New Drug Approvals

Home » Posts tagged 'rheumatoid arthritis'

Tag Archives: rheumatoid arthritis

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

Blog Stats

  • 4,234,988 hits

Flag and hits

Flag Counter

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

Join 2,800 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 2,800 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, CLEANCHEM LABS 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

Personal Links

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Azeliragon


Azeliragon.png

Azeliragon

C32H38ClN3O2, 532.1 g/mol

CAS 603148-36-3

TTP488

UNII-LPU25F15UQ

LPU25F15UQ

TTP-488; PF-04494700

3-[4-[2-butyl-1-[4-(4-chlorophenoxy)phenyl]imidazol-4-yl]phenoxy]-N,N-diethylpropan-1-amine

MOA:RAGE inhibitor

Indication:Alzheimer’s disease (AD)

Status:Phase III (Active), Dementia, Alzheimer’s type
Company:vTv Therapeutics (Originator)

Azeliragon

Azeliragon is in phase III clinical for the treatment of Alzheimer’s type dementia.

Azeliragon was originally by TransTech Pharma (now vTv Therapeutics), then licensed to Pfizer in 2006.

Pfizer discontinued the research in 2011, now vTv Therapeutics continues the further reaserch.

vTv Therapeutics  (previously TransTech Pharma) is developing azeliragon, an orally active antagonist of the receptor for advanced glycation end products (RAGE), for the treatment of Alzheimer’s disease (AD) in patients with diabetes.  In June 2019, this was still the case .

Azeliragon was originally developed at TransTech Pharma. In September 2006, Pfizer entered into a license agreement with the company for the development and commercialization of small- and large-molecule compounds under development at TransTech. Pursuant to the collaboration, Pfizer gained exclusive worldwide rights to develop and commercialize TransTech’s portfolio of RAGE modulators, including azeliragon.

Reference:

1. WO03075921A2.

2. US2008249316A1.

US 20080249316

VTV Therapeutics

Azeliragon (TTP488) is an orally bioavailable small molecule that inhibits the receptor for advanced glycation endproducts (RAGE). A Phase 2 clinical trial to evaluate azeliragon as a potential treatment of mild-AD in patients with type 2 diabetes is ongoing.  The randomized, double-blind, placebo-controlled multicenter trial is designed as sequential phase 2 and phase 3 studies operationally conducted under one protocol. For additional information on the study, refer to NCT03980730 at Clinicaltrials.gov.

RAGE is an immunoglobulin-like cell surface receptor that is overexpressed in brain tissues of patients with AD. The multiligand nature of RAGE is highlighted by its ability to bind diverse ligands such as advanced glycation end-products (AGEs), linked to diabetic complications and β-amyloid fibrils, a hallmark of AD. The association between type 2 diabetes and AD is well documented. A linear correlation between circulating hemoglobin A1c (HbA1c) levels and cognitive decline has been demonstrated in the English Longitudinal Study of Ageing.

PATENT

WO-2019190823

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019190823&tab=PCTDESCRIPTION&_cid=P12-K1K59I-21476-1

Novel crystalline forms of [3-(4-{2-butyl-1-[4-(4-chlorophenoxy)phenyl]-1H-imidazol-4-yl}phenoxy)-propyl]-diethylamine and its salt ( azeliragon ) (deignated as forms III and IV) as RAGE inhibitors useful for treating  psoriasis, rheumatoid arthritis and Alzheimer’s disease.

The Receptor for Advanced Glycation Endproducts (RAGE) is a member of the immunoglobulin super family of cell surface molecules. Activation of RAGE in different tissues and organs leads to a number of pathophysiological consequences. RAGE has been implicated in a variety of conditions including: acute and chronic inflammation (Hofmann et al., Cell 97:889-901 (1999)), the development of diabetic late complications such as increased vascular permeability (Wautier et al., J. Clin. Invest. 97:238-243 (1995)), nephropathy (Teillet et al., J. Am. Soc. Nephrol. 11 : 1488- 1497 (2000)), atherosclerosis (Vlassara et. al., The Finnish Medical Society DUODECIM, Ann. Med. 28:419-426 (1996)), and retinopathy (Hammes et al., Diabetologia 42:603-607 (1999)). RAGE has also been implicated in Alzheimer’s disease (Yan et al., Nature 382: 685-691 , (1996)), erectile dysfunction, and in tumor invasion and metastasis (Taguchi et al., Nature 405: 354-357, (2000)).

Binding of ligands such as advanced glycation endproducts (AGEs), S100/calgranulin/EN-RAGE, b-amyloid, CML (Ne-Carboxymethyl lysine), and amphoterin to RAGE has been shown to modify expression of a variety of genes. For example, in many cell types interaction between RAGE and its ligands generates oxidative stress, which thereby results in activation of the free radical sensitive transcription factor NF-kB, and the activation of NF-kB regulated genes, such as the cytokines IL- 1 b, TNF- a, and the like. In addition, several other regulatory pathways, such as those involving p21 ras.

MAP kinases, ERK1 and ERK2, have been shown to be activated by binding of AGEs and other ligands to RAGE. In fact, transcription of RAGE itself is regulated at least in part by NF-kB. Thus, an ascending, and often detrimental, spiral is fueled by a positive feedback loop initiated by ligand binding. Antagonizing binding of physiological ligands to RAGE, therefore, is our target, for down-regulation of the pathophysiological changes brought about by excessive concentrations of AGEs and other ligands for RAGE.

Pharmaceutically acceptable salts of a given compound may differ from each other with respect to one or more physical properties, such as solubility and dissociation, true density, melting point, crystal shape, compaction behavior, flow properties, and/or solid state stability. These differences affect practical parameters such as storage stability, compressibility and density (important in formulation and product manufacturing), and dissolution rates (an important factor in determining bio-availability). Although U.S. Patent No. 7,884,219 discloses Form I and Form II of COMPOUND I as a free base, there is a need for additional drug forms that are useful for inhibiting RAGE activity in vitro and in vivo, and have properties suitable for large-scale manufacturing and formulation. Provided herein

PATENT

WO03075921

PATENT

WO2019190822

PATENT

WO2008123914

Publications

Links to the following publications and presentations, which are located on outside websites, are provided for informational purposes only and do not constitute the opinions or views of vTv Therapeutics

Presentations and Posters

Links to the following publications and presentations, which are located on outside websites, are provided for informational purposes only and do not constitute the opinions or views of vTv Therapeutics

///////////Azeliragon, psoriasis, rheumatoid arthritis, Alzheimer’s disease, TTP-488,  PF-04494700, RAGE inhibitors, TransTech Pharma, PHASE 3, Dementia, Alzheimer’s type,

CCCCC1=NC(=CN1C2=CC=C(C=C2)OC3=CC=C(C=C3)Cl)C4=CC=C(C=C4)OCCCN(CC)CC

Ritlectinib, PF 06651600


Image result for PF-06651600

Image result for PF-06651600

Ritlectinib

PF-06651600

CAS 1792180-81-4

C₁₅H₁₉N₅O, 285.34, UNII-2OYE00PC25

1-((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1-yl)prop-2-en-1-one

Image result for PF-06651600

 1-[(2S,5R)-2-Methyl-5-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-1-piperidinyl]-2-propen-1-one malonate

PF-06651600 malonate
CAS: 2140301-97-7 (malonate)
Chemical Formula: C18H23N5O5

Molecular Weight: 389.412

PHASE 2  alopecia areata, rheumatoid arthritis, Crohn’s disease, and ulcerative colitis.

Pfizer is developing ritlecitinib, an irreversible, covalent and selective dual JAK3/TEC inhibitor, for treating AA, RA, vitiligo and inflammatory bowel diseases, including UC and CD. In July 2021, this drug was reported to be in phase 3 clinical development.

PF-06651600 is a potent and selective JAK3 inhibitor. PF-06651600 is a potent and low clearance compound with demonstrated in vivo efficacy. The favorable efficacy and safety profile of this JAK3-specific inhibitor PF-06651600 led to its evaluation in several human clinical studies. JAK3 was among the first of the JAKs targeted for therapeutic intervention due to the strong validation provided by human SCID patients displaying JAK3 deficiencies

Pfizer has established a leading kinase research capability with multiple unique kinase inhibitors in development as potential medicines. PF-06651600 is a highly selective and orally bioavailable Janus Kinase 3 (JAK3) inhibitor that represents a potential immunomodulatory therapy. With the favorable efficacy, safety profile, and ADME properties, this JAK3-specific covalent inhibitor has been under clinical investigation for the treatment of alopecia areata, rheumatoid arthritis, Crohn’s disease, and ulcerative colitis. Supported by positive results from a Phase 2 study, 1 was granted Breakthrough Therapy designation by the FDA on Sept. 5, 2018 for treatment of alopecia areata.

SYN

PAPER

J. Med. Chem. 201760 (5), 19711993DOI: 10.1021/acs.jmedchem.6b01694

https://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.6b01694

Paper

Process Development and Scale Up of a Selective JAK3 Covalent Inhibitor PF-06651600, 

Yong Tao*

Cite This:Org. Process Res. Dev.2019XXXXXXXXXX-XXX

Publication Date:July 19, 2019

https://doi.org/10.1021/acs.oprd.9b00198

A scalable process for PF-06651600 (1) has been developed through successful enabling of the first generation syntheis. The synthesis highlights include the following: (1) replacement of costly PtO2 with a less expensive 5% Rh/C catalyst for a pyridine hydrogenation, (2) identification of a diasteroemeric salt crystallization to isolate the enantiomerically pure cis-isomer directly from a racemic mixture of cis/trans isomers, (3) a high yielding amidation via Schotten–Baumann conditions, and (4) critical development of a reproducible crystallization procedure for a stable crystalline salt (1·TsOH), which is suitable for long-term storage and tablet formulation. All chromatographic purifications, including two chiral SFC chromatographic separations, were eliminated. Combined with other improvements in each step of the synthesis, the overall yield was increased from 5% to 14%. Several multikilogram batches of the API have been delivered to support clinical studies.

https://pubs.acs.org/doi/10.1021/acs.oprd.9b00198

1-((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1-yl)prop-2-en-1-one p-Toluenesulfonate (1·TsOH)

1·TsOH (4.41 kg, 9.64 mol) as a white powder in 89.6% yield (accounting for the amount of seed charged). Achiral HPLC purity: 99.6% with 0.22% of dimer 15. Chiral SFC purity: >99.7%. Mp 199 °C. Rotomers observed for NMR spectroscopies. 1H NMR (400 MHz, DMSO-d6): δ ppm 12.68 (brs, 1H), 9.22 (brs, 1H), 8.40 (s, 1H), 7.50 (d, J = 8.2 Hz, 2H), 7.45 (m, 1H), 7.12 (d, J = 8.2 Hz, 2H), 6.94 (d, J = 1.2 Hz, 1H), 6.84 (m, 1H), 6.13 (m, 1H), 5.70 (m, 1H), 4.81 (m, 0.5H), 4.54 (m, 0.5H), 4.41 (m, 0.5H), 4.12 (m, 0.5H), 3.99 (m, 1H), 3.15 (m, 0.5H), 2.82 (m, 0.5H), 2.29 (s, 3H), 1.91–1.72 (m, 4H), 1.24–1.17 (m, 3H). 13C NMR (100 MHz, DMSO-d6): δ ppm 165.52, 165.13, 150.50, 145.64, 143.06, 138.48, 129.51, 129.24, 128.67, 127.99, 127.73, 125.97, 125.02, 102.30, 49.53, 48.92, 47.27, 43.83, 42.96, 29.37, 28.41, 25.22, 21.28, 16.97, 15.51. HRMS (ESI) m/z: calculated for C15H20N5O [M + H]+286.1668; observed 286.1692.

PAPER

Telliez JB, et al. Discovery of a JAK3-Selective Inhibitor: Functional Differentiation of JAK3-Selective Inhibition over pan-JAK or JAK1-Selective Inhibition. ACS Chem Biol. 2016 Dec 16;11(12):3442-3451.

PATENT

WO2015083028

PATENT

WO 2015083028

https://patents.google.com/patent/WO2015083028A1

PATENT

WO 2020084435

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020084435&tab=PCTDESCRIPTION&_cid=P12-KIL68Y-23557-1

1 -((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one has the structural formula:

The synthesis of 1 -((2S,5R)-5-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one is described in WO2015/083028, commonly assigned to the assignee of the present invention and which is incorporated herein by reference in its entirety. 1 -((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one is useful as an inhibitor of protein kinases, such as the enzyme Janus Kinase (JAK) and as such is useful therapy as an immunosuppressive agent for organ transplants, xeno transplantation, lupus, multiple sclerosis, rheumatoid arthritis, psoriasis, Type I diabetes and complications from diabetes, cancer, asthma, atopic dermatitis, autoimmune thyroid disorders, ulcerative colitis, Crohn’s disease, alopecia, vitiligo, Alzheimer’s disease, leukemia and other indications where immunosuppression would be desirable. See ACS Chem. Biol. , 2016, 11 (12), pp 3442-3451 . The present invention relates to a novel p-toluenesulfonic acid salt and crystalline solid form of the said salt of 1 -((2S,5R)-5-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one that demonstrate improved properties for use in a pharmaceutical dosage form, particularly for oral dosage forms.

Preparations

Scheme 1. Synthesis of 1

Scheme 2. Alternate Synthesis of Intermediates 7 and 10

1. K2C03, MIBK/water 1. H2, H2O

2. EtOAc, aq. NaCI Pd(OH)2/C (wet)

3. MeOH, H20 2. NaOH, MeOH

7 + 8 –  9 – – 10 . H2O

89% 89%

Scheme 3. First Alternate Preparation of 1

Scheme 4. Second Alternate Preparation of 1

Preparation 1

ferf-Butyl (6-methylpyridin-3-yl)carbamate (3). To a 3000L reactor was charged 2 (72.00 kg, 665.8 mol) and THF (660 kg). A solution of NH4CI (1 .07 kg , 20 mol) in water (72 kg, 4000 mol) was added. The mixture was heated to 57 °C and Di-f-butyl dicarbonate (220.0 kg, 1003 mol) was added slowly with rinse of THF (45 kg) while maintaining the temperature between 55 – 60 °C. The mixture was stirred at 55 – 60 °C for 10 h. Upon reaction completion, the slurry was cooled to 20 °C and ethyl acetate (654 kg) and water (367 kg) were added. The organic phase was separated, washed by water (2 x 360 kg) and stirred with active carbon (22 kg) for 5 h. The mixture was filtered through a layer of diatomaceous earth (22 kg) with THF rinse and the filtrates were concentrated under vacuum at <40 °C to a residual volume of ~370 L. n-Heptane (500 kg) was added slowly over 1 h and the resulting slurry was cooled to 20 °C and stirred for 2 h. The solid was collected by centrifuge with an n-heptane wash (420 kg), then dried at 45 °C under vacuum for 20 h to give 3 (131 .15 kg, 629.7 mol) as a white powder in 94.5% yield. HPLC purity: 99.9%. 1H NMR (400 MHz, DMSO-c/6): d ppm 9.42 (brs, 1 H), 8.48 (d, J = 1 .9 Hz, 1 H), 7.75 (d, J = 8.6 Hz, 1 H), 7.13 (d, J = 8.6 Hz, 1 H), 2.38 (s, 3H), 1 .49 (s, 9H). 13C NMR (100 MHz, DMSO-d6y d ppm 153.34, 151 .56, 139.75, 134.13, 126.10, 123.09, 79.87, 28.56, 23.70. HRMS (ESI) m/z: calculated for C11H17N2O2 [M + H]+ 209.1290; observed 209.1285.

Preparation 2

ferf-Butyl (6-methylpiperidin-3-yl)carbamate (rac-4). To a 3000L reactor was charged 3 (137.0 kg, 667.8 mol), ethanol (988 kg) and acetic acid (139 kg). The reactor was purged with nitrogen three times and 5 wt% Rhodium on carbon (wet, 27.4 kg, 20 wt% loading relative to 3) was added. The reactor was purged with nitrogen three times and then with hydrogen three times. The hydrogen pressure was adjusted to 0.34 – 0.38 MPa and the reactor temperature was adjusted to 47 °C. The mixture was stirred at 45 – 60 °C under hydrogen pressure at 0.34 – 0.38 MPa for 10 h. Upon reaction completion, the reactor was cooled to 20 °C and flushed with nitrogen. The mixture was filtered through a layer of diatomaceous earth (20 kg) with an ethanol rinse (1320 kg) and the filtrates were concentrated under vacuum at <50 °C to a residual volume of ~350 L. n-Heptane (571 kg) was added and the mixture was concentrated under vacuum at <50 °C to a residual volume of~350 L. This operation was repeated twice until the residual acetic acid <8.0%. Ethanol (672 kg) was added and the mixture was concentrated under vacuum at <50 °C to a residual volume of ~350 L. This operation was repeated twice until the residual n-heptane was <0.2% and water was <0.2%. Ethanol (889 kg) was added and the solution (1254 kg) was transferred to drums for use in the subsequent classical resolution step. Achiral HPLC assay indicated that the solution contained 10.8 wt% of the total reduced product (rac-4) in 96% mass recovery and chiral SFC showed that the solution contained 36.3% of the desired stereoisomer cis-4.

Preparation 3

ferf-Butyl ((3R,6S)-6-methylpiperidin-3-yl)carbamate (R)-2-(3,5-dinitrobenzamido)-2-phenylacetic acid salt (15). To a 2000L reactor (R1 ) was charged rac-4 as a 10.8 wt% solution in ethanol (620.5 kg, ~312.7 mol. of all 4 isomers). The solution was concentrated under vacuum at <45 °C to a residual volume of ~210 L and then cooled to 20 °C. To a 3000 L reactor (R2) was charged (R)-2-(3,5-dinitrobenzamido)-2-phenylacetic acid 14 (47.0 kg, 136.1 mol) and ethanol (1 125 kg). With high speed agitation, reactor R2 was heated to 70 °C, stirred at 68 – 70 °C for ~2 h to dissolve all solid 14, and then seeded with crystalline 15 (1 1 g). The solution containing 4 in reactor R1 was slowly transferred to reactor R2 over 30 min with ethanol rinse (160 kg). Reactor R2 was stirred at ~74 °C for 3 h and then cooled to 22 °C with a linear cooling rate over a period of 5 h and stirred for 16 h. The solid was collected by centrifuge with ethanol wash (2 x 200 kg). The wet cake (with 97.1 % e.e.) was charged back to reactor R2. The slurry was heated to 74 °C and the mixture was stirred for 17 h. The mixture was then cooled to 22 °C with a linear cooling rate over a period of 5 h and stirred for 4 h. The solid was collected by centrifuge with ethanol wash (2 x 200 kg) and dried at 35 °C under vacuum for 25 h to give 15 (56.05 kg, 100.2 mol) as a white powder in 30.7% yield over 2 steps. Chiral HPLC purity: 99.1 %. 1H NMR (400 MHz, DMSO-d6): d ppm 9.46 (d, J = 7.0 Hz, 1 H), 9.07 (d, J = 2.2 Hz, 2H), 8.96 (t, J = 2.2 Hz, 1 H), 7.49 (d, J = 7.3 Hz, 2H), 7.30 (t, J = 7.3 Hz, 2H), 7.23 (t, J = 7.3, 1 H), 7.1 1 (m, 1 H), 5.31 (d, J = 7.0 Hz, 1 H), 3.66 (m, 1 H), 2.98 (m, 3H), 1 .63 (m, 2H), 1 .45 (m, 2H), 1 .40 (s, 9H), 1 .1 1 (d, J = 6.7 Hz, 3H). 13C NMR (100 MHz, DMSO-d6): d ppm 172.71 , 161 .71 , 155.42, 148.51 , 141 .27, 137.70, 128.29, 128.25, 128.02, 127.05, 121 .12, 78.49, 59.74, 50.66, 46.29, 43.34, 28.66, 26.88, 26.1 1 , 18.60.

Preparation 4

Benzyl (2S,5R)-5-amino-2-methylpiperidine-1 -carboxylate hydrochloride (7»HCI) -telescoped process. To a 2000L reactor was charged 15 (70.0 kg, 125 mol) and MTBE (500 kg). The mixture was cooled to 12 °C and 6.9 wt% aqueous NaOH solution (378 kg, 652 mol) was added slowly while maintaining the temperature between 10 – 25 °C. The mixture was stirred at 18 °C for 1 h . The organic phase was separated and washed with 3.8 wt% aqueous NaOH solution (2 x 221 kg) and then 25 wt% aqueous NaCI solution (2 x 220 kg). The organic layer (containing the free base cis-4) was concentrated under vacuum at <40 °C to a residual volume of ~300 L and then cooled to 20 °C. NaHCOs (53 kg, 632 mol) and water (200 kg) were added and the mixture was cooled to 7 °C. Benzyl chloroformate (32.30 kg, 189.3 mol) was added slowly while maintaining the temperature between 5 – 20 °C. The mixture was stirred at 17 °C for 20 h. Upon reaction completion, the mixture was cooled to 12 °C, 25 wt% aqueous ammonium hydroxide solution (79 kg, 1 160 mol) was added slowly while maintaining the temperature between 10 – 20 °C, and the mixture was stirred at 15 °C for 1 h. The organic phase was separated and washed with 25 wt% aqueous NaCI solution (3 x 90 kg). The organic layer (containing 5) was concentrated under vacuum at <45 °C to a residual volume of ~150 L. Isopropyl acetate (310 kg) was added and the mixture was concentrated under vacuum at <45 °C to a residual volume of ~150 L. This operation was repeated twice to meetthe criteria of water <0.1 % (by KF). Isopropyl acetate (130 kg) was then added and the mixture was cooled to -3 °C. 4-5N HCI in methanol (181 kg, ~730 mol) was added slowly while maintaining the temperature between -5 to 5 °C, and the mixture was stirred at 3 °C for 12 h. Upon reaction completion, the mixture was cooled to -3 °C and MTBE (940 kg) was added slowly while maintaining the temperature between -5 to 5 °C. The resulting slurry was stirred at 3 °C for 3 h. The solid was collected by centrifuge with MTBE washes (4 x 70 kg), and then dried at 45 °C under vacuum for 20 h to give 7»HCI (28.60 kg, 100.4 mol) as a white powder in 80.3% yield. Achiral HPLC purity: 100%. Chiral SFC purity: 99.8% e.e. 1H NMR (400 MHz, DMSO-d6): d ppm 8.36 (brs, 3H), 7.37 (m, 5H), 5.09 (s, 2H), 4.31 (m, 1 H), 4.16 (d, J = 8.2 Hz, 1 H), 3.00 (m, 2H), 1 .82 (m, 2H), 1 .59 (m, 2H), 1 .1 1 (d, J = 7.0 Hz, 3H). 13C NMR (100 MHz, DMSO-d6): d ppm 154.71 , 137.24, 128.92, 128.34, 128.00, 66.89, 47.20, 45.66, 40.68, 28.16, 23.02, 15.67. HRMS (ESI) m/z. calculated for C H N O [M + H]+ 249.1603; observed 249.1598.

Preparation 5

Benzyl (2S, 5R)-5-((2-chloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2 -methyl-piperidine-1 -carboxylate (9). To a 2000L reactor was charged 7»HCI (88.6 kg, 31 1 .12 mol), 8 (56.0 kg, 298 mol), K2C03 (133.0 kg, 962.3 mol), water (570 kg) and MIBK (101 kg). The mixture was heated to 90 °C and stirred at this temperature for 22 h. Upon reaction completion, the mixture was cooled to 56 °C and ethyl acetate (531 kg) was added. After cooling the mixture to 22 °C, the organic phase was separated, washed with water (570 kg) and concentrated under vacuum at <40 °C to a residual volume of ~220 L. Methanol (360 kg) was added slowly over a period of 1 h and the mixture concentrated under vacuum at <50 °C to a residual volume of ~220 L. This operation was repeated three times until residual MIBK reached <5 wt%. Methanol (270 kg) was added, followed by seeding with 9 (120 g). The mixture was stirred at 22 °C for >4 h and water (286 kg) was added slowly over 4 h. The slurry was stirred for 10 h and the solid was then collected by centrifuge. The wet cake (165.6 kg) was charged back to a clean reactor and water (896 kg) was added. The slurry was heated to 55 °C and stirred at this temperature for 7 h; and then cooled to 22 °C and stirred at this temperature for 2 h. The solid was collected by centrifuge with water wash (3 x 170 kg) and dried at 55 °C under vacuum for 20 h to give 9 (106.62 kg, 266.6 mol) as a white powder in 89.5% yield. Achiral HPLC purity: 99.7%. 1H NMR (400 MHz, DMSO-d6): d ppm 1 1 .71 (brs, 1 H), 7.72 (d, J = 7.9 Hz, 1 H), 7.38 (m, 5H), 7.10 (s, 1 H), 6.57 (d, J = 2.7 Hz, 1 H), 5.1 1 (m, 2H), 4.39 (m, 1 H), 4.17 (m, 1 H), 4.01 (m, 1 H), 3.36 (s, 2H), 2.77 (m, 1 H), 1 .73-1 .81 (m, 4H), 1 .16 (d, J = 6.6 Hz, 3H). 13C NMR (100 MHz, DMSO-d6): d ppm 156.65, 154.74, 153.04, 151 .31 , 137.43, 128.89, 128.27, 127.96, 122.13, 101 .65, 99.51 , 66.75, 49.10, 47.32, 45.64, 42.98, 29.05, 25.08. HRMS (ESI) m/z. calculated for C20H22CIN5O2 [M + H]+ 400.1540; observed 400.1535.

Preparation 6

N-((3R,6S)-6-methylpiperidin-3-yl)-7H-pyrrolo[2,3-dlpyrimidin-4-amine monohydrate (10·H2O) To a 1600L reactor was charged water (570 kg). The reactor was purged with nitrogen three times. 10% Pd(OH)2/C (wet, 3.2 kg) and 9 (53.34 kg, 133.2 mol) were added with water rinses (2 x 55 kg). The reactor was purged with nitrogen three times and then with hydrogen three times. The hydrogen pressure was adjusted to 0.34 – 0.38 MPa and the reactor temperature was adjusted to 77 °C. The mixture was stirred at 75 – 80 °C under a hydrogen pressure of 0.34 -0.38 MPa for 10 h. Upon reaction completion, the reactor was cooled to 20 °C and purged with nitrogen. The mixture was filtered through a layer of diatomaceous earth (8 kg) with a water rinse (460 kg), and the filtrates were transferred to a 3000L reactor. Methanol (260 kg) was added, followed by slow addition of 50 wt% aqueous sodium hydroxide (12.0 kg , 150 mol) while maintaining the temperature between 15 – 25 °C. The slurry was heated to 55 °C and stirred for 2 h; then cooled to 22 °C and stirred for 10 h. The solid was collected by centrifuge with a 10:1 water/methanol wash (3 x 1 10 kg) and then dried at 55 °C under vacuum for 20 h to give 10·H2O (30.90 kg, 266.6 mol) as a white powder in 89.1 % yield. Achiral HPLC purity: 99.7%. Chiral SFC

purity: 99.8% e.e. 1H NMR (400 MHz, DMSO-d6): d ppm 1 1 .48 (brs, 1 H), 8.08 (s, 1 H), 7.07 (s, 1 H), 6.85 (d, J = 7.3 Hz, 1 H), 6.64 (s, 1 H), 4.16 (m, 1 H), 3.35 (brs, 2H), 2.96 (d, J = 12.7 Hz, 1 H), 2.82 (d, J = 12.7 Hz, 1 H), 2.67 (m, 1 H), 2.04 (brs, 1 H), 1 .92 (m, 1 H), 1 .63 (m, 1 H), 1 .44 (m, 1 H), 1 .33 (m, 1 H), 1 .03 (d, J = 6.2 Hz, 3H). 13C NMR (100 MHz, DMSO-d6): d ppm 155.95, 151 .87, 150.74, 121 .20, 102.97, 99.20, 51 .27, 49.94, 44.78, 29.97, 28.69, 22.35. HRMS (ESI) m/z\ calculated for C12H17N5 [M + H]+ 232.1562; observed 232.1558.

Preparation 7

1 -((2S,5R)-5-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one (1 ). To a 100L reactor was charged water (18.0 L), 10·H2q (3.60 kg, 14.4 mol) and THF (36.0 L). The mixture was heated to 53 °C and stirred for 15 min to dissolve all the solids. The solution was then cooled to 18 °C and K3PO4 (6.38 kg, 30.1 mol) was added. The mixture was stirred at 18 °C for 10 min to dissolve all the solids, and then cooled to 10 °C. 3-Chloropropionyl chloride (2.20 kg, 17.3 mol) was added while maintaining the temperature <20 °C. The mixture was then stirred at 20 °C for 2 h. Upon reaction completion, 2 N aqueous NaOH solution (23.50 kg, 43.76 mol) was added while maintaining the temperature <25 °C. The mixture was stirred at 22 °C for >12 h until the elimination reaction was complete (11 <0.2%). KH2PO4 (10.32 kg, 75.8 mol) was added and the mixture was stirred at 20 °C for 10 min. The organic phase was separated and then washed with 23.5 wt% aqueous NaCI solution (2 x 8.5 kg). The isolated organic phase was concentrated under vacuum at <30 °C to a residual volume of ~10 L, whereupon MEK (39.6 L) was added. This operation was repeated once or twice until residual THF was <1 % and water was <2%. MgS04 (0.96 kg), Silica gel (4.90 kg) and Darco™ G-60 (0.48 kg) were added to the MEK solution, and the mixture was stirred at 20 °C for 1 h, then filtered through a layer of Diatomaceous Earth with a MEK rinse (76 L). The combined filtrates were concentrated under vacuum at <30 °C to a residual volume of ~8 L. The concentration of the residual solution was measured by qNMR, and the solution was transferred to a container with a rinse using the calculated amount of MEK to adjust the final concentration to 30 wt%. Thus, a 30 wt% solution of 1 in MEK (1 1 .09 kg, 1 1 .66 mol of 1) with 98.7% purity was obtained in 81 % yield, which was stored in a cold room (2 – 8 °C) for the next step.

Preparation 8

1 -((2S,5R)-5-((7H-pyrrolo[2,3-cdpyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one p-toluenesulfonate (1»TsOH). To a 20L reactor was charged a 30 wt% solution of 1 in MEK (9.80 kg, 10.30 mol of 1) and silica gel (0.74 kg). The mixture was stirred at 22 °C for 15 min and filtered through a 0.45 micron Teflon cartridge filter with a MEK rinse (7.89 kg, 9.8 L), collecting in a 100L reactor. Water (1 .27 L) was added, followed by a solution of p-toluenesulfonic acid monohydrate (2.18 kg, 1 1 .3 mol) in MEK (4.75 kg, 5.9 L) with a MEK rinse (3.14 kg, 3.9 L), followed by the addition of 1 »TsOH seed (188 g, 0.41 mol). The mixture was stirred at 22 °C for

4 h to form a slurry and MEK (31 .56 kg, 39.2 L) was added slowly over a period of 3 h. The slurry was stirred at 22 °C for an additional 2 h and then filtered. The cake was washed with MEK (4.02 kg, 5 L) and then dried at 50 °C under vacuum for 10 h to give 1 »TsOH (4.41 kg, 9.64 mol) as a white powder in 89.6% yield (accounting for the amount of seed charged). Achiral HPLC purity: 99.6% with 0.22% of dimer 15. Chiral SFC purity: >99.7%. m.p. 199 °C. Rotomers observed for NMR spectroscopies. Ή NMR (400 MHz, DMSO-d6): d ppm 12.68 (brs, 1 H), 9.22 (brs, 1 H), 8.40 (s, 1 H), 7.50 (d, J = 8.2 Hz, 2H), 7.45 (m, 1 H), 7.12 (d, J = 8.2 Hz, 2H), 6.94 (d, J = 1 .2 Hz, 1 H), 6.84 (m, 1 H), 6.13 (m, 1 H), 5.70 (m, 1 H), 4.81 (m, 0.5H), 4.54 (m, 0.5H), 4.41 (m, 0.5H), 4.12 (m, 0.5H), 3.99 (m, 1 H), 3.15 (m, 0.5H), 2.82 (m, 0.5H), 2.29 (s, 3H), 1 .91 -1 .72 (m, 4H), 1 .24-1 .17 (m, 3H). 13C NMR (100 MHz, DMSO-c/6): d ppm 165.52, 165.13, 150.50, 145.64, 143.06, 138.48, 129.51 , 129.24, 128.67, 127.99, 127.73, 125.97, 125.02, 102.30, 49.53, 48.92, 47.27, 43.83, 42.96, 29.37, 28.41 , 25.22, 21 .28, 16.97, 15.51 . HRMS (ESI) m/z: calculated for Ci5H2oN50 [M + H]+ 286.1668; observed 286.1692.

Comparative Example

Preparation of 1 -((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methyl-piperidin-1 -yl)prop-2-en-1 -one Malonic Acid Salt (Form 1 )

A 250 ml_ round bottom flask was charged with 1 -((2S,5R)-5-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one (4.10 g, 14.4 mmol), MEK (Methyl Ethyl Ketone (15.0 ml_/g, 687 mmol, 49.5 g, 61 .5 ml_)). To the solution, malonic acid (0.950 equiv. 13.7 mmol, 1 .42 g) was added in one portion. The mixture was heated to 50 °C and stirred at 50 °C for 15min. The heating was turned off and the slurry was stirred for 16 hours. The resulting white slurry was filtered. The filter cake was washed with MEK (2 X 5 ml_) and dried in a vacuum oven (40 °C) for 2 hours give 1 -((2S,5R)-5-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1 -yl)prop-2-en-1 -one malonic acid salt (Form 1) (4.48 g, 1 1 .5 mmol, 4.48 g, 80.1 % Yield) as white powder.

PATENT

WO-2021136482

Improved process for preparing ritlecitinib (PF06651600), useful for treating alopecia areata (AA), rheumatoid arthritis (RA), Crohn’s disease (CD) and ulcerative colitis (UC). Also claims novel intermediates of ritlecitinib and their preparation method.

PATENT

CN111732591

crystalline polymorphic form of ritlecitinib L-tartrate. Jiangsu Alicorn focuses on developing generic, innovative and first-line drugs, whose website lists an undisclosed JAK inhibitor under its product’s list.

REFERENCES

1: D’Amico F, Fiorino G, Furfaro F, Allocca M, Danese S. Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials. Expert Opin Investig Drugs. 2018 Jul;27(7):595-599. doi: 10.1080/13543784.2018.1492547. Epub 2018 Jul 6. Review. PubMed PMID: 29938545.

2: Robinette ML, Cella M, Telliez JB, Ulland TK, Barrow AD, Capuder K, Gilfillan S, Lin LL, Notarangelo LD, Colonna M. Jak3 deficiency blocks innate lymphoid cell development. Mucosal Immunol. 2018 Jan;11(1):50-60. doi: 10.1038/mi.2017.38. Epub 2017 May 17. PubMed PMID: 28513593; PubMed Central PMCID: PMC5693788.

3: Thorarensen A, Dowty ME, Banker ME, Juba B, Jussif J, Lin T, Vincent F, Czerwinski RM, Casimiro-Garcia A, Unwalla R, Trujillo JI, Liang S, Balbo P, Che Y, Gilbert AM, Brown MF, Hayward M, Montgomery J, Leung L, Yang X, Soucy S, Hegen M, Coe J, Langille J, Vajdos F, Chrencik J, Telliez JB. Design of a Janus Kinase 3 (JAK3) Specific Inhibitor 1-((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1-yl)prop -2-en-1-one (PF-06651600) Allowing for the Interrogation of JAK3 Signaling in Humans. J Med Chem. 2017 Mar 9;60(5):1971-1993. doi: 10.1021/acs.jmedchem.6b01694. Epub 2017 Feb 16. PubMed PMID: 28139931.

4: Telliez JB, Dowty ME, Wang L, Jussif J, Lin T, Li L, Moy E, Balbo P, Li W, Zhao Y, Crouse K, Dickinson C, Symanowicz P, Hegen M, Banker ME, Vincent F, Unwalla R, Liang S, Gilbert AM, Brown MF, Hayward M, Montgomery J, Yang X, Bauman J, Trujillo JI, Casimiro-Garcia A, Vajdos FF, Leung L, Geoghegan KF, Quazi A, Xuan D, Jones L, Hett E, Wright K, Clark JD, Thorarensen A. Discovery of a JAK3-Selective Inhibitor: Functional Differentiation of JAK3-Selective Inhibition over pan-JAK or JAK1-Selective Inhibition. ACS Chem Biol. 2016 Dec 16;11(12):3442-3451. Epub 2016 Nov 10. PubMed PMID: 27791347.

5: Walker G, Croasdell G. The European League Against Rheumatism (EULAR) – 17th Annual European Congress of Rheumatology (June 8-11, 2016 – London, UK). Drugs Today (Barc). 2016 Jun;52(6):355-60. doi: 10.1358/dot.2016.52.6.2516435. PubMed PMID: 27458612.

////////////PF-06651600, PF 06651600, PF06651600, Breakthrough Therapy designation, PHASE 2,   alopecia areata, rheumatoid arthritis, Crohn’s disease,  ulcerative colitis, Ritlectinib

C=CC(N1[C@@H](C)CC[C@@H](NC2=C3C(NC=C3)=NC=N2)C1)=O

Piclidenoson, иклиденозон , بيكليدينوسون , 匹利诺生 ,


img

Thumb

ChemSpider 2D Image | Piclidenoson | C18H19IN6O4

DB05511.png

CF 101, Piclidenoson

ALB-7208

CAS 152918-18-8
Chemical Formula: C18H19IN6O4
Molecular Weight: 510.28

(2S,3S,4R,5R)-3,4-Dihydroxy-5-{6-[(3-iodobenzyl)amino]-9H-purin-9-yl}-N-methyltetrahydro-2-furancarboxamide

N6-(3-Iodobenzyl)adenosine-5′-N-methyluronamide

β-D-Ribofuranuronamide, 1-deoxy-1-[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-

1-Deoxy-1-[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-β-D-ribofuranuronamide

10136
1-Deoxy-1-[6-[((3-Iodophenyl)methyl)amino]-9H-purin-9-yl]-N-methyl-β-D-ribofuranuronamide
30679UMI0N
UNII-30679UMI0N
Пиклиденозон [Russian] [INN]
بيكليدينوسون [Arabic] [INN]
匹利诺生 [Chinese] [INN]

CF 101 (known generically as IB-MECA) is an anti-inflammatory drug for rheumatoid arthritis patients. Its novel mechanism of action relies on antagonism of adenoside A3 receptors. CF101 is supplied as an oral drug and has an excellent safety profile. It is also being considered for the treatment of other autoimmune-inflammatory disorders, such as Crohn’s disease, psorasis and dry eye syndrome.

Image result for CF 101, Piclidenoson

  • Originator Can-Fite BioPharma
  • Class Amides; Anti-inflammatories; Antineoplastics; Antipsoriatics; Antirheumatics; Eye disorder therapies; Iodobenzenes; Neuroprotectants; Purine nucleosides; Ribonucleosides; Small molecules
  • Mechanism of Action Adenosine A3 receptor agonists; Immunosuppressants; Interleukin 23 inhibitors; Interleukin-17 inhibitors
  • Phase III Plaque psoriasis; Rheumatoid arthritis
  • Phase II Glaucoma; Ocular hypertension
  • Phase I Uveitis
  • Preclinical Osteoarthritis
  • Discontinued Colorectal cancer; Dry eyes; Solid tumours
  • 05 Feb 2019 Can-Fite BioPharma receives patent allowance for A3 adenosine receptor (A3AR) agonists in USA
  • 05 Feb 2019 Can-Fite BioPharma receives patent allowance for A3 adenosine receptor (A3AR) agonists in North America, South America, Europe and Asia
  • 21 Aug 2018 Phase-III clinical trials in Plaque psoriasis (Monotherapy) in Israel (PO)

Piclidenoson, also known as CF101, is a specific agonist to the A3 adenosine receptor, which inhibits the development of colon carcinoma growth in cell cultures and xenograft murine models. CF101 has been shown to downregulate PKB/Akt and NF-κB protein expression level. CF101 potentiates the cytotoxic effect of 5-FU, thus preventing drug resistance. The myeloprotective effect of CF101 suggests its development as an add-on treatment to 5-FU.

Piclidenoson is known to be a TNF-α synthesis inhibitor and a neuroprotectant. use as an A3 adenosine receptor agonist, useful for treating rheumatoid arthritis (RA), psoriasis, osteoarthritis and glaucoma.

Can-Fite BioPharma , under license from the National Institutes of Health (NIH), is developing a tablet formulation of CF-101, an adenosine A3 receptor-targeting, TNF alpha-suppressing low molecular weight molecule for the potential treatment of psoriasis, RA and liver cancer. The company is also investigating a capsule formulation of apoptosis-inducing namodenoson, the lead from a program of adenosine A3 receptor agonist, for treating liver diseases, including hepatocellular carcinoma (HCC). In January 2019, preclinical data for the treatment of obesity were reported. Also, see WO2019105217 , WO2019105359 and WO2019105082 , published alongside.

PATENT

WO-2019105388

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019105388&tab=FULLTEXT&maxRec=1000

Novel crystalline forms of CF-101 (also known as piclidenoson; designated as Forms CS1, CS2 and CS3), processes for their preparation, compositions comprising them and their use as an A3 adenosine receptor agonist for treating rheumatoid arthritis, psoriasis, osteoarthritis and glaucoma are claimed

CF-101 was developed by Kan-Fete Biomedical Co., Ltd. By the end of 2018, CF-101 is in clinical phase III for the treatment of autoimmune diseases such as rheumatoid arthritis, osteoarthritis and psoriasis, as well as glaucoma. CF-101 is an A3 adenosine receptor (A3AR) agonist, and adenosine plays an important role in limiting inflammation through its receptor. Adenosine can produce anti-inflammatory effects by inhibiting TNF-a, interleukin-1, and interleukin-6. Studies have shown that A3AR agonists are in different experimental autoimmune models, such as rheumatoid arthritis, Crohn’s disease, and silver swarf. In the disease, it acts as an anti-inflammatory agent by improving the inflammatory process.
The chemical name of CF-101 is: 1-deoxy-I-(6-{[(3-iodophenyl)methyl]amino}-9H-fluoren-9-yl)-N-methyl-bD-ribofuranose Carbonamide (hereinafter referred to as “Compound I”) has the following structural formula:
A crystal form is a solid in which a compound molecule is orderedly arranged in a microstructure to form a crystal lattice, and a drug polymorphism phenomenon means that two or more different crystal forms of a drug exist.
Due to different physical and chemical properties, different crystal forms of drugs may have different dissolution and absorption in the body, which may affect the clinical efficacy and safety of the drug to a certain extent; especially for poorly soluble solid drugs, the crystal form will have greater influence. Therefore, the drug crystal form is inevitably an important part of drug research and an important part of drug quality control. Most importantly, the study of crystal forms is beneficial to find a crystal form that is clinically therapeutically meaningful and has stable and physicochemical properties.
There are no reports of CF-101 related crystal forms so far. Amorphous is generally not suitable as a medicinal form, and the molecules in the amorphous material are disorderly arranged, so they are in a thermodynamically unstable state. Amorphous solids are in a high-energy state, and generally have poor stability. During the production and storage process, amorphous drugs are prone to crystal transformation, which leads to the loss of consistency in drug bioavailability, dissolution rate, etc., resulting in changes in the clinical efficacy of the drug. In addition, the amorphous preparation is usually a rapid kinetic solid precipitation process, which easily leads to excessive residual solvents, and its particle properties are difficult to control by the process, making it a challenge in the practical application of the drug.
Therefore, there is a need to develop a crystalline form of CF-101 that provides a usable solid form for drug development. The inventors of the present application have unexpectedly discovered the crystalline forms CS1, CS2 and CS3 of Compound I, which have melting point, solubility, wettability, purification, stability, adhesion, compressibility, fluidity, dissolution in vitro and in vivo, and biological effectiveness. There is an advantage in at least one of the properties and formulation processing properties. Crystalline CS1 has advantages in physical and chemical properties, especially physical and chemical stability, low wettability, good solubility and good mechanical stability. It provides a new and better choice for the development of drugs containing CF-101, which is very important. The meaning.
Figure 7 is a 1 H NMR spectrum of the crystalline form CS3 obtained according to Example 7 of the present invention
The nuclear magnetic data of the crystalline form CS3 obtained in Example 7 was: { 1 H NMR (400 MHz, DMSO) δ 8.82 – 8.93 (m, 1H), 8.53 – 8.67 (m, 1H), 8.45 (s, 1H), 8.31 ( s, 1H), 7.73 (s, 1H), 7.59 (d, J = 7.7 Hz, 1H), 7.36 (d, J = 7.7 Hz, 1H), 7.11 (t, J = 7.8 Hz, 1H), 5.98 ( d, J = 7.4 Hz, 1H), 5.74 (s, 1H), 5.56 (s, 1H), 4.64 (d, J = 29.3 Hz, 3H), 4.32 (s, 1H), 4.15 (s, 1H), 2.71 (d, J = 4.6 Hz, 3H), 1.91 (s, 3H).}. Form CS3 has a single peak at 1.91, corresponding to the hydrogen chemical shift of the acetic acid molecule. According to the nuclear magnetic data, the molar ratio of acetic acid molecule to CF-101 is 1:1, and its 1 H NMR is shown in FIG.7

PAPER

Journal of medicinal chemistry (1994), 37(5), 636-46

https://pubs.acs.org/doi/pdf/10.1021/jm00031a014

PAPER

Journal of medicinal chemistry (1998), 41(10), 1708-15

https://pubs.acs.org/doi/abs/10.1021/jm9707737

PAPER

Bioorganic & Medicinal Chemistry (2006), 14(5), 1618-1629

PATENT

WO 2015009008

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015009008

Example 1
Preparation Example 1: Synthesis of Compound (5) (S) -2 – ((R) -1- (2-Chloro-6- (3-iodobenzylamino) -9H- purin- Hydroxyethoxy) -3-hydroxy-N-methylpropanamide)
Scheme 1
Step 1: A solution of (2R, 3S, 4S, 5R) -2- (benzoyloxymethyl) -5- (2,6- dichloro-9H- purin-9- yl) tetrahydrofuran- Preparation of benzoate (7)
Starting material A mixture of (2R, 3R, 4S, 5R) -2-acetoxy-5- (benzoyloxymethyl) tetrahydrofuran-3,4-diyldibenzoate (7.5 g, 14.9 mmol) (3.09 g, 16.4 mmol) was dissolved in acetonitrile (50 mL), and a solution of N, O-bis (trimethylsilyl) acetamid (8.9 mL, 36.4 mmol) was slowly added dropwise for 10-15 minutes Then, the mixture is stirred at 60 DEG C for 30 minutes. After cooling the reaction solution to -30 ° C, TiCl 4 (60 mL, 1 M methylene chloride solution, 59.5 mmol) is added dropwise, and the mixture is stirred at 60-65 ° C for 20 minutes. After confirming the completion of the reaction, methylene chloride (500 mL) and saturated sodium hydrogencarbonate solution (500 mL) are added. The reaction solution was stirred at 0 ° C for 30 minutes, and the organic layer was extracted and dried over anhydrous magnesium sulfate. After concentration under reduced pressure, the obtained residue was separated by column chromatography to obtain the intermediate compound (2R, 3S, 4S, 5R) -2- (benzoyloxymethyl) -5- (2,6- dichloro- Yl) tetrahydrofuran-3,4-diyl dibenzoate (9.3 g, 98.8%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm 4.71-4.74 (dd, J = 12.22, 3.91 Hz, 1H), 4.85-4.93 (m, 2H), 6.12-6.14 (t, J = 4.89 Hz, 1H) (M, 1H), 6.16-6.19 (t, J = 5.38 Hz, 1H), 6.47-6.48 (d, J = 5.38 Hz, 1H), 7.35-7.38 4H), 7.54-7.61 (m, 3H), 7.92-7.93 (d, J = 7.33 Hz, 2H), 8.02-8.06 (m, 4H), 8.28 (s, 1H); 13C NMR (125 MHz; CDCl 3 ) δ 63.50, 71.59, 74.33, 81.56, 87.05, 128.14, 128.59 (3), 128.63 (2), 128.73 (2), 129.10, 129.63 (2), 129.88 (2), 129.92 (2), 131.38, 133.63, 133.87, 133.98, 143.81, 152.36, 152.64, 153.51, 165.13, 165.29, 166.03; mp = 76-80 [deg.] C.
Step 2: (2R, 3S, 4S, 5R) -2- (Benzoyloxymethyl) -5- (2-chloro-6- (3-iodobenzylamino) -9H- purin-9-yl) tetrahydrofuran -3,4-diyl dibenzoate (8)
The intermediate compound (204 mg, 0.32 mmol) and 3-iodobenzylamine hydrochloride (113 mg, 0.41 mmol) prepared in the above step 1 were dissolved in anhydrous ethanol (5 mL) under a nitrogen atmosphere, triethylamine (0.13 mL, 0.96 mmol) is stirred at room temperature for 24 hours. After confirming the completion of the reaction, the reaction solution was concentrated under reduced pressure, and the obtained residue was separated by column chromatography to obtain the intermediate compound (2R, 3S, 4S, 5R) -2- (benzoyloxymethyl) (3-iodobenzylamino) -9H-purin-9-yl) tetrahydrofuran-3,4-diyldibenzoate (230 mg, 86.14%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm 4.71-4.90 (m, 5H), 6.13-6.17 (m, 2H), 6.33 (.. Br s, 1H), 6.43-6.44 (d, J = 4.89 Hz (M, 4H), 7.31-7.33 (m, 6H), 7.33-7.46 (m, 6H) 7.70-7.71 (t, J = 1.46 Hz, 1H), 7.88 (s, 1H), 7.94-7.96 (m, 2H), 7.99-8.01 (m, 2H), 8.07-8.09 (m, 2H); 13 C NMR (125 MHz; CDCl 3) δ 43.91, 63.79, 71.56, 74.43, 80.97, 86.40, 94.49, 119.07, 127.14, 128.40, 128.52 (3), 128.62 (2), 128.71, 129.32, 129.68 (3), 129.86 (2), 129.93 (2), 130.39 (2), 133.41, 133.69, 133.78, 136.72, 136.80, 138.39, 140.20, 150.05, 155.00, 165.17, 165.31, 166.11; mp = 80-84 [deg.] C.
Step 3: ((3aR, 4R, 6R, 6aR) -6- (2-Chloro-6- (3-iodobenzylamino) -9H- purin-9- yl) -2,2- dimethyltetrahydrofur [3,4-d] [1,3] dioxol-4-yl) methanol (9)
The intermediate compound (20 g, 24.09 mmol) prepared in the above step 2 was dissolved in methanolic ammonia (1 L) and stirred at room temperature for 3 days. The reaction mixture was concentrated under reduced pressure to obtain a triol intermediate. The triol intermediate thus obtained (20 g, 38.63 mmol) was dissolved in anhydrous acetone (400 mL), and 2,2-dimethoxypropane (23.68 mL, 193.15 mmol) and p-toluenesulfonic acid monohydrate (7.34 g, 38.63 mmol) was added dropwise thereto, followed by stirring at room temperature for 12 hours. After confirming the completion of the reaction, saturated sodium hydrogencarbonate solution (400 mL) was added thereto. The reaction mixture was concentrated under reduced pressure. The organic layer was extracted with chloroform (4 x 250 mL), washed with a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. The reaction mixture was concentrated under reduced pressure, and the obtained residue was then separated by column chromatography to obtain the intermediate compound ((3aR, 4R, 6R, 6aR) -6- (2-Chloro-6- (3-iodobenzylamino) Yl] -2,2-dimethyltetrahydrofuro [3,4-d] [1,3] dioxol-4-yl) methanol (12 g, 89.35%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 )? Ppm 1.36 (s, 3H), 1.62 (s, 3H), 3.77-3.80 (dd, J = 12.71, 1.95 Hz, 1H), 3.95-3.98 12.71, 1.95 Hz, 1H), 4.48-4.49 (d, J = 1.46 Hz, 1H), 4.68 (br. s., 1H, exchangeable with d 2 O, OH), 4.74 (br. s., 2H), (D, J = 5.86, 1.46 Hz, 1H), 5.15-5.17 (t, J = 5.38 Hz, 1H), 5.77-5.78 (d, J = 4.40 Hz, 1H), 6.81 (br s. , 1H, exchangeable with d 2 O, NH), 7.03-7.06 (t, J = 7.82 Hz, 1H), 7.30-7.31 (d, J = 7.33 Hz, 1H), 7.59-7.61 (d, J = 7.82 Hz , & Lt; / RTI & gt; 1H), 7.67 (s, 1H), 7.70 (s, 1H); 13 C NMR (125 MHz; CDCl 3) [delta] 25.26, 27.63, 43.93, 63.37, 81.52, 82.98, 86.12, 93.89, 94.55, 114.18, 120.09, 127.19, 130.44, 136.86 (2), 139.93, 140.01, 148.80, 154.50, 155.14; mp = 82-86 [deg.] C.
Step 4: (2S, 5R) -5- (2-Chloro-6- (3-iodobenzylamino) -9H- purin-9- yl) -3,4- dihydroxy- -2-carboxamide & lt; / RTI & gt; (10)
The intermediate compound (15 g, 26.89 mmol) prepared in step 3 was dissolved in a solution of acetonitrile-water (130 mL, 1: 1) and then (diacetoxy iodo) -benzene (19 g, 59.16 mmol) 2,2,6,6-Tetramethyl 1-piperidinyloxyl (840 mg, 5.37 mmol) was added dropwise, followed by stirring at room temperature for 4 hours. After confirming the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain an acid intermediate without purification. The obtained intermediate (15 g, 26.23 mmol) was dissolved in anhydrous ethanol (500 mL) under a nitrogen stream, cooled to 0 ° C, thionyl chloride (9.52 mL, 131.17 mmol) was slowly added dropwise and the mixture was stirred at room temperature for 12 hours. After confirming the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain an ethyl ester intermediate without purification. Methylamine (750 mL, 2 N THF solution) was added dropwise to the resulting ethyl ester intermediate (15.5 g, 25.84 mmol) and the mixture was stirred at room temperature for 12 hours. After confirming the completion of the reaction, the reaction mixture was concentrated under reduced pressure. The obtained residue was purified by column chromatography to obtain the intermediate compound (2S, 5R) -5- (2-Chloro-6- (3- -9-yl) -3,4-dihydroxy-N-methyltetrahydrofuran-2-carboxamide (5 g, 31.80%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 )? Ppm 2.72-2.73 (d, J = 3.91 Hz, 3H), 4.17 (brs, 1H), 4.33 (s, 1H), 4.55-4.56 (D, J = 6.35 Hz, 1H, exchangeable with D 2 O, 2′-OH), 5.71-5.72 d, J = 3.91 Hz, 1H, exchangeable with d 2 O, 3′-OH), 5.92-5.93 (d, J = 7.33 Hz, 1H), 7.11-7.14 (t, J = 7.82 Hz, 1H), 7.35 (D, J = 6.84 Hz, 1H), 7.59-7.61 (d, J = 7.82 Hz, 1H), 7.75 (s, 1H), 8.27-8.28 exchangeable with D 2 O, NH), 8.48 (s, 1 H), 8.98-8.99 (br. t, J = 5.86 Hz, 1H, exchangeable with D 2 O, N 6 H); 13 C NMR (125 MHz; DMSO-d 6) [delta] 26.07, 43.02, 72.79, 73.42, 84.95, 88.10, 95.12, 119.46, 127.31, 130.99, 136.06, 136.51, 141.57, 142.23, 150.00, 153.44, 155.31, 170.14; mp = 207-209 [deg.] C.
Step 5: (S) -2 – ((R) -1- (2-Chloro-6- (3-iodobenzylamino) -9H- purin-9- yl) -2-hydroxyethoxy) -3 – & lt; / RTI & gt; hydroxy-N-methylpropanamide (5)
The intermediate compound (2.0 g, 3.67 mmol) prepared in step 4 was dissolved in water / methanol (210 mL, 1: 2), cooled to 0 ° C and then sodium per iodate (1.57 g, 7.34 mmol) And then stirred at the same temperature for 2 hours. After completion of the reaction was confirmed, sodium borohydride (694 mg, 18.35 mmol) was added and stirred for 1 hour. After confirming the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was concentrated under reduced pressure using toluene (3 x 50 mL). The residue was separated by column chromatography to obtain the title compound (1.58 g, 79%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 ) δ ppm 2.40 (.. Br s, 3H), 3.53-3.60 (m, 1H), 3.71-3.73 (d, J = 10.27 Hz, 1H), 3.85 (br . s., 1H), 3.95 (br. s., 2H), 4.60 (br. s., 2H), 4.98-5.00 (t, J = 5.38 Hz, 1H, exchangeable with D 2 O, OH), 5.19 -5.20 (t, J = 5.86 Hz, 1H, exchangeable with D 2 O, OH), 5.78-5.80 (t, J = 5.38 Hz, 1H), 7.10-7.13 (t, J = 7.82Hz, 1H), 7.35 -7.36 (d, J = 6.84Hz, 1H), 7.59-7.60 (d, J = 5.86 Hz, 2H, exchangeable with D 2 O, NH), 7.73 (br. s., 1H), 8.30 (s, 1H ), 8.85 (br s, 1H, exchangeable with D 2 O, NH); 13 C NMR (125 MHz; DMSO-d 6) [delta] 25.59, 42.98, 62.02, 62.25, 80.43, 84.90, 95.11, 118.57, 127.27, 130.96, 136.03, 136.45, 140.78, 142.34, 150.69, 153.53, 155.17, 169.31; HRMS (FAB) m / z calcd for C 18 H20 ClIN 6 O 4 [M + Na] + 546.0279, found 569.0162; mp = 226-229 [deg.] C.
Example 2
Preparation Example 2: Synthesis of Compound (11) ((R) -2- (1- (2-Chloro-6- (3-iodobenzylamino) -9H- purin-9-yl) -2- hydroxyethoxy) Propane-1,3-diol)
Scheme 2
The intermediate compound (230 mg, 0.27 mmol) prepared in Step 2 of Example 1 was dissolved in methanolic ammonia (25 mL) and stirred at room temperature for 3 days. The reaction mixture was concentrated under reduced pressure to obtain a triol intermediate. The obtained triol intermediate (248 mg, 0.47 mmol) was treated in the same manner as in Step 5 of Example 1 to obtain the desired compound (109 mg, 75.69%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 )? Ppm 3.13-3.17 (m, 1H), 3.22-3.26 (m, 1H), 3.43-3.47 (m, 2H), 3.54-3.56 (Br s, 2H), 4.41-4.42 (br t, J = 5.38 Hz, 1H, exchangeable with D 2 O, OH), 4.60 , J = 5.38 Hz, 1H, exchangeable with D 2 O, OH), 5.13 (br. s., 1H, exchangeable with D 2 O, OH), 5.80-5.82 (t, J = 4.89 Hz, 1H), 7.11 (D, J = 7.33 Hz, 1H), 7.36-7.37 (d, J = 7.33 Hz, 1H), 7.59-7.60 s, 1 H), 8.82 (br s, 1H, exchangeable with D 2 O, NH); 13 C NMR (125 MHz; DMSO-d 6) [delta] 43.01, 61.12, 61.23, 62.64, 80.90, 84.53, 95.12, 118.56, 127.36, 130.99, 136.04, 136.58, 140.68, 142.44, 150.73, 153.40, 155.12; HRMS (FAB) m / z calcd for C 17 H 19 ClIN 5 O 4 [M + Na] + 519.0170, found 542.0054; mp = 170-172 [deg.] C.
Example 3
Preparation Example 3: Synthesis of Compound (12) ((S) -3-Hydroxy-2 – ((R) -2-hydroxy- 1- (6- (3-iodobenzylamino) -9H- Yl) ethoxy) -N-methylpropanamide & lt; / RTI & gt;
Scheme 3
Step 1: ((3aR, 4R, 6R, 6aR) -6- (6-Chloro-9H- purin-9- yl) -2,2- dimethyltetrahydrofuro [3,4 d] [1,3 ] Dioxol-4-yl) methanol (14)
(Hydroxymethyl) tetrahydrofuran-3,4-diol (4.8 g, 16.74 mmol) and 2,2 & lt; RTI ID = 0.0 & -Dimethoxypropane (10.26 mL, 83.71 mmol) was dissolved in anhydrous acetone (120 mL) under a nitrogen stream, p-toluenesulfonic acid monohydrate (3.18 g, 16.74 mmol) was added dropwise and the mixture was stirred at room temperature for 4 hours . After confirming the completion of the reaction, the reaction is terminated with a saturated sodium hydrogencarbonate solution. The reaction solution was concentrated under reduced pressure, and the organic layer was extracted with chloroform (4 x 20 mL), washed with a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. After concentration under reduced pressure, the obtained residue was separated by column chromatography to obtain the intermediate compound ((3aR, 4R, 6R, 6aR) -6- (6-Chloro-9H-purin-9- 3,4-d] [1,3] dioxol-4-yl) methanol (4.87 g, 89.03%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 )? Ppm 1.38 (s, 3H), 1.65 (s, 3H), 3.80-3.83 (dd, J = 12.22, 1.46 Hz, 1H), 3.95-3.98 (Dd, J = 5.86, 1.46 Hz, 1H), 5.19 (d, J = 8.6 Hz, 1H), 4.53-4.55 5.21 (dd, J = 5.86, 4.40 Hz, 1H), 5.99-6.00 (d, J = 4.89 Hz, 1H), 8.25 (s, 1H), 8.75 (s, 1H); 13 C NMR (125 MHz; CDCl 3 )? 25.22, 27.55, 63.22, 81.51, 83.35, 86.43, 94.02, 114.51, 133.25, 144.73, 150.50, 151.71, 152.31; mp = 146-150 [deg.] C.
Step 2: ((3aR, 4R, 6R, 6aR) -6- (6-Chloro-9H-purin-9- yl) -2,2- dimethyltetrahydrofuro [3,4- d] 3] dioxol-4-yl) methyl benzoate (15)
The intermediate compound (2.8 g, 8.56 mmol) prepared in Step 1 was dissolved in anhydrous methylene chloride (100 mL), and then cooled to 0 ° C. Triethylamine (3.6 mL, 25.70 mmol) and dimethylaminopyridine (21 mg, 0.17 mmol). Benzoyl chloride (1.5 mL, 12.85 mmol) is slowly added dropwise at the same temperature and then stirred at room temperature for 2 hours. After confirming the completion of the reaction, the reaction is terminated with a saturated sodium hydrogencarbonate solution. The reaction solution was concentrated under reduced pressure, the organic layer was extracted with methylene chloride, washed with a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. After concentration under reduced pressure, the obtained residue was separated by column chromatography to obtain the intermediate compound ((3aR, 4R, 6R, 6aR) -6- (6-Chloro-9H-purin-9- 3,4-d] [1,3] dioxol-4-yl) methyl benzoate (3.68 g, 99.72%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm 1.40 (s, 3H), 1.62 (s, 3H), 4.42-4.46 (dd, J = 11.73, 3.9 Hz, 1H), 4.61-4.64 (m, 2H) (D, J = 7.33 Hz, 2H), 5.51-5.13 (d, J = 2.93 Hz, 1H), 5.53-5.54 7.47-7.50 (t, J = 7.33 Hz, 1 H), 7.79-7.81 (d, J = 7.82 Hz, 2H), 8.21 (s, 1H), 8.64 (s, 1H); 13 C NMR (125 MHz; CDCl 3 ) δ 25.36, 27.15, 63.99, 81.42, 84.07, 85.04, 91.87, 114.92, 128.31 (2), 129.07, 129.39 (2), 132.42, 133.33, 144.10, 150.79, 151.40, 152.02 , 165.80; mp = 50-54 [deg.] C.
Step 3: ((3aR, 4R, 6R, 6aR) -6- (6- (3-Iodobenzylamino) -9H- purin- 9-yl) -2,2 dimethyltetrahydrofuro [3,4 -d] [1,3] dioxol-4-yl) methyl benzoate (16)
The intermediate compound (1.24 g, 2.87 mmol) prepared in the above step 2 was prepared in the same manner as in step 2 of Example 1 to give the intermediate compound ((3aR, 4R, 6R, 6aR) -6- (6- Yl) -2,2-dimethyltetrahydrofuro [3,4-d] [1,3] dioxol-4-yl) methyl benzoate (1.73 g, 96.11 %).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 )? Ppm 1.42 (s, 3H), 1.63 (s, 3H), 4.45-4.59 (m, 1H), 4.59-4.61 (m, 2H), 4.80 (br s. J = 5.38 Hz, 1H), 5.17 (d, J = 3.43 Hz, 1H), 5.58-5.59 , 7.32-7.05 (t, J = 7.33 Hz, 2H), 7.49-7.52 (t, J = = 7.33 Hz, 1 H), 7.58-7.60 (d, J = 7.33 Hz, 1 H), 7.71 (s, (br. s., 1 H); 13 C NMR (125 MHz; CDCl 3 ) δ 25.47, 27.21, 43.81, 64.35, 81.71, 84.21, 85.03, 91.38, 94.55, 114.61, 120.52, 126.85, 128.32 (3), 129.43, 129.62 (2), 130.34, 133.18 , 136.53, 139.16, 140.99, 148.68, 153.34, 154.60, 166.02; mp = 68-72 [deg.] C.
Step 4: ((2R, 3R, 4R, 5R) -3,4-Bis (tert- butyldimethylsilyloxy) -5- (6- (3- iodobenzylamino) -9H-purin- ) Tetrahydrofuran-2-yl) methyl benzoate (17)
The intermediate compound (4.93 g, 7.85 mmol) prepared in the above step 3 was dissolved in 80% acetic acid (250 mL), and the mixture was refluxed at 100 ° C for 12 hours. After completion of the reaction was confirmed, the reaction solution was concentrated under reduced pressure, toluene (4 x 50 mL) was added, and the filtrate was concentrated under reduced pressure to obtain a diol intermediate without purification. The obtained diol intermediate (8.5 g, 14.47 mmol) was dissolved in anhydrous pyridine (250 mL), followed by addition of tetrabutyldimethylsilyl triflate (TBDMSOTf) (13.3 mL, 57.88 mmol) followed by stirring at 50 ° C for 5 hours. After confirming the completion of the reaction, the reaction solution was partitioned into methylene chloride / water. The organic layer was washed with water, saturated sodium hydrogencarbonate solution and saturated saturated sodium bicarbonate solution, and then dried over anhydrous magnesium sulfate. After concentration under reduced pressure, the obtained residue was separated by column chromatography to obtain the intermediate compound ((2R, 3R, 4R, 5R) -3,4-bis (tert-butyldimethylsilyloxy) -9H-purin-9-yl) tetrahydrofuran-2-yl) methylbenzoate (4.47 g, 69.73%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm -0.17 (s, 3H), 0.01 (s, 3H), 0.1 (s, 3H), 0.12 (s, 3H), 0.83 (s, 9H), 0.93 ( (t, J = 4.40 Hz, 1H), 4.74-4.78 (dd, J = J = 4.40 Hz, 1H), 4.82 (br s, 2H), 5.07-5.09 (t, J = 4.40 Hz, 1H), 5.88-5.89 (d, J = 7.82 Hz, 1H), 7.31-7.33 (d, J = 7.82 Hz, 1H), 7.38-7.41 (t, J = 7.82 Hz, 1H) , 7.52-7.55 (t, J = 7.82 Hz, 1H), 7.59-7.60 (d, J = 7.82 Hz, 1H), 7.72 (s, 1H), 7.84 dd, J = 8.31, 0.97 Hz, 2H), 8.32 (s, 1H); 13 C NMR (125 MHz; CDCl 3)? -0.00, 0.11, 0.26, 0.54, 30.63 (3), 34.61 (2), 49.19, 68.45, 77.09, 79.28, 87.24, 94.71, 99.46, 125.63, 131.74, 133.32 , 134.59, 135.23, 138.10, 141.41, 141.46, 144.66, 145.99, 153.87, 157.99, 159.53, 171.15; mp = 68-70 [deg.] C.
Step 5: ((2R, 3R, 4R, 5R) -3,4-Bis (tert-butyldimethylsilyloxy) -5- (6- (3- iodobenzylamino) -9H-purin- ) Tetrahydrofuran-2-yl) methanol (18)
The intermediate compound (1.28 g, 1.56 mmol) prepared in step 4 was dissolved in anhydrous methanol (100 mL), 25% sodium methoxide / methanol (15 mL) was added, and the mixture was stirred at room temperature for 12 hours. After confirming the completion of the reaction, the reaction solution was concentrated under reduced pressure, and the obtained residue was separated by column chromatography to obtain the intermediate compound ((2R, 3R, 4R, 5R) -3,4- bis (tert- butyldimethylsilyloxy) Yl) tetrahydrofuran-2-yl) methanol (960 mg, 86.48%) was obtained as a pale-yellow amorphous solid.
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm -0.58 (s, 3H), -0.13 (s, 3H), 0.11-0.13 (d, J = 7.33 Hz, 6H), 0.74 (s, 9H), 0.95 (s, 9H), 3.68-3.71 (d, J = 12.71 Hz, 1H), 3.92-3.95 (d, J = 12.71 Hz, (D, J = 7.33, 4.40 Hz, 1H), 5.75-5.77 (d, J = 7.82 Hz, 1H), 6.39 (br s). J = 7.82 Hz, 1H), 7.30-7.32 (d, J = 7.82 Hz, 1H), 7.59-7.61 (d, J = 7.82 Hz, 1H), 7.70 (s, 1H), 7.76 (br s, 1H), 8.35 (br s, 1H); 13 C NMR (125 MHz; CDCl 3 ) δ 0.00, 1.30, 1.35, 1.38, 31.62 (3), 31.75 (3), 35.61 (2), 49.54, 68.95, 79.91, 79.96, 95.50, 96.96, 100.51, 127.37, 132.70, 136.30, 142.42, 142.57, 146.54, 146.61, 153.78, 158.46, 160.79; mp = 82-86 [deg.] C.
Step 6: (2S, 5R) -3,4-Bis (tert-butyldimethylsilyloxy) -5- (6- (3- iodobenzylamino) -9H- purin- Preparation of tetrahydrofuran-2-carboxamide (19)
The intermediate compound (450 mg, 0.63 mmol) prepared in Step 5 and pyridinium dichromate (5.47 g, 14.54 mmol) were dissolved in DMF (50 mL) under a nitrogen stream, followed by stirring at room temperature for 12 hours. After confirming completion of the reaction, the resulting solid was washed with water to obtain an acid intermediate. The obtained intermediate (450 mg, 0.62 mmol) was dissolved in anhydrous ethanol (10 mL) under a nitrogen stream, cooled to 0 ° C, thionyl chloride (0.25 mL, 3.10 mmol) was slowly added dropwise and the mixture was stirred at room temperature for 5 hours Lt; / RTI & gt; After completion of the reaction was confirmed, the reaction solution was concentrated under reduced pressure, and the residue was partitioned into ethyl acetate / water. The organic layer was washed with water and saturated aqueous sodium chloride solution, and dried over anhydrous magnesium sulfate. After concentration under reduced pressure, an intermediate ethyl ester was obtained. The ethyl ester thus obtained is added with a methylamine / 2N-THF solution under a nitrogen stream, followed by stirring at room temperature for 12 hours. After confirming the completion of the reaction, the reaction mixture was concentrated under reduced pressure and the residue was purified by column chromatography to obtain the intermediate compound (2S, 5R) -3,4-bis (tert-butyldimethylsilyloxy) -5- -9H-purin-9-yl) -N-methyltetrahydrofuran-2-carboxamide (400 mg, 85.65%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm -0.61 (s, 3H), -0.16 (s, 3H), 0.15 (s, 3H), 0.25 (s, 3H), 0.71 (s, 9H), 0.97 (s, 9H), 2.93-2.94 (d, J = 4.40 Hz, 3H), 4.33-4.34 (d, J = 3.42 Hz, (D, J = 7.33 Hz, 1H), 6.42 (br s, 1H), 7.03-7.06 (t, J = 7.82 Hz, 1H), 7.30-7.32 ), 7.59-7.61 (d, J = 7.82 Hz, 1H), 7.70 (s, 1H), 7.75 (s, 1H), 8.36 , 1H); 13 C NMR (125 MHz; CDCl 3 ) δ 0.00, 1.19, 1.21, 1.40, 23.71, 24.00, 31.53 (3), 31.58, 31.78 (2), 35.64, 49.60, 78.09, 81.25, 92.53, 95.48, 100.56, 127.30 , 132.72, 136.34, 142.44, 142.61, 146.64, 146.79, 154.27, 158.70, 160.96, 175.92; mp = 80-84 [deg.] C.
Step 7: (2S, 5R) -3,4-Dihydroxy-5- (6- (3-iodobenzylamino) -9H- purin-9- yl) -N- methyltetrahydrofuran- Manufacture of Radiate (20)
The intermediate compound (65 mg, 0.08 mmol) prepared in Step 6 was dissolved in anhydrous THF under a nitrogen stream, and then tetrabutylammonium fluoride (TBAF) (0.44 mL, 0.43 mmol, (1 M solution THF) Stir at room temperature for 1 hour. After confirming the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by column chromatography to obtain the intermediate compound (2S, 5R) -3,4-dihydroxy-5- (6- (3-iodobenzylamino) -9H-purin-9-yl) -N-methyltetrahydrofuran-2-carboxamide (52 mg, 95.45%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 ) δ ppm 2.70-2.71 (d, J = 4.40 Hz, 3H), 4.14-4.16 (t, J = 3.91 Hz, 1H), 4.31 (s, 1H), 4.57 (D, J = 4.40 Hz, 1H), 4.67 (d, 1H), 5.96-5.97 (d, J = 7.33 Hz, 1H), 7.09-7.12 (t, J = 7.82 Hz, 1H), 7.35-7.36 (d, J = 7.82 Hz, 1H), 7.56-7.58 1H, J = 7.82 Hz, 1H), 7.72 (s, 1H), 8.29 (s, 1H), 8.42 (s, 1H), 8.53 (br s., 1H), 8.85-8.86 (m, 1H); 13 C NMR (125 MHz; DMSO-d 6 )? 25.81, 42.74, 72.56, 73.49, 85.09, 88.26, 95.06, 120.42, 127.11, 130.95, 135.86, 136.13, 141.17, 143.10, 148.70, 152.94, 154.86, 170.34; mp = 178-182 [deg.] C.
Step 8: (S) -3-Hydroxy-2 – ((R) -2-hydroxy-1- (6- (3-iodobenzylamino) -9H- purin- Preparation of N-methylpropanamide (12)
The intermediate compound (52 mg, 0.10 mmol) prepared in the above Step 7 was treated in the same manner as in Step 5 of Example 1 to obtain the title compound (35 mg, 83.33%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 .) Δ ppm 2.35 (s, 3H), 3.5-3.6 (m, 1H), 3.72-3.74 (d, J = 9.29 Hz, 1H), 3.86 (br s. 2H), 4.99 (br s, 1H, exchangeable with D2O, OH), 5.21 (br. S., 1H), 4.00 (d, J = (d, J = 6.84 Hz, 1H), 7.56 d, J = 6.35 Hz, 2H, exchangeable with D 2 O, NH), 7.71 (s, 1H), 8.22 (s, 1H), 8.30 (s, 1H), 8.37 (br. s., 1H, exchangeable with D2O, NH); 13 C NMR (125 MHz; DMSO-d 6 ) δ 25.53, 42.81, 61.98, 62.26, 80.30, 84.62, 95.09, 119.43, 127.11, 130.91, 135.81, 136.16, 140.19, 143.31, 149.79, 152.98, 154.66, 169.42; HRMS (FAB) m / z calcd for C 1821 IN 6 O 4 [M + Na] +512.0669, found 535.0578; mp = 176-182 [deg.] C.
Example 4
Production Example 4: Synthesis of Compound (21) ((R) -2- (2-hydroxy-1- (6- (3-iodobenzylamino) -9H- purin- 3-diol)
Scheme 4
Step 1: ((3aR, 4R, 6R, 6aR) -6- (6- (3-Iodobenzylamino) -9H-purin-9- yl) -2,2-dimethyltetrahydrofuro [ 4-d] [1,3] dioxol-4-yl) methanol (22)
The intermediate compound (1.73 g, 2.75 mmol) prepared in the step 2 of Example 1 was treated in the same manner as in the step 5 of Example 3 with (3aR, 4R, 6R, 6aR) -6- L, 3-dioxol-4-yl) methanol (1.04 g, 93.69 & lt; RTI ID = 0.0 & gt; %).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CDCl 3 ) δ ppm 1.36 (s, 3H), 1.63 (s, 3H), 3.75-3.80 (t, J = 11.24 Hz, 1H), 3.95-3.97 (d, J = 12.71 Hz , 5.19 (br, s, 2H), 5.10-5.11 (d, J = 4.89 Hz, 1H), 5.19 = 3.91 Hz, 1H), 6.58 (br s, 2H), 7.01-7.04 (t, J = 7.82 Hz, 1H), 7.29-7.30 (d, J = 6.84 Hz, 1H), 7.58-7.59 , J = 7.33 Hz, 1H), 7.69 (br s, 2H), 8.33 (br s, 1H); 13 C NMR (125 MHz; CDCl 3 ) δ 25.24, 27.66, 29.65, 63.36, 81.68, 83.03, 86.12, 94.26, 94.54, 113.93, 121.24, 126.80, 130.32, 136.52, 136.58, 139.71, 140.72, 147.66, 152.73, 154.94 ; mp = 72-76 [deg.] C.
Step 2: (2R, 3S, 4R, 5R) -2- (hydroxymethyl) -5- (6- (3-iodobenzylamino) -9H- purin-9- yl) tetrahydrofuran- – Preparation of diol (23)
The intermediate compound (250 mg, 0.47 mmol) prepared in the above step 1 was dissolved in 80% acetic acid (250 mL), and the mixture was heated under reflux at 100 ° C for 12 hours. After confirming the completion of the reaction, the reaction solution was concentrated under reduced pressure, toluene (4 x 50 mL) was added, and the mixture was concentrated under reduced pressure. The obtained residue was purified by column chromatography to obtain the intermediate (2R, 3S, 4R, Methyl) -5- (6- (3-iodobenzylamino) -9H-purin-9-yl) tetrahydrofuran-3,4-diol (177 mg, 76.95%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; DMSO-d 6 )? Ppm 3.56 (br s, 1H), 3.67-3.69 (d, J = 10.27 Hz, 1H), 3.97 ), 4.61-4.67 (m, 3H), 5.17 (d, J = 2.93 Hz, 1H, exchangeable with D 2 O, OH), 5.35-5.36 (t, J = 5.38 Hz, 1H, exchangeable with D 2 O, OH), 5.43-5.44 (d, J = 5.38 Hz, 1H, exchangeable with d 2O, OH), 5.89-5.90 (d, J = 4.89 Hz, 1H), 7.09-7.11 (t, J = 7.33 Hz, 1H), 7.35-7.36 (d, J = 6.84 Hz, 1H), 7.56-7.58 (d, J = 7.33 Hz, 1H), 7.72 ), 8.45 (br s, 1H, exchangeable with D 2 O, NH); 13 C NMR (125 MHz; DMSO-d 6) [delta] 42.69, 62.08, 71.06, 73.97, 86.32, 88.42, 95.09, 120.24, 127.08, 130.91, 135.81, 136.16, 140.47, 143.22, 149.00, 152.76, 154.79; mp = 174-178 [deg.] C.
Step 3: (R) -2- (2-Hydroxy-1- (6- (3-iodobenzylamino) -9H-purin-9-yl) ethoxy) propane- )
The intermediate compound (77 mg, 0.15 mmol) prepared in the above Step 2 was treated in the same manner as in Step 5 of Example 1 to obtain the desired compound (62 mg, 80.51%).
The analytical data of the obtained compound are as follows.
1 H NMR (500 MHz; CD 3 OD)? Ppm 3.42 – 3.44 (d, J = 5.38 Hz, 2H), 3.54-3.58 (m, 1H), 3.65-3.68 ), 3.75-3.78 (dd, J = 11.73,4.44 Hz, 1H), 4.01-4.02 (d, J = 5.38 Hz, 2H), 4.53 (s, 2H), 6.04-6.06 J = 7.82 Hz, 1H), 7.76 (s, 1H), 7.07-7.10 (t, J = 7.82 Hz, 1H), 7.38-7.40 (d, J = 7.82 Hz, 1H), 7.59-7.60 ), 8.27 (s, 1 H), 8.29 (s, 1 H); 13 C NMR (125 MHz; CD 3 OD)? 42.88, 60.80, 61.25, 62.76, 80.26, 84.02, 93.52, 119.00, 126.44, 129.93, 135.90, 136.10, 139.65, 141.72, 148.97, 152.52, 154.53; HRMS (FAB) m / z calcd for C 17 H 20 IN 5 O 4 [M + H] +485.0560, found 486.0625; mp = 72-76 [deg.] C.

PATENT

WO 2008111082

REFERENCES

1: Avni I, Garzozi HJ, Barequet IS, Segev F, Varssano D, Sartani G, Chetrit N, Bakshi E, Zadok D, Tomkins O, Litvin G, Jacobson KA, Fishman S, Harpaz Z, Farbstein M, Yehuda SB, Silverman MH, Kerns WD, Bristol DR, Cohn I, Fishman P. Treatment of Dry Eye Syndrome with Orally Administered CF101 Data from a Phase 2 Clinical Trial. Ophthalmology. 2010 Mar 19. [Epub ahead of print] PubMed PMID: 20304499.

2: Bar-Yehuda S, Rath-Wolfson L, Del Valle L, Ochaion A, Cohen S, Patoka R, Zozulya G, Barer F, Atar E, Piña-Oviedo S, Perez-Liz G, Castel D, Fishman P. Induction of an antiinflammatory effect and prevention of cartilage damage in rat knee osteoarthritis by CF101 treatment. Arthritis Rheum. 2009 Oct;60(10):3061-71. PubMed PMID: 19790055.

3: Borea PA, Gessi S, Bar-Yehuda S, Fishman P. A3 adenosine receptor: pharmacology and role in disease. Handb Exp Pharmacol. 2009;(193):297-327. Review. PubMed PMID: 19639286.

4: Moral MA, Tomillero A. Gateways to clinical trials. Methods Find Exp Clin Pharmacol. 2008 Mar;30(2):149-71. PubMed PMID: 18560631.

5: Silverman MH, Strand V, Markovits D, Nahir M, Reitblat T, Molad Y, Rosner I, Rozenbaum M, Mader R, Adawi M, Caspi D, Tishler M, Langevitz P, Rubinow A, Friedman J, Green L, Tanay A, Ochaion A, Cohen S, Kerns WD, Cohn I, Fishman-Furman S, Farbstein M, Yehuda SB, Fishman P. Clinical evidence for utilization of the A3 adenosine receptor as a target to treat rheumatoid arthritis: data from a phase II clinical trial. J Rheumatol. 2008 Jan;35(1):41-8. Epub 2007 Nov 15. PubMed PMID: 18050382

/////////////CF 101, Piclidenoson, CF101, CF-101, CF 101, ALB-7208,  ALB 7208, ALB7208,  IB MECA, Phase III,  Plaque psoriasis, Rheumatoid arthritis, UNII-30679UMI0N, Пиклиденозон بيكليدينوسون 匹利诺生 , Can-Fite BioPharma

CNC(=O)[C@H]1O[C@H]([C@H](O)[C@@H]1O)N1C=NC2=C(NCC3=CC(I)=CC=C3)N=CN=C12

Peficitinib hydrobromide, ペフィシチニブ臭化水素酸塩


1353219-05-2.png

Structure of PEFICITINIB HYDROBROMIDE

img

ChemSpider 2D Image | PEFICITINIB HYDROBROMIDE | C18H23BrN4O2

Peficitinib hydrobromide

ペフィシチニブ臭化水素酸塩

ASP015K,

Rheumatoid Arthritis

1H-Pyrrolo(2,3-b)pyridine-5-carboxamide, 4-((5-hydroxytricyclo(3.3.1.13,7)dec-2-yl)amino)-, hydrobromide (1:1), stereoisomer

4-{[(1R,2s,3S,5r)-5-Hydroxyadamantan-2-yl]amino}-1H-pyrrolo[2,3-b]pyridine-5-carboxamide hydrobromide (1:1)

1H-Pyrrolo[2,3-b]pyridine-5-carboxamide, 4-[[(1R,3S)-5-hydroxytricyclo[3.3.1.13,7]dec-2-yl]amino]-, hydrobromide (1:1)

U55XHZ5X6P

Formula
C18H22N4O2. HBr
CAS
1353219-05-2 HBR
944118-01-8 BASE
Mol weight
407.3048

PMDA, 2019/3/26 JAPAN APPROVED, Smyraf

Image result for Peficitinib hydrobromide

Peficitinib hydrobromide is used in the treatment of Psoriasis and Rheumatoid Arthritis

Peficitinib (formerly known as ASP015K) is a pyrrolo[2,3-b]pyridine derivative orally administered once-daily JAK inhibitor in development for the treatment of Rheumatoid Arthritis. In preclinical studied Peficitinib inhibited JAK1 and JAK3 with IC50 of 3.9 and 0.7 nM, respectively. Peficitinib also inhibited IL-2-dependent T cell proliferation in vitro and STAT5 phosphorylation in vitro and ex vivo. Furthermore, Peficitinib dose-dependently suppressed bone destruction and paw swelling in an adjuvant-induced arthritis model in rats via prophylactic or therapeutic oral dosing regimens.In clinical trials, Peficitinib treatment prescribed at 50, 100 and 150 mg amounts each showed statistically significantly higher ACR20 response rates compared to the placebo and response rates increased up to the 150 mg dosage. Adverse events included neutropenia, headache, and abdominal pain. The treatment-emergent adverse events occurring more frequently in the Peficitinib group compared with the placebo group included diarrhea, nasopharyngitis, and increased serum creatine phosphokinase activity. No cases of serious infections were reported. Herpes zoster occurred in four patients (two each in the peficitinib 25 and 100 mg cohorts). The authors concluded that treatment with peficitinib as monotherapy for 12 weeks in Japanese patients with moderate to severe RA is efficacious and showed an acceptable safety profile.

SYN

CLIP

Bioorganic & Medicinal Chemistry

Volume 26, Issue 18, 1 October 2018, Pages 4971-4983

Discovery and structural characterization of peficitinib (ASP015K) as a novel and potent JAK inhibitor

Abstract

Janus kinases (JAKs) are considered promising targets for the treatment of autoimmune diseases including rheumatoid arthritis (RA) due to their important role in multiple cytokine receptor signaling pathways. Recently, several JAK inhibitors have been developed for the treatment of RA. Here, we describe the identification of the novel orally bioavailable JAK inhibitor 18, peficitinib (also known as ASP015K), which showed moderate selectivity for JAK3 over JAK1, JAK2, and TYK2 in enzyme assaysChemical modification at the C4-position of lead compound 5 led to a large increase in JAK inhibitory activity and metabolic stability in liver microsomes. Furthermore, we determined the crystal structures of JAK1, JAK2, JAK3, and TYK2 in a complex with peficitinib, and revealed that the 1H-pyrrolo[2,3–b]pyridine-5-carboxamide scaffold of peficitinib forms triple hydrogen bonds with the hinge region. Interestingly, the binding modes of peficitinib in the ATP-binding pockets differed among JAK1, JAK2, JAK3, and TYK2. WaterMap analysis of the crystal structures suggests that unfavorable water molecules are the likely reason for the difference in orientation of the 1H-pyrrolo[2,3-b]pyridine-5-carboxamide scaffold to the hinge region among JAKs.

Image result for Peficitinib hydrobromide

Image result for Peficitinib hydrobromide

Image result for Peficitinib hydrobromide

PATENT

WO 2011162300

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2011162300&tab=FULLTEXT&queryString=%28PA%2FAstellas%29+&recNum=126&maxRec=386

Diseases that have good JAK3 inhibitory activity and are caused by undesired cytokine signaling (eg rejection in living transplantation, rheumatism, psoriasis, autoimmune diseases, asthma, atopic dermatitis, Alzheimer’s disease, atherosclerosis etc. Patent Document 1 discloses fused heterocyclic compounds and salts thereof which are useful as therapeutic agents and / or prophylactic agents for diseases (eg, cancer, leukemia, etc.) caused by abnormal cytokine signaling. Among them, 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo represented by the following formula (I) disclosed in the example compound Ex121 [2,3-b] pyridine-5-carboxamide exhibits good activity, and is particularly a compound expected as a therapeutic agent for suppressing rejection during organ / tissue transplantation, rheumatism, psoriasis and the like.
[Chemical formula 1]
 The solid stability of a compound which has become a drug development candidate is an important factor both in industrial operation and in maintaining quality. In the stability of the drug substance itself, it is necessary to evaluate the stability of the quality necessary to maintain the efficacy and safety of the drug, and to obtain the information necessary for setting the storage method and the shelf life of the drug. For this reason, the stability test is considered to be one of the most important tests in the manufacture of pharmaceuticals (Heat measurement, 2004, 31 (2), pp. 80-86).
 Patent Document 1 discloses the free form of the compound of the formula (I) but does not disclose as a crystal. There is a need for a drug substance which is more suitable for formulation, and is physically and chemically stable from the viewpoint of quality assurance.
Example 1
(Production Method of Hydrobromide Salt Form B 45)
(In the Case of Addition of Seed Crystals )
After nitrogen substitution, the reaction vessel was charged with 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy- 2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide (145.0 kg), water (377 L), EtOH (1508 L), 48% hydrobromic acid (74.9 kg) It charged sequentially at room temperature and started stirring. 48% hydrobromic acid was added, taking care that the pH was in the range of 1.5 to 1.9. The reaction mixture was heated and stirred until the internal temperature reached 70 ° C. or higher. After confirming that the solution was completely dissolved, the solution was stirred for 5 minutes or more, and the solution was subjected to clear filtration at an internal temperature of 70 ° C. or higher, and the pot and line were washed with warm EtOH (290 L). At an internal temperature of about 50 ° C., 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide odor Hydrochloric acid salt seed crystals (B45, 145 g) were added, and the mixture was ripened and stirred overnight at an internal temperature of 40 to 50 ° C. Subsequently, the mixture was cooled to an internal temperature of 20 to 30 ° C. over 1 hour or more, and the mixture was aged and stirred at the same temperature for 1 hour or more. At an internal temperature of 20 to 30 ° C., EtOAc (4350 L) was added dropwise over 1 hour, and the mixture was aged and stirred overnight at the same temperature. The precipitated crystals were filtered. The wet crystals were washed with a solution of EtOH / EtOAc (145 L / 290 L). The wet crystals are dried under reduced pressure at an external temperature of 40 ° C. overnight under reduced pressure to give 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3 -b] Pyridine-5-carboxamide hydrobromide crystal (B45, 161 kg) was obtained.

[0037]
(Another method of producing hydrobromide salt B45 type crystal)
(In the case of no addition of seed crystals) After
sufficiently drying the reaction vessel and replacing with nitrogen, water (585 L) is charged and subsequently 4- {[ (1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide (225 kg), EtOH (2250 L) was charged Stirring was started. The internal temperature was adjusted to 25 ° C., and 48% hydrobromic acid (127.8 kg) was charged at the same temperature, and the vessel and kettle wall were washed with EtOH (90 L). After the completion of the charging, it was confirmed that the reaction solution had been dissolved, the pH was measured, and the pH was confirmed to be in the range of 1.5 to 1.9. When the pH was out of the range, the pH was adjusted to a predetermined pH using 48% hydrobromic acid (48% hydrobromic acid: about 11.6 kg). The temperature was raised until the internal temperature reached 70 ° C., and after confirmation of dissolution, the mixture was stirred for 5 minutes or more. The solution was subjected to clear filtration while maintaining the internal temperature at 60 ° C. or higher, and washed through a filter from a dissolution vessel with warm EtOH (450 L) preheated to 50 ° C. or higher. The clarified filtrate was gradually cooled to an internal temperature of 45 ° C., and filtered EtOAc (6750 L) was added dropwise over 6 hours at an internal temperature of 45 ° C. After the dropping was completed, the mixture was stirred at an internal temperature of 45 ° C. for 10 hours or more. Subsequently, it was cooled to an internal temperature of 25 ° C. using a follow-up temperature control cooler, and stirred at an internal temperature of 25 ° C. for 3 hours. The predetermined supernatant concentration and the crystal form of the precipitated crystals were confirmed and filtered. A mixed solvent of EtOH / EtOAc (225 L / 450 L) was prepared and cake washed using this mixed solvent. The obtained wet crystals are dried under reduced pressure at an external temperature of 40 ° C. for 10 hours or more, and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [ 2,3-b] pyridine-5-carboxamido hydrobromide crystal (B45, 250 kg) was obtained.

[0038]
1 H-NMR (600 MHz, d 6 -DMSO) δ: 1.49 (2 H, m), 1. 68 (2 H, m), 1.71 (2 H, m), 1. 80 (2 H, m), 1. 91 (2 H, m), 2.10 (1H, m), 2.20 (2 H, m), 3. 70-4.00 (1 H, brs), 4. 28 (1 H, m), 6. 66 (1 H, m), 7. 39 (1 H, m), 7. 75 (1 H, brs), 8. 38 (1 H, brs), 8.5 5 (1 H, s), 11. 17 (1 H, d, 7.8 Hz), 12.5 (1 H, brs), 14. 17 (1 H, brs)
Elemental analysis: theoretical value: C 53.08%, H 5.69% , N 13.76%, O 7.86% , Br 19.62%;
Found:. C 53.02%, H 5.74 %, N 13.73%, Br 19.42%
molecular composition: C 18 H 22 N 4 O . HBr
MS: 327.0 (M From the result of + H) +
elemental analysis, 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5 The carboxamido hydrobromide was a monohydrobromide.

[0039]
Example 2
(hydrobromide A87 crystal)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] Pyridine-5-carboxamide (6.0 g) was charged in EtOH / water (57.6 mL / 14.4 mL). At 50-60 ° C., 48% hydrobromic acid was added, stirred for 15 minutes more, and washed with EtOH (18 mL). At 45 ° C.-55 ° C. EtOAc (180 mL) was added dropwise over 30 minutes. Crystals were precipitated upon stirring at 15 ° C to 25 ° C. The crystals were collected by filtration and washed with a mixed solvent of EtOH / EtOAc (6 mL / 12 mL). The crystals are dried under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide odor Seed crystals of hydrofluoride (Form A87, 6.11 g) were obtained.
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide (3.0 g), EtOH (24 mL), water (6 mL), and 48% hydrobromic acid (1.55 g) were charged sequentially at room temperature. After charging, the mixture was heated to an internal temperature of 60 ° C. or higher and stirred. After confirming that the solution was completely in solution, the solution was subjected to clear filtration at an internal temperature of 60 ° C. or higher, and washed with warm EtOH (9 mL). EtOH (21 mL) is added dropwise at an internal temperature of 70 ° C. or higher, and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H at an internal temperature of 70 ° C. Seed crystals (A87, 30 mg) of pyrrolo [2,3-b] pyridine-5-carboxamide hydrobromide were added, and the mixture was ripened and stirred overnight at an internal temperature of 65 to 70 ° C. Subsequently, it was cooled to an internal temperature of 20 to 30 ° C., and ripening stirring was carried out at the same temperature overnight. At an internal temperature of 20 to 30 ° C., EtOAc (90 mL) was added dropwise over 1 hour, and the mixture was aged and stirred at the same temperature for 1 hour or more. The precipitated crystals were collected by filtration. The wet crystals were washed with a solution of EtOH / EtOAc (3 mL / 12 mL). The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide crystal (Form A87, 3.09 g) was obtained.

[0040]
Example 3
(hydrobromide A61 crystal)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] Pyridine-5-carboxamide (5.0 g), EtOH (48 mL), water (12 mL), and 48% hydrobromic acid (2.58 g) were charged sequentially at room temperature. After charging, the mixture was heated to an internal temperature of 70 ° C. and stirred. After confirming complete dissolution, the solution was clarified by filtration at an internal temperature of 70 ° C., and washed with warm EtOH (15 mL). The internal temperature was cooled to 50 to 60 ° C., and EtOAc (150 mL) was added dropwise over 1 hour at the same temperature. After the addition was completed, the solution was gradually cooled to 20 to 30 ° C., and the mixture was aged and stirred at the same temperature for 1 hour or more. The precipitated crystals were collected by filtration. The wet crystals were washed with a solution of EtOH / EtOAc (5 mL / 10 mL). The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide crystal (Form A61, 5.19 g) was obtained.

[0041]
Example 4
(hydrobromide A36 type crystal)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] To a suspension of pyridine-5-carboxamide (500 mg) in EtOAc, 48% hydrobromic acid (258 μL) was added, and the mixture was stirred with heating under reflux for 1 hour, and further allowed to cool to room temperature. The precipitated crystals were collected by filtration and washed with EtOAc. The resulting crystals are dried at 60 ° C. under reduced pressure to give 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-]. b] Pyridine-5-carboxamide monohydrobromide crystal (Form A36, 625 mg) was obtained.

[0042]
Example 5
(B11-type crystal of hydrobromide monohydrate)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2 , 3-b] Pyridine-5-carboxamide (5.0 g), EtOH (48 mL), water (12 mL), 48% hydrobromic acid (2.58 g) were sequentially charged at room temperature. After charging, the mixture was heated to an internal temperature of 70 ° C. or higher and stirred. After confirming that the solution had completely dissolved, the solution was subjected to clear filtration at an internal temperature of 70 ° C. or higher, and washed with warm EtOH (15 mL). 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide odor at an internal temperature of about 35 ° C. Hydrochloric acid salt seed crystals (A87, 49.0 mg) were added, and the mixture was aged with an internal temperature of 30 to 40 ° C. for 4 hours. Subsequently, the mixture was cooled to an internal temperature of 20 to 30 ° C., and aged and stirred overnight at the same temperature. At an internal temperature of 20-25 ° C., EtOAc (150 mL) was added dropwise over 1 hour, and the mixture was aged and stirred at the same temperature for 30 minutes or longer. The precipitated crystals were collected by filtration. The wet crystals were washed with a solution of EtOH / EtOAc (5 mL / 10 mL). The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide monohydrate crystal (Form B11, 5.24 g) was obtained.

[0043]
Example 6
(B21-type crystal of hydrobromide dihydrate)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2 , 3-b] Pyridine-5-carboxamide (5.0 g), EtOH (18 mL), water (12 mL), 48% hydrobromic acid (2.58 g) were sequentially charged at room temperature. After charging, the mixture was heated to an internal temperature of 60 ° C. or higher and stirred. After confirming that the solution was completely dissolved, the solution was subjected to clear filtration at an internal temperature of 60 ° C. or higher, and washed with warm EtOH (10 mL). The mixture was cooled to an internal temperature of about 45 to 50 ° C. and aged for 2 hours while stirring. Subsequently, the reaction solution is cooled to an internal temperature of 20 to 30 ° C., and aged at the same temperature and stirred overnight. At an internal temperature of 20 to 30 ° C., EtOAc (160 mL) was added dropwise over 1 hour, and the mixture was aged and stirred for 1 hour or more at the same temperature. The precipitated crystals were filtered. The wet crystals were washed with a solution of EtOH / EtOAc (3 mL / 12 mL). The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide dihydrate crystals (Form B21, 6.05 g) were obtained.

[0044]
Example 7
(Tautomerism of each crystal)

[0045]
Example 7-1
(Crystal form conversion; hydrobromide B21 → A61)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H- Pyrrolo [2,3-b] pyridine-5-carboxamide hydrobromide dihydrate (Form B21, 300 mg) and EtOH (3 mL) were sequentially charged at room temperature and suspended overnight. After suspension, the crystals were collected by filtration at room temperature and the wet crystals were washed with EtOH. The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide crystal (Form A61, 258 mg) was obtained.

[0046]
Example 7-2
(Crystal form conversion; hydrobromide B11 → B21)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H- Pyrrolo [2,3-b] pyridine-5-carboxamide hydrobromide monohydrate (form B11, 2.0 g), EtOH (7 mL), water (3 mL) were sequentially charged at room temperature and suspended overnight. It became cloudy. After suspension, the crystals were collected by filtration at room temperature and the wet crystals were washed with 70% aqueous EtOH. The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide dihydrate crystals (Form B21, 1.54 g) were obtained.

[0047]
Example 7-3
(Crystal form conversion; hydrobromide A61 form → B21 form)
4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H- Pyrrolo [2,3-b] pyridine-5-carboxamide hydrobromide (form A61, 1.0 g), EtOH (3.5 mL) and water (1.5 mL) were sequentially charged at room temperature and suspended overnight. After suspension, the crystals were filtered at room temperature and the wet crystals were washed with 70% aqueous EtOH. The wet crystals are dried overnight under vacuum and 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2,3-b] pyridine-5-carboxamide Hydrobromide dihydrate crystals (Form B21, 827 mg) were obtained.

[0048]
Reference Example 1
( Example of Preparation of Monohydrate Crystalline Compound (I) Free Form)
4-Chloro-1H-pyrrolo [2,3-b] pyridine-5-carboxamide (44.5 g) under nitrogen atmosphere 1s, 3R, 4s, 5S) -4-aminoadamantan-1-ol (57.0 g) and tributylamine (162.6 mL) were charged in NMP (222.5 mL), and heated and stirred at a bath temperature of 200 ° C. for 2.5 hours. The reaction solution was allowed to cool, and then the reaction solution was added dropwise while stirring in water / Et 2 O (6 L / 0.5 L), followed by stirring for 30 minutes. The obtained solid was collected by filtration, washed twice with water (400 mL), washed twice with Et 2 O (300 mL), and dried. The resulting solid was warmed to dissolve in MeOH (1.8 L) and filtered hot. The resulting mother liquor was concentrated under reduced pressure and MeOH (1.8 L) was added to the residue and heated to dissolve. The resulting solution was allowed to cool and stir, and then stirred at room temperature and aged overnight. The precipitated solid was collected by filtration, washed with EtOH and dried under reduced pressure. The resulting solid was suspended in EtOH (250 mL) and stirred at room temperature for 1 h. The solid was collected by filtration, washed with EtOH and dried under reduced pressure. The obtained solid was suspended in water (900 mL) and stirred at a bath temperature of 70 ° C. for 2 hours. The solid was collected by filtration, washed with water and dried under reduced pressure. Furthermore, the solid was suspended in water (900 mL) and stirred at a bath temperature of 70 ° C. for 2 hours. The solid is collected by filtration, washed with water and then dried under reduced pressure to give 4-{[(1R, 2s, 3S, 5s, 7s) -5-hydroxy-2-adamantyl] amino} -1H-pyrrolo [2 , 3-b] Pyridine-5-carboxamide monohydrate crystal (Form A01, 44 g) was obtained.

REFERENCES

1: D’Amico F, Fiorino G, Furfaro F, Allocca M, Danese S. Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials. Expert Opin Investig Drugs. 2018 Jul;27(7):595-599. doi: 10.1080/13543784.2018.1492547. Epub 2018 Jul 6. Review. PubMed PMID: 29938545.

2: Sands BE, Sandborn WJ, Feagan BG, Lichtenstein GR, Zhang H, Strauss R, Szapary P, Johanns J, Panes J, Vermeire S, O’Brien CD, Yang Z, Bertelsen K, Marano C; Peficitinib-UC Study Group. Peficitinib, an Oral Janus Kinase Inhibitor, in Moderate-to-Severe Ulcerative Colitis: Results From a Randomized, Phase 2 Study. J Crohns Colitis. 2018 Jun 15. doi: 10.1093/ecco-jcc/jjy085. [Epub ahead of print] PubMed PMID: 29917064.

3: Veale DJ, McGonagle D, McInnes IB, Krueger JG, Ritchlin CT, Elewaut D, Kanik KS, Hendrikx T, Berstein G, Hodge J, Telliez JB. The rationale for Janus kinase inhibitors for the treatment of spondyloarthritis. Rheumatology (Oxford). 2018 Apr 3. doi: 10.1093/rheumatology/key070. [Epub ahead of print] PubMed PMID: 29618084.

4: Cline A, Cardwell LA, Feldman SR. Advances in treating psoriasis in the elderly with small molecule inhibitors. Expert Opin Pharmacother. 2017 Dec;18(18):1965-1973. doi: 10.1080/14656566.2017.1409205. Epub 2017 Nov 27. Review. PubMed PMID: 29171774.

5: Baker KF, Isaacs JD. Novel therapies for immune-mediated inflammatory diseases: What can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn’s disease and ulcerative colitis? Ann Rheum Dis. 2018 Feb;77(2):175-187. doi: 10.1136/annrheumdis-2017-211555. Epub 2017 Aug 1. Review. PubMed PMID: 28765121.

6: Zhu T, Howieson C, Wojtkowski T, Garg JP, Han D, Fisniku O, Keirns J. The Effect of Verapamil, a P-Glycoprotein Inhibitor, on the Pharmacokinetics of Peficitinib, an Orally Administered, Once-Daily JAK Inhibitor. Clin Pharmacol Drug Dev. 2017 Nov;6(6):548-555. doi: 10.1002/cpdd.344. Epub 2017 Mar 16. PubMed PMID: 28301084.

7: Genovese MC, Greenwald M, Codding C, Zubrzycka-Sienkiewicz A, Kivitz AJ, Wang A, Shay K, Wang X, Garg JP, Cardiel MH. Peficitinib, a JAK Inhibitor, in Combination With Limited Conventional Synthetic Disease-Modifying Antirheumatic Drugs in the Treatment of Moderate-to-Severe Rheumatoid Arthritis. Arthritis Rheumatol. 2017 May;69(5):932-942. doi: 10.1002/art.40054. PubMed PMID: 28118538.

8: Ito M, Yamazaki S, Yamagami K, Kuno M, Morita Y, Okuma K, Nakamura K, Chida N, Inami M, Inoue T, Shirakami S, Higashi Y. A novel JAK inhibitor, peficitinib, demonstrates potent efficacy in a rat adjuvant-induced arthritis model. J Pharmacol Sci. 2017 Jan;133(1):25-33. doi: 10.1016/j.jphs.2016.12.001. Epub 2016 Dec 23. PubMed PMID: 28117214.

9: Zhu T, Parker B, Wojtkowski T, Nishimura T, Garg JP, Han D, Fisniku O, Keirns J. Drug Interactions Between Peficitinib, an Orally Administered, Once-Daily Janus Kinase Inhibitor, and Rosuvastatin in Healthy Subjects. Clin Pharmacokinet. 2017 Jul;56(7):747-757. doi: 10.1007/s40262-016-0474-4. PubMed PMID: 27878567.

10: Semerano L, Decker P, Clavel G, Boissier MC. Developments with investigational Janus kinase inhibitors for rheumatoid arthritis. Expert Opin Investig Drugs. 2016 Dec;25(12):1355-1359. Epub 2016 Oct 31. PubMed PMID: 27748152.

11: Kivitz AJ, Gutierrez-Ureña SR, Poiley J, Genovese MC, Kristy R, Shay K, Wang X, Garg JP, Zubrzycka-Sienkiewicz A. Peficitinib, a JAK Inhibitor, in the Treatment of Moderate-to-Severe Rheumatoid Arthritis in Patients With an Inadequate Response to Methotrexate. Arthritis Rheumatol. 2017 Apr;69(4):709-719. doi: 10.1002/art.39955. PubMed PMID: 27748083.

12: Lam S. JAK inhibitors: A broadening approach in rheumatoid arthritis. Drugs Today (Barc). 2016 Aug;52(8):467-469. PubMed PMID: 27722215.

13: Roskoski R Jr. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol Res. 2016 Sep;111:784-803. doi: 10.1016/j.phrs.2016.07.038. Epub 2016 Jul 26. Review. PubMed PMID: 27473820.

14: Iwata S, Tanaka Y. Progress in understanding the safety and efficacy of Janus kinase inhibitors for treatment of rheumatoid arthritis. Expert Rev Clin Immunol. 2016 Oct;12(10):1047-57. doi: 10.1080/1744666X.2016.1189826. Epub 2016 Jun 6. Review. PubMed PMID: 27253519.

15: Cao YJ, Sawamoto T, Valluri U, Cho K, Lewand M, Swan S, Lasseter K, Matson M, Holman J Jr, Keirns J, Zhu T. Pharmacokinetics, Pharmacodynamics, and Safety of ASP015K (Peficitinib), a New Janus Kinase Inhibitor, in Healthy Subjects. Clin Pharmacol Drug Dev. 2016 Nov;5(6):435-449. doi: 10.1002/cpdd.273. Epub 2016 Jun 30. PubMed PMID: 27162173.

16: Nielsen OH, Seidelin JB, Ainsworth M, Coskun M. Will novel oral formulations change the management of inflammatory bowel disease? Expert Opin Investig Drugs. 2016 Jun;25(6):709-18. doi: 10.1517/13543784.2016.1165204. Epub 2016 Mar 28. Review. PubMed PMID: 26967267.

17: Yiu ZZ, Warren RB. Novel Oral Therapies for Psoriasis and Psoriatic Arthritis. Am J Clin Dermatol. 2016 Jun;17(3):191-200. doi: 10.1007/s40257-016-0179-3. Review. PubMed PMID: 26923915.

18: Takeuchi T, Tanaka Y, Iwasaki M, Ishikura H, Saeki S, Kaneko Y. Efficacy and safety of the oral Janus kinase inhibitor peficitinib (ASP015K) monotherapy in patients with moderate to severe rheumatoid arthritis in Japan: a 12-week, randomised, double-blind, placebo-controlled phase IIb study. Ann Rheum Dis. 2016 Jun;75(6):1057-64. doi: 10.1136/annrheumdis-2015-208279. Epub 2015 Dec 15. PubMed PMID: 26672064; PubMed Central PMCID: PMC4893099.

19: Oda K, Cao YJ, Sawamoto T, Nakada N, Fisniku O, Nagasaka Y, Sohda KY. Human mass balance, metabolite profile and identification of metabolic enzymes of [¹⁴C]ASP015K, a novel oral janus kinase inhibitor. Xenobiotica. 2015;45(10):887-902. doi: 10.3109/00498254.2015.1026864. Epub 2015 May 19. PubMed PMID: 25986538.

20: Nakada N, Oda K. Identification and characterization of metabolites of ASP015K, a novel oral Janus kinase inhibitor, in rats, chimeric mice with humanized liver, and humans. Xenobiotica. 2015;45(9):757-65. doi: 10.3109/00498254.2015.1019594. Epub 2015 Jun 12. PubMed PMID: 25869242.

/////////////////Peficitinib hydrobromide, Smyraf, JAPAN 2019, ペフィシチニブ臭化水素酸塩  , ASP015K, Rheumatoid Arthritis

O=C(C1=CN=C(NC=C2)C2=C1N[C@@H]3[C@]4([H])C[C@@]5([H])C[C@](C4)(O)C[C@]3([H])C5)N.[H]Br

BMS 986142


Image result for BMS-986142

img

BMS-986142

(2S,5R,3S)-6-fluoro-5-(3-(8-fluoro-1-methyl-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)-2-methylphenyl)-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-1H-carbazole-8-carboxamide

6-Fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide

Molecular Formula, C32-H30-F2-N4-O4, Molecular Weight, 572.609, RN: 1643368-58-4
UNII: PJX9GH268R

  • Originator Bristol-Myers Squibb
  • Class Anti-inflammatories; Antirheumatics; Small molecules
  • Mechanism of Action Agammaglobulinaemia tyrosine kinase inhibitors
  • Phase II Rheumatoid arthritis; Sjogren’s syndrome
  • 24 Jun 2018 Biomarkers information updated
  • 07 Jun 2018 Bristol-Myers Squibb completes a phase II trial in Rheumatoid arthritis (Treatment-experienced) in Argentina, Austria, Belgium, Brazil, Canada, Chile, Colombia, Czech Republic, France, Germany, Israel, Italy, Japan, Mexico, Netherlands, Poland, Russia, South Africa, South Korea, Spain, Taiwan, USA (PO) (NCT02638948) (EudraCT2015-002887-17)
  • 01 Oct 2016 Phase-II clinical trials in Sjogren’s syndrome in Puerto Rico (PO) (NCT02843659) after October 2016
  •  phase II clinical development at Bristol-Myers Squibb for the treatment of patients with moderate to severe rheumatoid arthritis and for the treatment of moderate to severe primary Sjogren’s syndrome.

BMS-986142 is a potent, selective, reversible BTK inhibitor. BMS-986142 shows BTK IC50 = 0.5nM; human WB IC50 = 90 nM. In molecule of BMS-986142, two atropisomeric centers were rotationally locked to provide a single, stable atropisomer, resulting in enhanced potency and selectivity as well as a reduction in safety liabilities. With significantly enhanced potency and selectivity, excellent in vivo properties and efficacy, and a very desirable tolerability and safety profile, BMS-986142 was advanced into clinical studies substituted tetrahydrocarbazole and 10 carbazole carboxamide compounds useful as kinase inhibitors, including the modulation of Bruton’s tyrosine kinase (Btk) and other Tec family kinases such as Itk. Provided herein are substituted tetrahydrocarbazole and carbazole carboxamide compounds, compositions comprising such compounds, and methods of their use. The invention further pertains to pharmaceutical compositions containing at least one compound 15 according to the invention that are useful for the treatment of conditions related to kinase modulation and methods of inhibiting the activity of kinases, including Btk and other Tec family kinases such as Itk, in a mammal. Protein kinases, the largest family of human enzymes, encompass well over 500 proteins. Btk is a member of the Tec family of tyrosine kinases, and is a regulator of 20 early B-cell development, as well as mature B-cell activation, signaling, and survival. B-cell signaling through the B-cell receptor (BCR) leads to a wide range of biological outputs, which in turn depend on the developmental stage of the B-cell. The magnitude and duration of BCR signals must be precisely regulated. Aberrant BCR- mediated signaling can cause disregulated B-cell activation and/or the formation of 25 pathogenic auto-antibodies leading to multiple autoimmune and/or inflammatory diseases. Mutation of Btk in humans results in X-linked agammaglobulinaemia (XLA). This disease is associated with the impaired maturation of B-cells, diminished immunoglobulin production, compromised T-cell-independent immune responses and marked attenuation of the sustained calcium signal upon BCR stimulation. 30 Evidence for the role of Btk in allergic disorders and/or autoimmune disease and/or inflammatory disease has been established in Btk-deficient mouse models. For example, in standard murine preclinical models of systemic lupus erythematosus (SLE), Btk deficiency has been shown to result in a marked amelioration of disease progression. Moreover, Btk deficient mice are also resistant to developing collagen-induced arthritis and are less susceptible to Staphylococcus-induced arthritis.

A large body of evidence supports the role of B-cells and the humoral immune system in the pathogenesis of autoimmune and/or inflammatory diseases. Protein-based therapeutics (such as RITUXAN®) developed to deplete B-cells, represent an important approach to the treatment of a number of autoimmune and/or inflammatory diseases. Because of Btk’s role in B-cell activation, inhibitors of Btk can be useful as inhibitors of B-cell mediated pathogenic activity (such as autoantibody production).

Btk is also expressed in mast cells and monocytes and has been shown to be important for the function of these cells. For example, Btk deficiency in mice is associated with impaired IgE-mediated mast cell activation (marked diminution of TNF-alpha and other inflammatory cytokine release), and Btk deficiency in humans is associated with greatly reduced TNF-alpha production by activated monocytes.

Thus, inhibition of Btk activity can be useful for the treatment of allergic disorders and/or autoimmune and/or inflammatory diseases including, but not limited to: SLE, rheumatoid arthritis, multiple vasculitides, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis, allergic rhinitis, multiple sclerosis (MS), transplant rejection, type I diabetes, membranous nephritis, inflammatory bowel disease, autoimmune hemolytic anemia, autoimmune thyroiditis, cold and warm agglutinin diseases, Evans syndrome, hemolytic uremic syndrome/thrombotic thrombocytopenic purpura (HUS/TTP), sarcoidosis, Sj5gren’s syndrome, peripheral neuropathies (e.g., Guillain-Barre syndrome), pemphigus vulgaris, and asthma. In addition, Btk has been reported to play a role in controlling B-cell survival in certain B-cell cancers. For example, Btk has been shown to be important for the survival of BCR-Abl-positive B-cell acute lymphoblastic leukemia cells. Thus inhibition of Btk activity can be useful for the treatment of B-cell lymphoma and leukemia. In view of the numerous conditions that are contemplated to benefit by treatment involving modulation of protein kinases, it is immediately apparent that new compounds capable of modulating protein kinases such as Btk and methods of using these compounds should provide substantial therapeutic benefits to a wide variety of patients.

U.S. Patent No. 8,084,620 and WO 2011/159857 disclose tricyclic carboxamide compounds useful as kinase inhibitors, including the modulation of Btk and other Tec family kinases. There still remains a need for compounds useful as Btk inhibitors and yet having selectivity over Jak2 tyrosine kinase. Further, there still remains a need for compounds useful as Btk inhibitors that have selectivity over Jak2 tyrosine kinase and also have improved potency in the whole blood BCR-stimulated CD69 expression assay. Applicants have found potent compounds that have activity as Btk inhibitors. Further, applicants have found compounds that have activity as Btk inhibitors and are selective over Jak2 tyrosine kinase. Further still, applicants have found compounds that have activity as Btk inhibitors, are selective over Jak2 tyrosine kinase, and have improved potency in the whole blood BCR-stimulated CD69 expression assay. These compounds are provided to be useful as pharmaceuticals with desirable stability, bioavailability, therapeutic index, and toxicity values that are important to their drugability.

SYN

CLIP

Adventures in Atropisomerism: A Case Study from BMS – Not a Real Doctor

Dennis Hu

Scheme 2. Highlights from optimization of the first intermediate with axial chirality.

Image result for BMS-986142

Image result for BMS-986142

CLIP

https://cen.acs.org/pharmaceuticals/drug-development/Giving-atropisomers-another-chance/96/i33

Image result for BMS-986142

Yet another atropisomeric kinase inhibitor, of Bruton’s tyrosine kinase (BTK), currently being evaluated in Phase II clinical trials for rheumatoid arthritis, comes from Bristol Myers-Squibb. BMS-986142 contains one point-chiral center and two atropisomeric chiral axes, making it a diastereomeric compound with eight possible isomers. The less stable atropisomeric axis has a half-life on the order of hours to days, which means it can’t be heated above about 45 °C without the compound morphing. To keep the molecule from racemizing, the team had to design its synthetic routes and analysis with a close eye on temperature.

During the discovery stage, BMS analytical chemist Jun Dai and the team developed methods to analyze the compounds’ isomers. She estimates that the researchers screened at least twice as many separation methods for atropisomers as they would have for normal chiral compounds because of the atropisomers’ potential for temperature-dependent conversion. “It was challenging but rewarding,” she says.

To determine the proportion of early atropisomers with half-lives of minutes to hours, the team ran high-performance liquid chromatography analysis at low temperature, chilling the column with ice or cooling equipment. Isolating some atropisomeric compounds required researchers to use ice-bath cooling during fraction collection and even solvent evaporation. The medicinal chemistry route to BMS-986142 required three chiral column purifications to obtain a single diastereomer with the best binding properties (J. Chromatogr. A 2017, DOI: 10.1016/j.chroma.2017.01.016).

Process synthesis, however, generally isn’t amenable to column chromatography steps, which can take weeks to months on a large scale. “To be honest, when I first saw it, I really wasn’t sure how we were going to make it,” says BMS chemist Thomas Razler, who led the process chemistry efforts to scale-up BMS-986142.

The researchers say extensive knowledge sharing between medicinal, analytical, and process teams about the atropisomeric compound was key to the program’s success. The process team took advantage of the fact that the diastereomeric forms of BMS-986142 had very different solubility profiles, enabling the chemists to replace all chiral chromatography with simpler crystallization steps and produce more than 200 kg of a single enantiomer and diastereomer (Org. Lett. 2018, DOI: 10.1021/acs.orglett.8b01218).

Although the final molecule is stable as a solid, the team says that in solution, the risk of racemization is higher. Citing ongoing work in that area of development, Razler declined to elaborate on how the molecule behaves in its formulation but notes the team hopes to publish that information next year. The atropisomerism is still an issue, he says, but a fascinating one.

Paper

Organic Letters, 20(13), 3736-3740; 2018

Adventures in Atropisomerism: Total Synthesis of a Complex Active Pharmaceutical Ingredient with Two Chirality Axes

Chemical & Synthetic DevelopmentBristol-Myers Squibb Company1 Squibb Drive, New Brunswick, New Jersey 08901, United States
Org. Lett.201820 (13), pp 3736–3740
DOI: 10.1021/acs.orglett.8b01218
Abstract Image

A strategy to prepare compounds with multiple chirality axes, which has led to a concise total synthesis of compound 1A with complete stereocontrol, is reported.

Figure

Figure

https://pubs.acs.org/doi/suppl/10.1021/acs.orglett.8b01218/suppl_file/ol8b01218_si_001.pdf

(2S,5R)-6-fluoro-5-(3-(8-fluoro-1-methyl-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)-2-methylphenyl)-2-(2-hydroxypropan-2-yl)-2,3,4,9- tetrahydro-1H-carbazole-8-carboxamide (1A).

1H NMR (500 MHz, DMSO-d6) 10.78 (s, 1H), 8.07 (br. s., 1H), 7.95 (d, J=7.8 Hz, 1H), 7.72 (dd, J=14.2, 8.0 Hz, 1H), 7.56 (d, J=10.8 Hz, 1H), 7.45 (br. s., 1H), 7.42 – 7.36 (m, 1H), 7.34 (d, J=6.9 Hz, 1H), 7.34 – 7.31 (m, 1H), 7.29 (dd, J=7.5, 1.3 Hz, 1H), 4.17 (s, 1H), 3.73 (d, J=8.0 Hz, 3H), 2.91 (dd, J=16.8, 4.4 Hz, 1H), 2.48 – 2.37 (m, 1H), 1.98 – 1.89 (m, 2H), 1.87 (d, J=11.0 Hz, 1H), 1.76 (s, 3H), 1.59 (td, J=11.5, 4.1 Hz, 1H), 1.20 – 1.12 (m, 1H), 1.11 (s, 6H). 13C NMR (125.8 MHz, DMSO-d6) 168.2 (d, J=1.8 Hz, 1C), 160.1 (d, J=3.6 Hz, 1C), 151.9 (d, J=228.9 Hz, 1C), 150.5 (d, J=41.8 Hz, 1C), 148.7 (d, J=205.3 Hz, 1C), 139.2, 135.1, 135.0, 134.8, 131.4, 130.6, 130.0 (d, J=7.3 Hz, 1C), 128.5, 127.1 (d, J=4.5 Hz, 1C), 125.7, 124.3 (d, J=2.7 Hz, 1C), 123.6 (d, J=8.2 Hz, 1C), 123.0 (d, J=23.6 Hz, 1C), 120.8 (d, J=20.0 Hz, 1C), 118.4, 115.3 (d, J=7.3 Hz, 1C), 108.8 (d, J=5.4 Hz, 1C), 106.7 (d, J=28.2 Hz, 1C), 70.4, 45.4, 34.3 (d, J=14.5 Hz, 1C), 27.1, 26.8, 24.8, 24.7, 22.1, 14.5. mp 222-225 °C. IR (neat) 3487, 3418, 3375, 2967, 1651, 1394, 756 cm-1; HRMS (ESI) m/z: calcd for C32H30F2N4O4 [M+H]+ 573.2308, found 573.2312.

Chiral HPLC Analysis: Gradient: Complex Start % B: 0 7 Min. 55% 11 Min. 55% 14 Min. 100% Stop Time: 17 min Flow Rate: 1.5 ml/min Wavelength1: 225 Wavelength2: 256 Solvent Pair: S194/S195 (TFA) Solvent A: A1=0.05%TFA Water:ACN (95:5) S194 Solvent B: B1=0.05%TFA Water:ACN (5:95) S195 Column 1 : 1: Chiralcel OX-3R 3um 4.6 x 150 mm SN = OX3RCD-TE001 Oven Temperature: 50

Clip

Adventures in Atropisomerism: Development of a Robust, Diastereoselective, Lithium-Catalyzed Atropisomer-Forming Active Pharmaceutical Ingredient Step

Chemical and Synthetic DevelopmentBristol-Myers Squibb CompanyOne Squibb Drive, New Brunswick, New Jersey08903, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00246
Abstract Image

The final step in the route to BMS-986142, a reversible inhibitor of the BTK enzyme, involves the diastereoselective construction of a chiral axis during the base-mediated cyclization of the quinazolinedione fragment. Optimization of the reaction to minimize formation of the undesired atropisomer led to the discovery that the amount of base and nature of the counterion play a vital role in the diastereoselectivity of the reaction. The highest diastereoselectivities were observed with a catalytic amount of LiOt-Bu. Development of a crystallization to selectively purge the undesired atropisomer is reported. Interestingly, ripening of the crystalline API was observed and further investigated, leading to a significant increase in the purity of the active pharmaceutical ingredient.

(2S,5R)-6-fluoro-5-(3-(8-fluoro-1-methyl-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)-2-methylphenyl)-2-(2-hydroxypropan-2-yl)-2,3,4,9- tetrahydro-1H-carbazole-8-carboxamide 1A

white crystalline solid (80.52g, 6 wt % MeOH, 89.4% corrected yield).

1H NMR (500 MHz, DMSO-d6) 10.78 (s, 1H), 8.07 (br. s., 1H), 7.95 (d, J=7.8 Hz, 1H), 7.72 (dd, J=14.2, 8.0 Hz, 1H), 7.56 (d, J=10.8 Hz, 1H), 7.45 (br. s., 1H), 7.42 – 7.36 (m, 1H), 7.34 (d, J=6.9 Hz, 1H), 7.34 – 7.31 (m, 1H), 7.29 (dd, J=7.5, 1.3 Hz, 1H), 4.17 (s, 1H), 3.73 (d, J=8.0 Hz, 3H), 2.91 (dd, J=16.8, 4.4 Hz, 1H), 2.48 – 2.37 (m, 1H), 1.98 – 1.89 (m, 2H), 1.87 (d, J=11.0 Hz, 1H), 1.76 (s, 3H), 1.59 (td, J=11.5, 4.1 Hz, 1H), 1.20 – 1.12 (m, 1H), 1.11 (s, 6H).

13C NMR (125.8 MHz, DMSO-d6) 168.2 (d, J=1.8 Hz, 1C), 160.1 (d, J=3.6 Hz, 1C), 151.9 (d, J=228.9 Hz, 1C), 150.5 (d, J=41.8 Hz, 1C), 148.7 (d, J=205.3 Hz, 1C), 139.2, 135.1, 135.0, 134.8, 131.4, 130.6, 130.0 (d, J=7.3 Hz, 1C), 128.5, 127.1 (d, J=4.5 Hz, 1C), 125.7, 124.3 (d, J=2.7 Hz, 1C), 123.6 (d, J=8.2 Hz, 1C), 123.0 (d, J=23.6 Hz, 1C), 120.8 (d, J=20.0 Hz, 1C), 118.4, 115.3 (d, J=7.3 Hz, 1C), 108.8 (d, J=5.4 Hz, 1C), 106.7 (d, J=28.2 Hz, 1C), 70.4, 45.4, 34.3 (d, J=14.5 Hz, 1C), 27.1, 26.8, 24.8, 24.7, 22.1, 14.5.

mp 222-225 °C.

IR (neat) 3487, 3418, 3375, 2967, 1651, 1394, 756 cm-1;

HRMS (ESI) m/z: calcd for C32H30F2N4O4 [M+H]+ 573.2308, found 573.2312.

Chiral HPLC Analysis: Gradient: Complex Start % B: 0 7 Min. 55% 11 Min. 55% 14 Min. 100% Stop Time: 17 min Flow Rate: 1.5 ml/min Wavelength1: 225 Wavelength2: 256 Solvent Pair: S194/S195 (TFA) Solvent A: A1=0.05%TFA Water:ACN (95:5) S194 Solvent B: B1=0.05%TFA Water:ACN (5:95) S195 Column 1 : 1: Chiralcel OX-3R 3um 4.6 x 150 mm SN = OX3RCD-TE001 Oven Temperature: 50…..https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.8b00246/suppl_file/op8b00246_si_001.pdf

PAPER

Discovery of 6-Fluoro-5-(R)-(3-(S)-(8-fluoro-1-methyl-2,4-dioxo-1,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-1H-carbazole-8-carboxamide (BMS-986142): A Reversible Inhibitor of Bruton’s Tyrosine Kinase (BTK) Conformationally Constrained by Two Locked Atropisomers

Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
J. Med. Chem.201659 (19), pp 9173–9200
DOI: 10.1021/acs.jmedchem.6b01088
Publication Date (Web): September 1, 2016
Copyright © 2016 American Chemical Society
*Phone: 609-252-6778. E-mail: scott.watterson@bms.com.
Abstract Image

Bruton’s tyrosine kinase (BTK), a nonreceptor tyrosine kinase, is a member of the Tec family of kinases. BTK plays an essential role in B cell receptor (BCR)-mediated signaling as well as Fcγ receptor signaling in monocytes and Fcε receptor signaling in mast cells and basophils, all of which have been implicated in the pathophysiology of autoimmune disease. As a result, inhibition of BTK is anticipated to provide an effective strategy for the clinical treatment of autoimmune diseases such as lupus and rheumatoid arthritis. This article details the structure–activity relationships (SAR) leading to a novel series of highly potent and selective carbazole and tetrahydrocarbazole based, reversible inhibitors of BTK. Of particular interest is that two atropisomeric centers were rotationally locked to provide a single, stable atropisomer, resulting in enhanced potency and selectivity as well as a reduction in safety liabilities. With significantly enhanced potency and selectivity, excellent in vivo properties and efficacy, and a very desirable tolerability and safety profile, 14f (BMS-986142) was advanced into clinical studies.

HPLC purity: 99.9%; tr = 11.05 min (Method A); 99.9%; tr = 10.72 min (Method B). Chiral purity: 99.8% ie;

Optical rotation: [α]D20 (c = 2.10, CHCl3) = +63.8°;

LCMS (ESI) m/z calcd for C32H30F2N4O4 [M + H]+ 573.2. Found: 573.5. Anal. calcd for C32H30F2N4O4, 0.72% H2O: C 65.56, H 5.42, N 9.55. Found: C 65.69, H 5.40, N 9.52.

 1H NMR (500 MHz, DMSO-d6) δ 10.78 (s, 1H), 8.07 (br. s., 1H), 7.95 (d, J = 7.8 Hz, 1H), 7.72 (dd, J = 14.2, 8.0 Hz, 1H), 7.56 (d, J = 10.8 Hz, 1H), 7.45 (br. s., 1H), 7.42–7.36 (m, 1H), 7.34 (d, J = 6.9 Hz, 1H), 7.34–7.31 (m, 1H), 7.29 (dd, J = 7.5, 1.3 Hz, 1H), 4.17 (s, 1H), 3.73 (d, J = 8.0 Hz, 3H), 2.91 (dd, J = 16.8, 4.4 Hz, 1H), 2.48–2.37 (m, 1H), 1.98–1.89 (m, 2H), 1.87 (d, J = 11.0 Hz, 1H), 1.76 (s, 3H), 1.59 (td, J = 11.5, 4.1 Hz, 1H), 1.20–1.12 (m, 1H), and 1.11 (s, 6H). 1

3C NMR (126 MHz, DMSO-d6) δ 168.2 (d, J = 1.8 Hz, 1C), 160.1 (d, J = 3.6 Hz, 1C), 151.9 (d, J = 228.9 Hz, 1C), 150.5 (d, J = 41.8 Hz, 1C), 148.7 (d, J= 205.3 Hz, 1C), 139.2, 135.1, 135.0, 134.8, 131.4, 130.6, 130.0 (d, J = 7.3 Hz, 1C), 128.5, 127.1 (d, J = 4.5 Hz, 1C), 125.7, 124.3 (d, J = 2.7 Hz, 1C), 123.6 (d, J = 8.2 Hz, 1C), 123.0 (d, J = 23.6 Hz, 1C), 120.8 (d, J = 20.0 Hz, 1C), 118.4, 115.3 (d, J = 7.3 Hz, 1C), 108.8 (d, J = 5.4 Hz, 1C), 106.7 (d, J = 28.2 Hz, 1C), 70.4, 45.4, 34.3 (d, J = 14.5 Hz, 1C), 27.1, 26.8, 24.8, 24.7, 22.1, and 14.5. 

19F-NMR (470 MHz, DMSO-d6) δ −121.49 (dt, J = 22.9, 11.4 Hz, 1F), and −129.56 (d, J = 11.4 Hz, 1F).

PATENT

WO 2014210085

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=850E1F706BE58D54C2B9AEE37AE6831C.wapp2nC?docId=WO2014210085&tab=PCTDESCRIPTION&queryString=EN_ALL%3Anmr+AND+PA%3A%28Bristol-Myers+Squibb%29+&recNum=19&maxRec=4726

Atropisomers are stereoisomers resulting from hindered rotation about a single bond axis where the rotational barrier is high enough to allow for the isolation of the individual rotational isomers. (LaPlante et al., J. Med. Chem., 54:7005-7022 (2011).)

Th compounds of Formula (A):

have two stereogenic axes: bond (a) between the tricyclic tetrahydrocarbazole/carbazole group and the phenyl group; and bond (b) between the asymmetric heterocyclic dione group Q and the phenyl group. Due to the non-symmetric nature of the substitutions on the rings connected by the single bonds labeled a and b, and due to limited rotation about these bonds caused by steric hindrance, the compounds of Formula (A) can form rotational isomers. If the rotational energy barriers are sufficiently high, hindered rotations about bond (a) and/or bond (b) occur at rates that are slow enough to allow isolation of the separated atropisomers as different compounds. Thus, the compounds of Formula (A) can form four rotational isomers, which under certain conditions, such as chromatography on a chiral stationary phase, can be separated into individual atropisomers. In solution, the compounds of Formula (A) can be provided as a mixture of four diastereomers, or mixtures of two pairs of diastereomers, or single atropisomers.

For the compounds of Formula (A), the pair of rotational isomers formed by hindered rotation about stereogenic axis (a) can be represented by the compounds of Formula (I) and Formula (B) having the structures:

The compounds of Formula (I) and the compounds of Formula (B) were found to be separable and stable in solution at ambient and physiological temperatures. Additionally, rotational isomers are formed by hindered rotation about stereogenic axis (b). These two atropisomers of the compounds of Formula (I) were also found to be separable and stable in solution at ambient and physiological temperatures.

Chiral compounds, such as the compounds of Formula (A), can be separated by various techniques including Supercritical Fluid Chromatography (SFC). SFC, which is form of normal phase HPLC, is a separation technique that uses super/subcritical fluid CO2 and polar organic modifiers such as alcohols as mobile phases. (White et al, J. Chromatography A, 1074: 175-185 (2005).

Example 28

6-Fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide (single atropisomer)


(28)

Following the procedure used to prepare Example 27, (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro- lH-carbazole-8-carboxamide (single enantiomer) [Intermediate 26] (0.045 g, 0.122 mmol) and 8-fluoro-l-methyl-3-(S)-(2-methyl-3-(4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)quinazoline-2,4(lH,3H)-dione

[Intermediate 10] (0.065 g, 0.158 mmol) were converted into 6-fluoro-5-(3-(S)-(8-fluoro-1 -methyl-2,4-dioxo- 1 ,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-

hydroxypropan-2-yl)-2,3,4,9-tetrahydro- lH-carbazole-8-carboxamide (mixture of two atropisomers) as a yellow solid (0.035 g, 49% yield). Separation of a sample of this material by chiral super-critical fluid chromatography, using the conditions used to separate Example 27, provided (as the first peak to elute from the column) 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide. The chiral purity was determined to be greater than 99.5%. The relative and absolute configurations were determined by x-ray crystallography. Mass spectrum m/z 573 (M+H)+XH NMR (500 MHz, DMSO-d6) δ 10.77 (s, 1H), 8.05 (br. s., 1H), 7.94 (dd, J=7.9, 1.2 Hz, 1H), 7.56-7.52 (m, 1H), 7.43 (br. s., 1H), 7.40-7.36 (m, 1H), 7.35-7.30 (m, 2H), 7.28 (dd, J=7.5, 1.4 Hz, 1H), 4.15 (s, 1H), 3.75-3.70 (m, 3H), 2.90 (dd, J=16.8, 4.6 Hz, 1H), 2.47-2.39 (m, 1H), 1.93-1.82 (m, 3H), 1.74 (s, 3H), 1.57 (td, J=1 1.7, 4.2 Hz, 1H), 1.16-1.11 (m, 1H), and 1.10 (d, J=1.9 Hz, 6H). [a]D: +63.8° (c 2.1, CHC13). DSC melting point onset temperature = 202.9 °C (heating rate = 10 °C/min.).

The absolute configuration of Example 28 was confirmed by single crystal x-ray analysis of crystals prepared by dissolving the compound in excess methanol and slowly evaporating the solvent at room temperature to provide a di-methanol solvate (crystalline form M2-1). Unit cell dimensions: a = 9.24 A, b = 7.97 A, c = 22.12 A, a = 90.0°, β = 94.1°, γ = 90.0°; Space group: P2i; Molecules of Example 28/asymmetric unit: 1 ;

Volume/Number of molecules in the unit cell = 813 A3; Density (calculated) = 1.301 g/cm3. Fractional atomic coordinates at 173 K are given in Table 6, and a depiction of the structure is given in Figure 5.

Alternative Synthesis of Example 28:

A mixture of (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide [Intermediate 1 1] (5.00 g, 13.54 mmol), 8-fluoro-l-methyl-3-(S)-(2-methyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)quinazoline-2,4(lH,3H)-dione [Intermediate 10] (6.67 g, 16.25 mmol), tripotassium phosphate (2 M in water) (20.31 mL, 40.6 mmol), and tetrahydrofuran (25 mL) was subjected to 3 evacuate-fill cycles with nitrogen. The mixture was treated with l, l’-bis(di-/er/-butylphosphino)ferrocene palladium dichloride (0.441 g, 0.677 mmol) and the mixture was subjected to 2 more evacuate- fill cycles with nitrogen. The mixture was stirred at room temperature overnight, then was diluted with EtOAc, washed sequentially with water and brine, and dried and concentrated. The residue was purified by column chromatography on silica gel, eluting with EtOAc-hexanes (sequentially 50%, 62%, 75% and 85%), to provide 6-fluoro-5-(3-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3-(S)-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide as a white solid (6.58 g, 85% yield).

Material prepared by this method (40.03 g, 69.9 mmol) was separated by chiral super-critical fluid chromatography to give (2S, 5R)-6-fluoro-5-(3-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide. Further purification was achieved by suspending this material in methanol, sonicating for 5 min, collection of the solid by filtration, rinsing the collected solid with methanol and drying at room temperature under reduced pressure to give a white solid (22.0 g, 90% yield).

2R ANALOGUE

Example 27

6-Fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(R)-(2-hydroxypropan-2-yl)-2,3 ,4,9-tetrahydro- 1 H-carbazole-8- carboxamide (single atropisomer)

Preparation 27A: 6-Fluoro-5-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(R)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (mixture of 2 atropisomers)

A mixture of (R)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (single enantiomer) [Intermediate 25] (5.00 g, 13.5 mmol), 8-fluoro-l-methyl-3-(S)-(2-methyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl) quinazoline-2,4(lH,3H)-dione [Intermediate 10] (6.94 g, 16.9 mmol), 2 M aqueous K3PO4 (20.3 mL, 40.6 mmol) and THF (60 mL) was subjected to three evacuate-fill cycles with nitrogen. The mixture was treated with 1 , l’-bis(di-tert-butylphosphino) ferrocene palladium(II) chloride (441 mg, 677 μιηοΐ) and subjected to two more evacuate-fill cycles with nitrogen. The mixture was stirred at room temperature overnight. The mixture was diluted with EtOAc, washed sequentially with water and brine, and dried and concentrated. The residue was purified by column chromatography on silica gel, eluting with EtOAc-hexanes (sequentially 50%, 62%, 75% and 85%), to give 6-fluoro-5-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(R)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (mixture of two atropisomers) as an off-white solid (6.77 g, 87% yield). Mass spectrum m/z 573 (M+H)+. ¾ NMR (500 MHz, DMSO-d6) δ 10.79-10.74 (m, 1H), 8.05 (br. s., 1H), 7.98-7.93 (m, 1H), 7.76-7.69 (m, 1H), 7.57-7.51 (m, 1H), 7.43 (br. s., 1H), 7.40-7.26 (m, 4H), 4.19-4.13 (m, 1H), 3.74-3.68 (m, 3H), 2.94-2.84 (m, 1H), 2.49-2.35 (m, 2H), 1.92-1.80 (m, 3H), 1.76-1.68 (m, 3H), 1.62-1.52 (m, 1H), and 1.12-1.06 (m, 6H).

Example 27:

A sample of 6-fluoro-5-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(R)-(2-hydroxypropan-2-yl)-2, 3,4,9-tetrahydro-lH-carbazole-8-carboxamide (mixture of two atropisomers) was separated by chiral super-critical fluid chromatography as follows: column: CHIRALPAK® AS-H (3 x 25 cm, 5 μιη); Mobile Phase: C02-MeOH (70:30) at 120 mL/min, 35 °C, 100 bar; sample preparation: 9 mg/mL in MeOH; injection: 1.7 mL. The first peak eluting from the column provided 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(R)-(2 -hydroxypropan-2-yl)-2, 3,4,9-tetrahydro-lH-carbazole-8-carboxamide. The chiral purity was determined to be greater than 99.5%. Mass spectrum m/z 573 (M+H)+XH NMR (500 MHz, DMSO-d6) δ 10.76 (s, 1H), 8.05 (br. s., 1H), 7.96 (d, J=7.8 Hz, 1H), 7.72 (ddd, J=14.3, 8.0, 1.2 Hz, 1H), 7.55 (d, J=10.8 Hz, 1H), 7.44 (br. s., 1H), 7.40-7.36 (m, 1H), 7.35-7.28 (m, 3H), 4.18 (s, 1H), 3.72

PATENT

WO 2018118830

https://patentscope.wipo.int/search/de/detail.jsf?docId=WO2018118830&tab=PCTDESCRIPTION&office=&prevFilter=%26fq%3DICF_M%3A%22C07D%22%26fq%3DPAF_M%3A%22BRISTOL-MYERS+SQUIBB+COMPANY%22&sortOption=Ver%C3%B6ffentlichungsdatum+ab&queryString=&recNum=1&maxRec=1018

The present invention generally relates to processes for preparing a

tetrahydrocarbazole carboxamide compound.

Protein kinases, the largest family of human enzymes, encompass well over 500 proteins. Btk is a member of the Tec family of tyrosine kinases, and is a regulator of early B-cell development, as well as mature B-cell activation, signaling, and survival.

B-cell signaling through the B-cell receptor (BCR) leads to a wide range of biological outputs, which in turn depend on the developmental stage of the B-cell. The magnitude and duration of BCR signals must be precisely regulated. Aberrant BCR-mediated signaling can cause disregulated B-cell activation and/or the formation of pathogenic auto-antibodies leading to multiple autoimmune and/or inflammatory diseases. Mutation of Btk in humans results in X-linked agammaglobulinaemia (XLA). This disease is associated with the impaired maturation of B-cells, diminished immunoglobulin production, compromised T-cell-independent immune responses and marked attenuation of the sustained calcium signal upon BCR stimulation.

Evidence for the role of Btk in allergic disorders and/or autoimmune disease and/or inflammatory disease has been established in Btk-deficient mouse models. For example, in standard murine preclinical models of systemic lupus erythematosus (SLE), Btk deficiency has been shown to result in a marked amelioration of disease progression. Moreover, Btk deficient mice are also resistant to developing collagen-induced arthritis and are less susceptible to Staphylococcus-induced arthritis.

A large body of evidence supports the role of B-cells and the humoral immune system in the pathogenesis of autoimmune and/or inflammatory diseases. Protein-based therapeutics (such as Rituxan) developed to deplete B-cells, represent an important approach to the treatment of a number of autoimmune and/or inflammatory diseases. Because of Btk’s role in B-cell activation, inhibitors of Btk can be useful as inhibitors of B-cell mediated pathogenic activity (such as autoantibody production).

Btk is also expressed in mast cells and monocytes and has been shown to be important for the function of these cells. For example, Btk deficiency in mice is

associated with impaired IgE-mediated mast cell activation (marked diminution of TNF-alpha and other inflammatory cytokine release), and Btk deficiency in humans is associated with greatly reduced TNF-alpha production by activated monocytes.

Thus, inhibition of Btk activity can be useful for the treatment of allergic disorders and/or autoimmune and/or inflammatory diseases including, but not limited to: SLE, rheumatoid arthritis, multiple vasculitides, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis, allergic rhinitis, multiple sclerosis (MS), transplant rejection, type I diabetes, membranous nephritis, inflammatory bowel disease, autoimmune hemolytic anemia, autoimmune thyroiditis, cold and warm agglutinin diseases, Evan’s syndrome, hemolytic uremic syndrome/thrombotic thrombocytopenic purpura (HUS/TTP), sarcoidosis, Sjogren’s syndrome, peripheral neuropathies (e.g., Guillain-Barre syndrome), pemphigus vulgaris, and asthma.

In addition, Btk has been reported to play a role in controlling B-cell survival in certain B-cell cancers. For example, Btk has been shown to be important for the survival of BCR-Abl-positive B-cell acute lymphoblastic leukemia cells. Thus inhibition of Btk activity can be useful for the treatment of B-cell lymphoma and leukemia.

Atropisomers are stereoisomers resulting from hindered rotation about a single bond axis where the rotational barrier is high enough to allow for the isolation of the individual rotational isomers. (LaPlante et al., J. Med. Chem. 2011, 54, 7005-7022).

US Patent 9,334,290 discloses substituted tetrahydrocarbazole and carbazole compounds useful as Btk inhibitors, including 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide as Example 28. 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide, referred to herein as Compound 8, has two stereogenic axes:

(i) bond “a” between the tricyclic tetrahydrocarbazole/carbazole group and the phenyl group; and (ii) bond “b” between the substituted tetrahydroquinazolinedione group and the phenyl group. Compound 8 has non-symmetric substitutions on the rings connected by the single bonds labeled “a” and “b”, and limited rotation about these bonds caused by steric hindrance. As the rotational energy barriers are sufficiently high, hindered rotations about bond (a) and bond (b) occur at rates that are slow enough to allow isolation of Compound 8 and the other atropisomers of Compound 8 as four individual diastereomeric atropisomer compounds. These four rotational isomers can be separated by

chromatography on a stationary phase to provide chiral mixtures of two atropisomers or individual atropisomers.

US Patent 9,334,290 discloses a multistep synthesis process for preparing the Compound 8. This process is shown schematically in Figures 2-4. The disclosed process includes three chiral separations from racemic mixtures including (i) a chiral separation of a racemic mixture of chiral enantiomers (FIG.2); (ii) chiral separation of a mixture of atropisomers along bond “b” between the substituted tetrahydroquinazolinedione group and the phenyl group (FIG.3); and chiral separation of a mixture of atropisomers along bond “a” between the tricyclic tetrahydrocarbazole/carbazole group and the phenyl group (FIG.4). In each one of these chiral separations, the maximum yield of the desired enantiomer or atropisomer from the racemic mixture is 50%.

There are difficulties associated with the adaptation of this multistep synthesis disclosed in US Patent 9,334,290 to a larger scale synthesis, such as production in a pilot plant or a manufacturing plant for commercial production. Additionally, it is desired to have a process that provides higher yields and/or reduces waste.

Applicants have discovered a synthesis process for the preparation of Compound 8 that provides higher yields, reduces waste, and/or is adaptable to large scale manufacturing.

he invention is illustrated by reference to the accompanying drawing described below.

FIG.1 shows the stereoselective synthesis scheme for the preparation of 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide, Compound 8, according to the processes of second aspect, the third aspect, and the first aspect of the invention.

FIG.2 shows the synthesis scheme disclosed in US 9,334,290 for the preparation of (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide, Compound 5 (Intermediate 26 in US 9,334,290).

FIG.3 shows the synthesis scheme disclosed in US 9,334,290 for the preparation of 8-fluoro-l-methyl-3-(S)-(2-methyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phenyl)quinazoline-2,4(lH,3H)-dione, Intermediate 10 in US 9,334,290.

FIG.4 shows the synthesis scheme disclosed in US 9,334,290 for the preparation of Compound 8 from the coupling reaction of 8-fluoro-l -methyl-3-(S)-(2-methyl-3- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phenyl)quinazoline-2,4(lH,3H)-dione, Intermediate 10, and (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro- lH-carbazole-8-carboxamide, Compound 5, to provide a racemic mixture of Example 27 in US 9,334,290; and the chiral separation of Example 27 to provide Compound 8.

wherein R is Ci-8 alkyl or benzyl;

in the presence of:

(i) one or more bases selected from lithium bases, sodium bases, potassium bases, cesium bases, l,8-diazabicycloundec-7-ene, and 1,1,3,3-tetramethylguanidine; and

(ii) a solvent selected from n-butyl acetate (nBuOAc), cyclopentyl methyl ether (CPME), dimethoxy ethane (DME), dimethylacetamide (DMAc), dimethylformamide (DMF), 1,4-dioxane, ethyl acetate (EtOAc), isobutyl acetate (iBuOAc), isopropyl acetate (IP Ac), isopropyl alcohol (IP A), methanol (MeOH), methyl acetate (MeOAc), methyl isobutyl ketone (MIBK), N-methyl-2-pyrrolidone (NMP), 2-methyltetrahydrofuran (MeTHF), tetrahydrofuran (THF), tetrahydropyran (THP), and mixtures thereof;

to provide said Compound 8.

Intermediate Al

2-amino-4 robenzoic acid


(Al)

5% Pt/C (50% water-wet) (60 g, 6 wt%) was charged to a nitrogen blanketed vessel containing isopropyl acetate (22 L) and 4-bromo-5-fluoro-2-nitrobenzoic acid (1.00 kg, 3.79 mol). The headspace was exchanged three times with nitrogen and followed three times with hydrogen. The reaction mixture was stirred at 25 °C under an atmosphere of hydrogen. After 40 hours, the reaction was complete and the headspace was exchanged three times with nitrogen. The reaction mixture was filtered. The reaction vessel and filter train were rinsed with isopropyl acetate (5 L). The combined organic layers were concentrated under reduced pressure to 5.0 L. The solvent was then exchanged to toluene under reduced pressure and the resulting solids were isolated by filtration, washed with toluene, and dried at 50 °C under reduced pressure to afford 0.59 kg (66% yield) of 2-amino-4-bromo-5-fluorobenzoic acid as a white to off-white crystalline solid.

Additional 2-amino-4-bromo-5-fluorobenzoic acid was obtained by washing the spent catalyst twelve times with 2.75: 1 w/w THF in water (9.0 L). Each portion of wash was allowed to soak the spent catalyst for 30 minutes. The filtrate was concentrated to 10 L. The resulting solids were isolated by filtration, washed with water (1.0 L), and dried at 40 °C under reduced pressure to afford 0.15 kg (17% yield) of 2-amino-4-bromo-5-fluorobenzoic acid as an off-white crystalline solid. ¾ NMR (400 MHz, DMSO-de) δ 8.74 (br s, 2H), 7.50 (d, J=9.6 Hz, 1H), 7.08 (d, J=6.1 Hz, 1H). 13C NMR (101 MHz, DMSO-de) 5 168.2, 149.5, 148.8, 147.2, 119.9, 117.0, 116.8, 114.8, 114.6, 109.1.

HPLC Conditions: Column: Waters X-bridge C-18 (150X4.6mm, 3.5μ); Column

Temeprature: 30 °C; Solvent A: 0.05% TFA in water: acetonitrile (95:05 v/v); Solvent B: 0.05%TFA in water: acetonitrile:methanol (05:75:20 v/v); Diluent: 0.25 mg/ml in acetonitrile; Gradient: %B: 0 min. 5%; 20 min. 95%; 25 min. 95%; 26 min. 5%; stop time 30 min; Flow Rate: 0.8 ml/min; Wavelength: 230 nm; The retention time of 2-amino-4-bromo-5-fiuorobenzoic acid was 13.2 min. The retention time of 4-bromo-5-fluoro-2-nitrobenzoic acid was 12.9 min.

Intermediate A2

4-bromo-5-fluoro- -hydrazinylbenzoic acid hydrochloride

A solution of sodium nitrite (100.0 g, 6.38 mol) and water (1.8 L) was slowly charged to a cold slurry (0 °C) of 2-amino-4-bromo-5-fluorobenzoic acid (1.00 kg, 4.27 mol) in water (2.2 L) containing 35% HCl (2.1 kg, 20.15 mol). The reaction mixture slurry was stirred at 0 °C for 5 hours. The resultant cold diazonium salt slurry was charged over 4 hours to a cold solution (0 °C) of sodium bisulfite (2.66 kg, 25.0 mol in water (7.5 L). The diazonium reaction vessel was rinsed with cold water (2.5 L). The rinse water was transferred slowly to the reaction mixture. After 40 minutes, the reaction mixture was warmed to 20 °C over one hour. The reaction mixture slurry was stirred at 20 °C for 3 hours. After 3 hours, the reaction mixture was slowly transferred to a 60 °C solution of 35% HCl (15.0 kg, 144.0 mol) and water (3.0 L). The vessel was rinsed with water (2.5 L); and transferred to 35% HCl and water reaction mixture. The reaction mixture was stirred at 60 °C for 2 hours. The product was isolated by filtration and washed with water (3.0 L). The wet cake was charged back to the reactor and was

slurried with isopropyl acetate (9.0 L) for 1 hour at 20 °C. The product was isolated by filtration, washed with isopropyl acetate (1.0 L), and dried at 45-50 °C under reduced pressure to afford 0.99 kg (81 % yield) of 4-bromo-5-fluoro-2-hydrazinylbenzoic acid hydrochloride as an off-white crystalline solid in 95% purity. ¾ NMR (400 MHz, DMSO-de) δ 10.04 (br s, 3H), 9.00 (br s, 1H), 7.74 (d, J=9.1 Hz, 1H), 7.61 (d, J=5.8 Hz, 1H). 13C NMR (101 MHz, DMSO-de) δ 167.3, 153.0, 150.6, 144.5, 119.2, 1 18.0, 114.6. HPLC analysis: Column: Zorbax Eclipse Plus C 18 3.5 um, 150 x 4.6 mm ID; Column Temeprature: 30 °C; Solvent A: 10 mM ammonium formate in water:MeOH (90: 10 v/v); Solvent B: MeOH : ACN (70:30 v/v); Diluent: 50% CH3CN(aq); Gradient: %B: 0 min. 0%; 15 min. 90%; 18 min. 100%; stop time 18 min; Flow Rate: 1.0 ml/min; Wavelength: 240 nm. The retention time of the diazonium salt intermediate was 3.7 min. The retention time of the mono-sulfamic acid intermediate was 5.2 min. The retention time of 4-bromo-5-fluoro-2-hydrazinylbenzoic acid hydrochloride was 8.0 min. The retention time of 2-amino-4-bromo-5-fluorobenzoic acid was 8.7 min.

INTERMEDIATE Bl

(3-amino-2-methylphenyl)boronic acid hydrochloride

A 500 mL ChemGlass reactor (Reactor A) was equipped with mechanical stirrer and a nitrogen inlet. To the reactor was added 150 ml of methyl tetrahydrofuran. Next, Pd(OAc)2 (241 mg, 0.02 eq) was added, followed by the addition of P(o-tolyl)3 ligand (654 mg, 0.04 eq). The containers holding the Pd(OAc)2 and P(o-tolyl)3 were rinsed with 15 ml of methyl tetrahydrofuran, and the rinse solvents were added to the reactor. The reactor was sealed, evacuated to less than 150 mbar, and filled with nitrogen gas. This was repeated an additional four times to reduce the oxygen level to below 400 ppm. The reaction mixture was stirred for 30 min. Next, 10 g (1.0 eq) of 3-bromo-2-methyl aniline was charged to the inerted reactor. The container that held the 3-bromo-2-methyl aniline was rinsed with 15 ml of Me-THF and added into the reactor. KOAc (15.6 g, 3 eq) was added to the reactor. A slurry formed. The reaction mixture was inerted by using three vacuum/nitrogen cycles to an oxygen endpoint of less than 400 ppm.

A second 500 ml ChemGlass reactor was charged with 150 mL of MeOH, followed by the addition of 7.2 g (1.5 eq) of B2(OH)4. The resultant slurry was agitated at 25 °C. After 30 min, the B2(OH)4 was fully dissolved. The homogeneous solution was inerted by using 5 vacuum/nitrogen purge cycles to reduce the oxygen level to less than 400 ppm. The B2(OH)4/MeOH solution was transferred to Reactor A under a nitrogen atmosphere.

The reactor was inerted using three vacuum/nitrogen cycles with agitation to reduce the oxygen level to less than 400 ppm. The batch was heated to 50 °C (internal batch temperature). A slurry was observed when the temperature reached 40 °C. After reacting for 3 hrs, HPLC analysis of the reaction mixture showed 0.2 AP starting material remained. N-acetyl cysteine (2.0 g, 0.2 g/g) was added to Reactor A. The reaction mixture was stirred at 50 °C (internal batch temperature) for 30 min. The reaction stream was concentrated through distillation to 5 ml/g (~ 50 ml). Methyl tetrahydrofuran (200 ml, 20 ml/g) was charged to the slurry. The slurry was then concentrated via distillation to 150 ml (15 ml/g). Methyl tetrahydrofuran (150 ml, 15 ml/g) was charged to the reaction mixture. The slurry was cooled to 20 °C (batch temperature). Brine (26 wt%, 25 ml, 2.5 ml/g) was charged followed by the addition of aqueous Na2C03 (20 wt%, 15 ml, 1.5 ml/g). The reaction mass was agitated at a moderate rate (50~75/min) for 30 min. Celite (1 g, 0.1 g/g) was charged to the bi-phasic solution. The resultant slurry was agitated for 30 min. The slurry was filtered and transferred to Reactor B. The Celite cake was washed with 10 ml of methyl tetrahydrofuran. The bottom, lean aqueous phase was split from the organic phase and discarded. Brine (26 wt%, 25 ml, 2.5 ml/g) was charged followed by the addition of aqueous Na2C03 (20 wt%, 15 ml, 1.5 ml/g) to the organic solution. The resultant bi-phasic solution was agitated at a moderate rate (75 rpm) for 30 min. The bottom, lean aqueous phase was split from the organic phase and discarded. B2(OH)4 analysis of the rich organic solution did not detect B2(OH)4.

In Reactor B, the rich organic phase was concentrated via distillation to 50 ml (5 ml/g). The concentrated solution was cooled to 0-5 °C (batch temp). Concentrated HC1 (1.06 kg, 2.0 eq) was charged to the solution over 30 min with the batch temperature maintained below 10 °C. Once the concentrated HC1 was added, a slurry formed. The

slurry was agitated for 2 h at 5 °C. The slurry was filtered. The wet cake was washed with methyl tetrahydrofuran (2 X 20 ml). The cake was collected and dried at 50 °C under 100 mbar vacuum for 6 h to afford 8.4 g of 3-amino-2-methylphenyl)boronic acid hydrochloride as a white solid (83.5 % yield). ¾ NMR (500 MHz, D20) δ 7.48-7.23 (m, 3H), 4.78 (br s, 5 H); 2.32 (s, 3H). 13C NMR (126 MHz, D2O) δ 135.2, 134.7, 130.1, 128.0, 124.3, 17.4.

HPLC analysis: Column: Zorbax Eclipse Plus CI 8 3.5 um, 150 x 4.6 mm ID; Solvent A: 10 mM ammonium formate in water: MeOH=90: 10); Solvent B: CH3CN: MeOH (30:70 v/v); Gradient: % B: 0 Min. 0%; 1 Min. 0%; 15 Min. 90%; 15.1 Min. 0%; Stop Time: 20 min; Flow Rate: 1 ml/min; wavelength: 240 nm. The retention time of (3-amino-2-methylphenyl)boronic acid hydrochloride was 4.4 min. The retention time of (3-amino-2-methylphenyl)boronic acid hydrochloride was 17.8 min.

Intermediate CI

7-fluoro-l-methylindoline-2,3-dione

N,N-dimethylformamide (540.0 mL, 6980 mmol, 100 mass%) was added to a 2-L ChemGlass reactor equipped with a mechanical agitator, a temperature probe, and a cooling/heating circulator. Next, 7-fluoroindoline-2,3-dione (135.0 g, 817.6 mmol, 100 mass%) was added at 25 °C and dissolved to form a dark red solution. The charging ports and the beaker that contained the 7-fluoroindoline-2,3-dione were washed with N,N-dimethylformamide (135.0 mL, 1750 mmol, 100 mass%) and the rinse solution was poured into the reactor. Next, cesium carbonate 60-80 mesh (203.66 g, 625.05 mmol, 100 mass%) was added portion-wise to the reaction mixture. The addition was exothermic and the temperature of the reaction mixture increased from 20 to 25.5 °C. The color of the reaction mixture changed from a dark red solution to a black solution. The reactor jacket temperature was set to 0 °C. Next, iodomethane (56.5 mL, 907 mmol, 100 mass%) was added slowly via an additional funnel at ambient temperature, (iodomethane

temperature) while maintaining the batch temperature at less than 30 °C. Upon stirring, the reaction was exothermic, reaching a temperature of 29.3 °C. The batch temperature decreased to 26.3 °C after 85% of iodomethane was added, and the reaction mixture turned from black to an orange. After the addition of the iodomethane was completed, the jacket temperature was raised to 25.5 °C. The reaction mixture was stirred at 25 °C for 2 hrs.

The reddish orange-colored reaction mixture was transferred to a 1 L Erlenmeyer flask. The reaction mixture was filtered through a ceramic Buchner funnel with a No.1 Whatman filter paper to remove solid CS2CO3 and other solid by-products. In addition to a light-colored powder, there were yellow to brown colored rod-shaped crystals on top of the cake, which were water soluble. The filtrate was collected in a 2-L Erlenmeyer flask. The solids cake was washed with N,N-dimethylformamide (100.0 mL, 1290 mmol, 100 mass%). The DMF filtrate was collected in a 2-L Erlenmeyer flask.

To a separate 5-L ChemGlass reactor was charged water (3000.0 mL, 166530 mmol, 100 mass%). Next, 1.66 g of 7-fluoro-l-methylindoline-2,3-dione was added as seed to the water to form an orange colored suspension. The DMF filtrate was charged to the 5-L reactor slowly while maintaining the batch temp, at less than 29 °C over a period of 60 min. Stirring was maintained at 290 rpm. The orange solids precipitated instantly. The 2-L Erlenmeyer flask was rinsed with N,N-dimethylformamide (55.0 mL, 711 mmol, 100 mass%) and charged to the 5-L reactor. The slurry was cooled to 25 °C and agitated at 200 rpm for 12 hrs. The mixture remained as a bright orange-colored suspension. The slurry was filtered over a No. l Whatman filter paper in a 9 cm diameter ceramic Buchner funnel to a 4L Erlenmeyer flask to provide a bright orange-colored cake. The cake was washed with 1200 mL of water via rinsing the 5000 mL reactor (400 mL x 2), followed by 300 mL of deionized water introduced directly on the orange cake. The wet cake was dried under suction for 40 min at ambient temperature until liquid was not observed to be dripping from the cake. The cake was introduced into a vacuum oven (800 mbar) with nitrogen sweeping at ambient temperature for 1 hr, at 40-45 °C for overnight, and at 25 °C for 1 day to provide 7-fluoro-l-methylindoline-2,3-dione (Q, 130.02 g, 725.76 mmol, 100 mass%, 88.77% yield) as a bright orange-colored solid. ¾ NMR (400 MHz, DMSO-de) δ 7.57 (ddd, J=12.0, 8.5, 1.0 Hz, 1H), 7.40 (dd, J=7.3, 1.0 Hz, 1H), 7.12 (ddd, J=8.5, 7.5, 4.0 Hz, 1H), 3.29 (d, J=3.0 Hz, 3H). 13C NMR (101 MHz, DMSO-de) δ 182.3, 158.2, 148.8, 146.4, 137.2, 125.9, 124.3, 120.6, 28.7.

Intermediate C2

3-fluoro-2-(methylamino)benzoic acid

To a 1-L three neck round bottom flask equipped with a mechanical overhead agitator, a thermocouple, and an ice-water bath was charged NaOH (5.0 N) in water (140.0 mL, 700 mmol, 5.0 mol/L) followed by deionized water (140.0 mL, 7771 mmol, 100 mass%) to form a colorless transparent solution (T = 20.2 °C). 7-fluoro-l-methylindoline-2,3-dione (R, 25 g, 139.55 mmol, 100 mass%) was charged portion-wise while controlling the batch temperature at less than 24 °C with an ice-water bath to provide cooling. 7-fluoro-l-methylindoline-2,3-dione was charged and 50 mL of water was used to rinse off the charging funnel, the spatula, and the charging port. The reaction mixture was a thick yellow-green hazy suspension. The yellow-greenish suspension was cooled to 5.0 °C with an ice-water bath. The mixture was stirred for 15 min. Next, hydrogen peroxide (50% wt.) in water (11.0 mL, 179 mmol, 50 mass%) was charged to a 60 mL additional funnel with deionized (4.0 mL, 220 mmol, 100 mass%). The concentration of H2O2 post dilution was ~ 36.7%. The dilute hydrogen peroxide solution was added over a period of 11 minutes to the 1 L round bottom flask cooled with an ice-water bath and stirred at 350 rpm. The reaction mixture color was observed to become lighter in color and less viscous after 5 mL of the peroxide solution was added. After adding 10 mL of peroxide solution, the reaction mixture became clear with visible solids. At the end of addition, the reaction mixture was a green-tea colored transparent solution. The ice-water bath was removed (batch temperature was 16.6 °C), and the transparent, greenish yellow reaction mixture was allowed to warm to ambient temperature (21.0 °C), stirred for 1 hr.

After the reaction was complete, (1.0 hr), the reaction mixture was cooled to 4.3 °C with an ice-water bath. The reaction mixture was neutralized by the addition 6.0 N HCl (aq.) over a period of 3 hours to minimize foaming and the exotherm, resulting in the formation of a yellow-green suspension. The ice-bath was removed and the quenched reaction mixture was stirred at ambient temperature for 20 min. The yellow-green colored reaction mixture was transferred to a 2 L separatory funnel. Dichloromethane (300.0 mL, 4680 mmol, 100 mass%) was charged to the separatory funnel via rinsing the 1 L 3-necked round bottom flask. The separatory funnel was shaken vigorously, then allowed to settle (phase split was fast). Gas evolution was minor. The top aqueous layer was dark amber in color. The bottom dichloromethane layer was tea-green in color. The bottom rich dichloromethane layer was transferred to a clean 1 L Erlenmeyer flask. Next, the 1 L three necked round bottom flask was rinsed again with dichloromethane (200.0 mL, 3120 mmol, 100 mass%). The dichloromethane rinse was added to the separatory funnel. The separatory funnel was shaken vigorously and allowed to settle (phase split was fast). The top aqueous layer was amber in color (lighter); the bottom

dichloromethane layer was lighter green. The bottom rich dichloromethane layer was transferred to the 1 L Erlenmeyer flask. Dichloromethane (200.0 mL, 3120 mmol, 100 mass%) was charged to the separatory funnel and the separatory funnel was shaken vigorously. The contents were allowed to settle (phase split was fast). The bottom rich dichloromethane layer was transferred to the same 1 L Erlenmeyer flask. Peroxide test strip showed > 10 mg/Liter peroxide concentration. The total volume of the aqueous layer was 540 mL.

In a separate 250-mL Erlenmeyer flask was added sodium thiosulfate

pentahydrate (20.0 g, 80.6 mmol, 100 mass%) followed by deionized water (180.0 mL, 9992 mmol, 100 mass%) to form a colorless solution (10% wt. solution). The sodium thiosulfate solution was added to the combined dichloromethane rich solution in the 1 L Erlenmeyer flask. The contents of the flask were stirred vigorously for 10 hrs at ambient temperature. Peroxide strip did not detect the presence of peroxides in the bottom DCM layer. The top Na2S203 layer was amber in color, the bottom dichloromethane layer was much lighter in color, but was still amber in color. After 10 hrs, the mixture was transferred to a 1 L separatory funnel. The top aqueous layer was discarded.

The dichloromethane solution was washed with 150.0 mL of saturated brine solution. After phase split, the bottom rich dichloromethane layer was transferred to a 1 L flask. The dichloromethane solution was distilled to approximately 150 mL to obtain an amber-colored solution. Next, dichloromethane (120 mL, 1872 mmol, 100 mass%) was added and the mixture was heated to 35-40 °C to fully dissolve the solids. The amber solution was filtered through a 0.45 micron PTFE membrane Zap Cap filtration unit into a 1 L flask. The filtrate was transferred into a 3-neck 1 L round bottom flask fitted with a thermocouple, a heating mantle, a mechanical agitator, and a condenser with a nitrogen inlet. To the flask was charged dichloromethane (120 mL, 1872 mmol, 100 mass%) via rinsing the 1 L flask. The contents of the flask were concentrated under reduced pressure to approximately 140 mL to afford a yellow-green-colored suspension. The mixture was heated to 40.5 °C (refluxing) with stirring at 155 rpm to form a green-colored suspension with white solid pieces. After refluxing for 5 min, heptane (100.0 mL, 683 mmol, 100 mass%) was charged to the above mixture. The batch temperature dropped from 41.3 °C to 33.8 °C and the reaction mixture was a suspension. The mixture was heated to 45 °C. The mixture remained as a suspension with supernatant being amber with white solids. The refluxing was mild. After 36 minutes, (batch temp. = 43.8 °C), heptane (120.0 mL, 819 mmol, 100 mass%) was added to the mixture. The batch temperature dropped to 38.0 °C. The reaction mixture was a suspension. The mixture was heated to 40-45 °C and seeded with 0.3 g of 3-fluoro-2-(methylamino)benzoic acid. The reaction mixture remained as a suspension with supernatant being amber and solid pieces of white color. At t = 1 h 25 min (T = 45.4 °C) heptane (100.0 mL, 683 mmol, 100 mass%) was charged to the mixture causing the temperature to drop to 41.0 °C. At t = 2 h l3 min, (T = 45.6 °C) additional heptane (100.0 mL, 683 mmol, 100 mass%) was added to the mixture causing temperature to drop to 41.7 °C. At t = 3 h 07 min, (T = 45.5 °C), the heating was stopped. The mixture was allowed to cool to 20-25 °C under a nitrogen blanket. The suspension was agitated at ambient temperature for 12 hrs. The mixture was filtered using No.1 Whatman filter paper fitted in a ceramic Buchner funnel to a 1 L Erlenmeyer flask. The solids were observed to settle quickly. The mother liquor was green in color. The bottom half of the round bottom flask was coated with a thin dark amber or brown film, which was water soluble. The 1 L round bottom flask was washed with 150 mL of heptane, and then the heptane was used to wash the collected off-white-colored solid.

The filter cake was allowed to dry at ambient temperature with suction for 10 min., then dried in a vacuum oven with nitrogen sweeping at 45-50 °C for 4 hrs, followed by drying at ambient temperature for 10 hrs, with nitrogen sweeping. 3-fluoro-2-(methylamino)benzoic acid (16.1 g) was isolated in 68.1 % yield. ¾ NMR (400 MHz, DMSO-de) δ 7.61 (d, J=7.7 Hz, IH), 7.23 (dq, J=7.9, 1.6 Hz, IH), 6.57 (td, J=8.0, 4.4 Hz, IH), 3.02 (d, J=6.8 Hz, 4H). 13C NMR (101 MHz, DMSO-de) δ 169.5, 153.1, 150.7, 141.8, 141.7, 127.4, 127.4, 120.9, 120.7, 114.8, 114.7, 114.4, 114.3, 32.8.

Intermediate C3

3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic

A 20 L jacketed glass reactor with an overhead mechanical agitator, a

thermocouple, a nitrogen inlet, a glass baffle, and a condenser rinsed with 4 liters of dichloromethane followed by nitrogen sweeping through bottom valve overnight. To the reactor was charged 3-fluoro-2-(methylamino)benzoic acid (1004.7 g, 5939.7 mmol, 100 mass%) followed by dichloromethane (6000 mL, 93400 mmol, 99.8 mass%) to form an off-white-colored suspension. Next, cesium carbonate (1035.2 g, 3170 mmol, 99.9 mass%) was added followed the addition of water (6000 g, 333056 mmol, 99 mass%) at ambient temperature. The batch temperature rose from 17.0 °C to 29.6 °C prior to addition of the water. Gas evolution was observed during the water charging. The colorless biphasic mixture was stirred for 15 min. The batch temperature was approximately 18.8 °C. Next, n-propyl chloroformate (806.0 g, 6445.4 mmol, 98 mass%) was charged to an addition funnel. The reaction mixture was cooled to 15.0 °C with a glycol circulator. The n-propyl chloroformate was added from the addition funnel to the mixture while maintaining the batch temperature between 15.0 and 20.0 °C over 1 hr with stirring at 156 rpm. At the end of the addition, the batch temperature was 18.1 °C. The jacket temperature was increased to 20 °C. The white milky reaction mixture was agitated for 90 minutes.

The agitation was stopped and the reaction mixture was allowed to settle for phase split for 50 min. The hazy, bottom rich dichloromethane layer split from the aqueous layer and was transferred to a carboy. Next, 500 g of anhydrous Na2S04 (s) and 100 g of 60-200 mesh silica gel was added to the dichloromethane solution of 3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid in the carboy. The dichloromethane solution was allowed to dry overnight.

The dichloromethane solution containing the 3-fluoro-2-(methyl

(propoxycarbonyl)amino)benzoic acid was transferred from the carboy to a clean 20 L reactor via a 10 micron Cuno® in-line filter under vacuum to remove solid Na2S04 and silica gel. The carboy was rinsed with 1 liter x 2 of dichloromethane to remove residual solids. The dichloromethane was distilled off in the 20 L reactor with the jacket temperature set at 32 °C, the batch temperature at 15 °C, and vacuum set to 200-253 torr. At the end of distillation, the crude product was a thick light-amber-colored syrup. The solution was concentrated to 3 L of dichloromethane, and refilled with 3 L of dichloromethane each time to a final fill volume of 6 L. Next, 1 liter of dichloromethane was charged via vacuum to the residue in the 20-L reactor. The solution of 3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid became hazier. The solution was filtered using a Buchner funnel with a No.1 filter paper into a new carboy. The reactor was rinsed with 500 mL x 2 of dichloromethane and the rinse was filtered through the same Buchner funnel. All the filtrates were combined in a carboy and stored at the ambient temperature under nitrogen. Yellow-colored solids were observed to settle at the bottom of the carboy. The solution of 3-fluoro-2-(methyl (propoxycarbonyl)amino)benzoic acid in dichloromethane was transferred back to the clean 20-L reactor via vacuum and a 1 micron Cuno® in-line filter. The filtrate was still slightly hazy. The carboy was rinsed with 300 mL x 3 of dichloromethane and the rinses were transferred to the reactor via the 1 micron Cuno® filter. The reactor walls were rinsed with 500-mL of dichloromethane. The dichloromethane solution was concentrated by distillation under reduced pressure until the volume was less than 2.0 liters.

The temperature of the reactor jacket was lowered to 30 °C. The vacuum was broken and the reactor was filed with nitrogen. To the reactor was added 2 liters of cyclohexane followed by 5.0 g of 3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid crystalline seed. The seeds did not dissolve. The mixture was allowed to stir at 30 °C for 5-10 min to form a thick slurry. Additional cyclohexane (2.0 L) was added over 2 minutes. The jacket temperature was lowered to 25 °C. The mixture was allowed to stir for 40 min. Additional cyclohexane (2.0 L) was added over 2 minutes. The j acket temperature was lowered to 23 °C. The suspension was maintained at 23 °C for 60 min. Additional cyclohexane (2.0 L) was added over 2 minutes. The suspension was stirred for 20 min. The jacket temperature was lowered to 19.0 °C. The suspension was maintained at 19-21 °C for 10 hrs. The slurry settled well after overnight aging. A sample of the supernatant was obtained and assessed for the loss based on 9.5 L total volume. The slurry was filtered to collect solids via a ceramic Buchner funnel with a No. l Whatman filter paper. The solids were crystalline and white when dry. The wet cake was washed with cyclohexane (~ 2000 mL x 3) followed by drying for 10 min. The cake volume was 4933 cm3. The wet cake was transferred to four Pyrex glass trays for heated drying. The drying was continued in a vacuum oven at ~ 35-40 °C with nitrogen sweeping for 12 hrs to afford 1302.9 g of 3-fluoro-2-(methyl(propoxycarbonyl)amino) benzoic acid in 85.9 % yield. ¾ NMR (400 MHz, DMSO-de) (3: 1 mixture of rotamers) δ 13.2 (br s, 1H), 7.72-7.67 (m, 1H), 7.58-7.52 (m, 1H), 7.49-7.43 (m, 1H), 4.06-3.95 (m, 0.50H), 3.90 – 3.80 (m, 1.50H) 3.12 (s 0.75H), 3.12 (s 2.25H), 1.67 – 1.58 (m, 0.50H), 1.42 – 1.34 (m5 1.50H), 0.93 (t, J=7.5 Hz, 0.75H), 0.67 (t, J=7.5 Hz, 2.25H). 13C NMR (101 MHz, DMSO-de) (mixture of rotamers) δ 165.8, 159.0, 156.6, 154.3, 131.6, 131.0, 128.7, 128.6, 126.3, 1 19.9, 119.7, 66.6, 66.4, 36.9, 36.4, 36.4, 21.8, 21.5, 10.0, 9.8.

HPLC Analysis: Column: Agilent ZORBAX Eclipse Plus C18 3.5um 4.6X150 mm; Column Temeprature: 40 °C; Solvent A: 0.01M NH4OOCH in water:MeOH (90: 10 v/v); Solvent B: O.OIM NH4OOCH in MeOH:CH3CN (70:30 v/v); Diluent: 0.25 mg/ml in acetonitrile; Gradient: %B: 0 min. 10%; 10 min. 30%; 20 min. 90%; 20.1 min. 10%; stop time 25 min; Flow Rate: 1.0 ml/min; Wavelength: 220 nm;

The retention time of 7-fluoro-l-methylindoline-2,3-dione was 10.7 minutes.

The retention time of 7-fluoroindoline-2,3-dione was 6.8 minutes. The retention time of 3-fluoro-2-(methylamino)benzoic acid was 5.9 minutes. The retention time of 3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid was 12.0 minutes.

Compound 1

(S)-3-(prop-l -en-2-yl)cyclohexan-l-one

Catalyst Preparation: Rhodium (I) (S)-(+)-5,5′-bis[di(3,5-di-tert-butyl-4-methoxyphenyl) phosphino] -4,4′-bi- 1 ,3-benzodioxole

Methanol (320 mL) was charged into a 0.5 L inerted reactor equipped with an overhead agitator, nitrogen sparging tube and an outlet connected to an oxygen meter. The reactor was inerted by sparging nitrogen subsurface through methanol until <300 ppm 02 was detected in the headspace. S-(+) DTBM-SEGPHOS (77.3 g, 65.6 mmol) and [Rh(cod)Cl]2 (15.4 g, 31 mmol) were charged and the nitrogen sparging continued until <300 ppm C was detected in the headspace. The mixture was agitated at room temperature under constant positive nitrogen pressure for 30 min by sweeping a low flow of nitrogen through the headspace. The initial yellow slurry gradually transformed into a deep-red solution containing a small amount of solids (excess ligand). The ligation completion was confirmed by 1P NMR by disappearance of the ligand peak at 13.1 ppm (s) and the appearance of the new singlets at 26.10 ppm and 27.01 ppm for the ligated species.

Synthesis of the Compound I

A 20 L jacketed Chemglass reactor, equipped with an overhead agitator, a thermocouple, nitrogen sparging tube, a sampling port, a condenser connected to the glycol supply and a nitrogen outlet connected sequentially to a bubbler, flow meter and an oxygen meter, was inerted using a vigorous nitrogen sweep. A Teledyne 3110 oxygen meter was used to monitor the progress of inertion. A vigorous nitrogen sweep was implemented prior to reagent charges until the oxygen reading was <300 ppm.

Heptane (4.0 L), 2-cyclohexen-l-one (1 kg, 10.4 M) in heptane (1.0 L), isopropenyl pinacol boronate (1.92 kg, 11.4 M, 1.1 eq) in heptane (1.0 L), DIPEA (0.91 L, 0.67 kg, 0.50 eq), a solution of 2,2-dimethy 1-1, 3 -propanediol (1.19 kg, 1.1 eq) in methanol (0.12L) in water (3 L), and additional heptane (2.55L) were sequentially charged to the reactor via vacuum. Nitrogen sparging subsurface through the agitated bi phasic mixture continued after the charges until an oxygen level of <300 ppm was

reached in the headspace prior to the catalyst charge. Then the nitrogen flow was reduced to maintain a slight positive pressure in the reactor.

The catalyst light slurry was transferred from the bottom value of the 0.5 L reactor’s bottom into the 20 L reactor through an inerted Teflon tubing by applying slight positive pressure of nitrogen. The contents of the small reactor was transferred including the excess of the undissolved solid.

The jacket was set to 60 °C on the 20 L reactor and the biphasic mixture was vigorously heated and agitated under nitrogen at 55-58 °C. After the transfer, the nitrogen flow was reduced to maintain a slight positive pressure and to minimize solvent loss. After completion of the reaction, the reaction mixture was cooled to 20-25 °C. The phases were separated and the organic phase was washed with IN HC1 aq (v=5.7 L, 0.55 eq) to remove DIPEA, and with water (2.5 L). Two back-extractions with heptane (2 x 2L) from the original aqueous phase were performed to bring back an additional 8 mol% of the product. All organic phases were combined and polished filtered back to the cleaned reactor. Heptane was removed under reduced pressure (30-40 °C at 45-55 torr) to give the crude product, which was transferred to a 2 L 4-necked round bottom flask, equipped with a mechanical stirrer, a thermocouple, a 30 cm Vigreaux column, a distillation adapter containing a thermocouple to measure the vapor temperature, a condenser (glycol) and a Teflon tubing attached to a receiver flask. Distillation was performed at a pressure of 10 torr with the main fraction containing the product boiling at 85-92 °C to afford 1.18 kg (85 mol % as is, 82.1 % corrected) of (S)-3-(prop-l-en-2-yl)cyclohexan-l-one. Chiral GC: Supelco AlphaDex 120 30 x 0.25 mm x 0.25 μπι, inlet 200 °C, split ratio 30: 1, carrier gas: helium, constant flow 1.9 mL/min, oven program: 80 °C to 110 °C at 2 °C /min, then 20 °C /min to 220 °C, detector: FID 250 °C; RT for the desired product: 14.4 min. Chemical purity: 97.1 GCAP. Chiral purity: ee = 99.6 %. ¾ NMR (CDCh): 1.57-1.70 (m, 12H), 1.75 (s, 3H), 1.91-1.96 (m, 1H), 2.05-2.12 (m, 1H), 2.26-2.46 (m, 5H), 4.73 (s, 1H), 4.78 (s, 1H).

Compound 2

(S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l-en-2-yl)cyclohexylidene)hydrazinyl)benzoic acid 

(S)-3 -(prop- l -en-2-yl)cyclohexan-l -one (50.00 mL, 33.4 mmol, 0.667 mmol/mL) solution in heptane was added to a Chemglass reactor. Next, 75 mL of MeOH was added. The MeOH solution was distilled at 60 torr/50 °C jacket temperature and 75 mL of constant volume with the addition of 300 mL of MeOH. The contents of the reactor were cooled to 20 °C. 2-amino-4-bromo-5-fluorobenzoic acid (8.5415 g, 29.918 mmol) was added to the reactor. The reaction mixture was stirred at 20 °C. After, 30 minutes, the solid material was dissolved to form a clear brown solution. After 2.0 h, water (25.0 mL) was added over 25 min to the reaction mixture under slow agitation (RPM = 100). After an additional 1.0 h, the slurry was filtered (fast; < 3 seconds). The cake was washed with 2×25 mL of MeOH/H20 (3:2). The cake was dried at 55 °C under vacuum overnight to afford (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l -en-2-yl)cyclohexylidene)

hydrazinyl)benzoic acid (10.5701 g; 95.7% yield). HPLC method: Column: Zorbax Eclipse plus 1.8 um C8 (4.6 X 50 mm); inj ection volume: 10 μί; Mobile Phase A: 0.05% TFA in acetonitrile: water (5 :95, v/v); Mobile Phase B: 0.05% TFA in water: acetonitrile (5:95, v/v); Gradient (%B) 0 min (30%), 14 min (100%), 15 min (30%); Flow Rate: 1.0 mL/min; Wavelength: 240 nm for IPC; Column temp: 25 °C; IPC Sample Prep:

Dissolved 10 of the reaction mixture and dilute with MeOH to 1.5 mL; HPLC results: Intermediate A2, 0.87 min; Compound 2, 9.97 min. ¾ NMR (400 MHz, DMSO-de) δ 13.54 (s, 1H), 10.76 (d, J = 26.5 Hz, 1H), 7.73 (appt triplet, J = 6.32 Hz, 1H), 7.64 (dd, J = 9.35, 1.26 Hz, 1H), 4.77-4.75 (m, 2H), 2.68-2.61 (m, 1H), 2.46-2.44 (m, 1H), 2.27-2.12 (m, 2H), 2.06-1.97 (m, 1H), 1.96-1.86 (m, 1H), 1.82-1.80 (m, 1H), 1.75-1.74 (m, 3H), 1.50-1.41 (m, 2H). 13C NMR (100 MHz, DMSO-de) δ 168.67, 152.76, 152.73, 150.71 , 148.41 , 148.38, 148.20, 145.10, 117.45, 117.21 , 116.45, 1 16.40, 1 15.76, 1 15.74, 1 15.54, 1 15.52, 109.64, 109.39, 108.88, 108.85, 108.83, 108.80, 44.80, 43.72, 34.22, 30.89, 30.08, 30.05, 25.42, 25.39, 24.15, 20.60, 20.44.

Compound 3

(S)-5-bromo-6-fluoro-2-(prop-l-en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxylic acid

Zinc chloride (8.7858 g, 64.46 mmol) and (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop- 1- en-2-yl)cyclohexylidene)hydrazinyl)benzoic acid (17.0011 g, 46.05 mmol) were added to a Chemglass reactor. Next, isopropyl acetate (170 mL) was added. The contents of the reactor were heated at 69.5 °C for 71 h and then cooled to room temperature. 2-MeTHF (205 mL) and HC1 (1 mol/L) in water (85 mL) were added. The reaction mixture was stirred at room temperature for 0.5 h. The layers were allowed to separate. The organic layer was washed with water (85 mL). The layers were separated and the organic layer was polish-filtered. The rich organic layer was distilled at 220 torr and 70 °C jacket temperature to 85 mL (5.0 mL/g (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l-en-2-yl)cyclohexylidene)hydrazinyl) benzoic acid). Next, the solution was distilled at 120 mL (7.0 mL/g (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l-en-2-yl)cyclohexylidene)hydrazinyl) benzoic acid) constant volume under 220 torr and 70 °C jacket temperature with continuous addition of acetonitrile (350 mL, 20 mL/g). Additional CFbCN was added to make the slurry volume = 153 mL (9.0 mL/g (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l-en- 2- yl)cyclohexylidene) hydrazinyl)benzoic acid). The slurry was heated to 82 °C batch temperature. After 3.0 h, the slurry was cooled to 20 °C over 2.0 h. The slurry was stirred at 20 °C for an additional 14 h. The slurry was filtered and the cake was washed with acetonitrile (2 x 17 mL, 1.0 mL/g (S,E)-4-bromo-5-fluoro-2-(2-(3-(prop-l-en-2-yl)cyclohexylidene) hydrazinyl)benzoic acid). The wet cake was dried in a vacuum oven at a temperature range of 50-55 °C overnight to afford (S)-5-bromo-6-fluoro-2-(prop-l-en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxylic acid (7.8991 g; 48.7% yield). HPLC method: Column: Agilent Zorbax Eclipse plus 1.8 μπι C8 (4.6 X 50 mm);

Injection Volume: 10 μί; Mobile Phase A: 0.05% TFA in acetonitrile: water (5:95, v/v); Mobile Phase B: 0.05% TFA in water: acetonitrile (5:95, v/v); Gradient (%B) 0 min

(30%), 14 min (100%), 15 min (100%); Flow Rate: 1.0 mL/min; Wavelength: 240 nm for IPC and Isolated product; Column temp: 25 °C; IPC Sample Prep: 1 mL/100 mL in tetrahydrofuran; Isolated Sample Prep: 0.25 mg/mL in tetrahydrofuran; HPLC results: Compound 3, 8.86 min; Compound 2, 10.0 min. ¾ NMR (400 MHz, DMSO-de) δ 13.41 (s, 1H), 11.03 (s, 1H), 7.45 (d, J = 9.85 Hz, 1H), 4.79 (appt d, J = 4.55Hz, 2H), 3.21-3.17 (m, 1H), 2.95 (dd, J = 17.18, 4.80 Hz, 1H), 2.91-2.83 (m, 1H), 2.61 (dd, J = 16.93, 10.61 Hz, 1H), 2.41-2.35 (m, 1H), 2.01-1.95 (m, 1H), 1.79 (s, 3H), 1.67-1.57 (m, 1H). 13C NMR (100 MHz, DMSO-de) δ 166.64, 166.61, 152.72, 150.42, 148.44, 139.96, 131.90, 127.44, 127.43, 112.40, 112.33, 109.67, 109.54, 109.39, 109.19, 109.14, 28.28, 27.79, 22.20, 20.69.

Compound 4

(S)-5-bromo-6-fluoro-2-(prop- -en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide

Acetonitrile (70 mL) was added to a Chemglass reactor, followed by the addition of (S)-5-bromo-6-fluoro-2-(prop-l-en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxylic acid (7.0150 g). Next, Ι,Γ-carbonyldiimidazole (4.2165 g, 26.004 mmol) was added. The reaction mixture was stirred (RPM = 100) for 5.0 hr at 20 °C. The slurry was cooled to 3 °C. Ammonia (30 mL, 200 mmol, 30 mass%) was added in less than 2 min. The slurry was stirred at 3 °C for 17.5 h. Water (70 mL) was added over 5 min. The slurry was stirred at 3 °C for 3 h. The slurry was filtered and the wet cake was washed with 2×50 mL of CH3CN/H2O (1 : 1). The wet cake was dried at 55 °C under vacuum overnight to afford (S)-5-bromo-6-fluoro-2-(prop-l-en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (5.2941 g; 75.8% yield). HPLC Method; Column: Agilent Zorbax Eclipse plus 1.8 μιη C8 (4.6 X 50 mm); Injection Volume: 10 μί; Mobile Phase A: 0.05% TFA in acetonitrile: water (5:95, v/v); Mobile Phase B: 0.05% TFA in water: acetonitrile (5:95, v/v); Gradient (%B) 0 min (0%), 8 min (100%), 10 min (100%); Flow Rate: 1.0 mL/min; Wavelength: 240 nm for IPC and Isolated product; Column temp: 25 °C; IPC Sample

Prep: Dissolved 10 of the reaction mixture into 1.0 mL 0.05 v% DBU/MeOH;

Product sample preparation: Dissolved product in MeOH at 1 mg/mL; HPLC results: Compound 4, 6.39 min; Compound 3, 6.80 min. ¾ NMR (400 MHz, DMSO-de) δ 11.05 (s, 1H), 8.11 (s, 1H), 7.59 (d, J = 10.36 Hz, 1H), 7.55 (br s, 1H), 4.78 (br s, 2H), 3.18 (br d, J = 14.65 Hz, 1H), 2.94 (dd, J = 16.93, 4.80 Hz, 1H), 2.88-2.82 (m, 1H), 2.62 (dd, J = 16.93, 10.61 Hz, 1H), 2.40-2.34 (m, 1H), 1.98 (d, J = 11.87 Hz, 1H), 1.78 (s, 3H), 1.66-1.56 (m, 1H). 13C NMR (100 MHz, DMSO-de) δ 167.64, 152.68, 150.38, 148.47, 139.47, 131.71, 127.02, 127.01, 115.36, 115.28, 109.53, 108.66, 108.61, 107.47, 107.19, 28.24, 27.87, 22.21, 20.67.

Compound 5

(S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide

Dichloromethane (100 mL) and (S)-5-bromo-6-fluoro-2-(prop-l-en-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (PPP, 10.0016 g, 28.48 mmol) were added to a 250 mL Chemglass reactor. The slurry was cooled to 5 °C. Next, trifluoroacetic acid (14.68 g, 128.7 mmol) was added over 0.5 h with agitation (RPM = 250) while maintaining the internal temperature at less than 10 °C). The temperature was raised to 14 °C and the reaction mixture was stirred at 14 °C for 17.5 h. Next, 60 mL of MeOH was added to dissolve the thin slurry. The solution was cooled to -10 °C. The solution was distilled at 80 torr while the jacket temperature was gradually raised from -10 °C to 20 °C. The solution was distilled to about 60 mL volume. The internal temperature changed from -7 °C to -2 °C. The solution became a heavy slurry. The distillation was continued at 80 torr at 20 °C jacket temperature at 60 mL volume with the addition of 120 mL MeOH. The intemal temperature changed from -2 °C to 15 °C. The solution became a heavy slurry. The distillation became slow. The vacuum pressure was changed to 60 torr, and the distillation was continued with a 20 °C jacket temperature to 40 mL slurry volume. The batch temperature went from 12 °C to 13 °C.

MeOH (20 mL) was sprayed to wash solid crust off the reactor wall, but was not effective. Aqueous N¾ (30.0 mL, 400 mmol, 28 mass%) was sprayed to the slurry (pH = 10.59). Some solid crust on the upper reactor wall still remained. The slurry was stirred at 20 °C for 0.5 h (pH = 10.58), then heated to 70 °C in 15 min. All the solid crust on the upper reactor wall dissolved. Next, water (40 mL) was added over a period of 15 min. The solution remained as a clear solution at 70 °C.

The slurry was seeded with solid (S)-5-bromo-6-fluoro-2-(2 -hydroxy propan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (~ 5 mg). The seeds remained but there was little additional crystallization was observed at 70 °C. The slurry was heated at 70 °C (jacket temperature = 80 °C) for 0.5 h, and then cooled down to 20 °C in 0.5 h. At 65 °C the mixture became cloudy. The mixture was stirred at 20 °C for 65 h. The mixture was filtered. The cake was washed with 2×15 mL of MeOH/LhO (1 : 1). The wet cake was dried at 65 °C under vacuum for 24 h, giving (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (9.1741 g, 87.3% yield).

(S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide was recrystallization in MeOH/MTBE/n-Heptane (1 :4:8).

(S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (8.0123 g) was added to a reactor. Next, MeOH (8.0 mL) and MTBE (32.0 mL) were added. The mixture was heated to 45 °C to dissolve the slurry. Heptane (64 mL) was added over a period of 15 min at 45 °C. The slurry was stirred at 45 °C for an additional 0.5 h and then cooled to 5 °C in 1.0 h. Stirring was continued at 5 °C for an additional 1.0 h. The slurry was filtered and the wet cake was washed with 2×20 mL of n-heptane. The wet cake was dried at 65 °C under vacuum for 16 h to afford (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (6.9541 g; 86.8%).

(S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (8.0123 g) was added to a reactor. Next, MeOH (8.0 mL) and MTBE (32.0 mL) were added. The mixture was heated to 45 °C to dissolve the slurry. Heptane (64 mL) was added over a period of 15 min at 45 °C. The slurry was stirred at 45 °C for an additional 0.5 h and then cooled to 5 °C in 1.0 h. Stirring was continued at 5 °C for an additional 1.0 h. The slurry was filtered and the wet cake was washed with 2×20 mL of n-heptane. The wet cake was dried at 65 °C under vacuum for 16 h to afford (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (6.9541 g; 86.8%). HPLC method Column: Phenomenex Kinetex C18 2.6um 100A 4.6X150mm SN:538219-97; Injection Volume 5 μί; Mobile Phase A: 0.05% TFA in acetonitrile:water (5:95, v/v); Mobile Phase B: 0.05% TFA in

water: acetonitrile (5 :95, v/v); Gradient (%B) 0 min (32%), 5 min (38%), 1 1 min (38%), 18 min (68%), 22 min (68%), 30 min (90%), 31 min (100%); Flow Rate: 1.0 mL/min; Wavelength: 220 nm for IPC and Isolated product; Column temp: 25 °C; IPC Sample Prep: 1 μΙ71 mL in tetrahydrofuran; Isolated Sample Prep: 0.25 mg/mL in

tetrahydrofuran; HPLC results: Compound 5, 9.58 min; Compound 4, 19.98 min; ¾ NMR (400 MHz, DMSO-de) δ 10.99 (s, 1H), 8.10 (s, 1H), 7.57 (d, J = 10.36 Hz, 1H), 7.54 (br s, 1H), 4.27 (s, 1H), 3.26 (dd, J = 15.66, 4.29 Hz, 1H), 2.93 (dd, J = 17.18, 4.55 Hz, 1H), 2.76-2.68 (m, 1H), 2.44 (dd, J = 16.17, 1 1.87 Hz, 1H), 2.12 (br d, J = 1 1.12 Hz, 1H), 1.69-1.62 (m, 1H), 1.31 (ddd, J = 25.01, 12.38, 5.31 Hz, 1H), 1.14 (s, 6H). 13C

NMR (100 MHz, DMSO-de) δ 167.67, 152.64, 150.34, 140.46, 131.77, 127.03, 127.02, 1 15.28, 1 15.21, 109.09, 109.05, 107.30, 107.03, 101.43, 101.19, 70.37, 44.96, 27.17, 26.73, 24.88, 24.36, 22.85.

Compound 6

(2S)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro- lH-carbazole-8-carboxamide

Catalyst activation

Into a 1 Liter Chemglass reactor (Reactor A) were added Me-THF (4 L/kg) followed by (R)-BINAP (0.0550 mol/mol, 7.45 mmol) and Pd(OAc)2 (0.0500 mol/mol, 6.77 mmol). Additional Me-THF (1 L/kg) was added. The mixture was stirred at 25 °C

for 1 h. Next, 4-bromo-3-fluoro-7-(l-hydroxy-l-methyl-ethyl)-6,7,8,9-tetrahydro-5H-carbazole-l-carboxamide (0.10 equiv, 13 mmol) was added into the mixture in Reactor A, followed by the addition of 2-methyltetrahydrofuran (0.50 L/kg) and water (0.5 L/kg).

The overhead space of Reactor A was sparged with nitrogen at 1 mL/second for 40 min at 25 °C. The resulting mixture was then stirred at 70 °C for 3 h under a positive pressure of nitrogen (1.05 atm). The resulting mixture containing the activated catalyst was cooled to

25 °C and kept at 25 °C under a positive pressure of nitrogen before use.

To a 500 mL Chemglass reactor (Reactor B) were added water (6 L/kg) followed by K3PO4 (6 equiv., 813 mmol). The addition was exothermic. The mixture was stirred till the base was fully dissolved. The overhead space of Reactor B was sparged with nitrogen at 1 mL/second for 60 min at 25 °C. The K3PO4 solution in Reactor B was then kept under a positive pressure of nitrogen before use.

To Reactor A, which contained the activated catalyst, was added 4-bromo-3-fluoro-7-(l-hydroxy-l-methyl-ethyl)-6,7,8,94etrahydro-5H-carbazole-l-carboxarnide (0.90 equiv., 122 mmol), followed by THF (2.5 L/kg). Then (3-amino-2-methyl-phenyl)boronic acid hydrochloride (1.15 equiv., 156 mmol) and MeOH (2 L/kg) were added to Reactor A. The overhead space of Reactor A was sparged with nitrogen at 1 mL/second for 40 min. Then the reaction mixture in Reactor A was cooled to -10 °C under a positive pressure of nitrogen.

The K3PO4 aqueous solution in Reactor B was then transferred into Reactor A via a cannula while both reactors were kept under a positive pressure of N2. The rate of transfer was controlled so that the inner temperature in Reactor A was below 0 °C throughout the operation.

The resulting biphasic reaction mixture was stirred at 5 °C under a positive pressure of nitrogen. After 2.5 h at 5 °C, HPLC analysis of the reaction mixture showed

0.3 AP starting material remained. The reaction mixture was then warmed to 25 °C and stirred at 25 °C for 30 min. HPLC analysis of the reaction mixture showed 0.0 AP starting material remained.

N-acetyl-L-cysteine (1 kg/kg, 306 mmol) and water (2.5 L/kg) were added into Reactor A. The resulting mixture was stirred at 40 °C for 2 h then cooled to 25 °C. The bottom layer (aqueous layer) was discharged and the top layer (organic layer) was retained in the reactor.

Afterwards, THF (1 L/kg) and NaCl solution (13 mass%) in water (7 L/kg) were added into Reactor A, and the resulting mixture was stirred at 25 °C for lh. The bottom layer (aqueous layer) was discharged and the top layer (organic layer) was retained in the reactor.

The organic layer was filtered through a polyethylene filter. Then the reactor was rinsed with Me-THF (0.50 L/kg). The rinse was filtered through the polyethylene filter and combined with the filtrate. The solution was transferred into a clean 1 L reactor (Reactor C).

The mixture in Reactor C was concentrated under reduced pressure to 8.8 L/kg. (2 L/kg solvent was removed by distillation). At 50 °C, n-BuOH (4 L/kg) was added slowly over 2 h. The mixture was then stirred at 50 °C for 2.5 h, and a slurry was obtained.

The solvent was swapped to n-BuOH through constant volume distillation. During this operation, n-BuOH (8 L/kg) was used and 8 L/kg solvent was removed from Reactor C. The resulting mixture was stirred at 55 °C for 1 h and cooled to 25 °C over 1 h.

The slurry in Reactor C was filtered. The reactor rinsed with n-BuOH (2 L/kg).

The cake was then washed with this reactor rinse, followed by heptane (8 L/kg). The product was dried under vacuum at 55 °C for 24 h to afford (2S,5R)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide, which was isolated as an off-white solid powder (46.2 g, 86% yield).

HPLC analysis: (2S,5R)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide: 98.1 AP (19.2 min); (2S,5S)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide: 1.8 AP (19.9 min), (S)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide: 0.1 AP (20.9 min). Column: Waters XBridge BEH C18 S-2.5um 150 X 4.6mm; Solvent A: 10 mM sodium phosphate buffer pH 7; Solvent B: CH3CN:MeOH (50:50 v/v); Gradient: % B: 0 Min. 5%; 4 Min. 30%; 41 Min. 95%; 47 Min. 95%; Stop Time: 48 min; Flow Rate: 0.7 ml/min wavelength: 240 nm. ¾ NMR (500 MHz, DMSO-de) δ 10.76 (s, 1H), 8.09 (br s, 1H), 7.54 (d, J=10.7 Hz, 1H), 7.47 (br s, 1H), 6.96 (t, J=7.7 Hz, 1H), 6.72 (d, J=7.9 Hz, 1H), 6.41 (d, J=7.3 Hz, 1H), 4.90 (s, 2H), 4.19 (s, 1H), 2.91 (br dd, J=16.6, 4.0 Hz, 1H), 2.50-2.39 (m, 1H), 2.05-1.93 (m, 1H), 1.88-1.75 (m, 5H), 1.64-1.53 (m, 1H), 1.21-1.11 (m, 1H), 1.09 (s, 6H). 13C NMR (126 MHz, DMSO-de) δ 169.0 (d, J=2.7 Hz), 152.5 (d, J=229.8 Hz), 146.7, 139.1,

134.4, 132.0, 127.7 (d, J=4.5 Hz), 125.6, 123.3 (d, J=20.0 Hz), 120.5, 119.2, 1 15.1 (d, J=7.3 Hz), 1 14.3, 109.5(d, J=4.5 Hz), 107.2 (d, J=27.3 Hz), 70.9, 45.9, 27.6, 27.2, 25.3, 25.0, 22.7, 14.7.

Compound 7

propyl (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro- lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl)carbamate

N, N-Dimethylformamide (7.0 L, 7 L/kg) was charged into a reactor followed by the addition of (2S)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (1 kg, 2528 mmol, 1.0 eq.). 3-Fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid (0.774 kg, 3034 mmol, 1.2 eq.) was added to the reactor, followed by the addition of 1 -methylimidazole (0.267 kg, 3287 mmol, 1.3 eq) and methanesulfonic acid (0.122 kg, 1264 mmol, 0.5 eq.) at 20 °C. The reaction mixture was stirred for at 20 °C for 30 min to completely dissolve the reaction contents. The reaction mixture was cooled to 10 °C and EDAC (l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride) (0.679 kg, 3540 mmol, 1.4 eq) was charged into the reactor. An exotherm of approximately 4 °C was observed. The reaction mixture was stirred at 10 °C for 4 h.

After 4 hrs, the reaction mixture was warmed to 20 °C. Isopropyl acetate (25 L, 25 L/kg) was added to the reaction mixture followed by 25 wt% aqueous sodium chloride solution (2.5 L, 2.5 L/kg) and 1.0 M aqueous hydrochloric acid (2.5 L, 2.5 L/kg). The reaction mixture was stirred for 30 min. The agitation was stopped and the bottom aqueous layer was separated. Water (5 L, 5 L/kg) was charged to the rich organic solution and stirred for 30 min. The agitation was stopped and the bottom aqueous layer was separated. Next, 2.5% aqueous sodium bicarbonate solution (10 L, 10 L/kg) was charged to the rich organic solution and stirred for 30 min. The agitation was stopped and the bottom aqueous layer was separated. Water (10 L, 10 L/kg) was charged to the rich organic solution and stirred for 30 min. The agitation was stopped and the bottom aqueous layer was separated. The rich organic solution was concentrated under reduced pressure (90 mbar and 40 °C jacket temperature) to 7 L/kg volume. Dichloromethane (5 L, 5 L/kg) was charged to the product rich isopropyl acetate solution at 20 °C. Seeds of propyl (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl)carbamate (10 g, 1%) were charged and a thin slurry formed. Heptane (7 L, 7 L/kg) was charged to the above slurry slowly over 1 hr at 25 °C and stirred for another 1 h before cooling 20 °C over 30 min. The resultant slurry was stirred for 4-6 hrs at 20 °C. The slurry was filtered over a laboratory Buchner funnel. The wet cake was washed with a dichloromethane-heptane mixture (10:7 ratio, 12 vol). The wet cake was dried in a vacuum oven at 25 mm Hg vacuum and 50 °C until the residual heptane was <13 wt% in the solid to provide 1.5 kg of propyl (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl) carbamate in 94% yield. The product was a mixture of four amide rotational isomers. ¾ NMR (400 MHz, DMSO-de) δ 10.79 (br s, 1H), 9.96 (m, 1H), 8.07 (br s, 1H), 7.50 (m, 6H), 7.29 (m, 1H), 7.09 (m, 1H), 4.15 (m, 1H), 3.89 (m, 2H), 3.19 (br s, 1H), 3.13 (br s, 2H), 2.90 (m, 1H), 2.44 (m, 1H), 1.97 (m, 3H), 1.82 (m, 3H), 1.50 (m, 3H), 1.26 (m, 5H), 1.09 (m, 7H), 0.85 (m, 4H), 0.70 (m, 2H). 13C NMR (101 MHz, DMSO-de) δ 168.33, 168.32, 164.85, 164.55, 159.38, 159.16, 156.93, 156.69, 154.90, 154.74, 153.14, 150.86, 139, 15, 139.11, 137.96, 137.89, 137.36, 137.23, 135.75, 135.68, 135.64, 134.77, 134.68, 132.57, 132.51, 132.46, 132.42, 131.50, 128.98 (m), 128.26 (m), 127.05, 127.01, 125.99, 125,76, 124.97, 124.83, 124.06, 121.48, 121.40, 121.28, 121.20, 117.90, 117.86, 117.70, 117.65, 115.19, 115.15, 115.12, 115.07, 108.69, 108.65, 106.87, 106.60, 70.39, 66.83, 66.80, 66.73, 45.32, 37.38, 37.15, 31.23, 28.35, 27.05, 26.68, 24.85, 24.61, 22.27, 22.07, 21.84, 21.75, 14.98, 14.93, 14.86, 14.84, 13.87, 10.11, 9.89.

HPLC Analysis: Column: Zorbax Eclipse Plus C18 3.5 um, 150 x 4.6 mm ID;

Solvent A: 10 mM ammonium formate in water-MeOH (90: 10); Solvent B: C¾CN :

MeOH (30:70 v/v); Gradient: % B: 0 Min. 50%; 25 Min. 81 %; 26 Min. 100%; 30 Min. 100%; Stop Time: 30 min; Flow Rate: 1 ml/min; Wavelength: 240 nm. The retention time of propyl (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl) carbamate wasl4.6 min. The retention time of 3-fluoro-2-(methyl(propoxycarbonyl) amino)benzoic acid was 2.6 min. The retention time of (2S)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide was 6.1 min.

Compound 8

6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l ,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide


(8)

To a 1 L round bottom flask with stir bar was added propyl (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl)carbamate (100 g, 148 mmol, 93.5 mass%) followed by MeTHF (500 mL, 4990 mmol, 100 mass%). The mixture was stirred at room temperature for 10 minutes to ensure complete dissolution. Next, 150 mL of MeTHF was added, and an azeotropic distillation to remove water was performed at 50 °C and 70 torr. The KF was measured to be 424 ppm. This solution is termed the “Compound 8 solution.”

To a 2 L Chemglass reactor was charged MeTHF (2000 mL, 19900 mmol, 100 mass%) followed by lithium fert-butoxide (7.9 mL, 7.9 mmol, 1 mol/L). The KF of MeTHF was measured to be 622 ppm. The Compound 8 solution was added dropwise

over 2 hours at room temperature via a Simdos pump. After the addition was complete, the reaction mixture was maintained at temperature for 15 minute.

MeOH (200 mL, 4940 mmol, 100 mass%) was then added to the reactor followed by the addition of acetic acid (0.5 mL, 9 mmol, 100 mass%). The reaction mixture was distilled to 5 volumes of organics (60 mbar pressure, jacket temperature = 40 °C). After the distillation, acetone (150 mL, 2000 mmol, 100 mass%) was added to the thick slurry as the solution warmed to 35 °C. Once at 35 °C, MeOH (550 mL, 13600 mmol, 100 mass%) was charged to the reactor, re-dissolving the batch to provide a yellow solution. The reaction mixture was cooled over 1 hour to 20 °C resulting in crystallization of the product. Ten heat cycles were performed. Starting at 20 °C, the batch was heated to 35 °C over 45 minutes, held at 35 °C for 10 minutes, cooled 20 °C over 60 minutes, and held at 20 °C for 10 minutes. After the heat cycles, the slurry was maintained at room temperature for 1 hour at room temperature. Heptane (1100 mL, 7510 mmol, 100 mass%) was added over 4 hours at 20 °C with agitation via a Simdos pump. After the addition, the slurry aged to 20 °C overnight. The product was isolated by vacuum filtration and washed twice with MeOH (200 mL, 4940 mmol, 100 mass%). The product was dried on a filter with vacuum for 1.5 h to afford 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide at 89.4% corrected yield (80.52g, 6 wt % MeOH, Purity by HPLC: 99.32 AP; Retention time (11.65 min)).

¾ NMR (500MHz, DMSO-de) 10.78 (s, 1H), 8.07 (br. s., 1H), 7.95 (d, J=7.8 Hz, 1H), 7.72 (dd, J=14.2, 8.0 Hz, 1H), 7.56 (d, J=10.8 Hz, 1H), 7.45 (br. s., 1H), 7.42-7.36 (m, 1H), 7.34 (d, J=6.9 Hz, 1H), 7.34-7.31 (m, 1H), 7.29 (dd, J=7.5, 1.3 Hz, 1H), 4.17 (s, 1H), 3.73 (d, J=8.0 Hz, 3H), 2.91 (dd, J=16.8, 4.4 Hz, 1H), 2.48-2.37 (m, 1H), 1.98-1.89 (m, 2H), 1.87 (d, J=11.0 Hz, 1H), 1.76 (s, 3H), 1.59 (td, J=l 1.5, 4.1 Hz, 1H), 1.20-1.12 (m, 1H), 1.11 (s, 6H).

13C NMR (126MHz, DMSO-de) 168.2 (d, J=1.8 Hz, 1C), 160.1 (d, J=3.6 Hz, 1C), 151.9 (d, J=228.9 Hz, 1C), 150.5 (d, J=41.8 Hz, 1C), 148.7 (d, J=205.3 Hz, 1C), 139.2, 135.1, 135.0, 134.8, 131.4, 130.6, 130.0 (d, J=7.3 Hz, 1C), 128.5, 127.1 (d, J=4.5 Hz, 1C), 125.7, 124.3 (d, J=2.7 Hz, 1C), 123.6 (d, J=8.2 Hz, 1C), 123.0 (d, J=23.6 Hz, 1C), 120.8 (d, J=20.0 Hz, 1C), 118.4, 115.3 (d, J=7.3 Hz, 1C), 108.8 (d, J=5.4 Hz, 1C), 106.7 (d, J=28.2 Hz, 1C), 70.4, 45.4, 34.3 (d, J=14.5 Hz, 1C), 27.1, 26.8, 24.8, 24.7, 22.1, 14.5.

HPLC Analysis: Column: Chiralcel OX-3R 3um 4.6 x 150 mm; Oven

Temperature: 50 °C; Solvent A: 0.05%TFA Water/ ACN (95:5); Solvent B: 0.05%TFA Water/ ACN (5:95); Gradient % B: 0 Min. 0%; 7 Min. 55%; 11 Min. 55%; 14 Min. 100%; Stop Time: 17 Min.; Flow Rate: 1.5 ml/min; wavelength: 225 nm. (2-((3-((2S)-8-carbamoyl-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazol-5-yl)-2-methylphenyl)carbamoyl)-6-fluorophenyl)(methyl)carbamate: 0.00 AP (9.85 min).

Alternative Preparation of Compound 8

To a 2.5 L Chemglass reactor with agitator were added 2-Me-THF (162.4 g, 1885 mmol, 100 mass%, 189 mL, 11.83) and DMF (179.5 g, 2456 mmol, 100 mass%, 190 mL, 15.41), followed by the addition of (2S)-5-(3-amino-2-methylphenyl)-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (63.03 g, 63.03 mL, 159.4 mmol, 63.03 g), 3-fluoro-2-(methyl(propoxycarbonyl)amino)benzoic acid (44.77 g, 44.77 mL, 175.4 mmol, 44.77 g), and 1 -Me-Imidazole (16.99 g, 16.48 mL, 206.9 mmol, 16.99 g). With agitation, MSA (7.66 g, 5.23 mL, 79.7 mmol, 7.66 g) was added at -20 °C, and a slight exotherm to 26 °C was observed. The reaction mixture was cooled to 10 °C and ED AC (42.73 g, 42.73 mL, 222.9 mmol, 42.73 g) was added as a solid followed by a DMF rinse (60.4 g, 63.9 mL, 826 mmol, 60.4 g). The reaction mixture was aged overnight at 10 °C with agitation. An aliquot was taken and subjected to HPLC analysis to confirm reaction completion.

The batch temperature was increased to 15 °C, and 2-Me-THF (923.96 g, 10727 mmol, 100 mass%, 1080 mL, 67.31) was charged to the reactor, followed by a saturated aqueous brine solution (158 mL, 835.8 mmol, 26 mass%, 158 mL, 5.244) and an aqueous 2.0 M HCl solution (78 mL, 78 mmol, 1.0 mol/L, 78 mL, 0.49). The batch temperature was then increased to 20 °C. The biphasic mixture was agitated for 15 min and allowed to settle for 5 min. An saturated aqueous brine solution (157 mL, 830.5 mmol, 26 mass%, 157 mL, 5.211) and an aqueous 2.0 M HCl solution (78 mL, 78 mmol, 1.0 mol/L, 78 mL, 0.49) were then added to the reactor. The biphasic mixture was agitated for 15 min, allowed to settle for 5 min, and the aqueous layer was removed. Water (634.6 g, 35230 mmol, 100 mass%, 634.6 mL, 221.0) was then added to the reactor. The biphasic mixture was agitated for 15 min, allowed to settle for 5 min, and the aqueous layer was removed. Next, 10 w/w% aqueous NaHCC solution (164.2 g, 97.73 mmol, 5 mass%,

158.2 mL, 0.6132) and water (476.3 g, 26440 mmol, 100 mass%, 476.3 mL, 165.9) were added to the reactor. The biphasic mixture was agitated for 15 min, settled for 5 min, and the aqueous layer was removed. A saturated aqueous brine solution (752.9 g, 3349 mmol, 26 mass%, 633.2 mL, 21.02) was then added to the reactor. The biphasic mixture was agitated for 30 min, allowed to settle for 5 min, and the aqueous layer was removed.

The organic stream was distilled to 6 volumes (380 mL) at a pressure of 200 mbar, a jacket temperature of 60 °C, and a batch temperature of -35 °C. 2-Me-THF (765 g, 8881.6 mmol, 100 mass%, 891 mL, 55.73) was charged to the reactor. The organic solution was distilled to 6 volumes (380 mL) at a pressure of 200 mbar, a jacket temperature of 60 °C, and a batch temperature of -35 °C. 2-Me-THF (268.5 g, 3117 mmol, 100 mass%, 313 mL, 19.56) was charged to the reactor. The organic solution was distilled to 6 volumes (380 mL) at a pressure of 200 mbar, a jacket temperature of 60 °C, and a batch temperature of -35 °C. The concentrated stream was polish filtered through a 0.4 μιη PTFE filter. The reactor was rinsed with 2-Me-THF (134.6 g, 1563 mmol, 100 mass%, 157 mL, 9.806) and the rinse was passed through the PTFE filter. This solution was termed “organic solution.”

To a clean, dry, 2.5 L Chemglass reactor were added LiOtBu 1.0 M in THF (9.91 g, 11.2 mmol, 1 mol/L, 11.2 mL, 0.0700) and 2-Me-THF (1633.3 g, 18963 mmol, 100 mass%, 1900 mL, 119.0). The organic solution was charged to the reactor, with agitation, over 2 hours (at a rate of -100 mL/h) via a sim-dos pump. The reaction mixture was aged 10 minutes upon completion of the addition. An aliquot was taken and subjected to HPLC analysis to confirm reaction completion.

Acetic acid (1.03 g, 17.2 mmol, 100 mass%, 0.983 mL, 0.108) and methanol (150 g, 4681.41 mmol, 100 mass%, 189 mL, 29.37) were charged to the reactor. The organic stream was distilled to 16.5 vol Me-THF. Acetone (638.4 g, 10990 mmol, 100 mass%, 810 mL, 68.97) was added to the reactor and the organic stream was distilled to 9 vol at a pressure of 100 mbar and ajacket temperatures of less than 40 °C. The organic stream was heated to 35 °C, and methanol (400 g, 12483.8 mmol, 100 mass%, 505 mL, 78.33) was added. The stream was cooled to 20 °C to induce crystallization.

Heat cycles were performed for -15 h by heating the batch to 35 °C over 20 min, holding for 10 min, cooling to 20 °C over 20 min, and holding 10 min. After the heat cycles, heptane (686 g, 6846.10 mmol, 100 mass%, 1000 mL, 42.96) was added over 4 hours via a sim-dos pump. The slurry was aged for 2 h. The product was filtered, washed with methanol (152.2 g, 4750 mmol, 100 mass%, 192 mL, 29.81) to afford 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l -methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (68.4 g, 1 19 mmol, 100 mass%, 75.0% Yield, 68.4 mL, 0.750).

Comparative Process Disclosed in US 9,334,290

Intermediates 25 and 26

(R)-5-Bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- carboxamide (1-25), and

(S)-5-Bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8- -26)

A sample of racemic 5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide [Intermediate 24] was separated by chiral supercritical fluid chromatography as follows: column: CHIRALPAK® OD-H (3 x 25 cm, 5μηι); Mobile Phase: CC -MeOH (70:30) at 150 mL/min, 40 °C. The first peak eluting from the column provided (R)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide [Intermediate 25]. The second peak eluting from the column provided (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide [Intermediate 26]. The mass spectra and ¾ NMR spectra of the two enantiomers were the same. Mass spectrum m/z 369, 371 (M+H)+. ¾ NMR (500 MHz, DMSO-de) δ 10.96 (s, 1H), 8.07 (br. s., 1H), 7.55 (d, J=10.3 Hz, 1H), 7.50 (br. s., 1H), 4.24 (s, 1H), 3.26 (dd, J=15.8, 4.4 Hz, 1H), 2.93 (dd, J=17.1, 4.6 Hz, 1H), 2.72 (t, J=11.7 Hz, 1H), 2.48-2.40 (m, 1H), 2.12 (d, J=9.2 Hz, 1H), 1.70-1.62 (m, 1H), and 1.32 (qd, J=12.4, 5.3 Hz, 1H).

Alternative SFC Separation to Give Intermediate 26:

CHIRALPAK® AD-H (3 x 25 cm, 5 μηι); Mobile Phase: C02-MeOH (55:45) at

150 mL/min, 40 °C. The first peak eluting from the column provided (S)-5-bromo-6- fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxarnide

[Intermediate 26]. The second peak eluting from the column provided (R)-5-bromo-6- fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxarnide

[Intermediate 25].

Example 28

6-Fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-

Following the procedure used to prepare Example 27, (S)-5-bromo-6-fluoro-2-(2- hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (single enantiomer) [Intermediate 26] (0.045 g, 0.122 mmol) and 8-fluoro-l-methyl-3-(S)-(2-methyl-3- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)quinazoline-2,4(lH,3H)-dione

[Intermediate 10] (0.065 g, 0.158 mmol) were converted into 6-fluoro-5-(3-(S)-(8-fluoro- l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2- hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide (mixture of two atropisomers) as a yellow solid (0.035 g, 49% yield). Separation of a sample of this material by chiral super-critical fluid chromatography, using the conditions used to separate Example 27, provided (as the first peak to elute from the column) 6-fluoro-5-(R)-(3-(S)-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxarnide. The chiral purity was determined to be greater than 99.5%. The relative and absolute configurations were determined by x-ray crystallography. Mass spectrum m/z 573 (M+H)+. ¾ NMR (500 MHz, DMSO-de) δ 10.77 (s, 1H), 8.05 (br. s., 1H), 7.94 (dd, J=7.9, 1.2 Hz, 1H), 7.56-7.52 (m, 1H), 7.43 (br. s., 1H), 7.40-7.36 (m, 1H), 7.35-7.30 (m, 2H), 7.28 (dd, J=7.5, 1.4 Hz, 1H), 4.15 (s, 1H), 3.75-3.70 (m, 3H), 2.90 (dd, J=16.8, 4.6 Hz, 1H), 2.47-2.39 (m, 1H), 1.93-1.82 (m, 3H), 1.74 (s, 3H), 1.57 (td, J=l 1.7, 4.2 Hz, 1H), 1.16-1.11 (m, 1H), and 1.10 (d, J=1.9 Hz, 6H). [a]D: +63.8° (c 2.1, CHCh). DSC melting point onset temperature = 202.9 °C (heating rate = 10 °C/min.).

Alternative Synthesis of Example 28:

A mixture of (S)-5-bromo-6-fluoro-2-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide [Intermediate 26] (5.00 g, 13.54 mmol), 8-fluoro-l-methyl-3-(S)-(2-methyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)quinazoline-2,4(lH,3H)-dione [Intermediate 10] (6.67 g, 16.25 mmol), tripotassium phosphate (2 M in water) (20.31 mL, 40.6 mmol), and tetrahydrofuran (25 mL) was subjected to 3 evacuate-fill cycles with nitrogen. The mixture was treated with l,l’-bis(di-fert-butylphosphino)ferrocene palladium dichloride (0.441 g, 0.677 mmol) and the mixture was subjected to 2 more evacuate-fill cycles with nitrogen. The mixture was stirred at room temperature overnight, then was diluted with EtOAc, washed sequentially with water and brine, and dried and concentrated. The residue was purified by column chromatography on silica gel, eluting with EtOAc-hexanes (sequentially 50%, 62%, 75% and 85%), to provide 6-fluoro-5-(3-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3-(S)-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide as a white solid (6.58 g, 85% yield).

Material prepared by this method (40.03 g, 69.9 mmol) was separated by chiral super-critical fluid chromatography to give (2S, 5R)-6-fluoro-5-(3-(8-fluoro-l-methyl-2,4-dioxo-l,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-lH-carbazole-8-carboxamide. Further purification was achieved

by suspending this material in methanol, sonicating for 5 min, collection of the solid by filtration, rinsing the collected solid with methanol and drying at room temperature under reduced pressure to give a white solid (22.0 g, 90% yield).

REFERENCES

1: Watterson SH, De Lucca GV, Shi Q, Langevine CM, Liu Q, Batt DG, Beaudoin Bertrand M, Gong H, Dai J, Yip S, Li P, Sun D, Wu DR, Wang C, Zhang Y, Traeger SC, Pattoli MA, Skala S, Cheng L, Obermeier MT, Vickery R, Discenza LN, D’Arienzo CJ, Zhang Y, Heimrich E, Gillooly KM, Taylor TL, Pulicicchio C, McIntyre KW, Galella MA, Tebben AJ, Muckelbauer JK, Chang C, Rampulla R, Mathur A, Salter-Cid L, Barrish JC, Carter PH, Fura A, Burke JR, Tino JA. Discovery of 6-Fluoro-5-(R)-(3-(S)-(8-fluoro-1-methyl-2,4-dioxo-1,2-dihydroquinazolin-3(4H)-yl )-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-1H-carbazole-8-carboxamide (BMS-986142): A Reversible Inhibitor of Bruton’s Tyrosine Kinase (BTK) Conformationally Constrained by Two Locked Atropisomers. J Med Chem. 2016 Oct 13;59(19):9173-9200. PubMed PMID: 27583770.

(a) Watterson, S. H.De Lucca, G. V.Shi, Q.Langevine, C. M.Liu, Q.Batt, D. G.Bertrand, M. B.Gong, H.Dai, J.Yip, S.Li, P.Sun, D.Wu, D.-R.Wang, C.Zhang, Y.Traeger, S. C.Pattoli, M. A.Skala, S.Cheng, L.Obermeier, M. T.Vickery, R.Discenza, L. N.D’Arienzo, C. J.Zhang, Y.Heimrich, E.Gillooly, K. M.Taylor, T. L.Pulicicchio, C.McIntyre, K. W.Galella, M. A.Tebben, A. J.Muckelbauer, J. K.Chang, C.Rampulla, R.Mathur, A.Salter-Cid, L.Barrish, J. C.Carter, P. H.Fura, A.Burke, J. R.Tino, J. A. Discovery of 6-Fluoro-5-(R)-(3-(S)-(8-fluoro-1-methyl-2,4-dioxo-1,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-1H-carbazole-8-carboxamide (BMS-986142): A Reversible Inhibitor of Bruton’s Tyrosine Kinase (BTK) Conformationally Constrained by Two Locked AtropisomersJ. Med. Chem. 2016599173DOI: 10.1021/acs.jmedchem.6b01088
(b) De Lucca, G. V.Shi, Q.Liu, Q.Batt, D. G.Bertrand, M. B.Rampulla, R.Mathur, A.Discenza, L.D’Arienzo, C.Dai, J.Obermeier, M.Vickery, R.Zhang, Y.Yang, Z.Marathe, P.Tebben, A. J.Muckelbauer, J. K.Chang, C. J.Zhang, H.Gillooly, K.Taylor, T.Pattoli, M. A.Skala, S.Kukral, D. W.McIntyre, K. W.Salter-Cid, L.Fura, A.Burke, J. R.Barrish, J. C.Carter, P. H.Tino, J. A. Small Molecule Reversible Inhibitors of Bruton’s Tyrosine Kinase (BTK): Structure–Activity Relationships Leading to the Identification of 7-(2-Hydroxypropan-2-yl)-4-[2-methyl-3-(4-oxo-3,4-dihydroquinazolin-3-yl)phenyl]-9H-carbazole-1-carboxamide (BMS-935177)J. Med. Chem. 2016597915DOI: 10.1021/acs.jmedchem.6b00722
Watterson, S.H.; De Lucca, G.V.; Shi, Q.; et al.
Twisted road to the discovery of BMS-986142: Using conformationally locked atropisomers to drive potency in a reversible inhibitor of Brutonas tyrosine kinase (BTK)
255th Am Chem Soc (ACS) Natl Meet (March 18-22, New Orleans) 2018, Abst MEDI 6

////////////BMS-986142, BMS 986142, BMS986142,  phase II,  clinical development,  Bristol-Myers Squibb, rheumatoid arthritis, primary Sjogren’s syndrome,

CN1C(=O)N(C(=O)c2cccc(F)c12)c3cccc(c3C)c4c(F)cc(C(=O)N)c5[nH]c6C[C@H](CCc6c45)C(C)(C)O

BMS-986195


img
BMS-986195
  • Molecular FormulaC20H23FN4O2
  • Average mass370.421 Da
  • CAS: 1912445-55-6
1H-Indole-7-carboxamide, 5-fluoro-2,3-dimethyl-4-[(3S)-3-[(1-oxo-2-butyn-1-yl)amino]-1-piperidinyl]-
4-[(3S)-3-(2-Butynoylamino)-1-piperidinyl]-5-fluor-2,3-dimethyl-1H-indol-7-carboxamid
(S)-4-(3-(2-Butynoylamino)piperidin-1-yl)-5-fluoro-2,3-dimethyl-1H-indole-7-carboxamide
(S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimeth -lH-indole-7-carboxamide
  • Originator Bristol-Myers Squibb
  • Class Anti-inflammatories; Antirheumatics
  • Mechanism of Action Agammaglobulinaemia tyrosine kinase inhibitors

Highest Development Phases

  • Phase I Rheumatoid arthritis

Most Recent Events

  • 30 Jan 2018 Bristol-Myers Squibb completes a phase I trial in Rheumatoid arthritis (In volunteers, In adults, Combination therapy) in USA (PO) (NCT03262740)
  • 10 Nov 2017 Bristol-Myers Squibb completes a phase I drug-drug interaction trial in Healthy volunteers (NCT03131973)
  • 03 Nov 2017 Safety, pharmacokinetic, and pharmacodynamic data from a pharmacokinetic trial in healthy volunteers presented at the 81st American College of Rheumatology and the 52nd Association of Rheumatology Health Professionals Annual Scientific Meeting (ACR/ARHP-2017)
  • Image result for BMS-986195

BMS-986195 is a potent, covalent, irreversible inhibitor of Bruton’s tyrosine kinase (BTK), a member of the Tec family of non-receptor tyrosine kinases essential in antigen-dependent B-cell signaling and function. BMS-986195 is more than 5000-fold selective for BTK over all kinases outside of the Tec family, and selectivity ranges from 9- to 1010-fold within the Tec family. BMS-986195 inactivated BTK in human whole blood with a rapid rate of inactivation (3.5×10-4 nM-1·min-1) and potently inhibited antigen-dependent interleukin-6 production, CD86 expression and proliferation in B cells (IC50 <1 nM) without effect on antigen-independent measures in the same cells.

Bristol-Myers Squibb is developing BMS-986195, an oral candidate for the treatment of rheumatoid arthritis. A phase I clinical trial in healthy adult volunteers is ongoing.

Image result

Structure of BMS986195.
Credit: Tien Nguyen/C&EN

Presented by: Scott H. Watterson, principal scientist at Bristol-Myers Squibb

Target: Bruton’s tyrosine kinase (BTK)

Disease: Autoimmune diseases such as rheumatoid arthritis

Reporter’s notes: Completing another set of back-to-back presentations on the same target, Watterson revealed another BTK inhibitor also in Phase II clinical trials. Chemists made BMS-986195 in seven steps, and the molecule showed high levels of BTK inactivation in mice. The team aimed to develop an effective compound that required low doses and that had low metabolic degradation.

Patent

WO 2016065226

Inventor Saleem AhmadJoseph A. TinoJohn E. MacorAndrew J. TebbenHua GongQingjie LiuDouglas G. BattKhehyong NguScott Hunter WattersonWeiwei GuoBertrand Myra Beaudoin

Original Assignee Bristol-Myers Squibb Company

https://patents.google.com/patent/WO2016065226A1/en

PATENT

WO 2018045157

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=E81EF2BDB127473D100AAA55455FC42B.wapp1nA?docId=WO2018045157&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

otein kinases, the largest family of human enzymes, encompass well over 500 proteins. Btk is a member of the Tec family of tyrosine kinases, and is a regulator of early B-cell development, as well as mature B-cell activation, signaling, and survival.

B-cell signaling through the B-cell receptor (BCR) leads to a wide range of biological outputs, which in turn depend on the developmental stage of the B-cell. The magnitude and duration of BCR signals must be precisely regulated. Aberrant BCR-mediated signaling can cause dysregulated B-cell activation and/or the formation of pathogenic auto-antibodies leading to multiple autoimmune and/or inflammatory diseases. Mutation of Btk in humans results in X-linked agammaglobulinaemia (XLA). This disease is associated with the impaired maturation of B-cells, diminished immunoglobulin production, compromised T-cell-independent immune responses and marked attenuation of the sustained calcium signal upon BCR stimulation.

Evidence for the role of Btk in allergic disorders and/or autoimmune disease and/or inflammatory disease has been established in Btk-deficient mouse models. For example, in standard murine preclinical models of systemic lupus erythematosus (SLE), Btk deficiency has been shown to result in a marked amelioration of disease progression. Moreover, Btk deficient mice are also resistant to developing collagen-induced arthritis and are less susceptible to Staphylococcus-induced arthritis.

A large body of evidence supports the role of B-cells and the humoral immune system in the pathogenesis of autoimmune and/or inflammatory diseases. Protein-based therapeutics (such as Rituxan) developed to deplete B-cells, represent an important approach to the treatment of a number of autoimmune and/or inflammatory diseases.

Because of Btk’s role in B-cell activation, inhibitors of Btk can be useful as inhibitors of B-cell mediated pathogenic activity (such as autoantibody production).

Btk is also expressed in mast cells and monocytes and has been shown to be important for the function of these cells. For example, Btk deficiency in mice is associated with impaired IgE -mediated mast cell activation (marked diminution of T F-alpha and other inflammatory cytokine release), and Btk deficiency in humans is associated with greatly reduced TNF-alpha production by activated monocytes.

Thus, inhibition of Btk activity can be useful for the treatment of allergic disorders and/or autoimmune and/or inflammatory diseases including, but not limited to: SLE, rheumatoid arthritis, multiple vasculitides, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis, allergic rhinitis, multiple sclerosis (MS), transplant rejection, type I diabetes, membranous nephritis, inflammatory bowel disease, autoimmune hemolytic anemia, autoimmune thyroiditis, cold and warm agglutinin diseases, Evan’s syndrome, hemolytic uremic syndrome/thrombotic thrombocytopenic purpura (HUS/TTP), sarcoidosis, Sjogren’s syndrome, peripheral neuropathies (e.g., Guillain-Barre syndrome), pemphigus vulgaris, and asthma.

In addition, Btk has been reported to play a role in controlling B-cell survival in certain B-cell cancers. For example, Btk has been shown to be important for the survival of BCR-Abl-positive B-cell acute lymphoblastic leukemia cells. Thus inhibition of Btk activity can be useful for the treatment of B-cell lymphoma and leukemia.

In view of the numerous conditions that are contemplated to benefit by treatment involving modulation of protein kinases, it is immediately apparent that new compounds capable of modulating protein kinases such as Btk and methods of using these compounds should provide substantial therapeutic benefits to a wide variety of patients.

WO 2016/065226 discloses indole carboxamide compounds useful as Btk inhibitors, including (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide (Example 223), which has the structure:

Also disclosed is multistep synthesis process for preparing (S)-4-(3-(but-2-ynamido) piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide.

There are difficulties associated with the adaptation of the multistep synthesis disclosed in WO 2016/065226 to larger scale synthesis, such as production in a pilot plant or a manufacturing plant for commercial production. Further, there is a continuing need to find a process that has few synthesis steps, provides higher yields, and/or generates less waste.

Applicants have discovered a new synthesis process for the preparation of (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide that has fewer synthesis steps and/or provides higher yields than the process disclosed in WO 2016/065226. Furthermore, this process contains no metal-catalyzed steps, no genotoxic intermediates, and is adaptable to large scale manufacturing.

EXAMPLE 1

(S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide

Step 1 : Preparation of Methyl (S)-2-amino-4-(3-((tert-butoxycarbonyl)amino)piperidin-l-yl)-5-fluorobenz

To a 250 mL ChemGlass reactor were charged methyl 2-amino-4,5-difluoro-benzoate (11.21 g, 59.90 mmol), tert-butyl N-[(3S)-3-piperidyl]carbamate (10 g, 49.930 mmol), potassium phosphate, dibasic (10.44 g, 59.94 mmol), and dimethyl sulfoxide (100 mL, 1400 mmol). The resulting thin slurry was heated to 95 to 100 °C and agitated at this temperature for 25 hours. The mixture was cooled to 50 °C. Methanol (100 mL) was added and followed by slow addition of water (50 mL). The mixture was aged at 50 °C for 30 minutes to result in a thick white slurry. Additional water (150 mL) was slowly charged to the above mixture and agitated at 50 °C for 1 hour. The slurry was cooled to 20 °C in 1 hour and aged at this temperature for 4 hours. The slurry was filtrated. The wet cake washed with 25% MeOH in water (30 mL), water (100 mL) and dried under vacuum at 60 °C for 24 h. Methyl (S)-2-amino-4-(3-((tert-butoxycarbonyl)amino) piperidin-l-yl)-5-fluorobenzoate was obtained as a white solid (7 g, yield: 72.5%). ¾ MR (400MHz, METHANOLS) δ 7.34 (d, J=14.6 Hz, 1H), 6.27 (d, J=7.3 Hz, 1H), 3.83-3.71 (s, 3H), 3.68-3.57 (m., 1H), 3.50 -3.40 (m 1H), 3.39 -3.31 (m, 1H), 3.31-3.26 (m, 1H), 2.86-2.70 (m, 1H), 2.64 (t, J=10.0 Hz, 1H), 1.97-1.84 (m, 1H), 1.84-1.74 (m, 1H), 1.73-1.61 (m, 1H), 1.44 (s, 9H), 1.38 (m, 1H). LC-MS [M+H] 368.

Step 2: Preparation of Methyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate

To a reactor were charged methyl (S)-2-amino-4-(3-((tert-butoxycarbonyl)amino) piperidin-l-yl)-5-fluorobenzoate (5.0 g), DPPOH (diphenyl phosphate, 6.81 g, 2 eq) and 3-hydroxybutanone (1.2 eq, 1.44 g), followed by addition of isopropyl acetate (100 mL, 20 mL/g). The mixture was allowed to warm up to 70 to 75 °C, resulting in a yellow solution. The solution was stirred at 70 to 75 °C for 30 h to complete the cyclization.

Water (2 mL) was added and the mixture was aged at 70 °C over 24 h to remove the Boc group. The mixture was cooled to room temperature. Next, aqueous 20% K3PO4 solution (50 mL) was added and the mixture was stirred for 15 min. The organic layer was separated and washed with water (50 mL). The organic layer was then concentrated under vacuum (200 Torr) to -50 mL. The resulting slurry was stirred at 50 °C for 2 h and then heptane (100 mL) was added over 1 h. The mixture was cooled to room

temperature, stirred for 20 h, and then filtered. The cake was washed with heptane (50 mL). Methyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate, DPPOH salt was obtained as a light yellow solid. The wet-cake was added to a reactor. Isopropyl acetate (100 mL) was added, followed by addition of aqueous K3PO4 solution (4 g in water 50 mL). The mixture was stirred at room temperature for -half-hour, resulting in a two phase clear solution (pH >10 for aqueous). The organic layer was separated and washed with water (50 mL), and then concentrated under vacuum to a volume of 15 mL. The resulting slurry was stirred at room temperature for 4 h, then heptane (75 mL) was added over 1 h. The mixture was aged at room temperature for 24 h, then concentrated to a volume to -50 mL. The slurry was filtered. The cake was washed with heptane 20 mL and dried under vacuum at 50 °C for 24 h. Methyl (S)-4-(3- aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate was obtained as a light yellow solid (2.76 g, yield: 69%). ¾ NMR (400MHz, DMSO-d6) δ 10.64 (s, 1H), 7.33 (d, J=13.7 Hz, 1H), 3.89 (s, 3H), 3.14 (br. m., 1H), 3.07-2.90 (m, 2H), 2.84 (br. m., 1H), 2.70 (br. m., 1H), 2.35 (s, 3H), 2.33 (s, 3H), 1.87 (br. m., 1H), 1.67 (br. m., 3H). LC-MS: M+H= 320.

Alternative Preparation

Step 2: Preparation of ethyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate trifluoroacetic acid salt

To a reactor were charged ethyl (S)-2-amino-4-(3-((tert-butoxycarbonyl)amino) piperidin-l-yl)-5-fluorobenzoate (1.0 g, limiting reagent), DPPOH (diphenyl phosphate, 1.97 g, 3.0 eq) and 3-hydroxybutanone (1.4 eq, 0.32 g), followed by addition of toluene (20 mL, 20 mL/g). The mixture was allowed to warm up to 80-90 °C, resulting in a yellow solution. The solution was stirred at 80-90 °C for 10 h to complete the

cyclization. Water (0.4 mL, 0.4 ml/g) was added and the mixture was aged at 80-90 °C for 8 hours. The mixture was cooled to room temperature. Next, aqueous 20% K3PO4 solution (15 mL, 15 mL/g) was added and the mixture was stirred for 0.5 hour. The organic layer was separated and the aqueous layer was washed with toluene (7.5 mL, 7.5 mL/g). To combined organic layers water (10 mL, 10 mL/g) was added and the mixture was stirred for 0.5 hour. The organic layer was separated. To the organic layer water (10 mL, 10 mL/g) was added and the mixture was stirred for 0.5 hour. The organic layer was separated. The organic layer was concentrated under vacuum (100 Torr) to 8 mL (8 ml/g). Following concentration the reaction mixture was cooled to 20-25 °C and MTBE (20 mL, 20 mL/g) was added. Trifluoroacetic acid (1.2 eq., 0.36 g) was slowly added to make the salt maintaining temperature at 20-25 °C. The resulting slurry was aged for 4 hours and then filtered. The filtered solids are washed with MTBE (8 mL, 8 mL/g) and the cake

was dried under vacuum at 50 °C. (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate trifluoroacetic acid salt was obtained as a white to tan crystalline material (85% yield, 1.0 g). ¾ NMR (400 MHz, DMSO-d6) δ 10.74 (s, 1H), 8.16-7.88 (m, 2H), 7.37 (d, 7=13.6 Hz, 1H), 4.38 (q, 7=7.1 Hz, 2H), 3.18-3.01 (m, 3H), 2.96 (br s, 1H), 2.35 (s, 6H), 2.30 (s, 1H), 2.12 (br d, 7=9.3 Hz, 1H), 1.78 (br s, 2H), 1.45-1.31 (m, 4H), 1.10 (s, 1H). 13C NMR (101 MHz, DMSO-d6) δ 165.1, 165.1, 158.4, 158.1, 135.4, 134.7, 134.6, 132.2, 128.8, 128.2, 126.9, 126.8, 118.7, 115.7, 110.6, 110.3,108.7, 108.6, 106.6, 106.5, 83.5, 79.8, 60.5, 54.9, 51.7, 48.7, 47.2, 28.4, 26.8, 23.6, 14.2, 11.1, 10.2

Step 3A: Preparation of (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide

A 40 mL vial was charged with methyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate (1.5 g, 4.70 mmol), followed by the addition of N,N-dimethylformamide (12.0 mL, 8.0 mL/g). The vial was purged with N2. Formamide (1.49 mL, 37.6 mmol) was added followed by sodium methoxide solution in methanol (35 wt%, 1.29 mL, 3.76 mmol). The resulting solution was heated at 50 °C over 8 hours. The reaction mixture was cooled down to room temperature and the reaction was quenched with water (12.0 mL, 8.0 mL/g). 2-methyltetrahydrofuran (30 mL, 20 mL/g) was added to the mixture. The mixture was shaken vigorously. The layers were separated and the aqueous layer was extracted with 2-methyltetrahydrofuran (15 mL, 10 mL/g) two more times. Organic extracts were then washed with brine and water (15 mL each, 10 mL/g). The organic layer was evaporated. Solids were dried in vacuo at 60 °C to afford (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide as a yellow solid (1.04 g, 69% yield). ¾ NMR (500MHz, DMSO-d6) δ 10.60 (br. s.,

1H), 7.91 (br. s., 1H), 7.40 (d, 7=14.0 Hz, 1H), 7.32 (br. s., 1H), 3.10 (br. s., 1H), 2.98 (br. s., 2H), 2.82 (br. s., 1H), 2.68 (br. s., 1H), 2.34 (br. s., 3H), 2.30 (br. s., 3H), 1.88 (br. s., 1H), 1.67 (br. s., 2H), 1.45 (br. s., 2H), 1.05 (br. s., 1H). LCMS [M+H] 305.24.

Step 3B: Alternative Preparation of (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide

A 100 mL Hastelloy high pressure EasyMax reactor was charged with methyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate (1.5 g, 4.70 mmol), followed by addition of 7 N ammonia solution in methanol (45.0 mL, 30.0 mL/g) followed by addition of l,3,4,6,7,8-hexahydro-2H-pyrimido[l,2-a]pyrimidine (1.33 g, 9.39 mmol). The reactor was sealed and purged with N2 three times. The reactor was then heated to 80 °C for 24 hrs. The reaction mixture was cooled to room temperature and the vessel contents were purged with N2 three times. Volatiles were concentrated to ~6 mL (4 mL/g) and water (24 mL, 16 mL/g) was added. The yellow precipitate was collected and filtered. The precipitate was washed with methanol/water mixture (20:80 v/v, 6 mL, 4 mL/g), and then water (18 mL, 12 mL/g). The solids were dried in vacuo at 60 °C to afford (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide as a yellow crystalline material (0.93 g, 62% yield). ¾ MR (500MHz, DMSO-de) δ 10.60 (br. s., 1H), 7.91 (br. s., 1H), 7.40 (d, J=14.0 Hz, 1H), 7.32 (br. s., 1H), 3.10 (br. s., 1H), 2.98 (br. s., 2H), 2.82 (br. s., 1H), 2.68 (br. s., 1H), 2.34 (br. s., 3H), 2.30 (br. s., 3H), 1.88 (br. s., 1H), 1.67 (br. s., 2H), 1.45 (br. s., 2H), 1.05 (br. s., 1H). LCMS [M+H] 305.24.

Alternative Preparation:

Step 3C: Preparation of (,S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide 2-butynoic acid salt

Ethyl (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxylate trifluoroacetic acid salt (1.0 g, limiting reagent) and formamide (5 mL, 5 mL/g) were added to a nitrogen inerted reactor. The temperature was maintained at 20-25 °C. To the reactor was added a solution of 20 wt% potassium t-butoxide in THF. The reaction mixture was allowed to sit for 6 hours. To reaction mixture was added Me-THF (15 mL, 15 mL/g) and 12.5 wt % aqueous NaCl (5 mL, 5 mL/g). The reaction mixture was stirred for 0.5 hour. The organic layer was separated, 5 wt% aqueous NaCl (1 mL, 1 mL/g) and 0.25 N aqueous NaOH (4 mL, 4 mL/g) were added, and then stirred for 0.5 hour. The organic layer was separated and 5 wt% aqueous NaCl (5 mL, 5 mL/g) was added, the mixture was stirred for 0.5 hour, and organic phase was separated. The rich organic phase was dried distillation at a pressure of 100 mtorr with Me-THF to obtain KF in 1.5-4wt% range at 5 mL Me-THF volume. The volume was adjusted to 15 mL Me-THF by adding Me-THF (10 mL, 10 mL/g) and EtOH (4 mL, 4 mL/g). Next, 2-butynoic acid (1.0 eq., 0.19 g) was added and the mixture was agitated for 10 hrs. The resulting slurry was filtered. The cake was washed with Me-THF (10 mL, 10 mL/g) and dried under vacuum at 75 °C to afford (,S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide 2-butynoic acid salt (0.7 g, 80% yield) as white crystalline powder. ¾ NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 7.98 (br s, 1H), 7.50-7.32 (m, 2H), 3.32 (br d, J=8.6 Hz, 2H), 3.21 (br t, J=10.5 Hz, 1H), 3.13-2.89 (m, 3H), 2.32 (d, J=5.1 Hz, 5H), 2.11 (br d, J=10.9 Hz, 1H), 1.81-1.67 (m, 4H), 1.55-1.28 (m, 1H).

Step 4A: Preparation of (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide

To Reactor-1 was charged N,N-dimethylformamide (DMF, 12.77 kg, 13.5 L). Reactor-1 was purged with N2 to inert. (S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide (3.0 kg, 1.0 equiv) was charged followed by 2-butynoic acid (0.854 kg, 1.04 equiv). Reactor-1 was rinsed with DMF (1.42 kg, 1.5 L). The mixture was sparged with N2 for 20 min. Triethylamine (2.99 kg, 3.0 equiv) was charged followed by a DMF rinse (1.42 kg, 1.5 L). TBTU (O-(Benzotriazol-l-yl)-N,N,N’,N’-tetramethyluronium tetrafluorob orate, 3.256 kg, 1.04 equiv) was charged followed by a DMF rinse (1.42 kg, 1.5 L). The reaction mixture was agitated for 1.5 h at 20 °C. MeTHF (46.44 kg, 60 L) was charged to the batch. The reaction was quenched with LiCl (20 wt%, 26.76 kg, 24 L) at 20 °C. The bottom aqueous layer was discharged as waste. The organic layer was washed with 2N HCl solution (24.48 kg, 24 L), 10 wt% sodium bicarbonate solution (25.44 kg, 24 L) and deionized water (24.0 kg, 24 L). THF (26.61 kg, 30 L) was charged into Reactor-1. The rich organic stream in MeTHF/TFIF was polish filtered. The stream was distilled down to 15 L at 75-100 Torn Constant volume distillation was carried out at 15 L with THF feed (39.92 kg, 45 L). The stream was heated to 60 °C for 1 hr and cooled to 50 °C. MTBE (33.30 kg, 45 L) was charged slowly over 2 h. The slurry was aged at 50 °C for 4 h and cooled to 20 °C over 2 h, and aged at 20 °C for >2 h. The 1st drop slurry was filtered and was rinsed with MTBE (8.88 kg, 12 L) twice. Wet cake was dried under vacuum 60 to 70 °C at 25 mbar overnight (>15 h). Reactor-1 was thoroughly cleaned with IPA. The dry cake was charged into Reactor-1 followed by the charge of IPA (47.10 kg, 60 L). The batch was heated to 60 °C to achieve full dissolution and cooled to 40 °C. Rich organic (24 L) was transferred to Reactor-2 for crystallization. The stream was distilled at 24 L constant volume and 100 mbar using remaining rich organic from reactor-1 as distillation feed. Following distillation completion, the batch was heated to 60 °C, aged at 60 °C for 2 h, cooled to 20 °C over 2 h, and aged at 20 °C over 2 h. The slurry was filtered. IPA (1.18 kg) was used to rinse the reactor and washed the cake. The wet cake was dried under vacuum at 70 °C and 25 mbar for >15 h. The dry cake (2.196 kg, 63.2% yield) was discharged as an off-white crystalline solid. ¾ NMR (400MHz, DMSO-d6): δ 10.62 (s, 1H), 8.48 (d, J= 7.1 Hz, 1H), 7.91 (s, 1H), 7.39 (d, J=7.4 Hz, 1H), 7.33 (s, 1H), 3.88 (m, 1H), 3.11 (t, J= 8.0 Hz, 1H), 3.0 (m, 1H), 2.96 (m, 1H), 2.78 (t, J= 10.0 Hz, 1H), 2.35 (s, 3H), 2.30 (s, 3H), 1.92 (s, 3H), 1.86 (m, 1H), 1.31 (m, 1H), 1.70 (m, 2H); 13C NMR (400 MHz, DMSO-d6): δ 168.2, 153.2, 151.9, 134.4, 133.2, 132.1, 126.5, 112.3, 108.4, 106.0, 82.3, 75.7, 56.9, 51.9, 46.3, 29.7, 24.4, 11.1, 10.2, 3.0; LC-MS: M+H= 371.2.

Step 4B: Alternative preparation of (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimeth -lH-indole-7-carboxamide

To Reactor-1 was charged N,N-dimethylformamide (DMF 4.5 mL, 4.5 mL/g). Reactor-1 was purged with N2 to inert. (,S)-4-(3-aminopiperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide 2-butynoic acid salt (1.0 g, limiting reagent) was charged followed by 2-butynoic acid (0.065g, 0.3 equiv.). The mixture was inerted with N2 for 20 min. N-methylmorpholine (0.78 g, 3.0 equiv) was charged. Next,

diphenylphosphinic chloride (0.79 g, 1.3 equiv) was charged over 0.5 h while maintaining the reaction temperature at 20-25 °C. The reaction mixture was agitated for 1.5 hour at 20 °C. Me-THF (14 mL, 14 mL/g) was charged to the reaction mixture. The reaction was quenched with the addition of aqueous NaCl (12.5 wt%, 6 mL, 6 mL/g) at 20 °C. The bottom aqueous layer was discharged as waste. Aqueous NaCl (12.5 wt%, 6 mL, 6 mL/g) at 20 °C was added to the organic layer, stirred for 0.5 hour and the bottom aqueous layer was discharged to waste. Deionized water (6 mL, 6 mL/g) was charged to the organic layer, stirred for 0.5 hour and the bottom aqueous layer was discharged to waste. THF (8 mL, 8 mL/g) was charged into Reactor-1 and the mixture was

concentrated under vacuum to remove Me-THF and water, and reconstituted in 4 L/kg of THF. The mixture was heated to 60 °C and stirred for 1 hour; the temperature was reduced to 50 °C and MTBE (12 mL, 12 mL/g) was added. The mixture was aged for 4 hours while maintaining the temperature of 50 °C and then cooled to room temperature. The solids were filtered and washed with MTBE (6.5 mL, 6.5 mL/g). The solids of crude were dried at 70 °C under vacuum for 12 hours.

Crude (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide was charged to Reactor-2, followed by THF (12 mL, 12 mL/g). The mixture was stirred for 0.5 hour. The solution was polish filtered. The solution was concentrated under vaccuum to remove THF and reconstituted in EtOH (7 mL, 7 mL/g). (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide seeds (0.01 g, 0.01 g/g) were added, the mixture was heated to 60 °C and aged for 2 hours, n-heptane (21 mL, 21 mL/g) was added slowly over 4 hours. The mixture was aged for additional 2 hours at 60 °C, followed by cooldown to room temperature. The slurry was filtered, washed with n-heptane (6 mL, 6 mL/g), and dried under vacuum at 70 °C for 12 hours. The dry cake (0.68 g, 71% yield) was discharged as an off-white crystalline solid. ¾ NMR (400MHz, DMSO-d6): δ 10.62 (s, 1H), 8.48 (d, J= 7.1 Hz, 1H), 7.91 (s, 1H), 7.39 (d, J=7.4 Hz, 1H), 7.33 (s, 1H), 3.88 (m, 1H), 3.11 (t, J= 8.0 Hz, 1H), 3.0 (m, 1H), 2.96 (m, 1H), 2.78 (t, J= 10.0 Hz, 1H), 2.35 (s, 3H), 2.30 (s, 3H), 1.92 (s, 3H), 1.86 (m, 1H), 1.31 (m, 1H), 1.70 (m, 2H); 13C MR (400 MHz, DMSO-d6): δ 168.2, 153.2, 151.9, 134.4, 133.2, 132.1, 126.5, 112.3, 108.4, 106.0, 82.3, 75.7, 56.9, 51.9, 46.3, 29.7, 24.4, 11.1, 10.2, 3.0; LC-MS: M+H= 371.2.

Applicants have discovered a new synthesis process for the preparation of (S)-4- (3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide which offers significant advantages.

The new synthesis process utilizes fewer synthesis steps (4 vs 8) than the process disclosed in WO 2016/065226.

Additionally, the process of the present invention provided (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide at an overall

yield of 22% (step 1 : 73.%, step 2: 69%, step 3 : 69%, step 4: 63%). In comparison, (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide was prepared according to the process of WO 2016/065226, which provided (S)-4-(3-(but-2-ynamido)piperidin-l-yl)-5-fluoro-2,3-dimethyl-lH-indole-7-carboxamide at an overall yield of 2.9% yield (step 1 : 91%, step 2: 71%, step 3 : 35%, step 4: 88%, step 5: 80%, step 6: 29%, step 7: 99%, step 8: 63%).

Furthermore, the process of the present invention does not include any transition metal-catalyzed steps, no genotoxic intermediates, and is adaptable to large scale manufacturing. In comparison, the process disclosed in WO 2016/065226 employed lead (Pb) in process step (8) and included a potentially genotoxic hydrazine intermediate in process step 8.

The process of the present invention has an estimated manufacturing cycle time of approximately 6 months versus a estimated manufacturing cycle time of approximately 12 months for the process disclosed in WO 2016/065226.

REFERENCE

http://acrabstracts.org/abstract/bms-986195-is-a-highly-selective-and-rapidly-acting-covalent-inhibitor-of-brutons-tyrosine-kinase-with-robust-efficacy-at-low-doses-in-preclinical-models-of-ra-and-lupus-nephritis/

/////////////////BMS-986195, Phase I,  Rheumatoid arthritis, BMS

NC(=O)c2cc(F)c(c1c(C)c(C)nc12)N3CCC[C@@H](C3)NC(=O)C#CC

GSK 2982772


str1Image result

CAS: 1622848-92-3 (free base),  1987858-31-0 (hydrate)

Chemical Formula: C20H19N5O3

Molecular Weight: 377.404

5-Benzyl-N-[(3S)-5-methyl-4-oxo-2,3,4,5-tetrahydro-1,5-benzoxazepin-3-yl]-4H-1,2,4-triazole-3-carboxamide

(S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide

  • 3-(Phenylmethyl)-N-[(3S)-2,3,4,5-tetrahydro-5-methyl-4-oxo-1,5-benzoxazepin-3-yl]-1H-1,2,4-triazole-5-carboxamide
  • (S)-5-Benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4H-1,2,4-triazole-3-carboxamide

GSK2982772 is a potent and selective receptor Interacting Protein 1 (RIP1) Kinase Specific Clinical Candidate for the Treatment of Inflammatory Diseases. GSK2982772 is, currently in phase 2a clinical studies for psoriasis, rheumatoid arthritis, and ulcerative colitis. GSK2982772 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking many TNF-dependent cellular responses. RIP1 has emerged as an important upstream kinase that has been shown to regulate inflammation through both scaffolding and kinase specific functions.

GSK-2982772, an oral receptor-interacting protein-1 (RIP1) kinase inhibitor, is in phase II clinical development at GlaxoSmithKline for the treatment of active plaque-type psoriasis, moderate to severe rheumatoid arthritis, and active ulcerative colitis. A phase I trial was also completed for the treatment of inflammatory bowel disease using capsule and solution formulations.

  • Originator GlaxoSmithKline
  • Class Antipsoriatics
  • Mechanism of Action Receptor-interacting protein serine-threonine kinase inhibitors

Highest Development Phases

  • Phase II Plaque psoriasis; Rheumatoid arthritis; Ulcerative colitis
  • Phase I Inflammatory bowel diseases

Most Recent Events

  • 15 Dec 2016 Biomarkers information updated
  • 01 Nov 2016 Phase-II clinical trials in Ulcerative colitis (Adjunctive treatment) in USA (PO) (NCT02903966)
  • 01 Oct 2016 Phase-II clinical trials in Rheumatoid arthritis in Poland (PO) (NCT02858492)

PHASE 2 Psoriasis, plaque GSK

Inflammatory Bowel Disease, Agents for
Rheumatoid Arthritis, Treatment of
Antipsoriatics
Inventors Deepak BANDYOPADHYAYPatrick M. EidamPeter J. GOUGHPhilip Anthony HarrisJae U. JeongJianxing KangBryan Wayne KINGShah Ami LakdawalaJr. Robert W. MarquisLara Kathryn LEISTERAttiq RahmanJoshi M. RamanjuluClark A SehonJR. Robert SINGHAUSDaohua Zhang
Applicant Glaxosmithkline Intellectual Property Development Limited

Deepak Bandyopadhyay

Deepak BANDYOPADHYAY

Data Science and Informatics Leader | Innovation Advocate

GSK 

 University of North Carolina at Chapel Hill

He is  a data scientist and innovator with experience in both early and late stages of drug development. his current role involves the late stage of drug product development. I’m leading a project to bring GSK’s large molecule process and analytical data onto our big data platform and develop new data analysis and modeling capabilities. Also, working within GSK’s Advanced Manufacturing Technology (AMT) initiative provides plenty of other opportunities to impact how we make medicines.

Previously as a computational chemist (i.e. a data scientist in drug discovery), he worked with scientists from many domains, including chemists, biologists, and other informaticians. he enjoys digging into all the computational aspects of life science research, and solving data challenges by exploiting adjacencies and connections – between diverse fields of knowledge, and the equally diverse scientists trained in them. 

He has supported multiple drug discovery projects at GSK starting from target identification (“how should we modulate disease X?”) through to candidate selection and early clinical development (“let’s see if what we discovered can become a medicine”). Deriving insight by custom data integration is one of my specialties; recently he designed and implemented a platform for integrating data sets from multiple experiments that will be used by GSK screening scientists to find and combine hits. 

A trained computer scientist and cheminformatician, he is  an active member of the algorithms, data science and internal innovation communities at GSK, leading many of these efforts. 

His Ph.D. work introduced new computational geometry techniques for structural bioinformatics and protein function prediction. I have touched on several other subject areas:

* data mining/machine learning (predictive modeling and graph mining), 
* computer graphics and augmented reality (one of the pioneers of projection mapping)
* robotics (keen current interest and future aspiration)

Receptor-interacting protein- 1 (RIP1) kinase, originally referred to as RIP, is a TKL family serine/threonine protein kinase involved in innate immune signaling. RIPl kinase is a RHIM domain containing protein, with an N-terminal kinase domain and a C-terminal death domain ((2005) Trends Biochem. Sci. 30, 151-159). The death domain of RIPl mediates interaction with other death domain containing proteins including Fas and TNFR-1 ((1995) Cell 81 513-523), TRAIL-Rl and TRAIL-R2 ((1997) Immunity 7, 821-830) and TRADD ((1996) Immunity 4, 387-396), while the RHIM domain is crucial for binding other RHFM domain containing proteins such as TRIF ((2004) Nat Immunol. 5, 503-507), DAI ((2009) EMBO Rep. 10, 916-922) and RIP3 ((1999) J. Biol. Chem. 274, 16871-16875); (1999) Curr. Biol. 9, 539-542) and exerts many of its effects through these interactions. RIPl is a central regulator of cell signaling, and is involved in mediating both pro-survival and programmed cell death pathways which will be discussed below.

The role for RIPl in cell signaling has been assessed under various conditions

[including TLR3 ((2004) Nat Immunol. 5, 503-507), TLR4 ((2005) J. Biol. Chem. 280,

36560-36566), TRAIL ((2012) J .Virol. Epub, ahead of print), FAS ((2004) J. Biol. Chem. 279, 7925-7933)], but is best understood in the context of mediating signals downstream of the death receptor TNFRl ((2003) Cell 114, 181-190). Engagement of the TNFR by TNF leads to its oligomerization, and the recruitment of multiple proteins, including linear K63-linked polyubiquitinated RIPl ((2006) Mol. Cell 22, 245-257), TRAF2/5 ((2010) J. Mol. Biol. 396, 528-539), TRADD ((2008) Nat. Immunol. 9, 1037-1046) and cIAPs ((2008) Proc. Natl. Acad. Sci. USA. 105, 1 1778-11783), to the cytoplasmic tail of the receptor. This complex which is dependent on RIPl as a scaffolding protein (i.e. kinase

independent), termed complex I, provides a platform for pro-survival signaling through the activation of the NFKB and MAP kinases pathways ((2010) Sci. Signal. 115, re4).

Alternatively, binding of TNF to its receptor under conditions promoting the

deubiquitination of RIPl (by proteins such as A20 and CYLD or inhibition of the cIAPs) results in receptor internalization and the formation of complex II or DISC (death-inducing signaling complex) ((2011) Cell Death Dis. 2, e230). Formation of the DISC, which contains RIPl, TRADD, FADD and caspase 8, results in the activation of caspase 8 and the onset of programmed apoptotic cell death also in a RIPl kinase independent fashion ((2012) FEBS J 278, 877-887). Apoptosis is largely a quiescent form of cell death, and is involved in routine processes such as development and cellular homeostasis.

Under conditions where the DISC forms and RJP3 is expressed, but apoptosis is inhibited (such as FADD/caspase 8 deletion, caspase inhibition or viral infection), a third RIPl kinase-dependent possibility exists. RIP3 can now enter this complex, become phosphorylated by RIPl and initiate a caspase-independent programmed necrotic cell death through the activation of MLKL and PGAM5 ((2012) Cell 148, 213-227); ((2012) Cell 148, 228-243); ((2012) Proc. Natl. Acad. Sci. USA. 109, 5322-5327). As opposed to apoptosis, programmed necrosis (not to be confused with passive necrosis which is not programmed) results in the release of danger associated molecular patterns (DAMPs) from the cell.

These DAMPs are capable of providing a “danger signal” to surrounding cells and tissues, eliciting proinflammatory responses including inflammasome activation, cytokine production and cellular recruitment ((2008 Nat. Rev. Immunol 8, 279-289).

Dysregulation of RIPl kinase-mediated programmed cell death has been linked to various inflammatory diseases, as demonstrated by use of the RIP3 knockout mouse (where RIPl -mediated programmed necrosis is completely blocked) and by Necrostatin-1 (a tool inhibitor of RIPl kinase activity with poor oral bioavailability). The RIP3 knockout mouse has been shown to be protective in inflammatory bowel disease (including Ulcerative colitis and Crohn’s disease) ((2011) Nature 477, 330-334), Psoriasis ((2011) Immunity 35, 572-582), retinal-detachment-induced photoreceptor necrosis ((2010) PNAS 107, 21695-21700), retinitis pigmentosa ((2012) Proc. Natl. Acad. Sci., 109:36, 14598-14603), cerulein-induced acute pancreatits ((2009) Cell 137, 1100-1111) and Sepsis/systemic inflammatory response syndrome (SIRS) ((2011) Immunity 35, 908-918). Necrostatin-1 has been shown to be effective in alleviating ischemic brain injury ((2005) Nat. Chem. Biol. 1, 112-119), retinal ischemia/reperfusion injury ((2010) J. Neurosci. Res. 88, 1569-1576), Huntington’s disease ((2011) Cell Death Dis. 2 el 15), renal ischemia reperfusion injury ((2012) Kidney Int. 81, 751-761), cisplatin induced kidney injury ((2012) Ren. Fail. 34, 373-377) and traumatic brain injury ((2012) Neurochem. Res. 37, 1849-1858). Other diseases or disorders regulated at least in part by RIPl -dependent apoptosis, necrosis or cytokine production include hematological and solid organ malignancies ((2013) Genes

Dev. 27: 1640-1649), bacterial infections and viral infections ((2014) Cell Host & Microbe 15, 23-35) (including, but not limited to, tuberculosis and influenza ((2013) Cell 153, 1-14)) and Lysosomal storage diseases (particularly, Gaucher Disease, Nature Medicine Advance Online Publication, 19 January 2014, doi: 10.1038/nm.3449).

A potent, selective, small molecule inhibitor of RIP1 kinase activity would block RIP 1 -dependent cellular necrosis and thereby provide a therapeutic benefit in diseases or events associated with DAMPs, cell death, and/or inflammation.

str1

Patent

WO 2014125444

Example 12

Method H

(S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide

A mixture of (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one, hydrochloride (4.00 g, 16.97 mmol), 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid, hydrochloride (4.97 g, 18.66 mmol) and DIEA (10.37 mL, 59.4 mmol) in isopropanol (150 mL) was stirred vigorously for 10 minutes and then 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (T3P) (50% by wt. in EtOAc) (15.15 mL, 25.5 mmol) was added. The mixture was stirred at rt for 10 minutes and then quenched with water and concentrated to remove isopropanol. The resulting crude material is dissolved in EtOAc and washed with 1M HC1, satd. NaHC03 and brine. Organics were concentrated and purified by column chromatography (220 g silica column; 20-90% EtOAc/hexanes, 15 min.; 90%, 15 min.) to give the title compound as a light orange foam (5.37 g, 83%). 1H NMR (MeOH-d4) δ: 7.40 – 7.45 (m, 1H), 7.21 – 7.35 (m, 8H), 5.01 (dd, J = 11.6, 7.6 Hz, 1H), 4.60 (dd, J = 9.9, 7.6 Hz, 1H), 4.41 (dd, J = 11.4, 9.9 Hz, 1H), 4.17 (s, 2H), 3.41 (s, 3H); MS (m/z) 378.3 (M+H+).

Alternative Preparation:

To a solution of (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one hydrochloride (100 g, 437 mmol), 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid hydrochloride (110 g, 459 mmol) in DCM (2.5 L) was added DIPEA (0.267 L, 1531 mmol) at 15 °C. The reaction mixture was stirred for 10 min. and 2,4,6-tripropyl-l, 3, 5,2,4,6-trioxatriphosphinane 2,4,6-trioxide >50 wt. % in ethyl acetate (0.390 L, 656 mmol) was slowly added at 15 °C. After stirring for 60 mins at RT the LCMS showed the reaction was complete, upon which time it was quenched with water, partitioned between DCM and washed with 0.5N HCl aq (2 L), saturated aqueous NaHC03 (2 L), brine (2 L) and water (2 L). The organic phase was separated and activated charcoal (100 g) and sodium sulfate

(200 g) were added. The dark solution was shaken for 1 h before filtering. The filtrate was then concentrated under reduced pressure to afford the product as a tan foam (120 g). The product was dried under a high vacuum at 50 °C for 16 h. 1H MR showed 4-5% wt of ethyl acetate present. The sample was dissolved in EtOH (650 ml) and stirred for 30 mins, after which the solvent was removed using a rotavapor (water-bath T=45 °C). The product was dried under high vacuum for 16 h at RT (118 g, 72% yield). The product was further dried under high vacuum at 50 °C for 5 h. 1H NMR showed <1% of EtOH and no ethyl acetate. 1H NMR (400 MHz, DMSO-i¾) δ ppm 4.12 (s, 2 H), 4.31 – 4.51 (m, 1 H), 4.60 (t, J=10.36 Hz, 1 H), 4.83 (dt, 7=11.31, 7.86 Hz, 1 H), 7.12 – 7.42 (m, 8 H), 7.42 – 7.65 (m, 1 H), 8.45 (br. s., 1 H), 14.41 (br. s., 1 H). MS (m/z) 378 (M + H+).

Crystallization:

(S)-5-Benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (100 mg) was dissolved in 0.9 mL of toluene and 0.1 mL of methylcyclohexane at 60 °C, then stirred briskly at room temperature (20 °C) for 4 days. After 4 days, an off-white solid was recovered (76 mg, 76% recovery). The powder X-ray diffraction (PXRD) pattern of this material is shown in Figure 7 and the corresponding diffraction data is provided in Table 1.

The PXRD analysis was conducted using a PANanalytical X’Pert Pro

diffractometer equipped with a copper anode X-ray tube, programmable slits, and

X’Celerator detector fitted with a nickel filter. Generator tension and current were set to 45kV and 40mA respectively to generate the copper Ka radiation powder diffraction pattern over the range of 2 – 40°2Θ. The test specimen was lightly triturated using an agate mortar and pestle and the resulting fine powder was mounted onto a silicon background plate.

Table 1.

Paper

Discovery of a first-in-class receptor interacting protein 1 (RIP1) kinase specific clinical candidate (GSK2982772) for the treatment of inflammatory diseases
J Med Chem 2017, 60(4): 1247

http://pubs.acs.org/doi/pdf/10.1021/acs.jmedchem.6b01751

RIP1 regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP1 kinase that are suitable for advancement into the clinic have yet to be described. Herein, we report our lead optimization of a benzoxazepinone hit from a DNA-encoded library and the discovery and profile of clinical candidate GSK2982772 (compound 5), currently in phase 2a clinical studies for psoriasis, rheumatoid arthritis, and ulcerative colitis. Compound 5 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking many TNF-dependent cellular responses. Highlighting its potential as a novel anti-inflammatory agent, the inhibitor was also able to reduce spontaneous production of cytokines from human ulcerative colitis explants. The highly favorable physicochemical and ADMET properties of 5, combined with high potency, led to a predicted low oral dose in humans.

J. Med. Chem. 2017, 60, 1247−1261

(S)-5-Benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b]- [1,4]oxazepin-3-yl)-4H-1,2,4-triazole-3-carboxamide (5).

EtOAc solvate. 1 H NMR (DMSO-d6) δ ppm 14.41 (br s, 1 H), 8.48 (br s, 1 H), 7.50 (dd, J = 7.7, 1.9 Hz, 1 H), 7.12−7.40 (m, 8 H), 4.83 (dt, J = 11.6, 7.9 Hz, 1 H), 4.60 (t, J = 10.7 Hz, 1 H), 4.41 (dd, J = 9.9, 7.8 Hz, 1 H), 4.12 (s, 2 H), 3.31 (s, 3 H). Anal. Calcd for C20H20N5O3·0.026EtOAc·0.4H2O C, 62.36; H, 5.17; N, 18.09. Found: C, 62.12; H, 5.05; N, 18.04.

Synthesis of (<it>S</it>)-3-amino-benzo[<it>b</it>][1,4]oxazepin-4-one via Mitsunobu and S<INF>N</INF>Ar reaction for a first-in-class RIP1 kinase inhibitor GSK2982772 in clinical trials
Tetrahedron Lett 2017, 58(23): 2306
Harris, P.A.
Identification of a first-in-class RIP1 kinase inhibitor in phase 2a clinical trials for immunoinflammatory diseases
ACS MEDI-EFMC Med Chem Front (June 25-28, Philadelphia) 2017, Abst 

Harris, P.
Identification of a first-in-class RIP1 kinase inhibitor in phase 2a clinical trials for immuno-inflammatory diseases
253rd Am Chem Soc (ACS) Natl Meet (April 2-6, San Francisco) 2017, Abst MEDI 313

1H NMR AND 13C NMR PREDICT

////////////GSK 2982772, phase 2, Plaque psoriasis, Rheumatoid arthritis, Ulcerative colitis

CN3c4ccccc4OC[C@H](NC(=O)c2nnc(Cc1ccccc1)n2)C3=O

AZD 9567


SCHEMBL17643955.png

str1

AZD 9567

CAS 1893415-00-3

1893415-64-9  as MONOHYDRATE

2,2-Difluoro-N-[(1R,2S)-3-methyl-1-[[1-(1-methyl-6-oxo-1,6-dihydropyridin-3-yl)-1H-indazol-5-yl]oxy]-1-phenylbutan-2-yl]propanamide

Propanamide, N-[(1S)-1-[(R)-[[1-(1,6-dihydro-1-methyl-6-oxo-3-pyridinyl)-1H-indazol-5-yl]oxy]phenylmethyl]-2-methylpropyl]-2,2-difluoro-

2,2-difluoro-N-[(1R,2S)-3-methyl-1-[1-(1-methyl-6-oxopyridin-3-yl)indazol-5-yl]oxy-1-phenylbutan-2-yl]propanamide

2,2-difluoro- V-[(lR,25)-3-methyl-l-{[l-(l-methyl-6-oxo-l,6-dihydropyridin-3-yl)-lH-indazol-5-yl]oxy}-l-phenylbutan-2-yl]propanamide

MF C27 H28 F2 N4 O3, MF 494.533

AstraZeneca INNOVATOR

AZD-9567, a glucocorticoid receptor modulator, is in early clinical development at AstraZeneca in healthy male volunteers.

Phase I Rheumatoid arthritis

  • Originator AstraZeneca
  • Class Antirheumatics
  • Mechanism of Action Glucocorticoid receptor modulators
    • 01 Sep 2016 AstraZeneca completes a phase I trial (In volunteers) in Germany (NCT02512575)
    • 24 May 2016 Phase-I clinical trials in Rheumatoid arthritis (In volunteers) in United Kingdom (PO) (NCT02760316)
    • 24 May 2016 AstraZeneca initiates a phase I trial in Rheumatoid arthritis (In volunteers) in Germany (PO) (NCT02760316)
     
Inventors Lena Elisabeth RIPA, Karolina Lawitz, Matti Juhani Lepistö, Martin Hemmerling, Karl Edman, Antonio Llinas
Applicant Astrazeneca

Warning: Chancellor George Osborne told Scotland it could be forced to give up the pound if it became independent of the rest of the UK. He is pictured yesterday with Jan Milton-Edwards during a visit to the Macclesfield AstraZeneca site in Cheshire

Macclesfield AstraZeneca site in Cheshire

Image result

Glucocorticoids (GCs) have been used for decades to treat acute and chronic inflammatory and immune conditions, including rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (“COPD”), osteoarthritis, rheumatic fever, allergic rhinitis, systemic lupus erythematosus, Crohn’s disease, inflammatory bowel disease, and ulcerative colitis. Examples of GCs include dexamethasone, prednisone, and

prednisolone. Unfortunately, GCs are often associated with severe and sometimes irreversible side effects, such as osteoporosis, hyperglycemia, effects on glucose metabolism (diabetes mellitus). skin thinning, hypertension, glaucoma, muscle atrophy. Cushing’s syndrome, fluid homeostasis, and psychosis (depression ). These side effects can particularly limit the use of GCs in a chronic setting. Thus, a need continues to exist for alternative therapies that possess the beneficial effects of GCs, but with a reduced likel ihood of side effects.

GCs form a complex with the GC receptor ( GR ) to regulate gene transcription. The GC-GR complex translocates to the cell nucleus, and then binds to GC response elements (GREs) in the promoter regions of various genes. The resulting GC-GR- GRE complex, in turn, activates or inhibits transcription of proximally located genes. The GC-GR complex also (or alternatively) may negatively regulate gene transcription by a process that does not involve DNA binding. In this process, termed transrepression, the GC-GR complex enters the nucleus and directly interacts (via protein-protein interaction) with other transcription factors, repressing their ability to induce gene transcription and thus protein expression.

Some of the side effects of GCs are believed to be the result of cross-reactivity with other steroid receptors (e.g., progesterone, androgen, mineralocorticoid, and estrogen receptors), which have somewhat homologous ligand binding domains; and/or the inability to selectively modulate gene expression and downstream signaling. Consequently, it is believed that an efficacious selective GR modulator (SGRM), which binds to GR with greater affinity relative to other steroid hormone receptors, would provide an alternative therapy to address the unmet need for a therapy that possesses the beneficial, effects of GCs, while, at the same time, having fewer side effects.

A range of compounds have been reported to have SGRM activity. See, e.g., WO2007/0467747, WO2007/114763, WO2008/006627, WO2008/055709, WO2008/055710, WO2008/052808, WO2008/063116, WO2008/076048,

WO2008/079073, WO2008/098798, WO2009/065503, WO2009/142569,

WO2009/142571, WO2010/009814, WO2013/001294, and EP2072509. Still, there continues to be a need for new SGRMs that exhibit, for example, an improved potency, efficacy, effectiveness in steroid-insensitive patients, selectivity, solubility allowing for oral administration, pharmacokinetic profile allowing for a desirable dosing regimen, stability on the shelf {e.g., hydro lytic, thermal, chemical, or photochemical stability), crystallinity, tolerability for a range of patients, side effect profile and/or safety profile.

PATENT

WO 2016046260

Scheme 1 below illustrates a general protocol for making compounds described in this specification, using either an Ullman route or an aziridine route.

Scheme 1

In Scheme 1, Ar is

[182] The amino alcohol reagent used in Scheme 1 may be made using the below Scheme 2.

Scheme 2

The Grignard reagent (ArMgBr) used in Scheme 2 can be obtained commercially, or, if not, can generally be prepared from the corresponding aryl bromide and Mg and/or iPrMgCl using published methods.

[183] The iodo and hydroxy pyridone indazole reagents used in Scheme 1 may be made using the below Scheme 3A or 3B, respectively.

Scheme 3A

[184] Scheme 4 below provides an alternative protocol for making compounds described in this specification.

Scheme 4

Example 1. Preparation of 2,2-difluoro- V-[(lR,2S)-3-methyl-l-{[l-(l-methyl-6-oxo-l,6-dihydropyridin-3-yl)-lH-indazol-5-yl]oxy}-l-phenylbutan-2-yl]propanamide.

[199] Step A. Preparation of 5-[5-[(te^butyldimethylsilyl)oxy]-lH-indazol-l-yl]-l-methyl-l,2-dihydropyridin-2-one.

Into a 2 L 4-necked, round-bottom flask, purged and maintained with an inert atmosphere of N2, was placed a solution of 5-[(tert-butyldimethylsilyl)oxy]-lH-indazole (805 g, 3.2 mol) in toluene (8 L), 5 -iodo-1 -methyl- 1 ,2-dihydropyridin-2-one (800 g, 3.4 mol) and

K3PO4 (1.2 kg, 5.8 mol). Cyclohexane-l,2-diamine (63 g, 0.5 mol) was added followed by the addition of Cul (1.3 g, 6.8 mmol) in several batches. The resulting solution was stirred overnight at 102°C. The resulting mixture was concentrated under vacuum to yield 3.0 kg of the title compound as a crude black solid. LC/MS: m/z 356 [M+H]+.

[200] Step B. Preparation of 5-(5-hydroxy-lH-indazol-l-yl)-l-methylpyridin-2(lH)-one.

Into a 2 L 4-necked, round-bottom flask was placed 5-[5-[(fert-butyldimethylsilyl)oxy]-lH-indazol-l-yl]-l-methyl-l,2-dihydropyridin-2-one (3.0 kg, crude) and a solution of HCl (2 L, 24 mol, 36%) in water (2 L) and MeOH (5 L). The resulting solution was stirred for 1 hr at 40°C and then evaporated to dryness. The resulting solid was washed with water (4 x 5 L) and ethyl acetate (2 x 0.5 L) to afford 480 g (61%, two steps) of the title product as a brown solid. LC/MS: m/z 242 [M+H]+. 1HNMR (300 MHz, DMSO-d6): δ 3.52 (3H, s),6.61 (lH,m),7.06 (2H,m),7.54 (lH,m), 7.77 (lH,m), 8.19 (2H, m) 9.35 (lH,s).

[201] Ste C. Preparation of tert-butyl((lR,25)-l-hydroxy-3-methyl-l-phenylbutan-2-yl)carbamate.

(S)-tert-butyl 3 -methyl- l-oxo-l-phenylbutan-2-ylcarbamate (1.0 kg, 3.5 mol) was dissolved in toluene (4 L). Afterward, 2-propanol (2 L) was added, followed by triisopropoxyaluminum (0.145 L, 0.73 mol). The reaction mixture was heated at 54-58°C for 1 hr under reduced pressure (300-350 mbar) to start azeothropic distillation. After the collection of 0.75 L condensate, 2-propanol (2 L) was added, and the reaction mixture was stirred overnight at reduced pressure to afford 4 L condensate in total. Toluene (3 L) was added at 20°C, followed by 2M HC1 (2 L) over 15 min to keep the temperature below 28°C. The layers were separated (pH of aqueous phase 0-1) and the organic layer was washed successively with water (3 L), 4% NaHCCte (2 L) and water (250 mL). The volume of the organic layer was reduced from 6 L at 50°C and 70 mbar to 2.5 L. The resulting mixture was heated to 50°C and heptane (6.5 L) was added at 47-53°C to maintain the material in solution. The temperature of the mixture was slowly decreased to 20°C, seeded with the crystals of the title compound at 37°C (seed crystals were prepared in an earlier batch made by the same method and then evaporating the reaction mixture to dryness, slurring the residue in heptane, and isolating the crystals by filtration), and allowed to stand overnight. The product was filtered off, washed with heptane (2 x 1 L) and dried under vacuum to afford 806 g (81%) of the title compound as a white solid. 1HNMR (500 MHz, DMSO-d6): δ 0.81 (dd, 6H), 1.16 (s, 8H), 2.19 (m, 1H), 3.51 (m, 1H), 4.32 (d, 1H), 5.26 (s, 1H), 6.30 (d, 1H), 7.13 – 7.2 (m, 1H), 7.24 (t, 2H), 7.3 – 7.36 (m, 3H).

[202] Step D. Preparation of (lR,2S)-2-amino-3-methyl-l-phenylbutan-l-ol hydrochloride salt.

To a solution of HC1 in propan-2-ol (5-6 N, 3.1 L, 16 mol) at 20°C was added tert-butyl((li?,25)-l-hydroxy-3-methyl-l-phenylbutan-2-yl)carbamate (605 g, 2.2 mol) in portions over 70 min followed by the addition of MTBE (2 L) over 30 min. The reaction mixture was cooled to 5°C and stirred for 18 hr. The product was isolated by filtration and dried to afford 286 g of the title compound as an HC1 salt (61% yield). The mother liquor was concentrated to 300 mL. MTBE (300 mL) was then added, and the resulting precipitation was isolated by filtration to afford additional 84 g of the title compound as a HC1 salt (18% yield). Total 370 g (79%). 1HNMR (400 MHz, DMSO-d6): δ 0.91 (dd, 6H), 1.61 – 1.81 (m, 1H), 3.11 (s, 1H), 4.99 (s, 1H), 6.08 (d, 1H), 7.30 (t, 1H), 7.40 (dt, 4H), 7.97 (s, 2H).

[203] Step E. Preparation of (2S,35)-2-isopropyl-l-(4-nitrophenylsulfonyl)-3-phenylaziridine.

(li?,25)-2-Amino-3-methyl-l-phenylbutan-l-ol hydrochloride (430 g, 2.0 mol) was mixed with DCM (5 L) at 20°C. 4-Nitrobenzenesulfonyl chloride (460 g, 2.0 mol) was then added over 5 min. Afterward, the mixture was cooled to -27°C. Triethylamine (1.0 kg, 10 mol) was slowly added while maintaining the temperature at -18°C. The reaction mixture was cooled to -30°C, and methanesulfonyl chloride (460 g, 4.0 mol) was added slowly while maintaining the temperature at -25 °C. The reaction mixture was then stirred at 0°C for 16 hr before adding triethylamine (40 mL, 0.3 mol; 20 mL ,0.14 mol and 10 mL, 0.074 mol) w at 0°C in portions over 4 hr. Water (5 L) was subsequently added at 20°C, and the resulting layers were separated. The organic layer was washed with water (5 L) and the volume reduced to 1 L under vacuum. MTBE (1.5 L) was added, and the mixture was stirred on a rotavap at 20°C over night and filtered to afford 500 g (70%) of the title product as a solid. 1HNMR (400 MHz, CDCls): δ 1.12 (d, 3H), 1.25 (d, 3H), 2.23 (ddt, 1H), 2.89 (dd, 1H), 3.84 (d, 1H), 7.08 – 7.2 (m, 1H), 7.22 – 7.35 (m, 4H), 8.01 – 8.13 (m, 2H), 8.22 – 8.35 (m, 2H)

[204] Step F. Preparation of V-((lR,2S)-3-methyl-l-(l-(l-methyl-6-oxo-l,6-dihydropyridin-3-yl)-lH-indazol-5-yloxy)-l-phenylbutan-2-yl)-4-nitrobenzenesulfonamide.

[205] (25′,35)-2-Isopropyl-l-(4-nitrophenylsulfonyl)-3-phenylaziridine (490 g, 1.3 mol) was mixed with 5-(5-hydroxy-lH-indazol-l-yl)-l-methylpyridin-2(lH)-one (360 g, 1.4 mol) in acetonitrile (5 L) at 20°C. Cesium carbonate (850 g, 2.6 mol) was added in portions over 5 min. The reaction mixture was then stirred at 50°C overnight. Water (5 L) was added at 20°C, and the resulting mixture was extracted with 2-methyltetrahydrofuran (5L and 2.5 L). The combined organic layer was washed successively with 0.5 M HC1 (5 L), water (3 x 5L) and brine (5L). The remaining organic layer was concentrated to a thick oil, and then MTBE (2 L) was added. The resulting precipitate was filtered to afford 780 g (purity 71% w/w) of the crude title product as a yellow solid, which was used in the next step without further purification. 1HNMR (400 MHz, DMSO-d6): δ 0.93 (dd, 6H), 2.01 -2.19 (m, 1H), 3.50 (s, 3H), 3.74 (s, 1H), 5.00 (d, 1H), 6.54 (d, 1H), 6.78 (d, 1H), 6.95 -7.15 (m, 4H), 7.23 (d, 2H), 7.49 (d, 1H), 7.69 (dd, 1H), 7.74 (d, 2H), 8.00 (s, 1H), 8.08 (d, 2H), 8.13 (d, 2H).

[206] Step G. Preparation of 2,2-difluoro- V-[(lR,25)-3-methyl-l-{[l-(l-methyl-6-oxo-l,6-dihydropyridin-3-yl)-lH-indazol-5-yl]oxy}-l-phenylbutan-2-yl]propanamide.

[207] N-((lR,2S)-3-Methyl- 1 -(1 -(1 -methyl-6-oxo- 1 ,6-dihydropyridin-3-yl)- \H-indazol-5-yloxy)-l-phenylbutan-2-yl)-4-nitrobenzenesulfonamide (780 g, 71%w/w) was mixed with DMF (4 L). DBU (860 g, 5.6 mol) was then added at 20°C over 10 min. 2-Mercaptoacetic acid (170 g, 1.9 mol) was added slowly over 30 min, keeping the temperature at 20°C. After 1 hr, ethyl 2,2-difluoropropanoate (635 g, 4.60 mol) was added over 10 min at 20°C. The reaction mixture was stirred for 18 hr. Subsequently, additional ethyl 2,2-difluoropropanoate (254 g, 1.8 mol) was added, and the reaction mixture was stirred for an additional 4 hr at 20°C. Water (5 L) was then slowly added over 40 min, maintaining the temperature at 20°C. The water layer was extracted with isopropyl acetate (4 L and 2 x 2 L). The combined organic layer was washed with 0.5M HC1 (4 L) and brine (2 L). The organic layer was then combined with the organic layer from a parallel reaction starting from 96 g of N-((li?,25)-3-methyl-l-((l-(l-methyl-6-oxo-l,6-dihydropyridin-3-yl)- lH-indazol-5-yl)oxy)- 1 -phenylbutan-2-yl)-4-nitrobenzenesulfonamide, and concentrated to approximate 1.5 L. The resulting brown solution was filtered. The filter was washed twice with isopropyl acetate (2 x 0.5 L). The filtrate was evaporated until a solid formed. The solid was then co evaporated with 99.5% ethanol (1 L), affording 493 g (77%, two steps) of an amorphous solid.

[208] The solid (464 g, 0.94 mol) was dissolved in ethanol/water 2: 1 (3.7 L) at 50°C. The reaction mixture was then seeded with crystals () of the title compound (0.5 g) at 47°C, and a slight opaque mixture was formed. The mixture was held at that temperature for 1 hr. Afterward, the temperature was decreased to 20°C over 7 hr, and kept at 20°C for 40 hr. The solid was filtrated off, washed with cold (5°C) ethanol/water 1 :2 (0.8 L), and dried in vacuum at 37°C overnight to afford 356 g (0.70 mol, 74%, 99.9 % ee) of the title compound as a monohydrate. LC/MS: m/z 495 [M+H]+. ‘HNMR (600 MHz, DMSO-d6) δ 0.91 (dd, 6H), 1.38 (t, 3H), 2.42 (m, 1H), 3.50 (s, 3H), 4.21 (m, 1H), 5.29 (d, 1H), 6.53 (d, 1H), 7.09 (d, 1H), 7.13 (dd, 1H), 7.22 (t, 1H), 7.29 (t, 2H), 7.47 (d, 2H), 7.56 (d, 1H), 7.70 (dd, 1H), 8.13 (d, 1H), 8.16 (d, 1H), 8.27 (d, 1H).

[209] The seed crystals may be prepared from amorphous compound prepared according to Example 2 using 2,2-difluoropropanoic acid, followed by purification on HPLC. The compound (401 mg) was weighed into a glass vial. Ethanol (0.4 mL) was added, and the vial was shaken and heated to 40°C to afford a clear, slightly yellow solution. Ethanol/Water (0.4 mL, 50/50% vol/vol) was added. Crystallization started to

occur within 5 min, and, after 10 min, a white thick suspension formed. The crystals were collected by filtration

/////////////AZD 9567, AstraZeneca, lucocorticoid receptor modulator, Rheumatoid arthritis, phase 1, Lena Elisabeth RIPA, Karolina Lawitz, Matti Juhani Lepistö, Martin Hemmerling, Karl Edman, Antonio Llinas

3rd speaker this afternoon in 1st time disclosures is Lena Ripa of @AstraZeneca on a glucocorticoid receptor modulator

str2

CC(F)(F)C(=O)N[C@@H](C(C)C)[C@H](Oc1cc2cnn(c2cc1)C=3C=CC(=O)N(C)C=3)c4ccccc4

CEP 33779


img

CEP-33779, CEP33779
CAS 1257704-57-6
Chemical Formula: C24H26N6O2S
Molecular Weight: 462.57
Elemental Analysis: C, 62.32; H, 5.67; N, 18.17; O, 6.92; S, 6.93

N-(3-(4-methylpiperazin-1-yl)phenyl)-8-(4-(methylsulfonyl)phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine

PRECLINICAL Treatment of Rheumatoid Arthritis, Agents for Colorectal Cancer Therapy Systemic Lupus Erythematosus,

Jak2 Inhibitors

Image result for teva logo

Matthew A. Curry, Bruce D. Dorsey, Benjamin J. Dugan, Diane E. Gingrich, Eugen F. Mesaros, Karen L. Milkiewicz,
Applicant Cephalon, Inc.

Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380, United States

Image result for Cephalon, Inc.

Matt Curry

 Matthew A. Curry

Bruce Dorsey

Bruce Dorsey

Image result for Cephalon, Inc. Benjamin J. Dugan

Benjamin Dugan

Benjamin J. Dugan received a B.S. degree in Chemistry from the University of Delaware in 1993 under the tutelage of the late Dr. Cynthia McClure. He began his career at FMC Corporation in the agricultural products division. In 2006, he moved to Cephalon, Inc., acquired by Teva Pharmaceutical Industries Ltd. in 2011, and engaged in oncology research focused on small molecule, ATP competitive, kinase inhibitors culminating with the discovery of CEP-33779. He is currently a Research Scientist focused on the development of novel, bioactive small molecules for treatment of central nervous system disorders.

Cephalon Inc.
Malvern, United States

Image result for Cephalon, Inc. Diane E. Gingrich

Members of the Cephalon research team that discovered CEP-5214 and CEP-7055 include (from left) Hudkins, Thelma S. Angeles, Bruce A. Ruggeri, and Diane E. Gingrich. CEPHALON PHOTO

Eugen F. Mesaros

Cephalon Inc.
Malvern, United States
Image result for cephalon Karen L. Milkiewicz

Lupus (systemic lupus erythematosus, SLE) is a chronic autoimmune disease characterized by the presence of activated T and B cells, autoantibodies and chronic inflammation that attacks various parts of the body including the joints, skin, kidneys, CNS, cardiac tissue and blood vessels. In severe cases, antibodies are deposited in the cells (glomeruli) of the kidneys, leading to inflammation and possibly kidney failure, a condition known as lupus nephritis.

Although the cause of lupus remains unknown, manifestations of the disease have been linked to genetic polymorphisms, environmental toxins and pathogens (Morel;

Fairhurst, Wandstrat et al. 2006). In addition, gender, hormonal influences and cytokine dysregulation have been tightly linked to the development of lupus (Aringer and Smolen 2004; Smith-Bouvier, Divekar et al. 2008). Lupus affects nine times as many women as men. It may occur at any age, but appears most often in people between the ages of 10 and 50 years. African Americans and Asians are affected more often than people from other races.

There is no cure for lupus. Current treatments for lupus are aimed at controlling symptoms and are limited to toxic and immunosuppressive agents with severe side-effects such as high dose glucocorticoids and/or hydroxchloroquine. Severe disease (e.g., patients that have signs of renal involvement) require more aggressive drugs including

mycophenolate mofetil (MMF), azathioprine (AZA) and/or cyclophosphamide (CTX) (Bertsias and Boumpas 2008). CTX, AZA and MMF are very toxic and

immunosuppressive, and only 50% of treated patients enter complete remission, with relapse rates up to 30% over a 2-year period.

Memory B cells, and more important, long-lived plasma cells (LL-PCs) which differentiate from memory B cells, are key cell types involved in lupus (Neubert, Meister et al. 2008; Sanz and Lee 2010). Long-lived plasma cells synthesize and secrete large quantities of high-affinity isotype switched antibodies (Meister, Schubert et al. 2007;

Muller, Dieker et al. 2008). Circulating antinuclear antibodies (ANAs) increase the chances of antibody depositing onto self tissues, forming immune-complexes and eventually leading to tissue destruction, epitope spreading and involvement of other organ systems. LL-PCs are commonly found to be chemo- and radio-resistant, over expressing various heat shock proteins and drug pumps (Obeng, Carlson et al. 2006; Neubert, Meister et al. 2008). In addition, LL-PCs primarily reside in the bone marrow where they are protected from current lupus therapies such as cyclophosphamide and glucocorticoids.

A need exists for new treatments for lupus, including lupus nephritis. A need particularly exists for lupus treatments that can target and reduce LL-PCs.

str0

CEP-33779 is a highly selective, orally active, small-molecule inhibitor of JAK2. CEP-33779 induced regression of established colorectal tumors, reduced angiogenesis, and reduced proliferation of tumor cells. Tumor regression correlated with inhibition of STAT3 and NF-κB (RelA/p65) activation in a CEP-33779 dose-dependent manner. The ability of CEP-33779 to suppress growth of colorectal tumors by inhibiting the IL-6/JAK2/STAT3 signaling suggests a potential therapeutic utility of JAK2 inhibitors in multiple tumors types, particularly those with a strong inflammatory component.

str0

{[8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine} (1)

LC/MS: (M+H+)+ = 463.2;
1H NMR (DMSO, 400 MHz) δ 9.61 (s, 1H), 8.85 (d, J = 6.8 Hz, 1H), 8.43 (d, J = 6.8 Hz, 2H), 8.06 (d, J = 6.8 Hz, 2H), 7.96 (d, J = 7.5 Hz, 1H), 7.59 (s, 1H), 7.17 (t, J = 6.8 Hz, 1H), 7.11 (t, J = 8.0 Hz, 1H), 7.05 (d, J = 8.6 Hz 1H), 6.49 (d, J = 8.0 Hz, 1H), 3.30 (s, 3H), 3.13 (m, 4H), 2.48 (m, 4H), 2.24 (s, 3H).
CEP-33779 Diglycolate Salt
1H NMR (DMSO, 400 MHz) δ 9.61 (s, 1H), 8.85 (d, J = 6.7 Hz, 1H), 8.43 (d, J = 6.7 Hz, 2H), 8.06 (d, J = 6.7 Hz, 2H), 7.97 (d, J = 7.5 Hz, 1H), 7.59 (s, 1H), 7.18 (d, J = 6.7 Hz, 1H), 7.11 (m, 1H), 7.05 (d, J = 8.6 Hz, 1H), 6.50 (d, J = 8.0 Hz, 1H), 3.89 (s, 4H), 3.30 (s, 3H), 3.13 (m, 4H), 2.48 (m, 4H), 2.24 (s, 3H).
DSC: Endotherm onset at 153.0 °C; Peak at 155.8 °C.

PATENT

WO 2010141796

https://www.google.com/patents/WO2010141796A3?cl=en

Example 35 [8-(4-Methanesulfonyl-phenyl)-[ 1 ,2,4]triazolo[ 1 ,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-

1 -yl)-phenyl]-amine

Figure imgf000156_0001

35 a) l-(3-Bromo-phenyl)-4-methyl-piperazine was prepared from l-(3-bromo-phenyl)- piperazine (1.33 g, 5.52 mmol) in a manner analogous to Step 32a. The reaction product was isolated as a pale yellow oil (1.4 g, 100%). 1H NMR (400 MHz, CDCl3, δ, ppm): 7.10 (dd, J=8.2, 8.2 Hz, IH), 7.04 (dd, J=2.1, 2.1 Hz, IH), 6.95 (ddd, J=I. S, 1.7, 0.7 Hz, IH), 6.83 (ddd, J=8.3, 2.4, 0.6 Hz, IH), 3.23-3.18 (m, 4H), 2.58-2.54 (m, 4H), 2.35 (s, 3H). MS = 255, 257 (MH)+. 35b) [8-(4-Methanesulfonyl-phenyl)-[ 1 ,2,4]triazolo[ 1 ,5-a]pyridin-2-yl]-[3-(4-methyl- piperazin-l-yl)-phenyl]-amine was prepared from 8-(4-methanesulfonyl-phenyl)- [l,2,4]triazolo[l,5-a]pyridin-2-ylamine (75.0 mg, 0.260 mmol) and l-(3-bromo-phenyl)-4- methyl-piperazine (80.0 mg, 0.314 mmol) with 2,2′-bis-dicyclohexylphosphanyl-biphenyl (30.0 mg, 0.0549 mmol) as the ligand in a manner analogous to Step 2d and was isolated as a yellow solid (0.072 g, 60%).

MP = 232-234 0C.

1H NMR (400 MHz, CDCl3, δ, ppm): 8.49 (d, J=I 2 Hz, IH), 8.25 (d, J=I .5 Hz, 2H), 8.08 (d, J=I .9 Hz, 2H), 7.65 (d, J=I .1 Hz, IH), 7.38 (s, IH), 7.27-7.20 (m, IH), 7.04-6.95 (m, 2H), 6.84 (s, IH), 6.60 (d, J=8.0 Hz, IH), 3.30-3.25 (m, 4H), 3.10 (s, 3H), 2.63-2.58 (m, 4H), 2.38 (s, 3H).

MS = 463 (MH)+.

PATENT

WO 2012078504

PATENT

WO 2012078574

https://google.com/patents/WO2012078574A2?cl=da

COMPOUND A is a JAK2 inhibitor with the chemical name [8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine. COMPOUND A has the following structure:

COMPOUND A

COMPOUND A was prepared in a manner analogous to the five-step method described below (see Example 35 of International Application No. PCT/US10/37363):

Step 1 : To a solution of 1-(3-bromo-phenyl)-piperazine (about 1 g) and acetic acid (about 0.4 mL) in methanol (about 25 mL) is added 37% formaldehyde in water/methanol (about 56.7:37:6.3, water:formaldehyde:methanol; about 5 mL). The mixture is stirred at room temperature for about 18 hours. The suspension is cooled to about 5°C in an ice/water bath and sodium cyanoborohydride (about 5 g) is added in small portions. The mixture is stirred and warmed to room temperature for about 18 hours. The mixture is slowly poured into saturated aqueous ammonium chloride (about 200 mL) and stirred for about 1 hour. The mixture is extracted with dichloromethane (3 x about 75 mL). The combined organic layers are dried over magnesium sulfate, filtered and evaporated. The material is placed under high vacuum for about 18 hours to yield 1-(3-bromo-phenyl)-4-methyl-piperazine as a pale yellow oil (about 1 g). 1H NMR (400 MHz, CDCl3, δ, ppm): 7.10 (dd, J=8.2, 8.2 Hz, 1H), 7.04 (dd, J=2.1, 2.1 Hz, 1H), 6.95 (ddd, J=7.8, 1.7, 0.7 Hz, 1H), 6.83 (ddd, J=8.3, 2.4, 0.6 Hz, 1H), 3.23-3.18 (m, 4H), 2.58-2.54 (m, 4H), 2.35 (s, 3H). MS = 255, 257 (MH)+.

Step 2: To a solution of 3-bromo-pyridin-2-ylamine (about 10 g) in 1,4-dioxane (about 100 mL) is added dropwise ethoxycarbonyl isothiocyanate (about 7 mL). The mixture is stirred under an atmosphere of nitrogen for about 18 hours. The volatiles are evaporated to yield a waxy solid. The recovered material is triturated with hexane (about 250 mL). N-(3-bromo-2-pyridinyl)-N’-carboethoxy-thiourea is isolated and used without further purification. 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 11.46 (s, 1H), 11.43 (s, 1H), 8.49 (dd, J=4.6, 1.5 Hz, 1H), 8.18 (dd, J=8.0, 1.5 Hz, 1H), 7.33 (dd, J=8.0, 4.7 Hz, 1H), 4.23 (q, J=7.1 Hz, 2H), 1.27 (t, J=7.2 Hz, 3H). MS = 215 (MH)+.

Step 3: To a stirred suspension of hydroxylamine hydrochloride (about 17 g) and Ν,Ν-diisopropylethylamine (about 26 mL) in a mixture of methanol (about 70 mL) and

ethanol (about 70 mL) is added N-(3-bromo-2-pyridinyl)-N’-carboethoxy-thiourea. The mixture is stirred for about 2 hours at room temperature then heated to about 60°C for about 18 hours. The suspension is cooled to room temperature, filtered and rinsed with methanol, water then methanol. 8-Bromo-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine is isolated as an off-white solid (about 8 g). 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 8.58 (d, J=6.4 Hz, 1H), 7.73 (d, J=7.6 Hz, 1H), 6.80 (t, J=7.0 Hz, 1H), 6.25 (s, 2H). MS = 213, 215 (MH)+.

Step 4: An oven dried tube is charged with palladium acetate (about 0.2 g) and triphenylphosphine (about 0.6 g). The tube is evacuated under high vacuum and backflushed under a stream of nitrogen for about 5 minutes. A suitable solvent such as

1,4-dioxane (about 10 mL) is added and the mixture is stirred under nitrogen for a suitable time (e.g., for about 10 minutes). 8-Bromo-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine (about 0.75 g), (4-methylsulfonylphenyl)boronic acid (about 1 g), a suitable solvent, such as N,N-dimethylformamide (about 10 mL) and a suitable base, such as about 1.5 M of sodium carbonate in water (about 10 mL) are added. The mixture is stirred for about 2 minutes at room temperature under nitrogen then the tube is sealed and heated at about 80°C for about 18 hours. The mixture is transferred to a round bottom flask and the volatiles are evaporated under reduced pressure. The product is isolated in a suitable manner. For example, water (about 100 mL) may be added and the mixture stirred. The solid may then be collected by filtration, and optionally rinsed with water, air dried, triturated with ether/dichloromethane (about 4: 1; about 10 mL), filtered and rinsed with ether. 8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine is isolated as a tan solid (about 0.6 g). MP = 236-239 °C. 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 8.63 (d, J=6.3 Hz, 1H), 8.38 (d, J=7.9 Hz, 2H), 8.03 (d, J=7.9 Hz, 2H), 7.84 (d, J= 7.3 Hz, 1H), 7.03 (t, J=7.0 Hz, 1H), 6.21 (br s, 2H), 3.28 (s, 3H). MS = 289 (MH)+.

Step 5: To an oven dried tube is added palladium acetate (about 10 mg) and 2,2′-bis-dicyclohexylphosphanyl-biphenyl (about 30 mg), 8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine (about 75 mg), 1-(3-bromo-phenyl)-4-methyl-piperazine (about 80 mg), a suitable base, such as cesium carbonate (about 270 mg) and a suitable solvent, such as 1,4-dioxane (about 5 mL). The tube is evacuated and backflushed with nitrogen three times. The tube is sealed and heated at about 80°C for about 72 hours. The mixture is cooled to room temperature and the product isolated in a suitable manner.

For example, the cooled mixture may be diluted with dichloromethane (about 10 mL), filtered through a plug of diatomaceous earth, rinsed with dichloromethane and evaporated. The material may then be purified, e.g., via chromatography, e.g., utilizing an ISCO automated purification apparatus (e.g., amine modified silica gel column 5%→100% ethyl acetate in hexanes). [8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine (i.e., COMPOUND A) is isolated as a yellow solid (about 0.07 g). MP = 232-234 °C. 1H NMR (400 MHz, CDCl3, δ, ppm): 8.49 (d, J=7.2 Hz, 1H), 8.25 (d, J=7.5 Hz, 2H), 8.08 (d, J=7.9 Hz, 2H), 7.65 (d, J=7.7 Hz, 1H), 7.38 (s, 1H), 7.27-7.20 (m, 1H), 7.04-6.95 (m, 2H), 6.84 (s, 1H), 6.60 (d, J=8.0 Hz, 1H), 3.30-3.25 (m, 4H), 3.10 (s, 3H), 2.63-2.58 (m, 4H), 2.38 (s, 3H). MS = 463 (MH)+.

PATENT

WO 2015089153

https://www.google.com/patents/WO2015089153A1?cl=un

This disclosure relates to a l,2,4 riazolo[l,5a]pyridine derivative, [8-(4 methanesulfonyl-phenyl)-[ 1 ,2,4]triazoio[1 ,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin- 1 -yl phenyl] -amine, re g structure:

or a pharmaceutical salt thereof, and its use in the treatment of multiple sclerosis.

Compound A is a potent, orally active, small molecule inhibitor of JA 2. See, e.g..International Application No. PCT/USlO/37363, U.S. Patent Nos. 8,501,936 and ,633,173, and U.S. Published Patent Application Nos. 2013/0267535 and 2014/0024655, each of which is incorporated by reference herein. Compound A can be prepared, for example, using methods analogous to Example 35 of International Application No.PCT/US 10/37363.

PAPER

A Selective, Orally Bioavailable 1,2,4-Triazolo[1,5-a]pyridine-Based Inhibitor of Janus Kinase 2 for Use in Anticancer Therapy: Discovery of CEP-33779

Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380, United States
J. Med. Chem., 2012, 55 (11), pp 5243–5254
DOI: 10.1021/jm300248q
Publication Date (Web): May 10, 2012
Copyright © 2012 American Chemical Society
*Phone: 610-738-6733. Fax: 610-738-6643. E-Mail: bdugan@cephalon.com.

Abstract

Abstract Image

Members of the JAK family of nonreceptor tyrosine kinases play a critical role in the growth and progression of many cancers and in inflammatory diseases. JAK2 has emerged as a leading therapeutic target for oncology, providing a rationale for the development of a selective JAK2 inhibitor. A program to optimize selective JAK2 inhibitors to combat cancer while reducing the risk of immune suppression associated with JAK3 inhibition was undertaken. The structure–activity relationships and biological evaluation of a novel series of compounds based on a 1,2,4-triazolo[1,5-a]pyridine scaffold are reported. Para substitution on the aryl at the C8 position of the core was optimum for JAK2 potency (17). Substitution at the C2 nitrogen position was required for cell potency (21). Interestingly, meta substitution of C2-NH-aryl moiety provided exceptional selectivity for JAK2 over JAK3 (23). These efforts led to the discovery of CEP-33779 (29), a novel, selective, and orally bioavailable inhibitor of JAK2.

[8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine (29)

 1H NMR (CDCl3) δ 8.49 (dd, J = 6.6, 1.0 Hz, 1H), 8.25 (d, J = 8.4 Hz, 2H), 8.08 (d, J = 8.4 Hz, 2H), 7.66 (dd, J = 7.5, 0.9 Hz, 1H), 7.39–7.36 (m, 1H), 7.23 (t, J = 8.2 Hz, 1H), 7.02 (t, J = 7.1 Hz, 1H), 6.97 (dd, J = 7.8, 1.4 Hz, 1H), 6.88 (s, 1H), 6.60 (dd, J = 8.3, 1.8 Hz, 1H), 3.30–3.25 (m, 4H), 3.10 (s, 3H), 2.63–2.58 (m, 4H), 2.38 (s, 3H).
13C NMR (CDCl3) δ 162.65, 152.28, 148.87, 141.00, 140.91, 140.05, 129.64, 129.29, 128.18, 127.85, 127.76, 124.77, 112.03, 109.40, 108.59, 104.80, 55.19, 49.02, 46.19, 44.59;
mp 208–211 °C.
High resolution mass spectrum (ESI+) m/z 463.1925 [(M + H)+calcd for C24H26N6O2S: 463.1916]. HPLC: 95 A%.

PAPER

An Improved Synthesis of the Free Base and Diglycolate Salt of CEP-33779; A Janus Kinase 2 Inhibitor

Chemical Process Research and Development, Teva Branded Pharmaceutical Products R&D Inc., 383 Phoenixville Pike, Malvern, Pennsylvania 19355, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00311
Publication Date (Web): November 30, 2016
Copyright © 2016 American Chemical Society

Abstract

Abstract Image

CEP-33779 is a triazole that has been reported to show highly selective inhibition of Janus kinase 2 (JAK2). An efficient process to form CEP-33779 will be presented that uses multiple palladium couplings to provide the drug substance in a convergent manner. The existing medicinal chemistry route was modified to avoid chromatographic purification, improve safety, and utilize palladium ligands which are available in quantities amenable to scale-up. Challenges faced during the development of the new process included optimization of conditions for Buchwald–Hartwig and Suzuki couplings, control of homocoupled impurities and removal of residual palladium. In addition, a screen of conditions to form a diglycolate salt of the parent compound are also presented.

REFERENCES

1: Dugan BJ, Gingrich DE, Mesaros EF, Milkiewicz KL, Curry MA, Zulli AL, Dobrzanski P, Serdikoff C, Jan M, Angeles TS, Albom MS, Mason JL, Aimone LD, Meyer SL, Huang Z, Wells-Knecht KJ, Ator MA, Ruggeri BA, Dorsey BD. A selective, orally bioavailable 1,2,4-triazolo[1,5-a]pyridine-based inhibitor of Janus kinase 2 for use in anticancer therapy: discovery of CEP-33779. J Med Chem. 2012 Jun 14;55(11):5243-54. doi: 10.1021/jm300248q. Epub 2012 May 18. PubMed PMID: 22594690.

2: Tagoe C, Putterman C. JAK2 inhibition in murine systemic lupus erythematosus. Immunotherapy. 2012 Apr;4(4):369-72. doi: 10.2217/imt.12.20. PubMed PMID: 22512630.

3: Seavey MM, Lu LD, Stump KL, Wallace NH, Hockeimer W, O’Kane TM, Ruggeri BA, Dobrzanski P. Therapeutic efficacy of CEP-33779, a novel selective JAK2 inhibitor, in a mouse model of colitis-induced colorectal cancer. Mol Cancer Ther. 2012 Apr;11(4):984-93. doi: 10.1158/1535-7163.MCT-11-0951. Epub 2012 Feb 14. PubMed PMID: 22334590.

4: Lu LD, Stump KL, Wallace NH, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Mason JL, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. Depletion of autoreactive plasma cells and treatment of lupus nephritis in mice using CEP-33779, a novel, orally active, selective inhibitor of JAK2. J Immunol. 2011 Oct 1;187(7):3840-53. doi: 10.4049/jimmunol.1101228. Epub 2011 Aug 31. PubMed PMID: 21880982.

5: Stump KL, Lu LD, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. A highly selective, orally active inhibitor of Janus kinase 2, CEP-33779, ablates disease in two mouse models of rheumatoid arthritis. Arthritis Res Ther. 2011 Apr 21;13(2):R68. doi: 10.1186/ar3329. PubMed PMID: 21510883; PubMed Central PMCID: PMC3132063.

/////////////CEP-33779, CEP33779, CEP 33779, 1257704-57-6, PRECLINICAL, TEVA,  Rheumatoid Arthritis, Colorectal Cancer Therapy, Systemic Lupus Erythematosus,

Jak2 Inhibitors

O=S(C1=CC=C(C2=CC=CN3C2=NC(NC4=CC=CC(N5CCN(C)CC5)=C4)=N3)C=C1)(C)=O

str1 str2

str0

Pfizer’s Fosdagrocorat, PF-04171327 for Rheumatoid Arthritis


Fosdagrocorat, PF-04171327,

CAS 1044535-58-1

(2R,4aS,10aR)-4a-Benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-1,2,3,4,4a,9,10,10a-octahydrophenanthren-2-yl dihydrogen phosphate

2-Phenanthrenecarboxamide, 4b,5,6,7,8,8a,9,10-octahydro-N-(2-methyl-3-pyridinyl)-4b-(phenylmethyl)-7-(phosphonooxy)-7-(trifluoromethyl)-, (4bS,7R,8aR)-

(2R,4aS,10aR)-4a-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-1,2,3,4,4a,9,10,10a-octahydrophenanthren-2-yl dihydrogen phosphate

MF C29H30F3N2O5P
Exact Mass: 574.1844

 

  • PF 04171327
  • PF-04171327
  • UNII-HPI19004QS
  • Selective Glucocorticoid Receptor Modulator

phase 2 .Rheumatoid Arthritis

Glucocorticoid receptor modulators

Pfizer

  • 03 Sep 2015Phase II development of fosdagrocorat is ongoing
  • 01 Jun 2014Pfizer completes a phase II trial in Rheumatoid arthritis in US, Bulgaria, Colombia, the Czech Republic, Germany, Hungary, India, South Korea, Malaysia, Mexico, Poland, Romania, Russia, Serbia, Slovakia, South Africa, Spain and the Ukraine (NCT01393639)
  • 30 Sep 2011Phase-II clinical trials in Rheumatoid arthritis in Bulgaria, Colombia, Germany, India, Malaysia, Mexico, Poland, Romania and South Africa (PO)

 

Fosdagrocorat, also known as PF-04171327, a dissociated agonist of the glucocorticoid receptor (DAGR), a selective high-affinity partial agonist of the GR with potent anti-inflammatory activity at exposures that provide less undesirable effects on bone and glucose metabolism compared with prednisone (pred).

Glucocorticoid receptor modulators are glucocorticoid receptor ligands that are used to treat a variety of conditions because of their powerful anti-inflammatory, antiproliferative and immunomodulatory activity. J. Miner, et al., Expert Opin. Investig. Drugs (2005) 14(12):1527-1545.
Examples of glucocorticoid receptor modulators include dexamethasone, prednisone, prednisolone, RU-486, and as described in WO 2000/66522 and WO 2004/005229.
Treatment with glucocorticoid receptor modulators is often associated with side effects, such as bone loss and osteoporosis.
Identifying a glucocorticoid receptor modulator that is efficacious, potent, and has mitigated side-effects fulfills a medical need.

1044535-58-1.png

SYNTHESIS COMING…………

PATENT

WO 2008093227/US 20100286214

https://www.google.com/patents/WO2008093227A1?cl=en

SCHEME A

The 1 (/?)-Benzyl-5-bromo-9(S)-hydro-10(R)-hydroxy-10(R)-methyl-tricyclo[7.3.1.027]trideca-2,4,6-trien-13-one of Formula A-8 was prepared using the protocol described in Scheme A, which is generally disclosed in WO 00/66522. Ph depicts Phenyl. Bn depicts Benzyl. Compound A-1 can be purchased (for example, VOUS and Riverside; CAS No. 4133-35-1 ). Compound A-2 can be prepared as described in Org. Syn. 1971 , 51 , 109-112.

SCHEME B

The (4βS,7R,8αR)-4β-benzyl-7-hydroxy-Λ/-(2-methylpyridin-3-yl)-7-(trifluoromethyl)-4b,5,6,7,8α,9,10-octahydrophenanthrene-2-carboxamide was prepared as described in Scheme B.

SCHEME C

The (2R,4αS, 10αR)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-1 ,2,3,4,4α,9,10,10α-octahydrophenanthren-2-yl dihydrogen phosphate of C-3 was prepared as described in Scheme C. Bn depicts benzyl.

SCHEME D

The (2R,4αS,10αR)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-1 ,2,3,4,4α,9,10,10α-octahydrophenanthren-2-yl dihydrogen phosphate of C-3 was prepared as described in Scheme D. Bn depicts benzyl. Ph depicts phenyl.

SCHEME E


The (2R,4αS, 10αR)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoy[)-2-(trifluoromethyl)-1 ,2,3,4,4α,9,10,10α-octahydrophenanthren-2-yl dihydrogen phosphate of C-3 was prepared as described in Scheme E. Bn depicts benzyl. Ph depicts phenyl.

Starting Material A-8 is 1(R)~Benzyl-5-bromo-9(S)-hydro-10(R)-hydroxy-10(R)-methyl-tricyclo[7.3.1.027]trideca-2,4,6-trien-13-one as depicted by the following formula:

Preparation 1 : (S)-4a-benzyl-7-bromo-2-ethoxy-3,4,4a,9-tetrahydrophenanthrene

Starting Material A-8 (450 g; 1.17 moles) was dissolved in ethanol (4.5 L) at ambient temperature. 21% sodium ethoxide in ethanol (44 mL; 0.12 moles) was added and the mixture was heated to reflux for three hours. Once the Starting Material A-8 was consumed, the reaction mixture was chilled to -250C. Acetyl chloride (250 mL; 3.51 moles) was slowly added to the mixture while the temperature was maintained near -25°C. After the addition was complete, the mixture was warmed to O0C and held there until the intermediate enone was consumed. The mixture was slurry at this point. 21 % sodium ethoxide in ethanol (1.31 L; 3.51 moles) was added to the mixture while the temperature was maintained between -5°C and 50C. If the mixture was not basic, more sodium ethoxide was added. The temperature of the mixture was increased to 25°C and then diluted with water (5.9 L). The mixture was filtered and the solid was washed with water (3 X). The title compound (440 g; 85 area %) was obtained as a beige solid. 1H NMR (DMSO) δ ppm: 1.27 (t, 3H), 1.65 (dt, 1 H), 2.06 (d, 1 H), 2.21 (dd, 1 H)1 2.49 (m, 1 H), 2.65 (m, 2H), 2.89 (m, 2H), 3.85 (q, 2H), 5.45 (m, 2H), 6.44 (d, 2H), 6.98 (t, 2H), 7.06 (m, 2H), 7.25 (d, 1 H), 7.33 (dd, 1 H).

Preparation 2: (S)-4a-benzyl-7-bromo-2,2-(1,2-ethylenedioxy)-1,2,3,4,4a,9-hexahydrophenanthrene

The (S)-4α-benzyl-7-bromo-2-ethoxy-3,4,4α,9-tetrahydrophenanthrene (1270 g; 3.2 moles; 85 area %, which may be prepared as described in Preparation 1 ) was dissolved in toluene (6.45 L). The ethylene glycol (898 mL; 16.1 moles) and p-toluenesulfonic acid (6.1 g; 0.03 moles) were added and the reaction heated to reflux. Solvent (1 L) was distilled from the mixture and replaced with fresh toluene (1 L). This distillation process was repeated twice more. More p-toluenesulfonic acid (6.1 g) was added each time fresh toluene was added. During the reaction, two intermediates (detected by LC) were formed as the substrate was converted into product. The end point of the reaction was an equilibrium point between the two intermediates and the product. Once the endpoint was reached, the mixture was cooled to ambient temperature. The mixture was washed with 0.5 M NaOH (2 L). The phases separated quickly and both were dark with a small rag layer. The mixture was washed with water (2 L). The phases
separated very slowly. The mixture was dried by azeotropic distillation. Methanol (4 L) was added to the mixture and solvent (4 L) was distilled from the mixture. The methanol addition and solvent distillation were repeated twice more. Methanol was added to the mixture and precipitation occurred a few minutes later. More methanol (4 L) was added to the mixture and then brought to reflux. After 30 minutes, the mixture was cooled to 00C. The mixture was filtered and the solid was washed with chilled methanol (2 X 2L). The solid was dried in a vacuum oven at 65°C. The title compound (882 g; 98 area %) was obtained as a beige solid. 1H NMR (DMSO) δ ppm: 1.71 (m, 2H), 2.06 (m, 2H), 2.31 (dd, 1 H), 2.39 (m, 1 H), 2.68 (d, 1 H), 2.77 (m, 1 H), 2.86 (dd, 1 H), 3.36 (d, 1 H), 3.86 (m, 4H), 5.45 (m, 1 H), 6.50 (m, 2H), 7.00 (m, 4H), 7.37 (dd, 1 H), 7.44 (d, 1 H).

Preparation 3: (S)-methyl 4β-benzyl-7,7-(1,2-ethylenedioxy)-4β,5,6,7,8,10-hexahydrophenanthrene-2-carboxylate

The (S)-4α-benzyl-7-bromo-2,2-(1 ,2-ethylenedioxy)-1 ,2,3,4,4α,9-hexahydrophenanthrene (719 g; 1.75 moles, which may be prepared as described in Preparation 2) was dissolved in tetrahydrofuran (7.19 L) and chilled to -7O0C. The 1.6 M n-butyl lithium in hexane (2270 mL; 2.27 moles) was added at a rate such that the temperature was maintained below -6O0C. The mixture held an additional 15 minutes after the addition. Carbon dioxide (108 g; 2.45 moles) was added while the temperature was maintained below -60°C. The mixture held an additional 15 minutes after the addition. The mixture was warmed to ambient temperature. Solvent (7 L) was distilled from the mixture at atmospheric pressure. DMF (7 L) was added to the mixture. The mixture was cooled to ambient temperature. Methyl iodide (152 mL; 2.45 moles) was added and the mixture was held until the reaction was completed (~1 hour). The mixture was heated to 7O0C and solvent was distilled by gradually reducing the pressure to 70 mmHg. Once distillation had ceased, the mixture was cooled to room
temperature. Water (6.5 L) was slowly added to the mixture to precipitate the product. The mixture was filtered and the solid washed with water (3 X). The solid was dried on the filter. The crude product (736 g; 74 area %) was obtained as a beige solid. The product was purified by chromatography. 463 g of product was recovered from the chromatography. This material was separated from n-heptane (6130 mL). 394 g of the title compound was recovered. Another 70 g of title compound was recovered from the mother liquor by chromatography. 1H NMR (DMSO) δ ppm: 1.74 (m, 2H), 2.10 (m, 2H)1 2.33 (dd, 1 H), 2.45 (m, 1 H), 2.72 (d, 1 H), 2.79 (m, 1 H), 2.94 (dd, 1 H), 3.40 (d, 1 H), 3.87 (m, 7H), 5.49 (m, 1 H), 6.47 (m, 2H), 6.93 (m, 2H), 7.01 (m, 1 H), 7.42 (d, 1 H), 7.64 (d, 1 H), 7.79 (dd, 1 H).

Preparation 4: (4βS,8α/?)-methyl 4β-benzyl-7,7-(1,2-ethylenedioxy)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate

The (S)-methyl 4β-benzyl-7,7-(1 ,2-ethylenedioxy)-4β,5,6,7,8,10-hexahydrophenanthrene-2-carboxylate (201 g; 0.515 moles, which may be prepared as described in Preparation 3) and 50 ml of ethylene glycol was dissolved in toluene (2.0 L) in an autoclave. To this was added 10 grams of a 5% Pd/C (dry catalyst). The autoclave was then sealed and purged with nitrogen (three cycles) followed by hydrogen (three cycles). The reaction was run for 18 hours with a pressure of 80 psig and temperature of 50 0C. HPLC analysis for completion and selectivity (typical selectivity’s are: 95 to 5, Trans to Cis). The suspension was filtered through Celite® to remove the catalyst and the toluene solution is concentrated at 50 0C, under vacuum, to
approximately 200 ml. While still at 50 0C, 1 L of 1-butanol was added and the solution heated to 60 0C, until clear. Upon cooling, the resulting solid title compound was isolated by vacuum filtration (196 grams; 97%; Trans to Cis 95.75 to 4.24). 1H NMR (300 MHz, CDCI3) δ ppm: 7.79 (bs, 1 H1 Ar-H), 7.47 (d, J= 9 Hz, 1 H, Ar-H), 7.13-7.05 (cm, 3H, Ar-H), 6.56-6.53 (cm, 2H, Ar-H), 6.43 (d, J= 9 Hz, 1 H, Ar-H), 4.04-3.93 (cm, 4H, 2-CH2), 3.89 (s, 3H, CH3),3.08-3.03 (cm, 3H, CH2, CH-H), 2.63 (d, J= 15 Hz, CH-H), 2.22-1.72 (cm, 8H, 4-CH2), 1.57 (cm, 1 H, CH-H).; 13CNMR (CDCI3, δ): 167.7, 149.2, 137.7, 136.4, 131.1 , 130.5, 127.8, 127.7, 127.4, 126.3, 125.5, 108.9, 64.6, 64.5, 52.1 , 40.5, 39.8, 38.3, 35.8, 31.6, 30.3, 27.9, 24.6.

Preparation 5: (4βS,8α/?)-methyl 4β-benzyl-7-oxo-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate

ThΘ (4βS,8αR)-mΘthyl 4β-benzyl-7,7-(1 ,2-ethylenΘdioxy)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate (150 g, 382 mmol, which may be prepared as described in Preparation 4) was dissolved in dichloromethane (630 ml). Water (270 ml) was added with stirring followed by trifluoroacetic acid (73 ml. 1150 mmol) via drop funnel over 30 minutes, maintaining the internal temperature below 3O0C. After the addition was complete, the reaction was heated at 4O0C for 2 hours. In process check indicated incomplete reaction with around 9% (area percent) starting material. The layers were separated and fresh water (270 ml) and trifluoroacetic acid (31 ml) was added. The reaction mixture was heated at 4O0C for 1 hour. This process was continued until the starting material was consumed. The organic phase was washed with 5% aqueous sodium bicarbonate (300 ml), water (300 ml) and dried over MgSO4 and concentrated to dryness to give 126.4 g of the title compound (representing a 95% yield). 1H NMR (DMSO) δ ppm: 7.70 (s, 1 H), 7.37 (d, J=8.4 Hz, 1 H), 7.11 (m, 3H), 6.6 (d, J= 5.70 Hz, 2H), 6.45 (d, J=8.4 Hz, 1H), 3.80 (s, 3H), 3.80 (m, 2H), 3.04-1.48 (m, 11 H).

Preparation 6: (4βS,7f?,8α/?)-methyl 4β-benzyl-7-hydroxy-7-(trifluoromethyl)-4β,5J6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate


The (4βS,8αf?)-methyl 4β-benzyl-7-oxo-4β,5,6,7,8I8α,9,10-octahydrophenanthrene-2-carboxylate (118g, 0.339 mole, which may be prepared as described in Preparation 5) dissolved in dichloromethane was chilled to -5O0C. The solution became turbid. 1.0 M Tetrabutylammonium fluoride a solution in THF (3.4 ml, 0.003 mol) was added with no appreciable temperature change. Trifluorotrimethylsilane (79 ml, 0.51 mol) was added over 20 minutes with a color change to bright orange to light red in color. The reaction mixture was held at -50 0C for about 2 hours and then allowed to warm to 0 0C.
Tetrabutylammonium fluoride (340 ml, 0.34 moles) was added very slowly at 0 0C, to the reaction mixture over 45 minutes. An exotherm was observed with gas evolution. The reaction mixture was stirred 10 minutes and HPLC analysis indicated complete desilylialation. Water (1 L) was added to the reaction mixture and with vigorous stirring and allowed to warm to room temperature. The organic layer was washed with water (1 L). The organic layer was concentrated and chromatographed to produce 72 g, 51 % of the title compound, with an additional 32 g of impure product. 1H NMR (DMSO) δ ppm: 7.70 (s, 1 H), 7.37 (d, J=8.1 Hz, 1 H)1 7.09 (m, 3H), 6.5 (dd, J=1.2, 6.6 Hz, 2H), 6.38 (d, J=8.4 Hz, 1 H), 3.80 (s, 3H), 3.80 (m, 2H), 3.09-1.21 (m, 13H).

Preparation 7: (4βS,7/?,8α/?)-methyl 4β-benzyl-7-(bis(benzyloxy)phosphoryloxy)-7-(trifluoromethyl)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate

The (4βS,7R,8αf?)-methyl 4β-benzyl-7-hydroxy-7-(trifluoromethyl)-4β)5,6,7)8,8α,9,10-octahydrophenanthrene-2-carboxylate (5.0 g; 11.9 mmol, which may be prepared as in Preparation 6) and 5-methyltetrazole (3.6 g; 43.0 mmol) were mixed together in dichloromethane (50 mL) at ambient temperature. Dibenzylphosphoramidite (8.3 mL; 25.1 mmol) was added and the mixture was stirred until the reaction was completed (1 hour). The mixture was chilled to 00C and 30% hydrogen peroxide (10 mL) was added. The reaction was stirred until the oxidation was completed (30 minutes). The aqueous phase was separated from the organic phase. The organic phase was washed with 10% sodium meta-bisulfite (50 ml_). The organic phase was dried with anhydrous magnesium sulfate and concentrated. The crude product was purified by silica gel chromatography with 15% ethyl acetate in hexanes. The purified title compound (8.41 g; 94% yield) was obtained as a colorless oil that contained 6% ethyl acetate by weight. 1H NMR (DMSO): δ 1.31 (t, 1 H), 1.63-1.92 (m, 3H), 2.05-2.35 (m, 3H), 2.63 (d, 1 H), 2.75-3.16 (m, 4H), 3.80 (s, 3H), 5.13 (m, 4H), 6.43 (d, 1 H), 6.49 (m, 2H), 7.04-7.17 (m, 3H), 7.33-7.42 (m, 12H), 7.71 (d, 1 H).

Preparation 8: dibenzyl (2f?,4αS,10αR)-4α-benzyl-7-((2-methylpyridin-3-o yl)carbamoyl)-2-(trifluoromethyl)-1 ,2,3,4,4α,9,10,10α-octahydrophenanthren-2-yI phosphate

The (4βS,7R,8αf?)-methyl 4β-benzyl-7-(bis(benzyloxy)phosphoryloxy)-7- (trifluoromethyl)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate (7.9 g; 11.6 5 mmol, which may be prepared as in Preparation 7) and 3-amino-2-picoline (1.3 g; 12.2 mmol) were mixed together in tetrahydrofuran (80 ml_) and chilled to 0°C. The 1 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (24 ml_; 24.4 mmol) was added while maintaining the temperature below 100C. The mixture was stirred for 30 minutes. Water (50 mL) was added to the reaction mixture. The mixture was extracted with ethyl acetate. The organic extract was washed with water. The organic phase was dried with anhydrous magnesium sulfate and concentrated. The crude product was purified by silica gel chromatography with 70% ethyl acetate in hexanes. The purified title compound (6.79 g; 68% yield) was obtained as a yellow gum that contained 6% ethyl acetate by weight. 1H NMR (DMSO): δ 1.33 (t, 1 H), 1.66-1.93 (m, 3H), 2.08-2.34 (m, 3H), 2.41 (s, 3H), 2.68 (d, 1 H), 2.76-3.19 (m, 4H), 5.14 (m, 4H), 6.47 (d, 1 H), 6.56 (m, 2H), 7.07-7.19 (m, 3H), 7.20-7.53 (m, 12H), 7.71 (d, 1 H), 7.76 (s, 1 H), 8.32 (d, 1 H), 9.93 (s, 1 H).

Example 1 : (4βS,7/?,8αR)-4β-benzyl-7-hydroxy-W-(2-methylpyridin-3-yl)-7-(trifluoromethyl)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxamide

The (4βS,7ft,8αR)-methyl 4β-benzyl-7-hydroxy-7-(trifluoromethyl)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxylate (10 g; 23.9 mmol, which may be prepared as described in Preparation 6), and 3-amino-2-picoline (2.71 g; 25.1 mmol) were dissolved in toluene (200 ml_). The 1 M lithium bis(trimethylsilyl)amide in tetrahydrofuran (74.1 mL; 74.1 mmol) was added at a rate such that the temperature was maintained below 350C. There was a mild exotherm and a solid precipitated during the addition. The mixture was held an additional 30 minutes after the addition. Water (250 mL) was added to the mixture. There was a mild exotherm and the solid dissolved. Ethyl acetate (50 mL) was added to the mixture to ensure the product did not precipitate. Stirring was stopped to allow the phases to separate. The aqueous phase was removed. The organic phase was washed with water (250 mL). Solvent (230 mL) was distilled at atmospheric pressure from the organic phase. The mixture was cooled to ambient temperature. The mixture was filtered and the solid was washed with toluene (2 times) followed by heptane (2 times). The solid was dried in a vacuum oven at 700C. The title compound of the present example (10 g) was obtained as a beige solid. 1H NMR (DMSO) δ ppm: 1.32 (m, 1 H), 1.82 (m, 4H), 2.10 (m, 4H), 2.41 (s, 3H), 2.68 (d, 1 H), 3.08 (m, 3H), 6.00 (s, 1H), 6.43 (d, 1 H), 6.59 (m, 2H), 7.12 (m, 3H), 7.25 (dd, 1H), 7.44 (dd, 1H), 7.71 (dd, 1 H), 7.75 (d, 1 H), 8.31 (dd, 1 H), 9.91 (s, 1 H).

Example 2: (2f?,4αS,10αR)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-i ,2,3,4,4α,9,10,1 Oα-octahydrophenanthren-2-yl dihydrogen phosphate

The dibenzyl (2R,4αS, 10αR)-4α-bθnzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2-(trifluoromethyl)-1 ,2,3,4,4a,9,10,10a-octahydrophenanthren-2-yl phosphate (6 g; 7.9 mmol, which may be prepared as described in Preparation 8) was dissolved in methanol (120 ml_). 5% palladium on carbon (63% water) (1.3 g; 0.4 mmol) was added to the mixture. The mixture was treated with hydrogen (50 psi) at room temperature. The reaction stalled with 12% of the monobenzylic intermediate remaining. The mixture was filtered through a pad of Celite®. Fresh catalyst (1.3 g) was added to the solution and resubmitted to the hydrogenation conditions. Once the reaction was completed, the mixture was filtered through a pad of Celite®. The solution was concentrated to about 60 ml_ by distillation and not by using a rotary evaporator. During the distillation a white solid precipitated. The mixture was cooled to ambient temperature. The mixture was filtered and the solid washed with methanol. The solid was dried in a vacuum oven at 700C. The compound of the present example (3.36 g; 75% yield) was obtained as a white solid and had an LC purity of 98 area %. 1H NMR (DMSO): δ 1.33 (t, 1 H)1 1.69-1.98 (m, 3H), 2.07-2.29 (m, 3H)1 2.42 (s, 3H), 2.61-2.80 (m, 2H)1 2.93-3.19 (m, 3H)1 3.30 (d, 1 H), 6.50 (d, 1 H), 6.64 (m, 2H), 7.08-7.20 (m, 3H), 7.29 (dd, 1 H), 7.48 (dd, 1 H), 7.75 (dd, 2H), 8.33 (dd, 1 H), 9.96 (s, 1 H).

 

PATENT

WO 2008093236

http://www.google.co.in/patents/WO2008093236A1?cl=en

 

Example 1 : (4βS,7/?,8α/?)-4β-benzyl-7-hydroxy-N-(2-methylpyridin-3-yl)-7- (trifluoromethyl)-4β,5,6,7,8,8α,9,10-octahydrophenanthrene-2-carboxamide

Figure imgf000042_0001

The (4βS,7R,8α/?)-methyl 4β-benzyl-7-hydroxy-7-(trifluoromethyl)-4β,5,6J7,8,δα,9, 10- octahydrophenanthrene-2-carboxylate (10 g; 23.9 mmol, which may be prepared as described in Preparation 6), and 3-amino-2-picoline (2.71 g; 25.1 mmol) were dissolved in toluene (200 ml_). The 1 M lithium bis(trimethylsilyl)amide in tetrahydrofuran (74.1 ml_; 74.1 mmol) was added at a rate such that the temperature was maintained below 350C. There was a mild exotherm and a solid precipitated during the addition. The mixture was held an additional 30 minutes after the addition. Water (250 ml_) was added to the mixture. There was a mild exotherm and the solid dissolved. Ethyl acetate (50 ml_) was added to the mixture to ensure the product did not precipitate. Stirring was stopped to allow the phases to separate. The aqueous phase was removed. The organic phase was washed with water (250 ml_). Solvent (230 ml_) was distilled at atmospheric pressure from the organic phase. The mixture was cooled to ambient temperature. The mixture was filtered and the solid was washed with toluene (2 times) followed by heptane (2 times). The solid was dried in a vacuum oven at 700C. The title compound of the present example (10 g) was obtained as a beige solid. 1H NMR (DMSO) δ ppm: 1.32 (m, 1H), 1.82 (m, 4H), 2.10 (m, 4H), 2.41 (s, 3H), 2.68 (d, 1 H), 3.08 (m, 3H), 6.00 (s, 1 H), 6.43 (d, 1 H), 6.59 (m, 2H), 7.12 (m, 3H), 7.25 (dd, 1 H), 7.44 (dd, 1 H), 7.71 (dd, 1 H), 7.75 (d, 1 H), 8.31 (dd, 1 H), 9.91 (s, 1 H).

Example 2: (2f?,4αS,10α/?)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2- (trifluoromethyl)-1,2,3,4,4α,9,10,10α-octahydrophenanthren-2-yl dihydrogen phosphate

Figure imgf000043_0001

The dibenzyl (2R,4αS,10αR)-4α-benzyl-7-((2-methylpyridin-3-yl)carbamoyl)-2- (trifluoromethyl)-1 ,2,3,4,4a,9,10,10a-octahydrophenanthren-2-yl phosphate (6 g; 7.9 mmol, which may be prepared as described in Preparation 8) was dissolved in methanol (120 ml_). 5% palladium on carbon (63% water) (1.3 g; 0.4 mmol) was added to the mixture. The mixture was treated with hydrogen (50 psi) at room temperature. The reaction stalled with 12% of the monobenzylic intermediate remaining. The mixture was filtered through a pad of Celite®. Fresh catalyst (1.3 g) was added to the solution and resubmitted to the hydrogenation conditions. Once the reaction was completed, the mixture was filtered through a pad of Celite®. The solution was concentrated to about 60 ml_ by distillation and not by using a rotary evaporator. During the distillation a white solid precipitated. The mixture was cooled to ambient temperature. The mixture was filtered and the solid washed with methanol. The solid was dried in a vacuum oven at 7O0C. The compound of the present example (3.36 g; 75% yield) was obtained as a white solid and had an LC purity of 98 area %. 1H NMR (DMSO): δ 1 .33 (t, 1 H), 1 .69- 1.98 (m, 3H), 2.07-2.29 (m, 3H), 2.42 (s, 3H), 2.61 -2.80 (m, 2H), 2.93-3.19 (m, 3H), 3.30 (d, 1 H), 6.50 (d, 1 H), 6.64 (m, 2H), 7.08-7.20 (m, 3H), 7.29 (dd, 1 H), 7.48 (dd, 1 H), 7.75 (dd, 2H), 8.33 (dd, 1 H), 9.96 (s, 1 H).

REFERENCES

https://www.pfizer.com/sites/default/files/product-pipeline/July%2028%202015%20Pipeline%20Update.pdf

https://clinicaltrials.gov/ct2/show/NCT00938587

////////

Cc1c(cccn1)NC(=O)c2ccc3c(c2)CC[C@H]4[C@]3(CC[C@@](C4)(C(F)(F)F)OP(=O)(O)O)Cc5ccccc5

O=P(O)(O[C@@]1(C(F)(F)F)C[C@@]2([H])CCC3=C(C=CC(C(NC4=CC=CN=C4C)=O)=C3)[C@]2(CC5=CC=CC=C5)CC1)O

 

%d bloggers like this: