New Drug Approvals
Follow New Drug Approvals on WordPress.com

FLAGS AND HITS

Flag Counter
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO

Archives

Categories

Join me on Linkedin

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

Join me on Researchgate

Anthony Melvin Crasto Dr.

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

MYSELF

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

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

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

DR ANTHONY MELVIN CRASTO Ph.D

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

Verified Services

View Full Profile →

Recent Posts

ABBV 744


ABBV-744 Chemical Structure

ABBV 744

N-Ethyl-4-[2-(4-fluoro-2,6-dimethylphenoxy)-5-(1-hydroxy-1-methylethyl)phenyl]-6,7-dihydro-6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridine-2-carboxamide

1H-Pyrrolo[2,3-c]pyridine-2-carboxamide, N-ethyl-4-[2-(4-fluoro-2,6-dimethylphenoxy)-5-(1-hydroxy-1-methylethyl)phenyl]-6,7-dihydro-6-methyl-7-oxo-

Molecular Weight

491.55

Formula

C₂₈H₃₀FN₃O₄

CAS No.

2138861-99-9

ABBV-744 is a highly BDII-selective BET bromodomain inhibitor, used in the research of inflammatory diseases, cancer, and AIDS.

Acute Myeloid Leukemia (AML)

Phase I, AbbVie is evaluating oral agent ABBV-744 in early clinical trials for the treatment of metastatic castration resistant prostate cancer (CRPC) and for the treatment of relapsed or refractory acute myeloid leukemia (AML).

PATENT

WO 2017177955

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017177955&tab=FULLTEXT

Bromodomains refer to conserved protein structural folds which bind to N-acetylated lysine residues that are found in some proteins. The BET family of bromodomain containing proteins comprises four members (BRD2, BRD3, BRD4 and BRDt) . Each member of the BET family employs two bromodomains to recognize N-acetylated lysine residues typically, but not exclusively those found on transcription factors (Shi, J., et al. Cancer Cell 25 (2) : 210-225 (2014) ) or on the amino-terminal tails of histone proteins. Numbering from the N-terminal end of each BET protein the tandem bromodomains are typically labelled Binding Domain I (BDI) and Binding Domain II (BDII) . These interactions modulate gene expression by recruiting transcription factors to specific genome locations within chromatin. For example, histone-bound BRD4 recruits the transcription factor P-TEFb to promoters, resulting in the expression of a subset of genes involved in cell cycle progression (Yang et al., Mol. Cell. Biol. 28: 967-976 (2008) ) . BRD2 and BRD3 also function as transcriptional regulators of growth promoting genes (LeRoy et al., Mol. Cell 30: 51-60 (2008) ) . BET family members were recently established as being important for the maintenance of several cancer types (Zuber et al., Nature 478: 524-528 (2011) ; Mertz et al; Proc. Nat’l. Acad. Sci. 108: 16669-16674 (2011) ; Delmore et al., Cell 146: 1-14, (2011) ; Dawson et al., Nature 478: 529-533 (2011) ) . BET family members have also been implicated in mediating acute inflammatory responses through the canonical NF-KB pathway (Huang et al., Mol. Cell. Biol. 29: 1375-1387 (2009) ) resulting in the upregulation of genes associated with the production of cytokines (Nicodeme et al., Nature 468: 1119-1123, (2010) ) . Suppression of cytokine induction by BET bromodomain inhibitors has been shown to be an effective approach to treat inflammation-mediated kidney disease in an animal model (Zhang, et al., J. Biol. Chem. 287: 28840-28851 (2012) ) . BRD2 function has been linked to pre-disposition for dyslipidemia or improper regulation of adipogenesis, elevated inflammatory profiles and increased susceptibility to autoimmune diseases (Denis, Discovery Medicine 10: 489-499 (2010) ) . The human immunodeficiency virus utilizes BRD4 to initiate transcription of viral RNA from stably integrated viral DNA (Jang et al., Mol. Cell, 19: 523-534 (2005) ) . BET bromodomain inhibitors have also been shown to reactivate HIV transcription in models of latent T cell infection and latent monocyte infection (Banerjee, et al, J. Leukocyte Biol. doi: 10.1189/jlb. 0312165) . BRDt has an important role in spermatogenesis that is blocked by BET bromodomain inhibitors (Matzuk, et al., Cell 150: 673-684 (2012) ) . Thus, compounds that inhibit the binding of BET family bromodomains to their cognate acetylated lysine proteins are being pursued for the treatment of cancer, inflammatory diseases, kidney diseases, diseases involving metabolism or fat accumulation, and some viral infections, as well as for providing a method for male contraception. Accordingly, there is an ongoing medical need to develop new drugs to treat these indications.

FIDANZE, Steven D., et al. BROMODOMAIN INHIBITORS. WO 2017177955 A1.

////////////ABBV 744, Acute Myeloid Leukemia, AML,  Phase 1 , AbbVie

CC(O)(C)C1=CC(C(C2=C3NC(C(NCC)=O)=C2)=CN(C)C3=O)=C(OC4=C(C)C=C(F)C=C4C)C=C1

NIDUFEXOR


Nidufexor Chemical Structure

Nidufexor.png

NIDUFEXOR

LMB763

4-[[benzyl-(8-chloro-1-methyl-4H-chromeno[4,3-c]pyrazole-3-carbonyl)amino]methyl]benzoic acid

Nidufexor is a farnesoid X receptor (FXR) agonist.

Molecular Weight

487.93

Formula

C₂₇H₂₂ClN₃O₄

CAS No.

1773489-72-7

PHASE 2 Treatment of Liver and Biliary Tract Disorders,
Agents for Diabetic Nephropathy, NOVARTIS

Nidufexor

1773489-72-7LMB-763UNII-CJ1PL0TE6JCJ1PL0TE6JBCP28929EX-A1854

Nidufexor pound LMB-763 pound(c)

ZINC584641402

4-((N-benzyl-8-chloro-1-methyl-1,4-dihydrochromeno[4,3-c]pyrazole-3-carboxamido)methyl)benzoic acid

HY-109096

CS-0039398

https://pubs.acs.org/doi/pdf/10.1021/acs.jmedchem.9b01621

1 (7.6 g, 89% yield) as a white solid. Melting point: 232.6 °C.

1 H NMR (400 MHz, DMSO): δ 12.93 (s, 1H), 7.96−7.85 (m, 2H), 7.71 (dd, J = 7.1, 2.5 Hz, 1H), 7.42−7.20 (m, 8H), 7.06 (dd, J = 8.7, 1.9 Hz, 1H), 5.45 (d, J = 3.9 Hz, 2H), 5.25 (d, J = 9.2 Hz, 2H), 4.58 (d, J = 12.1 Hz, 2H), 4.12 (d, J = 16.6 Hz, 3H).

13C NMR (101 MHz, DMSO-d6): δ 167.07, 162.21, 151.98, 142.65, 139.18, 132.20, 132.67, 129.70, 129.50, 129.50, 128.53, 128.53, 127.43, 127.43, 127.43, 127.43, 127.43, 125.53, 122.24, 119.0, 117.09, 116.64, 64.51, 50.68, 48.24. LC-MS m/z: 488.2/490.2 (M +H)+ ; chlorine pattern; method 3; RT = 1.41 min.

Elemental Analysis calcd for C27H22ClN3O4: C 66.46, H 4.54, N 8.61; found: C 66.43, H 4.56, N 8.62.

TRIS Salt Formation. Methanol (400 mL) was added to a mixture of 1 (4.0 g, 8.2 mmol) and 2-amino-2-hydroxymethylpropane-1,3-diol (TRIS, 1.0 g, 8.2 mmol). The mixture was heated to 70 °C for 0.5 h. After cooling to room temperature, the solvent was removed in vacuum. The residue was sonicated in dichloromethane (10 mL) and concentrated again. The resulting white solid was dried under vacuum overnight. The crude material was crystallized by slurring the solid residue in a 4:1 mixture of acetonitrile and methanol (5 mL). The mixture was stirred at room temperature for 24 h to give 4-((N-benzyl-8-chloro-1-methyl-1,4-dihydrochromeno- [4,3-c]pyrazole-3-carboxamido)methyl)benzoic acid TRIS salt as a white salt (3.7 g, 73% yield). Melting point: 195.6 °C. 1 H NMR (400 MHz, DMSO): δ 7.92−7.80 (m, 2H), 7.78−7.64 (m, 1H), 7.41− 7.19 (m, 8H), 7.13−7.00 (m, 1H), 5.44 (s, 2H), 5.25−5.14 (m, 2H), 4.61−4.48 (m, 2H), 4.18−4.03 (m, 3H), 3.39 (s, 7H). TRIS OH masked by water peak. LC-MS m/z: 488.0/490.0 (M+H)+ ; chlorine pattern, method 3. RT = 1.58 min. Elemental Analysis calc for C31H33ClN4O7: C 61.00, H 5.36, N 9.15; found: C 60.84, H 5.34, N 9.13.

https://pubs.acs.org/doi/suppl/10.1021/acs.jmedchem.9b01621/suppl_file/jm9b01621_si_001.pdf

Patent

WO 2015069666

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015069666&tab=PCTDESCRIPTION

///////NIDUFEXOR, LMB 763, Phase II, PHASE 2, Liver and Biliary Tract Disorders,  Diabetic Nephropathy, NOVARTIS

CN1C(C2=CC(Cl)=CC=C2OC3)=C3C(C(N(CC4=CC=CC=C4)CC5=CC=C(C(O)=O)C=C5)=O)=N1

LNP 023


LNP023

4-[(2S,4S)-4-Ethoxy-1-[(5-methoxy-7-methyl-1H-indol-4-yl)methyl]piperidin-2-yl]benzoic acid.png

LNP 023

CAS 1644670-37-0

ROTATION +

4-((2S,4S)-4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl)benzoic acid

M.Wt 422.525
Formula C25H30N2O4

4-[(2S,4S)-4-ethoxy-1-[(5-methoxy-7-methyl-1H-indol-4-yl)methyl]piperidin-2-yl]benzoic acid

LNP023

RENRQMCACQEWFC-UGKGYDQZSA-N

PATENT US9682968, Example-26a

BDBM160475

ZINC223246892

HY-127105

CS-0093107

4-((2S,4S)-(4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl))benzoic acid

4-[(2~{S},4~{S})-4-ethoxy-1-[(5-methoxy-7-methyl-1~{H}-indol-4-yl)methyl]piperidin-2-yl]benzoic acid

LNP023 (LNP-023) is a highly potent, reversible, selective inhibitor of factor B (IC50=10 nM), the proteolytically active component of the C3 and C5 convertases.

LNP023 (LNP-023) is a highly potent, reversible, selective inhibitor of factor B (IC50=10 nM), the proteolytically active component of the C3 and C5 convertases; shows direct, reversible, and high-affinity binding to human FB with Kd of 7.9 nM in SPR assays, demonstrates potent inhibition of AP-induced MAC formation in 50% human serum with IC50 of 0.13 uM; shows no inhibition of factor D (FD), as well as classical or lectin complement pathway activation (up to 100 uM), and no significant effects (up to 10 μM) in a broad assay panel of receptors, ion channels, kinases, and proteases; blocks zymosan-induced MAC formation membrane attack complex (MAC) with IC50 of 0.15 uM, prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats afer oral adminstration; also prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes.

Other Indication

Phase 2 Clinical

PATENT

WO 2015009616

https://patents.google.com/patent/WO2015009616A1/en

PATENT

https://patents.google.com/patent/US9682968B2/en

Example-26Example-26a4-((2S,4S)-(4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl))benzoic acid ((+) as TFA Salt)

Figure US09682968-20170620-C00315

A mixture of methyl 4-((2S,4S)-4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl)benzoate, Intermediate 6-2b peak-1 (tr=1.9 min), (84 mg, 0.192 mmol) and LiOH in H2O (1 mL, 1 mmol) in THF (1 mL)/MeOH (2 mL) was stirred at room temperature for 16 h, and then concentrated. The resulting residue was purified by RP-HPLC (HC-A) to afford the title compound. Absolute stereochemistry was determined by comparison with enantiopure synthesis in Example-26c. 1H NMR (TFA salt, 400 MHz, D2O) δ 8.12 (d, J=8.19 Hz, 2H), 7.66 (br. d, J=8.20 Hz, 2H), 7.35 (d, J=3.06 Hz, 1H), 6.67 (s, 1H), 6.25 (d, J=3.06 Hz, 1H), 4.65 (dd, J=4.28, 11.49 Hz, 1H), 4.04 (d, J=13.00 Hz, 1H), 3.87-3.98 (m, 2H), 3.53-3.69 (m, 5H), 3.38-3.50 (m, 1H), 3.20-3.35 (m, 1H), 2.40 (s, 3H), 2.17-2.33 (m, 2H), 2.08 (br. d, J=15.70 Hz, 1H), 1.82-1.99 (m, 1H), 1.28 (t, J=7.03 Hz, 3H); HRMS calcd. for C26H31N2O(M+H)423.2284, found 423.2263.

PATENT

WO 2020016749

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=D7DA400C5FC41AD0EA9F0AB9B74A1D86.wapp1nB?docId=WO2020016749&tab=PCTDESCRIPTION

The present invention relates to a process for the preparation of phenylpiperidinyl indole derivatives. More particularly, the present invention relates to a process for the preparation of the compound of formula (I)

also referred to as 4-((2S,4S)-(4-ethoxy-1 -((5-methoxy-7-methyl-1 /-/-indol-4-yl)methyl)piperidin-2-yl))benzoic acid, or a pharmaceutically acceptable salt thereof, which is capable of inhibiting the activation of the alternative pathway of the complement system. The complement system plays a major role in the innate and adaptive immunity system and comprises a group of proteins that are normally present in an inactive state. These proteins are organized in three activation pathways: the classical, the lectin, and the alternative pathways (Holers, In Clinical Immunology: Principles and practice, ed. R.R. Rich, Mosby Press; 1996, 363-391 ). Molecules from microorganisms, antibodies or cellular components can activate these pathways resulting in the formation of protease complexes known as the C3-convertase and the C5-convertase. The classical pathway is a calcium / magnesium-dependent cascade, which is normally activated by the formation of antigen-antibody complexes. It can also be activated in an antibody-independent manner by the binding of C-reactive protein complexed to

ligand and by many pathogens including gram-negative bacteria. The alternative pathway is a magnesium-dependent cascade, which is activated by deposition and activation of C3 on certain susceptible surfaces (e.g. cell wall polysaccharides of yeast and bacteria, and certain biopolymer materials). The alternative pathway (AP) utilizes C3 fragments (C3b) to opsonize the pathogens hence targeting them for phagocytosis without the need for antibodies. Hyperactivity of the complement system, and in particular in its AP, plays a role in a large number of complement-driven diseases, such as C3 glomerulopathy (C3G), paroxysmal nocturnal hemoglobinuria (PNH) and IgA nephropathy (IgAN). Phenylpiperidinyl indole derivatives, such as compound of formula (I), or a pharmaceutically acceptable salt thereof, play a role in the inhibition of complement factor B, a known critical enzyme for activation of the alternative complement pathway (Lesavre et al J. Exp. Med. 1978, 148, 1498-1510; Volanakis et al New Eng. J. Med. 1985, 312, 395-401 ), which may also be a suitable target for the inhibition of the amplification of the complement pathways. The phenylpiperidinyl indole derivatives, such as compound of formula (I), or a pharmaceutically acceptable salt thereof, and a method for preparing such derivatives, are described in WO2015/009616. In particular, compound of formula (I) is described in example 26, of WO2015/009616. One of the drawbacks of the synthesis was the use of hazardous chemicals (such as sodium hydride, or dimethylacetamide, which represent safety concerns on a larger scale) and the poor enantio- and diastereo-selectivity of the steps, leading to unwanted stereoisomers.

Thus, there is a need to provide an alternative reaction route in a process for producing compound of formula (I), or a pharmaceutically acceptable salt thereof, generating less by products, and easier to handle on a large scale.

Scheme 1 , vide infra.

Compound of fformula (II)


ormu a ( )


formula (1)

Scheme 1

1. Asymmetric synthesis of compound of formula (II): .

One aspect of the present invention relates to an asymmetric process for preparing a compound of formula (II), or salt thereof, as outlined in Scheme 2 below, wherein the stereocenters in position 2 and in position 4 on the piperidine are obtained in high enantio- and diastereo-selectivity.

formula (ii)

Scheme 2

Example 1 : Synthesis of Benzyl-2-r4-(methoxycarbonyl)phenyl1-4-oxopiperidine-1 -carboxylate according to the following sequence:

R = Methyl R = Methyl R =: Methyl

Step 1 : Synthesis of Benzyl-2-[4-(methoxycarbonyl)phenyl]-4-oxo-3, 4-dihydro pyridine-1(2W)-carboxylate (C3, wherein Pi = Cbz and R = methyl)

iPrMgCI (2N THF, 109.96 g, 54.98 ml_, 2.0 eq) was charged in a reactor. A solution of bis[2 -(N,N-dimethylaminoethyl)] ether (2.5 eq, 22.03 g, 137.46 mmol) in THF (24 ml.) was added at 15 – 25 °C. The mixture was stirred for 1 hour. A solution of C1 (20.17 g, 76.98 mmol, 1 .4 eq) in THF (102 ml.) was added slowly at 15 – 25 °C. The mixture was heated to 25 – 30 °C, stirred for more than 1 hour, and checked by HPLC. The mixture was cooled to -30 °C. A solution of C2 (methyl 4-iodobenzoate, 6.0 g, 54.98 mmol, 1 .0 eq) in THF (20 ml.) was added, followed by a solution of benzyl chloroformate (1 .15 eq, 10.79 g, 63.23 mmol) in THF (36 ml_). The mixture was stirred for 2 hours and quenched with AcOH (6.60 g, 109.96 mmol, 2 eq). Isopropyl acetate (60 ml.) was added. Hydrogen chloride (15%, 90 g) was added to adjust the pH = 1 – 2. The organic layer was separated and washed with brine (15%, 100 g), and concentrated. Isopropyl acetate (160 ml.) was added and concentrated to remove the THF. The crude product was recrystallized in Isopropyl

acetate (1 14 ml.) and n-heptane (120 ml_). The product was dried at 60 °C to provide C3 as light yellow solid (16.0 g, 79.65 % yield). 1 H-NMR (400 MHz, DMSO-d6) d (ppm) = 8.1 1 (dd, J=8.39, 1.01 Hz, 1 H), 7.91 (d, J=8.39 Hz, 2H), 7.33 – 7.37 (m, 6H), 5.82 (d, J= 7.20 Hz, 1 H), 5.20 – 5.35 (m, 3H) , 3.83 (s, 3H), 3.41 (br. s, 1 H), 3.31 (dd, J=16.64, 7.52 Hz, 1 H), 2.66 (br. d, J=16.55 Hz, 1 H).

Step 2: Synthesis of Benzyl-2-[4-(methoxycarbonyl)phenyl]-4-oxopiperidine-1 -carboxylate (C4, wherein Pi = Cbz and R = methyl)

A solution of C3 (25 g, 68.42 mmol, 1 .0 eq) in AcOH (200 ml.) was heated to 50 – 60 °C to form a clear solution. The solution was then cooled to 35 °C. Zn powder (13.42 g, 205.26 mmol, 3.0 eq) was added portionwise while keeping the inner temperature at 35 – 40 °C. After addition, the mixture was stirred for more than 8 hours and checked by HPLC. THF (250 ml.) was added. The mixture was cooled to 25 °C, filtered, and the filter cake was washed with THF (125 volume). The filtrate was concentrated to dryness. Isopropanol (375 ml.) was added. The solution was cooled to 0 – 5 °C. EDTA-4Na.2H20 (40 g) in water (200 ml.) was added. The mixture was neutralized to pH = 9 – 10 with 30% sodium hydroxide solution and stirred for 2 hours. The organic layer was collected, washed with brine (15%, 250 g) and concentrated to about 50 ml_. MTBE (100 ml.) was added and concentrated to about 50 ml_. MTBE (80 ml.) was added followed by n-heptane (20 ml.) dropwise. Then the mixture was cooled to 0 °C gradually. The mixture was filtered and the filter cake was dried to afford C4 as a light yellow solid (20.1 1 g, 80.0 % yield). 1 H NMR (400 MHz, CDCIs) d (ppm)= 7.99 (d, J=8.31 Hz, 2H), 7.27 – 7.39 (m, 7H), 5.83 (br. s, 1 H), 5.14 – 5.28 (m, 2H), 4.20 – 4.42 (m, 1 H), 3.92 (s, 3H), 3.12 – 3.33 (m, 1 H), 2.84 – 3.04 (m, 2H), 2.46 – 2.65 (m, 1 H), 2.23 – 2.45 (m, 1 H).

Example 2: Synthesis of Benzyl -4-hvdroxy-2-(4-(methoxycarbonyl)phenyl)piperidine-1- 

carboxylate (C5. wherein Pi = Cbz and R = methyl)

P1 = Cbz P i = Cbz

R = Methyl R = Methyl

A 0.1 M pH = 7.0 PBS was prepared with disodium phosphate dodecahydrate (22.2 g), sodium dihydrogen phosphate dihydrate (6.2 g) and purified water (999 g). To a reactor equipped with a pH meter 0.1 M pH = 7.0 PBS (499 g), D-glucose (40.2 g, 233.14 mmol, 2.0 eq), NADP (EnzymeWorks, 0.72 g), GDH (EnzymeWorks, 0.41 g) and KRED-EW124 (EnzymeWorks, 2.05 g)

were added, followed by addition of emulsion of C4 (41 g, 1 1 1 .60 mmol, 1 .0 eq) in DMSO (102.5 ml_). The mixture was heated to JT < 45 °C, IT 41 ± 3 °C and stirred at IT 41 ± 3 °C for > 16 h while controlling pH 6.9-7.2 by adding 1 M sodium hydroxide solution. A mixture of NADP (0.29 g), GDH (0.16 g) and KRED-EW124 (0.82 g, #Enzyme Works Inc. China) in 0.1 M pH = 7.0 PBS (1 1 g) were charged and stirred at IT 41 ± 3 °C for > 20 hours. The reaction was monitored by HPLC.

The reaction was filtered to afford white wet cake. To a 1 .0 L Radleys reactor equipped with anchor agitator crude C5 wet cake (80 g) and acetonitrile (500 ml.) were charged. The mixture was stirred to form a light yellow suspension (700 RPM). The suspension was heated to IT = 70 ± 5 °C and stirred for 4 hours, and then cooled to IT = 25 ± 5 °C. The suspension was filtered and the cake was washed with acetonitrile (75 ml_). To a clean 500 ml. Radleys reactor equipped with anchor agitator the resulting mother liquor was charged. The mother liquid was concentrated to about 95 g, solvent exchanged with three portions of toluene (105 g) to 95 g residue. Toluene (170 g) was charged and the reaction was checked by GC (acetonitrile / (toluene + acetonitrile) < 1 .2%). The suspension was heated to IT = 80 ± 5 °C, held for 1 hour, cooled to IT = 45 ± 3 °C and adjusted the agitation speed to low mode. Sequential operations of seeding and aging for 2 hours, charging n-heptane (10.2 g) in 0.5 hours and aging for 1 hour, charging n-heptane (34 g) over 1 .5 hours and aging for 0.5 hours were carried out. The mixture was cooled to IT = 10 ± 3 °C over 7 hours and maintained at 10 ± 3 °C for 2 hours. The mixture was filtered and the cake was washed with cold mixed solvents of toluene (50 ml.) and n-heptane (10 ml.) to afford a light yellow solution of C5 (330 g, trans/cis = 90/10, assay 6.8%, yield 52%). The mother liquor was telescoped to the next step. 1 H-NMR (400 MHz, CDCI3, mixture of two isomers, data for the minor isomer is shown in brackets): d (ppm) = 7.99 (d, J=8.44 Hz, 2H) [7.92 (d, J=8.44 Hz, 0.04H)], 7.23 – 7.39 (m, 7H) [7.10 – 7.18 (m, 0.21 H)], 5.69 (br. s, 1 H) [5.40-5.42 (m, 0.1 1 H)], 5.19 (s, 2H) [5.14 (s, 0.23H)], 4.26 (br. d, J=13.33 Hz, 1 H) [4.18-4.20(m, 0.13H)], 3.91 (s, 3H) [3.90 (s, 0.4H)], 3.67 – 3.79 (m, 1 H) [3.38-3.45 (m, 0.1 1 H)], 2.83 (td, J=13.51 , 2.81 Hz, 1 H), 2.64 (br. d, J=13.33 Hz, 1 H) [2.41 -2.47 (m, 0.12H)], 1 .81-1 .91 (m, 2H) [2.17-2.22 (m, 0.12H)], 1 .72 – 1 .77 (m, 1 H), 1 .45 – 1 .56 (m, 1 H). HRMS: Calcd for C21 H24NO5 (M+H): 370.1654m, found 370.1662.

Example 3: Synthesis of Methyl 4-r(2S,4S)-4-ethoxypiperidin-2-yl1benzoate (Compound of formula according to the following sequence:

R = Methyl R = Methyl R = Methyl

Step 1 : Synthesis of Benzyl (4S)-4-((tert-butyldimethylsilyl)oxy)-2-(4-(methoxycarbonyl) phenyl)piperidine-1 -carboxylate (C8, wherein Pi = Cbz, P2 = TBS and R = methyl).

To a 500 ml. Radleys Reactor charged with C5 in a toluene/heptane solution (1 .0 eq, 145.67 g from previous step, assay 6.07%, 23.94 mmol). The solution was concentrated to about 25 g. Then dichloromethane (1 17.1 g) was charged and the solution was cooled to 23 ± 4 °C. To the clear solution, imidazole (3.42 g, 50.26 mmol, 2.1 eq) and TBS-CI (6.13 g, 40.69 mmol, 1 .7 eq) were introduced. The yellow suspension was stirred at 23 ± 4 °C for 10 hours. The reaction was monitored by HPLC. Then 10% Na2CC>3 (70.7 g) was charged and the mixture was stirred for 1 hours. The organic phase was washed with 5% brine (53 g) and concentrated to about 30 g. Then the solvent was exchange with toluene (45 g) to about 25 g. The residue was diluted with dichloromethane (66 g) and the mixture was filtered through a pad of 200-300 mesh silica gel (1 .66 g). The silica gel was eluted with another portion of dichloromethane (17.5 g). The eluent was concentrated and the residue was subjected to solvent exchange with acetonitrile (71 .1 g + 98.2 g) to 90 g (yield 100%). C8 in acetonitrile solution was used in the next step. 1 H-NMR (400 MHz, CDCI3, mixture of two isomers, data for the minor isomer is shown in brackets): d (ppm) = 8.01 (d, J=8.44 Hz, 2H) [7.94 (d, J=8.44 Hz, 0.17H)], 7.26 – 7.34 (m, 7H) [7.09 – 7.18 (m, 0.13H)], 5.65 (br. d, J=2.04 Hz, 1 H) [5.41 (br. d, J=2.04 Hz, 0.08H)], 5.19 (s, 2H) [5.13 (s, 0.16H)], 4.22 (br. d, J=13.69 Hz, 1 H) [4.10-4.14(m, 0.19H)], 3.92 (s, 3H) [3.90 (s, 0.3H)], 3.62 – 3.69 (m, 1 H) [3.43-3.50 (m, 0.08H)], 2.81 (td, J=13.54, 2.87 Hz, 1 H), 2.49 (br. d, J=13.57 Hz, 1 H) [2.31 -2.35 (m, 0.1 OH)], 1.84-1 .92 (m, 1 H) [2.08-2.14 (m, 0.07H)], 1 .74 – 1 .75 (m, 1 H), 1 .48 – 1 .59 (m, 1 H), 0.86 (s, 9H) [0.56 (s, 0.65H)], 0.03 (s, 3H) [0.09 (s, 0.27H)].

Step 2: Synthesis of Benzyl (4S)-4-ethoxy-2-(4-(methoxycarbonyl)phenyl)piperidine-1 -carboxylate (C9, wherein Pi = Cbz, R = methyl)

To a 250 ml. Radleys Reactor equipped with impeller agitator C8 in acetonitrile solution (135.5 g, assay 12.53%, 35.10 mmol) was charged and rinsed with acetonitrile (with 8.5 g). Et3SiH (12.25 g, 105.31 mmol, 3.0 eq) was charged. The reactor was cooled to IT = 4 ± 5 °C. TESOTf (1 .392 g,

5.265 mmol, 0.15 eq) was charged. A solution of 2,4,6-trimethyM ,3,5-trioxane (4.64 g, 35.10 mmol, 1 .0 eq) in acetonitrile (7.9 g) was added to the mixture in 60 min at IT = 4 ± 5 °C. After addition, the mixture was stirred for 15 min and followed by HPLC. To the reaction mixture was charged 5% aqueous Na2CC>3 (21 .22 g) and water (30 g). Followed by n-heptane (20.4 g) and the mixture was stirred at 25 ± 5 °C for 30 min. Phase cut and the bottom acetonitrile phase was collected. The acetonitrile phase was concentrated to about 65 g. MTBE (100.6 g) and 5% aqueous Na2CC>3 (43.44 g) were charged to the residual acetonitrile solution. The mixture was stirred for 30 min. The upper MTBE phase was collected and filtered via Charcoal film. The charcoal film was washed with MTBE (7.4 g). The mother liquor was concentrated to about 35 g. To the residue methanol (79.2 g) was charged and the solution was concentrated to 70 g. The solution was telescoped to the next step. 1 H NMR (400 MHz, CDCI3, mixture of two isomers, data for the minor isomer is shown in brackets) d (ppm) = 8.01 (d, J= 8.31 Hz, 2H) [7.96 (d, J= 8.31 Hz, 0.21 H)], 7.29 – 7.32 (m, 7H) [7.07 – 7.22 (m, 0.40H)], 5.68 (br. s, 1 H) [5.32 – 5.34 (m, 0.10H)], 5.19 (s, 2H) [5.1 1 (s, 0.19H)], 4.27 (br. d, J=13.08 Hz, 1 H) [4.05 – 4.14 (m, 0.15H)], 3.91 (s, 3H) [3.89 (s, 0.15H)], 3.41 – 3.54 (m, 2H) [3.14 – 3.25 (m, 0.21 )], 3.30 – 3.40 (m, 1 H) [3.86 – 3.75 (m, 0.13H)], 2.84 (td, J=13.51 , 2.81 Hz, 1 H), 2.66 (br. d, J=13.20 Hz, 1 H), 1 .62 – 1 .95 (m, 2H), 1 .40 – 1 .53 (m, 1 H), 1 .18 (t, J= 6.97 Hz, 3H).

Step3: Synthesis of Methyl 4-((4S)-4-ethoxypiperidin-2-yl)benzoate (removal of the protecting group Pi = Cbz – R = methyl)

To a 500 ml. autoclave charged with 10% Pd/C (50% wet, 3.83 g), C9 solution in methanol (assay 19.97%, 192 g, 96.46 mmol) and methanol (28 g). The reactor was purged with vacuum/H2, three times. The mixture was hydrogenated at 3 bar and at a temperature of 25 ± 4 °C for 4 hours. The mixture was filtered and the Pd/C cake was washed with methanol (20 g). The mother liquor was concentrated to 48 g, solvent swapped twice with 142 g isopropyl acetate to 106 g, cooled to 8 ± 5 °C, and 3% hydrogen chloride solution (90.2 g) was added. After phase separation, the aqueous phase was collected and washed with isopropyl acetate (86.4 g). To the aqueous phase MTBE (72 g) and 10% Na2C03 (99.2 g) were added. After phase separation, the aqueous phase was extracted with MTBE (72 g). The combined MTBE phase was washed with water (40 g). The MTBE solution was introduced into the next step. 1 H NMR (400 MHz, CDCI3, mixture of two isomers, data for the minor isomer is shown in brackets) d (ppm) = 7.96 (m, J= 8.31 Hz, 2H), 7.40 – 7.46 (m, 2H), 4.06 (dd, J=1 1 .62, 2.45 Hz, 1 H), 3.88 (s, 3H), 3.70 – 3.79 (m, 1 H) [3.64 – 3.69 (m, 0.12H)], 3.48 -3.56 (m, 2H) [3.38 – 3.45(m, 0.1 1 H)], 3.1 1 – 3.18 (m, 1 H) [3.21 – 3.26 (m, 0.1 1 H)], 2.88 – 2.97 (m, 1 H) [2.73 – 2.80 (m, 0.12H )], 1 .94 – 2.00 (m, 1 H) [ 2.14 – 2.19 (m, 0.10H)], 1.84 – 1 .89 (m, 1 H) [2.02 – 2.07 (m, 0.12H)], 1 .75 (S, 1 H), 1 .65 – 1 .70 (m, 1 H) [1 .45 – 1 .49 (m, 0.10H)], 1 .59 – 1 .64 (m, 1 H) [1 .36 – 1 .42 (m, 0.1 1 H)], 1 .22 – 1 .25 (t, 3H) [1 .17 – 1 .20 (t, J= 6.97, 0.24H)].

Step 4: Synthesis of Methyl 4-[(2S,4S)-4-ethoxypiperidin-2-yl]benzoate (Compound of formula (II) – R = methyl).

To a 500 ml. one neck flask was added the crude solution of step 3 (above) in MTBE (telescoped from last step, 1 10 g, assay 10.52%, light yellow solution, 43.95 mmol). The solution was concentrated to 18.4 g and the solvent was exchanged (JT = 60 °C) with 55 g of n-heptane twice to get 35 g yellow solution. The solution was transferred to 100 ml. Easy Max equipped with impeller agitator. The solution was heated to 50 °C with 300 RPM , aged for 30 min, cooled to 41 ± 2 °C and seed was added. The agitation was adjusted to low speed. The mixture was aged at 41 ± 2 °C for 2 hours, cooled to 35 ± 2 °C in 8 – 10 hours and then aged at 35 ± 2 °C for 1 – 2 hours n-heptane (7.9 g) was added dropwise. The agitation was adjusted to medium speed. The mixture was cooled to IT = 25 ± 2 °C in 1 hour and aged at 25 ± 2 °C for 10 – 20 minutes. The mixture was filtered. The filtrate was re-charged to the reactor for rinsing the solid on the reactor wall. The mixture was filtered and the filter cake was washed with pre-cooled (-5 °C) n-heptane (7.9g). The cake was dried at 40 °C for > 10 hours to afford 6.4 g of white solid (50% yield). 1H NMR (400 MHz, CDCIs) d (ppm) = 7.99 (m, J=8.31 Hz, 2H), 7.45 (m, J=8.19 Hz, 2H), 4.09 (dd, J=1 1 .62, 2.20 Hz, 1 H), 3.90 (s, 3H), 3.75 (t, J=2.81 Hz, 1 H), 3.53 (q, J= 6.97 Hz, 2H), 3.17 (td, J=12.13, 2.63 Hz, 1 H), 2.91 – 2.99 (m, 1 H), 1.99 (dd, J=13.57, 2.69 Hz, 1 H), 1 .88 (dt, J=13.79, 2.58 Hz, 1 H), 1 .69 – 1 .79 (m, 1 H), 1 .57 – 1 .68 (m, 2H), 1 .25 (t, J= 7.03 Hz, 3H).

Example 4: Enantioselective synthesis of compound according to the following

sequence:

Step 1 : Synthesis of Benzyl 4-oxo-3,4-dihydropyridine-1 (2H)-carboxylate (C6, wherein Pi = Cbz and R = methyl)

To a 2.0 L reactor, 4-methoxypyridine (C1 , 45.0 g, 412.39 mmol, 1 .0 eq) and methanol (900 ml.) were added. The mixture was cooled to -75 °C with dry ice/acetone bath. A solution of benzyl

chloroformate (73.86 g, 432.99 mmol, 1 .05 eq) in THF (90 ml.) was charged dropwise while keeping IT < -70 °C. The reaction was stirred for 1 hour to afford a white suspension at -70 °C. Sodium borohydride (16.38 g, 432.99 mmol, 1 .05 eq) was added in portions while keeping IT < -70 °C. The reaction was stirred at -70 °C for 2 hours. Water (200 g) was added and the cooling bath was removed. A solution of 36% hydrogen chloride (16.72 g, 164.95 mmol, 0.4 eq) in water (50 ml.) was added in 10 min at 0 – 5 °C and stirred for 1 hour. Then 20% Na2CC>3 (85.5 g) was added to adjust pH = 7 while maintained IT < 5 °C. Organic solvents were removed under vacuum. The resulting residue was extracted with dichloromethane (450 ml_). The dichloromethane phase was washed with 3wt% hydrogen chloride (151 ml.) and 3 wt% Na2C03 (151 ml_). After solvent exchange with MTBE, about 4 volume (180 ml) of the MTBE mixture was obtained. The mixture was heated to 50 °C to afford a solution and then cooled to 45 °C. Crystal seed of C6 was charged and the mixture was aged at 40 – 45 °C for 7 hours. The mixture was cooled to 10 – 15 °C in 3 hours. The white suspension was filtered and the wet cake was rinsed with cold MTBE (45 ml_). The cake was dried under vacuum at 40 – 50 °C for 2 hours to afford C6 as a white powder (91.56 g, 60% yield). 1H NMR (400 MHz, CDCI3): d (ppm) = 7.85 (br. s, 1 H), 7.37 – 7.43 (m, 5H), 5.43 (br. s, 1 H), 5.26 (s, 2H), 4.05 (t, J=7.34 Hz, 2H), 2.54 – 2.58 (m, 2H).

Step 2: Synthesis of Benzyl (S)-2-(4-(methoxycarbonyl)phenyl)-4-oxopiperidine-1 -carboxylate ((S)-C4, wherein Pi = Cbz and R = methyl)

Method 1 : A 500 ml Radleys reactor was purged 3 times with vacuum/N2. C6 (8 g, 34.60 mmol, 1.0 eq), C7 (9.34 g, 51.89 mmol, 1 .5 eq), tert- Amyl alcohol (160 ml.) and deionized water (16 ml.) were added. The mixture was stirred for > 40 minutes to give a clear colorless solution. The solution was purged 4 times with vacuum / N2 and bubbled with N2 via a syringe needle for 1 hour. To the colorless solution was charged the mixed solid of (S)-XylBINAP (0.381 g, 0.519 mmol, 0.015 eq) and Rh(Acac)(C2H4)2 (0.134 g, 0.519 mmol, 0.015 eq). The mixture was continued to bubble with N2 for 15 minutes and purged 4 times with vacuum / N2. The suspension was stirred for another 2 hours to dissolve (S)-XylBINAP. The reaction mixture was stirred at 55 ± 4 °C for 15 hours. The reaction was followed by HPLC. The mixture was cooled and treated with 7.7% sodium hypochlorite (1 g, 1 .04 mmol, 0.03 eq) for 1 .5 hours at 40 ± 4 °C. tert- Amyl alcohol was distilled off. The residue was extracted with isopropyl acetate (64 ml.) and ethyl acetate (8 ml.) and filtered. The organic phase was washed with 5% NaHC03 (50 g) then with 15% brine (40 g) at 50 ± 5 °C. Some solvents were removed and ethyl acetate (21 .6 g) was added. The solution was treated with Smopex-234 (1 .2 g) at IT =55 ± 5 °C for 2 hours then filtered via 200 – 300 mesh silica gel (1 .6 g). After solvent exchange with n-heptane, MTBE (44.4 g) was added. The mixture was cooled to IT = 42 ± 3 °C. (S)-C4 seed (10 mg) was added. The mixture was aged for 2 hours and cooled to IT =

31 ± 3 °C in 3 hours n-heptane (23.2 g) was then charged in 1 – 2 hours. The mixture was aged for 2 hours and cooled to IT = 20 ± 3 °C in 2 hours. The mixture was filtered and the cake was washed with a mixed solvent of MTBE (4.4 g) and n-heptane (4.1 g). Dried the wet cake at 60 °C for > 5 hours to afford (S)-C4 (7.63 g, 60% yield) as yellow powder. 1H NMR (400 MHz, CDCI3): d (ppm) = 7.99 (d, J=8.44 Hz, 2 H), 7.28 – 7.37 (m, 7H), 5.82 (br. s, 1 H), 5.14 – 5.28 (m, 2H), 4.30 (br. s, 1 H), 3.91 (s, 3H), 3.22 (br. d, J=8.31 Hz, 1 H), 2.84 – 3.03 (m, 2H), 2.46 – 2.64 (m, 1 H), 2.38 (br. d, J=16.26 Hz, 1 H).

Method 2: To a 500 ml Radleys reactor purged 3 times with vacuum/N2, C6 (8 g, 34.60 mmol, 1 .0 eq), C7 (9.34 g, 51 .89 mmol, 1 .5 eq), fe/f-Amyl alcohol (160 ml.) and deionized water (16 ml.) were added. The mixture was stirred for roughly 40 minutes to give a clear colorless solution. The solution was purged 4 times with vacuum / N2 and bubbled with N2 via a syringe needle for 1 hour. To the colorless solution, was charged the mixed solid of (R, R)-Ph-BPE-Rh(Acac) (0.005 eq., 0.122 g, 0.173 mmol). The mixture was continued to bubble with N2 for 15 minutes and purged with vacuum / N2. The reaction mixture was stirred at 55 ± 4 °C for 15 hours. The reaction was followed by HPLC. Tert- amyl alcohol was distilled off. The residue was extracted with isopropyl acetate (64 ml.) and ethyl acetate (8 ml_), and then filtered. The organic phase was washed with 5% NaHC03 (50 g), then with 15% brine (40 g) at 50 ± 5 °C. Some solvents were removed and ethyl acetate (21 .6 g) was added. The solution was treated with Smopex-234 (1 .2 g) at IT = 55 ± 5 °C for 2 hours then filtered via 200 – 300 mesh silica gel (1 .6 g). After solvent exchange with n-heptane, MTBE (44.4 g) was added. The mixture was cooled to IT = 42 ± 3 °C. (S)-C4 seed (10 mg) was added. The mixture was aged for 2 hours and cooled to IT = 31 ± 3 °C in 3 hours n-heptane (23.2 g) was then charged in 1 – 2 hours. The mixture was aged for 2 hours and cooled to IT = 20 ± 3 °C in 2 hours. The mixture was filtered and the cake was washed with a mixed solvent of MTBE (4.4 g) and n-heptane (4.1 g). The wet cake was dried at 60 °C for roughly 5 hours to afford (S)-C4 (10.17 g, 80% yield) as yellow powder. 1 H NMR (400 MHz, CDCI3) d (ppm) = 7.99 (d, J=8.44 Hz, 2 H), 7.28 – 7.37 (m, 7H), 5.82 (br. s, 1 H), 5.14 – 5.28 (m, 2H), 4.30 (br. s, 1 H), 3.91 (s, 3H), 3.22 (br. d, J=8.31 Hz, 1 H), 2.84 – 3.03 (m, 2H), 2.46 – 2.64 (m, 1 H), 2.38 (br. d, J=16.26 Hz, 1 H).

Method 3: To a 500 ml Radleys reactor purged 3 times with vacuum/N2. C6 (8 g, 34.60 mmol, 1 .0 eq), C7 (9.34 g, 51 .89 mmol, 1 .5 eq), tert- amyl alcohol (160 ml.) and deionized water (16 ml.) were added. The mixture was stirred for roughly 40 minutes to give a clear colorless solution. The solution was purged 4 times with vacuum / N2, and bubbled with N2 via a syringe needle for 1 hour. To the colorless solution was charged the mixed solid of (S)-XylBINAP-Rh(Acac) (0.01 eq., 0.324

g, 0.346 mmol). The mixture was continued to bubble with N2 for 15 minutes and purged with vacuum / N2. The reaction mixture was stirred at 55 ± 4 °C for 15 hours. The reaction was followed by HPLC. Tert- amyl alcohol was distilled off. The residue was extracted with isopropyl acetate (64 mL) and ethyl acetate (8 mL), and then filtered. The organic phase was washed with 5% NaHC03 (50 g), then with 15% brine (40 g) at 50 ± 5 °C. Some solvents were removed and ethyl acetate (21 .6 g) was added. The solution was treated with Smopex-234 (1 .2 g) at IT =55 ± 5 °C for 2 hours then filtered via 200 – 300 mesh silica gel (1 .6 g). After solvent exchange with n-heptane, MTBE (44.4 g) was added. The mixture was cooled to IT = 42 ± 3 °C. (S)-C4 seed (10 mg) was added. The mixture was aged for 2 hours and cooled to IT = 31 ± 3 °C in 3 hours n-heptane (23.2 g) was then charged in 1 – 2 hours. The mixture was aged for 2 hours and cooled to IT = 20 ± 3 °C in 2 hours. The mixture was filtered, and the cake was washed with a mixed solvent of MTBE (4.4 g) and n-heptane (4.1 g). The wet cake was dried at 60 °C for roughly 5 hours to afford (S)-C4 (10.30 g, 81 % yield) as yellow powder. 1H NMR (400 MHz, CDCI3) d (ppm) = 7.99 (d, J=8.44 Hz, 2 H), 7.28 – 7.37 (m, 7H), 5.82 (br. s, 1 H), 5.14 – 5.28 (m, 2H), 4.30 (br. s, 1 H), 3.91 (s, 3H), 3.22 (br. d, J=8.31 Hz, 1 H), 2.84 – 3.03 (m, 2H), 2.46 – 2.64 (m, 1 H), 2.38 (br. d, J=16.26 Hz, 1 H).

Example 5: Synthesis of Benzyl -4-hvdroxy-2-(4-(methoxycarbonyl)phenyl)piperidine- 

1-carboxylate f(S)-C5, wherein Pi = Cbz and R = methyl)

R = Methyl R = Methyl

Preparation of 0.1 M PBS, pH 7.0, with 0.1 % TPGS buffer solution: To a 500 ml. Radleys reactor equipped with impeller agitator was charged Na2HP04.12H20 (8.63 g), NaH2P04.2H20 (2.41 g), Tap Water (388.6 g) and TPGS-750-M.001 (0.388 g). The mixture was stirred for > 3 hours at IT = 60 ± 5 °C and then cooled to IT = 51 ± 3 °C. 80 g of the buffer solution was taken from the reactor to a flask and cooled to < 35 °C. Check pH value of the buffer solution (7.0 ± 0.5). To the above Radleys reactor (S)-C4 (20.0 g, 54.4 mmol, 1 .0 eq), Isopropanol (16.36 g, 272.2 mmol, 5.0 eq) and 0.1 % TPGS buffer solution (60 g) were added. To a 25 mL flask was charged KRED-P3-G09 (0.4 g, #Codexis), NADP+ (0.1 g) and 0.1 % TPGS buffer solution (60 g) from the above flask. All the solid was dissolved. The solution of enzyme was charged to the 500 mL Reactor at IT =50 ± 5 °C. Rinsed the 25 mL flask with 0.1 % TPGS buffer (10 g) and transferred the solution to the 500 mL reactor at IT =50 ± 5 °C. The mixture was stirred with agitation speed > 500 RPM at 51 ± 3 °C for >

8 hours. The reaction was followed by HPLC. To the reactor 2-MeTHF (200 mL) was added and the mixture was stirred for > 60 minutes at 50 ± 5 °C. The mixture was held for > 50 minutes without agitation and the bottom aqueous phase was separated. The organic phase was washed twice with another 200 g of water at 50 ± 5 °C. The organic phase was concentrated to about 70 g. After solvent exchange with twice 158 g acetonitrile to give about 80 g solution, which was cooled to < 30 °C then filtered via MCC. MCC cake was washed with isopropyl acetate (40 mL/35.5 g) to afford (S)-C5 in a light color solution (1 14.3 g, assay 16.95% 96.34% yield). The acetonitrile / isopropyl acetate solution was telescoped to the next step directly. 1 H NMR (400 MHz, CDCI3): d (ppm) = 7.98 (d, J=8.44 Hz, 2H), 7.23 – 7.38 (m, 7H), 5.61 – 5.72 (m, 1 H), 5.18 (s, 2H), 4.23 (br. d, J=13.33 Hz, 1 H), 3.90 (s, 3H), 3.62 – 3.75 (m, 1 H), 2.81 (td, J=13.51 , 2.81 Hz, 1 H), 2.62 (br. d, J=13.33 Hz, 1 H), 2.45 (br. s, 1 H), 1 .79 – 1 .91 (m, 2H), 1 .41 – 1 .56 (m, 1 H).

Example 6: Asymmetric synthesis of Methyl 4-r(2S.4S)-4-ethoxypiperidin-2-yl1benzoate

(Compound of formula . or a salt thereof. – R= methyl) according to the following

sequence:

(S)-C5 (S)-C9 Compound of (Pi = Cbz) (Pi = Cbz, P2 = TBS) (Pi = Cbz) formula (II) R = Methvl R = Methyl R = Methyl R = Methyl

Step 1 : Synthesis of Benzyl (2S,4S)-4-{[tert-butyl(dimethyl)silyl]oxy}-2-[4-(methoxy carbonyl) phenyl]piperidine-1 -carboxylate ((S)-(C8), wherein Pi = Cbz, P2 = TBS, and R = methyl).

To a 500 ml Radleys Reactor was charged with (S)-C5 solution (in acetonitrile / isopropyl acetate, 271 .8 g, assay 14.72%, contained 40.0 g of (S)-C5, 108.31 mmol, 1 .0 eq) from the previous step. After solvent exchange with isopropyl acetate (159.8 g / 180 ml_), 100 g clear solution was obtained. Isopropyl acetate (176 g /198 ml_), imidazole (26.54 g, 389.90 mmol, 3.6 eq) and TBS-CI (27.75 g, 184.12 mmol, 1 .7 eq) were added. The yellow suspension was stirred at 55 ± 4 °C for 7 hours. The reaction was followed by HPLC. The reaction mixture was cooled to 23 ± 4 °C and filtered through MCC (2 g). The cake was washed with isopropyl acetate (88.8 g / 100 ml_). 6% NaHC03 (240 g) was added and the mixture was stirred for 20 minutes. The organic phase was washed with 5% brine (2×240 g) and concentrated to about 105 g. After solvent exchange with toluene (120 g / 135.4 ml_), 105 g solution was obtained. Dichloromethane (298 g / 224.5 ml.) was added and the solution was filtered via 200-300 mesh silica gel (4.4 g). The silica gel was eluted with another portion of dichloromethane (44 g / 33 ml_). The mother liquor was concentrated and the solvent was exchanged with acetonitrile (2×280 ml_, 442.4 g in total) to 100 g. The residue was diluted with acetonitrile (105 g / 132.9 ml.) to afford a light yellow solution (205 g, assay 25.55%, 100% yield), which was used for the next step directly. 1 H NMR (400 MHz, CDCI3) d (ppm) = 8.01 (d, J=8.44 Hz, 2 H), 7.23 – 7.37 (m, 7 H), 5.60 – 5.70 (m, 1 H), 5.18 (s, 2H), 4.22 (br. d, J=13.45 Hz, 1 H), 3.90 (s, 3H), 3.62 – 3.71 (m, 1 H), 2.82 (td, J=13.51 , 2.81 Hz, 1 H), 2.49 (br. d, J=13.45 Hz, 1 H), 1.83 – 1 .96 (m, 1 H), 1 .75 – 1 .80 (m, 1 H), 1 .47 – 1.60 (m, 1 H), 0.86 (s, 9H), 0.03 (s, 3H), 0.00 (s, 3H).

Step 2: Synthesis of Benzyl (2S, 4S)-4-ethoxy-2-[4-(methoxycarbonyl)phenyl]piperidine-1 -carboxylate ((S)-C9, wherein Pi = Cbz amd R = methyl)

To a 500 ml. Radleys Reactor equipped with impeller agitator (S)-C8 in an acetonitrile solution (170.8 g, assay 29.28%, 103.38 mmol, 1 .0 eq) and fresh acetonitrile (220 g) were charged, followed by Et3SiH (36.06 g, 310.13 mmol, 3.0 eq). The mixture was cooled to IT =4 ± 5 °C and TESOTf (5.47 g, 20.68 mmol, 0.2 eq) was charged. To the mixture was charged a solution of 2,4,6-trimethyl-1 ,3,5-trioxane (13.66 g, 103.38 mmol, 1 .0 eq) in acetonitrile (23 g) over 60 minutes at IT =4 ± 5 °C. Upon addition, the mixture was stirred for 15 minutes. The reaction was followed by HPLC. To the reaction mixture was charged 5% aqueous sodium hydroxide (16.54 g, 20.68 mmol, 0.2 eq) and 20 g water, followed by n-heptane (60 g). The mixture was stirred for 30 minutes at 20 ± 5 °C. The bottom acetonitrile phase was collected. To the acetonitrile phase was charged with MTBE (1 1 1 g) and 10% brine (300 g). The mixture was stirred for 30 minutes. The upper MTBE phase was washed with 10% brine (2×300 g), concentrated to 90 g. MTBE (185 g) and water (150 g) were charged. After phase separation at 38 ± 4 °C and solvent exchange of the organic layer with isopropyl acetate (2×266.4 g), 205 g solution was obtained, which was filtered through Charcoal film slowly. The charcoal film was washed with isopropyl acetate (22.2 g) to afford as a light yellow solution (223 g, 100% yield). The solution was telescoped to the next step directly. 1 H NMR (400 MHz, CDCI3) d (ppm) = 8.01 (d, J=8.44 Hz, 2H), 7.25 – 7.38 (m, 7H), 5.68 (br. s, 1 H), 5.19 (s, 2H), 4.27 (br. d, J=13.33 Hz, 1 H), 3.92 (s, 3H), 3.42 – 3.54 (m, 2H), 3.34 (ddd, J=10.88, 6.91 , 4.22 Hz, 1 H), 2.84 (td, J=13.51 , 2.81 Hz, 1 H), 2.66 (br. d, J=13.20 Hz, 1 H), 1 .96 (br. d, J=10.51 Hz, 1 H), 1 .75 – 1 .90 (m, 1 H), 1 .33 – 1 .53 (m, 1 H), 1 .18 (t, J= 6.97 Hz, 3H).

Step 3: Synthesis of Methyl 4-((2S,4S)-4-ethoxypiperidin-2-yl)benzoate (compound of Formula (II), or a salt thereof – R= methyl)

To a 500 ml. autoclave which was purged with vacuum / N2 (S)-C9 in an isopropyl acetate solution (278.4 g, assay 17.96%, 50 g of (S)-C9, 125.80 mmol) and 10% Pd/C (5.0 g, 50% wet) were

charged. The reactor was purged with vacuum / H2 and stirred for > 7 hours at 25 ± 5 °C. The reaction was followed by HPLC analysis. Filtered the reaction mixture via MCC (7.7 g) which was pre-washed with isopropyl acetate . Rinsed the reactor and MCC with isopropyl acetate (39 g). The mother liquor was combined to afford compound of formula (II) as a light yellow solution (315 g, assay 10.0%, 95.1 % yield). 1 H NMR (400 MHz, CDCI3) d (ppm) = 7.99 (m, J=8.31 Hz, 2H), 7.45 (m, J=8.19 Hz, 2H), 4.09 (dd, J=1 1 .62, 2.20 Hz, 1 H), 3.90 (s, 3H), 3.75 (t, J=2.81 Hz, 1 H), 3.53 (q, J= 6.97 Hz, 2H), 3.17 (td, J=12.13, 2.63 Hz, 1 H), 2.91 – 2.99 (m, 1 H), 1 .99 (dd, J=13.57, 2.69 Hz, 1 H), 1 .88 (dt, J=13.79, 2.58 Hz, 1 H), 1 .69 – 1 .79 (m, 1 H), 1 .57 – 1 .68 (m, 2H), 1 .25 (t, J= 7.03 Hz, 3H).

Step 4: Synthesis of the maleic salt of compound of formula (II) (R = methyl)

To a 500 ml. Radleys Reactor equipped with impeller agitator a solution of methyl 4-((2S,4S)-4-ethoxypiperidin-2-yl)benzoate (381 g, assay 10.03%, 145.12 mmol, 1 .0 eq) from the previous step was charged. The solution was concentrated to 281 g and fresh isopropyl acetate (28.6 g) was added. Then a solution of maleic acid (8.45 g, 72.56 mmol, 0.5 eq) in acetone (30.5 ml.) was added at 51 ± 3 °C in 30 minutes. After stirring for 15 minutes, a seed of the maleic salt of compound of formula (II) was added and the mixture was aged for 2 hours. A solution of maleic acid (8.45 g, 72.56 mmol, 0.5 eq) in acetone (30.5 ml.) was charged at 51 ± 3 °C in 60 minutes and the mixture was aged for 2 hours. The mixture was cooled to IT = 10 ± 3 °C in 6 hours and stirred for > 120 minutes. The mixture was filtered and the filter cake was washed with pre-cooled isopropyl acetate (44.4 g). The cake was dried under high vacuum at 55 °C for 5 – 12 hours to afford maleic salt of compound of formula (II) as white solid (49.8 g, Yield 90.4%). 1 H NMR (400 MHz, CDCIs) d (ppm) 9.35 – 9.78 (m, 2H), 8.02 (m, J=8.31 Hz, 2H), 7.58 (m, J=8.31 Hz, 2H), 6.17 (s, 2H), 4.56 (br. d, J=1 1.13 Hz, 1 H), 3.90 (s, 3H), 3.86 (s, 1 H), 3.48 – 3.57 (m, 2H), 3.38 – 3.44 (m, 2H), 2.42 (br. t, J=13.57 Hz, 1 H), 1 .98 – 2.20 (m, 3H), 1 .24 (t, J= 6.97 Hz, 3H).

The maleic salt of compound of formula (II) may be characterized by a x-ray powder diffraction pattern (XRPD) comprising four or more 2Q values (CuKa l=1 .5418 A) selected from the group consisting of 5.893, 6.209, 1 1 .704, 13.014, 16.403, 17.295, 17.592, 18.629, 18.942, 21 .044, 21 .733, 21 .737, 22.380, 23.528, 24.195, 26.013, 26.825, 29.017, 29.515, 32.250, 35.069, 35.590, and 37.932, measured at a temperature of about 22 °C and an x-ray wavelength, l, of 1 .5418 A.

Example 7: Synthesis of fert-butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate

(Compound of formula (III), or a salt thereof) according to the following seguence:

Step 1 : Synthesis of 7-methyl-1 H-indol-5-ol (C11 )

To a 250 ml. flask equipped with a thermometer 3.4% Na2HP04 (100 g, pH = 8.91 ) was charged, followed by addition of Fremy’s salt (4.84 g, 2.4 eq). The mixture was stirred at 20 ± 5 °C until a clear solution was formed. A solution of 7-methylindoline in acetone (9.1 g, 1 1 %) was added in one portion. The mixture was stirred at 20 ± 5 °C for 1 .5 hours. Then sodium sulfite (0.38 g) was added. The mixture was extracted with ethyl acetate (100 ml. x 2) The combined organic extracts were dried over anhydrous sodium sulfate, filtered and concentrated. To the residue 20ml_ acetonitrile was added. The solution was used directly in the next step.

Step 2: Synthesis of fert-butyl 5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C12, wherein P3 = Boc)

The above as prepared solution was cooled to 0 ± 5 °C. DMAP (0.34 g, 0.4 eq) was charged followed by addition of (Boc)20 (4.9 g, 3.0 eq). The mixture was warmed to 20 ± 5 °C, stirred at 20 ± 5 °C for 30 minutes and concentrated. To the residue was added methanol (40 ml_). The mixture was cooled to 0 ± 5 °C. Potassium carbonate (5.1 g, 5.0 eq) was added. The mixture was stirred at 0 ± 5 °C for 4 hours, warmed to 20 ± 5 °C and stirred for additional 2 hours. The mixture was cooled to 0 ± 5 °C. Acetic acid (2 g) was added. pH was 7-8. The mixture was filtered and the filter cake was washed with methanol (10 mL x 2). The filtrate was concentrated and ethyl acetate (30 ml.) was added. The mixture was washed with water (20 ml.) and 5% brine (20 ml_). The organic layer was concentrated to afford a dark oil, which was slurried with (3:2) n-heptane: Ethyl acetate (5 g) to afford a yellow solid. The solid was collected by filtration and dried to give C12 as yellow solid. 27.4% isolate yield from C10. 1 H-NMR (400 MHz, DMSO-d6): d (ppm) = 9.13 (s, 1 H), 7.52 (d, J= 3.67 Hz, 1 H), 6.74 (d, J= 2.2 Hz, 1 H), 6.56 (m, 1 H), 6.50 (d, J= 3.67 Hz, 1 H), 2.45 (s, 3 H), 1.57 (s, 9 H). LCMS (m/z): positive mode 248.1 [M]+, LCMS (m/z): negative mode 246.1 [M-1 ]-.

Step 3: Synthesis of fert-butyl 4-formyl-5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C13, wherein P3 = Boc)

To a solution of fe/f-butyl 5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C12) (53.8% assay, 1 .0 g, 2.2 mmol) in THF (20 ml.) was added dropwise the solution of CH3MgBr in THF (1 N, 2.2 ml_, 2.2 mmol). The resulting mixture was stirred at 20 – 25 °C for 10 minutes. (CHO)n (0.2 g, 6.53 mmol)

was added to the mixture. The reaction mixture was heated to 65 – 70 °C and stirred for 1 hours. The reaction mixture was cooled to 20 – 25 °C. Saturated NH4CI (20 ml.) and MTBE (20 ml.) were added. The mixture was separated and the aqueous layer was extracted with MTBE (20 ml_). The organic layers were combined and concentrated to give compound C13 as yellow solid (0.7 g, 79% assay, 92% yield). 1H-NMR (400 MHz, DMSO-d6) d (ppm) = 10.74 (s, 1 H), 10.54 (s, 1 H), 7.82 (d, J= 4.0 Hz, 1 H), 7.34 (d, J= 4.0 Hz, 1 H), 6.81 (s, 1 H), 2.59 (s, 3H), 1 .65 (s, 9H). LCMS (m/z): positive mode 290.1 [M]+.

Step 4: Synthesis of fert-Butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate (Compound of formula (III)).

To a solution of compound C13 (50 mg, 0.182 mmol) in dry DMF (3 ml.) was added K2CO3 (50.2 mg, 0.363 mmol). The mixture was stirred for 10 minutes and then dimethyl sulfate (25.2 mg, 0.20 mmol) was added. The reaction mixture was stirred for 1 hours and poured into ice-water (12 ml_). The mixture was filtered and the filter cake was washed with water. The cake was dried under vacuum to give tert- Butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate (Compound of formula (III)) as pale solid (48 mg, 91 % yield). 1 H-NMR (400 MHz, DMSO-d6) d (ppm) = 10.51 (s, 1 H), 7.80 (d, J= 4.0 Hz, 1 H), 7.31 (d, J= 4.0 Hz, 1 H), 6.81 (s, 1 H), 3.95 (s, 3H), 2.61 (s, 3H), 1 .59 (s, 9H). LCMS (m/z): negative mode 274.1 [M-1 ]-.

Example 8: Synthesis of fert-butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate

(Compound of formula (III), or a salt thereof) according to the following sequence:


f available (P3 = Boc) 
formula (ill)

Step 1 : Synthesis of 5-(benzyloxy)-1 ,3-dimethyl-2 -nitrobenzene

To a solution of commercially available 3,5-dimethyl-4-nitrophenol (100.0 g, 590.4 mmol) in DMF (500 ml_), CS2CO3 (230.8 g, 708.5 mmol) was added and the resulting mixture was stirred for 10 minutes. Then, (bromomethyl)benzene (104.1 g, 590.4 mmol) was added dropwise to the mixture within 30 minutes. The reaction mixture was stirred at 20-25 °C for 1 hour, and then poured into ice-water (1800 ml_). The solid separated out was collected by filtration and washed with water (500 ml_). The cake was dissolved in ethyl acetate (500 ml.) and the solution was washed with a saturated solution of NaCI (50 ml_), was separated, and the solution was concentrated to give 5-(benzyloxy)-l ,3-dimethyl-2-nitrobenzene 2 (147 g, 97.8% yield) as brown solid. HPLC purity

99.7%. 1H-NMR (400 MHz, DMSO-d6) d (ppm) = 7.42 (m, 5 H), 6.94 (s, 2H), 5.16 (s, 2 H), 2.25 (s, 6 H); LCMS (m/z): negative mode 256.2 [M-1 ]-

Step 2: Synthesis of fert-butyl 5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C12, wherein P3 = Boc)

To a solution of 5-(benzyloxy)-1 ,3-dimethyl-2-nitrobenzene (60.0 g, 233.2 mmol, from Step 1) in DMF (300 ml.) were added DMF-DMA (87.8 g, 699.6 mmol) and pyrrolidine (50.3 g, 699.6 mmol). The solution was heated to 85-90 °C and stirred for 19 hours under nitrogen, then the mixture was cooled to 20-25 °C. The volatile components (DMF-DMA, pyrrolidine and DMF) were removed at 65-70 °C on a rotary evaporator. The crude mixture was dissolved in ethyl acetate (300 ml_), and Raney Nickel (6.0 g) was added. The reaction mixture was subjected to catalytic hydrogenation under atmospheric pressure, overnight. Then, the reaction mixture was put under nitrogen. The mixture was filtrated and the filtrate was concentrated to provide 5-(benzyloxy)-7-methyM H-indole as a black oil. 5-(benzyloxy)-7-methyl-1 H-indole was used without further purification into the next step.

5-(benzyloxy)-7-methyl-1 H-indole was dissolved in acetonitrile (300 ml_), (Boc)20 (53.6 g, 233.2 mmol) and DMAP (5.7 g, 46.6 mmol) were added. The reaction mixture was stirred at 20-25 °C for 1 hour. Acetonitrile was removed on a rotary evaporator, and the residual mixture was dissolved in ethyl acetate (300 ml_). The solution was washed with a saturated aqueous solution of NaHC03 and then concentrated to give a crude oil which was purified by column chromatography (Si02, 500 g) using a mixture of heptane / MTBE (1 :10) to provide the intermediate tert-butyl 5-(benzyloxy)-7-methyl-1 H-indole-1 -carboxylate as a brown oil (42.1 g, 49.2% yield). HPLC purity 93.5%. 1 H-NMR (400 MHz, DMSO-d6) d (ppm) = 7.59 (d, J= 3.67 Hz, 1 H), 7.40 (m, 5 H), 7.04 (d, J= 2.45 Hz, 1 H), 6.81 (d, J= 2.2 Hz, 1 H), 6.57 (d, J= 3.67 Hz, 1 H), 5.1 1 (s, 2 H), 2.51 (s, 3 H), 1.58 (s, 9 H). LCMS (m/z): negative mode 336.2 [M-1 ]- To a solution of intermediate tert-butyl 5-(benzyloxy)-7-methyl-1 H-indole-1-carboxylate (36.7 g, 100 mmol) in ethanol (250 mL), under nitrogen, 10% Pd/C (10.6 g, 10 mmol) and ammonium formate (6.8 g, 105 mmol) were added. The solution was heated to 45-50 °C and stirred for 5 hours under nitrogen. Then the mixture was cooled to room temperature, filtered, and the filtrate was concentrated to give a residue oil. The residual oil was dissolved in ethyl acetate (250 mL), the solution was washed with a saturated aqueous solution of NaCI (100 mL), the phases were separated. The organic layers were collected and concentrated. The obtained crude mixtures was slurried with a (1 :15) mixture of MTBE / Heptane (160 mL) for 2 hours. The precipitate was filtered and washed with heptane (50 mL). The cake was dried under vacuum to give tert-butyl 5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C12) as a tawny solid (21 .8 g, 87.2% yield). HPLC purity 97.7%. 1 H-NMR (400 MHz, DMS0-d6) d (ppm) = 9.13 (s, 1 H), 7.52 (d, J= 3.67 Hz, 1 H), 6.74 (d, J= 2.2 Hz, 1 H), 6.56 (m, 1 H), 6.50 (d, J= 3.67 Hz, 1 H), 2.45 (s, 3 H), 1 .57 (s, 9 H). LCMS (m/z): negative mode 246.2 [M-1 ]-

Step 3: Synthesis of fert-butyl 4-formyl-5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C13, wherein P3 = Boc)

To a mixture of MgCI2 (1 1 .6 g, 1 19.7 mmol) and (CHO)n (5.0 g, 159.6 mmol), in THF (150 ml), under nitrogen, triethylamine (17.8 ml_, 127.7 mmol) was added dropwise and the resulting mixture was stirred at 20-25 °C for 10 minutes. Then, tert-butyl 5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C12) (10.0 g, 39.9 mmol) was added to the mixture. The reaction mixture was heated to 65-70 °C and stirred for 3 hours. The reaction mixture was cooled to 20-25 °C, followed by addition of 2N HCI (70 ml) and isopropyl acetate (150 ml). The mixture was separated and the organic layer was washed with a 5% NaCI solution. Then, the solution was concentrated to give a crude solid. The solid was slurried with ethanol (100 ml.) for 1 hour. The solid precipitate was filtrated, and washed with ethanol (20 ml_). The cake was dried under vacuum to give tert-butyl 4-formyl-5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C13) as a tawny solid (7.2 g, 63.9% yield). HPLC purity 96.5%. The filtrate solution was concentrated to 20 mL, then stirred for 1 hour. The solid was filtrated, and washed with ethanol (5 mL). The cake was dried by vacuum to give an additional amount of tert-butyl 4-formyl-5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C13) as a tawny solid (1 .1 g, 95.3% assay, 9.5% yield.). HPLC purity 90.5%. 1 H-NMR (400 MHz, DMSO-d6) d (ppm) = 10.69 (s, 1 H), 10.47 (s, 1 H), 7.75 (d, J= 3.35 Hz, 1 H), 7.27 (d, J= 3.55 Hz, 1 H), 6.74 (s, 1 H), 2.51 (s, 3 H), 1 .59 (s, 9 H); LCMS (m/z): negative mode 274.2 [M-1 ]-.

Step 4: Synthesis of fert-Butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate (Compound of formula (III)).

To a suspension of tert-butyl 4-formyl-5-hydroxy-7-methyl-1 H-indole-1 -carboxylate (C13) (6.0 g, 21 .3 mmol) in MeCN (60 mL), 50% K2C03 solution (20 mL) and dimethyl sulfate (2.26 mL, 23.4 mmol) were added. The resulting mixture was stirred at 35-40 °C for 3 hours. The reaction mixture was cooled to 20-25 °C and isopropyl acetate (30 mL) was added. The mixture was then extracted; the water layer was extracted with isopropyl acetate (15 mL), the organic layers were combined and concentrated to give a crude residual. The crude residual was dissolved in isopropyl acetate (60 mL), the solution was washed with a statured NH4CI solution, and then concentrated to give a crude product (6.6 g). The crude was slurried with ethyl acetate / Heptane (100 mL, 1/50) for 3 hours. The solid was filtrated, washed with heptane (20 mL). The cake was dried under vacuum to give tert-butyl 4-formyl-5-methoxy-7-methyl-1 H-indole-1 -carboxylate (Compound of formula (III))

as a pink solid (5.5 g, 87.8% yield). HPLC purity 99.3%. 1 H-NMR (400 MHz, DMSO-d6) d (ppm) = 10.52 (s, 1 H), 7.79 (d, J= 3.67 Hz, 1 H), 7.31 (d, J= 3.67 Hz, 1 H), 7.02 (s, 1 H) , 3.95 (s, 3 H), 2.61 (s, 3 H), 1 .60 (s, 9 H); LCMS (m/z): positive mode 290 [M]+.

Example 9: Synthesis of Compound of formula , or salt thereof (R = methyl).

Method 1 (Pa = Boc and R = methyl): To a vessel were added lr(CO)2acac (1 mg, 0.1 mol%), compound of formula (II) (maleic salt, 3 mmol, 1 .137g), compound of formula (III) (3 mmol, 0.867g) in 9 ml. of degassed ethanol. The autoclave was purged 3 times with nitrogen and 3 times with H2 under stirring (250 RPM). The reactions were run for 24 hours at 75 °C under 20 bar of H2 at 700 RPM. An aliquot of the reaction was diluted in methanol and was analyzed by HPLC. Compound of formula (C15) was obtained after 24 hours in 88% conversion.

Method 2 (Pa = Boc and R = methyl): To a vessel were added lrCI3, xH20 (0.05 mol%, 0.9 mg, anhydrous), compound of formula (II) (maleic salt, 6 mmol, 2.274 g ), compound of formula (III) (6 mmol, 1 .735g) in 12 ml. of degassed ethanol. The autoclave was purged 3 times with nitrogen and 3 times with carbon monoxide (CO) (250 RPM). The autoclave was pressurized with 1 bar of CO and 19 bar of H2 and run for 24 hours at 75 °C under 20 bar of H2 / CO at 700 RPM. An aliquot of the reaction was diluted in methanol and was analyzed by HPLC. Compound of formula (C15) was obtained after 24 hours in 62% conversion.

1H NMR (400 MHz, DMSO-d6) d ppm 8.13 (d, J=8.16 Hz, 2H), 7.77 (br. d, J=7.84 Hz, 2H), 7.62 -7.68 (m, 1 H), 6.85 (s, 1 H), 6.80 (d, J= 3.76 Hz, 1 H), 4.01 (s, 3H), 3.92 (s, 3H), 3.73 (br. s, 1 H), 3.55 – 3.67 (m, 4H), 3.39 – 3.42 (m, 1 H), 2.60 – 2.70 (m, 5H), 1 .99 – 2.02(br. d, 1 H), 1 .82 – 1.90 (m, 2H), 1.74 (s, 9H), 1 .64 – 1 70(m, 1 H), 1 .35 (t, J= 6.97 Hz, 3H).

1. Schubart A, et al. Proc Natl Acad Sci U S A. 2019 Mar 29. pii: 201820892.

 Proceedings of the National Academy of Sciences of the United States of America (2019), 116(16), 7926-7931.

//////LNP 023, BDBM160475, ZINC223246892HY-127105CS-0093107, LNP023

O=C(O)c1ccc(cc1)[C@@H]4C[C@H](CCN4Cc2c(OC)cc(C)c3nccc23)OCC

Eptinezumab エプチネズマブ;


Fig. 4.7

Eptinezumab

エプチネズマブ;

(Heavy chain)
EVQLVESGGG LVQPGGSLRL SCAVSGIDLS GYYMNWVRQA PGKGLEWVGV IGINGATYYA
SWAKGRFTIS RDNSKTTVYL QMNSLRAEDT AVYFCARGDI WGQGTLVTVS SASTKGPSVF
PLAPSSKSTS GGTAALGCLV KDYFPEPVTV SWNSGALTSG VHTFPAVLQS SGLYSLSSVV
TVPSSSLGTQ TYICNVNHKP SNTKVDARVE PKSCDKTHTC PPCPAPELLG GPSVFLFPPK
PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN AKTKPREEQY ASTYRVVSVL
TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP QVYTLPPSRE EMTKNQVSLT
CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS
VMHEALHNHY TQKSLSLSPG K
(Light chain)
QVLTQSPSSL SASVGDRVTI NCQASQSVYH NTYLAWYQQK PGKVPKQLIY DASTLASGVP
SRFSGSGSGT DFTLTISSLQ PEDVATYYCL GSYDCTNGDC FVFGGGTKVE IKRTVAAPSV
FIFPPSDEQL KSGTASVVCL LNNFYPREAK VQWKVDNALQ SGNSQESVTE QDSKDSTYSL
SSTLTLSKAD YEKHKVYACE VTHQGLSSPV TKSFNRGEC
(Disulfide bridge: H22-H95, H138-H194, H214-L219, H220-H’220, H223-H’223, H255-H315, H361-H419, H’22-H’95, H’138-H’194, H’214-L’219, H’255-H’315, H’361-H’419, L22-L89, L139-L199, L’22-L’89, L’139-L’199)

Formula
C6352H9838N1694O1992S46
cas
1644539-04-7
Mol weight
143281.2247

Antimigraine, Anti-calcitonin gene-related peptide (GCRP) antibody

Immunoglobulin G1, anti-(calcitonin gene-related peptide) (human-oryctolagus cuniculus monoclonal ALD403 heavy chain), disulfide with human-oryctolagus cuniculus monoclonal ALD403 kappa-chain, dimer

Approved 2020 fda

ALD403, UNII-8202AY8I7H

Humanized anti-calcitonin gene-related peptide (CGRP) IgG1 antibody for the treatment of migraine.

Eptinezumab, sold under the brand name Vyepti, is a medication for the preventive treatment of migraine in adults.[2] It is a monoclonal antibody that targets calcitonin gene-related peptides (CGRP) alpha and beta.[3][4] It is administered by intravenous infusion every three months.[2]

Image result for Eptinezumab

Eeptinezumab-jjmr was approved for use in the United States in February 2020.[5]

Image result for Eptinezumab

References

  1. ^ “Alder BioPharmaceuticals Initiates PROMISE 2 Pivotal Trial of Eptinezumab for the Prevention of Migraine”. Alder Biopharmaceuticals. 28 November 2016.
  2. Jump up to:a b “Vyeptitm (eptinezumab-jjmr) injection, for intravenous use” (PDF). U.S. Food and Drug Administration (FDA). Retrieved 24 February2020.
  3. ^ Dodick DW, Goadsby PJ, Silberstein SD, Lipton RB, Olesen J, Ashina M, et al. (November 2014). “Safety and efficacy of ALD403, an antibody to calcitonin gene-related peptide, for the prevention of frequent episodic migraine: a randomised, double-blind, placebo-controlled, exploratory phase 2 trial”. The Lancet. Neurology13 (11): 1100–1107. doi:10.1016/S1474-4422(14)70209-1PMID 25297013.
  4. ^ “International Nonproprietary Names for Pharmaceutical Substances (INN)” (PDF)WHO Drug Information. WHO. 31 (1). 2017.
  5. ^ “Vyepti: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 24 February 2020.

External links

Image result for Eptinezumab

Eptinezumab
Monoclonal antibody
Type Whole antibody
Source Humanized
Target CALCACALCB
Clinical data
Trade names Vyepti
Other names ALD403,[1] eeptinezumab-jjmr
License data
Routes of
administration
IV
Drug class Calcitonin gene-related peptide antagonist
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
ChemSpider
  • none
UNII
KEGG
Chemical and physical data
Formula C6352H9838N1694O1992S46
Molar mass 143283.20 g·mol−1

Biologics license application submitted for eptinezumab, an anti-CGRP antibody for migraine prevention

Alder BioPharmaceuticals has submitted a biologics license application (BLA) for eptinezumab, a humanized IgG1 monoclonal antibody that targets calcitonin gene-related peptide (CGRP), for migraine prevention. If the US Food and Drug Administration grants approval, Alder will be on track to launch the drug in Q1 2020. The BLA included data from the PROMISE 1 and PROMISE 2 studies, which evaluated the effects of eptinezumab in episodic migraine patients (n=888) or chronic migraine patients (n=1,072), respectively.  In PROMISE 1, the primary and key secondary endpoints were met, and the safety and tolerability were similar to placebo, while in PROMISE 2, the primary and all key secondary endpoints were met, and the safety and tolerability was consistent with earlier eptinezumab studies.

Alder announced one-year results from the PROMISE 1 study in June 2018, which indicated that, following the first quarterly infusion, episodic migraine patients treated with 300 mg eptinezumab experienced 4.3 fewer monthly migraine days (MMDs) from a baseline of 8 MMDs, compared to 3.2 fewer MMDs for placebo from baseline (p= 0.0001). At one year after the third and fourth quarterly infusions, patients treated with 300 mg eptinezumab experienced further gains in efficacy, with a reduction of 5.2 fewer MMDs compared to 4.0 fewer MMDs for placebo-treated patients.  In addition, ~31% of episodic migraine patients achieved, on average per month, 100% reduction of migraine days from baseline compared to ~ 21% for placebo. New 6-month results from the PROMISE 2 study were also released in June 2018.  These results indicated that, after the first quarterly infusion, chronic migraine patients dosed with 300 mg of eptinezumab experienced 8.2 fewer MMDs, from a baseline of 16 MMDs, compared to 5.6 fewer MMDs for placebo from baseline (p <.0001). A further reduction in MMDs was seen following a second infusion; 8.8 fewer MMDs for patients dosed with 300 mg compared to 6.2 fewer MMDs for those with placebo. In addition, ~ 21% of chronic migraine patients achieved, on average, 100% reduction of MMDs from baseline compared to 9% for placebo after two quarterly infusions of 300 mg of eptinezumab.

If approved, eptinezumab would become the fourth antibody therapeutic for migraine prevention on the US market, following the approval of erenumab-aooe (Aimovig; Novartis), galcanezumab-gnlm (Emgality; Eli Lilly & Company) and fremanezumab-vfrm (Ajovy; Teva Pharmaceuticals) in 2018.

//////////Eptinezumab, Monoclonal antibody, Peptide, エプチネズマブ  , fda 2020, approvals 2020

Amisulpride, アミスルプリド ,


71675-85-9.png

ChemSpider 2D Image | Amisulpride | C17H27N3O4S

Amisulpride.svg

Amisulpride

FDA 2020, Barhemsys APPROVED, 2020/2/27

Name
Amisulpride (INN);
Deniban (TN);
Solian (TN)
アミスルプリド;
Formula
C17H27N3O4S
CAS
71675-85-9
Mol weight
369.479

Antipsychotic, Dopamine receptor antagonist, Neuropsychiatric agent

amisulpride(标准品)

275-831-7 [EINECS]
Synthesis ReferenceUS4401822
4-Amino-N-[(1-ethyl-2-pyrrolidinyl)methyl]-5-(ethylsulfonyl)-2-methoxybenzamide
Amisulpride
CAS Registry Number: 71675-85-9
CAS Name: 4-Amino-N-[(1-ethyl-2-pyrrolidinyl)methyl]-5-(ethylsulfonyl)-2-methoxybenzamide
Additional Names: 4-amino-N-[(1-ethyl-2-pyrrolidinyl)methyl]-5-(ethylsulfonyl)-o-anisamide; aminosultopride
Manufacturers’ Codes: DAN-2163
Trademarks: Deniban (Synthelabo); Socian (Synthelabo); Solian (Synthelabo); Sulamid (Baldacci)
Molecular Formula: C17H27N3O4S
Molecular Weight: 369.48
Percent Composition: C 55.26%, H 7.37%, N 11.37%, O 17.32%, S 8.68%
Literature References: Dopamine receptor antagonist. Prepn: M. Thominet et al., BE 872585eidem, US 4401822 (1979, 1983 both to Soc. d’Etudes Sci. Ind. de l’Ile-de-France).
Crystal structure: H. L. DeWinter et al., Acta Crystallogr. C46, 313 (1990). Psychopharmacology: G. Perrault et al., J. Pharmacol. Exp. Ther. 280, 73 (1997). HPLC determn in plasma and urine: B. Malavasi et al., J. Chromatogr. B 676, 107 (1996). Series of articles on pharmacology and clinical efficacy in schizophrenia: Int. Clin. Psychopharmacol. 12, Suppl. 2, S11-S36 (1997).
Properties: Crystals from acetone, mp 126-127°. LD50 in male mice (mg/kg): 56-60 i.v.; 175-180 i.p.; 224-250 s.c.; 1024-1054 orally (Thominet).
Melting point: mp 126-127°
Toxicity data: LD50 in male mice (mg/kg): 56-60 i.v.; 175-180 i.p.; 224-250 s.c.; 1024-1054 orally (Thominet)
Therap-Cat: Antipsychotic.
Keywords: Antipsychotic; Benzamides; Dopamine Receptor Antagonist.
Amisulpride (trade name Solian) is an antipsychotic drug sold by Sanofi-Aventis.  but is approved for use in Europe and Australia for the treatment of psychoses and schizophrenia. Additionally, it is approved in Italy for the treatment of dysthymia (under the brand name Deniban). Amisulpride is a selective dopamine antagonist.

Amisulpride is an antiemetic and antipsychotic medication used at lower doses intravenously to prevent and treat postoperative nausea and vomiting; and at higher doses orally and intramuscularly to treat schizophrenia and acute psychotic episodes. It is sold under the brandnames Barhemsys[6] (as an antiemetic) and Solian, Socian, Deniban and others (as an antipsychotic).[2] It is also used to treat dysthymia.[7]

It is usually classed with the atypical antipsychotics. Chemically it is a benzamide and like other benzamide antipsychotics, such as sulpiride, it is associated with a high risk of elevating blood levels of the lactation hormone, prolactin (thereby potentially causing the absence of the menstrual cycle, breast enlargement, even in males, breast milk secretion not related to breastfeeding, impaired fertility, impotence, breast pain, etc.), and a low risk, relative to the typical antipsychotics, of causing movement disorders.[8][9][10] It has also been found to be modestly more effective in treating schizophrenia than the typical antipsychotics.[9]

Amisulpride is approved for use in the United States in adults for the prevention of postoperative nausea and vomiting (PONV), either alone or in combination with an antiemetic of a different class; and to treat PONV in those who have received antiemetic prophylaxis with an agent of a different class or have not received prophylaxis.[6]

Amisulpride is believed to work by blocking, or antagonizing, the dopamine D2 receptor, reducing its signalling. The effectiveness of amisulpride in treating dysthymia and the negative symptoms of schizophrenia is believed to stem from its blockade of the presynapticdopamine D2 receptors. These presynaptic receptors regulate the release of dopamine into the synapse, so by blocking them amisulpride increases dopamine concentrations in the synapse. This increased dopamine concentration is theorized to act on dopamine D1 receptors to relieve depressive symptoms (in dysthymia) and the negative symptoms of schizophrenia.[7]

It was introduced by Sanofi-Aventis in the 1990s. Its patent expired by 2008, and generic formulations became available.[11] It is marketed in all English-speaking countries except for Canada and the United States.[10] A New York City based company, LB Pharmaceuticals, has announced the ongoing development of LB-102, also known as N-methyl amisulpride, an antipsychotic specifically targeting the United States.[12][13] A poster presentation at European Neuropsychopharmacology[14] seems to suggest that this version of amisulpride, known as LB-102 displays the same binding to D2, D3 and 5HT7 that amisulpride does.[15][16]

Medical uses

Schizophrenia

In a 2013 study in a comparison of 15 antipsychotic drugs in effectiveness in treating schizophrenic symptoms, amisulpride was ranked second and demonstrated high effectiveness. 11% more effective than olanzapine (3rd), 32-35% more effective than haloperidolquetiapine, and aripiprazole, and 25% less effective than clozapine (1st).[9] Although according to other studies it appears to have comparable efficacy to olanzapine in the treatment of schizophrenia.[17][18] Amisulpride augmentation, similarly to sulpirideaugmentation, has been considered a viable treatment option (although this is based on low-quality evidence) in clozapine-resistant cases of schizophrenia.[19][20] Another recent study concluded that amisulpride is an appropriate first-line treatment for the management of acute psychosis.[21]

Contraindications

Amisulpride’s use is contraindicated in the following disease states[2][22][8]

Neither is it recommended to use amisulpride in patients with hypersensitivities to amisulpride or the excipients found in its dosage form.[2]

Adverse effects

Very Common (≥10% incidence)[1]
  • Extrapyramidal side effects (EPS; including dystonia, tremor, akathisiaparkinsonism). Produces a moderate degree of EPS; more than aripiprazole (not significantly, however), clozapine, iloperidone (not significantly), olanzapine (not significantly), quetiapine (not significantly) and sertindole; less than chlorpromazine (not significantly), haloperidol, lurasidone (not significantly), paliperidone (not significantly), risperidone (not significantly), ziprasidone (not significantly) and zotepine (not significantly).[9]
Common (≥1%, <10% incidence)[1][2][23][22][8]
  • Hyperprolactinaemia (which can lead to galactorrhoea, breast enlargement and tenderness, sexual dysfunction, etc.)
  • Weight gain (produces less weight gain than chlorpromazine, clozapine, iloperidone, olanzapine, paliperidone, quetiapine, risperidone, sertindole, zotepine and more (although not statistically significantly) weight gain than haloperidol, lurasidone, ziprasidone and approximately as much weight gain as aripiprazole and asenapine)[9]
  • Anticholinergic side effects (although it does not bind to the muscarinic acetylcholine receptors and hence these side effects are usually quite mild) such as
– constipation
– dry mouth
– disorder of accommodation
– Blurred vision
Rare (<1% incidence)[1][2][23][22][8]

Hyperprolactinaemia results from antagonism of the D2 receptors located on the lactotrophic cells found in the anterior pituitary gland. Amisulpride has a high propensity for elevating plasma prolactin levels as a result of its poor blood-brain barrier penetrability and hence the resulting greater ratio of peripheral D2 occupancy to central D2 occupancy. This means that to achieve the sufficient occupancy (~60–80%[24]) of the central D2 receptors in order to elicit its therapeutic effects a dose must be given that is enough to saturate peripheral D2receptors including those in the anterior pituitary.[25][26]

  • Somnolence. It produces minimal sedation due to its absence of cholinergic, histaminergic and alpha adrenergic receptor antagonism. It is one of the least sedating antipsychotics.[9]

Discontinuation

The British National Formulary recommends a gradual withdrawal when discontinuing antipsychotics to avoid acute withdrawal syndrome or rapid relapse.[27] Symptoms of withdrawal commonly include nausea, vomiting, and loss of appetite.[28] Other symptoms may include restlessness, increased sweating, and trouble sleeping.[28] Less commonly there may be a felling of the world spinning, numbness, or muscle pains.[28] Symptoms generally resolve after a short period of time.[28]

There is tentative evidence that discontinuation of antipsychotics can result in psychosis.[29] It may also result in reoccurrence of the condition that is being treated.[30] Rarely tardive dyskinesia can occur when the medication is stopped.[28]

Overdose

Torsades de pointes is common in overdose.[31][32] Amisulpride is moderately dangerous in overdose (with the TCAs being very dangerous and the SSRIs being modestly dangerous).[33][34]

Interactions

Amisulpride should not be used in conjunction with drugs that prolong the QT interval (such as citalopramvenlafaxinebupropionclozapinetricyclic antidepressantssertindoleziprasidone, etc.),[33] reduce heart rate and those that can induce hypokalaemia. Likewise it is imprudent to combine antipsychotics due to the additive risk for tardive dyskinesia and neuroleptic malignant syndrome.[33]

Pharmacology

Pharmacodynamics

Amisulpride and its relatives sulpiridelevosulpiride, and sultopride have been shown to bind to the high-affinity GHB receptor at concentrations that are therapeutically relevant (IC50 = 50 nM for amisulpride).[37]Amisulpride functions primarily as a dopamine D2 and D3 receptor antagonist. It has high affinity for these receptors with dissociation constantsof 3.0 and 3.5 nM, respectively.[36] Although standard doses used to treat psychosis inhibit dopaminergic neurotransmission, low doses preferentially block inhibitory presynaptic autoreceptors. This results in a facilitation of dopamine activity, and for this reason, low-dose amisulpride has also been used to treat dysthymia.[2]

Amisulpride, sultopride and sulpiride respectively present decreasing in vitro affinities for the D2 receptor (IC50 = 27, 120 and 181 nM) and the D3 receptor (IC50 = 3.6, 4.8 and 17.5 nM).[39]

Though it was long widely assumed that dopaminergic modulation is solely responsible for the respective antidepressant and antipsychoticproperties of amisulpride, it was subsequently found that the drug also acts as a potent antagonist of the serotonin 5-HT7 receptor (Ki = 11.5 nM).[36] Several of the other atypical antipsychotics such as risperidone and ziprasidone are potent antagonists at the 5-HT7 receptor as well, and selective antagonists of the receptor show antidepressant properties themselves. To characterize the role of the 5-HT7 receptor in the antidepressant effects of amisulpride, a study prepared 5-HT7 receptor knockout mice.[36] The study found that in two widely used rodent models of depression, the tail suspension test, and the forced swim test, those mice did not exhibit an antidepressant response upon treatment with amisulpride.[36] These results suggest that 5-HT7 receptor antagonism mediates the antidepressant effects of amisulpride.[36]

Amisulpride also appears to bind with high affinity to the serotonin 5-HT2B receptor (Ki = 13 nM), where it acts as an antagonist.[36] The clinical implications of this, if any, are unclear.[36] In any case, there is no evidence that this action mediates any of the therapeutic effects of amisulpride.[36]

Society and culture

Brand names

Brand names include: Amazeo, Amipride (AU), Amival, Solian (AUIERUUKZA), Soltus, Sulpitac (IN), Sulprix (AU), Midora (RO) and Socian (BR).[40][41]

Availability

Amisulpride was not approved by the Food and Drug Administration for use in the United States until February 2020, but it is used in Europe,[41]Israel, Mexico, India, New Zealand and Australia[2] to treat psychosis and schizophrenia.[42][43]

Amisulpride was approved for use in the United States in February 2020.[44][6]

CLIP

Dopamine receptor antagonist. Prepn: M. Thominet et al., BE 872585; eidem, U.S. Patent 4,401,822 (1979, 1983 both to Soc. d’Etudes Sci. Ind. de l’Ile-de-France).

CLIP

4-Amino-N-((1-ethyl-2-pyrrolidinyl)methyl)-5-(ethylsulfonyl)-o-anisamide, could be produced through many synthetic methods.

Following is one of the synthesis routes:
Firstly, the acetylation of 5-aminosalicylic acid (I) with acetic anhydride in hot acetic acid affords 5-acetaminosalicylic acid (II), which is methylated with dimethyl sulfate and K2CO3 in refluxing acetone producing methyl 2-methoxy-5-acetaminobenzoate (III). Secondly, nitration of (III) with HNOin acetic acid affords methyl 2-methoxy-4-nitro-5-acetaminobenzoate (IV), which is deacetylated with H2SO4 in refluxing methanol to give methyl 2-methoxy-4-nitro-5-aminobenzoate (V). Next, the diazotation of (V) with NaNO2-HCl, followed by reaction with sodium ethylmercaptide, oxidation with H2O2 and hydrolysis with NaOH in ethanol yields 2-methoxy-4-nitro-5-(ethylsulfonyl)benzoic acid (VI), which is condensed with N-ethyl-2-aminomethylpyrrolidine (VII) in the presence of ethyl chloroformate and triethylamine in dioxane affording 2-methoxy-4-nitro-N-[(1-ethyl-2-pyrrolidinyl) methyl]-5-(ethylsulfonyl)benzamide (VIII). At last, this compound is reduced with H2 over Raney-Ni in ethanol.

Production Route of Amisulpride

CLIP

BE 0872585; ES 476755; FR 2415099; GB 2083458; JP 54145658; US 4294828; US 4401822

Alkylation of 2-methoxy-4-amino-5-mercaptobenzoic acid (X) with diethyl sulfate acid Na2CO3 gives 2-methoxy-4-amino-5-ethylthiobenzoic acid (XI), which is oxidized with H2O2 in acetic acid yielding 2-methoxy-4-amino-5-(ethylsulfonyl)benzoic acid (XII). Finally, this compound is condensed with (VII) by means of ethyl chloroformate.

CLIP

FR 2460930

Acetylation of 5-aminosalicylic acid (I) with acetic anhydride in hot acetic acid gives 5-acetaminosalicylic acid (II), which is methylated with dimethyl sulfate and K2CO3 in refluxing acetone yielding methyl 2-methoxy-5-acetaminobenzoate (III). Nitration of (III) with HNO3 in acetic acid affords methyl 2-methoxy-4-nitro-5-acetaminobenzoate (IV), which is deacetylated with H2SO4 in refluxing methanol to give methyl 2-methoxy-4-nitro-5-aminobenzoate (V). The diazotation of (V) with NaNO2-HCl, followed by reaction with sodium ethylmercaptide, oxidation with H2O2 and hydrolysis with NaOH in ethanol yields 2-methoxy-4-nitro-5-(ethylsulfonyl)benzoic acid (VI), which is condensed with N-ethyl-2-aminomethylpyrrolidine (VII) by means of ethyl chloroformate and triethylamine in dioxane affording 2-methoxy-4-nitro-N-[(1-ethyl-2-pyrrolidinyl) methyl]-5-(ethylsulfonyl)benzamide (VIII). Finally, this compound is reduced with H2 over Raney-Ni in ethanol.

CLIP

Treatment of thiourea (I) with iodomethane provided S-methylthiouronium iodide (II). This was further condensed with N-methylpiperazine (III) to afford the intermediate piperazine-1-carboxamidine (IV)

CLIP

Regioselective lithiation of 1,2,4-trichlorobenzene (V) with n-BuLi at -60 C, followed by quenching of the resultant organolithium compound (VI) with N,N-dimethylformamide yielded 2,3,5-trichlorobenzaldehyde (VII) (1), which was then reduced with NaBH4 to provide alcohol (VIII). Bromination of (VIII) using PBr3 afforded compound (IX), whose bromide atom was displaced with KCN to give the trichlorophenylacetonitrile (X). Claisen condensation of (X) with ethyl formate in the presence of NaOEt furnished the oxo nitrile sodium enolate (XI), which was subsequently O-alkylated with iodomethane yielding the methoxy acrylonitrile (XII). Finally, cyclization of (XII) with the piperazine-1-carboxamidine (IV) in EtOH gave rise to the target pyrimidine derivative

PATENT

https://patents.google.com/patent/US20130096319A1/en

Amisulpride is represented by the formula (I) as given below.

Figure US20130096319A1-20130418-C00001

The product patent U.S. Pat. No. 4,401,822 describes preparation of amisulpride as shown in scheme (I)

Figure US20130096319A1-20130418-C00002

The synthesis of amisulpride involves oxidation of 2-methoxy-4-amino-5-ethyl-thio benzoic acid (III) using acetic acid and hydrogen peroxide at 40-45° C. for few hours to obtain 2-methoxy-4-amino-5-ethyl-sulfonyl benzoic acid (IV). In our attempt to repeat this reaction, we found that almost 22 hours were required for completion and the purity of compound (IV) was 87.6%.

    • [0006]
      Thus, the product patent method suffers from the disadvantages such as high reaction time, low yield and low purity.
    • [0007]
      Liu Lie et al, Jingxi Huagong Zhongjianti 2008, 38 (3), 29-32 describes the process for the preparation of 2-methoxy-4-amino-5-ethyl-sulfonyl benzoic acid (IV) as shown in scheme (II).
    • Figure US20130096319A1-20130418-C00003
    • [0008]
      4-amino salicylic acid (VI) is treated with dimethyl sulphate in the presence of potassium hydroxide and acetone to give 4-amino-2-methoxy-methyl benzoate in 4 hours, which is further treated with potassium thiocynate to give compound of formula (VIII). 4-Amino-2-,methoxy-5-thiocyanatobenzoate (VIII) is treated with bromoethane to give 4-amino-5-ethylthio-2-methoxy benzoic acid (IX) which is further converted to 2-methoxy-4-amino-5-ethyl-sulfonyl benzoic acid (IV) via oxidation with hydrogen peroxide and acetic acid.
    • [0009]
      The yield of conversion of compound (VIII) to compound (IX) is 57% and the overall yield of compound (IV) from compound (VI) is 24% only. Thus, the above process suffers from the disadvantages such as low yield and in that it uses bromoethane which is skin and eye irritant and has carcinogenic effects.
    • [0010]
      Therefore, there is, an unfulfilled need to provide industrially feasible process for the preparation of 2-methoxy-4-amino-5-ethyl-sulfonyl benzoic acid (IV) and amisulpride (I) with higher purity and yield, since it is one of the key intermediates in the manufacture of amisulpride.

SUMMARY OF THE INVENTION

The present invention is related to a novel process for the preparation of amisulpride (I) that involves: (i) methylation of 4-amino-salicylic-acid (VI) with dimethyl sulphate and base, optionally in presence of TBAB to obtain 4-amino-2-methoxy methyl benzoate (VII) and (ii) oxidation of 4-amino-2-methoxy-5-ethyl thio benzoic acid (IX) or 4-amino-2-methoxy-5-ethyl thio methyl benzoate (X) with oxidizing agent in the presence of sodium tungstate or ammonium molybdate to give 2-methoxy-4-amino-5-ethyl-sulfonyl benzoic acid (IV) or 2-methoxy-4-amino-5-ethyl-sulfonyl methyl benzoate (XI) respectively.
    • Example 13

    • [0097]
      Preparation of crude amisulpride
    • [0098]
      To a stirring mixture of 4-amino-2-methoxy-5-ethyl sulphonyl benzoic acid (IV) and acetone (5.0 L) at 0-5° C., triethyl amine (0.405 Kg) was added and stirred followed by addition of ethyl chloroformate (0.368 Kg). N-ethyl-2-amino methyl pyrrolidine (0.627 Kg) was added to the reaction mass at 5-10° C. Temperature of reaction mass was raised to 25-30° C. and stirred for 120 min. To the same reaction mass triethyl amine (0.405 Kg) and ethyl chloroformate (0.368 Kg) was added with maintaining the temperature. Reaction mass was stirred for 120 min. After completion of reaction, water (4.0 L) was added. Reaction mass was filtered and washed with water (2.0 L). Filtrate was collected and water was added (9.0 L). pH of the reaction mass was adjusted to 10.8-11.2 by using 20% NaOH solution. Reaction mass was stirred for 240-300 min, filtered and washed with water. Solid was dried under vacuum
    • [0099]
      Yield : 70%
    • [0100]
      Purity: 98%

Example 14

  • [0101]
    Purification of amisulpride
  • [0102]
    Amisulpride (1 kg) was charged in acetone (6 liters) and the reaction mixture was heated till a clear solution was obtained. Slurry of activated carbon (0.1 kg in 1 liter) was added in acetone. The reaction mass was stirred at 50-55 ° C. for 60 minutes and filtered hot. The filtrate was concentrated and further heated to dissolve the solid. The reaction mass was cooled to 0-5° C., stirred and filtered. The precipitated solid was washed with acetone and dried.
  • [0103]
    Yield: 750 gm (75%)
  • [0104]
    HPLC purity: 99.8% (quantitative)
  • [0105]
    M.P.: 125° C.
  • [0106]
    DSC: shows endotherm at 133° C.
  • [0107]
    Particle size: d10=0.637, d50=6.0, d90=13.325 microns

CLIP

https://watermark.silverchair.com/bmw186.pdf?token=AQECAHi208BE49Ooan9kkhW_Ercy7Dm3ZL_9Cf3qfKAc485ysgAAAmEwggJdBgkqhkiG9w0BBwagggJOMIICSgIBADCCAkMGCSqGSIb3DQEHATAeBglghkgBZQMEAS4wEQQM_rfBl_qrJE7Y7K67AgEQgIICFOQ9ug62uUxOD4oCuuUGlGD3N04qUgCHew1O5UIyknvohf-_QUaJclqSZM6k5UhPTLgjkYyVMVgS04HMcDKUVXr1cMUfV6cExwayFb8z3MQUF4Ny6s8hPuAMJO4XsTm4qh0nnEykHwgMonNWdDr32D4B7NuEVwGE_5Z-d1yQvAdkNeCmEbHIaue3OTiocWodCsAv8yUdnXf1AtreXJkvsiAQtk4oCddsM_a2njiXJAc-VcFgTImCvsaCY-_eWT91Dc3gb7fpEAJSPLl06xx30GziAvF_hl5P33TaMFmVm_p-0rJGWi-_x92Tlo1CkuR1N1oWlcnuBSPqKeX3tbMO3phnIYtbDPycftd6UKI2f9-zyMRHgSId4xJCpaxvy6fndrWZ1qrHTyQLt_XqncL7zD8aYHER67kV3g30ZgAtcivHoMSHj9h4wGD5WLZ5-M4cZ0dpUyKx3E2njYBEBe0LNQyqDmP8HKpM_RBN2C2nuD2h1fJkiwf2kLAdlBC6gOhjl60XqU_7ARJZf_86kR3OhUJ5f8Ey2R-k3zwDHEc3tU10AlEky9ne-UWVHGjOCd9L-SV-eXfjOnaERGw9EHahxajGBCRuqa07-BtbV0mr53AKyaS5YUTQ2EZ7P3WarhImsJpYiQxWAuSlYn2F11RTMu_KjP7-DMXbX6pcq20axI2NNwrBtfsDXFbQWZ8q9R0FYGsUS90

References

  1. Jump up to:a b c d “Amisulpride 100 mg Tablets – Summary of Product Characteristics (SmPC)”(emc). 5 July 2019. Retrieved 26 February 2020.
  2. Jump up to:a b c d e f g h i j k “Solian tablets and solution product information” (PDF)TGA eBusiness Services. Sanofi-Aventis Australia Pty Ltd. 27 September 2019. Retrieved 26 February2020.
  3. Jump up to:a b c Rosenzweig, P.; Canal, M.; Patat, A.; Bergougnan, L.; Zieleniuk, I.; Bianchetti, G. (2002). “A review of the pharmacokinetics, tolerability and pharmacodynamics of amisulpride in healthy volunteers”. Human Psychopharmacology17 (1): 1–13. doi:10.1002/hup.320PMID 12404702.
  4. ^ Caccia, S (May 2000). “Biotransformation of Post-Clozapine Antipsychotics Pharmacological Implications”. Clinical Pharmacokinetics38 (5): 393–414. doi:10.2165/00003088-200038050-00002PMID 10843459.
  5. ^ Noble, S; Benfield, P (December 1999). “Amisulpride: A Review of its Clinical Potential in Dysthymia”. CNS Drugs12 (6): 471–483. doi:10.2165/00023210-199912060-00005.
  6. Jump up to:a b c “Barhemsys (amisulpride) injection, for intravenous use” (PDF). U.S. Food and Drug Administration (FDA). February 2020. Retrieved 26 February 2020.
  7. Jump up to:a b Pani L, Gessa GL (2002). “The substituted benzamides and their clinical potential on dysthymia and on the negative symptoms of schizophrenia”. Molecular Psychiatry7 (3): 247–53. doi:10.1038/sj.mp.4001040PMID 11920152.
  8. Jump up to:a b c d Rossi, S, ed. (2013). Australian Medicines Handbook (2013 ed.). Adelaide: The Australian Medicines Handbook Unit Trust. ISBN 978-0-9805790-9-3.
  9. Jump up to:a b c d e f g Leucht, S; Cipriani, A; Spineli, L; Mavridis, D; Orey, D; Richter, F; Samara, M; Barbui, C; Engel, RR; Geddes, JR; Kissling, W; Stapf, MP; Lässig, B; Salanti, G; Davis, JM (September 2013). “Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis”. Lancet382 (9896): 951–962. doi:10.1016/S0140-6736(13)60733-3PMID 23810019.
  10. Jump up to:a b Brayfield, A, ed. (June 2017). “Amisulpride: Martindale: The Complete Drug Reference”MedicineComplete. Pharmaceutical Press. Retrieved 5 August 2017.
  11. ^ De Silva, V; Hanwella, R (April 2008). “Pharmaceutical patents and the quality of mental healthcare in low- and middle-income countries”. The Psychiatrist32 (4): 121–23. doi:10.1192/pb.bp.107.015651.
  12. ^ “Pipeline”LB Pharmaceuticals. Retrieved 29 August 2019.
  13. ^ “About Us”LB Pharmaceuticals. Retrieved 26 February 2020.
  14. ^ “Data presented at 2017 ECNP meeting (European Neuropsychopharmacology, 2017, 27 (S4), S922-S923)”LB Pharmaceuticals. Retrieved 26 February 2020.
  15. ^ “Building a translational bridge from animals to man for clinical candidate LB-102, a next-generation benzamide antipsychotic (P.101)” (PDF)LB Pharmaceuticals. Retrieved 29 August 2019.
  16. ^ Grattan V, Vaino AR, Prensky Z, Hixon MS (August 2019). “Antipsychotic Benzamides Amisulpride and LB-102 Display Polypharmacy as Racemates, S Enantiomers Engage Receptors D2 and D3, while R Enantiomers Engage 5-HT7”ACS Omega4 (9): 14151–4. doi:10.1021/acsomega.9b02144ISSN 2470-1343PMC 6714530PMID 31497735.
  17. ^ Komossa, K; Rummel-Kluge, C; Hunger, H; Schmid, F; Schwarz, S; Silveira da Mota Neto, JI; Kissling, W; Leucht, S (January 2010). “Amisulpride versus other atypical antipsychotics for schizophrenia”The Cochrane Database of Systematic Reviews (1): CD006624. doi:10.1002/14651858.CD006624.pub2PMC 4164462PMID 20091599.
  18. ^ Leucht, S; Corves, C; Arbter, D; Engel, RR; Li, C; Davis, JM (January 2009). “Second-generation versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis”. Lancet373 (9657): 31–41. doi:10.1016/S0140-6736(08)61764-XPMID 19058842.
  19. ^ Solanki, RK; Sing, P; Munshi, D (October–December 2009). “Current perspectives in the treatment of resistant schizophrenia”Indian Journal of Psychiatry51 (4): 254–60. doi:10.4103/0019-5545.58289PMC 2802371PMID 20048449.
  20. ^ Mouaffak, F; Tranulis, C; Gourevitch, R; Poirier, MF; Douki, S; Olié, JP; Lôo, H; Gourion, D (2006). “Augmentation Strategies of Clozapine With Antipsychotics in the Treatment of Ultraresistant Schizophrenia”. Clinical Neuropharmacology29 (1): 28–33. doi:10.1097/00002826-200601000-00009PMID 16518132.
  21. ^ Nuss, P.; Hummer, M.; Tessier, C. (2007). “The use of amisulpride in the treatment of acute psychosis”Therapeutics and Clinical Risk Management3 (1): 3–11. doi:10.2147/tcrm.2007.3.1.3PMC 1936283PMID 18360610.
  22. Jump up to:a b c Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. ISBN 978-0-85711-084-8.
  23. Jump up to:a b Truven Health Analytics, Inc. DRUGDEX System (Internet) [cited 2013 Sep 19]. Greenwood Village, CO: Thomsen Healthcare; 2013.
  24. ^ Brunton, L; Chabner, B; Knollman, B (2010). Goodman and Gilman’s The Pharmacological Basis of Therapeutics (12th ed.). New York: McGraw-Hill Professional. ISBN 978-0-07-162442-8.
  25. ^ McKeage, K; Plosker, GL (2004). “Amisulpride: a review of its use in the management of schizophrenia”. CNS Drugs18 (13): 933–956. doi:10.2165/00023210-200418130-00007ISSN 1172-7047PMID 15521794.
  26. ^ Natesan, S; Reckless, GE; Barlow, KB; Nobrega, JN; Kapur, S (October 2008). “Amisulpride the ‘atypical’ atypical antipsychotic — Comparison to haloperidol, risperidone and clozapine”. Schizophrenia Research105 (1–3): 224–235. doi:10.1016/j.schres.2008.07.005PMID 18710798.
  27. ^ Joint Formulary Committee, BMJ, ed. (March 2009). “4.2.1”. British National Formulary (57 ed.). United Kingdom: Royal Pharmaceutical Society of Great Britain. p. 192. ISBN 978-0-85369-845-6Withdrawal of antipsychotic drugs after long-term therapy should always be gradual and closely monitored to avoid the risk of acute withdrawal syndromes or rapid relapse.
  28. Jump up to:a b c d e Haddad, Peter; Haddad, Peter M.; Dursun, Serdar; Deakin, Bill (2004). Adverse Syndromes and Psychiatric Drugs: A Clinical Guide. OUP Oxford. p. 207–216. ISBN 9780198527480.
  29. ^ Moncrieff J (July 2006). “Does antipsychotic withdrawal provoke psychosis? Review of the literature on rapid onset psychosis (supersensitivity psychosis) and withdrawal-related relapse”. Acta Psychiatrica Scandinavica114 (1): 3–13. doi:10.1111/j.1600-0447.2006.00787.xPMID 16774655.
  30. ^ Sacchetti, Emilio; Vita, Antonio; Siracusano, Alberto; Fleischhacker, Wolfgang (2013). Adherence to Antipsychotics in Schizophrenia. Springer Science & Business Media. p. 85. ISBN 9788847026797.
  31. ^ Isbister, GK; Balit, CR; Macleod, D; Duffull, SB (August 2010). “Amisulpride overdose is frequently associated with QT prolongation and torsades de pointes”. Journal of Clinical Psychopharmacology30 (4): 391–395. doi:10.1097/JCP.0b013e3181e5c14cPMID 20531221.
  32. ^ Joy, JP; Coulter, CV; Duffull, SB; Isbister, GK (August 2011). “Prediction of Torsade de Pointes From the QT Interval: Analysis of a Case Series of Amisulpride Overdoses”. Clinical Pharmacology & Therapeutics90 (2): 243–245. doi:10.1038/clpt.2011.107PMID 21716272.
  33. Jump up to:a b c Taylor, D; Paton, C; Shitij, K (2012). Maudsley Prescribing Guidelines in Psychiatry(11th ed.). West Sussex: Wiley-Blackwell. ISBN 978-0-47-097948-8.
  34. ^ Levine, M; Ruha, AM (July 2012). “Overdose of atypical antipsychotics: clinical presentation, mechanisms of toxicity and management”. CNS Drugs26 (7): 601–611. doi:10.2165/11631640-000000000-00000PMID 22668123.
  35. ^ Roth, BL; Driscol, J. “PDSP Ki Database”Psychoactive Drug Screening Program (PDSP). University of North Carolina at Chapel Hill and the United States National Institute of Mental Health. Retrieved 14 August 2017.
  36. Jump up to:a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah ai aj ak al am an ao ap aq ar asat au av aw Abbas AI, Hedlund PB, Huang XP, Tran TB, Meltzer HY, Roth BL (2009). “Amisulpride is a potent 5-HT7 antagonist: relevance for antidepressant actions in vivo”Psychopharmacology205 (1): 119–28. doi:10.1007/s00213-009-1521-8PMC 2821721PMID 19337725.
  37. Jump up to:a b Maitre, M.; Ratomponirina, C.; Gobaille, S.; Hodé, Y.; Hechler, V. (April 1994). “Displacement of [3H] gamma-hydroxybutyrate binding by benzamide neuroleptics and prochlorperazine but not by other antipsychotics”. European Journal of Pharmacology256(2): 211–214. doi:10.1016/0014-2999(94)90248-8PMID 7914168.
  38. ^ Schoemaker H, Claustre Y, Fage D, Rouquier L, Chergui K, Curet O, Oblin A, Gonon F, Carter C, Benavides J, Scatton B (1997). “Neurochemical characteristics of amisulpride, an atypical dopamine D2/D3 receptor antagonist with both presynaptic and limbic selectivity”. J. Pharmacol. Exp. Ther280 (1): 83–97. PMID 8996185.
  39. ^ Blomme, Audrey; Conraux, Laurence; Poirier, Philippe; Olivier, Anne; Koenig, Jean-Jacques; Sevrin, Mireille; Durant, François; George, Pascal (2000), “Amisulpride, Sultopride and Sulpiride: Comparison of Conformational and Physico-Chemical Properties”, Molecular Modeling and Prediction of Bioactivity, Springer US, pp. 404–405, doi:10.1007/978-1-4615-4141-7_97ISBN 9781461368571
  40. ^ “Amisulpride international”Drugs.com. 3 February 2020. Retrieved 26 February 2020.
  41. Jump up to:a b “Active substance: amisulpride” (PDF). 28 September 2017. EMA/658194/2017; Procedure no.: PSUSA/00000167/201701. Retrieved 26 February 2020.
  42. ^ Lecrubier, Y.; et al. (2001). “Consensus on the Practical Use of Amisulpride, an Atypical Antipsychotic, in the Treatment of Schizophrenia”. Neuropsychobiology44 (1): 41–46. doi:10.1159/000054913PMID 11408792.
  43. ^ Kaplan, A. (2004). “Psychotropic Medications Around the World”Psychiatric Times21(5).
  44. ^ “Barhemsys: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 26 February 2020.

External links

Amisulpride
Amisulpride.svg
Amisulpride-xtal-1990-ball-and-stick-model.png
Clinical data
Trade names Solian, Barhemsys, others
Other names APD421
AHFS/Drugs.com International Drug Names
License data
Pregnancy
category
  • AU: C
Routes of
administration
By mouthintravenous
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability 48%[3][2]
Protein binding 16%[2]
Metabolism Hepatic (minimal; most excreted unchanged)[2]
Elimination half-life 12 hours[3]
Excretion Renal[3] (23–46%),[4][5]Faecal[2]
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.068.916 Edit this at Wikidata
Chemical and physical data
Formula C17H27N3O4S
Molar mass 369.48 g/mol g·mol−1
3D model (JSmol)

  1. Rosenzweig P, Canal M, Patat A, Bergougnan L, Zieleniuk I, Bianchetti G: A review of the pharmacokinetics, tolerability and pharmacodynamics of amisulpride in healthy volunteers. Hum Psychopharmacol. 2002 Jan;17(1):1-13. [PubMed:12404702]
  2. Moller HJ: Amisulpride: limbic specificity and the mechanism of antipsychotic atypicality. Prog Neuropsychopharmacol Biol Psychiatry. 2003 Oct;27(7):1101-11. [PubMed:14642970]
  3. Weizman T, Pick CG, Backer MM, Rigai T, Bloch M, Schreiber S: The antinociceptive effect of amisulpride in mice is mediated through opioid mechanisms. Eur J Pharmacol. 2003 Oct 8;478(2-3):155-9. [PubMed:14575800]
  4. Leucht S, Pitschel-Walz G, Engel RR, Kissling W: Amisulpride, an unusual “atypical” antipsychotic: a meta-analysis of randomized controlled trials. Am J Psychiatry. 2002 Feb;159(2):180-90. [PubMed:11823257]
  5. Rehni AK, Singh TG, Chand P: Amisulpride-induced seizurogenic effect: a potential role of opioid receptor-linked transduction systems. Basic Clin Pharmacol Toxicol. 2011 May;108(5):310-7. doi: 10.1111/j.1742-7843.2010.00655.x. Epub 2010 Dec 22. [PubMed:21176108]

Patent

Publication numberPriority datePublication dateAssigneeTitle
US4052445A *1975-02-011977-10-04Deutsche Gold- Und Silber-Scheideanstalt Vormals RoesslerProcess for the production of alkyl sulfonic acids
Family To Family Citations
FR2415099B11978-01-201981-02-20Ile De France
US20100105755A1 *2008-09-122010-04-29Auspex Pharmaceuticals, Inc.Substituted benzamide modulators of dopamine receptor

Non-Patent

Title
Jeyakumar et al. (Tetrahedron Letters 47 (2006) 4573-4576) *
Sato et al. (Tetrahedron 57 (2001) 2469-2476) *
WO2019113084A1 *2017-12-052019-06-13Sunovion Pharmaceuticals Inc.Crystal forms and production methods thereof
Family To Family Citations
CN102807516A *2012-08-162012-12-05四川省百草生物药业有限公司Intermediate in amisulpride and method for preparing amisulpride by using intermediate
CN103819383A *2012-11-192014-05-28上海美迪西生物医药有限公司Synthesis method for amisulpride
CN103319385B *2013-06-182015-07-08苏州诚和医药化学有限公司Method for synthesizing 2-methoxy-4-amino-5-ethylsulfonyl benzoic acid
CN103450058B *2013-09-182015-10-14广安凯特医药化工有限公司A kind of preparation method of amisulpride acid
CN103553989B *2013-11-082015-03-11苏州诚和医药化学有限公司Synthetic method of 2-methoxyl-4-amino-5-ethyl sulfuryl methyl benzoate
CN104725292B *2015-03-232017-07-25湖北荆江源制药股份有限公司A kind of preparation method of (S) () Amisulpride
CN105237422A *2015-09-062016-01-13南京理工大学Synthetic method of 4-amino-5-chloro-2-methoxyl benzoic acid

///////////////Amisulpride, アミスルプリド , 标准品 , FDA 2020, 2020 APPROVALS, Barhemsys, SOLIAN,  Antipsychotic, Benzamides,  Dopamine Receptor Antagonist,

CCN1CCCC1CNC(=O)C1=CC(=C(N)C=C1OC)S(=O)(=O)CC

Rimegepant sulfate, リメゲパント硫酸塩;


ChemSpider 2D Image | Rimegepant | C28H28F2N6O3

Rimegepant.svg

Rimegepant

  • Molecular FormulaC28H28F2N6O3
  • Monoisotopic mass534.219116 Da
1289023-67-1 [RN]
1-Piperidinecarboxylic acid, 4-(2,3-dihydro-2-oxo-1H-imidazo[4,5-b]pyridin-1-yl)-, (5S,6S,9R)-5-amino-6-(2,3-difluorophenyl)-6,7,8,9-tetrahydro-5H-cyclohepta[b]pyridin-9-yl ester
9751
BMS 927711

Antimigraine, Calcitonin receptor-like receptor antagonist

Treatment of migraine

Rimegepant sulfate.png

str1

Structure of RIMEGEPANT SULFATE

Rimegepant sulfate (USAN)

リメゲパント硫酸塩;

Formula
(C28H28F2N6O3)2. H2SO4. 3H2O
CAS
1374024-48-2
Mol weight
1221.2386

Nurtec ODT, FDA 2020, 2020/2/27 fda approved

Biohaven Pharmaceuticals developed Rimegepant, also known as BMS-927711, acquired in 2016 from Bristol-Myers Squibb, Rimegepant, also known as BMS-927711. Rimegepant is a potent, selective, competitive and orally active calcitonin gene-related peptide (CGRP) antagonist in clinical trials for treating migraine. Rimegepant has shown in vivo efficacy without vasoconstrictor effect; it is superior to placebo at several different doses (75 mg, 150 mg, and 300 mg) and has an excellent tolerability profile.

Rimegepant is a medication for the treatment of an acute migraine with or without aura (a sensory phenomenon or visual disturbance) in adults. However, it is not to be used prophylactically. In the US, it is marketed under the brand name, Nurtec ODT.[1]

It is not indicated for the preventive treatment of migraine.[1] It is taken by mouth, to dissolve on the tongue.[1] It takes effect within an hour and can provide relief for up to 48 hours, according to Biohaven. It is not a narcotic and has no addictive potential, and consequently will not be designated a controlled substance. It works by blocking CGRP receptors. 86% of patients did not require additional rescue medication within 24 hours of a single dose of Nurtec. All this info was obtained from a press release from Biohaven. (https://www.prnewswire.com/news-releases/biohavens-nurtec-odt-rimegepant-receives-fda-approval-for-the-acute-treatment-of-migraine-in-adults-301013021.html)

Rimegepant was approved for use in the United States as of February 27th, 2020 by the U.S. Food and Drug Administration (FDA) to be produced and marketed by Biohaven Pharmaceuticals.[2]

Charlie Conway

Charlie Conway, Chief Scientific Officer at Biohaven Pharmaceuticals

Vlad Coric, M.D.

Vlad Coric, M.D., CEO at Biohaven

No alternative text description for this image

 

clip

https://www.biohavenpharma.com/investors/news-events/press-releases/02-27-2020

BIOHAVEN’S NURTEC™ ODT (RIMEGEPANT) RECEIVES FDA APPROVAL FOR THE ACUTE TREATMENT OF MIGRAINE IN ADULTS
– First and only calcitonin gene-related peptide (CGRP) receptor antagonist available in a fast-acting orally disintegrating tablet (ODT)- A single oral dose of NURTEC ODT 75 mg can provide fast pain relief and return patients to normal function within one hour, and deliver sustained efficacy that lasts up to 48 hours for many patients- 86 percent of patients treated with a single dose of NURTEC ODT did not use a migraine rescue medication within 24 hours- Biohaven to host investor conference call on Friday, February 28, 2020 at 8:00 am ET

NEW HAVEN, Conn., Feb. 27, 2020 /PRNewswire/ — Biohaven Pharmaceutical Holding Company Ltd. (NYSE: BHVN) today announced that the U.S. Food and Drug Administration (FDA) has approved NURTEC™ ODT (rimegepant) for the acute treatment of migraine in adults. NURTEC ODT is the first FDA-approved product for Biohaven, a company dedicated to advancing innovative therapies for neurological diseases.

Nurtec™ ODT convenient 8-count package

NURTEC™ ODT Convenient 8-count Package

 NURTEC™ ODT zoom in showing one individual quick-dissolving tablet (not actual size)

A single quick-dissolving tablet of NURTEC ODT can provide fast pain relief and return patients to normal function within one hour, and deliver sustained efficacy that lasts up to 48 hours for many patients. NURTEC ODT disperses almost instantly in a person’s mouth without the need for water, offering people with migraine a convenient, discreet way to take their medication anytime and anywhere they need it. NURTEC ODT is not indicated for the preventive treatment of migraine. Biohaven expects topline results from its prevention of migraine trial later this quarter.

Vlad Coric, M.D., CEO of Biohaven commented, “The FDA approval of NURTEC ODT marks an important milestone for the migraine community and a transformative event for Biohaven. Millions of people suffering from migraine are often not satisfied with their current acute treatment, at times having to make significant tradeoffs because of troublesome side effects and reduced ability to function. NURTEC ODT is an important new oral acute treatment for migraine that offers patients the potential to quickly reduce and eliminate pain and get back to their lives.” Dr. Coric added, “We believe NURTEC ODT will be the first of many innovative Biohaven medicines to become available to treat devastating neurological diseases, a therapeutic category many other companies have abandoned. We are dedicated to helping patients with these conditions, who often have limited or no treatment options, live better, more productive lives.”

NURTEC ODT, with its novel quick-dissolve oral tablet formulation, works by blocking CGRP receptors, treating a root cause of migraine. NURTEC ODT is not an opioid or narcotic, does not have addiction potential and is not scheduled as a controlled substance by the U.S. Drug Enforcement Administration.

NURTEC ODT may offer an alternative treatment option, particularly for patients who experience inadequate efficacy, poor tolerability, or have a contraindication to currently available therapies. More than 3,100 patients have been treated with rimegepant with more than 113,000 doses administered in clinical trials, including a one-year long-term safety study. In the pivotal Phase 3 trial, NURTEC ODT was generally well tolerated; the most common adverse reaction was nausea (2%) in patients who received NURTEC ODT compared to 0.4% of patients who received placebo.

Mary Franklin, Executive Director of the National Headache Foundation commented, “Everyone knows someone living with migraine, yet it remains an invisible disease that is often overlooked and misunderstood. Almost all people with migraine need an acute treatment to stop a migraine attack as it occurs, which can happen without warning. The approval of NURTEC ODT is exciting for people with migraine as it provides a new treatment option to help people regain control of their attacks and their lives.”

Peter Goadsby, M.D., Ph.D., Professor of Neurology and Director of the King’s Clinical Research Facility, King’s College Hospital commented, “I see many patients in my practice whose lives are disrupted by migraine, afraid to go about everyday life in case of a migraine attack. Many feel unsure if their acute treatment will work and if they can manage the side effects. With the FDA approval of NURTEC ODT, there is renewed hope for people living with migraine that they can get back to living their lives without fear of the next attack.”

The FDA approval of NURTEC ODT is based on results from the pivotal Phase 3 clinical trial (Study 303) and the long-term, open-label safety study (Study 201). In the Phase 3 trial, NURTEC ODT achieved statistical significance on the regulatory co-primary endpoints of pain freedom and freedom from most bothersome symptom (MBS) at two hours post dose compared to placebo. NURTEC ODT also demonstrated statistical superiority at one hour for pain relief (reduction of moderate or severe pain to no pain or mild pain) and return to normal function. The benefits of pain freedom, pain relief, return to normal function and freedom from MBS were sustained up to 48 hours for many patients. Importantly, these benefits were seen with only a single dose of NURTEC ODT. Eighty-six percent of patients treated with NURTEC ODT did not require rescue medication (e.g. NSAIDS, acetaminophen) within 24 hours post dose. The long-term safety study assessed the safety and tolerability of rimegepant with multiple doses used over up to one year. The study evaluated 1,798 patients, who used rimegepant 75 mg as needed to treat migraine attacks, up to one dose per day. The study included 1,131 patients who were exposed to rimegepant for at least six months, and 863 who were exposed for at least one year, all of whom treated an average of at least two migraine attacks per month. The safety of treating more than 15 migraines in a 30-day period has not been established.

NURTEC ODT is contraindicated in patients with a history of hypersensitivity to rimegepant, NURTEC ODT, or to any of its components. Hypersensitivity reactions with dyspnea and severe rash, including delayed serious hypersensitivity days after administration, occurred in less than 1% of subjects taking NURTEC ODT in clinical studies.

Biohaven Conference Call Information
Biohaven is hosting a conference call and webcast on Friday, February 28, 2020, at 8:00 a.m. ET.  Participants are invited to join the conference by dialing 877-407-9120 (toll-free) or 412-902-1009 (international). To access the audio webcast with slides, please visit the “Events & Presentations” page in the Investors section of the Company’s website.

Biohaven’s Commitment to Patient Access 
Biohaven is committed to supporting the migraine community by eliminating barriers to medication access. The company has launched a patient support program. For more information and to enroll, please call 1-833-4-NURTEC or visit www.nurtec.com.

NURTEC ODT will be available in pharmacies in early March 2020 in packs of eight tablets. Each eight tablet pack covers treatment of eight migraine attacks with one dose, as needed, up to once daily.  Sample packs containing two tablets will also be made available to healthcare providers. Patients with migraine should discuss with their primary care provider or neurologist whether NURTEC ODT is appropriate for them.

About NURTEC ODT
NURTEC™ ODT (rimegepant) is the first and only calcitonin gene-related peptide (CGRP) receptor antagonist available in a quick-dissolve ODT formulation that is approved by the U.S. Food and Drug Administration (FDA) for the acute treatment of migraine in adults. The activity of the neuropeptide CGRP is thought to play a causal role in migraine pathophysiology. NURTEC ODT is a CGRP receptor antagonist that works by reversibly blocking CGRP receptors, thereby inhibiting the biologic activity of the CGRP neuropeptide. The recommended dose of NURTEC ODT is 75 mg, taken as needed, up to once daily. For more information about NURTEC ODT, visit www.nurtec.com.

About Migraine
Nearly 40 million people in the U.S. suffer from migraine and the World Health Organization classifies migraine as one of the 10 most disabling medical illnesses. Migraine is characterized by debilitating attacks lasting four to 72 hours with multiple symptoms, including pulsating headaches of moderate to severe pain intensity that can be associated with nausea or vomiting, and/or sensitivity to sound (phonophobia) and sensitivity to light (photophobia). There is a significant unmet need for new acute treatments as more than 90 percent of migraine sufferers are unable to work or function normally during an attack.

About CGRP Receptor Antagonism
Small molecule CGRP receptor antagonists represent a novel class of drugs for the treatment of migraine. This unique mode of action potentially offers an alternative to current agents, particularly for patients who have contraindications to the use of triptans, or who have a poor response to triptans or are intolerant to them.

What is NURTEC ODT? 
NURTEC™ ODT (rimegepant) is indicated for the acute treatment of migraine with or without aura in adults.

No alternative text description for this image

Raising the “flag of freedom from migraine” over Biohaven headquarters in New Haven CT

Mechanism of action

Rimegepant is a small molecule calcitonin gene-related peptide (CGRP) receptor antagonist.[3]

PATENTS

WO 2011046997

PATENT

WO 2012050764

https://patents.google.com/patent/WO2012050764A1

The disclosure generally relates to a synthetic process for preparing compounds of formula I including the preparation of chemical intermediates useful in this process. CGRP inhibitors are postulated to be useful in pathophysiologic conditions where excessive CGRP receptor activation has occurred. Some of these include neurogenic vasodilation, neurogenic inflammation, migraine, cluster headache and other headaches, thermal injury, circulatory shock, menopausal flushing, and asthma. CGRP antagonists have shown efficacy in human clinical trials. See Davis CD, Xu C. Curr Top Med Chem. 2008 8(16):1468-79; Benemei S, Nicoletti P, Capone JG, Geppetti P. Curr Opin Pharmacol 2009 9(1):9-14. Epub 2009 Jan 20; Ho TW, Ferrari MD, Dodick DW, Galet V, Kost J, Fan X, Leibensperger H, Froman S, Assaid C, Lines C, Koppen H, Winner PK. Lancet. 2008 372:2115. Epub 2008 Nov 25; Ho TW, Mannix LK, Fan X, Assaid C, Furtek C, Jones CJ, Lines CR, Rapoport AM; Neurology 2008 70: 1304. Epub 2007 Oct 3.

CGRP receptor antagonists have been disclosed in PCT publications WO 2004/092166, WO 2004/092168, and WO 2007/120590. The compound (5S,6S,9R)- 5-amino-6-(2,3-difluorophenyl)-6,7,8!9-tetrahydiO-5H-cyclohepta[b]pyridin-9-yl 4- (2-oxo-2,3-dihydiO-lH-imidazo[4,5-b]pyridin-l-yl)piperidine-l-carboxylate is an inhibitor of the calcitonin gene-related peptide (CGRP) receptor.

Figure imgf000004_0001
Figure imgf000005_0001

cheme 1 illustrates a synthesis of formula I compounds. heme 1,

Figure imgf000011_0001

DESCRIPTION OF SPECIFIC EMBODIMENTS

Figure imgf000012_0001

( 6S, 9R)-6~ (2, 3 -difluorophenyl)-9-(triisopropylsiIyloxy) – 6, 7, 8, 9-tetrahydro-5H- cyclohepta[b]pyridin-5 -amine. To a 100 mL hastelloy autoclave reactor was charged (6S,9R)-6-(2,3-difluorophenyl)-9-(triisopiOpylsilyloxy)-6,7,8,9-tetrahydi -5H- cyclohepta[b]pyridin-5-one (5.00 g, 1 1.22 mmol), 1,4-dioxane (50 mL) and titanium tetra(isopropoxide) (8.33 mL, 28.11 mmol). The reactor was purged three times with nitrogen and three times with ammonia. After the purge cycle was completed, the reactor was pressurized with ammonia to 100 psig. The reaction mixture was heated to 50°C (jacket temperature) and stirred at a speed to ensure good mixing. The reaction mixture was aged at 100 psig ammonia and 50°C for 20 h. The mixture was then cooled to 20°C then 5 % Pd/Alumina (1.0 g, 20 wt%) was charged to the autoclave reactor. The reactor was purged three times with nitrogen and three times with hydrogen. After the purged cycle completed, the reactor was pressurized with hydrogen to 100 psig and mixture was heated to 50°C (jacket temperature) and stirred at a speed to ensure good mixing. The reaction mixture was aged at 100 psig H2 and 50°C for 23h (reactor pressure jumped to -200 psig due to soluble ammonia in the mixture). The mixture was then cooled to 20 °C then filtered then transferred to a 100 ml 3-necked flask. To the mixture water (0.55 mL) was added drop wise, which resulted in yellow slurry. The resulting slurry was stirred for 30 mm then filtered, then the titanium dioxide cake was washed with 1,4-dioxane (30 mL). The filtrate was collected and the solvent was removed. The resulting oil was dissolved in isopropanol (40 mL). To the solution ~5N HC1 in isopropanol (9.0 ml) was added drop wise resulting in a thick slurry. To the slurry isopropyi acetate (60 ml) was added and heated to 45 °C for 10 min and then cooled to 22 °C over approximately 3 h to afford a white solid (3.0 g, 51.5 %). Ή NMR (500 MHz, CD3OD)

δ ppm 8.89 (d, J= 5.3, 1H), 8,42 (bs, 1H), 8.05 (bs, 1H), 7.35 (dd, J= 8.19 , 16.71), 7.2 (bs, 2H), 7.22 (m, 1H) 7.15 (m, 1H), 5.7 (dd, J = 1.89, J = 8.51), 5.4 (m, 1H), 3.5 ( m, 1H), 1.9-2.5 (B, 4h) 1.4 (sept, J = 15.13,3H), 1.2 (t, J= Ί.5Ί 18H); 13C NMR (125 MHz, CD3OD) δ 153.5, 151.6, 151.5, 151.3, 149.4, 143.4, 135.03, 129.8, 129.8, 127.8, 126.8, 126.4, 118.6, 72.4, 54.1, 41.4, 34.3, 32.3, 25.4, 18.6, 18.5, 13.7, 13.6, 13.5, 13.3.

Example 2

Figure imgf000013_0001

(6S,9R)-5-cmino-6-(2 -difluorophenyl)-6, 7,8,9-tetrahydro~5H-cyclohepta[b^ 9-o To a 250 ml flask was charged (6S,9R)-6-(253-difluoiOphenyl)-9-

(tnisopiOpylsilyloxy)-6,7,8,9-tetrahydro-5H-cyclohepta[b]pyridin-5-amine di HC1 salt (15 g, 25.88 mtnol) and a solution of isopropanol: water (45 mL : 15 mL). The mixture was heated to 82 °C for 6h then dried via azeotropic distillation at atmospheric pressure using isopropanol until the KF was less than < 3 %. After fresh isopropanol (25 ml) was added, the mixture was heated to 70 °C and then isopropyl acetate (45 ml) was added that resulting in a white slurry. The slurry cooled to 22 °C for 15 min to afford a white solid (9.33 g, 99%). 1H NMR (500 MHz CD3OD) δ 8.77 (d, J= 5.7 Hz, 1H), 8.47 (d, J= 7.9 Hz, 1H), 8.11 (dd, J= 6.0, 8.2 Hz, 1H), 7.21-7.32 (m, 3H), 5.53 (dd, J= 3.8, 9.8 Hz, 1H) 5.33 (d, J = 9.8 Hz, 1H), 3.5 (bm, 1H), 2.25- 2.40 (m, 2H), 2.15 (bm, 1H), 1.90 (bm, 1H); 13C NMR (125 MHz, MeOD) δ 159.4, 153.9, 151.9 and 151.8, 149.7, 143.6, 141.8, 135.7, 130.6, 127.7, 126.8, 1 18.9, 70.0, 54.9, 42.2, 34.5, 33.4. Example 3

Figure imgf000014_0001

(5S, 6S, 9R)-5-amino-6-(2, 3-difluorophenyl)-6, 7>8,9-tetrahydro-5H- cyclohepta[b ]pyridin-9~yl~4-(2-oxo-2, 3-dihydro-lH-imidazo[4, 5-b ]pyridin-l- yl)piperidine-l-carboxylate. To a round bottom flask was charged (5S,6S,9R)-5- amino-6-(2,3-difluorophenyl)-6,7,8,9-tetrahydro-5H-cyclohepta[b]pyridin-9-ol dihydrochloride (1.00 g, 2.73 mmol) and dichloromethane (15 mL). A solution of sodium carbonate (0.58 g, 5.47 mmol), 20 wt% aqueous sodium chloride (5 mL), and water (10 mL) was added and the biphasic mixture was aged for 30 min. The phases were allowed to separate and the organic stream was retained. The dichloromethane solvent was then switched with azeotropic drying to tetrahydrofuran, with a final volume of (15 mL). At 20 °C was added, l-(l-(lH~imidazole-l-carbonyl)piperidin- 4-yl)-lH-imidazo[4,5-b]pyridin-2(3H)-one (0.95 g, 3.01 mmol), followed by a 20 wt% potassium ter/-butoxide solution in THF (4 mL, 6.20 mmol). The thin slurry was aged for lh, and then the reaction was quenched with the addition of 20 wt% aqueous sodium chloride (5 mL) and 20 wt% aqueous citric acid (2.5 mL). The layers were allowed to separate and the organic rich layer was retained. The organic layer was washed with 20 wt% aqueous sodium chloride (1 mL). The organic tetrahydrofuran stream was then concentrated in vacuo to afford an oil which was resuspended in dichloromethane (20 mL) and dried with MgS04. The

dichloromethane stream was concentrated in vacuo to afford an oil, which was crystallized from ethanohheptane to afford a white solid (1.14 g, 78.3%). LCMS: [M+H] = 535: 1H MR (600 MHz, 6-DMSO) δ 11.58 (IH, bs), 8.45 (IH, bd), 8.03 (IH, d, J= 7.3 Hz), 7.91 (IH, bs), 7.54 (IH, bd), 7.36 (IH, bm), 7.34 (IH, bm), 7.28 (IH, m), 7.21 (IH, m), 7.01 (IH, bs), 6.01 (IH, dd, J= 3.2, 9.8 Hz), 4.48 (IH, d, J= 9.5 Hz), 4.43 (IH, bm), 4.38 (IH, bm), 4.11 (IH, bm), 3.08 (IH, bm), 2.93 (IH, bm), 2.84 (IH, m), 2.62 (IH, bm), 2.20 (2H, bm), 2.13 (IH, bm), 2.12 (IH, bm), 1.75 (IH, bm), 1.72 (1H, bm), 1.66 (1H, bm); C NMR (125 MHz, i/6-DMSO) δ 156.6, 154.2, 153.0, 149.8, 148.1, 146.4, 143.5, 139.6, 137.4, 134.0, 132.8, 124.7, 124.5, 123.3, 122.2, 116.3, 115.0, 114.3, 73.7, 52.8, 50.0, 43.8, 43.3, 32.0, 30.3, 28.6; nip 255°C.

Example 4

Figure imgf000015_0001

l-(l-(lH^mdazole-l-carbonyl)piperidin-4-yl)-lH-imidazo

To a round bottom flask was added, Ι,Γ-carbonyldiimidazole (8.59 g, 51.4 mmoi), diisopropylethylamine (12.6 mL, 72.2 mmol) and tetrahydrofuran (100 niL). This mixture was warmed to 40°C and aged for 10 min, after which l-(piperidin-4-yl)-lH- imidazo[4,5-b]pyridin-2(3H)-one dihydrochloride (10 g, 34,3 mmol) was added. The slurry was aged at 40 °C for 3 h, and then upon reaction completion, the solvent was swapped to acetonitrile which afforded an off white solid (9.19 g, 85.9%). LCMS: [M+H] = 313; Ή NMR (400 MHz, 6-DMSO) δ 11.58 (1H, s), 8.09 (1H, s), 7.97 (1H, d, J= 8.0 Hz), 7.73 (1H, d, J= 4.0 Hz), 7.53 (1H, s), 7.05 (1H, s), 7.00 (1H, dd, J= 4.0, 8.0 Hz), 4.52, (1H, dd, J= 8.0, 12.0 Hz), 4.05 (2H, bd, J= 8,0 Hz), 3.31 (2H, m), 2.34 (2H, m), 1.82 (2H, bd, J = 12.0 Hz); 13C NMR (100 MHz, i/6~DMSO) δ 153.0, 150.4, 143.4, 139.8, 137.2, 128.9, 123.0, 1 18.7, 116.4, 115.2, 49.3, 45.1 , 28.5; mp 226°C.

Example 5

Figure imgf000015_0002

l-(l-(lH-imidazole-l-carbonyl)piperidin-4-yl)-lH-imidazo[4,5

To a 250 ml round bottom flask was added 3-N-piperidin-4-ylpyridine-2, 3 -diamine dihydrochloride (10 g, 52 mmol) and acetonitrile (100 mL). Triethyl amine (11.44 g, 1 13 mmol) and 1 , -Carbonyldiimidazole (18.34 g, 113 mmol) were added at ambient temperature and the mixture was stirred for 2 h. The solvent was evaporated under vacuum to—30 ml reaction volume and isopropyl acetate (50 mL) was added into the resulting sluny at 40°C. The slurry was cooled to 10-15 °C and then stirred for 1 h to afford an off white solid (10 g, 85%).

PATENT

US 20130225636

EP 2815749

PAPER

 Journal of Medicinal Chemistry (2012), 55(23), 10644-10651.

https://pubs.acs.org/doi/full/10.1021/jm3013147

Calcitonin gene-related peptide (CGRP) receptor antagonists have demonstrated clinical efficacy in the treatment of acute migraine. Herein, we describe the design, synthesis, and preclinical characterization of a highly potent, oral CGRP receptor antagonist BMS-927711 (8). Compound 8 has good oral bioavailability in rat and cynomolgus monkey, attractive overall preclinical properties, and shows dose-dependent activity in a primate model of CGRP-induced facial blood flow. Compound 8 is presently in phase II clinical trials.

PAPER

Organic letters (2015), 17(24), 5982-5.

https://pubs.acs.org/doi/full/10.1021/acs.orglett.5b02921

An asymmetric synthesis of novel heterocyclic analogue of the CGRP receptor antagonist rimegepant (BMS-927711, 3) is reported. The cycloheptane ring was constructed by an intramolecular Heck reaction. The application of Hayashi–Miyaura and Ellman reactions furnished the aryl and the amine chiral centers, while the separable diastereomeric third chiral center alcohols led to both carbamate and urea analogues. This synthetic approach was applicable to both 6- and 5-membered heterocycles as exemplified by pyrazine and thiazole derivatives.

History

Originally discovered at Bristol-Myers Squibb,[4] it was under development by Biohaven Pharmaceuticals and is now also being marketed in the US by the same company after receiving FDA approval late February 2020.[5]

References

  1. Jump up to:a b c “Nurtec ODT (rimegepant) orally disintegrating tablets, for sublingual or oral use” (PDF). February 2020. Retrieved 27 February 2020.
  2. ^ “Nurtec ODT: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 28 February 2020.
  3. ^ Diener HC, Charles A, Goadsby PJ, Holle D (October 2015). “New therapeutic approaches for the prevention and treatment of migraine”. The Lancet. Neurology14 (10): 1010–22. doi:10.1016/S1474-4422(15)00198-2PMID 26376968.
  4. ^ “Rimegepant – Biohaven Pharmaceuticals Holding Company”Adis Insight. Springer Nature Switzerland AG.
  5. ^ “Rimegepant (BHV-3000) – for acute treatment of Migraine”. Biohaven Pharmaceuticals.

External links

Rimegepant
Rimegepant.svg
Clinical data
Trade names Nurtec ODT
Other names BHV-3000, BMS-927711
License data
Routes of
administration
By mouth
Drug class calcitonin gene-related peptide receptor antagonist
ATC code
  • none
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
CompTox Dashboard(EPA)
Chemical and physical data
Formula C28H28F2N6O3
Molar mass 534.568 g·mol−1
3D model (JSmol)

//////////Rimegepant , リメゲパント硫酸塩, Rimegepant sulfate,  migraine, BMS-927711, fda 2020

TRANILAST


Tranilast.svg

ChemSpider 2D Image | Tranilast | C18H17NO5

Tranilast

  • Molecular FormulaC18H17NO5
  • Average mass327.331 Da
2-{[(2E)-3-(3,4-Dimethoxyphenyl)prop-2-enoyl]amino}benzoic acid
3,4-DAA
5070
53902-12-8 [RN]

Tranilast (INN, brand name Rizaben) is an antiallergic drug. It was developed by Kissei Pharmaceuticals and was approved in 1982 for use in Japan and South Korea for bronchial asthma. Indications for keloid and hypertrophic scar were added in the 1980s.

Kissei  has developed and launched tranilast in Japan and South Korea for the treatment of allergic rhinitis, asthma and atopic dermatitis. Kissei, in collaboration with  GlaxoSmithKline  was additionally developing tranilast for the prevention of restenosis following percutaneous transluminal coronary angioplasty.

Medical uses

It is used Japan, South Korea, and China to treat asthma, keloid scars, and hypertrophic scars, and as an ophthalmic solution for allergic pink eye.[1]

It should not be taken women who are or might become pregnant, and it is secreted in breast milk.[1]

Interactions

People who are taking warfarin should not also take tranilast, as they interact.[1] It appears to inhibit UGT1A1 so will interfere with metabolism of drugs that are affected by that enzyme.[1]

Adverse effects

When given systemically, tranilast appears to cause liver damage; in a large well-conducted clinical trial it caused elevated transaminases three times the upper limit of normal in 11 percent of patients, as well as anemia, kidney failure, rash, and problems urinating.[1]

Given systemically it inhibits blood formation, causing leukopeniathrombocytopenia, and anemia.[1]

Society and culture

As of March 2018 it was marketed in Japan, China, and South Korea under the brand names Ao Te Min, Arenist, Brecrus, Garesirol, Hustigen, Krix, Lumios, Rizaben, Tramelas, Tranilast and it was marketed as a combination drug with salbutamol under the brand name Shun Qi.[2]

In 2016 the FDA proposed that tranilast be excluded from the list of active pharmaceutical ingredients that compounding pharmacies in the US could formulate with a prescription.[1]

Pharmacology

It appears to work by inhibiting the release of histamine from mast cells; it has been found to inhibit proliferation of fibroblasts but its biological target is not known.[3] It has been shown to inhibit the release of many cytokines in various cell types, in in vitro studies.[3] It has also been shown to inhibit NALP3 inflammasome activation and is being studied as a treatment for NALP3-driven inflammatory diseases.[4]

Chemistry

Tranilast is an analog of a metabolite of tryptophan, and its chemical name is 3′,4′-dimethoxycinnamoyl) anthranilic acid (N-5′).[3]

It is almost insoluble in water, easily soluble in dimethylsulfoxide, soluble in dioxane, and very slightly soluble in ether. It is photochemically unstable in solution.[3]

File:Tranilast synthesis.svg

Orally active anti-allergic agent. Prepn: K. Harita et al., DE 2402398; idem, US 3940422 (1974, 1976 both to Kissei).

Y. Kamijo, M. Kobayashi, and A. Ajisawa, Jpn. Kokai, 77/83,428 (1977) via Chem. Abstr.,

88:6,569f (1978).

Research

After promising results in three small clinical trials, tranilast was studied in a major clinical trial (the PRESTO trial) by SmithKline Beecham in partnership with Kissei for prevention of restenosis after percutaneous transluminal coronary revascularization,[5] but was not found effective for that application.[1][6]

As of 2016, Altacor was developing a formulation of tranilast to prevent of scarring following glaucoma surgery and had obtained an orphan designation from the EMA for this use.[7][8]

History

It was developed by Kissei and first approved in Japan and South Korea for asthma in 1982, and approved uses for keloid and hypertrophic scars were added later in the 1980s.[3]

PATENT

tranilast product case US03940422 , expired in all the regional territories.

PATENT

WO2013144916 claiming tranilast complexes and cocrystals with nicotinamide, saccharin, gentisic acid, salicylic acid, urea, 4-aminobenzoic acid and 2,4-dihydroxybenzoic acid

Patent

WO-2020035546

Nuformix Ltd

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020035546&tab=PCTDESCRIPTION&_cid=P11-K75NGV-11408-1

Novel crystalline forms of tranilast or its salts as histamine H1 receptor antagonist useful for treating allergy, allergic rhinitis and atopic dermatitis.

Tranilast, (2-[[3-(3,4-dimethoxyphenyl)-l-oxo-2-propenyl]amino] benzoic acid, shown below), was originally developed as an anti-allergy drug due to its ability to inhibit the release of inflammatory mediators, such as histamine, from mast cells and basophils (P. Zampini. IntJ Immunopharmacol. 1983;

Tranilast

Tranilast has been marketed in Japan, China and South Korea by Kissei Pharmaceutical Co. Ltd, for allergic conditions such as allergic conjunctivitis, bronchial asthma, allergic rhinitis and atopic dermatitis, under the Rizaben® brand name for more than thirty years. More recently tranilast has also been shown to have anti-proliferative properties. Tranilast was shown to inhibit the proliferation of fibroblasts and suppress collagen synthesis (M. Isaji. Biochem Pharmacol. 1987; 36: 469-474) and also to inhibit the transformation of fibroblasts to myofibroblasts and their subsequent contraction (M. Isaji. Life Sci. 1994; 55: 287-292). This additional behaviour led to tranilast gaining additional approval for the treatment of keloids and hypertrophic scars.

[004] Over recent years many researchers have explored the anti-proliferative effects of tranilast to assess its potential in fibrotic and cancerous conditions. Its anti-proliferative action is believed to be due to its ability to inhibit transforming growth factor beta (TGF-b) (H. Suzawa. Jpn J Pharmacol. 1992 Oct; 60(2): 91-96). Fibrosis is a condition that can affect most organs of the body and fibroblast proliferation, differentiation and collagen synthesis are known to be key factors in the progression of most types of fibrosis. Tranilast has been shown in-vivo to have potential beneficial effects in

numerous fibrotic conditions. Tranilast has been shown in-vivo to have potential in lung fibrosis (M. Kato. Eur RespirJ. 2013; 42(57): 2330), kidney fibrosis (DJ Kelly, J Am Soc Nephrol. 2004; 15(10): 2619-29), cardiac fibrosis (J Martin, Cardiovasc Res. 2005; 65(3): 694-701), ocular fibrosis (M J Moon, BMC Opthalmol. 2016; 16: 166) and liver fibrosis (M Uno, Hepatology. 2008; 48(1): 109-18.

[005] Tranilast’s anti-tumor action has also recently been demonstrated, in-vitro and in-vivo. Tranilast has been shown to inhibit the proliferation, apoptosis and migration of several cell lines including breast cancer (R. Chakrabarti. Anticancer Drugs. 2009 Jun; 20(5): 334-45) and prostate cancer (S. Sato. Prostate. 2010 Feb; 70(3): 229-38) cell lines. In a study of mammary carcinoma in mice tranilast was found to produce a significant reduction in metastasis (R. Chakrabarti. Anticancer Drugs. 2009 Jun; 20(5): 334-45). In a pilot study in humans, tranilast was shown to have the potential to improve the prognosis of patients with advanced castration-resistant prostate cancer (K. Izumi. Anticancer Research. 2010 Jul; 30: 73077-81). In-vitro studies also showed the therapeutic potential of tranilast in glioma (M Platten. IntJ Cancer. 2001; 93:53-61), pancreatic cancer (M Hiroi, J Nippon Med Sch. 2002; 69: 224-234) and gastric carcinoma (M Yashiro, Anticancer Res. 2003; 23: 3899-3904).

[006] Given the wide range of fibrotic conditions and cancers for which tranilast could have a potential therapeutic benefit, as well as the different patient types and specific areas of the body requiring treatment, it is anticipated that patients would benefit from having multiple delivery methods for the administration of tranilast so as to best suit the patient’s needs. The pharmaceutical compositions could include, for example, a solid oral dosage, a liquid oral dosage, an injectable composition, an inhalable composition, a topical composition or a transdermal composition.

[007] Kissei Pharmaceutical Co. Ltd explored the anti-proliferative effect of tranilast in the prevention of restenosis associated with coronary intervention. In a Phase II clinical study Kissei found that the current approved dose of tranilast (300 mg/day) was insufficient to prevent restenosis and that a higher dose of 600 mg/day was needed to achieve a decrease in restenosis rates (H. Tamai, Am Heart J.1999; 138(5): 968-75). However, it was found that a 600 mg daily dosage can result in a ten-fold inter-patient variation in plasma concentrations of the drug (30-300 pmol/L) (H Kusa ma. Atherosclerosis. 1999; 143: 307-313) and in the Phase III study of tranilast for the prevention of restenosis the dose was further increased to 900mg daily (D Holmes, Circulation. 2002; 106(10): 1243-1250).

[008] The marketed oral form of tranilast (Rizaben®) contains tranilast in its pure crystalline form. Crystalline tranilast has extremely low aqueous solubility (solubility of 14.5 pg/ml in water and 0.7 pg/ml in pH 1.2 buffer solution (Society of Japanese Pharmacopoeia. 2002)). Whilst, high energy amorphous forms are often used as a means of improving the solubility of poorly soluble drug

compounds, literature shows that an amorphous form of tranilast is not completely photostable in the solid state and that it undergoes photodegradation on storage when exposed to light (S. Onoue. EurJ Pharm Sci. 2010; 39: 256-262).

[009] It is expected that the very low solubility of tranilast is a limiting factor in the oral bioavailability of the drug. Given the limited time any drug has to firstly dissolve in the

gastrointestinal tract and then be absorbed into the bloodstream, this issue will become even more limiting as the oral dose of tranilast is increased. The poor solubility of tranilast is also possibly a key factor in the high inter-patient variability reported for higher dose tranilast pharmacokinetics. As a BCS class II drug (low solubility/high permeability) it is expected that absorption from the gastrointestinal tract is hampered by the dissolution rate of the drug in gastrointestinal media as well as its overall solubility. For treatment of chronic proliferative diseases such as fibrosis and cancer it is vital for the delivery method of a drug to produce consistent, predictable plasma levels that are maintained above the minimum effective concentration. To achieve efficacious oral delivery of tranilast at higher doses there is a need for new solid forms of the drug with both high solubility and rapid dissolution rates.

[010] Given the severity of conditions involving cancer or fibrosis there is also a need for systemic treatment options by which tranilast can be delivered by healthcare specialists that do not require the patient to swallow solid oral dosage forms. Alternative dosage forms suitable for these needs could include, for example, injectable compositions, liquid oral formulations or nebulized inhaled formulations. These would require a liquid formulation of tranilast suitable for systemic delivery. [Oil] Given the potential of tranilast to treat ocular diseases, such as allergic conjunctivitis, Kissei Pharmaceutical Co. Ltd recognised the need to develop an eye drop formulation of tranilast for localised treatment. However, as well as having very low aqueous solubility, tranilast is also photochemically unstable when stored in solution, resulting in significant degradation (N Hori, Chem. Pharm. Bull. 1999; 47(12): 1713-1716). Therefore, the only way Kissei were able to achieve an eye drop liquid composition of tranilast was to use both solubilising and stabilising agents in the formulation (US Patent 5356620). The resulting 0.5% (w/v) eye drop formulation is currently also marketed under the Rizaben® brand name. However, the focus of this formulation and of the subsequent research that has attempted to produce alternative solution formulations of tranilast has always been solely on external delivery of tranilast using compositions such as eye drops and skin ointments etc. None of the liquid formulations of tranilast previously described have been produced for systemic delivery such as for oral or IV delivery. Excipients used in the previously reported external preparations are not suitable for systemic delivery. Also, despite the successful

development of an eye drop formulation of tranilast, the package insert of the marketed Rizaben® eye drops states that the product should not be stored in a refrigerator as crystals may precipitate.

[012] Thus, there remains a need for aqueous pharmaceutical compositions of tranilast suitable for systemic delivery. Given the potential photochemical degradation issue of long term storage of tranilast in solution and also the disadvantage of the larger storage facilities needed to store bulkier solution based formulations it would also be advantageous to develop a stable highly soluble solid form of tranilast that can be quickly dissolved at the time of treatment by the patient or healthcare provider to produce the required liquid formulation.

[013] Following efforts to make a liquid formulation of tranilast, Kissei made the statement that tranilast and pharmaceutically acceptable salts thereof are too insoluble in water to prepare an aqueous solution (US Patent 5356620). Since that US patent the only crystalline pharmaceutically acceptable salt to have been published is the sodium salt (N Geng, Cryst. Growth Des. 2013; 13: 3546-3553). In line with the findings of Kissei the authors of this paper stated that the apparent solubility of the crystalline tranilast sodium salt is even less than that of pure tranilast. Also, when they performed a dissolution study of tranilast in a sodium containing media they found that as the tranilast dissolved it gradually precipitated out of solution as its sodium salt indicating that the sodium salt has a lower thermodynamic solubility than the pure drug. The authors of this paper also successfully prepared the non-pharmaceutically acceptable crystalline cytosine salt of tranilast. Despite this crystalline cytosine salt showing approximately a two-fold solubility improvement over pure crystalline tranilast, not only would this crystalline cytosine salt not be suitable for systemic delivery to a patient due to cytosine not having FDA acceptability but this improvement in solubility would not be great enough to produce high dose tranilast liquid formulations such as an injectable formulation.

[014] Patent application EP1946753 discloses an attempt to prepare an external preparation of tranilast and claims the preparation of ionic liquid salts of tranilast with organic amines. The inventors claim that blending tranilast with the organic amine results in a liquid form. This application does not disclose the formation of any solid state, crystalline tranilast salts with organic amines. They demonstrate that these ionic liquid forms of tranilast have higher solubility in solvents suitable for external application to the skin and that these preparations have higher photostability than pure tranilast in the same formulation. However, this improved photostability still results in a significant proportion of the tranilast being photo-degraded and would not be suitable for long term storage. Also, the solvents used for preparation of these ionic liquid salt formulations are not suitable for internal delivery of tranilast. Moreover, there is no mention in EP1946753 of improved solubility in aqueous or bio-relevant media.

PATENT

US20150119428

https://patents.google.com/patent/US20150119428

  • Tranilast, (2-[[3-(3,4-dimethoxyphenyl)-1-oxo-2-propenyl]amino]benzoic acid), shown below, is a therapeutic agent that exhibits an anti-allergic effect. It has been shown to inhibit the release of inflammatory mediators, such as histamine, from mast cells and basophils (P. Zampini. Int J Immunopharmacol. 1983; 5(5): 431-5). Tranilast has been used as an anti-allergic treatment, for several years in Japan and South Korea, for conditions such as allergic conjunctivitis, bronchial asthma, allergic rhinitis and atopic dermatitis.
  • Figure US20150119428A1-20150430-C00001
  • [0004]
    Tranilast is currently marketed in Japan and South Korea by Kissei Pharmaceutical Co. Ltd under the Rizaben® brand name. As well as displaying an anti-allergic effect tranilast has been shown to possess anti-proliferative properties. Tranilast was found to inhibit the proliferation of fibroblasts and suppress collagen synthesis (M. Isaji. Biochem Pharmacol. 1987; 36: 469-474) and also to inhibit the transformation of fibroblasts to myofibroblasts and their subsequent contraction (M. Isaji. Life Sci. 1994; 55: 287-292). On the basis of these effects tranilast is now also indicated for the treatment of keloids and hypertrophic scars. Its anti-fibrotic action is believed to be due to its ability to inhibit transforming growth factor beta (TGF-β) (H. Suzawa. Jpn J Pharmacol. 1992 October; 60(2): 91-96). TGF-β induced fibroblast proliferation, differentiation and collagen synthesis are known to be key factors in the progression of idiopathic pulmonary fibrosis and tranilast has been shown in-viva to have potential in the treatment of this chronic lung disease (T. Jiang. Afr J Pharm Pharmaco. 2011; 5(10): 1315-1320). Tranilast has also been shown in-vivo to be have potential beneficial effects in the treatment of airway remodelling associated with chronic asthma (S. C. Kim. J Asthma 2009; 46(9): 884-894.
  • [0005]
    It has been reported that tranilast also has activity as an angiogenesis inhibitor (M. Isaji. Br. J Pharmacol. 1997; 122(6): 1061-1066). The results of this study suggested that tranilast may be beneficial for the treatment of angiogenic diseases such as diabetic retinopathy and age related macular degeneration. As well as showing inhibitory effects on mast cells and fibroblasts, tranilast has also demonstrated an ability to diminish tumor necrosis factor-alpha (TNF-α) from cultured macrophages (H. O. Pae. Biochem Biophys Res Commun. 371: 361-365) and T-cells (M. Platten. Science. 310: 850-855), and inhibited NF-kB-dependent transcriptional activation in endothelial cells (M. Spieker. Mol Pharmacol. 62: 856-863). Recent studies have revealed that tranilast attenuates inflammation and inhibits bone destruction in collagen induced arthritis in mice suggesting the possible usefulness of tranilast in the treatment of inflammatory conditions such as arthritis (N. Shiota. Br. Pharmacol. 2010; 159 (3): 626-635).
  • [0006]
    As has recently been demonstrated, in-vitro and in-vivo, tranilast also possesses an anti-tumor action. Tranilast has been shown to inhibit the proliferation, apoptosis and migration of several cell lines including breast cancer (R. Chakrabarti. Anticancer Drugs. 2009 June; 20(5): 334-45) and prostate cancer (S. Sato. Prostate. 2010 February; 70(3): 229-38) cell lines. In a study of mammary carcinoma in mice tranilast was found to produce a significant reduction in metastasis (R. Chakrabarti. Anticancer Drugs. 2009 June; 20(5): 334-45). In a pilot study in humans, tranilast was shown to have the potential to improve the prognosis of patients with advanced castration-resistant prostate cancer (K. Izurni. Anticancer Research. 2010 July; 30: 73077-81).
  • [0007]
    It has been reported that tranilast has the ability to induce or enhance neurogenesis and, therefore, could be used as an agent to treat neuronal conditions such as cerebral ischernia, glaucoma, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease, neurodegenerative trinucleotide repeat disorders, neurodegenerative lyosomal storage diseases, spinal cord injury and trauma, dementia, schizophrenia and peripheral neuropathy (A. Schneider. EP2030617).
  • [0008]
    Tranilast’s beneficial properties have been reported to have utility in several ocular conditions. Tranilast is currently approved in Japan and Korea far the treatment of allergic conjunctivitis. WO2010137681 claims the use of tranilast as a prophylactic or therapeutic agent for the treatment of retinal diseases. The anti-fibrotic properties of tranilast have been reported to be of benefit in maintaining the filtering blob during glaucoma surgery and this has been demonstrated in a pilot study in humans (E. Chihara.J Glaucoma. 1999; 11(2): 127-133). There have also been several reported cases of the beneficial use of tranilast in the prevention of postoperative recurrence of pterygium (C. Fukui. Jap J Opthalmol. 1999; 12: 547-549). Tsuji recently reported that tranilast may be beneficial not only in the prevention of ptergium recurrence, but also for the inhibition of symblepharon and granuloma formation (A. Tsuji. Tokai J Exp Clin Med. 2011; 36(4): 120-123). Collectively it has been demonstrated that tranilast possesses anti-allergic, anti-fibrotic, anti-inflammatory, anti-tumor, neurogenesis enhancing end angiogenesis inhibitory properties and as such may be useful for the treatment of diseases associated with such properties.
  • [0009]
    Tranilast occurs as a yellow crystalline powder that is identified by CAS Registry Number: 53902-12-8. As is typical of cinnamic acid derivatives (G. M. J. Schmidt J Chem. Soc. 1964: 2000) tranilast is photochemically unstable when in solution, tranforming into cis-isomer and dimer forms on exposure to light (N. Hori. Cehm Pharm Bull. 1999; 47: 1713-1716). Although pure crystalline tranilast is photochemically stable in the solid state it is practically insoluble in water (14.5 μg/ml) and acidic media (0.7 μg/ml in pH 1.2 buffer solution) (Society of Japanese Pharmacopoeia. 2002). Although tranilast has shown activity in various indications, it is possible that the therapeutic potential of the drug is currently limited by its poor solubility and photostability. High energy amorphous forms are often used as a means of improving the solubility of poorly soluble APIs, however, literature shows that amorphous solid dispersions of tranilast are not completely photostable in the solid state and that they undergo photodegradation on storage when exposed to light (S. Onoue. Eur J Pharm Sci. 2010; 39: 256-262). US20110136835 describes a combination of tranilast and allopurinol and its use in the treatment of hyperuricemia associated with gout and has one mention of a “co-crystal form”, but lacks any further description or characterization.

Patent

Publication numberPriority datePublication dateAssigneeTitle
Family To Family Citations
JP2001072605A *1999-09-032001-03-21Lion CorpTransdermal and transmucosal absorption-promoting agent composition
JP2001187728A *1999-12-282001-07-10Lion CorpOphthalmic composition
JP2011225626A *2001-02-012011-11-10Rohto Pharmaceutical Co LtdEye lotion
US6585997B22001-08-162003-07-01Access Pharmaceuticals, Inc.Mucoadhesive erodible drug delivery device for controlled administration of pharmaceuticals and other active compounds
CA2548281C2003-12-092013-11-12Medcrystalforms, LlcMethod of preparation of mixed phase co-crystals with active agents
JP2005314229A *2004-03-312005-11-10Rohto Pharmaceut Co LtdTranilast-containing medicine composition
JP4843824B22004-08-182011-12-21株式会社 メドレックスTopical preparation
JP2007051089A *2005-08-182007-03-01Medorekkusu:KkPreparation for external use
WO2007046544A1 *2005-10-212007-04-26Medrx Co., Ltd.Preparation for external application comprising salt of mast cell degranulation inhibitor having carboxyl group with organic amine
WO2008078730A1 *2006-12-262008-07-03Translational Research, Ltd.Preparation for transnasal application
EP2030617A12007-08-172009-03-04Sygnis Bioscience GmbH & Co. KGUse of tranilast and derivatives thereof for the therapy of neurological conditions
CN101683330A *2008-09-232010-03-31沈阳三川医药科技有限公司Oral compound pharmaceutic preparation containing tranilast and salbutamol
US20110136835A1 *2009-09-142011-06-09Nuon Therapeutics, Inc.Combination formulations of tranilast and allopurinol and methods related thereto
EP2429495A4 *2009-05-152014-01-22Shin Nippon Biomedical Lab LtdIntranasal pharmaceutical compositions with improved pharmacokinetics
WO2010137681A12009-05-292010-12-02参天製薬株式会社Prophylactic or therapeutic agent for retinal diseases comprising tranilast, method for prevention or treatment of retinal diseases, and tranilast or pharmaceutically acceptable salt thereof and use thereof
JP2011093849A *2009-10-302011-05-12Kissei Pharmaceutical Co LtdEasily dissolvable powder inhalant composed of tranilast
JPWO2011096241A1 *2010-02-022013-06-10テルモ株式会社Bioabsorbable stent
AU2013239114B2 *2012-03-302017-07-20Nuformix LimitedTranilast compositions and cocrystals

Family To Family Citations
AU2013239114B2 *2012-03-302017-07-20Nuformix LimitedTranilast compositions and cocrystals
US10155757B22015-03-102018-12-18Vectura LimitedCrystalline form of a JAK3 kinase inhibitor
CN106344550A *2016-09-282017-01-25江苏省人民医院Application of tranilast to preparation of medicines for treating pneumoconiosis
CN107286210A *2017-06-192017-10-24昆药集团股份有限公司A kind of Acegastrodine compound and preparation method thereof, preparation and application

References

  1. Jump up to:a b c d e f g h “FDA Proposed Rules” (PDF)Federal Register81 (242): 91071–91082. December 16, 2016. Another version of same published at here
  2. ^ “International brands for Tranilast”. Drugs.com. Retrieved 10 March 2018.
  3. Jump up to:a b c d e Darakhshan, S; Pour, AB (January 2015). “Tranilast: a review of its therapeutic applications”. Pharmacological Research91: 15–28. doi:10.1016/j.phrs.2014.10.009PMID 25447595.
  4. ^ Y. Huang et al, “Tranilast directly targets NLRP3 to treat inflammasome-driven diseases.”EMBO Mol Med., 10(4), 2018
  5. ^ “Kissei’s existing business flat but R&D pipeline should lead to growth”The Pharma Letter. 8 September 2000.
  6. ^ Holmes, D. R; Savage, M; Lablanche, J. M; Grip, L; Serruys, P. W; Fitzgerald, P; Fischman, D; Goldberg, S; Brinker, J. A; Zeiher, A. M; Shapiro, L. M; Willerson, J; Davis, B. R; Ferguson, J. J; Popma, J; King Sb, 3rd; Lincoff, A. M; Tcheng, J. E; Chan, R; Granett, J. R; Poland, M (2002). “Results of Prevention of REStenosis with Tranilast and its Outcomes (PRESTO) Trial”. Circulation106 (10): 1243–50. doi:10.1161/01.CIR.0000028335.31300.DAPMID 12208800.
  7. ^ “Tranilast – Altacor: ALT-401”AdisInsight. Retrieved 10 March 2018.
  8. ^ “EU/3/10/756 Orphan Designation”. European Medicines Agency. 6 August 2010. Retrieved 10 March 2018.
Tranilast
Tranilast.svg
Clinical data
AHFS/Drugs.com International Drug Names
Routes of
administration
Oral
ATC code
  • none
Legal status
Legal status
  • US: Not FDA approved
  • In general: ℞ (Prescription only)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
ChemSpider
UNII
ChEBI
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.150.125 Edit this at Wikidata
Chemical and physical data
Formula C18H17NO5
Molar mass 327.336 g·mol−1
3D model (JSmol)

///////////////Tranilast,  Rizaben, antiallergic,  Kissei Pharmaceuticals,  Japan, South Korea,  bronchial asthma,  keloid,  hypertrophic scar

Enavogliflozin, DWP-16001


Image result for south korea flag gif

str1

Enavogliflozin, DWP-16001

(2S,3R,4R,5S,6R)-2-(7-chloro-6-(4-cyclopropylbenzyl)-2,3-dihydrobenzofuran-4-yl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol

(2S,3R,4R,5S,6R)-2-(7-chloro-6-(4-cyclopropylbenzyl)-2,3-dihydrobenzofuran-4-yl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol

CAS: 1415472-28-4
Chemical Formula: C24H27ClO6
Molecular Weight: 446.92
Elemental Analysis: C, 64.50; H, 6.09; Cl, 7.93; O, 21.48

Green Cross Corp INNOVATOR

Daewoong Pharmaceutical Co Ltd

Enavogliflozin is an antidiabetic (hypoglycemic).

Daewoong is investigating DWJ-304 , a sodium/glucose cotransporter 2 (SGLT-2) inhibitor, for treating type 2 diabetes. By February 2017, preclinical development was underway. Daewoong is developing DWP-16001 , presumed to be enavogliflozin, a SGLT-2 inhibitor, for treating type 2 diabetes. In September 2019, launch was expected in 2023.

Enavogliflozin (DWP16001/GCC5694A) is a selective SLC5A2 and SGLT2 inhibitor developed to treat diabetes and obesity.[1][2][3][4][5] It was developed by GC Pharma[6][7] and Daewoong Pharmaceutical.[7]

Enavogliflozin has been approved for clinical use in South Korea,[7] and Ecuador,[8] and applied for approval in BrazilMexicoPeru, and Colombia.[8]

PATENT

WO2012165914

DWP-16001 expire in  EU states until June 2032 and  US in November 2033.

PATENT

US 2014274918

PATENT

US2019169174

Paragraph 0305; 0340; 0347

H NMR (400 MHz, CD3OD) δ 7.02 (d, J=8.0 Hz, 2H), 6.92 (d, J=8.0 Hz, 2H), 6.81 (s, 1H), 4.59 (t, J=8.8 Hz, 2H), 4.11 (d, J=9.2 Hz, 1H), 3.96 (ABq, ΔvAB=19.0 Hz, JAB=15.2 Hz, 2H), 3.87-3.84 (m, 1H), 3.67-3.63 (m, 1H), 3.47-3.37 (m, 3H), 3.35-3.33 (m, 3H), 1.85-1.79 (m, 1H), 0.91-0.86 (m, 2H), 0.61-0.57 (m, 2H)

PATENT

WO2017217792 , claiming process for preparing diphenylmethane derivative.

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017217792&tab=FULLTEXT&_cid=P21-K72SDW-38338-1

1H NMR(400 MHz, CD 3OD) δ 7.02(d, J = 8.0 Hz, 2H), 6.92(d, J = 8.0 Hz, 2H), 6.81(s, 1H), 4.59(t, J = 8.8 Hz, 2H), 4.11(d, J = 9.2 Hz, 1H), 3.96(ABq, Δv AB = 19.0 Hz, J AB = 15.2 Hz, 2H), 3.87-3.84(m, 1H), 3.67-3.63(m, 1H), 3.47-3.37(m, 3H), 3.35-3.33(m, 3H), 1.85-1.79(m, 1H), 0.91-0.86(m, 2H), 0.61-0.57(m, 2H); [M+Na] + 469.

PATENT

WO-2020036382

The present invention relates to a method for producing an intermediate useful for the synthesis of a diphenylmethane derivative that can be used as a SGLT inhibitor. A method for synthesizing a compound of formula 7 according to the present invention has solved the problem of an existing synthesis process which requires an additional process due to the synthesis of Grignard reagent and the management of a related substance. In addition, the process can be simplified by minimizing the formation of the related substance and eliminating the need for reprocessing of reaction products, thereby becoming capable of maximizing a yield of a diphenylmethane derivative.

Process for preparing intermediates of SGLT inhibitor and their use for the synthesis of diphenyl-methane derivative, which can be used as SGLT inhibitors.

Sodium-dependent glucose cotransporters (SGLT) allow the transport of Na + along the concentration gradient simultaneously with the transport of glucose across the concentration gradient. Currently two important SGLT isoforms have been cloned, known as SGLT1 and SGLT2. SGLT1 is located in the intestine, kidney and heart and regulates cardiac glucose transport. SGLT1 is a high affinity low dose transporter and therefore only accounts for a portion of renal glucose reuptake. In contrast, SGLT2 is a low affinity, high dose transporter located primarily in the apica domain of epithelial cells in the early proximal manure tubules. In healthy individuals, over 99% of the plasma glucose filtered out of the renal glomeruli is reabsorbed and less than 1% of the total filtered glucose is excreted in the urine. It is estimated that 90% of renal glucose reuptake is promoted by SGLT2 and the remaining 10% is mediated by SGLT1 in the late proximal canal. Genetic mutations in SGLT2 do not have a particular adverse effect on carbohydrate metabolism but cause increased kidney glucose secretion of about 140 g / day following mutation. Human mutation studies have been the subject of therapeutic studies because SGLT2 is believed to be responsible for most renal glucose resorption.

[3]
Korean Unexamined Patent Publication No. 2017-0142904 discloses a method for producing a diphenylmethane derivative having inhibitory activity against SGLT2. Since the above document prepares diphenylmethane derivatives by a convergent synthesis method in which each group is individually synthesized and then coupled, the synthesis route is more concise and yield is higher than the linear synthesis method disclosed in the prior art. It is disclosed that it can increase and reduce the risks inherent in sequential synthesis pathways.

[4]
However, the preparation method of the diphenylmethane derivative according to Korean Patent Publication No. 2017-0142904 uses a heavy metal such as pyridinium chlorochromate (PCC) to burden safety management, and the Grignard reagent. In addition to the need for a separate manufacturing process, the cost of the additional process is not only incurred, but also the management of the flexible material is necessary because the flexible material from the Grignard reagent manufacturing process is included in the final product. In addition, since the product generated after the reaction between the intermediate and the Grignard reagent includes additional flexible materials, there is a problem that a reprocessing process of such flexible materials is required.
Step 3. 4- bromo- 7- chloro -6- (4- cyclopropylbenzyl ) -2,3- dihydrobenzofuran (Compound 6)
 
(4-bromo-7-chloro-2,3-dihydrobenzofuran-6-yl) (4-cyclopropylphenyl) methanone (Compound 5) in a mixture of dichloromethane (9.7 mL) and acetonitrile (9.7 mL) at -15 ° C. g, 2.57 mmol) was added Et 3 SiH (1.2 mL, 7.71 mmol) and BF 3 -Et 2 O (0.79 mL, 6.42 mmol) in this order. The reaction mixture was allowed to warm to room temperature and then stirred for 4 hours. After completion of the reaction by TLC, the reaction solution was added with saturated NaHCO 3aqueous solution (40 mL) to terminate the reaction, and extracted with ethyl acetate. The organic layer obtained by extraction was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The concentrated residue was purified by silica gel chromatography to give the title compound 6 (0.84 g, 89.9%) as an off-white solid.

[387]
1 H NMR (500 MHz, CDCl 3): δ 7.07 (d, J = 10.0 Hz, 2H), 6.99 (d, J = 10.0 Hz, 2H), 6.80 (s, 1H), 4.70 (t, J = 11.0 Hz , 2H), 3.97 (s, 2H), 3.26 (t, J = 11.0 Hz, 2H), 1.88-1.84 (m, 1H), 0.95-0.90 (m, 2H), 0.68-0.64 (m, 2H); LC-MS: [M + H] & lt; + & gt; 363.
 

REFERENCES

1: Markiewicz M, Jungnickel C, Stolte S, Białk-Bielińska A, Kumirska J, Mrozik W. Ultimate biodegradability and ecotoxicity of orally administered antidiabetic drugs. J Hazard Mater. 2017 Jul 5;333:154-161. doi: 10.1016/j.jhazmat.2017.03.030. Epub 2017 Mar 16. PubMed PMID: 28349868.

2: Holt RI. Trials of new anti-diabetes agents. Diabet Med. 2017 Feb;34(2):147. doi: 10.1111/dme.13306. PubMed PMID: 28090726.

References

  1.  Hwang, Jun Gi; Lee, SeungHwan; Huh, Wan; Han, Jumi; Oh, Jaeseong; Jang, In-Jin; Yu, Kyung-Sang (September 2022). “Dose-dependent glucosuria of DWP16001, a novel selective sodium–glucose cotransporter-2 inhibitor, in healthy subjects”British Journal of Clinical Pharmacology88 (9): 4100–4110. doi:10.1111/bcp.15348PMID 35395697.
  2.  Kim, Ju-Hyun; Kim, Dong Kyun; Choi, Won-Gu; Ji, Hye-Young; Choi, Ji-Soo; Song, Im-Sook; Lee, Sangkyu; Lee, Hye Suk (11 September 2020). “In Vitro Metabolism of DWP16001, a Novel Sodium-Glucose Cotransporter 2 Inhibitor, in Human and Animal Hepatocytes”Pharmaceutics12 (9): 865. doi:10.3390/pharmaceutics12090865PMC 7558535PMID 32932946.
  3.  Kim, Byungwook; Huh, Ki Young; Hwang, Jun Gi; Nah, JaeJin; Huh, Wan; Cho, Jae Min; Jang, In-Jin; Yu, Kyung-Sang; Kim, Yun; Lee, SeungHwan (April 2023). “Pharmacokinetic and pharmacodynamic interaction between DWP16001, an sodium–glucose cotransporter 2 inhibitor and metformin in healthy subjects”. British Journal of Clinical Pharmacology89 (4): 1462–1470. doi:10.1111/bcp.15613PMID 36422809S2CID 253838705.
  4.  Rhee, Beomseok; Mahbubur, Rahman Md; Jin, Changfan; Choi, Ji-Soo; Lim, Hyun-Woo; Huh, Wan; Park, Joon Seok; Han, Jumi; Kim, Sokho; Lee, Youngwon; Park, Jinho (December 2022). “Evaluation of safety and anti-obesity effects of DWP16001 in naturally obese dogs”BMC Veterinary Research18 (1): 237. doi:10.1186/s12917-022-03324-2PMC 9214997PMID 35733159.
  5.  Yoon, Sukyong; Park, Min Soo; Jin, Byung Hak; Shin, Hyobin; Na, Jaejin; Huh, Wan; Kim, Choon Ok (3 July 2023). “Pharmacokinetic and pharmacodynamic interaction of DWP16001, a sodium-glucose cotransporter-2 inhibitor, with phentermine in healthy subjects”. Expert Opinion on Drug Metabolism & Toxicology19 (7): 479–485. doi:10.1080/17425255.2023.2249397PMID 37593838S2CID 265846294.
  6.  Kong, Young Kyu; Song, Kwang-Seop; Jung, Myung Eun; Kang, Misuk; Kim, Hyeon Jung; Kim, Min Ju (2022-01-15). “Discovery of GCC5694A: A potent and selective sodium glucose co-transporter 2 inhibitor for the treatment of type 2 diabetes”Bioorganic & Medicinal Chemistry Letters56: 128466. doi:10.1016/j.bmcl.2021.128466ISSN 0960-894X.
  7.  Kim, Min-Soo; Song, Yoo-Kyung; Choi, Ji-Soo; Ji, Hye Young; Yang, Eunsuk; Park, Joon Seok; Kim, Hyung Sik; Kim, Min-Joo; Cho, In-Kyung; Chung, Suk-Jae; Chae, Yoon-Jee; Lee, Kyeong-Ryoon (2023-03-14). “Physiologically Based Pharmacokinetic Modelling to Predict Pharmacokinetics of Enavogliflozin, a Sodium-Dependent Glucose Transporter 2 Inhibitor, in Humans”Pharmaceutics15 (3): 942. doi:10.3390/pharmaceutics15030942ISSN 1999-4923PMC 10058973PMID 36986803.
  8.  “Daewoong Pharmaceutical’s diabetes drug wins approval in Ecuador – 매일경제 영문뉴스 펄스(Pulse)”Pulse (in Korean). 2024. Retrieved 2025-04-04.
Enavogliflozin
Clinical data
Other names DWP16001
ATC code
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C24H27ClO6
Molar mass 446.92 g·mol−1
3D model (JSmol)

/////// DWJ-304, Daewoong Pharmaceutical, DWP-16001, SGLT-2 inhibitor, type 2 diabetes, KOREA, Enavogliflozin, GCC 5694A

ClC1=C2C(CCO2)=C([C@@H]3O[C@H](CO)[C@@H](O)[C@H](O)[C@H]3O)C=C1CC4=CC=C(C5CC5)C=C4

SYN

Synthesis 2024, 56, 906–943

Enavogliflozin (17) (DWP-16001) was developed by Green Cross Corp. and Daewoong Pharmaceuticals Co. Ltd.with the intention of addressing type 2 diabetes. Its chemical name is (2S,3R,4R,5S,6R)-2-(7-chloro-6-(4-cyclopropylbenzyl)-2,3-dihydrobenzofuran-4-yl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol. In clinical trials, this medication exhibited remarkable efficacy as both an antidiabetic agent and an SGLT inhibitor. The initial synthetic pathway for producing envogliflozin (17), in addition to other C-aryl-glycoside-type derivatives, was documented in the United States, specifically through patent application
number US9034921B2.75 Enavogliflozin (17) belongs to the category of C-aryl glycoside derivatives and its synthesis encompasses 19 steps, ultimately achieving an overall yield of 12%.
The process starts with the preparation of aglycone key intermediate 290 (Scheme 50), and involves a series chemical transformations starting from the commercially available 3-methoxy-2-nitrobenzoic acid (275). Following the successful synthesis of the aglycone intermediate 290, the process was advanced by employing n-BuLi for lithium–halogen exchange on 290 and subsequent addition of lithiated 290 to O-silyl-protected compound 22 at –78 °C (Scheme51). This sequence yielded the TMS-protected lactol inter
mediate 291 in quantitative yield. By subjecting this intermediate to treatment with MsOH/MeOH, the desired product 292 was obtained, achieving a 2-step overall yield of 88%. During these reactions, the O-silyl groups of the C-glucoside 292 were cleaved. Furthermore, the reduction of intermediate 292 was executed in the presence of triethylsilane and boron trifluoride–diethyl etherate complex, lead
ing to the formation of desmethoxy intermediate 293 in 100% yield. Subsequent acetylation of the four hydroxy groups was performed using acetic anhydride and a catalytic quantity of DMAP, producing the tetra-acetyl intermediate 294 in a yield of 59%. Ultimately, removal of the acetyl groups was achieved in the presence of NaOH, culminating in the generation of the final product, enovagliflozin (17),
with a 100% yield (Scheme 51). This synthetic procedure is plagued by significant limitations, including an extended route to obtain the aglycone intermediate 290, the application of protection and deprotection chemistry, and the necessity of cryogenic conditions to obtain the lactol intermediate 291. According to the reaction sequences given in Schemes 50 and 51, the overall yield of the final compound is calculated to be 12% via a total of 19 steps. In another approach, enavogliflozin (17) was synthesized in 27 steps with an overall yield of 0.5% (Schemes 52and 53).76 Initially, the synthesis of O-allyl aglycone intermediate 299 was achieved in nine steps starting from 3 methoxy-2-nitrobenzoic acid (275) (see Scheme 51). Methylation of compound 275 using methyl iodide and potassium carbonate in DMF afforded methyl ester 276 in 98% yield. Reduction of the NO2 group of 276 was carried out using Pd/C to afford aryl amine 277 in excellent yield. Next, bromination of 277 was facilitated by using NBS in DMF and
ethyl acetate to afford the brominated compound 278 in 86% yield. Chlorination was then carried out on intermediate 278 under diazotization reaction conditions to afford 279. The ester group of 279 was hydrolyzed under basic conditions to afford the aryl carboxylic acid 295. Subsequently, the preparation of acid chloride 296 from 295 was achieved using oxalyl chloride and a catalytic amount of DMF, which was coupled with benzene under Friedel Crafts acylation conditions to give the aryl benzophenone
intermediate 297. Reduction of the keto group of 297 was achieved by using triethylsilane and TFA/TfOH. Finally, the O-allyl aglycone intermediate 299 was obtained, when in termediate 298 was subjected to O-allylation using allylbromide and potassium carbonate in acetone. Next, the O-allyl aglycone intermediate 299 was subjected to a Br/Li exchange reaction using n-BuLi and addition of the obtained lithiated compound was carried out on gluconolactone 22 to afford the lactol intermediate 300 in quantitative yield (Scheme 53). The hydroxy group of 300 was methylated using methanesulfonic acid in methanol to
give 301 in 88% yield over two steps from 299. Demethoxylation and TMS cleavage was carried out on 301 using triethylsilane and BF3·Et2O to furnish intermediate 302. This hydroxy intermediate was protected using acetic anhydride and DMAP to afford the acetylated compound 303, deprotection of which with sodium methoxide gave product 304 in 100% yield. Benzylation of 304 was carried out using BnBr and NaH to give tetra-O-benzylated compound 305. Next, the O-allyl group of 305 was reduced to give alcohol 306 in 95% yield. Bromination followed by O-alkylation of the intermediate 306 then furnished compound 308. The hydroxy group of 308 was replaced by a chlorine atom under treatment with CCl4 and PPh3 to afford 309. Using n-BuLi, intramolecular cyclization was carried out to give com
pound 310 in 69% yield and subsequent debenzylation by treatment with Pd/C and H2 afforded compound 311. Again, acetylation of the free hydroxy groups of 311 was achieved using acetic anhydride and DMAP to give the O-acetylated intermediate 312. A Friedel–Crafts reaction on the aryl moiety of intermediate 312 gave acylated product 313 in 93% yield. The keto group of 313 was reduced using sodium borohydride in methanol to give the 314, which was further reduced under acidic conditions to give the alkene intermediate 315. The Simmons–Smith cyclopropanation was achieved on the alkene intermediate 315 to give compound 294 in 60% yield. Finally, the acetyl groups were removed
from the sugar moiety of 294 to give enavogliflozin (17) in 47% yield. This synthetic route also contains major disadvantages in terms of the use of protection/deprotection strategies, a lengthy linear process and employs several harmful reagents.

(75) Choi, S.; Song, K. S.; Lee, S. H.; Kim, M. J.; Seo H. J.; Park, E.-J.; Kong, Y.; Park, S. O.; Kang, H.; Jung, M. E.; Lee, K.; Kim, H. J.; Lee, J. S.; Lee, M. W.; Kim, M.-S.; Hong, D. H.; Kang, M. US9034921B2, 2015.
(76) Yoon, H.-K.; Park, S.-H.; Yoon, J.-S.; Choi, S.; Seo, H. J.; Park, E.-J.; Kong, Y.; Song, K.-S.; Kim, M. J.; Park S. O. WO2017217792A1, 2017.

.

,

LYS 228


2D chemical structure of 1810051-96-7

LYS228

BOS-228
LYS-228

Molecular Formula, C16-H18-N6-O10-S2

Molecular Weight, 518.4783

(3S,4R)-3-((Z)-2-(2-Ammoniothiazol-4-yl)-2-((1-carboxycyclopropoxy)imino)acetamido)-2-oxo-4-((2-oxooxazolidin-3-yl)methyl)azetidine-1-sulfonate

RN: 1810051-96-7
UNII: 29H7N9XI1B

Unii-005B24W9YP.png

UNII-005B24W9YP

005B24W9YP

Lys-228 trihydrate

2091840-43-4

Yclopropanecarboxylic acid, 1-(((Z)-(1-(2-amino-4-thiazolyl)-2-oxo-2-(((3S,4R)-2-oxo-4-((2-oxo-3-oxazolidinyl)methyl)-1-sulfo-3-azetidinyl)amino)ethylidene)amino)oxy)-, hydrate (1:3)

1-[(Z)-[1-(2-amino-1,3-thiazol-4-yl)-2-oxo-2-[[(3S,4R)-2-oxo-4-[(2-oxo-1,3-oxazolidin-3-yl)methyl]-1-sulfoazetidin-3-yl]amino]ethylidene]amino]oxycyclopropane-1-carboxylic acid;trihydrate

BOS-228 (LYS-228) is a monobactam discovered at Novartis and currently in phase II clinical development at Boston Pharmaceuticals for the treatment of complicated urinary tract infection and complicated intraabdominal infections in adult patients.

The compound has been granted fast track and Qualified Infectious Disease Product (QIDP) designation from the FDA.

In October 2018, Novartis licensed to Boston Pharmaceuticals worldwide rights to the product.

Paper

https://pubs.acs.org/doi/10.1021/acs.oprd.9b00330

Patent

US 20150266867

PATENT

WO 2017050218

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017050218&tab=FULLTEXT

Compound X: 1- ( ( (Z) – (1- (2-aminothiazol-4-yl) -2-oxo-2- ( ( (3S, 4R) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) -1-sulfoazetidin-3-yl) amino) ethylidene) amino) oxy) cyclopropanecarboxylic acid.

[0126]
Step 1: Benzhydryl 1- ( ( (Z) – (1- (2- ( (tert-butoxycarbonyl) amino) thiazol-4-yl) -2-oxo-2- ( ( (3S, 4R) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) azetidin-3-yl) amino) ethylidene) amino) oxy) cyclopropanecarboxylate. To a solution of (Z) -2- ( (1- ( (benzhydryloxy) carbonyl) cyclopropoxy) imino) -2- (2- ( (tert-butoxycarbonyl) amino) thiazol-4-yl) acetic acid (854 mg, 1.59 mmol) prepared according to published patent application US2011/0190254, Intermediate B (324 mg, 1.75 mmol) and HATU (785 mg, 2.07 mmol) in DMF (7.9 mL) , DIPEA was added (832 μL, 4.77 mmol) . After 1 h of stirring, it was poured into water and extracted with EtOAc. Brine was added to the aqueous layer, and it was further extracted with ethyl acetate (EtOAc) (3x) . The combined organic layers were dried over Na 2SO 4 and concentrated in vacuo. The crude residue was purified via silica gel chromatography (0-10%MeOH-DCM) to afford the title compound (1.09 g, 97%) as a beige foam. LCMS: R t = 0.97 min, m/z =705.3 (M+1) Method 2m_acidic.

[0127]
Instead of HATU, a variety of other coupling reagents can be used, such as any of the typical carbodiimides, or CDMT (2-chloro-4, 6-dimethoxy-1, 3, 5-triazine) and N-methylmorpholine to form the amide bond generated in Step 1.

[0128]
Step 2: (3S, 4R) -3- ( (Z) -2- ( (1- ( (benzhydryloxy) carbonyl) cyclopropoxy) imino) -2- (2- ( (tert-butoxycarbonyl) amino) thiazol-4-yl) acetamido) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) azetidine-1-sulfonic acid. Benzhydryl 1- ( ( (Z) – (1- (2- ( (tert-butoxycarbonyl) amino) thiazol-4-yl) -2-oxo-2- ( ( (3S, 4R) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) azetidin-3-yl) amino) ethylidene) amino) oxy) cyclopropanecarboxylate (1.00 g, 1.42 mmol) in DMF (7.0 mL) at 0 ℃ was treated with SO 3·DMF (448 mg, 2.84 mmol) . After 2 h of stirring at rt, the solution was poured into ice-cold brine and extracted with EtOAc (3x) . The combined organic layers were dried over Na 2SO 4 and concentrated in vacuo, affording the title compound (assumed quantitative) as a white solid. LCMS: Rt =0.90 min, m/z = 785.2 (M+1) Method 2m_acidic.

[0129]
Step 3: 1- ( ( (Z) – (1- (2-aminothiazol-4-yl) -2-oxo-2- ( ( (3S, 4R) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) -1-sulfoazetidin-3-yl) amino) ethylidene) amino) oxy) cyclopropanecarboxylic acid.

[0130]

[0131]
To a solution of (3S, 4R) -3- ( (Z) -2- ( (1- ( (benzhydryloxy) carbonyl) cyclopropoxy) imino) -2- (2- ( (tert-butoxycarbonyl) amino) thiazol-4-yl) acetamido) -2-oxo-4- ( (2-oxooxazolidin-3-yl) methyl) azetidine-1-sulfonic acid (1.10 g, 1.40 mmol) in DCM (1.5 mL) at 0℃, TFA (5.39 mL, 70.0 mmol) was added, and after 10 minutes, the ice bath was removed. Additional TFA (3.24 mL, 42.0 mmol) was added after 1 hr at rt and the solution was diluted with DCM and concentrated in vacuo after an additional 30 min. Optionally, anisole may be added to the TFA reaction to help reduce by-product formation, which may increase the yield of desired product in this step. The crude residue was purified by reverse phase prep HPLC (XSelect CSH, 30 x 100 mm, 5 μm, C18 column; ACN-water with 0.1%formic acid modifier, 60 mL/min) , affording the title compound (178 mg, 23%) as a white powder. LCMS: R t = 0.30 min, m/z = 518.9 (M+1) Method 2m_acidic; 1H NMR (400 MHz, DMSO-d 6) δ 9.27 (d, J = 9.0 Hz, 1H) 6.92 (s, 1H) 5.23 (dd, J = 9.1, 5.7 Hz, 1H) 4.12-4.23 (m, 3H) 3.72-3.62 (m, 2H assumed; obscured by water) 3.61-3.52 (m, 1H assumed; obscured by water) 3.26 (dd, J = 14.5, 5.9 Hz, 1H) 1.36 (s, 4H) . 1H NMR (400 MHz, D 2O) δ 7.23 (s, 1H) , 5.48 (d, J = 5.8 Hz, 1H) , 4.71-4.65 (m, 1H) , 4.44 (t, J = 8.2 Hz, 2H) , 3.89-3.73 (m, 3H) , 3.54 (dd, J = 14.9, 4.9 Hz, 1H) , 1.65-1.56 (m, 2H) , 1.56-1.46 (m, 2H) . The product of this process is amorphous. Compound X can be crystallized from acetone, ethanol, citrate buffer at pH 3 (50 mM) , or acetate buffer at pH 4.5 (50 mM) , in addition to solvents discussed below.

PAPER

Bioorganic & Medicinal Chemistry Letters (2018), 28(4), 748-755.

https://www.sciencedirect.com/science/article/pii/S0960894X18300064

PATENT

WO 2019026004

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019026004&tab=PCTDESCRIPTION

Over the past several decades, the frequency of antimicrobial resistance and its association with serious infectious diseases have increased at alarming rates. The increasing prevalence of resistance among nosocomial pathogens is particularly disconcerting. Of the over 2 million (hospital-acquired) infections occurring each year in the United States, 50 to 60% are caused by antimicrobial-resistant strains of bacteria. The high rate of resistance to commonly used antibacterial agents increases the morbidity, mortality, and costs associated with nosocomial infections. In the United States, nosocomial infections are thought to contribute to or cause more than 77,000 deaths per year and cost approximately $5 to $10 billion annually.

Important causes of Gram-negative resistance include extended-spectrum 13- lactamases (ESBLs), serine carbapenemases (KPCs) and metallo-13-lactamases (for example NDM-1 ) in Klebsiella pneumoniae, Escherichia coli, and Proteus mirabilis, high-level third-generation cephalosporin (AmpC) 13-lactamase resistance among Enterobacter species and Citrobacter freundii, and multidrug-resistance genes observed in Pseudomonas, Acinetobacter, and Stenotrophomonas. The problem of antibacterial resistance is compounded by the existence of bacterial strains resistant to multiple antibacterials. For example, Klebsiella pneumonia harboring NDM-1 metallo-13- lactamase carries frequently additional serine-13-lactamases on the same plasmid that carries the NDM-1 .

Thus there is a need for new antibacterials, particularly antibacterial compounds that are effective against existing drug-resistant microbes, or are less susceptible to development of new bacterial resistance. Monobactam antibiotic, which is referred to herein as Compound X, is primarily effective against Gram-negative bacteria, including strains that show resistance to other monobactams.

The present invention relates to a process for the preparation of monobactam antibiotic Compound X and intermediates thereof.

More particularly, the present invention relates to a process for the preparation of Compound X

Compound X

also referred to as 1 -(((Z)-(1 -(2-aminothiazol-4-yl)-2-oxo-2-(((3S,4R)-2-oxo-4-((2-oxooxazolidin-3-yl)methyl)-1 -sulfoazetidin-3-yl)amino)ethylidene)amino)oxy)cyclopropanecarboxylic acid, or a salt thereof, or a solvate including hydrate thereof.

Patent application number PCT/US2015/02201 1 describes certain monobactam antibiotics. Compound X may be prepared using the method disclosed in PCT/US2015/02201 1 , in particular example 22, and in PCT/CN2016/099482.

A drawback from these processes is that they exhibit a large number of process steps and intermediate nitrogen protection/deprotection steps, reducing the overall yield and efficiency. Furthermore, these processes require several chromatographic purification steps to be carried out in course of the processes. We have found that the preparation of Compound X, as previously prepared on a manufacturing scale, possesses a number of disadvantages, in particular poor handling characteristics.

It would thus be beneficial to develop alternative or improved processes for the production of Compound X that do not suffer from some or all of these disadvantages.

Compound x Compound x

Scheme 1

Preparation of Compound X from Intermediates 22 and 2A

Scheme 3

Examples

The Following examples are merely illustrative of the present disclosure and they should not be considered as limiting the scope of the disclosure in any way, as these examples and other equivalents thereof will become apparent to those skilled in the art in the light of the present disclosure, and the accompanying claims.

Synthesis of Compound 8 (R = benzyl)

1 .50kg oxazolidin-2-one (7b) was charged into the reactor. 7.50kg THF was charged and the stirring started. The mixture was cooled to 10~20°C. 2.18kg potassium fert-butoxide was charged intol 2.00kg THF and stirred to dissolve.

The potassium fert-butoxide solution was added dropwise into the reactor while maintaining the temperature at 10-20 °C. The reaction was stirred for 1 ~2hrs at 10-20 °C after the addition. The solution of 2.36kg methyl-2-chloroacetate (7a) in 3.00kg of THF was added to the reactor while maintaining the temperature at 10-20 °C. The reaction mixture was stirred for 16-18 h at 20-25 °C. The IPC (in process control) showed completion of the reaction. The mixture was centrifuged and the wet cake was washed with 7.50kg THF. The filtrate was concentrated and the crude 7 was provided as reddish brown liquid, which was used for the next step without further purification,

1H NMR (400 MHz, CHLOROFORM- /) δ ppm 3.65 – 3.71 (m, 2 H) 3.74 (s, 3 H) 4.02 (s, 2 H) 4.34 – 4.45 (m, 2 H).

The dried reactor was exchanged with N2 three times. 3.71 kg LiHMDS solution in THF/Hep (1 M) and 1 .30kg THF were charged under nitrogen protection. The stirring was started and the solution was cooled to -70—60 °C. The solution of 0.71 kg benzyl acetate (6) in 5.20 kg THF was added dropwisely at -70— 60 °C, and the resulted mixture was stirred for 1 -1 .5 h after the addition. The solution of 0.65kg 7 in 3.90kg THF was added dropwise while maintaining the temperature at -70—60 °C, then stirred for 30-40 minutes. The reaction mixture was warmed to 20-25 °C and stirring was continued for 0.5-1 .0 h. IPC showed 6 was less than1 .0% (Otherwise, continue the reaction till IPC passes). The reaction mixture was poured into 13.65 kg aqueous citric acid below 10 °C. The mixture was stirred for 15-20 minutes after the addition. Phases were separated and the organic layer was collected. The aqueous layer was extracted with EA (6.50kg * 2). The organic layer was combined, washed by 6.50 kg 28% NaCI solution and dried with 0.65

kg anhydrous MgSC . The mixture was filtered and the wet cake was washed with 1 .30kg EA. The filtrate was concentrated under vacuum to provide crude 8. The crude 8 was stirred in 2.60 kg MTBE at 20-25 °C for 1 -1 .5 h. The mixture was cooled to 0-10 °C and stirred for 1 .5-2.0 h and filtered. The filter cake was washed with 0.65kg pre-cooled MTBE and dried under vacuum (<-0.096Mpa) at 20-25 °C for 12~16hrs till a constant weight to give 513 g of 8 as a white solid, Yield: 45%, HPLC purity 96.4%,1 H NMR (400 MHz, CHLOROFORM-c δ ppm 3.48 – 3.55 (m, 1 H) 3.56 – 3.63 (m, 2 H) 3.66 – 3.74 (m, 1 H) 4.17 – 4.26 (m, 2 H) 4.31 – 4.44 (m, 2H) 5.12 – 5.24 (m, 2 H) 7.30 – 7.44 (m, 5 H).

Synthesis of Compound 9 (R = benzyl)

The dried reactor was charged with 3.75kg HOAc and 1 .50 kg 8. The stirring was started and the reaction mixture was cooled to 0-5 °C. 3.53kg aqueous NaN02 was added dropwise at 0-10 °C, and the reaction mixture was stirred for 15-30 minutes after the addition. IPC showed 8 was less than 0.2%. The reaction mixture was treated with 7.50kg EA and 7.50 kg water. Phases were separated and the organic layer was collected. The aqueous layer was extracted with EA (7.50kg * 2). The organic layers were combined, washed with 7.50 kg 28% NaCI solution, and concentrated under vacuum to provide crude 9. The crude 9 was slurried with 5.25 kg water at 10-20 °C for 3~4hrs, and filtered. The wet cake was washed with 1 .50kg water. The solid was dried under vacuum (<-0.096 Mpa) at 45-50 °C for 5-6 h till a constant weight to give 1 .44 Kg of 9, yield: 86.9%, HPLC purity 92.9%,1H NMR (400 MHz, CHLOROFORM- /) δ ppm 3.60 – 3.76 (m, 2 H) 4.44 (t, J=8.07 Hz, 2 H) 4.60 (s, 2 H) 5.25 – 5.41 (m, 2 H) 7.30 – 7.43 (m, 5 H) 1 1 .62 (br s, 1 H).

Synthesis of Compound 9a (R = benzyl)

9

The dried reactor was charged with 0.58 kg Zn, 4.72kg (Βο Ο, 6.00 kg water, 1 .20 kg NH4CI and 6.00kg THF. The reaction mixture was stirred and heated to 50-55 °C. The solution of 0.60 kg 9 in 4.20kg THF

was added dropwisely while maintaining the temperature at 50-55 °C. The reaction mixture was stirred for 0.5-1 .Ohrs after the addition. IPC showed 9 was less than 0.1 %. The reaction mixture was treated withl .50 kg ethyl acetate and stirred for 15-20 minutes. Phase was separated and the water layer was extracted by1 .50 kg ethyl acetate. The organic layers were combined, washed with 6.00 kg 28% NaCI solution and concentrated under vacuum to provide crude 9a. The crude 9a was stirred with 3.60kg*2 n-heptane to remove excess (Βο Ο. The residue was purified by silica gel chromatography column eluted with ethyl acetate: Heptane= 1 :1 to provide crude 9a solution. The solution was concentrated under reduced pressure to obtain crude 9a. The crude 9a was slurried with 1 .80 kg MTBE for 2.0-3. Ohrs, filtered, and the wet cake was washed with MTBE. The solid was dried under vacuum (<-0.096 Mpa) at 50-55 °C for 16-18 h till a constant weight to give 392 g of 9a as a white solid, Yield: 51 %, HPLC purity 98.1 %,1H NMR (400 MHz, DMSO-cfe) δ ppm 1.17 – 1 .57 (m, 9 H) 3.39 – 3.61 (m, 2 H) 4.20 – 4.45 (m, 3 H) 5.10 – 5.32 (m, 3 H) 5.75 (s, 1 H) 7.38 (br s, 5 H) 7.75 – 7.99 (m, 1 H).

Synthesis of compound (VII) (R = benzyl, X = CI)

9a VII

The dried reactor was charged with 13.0kg HCI in IPA and the stirring was started. 1 .33 kg 9a was charged in portions at 20-25 °C. The mixture was stirred at 20-25 °C for 3-4 h. IPC showed 9a was less than 0.1 %. The reaction solution was concentrated under vacuum 40-45 °C. The residue was treated with 21 .58kg MTBE at 20-25 °C for 3-4 h. The mixture was filtered and the wet cake was washed with 2.60kg MTBE. The solid was dried under vacuum (<-0.096 Mpa) at 45-50 °C for 5-6 h till a constant weight to give 1 .045 Kg of compound VII (R = benzyl, X = CI) as a yellow solid, Yield: 93.7%, HPLC purity 99.2%,1 H NMR (400 MHz, DMSO-cfe) δ ppm 3.16 – 3.74 (m, 3 H) 4.10 – 4.35 (m, 4 H) 5.09 – 5.39 (m, 2 H) 7.27 – 7.60 (m, 5 H) 8.72 (br s, 2 H).

Synthesis of compound (Vile) (R = benzyl)

VII Vile

To an autoclave (3L) were added VII (R = benzyl, X = CI) (100 g, 304.2 mmol, 1 .0 equiv.), DCM (2650 g, 26.5 equiv., w/w) and (S-BINAP)RuCl2 (2.4 g, 3.04 mmol, 0.01 equiv.), successively. Air in the autoclave was replaced with N2 5 times. N2 in the autoclave was was replaced with H2 5 times. The solution was stirred with 250-260 r/min and H2 (2.1 ±0.1 MPa) at 40±5°C for 24 h. The reaction mixture was filtered, and the filter cake was washed with DCM (400 g, 4.0 equiv., w/w). The filter cake was slurried with IPA (785 g, 7.85 equiv., w/w) and H2O (40 g, 0.4 equiv., w/w) overnight (18-20 h). The mixture was filtered. The filter cake was washed with IPA (200 g, 2.0 equiv., w/w) and dried at 45±5°C overnight (18-20 h). Vile (R = benzyl) was obtained as off-white solid, 80.4 g, 79.9% yield, 95.5% purity, 97.6% de, >99.5% ee. 1H NMR (400 MHz, DMSO-cfe) δ ppm 3.34-3.38 (m, 2 H) 3.50-3.52 (m, 1 H) 3.60-3.62 (m, 1 H) 4.18-4.24 (m, 4 H) 5.23 (s, 2H) 6.16 (s, 1 H) 7.32 (m, 5H) 8.74 (s, 1 H).

Alternative synthesis of compound 9a (R = benzyl)

5b

Mg(OtBu)2

To a flask was added 5a (1 .88 g, 12.93 mmol), THF (40 mL), and CDI (2.20 g, 13.58 mmol) at 25 °C. The mixture was stirred for 3 h. To the reaction mixture was added 5b (2.00 g, 6.47 mmol), and Mg(OfBu)2 (2.21 g, 12.93 mmol). The reaction mixture was stirred at 25 °C for 24 h. The reaction mixture was concentrated under vacuum to remove most of the THF solvent. To the concentrated solution was added MTBE (40 mL), followed by addition of an aqueous solution of HCI (1 M, 60mL) to adjust to pH = 2-3. Two phases were separated, and the water phase was extracted with MTBE (20 mL). The combined organic phase was washed with aqueous NaHCC (5%, 50 mL) and brine (20%, 40 mL). The organic phase was concentrated to a weight of -19 g, and a lot of white solid was obtained in the concentration process. The suspension was cooled to 0 °C, and filtered. The filter cake was washed with cold MTBE (5 mL) and dried under vacuum to obtain product 9a (1 .6g, 63% yield).

Synthesis of compound (Vile) (R = benzyl, PG = Cbz)

Vile Vile

To a flask (5 L) were added Vile (R = benzyl) (140 g, 423.2 mmol, LOequiv.), H20 (1273 g, 9.09 equiv., w/w) and toluene (2206 g, 15.76 equiv., w/w). The solution was stirred and cooled to 0-5 °C with ice bath. Then NaHCOa (78.4 g, 933 mmol, 2.22 equiv.) was added and CbzCI (89.6 g, 527 mmol, 1 .24 equiv.) was dropped into the stirring solution, respectively. The solution was stirred at 30±5 °C overnight (18-20 h). Heptane (3612 g, 25.8 equiv., w/w) was added dropwise to the stirring solution over 1 h at 20-30 °C. The mixture was filtered. The filter cake was washed with heptane (280 g, 2.00 equiv., w/w) and MTBE (377 g, 2.69 equiv., w/w), respectively. The filter cake was dried at 45±5°C overnight (18-20 h). Vile (R = benzyl, PG = Cbz) was obtained as an off-white solid, 169.4 g, 93% yield, 96.7% purity, 98% de, >99.5% ee, 1 H NMR (400 MHz, DMSO-cfe) δ ppm 3.23-3.24 (m, 1 H) 3.30 (m, 1 H) 3.51 -3.55 (m, 2 H) 3.99 (s, 1 H) 4.17-4.21 (m, 3 H) 5.02-5.03 (m, 2H) 5.12 (s, 2H) 5.46-5.48 (d, 1 H) 7.33-7.36 (m, 10H) 7.75-7.73 (d, 1 H).

Synthesis of compound (IV) (PG = Cbz)

Vile IV

Vile (R = benzyl) (220 g, 513.5 mmol, 1 .0 equiv.) was dissolved in THF (1464g, 6.65 equiv., w/w). The solution was filtered. The filter cake was washed with THF (488g, 2.22 equiv., w/w). The filtrate (Vile) was collected. To an autoclave (3L) were added the filtrate (Vile). The reactor was cooled down to -75 – -65 °C with dry-ice/EtOH bath, and bubbled with NH3 for not less than 4 h. Then the solution was stirred at 25±5 °C with NH3 (0.5-0.6 MPa) for 24 h. The autoclave was deflated to release NH3. The reaction solution was concentrated with a rotary evaporator to remove THF until the residue was around 440 g. The residue was slurried with EA (2200 g, 10 equiv., w/w) at 70±2 °C, then cooled to 25±5 °C and stirred for 16-18 h. The mixture was filtered. The filter cake was washed with EA (440 g). The filter cake was slurried with EA (1320 g, 6.00 equiv. w/w), and the temperature was raised to 70±2 °C, then cooled to 25±5 °C and stirred for 16-20 h. The mixture was filtered. The filter cake was washed with EA, and dried at 50±5 °C overnight (18-20 h). IV (PG = Cbz) was obtained as off-white solid, 141 g, 81 .5% yield, 99.1 % purity, >99.5% assay, 1H NMR (400 MHz, DMSO-cfe) δ ppm 3.12 – 3.23 (m, 2 H) 3.31 (br s, 1 H) 3.56 (t, J=8.01 Hz, 2 H) 3.88 (quin, J=6.02 Hz, 1 H) 3.93 – 4.03 (m, 1 H) 4.20 (t, J=8.01 Hz, 2 H) 5.02 (s, 2 H) 5.27 (d, J=5.87 Hz, 1 H) 7.12 (s, 1 H) 7.22 – 7.45 (m, 5 H).

Synthesis of compound (III) (PG = Cbz, LG = S02CH3)

IV III

To a flask was added IV (PG = Cbz) (14.00 g, 41 .50 mmol, 1 .00 equiv), and dry 1 , 2-dimethoxyethane (300 mL) under N2. The mixture was stirred at -5°C ~ 0°C for 1 h to obtain a good suspension. MsCI (7.89 g, 68.89 mmol, 5.33 mL, 1 .66 eq) in 1 , 2-dimethoxyethane (20.00 mL) was added dropwise during 30 min, and Et3N (12.60 g, 124.50 mmol, 17.26 mL, 3.00 eq) in 1 , 2-dimethoxyethane (20.00 mL) was added dropwise during 30 min side to side. The reaction mixture was stirred for additional 5 min at -5°C ~ 0°C, and was quenched with water (6 mL). The reaction mixture was concentrated to remove DME. The solid was slurried in water (250 mL) and MTBE (125 mL) for 1 h. The solid was collected by filtration, and then slurried in water (250 mL) for 1 hr. The solid was collected by filtration, and washed with water (25 mL) to give white solid. The solid was slurried in EA (150 mL) and dried in vacuum at 60°C for 24 h to give III (PG = Cbz, LG = SO2CH3) (15.00 g, 36.1 1 mmol, 87.01 % yield), 1H NMR (400 MHz, DMSO-cfe) δ ppm 3.17 (s, 3 H) 3.26 (br d, J=15.04 Hz, 1 H) 3.47 – 3.57 (m, 1 H) 3.64 (br d, J=6.36 Hz, 2 H) 4.22 (br dd, J=17.79, 8.50 Hz, 2 H) 4.50 (br s, 1 H) 4.95 – 5.17 (m, 3 H) 7.21 – 7.56 (m, 5H) 7.43 (s, 1 H) 7.63 – 7.89 (m, 2 H).

Synthesis of compound II (PG = Cbz, LG = SO2CH3, M+ = NBu4+)

O OMs o CISO3H, 2-picoline – ° O ?yO

HN Bu4NHS04< NHCbz

“Cbz

III II

To a flask was added 2-picoline (1 1 .50 g, 12.23 mL) and DMF (10 mL). The solution was cooled to 5 SC, followed by slow addition of chlorosulfonic acid (7.20 g, 4.14 mL). The temperature was increased to 20 SC. Ill (PG = Cbz, LG = SO2CH3) (5.13 g, 12.35 mmol) was added to the reaction mixture. The reaction mixture was heated to 42 SC for 18h. IPC (in process control) showed complete conversion of starting material. The reaction was cooled to 20 SC and dropwise added to a solution of tetrabutylammonium hydrogen sulfate (4.6 g, 13.6 mmol) in the mixed solvents of dichloromethane (100 mL) and water (100 mL) at 5SC. The phases were separated and the water phase was extracted with dichloromethane (2*50mL). The combined organic phase was washed with water (5*100mL). The organic phase was concentrated to dryness and purified by column chromatography (dichloromethane/methanol = 15/1 v/v) to afford II (PG = Cbz, LG = SO2CH3, M+ = NBii4+) (8.4 g, 92.30%), 1 H NMR (400 MHz, CHLOROFORM-c/) δ ppm 0.99 (t, J=7.34 Hz, 12 H) 1 .36 – 1 .50 (m, 8 H) 1 .54 – 1 .76 (m, 8 H) 3.15 (br d, J=8.31 Hz, 2 H) 3.21 – 3.35 (m, 8 H) 3.47 (br dd, J=14.73, 7.27 Hz, 1 H) 3.54 – 3.65 (m, 1 H) 3.67 – 3.81 (m, 2 H) 4.17 – 4.32 (m, 1 H) 4.39 – 4.62 (m, 1 H) 4.74 (br s, 1 H) 5.1 1 (s, 3 H) 5.32 – 5.50 (m, 1 H) 6.47 (br s, 1 H) 7.29 – 7.47 (m, 5 H) 8.69 – 8.94 (m, 1 H).

Synthesis of compound (IA)

A solution of II (PG = Cbz, LG = SO2CH3, M+ = NBu4+) (4.0 g) in dichloromethane (38 mL) was pumped to tube A at rate of 2.0844 mL/min, and a solution of KHCO3 (3.0 g) in water (100 mL) was pumped to tube B at a rate of 1 .4156 mL/min side to side. These two streams were mixed in a cross-mixer then flowed to a tube coil that was placed in an oil bath at 100 °C. The residence time of the mixed stream in the coil was 2 min. The reaction mixture flowed through a back-pressure regulator that was set at ~ 7 bars, and was collected to a beaker. After completion of the collection, two phases was separated. The organic phase was concentrated to dryness. The residue was slurried in ethyl acetate (5 mL). The solid was filtered and the filter cake was dried to give IA (2.6 g, 75%),

1H NMR (400 MHz, CHLOROFORM-c/) δ ppm 1.00 (t, J=7.27 Hz, 12 H) 1 .42 (sxt, J=7.31 Hz, 8 H) 1 .62 (quin, J=7.83 Hz, 8 H) 3.13 – 3.39 (m, 8 H) 3.54 – 3.69 (m, 2 H) 3.81 (dd, J=14.98, 2.51 Hz, 1 H) 3.96 – 4.13 (m, 1 H) 4.22 – 4.47 (m, 3 H) 4.99 – 5.23 (m, 3 H) 6.42 (br d, J=9.29 Hz, 1 H) 7.26 – 7.44 (m, 5 H).

Synthesis of compound 2A

Step 1

To a stirring solution of compound 16b (2 g, 10.14mmol, 1 .0 eq) in DMF (20 ml_) was added CS2CO3 (5.29g, 16.22 mmol, 1 .6 eq), then the resulting solution was stirred at room temperature for 10mins, then compound 16a (5.27g, 20.28mmol, 2eq) was added dropwise to the mixture for 2 minutes, then the resulting solution was stirred for another 2 hours. TLC showed the starting material was consumed completely. The mixture was added with water (60mL) and extracted with MTBE (20mL*3). The combined organic layers were dried over anhydrous sodium sulfate and concentrated. The crude was slurried in heptane to give 1 .65 g 16 as a white solid (Yield: 57%), 1H NMR (400 MHz, DMSO-cfe) δ ppm 7.48-7.28 (m, 10 H), 5.00-4.96 (t, J=6.0 Hz, 1 H), 3.81 (s, 3H), 3.44-3.42 (m, 2H), 2.40-2.37 (m, 2H).

Compound 16 (1 g, 2.66mmol, 1 eq) was dissolved in THF (20mL) under Nitrogen, and cooled to -40 °C. NaHMDS (1 .6mL, 2.0M THF solution, 1 .2 eq) was added dropwise. The reaction was stirred for 1 h at -40 °C. HPLC indicated the reaction was finished. The reaction was quenched with 10% Citric acid, extracted with MTBE (25 ml_ x 2). The combined organic layers were washed with brine (30 ml_), dried with Na2S04, filtered and concentrated to give 17 as a yellow solid, which was used for the next step without purification (assay yield: 65%); 1H NMR (400 MHz, DMSO-cfe) δ ppm 7.27-7.13 (m, 10 H), 3.46 (s, 3H), 1 .21 -1 .17(dd, J=7.2, 10.4 Hz, 2H ); 1 .14-1 .1 1 (dd, J=7.2, 10.4 Hz, 2H).

Step 3

Compound 17 (100 mg) was dissolved in methanol (5 mL) and 2.0 M HCI IPAC solution (5 mL). The solution was heated at 45 °C for 3 days. HPLC indicated the reaction was finished. The reaction was cooled to room temperature and was diluted with 10 mL water. The reaction mixture was washed with MTBE (10 mL x 2), organic layer was discarded and the aqueous layer was concentrated to give compound 2A HCI (32 mg, 62% yield), 1 H NMR (400 MHz, DMSO-cfe) δ ppm 3.80-3.44 (br, 4H), 1 .56 (s, 2H), 1 .38 (s, 2H).

Step 4

To a solution of 2A HCI (0.70 g, 4.57 mmol) in methanol (5 mL) was added triethylamine (1 .26 mL, 9.14 mmol) at room temperature. The solution was stirred for 20 min, and the solvent was removed under vacuum. To the residue was added IPAC (10 mL) leading to precipitation. The solid was filtered, and the filtrate was concentrated to provide 2A (0.50g, 94% yield) containing ca. 6 wt% Et3N-HCI.

Synthesis of Compound X from compound of formula (I), (IA)

Compound x

To a flask was charged 21 (1 .00 g, 68.43 wt%, 2.50 mmol) and DMF (10 mL). The suspension was cooled to -20 °C, to which was added diphenylphosphinic chloride (0.52 mL, 2.75 mmol). The solution was stirred at -20 °C for 30 min, followed by addition of a mixed solution of (IA) (1 .52g, 3.00 mmol) and triethylamine (0.52 mL, 3.76 mmol) in DMF (2mL). The reaction mixture was stirred at 20 °C for 20 h, followed by addition of MTBE (20 mL). The reaction mixture was adjusted to pH = 2-3 using aqueous HCI solution (37%). To the mixture was added isopropanol (100 mL). The resulting mixture was stirred for 4 h to obtain a suspension. The suspension was filtered and the filter cake was dried under vacuum to afford crude 22 (1 .17 g). The crude 22 was slurried in a combined solvent of THF/H2O (= 12 mL / 3mL), and filtered to afford 22 (0.744 g, 75 wt% by Q-NMR, 53.3% yield). 1H NMR (400 MHz, DMSO-cfe) δ ppm 3.47 – 3.55 (m, 2 H) 3.59 – 3.63 (m, 2 H) 4.13 – 4.21 (m, 3 H ) 5.05 (dd, J=8.8, 5.6 Hz, 1 H) 8.22 (s, 1 H) 9.73 (d, J=8.7 Hz, 1 H).

To a suspension of 22 (580 mg, 75 wt%, 1 .037 mmol) in DMAC (1 .5 mL) was added 2A (214.3 mg, 85 wt%, 1 .556 mmol). The reaction was stirred at 25 °C for 3 days, and in process control showed 22, Compound X = 4/96, and Z/E = 91 /9. the mixture was slowly added into 15ml acetone to precipitate yellowish solid. The reaction mixture was filtered to afford Compound X (0.7 g, 34 wt% by QNMR, 44% yield).

Synthesis of compound 3 (R2 = CH(Ph)2)

R2 = CH(Ph)2

2-(2-aminothiazol-4-yl)-2-oxoacetic acid (Y) (10.00 g, 47.93 mmol) and compound W (R2 = CH(Ph)2) (13.31 g, 46.98 mmol) were suspended in DMAC (40 mL), followed by addition of triethylamine (5.01 mL, 35.95 mmol). The reaction mixture was stirred at 20 °C for 5 h. HPLC showed completion of the reaction, and Z/E

= 97/3. To the reaction mixture was added water (120 mL) with stirring. The mixture was stirred for 20 min to obtain a suspension. The suspension was filtered and the filter cake was washed with water (50 mL).

The filter cake was slurried in a combined solvent of THF/ethyl acetate (50 mL / 50 mL) at 60 °C and cooled to 20 °C. The solid was filtered and dried at 50 °C for 3 h to get 3 (R2 = CH(Ph)2) (19.5 g, 88% yield). 1H

NMR (400 MHz, DMSO-cfe) δ ppm 1.37 -1 .42 (m, 2 H) 1 .44 – 1 .49 (m, 2 H) 6.87 (s, 1 H) 6.94 (s, 1 H) 7.22

– 7.30 (m, 6 H) 7.45 – 7.49 (m, 4 H).

Alternative Synthesis of Compound X from compound of formula (I), (IA)

Compound x

IA (40.14 g, 62.63 mmol) was dissolved in methanol (200 ml_), followed by addition of Pd/C (10%, 1 .1 g). The reaction mixture was maintained under hydrogen atmosphere (1 -2 bar) at 20 °C for 24 h. In process control showed completion of the reaction. The reaction mixture was filtered. The filtrate was concentrated to give an oil of IB (M+ = NBu4+) (58.20 g, 55 wt% by Q-NMR, 100% yield). 1 H NMR (400 MHz, DMSO-cfe) δ ppm 0.93 (t, J=7.3 Hz, 12 H) 1 .23 – 1 .36 (m, 8 H) 1 .57 (m, 8 H) 2.99 – 3.28 (m, 8 H) 3.37 (dd, J=14.3, 7.5 Hz, 1 H) 3.65 – 3.70 (m, 3 H) 3.84 – 3.88 (m, 1 H) 4.08 (d, J=5.6 Hz, 1 H) 4.18 – 4.22 (m, 2 H).

3 (R2 = CH(Ph)2) (0.95 g, 2.17 mmol) was dissolved in THF (20 ml_). To the solution was added /V-methyl morpholine (0.77 g, 7.60 mmol) and 2-chloro-4,6-dimethoxy-1 ,3,5-triazine (0.57 g, 3.26 mmol). The reaction mixture was stirred at 20 °C for 1 h followed by addition of IB (M+ = NBu +) (2.70 g, 48.98 wt%, 2.61 mmol). The reaction was stirred at 20 °C for 5 h. In process control showed completion of the reaction. To the reaction mixture was added ethyl acetate (20 ml_). The organic phase was washed with brine (10 ml_). Solvent was removed. Acetone (40ml) was added to dissolve residue. TFA (1 .24 g, 10.86 mmol) dissolved in acetone (3 ml) was added slowly. The white solid was filtered and washed by acetone (10 ml) two times. Dried at 40 °C for 5h to get compound 4 (R2 = CH(Ph)2). 1 H NMR (400 MHz, DMSO-cfe) δ ppm 1 .49 – 1 .55 (m, 4 H) 3.27 (dd, J=14.4, 6.2 Hz, 1 H) 3.49 – 3.65 (m, 2 H) 3.71 (dd, J=14.4, 6.2 Hz, 1 H) 4.04 – 4.10 (m, 1 H) 4.07 (dd, J=16.0, 8.6 Hz, 1 H) 4.17 (dd, J=1 1 .8, 6.0 Hz, 1 H) 5.28 (dd, J=9.0, 5.7 Hz, 1 H) 6.88 (s, 1 H) 7.03 (s, 1 H) 7.18 – 7.32 (m, 6 H) 7.43 (m, 4 H) 9.45 (d, J=9.0 Hz, 1 H).

Crude 4 (R2 = CH(Ph)2) (2.13 g) was dissolved in dichloromethane (20 ml_). The solution was cooled to 0 °C. To the solution was added anisole (0.68 ml_, 6.24 mmol) and trifluoroacetic acid (2.16 ml_, 28.08 mmol). The reaction was warmed to 20 °C, and stirred for 15 h. In process control showed completion of the

reaction. The aqueous phase was separated and added to acetone (40 mL) to obtain a suspension. The suspension was filtered to afford Compound X (0.98 g, 54.5% yield over two steps). 1 H NMR (400 MHz, DMSO-c/e) δ ppm 1.40 (m, 4 H) 3.26 (dd, J=14.4, 6.0 Hz, 1 H) 3.54 – 3.69 (m, 3 H) 4.14 – 4.21 (m, 3 H) 5.25 (dd, J= 8.9, 5.7 Hz, 1 H) 7.02 (s, 1 H) 9.38 (d, J=9.0 Hz, 1 H).

REF

Synthesis and optimization of novel monobactams with activity against carbapenem-resistant Enterobacteriaceae – Identification of LYS228
57th Intersci Conf Antimicrob Agents Chemother (ICAAC) (June 1-5, New Orleans) 2017, Abst SATURDAY-297

//////////////LYS228, LYS 228, BOS-228, LYS-228, monobactam, Novartis, phase II,  Boston Pharmaceuticals, complicated urinary tract infection, complicated intraabdominal infections,  fast track, Qualified Infectious Disease Product, QIDP,

Nc1nc(cs1)\C(=N\OC2(CC2)C(=O)O)\C(=O)N[C@H]3[C@@H](CN4CCOC4=O)N(C3=O)S(=O)(=O)O

ADX-103


2-(5-Amino-2-phenyl-1,3-benzoxazol-6-yl)propan-2-ol.png

ADX-103

CAS 916056-81-0

Preclinical, Antiinflammatory Ophthalmic Agents, Diabetic Retinopathy,

Agents for Ophthalmic Drugs
MF C16 H16 N2 O2

5-Amino-α,α-dimethyl-2-phenyl-6-benzoxazolemethanol

MW 268.31
6-Benzoxazolemethanol, 5-amino-α,α-dimethyl-2-phenyl-

Aldeyra Therapeutics Inc
ADX-103 , an aldehyde trap being investigated by Aldeyra for the treatment of dry eye syndrome; in May 2018, preclinical data were presented at 2018 ARVO Meeting in Honolulu, HI. Aldeyra, in collaboration with an undisclosed company, is also investigating an anti-inflammatory agent for treating ocular inflammation.

PATENT

WO-2020033344

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020033344&tab=PCTDESCRIPTION&_cid=P21-K6SRJF-10276-1

Novel crystalline forms of a specific benzoxazole and it’s salts, process for their preparation, and compositions comprising them are claimed, useful for treating dry eye, inflammation and diabetes, through action as an aldehyde scavenger.

It has now been found that compounds of the present invention, and compositions thereof, are useful for treating, preventing, and/or reducing a risk of a disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis. In general, salt forms or freebase forms, and pharmaceutically acceptable compositions thereof, are useful for treating or lessening the severity of a variety of diseases or disorders as described in detail herein. Such compounds are represented by the chemical structure below, denoted as compound A:

or a pharmaceutically acceptable salt thereof.

[0008] Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with toxic aldehydes. Such diseases, disorders, or conditions include those described herein.

[0009] Compounds provided by this invention are also useful for the study of certain aldehydes in biology and pathological phenomena.

Scheme 1 – Synthesis of Compound A

Step 1: Synthesis of Compound A2

[00549] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with methanol (10L). Compound A1 (2.0kg) was added, followed by further methanol to rinse (9L). The reaction mixture was warmed to Tjacket=40°C. Once temperature had stabilized, sulfuric acid (220 mL, 0.4eq.) was slowly added. Once addition was complete, agitation was maintained for 30 mins then the vessel was heated to Tjmt=62°C. Reaction progress was

monitored by LC-MS analysis of reaction mixture. The reaction does not go to completion but is deemed complete when no change is apparent in ratio of starting material : product.

[00550] The vessel contents were cooled to Tjmt=24°C and stirred 60 minutes before filtration under vacuum. The filter cake was air dried for 2 hours and the contents then dissolved in ethyl acetate (18L) which was then washed sequentially with saturated sodium bicarbonate (8L), water (8L) and brine (8L) before drying over sodium sulfate, filtration and evaporation in vacuo. Compound A2 (1.5kg, 68.1%) was obtained as a bright orange powder.

Step 2: Synthesis of Compound A3

[00551] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with /V,/V-dimethylformamide (16L). Compound A2 (1.5kg) was added and the brown reaction mixture set to cool to Tint<20oC. Once temperature had stabilized, A-bromosucci ni mi de (l.5kg, 1.1 eq.) was added portion wise, maintaining Tint<27°C. Once addition was complete, the reaction was allowed to stir until starting material content was <1% AUC (250nm) by LCMS analysis.

[00552] A secondary jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with ethyl acetate (16L) and deionized water (22L). The reaction mixture was vacuum transferred into this vessel and held at high agitation for not less than 30 minutes. The aqueous layer was discharged and the organic layer washed with saturated sodium chloride (2 x 8L) then dried over sodium sulfate before evaporation in vacuo to Compound A3 as a deep brown oil (2.lkg, 100.8%), suitable for use in following step without purification.

Step 3: Synthesis of Compound A4

[00553] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with dichloromethane (9L). Compound A3 (2.lkg) was added and the reaction mixture cooled to Tmt<l°C. A solution of Di-/er/-butyl dicarbonate (3.6kg, 2.2 eq.) in dichloromethane (0.5L) was added followed by a solution of A, A-di methyl ami nopyri di ne (92g, 0.1 eq.) in dichloromethane (0.5L). The resultant clear brown solution was stirred for 30 minutes whereupon pyridine (1.3L, 1.7 eq.) was dropwise added, maintaining Tint<5°C. Upon complete addition internal temperature was ramped from Tint=l°C to Tint=20°C over 18 hours.

[00554] The reaction mixture was sequentially washed with saturated sodium chloride (3 x 4.5L), 10 % w/v aqueous citric acid (2 x 4L), saturated sodium bicarbonate (4L), aqueous hydrochloric acid (1M, 4L), saturated sodium bicarbonate (4L) and saturated sodium chloride (4L) then dried over sodium sulfate and evaporated in vacuo with one azeotropic distillation with toluene (2L) to a very dark, heavy tar (3.4kg).

[00555] The isolated tar was mixed with absolute ethanol (3.1L) for 2 days whereupon it was filtered providing light cream colored, granular solids and a black mother liquor. The solids were washed with ice-cold ethanol (3 x 1L) and dried to constant mass. Compound A4 was obtained as off- white granules (1.7 kg, 50.2%).

Step 4: Synthesis of Compound AS

[00556] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with reagent alcohol (6.1 L) and Compound A4 (0.8kg), Tmt<20°C. Iron powder (0.5kg, 5.0 eq.) was added and the suspension stirred vigorously for 30 minutes. Acetic acid (glacial, 1.6L, 15.7 eq.) was added, maintaining Tint<30C.

[00557] Once LCMS confirmed complete consumption of starting material, ethyl acetate (10.2L) and water (10.2L) were added. Sodium bicarbonate (2.3kg, 15.9 eq.) was added portion wise and the layers separated once gas evolution had ceased. The aqueous layer was washed with ethyl acetate until LCMS indicated no further product was being extracted (8 x 2L) and the combined organic layers were sequentially washed with deionized water (6L) then saturated sodium chloride (6L) before drying over magnesium sulfate and evaporation in vacuo. Compound A5 was obtained as a light orange solid (0.7kg, 91.5%).

Step 5: Synthesis of Compound A6

[00558] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with dichloromethane (9L), Compound A5 (0.7kg), and the reaction mixture cooled to Tint 20°C. Benzoyl chloride (0.3L, 1.5 eq.) was added and the reaction stirred 15 minutes. N,N-dimethylaminopyridine (7g, 0.04 eq.) in dichloromethane (0.1L) was added and the reaction stirred 15 minutes. Pyridine (0.5L, 2.5 eq.) was dropwise added, maintaining Tint<20°C. Upon complete addition the reaction was stirred until LCMS indicated consumption of starting material.

[00559] The reaction mixture was washed with deionized water (11L) and the organic layer extracted sequentially with aqueous hydrochloric acid (1M, 3 x 5L), saturated aqueous sodium bicarbonate (11 L), saturated sodium chloride (11 L), dried over magnesium sulfate and evaporated in vacuo. Compound A6 was obtained as a cream colored solid, suitable for use without further purification (0.9kg, 100.7%).

Step 6: Synthesis of Compound A 7

[00560] A 30L jacketed vessel equipped with mechanical agitation, baffle and nitrogen bleed was charged with l,2-dimethoxy ethane (16L) and temperature set to Tint=2l°C. Compound A6 (0.9kg) was added and stirred to dissolution. Copper iodide (0.3kg, 1.0 eq.) was added and the mixture stirred 15 minutes. l, lO-phenanthroline (0.3kg, 1.2 eq.) was added and the mixture stirred 15 minutes. Cesium carbonate (l .5kg, 3.0 eq.) was added and the reaction was stirred for 15 minutes. The reaction temperature was ramped to Tint=80-85oC and maintained for 23 hours whereupon it was cooled to Tmt=20°C.

[00561] The reaction mixture was filtered through a celite pad, washing sequentially with deionized water (8L) and ethyl acetate (8L). The organic layer was extracted sequentially with deionized water (2 x 5L), saturated sodium chloride (4L), dried over sodium sulfate and evaporated in vacuo. Compound A7 was obtained as a brown solid, suitable for use without further purification (0.8kg, 104.1%).

Step 7: Synthesis of Compound A8

[00562] A 12L 3 -neck round bottom flask with nitrogen bleed and mechanical stirring was charged with a solution of Compound A7 (0.8kg) in dichloromethane (3.6L) and cooled to Tmt<5°C in an ice bath. Hydrochloric acid in dioxane (4M, 1 2L, 3.1 eq.) was added dropwise with vigorous stirring, maintaining Tmt<25°C. Once addition was complete, the reaction mixture was allowed to stir for 18 hours at Tint=20-25oC.

[00563] The reaction mixture was filtered and the filter cake washed with dichloromethane (2 x 1L) and dried to constant mass. The hydrochloride salt of Compound A8 was isolated as an off-white solid (0.5kg, 88.7%).

Step 8: Synthesis of Compound A

[00564] A 12L 3 -neck round bottom flask with nitrogen bleed and mechanical stirring was charged with a solution of Compound A8 (0.5kg) in tetrahydrofuran (4.8L) and cooled to Tint<-30°C in a dry-ice / acetone bath. Methylmagnesium bromide (3.4M in 2-methyltetrahydrofuran, 2.4L, 5.0eq.) was added slowly, maintaining Tmt<-lO°C. Once addition was complete, the reaction was allowed to warm to room temperature overnight.

[00565] Saturated aqueous ammonium chloride (2L) and ethyl acetate (2L) were added and the reaction mixture stirred for 30 minutes. The aqueous layer was extracted with further ethyl acetate (2 x 2L) and the combined organic layers washed with saturated sodium chloride (2L), dried over sodium sulfate and evaporated in vacuo to a dark heavy oil. The heavy oil was purified by column chromatography on silica gel, eluting with ethyl acetate : heptane 1 : 19 to 1 : 1. Pure Compound A was obtained after evaporation and drying as a brown powder (99.8 g, 23.0%).

Example 1 – Preparation of Free Base Forms A, B and C of Compound A

Compound A

Primary Polymorph Screen

[00566] Based on solubility screen results, a primary polymorph screen using an initial set of 24 solvents, as shown in Table 18, was performed as follows: A) To 24 x 20 mL vials, approximately 50 mg of the received ADX-103 was added; B) The solids were then slurried in 2 mL of the solvents and left placed in an incubator/shaker to temperature cycle between ambient and 40 °C in 4 hour cycles; C) After 72 hours temperature cycling, the mother liquors were removed from the vials and split evenly between 4 x 2 mL vials. The vials were then split between evaporation, crash cooling to 2 °C and -18 °C and anti-solvent addition; and D) Any solids

recovered were analysed by XRPD, any new patterns identified were also analysed by TG/DTA and PLM.

Table 18. Solvents Selected for Initial Primary Polymorph Screen

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018039197&tab=PCTDESCRIPTION&_cid=P21-K6SRNE-12791-1

WO2018039197 , as compound I-8.

PATENT

WO 2006127945

WO 2011072141

WO 2014116593

US 20150344447

WO 2020028820

////////////ADX-103, Preclinical, Antiinflammatory,  Ophthalmic Agents, Diabetic Retinopathy, Aldeyra Therapeutics Inc,

CC(C)(O)c1cc2oc(nc2cc1N)c3ccccc3

Follow New Drug Approvals on WordPress.com

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

Join 37.9K other subscribers

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

DISCLAIMER

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