Quantcast
Channel: New Drug Approvals
Viewing all 2871 articles
Browse latest View live

Chenodeoxycholic acid, ケノデオキシコール酸

$
0
0

Skeletal formula of chenodeoxycholic acid

ChemSpider 2D Image | chenodeoxycholic acid | C24H40O4Chenodeoxycholic acid.png

Chenodeoxycholic acid

Chenodiol

  • Molecular FormulaC24H40O4
  • Average mass392.572
UNII-0GEI24LG0J
ケノデオキシコール酸
474-25-9 [RN]
chenodeoxycholic acid [JP15] [Wiki]
(+)-chenodeoxycholic acid
(3a,5b,7a)-3,7-dihydroxy-cholan-24-oic acid
(3α,5β,7α,8ξ,20R)-3,7-Dihydroxycholan-24-säure[German] [ACD/IUPAC Name]
(4R)-4-[(3R,5S,7R,8R,9S,10S,13R,14S,17R)-3,7-Dihydroxy-10,13-dimethylhexadecahydro-1H-cyclopenta[a]phenanthren-17-yl]pentanoic acid
0GEI24LG0J
17b-(1-Methyl-3-carboxypropyl)etiocholane-3a,7a-diol
207-481-8[EINECS]
3a,7a-dihydroxy-5b-cholan-24-oic acid
3a,7a-dihydroxy-5b-cholanic acid; anthropodesoxycholic acid; gallodesoxycholic acid; 17b-(1-methyl-3-carboxypropyl)etiocholane-3a,7a-diol; chenic acid; chenodeoxycholic acid; CDC
Chenodeoxycholate;
Chenodeoxycholic acid;
3alpha,7alpha-Dihydroxy-5beta-cholanic acid;
Chenodiol

Synthesis ReferenceHenry Francis Frost, Fritz Fabian, Christopher James Sharpe, William Arthur Jones, “Process for preparing chenodeoxycholic acid.” U.S. Patent US4022806, issued October, 1974. US4022806

First ref 

  • By Windaus, A.; Bohne, A.; Schwarzkopf, E.
  • From Z. physiol. Chem. (1924), 140, 177-85
  • By Wieland, Heinrich; Reverey, Gustav
  • From Z. physiol. Chem. (1924), 140, 186-202.  

Title: Chenodiol
CAS Registry Number: 474-25-9
CAS Name: (3a,5b,7a)-3,7-Dihydroxycholan-24-oic acid
Additional Names: 3a,7a-dihydroxy-5b-cholanic acid; anthropodesoxycholic acid; gallodesoxycholic acid; 17b-(1-methyl-3-carboxypropyl)etiocholane-3a,7a-diol; chenic acid; chenodeoxycholic acid; CDC
Trademarks: Chendol (CP Pharm.); Chenocol (Astellas); Chenofalk (Falk); Chenossil (Sanofi-Aventis); Cholanorm (Grñenthal); Fluibil (Zambon)
Molecular Formula: C24H40O4
Molecular Weight: 392.57
Percent Composition: C 73.43%, H 10.27%, O 16.30%
Literature References: A major bile acid in many vertebrates, occurring as the N-glycine and/or N-taurine conjugate. With other bile acids, forms mixed micelles with lecithin in bile which solubilize cholesterol and thus facilitates its excretion. Facilitates fat absorption in the small intestine by micellar solubilization of fatty acids and monoglycerides. Has cathartic properties since it induces fluid secretion from large intestine. Main constituent of the bile of hens, geese and other fowl; occurs in appreciable amounts in the bile of hamster, hog, guinea pig, bear and man. Epimeric with ursodiol, q.v. Isoln: Windhaus et al.,Z. Physiol. Chem.140, 177 (1924); Wieland, Reveney, ibid. 186. Configuration: Lettré, Ber.68, 766 (1935). Prepn from cholic acid: Fieser, Rajagopalan, J. Am. Chem. Soc.72, 5530 (1950); Hauser et al.,Helv. Chim. Acta43, 1595 (1960); Hofmann, Acta Chem. Scand.17, 173 (1963). Alternate prepns: Sato, Ikekawa, J. Org. Chem.24, 1367 (1959); T. Iida, F. C. Chang, ibid.46, 2786 (1981). Stereoselective total synthesis: T. Kametani et al.,J. Am. Chem. Soc.103, 2890 (1981). Asymmetric total synthesis of (+)-form:eidem,J. Org. Chem.47, 2331 (1982). Dissolution of cholesterol gallstones: Danzinger et al.,N. Engl. J. Med.286, 1 (1972); Bell et al.,LancetII, 1213 (1972). Use in long-term treatment of cerebrotendinous xanthomatosis: V. M. Berginer et al.,N. Engl. J. Med.311, 1649 (1984). Monograph on bile acids: The Bile Acids, 2 vols., P. P. Nair, D. Kritchevsky, Eds. (Plenum Press, New York, 1971, 1973). Review of pharmacology and therapeutic use of chenodeoxycholic acid: J. H. Iser, A. Sali, Drugs21, 90-119 (1981). Effect on cholesterol and bile acid metabolism: G. S. Tint et al.,Gastroenterology91, 1007 (1986).
Properties: Needles from ethyl acetate + heptane, mp 119°. [a]D20 +11.5° (dioxane). Freely sol in methanol, alc, acetone, acetic acid; more sol in ether and ethyl acetate than deoxycholic acid. Practically insol in water, petr ether, benzene. High solvent power for alkali soaps, but does not form “choleic” acid addition compds as does deoxycholic acid. Forms beautiful cryst salts of Na, K and Ba. While the acid is tasteless, the Na salt tastes slightly sweet at first, then bitter.
Melting point: mp 119°
Optical Rotation: [a]D20 +11.5° (dioxane)
Derivative Type: Diformate
Molecular Formula: C25H40O6
Molecular Weight: 436.58
Percent Composition: C 68.78%, H 9.23%, O 21.99%
Properties: Clusters of needles from alc; mp with slight effervescence at 137°, upon further heating solidifies again, and finally melts around 172°.
Melting point: mp with slight effervescence at 137°
Derivative Type: Methyl ester
Molecular Formula: C25H42O4
Molecular Weight: 406.60
Percent Composition: C 73.85%, H 10.41%, O 15.74%
Properties: Fine needles from benzene + heptane, mp 90-91°. [a]D25 +20°.
Melting point: mp 90-91°
Optical Rotation: [a]D25 +20°
Therap-Cat: Anticholelithogenic.
Keywords: Cholelitholytic Agent.
SPECIFIC ROTATION
+13.23 °   ethanol ,  589.3 nm;  21 °C, Yonemura, Sadatomo; Journal of Biochemistry 1926, Vol6, Pg287-96
+12.5 °  chloroform, 589.3 nm; 17 °C  Plattner, Pl. A.; Helvetica Chimica Acta 1944, Vol27, Pg748-57
MP
Chenodeoxycholic acid (or Chenodiol) is an epimer of ursodeoxycholic acid (DB01586). Chenodeoxycholic acid is a bile acid naturally found in the body. It works by dissolving the cholesterol that makes gallstones and inhibiting production of cholesterol in the liver and absorption in the intestines, which helps to decrease the formation of gallstones. It can also reduce the amount of other bile acids that can be harmful to liver cells when levels are elevated.

Chenodeoxycholic acid (also known as chenodesoxycholic acidchenocholic acid and 3α,7α-dihydroxy-5β-cholan-24-oic acid) is a bile acid. It occurs as a white crystalline substance insoluble in water but soluble in alcohol and acetic acid, with melting point at 165–167 °C. Salts of this carboxylic acid are called chenodeoxycholates. Chenodeoxycholic acid is one of the main bile acids produced by the liver.[1]

It was first isolated from the bile of the domestic goose, which gives it the “cheno” portion of its name (Greek: χήν = goose).[2]

Chenodeoxycholic acid and cholic acid are the two primary bile acids in humans. Some other mammals have muricholic acid or deoxycholic acid rather than chenodeoxycholic acid.[1]

Chenodeoxycholic acid is synthesized in the liver from cholesterol by a process which involves several enzymatic steps.[1] Like other bile acids, it can be conjugated in the liver with taurine or glycine, forming taurochenodeoxycholate or glycochenodeoxycholate. Conjugation results in a lower pKa. This means the conjugated bile acids are ionized at the usual pH in the intestine and will stay in the gastrointestinal tract until reaching the ileum where most will be reabsorbed. Bile acids form micelles which facilitate lipid digestion. After absorption, they are taken up by the liver and resecreted, so undergoing an enterohepatic circulation. Unabsorbed chenodeoxycholic acid can be metabolised by bacteria in the colon to form the secondary bile acid known as lithocholic acid.

Chenodeoxycholic acid is the most potent natural bile acid at stimulating the nuclear bile acid receptor, farnesoid X receptor (FXR).[3]The transcription of many genes is activated by FXR.

Indication

Chenodiol is indicated for patients with radiolucent stones in well-opacifying gallbladders, in whom selective surgery would be undertaken except for the presence of increased surgical risk due to systemic disease or age. Chenodiol will not dissolve calcified (radiopaque) or radiolucent bile pigment stones.

Associated Conditions

Pharmacodynamics

It acts by reducing levels of cholesterol in the bile, helping gallstones that are made predominantly of cholesterol to dissolve. Chenodeoxycholic acid is ineffective with stones of a high calcium or bile acid content.

Mechanism of action

Chenodiol suppresses hepatic synthesis of both cholesterol and cholic acid, gradually replacing the latter and its metabolite, deoxycholic acid in an expanded bile acid pool. These actions contribute to biliary cholesterol desaturation and gradual dissolution of radiolucent cholesterol gallstones in the presence of a gall-bladder visualized by oral cholecystography. Bile acids may also bind the the bile acid receptor (FXR) which regulates the synthesis and transport of bile acids.

EMA

On 16 December 2014, orphan designation (EU/3/14/1406) was granted by the European Commission to Sigma-Tau Pharma Ltd, United Kingdom, for chenodeoxycholic acid for the treatment of inborn errors in primary bile acid synthesis.

The sponsorship was transferred to sigma-tau Arzneimittel GmbH, Germany, in May 2015.

Chenodeoxycholic acid has been authorised in the EU as Chenodeoxycholic acid sigma-tau since 10 April 2017.

The name of the product changed to Chenodeoxycholic acid Leadiant in May 2017.

The sponsorship was transferred to Leadiant GmbH, Germany, in June 2017.

On 16 February 2017, the Committee for Orphan Medicinal Products (COMP) concluded its review of the designation EU/3/14/1406 for Chenodeoxycholic acid sigma-tau (chenodeoxycholic acid) as an orphan medicinal product for the treatment of inborn errors in primary bile acid synthesis. The COMP assessed whether, at the time of marketing authorisation, the medicinal product still met the criteria for orphan designation. The Committee looked at the seriousness and prevalence of the condition, and the existence of other methods of treatment. As other methods of treatment are authorised in the European Union (EU), the COMP also considered whether the medicine is of significant benefit to patients with inborn errors in primary bile acid synthesis. The COMP recommended that the orphan designation of the medicine be maintained1.


1 The maintenance of the orphan designation at time of marketing authorisation would, except in specific situations, give an orphan medicinal product 10 years of market exclusivity in the EU. This means that in the 10 years after its authorisation similar products with the same therapeutic indication cannot be placed on the market.

http://www.ema.europa.eu/docs/en_GB/document_library/Orphan_designation/2015/02/WC500183233.pdf

Therapeutic applications

Chenodeoxycholic acid has been used as medical therapy to dissolve gallstones.[4]

Chenodeoxycholic acid can be used in the treatment of cerebrotendineous xanthomatosis.[5]

The Australian biotechnology company Giaconda has tested a treatment for Hepatitis C infection that combines chenodeoxycholic acid with bezafibrate.[6]

As diarrhea is a complication of chenodeoxycholic acid therapy, it has also been used to treat constipation.[7][8]

In supramolecular chemistrymolecular tweezers based on a chenodeoxycholic acid scaffold is a urea receptor that can contain anionsin its binding pocket in order of affinity: H2PO4 (dihydrogen phosphate) > Cl > Br > I reflecting their basicities (tetrabutylammonium counter ion).[9]

Molecular tweezer based on chenodeoxycholic acid
PAPER
1H and 13C NMR characterization and stereochemical assignments of bile acids in aqueous media
Lipids (2005), 40, (10), 1031-1041.
https://onlinelibrary.wiley.com/doi/abs/10.1007/s11745-005-1466-1

PAPER

Improved Chemical Synthesis, X-Ray Crystallographic Analysis, and NMR Characterization of (22R)-/(22S)-Hydroxy Epimers of Bile Acids
Lipids (2014), 49, (11), 1169-1180.

Improved Chemical Synthesis, X‐Ray Crystallographic Analysis, and NMR Characterization of (22R)‐/(22S)‐Hydroxy Epimers of Bile Acids

PAPER

A Practical and Eco-friendly Synthesis of Oxo-bile Acids

By Han, Young Taek and Yun, HwayoungFrom Organic Preparations and Procedures International, 48(1), 55-61; 2016

DOI:10.1080/00304948.2016.1127101

General Procedure

An aqueous solution of 0.2 M NaBrO3 (1.5 equiv. per hydroxy group) was added dropwise to a slurry of bile acid (1 equiv.) and ceric ammonium nitrate (0.05 equiv.) in 20% aqueous acetonitrile (0.2 M) at 80°C over 20 min. The bile acid slowly dissolved in a few minutes, and then the color of the reaction mixture changed to orange. The reaction mixture was stirred at the same temperature and the progress of the reaction was monitored by TLC on silica gel (1:20 MeOH-CH2Cl2) until disappearance of the starting material and partially oxidized intermediates. It was then cooled in an ice bath and quenched with aqueous Na2S2O3 solution. Water was added slowly to the resulting white suspension until no more oxo-bile acid precipitated. The white solid was collected, washed with water until the filtrate was colorless, and then dried in vacuo at 50°C. Methyl 3,7α-Diacetoxy-12-oxo-5β-cholanoate(3),21 was obtained in 92% yield (275 mg) as a white solid from 300 mg (0.590 mmol) of 2 via the general procedure. mp. 176-178°C, lit.22 mp. 178-179°C, IR (thin film, neat): 2947 (m), 2873 (s), 1736 (w), 1706 (w), 1436 (s), 1365 (m) cm-1; 1H-NMR (400 MHz, CDCl3): δ 4.96 (m, 1H, 7-CH), 4.55 (m, 1H, 3-CH), 3.64 (s, 3H), 2.49 (t, 1H, J = 12.6 Hz), 2.41-0.80 (m, 23H), 2.01 (s, 3H), 2.00 (s, 3H), 1.01 (s, 3H, 18-CH3), 1.00 (s, 3H, 19-CH3), 0.83 (d, 3H, J = 6.6 Hz, 21-CH3); 13C-NMR (CDCl3, 100 MHz): δ 214.0 (12-C), 174.6 (24-C), 170.7 (C = O), 170.2 (C = O), 73.5 (3-C), 70.5 (7-C), 57.1 (13-C), 53.1 (14-C), 51.5 (CH3O), 46.3 (17-C), 40.5 (5-C), 37.9 (11-C), 37.8 (4-C), 37.6 (8-C), 35.54 (9-C), 35.52 (20-C), 34.9 (1-C), 34.5 (10-C), 31.3 (6-C), 31.2 (22-C), 30.4 (23-C), 27.4 (16-C), 26.5 (2-C), 23.8 (15-C), 22.1 (19-C), 21.51 (CH3CO2), 21.46 (CH3CO2), 18.6 (21-C), 11.5 (18-C); LR-MS (FABC) m/z 505 (M+H +). HR-MS (FABC): Calcd for C29H45O7 (M+H +): 505.3165. Found 505.3161.

next step

R:KOH, R:N2H4

NOTE STARTING  IS BILE ACID AS BELOW

Cholan-24-oic acid, 3,7-bis(acetyloxy)-12-oxo-, methyl ester, (3α,5β,7α)-

  • 5β-Cholan-24-oic acid, 3α,7α-dihydroxy-12-oxo-, methyl ester, diacetate (8CI)
  • 5β-Cholanic acid, 3α,7α-dihydroxy-12-oxo-, methyl ester, diacetate (6CI,7CI)
  • 3α,7α-Diacetoxy-12-oxo-5β-cholan-24-oic acid methyl ester
  • Methyl 3α,7α-diacetoxy-12-oxo-5β-cholan-24-oate
  • Methyl 3α,7α-diacetoxy-12-oxo-5β-cholanate
CAS 28535-81-1
C29 H44 O7  504.66
Cholan-24-oic acid, 3,7-bis(acetyloxy)-12-oxo-, methyl ester, (3α,5β,7α)-

PAPER

https://pubs.acs.org/doi/pdf/10.1021/jo01091a623

Journal of Organic Chemistry
Volume24
Pages1367-8
Journal
1959

DOI:10.1021/jo01091a623

Chenodeoxycholic acid (V). Five hundred mg. of the above ester IV was hydrolyzed with 80 ml. of ethanolic 5% potassium hydroxide for 4 hr. After partial concentration of the volume and addition of water, the reaction product was acidified with hydrochloric acid. The resulting precipitate was collected, dried, and crystallized from ethyl acetate. A quantitative crop (400 mg.) of prisms melting at 143- 145° were obtained. Recrystallization from the same solvent yielded a product of m.p. 145-146°, [ ]2 +10.7° (dioxane). Anal. Caled, for C24H40O4: C, 73.43; H, 10.27. Found: C, 73.49; H, 10.31.

NOTE I IS BILE ACID

Cholan-24-oic acid, 3,7-bis(acetyloxy)-12-oxo-, methyl ester, (3α,5β,7α)-

  • 5β-Cholan-24-oic acid, 3α,7α-dihydroxy-12-oxo-, methyl ester, diacetate (8CI)
  • 5β-Cholanic acid, 3α,7α-dihydroxy-12-oxo-, methyl ester, diacetate (6CI,7CI)
  • 3α,7α-Diacetoxy-12-oxo-5β-cholan-24-oic acid methyl ester
  • Methyl 3α,7α-diacetoxy-12-oxo-5β-cholan-24-oate
  • Methyl 3α,7α-diacetoxy-12-oxo-5β-cholanate
CAS 28535-81-1
C29 H44 O7  504.66
Cholan-24-oic acid, 3,7-bis(acetyloxy)-12-oxo-, methyl ester, (3α,5β,7α)-

PATENT

https://patents.google.com/patent/CN102060902A/en

chenodeoxycholic acid (3 α, 7 α – dihydroxy _5 β – cholestane-24-oic acid) Chenodeoxycholic Ac id (referred to as CDCA), clinically used to correct dissolving cholesterol calculi and bile saturation drugs, the main function is to reduce the cholesterol in the bile saturation, large doses can inhibit the synthesis of cholesterol CDCA and increasing bile gallstone patients cholesterol level in a non-saturated, thereby preventing the formation of cholesterol gallstones of cholesterol and promote stone dissolve and fall off. It also has significant anti-asthmatic, anti-inflammatory, antitussive and expectorant effects.

[0003] Synthesis of chenodeoxycholic acid or ursodeoxycholic acid (3 α, 7β_ -5β_ dihydroxy-cholestane-24-oic acid, ursodeoxycholic Acid, referred UDCA), a key intermediate. Ursodeoxycholic acid is the main active ingredient of precious Chinese medicine bear bile, used in a variety of clinical hepatobiliary disease and dyspepsia. Currently we bear bile resources are scarce, mainly used synthetic chemical ursodeoxycholic acid as a clinical treatment. Therefore, the preparation of chenodeoxycholic acid is also important for the preparation of ursodeoxycholic acid.

[0004] CDCA mainly come from poultry or livestock bile extraction. Traditional extraction process complicated operation, low yield, (pharmaceutical industry, 1987,18 (9), 416; Chinese Journal of Biochemical Pharmaceutics, 1996,17 (1), 17; Applied Technology, 1998, (4), 9; CN1850846A ) can not meet the needs of modern industry. Chemical synthesis of chenodeoxycholic acid have also been reported (Japanese Journal of Chemistry 1955,76 (3), 297 -J Org Chem 1982,47 (2): 2331; Journal of Biochemical Pharmaceutics 1987,1,6 -, Tap Chi Duoc ^ oc2004 , 44 (1), 11; CN1869043A), but lower yield widespread pollution major problem, especially in the oxidation reaction is often used to expensive, and polluting agents.Therefore, to reduce pollution, reduce environmental hazards, streamline operations, improve yield, reduce costs, important for the synthesis of chenodeoxycholic acid.

 Figure CN102060902AD00041

n particular by the following steps:

(1) Preparation of cholate: bile acid in alcohol, concentrated hydrochloric acid as catalyst, at reflux, cooling and crystallization, filtration, and washed with methanol.

[0008] (2) Preparation of 3α, 7α- diacetyl hydroxy -12α- cholate: bile acid ester was dissolved in dichloromethane and triethylamine was added with stirring acetic anhydride and the catalyst N, N- dimethyl pyridine, methylene chloride was distilled off, poured into water, filtered to give 3α, 7α- diacetyl -12 α – hydroxy cholate.

[0009] (3) 3α, 7α- diacetyl -12– Preparation oxo chenodeoxycholic acid ester: Take 3 α, 7 α – diacetyl -12 α – hydroxy cholate dissolved in ethyl acetate and methanol, bromide and tetrabutylammonium bromide as catalyst, and acetic acid was added dropwise under stirring hypochlorite, the organic solvent was distilled off and filtered, to give 12-oxo-3,7-diacetyl Chenodeoxy cholate.

[0010] (4) i2 – Preparation oxo chenodeoxycholic acid: 3,7-diacetyl-12-oxo-chenodeoxycholic acid ester added ethanol – sodium hydroxide solution, at reflux.PH adjusted with hydrochloric acid value of the reaction system acidic, ethanol was distilled off, and filtered to give 12- oxo crude chenodeoxycholic acid, fine recrystallization.

[0011] Preparation of chenodeoxycholic acid (5): 12- oxo take chenodeoxycholic acid, ethylene glycol and solid sodium hydroxide, hydrated corpus, refluxed for 2 hours, gradually warming evaporated partially hydrated corpus, continue to heat up to 150 ° C, continued to reflux, cooled to room temperature, poured into water, adjusting the PH with hydrochloric acid, the white precipitate was filtered, washed with water to give crude chenodeoxycholic acid, recrystallization

Azusa mouth

M ο not mesh

[0012] Step (1): cholic acid to alcohol weight to volume ratio of 1: 2 ~ 5, the volume ratio of concentrated hydrochloric acid to alcohol is 10 wide: 100, 5-5 hours reflux time was 0.5.

[0013] Step (2): cholate: acetic anhydride molar ratio = 1: 2 ~ 5, the reaction temperature, time; Tl2O hours; cholate was added per mole of N, N- dimethylpyridine wide 5g.

[0014] Step (; 3): The hypochlorite is sodium hypochlorite or calcium hypochlorite; bromide is sodium bromide, potassium bromide and the like.

[0015] Step (4): recrystallization from a solvent with an alcohol such: as methanol or ethanol.

[0016] Step (5): recrystallization solvent is a water-miscible organic solvents, such as: methanol, ethanol, acetonitrile, acetone and the like.

[0017] Step (cholate was used ¾ of methyl cholate, ethyl cholate, cholic acid or cholic acid propyl ester; Step (3) used as 3 [alpha], 7 α – diacetyl -12 α – hydroxy cholate as 3 α, 7 α – diacetyl -12 α – hydroxy methyl cholate, 3 α, 7α- diacetyl -12 α – hydroxy bile acid ethyl ester, 3 α, 7α- diacetyl yl -12 α – hydroxy acid or ester 3α, 7α- diacetyl -12 α – hydroxy acid ester.

[0018] The invention has the advantages: in cholic acid as raw materials, and the choice of bromide tetrabutylammonium bromide as catalyst, in a non-polluting oxidizing agent is hypochlorite, Intermediate 3 α, 7 α – Diacetyl _12_ oxo chenodeoxycholic acid ester yield of 90% or more, thereby improving the yield of the final product of chenodeoxycholic acid, 99% yield, low cost and no pollution, very convenient for industrial production. detailed description

[0019] The present invention will be better described, for example is as follows:

(1) Preparation of methyl cholate: bile acid 5. lg, 15ml of anhydrous methanol, heating the whole solution. Refluxed for 3 hours, was added 0. 4ml concentrated hydrochloric acid, the reaction was stopped after 30min, after slow cooling, and filtered to give methyl cholate 5. 05g, 95% yield. 1HNMR (CDCl3):. Δ 0. 70 (s, 3H, 18- CH3), 0.90 (s, 3H, 19- CH3), 0.98 (d, 3H, 21-CH3), 3 50 (m, 1H, 3 β -H), 3. 67 (s, 3H, OCH3), 3. 87 (s, 1H, 7 β -H), 3. 99 (s, 1H, 12 β -H).

[0020] (2) Preparation of 3α, 7α- methyl cholate diacetyl-hydroxy -12α-: bile acid methyl ester 4. 71g (Ilmmol) IOOml was placed in a flask, was added methylene chloride 30ml, triethylamine 3 . Chiu 1, stirred at room temperature, was added dropwise acetic anhydride 2. 7ml (28. 6mmo 1), followed by addition of 20mg N, N- dimethylpyridine catalyst, the reaction time of 7 hours, methylene chloride was distilled off, into the water, filtered to give a white solid. The crude product was recrystallized from methanol to give white crystals 4. 05g, yield 67.2%. 1H NMR (CDCl3) δ: 4.90 (m, 1H, 7 β -H), 4. 59 (s, 1H, 3 β -H), 4 01 (s, 1H, 12 β -H), 3 67.. (s, 3Η, OCH3), 2. 08 (s, 3Η, CH3CO), 2. 02 (s, 3Η, CH3CO), 0. 98 (s, 3Η, 21-CH3), 0. 93 (s, 3Η , 19-CH3), 0.69 (s, 3Η, 18_CH3).

[0021] (3) 3α, 7α – 12-oxo-diacetyl chenodeoxycholic acid methyl ester prepared: Take 3 α, 7 α – diacetyl -12 α- hydroxy methyl cholate 1.917 g ( 3. 79mmol) was placed in a 50ml round bottom flask, 12ml of ethyl acetate was added, 5ml methanol, stirring at room temperature, was added 0. 25g 0. Ig of potassium bromide and tetrabutylammonium bromide. Was added dropwise a solution of acetic acid and 6g of sodium hypochlorite (7%) (5.62mmol), for 10 hours. Methanol was distilled off under reduced pressure and ethyl acetate, filtered, washed with water, and dried to give crude 1.915g, 1.75g as a white solid after recrystallization from methanol, yield 91.2%. 1H bandit R (CDCl3) δ:.. 4. 99 (d, 1H, 7 β-H), 4 60 (m, 1H, 3 β-H), 3 67 (s, 3H, OCH3), 2. 07 (s, 6H, CH3CO), 1. 03 (s, 6H, I8-CH3 and 19-CH3), 0. 82 (d, 3H, 21-CH3) ο

[0022] (4) 12- oxo chenodeoxycholic acid Preparation: Take 3 α, 7 α – diacetyl _12_ oxo chenodeoxycholic acid methyl ester 1. 56g, was dissolved in 30ml 95% ethanol was added 3. 2g of sodium hydroxide, heated at reflux for 5 hours. PH adjusted with hydrochloric acid value of the reaction system, most of the ethanol was distilled off, filtered, washed with water, and dried to give a white solid 12- oxo-1 crude chenodeoxycholic acid, recrystallized from methanol ^ g 1. 25g, yield rate of 96%. Tun bandit R (CDCl3) δ:. 3.96 (d, 1H, 7 β-H), 3 47 (m, 1H, 3 β-H), 1.03 (s, 3H, 19_CH3), 0.89 (s, 3H, 18_CH3 ), 0 · 70 (d, 3 H, 21_CH3).

[0023] Preparation of chenodeoxycholic acid (5): 12- oxo take chenodeoxycholic acid 0. 9g, 15ml ethylene glycol was added solid sodium hydroxide and 1. 5g, 15ml hydrated corpus (80%) , 120 ° C reflux for 2 hours, change return device is a distillation apparatus, was gradually warmed evaporated amount hydrated corpus, continue to heat up to 150 ° C, continuing reflux for 4h, cooled to room temperature, poured into water, adjusted with HCl of PH3, white precipitated, was filtered cake was washed with water, and dried to give crude chenodeoxycholic acid 0. 92g, recrystallized from methanol to give 0. 86g, 99 (s, 1H, C00H).

Paper

https://pubs.acs.org/doi/abs/10.1021/ja01168a045

Reactions of 2-Arylcyclohexanones. IV. Michael Addition of Malonic Ester to 2-Phenyl-Δ2-cyclohexenone

J. Am. Chem. Soc.195072 (12), pp 5529–5530
DOI: 10.1021/ja01168a045
Publication Date: December 1950
PAPER
Hauser et al., Helv. Chim. Acta 43, 1595 (1960);
Paper

The Preparation of Chenodeoxycholic Acid and Its Glycine and Taurine Conjugates.Hofmann, Alan F.

Pages: 173-186.
DOI number: 10.3891/acta.chem.scand.17-0173
Download as: PDF DjVu
PAPER
Sato, Ikekawa, J. Org. Chem. 24, 1367 (1959)

Preparation of Chenodeoxycholic Acid

J. Org. Chem.195924 (9), pp 1367–1368
DOI: 10.1021/jo01091a623
Publication Date: September 1959
PAPER
J. Org. Chem. 47, 2331 (1982)

Further studies on the synthesis of thienamycin: a facile and stereoselective synthesis of a bicyclic .beta.-keto ester by 1,3-dipolar cycloaddition

J. Org. Chem.198247 (12), pp 2328–2331
DOI: 10.1021/jo00133a019
PAPER
PATENT

Chenodeoxycholic acid (3α, 7α- -5β- dihydroxy-cholestane acid) Chenodeoxycholic Acid (referred to as CDCA), a medicine for treating gallstones. 1848 first discovered in goose bile, 1924, known as the CDCA. By reducing cholesterol absorption, synthesis, the bile cholesterol decreased, thereby suppressing cholesterol gallstone formation and promote dissolution, and can reduce cholesterol saturation.

Chenodeoxycholic acid addition pharmaceutically itself, but also as the preparation of ursodeoxycholic acid (3α, 7β- -5β- dihydroxy bile acid, abbreviated UDCA) starting material. Ursodeoxycholic acid is the main active ingredient contained bile valuable medicine, in clinical treatment of various gastrointestinal diseases and bladder diseases. But the limited sources of bear bile medicine, and contrary to the principles of animal protection. So, dwindling source of natural bear bile, can not meet the medical requirements. Therefore, the preparation of chenodeoxycholic acid is also of great significance for further preparation of ursodeoxycholic acid.

CDCA bile extracted mainly from poultry or animal bile extraction methods in the past as it involves toxic chemicals (animal biological pharmacy, 1981, People’s Medical Publishing House, P259; pharmaceutical industry, 1987,18 (2): 75-76; ) or unsafe to use a large amount of organic solvent (Chinese Journal of biochemical Pharmaceutics, 1996,17 (1): 17; application technology, 1998,4: 9-10; US Patent, 3,965,131; US Patent, 4,331,607; USPatent, 4,163,017), can not be meet the requirements of modern industry, CDCA and low purity prepared costly.

PATENT

https://patents.google.com/patent/WO2007069814A1/en

Chenodeoxycholic acid is generally contained in bile of cow, swine, bear, or poultry such as chicken or goose, as well as in bile of human. Chenodeoxycholic acid is used as starting material for the preparation of ursodeoxycholic acid which is effective to alleviate biliary system diseases, hyperlipidemia, cholelithiasis, and chronic liver diseases, and a typical process for preparing ursodeoxycholic acid known in the art is as follows.

A typical process for preparing chenodeoxycholic acid comprises the steps of: esterifying cholic acid (3α,7α,12θ!-trihydroxy cholic acid) with methyl; protecting the hydroxyl group of 3α and Ia position by acetylating them with anhydrous acetic acid; oxidizing the hydroxyl group of 12α position to carbonyl group by using chromic acid, and then removing the carbonyl group by Wolff-kichner reduction reaction; hydrolyzing and deprotecting the obtained product to yield chenodeoxycholic acid. The above process requires the reaction to be maintained at a high temperature of more than 200 °C , and the supply of raw material may be interrupted by bovine spongiform encephalopathy, etc. Bile ,of poultry contains chenodeoxycholic acid, lithocholic acid, and a small amount of cholic acid. Thus, the process for separating chenodeoxycholic acid from poultry is well known in the art, but is not economically reasonable due to the supply decrease of raw material and low yield [see, Windhaus et al, I Physiol. Chem., 140, 177-185 (1924)].

US Patent No. 4,186,143 disclosed a process for purely separating and purifying chenodeoxycholic acid from chenodeoxycholic acid mixture derived from natural swine bile. This process comprises the major steps of: pre-treatment to remove 3ohydroxy-6- oxo-5/3-cholic acid by saponification of bile; esterification of bile acid; acetylation of bile acid ester; removal of intermediate product by using non-polar organic solvent; crystallization of acetylated ester of formula I; deprotection; and production of the compound of formula I by using crystallization in organic solvent. However, this patent does not describe HPLC content for acetylated ester of formula I, and the purity of the final product is very low since the specific rotatory power is [ofo25 +13.8° (c=l, CHCl3), and the melting point is 119-121 °C [STD: [α]D 25 +15.2°(c=l, CHCl3), melting point 127- 129 “C]. Also, the crystallization for purifying the final product requires a very long time (i.e., 16-48 hours), and the entire process is complex as eight (8) steps. Thus, when purifying the compound of formula I by using the above process, the yield of the final product becomes low, and the reaction time is as long as 12 days. Therefore, the process is not economically reasonable.

Step 6: Deprotection and crystallization of chenodeoxycholic acid

To 220ml of water were added 24.5g of chenodeoxycholic acid-diacetate-ester and 29.5g of sodium hydroxide, and then the solution was stirred with reflux for 4 hours. To the solution was added 370ml of water. The solution’s pH is adjusted to 2.0-3.0 by using 59ml of hydrochloric acid. Then, the solution was stirred at 35-45 °C for 1 hour, and then filtered. The filtered material was washed with 24.5ml of water and dried in vacuum at 70 °C to obtain 19.5g of pure chenodeoxycholic acid, m.p.: 160-161 °C, [α]o25 +13.0°(c=l, CHCl3).

Step 8: Production of the compound of formula I

The reaction solution was extracted by using ethyl acetate, and aqueous layer was discarded therefrom. Ethyl acetate layer in the solution was washed with 6% saline, and the solution was distilled to about 90ml. This solution was cooled, kept cool for one day after adding 90ml of hexane, and filtered. Thus filtered material was washed with 20ml of hexane, and dried in vacuum at 60 °C to produce 12.7g of chenodeoxycholic acid. m.p. 142-1450C; [α]D 25 +13.0°(c=l, CHCl3). INDUSTRIAL APPLICABILITY The present invention can purify chenodeoxycholic acid of formula I from swine bile solid in high yield and purity. Also, the present invention is suitable for industrial purification by reducing the purification time.

PATENT

https://patents.google.com/patent/CN102060902A/en

chenodeoxycholic acid (3 α, 7 α – dihydroxy _5 β – cholestane-24-oic acid) Chenodeoxycholic Ac id (referred to as CDCA), clinically used to correct dissolving cholesterol calculi and bile saturation drugs, the main function is to reduce the cholesterol in the bile saturation, large doses can inhibit the synthesis of cholesterol CDCA and increasing bile gallstone patients cholesterol level in a non-saturated, thereby preventing the formation of cholesterol gallstones of cholesterol and promote stone dissolve and fall off. It also has significant anti-asthmatic, anti-inflammatory, antitussive and expectorant effects.

[0003] Synthesis of chenodeoxycholic acid or ursodeoxycholic acid (3 α, 7β_ -5β_ dihydroxy-cholestane-24-oic acid, ursodeoxycholic Acid, referred UDCA), a key intermediate. Ursodeoxycholic acid is the main active ingredient of precious Chinese medicine bear bile, used in a variety of clinical hepatobiliary disease and dyspepsia. Currently we bear bile resources are scarce, mainly used synthetic chemical ursodeoxycholic acid as a clinical treatment. Therefore, the preparation of chenodeoxycholic acid is also important for the preparation of ursodeoxycholic acid.

[0004] CDCA mainly come from poultry or livestock bile extraction. Traditional extraction process complicated operation, low yield, (pharmaceutical industry, 1987,18 (9), 416; Chinese Journal of Biochemical Pharmaceutics, 1996,17 (1), 17; Applied Technology, 1998, (4), 9; CN1850846A ) can not meet the needs of modern industry. Chemical synthesis of chenodeoxycholic acid have also been reported (Japanese Journal of Chemistry 1955,76 (3), 297 -J Org Chem 1982,47 (2): 2331; Journal of Biochemical Pharmaceutics 1987,1,6 -, Tap Chi Duoc ^ oc2004 , 44 (1), 11; CN1869043A), but lower yield widespread pollution major problem, especially in the oxidation reaction is often used to expensive, and polluting agents.Therefore, to reduce pollution, reduce environmental hazards, streamline operations, improve yield, reduce costs, important for the synthesis of chenodeoxycholic acid.

 Preparation of chenodeoxycholic acid.

[0007]

Figure CN102060902AD00041
Preparation of chenodeoxycholic acid (5): 12- oxo take chenodeoxycholic acid 0. 9g, 15ml ethylene glycol was added solid sodium hydroxide and 1. 5g, 15ml hydrated corpus (80%) , 120 ° C reflux for 2 hours, change return device is a distillation apparatus, was gradually warmed evaporated amount hydrated corpus, continue to heat up to 150 ° C, continuing reflux for 4h, cooled to room temperature, poured into water, adjusted with HCl of PH3, white precipitated, was filtered cake was washed with water, and dried to give crude chenodeoxycholic acid 0. 92g, recrystallized from methanol to give 0. 86g, 99% yield. .. 1HnMR (CD3SOCD3) S: 0.60 (s, 3H, 18- CH3), 0 90 (s, 3H, 19_CH3), 0.95 (d, 3H, 21-CH3), 3 47 (s, IH, 3 β – H), 3. 96 (s, 1H, 7 β -H), 11. 94 (s, 1H, C00H).
PATENT

Cholic acid esters prepared by (1) Weigh 50 g of cholic acid, dissolved in 150 ml of anhydrous methanol was added 5 ml of concentrated hydrochloric acid was refluxed for 30 minutes, cooled slowly into the freezer, the available capacity methyl cholate It was 95%.

(2) hydroxy -12α- diacetyl – Preparation of methyl cholate methyl cholate weighed 50 g, was dissolved in 100 ml of pyridine was purified, dissolved completely, 100 ml of acetic anhydride was stirred at room temperature for 3 to 4 hours, poured into 500 ml of water, a white precipitate in the refrigerator, filtered the next day, diacetyl -12α- available hydroxy – methyl cholate, yield 40%.

(3) 3α, 7α–diacetoxy-12-oxo – Preparation of methyl cholanic acid prepared above was weighed 25 g of crude product, dissolved in 250 ml of acetone, filtered to remove insolubles, the stirring conditions , the Jones reagent was slowly added, at room temperature for 30 minutes, filtered, water was added to the filtrate precipitated white precipitate was filtered available 3α, 7α–diacetoxy-12-oxo – methyl-cholanic acid. The yield was 100%.

(4) 12- oxo – Preparation of chenodeoxycholic acid in ethanol 10% – sodium hydroxide solution and saponified for 1 hour at room temperature, the solution was acidified, poured into water to give 12- oxo – chenodeoxycholic acid , 100% yield.Recrystallized in absolute ethanol.

Preparation of chenodeoxycholic acid (5) was weighed 12- oxo – chenodeoxycholic acid, 20 grams, was added 300 ml of ethylene glycol and 30 g of solid sodium hydroxide and 300 ml of hydrazine hydrate (85%), 100 ℃ refluxed for 2 hours, warming gradually raised to 130. ℃, generated by hydrazine hydrate was distilled off, continue to heat up to 185 ~ 190 ℃, continued reflux for 4 hours, cooled to a lower temperature, poured into water and heat, PH adjusted with hydrochloric acid (20%) 3, a white precipitate was filtered cake was washed with water to give chenodeoxycholic acid.

(6) Purification of chenodeoxycholic acid obtained weighed amount of chenodeoxycholic acid, dissolved with a small amount of ethanol, was impregnated on a silica gel column petroleum ether, liquid flow linear velocity by column chromatography 1 ~ 5cm / control points, with petroleum ether: acetone = 2, begins to elute, detected by TLC chromatography therebetween, Junichi appearance of spots to be chenodeoxycholic acid appears to start collecting the eluate until no Chenodeoxy acid spots, distillation under reduced pressure and dried to give pure higher chenodeoxycholic acid.

PATENTS

Publication numberPriority datePublication dateAssigneeTitle
WO2007069814A1 *2005-12-122007-06-21Daewoong Pharmaceutical Co., Ltd.Purification process for chenodeoxycholic acid
WO2007078039A1 *2005-12-302007-07-12Daewoong Pharmaceutical Co., Ltd.Purification process for chenodeoxycholic acid
CN100484952C2005-12-132009-05-06山东博尔德生物科技有限公司Method for producing high-purity chenodeoxy cholic acid from poultry and livestock bile
CN102060902A *2011-01-212011-05-18郑州大学Chenodeoxycholic acid synthesis method
CN102286051A *2011-08-152011-12-21上海华震科技有限公司A method for separating chenodeoxycholic acid and ursodeoxycholic acid
CN102690856A *2012-05-302012-09-26绵阳劲柏生物科技有限责任公司Process using microbial solution to prepare free bile acid
CN102703556A *2012-05-302012-10-03绵阳劲柏生物科技有限责任公司Method for separating chenodeoxycholic acid from duck bile by using macroporous resin
CN101830956B2008-11-192012-11-21毕小升Preparation method for separating and purifying chenodeoxycholic acid in porcine bile paste or leftovers
CN102827234A *2012-08-302012-12-19苏州天绿生物制药有限公司Method for separating and purifying chenodeoxycholic acid from duck gall
CN103360454A *2013-05-062013-10-23广西大学Method for separating and purifying chenodeoxycholic acid from goose bile
US3919266A1972-09-211975-11-11Intellectual Property Dev CorpProduction of bile acids
FR2429224A1 *1978-06-191980-01-18Canada Packers LtdChenodeoxycholic acid recovery from porcine bile – useful for dissolving gall stones in vivo
JPS60181096A *1984-02-281985-09-14Tokyo Tanabe Co LtdPurification of bile acid
EP0386538A21989-03-061990-09-12ERREGIERRE INDUSTRIA CHIMICA SpaProcess for preparing high purity 3-alpha-7-beta-dihydroxycholanic acid
JPH03227998A *1990-02-021991-10-08Showa Denko KkMethod for purifying chenodeoxycholic acid
CN1528779A *2003-09-292004-09-15华东理工大学Method for preparing cheodexycholic acid
Family To Family Citations
GB1450939A *1973-12-191976-09-29Intellectual Property
US4186143A *1977-06-201980-01-29Canada Packers LimitedChenodeoxycholic acid recovery process
KR100658512B1 *2005-12-302006-12-11주식회사 대웅제약Purification process for chenodeoxycholic acid

References

  1. Jump up to:a b c Russell DW (2003). “The enzymes, regulation, and genetics of bile acid synthesis”Annu. Rev. Biochem72: 137–74. doi:10.1146/annurev.biochem.72.121801.161712PMID 12543708.
  2. Jump up^ Carey MC (December 1975). “Editorial: Cheno and urso: what the goose and the bear have in common”. N. Engl. J. Med293 (24): 1255–7. doi:10.1056/NEJM197512112932412PMID 1186807.
  3. Jump up^ Parks DJ, Blanchard SG, Bledsoe RK, et al. (May 1999). “Bile acids: natural ligands for an orphan nuclear receptor”Science284 (5418): 1365–8. doi:10.1126/science.284.5418.1365PMID 10334993.
  4. Jump up^ Thistle JL, Hofmann AF (September 1973). “Efficacy and specificity of chenodeoxycholic acid therapy for dissolving gallstones”N. Engl. J. Med289 (13): 655–9. doi:10.1056/NEJM197309272891303PMID 4580472.
  5. Jump up^ Berginer VM, Salen G, Shefer S (December 1984). “Long-term treatment of cerebrotendinous xanthomatosis with chenodeoxycholic acid”N. Engl. J. Med311 (26): 1649–52. doi:10.1056/NEJM198412273112601PMID 6504105.
  6. Jump up^ Giaconda. “Press release”. Retrieved 5 April 2014.
  7. Jump up^ Bazzoli F, Malavolti M, Petronelli A, Barbara L, Roda E (1983). “Treatment of constipation with chenodeoxycholic acid”. J. Int. Med. Res11 (2): 120–3. PMID 6852359.
  8. Jump up^ Rao AS, Wong BS, Camilleri M, et al. (November 2010). “Chenodeoxycholate in females with irritable bowel syndrome-constipation: a pharmacodynamic and pharmacogenetic analysis”Gastroenterology139 (5): 1549–58, 1558.e1. doi:10.1053/j.gastro.2010.07.052PMC 3189402Freely accessiblePMID 20691689.
  9. Jump up^ Ki Soo Kim, Hong-Seok Kim Molecular Tweezer Based on Chenodeoxycholic Acid:Synthesis, Anion Binding Properties. Bulletin of the Korean Society 1411-1413 2004 Article ArchivedSeptember 27, 2007, at the Wayback Machine.
Chenodeoxycholic acid
Skeletal formula of chenodeoxycholic acid
Ball-and-stick model of the chenodeoxycholic acid molecule
Names
IUPAC names
chenodiol
OR
3α,7α-dihydroxy-5β-cholanic acid
OR
5β-cholanic acid-3α,7α-diol
OR
(R)-((3R,5S,7R,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethylhexadecahydro-1H-cyclopenta[a]phenanthren-17-yl)pentanoic acid
Identifiers
3D model (JSmol)
ChEBI
ChEMBL
ChemSpider
DrugBank
ECHA InfoCard 100.006.803
EC Number 207-481-8
KEGG
PubChem CID
UNII
Properties
C24H40O4
Molar mass 392.57 g/mol
Melting point 165 to 167 °C (329 to 333 °F; 438 to 440 K)
Pharmacology
A05AA01 (WHO)
License data
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
No verify (what is YesNo ?)
Infobox references

////////////////////Chenodeoxycholic acid,  ケノデオキシコール酸 , orphan designation

[H][C@@]1(CC[C@@]2([H])[C@]3([H])[C@H](O)C[C@]4([H])C[C@H](O)CC[C@]4(C)[C@@]3([H])CC[C@]12C)[C@H](C)CCC(O)=O


Gadobenate Dimeglumine

$
0
0

Gadobenate dimeglumine.png

2D chemical structure of 127000-20-8

ChemSpider 2D Image | UNII:3Q6PPC19PO | C36H62GdN5O21

Gadobenate Dimeglumine

Gadobenate Dimeglumine

Molecular Formula: C36H62GdN5O21
Molecular Weight: 1058.156 g/mol

cas 113662-23-0 FREEFORM

INGREDIENT UNII CAS
Gadobenate dimeglumine 3Q6PPC19PO 127000-20-8

Used in MR imaging of liver.

UNII-15G12L5X8K

  1. 3,6,9-triaza-12-oxa-3,6,9-tricarboxymethylene-10-carboxy-13-phenyltridecanoic acid, gadolinium
  2. B 19036
  3. B-19036
  4. gadobenate dimeglumine
  5. gadobenic acid
  6. gadobenic acid, dimeglumine salt
  7. gadolinium-benzyloxypropionyl tetraacetate
  8. gadolinium-BOPTA-Dimeg
  9. Gd(BOPTA)2
  10. Gd-BOPTA
  11. Multihance (TN)
  12. E-7155

2-[2-[carboxylatomethyl-[2-[carboxylatomethyl(carboxymethyl)amino]ethyl]amino]ethyl-(carboxymethyl)amino]-3-phenylmethoxypropanoate;gadolinium(3+);(2R,3R,4R,5S)-6-(methylamino)hexane-1,2,3,4,5-pentol

gadolinium(3+) ion bis((2R,3R,4R,5S)-6-(methylamino)hexane-1,2,3,4,5-pentol) 8-(carboxylatomethyl)-5,11-bis(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecane-4,13-dioate

Gadolinium hydrogen 4-carboxylato-5,8,11-tris(carboxylatomethyl)-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oate – 1-deoxy-1-(methylamino)-D-glucitol (1:2:1:2)

4-Carboxylato-5,8,11-tris(carboxylatométhyl)-1-phényl-2-oxa-5,8,11-triazatridécan-13-oate de gadolinium et de hydrogène – 1-désoxy-1-(méthylamino)-D-glucitol (1:1:2:2)

Gadolinate(2-), (4-carboxy-5,8,11-tris(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)-N5,N8,N11,O4,O5,O8,O11,O13)-, dihydrogen, comp. with 1-deoxy-1-(methylamino)-D-glucitol (1:2)
gadolinium(3+) bis(meglumine) 8-(carboxylatomethyl)-5,11-bis(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecane-4,13-dioate
gadolinium(3+) ion bis((2R,3R,4R,5S)-6-(methylamino)hexane-1,2,3,4,5-pentol) 8-(carboxylatomethyl)-5,11-bis(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecane-4,13-dioate
gadolinium(3+) ion bis(N-methyl-D(-)-glucamine) 8-(carboxylatomethyl)-5,11-bis(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecane-4,13-dioate
GADOLINIUM(3+) ION DIHYDROGEN BIS(N-METHYL-D(-)-GLUCAMINE) 5,8,11-TRIS(CARBOXYLATOMETHYL)-1-PHENYL-2-OXA-5,8,11-TRIAZATRIDECANE-4,13-DIOATE
  • D-Glucitol, 1-deoxy-1-(methylamino)-, [4-carboxy-5,8,11-tris(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)-N5,N8,N11,O4,O5,O8,O11,O13]gadolinate(2-) (2:1)
  • 2-Oxa-5,8,11-triazatridecan-13-oic acid, 4-carboxy-5,8,11-tris(carboxymethyl)-1-phenyl-, gadolinium complex
  • Gadolinate(2-), [4-(carboxy-κO)-5,8,11-tris[(carboxy-κO)methyl]-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)-κN5,κN8,κN11,κO13]-, dihydrogen, compd. with 1-deoxy-1-(methylamino)-D-glucitol (1:2) (9CI)
  • Gadolinate(2-), [4-carboxy-5,8,11-tris(carboxymethyl)-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)-N5,N8,N11,O4,O5,O8,O11,O13]-, dihydrogen, compd. with 1-deoxy-1-(methylamino)-D-glucitol (1:2)
  • B 19036/7

Hygroscopic powder

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 794

Melting Point

124 deg C

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 794

Spectral Properties

Specific optical rotation: -26.9 deg at 20 deg C/365 deg C (c = 1.45 in water)

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 794

Absorption maximum: 257.8 nm (epsilon 203)

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 794
Title: Gadobenate Dimeglumine
CAS Registry Number: 127000-20-8
CAS Name: 1-Deoxy-1-(methylamino)-D-glucitol [4-(carboxy-kO)-5,8,11-tris[(carboxy-kO)methyl]-1-phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)-kN5,kN8,kN11,kO13]gadolinate(2-) (2:1)
Additional Names: gadolinium benzyloxypropionictetraacetate dimeglumine; Gd-BOPTA/Dimeg
Manufacturers’ Codes: B-19036/7
Trademarks: MultiHance (Bracco)
Molecular Formula: C36H62GdN5O21
Molecular Weight: 1058.15
Percent Composition: C 40.86%, H 5.91%, Gd 14.86%, N 6.62%, O 31.75%
Literature References: Intravascular paramagnetic MRI contrast agent.
Prepn: E. Felder et al.,EP230893eidem,US4916246(1987, 1990 both to Bracco); F. Ungerri et al.,Inorg. Chem.34, 633 (1995). HPLC determn in biological samples: T. Arbughi et al.,J. Chromatogr. B713, 415 (1998). Physicochemical properties: C. de Haen et al.,J. Comput. Assist. Tomogr.23, Suppl. 1, S161 (1999). Pharmacology: P. Tirone et al.,ibid. S195. Pharmacokinetics: V. Lorusso et al.,ibid. S181. Toxicology: A. Morisetti et al.,ibid. S207. Clinical study in MRI of liver lesions: J. Petersein et al.Radiology215, 727 (2000). Review of clinical studies: B. Hamm et al.,J. Comput. Assist. Tomogr.23, Suppl. 1, S53-S60 (1999).
Properties: Hygroscopic powder. mp 124°. Freely sol in water, sol in methanol. Practically insol in n-butanol, n-octanol, chloroform. Abs max 257.8 nm (e 203). [a]36520 -26.9° (c = 1.45 in water). Prepd as 0.5M soln, osmolality (37°) 1.97 mol/kg. d20 1.22. Viscosity (mPa.s): 9.2 (20°), 5.3 (37°). LD50 i.v. in mice (mmol/kg): 5.7 (at 1 mL/min), 7.9 (at 0.2 mL/min); LD50 i.v. in rats (mmol/kg): 6.6 (at 6 mL/min), 9.2 (at 1 mL/min) (Morisetti).
Melting point: mp 124°
Optical Rotation: [a]36520 -26.9° (c = 1.45 in water)
Absorption maximum: Abs max 257.8 nm (e 203)
Density: d20 1.22
Toxicity data: LD50 i.v. in mice (mmol/kg): 5.7 (at 1 mL/min), 7.9 (at 0.2 mL/min); LD50 i.v. in rats (mmol/kg): 6.6 (at 6 mL/min), 9.2 (at 1 mL/min) (Morisetti)
Therap-Cat: Diagnostic aid (MRI contrast agent).
Keywords: Diagnostic Aid (MRI Contrast Agent).

Launched – 1998 Bracco,

Imaging, magnetic resonance

MultiHance injection is supplied as a sterile, nonpyrogenic, clear, colorless to slightly yellow aqueous solution intended for intravenous use only. Each mL of MultiHance contains 529 mg gadobenate dimeglumine and water for injection. MultiHance contains no preservatives.

Gadobenate dimeglumine is a gadolinium-based, paramagnetic contrast agent that was launched by Bracco in 1998 for use in magnetic resonance imaging (MRI). The drug is administered as an injection, and is approved in the U.S. for use in imaging of the central nervous system in adults and for visualization of lesions, abnormalities in the blood brain barrier, or abnormal vascularity of the brain, spine and associated tissues. Commercialization took place in 2010. In 2012, the product was approved and launched in the U.S. as a contrast agent for magnetic resonance angiography (MRA) to evaluate adults with known or suspected renal or aorto-ilio-femoral occlusive vascular disease

Gadobenate dimeglumine is chemically designated as (4RS)-[4-carboxy-5,8,11-tris(carboxymethyl)-1phenyl-2-oxa-5,8,11-triazatridecan-13-oato(5-)] gadolinate(2-) dihydrogen compound with 1-deoxy-1(methylamino)-D-glucitol (1:2) with a molecular weight of 1058.2 and an empirical formula of C22H28GdN3O11 • 2C7H17NO5. The structural formula is as follows:

Image result for Gadobenate Dimeglumine SYNTHESIS

Prescription Drug Products

Prescription Drug Products: 1 of 2 (RX Drug Ingredient)
Drug Ingredient GADOBENATE DIMEGLUMINE
Proprietary Name MULTIHANCE MULTIPACK
Applicant BRACCO (Application Number: N021358)
Prescription Drug Products: 2 of 2 (RX Drug Ingredient)
Drug Ingredient GADOBENATE DIMEGLUMINE
Proprietary Name MULTIHANCE
Applicant BRACCO (Application Number: N021357)

PATENT

US4916246 Paramagnetic chelates useful for NMR imaging
1990-04-10

Gadolinium-Based, paramagnetic contrast agent launched by Bracco in 1998 for use in magnetic resonance imaging (MRI).

Gadobenate Dimeglumine is a gadolinium-based paramagnetic contrast agent. When placed in a magnetic field, gadobenate dimeglumine produces a large magnetic moment and so a large local magnetic field, which can enhance the relaxation rate of nearby protons; as a result, the signal intensity of tissue images observed with magnetic resonance imaging (MRI) may be enhanced. Because this agent is preferentially taken up by normal functioning hepatocytes, normal hepatic tissue is enhanced with MRI while tumor tissue is unenhanced. In addition, because gadobenate dimeglumine is excreted in the bile, it may be used to visualize the biliary system using MRI.

Image result for Gadobenate Dimeglumine SYNTHESIS

FDA

https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/021357s000_Multihance_Chemr.pdf

Gadobenate Dimeglumine is an MRI contrast agent used primarily for MR imaging of the liver. It can also be used for MRI of the heart, as well as and central nervous system in adults to visualize lesions with abnormal brain vascularity or abnormalities in the blood brain barrier, the brain, spine, or other associated tissues.

Gadobenate Dimeglumine is an MRI contrast agent used primarily for MR imaging of the liver. It can also be used for visualizing the CNS and heart. In contrast to conventional extracellular fluid contrast agents, gadobenate dimeglumine is characterized by a weak and transient binding capacity to serum proteins. This binding leads to an increased relaxivity of gadobenate dimeglumine and, consequently, to a considerably increased signal intensity over that of other agents.

The drug is administered as an injection, and is approved in the U.S. for use in imaging of the central nervous system in adults and for visualization of lesions, abnormalities in the blood brain barrier, or abnormal vascularity of the brain, spine and associated tissues

Gadobenic acid (INN, trade name MultiHance) is a complex of gadolinium with the ligand BOPTA. In the form of the methylglucaminesalt meglumine gadobenate (INNm) or gadobenate dimeglumine (USAN), it is used as a gadolinium-based MRI contrast medium.[1]

BOPTA is a derivative of DTPA in which one terminal carboxyl group, –C(O)OH is replaced by -C–O–CH2C6H5. Thus gadobenic acid is closely related to gadopentetic acid. BOPTA itself was first synthesized in 1995. [2] In the “gadobenate” ion gadolinium ion is 9-coordinate with BOPTA acting as an 8-coordinating ligand. The ninth position is occupied by a water molecule, which exchanges rapidly with water molecules in the immediate vicinity of the strongly paramagnetic complex, providing a mechanism for MRI contrast enhancement139La NMR studies on the diamagnetic La-BOPTA2− complex suggest that the Gd complex maintains in solution the same kind of coordination as found, by X-ray crystallography, in the solid state for Gd-BOPTA disodium salt.[2]

2D chemical structure of 113662-23-0

ChemSpider 2D Image | 6688 | C22H28GdN3O11

MW: 670.7469

Gadobenic Acid [INN:BAN]
113662-23-0

PATENT

https://encrypted.google.com/patents/US20090155181

  • Gadolinium-based contrast agents are commonly used to improve visibility of internal structures when a patient undergoes magnetic resonance imaging (MRI). These agents are typically administered intravenously immediately prior to imaging. Many contrast agents used in MRI cause toxicity in various areas of the body if they are not excreted rapidly by the kidney. These include for example, chelated organic gadolinium compounds which are not nephrotoxic in themselves, but which if retained in the body for extended periods of time release gadolinium ions which are toxic to various organs and cells of the body including skin, nerves, etc. The problems particularly occur in patients who are at risk for reduced kidney function. Serious diseases including nephrogenic systemic fibrosis (NSF) are among the consequences of this problem. (see, for example, Briguori et al., Catheter Cardiovasc. Intery (2006) 67(2): 175-80; Grobner et al., Kidney Int. (2007) 72(3): 260-4; Nortier et al., Nephrol. Dial. Transplant (2007) 22(11): 3097-101).
  • [0003]
    The FDA requested a boxed warning for contrast agents used to improve MRI images on May 23, 2007 stating that patients with severe kidney insufficiency who receive gadolinium-based agents are at risk for developing NSF, a debilitating and potentially fatal disease. In addition, patients just before or just after liver transplantation, or those with chronic liver disease, are also at risk for developing NSF if they are experiencing kidney insufficiency of any severity. The boxed warning is now included in each of the five gadolinium-based contrast agents currently approved for use in the United States. Thus, a need exists to reduce the toxicity that is caused by contrast agents in patients with risk factors for compromised renal function.

PATENT

https://patents.google.com/patent/CN104606686A/en

gadobenate dimeglumine according to the present invention is a pharmaceutical composition, a chemical reaction equation I and the preparation was prepared as follows:

Figure CN104606686AD00051

Example 1 were added to the vessel IOOmL 7. 7gBOPTA and 4. 2g thirty-two gadolinia weighed, followed by addition of 47mL water for injection, stirring and heated to 60 ° C. After incubation the reaction at this temperature for 1.5h, added the same amount in ten batches 5.77g meglumine. After each addition was complete meglumine, taking a small sample using a pH meter to monitor the reaction solution pH. If the sample pH <6. 9, the reaction was continued until the reaction solution was added next batch PH interposed between Meglumine 6.9 ~ 7.3. After the addition of meglumine, the reaction was continued heating and stirring 1.5 hours. Followed by addition of decolorizing charcoal 〇.16g pharmaceutically acceptable, holding the temperature, stirred for 1.5 hours. Finally hot filtration, the filtrate was collected, concentrated in vacuo to give a white solid 14. 2g.

[0022] Example 2 were added to the vessel IOOmL 7. 7gBOPTA weighed and 5. Ig trioxide followed by addition of 68mL of water for injection, stirring and heated to 65 ° C. After incubation the reaction I. 5h at that temperature, was added an equal amount of sub-batches twelve 6. 24g meglumine. After each addition was complete meglumine, taking a small sample using a pH meter to monitor the reaction solution pH. If the sample pH <6. 9, the reaction was continued until the reaction solution was added at a pH between batch Meglumine between 6.9 ~ 7.3.After the addition of meglumine, the reaction solution and heating was continued for 2 hours. Followed by addition of decolorizing charcoal 〇.23g pharmaceutically acceptable, holding the temperature, stirring for 2 hours. Finally hot filtration, the filtrate was collected, concentrated in vacuo to give a white solid 14. 8g.

[0023] Example 3 were added to the vessel IOOOmL 77gBOPTA weighed 42g and gadolinium oxide, followed by addition of 470mL injection water and heated with stirring to 60 ° C. After incubation the reaction at this temperature for 2h, the same amount was added in ten 58g batches meglumine. After each addition was complete meglumine, small sample, monitoring the reaction solution with a PH meter pH. If the sample pH <6. 9, the reaction was continued until the reaction solution was added next batch PH interposed between Meglumine 6.9 ~ 7.3. After the addition of meglumine, the reaction solution and heating was continued for 2 hours. Followed by addition of 2. 5g medicinal charcoal decolorization, holding the temperature, stirring for 2 hours. Finally hot filtration, the filtrate was collected, and concentrated to give a white solid 131g.

PATENT

https://patents.google.com/patent/CN104606686A/en

Magnetic resonance imaging (MRI) is a tomographic image, which is obtained using a magnetic resonance phenomenon of electromagnetic signals from the body, the body information and reconstructed. Currently in clinical use development speed is very fast, widely used in nerve, spinal cord, heart and great vessels, joint bone, soft tissue and pelvic enhanced prosecution, has a three-dimensional object to be measured non-destructive and can perform high-resolution imaging and so on.

[0003] paramagnetic contrast agent is a contrast agent suitable for diagnostic magnetic resonance imaging (MRI), and into the body tissue can shorten the imaging time of protons, thereby enhancing the image sharpness and contrast. The paramagnetic contrast agents include Gd-DTPA and gadobenate dimeglumine.

[0004] Patent No. US5733528 discloses a metal chelate Gd-DTPA is applied to a magnetic resonance imaging (MRI) imaging study based on human organs; CN100325733C Patent discloses a gadolinia and diethylene triamine pentaacetic acid (DTPA) complex , to give after separation of gadolinium diethylenetriamine pentaacetic acid (Gd-DTPA), Gd-DTPA method of re-chelated meglumine obtained.

[0005] gadobenate dimeglumine gadolinium DTPA derivatives, widely used as paramagnetic contrast agents for magnetic resonance imaging. Clinical research shows that traitor, compared to Gd-DTPA, Gd Tony dimeglumine showed obvious advantages in terms of allergy, side effects and efficacy and so on. Moreover, Gd-DTPA was prepared using the purified and then after intermediate isolation, reacted not only with the stepwise synthesis of certain other compounds prepared by reacting the starting material many steps, long reaction period, and intermediate separation and purification operation complicated and likely to cause loss of product increased production costs. Therefore, the development of a simple method of synthesis Gadobenate dimeglumine is of great significance.

Patent

WO 2007031390

WO 2011073236

WO 2000002847

CN 102603550

PATENT

IN 201203216

IN 2012MU03216

The last step is Coordination of a Metal 12064-62-9, Gadolinium(III) oxide, to Carbon and Heteroatom

FIRST REPORT

  • By Vittadini, Giorgio; Felder, Ernst; Musu, Carlo; Tirone, Piero
  • From Investigative Radiology (1990), 25(Suppl. 1), S59-S60.

PRODUCT PATENT

  • By Cavagna, Friedrich; Dapra, Massimo; De Haen, Christoph; Maggioni, Fabio; Vicinanza, Eleonora
  • From Ital. Appl. (1992), IT 91MI1422 A1

AND WO 2011073236

References

  1. Jump up^ Sweetman, Sean C., ed. (2009). “Contrast Media”. Martindale: The Complete Drug Reference (36th ed.). London: Pharmaceutical Press. p. 1478. ISBN 978-0-85369-840-1.
  2. Jump up to:a b Uggeri, F.; Aime, S., Anelli, P.L., Botta, M., Brocchetta, M., De Haën, C., Ermondi, G., Grandi, M., Paoli, P. (1995). “Novel contrast agents for magnetic resonance imaging. Synthesis and characterization of the ligand BOPTA and its Ln(III) complexes (Ln = Gd, La, Lu). X-ray structure of disodium (TPS-9-145337286-C-S)-[4-carboxy-5,8,11-tris(carboxymethyl)-1-phenyl-2-oxa-5,8, 11-triazatridecan-13-oato(5-)]gadolinate(2-) in a mixture with its enantiomer”. Inorg. Chem34 (3): 633–642. doi:10.1021/ic00107a017.
Gadobenic acid
Structure of Gadobenic acid.png
Clinical data
AHFS/Drugs.com International Drug Names
ATC code
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C22H28GdN3O11
Molar mass 667.72 g/mol
3D model (JSmol)

////////////////Gadobenate Dimeglumine, 113662-23-0, x ray contrast agent, b 1906, Gd(BOPTA)2, Gd-BOPTA, MultihanceE-7155

CNCC(C(C(C(CO)O)O)O)O.CNCC(C(C(C(CO)O)O)O)O.C1=CC=C(C=C1)COCC(C(=O)[O-])N(CCN(CCN(CC(=O)O)CC(=O)[O-])CC(=O)[O-])CC(=O)O.[Gd+3]

THELIATINIB

$
0
0

img str1

THELIATINIB

CAS: 1353644-70-8
Chemical Formula: C25H26N6O2

Molecular Weight: 442.523

HMPL-309; HMPL 309; HMPL309; Theliatinib.

  • Originator Hutchison MediPharma
  • Class Antineoplastics; Small molecules
  • Mechanism of Action Epidermal growth factor receptor antagonists

Highest Development Phases

  • Phase I Oesophageal cancer; Solid tumours

Most Recent Events

  • 29 Sep 2017 Efficacy and adverse events data from a phase I trial in Oesophageal cancer released by Hutchison Pharma
  • 13 Mar 2017 Phase-I clinical trials in Oesophageal cancer (First-line therapy) in China (PO) before March 2017 (Hutchison MediPharma pipeline, July 2017)
  • 02 Aug 2016 Hutchison MediPharma plans a phase Ib proof-of-concept trial for Oesophageal cancer, and Head and Neck cancer in China

Theliatinib, also known as HMPL-309, is a novel small molecule, epidermal growth factor receptor tyrosine kinase inhibitor with potential antineoplastic and anti-angiogenesis activities. In vitro studies suggest that Theliatinib is a potent EGFR kinase inhibitor with good kinase selectivity and in vivo data demonstrated broad spectrum anti-tumor activity via oral dosing in multiple xerographs such as A-431, Bcap-37 and Fadu.

PRODUCT PATENT

  • By Zhang, Weihan; Su, Wei-Guo; Yang, Haibin; Cui, Yumin; Ren, Yongxin; Yan, Xiaoqiang

WO2012000356 , covering quinazoline compounds as EGFR inhibitors

https://encrypted.google.com/patents/WO2012000356A1?cl=pt-PT&hl=en&output=html_text

Example 3:

(3aR,6aR)-N-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yl)-l-methyl-hexahydropyrrolo [3,4-b]pyrrole-5(lH)-carboxamide

[060] To a solution of Compound 3-a (40 g, 0.138 mol, prepared according to procedures disclosed in WO2010002845), pyridine (40 mL, 0.495 mol) and DMF (anhydrous, 22 mL) in anhydrous THF (500 mL), was added phenyl carbonochloridate 3-b (22 mL, 0.175 mol) dropwise at -10°C. The mixture was stirred at room temperature for 12 hours. The precipitates were filtered and then suspended in saturated NaHC03 solution (500 mL). The solid was filtered, washed with H20 and EtOAc, and dried in vacuum to give compound 3-c (46 g).

A mixture of compound 3-c (1 g, 2.44 mmol) and compound 3-d (369 mg, 2.92 mmol) in dioxane (30mL) was stirred at 70°C for 5 hours, and then cooled to the ambient temperature. The precipitates were filtered, washed with EtOAc, and dried in vacuum to give compound 3 (0.8 g). MS (m/e): 443.4 (M+l)+.

PATENT

https://patents.google.com/patent/WO2010002845A2/en

PATENT

US 9168253

https://patents.google.com/patent/US9168253

Example 3 (3aR,6aR)—N-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yl)-1-methyl-hexahydropyrrolo[3,4-b]pyrrole-5(1H)-carboxamide

Figure US09168253-20151027-C00004

To a solution of Compound 3-a (40 g, 0.138 mol, prepared according to procedures disclosed in WO2010002845), pyridine (40 mL, 0.495 mol) and DMF (anhydrous, 22 mL) in anhydrous THF (500 mL), was added phenyl carbonochloridate 3-b (22 mL, 0.175 mol) dropwise at −10° C. The mixture was stirred at room temperature for 12 hours. The precipitates were filtered and then suspended in saturated NaHCO3solution (500 mL). The solid was filtered, washed with H2O and EtOAc, and dried in vacuum to give compound 3-c (46 g). A mixture of compound 3-c (1 g, 2.44 mmol) and compound 3-d (369 mg, 2.92 mmol) in dioxane (30 mL) was stirred at 70° C. for 5 hours, and then cooled to the ambient temperature. The precipitates were filtered, washed with EtOAc, and dried in vacuum to give compound 3 (0.8 g). MS (m/e): 443.4 (M+1)+.

PATENT

THELIATINIB BY HUTCHISON

WO-2018099451

The present invention belongs to the field of pharmacy and provides a crystal form of a compound (3aR,6aR)-N-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yl)-1-methyl-hexahydropyrrolo[3,4-b]pyrrole-5(1H)-carboxamide, a pharmaceutical composition thereof, and a preparation method therefor and the use thereof.
(FR)La présente invention concerne le domaine de la pharmacie et fournit une forme cristalline d’un composé (3aR,6aR)-N-(4-(3-éthynylphénylamino)-7-méthoxyquinazolin-6-yl)-1-méthyl-hexahydropyrrolo[3,4-b]pyrrole-5(1H)-carboxamide, une composition pharmaceutique de celui-ci, et son procédé de préparation et son utilisation.

Novel crystalline forms of the compound presumed to be theliatinib , processes for their preparation and compositions comprising them are claimed. Also claimed is their use for treating lung cancer, colon cancer, breast cancer, ovary cancer, prostate cancer, stomach cancer, kidney cancer, liver cancer, brain cancer, esophageal cancer, bone cancer and leukemia.

Hutchison Medipharma is developing theliatinib, a small molecule EGFR tyrosine kinase and AKT cell proliferation pathway inhibitor, for treating cancer, including brain tumor, esophageal tumor and NSCLC; in September 2017, positive preliminary data were presented. Hutchison is also developing epitinib succinate , for treating cancer including glioblastoma.

Binding of epidermal growth factor (EGF) to epidermal growth factor receptor (EGFR) activates tyrosine kinase activity and triggers a response that leads to cell proliferation. Overexpression and/or overactivation of EGFR can lead to uncontrolled cell division, and uncontrolled cell division can be a cause of cancer. Therefore, compounds that inhibit the over-expression and/or over-activation of EGFR are candidates for treating tumors.
Relevant compounds of the present invention (3aR, 6aR)-N-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yl)-1-methyl-hexahydropyrrolo [3, 4-b]pyrrole-5(1H)-carboxamide, whose chemical structure is shown in Formula A, has the effect of effectively inhibiting overexpression and/or overactivation of EGFR. Therefore, it can be used for the treatment of diseases associated with overexpression and/or overactivation of EGFR, such as the treatment of cancer.
Before discovering the crystal form of a compound, it is difficult to predict (1) whether a particular compound exists in crystalline form; (2) how an unknown crystal form is made; (3) what the properties of the crystal form would be, such as stability , bioavailability and so on.
Since the properties of the solid depend on the structure and the nature of the compound itself, different solid forms of the compound often exhibit different physical and chemical properties. Differences in chemical properties can be measured, analyzed, and compared using a variety of analytical techniques that ultimately can be used to distinguish these different solid forms. Differences in physical properties, such as solubility and bioavailability, are also important in describing the solid form of the drug compound. Likewise, in the development of pharmaceutical compounds, such as compounds of Formula A, the new crystalline and amorphous forms of the pharmaceutical compounds are also important.

Patent CN102906086A discloses compound (3aR,6aR)-N-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yl)-1-methyl-hexahydropyrrolo[3 4-b]pyrrole-5(1H)-carboxamide and its preparation method.

Experimental part
 
The starting material of the compound of formula A used in the examples was prepared according to CN102906086A
PATENT

Example 3: (3aR, 6aR) -N- (4- (3- ethynyl-phenylamino) -7-methoxy-quinazolin-6-yl) -1-methyl-hexahydro-pyrrolo [3,4-b] pyrrol -5 (IH) – carboxamide

[0102]

Figure CN102906086AD00131

[0103] at -10 ° C, to (40g, 0. 138mol, was prepared in accordance with the operation disclosed in W02010002845) Compound 3-a, pyridine (40mL, O. 495mol) and DMF (anhydrous, 22mL) in dry solution (500 mL) in THF dropwise phenyl chloroformate 3-b (22mL, O. 175mol). The mixture was stirred at room temperature for 12h. The precipitate was filtered off, and then it was suspended in saturated NaHCO3 solution (500mL). The solid was filtered off, washed with H2O and EtOAc, and dried in vacuo to give compound 3_c (46g). Compound 3-c (lg, 2. 44mmol) and the compound 3_d (369mg, 2. 92mmol) in a mixture of two anger dioxane (30mL) was stirred at 70 ° C 5 h, then cooled to ambient temperature. The precipitate was filtered off, washed with EtOAc, and dried in vacuo to give compound 3 (O. 8g). MS (m / e): 443. 4 (M + 1) +.

Theliatinib (HMPL-309)

Theliatinib (HMPL-309) is a novel small molecule, epidermal growth factor receptor tyrosine kinase inhibitor with potential antineoplastic and anti-angiogenesis activities. Theliatinib is being developed as an oral formulation for the treatment of solid tumors like non-small cell lung cancer.

Theliatinib pre-clinical studies were conducted in China. In vitro studies suggest that Theliatinib is a potent EGFR kinase inhibitor with good kinase selectivity and in vivo data demonstrated broad spectrum anti-tumor activity via oral dosing in multiple xerographs such as A-431, Bcap-37 and Fadu. Non-clinical safety studies have indicated that Theliatinib is generally well tolerated in animals.

In November 2012, HMP initiated the first-in-human clinical trials of theliatinib.

Patent Citations (4)

Publication number Priority date Publication date  AssigneeTitle
CN101094840A *2004-12-292007-12-26韩美药品株式会社Quinazoline derivatives for inhibiting cancer cell growth and method for the preparation thereof
CN101619043A *2008-06-302010-01-06和记黄埔医药(上海)有限公司Quinazoline derivant and medical application thereof
WO2010002845A2 *2008-06-302010-01-07Hutchison Medipharma Enterprises LimitedQuinazoline derivatives
CN102311438A *2010-06-302012-01-11和记黄埔医药(上海)有限公司Quinazoline compound
CN106117182A *2016-06-202016-11-16中国药科大学Quinazoline-N-phenethyl tetrahydroisoquinoline compound and preparation method and application thereof

REFERENCES

1: Ren Y, Zheng J, Fan S, Wang L, Cheng M, Shi D, Zhang W, Tang R, Yu Y, Jiao L,
Ni J, Yang H, Cai H, Yin F, Chen Y, Zhou F, Zhang W, Qing W, Su W. Anti-tumor
efficacy of theliatinib in esophageal cancer patient-derived xenografts models
with epidermal growth factor receptor (EGFR) overexpression and gene
amplification. Oncotarget. 2017 Apr 19. doi: 10.18632/oncotarget.17243. [Epub
ahead of print] PubMed PMID: 28472779.

//////THELIATINIB, HMPL-309, HMPL 309, HMPL309, Phase I,  Oesophageal cancer,  Solid tumours

 O=C(N1C[C@]2([H])N(C)CC[C@]2([H])C1)NC3=CC4=C(NC5=CC=CC(C#C)=C5)N=CN=C4C=C3OC

Alatrofloxacin Mesylate

$
0
0

 

Alatrofloxacin.svg

Alatrofloxacin mesylate.png

Alatrofloxacin Mesylate

Chemical Names: Alatrofloxacin mesylate; UNII-2IXX802851; 146961-77-5; Alatrofloxacin mesylate [USAN]; 157605-25-9; 2IXX802851
Molecular Formula: C27H29F3N6O8S
Molecular Weight: 654.618 g/mol
CAS No. 146961-76-4 (Alatrofloxacin );
157605-25-9 (Alatrofloxacin Mesylate);
Chemical Name (1α, 5α, 6α)-L-alanyl-N-[3-[6-carboxy-8-(2,4-difluorophenyl)-3-fluoro-5,8-dihydro-5-oxo-1,8-naphthyridine-2-yl]-3-azabicyclo[3.1.0]hex-6-yl]-L-alaninamide, monomethanesulfonate

Research Code:CP-116517-27; CP-116517,    Trade Name:Trovan I.V.®          MOA:Quinolone antibiotic            Indication:Life- or limb-threatening infections caused by susceptible strains          Status:Withdrawn    Company:Pfizer (Originator)

Alatrofloxacin (Trovan IV) is a fluoroquinolone antibiotic developed by Pfizer, delivered as a mesylate salt.[1]

Trovafloxacin and alatrofloxacin were both withdrawn from the U.S. market in 2001

Alatrofloxacin mesylate was first approved by the U.S. Food and Drug Administration (FDA) on Dec 18, 1997. It was developed and marketed as Trovan I.V. ® by Pfizer in the US.

Alatrofloxacin mesylate is a fluoronaphthyridone related to the fluoroquinolones with in vitro activity against a wide range of gram-negative and gram-positive aerobic and anaerobic microorganisms. The bactericidal action of alatrofloxacin results from inhibition of DNA gyrase and topoisomerase IV. Trovan I.V.® is indicated for the treatment of patients initiating therapy in in-patient health care facilities (i.e., hospitals and long term nursing care facilities) with serious, life- or limb-threatening infections caused by susceptible strains of the designated microorganisms in the conditions listed below.

Trovan I.V.® is available as injection solution for intravenous use, containing 7.86 mg/ml of Alatrofloxacin mesylate. The recommended starting dose is 200 mg or 300 mg administered intravenously.

Alatrofloxacin mesylate was withdrawn from the U.S. market in 2001.

Image result for Alatrofloxacin mesylate

Alatrofloxacin mesilate

    • Synonyms:CP 116517, CP 116517-27
    • ATC:J01MA
  • Use:antibiotic, prodrug of trovafloxacin
  • Chemical name:l-Alanyl-N-[(1α,5α,6α)-3-[6-carboxy-8-(2,4-difluorophenyl)-3-fluoro-5,8-dihydro-5-oxo-1,8-naphthyridin-2-yl]-3-azabicyclo[3.1.0]hex-6-yl]-l-alaninamide monomethanesulfonate
  • Formula:C26H25F3N6O5 • CH4O3S
  • MW:654.62 g/mol
  • CAS-RN:146961-77-5

Derivatives

base

  • Formula:C26H25F3N6O5
  • MW:558.52 g/mol
  • CAS-RN:146961-76-4

Substance Classes

Synthesis Path

Substances Referenced in Synthesis Path

CAS-RN Formula Chemical Name CAS Index Name
27317-69-7 C11H20N2O5 Ntert-butoxycarbonyl-l-alanyl-l-alanine L-Alanine, N-[(1,1-dimethylethoxy)carbonyl]-L-alanyl-
186772-86-1 C33H37F3N6O7 N-[(1,1-dimethylethoxy)carbonyl]-l-alanyl-N-[(1α,5α,6α)-3-[8-(2,4-difluorophenyl)-6-(ethoxycarbonyl)-3-fluoro-5,8-dihydro-5-oxo-1,8-naphthyridin-2-yl]-3-azabicyclo[3.1.0]hex-6-yl]-l-alaninamide L-Alaninamide, N-[(1,1-dimethylethoxy)carbonyl]-L-alanyl-N-[(1α,5α,6α)-3-[8-(2,4-difluorophenyl)-6-(ethoxycarbonyl)-3-fluoro-5,8-dihydro-5-oxo-1,8-naphthyridin-2-yl]-3-azabicyclo[3.1.0]hex-6-yl]-
171176-56-0 C22H19F3N4O3 ethyl (1α,5α,6α)-7-(6-amino-3-azabicyclo[3.1.0]hex-3-yl)-1-(2,4-difluorophenyl)-6-fluoro-1,4-dihydro-4-oxo-1,8-naphthyridine-3-carboxylate 1,8-Naphthyridine-3-carboxylic acid, 7-(6-amino-3-azabicyclo[3.1.0]hex-3-yl)-1-(2,4-difluorophenyl)-6-fluoro-1,4-dihydro-4-oxo-, ethyl ester, (1α,5α,6α)-
134575-66-9 C27H27F3N4O5 ethyl (1α,5α,6α)-1-(2,4-difluorophenyl)-7-[6-[[(1,1-dimethylethoxy)carbonyl]amino]-3-azabicyclo[3.1.0]hex-3-yl]-6-fluoro-1,4-dihydro-4-oxo-1,8-naphthyridine-3-carboxylate 1,8-Naphthyridine-3-carboxylic acid, 1-(2,4-difluorophenyl)-7-[6-[[(1,1-dimethylethoxy)carbonyl]amino]-3-azabicyclo[3.1.0]hex-3-yl]-6-fluoro-1,4-dihydro-4-oxo-, ethyl ester, (1α,5α,6α)-
75-75-2 CH4O3S methanesulfonic acid Methanesulfonic acid

Trade Names

Country Trade Name Vendor Annotation
D TROVAN Pfizer wfm
F Turvel Pfizer wfm
GB Turvel Pfizer wfm
I Turvel Pfizer wfm
USA Trovan Pfizer wfm

(wfm = withdrawn from market)

Formulations

  • vial 200 mg/40 ml, 300 mg/60 ml (5 mg/ml) (as mesilate)

References

    • US 5 164 402 (Pfizer; 17.11.1992; appl. 4.2.1991; WO-prior. 16.8.1989).
    • US 5 229 396 (Pfizer; 20.7.1993; appl. 24.7.1992).
    • WO 9 700 268 (Pfizer; appl. 27.3.1996; USA-prior. 15.6.1995).
    • US 5 763 454 (Pfizer; 9.6.1998; appl. 21.5.1997; WO-prior. 6.6.1995).
  • polymorphs:

    • US 6 080 756 (Pfizer; 27.6.2000; appl. 30.1.1998; WO-prior. 5.7.1996).

References
“Center for Drug Evaluation and Research – Application Number: 020759/020760 – Chemistry Review(s)” (PDF). Food and Drug Administration. Retrieved 29 August 2014.

Alatrofloxacin
Alatrofloxacin.svg
Clinical data
AHFS/Drugs.com Micromedex Detailed Consumer Information
MedlinePlus a605016
Pregnancy
category
  • US: C (Risk not ruled out)
Routes of
administration
Intravenous
ATC code
  • none
Legal status
Legal status
  • Withdrawn
Pharmacokinetic data
Bioavailability N/A
Protein binding 76% (trovafloxacin)
Metabolism Quickly hydrolyzed to trovafloxacin
Elimination half-life 9 to 12 hours (trovafloxacin)
Excretion Fecal and renal(trovafloxacin)
Identifiers
CAS Number
ChemSpider
UNII
ChEMBL
Chemical and physical data
Formula C26H25F3N6O5
Molar mass 558.509 g/mol
3D model (JSmol)

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

RG 7604,Taselisib

$
0
0

Taselisib skeletal.svgChemSpider 2D Image | Taselisib | C24H28N8O2  Taselisib.png

  • Molecular FormulaC24H28N8O2
  • Average mass460.531 Da

RG7604,Taselisib

GDC-0032, GDC0032;GDC 0032, RO5537381

1282512-48-4 [RN]
1H-Pyrazole-1-acetamide, 4-[5,6-dihydro-2-[3-methyl-1-(1-methylethyl)-1H-1,2,4-triazol-5-yl]imidazo[1,2-d][1,4]benzoxazepin-9-yl]-α,α-dimethyl-
UNII:L08J2O299M
10.1021/jm4003632
2-(4-(2-(1-isopropyl-3-methyl-1H-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide
2-{3-[2-(1-Isopropyl-3-methyl-1H-1,2–4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide
POLYMORPHS almost A to Z, US9266903
Taselisib (GDC-0032) is an experimental cancer drug in development by Roche. Molecule is a complex heterocycle with no chiral centres, hazardous materials are used in synthesis, preparation of impurities is a challenge. Taselisib is in phase III with Roche , clinical trials for treatment of metastatic breast cancer and non-small cell lung cancer

Taselisib (GDC-0032) is an experimental cancer drug in development by Roche. It is a small molecule inhibitor targeting phosphoinositide 3-kinase subtype PIK3CA.[1]

Taselisib is in phase III with Roche , clinical trials for treatment of metastatic breast cancer and non-small cell lung cancer.[2]

Taselisib is a phosphatidylinositol 3-kinase (PI3Kalpha) inhibitor in phase III clinical studies at Roche for the treatment of postmenopausal women with histologically or cytologically confirmed locally advanced or metastatic estrogen-receptor positive (ER+) breast cancer.

Taselisib is an orally bioavailable inhibitor of the class I phosphatidylinositol 3-kinase (PI3K) alpha isoform (PIK3CA), with potential antineoplastic activity. Taselisib selectively inhibits PIK3CA and its mutant forms in the PI3K/Akt/mTOR pathway, which may result in tumor cell apoptosis and growth inhibition in PIK3CA-expressing tumor cells. By specifically targeting class I PI3K alpha, this agent may be more efficacious and less toxic than pan PI3K inhibitors. Dysregulation of the PI3K/Akt/mTOR pathway is frequently found in solid tumors and causes increased tumor cell growth, survival, and resistance to both chemotherapy and radiotherapy. PIK3CA, which encodes the p110-alpha catalytic subunit of the class I PI3K, is mutated in a variety of cancer cell types and plays a key role in cancer cell growth and invasion.

str1

PRODUCT PATENT

WO 2011036280

Inventors Nicole BlaquiereSteven DoDanette DudleyAdrian J. FolkesRobert HealdTimothy HeffronMark JonesAleksandr KolesnikovChudi NdubakuAlan G. OliveroStephen PriceSteven StabenLan WangLess «
Applicant F. Hoffmann-La Roche Ag

https://encrypted.google.com/patents/WO2011036280A1?cl=en

Discovery of 2-(3-(2-(1-Isopropyl-3-methyl-1H-1,2-4-triazol-5-yl)-5,6-dihydrobenzo(f)imidazo(1,2-d)(1,4)oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide (GDC-0032): A -sparing phosphoinositide 3-kinase inhibitor with high unbound exposure and robust in vivo antitumor activity
J Med Chem 2013, 56(11): 4597

Condensation of 4-bromo-2-hydroxybenzaldehyde  with glyoxal  in the presence of NH3 in MeOH gives 5-bromo-2-(1H-imidazol-2-yl)phenol

Which upon annulation with 1,2-dibromoethane  in the presence of Cs2CO3 in DMF at 90 °C yields 9-bromo-5,6-dihydroimidazo[1,2-d][1,4]benzoxazepine .

Iodination of oxazepine  with NIS in DMF provides 9-bromo-2,3-diiodo-5,6-dihydroimidazo[1,2-d][1,4]benzoxazepine,

Which upon mono-deiodination by means of EtMgBr in THF at -15 °C affords 9-bromo-2-iodo-5,6-dihydroimidazo[1,2-d][1,4]benzoxazepine .

Amidation of iodide  with CO in the presence of PdCl2(PPh3)2 and HMDS in DMF at 70 °C produces the intermediate,

Which upon reaction with N,N-dimethylacetamide dimethyl acetal  in the presence of DME at 65 °C furnishes intermediate . Intramolecular cyclization of this compound with isopropylamine hydrochloride  in AcOH generates triazole derivative,

Which upon Suzuki coupling with dioxaborolane derivative in the presence of Pd(PPh3)4 and KOAc in CH3CN/H2O at 120 °C yields the target compound Taselisib.

Genentech BioOncology® logo

Taselisib has been used in trials studying the treatment and basic science of LYMPHOMA, Breast Cancer, Ovarian Cancer, Solid Neoplasm, and HER2/Neu Negative, among others.

Solubility (25°C)

In vitro DMSO 70 mg/mL warmed (151.99 mM)
Water Insoluble
Ethanol Insoluble warmed

Biological Activity

Description Taselisib (GDC 0032) is a potent, next-generation β isoform-sparing PI3K inhibitor targeting PI3Kα/δ/γ with Ki of 0.29 nM/0.12 nM/0.97nM, >10 fold selective over PI3Kβ.
Features A beta isoform-sparing PI3K inhibitor.
Targets
PI3Kδ [1]
(Cell-free assay)
PI3Kα [1]
(Cell-free assay)
PI3Kγ [1]
(Cell-free assay)
PI3Kβ [1]
(Cell-free assay)
C2β [1]
(Cell-free assay)
View More
0.12 nM(Ki) 0.29 nM(Ki) 0.97 nM(Ki) 9.1 nM(Ki) 292 nM
In vitro GDC-0032 is an orally bioavailable, potent, and selective inhibitor of Class I PI3Kα, δ, and γ isoforms, with 30 fold less inhibition of the PI3K β isoform relative to the PI3Kα isoform. Preclinical data show that GDC-0032 has increased activity against PI3Kα isoform (PIK3CA) mutant and HER2-amplified cancer cell lines. GDC-0032 inhibits MCF7-neo/HER2 cells proliferation with IC50 of 2.5 nM. [1]
Cell Data
Cell Lines Assay Type Concentration Incubation Time Formulation Activity Description PMID
human MOLM16 cells Proliferation assay 72 h Antiproliferative activity against human MOLM16 cells after 72 hrs by Cell Titer-Blue assay 22727640
In vivo GDC-0032 pharmacokinetics is approximately dose proportional and time independent with a mean t1/2 of 40 hours. The combination of GDC-0032 enhances activity of fulvestrant resulting in tumor regressions and tumor growth delay (91% tumor growth inhibition (TGI)). In addition, the combination of GDC-0032 with tamoxifen enhances the efficacy of tamoxifen in vivo (102%TGI for GDC-0032). [1]

PATENT

WO 2014140073

The invention relates to methods of making the PI3K inhibitor I (GDC-0032), named as 2-(4-(2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[l,2- d][l,4]oxazepin-9-yl)-lH-pyrazol-l-yl)-2-methylpropanamide, having the structure:

Figure imgf000003_0001

and stereoisomers, geometric isomers, tautomers, and pharmaceutically acceptable salts thereof.

Another aspect of the invention includes novel intermediates useful for preparing GDC- 0032 and having the structures:

Figure imgf000003_0002
Figure imgf000004_0001
Figure imgf000005_0001

The following Schemes 1-15 illustrate the chemical reactions, processes, methodology for the synthesis of GDC-0032, Formula I, and certain intermediates and reagents. Scheme 1:

Figure imgf000010_0001
Figure imgf000010_0002

Scheme 1 shows the synthesis of intermediate isopropylhydrazine hydrochloride 4 from Boc-hydrazine 1. Condensation of 1 with acetone and magnesium sulfate gave Boc-hydrazone, tert-butyl 2-(propan-2-ylidene)hydrazinecarboxylate 2 (Example 1). Palladium-catalyzed hydrogenation of 2 in acetic acid and methanol gave Boc-isopropyl-hydrazine 3 (Example 2) which was treated in situ with hydrogen chloride gas to give 4 (Example 3).

Alternatively, the double bond of 2 can be reduced with a hydride reagent such as sodium cyanoborohydride (Example 2).

Scheme 2:

Figure imgf000010_0003

Scheme 2 shows the synthesis of l-isopropyl-3-methyl-lH-l,2,4-triazole 7 from methyl acetimidate hydrochloride 5 and isopropylhydrazine hydrochloride 4. Reaction of 5 and 4 in triethylamine and methanol followed by cyclization of condensation product, N’- isopropylacetohydrazonamide 6 (Example 4) with triethyl orthoformate (triethoxymethane) gave 7 (Example 5). Alternatively, 4 and acetamidine can be reacted to give 6.

Or, 4 can be reacted with acetonitrile and an acid to form the corresponding salt of 6. Scheme 3:

Figure imgf000011_0001

0 K2C03, H20, MTBE w

Scheme 3 shows the synthesis of intermediate, 2-chloro-N-methoxy-N-methylacetamide 10. Reaction of 2-chloroacetyl chloride 8 and Ν,Ο-dimethylhydroxylamine hydrochloride 9 in aqueous potassium carbonate and methyl, tert-butyl ether (MTBE) gave 10 (Example 6).

Scheme 4:

Figure imgf000011_0002

Scheme 4 shows the synthesis of intermediate 4-bromo-2-fluorobenzimidamide hydrochloride 12 formed by reaction of 4-bromo-2-fluorobenzonitrile 11 with lithium hexamethyldisilazide (LiHMDS) in tetrahydrofuran (Example 7). Alternatively, 11 is treated with hydrogen chloride in an alcohol, such as ethanol, to form the imidate, ethyl 4-bromo-2- fluorobenzimidate hydrochloride, followed by ammonia in an alcohol, such as ethanol, to form 12 (Example 7).

Scheme 5:

Figure imgf000012_0001

Scheme 5 shows the synthesis of 5-(2-(4-bromo-2-fluorophenyl)-lH-imidazol-4-yl)-l- isopropyl-3 -methyl- lH-l,2,4-triazole V from l-isopropyl-3-methyl-lH-l,2,4-triazole 7.

Deprotonation of 7 with n-butyllithium and acylation with 2-chloro-N-methoxy-N- methylacetamide 10 gave intermediate 2-chloro-l-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5- yl)ethanone 13 (Example 8). Cyclization of 13 with 4-bromo-2-fluorobenzimidamide hydrochloride 12 and potassium hydrogen carbonate in water and THF (tetrahydrofuran) formed the imidazole V (Example 9).

Scheme 6:

Figure imgf000012_0002

Scheme 6 shows the synthesis of 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5- yl)-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III from V. Alkylation of the imidazole nitrogen of V with a 2-hydroxyethylation reagent such as, l,3-dioxolan-2-one, gave 2-(2-(4- bromo-2-fluorophenyl)-4-( 1 -isopropyl-3-methyl- 1 H- 1 ,2,4-triazol-5 -yl)- 1 H-imidazol- 1 – yl)ethanol 14 (Example 10). Cyclization of 14 with an aqueous basic reagent, such as methyltributylammonium chloride in aqueous potassium hydroxide, gave III, which can be cystallized from ethanol and water (Example 11). Scheme 7:

Figure imgf000013_0001

IV

Scheme 7 shows the synthesis of ethyl 2-(4-bromo-lH-pyrazol-l-yl)-2-methylpropanoate IV starting from 2-bromo-2-methylpropanoic acid 15. Alkylation of pyrazole with 15 gave 2- methyl-2-(lH-pyrazol-l-yl)propanoic acid 16 (Example 12). Esterification of 16 with sulfuric acid in ethanol gave ethyl 2-methyl-2-(lH-pyrazol-l-yl)propanoate 17 (Example 13).

Regiospecific bromination of 17 with N-bromosuccinimide (NBS) gave IV (Example 14). Alternatively, 16 was treated in situ with a brominating reagent such as l,3-dibromo-5,5- dimethylhydantoin (DBDMH) to give 2-(4-bromo-lH-pyrazol-l-yl)-2-methylpropanoic acid which was esterified to give IV, where R is ethyl. Other esters can also be prepared, such as methyl, iso-propyl, or any alkyl, benzyl or aryl ester.

Scheme 8:

Figure imgf000014_0001

Scheme 8 shows an alternative synthesis of ethyl 2-(4-bromo-lH-pyrazol-l-yl)-2- methylpropanoate IV starting from ethyl 2-bromo-2-methylpropanoate 18. Alkylation of pyrazole with 18 in the presence of a base such as sodium tert-butyloxide or cesium carbonate gave a mixture of ethyl 2-methyl-2-(lH-pyrazol-l-yl)propanoate 17 and ethyl 2-methyl-3-(lH- pyrazol-l-yl)propanoate 19. Bromination of the mixture with l,3-dibromo-5,5- dimethylimidazolidine-2,4-dione (DBDMH) gave a mixture containing IV, ethyl 3-(4-bromo- lH-pyrazol-l-yl)-2-methylpropanoate 20, and 4-bromo-lH-pyrazole 21 which was treated with a strong base under anhydrous conditions, such as lithium hexamethyldisilazide in tetrahydrofuran. Acidification with hydrochloric acid gave IV.

Scheme 9:

Pd(O) catalyst

Figure imgf000015_0001

KOAc, EtOH

Figure imgf000015_0002

Scheme 9 shows the synthesis of 2-(4-(2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)- 5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepin-9-yl)-lH-pyrazol-l-yl)-2-methylpropanamide, GDC-0032, 1 from ethyl 2-(4-bromo- 1 H-pyrazol- 1 -yl)-2-methylpropanoate IV (CAS Registry Number: 1040377-17-0, WO 2008/088881) and 9-bromo-2-(l-isopropyl-3-methyl-lH- 1,2,4- triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III (CAS Registry Number: 1282514-63-9, US 2012/0245144, US 8242104). Other esters besides ethyl can also be used which can be hydrolyzed with aqueous base, such as methyl, iso-propyl, or any alkyl, benzyl or aryl ester. In a one-pot Miyaura Borylation /Suzuki, Buchwald system, ethyl 2-(4-bromo-lH- pyrazol-l-yl)-2-methylpropanoate IV is reacted with 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(l,3,2- dioxaborolane), CAS Reg. No. 73183-34-3, also referred to as B2Pin2, and a palladium catalyst such as XPhos (2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl, CAS Reg. No. 564483- 18-7), with a salt such as potassium acetate, in a solvent such as ethanol, at about 75 °C to form the intermediate ethyl 2-methyl-2-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazol- l-yl)propanoate 22 (Example 15, CAS Registry Number: 1201657-32-0, US 8242104, US 8263633, WO 2009/150240).

Figure imgf000016_0001

XPhos ligandIntermediate 22 can be isolated or reacted in situ (one pot) with III to form 23.

A variety of low valent, Pd(II) and Pd(0) palladium catalysts can be used during the Suzuki coupling step to form 23 (Example 16) from 22 and III, including PdCl2(PPh3)2, Pd(t- Bu)3, PdCl2 dppf CH2C12, Pd(PPh3)4, Pd(OAc)/PPh3, Cl2Pd[(Pet3)]2, Pd(DIPHOS)2, Cl2Pd(Bipy), [PdCl(Ph2PCH2PPh2)]2, Cl2Pd[P(o-tol)3]2, Pd2(dba)3/P(o-tol)3, Pd2(dba)/P(furyl)3,

Cl2Pd[P(furyl)3]2, Cl2Pd(PMePh2)2, Cl2Pd[P(4-F-Ph)3]2, Cl2Pd[P(C6F6)3]2, Cl2Pd[P(2-COOH- Ph)(Ph)2]2, Cl2Pd[P(4-COOH-Ph)(Ph)2]2, and encapsulated catalysts Pd EnCat™ 30, Pd EnCat™ TPP30, and Pd(II)EnCat™ BINAP30 (US 2004/0254066).

The ester group of 23 is saponified with an aqueous basic reagent such as lithium hydroxide, to give 2-(4-(2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepin-9-yl)- IH-pyrazol- 1 -yl)-2-methylpropanoic acid II (Example 17). Intermediate 23 can be isolated or further reacted in situ with the aqueous basic reagent to form II. The carboxylic acid group of II is activated with an acyl activating reagent such as di(lH-imidazol-l-yl)methanone (carbonyl diimidazole, CDI) or Ν,Ν,Ν’,Ν’-tetramethyl- 0-(7-azabenzotriazol-l-yl)uronium hexafluorophosphate (HATU), and then reacted with an alcoholic ammonia reagent, such as ammonia dissolved in methanol, ethanol, or isopropanol, aqueous ammonium hydroxide, aqueous ammonium chloride, or ammonia dissolved in THF, to give I (Example 18).

A variety of solid adsorbent palladium scavengers can be used to remove palladium after the Suzuki coupling step to form compound I. Exemplary embodiments of palladium scavengers include FLORISIL®, SILIABOND®Thiol, and SILIABOND® Thiourea. Other palladium scavengers include silica gel, controlled-pore glass (TosoHaas), and derivatized low crosslinked polystyrene QUADRAPURE™ AEA, QUADRAPURE™ IMDAZ, QUADRAPURE™ MPA, QUADRAPURE™ TU (Reaxa Ltd., Sigma-Aldrich Chemical Co.).

Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003

Scheme 10 shows the synthesis of 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5- yl)-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III from 4-bromo-2-fluorobenzonitrile 11. Addition of hydroxylamine to the nitrile of 11 gave 4-bromo-2-fluoro-N-hydroxybenzimidamide 24. Michael addition of 24 to ethyl propiolate gave ethyl 3-(4-bromo-2- fluorobenzimidamidooxy)acrylate 25. Heating 25 in a high-boiling solvent such as toluene, xylene, ethylbenzene, or diphenyl oxide gave cyclized imidazole, ethyl 2-(4-bromo-2- fluorophenyl)-lH-imidazole-4-carboxylate 26, along with by-product pyrimidine, 2-(4-bromo-2- fluorophenyl)pyrimidin-4-ol. Alternatively, 25 can be cyclized to 26 with catalytic Lewis acids such as Cu(I) or Cu(II) salts. Alkylation of 26 with a 2-hydroxyethylation reagent, such as 1,3- dioxolan-2-one, in a base, such as N-methylimidazole or cesium carbonate, gave ethyl 2-(4- bromo-2-fluorophenyl)-l-(2-hydroxyethyl)-lH-imidazole-4-carboxylate 27. Ring-cyclization of 27 with an aqueous basic reagent, such as potassium hydroxide, lithium hydroxide, and methyl tributylammonium hydrochloride, gave 9-bromo-5,6-dihydrobenzo[f]imidazo[l,2- d][l,4]oxazepine-2-carboxylic acid 28. Addition of acetamidine to 28 with triphenylphosphine gave 9-bromo-N-(l-iminoethyl)-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2- carboxamide 29. Ring-cyclization of 29 with isopropylhydrazine hydrochloride 4 in acetic acid gave 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[l,2- d][l,4]oxazepine III.

Alternatively, 28 can be reacted with N’-isopropylacetohydrazonamide 6 to give III (Scheme 12).

Scheme 11 :

Figure imgf000018_0001

Scheme 11 shows the synthesis of 5-(2-(4-bromo-2-fluorophenyl)-lH-imidazol-4-yl)-l- isopropyl-3 -methyl- lH-l ,2,4-triazole V from 4-bromo-2-fluorobenzimidamide hydrochloride 12. 3-Chloro-2-oxopropanoic acid and 12 are reacted with base to give 2-(4-bromo-2-fluorophenyl)- lH-imidazole-4-carboxylic acid 30. Alternatively, 3-bromo-2-oxopropanoic acid can be reacted with 12 to give 30. Reaction of 30 with N’-isopropylacetohydrazonamide 6 and coupling reagent HBTU (N,N,N’,N’-tetramethyl-0-(lH-benzotriazol-l-yl)uronium hexafluorophosphate, O- (Benzotriazol-l-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate, CAS Ref. No. 94790- 37-1) in DMF gives intermediate, 2-(4-bromo-2-fluorophenyl)-N-(l-(2- isopropylhydrazinyl)ethylidene)-lH-imidazole-4-carboxamide 31 which need not be isolated and cyclizes upon heating to give V.

Alternatively, 5-(2-(4-chloro-2-fluorophenyl)-lH-imidazol-4-yl)-l-isopropyl-3-methyl- lH-l,2,4-triazole 44, the chloro version of V, can be prepared from 4-chloro-2-fluorobenzonitrile 38 (Scheme 15) Scheme 12:

Figure imgf000019_0001

Scheme 12 shows an alternative synthesis of 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4- triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III from 4-bromo-2- fluorobenzonitrile 11. Alkylation of 11 with tert-butyl 2-hydroxyethylcarbamate gives tert-butyl 2-(5-bromo-2-cyanophenoxy)ethylcarbamate 32. Cyclization of 32 under acidic conditions, such as hydrochloric acid in ethanol, gives 8-bromo-3,4-dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33. It will be noted that 33 has an alternative tautomeric form where the double bond is inside the oxazepine ring. Formation of the imidazole ring occurs by reaction of 3-bromo-2- oxopropanoic acid (X = Br, R = OH), or other 3-halo-2-oxopropanoic acid or ester (R = alkyl), and 33 to give 9-bromo-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxylic acid 28. Coupling of 28 with N’-isopropylacetohydrazonamide 6 and a coupling reagent such as HBTU, HATU or CDI in DMF gives intermediate, 9-bromo-N-(l-(2-isopropylhydrazinyl)ethylidene)- 5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxamide 34, which need not be isolated and forms 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III upon heating.

Alternatively, N’-isopropylacetohydrazonamide 6 is used as monohydrochloride salt, which has to be set free under the reaction conditions with an appropriate base, such as K2CO3. Scheme 13:

Figure imgf000020_0001

Scheme 13 shows an alternative synthesis of 8-bromo-3,4-dihydrobenzo[f][l,4]oxazepin- 5(2H)-imine 33 from 4-bromo-2-fluorobenzonitrile 11. Reaction of 11 with sodium methoxide in methanol gives methyl 4-bromo-2-fluorobenzimidate 35. Alkylation of 35 with 2- aminoethanol gives 4-bromo-2-fluoro-N-(2-hydroxyethyl)benzimidamide 36, followed by cyclization to 33.

Scheme 14:

Figure imgf000020_0002

37

11

Scheme 14 shows another alternative synthesis of 8-bromo-3,4- dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33 from 4-bromo-2-fluorobenzonitrile 11. Reaction of 11 with 2-aminoethanol and potassium tert-butoxide displaces fluorine to give 2-(2- aminoethoxy)-4-bromobenzonitrile hydrochloride 37. Ring closure of 37 with

trimethylaluminum gave 33. Alternatively, other trialkylaluminum reagents can be used, or magnesium alkoxide reagents such as magnesium ethoxide (magnesium bisethoxide, CAS Reg. No. 2414-98-4) to cyclize 37 to 33.

Figure imgf000021_0001
Figure imgf000021_0002

Scheme 15 shows the synthesis of 5-(2-(4-chloro-2-fluorophenyl)-lH-imidazol-4-yl)-l- isopropyl-3 -methyl- lH-l,2,4-triazole 44 from 4-chloro-2-fluorobenzonitrile 38. Addition of hydroxylamine to the nitrile of 38 gave 4-chloro-2-fluoro-N-hydroxybenzimidamide 39.

Michael addition of 39 to ethyl propiolate gave ethyl 3-(4-chloro-2- fluorobenzimidamidooxy)acrylate 40. Heating 40 in diphenyl oxide gave cyclized imidazole, ethyl 2-(4-chloro-2-fluorophenyl)-lH-imidazole-4-carboxylate 41. Saponification of the ester of 41 with aqueous sodium hydroxide in tetrahydrofuran gave 2-(4-chloro-2-fluorophenyl)-lH- imidazole-4-carboxylic acid 42. Reaction of 42 with N’-isopropylacetohydrazonamide 6 and coupling reagent HBTU in DMF gives intermediate, 2-(4-chloro-2-fluorophenyl)-N-(l-(2- isopropylhydrazinyl)ethylidene)-lH-imidazole-4-carboxamide 43 which cyclizes upon heating to give 44.

EXAMPLES

Example 1 tert-butyl 2-(propan-2-ylidene)hydrazinecarboxylate 2

To a solution of tert-butyl hydrazinecarboxylate 1 (CAS Reg. No. 870-46-2) (25.1 g, 0.190 mol) in acetone (185 mL) was added the magnesium sulfate (6 g) and 12 drops acetic acid (Wu et al (2012) Jour. Med. Chem. 55(6):2724-2736; WO 2007/056170; Zawadzki et al (2003) Polish Jour. Chem. 77(3):315-319). The mixture was heated to reflux for 2.5 h and cooled to rt and filtered. The filtrate was concentrated to give tert-butyl 2-(propan-2- ylidene)hydrazinecarboxylate 2 (CAS Reg. No. 16689-34-2) as an off-white solid (32 g, 98%) (used in the next step without further purification). LC-MS [M+H]+ = 172.9, RT = 2.11 min. 1H NMR 300 MHz (CDC13) d 7.35 (br s, 1H, NH), 2.04 (s, 3H), 1.82 (s, 3H), 1.54 (s, 9H); 13C NMR 300 MHz (CDC13) d 152.9, 149.7, 80.7, 28.1, 25.3, 15.9. Example 2 tert-butyl 2-isopropylhydrazinecarboxylate 3

tert-Butyl 2-(propan-2-ylidene)hydrazinecarboxylate 2 was reduced with palladium catalyst on carbon with hydrogen gas in acetic acid and methanol to give tert-butyl 2- isopropylhydrazinecarboxylate 3 (CAS Reg. No. 16689-35-3).

Alternatively, tert-Butyl 2-(propan-2-ylidene)hydrazinecarboxylate 2 (0.51 g, 3.0 mmol) was dissolved in 20 mL of THF, treated with NaB¾CN (0.19 g, 3.0 mmol) and a few mg of bromocresol green, followed by a solution of p-toluenesulfonic acid (0.57 g, 3.0 mmol) in 1.5 mL of THF which was added dropwise over approximately 1 h to maintain the reaction pH between 3.5-5.0. After stirring at room temperature for an additional hour, the solvent was removed by rotary evaporation, and the residue was partitioned between EtOAc (30 mL) and brine. The organic phase was extracted with sat. NaHCC>3, 20 mL and brine, evaporated to a residue and dissolved in 10 mL of ethanol. The ethanolic solution was treated with 3.6 mL of 1M NaOH solution (3.6 mmol) and left to stir at rt for 30 min. The solvent was removed by rotary evaporation and the residue was taken up into ethyl acetate and extracted with water. The organic layer was evaporated under reduced pressure and the residue was purified by column chromatography using 5 % MeOH in DCM as eluent to collect tert-butyl 2- isopropylhydrazinecarboxylate 3 (0.4 g, 77 % yield): mp = 47-49 °C; Rf = 0.44 (5 % MeOH in DCM); IH NMR 300 MHz (CDC13) d 6.03 (s, N-H, IH), 3.92 (s, N-H, IH), 3.14 (m, IH), 1.46 (s, 9H), 1.02 (d, 6H, J = 6 Hz); 13C NMR 300 MHz (CDC13) d 157.2, 80.8, 51.2, 28.7, 21.0.

Example 3 isopropylhydrazine hydrochloride 4

tert-butyl 2-isopropylhydrazinecarboxylate 3 was treated with hydrochloric acid to remove the Boc protecting group and give 4 (CAS Reg. No. 16726-41-3).

Example 4 N’-isopropylacetohydrazonamide 6

Methyl acetimidate hydrochloride 5 (CAS Reg. No. 14777-27-6), isopropylhydrazine hydrochloride 4, and triethylamine were reacted in methanol to give 6 (CAS Reg. No. 73479-06- 8).

Example 5 l-isopropyl-3 -methyl- lH-l,2,4-triazole 7

N’-isopropylacetohydrazonamide 6 was treated with triethylorthoformate in ethanol, followed by triethylamine and tetrahydrofuran to give 7 (CAS Reg. No. 1401305-30-3). Example 6 2-chloro-N-methoxy-N-methylacetamide 10

To a solution of 21.2 kg potassium carbonate K2CO3 (153.7 mol, 3.0 eq) in 30 L H20 was added, Ν,Ο-dimethylhydroxylamine 9 (CAS Reg. No. 1117-97-1) (5.0 kg, 51.3 mol, 1.0 eq) at 15-20 °C. The reaction was stirred at rt for 30min and 30 L methyl tert-butyl ether (TBME) was added. After stirred for 30min, the mixture was cooled to 5°C, and 11.6 kg of 2-Chloroacetyl chloride 8 (CAS Reg. No. 79-04-9 (102.7 mol, 2.0 eq) were added slowly. The reaction was stirred at rt overnight. Organics were separated from aqueous, and aqueous was extracted with TBME (30 L). The combined organics were washed with H20 (50 L), brine (50 L) and dried over Na2S04. Filtered and concentrated under vacuum afforded 5.1 kg of 2-chloro-N-methoxy- N-methylacetamide 10 (CAS Reg. No. 67442-07-3) as a white solid.

Example 7 4-bromo-2-fluorobenzimidamide hydrochloride 12

To 35.0 L of lithium hexamethyldisilazide LiHMDS (35.0 mol, 1.4 eq, 1.0 M in THF) under N2 was added a THF solution of 4-Bromo-2-fluorobenzonitrile 11 (CAS Reg. No. 105942- 08-3) (5.0 kg in 10 L THF) at 10 °C, the mixture was stirred at rt for 3h. Cooled to -20°C and 8.3 L of HCl-EtOH (6.6 M) were added. The mixture was stirred at -10 °C for additional lh, filtered. The wet cake was washed with EA (10 L) and H20 (6 L). Drying in vacuo yielded 5.8 kg 4- bromo-2-fluorobenzimidamide hydrochloride 12 (CAS Reg. No. 1187927-25-8) as an off-white solid.

Alternatively, to a 200-L vessel was charged 4-bromo-2-fluorobenzonitrile 11 (10 kg, 50.00 mol, 1.00 equiv) and ethanol (100 L) followed by purging 40 kg Hydrogen chloride (g) at – 10 °C with stirring (Scheme 4). The resulting solution was allowed to react for an additional 36 h at 10 °C. The reaction progress was monitored by TLC until 11 was consumed completely. The resulting mixture was concentrated under vacuum while maintaining the temperature below 60 °C. The volume was concentrated to 10-15 L before 60 L MTBE was added to precipitate the product. The precipitates were collected by filtration to afford in 12 k g of ethyl 4-bromo-2- fluorobenzimidate hydrochloride 12 as a white solid. (Yield: 85%). 1H NMR δ 7.88-7.67 (m), 4.89 (br s), 4.68 (q), 3.33 (m), 1.61 (t). MS M+l: 245.9, 248.0.

To a 200L vessel, was charged ethyl 4-bromo-2-fluorobenzimidate hydrochloride (12.5 k g, 44mol, 1.00 equiv, 99%) and ethanol (125 L) followed by purging NH3 (g) at -5 °C for 12 h. The resulting solution was stirred at 30 °C for an additional 24 h. The reaction progress was monitored by TLC until SM was consumed completely. The precipitates were filtered and the filtrate was concentrated under vacuum. The product was precipitated and collected by filtration to afford 6.1 kg (54.5%) of 4-bromo-2-fluorobenzamidine hydrochloride 12 as a white solid. 1H NMR δ 9.60 (br), 7.91-7.64 (m), 3.40 (s), 2.50 (m). MS M+l: 216.9, 219.9.

Example 8 2-chloro-l-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)ethanone 13

To a 10L four necked flask was charged l-Isopropyl-3-methyl-lH-l,2,4-triazole 7 (400 g) in THF (2.5 L). The resulting solution was cooled to -40 °C and 2.5 M n-butyllithium BuLi in n- hexanes (1.41 L) was added while keeping the internal temp, below -20°C. The resulting yellow suspension was stirred at -40°C for 1 hour before being transferred. To a 20L flask was charged 2-chloro-N-methoxy-N-methylacetamide 10 (485 g) in THF (4 L). The resulting solution was cooled to -40 °C at which point a white suspension was obtained, and to this was added the solution of lithiated triazole 7 keeping the internal temp, below -20°C. At this point a yellow orange solution was obtained which was stirred at – 30°C for lhour. Propionic acid (520 mL) was added keeping the internal temp, below -20°C. The resulting off-white to yellowish suspension was warmed to -5 °C over 30 minutes. Citric acid (200 g) in water (0.8 L) was added and after stirring for 5 minutes a clear biphasic mixture was obtained. At this point stirring was stopped and the bottom aqueous layer was removed. The organic phase was washed with 20w% K3PO4 solution (1 L), 20w% K2HP04 solution (2 L), and 20w% NaCl solution (1 L). The organics was reduced to ca 4L via distillation under vacuum to afford 2-chloro-l-(l-isopropyl-3- methyl-lH-l,2,4-triazol-5-yl)ethanone 13 as a dark amber liquid which was used “as is” in the next step.

Example 9 5-(2-(4-bromo-2-fluorophenyl)- lH-imidazol-4-yl)-l -isopropyl-3-methyl- lH-l,2,4-triazole V

To a 10 L four- neck flask were charged with THF (5.6 L), 4-bromo-2- fluorobenzimidamide hydrochloride 12 (567 g), KHCO3 (567 g) and water (1.15 L). The resulting white suspension was heated to 60°C over 2 hours. At this point a hazy solution was obtained to which was added a solution of 2-Chloro-l-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5- yl)ethanone 13 in THF (2 L). This solution was stirred at 60-65 °C for 24 hours. Then the aqueous bottom layer was removed. The organic layer was concentrated under vacuum. The residue was slurried in a mixture of MIBK (1.25 L) and toluene (0.7 L), and the precipitated product was filtered giving 552 g of 5-(2-(4-bromo-2-fluorophenyl)-lH-imidazol-4-yl)-l- isopropyl-3 -methyl- lH-l,2,4-triazole V (98.0% purity, 254 nm) as a brown solid Example 10 2-(2-(4-bromo-2-fluorophenyl)-4-(l-isopropyl-3-methyl-lH-l,2,4-triazol- 5-yl)- 1 H-imidazol- 1 -yl)ethanol 14

5-(2-(4-Bromo-2-fluorophenyl)-lH-imidazol-4-yl)-l-isopropyl-3-methyl-lH- 1,2,4- triazole V (2.75 kg, 7.55 mol) was added to a solution of 3-dioxolan-2-one (ethylene carbonate, 3.99 kg, 45.3 mol) inN-methylimidazole (12 L) at 50 °C. The suspension was heated at 80°C for 7 h until the reaction was judged complete by HPLC. The solution of 14 was cooled to 35 °C and used directly in the subsequent cyclization.

Example 11 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepine III

To a solution of 2-(2-(4-Bromo-2-fluorophenyl)-4-(l-isopropyl-3-methyl-lH-l,2,4- triazol-5-yl)-l H-imidazol- l-yl)ethanol (7.55 mmol) 14 inN-methylimidazole(12 L) at 35 °C was added methyl tributylammonium chloride (115 g, 0.453 mol), toluene (27.5 L) and 35% potassium hydroxide solution (10.6 kg, 25 mol in 22 L of water). The biphasic solution was stirred vigorously at 65 °C for 18 h when it was judged complete by HPLC. Stirring was stopped but heating was continued and the bottom aqueous layer was removed. Added isopropyl acetate (13.8 L) and the organic phase was washed twice with water (13.8 L and 27.5 L). The solvent was removed via vacuum distillation and after 30 L had been removed, isopropanol (67.6 L) was added. Vacuum distillation was resumed until an additional 30 L of solvent had been removed. Added additional isopropanol (28.8 L) and continued vacuum distillation until the volume was reduced by 42 L. Added isopropanol (4L) and the temperature was increased to >50 °C. Added water (28 L) such that the internal temperature was maintained above 50 °C, then heated to 75 °C to obtain a clear solution. The mixture was allowed to cool slowly and the product crystallized out of solution. The resulting suspension was cooled to 0 °C, held for 1 h then filtered and the cake was washed with water (5.5 L). The cake was dried at 45 °C under a nitrogen sweep to give III as a tan solid (3.30 kg, 71.6 wt %, 80.6% yield).

Example 12 2-methyl-2-(lH-pyrazol-l-yl)propanoic acid 16

2-Bromo-2-methylpropanoic acid 15 and pyrazole were reacted in triethylamine and 2- methyltetrahydrofuran to give 16.

Example 13 ethyl 2-methyl-2-(lH-pyrazol-l-yl)propanoate 17

2-Methyl-2-(lH-pyrazol-l-yl)propanoic acid 16 was treated with sulfuric acid in ethanol to give 17. Alternatively, pyrazole (10 g, 147 mmol, 1.0 eq.) was dissolved in DMF (500 ml) at room temperature (Scheme 8). 2-Bromoisobutyrate 18 (22 ml, 147 mmol, 1.0 eq.), cesium carbonate CS2CO3 (53 g, 162 mmol, 1.1 eq) and catalytic sodium iodide Nal (2.2 g, 15 mmol, 0.1, eq) were added to the mixture that was then heated to 60 °C for 24 hr. Reaction was followed by 1H NMR and pyrazole was not detected after 24 hr. The reaction mixture was quenched with a saturated solution of NaHCC>3 (200 ml) and ethyl acetate EtOAc (150 ml) was added and organics were separated from aqueous. Organics were dried over Na2S04, filtered and concentrated under vacuum to afford an oil which was purified by flash chromatography to give 17.

Example 14 Ethyl 2-(4-bromo-lH-pyrazol-l-yl)-2-methylpropanoate IV

Method A: Ethyl 2-methyl-2-(lH-pyrazol-l-yl)propanoate 17 was reacted with N- bromosuccinimide (NBS) in 2-methyltetrahydrofuran to give IV (CAS Reg. No. 1040377-17-0).

Method B: Ethyl 2-bromo-2-methylpropanoate 18 and pyrazole were reacted with sodium tert-butoxide in dimethylformamide (DMF) to give a mixture of ethyl 2-methyl-2-(lH- pyrazol-l-yl)propanoate 17 and ethyl 2-methyl-3-(lH-pyrazol-l-yl)propanoate 19 which was treated with l,3-dibromo-5,5-dimethylimidazolidine-2,4-dione to give a mixture of IV, ethyl 3- (4-bromo-lH-pyrazol-l-yl)-2-methylpropanoate 20, and 4-bromo-lH-pyrazole 21. The mixture was treated with a catalytic amount of lithium hexamethyldisilazide in tetrahydrofuran followed by acidification with hydrochloric acid to give IV.

Example 15 ethyl 2-methyl-2-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazol-l-yl)propanoate 22

To a 50 L glass reactor was charged ethyl 2-(4-bromo-lH-pyrazol-l-yl)-2- methylpropanoate IV (1.00 kg, 3.85 mol, 1.00 equiv), potassium acetate, KOAc (0.47 kg, 4.79 mol 1.25 equiv), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(l,3,2-dioxaborolane),

bis(pinacolato)diboron, B2Pin2 (1.22 kg, 4.79 mol, 1.25 equiv) and ethanol (10 L, 10 vol) and the mixture was stirred until a clear solution was obtained. The solution was vacuum/degassed 3x with nitrogen. To this mixture was charged XPhos ligand (0.023 kg, 0.048 mol, 1.0 mol ) and the Pd precatalyst (0.018 kg, 0.022 mol, 0.5 mol ) resulting in a homogeneous orange solution. The solution was vacuum/degassed once with nitrogen. The internal temperature of the reaction was set to 75 °C and the reaction was sampled every 30 min once the set temperature was reached and was monitored by LC (IPC method: XTerra MS Boronic). After 5 h, conversion to 22 (CAS Reg. No. 1201657-32-0) was almost complete, with 1.3% IV remaining. Example 16 ethyl 2-(4-(2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepin-9-yl)- IH-pyrazol- 1 -yl)-2-methylpropanoate 23

Ethyl 2-methyl-2-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazol-l- yl)propanoate 22 and 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III were reacted under Suzuki conditions with palladium catalyst, in isopropanol and aqueous phosphate buffer to give 23.

A 1M solution of K3PO4 (1.60 kg in 7.6 L of water, 7.54 mol, 2.00 equiv) was charged to the above reaction mixture from Example 15, followed by the addition of a solution of III in THF (1.33 kg in 5.0 L, 3.43 mol, 0.90 equiv) over 2 min. The reaction mixture was warmed to 75 °C (internal temperature) over 45 min and stirred for 13 h at 75 °C, then analyzed by HPLC (III not detected) showing the formation of 23.

Example 17 2-(4-(2-( 1 -isopropyl-3-methyl- 1 H- 1 ,2,4-triazol-5-yl)-5 ,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepin-9-yl)- IH-pyrazol- 1 -yl)-2-methylpropanoic acid II

Ethyl 2-(4-(2-(l -isopropyl-3 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-5 ,6- dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepin-9-yl)-lH-pyrazol-l-yl)-2-methylpropanoate 23 was treated with aqueous lithium hydroxide to give II.

The ester saponification reaction was initiated with the addition of 3.5 M aqueous LiOH (0.74 kg in 5.0 L, 17.64 mol, 5 equiv) to the reaction mixture from Example 16 and allowed to warm to 75 °C. The mixture was sampled every 30 min (IPC method: XTerra MS Boronic) and the saponification was complete after 4.5 h (with less than 0.3% 23 remaining). The reaction mixture was concentrated via distillation to approximately half volume (starting vol = 37 L; final vol = 19 L) to remove EtOH and THF, resulting in tan-brown slurry. Water (5 L, 5 vol) was charged to the mixture and then distilled (starting vol = 25 L; final vol = 21 L). The temperature was set at 60 °C (jacket control) and then charged with isopropyl acetate, IP Ac (4 L, 4 vol). The biphasic mixture was stirred a minimum of 5 min and then the layers allowed to separate for a minimum of 5 min. The bottom aqueous layer was removed into a clean carboy and the organics were collected into a second carboy. The extraction process was repeated a total of four times, until the organic layer was visibly clear. The aqueous mixture was transferred back to the reactor and then cooled to 15 °C. A 6 M solution of HC1 (6.4 L, 38.40 mol, 10 equiv) was charged slowly until a final pH = 1 was obtained. The heterogeneous mixture was then filtered. The resulting solids were washed twice with 5 L (2 x 5 vol) of water. The filter was then heated to 80 °C and the vacuum set to -10 Psi (with nitrogen bleed) and the solids were dried for 24 h (KF = 2.0 % H20) to give 1.54 kg (95% corrected yield) of II as a white solid; 98% wt, 97.3 % pure.

Example 18 2-(4-(2-( 1 -isopropyl-3-methyl- 1 H- 1 ,2,4-triazol-5-yl)-5 ,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepin-9-yl)- lH-pyrazol- 1 -yl)-2-methylpropanamide I (GDC-0032)

2-(4-(2-(l-Isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[l,2- d][l,4]oxazepin-9-yl)-lH-pyrazol-l-yl)-2-methylpropanoic acid II was treated with di(lH- imidazol-l-yl)methanone (carbonyldiimidazole, CDI) in tetrahydrofuran followed by methanolic ammonia to give crude I.

Solid II (1.44 kg, 3.12 mol, 1.00 equiv) was transferred into a 20 L bottle and then THF

(10 L, 7 vol) was charged. The slurry was transferred under reduced pressure into a second 50 L reactor and additional THF (5 L, 3 vol) was added for the rinse. The internal temperature of the slurry was set to 22 °C and Γ1 -carbonyldiimidazole, CDI (0.76 Kg, 5.12 mol, 1.50 equiv) was charged to the mixture and a clear solution was observed after 5 min. The reaction mixture was sampled every 30 min and analyzed by HPLC (IPC: XTerra MS Boronic method) which showed almost complete conversion to the acyl-imidazole intermediate and 1.2% remaining II after 30 min. An additional portion of CDI (0.07 kg, 0.15 mol, 0.14 equiv) was added, and the reaction mixture was stirred for 1 h and then analyzed by HPLC (IPC: XTerra MS Boronic method) which showed 0.8% remaining II.

Into a second 50-L reactor, was added NH3/MeOH (1.5 L, 10.5 mol, 3.37 equiv) and THF

(5 L, 3 vol). The acyl-imidazole intermediate was transferred to a second reactor under reduced pressure (transfer time -10 min). The internal temperature was then set to 45 °C and the volume of solvent was distilled down from 35 L to 12 L. Water (6 L, 4 vol) was then added to the mixture that was further distilled from 18 L to 11 L. Finally, another portion of water (6 L, 4 vol) was added and the solvents were distilled one last time from 17 L to 14 L, until no more THF was coming out. The reaction was then cooled down to 10 °C (internal temperature). The white slurry was filtered and the filter cake was washed with water (2 x 6 L, 2 x 4 vol). The solids were then dried at 80 °C (jacket temp) in the Aurora filter for 24 h (KF = 1.5 % H20) under vacuum to give 1.25 kg crude I, GDC-0032 (84% corrected yield, 96% wt, 97.3 % pure by HPLC) as a white solid.

A slurry of crude I (1.15 kg, 2.50 moles) in MeOH (6 L, 5 vol) was prepared and then charged to a 50 L glass reactor. Additional MeOH (24 L, 21 vol) was added to the mixture, which was then heated to 65 °C. A homogenous mixture was obtained. Si-thiol (Silicycle, Inc., 0.23 kg, 20% wt) was added to the solution via the addition port and the mixture was stirred for 3 hours. It was then filtered warm via the Aurora filter (jacket temperature = 60 °C, polish filtered and transferred directly into a second 50 L reactor with reduced pressure. The solution was then heated back to 65 °C internal temperature (IT). The homogeneous solution was cooled down to 54 °C and I seeds (12 g, 1 % wt) in MeOH (50 mL) were added with reduced pressure applied to the reactor. The mixture was then cooled down to 20 °C over 16 hours. The solids were then filtered via the Aurora filter and dried at 80 °C for 72 hours to give 921 g, 80% yield of I as a methanoate solvate (form A by XRPD,) and transferred to a pre-weighed charge-point bag.

In an isolator, the solids were slurred in IP Ac (8 L, 7 vol) and transferred to a clean 10 L reactor. The mixture was stirred for 1 h at 60 °C (IT). The solids were then filtered via the Aurora system and dried at 80 °C (jacket) for 96 h. A sample of I was removed and analyzed by GC (IP Ac = 1 %). To attempt more efficient drying, the API was transferred to two glass trays in an isolator and sealed with a drying bag before being dried in a vacuum oven set at 100 °C for 16 h. GC (IPC: Q12690V2) showed 1 % solvent was still present. The process afforded 760 g (68% corrected yield, 68% wt, 99.9 % purity by LC) of a white solid (form B by XRPD).

Crude I (340.7 g) was charged to a 2-1 . 1 1 DPI · bottle and slurried with 0.81 ,

isoamylalcohol (I A A). The slurry was transferred to a 20 L reactor and diluted with 6.7 L round- bottom flask (22 vol total). The white slurry was heated until a solution was observed (internal temperature rose to 118 °C and then cooled to 109 °C). The solution was polish filtered (0.2 μ .Μ filter). A flask was equipped with overhead stirring and the filtrate was slurried in isoamyl alcohol (344 ml ., 21 vol). The mixture was warmed to 95 °C (internal) until the solids dissolved. A slurry of charcoal (10 wt%, 0.16g) and silicycle thiol (10 wt%, 0.16g) in isoamyl alcohol (1 vol, 1 6 ml . ) was charged and the mixture was stirred at 90-95 °C for 1 h and then filtered (over Celite® pad). The clear amber colored solution was cooled to 73 °C (seeding temp range = 70 ±5 °C) and a GDC-0032 I seed (10 wt%, 0.16g) was added. The temperature of the heating mantle was turned off and the mixture was allowed to cool to room temperature overnight with stirrin (200 rpni). After 17 hr, the white solids were filtered starting with slow gravity filtration and then vacuum was applied. The solids were suction dried for 20 min with mixing until a free flowing powder was obtained. ( rude weight prior to oven drying = 16 g. The solids were oven- dried at 100 °C for 24 h and then sampled for testing. Drying continued at 100 °C for another 24 hr. I l l NMR (DMSO d6) δ 8.38 (t), 8.01 (s), 7.87 (s), 7.44, 7.46 (d), 7.36 (s), 7.18 (br s), 6.81

(br s), 5.82 (m), 3.99 (s), 2.50 (s), 2.26 (s), 1.75 (s), 1.48, 1.46 (d).

Purified 2-(4-(2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepin-9-yl)- lH-pyrazol- 1 -yl)-2-methylpropanamide I (GDC-0032) was dry granulation formulated in tablet form by the roller compaction method (He et al (2007) Jour, of Pharm. Sci., 96(5):1342-1355) with excipients including lactose, microcrystalline cellulose (AVICEL® PH 01, FMC BioPolymer, 50 μΜ particle),

croscarmellose sodium (Ac-Di-Sol®, FMC BioPolymer), and magnesium stearate.

Example 19 4-bromo-2-fluoro-N-hydroxybenzimidamide 24

To a solution of 4-Bromo-2-fluorobenzonitrile 11 (800 g, 4 mol, 1 eq), hydroxylamine hydrochloride (695 g, 10 mol, 2.5 eq) in MeOH (2 L, 2.5 vol) was added Et3N (485 g, 4.8 mol, 1.2 eq), then the mixture was stirred at 60 °C for 40 min and checked by HPLC (no nitrile remaining). Reaction was then quenched by H20 (30 L), and lots of off-white solid was separated out, and then filtered, the filter cake was washed with water (10 L x 2) and 1350 g wet 4-bromo-2-fluoro-N-hydroxybenzimidamide 24 was obtained with 96% purity.

Example 20 ethyl 3-(4-bromo-2-fluorobenzimidamidooxy)acrylate 25

To a solution of 4-Bromo-2-fluoro-N-hydroxybenzimidamide 24 (800 g, 3.43 mol, 1 eq) and Amberlyst® A21 (20 wt%, 160 g) in PhMe (12 L, 15 vol) was added ethyl propiolate (471 g, 4.8 mol, 1.4 eq) at 10 °C. The reaction was stirred at 50 °C overnight and checked by LC-MS (ca 14A% of starting material 24 was left). Reaction was then filtered and the filtrate was concentrated under vacuum, and 1015 g ethyl 3-(4-bromo-2-fluorobenzimidamidooxy)acrylate 25 was obtained as a yellow oil with 84.9% LC purity (yield: 89%).

Example 21 ethyl 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4-carboxylate 26

A solution of ethyl 3-(4-bromo-2-fluorobenzimidamidooxy)acrylate 25 (300 g, 0.91 mol, 1 eq) in diphenyl oxide (900 mL, 3 vol) was stirred at 190 °C under N2 for 1 h and checked by LC-MS (no 25 remaining). Cooled the mixture to rt and TBME (600 mL, 2 vol of 25) was added, and then PE (1.8 L, 6 vol of 25) was dropwise added to separate out solids. The mixture was stirred at rt for 20 min, and filtered to give 160 g wet cake. The wet cake was washed with PE (1 L) and dried to afford 120 g ethyl 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4-carboxylate 26 with 92% LC purity as brown solids. Example 22 ethyl 2-(4-bromo-2-fluorophenyl)- 1 -(2-hydroxyethyl)- 1 H-imidazole-4- carboxylate 27

Ethyl 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4-carboxylate 26 and l,3-dioxolan-2- one and N-methylimidazole were reacted to give 27.

Example 23 9-bromo-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxylic acid 28

Ethyl 2-(4-bromo-2-fluorophenyl)-l -(2-hydroxyethyl)- lH-imidazole-4-carboxylate 27, potassium hydroxide and methyl tributylammonium hydrochloride were reacted at 65 °C, cooled, and concentrated. The mixture was dissolved in ethanol and water to crystallize 28.

Example 24 9-bromo-N-(l-iminoethyl)-5,6-dihydrobenzo[f]imidazo[l,2- d] [ 1 ,4]oxazepine-2-carboxamide 29

9-Bromo-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxylic acid 28, triphenylphosphine, and acetamidine were reacted to give 29.

Example 25 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine III

9-Bromo-N-( 1 -iminoethyl)-5 ,6-dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepine-2- carboxamide 29 was reacted with isopropylhydrazine hydrochloride 4 in acetic acid to give III.

Example 26 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4-carboxylic acid 30

3-Chloro-2-oxopropanoic acid and 4-bromo-2-fluorobenzimidamide hydrochloride 12 are reacted with base to give 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4-carboxylic acid 30.

Alternatively, to a solution of ethyl 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4- carboxylate 26 (1350 g, 4.3 mol) in THF (8.1 L, 6 vol) and H20 (4 L, 3 vol) was added NaOH (520 g, 13 mol, 3 eq), and the reaction was stirred at 65 °C for 48 h till it completed (checked by LC-MS). Adjust the mixture with 2 M HC1 to pH = 5, and product was separated out as a yellow solid, filtered to give 2.2 kg wet cake, the wet cake was washed with H20 (1.5 L), DCM (1.5 L x 3), PE (1 L), and dried to afford 970 g pure 2-(4-bromo-2-fluorophenyl)-lH-imidazole-4- carboxylic acid 30 (Scheme 10). Example 27 5-(2-(4-bromo-2-fluorophenyl)-lH-imidazol-4-yl)-l-isopropyl-3-methyl- lH-l,2,4-triazole V

Reaction of 30 with N’-isopropylacetohydrazonamide 6 and coupling reagent HBTU in DMF gives intermediate, 2-(4-bromo-2-fluorophenyl)-N-(l-(2-isopropylhydrazinyl)ethylidene)- lH-imidazole-4-carboxamide 31 which cyclizes upon heating to give V.

Example 28 tert-butyl 2-hydroxyethylcarbamate gives tert-butyl 2-(5-bromo-2- cyanophenoxy)ethylcarbamate 32

Alkylation of 4-bromo-2-fluorobenzonitrile 11 with tert-butyl 2-hydroxyethylcarbamate gives 32.

Example 29 8-bromo-3,4-dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33

Cyclization of tert-butyl 2-hydroxyethylcarbamate gives tert-butyl 2-(5-bromo-2- cyanophenoxy)ethylcarbamate 32 under acidic conditions, such as hydrochloric acid in ethanol, gives 33.

Example 30 9-bromo-5,6-dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxylic acid 28

Reaction of 3-bromo-2-oxopropanoic acid and 8-bromo-3,4- dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33 gives 28 (CAS Reg. No. 1282516-74-8).

Example 31 9-bromo-2-(l-isopropyl-3-methyl-lH-l,2,4-triazol-5-yl)-5,6- dihydrobenzo[f]imidazo[ 1 ,2-d] [ 1 ,4]oxazepine III

Coupling of 28 with N’-isopropylacetohydrazonamide 6 and coupling reagent HBTU in

DMF gives intermediate, 9-bromo-N-(l-(2-isopropylhydrazinyl)ethylidene)-5,6- dihydrobenzo[f]imidazo[l,2-d][l,4]oxazepine-2-carboxamide 34, which forms III upon heating.

Example 32 methyl 4-bromo-2-fluorobenzimidate 35

Reaction of 4-bromo-2-fluorobenzonitrile 11 with sodium methoxide in methanol gives 35.

Example 33 8-bromo-3,4-dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33

Alkylation of methyl 4-bromo-2-fluorobenzimidate 35 with 2-aminoethanol gives 4- bromo-2-fluoro-N-(2-hydroxyethyl)benzimidamide 36, followed by cyclization to 33 (Scheme 13). Alternatively, reaction of 11 with 2- aminoethanol and potassium tert-butoxide displaces fluorine to give 2-(2-aminoethoxy)-4-bromobenzonitrile hydrochloride 37. Ring closure of 37 with trimethylaluminum gave 33 (Scheme 14). A solution of 11 (10 g, 50 mmol) and 2- aminoethanol (3.1 mL, 50.8 mmol) in 2-methyltetrahydrofuran (80 mL) was cooled to 0 °C and a solution of 1M potassium tert-butoxide in tetrahydrofuran (55 mL, 55 mmol) was slowly added while maintaining the solution temperature below 5 °C. The reaction was stirred at 0 °C for 30 min until judged complete by HPLC at which point it was warmed to 25 °C. A solution of 0.5M HC1 in isopropanol (100 mL, 50 mmol) was added and the desired HC1 salt 3 crystallized directly from the solution. The solid was collected by filtration and dried under vacuum with a nitrogen bleed to give 2-(2-aminoethoxy)-4-bromobenzonitrile hydrochloride 37 as a white solid. (12.1 g, 87 % yield).

To a flask was charged 37 (9.00 g, 32.4 mmol) and toluene (90.0 ml). The suspension was cooled to 0 °C and was added trimethylaluminum (1.8 equiv., 58.4 mmol, 2M in toluene) drop-wise over 30 minutes. The suspension was then stirred at room temperature for 1 h and then warmed to 100 °C. After 5 h, the solution was cooled to 0 °C and quenched with aqueous NaOH (2N, 90.0 ml). The suspension was extracted with EtOAc (4 x 90 ml) and the combined extracts were dried over then filtered through Celite®. The solution was concentrated and the residue triturated with EtOAc to afford 8-bromo-3,4-dihydrobenzo[f][l,4]oxazepin-5(2H)-imine 33 (6.26 g, 26.0 mmol, 80% yield) as white crystalline solid.

Example 34 4-chloro-2-fluoro-N-hydroxybenzimidamide 39

To a solution of 4-chloro-2-fluorobenzonitrile 38 (400 g, 2.58 mol, 1.0 eq),

hydroxylamine hydrochloride (448 g, 6.45 mol, 2.5 eq) in MeOH (1 L, 2.5 vol) was added Et3N (313 g, 3.1 mol, 1.2 eq), then the mixture was stirred at 60 °C for 40 min and checked by HPLC (no nitrile remaining). Reaction was then quenched by H20 (10 L), and lots of off-white solid was separated out, and then filtered, the filter cake was washed with water (10 L x 2) and 378 g 4-chloro-2-fluoro-N-hydroxybenzimidamide 39 was obtained with 93% purity (Scheme 15).

Example 35 ethyl 3-(4-chloro-2-fluorobenzimidamidooxy)acrylate 40

To a solution of 4-chloro-2-fluoro-N-hydroxybenzimidamide 39 (378 g, 2 mol, 1.0 eq) and Amberlyst® A21 (20 wt%, 75.6 g) in toluene PhMe (5.6 L, 15 vol) was added ethyl propiolate (275 g, 2.8 mol, 1.4 eq) at 30 °C. The reaction was stirred at 30 °C overnight and checked by LC-MS. Reaction was then filtered and the filtrate was concentrated under vacuum, and 550 g ethyl 3-(4-chloro-2-fluorobenzimidamidooxy)acrylate 40 was obtained as a yellow oil with 83% LC purity (Scheme 15).

Example 36 ethyl 2-(4-chloro-2-fluorophenyl)-lH-imidazole-4-carboxylate 41

A solution of ethyl 3-(4-chloro-2-fluorobenzimidamidooxy)acrylate 40 (550 g, 1.9 mol, 1.0 eq, 83% LC purity) in diphenyl oxide (1.65 L, 3 vol) was stirred at 190 °C under N2 for 1 h and checked by LC-MS (no 40 remaining). Cooled the mixture to rt and PE (10 L) was added dropwise. The mixture was stirred at rt for 20 min, and filtered to give 400 g wet cake, after purified by chromatography on silica gel (PE / EA=1 / 5) to get 175 g pure ethyl 2-(4-chloro-2- fluorophenyl)-lH-imidazole-4-carboxylate 41 with 98% LC purity (Scheme 15).

Example 37 2-(4-chloro-2-fluorophenyl)-lH-imidazole-4-carboxylic acid 42

To a solution of ethyl 2-(4-chloro-2-fluorophenyl)-lH-imidazole-4-carboxylate 41 (175 g, 4.3 mol) in THF (1 L, 6 vol) and H20 (500 mL, 3 vol) was added NaOH (78 g, 1.95 mol, 3.0 eq), and the reaction was stirred at 65 °C for 48 h till it completed (checked by LC-MS). Adjust the mixture with 2 N HC1 to pH = 5, and product was separated out as a yellow solid, filtered to give 210 g wet cake, the wet cake was washed with H20 (300 mL), DCM (3 x 300 mL), PE (500 mL), and dried to afford 110 g pure 2-(4-chloro-2-fluorophenyl)-lH-imidazole-4-carboxylic acid 42 (CAS Reg. No. 1260649-87-3) (Scheme 15 ). I l l NMR (DMSO d6) δ: 12.8 (br s), 8.0, 7.9 (br s), 7.46, 7.4 (m).

PATENT

US 2014275523

SYNTHESIS

Taselisib_药物数据_原料药API_CCIS-CHEM化学平台科研物资一站式采购平台 …

化学试剂

参考文献:

Image result for taselisib

CLIP

http://www.ccis-chem.com/goods.php?id=194272

商品规格

Taselisib

Taselisib是罗氏集团及其下属公司Genentech和Chugai研发,目前治疗绝经后妇女雌激素受体阳性(ER +)乳腺癌和非小细胞肺癌(NSCLC)的三期临床研究均在进行中。

基本信息更新时间:2016-02-01

药品名称:
Taselisib
研发代码:
GDC-0032; RG-7604
商品名称:
作用机制:
PI3K inhibitor; Cytochrome P450 CYP3A4 Inhibitors
适应症:
乳腺癌,非小细胞肺癌
研发阶段:
临床三期 (进行中)
研发公司:
罗氏 (原研)

化学信息更新时间:2015-08-27

分子量 460.53
分子式 C24H28N8O2
CAS号 1282512-48-4 (Taselisib);
化学名称 1H-Pyrazole-1-acetamide, 4-[5,6-dihydro-2-[3-methyl-1-(1-methylethyl)-1H-1,2,4-triazol-5-yl]imidazo[1,2-d][1,4]benzoxazepin-9-yl]-a,a-dimethyl-
Fudosteine药品(游离态)参数:
MW HD HA FRB* PSA* cLogP*
460.53 2 10 5 119 2.548±1.034

化学合成路线及相关杂质更新时间:2015-12-15

参考文献:J. Med. Chem. 2013, 56, 4597−4610

参考文献:WO2014140073A1

PAPER

J Med Chem 2013, 56(11): 4597

Discovery of 2-{3-[2-(1-Isopropyl-3-methyl-1H-1,2–4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (GDC-0032): A β-Sparing Phosphoinositide 3-Kinase Inhibitor with High Unbound Exposure and Robust in Vivo Antitumor Activity

Departments of Discovery Chemistry, Drug Metabolism and Pharmacokinetics, §Translational Oncology, and Biochemical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
 Argenta Discovery, 8-9 Spire Green Centre, Flex Meadow, Harlow, Essex, CM19 5TR, United Kingdom
J. Med. Chem.201356 (11), pp 4597–4610
DOI: 10.1021/jm4003632
*Phone: 650-225-2923 (C.O.N.); +1-(650)-467-3214 (T.P.H.). E-mail: chudin@gene.com (C.O.N.); theffron@gene.com (T.P.H.).
Abstract Image

Dysfunctional signaling through the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway leads to uncontrolled tumor proliferation. In the course of the discovery of novel benzoxepin PI3K inhibitors, we observed a strong dependency of in vivo antitumor activity on the free-drug exposure. By lowering the intrinsic clearance, we derived a set of imidazobenzoxazepin compounds that showed improved unbound drug exposure and effectively suppressed growth of tumors in a mouse xenograft model at low drug dose levels. One of these compounds, GDC-0032 (11l), was progressed to clinical trials and is currently under phase I evaluation as a potential treatment for human malignancies.

2-{3-[2-(1-Isopropyl-3-methyl-1H-1,2–4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (11l)

1H NMR (500 MHz, DMSO) δ 8.42 (s, 1H), 8.37 (d, J = 8.3 Hz, 1H), 8.02 (s, 1H), 7.89 (s, 1H), 7.46 (dd, J = 8.3, 1.8 Hz, 1H), 7.36 (d, J = 1.8 Hz, 1H), 7.22 (s, 1H), 6.87 (s, 1H), 5.90–5.73 (m, 1H), 4.62–4.42 (m, 4H), 2.50 (dt, J = 3.6, 1.7 Hz, 5H), 2.26 (s, 3H), 1.74 (s, 6H), 1.47 (d, J = 6.5 Hz, 6H). 13C NMR (126 MHz, DMSO) δ 173.78, 158.24, 155.88, 147.31, 143.94, 136.64, 134.60, 130.26, 129.88, 126.42, 123.62, 120.32, 119.31, 116.17, 115.26, 68.31, 64.48, 49.89, 49.70, 40.06, 39.97, 39.89, 39.80, 39.72, 39.63, 39.56, 39.47, 39.30, 39.13, 38.96, 25.47, 22.34, 13.82. HRMS (ESI+): m/z (M + H+) calcd: 461.2413, found: 461.2427. Melting point: 259 °C.
POLYMORPHS almost A to Z
GDC-0032, also known as taselisib, RG7604, or the IUPAC name: 2-(4-(2-(1-isopropyl-3-methyl-1H-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide, has potent PI3K activity (Ndubaku et al (2013) J. Med. Chem. 56(11):4597-4610; WO 2013/182668; WO 2011/036280; U.S. Pat. No. 8,242,104; U.S. Pat. No. 8,343,955) and is being studied in patients with locally advanced or metastatic solid tumors (Juric et al “GDC-0032, a beta isoform-sparing PI3K inhibitor: Results of a first-in-human phase Ia dose escalation study”, 2013 (Apr. 7) Abs LB-64 American Association for Cancer Research Annual Meeting).

the invention relates to polymorph forms of the PI3K inhibitor I (taselisib, GDC-0032, RG7604, CAS Reg. No. 1282512-48-4, Genentech, Inc.), named as 2-(4-(2-(1-isopropyl-3-methyl-1H-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide, having the structure:

Figure US09266903-20160223-C00001

and stereoisomers, geometric isomers, tautomers, and pharmaceutically acceptable salts thereof.

Publication numberPriority datePublication dateAssigneeTitle
WO2011036280A12009-09-282011-03-31F. Hoffmann-La Roche AgBenzoxazepin pi3k inhibitor compounds and methods of use
WO2014140073A12013-03-132014-09-18F. Hoffmann-La Roche AgProcess for making benzoxazepin compounds

References

Patent ID Patent Title Submitted Date Granted Date
US9670228 BENZOXAZEPIN PI3K INHIBITOR COMPOUNDS AND METHODS OF USE
2016-12-05
US9546178 BENZOXAZEPIN PI3K INHIBITOR COMPOUNDS AND METHODS OF USE
2015-11-03
2016-02-25
US9303043 PROCESS FOR MAKING BENZOXAZEPIN COMPOUNDS
2014-03-12
2014-09-18
US8785626 Benzoxazepin PI3K inhibitor compounds and methods of use
2013-10-07
2014-07-22
US2016135446 CELL STABILIZATION
2014-06-13
2016-05-19
Patent ID Patent Title Submitted Date Granted Date
US2016375033 METHODS OF TREATMENT WITH TASELISIB
2016-06-28
US2016045515 MUTANT SELECTIVITY AND COMBINATIONS OF A PHOSPHOINOSITIDE 3-KINASE INHIBITOR COMPOUND AND CHEMOTHERAPEUTIC AGENTS FOR THE TREATMENT OF CANCER
2015-07-06
2016-02-18
US2016117439 SUPERIOR BIOINFORMATICS PROCESS FOR IDENTIFYING AT RISK SUBJECT POPULATIONS
2015-10-26
2016-04-28
US2017096492 DOSAGE AND ADMINISTRATION OF ANTI-IGF-1R, ANTI-ErbB3 BISPECIFIC ANTIBODIES, USES THEREOF AND METHODS OF TREATMENT THEREWITH
2016-08-19
US9481690 Polymorphs of 2-(4-(2-(1-(isopropyl-3-methyl-1H-1, 2, 4, triazol-5-yl)-5, 6-dihydrobenzo[f] imidazo[1, 2-d][1, 4]oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide, methods of production, and pharmaceutical uses thereof
2016-01-08
2016-11-01
Patent ID Patent Title Submitted Date Granted Date
US9266903 POLYMORPHS OF 2-(4-(2-(1-ISOPROPYL-3-METHYL-1H-1, 2, 4-TRIAZOL-5-YL)-5, 6-DIHYDROBENZO[F]IMIDAZO[1, 2-D][1, 4]OXAZEPIN-9-YL)-1H-PYRAZOL-1-YL)-2-METHYLPROPANAMIDE, METHODS OF PRODUCTION, AND PHARMACEUTICAL USES THEREOF
2014-12-15
2015-06-18
US2016279142 COMBINATIONS OF A PHOSPHOINOSITIDE 3-KINASE INHIBITOR COMPOUND AND A CDK4/6 INHIBITOR COMPOUND FOR THE TREATMENT OF CANCER
2016-03-24
2016-09-29
US2016117443 BIOINFORMATICS PROCESS FOR IDENTIFYING AT RISK SUBJECT POPULATIONS
2015-10-26
2016-04-28
Patent ID Patent Title Submitted Date Granted Date
US2016220537 COMPOSITIONS TO IMPROVE THE THERAPEUTIC BENEFIT OF BISANTRENE AND ANALOGS AND DERIVATIVES THEREOF
2014-07-25
2016-08-04
US2016113932 TREATMENT OF CANCERS USING PI3 KINASE ISOFORM MODULATORS
2014-05-30
2016-04-28
US2014113879 BLOCK COPOLYMERS FOR STABLE MICELLES
2013-09-16
2014-04-24
US2014114051 BLOCK COPOLYMERS FOR STABLE MICELLES
2013-09-16
2014-04-24
US2014127271 BLOCK COPOLYMERS FOR STABLE MICELLES
2013-09-16
2014-05-08
Patent ID Patent Title Submitted Date Granted Date
US9555030 THERAPEUTIC USES OF SELECTED PYRAZOLOPYRIMIDINE COMPOUNDS WITH ANTI-MER TYROSINE KINASE ACTIVITY
2015-04-03
2015-10-15
US9555031 THERAPEUTIC USES OF SELECTED PYRROLOPYRIMIDINE COMPOUNDS WITH ANTI-MER TYROSINE KINASE ACTIVITY
2015-04-03
2015-10-15
US9649309 THERAPEUTIC USES OF SELECTED PYRIMIDINE COMPOUNDS WITH ANTI-MER TYROSINE KINASE ACTIVITY
2015-04-03
2015-10-15
US2015258080 THERAPEUTIC COMBINATIONS WITH ESTROGEN RECEPTOR MODULATORS
2015-03-12
2015-09-17
US2016166546 COMBINATORIAL METHODS TO IMPROVE THE THERAPEUTIC BENEFIT OF BISANTRENE AND ANALOGS AND DERIVATIVES THEREOF
2014-07-25
2016-06-16
Patent ID Patent Title Submitted Date Granted Date
US2016346408 IRON STABILIZED MICELLES AS MAGNETIC CONTRAST AGENTS
2016-05-26
US2016264732 BLOCK COPOLYMERS FOR STABLE MICELLES
2016-03-10
2016-09-15
US2016220569 CDK4/6 Inhibitor Dosage Formulations For The Protection Of Hematopoietic Stem And Progenitor Cells During Chemotherapy
2016-02-03
2016-08-04
US2015320754 COMBINATION THERAPIES
2015-04-15
2015-11-12
US2015320755 COMBINATION THERAPIES
2015-04-15
2015-11-12
Patent ID Patent Title Submitted Date Granted Date
US8242104 Benzoxazepin P13K inhibitor compounds and methods of use
2011-03-31
2012-08-14
US2017182043 Anti-Neoplastic Combinations and Dosing Regimens using CDK4/6 Inhibitor Compounds to Treat RB-Positive Tumors
2017-03-13
US2017246171 Treatment Of RB-Negative Tumors Using Topoisomerase Inhibitors In Combination With Cyclin Dependent Kinase 4/6 Inhibitors
2017-03-13
US2017136043 THERAPEUTIC USES OF SELECTED PYRROLOPYRIMIDINE COMPOUNDS WITH ANTI-MER TYROSINE KINASE ACTIVITY
2017-01-30
US2017135960 AGGREGATING MICROPARTICLES FOR MEDICAL THERAPY
2016-11-11
Patent ID Patent Title Submitted Date Granted Date
US9603850 MERTK-SPECIFIC PYRAZOLOPYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
US9198918 BENZOXAZEPIN PI3K INHIBITOR COMPOUNDS AND METHODS OF USE
2014-06-05
2014-09-25
US2014377258 Treatment Of Cancers Using PI3 Kinase Isoform Modulators
2014-05-30
2014-12-25
US2015283142 TREATMENT OF CANCERS USING PI3 KINASE ISOFORM MODULATORS
2013-11-01
2015-10-08
US8586574 Benzoxazepin PI3K inhibitor compounds and methods of use
2012-11-20
2013-11-19
Patent ID Patent Title Submitted Date Granted Date
US2017121421 ANTIBODY CONJUGATES COMPRISING TOLL-LIKE RECEPTOR AGONIST
2016-10-25
US9643980 Benzoxazepin oxazolidinone compounds and methods of use
2016-07-01
2017-05-09
US9650393 BENZOXAZEPIN OXAZOLIDINONE COMPOUNDS AND METHODS OF USE
2016-07-01
US2015291606 MERTK-SPECIFIC PYRROLOPYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
US2015291609 MERTK-SPECIFIC PYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
Taselisib
Taselisib skeletal.svg
Clinical data
ATC code
  • None
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
ChEMBL
Chemical and physical data
Formula C24H28N8O2
Molar mass 460.54 g·mol−1
3D model (JSmol)

Taselisib

(GDC-0032, RG7604)

BREAST
  • PHASE II,
  • III

This compound and its uses are investigational and have not been approved by the US Food and Drug Administration. Efficacy and safety have not been established. The information presented should not be construed as a recommendation for use. The relevance of findings in preclinical studies to humans is currently being evaluated.

Taselisib, a PI3K inhibitor

Taselisib, an investigational PI3K inhibitor, is currently in clinical development based on its potential selectivity for the PI3Kα isoform.1,2 Preclinical data have shown that taselisib induced growth inhibition in PI3Kα-mutant cell lines.Taselisib continues to be investigated in ongoing clinical studies.

1Taselisib is an investigational PI3K inhibitor currently being studied for its potential to selectively inhibit the PI3Kα isoform.1,2

2Taselisib is designed to bind to the ATP-binding pocket of PI3Kα to potentially prevent subsequent downstream signaling.1

3In preclinical studies, taselisib induced growth inhibition in PI3Kα-mutant xenograft mouse models.1 Taselisib continues to be investigated in ongoing clinical studies.

References

  1. Lopez S, Schwab CL, Cocco E, et al. Taselisib, a selective inhibitor of PIK3CA, is highly effective on PIK3CA-mutated and HER2/neu amplified uterine serous carcinoma in vitro and in vivo. Gynecol Oncol.2014;135:312-317. PMID: 25172762
  2. Ndubaku CO, Heffron TP, Staben ST, et al. Discovery of 2-{3-[2-(1-isopropyl-3-methyl-1H-1,2-4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (GDC-0032): a β-sparing phosphoinositide 3-kinase inhibitor with high unbound exposure and robust in vivo antitumor activity. J Med Chem. 2013;56:4597-4610. PMID: 23662903

//////////////////RG7604, Taselisib, PHASE 3,  metastatic breast cancer,  non-small cell lung cancer, RO5537381, Roche

CC1=NN(C(=N1)C2=CN3CCOC4=C(C3=N2)C=CC(=C4)C5=CN(N=C5)C(C)(C)C(=O)N)C(C)C

Larotrectinib, ларотректиниб , 拉罗替尼 ,

$
0
0

Image result for LarotrectinibImage result for Larotrectinib

Image result for LarotrectinibImage result for Larotrectinib

Larotrectinib

ARRY-470, LOXO-101, PF9462I9HX

Molecular Formula: C21H22F2N6O2
Molecular Weight: 428.444 g/mol
(3S)-N-{5-[(2R)-2-(2,5-Difluorphenyl)-1-pyrrolidinyl]pyrazolo[1,5-a]pyrimidin-3-yl}-3-hydroxy-1-pyrrolidincarboxamid
(S)-N-{5-[(R)-2-(2,5-Difluorophenyl)pyrrolidin-1-yl]pyrazolo[1,5-a]pyrimidin-3-yl}-3-hydroxypyrrolidine-1-carboxamide
10360
1223403-58-4 [RN]
UNII:PF9462I9HX
ларотректиниб [Russian] [INN]
拉罗替尼 [Chinese] [INN]
(3S)-N-[5-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl]pyrazolo[1,5-a]pyrimidin-3-yl]-3-hydroxypyrrolidine-1-carboxamide
NTRK-fusion solid tumours
TRK inhibitor
orphan drug designation in the U.S
In 2013, Array Biopharma licensed the product to Loxo Oncology for development and commercialization in the U.S. In 2016, breakthrough therapy designation was received in the U.S. for the treatment of unresectable or metastatic solid tumors with NTRK-fusion proteins in adult and pediatric patients who require systemic therapy and who have either progressed following prior treatment or who have no acceptable alternative treatments. In 2017, Bayer acquired global co-development and commercialization rights from Loxo Oncology.
  • Originator Array BioPharma
  • Developer Array BioPharma; Loxo Oncology; National Cancer Institute (USA)
  • Class Antineoplastics; Pyrazoles; Pyrimidines; Pyrrolidines; Small molecules
  • Mechanism of Action Tropomyosin-related kinase antagonists
  • Orphan Drug Status Yes – Solid tumours; Soft tissue sarcoma

Highest Development Phases

  • Preregistration Solid tumours
  • Phase II Histiocytosis; Non-Hodgkin’s lymphoma
  • Phase I/II CNS cancer
  • Preclinical Precursor cell lymphoblastic leukaemia-lymphoma

Most Recent Events

  • 29 May 2018 FDA assigns PDUFA action date of 26/11/2018 for larotrectinib for Solid tumors
  • 29 May 2018 Larotrectinib receives priority review status for Solid tumors in the US
  • 29 May 2018 The US FDA accepts NDA for larotrectinib for Solid tumours for review

Image result for LarotrectinibImage result for Larotrectinib

Larotrectinib sulfate

(3S)-N-[5-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl]pyrazolo[1,5-a]pyrimidin-3-yl]-3-hydroxypyrrolidine-1-carboxamide;sulfuric acid

Larotrectinib (LOXO-101) sulfate is an oral potent and selective ATP-competitive inhibitor of tropomyosin receptor kinases (TRK).

    • Crystalline Form (I-HS) OF

SULFATE SALT REPORTED IN https://patents.google.com/patent/US20170165267

nmr  http://file.selleckchem.com/downloads/nmr/s796001-loxo-101-methanol-hnmr-selleck.pdf

Figure US20170165267A1-20170615-C00006Figure US20170165267A1-20170615-C00007

Molecular Weight 526.51
Formula C21H22F2N6O2.H2O4S
CAS No. 1223405-08-0
  1. LOXO-101 sulfate
  2. Larotrectinib sulfate
  3. LOXO-101 (sulfate)
  4. 1223405-08-0
  5. UNII-RDF76R62ID
  6. RDF76R62ID
  7. ARRY-470 sulfate
  8. LOXO-101(sulfate)
  9. Larotrectinib sulfate [USAN]
  10. PXHANKVTFWSDSG-QLOBERJESA-N
  11. HY-12866A
  12. s7960
  13. AKOS030526332
  14. CS-5314

LOXO-101 is a small molecule that was designed to block the ATP binding site of the TRK family of receptors, with 2 to 20 nM cellular potency against the TRKA, TRKB, and TRKC kinases. IC50 value: 2 – 20 nM Target: TRKA/B/C in vitro: LOXO-101 is an orally administered inhibitor of the TRK kinase and is highly selective only for the TRK family of receptors. LOXO-101 is evaluated for off-target kinase enzyme inhibition against a panel of 226 non-TRK kinases at a compound concentration of 1,000 nM and ATP concentrations near the Km for each enzyme. In the panel, LOXO-101 demonstrates greater than 50% inhibition for only one non-TRK kinase (TNK2 IC50, 576 nM). Measurement of proliferation following treatment with LOXO-101 demonstrates a dose-dependent inhibition of cell proliferation in all three cell lines. The IC50 is less than 100 nM for CUTO-3.29 and less than 10 nM for KM12 and MO-91, consistent with the known potency of this drug for the TRK kinase family. [1] LOXO-101 demonstrates potent and highly-selective inhibition of TRKA, TRKB, and TRKC over other kinase- and non-kinase targets. LOXO-101 is a potent, ATP-competitive TRK inhibitor with IC50s in low nanomolar range for inhibition of all TRK family members in binding and cellular assays, with 100x selectivity over other kinases. [2] in vivo: Athymic nude mice injected with KM12 cells are treated with LOXO-101 orally daily for 2 weeks. Dose-dependent tumor inhibition is observed, demonstrating the ability of this selective compound to inhibit tumor growth in vivo. [1]

Image result for Larotrectinib

DOI

https://doi.org/10.1038/nrd.2018.4

SYNTHESIS

WO 2010048314

Synthesis of larotrectinib

N-Boc-pyrrolidine as starting material The method involves enantioselective deprotonation, transmetalation with ZnCl2, Negishi coupling with 2-bromo-1,4-difluorobenzene,

N-arylation with 5-chloropyrazolo[1,5-a]pyrimidine, nitration, nitro reduction and condensation with CDI and 3(S)-pyrrolidinol.

PRODUCT Patent

WO 2010048314

https://patents.google.com/patent/WO2010048314A1

InventorJulia HaasSteven W. AndrewsYutong JiangGan Zhang

Original AssigneeArray Biopharma Inc.

Priority date 2008-10-22

Example 14


(S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-alpyrimidin-3-yl)- 3 -hydroxypyrrolidine- 1 -carboxamide

[00423] To a DCM (0.8 mL) solution of (R)-5-(2-(2,5-difiuorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidin-3-amine (Preparation B; 30 mg, 0.095 mmol) was added CDI (31 mg, 0.19 mmol) at ambient temperature in one portion. After stirring two hours, (S)-pyrrolidin-3-ol (17 mg, 0.19 mmol) [purchased from Suven Life Sciences] was added in one portion. The reaction was stirred for 5 minutes before it was concentrated and directly purified by reverse-phase column chromatography, eluting with 0 to 50% acetonitrile/water to yield the final product as a yellowish foamy powder (30 mg, 74% yield). MS (apci) m/z = 429.2 (M+H).

Example 14A


(S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolori,5-alpyrimidin-3-yl)- 3 -hydroxypyrrolidine- 1 -carboxamide sulfate

[00424] To a solution of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolo [ 1 ,5 -a]pyrimidin-3 -yl)-3 -hydroxypyrrolidine- 1 -carboxamide (4.5 mg, 0.011 mmol) in methanol (1 mL) at ambient temperature was added sulfuric acid in MeOH (105 μL, 0.011 mmol). The resulting solution was stirred for 30 minutes then concentrated to provide (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidin-3-yl)-3 -hydroxypyrrolidine- 1 -carboxamide sulfate (5.2 mg, 0.0099 mmol, 94 % yield) as a yellow solid.

PATENT

WO 2017201241 

Examples

Preparation of 10:

1)

(R,E)-N-(2,5-difluorobenzylidene)-2-methylpropane-2-sulfinamide (17): Compound 16 and (R)-2-methylpropane-2-sulfinamide (1.05 eq.) were charged to a reactor outfitted with a mechanical stirrer, reflux condensor, J-Kem temperature probe under N2. DCM (3 mL/g of 14) was added (endothermic from 22 °C to about 5 °C) followed by addition of cesium carbonate (0.70 eq.) (exothermic to -50 °C). Once the addition was complete, the reaction mixture was stirred at room temperature for 3 h (slowly cools from about 40 °C). When the reaction was called complete (HPLC) the mixture was filtered through Celite. The Celite pad (0.3 wt eq) was equilibrated with DCM (1 mL/g of 16), and the reaction mixture was poured through the pad. The Celite cake was washed with DCM (2 x 1 mL/g), and the filtrate concentrated partially to leave about 0.5 to 1 mL/g DCM remaining. The orange solution was stored at room temperature (generally overnight) and used directly in the next reaction. (100% yield was assumed).

2)

(R)-N-((R)-l-(2,5-difluorophenyl)-3-(l,3-dioxan-2-yl)propyl)-2-methylpropane-2-sulfinamide (19): To a reactor equipped with overhead stirring, reflux condensor, under

nitrogen, was added magnesium turnings (2.0 eq), and THF (8 mL/g of 17). The mixture was heated to 40 °C. Dibal-H (25% wt in toluene, 0.004 eq) was added to the solution, and the suspension heated at 40 °C for 25 minutes. A solution of 2-(2-bromoethyl)-l,3-dioxane (18) (2 eq) in THF (4.6 mL/g of 17) was added dropwise to the Mg solution via addition funnel. The solution temperature was maintained < 55 °C. The reaction progress was monitored by GC. When the Grignard formation was judged complete, the solution was cooled to -30 °C, and 17 (1.0 eq, in DCM) was added dropwise via addition funnel. The temperature was kept between -30 °C and -20 °C and the reaction was monitored for completion (FIPLC). Once the reaction was called complete, the suspension (IT = -27.7 °C) was vacuum transferred to a prepared and cooled (10 °C) 10% aqueous citric acid solution (11 mL/g of 17). The mixture temperature rose to 20 °C during transfer. The milky solution was allowed to stir at ambient temperature overnight. MTBE (5.8 mL/g) was added to the mixture, and it was transferred to a separatory funnel. The layers were allowed to separate, and the lower aqueous layer was removed. The organic layer was washed with sat. NaHC03 (11 mL/g) and then sat. NaCl (5.4 mL/g). The organic layer was removed and concentrated to minimum volume via vacuum distillation. MTBE (2 mL/g) was added, and the mixture again concentrated to minimum volume. Finally MTBE was added to give 2 mL/g total MTBE (GC ratio of MTBE:THF was about 9: 1), and the MTBE mixture was heated to 50 °C until full dissolution occurred. The MTBE solution was allowed to cool to about 35 °C, and heptane was added portion -wise. The first portion (2 mL/g) is added, and the mixture allowed to stir and form a solid for 1-2 h, and then the remainder of the heptane is added (8 mL/g). The suspension was allowed to stir for >lh. The solids were collected via filtration through polypropylene filter cloth (PPFC) and washed with 10% MTBE in heptane (4 mL/g. The wet solid was placed in trays and dried in a vacuum oven at 55 °C until constant weight (3101 g, 80.5%, dense white solid, 100a% and 100wt%).

3)

(R)-2-(2,5-difluorophenyl)pyrrolidine (R)-2-hydroxysuccinate (10): To a flask containing 4: 1 TFA:water (2.5 mL/g, pre-mixed and cooled to <35 °C before adding 19) was added (R)-N-((R)-l-(2,5-difluorophenyl)-3-(l,3-dioxan-2-yl)propyl)-2-methylpropane-2-sulfinamide (19) (1 eq). The mixture temperature rose from 34 °C to 48 °C and was stirred at ambient temperature for 1 h. Additional TFA (7.5 mL/g) was added, followed by triethylsilane (3 eq) over 5 minutes. The biphasic mixture was stirred vigorously under nitrogen for 21 h until judged complete (by GC, <5% of imine). The mixture was then concentrated under vacuum until -10 kg target mass (observed 10.8 kg after concentration). The resulting concentrate was transferred to a separatory funnel and diluted with MTBE (7.5 mL/g), followed by water (7.5 mL/g). The layers were separated. The MTBE layer was back-extracted with 1M HC1 (3 mL/g). The layers were separated, and the aqueous layers were combined in a round-bottomed flask with DCM (8 mL/g). The mixture was cooled in an ice bath and 40% NaOH was charged to adjust the pH to >12 (about 0.5 mL/g; the temperature went from 24 °C to 27 °C, actual pH was 13), and the layers separated in the separatory funnel. The aqueous layer was back-extracted twice with DCM (2 x 4 mL/g). The organic layers were concentrated to an oil (<0.5 mL/g) under vacuum (rotovap) and EtOH (1 mL/g based on product) was added. The yellow solution was again concentrated to an oil (81% corrected yield, with 3% EtOH, 0.2% imine and Chiral HPLC showed 99.7%ee).

Salt formation: To a solution of (R)-2-(2,5-difluorophenyl)pyrrolidine 10 (1 eq) in EtOH (15 mL/g) was added Z)-(+)-Malic Acid (1 eq). The suspension was heated to 70 °C for 30 minutes (full dissolution had occurred before 70 °C was reached), and then allowed to cool to room temperature slowly (mixture was seeded when the temperature was < 40 °C). The slurry was stirred at room temperature overnight, then cooled to <5 °C the next morning. The suspension was stirred at <5 °C for 2h, filtered (PPFC), washed with cold EtOH (2 x 2 mL/g), and dried (50-55 °C) under vacuum to give the product as a white solid (96% based on 91% potency, product is an EtOH solvate or hemi- solvate).

Preparation of the compound of Formula I:

1)

(R)-5-(2-(2,5-difluorophenyl)pyrrolidin-l-yl)-3-nitropyrazolo[l,5-a]pyrimidine (11):

Compound 5 and 10 (1.05 eq) were charged to a reactor outfitted with a mechanical stirrer, J-Kem temperature probe, under N2. EtOH and THF (4: 1, 10 mL/g of 5) were added and the mixture was cooled to 15-25 °C. Triethylamine (3.5 eq) was added and the internal temp generally rose from 17.3 – 37.8 °C. The reaction was heated to 50 – 60 °C and held at that temperature for 7 h. Once the reaction is judged complete (HPLC), water (12 mL/g of 5) is added maintaining the temperature at 50 – 60 °C. The heat is removed and the suspension was slowly cooled to 21 °C over two h. After stirring at -21 °C for 2 h, the suspension was centrifuged and the cake was washed with water (3 x 3 mL/g of 5). The solid was transferred to drying trays and placed in a vacuum oven at 50 – 55 °C to give 11.

2)

(R)-5-(2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidin-3-amine fumarate Pt/C hydrogenation (12 fumarate): To a Parr reactor was charged 11 (1.0 eq), 5% Pt/C ~ 50 wt% water (2 mol% Pt / Johnson Matthey B 103018-5 or Sigma Aldrich 33015-9), and MeOH (8 mL/g). The suspension was stirred under hydrogen at 25-30 psi and the temperature was maintained below 65 °C for ~8 h. When the reaction was called complete (HPLC), the reaction was cooled to 15 – 25 °C and the hydrogen atmosphere was replaced with a nitrogen atmosphere. The reaction mixture was filtered through a 2 micron bag filter and a 0.2 micron line filter in series. The filtrate from the Pt/C hydrogenation was transferred to a reactor under nitrogen with mechanical stirring and then MTBE (8 mL/g) and fumaric acid (1.01 eq) were charged. The mixture was stirred under nitrogen for 1 h and solids formed after -15 min. The mixture was cooled to -10 to -20 °C and stirred for 3 h. The suspension was filtered (PPFC), washed with MTBE (-2.5 mL/g), and the solids was dried under vacuum at 20-25 °C with a nitrogen bleed to yield an off-white solid (83% yield).

3)

Phenyl (5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)-3,3a-dihydropyrazolo[l,5-a]pyrimidin-3-yl)carbamate (13): To a 5 to 15°C solution of 12-fumarate (1.0 eq) in 2-MeTHF (15 mL/g) was added a solution of potassium carbonate (2.0 eq.) in water (5 mL/g) followed by phenyl chloroformate (1.22 eq.) (over 22 min, an exotherm from 7 °C to 11 °C occurred). The mixture was stirred for 2 h and then the reaction was called complete (HPLC). The stirring ceased and the aqueous layer was removed. The organic layer was washed with brine (5 mL/g) and concentrated to ca. 5 mL/g of 2-MeTHF under vacuum and with heating to 40 °C. To the 2-MeTHF solution was added heptanes (2.5 mL/g) followed by seeds (20 mg, 0.1 wt%). This mixture was allowed to stir at room temperature for 2 h (until a solid formed), and then the remainder of the heptanes (12.5 mL/g) was added. The mixture was stirred at ambient temperature for 2 h and then the solids were collected via filtration (PPFC), washed with 4: 1 heptanes :MeTHF (2 x 2 mL/g), and dried to give 13 (96%).

4)

(S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-l-carboxamide hydrogen sulfate: To a flask containing 13 (1.0 eq) was added a solution of (S)-pyrrolidin-3-ol (1.1 eq.) in EtOH (10 mL/g). The mixture was heated at 50 – 60 °C for 5 h, called complete (HPLC), and then cooled to 20-35 °C. Once <35°C, the reaction was polish-filtered (0.2 micron) into a clean reaction vessel and the mixture was cooled to -5 to 5 °C. Sulfuric acid (1.0 eq.) was added over 40 minutes, the temperature rose to 2 °C and the mixture was seeded. A solid formed, and the mixture was allowed to stir at -5 to 5 °C for 6.5 h. Heptanes (10 mL/g) was added, and the mixture stirred for 6.5 h. The

suspension was filtered (PPFC), washed with 1 : 1 EtOH:heptanes (2 x 2 mL/g), and dried (under vacuum at ambient temperature) to give Formula I (92.3%).

Preparation of the hydrogen sulfate salt of the compound of Formula I:

Concentrated sulfuric acid (392 mL) was added to a solution of 3031 g of (S)-N-(5- ((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)-pyrazolo[l,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-l-carboxamide in 18322 mL EtOH to form the hydrogen sulfate salt. The solution was seeded with 2 g of (,S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl)-pyrazolo[l,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-l-carboxamide hydrogen sulfate and the solution was stirred at room temperature for at least 2 hours to form a slurry of the hydrogen sulfate salt. Heptane (20888 g) was added and the slurry was stirred at room temperature for at least 60 min. The slurry was filtered and the filter cake was washed with 1 : 1 heptane/EtOH. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius).

The dried hydrogen sulfate salt (6389 g from 4 combined lots) was added to a 5 :95 w/w solution of water/2-butanone (total weight 41652 g). The mixture was heated at about 68° Celsius with stirring until the weight percent of ethanol was about 0.5%, during which time a slurry formed. The slurry was filtered, and the filter cake was washed with a 5 :95 w/w solution of water/2-butanone. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius) to provide the crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-l-yl)-pyrazolo[l,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-l-carboxamide hydrogen sulfate.

PATENT

US2017165267

https://patents.google.com/patent/US20170165267

Provided herein is a novel crystalline form of the compound of Formula I:

[0000]

Figure US20170165267A1-20170615-C00001

also known as (S)—N-(5-((R)-2-(2, 5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide. In particular, the novel crystalline form comprises the hydrogen sulfate salt of the compound of Formula I in a stable polymorph form, hereinafter referred to as crystalline form (I-HS) and LOXO-101, which can be characterized, for example, by its X-ray diffraction pattern—the crystalline form (I-HS) having the formula:

[0000]

Figure US20170165267A1-20170615-C00002

In some embodiments of the above step (c), the base is an alkali metal base, such as an alkali metal carbonate, such as potassium carbonate.

Figure US20170165267A1-20170615-C00004

Preparation of 5-chloro-3-nitropyrazolo[1,5-a]pyrimidine Step A—Preparation of sodium pyrazolo[1,5-a]pyrimidin-5-olate

A solution of 1H-pyrazol-5-amine and 1,3-dimethylpyrimidine-2,4(1H,3H)-dione (1.05 equiv.) were charged to a round bottom flask outfitted with a mechanical stirrer, a steam pot, a reflux condenser, a J-Kem temperature probe and an Nadaptor for positive Npressure control. Under mechanical stirring the solids were suspended with 4 vol. (4 mL/g) of absolute EtOH under a nitrogen atmosphere, then charged with 2.1 equivalents of NaOEt (21 wt % solution in EtOH), and followed by line-rinse with 1 vol. (1 mL/g) of absolute EtOH. The slurry was warmed to about 75° Celsius and stirred at gentle reflux until less than 1.5 area % of 1H-pyrazol-5-amine was observed by TRK1PM1 HPLC to follow the progression of the reaction using 20 μL of slurry diluted in 4 mL deionized water and 5 μL injection at 220 nm.

After 1 additional hour, the mixture was charged with 2.5 vol. (2.5 mL/g) of heptane and then refluxed at 70° Celsius for 1 hour. The slurry was then cooled to room temperature overnight. The solid was collected by filtration on a tabletop funnel and polypropylene filter cloth. The reactor was rinsed and charged atop the filter cake with 4 vol. (4 mL/g) of heptane with the cake pulled and the solids being transferred to tared drying trays and oven-dried at 45° Celsius under high vacuum until their weight was constant. Pale yellow solid sodium pyrazolo[1,5-a]-pyrimidin-5-olate was obtained in 93-96% yield (corrected) and larger than 99.5 area % observed by HPLC (1 mg/mL dilution in deionized water, TRK1PM1 at 220 nm).

Step B—Preparation of 3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one

A tared round bottom flask was charged with sodium pyrazolo[1,5-a]pyrimidin-5-olate that was dissolved at 40-45° Celsius in 3.0 vol. (3.0 mL/g) of deionized water, and then concentrated under high vacuum at 65° Celsius in a water-bath on a rotary evaporator until 2.4× weight of starting material was observed (1.4 vol/1.4 mL/g deionized water content). Gas chromatography (GC) for residual EtOH (30 μL of solution dissolved in ˜1 mL MeOH) was performed showing less than 100 ppm with traces of ethyl nitrate fumes being observed below upon later addition of HNO3. In some cases, the original solution was charged with an additional 1.5 vol. (1.5 mL/g) of DI water, then concentrated under high vacuum at 65° Celsius in a water-bath on a rotary evaporator until 2.4× weight of starting material was observed (1.4 vol/1.4 mL/g DI water content). Gas chromatograph for residual EtOH (30 μL of solution dissolved in about 1 mL MeOH) was performed showing <<100 ppm of residual EtOH without observing any ethyl nitrate fumes below upon later addition of HNO3.

A round bottom vessel outfitted with a mechanical stirrer, a steam pot, a reflux condenser, a J-Kem temperature probe and an Nadaptor for positive Npressure control was charged with 3 vol. (3 mL/g, 10 equiv) of >90 wt % HNOand cooled to about 10° Celsius under a nitrogen atmosphere using external ice-water cooling bath under a nitrogen atmosphere. Using a pressure equalizing addition funnel, the HNO3solution was charged with the 1.75-1.95 volumes of a deionized water solution of sodium pyrazolo[1,5-a]pyrimidin-5-olate (1.16-1.4 mL DI water/g of sodium pyrazolo[1,5-a]pyrimidin-5-olate) at a rate to maintain 35-40° Celsius internal temperature under cooling. Two azeotropes were observed without any ethyl nitrate fumes. The azeotrope flask, the transfer line (if applicable) and the addition funnel were rinsed with 2×0.1 vol. (2×0.1 mL/g) deionized water added to the reaction mixture. Once the addition was complete, the temperature was gradually increased to about 45-50° Celsius for about 3 hours with HPLC showing >99.5 area % conversion of sodium pyrazolo[1,5-a]pyrimidin-5-olate to 3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one.

Step C—Preparation of 5-chloro-3-nitropyrazolo[1,5-a]pyrimidine

3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one was charged to a round bottom flask outfitted with a mechanical stirrer, a heating mantle, a reflux condenser, a J-Kem temperature probe and an Nadaptor for positive N2pressure control. Under mechanical stirring the solids were suspended with 8 volumes (8 mL/g) of CH3CN, and then charged with 2,6-lutitine (1.05 equiv) followed by warming the slurry to about 50° Celsius. Using a pressure equalizing addition funnel, the mixture was dropwise charged with 0.33 equivalents of POCl3. This charge yielded a thick, beige slurry of a trimer that was homogenized while stirring until a semi-mobile mass was observed. An additional 1.67 equivalents of POClwas charged to the mixture while allowing the temperature to stabilize, followed by warming the reaction mixture to a gentle reflux (78° Celsius). Some puffing was observed upon warming the mixture that later subsided as the thick slurry got thinner.

The reaction mixture was allowed to reflux until complete dissolution to a dark solution and until HPLC (20 μL diluted in 5 mL of CH3CN, TRK1PM1 HPLC, 5 μL injection, 268 nm) confirmed that no more trimer (RRT 0.92) was present with less than 0.5 area % of 3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one (RRT 0.79) being observed by manually removing any interfering and early eluting peaks related to lutidine from the area integration. On a 1.9 kg scale, 0 area % of the trimer, 0.25 area % of 3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one, and 99.5 area % of 5-chloro-3-nitropyrazolo[1,5-a]pyrimidine was observed after 19 hours of gentle reflux using TRK1PM1 HPLC at 268 [0000]

Figure US20170165267A1-20170615-C00005

Preparation of (R)-2-(2,5-difluorophenyl)-pyrrolidine (R)-2-hydroxysuccinate Step A—Preparation of tert-butyl(4-(2,5-difluorophenyl)-4-oxobutyl)-carbamate

2-bromo-1,4-difluorobenzene (1.5 eq.) was dissolved in 4 volumes of THF (based on weight of tert-butyl 2-oxopyrrolidine-1-carboxylate) and cooled to about 5° Celsius. A solution of 2.0 M iPrMgCl in THF (1.4 eq.) was added over 2 hours to the mixture while maintaining a reaction temperature below 25° Celsius. The solution was allowed to cool to about 5° Celsius and stirred for 1 hour (GC analysis confirmed Grignard formation). A solution of tert-butyl 2-oxopyrrolidine-1-carboxylate (1.0 eq.) in 1 volume of THF was added over about 30 min while maintaining a reaction temperature below 25° Celsius. The reaction was stirred at about 5° Celsius for 90 min (tert-butyl 2-oxopyrrolidine-1-carboxylate was confirmed to be less than 0.5 area % by HPLC). The reaction was quenched with 5 volumes of 2 M aqueous HCl while maintaining a reaction temperature below 45° Celsius. The reaction was then transferred to a separatory funnel adding 10 volumes of heptane and removing the aqueous layer. The organic layer was washed with 4 volumes of saturated aqueous NaCl followed by addition of 2×1 volume of saturated aqueous NaCl. The organic layer was solvent-switched to heptane (<1% wt THF confirmed by GC) at a distillation temperature of 35-55° Celsius and distillation pressure of 100-200 mm Hg for 2×4 volumes of heptane being added with a minimum distillation volume of about 7 volumes. The mixture was then diluted to 10 volumes with heptane while heating to about 55° Celsius yielded a denser solid with the mixture being allowed to cool to room temperature overnight. The slurry was cooled to less than 5° Celsius and filtered through polypropylene filter cloth. The wet cake was washed with 2×2 volumes of heptane. The solids were dried under vacuum at 55° Celsius until the weight was constant, yielding tert-butyl(4-(2,5-difluorophenyl)-4-oxobutyl)-carbamate as a white solid at about 75% to 85% theoretical yield.

Step B—Preparation of 5-(2,5-difluorophenyl)-3,4-dihydro-2H-pyrrole

tert-butyl(4-(2,5-difluorophenyl)-4-oxobutyl)-carbamate was dissolved in 5 vol. of toluene with 2.2 eq. of 12M HCl being added observing a mild exotherm and gas evolution. The reaction was heated to 65° Celsius for 12-24 hours and monitored by HPLC. Upon completion the reaction was cooled to less than 15° Celsius with an ice/water bath. The pH was adjusted to about 14 with 3 equivalents of 2M aqueous NaOH (4.7 vol.). The reaction was stirred at room temperature for 1-2 hours. The mixture was transferred to a separatory funnel with toluene. The aqueous layer was removed and the organic layer was washed with 3 volumes of saturated aqueous NaCl. The organic layer was concentrated to an oil and redissolved in 1.5 volumes of heptane. The resulting suspension was filtered through a GF/F filter paper and concentrated to a light yellow oil of 5-(2,5-difluorophenyl)-3,4-dihydro-2H-pyrrole with a 90% to 100% theoretical yield.

Step C—Preparation of (R)-2-(2,5-difluorophenyl)-pyrrolidine

Chloro-1,5-cyclooctadiene iridium dimer (0.2 mol %) and (R)-2-(2-(diphenylphosphino)phenyl)-4-isopropyl-4,5-dihydrooxazole (0.4 mol %) were suspended in 5 volumes of MTBE (based on 5-(2,5-difluorophenyl)-3,4-dihydro-2H-pyrrole) at room temperature. The mixture was stirred for 1 hour and most of the solids dissolved with the solution turning dark red. The catalyst formation was monitored using an HPLC/PDA detector. The reaction was cooled to less than 5° Celsius and 5-(2,5-difluorophenyl)-3,4-dihydro-2H-pyrrole (1.0 eq.) was added using a 0.5 volumes of MTBE rinse. Diphenylsilane (1.5 eq.) was added over about 20 minutes while maintaining a reaction temperature below 10° Celsius. The reaction was stirred for 30 minutes below 10° Celsius and then allowed to warm to room temperature. The reaction was stirred overnight at room temperature. The completion of the reaction was confirmed by HPLC and then cooled to less than 5° Celsius. The reaction was quenched with 5 volumes of 2M aqueous HCl maintaining temperature below 20° Celsius. After 10 minutes the ice/water bath was removed and the reaction temperature was allowed to increase to room temperature while stirring for 2 hours. The mixture was transferred to a separatory funnel with 3 volumes of MTBE. The aqueous layer was washed with 3.5 volumes of MTBE followed by addition of 5 volumes of MTBE to the aqueous layer while adjusting the pH to about 14 by adding 0.75 volumes of aqueous 50% NaOH. The organic layer was washed with 5 volumes of aqueous saturated NaCl, then concentrated to an oil, and diluted with 3 volumes of MTBE. The solution was filtered through a polypropylene filter cloth and rinsed with 1 volume of MTBE. The filtrate was concentrated to an oil of (R)-2-(2,5-difluorophenyl)-pyrrolidine with a 95% to 100% theoretical yield and with 75-85% ee.

Step D—Preparation of (R)-2-(2,5-difluorophenyl)-pyrrolidine (R)-2-hydroxy-succinate

(R)-2-(2,5-difluorophenyl)-pyrrolidine (1.0 eq.) was transferred to a round bottom flask charged with 15 volumes (corrected for potency) of EtOH (200 prf). D-malic acid (1.05 eq.) was added and the mixture was heated to 65° Celsius. The solids all dissolved at about 64° Celsius. The solution was allowed to cool to RT. At about 55° Celsius the solution was seeded with (R)-2-(2,5-difluorophenyl)-pyrrolidine (R)-2-hydroxy-succinate (about 50 mg, >97% ee) and stirred at room temperature overnight. The suspension was then filtered through a polypropylene filter cloth and washed with 2×1 volumes of EtOH (200 prf). The solids were dried under vacuum at 55° Celsius, yielding (R)-2-(2,5-difluorophenyl)-pyrrolidine (R)-2-hydroxy-succinate with a 75% to 90% theoretical yield and with >96% ee.

Referring to Scheme 1, suitable bases include tertiary amine bases, such as triethylamine, and K2CO3. Suitable solvents include ethanol, heptane and tetrahydrofuran (THF). The reaction is conveniently performed at temperatures between 5° Celsius and 50° Celsius. The reaction progress was generally monitored by HPLC TRK1PM1.

Figure US20170165267A1-20170615-C00006

Figure US20170165267A1-20170615-C00007

[0247]

Compounds II (5-chloro-3-nitropyrazolo[1,5-a]pyrimidine) and III ((R)-2-(2,5-difluorophenyl)-pyrrolidine (R)-2-hydroxysuccinate, 1.05 eq.) were charged to a round bottom flask outfitted with a mechanical stirrer, a J-Kem temperature probe and an Nadaptor for positive Npressure control. A solution of 4:1 EtOH:THF (10 mL/g of compound II) was added and followed by addition of triethylamine (NEt3, 3.50 eq.) via addition funnel with the temperature reaching about 40° Celsius during addition. Once the addition was complete, the reaction mixture was heated to 50° Celsius and stirred for 0.5-3 hours to yield compound IV.

To a round bottom flask equipped with a mechanical stirrer, a J-Kem temperature probe, and an Ninlet compound IV was added and followed by addition of tetrahydrofuran (10 mL/g of compound IV). The solution was cooled to less than 5° Celsius in an ice bath, and Zn (9-10 eq.) was added. 6M HCl (9-10 eq.) was then added dropwise at such a rate to keep the temperature below 30° Celsius (for 1 kg scale the addition took about 1.5 hours). Once the exotherm subsided, the reaction was allowed to warm to room temperature and was stirred for 30-60 min until compound IV was not detected by HPLC. At this time, a solution of potassium carbonate (K2CO3, 2.0 eq.) in water (5 mL/g of compound IV) was added all at once and followed by rapid dropwise addition of phenyl chloroformate (PhOCOCl, 1.2 eq.). Gas evolution (CO2) was observed during both of the above additions, and the temperature increased to about 30° Celsius after adding phenyl chloroformate. The carbamate formation was stirred at room temperature for 30-90 min. HPLC analysis immediately followed to run to ensure less than 1 area % for the amine being present and high yield of compound VI in the solution.

To the above solution amine VII ((S)-pyrrolidin-3-ol, 1.1 eq. based on theoretical yield for compound VI) and EtOH (10 mL/g of compound VI) was added. Compound VII was added before or at the same time as EtOH to avoid ethyl carbamate impurities from forming. The above EtOH solution was concentrated to a minimum volume (4-5 mL/g) using the batch concentrator under reduced pressure (THF levels should be <5% by GC), and EtOH (10 mL/g of compound VI) was back-added to give a total of 10 mL/g. The reaction was then heated at 50° Celsius for 9-19 hours or until HPLC shows that compound VI is less than 0.5 area %. The reaction was then cooled to room temperature, and sulfuric acid (H2SO4, 1.0 eq. to compound VI) was added via addition funnel to yield compound I-HS with the temperature usually exotherming at about 30° Celsius.

Example 1 Preparation of Crystalline Form (I-HS) (Method 1)

(S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide (0.500 g, 1.17 mmol) was dissolved in EtOH (2.5 mL) and cooled to about 5° Celsius. Concentrated sulfuric acid (0.0636 mL, 1.17 mmol) was added to the cooled solution and stirred for about 10 min, while warming to room temperature. Methyl tert-butyl ether (MTBE) (2 mL) was slowly added to the mixture, resulting in the product gumming out. EtOH (2.5 mL) was then added to the mixture and heated to about reflux until all solids were dissolved. Upon cooling to room temperature and stirring for about 1 hour, some solids formed. After cooling to about 5° Celsius, the solids were filtered and washed with MTBE. After filtration and drying at air for about 15 minutes, (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate was isolated as a solid.

Example 2 Preparation of Crystalline Form (I-HS) (Method 2)

Concentrated sulfuric acid (392 mL) was added to a solution of 3031 g of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide in 18322 mL EtOH to form the hydrogen sulfate salt. The solution was seeded with 2 g of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate and the solution was stirred at room temperature for at least 2 hours to form a slurry of the hydrogen sulfate salt. Heptane (20888 g) was added and the slurry was stirred at room temperature for at least 60 min. The slurry was filtered and the filter cake was washed with 1:1 heptane/EtOH. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius).

The dried hydrogen sulfate salt (6389 g from 4 combined lots) was added to a 5:95 w/w solution of water/2-butanone (total weight 41652 g). The mixture was heated at about 68° Celsius with stirring until the weight percent of ethanol was about 0.5%, during which time a slurry formed. The slurry was filtered, and the filter cake was washed with a 5:95 w/w solution of water/2-butanone. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius) to provide the crystalline form of (S)—N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate.

Example 3 Preparation of Amorphous Form AM(HS)

To a solution of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide (9.40 g, 21.94 mmol) in MeOH (220 mL) was slowly added sulfuric acid (0.1 M in MeOH, 219.4 mL, 21.94 mmol) at ambient temperature under rapid stirring. After 30 minutes, the reaction was first concentrated by rotary evaporator to near dryness, then on high vacuum for 48 h to provide amorphous form of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide sulfate (11.37 g, 21.59 mmol, 98.43% yield). LCMS (apci m/z 429.1, M+H).

PATENT

CN 107987082

PATENT

https://patents.google.com/patent/US20170281632A1/en

WO 2010/048314 discloses in Example 14A a hydrogen sulfate salt of (S)—N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide. WO 2010/048314 does not disclose the particular form of the hydrogen sulfate salt described herein when prepared according to the method of Example 14A in that document. In particular, WO 2010/048314 does not disclose crystalline form (l-HS) as described below.

(S)—N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide having the formula (I):

Figure US20170281632A1-20171005-C00001

Example 1 Preparation of Crystalline Form (I-HS) (Method 1)

(S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide (0.500 g, 1.17 mmol) was dissolved in EtOH (2.5 mL) and cooled to about 5° Celsius. Concentrated sulfuric acid (0.0636 mL, 1.17 mmol) was added to the cooled solution and stirred for about 10 min, while warming to room temperature. Methyl tert-butyl ether (MTBE) (2 mL) was slowly added to the mixture, resulting in the product gumming out. EtOH (2.5 mL) was then added to the mixture and heated to about reflux until all solids were dissolved. Upon cooling to room temperature and stirring for about 1 hour, some solids formed. After cooling to about 5° Celsius, the solids were filtered and washed with MTBE. After filtration and drying at air for about 15 minutes, (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidi n-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate was isolated as a solid.

Example 2 Preparation of Crystalline Form (I-HS) (Method 2)

Concentrated sulfuric acid (392 mL) was added to a solution of 3031 g of (S)—N-(5-((R)-2-(2, 5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1, 5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide in 18322 mL EtOH to form the hydrogen sulfate salt. The solution was seeded with 2 g of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate and the solution was stirred at room temperature for at least 2 hours to form a slurry of the hydrogen sulfate salt. Heptane (20888 g) was added and the slurry was stirred at room temperature for at least 60 min. The slurry was filtered and the filter cake was washed with 1:1 heptane/EtOH. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius).

The dried hydrogen sulfate salt (6389 g from 4 combined lots) was added to a 5:95 w/w solution of water/2-butanone (total weight 41652 g). The mixture was heated at about 68° Celsius with stirring until the weight percent of ethanol was about 0.5%, during which time a slurry formed. The slurry was filtered, and the filter cake was washed with a 5:95 w/w solution of water/2-butanone. The solids were then dried under vacuum at ambient temperature (oven temperature set at 15° Celsius) to provide the crystalline form of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate.

Example 3 Preparation of Amorphous Form AM(HS)

To a solution of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide (9.40 g, 21.94 mmol) in MeOH (220 mL) was slowly added sulfuric acid (0.1 M in MeOH, 219.4 mL, 21.94 mmol) at ambient temperature under rapid stirring. After 30 minutes, the reaction was first concentrated by rotary evaporator to near dryness, then on high vacuum for 48 h to provide amorphous form of (S)—N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide sulfate (11.37 g, 21.59 mmol, 98.43% yield). LCMS (apci m/z 429.1, M+H).

References

External links

Larotrectinib
Larotrectinib.svg
Identifiers
CAS Number
ChemSpider
UNII
Chemical and physical data
3D model (JSmol)
Patent ID Patent Title Submitted Date Granted Date
US8865698 Method of treatment using substituted pyrazolo[1, 5-a]pyrimidine compounds
2013-07-16
2014-10-21
US8513263 Substituted Pyrazolo[1, 5-a]Pyrimidine Compounds as TRK Kinase Inhibitors
2011-08-11
US2017165267 CRYSTALLINE FORM OF (S)-N-(5-((R)-2-(2, 5-DIFLUOROPHENYL)-PYRROLIDIN-1-YL)-PYRAZOLO[1, 5-A]PYRIMIDIN-3-YL)-3-HYDROXYPYRROLIDINE-1-CARBOXAMIDE HYDROGEN SULFATE
2017-01-05
US2017260589 POINT MUTATIONS IN TRK INHIBITOR-RESISTANT CANCER AND METHODS RELATING TO THE SAME
2016-10-26
US9676783 METHOD OF TREATMENT USING SUBSTITUTED PYRAZOLO[1, 5-A] PYRIMIDINE COMPOUNDS
2015-09-04
2016-08-11
Patent ID Patent Title Submitted Date Granted Date
US9447104 METHOD OF TREATMENT USING SUBSTITUTED PYRAZOLO[1, 5-a]PYRIMIDINE COMPOUNDS
2014-09-18
2015-01-01
US9127013 Method of treatment using substituted pyrazolo[1, 5-a] pyrimidine compounds
2015-01-14
2015-09-08
Patent ID Patent Title Submitted Date Granted Date
US9676783 METHOD OF TREATMENT USING SUBSTITUTED PYRAZOLO[1, 5-A] PYRIMIDINE COMPOUNDS
2015-09-04
2016-08-11
US2015073036 NOVEL NTRK1 FUSION MOLECULES AND USES THEREOF
2014-08-29
2015-03-12
US2017114067 METHOD OF TREATMENT USING SUBSTITUTED PYRAZOLO[1, 5-A] PYRIMIDINE COMPOUNDS
2017-01-05
US2016137654 CRYSTALLINE FORM OF (S)-N-(5-((R)-2-(2, 5-DIFLUOROPHENYL)-PYRROLIDIN-1-YL)-PYRAZOLO[1, 5-A]PYRIMIDIN-3-YL)-3-HYDROXYPYRROLIDINE-1-CARBOXAMIDE HYDROGEN SULFATE
2015-11-16
2016-05-19
US2015133429 METHOD OF TREATMENT USING SUBSTITUTED PYRAZOLO[1, 5-a] PYRIMIDINE COMPOUNDS
2015-01-14
2015-05-14
Patent ID Patent Title Submitted Date Granted Date
US2015366866 METHODS OF TREATING CHOLANGIOCARCINOMA
2014-01-17
2015-12-24
US8865698 Method of treatment using substituted pyrazolo[1, 5-a]pyrimidine compounds
2013-07-16
2014-10-21
US8513263 Substituted Pyrazolo[1, 5-a]Pyrimidine Compounds as TRK Kinase Inhibitors
2011-08-11
US2017165267 CRYSTALLINE FORM OF (S)-N-(5-((R)-2-(2, 5-DIFLUOROPHENYL)-PYRROLIDIN-1-YL)-PYRAZOLO[1, 5-A]PYRIMIDIN-3-YL)-3-HYDROXYPYRROLIDINE-1-CARBOXAMIDE HYDROGEN SULFATE
2017-01-05
US2017260589 POINT MUTATIONS IN TRK INHIBITOR-RESISTANT CANCER AND METHODS RELATING TO THE SAME
2016-10-26

///////////Larotrectinib, UNII:PF9462I9HX, ларотректиниб , 拉罗替尼 , ARRY-470, LOXO-101, PF9462I9HX, phase 3,  Array BioPharma, Loxo Oncology, National Cancer Institute, BAYER, orphan drug designation, breakthrough therapy designation

C1CC(N(C1)C2=NC3=C(C=NN3C=C2)NC(=O)N4CCC(C4)O)C5=C(C=CC(=C5)F)F.OS(=O)(=O)O

Fedratinib

$
0
0

Fedratinib structure.svgFedratinib.png

ChemSpider 2D Image | Fedratinib | C27H36N6O3SFigure imgf000121_0001

FEDRATINIB

SAR-302503; TG-101348, 6L1XP550I6, 936091-26-8 [RN], WHO 9707

Molecular Formula: C27H36N6O3S
Molecular Weight: 524.684 g/mol

FLT3, JAK2

http://www.ama-assn.org//resources/doc/usan/fedratinib.pdf

Fedratinib had been in phase III clincial trials by Sanofi for the treatment of myelofibrosis.

However, Sanofi had discontinued this research because of the safety issues. Orphan drug designation was assigned in the U.S. and in Japan for this indication. In 2017, the clinical hold was lifted in the U.S. by Impact Biomedicines.

MYELOFIBROSIS (MF), SANOFI , phase 3

Benzenesulfonamide, N-(1,1-dimethylethyl)-3-[[5-methyl-2-[[4-[2-(1-pyrrolidinyl)ethoxy]phenyl]amino]-4-pyrimidinyl]amino]-

N-tert-butyl-3-{[5-methyl-2-({4-[2-(pyrrolidin-1-yl)ethoxy]phenyl}amino)pyrimidin-4-yl]amino}benzenesulfonamide

N-tert-butyl-3-[[5-methyl-2-[4-(2-pyrrolidin-1-ylethoxy)anilino]pyrimidin-4-yl]amino]benzenesulfonamide

USAN (AB-104) FEDRATINIB
THERAPEUTIC CLAIM Antineoplastic
CHEMICAL NAMES
1. Benzenesulfonamide, N-(1,1-dimethylethyl)-3-[[5-methyl-2-[[4-[2-(1-
pyrrolidinyl)ethoxy]phenyl]amino]-4-pyrimidinyl]amino]-
2. N-tert-butyl-3-[(5-methyl-2-{4-[2-(pyrrolidin-1-yl)ethoxy]anilino}pyrimidin-4-
yl)amino]benzenesulfonamide

MOLECULAR FORMULA C27H36N6O3S
MOLECULAR WEIGHT 524.7
SPONSOR Sanofi
CODE DESIGNATIONS SAR302503; TG101348
CAS REGISTRY NUMBER……….936091-26-8

WHO 9707

TG-101348 , a dual-acting JAK2/FLT3 small molecule kinase inhibitor, has been evaluated in phase III clinical development at Sanofi (formerly known as sanofi-aventis) for the oral treatment of intermediate-2 or high risk primary myelofibrosis, post-polycythemia vera myelofibrosis or post-essential thrombocythemia myelofibrosis with splenomegaly. However, development of the compound has been discontinued due to safety issues.

In preclinical models of myeloproliferative diseases, TG-101348, administered orally, was shown to reduce V617F-expressing cell populations in a dose-dependent manner without adversely impacting normal hematopoiesis. The reduction of V617F- expressing cell populations correlated with improved survival and reduced morbidity. Orphan drug designation was assigned in the U.S. and in Japan for the treatment of secondary and primary myelofibrosis. In July 2010, TargeGen was acquired by Sanofi. In 2013, orphan drug designation was assigned by the FDA for the treatment of polycythemia vera.

Fedratinib is an orally bioavailable, small-molecule, ATP-competitive inhibitor of Janus-associated kinase 2 (JAK2) with potential antineoplastic activity. Fedratinib competes with JAK2 as well as the mutated form AK2V617F for ATP binding, which may result in inhibition of JAK2 activation, inhibition of the JAK-STAT signaling pathway, and the induction of tumor cell apoptosis. JAK2 is the most common mutated gene in bcr-abl-negative myeloproliferative disorders (MPDs); the mutated form JAK2V617F has a valine-to-phenylalanine modification at position 617 and plays a key role in tumor cell proliferation and survival.

Fedratinib has been used in trials studying the treatment and basic science of Solid Tumor, Myelofibrosis, Renal Impairment, Neoplasm Malignant, and Hepatic Impairment, among others.

Fedratinib (TG101348SAR302503) is an orally available inhibitor of Janus kinase 2 (JAK-2) developed for the treatment of patients with myeloproliferative diseases including myelofibrosis. Fedratinib acts as a competitive inhibitor of protein kinase JAK-2 with IC50=6 nM; related kinases FLT3 and RET are also sensitive, with IC50=25 nM and IC50=17 nM, respectively. Significantly less activity was observed against other tyrosine kinases including JAK3 (IC50=169 nM).[1] In treated cells the inhibitor blocks downstream cellular signalling (JAK-STAT) leading to suppression of proliferation and induction of apoptosis.

Myelofibrosis is a myeloid malignancy associated with anemia, splenomegaly, and constitutional symptoms. Patients with myelofibrosis frequently harbor JAK-STAT activating mutations that are sensitive to TG101348. Phase I trial results focused on safety and efficacy of Fedratinib in patients with high- or intermediate-risk primary or post–polycythemia vera/essential thrombocythemia myelofibrosis have been published in 2011.[2]

Fedratinib was originally discovered at TargeGen. In 2010, Sanofi-Aventis acquired TargeGen and continued development of fedratinib until 2013. In 2016, Impact Biomedicines acquired the rights to fedratinib from Sanofi and continued its development for the treatment of myelofibrosis and polycythemia vera. In January 2018, Celgene acquired Impact Biomedicines.[3]

Image result for Fedratinib SYNTHESIS

SYN

WO2007053452A1. +Bioorganic & Medicinal Chemistry Letters, 27(12), 2668-2673; 2017

Condensation of 3-bromo-N-tertbutylbenzylsulfonamide with 2-chloro-5-methyl-pyrimidin-4-ylamine  in the presence of Pd2(dba)3, Xantphos, Cs2CO3 in refluxing dioxane gives sulfonamide derivative , which is coupled with 4-[2-pyrrolidin-1-yl-ethoxy]phenylamine  in AcOH at 150°C to provide the title compound

PRODUCT PATENT

WO2007053452A1.

Inventors Jon Jianguo CaoJohn HoodDan LohseChi Ching MakPherson Andrew McGlenn NoronhaVed PathakJoel RenickRichard M. SollBinqi ZengLess «
Applicant Targegen, Inc.

https://encrypted.google.com/patents/WO2007053452A1?cl=en

EXAMPLE 90. 7V-fe^-Butyl-3-{5-methyl-2-14-(2-pyrrolidm-l-yl-ethoxy)-phenylaminol- pyrimidin-4-ylaminol-benzenesuIfonamide (Compound LVII)

Figure imgf000121_0001

LVII

[0203] A mixture of intermediate 33 (0.10 g, 0.28 mmol) and 4-(2-pyrrolidin-l-yl- ethoxy)-phenylamine (0.10 g, 0.49 mmol) in acetic acid (3 mL) was sealed in a microwave reaction tube and irradiated with microwave at 150 °C for 20 min. After cooling to room temperature, the cap was removed and the mixture concentrated. The residue was purified by HPLC and the corrected fractions combined and poured into saturated NaHCO3 solution (30 mL). The combined aqueous layers were extracted with EtOAc (2 x 30 mL) and the combined organic layers washed with brine, dried over anhydrous Na2SO4and filtered. The filtrate was concentrated and the resulting solid dissolved in minimum atnount of EtOAc and hexanes added until solid precipitated. After filtration, the title compound was obtained as a white solid (40 mg, 27%).

[0204] 1H NMR (500 MHz, DMSO-d6): δ 1.12 (s, 9H), 1.65-1.70 (m, 4H), 2.12 (s, 3H), 2.45-2.55 (m, 4H), 2.76 (t, J= 5.8 Hz, 2H), 3.99 (t, J= 6.0 Hz, 2H), 6.79 (d, J= 9.0 Hz, 2H), 7.46-7.53 (m, 4H), 7.56 (s, IH), 7.90 (s, IH), 8.10-8.15 (m, 2H), 8.53 (s, IH), 8.77 (s, IH). MS (ES+): m/z 525 (M+H)+. it ιr

PATENTS

WO 2013059548

PAPER

Bioorganic & Medicinal Chemistry Letters, 27(12), 2668-2673; 2017

PATENT

WO 2012061833

The compound and the pharmaceutical compositions described herein can be used for treating or delaying development of myelofibrosis in a subject. N-teft-Butyl-3-[(5-methyl-2-{ [4- (2-pyrrolidin-l-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide has the following chemical structure:

Figure imgf000018_0001

Example 4. Synthesis of TG101348

Example 4.1 N-fer^-Butyl-3-(2-chloro-5-methyl-pyrimidin-4-ylamino)-benzenesulfonamide

(Intermediate)

Example 4.1(a)

Figure imgf000053_0001

1 2 Intermediate

[0162] A mixture of 2-chloro-5-methyl-pyrimidin-4-ylamine (1) (0.4 g, 2.8 mmol), 3-bromo-N- teft-butyl-benzenesulfonamide (2) (1.0 g, 3.4 mmol), Pd2(dba¾ (0.17 g, 0.19 mmol), Xantphos (0.2 g, 3.5 mmol) and cesium carbonate (2.0 g, 6.1 mmol) was suspended in dioxane (25 mL) and heated at reflux under the argon atmosphere for 3 h. The reaction mixture was cooled to room temperature and diluted with DCM (30 mL). The mixture was filtered and the filtrate

concentrated in vacuo. The residue was dissolved in EtOAc and hexanes added until solid precipitated. After filtration, the title compound (1.2 g, 98%) was obtained as a light brown solid. It was used in the next step without purification. MS (ES+): m/z 355 (M+H)+.

Example 4.1(b)

Figure imgf000053_0002

SM2 Intermediate[0163] The Intermediate was synthesized from 2,4-dichloro-5-methylpyrimidine (SMI) and N-t- butyl-3-aminobenzenesulfonamide (SM2) in the following steps: (1) Mix MeOH (6.7UOa) and SMI (Combi Blocks) (UOa); (2) Add SM2 (1.15UOa, 082eq) and H20 (8.5UOa); (3) Heat 45°C, 20h, N2, IPC CPL SM2<2%; (4) Cool 20°C; (5) Centrifuge, N2; (6) Wash H20 (2.1UOa) + MeOH (1.7UOa); (7) Mix solid in H20 (4.3UOa) + MeOH (3.4UOa); (8) Centrifuge, N2; (9) Wash H20 (2.1UOa) + MeOH (1.7UOa); and (10) Dry 45°C, vacuum, 15h. Obtained

Intermediate, mass 49.6kg (UOb); Yield 79%; OP: 99.6%.

Example 4.2 N-½ri-Butyl-3-[(5-methyl-2-{ [4-(2-pyrrolidin-l- ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide

Figure imgf000054_0001

Intermediate TG101348

Example 4.2(a)

[0164] A mixture of N-ieri-Butyl-3-(2-chloro-5-methyl-pyrimidin-4-ylamino)- benzenesulfonamide (Intermediate) (0.10 g, 0.28 mmol) and 4-(2-pyrrolidin-l-yl-ethoxy)- phenylamine (3) (0.10 g, 0.49 mmol) in acetic acid (3 mL) was sealed in a microwave reaction tube and irradiated with microwave at 150 °C for 20 min. After cooling to room temperature, the cap was removed and the mixture concentrated. The residue was purified by HPLC and the corrected fractions combined and poured into saturated NaHCC^ solution (30 mL). The combined aqueous layers were extracted with EtOAc (2 x 30 mL) and the combined organic layers washed with brine, dried over anhydrous Na2S04 and filtered. The filtrate was concentrated and the resulting solid dissolved in minimum amount of EtOAc and hexanes added until solid precipitated. After filtration, the title compound was obtained as a white solid (40 mg, 27%). ]H NMR (500 MHz, DMSO-d6): δ 1.12 (s, 9H), 1.65-1.70 (m, 4H), 2.12 (s, 3H), 2.45-2.55 (m, 4H), 2.76 (t, /=5.8 Hz, 2H), 3.99 (t, 7=6.0 Hz, 2H), 6.79 (d, 7=9.0 Hz, 2H), 7.46-7.53 (m, 4H), 7.56 (s, 1H), 7.90 (s, 1H), 8.10-8.15 (m, 2H), 8.53 (s, 1H), 8.77 (s, 1H). MS (ES+): m/z 525 (M+H)+.

Example 4.2(b)

[0165] N-½ri-Butyl-3-[(5-methyl-2-{ [4-(2-pyrrolidin-l-ylethoxy)phenyl]amino}pyrimidin-4- yl)amino]benzenesulfonamide dihydrochloride monohydrate was prepared from 4-[2-(l- pyrrolidinyl)ethoxy] aniline dihydrochloride (SM3) and Intermediate following steps (A) and (B).

[0166] Step (A), preparation of free base of SM3 (3) from SM3, comprised steps (1) – (9): (1) Solubilize NaOH (0.42UOb) in H20 (9UOb); (2) Cool <20°C, N2; (3) Add TBME (6UOb) then SM3 (Malladi Drugs) (1.06UOb); (4) Mix >20mn then stop; (5) Drain Aq Ph then extract by TBME (3UOb); (6) Combine Or Ph; (7) Concentrate, vacuum, T<40°C, to an Oil; (8) Solubilize in IPA (2.5UOb); and (9) Calculate dry extract 23%.

[0167] Step (B) comprised the steps (1) – (6): (1) Mix IPA (10.5UOb) and Intermediate (UOb); (2) Add free base of SM3 (0.75UOb, 1.33eq/ interm); (3) add HC1 cone (0.413UOb); (4) Heat 70°C, 20h, N2, IPC CPL Interm<2%; (5) Cool <20°C; (2) Centrifuge, N2; (3) Wash IPA (3UOb); (4) Dry 50°C, vacuum, 26h; (5) De-lump in Fitzmill; and (6) polybag (x2) / poly drum. Obtained TG101348 dihydrochloride monohydrate, mass 83.8kg; Yield 98%; OP: 99.5%. Example 5 Capsule Form of TG101348 and Process of Making TG101348

PATENT

WO 2010017122

US 2007259904

WO 2007053452

Paper

JAK inhibitors: pharmacology and clinical activity in chronic myeloprolipherative neoplasms.

Treliński J, Robak T.

Curr Med Chem. 2013;20(9):1147-61.

JAK2 inhibitors for myelofibrosis: why are they effective in patients with and without JAK2V617F mutation?

Santos FP, Verstovsek S.

Anticancer Agents Med Chem. 2012 Nov;12(9):1098-109. Review.

Octa-arginine mediated delivery of wild-type Lnk protein inhibits TPO-induced M-MOK megakaryoblastic leukemic cell growth by promoting apoptosis.

Looi CY, Imanishi M, Takaki S, Sato M, Chiba N, Sasahara Y, Futaki S, Tsuchiya S, Kumaki S.

PLoS One. 2011;6(8):e23640. doi: 10.1371/journal.pone.0023640. Epub 2011 Aug 10

PATENT

us2007191405

Example 90 N-tert-Butyl-3-{5-methyl-2-[4-(2-pyrrolidin-1-yl-ethoxy)-phenylamino]-pyrimidin-4-ylamino}-benzenesulfonamide (Compound LVII)

Figure US20070191405A1-20070816-C00156

A mixture of intermediate 33 (0.10 g, 0.28 mmol) and 4-(2-pyrrolidin-1-yl-ethoxy)-phenylamine (0.10 g, 0.49 mmol) in acetic acid (3 mL) was sealed in a microwave reaction tube and irradiated with microwave at 150° C. for 20 min. After cooling to room temperature, the cap was removed and the mixture concentrated. The residue was purified by HPLC and the corrected fractions combined and poured into saturated NaHCOsolution (30 mL). The combined aqueous layers were extracted with EtOAc (2×30 mL) and the combined organic layers washed with brine, dried over anhydrous Na2SOand filtered. The filtrate was concentrated and the resulting solid dissolved in minimum amount of EtOAc and hexanes added until solid precipitated. After filtration, the title compound was obtained as a white solid (40 mg, 27%).

1H NMR (500 MHz, DMSO-d6): δ 1.12 (s, 9H), 1.65-1.70 (m, 4H), 2.12 (s, 3H), 2.45-2.55 (m, 4H), 2.76 (t, J=5.8 Hz, 2H), 3.99 (t, J=6.0 Hz, 2H), 6.79 (d, J=9.0 Hz, 2H), 7.46-7.53 (m, 4H), 7.56 (s, 1H), 7.90 (s, 1H), 8.10-8.15 (m, 2H), 8.53 (s, 1H), 8.77 (s, 1H). MS (ES+): m/z 525 (M+H)+.

Example 76 N-tert-Butyl-3-(2-chloro-5-methyl-pyrimidin-4-ylamino)-benzenesulfonamide (Intermediate 33)

Figure US20070191405A1-20070816-C00142

A mixture of 2-chloro-5-methyl-pyrimidin-4-ylamine (0.4 g, 2.8 mmol), 3-bromo-N-tert-butyl-benzenesulfonamide (1.0 g, 3.4 mmol), Pd2(dba)(0.17 g, 0.19 mmol), Xantphos (0.2 g, 3.5 mmol) and cesium carbonate (2.0 g, 6.1 mmol) was suspended in dioxane (25 mL) and heated at reflux under the argon atmosphere for 3 h. The reaction mixture was cooled to room temperature and diluted with DCM (30 mL). The mixture was filtered and the filtrate concentrated in vacuo. The residue was dissolved in EtOAc and hexanes added until solid precipitated. After filtration, the title compound (1.2 g, 98%) was obtained as a light brown solid. It was used in the next step without purification. MS (ES+): m/z 355 (M+H)+.

PATENT

https://encrypted.google.com/patents/US20090286789

    Example 90N-tert-Butyl-3-{5-methyl-2-[4-(2-pyrrolidin-1-yl-ethoxy)-phenylamino]-pyrimidin-4-ylamino}-benenesulfonamide (Compound LVII)
  • [0308]
    Figure US20090286789A1-20091119-C00143
  • [0309]
    A mixture of intermediate 33 (0.10 g, 0.28 mmol) and 4-(2-pyrrolidin-1-yl-ethoxy)-phenylamine (0.10 g, 0.49 mmol) in aeetie acid (3 mL) was sealed in a microwave reaction tube and irradiated with microwave at 150° C. for 20 min. After cooling to room temperature, the cap was removed and the mixture concentrated. The residue was purified by HPLC and the corrected fractions combined and poured into saturated NaIICOsolution (30 mL). The combined aqueous layers were extracted with EtOAc (2×30 mL) and the combined organic layers washed with brine, dried over anhydrous Na2SOand filtered. The filtrate was concentrated and the resulting solid dissolved in minimum amount of EtOAc and hexanes added until solid precipitated. After filtration, the title compound was obtained as a white solid (40 mg, 27%).
  • [0310]
    1H NMR (500 MHz, DMSO-d6): δ 1.12 (s, 9H), 1.65-1.70 (m, 4H), 2.12 (s, 3H), 2.45-2.55 (m, 4H), 2.76 (t, J=5.8 Hz, 2H), 3.99 (t, J=6.0 Hz, 2H), 6.79 (d, J=9.0 Hz, 2H), 7.46-7.53 (m, 4H), 7.56 (s, 1H), 7.90 (s, 1H), 8.10-8.15 (m, 2H), 8.53 (s, 1H), 8.77 (s, 1H). MS (ES+): m/z 525 (M+H)+.

PATENT

WO 2015117053

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015117053&recNum=4&maxRec=26794&office=&prevFilter=&sortOption=&queryString=FP%3A%28%22cancer%22%29+AND+EN_ALL%3Anmr&tab=PCTDescription

References

  1. Jump up^ Pardanani, A.; Hood, J.; Lasho, T.; Levine, R. L.; Martin, M. B.; Noronha, G.; Finke, C.; Mak, C. C.; Mesa, R.; Zhu, H.; Soll, R.; Gilliland, D. G.; Tefferi, A. (2007). “TG101209, a small molecule JAK2-selective kinase inhibitor potently inhibits myeloproliferative disorder-associated JAK2V617F and MPLW515L/K mutations”. Leukemia21 (8): 1658–1668. doi:10.1038/sj.leu.2404750PMID 17541402.
  2. Jump up^ Pardanani, A.; Gotlib, J. R.; Jamieson, C.; Cortes, J. E.; Talpaz, M.; Stone, R. M.; Silverman, M. H.; Gilliland, D. G.; Shorr, J.; Tefferi, A. (2011). “Safety and Efficacy of TG101348, a Selective JAK2 Inhibitor, in Myelofibrosis”Journal of Clinical Oncology29 (7): 789–796. doi:10.1200/JCO.2010.32.8021PMC 4979099Freely accessiblePMID 21220608.
  3. Jump up^ “Celgene to Acquire Impact Biomedicines, Adding Fedratinib to Its Pipeline of Novel Therapies for Hematologic Malignancies (NASDAQ:CELG)”ir.celgene.com. Retrieved 2018-01-18.

External links

Cited Patent Filing date Publication date Applicant Title
WO2009073575A2 * Nov 28, 2008 Jun 11, 2009 Oregon Health & Science University Methods for treating induced cellular proliferative disorders
US20090088410 * Dec 5, 2008 Apr 2, 2009 Celgene Corporation Methods for the treatment and management of myeloproliferative diseases using 4-(amino)-2-(2,6-dioxo(3-piperidyl)-isoindoline-1,3-dione in combination with other therapies
US20090286789 * Oct 14, 2008 Nov 19, 2009 Targegen, Inc. Bi-Aryl Meta-Pyrimidine Inhibitors of Kinases
Reference
1 * See also references of EP2635282A4
Citing Patent Filing date Publication date Applicant Title
US8604042 Aug 24, 2010 Dec 10, 2013 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
Patent ID Patent Title Submitted Date Granted Date
US8748428 USE OF A PKC INHIBITOR
2011-10-06
US8133900 Use of bi-aryl meta-pyrimidine inhibitors of kinases
2009-11-19
2012-03-13
US8138199 Use of bi-aryl meta-pyrimidine inhibitors of kinases
2009-11-05
2012-03-20
US2016332993 DIAMINOPYRIMIDINE BENZENESULFONE DERIVATIVES AND USES THEREOF
2015-02-02
US7825246 Bi-aryl meta-pyrimidine inhibitors of kinases
2007-11-08
2010-11-02
Patent ID Patent Title Submitted Date Granted Date
US2013243853 COMPOSITIONS AND METHODS FOR TREATING MYELOFIBROSIS
2013-05-06
2013-09-19
US9198911 Methods for Treating Hair Loss Disorders
2013-05-02
2014-03-06
US9089574 ANTIVIRAL JAK INHIBITORS USEFUL IN TREATING OR PREVENTING RETROVIRAL AND OTHER VIRAL INFECTIONS
2012-11-30
2014-11-06
US2014170157 METHOD OF SELECTING THERAPEUTIC INDICATIONS
2012-06-15
2014-06-19
US2011269721 METHODS OF TREATING THALASSEMIA
2011-11-03
Patent ID Patent Title Submitted Date Granted Date
US2016264732 BLOCK COPOLYMERS FOR STABLE MICELLES
2016-03-10
2016-09-15
US9763866 METHODS FOR TREATING HAIR LOSS DISORDERS
2016-03-10
2016-09-08
US9730877 METHODS FOR TREATING HAIR LOSS DISORDERS
2015-08-07
2016-03-03
US9662332 ANTIVIRAL JAK INHIBITORS USEFUL IN TREATING OR PREVENTING RETROVIRAL AND OTHER VIRAL INFECTIONS
2015-07-24
2016-02-25
US2014357557 CYCLODEXTRIN-BASED POLYMERS FOR THERAPEUTIC DELIVERY
2014-05-30
2014-12-04
Patent ID Patent Title Submitted Date Granted Date
US8604042 BI-ARYL META-PYRIMIDINE INHIBITORS OF KINASES
2011-09-01
US8791100 ARYL BENZYLAMINE COMPOUNDS
2011-08-04
US7528143 Bi-aryl meta-pyrimidine inhibitors of kinases
2007-08-16
2009-05-05
US2016346408 IRON STABILIZED MICELLES AS MAGNETIC CONTRAST AGENTS
2016-05-26
US2016303205 Combination Therapies for Lysosomal Storage Diseases
2016-04-13
Fedratinib
Fedratinib structure.svg
Names
IUPAC name
Ntert-Butyl-3-{5-methyl-2-[4-(2-pyrrolidin-1-yl-ethoxy)-phenylamino]-pyrimidin-4-ylamino}-benzenesulfonamide
Other names
SAR302503; TG101348
Identifiers
3D model (JSmol)
Properties
C27H36N6O3S
Molar mass 524.68 g·mol−1
Density 1.247 ± 0.06 g/cm3
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
Infobox references

////////////////FEDRATINIB, SAR-302503,  TG-101348, SANOFI, PHASE 3, TG101348,  SAR302503, TG 101348, SAR 302503, Orphan drug designation 

CC1=CN=C(N=C1NC2=CC(=CC=C2)S(=O)(=O)NC(C)(C)C)NC3=CC=C(C=C3)OCCN4CCCC4

Remimazolam

$
0
0

Remimazolam.svgChemSpider 2D Image | Remimazolam | C21H19BrN4O2GHUYIIGPWBMOGY-KRWDZBQOSA-N.png

Figure imgf000062_0002

Remimazolam

  • Molecular FormulaC21H19BrN4O2
  • Average mass439.305 Da
3-[(4S)-8-bromo-1-methyl-6-(2-pyridyl)-4H-imidazo[1,2-a][1,4]benzodiazepin-4-yl]propionic acid methyl ester
CNS-7056
methyl 3-[(4S)-8-bromo-1-methyl-6-(2-pyridyl)-4H-imidazo[1,2-a][1,4]benzodiazepin-4-yl]propanoate
methyl 3-[(4S)-8-bromo-1-methyl-6-pyridin-2-yl-4H-imidazo[1,2-a][1,4]benzodiazepin-4-yl]propanoate
methyl 3-[(7S)-12-bromo-3-methyl-9-(pyridin-2-yl)-2,5,8-triazatricyclo[8.4.0.02,6]tetradeca-1(14),3,5,8,10,12-hexaen-7-yl]propanoate
MFCD18633229
UNII:7V4A8U16MB

CAS 308242-62-8 [RN]

PHASE 3, PAION, Anesthesia

4H-Imidazo[1,2-a][1,4]benzodiazepine-4-propanoic acid, 8-bromo-1-methyl-6-(2-pyridinyl)-, methyl ester, (4S)-

7V4A8U16MB
9232
Methyl 3-[(4S)-8-bromo-1-methyl-6-(2-pyridinyl)-4H-imidazo[1,2-a][1,4]benzodiazepin-4-yl]propanoate
Methyl 3-[(4S)-8-bromo-l-methyl-6-(2-pyridinyl)-4H-imidazo[l,2- a] [ 1 ,4]benzodiazepin-4-yl]propanoate
methyl 3-[(4S)-8-bromo-2-methyl-6-pyridin-2-yl-4H-imidazo[1,2-a][1,4]benzodiazepin-4-yl]propanoate
D0L5KU; GTPL8442; SCHEMBL13862667; Short-acting sedatives, Therasci; CNS-7056B; CNS-7056X
  1. CNS 7056
  2. methyl 3-(8-bromo-1-methyl-6-(2-pyridinyl)-4H-imidazo(1,2-a)(1,4)benzodiazepin-4-yl)propanoate
  3. ONO 2745
  4. ONO-2745
  5. ONO2745

Remimazolam[1] (CNS-7056) is a benzodiazepine derivative drug, developed by PAION, in collaboration with Japanese licensee Ono Pharmaceutical as an alternative to the short-acting imidazobenzodiazepine midazolam, for use in induction of anaesthesia and conscious sedation for minor invasive procedures. Remimazolam was found to be both faster acting and shorter lasting than midazolam, and human clinical trials showed a faster recovery time and predictable, consistent pharmacokinetics, suggesting some advantages over existing drugs for these applications.[2][3]

Remimazolam (CNS-7056) is a water-soluble, rapid and short-acting GABA (A) benzodiazepine (BZ) site receptor agonist in phase III trials at PAION as procedural sedation in patients undergoing colonoscopy or diagnostic endoscopy of the upper gastrointestinal tract, and also with patients undergoing bronchoscopy.

PAION AG and its subsidiary PAION Inc, following its acquisition of CeNeS Pharmaceuticals (following CeNeS’ acquisition of TheraSci ), and licensees Mundipharma , Yichang Humanwell Pharmaceutical , Pendopharm , Cosmo and R-Pharm are developing remimazolam, the lead from a series of short-acting GABA A receptor agonists, as an iv sedative and/or anesthetic for potential use in day case surgical and non-surgical procedures

Image result for remimazolam

(Salt/Parent)
1
Remimazolam [INN]
308242-62-8
2D chemical structure of 308242-62-8
MW: 439.3111
2
Remimazolam besilate
1001415-66-2
2D chemical structure of 1001415-66-2
MW: 597.4875
3
Remimazolam tosylate
1425904-79-5
2D chemical structure of 1425904-79-5
MW: 611.5143

Trials

Phase I[4] and Ib[5] dose-finding studies for procedural sedation with patients recovering faster from remimazolam than midazolam. Phase II trials comparing remimazolam to the standard anesthesia protocols for cardiac surgery and colonoscopy were presented at major conferences in October 2014.[6]

A phase IIa trial comparing remimazolam to midazolam for upper endoscopy was published in December 2014, finding a similar safety profile.[7] Remimazolam was originally discovered in the late 1990s at Glaxo Wellcome in their labs in Research Triangle Park, NC.

BY CHENGDU

WO-2018103119

Novel crystalline forms of hydrobromate salt of remimazolam , processes for their preparation and compositions comprising them are claimed.

Remazolam, whose structure is shown in formula (I), has the chemical name 3-[(4S)-8-bromo-1-methyl-6-(2-pyridyl)-4H-imidazole [1,2] -a] methyl [1,4]benzodiazepin- 4-yl]propanoate.
This compound is currently known as a CNS (Central Nervous System) inhibitor and has sedative, hypnotic, anxiolytic, muscle relaxing, and anticonvulsant effects. It is currently used intravenously in the following clinical treatment programs: preoperative sedation, anxiolysis and forgetfulness during surgery; awake sedation during short-term diagnosis, surgery, or endoscopic procedures; and administration of other anesthetics and analgesia Before and/or at the same time as a component for induction and maintenance of general anesthesia; ICU sedation and the like. It is reported in patent application CN101501019 that the free base stability of the compound is poor, and it is only suitable for storage at a low temperature of 5°C. Under conditions of 40°C/75% relative humidity (open), the sample is deliquescent and discolored, and the content is significantly reduced.
Due to the stability problem of the free base of the compound, researchers from various countries have studied the salts of the compound. For example, patent applications CN101501019B and WO2008/007081A1 respectively report the besylate and ethanesulfonate of the compound of formula (I). And shows that the above salts have good thermal stability, low hygroscopicity, and high water solubility, and that CN104968348A clearly states that the above benzenesulfonates and ethanesulfonates are the most preferred compounds of formula (I). Salts.
Immediately afterwards, CN 103221414B proposes a toxilate salt of a compound of formula (I) and indicates that the toxitic acid salt is less toxic than benzene sulphonate, and the thermal stability, water solubility and the like of certain crystal forms are even higher. For good.
To sort out the existing technology information, you can draw the following related content (Table 1):
Table 1
From the above table, it can be seen that regardless of whether it is a free base of remazolam or a known salt derivative of remazolam, the water solubility is not higher than 11 mg/ml, and only in the slightly soluble range, which will increase The safety risk of its use in clinical use requires resolving and dissolving for a long time during clinical reconstitution. It may also leave insoluble materials, resulting in inaccurate drug dosage and potential safety risks. In addition, it is used for general anesthesia. Indications with a large demand will increase the amount of diluent and cause extreme inconvenience for clinical use. Therefore, the solubility of the known salt derivatives of remazolam is a big disadvantage and needs to be further improved.
The raw material remazolam of the compound of the formula (I) used in the present invention can be obtained by purchasing a commercially available product or can be prepared according to a known method (for example, patent US200,700,934,75A, etc.).
Example 1 Preparation of Form III Hydrobromide Salt of Compound of Formula (I)
Accurately weigh 1.8 g of the compound of formula (I) into a 100 mL three-necked flask, add 8.2 mL of isopropanol and stir to dissolve it completely, then dissolve 0.83 g of 47% aqueous hydrobromic acid in 6.3 mL of isopropanol and drip To the solution of the compound of formula (I) in isopropanol, the crystals were stirred, filtered, and dried at 55°C under reduced pressure to give the hydrobromide salt of the compound of formula (I).
The X-ray diffraction pattern of this crystal is shown in FIG. 1, the DSC and TGA patterns are shown in FIG. 2, and the melting point is 163 DEG C. It is defined that the crystal form is the hydrobromide III crystal form of the compound of Formula (I).

PATENT

WO0069836

Family members of remimazolam’s product case WO0069836 , have production in most of the EU states until May 2020 and expire in the US in April 2020.

PRODUCT PATENT

WO 2000069836

https://encrypted.google.com/patents/WO2000069836A1?cl=en

Inventors Paul L. FeldmanDavid Kendall JungIstvan KaldorGregory J. PacofskyJeffrey A. StaffordJeffrey H. TidwellLess «
Applicant Glaxo Group Limited

Example Ic-8

Methyl 3-[(4S)-8-bromo-l-methyl-6-(2-pyridinyl)-4H-imidazo[l,2- a] [ 1 ,4]benzodiazepin-4-yl]propanoate

Figure imgf000062_0002

A solution of the C7-bromo-benzodiazepine Ex 1-10 (7.31 g, 18.2 mmol) in THF (21 mL) was added to a suspension of NaH (870 mg of 60% oil dispersion, 21.8 mmol) in THF (70 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 30 min, warmed to room temperature and stirred for 30 min, then cooled to 0 °C. Bis- morpholinophosphorochloridate (6.48 g, 25.5 mmol) was added, the mixture was allowed to warm to room temperature over 4.5 h, and the mixture was filtered with additional THF (ca. 10 mL). A mixture of the filtrate and DL-l-amino-2-propanol (2.80 mL, 36.4 mmol) was stirred at room temperature for 18 h and concentrated under reduced pressure. The residue was diluted with EtOAc (ca. 250 mL), washed with saturated aqueous NaHCO3 (1 x 75 mL), H2O (2 x 75 mL), saturated aqueous NaCl (1 x 75 mL), dried (Na SO ), and concentrated under reduced pressure. Purification by flash chromatography, elution with 19:1 EtOAc-MeOH, gave 3.06 g

(37%) of the adduct as a foam; ESIMS 459 (M+H, base).

A mixture of DMSO (1.88 mL, 26.6 mmol) and oxalyl chloride (1.16 mL, 13.3 mmol) in CH2C12 (40 mL) was stirred at -78 °C for 30 min. A solution of the alcohol prepared above (3.05 g, 6.64 mmol) in CH2C1 (26 mL) was added. The reaction mixture was warmed to -15 °C and stirred 1 h, cooled to -78 °C, treated with

E-3N (5.55 mL, 39.9 mmol), and allowed to warm to room temperature over 3 h. The mixture was diluted with EtOAc (ca. 500 mL), washed with saturated aqueous NaHCO3 (1 x 100 mL), H2O (1 x 100 mL), saturated aqueous NaCl (1 x 100 mL), dried (Na SO ), and concentrated under reduced pressure to give a foam. A mixture of this foam and a catalytic amount ofp-toluenesulfonic acid was stirred at room temperature for 18h, neutralized by the addition of saturated aqueous NaHCO3 and diluted with EtOAc (ca. 500 mL). The layers were separated and the organic phase was washed with saturated aqueous NaHCO3 (1 x 100 mL), H2O (2 x 100 mL), saturated aqueous NaCl (1 x 100 mL), dried (Na SO ), and concentrated under reduced pressure. Purification by flash chromatography, elution with 19: 1 EtOAc-

MeOH, gave 2.56 g (88%) of Ic-8 as a foam; 1H NMR (400 MHz, CDC13) δ 8.57 (d, J = 4.6 Hz, lH), 8.17 (d J = 7.8 Hz, IH), 7.79 (dd, J = 7.7, 6.2 Hz, IH), 7.71 (dd, J = 8.6, 2.2 Hz, IH), 7.64 (d, J – 2.2 Hz, IH), 7.34 (dd, J = 7.5, 5.0 Hz, IH), 7.30 (d, J = 8.6 Hz, IH), 6.86 (s, IH), 4.05 (m, 1 H), 3.67 (s, 3H), 2.80 (comp, 4H), 2.34 (s, 3H); ESIMS 461 (M+Na, base), 439 (M+H); Anal, calcd. for C2]H19BrN4O2-0.25 H2O: C,

58.63; H, 4.43; N, 12.62. Found: C, 56.88; H, 4.43; N, 12.23.

Example lc-8 was formulated in an aqueous vehicle at a concentration of 10 mg/ml. Accordingly, 10 mg of compound (and 9 mg NaCl) was dissolved in 0.63 ml of 0.1 N HCl. Slowly and while stirring, 0.37 ml of 0.1 N NaOH was added. Adjustments are made to the dose volume depending on the dose being administered.

PATENT

CN 103232454

https://patents.google.com/patent/CN103232454A/en

The compounds of the following formula I:

[0003]

Figure CN103232454AD00051

Wherein R1 is bromine, R2 and R3 is methyl, [0004] because it contains the specific configuration, W000 / 69836 reported in the compound (60 specification Example Ic-8) is a short-acting central nervous system (CNS) to suppress agents, including having a sedative-hypnotic, anxiolytic, muscle relaxant and anticonvulsant effect.They can be used for intravenous administration in the clinical treatment: preoperative sedation, such as during surgery, and forgetting anxiolytic purposes; in short diagnostic, operative or endoscopic conscious sedation during the procedure; administration of other anesthetics and analgesics before and / or simultaneously, as a component for the induction and maintenance of general anesthesia in; the ICU sedation, according CN101501019A (PA10N, application No. CN200780028964.5) reports, free base of the compound is not very stable, only suitable stored at low temperatures 5 ° C, at 40 ° C / 75% relative humidity (open) condition, the sample storage deliquescence, to the orange color turned yellow, with respect to the initial content and significantly reduced the content of the display. Thus the synthesis of salts of compounds of formula It (the I), hoping to increase the chemical stability thereof, for use in the preparation of medicaments.

[0005] existing CN101501019A and US20100075955A1 (TILBR00K) reported the benzenesulfonate salt of a compound of formula I, ethanesulfonate.CN102964349A (Henry, Application No. 201110456864.0) reported for compounds of formula ITosylate.

[0006] have reported the presence of a compound of formula I or a salt thereof concerns stability, which is disadvantageous for these compounds used in the clinical treatment of related diseases.

HPLC method [A]:

[0022] According to Chinese Pharmacopoeia 2010 Appendix VD High Performance Liquid Chromatography;

[0023] using Daicel Chrialcel OJ-H (5 μ m) 4.6 X 250mm using chiral chromatographic columns (guard column, if necessary Daicel Chrialcel OJ-H column analysis protected 5 μ m4.0 X IOmm, which is Japan Series Cat (Daicel ) brand), hexane: ethanol = 93: 7 (v / v) as the mobile phase, a flow rate of 1.0ml / min, column temperature 40 ° C, detection wavelength 225nm;

Bulk drug preparation of the present invention: [0204] Example 1

[0205] Preparation Example 4 taking the resulting compound of formula I lg, were added to 8ml of ethanol at 50 ° C – lactic acid – water (volume ratio of the three 45: 2: 53) mixed solution was stirred to dissolve; filtration, the filtrate was 5 ° C was allowed to stand at a temperature of 10~12 hours recrystallized, crystals were filtered off, 40 ° C and dried in vacuo; the above operation was repeated once, to give a compound of formula I may be formulated bulk drug used as a pharmaceutical formulation, was recrystallized twice yield rate of 86.1%.Chromatographic purity of product by HPLC 99.22% [B]; R & lt isomer impurity content of 0.39% relative peak area ratio (I / Ix) = 255 HPLC [Method A].

PATENT

EP 2305647

PATENT

WO 2011032692,

See also

References

  1. Jump up^ EP Patent 1183243
  2. Jump up^ Rogers WK, McDowell TS (December 2010). “Remimazolam, a short-acting GABA(A) receptor agonist for intravenous sedation and/or anesthesia in day-case surgical and non-surgical procedures”. IDrugs : the Investigational Drugs Journal13 (12): 929–37. PMID 21154153.
  3. Jump up^ Saari TI, Uusi-Oukari M, Ahonen J, Olkkola KT (March 2011). “Enhancement of GABAergic activity: neuropharmacological effects of benzodiazepines and therapeutic use in anesthesiology”. Pharmacological Reviews63 (1): 243–67. doi:10.1124/pr.110.002717PMID 21245208.
  4. Jump up^ “A placebo- and midazolam-controlled phase I single ascending-dose study evaluating the safety, pharmacokinetics, and pharmacodynamics of remimazolam (CNS 7056): Part I. Safety, efficacy, and basic pharmacokinetics”. Anesth. Analg.accessdate =115: 274–83. Aug 2012. doi:10.1213/ANE.0b013e31823f0c28PMID 22190555.
  5. Jump up^ “A phase Ib, dose-finding study of multiple doses of remimazolam (CNS 7056) in volunteers undergoing colonoscopy”. Anesth. Analg117: 1093–100. Nov 2013. doi:10.1213/ANE.0b013e3182a705aePMID 24108261.
  6. Jump up^ “Two Scientific Remimazolam Presentations Are Accepted for ASA and ACG Meeting in October 2014”MarketWired. Oct 1, 2014. Retrieved 2014-10-24.
  7. Jump up^ “A Phase IIa, Randomized, Double-Blind Study of Remimazolam (CNS 7056) Versus Midazolam for Sedation in Upper Gastrointestinal Endoscopy”. Anesthesia120: 771–80. Dec 11, 2014. doi:10.1213/ANE.0000000000000548PMID 25502841

PATENTS

WO2016011943A1 *2014-07-232016-01-28李勤耕New benzodiazepine derivative and use thereof

WO2000069836A1 *1999-05-142000-11-23Glaxo Group LimitedShort-acting benzodiazepines
WO2008007081A1 *2006-07-102008-01-17Cenes LimitedShort-acting benzodiazepine salts and their polymorphic forms
CN101501019A *2006-07-102009-08-05Paion英国有限公司Short-acting benzodiazepine salts and their polymorphic forms
WO2012062439A1 *2010-11-082012-05-18Paion Uk Ltd.Dosing regimen for sedation with cns 7056 (remimazolam)
CN102753525A *2009-09-182012-10-24Paion英国有限公司Process for preparing 3-[(4s)-8-bromo-1-methyl-6-(2-pyridinyl)-4h-imidazol[1,2-a][1,4]benzodiazepine-4-yl]propionic acid methyl ester or the benzene sulfonate salt thereof, and compounds useful in that process
CN102964349A *2011-08-312013-03-13江苏恒瑞医药股份有限公司Tosilate of benzodiazepine derivative, its crystal forms, their preparation method and application
Patent ID Patent Title Submitted Date Granted Date
US9737547 DOSING REGIMEN FOR SEDATION WITH CNS 7056 (REMIMAZOLAM)
2017-01-06
US2015224114 DOSING REGIMEN OF SEDATIVE
2013-08-30
2015-08-13
US2015148338 COMPOSITIONS COMPRISING SHORT-ACTING BENZODIAZEPINES
2013-05-22
2015-05-28
US9777007 SHORT-ACTING BENZODIAZEPINE SALTS AND THEIR POLYMORPHIC FORMS
2015-11-23
2016-06-23
US8642588 SHORT-ACTING BENZODIAZEPINE SALTS AND THEIR POLYMORPHIC FORMS
2010-03-25
Patent ID Patent Title Submitted Date Granted Date
US2017217965 NEW BENZODIAZEPINE DERIVATIVE AND USE THEREOF
2015-07-22
US9656987 OXIDATION REACTION EXCELLENT IN CONVERSION RATE
2014-03-03
2016-01-14
US9156842 PROCESS FOR PREPARING 3-[(4S)-8-BROMO-1-METHYL-6-(2-PYRIDINYL)-4H-IMIDAZO[1, 2-A][1, 4]BENZODIAZEPINE-4-YL]PROPIONIC ACID METHYL ESTER OR THE BENZENE SULFONATE SALT THEREOF, AND COMPOUNDS USEFUL IN THAT PROCESS
2010-09-15
2012-12-27
US7435730 Short-acting benzodiazepines
2007-06-14
2008-10-14
US7528127 Short-acting benzodiazepines
2007-06-14
2009-05-05
Patent ID Patent Title Submitted Date Granted Date
US7485635 Short-acting benzodiazepines
2007-04-26
2009-02-03
US2015313913 POSITIVE ALLOSTERIC MODULATORS OF THE GABA-A RECEPTOR IN THE TREATMENT OF AUTISM
2014-02-04
2015-11-05
US9561236 DOSING REGIMEN FOR SEDATION WITH CNS 7056 (REMIMAZOLAM)
2011-11-07
2014-03-20
US2017044135 PROCESS FOR PREPARING 3-[(4S)-8-BROMO-1-METHYL-6-(2-PYRIDINYL)-4H-IMIDAZO[1, 2-A][1, 4]BENZODIAZEPINE-4-YL]PROPIONIC ACID METHYL ESTER OR THE BENZENE SULFONATE SALT THEREOF, AND COMPOUNDS USEFUL IN THAT PROCESS
2016-10-27
US9512078 PROCESS FOR PREPARING 3-[(4S)-8-BROMO-1-METHYL-6-(2-PYRIDINYL)-4H-IMIDAZO[1, 2-A][1, 4]BENZODIAZEPINE-4-YL]PROPIONIC ACID METHYL ESTER OR THE BENZENE SULFONATE SALT THEREOF, AND COMPOUNDS USEFUL IN THAT PROCESS
2015-09-01
2015-12-24
Patent ID Patent Title Submitted Date Granted Date
US2017217925 PROCESS FOR PREPARING 3-[(S)-7-BROMO-2-(2-OXOPROPYLAMINO)-5-PYRIDIN-2-YL-3H-1, 4-BENZODIAZEPIN-3-YL]PROPIONIC ACID METHYL ESTER
2017-04-14
US9193730 SHORT-ACTING BENZODIAZEPINE SALTS AND THEIR POLYMORPHIC FORMS
2010-04-01
US7473689 Short-acting benzodiazepines
2007-06-14
2009-01-06
US7160880 Short-acting benzodiazepines
2007-01-09
WO0069836 SHORT-ACTING BENZODIAZEPINES
2000-11-23
Remimazolam
Remimazolam.svg
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
Chemical and physical data
Formula C21H19BrN4O2
Molar mass 439.304 g/mol
3D model (JSmol)

//////////////CNS-7056 , CNS-7056X , ONO-2745  , CNS 7056 , CNS 7056X , ONO 2745, REMIMAZOLAM, PHASE 3, PHASE 3, PAION, Anesthesia, 308242-62-8

CC1=CN2C3=C(C=C(C=C3)Br)C(=NC(C2=N1)CCC(=O)OC)C4=CC=CC=N4


TIPIFARNIB, типифарниб , تيبيفارنيب , 替匹法尼 ,

$
0
0

Tipifarnib.svgDB04960.pngChemSpider 2D Image | tipifarnib | C27H22Cl2N4O

str1

TIPIFARNIB

R-115777, NSC-702818

Categories

UNIIMAT637500A

CAS number 192185-72-1 +form
192185-68-5 (racemate)
192185-69-6 (racemic; fumarate)
192185-70-9 (racemic; diHCl)

(+)-(R)-6-[1-Amino-1-(4-chlorophenyl)-1-(1-methylimidazol-5-yl)methyl]-4-(3-chlorophenyl)-1-methylquinolin-2(1H)-one

2(1H)-Quinolinone, 6-[(R)-amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-1-methyl-

Weight Average: 489.396
Chemical Formula C27H22Cl2N4O

типифарниб [Russian] [INN]
تيبيفارنيب [Arabic] [INN]
替匹法尼 [Chinese] [INN]
(R)-(+)-R115777
(R)-6-(Amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl)-4-(3-chlorophenyl)-1-methyl-2(1H)-quinolinone
(R)-6-(amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl)-4-(3-chlorophenyl)-1-methylquinolin-2(1H)-one
2 (1H))-Quinolinone,6-(amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl)-4-(3-chlorophenyl)-1-methyl-, 2(1H )-quinolinone
Title: Tipifarnib
CAS Registry Number: 192185-72-1; 192185-68-5 (unspecified stereo)
CAS Name: 6-[(R)-Amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-1-methyl-2(1H)-quinolinone
Manufacturers’ Codes: R-115777
Trademarks: Zarnestra (Janssen)
Molecular Formula: C27H22Cl2N4O
Molecular Weight: 489.40
Percent Composition: C 66.26%, H 4.53%, Cl 14.49%, N 11.45%, O 3.27%
Literature References: Farnesyl transferase inhibitor. Prepn: M. G. Venet et al., WO 9721701eidemUS 6037350 (1997, 2000 both to Janssen). Review of syntheses: P. R. Angibaud et al., Eur. J. Org. Chem. 2004, 479-486. Inhibition of farnesyl protein transferase and antitumor effects in vivo: D. W. End et al., Cancer Res. 61, 131 (2001). Clinical pharmacology and pharmacokinetics: J. Zujewski et al., J. Clin. Oncol. 18, 927 (2000). Accelerator mass spec determn in biological samples: R. C. Garner et al., Drug Metab. Dispos. 30, 823 (2002). Clinical evaluation in hematologic malignancies: J. Cortes et al., Blood 101, 1692 (2003). Review of clinical experience: P. Norman, Curr. Opin. Invest. Drugs 3, 313-319 (2002).
Properties: Crystals from 2-propanol, mp 234°. [a]D20 +22.86° (c = 0.98 in methanol).
Melting point: mp 234°
Optical Rotation: [a]D20 +22.86° (c = 0.98 in methanol)
Therap-Cat: Antineoplastic.
Keywords: Antineoplastic; Farnesyl Transferase Inhibitors.

PRODUCT PATENT

WO 9721701

Tipifarnib (R-115777) is a substance that is being studied in the treatment of acute myeloid leukemia (AML) and other types of cancer. It belongs to the family of drugs called farnesyltransferase inhibitors. It is also called Zarnestra. In June 2005, the FDA issued a Not Approvable Letter for Zarnestra.

Investigated for use/treatment in colorectal cancer, leukemia (myeloid), pancreatic cancer, and solid tumors.

Drug had been granted orphan drug designation by the FDA for the treatment of AML in 2004. In 2005, the Committee for Orphan Medicinal Products of the European Medicines Agency (EMEA) adopted a positive opinion on orphan medicinal product designation for the drug. In 2014, Eiger BioPharmaceuticals licensed the product for worldwide development for the treatment of viral diseases and Kura Oncology licensed development and commercialization rights for the treatment cancer indications.

Pharmacodynamics

R115777, a nonpeptidomimetic farnesyl transferase inhibitor, suppresses the growth of human pancreatic adenocarcinoma cell lines. This growth inhibition is associated with modulation in the phosphorylation levels of signal transducers and activators of transcription 3 (STAT3) and extracellular signal-regulated kinases (ERK)

Tipifarnib (INN,[1]:213 proposed trade name Zarnestra) is a farnesyltransferase inhibitor that is being investigated in patients 65 years of age and older with newly diagnosed acute myeloid leukemia (AML). It inhibits the Ras kinase in a post-translational modification step before the kinase pathway becomes hyperactive. It inhibits prenylation of the CaaX tail motif, which allows Ras to bind to the membrane where it is active. Without this step the protein cannot function.

It is also being tested in clinical trials in patients in certain stages of breast cancer.[2] It is also investigated as a treatment for multiple myeloma.[3]

For treatment of progressive plexiform neurofibromas associated with neurofibromatosis type I, it successfully passed phase I clinical trials but was suspended (NCT00029354) in phase II.[4][5] The compound was discovered by and is under investigation by Johnson & Johnson Pharmaceutical Research & Development, L.L.C, with registration number R115777.Approval process

Tipifarnib was submitted to the FDA by Johnson & Johnson for the treatment of AML in patients aged 65 and over with a new drug application (NDA) to the FDA on January 24, 2005.

In June 2005, the FDA issued a “not approvable” letter for tipifarnib.[6]Progeria

Confocal microscopy photographs of the descending aortas of two 15-month-old progeria mice, one untreated (left picture) and the other treated with the farnsyltransferase inhibitor drug tipifarnib (right picture). The microphotographs show prevention of the vascular smooth muscle cell loss that is otherwise rampant by this age. Staining was smooth muscle alpha-actin (green), lamins A/C (red) and DAPI (blue). (Original magnification, ×40)

It was shown on a mouse model of Hutchinson–Gilford progeria syndrome that dose-dependent administration of tipifarnib can significantly prevent both the onset of the cardiovascular phenotype as well as the late progression of existing cardiovascular disease.[7]

PATENT

TIPIFARNIB BY SOLIPHARMA

WO-2018103027

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018103027&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

Crystalline form (I, II, III and IV) of tipifarnib . Useful for the treatment and/or prevention of abnormal cell growth diseases such as lung cancer, pancreatic cancer, colon cancer, melanoma, neuroblastoma or glioma. first filing from Solipharma claiming tipifarnib which was developing by Kura Oncology , under license from Johnson & Johnson subsidiary J&JPRD (now Janssen Research & Development).

Tipifarnib is a farnesyltransferase inhibitor that acts on H-RAS or N-RAS mutant cells and has antiproliferative effects. It can block the farnesylation modification of RAS protein, thereby disturbing its localization on the inner surface of the plasma membrane and subsequent activation of downstream signaling pathways, and has an effective anti-tumor disease activity.
Tipifarny’s chemical name is (R)-(+)-6-[amino(4-chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl]-4-(3-chloro) Phenyl) 1-methyl-2(1H)-quinolinone, English name Tipifarnib; its chemical structure is shown below:
The patent document CN1101392C reports the preparation method of typrivadina, which is a racemate and does not disclose any characterization data; the patent document CN100567292C reports the preparation method of typ fenfanide, which is a mixture of certain enantiomeric excesses. Only the melting point of the mixture is mentioned; the patent document CN1246318C reports the preparation method of typifanidin and the method for the resolution and purification of tepifefene in its enantiomers. The present inventors have found that the form of typifene prepared according to the method provided by CN1246318C is in the crystalline state (herein referred to as “Form A”), but it has a defect of low crystallinity and poor stability of the crystal, and the patent The typifanibs reported in the documents CN1101392C and CN100567292C are both mixtures and lack the characteristic data accurately reflecting their physical form and cannot be fully disclosed.
PATENT

Cyclization of 3-(3-chlorophenyl)-N-phenyl-2-propenamide by means of polyphosphoric acid (PPA) at 100 °C gives 4-(3-chlorophenyl)-1,2,3,4-tetrahydroquinolin-2-one ,

Which is condensed with 4-chlorobenzoic acid by means of PPA at 140 °C to yield 6-(4-chlorobenzoyl)-4-(3-chlorophenyl)-1,2,3,4-tetrahydroquinolin-2-one

The dehydrogenation of compound  by means of Br2 in bromobenzene at 160 °C affords 6-(4-chlorobenzoyl)-4-(3-chlorophenyl)quinolin-2-one,

Which is N-alkyalted with iodomethane in the presence of BnNMe3Cl and NaOH in THF to provide 6-(4-chlorobenzoyl)-4-(3-chlorophenyl)-1-methylquinolin-2-one.

Condensation of compound  with 1-methylimidazole  by means of BuLi in THF gives the triaryl carbinol (N-1),

Which is finally treated with NH3 in THF to afford the target Tipifarnib, R-115777 .

Scheme SHOWING COMPLICATIONS

PATENT

WO 2005105782

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2005105782

Farnesyltransf erase inhibitors block the main post-translational modification of the Ras protein, thus interfering with its localization to the inner surface of the plasma
10 membrane and subsequent activation of the downstream effectors. Although initially developed as a strategy to target Ras in cancer, farnesyltransferase inhibitors have
subsequently been acknowledged as acting by additional and more complex
mechanisms that may extend beyond Ras involving GTP-binding proteins, kinases,
centromere-binding proteins and probably other f arnesylated proteins.
15
A particular farnesyltransferase inhibitor is described in WO 97/21701, namely (R)-(+)- 6-[amino(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l- methyl-2(liϊ)-quinolinone. The absolute stereochemical configuration of the compound was not determined in the experiments described in the above-mentioned patent
20 specification, but the compound was identified by the prefix “(B)” to indicate that it was the second compound isolated from column chromatography. The compound thus obtained has been found to have the (R)-(+)-configuration. This compound will be
referred to below by its published code number Rl 15777 and has the following formula

Rl 15777 (Tipifamib) is a potent, orally active inhibitor of f arnesylprotein transferase.
It is one of the most advanced of the farnesylprotein transferase inhibitors currently
reported to be in clinical development, being one of the agents that have progressed to phase III studies.
30 Rl 15777 has been found to have very potent activity against neoplaslic diseases.
Antineoplastic activity in solid tumors, such as breast cancer, as well as in haematological malignancies, such as leukemia, have been observed. Also combination studies have been carried out demonstrating that R 115777 can be safely combined with several highly active anticancer drugs.

In WO 01/53289, the racemates (±) (4-(3-chloro-phenyl)-6-[(6-chloro-pyridin-3-yl)-(4-methoxy-benzylamino)-(3-methyl-3-f: -imidazol-4-yl)-methyl]-l-cyclopropylmethyl-liϊ-quinolin-2-one (racemate 1) and (±) 4-(3-chloro-phenyl)-6-[(6-chloro-pyridin-3-yl)-[(4-methoxy-benzylidene)-amino]-(3-methyl-3jr7-imidazol-4-yl)-methyl]-l-cyclopropylmethyl-liϊ-quinolin-2-one (racemate 2) are prepared.

racemate 1 racemate 2

After chiral molecule separation using column chromatography, either the benzylamino or the benzilidine moiety of the resulting (+) and /or (-) enantiomers are converted to an amino group under acidic conditions.

The synthesis of Rl 15777 as originally described in WO 97/21701, is presented in scheme 1.

Herein, in step 1, the intermediate 1-methyl imidazole in tetrahydrofuran, is mixed with a solution of ra-butyllithium in a hexane solvent to which is added chlorotriethylsilane (triethylsilyl chloride), followed by a further addition of ra-butyllithium in hexane, the resulting mixture being cooled to -78°C before the addition of a solution of a compound of formula (I), i.e. 6-(4-chlorobenzoyl)-4-(3-chlorophenyl)-l-methyl-2(12ϊ)-quinolinone in tetrahydrofuran. The reaction mixture is subsequently brought to room temperature, and then hydrolysed, extracted with ethyl acetate and the organic layer worked up to obtain a compound of formula (II), i.e. (±)-6-[hydroxy(4-chlorophenyl) (l-methyl-liϊ-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lia- )-quinolinone.

In step 2, the hydroxy compound of formula (II) is chlorinated with thionylchloride to form a compound of formula (III), i.e. (±)-6-[chloro(4-chlorophenyl)(l -methyl- liJ-imidazol-5-yl)methyl]-4-(3-chloroρhenyl)-l-methyl-2(li3)-quinolinone.

In step 3, the chloro compound of formula (III) is treated, with NEaL OH in
tetrahydrofuran to form the amino compound of formula (IV), i.e. (±)-6-[amino(4-chlorophenyl)(l-methyl-l -imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l-methyl- 2(l/J)-quinolinone.

In step 4, the amino compound of formula (IV) is separated into its enantiomers by chiral column chromatography over Chiracel OD (25 cm; eluent: 100% ethanol; flow: 0.5 ml/rnin; wavelength: 220 nm). The pure (B)-fractions are collected and recrystallised from 2-propanol resulting in Rl 15777, the compound of formula (V).

Scheme 1

However, the procedure described in WO97/21701 has a number of disadvantages. For example, during the first step, the procedure results in the undesired formation of a corresponding compound of formula (XI), i.e. 6-[hydroxy(4-chlorophenyl) (1-methyl-lJrJ-imidazol-2-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(liϊ)-quinolinone)Jn which the imidazole ring is attached to the remainder of the molecule at the 2-position of the ring, instead of the desired 5-position. At the end of the procedure, this results in the formation of a compound of formula (XII), i.e.6-[amino(4-chlorophenyl)(l-methyl-lϊJ-imidazol-2-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lβ -quinolinone.

(XI) CXH)

The use of n-butyllithium during the conversion of a compound of formula (I) in a compound of formula (II) is also undesirable in a commercial process in view of its pyrophoric nature and the formation of butane, a flammable gas, as the by-product. Also the carrying out of this process step, at a temperature as low as -78°C, is inconvenient and costly on a commercial scale.
Finally, the purification of compound (V) using chiral chromatography is expensive and disadvantageous in view of the large amounts of solvent needed and the specialised equipment required to perform a large scale chiral chromatography.

Another process for the synthesis of Rl 15777 as described in WO 02/072574, is presented in scheme 2.

Herein, in step 1, 1-methyl imidazole in tetrahydrofuran is mixed with a solution of n-hexyllithium in a hexane solvent to which is added tri-iso-butylsilyl chloride, followed by a further addition of n-hexyllithium in hexane. The compound of formula (I) in tetrahydrofuran is then added to the reaction mixture, keeping the temperature between -5°C and 0°C. The resulting product of formula (II) is isolated by salt formation.

In step 2, the chlorination reaction is effected by treatment of the compound of formula (II) with thionyl chloride in 1 ,3-dimethyl-2-imidazolidinone.

In step 3, the chloro compound of formula (III) is treated with a solution of ammonia in methanol. After the addition of water, the compound of formula (IV), precipitates and can be isolated.

In step 4, the compound of formula (IV) can be reacted with L-(-)-dibenzoyl tartaric acid (DBTA) to form the diastereomeric tartrate salt with formula (VI) i.e. R-(-)-6-[amino(4-chlorophenyl)(l-methyl-ljt-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(l Z)-quinolinone [R-(R*,RH!)]-2,3-bis(benzoyloxy)butanedioate (2:3).

Finally, in step 5, the compound of formula (VI) is treated with aqueous ammonium hydroxide, to form the crude compound of formula (V) which is then purified by recrystallisation from ethanol to the pure compound (V).

(VI) (V)
Scheme 2

However, in view of the fact that water is present during the third and the fifth step of this procedure, there is significant formation of the hydroxy compound of formula (II).

This is important because the compounds of formula (II) and (V) are difficult to separate. In order to keep the quality of the final product (V) as high as possible, it is critical to limit the formation of compound (II).

The major drawback of the above described processes is the generation of large amounts of the other enantiomer that subsequently must be recycled.

Attempts were made to develop processes that solve this problem. One of the possibilities was to enter chirality in the first step of the procedure. A first study was carried out in order to determine if the conversion of an enantiomer of the hydroxy compound of formula (II) into a compound of formula (IV) could preserve chirality. Several experimental conditions have been tested starting with an enantiomer of a compound of formula (II), but racemisation always occurred.

Another possibility was to try entering chirality by adding N-methylimidazole under the reaction conditions described herein above under steps 1 of WO97/21701 and WO 02/072574, to an N-Ct-6alkyl-(S(R))-sulfinylketimine prepared from the compound of formula (I). It turned out that the resulting N-Cι-6alkyl-(S(R))-sulfinylamide of the compound of formula (I) was in the desired R-configuration and could be used for conversion into compound (V).
These results are completely unexpected, especially in view of Shaw et al.
(Tetrahedron Letters: 42, 7173-7176). Already in 2001, Shaw et al. disclosed an asymmetric synthesis process for the production of α-aryl-α-heteroaryl alkylamines using organometallic additions to N-tert-butanesulfinyl ketimines. However, the configuration and the yield of the final enantiomer formed with this process, was depending on the configuration of the N-tert-butanesulfinyl moiety of the ketimines, the composition of the aryl and/or the heteroaryl moieties of the ketimines, as well as on the organo- and the metallic moiety of the organometallic reagent. Furthermore, the use of heteroaryllithium reagents were described in this document, as being in particular disadvantageous, in view of their instability.

Thus the present invention solves the above described problems. It provides a new process for the preparation of the compound of formula (V) without the need to recycle one of the enantiomers while minimising the formation of undesired isomers and impurities and under conditions which offer economic advantages for operation on a commercial scale.

A. Preparation of intermediates

Example AJ
a) Preparation of /V-r(4-chlorophenyl)((,4- -chlorophenyl’)-l-methyl-l f-quinolin-2-one’)-6-yDmethylenel-2-methyl-2-propanesulfinamide TSfR-)! (com ound 15)


Ti(OEt) (0.0122 mol) was added to a mixture of compound (I) (0.0024 mol) and (R)-(+)-2-methyl-2-propane-sulfinamide (0.0024 mol) in DCM (15ml). The mixture was stirred and refluxed for 4 days, then cooled to room temperature. Ice water was added. The mixture was filtered over celite. Celite was washed with DCM. The organic layer was extracted with saturated sodium chloride. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. This fraction was purified by column chromatography over silica gel (40 μm) (eluent: DCM/MeOH 98/2). The pure fractions were collected and the solvent was evaporated, yielding 0.95g of compound 15 _ (76%), melting point: 115°C.

b) Preparation of (R)-N-r(4-chlorophenyl1((4-(3-chlorophenyl)-l-methyl-lic/-quinoline- 2-one -6-ylVl-methyl-l/j-imidazole-5-yl’)methyll-2-methyl-2-propanesulfinamide rS(R)l (compound 161

(compound 16)

n-Butyllithium (1.34ml, 0.002 mol) was added dropwise at -70°C to a mixture of 1-methylimidazole (0.0021 mol) in THF (4.5ml). The mixture was stirred at -70°C for 15 minutes. Triethylsilyl chloride (0.0021 mol) was added. The mixture was stirred at -70°C for 15 minutes. n-Butyllithium (1.34ml, 0.0021 mol) was added dropwise. The mixture was stirred at -70°C for 15 minutes. A solution of compound 15 (0.0019 mol) in THF (5.5ml) was added. The mixture was stirred at -70°C for 45 minutes, poured out into ice water and extracted with EtOAc. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-40 m)(eluent: DCM/MeOH/ΝEUOH 95/5/0.5), yielding 0.59g (52%) of compound 16, diastereomeric excess 24%.

c) Preparation of the (B)-diastereomer (compound 18) of compound 16

(compound 18)

Compound 16 was purified by column chromatography over silica gel (15-40μm) (eluent: DCM/MeOH/NHtOH 95/5/0.5). Two fractions were collected and the solvent was evaporated, yielding 0.304g diastereomer (B) (compound 18) (27%), melting point 174°C.

Example A.2
a) Preparation of jV-r(4-chlorophenyl¥(4-(3-chlorophenyl)-l-methyl-l JJ-quinolin-2-one)-6-yl)methylene1-4-methylphenylsulfιnamidesulfιnamide fS(S)l (compound 17)

(compound 17)

Ti(OEt)4 (0.0122 mol) was added to a mixture of compound (I) (0.0123 mol) and (S)-(+)-j5-toluenesulfinamide (0.0123 mol) in DCM (80ml). The mixture was stirred and refluxed for 4 days, then cooled to room temperature. Satured sodium chloride was added. The mixture was filtered over celite. Celite was washed with DCM. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. A fraction was purified by column chromatography over silica gel (40 μm) (eluent: DCM MeOH 98/2). The fractions were collected and the solvent was evaporated, yielding 0.65g of pure compound 17 .

The pure compound N-[(4-chlorophenyl)((4-(3-chlorophenyl)-l-methyl-l-tf-quinolin-2-one)-6-yl)methylene]-2-methyl-2-propanesulfinamide [S(R)] can be obtained in an analogues way.

B. Preparation of final compounds

Example BJ
a Preparation of compound (V)

Hydrochloric acid in isopropanol was added to a solution of compound 16 (0.00003 mol) in methanol (0J ml). The mixture was stirred at room temperature for 30 minutes. The mixture was added to potassium carbonate (10%) on ice. The organic layer was separated, washed with a solution of saturated sodium chloride, dried (MgS04), filtered, and evaporated giving 0,017 g (100%) of compound (V), enantiomeric excess 22%, content of compound (II) < 1%.

PATENT

WO 2005105783

https://encrypted.google.com/patents/WO2005105783A1?cl=en

A. Preparation of intermediates

Example A.1

a) Preparation of N-r(4-chlorophenyl’)(l-methyl-lH-imidazol-5-yl)methylene)l-2- methyl-2-propanesulfinamide KSfl l (compound 25)

Figure imgf000016_0001

(compound 25) Ti(OEt)4 (0.0162 mol) was added to a mixture of (4-chlorophenyiχi-methyl-lH- imidazol-5-yl)methanone (0.0032 mol) and (R)-(+)-2-methyl-2-propane-sulfinamide (0.0032 mol) in DCE (7ml). The mixture was stirred and refluxed for 6 days, then cooled to room temperature. Ice water was added. The mixture was filtered over celite. Celite was washed with DCM. The organic layer was extracted with saturated sodium chloride. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. This fraction was purified by column chromatography over silica gel (40 μm) (eluent: DCM/MeOH/NH OH 97/3/0.5), yielding 0.475g of compound 25 (46%).

The compound N-[(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methylene)]-2-methyl- 2-propanesulfinamide [(S(S)] can be obtained in an analogous way.

b) Preparation of N-r(4-chlorophenyl)((4-(3-chlorophenyl)-2-methoχy-quinoline-6- yl l-methyl-lH-imidazole-5-yl)methyn-2-methyl-2-propanesulfinamide TS(R)1 (compound 26)

Figure imgf000017_0001

(compound 26)

n-Butyllithium (0.00081 mol) in hexane, was added dropwise at -78°C to a mixture of 6-bromo-4-(3-chlorophenyl)-2-methoxy-quinoline (0.00081 mol) in THF (3 ml) under nitrogen flow. The mixture was stirred at -78°C for 30 minutes. A solution of compound 25 (0.00065 mol) in THF (0.6 ml) was added . The mixture was stirred at – 78°C for 1 hour and 30 minutes, poured out into ice water and extracted with EtOAc. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. This fraction was purified by column chromatography over silica gel (40μm)(eluent: DCM eOH/NB OH 97/3/0.1). The pure fractions were collected and the solvent was evaporated, yielding 0.138g (36 %) of compound 26, melting point 153°C.

The compound N-[(4-chlorophenyl)((4-(3-chlorophenyl)-2-methoxy-quinoline-6-yl)(l- methyl-lH-imidazole-5-yl)methyl]-2-methyl-2-propanesulfmamide [S(S)] can be obtained in an analogous way

c) Preparation of (S)-l-,4-chlorophenylV l-r4-(3-chlorophenylV2-methoxy-quinoline-6- yll-l-(l-methyl-l/J-imidazole-5-yl)-methylamine (compound 27)

Figure imgf000017_0002

(compound 27) Hydrochloric acid in isopropanol was added to a solution of compound 26 (0.000018 mol) in methanol (4.2 ml). The mixture was stirred at room temperature for 30 minutes. The mixture was added to potassium carbonate (10%) on ice and extracted with ethyl acetate. The organic layer was separated, washed with a solution of saturated sodium chloride, dried (MgS0 ), filtered, and evaporated giving 0,086 g (100%) of compound 27, melting point 96°C, enantiomeric excess 88%. d) Preparation of (SV6-ramino(4-chlorophenyl¥l-methyl-l #-imidazol-5-yDmethyH-4- (3-chlorophenyD-lH)-quinorin-2-one (compound 28)

Figure imgf000018_0001

(compound 28) Compound 27 (0.00038 mol) in hydrochloric acid 3N (9.25 ml) and THF (9.25 ml), was stirred at 60°C for 24 hours and evaporated, giving 0,18 g (100%) of compound 28, melting point 210°C.

Example A.2

a) Preparation of N-r(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl’)methylene)1-p-

Figure imgf000018_0002

(compound 29) Ti(OEt)4 (0.0419 mol) was added to a mixture of (4-chlorophenyl)(l-methyl-lH- imidazol-5-yl)methanone (0.0084 mol) and (S)-(+)-p-_toluenesulfinamide (0.0084 mol) in DCE (18ml). The mixture was stirred and refluxed for 7 days, then cooled to room temperature. Ice water was added. The mixture was filtered over celite. Celite was washed with DCM. The organic layer was extracted with saturated sodium chloride. The organic layer was separated, dried (MgS04), filtered, and the solvent was evaporated. This fraction was purified by column chromatography over silica gel (40 μm) (eluent: DCM/MeOH/ΝHiOH 97/3/0.5), yielding 1.15 g of compound 29 (38%).

The compound N-[(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methylene)]-p- toluenesulfinamide [(S(R)] can be obtained in an analogues way. B. Preparation of final compounds

Example B.l a) Preparation of (S)-6-ramino(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methyll-4-

Figure imgf000019_0001

Compound 28 (0.00038 mol) was added to a solution of THF (1.8 ml) and NaOH ION (1.8 ml). BTEAC (0.0019 mol) and methyliodide (0.00076 mol) were added and the mixture was stirred for 2 hours at room temperature. EtOAc was added. The organic layer was separated, dried (MgS04), filtered, and evaporated giving 0,149 g (83%) of compound 30, enantiomeric excess 86%.

PATENT

WO 02/072574

https://encrypted.google.com/patents/WO2002072574A1?cl=en

Preparation of compound (III):

110 ml of dry tetrahydrofuran was added to 7.6 ml of 1-methylimidazole (0.0946 mole) and the resulting solution cooled to -15°C.37.8 ml of n-hexyllithium 2.5 M in n-hexane (0.0946 mole) was added, while the temperature during addition was kept between – 5°C and 0°C. After addition, the reaction mixture was stirred for 15 minutes, while cooling to -12°C. 26.2 ml of tri-w o-butylsilyl chloride (0.0964 mole) was added, while the temperature during addition was kept between -5° and 0°C. After addition, the reaction mixture was stirred for 15 minutes, while cooling to -13°C. 37.2 ml of n- hexyllithium 2.5 M in n-hexane (0.0930 mole) was added, while the temperature during addition was kept between -5°C and 0°C (some precipitation occured). After addition, the reaction mixture was stirred for 15 minutes, while cooling to -14°C. 128 ml of dry tetrahydrofuran was added to 26.22 g of 6-(4-chlorobenzoyl)-4-(3-chlorophenyl)-l- methyl-2(lH)-quinolinone (compound (II)) (0.0642 mole) and stirred until dissolution. This solution was added to the reaction mixture, while the temperature during addition was kept between -5°C and 0°C. After addition, the reaction mixture was stirred for 15 minutes between -5°C and 0°C. 128 ml of water was added to the reaction mixture, followed by the addition of 10.6 ml of acetic acid. The mixture was then heated to 40°C and stirred for 2 hours. The layers were separated and the organic layer washed with 32 ml water. 64 ml water and 7.8 ml aqueous NaOΗ 50% were added to the organic layer which was stirred for 1 hour at ambient temperature. The layers were separated and the organic layer concentrated under reduced pressure, yielding 51.08 g of a brown oil (46.6 wt% 4-(3-chlorophenyl)-6-[(4-chlorophenyl)hydroxy(l-methyl-lH-imidazol-5- yl)methyl]-l-methyl-2(lH)-quinolinone (compound HI); 75.6 % yield).

The product can be isolated via the procedures mentioned above. The resulting product was analysed by hplc using the following conditions :-

Column: Ηypersil C18-BD 3μm, 100mm x 4 mm (i.d.)

Mobile phase:

Solvent A: 0.5% NΗLjOAc

Solvent B: CΗ3CN

Gradient: Time %A %B

0 100 0

15 0 100

18 0 100 19 100 0 23 100 0 Detector: UV 254nm Solvent: DMF The product was found to have a C5:C2 ratio of 99.8:0.2. In contrast using n-butyllithium in place of n-hexyllithium, triethylsilyl chloride in place of tri-i.ro- butylsilyl chloride and conducting the process at -70°C, i.e. generally in accordance with prior art procedures discussed above, the resulting product had a C5:C2 ratio of 95:5, a significant difference in commercial terms.

Preparation of compound (IV)

A 1 liter reaction vessel was charged with 105.4 g of 4-(3-chlorophenyl)-6-[(4- chlorophenyl)hydroxy ( 1 -methyl- 1 H-imidazol-5-yl)methyl] – 1 -methyl-2( 1 H)- quinolinone hydrochloric acid salt (compound (IΗ)and 400 ml of N,N- dimethylimidazolidinone added at 22°C. The mixture was stirred vigorously for 15 minutes at 22°C and became homogeneous. 32.1 ml of thionyl chloride was added over 10 minutes to the reaction mixture, the reaction temperature rising from 22°C to 40°C. After addition of the thionyl chloride, the reaction mixture was cooled from 40°C to 22°C and stirred for three hours at the latter temperature to provide a solution of 4-(3- chlorophenyl)-6-[chloro-(4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methyl]-l- methyl-2(lH)-quinolinone (compound (IN).

Preparation of unresolved compound (I)

429 ml of ammonia in methanol 7Ν was cooled to 5°C in a 3 liter reaction vessel and the solution of compound (IN), obtained in the previous stage, added, while stirring, over 10 minutes, with an exothermic reaction, the temperature rising from 5°C to 37°C. After the addition was complete, the reaction mixture was cooled to 22°C and stirred for 20 hours. 1000ml of water was then added over 20 minutes, the addition being slightly exothermic so the reaction mixture was cooled to keep the temperature below 30°C. The mixture was then stirred for 22 hours at 22°C, the resulting precipitate filtered off and the precipitate washed three times with 100ml of water to provide a yield of 70-75% of 6-[arnino(4-chlorophenyl)-l-methyl-lH-imidazol-5-ylmethyl]-4-(3- chlorophenyl)-l-methyl-2(lH)-quinolinone. Resolution of compound (I)

a) A 3 liter reaction vessel was charged with 146.8 g of 6-[amino(4-chlorophenyl)(l- methyl-lH-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lH)-quinolinone and 301.1 g of L-(-)-dibenzoyl-tartaric acid monohydrate, 1200ml of acetone was added and the reaction mixture stirred vigorously for 10 minutes at 22°C to form a solution which was seeded with lOOmg of the final tartrate salt product (obtained from previous screening experiments) and then stirred for 22 hours at 22°C. The resulting precipitate was filtered off and the precipitate was washed twice with 75 ml of acetone and the product dried at 50°C in vacuo to yield 114.7g of R-(-)-6-[amino(4-chlorophenyl)(l- methyl-lΗ-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lΗ)-quinolinone [R- (R*,R*)]-2,3-bis(benzoyloxy)butanedioate (2:3).

b) 41.08 g of the product of stage a) and 80 ml ethanol were stirred for 15 minutes at 22°C. 12.0 ml concentrated aqueous ammonium hydroxide was added over 2 minutes, and the reaction mixture stirred for 1 hour at 25°C. 160 ml water was added over 10 minutes at 25 °C and the mixture heated to reflux and stirred at reflux for 1 hour. The reaction mixture was then cooled to 20°C and stirred for 16 hours at 20°C. The product was filtered, washed twice with 8 ml water and dried at 50°C in vacuo to yield 16.87 g of (R)-(+)-6-[amino(4-chloro-phenyl)(l-methyl-lH-imidazol-5-yl)methyl]-4-(3- chlorophenyl)-l-methyl-2(lH)-quinolinone (compound (I)).

Purification of compound (I)

265 ml of ethanol was added to 19.9g of compound (I), obtained as described in the previous stage, and the mixture warmed while stirring to reflux temperature (78 °C) and then stirred at reflux temperature for 15 minutes before cooling the solution to 75 °C. 1.0 g of activated carbon (Norit A Supra) was then added to the mixture which was stirred at reflux temperature for 1 hour, filtered while warm and the filter then washed with 20 ml warm ethanol. The filtrate and wash solvent were combined (the product spontaneously crystallizes at 48°C), and the mixture warmed to reflux temperature and concentrated by removing 203 ml of ethanol. The resulting suspension was cooled to 22°C, stirred for 18 hours at 22°C, cooled to 2°C and stirred for 5 more hours at 2°C. The precipitate was filtered and washed with 4 ml ethanol and the product dried at 50°C in vacuo to yield 17.25 g of purified compound (I) which complies with the infrared spectrum of reference material.

PAPER

Practical route to 2-quinolinones via a pd-catalyzed c-h bond Activation/C-C bond Formation/Cyclization cascade reaction
Org Lett 2015, 17(2): 222

https://pubs.acs.org/doi/10.1021/ol503292p

Practical Route to 2-Quinolinones via a Pd-Catalyzed C–H Bond Activation/C–C Bond Formation/Cyclization Cascade Reaction

Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
Org. Lett.201517 (2), pp 222–225
DOI: 10.1021/ol503292p
Publication Date (Web): December 29, 2014
Copyright © 2014 American Chemical Society
Abstract Image

Quinolinone derivatives were constructed via a Pd-catalyzed C–H bond activation/C–C bond formation/cyclization cascade process with simple anilines as the substrates. This finding provides a practical procedure for the synthesis of quinolinone-containing alkaloids and drug molecules. The utility of this method was demonstrated by a formal synthesis of Tipifarnib.

SEE https://pubs.acs.org/doi/suppl/10.1021/ol503292p/suppl_file/ol503292p_si_001.pdf

4-(3-chlorophenyl)-6-(4-chlorobenzyl)-2-quinolinone 5:

str1

0.5 mmol 4-Amino-4′-chlorodiphenylmethane 4, 1mmol acetic anhydride and 2 mL toluene were added into the Schlenk tuble. The mixture was stirred at r.t. for 5 minutes, then 0.5 mmol TsOH•H2O, 2.5 mmol (2E)-3-(3-chlorophenyl) propenoate, 1.5 mmol Na2S2O8 and 5 mmol % Pd(OAc)2 were added into the reaction system in one time. The mixture was heated at 100 oC for 36 h and cooled down to room temperature, quenched with 50 mL saturated sodium bicarbonate solution and extracted thrice with ethyl acetate (30 mL) and the combined organic phase was dried over Na2SO4. After evaporation of the solvents the residue was purified by silica gel chromatography to afford 5 as pale yellow solid (elute: hexane-EtOAc) (180 mg, 95%).

1H NMR (400 MHz, d6-DMSO) ppm: 11.87 (s, 1H), 7.59-7.52 (m, 2H), 7.50-7.47 (m, 1H), 7.42-7.37 (m, 2H), 7.35-7.28 (m, 3H), 7.19-7.14 (m, 3H), 6.41 (s, 1H), 3.92 (s, 2H).

13C NMR (100 MHz, d6-DMSO): 161.50, 150.09, 140.52, 139.13, 138.25, 134.89, 133.85, 132.04, 131.16, 130.99, 130.95, 129.17, 128.88, 128.80, 127.94, 125.84, 122.30, 118.44, 116.55, 39.92.

HRMS (ESI) Calcd. for C22H15Cl2NO: [M + H]+ , 380.0609. Found: m/z 380.0613.

4-(3-chlorophenyl)-6-(4-chlorobenzoyl)-2-quinolinone 6:1

str2

4-(3-chlorophenyl)-6-(4-chlorobenzyl)-2-quinolinone 5 (0.2 mmol), iodine (0.002 mmol), pyridine (0.002 mmol) and aqueous tert-butylhydroperoxide (70%, 0.5 ml) were sealed in a 5 mL tube, then stirred at 80 oC overnight. After cooling to room temperature, the mixture was purified by a short silica gel chromatography column to afford 6 as pale yellow solid (elute: DCM/acetone = 2/1) (77 mg, 98%).

1H NMR (400 MHz, d6-DMSO) ppm: 12.31 (s, 1H), 8.00 (dd, J = 8.40 Hz, 1.60 Hz, 1H), 7.76 (d, J = 8.40 Hz, 2H), 7.74 (d, J = 1.60 Hz, 1H) 7.68 (s, 1H), 7.60 (d, J = 8.40 Hz, 2H), 7.55-7.50 (m, 4H), 6.57 (s, 1H).

13C NMR (100MHz, d6-DMSO): 193.48, 161.83, 150.38, 143.00, 138.46, 137.74, 136.36, 133.92, 132.04, 131.85, 131.16, 130.20, 129.93, 129.57, 129.08, 128.99, 128.11, 123.01, 117.81, 116.74. HRMS (ESI) Calcd. for C22H13Cl2NO2: [M + H]+ , 394.0402. Found: m/z 394.0405.

Reference: 1. Zhang, J.; Wang, Z.; Wang, Y.; Wan, C.; Zheng, X.; Wang, Z. Green Chem. 2009, 11, 1973. 2. (a) Angibaud, P.; Venet, M.; Filliers, W.; Broeckx, R.; Ligny, Y.; Muller, P.; Poncelet, V.; End, D. Eur. J. Org. Chem. 2004, 479. (b) Filliers, W.; Broeckx, R.; Angibaud, P. U.S. patent, US7572916, 2009.

NMR SIMULATION

PREDICTED VALUES

1H NMR: δ 3.42 (3H, s), 3.63 (3H, s), 6.57 (1H, s), 6.67 (1H, d, J = 1.7 Hz), 7.27 (1H, dd, J = 8.3, 1.5 Hz), 7.36-7.59 (8H, 7.46 (ddd, J = 8.3, 1.5, 0.5 Hz), 7.41 (ddd, J = 8.1, 8.1, 0.5 Hz), 7.39 (ddd, J = 8.1, 1.6, 1.5 Hz), 7.49 (ddd, J = 8.1, 1.7, 1.5 Hz), 7.55 (ddd, J = 8.3, 1.6, 0.5 Hz), 7.58 (d, J = 1.7 Hz)), 7.66 (1H, dd, J = 8.3, 0.5 Hz), 7.71 (1H, dd, J = 1.5, 0.5 Hz), 7.84 (1H, ddd, J = 1.7, 1.6, 0.5 Hz).

13C NMR PREDICT

str1

COSY PREDICT

HSQC PREDICT

References

  1. Jump up^ “International Nonproprietary Names for Pharmaceutical Substances (INN). Recommended International Nonproprietary Names (Rec. INN): List 46” (PDF). World Health Organization. Retrieved 16 November 2016.
  2. Jump up^ Sparano, JA; Moulder, S; Kazi, A; Coppola, D; Negassa, A; Vahdat, L; Li, T; Pellegrino, C; Fineberg, S; Munster, P; Malafa, M; Lee, D; Hoschander, S; Hopkins, U; Hershman, D; Wright, JJ; Kleer, C; Merajver, S; Sebti, SM (15 April 2009). “Phase II Trial of Tipifarnib plus Neoadjuvant Doxorubicin-Cyclophosphamide in Patients with Clinical Stage IIB-IIIC Breast Cancer” (PDF). Clinical Cancer Research15 (8): 2942–48. doi:10.1158/1078-0432.CCR-08-2658PMC 2785076Freely accessiblePMID 19351752. Retrieved 16 November 2016.
  3. Jump up^ Alsina, M; Fonseca, R; Wilson, EF; Belle, AN; Gerbino, E; Price-Troska, T; Overton, RM; Ahmann, G; Bruzek, LM; Adjei, AA; Kaufmann, SH; Wright, JJ; Sullivan, D; Djulbegovic, B; Cantor, AB; Greipp, RP; Dalton, WS; Sebti, SM (1 May 2004). “Farnesyltransferase Inhibitor Tipifarnib Is Well Tolerated, Induces Stabilization of Disease, and Inhibits Farnesylation and Oncogenic/Tumor Survival Pathways in Patients with Advanced Multiple Myeloma” (PDF). Blood103 (9): 3271–7. doi:10.1182/blood-2003-08-2764PMID 14726402. Retrieved 16 November 2016.
  4. Jump up^ “R115777 in Treating Patients With Advanced Solid Tumors”
  5. Jump up^ “R115777 to Treat Children With Neurofibromatosis Type 1 and Progressive Plexiform Neurofibromas”
  6. Jump up^ “Johnson & Johnson Pharmaceutical Research & Development, L.L.C. Receives Not Approvable Letter From FDA for Tipifarnib Based on Phase II Data”. PR Newswire. Jun 30, 2005. Retrieved 16 November 2016.
  7. Jump up^ Capell, BC; Olive, M; Erdos, MR; Cao, K; Faddah, DA; Tavarez, UL; Conneely, KN; Qu, X; San, H; Ganesh, SK; Chen, X; Avallone, H; Kolodgie, FD; Virmani, R; Nabel, EG; Collins, FS (6 October 2008). “A Farnesyltransferase Inhibitor Prevents Both the Onset and Late Progression of Cardiovascular Disease in a Progeria Mouse Model” (PDF). Proceedings of the National Academy of Sciences105 (41): 15902–7. doi:10.1073/pnas.0807840105PMC 2562418Freely accessiblePMID 18838683. Retrieved 16 November 2016.
Tipifarnib
Tipifarnib.svg
Clinical data
Synonyms R115777
ATC code
  • None
Legal status
Legal status
  • Investigational
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C27H22Cl2N4O
Molar mass 489.40 g·mol−1
3D model (JSmol)
PATENT 
Cited Patent Filing date Publication date Applicant Title
WO1997021701A1 * Oct 16, 1996 Jun 19, 1997 Janssen Pharmaceutica N.V. Farnesyl protein transferase inhibiting (imidazol-5-yl)methyl-2-quinolinone derivatives
WO2001051127A1 * Jan 9, 2001 Jul 19, 2001 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2001053289A1 * Nov 29, 2000 Jul 26, 2001 Pfizer Products Inc. Anticancer compound and enantiomer separation method useful for synthesizing said compound
WO2002020015A1 * Aug 30, 2001 Mar 14, 2002 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2002072574A1 * Mar 5, 2002 Sep 19, 2002 Janssen Pharmaceutica N.V. Process for the preparation of imidazole compounds
WO2002079147A2 * Mar 26, 2002 Oct 10, 2002 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
NON-PATENT CITATIONS
Reference
1 * SHAW A W ET AL: “Asymmetric synthesis of alpha,alpha-diaryl and alpha-aryl-alpha-heteroaryl alkylamines by organometallic additions to N-tert-butanesulfinyl ketimines” TETRAHEDRON LETTERS, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 42, no. 41, 8 October 2001 (2001-10-08), pages 7173-7176, XP004304959 ISSN: 0040-4039 cited in the application
Citing Patent Filing date Publication date Applicant Title
US9707221 Nov 8, 2016 Jul 18, 2017 Kura Oncology, Inc. Methods of treating cancer patients with farnesyltransferase inhibitors

//////////////////TIPIFARNIB , R-115777, типифарниб تيبيفارنيب 替匹法尼 , NSC-702818  , phase 3, orphan drug designation, NSC 702818, R 115777, Kura Oncology, Zarnestra, Janssen

CN1C=NC=C1[C@@](N)(C1=CC=C(Cl)C=C1)C1=CC2=C(C=C1)N(C)C(=O)C=C2C1=CC(Cl)=CC=C1

Glasdegib, PF-04449913

$
0
0

Glasdegib.svgChemSpider 2D Image | Glasdegib | C21H22N6OGlasdegib.png

str1

Glasdegib (PF-04449913)

1-[(2R,4R)-2-(1H-Benzimidazol-2-yl)-1-methyl-4-piperidinyl]-3-(4-cyanophenyl)urea [ACD/IUPAC Name]
1-[(2R,4R)-2-(1H-benzimidazol-2-yl)-1-methylpiperidin-4-yl]-3-(4-cyanophenyl)urea
CAS 1095173-27-5 [RN]Orphan Drug Status

Glasdegib

  • Molecular FormulaC21H22N6O
  • Average mass374.439 Da
  • Urea, N-[(2R,4R)-2-(1H-benzimidazol-2-yl)-1-methyl-4-piperidinyl]-N’-(4-cyanophenyl)- [ACD/Index Name]
    гласдегиб [Russian] [INN]
    غلاسديغيب [Arabic] [INN]
    格拉德吉 [Chinese] [INN]

FACT SHEET   https://www.pfizer.com/files/news/asco/Glasdegib-Fact-Sheet-6JUNE2018.pdf

Glasdegib (PF-04449913) is an experimental cancer drug developed by Pfizer. It is a small molecule inhibitor of the Sonic hedgehog pathway, which is overexpressed in many types of cancer. It inhibits smoothened receptor, as do most drug in its class.[1]

Four phase II clinical trials are in progress. One is evaluating the efficacy of glasdegib in treating myelofibrosis in patients who were unable to control the disease with ruxolitinib.[2] Another is a combination trial of glasdenib with ARA-Cdecitabinedaunorubicin, or cytarabine for the treatment of acute myeloid leukemia.[3] The third is for the treatment of myelodysplastic syndrome and chronic myelomonocytic leukemia.[4] The fourth administers glasdegib to patients at high risk for relapse after stem cell transplants in acute lymphoblastic or myelogenous leukemia.[5]

  • OriginatorPfizer
  • DeveloperGrupo Espanol de Trasplante Hematopoyetico y Terapia Celular; H. Lee Moffitt Cancer Center and Research Institute; Netherlands Cancer Institute; Pfizer
  • ClassAntineoplastics; Benzimidazoles; Phenylurea compounds; Piperidines; Small molecules
  • Mechanism of ActionHedgehog cell-signalling pathway inhibitors; SMO protein inhibitors
  • Orphan Drug StatusYes – Acute myeloid leukaemia; Myelodysplastic syndromes
  • New Molecular EntityYes

Highest Development Phases

  • Phase IIIAcute myeloid leukaemia
  • Phase IIChronic myeloid leukaemia; Colorectal cancer; Myelodysplastic syndromes; Myelofibrosis; Non-small cell lung cancer
  • Phase I/IIChronic myelomonocytic leukaemia; Glioblastoma; Graft-versus-host disease
  • Phase ICancer; Haematological malignancies
  • No development reportedSolid tumours

Most Recent Events

  • 20 Apr 2018Phase-III clinical trials in Acute myeloid leukaemia (Combination therapy, First-line therapy) in Japan (PO) (NCT03416179)
  • 02 Apr 2018Pfizer terminates a phase II trial in Myelofibrosis (Second-line therapy or greater) in USA, Japan, Austria, France, Spain and United Kingdom (PO) (NCT02226172) (EudraCT2014-001048-40)
  • 06 Feb 2018Phase-I/II clinical trials in Glioblastoma (Newly diagnosed) in Spain (PO) (EudraCT2017-002410-31)

Glasdegib is an orally bioavailable small-molecule inhibitor of the Hedgehog (Hh) signaling pathway with potential antineoplastic activity. Glasdegib appears to inhibit Hh pathway signaling. The Hh signaling pathway plays an important role in cellular growth, differentiation and repair. Constitutive activation of Hh pathway signaling has been observed in various types of malignancies.

Glasdegib is under investigation for the treatment of Acute Myeloid Leukemia.

SYNTHESIS

Discovery of PF-04449913, a Potent and Orally Bioavailable Inhibitor of Smoothened

https://pubs.acs.org/doi/abs/10.1021/ml2002423

 Michael J. Munchhof LLC, 266 West Road, Salem, Connecticut 06420, United States
 Pfizer Global Research and Development, Groton, Connecticut 06340, United States
§ 24 Queen Eleanor Drive, Gales Ferry, Connecticut 06335, United States
 INC Research, Old Lyme, Connecticut 06371, United States
 Reiter.MedChem, 32 West Mystic Avenue, Mystic, Connecticut 06355, United States
# Bristol-Meyers Squibb, Princeton, New Jersey 08540, United States
ACS Med. Chem. Lett.20123 (2), pp 106–111
DOI: 10.1021/ml2002423
Publication Date (Web): December 21, 2011
Copyright © 2011 American Chemical Society
*Tel: 860-287-5924. E-mail: mikemunchhof@yahoo.com.
Abstract Image

Inhibitors of the Hedgehog signaling pathway have generated a great deal of interest in the oncology area due to the mounting evidence of their potential to provide promising therapeutic options for patients. Herein, we describe the discovery strategy to overcome the issues inherent in lead structure 1 that resulted in the identification of Smoothened inhibitor 1-((2R,4R)-2-(1H-benzo[d]imidazol-2-yl)-1-methylpiperidin-4-yl)-3-(4-cyanophenyl)urea (PF-04449913, 26), which has been advanced to human clinical studies

1-((2R,4R)-2-(1H-benzo[d]imidazol-2-yl)-1-methylpiperidin-4-yl)-3-(4-cyanophenyl)urea (26)

https://pubs.acs.org/doi/suppl/10.1021/ml2002423/suppl_file/ml2002423_si_001.pdf

str1

Product was purified by Companion (ReadySep 40g, silica gel packed) with CH3OH/CH2Cl2 from 1-5% to give the title compound as an off-white solid 915mg (73%). LC-MS 375.3.

1H NMR(acetone-D6): δ 1.81 (m, 2H), 1.9- 2.05 (m, 2H), 2.10 (m, 1H), 2.17 (s, 3H), 2.52 (m, 1H), 2.94 (m, 1H), 3.86 (m, 1H), 4.2 (m, 1H), 6.4 (d, 1H), 7.16 (m, 2H), 7.52 (m, 2H), 7.60 (m, 2H), 7.62 (m, 2H), 8.46 (s, 1H).

The dihydrochloride salt was prepared by adding 4M HCl in dioxane (1.22mL, 4.86 mmol) to a solution of 1-((2R,4R)-2-(1H-benzo[d]imidazol-2-yl)-1-methylpiperidin-4-yl)-3-(4- cyanophenyl)urea (910 mg’s, 2.43mmol) in methanol (10mL). The mixture was stirred at at 230C for 10 minutes. The solution was concentrated to give a white solid, 1082 mg’s as the 2 .HCl monohydrate salt. M.P. > 125 0C with dehydration above 130 0C. Analytical calculated for free base C21H22N6O: C 67.38%, H 5.88%, N 22.46%; Found: C 67.16%, H 5.54%, N 22.18%. Purity of the dihydrochloride monohydrate salt was determined to be > 99.9% by analytical HPLC using a Xbridge C18; 3.5µm column and eluting with 95:5 0.1% Perchloric Acid (HClO4) solution in water and acetonitrile, over a gradient of 25 minutes, with and ending solvent ratio of 5:95. Enantiomeric purity of the dihydrochloride monohydrate salt was > 99.9% by chiral HPLC using a Chiralcel OJ column and eluting with 96:4 Heptane:Ethanol(with 0.1% diethylamine).

Syn 2

Development of a Concise, Asymmetric Synthesis of a Smoothened Receptor (SMO) Inhibitor: Enzymatic Transamination of a 4-Piperidinone with Dynamic Kinetic Resolution

https://pubs.acs.org/doi/10.1021/ol403630g

Chemical Research & Development, Analytical Research & Development, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
Org. Lett.201416 (3), pp 860–863
DOI: 10.1021/ol403630g
Publication Date (Web): January 22, 2014
Copyright © 2014 American Chemical Society
Abstract Image

A concise, asymmetric synthesis of a smoothened receptor inhibitor (1) is described. The synthesis features an enzymatic transamination with concurrent dynamic kinetic resolution (DKR) of a 4-piperidone (4) to establish the two stereogenic centers required in a single step. This efficient reaction affords the desired anti amine (3) in >10:1 dr and >99% ee. The title compound is prepared in only five steps with 40% overall yield.

https://pubs.acs.org/doi/suppl/10.1021/ol403630g/suppl_file/ol403630g_si_001.pdf

1-((2R,4R)-2-(1H-Benzo[d]imidazol-2-yl)-1-methylpiperidin-4-yl)-3-(4-cyanophenyl)urea (1)

1 as white solids3 (27.1 g, 99.5 wt%, 90.0% corrected yield, > 99.0 UPLC area% purity): m.p. 223–224 °C; UPLC tR 2.11 min; 1 H NMR (DMSO-d6) δ 12.39 (s, 1H), 8.94 (s, 1H), 7.69 (m, 2 H), 7.57 (m, 3 H), 7.43 (m, 1 H), 7.13 (m, 2H), 6.75 (d, J = 7.2 Hz, 1H), 4.08 (m, 1H), 3.63 (dd, J = 10.3, 3.5 Hz, 1H), 2.89 (dt, J = 12.0, 4.0 Hz, 1H), 2.40 (td, J = 11.9, 3.1 Hz, 1H), 2.06 (s, 3H), 1.98–2.10 (m, 1H), 1.83–1.95 (m, 2H), 1.72 (m, 1H); 13C NMR (DMSO-d6) δ 155.7, 153.9, 144.8, 142.7, 134.3, 133.2, 121.8, 120.9, 119.4, 118.5, 117.3, 111.2, 102.4, 58.6, 49.9, 43.7, 42.4, 36.0, 29.8. HRMS (EI) calcd. for C21H23N6O [M+H]+ : 375.1928; Found 375.1932.

To the crude solution of 3 in DMSO-H2O (UPLC assay ~55.0 mg/mL, 104 mL, ~5.74 g of 3, 24.9 mmol) from the enzymatic transamination reaction (vide supra) was added THF (57.0 mL) followed by 17 (mixture with imidazole, 9.31 gm, 74.0 wt%, 31.2 mmol). The mixture was then stirred at rt for three hours. Once the reaction was complete (<1 % of 3 remaining by UPLC), methanol (10.1 mL, 249 mmol) was added followed by 2-MeTHF (57.0 mL). The layers were separated and the aqueous was extracted with 2-MeTHF (57.0 mL). The combined organic layers were then washed with 2 × 50.0 mL water and 2 × 50.0 mL of 10% aqueous NaCl solution. The organic solution was then concentrated under vacuum and the solvent was switched to acetonitrile to give a slurry with a final volume of ~90.0 mL. The slurry was stirred at rt for three hours and filtered, and the solids were washed with 2 × 10.0 mL of acetonitrile and dried in oven at 60 °C for two hours. The solids (~7.90 gm) were then slurried in 70.0 mL of acetonitrile. The slurry was heated to 60 °C for two hours, cooled to rt, filtered, and the solids were dried in oven under vacuum at 60 °C for 12 hours to give 1 as white solids (7.64 g, 98.0 wt%, 80.0% corrected yield, > 98 UPLC area% purity). The analytical data were identical to that obtained with method A.

References

1. Lin TL, Matsui W. Hedgehog pathway as a drug target: smoothened inhibitors in development. Onco Targets Ther. 2012;5:47-58.

2. Munchhof MJ, Li Q, Shavnya A, et al. Discovery of PF-04449913, a potent and orally bioavailable inhibitor of smoothened. ACS Med Chem Lett. 2012;3(2):106-111.

3. Clement V, Sanchez P, de Tribolet N, et al. Hedgehog-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr Biol. 2007;17(2):165-172.

4. Deschler, B. and Lübbert, M. (2006), Acute myeloid leukemia: Epidemiology and etiology. Cancer, 107: 2099–2107. doi: 10.1002/cncr.22233.

5. American Cancer Society. Key statistics for acute myeloid leukemia. Available at https://www.cancer.org/cancer/acute-myeloid-leukemia/about/key-statistics.html. Accessed January 25, 2018.

6. SEER Cancer Stat Facts: Acute Myeloid Leukemia. National Cancer Institute. Bethesda, MD, April 2017. Available at: http://seer.cancer.gov/statfacts/html/amyl.html. Accessed January 25, 2018.

7. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid leukemia. Blood 2006; 107(9): 3481-5.

8. Estey E. Acute myeloid leukemia and myelodysplastic syndromes in older patients. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 2007; 25(14): 1908-15.

9. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 2012; 30(21): 2670-7.

10. Ornstein MC, Mukherjee S, Sekeres MA. More is better: combination therapies for myelodysplastic syndromes. Best Pract Res Clin Haematol. 2015;28(1):22-31.

11. American Cancer Society. What are the key statistics about myelodysplastic syndromes? Available at: http://www.cancer.org/cancer/myelodysplasticsyndrome/detailedguide/myelo-dysplastic-syndromes-key-statistics. Accessed January 25, 2018. 12. Ma X, Does M, Raza A, et al. Myelodysplastic syndromes: incidence and survival in the United States. Cancer. 2007;109(8):1536-1542

Glasdegib
Glasdegib.svg
Clinical data
Synonyms PF-04449913
Identifiers
CAS Number
ChemSpider
KEGG
Chemical and physical data
Formula C21H22N6O
Molar mass 374.45 g·mol−1
3D model (JSmol)
 to 3 of 3
Patent ID Patent Title Submitted Date Granted Date
US8431597 Benzimidazole derivatives
2012-02-24
2013-04-30
US8148401 BENZIMIDAZOLE DERIVATIVES
2009-01-01
2012-04-03
US9611330 COMPOSITIONS AND METHODS FOR CANCER AND CANCER STEM CELL DETECTION AND ELIMINATION
2012-09-07
2014-10-09

////////////Glasdegib, PF-04449913, гласдегиб غلاسديغيب 格拉德吉 , PF04449913, PF 04449913, phase 3, aml, Orphan Drug Status

CN1CCC(CC1C2=NC3=CC=CC=C3N2)NC(=O)NC4=CC=C(C=C4)C#N

Capmatinib, капматиниб , كابماتينيب , 卡马替尼 ,

$
0
0
  • ThumbChemSpider 2D Image | Capmatinib | C23H17FN6OChemSpider 2D Image | Capmatinib | C23H17FN6OCapmatinib.png

Capmatinib / INC280/ INCB 28060

INC280 / INCB-28060 FREE BASE

UNIITY34L4F9OZ

CAS number 1029712-80-8

WeightAverage: 412.428
Chemical FormulaC23H17FN6O

2-fluoro-N-methyl-4-{7-[(quinolin-6-yl)methyl]imidazo[1,2-b][1,2,4]triazin-2-yl}benzamide

Capmatinib dihydrochloride; CAS 1197376-85-4

1029712-80-8 [RN]
2-Fluoro-N-méthyl-4-[7-(6-quinoléinylméthyl)imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide [French] [ACD/IUPAC Name]
капматиниб [Russian] [INN]
كابماتينيب [Arabic] [INN]
卡马替尼 [Chinese] [INN]

Scheme 1

Scheme 2

Method C


Capmatinib has been used in trials studying the treatment of Melanoma, Gliosarcoma, Solid Tumors, Colorectal Cancer, and Hepatic Impairment, among others.

Capmatinib is an orally bioavailable inhibitor of the proto-oncogene c-Met (also known as hepatocyte growth factor receptor (HGFR)) with potential antineoplastic activity. Capmatinib selectively binds to c-Met, thereby inhibiting c-Met phosphorylation and disrupting c-Met signal transduction pathways. This may induce cell death in tumor cells overexpressing c-Met protein or expressing constitutively activated c-Met protein. c-Met, a receptor tyrosine kinase overexpressed or mutated in many tumor cell types, plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis.

Capmatinib has been used in trials studying the treatment of Melanoma, Gliosarcoma, Solid Tumors, Colorectal Cancer, and Hepatic Impairment, among others

Protein kinases (PKs) are a group of enzymes that regulate diverse, important biological processes including cell growth, survival and differentiation, organ formation and morphogenesis, neovascularization, tissue repair and regeneration, among others. Protein kinases exert their physiological functions through catalyzing the phosphorylation of proteins (or substrates) and thereby modulating the cellular activities of the substrates in various biological contexts, hi addition to the functions in normal tissues/organs, many protein kinases also play more specialized roles in a host of human diseases including cancer. A subset of protein kinases (also referred to as oncogenic protein kinases), when dysregulated, can cause tumor formation and growth, and further contribute to tumor maintenance and progression (Blume- Jensen P et al, Nature 2001, 411(6835):355-365). Thus far, oncogenic protein kinases represent one of the largest and most attractive groups of protein targets for cancer intervention and drug development. c-Met, a proto-oncogene, is a member of a distinct subfamily of heterodimeric receptor tyrosine kinases which include Met, Ron, and Sea (Birchmeier, C. et al., Nat. Rev. MoI. Cell Biol. 2003, 4(12):915-925; Christensen, J. G. et al., Cancer Lett. 2005, 225(1): 1-26). The only high affinity ligand for c-Met is the hepatocyte growth factor (HGF), also known as scatter factor (SF). Binding of HGF to c-Met induces activation of the receptor via autophosphorylation resulting in an increase of receptor dependent signaling. Both c-Met and HGF are widely expressed in a variety of organs, but their expression is normally confined to the cells of epithelial and mesenchymal origin, respectively. The biological functions of c-Met (or c-Met signaling pathway) in normal tissues and human malignancies such as cancer have been well documented (Christensen, J.G. et al., Cancer Lett. 2005, 225(l):l-26; Corso, S. et al., Trends in MoI. Med. 2005, l l(6):284-292).

HGF and c-Met are each required for normal mammalian development, and abnormalities reported in both HGF- and c-Met-null mice are consistent with proximity of embryonic expression and epithelial-mesenchymal transition defects during organ morphogenesis (Christensen, J.G. et al., Cancer Lett. 2005, 225(1): 1-26). Consistent with these findings, the transduction of signaling and subsequent biological effects of HGF/c-Met pathway have been shown to be important for epithelial-mesenchymal interaction and regulation of cell migration, invasion, cell proliferation and survival, angiogenesis, morphogenesis and organization of three-dimensional tubular structures (e.g. renal tubular cells, gland formation) during development. The specific consequences of c-Met pathway activation in a given cell/tissue are highly context-dependent.

Dysregulated c-Met pathway plays important and sometimes causative (in the case of genetic alterations) roles in tumor formation, growth, maintenance and progression (Birchmeier, C. et al., Nat. Rev. MoI. Cell. Biol. 2003, 4(12):915-925; Boccaccio, C. et al., Nat. Rev. Cancer 2006, 6(8):637-645; Christensen, J.G. et al., Cancer Lett. 2005, 225(1): 1-26). HGF and/or c-Met are overexpressed in significant portions of most human cancers, and are often associated with poor clinical outcomes such as more aggressive disease, disease progression, tumor metastasis and shortened patient survival. Further, patients with high levels of HGF/c-Met proteins are more resistance to chemotherapy and radiotherapy, hi addition to the abnormal HGF/c-Met expression, c-Met receptor can also be activated in cancer patients through genetic mutations (both germline and somatic) and gene amplification. Although gene amplification and mutations are the most common genetic alterations that have been reported in patients, the receptor can also be activated by deletions, truncations, gene rearrangement, as well as abnormal receptor processing and defective negative regulatory mechanisms.

The various cancers in which c-Met is implicated include, but are not limited to: carcinomas (e.g., bladder, breast, cervical, cholangiocarcinoma, colorectal, esophageal, gastric, head and neck, kidney, liver, lung, nasopharygeal, ovarian, pancreas, prostate, thyroid); musculoskeletal sarcomas (e.g., osteosarcaoma, synovial sarcoma, rhabdomyosarcoma); soft tissue sarcomas (e.g., MFH/fibrosarcoma, leiomyosarcoma, Kaposi’s sarcoma); hematopoietic malignancies (e.g., multiple myeloma, lymphomas, adult T cell leukemia, acute myelogenous leukemia, chronic myeloid leukemia); and other neoplasms (e.g., glioblastomas, astrocytomas, melanoma, mesothelioma and Wilm’s tumor (www.vai.org/met/; Christensen, J. G. et al., Cancer Lett. 2005, 225(1): 1-26).

The notion that the activated c-Met pathway contributes to tumor formation and progression and could be a good target for effective cancer intervention has been further solidified by numerous preclinical studies (Birchmeier, C. et al., Nat. Rev. MoI. Cell Biol. 2003, 4(12):915-925; Christensen, J.G. et al., Cancer Lett. 2005, 225(l):l-26; Corso, S. et al., Trends in MoI. Med. 2005, 11(6):284-292). For example, studies showed that the tpr-met fusion gene, overexpression oϊc-met and activated c-met mutations all caused oncogenic transformation of various model cell lines and resulted in tumor formation and metastasis in mice. More importantly, significant anti-tumor (sometimes tumor regression) and anti-metastasis activities have been demonstrated in vitro and in vivo with agents that specifically impair and/or block HGF/c-Met signaling. Those agents include anti-HGF and anti-c-Met antibodies, HGF peptide antagonists, decoy c-Met receptor, c-Met peptide antagonists, dominant negative c-Met mutations, c-Met specific antisense oligonucleotides and ribozymes, and selective small molecule c-Met kinase inhibitors (Christensen, J.G. et al., Cancer Lett. 2005, 225(1): 1-26).

In addition to the established role in cancer, abnormal HGF/c-Met signaling is also implicated in atherosclerosis, lung fibrosis, renal fibrosis and regeneration, liver diseases, allergic disorders, inflammatory and autoimmune disorders, cerebrovascular diseases, cardiovascular diseases, conditions associated with organ transplantation (Ma, H. et al., Atherosclerosis. 2002, 164(l):79-87; Crestani, B. et al., Lab. Invest. 2002, 82(8):1015-1022; Sequra-Flores, A. A. et al., Rev. Gastroenterol. Mex. 2004, 69(4)243-250; Morishita, R. et al., Curr. Gene Ther. 2004, 4(2)199-206; Morishita, R. et al., Endocr. J. 2002, 49(3)273-284; Liu, Y., Curr. Opin. Nephrol. Hypertens. 2002, l l(l):23-30; Matsumoto, K. et al., Kidney Int. 2001, 59(6):2023-2038; Balkovetz, D.F. et al., Int. Rev. Cytol. 1999, 186:225-250; Miyazawa, T. et al., J. Cereb. Blood Flow Metab. 1998, 18(4)345-348; Koch, A.E. et al., Arthritis Rheum. 1996, 39(9):1566-1575; Futamatsu, H. et al., Circ. Res. 2005, 96(8)823-830; Eguchi, S. et al., Clin. Transplant. 1999, 13(6)536-544).

Inhibitors of c-Met and other kinases are reported in U.S. Ser. No. 11/942,130, including the compound 2-fluoro-N-methyl-4-[7-(quinolin-6-ylmethyl)imidazo[l ,2-b][l ,2,4]triazin-2-yl]benzamide (I) having the structure indicated below.

(I)

New or improved forms of existing agents which inhibit kinases such as c-Met are continually needed for developing more effective pharmaceuticals to treat cancer and other diseases. The salts, compositions, and methods described herein are directed toward these needs and other ends.

PATENT

Example 8 4-Bromo-3-fluoro-N-methoxy-iV-methylbenzamide (3)

To a suspension of 4-bromo-3-fluorobenzoic acid (1, 967.9 g, 4.4 mol) in dichloromethane (5.9 L) and DMF (21 mL) was slowly added a solution of oxalyl chloride ((COCl)2, 560 mL, 6.4 mol, 1.45 equiv) in dichloromethane (520 mL) at room temperature. The resulting reaction mixture was stirred at room temperature for 20 h and then cooled to 0 0C by ice-water bath. iV,0-dimethyl hydroxylamine hydrochloride (826 g, 8.4 mol, 1.9 equiv) was added followed by slow addition of triethylamine (TEA, 2.5 L, 17.7 mol, 4.0 equiv) at 0 0C. The reaction mixture was then gradually warmed to room temperature and stirred at room temperature overnight. Once the coupling reaction was complete, the reaction mixture was washed with saturated aqueous sodium bicarbonate solution (NaHCO3, 2 L). The aqueous phase was back extracted with dichloromethane (1 L). The combined organic phases were washed with water (1 L), brine (1 L), and concentrated under reduced pressure. The resulting solid residue was dissolved into methyl tert-butyl ether (MTBE, 5 L), washed sequentially with water (5 x 1 L), brine (1 L), and dried over anhydrous sodium sulfate (Na2SO4). The filtrated solution was concentrated under reduced pressure and the resulting solid was dried in a vacuum oven at 45 0C to afford 4-bromo-3-fluoro-Λr-methoxy-Λr-methylbenzamide (3, 1106 g, 1153 g theoretical, 95.9% yield) which was used for the subsequent reaction without further purification. For 3: 1H NMR (400 MHz, DMSO-J6) δ ppm 7.78 (t, IH, J= 7.47 Hz), 7.56 (dd, IH, J= 9.3, 1.6 Hz), 7.18 (d, IH, J= 8.1 Hz), 3.53 (s, 3H), 3.25(s, 3H); C9H9BrFNO2 (MW 262.08), LCMS (EI) mle 262.0/ 264.0 (M+ + H).

Scheme 1 (Examples 8-14)

C7H4BrFO2 C7H3BrCIFO C9H9BrFNO2 MoI. Wt: 219.01 MoI. Wt: 237.45 MoI. Wt: 26208

C8H6BrFO C8H4BrFO2 Ci2Hi4BrFO3 MoI. Wt: 217.04 MoI. Wt: 231.02 MoI. Wt: 305.14

13

C22H13FN3 MoI. Wt: 380.38

Example 9 l-(4-Bromo-3-fluorophenyl)ethanone (4)

To a solution of crude 4-bromo-3-fluoro-N-methoxy-iV-methylbenzamide (3, 1106 g, 4.2 mol) in anhydrous tetrahydrofuran (THF, 11 L) was slowly added a 3.0 M solution of methylmagnesium chloride (MeMgCl, 2.5 L, 7.5 mol, 1.7 equiv) in THF at 0 0C. The resulting reaction mixture was stirred at 0 0C for 2 h and then quenched very carefully with saturated aqueous ammonium chloride (NH4Cl, 1.5 L). The resulting solution was concentrated under reduced pressure to remove most of THF. The residue was then diluted with ethyl acetate (EtOAc, 5 L) and the resulting solution was washed with water (2 L). The aqueous phase was extracted with ethyl acetate (EtOAc, 2 x 2 L). The combined organic phases were washed with water (2 L), brine (2 L) and dried over anhydrous sodium sulfate (Na2SO4). The filtered solution was concentrated under reduced pressure and the resulting solid was dried in a vacuum oven at 45 0C to afford l-(4-bromo-3-fluorophenyl)ethanone (4, 890.8 g, 911.6 g theoretical, 97.7% yield) as a solid which was used in the subsequent reaction without further purification. For 4: 1H NMR (400 MHz, OMSO-d6) δ ppm 7.89-7.84 (m, 2H), 7.71 (dd, IH, J= 8.30, 1.87 Hz), 2.57 (s, 3H).

Example 10 2-(4-Bromo-3-fluorophenyl)-2-oxoacetaldehyde (5)

To a solution of l-(4-bromo-3-fluorophenyl)ethanone (4, 890.8 g, 4.1 mol) in DMSO (4 L) was slowly added a solution of 48% aqueous hydrogen bromide (HBr, 1420 mL, 12.5 mol, 3.0 equiv). The reaction temperature was gradually increased from 2O0C to 50 0C during the course of the addition. The reaction mixture was subsequently heated to 60 0C and stirred at 60 0C overnight. The resulting dimethyl sulfide was removed by distillation and the residue was poured into ice water (28 L). The resulting yellow precipitate was collected by filtration (save the filtrate) and washed with water (5 L). The yellow solid was dissolved in ethyl acetate (EtOAc, 5 L), washed with brine (1 L) and dried over anhydrous sodium sulfate (Na2SO4). The solution was then concentrated under the reduced pressure and the resulting solid was dried in a vacuum oven at 45 0C to give the desired product, 2-(4-bromo-3-fluorophenyl)-2-oxoacetaldehyde, as its hydrate (hydrate of 5, 730.6 g, 1020.9 g theoretical, 71.6% yield). The aqueous phase (filtrate) was extracted with ethyl acetate (3 x 5 L) and the combined organic phase was washed with water (2 x 2 L), brine (2 L) and dried over anhydrous sodium sulfate (Na2SO4). The solution was concentrated under reduced pressure and the resulting solid was dried in a vacuum oven at 45 0C to give the second crop of 2-(4-bromo-3-fluorophenyl)-2-oxoacetaldehyde hydrate (hydrate of 5, 289.4 g, 1020.9 g theoretical, 28.3% yield; total 1020 g, 1020.9 g theoretical, 99.9% yield) which was used in the subsequent reaction without further purification. For hydrate of 5: H NMR (400 MHz, DMSO-</6) δ ppm 8.00-7.70 (m, 3H), 6.69 (br s, 2H), 5.59 (s, IH).

Example 11 l-(4-Bromo-3-fluorophenyl)-2,2-diethoxyethanone (6)

A 22 L flask was charged with the hydrate of (4-bromo-3-fluorophenyl)-2-oxoacetaldehyde (5, 1020 g, 4.41 mol), toluene (7.5 L), triethyl orthoformate (1633 g, 1.8 L, 11.04 mol, 2.5 equiv), para-toluene sulfonic acid (33.5 g, 0.176 mol, 0.4 equiv) at room temperature, and the resulting reaction mixture was heated to 110 0C and stirred at 1 10 0C for 6 h. When HPLC showed that the reaction was complete, the reaction mixture was cooled down to room temperature before being poured into a 50 L separation funnel along with ethyl acetate (7.5 L) and the saturated aqueous sodium bicarbonate solution (NaHCO3, 3 L). The mixture was stirred and the layers were separated. The aqueous layer was extracted with ethyl acetate (2 L). The combined organic layers were washed with brine (4 L), dried with sodium sulfate (Na2SO4), and concentrated under the reduced pressure to afford crude l-(4-bromo-3-fluorophenyl)-2,2-diethoxyethanone (6, 1240 g, 1345.7 g theoretical, 92.1% yield) which was used in the subsequent reaction without further purification. For 6: 1H NMR (400 MHz, DMSO-J6) δ ppm 7.94-7.94 (m, 2H), 7.78 (dd, IH, J= 8.51, 2.08 Hz), 5.40 (s, IH), 3.77-3.60 (m, 4H), 1.16-1.14 (m, 6H).

Example 12 6-(4-Bromo-3-fluorophenyl)-l,2,4-triazin-3-amine (7)

A 22 L flask was charged with l-(4-bromo-3-fluorophenyl)-2,2-diethoxyethanone (6, 1240 g, 4.07 mol), ethanol (11 L), water (1.4 L), potassium hydroxide (KOH, 910 g, 16.3 mol, 4.0 equiv), and aminoguanidine bicarbonate (1105 g, 8.13 mol, 2.0 equiv) at room temperature. The resulting reaction mixture was then heated to 75 0C for 14 h. When HPLC showed the condensation reaction was deemed complete, the reaction mixture was cooled down to room temperature before being filtered. The filtrate was then concentrated under the reduced pressure to remove the most of the solvents. The residual aqueous solution was extracted with ethyl acetate (EtOAc, 3 x 6 L). The organic layers were combined and concentrated under the reduced pressure to give a dark brown solid. This solid was dissolved in ethanol (4 L) and the resulting solution was treated with a solution of 0.2 M aqueous hydrochloric acid solution (4 L). The resulting slurry was subsequently heated to 50 0C for 6 h before being allowed to cool down to room temperature. A solution of saturated aqueous sodium bicarbonate solution (NaHCO3, 2 L) was slowly added to the slurry and the resulting mixture was then concentrated under the reduced pressure to remove most of the solvents. The aqueous residue was then treated with ethyl acetate (20 L) to dissolve the solids. The two layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 2 L). The combined organic layers were concentrated under the reduced pressure. The dark brown solids were treated with methyl ter/-butyl ether (MTBE, 4 L) and the resulting slurry was heated to 30 0C and stirred at 30 0C for 30 min. The mixture was filtered and the solids (green to orange in color) were collected (save the filtrate) and washed with methyl tert-buty\ ether (MTBE, 2 L) to give the first crop of the crude desired product (7). The filtrate was evaporated under the reduced pressure, and the resulting dark brown solids were treated with methyl tert-butyl ether (MTBE, 2 L). The resulting slurry was heated to 30 0C and stirred at 30 0C for 30 min. The mixture was filtered to give the second crop of the crude desired product (7) which was washed with MTBE (1 L). The combined solids were dried in vacuum at 40 – 45 0C to afford 6-(4-bromo-3-fluorophenyl)-l,2,4-triazin-3-amine (7, 585 g, 1095.1 g theoretical, 53.4 % yield) which was used in the subsequent reaction without further purification. For 7: 1H NMR (400 MHz, DMSO-J6) δ ppm 8.86 (s, IH), 7.97 (d, IH, J= 10.79 Hz), 7.81 (m, 2H), 7.52 (br s, 2H); C9H6BrFN4 (MW 269.07), LCMS (EI) mle 269.0/271.1 (M+ + H).

Example 13 6-((2-(4-Bromo-3-fluorophenyl)imidazo[l,2-6][l,2,4]triazin-7-yl)methyl)quinoline (12) l-(2-Chloro-l-hydroxy-3-(quinolin-6-yl)propyl)pyrrolidine-2,5-dione (11, 228 g, 0.74 mol, 1.1 equiv) and 6-(4-bromo-3-fluorophenyl)-l,2,4-triazin-3-amine (7, 181 g, 0.673 mol) were suspended in 1-butanol (1800 mL) and the resulting suspension was heated to 110 0C and stirred at 110 0C for 18 h (the reaction mixture becomes homogeneous at this point). The reaction mixture was then gradually cooled down to room temperature before being further cooled down to 10 0C in an ice bath. The resulting yellow solid was collected by filtration (save the 1 -butanol filtrates), washed with cold 1-butanol (3 x 100 mL) and dried by suction. This solid was then suspended in the saturated aqueous sodium bicarbonate solution (NaHCO3, 500 mL) and the resulting suspension was stirred at room temperature for 1 h to neutralize the corresponding hydrochloride salt. The free base was then filtered, washed with water (500 mL) and dried in a vacuum oven at 45 0C for 18 h to afford the first crop of the crude 6-((2-(4-bromo-3-fluorophenyl)imidazo[l,2-6][l,2,4]triazin-7-yl)methyl)quinoline (12, 125.1 g, 292.3 g theoretical, 42.8% yield). The 1-butanol filtrates were then concentrated under the reduced pressure and the resulting solids were dissolved in dichloromethane (CH2Cl2, 2 L). The solution was wash with the saturated aqueous sodium bicarbonate solution (NaHCO3, 1 L), dried over sodium sulfates (Na2SO4), and concentrated under the reduced pressure. The residue was then purified by flash column chromatography (SiO2, O – 10% MeOH-CH2Cl2 gradient elution) to afford the second crop of 6-((2-(4-bromo-3-fluorophenyl)imidazo[l,2-&][l ,2,4]triazin-7-yl)methyl)-quinoline (12, 19.7 g, 292.3 g theoretical, 6.7% yield; total 144.8 g, 292.3 g theoretical, 49.5% yield) as yellow solids. For 12: 1H NMR (400 MHz, DMSO-^6) δ ppm 9.23 (s, IH), 9.11 (dd, IH, J= 4.98, 1.55 Hz), 8.85 (d, IH, J= 8.09 Hz), 8.25 – 8.18 (m, 2H), 8.12 -8.00 (m, 3H), 7.93 – 7.86 (m, 3H), 4.70 (s, 2H); C21H13BrFN5 (MW 434.26), LCMS (EI) mle 434.00/435.95 (M+ + H).

Example 14 2-Fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-6][l,2,4]triazin-2-yl)benzonitrile (13)

6-((2-(4-Bromo-3-fluorophenyl)imidazo[ 1 ,2-b] [ 1 ,2,4]triazin-7-yl)methyl)quinoline (12, 200 g, 0.461 mol), zinc cyanide (ZnCN2, 32.7 g, 0.277 mol, 0.6 equiv), zinc powder (Zn, 6.0 g, 0.093 mol, 0.2 equiv) and Pd(dppf)2Cl2 (22.6 g 0.028 mol, 0.06 eqiv) were suspended in premixed solution of ΛyV-dimethyl acetamide (DMAC, 2000 mL) and water (H2O, 40 mL). The resulting suspension was then degassed with a stream of nitrogen for 20 min before being heated to 110 0C and stirred at 110 0C for 1 – 2 h (homogeneous solution was observed). When LC/MS indicated the reaction was deemed complete, the reaction mixture was cooled first to room temperature and then in an ice bath to 5 0C. The cooled reaction mixture was diluted with a mixture of the saturated aqueous ammonium chloride solution (aq. NH4Cl), the concentrated ammonium hydroxide aqueous solution (aq. NH4OH), and water (4:1 :4 by volumn, 8.1 L) and the resulting mixture was stirred at room temperature for 30 min. The resulting solids were collected by filtration and dried in a vacuum oven overnight at 45 0C to afford the crude desired product (13). This crude material was then purified by flash chromatography (SiO2, gradient elution with 1% triethylamine in dichloromethane, 2.5 % acetone and 1% triethylamine in dichloromethane, 5.0 % acetone and 1% triethylamine in dichloromethane, and 10.0 % acetone and 1% triethylamine in dichloromethane sequentially) to afford the pure 2-fluoro-4-(7-(quinolin-6-ylmethyl)-imidazo[l,2-έ][l,2,4]triazin-2-yl)benzonitrile (13, 127.4 g, 175.4 g theoretical, 72.6% yield) as yellow solids. For 13: 1H NMR (400 MHz, DMSO-^6) δ ppm 9.24 (s, IH), 8.81 (dd, IH, J= 4.15, 1.66 Hz), 8.26 – 8.12 (m, 4H), 8.02 (s, IH), 7.95 – 7.93 (m, 2H), 7.76 (dd, IH, J= 8.71, 2.08 Hz), 7.47 (dd, IH, J= 8.70, 4.15 Hz), 4.62 (s, 2H); C22HnFN6 (MW 380.38), LCMS (EI) mle 381.0 (M+ + H).

Example 15 6-(3,3-Diethoxyprop-l-ynyl)quinoline (22)

A mixture of 6-bromoquinoline (8, 2.63 g, 12.6 mmol), propargylaldehyde diethyl acetal (3.73 niL, 25.2 mmol, 2.0 equiv), triethylamine (TEA, 12.7 mL, 90.8 mmol, 7.2 equiv), copper(I) iodide (CuI, 24.0 mg, 0.126 mmol, 0.01 equiv), and triphenylphosphine (PPh3, 0.39716 g, 1.5142 mmol, 0.12 equiv) in JV^V-dimethylformamide (DMF, 15.6 mL, 202 mmol) was degassed with nitrogen bubbling for 5 min. Palladium acetate (Pd(OAc)2, 0.08499 g, 0.3786 mmol, 0.03 equiv) was added and the mixture was degassed with nitrogen bubbling for 5 min. The reaction mixture was heated to 90 0C under nitrogen with stirring. After 3 h and 10 min, HPLC indicated that the reaction was complete. The reaction mixture was diluted with ethyl acetate (EtOAc, 100 mL) and washed with water (H2O, 2 x 100 mL). The aqueous layer was extracted with ethyl acetate (EtOAc, 20 mL). The combined organic extracts were then concentrated under the reduced pressure to give the crude product as a black oil. The crude product was purified by flash column chromatography (SiO2, 0 – 40% EtOAc in hexane gradient elution) to afford 6-(3,3-diethoxyprop-l-ynyl)quinoline (22, 3.2 g, 3.22 g theoretical, 99% yield) as a colorless oil. For 22: 1H NMR (400 MHz, OMSO-d6) δ ppm 8.92 (dd, IH, J= 4.35 Hz, 1.86 Hz), 8.36 (d, IH, J= 8.40 Hz, 1.66 Hz), 8.20 (d, IH, J= 1.78 Hz), 7.99 (d, IH, J= 8.71 Hz), 7.76 (dd, IH, J= 8.71 Hz, 1.87 Hz), 7.57 (dd, IH, J= 8.09 Hz, 4.05 Hz), 5.58 (s, IH), 3.75 – 3.55 (m, 4H), 1.17 (t, 6H, J= 7.16 Hz); Ci6H17NO2 (MW 255.31), LCMS (EI) m/e 256.0 (M+ + H).

Scheme 2 (Examples 15-18)

Method C


Example 16 6-(3,3-Diethoxypropyl)quinoline (23)

Method A. 3,3-Diethoxy-l-propene (548 g, 4.2 mol, 1.75 equiv) was added to a 22 L flask charged with 0.5 M solution of 9-borabicyclo[3.3.1] nonane in tetrahydrofuran (9-BBN solution in THF, 8.4 L, 4.2 mol, 1.75 equiv) at room temperature (the internal temperature raised to 40 0C) over 1 h.. The resulting reaction mixture was stirred at room temperature for overnight. At which time 1H NMR of an aliquot of the reaction mixture indicated that all the 3,3-diethoxy-1-propene had been consumed. 6-Bromoquinoline (8, 500 g, 2.4 mol, 1.0 equiv), potassium carbonate (K2CO3, 662 g, 4.8 mol, 2.0 equiv), tricyclohexylphosphine (67.4 g, 0.24 mol, 0.1 equiv), palladium acetate (Pd(OAc)2, 27 g, 0.12 mol, 0.05 equiv) and water (90 mL) were added to the reaction mixture in that order followed by degassing with nitrogen for 0.5 h. The reaction mixture was then heated to reflux for 4 h. Once TLC and LC/MS showed that the starting material had been consumed, the reaction mixture was cooled to room temperature with stirring before being quenched with water (7.5 L) and ethyl acetate (EtOAc, 7.5 L). The layers were separated and the aqueous layer was extracted with ethyl acetate (EtOAc, 4 L). The combined organic layers were washed with a saturated brine solution (NaCl, 4 L), dried over magnesium sulfate (MgSO4) and concentrated under the reduced pressure. The residue was purified by column chromatography (SiO2, 10 – 60% of ethyl acetate in heptane gradient elution) to afford 6-(3,3-diethoxypropyl)quinoline (23, 520 g, 622.4 g theoretical, 83.5% yield) as a colorless oil. For 23: 1HNMR (DMSO-</6, 300MHz) δ ppm 8.81 (dd, IH, J= 4.23 Hz, 1.73 Hz), 8.28 (d, IH, J= 8.07 Hz), 7.91 (d, IH, J= 8.62 Hz ), 7.75 (s, IH), 7.61 (dd, lH, J= 8.63 Hz, 1.92 Hz), 7.46 (dd, IH, J= 8.25 Hz, 4.22 Hz), 4.46 (t, IH, J= 5.60 Hz), 3.61 – 3.38 (m, 4H), 2.79 (t, 2H, J= 8.53 Hz), 1.95 -1.85 (m, 2H), 1.11 (t, 6H, J= 6.84 Hz); Ci6H21NO2 (MW 259.34), LCMS (EI) m/e 260.2 (M+ + H).

Method A-Alternative. 9-BBN was generated in situ and used to prepare compound 23 as discribed as follows: under a nitrogen atmosphere anhydrous 1 ,2-dimethoxyethane (DME, 47.0 mL) was charged into a 500 mL 3-neck flask equipped with a distillation apparatus. Borane-dimethyl sulfide complex (12.1 g, 151 mmol, 2 equiv) was added and the solution temperature increased from 20 to 22 0C. To this solution, 1 ,5-cyclooctadiene (16.3 g, 151 mmol, 2 equiv) was added dropwise over a period of 30 min to maintain a reaction temperature of 50 – 60 0C, during which time a small amount of dimethyl sulfide was collected by the distillation apparatus. The reaction mixture was then distilled under nitrogen until the distillate temperature reach 84 0C. The distillates collected had a volume of ~ 21 mL. The oil bath was removed and anhydrous THF (49 mL) was added. A small sample of the reaction mixture was taken for 1H NMR analysis and the result indicated the olefin was consumed. This 9-BBN solution was used directly for the next step.

To the above 9-BBN solution, 3,3-diethoxy-l-propene (19.3 g, 142 mmol, 1.89 equiv) was added dropwise while maintaining the temperature below 30 0C. The reaction is slightly exothermal and white precipitate slowly dissolved. The reaction mixture was then stirred at room temperature for 18 h.

To the solution prepared above, 6-bromoquinoline (8, 15.7 g, 75.4 mmol, 1 equiv), tricyclohexylphosphine (1.27 g, 4.52 mmol, 0.06 equiv), potassium carbonate (20.8 g, 151 mmol, 2 equiv), and water (0.421 mL, 23.4 mmol) were added. The mixture was degassed with nitrogen bubbling for 10 – 15 min. Palladium acetate (Pd(OAc)2, 0.508 g, 2.26 mmol, 0.03 equiv) was added and the nitrogen bubbling was continued for an additional 10 min. The reaction mixture was heated to 75 0C and maintained at 75 – 78 0C for 2 – 3 h. When HPLC showed the completion of the reaction, the heating was discontinued and the reaction mixture was cooled to room temperature. Ethyl acetate (EtOAc, 162 mL) and water (H2O, 162 mL) were added and the organic layer was separated. The aqueous layer was extracted with ethyl acetate (EtOAc, 2 x 60 mL) and the combined organic extracts were dried over sodium sulfate (Na2SO4) and concentrated under the reduced pressure. The residue was purified by flash column chromatography (silica gel, 0 – 40% EtOAc in hexane gradient elution) to afford 6-(3,3-diethoxypropyl)quinoline (23, 17.6 g, 19.6 g theoretical, 90% yield) as a clear oil, which was found to be identical to the meterial made from Method A in every comparable aspect.

Method B. A mixture of 6-(3,3-diethoxyprop-l-yn-l-yl)quinoline (22, 56 mg, 0.22 mmol) and 10% palladium on carbon (5 mg) in THF (5 mL) was hydrogenated under H2 at 1 atm for 6 h. The reaction mixture was filtered through a celite bed and the celite bed was washed with THF (2 x 2 mL). The combined filtrates were concentrated under the reduced pressure to afford 6-(3,3-diethoxypropyl)quinoline (23, 56 mg, 57 mg theoretical, 98% yield) as a clear oil, which was found to be sufficiently pure to be used in the subsequent reaction without further purification and was identical to the meterial made from Method A in every comparable aspect.

Example 17 3-(Quinolin-6-yl)propanal (9)

Method 1. A 22 L flask was charged with tris(dibenzylideneacetone)dipalladium(0) (70.0 g, 0.076 mol, 0.015 equiv), tri-tert-butylphosphonium tetrafluoroborate (44 g, 0.152 mol, 0.03 equiv), and dioxane (12 L) at room temperature. The resulting solution was then degassed with a steady stream of nitrogen for 20 min before 6-bromoquinoline (8, 1055 g, 5.07 mol, 1.0 equiv), allyl alcohol (588 g, 10.1 mol, 2.0 equiv), and 7V-methyl-iV-cyclohexylcyclohexylamine (1186 g, 6.08 mol, 1.2 equiv) were added at room temperature. The resulting reaction mixture was stirred at 50 – to 55 °C for 8 – 12 h. When TLC and LC/MS showed that the reaction was deemed complete, the reaction mixture was cooled to room temperature before methyl fert-butyl ether (MTBE, 10 L) was added to the reaction mixture. The resulting mixture was stirred at room temperature for 10 min before being filtered through a plug of celite. The filtrate was concentrated under the reduced pressure and the residue was purified by flash column chromatography (SiO2, 20 – 80 % ethyl acetate in heptane gradient elution) to afford 3-(quinolin-6-yl)propanal (9, 495 g, 939.1 g theoretical, 52.7%) as a yellow oil, which solidified partially upon standing at 0 – 5 0C. For 9: 1H NMR (400 MHz, DMSO-J6) δ ppm 9.75 (t, IH, J= 1.24 Hz), 8.83 (dd, IH, J= 4.15 Hz, 1.66 Hz), 8.25 (dd, IH, J= 8.3, 1.03 Hz), 7.93 (d, IH, J= 8.71 Hz), 7.76 (d, IH, J= 1.45 Hz), 7.64 (dd, IH, J= 8.72 Hz, 2.08 Hz), 7.48 (dd, IH, J= 8.30 Hz, 4.36 Hz), 3.05 (t, 2H, J= 7.26 Hz), 2.89 (t, 2H, J= 7.26 Hz); Ci2HnNO (MW 185.22), LCMS (EI) We 186 (M+ + H).

Method 2. A solution of 6-(3,3-diethoxypropyl)quinoline (23, Method A of Example 16, 520 g , 2.08 mol, 1.0 equiv) in ethyl acetate (EtOAc, 2.2 L) was cooled to 0 0C before a 2 N aqueous hydrochloric acid (HCl) solution (2.2 L) was added over 1 h while keeping the reaction temperature below 5 0C. The resulting reaction mixture was stirred for an additional 2 h at 0 – 5 0C. When TLC and HPLC/MS indicated the reaction was complete, the reaction was quenched with an ice cold 3 N aqueous sodium hydroxide (NaOH) solution at 0 °C until the pH was between 8 to 9. The layers were separated and the aqueous layer was extracted with ethyl acetate (EtOAc, 2 L). The combined organic layers were washed with brine (2 L), dried with sodium sulfate (Na2SO4), and concentrated under the reduced pressure to afford crude 3-(quinolin-6-yl)propanal (9, 385.3 g, 385.3 g theoretical, 100%) as a yellow oil, which was found to be identical to the material obtained from Method 1 in every comparable aspect. Since this crude material was found to be sufficiently pure, it was used directly in subsequent reaction without further purification.

Method 5. A 22 L flask charged with 0.5 M solution of 9-borabicyclo[3.3.1] nonane in tetrahydrofuran (9-BBN, 5.75 L, 2.89 mol, 2.0 equiv) and tetrahydrofuran (THF, 6 L) was treated with 3,3-diethoxy-l-propene (393 g, 3.02 mol, 2.10 equiv) at 0 – 5 0C and the resulting reaction mixture was subsequently warmed to room temperature and stirred at room temperature for 14 h. 6-Bromoquinoline (8, 300 g, 1.44 mol, 1.0 equiv), palladium acetate (Pd(OAc)2, 16.1 g, 0.072 mol, 0.05 equiv), potassium carbonate (K2CO3, 398 g, 2.89 mol, 2.0 equiv), tricyclohexylphosphine (22.3 g, 0.079 mol, 0.055 equiv), and water (52 g, 2.8 mol) were added to the reaction mixture at room temperature before being degassed with nitrogen for 1 h. The resulting reaction mixture was heated to 75 0C for 1 h. When TLC and LC/MS showed the reaction was deemed complete, the reaction mixture was cooled to room temperature and water (2 L) was added to dissolve the salts. The resulting mixture was then concentrated under the reduced pressure to a volume of approximately 4 L before being filtered through a plug of Celite. The Celite plug was washed with ethyl acetate (EtOAc, 2 L). The filtrate was concentrated under the reduced pressure to a volume of approximately 2 L and this residual solution was then added slowly over 5 min to a flask containing a 2.0 M aqueous hydrochloric acid (HCl) solution (2 L) at 0 – 5 °C. The resulting solution was stirred at 0 – 5 °C for 14 h before being quenched with saturated aqueous sodium bicarbonate (NaHCO3) solution at 0 0C until the pH was between 8 to 9. The layers were separated and the aqueous layer was extracted with ethyl acetate (EtOAc, 2 L). The combined organic layers were washed with brine (1 L), dried with sodium sulfate (Na2SO4), and concentrated under the reduced pressure. The residue, which contains the crude 3-(quinolin-6-yl)propanal (9) was purified by flash column chromatography (SiO2, 20 – 80 % ethyl acetate in heptane gradient elution) to afford 3-(quinolin-6-yl)propanal (9, 139 g, 266.7 g theoretical, 52.1%) as a yellow oil, which was found to be identical to the material obtained from Methods 1 and 2.

Example 18 l-(2-Chloro-l-hydroxy-3-(quinolin-6-yI)propyl)pyrrolidine-2,5-dione (11)

Method I. A solution of 3-(quinolin-6-yl)propanal (9, 407 g, 2.2 mol, 1.0 equiv) in chloroform (CHCl3, 1700 mL) was cooled to 0 0C before proline (52 g, 0.44 mol, 0.2 equiv) and iV-chlorosuccinimide (NCS, 303 g, 2.31 mol, 1.05 equiv) were added. The resulting reaction mixture was allowed to slowly warm to room temperature (becomes homogeneous) and stirred at room temperature for overnight. The reaction was exothermal to around 40 0C when it reaches room temperature and a precipitate had formed at this point. Once TLC and LC/MS showed that the reaction was deemed complete, the reaction mixture was diluted with ethyl acetate (EtOAc, 1700 mL) and the resulting mixture was cooled to 0 0C. The solid was collected by filtration and the collected wet solid cake was placed in a flask and triturated with water (750 mL). The resulting suspension was stirred at room temperature for 30 min before the solids were collected by filtration. The collected solids were washed with water (250 mL) and methyl tert-bntyl ether (MTBE, 500 mL) and dried in a vacuum oven at 45 0C to constant weight to afford l-(2-chloro-l-hydroxy-3-(quinolin-6-yl)propyl)pyrrolidine-2,5-dione (11, 378.7 g, 701.3 g theoretical, 54 % yield) as off-white powder. For 11: 1HNMR (DMSO-J6, 400MHz) δ ppm 8.86 (dd, IH, J= 4.15 Hz, 1.66 Hz), 8.33 (dd, IH, J= 8.51 Hz, 1.04 Hz), 7.98 (d, IH, J= 8.72 Hz), 7.85 (d, IH, J= 1.66 Hz), 7.68 (dd, IH, J= 8.51 Hz, 1.87 Hz), 7.51 (dd, IH, J= 8.29 Hz, 4.15 Hz), 7.36 (d, IH, J = 7.05 Hz), 5.28 (dd, IH, J= 9.54 Hz, 6.85 Hz), 5.07 (dt, IH, J= 9.75 Hz, 2.70 Hz), 3.65 (dd, IH, J= 14.52 Hz, 2.49 Hz), 3.09 (dd, IH, J= 14.52 Hz, 9.75 Hz), 2.64 (s, 4H); C16H15ClN2O3 (MW 318.75), LCMS (EI) m/e 319.2 (M+ + H).

Method II. A solution of 3-quinolin-6-ylpropanal (9, 74.8 g, 0.404 mol) in acetonitrile (202 mL, 3.87 mol) was cooled to 0 0C before L-proline (4.70 g, 0.0404 mol, 0.10 equiv), benzoic acid (4.96 g, 0.0404 mol, 0.10 equiv), and iV-chlorosuccinimide (NCS, 57.8 g, 0.424 mol, 1.05 equiv) were added at 0 0C. The reaction mixture was stirred at 0 °C for 3 h and the resulting clear solution was allowed to warm to room temperature and stirred at room temperature for 18 h. The reaction mixture became a thick suspension and LCMS showed the completion of the reaction. Ethyl acetate (EtOAc, 202 mL) was added to the reaction mixture and the resulting mixture was stirred at room temperature for 1 h. The solids were collected by filtration, washed with ethyl acetate (EtOAc, 100 mL) and dried under vacuum at 40 – 45 0C to constant weight to afford l-(2-chloro-l-hydroxy-3-(quinolin-6-yl)propyl)pyrrolidine-2,5-dione (11, 88.8 g, 128.8 g theoretical, 69 % yield) as an off-white powder, which was found to be identical to the material made from method I in every comparable aspect.

Scheme 3 (Examples 19-21)

15 21, dihydrochloride

C23H17FN6O C23H19Q2FN6O MoI Wt 412 42 MoI Wt 485 34

Example 19

2-Fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-Z>] [l,2,4]triazin-2-yl)benzoic acid (14)

A suspension of 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l ,2-&][l ,2,4]triazin-2-yl)benzonitrile (13, 277.5 g, 0.73 mol, 1.0 equiv) in concentrated hydrochloric acid (2500 mL) and water (250 mL) was heated to 1000C (homogenous at this point) and stirred at around 100 0C for 18 h. When LC/MS indicated the reaction was deemed complete, the reaction mixture was cooled down to 70 – 80 0C before being diluted with water (2500 mL). The resulting diluted reaction mixture was then cooled down to room temperature (yellow solid forms at 40 – 50 0C) and subsequent to 0 – 5 0C. The solids were then collected by filtration, washed with a small amount of IN aqueous HCl (100 mL), and dried in a vacuum oven at 45 0C to constant weight to afford 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-έ][l,2,4]triazin-2-yl)benzoic acid (14, 271 g, 291.5 g theoretical, 93% yield) as yellow to bright-yellow powders. For 14: 1H NMR (400 MHz, OMSO-d6) δ ppm 9.34 (s, IH), 9.23 (dd, IH, J- 5.19 Hz, 1.45 Hz), 9.08 (d, IH, J= 8.29 Hz), 8.38 (d, IH, J= 8.92 Hz), 8.30 (d, IH, J= 1.24 Hz), 8.18 (dd, IH, J= 8.72 Hz, 1.87 Hz), 8.12 (s, IH), 8.08 – 8.00 (m, 4H), 4.75 (s, 2H); C22H16Cl2FN5O2 (MW 472.30), C22H14FN5O2 (free base: MW 399.38), LCMS (EI) mle 400.0 (M+ + H).

Example 20 2-Fluoro-7V-methyl-4-(7-(quiiiolin-6-ylmethyl)imidazo[l,2-^][l,2,4]triazin-2-yl)benzainide

(15).

A suspension of 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l ,2-b][l ,2,4]triazin-2-yl)benzoic acid (14, 431.4 g, 0.914 mol, 1.0 equiv) and (benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyBOP, 570 g, 1.1 mol, 1.2 equiv) in ΛyV-dimethylformamide (DMF, 3700 mL) was treated with a solution of 2 M methylamine in THF (1830 mL, 3.656 mol, 4.0 equiv) over 15 min at room temperature. The reaction temperature increased to 30 0C during the addition of methylamine and the reaction mixture became homogeneous once the addition of methylamine was complete. Triethylamine (TEA, 382 mL, 2.742 mol, 3.0 equiv) was then added to the reaction mixture and the resulting reaction mixture was stirred at room temperature for 2 – 4 h. When LC/MS showed the coupling reaction was deemed complete, the reaction mixture was treated with water (950 mL). The resulting suspension was cooled down to 0 – 5 0C in an ice-bath and stirred at 0 – 5 0C for 30 min. The solids were collected by filtration and washed with water (200 mL). The wet solid cake was then suspended in a mixture of water and acetonitrile (1/1 by volume, 2000 mL) and the resulted suspension was stirred at room temperature for Ih. The solids were collected by filtration, washed with water and acetonitrile, and dried in a vacuum oven at 40 – 45 0C to constant weight to afford 2-fluoro-Λ/-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[l ,2-έ>][l ,2,4]triazin-2-yl)benzamide (15, 322 g, 377 g theoretical, 85.4% yield) as yellow to bright-yellow powders. For 15: 1H NMR (400 MHz, DMSO-J6) δ ppm 9.20 (s, IH), 8.82 (dd, IH, J= 4.05, 1.56 Hz), 8.38 (br m, IH), 8.27 (dd, IH, J= 8.50 Hz, 1.25 Hz), 8.06 – 7.93 (m, 5H), 7.81 – 7.74 (m, 2H), 7.49 (dd, IH, J= 8.40 Hz, 4.35 Hz), 4.62 (s, 2H), 2.78 (d, 3H, J= 4.36 Hz); C23H17FN6O (MW 412.42), LCMS (EI) mle 413.1 (M+ + H).

Example 21

2-Fluoro-Λr-methyl-4-(7-(quinoIin-6-ylmethyl)imidazo[l,2-6][l,2,4]triazm-2-yl)benzamide dihydrochloride (21, dihydrochloride)

A suspension of 2-fluoro-iV-methyl-4-[7-(quinolin-6-ylmethyl)imidazolo[l ,2-6][l,2,4]triazin-2-yl]benzamide (15, 421.2 g, 1.021 mol) in methanol (MeOH, 6600 mL) was heated to 55 0C before a premixed solution of aqueous concentrated hydrochloric acid (cone.

HCl, 37 wt.%, 12 M, 420 mL, 5.10 mol, 5.0 equiv) in isopropyl alcohol (IPA, 1510 mL) was added dropwise at 55 0C. The resulting clear solution was stirred at 55 0C for 30 min before methyl tert-butyl ether (MTBE, 6750 mL) was added via an additional runnel over 30 min. The solids were slowly precipitated out after addition of methyl tert-butyl ether. The resulting mixture was stirred at 55 0C for an additional 1 h before being gradually cooled down to room temperature. The mixture was stirred at room temperature for overnight. The solids were collected by filtration, washed with methyl tert-butyl ether (MTBE, 3 x 500 mL), and dried in vacuum oven at 45 – 55 0C to constant weight. The desired 2-fluoro-Λr-methyl-4-[7-(quinolin-6-ylmethyl)imidazolo[l,2-£][l,2,4]triazin-2-yl]benzamide dihydrochloride (21, dihydrochloride, 470.7 g, 495.5 g theoretical, 95% yield) was obtained as off-white to light yellow crystalline solids. For 21 (dihydrochloride): mp (decom.) 222 0C; 1H NMR (400 MHz, DMSO-J6) δ ppm 9.46 (s, IH), 9.25 (dd, IH, J= 5.4 Hz, 1.4 Hz), 9.12 (d, IH, J= 8.3 Hz), 8.51 (m, IH), 8.47 (d, IH, J= 0.9 Hz), 8.34 (d, IH, J= 1.3 Hz), 8.23 (s, IH), 8.21 (dd, IH, J= 9.0 Hz, 1.8 Hz), 8.09-8.02 (m, 3H), 7.79 (dd, IH, J= 7.5 Hz, 8.3 Hz), 4.77 (s, 2H), 2.78 (s, 3H, J= 4.5 Hz); 13C NMR (100 MHz, DMSO-^6) δ ppm 163.4, 159.4 (d, J= 249.9 Hz), 145.8, 145.4, 144.5, 143.8, 140.4, 138.8, 136.8, 135.9, 135.7 (J= 8.6 Hz), 131.2 ( J= 3.1 Hz), 130.7, 128.7, 128.2, 126.2 (J- 14.9 Hz), 126.0, 123.1 (J= 3 Hz), 122.5, 121.0, 114.9 (J= 5.6 Hz), 28.4, 26.3; 19F NMR (376.3 MHz, DMSO-^6) δ ppm -113.2; C23H17FN6O (free base, MW 412.42), LCMS (EI) mle 413.1 (M+ + H) and 435.0 (M+ + Na).

Scheme 4 (Examples 22-25)

C7H3BrFN C13H15BFN2O2 MoI. Wt: 200.01 MoI. Wt: 247.07 Example 22 l,2,4-Triazin-3-amine (16)

An aqueous solution of glyoxal (57 Kg of 40 wt% aqueous solution, 393 mol, 0.73 equiv) was added to a suspension of aminoguanidine bicarbonate (73 Kg, 536.3 mol) in water (400 L) at room temperature. The evolution of carbon dioxide (CO2) began almost immediately. The reaction mixture was then stirred at room temperature for 18 h and the evolution of gas had virtually ceased after about 2 h. The reaction mixture was then filtered, and the filtrate was evaporated to dryness under the reduced pressure. The residue was then extracted with cold methanol (MeOH, 3 x 120 L), and the combined methanol solution was cooled down to 0 – 5 0C before being filtered to remove the residual solids. The filtrate was then concentrated under the reduced pressure, and the residue was recrystallized in acetonitrile to afford l,2,4-triazin-3-amine (16, 34 Kg, 37.76 Kg theoretical, 90% yield) as fine, white needles. For 16: 1H NMR (400 MHz, DMSO-J6) δ ppm 8.54 (d, IH, J- 2.33 Hz), 8.20 (d, IH, J= 2.33 Hz), 7.15 (br s, 2H).

Example 23 6-Bromo-l,2,4-triazin-3-amine (17)

A solution of 1 ,2,4-triazin-3-amine (16, 33 Kg, 343.4 mol) in water (500 L) and acetonitrile (300 L) was treated with jV-bromosuccinimide (NBS, 66 Kg, 370 mol, 1.08 equiv) at 5 – 15 0C, and the resulting reaction mixture was stirred at 10 – 15 0C for 1 – 4 h. When TLC and LC/MS showed that the bromination reaction was deemed complete, the reaction mixture was treated with an aqueous solution of saturated sodium carbonate (Na2CO3). The resulting solution was then extracted with ethyl acetate (EtOAc, 3 x 500 L). The combined organic extracts were washed with water (2 x 100 L), dried over magnesium sulfate (MgSO4), and concentrated under the reduced pressure to afford 6-bromo-l,2,4-triazin-3-amine (17, 10.3 Kg, 60 Kg theoretical, 17.2% yield) as yellow to brown powders. For 17: 1H NMR (400 MHz, DMSO-J6) δ ppm 8.39 (s, IH), 7.47 (br, 2H); C3H3BrN4 (MW 174.99), LCMS (EI) mle 175.0/176.9 (M+ + H).

Example 24 2-Fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile (19) Step 1. A solution of 2-fluro-4-bromobenzonitrile (18, 12.5 Kg, 62.5 mol) in anhydrous tetrahydrofuran (THF, 30 L) was treated with a solution of isopropylmagnesium chloride generated from magnesium (Mg, 1.8 Kg, 150 mol, 1,2 equiv) an 2-chloropropane (7.2 Kg, 92 mol, 1.47 equiv) in THF (20 L) and 2-(2-(dimethylamino)ethoxy)-τV/vr-dimethylethanamine (11 Kg, 69 mol, 1.1 equiv) at room temperature. The resulting mixture was then stirred at 12 – 20 0C for an additional 2 h before being treated with trimethylborate (9 Kg, 86.7 mol, 1.4 equiv) at 10 -15 0C. The reaction mixture was stirred at 7 – 16 0C for 40 min. When TLC and LC/MS showed that the reaction was deemed complete, the reaction mixture was quenched with 1 N aqueous hydrochloric acid (HCl, 35 Kg) at room temperature. The quenched aqueous reaction mixture was then extracted with ethyl acetate (EtOAc, 4 x 35 L). The combined organic extracts were washed with water (50 L), dried over magnesium sulfate (MgSO4), and concentrated under the reduced pressure. The residual solids were then recrystallized from acetonitrile (20 L) and hexanes (45 L) to afford the corresponding crude 3-fluoro-4-cyanophenyl boronic acid (5.0 Kg, 48% yield).

Step 2. A suspension of the crude 3-fluoro-4-cyanophenyl boronic acid (9.2 Kg, 55.8 mol) in cyclohexane (150 L) was treated with pinacol (13.2 Kg, 111.6 mol, 2.0 equiv) at room temperature, and the resulting reaction mixture was warmed to 40 0C for 4 h. When TLC and LC/MS showed that the reaction was deemed complete, the reaction mixture was cooled down to room temperature before being washed with water (2 x 75 L). The organic layer was then dried over magnesium sulfate (MgSO4) and concentrated under the reduced pressure to afford 2-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile (19, 11.8 Kg, 13.8 Kg theoretical, 85.6% yield) as a light yellow solid. For 19: 1H NMR (300 MHz, DMSO-J6) δ ppm 7.92 (t, IH, J- 7.00 Hz), 7.62 (m, 2H), 1.29 (s, 12 H).

Example 25 4-(3-Amino-l,2,4-triazin-6-yI)-2-fluorobenzonitrile (20).

A mixture of 6-bromo- 1,2, 4-triazin-3 -amine (17, 100.0 g, 571.47 mmol) and 2-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile (19, 145.43 g, 588.61 mmol, 1.03 equiv) in 1,4-dioxane (1200 mL) was stirred at room temperature for 10 min before potassium carbonate (K2CO3, 355.4 g, 2572 mmol) in water (600 mL) was added to give a deep red solution. The mixture was degassed by bubbling with nitrogen for 10 min before 1,1′-bis(diphenyl phosphino)ferrocene dichloropalladium(II) complex with dichloromethane (1 :1) (Pd(dppf)2Cl2, 14.14 g, 17.14 mmol, 0.03 equiv) was added at room temperature. The resulting reaction mixture was degassed by bubbling with nitrogen for 10 min and then heated at 86 0C under nitrogen. After 2 h, HPLC showed that the reaction was deemed complete, and the reaction mixture was cooled to room temperature and then to 0 – 5 0C with an ice-water bath. 1 ,4-Dioxane (400 mL) was added to the cooled reaction mixture before a solution of 3.3 M aqueous hydrochloric acid solution (HCl, 1900 mL) was added dropwise with stirring to adjust pH to 0.40- 0.93. The mixture was stirred at room temperature for 30 min and filtered. The solid collected was stirred with 1,4-dioxane (260 mL) and then added IN HCl (400 mL). The mixture was stirred at room temperature for 10 min and filtered. The filtrate was combined with the filtrate obtained earlier and washed with ethyl acetate (EtOAc, 2 x 2 L). The combined ethyl acetate extracts was extracted with 1 N aqueous hydrochloric acid solution (HCl, 3 x 200 mL). The combined aqueous solution was then treated with activated charcoal (20 g) and stirred at room temperature for 30 min. The mixture was filtered through a celite bed and the filtrate was cooled to 0 – 5 0C with an ice- water bath. A solution of 50% of sodium hydroxide in water (NaOH, 240 mL, 4500 mmol) was added drowise at 5-12 0C to adjust pH tolθ.6 – 11.6. The mixture was stirred at 0 – 5 0C for 30 min and then filtered. The solids collected were washed with aqueous ammonium hydroxide (1 to 3 of 28% concentrated NH4OH to water, 1900 mL) and dried under vacuum at 40 – 45 0C to constant weight to afford 4-(3-amino-l,2,4-triazin-6-yl)-2-fluorobenzonitrile (20, 101.2 g, 122.9 g theoretical, 82.3% yield) as a off-white powder. For 20: 1H NMR (400 MHz, DMSO-J6) δ ppm 8.94 (s, IH), 8.12 (d, IH, J= 11.41 Hz), 8.08 – 8.00 (m, 2 H), 7.71 (br s, 2 H); Ci0H6FN5 (MW 215.19), LCMS (EI) mle 215.9 (M+ + H).

Scheme 5 (Example 26)

20 13

C10H6FN5 C16H15ON2O3 C22H13FN6 MoI. Wt: 215.19 MoI. Wt 318.75 MoI. Wt: 380.38 Example 26 2-Fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-Z>][l,2,4]triazin-2-yl)benzonitrile (13).

Step 1. A 22 L reactor equipped with a overhead stirring, a thermocouple, a distillation apparatus, and a nitrogen inlet was purged with nitrogen before 4-(3-amino-l,2,4-triazin-6-yl)-2-fluorobenzonitrile (20, 300 g, 1.39 mol), l-(2-chloro-l-hydroxy-3-(quinolin-6-yl)propyl)pyrrolidine-2,5-dione (11, 635 g, 1.99 mol, 1.43 equiv), and ethylene glycol (3.0 L) were charged to the reactor at room temperature. The resulting reaction mixture was heated to 130-140 °C with nitrogen bubbled through continuously. The distillate was collected with the distillation apparatus. After 3 – 4 h, HPLC indicated the reaction was deemed complete (presence of < 1.5% of starting material 20). The reaction mixture was gradually cooled to room temperature. A 2.5% aqueous sodium carbonate solution (Na2CO3, 14.1 L) was added with stirring to the reactor over 60 min and the mixture was stirred at room temperature for 1 – 2 h. The mixture was then filtered, and the solid was washed with water (9.6 L) and dried under vacuum to afford the desired crude product (13, 980.4 g), which was combined with several other batches for purification as described below.

Step 2. A solution of crude product (13, 2754 g) in methylene chloride (CH2Cl2, 37.8 L) and methanol (0.54 L) was treated with silica gel (SiO2, 2700 g) at room temperature, and the resulting mixture was stirred at room temperature for 90 min. The mixture was filtered and the filter cake was washed with a mixture OfCH2Cl2(18 L) and methanol (0.26 L). The combined filtrates were treated with silica gel (SiO2J 800 g) and the resulting mixture was stirred at room temperature for 90 min and then filtered. The filter cake was washed with a mixture of CH2Cl2 (18 L) and methanol (0.26 L). The combined filtrates were concentrated under the reduced pressure at 20 – 60 0C to about 8 – 12 L. The residue was treated with a mixture of isopropanol (IPA) and water (1 : 1 , 9 L) in portions and the distillation was continued at 1 atm pressure until the temperature reached 68 – 75 0C. The mixture was cooled to room temperature and the solids were collected by filtration. The solids collected were washed with isopropanol (IPA, 3.6 L) and dried under vacuum at 40 – 45 0C to constant weight to afford pure 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-ό][l,2,4]triazin-2-yl)benzonitrile (13, 940.27g) as a bright yellow powder.

The above reaction and purification process gave product 13 in 59 – 64% yield. The spectroscopic data of compound 13 made by this synthetic process was found to be identical to those obtained from material made by cyanation of compound 12 described previously. For 13: 1H NMR (400 MHz, OMSO-d6) δ ppm 9.24 (s, IH), 8.81 (dd, IH5 J= 4.15, 1.66 Hz), 8.26 – 8.12 (m, 4H), 8.02 (s, IH), 7.95 – 7.93 (m, 2H), 7.76 (dd, IH, J= 8.71, 2.08 Hz), 7.47 (dd, IH, J = 8.70, 4.15 Hz), 4.62 (s, 2H); C22Hi3FN6 (MW 380.38), LCMS (EI) m/e 381.0 (M+ + H).

Scheme 6 (Examples 27-29)

14 15

C22H14FN5O2 C23H17FN6O MoI. Wt: 399.38 MoI. Wt: 412.42

I I) SOd2 aq. HCl/ acetone { 2) MeNH2

15 (.{hydrochloride

C23H17FN6O C23H19CI2FN6O MoI. Wt: 412.42 MoI. Wt: 485.34

Example 27

2-Fluoro-4-(7-(quinolin-6-ylmethyl)imidazo [1,2-6] [1,2,4] triazin-2-yl)benzoic acid (14).

To a 22 L reactor equipped with a overhead stirring, a thermocouple, and a nitrogen inlet was charged compound 13 (900 g, 2.37 mol), water (0.9 L), and concentrated HCl (9.1 L) at room temperature. The resulting reaction mixture was heated at 100 0C for 12 h. When HPLC showed the reaction was complete, the reaction mixture was cooled to 90 0C and water (4.9 L) was added over 15 min while maintaining the temperature at 65 – 90 0C. The reaction mixture was further cooled to room temperature and stirred at room temperature for 3 h. The solids were collected by filtration, washed with water (1.2 L) and dried in vacuum at 40 – 45 0C to constant weight to afford 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-6][l,2,4]triazin-2-yl)benzoic acid (14, 945 g, 946.5 g theoretical, 99.8% yield) as a light yellow solid, which was found to be identical to the material made by earlier method. For 14: 1H NMR (400 MHz, DMSO-J6) δ ppm 9.34 (s, IH), 9.23 (dd, IH, J= 5.19 Hz, 1.45 Hz), 9.08 (d, IH5 J= 8.29 Hz), 8.38 (d, IH, J = 8.92 Hz), 8.30 (d, IH, J= 1.24 Hz), 8.18 (dd, IH, J= 8.72 Hz, 1.87 Hz), 8.12 (s, IH), 8.08-8.00 (m, 4H), 4.75 (s, 2H); C22Hi6Cl2FN5O2 (MW 472.30), C22H14FN5O2 (free base: MW 399.38), LCMS (EI) mle 400.0 (M+ + H).

Example 28

2-Fluoro-iV-methyl-4-(7-(quinolin-6-yImethyl)iinidazo [ 1 ,2-6] [ 1 ,2,4] triazin-2-yl)benzamide

(15).

Method A. To a stirred solution of 2-fluoro-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-6][l,2,4]triazin-2-yl)benzoic acid (14, 1000 g, 2.12 mol) in acetonitrile (5 L) and CH2Cl2(10 L) were charged HOBt (358 g, 2.65 mol, 1.25 equiv), and EDC hydrochloride (508.4 g, 2.65 mol, 1.25 equiv) at room temperature. Another portion OfCH2Cl2 (10 L) was then added to the reaction mixture and the resulting reaction mixture was stirred at room temperature for 20 min. A 2.0 M solution of methylamine (MeNH2) in THF (3.44 L, 6.88 mol, 3.25 equiv) was added with stirring while maintaining the temperature at 15 – 30 0C. The reaction mixture was stirred at room temperature for 2 h before an additional portion of 2.0 M solution of methylamine (MeNH2) in THF (1.06 L, 2.12 mol, 1 equiv) was added. The reaction mixture was stirred at room temperature for 1 h and a second portion of EDC hydrochloride (406 g, 2.12 mol, 1 equiv) was added and the stirring was continued for 6 h. When HPLC showed less than 1 % of starting material (14) was remaining, the reaction mixture was concentrated under the reduced pressure at < 50 0C. During distillation acetonitile (20 L) was added and distillation was continued until the remaining volume was about 20 L. The residue was treated with an aqueous solution of 2.5% sodium carbonate (Na2CO3, 40 L) and the resulting mixture was stirred at room temperature for 30 min. The solids were collected by filtration, washed with water (3 x 4.0 L), air dried by pulling vacuum on the filter to afford the crude desired product (15). The crude solids were treated with CH2Cl2 (17.6 L) and MeOH (5.2 L) at room temperature and resulting mixture was stirred until a clear solution was obtained. The solution was filtered to remove insoluble materials. With vigorous stirring a 2.5% aqueous solution of sodium carbonate (Na2CO3, 17.6 L) was added to the filtrate and the mixture was stirred at room temperature for 60 min to give a suspension. Heptane (20 L) was added and the mixture was stirred for an additional 60 min. The mixture was filtered and the solid was washed sequentially with water (3 x 4.0 L) and heptane (4.0 L), and dried in vacuum to afford 2-fluoro-./V-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-b][l,2,4]triazin-2-yl)benzamide (15, 1095.3 g, 874.3 g theoretical) as a bright yellow solid, which was found to be not totally dry and to contain ~ 25% residual solvents. This wet solid was used directly for the subsequent dihydrochloride salt (21) formation reaction without further drying. A small sample was dried completely for spectroscopic analyses and the data were consistent with those obtained by earlier method: For 15: 1H NMR (400 MHz, DMSO-J6) δ ppm 9.20 (s, IH), 8.82 (dd, IH, J= 4.05, 1.56 Hz), 8.38 (br m, IH), 8.27 (dd, IH, J = 8.50 Hz, 1.25 Hz), 8.06 – 7.93 (m, 5H), 7.81 – 7.74 (m, 2H), 7.49 (dd, IH, J= 8.40 Hz, 4.35 Hz), 4.62 (s, 2H), 2.78 (d, 3H, J= 4.36 Hz); C23HnFN6O (MW 412.42), LCMS (EI) m/e 413.1 (M+ + H).

Method B. 2-Fluoro-4-[7-(quinolin-6-ylmethyl)imidazo[l,2-b][l,2,4]triazin-2-yl]benzoic acid dihydrochloride (14, 50.00 g, 0.1059 mol) was added toluene (300 mL) and followed by thionyl chloride (SOCl2, 77.2 mL, 1.06 mol, 10.0 equiv) at room temperature. The resulting reaction mixture was heated at 72 0C under N2 and the reaction was followed by HPLC analysis of the disappearance of the starting material benzoic acid (14). After 48 h, HPLC indicated ~4% starting material remaining and the reaction was stopped. The reaction mixture was concentrated to dryness by vacuum distillation at 40-50 0C. The residual solids were added toluene (300 mL) and the solvent was removed by vacuum distillation at 40-50 0C. THF (250 mL) was added and the mixture was cooled with an ice-water bath. A 2.0 M of methylamine (MeNH2) in THF (529 mL, 1.06 mol, 10 equiv) was added dropwise. The resulting reaction mixture was allowed to warm up to room temperature and stirred at room temperature for 17 h. Water (600 mL) was added to the reaction mixture and THF (400 – 500 mL) was removed by vacuum distillation at 40 0C. Sodium carbonate (15.60 g, 0.147 mol) was added and the mixture was stirred at room temperature for 30 min. The mixture was filtered and the solid was washed with water (3 x 30 mL) and dried. The solid was dissolved in pre-mixed methylene chloride (CH2Cl2, 1000 mL) and methanol (MeOH, 300 mL). With vigorous stirring, a solution of 0.236 M of sodium carbonate (Na2CO3) in water (1000 mL) was added dropwise. Solid was slowly precipitated out after addition of aqueous solution of sodium carbonate (Na2CO3). Hexane (1000 niL) was then added dropwise with stirring. The mixture was stirred at room temperature for 30 – 40 min and the solids were collected by filtration. The solids collected were washed with water (3 x 200 mL) and dried in vacuum at 40 – 500C to constant weight to afford 2-fluoro-Λr-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[l,2-ό][l,2,4]triazin-2-yl)benzamide (15, 42.2 g, 43.67 g theoretical, 96.6% yield) as a bright yellow solid, which was found to be identical to the material made by Method A in every comparable aspect. For 15: 1H NMR (400 MHz, DMSO-J6) δ ppm 9.20 (s, IH), 8.82 (dd, IH, J= 4.05, 1.56 Hz), 8.38 (br m, IH), 8.27 (dd, IH, J= 8.50 Hz, 1.25 Hz), 8.06 – 7.93 (m, 5H), 7.81-7.74 (m, 2H), 7.49 (dd, IH, J= 8.40 Hz, 4.35 Hz), 4.62 (s, 2H), 2.78 (d, 3H, J= 4.36 Hz); C23H17FN6O (MW 412.42), LCMS (EI) mle 413.1 (M+ + H).

Example 29

2-Fluoro-iV-methyl-4-(7-(quinolin-6-ylmethyl)imidazo [ 1 ,2-b] [ 1 ,2,4] triazin-2-yl)benzamide dihydrochloride (21, dihydrochloride)

2-Fluoro-vV-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[l, 2-Z?][l, 2,4]triazin-2-yl)benzamide (15, 210O g, containing ~25% residual solvents) and filtered USP water (7.6 L) were charged into a 50 L reactor at room temperature. With stirring a solution of 6 M aqueous hydrochloric acid (HCl, 3 L) was added with an additional funnel. The resulting reaction mixture was stirred at room temperature for 1.5 h. Acetone (30.5 L) was added to the reactor with stirring during 1 h and the resulting mixture was stirred at room temperature for 2.5 h. The solids were collected by filtration, washed with acetone (2 x 4.3 L) and dried in vacuum to constant weight to afford 2-fluoro-iV-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[l ,2-b][ 1 ,2,4]triazin-2-yl)benzamide dihydrochloride (21, dihydrochloride, 1629.2 g, 1830.6 g theoretical, 89%) as a pale yellowish crystalline powder, which was found to be identical to the material made by previous method in every comparable aspect. For 21 (dihydrochloride): 1H NMR (400 MHz, DMSO-J6) δ ppm 9.46 (s, IH), 9.25 (dd, IH, J= 5.4 Hz, 1.4 Hz), 9.12 (d, IH, J= 8.3 Hz), 8.51 (m, IH), 8.47 (d, IH, J= 0.9 Hz), 8.34 (d, IH, J= 1.3 Hz), 8.23 (s, IH), 8.21 (dd, IH, J= 9.0, 1.8 Hz), 8.09 – 8.02 (m, 3H), 7.79 (dd, IH, J= 7.5, 8.3 Hz), 4.77 (s, 2H), 2.78 (s, 3H, J= 4.5 Hz); 13C NMR (100 MHz, DMSO-J6) δ ppm 163.4, 159.4 (d, J= 249.9 Hz), 145.8, 145.4, 144.5, 143.8, 140.4, 138.8, 136.8, 135.9, 135.7 (J= 8.6 Hz), 131.2 ( J= 3.1 Hz), 130.7, 128.7, 128.2, 126.2 (J= 14.9 Hz), 126.0, 123.1 (J= 3 Hz), 122.5, 121.0, 114.9 (J= 5.6 Hz), 28.4, 26.3; 19F NMR (376.3 MHz, DMSO-Z6) δ ppm -113.2; C23H17FN6O (free base, MW 412.42), LCMS (EI) mle 413.1 (M+ + H) and 435.0 (M+ + Na).\

PATENT

(S)-N-(54(R)-2-(2,5-difluoropheny1)-pyrrolidin-1-y1)-pyrazolo[1,5-a]pyrimidin-3-y1)-3-hydroxypyrrolidine-1-carboxamide.

Patent ID Patent Title Submitted Date Granted Date
US9605070 ANTIBODY MOLECULES TO TIM-3 AND USES THEREOF
2015-01-30
2015-08-06
US9683048 ANTIBODY MOLECULES TO PD-1 AND USES THEREOF
2015-01-23
2015-07-30
US8461330 IMIDAZOTRIAZINES AND IMIDAZOPYRIMIDINES AS KINASE INHIBITORS
2011-06-09
US8420645 SALTS OF 2-FLUORO-N-METHYL-4-[7-(QUINOLIN-6-YL-METHYL)-IMIDAZO[1, 2-b][1, 2, 4]TRIAZIN-2-YL]BENZAMIDE AND PROCESSES RELATED TO PREPARING THE SAME
2009-11-26
US7767675 IMIDAZOTRIAZINES AND IMIDAZOPYRIMIDINES AS KINASE INHIBITORS
2008-07-10
2010-08-03
Patent ID Patent Title Submitted Date Granted Date
US2017190777 ANTIBODY MOLECULES TO TIM-3 AND USES THEREOF
2017-03-17
US2016326178 IMIDAZOTRIAZINES AND IMIDAZOPYRIMIDINES AS KINASE INHIBITORS
2016-02-05
US2016046672 STAPLING eIF4E INTERACTING PEPTIDES
2014-02-28
2016-02-18
US2013324515 IMIDAZOTRIAZINES AND IMIDAZOPYRIMIDINES AS KINASE INHIBITORS
2013-05-16
2013-12-05
US2015148348 SALTS OF 2-FLUORO-N-METHYL-4-[7-(QUINOLIN-6-YL-METHYL)- IMIDAZO[1, 2-B][1, 2, 4]TRIAZIN-2-YL]BENZAMIDE AND PROCESSES RELATED TO PREPARING THE SAME
2014-10-28
2015-05-28
Patent ID Patent Title Submitted Date Granted Date
US2017198041 ANTIBODY MOLECULES TO TIM-3 AND USES THEREOF
2017-02-14
US2017258800 SALTS OF 2-FLUORO-N-METHYL-4-[7-(QUINOLIN-6-YL-METHYL)- IMIDAZO[1, 2-B][1, 2, 4]TRIAZIN-2-YL]BENZAMIDE AND PROCESSES RELATED TO PREPARING THE SAME
2016-11-18
US2016108123 ANTIBODY MOLECULES TO PD-L1 AND USES THEREOF
2015-10-13
2016-04-21
US2017209574 COMBINATION THERAPIES
2015-10-02
US2015259420 ANTIBODY MOLECULES TO LAG-3 AND USES THEREOF
2015-03-13
2015-09-17

/////////////////Capmatinib, INC 280, INC-280, капматиниб كابماتينيب 卡马替尼 , INCB 28060

CNC(=O)C1=CC=C(C=C1F)C1=NN2C(CC3=CC=C4N=CC=CC4=C3)=CN=C2N=C1

Alpelisib, BYL 719

$
0
0

Alpelisib.pngChemSpider 2D Image | Alpelisib | C19H22F3N5O2S

Alpelisib

(2S)-1-N-[4-methyl-5-[2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl]-1,3-thiazol-2-yl]pyrrolidine-1,2-dicarboxamide
PDT PAT WO 2010/029082
Chemical Names: Alpelisib; CAS 1217486-61-7; BYL-719; BYL719; UNII-08W5N2C97Q; BYL 719
Molecular Formula: C19H22F3N5O2S
Molecular Weight: 441.473 g/mol
  1. alpelisib
  2. 1217486-61-7
  3. BYL-719
  4. BYL719
  5. UNII-08W5N2C97Q
  6. BYL 719
  7. Alpelisib (BYL719)
  8. (S)-N1-(4-Methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide
  9. NVP-BYL719

Alpelisib is an orally bioavailable phosphatidylinositol 3-kinase (PI3K) inhibitor with potential antineoplastic activity. Alpelisib specifically inhibits PI3K in the PI3K/AKT kinase (or protein kinase B) signaling pathway, thereby inhibiting the activation of the PI3K signaling pathway. This may result in inhibition of tumor cell growth and survival in susceptible tumor cell populations. Activation of the PI3K signaling pathway is frequently associated with tumorigenesis. Dysregulated PI3K signaling may contribute to tumor resistance to a variety of antineoplastic agents.

Alpelisib has been used in trials studying the treatment and basic science of Neoplasms, Solid Tumors, BREAST CANCER, 3rd Line GIST, and Rectal Cancer, among others.
str1 str2
Image result for Alpelisib PHARMACODIA
 SYN 2Image result for Alpelisib PHARMACODIA
POLYMORPHS

(S)-pyrrolidine-l,2-dicarboxylic acid 2-amide l-(4-methyl-5-[2-(2,2,2-trifluoro-l,l- dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl)-amidei hereafter referred to as compound I,

Figure imgf000002_0001

is an alpha-selective phosphatidylinositol 3 -kinase (PI3K) inhibitor. Compound I was originally described in WO 2010/029082, wherein the synthesis of its free base form was described. There is a need for additional solid forms of compound I, for use in drug substance and drug product development. It has been found that new solid forms of compound I can be prepared as one or more polymorph forms, including solvate forms. These polymorph forms exhibit new physical properties that may be exploited in order to obtain new pharmacological properties, and that may be utilized in drug substance and drug product development. Summary of the Invention

In one aspect, provided herein is a crystalline form of the compound of formula I, or a solvate of the crystalline form of the compound of formula I, or a salt of the crystalline form of the compound of formula I, or a solvate of a salt of the crystalline form of the compound of formula I. In one embodiment, the crystalline form of the compound of formula I has the polymorph form SA, SB, Sc, or SD.

In another aspect, provided herein is a pharmaceutical composition comprising a crystalline compound of formula I. In one embodiment of the pharmaceutical composition, the crystalline compound of formula I has the polymorph form SA, SB,Sc, or So.

In another aspect, provided herein is a method for the treatment of disorders mediated by PI3K, comprising administering to a patient in need of such treatment an effective amount of a crystalline compound of formula I, particularly SA, SB, SC,or SD .

In yet another aspect, provided herein is the use of a crystalline compound of formula I, particularly SA, SB, SC, or SD, for the preparation of a medicament for the treatment of disorders mediated by PI3K.

In still another aspect, provided herein is a method for the treatment of disorders selected from benign or malignant tumor; a cancer selected from sarcoma; lung; bronchus; prostate; breast (including sporadic breast cancers and sufferers of Cowden disease);

pancreas; gastrointestinal cancer; colon; rectum; colon carcinoma; colorectal adenoma;

thyroid; liver; intrahepatic bile duct; hepatocellular; adrenal gland; stomach; gastric; glioma; glioblastoma; endometrial; melanoma; kidney; renal pelvis; urinary bladder; uterine corpus; uterine cervix; vagina; ovary; multiple myeloma; esophagus; a leukaemia; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; brain; a carcinoma of the brain; oral cavity and pharynx; larynx; small intestine; non-Hodgkin lymphoma; melanoma; villous colon adenoma; a neoplasia; a neoplasia of epithelial character; lymphomas; a mammary carcinoma; basal cell carcinoma; squamous cell carcinoma; actinic keratosis; tumor diseases, including solid tumors; a tumor of the neck or head; polycythemia vera; essential thrombocythemia; myelofibrosis with myeloid metaplasia; and Walden stroem disease; as well as polycythemia vera, essential thrombocythemia, myelofibrosis with myeloid metaplasia, asthma, COPD, ARDS, Loffler’s syndrome, eosinophilic pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma, eosinophil-related disorders affecting the airways occasioned by drug-reaction, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforme, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphisus, epidermolysis bullosa acquisita, autoimmune haematogical disorders (e.g., haemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia), systemic lupus erythematosus, polychondritis, scleroderma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven-Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (e.g., ulcerative colitis and Crohn’s disease), endocrine opthalmopathy, Grave’s disease, sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, primary biliary cirrhosis, uveitis (anterior and posterior), interstitial lung fibrosis, psoriatic arthritis, glomerulonephritis, cardiovascular diseases, atherosclerosis, hypertension, deep venous thrombosis, stroke, myocardial infarction, unstable angina, thromboembolism, pulmonary embolism, thrombolytic diseases, acute arterial ischemia, peripheral thrombotic occlusions, and coronary artery disease, reperfusion injuries, retinopathy, such as diabetic retinopathy or hyperbaric oxygen-induced retinopathy, and conditions characterized by elevated intraocular pressure or secretion of ocular aqueous humor, such as glaucoma, comprising administering to a patient in need of such treatment an effective amount of the crystalline compound of formula I, particularly polymorph forms SA, SB, SC, or SD-

In another aspect, provided herein is the use of the crystalline compound of formula I, particularly polymorph forms SA, SB, SC, or SD for the preparation of a medicament for the treatment of the disorders listed above. Brief Description of the Drawings

Figure I depicts the X-ray powder diffraction pattern of polymorph form A. Figure II depicts the FT-IR spectrum of polymorph form A. Figure III depicts the differential scanning calorimetry thermogram of polymorph form A. Figure IV depicts the X-ray powder diffraction pattern of polymorph form SA- Figure V depicts the X-ray powder diffraction pattern of polymorph form SB. Figure VI depicts the X-ray powder diffraction pattern of polymorph form Sc. Figure VII depicts the X-ray powder diffraction pattern of polymorph form SD.

Scheme 2. Synthesis of (S)-Pyrrolidine-1.2-dicarboxylic acid 2-amide l-((4-methyl-5-r2- (2,2,2-trifluoro- 1 , 1 -dimethyl-ethyl -pyridin-4-yl1-thiazol-2-yl} -amide)

Figure imgf000028_0001

Example 2: (S)-Pyrrolidine-1.2-dicarboxylic acid 2-amide 1 -((4-methyl-5- 2 -(2,2,2- trifluoro-1 J-dirhethyl-ethylVpyridin-4-yl -thia2ol-2-yll-amide

The title compound is prepared in analogy to the procedure described in Example 1 but with the following modifications. In Step 2.1 (corresponding to Step 1.1 of Example 1), the reaction mixture is stirred for 14 h at reflux. In Step 2.2 (corresponding to Step 1.2 of Example 1), the reaction mixture is stirred for 1 h at 85 °C and extracted with ethyl acetate after being quenched. In step 2.3 (corresponding to Step 1.3 of Example 1), the reaction mixture is stirred for 2.5 h at 120 °C. In Step 2.4 (corresponding to Step 1.4 of Example 1), the reaction mixture is stirred for 1 h at 83 °C and extracted with ethyl acetate after being quenched. In Step 2.5 (corresponding to Step 1.5 of Example 1), the reaction mixture is stirred for 1 h at 65 °C and trituration in methanol is not performed. In Step 2.6

(corresponding to Step 1.6 of Example 1), the crude product is not purified. In Step 2.7 (corresponding to Step 1.7 of Example 1), 3,3,3-trifluoro-2,2-dimethyl-propionyl chloride is used.

Title compound: ESI-MS: 442.0 [M+H]+; tR= 3.02 min (System 1); TLC: Rf = 0.35 (DCM/MeOH, 9: 1).

Example 3: Preparation of Polymorph Form A

(S)-Pyrrolidine-l,2-dicarboxylic acid 2-amide l-({4-methyl-5-[2-(2,2,2-trifluoro-l,l- dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl}-amide) (10.0 g) was suspended in ethanol/water (85:15 v/v; 75 mL) and the mixture was heated to 75 °C. The solution was clear-filtered into a second flask and the first flask was then washed with ethanol/water (4:6 v/v; 20 mL), followed by water (10 mL). The clear solution was stirred at 75 °C for an additional 30 minutes. The clear solution was then cooled to 2 °C over 2 hours and the obtained thick suspension was stirred at 2 °C for an additional hour. The mixture was then filtered, and the flask and filter cake were washed with ethanol/water (1 :1 v/v; 20 mL), followed by ethyl acetate (10 mL). The wet filter cake was returned to the flask and suspended in ethyl acetate (75 mL). the mixture was heated to 78 °C and was stirred under reflux for 1 hour. During this time, 15 mL ethyl acetate was distilled off. The mixture was then cooled to 2 °C over 2 hours and the suspension was stirred at 2 °C for an additional hour. The mixture was filtered, and the flask and filter cake were washed with cold ethyl acetate (12 mL). The filter cake was then dried under 1-50 mbar vacuum at 50 °C to yield the polymorph form A (7.3 g).

Publication numberPriority datePublication dateAssigneeTitle
WO2010029082A12008-09-102010-03-18Novartis AgOrganic compounds
WO2012016970A1 *2010-08-022012-02-09Novartis AgA crystalline form of (s)-pyrrolidine-1,2-dicarboxylic acid 2-amide 1-(4 -methyl-5-[2-(2,2,2-trifluoro-1,1-dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl)-amide and its use as pi3k inhibitor
KR20070113188A *2004-10-072007-11-28베링거 인겔하임 인터내셔날 게엠베하Thiazolyldihydroindazoles
EP2016075A1 *2006-05-032009-01-21AstraZeneca ABThiazole derivatives and their use as anti-tumour agents
WO2016051374A1 *2014-10-032016-04-07Novartis AgPharmaceutical compositions comprising alpelisib
CN105979947A *2013-12-062016-09-28诺华股份有限公司Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
 PATENTS
Patent ID Patent Title Submitted Date Granted Date
US9815898 ANTIBODY MOLECULES TO PD-1 AND USES THEREOF
2017-05-15
US2017210733 BENZOXAZEPIN OXAZOLIDINONE COMPOUNDS AND METHODS OF USE
2017-04-07
US2017210804 ANTIBODY MOLECULES TO LAG-3 AND USES THEREOF
2017-03-24
US2017190777 ANTIBODY MOLECULES TO TIM-3 AND USES THEREOF
2017-03-17
US2017166550 BENZOTHIOPHENE-BASED SELECTIVE ESTROGEN RECEPTOR DOWNREGULATORS
2016-12-09
Patent ID Patent Title Submitted Date Granted Date
US2015291606 MERTK-SPECIFIC PYRROLOPYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
US2015291609 MERTK-SPECIFIC PYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
US9603850 MERTK-SPECIFIC PYRAZOLOPYRIMIDINE COMPOUNDS
2015-04-03
2015-10-15
US2015259420 ANTIBODY MOLECULES TO LAG-3 AND USES THEREOF
2015-03-13
2015-09-17
US9605070 ANTIBODY MOLECULES TO TIM-3 AND USES THEREOF
2015-01-30
2015-08-06
Patent ID Patent Title Submitted Date Granted Date
US2016108123 ANTIBODY MOLECULES TO PD-L1 AND USES THEREOF
2015-10-13
2016-04-21
US2017209574 COMBINATION THERAPIES
2015-10-02
US2017224836 ANTI-CDH6 ANTIBODY DRUG CONJUGATES
2015-08-07
US2017189409 MEDICAL USE
2015-05-21
US2015320880 ANTIBODY DRUG CONJUGATES
2015-05-20
2015-11-12

/////////////////Alpelisib,  CAS,  1217486-61-7, BYL-719, BYL719, UNII-08W5N2C97Q, BYL 719

CC1=C(SC(=N1)NC(=O)N2CCCC2C(=O)N)C3=CC(=NC=C3)C(C)(C)C(F)(F)F

ABL 001, Asciminib

$
0
0

img

Image result for ABL001 / Asciminib

ABL001 / Asciminib

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

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

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

PHASE 3, Chronic Myeloid Leukemia, NOVARTIS

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

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

Highest Development Phases

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

Most Recent Events

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

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

PATENT

WO2013171639

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

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

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

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

Example 9

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

PAPER

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

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

PAPER

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

PATENT

US 20170216289

PAPER

  • By El Rashedy, Ahmed A.; Olotu, Fisayo A.; Soliman, Mahmoud E. S.
  • From Chemistry & Biodiversity (2018), 15(3), n/a.
Patent ID Patent Title Submitted Date Granted Date
US2016108123 ANTIBODY MOLECULES TO PD-L1 AND USES THEREOF
2015-10-13
2016-04-21
US2014343086 COMPOUNDS AND COMPOSITIONS FOR INHIBITING THE ACTIVITY OF ABL1, ABL2 AND BCR-ABL1
2014-07-31
2014-11-20
US8829195 Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
2013-05-13
2014-09-09

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

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

BMS 986205, ONO 7701

$
0
0

ChemSpider 2D Image | BMS 986205 | C24H24ClFN2Oimg

BMS 986205

(2R)-N-(4-Chlorophenyl)-2-[cis-4-(6-fluoro-4-quinolinyl)cyclohexyl]propanamide
Cyclohexaneacetamide, N-(4-chlorophenyl)-4-(6-fluoro-4-quinolinyl)-α-methyl-, cis-
Cyclohexaneacetamide, N-(4-chlorophenyl)-4-(6-fluoro-4-quinolinyl)-α-methyl-, cis-(αR)-
(i?)-N-(4-chlorophenyl)-2- c 5-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide

CAS: 1923833-60-6

Phase III Head and neck cancer; Malignant melanoma

BMS-986205, ONO-7701,  F- 001287

  • Molecular Formula C24H24ClFN2O
  • Average mass 410.912 Da

BMS986205, BMS 986205, ONO-7701

Cyclohexaneacetamide, N-(4-chlorophenyl)-4-(6-fluoro-4-quinolinyl)-α-methyl-, cis-(αR)-

A potent and selective IDO1 (indoleamine 2,3-dioxygenase 1) inhibitor.

Alternate Name (R)-N-(4-chlorophenyl)-2-((1s,4S)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propenamide
Appearance Crystalline solid
CAS # 1923833-60-6
Molecular Formula C₂₄H₂₄ClFN₂O
Molecular Weight 410.92
  • Originator Bristol-Myers Squibb
  • Developer Bristol-Myers Squibb; Ono Pharmaceutical
  • Class Antineoplastics; Cyclohexanes; Quinolines; Small molecules
  • Mechanism of Action Indoleamine-pyrrole 2,3-dioxygenase inhibitors

Highest Development Phases

  • Phase II IHead and neck cancer; Malignant melanoma
  • Phase I/II Cancer
  • Phase I Solid tumours

Most Recent Events

  • 01 Jun 2018Efficacy and adverse events data from a phase I/IIa trial in Bladder cancer (Combination therapy, Late-stage disease) presented at the 54th Annual Meeting of the American Society of Clinical Oncology (ASCO- 2018)
  • 08 May 2018Bristol-Myers Squibb plans the CheckMate 9UT phase II trial for Bladder Cancer in USA, Canada, Italy, Mexico, Netherlands, Spain and United Kingdom , (NCT03519256)
  • 30 Apr 2018Bristol-Myers Squibb withdraws a phase III trial for Non-small cell lung cancer (First-line therapy, Combination therapy, Late-stage disease) in USA, Austria, Australia, Brazil, Canada, Czech Republic, France, Germany, Greece, Italy, Japan, South Korea, Mexico, Spain, Switzerland, Taiwan and Turkey prior to enrolment (NCT03417037)

WO 2016073770

Inventors Hilary Plake BeckJuan Carlos JaenMaksim OSIPOVJay Patrick POWERSMaureen Kay REILLYHunter Paul SHUNATONAJames Ross WALKERMikhail ZIBINSKYJames Aaron BalogDavid K WilliamsJay A MARKWALDEREmily Charlotte CHERNEYWeifang ShanAudris Huang
Applicant Flexus Biosciences, Inc.

Image result for BMS 986205

Image result for BMS 986205
Bristol-Myers Squibb
, following its acquisition of Flexus Biosciences, is developing BMS-986205 (previously F- 001287), the lead from an immunotherapy program of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors for the potential treatment of cancer. In February 2016, a phase I/IIa trial was initiated .

BMS-986205 (ONO-7701) is being evaluated at Bristol-Myers Squibb in phase I/II clinical trials for the oral treatment of adult patients with advanced cancers in combination with nivolumab. Early clinical development is also ongoing at Ono in Japan for the treatment of hematologic cancer and for the treatment of solid tumors.

In April 2017, data from the trial were presented at the 108th AACR Annual Meeting in Washington DC. As of February 2017, the MTD had not been reached, but BMS-986205 plus nivolumab treatment was well tolerated, with only two patients discontinuing treatment due to DLTs. The most commonly reported treatment-related adverse events (TRAEs) were decreased appetite, fatigue, nausea, diarrhea, and vomiting. Grade 3 TRAEs were reported in three patients during the combination therapy; however, no grade 3 events were reported during BMS-986205 monotherapy lead-in. No grade 4 or 5 TRAEs were reported with BMS-986205 alone or in combination with nivolumab

Indoleamine 2,3-dioxygenase (IDO; also known as IDOl) is an IFN-γ target gene that plays a role in immunomodulation. IDO is an oxidoreductase and one of two enzymes that catalyze the first and rate-limiting step in the conversion of tryptophan to N-formyl-kynurenine. It exists as a 41kD monomer that is found in several cell populations, including immune cells, endothelial cells, and fibroblasts. IDO is relatively well-conserved between species, with mouse and human sharing 63% sequence identity at the amino acid level. Data derived from its crystal structure and site-directed mutagenesis show that both substrate binding and the relationship between the substrate and iron-bound dioxygenase are necessary for activity. A homolog to IDO (ID02) has been identified that shares 44% amino acid sequence homology with IDO, but its function is largely distinct from that of IDO. (See, e.g., Serafini P, et al, Semin. Cancer Biol, 16(l):53-65 (Feb. 2006) and Ball, H.J. et al, Gene, 396(1):203-213 (Jul. 2007)).

IDO plays a major role in immune regulation, and its immunosuppressive function manifests in several manners. Importantly, IDO regulates immunity at the T cell level, and a nexus exists between IDO and cytokine production. In addition, tumors frequently manipulate immune function by upregulation of IDO. Thus, modulation of IDO can have a therapeutic impact on a number of diseases, disorders and conditions.

A pathophysiological link exists between IDO and cancer. Disruption of immune homeostasis is intimately involved with tumor growth and progression, and the production of IDO in the tumor microenvironment appears to aid in tumor growth and metastasis. Moreover, increased levels of IDO activity are associated with a variety of different tumors (Brandacher, G. et al, Clin. Cancer Res., 12(4): 1144-1151 (Feb. 15, 2006)).

Treatment of cancer commonly entails surgical resection followed by chemotherapy and radiotherapy. The standard treatment regimens show highly variable degrees of long-term success because of the ability of tumor cells to essentially escape by regenerating primary tumor growth and, often more importantly, seeding distant metastasis. Recent advances in the treatment of cancer and cancer-related diseases, disorders and conditions comprise the use of combination therapy incorporating immunotherapy with more traditional chemotherapy and radiotherapy. Under most scenarios, immunotherapy is associated with less toxicity than traditional chemotherapy because it utilizes the patient’s own immune system to identify and eliminate tumor cells.

In addition to cancer, IDO has been implicated in, among other conditions, immunosuppression, chronic infections, and autoimmune diseases or disorders (e.g. , rheumatoid arthritis). Thus, suppression of tryptophan degradation by inhibition of IDO activity has tremendous therapeutic value. Moreover, inhibitors of IDO can be used to enhance T cell activation when the T cells are suppressed by pregnancy, malignancy, or a virus (e.g., HIV). Although their roles are not as well defined, IDO inhibitors may also find use in the treatment of patients with neurological or neuropsychiatric diseases or disorders (e.g., depression).

Small molecule inhibitors of IDO have been developed to treat or prevent IDO-related diseases. For example, the IDO inhibitors 1-methyl-DL-tryptophan; p-(3-benzofuranyl)-DL-alanine; p-[3-benzo(b)thienyl]-DL-alanine; and 6-nitro-L-tryptophan have been used to modulate T cell-mediated immunity by altering local extracellular concentrations of tryptophan and tryptophan metabolites (WO 99/29310). Compounds having IDO inhibitory activity are further reported in WO 2004/094409.

In view of the role played by indoleamine 2,3-dioxygenase in a diverse array of diseases, disorders and conditions, and the limitations (e.g., efficacy) of current IDO inhibitors, new IDO modulators, and compositions and methods associated therewith, are needed.

In April 2017, preclinical data were presented at the 108th AACR Annual Meeting in Washington DC. BMS-986205 inhibited kynurenine production with IC50 values of 1.7, 1.1 and > 2000 and 4.6, 6.3 and > 2000 nM in human (HeLa, HEK293 expressing human IDO-1 and tryptophan-2, 3-dioxygenase cell-based assays) and rat (M109, HEK293 expressing mouse ID0-1 and -2 cell-based assays) respectively. In human SKOV-3 xenografts (serum and tumor) AUC (0 to 24h; pharmacokinetic and pharmacodynamic [PK and PD])) was 0.8, 4.2 and 23 and 3.5, 11 and 40 microM h, respectively; area under the effect curve (PK and PD) was 39, 32 and 41 and 60, 63 and 76% kyn, at BMS-986205 (5, 25 and 125 mg/kg, qd×5), respectively

In April 2017, preclinical data were presented at the 253rd ACS National Meeting and Exhibition in San Francisco, CA. BMS-986205 showed potent and selective inhibition of IDO-1 enzyme (IC50 = 1.7nM) and potent growth inhibition in cellular assays (IC50 = 3.4 nM) in SKOV3 cells. A good pharmacokinetic profile was seen at oral and iv doses in rats, dogs and monkeys. The compound showed good oral exposure and efficacy in in vivo assays

Preclinical studies were performed to evaluate the activity of BMS-986205, a potent and selective optimized indoleamine 2, 3-dioxygenase (IDO)- 1inhibitor, for the treatment of cancer. BMS-986205 inhibited kynurenine production with IC50 values of 1.7, 1.1 and > 2000 and 4.6, 6.3 and > 2000 nM in human (HeLa, HEK293 expressing human IDO-1 and tryptophan-2, 3-dioxygenase cell-based assays) and rat (M109, HEK293 expressing mouse ID0-1 and -2 cell-based assays) respectively. BMS-986205 was also found to be potent when compared with IDO-1from other species (human < dog equivalent monkey equivalent mouse > rat). In cell-free systems, incubation of inhibitor lead to loss of heme absorbance of IDO-1 which was observed in the presence of BMS-986205 (10 microM), while did not observed with epacadostat (10 microM). The check inhibitory activity and check reversibility (24 h after compound removal) of BMS-986205 was found to be < 1 and 18% in M109 (mouse) and < 1 and 12% SKOV3 (human) cells, respectively. In human whole blood IDO-1, human DC mixed lymphocyte reaction and human T cells cocultured with SKOV3 cells- cell based assays, BMS-986205 showed potent cellular effects (inhibition of kynurenine and T-cell proliferation 3H-thymidine) with IC50 values of 2 to 42 (median 9.4 months), 1 to 7 and 15 nM, respectively. In human SKOV-3 xenografts (serum and tumor) AUC (0 to 24h; pharmacokinetic and pharmacodynamic [PK and PD])) was 0.8, 4.2 and 23 and 3.5, 11 and 40 microM h, respectively; area under the effect curve (PK and PD) was 39, 32 and 41 and 60, 63 and 76% kyn, at BMS-986205 (5, 25 and 125 mg/kg, qd×5), respectively. In vivo human-SKOV3 and hWB-xenografts, IC50 values of BMS-986205 were 3.4 and 9.4 NM, respectively. The ADME of BMS-986205 at parameters iv/po dose was 0.5/2, 0.5/1.5 and 0.5/1.2 mg/kg, respectively; iv/clearance was 27, 25 and 19 ml, min/kg, respectively; iv Vss was 3.8, 5.7 and 4.1 l/kg, respectively; t1/2 (iv) was 3.9, 4.7 and 6.6 h, respectively; fraction (po) was 64, 39 and 10%, respectively. At the time of presentation, BMS-986205 was being evaluated in combination with nivolumab.

The chemical structure and preclinical profile was presented for BMS-986205 ((2R)-N-(4-Chlorophenyl)-2-[cis-4-(6-fluoroquinolin-4-yl)cyclohexyl]propanamide), a potent IDO-1 inhibitor in phase I for the treatment of cancer. This compound showed potent and selective inhibition of IDO-1 enzyme (IC50 = 1.7nM) and potent growth inhibition in cellular assays (IC50 = 3.4 nM) in SKOV3 cells. The pharmacokinetic profile in rats dosed at 0.5 mg/kg iv and 2 mg/kg po, with clearance, Vss, half-life and bioavailability of 27 ml/min/kg, 3.8 l/kg, 3.9 h and 4%, respectively; in dogs at 0.5 iv and 1.5 po mg/kg dosing results were 25 ml/min/kg, 5.7 l/kg, 4.7 h and 39%; and, in cynomolgus monkeys with the same doses as dogs results were 19 ml/min/kg, 4.1 l/kg, 6.6 h and 10%, respectively. The compound showed good oral exposure and efficacy in in vivo assays.

BMS-986158: a BET inhibitor for cancerAshvinikumar Gavai of Bristol Myers Squibb (BMS) gave an overview of his company’s research into Bromodomian and extra-terminal domain (BET) as oncology target for transcriptional suppression of key oncogenes, such as MYC and BCL2. BET inhibition has been defined as strong rational strategy for the treatment of hematologic malignancies and solid tumors. From crystal-structure guided SAR studies, BMS-986158, 2-{3-(1,4-Dimethyl-1H-1,2,3-triazol-5-yl)-5-[(S)-(oxan-4-yl)(phenyl)methyl]-5H-pyrido[3,2-b]indol-7-yl}propan-2-ol, was chosen as a potent BET inhibitor, showing IC50 values for BRD2, BRD3 and BRD4 activity of 1 nM; it also inhibited Myc oncogene (IC50 = 0.5 nM) and induced chlorogenic cancer cell death. In vitro the compound also displayed significant cytotoxicity against cancer cells.  When administered at 0.25, 0.5 and 1 mg/kg po, qd to mice bearing human lung H187 SCLC cancer xenograft, BMS-986158 was robust and showed efficacy as a anticancer agent at low doses. In metabolic studies, it showed t1/2 of 36, 40 and 24 min in human, rat and mice, respectively, and it gave an efflux ratio of 3 in Caco-2 permeability assay. In phase 1/II studies, BMS-986158 was well tolerated at efficacious doses and regimens, and drug tolerable toxicity at efficacy doses and regimens. Selective Itk inhibitors for inflammatory disordersThe development of highly selective Itk inhibitors for the treatment of diseases related to T-cell function, such as inflammatory disorders, was described by Shigeyuki Takai (Ono Pharmaceutical). Inhibitory properties of a hit compound, ONO-8810443, were modified via X-ray structure and Molecular Dynamics stimulation to get ONO-212049 with significant kinase selectivity (140-fold) against Lck, a tyrosine kinase operating upstream of Itk in the TCR cascade. Further modifications identified final lead compound ONO-7790500 (N-[6-[3-amino-6-[2-(3-methoxyazetidin-1-yl)pyridin-4-yl]pyrazin-2-yl]pyridin-3-yl]-1-(3-methoxyphenyl)-2,3-dimethyl-5-oxopyrazole-4-carboxamide), which selectively inhibited Itk (IC50 = < 0.004 microM) over Lck (IC50 = 9.1 microM; SI 2000-fold) and suppressed Jurkat T-cell proliferation (IC50 = 0.014 microM). This compound suppressed alphaCD3/CDP28 CD4+T-cell stimulation (IC50 = 0.074 microM) with selectivity over PMA/Ionomycin (IC50 = > 10 microM). ONO-7790500 also exhibited in vivo IL-2 inhibitory properties (62% inhibition at 30 mg/kg po) in mice. In pharmacokinetic studies in balb/c mice, the compound administered orally (10 mg/kg) showed a Cmax of 1420 ng/ml, AUClast of 11,700 ng*h/ml, t1/2 of 5.3 h and oral bioavailability of 68%. Administration iv at 0.3 mg/kg gave an AUC last of 610 ng*h/ml, t1/2 of 3.8 h, Vss of 1260 ml/kg and Cl of 5.1 ml/min/kg. ADMET data showed ONO-7790500 did not have relevant activity in cytochromes and hERG channels (IC50 > 10 microM) in toxicological studies, and gave a PAMPA value of 5.0 x 10(-6) cm/s. Fused imidazole and pyrazole derivatives as TGF-beta inhibitorsDual growth and differentiation factor-8 (GDF-8; also known as myostatin) and TGF-beta inhibitors were described. Both targets belong to TGF-beta superfamily consisting of a large group of structurally related cell regulatory proteins involved in fundamental biological and pathological processes, such as cell proliferation or immunomodulation. Myostatin (GDF8) is a negative regulator negative regulator of skeletal muscle growth and has also been related to bone metabolism. Investigators at Rigel Pharmaceuticals found that compounds designed to be GDF-8 inhibitors were able to inhibit TGF-beta as well, this could be an advantage for the treatment of diseases associated with muscle and adipose tissue disorders, as well as potentially immunosuppressive disorders. Jiaxin Yu from the company described  new fused imidazole derivatives, of which the best compound was 6-[2-(2,4,5-Trifluorophenyl)-6,7-dihydro-5H-pyrrolo[1,2-a]imidazol-3-yl]quinoxaline. This compound was very potent at TGF-beta Receptor Type-1 (ALK5) inhibition with an IC50 value of 1nM. In an in vivo mouse assay this compound showed good activity at 59.7 mg/kg, po, and good plasma exposure; inhibition of GDF-8 and TGFbeta growth factors was 90 and 81.6 %, respectively.Rigel’s Ihab Darwish described a series of fused pyrazole derivatives, with the best compound being 6-[2-(2,4-Difluorophenyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl][1,2,4]triazolo[1,5-a]pyridine. This compound showed an IC50 of 0.06 and 0.23 microM for GDF-8 and TGFbeta, respectively, in the pSMAD (MPC-11) signaling inhibition test. The compound had a good pharmacokinetic profile, with 40% of bioavailability in mice after a 5-mg/kg po dose. An iv dose of 1 mg/kg showed t1/2 of 0.7 h and Vss of 1.0 l/h/kgDiscovery of selective inhibitor of IDO BMS-986205 for cancerIndoleamine-2,3-dioxygenase (IDO)-1 enzyme initiates and regulates the first step of the kynurenine pathway (KP) of tryptophan metabolism, and evidence has shown that overexpression of IDO-1 in cancer tumors is a crucial mechanism facilitating tumor immune evasion and persistence. The chemical structure and preclinical profile of BMS-986205 was presented by Aaron Balog from BMS. BMS-986205 ((2R)-N-(4-Chlorophenyl)-2-[cis-4-(6-fluoroquinolin-4-yl)cyclohexyl]propanamide),  is a potent IDO-1 inhibitor in phase I for the treatment of cancer. This compound showed potent and selective inhibition of IDO-1 enzyme (IC50 = 1.7nM) and potent growth inhibition in cellular assays (IC50 = 3.4 nM) in SKOV3 cells. The pharmacokinetic profile in rats dosed at 0.5 mg/kg iv and 2 mg/kg po, with clearance, Vss, half-life and bioavailability of 27 ml/min/kg, 3.8 l/kg, 3.9 h and 4%, respectively; in dogs at 0.5 iv and 1.5 po mg/kg dosing results were 25 ml/min/kg, 5.7 l/kg, 4.7 h and 39%; and, in cynomolgus monkeys with the same doses as dogs results were 19 ml/min/kg, 4.1 l/kg, 6.6 h and 10%, respectively. The compound showed good oral exposure and efficacy in in vivo assays.Three further reports have been published from this meeting .The website for this meeting can be found at https://www.acs.org/content/acs/en/meetings/spring-2017.html.

SYNTHESIS

1 Wittig  NaH

2 REDUCTION H2, Pd, AcOEt, 4 h, rt, 50 psi

3 Hydrolysis HCl, H2O, Me2CO, 2 h, reflux

4  4-Me-2,6-(t-Bu)2-Py, CH2Cl2, overnight, rt

5 SUZUKI AcOK, 72287-26-4, Dioxane, 16 h, 80°C

6  Heck Reaction,  Suzuki Coupling, Hydrogenolysis of Carboxylic Esters, Reduction of Bonds, HYDROGEN

7 Et3N, THF, rt – -78°C , Pivaloyl chloride, 15 min, -78°C; 1 h, 0°C ,THF, 0°C – -78°C, BuLi, Me(CH2)4Me, 15 min, -78°C, R:(Me3Si)2NH •Na, THF, 10 min, -50°C , HYDROLYSIS,  (PrP(=O)O)3, C5H5N, AcOEt, 5 min, rt

Product Patent

WO2016073770

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=289DBE79BEFC6ADC558C89E7A74B19DB.wapp2nB?docId=WO2016073770&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Example 19

(i?)-N-(4-chlorophenyl)-2- c 5-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide

Example 19 : (i?)-N-(4-chlorophenyl)-2-(cz5-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide

[0277] Prepared using General Procedures K, B, E, L, M, N, and O. General Procedure L employed 2-(4-(6-fluoroquinolin-4-yl)-cyclohexyl)acetic acid (mixture of

diastereomers), and ( ?)-2-phenyl-oxazolidinone. General Procedure M employed the cis product and iodomethane. The auxiliary was removed following General Procedure N and the desired product formed employing General Procedure O with 4-chloroaniline.

Purified using silica gel chromatography (0% to 100% ethyl acetate in hexanes) to afford Example 19. 1H NMR of czs-isomer (400 MHz; CDC13): δ 9.14 (s, 1H), 8.70 (d, J= 4.6 Hz, 1H), 8.06 (dd, J= 9.2 Hz, J= 5.6 Hz, 1H), 7.58-7.64 (m, 3H), 7.45 (ddd, J= 9.3 Hz, J= 7.8 Hz, J= 2.7 Hz, 1H), 7.19-7.24 (m, 2H), 7.15 (d, J= 4.6Hz, 1H), 3.16-3.26 (m, 1H), 2.59-2.69 (m, 1H), 2.08-2.16 (m, 1H), 1.66-1.86 (m, 7H), 1.31-1.42 (m, 1H), 1.21 (d, J= 6.8Hz, 3H) ppm. m/z 411.2 (M+H)+.

PAPER

Bioorganic & Medicinal Chemistry Letters (2018), 28(3), 319-329.

https://www.sciencedirect.com/science/article/pii/S0960894X17312180

PATENT

WO 2018022992

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018022992&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

PATENT

WO 2018071500

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018071500&redirectedID=true

REFERENCES

23-Feb-2015
Bristol-Myers Squibb To Expand Its Immuno-Oncology Pipeline with Agreement to Acquire Flexus Biosciences, Inc
Bristol-Myers Squibb Co; Flexus Biosciences Inc

17-Dec-2014
Flexus Biosciences, a Cancer Immunotherapy Company Focused on Agents for the Reversal of Tumor Immunosuppression (ARTIS), Announces $38M Financing
Flexus Biosciences Inc

2015106thApril 21Abs 4290
Potent and selective next generation inhibitors of indoleamine-2,3-dioxygenase (IDO1) for the treatment of cancer
American Association for Cancer Research Annual Meeting
Jay P. Powers, Matthew J. Walters, Rajkumar Noubade, Stephen W. Young, Lisa Marshall, Jan Melom, Adam Park, Nick Shah, Pia Bjork, Jordan S. Fridman, Hilary P. Beck, David Chian, Jenny V. McKinnell, Maksim Osipov, Maureen K. Reilly, Hunter P. Shunatona, James R. Walker, Mikhail Zibinsky, Juan C. Jaen

2017108thApril 04Abs 4964
Structure, in vitro biology and in vivo pharmacodynamic characterization of a novel clinical IDO1 inhibitor
American Association for Cancer Research Annual Meeting
John T Hunt, Aaron Balog, Christine Huang, Tai-An Lin, Tai-An Lin, Derrick Maley, Johnni Gullo-Brown, Jesse Swanson, Jennifer Brown

2017253rdApril 05Abs MEDI 368
Discovery of a selective inhibitor of indoleamine-2,3-dioxygenase for use in the therapy of cancer
American Chemical Society National Meeting and Exposition
Aaron Balog

April 2-62017
American Chemical Society – 253rd National Meeting and Exhibition (Part IV) – OVERNIGHT REPORT, San Francisco, CA, USA
Casellas J, Carceller V

////////////////PHASE 1, BMS 986205, 1923833-60-6, BMS-986205, ONO-7701,Bristol-Myers Squibb,  Antineoplastics,  F- 001287

 C[C@H]([C@H]1CC[C@@H](C2=CC=NC3=CC=C(F)C=C23)CC1)C(NC4=CC=C(Cl)C=C4)=O

Wrapping up ‘s 1st time disclosures is Aaron Balog of @bmsnews talking about an IOD-1 inhibitor to treat cancer 

str0

////////////////BMS986205, BMS 986205, BM-986205, ONO-7701, Phase III,  Head and neck cancer, Malignant melanoma, 1923833-60-6

CC(C1CCC(CC1)C2=C3C=C(C=CC3=NC=C2)F)C(=O)NC4=CC=C(C=C4)Cl

RG7440, Ipatasertib, アイパタセルチブ;

$
0
0

1001264-89-6.png

Ipatasertib.svg

Ipatasertib

GDC-0068 , RG7440

CAS 1001264-89-6, C24H32ClN5O2, 457.9962

アイパタセルチブ;
イパタセルチブ;

Antineoplastic, AKT serine/threonine kinase inhibitor

2(S)-(4-Chlorophenyl)-1-[4-[7(R)-hydroxy-5(R)-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl]piperazin-1-yl]-3-(isopropylamino)propan-1-one

(2S)-2-(4-Chlorophenyl)-1-(4-((5R,7R)-7-hydroxy-5-methyl-6,7-dihydro-5H-cyclopenta(d)pyrimidin-4-yl)piperazin-1-yl(-3-((propan-2-yl)amino)propan-1-one

1-Propanone, 2-(4-chlorophenyl)-1-(4-((5R,7R)-6,7-dihydro-7-hydroxy-5-methyl-5H-cyclopentapyrimidin-4-yl)-1-piperazinyl)-3-((1-methylethyl)amino)-,  (2S)-

2D chemical structure of 1396257-94-5

Ipatasertib dihydrochloride
1396257-94-5

Ipatasertib (RG7440) is an experimental cancer drug in development by Roche. It is a small molecule inhibitor of Akt. It was discovered by Array Biopharma and is currently in phase II trials for treatment of breast cancer.[1]

In vitro, ipatasertib showed activity against all three isoforms of Akt.[2]

Ipatasertib is an orally-available protein kinase B (PKB/Akt) inhibitor in phase III clinical development at Genentech for the treatment of metastatic castration-resistant prostate cancer in combination with abiraterone and prednisone.

In 2014, orphan drug designation was assigned in the U.S. for the treatment of gastric cancer including cancer of the gastro-esophageal junction.

Ipatasertib. An orally bioavailable inhibitor of the serine/threonine protein kinase Akt (protein kinase B) with potential antineoplastic activity. Ipatasertib binds to and inhibits the activity of Akt in a non-ATP-competitive manner, which may result in the inhibition of the PI3K/Akt signaling pathway and tumor cell proliferation and the induction of tumor cell apoptosis. Activation of the PI3K/Akt signaling pathway is frequently associated with tumorigenesis and dysregulated PI3K/Akt signaling may contribute to tumor resistance to a variety of antineoplastic agents. Check for active clinical trials using this agent.

PROBLEM 

It has been found that ipatasertib exhibits a very high solubility (>1 g/g water; >2 g/g water/ethanol 1:1) and a very high hygroscopicity (˜6% at 50% RH, >35% at 95% RH). Whereas poor solubility is often a limiting factor in the development of galenical formulations of other API’s (active pharmaceutical ingredient), a high solubility can equally be problematic for the process performance. Due to this very high intrinsic hygroscopicity of the API, ipatasertib drug substance tends to auto-dissolve to a honey-like viscous liquid at increased humidity. Such high solubility and hygroscopicity may pose serious problems for processing as well as for stability and shelf-life of the final product. Therefore, conventional pharmaceutical compositions comprising ipatasertib and processes for the manufacture of pharmaceutical compositions comprising wetting (e.g. wet granulation) are difficult due to the high solubility and high hygroscopicity of the API.

SYN

 Ipatasertib pk_prod_list.xml_prod_list_card_pr?p_tsearch=A&p_id=691990

Bromination of (+)-(R)-pulegone (I) with Br2 in the presence of NaHCO3 in Et2O, followed by ring contraction via Favorskii rearrangement with NaOEt in EtOH, and treatment with semicarbazide hydrochloride and NaOAc in refluxing EtOH/H2O gives rise to cyclopentanecarboxylate (II) (1). Subsequent ozonolysis of olefin (II) by means of O3 in EtOAc at -78 °C, and reductive treatment with Zn in AcOH provides beta-ketoester (III). Reaction of ketoester (III) with ammonium acetate (IVa) in MeOH/CH2Cl2 yields enamine (V), which upon cyclization with ammonium formate (IVb) and formamide (VI) at 150 °C provides cyclopentapyrimidinol (VII). Chlorination of pyrimidinol (VII) using POCl3 in refluxing CH2Cl2 results in 4-chloro-5(R)-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidine (VIII), which is condensed with N-Boc-piperazine (IX) in the presence of DIEA in refluxing BuOH to produce piperazinyl cyclopentapyrimidine (X). Oxidation of compound (X) using mCPBA and NaHCO3 in CHCl3 furnishes N-oxide (XI). Subsequent rearrangement of N-oxide (XI) using Ac2O in CH2Cl2 at 100 °C yields acetate (XII). This compound (XII) is hydrolyzed with LiOH in H2O/THF to give alcohol (XIII), which upon Swern oxidation with (COCl)2, DMSO and Et3N in CH2Cl2 at -78 °C affords ketone (XIV) (1-6). Asymmetric transfer hydrogenation of ketone (XIV) in the presence of RuCl[(R,R)-TsDPEN(p-cymene)], HCOOH and Et3N in CH2Cl2, followed by protection with PNBCl in the presence of Et3N in CH2Cl2, and hydrolysis with LiOH in H2O/THF gives rise to alcohol (XV) (1-6). Also, intermediate (XV) can be produced by enzymatic reduction of ketone (XI) using KRED-101 in the presence of GDH, NADP, KOH and PEG-400, KRED-X1.1-P1F01 in the presence of glucose and NAD in DMSO/i-PrOH or KRED-X1.1-P1B06, KRED-X1.1-P1F01 or KRED-X1.1-P1H10 in the presence of NADP in DMSO/i-PrOH or i-PrOH (11,12). In an alternative method, asymmetric transfer hydrogenation of ketone (XIV) in the presence of RuCl[(R,R)-MsDPEN(p-cymene)], HCOOH and Et3N in CH2Cl2, followed by O-protection of the resultant cis/trans mixture of alcohols with PNBCl and Et3N or protection with pivaloyl chloride in the presence of DIEA in CH2Cl2, followed by separation of the resulting cis/trans mixture of esters by means of HPLC. Hydrolysis of trans ester with LiOH in THF yields alcohol (XV) (11). N-Deprotection of piperazine derivative (XV) by means of HCl in CH2Cl2, i-PrOH or toluene at 62 °C provides amine dihydrochloride (XVI) (1-7,11,12), which is then coupled with aminoacid derivative (XVIIa) (1-7,11) or its sodium salt (XVIIb) (12,13) in the presence of DIEA and HBTU in CH2Cl2 or NMM and T3P in i-PrOH or toluene to produce amide (XVIII) (1-7,11-13). Finally, Boc-deprotection of precursor (XVIII) by means of HCl in MeOH/Et2O, PrOH, i-PrOH or toluene at 57 °C furnishes the target GDC-0068

 Ipatasertib pk_prod_list.xml_prod_list_card_pr?p_tsearch=A&p_id=691990

Synthesis of intermediate (XVII): Condensation of methyl (4-chlorophenyl)acetate (XIX) with formaldehyde (XX) in the presence of NaOMe in DMSO gives beta-hydroxyester (XXI). Subsequent dehydration of alcohol (XXI) using MsCl and Et3N in CH2Cl2 provides arylacrylate (XXII), which upon conjugate addition with isopropylamine (XXIII) in the presence of Boc2O in THF yields N-Boc beta-aminoester (XXIV). Basic hydrolysis of ester (XXIV) using KOSiMe3 in THF generates the potassium carboxylate (XXV), which upon condensation with 4(R)-benzyl-2-oxazolidinone (XXVI) via activation with pivaloyl chloride and BuLi in THF at -78 °C affords the N-acyl oxazolidinone (XXVII) (2-6). Finally, removal of the chiral auxiliary group of (XXVII) using LiOH and H2O2 in THF/H2O furnishes the key intermediate (XVII) (1-6,11). Alternative synthesis of intermediate (XXVII): Protection of isopropylamine (XXIII) with Boc2O in toluene affords tert-butyl isopropylcarbamate (XXVIII), which upon N-alkylation with bromomethyl methyl ether (XXIX) in the presence of NaHMDS in 2-MeTHF gives tert-butyl isopropyl(methoxymethyl)carbamate (XXX) (11). Condensation of 4(R)-benzyl-2-oxazolidinone (XXVI) with 2-(4-chlorophenyl)acetyl chloride (XXXIIa) using BuLi in THF at -50 °C (1) or with 2-(4-chlorophenyl)acetic acid (XXXIIb) via activation with pivaloyl chloride and Et3N in refluxing toluene (11) affords N-acyl oxazolidinone(XXXI). After conversion of intermediate (XXXI) to its titanium enolate with TiCl4 and DIEA in CH2Cl2 at -50 °C, diastereoselective Mannich reaction with formaldehyde hemiaminal (XXX) affords adduct (XXVII)

PAPER

Synthesis of Akt inhibitor ipatasertib. Part 2. Total synthesis and first kilogram scale-up
Org Process Res Dev 2014, 18(12): 1652

https://pubs.acs.org/doi/full/10.1021/op500270z

https://pubs.acs.org/doi/suppl/10.1021/op500270z/suppl_file/op500270z_si_001.pdf

Synthesis of Akt Inhibitor Ipatasertib. Part 2. Total Synthesis and First Kilogram Scale-up

 Small Molecule Process Chemistry, Genentech, Inc., a member of the Roche Group, 1 DNA Way, South San Francisco, California 94080-4990, United States
 Array BioPharma Inc., 3200 Walnut Street, Boulder, Colorado 80301, United States
Org. Process Res. Dev.201418 (12), pp 1652–1666
DOI: 10.1021/op500270z
*E-mail: travisr@gene.com.
Abstract Image

Herein, the first-generation process to manufacture Akt inhibitor Ipatasertib through a late-stage convergent coupling of two challenging chiral components on multikilogram scale is described. The first of the two key components is a trans-substituted cyclopentylpyrimidine compound that contains both a methyl stereocenter, which is ultimately derived from the enzymatic resolution of a simple triester starting material, and an adjacent hydroxyl group, which is installed through an asymmetric reduction of the corresponding cyclopentylpyrimidine ketone substrate. A carbonylative esterification and subsequent Dieckmann cyclization sequence was developed to forge the cyclopentane ring in the target. The second key chiral component, a β2-amino acid, is produced using an asymmetric aminomethylation (Mannich) reaction. The two chiral intermediates are then coupled in a three-stage endgame process to complete the assembly of Ipatasertib, which is isolated as a stable mono-HCl salt.

(S)-2-(4-Chlorophenyl)-1-(4-((5R,7R)-7-hydroxy-5-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)piperazin-1-yl)-3-(isopropylamino)propan-1-one, Ipatasertib Mono-HCl

 Ipatasertib mono-HCl (3.23 kg, 80% yield) as an off-white solid. Analytical results: 99.7 A% [0.26% S,R,S-diastereomer observed)]; impurity 23 (M399) was not detected (<0.02 A%) [Method 2.2]; ruthenium content by IPC-AES = 5 ppm; analysis for PF6 anion by CAD-HPLC resulted in not detected [Method 2.3]; residual solvent = 0.4% EtOAc; ion chromatography (IC) = 8.5% chloride (1.14 salt equivalent); DSC = 141 °C; FTIR (neat) 3269 (br OH), 2961–2865 (N–H stretch), 1637 (C═O stretch); 1H NMR (600 MHz, DMSO-d6) 9.39 (s, 1H), 8.64 (s, 1H), 8.49 (s, 1H), 7.49 (q, J = 2.9 Hz, 2H), 7.41 (q, J = 2.9 Hz, 2H), 5.58 (s, 1H), 4.91 (t, J = 6.9 Hz, 1H), 4.78 (dd, J = 8.9, 4.5 Hz, 1H), 3.81 (m, J = 3.3 Hz, 1H), 3.68 (m, J = 3.3 Hz, 1H), 3.67 (m, J = 3.1 Hz, 1H), 3.65 (m, J = 3.2 Hz, 1H), 3.63 (m, J = 3.6 Hz, 1H), 3.59 (m, J = 4.3 Hz, 1H), 3.51 (m, J = 3.5 Hz, 1H), 3.46 (m, J = 3.5 Hz, 1H), 3.36 (m, J = 3.2 Hz, 1H), 3.30 (m, J = 5.7 Hz, 1H), 3.21 (m, J = 3.4 Hz, 1H), 2.98 (m, J = 5.8 Hz, 1H), 1.97 (m, J = 4.8 Hz, 2H), 1.26 (d, J = 6.6 Hz, 3H), 1.25 (d, J = 7.0 Hz, 3H); 13C NMR (150 MHz, DMSO-d6) 170.2, 168.2, 159.4, 155.2, 135.3, 132.5, 129.7 (2C), 129.1 (2C), 120.8, 71.7, 50.4, 47.0, 44.8, 44.5, 44.1, 41.4, 40.8, 34.5, 19.8, 18.4, 18.1; HRMS calcd for C24H32ClN5O2 457.2245; found [M+H]+ 458.2306.

str1

 Ipatasertib freebase (3.9 kg, 98.2 A% containing ~1.2% impurity 23 (M399) and impurity M416 at 0.2 A% [Method 2.2]) as tan solid. By CAD-HPLC (see Figure S1-2), the PF6 anion was present in ~0.86 A% [Method 2.3]; Ion chromatography (IC) = 4.0% chloride (0.56 salt equivalent); 1 H NMR (600 MHz, DMSO-d6) 8.44 (s, 1H), 7.45 (d, J = 8.5 Hz, 2H), 7.40 (d, J = 8.5 Hz, 2H), 5.48 (br s, 1H), 4.86 (t, J = 6.9 Hz, 1H), 4.58 (dd, J = 7.3, 4.6 Hz, 1H), 3.74 (m, 1H), 3.40 (m, 1H), 3.63 (m, 2H), 3.61 (m, 1H), 3.42 (m, 1H), 3.57 (m, 1H), 3.18 (m, 1H), 3.50 (m, J = 2.9 Hz, 1H), 3.09 (m, J = 3.1 Hz, 1H), 3.42 (m, 1H), 2.87 (m, J = 4.7 Hz, 1H), 2.00 (m, 1H), 1.92 (m, J = 3.1 Hz, 1H), 1.15 (d, J = 6.4 Hz, 6H), 1.03 (d, J = 6.9 Hz, 3H); 13C NMR (150 MHz, DMSO-d6) 172.0, 169.0, 159.6, 156.3, 136.3, 132.1, 129.7 (2C), 128.9 (2C), 120.9, 72.0, 49.4, 48.7, 45.4, 44.9, 44.8, 44.6, 41.4, 40.9, 34.3, 20.1, 19.9, 19.7; HRMS calcd for C24H32ClN5O2 [M+H]+ 458.2317; found 458.2312. See supporting information (S2) for the NMR spectra (DMSO-d6) of Ipatasertib freebase: ( 1 H) S2, Figure S2-5.12 and ( 13C) Figure S2-5.13.

https://pubs.acs.org/doi/suppl/10.1021/op500270z/suppl_file/op500270z_si_002.pdf

Table S2-1 1 H NMR Assignments of Ipatasertib mono-HCl. S2-52 Figure S2-5.10. 13C NMR (DMSO-d6) spectrum of Ipatasertib mono-HCl. S2-53 Table S2-2 13C NMR Assignments of Ipatasertib mono-HCl. S2-54 Table S2-3 Characteristic Ipatasertib mono-HCl Infrared Signals. S2-55 Figure S2-5.11. FTIR Spectrum of Ipatasertib mono-HCl. S2-56 Figure S2-5.12. XRPD Pattern of Ipatasertib mono-HCl. S2-57

PAPER

https://pubs.acs.org/doi/abs/10.1021/op500271w

https://pubs.acs.org/doi/suppl/10.1021/op500271w/suppl_file/op500271w_si_001.pdf

Synthesis of Akt Inhibitor Ipatasertib. Part 1. Route Scouting and Early Process Development of a Challenging Cyclopentylpyrimidine Intermediate

 Array BioPharma Inc., 3200 Walnut Street, Boulder, Colorado 80301, United States
 Genentech Inc., a member of the Roche Group, 1 DNA Way, South San Francisco, California 94080-4990, United States
Org. Process Res. Dev.201418 (12), pp 1641–1651
DOI: 10.1021/op500271w
Abstract Image

Herein, the route scouting and early process development of a key cyclopentylpyrimidine ketone intermediate toward the synthesis of Akt inhibitor Ipatasertib are described. Initial supplies of the intermediate were prepared through a method that commenced with the natural product (R)-(+)-pulegone and relied on the early construction of a methyl-substituted cyclopentyl ring system. The first process chemistry route, detailed herein, enabled the synthesis of the ketone on a hundred-gram scale, but it was not feasible for the requisite production of multikilogram quantities of this compound and necessitated the exploration of alternative strategies. Several new synthetic approaches were investigated towards the preparation of the cyclopentylpyrimidine ketone, in either racemic or chiral form, which resulted in the discovery of a more practical route that hinged on the initial preparation of a highly substituted dihydroxypyrimidine compound. The cyclopentane ring in the target was then constructed through a key carbonylative esterification and subsequent tandem Dieckmann cyclization–decarboxylation sequence that was demonstrated in a racemic synthesis. This proof-of-concept was later developed into an asymmetric synthesis of the cyclopentylpyrimidine ketone, which will be described in a subsequent paper, along with the synthesis of Ipatasertib.

PAPER

Discovery and preclinical pharmacology of a selective ATP-Competitive akt inhibitor (GDC-0068) for the treatment of human tumors
J Med Chem 2012, 55(18): 8110

PAPER

Asymmetric synthesis of akt kinase inhibitor ipatasertib
Org Lett 2017, 19(18): 4806

PATENT

WO 2008006040

PATENT

WO 2012135753

PATENT

WO 2012135759

PATENT

WO 2012135781

PATENT

WO 2013173784

PATENT

WO 2015073739

PATENT

WO 2012135779

PATENT

WO 2013173768

References

  1. Jump up^ https://www.clinicaltrials.gov/ct2/show/NCT02301988
  2. Jump up^ Lin K, Friedman L, Gloor S, Gross S, Liederer BM, Mitchell I, et al. Preclinical characterization of GDC-0068, a novel selective ATP competitive inhibitor of Akt. 22nd-EORTC-NCI-AACR-2010 2010; abstr. 79
Ipatasertib
Ipatasertib.svg
Clinical data
Routes of
administration
PO
ATC code
  • None
Identifiers
ChemSpider
KEGG
Chemical and physical data
Formula C24H32ClN5O2
Molar mass 458.00 g·mol−1
3D model (JSmol)

////////////// ipatasertib, orphan drug designation, GDC-0068 , RG7440, PHASE 3

CC(C)NC[C@@H](C(=O)N1CCN(CC1)c2ncnc3[C@H](O)C[C@@H](C)c23)c4ccc(Cl)cc4

It has been found that ipatasertib exhibits a very high solubility (>1 g/g water; >2 g/g water/ethanol 1:1) and a very high hygroscopicity (˜6% at 50% RH, >35% at 95% RH). Whereas poor solubility is often a limiting factor in the development of galenical formulations of other API’s (active pharmaceutical ingredient), a high solubility can equally be problematic for the process performance. Due to this very high intrinsic hygroscopicity of the API, ipatasertib drug substance tends to auto-dissolve to a honey-like viscous liquid at increased humidity. Such high solubility and hygroscopicity may pose serious problems for processing as well as for stability and shelf-life of the final product. Therefore, conventional pharmaceutical compositions comprising ipatasertib and processes for the manufacture of pharmaceutical compositions comprising wetting (e.g. wet granulation) are difficult due to the high solubility and high hygroscopicity of the API.


FDA approves first drug Epidiolex (cannabidiol) comprised of an active ingredient derived from marijuana to treat rare, severe forms of epilepsy

$
0
0
The U.S. Food and Drug Administration today approved Epidiolex (cannabidiol) [CBD] oral solution for the treatment of seizures associated with two rare and severe forms of epilepsy, Lennox-Gastaut syndrome and Dravet syndrome, in patients two years of age and older. This is the first FDA-approved drug that contains a purified drug substance derived from marijuana. It is also the first FDA approval of a drug for the treatment of patients with Dravet syndrome.

June 25, 2018

Release

The U.S. Food and Drug Administration today approved Epidiolex (cannabidiol) [CBD] oral solution for the treatment of seizures associated with two rare and severe forms of epilepsy, Lennox-Gastaut syndrome and Dravet syndrome, in patients two years of age and older. This is the first FDA-approved drug that contains a purified drug substance derived from marijuana. It is also the first FDA approval of a drug for the treatment of patients with Dravet syndrome.

CBD is a chemical component of the Cannabis sativa plant, more commonly known as marijuana. However, CBD does not cause intoxication or euphoria (the “high”) that comes from tetrahydrocannabinol (THC).

It is THC (and not CBD) that is the primary psychoactive component of marijuana.

“This approval serves as a reminder that advancing sound development programs that properly evaluate active ingredients contained in marijuana can lead to important medical therapies. And, the FDA is committed to this kind of careful scientific research and drug development,” said FDA Commissioner Scott Gottlieb, M.D. “Controlled clinical trials testing the safety and efficacy of a drug, along with careful review through the FDA’s drug approval process, is the most appropriate way to bring marijuana-derived treatments to patients. Because of the adequate and well-controlled clinical studies that supported this approval, prescribers can have confidence in the drug’s uniform strength and consistent delivery that support appropriate dosing needed for treating patients with these complex and serious epilepsy syndromes. We’ll continue to support rigorous scientific research on the potential medical uses of marijuana-derived products and work with product developers who are interested in bringing patients safe and effective, high quality products. But, at the same time, we are prepared to take action when we see the illegal marketing of CBD-containing products with serious, unproven medical claims. Marketing unapproved products, with uncertain dosages and formulations can keep patients from accessing appropriate, recognized therapies to treat serious and even fatal diseases.”

Dravet syndrome is a rare genetic condition that appears during the first year of life with frequent fever-related seizures (febrile seizures). Later, other types of seizures typically arise, including myoclonic seizures (involuntary muscle spasms). Additionally, status epilepticus, a potentially life-threatening state of continuous seizure activity requiring emergency medical care, may occur. Children with Dravet syndrome typically experience poor development of language and motor skills, hyperactivity and difficulty relating to others.

Lennox-Gastaut syndrome begins in childhood. It is characterized by multiple types of seizures. People with Lennox-Gastaut syndrome begin having frequent seizures in early childhood, usually between ages 3 and 5. More than three-quarters of affected individuals have tonic seizures, which cause the muscles to contract uncontrollably. Almost all children with Lennox-Gastaut syndrome develop learning problems and intellectual disability. Many also have delayed development of motor skills such as sitting and crawling. Most people with Lennox-Gastaut syndrome require help with usual activities of daily living.

“The difficult-to-control seizures that patients with Dravet syndrome and Lennox-Gastaut syndrome experience have a profound impact on these patients’ quality of life,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “In addition to another important treatment option for Lennox-Gastaut patients, this first-ever approval of a drug specifically for Dravet patients will provide a significant and needed improvement in the therapeutic approach to caring for people with this condition.”

Epidiolex’s effectiveness was studied in three randomized, double-blind, placebo-controlled clinical trials involving 516 patients with either Lennox-Gastaut syndrome or Dravet syndrome. Epidiolex, taken along with other medications, was shown to be effective in reducing the frequency of seizures when compared with placebo.

The most common side effects that occurred in Epidiolex-treated patients in the clinical trials were: sleepiness, sedation and lethargy; elevated liver enzymes; decreased appetite; diarrhea; rash; fatigue, malaise and weakness; insomnia, sleep disorder and poor quality sleep; and infections.

Epidiolex must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks. As is true for all drugs that treat epilepsy, the most serious risks include thoughts about suicide, attempts to commit suicide, feelings of agitation, new or worsening depression, aggression and panic attacks. Epidiolex also caused liver injury, generally mild, but raising the possibility of rare, but more severe injury. More severe liver injury can cause nausea, vomiting, abdominal pain, fatigue, anorexia, jaundice and/or dark urine.

Under the Controlled Substances Act (CSA), CBD is currently a Schedule I substance because it is a chemical component of the cannabis plant. In support of this application, the company conducted nonclinical and clinical studies to assess the abuse potential of CBD.

The FDA prepares and transmits, through the U.S. Department of Health and Human Services, a medical and scientific analysis of substances subject to scheduling, like CBD, and provides recommendations to the Drug Enforcement Administration (DEA) regarding controls under the CSA. DEA is required to make a scheduling determination.

The FDA granted Priority Review designation for this application. Fast-Track designation was granted for Dravet syndrome. Orphan Drug designation was granted for both the Dravet syndrome and Lennox-Gastaut syndrome indications.

The FDA granted approval of Epidiolex to GW Research Ltd.

Image result for Epidiolex (cannabidiol)
/////////// Epidiolex, cannabidiol, fda 2018, Dravet syndrome, epilepsy, Priority Review , Fast-Track designation, Orphan Drug designation

Afloqualone, アフロクアロン

$
0
0

Afloqualone.pngChemSpider 2D Image | Afloqualone | C16H14FN3OAfloqualone.svg

Afloqualone

Molecular Formula: C16H14FN3O
Molecular Weight: 283.306 g/mol

6-amino-2-(fluoromethyl)-3-(2-methylphenyl)quinazolin-4-one

HQ 495C033541, QA-3735, UNII:CO4U2C8ORZ

4(3H)-Quinazolinone, 6-amino-2-(fluoromethyl)-3-(2-methylphenyl)- [ACD/Index Name]
4831
56287-74-2 [RN], アフロクアロン

Afloqualone; 56287-74-2; Arofuto; Aroft; Afloqualon; Afloqualone [INN:JAN]

Afloqualone (Arofuto) is a quinazolinone family GABAergic drug and is an analogue of methaqualone developed in the 1970s by a team at Tanabe Seiyaku.[1] It has sedative and muscle-relaxant effects resulting from its agonist activity at the β subtype of the GABAareceptor ,[2] and has had some clinical use, although it causes photosensitization as a side-effect that can cause skin problems such as dermatitis.[3]

PATENT

CN 106496145

PATENT

CN 106496144

https://patents.google.com/patent/CN106496144A/en

Example 1:

[0027] A-fluoro-quinolin-one process, comprising the steps of:

[0028] A. To the hydrogenation apparatus 10g 6- nitro-2- (fluoromethyl) -3- (2-methylphenyl) -4- (3H) -1,3- phthalazinone , 150ml acid content of 0.1 ~ 0.4N n-butanol solution of acetic acid, lg palladium ruthenium bimetallic catalyst, hydrogen pressure 0.02 ~ 0.4MPa, reaction temperature of 25-50 ° C, after 1 hour, filtered to give the filtrate ;

[0029] B. washed catalyst with ethanol, at normal temperature, under reduced pressure to obtain a solution ⑴;

[0030] C. was added to the filtrate and the solution ⑴ water, 0.1N sodium hydroxide solution was added, the pH adjusted to 10.2 to 11.0, and stirred at 50-60 ° C 0.5 h, cooled to room temperature and filtered to give the crude fluoro-quinolin-one;

[0031] D.-fluoro-quinolin per gram of the recrystallization solvent was added 5 ~ 15ml crude ketone, wherein the recrystallization solvent is a volume ratio of 1: 1: 0.2 in a solution of isopropanol (m), a solution of acid butyl ester (II ) and water mixture; crystallized at room temperature, filtered to give a fluorine methaqualone.

[0032] Example 2:

[0033] A-fluoro-quinolin-one process, comprising the steps of:

[0034] A. hydrogenation apparatus added to 20g 6- nitro _2_ (fluoromethyl) -3- (2_-methylphenyl) -4- (3-1,3-phthalazinone buckle, acid content of the acid-containing 240ml 0.1 ~ 0.4N ethanol solution of hydrochloric acid, lg palladium ruthenium bimetallic catalyst, hydrogen pressure 0.02 ~ 0.4MPa, reaction temperature of 25-50 ° C. after 0.5 hours the reaction was filtered to obtain filtrate;

[0035] B. washed catalyst with ethanol, at normal temperature, under reduced pressure to obtain a solution ⑴;

[0036] C. was added to the filtrate and the solution ⑴ water, 0.1N sodium hydroxide solution was added, the pH adjusted to 10.2 to 11.0, and stirred at 50-60 ° C 1 hour, cooled to room temperature and filtered to give the crude fluoro-quinolin-one;

[0037] D.-fluoro-quinolin added per gram of crude ketone was recrystallized from 5 ~ 15ml of the solvent, wherein the recrystallization solvent is a volume ratio of 1: o.2: methanol solution of i (m), an ethyl acetate solution (II ) and water mixture; crystallized at room temperature, filtered to give a fluorine methaqualone.

[0038] Example 3:

[0039] – quinolin-fluoro-one kind of process, comprising the steps of:

[0040] A. Add 5g 6- nitro apparatus _2_ hydride (fluoromethyl) -3- (2-methylphenyl) -4- (3-1,3-Perot phthalazinone, 80ml methanol containing an acid in an amount of 0.1 ~ 0.4N solution of sulfuric acid, lg palladium ruthenium bimetallic catalyst, hydrogen pressure 0.02 ~ 0.4MPa, reaction temperature of 25-50 ° C. after 1.5 hours the reaction was filtered to obtain filtrate;

[0041] B. the catalyst was washed with ethanol, normal temperature under reduced pressure to obtain a solution (the I);

[0042] C. was added to the filtrate and the solution (I) water, 0.1N sodium hydroxide solution was added, the pH adjusted to 10.2 to 11.0, and stirred at 50-60 ° C 1 hour, cooled to room temperature and filtered to give fluoro-quinolin-one Crude;

[0043] D. methaqualone fluorine per gram of crude product were added 5 ~ 15ml recrystallization solvent, wherein the recrystallization solvent is a volume ratio of 1: 0.2: 0.2 ethanol solution (m), carboxylic acid butyl ester (II) and water mixture; crystallization at room temperature, and filtered to give fluoro-quinolin-one.

PAPER

6-Amino-2-(fluoromethyl)-3-(2-methylphenyl)quinazolin-4(3H)-one
Acta Crystallographica, Section E: Structure Reports Online (2007), 63, (7), o3109

http://scripts.iucr.org/cgi-bin/paper?S1600536807026670

PAPER

Synthesis of the metabolites of afloqualone and related compounds
Chemical & pharmaceutical bulletin (1983), 31, (4), 1158-65.

Seven main metabolites (3-9) of afloqualone (1, 6-amino-2-fluoromethyl-3-(o-tolyl)-4 (3H)-quinazolinone and related 4 (3H)-quinazolinone derivatives were synthesized. The metabolites 4 and 5 containing a sulfur atom were prepared by the reaction of 6-acetamido-2-chloromethyl-3-(o-tolyl)-4 (3H)-quinazolinone (11) with NaSCH3 followed by oxidation with H2O2. Reaction of 11 and N-acetyl-L-cysteine gave the mercapturic acid-conjugated metabolite 6. Condensation of 2-fluoroacetamido-5-nitrobenzoic acid (19) and 2-amino-benzyl alcohol (20) with dicyclohexylcarbodiimide (DCC) in the presence of 1-hydroxy-benzotriazole afforded 2-fluoromethyl-3-(o-hydroxymethylphenyl)-6-nitro-4 (3H)-quinazolinone (21), which was converted to the metabolites 7 and 8. Treatment of the 2-bromomethyl-4 (3H)-quinazolinone (24) with AgBF4-H2O in dimethylsulfoxide (DMSO) gave the 2-hydroxymethyl metabolite 9. None of the main metabolites (2-9) showed significant central nervous system depressant activity

https://www.jstage.jst.go.jp/article/cpb1958/31/4/31_4_1158/_article

References

  1. Jump up^ US Patent 3966731 – 2-Fluoromethyl-3-o-tolyl-6-amino-4(3H)-quinazolinone
  2. Jump up^ Ochiai T, Ishida R. Pharmacological studies on 6-amino- 2-fluoromethyl- 3-(O-tolyl)- 4(3H)- quinazolinone (afloqualone), a new centrally acting muscle relaxant. (II) Effects on the spinal reflex potential and the rigidity. Japanese Journal of Pharmacology. 1982 Jun;32(3):427-38.
  3. Jump up^ Ishikawa T, Kamide R, Niimura M. Photoleukomelanodermatitis (Kobori) induced by afloqualone. Journal of Dermatology. 1994 Jun;21(6):430-3.
Afloqualone
Afloqualone.svg
Clinical data
AHFS/Drugs.com International Drug Names
ATC code
  • none
Legal status
Legal status
  • US: Unscheduled
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C16H14FN3O
Molar mass 283.3
3D model (JSmol)

//////////////Afloqualone, HQ 495, アフロクアロン , C033541, QA-3735, UNII:CO4U2C8ORZ, 4831

CC1=CC=CC=C1N2C(=NC3=C(C2=O)C=C(C=C3)N)CF

Afloqualone

    • ATC:M03A
  • Use:muscle relaxant
  • Chemical name:6-amino-2-(fluoromethyl)-3-(2-methylphenyl)-4(3H)-quinazolinone
  • Formula:C16H14FN3O
  • MW:283.31 g/mol
  • CAS-RN:56287-74-2
  • LD50:397 mg/kg (M, p.o.);
    249 mg/kg (R, p.o.)

Derivatives

hydrochloride

  • Formula:C16H14FN3O • xHCl
  • MW:unspecified
  • CAS-RN:56287-75-3

Substance Classes

Synthesis Path

Substances Referenced in Synthesis Path

CAS-RN Formula Chemical Name CAS Index Name
108-24-7 C4H6O3 acetic anhydride Acetic acid, anhydride
69123-71-3 C7H5ClN2O3 2-amino-5-nitrobenzoyl chloride Benzoyl chloride, 2-amino-5-nitro-
23076-31-5 C14H13N3O3 N-(2-amino-5-nitrobenzoyl)-o-toluidine Benzamide, 2-amino-N-(2-methylphenyl)-5-nitro-
56287-72-0 C16H14FN3O4 2-[(fluoroacetyl)amino]-N-(2-methylphenyl)-5-nitrobenzamide Benzamide, 2-[(fluoroacetyl)amino]-N-(2-methylphenyl)-5-nitro-
359-06-8 C2H2ClFO fluoroacetyl chloride Acetyl chloride, fluoro-
56287-73-1 C16H12FN3O3 2-(fluoromethyl)-3-(2-methylphenyl)-6-nitro-4(3H)-quinazolinone 4(3H)-Quinazolinone, 2-(fluoromethyl)-3-(2-methylphenyl)-6-nitro-
616-79-5 C7H6N2O4 5-nitroanthranilic acid Benzoic acid, 2-amino-5-nitro-
95-53-4 C7H9N o-toluidine Benzenamine, 2-methyl-

Trade Names

Country Trade Name Vendor Annotation
J Aflomus Hishiyama
Airomate SawaiNippon Chemiphar
Arofuto Tanabe

Formulations

  • tabl. 20 mg

References

    • Tani, J. et al.: J. Med. Chem. (JMCMAR) 22, 95 (1979).
    • DOS 2 449 113 (Tanabe; appl. 15.10.1974; J-prior. 15.10.1973).
    • US 3 966 731 (Tanabe; 29.6.1976; J-prior. 15.10.1973)
Title: Afloqualone
CAS Registry Number: 56287-74-2
CAS Name: 6-Amino-2-(fluoromethyl)-3-(2-methylphenyl)-4(3H)-quinazolinone
Additional Names: 6-amino-2-fluoromethyl-3-(o-tolyl)-4(3H)-quinazolinone
Manufacturers’ Codes: HQ-495
Trademarks: Arofuto (Tanabe)
Molecular Formula: C16H14FN3O
Molecular Weight: 283.30
Percent Composition: C 67.83%, H 4.98%, F 6.71%, N 14.83%, O 5.65%
Literature References: A centrally acting muscle relaxant. Prepn: I. Inoue et al., DE 2449113eidem, US 3966731 (1975, 1976 to Tanabe); J. Tani et al., J. Med. Chem. 22, 95 (1979). Pharmacology: T. Ochiai, R. Ishida, Jpn. J. Pharmacol. 31, 491 (1981); 32,427 (1982). Metabolism: N. Otsuka et al., J. Pharmacobio-Dyn. 5, S-59 (1982); S. Furuuchi et al., Drug Metab. Dispos. 11, 371 (1983).
Properties: Pale yellow prisms from 2-propanol, mp 195-196°. LD50 in mice (mg/kg): 315.1 i.p. (Tani).
Melting point: mp 195-196°
Toxicity data: LD50 in mice (mg/kg): 315.1 i.p. (Tani)
Therap-Cat: Muscle relaxant (skeletal).
Keywords: Muscle Relaxant (Skeletal).

Alfuzosin, 塩酸アルフゾシン

$
0
0

Image result for alfuzosinChemSpider 2D Image | Alfuzosin | C19H27N5O4

Alfuzosin

  • Molecular FormulaC19H27N5O4
  • Average mass389.449 Da
N-{3-[(4-Amino-6,7-dimethoxy-2-quinazolinyl)(methyl)amino]propyl}tetrahydro-2-furancarboxamide
N-{3-[(4-amino-6,7-dimethoxyquinazolin-2-yl)(methyl)amino]propyl}tetrahydrofuran-2-carboxamide
SL 77499-10
UNII:90347YTW5F
Urion
Xatral
2-furancarboxamide, N-[3-[(4-amino-6,7-dimethoxy-2-quinazolinyl)methylamino]propyl]tetrahydro-
5357
cas 81403-80-7 [RN]

CAS

CAS: 81403-68-1  HCL SALT
90347YTW5F
塩酸アルフゾシン
Title: Alfuzosin
CAS Registry Number: 81403-80-7
CAS Name: N-[3-[(4-Amino-6,7-dimethoxy-2-quinazolinyl)methylamino]propyl]tetrahydro-2-furancarboxamide
Additional Names: N1-(4-amino-6,7-dimethoxyquinazol-2-yl)-N1-methyl-N2-(tetrahydrofuroyl-2)-propylenediamine
Manufacturers’ Codes: SL-77.499
Molecular Formula: C19H27N5O4
Molecular Weight: 389.45
Percent Composition: C 58.60%, H 6.99%, N 17.98%, O 16.43%
Literature References: a1-Adrenoceptor antagonist structurally similar to prazosin, q.v. Prepn: P. M. J. Manoury, DE 2904445idem, US 4315007 (1979, 1982 both to Synthelabo); and antihypertensive activity in rats: P. M. Manoury et al., J. Med. Chem. 29,19 (1986). Pharmacology: A. G. Ramage, Eur. J. Pharmacol. 129, 307 (1986). HPLC determn in biological fluids: P. Guinebault et al., J. Chromatogr. 353, 361 (1986). Pharmacology in humans: A. H. Deering, Br. J. Clin. Pharmacol. 25, 417 (1988). Clinical evaluation in essential hypertension: S. Leto Di Priolo et al., Eur. J. Clin. Pharmacol. 35, 25 (1988); A. K. Ghosh, S. Ghosh, Ger. Cardiovasc. Med. 1, 81 (1988). Clinical trial in benign prostatic hyperplasia (BPH): C. G. Roehrborn et al., BJU Int. 92, 257 (2003). Review of clinical experience in BPH: D. M. Weiner, F. C. Lowe, Expert Opin. Pharmacother. 4, 2057-2063 (2003).
Alfuzosin hydrochloride: sc-203812...

Alfuzosin hydrochloride (CAS 81403-68-1)

Derivative Type: Hydrochloride
CAS Registry Number: 81403-68-1
Manufacturers’ Codes: SL-77.499-10
Trademarks: Mittoval (Schering AG); Urion (Zambon); UroXatral (Sanofi-Synthelabo); Xatral (Sanofi-Synthelabo)
Molecular Formula: C19H27N5O4.HCl
Molecular Weight: 425.91
Percent Composition: C 53.58%, H 6.63%, N 16.44%, O 15.03%, Cl 8.32%
Properties: Crystals from ethanol + ether, mp 225° (Manoury, 1986), also reported earlier as mp 235° (dec) (Manoury, 1982). pKa 8.13.
Melting point: mp 225° (Manoury, 1986); mp 235° (dec) (Manoury, 1982)
pKa: pKa 8.13
Therap-Cat: Antihypertensive. In treatment of benign prostatic hypertrophy.
Keywords: Antihypertensive; Quinazoline Derivatives; Antiprostatic Hypertrophy; a-Adrenergic Blocker.

Alfuzosin (INN, provided as the hydrochloride salt) is a pharmaceutical drug of the α1 blocker class. As an antagonist of the α1adrenergic receptor, it works by relaxing the muscles in the prostate and bladder neck, making it easier to urinate. It is thus used to treat benign prostatic hyperplasia (BPH).[1]

Alfuzosin is marketed in the United States by Sanofi Aventis under the brand name Uroxatral and elsewhere under the tradenames Xat, Xatral, Prostetrol and Alfural. Alfuzosin was approved by the U.S. FDA for treatment of BPH in June 2003.

Side effects

The most common side effects are dizziness (due to postural hypotension), upper respiratory tract infectionheadachefatigue, and abdominal disturbances. Side effects include stomach pain, heartburn, and congested nose.[2] Adverse effects of alfuzosin are similar to that of tamsulosin with the exception of retrograde ejaculation.[3]

Contraindications

Alfuzosin should be used with caution in patients with severe renal insufficiency, and should not be prescribed to patients with a known history of QT prolongation who are taking medications known to prolong the QT interval.

Chemistry

Alfuzosin contains a stereocenter and is therefore chiral. There are two enantiomeric forms, (R)-alfuzosin and (S)-alfuzosin. The drug is used as a racemate, (RS)-alfuzosin, a 1: 1 mixture of the (R)- and (S)-forms.[4]

Enantiomers of alfuzosin
Strukturformel des (R)-Enantiomers
CAS number: 123739-69-5
Strukturformel des (S)-Enantiomers
CAS number.: 123739-70-8

Alfuzosin

    • ATC:G04CA01
  • Use:antihypertensive, α1-adrenoceptor antagonist, α-blocker, treatment of benign prostatic hypertrophy (BPH)
  • Chemical name:(±)-N-[3-[(4-amino-6,7-dimethoxy-2-quinazolinyl)methylamino]propyl]tetrahydro-2-furancarboxamide
  • Formula:C19H27N5O4
  • MW:389.46 g/mol
  • CAS-RN:81403-80-7

Derivatives

monohydrochloride

  • Formula:C19H27N5O4 • HCl
  • MW:425.92 g/mol
  • CAS-RN:81403-68-1

Substance Classes

Synthesis Path

Substances Referenced in Synthesis Path

CAS-RN Formula Chemical Name CAS Index Name
23680-84-4 C10H10ClN3O2 4-amino-2-chloro-6,7-dimethoxyquinazoline 4-Quinazolinamine, 2-chloro-6,7-dimethoxy-
5004-88-6 C9H12N2O3 2-amino-4,5-dimethoxybenzamide Benzamide, 2-amino-4,5-dimethoxy-
541-41-3 C3H5ClO2 chloroformic acid ethyl ester Carbonochloridic acid, ethyl ester
72104-44-0 C9H14N2O2 2-cyano-N-methyl-N-tetrahydrofuroylethylamine 2-Furancarboxamide, N-(2-cyanoethyl)tetrahydro-N-methyl-
27631-29-4 C10H8Cl2N2O2 2,4-dichloro-6,7-dimethoxyquinazoline Quinazoline, 2,4-dichloro-6,7-dimethoxy-
28888-44-0 C10H10N2O4 2,4-dihydroxy-6,7-dimethoxyquinazoline 2,4(1H,3H)-Quinazolinedione, 6,7-dimethoxy-
20357-25-9 C9H9NO5 4,5-dimethoxy-2-nitrobenzaldehyde Benzaldehyde, 4,5-dimethoxy-2-nitro-
4959-60-8 C9H10N2O5 4,5-dimethoxy-2-nitrobenzamide Benzamide, 4,5-dimethoxy-2-nitro-
28888-44-0 C10H10N2O4 6,7-dimethoxyquinazoline-2,4-dione 2,4(1H,3H)-Quinazolinedione, 6,7-dimethoxy-
541-41-3 C3H5ClO2 ethyl chloroformate Carbonochloridic acid, ethyl ester
693-05-0 C4H8N2 3-(methylamino)propanenitrile Propanenitrile, 3-(methylamino)-
81403-67-0 C9H18N2O2 N1-methyl-N2-tetrahydrofuroyltrimethylenediamine 2-Furancarboxamide, tetrahydro-N-[3-(methylamino)propyl]-
16874-33-2 C5H8O3 (±)-tetrahydrofuran-2-carboxylic acid 2-Furancarboxylic acid, tetrahydro-
167391-50-6 C8H12O5 tetrahydro-2-furancarboxylic acid anhydride with ethyl hydrogen carbonate 2-Furancarboxylic acid, tetrahydro-, anhydride with ethyl hydrogen carbonate
57-13-6 CH4N2O urea Urea
120-14-9 C9H10O3 veratraldehyde Benzaldehyde, 3,4-dimethoxy-

Trade Names

Country Trade Name Vendor Annotation
D Alfunar Apogepha
Alfusin TAD Pharma
Urion Sanofi-Aventis
Uroxatral Sanofi-Aventis
F Urion Zambon
Xatral Sanofi-Aventis
GB Xatral Sanofi-Aventis
I Mittoval Sanofi-Aventis
Xatral Sanofi-Aventis

Formulations

  • film tabl. 2.5 mg; retard tabl. 10 mg (hydrochloride)

References

    • Manoury, P.M. et al.: J. Med. Chem. (JMCMAR) 29, 19 (1986).
    • US 4 315 007 (Synthelabo; 9.2.1982; F-prior. 6.2.1978, 29.12.1978).
    • DE 2 904 445 (Synthelabo; appl. 16.8.1979; F-prior. 6.2.1978, 29.12.1978).
  • synthesis of 6,7-dimethoxyquinazoline-2,4-dione:

    • Althuis, T.H.; Hess, H.J.: J. Med. Chem. (JMCMAR) 20, 146 (1977).

SYN

Mathias Scheer, “Alfuzosin tablets and synthesis.” U.S. Patent US20060062845, issued March 23, 2006.

US20060062845

Syn,  DOI: 10.1021/jm00151a003 NB: (WO2009001369)

Image result for alfuzosin

Image result for alfuzosin

FTIR spectrum of alfuzosin hydrochloride 

CLIP

str1 str2

Add the following:
Alfuzosin Hydrochloride
Click to View Image

C19H27N5O4·HCl 425.91

2-Furancarboxamide, N-[3-[(4-amino-6,7-dimethoxy-2-quinazolinyl)methylamino]propyl]tetrahydro-, monohydrochloride (±).
(±)-N-[3-[(4-Amino-6,7-dimethoxy-2-quinazolinyl)methylamino]propyl]tetrahydro-2-furamide monohydrochloride [81403-68-1].
» Alfuzosin Hydrochloride contains not less than 99.0 percent and not more than 101.0 percent of C19H27N5O4·HCl, calculated on the anhydrous basis.
Packaging and storage— Preserve in tight, well-closed containers, protected from light and humidity. Store at room temperature.

Identification—

B: It meets the requirements of the test for Chloride 191.

pH 791between 4.0 and 5.5

Test solution: 20 mg per mL, in carbon dioxide-free water.

Optical rotation 7810.10 to +0.10

Test solution: 20 mg per mL, in carbon dioxide-free water.
Water, Method I 921not more than 0.5%.
Residue on ignition 281not more than 0.1%.

Related compounds—

Solution A— Dilute 5.0 mL of perchloric acid in 900 mL of water, adjust with 2 M sodium hydroxide solution to a pH of 3.5, and dilute with water to 1000 mL.
Mobile phase— Prepare a filtered and degassed mixture of Solution A, acetonitrile, and tetrahydrofuran (80:20:1). Make adjustments if necessary (see System Suitability under Chromatography 621).
System suitability solution— Dissolve an accurately weighed quantity of USP Alfuzosin System Suitability Mixture RS in Mobile phase, and dilute quantitatively with Mobile phase to obtain a solution containing about 0.4 mg per mL.
Test solution— Dissolve 40.0 mg of Alfuzosin Hydrochloride in Mobile phase, and dilute with Mobile phase to 100.0 mL.
Reference solution— Quantitatively dilute an accurately measured volume of the Test solution by a factor of 1000 with Mobile phase.

Chromatographic system (see Chromatography 621) The liquid chromatograph is equipped with a detector set at 254 nm and a 4.6-mm × 15-cm column that contains 5-µm packing L1. The flow rate is about 1.5 mL per minute. Chromatograph the System suitability solution, and record the peak responses as directed for Procedure: the peak-to-valley ratio is at least 5. [NOTE—The peak-to-valley ratio is determined as the ratio of the height above the baseline of the impurity A peak to the height above the baseline of the lowest point of the curve separating this impurity peak from the peak due to alfuzosin.]

Procedure— Separately inject equal volumes (about 10 µL) of the Reference solution and the Test solution, record the chromatograms, and measure the peak responses. Calculate the percentage of each impurity in the portion of Alfuzosin Hydrochloride taken by the formula:

100[r/ (1000 rS)]

in which 100 is the percentage conversion factor; rU is the peak response for any impurity obtained from the Test solution; 1000 is the dilution factor; and rS is the peak response for alfuzosin obtained from the Reference solution: the limits are as shown in the accompanying table. Disregard any peak with an area less than 0.05%.

Compound Relative 
Retention Time
Limit 
(%)
Alfuzosin 1.0
Impurity A1 1.2 *
Impurity D2 0.5 0.20
Any individual unspecified impurity 0.10
Total impurities 0.30
1  N-[3-[(4-Amino-6,7-dimethoxyquinazolin-2-yl)(methyl)amino]propyl]furan-2-carboxamide.
2  N-(4-Amino-6,7-dimethoxyquinazolin-2-yl)-N-methylpropane-1,3-diamine.
*  Impurity A, a component of USP Alfuzosin System Suitability Mixture RS, is not a specified impurity.
Assay— Dissolve about 300 mg of Alfuzosin Hydrochloride, accurately weighed, in a mixture of 40 mL of anhydrous acetic acid and 40 mL of acetic anhydride. Titrate with 0.1 M perchloric acid, determining the endpoint potentiometrically. Each mL of 0.1 M perchloric acid is equivalent to 42.59 mg of C19H27N5O4·HCl.USP32

Auxiliary Information— Please check for your question in the FAQs before contacting USP.

Topic/Question Contact Expert Committee
Monograph Daniel K. Bempong, Ph.D.
Senior Scientist
1-301-816-8143
(MDPS05) Monograph Development-Pulmonary and Steroids
Reference Standards Lili Wang, Technical Services Scientist
1-301-816-8129
RSTech@usp.org
USP32–NF27 Page 1449

Pharmacopeial Forum: Volume No. 34(1) Page 69

Chromatographic Column—

Chromatographic columns text is not derived from, and not part of, USP 32 or NF 27.

References

  1. Jump up^ Lepor, Herbert (2016). “Alpha-blockers for the Treatment of Benign Prostatic Hyperplasia”Urologic Clinics of North America43 (3): 311–23. doi:10.1016/j.ucl.2016.04.009PMC 2213889Freely accessiblePMID 27476124.
  2. Jump up^ “Alfuzosin”MedlinePlusUnited States National Library of Medicine. April 15, 2016.
  3. Jump up^ Hills, Robert K; Liu, Chenli; Zeng, Guohua; Kang, Ran; Wu, Wenqi; Li, Jiasheng; Chen, Kang; Wan, Show P. (2015). “Efficacy and Safety of Alfuzosin as Medical Expulsive Therapy for Ureteral Stones: A Systematic Review and Meta-Analysis”PLOS ONE10 (8): e0134589. doi:10.1371/journal.pone.0134589ISSN 1932-6203PMC 4526635Freely accessiblePMID 26244843. This article incorporates text available under the CC BY 4.0 license.
  4. Jump up^ Rote Liste Service GmbH (Hrsg.): Rote Liste 2017 – Arzneimittelverzeichnis für Deutschland (einschließlich EU-Zulassungen und bestimmter Medizinprodukte). Rote Liste Service GmbH, Frankfurt/Main, 2017, Aufl. 57, S. 159, ISBN 978-3-946057-10-9.

External links

Alfuzosin
Alfuzosin.svg
Clinical data
Pronunciation /ælˈfjuːzsɪn/ al-FEW-zoh-sin
Trade names Uroxatral, others
AHFS/Drugs.com Monograph
MedlinePlus a64002
Pregnancy
category
  • AU: B2
  • US: B (No risk in non-human studies)
Routes of
administration
By mouth (tablets)
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability 49%
Protein binding 82–90%
Metabolism Liver (CYP3A4-mediated)
Elimination half-life 10 hours
Excretion Feces (69%) and Urine (24%)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.108.671 Edit this at Wikidata
Chemical and physical data
Formula C19H27N5O4
Molar mass 389.46 g·mol−1
3D model (JSmol)

/////////////////塩酸アルフゾシン, Uroxatral, alfuzosin

COC1=C(OC)C=C2C(N)=NC(=NC2=C1)N(C)CCCNC(=O)C1CCCO1

Rebamipide, ребамипид , ريباميبيد ,瑞巴派特 ,

$
0
0

ChemSpider 2D Image | Rebamipide | C19H15ClN2O4DB11656.pngRebamipide.svg

Rebamipide

  • Molecular FormulaC19H15ClN2O4
  • Average mass370.786 Da
  • Monoisotopic mass370.072021 Da

OPC-12759
OPC-12759E
OPC-759

(±)-a-(p-Chlorobenzamido)-1,2-dihydro-2-oxo-4-quinolinepropionic acid
2-(4-Chlorobenzoylamino)-3-[2(1H)-quinolinon-4-yl]propionic acid
4-Quinolinepropanoic acid, α-[(4-chlorobenzoyl)amino]-1,2-dihydro-2-oxo- [ACD/Index Name]
4-quinolinepropanoic acid, α-[(4-chlorobenzoyl)amino]-2-hydroxy-
6454
CAS 90098-04-7 [RN]
a-[(4-Chlorobenzoyl)amino]-1,2-dihydro-2-oxo-4-quinolinepropanoic acid
LR583V32ZR
UNII:LR583V32ZR
ребамипид [Russian] [INN]
ريباميبيد [Arabic] [INN]
瑞巴派特 [Chinese] [INN]
(±)-2-(4-CHLOROBENZOYLAMINO)-3-(2(1H)-QUINOLINON-4-YL)-PROPIONIC ACID
obtain the white powder from dimethylformamide-water with its hemihydrate m.p. being 288-290°C (decomposition).
(-)-Configuration: from dimethylformamide to give colorless needles, mp 305~306 °C (decomposition). [α] D20-116.7 ° (C = 1.0, dimethylformamide).
(+)-Configuration: from dimethylformamide to give colorless needles, mp 305~306 °C (decomposition). [α] D20 + 116.9 ° (C = 1.0, dimethylformamide).
Rebamipide is a quinolone derivative that was launched in 1990 by Otsuka in Japan for the oral treatment of Helicobacter pylori-induced gastric inflammation after eradication therapy and peptic ulcer
Title: Rebamipide
CAS Registry Number: 90098-04-7
CAS Name: a-[(4-Chlorobenzoyl)amino]-1,2-dihydro-2-oxo-4-quinolinepropanoic acid
Additional Names: (±)-a-(p-chlorobenzamido)-1,2-dihydro-2-oxo-4-quinolinepropionic acid; 2-(4-chlorobenzoylamino)-3-[2(1H)-quinolinon-4-yl]propionic acid; proamipide
Manufacturers’ Codes: OPC-12759
Trademarks: Mucosta (Otsuka)
Molecular Formula: C19H15ClN2O4
Molecular Weight: 370.79
Percent Composition: C 61.55%, H 4.08%, Cl 9.56%, N 7.56%, O 17.26%
Literature References: Gastric cytoprotectant. Prepn: M. Uchida et al., DE 3324034eidem, US 4578381; (1984, 1986 both to Otsuka). Synthesis and pharmacology: M. Uchida et al., Chem. Pharm. Bull. 33, 3775 (1985); of enantiomers: eidem, ibid. 35, 853 (1987). Antiulcer activity in rats: K. Yamasaki et al., Eur. J. Pharmacol. 142, 23 (1987); K. Yamasaki et al., Jpn. J. Pharmacol. 49,441 (1989). HPLC determn in plasma and urine: Y. Shioya, T. Shimizu, J. Chromatogr. 434, 283 (1988).
Properties: White powder from DMF-water, mp 288-290° (dec) as hemihydrate.
Melting point: mp 288-290° (dec) as hemihydrate
Derivative Type: (-)-Form
Properties: Colorless needles from DMF, mp 305-306° (dec). [a]D20 -116.7° (c = 1.0 in DMF).
Melting point: mp 305-306° (dec)
Optical Rotation: [a]D20 -116.7° (c = 1.0 in DMF)
Derivative Type: (+)-Form
Properties: Colorless needles from DMF, mp 305-306° (dec). [a]D20 +116.9° (c = 1.0 in DMF).
Melting point: mp 305-306° (dec)
Optical Rotation: [a]D20 +116.9° (c = 1.0 in DMF)
Therap-Cat: Antiulcerative.
Keywords: Antiulcerative; Cytoprotectant (Gastric).
Rebamipide has been investigated for the treatment of Stomach Ulcer, Keratoconjunctivitis Sicca, and Gastric Adenoma and Early Gastric Cancer.
Rebamipide is a quinolinone derivative that stimulates endogenous PGE2 generation in gastric mucosa, enhancing gastric mucosal defense in a COX-2-dependent manner.
Rebamipide has been shown to inhibit the production of reactive oxygen species and to decrease cytokine release induced by H. pylori infection.
A daily oral dose of 100 mg/kg was found to be protective against the development of pyloric channel ulcers in Mongolian gerbils infected with H. pylori.
In addition to the stomach, rebamipide can also enhance secretion of mucin covering the conjunctiva and cornea, which is important for tear film adhesion.
Rebamipide, a gastroprotective drug, was developed in Japan and was proven to be superior to cetraxate, the former most prescribed drug of the same category, in 1989 in the treatment for gastric ulcers. The initially discovered basic mechanisms of action of rebamipide included its action as a prostaglandin inducer and oxygen free-radical scavenger. In the last 5 years, several basic and clinical studies have been performed for functional dyspepsia, chronic gastritis, NSAID-induced gastrointestinal injuries, gastric ulcer following eradication therapy for Helicobacter pylori, gastric ulcer after endoscopic surgery and ulcerative colitis. In addition, several molecules have been identified as therapeutic targets of rebamipide to explain its pleiotropic pharmacological actions.

Rebamipide, an amino acid derivative of 2-(1H)-quinolinone, is used for mucosal protection, healing of gastroduodenal ulcers, and treatment of gastritis. It works by enhancing mucosal defense, scavenging free radicals, and temporarily activating genes encoding cyclooxygenase-2.

Rebamipide is used in a number of Asian countries including Japan (marketed as Mucosta), South KoreaChina[1] and India (where it is marketed under the trade name Rebagen). It is also approved in Russia under the brand name Rebagit.[2] It is not approved by the Food and Drug Administration for use in the United States.

Studies have shown that rebamipide can fight the damaging effects of NSAIDs on the GIT mucosa, and more recently, the small intestine.[citation needed] It has also been studied for the treatment of Behçet’s disease.[3] It was shown to successfully treat pouchitis in a single-N study after first-line therapies for the condition were unsuccessful.[4] Some studies have shown effectiveness in presbyacusis(age-related hearing loss).[citation needed]

It has also been shown to alleviate signs and symptoms of dry eyes in a randomised controlled trial although this is not yet widely available clinically.[5]

SYN

Rebamipide (CAS NO.: 111911-87-6), with its systematic name of 4-Quinolinepropanic acid, alpha-((4-chlorobenzoyl)amino)-1,2-dihydro-2-oxo-, (+-)-, could be produced through many synthetic methods.

Following is one of the reaction routes:

Synthesis of Rebamipide

4-(Bromomethyl)quinolin-2(1H)-one (I) could react with hot phosphorus oxychloride to produce a mixture of 4-(bromomethyl)-2-chloroquinoline (II) and 2-chloro-4-(chloromethyl)quinoline (III), and then the mixture without separation is  ondensed with 2(S)-isopropyl-3,6-dimethoxy-2,5-dihydropyrazine (IVs) in the presence of butyllithium in hexane, affording (-)-2-chloro-4-[6(S)-isopropyl-2,5-dimethoxy-3,6-dihydropyrazin-3(R)-yl methyl]quinoline (Vr). The hydrolysis of (Vr) with HCl produces 3-(2-chloroquinolin-4-yl)-(R)-alanine methyl ester (VIr), which is treated with HCl and propylene oxide to afford 3-(2-oxo-2,3-dihydroquinolin-4-yl)-(R)-alanine (VIIr). At last, this compound is acylated with 4-chlorobenzoyl chloride (VIII) by means of K2CO3in acetone, affording (R)-OPC-12759.

The synthetic route of Rebamipide
Figure 2 The synthetic route of Rebamipide.

DE 3324034; US 4578381 ABOVE

The condensation of 4-(bromomethyl)quinolin-2(1H)-one (I) with diethyl acetamidomalonate (II) by means of sodium ethoxide in refluxing ethanol gives ethyl 2-acetamido-2-(ethoxycarbonyl)-3-(2-oxo-1,2-dihydroquinolin-4yl)propionate (III), which is submitted to a decarboxylative hydrolysis with refluxing 20% HCl yielding 3-(2-oxo-1,2-dihydroquinolin-4yl)alanine (IV). Finaily this compound is acylated with 4-chlorobenzoyl chloride by means of K2CO3 in acetone water.

SYN

Chem Pharm Bull 1991,39(11),2906 ABOVE

The synthesis of (R)- and (S)-isomers of OPC-12759 has been described: These optical isomers can be obtained in three different ways: 1) The reaction of 4-(bromomethyl)quinolin-2(1H)-one (I) with hot phosphorus oxychloride gives a mixture of 4-(bromomethyl)-2-chloroquinoline (II) and 2-chloro-4-(chloromethyl)quinoline (III), which, without separation, is condensed with 2(S)-isopropyl-3,6-dimethoxy-2,5-dihydropyrazine (IVs) by means of butyllithium in hexane, yielding (-)-2-chloro-4-[6(S)-isopropyl-2,5-dimethoxy-3,6-dihydropyrazin-3(R)-yl methyl]quinoline (Vr). The hydrolysis of (Vr) with HCl affords 3-(2-chloroquinolin-4-yl)-(R)-alanine methyl ester (VIr), which is treated with HCl and propylene oxide to give 3-(2-oxo-2,3-dihydroquinolin-4-yl)-(R)-alanine (VIIr). Finally, this compound is acylated with 4-chlorobenzoyl chloride (VIII) by means of K2CO3 in acetone, affording (R)-OPC-12759.

SYN

3) The methylation of 3-(2-oxo-1,2-dihydroquinolin-4-yl)-(R,S)-alanine (IX) with SOCl2 and methanol yields the corresponding methyl ester (X), which is submitted to optical resolution with D-(-)-mandelic acid, affording adducts (XII) and (XIII). The hydrolytic treatment of (XII) and (XIII) with HCl and propylene oxide finally yields isomers (VIIr) and (VIIs), already obtained. Racemic OPC-12759 can also be resolved into its optical isomers by treatment with brucine and fractionated crystallization.

Rebamipide

    • Synonyms:Proamipide
    • ATC:A02BX
  • Use:ulcer therapeutic
  • Chemical name:α-[(4-chlorobenzoyl)amino]-1,2-dihydro-2-oxo-4-quinolinepropanoic acid
  • Formula:C19H15ClN2O4
  • MW:370.79 g/mol
  • CAS-RN:90098-04-7
  • LD50:572 mg/kg (M, i.v.);
    700 mg/kg (R, i.v.);
    >2 g/kg (dog, p.o.)

Substance Classes

Synthesis Path

Substances Referenced in Synthesis Path

CAS-RN Formula Chemical Name CAS Index Name
39098-85-6 C4H5ClO2 acetoacetyl chloride Butanoyl chloride, 3-oxo-
62-53-3 C6H7N aniline Benzenamine
4876-10-2 C10H8BrNO 4-(bromomethyl)-2(1H)-quinolinone 2(1H)-Quinolinone, 4-(bromomethyl)-
128-08-5 C4H4BrNO2 N-bromosuccinimide 2,5-Pyrrolidinedione, 1-bromo-
122-01-0 C7H4Cl2O 4-chlorobenzoyl chloride Benzoyl chloride, 4-chloro-
1068-90-2 C9H15NO5 diethyl acetamidomalonate Propanedioic acid, (acetylamino)-, diethyl ester
4900-38-3 C19H22N2O6 ethyl 2-acetamido-2-(ethoxycarbonyl)-3-(2-oxo-1,2-dihydroquinolin-4-yl)propionate Propanedioic acid, (acetylamino)[(1,2-dihydro-2-oxo-4-quinolinyl)methyl]-, diethyl ester
5162-90-3 C12H12N2O3 3-(2-oxo-1,2-dihydroquinolin-4-yl)alanine 4-Quinolinepropanoic acid, α-amino-1,2-dihydro-2-oxo-
102-01-2 C10H11NO2 3-oxo-N-phenylbutanamide Butanamide, 3-oxo-N-phenyl-

Trade Names

Country Trade Name Vendor Annotation
J Mucosta Otsuka

Formulations

  • tabl. 100 mg

References

    • Uchida, M. et al.: Chem. Pharm. Bull. (CPBTAL) 33, 3775 (1985).
    • DOS 3 324 034 (Otsuka; appl. 7.4.1983; J-prior. 7.5.1982).
    • GB 2 123 825 (Otsuka; appl. 7.5.1983; J-prior. 7.5.1982).
  • oral and parenteral formulations:

    • JP 60 019 767 (Otsuka; appl. 7.11.1983).

PAPER

Magic Bullet! Rebamipide, a Superior Anti-ulcer and Ophthalmic Drug and Its Large-Scale Synthesis in a Single Organic Solvent via Process Intensification Using Krapcho Decarboxylation

https://pubs.acs.org/doi/10.1021/acs.oprd.7b00382#

Chemical Research Division, API R&D CentreMicro Labs Ltd.Plot No.43-45, KIADB Industrial Area, fourth phase, Bommasandra-Jigani Link Road, Bommasandra, Bangalore 560 105, Karnataka, India
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00382
Publication Date (Web): May 31, 2018
Copyright © 2018 American Chemical Society
Abstract Image

Rebamipide (1) is a superior drug compared to existing drugs for use in healing of peptic ulcers, gastrointestinal bleeding, and dyspepsia. It is also useful as an ophthalmic drug for the treatment of dry eye syndrome. Process intensification for its synthesis was achieved by (i) averting uncontrollable frothing using Krapcho decarboxylation instead of conventional acid hydrolysis, where uncontrollable frothing became chaotic, (ii) minimizing organic waste generation by using a single organic solvent, and (iii) avoiding anti-foaming agents (n-octanol, acetophenone) and acetic acid. With these trifling modifications, the overall yield of active pharmaceutical ingredient (API) was ≥83% with excellent purity (≥99.89%), and the process meets the metrics of “green” chemistry with an E-factor = 11.5. The developed hassle-free commercial process is viable for multi-kilogram synthesis of Rebamipide (1) as the key step, Krapcho decarboxylation is safe to run at 130–140 °C in DMSO, and it was proved to be effective by differential scanning calorimetry thermal screening studies. The characterization data of intermediates, process-related impurities, and API are reported. The carryover and process-related impurities were controlled efficiently. The present work can enhance the scope and worldwide adoptability of Rebamipide (1), which is currently limited to Asian countries.

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.7b00382/suppl_file/op7b00382_si_001.pdf

STR1 STR2 str3 str4 str5

Articles

  • Arakawa T, Watanabe T, Fukuda T, Yamasaki K, Kobayashi K (1995). “Rebamipide, novel prostaglandin-inducer accelerates healing and reduces relapse of acetic acid-induced rat gastric ulcer. Comparison with cimetidine”. Dig Dis Sci40 (11): 2469–72. doi:10.1007/BF02063257PMID 7587834.
  • Arakawa T, Kobayashi K, Yoshikawa T, Tarnawski A (1998). “Rebamipide: overview of its mechanisms of action and efficacy in mucosal protection and ulcer healing”. Dig Dis Sci43 (9 Suppl): 5S–13S. PMID 9753220.
  • Tarnawski AS, Chai J, Pai R, Chiou SK (2004). “Rebamipide activates genes encoding angiogenic growth factors and Cox2 and stimulates angiogenesis: a key to its ulcer healing action?”. Dig Dis Sci49 (2): 202–9. doi:10.1023/B:DDAS.0000017439.60943.5cPMID 15104358.
  • Takumida M, Anniko M (2009). “Radical scavengers for elderly patients with age-related hearing loss”. Acta Otolaryngol129 (1): 36–44. doi:10.1080/00016480802008215PMID 18607930.

References

  1. Jump up^ drugs.com
  2. Jump up^ “Russian State Register of Medicines. Registration Sertificate: Rebagit (rebamipide) Film-Coated Tablets” (in Russian). Retrieved 10 June 2017.
  3. Jump up^ Matsuda T, Ohno S, Hirohata S, Miyanaga Y, Ujihara H, Inaba G, Nakamura S, Tanaka S, Kogure M, Mizushima Y (2003). “Efficacy of rebamipide as adjunctive therapy in the treatment of recurrent oral aphthous ulcers in patients with Behcet’s disease: a randomised, double-blind, placebo-controlled study”. Drugs R D4 (1): 19–28. doi:10.2165/00126839-200304010-00002PMID 12568631.
  4. Jump up^ http://www.wjgnet.com/1007-9327/12/656.pdf Archived October 20, 2013, at the Wayback Machine.
  5. Jump up^ Kinoshita, S.; K. Oshiden; S. Awamura; H. Suzuki; N. Nakamichi (2013). “A randomized, multicenter phase 3 study comparing 2% rebamipide (OPC-12759) with 0.1% sodium hyaluronate in the treatment of dry eye”. Ophthalmology120 (6): 1158–65. doi:10.1016/j.ophtha.2012.12.022PMID 23490326.
Rebamipide
Rebamipide.svg
Clinical data
Trade names Mucosta (JP), Rebagen (KR,CNIN), Rebagit (RU)
AHFS/Drugs.com International Drug Names
Routes of
administration
Oral (tablets)
ATC code
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C19H15ClN2O4
Molar mass 370.786 g/mol
3D model (JSmol)

/////////Rebamipide, UNII:LR583V32ZR, ребамипид ريباميبيد ,瑞巴派特 , OPC-12759  , OPC-12759E  , OPC-759 , OPC 12759  , OPC 12759E  , OPC 759 , OTSUKA, JAPAN 1990

OC(=O)C(CC1=CC(O)=NC2=CC=CC=C12)NC(=O)C1=CC=C(Cl)C=C1

New ICH Guidelines: ICH Q13 on Conti Manufacturing and ICH Q14 on AQbD

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

ICH

New ICH Guidelines:

*ICH Q13* on Continuous Manufacturing &
🎛🎚

*ICH Q14* on ATP – QbD (Analytical target profile and quality by design)
⚗⏱

New ICH Guidelines: ICH Q13 on Conti Manufacturing and ICH Q14 on AQbD

In a press release from 22 June the International Council for Harmonisation (ICH) has announced that they will prepare new topics for the future. The Assembly agreed to begin working on two new topics for ICH harmonisation:

Analytical Procedure Development and Revision of Q2(R1) Analytical Validation (Q2(R2)/Q14)
and
Continuous Manufacturing (Q13)

The long anticipated revision of ICH Q2(R1) “Guideline on Validation of Analytical Procedures: Text and Methodology” has been approved and the work plan is scheduled to commence in Q3 2018. It is intended that the new guidelines will be consistent with ICH Q8(R2), Q9, Q10, Q11 and Q12 .

The AQbD approach is very important to collect information in order…

View original post 154 more words

Viewing all 2871 articles
Browse latest View live


<script src="https://jsc.adskeeper.com/r/s/rssing.com.1596347.js" async> </script>