Quantcast
Channel: DR ANTHONY MELVIN CRASTO Ph.D – New Drug Approvals
Viewing all 1646 articles
Browse latest View live

New Antiarthritic Drug Candidate S-2474

$
0
0

STR1

S-2474

(E)-(5)-(3,5-Di-tert-butyl-4-hydroxybenzylidene)-2-ethyl-1,2-isothiazolidine-1,1-dioxide

Shionogi Research Laboratories

cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO)

mp 135−137 °C.

S-2474,158089-95-3, 158089-96-4 ((Z)-isomer),C20-H31-N-O3-S,

E)-5-(3,5-Di-tert-butyl-4-hydroxybenzylidene)-2-ethylisothiazolidine 1,1-dioxide

  • Phenol, 2,6-bis(1,1-dimethylethyl)-4-[(2-ethyl-5-isothiazolidinylidene)methyl]-, S,S-dioxide, (E)-
  • 2,6-Bis(1,1-dimethylethyl)-4-[(E)-(2-ethyl-1,1-dioxido-5-isothiazolidinylidene)methyl]phenol
  • Phenol, 2,6-bis(1,1-dimethylethyl)-4-[(2-ethyl-1,1-dioxido-5-isothiazolidinylidene)methyl]-, (E)-

(E)-(5)-(3,5-Di-tert-butyl-4-hydroxybenzylidene)-2-ethyl-1,2-isothiazolidine-1,1-dioxide (S-2474, ), which was discovered at Shionogi Research Laboratories, shows potent inhibitory effects on both cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) and is anticipated to be promising as an antiarthritic drug

synthesis of novel γ-sultam derivatives containing the di-tert-butylphenol antioxidant moiety. Several compounds with lower alkyl groups at the 2-position of the γ-sultam skeleton showed potent inhibitory activities against PGE2 production via the COX pathway and LTB4 production via the 5-LO pathway, as well as production of IL-1 in in vitro assays. Extensive pharmacological characterizations revealed that 2-ethyl-γ-sultam derivative 10b displays multiple inhibition of COX, 5-LO, and IL-1 production similar to tenidap and also good selective COX-2 inhibition like NS-398 and celecoxib. It exerted excellent antiinflammatory activity without any ulcerogenic effects and was designated as S-2474 an agent having both NSAID and cytokine modulating properties. S-2474 is now being developed as a promising alternative antiarthritic drug candidate

SYNTHESIS

17th Symp Med Chem (Nov 19 1997 , Tsukuba), EP 0595546; JP 1994211819; US 5418230

The intermediate gamma-sultam (III) was prepared by condensation of 3-chloropropylsulfonyl chloride (I) with ethylamine, followed by cyclization of the resulting chloro sulfonamide (II) under basic conditions. Condensation of 3,5-di- tert-butyl-4- (methoxymethoxy) benzaldehyde (IV) with sultam (III) in the presence of LDA produced the aldol addition compound (V). Then, acid-promoted dehydration and simultaneous methoxymethyl group deprotection gave rise to a mixture of the desired E-benzylidene sultam and the corresponding Z-isomer (VII), which were separated by column chromatography.

PAPER

Novel Antiarthritic Agents with 1,2-Isothiazolidine-1,1-dioxide (γ-Sultam) Skeleton: Cytokine Suppressive Dual Inhibitors of Cyclooxygenase-2 and 5-Lipoxygenase

Shionogi Research Laboratories, Shionogi & Co., Ltd., Fukushima-ku, Osaka 553-0002, Japan, and Institute of Medical Science, St. Marianna University School of Medicine, Miyamae-ku, Kawasaki 216-8512, Japan
J. Med. Chem., 2000, 43 (10), pp 2040–2048
DOI: 10.1021/jm9906015
Abstract Image

Various 1,2-isothiazolidine-1,1-dioxide (γ-sultam) derivatives containing an antioxidant moiety, 2,6-di-tert-butylphenol substituent, were prepared. Some compounds, which have a lower alkyl group at the 2-position of the γ-sultam skeleton, showed potent inhibitory effects on both cyclooxygenase (COX)-2 and 5-lipoxygenase (5-LO), as well as production of interleukin (IL)-1 in in vitro assays. They also proved to be effective in several animal arthritic models without any ulcerogenic activities. Among these compounds, (E)-(5)-(3,5-di-tert-butyl-4-hydroxybenzylidene)-2-ethyl-1,2-isothiazolidine-1,1-dioxide (S-2474) was selected as an antiarthritic drug candidate and is now under clinical trials. The structure−activity relationships (SAR) examined and some pharmacological evaluations are described.

http://pubs.acs.org/doi/abs/10.1021/jm9906015

PAPER

Highly E-Selective and Effective Synthesis of Antiarthritic Drug Candidate S-2474 Using Quinone Methide Derivatives

Shionogi Research Laboratories, Shionogi & Company, Ltd., Fukushima-ku, Osaka 553-0002, Japan
J. Org. Chem., 2002, 67 (1), pp 125–128
DOI: 10.1021/jo0106795
 Abstract Image
We have developed an efficient and E-selective synthesis of an antiarthritic drug candidate (E)-(5)-(3,5-di-tert-butyl-4-hydroxybenzylidene)-2-ethyl-1,2-isothiazolidine-1,1-dioxide (S-2474), in which α-methoxy-p-quinone methide is used as a key intermediate. α-Methoxy-p-quinone methide was revealed to be an equiv. to a p-hydroxy protected benzaldehyde. It reacts smoothly with α-sulfonyl carbanion to give 1,6-addn. intermediates, which can be further processed to provide S-2474 directly in the presence of a base. This procedure gives S-2474 as an almost single isomer on the benzylidene double bond in excellent yield and thus is a very practical method adaptable to large-scale synthesis. The detailed mechanistic aspects are studied and discussed.
An improved synthesis has been reported. Acid -catalyzed ketalization of aldehyde (VIII) with trimethyl orthoformate provided the dimethyl acetal (IX) which, upon thermal decomposition in refluxing xylene, gave rise to the alpha-methoxy methylenequinone derivative (X ). This was then condensed with the lithio derivative of sultam (III) to form selectively the desired E-adduct. in an analogous procedure, aldehyde (VIII) was converted to the chloromethylene compound (XI) with methanesulfonyl chloride and triethylamine in refluxing CH2Cl2 . Condensation of (XI) with the lithiated sultam (III) furnished the desired E-benzylidene sultam.

PAPER

Development of One-Pot Synthesis of New Antiarthritic Drug Candidate S-2474 with High E-Selectivity

Chemical Development Department, CMC Development Laboratories, Shionogi & Co., Ltd., 1-3, Kuise Terajima 2-chome, Amagasaki, Hyogo 660-0813, Japan, and Shionogi Research Laboratories, Shionogi & Co., Ltd., 12-4, Sagisu 5-chome, Fukushima-ku, Osaka 553-0002, Japan
Org. Process Res. Dev., 2008, 12 (3), pp 442–446
DOI: 10.1021/op800008w

* To whom correspondence should be addressed. Telephone: +81-6-6401-8198 . Fax: +81-6-6401-1371. E-mail:takemasa.hida@shionogi.co.jp., †

Chemical Development Department, CMC Development Laboratories.

, ‡Shionogi Research Laboratories.

Abstract Image

A one-pot synthesis of S-2474 was developed to overcome the problems of a large number of steps, low stereoselectivity, low yield, a large amount of waste, and severe reaction conditions. Aldol-type condensation of 3,5-di-tert-butyl-4-hydroxybenzaldehyde and N-ethyl-γ-sultam was carried out with LDA and then quenched with water. Dehydration proceeded under basic conditions, providing S-2474 directly as a single isomer on the benzylidene double bond. The reaction mechanism appears to involve a quinone methide intermediate. Environmental assessment of the development of this compound is also discussed in this paper.

STR1

STR1

///////New,  Antiarthritic , Drug Candidate,  S-2474, Shionogi Research Laboratories, cyclooxygenase-2,  (COX-2),  5-lipoxygenase , (5-LO), PHASE 2, 158089-95-3, 158089-96-4, S2474, S 2474

CCN2CC\C(=C/c1cc(c(O)c(c1)C(C)(C)C)C(C)(C)C)S2(=O)=O


Filed under: Phase2 drugs Tagged: (5-LO), (COX-2), 158089-95-3, 158089-96-4, 5-lipoxygenase, Antiarthritic, Cyclooxygenase 2, Drug Candidate, new, phase 2, S-2474, S2474, Shionogi Research Laboratories

ацетазоламид , أسيتازولاميد [, 乙酰唑胺 , ACETAZOLAMIDE

$
0
0

ChemSpider 2D Image | acetazolamide | C4H6N4O3S2

ACETAZOLAMIDE
ацетазоламид ,  أسيتازولاميد [,  乙酰唑胺 ,
CAS 59-66-5
Acetamide, N-(5-(aminosulfonyl)-1,3,4-thiadiazol-2-yl)-
MW 222.245,MF  C4H6N4O3S2
Title: Acetazolamide
CAS Registry Number: 59-66-5
CAS Name: N-[5-(Aminosulfonyl)-1,3,4-thiadiazol-2-yl]acetamide
Additional Names: 5-acetamido-1,3,4-thiadiazole-2-sulfonamide; 2-acetylamino-1,3,4-thiadiazole-5-sulfonamide
Manufacturers’ Codes: 6063
Trademarks: Acetamox (Tobishi-Santen); Atenezol (Tsuruhara); Défiltran (Gallier); Diamox (Barr); Didoc (Sawai); Diuriwas (IFI); Donmox (Horita); Edemox (Wassermann); Fonurit (Chinoin); Glaupax (Erco)
Molecular Formula: C4H6N4O3S2
Molecular Weight: 222.25
Percent Composition: C 21.62%, H 2.72%, N 25.21%, O 21.60%, S 28.85%
Literature References: Carbonic anhydrase inhibitor. Prepn: R. O. Roblin, J. W. Clapp, J. Am. Chem. Soc. 72, 4890 (1950); J. W. Clapp, R. O. Roblin, US 2554816 (1951 to Am. Cyanamid). HPLC determn in pharmaceuticals: Z. S. Gomaa, Biomed. Chromatogr. 7, 134 (1993). Effect on retinal circulation: S. M. B. Rassam et al., Eye 7, 697 (1993). Clinical trial in postoperative elevation of intraocular pressure: I. D. Ladas et al., Br. J. Ophthalmol. 77, 136 (1993). Comprehensive description: J. Parasrampuria, Anal. Profiles Drug Subs. Excip. 22, 1-32 (1993). Review of efficacy in acute mountain sickness: L. D. Ried et al.,J. Wilderness Med. 5, 34-48 (1994).
Properties: Crystals from water, mp 258-259° (effervescence). Weak acid. pKa 7.2. Sparingly sol in cold water. Slightly sol in alcohol, acetone. Practically insol in carbon tetrachloride, chloroform, ether. Soly (mg/ml): polyethylene glycol-400 87.81; propylene glycol 7.44; ethanol 3.93; glycerin 3.65; water 0.72.
Melting point: mp 258-259° (effervescence)
pKa: pKa 7.2
Derivative Type: Sodium salt
CAS Registry Number: 1424-27-7
Trademarks: Vetamox (Am. Cyanamid)
Therap-Cat: Antiglaucoma; diuretic; in treatment of acute mountain sickness.
Therap-Cat-Vet: Diuretic.
Keywords: Antiglaucoma; Carbonic Anhydrase Inhibitor; Diuretic; Sulfonamide Derivatives.
Starting reaction occurs in-between hydrazine hydrate and ammonium thiocyanate that produces 1, 2-bis (thiocarbamoyl) hydrazine which on further treatment with phosgene undergoesrearrangements, particularly  molecular rearrangement through loss of ammonia to form 5-amino-2-mercapto-1, 3, 4-thiadiazole. Upon acylation of 5-amino-2-mercapto-1, 3, 4-thiadiazole gives a corresponding amide which on oxidation with aqueous chlorine affords the 2-sulphonyl chloride. The final step essentially consists of amidation by treatment with ammonia.

STR1 STR2

STR1 STR2 STR3

1H NMR

Paper

14N NQR, 1H NMR and DFT/QTAIM study of hydrogen bonding and polymorphism in selected solid 1,3,4-thiadiazole derivatives

*
Corresponding authors
a»Jozef Stefan« Institute, Jamova 39, 1000 Ljubljana, Slovenia
E-mail: janez.seliger@fmf.uni-lj.si
Fax: +386 1 2517281
Tel: +386 1 4766576
bFaculty of Mathematics and Physics, University of Ljubljana, Jadranska 19, 1000 Ljubljana, Slovenia
cFaculty of Physics, Adam Mickiewicz University, Umultowska 85, 61-614 Poznań, Poland
Phys. Chem. Chem. Phys., 2010,12, 13007-13019

DOI: 10.1039/C0CP00195C, http://pubs.rsc.org/en/content/articlelanding/2010/cp/c0cp00195c#!divAbstract

Graphical abstract: 14N NQR, 1H NMR and DFT/QTAIM study of hydrogen bonding and polymorphism in selected solid 1,3,4-thiadiazole derivatives

The 1,3,4-thiadiazole derivatives (2-amino-1,3,4-thiadiazole, acetazolamide, sulfamethizole) have been studied experimentally in the solid state by 1H–14N NQDR spectroscopy and theoretically by Density Functional Theory (DFT). The specific pattern of the intra and intermolecular interactions in 1,3,4-thiadiazole derivatives is described within the QTAIM (Quantum Theory of Atoms in Molecules)/DFT formalism. The results obtained in this work suggest that considerable differences in the NQR parameters permit differentiation even between specific pure association polymorphic forms and indicate that the stronger hydrogen bonds are accompanied by the larger η and smaller ν and e2Qq/h values. The degree of π-electron delocalization within the 1,3,4-thiadiazole ring and hydrogen bonds is a result of the interplay between the substituents and can be easily observed as a change in NQR parameters at N atoms. In the absence of X-ray data NQR parameters can clarify the details of crystallographic structure revealing information on intermolecular interactions.

////////////ацетазоламид ,  أسيتازولاميد [,  乙酰唑胺 , ACETAZOLAMIDE

CC(=O)NC1=NN=C(S1)S(N)(=O)=O


Filed under: GENERIC DRUG Tagged: ACETAZOLAMIDE, ацетазоламид, 乙酰唑胺, أسيتازولاميد [

Gemfibrozil

$
0
0

Gemfibrozil.svg

Gemfibrozil
CAS: 25812-30-0
 5-(2,5-Dimethylphenoxy)-2,2-dimethylpentanoic acid
2,2-dimethyl-5-(2,5-xylyloxy)valeric acid
Manufacturers’ Codes: CI-719
Trademarks: Decrelip (Ferrer); Genlip (Teofarma); Gevilon (Pfizer); Lipozid (Pfizer); Lipur (Pfizer); Lopid (Pfizer)
MF: C15H22O3
MW: 250.33
Percent Composition: C 71.97%, H 8.86%, O 19.17%
Properties: Crystals from hexane, mp 61-63°. bp0.02 158-159°. LD50 in mice, rats (mg/kg): 3162, 4786 orally (Kurtz).
Melting point: mp 61-63°
Boiling point: bp0.02 158-159°
Toxicity data: LD50 in mice, rats (mg/kg): 3162, 4786 orally (Kurtz)
Therap-Cat: Antilipemic.
 

Gemfibrozil

5-(2,5-Dimethylphenoxy)-2,2-dimethylpentanoic Acid

Gemfibrozil is classified as a fibric acid derivative and is used in the treatment of hyperlipidaemias. It has effects on plasma-lipid concentrations similar to those described under bezafibrate. The major effects of gemfibrozil have been a reduction in plasma-triglyceride concentrations and an increase in high-density lipoprotein (HDL) cholesterol concentrations. A reduction in very-low-density lipoprotein (VLDL)-triglyceride appears to be largely responsible for the fall in plasma triglyceride although reductions in HDL and low-density lipoprotein (LDL)-triglycerides have also been reported.
The effects of gemfibrozil on total cholesterol have been more variable: in general, LDL-cholesterol may be decreased in patients with pre-existing high concentrations and raised in those with low concentrations. The increase in HDL-cholesterol concentrations has resulted in complementary changes to the ratios of HDL-cholesterol to LDL-cholesterol and to total cholesterol. Gemfibrozil has successfully raised HDL-cholesterol concentrations in patients with isolated low levels of HDL-cholesterol but otherwise normal cholesterol concentrations.The Helsinki heart study assessed gemfibrozil for the primary prevention of ischaemic heart disease in middle-aged men with hyperlipidaemia. The usual dose, by mouth, is 1.2 g daily in two divided doses given 30 min before the morning and evening meals. Gemfibrozil is available as tablets for oral administration (Lopid: USP).

IR (KBr, cm–1): 2959.03, 2919.78, 2877.65, 1709.42, 1613.44, 1586.60, 1511.07, 1473.81, 1414.01, 1387.89, 1317.61, 1286.34, 1271.91, 1214.39, 1159.26, 1048.83, 996.57, 803.75;

1H NMR (DMSO, 500 MHz, δ ppm): 1.12 (s, 6H), 1.60 and 1.67 (m, 4H), 2.08 (s, 3H), 2.24 (s, 3H), 3.90 (t, 2H), 6.62 (d, 1H), 6.70 (s, 1H), 6.97 (d, 1H);

13C NMR and DEPT (DMSO, 500 MHz, δ ppm): 15.39 (CH3), 20.94 (CH3), 24.67 (CH2), 24.87 (CH3, CH3), 36.43 (CH2), 40.91 (C), 67.57 (CH2), 112.07 (CH), 120.45 (CH), 122.44 (C), 129.96 (CH), 135.93 (C), 156.43 (C), 178.56 (C);

MS M/Z (ESI): 251.16 [(MH)+].

STR1

Solvent:CDCl3Instrument Type:JEOLNucleus:1HFrequency:400 MHzChemical Shift Reference:TMS

 

1H NMR spectrum of C15H22O3 in CDCL3 at 400 MHz

Gemfibrozil is the generic name for an oral drug used to lower lipid levels. It belongs to a group of drugs known as fibrates. It is most commonly sold as the brand name, Lopid. Other brand names include Jezil and Gen-Fibro.

history

Gemfibrozil was selected from a series of related compounds synthesized in the laboratories of the American company Parke Davisin the late 1970s. It came from research for compounds that lower plasma lipid levels in humans and in animals.[1]

Actions

Therapeutic effects

Nontherapeutic effects and toxicities

Indications

Contraindications and precautions

  • Gemfibrozil should not be given to these patients:
    • Hepatic dysfunction
  • Gemfibrozil should be used with caution in these higher risk categories:
    • Biliary tract disease
    • Renal dysfunction
    • Pregnant women
    • Obese patients

Drug interactions

Environmental data

Gemfibrozil has been detected in biosolids (the solids remaining after wastewater treatment) at concentrations up to 2650 ng/g wet weight.[3] This indicates that it survives the wastewater treatment process.

SYNTHESIS

STR1

The sodium isobutyrate (I) is metallated with lithium diisopropylamide, and the resulting compound is alkylated with 3- (2,5-dimethylphenoxy) propyl bromide.

PATENT

Paul, L. C. 2,2-Dimethyl-ω-aryloxy alkanoic acids and salts and ester thereof. U.S. 3,674,836, 1972.

http://www.google.co.in/patents/US3674836

CLIP

Production of Gemfibrozil
(1)2,5-Dimethylphenol and 1-Bromo-3-chloropropane reaction of 1-(2,5-dimethylphenoxy)-3-chloropropane. The reaction is carried out in toluene, adding new clean off reflux 5h. Just as follows:

Production of Gemfibrozil

(2)N/A can be used to manufacture Gemfibrozil.

Production of Gemfibrozil

PAPER

Improved Process for Preparation of Gemfibrozil, an Antihypolipidemic

Chemical Research and Development, Aurobindo Pharma Ltd., Survey No. 71 and 72, Indrakaran (V), Sangareddy (M), Medak District-502329, Andhra Pradesh, India
Engineering Chemistry Department, AU College of Engineering, Andhra University, Visakhapatnam-530003, Andhra Pradesh, India
Org. Process Res. Dev., 2013, 17 (7), pp 963–966

An improved process for the preparation of gemfibrozil, an antihypolipodimic drug substance, with an overall yield of 80% and ∼99.9% purity (including three chemical reactions) is reported. Formation and control of possible impurities are also described. Finally, gemfibrozil is isolated from water without any additional solvent purification.

STR1

Literature References:

Serum lipid regulating agent. Prepn: P. L. Creger, DE 1925423; eidem, US 3674836 (1969, 1972, both to Parke, Davis).

Production: O. P. Goel, US 4126637 (1978 to Warner-Lambert).

Pharmacology: A. H. Kissebach et al.,Atherosclerosis 24, 199 (1976); M. T. Kahonen et al., ibid. 32, 47 (1979).

Series of articles on metabolism, clinical pharmacology, kinetics and toxicology: Proc. R. Soc. Med. 69, Suppl 2, 1-120 (1976).

Toxicity data: S. M. Kurtz et al., ibid. 15.

Clinical trial in hyperlipidemia: J. E. Lewis et al., Pract. Cardiol. 9, 99 (1983).

Clinical reduction of cardiovascular risk in patients with low HDL levels: H. B. Rubins et al., N. Engl. J. Med. 341, 410 (1999).

References

External links

Gemfibrozil
Gemfibrozil.svg
Systematic (IUPAC) name
5-(2,5-dimethylphenoxy)-2,2-dimethyl-pentanoic acid
Clinical data
Trade names Lopid
AHFS/Drugs.com Monograph
MedlinePlus a686002
Pregnancy
category
  • Category C
Routes of
administration
Oral
Legal status
Legal status
  • By Prescription
Pharmacokinetic data
Bioavailability Close to 100%
Protein binding 95%
Metabolism Hepatic (CYP3A4)
Biological half-life 1.5 hours
Excretion Renal 94%
Feces 6%
Identifiers
CAS Number 25812-30-0 Yes
ATC code C10AB04 (WHO)
PubChem CID 3463
IUPHAR/BPS 3439
DrugBank DB01241 Yes
ChemSpider 3345 Yes
UNII Q8X02027X3 Yes
KEGG D00334 Yes
ChEBI CHEBI:5296 Yes
ChEMBL CHEMBL457 Yes
Chemical data
Formula C15H22O3
Molar mass 250.333 g/mol

LOPID® (gemfibrozil tablets, USP) is a lipid regulating agent. It is available as tablets for oral administration. Each tablet contains 600 mg gemfibrozil. Each tablet also contains calcium stearate, NF; candelilla wax, FCC; microcrystalline cellulose, NF; hydroxypropyl cellulose, NF; hypromellose, USP; methylparaben, NF; Opaspray white; polyethylene glycol, NF; polysorbate 80, NF; propylparaben, NF; colloidal silicon dioxide, NF; pregelatinized starch, NF. The chemical name is 5-(2,5-dimethylphenoxy)2,2-dimethylpentanoic acid, with the following structural formula:

 

LOPID® (gemfibrozil) Structural Formula Illustration

The empirical formula is C15H22O3 and the molecular weight is 250.35; the solubility in water and acid is 0.0019% and in dilute base it is greater than 1%. The melting point is 58° –61°C. Gemfibrozil is a white solid which is stable under ordinary conditions.

/////////Gemfibrozil,  Antilipemic,  Fibrates, 25812-30-0,

CC1=CC(OCCCC(C)(C)C(O)=O)=C(C)C=C1


Filed under: Uncategorized Tagged: 25812-30-0, Antilipemic, Fibrates, Gemfibrozil

USFDA approves Indoco’s Allopurinol ANDA

$
0
0

usfda-approval-Allopurinol-forprint.jpg

Indoco Remedies Limited (India) | Facebook

https://www.facebook.com/Indoco-Remedies-Limited-India-317944458228011/

USFDA approves Indoco’s Allopurinol ANDA… Indoco Remedies Limited (India)’s … Indoco Remedies Limited (India) added a new photo

Allopurinol 3d structure.png

Allopurinol, sold under the brand name Zyloprim and generics, is a medication used primarily to treat excess uric acid in the bloodand its complications, including chronic gout. It is a xanthine oxidase inhibitor and is administered orally.

It is on the World Health Organization’s List of Essential Medicines, a list of the most important medication needed in a basic health system.

Allopurinol has been marketed in the United States since August 19, 1966, when it was first approved by FDA under the trade name Zyloprim. Allopurinol was marketed at the time by Burroughs-Wellcome. Allopurinol is now a generic drug sold under a variety of brand names, including Allohexal, Allosig, Milurit, Alloril, Progout, Ürikoliz, Zyloprim, Zyloric, Zyrik, and Aluron

Aditi Kare Panandikar, Managing Director, Indoco Remedies

click above

Aditi Kare Panandikar gets award

///////////Indoco Remedies LtdUSFDA,  approves,  Indoco’s,  Allopurinol,  ANDA, Aditi Kare Panandikar, Managing Director,


Filed under: ANDA, COMPANIES Tagged: Aditi Kare Panandikar, Allopurinol, anda, Approves, Indoco Remedies Ltd, Indoco's, Managing Director, usfda

Scaling up from mg to Kgs – Making your First GMP Batch

$
0
0

STR1

Scaling up from mg to Kgs – Making your First GMP Batch 

6th – 7th October 2016, Clearwater, USA

the course was very informative and it allowed me to see the big picture from discovery stage to pilot plant” 
Genentech

Course Outline:

  • Introduction
  • Making the first 100g non-GMP Batch
  • Non-GMP vs GMP preparation
  • Physical version and form
  • Process safety and raw materials supply
  • Scaling into fixed vessels
  • Technology transfer
  • Genotoxic impurities
  • Case studies and Review

Who should attend:

  • Project managers
  • Project leaders
  • Bench chemists
  • New starters
  • MedChem Support teams

This course aims to provide attendees with a good understanding of the issues involved taking development candidates to the first in human trials.

Click here to Download the Course Brochure

Presented by Dr John Knight, JKonsult Ltd

John Knight

Managing Director at JKONSULT Ltd

STR1
Click here to Download the Course Brochure

“Brilliant Course, learn lots of tips and tricks”
Vertex

First incursion into Chemical Development has been very, very educational. John’s way of explaining the material has been wonderful.”
Almirall

Very clear and interesting sessions with a lot of relevant examples and not only theory.” 
Oribase Pharma
LINK
LITERATURE FROM INTERNET ON HIS TOPIC
//////////Scaling up,  mg to Kgs, Making,  First GMP Batch, SCIENTIFIC UPDATE,  JOHN KNIGHT, Clearwater, USA

Filed under: COMPANIES, GENERICS Tagged: Clearwater, First GMP Batch, JOHN KNIGHT, Making, mg to Kgs, Scaling up, SCIENTIFIC UPDATE, USA

FDA published generic user fee for 2017: for ANDA, DMF, and for Facility (API, FDF)

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

.

placeholder+image

http://www.raps.org/Regulatory-Focus/News/2016/07/26/25394/FDA-Lowers-ANDA-Fee-Rates-for-2017/

Generic drugmakers submitting abbreviated new drug applications (ANDAs) and prior approval supplements (PAS) will see their US Food and Drug Administration (FDA) fee rates drop in 2017, though all other rates, including those for drug master files (DMF) and facility fees will increase when compared to 2016.

For FY 2017, the generic drug fee rates are: ANDA ($70,480, down from $76,030 in 2016), PAS ($35,240, down from $38,020 in 2016), DMF ($51,140, up from $42,170 in 2016), domestic active pharmaceutical ingredient (API) facility ($44,234, up from $40,867 in 2016), foreign API facility ($59,234, up from $55,867 in 2016), domestic finished dose formulation (FDF) facility ($258,646, up from $243,905), and foreign FDF facility ($273,646, up from $258,905 in 2016).

The new fees are effective 1 October 2016 and will remain in effect through 30 September 2017.

FDA explained the increases and decreases in fees, noting that for ANDA…

View original post 413 more words


Filed under: Uncategorized

Arformoterol, (R,R)-Formoterol For Chronic obstructive pulmonary disease (COPD)

$
0
0

Arformoterol.svg

Arformoterol

  • MF C19H24N2O4
  • MW 344.405
(R,R)-Formoterol
Cas 67346-49-0
Chronic obstructive pulmonary disease (COPD)
  • Sunovion/Sepracor (Originator)
  • Asthma Therapy, Bronchodilators, Chronic Obstructive Pulmonary Diseases (COPD), Treatment of, RESPIRATORY DRUGS, beta2-Adrenoceptor Agonists
  • LAUNCHED 2007 , Phase III ASTHMA
Formamide, N-[2-hydroxy-5-[(1R)-1-hydroxy-2-[[(1R)-2-(4-methoxyphenyl)-1-methylethyl]amino]ethyl]phenyl]-

Arformoterol is a long-acting β2 adrenoreceptor agonist (LABA) indicated for the treatment of chronic obstructive pulmonary disease(COPD). It is sold by Sunovion, under the trade name Brovana, as a solution of arformoterol tartrate to be administered twice daily (morning and evening) by nebulization.[1]

Arformoterol inhalation solution, a long-acting beta2-adrenoceptor agonist, was launched in the U.S. in 2007 for the long-term twice-daily (morning and evening) treatment of bronchospasm in patients with chronic obstructive pulmonary disease (COPD), including chronic bronchitis and emphysema. The product, known as Brovana(TM), for use by nebulization only, is the first long-acting beta2-agonist to be approved as an inhalation solution for use with a nebulizer. The product was developed and is being commercialized by Sunovion Pharmaceuticals (formerly Sepracor)

Arformoterol.png

It is the active (R,R)-(−)-enantiomer of formoterol and was approved by the United States Food and Drug Administration (FDA) on October 6, 2006 for the treatment of COPD.

Arformoterol is a bronchodilator. It works by relaxing muscles in the airways to improve breathing. Arformoterol inhalation is used to prevent bronchoconstriction in people with chronic obstructive pulmonary disease, including chronic bronchitis and emphysema. The use of arformoterol is pending revision due to safety concerns in regards to an increased risk of severe exacerbation of asthma symptoms, leading to hospitalization as well as death in some patients using long acting beta agonists for the treatment of asthma.

Arformoterol is an ADRENERGIC BETA-2 RECEPTOR AGONIST with a prolonged duration of action. It is used to manage ASTHMA and in the treatment of CHRONIC OBSTRUCTIVE PULMONARY DISEASE.

 
Arformoterol is a beta2-Adrenergic Agonist. The mechanism of action of arformoterol is as an Adrenergic beta2-Agonist.
Arformoterol is a long-acting beta-2 adrenergic agonist and isomer of formoterol with bronchodilator activity. Arformoterol selectively binds to and activates beta-2 adrenergic receptors in bronchiolar smooth muscle, thereby causing stimulation of adenyl cyclase, the enzyme that catalyzes the conversion of adenosine triphosphate (ATP) to cyclic-3′,5′-adenosine monophosphate (cAMP). Increased intracellular cAMP levels cause relaxation of bronchial smooth muscle and lead to a reduced release of inflammatory mediators from mast cells. This may eventually lead to an improvement of airway function.

Arformoterol tartrate

  • Molecular FormulaC23H30N2O10
  • Average mass494.492
  •  cas 200815-49-2
  • 183-185°C
Butanedioic acid, 2,3-dihydroxy-, (2R,3R)-, compd. with formamide, N-[2-hydroxy-5-[(1R)-1-hydroxy-2-[[(1R)-2-(4-methoxyphenyl)-1-methylethyl]amino]ethyl]phenyl]- (1:1) [ACD/Index Name]
N-{2-hydroxy-5-[(1R)-1-hydroxy-2-{[(1R)-2-(4-methoxyphenyl)-1-methylethyl]amino}ethyl]phenyl}formamide 2,3-dihydroxybutanedioate (salt)
N-[2-Hydroxy-5-[(1R)-1-hydroxy-2-[[(1R)-2-(4-methoxyphenyl)-1-methylethyl]amino]ethyl]phenyl]formamide (+)-(2R,3R)-Tartaric Acid; (-)-Formoterol 1,2-Dihydroxyethane-1,2-dicarboxylic Acid; (R,R)-Formoterol Threaric Acid; Arformoterol d-Tartaric Acid; Arformoterol d-α,β-Dihydroxysuccinic Acid
(R,R)-Formoterol-L-(+)-tartrate
200815-49-2 CAS
Arformoterol tartrate (USAN)
Brovana
UNII:5P8VJ2I235
Arformoterol Tartrate, can be used in the synthesis of Omeprazole (O635000), which is a proton pump inhibitor, that inhibits gasteric secretion, also used in the treatment of dyspepsia, peptic ulcer disease, etc. Itis also the impurity of Esomeprazole Magnesium (E668300), which is the S-form of Omeprazole, and is a gastric proton-pump inhibitor. Also, It can be used for the preparation of olodaterol, a novel inhaled β2-adrenoceptor agonist with a 24h bronchodilatory efficacy.
 

SYNTHESIS

PATENT

us-9309186

Example 1

Synthesis of (R,R)-Formoterol-L-tartrate Form D

A solution containing 3.9 g (26 mmol) of L-tartaric acid and 36 mL of methanol was added to a solution of 9 g (26 mmol) of arformoterol base and 144 mL methanol at 23.degree. C. Afterwards, the resulting mixture was seeded with form D and stirred at 23.degree. C. for 1 hour. It was then further cooled to 0-5.degree. C. for 1 hour and the product collected by filtration and dried under inlet air (atmospheric pressure) for 16 hours to provide 11.1 g (86% yield) (99.7% chemical purity, containing 0.14% of the degradation impurity (R)-1-(3-amino-4-hydroxyphenyl)-2-[[(1R)-2-(4-methoxyphenyl)-1-methylethy- l]amino]ethanol) of (R,R)-formoterol L-tartrate form D, as an off white powder. .sup.1H-NMR (200 MHz, d.sub.6-DMSO) .delta.: 1.03 (d, 3H); 2.50-2.67 (m, 5H); 3.72 (s, 3H); 3.99 (s, 2H); 4.65-4.85 (m, 1H); 6.82-7.15 (m, 5H); 8.02 (s, 1H); 8.28 (s, 1H); 9.60 (s, NH). No residual solvent was detected (.sup.1H-NMR).

PSD: d.sub.50=2.3 .mu.m.

 PAPER
Tetrahedron Letters, Vol. 38, No. 7, pp. 1125-1128, 1997
Enantio- and Diastereoselective Synthesis of all Four Stereoisomers of Formoterol
 STR1
STR1
PAPER

Taking Advantage of Polymorphism To Effect an Impurity Removal:  Development of a Thermodynamic Crystal Form of (R,R)-FormoterolTartrate

Chemical Research and Development, Sepracor Inc., 111 Locke Drive, Marlborough, Massachusetts 01752, U.S.A.
Org. Proc. Res. Dev., 2002, 6 (6), pp 855–862
DOI: 10.1021/op025531h

Abstract

Abstract Image

The development and large-scale implementation of a novel technology utilizing polymorphic interconversion and crystalline intermediate formation of (R,R)-formoterol l-tartrate ((R,R)-FmTA, 1) as a tool for the removal of impurities from the final product and generation of the most thermodynamically stable crystal form is reported. The crude product was generated by precipitation of the free base as the l-tartrate salt in a unique polymorphic form, form B. Warming the resultant slurry effected the formation of a partially hydrated stable crystalline intermediate, form C, with a concomitant decrease in the impurity levels in the solid. Isolation and recrystallization of form C provided 1 in the thermodynamically most stable polymorph, form A.

SYN1
SYN 2
SYN 3
 SYN 4
SYN 5

PATENT

Formoterol, (+/-)N-[2-hydroxy-5-[1-hydroxy-2-[[2-(p-methoxyphenyl)-2-propylamino]ethyl]phenyl]-formamide, is a highly potent and β2-selective adrenoceptor agonist having a long lasting bronchodilating effect when inhaled. Its chemical structure is depicted below:
Figure imgb0001
Formoterol has two chiral centres, each of which can exist into two different configurations. This results into four different combinations, (R,R), (S,S), (S,R) and (R,S). Formoterol is commercially available as a racemic mixture of 2 diasteromers (R,R) and (S,S) in a 1:1 ratio. The generic name Formoterol always refers to its racemic mixture. Trofast et al. (Chirality, 1, 443, 1991) reported on the potency of these isomers, showing a decrease in the order of (R,R)>(R,S)≥(S,R)>(S,S). The (R,R) isomer, also known as Arformoterol, being 1000 fold more potent than the (S,S) isomer. Arformoterol is commercialised by Sepracor as Brovana
Formoterol was first disclosed in Japanese patent application (Application N° 13121 ) whereby Formoterol is synthesised by N-alkylation using a phenacyl bromide as described in the scheme below:
Figure imgb0002
Afterwards, a small number of methods have been reported so far, regarding the synthesis of the (R,R) isomer, also referred as (R,R)-Formoterol and Arformoterol.
Murase et al. [Chem. Pharm. Bull. 26(4) 1123-1129(1978)] reported the preparation of (R,R)-Formoterol from a racemic mixture of the (R,R) and (S,S) isomers by optical resolution using optically active tartaric acid. Trofast et al. described a method in which 4-benzyloxy-3-nitrostyrene oxide was coupled with a optically pure (R,R)- or (S,S)-N-phenylethyl-N-(1-p-methoxyphenyl)-2-(propyl)amine to give a diastereomeric mixture of Formoterol precursors. These precursors were further separated by HPLC in order to obtain pure Formoterol isomers. Both synthetic processes undergo long synthetic procedures and low yields.
Patent publication EP0938467 describes a method in which Arformoterol is prepared via the reaction of the optically pure (R) N-benzyl-2-(4-methoxyphenyl)-1-(methylethylamine) with an optically pure (R)-4-benzyloxy-3-nitrostyrene oxide or (R)-4-benzyloxy-3-formamidostyrene oxide followed by formylation of the amino group. This method requires relatively severe reaction conditions, 24 h at a temperature of from 110 up to 130 °C as well as a further purification step using tartaric acid in order to eliminate diastereomer impurities formed during the process.
WO2009/147383 discloses a process for the preparation of intermediates of Formoterol and Arformoterol which comprises a reduction of a ketone intermediate of formula:
Figure imgb0003
Using chiral reductive agent with an enantiomeric excess of about 98% which requires further purification steps to obtain a product of desired optical purity.
 R,R)-Formoterol (Arformoterol) or a salt thereof from optically pure and stable intermediate (R)-2-(4-Benzyloxy-3-nitro-phenyl)-oxirane (compound II), suitable for industrial use, in combination with optically pure amine in higher yields, as depicted in the scheme below:
Figure imgb0011

Compound (R, R)-1-(4-Benzyloxy-3-nitro-phenyl)-2-[[2-(4-methoxy-phenyl)-1-methylethyl]-(1-phenyl-ethyl)-amino]-ethanol (compound VI), having the configuration represented by the following formula:

Figure imgb0018

Examples(R)-2-(4-Benzyloxy-3-nitro-phenyl)-oxirane (II)

A solution of 90 g (0.25 mol) of (R)-1-(4-Benzyloxy-3-nitro-phenyl)-2-bromo-ethanol (compound I) in 320 mL of toluene and 50 mL of MeOH was added to a stirred suspension of 46 g (0.33 mol) of K2CO3 in 130 mL of toluene and 130 mL of MeOH. The mixture was stirred at 40°C for 20 h and washed with water (400 mL). The organic phase was concentrated under reduced pressure to a volume of 100 mL and stirred at 25 °C for 30 min. It was then further cooled to 0-5°C for 30 min. and the product collected by filtration and dried at 40 °C to provide 67.1 g (97% yield) (98% chemical purity, 100% e.e.) of compound II as an off-white solid. 1 H-NMR (200 MHz, CDCl3) δ: 2.80-2.90 (m, 2H); 3.11-3.20 (m, 2H), 3.80-3.90 (m, 1H); 5.23 (s, 2H); 7.11 (d, 2H); 7.41 (m, 5H), 7.76 (d, 2H).

Preparation of (R,R)-[2-(4-Methoxy-phenyl)-1-methyl-ethyl]-(1-phenyl-ethyl)-amine (III)

A solution of 13 g (78.6 mmol) of 1-(4-Methoxy-phenyl)-propan-2-one and 8.3 g (78.6 mmol) of (R)-1-Phenylethylamine in 60 mL MeOH was hydrogenated in the presence of 1.7 g of Pt/C 5% at 10 atm. and 30 °C for 20 h. The mixture was filtered though a pad of diatomaceous earth and concentrated under reduced pressure to give compound III as an oil. The obtained oil was dissolved in 175 mL of acetone, followed by addition of 6.7 mL (80.9 mmol) of a 12M HCl solution. The mixture was stirred at 23 °C for 30 min and at 0-5 °C for 30 min. The product collected by filtration and dried at 40 °C to provide 13.8 g of the hydrochloride derivate as a white solid. The obtained solid was stirred in 100 mL of acetone at 23 °C for 1h and at 0-5 °C for 30 min, collected by filtration and dried at 40 °C to provide 13.2 g of the hydrochloride derivate as a white solid. This compound was dissolved in 100 mL of water and 100 mL of toluene followed by addition of 54 mL (54 mmol) of 1N NaOH solution. The organic phase was concentrated to give 11.7 g (55% yield) (99% chemical purity and 100% e.e) of compound III as an oil.1H-NMR (200 MHz, CDCl3) δ: 0.88 (d, 3H); 1.31 (d, 3H), 2.40-2.50 (m, 1H); 2.60-2.80 (m, 2H); 3.74 (s, 3H); 3.90-4.10 (m, 1H); 6.77- 6.98 (m, 4H), 7.31 (s, 5H).

Synthesis of (R,R)-1-(4-Benzyloxy-3-nitro-phenyl)-2-[[2-(4-methoxy-phenyl)-1-methyl-ethyl]-(1-phenyl-ethyl)-amino]-ethanol (IV)

A 1-liter flask was charged with 50g (0.18 mol) of II and 50g (0.18 mol) of III and stirred under nitrogen atmosphere at 140 °C for 20 h. To the hot mixture was added 200 mL of toluene to obtain a solution, which was washed with 200 mL of 1N HCl and 200 mL of water. The organic phase was concentrated under reduced pressure to give 99 g (99% yield) (88% chemical purity) of compound IV as an oil. Enantiomeric purity 100%. 1H-NMR (200 MHz, CDCl3) δ: 0.98 (d, 3H); 1.41 (d, 3H), 2.60-2.90 (m, 4H); 3.20-3.30 (m, 1H); 3.74 (s, 3H); 4.10-4.20 (m, 1H); 4.30-4.40 (m, 1H), 5.19 (s, 2H); 6.69-7.42 (m, 16H); 7.77 (s, 1H).

Synthesis of (R, R)-1-(3-Amino-4-benzyloxy-phenyl)-2-[[2-(4-methoxy-phenyl)-1-methyl-ethyl]-(1-phenyl-ethyl)-amino]-ethanol (V)

A solution of 99 g (0.18 mol) of IV in 270 mL IPA and 270 mL toluene was hydrogenated in the presence of 10 g of Ni-Raney at 18 atm and 40 °C for 20 h. The mixture was filtered though a pad of diatomaceous earth and the filtrate was concentrated under reduced pressure to give 87 g (92% yield) (83% chemical purity, 100 % e.e.) of compound V as an oil. 1H-NMR (200 MHz, CDCl3) δ: 0.97 (d, 3H); 1.44 (d, 3H), 2.60-2.90 (m, 4H); 3.20-3.30 (m, 1H); 3.74 (s, 3H); 4.10-4.20 (m, 1H); 4.30-4.40 (m, 1H), 5.07 (s, 2H); 6.67-6.84 (m, 7H); 7.25-7.42 (m, 10H).

Synthesis of (R,R)-N-(2-Benzyloxy-5-{1-hydroxy-2-[[2-(4-methoxy-phenyl)-1-methyl-ethyl]-(1-phenyl-ethyl)-amino]-ethyl)-phenyl)-formamide (VI)

24 mL (0.63 mol) of formic acid was added to 27 mL (0.28 mol) of acetic anhydride and stirred at 50 °C for 2 h under nitrogen atmosphere. The resulting mixture was diluted with 100 mL of CH2Cl2 and cooled to 0 °C. A solution of 78 g (0.15 mol) of V in 300 mL de CH2Cl2 was slowly added and stirred for 1h at 0 °C. Then, 150 mL of 10% K2CO3 aqueous solution were added and stirred at 0 °C for 15 min. The organic phase was washed twice with 400 mL of 10% K2CO3 aqueous solution and concentrated under reduced pressure to give 80 g (97% yield, 100% e.e.) (75% chemical purity) of compound VI as an oil. 1H-NMR (200 MHz, CDCl3) δ: 0.98 (d, 3H); 1.42 (d, 3H), 2.60-2.90 (m, 4H); 3.20-3.30 (m, 1H); 3.75 (s, 3H); 4.10-4.20 (m, 1H); 4.30-4.40 (m, 1H), 5.09 (s, 2H); 6.67-7.41 (m, 17H); 8.4 (d, 1H).

Synthesis (R,R)-N-(2-Hydroxy-5-{1-hydroxy-2-[2-(4-methoxy-phenyl)-1-methyl-ethylamino]-ethyl}-phenyl)-formamide (VII)

A solution of 8.5 g (16 mmol) of VI, previous purified by column chromatography on silica gel (AcOEt/heptane, 2:3), in 60 mL ethanol was hydrogenated in the presence of 0.14 g of Pd/C 5% at 10 atm. and 40 °C for 20 h. The mixture was filtered though a pad of diatomaceous earth and concentrated under reduced pressure to give 5 g (93% yield) (91% chemical purity, 100% e.e.) of compound VII as foam. m. p.= 58-60 °C. 1H-NMR (200 MHz, d6-DMSO) δ: 0.98 (d, 3H); 2.42-2.65 (m, 5H); 3.20-3.40 (m, 1H); 3.71 (s, 3H); 4.43-4.45 (m, 1H); 6.77-7.05 (m, 5H); 8.02 (s, 1H), 8.26 (s, 1H).

Synthesis (R,R)-N-(2-Hydroxy-5-{1-hydroxy-2-[2-(4-methoxy-phenyl)-1-methyl-ethylamino]-ethyl}-phenyl)-formamide (VII)

A solution of 46 g (0.08 mol) of VI, crude product, was dissolved in 460 mL ethanol and hydrogenated in the presence of 0.74 g of Pd/C 5% at 10 atm. and 40 ° C for 28 h. The mixture was filtered though a pad of diatomaceous earth and the filtrate was concentrated under reduced pressure to give 24 g (83% yield) (77% chemical purity, 100% e.e.) of compound VII as a foam. m. p. = 58-60 °C. 1H-NMR (200 MHz, d6-DMSO) δ: 0.98 (d, 3H); 2.42-2.65 (m, 5H); 3.20-3.40 (m, 1H); 3.71 (s, 3H); 4.43-4.45 (m, 1H); 6.77-7.05 (m, 5H); 8.02 (s, 1H), 8.26 (s, 1H).

The HPLC conditions used for the determination of the Chemical purity % are described in the table below:

  • HPLC Column Kromasil 100 C-18
    Dimensions 0.15 m x 4.6 mm x 5 µm
    Buffer 2.8 ml TEA (triethylamine) pH=3.00 H3PO4 (85%) in 1 L of H2O
    Phase B Acetonitrile
    Flow rate 1.5 ml miN-1
    Temperature 40 °C
    Wavelength 230 nm

    The HPLC conditions used for the determination of the enantiomeric purity % are described in the table below:

    HPLC Column Chiralpak AD-H
    Dimensions 0.25 m x 4.6 mm
    Buffer n-hexane : IPA : DEA (diethyl amine) : H2O 85:15:0.1:0.1
    Flow rate 0.8 ml min-1
    Temperature 25 °C
    Wavelength 228 nm

PATENT

Example 1

(R) -2- (4- benzyloxy-3-nitrophenyl) oxirane (I) (9. 86g, 36mmol) and (R) -I- (4- methoxy- phenyl) -N – [(R) -I- phenyl-ethyl] -2-amino-propane (II) (10. 8g, 40mmol) cast in the reaction flask, the reaction 20 hours at 140 ° C, the chiral Intermediate (III) (17. 3g, yield 88%). HPLC: de values of> 90%; MS (ESI) m / z: 541 3 (M ++ 1); 1H-NMR (CDCl3):.. Δ 0. 96 (d, 3H), 1 49 (d, 3H ), 2 · 15 (q, 1Η), 2 · 67 (dq, 2H), 2. 99 (dq, 2H), 3. 74 (s, 3H), 4. 09 (d, 1H), 4. 56 (q, 1H), 5. 24 (s, 2H), 6. 77 (dd, 4H), 7. 10 (d, 1H), 7. 25-7. 5 (m, 11H), 7. 84 ( s, 1H).

 Example 2

 (R) -2- (4- benzyloxy-3-nitrophenyl) oxirane (I) (9. 86g, 36mmol) and (R) -I- (4- methoxybenzene yl) -N – [(R) -I- phenyl-ethyl] -2-amino-propane (II) (10. 8g, 40mmol) and toluene 100ml, 110 ° C0-flow reactor 36 hours, the solvent was distilled off succeeded intermediates (III) (16. 8g, yield 85%).

Example 3

(R) -2- (4- benzyloxy-3-nitrophenyl) oxirane (I) (9. 86g, 36mmol) and (R) -I- (4- methoxybenzene After [(R) -I- phenyl-ethyl] -2-amino-propane (II) (10. 8g, 40mmol) and dichloromethane 100ml, 30 ° C for 48 hours, and the solvent was distilled off – yl) -N succeeded intermediates (III) (15. Sg, yield 80%).

Example 4

 (R) -2- (4- benzyloxy-3-nitrophenyl) oxirane (I) (9. 86g, 36mmol) and (R) -I- (4- methoxybenzene yl) -N – [(R) -I- phenyl-ethyl] -2-amino-propane (II) (8. 75g, 32mmol) cast in the reaction flask, the reaction 20 hours at 140 ° C, the chiral intermediate form (III) (16. 3g, 83% yield).

Example 5

 (R) -2- (4- benzyloxy-3-nitrophenyl) oxirane (I) (9. 86g, 36mmol) and (R) -I- (4- methoxybenzene yl) -N – [(R) -I- phenyl-ethyl] -2-amino-propane (II) (14. 6g, 54mmol) cast in the reaction flask, the reaction 20 hours at 140 ° C, the chiral intermediate form (III) (17. 5g, 89% yield).

STR1

Scheme

chirality 1991, 3, 443-50
Fumaric acid (0.138 mmol, 16 mg) was added to the residue dissolved in methanol. Evaporation of the solvent gave the
product (SS) W semifumarate (109 mg) characterized by ‘HNMR (4-D MSO) 6 (ppm) 1.00 (d, 3H, CHCH,), 4.624.70 (m, lH,
CHOH), 3.73 (s, 3H, OCH,), 6.M.9 (m, 3H, aromatic), 7.00 (dd,4H, aromatic), 6.49 (s, 1@ CH = CH fumarate). MS of disilylated
(SS) W: 473 (M +<H3,7%); 367 (M ‘<8H90, 45%); 310 61%). The (RSS) fraction was treated in the same manner
giving the product (R;S) W semifumarate, which was characterized by ‘H-NMR (4-DMSO) 6 (ppm) 1.01 (d, 3H, CHCH,),
3.76 (s, 3H, OC&), 6.49 (s, lH, CH=CH, fumarate) 6.M.9 (m, 3H, aromatic), 7.0 (dd, 4H, aromatic). MS of disilylated (R;S)
(M’X~~HIGNO1,7 %); 178 ( C I ~H~ ~N95O%,) ; 121 (CsH90, W. 473 (M’4H3, 5%); 367 (M’4gH90, 48%); 310
(M +–CI~HIGNO18, %); 178 (CIIHIGNO, 95%); 121 (CsH90, 52%). The structural data for the (RR) and (S;R) enantiomers
were in accordance with the proposed structures. The enantiomeric purity obtained for the enantiomers in each batch is
shown in Table 1.
STR1
Scheme
The enantioselective reduction of phenacyl bromide (I) with BH3.S(CH3)2 in THF catalyzed by the chiral borolidine (II) (obtained by reaction of (1R,2S)-1-amino-2-indanol (III) with BH3.S(CH3)2 in THF) gives the (R)-2-bromo-1-(4-benzyloxy-3-nitrophenyl)ethanol (IV), which is reduced with H2 over PtO2 in THF/toluene yielding the corresponding amino derivative (V). The reaction of (V) with formic acid and Ac2O affords the formamide (VI), which is condensed with the chiral (R)-N-benzyl-N-[2-(4-methoxyphenyl)-1-methylethyl]amine (VII) in THF/methanol providing the protected target compound (VIII). Finally, this compound is debenzylated by hydrogenation with H2 over Pd/C in ethanol. The intermediate the chiral (R)-N-benzyl-N-[2-(4-methoxyphenyl)-1-methylethyl]amine (VII) has been obtained by reductocondensation of 1-(4-methoxyphenyl)-2-propanone (IX) and benzylamine by hydrogenation with H2 over Pd/C in methanol yielding racemic N-benzyl-N-[2-(4-methoxyphenyl)-1-methylethyl]amine (X), which is submitted to optical resolution with (S)-mandelic acid to obtain the desired (R)-enantiomer (VII).
Org Process Res Dev1998,2,(2):96

Large-Scale Synthesis of Enantio- and Diastereomerically Pure (R,R)-Formoterol

Process Research and Development, Sepracor Inc., 111 Locke Drive, Marlborough, Massachusetts 01752
Org. Proc. Res. Dev., 1998, 2 (2), pp 96–99
DOI: 10.1021/op970116o

Abstract

(R,R)-Formoterol (1) is a long-acting, very potent β2-agonist, which is used as a bronchodilator in the therapy of asthma and chronic bronchitis. Highly convergent synthesis of enantio- and diastereomerically pure (R,R)-formoterol fumarate is achieved by a chromatography-free process with an overall yield of 44%. Asymmetric catalytic reduction of bromoketone 4 using as catalyst oxazaborolidine derived from (1R, 2S)-1-amino-2-indanol and resolution of chiral amine 3 are the origins of chirality in this process. Further enrichment of enantio- and diastereomeric purity is accomplished by crystallizations of the isolated intermediates throughout the process to give (R,R)-formoterol (1) as the pure stereoisomer (ee, de >99.5%).

STR1
STR1

Scheme

The intermediate N-benzyl-N-[1(R)-methyl-2-(4-methoxyphenyl)ethyl]amine (IV) has been obtained as follows: The reductocondensation of 1-(4-methoxyphenyl)-2-propanone (I) with benzylamine (II) by H2 over Pd/C gives the N-benzyl-N-[1-methyl-2-(4-methoxyphenyl)ethyl]amine (III) as a racemic mixture, which is submitted to optical resolution with L-mandelic acid in methanol to obtain the desired (R)-enantiomer (IV). The reaction of cis-(1R,2S)-1-aminoindan-2-ol (V) with trimethylboroxine in toluene gives the (1R,2S)-oxazaborolidine (VI), which is used as chiral catalyst in the enantioselective reduction of 4-benzyloxy-3-nitrophenacyl bromide (VII) by means of BH3/THF, yielding the chiral bromoethanol derivative (VIII). The reaction of (VIII) with NaOH in aqueous methanol affords the epoxide (IX), which is condensed with the intermediate amine (IV) by heating the mixture at 90 C to provide the adduct (X). The reduction of the nitro group of (X) with H2 over PtO2 gives the corresponding amino derivative (XI), which is acylated with formic acid to afford the formamide compound (XII). Finally, this compound is debenzylated by hydrogenation with H2 over Pd/C in ethanol, providing the target compound.
The synthesis of the chiral borolidine catalyst (II) starting from indoline (I), as well as the enantioselective reduction of 4′-(benzyloxy)-3′-nitrophenacyl bromide (III), catalyzed by borolidine (II), and using various borane complexes (borane/dimethylsulfide, borane/THF and borane/diethylaniline), has been studied in order to solve the problems presented in large-scale synthesis. The conclusions of the study are that the complex borane/diethylaniline (DEANB) is the most suitable reagent for large-scale reduction of phenacyl bromide (III) since the chemical hazards and inconsistent reagent quality of the borane/THF and borane/dimethylsulfide complexes disqualified their use in large-scale processes. The best reaction conditions of the reduction with this complex are presented.
PATENT

Formoterol is a long-acting β2-adrenoceptor agonist and has a long duration of action of up to 12 hours. Chemically it is termed as Λ/-[2-hydroxy-5-[1-hydroxy-2-[[2-(4- methoxyphenyl)propan-2-yl]amino]ethyl]phenyl]-formamide. The structure of formoterol is as shown below.

Figure imgf000003_0001

The asterisks indicate that formoterol has two chiral centers in the molecule, each of which can exist in two possible configurations. This gives rise to four diastereomers which have the following configurations: (R,R), (S1S), (S1R) and (R1S).

(R1R) and (S1S) are mirror images of each other and are therefore enantiomers. Similarly (S1R) and (R1S) form other enatiomeric pair.

The commercially-available formoterol is a 50:50 mixture of the (R1R)- and (S1S)- enantiomers. (R,R)-formoterol is an extremely potent full agonist at the β2-adrenoceptor and is responsible for bronchodilation and has anti-inflammatory properties. On the other hand (S,S)-enantiomer, has no bronchodilatory activity and is proinflammatory.

Murase et al. [Chem.Pharm.Bull., .26(4)1123-1129(1978)] synthesized all four isomers of formoterol and examined for β-stimulant activity. In the process, racemic formoterol was subjected to optical resolution with tartaric acid.

In another attempt by Trofast et al. [Chirality, 3:443-450(1991 )], racemic 4-benzyloxy-3- nitrostryrene oxide was coupled with optically pure N-[(R)-1-phenylethyl]-2-(4- methoxyphenyl)-(R)1-methylethylamine to give diastereomeric mixtures of intermediates, which were separated by column chromatography and converted to the optically pure formoterol.

In yet another attempt, racemic formoterol was subjected to separation by using a chiral compound [International publication WO 1995/018094].

WO 98/21175 discloses a process for preparing optically pure formoterol using optically pure intermediates (R)-N-benzyl-2-(4-methoxyphenyl)-1-methylethyl amine and (R)-4- benzyloxy-3-formamidostyrene oxide.

Preparation of optically pure formoterol is also disclosed in IE 000138 and GB2380996.

Example 7

Preparation of Arformoterol

4-benzyloxy-3-formylamino-α-[N-benzyl-N-(1-methyl-2-p- methoxyphenylethyl)aminomethyl]benzyl alcohol (120gms, 0.23M), 10% Pd/C (12 gms) and denatured spirit (0.6 lit) were introduced in an autoclave. The reaction mass was hydrogenated by applying 4 kg hydrogen pressure at 25-300C for 3 hrs. The catalyst was removed by filtration and the, clear filtrate concentrated under reduced pressure below 400C to yield the title compound. (63 gms, 80%).

Example 8

Preparation of Arformoterol Tartrate

Arformoterol base (60 gms, 0.17M), 480 ml IPA , 120 ml toluene and a solution of l_(+)- tartaric acid (25.6 gms, 0.17M) in 60 ml distilled water were stirred at 25-300C for 2 hrs and further at 40°- 45°C for 3 hrs. The reaction mass was cooled to 25-300C and further chilled to 200C for 30 mins. The solid obtained was isolated by filtration to yield the title compound. (60 gms, 70%),

The tartrate salt was dissolved in hot 50% IPA-water (0.3 lit), cooled as before and filtered to provide arformoterol tartrate. (30 gms, 50 % w/w). having enantiomeric purity greater than 99%.

 PAPER
Organic Process Research & Development 2000, 4, 567-570
 Modulation of Catalyst Reactivity for the Chemoselective Hydrogenation of a Functionalized Nitroarene: Preparation of a Key Intermediate in the Synthesis of (R,R)-Formoterol Tartrate………..http://pubs.acs.org/doi/abs/10.1021/op000287k

Modulation of Catalyst Reactivity for the Chemoselective Hydrogenation of a Functionalized Nitroarene:  Preparation of a Key Intermediate in the Synthesis of (R,R)-Formoterol Tartrate

Chemical Research and Development, Sepracor Inc., 111 Locke Drive, Marlborough, Massachusetts 01752, U.S.A.
Org. Proc. Res. Dev., 2000, 4 (6), pp 567–570
DOI: 10.1021/op000287k
In the synthesis of the β2-adrenoceptor agonist (R,R)-formterol, a key step in the synthesis was the development of a highly chemoselective reduction of (1R)-2-bromo-1-[3-nitro-4-(phenylmethoxy)phenyl]ethan-1-ol to give (1R)-1-[3-amino-4-(phenylmethoxy)phenyl]-2-bromoethan-1-ol. The aniline product was isolated as the corresponding formamide. The reaction required reduction of the nitro moiety in the presence of a phenyl benzyl ether, a secondary benzylic hydroxyl group, and a primary bromide, and with no racemization at the stereogenic carbinol carbon atom. The development of a synthetic methodology using heterogeneous catalytic hydrogenation to perform the required reduction was successful when a sulfur-based poison was added. The chemistry of sulfur-based poisons to temper the reacitivty of catalyst was studied in depth. The data show that the type of hydrogenation catalyst, the oxidation state of the poison, and the substituents on the sulfur atom had a dramatic effect on the chemoselectivity of the reaction. Dimethyl sulfide was the poison of choice, possessing all of the required characteristics for providing a highly chemoselective and high yielding reaction. The practicality and robustness of the process was demonstrated by preparing the final formamide product with high chemoselectivity, chemical yield, and product purity on a multi-kilogram scale.
 STR1

 PAPER

Tetrahedron: Asymmetry 11 (2000) 2705±2717
An ecient enantioselective synthesis of (R,R)-formoterol, a potent bronchodilator, using lipases
Francisco Campos, M. Pilar Bosch and Angel Guerrero*
STR1
STR1
STR1
STR1
STR1
 formoterol (R,R)-1 as amorphous solid. Rf: 0.27 (SiO2, AcOEt:MeOH, 1:1).‰Š20D=-41.5 (CHCl3, c 0.53).
IR, : 3383, 2967, 2923, 1674, 1668, 1610, 1514, 1442, 1247, 1033,815 cm^1.
1H NMR (300 MHz, CDCl3), : 8.11 (b, 1H), 7.46 (b, 1H), 6.99 (d, J=8.4 Hz, 2H), 6.9±6.7 (c, 4H), 4.46 (m, 1H), 4.34 (b, 3H interchangeable), 3.74 (s, 3H), 2.90±2.45 (c, 5H), 1.02 (d,J=5.7 Hz, 3H) ppm.
13C NMR (75 MHz, CDCl3), : 160.2, 158.3, 147.7, 133.4, 130.6, 130.2 (2C),125.7, 123.7, 119.5, 117.8, 114.0 (2C), 71.3, 55.3, 54.7, 53.6, 42.0, 19.4 ppm.
CI (positive, LC-MS)(m/z, %) 435 (M+1, 100).
The tartrate salt was prepared by dissolving 13.8 mg (0.04 mmol) of(R,R)-1 and 6.0 mg (0.04 mmol) of (l)-(+)-tartaric acid in 150 mL of 85% aqueous isopropanol.
The solution was left standing overnight and the resulting crystalline solid (7.6 mg) puri®ed on areverse-phase column (1 g, Isolute SPE C18) using mixtures of MeOH±H2O as eluent. The solventwas removed under vacuum and the aqueous solution lyophilized (^35C, 0.6 bar) overnight. The(l)-(+)-tartrate salt of (R,R)-1 showed an ‰Š20D=-29.4 (H2O, c 0.61) (>99% ee based on the
reported value 34). 34=Hett, R.; Senanayake, C. H.; Wald, S. A. Tetrahedron Lett. 1998, 39, 1705.
PAPER

Diethylanilineborane:  A Practical, Safe, and Consistent-Quality Borane Source for the Large-Scale Enantioselective Reduction of a Ketone Intermediate in the Synthesis of (R,R)-Formoterol

Chemical Research and Development, Sepracor Incorporated, 111 Locke Drive, Marlborough, Massachusetts 01752, U.S.A.
Org. Proc. Res. Dev., 2002, 6 (2), pp 146–148
DOI: 10.1021/op015504b

Abstract

Abstract Image

The development of a process for the use of N,N-diethylaniline−borane (DEANB) as a borane source for the enantioselective preparation of a key intermediate in the synthesis of (R,R)-formoterol l-tartrate, bromohydrin 2, from ketone 3 on kilogram scale is described. DEANB was found to be a more practical, safer, and higher-quality reagent when compared to other more conventional borane sources:  borane−THF and borane−DMS.

PAPER

http://nopr.niscair.res.in/bitstream/123456789/8917/1/IJCB%2044B(1)%20167-169.pdf

str1

str1

PAPER

http://www.bioorg.org/down/Hetetorcycles_07_2243.pdf?ckattempt=1

str1

str1

str1

PAPER

Drugs R D. 2004;5(1):25-7.

Arformoterol: (R,R)-eformoterol, (R,R)-formoterol, arformoterol tartrate, eformoterol-sepracor, formoterol-sepracor, R,R-eformoterol, R,R-formoterol.

Abstract

Sepracor in the US is developing arformoterol [R,R-formoterol], a single isomer form of the beta(2)-adrenoceptor agonist formoterol [eformoterol]. This isomer contains two chiral centres and is being developed as an inhaled preparation for the treatment of respiratory disorders. Sepracor believes that arformoterol has the potential to be a once-daily therapy with a rapid onset of action and a duration of effect exceeding 12 hours. In 1995, Sepracor acquired New England Pharmaceuticals, a manufacturer of metered-dose and dry powder inhalers, for the purpose of preparing formulations of levosalbutamol and arformoterol. Phase II dose-ranging clinical studies of arformoterol as a longer-acting, complementary bronchodilator were completed successfully in the fourth quarter of 2000. Phase III trials of arformoterol began in September 2001. The indications for the drug appeared to be asthma and chronic obstructive pulmonary disease (COPD). However, an update of the pharmaceutical product information on the Sepracor website in September 2003 listed COPD maintenance therapy as the only indication for arformoterol. In October 2002, Sepracor stated that two pivotal phase III studies were ongoing in 1600 patients. Sepracor estimates that its NDA submission for arformoterol, which is projected for the first half of 2004, will include approximately 3000 adult subjects. Sepracor stated in July 2003 that it had completed more than 100 preclinical studies and initiated or completed 15 clinical studies for arformoterol inhalation solution for the treatment of bronchospasm in patients with COPD. In addition, Sepracor stated that the two pivotal phase III studies in 1600 patients were still progressing. In 1995, European patents were granted to Sepracor for the use of arformoterol in the treatment of asthma, and the US patent application was pending.

CLIP

str1

str1

str1

PAPER

doi:10.1016/j.cclet.2008.01.012

http://www.sciencedirect.com/science/article/pii/S1001841708000132

Volume 19, Issue 3, March 2008, Pages 279–280

Cover image

New method in synthesizing an optical active intermediate for (R,R)-formoterol

  • Key Laboratory of Drug Targeting Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China\

Abstract

(R)-1-(4-Methoxyphenyl)propan-2-amine 2a, an optical active intermediate for (R,R)-formoterol, was synthesized from d-alanine in 65% overall yield by using a simple route, which contained protecting amino group, cyclization, coupling with Grignard reagent, reduction and deprotection.

References

Muller, P., et al.: Arzneimittel-Forsch., 33, 1685 (1983); Wallmark, B., et al.: Biochim. Biophys. Acta., 778, 549 (1984); Morii, M., et al.: J. Biol. chem., 268, 21553 (1993); Ritter, M., et al.: Br. J. Pharmacol., 124, 627 (1998); Stenhoff, H., et al.: J. Chromatogr., 734, 191 (1999), Johnson, D.A., et al.: Expert Opin. Pharmacother., 4, 253 (2003); Bouyssou, T., et al.: Bio. Med. Chem. Lett. 20, 1410, (2010);

External links

EP0390762A1 * 23 Mar 1990 3 Oct 1990 Aktiebolaget Draco New bronchospasmolytic compounds and process for their preparation
EP0938467A1 7 Nov 1997 1 Sep 1999 Sepracor, Inc. Process for the preparation of optically pure isomers of formoterol
EP1082293A2 20 May 1999 14 Mar 2001 Sepracor Inc. Formoterol polymorphs
WO2009147383A1 2 Jun 2009 10 Dec 2009 Cipla Limited Process for the synthesis of arformoterol
Reference
1 * HETT R ET AL: “Enantio- and Diastereoselective Synthesis of all Four Stereoisomers of Formoterol” TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL LNKD- DOI:10.1016/S0040-4039(97)00088-9, vol. 38, no. 7, 17 February 1997 (1997-02-17), pages 1125-1128, XP004034214 ISSN: 0040-4039
2 * LING HUANG ET AL.: “The Asymmetric Synthesis of (R,R)-Formoterol via Transfer Hydrogenation with Polyethylene Glycol Bound Rh Catalyst in PEG2000 and Water” CHIRALITY, vol. 22, 30 April 2009 (2009-04-30), pages 206-211, XP002592699
3 MURASE ET AL. CHEM. PHARM. BULL. vol. 26, no. 4, 1978, pages 1123 – 1129
4 TROFAST ET AL. CHIRALITY vol. 1, 1991, page 443
5 * TROFAST J ET AL: “STERIC ASPECTS OF AGONISM AND ANTAGONISM AT BETA-ADRENICEPTORS: SYNTHESIS OF AND PHARMACOLOGICAL EXPERIMENTS WITH THE ENANTIOMERS OF FORMOTEROL AND THEIR DIASTEREOMERS” CHIRALITY, WILEY-LISS, NEW YORK, US LNKD- DOI:10.1002/CHIR.530030606, vol. 3, no. 6, 1 January 1991 (1991-01-01) , pages 443-450, XP002057060 ISSN: 0899-0042
6 WILKINSON, H.S ET AL. ORGANIC PROCESS RESEARCH AND DEVELOPMENT vol. 6, 2002, pages 146 – 148

Durham E-Theses A Solid-state NMR Study of Formoterol Fumarate

Arformoterol
Arformoterol.svg
Arformoterol ball-and-stick model.png
Systematic (IUPAC) name
N-[2-hydroxy-5-[(1R)-1-hydroxy-2-[[(2R)-1-(4-methoxyphenyl) propan-2-yl]amino]ethyl] phenyl]formamide
Clinical data
Trade names Brovana
AHFS/Drugs.com Monograph
MedlinePlus a602023
License data
Pregnancy
category
  • US: C (Risk not ruled out)
Routes of
administration
Inhalation solution fornebuliser
Legal status
Legal status
Pharmacokinetic data
Protein binding 52–65%
Biological half-life 26 hours
Identifiers
CAS Number 67346-49-0 Yes
ATC code none
PubChem CID 3083544
IUPHAR/BPS 7479
DrugBank DB01274 Yes
ChemSpider 2340731 Yes
UNII F91H02EBWT Yes
ChEBI CHEBI:408174 Yes
ChEMBL CHEMBL1201137 
Chemical data
Formula C19H24N2O4
Molar mass 344.405 g/mol

 

Formoterol

Formoterol

CAS Registry Number: 73573-87-2
CAS Name: relN-[2-Hydroxy-5-[(1R)-1-hydroxy-2-[[(1R)-2-(4-methoxyphenyl)-1-methylethyl]amino]ethyl]phenyl]formamide
Additional Names: 3-formylamino-4-hydroxy-a-[N-[1-methyl-2-(p-methoxyphenyl)ethyl]aminomethyl]benzyl alcohol; (±)-2¢-hydroxy-5¢-[(RS)-1-hydroxy-2-[[(RS)-p-methoxy-a-methylphenethyl]amino]ethyl]formanilide
Molecular Formula: C19H24N2O4
Molecular Weight: 344.40
Percent Composition: C 66.26%, H 7.02%, N 8.13%, O 18.58%
Literature References: Selective b2-adrenergic receptor agonist. Mixture of R,R (-) and S,S (+) enantiomers. Prepn: M. Murakamiet al., DE 2305092; eidem, US 3994974 (1973, 1976 both to Yamanouchi); K. Murase et al., Chem. Pharm. Bull. 25, 1368 (1977). Absolute configuration and activity of isomers: eidem, ibid. 26, 1123 (1978). Toxicity studies: T. Yoshida et al., Pharmacometrics26, 811 (1983). HPLC determn in plasma: J. Campestrini et al., J. Chromatogr. B 704, 221 (1997). Review of pharmacology: G. P. Anderson, Life Sci. 52, 2145-2160 (1993); and clinical efficacy: R. A. Bartow, R. N. Brogden, Drugs 55, 303-322 (1998).
Derivative Type: Fumarate dihydrate
CAS Registry Number: 43229-80-7
Manufacturers’ Codes: BD-40A
Trademarks: Atock (Yamanouchi); Foradil (Novartis); Oxeze (AstraZeneca)
Molecular Formula: (C19H24N2O4)2.C4H4O4.2H2O
Molecular Weight: 840.91
Percent Composition: C 59.99%, H 6.71%, N 6.66%, O 26.64%
Properties: Crystals from 95% isopropyl alcohol, mp 138-140°. pKa1 7.9; pKa2 9.2. Log P (octanol/water): 0.4 (pH 7.4). Freely sol in glacial acetic acid; sol in methanol; sparingly sol in ethanol, isopropanol; slightly sol in water. Practically insol in acetone, ethyl acetate, diethyl ether. LD50 in male, female, rats, mice (mg/kg): 3130, 5580, 6700, 8310 orally; 98, 100, 72, 71 i.v.; 1000, 1100, 640, 670 s.c.; 170, 210, 240, 210 i.p. (Yoshida).
Melting point: mp 138-140°
pKa: pKa1 7.9; pKa2 9.2
Log P: Log P (octanol/water): 0.4 (pH 7.4)
Toxicity data: LD50 in male, female, rats, mice (mg/kg): 3130, 5580, 6700, 8310 orally; 98, 100, 72, 71 i.v.; 1000, 1100, 640, 670 s.c.; 170, 210, 240, 210 i.p. (Yoshida)
Derivative Type: R,R-Form
CAS Registry Number: 67346-49-0
Additional Names: Arformoterol
Derivative Type: R,R-Form L-tartrate
CAS Registry Number: 200815-49-2
Additional Names: Arformoterol tartrate
Molecular Formula: C19H24N2O4.C4H6O6
Molecular Weight: 494.49
Percent Composition: C 55.86%, H 6.12%, N 5.67%, O 32.36%
Literature References: Prepn: Y. Gao et al., WO 9821175; eidem, US 6040344 (1998, 2000 both to Sepracor). Pharmacology: D. A. Handley et al., Pulm. Pharmacol. Ther. 15, 135 (2002).
Properties: Off-white powder, mp 184°.
Melting point: mp 184°
Therap-Cat: Antiasthmatic.
Keywords: ?Adrenergic Agonist; Bronchodilator; Ephedrine Derivatives.

//////Arformoterol, (R,R)-Formoterol, (R,R)-Formoterol-L-(+)-tartrate, 200815-49-2, Arformoterol tartrate , Brovana, UNII:5P8VJ2I235, Sepracor, Asthma Therapy, Bronchodilators, Chronic Obstructive Pulmonary Diseases, COPD ,  RESPIRATORY DRUGS, beta2-Adrenoceptor Agonists, Phase III, 2007, Sunovion

COC1=CC=C(C[C@@H](C)NC[C@H](O)C2=CC(NC=O)=C(O)C=C2)C=C1


Filed under: GENERICS Tagged: (R)-, 2007, 200815-49-2, Arformoterol, Arformoterol tartrate, Asthma Therapy, beta2-Adrenoceptor Agonists, Bronchodilators, Brovana, Chronic Obstructive Pulmonary Diseases, COPD, Phase III, R)-Formoterol, R)-Formoterol-L-(+)-tartrate, RESPIRATORY DRUGS, Sepracor, Sunovion, UNII:5P8VJ2I235

APIC is working on an enhanced ICH Q7 How to do Guide

$
0
0

Acting on the publication of the ICH Q7 Q&A Document in June last year APIC has decided to elaborate another revision of its “ICH Q7 How to do Document” taking into account these Q&As. Here you will get to know why this upcoming How to do Document is a valuable support for API and API intermediate manufacturers regarding the practical implementation of the ICH Q7 What to do principles.

see

http://www.gmp-compliance.org/enews_05493_APIC-is-working-on-an-enhanced-ICH-Q7-How-to-do-Guide_15332,S-WKS_n.html

The first edition of the “How to do Document – Interpretation of the ICH Q7 Guide” was published by APIC shortly after the ICH Q7 Guideline appeared as Step 4 document in November 2000. In the meantime it has undergone a numer of revisions and the current version of the How to do Document to be found on the publications section of APIC’s website is an update from August 2015 (version 8). This document was written by experts from the European Industry (CEFIC APIC) and is essentially an interpretation of “how to” implement the requirements of the ICH Q7 Guide based on practical experience.

On the occasion of the publication of the ICH Q7 Questions & Answers Document in June 2015 another revision of APIC’s How to do Document was necessary. APIC is currently working on this new revision (version 9) taking into account the new ICH Q7 Q&A Document which will be incorporated into the How to do Document as a new chapter 21. In this chapter each Question/Answer of the Q&A document is examined and commented with respect to its practical implementation. The following example illustrates what these practical comments look like:

ICH Q7 Q&A – Question
For dedicated equipment, is ‘visually clean’ acceptable for verification of cleaning effectiveness, (i.e., no expectation for specific analytical determination)?

ICH Q7 Q&A – Answer
‘Visually clean’ may be acceptable for dedicated equipment based on the ability to visually inspect and sufficient supporting data from cleaning studies (e.g., analytical determination to demonstrate cleaning effectiveness) [ICH Q7, Section 12.76]. Equipment should be cleaned at appropriate intervals (e.g., time or number of batches) to prevent build-up and carryover of contaminants (e.g., degradants or objectionable levels of microorganisms) so that they do not adversely alter the quality of the API [ICH Q7, Sections 5.23, 12.7].

Comment in APIC’s How to do Document
“dedicated equipment” can be defined in various ways such as:
– a reactor that is used solely for 1 API process
– a reactor used for different intermediate steps of the same API.
– a reactor used for different steps in the same intermediate or API
– a reactor solely used for 1 stage in 1 process
Whatever definition is used it should be documented and justified.
When visual inspection is applied following points should be considered:
– adequate lighting
– fully dried
– difficult to clean spots visually inspectable
– use of cameras, endoscopy
– limit of detection of visual cleanliness
– dirty hold time / clean hold time
– Campaign length

While the purpose of the ICH Q7 Q&A Document is to clarify some equivocal issues of the ICH Q7 Guideline the comments of APIC’s ICH Q7 Q&A How to do Document intend to give support on an even more practical level. So after its finalisation and publication later this year the API industry will have another very useful document available which facilitates the implementation of the ICH Q7 principles.

At the pre-Conference Session to the 19th APIC/CEFIC European Conference on Active Pharmaceutical Ingredients on 22 November 2016 in Barcelona APIC will launch the ICH Q7 Q&A How to do Document as a stand alone document. All participants will receive a copy.
///////////////


Filed under: Regulatory Tagged: apic, ICH Q7 Q&A

New WHO guidance on variations to multisource pharmaceutical products

$
0
0

The World Health Organizaton (WHO) recently published a new Annex 10 on handling of variations. Find out more about the new WHO general guidance on variations to multisource pharmaceutical products.

see http://www.gmp-compliance.org/enews_05425_New-WHO-guidance-on-variations-to-multisource-pharmaceutical-products_15152,15173,Z-RAM_n.html

The World Health Organizaton recently published a new Annex 10: WHO general guidance on variations to multisource pharmaceutical products.

The document is “intended to serve as a guide for establishing national requirements for the regulation of post-approval changes.” Proposed categories and reporting procedures are provided. Modifications may be justified in light of risk-benefit and legal considerations specific to each national medicines regulatory authority (NMRA).

The guideline can be used with respect to changes to the quality sections of product dossiers and should be read in context with the Guidelines on submission of documentation for a multisource (generic) finished product: quality part as well as other related WHO guidelines or applicable national guidelines.

The guideline emphasizes that “even well-resourced agencies find it difficult to evaluate all the pharmaceutical changes that are made to all products.” Therefore, “a shift towards increased self-assessment of changes by the marketing authorization (MA) holder” has been observed. In this context “it is necessary, to define those changes that can be made without the NMRA´s prior approval (self-assessable changes) and those that require prior approval based on an understanding of the risk and how best to manage the risk.”

Description of the reporting categories are discussed in section 5 of the Annex:

  • Notifications: They can be made for changes to the product that may have no potential or minimal potential to have a negative impact on the quality, safety and efficacy (QSE) of a finished pharmaceutical product (FPP). Implementation of such variations may be possible without prior approval by the NMRA (e.g. submission of these variations as annual notifications).
  • Minor variations: Changes with moderate or negative impact on the QSE. Such changes must be submitted to the NMRA with all required documentation prior to implementation. The MA holder may implement the change if no objection has been issued within a specific time period by the NMRA.
  • Major variations: They have a significant potential to have a negative impact on the QSE. A major variation should be reviewed and approved by the NMRA prior to implementation of the change. To increase the efficiency NMRAs may accept grouping of variations under specific circumstances (see also FDA´s new policy regarding grouping of CMC changes).
  • Editorial changes: They typically need not to be submitted and can be included as a notification together with a subsequent variation concerning that part of the dossier.

Certain changes can be so fundemental that they require a submission of a new dossier. Examples of such changes are:

  • Change of the API to a different API,
  • Inclusion of an additional API in a multicomponent product,
  • Removal of one API from a multicomponent product,
  • Change in the dose and/ or strength of one or more APIs,
  • Change from an immediate-release product to an extended- or delayed-release dosage form or vice versa,
  • Change from a liquid to a powder for reconstitution or vice versa,
  • Changes in the route of administration.

For any change in the SmPC, patient information leaflet (PIL)  and/or labels the NMRAS should be notified and submission of the revised product information and/or labelling is expected as per country-specific requirements.

Proposed recommendations on the regulatory procedures for the reporting of changes to the NMRA are discussed in section 8 of the Annex:

  • General: It is essential that different branches of the NMRA interact and exchange information effectively. They should, for example, discuss whether a change may require a GMP inspection or may be reviewed during the next routine inspection.
  • Presubmission meetings: Procedures should be established to allow MA holders the opportunity to obtain advice prior to submitting variations.
  • Proposed documentation for minor variations: Basic information like
    – covering letter,
    – application form,
    – a list of subsections of the current dossier affected by the change(s),
    – a list and description of each change, reason for change(s) and the date each change was implemented,
    – relevant summary of data from studies and tests performed,
    – copies of the updated subsections of the original dossier,
    should be included as part of the immediate or annual notification, where prior approval is not required.
  • Proposed documentation for variations requiring prior approval: Additionally to the documents mentioned under “minor variations”:
    – replacement of the relevant subsections of the dossier in accordance with the acceptable dossier format for the NMRAs concerned, with the proposed changes clearly annotated,
    – copies of the SmPC, PIL and labels, if relevant,
    – registration status and date of the proposed change(s) in other countries and/or agencies that have already approved the variation(s), especially the country of origin and the reference agencies.
  • Review procedures: NMRA should adopt a risk-based review strategy for assessment, concentratiing most effort on those changes considered to carry the greatest risk. NMRA may consider whether it will rely on decisions and/or assessment reports made by other national authorities or prepare its own full assessment report or use a combination of those approaches.

For more Information please see the new Annex 10 – WHO general guidance on variations to multisource pharmaceutical products.

//////////////Annex 10,  WHO general guidance, multisource pharmaceutical products.


Filed under: Regulatory Tagged: Annex 10, multisource pharmaceutical products, WHO general guidance

Eribulin, エリブリンメシル酸塩 an Antineoplastic

$
0
0

Eribulin

Eribulin mesylate

エリブリンメシル酸塩

CAS 441045-17-6 MESYLATE

C41H63NO14S, 826.00222 g/mol

halichrondrin B analog, B1939, E7389, ER-086526,Halaven

CAS 253128-41-5  FREE FORM

(1S,3S,4R)-3-tert-butoxycarbonylamino-4-hydroxycyclopentanecarboxylic acid methyl ester;

(1S,3S,6S,9S,12S,14R,16R,18S,20R,21R,22S,26R,29S,31R,32S,33R,35R,36S)-20-[(2S)-3-Amino-2-hydroxypropyl]-21-methoxy-14-methyl-8,15-bis(methylene)-2,19,30,34,37,39,40,41-octaoxanonacyclo[24.9.2.13,32.13,33.16,9.112,16.018,22.029,36.031,35]hentetracontan-24-one;

2-(3-Amino-2-hydroxypropyl)hexacosahydro-3-methoxy- 26-methyl-20,27-bis(methylene)11,15-18,21-24,28-triepoxy- 7,9-ethano-12,15-methano-9H,15H-furo(3,2-i)furo(2′,3′-5,6) pyrano(4,3-b)(1,4)dioxacyclopentacosin-5-(4H)-one

(2R,3R,3aS,7R,8aS,9S,10aR,11S,12R,13aR,13bS,15S,18S,21S,24S,26R,28R,29aS)-2-((2S)-3-amino-2-hydroxypropyl)-3-methoxy-26-methyl-20,27-dimethylidenehexacosahydro-11,15:18,21:24,28-triepoxy-7,9-ethano-12,15-methano-9H,15H-furo(3,2-i)furo(2′,3′:5,6)pyrano(4,3-b)(1,4)dioxacyclopentacosin-5(4H)-one methanesulfonate (salt)

11,15:18,21:24,28-Triepoxy-7,9-ethano-12,15-methano-9H,15H-furo(3,2-i)furo(2′,3′:5,6)pyrano(4,3-b)(1,4)dioxacyclopentacosin-5(4H)-one, 2-((2S)-3- amino-2-hydroxypropyl)hexacosahydro-3-methoxy-26-methyl-20,27-bis(methylene)-, 2R,3R,3aS,7R,8aS,9S,10aR,11S,12R,13aR,13bS,15S,18S,21S,24S,26R,28R,29aS)-, methanesulfonate (salt)

エリブリンメシル酸塩
Eribulin Mesilate

C40H59NO11▪CH4O3S : 826
[441045-17-6]

Eribulin mesylate is the mesylate salt of a synthetic analogue of halichondrin B, a substance derived from a marine sponge (Lissodendoryx sp.) with antineoplastic activity.

E7389 is the mesylate salt of a synthetic analogue of halichondrin B, a substance derived from a marine sponge (Lissodendoryx sp.) with antineoplastic activity. Eribulin binds to the vinca domain of tubulin and inhibits the polymerization of tubulin and the assembly of microtubules, resulting in inhibition of mitotic spindle assembly, induction of cell cycle arrest at G2/M phase, and, potentially, tumor regression.

Halichondrin B, a large polyether macrolide, was isolated 25 years ago from the marine sponge Halichondria okadai

Halichondria okadaiHalaven.png

ERBULIN

The anti-cancer drug made from a sea-spongeEribulin is an anticancer drug marketed by Eisai Co. under the trade name Halaven. Eribulin mesylate was approved by the U.S. Food and Drug Administration on November 15, 2010, to treat patients with metastatic breast cancer who have received at least two prior chemotherapy regimens for late-stage disease, including both anthracycline– and taxane-based chemotherapies.[1] It was approved by Health Canada on December 14, 2011 for treatment of patients with metastatic breast cancer who have previously received at least two chemotherapeutic regimens for the treatment of metastatic disease. [2]

Eribulin is also being investigated by Eisai Co. for use in a variety of other solid tumors, including non-small cell lung cancer, prostate cancer and sarcoma.[3]

Eribulin has been previously known as E7389 and ER-086526, and also carries the US NCI designation NSC-707389.

Eribulin mesylate is an analogue of halichondrin B, which in 1986 was isolated from the marine sponge Halichondria okadai toxic Pacific.Halichondrin B has a significant anti-tumor activity. The Eribulin synthetically obtained has a simpler but still complex molecular structure.Taxanes such as to inhibit the spindle apparatus of the cell, but it is engaged in other ways.

Drug substance, eribulin mesylate, is a It is a structurally simplified synthetic analogue of halichondrin B, a natural product isolated from the marine sponge Halichondira okadai. Eribulin mesylate is a white powder which is freely soluble in water, methanol, ethanol, 1-octanol, benzyl alcohol, dichloromethane, dimethylsulfoxide, N-methylpyrrolidone and ethyl acetate. It is soluble in acetone, sparingly soluble in acetonitrile, and practically insoluble in tertbutyl methyl ether, n-heptane and n-pentane. Eribulin mesylate is characterized by ion chromatography for counter ion content, and spectroscopic analyses (mass, ultraviolet, nuclear magnetic resonance, single crystal X-ray crystallography, and circular dichroism) for molecular structure and absolute configuration. Bulk drug substance is hygroscopic and sensitive to light, heat, and acid hydrolysis,,,,,,……..http://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/201532orig1s000chemr.pdf

 

STR1

Melvin Yu received his B.S. from MIT, and both his M.A. and Ph.D. degrees from Harvard University while studying under Professor Yoshito Kishi. In 1985, he joined Eli Lilly, and in 1993 he relocated to Eisai Inc. where he led the chemistry team that discovered Halaven. He was then responsible for the initial route nding and synthesis scale-up effort that ultimately provided the rst multi-gram batch of eribulin mesylate. Mel retains a strong interest in natural products as the inspiration of new chemotherapeutic agents, and in this context recently expanded his area of research to include cheminformatics and compound library design.

STR1

Wanjun Zheng received a Ph.D. in organic chemistry from Wesleyan University in 1994 under the direction of Professor Peter A. Jacobi working on synthetic methodology development and its application in natural product synthesis. He spent over two years as a postdoctoral research fellow in Harvard University under Professor Yoshito Kishi working on the complete structure determination of maitotoxin. He joined Eisai in 1996 and has since been contributing and leading many drug discovery projects including project in the discovery of Halaven.

STR2

Boris M. Seletsky earned his PhD in 1987 from Shemyakin Institute of Bioorganic Chemistry in Moscow, Russia working on new methods in steroid synthesis under direction of Dr George Segal and Professor Igor Torgov. Aer several years of natural product research at the same Institute, he moved on to postdoctoral studies in stereoselective synthesis with Professor Wolfgang Oppolzer at the University of Geneva, Switzerland, and Professor James A. Marshall at the University of South Carolina. Boris joined Eisai in 1994, and has contributed to many oncology drug discovery projects with considerable focus on natural products as chemical leads, culminating in the discovery of Halaven.

PAPER

http://www.sciencedirect.com/science/article/pii/S0960894X0401100X

Volume 14, Issue 22, 15 November 2004, Pages 5551–5554

Macrocyclic ketone analogues of halichondrin B

This paper is dedicated to memory of Bruce F. Wels, our friend and colleague
  • a Department of Medicinal Chemistry, Eisai Research Institute, 4 Corporate Drive, Andover, MA 01810, USA
  • b Department of Anticancer Research, Eisai Research Institute, 4 Corporate Drive, Andover, MA 01810, USA
  • c Advisory Board, Eisai Research Institute, 4 Corporate Drive, Andover, MA 01810, USA

Image for unlabelled figure

PAPER

From micrograms to grams: scale-up synthesis of eribulin mesylate

*Corresponding authors
aEisai Inc., Andover, USA
E-mail: Melvin_Yu@eisai.com
Nat. Prod. Rep., 2013,30, 1158-1164

DOI: 10.1039/C3NP70051H, http://pubs.rsc.org/is/content/articlelanding/2013/np/c3np70051h#!divAbstract

Covering: 1993 to 2002

The synthesis of eribulin mesylate from microgram to multi-gram scale is described in thisHighlight. Key coupling reactions include formation of the C30a to C1 carbon–carbon bond and macrocyclic ring closure through an intramolecular Nozaki–Hiyama–Kishi reaction.

Graphical abstract: From micrograms to grams: scale-up synthesis of eribulin mesylate

The synthesis of the C27–C35 tetrahydrofuran fragment.

The synthesis of the C14–C21 aldehyde subfragment.

CLIP

In 1986, two Japanese chemists Hirata and Uemura [Y. Hirata, D. Uemura, Pure Appl. Chem. 58 (1986) 701.] isolated a naturally-occurring compound from the marine sponge Halichondria okadai (picture above, right). The compound was named Halichondrin B, and it immediately began to generate great excitement when it was realised that it was extremely potent at killing certain types of cancer cells in small-scale tests. As a result of this discovery, it was immediately given top priority to be tested against a wide range of other cancers, and became one of the first compounds to be evaluated using the novel 60-cell line method developed by the US National Cancer Institute (NCI). In this technique, 60 different types of human tumor cells (including leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney) are tested with the potential anti-cancer molecule delivered at a single dose of 10 μM concentration. This process can be run in parallel, with dozens of different molecules being tested against all 60 cancer cell lines at the same time in a huge array. Any molecules which exhibit significant growth inhibition are prioritised, and the test repeated on them, but this time at five different concentration levels.

Halichondrin B
Halichondrin B – the part of the molecule used to make Eribulin is shown in blue.

Unfortunately, the concentration of Halichondrin B in the sea sponge wasn’t enough to enable commercial production for use in chemotherapy. For example, a ton of sea sponges could only produce 300 mg of Halichondrin B! The race was on to try to synthesise Halichondrin B in the lab, which wasn’t easy due to its large size (molecular weight 1110) and complex structure. However, only 6 years later, chemists at Harvard University published the complete chemical synthesis of this molecule………..T.D. Aicher, K.R. Buszek, F.G. Fang, C.J. Forsyth, S.H. Jung, Y. Kishi, M.C. Matelich, P.M. Scola, D.M. Spero, S.K. Yoon, J. Am. Chem. Soc. 114 (1992) 3162

Although this was a great achievement, Halichondrin B was still far too complex and the sythesis route too expensive to do on a large scale. The molecule needed to be stripped down to its essential components, while keeping, or even improving, its anti-cancer efficacy. Many tests were performed, but eventually the work led to te development of the structurally-simplified and pharmaceutically-optimized analog, which was named Eribulin [3,4]. Eribulin mesylate was approved by the U.S. Food and Drug Administration in 2010, to treat patients with metastatic breast cancer [5], and it is currently being marketed by Eisai Co. under the trade nameHalaven . It is also being investigated for use in a variety of other solid tumors, including lung cancer, prostate cancer and sarcoma .

EribulinERIBULIN

M.J. Towle, K.A. Salvato, J. Budrow, B.F. Wels, G. Kuznetsov, K.K. Aalfs, S. Welsh, W. Zheng, B.M. Seletsk, M.H. Palme, G.J. Habgood, L.A. Singer, L.V. Dipietro, Y. Wang, J.J. Chen, D.A. Quincy, A. Davis, K. Yoshimatsu, Y. Kishi, M.J. Yu, B.A. Littlefield, Cancer Res. 61 (2001) 1013.

M.J. Yu, Y. Kishi, B.A. Littlefield, in D.J. Newman, D.G.I. Kingston, G.M. Cragg, Anticancer agents from natural products, Washington, DC, Taylor and Francis (2005).

http://healthmad.com/conditions-and-diseases/breast-cancer-cure-from-the-sea/

http://www.clinicaltrials.gov/ct2/results?term=eribulin+OR+E7389

M.A. Jordan, L. Wilson, Nature Revs: Cancer 4 (2004) 253.

ERIBULIN

Patent Data

Appl No Prod No Patent No Patent
Expiration
Drug Substance
Claim
Drug Product
Claim
Patent Use
Code
Delist
Requested
N201532 001 6214865 Jul 20, 2023 Y
N201532 001 6469182 Jun 16, 2019 U – 1096
N201532 001 7470720 Jun 16, 2019 Y
N201532 001 8097648 Jan 22, 2021 U – 1096

Exclusivity Data

Appl No Prod No Exclusivity Code Exclusivity Expiration
N201532 001 NCE Nov 15, 2015

The substance inhibits the polymerization of tubulin into microtubules and encapsulates tubulin molecules in non-productive aggregates from. The lack of training of the spindle apparatus blocks the mitosis and ultimately induces apoptosis of the cell. Eribulin differs from known microtubule inhibitors such as taxanes and vinca alkaloids by the binding site on microtubules, also it does not affect the shortening. This explains the effectiveness of the new cytostatic agent in taxane-resistant tumor cell lines with specific tubulin mutations.

Structure and mechanism

Structurally, eribulin is a fully synthetic macrocyclic ketone analogue of the marine sponge natural product halichondrin B,[4][5] the latter being a potent naturally-occurring mitotic inhibitor with a unique mechanism of action found in the Halichondria genus of sponges.[6][7] Eribulin is a mechanistically-unique inhibitor of microtubule dynamics,[8][9] binding predominantly to a small number of high affinity sites at the plus ends of existing microtubules.[10] Eribulin exerts its anticancer effects by triggering apoptosis of cancer cells following prolonged and irreversible mitotic blockade.[11][12]

A new synthetic route to E7389 was published in 2009.[13]

clip

Eisai R&D Management Co., Ltd.

13/9/2013

Halaven is a novel anticancer agent discovered and developed in-house by Eisai and is currently approved in more than 50 countries, including Japan, the United States and in Europe. In Russia, Halaven was approved in July 2012 for the treatment of locally advanced or metastatic breast cancer previously treated with at least two chemotherapy regimens including an anthracycline and a taxane. Approximately 50,000 women in Russia are newly diagnosed with breast cancer each year, with this type of cancer being the leading cause of death in women aged 45 to 55 years. read all at…………………….

http://www.dddmag.com/news/2013/09/eisai-launches-halaven-cancer-drug-russia

Eribulin mesylate (Halaven; Eisai) — a synthetic analogue of the marine natural product halichondrin B that interferes with microtubule dynamics — was approved in November 2010 by the US Food and Drug Administration for the treatment of metastatic breast cancer.

Family members of the product patent, WO9965894, have SPC protection in the EU until 2024 and one of its Orange Book listed filings, US8097648, has US154 extension till January 2021.

The drug also has NCE exclusivity till November 2015.

HALAVEN (eribulin mesylate) Injection is a non-taxane microtubule dynamics inhibitor. Eribulin mesylate is a synthetic analogue of halichondrin B, a product isolated from the marine sponge Halichondria okadai. The chemical name for eribulin mesylate is 11,15:18,21:24,28-Triepoxy-7,9-ethano12,15-methano-9H,15H-furo[3,2-i]furo[2′,3′:5,6]pyrano[4,3-b][1,4]dioxacyclopentacosin-5(4H)-one, 2[(2S)-3-amino-2-hydroxypropyl]hexacosahydro-3-methoxy-26-methyl-20,27-bis(methylene)-, (2R,3R,3aS,7R,8aS,9S,10aR,11S,12R,13aR,13bS,15S,18S,21S,24S,26R,28R,29aS)-, methanesulfonate (salt).

It has a molecular weight of 826.0 (729.9 for free base). The empirical formula is C40H59NO11 •CH4O3S. Eribulin mesylate has the following structural formula:

HALAVEN® (eribulin mesylate) Structural Formula Illustration

HALAVEN is a clear, colorless, sterile solution for intravenous administration. Each vial contains 1 mg of eribulin mesylate as a 0.5 mg/mL solution in ethanol: water (5:95).

Full-size image (23 K)

Full-size image (15 K)

complete syn is available here

http://www.sciencedirect.com/science/article/pii/S0968089611010674

http://www.drugdevelopment-technology.com/projects/halaven-cancer/halaven-cancer1.html

Nitrogen: dark blue, oxygen: red, hydrogen: light blue
graphics: Wurglics, Frankfurt am Main

clip

Macrocyclization process for preparing a macrocyclic intermediate of halichondrin B analogs, in particular eribulin, from a non-macrocyclic compound, using a carbon-carbon bond-forming reaction.

http://www.pnas.org/content/108/17/6699/F1.expansion.html

http://www.nature.com/nrd/journal/v8/n1/fig_tab/nrd2487_F6.html

UPDATED

WO 2015066729

Eisai has developed and launched eribulin mesylate for treating breast cancer.  Follows on from WO2014208774, claiming use of a combination comprising eribulin mesylate and lenvatinib mesylate, for treating cancer.

Macrocyclization reactions and intermediates useful in the synthesis of analogs of halichondrin B

By: Fang, Francis G.; Kim, Dae-Shik; Choi, Hyeong-Wook; Chase, Charles E.; Lee, Jaemoon

Assignee: Eisai R&D Management Co., Ltd., Japan

The invention provides methods for the synthesis of eribulin or a pharmaceutically acceptable salt thereof (e.g., eribulin mesylate) through a macrocyclization strategy.  The macrocyclization strategy of the present invention involves subjecting a non-macrocyclic intermediate to a carbon-carbon bond-forming reaction (e.g., an olefination reaction (e.g., Horner-Wadsworth-Emmons olefination), Dieckmann reaction, catalytic Ring-Closing Olefin Metathesis, or Nozaki-Hiyama-Kishi reaction) to afford a macrocyclic intermediate.  The invention also provides compds. useful as intermediates in the synthesis of eribulin or a pharmaceutically acceptable salt thereof and methods for prepg. the same.

CLIPS

http://www.chemistry-blog.com/2012/09/15/from-natural-product-to-pharmaceutical/

In a recent discussion (Nicolau), about the suggested move of Prof. NicoIau from Scripps, the issue of the practicality of natural product total synthesis was raised. Here is a wonderful example of just that very usefulness, a wonderful piece of science extending over many years. It concerns the journey from Halichondrin B to Eribulin (E7389) a novel anti-cancer drug. The two compounds have the following structures:

I think you can see the relationship and as a development chemist I am glad they managed to simplify things (a bit).

Both compounds have an enormous number of possible isomers: Halichondrin B, with 32 stereocenters has 232possible isomers; Eribulin has 19 with 219 isomers (if I have counted correctly, it does not really matter, there are lots of isomers). Remarkable is the fact that only one of these isomers is active in the given area of anti-cancer agents.

An excellent review of the biology and chemistry of these compounds has been published by Phillips etal1. This review is an excellent read and is to be commended. Another one written by Kishi2, is also full of information about the discovery of E7389 and I hope you will all get a chance to read this chapter.

The history of Halichondrin B goes back to 1987 when Blunt2-5 isolated it with other similar compounds from extraction of 200Kg of a sponge. Independently Pettit isolated the same compound from a different species4. The appearance of this compound in different species of sponge may indicate that it is produced by a symbiote.

The biological activity of Halichondrin B is amazing. When evaluated against B-16 melanoma cells it was found to have an IC50 of 0.093ng/mL. Against various cancers, generated in mice, it was shown to be affective at a daily dose of 5ug/kg, which resulted in a doubling of the survival rate. It has also been demonstrated that Halichondrin acts as a microtubule destabiliser and mitoitic spindle poison. It was proven that it is has tremendous in vivo activity against a variety of drug resistant cancers, lung, colon, breast, ovarian to mention a few. Consequently the National Cancer Institute selected it for pre-clinical trials and it’s here that the problems began. According to reference 1 the entire clinical development would require some 10g, and if successful the annual production amount would be between 1-5 kg. Blunt and co-workers managed to isolate 310mg from 1000kg-harvested sponge therefore, the only way to obtain the amounts required is total chemical synthesis. But synthesising 1-5 kg of such a compound would indeed be a mammoth task.

Kishi synthesised this compound7 in 1992 starting from carbohydrate precursors employing the Nozaki-Hiyama-Kishi Ni/Cr reaction, several times, in the long synthetic sequence8, 9. Now as an aside I have used this reaction on scale several times and although it works well its success is very dependant upon the quality of the chromium source and also the presence of other trace transition metals.

In collaboration with Eisai work on the SAR of Halichondrin began. They had a good start: Thanks to the total syntheses of Kishi several advanced intermediates were available for biological screening and one popped out of the screen as being very active:

The first active lead compound

As one can see the complete left hand side of Halichondrin has gone! However, this compound was not active in vivo. Many derivatives and analogues of this compound were prepared: furans, diols, ketones and so on and a lead emerged from this complex SAR study, ER-076349. The vicinal diol was used as a handle for further refinement and lead ultimately to E7389, the clinical candidate.

It can be synthesised in around 35 steps from simple starting materials.

Going through all this work in a few sentences really belittles the tremendous amount of effort that went into discovery and development of this compound and the people involved are to be applauded for their dedication.

Kishi continues to optimise the synthesis of Eribulin as judged by a recent publication10. Where he describes an approach to the amino-alcohol-tetrahydrofuran part of Eribulin (top left fragment, compound 1 below). The retro-synthetic analysis is shown below. The numbering corresponds to that of Eribulin.

The first generation synthesis consisted of 20 steps and delivered compound 1 about 5% yield, the second-generation route was completed in 12 steps with a yield of 48%. One of the highlights includes a remarkable asymmetric hydrogenation11 with Crabtree’s catalyst12:

This selectivity was not just luck; it seems to quite general, at least in this system. I always wonder how long it took them to stumble across this catalyst, but then I suppose that Eisai like most of the large pharma. companies has a hydrogenation group that probably indulges in catalyst screening.

The C34-C35 diol was obtained by a Sharpless asymmetric hydroxylation, here the diastereoisomeric ratio was not very high, only about 3:1 in favour of the desired isomer. Fortunately the undesired isomer could be removedcompletely by crystallisation.

This is a remarkable story and references 1 and 2 are worth reading to obtain the complete picture and learn lots of new chemistry as well. Eisai filed a NDA and the FDA approved the compound in 2010 for the treatment of metastatic breast cancer.

Patent

https://www.google.com/patents/WO2013142999A1?cl=en

EXAM PLE 23 : Preparation of Eribulin :

Figure imgf000049_0001

[00120] Compound E-12A (133 mg, 160 μηιοΙ, 1.0 eq) was dissolved in anhydrous dichloromethane (20 mL) and cooled to 0 °C. To this solution was sequentially added 2,6-lutidine (0.09 m L, 0.8 mmol, 5.0 eq), and trimethyl silyl triflate (TMSOTf) (0.12 m L, 0.64 mmol, 4.0 eq) and the cooling bath was removed . The reaction was stirred at room temperature for 1.5 hours and another portion of 2,6-lutidine (5.0 eq) and TMSOTf (4.0 eq) were added at room temperature. The reaction was further stirred for 1 hour and quenched with water (10 m L). The layers were separated and the organic phase was washed with additional water (2x 10 m L), brine (10 m L), dried over MgS04 and concentrated under reduced pressure. The residue was dissolved in MeOH (10 m L), a catalytic amount of K2C03 was added at room temperature and the resulting mixture was stirred for 2 hours. The reaction was diluted with dichloromethane and quenched with water (10 mL). The layers were separated and the aqueous phase was further extracted with dichloromethane (5 x 10 m L). The combined organic layers were washed with brine (20 m L), dried over MgS04, filtered and concentrated. The residue was dissolved in dichloromethane and purified by column chromatography on silica gel, using 1 : 9 MeOH : CH2CI2 to 1 : 9 : 90 N H4OH : MeOH : CH2CI2 as eluent. The product was afforded as a white amorphous solid (103 mg, 88%) . [00121] EXAMPLE 23 : Preparation of compound of formula 4a

Figure imgf000050_0001

D-Gulonolactone 4a

[00122] The compound of formula 4a was prepared from D-Gulonolactone according to the conditions described in PCT publication number WO 2005/118565. [00123] EXAMPLE 24: Preparation of Eribulin mesylate (3)

[00124] Eribulin mesylate (3) was prepared from Eribulin according to the conditions described in US patent application publication number US

2011/0184190.

PATENT

https://www.google.com/patents/EP2528914A1?cl=en

Halichondrin B analogs, e.g., eribulin or pharmaceutically acceptable salts thereof, can be synthesized from the C14-C35 fragment as described in U.S. Patent No. 6,214,865 and International Publication No. WO 2005/118565. In one example described in these references, the C14-C35 portion, e.g., ER- 804028, of the molecule is coupled to the C1-C13 portion, e.g., ER-803896, to produce ER-804029, and additional reactions are carried out to produce eribulin (Scheme 1):

Figure imgf000022_0001

Scheme 1

eribulin, eribulin mesylate

Scheme 2

ER-804028

Figure imgf000042_0001

Compound AE (280 mg, 0.281 mmol, 1 eq) was dissolved in CH2C12 and cooled to 0 °C. Pyridine (0.045 ml, 0.56 mmol, 2.0 eq) was added followed by Ms20 (58.8 mg, 0.338 mmol, 1.20 eq). The reaction was allowed to warm to room temperature, and stirring was continued for an additional 1 h. The reaction mixture was cooled to 0 °C, diluted with MTBE (5.6 ml), washed with saturated NaHC03 (0.84 g), and concentrated to give crude product as colorless film. The crude was azeotropically dried with heptane (3 ml χ 2) and re-dissolved in THF (7.0 ml). The mixture was cooled to 0 °C and treated with 25 wt% NaOMe (0.13 ml). After 10 min, the reaction was allowed to warm to room temperature, and stirring was continued for an additional 30 min. The mixture was treated with additional 25 wt% NaOMe (0.045 ml), and stirring was continued for an additional 20 min. The reaction mixture was diluted with heptane (7.0 ml) and washed with water (1.4 ml). The organic layer was separated, sequentially washed with: 1) 20 wt% NH4C1 (0.84 g) and 2) 20 wt% NaCl (3 g), and concentrated to give crude product as brownish oil. The crude was purified by Biotage (Uppsala, Sweden) 12M (heptane-MTBE 2:3 v/v) to give ER-804028 (209 mg, 0.245 mmol, 87%) as pale yellow oil. 1H NMR (400 MHz, CDC13): δ 7.89 (2H, m), 7.64 (IH, m), 7.56 (2H, m), 4.85 (IH, d, J= 1.6 Hz), 4.80 (IH, s), 4.72 (IH, s), 4.61 (IH, d, J= 1.6 Hz), 4.23 (IH, br), 3.91 (IH, m), 3.79 (IH, m), 3.76 (2H, m), 3.63 (IH, m), 3.50-3.60 (4H, m), 3.43 (IH, dd, J= 5.6 Hz, 10.0 Hz), 3.38 (3H, s), 3.32 (IH, m), 2.98 (2H, m), 2.61 (IH, br), 2.56 (IH, m), 2.50 (IH, m), 2.08-2.22 (3H, m), 1.96 (IH, m), 1.84 (IH, m), 1.78 (IH, m), 1.70 (IH, m), 1.42-1.63 (6H, m), 1.28-1.42 (2H, m), 1.01 (3H, d, J= 6.8 Hz), 0.84 (18H, s), 0.05 (3H, s), 0.04 (3H, s), 0.00 (3H, s), -0.01 (3H, s); and 13C NMR (100 MHz, CDC13): δ 150.34, 150.75, 139.91, 134.18, 129.73 (2C), 128.14 (2C), 105.10, 85.97, 80.92, 79.72, 78.50, 77.45, 77.09, 75.53, 71.59, 68.04, 62.88, 58.27, 57.73, 43.51, 42.82, 39.16, 37.68, 35.69, 33.31, 32.41, 31.89, 31.48, 29.79, 26.21 (3C), 26.17 (3C), 18.58, 18.38, 18.13, -3.85, – 4.71, -5.12 (2C).

CLIP

Eribulin mesylate (Halaven)
Eribulin is a highly potent cytotoxic agent approved in the US for the treatment of metastatic breast cancer for patients who have
received at least two previous chemotherapeutic regimens.30 Eribulin was discovered and developed by Eisai and it is currently
undergoing clinical evaluation for the treatment of sarcoma (PhIII) and non-small cell lung cancer which shows progression after platinum-based chemotherapy and for the treatment of prostate cancer (PhII). Early stage clinical trials are also underway to evaluate
eribulin’s efficacy against a number of additional cancers. Eribulin is a structural analog of the marine natural product halichondrin B.
Its mechanism of action involves the disruption of mitotic spindle formation and inhibition of tubulin polymerization which results
in the induction of cell cycle blockade in the G2/M phase and apoptosis.31 Several synthetic routes for the preparation of eribulin have
been disclosed,32–35 each of which utilizes the same strategy described by Kishi and co-workers for the total synthesis of halichondrin B.36 Although the scales of these routes were not disclosed in all cases, this review attempts to highlight what appears to be the production-scale route based on patent literature.37,38 Nonetheless, the synthesis of eribulin represents a significant accomplishment in the field of total synthesis and brings a novel chemotherapeutic option to cancer patients.
The strategy to prepare eribulin mesylate (V) employs a convergent synthesis featuring the following: the late stage coupling of
sulfone 22 and aldehyde 23 followed by macrocyclization under Nozaki–Hiyami–Kishi coupling conditions, formation of a challenging
cyclic ketal, and installation of the primary amine (Scheme 5).Sulfone 22 was further simplified to aldehyde 24 and vinyl triflate 25 which were coupled through a Nozaki–Hiyami–Kishi reaction.

STR1 STR2
The schemes that follow will describe the preparation of fragments 23, 24 and 25 along with how the entire molecule was assembled.
The synthesis of the C1–C13 aldehyde fragment 23 is described in Scheme 6. L-Mannonic acid-lactone 26 was reacted with cyclohexanone in p-toluene sulfonic acid (p-TSA) to give the biscyclohexylidene ketal 27 in 84% yield. Lactone 27 was reduced with
diisobutylaluminum hydride (DIBAL-H) to give lactol 28 followed by condensation with the ylide generated from the reaction of
methoxymethylene triphenylphosphorane with potassium tertbutoxide to give a mixture of E and Z vinyl ethers 29 in 81% yield.
Dihydroxylation of the vinyl ether of 29 using catalytic osmium teteroxide and N-methylmorpholine-N-oxide (NMO) with concomitant cyclization produced diol 30 in 52% yield. Bis-acetonide 30 was then reacted with acetic anhydride in acetic acid in the presence of ZnCl2 which resulted in selective removal of the pendant ketal protecting group. These conditions also affected peracylation, giving rise to tetraacetate 31 in 84% yield. Condensation of 31 with methyl 3-(trimethylsilyl)pent-4-enoate in the presence of boron trifluoride etherate in acetonitrile provided alkene 32. Saponification conditions using Triton B(OH) removed the acetate protecting groups within 32 and presumably induced isomerization of the alkene into conjugation with the terminal ester, triggering an intramolecular Michael attack of the 2-hydroxyl group, ultimately resulting in the bicylic-bispyranyl diol methyl ester 33 as a crystalline solid in 38% yield over two steps. Oxidative cleavage of the vicinal diol of 33 with sodium periodate gave aldehyde 34 which was coupled to (2-bromovinyl)trimethylsilane under Nozaki–Hiyami–Kishi conditions to give an 8.3:1 mixture of allyl alcohols 35 in 65% yield over two steps. Hydrolysis of the cyclohexylidine ketal 35 with aqueous acetic acid followed by recrystallization gave diastereomerically pure triol 36 which was reacted with tert-butyldimethylsilyl triflate (TBSOTf) to afford the tris-TBS ether 37 in good yield. Vinyl silane 37 was treated with NIS and catalytic tert-butyldimethylsilyl chloride (TBSCl) to give vinyl iodide 38 in 90% yield.
Reduction of the ester with DIBAL-H produced the key C1–C14 fragment 23 in 93% yield.
The preparation of the tetra-substituted tetrahydrofuran intermediate 24 is described in Scheme 7. D-Glucurono-6,3-lactone
39 was reacted with acetone and sulfuric acid to give the corresponding acetonide and the 5-hydroxyl group was then removed by converting it to its corresponding chloride through reaction with sulfuryl chloride (SO2Cl2) followed by hydrogenolysis
to give lactone 40 in good overall yield. Reduction of the lactone 40 with DIBAL-H gave the corresponding lactol which was condensed
with (trimethylsilyl)methylmagnesium chloride to afford silane 41. Elimination of the silyl alcohol of 41 was accomplished
under Peterson conditions with potassium hexamethyldisilazide (KHMDS) to afford the corresponding terminal alkene in 94% yield.
The secondary alcohol of this intermediate was alkylated with benzyl bromide to afford ether 42 in 95% yield. Asymmetric dihydroxylation of the alkene of 42 under modified Sharpless conditions using potassium osmate (VI) dehydrate (K2OsO4), potassium
ferricyanide (K3Fe(CN)6) and the (DHQ)2AQN ligand produced the vicinal diol which was then reacted with benzoyl chloride,
N-methylmorpholine, and DMAP to give di-benzoate 43 in excellent yield as a 3:1 mixture of diastereomeric alcohols. Allyl trimethylsilane was added to the acetal of 43 using TiCl3(OiPr) as the Lewis acid to give 44 in 83% yield. Re-crystallization of 44 from
isopropanol and n-heptane afforded 44 in >99.5% de in 71% yield.
Oxidation of the secondary alcohol of 44 under the modified Swern conditions generated the corresponding ketone which was condensed with the lithium anion of methyl phenyl sulfone to give a mixture of E and Z vinyl sulfones 45. Debenzylation of 45 using iodotrimethylsilane (TMSI) followed by chelation-controlled reduction of the vinyl sulfone through reaction with NaBH(OAc)3, and
then basic hydrolysis of the benzoate esters using K2CO3 in MeOH resulted in triol 46 as a white crystalline solid in 57% yield over the
five steps after re-crystallization. The vicinal diol of 46 was protected as the corresponding acetonide through reaction with 2,2-
dimethoxypropane and sulfuric acid and this was followed by methyl iodide-mediated methylation of the remaining hydroxyl
group to give methyl ether 47. The protecting groups within acetonide 47 were then converted to the corresponding bis-tert-butyldimethylsilyl ether by first acidic removal of the acetonide with aqueous HCl and reaction with TBSCl in the presence of imidazole to give bis-TBS ether 48. Then, ozonolysis of the olefin of 48 followed by hydrogenolysis in the presence of Lindlar catalyst afforded the key aldehyde intermediate 24 in 68% yield over the previous five steps after re-crystallization from heptane.
Two routes to the C14–C26 fragment 25 will be described as both are potentially used to prepare clinical supplies of eribulin.
The first route features a convergent and relatively efficient synthesis of 25, however it is limited by the need to separate enantiomers
and mixture of diastereomers via chromatographic methods throughout the synthesis.37 The second route to 25 is a
much lengthier synthesis from a step-counting perspective; however it takes full advantage of the chiral pool of starting materials
and requires no chromatographic separations and all of the products were carried on as crude oils until they could be isolated as
crystalline solids.38 The first route to fragment 25 is described in Scheme 8 and was initiated by the hydration of 2,3-dihydrofuran (49) using an aqueous suspension of Amberlyst 15 to generate the intermediate tetrahydro-2-furanol (50) which was then immediately reacted with 2,3-dibromopropene in the presence of tin and catalytic HBr to afford diol 51 in 45% for the two steps.

The primary alcohol of 51 was selectively protected as its tert-butyldiphenylsilyl ether using TBDPSCl and imidazole and the racemate was then separated using simulated moving bed (SMB) chromatography to give enantiopure 52 in 45% yield over the two steps. The secondary alcohol of 52 was reacted with p-toluenesulfonyl chloride and DMAP to give tosylate 53 in 78% yield which was used as a coupling partner later in the synthesis of this fragment. The synthesis of the appropriate coupling partner was initiated by condensing diethylmalonate with (R)-2-(3-butenyl)oxirane (54), followed by decarboxylation to give lactone 55 in 71% yield for the two step process. Methylation of the lactone with LHMDS and MeI provided 56 in 68% yield as a 6:1 mixture of diastereomers. The lactone 56 was reacted with the aluminum amide generated by the reaction of AlMe3 and N,O-dimethylhydroxylamine to give the corresponding Weinreb amide which was protected as its tert-butyldimethylsilyl ether upon reaction with TBSCl and imidazole to give 57 in 91% yield over the two steps. Dihydroxylation of the olefin of 57 by reaction with OsO4 and NMO followed by oxidative cleavage with NaIO4 gave the desired coupling partner aldehyde 58 in 93% yield. Aldehyde 58 was coupled with vinyl bromide 53 using an asymmetric Nozaki–Hiyami– Kishi reaction using CrCl2, NiCl2, Et3N and chiral ligand 66 (described in Scheme 9 below). The reaction mixture was treated with ethylene diamine to remove the heavy metals and give the secondary alcohol 59. This alcohol was stirred with silica gel in isopropanol to affect intramolecular cyclization to give the tetrahydrofuran 60 in 48% yield over the three step process. The Weinreb amide of 60 was reacted with methyl magnesium chloride to generate the corresponding methyl ketone which was converted to vinyl triflate 61 upon reaction with KHMDS and Tf2NPh. De-silylation of the primary and secondary silyl ethers with methanolic HCl gave the corresponding diol in 85% yield over two steps and the resulting mixture of diastereomers was separated using preparative HPLC to provide the desired diastereomer in 56% yield. The primary alcohol was protected as its pivalate ester with the use of pivaloyl chloride, DMAP and collidine; the secondary alcohol was converted     to a mesylate upon treatment with methanesulfonyl chloride (MsCl) and Et3N to give the C15–C27 fragment 25 in high yield.
The preparations of the chiral ligand 66 used in the coupling reaction in Scheme 8 along with the chiral ligand 67 utilized later
in the synthesis are described in Scheme 9. 2-Amino-3-methylbenzoic acid (62) was reacted with triphosgene to give benzoxazine
dione 63 in 97% yield, which then was reacted with either D- or L-valinol in DMF followed by aqueous LiOH to give alcohols 64
and 65, respectively in 65–75% yield for the two steps. Reaction of alcohol 64 or 65 with MsCl in the presence of DMAP effected formation of the dihydrooxazole ring and mesylation of the aniline to give the corresponding (R)-ligand 66 derived from D-valinol or the (S)-ligand 67 derived from L-valinol, respectively in high yield.
An alternative route to intermediate 25 is described in Scheme  10 and although much lengthier than the route described in
Scheme 8, it avoids chromatographic purifications as all of the products are carried on crude until a crystalline intermediate
was isolated and purified by re-crystallization. Quinic acid (68) was reacted with cyclohexanone in sulfuric acid to generate a protected
bicyclic lactone in 73% yield and the resulting tertiary alcohol was protected as its trimethylsilyl ether 69. Reduction of the
lactone 69 was accomplished with DIBAL-H and the resulting lactol  was treated with acetic acid to remove the TMS group and the resulting compound was reacted with acetic anhydride, DMAP and Et3N to give bis-acetate 70 in 65% yield for the three steps after re-crystallization. Methyl 3-(trimethylsilyl)pent-4-enoate was coupled to the acetylated lactol 70 in the presence of boron trifluoride etherate and trifluoroacetic anhydride to give adduct 71 in 62% yield. The acetate of 71 was removed upon reaction with sodium methoxide in methanol and the resulting tertiary alcohol cyclized on to the isomerized enone alkene to give the fused pyran ring. Reduction of the methyl ester with lithium aluminum hydride provided pyranyl alcohol 72. Mesylation of the primary alcohol was followed by displacement with cyanide anion to give nitrile 73.STR1 STR2

The nitrile was methylated upon reaction with KHMDS and MeI and the resulting product was purified by re-crystallization
to provide nitrile 74 in 66% over the previous five steps in a 34:1 diastereomeric ratio. Acid hydrolysis of the ketal of 74 liberated
the corresponding diol in 72% yield and this was reacted with 2-acetoxy-2-methylpropionyl bromide to give bromo acetate 75.
Elimination of the bromide was accomplished upon treatment with 1,8-diazabicycloundec-7-ene (DBU) to give alkene 76 in 63%
yield for two steps. Ozonolysis of the cyclohexene ring followed by reductive work-up with NaBH4 and basic hydrolysis of the acetate
produced a triol which upon reaction with NaIO4 underwent oxidative cleavage to give cyclic hemiacetal 77 in 75% yield over
the previous four steps. Wittig condensation with carbomethoxymethylene triphenylphosphorane gave the homologated unsaturated
ester 78. Catalytic hydrogenation of the alkene using PtO2 as the catalyst was followed by converting the primary alcohol to the
corresponding triflate prior to displacement with sodium iodide resulted in iodide 79 in 75% yield over four steps. The ester of 79
was reduced to the corresponding primary alcohol upon reaction with LiBH4 in 89% yield and the resulting iodoalcohol was treated
with Zn dust to affect reductive elimination of the iodide and decomposition of the pyran ring system to give the tetrahydrofuran
diol 80 in 90% yield. This diol was treated with methanolic HCl to affect an intramolecular Pinner reaction and this was followed
by protection of the primary alcohol as its tert-butyldiphenylsilyl ether to give lactone 81 The lactone was reacted with the
aluminum amide generated from AlMe3 and N,O-dimethylhydroxylamine and the resulting secondary alcohol was protected as
its tert-butyldimethylsilyl ether to give Weinreb amide 82 in 99% crude yield over four steps. Compound 82 is the diastereomerically
pure version of compound 60 and can be converted to compound 25 by the methods described in Scheme 8 absent the required
HPLC separation of diastereomers. With the three key fragments completed, the next step was to assemble them and complete the synthesis of eribulin. Aldehyde 24 was coupled to vinyl triflate 25 using an asymmetric Nozaki– Hiyami–Kishi reaction using CrCl2, NiCl2, Et3 N and chiral ligand 67 (Scheme 9) to give alcohol 83 (Scheme 11).

STR4

Formation of the THP ring was accomplished by reaction with KHMDS which allowed for displacement of the mesylate with the secondary alcohol and provided the THP containing product in 72% yield for the three steps. The pivalate ester group was removed with DIBAL-H to give the western fragment 22 in 92% yield.
The completion of the synthesis of eribulin is illustrated in Scheme 12. The lithium anion of sulfone 22 generated upon reaction
with nBuLi was coupled to aldehyde 23 to give diol 84 in 84% yield. Both of the alcohol functional groups of 84 were oxidized
using a Dess–Martin oxidation in 90% yield and the resulting sulfone was removed via a reductive cleavage upon reaction with
SmI2 to give keto-aldehyde 85 in 85% yield. Macrocyclization of 85 was accomplished via an asymmetric Nozaki–Hiyami–Kishi
reaction using CrCl2, NiCl2, Et3N and chiral ligand 67 to give alcohol 86 in 70% yield. Modified Swern oxidation of the alcohol provided the corresponding ketone in 91% yield and this was followed by removal of the five silyl ether protecting groups upon reaction with TBAF and subsequent cyclization to provide ketone 87. Compound 87 was treated with PPTS to provide the ‘caged’ cyclic ketal 88 in 79% over two steps. The vicinal diol of 88 was reacted with Ts2O in collidine to affect selective tosylation of the primary alcohol and this crude product was reacted with ammonium hydroxide to install the primary amine to give eribulin which was treated
with methanesulfonic acid in aqueous ammonium hydroxide to give eribulin mesylate (V) in 84% yield over the final three steps.

STR1  STR2 STR3

30. Zheng, W.; Seletsky, B. M.; Palme, M. H.; Lydon, P. J.; Singer, L. A.; Chase, C. E.;
Lemelin, C. A.; Shen, Y.; Davis, H.; Tremblay, L.; Towle, M. J.; Salvato, K. A.;
Wels, B. F.; Aalfs, K. K.; Kishi, Y.; Littlefield, B. A.; Yu, M. J. Bioorg. Med. Chem.
Lett. 2004, 14, 5551.
31. Wang, Y.; Serradell, N.; Bolós, J.; Rosa, E. Drugs Future 2007, 32, 681.
32. Chiba, H.; Tagami, K. J. Synth. Org. Chem. Jpn. 2011, 69, 600.
33. Choi, H.; Demeke, D.; Kang, F.-A.; Kishi, Y.; Nakajima, K.; Nowak, P.; Wan, Z.-
K.; Xie, C. Pure Appl. Chem. 2003, 75, 1.
34. Kishi, Y.; Fang, F.; Forsyth, C. J.; Scola, P. M.; Yoon, S. K. WO 9317690 A1, 1993.
35. Littlefield, B. A.; Palme, M.; Seletsky, B. M.; Towle, M. J.; Yu, M. J.; Zheng, W.
WO 9965894 A1, 1999.
36. Aicher, T. D.; Buszek, K. R.; Fang, F. G.; Forsyth, C. J.; Jung, S. H.; Kishi, Y.;
Matelich, M. C.; Scola, P. M.; Spero, D. M.; Yoon, S. K. J. Am. Chem. Soc. 1992,
114, 3162.
37. Austad, B.; Chase, C. E.; Fang, F. G. WO 2005118565 A1, 2005.
38. Chase, C.; Endo, A.; Fang, F. G.; Li, J. WO 2009046308 A1, 2009.

CLIP

http://www.rsc.org/chemistryworld/2015/06/longest-organic-syntheses-natural-product

Eribulin (Halaven)

Halichondrin B is a wicked molecule. In tests in mice, it is an extremely potent cancer cell killer, active at around 80 picomolar concentration. It also possesses a fiendish macrocyclic polyketide structure, with 32 stereocentres meaning that it could adopt over four billion different isomers – with just one that fights cancer.

Eribulin and halichondrin BEribulin is a cut-down derivative of halichondrin B, which maintains most of its activity with significantly reduced complexity

Its power is therefore inherently hard to harness. Halichondrin B was found in various sea sponge species in the 1980s, but getting 400mg  of the compound from a tonne of sponge was doing well. Clinical development required at least 10g, and annual production takes kilograms.

Although developing a synthetic route to halichondrin B looked just as tough as trying to extract it from sponges, Yoshito Kishi’s group at Harvard University in the US accepted the challenge. Frank Fang, one of the team, recalls how the Nozaki–Hiyama–Kishi (NHK) coupling reaction would prove critical. ‘Another feature that was impressed upon me was the importance of crystalline intermediates,’ Fang adds. These allowed simple purification by recrystallisation, rather than expensive and time-consuming chromatography.

Published in 1992, their method used several NHK couplings, forming carbon–carbon bonds between multifunctional vinyl halides and aldehydes via a nickel-catalysed, chromium-mediated process.4 The sprawling convergent synthesis, whose longest linear sequence involved 47 steps, prompted Japanese pharmaceutical company Eisai to collaborate with Kishi in exploring halichondrin B’s structure–activity relationship. On screening the team’s intermediates, one featuring the macrocyclic half of halichondrin B proved especially active. A series of medicinal chemistry refinements led to what would eventually becomeeribulin (marketed by Eisai as Halaven), promising a slightly simpler synthesis. It has ‘just’ 19 stereocentres, which along with other structural restrictions cuts the possible number of isomers to a mere 16,384.

Fang joined Eisai in 1998 as it selected eribulin for further development, and worked to develop a production process for a route that produced it from three fragments. He again strove to exploit recrystallisation and use the NHK reaction, although making it reliable enough for manufacturing was challenging. ‘There was an appreciation for the somewhat sensitive nature of the reaction, particularly the asymmetric variant,’ he recalls.

The Eisai researchers therefore studied the NHK procedure as they applied it to redesigning the synthesis for part of the eribulin molecule they refer to as the C14–C26 fragment. Featuring just one ring, this fragment isn’t the most structurally complex of the three, but is still very difficult to make. That’s because it is a long chain with several stereocentres, whose stereochemistry is hard to link together.

Fang’s team initially broke this section down into two sub-fragments, C14–C19 and C20–C26, using asymmetric NHK reactions on each, learning about the reaction’s parameters as they did so.5 They then used what they’d found out to devise NHK reactions linking the two sub-fragments and attaching the two fragments on either side, which included closing the eribulin macrocycle. ‘We gained knowledge through our studies on the C19–C20 NHK coupling and were ultimately able to utilise that knowledge to try to execute an asymmetric NHK reaction in fixed equipment on multi-kilogram scale and construct the C19–C20, C26–C27, and C13–C14 bonds,’ Fang explains.

Synthesis of eribulin Synthesis of eribulin relies heavily on Nozaki–Hiyama–Kishi (NHK) coupling reactions to make key C–C bonds

Halaven was approved in the US in 2010 to treat breast cancer and earned ¥2.89 billion in sales (£159 million) in 2014. The commercial route initially took 62 steps across a convergent synthesis bringing together three fragments, with a longest linear sequence of 30 steps. Fang’s team has since added seven steps to the C14–C26 fragment route, which counterintuitively cuts costs and waste by 80% by eliminating chromatography.6 ‘I am hopeful that we can find the lessons applicable in future work,’ Fang says.

Cheaper synthesis would appear welcome, given that Halaven’s price tag has been criticised. In the UK it currently costs £2,000 per 21 day treatment cycle according to data from the British National Formularyand the country’s National Institute for Health and Clinical Excellence (Nice). As a result, Nice refused to cover the drug, and in January 2015 the remaining funding in England looked set to be closed off with Halaven being taken off the Cancer Drugs Fund (CDF)’s list. But Eisai was told in March that the drug would stay on the list, pending reconsideration, after an appeal against the decision.

In defence, Fang claims that Halaven is actually one of the most affordable breast cancer treatments on the CDF. ‘Eisai was given no opportunity to lower the price of Halaven before NHS England announced that the treatment would be removed from the fund, despite this being something we were, and still are, very willing to do,’ he adds.

Cited Patent Filing date Publication date Applicant Title
WO2009124237A1 * Apr 3, 2009 Oct 8, 2009 Eisai R&D Management Co., Ltd. Halichondrin b analogs
US6214865 * Jun 16, 1999 Apr 10, 2001 Eisai Co., Ltd. Macrocyclic analogs and methods of their use and preparation
Reference
1 * DONG, C.-G. ET AL.: “New Syntheses of E7389 C 14-C35 and Halichondrin C 14- C38 Building Blocks: Reductive Cyclization and Oxy-Michael Cyclization Approaches“, J. AM. CHEM. SOC., vol. 131, 2009, pages 15642 – 15646, XP002629056
2 * See also references of EP2831082A4
3 * ZHENG, W. ET AL.: “Macrocyclic ketone analogues of halichondrin B“, BIOORG. MED. CHEM. LETT., vol. 14, 2004, pages 5551 – 5554, XP004598592
Citing Patent Filing date Publication date Applicant Title
WO2015000070A1 * May 30, 2014 Jan 8, 2015 Alphora Research Inc. Synthetic process for preparation of macrocyclic c1-keto analogs of halichondrin b and intermediates useful therein including intermediates containing -so2-(p-tolyl) groups
WO2015066729A1 * Nov 4, 2014 May 7, 2015 Eisai R&D Management Co., Ltd. Macrocyclization reactions and intermediates useful in the synthesis of analogs of halichondrin b
WO2015131286A1 * Mar 6, 2015 Sep 11, 2015 Alphora Research Inc. Crystalline derivatives of (s)-1-((2r,3r,4s,5s)-5-allyl-3-methoxy-4-(tosylmethyl)tetrahydrofuran-2-yl)-3-aminopropan-2-ol
CN103483352A * Oct 18, 2013 Jan 1, 2014 李友香 Medicinal bulk drug for resisting tumors
US9062020 Dec 24, 2012 Jun 23, 2015 Alphora Research Inc. 2-((2S,3S,4R,5R)-5-((S)-3-amino-2-hydroxyprop-1-yl)-4-methoxy-3-(phenylsulfonylmethyl)tetrahydrofuran-2-yl)acetaldehyde derivatives and process for their preparation
US9174956 Dec 14, 2012 Nov 3, 2015 Alphora Research Inc. Process for preparation of 3-((2S,5S)-4-methylene-5-(3-oxopropyl)tetrahydrofuran-2-yl)propanol derivatives and intermediates useful thereof
US9181152 Nov 29, 2012 Nov 10, 2015 Alphora Research Inc. Process for preparation of (3R)-2,4-di-leaving group-3-methylbut-1-ene
WO2012129100A1 * Mar 16, 2012 Sep 27, 2012 Eisai R&D Management Co., Ltd. Methods and compositions for predicting response to eribulin
WO2012166899A2 * May 31, 2012 Dec 6, 2012 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of thyroid and kidney cancer subjects to lenvatinib compounds
CA2828946A1 * Apr 16, 2012 Oct 26, 2012 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
US7982060 * Jun 3, 2005 Jul 19, 2011 Eisai R&D Management Co., Ltd. Intermediates for the preparation of analogs of Halichondrin B
P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

References

  1. ^“FDA approves new treatment option for late-stage breast cancer” (Press release). USFDA. 2010-11-15. Retrieved November 15, 2010.
  2. ^Notice of Decision for HALAVEN
  3. ^http://www.clinicaltrials.gov/ct2/results?term=eribulin+OR+E7389
  4. ^ Towle MJ, Salvato KA, Budrow J, Wels BF, Kuznetsov G, Aalfs KK, Welsh S, Zheng W, Seletsky BM, Palme MH, Habgood GJ, Singer LA, Dipietro LV, Wang Y, Chen JJ, Quincy DA, Davis A, Yoshimatsu K, Kishi Y, Yu MJ, Littlefield BA (February 2001). “In vitro and in vivo anticancer activities of synthetic macrocyclic ketone analogues of halichondrin B”. Cancer Res.61 (3): 1013–21. PMID11221827.
  5. ^ Yu MJ, Kishi Y, Littlefield BA (2005). “Discovery of E7389, a fully synthetic macrocyclic ketone analogue of halichondrin B”. In Newman DJ, Kingston DGI, Cragg, GM. Anticancer agents from natural products. Washington, DC: Taylor & Francis. ISBN0-8493-1863-7.
  6. ^ Hirata Y, Uemura D (1986). “Halichondrins – antitumor polyether macrolides from a marine sponge”. Pure Appl. Chem.58 (5): 701–710. doi:10.1351/pac198658050701.
  7. ^ Bai RL, Paull KD, Herald CL, Malspeis L, Pettit GR, Hamel E (August 1991). “Halichondrin B and homohalichondrin B, marine natural products binding in the vinca domain of tubulin. Discovery of tubulin-based mechanism of action by analysis of differential cytotoxicity data”. J. Biol. Chem.266 (24): 15882–9. PMID1874739.
  8.  Jordan MA, Kamath K, Manna T, Okouneva T, Miller HP, Davis C, Littlefield BA, Wilson L (July 2005). “The primary antimitotic mechanism of action of the synthetic halichondrin E7389 is suppression of microtubule growth”. Mol. Cancer Ther.4 (7): 1086–95. doi:10.1158/1535-7163.MCT-04-0345. PMID16020666.
  9.  Okouneva T, Azarenko O, Wilson L, Littlefield BA, Jordan MA (July 2008). “Inhibition of Centromere Dynamics by Eribulin (E7389) during Mitotic Metaphase”. Mol. Cancer Ther.7 (7): 2003–11. doi:10.1158/1535-7163.MCT-08-0095. PMC2562299. PMID18645010.
  10.  Smith JA, Wilson L, Azarenko O, Zhu X, Lewis BM, Littlefield BA, Jordan MA (February 2010). “Eribulin Binds at Microtubule Ends to a Single Site on Tubulin to Suppress Dynamic Instability”. Biochemistry49 (6): 1331–7. doi:10.1021/bi901810u. PMC2846717. PMID20030375.
  11. Kuznetsov G, Towle MJ, Cheng H, Kawamura T, TenDyke K, Liu D, Kishi Y, Yu MJ, Littlefield BA (August 2004). “Induction of morphological and biochemical apoptosis following prolonged mitotic blockage by halichondrin B macrocyclic ketone analog E7389”. Cancer Res.64 (16): 5760–6. doi:10.1158/0008-5472.CAN-04-1169. PMID15313917.
  12. ^ Towle MJ, Salvato KA, Wels BF, Aalfs KK, Zheng W, Seletsky BM, Zhu X, Lewis BM, Kishi Y, Yu MJ, Littlefield BA (January 2011). “Eribulin induces irreversible mitotic blockade: implications of cell-based pharmacodynamics for in vivo efficacy under intermittent dosing conditions”. Cancer Res.71 (2): 496–505. doi:10.1158/0008-5472.CAN-10-1874. PMID21127197.
  13. ^ Kim DS, Dong CG, Kim JT, Guo H, Huang J, Tiseni PS, Kishi Y (November 2009). “New syntheses of E7389 C14-C35 and halichondrin C14-C38 building blocks: double-inversion approach”. J. Am. Chem. Soc.131 (43): 15636–41. doi:10.1021/ja9058475. PMID19807076.

SEE          https://wordpress.com/post/newdrugapprovals.org/3955

Eribulin
Eribulin.svg
Systematic (IUPAC) name
2-(3-Amino-2-hydroxypropyl)hexacosahydro-3-methoxy- 26-methyl-20,27-bis(methylene)11,15-18,21-24,28-triepoxy- 7,9-ethano-12,15-methano-9H,15H-furo(3,2-i)furo(2′,3′-5,6) pyrano(4,3-b)(1,4)dioxacyclopentacosin-5-(4H)-one
Clinical data
Trade names Halaven
AHFS/Drugs.com Consumer Drug Information
MedlinePlus a611007
License data
Pregnancy
category
  • US: D (Evidence of risk)
Routes of
administration
Intravenous
Legal status
Legal status
Identifiers
CAS Number 253128-41-5 
ATC code L01XX41 (WHO)
PubChem CID 17755248
ChemSpider 21396142 Yes
UNII LR24G6354G Yes
ChEMBL CHEMBL1237028 
Chemical data
Formula C40H59NO11
Molar mass 729.90 g/mol

////////Halaven, ERIBULIN, anticancer drug ,  Eisai Co.  E7389,  ER-086526,  US NCI designation,  NSC-707389.   breast cancer,  liposarcoma, halichrondrin B analog, B1939, E7389, ER-086526, 441045-17-6, FDA 2010, 253128-41-5 , ERIBULIN MESYLATE, Antineoplastic, エリブリンメシル酸塩

CC1CC2CCC3C(=C)CC(O3)CCC45CC6C(O4)C7C(O6)C(O5)C8C(O7)CCC(O8)CC(=O)CC9C(CC(C1=C)O2)OC(C9OC)CC(CN)O.CS(=O)(=O)O

C[C@@H]1C[C@@H]2CC[C@H]3C(=C)C[C@@H](O3)CC[C@]45C[C@@H]6[C@H](O4)[C@H]7[C@@H](O6)[C@@H](O5)[C@@H]8[C@@H](O7)CC[C@@H](O8)CC(=O)C[C@H]9[C@H](C[C@H](C1=C)O2)O[C@@H]([C@@H]9OC)C[C@@H](CN)O.CS(=O)(=O)O

C[C@@H]1C[C@@H]2CC[C@H]3C(=C)C[C@@H](O3)CC[C@]45C[C@@H]6[C@H](O4)[C@H]7[C@@H](O6)[C@@H](O5)[C@@H]8[C@@H](O7)CC[C@@H](O8)CC(=O)C[C@H]9[C@H](C[C@H](C1=C)O2)O[C@@H]([C@@H]9OC)C[C@@H](CN)O.CS(=O)(=O)O

CREDIT

http://www.chm.bris.ac.uk/motm/eribulin/eribulinh.htm

253128-41-5  CAS

Eribulin


Filed under: FDA 2010 Tagged: 253128-41-5, 441045-17-6, anticancer drug, Antineoplastic, エリブリンメシル酸塩, B1939, E7389, Eisai Co. E7389, ER-086526, ERIBULIN, eribulin mesylate, FDA 2010, Halaven, halichrondrin B analog, liposarcoma, NSC-707389. breast cancer, US NCI designation

Lovastatin

$
0
0

Lovastatin3Dan.gifLovastatin.svg

Lovastatin
(+)-Mevinolin
(1S,3R,7S,8S,8aR)-1,2,3,7,8,8a-Hexahydro-3,7-dimethyl-8-[2-[(2R,4R)-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl]ethyl]-1-naphthalenyl (S)-2-Methylbutyrate
(2S)-2-Methylbutanoic acid (1S,3R,7S,8S,8aR)-1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-[2-[(2R,4R)-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl]ethyl]-1-naphthalenyl ester
[1S-[1a(R*),3a,7b,8b(2S*,4S*),8ab]]-2-Methylbutanoic Acid1,2,3,7,8,8a-Hexahydro-3,7-dimethyl-8-[2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl]-1-naphthalenyl Ester
1,2,6,7,8,8a-Hexahydro-b,d-dihydroxy-2,6-dimethyl-8-(2-methyl-1-oxobutoxy)-1-naphthaleneheptanoic Acid d-Lactone
2b,6a-Dimethyl-8a-(2-methyl-1-oxobutoxy)mevinic Acid Lactone
6a-Methylcompactin
75330-75-5

Lovastatin (Merck’s Mevacor) is a statin drug, used for lowering cholesterol (hypolipidemic agent) in those withhypercholesterolemia to reduce risk of cardiovascular disease. Lovastatin is a naturally occurring compound found in food such asoyster mushrooms,[2] red yeast rice,[3] and Pu-erh.[4]

Medical uses

The primary uses of lovastatin is for the treatment of dyslipidemia and the prevention of cardiovascular disease.[5] It is recommended to be used only after other measures, such as diet, exercise, and weight reduction, have not improved cholesterol levels.[5]

Pleurotus ostreatus, the oyster mushroom, naturally contains up to 2.8% lovastatin on a dry weight basis.[15]

Structure

 

 

History

 Compactin and lovastatin, natural products with a powerful inhibitory effect on HMG-CoA reductase, were discovered in the 1970s, and taken into clinical development as potential drugs for lowering LDL cholesterol.

However, in 1980, trials with compactin were suspended for undisclosed reasons (rumoured to be related to serious animal toxicity). Because of the close structural similarity between compactin and lovastatin, clinical studies with lovastatin were also suspended, and additional animal safety studies initiated.

In 1982 some small-scale clinical investigations of lovastatin, a polyketide derived natural product isolated from Aspergillus terrus, in very high-risk patients were undertaken, in which dramatic reductions in LDL cholesterol were observed, with very few adverse effects. After the additional animal safety studies with lovastatin revealed no toxicity of the type thought to be associated with compactin, clinical studies resumed.

Large-scale trials confirmed the effectiveness of lovastatin. Observed tolerability continued to be excellent, and lovastatin was approved by the US FDA in 1987.

Lovastatin at its maximal recommended dose of 80 mg daily produced a mean reduction in LDL cholesterol of 40%, a far greater reduction than could be obtained with any of the treatments available at the time. Equally important, the drug produced very few adverse effects, was easy for patients to take, and so was rapidly accepted by prescribers and patients. The only important adverse effect is myopathy/rhabdomyolysis. This is rare and occurs with all HMG-CoA reductase inhibitors.

 Mechanism of action

Lovastatin is an inhibitor of 3-hydroxy-3methylglutaryl-coenzyme A reductase (HMG-CoA reductase), an enzyme which catalyzes the conversion of HMG-CoA to mevalonate. Mevalonate is a required building block for cholesterol biosynthesis and lovastatin interferes with its production by acting as a competitive inhibitor for HMG-CoA which binds to the HMG-CoA reductase. Lovastatin, being inactive in the native form, the form in which it is administered, is hydrolysed to the β-hydroxy acid form in the body and it is this form which is active. Presumably, the reductase acts on the hydrolyzed lovastatin to reduce the carboxylic acid moiety.

Discovery, Biochemistry and Biology

 It is now generally accepted that a major risk factor for the development of coronary heart disease is an elevated concentration of plasma cholesterol, especially lowdensity lipoprotein (LDL) cholesterol. The objective is to decrease excess levels of cholesterol to an amount consistent with maintainence of normal body function. Cholesterol is biosynthesized in a series of more than 25 separate enzymatic reactions that initially involves 3 successive condensations of acetyl-CoA units to form a 6-carbon compound, 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA). This is reduced to mevalonate and then converted in a series of reactions to the isoprenes that are building blocks of squalene, the immediate precursor to sterols, which cyclizes to lanosterol (a methylated sterol) and further metabolized to cholesterol. A number of early attempts to block the synthesis of cholesterol resulted in agents that inhibited late in the biosynthetic pathway between lanosterol and cholesterol. A major rate limiting step in the pathway is at the level of the microsomal enzyme which catalyzes the conversion of HMG CoA to mevalonic acd and which has been considered to be a prime target for pharmacologic intervention for several years.

            HMG CoA reductase occurs early in the biosynthetic pathway and is among the first commited steps to cholesterol formulation. Inhibition of this enzyme could lead to accumulation of HMG CoA, a water-soluble intermediate that is then capable of being readily metabolized that is then capable of being readily metabolized to simpler molecules. This inhibition of reductase would nto lead to accumulation of lipophylic intermediates having a formal sterol ring.

            Lovastatin is the first specific inhibitor of HMG CoA reductase to receive approval for the treatment of hypercholesterolemia. The first breakthrough in efforts to find a potent, specific, competitive inhibitor of HMG CoA reductase occurred in 1976 when Endo et al reported discovery of mevastatin, a highly functionalized fungal metabolite, isolated from cultures of  Penicillium citrium. Mevastatin was demonstrated to be an unusually potent inhibitor of the target enzyme and of cholesterol biosynthesis. Subsequent to the first reports describing mevastatin, efforts were initiated to search for other naturally occurring inhibitors oh HMG CoA reductase. This led to the discovery of a novel fungal metabolite – Lovastatin. The structure of Lovastatin was determined to be different from that of mevastatin by the presence of a 6 alphamethyl group in the hexahydronaphthalene ring.

Key points from the study of the Biosynthesis of Lovastatin :-

– Lovastatin is comprised of 2 polyketide chains derived from acetate that are 8- and 4-

  carbons long coupled in head to tail fashion.

– 6 alphamethyl group and the methyl group on the 4-carbon side chain are derived from

  the methyl group of methionine, and

– 6 alphamethyl group is added before closure of the rings.

This implies that lovastatin is a unique compound synthesized by A. terreus and that mevastatin is not an intermediate in its fornmation.

Cholesterol Biosynthetic Pathway

 

The HMG CoA reductase reaction

 

Biosynthesis — Diels-Alder Catalyzed Cyclization

            In vitro formation of a triketide lactone using a genetically-modified protein derived from 6-deoxyerythronolide B synthase has been demonstrated. The stereochemistry of the molecule supports the intriguing idea that an enzyme-catalyzed Diels-Alder reaction may occur during assembly of the polyketide chain. It thus appears that biological Diels-Alder reactions may be triggered by generation of reactive triene systems on an enzyme surface.

 

Biosynthesis – Using Broadly specific Acyltransferase

It has been found that a dedicated acyltransferase, LovD, is encoded in the lovastatin biosynthetic pathway. LovD has a broad substrate specificity towards the acyl carrier, the acyl substrate and the decalin acyl acceptor. It efficiently catalyzes the acyl transfer from coenzyme A thoesters or N-acetylcysteamine (SNAC) thioesters to monacolin J.

            The biosynthesis of Lovastatin is coordinated by two iterative type I polyketide synthases and numerous accessory enzymes. Nonketide, the intermediate biosynthetic precursor of Lovastatin, is assembeled by the upstream megasynthase LovB (also known as lovastatin nonaketide synthase), enoylreductase LovC, and CYP450 oxygenases. The five carbon unit side chain is synthesized by LovF (also known as lovastatin diketide synthase) through a single condensation diketide undergoes methylation and reductive tailoring by the individual LovF catalytic domains to yield an α-S-methylbutyryl thioester covalently attached to the phosphopantetheine arm on the acyl carrier protein (ACP) domain of LovF. Encoded in the gene cluster is a 46kDa protein, LovD, which was initially identified as an esterase homolog. LovD, which was initially identified as an esterase homolog. LovD was suggested to catalyze the last step of lovastatin biosynthesis that regioselectively transacylates the acyl group from LovF to the C8 hydroxyl group of the Nonaketide to yield Lovastatin. 

 

  

K. Auclair, A. Sutherland, J. Kennedy, D. J. Witter, J. P. Van den Heever, C. R. Hutchinson and J. C. Vederas, Lovastatin Nonaketide Synthase Catalyses An Intramolecular Diels-Alder Reaction Of A Substrate Analogue, J. Am. Chem. Soc., 2000, 122, 11519-11520. DOI: 10.1021/ja003216+

JACS(Lov2)

http://pubs.rsc.org/en/content/articlelanding/2013/np/c2np20069d/unauth#!divAbstract

196264.fig.002

http://www.hindawi.com/journals/bmri/2012/196264/#B30

  1. Z. Jia, X. Zhang, Y. Zhao, and X. Cao, “Enhancement of lovastatin production by supplementing polyketide antibiotics to the submerged culture of Aspergillus terreus,” Applied Biochemistry and Biotechnology, vol. 160, no. 7, pp. 2014–2025, 2010. 

Patent

https://www.google.com/patents/US6307066

PATENT

https://www.google.com/patents/WO2002009697A1?cl=en

https://www.google.com/patents/EP0625208B1?cl=en

Total Synthesis

A major bulk of work in the synthesis of Lovastatin was done by M. Hirama in the 1980’s. Hirama synthesized Compactin and used one of the intermediates to follow a different path to get to Lovastatin. The synthetic sequence is shown in the schemes below. The γ-lactone was synthesized using Yamada methodology starting with aspartic acid. Lactone opening was done using lithium methoxide in methanol and then silylation to give a separable mixture of the starting lactone and the silyl ether. The silyl ether on hydrogenolysis followed by Collins oxidation gave the aldehyde. Stereoselective preparation of (E,E)-diene was accomplished by addition of trans-crotyl phenyl sulfone anion, followed by quenching with Ac2O and subsequent reductive elimination of sulfone acetate. Condensation of this with Lithium anion of dimethyl methylphosphonate gave compound 1.Compound 2 was synthesized as shown in the scheme in the synthetic procedure. Compounds 1 and 2 were then combined together using 1.3eq sodium hydride in THF followed by reflux in chlorobenzene for 82 hrs under nitrogen to get the enone 3.

Simple organic reactions were used to get to Lovastatin as shown in the scheme.

 

 

 

 

Pharmacopoeia Information

Lovastatin tablets are preserved in well closed, light resistant containers. Protected from light and stored either in a cool place or at controlled room temperature.

Lovastatin tablets are tested for Dissolution and Assay as per the USP.

Limit for Dissolution – Not less than 80% (Q) of the labeled amount of Lovastatin is dissolved in 30 mins.

Limit for Assay – Each tablet contains not less than 90% and not more than 110% of the labeled amount of Lovastatin, tested by HPLC analysis.

Lovastatin raw material contains 5 impurities – A, B, C, D and E (as shown below).

 

 

Market brands and other analogues  

There are other derivatives of Lovastatin which possess cholesterol reducing activity. Simvastatin (Zocor®) is another statin closely related to Lovastatin, differing only by the presence of a methyl group in the butanoyl ester moiety. Both effective in lowering total cholesterol.

Another statin having vastly different structure but a popular drug – Atorvastatin (Lipitor®), administered as a calcium salt is a pyrrole derivative and a synthetic compound rather than a natural product.

NMR

  1 H NMR spectrum of lovastatin, 300 MHz, solvent CDCl 3 . 

STR1 str2 STR3

STR1 str2 STR3UV LOVASTATIN

Figure 6. The mean FT-IR spectra (the calibration set) and variables selected after application of UVE-PLS for modelling lovastatin (triangles) and wavenumbers for characteristic peaks for lovastatin IR spectrum (dots).

PATENT

https://www.google.com/patents/EP0702679B1?cl=en

Lovastatin is produced as a secondary metabolite of the fungusAspergillus terreus (US 4,231,938) deposited in American Type Culture Collection under Nos. ATCC 20541, ATCC 20542, and Monascus ruberdeposited in Fermentation Research Institute Agency of Industrial Science and Technology, Ministry of International Trade and Industry, Japan (DE 30 06 216 A1) under No. Ferm 4822. Other kinds of microorganisms producing lovastatin are known as well, e.g. a mutant of the microorganism Aspergillus terreus andAspergillus oryzae marked ATCC 74135.

Lovastatin is chemically 1′,2′,6′,7′,8a’-hexahydro-3,5-dihydroxy-2′,6′-dimethyl-8′-2″-methyl-1″-oxobutoxy)-1-naphtalene heptanoic acid-5-lactone (Stubbs et al., 1986) of the formula (EP 0 033 537 A1)

Figure 00010001

An active form of lovastatin is also an acid, which is chemically 1,2,6,8,8a-hexahydro-β,δ-dihydroxy-1-naphtalene heptanoic acid (Alberts et al., 1980) of the formula (EP 0 022 478 A1)

Figure 00020001

The lactone form of lovastatin is used as an agent for reducing cholesterol level in blood (Scott M.G. and Vega G.L, 1985). It inhibits the biosynthesis of mevalonic acid by inhibition of 3-hydroxy-3-methylglutaryl A reductase coenzyme (HMG-CoA reductase, E.C. 1.1.1.34) (Zubay et al., 1984).

Prior Art

After the completed fermentation, lovastatin is present in the broth in the lactone form (compound I) and in the acid form (compound II). In the isolation process as disclosed in EP 0 033 536 A2, lovastatin is extracted from the broth with ethyl acetate. The extract is concentrated by vacuum distillation. Since lovastatin is present in the lactone form as well as in the acid form and only the lactone is of commercial interest, the acid form should be converted into the lactone. The lactonisation is carried out by the reflux of the concentrate in toluene at 106 °C for 2 hours. After the lactonisation is complete, the solution is concentrated to a small volume. A pure substance is obtained by means of purifying the concentrate on columns packed with silica gel, in the presence of solvents such as ethyl acetate or n-hexane. The collected fractions are again concentrated in vacuo and then pure lovastatin crystallizes in the lactone form.

Due to the sophisticated multi-step procedure and vigorous conditions applied during the isolation, the yields of lovastatin are generally low. Different solvents, which in part exhibit toxicity, are used such as benzene, toluene, acetonitrile or ethyl acetate. Hence working with these solvents endangers the health of the persons involved and poses a problem with respect to the environment.

https://www.google.com/patents/EP0702679B1?cl=en

The structure was confirmed by IR spectroscopy (Fig.1), mass spectroscopy (Fig. 2), NMR (Fig. 3) and UV spectroscopy (Fig. 4).

STR1 str2

IR spectrum of lovastatin.IR spectrum of lovastatin.

 Lovastatin

Title: Lovastatin
CAS Registry Number: 75330-75-5
CAS Name: (2S)-2-Methylbutanoic acid (1S,3R,7S,8S,8aR)-1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-[2-[(2R,4R)-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl]ethyl]-1-naphthalenyl ester
Additional Names: (1S,3R,7S,8S,8aR)-1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-[2-[(2R,4R)-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl]ethyl]-1-naphthalenyl (S)-2-methylbutyrate; 1,2,6,7,8,8a-hexahydro-b,d-dihydroxy-2,6-dimethyl-8-(2-methyl-1-oxobutoxy)-1-naphthaleneheptanoic acid d-lactone; 2b,6a-dimethyl-8a-(2-methyl-1-oxobutoxy)mevinic acid lactone; mevinolin; 6a-methylcompactin; monacolin K
Manufacturers’ Codes: MK-803
Trademarks: Lovalip (Merck & Co.); Mevacor (Merck & Co.); Mevinacor (Merck & Co.); Mevlor (Merck & Co.); Sivlor (Sidus)
Molecular Formula: C24H36O5
Molecular Weight: 404.54
Percent Composition: C 71.26%, H 8.97%, O 19.77%
Literature References: Fungal metabolite; potent inhibitor of HMG-CoA reductase, the rate controlling enzyme in cholesterol biosynthesis. Isoln from Monascus ruber: A. Endo, J. Antibiot. 32, 852 (1979); from Aspergillus terreus: R. L. Monaghan et al., US4231938 (1980 to Merck & Co.). Structure and biochemical properties: A. W. Alberts et al., Proc. Natl. Acad. Sci. USA 77, 3957 (1980). Total synthesis: M. Hirama, M. Iwashita, Tetrahedron Lett. 24, 1811 (1983). Review of syntheses: T. Rosen, C. H. Heathcock, Tetrahedron 42, 4909-4951 (1986). Biosynthesis: M. D. Greenspan, J. B. Yudkovitz, J. Bacteriol. 162, 704 (1985); R. N. Moore et al., J. Am. Chem. Soc. 107, 3694 (1985). HPLC determn in plasma and bile: R. J. Stubbs et al., J. Chromatogr. 383,438 (1986). Clinical pharmacology: S. M. Grundy, G. L. Vega, J. Lipid Res. 26, 1464 (1985). Clinical comparison with gemfibrozil,q.v.: M. J. Tikkanen et al., Am. J. Cardiol. 62, 35J (1988). Review of clinical experience: J. A. Tobert, Am. J. Cardiol. 62, 28J-34J (1988). Comprehensive description: G. S. Brenner et al., Anal. Profiles Drug Subs. Excip. 21, 277-305 (1992). Prevention of acute coronary events in men and women with average cholesterol levels: J. R. Downs et al., J. Am. Med. Ass

References

  1.  Neuvonen, PJ; Backman, JT; Niemi, M (2008). “Pharmacokinetic comparison of the potential over-the-counter statins simvastatin, lovastatin, fluvastatin and pravastatin.”. Clinical Pharmacokinetics. 47 (7): 463–74. doi:10.2165/00003088-200847070-00003. PMID 18563955.
  2.  . Gunde-Cimerman N; Cimerman A. (Mar 1995). “Pleurotus fruiting bodies contain the inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A reductase-lovastatin.”. Exp Mycol. 19 (1): 1–6.doi:10.1006/emyc.1995.1001. PMID 7614366.
  3. Jump up^ Liu J, Zhang J, Shi Y, Grimsgaard S, Alraek T, Fønnebø V (2006).“Chinese red yeast rice (Monascus purpureus) for primary hyperlipidemia: a meta-analysis of randomized controlled trials”.Chin Med. 1 (1): 4. doi:10.1186/1749-8546-1-4. PMC 1761143free to read.PMID 17302963.
  4. Jump up^ Zhao ZJ, Pan YZ, Liu QJ, Li XH (2013). “Exposure assessment of lovastatin in Pu-erh tea”. International Journal of Food Microbiology.164 (1): 26–31. doi:10.1016/j.ijfoodmicro.2013.03.018.PMID 23587710.
  5. ^ Jump up to:a b “Lovastatin”. The American Society of Health-System Pharmacists. Retrieved 3 April 2011.
  6. ^ Jump up to:a b “Mevacor, Altoprev (lovastatin) dosing, indications, interactions, adverse effects, and more”. Medscape Reference. WebMD. Retrieved 17 March 2014.
  7. ^ Jump up to:a b “Lovastatin”. MedlinePlus. U.S. National Library of Medicine. 15 June 2012. Retrieved 1 December 2012.
  8. Jump up^ “Lovastatin”. LactMed. U.S. National Library of Medicine. Retrieved 1 December 2012.
  9. Jump up^ Stöppler, Melissa. “Mevacor Side Effects Center”. RxList. Retrieved 1 December 2012.
  10. Jump up^ David G. Bailey, J. Malcolm, O. Arnold, J. David Spence (1998). “Grapefruit juice-drug interactions”. Br J Clin Pharmacol 46 (2): 101–110. doi:10.1046/j.1365-2125.1998.00764.x. PMC 1873672. PMID 9723817.
  11. Jump up^ Kantola T, Kivistö KT, Neuvonen PJ (Apr 1998). “Grapefruit juice greatly increases serum concentrations of lovastatin and lovastatin acid”. Clin Pharmacol Ther. 63 (4): 397–402. doi:10.1016/S0009-9236(98)90034-0. PMID 9585793.
  12. ^ Jump up to:a b Alberts AW (1998). “Discovery, biochemistry and biology of lovastatin”. The American Journal of Cardiology. 62 (15): 10J–15J.doi:10.1016/0002-9149(88)90002-1. PMID 3055919.
  13. Jump up^ Katz MS (2005). “Therapy insight: Potential of statins for cancer chemoprevention and therapy”. Nature Clinical Practice Oncology. 2(2): 82–9. doi:10.1038/ncponc0097. PMID 16264880.
  14. Jump up^ Rao S, Porter DC, Chen X, Herliczek T, Lowe M, Keyomarsi K (July 1999). “Lovastatin-mediated G1 arrest is through inhibition of the proteasome, independent of hydroxymethyl glutaryl-CoA reductase”. Proc. Natl. Acad. Sci. U.S.A. 96 (14): 7797–802.doi:10.1073/pnas.96.14.7797. PMC 22141free to read. PMID 10393901.
  15. Jump up^ Alarcón J, Aguila S, Arancibia-Avila P, Fuentes O, Zamorano-Ponce E, Hernández M (Jan–Feb 2003). “Production and purification of statins from Pleurotus ostreatus (Basidiomycetes) strains”. Z Naturforsch C. 58 (1–2): 62–4. doi:10.1515/znc-2003-1-211.PMID 12622228.
  16. Jump up^ Vederas JC, Moore RN, Bigam G, Chan KJ (1985). “Biosynthesis of the hypocholesterolemic agent mevinolin by Aspergillus terreus. Determination of the origin of carbon, hydrogen and oxygen by 13C NMR and mass spectrometry”. J Am Chem Soc. 107 (12): 3694–701.doi:10.1021/ja00298a046.
  17. Jump up^ Alberts AW, Chen J, Kuron G, Hunt V, Huff J, Hoffman C, Rothrock J, Lopez M, Joshua H, Harris E, Patchett A, Monaghan R, Currie S, Stapley E, Albers-Schonberg G, Hensens O, Hirshfield J, Hoogsteen K, Liesch J, Springer J (July 1980). “Mevinolin: a highly potent competitive inhibitor of hydroxymethlglutaryl-coenzyme A reductase and a cholesterol-lowering agent”. Proc Natl Acad Sci U S A. 77(7): 3957–61. doi:10.1073/pnas.77.7.3957. PMC 349746free to read.PMID 6933445.
  18. Jump up^ FDA Orange Book Detail for application N019643 showing approval for 20 mg tablets on Aug 31, 1987 and 40 mg tablets on Dec 14, 1988
  19. Jump up^ Endo, Akira (Oct 2004). “The origin of the statins”. Atheroscler. Suppl. 5 (3): 125–30.doi:10.1016/j.atherosclerosissup.2004.08.033. PMID 15531285.
  20. ^ Jump up to:a b Bobek P, Ozdín L, Galbavý S (1998). “Dose- and time-dependent hypocholesterolemic effect of oyster mushroom (Pleurotus ostreatus) in rats”. Nutrition. 14 (3): 282–6. doi:10.1016/S0899-9007(97)00471-1. PMID 9583372.
  21. Jump up^ Hossain S, Hashimoto M, Choudhury EK, et al. (July 2003). “Dietary mushroom (Pleurotus ostreatus) ameliorates atherogenic lipid in hypercholesterolaemic rats”. Clin Exp Pharmacol Physiol. 30(7): 470–5. doi:10.1046/j.1440-1681.2003.03857.x.PMID 12823261.
  22. Jump up^ Bobek P, Galbavý S (October 1999). “Hypocholesterolemic and antiatherogenic effect of oyster mushroom (Pleurotus ostreatus) in rabbits”. Nahrung. 43 (5): 339–42. doi:10.1002/(SICI)1521-3803(19991001)43:5<339::AID-FOOD339>3.0.CO;2-5.PMID 10555301.
  23. Jump up^ Opletal L, Jahodár L, Chobot V, et al. (December 1997). “Evidence for the anti-hyperlipidaemic activity of the edible fungus Pleurotus ostreatus”. Br. J. Biomed. Sci. 54 (4): 240–3. PMID 9624732.
  24. Jump up^ Bajaj M, Vadhera S, Brar AP, Soni GL (October 1997). “Role of oyster mushroom (Pleurotus florida) as hypocholesterolemic/antiatherogenic agent”. Indian J. Exp. Biol. 35(10): 1070–5. PMID 9475042.
  25. Jump up^ Bobek P, Ozdín L, Kuniak L, Hromadová M (March 1997). “[Regulation of cholesterol metabolism with dietary addition of oyster mushrooms (Pleurotus ostreatus) in rats with hypercholesterolemia]”.Cas. Lek. Cesk. (in Slovak). 136 (6): 186–90. PMID 9221192.
  26. Jump up^ Bobek P, Ozdín L, Kuniak L (August 1996). “Effect of oyster mushroom (Pleurotus Ostreatus) and its ethanolic extract in diet on absorption and turnover of cholesterol in hypercholesterolemic rat”.Nahrung. 40 (4): 222–4. doi:10.1002/food.19960400413.PMID 8810086.
  27. Jump up^ Bobek P, Ozdín O, Mikus M (1995). “Dietary oyster mushroom (Pleurotus ostreatus) accelerates plasma cholesterol turnover in hypercholesterolaemic rat”. Physiol Res. 44 (5): 287–91.PMID 8869262.
  28. Jump up^ Bobek P, Ozdin L, Kuniak L (1995). “The effect of oyster mushroom (Pleurotus ostreatus), its ethanolic extract and extraction residues on cholesterol levels in serum, lipoproteins and liver of rat”. Nahrung. 39(1): 98–9. doi:10.1002/food.19950390113. PMID 7898579.
  29. Jump up^ Bobek P, Ozdin L, Kuniak L (March 1994). “Mechanism of hypocholesterolemic effect of oyster mushroom (Pleurotus ostreatus) in rats: reduction of cholesterol absorption and increase of plasma cholesterol removal”. Z Ernahrungswiss. 33 (1): 44–50.doi:10.1007/BF01610577. PMID 8197787.
  30. Jump up^ Chorváthová V, Bobek P, Ginter E, Klvanová J (1993). “Effect of the oyster fungus on glycaemia and cholesterolaemia in rats with insulin-dependent diabetes”. Physiol Res. 42 (3): 175–9.PMID 8218150.
  31. Jump up^ Bobek P, Ginter E, Jurcovicová M, Kuniak L (1991). “Cholesterol-lowering effect of the mushroom Pleurotus ostreatus in hereditary hypercholesterolemic rats”. Ann. Nutr. Metab. 35 (4): 191–5.doi:10.1159/000177644. PMID 1897899.
  32. Jump up^ Khatun K, Mahtab H, Khanam PA, Sayeed MA, Khan KA (January 2007). “Oyster mushroom reduced blood glucose and cholesterol in diabetic subjects”. Mymensingh Med J. 16 (1): 94–9.doi:10.3329/mmj.v16i1.261. PMID 17344789.
  33. Jump up^ “FDA bans red yeast rice product” by Michael McCarthy, The Lancet, Volume 351, Issue 9116, Page 1637, 30 May 1998
  34. Jump up^ Cholesterol Treatment Upheld, The New York Times, February 18, 1999
  35. Jump up^ Coronary heart disease: MedLine Plus Medical Encyclopedia
  36. Jump up^ Endo, Akira; Kuroda M.; Tsujita Y. (December 1976). “ML-236A, ML-236B, and ML-236C, new inhibitors of cholesterogenesis produced by Penicillium citrinium”. Journal of Antibiotics (Tokyo). 29(12): 1346–8. doi:10.7164/antibiotics.29.1346. PMID 1010803.
  37. Jump up^ Witter, DJ; Vederas, JC (1996). “Putative Diels-Alder catalyzed cyclization during the biosynthesis of lovastatin”. J Org Chem. 61 (8): 2613–23. doi:10.1021/jo952117p. PMID 11667090.
  38. Jump up^ Hirama M, Vet M (1982). “A chiral total synthesis of compactin”. J. Am. Chem. Soc. 104 (15): 4251. doi:10.1021/ja00379a037.
  39. Jump up^ Hirama M, Iwashita; Iwashita, Mitsuko (1983). “Synthesis of (+)-Mevinolin starting from Naturally occurring building blocks and using an asymmetry inducing reaction”. Tetrahedron Lett. 24 (17): 1811–1812. doi:10.1016/S0040-4039(00)81777-3.
  40. Jump up^ Javernik S, Kreft S, Strukelj B, Vrecer F (2001). “Oxidation of lovastatin in the solid state and its stabilization with natural antioxidants”. Die Pharmazie. 56 (9): 738–40. PMID 11593996.
  41. Jump up^ Hartig K, Beck E (2005). “Assessment of lovastatin application as tool in probing cytokinin-mediated cell cycle regulation”. Physiologia Plantarum. 125 (2): 260–267. doi:10.1111/j.1399-3054.2005.00556.x.
Lovastatin
Lovastatin.svg
Lovastatin3Dan.gif
Systematic (IUPAC) name
(1S,3R,7S,8S,8aR)-8-{2-[(2R,4R)-4-Hydroxy-6-oxooxan-2-yl]ethyl}-3,7-dimethyl-1,2,3,7,8,8a-hexahydronaphthalen-1-yl (2S)-2-methylbutanoate
Clinical data
Trade names Mevacor
AHFS/Drugs.com Monograph
MedlinePlus a688006
Pregnancy
category
  • US: X (Contraindicated)
Routes of
administration
Oral
Legal status
Legal status
Pharmacokinetic data
Bioavailability <5%[1]
Protein binding >98%[1]
Metabolism Hepatic (CYP3A andCYP2C8 substrate)[1]
Biological half-life 2–5 hours[1]
Excretion Faeces (83%), urine (10%)[1]
Identifiers
CAS Number 75330-75-5 Yes
ATC code C10AA02 (WHO)
PubChem CID 53232
IUPHAR/BPS 2739
DrugBank DB00227 Yes
ChemSpider 48085 Yes
UNII 9LHU78OQFD Yes
KEGG D00359 Yes
ChEBI CHEBI:40303 Yes
ChEMBL CHEMBL503 Yes
Synonyms Monacolin K, Mevinolin
Chemical data
Formula C24H36O5
Molar mass 404.54 g/mol

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


Filed under: Uncategorized Tagged: Lovastatin

MICONAZOLE NITRATE , Миконазол , ミコナゾール硝酸塩

$
0
0

Miconazole            C18H14Cl4N2O    416.13             [22916478]

Miconazole Nitrate            C18H14Cl4N2O.HNO3              479.14             [22832877]

ミコナゾール硝酸塩 JP16
Miconazole Nitrate

C18H14Cl4N2O▪HNO3 : 479.14
[22832-87-7]

 

 

 

 

 

 

 


click on above image for clear view











MORE GRAPHS

13C






1D 1H, n/a spectrum for Miconazole

2D [1H,1H]-TOCSY  BELOW

2D [1H,1H]-TOCSY, n/a spectrum for Miconazole

1D DEPT90

1D DEPT90, n/a spectrum for Miconazole

1D DEPT135

1D DEPT135, n/a spectrum for Miconazole

 

2D [1H,13C]-HSQC

2D [1H,13C]-HSQC, n/a spectrum for Miconazole

2D [1H,13C]-HMBC

2D [1H,13C]-HMBC, n/a spectrum for Miconazole

2D [1H,1H]-COSY

2D [1H,1H]-COSY, n/a spectrum for Miconazole

2D [1H,13C]-HMQC

2D [1H,13C]-HMQC, n/a spectrum for Miconazole
Miconazole is an imidazole antifungal agent, developed by Janssen Pharmaceutica, commonly applied topically to the skin or tomucous membranes to cure fungal infections. It works by inhibiting the synthesis of ergosterol, a critical component of fungal cell membranes. It can also be used against certain species of Leishmania protozoa which are a type of unicellular parasites that also contain ergosterol in their cell membranes. In addition to its antifungal and antiparasitic actions, it also has some antibacterialproperties. It is marketed in various formulations under various brand names.

Miconazole is also used in Ektachrome film developing in the final rinse of the Kodak E-6 process and similar Fuji CR-56 process, replacing formaldehydeFuji Hunt also includes miconazole as a final rinse additive in their formulation of the C-41RA rapid access color negative developing process.
It is on the World Health Organization’s List of Essential Medicines, the most important medications needed in a basic health system.[1]

ALTERNATIVE ROUTES beginning with the racemic raw material will likely be more costly or more time-consuming to develop, Cox says. Crystallization might be tricky because the stereogenic center does not have a group that can readily undergo acid-base chemistry. Catalytic asymmetric chemistry will necessitate converting the raw material to an appropriate substrate and identifying effective, as well as usable, chemical catalysts or biocatalysts.
What happens to the unwanted enantiomer also depends on the economics. Reracemizing and feeding the racemate back into the process is ideal but not always practical. In the miconazole case, the raw material costs $32 per kg. It is unlikely that reracemizing would be less costly in this example, Cox explains.
People should not forget that the goal of chiral technologies–enantiopure product–also may be achieved with chemistry that already exists, notes David R. Dodds, founder of Dodds & Associates LLC, Manlius, N.Y., a consulting service for biotechnology and chemical companies. Process chemists seek the most robust, most productive, and least expensive synthetic route and aim to find it as fast as possible. Any reaction that can help reach this goal is useful. It is the overall process cost that will dictate which reactions will be used. And that cost covers not only reagents but also waste streams, utilities, equipment use, unit operations, and downstream requirements. Thus, it may be more commercially attractive to replace an elegant but expensive single reaction with several more mundane ones that have a lower total cost, he says. Such a situation is likely to arise when an asymmetric step requires an expensive chiral catalyst or chiral auxiliary.

Brief background information

 

Salt ATC Formula MM CAS
A01AB09 
A07AC01 
D01AC02 
G01AF04 
J02AB01 
S02AA13
18 H 14 Cl 4 N 2 O 416.14 g / mol 22916-47-8
mononitrate A01AB09 
A07AC01 
D01AC02 
G01AF04 
J02AB01 
S02AA13
18 H 14 Cl 4 N 2 O ⋅ HNO 3 479.15 g / mol 22832-87-7

Using

 

  • antifungal agent for topical use
  • antimycotic agent

Classes substance

 

  • Imidazoles, 1- (hlorfenetil) imidazoles

synthesis Way

 

Synthesis of a)

trade names

 

A country Tradename Manufacturer
Germany Castellani Hollborn
Daktar McNeil
Derma-Mikotral Rosen Pharma
Fungur HEXAL
Gyno-Daktar Janssen-Cilag, 1974
Gyno-Mikotral Rosen Pharma
Infektozoor Mundgel Infectopharm
Mikobeta betapharm
Mikotar Dermapharm
Mikoderm Engelhard
Mikotin Ardeypharm
Vobamik Almirall Hermal
France Daktapin Janssen-Cilag
Gyno-Daktapin Janssen-Cilag
Loramik Bioalliance
United Kingdom Gyno-Daktapin Janssen-Cilag
Italy Daktapin Janssen-Cilag
Mikonal Ecobi
Mikotef LPB
Miderm Mendelejeff
Nizakol PS Pharma
Pivanazolo Medestea
Prilagin Sofar
Japan Florid Mochida
USA Fungoid Pedinol
Ukraine GІNEZOL 7 Sagmel, Іnk., USA
MІKONAZOL-Darnitsa CJSC “Farmatsevtichna FIRMA” Darnitsa “, m. Kyiv, Ukraine
MІKOGEL BAT “Kiїvmedpreparat”, m. Kyiv, Ukraine
various generic drugs

Formulations

 

  • ampoule 200 mg / 20 ml;
  • cream 1%, 2 g / 100 g 20 mg / g;
  • losyon 1%;
  • ointment 1%;
  • 2% oral gel;
  • Powder 2 g / 100 g 20 mg / g (in the form mononitrate);
  • solution of 20 mg / ml;
  • 100 mg suppositories;
  • Tablets of 250 mg (free base form);
  • vaginal cream 20 mg / g;
  • bottles of 400 mg / 40 ml

references

 

  1. Synthesis of a)
    • DAS 1,940,388 (Janssen; appl 8.8.1969;. USA-prior 19.8.1968, 23.7.1969.).
    • US 3,717,655 (Janssen; 20.2.1973; appl 19.8.1968.).
    • US 3,839,574 (Janssen; 1.10.1974; prior 23.7.1969.).

Miconazole nitrate was prepared by Godefori et
al
[5­
7]. Imidazole 1 was coupled with
brominated 2,4‑dichloroacetophenone 2 and the resulting ketonic product 3
was reduced with sodium borohydride to its corresponding alcohol 4. The
latter compound 4 was then coupled with 2,4-dichlorotoluene by sodium borohydride
in hexamethylphosphoramide (an aprotic solvent) which was then extracted with
nitric acid to give miconazole nitrate.

 

 

2-     Miconazole was also
prepared by Molina Caprile [8] as follows:
Phenyl methyl ketone 1 was brominated to give
1-phenyl-2-bromoethanone 2. Compound 2 was treated with
methylsulfonic acid to yield the corresponding methylsulfonate 3.
Etherification of 3 gave the a‑benzyloxy derivative 4 and compound 4 was
then chlorinated to give the 2,4‑dichlorinated derivative in both aromatic ring
systems 5. Compound 5 reacted with imidazole in dimethylformamide
to give miconazole 6 [7] which is converted to miconazole nitrate.

 

3-     Ye
et al reported that the reduction of 2,4-dichlorophenyl-2-chloroethanone
1 with potassium borohydride in dimethylformamide to give 90% a‑chloromethyl-2,4-dichlorobenzyl
alcohol 2. Alkylation of imidazole with compound 2 in dimethyl­formamide
in the presence of sodium hydroxide and triethylbenzyl ammonium chloride, gave
1-(2,4‑dichlorophenyl-2-imidazolyl)ethanol 3 and etherification of 3
with 2,4-dichlorobenzyl chloride under the same condition, 62% yield of
miconazole [9].
4-     Liao
and Li enantioselectively synthesized and studied the antifungal activity of
optically active miconazole and econazole. The key step was the
enantioselective reduction of 2‑chloro-1-(2,4-dichlorophenyl)ethanone catalyzed
by chiral oxazaborolidine [10].
5-     Yanez
et al reported the synthesiz of miconazole and analogs through a
carbenoid intermediate. The process involves the intermolecular insertion of
carbenoid species to imidazole from a‑diazoketones with copper acetylacetonate as the key
reaction of the synthetic route [11].
5-11 as 1-7
1.             E.F. Godefori and J. Heeres, Ger. Pat. 1,940,388
(1970).
2.
E.F. Godefori and J. Heeres, U.S. Pat. 3,717,655
(1973).
3.
E.F. Godefori, J. Heeres, J. van Cutsem and P.A.J.
Janssen, J. Med. Chem., 12, 784 (1969).
4.
F. Molina Caprile, Spanish Patent ES 510870 A1
(1983).
5.
B. Ye, K. Yu and Q. Huang, Zhongguo Yiyao Gongye
Zazhi
, 21, 56 (1990).
6.
Y.W. Liao and H.X. Li, Yaoxue Xuebao, 28,
22 (1993).
7.
E.C. Yanez, A.C. Sanchez, J.M.S. Becerra, J.M.
Muchowski and C.R. Almanza, Revista de la Sociedad Quimica de Mexico, 48,
49 (2004).

MiconazoleTitle: Miconazole

CAS Registry Number: 22916-47-8
CAS Name: 1-[2-(2,4-Dichlorophenyl)-2-[(2,4-dichlorophenyl)methoxy]ethyl]-1H-imidazole
Additional Names: 1-[2,4-dichloro-b-[(2,4-dichlorobenzyl)oxy]phenethyl]imidazole
Molecular Formula: C18H14Cl4N2O
Molecular Weight: 416.13
Percent Composition: C 51.95%, H 3.39%, Cl 34.08%, N 6.73%, O 3.84%
Literature References: Prepn: E. F. Godefroi et al., J. Med. Chem. 12, 784 (1969); E. F. Godefroi, J. Heeres, DE 1940388;eidem, US 3717655 (1970, 1973 to Janssen). Clinical evaluation: Brugmans et al., Arch. Dermatol. 102, 428 (1970); Godts et al.,Arzneim.-Forsch. 21, 256 (1971). Review: P. Janssen, W. Van Bever, in Pharmacological and Biochemical Properties of Drug Substances vol. 2, M. E. Goldberg, Ed. (Am. Pharm. Assoc., Washington, DC, 1979) pp 333-354; R. C. Heel et al., Drugs 19, 7-30 (1980).
Derivative Type: Nitrate
CAS Registry Number: 22832-87-7
Manufacturers’ Codes: R-14889
Trademarks: Aflorix (Gramon); Albistat (Ortho); Andergin (ISOM); Brentan (Janssen); Conoderm (C-Vet); Conofite (Mallinckrodt); Daktar (Janssen); Daktarin (Janssen); Deralbine (Andromaco); Dermonistat (Ortho); Epi-Monistat (Cilag); Florid (Mochida); Fungiderm (Janssen); Fungisdin (Isdin); Gyno-Daktarin (Janssen); Gyno-Monistat (Cilag-Chemie); Micatin (J & J); Miconal Ecobi (Ecobi); Micotef (LPB); Monistat (Cilag-Chemie); Prilagin (Gambar); Vodol (Andromaco)
Molecular Formula: C18H14Cl4N2O.HNO3
Molecular Weight: 479.14
Percent Composition: C 45.12%, H 3.16%, Cl 29.60%, N 8.77%, O 13.36%
Properties: Crystals, mp 170.5° (Godefroi, Heeres, 1970); 184-185° (Godefroi).
Melting point: mp 170.5° (Godefroi, Heeres, 1970); 184-185° (Godefroi)
Derivative Type: (+)-Form nitrate
Properties: mp 135.3°. [a]D20 +59° (methanol).
Melting point: mp 135.3°
Optical Rotation: [a]D20 +59° (methanol)
Derivative Type: (-)-Form nitrate
Properties: mp 135°. [a]D20 -58° (methanol).
Melting point: mp 135°
Optical Rotation: [a]D20 -58° (methanol)
Therap-Cat: Antifungal (topical).
Therap-Cat-Vet: Antifungal (topical).
Keywords: Antifungal (Synthetic); Imidazoles.

References

  1. Jump up^ “WHO Model List of EssentialMedicines” (PDF)World Health Organization. October 2013. Retrieved 22 April 2014.
  2. Jump up^ British National Formulary ’45’ March 2003
  3. Jump up^ “Strange Beauty: Monistat Effectively Increases Hair Growth?”. Black Girl With Long Hair. Retrieved 12 April 2012.
  4. Jump up^ Ju, Jiang; Tsuboi, Ryoji; Kojima, Yuko; Ogawa, Hideoki (2005). “Topical application of ketoconazole stimulates hair growth in C3H/HeN mice”Journal of dermatology32: 243–247.
  5. Jump up^ S., Venturoli; O. Marescalchi; F. M. Colombo; S. Macrelli; B. Ravaioli; A. Bagnoli; R. Paradisi; C. Flamigni (April 1999). “A Prospective Randomized Trial Comparing Low Dose Flutamide, Finasteride, Ketoconazole, and Cyproterone Acetate-Estrogen Regimens in the Treatment of Hirsutism”The Journal of Clinical Endocrinology and Metabolism84 (4): 1304–1310. doi:10.1210/jc.84.4.1304. Retrieved 12 April 2012.
  6. Jump up^ Duret C, Daujat-Chavanieu M, Pascussi JM, Pichard-Garcia L, Balaguer P, Fabre JM, Vilarem MJ, Maurel P, Gerbal-Chaloin S (2006). “Ketoconazole and miconazole are antagonists of the human glucocorticoid receptor: consequences on the expression and function of the constitutive androstane receptor and the pregnane X receptor”. Mol. Pharmacol70 (1): 329–39. doi:10.1124/mol.105.022046PMID 16608920.
  7. Jump up^ Najm, Fadi J.; Madhavan, Mayur; Zaremba, Anita; Shick, Elizabeth; Karl, Robert T.; Factor, Daniel C.; Miller, Tyler E.; Nevin, Zachary S.; Kantor, Christopher (2015-01-01).“Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo”Nature522 (7555). doi:10.1038/nature14335.
  8. Jump up^ United States Patent 5461068

External links

Medical

Photographic

 

Miconazole
Miconazole2DCSD.svg
Miconazole ball-and-stick.png
Systematic (IUPAC) name
(RS)-1-(2-(2,4-Dichlorobenzyloxy)-2-(2,4-dichlorophenyl)ethyl)-1H-imidazole
Clinical data
Trade names Desenex, Monistat, Zeasorb-AF
AHFS/Drugs.com Monograph
MedlinePlus a601203
Pregnancy
category
  • AU: A
  • US: C (Risk not ruled out)
  • In Australia, it is category A when used topically. In the US, the pregnancy category is C for oral and topical treatment.
Routes of
administration
topicalvaginalsublabial,oral
Legal status
Legal status
  • AU: S2 (Pharmacy only)
  • UK: POM (Prescription only)
  • US: OTC
  • Schedule 2 in Australia for topical formulations, schedule 3 (Aus) for vaginal use and for oral candidiasis, otherwise schedule 4 in Australia
Pharmacokinetic data
Bioavailability n/a
Metabolism n/a
Biological half-life n/a
Excretion n/a
Identifiers
CAS Number 22916-47-8 Yes
ATC code A01AB09 (WHO)A07AC01 (WHO)D01AC02 (WHO)G01AF04 (WHO)J02AB01 (WHO)S02AA13 (WHO)
PubChem CID 4189
IUPHAR/BPS 2449
DrugBank DB01110 Yes
ChemSpider 4044 Yes
UNII 7NNO0D7S5M Yes
KEGG D00416 Yes
ChEBI CHEBI:6923 Yes
ChEMBL CHEMBL91 Yes
Chemical data
Formula C18H14Cl4N2O
Molar mass 416.127 g/mol
Chirality Racemic mixture

Day 2 of the 2016 Doodle Fruit Games! Find out more at g.co/fruit

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

 

Join me on Linkedin

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

Join me on Facebook FACEBOOK

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

 amcrasto@gmail.com


///////////


Filed under: GENERICS, Uncategorized Tagged: ミコナゾール硝酸塩, Миконазол, MICONAZOLE NITRATE

Commercial Production of Semi-Synthetic Artemisinin

$
0
0

STR1

Figure 1. Production of artemisinic acid or β-farnesene by engineered yeast. The sesquiterpene alkenes β-farnesene and amorphadiene are both derived from FPP (farnesyl diphosphate) by the action of specific enzymes introduced from plants: amorphadiene synthase (ADS) generates amorphadiene and β-farnesene synthase (FS) generates β-farnesene. Production strains express either ADS or FS, not both. Oxidation of amorphadiene to artemisinic acid is accomplished by the action of five plant enzymes expressed in the engineered yeast.17 Conversion of purified artemisinic acid to artemisinin is accomplished by in vitro organic chemistry. Isoprenoid production strains make little ethanol.

 

The antimalarial drug artemisinin and the specialty chemical β-farnesene are examples of natural product isoprenoids that can help solve global challenges, but whose usage has previously been limited by supply and cost impediments. This review describes the path to commercial production of these compounds utilizing fermentation of engineered yeast. Development of commercially viable yeast strains was a substantial challenge that was addressed by creation and implementation of an industrial synthetic biology pipeline. Using the engineered strains, production of β-farnesene from Brazilian sugarcane offers several environmental advantages. Among the many commercial applications of β-farnesene, its use as a feedstock for making biodegradable lubricants is highlighted. This example, along with others, highlight a powerful new suite of technologies that will become increasingly important for production of chemicals, spanning from pharmaceuticals through commodity chemicals.

 

STR1

Figure 2. Sanofi industrial semi-synthesis of artemisinin. The process starts with a moderate pressure catalytic diastereoselective hydrogenation of artemisinic acid to produce a high (95:5) ratio of the desired (R)-isomer. To avoid formation of a lactone byproduct, dihydro-epi-deoxyarteannuin B, during the photooxidation, the carboxylic acid is protected as a mixed anhydride. The final step combines formation of the intermediate hydroperoxide via photoxidation using a Hg vapor lamp and commercially available tetraphenylporphyin (TPP) as sensitizer with a Hock cleavage and rearrangement catalyzed by trifluoroacetic acid to give, after workup, the best yield reported to date of pure isolated artemisinin (55%).

Synthetic Biology and the Development of Commercial β-Farnesene Production Strains Semi-synthetic artemisinin is a pharmaceutical with a price point comparable to plant-derived artemisinin,20 namely above $150 per kg. β-Farnesene, however, is a specialty chemical with multiple uses (more details below); most specialty and commodity chemicals have significantly lower price points, often below $10 per kg. For these product categories, it is of paramount importance that fermentative production be as efficient as possible, with high yields (namely, grams of product made per gram of feed substrate), productivities (grams of product/liter of culture/hour) and concentration (also known as titer; grams of product per liter of culture). Developing yeast strains capable of the yield, productivity and titer required for chemical production requires extensive development, and has been enabled over the last decade by the new discipline of synthetic biology. Synthetic biology seeks to extend approaches and concepts from engineering and computation to redesign biology for a chosen function;21recent advances in the application of design automation, i.e., the use of software, hardware and robotics22 have enabled the creation and screening of hundreds of thousands of strain variants (created by both design and random mutagenesis) for the properties required for commercial production of β-farnesene. Notable enabling technologies developed for routine usage include rapid and reliable assembly of large (i.e., multiple kilobase) deoxyribonucleic acid (DNA) constructs;23-25 high throughput, cost effective, verification of structural DNA assemblies by both initial restriction digest26 and by low-cost DNA sequencing;27 and whole genome sequencing of yeast strains.28 In addition, there is a need to effectively identify the best new strains (akin to panning for gold!) through high throughput, rapid, and accurate methods to screen thousands of strains. Further, the results of small-scale (< 1 milliliter) tests must correspond to the results of large-scale (> 50,000 liter) production. Development and implementation of these technologies required considerable investment by Amyris. The outcome is a robust pipeline for efficient, cost-effective strain generation allied with screening for the properties required for commercial production of β-farnesene by fermentation (i.e., at a price point required for its use as a specialty chemical).

 

As the world’s population and economies grow, the demand for a wide variety of specialty, commodity, and pharmaceutical chemicals will outpace the supply available from current sources. There is an urgent need to develop alternative, sustainable sources of many existing chemicals and to develop abundant sources of currently scarce chemicals with novel beneficial properties. Synthetic biology and industrial fermentation, combined with synthetic chemistry, will be an increasingly important source of chemicals in the decades ahead; artemisinin and β-farnesene provide good examples of this relatively new approach to chemical production. Brazil’s plentiful sugar cane feedstock and fermentation expertise make it an excellent location for this type of manufacturing, which can expand and diversify the nation’s industrial base and international importance.

J. Braz. Chem. Soc. 2016, 27(8), 1339-1345

Developing Commercial Production of Semi-Synthetic Artemisinin, and of β-Farnesene, an Isoprenoid Produced by Fermentation of Brazilian Sugar

Kirsten R. Benjamin; Iris R. Silva; João P. Cherubim; Derek McPhee; Chris J. Paddon

How to cite this article

Genes encoding the biosynthetic pathway for production of a valuable product (e.g., farnesene) in a native organism are expressed in a heterologous microbial host (e.g., yeast). The engineered yeast produces farnesene by commercial fermentation. Copyright © 2016 Amyris, inc. All rights reserved.

http://dx.doi.org/10.5935/0103-5053.20160119

http://jbcs.sbq.org.br/imagebank/pdf/v27n8a04.pdf

Benjamin KR, Silva IR, Cherubim JP, Mcphee D, Paddon CJ. Developing Commercial Production of Semi-Synthetic Artemisinin, and of β-Farnesene, an Isoprenoid Produced by Fermentation of Brazilian Sugar. J. Braz. Chem. Soc. 2016;27(8):1339-1345

Kirsten R. Benjamin,a Iris R. Silva,b João P. Cherubim,c Derek McPheea and Chris J. Paddon*,a a Amyris, Inc., 5885 Hollis Street, Suite 100, CA 94608 Emeryville, USA b Amyris Brasil Ltda, Rua John Dalton 301-Bloco B-Edificio 3, Condominio Techno Plaza, 13069-330 Campinas-SP, Brazil c Amyris Brasil Ltda, Rodovia Brotas/Torrinha-km 7.5, 17380-000 Brotas-SP, Brazil

*e-mail: paddon@amyris.com
Chris Paddon

Chris Paddon, PhD

Dr. Paddon has a PhD in Biochemistry from Imperial College, London, but now considers himself a synthetic biologist. After postdoctoral work at the National Institutes of Health in Bethesda, MD, he worked in the pharmaceutical industry (GlaxoSmithKline), and then for two Bay Area biopharmaceutical companies (Affymax and Xenoport) before joining Amyris, Inc. in 2005 as its sixth employee and first scientist. He was project leader for the semi-synthetic artemisinin project at Amyris, Inc. and has subsequently led a number of other projects and programs there.

Chris Paddon is a Principal Scientist at Amyris, Inc. in Emeryville, CA. He was project leader for the Semi-Synthetic Artemisinin project, and subsequently led a number of projects at Amyris using synthetic biology for the production of natural products. He received his Bachelor’s degree in Microbiology from The University of Surrey (UK), and doctorate in Biochemistry from Imperial College (London, UK). Following postdoctoral work at The National Institutes for Health (Bethesda, MD) he joined the pharmaceutical industry, working for GSK (London, UK). He subsequently worked for Affymax (Palo Alto, CA) and Xenoport (Santa Clara, CA) before joining Amyris.

//////////// Commercial Production, Semi-Synthetic , Artemisinin,  farnesene, fermentation, natural product, lubricant


Filed under: antimalarials, MANUFACTURING Tagged: ARTEMISININ, Commercial Production, farnesene, fermentation, lubricant, natural product, Semi-Synthetic

Pharmaceutical Manufacturing Encyclopedia, 3rd Edition

$
0
0

If you have difficulty in viewing click,,,,,,,,,,http://www.allfordrugs.com/2016/08/07/pharmaceutical-manufacturing-encyclopedia-3rd-edition/

DESCRIPTION

This industry standard encyclopedia on pharmaceutical manufacturing processes has been completely updated to include FDA drugs approved up to the summer of 2004. The encyclopedia gives details for the manufacture of 2226 pharmaceuticals that are being marketed as a trade-named product somewhere in the world. Each entry includes:

ò Therapeutic function
ò Chemical and common name
ò Structural Formula
ò Chemical Abstracts Registry no.
ò Trade name, manufacturer, country, and year introduced
ò Raw Materials
ò Manufacturing Process

In addition, references are also cited under each drug’s entry to major pharmaceutical works where additional information can be obtained on synthesis and the pharmacology of the individual products.

 

STR1

//////////


Filed under: MANUFACTURING Tagged: 3rd Edition, Pharmaceutical Manufacturing Encyclopedia

Nacubactam, A diazabicyclooctane beta-lactamase inhibitor, for treating bacterial infection

$
0
0

 

Nacubactam

RG-6080,  FPI-1459,  OP-0595, WK ?, WK-?, WK?

 CAS 1452458-86-4,  MF C9 H16 N4 O7 S, MW 324.31
Sulfuric acid, mono[(1R,2S,5R)-2-[[(2-aminoethoxy)amino]carbonyl]-7-oxo-1,6-diazabicyclo[3.2.1]oct-6-yl] ester,

(2S,5R)-N-(2-amino ethoxy)-6-(sulfooxy)-7-oxo-1,6-diazabicyclo[3.2.1]octane-2-carboxamide

Beta lactamase inhibitor

Roche, under license from Meiji Seika Pharma and Fedora Pharmaceuticals is developing nacubactam hydrate

Meiji Seika Pharma Co., Ltd., Meiji Seikaファルマ株式会社

A diazabicyclooctane beta-lactamase inhibitor, for treating bacterial infection. In July 2016, nacubactam was reported to be in phase 1 clinical development

PATENTS , IN2015MU287, WO2016116878WO 2016120752, INDICATE INTEREST FROM WOCKHARDT

 

Sulfuric acid, mono[(1R,2S,5R)-2-[[(2-aminoethoxy)amino]carbonyl]-7-oxo-1,6-diazabicyclo[3.2.1]oct-6-yl] ester

A β-lactamase inhibitor potentially for the treatment of bacterial infections.

RG-6080; FPI-1459; OP-0595

CAS No. 1452458-86-4

Molecular Formula C9 H16 N4 O7 S
Formula Weight 324.31
  • Originator Fedora Pharmaceuticals
  • Developer Meiji Seika Pharma
  • Class Antibacterials; Azabicyclo compounds
  • Mechanism of Action Beta lactamase inhibitors
  • Phase I Bacterial infections

Most Recent Events

  • 13 Jan 2015  OP 0595 licensed to Roche worldwide, except Japan ,
  • 30 Nov 2014 Meiji Seika Pharma completes a phase I trial in Healthy volunteers in Australia (NCT02134834)
  • 01 May 2014 Phase-I clinical trials in Bacterial infections (in volunteers) in Australia (IV)

In September 2014, preclinical data were presented at the 54th ICAAC Meeting in Washington, DC. Nacubactam hydratedemonstrated Ki values of 0.24, 3 and 0.79 microM against AmpC P99 derived from Enterobacter cloacae, KPC-3, and CTX-M-15 enzymes, respectively; the Ki values were lower than that of cefepime

Bacterial infections continue to remain one of the major causes contributing towards human diseases. One of the key challenges in treatment of bacterial infections is the ability of bacteria to develop resistance to one or more antibacterial agents over time. Examples of such bacteria that have developed resistance to typical antibacterial agents include: Penicillin-resistant Streptococcus pneumoniae, Vancomycin-resistant Enterococci, and Methicillin-resistant Staphylococcus aureus. The problem of emerging drug-resistance in bacteria is often tackled by switching to newer antibacterial agents, which can be more expensive and sometimes more toxic. Additionally, this may not be a permanent solution as the bacteria often develop resistance to the newer antibacterial agents as well in due course. In general, bacteria are particularly efficient in developing resistance, because of their ability to multiply very rapidly and pass on the resistance genes as they replicate.

The persistent exposure of bacterial strains to a multitude of beta- lactam antibacterial agents has led to overproduction and mutation of beta-lactamases. These new extended spectrum beta-lactamases (ESBL) are capable of hydrolyzing penicillins, cephalosporins, monobactams and even carbapenems. Such a wide spread resistance to many of the existing beta-lactam antibacterial agents, either used alone or in combination with other agents, is posing challenges in treating serious bacterial infections.

Due to various reasons, the oral therapeutic options for treating bacterial infections (including those caused by ESBL strains) are limited. For example, a combination of amoxicillin and clavulanic acid is effective against Class A ESBLs producing bacteria. However, the usefulness of this combination is compromised against bacteria producing multiple or mixed beta-lactamase enzymes (such as, for example, bacteria producing Class A and Class C ESBLs concurrently), and Klebsiella pneumoniae carbapenemases (KPCs). Therefore, oral antibacterial agents or combinations with activity against a range of bacterial strains (including those producing multiple ESBLs and KPCs) are urgently desired.

Cephalosporin antibacterial agents are known for treatment for various bacterial infections. Surprisingly, it has been found that pharmaceutical compositions comprising a cephalosporin antibacterial agent and certain nitrogen containing bicyclic compound (disclosed in PCT/IB2013/053092, PCT/JP2013/064971 and PCT/IB 2012/002675) exhibit unexpectedly synergistic antibacterial activity, even against highly resistant bacterial strains.

SYNTHESIS

WO 2015046207,

STR1

CONTD…………………..

STR1

CONTD………………………………..

STR1

Patent

The novel heterocyclic compound in Japanese Patent 4515704 (Patent Document 1), preparation and shown for their pharmaceutical use, sodium trans-7-oxo-6- (sulfooxy) as a representative compound 1,6-diazabicyclo [3 .2.1] discloses an octane-2-carboxamide (NXL104). Preparation in regard to certain piperidine derivatives which are intermediates Patent 2010-138206 (Patent Document 2) and JP-T 2010-539147 (Patent Document 3) are shown at further WO2011 / 042560 (Patent Document 4) NXL104 to disclose a method for producing the crystals.

 In Patent 5038509 (Patent Document 5) (2S, 5R) -7- oxo -N- (piperidin-4-yl) -6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane – 2- carboxamide (MK7655) is shown, discloses the preparation of certain piperidine derivatives with MK7655 at Patent 2011-207900 (Patent Document 6) and WO2010 / 126820 (Patent Document 7).

 The present inventors also disclose the novel diazabicyclooctane derivative represented by the following formula (VII) in Japanese Patent Application 2012-122603 (Patent Document 8).

Patent Document 1: Japanese Patent No. 4515704 Pat

Patent Document 2: Japanese Patent Publication 2010-138206 Pat

Patent Document 3: Japanese patent publication 2010-539147 Pat

Patent Document 4: International Publication No. WO2011 / 042560 Patent

Patent Document 5: Japanese Patent No. 5038509 Pat

Patent Document 6: Japanese Patent Publication 2011-207900 Pat

Patent Document 7: International Publication No. WO2010 / 126820 Patent

Patent Document 8: Japanese Patent application 2012-122603 Pat.

[Chemical formula 1] (In the formula, R 3 are the same as those described below)

Reference Example

5 of 5 (2S, 5R)-N- (2-aminoethoxy) -7-oxo-6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane-2-carboxamide (VII-1)

Formula 43]

step 1 tert-butyl {2 – [({[( 2S, 5R) -6- benzyloxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl } amino) oxy] ethyl} carbamate  (IV-1)(2S, 5R)-6-(benzyloxy) -7-oxo-1,6-diazabicyclo [3.2.1] octane-2-carboxylic acid (4 .30g, dehydrated ethyl acetate (47mL) solution of 15.56mmol) was cooled to -30 ℃, isobutyl chloroformate (2.17g, washing included dehydration ethyl acetate 1mL), triethylamine (1.61g, washing included dehydration ethyl acetate 1 mL), successively added dropwise, and the mixture was stirred 1 hour at -30 ° C.. To the reaction solution tert- butyl 2-dehydration of ethyl acetate (amino-oxy) ethyl carbamate (3.21g) (4mL) was added (washing included dehydration ethyl acetate 1mL), raising the temperature over a period of 1.5 hours to 0 ℃, It was further stirred overnight. The mixture of 8% aqueous citric acid (56 mL), saturated aqueous sodium bicarbonate solution (40 mL), sequentially washed with saturated brine (40 mL), dried over anhydrous magnesium sulfate, filtered, concentrated to 5 mL, up to 6mL further with ethanol (10 mL) It was replaced concentrated. Ethanol to the resulting solution (3mL), hexane the (8mL) in addition to ice-cooling, and the mixture was stirred inoculated for 15 minutes. The mixture was stirred overnight dropwise over 2 hours hexane (75 mL) to. Collected by filtration the precipitated crystals, washing with hexane to give the title compound 5.49g and dried in vacuo (net 4.98 g, 74% yield). HPLC: COSMOSIL 5C18 MS-II 4.6 × 150 mm, 33.3 mM phosphate buffer / MeCN = 50/50, 1.0 mL / min, UV 210 nm, Retweeted 4.4 min; 1 H NMR (400 MHz, CDCl 3 ) [delta] 1.44 (s, 9H), 1.56-1.70 (m, 1H), 1.90-2.09 (m, 2H), 2.25-2.38 (m, 1H), 2.76 (d, J = 11.6 Hz, 1H), 3.03 (br.d., J = 11.6 Hz , 1H), 3.24-3.47 (m, 3H), 3.84-4.01 (m, 3H), 4.90 (d, J = 11.6 Hz, 1H), 5.05 (d, J = 11.6 Hz, 1H), 5.44 (br. . s, 1H), 7.34-7.48 (yd, 5H), 9.37 (Br.S., 1H); MS yd / z 435 [M + H] + .

Step 2

tert-butyl {2 – [({[( 2S, 5R) -6- hydroxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate

(V-1) tert-butyl {2 – [({[( 2S, 5R) -6- benzyloxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl ] carbonyl} amino) oxy] ethyl} carbamate (3.91 g, to a methanol solution (80 mL) of 9.01mmol), 10% palladium on carbon catalyst (50% water, 803 mg) was added, under hydrogen atmosphere and stirred for 45 minutes . The reaction mixture was filtered through Celite, after concentrated under reduced pressure to give 3.11g of the title compound (quantitative).

HPLC: COSMOSIL 5C18 MS-II 4.6 × 150 mm, 33.3 mM phosphate buffer / MeCN = 75/25, 1.0 mL / min, UV 210 nm, Retweeted 3.9 from min; 1 H NMR (400 MHz, CD 3 OD) [delta] 1.44 (s, 9H) , 1.73-1.83 (m, 1H), 1.86-1.99 (m, 1H), 2.01-2.12 (m, 1H), 2.22 (br.dd., J = 15.0, 7.0 Hz, 1H), 3.03 (d, J= 12.0 Hz, 1H), 3.12 (br.d., J = 12.0 Hz, 1H), 3.25-3.35 (m, 2H), 3.68-3.71 (m, 1H), 3.82-3.91 (m, 3H); MS M / Z 345 [M Tasu H] Tasu .

Step 3

Tetrabutylammonium tert- butyl {2 – [({[( 2S, 5R) -7- oxo-6 (sulfooxy) 1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl } amino) oxy] ethyl} carbamate

(VI-1) tert-butyl {2 – [({[( 2S, 5R) -6- hydroxy-7-oxo-1,6-diazabicyclo [3.2.1] oct 2-yl] carbonyl} amino) oxy] ethyl} carbamate (3.09g, in dichloromethane (80mL) solution of 8.97mmol), 2,6- lutidine (3.20mL), sulfur trioxide – pyridine complex (3 .58g) was added, and the mixture was stirred overnight at room temperature. The reaction mixture was poured into half-saturated aqueous sodium bicarbonate solution, washed the aqueous layer with chloroform, tetrabutylammonium hydrogen sulfate to the aqueous layer and (3.47 g) chloroform (30 mL) was added and stirred for 10 minutes. The aqueous layer was extracted with chloroform, drying the obtained organic layer with anhydrous sodium sulfate, filtered, and concentrated in vacuo to give the title compound 5.46g (91% yield).

HPLC: COSMOSIL 5C18 MS-II 4.6X150mm, 33.3MM Phosphate Buffer / MeCN = 80/20, 1.0ML / Min, UV210nm, RT 2.0 Min; 1 H NMR (400 MHz, CDCl 3 ) Deruta 1.01 (T, J = 7.4 Hz, 12H), 1.37-1.54 (m , 8H), 1.45 (s, 9H), 1.57-1.80 (m, 9H), 1.85-1.98 (m, 1H), 2.14-2.24 (m, 1H), 2.30- 2.39 (m, 1H), 2.83 (d, J = 11.6 Hz, 1H), 3.20-3.50 (m, 11H), 3.85-3.99 (m, 3H), 4.33-4.38 (m, 1H), 5.51 (br s , 1H), 9.44 (Br.S., 1H); MS yd / z 425 [M-Bu 4 N + 2H] + .

Step 4 (2S, 5R)-N- (2-aminoethoxy) -7-oxo-6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane-2-carboxamide (VII-1)

tetra butylammonium tert- butyl {2 – [({[( 2S, 5R) -7- oxo-6 (sulfooxy) 1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate (5.20g, 7.82mmol) in dichloromethane (25mL) solution of ice-cold under trifluoroacetic acid (25mL), and the mixture was stirred for 1 hour at 0 ℃. The reaction mixture was concentrated under reduced pressure, washed the resulting residue with diethyl ether, adjusted to pH7 with aqueous sodium bicarbonate, subjected to an octadecyl silica gel column chromatography (water), after freeze drying, 1.44 g of the title compound obtained (57% yield).

HPLC: COSMOSIL 5C18 MS-II 4.6X150mm, 33.3MM Phosphate Buffer / MeCN = 99/1, 1.0ML / Min, UV210nm, RT 3.1 Min; 1 H NMR (400 MHz, D 2O) Deruta 1.66-1.76 (M, 1H), 1.76-1.88 (m, 1H ), 1.91-2.00 (m, 1H), 2.00-2.08 (m, 1H), 3.02 (d, J = 12.0 Hz, 1H), 3.15 (t, J = 5.0 Hz , 2H), 3.18 (br d , J = 12.0 Hz, 1H), 3.95 (dd, J = 7.8, 2.2 Hz, 1H), 4.04 (t, J = 5.0 Hz, 2H), 4.07 (dd, J = 6.4 3.2 Hz &, 1H); MS yd / z 325 [M + H] + .

PATENT

Example 

64 tert-butyl {2 – [({[( 2S, 5R) -6- hydroxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy ] ethyl} carbamate (V-1) 

[of 124] 

tert- butyl {2 – [({[(2S, 5R) -6- benzyloxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl } carbamate (example 63q, net 156.42g, 360mmol) in methanol solution (2.4L) of 10% palladium carbon catalyst (50% water, 15.64g) was added, under an atmosphere of hydrogen, stirred for 1.5 hours did. The catalyst was filtered through celite, filtrate was concentrated under reduced pressure until 450mL, concentrated to 450mL by adding acetonitrile (1.5 L), the mixture was stirred ice-cooled for 30 minutes, collected by filtration the precipitated crystals, washing with acetonitrile, and vacuum dried to obtain 118.26g of the title compound (net 117.90g, 95% yield). Equipment data of the crystals were the same as those of the step 2 of Reference Example 3.

Example

65 (2S, 5R)-N- (2-aminoethoxy) -7-oxo-6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane-2-carboxamide (VI-1)

 

 tert- butyl {2 – [({[(2S, 5R) -1,6- -6- hydroxy-7-oxo-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate (example 64,537.61g, 1.561mol) in acetonitrile (7.8L) solution of 2,6-lutidine (512.08g), sulfur trioxide – pyridine complex (810.3g) was added, at room temperature in the mixture was stirred overnight. Remove insolubles and the mixture was filtered, the filtrate concentrated to 2.5 L, diluted with ethyl acetate (15.1L). The mixture was extracted with 20% phosphoric acid 2 hydrogencarbonate aqueous solution (7.8L), the resulting aqueous layer into ethyl acetate (15.1L), added tetrabutylammonium hydrogen sulfate (567.87g), was stirred for 20 min. The organic layer was separated layers, dried over anhydrous magnesium sulfate (425 g), after filtration, concentration under reduced pressure, substituted concentrated tetrabutylammonium tert- butyl with dichloromethane (3.1L) {2 – [({[(2S, 5R ) -7-oxo-6 (sulfooxy) 1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate was obtained 758g (net 586.27g, Osamu rate 84%).

 The tetra-butyl ammonium salt 719g (net 437.1g, 0.656mol) in dichloromethane (874mL) solution was cooled to -20 ℃, dropping trifluoroacetic acid (874mL) at 15 minutes, 1 the temperature was raised to 0 ℃ It was stirred time. The reaction was cooled to -20 ° C. was added dropwise diisopropyl ether (3.25L), and the mixture was stirred for 1 hour the temperature was raised to 0 ° C.. The precipitate is filtered, washed with diisopropyl ether to give the title compound 335.36g of crude and vacuum dried (net 222.35g, 99% yield).

 The title compound of crude were obtained (212.99g, net 133.33g) and ice-cold 0.2M phosphate buffer solution of pH5.3 mix a little at a time, alternating between the (pH6.5,4.8L). The solution was concentrated under reduced pressure to 3.6L, it was adjusted to pH5.5 at again 0.2M phosphate buffer (pH6.5,910mL). The solution resin purification (Mitsubishi Kasei, SP207, water ~ 10% IPA solution) is subjected to, and concentrated to collect active fractions, after lyophilization, to give the title compound 128.3 g (96% yield). Equipment data of the crystals were the same as those of step 3 of Reference Example 3.

PATENT

US 20140288051

WO 2014091268

WO 2013180197

US 20130225554

PATENT

IN2015MU287

PATENT

WO2013180197

Example 59
(2S, 5R) -N- (2- aminoethoxy) -7-oxo-6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane-2-carboxamide (II-059)

Figure JPOXMLDOC01-appb-C000130

Step 1
tert- butyl {2 – [({[(2S, 5R) -6- Benzyloxy-7-oxo-1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl } carbamate

Figure JPOXMLDOC01-appb-C000131

Acid of Example 9 or 16 (6b, 1.34g, 4.87mmol) in methylene chloride (35mL) solution of triethylamine (2.71mL), N- ethyl -N ‘- (3- dimethylaminopropyl) carbodiimide hydrochloride (1.41g), 1- hydroxybenzotriazole monohydrate (1.15g), were added tert- butyl of Reference Example 9, wherein 2- (amino-oxy) ethyl carbamate (1.12g), room temperature It was stirred overnight Te.Water was added to the reaction solution to a residue obtained by concentration under reduced pressure, and extracted with ethyl acetate. The resulting organic layer with 0.1M hydrochloric acid, saturated aqueous sodium bicarbonate solution, washed with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated.The resulting residue was purified by silica gel column and purified by chromatography (hexane / ethyl acetate = 8 / 2-0 / 10) to give the title compound 1.77g (84% yield).
[Α] D 20 -0.08 ° (c 0.29, CHCl 3); 1 H NMR (400 MHz, CDCl 3), δ: 1.44 (s, 9H), 1.56-1.70 (m, 1H), 1.90-2.09 (m , 2H), 2.25-2.38 (m, 1H), 2.76 (d, J = 11.6 Hz, 1H), 3.03 (br d, J = 11.6 Hz, 1H), 3.24-3.47 (m, 3H), 3.84-4.01 (m, 3H), 4.90 (d, J = 11.6 Hz, 1H), 5.05 (d, J = 11.6 Hz, 1H), 5.44 (br s, 1H), 7.34-7.48 (m, 5H), 9.37 (br s, 1H); MS m / z 435 [M + H] +; enantiomeric excess of 99.9% or higher ee (CHIRALPAK AD-H, 4.6x150mm, hexane / ethanol = 2/1, UV210nm, flow rate 1mL / min, retention time 4.95min (2R, 5S), 6.70min (2S, 5R).

Step 2
tert- butyl {2 – [({[(2S, 5R) -1,6- -6- hydroxy-7-oxo-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate

Figure JPOXMLDOC01-appb-C000132

Compound of the above Step 1 (3.91g, 9.01mmol) in methanol (80mL), 10% palladium on carbon catalyst (50% water, 803mg) was added, under hydrogen atmosphere and stirred for 45 minutes. The reaction mixture was filtered through Celite, then concentrated under reduced pressure, to give 3.11g of the title compound (quantitative).
1 H NMR (400 MHz, CD 3 OD), δ: 1.44 (s, 9H), 1.73-1.83 (m, 1H), 1.86-1.99 (m, 1H), 2.01-2.12 (m, 1H), 2.22 ( br dd, J = 15.0, 7.0 Hz, 1H), 3.03 (d, J = 12.0 Hz, 1H), 3.12 (br d, J = 12.0 Hz, 1H), 3.25-3.35 (m, 2H), 3.68-3.71 (m, 1H), 3.82-3.91 (m, 3H); MS m / z 345 [M + H] +.

Step 3
(2S, 5R) -N- (2- aminoethoxy) -7-oxo-6- (sulfooxy) 1,6-diazabicyclo [3.2.1] octane-2-carboxamide The above step 2 compound (3. 09g, in methylene chloride (80mL) solution of 8.97mmol), 2,6- lutidine (3.20mL), sulfur trioxide – was added pyridine complex (3.58g), and stirred at room temperature overnight. The reaction mixture was poured into half-saturated aqueous sodium bicarbonate solution, and washed the aqueous layer with chloroform, and tetrabutylammonium hydrogen sulfate (3.47g) and chloroform (30mL) was added to the aqueous layer and stirred for 10 minutes. After extracting the aqueous layer with chloroform, drying the resulting organic layer over anhydrous sodium sulfate, filtered, concentrated under reduced pressure tetrabutylammonium tert- butyl {2 – [({[(2S, 5R) -7- oxo – 6- (sulfooxy) 1,6-diazabicyclo [3.2.1] oct-2-yl] carbonyl} amino) oxy] ethyl} carbamate was obtained 5.46g (91% yield).
1 H NMR (400 MHz, CDCl 3), δ: 1.01 (t, J = 7.4 Hz, 12H), 1.37-1.54 (m, 8H), 1.45 (s, 9H), 1.57-1.80 (m, 9H), 1.85-1.98 (m, 1H), 2.14-2.24 (m, 1H), 2.30-2.39 (m, 1H), 2.83 (d, J = 11.6 Hz, 1H), 3.20-3.50 (m, 11H), 3.85- 3.99 (m, 3H), 4.33-4.38 (m, 1H), 5.51 (br s, 1H), 9.44 (br s, 1H); MS m / z 425 [M-Bu 4 N + 2H] +.

The tetrabutyl ammonium salt (5.20g, 7.82mmol) in methylene chloride (25mL) solution of under ice-cooling trifluoroacetic acid (25mL), and the mixture was stirred for 1 hour at 0 ℃. The reaction mixture was concentrated under reduced pressure, washed resulting residue with diethyl ether, at aqueous sodium bicarbonate was adjusted to pH7, it performs an octadecyl silica gel column chromatography (water), after freeze-drying, 1.44g of the title compound The obtained (57% yield).
[Α] D 24 -63.5 ° (c 0.83, H 2 O); 1 H NMR (400 MHz, D 2 O), δ: 1.66-1.76 (m, 1H), 1.76-1.88 (m, 1H), 1.91 -2.00 (m, 1H), 2.00-2.08 (m, 1H), 3.02 (d, J = 12.0 Hz, 1H), 3.15 (t, J = 5.0 Hz, 2H), 3.18 (br d, J = 12.0 Hz , 1H), 3.95 (dd, J = 7.8, 2.2 Hz, 1H), 4.04 (t, J = 5.0 Hz, 2H), 4.07 (dd, J = 6.4, 3.2 Hz, 1H); MS m / z 325 [ M + H] +.

PATENT

WO2016116878

ANTIBACTERIAL COMPOSITIONS OF A BETA-LACTAMASE INHIBITOR WITH A CEPHALOSPORINAbstract:

Pharmaceutical compositions comprising: (a) at least one cephalosporin antibacterial agent and (b) a compound of Formula (I) or a stereoisomer or a pharmaceutically acceptable derivative thereof are disclosed. Formula (I)

PATENT

WO 2016120752, WOCKHARDT, NEW PATENT, Nacubactam

Formula (I), chemically known as (25, 5i?)-N-(2-aminoethoxy)-6-(sulfooxy)-7-oxo-l ,6-diazabicyclo[3.2.1 ]octane-2-carboxamide has antibacterial properties and is disclosed in PCT International Patent Application No. PCT/IB2013/053092, PCT/JP2013/064971 and PCT/IB2012/002675. The present invention discloses a process for preparation of a compound of Formula (I).

Formula (I)

 

(VII) (VIII) (IX)

Scheme 2

Example 1

Synthesis of fert-butyl-r2-(aminooxy) ethyllcarbamate (III)

Preparation of fert-butyl-2-hydroxy ethylcarbamate (VIII):

Formula (VIII)

To a stirred solution of ethanolamine (50.0 g, 0.8186 mol) in dichloromethane (1000 ml), was added triethylamine (124 g, 1.228 mol) at 0°C. After 10 minutes, di-teri-butyl dicarbonate (VII, 214.15 g, 0.9823 mol) was added drop wise at 0°C under continuous stirring. Then reaction mass was allowed to warm to 25°C and stirred further for 3 hours. After completion of reaction, the resulting reaction mixture was poured into water (250 ml) and the organic layer was separated and dried over anhydrous sodium sulfate. The dried organic layer was concentrated under reduced pressure to obtain 130 g of the titled product as colorless oil in 98% yield.

Analysis:

Mass: 162 (M+l); for Molecular Weight of 161.2 and Molecular Formula of C7H15NO3.

1H NMR (400MHz, CDC13): δ 4.92(br s,lH), 3.72-3.68(q,2H), 3.30-3.26(q,2H), 2.33(br s,lH), 1.44(s,9H).

Preparation of A7-Boc-2-(2-aminoethoxy)isoindoline-l,3-dione (IX):

To a stirred solution of teri;butyl-2-hydroxy-ethylcarbamate (VIII, 50 g, 0.3106 mol) in tetrahydrofuran (500 ml), was added triphenylphosphine (89.5 g, 0.3416 mol) at 25°C. After stirring for 10 minutes, a solution of N-hydroxyphthalimide (50.66 g, 0.3106 mol) in dichloromethane (250 ml) was added to the reaction mass at 25 °C over a period of 10 minutes. After stirring for further 10 minutes, diisopropyl azodicarboxylate (69.1 g, 0.3416 mol) was added to the reaction mass in small portions (exothermic reaction was observed up to 34°C). The resulting reaction mass was stirred further at 25°C. After 16 hours, the reaction mass was concentrated under reduced pressure to obtain colorless oily material. The oily residue was diluted with diisopropyl ether (200 ml) and stirred for 30 minutes. The separated solid was filtered under suction. The filtrate was evaporated under reduced pressure and the residue subjected to di-isopropyl ether treatment (200 ml). This procedure was repeated once again. The filtrate was concentrated to obtain a solid product. The obtained solid was washed with diisopropyl ether (50 ml) and dried under reduced pressure. This solid contains small amount of triphenylphosphine oxide, along with the product. This was used as such for the next reaction without further purification.

Analysis:

Mass: 307.2 (M+l); for Molecular Weight of 306.3 and Molecular Formula of Ci5Hi8N205; 1H NMR of purified material (400MHz, CDC13): 7.85-7.25 (m,4H), 5.62(br s,lH), 4.26-4.23(t,2H), 3.46-3.42(q,2H), 1.46(s,9H).

Step 3: Preparation of fert-butyl-[ -(aminooxy) ethyl]carbamate (III):

Formula (III)

To a stirred solution of N-Boc-2-(2-aminoethoxy)isoindoline-l ,3-dione (IX, 97 g, 0.3167 mol) in dichloromethane (970 ml) was added hydrazine hydrate (31.7 g, 0.6334 mol) , at 0°C, drop wise, over a period of 45 minutes and the stirring continued further. After 2 hours, the reaction mass was filtered under suction. Filtrate was washed with water (485 ml), and the organic layer was diluted with an aq. solution of 10% potassium hydrogen sulfate (485 ml) and stirred for 15 minutes. The aqueous layer was separated, neutralized with solid sodium hydrogen carbonate and extracted with dichloromethane (2 x 485 ml). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain colorless oil, this was used as such for further reaction immediately (28g, overall yield of step II and step III was 60%)

Analysis:

Mass: 177.2 (M+l) for Molecular Weight of 176.2 and Molecular Formula of C7H16N2O3.

Example 2

Synthesis of (25,5R)-jV-(2-aminoethoxy)-6-(sulfooxy)-7-oxo-l,6-diaza-bicvclor3.2.11octane-2- carboxamide (I)

Step 1: Preparation of (25,5R)-iV-(2-Boc-aminoethoxy)-6-(benzyloxy)-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carboxamide (IV):

To a clear solution of sodium (25,5i?)-6-(benzyloxy)-7-oxo-l,6-diazabicyclo[3.2.1]octane-2-carboxylate (II, 42.67 g, 0.143 mol; prepared according to the procedure disclosed in Indian Patent Application No. 699/MUM/2013) in water (426 ml) was added EDC.HC1 (67.1 g, 0.349 mol) at 15°C

under stirring. After 10 minutes, a solution of teri-butyl-[2-(aminooxy) ethyl]carbamate (III, 28.0g, 0.159 mol; prepared as per the literature procedure depicted in Scheme 2) in dimethylformamide (56 ml) was added drop wise at 10°C under continuous stirring. The temperature of the reaction mass was allowed to warm to 25°C and then HOBt (21.5g, 0.159 mol) was added in small portions over a period of 15 minutes and the resulting mixture was further stirred at room temperature for 16 hours. The reaction was continuously monitored using thin layer chromatography using mixture of acetone and hexane (35 :65) as solvent system. After completion of reaction, the resulting mixture was filtered and the residue was washed with water (130 ml). The obtained white residue was suspended in water (130 ml) and the mixture stirred at 50°C for 3 hours. The resulting suspension was filtered, the residue dried under reduced pressure to obtain 51 g of (2S,5R)-N-(2-Boc-aminoethoxy)-6-(benzyloxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1]octane-2-carboxamide (IV) as off white solid in 73% yield.

Analysis:

Mass: 433.4 (M-l ); for Molecular Weight of 434.5 and Molecular Formula of C21H30N4O6;

1H-NMR (400MHz, CDC13): δ 9.32 (br s, 1H), 7.41 -7.26(m,5H), 5.41(br s, 1H), 5.06-4.88(dd, 2H), 3.98-3.96(d,lH), 3.91-3.90(m,2H), 3.39(m, 1H), 3.31-3.26(m, 2H), 3.04-3.01(d,lH), 2.77-2.74(d, 1H), 2.33-2.28(m, 1H), 2.03-1.93(m, 2H), 1.67-1.64(m, 1H), 1.44(s, 9H);

Purity as determined by HPLC: 99.4%.

Step 2: Preparation of (2S,5R)-iV-(2-Boc-aminoethoxy)-6-(hydroxy)-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carboxamide (V):

A solution of (25,5i?)-N-(2-Boc-aminoethoxy)-6-(benzyloxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1] octane-2-carboxamide (IV, 38 g, 0.0875 mol) in a mixture of dimethylformamide and dichloromethane (2: 8, 76 ml: 304 ml), containing 10% Pd/C (7.6 g, 50% wet) was hydrogenated at 50 psi hydrogen atmosphere at 25°C for 3 hours. The resulting mixture was filtered through a celite pad. The residue was washed with dichloromethane (75 ml). The solvent from the combined filtrate was evaporated

under reduced pressure to obtain 30 g (25,5i?)-N-(2-Boc-aminoethoxy)-6-(hydroxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1 ]octane-2-carboxamide (V) as an oil, which was used as such for the next reaction without further purification.

Analysis:

Mass: 343.3 (M-l ) for Molecular Weight of 344.3 and Molecular Formula of C14H24N4O6.

Step 3: Preparation of (25,5R)-iV-(2-Boc-aminoethoxy)-6-(sulfooxy)-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carboxamide,tetrabutyl ammonium salt (VI):

To a stirred solution of (25,5i?)-N-(2-Boc-aminoethoxy)-6-(hydroxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1 ]octane-2-carboxamide (V, 30.0 g, 0.0875 mol) in dimethylformamide (150 ml) was added sulphur trioxide dimethylformamide complex (16.06 g, 0.105 mol) in one portion, at 10°C. The reaction mass was stirred at the same temperature for 30 minutes and then allowed to warm to room temperature. After 2 hours, a solution of tetrabutylammonium acetate (31.6 g, 0.105 mol) in water (95 ml) was slowly added to the reaction mixture and stirred for another 2 hours. The solvent from the reaction mixture was evaporated under reduced pressure to obtain an oily residue. The oily mass was co-evaporated with xylene (2 x 60 ml) to obtain thick mass. This mass was partitioned between 1 : 1 mixture of dichloromethane (300 ml) and water (300 ml). The organic layer was separated and the aqueous layer re-extracted with dichloromethane (150 ml). The combined organic extracts were washed with water (3 x 150 ml) and dried over anhydrous sodium sulphate. The solvent was evaporated under reduced pressure and the resulting oily mass was triturated with ether (3 x 60 ml). Each time the ether layer was decanted and the residue was finally concentrated under reduced pressure to obtain the sticky mass. The so obtained material was purified by column chromatography over silica gel using mixture of methanol and dichloromethane as elution solvent. The solvent from the combined fractions was evaporated to obtain 47.5 g of (25,5i?)-N-(2-Boc-aminoethoxy)-6-(sulfooxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1 ]octane-2-carboxamide,tetrabutyl ammonium salt as white foam in 70% yield.

Analysis:

Mass: 423.4 (M-l) as free sulphonic acid; for Molecular Weight of 665.9 and Molecular Formula of C30H59N5O9 S;

1H- NMR (400MHz, CDC13): δ 9.52(br s, 1H), 5.53(br s, 1H), 4.33(s, 1H), 3.95-3.92(m,3H), 3.37-3.27(m, 1 1H), 2.87-2.84(d, 1H), 2.35-2.30(m, 1H), 2.17(m, 1H), 1.96-1.88(m, 2H), 1.74-1.60(m,8 H), 1.47-1.40(m, 17H), 1.02-0.98(m, 12H).

Step 4: Preparation of (2S R)-iV-(2-aminoethoxy)-6-(sulfooxy)-7-oxo-l,6-diaza-bicyclo[3.2.1]octane-2-carboxamide (I):

Formula (I)

To a stirred solution of (2S,5i?)-N-(2-Boc-aminoethoxy)-6-(sulfooxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1 ]octane-2-carboxamide, tetrabutyl ammonium salt (VI, 17 g, 0.0225 mol) in dichloromethane (85 ml) was added trifluoroacetic acid (85 ml) drop wise at -10°C over a period of 45 minutes. The resulting mass was further stirred at same temperature for 1 hour. The resulting reaction mixture was poured into cyclohexane (850 ml), stirred well for 30 minutes and the separated oily layer was collected. This procedure was repeated one more time and finally the separated oily layer was added to tert-butyl methyl ether (170 ml) under vigorous stirring at 25°C. The ether layer was removed by decantation from the precipitated solid. This procedure was repeated twice again with tert-butyl methyl ether (2 x 170 ml). The solid thus obtained was stirred with fresh dichloromethane (170 ml) for 30 minutes and filtered. The residual solid was dried at 45°C under reduced pressure to yield 7.3g of the titled compound in crude form. The obtained solid was further dissolved in water, (7.3 ml) and to this solution was added basic resin (Amberlyst A-26 -OH ion exchange resin, 4.4 g) under stirring. After 0.5 hour, the resin was filtered and to the filtrate isopropanol (51 ml) was added slowly at 25°C. The solution was further stirred for 12 hours. The separated solid was filtered and washed with additional isopropanol (7.5 ml) and dried under reduced pressure to obtain 4.3 g of (2S ,5R)-N-(2-aminoethoxy)-6-(sulfooxy)-7-oxo-l ,6-diaza-bicyclo[3.2.1 ]octane-2-carboxamide as off-white solid in 52 % yield.

Analysis:

Mass: 323.1 (M-l); for Molecular Weight of 324.31 and Molecular Formula of C9H16N4O7S; 1H-NMR (400MHz, D20): δ 4.07-4.06(d, 1H), 4.05-4.03(t, 2H), 3.96-3.94(d, 1H), 3.20(br s, 1H), 3.16-3.13(t, 2H), 3.02-2.99(d, 1H), 2.04-1.68(m, 4H);

Purity as determined by HPLC: 94.88%.

REF

http://www.pewtrusts.org/~/media/assets/2015/02/antibioticsinnovationproject_datatable_201502_v3.pdf?la=en

WO2015110969A3 * Jan 21, 2015 Nov 26, 2015 Wockhardt Limited Nitrogen containing compounds and their use as antibacterial agents
WO2015150941A1 * Mar 12, 2015 Oct 8, 2015 Wockhardt Limited A process for preparation of sodium (2s, 5r)-6-(benzyloxy)-7-oxo-1,6-diazabicyclo[3.2.1]octane-2-carboxylate
WO2016088863A1 * Dec 4, 2015 Jun 9, 2016 Meiji Seikaファルマ株式会社 Method for producing crystals of diazabicyclooctane derivative and stable lyophilized preparation
EP2931723A4 * Dec 11, 2012 Jun 1, 2016 Fedora Pharmaceuticals Inc New bicyclic compounds and their use as antibacterial agents and -lactamase inhibitors
US8933232 Mar 29, 2013 Jan 13, 2015 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole beta-lactamase inhibitors
US8933233 Mar 29, 2013 Jan 13, 2015 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole β-lactamase inhibitors
US8940897 Mar 29, 2013 Jan 27, 2015 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole β-lactamase inhibitors
US8962843 Mar 29, 2013 Feb 24, 2015 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole beta-lactamase inhibitors
US8962844 Mar 29, 2013 Feb 24, 2015 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole β-lactamase inhibitors
US9120795 Mar 14, 2014 Sep 1, 2015 Cubist Pharmaceuticals, Inc. Crystalline form of a β-lactamase inhibitor
US9120796 Oct 2, 2014 Sep 1, 2015 Cubist Pharmaceuticals, Inc. B-lactamase inhibitor picoline salt
US9309245 Apr 2, 2013 Apr 12, 2016 Entasis Therapeutics Limited Beta-lactamase inhibitor compounds
US9393239 Apr 15, 2014 Jul 19, 2016 Fedora Pharmaceuticals Inc. Bicyclic compounds and their use as antibacterial agents and betalactamase inhibitors

/////////////IN2015MU287, WO-2016120752, nacubactam, WOCKHARDT, NEW PATENT, WK ?, WK-?, WK?,  CAS 1452458-86-4C9 H16 N4 O7 S, 324.31, Beta lactamase inhibitor, Roche, Meiji Seika Pharma,  Fedora Pharmaceuticals, nacubactam hydrate , PHASE 1, A diazabicyclooctane beta-lactamase inhibitor, bacterial infection, July 2016,  phase 1 clinical development, RG-6080, 1452458-86-4, FPI-1459,  OP-0595, Phase I ,  β-lactamase inhibitor, bacterial infections, Fedora parmaceuticals, Meiji Seika Pharma

NCCONC(=O)[C@@H]2CC[C@@H]1C[N@]2C(=O)N1OS(=O)(=O)O


Filed under: PHASE 1, PHASE1, Uncategorized Tagged: 1452458-86-4, 324.31, A diazabicyclooctane beta-lactamase inhibitor, bacterial infection, Beta lactamase inhibitor, C9 H16 N4 O7 S, CAS 1452458-86-4, Fedora Pharmaceuticals, FPI-1459, IN2015MU287, July 2016, Meiji Seika Pharma, nacubactam, nacubactam hydrate, NEW PATENT, OP-0595, PHASE 1, phase 1 clinical development, RG-6080, Roche, WK ?, WO-2016120752, Wockhardt

DOLUTEGRAVIR, ドルテグラビルナトリウム

$
0
0

STR1

 

Dolutegravir.svgDolutegravir ball-and-stick model.png

Dolutegravir

ドルテグラビルナトリウム
  • Soltegravir

2H-Pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide, N-[(2,4-difluorophenyl)methyl]-3,4,6,8,12,12a-hexahydro-7-hydroxy-4-methyl-6,8-dioxo-, (4R,12aS)

(3R,11aS)—N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide

(4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide
Trade Name:Tivicay
Synonym:GSK1349572, S-349572, GSK572
Date of Approval: August 12, 2013 (US)
Indication:HIV infection
Drug class: Integrase strand transfer inhibitor
Company: ViiV Healthcare,GlaxoSmithKline

INNOVATOR …ViiV Healthcare 
CAS number: 1051375-16-6

1051375-19-9 (Dolutegravir Sodium)

MF:C20H19F2N3O5
MW:419.4

2H-Pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide, N-[(2,4-difluorophenyl)methyl]-3,4,6,8,12,12a-hexahydro-7-hydroxy-4-methyl-6,8-dioxo-, (4R,12aS)- [ACD/Index Name]
GSK 1349572
S-349572

Chemical Name: (4R,12aS)-N-[(2,4-difluorophenyl)methyl]-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a- hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide
Patent: US8129385
Patent expiration date: Oct 5, 2027
PCT patent application: W02006116764

ドルテグラビルナトリウム
Dolutegravir Sodium

C20H18F2N3NaO5 : 441.36
[1051375-19-9]

Dolutegravir (DTG, GSK1349572) is an integrase inhibitor being developed for the treatment of human immunodeficiency virus (HIV)-1 infection by GlaxoSmithKline (GSK) on behalf of Shionogi-ViiV Healthcare LLC. DTG is metabolized primarily by uridine diphosphate glucuronyltransferase (UGT)1A1, with a minor role of cytochrome P450 (CYP)3A, and with renal elimination of unchanged drug being extremely low (< 1% of the dose).

Dolutegravir sodium was approved by the U.S. Food and Drug Administration (FDA) on Aug 12, 2013, then approved by European Medicine Agency (EMA) on Jan 16, 2014, and approved by Pharmaceuticals and Medical Devices Agency of Japan (PMDA) on Mar 24, 2014, then approved by Center For Drug Evaluation (CFDA) on Dec 30, 2015. It was co-developed by GlaxoSmithKline & ViiV Healthcare Corporation, then marketed as Tivicay® by ViiV Healthcare in the US and EU and by GlaxoSmithKline & ViiV Healthcare Corporation in JP.

Dolutegravir sodium is an integrase inhibitor which blocks HIV replication by preventing the viral DNA from integrating into the genetic material of human immune cells (T-cells). This step is essential in the HIV replication cycle and is also responsible for establishing chronic infection. It is in combination with other antiretroviral agents for the treatment of HIV-1 infection in adults and children aged 12 years and older and weighing at least 40 kg.

Tivicay® is available as film-coated tablet for oral use, containing 50 mg of free Dolutegravir. The recommended dose is 50 mg Dolutegravir once daily without regards to meals.

APPROVALS

Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2013-08-12 Marketing approval Tivicay HIV infection Tablet, Film coated Eq. 50 mg Dolutegravir ViiV Priority
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2014-01-16 Marketing approval Tivicay HIV infection Tablet, Film coated 50 mg ViiV
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2014-03-24 Marketing approval Tivicay HIV infection Tablet, Film coated 50 mg ViiV, GlaxoSmithKline
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2015-12-30 Marketing approval Tivicay/特威凯 HIV infection Tablet 50 mg GlaxoSmithKline

CLIP

The European Commission has on 21 January 2014 Dolutegravir (Tivicay, ViiV) permit as part of combination therapy for the treatment of HIV-infected persons over the age of 12 years.Dolutegravir (Tivicay, ViiV) is an integrase inhibitor, in combination with other antiretroviral drugs in adults and adolescents can be used from 12 years for the treatment of HIV infection.

Source: Communication from the European Commission

Dolutegravir[1] is a FDA-approved drug[2] for the treatment of HIV infection. Dolutegravir is an integrase inhibitor. Known as S/GSK1349572 or just “572” the drug is marketed as Tivicay[3] by GlaxoSmithKline (GSK). In February, 2013 the Food and Drug Administration announced that it would fast track dolutegravir’s approval process.[4] On August 13, 2013, dolutegravir was approved by the FDA. On November 4, 2013, dolutegravir was approved by Health Canada.[5]

The oral HIV integrase inhibitor S-349572 was originated by Shionogi-GlaxoSmithKline and Shionogi-ViiV Healthcare. In 2013, the product was approved and launched in the U.S. for the treatment of HIV-1 in adults and children aged 12 years and older, in combination with other antiretroviral agents. A positive opinion was received in the E.U for this indication and, in 2014, approval was attained in Europe for this indication. Registration is pending in Japan.

In 2013, orphan drug designation in Japan was assigned to the compound.

Dolutegravir is approved for use in a broad population of HIV-infected patients. It can be used to treat HIV-infected adults who have never taken HIV therapy (treatment-naïve) and HIV-infected adults who have previously taken HIV therapy (treatment-experienced), including those who have been treated with other integrase strand transfer inhibitors. Tivicay is also approved for children ages 12 years and older weighing at least 40 kilograms (kg) who are treatment-naïve or treatment-experienced but have not previously taken other integrase strand transfer inhibitors.[6]

Dolutegravir has also been compared head-to-head with a preferred regimen from the DHHS guidelines in each of the three classes (i.e. 1.) nuc + non-nuc, 2.) nuc + boosted PI, and 3.) nuc + integrase inhibitor).

SPRING-2 compared dolutegravir to another integrase inhibitor, raltegravir, with both coformulated with a choice of TDF/FTC orABC/3TC. After 48 weeks of treatment 88% of those on dolutegravir had less than 50 copies of HIV per mL compared to 85% in the raltegravir group, thus demonstrating non-inferiority.[9]

The FLAMINGO study has been presented at scientific meetings but as of early 2014 has not yet been published. It is an open-label trial of dolutegravir versus darunavir boosted with ritonavir. In this trial 90% of those on dolutegravir based regimens had viral loads < 50 at 48 weeks compared to 83% in the darunavir/r.[10] This 7% difference was statistically significant for superiority of the dolutegravir based regimens.

Another trial comparing dolutegravir to efavirenz, SINGLE, was the first trial to show statistical superiority to an efavirenz/FTC/TDF coformulated regimen for treatment naive patients.[11] After 48 weeks of treatment, 88% of the dolutegravir group had HIV RNA levels < 50 copies / mL versus 81% of the efavirenz group. This has led one commentator to predict that it may replace efavirenz as the first line choice for initial therapy as it can also be formulated in one pill, once-a-day regimens.[12]

Doultegravir has also been studied in patients who have been on previous antiretroviral medications. The VIKING trial looked at patients who had known resistance to the first generation integrase inhibitor raltegravir. After 24 weeks 41% of patients on 50mg dolutegravir once daily and 75% of patients on 50mg twice daily (both along with an optimized background regimen) achieved an HIV RNA viral load of < 50 copies per mL. This demonstrated that there was little clinical cross-resistance between the two integrase inhibitors. [13]

Dolutegravir (also known as S/GSK1349572), a second-generation integrase inhibitor under development by GlaxoSmithKline and its Japanese partner Shionogi for the treatment of HIV infection, was given priority review status from the US Food and Drug Administration (FDA) in February, 2013.

GlaxoSmithKline  marketed the first HIV drug Retrovir in 1987 before losing out to Gilead Sciences Inc. (GILD) as the world’s biggest maker of AIDS medicines. The virus became resistant to Retrovir when given on its own, leading to the development of therapeutic cocktails.

The new once-daily drug Dolutegravir, which belongs to a novel class known as integrase inhibitors that block the virus causing AIDS from entering cells, is owned by ViiV Healthcare, a joint venture focused on HIV in which GSK is the largest shareholder.

Raltegravir (brand name Isentress) received approval by the U.S. Food and Drug Administration (FDA) on 12 October 2007, the first of a new class of HIV drugs, the integrase inhibitors, to receive such approval. it is a potent and well tolerated antiviral agent.  However, it has the limitations of twice-daily dosing and a relatively modest genetic barrier to the development of resistance, prompting the search for agents with once-daily dosing.

Elvitegravir, approved by the FDA on August 27, 2012 as part of theelvitegravir/cobicistat/tenofovir disoproxil fumarate/emtricitabine fixed-dose combination pill (Quad pill, brand name Stribild) has the benefit of being part of a one-pill, once-daily regimen, but suffers from extensive cross-resistance with raltegravir.

STR1DOLUTEGRAVIR

Gilead’s Atripla (Emtricitabine/Tenofovir/efavirenz), approved in 2006 with loss of patent protection in 20121, is the top-selling HIV treatment. The $3.2 billion medicine combines three drugs in one pill, two compounds that make up Gilead’s Truvada (Emtricitabine/Tenofovir) and Bristol- Myers Squibb Co.’s Sustiva (Efavirenz).

A three-drug combination containing dolutegravir and ViiV’s older two-in-one treatment Epzicom(Abacavir/Lamivudine, marketed outside US as Kivexa) proved better than Gilead’s market-leading Atripla  in a clinical trial released in July, 2012 (See the Full Conference Report Here), suggesting it may supplant the world’s best-selling AIDS medicine as the preferred front-line therapy. In the latest Phase III study, after 48 weeks of treatment, 88% of patients taking the dolutegravir-based regimen had reduced viral levels to the goal compared with 81% of patients taking Atripla. More patients taking Atripla dropped out of the study because of adverse events compared with those taking dolutegravir — 10% versus just 2% — which was the main driver of the difference in efficacy. The result was the second positive final-stage clinical read-out for dolutegravir, following encouraging results against U.S. company Merck & Co’s rival Isentress in April, 2012 (See the Conference Abstract Here)..

Dolutegravir is viewed by analysts as a potential multibillion-dollar-a-year seller, as its once-daily dosing is likely to be attractive to patients. The FDA is scheduled to issue a decision on the drug’s approval by August 17。

TIVICAY contains dolutegravir, as dolutegravir sodium, an HIV INSTI. The chemical name of dolutegravir sodium is sodium (4R,12aS)-9-{[(2,4-difluorophenyl)methyl]carbamoyl}-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazin-7-olate. The empirical formula is C20H18F2N3NaO5 and the molecular weight is 441.36 g/mol. It has the following structural formula:

TIVICAY (dolutegravir) Structural Formula Illustration

Dolutegravir sodium is a white to light yellow powder and is slightly soluble in water.

Each film-coated tablet of TIVICAY for oral administration contains 52.6 mg of dolutegravir sodium, which is equivalent to 50 mg dolutegravir free acid, and the following inactive ingredients: D-mannitol, microcrystalline cellulose, povidone K29/32, sodium starch glycolate, and sodium stearyl fumarate. The tablet film-coating contains the inactive ingredients iron oxide yellow, macrogol/PEG, polyvinyl alcohol-part hydrolyzed, talc, and titanium dioxide.

DOLUTEGRAVIR

File:Synthese Dolutegravir.png

http://blog.sina.com.cn/s/blog_de171b9b0101a1ah.html  BELOW

STR1

Dolutegravir Synthesis
Identifications:
1H NMR (Estimated) for Dolutegravir
Experimental: 1H NMR (CDCl3) δ  12.45 (s, 1H), 10.38 (br s, 1H), 8.30 (s, 1H), 7.40-7.30 (m, 1H), 6.85-6.75 (m, 2H), 5.26 (d, J = 5.8, 4.1 Hz, 2H), 5.05-4.95 (m, 1H), 4.64 (d, J = 5.9 Hz, 2H), 4.27 (dd, J = 13.4, 4.2 Hz, 1H), 4.12 (dd, J = 13.6, 6.0 Hz, 1H), 4.05 (t, J = 2.3 Hz, 1H), 4.02 (d, J = 2.2 Hz, 1H), 2.30-2.19 (m, 1H), 1.56 (dd, J = 14.0, 2.0 Hz, 1H), 1.42 (d, J = 7.0 Hz, 3H).

INTRODUCTION

Among viruses, human immunodeficiency virus (HIV), a kind of retrovirus, is known to cause acquired immunodeficiency syndrome (AIDS). The therapeutic agent for AIDS is mainly selected from a group of reverse transcriptase inhibitors (e.g., AZT, 3TC) and protease inhibitors (e.g., Indinavir), but they are proved to be accompanied by side effects such as nephropathy and the emergence of resistant viruses. Thus, the development of anti-HIV agents having the other mechanism of action has been desired.

On the other hand, a combination therapy is reported to be efficient in treatment for AIDS because of the frequent emergence of the resistant mutant. Reverse transcriptase inhibitors and protease inhibitors are clinically used as an anti-HIV agent, however agents having the same mechanism of action often exhibit cross-resistance or only an additional activity. Therefore, anti-HIV agents having the other mechanism of action are desired.

Under the circumstances above, an HIV integrase inhibitor has been focused on as an anti-HIV agent having a novel mechanism of action (Ref: Patent Documents 1 and 2). As an anti-HIV agent having such a mechanism of action, known are carbamoyl-substituted hydroxypyrimidinone derivative (Ref: Patent Documents 3 and 4) and carbamoyl-substituted hydroxypyrrolidione derivative (Ref: Patent Document 5). Further, a patent application concerning carbamoyl-substituted hydroxypyridone derivative has been filed (Ref: Patent Document 6, Example 8).

Other known carbamoylpyridone derivatives include 5-alkoxypyridine-3-carboxamide derivatives and γ-pyrone-3-carboxamide derivatives, which are a plant growth inhibitor or herbicide (Ref: Patent Documents 7-9).

Other HIV integrase inhibitors include N-containing condensed cyclic compounds (Ref: Patent Document 10).

  • [Patent Document 1] WO03/0166275
  • [Patent Document 2] WO2004/024693
  • [Patent Document 3] WO03/035076
  • [Patent Document 4] WO03/035076
  • [Patent Document 5] WO2004/004657
  • [Patent Document 6] JP Patent Application 2003-32772
  • [Patent Document 7] JP Patent Publication 1990-108668
  • [Patent Document 8] JP Patent Publication 1990-108683
  • [Patent Document 9] JP Patent Publication 1990-96506
  • [Patent Document 10] WO2005/016927
  • Patent Document 1 describes compounds (I) and (II), which are useful as anti-HIV drugs and shown by formulae:
    Figure imgb0001
    This document describes the following reaction formula as a method of producing compound (I).
    Figure imgb0002
    Figure imgb0003
    Furthermore, Patent Documents 2 to 6 describe the following reaction formula as an improved method of producing compound (I).
    Figure imgb0004
    Figure imgb0005
        [PATENT DOCUMENTS]
        • [Patent Document 1] International publication No.2006/116764 pamphlet
        • [Patent Document 2] International publication No.2010/011812 pamphlet
        • [Patent Document 3] International publication No.2010/011819 pamphlet
        • [Patent Document 4] International publication No.2010/068262 pamphlet
        • [Patent Document 5] International publication No.2010/067176 pamphlet
        • [Patent Document 6] International publication No.2010/068253 pamphlet
        • [Patent Document 7] US Patent 4769380A
        • [Patent Document 8] International applicationPCT/JP2010/055316

    [NON-PATENT DOCUMENTS]

      • [Non-Patent Document 1] Journal of Organic Chemistry, 1991, 56(16), 4963-4967
      • [Non-Patent Document 2] Science of Synthesis, 2005, 15, 285-387
      • [Non-Patent Document 3] Journal of Chemical Society Parkin Transaction. 1, 1997, Issue. 2, 163-169

A clip and its own references

Dolutegravir sodium (Tivicay®), developed and marketed by GlaxoSmithKline,45 was approved by the FDA in August 2013 as a novel integrase inhibitor for the treatment of HIV infection.46 Dolutegravir was fast-tracked by the FDA in February 2012,47 and joins an important class of drugs known as Integrase Strand Transfer inhibitors (INSTi’s).48 INSTi’s are characterized by their two-metal-chelating scaffolds, which are known to chelate Mg2+ cofactors in the enzyme active site,49, 50 interrupting function of HIV-1 integrase, which is essential for replication of viral DNA into host chromatin.49-51,52 Other drugs in this class, raltegravir and elvitegravir, are known to require either high dosages53 or PK boosting agents,54 respectively, with raltegravir also exhibiting substantial loss of potency in several major HIV-1 integrase mutation pathways.55 Dolutegravir was pursued with the goal of developing a INSTi with a once-daily, low-dosage treatment with improved resistance profile and without the
need for the use of a PK boosting agent.51, 56 Dolutegravir sodium has been approved for treating a broad
population of HIV-infected patients, including adults undergoing their first treatment as well as those
who have been treated with other integrase transfer strand inhibiting agents.46 The most likely process-scale synthesis of dolutegravir sodium, as described in Scheme 8, began with benzyl protection and alkylation of pyrone 46 with benzaldehyde, yielding alcohol 47 in 74% over 2 steps (Scheme 8).57, 58 Alcohol mesylation and in-situ elimination provided the styrenyl olefin 48 in 94% yield, which further underwent an oxidative cleavage of the olefin to generate 49 by sequential addition of RuCl3/NaIO4 and NaClO2 (56% overall yield). Treatment of pyranone 49 with 3-amino-propane-2-diol (50) in ethanol at elevated temperatures delivered the corresponding pyridinone in 83% yield, and this was followed by esterification and sodium periodate-mediated diol cleavage to furnish intermediate 51 in 71% overall yield across the two-step sequence.57, 58 Next, the key ring-forming step in the
synthesis of dolutegravir sodium consisted of cyclization of 51 with (R)-3-amino-butan-1-ol, a process which relies on substrate control to provide the desired tricyclic carbamoylpyridone system 52 in high stereoselectivity (20/1 in favor of the desired isomer).51 Previously, cyclization of systems such as 51 with unsubstituted amino alcohols were found to yield a mixture of diastereomeric products, therefore indicating the pivotal role of the chiral amino alcohol in influencing stereochemical bias during the overall cyclization step.51, 56 In practice, reaction of 51 with (R)-3-amino-butan-1-ol at 90 °C led to isolation of a single cyclization product 52, after recrystallization from EtOAc.57, 58 From 52, Nbromosuccinimide (NBS) bromination and subsequent treatment with amine 53 under palladiumcatalyzed
amidocarbonylative conditions led to amide 54 in 75% yield over 2 steps. Finally, removal of the benzyl group and subsequent crystallization using sodium hydroxide in water and ethanol provided dolutegravir sodium (VII) in 99% yield.57, 58

 

45 Johns, B. A.; Kawasuji, T.; Taishi, T.; Taoda, Y. WO Patent 2006116764A1, 2006.
46. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm364744.htm.
47. https://newdrugapprovals.org/2013/07/16/dolutegravir-biggest-rival-to-worlds-best-selling-hivdrug-atripla-may-get-fda-approval-by-august-2013/.
48. Pendri, A.; Meanwell, N. A.; Peese, K. M.; Walker, M. A. Expert Opin. Ther. Pat. 2011, 21,1173.
49. Johns, B. A.; Svolto, A. C. Expert Opin. Ther. Pat. 2008, 18, 1225.60
50. Johns, B. A.; Weatherhead, J. G.; Allen, S. H.; Thompson, J. B.; Garvey, E. P.; Foster, S. A.;
Jeffrey, J. L.; Miller, W. H. Bioorg. Med. Chem. Lett. 2009, 19, 1802.
51. Johns, B. A.; Kawasuji, T.; Weatherhead, J. G.; Taishi, T.; Temelkoff, D. P.; Yoshida, H.;Akiyama, T.; Taoda, Y.; Murai, H.; Kiyama, R.; Fuji, M.; Tanimoto, N.; Jeffrey, J.; Foster, S.A.; Yoshinaga, T.; Seki, T.; Kobayashi, M.; Sato, A.; Johnson, M. N.; Garvey, E. P.; Fujiwara,
T. J. Med. Chem. 2013, 56, 5901.
52. Kawasuji, T.; Johns, B. A.; Yoshida, H.; Taishi, T.; Taoda, Y.; Murai, H.; Kiyama, R.; Fuji, M.;Yoshinaga, T.; Seki, T.; Kobayashi, M.; Sato, A.; Fujiwara, T. J. Med. Chem. 2012, 55, 8735.
53. Lennox, J. L.; De Jesus, E.; Lazzarin, A.; Pollard, R. B.; Valdez Ramalho Madruga, J.; Berger,D. S.; Zhao, J.; Xu, X.; Williams-Diaz, A.; Rodgers, A. J.; Barnard, R. J. O.; Miller, M. D.; DiNubile, M. J.; Nguyen, B.-Y.; Leavitt, R.; Sklar, P. Lancet 2009, 374, 796.
54. Ramanathan, S.; Mathias, A. A.; German, P.; Kearney, B. P. Clin. Pharmacokinet. 2011, 50,229.
55. Ceccherini-Silberstein, F.; Malet, I.; D’Arrigo, R.; Antinori, A.; Marcelin, A.-G.; Perno, C.-F.AIDS Rev. 2009, 11, 17.
56. Kawasuji, T.; Johns, B. A.; Yoshida, H.; Weatherhead, J. G.; Akiyama, T.; Taishi, T.; Taoda, Y.;Mikamiyama-Iwata, M.; Murai, H.; Kiyama, R.; Fuji, M.; Tanimoto, N.; Yoshinaga, T.; Seki, T.;Kobayashi, M.; Sato, A.; Garvey, E. P.; Fujiwara, T. J. Med. Chem. 2013, 56, 1124.
57. Johns, B. A.; Duan, M.; Hakogi, T. WO Patent 2010068262A1, 2010.
58. Yoshida, H.; Taoda, Y.; Johns, B. A. WO Patent 2010068253A1, 2010.

CLIPS

Dolutegravir synthesis (EP2602260, 2013). LiHMDS as the non-nucleophilic strong base pulling compound 1 carbonyl group proton alpha position with an acid chloride after 2 and ring closure reaction to obtain 3 , 3 via primary amine 4 ring opening ring closure to obtain 5 , NBS the bromine under acidic conditions to obtain aldehyde acetal becomes 6 , 6 of the aldehyde and amino alcohols 7 and turn off the condensation reaction obtained by the ring 8 , alkaline hydrolysis 8 of bromine into a hydroxyl group and hydrolyzable ester obtained 9 after the 10 occurred acid condensation Dolutegravir.

CLIPS

Synthesis of Dolutegravir (S/GSK1349572, GSK1349572)

SYNTHESIS

2H-Pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide, N-[(2,4-difluorophenyl)methyl]-3,4,6,8,12,12a-hexahydro-7-hydroxy-4-methyl-6,8-dioxo-, (4R,12aS) ………..dolutegravir

PATENT

US8129385

STR1 STR2

Figure US08129385-20120306-C00099

Desired isomer

Example Z-1

(3R,11aS)—N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide sodium salt

Figure US08129385-20120306-C00116

a)

(3R,11aS)—N-[(2,4-Difluorophenyl)methyl]-3-methyl-5,7-dioxo-6-[(phenylmethyl)oxy]-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide. To a solution of 16a (409 mg, 0.87 mmol) in dichloroethane (20 mL) was added (2R)-2-amino-1-propanol (0.14 mL, 1.74 mmol) and 10 drops of glacial acetic acid. The resultant solution was heated at reflux for 2 h. Upon cooling, Celite was added to the mixture and the solvents removed in vacuo and the material was purified via silica gel chromatography (2% CH3OH/CH2Clgradient elution) to give (3R,11aS)—N-[(2,4-difluorophenyl)methyl]-3-methyl-5,7-dioxo-6-[(phenylmethyl)oxy]-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (396 mg, 92%) as a glass. 1H NMR (CDCl3) δ 10.38 (m, 1H), 8.42 (s, 1H), 7.54-7.53 (m, 2H), 7.37-7.24 (m, 4H), 6.83-6.76 (m, 2H), 5.40 (d, J=10.0 Hz, 1H), 5.22 (d, J=10.0 Hz, 1H), 5.16 (dd, J=9.6, 6.0 Hz, 1H), 4.62 (m, 2H), 4.41 (m, 1H), 4.33-4.30 (m, 2H), 3.84 (dd, J=12.0, 10.0 Hz, 1H), 3.63 (dd, J=8.4, 7.2 Hz, 1H), 1.37 (d, J=6.0 Hz, 3H); ES+MS: 496 (M+1).

b)

(3R,11aS)—N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide sodium salt. To a solution of (3R,11aS)—N-[(2,4-difluorophenyl)methyl]-3-methyl-5,7-dioxo-6-[(phenylmethyl)oxy]-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (396 mg, 0.80 mmol) in methanol (30 mL) was added 10% Pd/C (25 mg). Hydrogen was bubbled through the reaction mixture via a balloon for 2 h. The resultant mixture was filtered through Celite with methanol and dichloromethane.

The filtrate was concentrated in vacuo to give (3R,11aS)—N-[(2,4-difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide , DOLUTEGRAVIR   as a pink tinted white solid (278 mg, 86%).

1H NMR (CDCl3) δ 11.47 (m, 1H), 10.29 (m, 1H), 8.32 (s, 1H), 7.36 (m, 1H), 6.82 (m, 2H), 5.31 (dd, J=9.6, 3.6 Hz, 1H), 4.65 (m, 2H), 4.47-4.38 (m, 3H), 3.93 (dd, J=12.0, 10.0 Hz, 1H), 3.75 (m, 1H), 1.49 (d, J=5.6 Hz, 3H); ES+ MS: 406 (M+1).

DOLUTEGRAVIR NA SALT

The above material (278 mg, 0.66 mmol) was taken up in ethanol (10 mL) and treated with 1 N sodium hydroxide (aq) (0.66 ml, 0.66 mmol). The resulting suspension was stirred at room temperature for 30 min. Ether was added and the liquids were collected to provide the sodium salt of the title compound as a white powder (291 mg, 99%). 1H NMR (DMSO-d6) δ 10.68 (m, 1H), 7.90 (s, 1H), 7.35 (m, 1H), 7.20 (m, 1H), 7.01 (m, 1H), 5.20 (m, 1H), 4.58 (m, 1H), 4.49 (m, 2H), 4.22 (m, 2H), 3.74 (dd, J=11.2, 10.4 Hz, 1H), 3.58 (m, 1H), 1.25 (d, J=4.4 Hz, 3H).

UNDESIRED ISOMER

Example Z-9

(3S,11aR)—N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide sodium salt

Figure US08129385-20120306-C00124

The title compound was made in two steps using a similar process to that described in example Z-1. 16a (510 mg, 1.08 mmol) and (25)-2-amino-1-propanol (0.17 mL, 2.17 mmol) were reacted in 1,2-dichloroethane (20 mL) with acetic acid to give (3S,11aR)—N-[(2,4-difluorophenyl)methyl]-3-methyl-5,7-dioxo-6-[(phenylmethyl)oxy]-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (500 mg, 93%). This material was hydrogenated in a second step as described in example Z-1 to give (3S,11aR)—N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (386 mg, 94%) as a tinted white solid. 1H NMR (CDCl3) δ 11.46 (m, 1H), 10.28 (m, 1H), 8.32 (s, 1H), 7.35 (m, 1H), 6.80 (m, 2H), 5.30 (dd, J=10.0, 4.0 Hz, 1H), 4.63 (m, 2H), 4.48-4.37 (m, 3H), 3.91 (dd, J=12.0, 10.0 Hz, 1H), 3.73 (m, 1H), 1.48 (d, J=6.0 Hz, 3H); ES+ MS: 406 (M+1). This material (385 mg, 0.95 mmol) was treated with sodium hydroxide (0.95 mL, 1.0 M, 0.95 mmol) in ethanol (15 mL) as described in example Z-1 to provide its corresponding sodium salt (381 mg, 94%) as a white solid. 1H NMR (DMSO-d6) δ 10.66 (m, 1H), 7.93 (s, 1H), 7.33 (m, 1H), 7.20 (m, 1H), 7.01 (m, 1H), 5.19 (m, 1H), 4.59 (m, 1H), 4.48 (m, 2H), 4.22 (m, 2H), 3.75 (m, 1 H), 3.57 (m, 1H), 1.24 (d, J=5.6 Hz, 3H).

SYNTHESIS OF INTERMEDIATES

Figure US08129385-20120306-C00090

IN ABOVE SCHEME SYNTHESIS UPTO COMPD 9 MAY BE USEFUL IN SYNTHESIS BUT READERS DISCRETION IS SOUGHT IN THIS ?????????????????

1) Maltol 1 (189 g, 1.5 mol) was dissolved in dimethylformamide (1890 ml), and benzyl bromide (184 ml, 1.5 mol) was added. After the solution was stirred at 80° C. for 15 minutes, potassium carbonate (228 g, 1.65 mol) was added, and the mixture was stirred for 1 hour. After the reaction solution was cooled to room temperature, an inorganic salt was filtered, and the filtrate was distilled off under reduced pressure. To the again precipitated inorganic salt was added tetrahydrofuran (1000 ml), this was filtered, and the filtrate was distilled off under reduced pressure to obtain the crude product (329 g, >100%) of 3-benzyloxy-2-methyl-pyran-4-one 2 as a brown oil.

NMR (CDCl3) δ: 2.09 (3H, s), 5.15 (2H, s), 6.36 (1H, d, J=5.6 Hz), 7.29-7.41 (5H, m), 7.60 (1H, d, J=5.6 Hz).

2) The compound 2 (162.2 g, 750 mmol) was dissolved in ethanol (487 ml), and aqueous ammonia (28%, 974 ml) and a 6N aqueous sodium hydroxide solution (150 ml, 900 mmol) were added. After the reaction solution was stirred at 90° C. for 1 hour, this was cooled to under ice-cooling, and ammonium chloride (58 g, 1080 mmol) was added. To the reaction solution was added chloroform, this was extracted, and the organic layer was washed with an aqueous saturated sodium bicarbonate solution, and dried with anhydrous sodium sulfate. The solvent was distilled off under reduced pressure, isopropyl alcohol and diethyl ether were added to the residue, and precipitated crystals were filtered to obtain 3-benzyloxy-2-methyl-1H-pyridine-4-one 3 (69.1 g, 43%) as a pale yellow crystal.

NMR (DMSO-d6) δ: 2.05 (3H, s), 5.04 (2H, s), 6.14 (1H, d, J=7.0 Hz), 7.31-7.42 (5H, m), 7.46 (1H, d, J=7.2 Hz), 11.29 (1H, brs).

3) The above compound 3 (129 g, 699 mmol) was suspended in acetonitrile (1300 ml), and N-bromosuccinic acid imide (117 g, 659 mmol) was added, followed by stirring at room temperature for 90 minutes. Precipitated crystals were filtered, and washed with acetonitrile and diethyl ether to obtain 3-benzyloxy-5-bromo-2-methyl-pyridine-4-ol 4 (154 g, 88%) as a colorless crystal.

NMR (DMSO-d6) δ: 2.06 (3H, s), 5.04 (2H, s), 7.32-7.42 (5H, m), 8.03 (1H, d, J=5.5 Hz), 11.82 (1H, brs).

4) To a solution of the compound 4 (88 g, 300 mmol), palladium acetate (13.4 g, 60 mmol) and 1,3-bis(diphenylphosphino)propane (30.8 g, 516 mmol) in dimethylformamide (660 ml) were added methanol (264 ml) and triethylamine (210 ml, 1.5 mol) at room temperature. The interior of a reaction vessel was replaced with carbon monoxide, and the material was stirred at room temperature for 30 minutes, and stirred at 80 degree for 18 hours. A vessel to which ethyl acetate (1500 ml), an aqueous saturated ammonium chloride solution (1500 ml) and water (1500 ml) had been added was stirred under ice-cooling, and the reaction solution was added thereto. Precipitates were filtered, and washed with water (300 ml), ethyl acetate (300 ml) and diethyl ether (300 ml) to obtain 5-benzyloxy-4-hydroxy-6-methyl-nicotinic acid methyl ester 5 (44.9 g, 55%) as a colorless crystal.

NMR (DMSO-d6) δ: 2.06 (3H, s), 3.72 (3H, s), 5.02 (2H, s), 7.33-7.42 (5H, m), 8.07 (1H, s).

5) After a solution of the compound 5 (19.1 g, 70 mmol) in acetic anhydride (134 ml) was stirred at 130° C. for 40 minutes, the solvent was distilled off under reduced pressure to obtain 4-acetoxy-5-benzyloxy-6-methyl-nicotinic acid methyl ester 6 (19.9 g, 90%) as a flesh colored crystal.

NMR (CDCl3) δ: 2.29 (3H, s), 2.52 (3H, s), 3.89 (3H, s), 4.98 (2H, s), 7.36-7.41 (5H, m), 8.85 (1H, s).

6) To a solution of the compound 6 (46.2 g, 147 mmol) in chloroform (370 ml) was added metachloroperbenzoic acid (65%) (42.8 g, 161 mmol) in portions under ice-cooling, and this was stirred at room temperature for 90 minutes. To the reaction solution was added a 10% aqueous potassium carbonate solution, and this was stirred for 10 minutes, followed by extraction with chloroform. The organic layer was washed with successively with a 10% aqueous potassium carbonate solution, an aqueous saturated ammonium chloride solution, and an aqueous saturated sodium chloride solution, and dried with anhydrous sodium sulfate. The solvent was distilled off under induced pressure, and the residue was washed with diisopropyl ether to obtain 4-acetoxy-5-benzyloxy-6-methyl-1-oxy-nicotinic acid methyl ester 7 (42.6 g, 87%) as a colorless crystal.

NMR (CDCl3) δ: 2.30 (3H, s), 2.41 (3H, s), 3.90 (3H, s), 5.02 (2H, s), 7.37-7.39 (5H, m), 8.70 (1H, s).

7) To acetic anhydride (500 ml) which had been heated to stir at 130° C. was added the compound 7 (42.6 g, 129 mmol) over 2 minutes, and this was stirred for 20 minutes. The solvent was distilled off under reduced pressure to obtain 4-acetoxy-6-acetoxymethyl-5-benzyloxy-nicotinic acid methyl ester 8 (49.6 g, >100%) as a black oil.

NMR (CDCl3) δ: 2.10 (3H, s), 2.28 (3H, s), 3.91 (3H, s), 5.07 (2H, s), 5.20 (2H, s), 7.35-7.41 (5H, m), 8.94 (1H, s).

8) To a solution of the compound 8 (46.8 g, 125 mmol) in methanol (140 ml) was added a 2N aqueous sodium hydroxide solution (376 ml) under ice-cooling, and this was stirred at 50° C. for 40 minutes. To the reaction solution were added diethyl ether and 2N hydrochloric acid under ice-cooling, and precipitated crystals were filtered. Resulting crystals were washed with water and diethyl ether to obtain 5-benzyloxy-4-hydroxy-6-hydroxymethyl-nicotinic acid 9 (23.3 g, 68%) as a colorless crystal.

NMR (DMSO-d6) δ: 4.49 (2H, s), 5.19 (2H, s), 5.85 (1H, brs), 7.14-7.20 (2H, m), 7.33-7.43 (7H, m), 8.30 (1H, s), 10.73 (1H, t, J=5.8 Hz), 11.96 (1H, brs).

9) To a solution of the compound 9 (131 g, 475 mmol), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (219 g, 1140 mmol) and 1-hydroxybenzotriazole (128 g, 950 mmol) in dimethylformamide (1300 ml) was added 4-fluorobenzylamine (109 ml, 950 mmol), and this was stirred at 80° C. for 1.5 hours. After the reaction solution was cooled to room temperature, hydrochloric acid was added, followed by extraction with ethyl acetate. The extract was washed with a 5% aqueous potassium carbonate solution, an aqueous saturated ammonium chloride solution, and an aqueous saturated sodium chloride solution, and dried with anhydrous sodium sulfate. The solvent was distilled off under reduced pressure to obtain a mixture (175 g) of 10 and 11. the resulting mixture was dissolved in acetic acid (1050 ml) and water (1050 ml), and zinc (31.1 g, 475 mmol) was added, followed by heating to reflux for 1 hour. After the reaction solution was cooled to room temperature, a 10% aqueous potassium carbonate solution was added, followed by extraction with ethyl acetate. The extract was washed with an aqueous saturated ammonium chloride solution, and an aqueous saturated sodium chloride solution, and dried with anhydrous sodium sulfate. After the solvent was distilled off under reduced pressure, this was washed with diethyl ether to obtain 5-benzyloxy-N-(4-fluoro-benzyl)-4-hydroxy-6-hydroxymethyl-nicotinic acid amide 10 (107 g, 59%) as a colorless crystal.

NMR (DMSO-d6) δ: 4.45 (2H, d, J=4.3 Hz), 4.52 (2H, d, J=5.8 Hz), 5.09 (2H, s), 6.01 (1H, brs), 7.36-7.43 (5H, m), 8.31 (1H, s), 12.63 (1H, brs).

PATENT

SYNTHESIS

EP2602260A1

STR1

Example 3

Figure imgb0128

3H IS DOLUTEGRAVIR

Step 1

N,N-dimethylformamide dimethyl acetal (4.9 ml, 36.5 mmol) was added dropwise to compound 3A (5.0 g, 30.4 mmol) under cooling at 0°C. After stirring at 0°C for 1 hour, 100 ml of ethyl acetate was added to the reaction solution, and the organic layer was washed with a 0.5 N aqueous hydrochloric acid solution (50 ml). The aqueous layer was separated, followed by extraction with ethyl acetate (50 ml). The organic layers were combined, washed with a saturated aqueous solution of sodium bicarbonate and saturated saline in this order, and then dried over anhydrous sodium sulfate. The solvent was distilled off, and the obtained residue was purified by silica gel column chromatography (n-hexane-ethyl acetate: 1:1 (v/v) → ethyl acetate) to obtain 4.49 g (yield: 67%) of compound 3B as an oil.

1H-NMR (CDCl3)δ:1.32 (3H, t, J = 7.1 Hz), 2.90 (3H, br s), 3.29 (3H, br s), 4.23 (2H, q, J = 7.1 Hz), 4.54 (2H, s), 7.81 (1H, s).

Step 2

Lithium hexamethyldisilazide (1.0 M solution in toluene, 49 ml, 49.0 mmol) was diluted with tetrahydrofuran (44 ml). A tetrahydrofuran (10 ml) solution of compound 3B (4.49 g, 20.4 mmol) was added dropwise thereto under cooling at -78°C, and a tetrahydrofuran (10 ml) solution of ethyl oxalyl chloride (3.35 g, 24.5 mmol) was then added dropwise to the mixture. The mixture was stirred at -78°C for 2 hours and then heated to 0°C. 2 N hydrochloric acid was added to the reaction solution, and the mixture was stirred for 20 minutes, followed by extraction with ethyl acetate (200 ml x 2). The organic layer was washed with a saturated aqueous solution of sodium bicarbonate and saturated saline and then dried over anhydrous sodium sulfate. The solvent was distilled off, and the obtained residue was purified by silica gel column chromatography (n-hexane-ethyl acetate: 7:3 → 5:5 → 0:10 (v/v)) to obtain 1.77 g (yield: 31%) of compound 3C as a white solid.

1H-NMR (CDCl3)δ:1.36-1.46 (6H, m), 4.35-4.52 (8H, m), 8.53 (1H, s).

Step 3

Aminoacetaldehyde dimethyl acetal (0.13 ml, 1.20 mmol) was added to an ethanol (6 ml) solution of compound 3C (300 mg, 1.09 mmol) at 0°C, and the mixture was stirred at 0°C for 1.5 hours, then at room temperature for 18 hours, and at 60°C for 4 hours. The solvent in the reaction solution was distilled off under reduced pressure, and the obtained residue was then purified by silica gel column chromatography (n-hexane-ethyl acetate: 5:5 → 0:10 (v/v)) to obtain 252 mg (yield: 64%) of compound 3D as an oil.

1H-NMR (CDCl3)δ:1.36-1.47 (6H, m), 3.42 (6H, s), 3.90 (2H, d, J = 5.2 Hz), 4.37 (3H, q, J = 7.2 Hz), 4.50 (2H, q, J = 7.2 Hz), 8.16 (1H, s).

Step 4

62% H2SO4 (892 mg, 5.64 mmol) was added to a formic acid (10 ml) solution of compound 3D (1.02 g, 2.82 mmol), and the mixture was stirred at room temperature for 16 hours. The formic acid was distilled off under reduced pressure. To the residue, methylene chloride was added, and the mixture was pH-adjusted to 6.6 by the addition of a saturated aqueous solution of sodium bicarbonate. The methylene chloride layer was separated, while the aqueous layer was subjected to extraction with methylene chloride. The methylene chloride layers were combined and dried over anhydrous sodium sulfate. The solvent was distilled off to obtain 531.8 mg of compound 3E as a yellow oil.

1H-NMR (CDCl3) δ: 1.28-1.49 (6H, m), 4.27-4.56 (4H, m), 4.84 (2H, s), 8.10 (1H, s), 9.72 (1H, s).

Step 5

Methanol (0.20 ml, 5.0 mmol), (R)-3-amino-butan-1-ol (179 mg, 2.0 mmol), and acetic acid (0.096 ml, 1.70 mmol) were added to a toluene (5 ml) solution of compound 3E (531 mg, 1.68 mmol), and the mixture was heated to reflux for 4 hours. The reaction solution was cooled to room temperature, then diluted with chloroform, and then washed with a saturated aqueous solution of sodium bicarbonate. The aqueous layer was subjected to extraction with chloroform. The chloroform layers were combined, washed with saturated saline, and then dried over anhydrous sodium sulfate. The solvent was distilled off, and the obtained residue was purified by silica gel column chromatography (chloroform-methanol: 100:0 → 90:10) to obtain 309.4 mg of compound 3F as a brown oil.

1H-NMR (CDCl3) δ: 1.40 (3H, t, J = 7.1 Hz), 1.40 (3H, d, J = 7.1 Hz), 1.55-1.61 (1H, m), 2.19-2.27 (1H, m), 4.00 (1H, d, J = 1.5 Hz), 4.03 (1H, d, J = 2.5 Hz), 4.10 (1H, dd, J = 13.2, 6.3 Hz), 4.26 (1H, dd, J = 13.2, 3.8 Hz), 4.38 (2H, q, J = 7.1 Hz), 5.00-5.05 (1H, m), 5.31 (1H, dd, J = 6.4, 3.9 Hz), 8.10 (1H, s).

Step 6

Potassium trimethylsilanolate (333 mg, 2.34 mmol) was added to a 1,2-dimethoxyethane (2 ml) solution of compound 3F (159 mg, 0.47 mmol), and the mixture was stirred at room temperature for 7 hours. 1 N hydrochloric acid and saturated saline were added to the reaction solution, followed by extraction with chloroform. The chloroform layers were combined and dried over anhydrous sodium sulfate. The solvent was distilled off to obtain 34.4 mg (yield: 25%) of compound 3G as an orange powder.

1H-NMR (CDCl3) δ: 1.46 (3H, d, J = 3.5 Hz), 1.58-1.65 (1H, m), 2.26-2.30 (1H,m), 4.06-4.10 (2H, m), 4.31 (1H, dd, J = 13.8, 5.6 Hz), 4.48 (1H, dd, J = 13.6, 3.9 Hz), 5.03 (1H, t, J = 6.4 Hz), 5.36 (1H, dd, J = 5.5, 4.0 Hz), 8.44 (1H, s), 12.80 (1H, s), 14.90 (1H, s).

Step 7

Compound 3G (16 mg, 0.054 mmol) and 2,4-difluorobenzylamine (17 mg, 0.12 mmol) were dissolved in N,N-dimethylformamide (1 ml). To the solution, N,N,N’,N’-tetramethyl-O-(7-aza-benzotriazol-1-yl)uronium hexafluorophosphate (HATU) (53 mg, 0.14 mmol) and N-methylmorpholine (0.031 ml, 0.28 mmol) were added, and the mixture was stirred at room temperature for 16 hours. 2,4-difluorobenzylamine (17 mg, 0.12 mmol), HATU (64 mg, 0.17 mmol), and N-methylmorpholine (0.037 ml, 0.34 mmol) were further added thereto, and the mixture was stirred at room temperature for additional 16 hours. 0.5 N hydrochloric acid was added to the reaction solution, followed by extraction with ethyl acetate. The ethyl acetate layers were combined, washed with 0.5 N hydrochloric acid and then with saturated saline, and then dried over anhydrous sodium sulfate. The solvent was distilled off, and the obtained residue was purified by preparative high-performance liquid chromatography to obtain 12.5 mg (yield: 55%) of compound 3H as an orange solid.

DOLUTEGRAVIR

1H-NMR (DMSO-d6) δ: 1.36 (3H, d, J = 6.9 Hz), 1.55-1.60 (1H, m), 2.01-2.05 (1H, m), 3.92-3.94 (1H, m), 4.04 (1H, t, J = 12.6 Hz), 4.38-4.41 (1H, m), 4.57-4.60 (1H, m), 4.81-4.83 (1H, m), 5.46-5.49 (1H, m), 7.08-7.11 (1H, m), 7.25-7.30 (1H, m), 7.41 (1H, dd, J = 15.3, 8.7 Hz), 8.53 (1H, s), 10.38 (1H, s), 12.53 (1H, s).

ISOMERS OF DOLUTEGRAVIR

Reference Example 1

Figure imgb0145

Figure imgb0146

Step 1

Acetic acid (180 mg, 3.00 mmol) was added to a toluene (90 ml) solution of compound A-1 (4.39 g, 9.33 mmol) and (R)-3-aminobutan-1-ol (998 mg, 11.2 mmol), and the mixture was stirred at 50°C for 90 minutes. The reaction solution was allowed to cool to room temperature and then poured to a saturated aqueous solution of sodium bicarbonate. The organic layer was separated, while the aqueous layer was subjected to extraction three times with ethyl acetate. The combined extracts were washed with saturated saline and then dried over sodium sulfate. The solvent was distilled off to obtain 4.29 g of crude product A-2.

Step 2

The crude product A-2 obtained in the preceding step was dissolved in ethanol (40 ml). To the solution, a 2 N aqueous sodium hydroxide solution (20 ml) was added at room temperature, and the mixture was stirred at the same temperature for 2 hours. The reaction solution was neutralized to pH 7 using a 2 N aqueous hydrochloric acid solution. The solvent was directly distilled off. The obtained crude product A-3 was subjected to azeotropy with toluene (100 ml) and used in the next step without being purified.

Step 3

HOBt (1.65 g, 12.2 mmol) and WSC HCl (2.34 g, 12.2 mmol) were added at room temperature to a DMF (100 ml) solution of the crude product A-3 obtained in the preceding step, and the mixture was stirred at the same temperature for 15 hours. Water was added to the reaction solution, followed by extraction three times with ethyl acetate. The combined extracts were washed with water three times and then dried over sodium sulfate. The solvent was distilled off, and the obtained oil was subjected to silica gel column chromatography for purification. Elution was performed first with n-hexane-ethyl acetate (3:7, v/v) and then with only ethyl acetate. The fraction of interest was concentrated, and the obtained oil was then dissolved in ethyl acetate. The solution was crystallized with diisopropyl ether as a poor solvent. The obtained crystals were collected by filtration and dissolved again in ethyl acetate. The solution was recrystallized to obtain 1.84 g of compound A-4.

1HNMR (CDCl3) δ: 1.49 (3H, d, J = 6.6 Hz), 1.88-1.96 (1H, m), 2.13-2.26 (1H, m), 3.90-4.17 (4H, m), 4.42-4.47 (1H, m), 4.63 (2H, d, J = 6.0 Hz), 5.12-5.17 (1H, m), 5.17 (1H, d, J = 9.9 Hz), 5.33 (1H, d, J = 9.9 Hz), 6.77-6.87 (2H, m), 7.27-7.42 (4H, m), 7.59-7.62 (2H, m), 8.35 (1H, s), 10.41 (1H, t, J = 5.7 Hz).

Step 4

The compound A-4 was subjected to the hydroxy deprotection reaction described in Step F of the paragraph [0088] to obtain compound A-5.

1HNMR (DMSO-d6) δ:1.41 (3H, d, J = 6.3 Hz), 1.85-1.92 (1H, m), 1.50-1.75 (1H, m), 4.02-4.09 (3H, m), 4.28-4.34 (1H, m), 4.53 (2H, d, J = 5.7 Hz), 4.64 (1H, dd, J = 3.9 Hz, 12.6 Hz), 5.45 (1H, dd, J = 3.6 Hz, 9.3 Hz), 7.06 (1H, ddd, J = 2.7 Hz, 8.4 Hz, 8.4 Hz), 7.20-7.28 (1H, m), 7.35-7.42 (1H, m), 8.43 (1H, s),10.37 (1H, t, J = 6.0 Hz),12.37 (1H, brs).

Reference Example 2

Figure imgb0147

Compound A-1 was reacted with (S)-3-aminobutan-1-ol in Step 1. Compound B-5 was obtained in the same way as in Reference Example 1.

  • 1HNMR (DMSO-d6) δ:1.41 (3H, d, J = 6.3 Hz), 1.85-1.92 (1H, m), 1.50-1.75 (1H, m), 4.02-4.09 (3H, m), 4.28-4.34 (1H, m), 4.53 (2H, d, J = 5.7 Hz), 4.64 (1H, dd, J = 3.9 Hz, 12.6 Hz), 5.45 (1H, dd, J = 3.6 Hz, 9.3 Hz), 7.06 (1H, ddd, J = 2.7 Hz, 8.4 Hz, 8.4 Hz), 7.20-7.28 (1H, m), 7.35-7.42 (1H, m), 8.43 (1H, s),10.37 (1H, t, J = 6.0 Hz),12.37 (1H, brs).

PATENT

W02006116764

Figure imgf000122_0001

ENTRY 68

PATENT

WO 2010068262

STR1

PATENT

WO 2010068253

PATENT

WO 2011119566

PATENT

Synthesis

WO 2012018065

Example 3

Figure JPOXMLDOC01-appb-C000176

I was under cooling added dropwise at 0 ℃ (4.9 ml, 36.5 mmol) and N, N-dimethylformamide dimethyl acetal (5.0 g, 30.4 mmol) in the first step compound 3A. After stirring for 1 hour at 0 ℃, ethyl acetate was added to 100ml, the reaction mixture was washed with 0.5N aqueous hydrochloric acid (50 ml). Was extracted with ethyl acetate (50ml) and solution was separated and the aqueous layer. The organic layers were combined, washed successively with saturated aqueous sodium bicarbonate solution and saturated brine, and then dried over anhydrous sodium sulfate. After the solvent was distilled off, silica gel column chromatography and the residue obtained was – and purified by (n-hexane (v / v) → ethyl acetate 1:1) to an oil (67% yield) of Compound 3B 4.49 g I got a thing.
1 H-NMR (CDCl 3)δ: 1.32 (3H, t, J = 7.1 Hz), 2.90 (3H, br s), 3.29 (3H, br s), 4.23 (2H, q, J = 7.1 Hz), 4.54 (2H, s), 7.81 (1H, s).
Diluted with tetrahydrofuran (44 ml) (1.0M toluene solution, 49 ml, 49.0 mmol) the second step lithium hexamethyldisilazide, under cooling at -78 ℃, compound 3B (4.49 g, 20.4 mmol) in this After dropwise tetrahydrofuran (10 ml) was added dropwise tetrahydrofuran (3.35 g, 24.5 mmol) of ethyl oxalyl chloride and (10 ml) solution. After stirring for 2 hours at -78 ℃, I was warmed to 0 ℃. After washing (200 ml x 2), saturated aqueous sodium bicarbonate solution and the organic layer with saturated brine After stirring for 20 minutes, extracted with ethyl acetate by adding 2N hydrochloric acid, the reaction solution was dried over anhydrous sodium sulfate. After removal of the solvent, silica gel column chromatography and the residue obtained – was purified (n-hexane (v / v) ethyl acetate 7:3 → 5:5 → 0:10), compound 3C 1.77 g (yield I as a white solid 31%).
1 H-NMR (CDCl 3)δ :1.36-1 .46 (6H, m), 4.35-4.52 (8H, m), 8.53 (1H, s).
Was added at 0 ℃ (0.13 ml, 1.20 mmol) the aminoacetaldehyde dimethyl acetal ethanol (300 mg, 1.09 mmol) of the third step compound 3C to (6 ml) solution, 1 hour and 30 minutes at 0 ℃, 18 hours at room temperature , then I was stirred for 4 hours at 60 ℃. After the solvent was evaporated under reduced pressure and the reaction mixture by silica gel column chromatography and the residue obtained was – and purified by (n-hexane (v / v) ethyl acetate 5:5 → 0:10), compound 3D 252 mg (yield: I got as an oil 64%) rate.
1 H-NMR (CDCl 3)δ :1.36-1 .47 (6H, m), 3.42 (6H, s), 3.90 (2H, d, J = 5.2 Hz), 4.37 (3H, q, J = 7.2 Hz), 4.50 (2H, q, J = 7.2 Hz), 8.16 (1H, s).
Was added (892 mg, 5.64 mmol) and 2 SO 4 62-H% formic acid (1.02 g, 2.82 mmol) in a fourth step the compound for 3D (10 ml) solution was stirred at room temperature for 16 hours. Methylene chloride was added to the residue Shi distilled off under reduced pressure and formic acid was adjusted to pH = 6.6 by addition of saturated aqueous sodium bicarbonate. The solution was separated methylene chloride layer was extracted with methylene chloride and the aqueous layer. I was dried over anhydrous sodium sulfate combined methylene chloride layers. The solvent was then distilled off and was obtained as a yellow oil 531.8 mg compound 3E.
1H-NMR (CDCl3) δ: 1.28-1.49 (6H, m), 4.27-4.56 (4H, m), 4.84 (2H, s), 8.10 (1H, s), 9.72 (1H, s).
Amino – – butane – 1 – ol (179 mg, 2.0 mmol), methanol (0.20 ml, 5.0 mmol), (R) -3 toluene (531 mg, 1.68 mmol) in the fifth step to compound 3E (5 ml) solution was added (0.096 ml, 1.70 mmol) acetic acid was heated under reflux for 4 hours. After dilution with chloroform, cooled to room temperature, the reaction mixture was washed with a saturated aqueous sodium bicarbonate solution, and the aqueous layer was extracted with chloroform. After washing with saturated brine combined chloroform layer was dried over anhydrous sodium sulfate. The solvent was then distilled off, silica gel column chromatography and the residue obtained – and (chloroform methanol 100:0 → 90:10), was obtained as a brown oil 309.4 mg compound 3F.
1H-NMR (CDCl3) δ: 1.40 (3H, t, J = 7.1 Hz), 1.40 (3H, d, J = 7.1 Hz), 1.55-1.61 (1H, m), 2.19-2.27 (1H, m), 4.00 (1H, d, J = 1.5 Hz), 4.03 (1H, d, J = 2.5 Hz), 4.10 (1H, dd, J = 13.2, 6.3 Hz), 4.26 (1H, dd, J = 13.2, 3.8 Hz ), 4.38 (2H, q, J = 7.1 Hz), 5.00-5.05 (1H, m), 5.31 (1H, dd, J = 6.4, 3.9 Hz), 8.10 (1H, s).
1,2 (159 mg, 0.47 mmol) in the sixth step compound 3F – was added (333 mg, 2.34 mmol) and potassium trimethylsilanolate dimethoxyethane (2 ml) solution was stirred for 7 hours at room temperature. Brine was added to the 1N-hydrochloric acid to the reaction mixture, followed by extraction with chloroform. The combined chloroform layer was dried over anhydrous sodium sulfate. The solvent was removed by distillation, and I as an orange powder (25% yield) of compound 3G 34.4 mg.
1H-NMR (CDCl3) δ: 1.46 (3H, d, J = 3.5 Hz), 1.58-1.65 (1H, m), 2.26-2.30 (1H, m), 4.06-4.10 (2H, m), 4.31 (1H , dd, J = 13.8, 5.6 Hz), 4.48 (1H, dd, J = 13.6, 3.9 Hz), 5.03 (1H, t, J = 6.4 Hz), 5.36 (1H, dd, J = 5.5, 4.0 Hz) , 8.44 (1H, s), 12.80 (1H, s), 14.90 (1H, s).
2,4 (16 mg, 0.054 mmol) and the seventh step compound 3G – was dissolved in N, N-dimethylformamide (1 ml) (17 mg, 0.12 mmol) difluorobenzyl amine, N, N, N ‘, N was added (0.031 ml, 0.28 mmol) and N-methylmorpholine uronium hexafluorophosphate (HATU) (53 mg, 0.14 mmol), and ‘- tetramethyl-O-(yl 7 – aza – – benzo triazolopyrimidine -1) I was stirred at room temperature for 16 h. 2,4 – was added (0.037 ml, 0.34 mmol) and N-methylmorpholine (64 mg, 0.17 mmol) and (17 mg, 0.12 mmol), HATU difluorobenzylamine, and the mixture was stirred for 16 hours at room temperature. I was extracted with ethyl acetate addition of 0.5N-hydrochloric acid to the reaction mixture. 0.5N-hydrochloric acid and then was washed with saturated brine, and dried over anhydrous sodium sulfate and combined ethyl acetate layer. The solvent was then distilled off, and purified by preparative high performance liquid chromatography residue was obtained as an orange solid (55% yield) of compound 3H 12.5 mg.
1H-NMR (DMSO-d6) δ: 1.36 (3H, d, J = 6.9 Hz), 1.55-1.60 (1H, m), 2.01-2.05 (1H, m), 3.92-3.94 (1H, m), 4.04 (1H, t, J = 12.6 Hz), 4.38-4.41 (1H, m), 4.57-4.60 (1H, m), 4.81-4.83 (1H, m), 5.46-5.49 (1H, m), 7.08-7.11 (1H, m), 7.25-7.30 (1H, m), 7.41 (1H, dd, J = 15.3, 8.7 Hz), 8.53 (1H, s), 10.38 (1H, s), 12.53 (1H, s)

PAPER

http://pubs.acs.org/doi/abs/10.1021/jm400645w

Carbamoyl Pyridone HIV-1 Integrase Inhibitors 3. A Diastereomeric Approach to Chiral Nonracemic Tricyclic Ring Systems and the Discovery of Dolutegravir (S/GSK1349572) and (S/GSK1265744)

GlaxoSmithKline Research & Development, Infectious Diseases Therapeutic Area Unit, Five Moore Drive, Research Triangle Park, North Carolina 27709, United States
Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
J. Med. Chem., 2013, 56 (14), pp 5901–5916
DOI: 10.1021/jm400645w

J. Med. Chem. 2013, 56, 5901-5916.

Abstract Image

We report herein the discovery of the human immunodeficiency virus type-1 (HIV-1) integrase inhibitors dolutegravir (S/GSK1349572) (3) and S/GSK1265744 (4). These drugs stem from a series of carbamoyl pyridone analogues designed using a two-metal chelation model of the integrase catalytic active site. Structure–activity studies evolved a tricyclic series of carbamoyl pyridines that demonstrated properties indicative of once-daily dosing and superior potency against resistant viral strains. An inherent hemiaminal ring fusion stereocenter within the tricyclic carbamoyl pyridone scaffold led to a critical substrate controlled diastereoselective synthetic strategy whereby chiral information from small readily available amino alcohols was employed to control relative and absolute stereochemistry of the final drug candidates. Modest to extremely high levels of stereochemical control were observed depending on ring size and position of the stereocenter. This approach resulted in the discovery of 3 and 4, which are currently in clinical development.

STR1

(4R,12aS)-N-(2,4-Difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino-
[2,1-b][1,3]oxazine-9-carboxamide (3). 1H NMR (CDCl3) δ 12.45 (s, 1H),10.38 (br s, 1H), 8.30 (s, 1H), 7.40−7.30 (m, 1H), 6.85−6.75 (m, 2H),5.26 (d, J = 5.8, 4.1 Hz, 2H), 5.05−4.95 (m, 1H), 4.64 (d, J = 5.9 Hz,2H), 4.27 (dd, J = 13.4, 4.2 Hz, 1H), 4.12 (dd, J = 13.6, 6.0 Hz, 1H), 4.05(t, J = 2.3 Hz, 1H), 4.02 (d, J = 2.2 Hz, 1H), 2.30−2.19 (m, 1H), 1.56(dd, J = 14.0, 2.0 Hz, 1H), 1.42 (d, J = 7.0 Hz, 3H). ES+ LC/MS: m/zcalcd 419.13; found 420.13 (M + 1)+.
(4R,12aS)-N-(2,4-Difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino-
[2,1-b][1,3]oxazine-9-carboxamide (3) sodium salt.

1H NMR(DMSO-d6) δ 10.70 (t, J = 6.0 Hz, 1H), 7.89 (s, 1 H), 7.40−7.30 (m, 1H), 7.25−7.16 (m, 1H), 7.06−6.98 (m, 1H), 5.22−5.12 (m, 1H), 4.87−4.74 (m, 1H), 4.51 (d, J = 5.4 Hz, 2H), 4.35−4.25 (m, 1 H), 4.16 (dd, J =1.8, 14.1 Hz, 1 H), 4.05−3.90 (m, 1H), 3.86−3.74 (m, 1 H), 2.00−1.72(m, 1 H), 1.44−1.32 (m, 1 H), 1.24 (d, J = 6.9 Hz, 3H).

STR1

MORE UPDATES……………………………

Process for preparing integrase inhibitors such as dolutegravir and cabotegravir and their analogs, useful for treating viral infections eg HIV infection. Also claims a process for preparing intermediates of dolutegravir and cabotegravir.

(4R, 12aS)-N-[(2,4-Difluorophenyl)methyl]-3 ,4,6,8, 12, 12a-hexahydro-7-hydroxy-4-methyl-6,8-dioxo-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1-b][1 ,3]oxazine-9-carboxamide (Formula A):

Formula A

known by the INN name dolutegravir, is a new efficient antiviral agent from the group of HIV integrase inhibitors which is used in combination with some other antiviral agents for treatment of HIV infections, such as AIDS. The compound, which belongs to condensed polycyclic pyridines and was first disclosed in WO2006/1 16764, is marketed.

Another compound disclosed in WO2006/1 16764 is (3S, 1 1 aR)-N-[(2,4-difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, 1 1 ,1 1 a-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-d]pyrazine-8-carboxamide (Formula

Formula C

known by the INN name cabotegravir.

The complex structures of dolutegravir and cabotegravir present a synthetic challenge. The first description of the synthesis in WO2006/1 16764 shows a 16-steps synthesis (see Scheme A), which is industrially impractical due to its length and low overall yield.

Scheme A

WO 2010/068253 and WO 2006/1 16764 describe an alternative synthesis. The 1 1 -step synthesis, shown in Scheme B1 and Scheme B2, is based on bromination of the 9-position for further introduction of the carboxylic group. The synthesis relies on the use of expensive palladium catalysts and toxic selenium compounds. Furthermore, some variations of these approaches involve pyrone intermediates in several steps. In some cases pyrones are liquids which can complicate purification, while further reactions form complex mixtures.

doiutegravir

Scheme B2

In further alternative syntheses, acetoacetates were used as starting materials. Such an approach is challenging in terms of introducing the hydroxy group in the 7-position. The variation in Scheme C1 , described in WO2012/018065, starts from 4-benzyloxyacetoacetate. The procedure requires 9 steps, but use expensive reagents like palladium catalysts. Moreover, there is described a possibility of formation a co-crystal between an intermediate and hydroquinone, wherein however the additional step may diminish yields and make the process longer and time consuming.

Scheme C1

The variation in Scheme C2, described in WO2012/018065, starts from 4-chloroacetoacetate. The process is not optimal because of problems in steps which include pyrones and because of problems with conversion of 7-chloro to 7-hydroxy group which includes a disadvantageous use of silanolates with low yield (25%).

Scheme C2

The variation in Scheme C3, described in WO201 1/1 19566, starts from unsubstituted acetoacetate. For the introduction of the 7-hydroxy group, bromination is used and substitution of bromo with hydroxy is performed by a use of silanolates. The substitution of the bromine is achieved in a 43% yield.

Scheme C3

The variation in Scheme C4, described in WO201 1/1 19566, starts from 4-methoxyacetoacetate aiming at preparing dolutegravir or cabotegravir. The process uses lithium bases to affect a difficult to control selective monohydrolysis of a diester.

PATENT

WO 2016113372

Carbotegravir, New Patent, WO 2016113372, Lek Pharmaceutical and Chemical Co DD

LEK PHARMACEUTICALS D.D. [SI/SI]; Verovskova 57 1526 Ljubljana (SI)

MARAS, Nenad; (SI).
SELIC, Lovro; (SI).
CUSAK, Anja; (SI)

ViiV Healthcare is developing cabotegravir (first disclosed in WO2006088173), which in July 2016, was reported to be in phase 2 clinical development.

WO-2016113372

The object of the present invention is to provide short, simple, cost-effective, environmentally friendly and industrially suitable processes for beneficially providing dolutegravir and analogues thereof and cabotegravir and analogues thereof, in particular dolutegravir.

Scheme 1

According to an embodiment of the process of the invention the building block 3-aminobutanol can suitably be substituted with other aminoalcohols to give dolutegravir analogues. For example, using (S)-alaninol gives cabotegravir as the final product. Similarly, using amines other than 2,4-difluorobenzylamine in the amidation step results in the synthesis of other dolutegravir analogues.

According to the another preferred embodiment cabotegravir or a pharmaceutically acceptable salt thereof is prepared by the analogue process, which comprises providing a compound of formula (5c)

5c

converting the compound of formula (5c) to a compound of formula (6c)

6c

by carrying out a chlorination reaction, and converting the compound of formula (6c) to cabotegravir and/or a pharmaceutically acceptable salt thereof.

The compound of formula (5c) can preferably be provided by converting a compound of formula (3) to a compound of formula (4c)

Scheme 2

1. ) EtOCOCI, Et3N / Me2CO

2. ) 2,4-difiuorobenzylamine

Scheme 3

Analogous compound of formula 7c is a useful intermediate in the synthesis of cabotegravir. Scheme 3a

Scheme 4

Examples

The following examples are merely illustrative of the present invention and they should not be considered as limiting the scope of the invention in any way. The examples and modifications or other equivalents thereof will become apparent to those versed in the art in the light of the present entire disclosure. Particularly, all Examples related to the preparation of dolutegravir and intermediates thereof can be used by the analogy for the preparation of cabotegravir and intermediates thereof.

Example 1 :

Methyl acetoacetate (1 , 25.22 g) and dimethylformamide dimethyl acetal (DMFDMA, 35 mL) was heated at 50-55°C for 2 h, then methanol (60 mL), aminoacetaldehyde dimethyl acetal (24 mL) and acetic acid (4 mL) was added an the mixture was heated under reflux for one hour, then concentrated. MTBE (100 mL) was added and the mixture was kept at 5 °C overnight to crystallize. Upon filtration 46 g (92%) of product 2 was recovered.

1H NMR (DMSO-d6): δ 2.31 (s, 3H), 3.30 (s, 6H), 3.49 (m, 2H), 3.61 (s, 3H), 4.43 (m, 1 H), 8.02 (d, 1 H), 10.8 (bs, 1 H). 13C NMR (DMSO-d6): δ 30.52, 35.48, 50.53, 54.23, 98.99, 102.47, 160.70, 166.92, 197.21 .

Example 2:

Compound 2 (5.00 g) was dissolved in 2-propanol, dimethyl oxalate (7.02 g) was added and heated to 40 °C. Sodium methylate (25% in methanol; 20 mL) was slowly (10 min) added, the mixture was then heated to 50-55 °C and stirred at that temperature for 2-2.5 h. The mixture was cooled to ambient temperature, then sodium hydroxide solution (1 M, 65 mL) was added to the mixture and stirred for another 2 h, followed by addition of concentrated hydrochloric acid (1 1 mL) and stirred for another 2 h. The precipitate was filtered and dried to give 8.08 g (NMR assay 47%; 65% yield) of compound 3.

1H NMR (DMSO-d6): δ 2.50 (m, 2H), 3.30 (s. 6H), 4.49 (m, 1 H), 7.06 (s, 1 H); 8.70 (s, 1 H). 13C NMR (DMSO-d6): δ 55.23, 55.37, 102.34, 1 15.47, 120.24, 145.17, 162.71 , 165.22, 178.55.

Example 3:

Compound 2 (158.37 g) was dissolved in methanol (548 mL), followed by the addition of dimethyl oxalate (202.2 g). While keeping the temperature below 30°C, potassium ferf-butoxide (192.1 g) was added and reaction mixture was heated at 50 °C overnight. The suspension was then filtered and the filter cake washed with methanol. The filtrate was concentrated (approximately to 680 mL), then water (680 mL) was added, followed by addition of lithium hydroxide hydrate (143.7 g) while keeping the temperature below 40 °C. The suspension was then stirred at ambient temperature overnight and filtered. To the obtained filtrate, concentrated hydrochloric acid (339 mL) was added while keeping the temperature below 30 °C. The suspension was aged for 2 h and filtered to give 4 as a white powder (95.6 g, NMR assay 100%; 52% yield).

Example 4:

Compound 2 (5.00 g) was dissolved in 2-propanol, dimethyl oxalate (7.02 g) was added and heated to 40 °C. Sodium methylate (25% in methanol; 15 mL) was slowly (10 min) added then the mixture was heated to 50-55 °C and stirred at that temperature for 72 h. The mixture was concentrated and components were separated by flash column chromatography (ethyl acetate/methanol 9:1 to 6:4). Early fractions gave compound 22 upon concentration, late fractions gave compound 23.

Compound 22: 1H NMR (DMSO-d6): δ 2.49 (m, 2H), 3.28 (s, 6H), 3.73 (s, 3H), 3.85 (s, 3H), 4.41 (m, 1 H), 4.50 (m, 1 H), 6.65 (s, 1 H), 8.36 (s, 1 H). 13C NMR (DMSO-d6): δ 51.63, 53.36, 54.25, 55.47, 102.71 , 1 18.24, 123.60, 140.81 , 150.21 , 162.44, 164.49, 173.43.

Compound 23: 1H NMR (DMSO-d6): δ 2.49 (m, 2H), 3.26 (s, 6H); 3.70 (s, 3H); 4.33 (d, 1 H); 4.60 (m, 1 H), 6.19 (s, 1 H), 8.12 (s, 1 H). 13C NMR (DMSO-d6): δ 50.03, 51.34, 54.59, 54.85, 102.91 , 1 16.04, 1 18.19, 148.32, 152.12, 163.46, 165.24, 174.99

Example 5:

Compound 3 (5.5 g; assay 53%) was suspended in acetonitrile, acetic acid (6 mL) and methanesulfonic acid (2.5 mL) were added followed by the heating of mixture to 70 °C for 4 h. The suspension was filtered and filtrate cooled to ambient temperature. Triethylamine (6.6 mL) and (R)-3-amino-butan-1 -ol (1.24 mL) was added followed by heating the mixture at reflux temperature for 20-24 h. The mixture was filtered, filtrate concentrated and 1 M HCI (100 mL) was added, followed by extraction with dichloromethane (3 x 50 mL). Combined organic fractions were concentrated, 2-propanol was added (10 mL) and suspension was stirred at 70-80 °C for 10 min, left to cool to ambient temperature then filtered to give 2.19 g of compound 4 (73%).

1H NMR (DMSO-de): δ 1.31 (d, 3H), 1.52 (m, 1 H), 1 .97 (m, 1 H), 3.89 (m, 1 H), 4.01 (m, 1 H), 4.46 (m, 1 H), 4.64 (m, 1 H), 4.78 (m, 1 H), 5.50 (m, 1 H), 7.29 (s, 1 H), 8.88 (s, 1 H), 15.83 (s, 1 H). 13C NMR (DMSO-d6): δ 15.22, 29.14, 45.26, 51.13, 62.09, 76.03, 1 16.31 , 1 18.79, 140.53, 146.79, 155.36, 165.24, 178.75.

Example 6:

Compound 3 (14.55 g; assay 49%) was suspended in acetonitrile (125 mL), acetic acid (15 mL) and methanesulfonic acid (6.25 mL) were added followed by the heating of mixture to 70 °C for 4 h. The suspension was filtered and filtrate cooled to ambient temperature. Triethylamine (16.5 mL) and (S)-2-aminopropanol (2.45 mL) was added followed by heating the mixture at reflux temperature for 24 h. The insoluble product was filtered, washed with 2-propanol (20 mL) and dried to give (3S, 1 1 aR)-3-methyl-5,7-dioxo-2,3,5,7, 1 1 ,1 1 a-hexahydrooxazolo[3,2-a]pyrido[1 ,2-d]pyrazine-8-carboxylic acid (5.2 g, 75%).

1H NMR (DMSO-d6): δ 1.31 (d, J = 6.3 Hz, 3H), 3.65 (dd, J = 8.6, 6.8 Hz, 1 H), 4.13 (dd, J = 1 1.7, 10.3 Hz, 1 H), 4.28 (m, 1 H), 4.39 (dd, J = 8.6, 6.8 Hz, 1 H), 4.92 (dd, J = 12.3, 4.2 Hz, 1 H), 5.45 (dd, J = 10.2, 4.1 Hz, 1 H), 7.16 (s, 1 H), 8.84 (s, 1 H), 15.74 (s, 1 H).

Example 7:

Compound 4 (0.63 g) was dissolved in dichloromethane (15 mL), cooled to 5°C, then triethylamine (0.31 mL) was added, followed by ethyl chloroformate (0.26 mL), followed by slow (30 min) addition of 2,4-difluorobenzylamine. The mixture was then stirred at ambient temperature for 24 h. Water (10 mL) was added, organic phase was separated and washed with 1 M HCI (15 mL) and water (15 mL), concentrated and treated with 2-propanol to give the product 5 in a quantitative yield.

1H NMR (CDCI3): δ 1.39 (d, 3H), 1.52 (s, 1 H), 2.19 (m, 1 H), 4.00 (m, 2H), 4.16 (m, 1 H), 4.31 (m, 1 H), 4.62 (d, 2H), 5.00 (m, 1 H), 5.27 (m, 1 H), 6.80 (m 2H), 7.33 (m, 2H), 8.49 (s, 1 H), 10.48 (s, 1 H). 13C NMR (CDCI3): 15.50, 29.22, 36.43, 45.19, 51.83, 62.79, 103.71 , 103.91 , 1 1 1 .0, 1 1 1 .18, 120.59, 123.04, 130.40, 137.41 , 144.58, 156.27, 163. 87, 177.83.

Example 8:

To a suspension of 4 (2.84 g, 10 mmol) in a mixture of triethylamine (2.24 mL, 16 mmol) and acetone (50 mL) stirring on an ice bath was added ethyl chloroformate (1 .20 mL, 12 mmol). After stirring for 10 min, 2,4-difluorobenzylamine (1.21 mL, 10 mmol) was added and the mixture left stirring at room temperature for 1 h. The product was isolated by slowly diluting the reaction mixture with water (50 mL), partial concentration, filtration, washing with water (2 50 mL) and drying. There was obtained 5 as a white powder (3.48 g, 86%): mp 181.0-184.7 °C.1H NMR (DMSO-d6): δ 1.29 (d, J = 7.0 Hz, 3H), 1 .56 (dd, J = 13.9, 2.0 Hz, 1 H), 1 .93-2.06 (m, 1 H), 3.90 (ddd, J = 1 1.6, 5.0, 2.1 Hz, 1 H), 3.98 (td, J = 12.0, 2.2 Hz, 1 H), 4.45 (dd, J = 13.6, 6.6 Hz, 1 H), 4.72 (dd, J = 13.6, 3.8 Hz, 1 H), 4.74-4.81 (m, 1 H), 5.44 (dd, J = 6.6, 3.8 Hz, 1 H), 8.93 (s, 1 H), 15.14 (s, 1 H). 13C NMR (DMSO-d6): δ 15.78, 29.13, 44.89, 52.88, 61 .63, 75.61 , 1 13.54, 128.49, 136.42, 145.64, 154.62, 164.58, 174.58

Example 9:

To a suspension of 4 (1 1.36 g, 40 mmol) in acetonitrile (80 mL) stirring at room temperature was added TCCA (9.29 g, 38 mmol) and DABCO (0.23 g, 5 mol%). After stirring at room temperature for 1 h, the reaction was quenched with a mixture of DMSO (5.26 mL) and water (1.33 mL). The insoluble cyanuric acid was removed by filtration and the filtrate evaporated under reduced pressure to give viscous oil. This was triturated in methanol (20 mL) to induce crystallization. The product was filtered, washed with cold methanol (10 mL) and dried to give 7 as a yellowish powder (5.13 g, 41 %): mp 191 .3-198.7 °C.

Example 10:

Attempted chlorination of 23: Compound 23 (0.54g) was suspended in acetonitrile (10 mL) and trichlorocyanuric acid (0.44 g) was added and the solution was stirred at ambient temperature overnight. Precipitate was filtered. Only traces of a product corresponding to the compound 26 could be detected in the reaction mixture by LC-MS analysis. Conversion did not improve with time.

Example 11 :

Attempted chlorination of 3: Compound 3 (0.30 g) was suspended in acetonitrile (5 mL) and trichlorocyanuric acid (0.13 g) was added. The suspension was stirred at ambient temperature overnight. Only traces of a product corresponding to the compound 24 could be detected in the reaction mixture by LC-MS analysis.

Example 12:

9 10

Trichloroisocyanuric acid (0.23 g) was added in a single portion to a stirred solution of the diethyl 1 -(2,2-dimethoxyethyl)-4-oxo-1 ,4-dihydropyridine-2,5-dicarboxylate (9, 0.66 g) in dry acetonitrile (4 mL) at room temperature. The resulting suspension was stirred at room temperature for ca. 24 h. The reaction mixture was diluted with dichloromethane and filtrated. The filtrate was then concentrated in vacuo to afford crude oil (0.86 g). Purification by flash chromatography (eluting ethyl acetate/cyclohexane) furnished diethyl 3-chloro-1 -(2,2-dimethoxyethyl)-4-oxo-1 ,4-dihydropyridine-2,5-dicarboxylate, 10 as a yellow semi-solid (0.38 g). 1H NMR (CDCI3): δ 1.28 (t, J=7A Hz, 3H), 1 .37 (t, J=7.2 Hz, 3H), 3.35 (s, 6H), 3.89 (d, J=5.0 Hz, 2H), 4.27 (q, J=l A Hz, 2H), 4.43 (q, J=l A Hz, 2H), 4.48 (t, J=4.9 Hz, 1 H), 8.15 (s, 1 H). 13C NMR (CDCI3): δ 13.83, 14.13, 55.82, 57.09, 61.41 , 63.72, 102.52, 1 17.35, 126.90, 140.22, 146.92, 160.67, 164.13, 168.95.

Example 13:

Diethyl 1 -(2,2-dimethoxyethyl)-4-oxo-1 ,4-dihydropyridine-2,5-dicarboxylate (9, 0.64 g) was dissolved in anhydrous acetonitrile (6 mL) and treated sequentially with acetic acid (560 μί) and methanesulfonic acid (40 μί). The resulting mixture was heated to 62 °C and stirred for 4 h and more methanesulfonic acid (40 μΙ_) was added. After additional 2 h, more methanesulfonic acid (80 μΙ_) was added. This was repeated after additional 2 h, when more methanesulfonic acid (80 μΙ_) was added. The reaction mixture was stirred additional 17 h at 62 °C then was treated with a mixture of (R)-3-aminobutanol (0.22 g), triethylamine (0.5 mL) and acetonitrile (0.7 mL). The reaction mixture was stirred additional 22 h at 62 °C and then concentrated in vacuo. The crude material was partitioned between dichloromethane and 1 M HCI solution (15 mL). The combined organic phases were dried (Na2S04), filtered and concentrated in vacuo to afford the crude (4R, 12aS)-ethyl 4-methyl-6,8-dioxo-3,4,6,8, 12,12a-hexahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1 -b][1 ,3]oxazine-9-carboxylate (11 ) as a brownish oil (0.61 g).

1H NMR (CD3OD): δ 8.44 (s, 1 H), 7.16 (m, 1 H), 5.48 (t, J=4.8 Hz, 1 H), 4.86 (m, 1 H), 4.49 (dd, J=13.6, 4.0 Hz, 1 H), 4.30-4.25 (m, 3H), 4.09 (dt, J=12.1 , 2.3 Hz, 1 H), 3.96 (ddd, J=1 1.7, 5.0, 2.1 Hz, 1 H), 2.18-2.10 (m, 1 H), 1.60-1 .56 (m, 1 H) 1 .39 (d, J=7A Hz, 3H), 1.33 (t, J=7A Hz, 3H). 13C NMR (CDCI3): δ 8.45, 14.08, 15.39, 29.17, 45.04, 45.72, 51 .56, 60.86, 62.61 , 76.33, 1 19.54, 123.72, 136.96, 145.67, 156.26, 163.68, 175.43

Example 14:

10

Diethyl 3-chloro-1 -(2,2-dimethoxyethyl)-4-oxo-1 ,4-dihydropyridine-2,5-dicarboxylate (10, 1.23 g) was dissolved in 85% formic acid (25 mL) at room temperature. The mixture was warmed to 40 °C and stirred for 23 h. The reaction mixture was concentrated in vacuo, and then partitioned between dichloromethane and aqueous NaHC03 solution. The combined organic phases were dried (Na2S04), filtered and concentrated in vacuo to afford brownish oil (0.49 g). The crude oil was dissolved in anhydrous toluene (5 mL) and treated sequentially with (R)-3-aminobutanol (0.19 g), methanol (0.2 mL) and acetic acid (96 μί). The resulting mixture was heated to 90 °C and stirred for 20 h. The reaction mixture was cooled to room temperature and then partitioned between dichloromethane and aqueous NaHC03 solution. The combined organic phases were dried (Na2S04), filtered and concentrated in vacuo to afford the crude (4R,12aS)-Ethyl 7-chloro-4-methyl-6,8-dioxo-3,4,6,8,12, 12a-hexahydro-2H-pyrido[1 ‘,2’:4,5] pyrazino [2, 1-b][1 ,3]oxazine-9-carboxylate (12) as a brownish oil (0.24 g).

Example 15:

To a solution of 4 (5.68 g, 20 mmol) in dichloromethane (50 mL) stirring in an ice bath was added triethylamine (5.6 mL, 40 mmol), followed by ethyl chloroformate (2.61 mL, 26 mmol). After 20 min, ethanol (50 mL) was added. The mixture was then left stirring 24 h at room temperature and concentrated under reduced pressure. The residue was triturated in acetone (80 mL). The insoluble salt (triethylamine hydrochloride) was removed by filtration. The filtrate was evaporated under reduced pressure to give 11 as an amorphous solid in a quantitative yield (6.1 g).

Example 16:

To a stirring solution of 11 (0.94 g, 3.0 mmol) in acetonitrile (8 mL) heated at 40 °C was added TCCA in portions during 1 h (0.44 g, 1 .8 mmol). After an additional 1 h, the reaction mixture was diluted with a solution of NaHS03 (0.60 g) in water (60 mL), extracted with dichloromethane (50 mL) and the extract evaporated under reduced pressure to give a crude product which was purified by flash chromatography (CH2CI2 : MeOH, from 98 : 2 to 80 : 20) to give 12 (0.45 g, 44%).

1H NMR (CDCI3): δ 1.37 (t, J = 7.1 Hz, 3H), 1.38 (d, J = 7.0 Hz, 3H), 1 .56 (dq, J = 13.9, 2.2 Hz, 1 H), 2.21 (m, 1 H), 3.99 (d, J = 2.3 Hz, 1 H), 4.00 (t, J = 1.8 Hz, 1 H), 4.10 (dd, J = 13.2, 6.6 Hz, 1 H), 4.37-4.27 (m, 3H), 4.98 (m, 1 H), 5.35 (dd, J = 6.6, 3.8 Hz, 1 H), 8.07 (s, 1 H).

13C NMR (CDCI3): δ 14.20, 16.09, 29.34, 44.87, 53.73, 61.49, 62.29, 76.01 , 1 16.22, 133.1 1 , 134.18, 144.52, 155.48, 163.88, 169.98.

Example 17:

To a mixture of 7 (3.89 g, 12.2 mmol) in methanol (12 mL) was added sodium methylate (22.3 mL, 97.6 mmol). The reaction mixture was stirred for 24 h at 30 °C and then quenched with a slow addition of 3M hydrochloric acid (35 mL) while stirring in an ice bath. The mixture was concentrated under reduced pressure to remove most of the methanol, then extracted with dichloromethane (2 30 mL), the combined extracts washed with water (30 mL) and evaporated under reduced pressure. Methanol (20 mL) was added to the obtained amorphous residue and removed under reduced pressure to yield the solid 8 (3.69 g, 98%).

1H NMR (CDCI3): δ 15.04 (s, 1 H), 8.42 (s, 1 H), 5.29 (dd, J=5.6, 3.9 Hz, 1 H), 5.01 -4.96 (m, 1 H), 4.42 (dd, J=13.6, 3.6 Hz, 1 H), 4.25 (dd, J=13.6, 6.0 Hz, 1 H), 4.05 (s, 3H), 4.00-3.97 (m, 2H), 2.21 -2-14 (m, 1 H), 1.53 (dd, J=14.1 , 1.9 Hz, 1 H), 1.36 (d, J=7 Hz, 3H). 13C NMR (CDCI3): δ 176.35, 165.94, 155.03, 153.70, 143.08, 130.90, 1 15.94, 76.05, 62.65, 61.45, 53.86, 44.96, 29.43, 16.06.

Example 18:

To a suspension of 7 (2.55 g, 8.0 mmol) in a mixture of triethylamine (1 .46 mL, 10.4 mmol) and acetone (32 mL) stirring on an ice bath was added ethyl chloroformate (0.88 mL, 8.8 mmol). After stirring for 10 min, 2,4-difluorobenzylamine (1.07 mL, 8.8 mmol) was added and the mixture left stirring at room temperature for 1 h. The product was isolated by slowly diluting the reaction mixture with water (40 mL), filtration, washing with water (2 30 mL) and drying. There was obtained 2.91 g of 6 as a white powder (83%).

1H NMR (CDCI3): δ 1.30 (d, J = 7.0 Hz, 3H), 1 .49 (dd, J = 14.0, 2.2 Hz, 1 H), 2.14 (ddd, J = 14.6, 1 1.1 , 6.4 Hz, 1 H), 3.89-3.95 (m, 2H), 4.09-4.15 (m, 1 H), 4.26 (dd, J = 13.4, 3.8 Hz, 1 H), 4.55 (d, J = 5.8 Hz, 2H), 4.89-4.98 (m, 1 H), 5.18 (dd, J = 6.2, 3.8 Hz, 1 H), 6.68-6.79 (m, 2H), 7.23-7.31 (m, 1 H), 8.41 (s, 1 H), 10.24 (t, J = 5.8 Hz, 1 H). 13C NMR (CDCI3): δ 16.09, 26.95, 29.30, 36.79, 45.1 1 , 45.28, 53.86, 62.47, 75.93, 103.87 (t, J = 25.4 Hz), 1 1 1 .21 (dd, J = 21 .0, 3.4 Hz), 1 17.32, 130.58 (dd, J = 9.3, 5.8 Hz), 133.40, 143.54, 155.34, 163.16, 163.25, 163.35, 172.88.

Example 19:

To a suspension of 5 (1 .67 g, 4 mmol) in acetonitrile (20 mL) was added DABCO (23 mg, 5 mol%) and TCCA (0.62 g, 2.52 mmol). The mixture was stirred 18 h at 40 °C protected from light and then quenched with a mixture of DMSO (0.48 mL) and water (0.12 mL). The insoluble cyanuric acid was removed by filtration and washed with acetonitrile (5 mL). The filtrate was evaporated under reduced pressure to give viscous oil that was crystallized from a mixture of methanol (6 mL) and water (3 mL), by slowly cooling the solution from 60 °C to room

temperature. The product 6 was filtered, washed with cold methanol (5 mL) and dried to give an off-white powder (1.07 g, 61 %).

1H NMR (CDCI3): δ 1.30 (d, J = 7.0 Hz, 3H), 1 .49 (dd, J = 14.0, 2.2 Hz, 1 H), 2.14 (ddd, J = 14.6, 1 1.1 , 6.4 Hz, 1 H), 3.89-3.95 (m, 2H), 4.09-4.15 (m, 1 H), 4.26 (dd, J = 13.4, 3.8 Hz, 1 H), 4.55 (d, J = 5.8 Hz, 2H), 4.89-4.98 (m, 1 H), 5.18 (dd, J = 6.2, 3.8 Hz, 1 H), 6.68-6.79 (m, 2H), 7.23-7.31 (m, 1 H), 8.41 (s, 1 H), 10.24 (t, J = 5.8 Hz, 1 H). 13C NMR (CDCI3): δ 16.09, 26.95, 29.30, 36.79, 45.1 1 , 45.28, 53.86, 62.47, 75.93, 103.87 (t, J = 25.4 Hz), 1 1 1 .21 (dd, J = 21.0, 3.4 Hz), 1 17.32, 130.58 (dd, J = 9.3, 5.8 Hz), 133.40, 143.54, 155.34, 163.16, 163.25, 163.35, 172.88.

Example 20:

To a suspension of 6 (0.44 g) in anhydrous methanol (1 mL) was added a 25% methanolic solution of sodium methylate (1 .14 mL) and the mixture stirred for 4 h at 40 °C. The reaction was quenched with acetic acid (0.4 mL), diluted with water (8 mL), extracted with 2-methyltetrahydrofuran (12 mL), the extract washed with 1 M NaOH(aq) (8 mL), water (8 mL) and evaporated under reduced pressure. To the oily residue was added methanol (8 mL) and evaporated under reduced pressure to give 27 as a white solid (0.38 g, 88%).

Example 21 :

The suspension of (4R, 12aS)-7-chloro-N-(2,4-difluorobenzyl)-4-methyl-6,8-dioxo-3,4,6,8,12, 12a-hexahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1 -b][1 ,3]oxazine-9-carboxamide (6, 0.44 g) and solid sodium hydroxide (0.20 g) in absolute ethanol (2 mL) was stirred at room temperature for 24 h. The reaction was quenched with 2M H2S04 (1 .18 mL) and left stirring for 2 h at room temperature. The reaction mixture was filtered through fitted funnel rinsing with water (2 x 2 mL). The obtained white precipitate (0.38 g) was suspended in THF-water (1 :1 , 4.5 mL) and stirred at room temperature for ca. 2 h. The reaction mixture was filtered through fitted funnel rinsing with water (2 χ 1 mL) and dried in vacuo at 40°C to afford pure dolutegravir as a white solid (0.33 g, HPLC purity: 99.38%).

1H NMR (DMSO-d6): δ 12.51 (s, 1 H), 10.36 (t, J=5.9 Hz, 1 H), 8.50 (s, 1 H), 7.41-7.36 (m, 1 H), 7.26-7.21 (m, 1 H), 7.07-7.03 (m, 1 H), 5.45 (dd, J=5.4, 4.3 Hz, 1 H), 4.81 -4.76 (m, 1 H), 4.59-4.53 (m, 3H), 4.36 (dd, J=13.8, 5.8 Hz, 1 H), 4.05-4.00 (m, 1 H), 3.91-3.88 (m, 1 H), 2.05-1 .97 (m, 1 H), 1.55-1.52 (m, 1 H), 1 .33 (d, J=7.1 Hz, 3H). 13C NMR (DMSO-d6): δ 170.27, 163.68, 162.29, 161 .78 (dd), 159.82 (dd), 154.61 , 140.64, 130.74 (d), 130.67 (d), 122.37 (d), 1 16.73, 1 15.38, 1 1 1 .33 (d), 103.80 (t), 62.01 , 51 .16, 44.69, 35.74, 29.13, 15.21.

Example 22:

A suspension of dolutegravir (0.31 g) in methanol (4 mL) was cooled to 0 °C.25% Solution of sodium methoxide in methanol was added to the mixture and the resulting suspension was stirred at 0 °C for 2 h, then at room temperature for 23 h. The reaction mixture was then filtered through fitted funnel rinsing with methanol (3 x 10 mL). The white precipitate was dried overnight at room temperature to afford pure dolutegravir sodium as a white solid (0.26 g, HPLC purity: 99.84%).

1H NMR (DMSO-d6): δ 10.70 (t, J=5.8, 1H), 7.89 (s, 1H), 7.37-7.30 (m, 1H), 7.23-7.19 (m, 1H), 7.04-7.01 (m, 1H), 5.17 (m, 1H), 4.81 (t, J=6.4Hz, 1H), 4.51 (d, J=5.5Hz, 2H), 4.32-4.29 (m, 1H), 4.16 (dd, J=14.1, 4.8 Hz, 1H), 3.99-3.94 (m, 1H), 3.82-3.80 (m, 1H), 1.89-1.84 (m, 1H), 1.38 (d, J=12.9 Hz, 1H), 1.24 (d, J=7.0Hz, 3H).13C NMR (DMSO-d6): δ 177.93, 167.12, 166.08, 161.59 (dd), 161.13, 159.63 (dd), 134.26, 130.44 (d), 130.38 (d), 122.90 (d), 114.95, 111.23 (d), 108.78, 103.64 (t), 75.59, 61.95, 53.11, 43.01, 35.32, 29.22, 15.30.

Example 23:

The suspension of 6 (0.44 g) and solid sodium hydroxide (0.20 g) in absolute ethanol (2 mL) was stirred at room temperature for 24 h. The reaction was diluted with absolute ethanol (10 mL) and left stirring for ca. 30 min at room temperature. The reaction mixture was filtered through fitted funnel rinsing with absolute ethanol (3 x 10 mL) and dried in vacuo at room temperature to afford dolutegravir sodium as a pale yellow solid (0.43 g, HPLC purity: 98.80%). 1H NMR (DMSO-d6): δ 10.70 (t, J = 5.8 Hz, 1H), 7.89 (s, 1H), 7.37-7.30 (m, 1H), 7.23-7.19 (m, 1H), 7.04-7.01 (m, 1H), 5.17 (m, 1H), 4.81 (t, J = 6.4 Hz, 1H), 4.51 (d, J = 5.5 Hz, 2H), 4.32-4.29 (m, 1H), 4.16 (dd, J= 14.1, 4.8 Hz, 1H), 3.99-3.94 (m, 1H), 3.82-3.80 (m, 1H), 1.89-1.84 (m, 1H), 1.38 (d, J = 12.9 Hz, 1H), 1.24 (d, J = 7.0 Hz, 3H).13C NMR (DMSO-d6): δ 177.93, 167.12, 166.08, 161.59 (dd), 161.13, 159.63 (dd), 134.26, 130.44 (d), 130.38 (d), 122.90 (d), 114.95, 111.23 (d), 108.78, 103.64 (t), 75.59, 61.95, 53.11, 43.01, 35.32, 29.22, 15.30.

Example 24:

The suspension of (4R,12aS)-N-(2,4-difluorobenzyl)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12, 12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-6][1,3]oxazine-9-carboxamide (27, 0.43 g) and solid sodium hydroxide (0.20 g) in absolute ethanol (2.5 mL) was stirred at room temperature for ca.24 h. The reaction was diluted with mixture of water/ethanol (5 mL, 1:1) and left stirring for ca. 1.5 h at room temperature. The reaction mixture was filtered through fitted funnel rinsing with mixture of water/ethanol (3 x 5 mL, 1:1) and dried in vacuo at room temperature to afford 15 as a pale yellow solid (0.41 g, HPLC purity: 98.87%).

1H NMR (DMSO-de): δ 10.70 (t, J = 5.8 Hz, 1H), 7.89 (s, 1H), 7.37-7.30 (m, 1H), 7.23-7.19 (m, 1H), 7.04-7.01 (m, 1H), 5.17 (m, 1H), 4.81 (t, J = 6.4 Hz, 1H), 4.51 (d, J = 5.5 Hz, 2H), 4.32-4.29 (m, 1H), 4.16 (dd, J = 14.1, 4.8 Hz, 1H), 3.99-3.94 (m, 1H), 3.82-3.80 (m, 1H), 1.89-1.84 (m, 1H), 1.38 (d, J = 12.9 Hz, 1H), 1.24 (d, J = 7.0 Hz, 3H).13C NMR (DMSO-d6): δ 177.93, 167.12, 166.08, 161.59 (dd), 161.13, 159.63 (dd), 134.26, 130.44 (d), 130.38 (d), 122.90 (d), 114.95, 111.23 (d), 108.78, 103.64 (t), 75.59, 61.95, 53.11, 43.01, 35.32, 29.22, 15.30.

Example 25:

The suspension of {4R, 12aS)-7-chloro-4-methyl-6,8-dioxo-3,4, 6,8, 12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-6][1,3]oxazine-9-carboxylic acid (7, 0.31 g) and solid sodium hydroxide (0.20 g) in absolute ethanol (2.5 mL) was stirred at 50 °C for 3 days. The reaction was quenched with 2M H2S04 (1.2 mL) and left stirring for 7 h at room temperature. The reaction mixture was filtered through fitted funnel rinsing with water (3×5 mL) and ethanol (5 mL) dried in vacuo at 40°C to afford 28 as a pale yellow solid (0.17 g).

1H NMR (DMSO-d6): δ 15.37 (s, 1H), 12.76 (s, 1H), 8.66 (s, 1H), 5.51-5.49 (m, 1H), 4.80-4.78 (m, 1H), 4.65 (dd, J=13.8, 3.7 Hz, 1H), 4.43 (dd, J=13.8, 5.9 Hz, 1H), 4.05 (t, J^^.b Hz, 1H), 3.91 (dd, J=11.4, 3.1 Hz, 1H), 2.07-2.00 (m, 1H), 1.56 (d, J=13.8 Hz, 1H), 1.34 (d, J=7.0 Hz, 3H).13C NMR (DMSO-de): δ 172.21, 165.39, 161.73, 153.61, 141.11, 118.66, 112.99, 75.95, 62.03, 51.50, 44.90, 29.08, 15.18.

Example 26:

The suspension of (4R,12aS)-N-(2,4-difluorobenzyl)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8, 12, 12a-hexahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1 ,3]oxazine-9-carboxamide (27, 0.88 g) and solid sodium hydroxide (0.24 g) in absolute ethanol (20 mL) was stirred at 30 °C for 1.5 h. The reaction was quenched with 2M H2S04 (1 .5 mL) and left stirring for 3 hours at room temperature. The reaction mixture was filtered through fritted funnel and rinsed with water (3 x 2 mL) and ethanol (4 mL), and dried in vacuo at 40 °C to afford O-ethyl dolutegravir (29) as a pale yellow solid (0.25 g). The filtrate was extracted with ethyl acetate (3 x 5 mL). The combined organic layers were dried over MgS04, filtered and concentrated, then dried in vacuo at 40 °C to afford more 29 as a pale yellow solid (0.27 g).

1H NMR (CDCI3): δ 10.37 (t, J = 5.8 Hz, 1 H), 8.36 (s, 1 H), 7.37-7.32 (m, 1 H), 6.83-6.77 (m, 2H), 5.19 (dd, J = 5.9, 3.8 Hz, 1 H), 5.04-4.98 (m, 1 H), 4.61 (d, J = 6Hz, 2H), 4.26-4.22 (m, 3H), 4.1 1 (dd, J = 13.4, 5.9 Hz, 1 H), 3.97 (t, J = 2.4 Hz, 1 H), 3.96 (d, J = 2.4 Hz, 1 H), 2.21-2.14 (m, 1 H), 1.51 (dq, J = 14.0, 2.3 Hz, 1 H), 1 .47 (t, J = 7.0 Hz, 3H), 1 .35 (d, J = 7.1 Hz, 3H).

13C NMR (CDCI3): δ 174.78, 164.17, 162.49 (dd), 160.51 (dd), 155.72, 154.08, 142.32, 130.60 (dd), 129.33, 121 .51 (dd), 1 18.67, 1 1 1 .23 (dd), 103.78 (t), 76.15, 69.74, 62.58, 53.42, 44.58, 36.50 (d), 29.44, 16.04, 15.64.

Example 27:

The suspension of (4R, 12aS)-7-(benzyloxy)-4-methyl-3,4, 12,12a-tetrahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1-b][1 ,3]oxazine-6,8-dione (30, 0.68 g, prepared according to prior art) and solid sodium hydroxide (0.40 g) in absolute ethanol (5 mL) was stirred at 50 °C for 14 h. The reaction was quenched with formic acid (0.35 mL), water (2 mL) was added and mixture was left stirring for additional 1 h at room temperature. The reaction mixture was extracted with ethyl acetate (3 x 5 mL) and the combined organic layers concentrated to afford a crude oil. Purification by flash chromatography (eluting with CH2CI2/methanol) afforded 32 as an orange solid (0.26 g, 52 %).

The above procedure if done at room temperature in same time period, affords 31 as orange oil (0.24 g, 43 %).

Compound 32: 1H NMR (DMSO-d6): δ 7.64 (d, J = 7.4 Hz, 1 H), 6.20 (d, J = 7.3 Hz, 1 H), 5.40 (dd, J = 5.1 , 4.2 Hz, 1 H), 4.83-4.78 (m, 1 H), 4.35 (dd, J = 13.6, 3.9 Hz, 1 H), 4.13 (dd, J = 13.6, 5.4 Hz, 1 H), 4.05-4.00 (m, 1 H), 3.90-3.85 (m, 1 H), 2.03-1.95 (m, 1 H), 1.52 (dd, J = 13.9, 1 .9 Hz, 1 H), 1.33 (d, J = 7.1 Hz, 3H). 13C NMR (DMSO-d6): δ 170.96, 163.01 , 153.48, 137.96, 1 16.83, 1 13.52, 76.18, 62.05, 50.39, 44.53, 29.21 , 15.28.

Compound 31 : 1H NMR (DMSO-d6): δ 7.67 (d, J = 7.4 Hz, 1 H), 6.28 (d, J = 7.4 Hz, 1 H), 5.29 (dd, J = 5.4, 3.8 Hz, 1 H), 4.82-4.75 (m, 1 H), 4.32 (dd, J = 13.6, 3.6 Hz, 1 H), 4.10 (dd, J = 13.5, 5.6 Hz, 1 H), 4.03-3.93 (m, 3H), 3.85 (ddd, J = 1 1 .6, 5.0, 2.2 Hz, 1 H), 1.97-1 .89 (m, 1 H), 1 .48 (dd, J = 13.8, 2.1 Hz, 1 H), 1.27 (d, J = 7.1 Hz, 3H), 1.26 (d, J = 7.0 Hz, 3H). 13C NMR (DMSO-d6): δ 174.38, 156.1 1 , 150.82, 139.48, 1 16.39, 1 13.52, 75.92, 67.31 , 61 .80, 51 .36, 44.22, 29.29, 15.76, 15.36.

Exa

The transformation of 6 to dolutegravir with sodium hydroxide in ethanol was monitored for the interconversion of intermediates. The suspension of 6 (0.44 g) and solid sodium hydroxide (0.20 g) in ethanol (3.33 ml.) was stirred at 22 °C. Samples of the reaction mixture were taken after 3, 8 and 24 h for UPLC analysis. After 24 h, the reaction mixture was quenched with 2 M H2S04 (5 ml_), and left stirring at room temperature. The reaction mixture was filtered through fritted funnel, the product rinsed with water (30 ml.) and dried in vacuo at 50 °C overnight to afford dolutegravir as a white solid (0.27 g, 64 %).

The results of reaction monitoring:

Time UPLC analysis (area%)

Entry

(h) compound 6 compound 29 dolutegravir

1 3 h 37.50 20.63 39.99

2 8 h 0.78 15.46 80.32

3 24h 0.31 8.56 88.21

Example 29:

The effect of added water and reaction temperature was evaluated by monitoring 4 reactions in parallel. To the suspensions of 27 (0.86 g) in MeOH were added solid sodium hydroxide (0.40 g) or aqueous solution of NaOH (5 M, 2 ml.) (see Table below). The reactions were stirred in parallel at 50 °C or 22 °C. Samples were taken in timely intervals for UPLC analysis.

The results of reaction monitoring demethylation of 27 in MeOH:

Example 30:

The effect of added water and reaction temperature was evaluated by monitoring 4 reactions in parallel. To the suspensions of 6 (0.88 g) in EtOH were added solid sodium hydroxide (0.40 g) or aqueous solution of NaOH (5 M, 2 mL) (see Table below). The reactions were stirred in parallel at 50 °C or 22 °C. Samples were taken in timely intervals for UPLC analysis.

The results of reaction monitoring of the transformations of 6 in ethanol with NaOH:

dol. = dolutegravir

Exa

The effect of added water and reaction temperature was evaluated by monitoring 4 reactions in parallel. To the suspensions of 27 (0.88 g) in EtOH were added solid sodium hydroxide (0.40 g) or aqueous solution of NaOH (5 M, 2ml_) (see Table below). The reactions were stirred in parallel at 50 °C or 22 °C. Samples were taken in timely intervals for UPLC analysis.

The results of reaction monitoring of the transformations of 27 in ethanol with NaOH:

dol. = dolutegravir

Example 32:

Compound 3 (30 g, 1 10 mmol; assay 99%) was suspended in acetonitrile (450 mL), acetic acid (73 mL) and methanesulfonic acid (25 mL) were added. The reaction mixture was stirred 4 h at 70 °C. The clear red solution was cooled to 25 °C. Triethylamine (77 mL) and (S)-2-aminopropanol (17 mL) were added and the mixture was stirred at reflux temperature for 20 h. The reaction mixture was cooled to 25 °C and the insoluble product filtered, washed with 1 M HCI(aq) (60 mL), water (3 * 60 mL) and dried to give 4c (19.49 g, 67%): mp = 313-315 °C; 1H NMR (DMSO-d6): δ 1.31 (d, J = 6.3 Hz, 3H), 3.65 (dd, J = 8.6, 6.8 Hz, 1 H), 4.13 (dd, J = 1 1.7, 10.3 Hz, 1 H), 4.28 (m, 1 H), 4.39 (dd, J = 8.6, 6.8 Hz, 1 H), 4.92 (dd, J = 12.3, 4.2 Hz, 1 H), 5.45 (dd, J = 10.2, 4.1 Hz, 1 H), 7.16 (s, 1 H), 8.84 (s, 1 H), 15.74 (s, 1 H); 13C NMR (DMSO-d6) 16.5, 51.6, 52.9, 72.4, 81.6, 1 15.8, 1 18.1 , 141.5, 147.6, 153.4, 165.3, 179.0.

PATENT

WO2016016279, NOVEL HYDRATES OF DOLUTEGRAVIR SODIUM

LEK PHARMACEUTICALS D.D. [SI/SI]; Verovskova 57 1526 Ljubljana (SI).
SANDOZ AG [CH/CH]; Lichtstrasse 35 CH-4056 Basel (CH)

HOTTER, Andreas; (AT).
THALER, Andrea; (AT).
LEBAR, Andrija; (SI).
JANKOVIC, Biljana; (SI).
NAVERSNIK, Klemen; (SI).
KLANCAR, Uros; (SI).
ABRAMOVIC, Zrinka; (SI)

The present invention relates to novel hydrates of sodium dolutegravir and their methods of preparation. In addition, the invention relates to a novel crystalline form of sodium dolutegravir, which is a useful intermediate for the preparation of one of the new hydrates. The invention also relates to the use of the new hydrates for the production of pharmaceutical compositions.

Finally, the invention relates to pharmaceutical compositions comprising an effective amount of the novel hydrates, oral dosage forms comprising these pharmaceutical compositions, a process for preparing said oral dosage forms, and the use of such pharmaceutical compositions or dosage forms in the treatment of retroviral infections such as HIV infections -1.

Dolutegravir, chemically designated (4f?, 12aS)-/V-(2,4-difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8, 12, 12a-hexahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1- ?][1 ,3]oxazine-9-carboxamide, is a human immunodeficiency virus type 1 (HIV-1 ) integrase strand transfer inhibitor (INSTI) indicated in combination with other a nti retroviral agents for the treatment of HIV-1 infection. The marketed finished dosage form (TIVICAY™) contains dolutegravir as its sodium salt, chemically denominated sodium (4f?,12aS)-9-((2,4-difluorobenzyl)carbamoyl)-4-methyl-6,8-dioxo-3,4,6,8,12, 12a-hexahydro-2H-pyrido[1 ‘,2’:4,5]pyrazino[2, 1- ?][1 ,3]oxazin-7-olate, which is represented by the following general chemical formula (I):

(I)

WO 2010/068253 A1 discloses a monohydrate and an anhydrous form of dolutegravir sodium as well as a crystalline form of the free compound. Processes for the preparation of said forms are also provided in the application.

WO 2013/038407 A1 discloses amorphous dolutegravir sodium and processes for preparing the same.

Hydrates of pharmaceutical drug substances are of particular interest as they provide new opportunities for preparing novel pharmaceutical compositions with improved quality, activity and/or compliance. This is due to the fact that hydrates have different physicochemical properties compared to their anhydrous counterparts such as melting point, density, habitus, chemical and physical stability, hygroscopicity, dissolution rate, solubility, bioavailability etc., which influence the formulation process and also impact the final drug product.

If an anhydrous form is selected, phase changes during the formulation process induced by hydrate formation must be avoided. This can be particularly difficult if for example wet granulation is used with a substance that is able to form hydrates like dolutegravir sodium.

Hence, a stable hydrate of dolutegravir sodium would allow to easily formulate dolutegravir sodium in a controlled manner and subsequently also facilitate storage and packaging.

However, the so far known dolutegravir sodium monohydrate disclosed in WO 2010/068253 A1 shows excessive water uptake when exposed to moisture and on the other hand already dehydrates below 30% relative humidity.

Therefore, there is a need for hydrates of dolutegravir sodium with improved physicochemical properties, e.g. for hydrates which are stable over a broad humidity range, in particular for hydrates absorbing only low amounts of water at elevated humidity and on the other hand preserving their crystal structure also at dry conditions. In addition, there is a need for pharmaceutical compositions comprising these hydrates, and thus also for hydrates that allow for improved formulation of dolutegravir sodium in pharmaceutical compositions.

SUMMARY OF THE INVENTION

The present invention relates to novel hydrates of dolutegravir sodium and to processes for their preparation. Specifically, the present invention provides crystalline forms of dolutegravir sodium of formula (I) according to respective claims 1 , 5 and 6, with preferred embodiments being set forth in sub-claim 2. The present invention also provides processes for their preparation according to respective claims 3, 7 and 8, with preferred process embodiments being set forth in sub-claim 4. The present invention further provides the uses according to claims 9 and 16, and a pharmaceutical composition according to claim 10, and preferred embodiments thereof according to sub-claims 1 1 and 12. The present invention also provides a process for the preparation of the pharmaceutical composition according to claim

13, and preferred embodiments thereof according to sub-claim 14. The pharmaceutical composition for therapeutic use is set forth in claim 15.

The novel hydrates are physically and chemically stable over a broad humidity range, show only low water uptakes when exposed to moisture and are even stable at dry conditions. Therefore, the novel hydrates are especially suitable for the preparation of pharmaceutical compositions, e.g. in terms of time and costs.

In particular, it has been found that crystal Form HxA exhibits improved properties which allow for improved formulation of Form HxA in pharmaceutical compositions.

In addition, the present invention relates to a novel crystalline form of dolutegravir sodium, which, for the first time, allows the preparation of one of the novel hydrates and is therefore a valuable intermediate.

PATENT

1361/CHE/2013

Dolutegravir (I) is chemically known as (4/?,12aS)-N-[(2,4-difluorophenyl)methyl]-3,4,6,8,12,12a-hexahydro-7-hydroxy-4-methyl-6,8-dioxo-2//-pyrido[r,2′:4,5]pyrazino[2,l-b][l,3]oxazine-9-carboxamide. Dolutegravir is a human immunodeficiency virus type 1 (HIV-1) integrase strand transfer inhibitor (INSTI) indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection. Dolutegravir is being marketed under the trade name Tivicay®. US 8,129,385 disclosed Dolutegravir or its pharmaceutically acceptable salts thereof. US ‘385 also discloses a process for the preparation of Dolutegravir (I). The process involves the condensation of 5-benzyloxy-4-hydroxy-6-hydroxymethyl nicotinic acid (II) with 2,4-difluorobenzylamine (III) to produce 5-benzyloxy-N-(2,4-difluorobenzyl)-4-hydroxy-6-hydroxymethyl nicotinic acid amide (IV), which is further under goes oxidation using manganese dioxide (Mn02) to produce 5-benzyloxy-N-(2,4-difluorobenzyl)-6-formyl-4-hydroxy-nicotinic acid amide (V). This amide compound (V) is reacted with sodium chlorite (NaClCh) to produce 3-benzyloxy-5-(2,4-difluorobenzylcarbamoyl)-4- hydroxy-pyridine-2-carboxylic acid (VI), which is further treated with methanol (MeOH) to produce 3-benzyloxy-5-(2,4-difluorobenzyl)-4-hydroxy-pyridine-2-carboxylic acid methyl ester (VII).

The methyl ester compound (VII) is reacted with 3-bromopropene to produce l-allyl-3-benzyloxy-5-(2,4-difluorobenzyl)-4-oxo-l,4-dihydro-pyridine-2- carboxylic acid methyl ester (VIII), which is further reacted with potassium osmate dihydrate (K2OSO4.2H2O) to produce 3-benzyloxy-5-(2,4-difluorobenzylcarbamoyl)-4-oxo-l-(2-oxo-ethyl)-l,4-dihydropyridine-2-carboxylic acid methyl ester (IX). The compound (IX) is reacted with (R)-3-amino-l-butanol (X) to produce benzyloxy Dolutegravir (XI), which is deprotected by treating with TFA to produce Dolutegravir (I). The process is as shown in scheme-I below:

scheme1

The major disadvantage with the above prior-art process is that it involves large no of steps and tedious work-up procedures to isolate the required product. This results a longer period of time cycle is required to produce Dolutegravir (I), which in turn renders the process more costly and less eco friendly. Further the above processes are low yielding and with less purity. US 8,217,034 discloses variant process for the preparation of Dolutegravir.

This process involves the reaction of methyl l-(2,2-dihydroxyethyl)-4-oxo-3-[(phenylmethyl)oxy]-l,4-dihydro-2-pyridine carboxylate (XII) with (R)-3-amino-l-butanol (X) to produce (4R, 12o5)-4-methyl-7-[(phenylmethyl)oxy]-3,4,12,12a-tetrahydro-2//-pyrido[ 1 \2′,4,5] pyrazino[2,l-b][l,3]oxazine-6,8-dione (XIII), which is further undergoes bromination using NBS to produce (4R,12aS)-9-bromo-4-methyl-7-[(phenylmethyl)oxy]-3,4,12,12a-tetrahydro-2H-pyrido[r,2′:4,5]pyrazino[2,l-b][l,3]oxazine-6,8-dione (XIV). The bromo Compound (XIV) is condensed with 2,4-difluorobenzylamine (III) in the presence of Tetrakis(triphenylphosphine)palladium (Pd(PPh3)4) to produce benzyloxy Dolutegravir (XI), which is hydrogenated in the presence of Pd/C to produce Dolutegravir (I). The process is as shown in Scheme-II below:

scheme2

The major disadvantage with the above prior art process of preparing Dolutegravir is the use of expensive reagent tetrakis(triphenylphosphine)palladium (Pd(PPh3)4> in coupling step. Use of this reagent on industrial scale is not preferred, which makes the process more expensive. WO 2011/119566 discloses another variant process for the preparation of Dolutegravir.

This process involves the reaction of l-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l,4-dihydropyridine-3-carboxylic acid (XV) with acetic acid in presence of methane sulfonic acid to produce 5-methoxy-6-(methoxycarbonyl)-4-oxo-l-(2-oxoethyl)-l,4-dihydropyridine-3-carboxylic acid (XVI), which is further condensed with (R)-3-amino-l-butanol (X) to produce (4R,12aS)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2//-pyrido[ 1 ‘,2’:4,5]pyrazino[2,1 -b] [ 1,3]-oxazine-9-carboxylic acid (XVII). This acid Compound XVII is acylated with 2,4-difluorobenzylamine (III) in the presence of carbonyldiimidazole (CDI) to produce methoxy Dolutegravir (XVIII), which is demethylated in the presence of lithium bromide (LiBr) to produce Dolutegravir (I).

The process is as shown in Scheme-3 below:

scheme3

The major disadvantage of the above prior art process of preparing Dolutegravir is the use of expensive and highly moisture sensitive reagent, 1,1-carbonyldiimidazole (CDI), during acylation. Use of this reagent on industrial scale is not preferred due to anhydrous conditions required in the process. However, there is always a need for alternative preparative routes, which for example, involve fewer steps, use reagents that are less expensive and/or easier to handle, consume smaller amounts of reagents, provide a higher yield of product, have smaller and/or more eco-friendly waste products, and/or provide a product of higher purity. Hence, there is a need to develop cost effective and commercially viable process for the preparation of Dolutegravir of formula (I). The present invention is related to a process for the preparation of pure Dolutegravir of formula (I), wherein optically active acid addition salt of (R)-3-amino-l-butanol (X) is directly condensed with 5-methoxy-6-(methoxycarbonyl)-4-oxo-l-(2-oxoethyl)-l,4-dihydropyridine-3-carboxylic acid (XVI) instead of condensing with free base of (R)-3-amino-1-butanol (X). The present invention is also related to a process for the preparation of pure Dolutegravir of formula (I), wherein, inexpensive and easily handling condensing reagents in the condensation of (4R, 12aS)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2//-pyrido[l’,2′:4,5]pyrazino [2,l-b][l,3]oxazine-9-carboxylic acid (XVII) with 2,4-difluorobenzylamine (III).

In another embodiment, 5-methoxy-6-(methoxycarbonyl)-4-oxo-l-(2-oxoethyl)-l,4- dihydropyridine-3-carboxylic acid (XVI) used in the present invention is prepared by reacting 4-methoxyacetoacetate (XIX) with N,N-dimethyl-l,l- bis(methyloxy)methanamine (DMF-DMA) (XX) to produce methyl-2- (dimethylaminomethylene)-4-methoxy-3-oxo-butanoate(methyl-3-(dimethylamino)-2 [(methyloxy)acetyl]-2-propenoate) (XXI), which is reacted with aminoacetaldehyde dimethyl acetal (XXII) to produce methyl-2-(2,2-dimethoxyethylaminomethylene)-4-methoxy-3-oxo-butanoate(methyl-3-{[2,2-bis(methyloxy)ethyl]amino}-2-[(methyloxy) acetyl]-2-propenoate) (XXIII).

The compound (XXIII) is contacted with dimethyl ethanedioate in presence of alkali metal alkoxide to produce dimethyl-1-(2,2-dimethoxyethyl)-3-methoxy-4-oxo-l ,4-dihydropyridine-2,5-dicarboxylate (XXIV), which is selectively hydrolyzed with a base to produce l-[2,2-bis(methyloxy)ethyl]-5-(methyloxy)-6-[(methyloxy)carbonyl]-4-oxo-l ,4-dihydro-3-pyridinecarboxylic acid (XV). The compound (XV) is treated with a catalytic amount of a strong protic acid in the presence of acetic acid in an organic solvent to produce a reaction mixture containing 5- methoxy-6-(methoxycarbonyl)-4-oxo-l-(2-oxoethyl)-l,4-dihydropyridine-3-carboxylic acid (XVI), The process is as shown in Scheme-IV below:

scheme4

The following examples illustrate the nature of the invention and are provided for illustrative purposes only and should not be construed to limit the scope of the invention.

Example-1:

EXAMPLES: Example-1: Process for the preparation of Dolutegravir

Step-i: Preparation of (/?)-3-amino-l-butanol tartarate salt: D-(+) Tartaric acid (12.7 g, 0.085 mol) was added in to a solution of (i?,5)-3-amino-l-butnaol (7.5 g, 0.084 mol) in methanol (100 ml) at 40 °C. The reaction mixture was stirred for about 1 hour at 35-40 °C and the reaction mass was cooled to 0-5°C and maintained for 30-40 minutes. The obtained solid was filtered and washed with chilled methanol (10 ml) at 0-5 °C. The solid was dried to get (i?)-3-amino-l-butanol tartarate salt (8.0 g, 40%).

Step-ii: Preparation of (4rt,12a£)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[l’,2′;4,5]pyrazino[2,l-b][l,3]oxazine-9-carboxylic acid (XVII): l-[2,2-Bis(methyloxy)ethyl]-5-(methyloxy)-6-[(methyloxy)carbonyl]-4-oxo-l,4-dihydro-3-pyridinecarboxylic acid (XV) (lOOg; 0.3175 moles) was suspended in acetonitrile (800 ml) and heated to 80-82°C. A mixture of acetic acid (95.25 g), methanesulfonic acid (9.14 g; 0.09525 moles) and acetonitrile (200 ml) were added to the slurry at 80-82°C. The reaction mass was continued at 80-82°C to complete the reaction. After completion of the reaction, anhydrous sodium acetate (65 g) and (/?)-3-amino-l-butanol tartrate salt (79.68g; 0.3334 moles) were added at 20-25°C and stirred at 60-65°C to complete the reaction. The reaction mass was concentrated and acidified with IN aqueous hydrochloric acid (750 ml) and extracted with methylene chloride (1500 ml) at ice cold temperature. The organic layer was separated, concentrated, treated with hot methanol (350 ml) for 2 h, filtered, washed with methanol and dried to yield (4R,12aS)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[ 1′ ,2′ :4,5]pyrazino[2,1 -b] [ 1,3]oxazine-9-carboxylic acid (XVII) (72 g; HPLC purity: 99.07%).

Step-iii: Process for the preparation of Dolutegravir (I). Method A: Triethylamine (3.61 g; 0.0357 moles) was added to the suspension of (4R,12aS)-7- methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[ 1′ ,2′ :4,5]pyrazino[2,1 – b][l,3]oxazine-9-carboxylic acid (XVII) (10 g; 0.0325 moles) in methylene chloride (50 ml), and cooled to 10-15°C. Pivaloyl chloride (4.3 g; 0.0357 moles) was added to the reaction mass, and stirred at 10-15°C for 1 h. Thereafter, 2,4-difiuorobenzylamine (5.58 g; 0.0389 moles) was added at 10-15°C and then warmed to 20-25°C to complete the reaction. After completion of the reaction, IN aqueous hydrochloric acid (20 ml) was added, organic layer was separated, washed with 5% w/w aqueous sodium bicarbonate solution (10 ml) followed by 15% w/w aqueous sodium chloride solution (10 ml) and concentrated. To the concentrated mass, acetonitrile (100 ml) and Lithium bromide (5.08 g; 0.0584 moles) were added and heated to 65-70°C for 3 h to complete the reaction. After completion of the reaction, the reaction mass was acidified with 5N aqueous hydrochloric acid (40 ml), concentrated to about 50 ml and DM water was added to crystallize the product at 20-25°C. The slurry was stirred for 2 h, filtered, washed with DM water and dried to yield (4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a,-hexahydro-2H-pyrido[ 1′ ,2′ :4,5]pyrazino[2,1 -b] [ 1,3]oxazine-9-carboxamide (I) (11.5 g, HPLC purity: 99.63%).

Method B: Isobutyl chloroformate (4.65 gm, 0.03404 moles) in methylene chloride (10 ml) was added to the solution of N-methylmorpholine (3.45 gm, 0.03410 moles) and (4R,12aS)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[ 1′ ,2′ :4,5]pyrazino-[2,1 -b][l,3]oxazine-9-carboxy!ic acid (XVII) (10.0 gm, 0.03245 moles) in methylene chloride (60 ml) at -10 to 0°C in about 1 h. 2,4-Difloro benzyl amine (4.88 gm, 0.03409 moles) in methylene chloride (10 ml) was added to the cold reaction mass, and stirred at 20-30°C for completion of reaction. After completion of reaction, the reaction mass was washed with 5%w/w aqueous sodium bicarbonate solution (20 ml), IN hydrochloric acid (20 ml), DM water (20 ml) and concentrated. Acetonitrile (120 ml) and lithium bromide (4.8 gm, 0.05516 moles) were added to the concentrated mass, and stirred at 70-80°C for 3 h to complete the reaction. After completion of reaction, the reaction mass was acidified with 5N aqueous hydrochloric acid (40 ml) and concentrated to about 50 ml. DM Water (100 ml) was added to the concentrated reaction mass and stirred for 2 h at 25-30°C to crystallize the product. The product was filtered, washed with DM Water (50 ml) and dried to yield Dolutegravir (I) (10.7 gm, HPLC purity: 99.60%).

Example-2: Process for the preparation of Dolutegravir (I) (4R, 12aS)-N-(2,4-difluorobenzyl)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a,-hexahydro-2H-pyrido[r,2′:4,5]pyrazino[2,l-b][l,3]oxazine-9-carboxamide (XVIII) (2 g, 0.0046 moles) was suspended in isopropyl alcohol (20 ml) and lithium bromide (0.8 g, 0.00924 moles) was added and stirred at 70-80°C for 15 h to complete the reaction. After completion of reaction the reaction mass was acidified with 5N aqueous hydrochloric acid (5 ml) and concentrated. DM Water (20 ml) was added to the concentrated mass and stirred at 25-30°C to crystallize the product. The product was filtered, washed with DM Water and dried to yield Dolutegravir (I) (1.5 g, HPLC purity: 97.93%).

Dolutegravir

 
 
 

Experimental:

1H NMR (CDCl3) δ  12.45 (s, 1H), 10.38 (br s, 1H), 8.30 (s, 1H), 7.40-7.30 (m, 1H), 6.85-6.75 (m, 2H), 5.26 (d, J = 5.8, 4.1 Hz, 2H), 5.05-4.95 (m, 1H), 4.64 (d, J = 5.9 Hz, 2H), 4.27 (dd, J = 13.4, 4.2 Hz, 1H), 4.12 (dd, J = 13.6, 6.0 Hz, 1H), 4.05 (t, J = 2.3 Hz, 1H), 4.02 (d, J = 2.2 Hz, 1H), 2.30-2.19 (m, 1H), 1.56 (dd, J = 14.0, 2.0 Hz, 1H), 1.42 (d, J = 7.0 Hz, 3H).////////////LINK

Dolutegravir sodium

DOLUTEGRAVIR SODIUM.png

DOLUTEGRAVIR SODIUM; UNII-1Q1V9V5WYQ; Dolutegravir (sodium);  GSK1349572A; GSK 1349572A;  1051375-19-9

Molecular Formula: C20H18F2N3NaO5
Molecular Weight: 441.360596 g/mol


sodium;(4R,12aS)-9-[(2,4-difluorophenyl)methylcarbamoyl]-4-methyl-6,8-dioxo-3,4,12,12a-tetrahydro-2H-pyrido[5,6]pyrazino[2,6-b][1,3]oxazin-7-olate


Sodium(4R,12aS)-9-{[(2,4-Difluorophenyl)methyl]carbamoyl}-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazol-7-olate (1)

Characterization data of 1:
1H NMR (400 MHz, DMSO-d6) δ 10.6–10.7 (t, J = 6.0 Hz, 1H), 7.8 (s, 1H), 7.3 (dd, J = 8.4 and 7.2 Hz, 1H), 7.1–7.2 (m, 1H), 7.0 (t, J = 8.0 Hz, 1H), 5.1 (bs, 1H), 4.7–4.8 (m, 1H), 4.5 (d, J = 5.6 Hz, 2H), 4.2–4.3 (d, J = 11.2 Hz, 1H), 4.1 (m, 1H), 3.9 (m, 1H), 3.7–3.8 (m, 1H), 1.8 (m, 1H), 1.3 (d, J = 13.2 Hz, 1H), 1.2 (d, J = 6.8 Hz, 3H);
13C NMR (400 MHz, DMSO-d6) δ 177.9, 167.0, 166.0, 161.0, 159.9, 160.0, 162.4, 162.5, 158.6, 158.8, 161.1, 161.2, 134.2, 130.4, 130.5, 122–8, 123.0, 114.8, 111.0, 111.3, 108.6, 103.3, 103.8, 75.5, 61.8, 53.1, 42.9, 35.3, 29.1, 15.3;
 IR (KBr, cm–1): 3165, 3072, 2974, 2941, 2873, 1643, 1539, 1504, 1101;
ESI-MS m/z: 418.17.

References

  1. [1] American Medical Association (AMA), STATEMENT ON A NONPROPRIETARY NAME ADOPTED BY THE USAN COUNCIL (Dolutegravir) Accessed 3 December 2011.
  2.  FDA approves new drug to treat HIV infection http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm364744.htm Aug. 12, 2013
  3. “U.S. FDA approves GlaxoSmithKline’s HIV drug Tivicay”Reuters. 12 August 2013. Retrieved 13 February 2013.
  4. “GSK wins priority status for new HIV drug in U.S”Reuters. 16 February 2013. Retrieved 18 February 2013.
  5. “ViiV Healthcare receives approval for Tivicay™ (dolutegravir) in Canada for the treatment of HIV”. Retrieved 11 November 2013.
  6. FDA approves new drug to treat HIV infection http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm364744.htm Aug. 12, 2013
  7.  U.S. FDA approves GlaxoSmithKline’s HIV drug Tivicay http://www.reuters.com/article/2013/08/12/us-glaxosmithkline-hivdrug-idUSBRE97B0WU20130812 Mon Aug 12, 2013 6:40pm EDT
  8. “Dolutegravir Prescribing Information”. Retrieved 3 January 2014.
  9.  Raffi, F; Jaeger, H; Quiros-Roldan, E; Albrecht, H; Belonosova, E; Gatell, JM; Baril, JG; Domingo, P; Brennan, C; Almond, S; Min, S; extended SPRING-2 Study, Group (Nov 2013). “Once-daily dolutegravir versus twice-daily raltegravir in antiretroviral-naive adults with HIV-1 infection (SPRING-2 study): 96 week results from a randomised, double-blind, non-inferiority trial.”. The Lancet infectious diseases13 (11): 927–35. PMID24074642.
  10. http://www.natap.org/2013/ICAAC/ICAAC_24.htm
  11.  Walmsley, Sharon L.; Antela, Antonio; Clumeck, Nathan; Duiculescu, Dan; Eberhard, Andrea; Gutiérrez, Felix; Hocqueloux, Laurent; Maggiolo, Franco; Sandkovsky, Uriel; Granier, Catherine; Pappa, Keith; Wynne, Brian; Min, Sherene; Nichols, Garrett (7 November 2013). “Dolutegravir plus Abacavir–Lamivudine for the Treatment of HIV-1 Infection”. New England Journal of Medicine369 (19): 1807–1818. doi:10.1056/NEJMoa1215541.
  12.  Sax, Paul. “SINGLE Study Underscores Waning of the Efavirenz Era — But Probably Just in the USA – See more at:http://blogs.jwatch.org/hiv-id-observations/index.php/single-study-underscores-waning-of-the-efavirenz-era-but-probably-just-in-the-usa/2013/11/06/#sthash.A39SderN.dpuf”. Retrieved 19 December 2013.
  13.  Eron, JJ; Clotet, B; Durant, J; Katlama, C; Kumar, P; Lazzarin, A; Poizot-Martin, I; Richmond, G; Soriano, V; Ait-Khaled, M; Fujiwara, T; Huang, J; Min, S; Vavro, C; Yeo, J; VIKING Study, Group (2013 Mar 1). “Safety and efficacy of dolutegravir in treatment-experienced subjects with raltegravir-resistant HIV type 1 infection: 24-week results of the VIKING Study.”. The Journal of infectious diseases207 (5): 740–8. PMID23225901.
  14. WO2010011812A1 * Jul 23, 2009 Jan 28, 2010 Smithkline Beecham Corporation Chemical compounds
    WO2010011819A1 * Jul 23, 2009 Jan 28, 2010 Smithkline Beecham Corporation Chemical compounds
        • [Patent Document 1] International publication No.2006/116764 pamphlet
        • [Patent Document 2] International publication No.2010/011812 pamphlet
        • [Patent Document 3] International publication No.2010/011819 pamphlet
        • [Patent Document 4] International publication No.2010/068262 pamphlet
        • [Patent Document 5] International publication No.2010/067176 pamphlet
        • [Patent Document 6] International publication No.2010/068253 pamphlet
        • [Patent Document 7] US Patent 4769380A
        • [Patent Document 8] International applicationPCT/JP2010/055316

    [NON-PATENT DOCUMENTS]

      • [Non-Patent Document 1] Journal of Organic Chemistry, 1991, 56(16), 4963-4967
      • [Non-Patent Document 2] Science of Synthesis, 2005, 15, 285-387
      • [Non-Patent Document 3] Journal of Chemical Society Parkin Transaction. 1, 1997, Issue. 2, 163-169
Dolutegravir
Dolutegravir.svg
Dolutegravir ball-and-stick model.png
Systematic (IUPAC) name
(4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-pyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide
Clinical data
Trade names Tivicay
AHFS/Drugs.com Multum Consumer Information
MedlinePlus a613043
License data
Pregnancy
category
  • US: B (No risk in non-human studies)
Routes of
administration
Oral
Legal status
Legal status
Pharmacokinetic data
Bioavailability n/a[1]
Protein binding ≥98.9%
Metabolism UGT1A1 and CYP3A
Biological half-life ~14 hours
Excretion Feces (53%) and urine (18.9%)
Identifiers
CAS Number 1051375-16-6 
ATC code J05AX12 (WHO)
PubChem CID 54726191
IUPHAR/BPS 7365
ChemSpider 25051637 Yes
UNII DKO1W9H7M1 Yes
ChEMBL CHEMBL1229211 Yes
NIAID ChemDB 538122
Chemical data
Formula C20H19F2N3O5
Molar mass 419.38 g/mol
///////////GSK 1349572, S-349572, GSK 1349572, GSK-1349572, GSK1349572, Tivicay®, GSK1349572, GSK-1349572, S/GSK 1349572, S/GSK1349572, S/GSK1349572 (GSK1349572), S/GSK1349572, UNII:DKO1W9H7M1, 1051375-16-6, DOLUTEGRAVIR, 1051375-19-9,  ドルテグラビルナトリウム , Soltegravir
C[C@@H]1CCO[C@@H]2N1C(=O)c3c(c(=O)c(cn3C2)C(=O)NCc4ccc(cc4F)F)O
CC1CCOC2N1C(=O)C3=C(C(=O)C(=CN3C2)C(=O)NCC4=C(C=C(C=C4)F)F)[O-].[Na+]

Filed under: FDA 2013 Tagged: 1051375-16-6, 1051375-19-9, ドルテグラビルナトリウム, dolutegravir, FDA 2013, GSK 1349572, GSK1349572, S-349572, S/GSK 1349572, S/GSK1349572, S/GSK1349572 (GSK1349572), Soltegravir, Tivicay®, UNII:DKO1W9H7M1

Convert a Drowning Ocean into your Swimming Pool

$
0
0

Convert a drowning ocean into your swimming pool

Anthony Melvin Crasto Ph.D

If you find a situation in life which is similar to a drowning ocean, then rather than struggling for survival, pull few people to shore, you will find the drowning ocean like a swimming pool.

I suffered a paralytic stroke in 2007, called acute transverse mylitis and was bedridden, now wheelchair bound with 90% paralysis.

I faced a hopeless situation and in the process of pulling myself out of crisis helped millions in my field of profession to overcome day to day hurdles.

I worked on  a simple plan, collect information from “free” sources and put them in one place,  a week search then reduced to 5 minutes appreciation. A browsing junior gets the info and becomes happy to collect more on his own, no doubt the initial “leads” makes him confident to understand the subject.

Ego, anger, anxiety, crookedness, hatred, individualism, cruelty, all do not allow you to share, one accumulates wealth but not knowledge and becomes rusty, angry, with all sorts of prejudices. “Starts hiding everywhere” attitude seen.

One who shares, learns, accumulates and enhances his knowledge and skills. perfects them, invites criticism and corrects on the same, then shapes into a bright, honest, knowledgeable and liked individual.

In real life please pull people out of trouble, you will find your own troubles disappearing.

My single blog out of my 15 blogs has taken 13 lakh+ hits till Aug16 and 2 lakh+ viewers in US alone, with audiences in 212 countries

Link is NEW DRUG APPROVALS,  https://newdrugapprovals.wordpress.com/

Till date I have 25 lakh+ views on my blogs, 9.5 million google hits, 2.5 lakh + connections worldwide.

THANKS AND REGARDS,

DR ANTHONY MELVIN CRASTO

Email:  amcrasto@gmail.com

CALL: +919323115463

PS, THE VIEWS EXPRESSED ARE MY PERSONAL AND IN NO-WAY SUGGEST THE VIEWS OF THE PROFESSIONAL BODY OR THE COMPANY THAT I REPRESENT,

REF https://www.linkedin.com/pulse/convert-drowning-ocean-your-swimming-pool-anthony-melvin-crasto-ph-d?trk=pulse_spock-articles

////////


Filed under: Anthony crasto, BLOGS

Olopatadine

$
0
0

Olopatadine.svg

Olopatadine hydrochloride is an antihistamine (as well as anticholinergic and mast cell stabilizer), sold as a prescription eye dropmanufactured by Alcon in one of three strengths: 0.7% solution or Pazeo in the US, 0.2% solution or Pataday (also called Patanol Sin some countries), and 0.1% or Patanol (also called Opatanol in some countries). It is used to treat itching associated with allergicconjunctivitis (eye allergies). A decongestant nasal spray formulation is sold as Patanase, which was approved by the FDA on April 15, 2008.[1] It is also available as an oral tablet in Japan under the tradename Allelock, manufactured by Kyowa Hakko Kogyo.[2]

It should not be used to treat irritation caused by contact lenses. The usual dose for Patanol is 1 drop in each affected eye 2 times per day, with 6 to 8 hours between doses. Both Pazeo and Pataday are dosed 1 drop in each eye daily.

There is potential for Olopatadine as a treatment modality for steroid rebound (red skin syndrome).[3]

Olopatadine was developed by Kyowa Hakko Kogyo.[4]

Side Effects

Some known side effects include headache (7% of occurrence), eye burning and/or stinging (5%), blurred vision, dry eyes, foreign body sensation, hyperemia, keratitis, eyelid edema, pruritus, asthenia, sore throat (pharyngitis), rhinitis, sinusitis, and taste perversion.

Synthesis

Olopatadine synthesis:[5]

References

  1.  Drugs.com, Alcon’s Patanase Nasal Spray Approved by FDA for Treatment of Nasal Allergy Symptoms
  2.  Kyowa Hakko Kogyo Co., Ltd. (2007). “ALLELOCK Tablets 2.5 & ALLELOCK Tablets 5 (English)” (PDF). Retrieved2008-08-10.
  3. Jump up^ Tamura T; Matsubara M; Hasegawa K; Ohmori K; Karasawa A. (2005). “Olopatadine hydrochloride suppresses the rebound phenomenon after discontinuation of treatment with a topical steroid in mice with chronic contact hypersensitivity.”.
  4. Jump up^ Kyowa Hakko Kogyo Co., Ltd. (2002). “Company History”.Company Information. Kyowa Hakko Kogyo Co., Ltd. Retrieved16 September 2010.
  5. Jump up^ Ueno, K.; Kubo, S.; Tagawa, H.; Yoshioka, T.; Tsukada, W.; Tsubokawa, M.; Kojima, H.; Kasahara, A. (1976). “6,11-Dihydro-11-oxodibenz[b,e]oxepinacetic acids with potent antiinflammatory activity”. Journal of Medicinal Chemistry. 19 (7): 941.doi:10.1021/jm00229a017.

External links

 

 

Olopatadine
Olopatadine.svg
Systematic (IUPAC) name
{(11Z)-11-[3-(dimethylamino)propylidene]-6,11-
dihydrodibenzo[b,e]oxepin-2-yl}acetic acid
Clinical data
Trade names Patanol and others
AHFS/Drugs.com Monograph
MedlinePlus a602025
Pregnancy
category
  • C
Routes of
administration
Ophthalmic, intranasal, oral
Pharmacokinetic data
Biological half-life 3 hours
Identifiers
CAS Number 113806-05-6 Yes
ATC code S01GX09 (WHO)R01AC08 (WHO)
PubChem CID 5281071
DrugBank DB00768 Yes
ChemSpider 4444528 Yes
UNII D27V6190PM Yes
KEGG D08293 Yes
ChEMBL CHEMBL1189432 
Chemical data
Formula C21H23NO3
Molar mass 337.412 g/mol

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


Filed under: Uncategorized Tagged: Olopatadine

DAROLUTAMIDE

$
0
0

STR1

ODM-201.svg

ChemSpider 2D Image | ODM-201 | C19H19ClN6O2

Darolutamide

N-((S)-1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)-propan-2-yl)-5-(1-hydroxyethyl)-1H-pyrazole-3-carboxamide

N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)-propan-2-yl)-5-(l-hydroxyethyl)-lH-pyrazole-3-carboxamide

  • MF C19H19ClN6O2
  • MW 398.846

BAY 1841788; ODM-201

1H-Pyrazole-3-carboxamide, N-[(1S)-2-[3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl]-1-methylethyl]-5-(1-hydroxyethyl)-
BAY-1841788
N-{(2S)-1-[3-(3-Chlor-4-cyanphenyl)-1H-pyrazol-1-yl]-2-propanyl}-5-(1-hydroxyethyl)-1H-pyrazol-3-carboxamid
N-{(2S)-1-[3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl]-2-propanyl}-5-(1-hydroxyethyl)-1H-pyrazole-3-carboxamide
N-{(2S)-1-[3-(3-Chloro-4-cyanophényl)-1H-pyrazol-1-yl]-2-propanyl}-5-(1-hydroxyéthyl)-1H-pyrazole-3-carboxamide
ODM-201
1297538-32-9  CAS
UNII:X05U0N2RCO
  • Originator Orion
  • Developer Bayer HealthCare; Orion
  • Class Antineoplastics
  • Mechanism of Action Androgen receptor antagonists
  • Phase III Prostate cancer
  • Most Recent Events

    • 03 Jun 2016 Bayer and Orion plan the phase III ARASENS trial for Prostate cancer
    • 03 Jun 2016 Bayer and Orion expand the licensing agreement to include joint development of ODM 201 for Metastatic hormone-sensitive prostate cancer (mHSPC)
    • 06 May 2016 Long-term combined adverse events data from the the ARADES (phase I/II) and the ARAFOR (phase I) trials in Prostate cancer presented at the 111th Annual Meeting of the American Urological Association (AUA -2016)

Darolutamide (INN) (developmental code names ODM-201, BAY-1841788) is a non-steroidal antiandrogen, specifically, a full and high-affinity antagonist of the androgen receptor (AR), that is under development by Orion and Bayer HealthCare[1] for the treatment of advanced, castration-resistant prostate cancer (CRPC).[2][3]

Orion and licensee Bayer are co-developing darolutamide, an androgen receptor antagonist, for treating castration-resistant prostate cancer and metastatic hormone-sensitive prostate cancer. In August 2016, darolutamide was reported to be in phase 3 clinical development. The drug appears to be first disclosed in WO2011051540, claiming novel heterocyclic derivatives as tissue-selective androgen receptor modulators, useful for the treatment of prostate cancer.

Mode of action

Relative to enzalutamide (MDV3100 or Xtandi) and apalutamide (ARN-509), two other recent non-steroidal antiandrogens, darolutamide shows some advantages.[3] Darolutamide appears to negligibly cross the blood-brain-barrier.[3] This is beneficial due to the reduced risk of seizures and other central side effects from off-target GABAA receptor inhibition that tends to occur in non-steroidal antiandrogens that are structurally similar to enzalutamide.[3] Moreover, in accordance with its lack of central penetration, darolutamide does not seem to increase testosterone levels in mice or humans, unlike other non-steroidal antiandrogens.[3] Another advantage is that darolutamide has been found to block the activity of all tested/well-known mutant ARs in prostate cancer, including the recently-identified clinically-relevant F876L mutation that produces resistance to enzalutamide and apalutamide.[3] Finally, darolutamide shows higher affinity and inhibitory efficacy at the AR (Ki = 11 nM relative to 86 nM for enzalutamide and 93 nM for apalutamide; IC50 = 26 nM relative to 219 nM for enzalutamide and 200 nM for apalutamide) and greater potency/efficaciousness in non-clinical models of prostate cancer.[3]

ORM-15341 is the main active metabolite of darolutamide.[3] It, similarly, is a full antagonist of the AR, with an affinity (Ki) of 8 nM and an IC50 of 38 nM.[3]

Clinical trials

Darolutamide has been studied in phase I and phase II clinical trials and has thus far been found to be effective and well-tolerated,[4] with the most commonly reported side effects including fatigue, nausea, and diarrhea.[5][6] No seizures have been observed.[6][7] As of July 2015, darolutamide is in phase III trials for CRPC.[3]

Representative binding affinities of ODM-201, ORM-15341, enzalutamide, and ARN-509 measured in competition with [3H]mibolerone using wtAR isolated from rat ventral prostates (C). All data points are means of quadruplicates ±SEM. Ki values are presented in parentheses. D. Antagonism to wtAR was determined using AR-HEK293 cells treated with ODM-201, ORM-15341, enzalutamide, or ARN-509 together with 0.45 nM testosterone in steroid-depleted medium for 24 hours before luciferase activity measurements. All data points are means of triplicates ±SEM. IC50 values are presented in parentheses.

WHIPPANY, N.J., Sept. 16, 2014 /PRNewswire/ — Bayer HealthCare and Orion Corporation, a pharmaceutical company based in Espoo, Finland, have begun to enroll patients in a Phase III trial with ODM-201, an investigational oral androgen receptor inhibitor in clinical development. The study, called ARAMIS, evaluates ODM-201 in men with castration-resistant prostate cancer who have rising Prostate Specific Antigen (PSA) levels and no detectable metastases. The trial is designed to determine the effects of the treatment on metastasis-free survival (MFS).

“The field of treatment options for prostate cancer patients is evolving rapidly.  However, once prostate cancer becomes resistant to conventional anti-hormonal therapy, many patients will eventually develop metastatic disease,” said Dr. Joerg Moeller, Member of the Bayer HealthCare Executive Committee and Head of Global Development. “The initiation of a Phase III clinical trial for ODM-201 marks the starting point for a potential new treatment option for patients whose cancer has not yet spread.  This is an important milestone for Bayer in our ongoing effort to meet the unmet needs of men affected by prostate cancer.”

Earlier this year, Bayer and Orion entered into a global agreement under which the companies will jointly develop ODM-201, with Bayer contributing a major share of the costs of future development. Bayer will commercialize ODM-201 globally, and Orion has the option to co-promote ODM-201 in Europe. Orion will be responsible for the manufacturing of the product.

About the ARAMIS Study
The ARAMIS trial is a randomized, Phase III, multicenter, double-blind, placebo-controlled trial evaluating the safety and efficacy of oral ODM-201 in patients with non-metastatic CRPC who are at high risk for developing metastatic disease. About 1,500 patients are planned to be randomized in a 2:1 ratio to receive 600 mg of ODM-201 twice a day or matching placebo. Randomisation will be stratified by PSA doubling time (PSADT less than or equal to 6 months vs. > 6 months) and use of osteoclast-targeted therapy (yes vs. no).

The primary endpoint of this study is metastasis-free survival (MFS), defined as time between randomization and evidence of metastasis or death from any cause. The secondary objectives of this study are overall survival (OS), time to first symptomatic skeletal event (SSE), time to initiation of first cytotoxic chemotherapy, time to pain progression, and characterization of the safety and tolerability of ODM-201.

About ODM-201
ODM-201 is an investigational androgen receptor (AR) inhibitor that is thought to block the growth of prostate cancer cells. ODM-201 binds to the AR and inhibits receptor function by blocking its cellular function.

About Oncology at Bayer
Bayer is committed to science for a better life by advancing a portfolio of innovative treatments. The oncology franchise at Bayer now includes three oncology products and several other compounds in various stages of clinical development. Together, these products reflect the company’s approach to research, which prioritizes targets and pathways with the potential to impact the way that cancer is treated.

About Bayer HealthCare Pharmaceuticals Inc.
Bayer HealthCare Pharmaceuticals Inc. is the U.S.-based pharmaceuticals business of Bayer HealthCare LLC, a subsidiary of Bayer AG. Bayer HealthCare is one of the world’s leading, innovative companies in the healthcare and medical products industry, and combines the activities of the Animal Health, Consumer Care, Medical Care, and Pharmaceuticals divisions. As a specialty pharmaceutical company, Bayer HealthCare provides products for General Medicine, Hematology, Neurology, Oncology and Women’s Healthcare. The company’s aim is to discover and manufacture products that will improve human health worldwide by diagnosing, preventing and treating diseases.

Bayer® and the Bayer Cross® are registered trademarks of Bayer.

SYNTHESIS

STR1

str1

PATENT

US 2015203479

http://www.google.com/patents/WO2011051540A1?cl=en

PATENT

WO 2012143599

http://www.google.com/patents/US20140094474?cl=de

PATENTS

WO2011051540

https://www.google.com/patents/WO2011051540A1?cl=en

PATENT

IN 2011KO00570

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016120530&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

WO-2016120530

Compound of (I) (5 g) was dissolved in an acetonitrile and distilled water. The reaction mixture was heated at 75 °C and then slowly cooled down at RT and stirred at RT for 3 days. The solid obtained was filtered, washed twice with the acetonitrile: water and dried under vacuum at 40 °C and 60 °C to yield crystalline form of (I) (4.42 g) with 88% of yield (example 1, page 10).

Compound (I) can be synthetized using the procedures described in WO

201 1/051540.

Pure diastereomers (la) and (lb) can be suitably synthetized, for example, using ketoreductase enzymes (KREDs) for both S- and R-selective reduction of compound 1 to compound 2 as shown in Scheme 1, wherein R is H or Ci_6 alkyl.

Scheme 1.

For example, Codexis KRED-130 and KRED -NADH-110 enzymes are useful for obtaining excellent stereoselectivity, even stereospecificity. In Scheme 1 the starting material 1 is preferably an ester (R= Ci_6 alkyl), for example ethyl ester (R=ethyl), such as to facilitate extraction of the product into the organic phase as the compound where R=H has a tendency to remain in the water phase. Intermediate 2 can be protected, preferably with silyl derivatives such as tert-butyldiphenylsilyl, in order to avoid esterification in amidation step. In the case of R=Ci_6 alkyl, ester hydrolysis is typically performed before amidation step, preferably in the presence of LiOH, NaOH or KOH. Amidation from compound 3 to compound 5_is suitably carried out using EDCI HBTU, DIPEA system but using other typical amidation methods is also possible. Deprotection of 5 give pure diastereomers (la) and (lb).

Pyrazole ring without NH substitution is known tautomerizable functionality and is described here only as single tautomer but every intermediate and end product here can exist in both tautomeric forms at the same time.

The stereochemistry of the compounds can be confirmed by using optically pure starting materials with known absolute configuration as demonstrated in Scheme 2, wherein R=H or Ci_6 alkyl, preferably alkyl, for example ethyl. The end products of Scheme 2 are typically obtained as a mixture of tautomers at +300K 1H-NMR analyses in DMSO.

Scheme 2. Synthesis pathway to stereoisomers by using starting materials with known absolute configuration

The crystalline forms I, Γ and Γ ‘ of compounds (I), (la) and (lb), respectively, can be prepared, for example, by dissolving the compound in question in an

acetonitrile: water mixture having volume ratio from about 85: 15 to about 99: 1, such as from about 90: 10 to about 98:2, for example about 95:5, under heating and slowly cooling the solution until the crystalline form precipitates from the solution. The concentration of the compound in the acetonitrile: water solvent mixture is suitably about 1 kg of the compound in 5-25 liters of acetonitrile: water solvent mixture, for example 1 kg of the compound in 10-20 liters of acetonitrile: water solvent mixture. The compound is suitably dissolved in the acetonitrile: water solvent mixture by heating the solution, for example near to the reflux temperature, for example to about 60-80 °C, for example to about 75 °C, under stirring and filtering if necessary. The solution is suitably then cooled to about 0-50 °C, for example to about 5-35 °C, for example to about RT, over about 5 to about 24 hours, for example over about 6 to 12 hours, and stirred at this temperature for about 3 to 72 hours, for example for about 5 to 12 hours. The obtained crystalline product can then be filtered, washed, and dried. The drying is suitably carried out in vacuum at about 40 to 60 °C, for example at 55 °C, for about 1 to 24 hours, such as for about 2 to 12 hours, for example 2 to 6 hours.

The crystalline forms I, Γ and I” of compounds (I), (la) and (lb), respectively, are useful as medicaments and can be formulated into pharmaceutical dosage forms, such as tablets and capsules for oral administration, by mixing with pharmaceutical excipients known in the art.

The disclosure is further illustrated by the following examples.

Example 1. Crystallization of N-((S)- 1 -(3 -(3 -chloro-4-cyanophenyl)- 1 H-pyrazol- 1 -yl)-propan-2-yl)-5 -( 1 -hydroxyethyl)- 1 H-pyrazole-3 -carboxamide (I)

N-((iS)- 1 -(3 -(3 -chloro-4-cyanophenyl)- 1 H-pyrazol- 1 -yl)-propan-2-yl)-5 -( 1 -hydroxyethyl)-! H-pyrazole-3 -carboxamide (I) (5 g), 71.25 ml of acetonitrile, and 3.75 ml of distilled water were charged to a flask, and the mixture was heated up to 75 °C. The mixture was slowly cooled down to RT and stirred at RT for 3 days. The solid obtained was filtered and washed twice with acetonitrile: water (9.5 ml:0.5 ml). The product was dried under vacuum at 40 °C and finally at 60°C to obtain 4.42 g of crystalline title compound (yield of 88 %) which was used in X-ray diffraction study.

Example 3. Synthesis of N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)-propan-2-yl)-5-((S)- 1 -hy droxy ethyl)- lH-pyrazole-3-carboxamide (la)

a) Ethyl-5 -((S) 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxylate

HO

MgS04 x7H20 (341 mg), NADP monosodium salt (596 mg), D(+)-glucose (9.26 g) and optimized enzyme CDX-901 lyophilized powder (142 mg) were added to 0.2 mM of KH2P04 buffer (pH 7.0, 709 ml) to prepare solution I. To this solution I was added solution II which contained ethyl-5 -acetyl- 1 H-pyrazole-3 -carboxylate (8.509 g; 46.70 mmol), EtOH (28 ml) and K ED-130 (NADPH ketoreductase, 474 mg). The mixture was agitated at 30-32°C for 5.5 h (monitoring by HPLC) and allowed to cool to RT. The mixture was evaporated to smaller volume and the residue was agitated with diatomaceous earth and filtered. The mother liquid was extracted with 3×210 ml of EtOAc and dried. The solution was filtered through silica (83 g) and evaporated to dryness to give 7.40 g of the title compound. The optical purity was 100 % ee.

b) Ethyl 5-((S)-l -((tert-butyldiphenylsilyl)oxy)ethyl)- 1 H-pyrazole-3 -carboxylate

Diphenyl-tert-butyl chlorosilane (7.48 g, 27.21 mmol) was added in 26 ml of DMF to a mixture of compound of Example 3(a) (5.00 g, 27.15 mmol) and imidazole (2.81 g, 41.27 mmol) in DMF (50 ml) at RT. The mixture was stirred at RT for 24 h.

Saturated aqueous NaHC03 (56 ml) and water (56 ml) were added and the mixture was stirred at RT for 20 min. The mixture was extracted with 2×100 ml of EtOAc. Combined organic phases were washed with water (1×100 ml, 1×50 ml), dried (Na2S04), filtered and concentrated to give 10.92 g of crude title compound.

c) 5-((S)-l -((tert-Butyldiphenylsilyl)oxy)ethyl)- 1 H-pyrazole-3 -carboxylic acid

2 M NaOH (aq) (38.8 ml; 77.5 mmol) was added to a solution of the compound of Example 3(b) (10.9 g, 25.8 mmol) in 66 ml of THF. The mixture was heated up to reflux temperature. Heating was continued for 2.5 h and THF was removed in vacuum. Water (40 ml) and EtOAc (110 ml) were added. Clear solution was obtained after addition of more water (10 ml). Layers were separated and aqueous phase was extracted with 100 ml of EtOAc. Combined organic phases were dried (Na2S04), filtered and concentrated to give 9.8 g of the title compound.

d) 5-((S)- 1 -((tert-Butyldiphenylsilyl)oxy)ethyl)-N-((S)- 1 -(3-(3-chloro-4-cyano-phenyl)- 1 H-pyrazol- 1 -yl)propan-2-yl)- 1 H-pyrazole-3 -carboxamide

Under nitrogen atmosphere HBTU (0.84 g; 2.22 mmol), EDCIxHCl (3.26 g; 17.02 mmol) and (S)-4-(l-(2-aminopropyl)-lH-pyrazol-3-yl)-2-chlorobenzonitrile (3.86 g; 14.80 mmol) were added to a mixture of crude compound of Example 3(c) (8.68g; purity 77.4 area-%) and DIPEA (2.20 g; 17.02 mmol) in DCM (50 ml). The mixture was stirred at RT for 46 h (6 ml of DCM was added after 20 h). The mixture was washed with 3×20 ml of water, dried (Na2S04), filtered and concentrated to give 13.7 g of crude title compound.

e) N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((S)- 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxamide (la)

TBAF hydrate (Bu4NF x 3H20; 2.34 g; 7.40 mmol) in 10 ml of THF was added to the solution of the compound of Example 3(d) (9.43 g; 14.79 mmol) in THF (94 ml) at 0 °C under nitrogen atmosphere. Stirring was continued at RT for 21.5 h and the mixture was concentrated. DCM (94 ml) was added to the residue and the solution was washed with 3×50 ml of water, dried (Na2S04), filtered and concentrated. Crude product was purified by flash chromatography (EtOAc/n-heptane) to give 2.1 g of the title compound. 1H-NMR (400MHz; d6-DMSO; 300K): Major tautomer (-85 %): δ 1.11 (d, 3H), 1.39 (d, 3H), 4.24-4.40 (m, 2H), 4.40-4.50 (m, 1H), 6.41(s, 1H), 6.93 (d, 1H), 7.77-7.82 (m, 1H), 7.88-8.01 (m, 2H), 8.08 (s, 1H), 8.19 (d, 1H), 13.02 (broad s, 1H). Minor tautomer (-15 %) δ 1.07-1.19 (m, 3H), 1.32-1.41 (m, 3H), 4.24-4.40 (m, 2H), 4.40-4.50 (m, 1H), 6.80 (broad s, 1H), 6.91-6-94 (m, 1H), 7.77-7.82 (m, 1H), 7.88-8.01 (m, 2H), 8.05-8.09 (m, 1H), 8.31 (d, 1H), 13.10 (broad s, 1H).

Example 4. Crystallization of N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((S)- 1 -hy droxy ethyl)- lH-pyrazole-3-carboxamide (la)

N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((S)- 1 -hydroxyethyl)-lH-pyrazole-3-carboxamide (la) (5.00 g; 12.54 mmol) was mixed with 47.5 ml of ACN and 2.5 ml of water. The mixture was heated until compound (la) was fully dissolved. The solution was allowed to cool slowly to RT to form a precipitate. The mixture was then further cooled to 0 °C and kept in this temperature for 30 min. The mixture was filtered and the precipitate was dried under vacuum to obtain 4.50 g of crystalline title compound which was used in the X-ray diffraction study.

Example 6. Synthesis of N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)-propan-2-yl)-5-((R)- 1 -hy droxy ethyl)- lH-pyrazole-3-carboxamide (lb)

a) Ethyl-5 -((R)- 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxylate

Potassium dihydrogen phosphate buffer (Solution I) was prepared by dissolving potassium dihydrogen phosphate (11.350 g, 54.89 mmol) to water (333 ml) and adjusting pH of the solution to 7.0 by addition of 5 M solution of NaOH. MgS04 x 7 H20 (1.650 g), NAD monosodium salt (0.500 g), D(+)-glucose (10.880 g) and optimised enzyme CDX-901 lyophilised powder (0.200 g) were added to Solution I. To this solution (Solution II) were added KRED-NADH- 110 (0.467 g), ethyl-5-acetyl-1 H-pyrazole-3 -carboxylate (10.00 g; 54.89 mmol) and 2-methyltetrahydro-furan (16 ml). The mixture was agitated at 30° C for 11 h and allowed to cool to RT overnight. The pH of the mixture was kept at 7 by addition of 5 M solution of NaOH. The mixture was evaporated to a smaller volume. The evaporation residue was agitated for 10 min with diatomaceous earth (40 g) and activated charcoal (0.54 g), and filtered. Material on the filter was washed with water (40 ml) and the washings were combined with the filtrate. Layers were separated and aqueous phase was extracted with EtOAc (450 ml and 2×270 ml). Combined organic phases were dried over Na2S04, filtered and evaporated to dryness to give 9.85 g of the title compound (100 % ee).

b) Ethyl-5 -((R)- 1 -((tert-butyldiphenylsilyl)oxy)ethyl)- 1 H-pyrazole-3 -carboxylate

Imidazole (5.32 g; 78.08 mmol) was added to a DCM (67 ml) solution of the compound of Example 6(a) (9.85 g; 53.48). The mixture was stirred until all reagent was dissolved and tert-butyldiphenyl chlorosilane (13.21 ml; 50.80 mmol) was added to the mixture. The mixture was stirred for 1.5 h, 70 ml of water was added and stirring was continued for 15 min. Layers were separated and organic phase was washed with 2×70 ml of water and dried over Na2S04, filtered and concentrated to give 22.07 g of crude title compound.

c) 5 -((R)- 1 -((tert-Butyldiphenylsilyl)oxy)ethyl)- 1 H-pyrazole-3 -carboxylic acid

Compound of Example 6(b) (11.3 g; 26.74 mmol; theoretical yield from the previous step) was dissolved in 34 ml of THF and 50 ml of 2 M NaOH (aq.) was added. The mixture was heated under reflux temperature for 70 min. The mixture was extracted with 2×55 ml of EtOAc and combined organic phases were washed with brine, dried over Na2S04, filtered and concentrated. Evaporation residue was triturated in 250 ml of n-heptane, filtered and dried to give 17.58 g of crude title compound.

d) 5-((R)- 1 -((tert-Butyldiphenylsilyl)oxy)ethyl)-N-((S)- 1 -(3-(3-chloro-4-cyano-phenyl)- 1 H-pyrazol- 1 -yl)propan-2-yl)- 1 H-pyrazole-3 -carboxamide

A mixture of the compound of Example 6(c) (11.14 g; 26.75 mmol; theoretical yield from the previous step), 91 ml of DCM, HBTU (1.52 g; 4.01 mmol), EDCIxHCl

(5.90 g; 30.76 mmol), (S)-4-(l-(2-aminopropyl)-lH-pyrazol-3-yl)-2-chlorobenzo-nitrile (6.97 g; 26.75 mmol) and DIPEA (3.98 g; 30.76 mmol) was stirred at RT for 3 h and at 30° C for 22 h. The mixture was washed with 2×90 ml of 0.5 M HC1 and 4×90 ml of water, dried over Na2S04, filtered and concentrated. Crude product was purified by flash column chromatography (n-heptane-EtOAc) to give 16.97 g of title compound.

e) N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((R)- 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxamide (lb)

A mixture of the compound of Example 6(d) (6.09 g; 9.56 mmol), 61 ml of THF and TBAF was stirred at 40 °C for 6.5 h. The mixture was concentrated and 61 ml of EtOAc was added to the evaporation residue. Solution was washed with 2×50 ml of 0.5 M HC1 and 4×50 ml of water, dried over Na2S04, filtered and concentrated. Crude product was purified by flash column chromatography (n-heptane-EtOAc) to give 1.71 g of the title compound. 1H-NMR (400MHz; d6-DMSO; 300K): Major tautomer (~85%): 5 1.10 (d, 3H), 1.38 (d, 3H), 4.14-4.57 (m, 2H), 5.42 (d, 1H),

6.39(s, 1H), 6.86-6.98 (m, 1H), 7.74-7.84 (m, 1H), 7.86-8.02 (m, 2H), 8.08 (s, 1H), 8.21 (d, 1H), 13.04 (broad s, 1H). Minor tautomer (-15%) δ 0.95-1.24 (m, 3H), 1.25-1.50 (m, 3H), 4.14-4.57 (m, 2H), 4.60-4.90 (m, 1H), 5.08 (d, 1H), 6.78 (broad s, 1H), 6.86-6.98 (m, 1H), 7.77-7.84 (m, 1H), 7.86-8.02 (m, 2H), 8.02-8.12 (m, 1H), 8.32 (d, 1H), 13.1 1 (broad s, 1H).

Example 7. Crystallization of N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((R)- 1 -hy droxy ethyl)- 1 H-pyrazole-3 -carboxamide (lb)

N-((S)- 1 -(3-(3-chloro-4-cyanophenyl)- lH-pyrazol- 1 -yl)propan-2-yl)-5-((R)- 1 -hydroxyethyl)-l H-pyrazole-3 -carboxamide (lb) (3.7 g; 9.28 mmol) was mixed with 70 ml of ACN and 3.5 ml of water. The mixture was heated to reflux temperature until compound (lb) was fully dissolved. The solution was allowed to cool slowly. The mixture was filtered at 50 °C to obtain 6.3 mg of the precipitate. Mother liquid was cooled to 41 °C and filtered again to obtain 20.7 mg of the precipitate. Obtained mother liquid was then cooled to 36 °C and filtered to obtain 173 mg of the precipitate. The final mother liquid was cooled to RT, stirred overnight, cooled to 0 °C, filtered, washed with cold ACN: water (1 : 1) and dried to obtain 2.71 g of the precipitate. The precipitates were checked for optical purity and the last precipitate of crystalline title compound (optical purity 100 %) was used in the X-ray diffraction study.

Example 9. Synthesis of Ethyl-5 -((S) 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxylate

HO

Zinc trifluoromethanesulfonate (0.259 g; 0.713 mmol) and (S)-(-)-3-butyn-2-ol (0.25 g; 3.57 mmol) were added to 0.75 ml (5.35 mmol) of Et3N under nitrogen

atmosphere. Ethyldiazoacetate (0.45 ml; 4.28 mmol) was added slowly and the

mixture was heated at 100 °C for 2 h. The mixture was cooled to RT and 5 ml of water was added. The mixture was washed with 15 ml of DCM, 5 ml of water was added and phases were separated. Water phase was washed twice with DCM, all organic layers were combined, dried with phase separator filtration and evaporated to dryness to give 0.523 g of crude material. The product was purified by normal phase column chromatography (0-5 % MeOH:DCM) to give 0.165 mg of the title compound. 1H-NMR (400MHz; d6-DMSO; temp +300 K): Tautomer 1 (major 77%): δ 1.28 (t, 3H), 1.39 (d, 3H), 4.20-4.28 (m, 2H), (d, 1H), 4.75-4.85 (m, 1H) 5.43 (broad d, 1H), 6.54 (broad s, 1H), 13.28 (broad s, 1H). Tautomer 2 (minor 23%): δ 1.28 (t, 3H), 1.39 (d, 3H), 4.20-4.28 (m, 2H), 4.66-4.85 (m, 1H), 5.04-5.15 (broad s, 1H), 6.71 (broad s, 1H), 13.60 (broad s, 1H).

Exam le 10. Ethyl-5 -((R)- 1 -hydroxy ethyl)- 1 H-pyrazole-3 -carboxylate

Zinc trifluoromethanesulfonate (1.037 g; 2.85 mmol) and (R)-(+)-3-butyn-2-ol (1.00 g; 14.27 mmol) were added to 2.98 ml (21.40 mmol) of Et3N under nitrogen atmosphere. Ethyldiazoacetate (1.80 ml; 21.40 mmol) was added slowly and then refluxed for 3 h. The mixture was cooled to RT and 45 ml of water was added. The mixture was extracted with 3×50 ml of DCM, organic layers were combined, dried with phase separator filtration and evaporated to dryness to give 2.503 g of crude material which was purified by normal phase column chromatography (0-10 % MeOH:DCM) to give 0.67 lmg of the title compound. 1H-NMR (400MHz; d6-DMSO; temp +300 K): Tautomer 1 (major 78%): δ 1.28 (t, 3H), 1.39 (d, 3H), 4.18-4.35 (m, 2H), (d, 1H), 4.75-4.85 (m, 1H) 5.42 (broad d, 1H), 6.54 (s, 1H), 13.29 (broad s, 1H). Tautomer 2 (minor 22%): δ 1.28 (t, 3H), 1.39 (d, 3H), 4.18-4.35 (m, 2H), 4.66-4.85 (m, 1H), 5.09 (broad s, 1H), 6.71 (broad s, 1H), 13.61 (broad s, 1H).

References

  1.  “Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies.”. Sci Rep. 5: 12007. 2015. doi:10.1038/srep12007. PMC 4490394free to read. PMID 26137992.
  2.  Fizazi K, Albiges L, Loriot Y, Massard C (2015). “ODM-201: a new-generation androgen receptor inhibitor in castration-resistant prostate cancer”. Expert Rev Anticancer Ther. 15(9): 1007–17. doi:10.1586/14737140.2015.1081566. PMID 26313416.
  3.  Moilanen AM, Riikonen R, Oksala R, Ravanti L, Aho E, Wohlfahrt G, Nykänen PS, Törmäkangas OP, Palvimo JJ, Kallio PJ (2015). “Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies”. Sci Rep. 5: 12007.doi:10.1038/srep12007. PMC 4490394free to read. PMID 26137992.
  4.  “ODM-201 is safe and active in metastatic castration-resistant prostate cancer”. Cancer Discov. 4 (9): OF10. 2014. doi:10.1158/2159-8290.CD-RW2014-150. PMID 25185192.
  5. Pinto Á (2014). “Beyond abiraterone: new hormonal therapies for metastatic castration-resistant prostate cancer”. Cancer Biol. Ther. 15 (2): 149–55. doi:10.4161/cbt.26724.PMC 3928129free to read. PMID 24100689.
  6. Fizazi K, Massard C, Bono P, Jones R, Kataja V, James N, Garcia JA, Protheroe A, Tammela TL, Elliott T, Mattila L, Aspegren J, Vuorela A, Langmuir P, Mustonen M (2014). “Activity and safety of ODM-201 in patients with progressive metastatic castration-resistant prostate cancer (ARADES): an open-label phase 1 dose-escalation and randomised phase 2 dose expansion trial”. Lancet Oncol. 15 (9): 975–85. doi:10.1016/S1470-2045(14)70240-2. PMID 24974051.
  7.  Agarwal N, Di Lorenzo G, Sonpavde G, Bellmunt J (2014). “New agents for prostate cancer”. Ann. Oncol. 25 (9): 1700–9. doi:10.1093/annonc/mdu038. PMID 24658665.

External links

Fenner A. Prostate cancer: ODM-201 tablets complete phase I. Nat Rev Urol. 2015 Dec;12(12):654. doi: 10.1038/nrurol.2015.268. Epub 2015 Nov 3. PubMed PMID: 26526759.

2: Massard C, Penttinen HM, Vjaters E, Bono P, Lietuvietis V, Tammela TL, Vuorela A, Nykänen P, Pohjanjousi P, Snapir A, Fizazi K. Pharmacokinetics, Antitumor Activity, and Safety of ODM-201 in Patients with Chemotherapy-naive Metastatic Castration-resistant Prostate Cancer: An Open-label Phase 1 Study. Eur Urol. 2015 Oct 10. pii: S0302-2838(15)00964-1. doi: 10.1016/j.eururo.2015.09.046. [Epub ahead of print] PubMed PMID: 26463318.

3: Fizazi K, Albiges L, Loriot Y, Massard C. ODM-201: a new-generation androgen receptor inhibitor in castration-resistant prostate cancer. Expert Rev Anticancer Ther. 2015;15(9):1007-17. doi: 10.1586/14737140.2015.1081566. PubMed PMID: 26313416; PubMed Central PMCID: PMC4673554.

4: Bambury RM, Rathkopf DE. Novel and next-generation androgen receptor-directed therapies for prostate cancer: Beyond abiraterone and enzalutamide. Urol Oncol. 2015 Jul 7. pii: S1078-1439(15)00269-0. doi: 10.1016/j.urolonc.2015.05.025. [Epub ahead of print] Review. PubMed PMID: 26162486.

5: Moilanen AM, Riikonen R, Oksala R, Ravanti L, Aho E, Wohlfahrt G, Nykänen PS, Törmäkangas OP, Palvimo JJ, Kallio PJ. Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies. Sci Rep. 2015 Jul 3;5:12007. doi: 10.1038/srep12007. PubMed PMID: 26137992; PubMed Central PMCID: PMC4490394.

6: Thibault C, Massard C. [New therapies in metastatic castration resistant prostate cancer]. Bull Cancer. 2015 Jun;102(6):501-8. doi: 10.1016/j.bulcan.2015.04.016. Epub 2015 May 26. Review. French. PubMed PMID: 26022286.

7: Bjartell A. Re: activity and safety of ODM-201 in patients with progressive metastatic castration-resistant prostate cancer (ARADES): an open-label phase 1 dose-escalation and randomised phase 2 dose expansion trial. Eur Urol. 2015 Feb;67(2):348-9. doi: 10.1016/j.eururo.2014.11.019. PubMed PMID: 25760250.

8: De Maeseneer DJ, Van Praet C, Lumen N, Rottey S. Battling resistance mechanisms in antihormonal prostate cancer treatment: Novel agents and combinations. Urol Oncol. 2015 Jul;33(7):310-21. doi: 10.1016/j.urolonc.2015.01.008. Epub 2015 Feb 21. Review. PubMed PMID: 25708954.

9: Boegemann M, Schrader AJ, Krabbe LM, Herrmann E. Present, Emerging and Possible Future Biomarkers in Castration Resistant Prostate Cancer (CRPC). Curr Cancer Drug Targets. 2015;15(3):243-55. PubMed PMID: 25654638.

10: ODM-201 is safe and active in metastatic castration-resistant prostate cancer. Cancer Discov. 2014 Sep;4(9):OF10. doi: 10.1158/2159-8290.CD-RW2014-150. Epub 2014 Jul 9. PubMed PMID: 25185192.

11: Fizazi K, Massard C, Bono P, Jones R, Kataja V, James N, Garcia JA, Protheroe A, Tammela TL, Elliott T, Mattila L, Aspegren J, Vuorela A, Langmuir P, Mustonen M; ARADES study group. Activity and safety of ODM-201 in patients with progressive metastatic castration-resistant prostate cancer (ARADES): an open-label phase 1 dose-escalation and randomised phase 2 dose expansion trial. Lancet Oncol. 2014 Aug;15(9):975-85. doi: 10.1016/S1470-2045(14)70240-2. Epub 2014 Jun 25. PubMed PMID: 24974051.

12: Agarwal N, Di Lorenzo G, Sonpavde G, Bellmunt J. New agents for prostate cancer. Ann Oncol. 2014 Sep;25(9):1700-9. doi: 10.1093/annonc/mdu038. Epub 2014 Mar 20. Review. PubMed PMID: 24658665.

13: Pinto Á. Beyond abiraterone: new hormonal therapies for metastatic castration-resistant prostate cancer. Cancer Biol Ther. 2014 Feb;15(2):149-55. doi: 10.4161/cbt.26724. Epub 2013 Nov 1. Review. PubMed PMID: 24100689; PubMed Central PMCID: PMC3928129.

14: Yin L, Hu Q, Hartmann RW. Recent progress in pharmaceutical therapies for castration-resistant prostate cancer. Int J Mol Sci. 2013 Jul 4;14(7):13958-78. doi: 10.3390/ijms140713958. Review. PubMed PMID: 23880851; PubMed Central PMCID: PMC3742227.

15: Leibowitz-Amit R, Joshua AM. Targeting the androgen receptor in the management of castration-resistant prostate cancer: rationale, progress, and future directions. Curr Oncol. 2012 Dec;19(Suppl 3):S22-31. doi: 10.3747/co.19.1281. PubMed PMID: 23355790; PubMed Central PMCID: PMC3553559.

Darolutamide
ODM-201.svg
Systematic (IUPAC) name
N-((S)-1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-5-(1-hydroxyethyl)-1H-pyrazole-3-carboxamide[1]
Identifiers
ChemSpider 38772320
UNII X05U0N2RCO Yes
Chemical data
Formula C19H19ClN6O2
Molar mass 398.85 g·mol−1

//////////// Bayer HealthCare,  Orion,  Antineoplastics,  Androgen receptor antagonists, Phase III, Prostate cancer, BAY 1841788,  ODM-201

O=C(N[C@@H](C)Cn1ccc(n1)c2ccc(C#N)c(Cl)c2)c3cc(nn3)C(O)C

Day 8 of the 2016 Doodle Fruit Games! Find out more at g.co/fruit


Filed under: Phase3 drugs Tagged: Androgen receptor antagonists, Antineoplastics, BAY 1841788, Bayer HealthCare, ODM 201, ORION, Phase III, Prostate cancer

Flow Grignard and Lithiation: Screening Tools and Development of Continuous Processes for a Benzyl Alcohol Starting Material

$
0
0

ORGANIC CHEMISTRY SELECT

str1

Abstract Image

Efficient continuous Grignard and lithiation processes were developed to produce one of the key regulatory starting materials for the production of edivoxetine hydrochoride. For the Grignard process, organometallic reagent formation, Bouveault formylation, reduction, and workup steps were run in continuous stirred tank reactors (CSTRs). The lithiation utilized a hybrid approach where plug flow reactors (PFRs) were used for the metal halogen exchange and Bouveault formylation steps, while the reduction and workup steps were performed in CSTRs. Relative to traditional batch processing, both approaches offer significant advantages. Both processes were high-yielding and produced the product in high purity. The two main processes were directly compared from a number of perspectives including reagent and operational safety, fouling potential, process footprint, need for manual operation, and product yield and purity.

Flow Grignard and Lithiation: Screening Tools and Development of Continuous Processes for a Benzyl Alcohol Starting Material

View original post 103 more words


Filed under: Uncategorized
Viewing all 1646 articles
Browse latest View live