17, 100 g, 0.299 mol) in acetonitrile (ACN, 400 mL) at room temperature. The resultant solution was cooled to below 10° C. To the reaction mixture was added 6.0 N aqueous hydrochloric acid (HCl, 450 mL, 2.70 mol, 9.0 equiv), while the internal temperature was kept below 10° C. The resulting reaction mixture was then warmed to room temperature and an additional amount of 6.0 N aqueous hydrochloric acid (HCl, 1050 mL, 6.30 mol, 21.0 equiv) was slowly introduced to the reaction mixture at room temperature in 8 hours via the addition funnel. The reaction mixture was then cooled to 0° C. before being treated with 30% aqueous sodium hydroxide (NaOH, 860 mL, 8.57 mmol, 28.6 equiv) while the internal temperature was kept at below 10° C. The resulting reaction mixture was subsequently warmed to room temperature prior to addition of solid sodium bicarbonate (NaHCO3, 85.0 g, 1.01 mol, 3.37 equiv) in 1 hour. The mixture was then extracted with EtOAc (2×1.2 L), and the combined organic phase was washed with 16% aqueous sodium chloride solution (2×800 mL) and concentrated to approximately 1.0 L by vacuum distillation. Heptane (2.1 L) was added to the residue, and the resulting mixture was concentrated to 1.0 L by vacuum distillation. To the concentrated mixture was added heptane (2.1 L). The resulting white slurry was then concentrated to 1.0 L by vacuum distillation. To the white slurry was then added methyl tert-butyl ether (MTBE, 1.94 L). The white turbid was heated to 40° C. to obtain a clear solution. The resulting solution was concentrated to about 1.0 L by vacuum distillation. The mixture was stirred at room temperature for 1 hour. The white precipitate was collected by filtration with pulling vacuum. The filter cake was washed with heptane (400 mL) and dried on the filter under nitrogen with pulling vacuum to provide compound 18 (78.3 g, 90.1%) as an off-white solid. For 18: 1H NMR (300 MHz, (CD3)2SO) δ 8.68 (d, 3JHH=4.69 Hz, 1H, NCH in pyridine), 7.97 (dd, 3JHH=4.69 Hz, 4JHF=4.69 Hz, 1H, NCCH in pyridine), 3.92 (br s, 2H, one of NCH2 in piperidine rine, one of another NCH2 in piperidine ring, both in axial position), 3.54 (t, 3JHH=6.15 Hz, 2H, one of NCH2 in piperidine rine, one of another NCH2 in piperidine ring, both in equatorial position), 2.48 (t, 3JHH=6.44 Hz, 2H, NCCH2), 2.34 (t, 3JHE=6.15 Hz, 2H, NCCH2) ppm; 13C NMR (75 MHz, (CD3)2SO) δ 207.17 (C═O), 161.66 (N—C═O), 151.26 (d, 1JCF=266.89 Hz, C—F), 146.90 (d, 4JCF=6.05 Hz, NCH in pyridine), 135.56 (C—C═O), 134.78-135.56 (m, NCCF3), 128.27 (d, 3JCF=7.19 Hz, NCCH in pyridine), 119.52 (d×quartet, 1JCF=274.38 Hz, 3JCF=4.89 Hz, CF3), 45.10 (NC in piperidine ring) ppm, one carbon (NCC in piperidine ring) missing due to overlap with (CD3)2SO; 19F NMR (282 MHz, (CD3)2SO) δ-64.58 (d, 4JFF=15.85 Hz, F3C), −128.90 (d×quartet, 4JFF=15.85 Hz, 4JFH=4.05 Hz, FC) ppm; C12H10F4N2O2 (MW, 290.21), LCMS (EI) m/e 291.1 (M++H).

Example 53-[4-(7-{[2-(Trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile dihydrochloride (20)

Step 1. tent-Butyl 3-(cyanomethyl)-3-(4-(7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidine-1-carboxylate (19)

In a dried 30 L reactor equipped with a mechanic stirrer, a thermometer, an addition funnel and a vacuum outlet were placed 4-(1H-pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (5, 4.50 kg, 14.28 mol), tert-butyl 3-(cyanomethylene)azetidine-1-carboxylate (13, 3.12 kg, 16.08 mol, 1.126 equiv) in acetonitrile (9 L) at 20±5° C. To the resultant pink suspension was added 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU, 225 mL, 1.48 mol, 0.10 equiv) over 40 minutes. The batch temperature was kept between 10° C. and 20° C. during addition. The brown solution obtained was stirred at 20° C. for 3 hours. After the reaction was complete, water (18 L) was added with stirring over 80 minutes at 20° C. The mixture was seeded and the seeded mixture was stirred at room temperature for 12 hours. The solids were collected by filtration and the filter cake was washed with a mixture of acetonitrile and water (1:2, 9 L) and dried in a vacuum oven with nitrogen purge for 12 hours at 60° C. to provide the crude product (19, 7.34 kg) as a light yellow powder. The crude product obtained above was dissolved in methyl tert-butyl ether (MTBE, 22 L) at 60° C. in a 50 L reactor equipped with a mechanic stirrer, a thermometer, an addition funnel and a septum. Hexanes (22 L) was added over 1 hour at 60° C. The solution was then seeded, cooled to 20° C. over 3 hours and stirred at 20° C. for 12 hours. The precipitation was collected by filtration. The resultant cake was washed with a mixture of MTBE and hexane (1:15, 3 L) and dried in a vacuum oven for 10 hours at 50° C. to provide the compound 19 (6.83 kg, 13.42 mol, 94.0%) as a white powder. For 19: 1H NMR (400 MHz, CDCl3) δ 8.87 (s, 1H), 8.46 (d, J=0.6 Hz, 1H), 8.36 (d, J=0.7 Hz, 1H), 7.44 (d, J=3.7 Hz, 1H), 6.82 (d, J=3.7 Hz, 1H), 5.69 (s, 2H), 4.57 (d, J=9.6 Hz, 2H), 4.32 (d, J=9.5 Hz, 2H), 3.59-3.49 (m, 2H), 3.35 (s, 2H), 1.49 (s, 9H), 0.96-0.87 (m, 2H), −0.03-−0.10 (s, 9H) ppm; 13C NMR (101 MHz, CDCl3) δ 157.22, 153.67, 153.24, 151.62, 142.13, 130.16, 129.67, 124.47, 116.72, 115.79, 102.12, 82.54, 74.23, 68.01, 60.25, 58.23, 29.65, 29.52, 19.15, −0.26 ppm; C25H35N7O3Si (MW, 509.68), LCMS (EI) m/e 510.1 (M++H).

Step 2. 3-[4-(7-{[2-(Trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile dihydrochloride (20)

In a 2 L 4-necked round bottom flask equipped with a mechanical stirrer, a thermocouple, an addition funnel and a nitrogen inlet was added compound 19 (55.0 g, 0.108 mol) and methanol (MeOH, 440 mL) at 20±5° C. The resulting white turbid was stirred for 20 minutes at room temperature to provide a light yellow solution. A solution of hydrochloric acid (HCl) in isopropanol (5.25 M, 165 mL, 0.866 mol, 8.02 equiv) was then added to the reaction mixture via the addition funnel in 5 minutes. The resulting reaction mixture was then heated to 40° C. by a heating mantle. After 2 hours at 40° C., water (165 mL, 9.17 mol, 84.8 equiv) was added to the reaction mixture via the addition funnel to provide a light green solution at 40° C. Methyl tert-butyl ether (MTBE, 440 mL) was added to the resulting mixture via the addition funnel at 40° C. The resulting mixture was slowly cooled to 10° C. The solids were collected by filtration and washed with MTBE (2×220 mL). The white solids were dried in the filter under nitrogen with a pulling vacuum for 18 hours to afford compound 20 (52.2 g, KF water content 5.42%, yield 94.9%). For 20: 1H NMR (400 MHz, (CD3)2SO) δ 10.39 (brs, 1H), 10.16 (brs, 1H), 9.61 (s, 1H), 9.12 (s, 1H), 9.02 (s, 1H), 8.27-8.21 (d, J=3.8 Hz, 1H), 7.72-7.66 (d, J=3.8 Hz, 1H), 5.82 (s, 2H), 4.88-4.77 (m, 2H), 4.53-4.44 (m, 2H), 4.12 (s, 2H), 3.69-3.60 (m, 2H), 0.98-0.89 (m, 2H), 0.01 (s, 9H) ppm; 13C NMR (101 MHz, (CD3)2SO) δ 151.25, 146.45, 145.09, 140.75, 133.38, 132.44, 116.20, 116.09, 112.79, 102.88, 73.07, 66.14, 59.16, 53.69, 26.44, 17.15, −1.36 ppm; C20H29Cl2N7OSi (free base of 20, C20H27N7OSi, MW 409.56), LCMS (EI) m/e 410.2 (M++H).

Example 62-(1-(1-(3-Fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)-3-(4-(7-(2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (21)

In a 100 L dried reactor equipped with a mechanical stirrer, a thermocouple, a condenser, and a nitrogen inlet was added (20, 3.24 kg, 6.715 mol) and dichloromethane (32 L) at 20±5° C. The mixture was stirred at room temperature for 10 minutes before being treated with triethylamine (TEA, 1.36 kg, 13.44 mol, 2.00 equiv) at an addition rate which keeping the internal temperature at 15-30° C. Compound 18 (2.01 kg, 6.926 mol, 1.03 equiv) was then added to the reactor at room temperature. After 10 minutes, sodium triacetoxyborohydride (NaBH(OAc)3, 2.28 kg, 10.75 mol, 1.60 equiv) was added portion wise to the reactor in 1 hour while the internal temperature was kept at 15-30° C. The resulting reaction mixture was stirred at 15-30° C. for an additional one hour. Once the reductive amination reaction is deemed complete, the reaction mixture was treated with a 4% aqueous sodium bicarbonate solution (NaHCO3, 32 L) to adjust the pH to 7-8. After stirring for 30 minutes at room temperature, the two phases were separated. The aqueous phase was extracted with dichloromethane (29 L). The combined organic phase was sequentially washed with 0.1 N aqueous hydrochloric acid solution (16 L), 4% aqueous sodium bicarbonate solution (16 L), 8% aqueous sodium chloride solution (2×16 L). The resultant organic phase was partially concentrated and filtered. The filtrate was subjected to solvent exchange by gradually adding acetonitrile (65 L) under vacuum. The white solids were collected by filtration, washed with acetonitrile (10 L) and dried at 40-50° C. in a vacuum oven with nitrogen purge to afford compound 21 (4.26 kg, 6.23 mol, 92.9%). For 21: 1H NMR (500 MHz, (CD3)2SO) δ 8.84 (s, 1H), 8.76 (s, 1H), 8.66 (d, J=4.7 Hz, 1H), 8.43 (s, 1H), 7.90 (t, J=4.7 Hz, 1H), 7.78 (d, J=3.7 Hz, 1H), 7.17 (d, J=3.7 Hz, 1H), 5.63 (s, 2H), 4.07 (dt, J=11.1, 4.9 Hz, 1H), 3.75 (d, J=7.8 Hz, 2H), 3.57 (dd, J=10.2, 7.8 Hz, 2H), 3.55 (s, 2h), 3.52 (dd, J=8.5, 7.4 Hz, 2H), 3.41 (dq, J=13.3, 4.3 Hz, 1H), 3.26 (t, J=10.0 Hz, 1H), 3.07 (ddd, J=13.1, 9.4, 3.2 Hz, 1H), 2.56 (dt, J=8.5, 4.7 Hz, 1H), 1.81-1.73 (m, 1H), 1.63 (m, 1H), 1.29 (m, 1H), 1.21 (m, 1H), 0.82 (dd, J=8.5, 7.4 Hz, 2H), −0.12 (s, 9H) ppm; 13C NMR (101 MHz, (CD3)2SO) δ 161.68, (154.91, 152.27), 153.08, 152.69, 151.53, 147.69, 140.96, (136.19, 136.02), (136.48, 136.36, 136.13, 136.0, 135.78, 135.66, 135.43, 135.32), 131.43, 130.84, 129.03, (126.17, 123.42, 120.69), 117.99, 122.77, 118.78, 114.71, 102.02, 73.73, 67.04, 62.86, 61.88, 58.51, 45.63, 30.03, 29.30, 28.60, 18.52, 0.00 ppm; C32H37F4N9O2Si (MW, 683.77), LCMS (EI) m/e 684.2 (M++H).

Example 72-(3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(1-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile (22)

Figure US20130060026A1-20130307-C00025 BASE OF INCB 39110

To a 250 mL 4-necked round bottom flask equipped with a mechanical stirrer, a thermocouple, an addition funnel and a nitrogen inlet was added compound 21 (9.25 g, 13.52 mmol, KF water content 3.50%) and acetonitrile (74 mL) at 20±5° C. The resulting white slurry was cooled to below 5° C. Boron trifluoride diethyl etherate (BF3.OEt2, 6.46 mL, 51.37 mmol, 3.80 equiv) was then added at a rate while the internal temperature was kept at below 5.0° C. The reaction mixture was then warmed to 20±5° C. After stirring at 20±5° C. for 18 hours, the reaction mixture was cooled to 0-5° C. and an additional amount of BF3.OEt2 (0.34 mL, 2.70 mmol, 0.2 equiv) was introduced to the reaction mixture at below 5.0° C. The resulting reaction mixture was warmed to 20±5° C., and kept stirring at room temperature for an additional 5 hours. The reaction mixture was then cooled to 0-5° C. before water (12.17 mL, 0.676 mol, 50 equiv) was added. The internal temperature was kept at below 5.0° C. during addition of water. The resultant mixture was warmed to 20±5° C. and kept stirring at room temperature for 2 hours. The reaction mixture was then cooled to 0-5° C. and aqueous ammonium hydroxide (NH4OH, 5 N, 121.7 mmol, 9.0 equiv) was added. During addition of aqueous ammonium hydroxide solution, the internal temperature was kept at below 5.0° C. The resulting reaction mixture was warmed to 20±5° C. and stirred at room temperature for 20 hours. Once the SEM-deprotection was deemed complete, the reaction mixture was filtered, and the solids were washed with EtOAc (9.25 mL). The filtrates were combined and diluted with EtOAc (74 mL). The diluted organic solution was washed with 13% aqueous sodium chloride solution (46.2 mL). The organic phase was then diluted with EtOAc (55.5 mL) before being concentrated to a minimum volume under reduced pressure. EtOAc (120 mL) was added to the residue, and the resulting solution was stirred at 20±5° C. for 30 minutes. The solution was then washed with 7% aqueous sodium bicarbonate solution (2×46 mL) and 13% aqueous sodium bicarbonate solution (46 mL). The resultant organic phase was diluted with EtOAc (46 mL) and treated with water (64 mL) at 50±5° C. for 30 minutes. The mixture was cooled to 20±5° C. and the two phases were separated. The organic phase was treated with water (64 mL) at 50±5° C. for 30 minutes for the second time. The mixture was cooled to 20±5° C. and the two phases were separated. The resultant organic phase was concentrated to afford crude compound 22 (free base), which was further purified by column chromatography (SiO2, 330 g, gradient elution with 0-10% of MeOH in EtOAc) to afford analytically pure free base (22, 7.00 g, 93.5%) as an off-white solid. For 22:

 

1H NMR (400 MHz, (CD3)2SO) δ 12.17 (d, J=2.8 Hz, 1H), 8.85 (s, 1H), 8.70 (m, 2H), 8.45 (s, 1H), 7.93 (t, J=4.7 Hz, 1H), 7.63 (dd, J=3.6, 2.3 Hz, 1H), 7.09 (dd, J=3.6, 1.7 Hz, 1H), 4.10 (m, 1H), 3.78 (d, J=7.9 Hz, 2H), 3.61 (t, J=7.9 Hz, 1H), 3.58 (s, 2H), 3.46 (m, 1H), 3.28 (t, J=10.5 Hz, 1H), 3.09 (ddd, J=13.2, 9.5, 3.1 Hz, 1H), 2.58 (m, 1H), 1.83-1.75 (m, 1H), 1.70-1.63 (m, 1H), 1.35-1.21 (m, 2H) ppm;

13C NMR (101 MHz, (CD3)2SO) δ 160.28, (153.51, 150.86), 152.20, 150.94, 149.62, (146.30, 146.25), 139.48, (134.78, 134.61), (135.04, 134.92, 134.72, 134.60, 134.38, 134.26, 134.03, 133.92), 129.22, 127.62, 126.84, 121.99, 122.04, (124.77, 122.02, 119.19, 116.52), 117.39, 113.00, 99.99, 61.47, 60.49, 57.05, 44.23, 28.62, 27.88, 27.19 ppm;

C26H23F4N9O (MW, 553.51), LCMS (EI) m/e 554.1 (M′+H).

ADIPATE

Example 8

2-(3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(1-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipate (25)

Step 1. 2-(3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(1-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipate crude salt (24)

The process of making compound 22 in Example 7 was followed, except that the final organic phase was concentrated by vacuum distillation to the minimum volume to afford crude compound 22, which was not isolated but was directly used in subsequent adipate salt formation process. To the concentrated residue which containing crude compound 22 was added methanol (200 mL) at room temperature. The mixture was the concentrated by vacuum distillation to a minimum volume. The residue was then added methanol (75 mL) and the resulting solution was heated to reflux for 2 hours. Methyl isobutyl ketone (MIBK, 75 mL) was added to the solution and the resulting mixture was distilled under vacuum to about 30 mL while the internal temperature was kept at 40-50° C. Methanol (75 mL) was added and the resulting mixture was heated to reflux for 2 hours. To the solution was added MIBK (75 mL). The mixture was distilled again under vacuum to about 30 mL while the internal temperature was kept at 40-50° C. To the solution was added a solution of adipic acid (23, 2.15 g, 14.77 mmol) in methanol (75 mL). The resultant solution was then heated to reflux for 2 hours. MIBK (75 mL) was added. The mixture was distilled under vacuum to about 60 mL while the internal temperature was kept at 40-50° C. Heating was stopped and heptane (52.5 mL) was added over 1-2 hours. The resultant mixture was stirred at 20±5° C. for 3-4 hours. The white precipitates were collected by filtration, and the filter cake was washed with heptane (2×15 mL). The solid was dried on the filter under nitrogen with a pulling vacuum at 20±5° C. for 12 hours to provide compound 24 (crude adipate salt, 8.98 g, 12.84 mmol., 95.0%). For 24: 1H NMR (400 MHz, (CD3)2SO) δ 12.16 (s, 1H), 12.05 (brs, 2H), 8.85 (s, 1H), 8.72 (s, 1H), 8.69 (d, J=4.7 Hz, 1H), 8.45 (s, 1H), 7.93 (t, J=4.7 Hz, 1H), 7.63 (dd, J=3.6, 2.3 Hz, 1H), 7.09 (dd, J=3.6, 1.7 Hz, 1H), δ 4.11 (dt, J=11.0, 4.4 Hz, 1H), 3.77 (d, J=7.8 Hz, 2H), 3.60 (t, J=7.8 Hz, 2H), 3.58 (s, 2H), 3.44 (dt, J=14.4, 4.6 Hz, 1H), 3.28 (t, J=10.4 Hz, 1H), 3.09 (ddd, J=13.2, 9.6, 3.2 Hz, 1H), 2.58 (tt, J=8.6, 3.5 Hz, 1H), 2.28-2.17 (m, 4H), 1.83-1.74 (m, 1H), 1.67 (d, J=11.0 Hz, 1H), 1.59-1.46 (m, 4H), 1.37-1.21 (m, 2H) ppm; 13C NMR (101 MHz, (CD3)2SO) δ 174.38, 160.29, (153.52, 150.87), 152.20, 150.94, 149.63, (146.30, 146.25), 139.48, (134.79, 134.62), (135.08, 134.97, 134.74, 134.62, 134.38, 134.28, 134.04, 133.93), 129.21, 127.62, 126.84, 122.05, (124.75, 122.02, 119.29, 116.54), 117.39, 113.01, 99.99, 61.47, 60.50, 57.06, 44.24, 33.42, 30.70, 28.63, 27.89, 27.20, 24.07 ppm; C32H33F4N9O5 (Mol. Wt: 699.66; 24: C26H23F4N9O, MW 553.51), LCMS (EI) m/e 554.0 (M++H).

Step 2.

2-(3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(1-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipate (25)

In a 100 L dried reactor equipped with a mechanical stirrer, a thermocouple, an addition funnel and a nitrogen inlet was added compound 24 (3.40 kg, 4.86 mol) and acetone (23.8 L). The resulting white turbid was heated to 55-60° C. to provide a clear solution. The resultant solution was filtered through an in-line filter to another 100 L reactor. Heptane (23.8 L) was filtered through an in-line filter to a separated 50 L reactor. The filtered heptane was then charged to the acetone solution in the 100 L reactor at a rate while the internal temperature was kept at 55-60° C. The reaction mixture in the 100 L reactor was then cooled to 20±5° C. and stirred at 20±5° C. for 16 hours. The white precipitates were collected by filtration and the cake was washed with heptane (2×5.1 L) and dried on the filter under nitrogen with a pulling vacuum. The solid was further dried in a vacuum oven at 55-65° C. with nitrogen purge to provide compound 25 (3.11 kg, 92.2%) as white to off-white powder. For 25:

ADIPATE OF INCB 39110

1H NMR (400 MHz, (CD3)2SO) δ 12.16 (s, 1H), 12.05 (brs, 2H), 8.85 (s, 1H), 8.72 (s, 1H), 8.69 (d, J=4.7 Hz, 1H), 8.45 (s, 1H), 7.93 (t, J=4.7 Hz, 1H), 7.63 (dd, J=3.6, 2.3 Hz, 1H), 7.09 (dd, J=3.6, 1.7 Hz, 1H), δ 4.11 (dt, J=11.0, 4.4 Hz, 1H), 3.77 (d, J=7.8 Hz, 2H), 3.60 (t, J=7.8 Hz, 2H), 3.58 (s, 2H), 3.44 (dt, J=14.4, 4.6 Hz, 1H), 3.28 (t, J=10.4 Hz, 1H), 3.09 (ddd, J=13.2, 9.6, 3.2 Hz, 1H), 2.58 (tt, J=8.6, 3.5 Hz, 1H), 2.28-2.17 (m, 4H), 1.83-1.74 (m, 1H), 1.67 (d, J=11.0 Hz, 1H), 1.59-1.46 (m, 4H), 1.37-1.21 (m, 2H) ppm;

 

13C NMR (101 MHz, (CD3)2SO) δ 174.38, 160.29, (153.52, 150.87), 152.20, 150.94, 149.63, (146.30, 146.25), 139.48, (134.79, 134.62), (135.08, 134.97, 134.74, 134.62, 134.38, 134.28, 134.04, 133.93), 129.21, 127.62, 126.84, 122.05, (124.75, 122.02, 119.29, 116.54), 117.39, 113.01, 99.99, 61.47, 60.50, 57.06, 44.24, 33.42, 30.70, 28.63, 27.89, 27.20, 24.07 ppm;

 

C32H33F4N9O5 (Mol. Wt: 699.66; free base: C26H23F4N9O (MW, 553.51), LCMS (EI) m/e 554.0 (M++H).

 

…………………………

WO-2014138168

 http://www.google.com/patents/WO2014138168A1?cl=en

Processes for preparing JAK inhibitor (preferably INCB-39110) comprising the reaction of a substituted 1H-pyrazole compound with 4-chloro-7H-pyrrolo[2,3-d]pyrimidine in the presence of a base (eg cesium fluoride) and a solvent under Suzuki coupling conditions ([1,1′- bis(dicyclohexylphosphino)ferrocene]dichloropalladium (II)), followed by deprotection and then reaction with a piperidine derivative, and salt synthesis are claimed. Also claimed are novel intermediates and processes for their preparation. The compound is disclosed to be useful for treating disease mediated by JAK activity (targeting JAK-1 and 2), such as multiple sclerosis, rheumatoid arthritis, type I diabetes, inflammatory bowel disease, Crohn’s disease, COPD, prostate cancer, hepatic cancer, breast cancer, influenza, and SARS.

Example 1. Synthesis of 2-(3-(4-(7H-Pyrrolo[2,3-< ]pyrimidin-4-yl)-lH-pyrazol-l- yl)-l-(l-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3- yl)acetonitrile Adipate (9)20443-0253WO1 (INCY0124-WO1) PATENT

tert-Butyl 3-(cyanomethyl)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazol-l-yl)azetidine-l-carboxylate (3). To a 1-L flask equipped with a nitrogen inlet, a thermocouple, and a mechanical stirrer were sequentially added isopropanol (IP A, 200 mL), l,8-diazabicyclo[5,4,0]undec-ene (DBU, 9.8 g, 64.4 mmol, 0.125 equiv), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (1, 101 g, 520.51 mmol, 1.01 equiv) and tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (2, 100 g, 514.85 mmol) at ambient temperature to generate a reaction mixture as a

suspension. The resulting reaction mixture was heated to reflux in 30 minutes to provide a homogenous solution and the mixture was maintained at reflux for an additional 2 – 3 hours. After the reaction was complete as monitored by HPLC, n- heptane (400 mL) was gradually added to the reaction mixture in 45 minutes while maintaining the mixture at reflux. Solids were precipitated out during the w-heptane addition. Once w-heptane addition was complete, the mixture was gradually cooled to ambient temperature and stirred at ambient temperature for an additional 1 hour. The solids were collected by filtration, washed with w-heptane (200 mL), and dried under vacuum at 50 °C with nitrogen sweeping to constant weight to afford tert-butyl 3- 20443-0253WO1 (INCY0124-WO1) PATENT

(cyanomethyl)-3-(4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)- IH-pyrazol- 1 – yl)azetidine-l -carboxylate (3, 181 g, 199.9 g theoretical, 90.5%) as a white to pale yellow solid. For 3: XH NMR (400 MHz, DMSO-i¾) δ 8.31 (s, 1H), 7.74 (s, 1H), 4.45 – 4.23 (m, 2H), 4.23 – 4.03 (m, 2H), 3.56 (s, 2H), 1.38 (s, 9H), 1.25 (s, 12H) ppm; 13C NMR (101 MHz, DMSO-i/6) δ 155.34, 145.50, 135.88, 1 16.88, 107.08 (br), 83.15, 79.36, 58.74 (br), 56.28, 27.96, 26.59, 24.63 ppm; Ci9H29B 404 (MW 388.27),

LCMS (EI) mle 389 (M+ + H). teri-Butyl 3-(4-(7H-pyrrolo[2,3-< |pyrimidin-4-yl)-lH-pyrazol-l-yl)-3- (cyanomethyl)-azetidine-l-carboxylate (5). To a 1-L flask equipped with a nitrogen inlet, a thermocouple, and a mechanical stirrer were added 4-chloro-7H-pyrrolo[2,3- i/]pyrimidine (4, 39.6 g, 257.6 mmol), tert-butyl 3-(cyanomethyl)-3-(4-(4,4,5,5- tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- IH-pyrazol- 1 -yl)azetidine- 1 -carboxylate (3, 100 g, 257.6 mmol, 1.0 equiv), cesium fluoride (136.9 g, 901.4 mmol, 3.5 equiv), tert- butanol (250 mL), water (250 mL), and [l, l’-bis(di- cyclohexylphosphino)ferrocene]dichloropalladium(II) (Pd-127, 351.4 mg, 0.46 mmol, 0.0018 equiv) at ambient temperature. The resulting reaction mixture was de-gassed and refilled with nitrogen for 3 times before being heated to reflux and maintained at reflux under nitrogen for 20 – 24 hours. When HPLC showed the reaction was complete, the reaction mixture was cooled to 45 – 55 °C in 30 minutes, the two phases were separated, and the aqueous phase was discarded. To the organic phase was added w-heptane (125 mL) in 30 minutes at 45 – 55 °C. The resulting mixture was slowly cooled to ambient temperature in one hour and stirred at ambient temperature for an additional 2 hours. The solids were collected by filtration, washed with n- heptane (100 mL), and dried under vacuum at 50 °C with nitrogen sweeping to constant weight to afford tert-butyl 3-(4-(7H-pyrrolo[2,3-<i]pyrimidin-4-yl)-lH- pyrazol-l-yl)-3-(cyanomethyl)-azetidine-l -carboxylate (5, 96.8 g, 97.7 g theoretical, 99%) as a pale yellow solid. For 5: XH NMR (400 MHz, DMSO-i¾) δ 8.89 (s, 1H), 8.68 (s, 1H), 8.44 (s, 1H), 7.60 (d, J= 3.5 Hz, 1H), 7.06 (d, J= 3.6 Hz, 1H), 4.62 – 4.41 (m, 2H), 4.31 – 4.12 (m, 2H), 3.67 (s, 2H), 1.39 (s, 9H) ppm; 13C NMR (101 MHz, DMSO-i¾) δ 155.40, 152.60, 150.63, 149.15, 139.76, 129.53, 127.65, 122.25, 20443-0253WO1 (INCY0124-WO1) PATENT

116.92, 113.21, 99.71, 79.45, 58.34 (br), 56.80, 27.99, 26.83 ppm; Ci9H21 702 (MW 379.4), LCMS (EI) mle 380 (M+ + H).

2- (3-(4-(7H-Pyrrolo[2,3-< |pyrimidin-4-yl)-lH-pyrazol-l-yl)azetidin-3- yl)acetonitrile dihydrochloride salt (6). To a 0.5-L flask equipped with a nitrogen inlet, a thermocouple, an additional funnel, and a mechanical stirrer were added tert- butyl 3 -(4-(7H-pyrrolo [2,3 -<i]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)-3 – (cyanomethyl)azetidine-l-carboxylate (5, 15 g, 39.5 mmol), water (7.5 mL, 416 mmol) and dichloromethane (75 mL) at room temperature. The mixture was stirred at room temperature to generate a suspension. To the suspension was added a solution of 5 M hydrogen chloride (HQ) in isopropanol (55 mL, 275 mmol, 7.0 equiv) in 5 minutes. The resulting reaction mixture was then heated to gentle reflux and

maitained at reflux for 3-4 hours. After the reaction was completed as mornitored by HPLC, tert-butyl methyl ether (TBME, 45 mL) was added to the reaction suspension. The mixture was gradually cooled to room temperature, and stirred for an additional one hour. The solids were collected by filtration, washed with tert-butyl methyl ether (TBME, 45 mL) and dried under vacuum at 50 °C with nitrogen sweeping to constant weight to afford 2-(3-(4-(7H-pyrrolo[2,3-i/]pyrimidin-4-yl)-lH-pyrazol-l-yl)azetidin-

3- yl)acetonitrile dihydrochloride salt (6, 13.6 g, 13.9 g theoretical, 98%) as an off- white to light yellow solid. For 6: XH NMR (400 MHz, D20) δ 8.96 (s, 1H), 8.81 (s, 1H), 8.49 (s, 1H), 7.78 (d, J= 3.8 Hz, 1H), 7.09 (d, J= 3.7 Hz, 1H), 4.93 (d, J= 12.8 Hz, 2H), 4.74 (d, J= 12.5 Hz, 2H), 3.74 (s, 2H) ppm; 13C NMR (101 MHz, D20) δ 151.35, 143.75, 143.33, 141.33, 132.03, 131.97, 115.90, 114.54, 113.85, 103.18, 59.72, 54.45 (2C), 27.02 ppm; Ci4H15Cl2N7 (Ci4H13N7 for free base, MW 279.30), LCMS (EI) mle 280 (M+ + H).

2-(3-(4-(7H-Pyrrolo[2,3-< |pyrimidin-4-yl)-lH-pyrazol-l-yl)-l-(l-(3-fluoro-2- (trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile (8, Free Base). To a 0.5-L flask equipped with a nitrogen inlet, a thermocouple, an additional funnel, and a mechanical stirrer were added 2-(3-(4-(7H-pyrrolo[2,3-<i]pyrimidin-4- yl)-lH-pyrazol-l-yl)azetidin-3-yl)acetonitrile dihydrochloride salt (6, 20 g, 56.78 mmol), dichloromethane (200 mL) and triethylamine (TEA, 16.62 mL, 119.2 mmol, 20443-0253WO1 (INCY0124-WO1) PATENT

2.1 equiv) at ambient temperature. The mixture was stired at ambient temperature for 30 minutes before l-(3-fluoro-2-(trifluoromethyl)-isonicotinoyl)piperidin-4-one (7, 17.15 g, 57.91 mmol, 1.02 equiv) was added to the mixture. The mixture was then treated with sodium triacetoxyborohydride (25.34 g, 1 13.6 mmol, 2.0 equiv) in 5 minutes at ambient temperature (below 26 °C). The resulting reaction mixture was stirred at ambient temperature for 2 hours. After the reaction was complete as mornitored by HPLC, the reaction mixture was quenched with saturated aHC03 aqueous solution (200 mL). The two phases were separated and the aqueous phase was extracted with methylene chloride (200 mL). The combined organic phase was washed with 4% brine (100 mL) followed by solvent switch of methylene chloride to acetone by distillation. The resulting solution of the desired crude product (8) in acetone was directly used for the subsequent adipate salt formation. A small portion of solution was purified by column chromatography (S1O2, 0 – 10% of MeOH in EtOAc gradient elution) to afford the analytically pure 2-(3-(4-(7H-pyrrolo[2,3- i/]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)- 1 -( 1 -(3 -fluoro-2-

(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile (8 free base) as an off-white solid. For 8: ¾ NMR (400 MHz, DMSO-i¾) δ 12.17 (d, J= 2.8 Hz, 1H), 8.85 (s, 1H), 8.70 (m, 2H), 8.45 (s, 1H), 7.93 (t, J= A J Hz, 1H), 7.63 (dd, J= 3.6, 2.3 Hz, 1H), 7.09 (dd, J= 3.6, 1.7 Hz, 1H), 4.10 (m, 1H), 3.78 (d, J= 7.9 Hz, 2H), 3.61 (t, J= 7.9 Hz, 1H), 3.58 (s, 2H), 3.46 (m, 1H), 3.28 (t, J= 10.5 Hz, 1H), 3.09 (ddd, J = 13.2, 9.5, 3.1 Hz, 1H), 2.58 (m, 1H), 1.83 – 1.75 (m, 1H), 1.70 – 1.63 (m, 1H), 1.35 – 1.21 (m, 2H) ppm; 13C MR (101 MHz, DMSO-i/6) δ 160.28, (153.51, 150.86), 152.20, 150.94, 149.62, (146.30, 146.25), 139.48, (134.78, 134.61), (135.04, 134.92, 134.72, 134.60, 134.38, 134.26, 134.03, 133.92), 129.22, 127.62, 126.84, 121.99, 122.04, (124.77, 122.02, 1 19.19, 1 16.52), 117.39, 113.00, 99.99, 61.47, 60.49, 57.05, 44.23, 28.62, 27.88, 27.19 ppm;

(MW, 553.51), LCMS (EI) mle 554.1 (M+ + H).

2-(3-(4-(7H-Pyrrolo[2,3-< |pyrimidin-4-yl)-lH-pyrazol-l-yl)-l-(l-(3-fluoro-2- (trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile Adipate (9). To a 0.5-L flask equipped with a mechanical stirrer, a thermocouple, an addition funnel, and a nitrogen inlet was added a solution of crude 2-(3-(4-(7H-pyrrolo[2,3- 20443-0253WO1 (INCY0124-WO1) PATENT i/]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)- 1 -( 1 -(3 -fluoro-2-

(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile (8 free base, 31.38 g, 56.7 mmol) in acetone (220 mL) and adipic acid (8.7 g, 59.53 mmol, 1.05 equiv) at ambient temperature. The reaction mixture was then heated to reflux to give a solution. w-Heptane (220 mL) was gradually added to the reaction mixture at 40 – 50 °C in one hour. The resulting mixture was gradually cooled to ambient temperature in one hour and stirred at ambient temperature for an additional 16 hours. The solids were collected by filtration, washed with w-heptane (2 X 60 mL), and dried under vacuum at 50 °C with nitrogen sweeping to constant weight to afford 2-(3-(4-(7H- Pyrrolo[2,3 -i/]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)- 1 -(1 -(3 -fluoro-2- (trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipate (9,34.0 g, 39.7 g theoretical, 85.6% for two steps) as a white to off-white solid. 9:

XH NMR (400 MHz, DMSO-i/6) δ 12.16 (s, 1H), 12.05 (brs, 2H), 8.85 (s, 1H), 8.72 (s, 1H), 8.69 (d, J= A J Hz, 1H), 8.45 (s, 1H), 7.93 (t, J= A J Hz, 1H), 7.63 (dd, J= 3.6, 2.3 Hz, 1H), 7.09 (dd, J= 3.6, 1.7 Hz, 1H), 5 4.1 1 (dt, J= 1 1.0, 4.4 Hz, 1H), 3.77 (d, J= 7.8 Hz, 2H), 3.60 (t, J= 7.8 Hz, 2H), 3.58 (s, 2H), 3.44 (dt, J= 14.4, 4.6 Hz, 1H), 3.28 (t, J= 10.4 Hz, 1H), 3.09 (ddd, J= 13.2, 9.6, 3.2 Hz, 1H), 2.58 (tt, J= 8.6, 3.5 Hz, lH), 2.28 – 2.17 (m, 4H), 1.83 – 1.74 (m, 1H), 1.67 (d, J= 11.0 Hz, 1H), 1.59 – 1.46 (m, 4H), 1.37 – 1.21 (m, 2H) ppm;

 

13C MR (101 MHz, DMSO-i/6) δ 174.38, 160.29, (153.52, 150.87), 152.20, 150.94, 149.63, (146.30, 146.25), 139.48, (134.79, 134.62), (135.08, 134.97, 134.74, 134.62, 134.38, 134.28, 134.04, 133.93), 129.21, 127.62, 126.84, 122.05, (124.75, 122.02, 1 19.29, 1 16.54), 117.39, 113.01, 99.99, 61.47, 60.50, 57.06, 44.24, 33.42, 30.70, 28.63, 27.89, 27.20, 24.07 ppm;

C32H33F4N9O5 ( MW 699.66;Figure imgf000043_0001 for free base, MW, 553.51), LCMS (EI) mle 554.0 (M+ + H).

 

 

Example 2: Alternative Synthesis of 2-(3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl)-l-(l-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4- yl)azetidin-3-yl)acetonitrile 20443-0253WO1 (INCY0124-WO1) PATENT

Scheme II

………………………………..COMPD11……………………………………………………………………………………………………..COMPD  8 BASE

C26H3i BF4N603 C26H23F4N9O Mol. Wt: 562.37 Mol. Wt: 553.51

2- (Azetidin-3-ylidene)acetonitrile hydrochloride (2a). To a 0.5-L flask equipped with a nitrogen inlet, a thermocouple, and a mechanical stirrer were added tert-butyl

3- (cyanomethylene)azetidine-l-carboxylate (2, 30 g, 154.46 mmol) and

methylenechloride (300 mL) at ambient temperature. The solution was then treated with a solution of 5 M hydrogen chloride (HQ) in isopropanol solution (294.2 mL, 1.54 mol, 10 equiv) at ambient temperature and the resulting reaction mixture was stirred at ambient temperature for 18 hours. After the reaction was complete as monitored by HPLC, the suspension was added tert-butyl methyl ether (TBME, 150 mL), and the mixture was stirred at ambient temperature for 2 hours. The solids was collected by filtration, washed with w-heptane (2 X 100 mL), and dried on the filtration funnel at ambient temperature for 3 hours to afford 2-(azetidin-3- ylidene)acetonitrile hydrochloride (2a, 13.7 g, 20.2 g theoretical, 67.8 %) as a white solid. For 2a: XH NMR (500 MHz, DMSO-i¾) δ 9.99 (s, 2H), 5.94 (p, J= 2.5 Hz, 1H), 20443-0253WO1 (INCY0124-WO1) PATENT

4.85 – 4.80 (m, 2H), 4.77 – 4.71 (m, 2H) ppm; C NMR (126 MHz, DMSO-i¾) δ 155.65, 114.54, 94.78, 55.26, 54.63 ppm; C5H7C1N2 (MW 130.58; C5H6N2 for free base, MW 94.11), LCMS (EI) mle 95 (M+ + H).

2-(l-(l-(3-Fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3- ylidene)acetonitrile (10). To a 0.25-L flask equipped with a nitrogen inlet, a thermocouple, and a magnetic stirrer were added 2-(azetidin-3-ylidene)acetonitrile hydrochloride (2a, 4.5 g, 34.46 mmol), l-(3-fluoro-2-

(trifluoromethyl)isonicotinoyl)piperidin-4-one (7, 10 g, 34.46 mmol, 1.0 equiv), and methylenechloride (100 mL) at ambient temperqature and the resulting mixture was then treated with sodium triacetoxyborohydride (14.6 g, 68.93 mmol, 2.0 equiv) at ambient temperature. The reaction mixture was stirred at ambient temperature for 2 hours before being quenched with saturated sodium bicarbonate (NaHCOs) aqueous solution (50 mL). The two phases were separated and the aqueous phase was extracted with dichloromethane (200 mL). The combined organic phase was washed with water (50 mL) and brine (50 mL) and concentrated under reduced pressure to afford the crude desired product (10), which was purified by column chromatography (S1O2, 0 – 10 % of ethyl acetate in hexane gradient elution) to afford 2-(l-(l-(3- fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-ylidene)acetonitrile (10, 9.5 g, 12.7 g theoretical, 74.8 %) as a white solid. For 10: XH NMR (400 MHz, CDCI3) δ 8.57 (d, J= A J Hz, 1H), 7.54 (t, J= 4.6 Hz, 1H), 5.29 (p, J= 2.4 Hz, 1H), 4.18 – 4.08 (m, 1H), 4.08 – 4.03 (m, 2H), 3.98 – 3.94 (m, 2H), 3.57 – 3.39 (m, 2H), 3.17 – 3.04 (m, 1H), 2.56 (tt, J= 7.4, 3.5 Hz, 1H), 1.86 – 1.77 (m, 1H), 1.75 – 1.64 (m, 1H), 1.54 – 1.43 (m, 1H), 1.43 – 1.31 (m, lH) ppm; 13C MR (101 MHz, CDC13) δ 161.34, 160.73, 152.62 (d, J= 269.1 Hz), 145.75 (d, J= 6.1 Hz), 136.73 (qd, J = 36.1, 12.0 Hz), 134.56 (d, J= 16.9 Hz), 126.89, 120.58 (qd, J= 275.0, 4.9 Hz),

115.11, 92.04, 62.05, 60.57 (2C), 44.47, 39.42, 29.38, 28.47 ppm; Ci7H16F4N40 (MW 368.33), LCMS (EI) mle 369 (M++ H).

2-(l-(l-(3-Fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)-3-(4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazol-l-yl)azetidin-3-yl)acetonitrile (11). To a 25 mL flask equipped with a nitrogen inlet, a thermocouple, and a magnetic 20443-0253WO1 (INCY0124-WO1) PATENT stirrer were added 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (1, 210 mg, 1.08 mmol, 1.08 equiv), 2-(l-(l-(3-fluoro-2-

(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3 -ylidene)acetonitrile (10, 370 mg, 1.0 mmol) and acetonitrile (3 mL) at ambient temperature. The solution was then treated with l,8-diazabicyclo[5,4,0]undec-ene (DBU, 173 mg, 0.17 mL, 1.12 mmol, 1.12 equiv) at ambient temperature and the resulting reaction mixture was warmed to 50 °C and stirred at 50 °C for overnight. When the reaction was complete as

monitored by HPLC, the reaction mixture was directly load on a solica gel (S1O2) column for chromatographic purification (0 – 2.5 % MeOH in ethyl acetate gradient elution) to afford 2-(l-(l-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)-3- (4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazol-l-yl)azetidin-3- yl)acetonitrile

Figure imgf000010_0003COMPD 11

(11, 263 mg, 562.4 mg theoretical, 46.7 %) as a white solid.

For 11: ΧΗ NMR (400 MHz, DMSO-i/6) δ 8.64 (d, J= 4.7 Hz, 1H), 8.22 (d, J= 0.6 Hz, 1H), 7.88 (dd, J= A J Hz, 1H), 7.69 (s, 1H), 4.10 – 3.99 (m, 1H), 3.58 (d, J= 7.8 Hz, 2H), 3.52 – 3.42 (m, 2H), 3.44 (s, 2H), 3.41 – 3.33 (m, 1H), 3.28 – 3.15 (m, 1H), 3.03 (ddd, J= 12.9, 9.2, 3.2 Hz, 1H), 2.51 – 2.44 (m, 1H), 1.77 – 1.66 (m, 1H), 1.64 – 1.54 (m, 1H), 1.28 – 1.17 (m, 2H), 1.24 (s, 12H) ppm;

 

13C MR (101 MHz, DMSO-i/6) δ 160.22, 152.13 (d, J= 265.8 Hz), 146.23 (d, J= 5.7 Hz), 145.12, 135.41, 134.66 (d, J= 16.9 Hz), 134.43 (qd, J= 35.0, 1 1.7 Hz), 127.58, 120.61 (qd, J= 274.4, 4.6 Hz), 117.35, 106.59 (br), 83.10, 61.40, 60.53 (2C), 56.49, 44.17, 38.99, 28.55, 27.82, 27.02, 24.63 ppm; C26H3iBF4 603 (MW 562.37), LCMS (EI) mle 563 (M+ + H).

 

2-(3-(4-(7H-Pyrrolo[2,3-< |pyrimidin-4-yl)-lH-pyrazol-l-yl)-l-(l-(3-fluoro-2- (trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile (8). To a

25-mL flask equipped with a nitrogen inlet, a thermocouple, an additional funnel, and a magnetic stirrer were added 2-(l-(l-(3-fluoro-2-(trifluoromethyl)- isonicotinoyl)piperidin-4-yl)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazol-l-yl)azetidin-3-yl)acetonitrile (11, 307 mg, 0.546 mmol), 4-chloro-7H- pyrrolo[2,3-if|pyrimidine (4, 84.8 mg, 0.548 mmol, 1.0 equiv), sodium bicarbonate (NaHC03, 229 mg, 2.72 mmol, 5.0 equiv), water (1.6 mL), and 1,4-dioxane (1.6 mL) at ambient temperature. The mixture was then teated with

tetrakis(triphenylphosphine)palladium(0) (12.8 mg, 0.011 mmol, 0.02 equiv) at 20443-0253WO1 (INCY0124-WO1) PATENT ambient temperature and the resulting reaction mixture was de-gassed and refilled with nitrogen for 3 times before being heated to 85 °C. The reaction mixture was stired at 85 °C under nitrogen for overnight. When the reaction was complete as monitored by HPLC, the reaction mixture was concentrated to dryness under reduced pressure and the desired product, 2-(3-(4-(7H-pyrrolo[2,3-( Jpyrimidin-4-yl)-lH- pyrazol- 1 -yl)- 1 -( 1 -(3 -fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin- 3-yl)acetonitrile (8 free base, 135 mg, 302.2 mg theoretical, 44.6 %), was obtained as off- white solids by direct silica gel (S1O2) cloumn chromatography (0 – 10% of ethyl acetate in hexane gradient elution) purification of the dried reaction mixture. The compound obtained by this synthetic approach is identical in every comparable aspect to the compound 8 manufactured by the synthetic method as described above inExample 1.

……………………………………………….

A Double-Blind, Placebo-Controlled Study Exploring the Safety, Tolerability, and Efficacy of a 28 Day Course of INCB-039110 in Subjects With Active Rheumatoid Arthritis (NCT01626573)
ClinicalTrials.gov Web Site 2012, June 25

A double-blind, placebo-controlled study exploring the safety, tolerability, and efficacy of a 28-day course of escalating doses of an oral JAK 1 inhibitor (INCB039110) in subjects with stable, chronic plaque psoriasis
22nd Congr Eur Acad Dermatol Venereol (EADV) (October 3-6, Istanbul) 2013, Abst FC01.6

A randomized, dose-ranging, placebo-controlled, 84-day study of INCB039110, a selective janus kinase-1 inhibitor, in patients with active rheumatoid arthritis
77th Annu Sci Meet Am Coll Rheumatol (October 26-30, San Diego) 2013, Abst 1797

Safety Study of INCB-039110 in Combination With Gemcitabine and Nab-Paclitaxel in Subjects With Advanced Solid Tumors (NCT01858883)
ClinicalTrials.gov Web Site 2013, May

An Open-Label, Phase II Study Of The JAK1 Inhibitor INCB039110 In Patients With Myelofibrosis
55th Annu Meet Am Soc Hematol (December 7-10, New Orleans) 2013, Abst 663

WO2013036611A1 * Sep 6, 2012 Mar 14, 2013 Incyte Corporation Processes and intermediates for making a jak inhibitor
WO2013043962A1 * Sep 21, 2012 Mar 28, 2013 Merck Sharp & Dohme Corp. Cyanomethylpyrazole carboxamides as janus kinase inhibitors

Dasotraline, ダソトラリン

Inline image 1

ChemSpider 2D Image | Dasotraline | C16H15Cl2NDasotraline.svgDasotraline.png
(1R,4S)-4-(3,4-Dichlorphenyl)-1,2,3,4-tetrahydro-1-naphthalinamine
1-Naphthalenamine, 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-, (1R,4S)- [ACD/Index Name]
4D28EY0L5T
675126-05-3 [RN]
9885
SEP-225289
UNII:4D28EY0L5T
(1R,4S)-N-DESMETHYL SERTRALINE
(1R,4S)-trans-norsertraline
(1R,4S)-trans-norsertraline|SEP-225289
Dasotraline; 675126-05-3; UNII-4D28EY0L5T; (1R,4S)-trans-Norsertraline; Norsertraline, (1R,4S)-trans-; SEP-225289
ダソトラリン
SEP-225289; SEP-289, DSP-225289

Dasotraline,  SEP-225289, DSP-225289  

1R,4S Transnorsertraline

Generic Name:Dasotraline
Synonym: SEP-225289
Chemical Name:(1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine

4(S)-(3,4-Dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1(R)-ylamine hydrochloride
CAS Number:675126-05-3, Cas of THE DRUG SUBSTANCE hydrochloride is 675126-08-6
Indication:Attention deficit hyperactivity disorder (ADHD)
Drug Company:Sunovion Pharmaceuticals. Inc. in phase 2 as on sept 2014, Sunovion Pharmaceuticals Inc.

http://www.yaopha.com/2014/09/10/chemical-structures-of-drugs-in-clinical-pipeline-snapshot-sep-2014-yaopha%E4%B8%B4%E5%BA%8A%E8%8D%AF%E7%89%A9%E5%8C%96%E5%AD%A6%E7%BB%93%E6%9E%84%E5%BF%AB%E8%AE%AF/

PRONUNCIATION da soe tra’ leen
THERAPEUTIC CLAIM Treatment of attention deficit hyperactivity
disorder (ADHD)
CHEMICAL NAMES
1. 1-Naphthalenamine, 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-, (1R,4S)-
2. (1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine

MOLECULAR FORMULA C16H15Cl2N
MOLECULAR WEIGHT 292.2

SPONSOR Sunovion Pharmaceuticals. Inc.
CODE DESIGNATION SEP-225289
CAS REGISTRY NUMBER 675126-05-3
UNII 4D28EY0L5T
WHO NUMBER 9885

SEP-225289 is an antidepressant which had been in early clinical trials at Sepracor (now Sunovion Pharmaceuticals) for the treatment of major depressive disorder (MDD). In 2010, the company discontinued development of the compound for this indication. At present, phase II clinical trials are under way for the treatment of attention deficit/hyperactivity disorder (ADHD). In preclinical studies, the drug has been shown to be a potent and balanced reuptake inhibitor of serotonin, norepinephrine and dopamine (SNDRI). A drug candidate with a triple mechanism of action as such may provide a broader spectrum of therapy than currently marketed antidepressants.

Recently, drug candidates for blocking the monoamine reuptake transporters have sparked considerable interest in the pharmaceutical industry for treatment of central nervous system disorders. Various candidates are in clinical evaluation in addition to numerous others at the preclinical stage. Sertraline 2 is a selective serotonin reuptake inhibitor (SSRI), marketed by Pfizer as Zoloft for depression. (1R,4S)-4-(3,4-Dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine hydrochloride 1 is structurally similar to sertraline 2 and is currently under investigation for a number of potential central nervous system disorder indications at Sepracor.

Figure

ABOUT SERTRALINE

Sertraline2DACS2.svg

Sertraline-A-3D-balls.png
(1S,4S)-4-(3,4-dichlorophenyl)-N-methyl-1,2,3,4-tetrahydronaphthalen-1-amine

SERTRALINE

Clinicians recognize a distinction among central nervous system illnesses, and there have been many schemes for categorizing mental disorders. The Diagnostic and Statistical Manual of Mental Disorders, Fourth Ed., Text Revision, (hereinafter, the “DSM-IV-TR™”), published by the American Psychiatric Association, and incorporated herein by reference, provides a standard diagnostic system upon which persons of skill rely. According to the framework of the DSM-IV-TR™, the CΝS disorders of Axis I include: disorders diagnosed in childhood (such as, for example, attention deficit disorder or “ADD” and attention deficit / hyperactivity disorder or “ADHD”) and disorders diagnosed in adulthood. CΝS disorders diagnosed in adulthood include

(1) schizophrenia and psychotic disorders; (2) cognitive disorders;(3) mood disorders; (4) anxiety related disorders; (5) eating disorders; (6) substance related disorders; (7) personality disorders; and (8) “disorders not yet included” in the scheme.

Of particular interest to the present invention are adulthood disorders of DSM-IN-TR™ categories (1) through (7) and sexual disorders, currently classified in category (8). Mood disorders of particular interest include depression, seasonal affective disorder and bipolar disorder. Anxiety related disorders of particular interest are agoraphobia, generalized anxiety disorder, phobic anxiety, obsessive compulsive disorder (OCD), panic disorder, acute stress disorder, posttraumatic stress disorder, premenstrual syndrome, social phobia, chronic fatigue disorder, perimenopause, menopause and male menopause.

In general, treatment for psychoses, such as schizophrenia, for example, is quite different than treatment for mood disorders. While psychoses are treated with D2 antagonists such as olanzapine (the “typical” and “atypical” antipsychotics), mood disorders are treated with drugs that inhibit the neuronal reuptake of monoamines, in particular, serotonin (5-HT), norepinephrine (ΝE) and dopamine (DA).

[005] Common therapeutic agents for mood disorders include, but are not limited to, selective serotonin reuptake inhibitors (SSRI’s), including fluoxetine, citalopram, nefazodone, fluvoxamine, paroxetine, and sertraline.

Sertraline, whose chemical name (lS,4S)-c/5 4-(3,4-dichlorophenyl)- 1,2,3,4-tetrahydro-Ν-methyl-l-napthalenamine, is approved for the treatment of depression by the United States Food and Drug Administration, and is available under the trade name ZOLOFT® (Pfizer Inc., NY, NY, USA). In the human subject, sertraline has been shown to be metabolized to (lS,4S)-c« 4- (3,4-dichlorophenyl)-l,2,3,4-tetrahydro-l-napthalenamine, also known as desmethylsertraline or norsertraline. Desmethylsertraline has been described as “not contributing significantly to the serotonergic action of sertraline” Ronfield et al, Clinical Pharmacokinetcs, 32:22-30 (1997). Reports from Hamelin et al, Clinical Pharmacology & Therapeutics, 60:512 (1996) and Serebruany et al, Pharmacological Research, 43:453-461 (2001), have stated that norsertraline is “neurologically inactive”. These statements appear to be based on results observed in serotonin-induced syndrome and ptosis in mouse models in vivo, whereas the original Pfizer research papers suggested on the basis of data in vitro that desmethylsertraline was a selective serotonin uptake inhibitor. Koe et al, JPET, 226:686-700 (1983). Sanchez et al, Cellular and Molecular Neurobiology, 19: 467 (1999), speculated that despite its lower potency, desmethylsertraline might play a role in the therapeutic effects of sertraline but, there is presently no evidence in the literature to support this theory.

] The primary clinical use of sertraline is in the treatment of depression. In addition, United States Patent 4,981,870 discloses and claims the use of sertraline and norsertraline, as well as (lR,4S)-trans 4-(3,4-dichlorophenyl)- 1,2,3,4-tetrahydro-N-methyl-l-napthalenamine and (lS,4R)-trαra 4-(3 ,4- dichlorophenyl)- 1 ,2,3 ,4-tetrahydro-N-methyl- 1 -napthalenamine for the treatment of psychoses, psoriasis, rheumatoid arthritis and inflammation. The receptor pharmacology of the individual (1S,4R) and (1R,4S) enantiomers of trα«5 4-(3,4-dichlorophenyl)-l,2,3,4-tetrahydro-N-methyl-l -napthalenamine is described by Welch et al, J. Med. Chem., 27:1508-1515 (1984). Summary of the Invention

It has now been discovered that {\R,4S)-trans 4-(3,4-dichlorophenyl)- 1,2,3,4-tetrahydro-l-napthalenamine (P) and (lS,4R)-tra«_ 4-(3 ,4- dichlorophenyl)- 1,2,3, 4-tetrahydro-l-napthalenamine (Q) are useful in the treatment of CNS-related disorders that are modulated by monoamine activity, and produce diminished side effects as compared to the current standards of treatment. Treatable CNS disorders include, but are not limited to, mood disorders {e.g., depression), anxiety disorders {e.g., OCD), behavioral disorders {e.g., ADD and ADHD), eating disorders, substance abuse disorders and sexual function disorders. The compounds are also useful for the prophylaxis of migraine.

Compounds P and Q are represented by the formulae:

Figure imgf000005_0001

In one aspect, the present invention relates to a method for treating CNS disorders, which involves the administration of a therapeutically effective amount of P or Q, or a pharmaceutically acceptable salt of either.

In another aspect, the invention relates to trans- 4-(3,4-dichlorophenyl)- 1,2,3,4-tetrahydro-l-napthalenamine of the formula (PQ):

NH2

Figure imgf000006_0001

(PQ)

Skeletal formulae of chlorprothixene and tametraline, from which sertraline was derived

Norsertraline, sertraline’s chief active metabolite

………………………………………..

PATENT

http://www.google.com/patents/US20090149549

(Scheme 2).

Figure US20090149549A1-20090611-C00025

In a preferred embodiment, the compound prepared by the route of Scheme 2 is (1R,4S)-trans 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine. Even more preferred is the preparation of the compound substantially free of its cis isomer.

Example 1

Synthesis of N—((S)-4-(3,4-dichlorophenyl)-3,4-dihydronaphthalen-1-yl)acetamide (3)1.1. Synthesis of Oxime 2

A suspension formed from a mixture of (S)-tetralone 1 (56.0 g, 0.192 mol), hydroxylamine hydrochloride (14.7 g, 0.212 mol), and sodium acetate (17.4 g, 0.212 mol) in methanol (168 mL) was heated to reflux for 1 to 5 hours under a N2atmosphere. The progress of the reaction was monitored by HPLC. After the reaction was complete, the reaction mixture was concentrated in vacuo. The residue was diluted with toluene (400 mL) and 200 mL water. The organic layer was separated and washed with an additional 200 mL water. The organic layer was concentrated and dried to give crude solid oxime 2 (58.9 g, 100%), m. p. 117-120° C.

1H NMR (400 MHz, CDCl3) δ (ppm) 9.17 (br, 1H, OH), 7.98 (m, 1H), 7.36 (d, 1H, J=8.0 Hz), 7.29 (m, 2H), 7.20 (d, 1H, J=2.4 Hz), 6.91 (m, 2H), 4.11 (dd, 1H, J=7.2 Hz, 4.4 Hz), 2.82 (m, 2H), 2.21 (m, 1H), 2.08 (m, 1H). 13C NMR (100 MHz, CDCl3) δ 154.94, 144.41, 140.40, 132.83, 130.92, 130.82, 130.68, 130.64, 129.98, 129.38, 128.12, 127.64, 124.48, 44.52, 29.51, 21.27.

1.2. Synthesis of Enamide 3

The solution of the crude oxime 2 (59 g, 0.193 mol) in toluene (500 mL) was purged with N2 for 30 min. Et3P (25 g, 0.212 mol) was charged. After stirring for 10 min, acetic anhydride (21.6 g, 20 mL, 0.212 mol) was added. The reaction mixture was refluxed for 8 to 13 h. Progress of the reaction was monitored by HPLC. The reaction mixture was cooled to room temperature. 6N NaOH (aq) (86 mL, 0.516 mol) and 1.0 M (n-Bu)4NOH in methanol (1.0 mL) were added. The hydrolysis was complete in about 2 to 4 h. The organic layer was separated and diluted with EtOAc (300 mL) and 2-BuOH (30 mL). The diluted organic solution was washed with 1% HOAc (aq) solution (300 mL) and DI water (3×300 mL) and concentrated to about 350 mL of a slurry in vacuo. The slurry was diluted with heptane (100 mL) and 2-BuOH (4 mL) and heated to reflux to form a clear solution. Heptane (50 to 200 mL) was slowly added until a cloudy solution formed. The suspension was slowly cooled to rt. The product was filtered out, washed with 30% toluene and 70% heptane (3×100 mL) solution and dried in a vacuum oven to give 56.9 g white solid (enamide 3, 89% yield), m. p. 167-168° C.

(S)-Tetralone 1 (50.0 g, 0.172 mol) was slurried in methanol (150 mL) with hydroxylamine hydrochloride (13.1 g, 0.189 mol) and sodium acetate (15.5 g, 0.189 mol). The resulting suspension was heated to reflux for 2 to 6 h under an inert atmosphere with progress monitored by HPLC. On completion, the mixture was cooled to 25° C., diluted with toluene (300 mL) and quenched with 1.7 N NaOH (100 mL). The mixture was concentrated in vacuo under reduced pressure, the aqueous layer removed and the organic layer washed further with DI water (100 mL). Further toluene (300 mL) was charged to the vessel and water removed by azeotropic distillation. Once at ambient temperature, n-Bu3P (47.1 mL, 0.183 mol) was charged to the reactor, followed by acetic anhydride (32.5 mL, 0.344 mol). The reaction was heated to reflux and monitored by HPLC. After 20-24 h, the reaction was cooled to ambient temperature and quenched with 6 N NaOH (120 mL). This mixture was allowed to react for 2 to 6 h before the aqueous layer was removed. The organic phase was washed with DI water (100 mL). Concentration of the mixture in vacuo, cooling to room temperature and diluting with isopropanol (50 mL) was done prior to addition of heptane to assist with crystallization. An initial charge of heptane (50 mL) was followed by an additional 650 mL. Aging of the slurry followed by filtration, washing (4×100 mL heptane) and drying yielded a light yellow solid (enamide 3, 44.1 g, 77%).

1H NMR (400 MHz, CDCl3) δ (ppm) 7.35 (d, 1H, J=8.4 Hz), 7.26 (m, 3H), 7.17 (m, 1H), 7.05 (dd, 1H, J=8.0, 1.6 Hz), 7.00 (br, 1H), 6.87 (m, 0.82H, 82% NH rotamer), 6.80 (br, 0.18H, 18% NH rotamer), 6.31 (t, 0.82H, J=4.8 Hz, 82% H rotamer), 5.91 (br, 0.18H, 18% H rotamer), 4.12 (br, 0.18H, 18% H rotamer), 4.03 (t, 0.82H, J=8.0 Hz, 82% H rotamer), 2.72 (m, 1H), 2.61 (ddd, 1H, J=16.8, 8.0, 4.8 Hz), 2.17 (s, 2.46H, 82% CH3 rotamer), 1.95 (s, 0.54H, 18% CH3 rotamer). 100 MHz13CNMR (CDCl3) δ 169.3, 143.8, 137.7, 132.3, 131.8, 131.4, 130.5, 130.3, 130.2, 128.8, 128.1, 127.8, 127.2, 123.8, 122.5, 121.2, 117.5, 42.6, 30.3, 24.1.

Example 2Synthesis of N-((1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-yl)acetamide (4)

The enamide 3 (24 g, 72 mmol) was slurried in degassed isopropanol (200 mL). The resulting slurry was transferred to the appropriate reactor. Prior to the addition of the catalyst solution, the content of the reactor was purged with nitrogen. A solution of (R,R)-MeBPE(COD)RhBF4 catalyst (20.1 mg, 0.036 mmol, 0.05 mol %) in isopropanol (IPA) (100 mL) was added to the reactor. The content was cooled to 0° C. and purged with nitrogen three times. The reactor was then purged with hydrogen and pressurized to 90 psig. The reaction was aged with agitation at 0° C. for 7.5 h and conversion was monitored by the hydrogen uptake. The content was then warmed to RT and hydrogen was vented. After purging with nitrogen, the contents were drained. The reaction mixture was heated to 50° C. and filtered through a pad of Celite. The clear orange solution was concentrated to ˜50% volume (150 mL) and diluted with toluene (5.9 g, 5 wt %). The suspension was heated to 65° C. and water (14.7 mL) was added dropwise to form a cloudy solution. The slurry was slowly cooled to −10° C. and aged for 30 minutes. The solid was filtered and washed with cold IPA (2×45 mL). The cake was dried under vacuum at 45° C. overnight to afford 20.0 g (83% yield) of trans acetamide 4 (>99% de).

1H NMR (CDCl3) 400 MHz δ 7.34 (dd, 2H, J=7.9, 2.4 Hz), 7.23 (t, 1H, J=7.5 Hz), 7.15 (m, 2H), 6.85 (dd, 1H, J=8.2, 2.0 Hz), 6.82 (d, 1H, J=7.7 Hz), 5.72 (d, 1H, J=8.4 Hz), 5.31 (dd, 1H, J=13.2, 8.1 Hz), 4.10 (dd, 1H, J=7.0, 5.9 Hz), 2.17 (m, 2H), 2.06 (s, 3H), 1.87 (m, 1H). 1.72 (m, 1H); 13C NMR (CDCl3) 100 MHz δ 169.7, 146.9, 138.8, 137.7, 132.6, 130.8, 130.6, 130.5, 130.3, 128.4, 128.3, 127.9, 127.4, 47.9, 44.9, 30.5, 28.4, 23.8.

Example 3

Synthesis of (1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine Hydrochloride (5)

A solution of trans-acetamide 4 (9.0 g, 26.9 mmol), n-propanol (45 mL) and 5M hydrochloric acid (45 mL) was refluxed for approximately 48 h (90-93° C.). During this time, the reaction temperature was maintained at ≧90° C. by periodically collecting the distillate until the reaction temperature was >92° C. Additional n-propanol was added periodically to maintain the solution at its original volume. After the hydrolysis was complete, the solution was slowly cooled to 0° C., resulting in a slurry, which was aged for one hour at 0° C. The reaction mixture was filtered, and the cake was washed with 1:1 methanol/water (20 mL), followed by t-butyl methyl ether (20 mL). The wet-cake was dried under vacuum at 45 to 50° C. to afford 7.0 g of the amine hydrochloride 5 (80% yield).

1H NMR (DMSO-d6) δ 1.81-1.93 (m, 2H), 2.12-2.21 (m, 1H), 2.28-2.36 (m, 1H), 4.28 (t, 1H, J=6.8), 4.59 (br.s, 1H), 6.84 (d, 1H, J=7.6), 7.05 (dd, 1H, J=8.4, 1.6), 7.25 (t, 1H, J=7.6), 7.32 (t, 1H, J=7.6), 7.37 (d, 1H, J=1.6), 7.56 (d, 1H, J=8.4), 7.76 (d, 1H, J=7.2), 8.80 (br.s, 3H);

13C NMR (DMSO-d6) 147.4, 138.9, 133.6, 131.0, 130.5, 130.4, 130.1, 129.0, 128.9, 128.4, 128.2, 126.8, 47.9, 43.1, 27.8, 25.2.

INTERMEDIATE

Example 5 Catalytic Asymmetric Hydrogenation of the Enamide 3 Using (R,S,R,S)-MePenn Phos(COD)RhBF4 as the Catalyst

As shown in Scheme 4, the enamide 3 was subjected to homogeneous catalytic asymmetric hydrogenation in the presence of a chiral catalyst, H2, and a solvent. In this example the catalyst was derived from the complex of the transition metal rhodium with the chiral phosphine ligand, (1R,2S,4R,5S)—P,P-1,2-phenylenebis {(2,5-endo-dimethyl)-7-phosphabicyclo[2.2.1]heptane}(R,S,R,S-MePennPhos). The hydrogenations were carried out at a substrate concentration of about 0.12 M to about 0.24 M of compound 3.

Figure US20090149549A1-20090611-C00043

………………………………………..

Koenig, Stefan G.; Vandenbossche, Charles P.; Zhao, Hang; Mousaw, Patrick; Singh, Surendra P.; Bakale, Roger P.
Organic Letters, 2009 ,  vol. 11,  2  pG . 433 – 436

http://pubs.acs.org/doi/abs/10.1021/ol802482d

Abstract Image

Imidoyl chlorides, generated from secondary acetamides and oxalyl chloride, can be harnessed for a selective and practical deprotection sequence. Treatment of these intermediates with 2 equiv of propylene glycol and warming enables the rapid release of amine hydrochloride salts in good yields. Notably, the reaction conditions are mild enough to allow for a swift deprotection with no observed epimerization of the amino center.

Supporting Information             A Facile Deprotection of Secondary Acetamides

http://pubs.acs.org/doi/suppl/10.1021/ol802482d/suppl_file/ol802482d_si_001.pdf

(1R,4S)-4-(3,4-Dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine hydrochloride – Compound 1, Scheme 1 / Table 3, entry 1A:

decomp. > 290 °C.

1H NMR (400 MHz, DMSO-d6) δ 8.71 (s, 3H), 7.71 (d, 1H, J = 7.7 Hz), 7.53 (d, 1H, J = 8.1 Hz), 7.34 (s, 1H),
7.29 (m, 1H), 7.22 (m, 1H), 7.01 (d, 1H, J = 8.1 Hz), 6.81 (d, 1H, J = 7.7 Hz), 4.56 (s,
1H), 4.26 (s, 1H), 2.26 (m, 1H), 2.15 (m, 1H), 1.83 (m, 2H).

13C NMR (100 MHz, DMSO-d6) δ 147.3, 138.8, 133.5, 130.9, 130.5, 130.4, 130.0, 128.9, 128.8, 128.3, 128.1,
126.7, 47.8, 43.0, 27.7, 25.1.

NMR  GRAPHS GIVEN

Inline image 1

13 C NMR

Inline image 2

………………………………………..

Jerussi, T. P.; Fang, Q. K.; Currie, M. G. PCT Int. Appl. WO 2004042669 A1 200440325, 2004.

http://www.google.com/patents/WO2004024669A1?cl=en

Figure

The discovery route involved preparation of (S)-tetralone (4S)-3 from racemic tetralone(4RS)-3 via chromatographic separation of sulfinyl imine (Rs,4RS)-5 diastereomers, followed by hydrolysis. The sulfinyl imine isomers were generated by condensation with (R)-tert-butylsulfinamide ((R)-TBSA), (Rs)-4, in the presence of titanium ethoxide. The yield of sulfinyl imine diastereomer (Rs,4S)-5 was ∼15% after chromatographic purification. The low recovery yield was due to chromatographic loss and the instability of compound 5 on silica gel. The resulting (S)-tetralone (4S)-3 was converted to N-formyl amine (1RS,4S)-6 as a mixture of two diastereomers that were again separated by chromatography to afford the desired diastereomer(1R,4S)-6 in 17% yield over two steps. (1R,4S)-trans-norsertraline 1 was obtained after the acidic hydrolysis of (1R,4S)-6 in 71% yield. The overall yield of this route was less than 2% and involved two chromatographic purifications, making it impractical for an efficient large-scale synthesis of 1.

Jerussi, T. P.; Fang, Q. K.; Currie, M. G. PCT Int. Appl. WO 2004042669 A1 200440325, 2004.http://www.google.com/patents/WO2004024669A1?cl=en

……………………………………………

PAPER

Development of a large-scale stereoselective process for (1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine hydrochloride
Org Process Res Dev 2007, 11(4): 726

http://pubs.acs.org/doi/abs/10.1021/op7000589

Abstract Image

A convenient, multikilogram-scale, stereoselective process for the synthesis of (1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine hydrochloride 1 is described. The key steps involve synthesis of sulfinyl imine (Rs,4S)-5 from (S)-tetralone (4S)-3 and (R)-tert-butylsulfinamide (Rs)-4, and its stereoselective reduction with 9-BBN to produce the (1R)-amine center of 1. The process has been scaled up to multikilogram scale and gives 1 in an overall yield of >50% with a chemical purity of 99.7 A% by HPLC and stereochemical purity of >99.9% by chiral HPLC.

(1R,4S)-4-(3,4-Dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-ylamine HCl (1).
 
1H NMR (400 MHz, DMSO-d6) δ 1.81−1.93 (m, 2H), 2.12−2.21 (m, 1H), 2.28−2.36 (m, 1H), 4.28 (t, 1H, J = 6.8 Hz), 4.59 (br s, 1H), 6.84 (d, 1H, J = 7.6 Hz), 7.05 (dd, 1H, J = 8.4, 1.6 Hz), 7.25 (t, 1H, J = 7.6 Hz), 7.32 (t, 1H, J = 7.6 Hz), 7.37 (d, 1H, J = 1.6 Hz), 7.56 (d, 1H, J = 8.4 Hz), 7.76 (d, 1H, J = 7.2 Hz), 8.80 (br s, 3H).
 
13C NMR (100 MHz, DMSO-d6) δ 147.4, 138.9, 133.6, 131.0, 130.5, 130.4, 130.1, 129.0, 128.9, 128.4, 128.2, 126.8, 47.9, 43.1, 27.8, 25.2.
 
Anal. Calcd for C16H15Cl2N:  C, 58.47; H, 4.91; N, 4.26; Cl, 32.36. Found:  C, 58.44; H, 4.79; N, 4.21; Cl, 32.53.

……………………………………………………………..

WO 2004024669

http://www.google.com/patents/WO2004024669A1?cl=en

Preparation of compounds of the present invention is illustrated below in Scheme 1 and its accompanying narrative.

Figure imgf000007_0001

[0015] In the compound

Figure imgf000008_0001

of Scheme 1,

R is R,° , wherein R1, R2 and R3 are each independently alkyl. In a preferred embodiment of the compounds, R is tert-butyl.

[0016] N-[4-(3 ,4-dichlorophenyl)- 1 ,2,3 ,4-tefrahydronaphthalen- 1 -yl]formamide, the intermediate in the synthesis shown in Scheme 1 , exists in four stereoisomeric forms:

Figure imgf000008_0002

C (1S,4S) D (1R.4R) [0017] When N-[4-(3 ,4-dichlorophenyl)- 1 ,2,3,4-tetrahydronaρhthalen-l – yl]formamide is synthesized from achiral starting materials via non- stereoselective syntheses, all four isomers will be produced. The mixture can be readily separated into a racemic cis diastereomer and a racemic trans diastereomer by means, such as recrystallization or chromatography on achiral media, that rely on chemical and physical differences.

[0018] The trans diastereomer, represented as E below, is a 1 :1 mixture of A and B. When E is hydrolyzed, PQ is produced; when A is hydrolyzed, P is produced; when B is hydrolyzed, Q is produced. The cis diastereomer, represented as F below, is a 1 : 1 mix of C and D.

Figure imgf000009_0001

E = A + B F = C + D

……………………………………………………………………………………………

WO 2007006003

http://www.google.com/patents/WO2007006003A2?cl=en

Figure imgf000027_0001

Scheme 3

Production (lR,4S)-4-(3,4-dichloro-phenyl)-l,2,3,4-tetrahydro-naphthalen-l-ylamine HCl from 4-(S)-(3,4-dichloro-phenyl)-3,4-dihydro-2H-naphthalen-l-one.

Figure imgf000031_0001

(S)-(3,4-Dichloro-phenyl)-3,4- (1 R,4S)-4-(3,4-Dichloro-phenyl)-1 ,2,3,4-tetrahydro- d ιhydro-2H-naphthalen-1 -one naphthalen-1 -ylamine; [0080] Charge 4-(S)-(3,4-dichloro-phenyl)-3,4-dihydro-2H-naρhthalen-l-one (1 kg, 3.4 mol) and (R)-tert-butylsulphinamide (TBSA, 464 g, 3.8 mol) to a suitable reactor and dissolved in about 7 L THF. Add a 20%wt solution of Titanium ethoxide in ethanol (about 7.8 kg, 6.9 mol) and heat the mixture to about 70 0C for about 24h. The reaction is monitored by HPLC, and after the reaction is complete, cool the mixture to room temperature and added a 24% wt aqueous solution of NaCl to the mixture. The resultant slurry was filtered and washed multiple times with about 1 L total of ethyl acetate. The mother liquors and washes were concentrated to a minimum volume. The aqueous phase was extracted with about 5 L of ethyl acetate and evaporated to dryness.

[0081] The contents were then dissolved in about 7 L of THF and cooled to about —10 0C. About 9 kg, (~5 mol) of a 0.5 M solution of 9-borabicycIononane (9-BBN) in THF, was added slowly (about 3h) and the mixture was stirred at 0 0C until reaction completion. A 6N HCl/methanol (~2L) was added to the mixture and stirred until the hydrolysis reaction was complete. After neutralization with about 2 L of 6N aqueous NaOH, the mixture was distilled to remove THF and the residue (aqueous phase) was extracted twice with methyl t- butyl ether (2x6L). The organic phase was then washed with water. The organic phase was concentrated, then cooled to 00C followed by addition of 2N HCl in methyl t-butyl ether (3 L). The product slowly precipitated as the HCl salt during the addition. The slurry was filtered and washed with methyl t-butyl ether (2x2L). The product was dried under vacuum at about 45°C to afford about 850 g of Re-Crystallization of crude (lR,4S)-4-(3,4-dichloro-phenyl)- 1,2,3,4-tetrahydro-naphthalen-l-ylamine HCl.

[0082] The resulting (lR,4S)-4-(3,4-dichloro-phenyl)-l,2,3,4-tetrahydro- naphthalen-1-ylamine HCl (85Og) was charged to a suitable reactor and about 30 L of denatured ethanol was added. The mixture was heated to reflux, the volume was reduced to about 50% via distillation, and then cooled to 50°C. About 30 L of Hexane was added to the slurry to complete the product crystallization and then the slurry was cooled to about 00C. The product was isolated by filtration, the cake was washed with about 2 L of ethanol/hexane (1/3 v/v) and then about 2 L of ethyl acetate, followed by about 3 L of hexane. The wet cake was dried under vacuum at about 45°C to afford 630 g of product.

[0083] Another alternative process for preparation of compound P is presented below.

[0084] 4-(S)-(3,4-dichloro-phenyl)-3,4-dichloro-2H-naphthalen-l-one (4.11 kg) and (R)-tert-butylsulphinamide (TBSA, 1.9 kg) were charged to a suitable reactor and dissolved in 29 L THF. A 20%wt solution of titanium ethoxide in ethanol (31.6 kg) was added and the mixture was heated to 70 °C with stirring. The reaction is monitored by HPLC, and after the reaction was complete (20-24 h) the mixture was cooled to room temperature and added to 20 L of a 24 wt% aqueous solution of NaCl. The resultant slurry was filtered and washed 3 times with ethyl acetate (4.1 L). The mother liquors and washes were concentrated to a minimum volume. The aqueous phase was extracted with about 20 L of a 1 :1 mix of ethyl acetate and toluene. The organic phases were combined and concentrated to half volume to give a solution of 2. A purified sample of 2 was analyzed: m.p. 104 0C, 1HNMR (400 MHz, CDCl3) δ (ppm) 8.23 (dd, IH, J= 7.9, 0.9 Hz), 7.38 (ddd, IH, J= 14.7, 7.3, 1.5 Hz), 7.37 (d, IH, J= 8.4 Hz), 7.33 (d, IH, J= 7.7 Hz), 7.17 (d, IH, J= 1.8 Hz), 6.93 (d, IH, J= 7.7 Hz), 6.89 (dd, IH, J= 8.4, 2.2 Hz), 4.18 (dd, IH, J= 7.3, 4.8 Hz), 3.36 (ddd, IH, J= 17.5, 8.8, 4.4 Hz), 2.93 (ddd, IH, J= 17.6, 8.3, 4.2 Hz), 2.33 (m, IH), 2.15 (m, IH), 1.34 (s, 9H). 13C NMR (100 MHz, CDCl3) δ 175.8, 144.2, 142.7, 132.6, 130.8, 130.7, 129.7, 128.1, 127.6, 127.4, 57.8, 44.3, 31.1, 29.4, 22.8. HRMS calc for C20H2ICl2NOS 394.0799, found 394.0767.

[0085] The solution of imine (2) was cooled to -10 0C and 36.3 kg of a 0.5 M solution of 9-borabicyclononane (9-BBN) in THF, was added slowly (over 3h) and the mixture was stirred at 0 0C until reaction completion. A 4N HCl/methanol (8 L) was added to the mixture and stirred until the hydrolysis reaction was complete. After neutralization with about 15 kg of 6N aqueous NaOH (pH 8), the mixture was distilled to remove THF and methanol. The residue (aqueous phase) was extracted twice with methyl t-butyl ether (2 x 16L). The organic phase was then washed with water. The organic phase was concentrated, then cooled to 00C followed by addition of 2N HCl in methyl t- butyl ether (5.4 kg). The product precipitated as the HCl salt. The slurry was filtered, washed with methyl t-butyl ether (2 x 8L) and dried under vacuum at 450C to afford about 3.73 kg of crude (lR,4S)-4-(3,4-dichloro-phenyl)-l,2,3,4- tetrahydro-naphthalen-1-ylamine HCl (compound P).

A purified sample of P was analyzed:  NOTE P IS DASOTRALINE

m.p. 152 – 154 0C,

1H NMR (400 MHz, CDCl3) δ (ppm) 7.58 (d, IH, J= 7.7 Hz), 7.29 (m, 2H), 7.18 (br. t, IH, J= 7.5 Hz), 7.09 (d, IH, J= 1.8 Hz), 6.87 (d, IH, J= 7.7 Hz), 6.80 (dd, IH, J= 8.3, 2.0 Hz), 4.65 (dd, IH, J= 4.4, 4.4 Hz), 4.15 (t, IH, J= 5.5 Hz), 3.30 (d, IH, J= 3.7 Hz), 2.35 (m, IH), 1.95 (m, IH), 1.85 (m, IH), 1.75 (m, IH), 1.23 (s, 9H).

13C NMR (100 MHz, CDCl3) δ 147.1, 138.4, 138.0, 132.6, 130.8, 130.6, 130.5, 129.8, 128.3, 127.9, 55.8, 53.3, 44.0, 28.2, 27.7, 22.9.

HRMS calc for C20H23Cl2NOS 396.0956, found 396.0968.

[0086] The crude (lR,4S)-4-(3,4-dichloro-phenyl)-l,2,3,4-tetrahydro- naphthalen-1-ylamine HCl (3.63 kg) was charged to a suitable reactor and 128 L of denatured ethanol was added. The mixture was stirred at reflux and polish filtered. The volume was reduced to about 50% via distillation, and then cooled to 500C. 80 L of heptane was added to the slurry to complete the product crystallization and then the slurry was cooled to -5 °C. The product was filtered, the cake was washed twice with 5.7 L of ethanol/heptane (1/1 v/v) and then washed with 6 L of hexane. The wet cake was dried under vacuum at about 45°C to afford 2.57 kg of product. The product had a chemical purity of 99.65 A% and a diastereomeric purity in excess of 99%

…………………………………………………………………………

PATENT

WO 2011069032

http://www.google.com/patents/WO2011069032A2?cl=en

Transnorsertraline, i. e. , (1 R,4S)-trans-4-(3 ,4-dichlorophenyl)- 1 ,2,3 ,4-tetrahydro- 1 – naphthalenamine and (lS,4R)-trans-4-(3,4-dichlorophenyl)-l,2,3,4-tetrahydro-l- naphthalenamine are described in, for example, U.S. Patent No. 7,087,785 B2 (“the ‘785 patent”; incorporated herein by reference in its entirety), have the following chemical structures, respectively:

Figure imgf000002_0001

Uses of transnorsertraline in the treatment, prevention, or management of affective disorders and other various CNS disorders are also disclosed in the ‘785 patent. Such disorders include, but are not limited to, depression, mood disorders, anxiety disorders, behavioral disorders, eating disorders, substance abuse disorders, and sexual function disorders.

ref

A Randomized, Double-Blind, Parallel-Group, Multicenter Efficacy and Safety Study of SEP-225289 Versus Placebo in Adults With Attention Deficit Hyperactivity Disorder (ADHD) (NCT01692782)
ClinicalTrials.gov Web Site 2012, September 27

Characterization of the electrophysiological properties of triple reuptake inhibitors on monoaminergic neurons
Int J Neuropsychopharmacol 2011, 14(2): 211

PET evaluation of serotonin and dopamine transporter occupancy associated with administration of SEP-225289
Biol Psychiatry 2010, 67(9, Suppl. 1): Abst 102
[65th Annu Meet Soc Biol Psychiatry (SOBP) (May 20-22, New Orleans) 2010]

Koenig, Stefan G.; Vandenbossche, Charles P.; Zhao, Hang; Mousaw, Patrick; Singh, Surendra P.; Bakale, Roger P.
Organic Letters, 2009 ,  vol. 11, (2)  pg 433 – 436

Thalen, Lisa K.; Zhao, Dongbo; Sortais, Jean-Baptiste; Paetzold, Jens; Hoben, Christine; Baeckvall, Jan-E.
Chemistry – A European Journal, 2009 ,  vol. 15, ( 14)  pg. 3403 – 3410

US8134029 Jul 30, 2010 Mar 13, 2012 Sunovion Pharmaceuticals Inc. Treatment of CNS disorders with trans 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-napthalenamine
US8658700 Dec 4, 2012 Feb 25, 2014 Sunovion Pharmaceuticals Inc. Treatment of CNS disorders with trans 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-napthalenamine
US20010044474 * Dec 20, 2000 Nov 22, 2001 Curatolo William J. Hydrogel-driven layered drug dosage form
US20060257475 * Aug 17, 2006 Nov 16, 2006 Boehringer Ingelheim International Gmbh Stable Sertraline Hydrochloride Formulation and Method
US20080280993 * Jul 15, 2008 Nov 13, 2008 Sepracor Inc. Treatment of CNS Disorders With trans 4-(3,4-Dichlorophenyl)-1,2,3,4-Tetrahydro-1-Napthalenamine
Dasotraline is a serotoninnorepinephrine and dopamine reuptake inhibitor (SNDRI) that is under investigation for the treatment of Binge Eating Disorder, Adult Attention Hyperactivity Disorder, Attention Deficit Hyperactivity Disorder, and Adult Attention Deficit Hyperactivity Disorder

Dasotraline (INN)[1] (former developmental code name SEP-225,289) is a serotonin-norepinephrine-dopamine reuptake inhibitor(SNDRI) that is under development by Sunovion for clinical use.[2][3][4][5] In 2017, the U.S. Food and Drug Administration accepted Sunovion’s New Drug Application for review for the treatment of ADHD;[6] the NDA for dasotraline is expected to be decided by the end of August 2018.[7] The drug is no longer being developed for major depressive disorder (MDD), but is still under investigation for the treatment of attention-deficit hyperactivity disorder (ADHD) and eating disorders.[8][9] Structurally, dasotraline is a stereoisomer of desmethylsertraline, which is an active metabolite of the marketed selective serotonin reuptake inhibitor (SSRI) antidepressantsertraline (Zoloft) and an SNDRI similarly.

Side Effects

In phase I trials for ADHD, test subjects reported the following side effects:[10][11]

Syn1

SYN2

Clip

https://patents.google.com/patent/US20150196502

Transnorsertraline, i.e., (1R,4S)-trans-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine and (1S,4R)-trans-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine are described in, for example, U.S. Pat. No. 7,087,785 B2 (“the ‘785 patent”; incorporated herein by reference in its entirety), have the following chemical structures, respectively:

Figure US20150196502A1-20150716-C00001

Uses of transnorsertraline in the treatment, prevention, or management of affective disorders and other various CNS disorders are also disclosed in the ‘785 patent. Such disorders include, but are not limited to, depression, mood disorders, anxiety disorders, behavioral disorders, eating disorders, substance abuse disorders, and sexual function disorders.

2.2 Salts and Polymorphic Forms
Whether crystalline or amorphous, potential solid forms of a pharmaceutical compound include single-component and multiple-component solids. Single-component solids consist essentially of the pharmaceutical compound in the absence of other compounds. Variety among single-component crystalline materials may potentially arise, e.g., from the phenomenon of polymorphism, wherein multiple three-dimensional arrangements exist for a particular pharmaceutical compound (see, e.g., S. R. Byrn et al., Solid State Chemistry of Drugs, (1999) SSCI, West Lafayette).
Solid forms such as salts, crystal forms, e.g., polymorphic forms of a compound are known in the pharmaceutical art to affect, for example, the solubility, stability, flowability, fractability, and compressibility of the compound as well as the safety and efficacy of drug products based on the compound, (see, e.g., Knapman, K. Modern Drug Discoveries, 2000: 53).
The importance of studying polymorphs was underscored by the case of ritonavir, an HIV protease inhibitor that was formulated as soft gelatin capsules. About two years after the product was launched, the unanticipated precipitation of a new, less soluble polymorph in the formulation necessitated the withdrawal of the product from the market until a more consistent formulation could be developed (see S. R. Chemburkar et al., Org. Process Res. Dev., (2000) 4:413-417). Thus, the preparation of solid forms is of great importance in the development of a safe, effective, stable and marketable pharmaceutical compound.
New salts and polymorphic forms of transnorsertraline can further the development of formulations for the treatment, prevention or management of CNS diseases.
hydrochloride salt of transnorsertraline exists as a monohydrate.

Paper

Chemistry – A European Journal (2009), 15(14), 3403-3410.

A Chemoenzymatic Approach to Enantiomerically Pure Amines Using Dynamic Kinetic Resolution: Application to the Synthesis of Norsertraline

Dynamic transformation: A racemization catalyst and the enzyme Candida antarcticalipase B (CALB) were combined in a one‐pot dynamic kinetic resolution (DKR) of primary amines, which were transformed to their corresponding amides in up to 95 % yield and >99 % ee. This chemoenzymatic DKR was also applied to the synthesis of norsertraline (see scheme).

Racemization catalyst 5 c and the enzyme Candida antarctica lipase B were combined in a one‐pot dynamic kinetic resolution (DKR) of primary amines in which a wide range of amines were transformed to their corresponding amides in up to 95 % isolated yield and >99 % ee. The DKR protocol was applicable with either isopropyl acetate or dibenzyl carbonate as the acyl donor. In the latter case, release of the free amine from the carbamate products was carried out under very mild conditions. The racemization of (S)‐1‐phenylethylamine with several different Ru catalysts was also evaluated. Catalyst 5 c, of the Shvo type, was able to selectively racemize amines and was also compatible with the reaction conditions used for DKR. A racemization study of three different amines with varying electronic properties was also performed. Competitive racemization of a 1:1 mixture of the deuterated and non‐deuterated amine was carried out with 5 c and a primary kinetic isotope effect was observed for all three amines, providing support that the rate‐determining step is β‐hydride elimination. The chemoenzymatic DKR protocol was applied to the synthesis of norsertraline (16) by using a novel route starting from readily available 1,2,3,4‐tetrahydro‐1‐naphthylamine (1 o)………https://onlinelibrary.wiley.com/action/downloadSupplement?doi=10.1002%2Fchem.200802303&file=chem_200802303_sm_miscellaneous_information.pdf

Synthesis of (1R,4S)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-aminehydrochloride(NorSertraline, (1R,4S)-16)

1 H NMR (400 MHz, CD3OD) δ = 7.54 – 7.50 (m, 1H), 7.46 (d, J = 8.3 Hz, 1H), 7.40 (td, J = 3.7 Hz, 1.3 Hz, 1H), 7.33 (td, J = 3.7 Hz, 1.3 Hz, 1H), 7.20 (d, J = 2.1 Hz, 1H), 7.02-6.97 (m, 2H), 4.68 (dd, 1H), 4.33 (dd, 1H), 2.44 – 2.22 (m, 2H), 2.01 – 1.88 (m, 2H).

13C NMR (101 MHz, CD3OD) δ = 146.8, 138.9, 132.5, 131.9, 130.6, 130.3, 130.2, 130.0, 128.9, 128.3, 127.7, 127.3, 48.8, 43.5, 27.8, 24.9.

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013151877&recNum=109&docAn=US2013034529&queryString=EN_ALL:nmr%20AND%20PA:(Bristol-Myers%20Squibb)%20&maxRec=4406

PATENT

ApplicationPriority dateFiling dateRelationTitle
PCT/US2010/0588312009-12-042010-12-03DivisionFormulations, salts and polymorphs of transnorsertraline and uses thereof
US2013135131702013-01-092013-01-09Division
US156319092009-12-042017-06-23ContinuationFormulations, salts and polymorphs of transnorsertraline and uses thereof
US266864092009-12-042009-12-04US Provisional Application
PCT/US2010/0588312009-12-042010-12-03Formulations, salts and polymorphs of transnorsertraline and uses thereof
US2013135131702013-01-092013-01-09US Provisional Application
US146053342009-12-042015-01-26Formulations, salts and polymorphs of transnorsertraline and uses thereof
US146053342015-01-26Formulations, salts and polymorphs of transnorsertraline and uses thereof
US156319092017-06-23Formulations, salts and polymorphs of transnorsertraline and uses thereof

References

  1. Jump up^ “International Nonproprietary Names for Pharmaceutical Substances (INN)” (PDF). WHO Drug Information. WHO. 27 (4). 2013. Retrieved 4 November 2014.
  2. Jump up^ Chen, Zhengming; Skolnick, Phil (2007). “Triple uptake inhibitors: therapeutic potential in depression and beyond”. Expert Opinion on Investigational Drugs16 (9): 1365–77. doi:10.1517/13543784.16.9.1365PMID 17714023.
  3. Jump up^ DeLorenzo, C.; Lichenstein, S.; Schaefer, K.; Dunn, J.; Marshall, R.; Organisak, L.; Kharidia, J.; Robertson, B.; Mann, J. J.; Parsey, R. V. (2011). “SEP-225289 Serotonin and Dopamine Transporter Occupancy: A PET Study”Journal of Nuclear Medicine52 (7): 1150–5. doi:10.2967/jnumed.110.084525PMC 3856248Freely accessiblePMID 21680689.
  4. Jump up^ Ziegler, L.; Küffer, G.; Euler, E.; Wilhelm, K. (1990). “Arthrographische Darstellung von Ganglien im Handbereich” [Arthrographic imaging of ganglions of the hand]. RöFo (in German). 153 (2): 143–6. doi:10.1055/s-2008-1033352PMID 2168068.
  5. Jump up^ Guiard, B.; Chenu, F.; El Mansari, M.; Blier, P. (2009). “P.1.c.059 Electrophysiological properties of the triple reuptake inhibitor SEP 225289 on monoaminergic neurons”. European Neuropsychopharmacology19: S285. doi:10.1016/S0924-977X(09)70419-5.
  6. Jump up^ “Sunovion Announces FDA Acceptance for Review of New Drug Application for Dasotraline for the Treatment of ADHD” (Press release). Marlborough, Massachusetts: Sunovion. Business Wire. November 10, 2017. Retrieved 2018-05-01.
  7. Jump up^ “Pipeline Report: Brand Drugs” (PDF). Welldyne. February 2018. pp. 1, 4. Retrieved 1 May 2018.
  8. Jump up^ Clinical trial number NCT02276209 for “Dasotraline Adult ADHD Study” at ClinicalTrials.gov
  9. Jump up^ http://adisinsight.springer.com/drugs/800023450[full citation needed]
  10. Jump up^ Koblan, Kenneth S; Hopkins, Seth C; Sarma, Kaushik; Jin, Fengbin; Goldman, Robert; Kollins, Scott H; Loebel, Antony (2015). “Dasotraline for the Treatment of Attention-Deficit/Hyperactivity Disorder: A Randomized, Double-Blind, Placebo-Controlled, Proof-of-Concept Trial in Adults”Neuropsychopharmacology40 (12): 2745–52. doi:10.1038/npp.2015.124PMC 4864650Freely accessiblePMID 25948101.
  11. Jump up^ http://www.additudemag.com/adhdblogs/19/11101.html[full citation needed]

Further reading

Liming Shao Patent
  • US application 2007203111, Shao L, Wang F, Malcolm SC, Hewitt MC, Bush LR, Ma J, Varney MA, Campbell U, Engel SR, Hardy LW, Koch P, Campbell JE, “Cycloalkylamines as monoamine reuptake inhibitors”, published 2007-08-30, assigned to Sepracor Inc.
Asymmetry Patent
  • US patent 7129378, Han X, Krishnamurthy D, Senanayake CH, Lu Z-H, “Method of preparing amine stereoisomers”, published 2005-07-28, assigned to Apsinterm LLC
Dasotraline
Dasotraline.svg
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C16H15Cl2N
Molar mass 292.20 g·mol−1
3D model (JSmol)
Patent ID

Title

Submitted Date

Granted Date

US2017266133 METHODS AND COMPOSITIONS OF DASOTRALINE FOR TREATMENT OF ADHD
2015-05-12
US2017266134 DOSAGE OF DASOTRALINE AND METHOD FOR TREATMENT OF ADHD
2015-05-12
Patent ID

Title

Submitted Date

Granted Date

US2015196502 FORMULATIONS, SALTS AND POLYMORPHS OF TRANSNORSERTRALINE AND USES THEREOF
2015-01-26
2015-07-16
US2013065904 Combinations of Eszopiclone and Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-N-Methyl-1-Napthalenamine or Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine, and Methods of Treatment of Menopause and Mood, Anxiety, and Cognitive Disorders
2012-11-09
2013-03-14
US9072699 TREATMENT OF CNS DISORDERS WITH trans 4-(3, 4-DICHLOROPHENYL)-1, 2, 3, 4-TETRAHYDRO-1-NAPTHALENAMINE
2014-01-10
2014-05-08
US8329950 Process for preparation of trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-tetrahydro-1Napthalenamine
2012-01-25
2012-12-11
US2006128993 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine and its formamide
2006-06-15
Patent ID

Title

Submitted Date

Granted Date

US8097760 PREPARATION OF CHIRAL AMIDES AND AMINES
2009-06-11
2012-01-17
US9498452 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-naphthalenamine
2015-06-01
2016-11-22
US8344030 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine
2012-02-03
2013-01-01
US8134029 Treatment of CNS Disorders With trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine
2010-11-18
2012-03-13
US7589237 Treatment of CNS Disorders With trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine
2008-11-13
2009-09-15
Patent ID

Title

Submitted Date

Granted Date

US2012077818 COMPOSITIONS COMPRISING TRANSNORSERTRALINE AND SEROTONIN RECEPTOR 1A AGONISTS/ANTAGONISTS AND USES THEREOF
2010-05-12
2012-03-29
US2016016891 PREPARATION OF CHIRAL AMIDES AND AMINES
2015-05-18
2016-01-21
US2014057990 PREPARATION OF CHIRAL AMIDES AND AMINES
2013-08-01
2014-02-27
US9199946 PYRIMIDINONE CARBOXAMIDE INHIBITORS OF ENDOTHELIAL LIPASE
2013-03-29
2015-03-05
US8524950 Preparation of chiral amides and amines
2011-12-21
2013-09-03
Patent ID

Title

Submitted Date

Granted Date

US2008293726 Combinations of Eszopiclone and Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-N-Methyl-1-Napthalenamine or Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine, and Methods of Treatment of Menopause and Mood, Anxiety, and Cognitive Disorders
2008-11-27
US7105699 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine and its formamide
2006-01-19
2006-09-12
US2017157067 TREATMENT OF CNS DISORDERS WITH trans 4-(3, 4-DICHLOROPHENYL)-1, 2, 3, 4-TETRAHYDRO-1-NAPHTHALENAMINE
2016-10-20
US2014315910 Combinations of Eszopiclone and Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-N-Methyl-1-Napthalenamine or Trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine, and Methods of Treatment of Menopause and Mood, Anxiety, and Cognitive Disorders
2014-06-27
2014-10-23
US2007282007 TREATMENT OF PAIN DISORDERS WITH trans 4-(3, 4-DICHLOROPHENYL)-1, 2, 3, 4-TETRAHYDRO-1-NAPHTHALENAMINE AND ITS FORMAMIDE
2007-12-06
Patent ID

Title

Submitted Date

Granted Date

US8658700 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine
2012-12-04
2014-02-25
US8957114 Formulations, salts and polymorphs of transnorsertraline and uses thereof
2010-12-03
2015-02-17
US7790772 Treatment of CNS Disorders With trans 4-(3, 4-Dichlorophenyl)-1, 2, 3, 4-Tetrahydro-1-Napthalenamine
2009-11-26
2010-09-07
US7423179 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine
2006-09-28
2008-09-09
US7087785 Treatment of CNS disorders with trans 4-(3, 4-dichlorophenyl)-1, 2, 3, 4-tetrahydro-1-napthalenamine and its formamide
2004-05-13
2006-08-08

//////////ダソトラリン, SEP-225289,  SEP-289, DSP-225289, Dasotraline, 675126-05-3, UNII-4D28EY0L5T, (1R,4S)-trans-Norsertraline, Norsertraline

C1CC(C2=CC=CC=C2C1C3=CC(=C(C=C3)Cl)Cl)N

CARMEGLIPTIN………….a DPP-4 inhibitor

Figure

(2S,3S,11βS)-1-(2-Amino-9,10-dimethoxy-1,3,4,6,7,11β-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-(4S)-fluoromethyl-pyrrolidin-2-one Dihydrochloride

(2S,3S,11bS)-1-(2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4(S)-fluoromethyl-pyrrolidin-2-one

813452-14-1 (di-HCl)
916069-91-5 (mono-HCl)

Roche…….innovator

 

CARMEGLIPTIN, 813452-18-5, 结构式

 

CARMEGLIPTIN

813452-18-5

(2S,3S,11βS)-1-(2-Amino-9,10-dimethoxy-1,3,4,6,7,11β-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-(4S)-fluoromethyl-pyrrolidin-2-one

(S)-1-((2S,3S,11bS)-2-amino-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-3-yl)-4-(fluoromethyl)pyrrolidin-2-one

(S)-1-((2S,3S,11bS)-2-amino-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-3-yl)-4-(fluoromethyl)pyrrolidin-2-one
(S)-1-((2S,3S,11bS)-2-amino-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-3-yl)-4-(fluoromethyl)pyrrolidin-2-one
分子式: C20H28FN3O3
分子量: 377

813452-18-5, Carmegliptin, R-1579;carmegliptin, Carmegliptin (USAN/INN), SureCN419289, UNII-9Z723VGH7J, CHEMBL591118, CHEBI:699093, Ro-4876904, D08631, R-1579, B1Q

 

Type 2 diabetes is a chronic, progressive metabolic disease, affecting about 4% of the world population. The main goal of the management of type 2 diabetes is to achieve glycemic control as close to the nondiabetic range as practicable, in order to reduce the risk of late-stage complications.However, the therapeutic effect provided by existing medications is often not sustainable, since the multi-organ defects responsible for the disease are only insufficiently addressed.

Dipeptidyl peptidase-IV (DPP-IV) inhibitors have emerged as a new therapeutic option to treat type 2 diabetes.

Their rapid rise in popularity is due to the favourable safety profile (no hypoglycemia, no weight gain, no gastrointestinal problems—typical side effects associated with established anti-diabetic agents). DPP-IV is a ubiquitous serine protease, the inhibition of which prevents the degradation of glucagon-like peptide 1 (GLP-1). The resulting higher levels of GLP-1 have a beneficial impact on major players involved in the pathogenesis of type 2 diabetes: β-cells, liver, α-cells, gut, and brain.

Long-term studies with DPP-IV inhibitors in patients are underway in order to confirm the safety and sustainability of these effects, and, in particular, their ability to prevent the progressive loss of β-cell function.

 

SYNTHESIS

 

Figure

 

aReagents and conditions: a) HCO2Me, Δ; b) POCl3, MeCN; c) HO2CCH2CO2Et, neat, 120 °C; d) ethyl acrylate, neat; e) t-BuOK, neat (5 steps); f) NH4OAc, MeOH; g) NaBH4, TFA, THF; h) Boc2O, CH2Cl2; i) KOH, aq THF; j) DPPA, Et3N, TMSCH2CH2OH, PhMe, 80 °C; k) Et4NF, MeCN; l) chiral HPLC; m) Et3N, CH2Cl2; n) NaH, DMF; o) HCl, dioxane; p) HCl, 2-PrOH.

 

Full-size image (22 K)

Scheme 2.

Reagents and conditions: (a) NH4OAc, MeOH, rt, 95%; (b) NaBH4, TFA, THF, 0 °C; (c) Boc2O, CH2Cl2, 83% over 2 steps; (d) KOH, aq THF, rt; (e) DPPA, Et3N, 2-(trimethylsilyl)ethanol, toluene, 80 °C; (f) Et4NF, CH3CN, 50 °C, 56% over 3 steps; (g) Et3N, CH2Cl2, (h) NaH, cat. NaI, DMF; (i) HCl, 1,4-dioxane.

 

 

Carmegliptin (2.70) is an anti-diabetes drug which is currently in late stage clinical trials. It represents a further structural advancement from the other existing marketed drugs in this class, sitagliptin (2.71, Januvia) and vildagliptin (2.72, Zomelis, Figure 7). These compounds are all members of the dipeptidyl peptidase 4 class (DPP-4), a transmembrane protein that is responsible for the degradation of incretins; hormones which up-regulate the concentration of insulin excreted in a cell. As DPP-4 specifically cleaves at proline residues, it is unsurprising that the members of this drug class exhibit an embedded pyrrolidine ring (or mimic) and additional decoration (a nitrile or fluorinated alkyl substituent is present in order to reach into a local lipophilic pocket). One specific structural liability of the 2-cyano-N-acylpyrrolidinyl motif (2.73) is its inherent susceptibility towards diketopiperazine formation (2.74, Scheme 29) [80], however, one way to inhibit this transformation is to position a bulky substituent on the secondary amine nucleophile as is the case in vildagliptine (2.72).

[1860-5397-9-265-7]
Figure 7: Structures of DPP-4 inhibitors of the gliptin-type.
[1860-5397-9-265-i29]
Scheme 29: Formation of inactive diketopiperazines from cis-rotameric precursors.

A single crystal X-ray structure of carmegliptin bound in the human DPP-4 active site has been published indicating how the fluoromethylpyrrolidone moiety extends into an adjacent lipophilic pocket [81]. Additional binding is provided by π–π interaction between the aromatic substructure and an adjacent phenylalanine residue as well as through several H-bonds facilitated by the adjacent polar substituents (Figure 8).

 

[1860-5397-9-265-8]
Figure 8: Co-crystal structure of carmegliptin bound in the human DPP-4 active site (PDB 3kwf).

The reported synthesis of carmegliptin enlists a Bischler-Napieralski reaction utilising the primary amine 2.76 and methyl formate to yield the initial dihydroquinoline 2.77 as its HCl salt (Scheme 30) [82]. This compound was next treated with 3-oxoglutaric acid mono ethyl ester (2.78) in the presence of sodium acetate. Decarboxylation then yields the resulting aminoester 2.79 which was progressed through an intramolecular Mannich-type transformation using aqueous formaldehyde to allow isolation of enaminoester 2.80 after treatment of the intermediate with ammonium acetate in methanol.

The next step involves a very efficient crystallisation-induced dynamic resolution of the racemic material using the non-natural (S,S)-dibenzoyl-D-tartaric acid ((+)-DBTA). It is described that the desired (S)-enantiomer of compound 2.81 can be isolated in greater than 99% ee and 93% overall yield. This approach is certainly superior to the original separation of the two enantiomers (at the stage of the final product) by preparative chiral HPLC that was used in the discovery route (albeit it should be noted that both enantiomers were required for physiological profiling at the discovery stage).

Next, a 1,2-syndiastereoselective reduction of enaminoester 2.81 occurs with high diastereocontrol imposed by the convexed presentation of the substrate for the formal conjugate addition and subsequent protonation steps. This is followed by Boc-protection and interconversion of the ethyl ester to its amide derivative 2.82 in 80% overall yield for this telescoped process. The primary amide in 2.82 was then oxidised via a modern variant of the classical Hoffmann rearrangement using phenyliodine diacetate (PIDA).

Following extensive investigation it was found that slowly adding this reagent in a mixture of acetonitrile/water to a suspension of amide 2.82 and KOH gave clean conversion to the amine product in high yield. This new procedure was also readily scalable offering a cleaner, safer and more reliable transformation when compared to other related rearrangement reactions. During a further telescoped procedure amine 2.83 was treated with lactone 2.84 to regenerate the corresponding lactam after mesylate formation. Finally, removal of the Boc-group with aqueous hydrochloric acid furnished carmegliptin as its HCl salt.

 

[1860-5397-9-265-i30]
Scheme 30: Improved route to carmegliptin.
  1. Peters, J.-U. Curr. Top. Med. Chem. 2007, 7, 579–595……………..80
  2. Mattei, P.; Boehringer, M.; Di Gorgio, P.; Fischer, H.; Hennig, M.; Huwyler, J.; Koçer, B.; Kuhn, B.; Loeffler, B. M.; MacDonald, A.; Narquizian, R.; Rauber, E.; Sebokova, E.; Sprecher, U.Bioorg. Med. Chem. Lett. 2010, 20, 1109–1113. doi:10.1016/j.bmcl.2009.12.024………..81
  3. Albrecht, S.; Adam, J.-M.; Bromberger, U.; Diodone, R.; Fettes, A.; Fischer, R.; Goeckel, V.; Hildbrand, S.; Moine, G.; Weber, M. Org. Process Res. Dev. 2011, 15, 503–514. doi:10.1021/op2000207……….82

………………………………………………………………………………………………………………..

Org. Process Res. Dev. 2011, 15, 503–514. doi:10.1021/op2000207

http://pubs.acs.org/doi/full/10.1021/op2000207

 

Abstract Image

A short and high-yielding synthesis of carmegliptin (1) suitable for large-scale production is reported. The tricyclic core was assembled efficiently by a decarboxylative Mannich addition−Mannich cyclization sequence. Subsequent crystallization-induced dynamic resolution of enamine 7 using (S,S)-dibenzoyltartaric acid was followed by diastereoselective enamine reduction to give the fully functionalized tricyclic core with its three stereogenic centers. The C-3 nitrogen was introduced by Hofmann rearrangement of amide 28, and the resulting amine 10was coupled with (S)-fluoromethyl lactone 31. Following cyclization to lactam 13 and amine deprotection, 1 was obtained in 27−31% overall yield with six isolated intermediates.

Preparation of (2S,3S,11βS)-1-(2-Amino-9,10-dimethoxy-1,3,4,6,7,11β-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-(4S)-fluoromethyl-pyrrolidin-2-one Dihydrochloride (1)   CARMEGLIPTIN

A suspension of carbamate 13 (136 kg, 285 mol) in a mixture of H2O (112 kg) and acetone (122 kg) was treated at 50 °C within 60 min with 37% aq HCl (98.0 kg). After 90 min at 47−52 °C the solution was polish filtered through a 5 μm filter. The first reactor and the transfer lines were washed with a hot (47−52 °C) mixture of H2O (13.0 kg) and acetone (116 kg). The filtrate was cooled to 25 °C and treated at this temperature within 80 min with acetone (1600 kg) whereupon the product crystallized out. The resulting suspension was stirred for 1 h at 25 °C and subsequently centrifuged. The crystals were washed in two portions with acetone (391 kg) and dried at 50 °C and <30 mbar until constant weight to afford 122.4 kg (95%) of the title compound as colorless crystals with an assay (HPLC) of 98.8% (w/w).
1H NMR (400 MHz, D2O) δ 2.11−2.22 (m, 1H); 2.45 (dd, J = 17.6 Hz, 6.7 Hz; 1H); 2.76 (dd, J = 17.6 Hz, 9.55 Hz, 1H); 2.90−3.05 (m, 1H); 3.08−3.19 (m, 2H); 3.24−3.36 (m, 1H); 3.43 (dd, J = 9.8 Hz, 5.75 Hz, 1H); 3.49−3.58 (m, 1H); 3.70−3.84 (m, 4H); 3.87 (s, 3H); 3.88 (s, 3H); 4.12 (td, J = 11.6 Hz, 4.5 Hz, 1H); 4.45−4.55 (m, 1H); 4.56−4.68 (m, 3H); 6.91 (s, 1H), 6.95 (s, 1H).
 
 
IR (cm−1): 3237, 2925, 1682, 496, 478.
 
MS (ESI): m/z 378.3 ([M + H]+ (free amine)).
 
Anal. Calcd for C20H30Cl2FN3O3: C, 53.34; H, 6.71; N, 9.33; Cl, 15.74; F 4.22; O, 10.66. Found: C, 53.04; H, 6.43; N, 9.45; Cl, 15.66; F, 4.29; O, 11.09.
REF FOR ABOVE
Mattei, P.; Böhringer, M.; Di Giorgio, P.; Fischer, H.; Hennig, M.; Huwyler, J.; Kocer, B.; Kuhn, B.; Löffler, B. M.; MacDonald, A.; Narquizian, R.; Rauber, E.; Sebokova, E.; Sprecher, U. Bioorg. Med. Chem. Lett. 2010, 20, 1109

Böhringer, M.; Kuhn, B.; Lübbers, T.; Mattei, P.; Narquizian, R.; Wessel,H. P. (F. Hoffmann-La Roche AG). U.S. Pat. Appl. 2004/0259902, 2004.
…………………………………………………..
Discovery of carmegliptin: A potent and long-acting dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes
Bioorg Med Chem Lett 2010, 20(3): 1109
 http://www.sciencedirect.com/science/article/pii/S0960894X09017296

  • Discovery of carmegliptin: A potent and long-acting dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes

  • Pages 1109-1113
  • Patrizio Mattei, Markus Boehringer, Patrick Di Giorgio, Holger Fischer, Michael Hennig, Joerg Huwyler, Buelent Koçer, Bernd Kuhn, Bernd M. Loeffler, Alexander MacDonald, Robert Narquizian, Etienne Rauber, Elena Sebokova, Urs Sprecher
  • image
    Full-size image (16 K)

    Scheme 3.

    Reagents and conditions: (a) preparative HPLC (Chiralpak® AD column), heptane/2-propanol 85:15, 37% (b) BH3.Me2S, THF, 0 °C; (c) (MeOCH2CH2)2NSF3, CH2Cl2, 67% (2 steps); (d), SOCl2, ZnCl2, 80 °C, 72 h, 61%; (e) Et3N, CH2Cl2; (f) NaH, DMF, 56% (2 steps); (g) HCl, 1,4-dioxane, 91%; (h) HCl, 2-propanol, 86%.

 The synthesis of 8p is outlined ABOVE and required the enantiopure building blocks (S,S,S)-5 and 12. (S,S,S)-5 was obtained from the racemate by preparative chiral HPLC. Acid chloride 12 was prepared starting from (S)-paraconic acid (9).  Reduction of 9 with borane–dimethyl sulfide complex afforded hydroxymethyl lactone 10. Since 10 is known to racemise rather readily,  it was immediately treated with bis(2-methoxyethyl)aminosulfur trifluoride,  thereby affording fluoromethyl lactone 11. This was converted to 12 by reaction with thionyl chloride in the presence of zinc chloride.  The (S)-4-fluoromethyl-pyrrolidinone 8p was isolated as the dihydrochloride salt, a highly water soluble white crystalline solid, mp >275 °C.
…………………………………………………….
US 2013109859

The most preferred product is (2S,3S,11bS)-2-tert.-Butoxycarbonylamino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H pyrido[2,1-a]isoquinoline-3-carboxylic acid amide having the following structure:

It has been found that during the amidation of the ester epimerization takes place at position 3 and thus the 3R-epimer of the formula IVb is transformed to a larger extent in the 3S-epimer of formula V.

 

e) Preparation of (2S,3S,11bS)-1-(2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4(S)-fluoromethyl-pyrrolidin-2-one Dihydrochloride

A 2.5 L reactor equipped with a mechanical stirrer, a Pt-100 thermometer, a dropping funnel and a nitrogen inlet was charged with 619 g (1.30 mol) of (2S,3S,11bS)-3-((4S)-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester, 4.2 L isopropanol and 62 mL water and the suspension was heated to 40-45° C. In a second vessel, 1.98 L isopropanol was cooled to 0° C. and 461 mL (6.50 mol) acetyl chloride was added during 35 min, maintaining the temperature at 0-7° C. After completed addition, the mixture was allowed to reach ca. 15° C. and was then slowly added to the first vessel during 1.5 h. After completed addition the mixture was stirred for 18 h at 40-45° C., whereas crystallization started after 1 h. The white suspension was cooled to 20° C. during 2 h, stirred at that temperature for 1.5 h and filtered. The crystals were washed portionwise with 1.1 L isopropanol and dried for 72 h at 45° C./20 mbar, to give 583 g of the product as white crystals (100% yield; assay: 99.0%).

…………………………………………………….
US 2008071087
Figure US20080071087A1-20080320-C00035
(2S,3S,11bS)-(3-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)]-carbamic acid tert-butyl ester (8)
Example 8
Transformation of (2S,3S,11bS)-(3-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl) ]-carbamic acid tert-butyl ester into (S)-1-((2S,3S,11bS)-2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl) -4-fluoromethyl-pyrrolidin-2-one.a)
Preparation of 4-fluoromethyl-5H-furan-2-oneA 6 L reactor equipped with a mechanical stirrer, a Pt-100 thermometer, a dropping funnel and a nitrogen inlet was charged with 500 g (4.38 mmol) 4-hydroxymethyl-5H-furan-2-one and 2.0 L dichloromethane. The solution was cooled to −10° C. and 1.12 kg (4.82 mol) bis-(2-methoxyethyl)aminosulfur trifluoride (Deoxo-Fluor) was added during 50 min, maintaining the temperature at −5 to −10° C. with a cooling bath. During the addition a yellowish emulsion formed, which dissolved to an orange-red solution after completed addition. This solution was stirred for 1.5 h at 15-20° C., then cooled to −10° C. A solution of 250 ml water in 1.00 L ethanol was added during 30 min, maintaining the temperature between −5 and −10° C., before the mixture was allowed to reach 15-20° C. It was then concentrated in a rotatory evaporator to a volume of ca. 1.6 L at 40° C./600-120 mbar. The residue was dissolved in 2.0 L dichloromethane and washed three times with 4.0 L 1N hydrochloric acid. The combined aqueous layers were extracted three times with 1.4 L dichloromethane. The combined organic layers were evaporated in a rotatory evaporator to give 681 g crude product as a dark brown liquid. This material was distilled over a Vigreux column at 0.1 mbar, the product fractions being collected between 71 and 75° C. (312 g). This material was re-distilled under the same conditions, the fractions being collected between 65 and 73° C., to give 299 g 4-fluoromethyl-5H-furan-2-one (58% yield; assay: 99%).MS: m/e 118 M+, 74,59,41.b) Preparation of (S)-4-fluoromethyl-dihydro-furan-2-oneA 2 L autoclave equipped with a mechanical stirrer was charged with a solution of 96.0 g 4-fluoromethyl-5H-furan-2-one (8.27×10−1 mol) in 284 mL methanol. The autoclave was sealed and pressurized several times with argon (7 bar) in order to remove any traces of oxygen. At ˜1 bar argon, a solution of 82.74 mg Ru(OAc)2((R)-3,5-tBu-MeOBlPHEP) (6.62×10−5 mol) (S/C 12500) in 100 mL methanol was added under stirring from a catalyst addition device previously charged in a glove box (O2 content <2 ppm) and pressurized with argon (7 bar). The argon atmosphere in the autoclave was replaced by hydrogen (5 bar). At this pressure, the reaction mixture was stirred (˜800 rpm) for 20 h at 30° C. and then removed from the autoclave and concentrated in vacuo. The residue was distilled to afford 91.8 g (94%) (S)-4-fluoromethyl-dihydro-furan-2-one. The chemical purity of the product was 99.7% by GC-area.c) Preparation of (2S,3S,11bS)-3-(3-Fluoromethyl-4-hydroxy-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl esterA 1.5 L reactor equipped with a mechanical stirrer, a Pt-100 thermometer, a dropping funnel and a nitrogen inlet was charged with 50 g (128 mmol) (2S,3S,11bS)-3-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)-carbamic acid tert-butyl ester, 500 mL toluene and 2.51 g (25.6 mmol) 2-hydroxypyridine. To this slightly brownish suspension, 22.7 g (192 mmol) of (S)-4-fluoromethyl-dihydro-furan-2-one was added dropwise at RT. No exothermy was observed during the addition. The dropping funnel was rinsed portionwise with totally 100 mL toluene. The suspension was heated to reflux, whereas it turned into a dear solution starting from 60° C., after 40 min under reflux a suspension formed again. After totally 23 h under reflux, the thick suspension was cooled to RT, diluted with 100 mL dichloromethane and stirred for 30 min at RT. After filtration, the filter cake was washed portionwise with totally 200 mL toluene, then portionwise with totally 100 mL dichloromethane. The filter cake was dried at 50° C./10 mbar for 20 h, to give 60.0 g product (94% yield; assay: 100%).

MS: m/e 496 (M+H)+, 437.

d) Preparation of (2S,3S,11bS)-3-((4S)-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl esterA 1.5 L reactor equipped with a mechanical stirrer, a Pt-100 thermometer, a dropping funnel, a cooling bath and a nitrogen inlet was charged with 28 g (56.5 mmol) of (2S,3S,11bS)-3-(3-fluoromethyl-4-hydroxy-butyrylamino) -9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester and 750 mL THF. The mixture was cooled to 0° C. and a solution of 6.17 mL (79 mmol) methanesulfonic acid in 42 mL THF was added during 10 min, maintaining the temperature at 0-5° C. At 0° C. a solution of 12.6 mL (90.2 mmol) triethylamine in 42 mL THF was added during 15 min. The resulting suspension was stirred for 80 min at 0-5° C., whereas it became gradually thicker. Then 141 mL (141 mmol) 1 M lithium-bis(trimethylsilyl)amide were added to the mixture during 15 min, whereas the suspension dissolved. The solution was allowed to reach RT during 60 min under stirring. 500 mL water was added without cooling, the mixture was extracted and the aqueous phase was subsequently extracted with 500 mL and 250 mL dichloromethane. The organic layers were each washed with 300 mL half saturated brine, combined and evaporated on a rotatory evaporator. The resulting foam was dissolved in 155 mL dichloromethane, filtered and again evaporated to give 30.5 g crude product as a slightly brownish foam. This material was dissolved in 122 mL methanol, resulting in a thick suspension, which dissolved on heating to reflux. After 20 min of reflux the solution was allowed to gradually cool to RT during 2 h, whereas crystallization started after 10 min. After 2 h the suspension was cooled to 0° C. for 1 h, followed by −25° C. for 1 h. The crystals were filtered off via a pre-cooled glass sinter funnel, washed portionwise with 78 mL TBME and dried for 18 h at 45° C./20 mbar, to give 21.0 g of the title product as white crystals (77% yield; assay: 99.5%).

MS: m/e 478 (M+H)+, 437, 422.

e) Preparation of (2S,3S,11bS)-1-(2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4(S)-fluoromethyl-pyrrolidin-2-one dihydrochlorideA 2.5 L reactor equipped with a mechanical stirrer, a Pt-100 thermometer, a dropping funnel and a nitrogen inlet was charged with 619 g (1.30 mol) of (2S,3S,11bS)-3-((4S)-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester, 4.2 L isopropanol and 62 mL water and the suspension was heated to 40-45° C. In a second vessel, 1.98 L isopropanol was cooled to 0° C. and 461 mL (6.50 mol) acetyl chloride was added during 35 min, maintaining the temperature at 0-7° C. After completed addition, the mixture was allowed to reach ca. 15° C. and was then slowly added to the first vessel during 1.5 h. After completed addition the mixture was stirred for 18 h at 40-45° C., whereas crystallization started after 1 h. The white suspension was cooled to 20° C. during 2 h, stirred at that temperature for 1.5 h and filtered. The crystals were washed portionwise with 1.1 L isopropanol and dried for 72 h at 45° C./20 mbar, to give 583 g of the product as white crystals (100% yield; assay: 99.0%).

These compounds are useful intermediates for the preparation of DPP-IV inhibitors as disclosed in PCT International Patent Appl. WO 2005/000848. More preferably, the invention relates to a process for the preparation of (2S,3S,11bS)-(3-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)]-carbamic acid tert-butyl ester.

 

XXXXXXX

According to still another embodiment (Scheme 2, below) the (S)-4-fluoromethyl-dihydro-furan-2-one (VII) is directly coupled with the amino-pyrido[2,1-a]isoquinoline derivative (VI) to form the hydroxymethyl derivative of the pyrido[2,1-a]isoquinoline (VIII), which is then subsequently cyclized to the fluoromethyl-pyrrolidin-2-one derivative (IX). The latter can be deprotected to yield the desired pyrido[2,1-a]isoquinoline derivative (I).

In a further preferable embodiment, the process for the preparation of (S)-1-((2S,3S,11bS)-2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one or of a pharmaceutically acceptable salt thereof comprises the subsequent steps:

  • e) coupling of the (2S,3S,11bS)-3-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)-carbamic acid tert-butyl ester (amine of formula VI, wherein R2 and R3 are methoxy, R4 is hydrogen and Prot is Boc) with the (S)-4-fluoromethyl-dihydro-furan-2-one of formula
  • f) cyclization of the obtained (2S,3S,11bS)-3-(3-fluoromethyl-4-hydroxy-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester in the presence of a base, and
  • g) deprotecting the obtained (2S,3S,11bS)-3-((4S)-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester.

………………………………………………………….

PATENT

 

http://www.google.com.ar/patents/US7122555?cl=pt-PT

 

Example 23

RACEMIC

1-((RS,RS,RS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one

a) 4-Fluoromethyl-dihydro-furan-2-one

A solution of 4-hydroxymethyl-dihydro-furan-2-one (Tetrahedron 1994, 50, 6839; 1.02 g, 8.78 mmol) and bis(2-methoxyethyl)aminosulfur trifluoride (3.88 g, 17.6 mmol) in chloroform (4.4 mL) was stirred at 40° C. for 1 h, then poured onto ice and partitioned between sat. aq. sodium hydrogencarbonate solution and dichloromethane. The organic layer was washed with brine, dried (MgSO4), and evaporated. Chromatography (SiO2, heptane-ethyl acetate gradient) afforded the title compound (576 mg, 56%). Colourless liquid, MS (EI) 118.9 (M+H)+.

b) 3-Chloromethyl-4-fluoro-butyryl chloride

A mixture of 4-fluoromethyl-dihydro-furan-2-one (871 mg, 7.37 mmol), thionyl chloride (4.39 g, 36.9 mmol), and zinc chloride (60 mg, 0.44 mmol) was stirred 72 h at 80° C., then excess thionyl chloride was removed by distillation. Kugelrohr distillation of the residue (85° C., 0.2 mbar) afforded the title compound (450 mg, 35%). Colourless liquid, 1H-NMR (300 MHz, CDCl3): 4.65–4.55 (m, 1H), 4.50–4.40 (m, 1H), 3.70–3.60 (m, 2H), 3.25–3.05 (m, 2H), 2.80–2.60 (m, 1H).

c) (RS,RS,RS)-[3-(3-Chloromethyl-4-fluoro-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester

The title compound was produced in accordance with the general method of Example 5c from (RS,RS,RS)-(3-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)-carbamic acid tert-butyl ester (Example 5b) and 3-chloromethyl-4-fluoro-butyryl chloride. White solid, MS (ISP) 514.5 (M+H)+.

d) (RS,RS,RS)-[3-(4-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester

The title compound was produced in accordance with the general method of Example 5d from (RS,RS,RS)-[3-(3-chloromethyl-4-fluoro-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester. Off-white foam, MS (ISP) 478.5 (M+H)+.

e) 1-((RS,RS,RS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one

The title compound was produced in accordance with the general method of Example 1e from (RS,RS,RS)-[3-(4-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester. Light yellow oil, MS (ISP) 378.5 (M+H)+.
Examples 28 and 29

(SR)-1-((RS,RS,RS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one

Figure US07122555-20061017-C00040 UNDESIRED

and

 

(RS,RS,RS,RS)-1-(2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one

The title compounds were produced from 1-((RS,RS,RS)-2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one (Example 23) by chromatographic separation (SiO2, CH2Cl2/MeOH/NH4OH 80:1:0.2, then 95:5:0.25).

(SR)-1-((RS,RS,RS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one: Yellow oil, Rf=0.45 (CH2Cl2/MeOH/NH4OH 90:10:0.25).

(RS,RS,RS,RS)-1-(2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one: Light yellow solid, Rf=0.40 (CH2Cl2/MeOH/NH4OH 90:10:0.25).

Example 30

(S)-1-((S,S,S)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one Dihydrochloride

Figure US07122555-20061017-C00042 DESIRED

a) [(S,S,S)-3-(3-Chloromethyl-4-fluoro-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester

The title compound was produced in accordance with the general method of Example 5c from (S,S,S)-(3-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl)-carbamic acid tert-butyl ester (Example 16b) and 3-chloromethyl-4-fluoro-butyryl chloride (Example 23b). Off-white solid.

b) [(S,S,S)-3-((S)-4-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester and [(S,S,S)-3-((R)-4-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester

Sodium hydride (55–65% dispersion in oil, 1.14 g, 28.5 mmol) was added to a suspension of [(S,S,S)-3-(3-chloromethyl-4-fluoro-butyrylamino)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester (6.72 g, 13.1 mmol) in N,N-dimethylformamide (95 mL) at r.t., then after 1 h the reaction mixture was poured onto ice and partitioned between ethyl acetate and water. The organic layer was washed with brine, dried (MgSO4), and evaporated. Chromatography (SiO2, cyclohexane/2-propanol 4:1) afforded [(S,S,S)-3-((S)-4-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester (2.40 g, 38%) and the epimer, [(S,S,S)-3-((R)-4-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester (2.73 g, 44%).

[(S,S,S)-3-((S)-4-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester: Light yellow foam, Rf=0.6 (SiO2, cyclohexane/2-propanol 1:1).

[(S,S,S)-3-((R)-4-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester: Light yellow foam, Rf=0.4 (SiO2, cyclohexane/2-propanol 1:1).

    • c) (S)-1-((S,S,S)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one dihydrochloride

[(S,S,S)-3-((S)-4-Fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester (2.40 g, 5.02 mmol) was converted to (S)-1-((S,S,S)-2-amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one in accordance with the general method of Example 1e. The product was dissolved in 2-propanol (10 mL) and treated with hydrogen chloride (5–6 M in 2-propanol, 37 mL). The suspension formed was stirred for 64 h at r.t., then the precipitate was collected by filtration and dried, to afford the title compound (2.04 g, 91%). White solid, m.p. >300° C.

Example 31(R)-1-((S,S,S)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one dihydrochloride

Figure US07122555-20061017-C00043 UNDESIRED

The title compound was produced in accordance with the general method of Example 30c from [(S,S,S)-3-((R)-4-fluoromethyl-2-oxo-pyrrolidin-1-yl)-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido [2,1-a]isoquinolin-2-yl]-carbamic acid tert-butyl ester (Example 30b). White solid, m.p. >300° C.

 

DR ANTHONY MELVIN CRASTO

ANTHONY MELVIN CRASTO

MY BLOGS ON MED CHEM

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
アンソニー     安东尼   Энтони    안토니     أنتوني
blogs are
 

 MY CHINA, VIETNAM  AND JAPAN BLOGS

http://me.zing.vn/u/amcrasto

ICELAND, RUSSIA, ARAB

BOBRDOBRBLAND ICELAND100zakladokadfty

GROUPS

you can post articles and will be administered by me on the google group which is very popular across the world

OPD GROUPSPACESSCOOP OCIorganic-process-development GOOGLE, TVINX, MENDELEY WDT, SCIPEOPLE OPD, EPERNICUS OPDSYNTHETIC ORGANIC CHEMISTRYLinkedIn group, DIIGO OPD, LINKEDIN OPD, WDT LINKEDIN, WDTI ZING

shark

Albaconazole

Chemical structure for albaconazole

Albaconazole

Also known as: UNII-YDW24Y8IAB; UR-9825; 187949-02-6; UR 9825, W-0027
Molecular Formula: C20H16ClF2N5O2   Molecular Weight: 431.823146
(1R,2R)-7-chloro-3-[2-(2,4-difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]quinazolin-4(3H)-one

7-chloro-3-[(2R,3R)-3-(2,4-difluorophenyl)-3-hydroxy-4-(1,2,4-triazol-1-yl)butan-2-yl]quinazolin-4-one

Albaconazole (UR-9825) is a triazole antifungal. It has potential broad-spectrum activity.

Albaconazole is a broad-spectrum antifungal agent being evaluated in phase II clinical trials by Stiefel for the oral treatment of fungal infections, including toenail fungus, distal onychomycosis and subungual onychomycosis. Early clinical trials for the treatment of tinea pedis have been completed. In September 2005, Uriach, originator of albaconazole, granted Stiefel exclusive rights to develop and market albaconazole on a worldwide basis. In November 2006, Uriach’s R&D pipeline was transferred to Palau Pharma, a newly-created spin-out company. Under the terms of the agreement with Stiefel, Palau retains rights as comarketing partner in some European countries. In August 2013, Palau Pharma granted worldwide rights to Actavis. A triazole, albaconazole, has shown potent activity against a broad range of organisms, including pathogens resistant to other antifungals, such as fluconazole or itraconazole. It will be developed as an oral and topical formulation, and is expected to be available to the medical community for a variety of dermatological indications and fungal infections, including vulvovaginal candidiasis.

Albaconazole
Albaconazole.svg
Systematic (IUPAC) name
7-Chloro-3-[(2R,3R)-3-(2,4-difluorophenyl)-3-hydroxy-4-(1,2,4-triazol-1-yl)butan-2-yl]quinazolin-4-one
Clinical data
Identifiers
CAS number 187949-02-6 Yes
ATC code None
PubChem CID 208952
ChemSpider 181045 Yes
UNII YDW24Y8IAB Yes
KEGG D09702 Yes
ChEMBL CHEMBL298817 Yes
Chemical data
Formula C20H16ClF2N5O2 
Mol. mass 431.823146 g/mol
11-26-2003
Method for preparing pyrimidone derivatives with antifungal activity

 

The condensation of the chiral oxazolidinone (I) with 2,4-difluorophenacyl bromide (II) by means of NaHMDS in THF/Et2 O gives the chiral oxirane (III), which is treated with LiOH and H2O2 to eliminate the chiral auxiliary, yielding the carboxylic acid (IV). The cleavage of the oxirane ring of (IV) with 1,2,4-triazole (V) and NaH in hot DMF affords the chiral hydroxyacid (VI), which is submitted to Curtius rearrangement by means of DPPA in hot pyridine to provide the chiral oxazolidinone (VII). The cleavage of the oxazolidinone ring of (VII) by means of refluxing aq. HCl gives the chiral aminoalcohol (VIII), which is condensed with 2-amino-4-chlorobenzoic acid (IX) by means of DCC and HOBt to yield the corresponding amide (X). Finally, this compound is cyclized to the target quinazolinone by reaction with triethyl orthoformate in hot dioxane/NMP.

The condensation of the chiral oxazolidinone (I) with 2,4-difluorophenacyl bromide (II) by means of NaHMDS in THF/Et2 O gives the chiral oxirane (III), which is treated with LiOH and H2O2 to eliminate the chiral auxiliary, yielding the carboxylic acid (IV). The cleavage of the oxirane ring of (IV) with 1,2,4-triazole (V) and NaH in hot DMF affords the chiral hydroxyacid (VI), which is submitted to Curtius rearrangement by means of DPPA in hot pyridine to provide the chiral oxazolidinone (VII). The cleavage of the oxazolidinone ring of (VII) by means of refluxing aq. HCl gives the chiral aminoalcohol (VIII), which is condensed with 2-amino-4-chlorobenzoic acid (IX) by means of DCC and HOBt to yield the corresponding amide (X). Finally, this compound is cyclized to the target quinazolinone by reaction with triethyl orthoformate in hot dioxane/NMP.

EP 0783501; ES 2107376; ES 2120885; JP 1998508317; US 5807854; WO 9705130

…………………………………………..

 

The condensation of (R)-lactic acid (I) with morpholine (II) gives the corresponding morpholide (III), which is protected at the hydroxyl position with dihydropyran (IV) to yield the tetrahydropyranyl ether (V). The Grignard reaction of (V) with 2,4-difluorophenylmagnesium bromide (VI) affords the chiral 1-propanone (VII), which by a Corey’s diastereoselective epoxidation with trimethylsulfoxonium iodide is converted into the oxirane (VIII). The opening of the oxirane ring of (VIII) by means of 1,2,4-triazole (IX) and NaH provides the tertiary alcohol (X), which is treated with pyridine p-toluenesulfonate to give the deprotected diol (XI) as a (2R,3R) and (2R,3S) 4:1 diastereomeric mixture, from which the desired (2R,3R)-isomer (XII) was isolated by crystallization. The reaction of (XII) with Ms-Cl and TEA, followed by cyclization with NaOMe, yields the oxirane (XIII), which is finally condensed with 7-chloroquinazolin-4(3H)-one (XIV) by means of K2CO3 in hot NMP.

 

ES 2159488; WO 0166519

 

…………………………………………….

Alternatively, intermediate (XIII) can be obtained as follows: Heating of ethyl (S)-lactate (XIV) with morpholine affords amide (XVI), which then reacts with 3,4-dihydro-2H-pyran (A) in the presence of p-TsOH to give protected derivative (XVII). Grignard reaction between (XVII), bromo derivative (XVIII) and Mg turnings in THF yields protected ketone (XIX), which is treated with pyridinium p-toluenesulfonate (PPTS) (THP group removal) and reprotected by means of Tf2O and DIEA to give triflate derivative (XX). Conversion of (XX) into intermediate (XIII) is achieved by reaction with triazolone (VII) and NaH in THF.

Chem Pharm Bull 1993,41(6),1035-42

……………………………………

Alternatively, derivative (XXIX) can be obtained in an analogous way as its enantiomer (XIX). Diastereoselective epoxidation of (XXIX) with trimethylsulfoxonium iodide and NaH in DMSO provides oxirane (XXX) (3). THP group removal by means of PPTS in EtOH, followed by reaction with 3,5-dinitrobenzoyl chloride (XXXI) and NaHCO3 in CH2Cl2, yields a diastereomeric mixture from which dinitrobenzoate derivative (2R,3R)-(XXXII) is obtained by recrystallization (1). Hydrolysis of (2R,3R)-(XXXII) in MeOH by treatment with aqueous NaOH gives compound (2R,3R)-(XXXIII), which is converted into ester (2R,3S)-(XXXIV) by Mitsunobu reaction with benzoic acid, Ph3P and DEAD in THF. Subsequent debenzoylation of (2R,3S)-(XXXIV) with NaOMe in MeOH affords oxiranyl ethanol derivative (2R,3R)-(XXXV), which is first converted into its triflate derivative by means of Tf2O and DIEA in CH2Cl2, and then into triazolone derivative (2S,3R)-(XXXVI) by reaction with intermediate (VII) and NaH in CH2Cl2/DMF. Finally, oxirane derivative (2S,3R)-(XXXVI) reacts with triazole (XXVI) and NaH in DMF to furnish the desired product.

…………………………………………………..

ER-30346 is synthesized by thiazole ring formation of (2R,3R)-3-(2,4-difluorophenyl)-3-hydroxy-2-methyl-4-(1H-1,2,4-triazol-1-yl)thiobutanamide (I) and 4-bromoacetylbenzonitrile (II) by means of reflux in methanol. The thioamide (I) is obtained with excellent yield from a chiral nitrile (III) by heating with diethyl dithiophosphate in aqueous medium.

…………………………………………….

The nitrile (III), a chiral key intermediate of this synthesis, can be obtained by two different synthetic routes as follows: Route-a: The key step of this route is ring opening reaction of the trisubstituted oxirane (VII) by cyanide anion leading to the nitrile (III). The chiral oxirane (VII) is synthesized from (R)-lactic acid derivatives as already reported. The reaction of (VII) with diethylaluminum cyanide in toluene or lithium cyanide in tetrahydrofuran gives the nitrile (III) with high yield without any epimerization reaction.

…………………………………………..

The nitrile (III), a chiral key intermediate of this synthesis, can be obtained by two different synthetic routes as follows: Route-b: The starting material of this route is methyl (S)-3-hydroxy-2-methylpropionate (VIII), which contains one additional carbon between the hydroxyl group and the 2-position carbon of (R)-lactate, the starting material of route-a. The hydroxyl group of (VIII) is protected by triphenylmethyl group. Then, 2,4-difluorophenyl moiety is introduced to give the ketone (X). Direct conversion of the ketone (X) to the oxirane (XIV) by dimethylsulfoxonium methylide, the same condition for compound (IV) in route-a, does not proceed. The oxirane (XIV) having desired stereochemistry is obtained via oxidation reaction. The ketone (X) is converted to the exomethylene (XI) by Wittig reaction. The stereoselective oxidation of (XI) is achieved by means of osmium tetroxide in the presence of 4-methylmorpholine N-oxide to give the diol (XII) in 58% yield after separation of its epimer by column chromatography. After methanesulfonylation of the primary alcohol of (XII), a triazole moiety is introduced and the triphenylmethyl group is deprotected. Then, the primary hydroxyl group of (XVI) is oxidized under Swern oxidation condition to give the aldehyde (XVII), which is converted to the chiral nitrile intermediate (III) by means of heating with hydroxylamine-O-sulfonic acid.

 

………………………………..

J. Med. Chem., 1998, 41 (11), pp 1869–1882
DOI: 10.1021/jm9707277

A series of azole antifungal agents featuring a quinazolinone nucleus have been subjected to studies of structure−activity relationships. In general, these compounds displayed higher in vitro activities against filamentous fungi and shorter half-lives than the structures described in our preceding paper. The most potent products in vitro carried a halogen (or an isostere) at the 7-position of the quinazolinone ring. Using a murine model of systemic candidosis, oral activity was found to be dependent on hydrophobicity, which, in turn, modulated the compound’s half-life. The 7-Cl derivative, (1R,2R)-7-chloro-3-[2-(2,4-difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]quinazolin-4(3H)-one (20, UR-9825), was selected for further testing due to its high in vitro activity, low toxicity, good pharmacokinetic profile, and ease of obtention. Compound 20 is the (1R,2R) isomer of four possible stereoisomers. The other three isomers were also prepared and tested. The enantiomer (1S,2S) and the (1R,2S) epimer were inactive, whereas the (1S,2R) epimer retained some activity. In vitro 20 was superior to fluconazole, itraconazole, SCH-42427, and TAK-187 and roughly similar to voriconazole and ER-30346. In vivo, 20 was only moderately active in a mouse model of systemic candidosis when administration was limited to the first day. This was attributed to its short half-life in that species (t1/2 = 1 h po). Protection levels comparable to or higher than those of fluconazole, however, were observed in systemic candidosis models in rat and rabbit, where the half-life of the compound was found to be 6 and 9 h, respectively. Finally, 20 showed excellent protection levels in an immunocompromised rat model of disseminated aspergillosis. The compound showed low toxicity signs when administered to rats at 250 mg/kg qd or at 100 mg/kg bid during 28 days.

 

The condensation of the chiral oxazolidinone (I) with 2,4-difluorophenacyl bromide (II) by means of NaHMDS in THF/Et2 O gives the chiral oxirane (III), which is treated with LiOH and H2O2 to eliminate the chiral auxiliary, yielding the carboxylic acid (IV). The cleavage of the oxirane ring of (IV) with 1,2,4-triazole (V) and NaH in hot DMF affords the chiral hydroxyacid (VI), which is submitted to Curtius rearrangement by means of DPPA in hot pyridine to provide the chiral oxazolidinone (VII). The cleavage of the oxazolidinone ring of (VII) by means of refluxing aq. HCl gives the chiral aminoalcohol (VIII), which is condensed with 2-amino-4-chlorobenzoic acid (IX) by means of DCC and HOBt to yield the corresponding amide (X). Finally, this compound is cyclized to the target quinazolinone by reaction with triethyl orthoformate in hot dioxane/NMP.

J Med Chem 1998,41(11),1869

http://pubs.acs.org/doi/abs/10.1021/jm9707277

 (1R,2R)-7-Chloro-3-[2-(2,4-difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]quinazolin-4(3H)-one (20, UR-9825). Precipitated from EtOH/H2O (66% yield from amine 11):  white amorphous solid;

mp 93−110 °C (wide range);

IR (KBr) ν 1675, 1601, 1554, 1498 cm-1;

1H NMR (300 MHz, CDCl3) 8.58 (s, 1H, NCH-N), 8.26 (d, J = 8.6, 1H, arom), 8.11 (d, J = 5.7, trace rotamer), 7.76 (s, 2H, triazol),

7.74 (d, J = 5.3, 1H, arom), 7.5 (m, 2H, arom), 7.10 (s, trace rotamer), 6.9−6.7 (m, 2H, arom),

5.91 (dq, Jd = 2, Jq = 7, 1H, MeCH), 5.54 (d, J = 2, 1H, OH),

5.15 (d, J = 14.2 1H, CH(H)), 4.9−4.7 (m, trace rotamer), 4.30 (d, trace rotamer), 3.99 (d, J = 14.2, 1H, CH(H)),

1.46 (d, J = 6.9, trace rotamer), 1.29 (d, J = 7, 3H, CHMe);

GC−MS 224 (Tr-CH2COHAr, C10H8F2N3O), 208 (group N-ethylheterocycle, C10H9ClN2O);

[α]D −8.0° (c 1, CHCl3).

Chiral HPLC (column, CicloBond SN 1; eluent, MeOH: Et3NHOAc in H2O at pH7 1:1; retention times:  (S,S) (74) tR 12.6 min; (R,R) (20) tR 13.7 min). Area ratio:  0.01:99.99.

Anal. (C20H16ClF2N5O2) C, H, N. 

 

 

KEY
Albaconazole,UNII-YDW24Y8IAB, UR-9825, UR 9825, W-0027
Figure

Tecadenoson…………Atrial Fibrillation

Tecadenoson

 

Tecadenoson
CAS : 204512-90-3
N-[(3R)-Tetrahydro-3-furanyl]adenosine
(2R,3S,4R,5R)-2-(hydroxymethyl)-5-[6-[[(3R)-oxolan-3-yl]amino]purin-9-yl]oxolane-3,4-diol
 
Manufacturers’ Codes: CVT-510
UNII-GZ1X96601Z; AC1L4KMO;
Molecular Formula: C14H19N5O5
Molecular Weight: 337.33
Percent Composition: C 49.85%, H 5.68%, N 20.76%, O 23.71%
Therap-Cat: Antiarrhythmic.
 
Tecadenoson is a novel selective A1 adenosine receptor agonist that is currently being evaluated for the conversion of paroxysmal supraventricular tachycardia (PSVT) to sinus rhythm. It is being developed by CV Therapeutics, Inc.
 
Tecadenoson is an adenosine A1 agonist which had been in phase II clinical evaluation by Gilead Sciences for treatment of atrial fibrillation. The company was also conducting phase III clinical trials for the treatment of paroxysmal supraventricular tachycardia (PSVT); however, no recent developments have been reported for these indications.
Due to the fact that tecadenoson selectively stimulates the A1 receptor and slows electrical impulses in the heart’s conduction system without significantly stimulating the A2 receptor, the intravenous administration of CVT-510 may hold potential for rapid intervention in the control of atrial arrhythmias without lowering blood pressure.
 
 
 
 
 
 
 
 
The reaction of 3-tetrahydrofuroic acid (I) with diphenyl phosphoryl azide (DPPA) in refluxing dioxane gave the intermediate isocyanate (II), which was treated with benzyl alcohol (III) to yield carbamate (IV). Subsequent hydrogenolysis in the presence of Pd/C afforded racemic amine (V), which was resolved by treatment with S-(+)-10-camphorsulfonyl chloride (VI) in pyridine, followed by column chromatography and recrystallization from acetone of the resulting sulfonamide (VII). Then, hydrolysis in HCl-AcOH provided the S-amine (VIII). Condensation of amine (VIII) with 6-chloropurine riboside (IX) in the presence of triethylamine in refluxing MeOH furnished the title compound.
 
 
EP 0920438; EP 0992510; JP 2000501426; US 5789416; WO 9808855
……………………………
 
 
 
 
 
 
 
 
………………………….
 

CVT-510 (tecadenoson) has chemical structure (8 :

Figure imgf000011_0002
 
 
…………………………………….
 
Compound I can be prepared through reaction of the corresponding primary amino compound, R1NH2, through heating with commercially available 6-chloroadenosine in the appropriate solvent (e.g. n-butanol, dimethylformamide, and ethanol). The primary amino compound, R1NH2, is either commercially available or can be prepared as previously described (International Patent Application WO 98/08855).
 
Figure US06576619-20030610-C00008
 
 ……………………………
 
 
 

EXAMPLE 1

The compounds of this invention may be prepared by conventional methods of organic chemistry. The reaction sequence outlined below, is a general method, useful for the preparation of compounds of this invention.

According to this method, oxacycloalkyl carboxylic acid is heated in a mixture of dioxane, diphenylphosphoryazide and triethylamine for 1 hour. To this mixture is added benzyl alcohol and the reaction is further heated over night to give intermediate compound 1. Compound 1 is dissolved in methanol. Next, concentrated HC1, Pd/C is added and the mixture is placed under hydrogen at 1 atm. The mixture is stirred overnight at room temperature and filtered. The residue is recrystallized to give intermediate compound 2. 6-chloropurine riboside is combined and the mixture is compound 2 dissolved in methanol and treated with triethylamine. The reaction is heated to 80° C for 30 hours. Isolation and purification leads to Compound 3.

EXAMPLE 2

Compounds of this invention prepared according to the method of Example 1 were tested in two functional models specific for adenosine A, receptor agonist function. The first was the A , receptor mediated inhibition of isoproterenol stimulated cAMP accumulation in DDT cells. The EC50 of each derivative is shown in Table I. Also shown in Table I is the ability of each derivative to stimulate cAMP production in PC 12 cells, a function of agonist stimulation of adenosine A2 receptors. The ratio of the relative potency of each compound in stimulating either an A, receptor or an A2 receptor effect is termed the selectivity of each compound for the A, receptor. As can be seen in Table I, each derivative is relatively selective as an A, receptor agonist. The use of measuring cAMP metabolism as an assay for adenosine A , receptor function has been previously described (Scammells, P., Baker, S., Belardinelli, L., and Olsson, R. , 1994, Substituted 1 ,3-dipropylxanthines as irreversible antagonists of A, adenosine receptors. J. Med. Chem 37: 2794-2712, 1994).

Table I

Compound R EC50 (nM) ECS, (nM) A,/A2 A-/A, DDT cells PC 12 cells

I 4-arninopyran 12 970 0.012 80.0

II (±)-3-aminotetrahydrofuran 13 1400 0.0093 107.6

III (R)-3-aminotetrahydrofuran 1.08 448 0.0024 414

IV ( 1 )-caprolactam 161 181 0.889 1.12

V (S)-3-aminotetrahydrofuran 3.40 7680 0.00044 2258

Compounds were also tested in a whole organ model of A, receptor activation with respect to atrial and AV nodal function. In this model, guinea pig hearts are isolated and perfused with saline containing compound while atrial rate and AV nodal conduction time are assessed by electrographic measurement of atrial cycle length and AV intervals, as detailed in Belardinelli, L, Lu, J. Dennis, D. Martens, J, and Shryock J. (1994); The cardiac effects of a novel A,-adenosine receptor agonist in guinea pig isolated heart. J. Pharm. Exp. Therap. 271:1371-1382 (1994). As shown in Figure 1, each derivative was effective in slowing the atrial rate and prolonging the AV nodal conduction time of spontaneously beating hearts in a concentration-dependent manner, demonstrating efficacy as adenosine A, receptor agonists in the intact heart.

EXAMPLE 3

Preparation ofN-benzyloxycarbonyl-4-aminopyran.

A mixture of 4-pyranylcarboxylic acid (2.28 gm, 20 mmol), diphenylphosphorylazide (4.31 ml, 20 mmol), triethylamine (2.78 ml, 20 mmol) in dioxane (40 ml) was heated in a 100° C oil bath under dry nitrogen for 1 hour. Benzyl alcohol (2.7 ml, 26 mmol) was added, and heating was continued at 100° C for 22 hours. The mixture was cooled, filtered from a white precipitate and concentrated. The residue was dissolved in 2N HC1 and extracted twice with EtOAc. The extracts were washed with water, sodium bicarbonate, brine and then dried over MgSO4, and concentrated to an oil which solidified upon standing. The oil was chromatographed (30% to 60% EtO Ac/Hex) to give 1.85 g of a white solid (40%).

Preparation of 4-aminopyran.

N-benzyloxycarbonyl-4-aminopyran (1.85 gm, 7.87 mmol) was dissolved in MeOH (50 ml) along with cone. HC1 and Pd-C ( 10%, 300 mg). The vessel was charged with hydrogen at 1 atm and the mixture was allowed to stir for 18 hours at room temperature. The mixture was filtered through a pad of eelite and concentrated. The residue was co-evaporated twice with MeOH/EtOAc and recrystallized from MeOH/EtOAc to afford 980 mg (91 %) of white needles (mp 228-230° C).

Preparation of 6-(4-aminopyran)-purine riboside. A mixture of 6-chloropurine riboside (0.318 gm, 1. 1 mmol), 4-aminopyran-HCl

(0.220 mg,

1.6 mmol) and triethylamine (0.385 ml, 2.5 mmol) in methanol (10 ml) was heated to 80° C for 30 hours. The mixture was cooled, concentrated and the residue chromatographed (90: 10: 1, CH2 Cl2/MeOH/PrNH2). The appropriate fractions were collected and recliromatographed using a chromatotron

(2 mm plate, 90: 10: 1, CH2 Cl2/MeOH/PrNH2) to give an off white foam (0.37 gm, 95%).

EXAMPLE 4

Preparation of N-benzyloxycarbonyl-3-aminotetrahydrofuran. A mixture of 3-tetrahydrofuroic acid (3.5 gm, 30 mmol), diphenylphosphorylazide (6.82 ml, 32 mmol), triethylamine (5 ml, 36 mmol) in dioxane (35 ml) was stirred at RT for 20 min then heated in a 100° C oil bath under dry nitrogen for 2 hours. Benzyl alcohol (4.7 ml, 45 mmol) was added, and continued heating at 100° C for 22 hours. The mixture was cooled, filtered from a white precipitate and concentrated. The residue was dissolved in 2N HC1 and extracted twice using EtOAc. The extracts were washed with water, sodium bicarbonate, brine dried over MgSO4, and then concentrated to an oil which solidifies upon standing. The oil was chromatographed (30% to 60% EtO Ac/Hex) to give 3.4 g of an oil (51

%).

Preparation of 3-aminotetrahydrofuran.

N-benzyloxycarbonyl-3-aminotetrahydrofuran (3.4 gm, 15 mmol) was dissolved in MeOH (50 ml) along with cone. HC1 and Pd-C (10%, 300 mg). The vessel was charged with hydrogen at 1 atm and the mixture was allowed to stir for 18 hours at room temperature. The mixture was filtered through a pad of celite and concentrated. The residue was co-evaporated two times with MeOH/EtOAc and recrystallized from MeOH/EtOAc to give 1.9 g of a yellow solid.

Preparation of 6-(3-aminotetrahydrofuranyl)purine riboside. A mixture of 6-chloropurine riboside (0.5 gm, 1.74 mmol), 3-aminotetrahydrofuran

(0.325 gm, 2.6 mmol) and triethylamine (0.73 ml, 5.22 mmol) in methanol (10 ml) was heated to 80° C for 40 hours. The mixture was cooled, and concentrated. The residue was filtered through a short column of silica gel eluting with 90/10/1 (CH2Cl2/MeOH/PrNH2), the fractions containing the product were combined and concentrated. The residue was chromatorgraphed on the chromatotron (2 mm plate, 92.5/7.5/1 , CH2CL2/MeOH/P.NH2). The resulting white solid was recrystallized from MeOH/EtOAc to give 0.27 gm of white crystals (mp 128-130° C).

EXAMPLE 5

Resolution of 3-arninotetrahydrofuran hydrochloride

A mixture of 3-aminotetrahydrofuran hydrochloride (0.5 gm, 4 mmol) and

(S)-(+)-10-camphorsulfonyl chloride (1.1 gm, 4.4 mmol) in pyridine (10 ml) was stirred for 4 hours at room temperature and then concentrated. The residue was dissolved in EtOAc and washed with 0.5N HC1, sodium bicarbonate and brine. The organic layer was dried over MgSO4, filtered and concentrated to give 1. 17 g of a brown oil (97%) which was chromatographed on silica gel (25% to 70% EtOAc/Hex). The white solid obtained was repeatedly recrystallized from acetone and the crystals and supernatant pooled until an enhancement of greater than 90% by 1H NMR was acheived.

Preparation of 3-(S)-aminotetrahydrofuran hydrochloride.

The sulfonamide (170 mg, 0.56 mmol) was dissolved in cone. HCl/AcOH (2 mL each), stirred for 20 hours at room temperature, washed three times with CH2C12 (10 ml) and concentrated to dryness to give 75 mg (qaunt ) of a white solid

 

Preparation of 6-(3-(S)-aminotetrahydrofuranyl)puπne riboside.

A mixture of 6-chloropurιne riboside (30 mg, 0.10 mmol),

3-(S)-amιnotetrahydrofuran hydrochloride (19 mg, 0.15 mmol) and triethylamine (45 ml, 0.32 mmol) in methanol

(0.5 ml) was heated to 80° C for 18 hours. The mixture was cooled, concentrated and chromatographed with 95/5 (CH2Cl /MeOH) to give 8 mg (24%) of a white solid.

Chemical structure for tecadenoson
Literature References:
Selective adenosine A1-receptor agonist. Prepn: R. T. Lum et al., WO 9808855; eidem, US 5789416 (both 1998 to CV Therapeutics).
Clinical effect on AV nodal conduction: B. B. Lerman et al., J. Cardiovasc. Pharmacol. Ther. 6, 237 (2001).
Clinical evaluation in paroxysmal supraventricular tachycardia: E. N. Prystowsky et al., J. Am. Coll. Cardiol. 42, 1098 (2003); K. A. Ellenbogen et al., Circulation 111, 3202 (2005).
Review of pharmacology and clinical experience: A. Zaza, Curr. Opin. Invest. Drugs 3, 96-100 (2002); J. W. Cheung, B. B. Lerman, Cardiovasc. Drug Rev. 21, 277-292 (2003).
US7144871 * 19 Feb 2003 5 Dec 2006 Cv Therapeutics, Inc. Partial and full agonists of A1 adenosine receptors
US7696181 * 24 Aug 2006 13 Apr 2010 Cv Therapeutics, Inc. Partial and full agonists of A1 adenosine receptors
 
 
 
Keywords: Antiarrhythmic,  Adenosine Receptor Agonist, Tecadenoson, CVT-510, CV Therapeutics

RAVUCONAZOLE

Ravuconazole

BMS-207147, ER-30346
  • BMS 207147
  • ER 30346
  • Ravuconazole
  • UNII-95YH599JWV
CAS Registry Number: 182760-06-1
CAS Name: 4-[2-[(1R,2R)-2-(2,4-Difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]-4-thiazolyl]benzonitrile
Additional Names: (2R,3R)-3-[4-(4-cyanophenyl)thiazol-2-yl]-2-(2,4-difluorophenyl)-1-(1H-1,2,4-triazol-1-yl)-2-butanol
(2R,3R)-3-i4-(4-cvanophenyl)thiazol-2-yl1-1 -(1 H-1 ,2,4-triazol-1 -yl)-2-(2,4-difluorophenyl)- butan-2-ol
 [R-(R*,R*)]-4-[2-[2-(2,4-Difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]-4-thiazolyl]benzonitrile
4-[2-[(1R,2R)-2-(2,4-Difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,2,4-triazol-1-yl)propyl]-4-thiazolyl]benzonitrile
Molecular Formula: C22H17F2N5OS
Molecular Weight: 437.47
Percent Composition: C 60.40%, H 3.92%, F 8.69%, N 16.01%, O 3.66%, S 7.33%
Eisai (INNOVATOR)

poser

Properties: Colorless prisms from diisopropyl ether/n-hexane, mp 164-166°. [a]D24=  -29.1° (c = 1.03 in methanol).
Melting point: mp 164-166°
Optical Rotation: [a]D24=  -29.1° (c = 1.03 in methanol)
Therap-Cat: Antifungal.
http://www.google.com/patents/WO2011042827A1?cl=en…………m.p. 164-166° C; [a]=-30° (c=1 , methanol, 25° C); NMR (CDCI3): 1 .23(3H, d, J=8 Hz), 4.09(1 H, q, J=8Hz), 4.26(1 H, d, J=14Hz), 4.92(1 H, d, J=14Hz), 5.75(1 H, s), 6.75- 6.85(2H, m), 7.45-7.54(2H, m), 7.62(1 H, s), 7.69(1 H, s), 7.75(1 H, d, J=8Hz), 7.86(1 H, s), 8.03(1 H,d,J=8Hz). The analytical data were identical with published (US5648372 and Chem. Pharm. Bull. 1998, 46, 623-630).
Ravuconazole (BMS-207147 and ER-30346) is a potent triazole antifungal, being developed by Bristol-Myers Squibb, that is currently in phase I/II clinical trials.[1] The drug has a shown to have a similar spectrum of activity to voriconazole, with an increased half-life.[2] However, ravuconazole has limited activity against species of Fusarium, Scedosporium, and Zygomycetes.[3][4]
 
Ravuconazole
Ravuconazole.svg
Systematic (IUPAC) name
4-[2-[(2R,3R)-3-(2,4-Difluorophenyl)-3-hydroxy-4-(1,2,4-triazol-1-yl)butan-2-yl]-1,3-thiazol-4-yl]benzonitrile
Clinical data
Legal status
PHASE 2 AS ON SEPT 2014
Identifiers
CAS number 182760-06-1 Yes
ATC code None
PubChem CID 467825
NIAID ChemDB 057176
Chemical data
Formula C22H17F2N5OS 
Mol. mass 437.465086 g/mol

Chemical structure for Ravuconazole

DRUG PROCESS…do not miss this

http://www.drugprocess.com/pdf/Isavuconazole_DPLA_ProcessSummary.pdf =++++++++++++++++++++++

…………………………………………………

Thiazole antifungals. III. Stereocontrolled synthesis of an optically active triazolymethyloxirane precursor to antifungal oxazolidine derivatives
Chem Pharm Bull 1991, 39(9): 2241

https://www.jstage.jst.go.jp/article/cpb1958/39/9/39_9_2241/_pdf

……………………………………………………

Optically active antifungal azoles. I. Synthesis and antifungal activity of (2R,3R)-2-(2,4-difluorophenyl)-3-mercapto-1-(1H-1,2,4-triazol-1-yl)-2-butanol and its stereoisomers
Chem Pharm Bull 1993, 41(6): 1035

https://www.jstage.jst.go.jp/article/cpb1958/41/6/41_6_1035/_pdf

………………………………………………………….

A novel route for chiral synthesis of the triazole antifungal ER-30346
Chem Pharm Bull 1998, 46(7): 1125

https://www.jstage.jst.go.jp/article/cpb1958/46/7/46_7_1125/_pdf

……………………………………………………….

ER-30346 is synthesized by thiazole ring formation of (2R, 3R) -3- (2,4-difluorophenyl) -3-hydroxy-2-methyl-4- (1H-1,2,4-triazol-1-yl ) thiobutanamide (I) and 4-bromoacetylbenzonitrile (II) by means of reflux in methanol. The thioamide (I) is obtained with excellent yield from a chiral nitrile (III) by heating with diethyl dithiophosphate in aqueous medium.

Synthesis method
The nitrile (III), a chiral key intermediate of this synthesis, can be obtained by two different synthetic routes as follows: Route-a: The key step of this route is ring opening reaction of the trisubstituted oxirane (VII) by cyanide anion leading to the nitrile (III). The chiral oxirane (VII) is synthesized from (R) -lactic acid derivatives as already reported. The reaction of (VII) with diethylaluminum cyanide in toluene or lithium cyanide in tetrahydrofuran gives the nitrile (III) with high yield without any epimerization reaction.

 

The nitrile (III), a chiral key intermediate of this synthesis, can be obtained by two different synthetic routes as follows: Route-b: The starting material of this route is methyl (S) -3-hydroxy-2-methylpropionate (VIII ), which contains one additional carbon between the hydroxyl group and the 2-position carbon of (R) -lactate, the starting material of route-a. The hydroxyl group of (VIII) is protected by triphenylmethyl group. Then, 2,4 -difluorophenyl moiety is introduced to give the ketone (X). Direct conversion of the ketone (X) to the oxirane (XIV) by dimethylsulfoxonium methylide, the same condition for compound (IV) in route-a, does not proceed. The oxirane (XIV) having desired stereochemistry is obtained via oxidation reaction. The ketone (X) is converted to the exomethylene (XI) by Wittig reaction. The stereoselective oxidation of (XI) is achieved by means of osmium tetroxide in the presence of 4-methylmorpholine N-oxide to give the diol (XII) in 58% yield after separation of its epimer by column chromatography. After methanesulfonylation of the primary alcohol of (XII), a triazole moiety is introduced and the triphenylmethyl group is deprotected. Then, the primary hydroxyl group of (XVI) is oxidized under Swern oxidation condition to give the aldehyde (XVII), which is converted to the chiral nitrile intermediate (III) by means of heating with hydroxylamine-O-sulfonic acid.

 

The synthesis of (2S, 3S) -3- (2,4-difluorophenyl) -3-hydroxy-2-methyl-4- (1,2,4-triazol-1-yl) butyronitrile (XV), a key intermediate the synthesis of ER-30346 has been described: The tritylation of 3-hydroxy-2 (S) -methylpropionic acid methyl ester (I) with trityl chloride in hot pyridine gives the trityl ether (II), which is hydrolyzed with LiOH in H2O / THF / methanol yielding the free acid (III). The esterification of (III) with 2-mercaptopyridine (IV) by means of dicyclohexylcarbodiimide (DCC) in dichloromethane gives the thioester (V), which is treated with 2,4-difluorophenylmagnesium bromide (VI) in THF yielding the propiophenone (VII), which by treatment with methyltriphenylphosphonium bromide / NaH in THF is converted into the methylene derivative (VIII). The oxidation of (VIII) with OsO4 and N-methylmorpholine oxide in acetone affords, after column chromatography, the chiral diol (IX), which is monomesylated with mesyl chloride / triethylamine in dichlormethane giving the monoester (X). The reaction of (X) with 1,2,4-triazol (XI) and NaH in DMF yields (2R, 3S) -2- (2,4-difluorophenyl) -3-methyl-1- (1,2,4-triazol-1-yl) -4- (triphenylmethoxy) -2-butanol (XII), which is detritylated with p-toluenesulfonic acid in methanol affording the diol (XIII). The oxidation of (XIII) with oxalyl chloride / DMSO in dichloromethane gives the aldehyde (XIV), which is finally treated with hydroxylamine-O-sulfonic acid in water yielding the desired bytyronitrile intermediate (XV) already referenced.

http://www.google.com/patents/WO2011042827A1?cl=en

Example 1

(2R,3R)-3-i4-(4-cvanophenyl)thiazol-2-yl1-1 -(1 H-1 ,2,4-triazol-1 -yl)-2-(2,4-difluorophenyl)- butan-2-ol

To a solution of racemic 3-[4-(4-cyanophenyl)thiazol-2-yl]-1 -(1 H-1 ,2,4-triazol-1 -yl)-2-(2,4- difluorophenyl)-butan-2-ol (43.7 g) in acetone (800 ml) a solution of (1 R)-10- camphorsulfonic acid (23 g) in methanol (300 ml) was added and the mixture was heated under reflux until a clear solution was obtained. The solution was slowly cooled to rt, seeded with crystals of the title enantiomeric salt and let overnight. The solid was collected by filtration, washed with acetone and dried to provide (2R,3R)-3-[4-(4- cyanophenyl)thiazol-2-yl]-1 -(1 H-1 ,2,4-triazol-1 -yl)-2-(2,4-difluorophenyl)-butan-2-ol (1 R)- 10-camphorsulfonate as white solid. This crude salt was then taken up in methylenechloride (100 ml) and water (ca. 100 ml) and the mixture was basified with aqueous sodium hydroxide solution. The organic layer was separated and the aqueous phase washed twice with methylenechloride (50 ml) and combined. The organic phases were then washed twice with water (2×50 ml), dried with sodium sulfate, filtrated and the solvent removed under reduced pressure. The crude product was then mixed with isopropanol (ca. 150 ml), heated for 10 min, cooled to 0° C and stirred for ca. 2 hrs. The product was collected, washed with isopropanol and dried under reduced pressure to provide the enantiomerically pure title compound (17.5 g, 41 % yield, 99.1 % ee); m.p. 164-166° C; [a]=-30° (c=1 , methanol, 25° C); NMR (CDCI3): 1 .23(3H, d, J=8 Hz), 4.09(1 H, q, J=8Hz), 4.26(1 H, d, J=14Hz), 4.92(1 H, d, J=14Hz), 5.75(1 H, s), 6.75- 6.85(2H, m), 7.45-7.54(2H, m), 7.62(1 H, s), 7.69(1 H, s), 7.75(1 H, d, J=8Hz), 7.86(1 H, s), 8.03(1 H,d,J=8Hz). The analytical data were identical with published (US5648372 and Chem. Pharm. Bull. 1998, 46, 623-630).

…………………………

http://www.google.com/patents/WO1999045008A1?cl=en

Example 1

a) Preparation of (2R)-2′,5′-Difluoro-2-(3,4,5,6-tetrahydro-

2H-pyran-2-yloxy)-propiophenone A mixture of magnesium ( 7.25 g, 0.298 mol ) and iodine ( catalytic amount ) and l-bromo-2,5-difluorobenzene ( 20.0 g, 0.178 mol ) in THF ( 250ml ) was vigously stirred. The color of iodine was disappeared and the inner temperature rose up to 65°C. To this mixture was added additional l-bromo-2,5-difluorobenzene ( 30.0 g, 0.267 mol ) dropwise to maintain the inner temperature from 50 to 55°C over 45min. The resulting mixture was stirred at 55°C for 30min. then at r.t. for lhr. The – 21 –

mixture was cooled down to -5°C. To this mixture was added a solution of.4-[(2R)-2-(3,4,5,6-Tetrahydro-2H-pyran-2-yloxy)propionyl] morpholine ( 52.5 g, 0.216 mol ) in THF ( 150ml ) dropwise over 40min. And the resulting mixture was stirred at r.t. for 4hrs. The reaction mixture was cooled down to 5°C and saturated NH4C1 aq. ( 100ml ) was added carefully. The whole was diluted with H20 ( 600ml ) and extracted with EtOAc ( 400ml + 200ml x 2 ). The combined organic layer was dried over Na2S04 and concentrated in vacuo. The residue was chromatographed on silica gel ( n-hexane : EtOAc = 10 :1 ~ 5 : 1 ) to give (2R)-2′,5′- Difluoro-2-(3,4,5,6-tetrahydro-2H-pyran-2-yloxy)-propiophenone (47.3 g,

81 % ) as pale yellow syrup.

Physical form : colorless oil; FAB-MS: m/z 271(M+H)+; Η-NMR(CDCl;j): 1.42~1.90(9H,m),3.32~3.40(lHxl/2,m),3.69~3.77(lHxl/2,m),3.86~3.94 (lHxl/2,m),4.66(lHxl/2,t,J=3.6Hz),4.75(lHxl/2,t,J=3.6Hz),4.87(lHxl/2, q,J=6.6Hz),5.11(lHxl/2,q,J=6.9Hz),7.08~7.25(2H,m),7.49~7.55(lH,m).

b) Preparation of 2-(2,5-Difluorophenyl)-2-[(lR)-l-(3,4,5,6,- tetr ahy dro-2H-pyran-2-yloxy ) ethyl] oxir ane To a stirred mixture of NaH ( 60% in oil, 9.1g, 0.228mol ) in DMSO

(300ml ) was added portionwise trimethylsulfoxonium iodide ( 53.9g, 0.245 mol ) at the inner teperature with the range from 15°C to 18°C. over 20min. The ice bath was removed and the mixtuer was stirred at r.t. for 3hrs. The mixture was cooled down to 10°C. To this mixture was added a solution of (2R)-2′,5′-Difluoro-2-(3,4,5,6-tetrahydro-2H-pyran-2- yloxy)-propiophenone ( 47.3 g , 0.175 mol ) in DMSO (150ml ) dropwise over 20min. The resulting mixture was stirred at r.t. for 4hrs. The reaction mixture was poured into ice-water ( 800ml ). The whole was extracted with EtOAc ( 400ml + 200ml x 2 ). The combined organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo.

The residue was chromatograkkphed on silicagel ( n-hexane : EtOAc = – 22 –

8 : 1 ~ 5 : 1 ) to give 2-(2,5-Difluorophenyl)-2-[(lR)-l-(3,4,5,6,- tetrahydro-2H-pyran-2-yloxy)ethyl]oxirane (48.3 g, 97 % ). Physical form : pale yellow syrup, EI-MS: m/z 284 (M)+ ; 1H-NMR(CDC13): 1.15(3Hxl/2,dd,J=6.6,1.3Hz), 1.24(3Hxl/2,dd, J=6.6,1.3Hz), 1.52-1.87 (6H,m),2.83~2,90(lH,m),3.07

(lHxl/2,d,J=5.3Hz),3.36(lHxl/2,d,J=5.6Hz), 3.48~3.56(lH,m),3.82~3.92 (lH,m),4.00~4.16(lH,m),4.73~4.92(lH,m), 6.96~7.02(lH,m),7.09~7.15 (lH,m).

c) Preparation of (3R)-2-(2,5-difluorophenyl)-3-(3,4,5,6- tetrahydro-2H-pyran-2-yloxy)-l-(lH-l,2,4-triazol-l-yl)-2-butanol

To a stirred suspension of NaH ( 60 % in oil, 21.0 g, 0.525 mol ) in DMF (300ml ) was added portionwise 1,2,4-triazole ( 43.3 g, 0.627 mol ) at the inner temperature from 2°C to 11°C over 30min. The resulting mixture was stirred at r.t. for l.δhrs. To this mixture was added a solution of 2-(2,5-Difluorophenyl)-2-[(lR)-l-(3,4,5,6-tetrahydro-2H- pyran-2-yloxy)ethyl]oxirane ( 48.3 g, 0.170 mol ) in DMF ( 50 ml ). The mixture was stirred at 60°C for lhr. and then at 65°C for 14hrs. The reaction mixture was cooled down to 10°C and then poured into ice- water (800 mL ). The resulting mixture was extracted with EtOAc

(400ml + 200ml x 2 ). The combined organic layer was dried over Na2S04 and concentrated in vacuo. The residue was chromatographed on silicagel ( n-hexane : EtOAc = 4 : 1 ~ 1 : 5 ) to give (3R)-2-(2,5- difluorophenyl)-3-(3,4,5,6-tetrahydro-2H-pyran-2-yloxy)-l-(lH-l,2,4- triazol-l-yl)-2-butanol ( 43.9 g, 73 % ) and recovered starting material

(13.2 g, 27 % ).

Physical form : colorless syrup ; FAB-MS: m/z 354 (M+H)+ ; Η- NMR(CDCl3): 1.00(3Hxl/2,d,J=6.6Hz),1.13(3Hxl/2,d,J=6.6Hz), 1.42~1.88(6H,m),3.38~3.60 (lH,m),3.80~4.00(lH,m),4.32~5.02(5H,m),6.83~6.99 (2H,m),7.14-7.21

(lH,m),7.73(lHxl/2,s),7.74(lHxl/2,s),7.92(lHxl/2,s),7.95(lHxl/2,s). – 23 –

d) Preparation of (2R,3R)-2-(2,5-difluorophenyl)-l-(lH-l,2,4- triazol-l-yl)-2,3-butanediol

A mixture of (3R)-2-(2,5-difluorophenyl)-3-(3,4,5,6-tetrahydro-2H- pyran-2-yloxy)-l-(lH-l,2,4-triazol-l-yl)-2-butanol ( 43.9 g, 0.124 mol ) and PPTS ( 15.6 g, 62.1 mmol ) in EtOH ( 400ml ) was stirred at 55°C for 5hrs. The mixture was was evaporated to remove solvent down to 100ml. The residue was poured into ice-aqueous NaHC03 ( 500ml ). The whole was extracted with EtOAc ( 400ml + 200ml x 2 ). The combined organic layer was dried over Na2S04 and concentrated in vacuo. The residue was chromatographed on silicagel (CH2C12 : MeOH = 20 : 1) to give (2R,3R)-2-(2,5-difluorophenyl)-l-(lH-l,2,4-triazol-l-yl)-2,3- butanediol (18.0 g, 54 % ). Physical form : colorless syrup ; FAB-MS: m/z 270 (M+H)’ ; ‘H- NMR(CDC13): 0.99(3H,d,J=6.6Hz),2.61(lH,d,J=10.6Hz), 4.31-4.36

(lH,m),4.79,4.88

(2H,ABq,J=14.5Hz),4.84(lH,s),6.84~6.99(2H,m),7.13~7.19(lH,m),7.84(l H,s),7.85(lH,s).

e) Preparation of (2R,3S)-2-(2,5-Difluorophenyl)-3-methyl-2-

[ ( 1H- 1 ,2,4-triazol-l -yl) -methyl] -oxir ane

To a cold ( 0°C ) and stirred solution of (2R,3R)-2-(2,5-difluorophenyl)- l-(lH-l,2,4-triazol-l-yl)-2,3-butanediol ( 35.0 g, 0.130 mol ) and triethylamine ( 54.8 ml, 0.393 mol ) in CH2C12 ( 500ml ) was added a mesylchloride ( 12.1 ml, 0.156 mol ) dropwise over 5min. The resulting mixture was stirred at r.t. for l.δhrs. The reaction mixture was poured into ice-water ( 300ml ). The resulting mixture was shaken well and the organic layer was separated. The aqueous layer was further extracted with CH2C12 ( 150ml x 2 ). All the organic layers were combined, dried over Na2SO4 and concentrated in vacuo to give mesylate ( 46.7 g ) as crude syrup. The obtained mesylate was dissolved in MeOH ( 500ml ) – 24 –

and the solution was cooled down to 0°C. To this solution was added 28% NaOMe methanol solution (29.0 ml ). The mixture was stirred at 0°C for 50min. The reaction mixture was evaporated to reduce the volume of the solvent down to 150 ml. The residue was poured into ice- water ( 300ml ). The resulting mixture was extracted with ethylacetate (300ml + 200ml x 2 ). The combined organic layer was dried over Na.,S0 and concentrated in vacuo. The residue was cromatographed on silicagel (hexane : EtOAc = 1 : 3 ) to give (2R,3S)-2-(2,5-Difluorophenyl)- 3-methyl-2-[(lH-l,2,4-triazol-l-yl)-methyl]-oxirane (30.3 g, 93 %).

Physical form : white solid ; FAB-MS : m z 252 (M+H)+ ; ]H- NMR(CDC13): 1.64(3H,d,J=5.6Hz),3.19(lH,q,J=5.6Hz),4.42,4.97 (2H,ABq,J=14.8Hz), 6.75~6.81(lH,m),6.89~7.01(2H,m),7.83(lH,s),7.98 UH,s).

f) Preparation of (2S,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-

2-methyl-4-[l,2,4]triazol-l-yl-butyronitrile

A mixture of (2R,3S)-2-(2,5-Difluorophenyl)-3-methyl-2-[(lH-l,2,4- triazol-l-yl)-methyl]-oxirane ( 30.3 g, 0.121 mol ), trimethylsilylcyanide ( 65.0 ml ) and MgO ( 24.5 g ) in o-xylene ( 400 ml ) was stirred at 130°C for lOhrs. To this mixture was added additional trimethylsilylcyanide (20.0 ml ) and MgO ( 8.5 g ) and the resulting mixture was stirred at 130°C further for 6hrs. The reaction mixture was cooled down to r.t. The precipitate was filtered off and washed with CH2C12. The filtrate was concentrated in vacuo to give crude brown syrup.

This crude syrup was dissolved in THF ( 600ml ) and the solution was cooled down to 0°C. To this mixture was added 1.0 M tetra n- butylammoniumfluoride THF solution ( 133ml, 0.133 mol ) dropwise over 5min. The mixture was stirred at r.t. for 50min. The solvent was removed under reduced pressure down to 150ml. The residue was poured into ice-water ( 400ml ). The resulting mixture was extracted – 25 –

with EtOAc ( 300ml + 200ml x 2 ). The combined organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was chromatographed on silicagel ( n-hexane : EtOAc = 1 : 3 ) to give (2S,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-2-methyl-4-[l,2;4]triazol-l-yl- butyronitrile ( 30.5 g, 91 % ).

Physical form : colorless syrup ; FAB-MS : m/z 279 (M+H)+ ; Η- NMR(CDCl3): 1.19(3H,d,J=7.3Hz),3.33(lH,q,J=7.3Hz),4.82,5.00 (2H,ABq,J=13.9Hz), 5.56(lH,brs),6.89~7.04(2H,m),7.12~7.19(lH,m),7.85(lH,s),7.86(lH,s).

g) Preparation of (2R,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-

2-methyl-4- [ 1 ,2,4] triazol-1 -ylthiob tyramide

A mixture of (2S,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-2-methyl-4- [l,2,4]triazol-l-yl-butyronitrile ( 305 S> O.llOmol ), diethyldithio- phospate ( 235 ml ) and H2O ( 110 ml ) was stirre at 80°C for 2hrs. The reaction mixture was cooled down to r.t. n-Hexane ( 400ml ) and water (200 ml ) was added. The whole was shaken well and the aqueous layer was separated. The remaining organic layer was further extracted with H20 ( 100ml x 3 ). All the aqueous layer was combined. Cooled down to

0°C and neutralized and basified ( PH8 ) with NaHC03. This basic(PH8) aqueous layer was extracted with EtOAc ( 300ml + 100ml x 3 ). The combined organic layer was dried over Na2S04 and concentrated in vacuo to give dark brown syrup. By addition of CH2C12 ( 100ml ) to this crude syrup, precipitate was formed. The precipitate was filtered and washed with CH2C12-hexane ( 5 : 1 mixture ) to give (2R,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-2-methyl-4-[l,2,4]triazol-l- ylthiobutyramide ( 19.2 g, 56 % ) as white powder. On the oter hand, the filtrate was concentrated in vacuo and the residue was chromatographed on silica gel ( Wako-gel C-300, CH2C12 : MeOH = 20 :

1 ) to give additional (2R,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-2- – 26 –

methyl-4-[l,2,4]triazol-l-ylthiobutyramide ( 7.46 g, 22 % ) as pale brown amorphous powder.

Physical form : White solid ; FAB-MS : m/z 313 (M+H)+ ; ‘H-NMR (CDC13): 1.12(3H,d,J=7.3Hz),3.74(lH,q,J=7.3Hz), 4.55,5.12 (2H,ABq,J=14.5Hz), 5.84(lH,s),6.85~7.02(2H,m),7.15-7.22(lH,m),7.80

(1H,S),7.89(1H,S), 7.89(lH,brs),8.43(lH,brs).

h) Preparation of 4-{2-[(lR,2R)-2-(2,5-Difluoro-phenyl)-2- hydroxy-l-methyl-3-[l,2,4]triazol-l-yl-propyl]-thiazol-4-yl}- benzonitrile

A mixture of (2R,3R)-3-(2,5-Difluoro-phenyl)-3-hydroxy-2-methyl-4- [l,2,4]triazol-l-ylthiobutyramide ( 26.7 g, 85.4 mmol ) and a-bromo-4′- cyano-acetophenone ( 24.0 g, 0.107 mol ) in EtOH ( 500ml ) was refluxed for lhr. The reaction mixture was cooled down to r.t. And the solvent was removed under reduced pressure down to 150ml. The residue was poured into in to cold ( 0°C ) saturated NaHC03 aq. ( 400ml ). The resulting mixture was extracted with EtOAc ( 300ml + 150 ml x 2 ). The combined organic layer was washed with brine (200ml ), dried over Na2S04 and concentrated in vacuo. The residue was chromatographed on silica gel ( Wako-gel C-300, Hexane : EtOAc = 1 : 2 ) to give 4-{2-

[(lR,2R)-2-(2,5-Difluoro-phenyl)-2-hydroxy-l-methyl-3-[l,2,4]triazol-l- yl-propyl]-thiazol-4-yl}-benzonitrile ( 32.0 g, 86 % ).

Physical form : colorless heavy syrup ; ESI-MS : m/z 437 (M)+ ; ‘H-

NMR(CDCl3): 1.25(3H,d,J=7.3Hz),4.12(lH,q,J=7.3Hz),4.26,4.96 (2H,Abq,J=14.5Hz), 5.75(lH,s),6.89~7.07(2H,m),7.23~7.29(lH,m),7.65

(lH,s),7.71(lH,s),7.75,8.02 (4H,Abq,J=8.6Hz),7.85(lH,s). – 27 –

i) Preparation of 4-{4-[(tert-Butoxycarbonyl-methyl-amino)- acetoxy]-3,5-dimethyl-benzyl}-l-[(2R,3R)-3-[4-(4-cyano-phenyl)- thiazol-2-yl]-2-(2,5-difluoro-phenyl)-2-hydroxy-butyl]-lH- [l,2,4]triazol-4-ium bromide A mixture of 22.7mg of 4-{2-[(lR,2R)-2-(2,5-Difluoro-phenyl)-2-hydroxy- l-methyl-3-[l,2,4]triazol-l-yl-propyl]-thiazol-4-yl}-benzonitrile and 25.0mg of 4-tert-butoxycarbonyl-methyl-aminoacetoxy-3,5-dimethyl- benzyl bromide in CH3CN(1.5mL) was refluxed over 15hrs. The solvent was evaporated in vacuo and the residue was chromatographed on silica gel (Wakogel C-200, solvent:CH2Cl MeOH=10/l) to give 4-{4-[(tert-

Butoxycarbonyl-methyl-amino)-acetoxy]-3,5-dimethyl-benzyl}-l- [(2R,3R)-3-[4-(4-cyano-phenyl)-thiazol-2-yl]-2-(2,5-difluoro-phenyl)-2- hydroxy-butyl]-lH-[l,2,4]triazol-4-ium bromide (36.0mg, 84% as colorless heavy syrup) ; FAB-MS : m/z 743 (M-Br)’ ; Η-NMR(CDC1S): 1.23(3H,d,J=7.3Hz),

1.47(9H,s),2.14(6H,s),3.03(3H,s),4.15(lH,q,J=7.3Hz),4.25(2H,s), 4.98,5.16(2H,ABq,J=13.9Hz),5.39~5.54(2H,m),6.27(lH,s),6.89-7.07(4H, m),7.24~7.27(lH,m),7.58(lH,s),7.73,8.06(4H,ABq,J=8.58),8.07(lH,s),ll. 26 (lH,s).

j) Preparation of l-{(2R,3R)-3-[4-(4-cyano-phenyl)-thiazol-2-yl]- 2-(2,5-difluoro-phenyl)-2-hydroxy-butyl}-4-(3,5-dimethyl-4- methylaminoacetoxy-benzyl)-lH-[l,2,4]triazol-4-ium bromide To a solution of 36mg of 4-{4-[(tert-Butoxycarbonyl-methyl-amino)- acetoxy]-3,5-dimethyl-benzyl}-l-[(2R,3R)-3-[4-(4-cyano-phenyl)-thiazol-

2-yl]-2-(2,5-difluoro-phenyl)-2-hydroxy-butyl]-lH-[l,2,4]triazol-4-ium bromide in ethylacetate(2ml) was added dropwise 4N HC1 ethylacetate solution(lmL) and the mixture was stirred at r.t. for 4hrs.The precipitate was filtered and washed with diethylether to give 1- {(2R,3R)-3-[4-(4-cyano-phenyl)-thiazol-2-yl]-2-(2,5-difluoro-phenyl)-2- hydroxy-butyl}-4-(3,5-dimethyl-4-methylaminoacetoxy-benzyl)-lH- – 28 –

[l,2,4]triazol-4-ium bromide (24.5mg, 74% as HC1 salt and as white solid) ;

FAB-MS : m/z 643 (M-Br)+ ; Η-NMR(DMSO-d): 1.19(3H,d,J=7.3Hz), 2.11(6H,s),2.64(3H,s),4.15(lH,q,J=7.3Hz),4.41(2H,s),4.74,5.04(2H,ABq,J =14.5Hz),5.40(2H,s),6.76(lH,brs),7.10(2H,s),7.20~7.38(2H,m), 7.94,8.21

(4H,ABq,J=8.25),8.45(lH,s),9.07(lH,s),9.50(lH,brs),10.17(lH,s).

………………………

http://www.google.co.in/patents/US5648372

OR

http://www.google.co.in/patents/EP1394142A1

COMPD 21

Figure 01040001

    Example 88:Preparation of a compound of the structural formula:

  • Figure 01380001
  • 2-(2,4-Difluorophenyl)-3-thioamide-1-(1H-1,2,4-triazol-1-yl)-2-butanol (the raw material 2) (156 mg) was dissolved in EtOH (2 ml), and 2-bromo-4′-cyanoacetophenone (the raw material 3) (224 mg) was added to the solution, followed by heating and refluxing for 1 hour. The liquid reaction mixture was neutralized with a saturated aqueous solution of NaHCO3 and subjected to extraction with AcOEt. After the extract was washed with H2O and then a saturated aqueous solution of NaCl and dried over MgSO4, AcOEt was distilled out. The resultant residue was purified by chromatography on silica gel (SiO2: 20 g, eluted with CH2Cl2 and then with 1% solution of MeOH in CH2Cl2), and then crystallized from IPE, thereby obtaining the intended compound (109 mg). Physical properties of this compound are described below.

    mp:
    196-197°C.
    NMR:
    δ solvent (CDCl3)
    1.23(3H,d,J=8.0Hz), 4.09(1H,q,J=8.0Hz), 4.26(1H,d,J=14.3Hz), 4.92(1H,d,J=14.3Hz), 5.74(1H,s), 6.78-6.85(2H,m), 7.48-7.54(1H,m), 7.64(1H,s), 7.69(1H,s), 7.75(1H,d,J=8.1Hz), 7.85(1H,s), 8.03(1H,d,J=8.1Hz).
    MS:
    MH+ = 438.

References

  1.  National Cancer Institute. Ravuconazole in Preventing Fungal Infections in Patients Undergoing Allogeneic Stem Cell Transplantation. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- [cited 2010 Feb 18]. Available from:http://clinicaltrials.gov/ct2/show/NCT00064311?term=ravuconazole&spons_ex=Y&rank=1 NLM Identifier: NCT00064311.
  2.  The Aspergillus Website, Pasqualotto AC, Denning DW. Ravuconazole. Date accessed: 2010 Feb 18.
  3.  Pasqualotto AC, Thiele KO, Goldani LZ (2010). “Novel triazole antifungal drugs: focus on isavuconazole, ravuconazole and albaconazole”. Curr Opin Investig Drugs 11 (2): 165–74. PMID 20112166.
  4.  Pfaller, M. A.; Messer, S. A.; Hollis, R. J.; Jones, R. N.; Sentry Participants, Group (2002). “Antifungal Activities of Posaconazole, Ravuconazole, and Voriconazole Compared to Those of Itraconazole and Amphotericin B against 239 Clinical Isolates of Aspergillus spp. and Other Filamentous Fungi: Report from SENTRY Antimicrobial Surveillance Program, 2000”. Antimicrobial Agents and Chemotherapy46 (4): 1032. doi:10.1128/AAC.46.4.1032-1037.2002. PMC 127116. PMID 11897586.

Literature References:

Ergosterol biosynthesis inhibitor. Prepn (stereochemistry unspecified): T. Naito et al, EP 667346; eidem,US 5648372 (1995, 1997 both to Eisai); of optically acitve form: A. Tsuruoka et al., Chem. Pharm. Bull. 46, 623 (1998). Chiral synthesis: Y. Kaku et al., ibid. 1125.

In vitro comparative antifungal spectrum: J. C. Fung-Tomc et al., Antimicrob. Agents Chemother. 42, 313 1998. Antifungal activity in candidosis: K. V. Clemons, D. A. Stevens, ibid. 45, 3433 (2001); in aspergillosis: W. R. Kirkpatrick et al., J. Antimicrob. Chemother. 49, 353 (2002).

Clinical evaluation in onychomycosis: A. K. Gupta et al., J. Eur. Acad. Dermatol. Venereol. 19, 437 (2005).

Review of development and therapeutic potential: S. Arikan, J. H. Rex, Curr. Opin. Invest. Drugs 3, 555-561 (2002).

Extras you may need

moonwalk

http://www.google.com/patents/WO2011042827A1?cl=en

Scheme 1 :

Figure imgf000003_0001

The manufacturing process for Isavuconazole is similar: Since Isavuconazole differentiates from Ravuconazole by only another fluorine substitution on the aromatic ring (2,5- instead of 2,4-difluorophenyl), the identical synthesis has been used (US 6300353 from October 9, 2001 and Bioorg. & Med. Chem. Lett. 13, 191 (2003)). Consequently, also this manufacturing process, based on (R)-lactic acid, faces the same problems: to many steps, extremely low overall yield and in addition to US patent 6300353 claims even already known step as novel (claim 36).

Recent attempts to improve this concept as reported in WO 2007/062542 (Dec.1 , 2005), using less expensive, natural configured (S)-lactic acid, also failed: As already reported in US 6133485 and in US 2003/0236419, the second chiral center was formed from an optically active allyl alcohol prepared in a few steps from (S)-lactic acid. This allyl alcohol was subjected to Sharpless diastereoselective epoxidation providing first an opposite configured, epimeric epoxy alcohol which had to be then epimerized in an additional inversion step yielding finally the desired epoxy alcohol as the known precursor for Isavuconazole (US 6300353). It is obvious that this process using less expensive (S)- lactic acid makes the entire process with an inversion step even more complex than the original approach.

Elegant and more efficient process has been claimed in US 2004/0176432 from June 26, 2001 ) in which both chiral centers have been formed simultaneously, diastereo- and enantio-selectively pure in one single reaction step using chiral (R)-2-butynol as a chiral precursor in the presence of Pd(ll)-catalyst and diethyl zinc (Scheme 2).

Scheme 2:

Figure imgf000005_0001

Since water soluble, (R)-2-butynol is expensive, recently identical process has been published, in which instead of (R)-2-butynol less water soluble and therefore, less expensive (R)-4-phenyl-3-butyn-2-ol was used (Synthetic Commun. 39, 161 1 (2009)). Nevertheless, as incorrectly stated there, this process does not provide better diastereoselectivity than the original process using (R)-2-butynol: On the contrary disadvantage of this process is a very bad atom economy because huge phenyl group of (R)-4-phenyl-3-butyn-2-ol has to be “disposed” in oxidation step by the conversion of triple bond into carboxylic acid function.

……………………………

http://www.google.com/patents/WO2014023623A1?cl=en

The invention relates to a process for the manufacture of a

diastereomerically and enantiomerically enriched ester intermediate for isavuconazole or ravuconazole.

Isavuconazole and ravuconazole are triazole antifungal compounds. Processes for the manufacture of isavuconazole and ravuconazole were disclosed in patents WO99/45008, WO2007/062542 and WO03/002498 to Basilea. In WO2011/042827 a process for the manufacture of enantiomerically pure antifungal azoles such as ravuconazole and isavuconazole is disclosed, wherein a classical resolution of a racemic mixture is performed by the addition of an enantiopure chiral acid, then collection of the desired diastereomer followed by conversion of the salt into the enantiomerically pure form of the desired compound by treatment with a base or an ion-exchange resin. The disadvantages of using such classical resolution are that the chiral auxiliary needs to be applied in near stoichiometric amounts, and that additional process steps are required for recovery of these relatively high amounts of chiral reagent as well as for converting the salt into the free enantiopure product.

http://www.google.com/patents/US8076494

Reaction Scheme 1:

Figure US08076494-20111213-C00005

Lifitegrast, SAR 1118

Abstract Image
 Inline image 2

Lifitegrast, SAR 1118

SAR-1118-023

CAS 1025967-78-5

Xiidra (lifitegrast ophthalmic solution)

07/12/2016 08:48 AM EDT
The U.S. Food and Drug Administration approved Xiidra (lifitegrast ophthalmic solution) for the treatment of signs and symptoms of dry eye disease, on Monday, July 11, 2016. Xiidra is the first medication in a new class of drugs, called lymphocyte function-associated antigen 1 (LFA-1) agonist, approved by the FDA for dry eye disease.

L-​Phenylalanine, N-​[[2-​(6-​benzofuranylcarbonyl​)​-​5,​7-​dichloro-​1,​2,​3,​4-​tetrahydro-​6-​isoquinolinyl]​carbonyl]​-​3-​(methylsulfonyl)​-

INNOVATOR

SAR1118 is a white to off-white solid crystallized from methylethylketone. m.p. 154.4oC;
[α]D25=-5.0o(c =1% (w/w) inMeOH); solubility 90 μg/mL in water at 25oC(parent);
FT-IR(KBr): νmax3427, 3302, 3030, 2923, 1727, 1659, 1294, 1140, 826, 764 cm-1;
ESI-MS:m/z615.1[M+1]+, 637.0 [M+Na]+;
1H NMR (300 MHz,d6-DMSO): δ 12.90 (bs, 1H), 9.05 (d,J=6.0Hz,1H), 8.13 (d,J= 1.9 Hz,1H), 7.73 (m, 4H), 7.57 (m, 1H), 7.41 (bs, 1H), 7.05 (d,J= 1.9 Hz,1H),4.78 (bm, 3H),
3.63 (bm, 3H), 3.30 (m, 1H), 3.16 (s, 3H), 3.02 (m, 1H), 2.77 (m, 2H) ppm;
13CNMR (75.5 MHz,d6-DMSO): δ 172.1, 169.6, 163.6, 153.7, 147.8, 140.6, 125.7, 106.9, 53.1,
43.6, 36.4, 26.0 ppm;
Elemental analysis: calcd. for C29H24Cl2N2O7S: C 56.6%, H 3.9%, N 4.6%,S 5.2%, Cl 11.5%; found C 55.1%, H 4.0%, N 4.4%, S 5.2%, Cl 11.2%

PLEASE,  ALL THESE ARE NOT MINE,  FROM THE NETPLEASE NOT MINE FROM THE NET

ChemSpider 2D Image | Lifitegrast | C29H24Cl2N2O7S

SYN CONSTRUCTED FROM WO 2014018748,

3(2H)-Benzofuranone, 6-hydroxy- cas 6272-26-0

STR1

6-Benzofurancarboxylic acid cas 77095-51-3

NEXT…….

L-Phenylalanine, 3-bromo- cas 82311-69-1

L-Phenylalanine, 3-(methylsulfonyl)-, phenylmethyl ester, hydrochloride (1:1) cas  1194550-59-8

NEXT………..

WO 2014018748,

SAR 1118 ophthalmic solution from SARcode Bioscience (Brisbane, Calif.) is a first-in-class molecule that inhibits T-cell inflammation by blocking the binding of two key cellular surface proteins (LFA-1 and ICAM-1) that mediate the chronic inflammatory cascade, so it may be able to reduce the inflammation associated with dry-eye disease.

In September, the company initiated enrollment in a Phase III study (OPUS-1). This study will assess the safety and efficacy of SAR 1118 for the treatment of dry-eye disease. Approximately 588 patients will be randomized to receive SAR 1118 5.0% ophthalmic solution or placebo twice daily for 12 weeks. The primary outcome measures include inferior corneal fluorescein staining, vision-related function subscale of the Ocular Surface Disease Index, and safety and tolerability. The company plans to complete the study in the first half of 2012.
The Phase II trial was a randomized, placebo-controlled, multicen-ter trial that included 230 patients with dry eye. In this study, SAR 1118 demonstrated dose-dependent significant improvements in inferior corneal staining over 12 weeks. A statistically significant increase in tear production and improvement in vision-related functions were seen as early as two weeks after initiation of treatment. SAR 1118 was well-tolerated, and no serious ocular adverse events were reported.
Has been found to be an effective inhibitor of Lymphocyte Function- Associated Antigen- 1 (LFA- 1) interactions with the family of Intercellular Adhesion Molecules (ICAM), and has desirable pharmacokinetic properties, including rapid systemic clearance

A growing body of evidence points to a role for inflammation mediated by lymphocyte function-associated antigen-1 (LFA-1) and its ligand intercellular adhesion molecule-1 in the pathogenesis of diabetic macular oedema. This phase 1b clinical trial assessed the safety, tolerability, and pharmacokinetics of topically administered SAR 1118, a novel LFA-1 antagonist, in human subjects

Topical SAR 1118 was safe and well tolerated, and dose-dependent levels of drug were detected in aqueous. However, vitreous levels were below the threshold of detection with the concentrations tested. Further investigation of this medication for posterior segment applications would require intravitreal delivery or chemical modification of the drug.

In a Phase 2 dry eye trial, subjects receiving SAR 1118 demonstrated a reduction in corneal staining, increased tear production, and improved visual-related function as compared to placebo. These data were part of the scientific program of the Association for Research in Vision and Ophthalmology (ARVO) Annual Meeting held in Fort Lauderdale, Florida. SAR 1118 is a first-in-class topically administered small molecule integrin antagonist that inhibits T-cell mediated inflammation, a key component of dry eye disease.

In the randomized, placebo-controlled, multi-center trial, which included 230 subjects with dry eye disease, SAR 1118 demonstrated dose-dependent significant improvements (p<0.05) in inferior corneal staining over 12 weeks. As early as two weeks, a statistically significant(p<0.05) increase in tear production and improvement in visual-related functions (ability to read, drive at night, use a computer, and watch television) were observed, demonstrating early onset of action. Visual-related function was assessed using the Ocular Surface Disease Index (OSDI), a validated instrument designed to measure the severity of dry eye disease and the impact on vision-related function and quality of life. SAR 1118 was safe and well-tolerated with no serious ocular adverse events reported. Most ocular adverse events were transient and related to initial instillation.

“We are encouraged by the clinical effects of SAR 1118 in improving both signs and symptoms of dry eye, which supports the broad anti-inflammatory mechanism of this novel molecule,” commented Charles Semba, MD, Chief Medical Officer of SARcode Corporation. “We are excited to begin Phase 3 development in the later part of 2011, and have discussed appropriate and acceptable endpoints with the regulatory bodies to facilitate a smooth path towards approval.”

“It is well known that dry eye disease can have a deleterious effect on visual function, daily activities, workplace productivity, and quality of life. The potential to impact a patient’s quality of life in as early as 2 weeks could be a major advance in the current dry eye treatment model,” said Quinton Oswald, Chief Executive Officer of SARcode Corporation. “We hope to achieve similar results in our Phase 3 program.”

About Dry Eye Syndrome

Dry eye syndrome is a prevalent and often chronic condition estimated to affect approximately 20 million people in the US. It is among the most common diseases treated by ophthalmologists throughout the world, and has been shown to have a significant impact upon quality of life. Dry eye varies in severity and etiology, and symptoms most commonly manifest as discomfort, visual disturbances, and tear film instability due to decreased quality or quantity of tears. A major contributing factor towards the development of dry eye is inflammation caused by T-cell infiltration, proliferation and inflammatory cytokine production that can lead to reduction in tear film quality and ocular surface damage.

About SAR 1118 – SAR 1118 is a potent novel small molecule lymphocyte function-associated antigen-1 (LFA-1; CD11a/CD18; alphaLbeta2) antagonist under investigation for a broad range of ocular inflammatory conditions including dry eye and diabetic macular edema. LFA-1 is member of the integrin family of adhesion receptors found on the surface of all leukocytes and represents a therapeutic target central to a number of inflammatory stimuli. SAR 1118 has demonstrated potency in blocking LFA-1 binding to its cognate ligand, intercellular adhesion molecule-1 (ICAM-1; CD54), thereby inhibiting cell adhesion, cytokine production, and cellular proliferation in in vitro models.

About SARcode Corporation – SARcode Corporation, founded in 2006, is a venture-backed ophthalmic biopharmaceutical company based in Brisbane, CA. SARcode’s lead development program is a novel class of lymphocyte function-associated antigen-1 (LFA-1) antagonists for the treatment T-cell mediated inflammatory diseases. Institutional investors include Alta Partners and Clarus Venture Partners. For more information, visit http://www.sarcode.com

Inline image 1

Inline image 1

WO 2006125119

Tom Gadek

Tom Gadek

CEO and President at TearSolutions Inc.
Inventors Thomas GadekJohn Burnier
Applicant Sarcode Corporation

 

PATENT

http://www.google.com.mx/patents/WO2005044817A1?cl=en

EXAMPLE 14 [0305] This example describes the synthesis of

Figure imgf000097_0002

[0306] which was prepared according to Scheme 9 and the procedure below.

[0307] SCHEME 9

Figure imgf000097_0003

[0308] a) To a solution of 3-carboxylbenzenesulfonyl chloride (3.54 g, 16 mmol) in ethyl acetate (50 mL) at 0 °C was added concentrated ammonia (2.5 mL). The reaction was neutralized with HCl in dioance (20 mL), diluted with ethyl acetate (100 mL), dried with anhydrous sodium sulfate and filtered. Concentration of the filtrate yielded the title compound, which was used without purification. [0309] b) Crude compound 14.1 was dissolved in THF (50 mL), to it was added borane (1.0 M in THF, 50 L) over 20 minute period. After the reaction was stirred at room temperature for 15 hours, the reaction was diluted with brine (20 mL) and water (10 mL), extracted with ethyl acetate (100 mL). The organic extract was dried over anhydrous sodium sulfate and filtered. Concentration of the filtrate yielded the title compound, which was used without further purification. [0310] c) To crude compound 14.2 solution in DCM (100 mL) was added activated 4A molecular sieve powder (8 g), pyridinium dichromate (7.55 g, 20 mmol). After the reaction was stirred at room temperature for 2 hours, the reaction mixture was filtered through silica gel (50 g), rinsed with ethyl acetate. The residue after concentration of the filtrate was purified by silca gel column with 30-50% ethyl acetate in hexane to give compound 14.3 (477mg, 16%, 3 steps). ESI-MS (m/z): (M+H4″) 186. [0311] d) Compound 14.4 was made according to Example 8e except that compound 14.3 was used instead of compound 8.7. MS (ESI4) m/z: 260 (M+H4″). [0312] e) Compound 14 was made according to Example 3g except that compound 14.4 was used instead of compound 3.4. 1H NMR (400 MHz, CD3OD) δ 7.89 (s, 1 H), 7.80 (s, 1 H), 7.75 (m, 2 H), 7.64 (s, 1 H), 7.57(d, 1 H), 7.34 (d, 2 H), 6.93 9s, 1 H), 5.00 (m, 1 H), 3.99 (m, 1 H), 3.73 (m, 1 H), 3.40 (dd, 1 H), 3.12 (dd, 1 H), 2.89 (m, 2 H) ppm; ESI-MS (m/z) 616 (M+H4″). [0313] EXAMPLE 15 [0314] This example describes the synthesis of

Figure imgf000098_0001

which was prepared according to Scheme 10 and the procedure below. [0315] SCHEME 10 rr–λ I BuLi, THF m-CPBA

Figure imgf000099_0001

s ) 2. DMF CH2CI2

Figure imgf000099_0002

15.1 15.2

Figure imgf000099_0003

[0316] a) To a solution of 0.2 mol of furan in 200 mL of dry THF was added 0.2 mol of «-BuLi (1.6 M in hexanes) at -78 °C, the resulting solution was stirred at room temperature for 4 hours. Subsequently, the mixture was cooled to -78 °C and treated with 0.21 mol of dimethyl disulfide, and the mixture was stirred at room temperature overnight, followed by adding 10 mL of saturated aqueous NH C1. The mixture was concentrated at room temperature, and the residue was diluted with 200 mL of saturated aqueous NH4C1 and extracted with ether. The extract was then washed with brine and dried with anhydrous Na2SO . The solvent was removed, and the residue was distilled to collect, the fraction at 135-140 °C/760 mmHg to give compound 15.1 in 55% yield. 1H NMR (400 MHz, CD3C1): δ 7.50 (s, IH), 6.45 (m, IH), 6.39 (s, IH), 2.42 (s, 3H) ppm. [0317] b) To a solution of 0.1 mol of compound 15.1 in 100 mL of dry THF was added 0.1 mol of n- uLi (1.6 M in hexanes) at -78 °C, the resulting solution was stirred at room temperature for 4 hours. Subsequently, the mixture was cooled to -78 °C and treated with 0.12 mol of dry DMF, and the mixture was stirred at room temperature overnight. The reaction was quenched by adding 10 mL of saturated aqueous NH4C1, and the mixture was concentrated. The residue was diluted with 100 mL of brine and extracted with EtOAC. The extract was washed with brine and dried with anhydrous Na2SO4. The solvent was removed and the residue was purified to give the title compound in 65% yield. 1H NMR (400 MHz, CD3C1): δ 9.52 (s, IH), 7.24 (d, J= 3.4 Hz, IH), 6.42 (d, J= 3.4Hz, IH), 2.60 (s, 3H) ppm; ESI-MS (m/z) (M+H4) 143.1. [0318] c) A mixture of 50 mmol of compound 15.2 and 120 mmol of -CPBA in 100 mL of CH2C12 was stirred at room temperature overnight. The mixture was diluted with 150 mL of CH2C12, and the mixture was washed with saturated aqueous NaHCO3 for several times. The solution was then dried with anhydrous Na2SO4 and concentrated. The residue was purified to give compound 15.3 in 70% yield. 1H NMR (400 MHz, CD3C1): δ 9.83 (s, IH), 7.33 (m, 2H), 3.27 (s, 3H) ppm; ESI-MS (m/z): (M+H4″) 175.0.

[0319] d) Compound 15.4 was made according to Example 8e except that compound 15.3 was used instead of 8.7. ESI-MS (m/z): (M+H4″) 248.1. [0320] e) Compound 15 was made according to Example except that compound 15.4 was used instead of 3.4. 1H NMR (400 MHz, CD3OD): δ 7.92 (s, IH), 7.76 (m, IH), 7.67 (s, IH), 7.34 (m, IH), 7.13 (s, IH), 6.69 (s, IH), 6.49 (s, IH), 5.11 (m, IH), 4.73 and 4.88 (m, 2H), 3.76 and 4.02 (m, 2H), 3.46 (m, IH), 3.30 (m, IH), 3.17 (s, 3H), 2.94 (m, 2H) ppm; ESI-MS (m/z): (M+H4) 605.05. [0321]

…………………………………….

US 20110092707

http://www.google.com/patents/US20110092707

Formula I:

Figure US20110092707A1-20110421-C00002

has been found to be an effective inhibitor of Lymphocyte Function-Associated Antigen-1 (LFA-1) interactions with the family of Intercellular Adhesion Molecules (ICAM), and has desirable pharmacokinetic properties, including rapid systemic clearance. Improved forms, including crystalline forms, and their uses in treatment of disorders mediated by the interaction of LFA-1 and ICAM are described herein. Novel polymorphs of the compound of Formula I which may afford improved purity, stability, bioavailability and other like characteristics for use in pharmaceutical formulations and methods of use thereof are useful in treating disease.

Methods of Manufacture of the Compound of Formula I

In one embodiment, the compound of Formula I was synthesized as in the following Schemes 1-7. Alternate steps were used in the process as described below. The variants of this overall route yield superior yields, cost of goods and superior chiral purity compared to previously described methods. The final product of this synthesis yields crystalline Form A directly.

Figure US20110092707A1-20110421-C00009

A first alternative protecting strategy produces compound 5, a trityl protected species as shown in Scheme 1. The synthesis begins by reductively aminating 3, 5, dichlorobenzaldehyde, compound 1, with 1-chloro-2-aminoethane and sodium cyanoborohydride in 35% yield. Cyclization of compound 2 using aluminum chloride catalysis and ammonium chloride at 185° C. provided compound 3 in 91% yield. Protection of the free amine of compound 3 as the trityl protected species afforded compound 4 in 89% yield. A carboxylic acid functionality was introduced by treatment of compound 4 with n-butyllithium (nBuLi) and Tetramethylethylenediamine (TMEDA), with subsequent introduction of carbon dioxide, to produce compound 5 in 75% yield.

Figure US20110092707A1-20110421-C00010

Bromophenylalanine was used as the starting material for the right hand portion of the final molecule as shown in Scheme 2. t-Butylcarbamate (Boc) protection of the amino group was accomplished, using sodium bicarbonate (3 equivalents), t-butyl dicarbonate (Boc2O, 1.1 equivalent) in dioxane and water, to obtain compound 7 in 98% yield. A methyl sulfone functionality was introduced by treating the bromo compound 7 with copper iodide (0.4 equivalents), cesium carbonate (0.5 equivalents), L-proline (0.8 equivalents), and the sodium salt of methanesulfinic acid (3.9 equivalents) in dimethylsulfoxide (DMSO) at 95-100° C. for a total of 9 hours, with two further additions of copper iodide (0.2 equivalents) and L-proline (0.4 equivalents) during that period. Compound 8 was isolated in 96% yield. The carboxylic acid of compound 8 was converted to the benzyl ester, compound 9, in 99% yield, using benzyl alcohol (1.1 equivalent), dimethylaminopyridine (DMAP, 0.1 equivalent) and N-(3-dimethylaminopropyl)-N-ethylcarbodiimide (EDC, 1.0 equivalent). The amino group of compound 9 is deprotected by adding a 4N solution of HCl in dioxane to compound 9 at 0° C. in methylene chloride. The HCl salt of the free amino species, compound 10 was isolated in 94% yield.

Figure US20110092707A1-20110421-C00011

Compound 5 was treated with triethylamine (TEA, 5 equivalents) and 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU, 1.25 equivalents) for 10 minutes in dimethylformamide (DMF), and then compound 10 was added to the solution. After stirring at room temperature for 18 hours, the product, compound 11 was isolated in 70% yield. Removal of the trityl protecting group was accomplished by treating compound 1, with HCl in dioxane (4N, excess) at room temperature for 2 hours, diethyl ether added, and the solid product, compound 12, was isolated by filtration in 95% yield.

Figure US20110092707A1-20110421-C00012

The benzofuranyl carbonyl moiety of the compound of Formula I was prepared using two alternative schemes, Scheme 4 and Scheme 4″. In one embodiment, the benzofuranyl carbonyl moiety was prepared by protecting the hydroxyl group of compound 13 by reacting with tert-butyldimethylsilyl chloride (1.0 equivalents) and triethylamine (TEA, 1.1 equivalents) in acetone, to give compound 14 in 79% yield. A solution of compound 14 in methanol was then treated with sodium borohydride (1.0 equivalent) at room temperature overnight. The reaction was quenched with an addition of acetone, stirred at room temperature for a further 2.5 hours, aqueous HCl (4N) was added with the temperature controlled to below 28C, tetrahydrofuran (THF) was added, and the solution stirred overnight under argon and in the absence of light. The product, compound 15, was isolated quantitatively by extraction into methylene chloride, concentrated at low heat, and used without further purification. The triflate ester, compound 16, was produced in 69% yield from compound 15 by reacting it with N-phenyl-bis(trifluoromethanesulfonimide) (1.0 equivalent) in methylene chloride for 72 hours. Compound 16 in a mixture of DMF, methanol, and triethylamine, was added to a prepared solution of palladium acetate, diphenyl, DMF and methanol in an autoclave. Carbon monoxide was charged into the autoclave to a pressure of 8 bar, and the reaction mixture was heated at 70° C. for 6 hours. After workup, compound 17 was isolated in 91% yield. Lithium hydroxide (4 equivalents) in methanol and water was used to hydrolyze the ester and permit the isolation of compound 18 in 97% yield.

Figure US20110092707A1-20110421-C00013

In one embodiment, the benzofuranyl carbonyl moiety of the compound of Formula I was prepared according to Scheme 4″. By way of an Arbuzov reaction, diethyl 2-(1,3-dioxolan-2-yl)ethylphosphonate, compound 1″, was prepared from 2-(2-bromoethyl)-1,3-dioxolane by the addition of triethyl phosphate. After removal of ethyl bromide through distillation at 210° C. the crude reaction mixture was cooled and then by way of vacuum distillation, compound 1″ was collected as a colorless oil in 94% yield.

In the next step, n-butyllithium (2.15 equivalents) in hexane was cooled to −70° C. and diisopropylamine (2.25 equivalents) was added while keeping the temperature below −60° C. Compound 1″ (1 equivalent) dissolved in tetrahydrofuran (THF) was added over 30 min at −70° C. After 10 min, diethyl carbonate (1.05 equivalents) dissolved in THF was added over 30 min keeping the reaction temperature below −60° C. After stirring for one hour at −60° C., the reaction was allowed to warm to 15° C. and furan-2-carbaldehyde (1.3 equivalents) dissolved in THF was added. After stirring for 20 hrs at room temperature, the reaction was rotary evaporated to dryness to yield ethyl 2-(1,3-dioxolan2-yl)methyl-3-(furan-2-yl)acrylate, compound 5″. Crude compound 5″ was used directly in the next reaction.

The crude compound 5″ (1 equivalent) was dissolved in ethanol and added to a mixture of water and phosphoric acid (85%, 15 equivalents) over 30 min while keeping the temperature below 50° C. After stirring for 20 hrs at room temperature, another 200 ml of phosphoric acid (85%) was added and the mixture was heated to 50° C. for an additional two hrs. After removal of ethanol by rotary evaporation, the material was extracted with toluene, washed with water, dried with sodium sulfate, treated with charcoal, filtered and dried down to an oil. This oil was distilled to afford ethyl benzofuran-6-carboxylate, compound 6″, (bp 111-114.5° C.) which crystallized on standing. Compound 6″ was recovered at 57% yield based on compound 1″.

Compound 6″ (875 mmol) was dissolved in methanol and tetrahydrofuran (THF). Sodium hydroxide (4 M, 3 equivalents) was added and the reaction was stirred overnight. After concentration via rotary evaporation, the aqueous solution was extracted with methyl tert-butyl ether (MTBE), acidified to pH 2 with the addition of hydrochloric acid (HCl) and cooled resulting in fine crystals of benzofuran-6-carboxylic acid, i.e., compound 18. Compound 18 was isolated, washed with water and dried to a final yield of 97% yield.

Figure US20110092707A1-20110421-C00014

The benzofuran carboxylic acid 18 was treated with oxalyl chloride (1.2 equivalents) and a catalytic amount of DMF, stirring for 5.5 hours until a clear solution was obtained. The solvent was removed under reduced pressure and the acid chloride of compound 18 was stored under argon until use, on the next day. The acid chloride, in methylene chloride was added slowly to a methylene chloride solution of the compound of Formula I and diisopropylethylamine (DIPEA) which was cooled to 0-5° C. The reaction was not permitted to rise above 5° C., and after completion of addition, was stirred at 5° C. for a further 0.5 hour. Upon aqueous workup and extraction with methylene chloride, the product, compound 19, was isolated in quantitative yield.

Taking the compound of Formula I directly as the crude reaction product after transfer hydrogenolysis, and reconcentrating down from a solution in methylene chloride, the amorphous form of the compound of Formula I was obtained in 97% purity.

Figure US20110092707A1-20110421-C00015

An alternative protection strategy in this synthetic approach is illustrated in Scheme 6.

…………………….

WO 2014018748

http://www.google.com/patents/WO2014018748A1?cl=en

[0040] Methods of Manufacture of the Compound of Formula I

Figure imgf000009_0001

[0041] In one embodiment, the compound of Formula I is synthesized as in the following Schemes 1-7. The final product of this synthesis yields the compound of Formula I as an amorphous solid or as a crystalline form such as Forms A-E, or a pharmaceutically acceptable salt, either directly or indirectly. Variants of this overall route may provide superior yields, cost of goods, and/or superior chiral purity.

[0042] Protecting groups for amino and carboxy groups are known in the art. For example, see Greene, Protective Groups in Organic Synthesis, Wiley Interscience, 1981, and subsequent editions.

[0043] In various embodiments in the subsequent schemes, HATU is used as a reagent in amide- bond forming reactions. Alternatively, HATU is not used. In various embodiments, at least one amide-bond forming reaction is performed with thionyl chloride as a reagent in place of HATU. In various embodiments, all amide-bond forming reactions are performed with thionyl chloride as a reagent to form acid chlorides.

[0044] Scheme 1

Figure imgf000011_0001

[0045] A first alternative protecting strategy produces compound 5′, a protected species as shown in Scheme 1. The synthesis begins by reductively aminating 3, 5, dichlorobenzaldehyde, compound . Cyclization of compound 2′ provides compound 3′. Protection of the free amine of compound 3′ as a protected species provides compound 4′. A carboxylic acid functionality is introduced by treatment of compound 4′ with introduction of carbon dioxide, to produce compound 5′. In various embodiments, the protecting group of compound 4′ is a benzofuranyl carbonyl moiety derived from compound 18′.

[0046] In various embodiments, upon scaleup to multikilogram and larger scale reactions, treatment of compound 4′ with strong base (such as n-butyllithium (nBuLi) to generate a lithio species, or lithium diisopropyl amide (LDA) to generate the lithio species) is performed in flow mode rather than batchwise reaction due to instability of lithio species except at cold temperatures. Flow rates and residence times may be adjusted to maximize yield.

[0047] Scheme IB

Figure imgf000012_0001
Figure imgf000012_0002

5′ 4″”

[0048] In various embodiments, 6-hydroxy-l, 2,3, 4-tetrahydro-isoquino line (Compound 3″) is used as a starting material for Compound 5′. The starting material is chlorinated (x2) for example, with N-chlorosuccinimide. In various embodiments, the chlorination is performed in the presence of a sulfonic acid. In various embodiments, the sulfonic acid is selected from p- toluenesulfonic acid and methanesulfonic acid. Following protection of the amino group, the hydroxy group is functionalized, for example, as the triflate ester, which is carbonylated to yield the amino-protected methyl ester. Hydrolysis of the methyl ester yields the amino protected carboxylic acid.

[0049] Scheme 2

Figure imgf000012_0003

[0050] In various embodiments, bromophenyl alanine is used as the starting material for a portion of the final molecule as shown in Scheme 2. The starting material is protected with an amino protecting group to allow for introduction of a methyl sulfone functionality in compound 8′. Protecting groups are rearranged by introduction of an orthogonal protecting group for the carboxylic moiety, followed by deprotection of the amino group to provide compound 10′. In various embodiments, expensive or exotic bases are replaced with carbonate base such as potassium carbonate or calcium carbonate as a reagent.

[0051] Scheme 2A

Figure imgf000013_0001

10

[0052] In various embodiments, 3-methylsulfonylbenzaldehyde is converted into the 3- methylsulfonylphenylalanine derivative and functionalized to yield compound 10 as shown above.

[0053] Scheme 3

Figure imgf000014_0001

12′

[0054] Compounds 5′ and 10′ are joined through amide bond formation followed by deprotection of the remaining amino group in the presence of the carboxylic protecting group to yield compound 12′ or a salt thereof, such as the HCL salt.

[0055] Scheme 3

Figure imgf000014_0002

[0056] As an alternative to Scheme 3, compound 10″ is coupled with compound 5′ to yield the bromo compound 12″”, with subsequent introduction of a methyl sulfone functionality in place of bromine at a later step to produce compound 19′. Alternatively, instead of a bromine, compound 10″ includes X, where X is any halide (CI, I, Br, F) or a leaving group such as OTs, OTf, or the like.

[0057] Scheme 4

Figure imgf000015_0001

[0058] The benzofuranyl carbonyl moiety of the compound of Formula I can be prepared using various alternative schemes. In one embodiment, the benzofuranyl carbonyl moiety is prepared by protecting the hydroxyl group of compound 13′, reducing the carbonyl of compound 13′ to yield the benzofuranyl moiety, followed by carboxylation to yield compound 18′.

[0059] Scheme 4A

[0060] In one embodiment, compound 18′ is prepared from 6-hydroxybenzofuran via the triflate ester and the 6-carboxy methyl ester as intermediates, as shown in Example 4A.

[0061] Schem

Figure imgf000015_0002

[0062] The benzofuran carboxylic acid 18′ is coupled with compound 12′ (or a salt thereof) by amide bond formation to yield protected compound 19′, as shown in Scheme 5. Amide bond formation is known in the art

[0063] Schem

Figure imgf000016_0001

[0064] As an alternative to Schemes 3-5, compounds 18′ and 5″ may be coupled through amide bond formation followed by deprotection of the remaining carboxylic group to form compound 12″. Amide bond formation between compound 12″ and 10′ yields compound 19′ with a protected carboxylic group.

[0065] Scheme 5B

Figure imgf000017_0001

[0066] As an alternative to Schemes 1-5, compounds 12″ and 10″ may be coupled through amide bond formation followed by introduction of a methyl sulfone functionality in place of the bromine in converting compound 19″ to compound 19′ (similar to Scheme 2). Alternatively, instead of a bromine, compound 10″ includes X, where X is any halide (CI, I, Br, F) or a leaving group such as OTs, OTf, or the like. Compound 12″ can also be made using the following scheme:

Figure imgf000018_0001

[0067] Scheme 6

Figure imgf000018_0002

[0068] Final deprotection of compound 19′ to yield the compound of Formula I or a salt thereof is accomplished in a variety of ways. In various embodiments, the resulting compound of Formula I is provided in higher optical purity and/or higher overall purity and/or higher overall yield.

EXAMPLES

[00111] Example 1

Figure imgf000029_0001

Scheme El

[00112] Reductively aminating 3,5-dichlorobenzaldehyde, compound 1, with l-chloro-2- aminoethane and sodium cyanoborohydride provided 35% yield of compound 2. Cyclization of compound 2 using aluminum chloride catalysis and ammoniun chloride at 185°C provided compound 3 in 91% yield. Protection of the free amine of compound 3 as the trityl protected species afforded compound 4 in 89%> yield. A carboxylic acid functionality was introduced by treatment of compound 4 with n-butyllithium (nBuLi) and tetramethylethylenediamine (TMEDA), with subsequent introduction of carbon dioxide, to produce trityl protected compound 5 in 75% yield.

[00113] Example 1 A

Figure imgf000030_0001

2″

Figure imgf000030_0002

Scheme El A

[00114] To a glass reactor was charged 3,5-dichlorobenzaldehyde. Absolute ethanol was added to the batch slowly (this addition is mildly exothermic) and agitation started. 2,2- Diethoxyethyl amine (1.03 equiv) was slowly added to the batch, keeping the batch temperature at 20-78 °C. The batch was then heated to 76-78 °C for 2 h. GC-MS analysis indicated reaction completion (starting material < 1%). The batch was cooled to ambient temperature for work-up. The batch was concentrated in vacuo to a residue and azeotroped with heptanes (x2). The residue was cooled and held at 0-5 °C for 12 h to form a suspension. The solids were collected by filtration and the cake was washed with cold (0-5 °C) heptanes, and dried under hot nitrogen (45-50 °C) to afford Compound 2′ as a white solid (94% yield).

[00115] To a glass reactor was charged concentrated 95-98%) sulfuric acid (25.9 equiv).

The batch was heated to 120-125 °C and a solution of Compound 2′ in CH2CI2 was added slowly over 1 h, keeping the batch temperature between 120-125 °C. The batch was then stirred at 120— 125 °C for 6 h. The batch was cooled to < 50 °C. To a glass reactor was charged DI water and the batch temperature was adjusted to 0-5 °C. The reaction mixture was slowly transferred, keeping the batch temperature between 0-50 °C. DI water was used to aid the transfer. To the batch was added Dicalite 4200. The batch was filtered through a pad of Dicalite 4200. To the filtrate was added 50% aqueous sodium hydroxide solution slowly over 3 h, keeping the batch temperature between 0-50 °C to adjust the pH to 12. The resulting suspension was stirred at 45- 50 °C for 2 h and the solids were collected by filtration. The filter cake was slurried in DI water at 30-35 °C for 1 h. The batch was filtered. The cake was washed with heptanes and dried in vacuum oven at 45-50 °C for 22 h to give crude compound 2″ as a tan solid (75% yield), which was further purified by recrystallization.

[00116] To a reactor was added platinum dioxide (0.012 equiv), Compound 2″, and

MeOH (10 vol) and the suspension was stirred at room temperature under argon for 10 minutes. The reaction mixture was inerted with argon three times and then stirred under 125 psi of hydrogen at room temperature for 25 hours. HPLC analysis indicated complete reaction with less than 1% of the starting material remaining. After standing, the supernatant was decanted from the solids (catalyst) by vacuum. To the solids was added methanol and the slurry was mixed under nitrogen. The solids were allowed to settle on the bottom over several hours. The supernatant was decanted from the solids by vacuum. The combined supernatants were filtered through Celite under a blanket of nitrogen and the filter pad was washed with MeOH (x2). The combined filtrate and washes were concentrated to dryness. The residue was slurried in MTBE. The mixture was treated with 3 M HC1 while maintaining the temperature <40 °C resulting in the formation of a heavy precipitate. The mixture was stirred at 35-40 °C for 60 to 90 minutes. The batch was cooled to 0-5 °C, stirred for 60 to 90 minutes and then filtered. The filter cake was washed with cold DI water (x2) followed by a displacement wash with MTBE (x2). The filter cake was dried under reduced pressure to afford Compound 3 Hydrochloride Salt (86% yield). The hydrogenation catalyst can be recovered and re-used.

[00117] Compound 3 and trityl chloride were added to the reaction flask. DCM (10 vol) was added to the reactor and agitation was started to form slurry. The reaction mixture was cooled to 10-15 °C. N,N-Diisopropylethylamine (2.5 equiv) was slowly added to the reaction mixture, maintaining the temperature at 15-25 °C during the addition. Once addition was complete, the batch was stirred at 15 to 25 °C for a minimum of 60 minutes. The reaction was assayed by HPLC by diluting a sample with acetonitrile and then injecting it on the HPLC. The first assay after 30 minutes indicated that the reaction was complete with <1% of starting material observed by HPLC analysis. The reaction mixture was diluted with DI water (5 vol). The reaction mixture was stirred for 5 minutes after which it was transferred into a separation funnel and the phases were allowed to separate. The DCM layer was washed with DI water (5 vol) by stirring for 5 minutes and then allowing the phases to separate. The DCM layer was washed with brine (5 vol) by stirring for 5 minutes and then allowing the phases to separate. The DCM layer was dried over magnesium sulfate, filtered and the filter cake was washed with DCM (x2). The combined filtrate and washes were concentrated to a residue that was azeotroped with EtOAc (x2). The residue was suspended in EtOAc and stirred for 1 hour in a 40 °C water bath. The resulting slurry was cooled to 0-5 °C for 1 hour and then filtered. The filter cake was washed twice with EtOAc and then dried under reduced pressure to afford Compound 4.

[00118] Exam le IB

Figure imgf000032_0001

21 4″

[00119] To 1, 2,3, 4-tetrahydro-6-hydroxy-isoqino line in acetonitrile was added p- toluenesulfonic acid and N-chlorosuccinimide. The suspension was cooled to ambient temperature, and the product isolated by filtration for a yield of approximately 61% with purity greater than 95%. The isolated TsOH salt was recrystallized until purity was greater than 99.7%. To one equivalent of the TsOH salt suspended in methanol was added 2M sodium carbonate (0.55 eq.) and 1.2 eq. of Boc anhydride. The suspension was stirred at room temperature overnight. The reaction was monitored by HPLC. Upon completion, the mixture was cooled to below 10 °C, water was added, and the Boc-protected dichloro compound was isolated by filtraton. The product was washed and dried at 40 °C for a yield of 95% and purity of >97%. The Boc-protected dichloro compound was suspended in dichloromethane (10 volumes) and pyridine (5 volumes) was added. The mixture was cooled to below 2 °C, and triflic anhydride (1.25 eq) was added. The mixture was stirred at 0-2 °C for 10 minutes, and then poured into 10 volumes of 6%) aqueous sodium hydrogen carbonate solution. After washing with dichloromethane, the organic phases were combined and dried over magnesium sulphate. Following purification, the product (Compound 4′) was obtained in 90% yield and >98% purity. Compound 4′ was dissolved in dimethylformamide and methanol at room temperature. Diisopropylamine (4 eq) was added. Under CO atmosphere, l,3-bis(diphenylphosphino)propane (0.1 eq) and palladium acetate (0.1 eq) was added. The reaction was heated to refiux, and monitored by HPLC. Upon near completion, the mixture was cooled to ambient temperature. Workup with water, ethyl aceate, and brine yielded Compound 4″, which was used without further purification. Compound 4″ was dissolved in methanol and 2.4 M sodium hydroxide (10 volumes each) and refiuxed. The mixture was cooled to ambient temperature, and toluene was added. Following aqueous workup, the pH was adjusted to 2.3 with 3M hydrochloric acid, and crude product was isolated by filtration in 53% yield with greater than 80% purity.

[00120] Exam le 2

Figure imgf000033_0001

Scheme E2

[00121] t-Butylcarbamate (Boc) protection of the amino group of bromophenyl alanine was accomplished, using sodium bicarbonate (3 equivalents), t-butyl dicarbonate (Boc20, 1.1 equivalent) in dioxane and water, to obtain compound 7 in 98% yield. A methyl sulfone functionality was introduced by treating the bromo compound 7 with copper iodide (0.4 equivalents), cesium carbonate (0.5 equivalents), L-proline (0.8 equivalents), and the sodium salt of methanesulfinic acid (3.9 equivalents) in dimethylsulfoxide (DMSO) at 95-100°C for a total of 9 hours, with two further additions of copper iodide (0.2 equivalents) and L-proline (0.4 equivalents) during that period. Compound 8 was isolated in 96%> yield. The carboxylic acid of compound 8 was converted to the benzyl ester, compound 9, in 99% yield, using benzyl alcohol (1.1 equivalent), dimethylaminopyridine (DMAP, 0.1 equivalent) and N-(3- dimethylaminopropyl)-N-ethylcarbodiimide (EDC, 1.0 equivalent). The amino group of compound 9 is deprotected by adding a 4N solution of HC1 in dioxane to compound 9 at 0°C in methylene chloride. The HCl salt of the free amino species, compound 10 was isolated in 94% yield.

[00122] Example 2 A

[00123] Example 2 was repeated with potassium carbonate in place of cesium carbonate.

[00124] Example 2B

[00125] Boc-protected bromophenylalanine (Compound 7) (100g) was dissolved in

DMSO (400 mL) with stirring and degassing with argon. Sodium methane sulfmate (98g), copper iodide (28.7g), potassium carbonate (40 g) and L-proline (26.75g) were added at 28-30 °C. Reaction was heated to about 87 °C for about 17-19 hours. Reaction was cooled and quenched with crushed ice, stirred for 30-40 minutes, and the pH was adjusted from about 12 to about 3-4 with citric acid (350 g). Quenched reaction mixture was filtered, extracted with dichloromethane x3, washed with ammonium chloride solution, washed with sodium bisulphite solution, and washed with brine. Crude product in dichloromethane was concentrated in vacuo until moisture content was below about 0.5%, and used in next step without further isolation. Crude compound 8 in dichloromethane was charged with benzyl alcohol and DMPA with stirring under nitrogen. Reaction cooled to 0-5 °C. EDC-HCL (1.03 equiv) added with stirring for 30 minutes. Upon completion of reaction by TLC and HPLC, the reaction was quenched with sodium bicarbonate solution, the organic layer was separated, and the aqueous layer was extracted with dichloromethane. The organic layer was washed with citric acid solution, and combined organic layers were washed with brine solution. Dichloromethane was removed at 45- 50 °C, and the concentrate was used for next step without further isolation. The amino group of compound 9 was deprotected by adding a 4N solution of HCl in dioxane to compound 9 at 10- 15°C in methylene chloride. The HCl salt of the free amino species, compound 10 was isolated by filtration from diethyl ether. Isolation of compound 10 was performed through recrystallization using a dimethylformamide/dichloromethane solvent system.

[00126] Example 3

Figure imgf000035_0001

Scheme E3

[00127] Compound 5 was treated with triethylamine (TEA, 5 equivalents) and 2-(7-Aza- lH-benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate (HATU, 1.25 equivalents) for 10 minutes in dimethylformamide (DMF), and then compound 10 was added to the solution. After stirring at room temperature for 18 hours, the product, compound 11 was isolated in 70% yield. Removal of the trityl protecting group was accomplished by treating compound 11, with HC1 in dioxane (4 N, excess) at room temperature for 2 hours, diethyl ether added, and the solid product, compound 12, was isolated by filtration in 95% yield. The compound 12 exists in both amorphous and crystalline form and can be isolated in either form.

[00128] Example 3 A

[00129] Compound 5 was dissolved in isopropyl acetate and cooled to 20 to 25 °C.

Thionyl chloride was added, with cooling to 10 to 15 °C, and N-methylmorpholine was added slowly. The reaction was monitored by HPLC. Compound 10, water, and isopropyl acetate were stirred at 15 to 20°C until a solution was achieved. N-methylmorpholine was added followed by addition of the Compound 5 reaction mixture (acid chloride of Compound 5). The reaction was monitored by HPLC. Upon completion, the biphasic layers were allowed to settle, and the aqueous layer was removed. The upper organic layer was extracted with water, and the remaining organic layer was distilled under vacuum. Dioxane and IpAc were added with further distillation. Once dry, 4N anhydrous HC1 in dioxane was added. The mixture was stirred at 20 to 25°C for 12 hours, and checked for complete deprotection by HPLC. Once complete, the thick slurry was filtered, washed with IP Ac and dried under vacuum at 45 to 55°C. Yield of Compound 12 was 88%.

[00130] Example 4

[00131] The benzofuranyl carbonyl moiety of the compound of Formula I was prepared using various schemes, (Schemes E4, E4A, and E4B).

Figure imgf000036_0001

15

Phenyl-bis-triflate

Figure imgf000036_0002

18 ‘

Scheme E4

[00132] The benzofuranyl carbonyl moiety was prepared by protecting the hydroxyl group of compound 13 by reacting with tert-butyldimethylsilyl chloride (1.0 equivalents) and triethylamine (TEA, 1.1 equivalents) in acetone, to give compound 14 in 79% yield. A solution of compound 14 in methanol was then treated with sodium borohydride (1.0 equivalent) at room temperature overnight. The reaction was quenched with an addition of acetone, stirred at room temperature for a further 2.5 hours, aqueous HCl (4N) was added with the temperature controlled to below 28 °C, tetrahydrofuran (THF) was added, and the solution stirred overnight under argon and in the absence of light. The product, compound 15, was isolated quantitatively by extraction into methylene chloride, concentrated at low heat, and used without further purification. The triflate ester, compound 16, was produced in 69% yield from compound 15 by reacting it with N- phenyl-bis(trifluoromethanesulfonimide) (1.0 equivalent) in methylene chloride for 72 hours. Compound 16 in a mixture of DMF, methanol, and triethylamine, was added to a prepared solution of palladium acetate, l,3-Bis(diphenylphosphino)propane (dppp), DMF and methanol in an autoclave. Carbon monoxide was charged into the autoclave to a pressure of 8 bar, and the reaction mixture was heated at 70 °C for 6 hours. After workup, compound 17 was isolated in 91% yield. Lithium hydroxide (4 equivalents) in methanol and water was used to hydro lyze the ester and permit the isolation of compound 18′ in 97% yield.

[00133] Example 4A

[00134] Example 4 was repeated with triflic anhydride and sodium hydroxide as reagents for the ester hydrolysis.

[00135] Compound 15 (6-Hydroxybenzofuran) was stirred in dichloromethane and diisopropylethylamine. Triflic anhydride (1.2 eq.) was added, keeping the temperature below 20C. The reaction was monitored by HPLC. The reaction was quenched with methanol, solvent was removed with vacuum, and the crude residue of Compound 16 was used without further purification. Compound 16 as crude residue was dissolved in 4 volumes of dimethylformamide and 2 volumes methanol. To the solution was added 0.02 eq. of palladium acetate, 0.02 eq. of dppp, and CO under pressure. The reaction was monitored by HPLC. Following workup, Compound 17 was isolated as a crude oily residue without further purification. The residue of compound 17 was dissolved in methanol (5 volumes) and 1 volume of sodium hydroxide (27.65%) was added. The mixture was heated to 40C until full conversion of HPLC. The mixture was cooled to ambient temperature and 3 volumes of water were added. The pH was adjusted to about 2 with 3M hydrochloric acid. The suspension was filtered, washed with water, and dried to give Compound 18’ in about 75% overall yield with purity >99.5%.

[00136] Example 4B

Figure imgf000037_0001

Scheme E4B [00137] Diethyl 2-(l,3-dioxolan-2-yl)ethylphosphonate, compound 1″, was prepared from

2-(2-bromoethyl)-l,3-dioxolane by the addition of triethyl phosphate. After removal of ethyl bromide through distillation at 210°C the crude reaction mixture was cooled and then by way of vacuum distillation, compound 1″ was collected as a colorless oil in 94% yield.

[00138] In the next step, n-butyllithium (2.15 equivalents) in hexane was cooled to -70 °C and diisopropylamine (2.25 equivalents) was added while keeping the temperature below -60 °C. Compound 1″ (1 equivalent) dissolved in tetrahydrofuran (THF) was added over 30 min at -70 °C. After 10 min, diethyl carbonate (1.05 equivalents) dissolved in THF was added over 30 min keeping the reaction temperature below -60 °C. After stirring for one hour at -60 °C, the reaction was allowed to warm to 15 °C and furan-2-carbaldehyde (1.3 equivalents) dissolved in THF was added. After stirring for 20 hrs at room temperature, the reaction was rotary evaporated to dryness to yield ethyl 2-((l,3-dioxolan2-yl)methyl-3-(furan-2-yl)acrylate, which was used directly in the next reaction.

[00139] The crude compound (1 equivalent) was dissolved in ethanol and added to a mixture of water and phosphoric acid (85%>, 15 equivalents) over 30 min while keeping the temperature below 50°C. After stirring for 20 hrs at room temperature, another 200 ml of phosphoric acid (85%>) was added and the mixture was heated to 50 °C for an additional two hrs.

After removal of ethanol by rotary evaporation, the material was extacted with toluene, washed with water, dried with sodium sulfate, treated with charcoal, filtered and dried down to an oil. This oil was distilled to afford ethyl benzofuran-6-carboxylate, compound 6″, (bp 111-114.5°C) which crystallized on standing. Compound 6″ was recovered at 57%> yield based on compound

1″.

[00140] Compound 6″ (875 mmol) was dissolved in methanol and tetrahydrofuran (THF).

Sodium hydroxide (4 M, 3 equivalents) was added and the reaction was stirred overnight. After concentration via rotary evaporation, the aqueous solution was extracted with methyl tert-butyl ether (MTBE), acidified to pH 2 with the addition of hydrochloric acid (HC1) and cooled resulting in fine crystals of benzofuran-6-carboxylic acid, i.e., compound 18′. Compound 18′ was isolated, washed with water and dried to a final yield of 97%> yield.

[00141] Example 5

Figure imgf000039_0001

10% Pd/C, HCOOH/NEt3

MeOH/THF 5:1

Figure imgf000039_0002

Form A of Formula I

Scheme E5

[00142] The benzofuran carboxylic acid 18′ was treated with oxalyl chloride (1.2 equivalents) and a catalytic amount of DMF, stirring for 5.5 hours until a clear solution was obtained. The solvent was removed under reduced pressure and the acid chloride of compound 18′ was stored under argon until use, on the next day. The acid chloride, in methylene chloride was added slowly to a methylene chloride solution of the compound of Formula 12 and diisopropylethylamine (DIPEA) which was cooled to 0-5 °C. The reaction was not permitted to rise above 5°C, and after completion of addition, was stirred at 5°C for a further 0.5 hour. Upon aqueous workup and extraction with methylene chloride, the product, compound 19, was isolated in quantitative yield.

[00143] The benzyl ester of compound 19 was removed by transfer hydrogenolysis using

10% palladium on carbon, using formic acid and triethylamine in a 5: 1 mixture of methanol:THF, to produce the compound of Formula I in 95% yield.

[00144] A final step of slurrying in methyl ethylketone (MEK) produced Form A of the compound of Formula I. The product was washed with water to remove residual MEK. Alternatively, the product of the hydrogenolysis step was slurried in acetonitrile to yield Form A of the compound of Formula I.

[00145] Taking the compound of Formula I directly as the crude reaction product after transfer hydrogenolysis, and reconcentrating down from a solution in methylene chloride, the amorphous form of the compound of Formula I was obtained in 97% purity.

[00146] Example 6

[00147] An alternative protection strategy was performed in Scheme E6.

Figure imgf000040_0001

Scheme E6

[00148] Boc-protection was used for the ring nitrogen in the intermediates 21 and 22.

Compound 5 was deprotected with HC1 in dioxane to produce compound 23 in better than 97%> yield. Boc-protection was introduced, using di-tert-butyl dicarbonate (1.1 equivalent), and compound 21 was obtained in better than 95% yield. Compound 10 was coupled with compound 21 to obtain compound 22, using HATU and triethylamine in DMF. The product, compound 22, was obtained in quantitative yield, and greater than 90% purity. Deprotection with HC1 yielded the compound of Formula 12 in 97.4% yield.

[00149] Transfer hydrogeno lysis of compound 19 produced the compound of Formula I with optical purity of 98.5% (S) enantiomer compared to 79-94.5% (S) enantiomer optical purity obtained by hydrolysis of the corresponding methyl ester.

PATENT

http://www.google.co.in/patents/WO2009139817A2?cl=en

The compound of Formula I:

Figure imgf000002_0001

Formula I has been found to be an effective inhibitor of Lymphocyte Function- Associated Antigen -1 (LFA-I) interactions with the family of Intercellular Adhesion Molecules (ICAM), and has desirable pharmacokinetic properties, including rapid systemic clearance. Improved forms, including crystalline forms, and their uses in treatment of disorders mediated by the interaction of LFA-I and ICAM are described herein. Novel polymorphs of the compound of Formula I which may afford improved purity, stability, bioavailability and other like characteristics for use in pharmaceutical formulations and methods of use thereof are useful in treating disease.

Sch

Scheme 1

Figure imgf000032_0001

[00174] The first alternative protecting strategy produces compound 5, a trityl protected species as shown in Scheme 1. The synthesis begins by reductively aminating 3, 5, dichlorobenzaldehyde, compound 1, with 1-chloro- 2-aminoethane and sodium cyanoborohydride in 35% yield. Cyclization of compound 2 using aluminum chloride catalysis and ammoniun chloride at 185°C provided compound 3 in 91% yield. Protection of the free amine of compound 3 as the trityl protected species afforded compound 4 in 89% yield. A carboxylic acid functionality was introduced by treatment of compound 4 with n-butyllithium and Tetramethylethylenediamine (TMEDA), with subsequent introduction of carbon dioxide, to produce compound 5 in 75% yield. Scheme 2

l

Figure imgf000033_0001

[00175] Bromophenylalanine was used as the starting material for the right hand portion of the final molecule as shown in Scheme 2. t-Butylcarbamate (Boc) protection of the amino group was accomplished, using sodium bicarbonate (3 equivalents), f-butyl dicarbonate (BoC2O, 1.1 equivalent) in dioxane and water, to obtain compound

7 in 98% yield. A methyl sulfone functionality was introduced by treating the bromo compound 7 with copper iodide (0.4 equivalents), cesium carbonate (0.5 equivalents), L-proline ( 0.8 equivalents), and the sodium salt of methanesulfinic acid (3.9 equivalents) in dimethylsulfoxide (DMSO) at 95-1000C for a total of 9 hours , with two further additions of copper iodide (0.2 equivalents) ) and L-proline ( 0.4 equivalents) during that period. Compound

8 was isolated in 96% yield. The carboxylic acid of compound 8 was converted to the benzyl ester, compound 9, in 99% yield, using benzyl alcohol (1.1 equivalent), dimethylaminopyridine (DMAP, 0.1 equivalent) andN-(3- dimethylaminopropyl)-N-ethylcarbodiimide (EDC, 1.0 equivalent). The amino group of compound 9 is deprotected by adding a 4N solution of HCl in dioxane to compound 9 at O0C in methylene chloride. The HCl salt of the free amino species, compound 10 was isolated in 94% yield.

Scheme 3

Figure imgf000034_0001

DMF

Figure imgf000034_0002

[00176] Compound 5 was treated with triethylamine (TEA, 5 equivalents) and 2-(7-Aza-lH-benzotriazole-l-yl)- 1,1,3,3-tetramethyluronium hexafluorophosphate (HATU, 1.25 equivalents) for 10 minutes in dimethylformamide (DMF), and then compound 10 was added to the solution. After stirring at room temperature for 18 hours, the product, compound 11 was isolated in 70% yield. Removal of the trityl protecting group was accomplished by treating compound 1, with HCL in dioxane (4N, excess) at room temperature for 2 hours, diethyl ether added, and the solid product, compound 12, was isolated by filtration in 95% yield.

scheme 4

Figure imgf000034_0003
Figure imgf000034_0004

The benzofuranyl carbonyl moiety of the compound of Formula I was prepared by protecting the hydroxyl group of compound 13 by reacting with tert- butyldimethylsilyl chloride (1.0 equivalents) and TEA (1.1 equivalents) in acetone, to give compound 14 in 79% yield. A solution of compound 14 in methanol was then treated with sodium borohydride (1.0 equivalent) at room temperature overnight. The reaction was quenched with an addition of acetone, stirred at room temperature for a further 2.5 hours, aqueous HCl (4N) was added with the temperature controlled to below 28C, tetrahydrofuran (THF) was added, and the solution stirred overnight under argon and in the absence of light. The product, compound 15, was isolated quantitatively by extraction into methylene chloride, concentrated at low heat, and used without further purification. The triflate ester, compound 16, was produced in 69% yield from compound 15 by reacting it with N- phenyl-bis(trifluoromethanesulfonimide) (1.0 equivalent) in methylene chloride for 72 hours. Compound 16 in a mixture of DMF, methanol, and triethylamine, was added to a prepared solution of palladium acetate, diphenyl, DMF and methanol in an autoclave. Carbon monoxide was charged into the autoclave to a pressure of 8 bar, and the reaction mixture was heated at 700C for 6 hours. After workup, compound 17 was isolated in 91% yield. Lithium hydroxide (4 equivalents) in methanol and water was used to hydrolyze the ester and permit the isolation of compound 18 in 97% yield.

Scheme 5

Figure imgf000035_0001

10% Pd/C, HCOOH/NEt3 MeOH/THF 5:l

Figure imgf000035_0002

Form A of Formula I

The benzofuran carboxylic acid 18 was treated with oxalyl chloride (1.2 equivalents) and a catalytic amount of DMF, stirring for 5.5 hours until a clear solution was obtained. The solvent was removed under reduced pressure and the acid chloride of compound 18 was stored under argon until use, on the next day. The acid chloride, in methylene chloride was added slowly to a methylene chloride solution of the compound of Formula I and diisopropylethylamine (DIPEA) which was cooled to 0-50C. The reaction was not permitted to rise above 5°C, and after completion of addition, was stirred at 5°C for a further 0.5 hour. Upon aqueous workup and extraction with methylene chloride, the product, compound 19, was isolated in quantitative yield. The benzyl ester of compound 19 was removed by transfer hydrogenolysis using 10% palladium on carbon, using formic acid and triethylamine in a 5:1 mixture of methanol:THF, to produce the compound of Formula I in 95% yield. A final step of slurrying in methyl ethylketone (MEK) produced Form A of the compound of Formula I. The product was washed with water to remove residual MEK. Alternatively, the product of the hydrogenolysis step was slurried in acetonitrile yielded Form A of the compound of Formula I.

[00179] Taking the compound of Formula I directly as the crude reaction product after transfer hydrogenolysis, and reconcentrating down from a solution in methylene chloride, the amorphous form of the compound of Formula I was obtained in 97% purity.

Scheme 6

DMF

Figure imgf000036_0001

12 22

In this alternative approach, Boc-protection was used for the ring nitrogen in the intermediates 20, 21, and 22. Compound 5 was deprotected with HCl in dioxane to produce compound 20 in better than 97% yield. Boc- protection was introduced, using di-tert-butyl dicarbonate (1.1 equivalent), and compound 21 was obtained in better than 95% yield. Compound 10 was coupled with compound 21 to obtain compound 22, using HATU and triethylamine in DMF. The product, compound 22, was obtained in quantitative yield, and greater than 90% purity. Deprotection with HCl yields the compound of Formula I in 97.4% yield and the synthesis rejoins the process described in Scheme 5.

 

clip

Lifitegrast clinical efficacy for treatment of signs and symptoms of dry eye disease across three randomized controlled trials
Current Medical Research and Opinion (2016), 32, (10), 1759-1765. Publisher: (Taylor & Francis Ltd., )

PAPER

ACS Med. Chem. Lett., 2012, 3 (3), pp 203–206
DOI: 10.1021/ml2002482
Abstract Image

LFA-1/ICAM-1 interaction is essential in support of inflammatory and specific T-cell regulated immune responses by mediating cell adhesion, leukocyte extravasation, migration, antigen presentation, formation of immunological synapse, and augmentation of T-cell receptor signaling. The increase of ICAM-1 expression levels in conjunctival epithelial cells and acinar cells was observed in animal models and patients diagnosed with dry eye. Therefore, it has been hypothesized that small molecule LFA-1/ICAM-1 antagonists could be an effective topical treatment for dry eye. In this letter, we describe the discovery of a potent tetrahydroisoquinoline (THIQ)-derived LFA-1/ICAM-1 antagonist (SAR 1118) and its development as an ophthalmic solution for treating dry eye.

http://pubs.acs.org/doi/suppl/10.1021/ml2002482/suppl_file/ml2002482_si_001.pdf

str4STR1
Active Ingredient: LIFITEGRAST
Proprietary Name: XIIDRA
Dosage Form; Route of Administration: SOLUTION/DROPS; OPHTHALMIC
Strength: 5%
Reference Listed Drug: Yes
Reference Standard: Yes
TE Code:
Application Number: N208073
Product Number: 001
Approval Date: Jul 11, 2016
Applicant Holder Full Name: SHIRE DEVELOPMENT LLC
Marketing Status:  Prescription
Patent and Exclusivity Information

Patent and Exclusivity for: N208073

Product 001
LIFITEGRAST (XIIDRA) SOLUTION/DROPS 5%

Patent Data

Product No Patent No Patent Expiration Drug Substance Claim Drug Product Claim Patent Use Code
001 7314938 Mar 10, 2025 DS DP
001 7745460 Nov 5, 2024 DS DP U-1880
001 7790743 Nov 5, 2024 U-1880
001 7928122 Nov 5, 2024 DS DP
001 8084047 May 17, 2026 DS DP
001 8168655 May 9, 2029 U-1880
001 8367701 Apr 15, 2029 DP U-1880
001 8592450 May 17, 2026 U-1880
001 8927574 Nov 12, 2030 DP
001 9085553 Jul 25, 2033 DP
001 9216174 Nov 5, 2024 DP
001 9353088 Oct 21, 2030 DP
001 9447077 Apr 15, 2029 U-1900

Exclusivity Data

Product No Exclusivity Code Exclusivity Expiration
001 NCE Jul 11, 2021

“ALL FOR DRUGS, New drug approvals, Drug approvals international, ” CATERS TO EDUCATION GLOBALLY, No commercial exploits are done or advertisements added by me. This article is a compilation for educational purposes only.

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

///////

Cited Patent Filing date Publication date Applicant Title
US8084047 * Jul 23, 2009 Dec 27, 2011 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
Citing Patent Filing date Publication date Applicant Title
US8367701 Nov 4, 2011 Feb 5, 2013 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US8592450 Feb 16, 2012 Nov 26, 2013 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
US8758776 Jan 21, 2011 Jun 24, 2014 Sarcode Bioscience Inc. Compositions and methods for treatment
US8771715 Jan 21, 2011 Jul 8, 2014 Sarcode Bioscience Inc. Compositions and methods for treatment
WO2012121659A1 * Mar 8, 2012 Sep 13, 2012 Kat2Biz Ab C/O Interpares Konsult Ab Reduction of c-0 bonds by catalytic transfer hydrogenolysis
WO2014018748A1 * Jul 25, 2013 Jan 30, 2014 Sarcode Bioscience Inc. Lfa-1 inhibitor and polymorph thereof