US20080033175A1 - Process for preparation of 4-amino-3-quinolinecarbonitriles - Google Patents

Process for preparation of 4-amino-3-quinolinecarbonitriles Download PDF

Info

Publication number
US20080033175A1
US20080033175A1 US11/973,259 US97325907A US2008033175A1 US 20080033175 A1 US20080033175 A1 US 20080033175A1 US 97325907 A US97325907 A US 97325907A US 2008033175 A1 US2008033175 A1 US 2008033175A1
Authority
US
United States
Prior art keywords
amino
yield
mixture
suspension
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/973,259
Inventor
Karen Sutherland
Gregg Feigelson
Diane Boschelli
David Blum
Henry Strong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wyeth Holdings LLC
Original Assignee
Wyeth Holdings LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34215972&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080033175(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Wyeth Holdings LLC filed Critical Wyeth Holdings LLC
Priority to US11/973,259 priority Critical patent/US20080033175A1/en
Publication of US20080033175A1 publication Critical patent/US20080033175A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J31/00Catalysts comprising hydrides, coordination complexes or organic compounds
    • B01J31/02Catalysts comprising hydrides, coordination complexes or organic compounds containing organic compounds or metal hydrides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • C07D215/54Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • This invention relates to the manufacture of 4-amino-3-quinolinecarbonitriles, which can serve as intermediates for the synthesis of additional 4-amino-3-quinolinecarbonitrile analogues.
  • Such substituted quinolines, as well as the pharmaceutically acceptable salts thereof, inhibit the action of certain protein kinases (PK) thereby inhibiting the abnormal growth of certain cell types.
  • PK protein kinases
  • the compounds derived from this invention are for example, useful in the treatment of polycystic kidney disease, colonic polyps, cancer, and stroke in mammals.
  • This invention also relates to the manufacture of 7-amino-thieno[3,2-b]pyridine-6-carbonitriles useful as intermediates in the synthesis of 7-amino-thieno[3,2-b]pyridine-6-carbonitrile analogues.
  • the thieno[3,2-b]pyridine ring system will be numbered as indicated in the formula below:
  • the approach described herein is favorable due to the relative ease of preparation of the cyanoacetamide portion from cyanoacetic acid and the desired aniline and suitably substituted aniline that results in the carbocyclic ring.
  • 3-chloroaniline is reacted via a variety of cyanoacetamides to form enaminonitriles, which undergo cyclization to 4-aminoquinolines in some cases.
  • the conditions used to prepare the enaminonitrile are much harsher than those outlined herein and cyclization conditions in some examples do not result in cyclodehydration [Price, C. C., Boekelheide, V. (1946) J. Am. Chem. Soc., 68, 1246].
  • the compounds described in this invention contain significantly different substitution patterns.
  • the present invention relates to a process for the preparation of a 4-amino-3-quinolinecarbonitrile comprising combining an amine compound with cyanoacetic acid and an acid catalyst to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline in an alcoholic solvent and a trialkylorthoformate to yield a 3-amino-2-cyanoacrylamide, and then combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 4-amino-3-quinolinecarbonitrile.
  • the present invention also relates to a process for the preparation of a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile comprising combining an optionally up to disubstituted 3-amino thiophene with a cyanoacetamide and trialkylorthoformate in an alcoholic solvent to obtain a 3-amino-2-cyanoacrylamide and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile.
  • the present invention relates to a process for the preparation of a 4 amino-3-quinolinecarbonitrile by combining an amine compound with cyanoacetic acid and a peptide coupling reagent to obtain a solution, filtering the solution to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline, an alcoholic solvent, and a trialkylorthoformate to yield a 3-amino-2-cyanoacrylamide; and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride to yield a 4-amino-3-quinolinecarbonitrile.
  • the present invention includes a process for the preparation of a cyanoacetamide comprising combining dimethylformamide (DMF), an amine and cyanoacetic acid to yield a mixture; cooling the mixture; adding a solution of N,N′ dicyclohexylcarbodiimide in DMF so as to keep the temperature below 15° C. to yield a suspension; filtering the suspension and washing the resulting solid by-product to yield a filtrate; adding water to the filtrate to obtain a mixture; and filtering the mixture to yield a cyanoacetamide
  • This invention provides a high yielding, high purity, operationally simple process for the production of 4-amino-3-quinolinecarbonitriles optionally with substituents in 5, 6, 7, and 8 positions, comprising:
  • R 1 and R 2 are each independently hydrogen, alkyl of 1-6 carbon atoms, substituted and unsubstituted aryl;
  • R 3 , R 4 , R 5 and R 6 are each independently hydrogen, hydroxy, halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, alkoxy of 1-6 carbon atoms, -alkoxyalkoxy-, haloalkoxy-, -alkylheteroaryl, optionally substituted aryl, N-alkylpiperazino wherein the alkyl moiety is of 1-6 carbon atoms, pyrrolidino, morpholino, piperazino, -alkoxy[(N)-alkylpiperazino] wherein the alkyl and alkoxy moiety are of 1-6 carbon atoms, or optionally substituted heteroaryl.
  • the cyanoacetamide 1 can be prepared from the corresponding amine and cyanoacetic acid in refluxing toluene with an acid catalyst or in tetrahydrofuran (THF) with a peptide coupling reagent.
  • THF tetrahydrofuran
  • the examples contained herein provide preferred conditions and include use of 1.03 equivalents of cyanoacetic acid relative to amine and 1,3-diisopropylcarbodiimide in THF at 77-80° C.
  • the optimum procedure allows for filtration of the suspension then dilution with water to allow the product to precipitate and collecting by filtration. This procedure provides a near quantitative yield 1 of sufficient purity to be used in the subsequent step without further purification.
  • the condensation reaction of the cyanoacetamide with optionally up to tetra-substituted anilines can be performed with the hydrochloride salt or free base of the aniline with trialkylorthoformate using alcoholic solvents at 20-140° C.
  • the optimum conditions for this transformation result from conducting the reaction in iso-propanol at 80° C. with the hydrochloride salt or free base of the aniline and 2.0-7.0 equivalents of triethylorthoformate.
  • the cyclo-dehydration is accomplished with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene optionally with alcohols or amine bases as catalysts at 80-110° C.
  • the transformation may be done by heating the substrate in acetonitrile or butyronitrile with methanol and phosphorus oxychloride.
  • the product salt precipitates from the reaction and can be neutralized directly or isolated and neutralized in a separate vessel to produce 3 in good yield and high quality. Addition of base to the reaction, such as pyridine, triethylamine, or diisopropylamine can provide a significant rate acceleration.
  • R 1 and R 2 are each independently hydrogen, alkyl of 1-6 carbon atoms substituted and unsubstituted aryl;
  • R 3 and R 4 are each independently hydrogen, halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, -alkylheteroaryl, aryl, or heteroaryl.
  • the condensation reaction of the cyanoacetamide 1 with 3-aminothiophene can be performed with the hydrochloride salt or free base of 3-aminothiophene with trialkylorthoformate using alcoholic solvents at 20-140° C.
  • the optimum conditions for this transformation result from conducting the reaction in iso-propanol at 80° C. with 3-aminothiophene and 2.0-7.0 equivalents of triethylorthoformate. These conditions allow for precipitation of the product 4 isolated after filtration in good yield and high purity and can be used in the subsequent step without purification.
  • the cyclo-dehydration is accomplished with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene optionally with alcohols or amine bases as catalyst at 80-110° C.
  • the optimum procedure for this transformation is by heating the substrate in acetonitrile with phosphorus oxychloride.
  • the product salt precipitates from the reaction and can be neutralized directly or isolated and neutralized in a separate vessel to produce 7-amino-thieno[3,2-b]pyridine-6-carbonitriles of formula 5 in good yield and high quality.
  • the described route 1 is shown using the following examples 4, 6, 42, 43, 44 and 45.
  • the described route 2 is shown using the following examples 4, 46, 47, 48, 49, and 50.
  • the cyanoacetic acid is used in an amount of 1-1.5 equivalents relative to the amine, with a preferred concentration of 1.03 equivalents.
  • the optionally up to tetra-substituted aniline is a hydrochloride salt. In another embodiment the optionally up to tetra-substituted aniline is a free base.
  • alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl(alicyclic)groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has 6 or fewer carbon atoms in its backbone.
  • alkyl can be used alone or as part of a chemical name as in for example, “trialkylorthoformate”.
  • alkenyl and alkynyl refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one double or triple carbon-carbon bond, respectively.
  • alkoxy refers to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • alkoxy can be used alone or as part of a chemical name as in for example, “alkoxy-enaminonitrile”.
  • Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, alkenylamines, alkynylamines, alkenylamides, alkynylamides, alkenylimines, alkynylimines, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls, alkenoxyls, alkynoxyls, metalloalkenyls and metalloalkynyls.
  • aryl as used herein includes 4-, 5-, 6-, 7- and 10-membered carbocyclic single ring or fused multiple ring aromatic groups, which may be substituted or unsubstituted.
  • heteroaryl refers to a 4 to 10 aromatic membered ring structure, which ring structure includes one to four heteroatoms.
  • Heteroaryls include, but are not limited to, pyrrolidine, oxolane, thiolane, piperidine, piperazine, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and morpholine.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur, phosphorus, and selenium.
  • halogen refers to an atom of fluorine, chlorine, bromine, or iodine.
  • the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents of organic compounds include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • the acid and base catalysts of this invention include a substance that increases the rate of a reaction without modifying the overall standard Gibbs energy change in the reaction.
  • the acid and base catalysts include for example, pyridine and 3-nitrophenylboronic acid.
  • the peptide coupling reagent includes for example, 1,3-diisopropylcarbodiimide, 1,3-[(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride, and dicyclohexylcarbodiimide (DCC).
  • DCC dicyclohexylcarbodiimide
  • the alcoholic solvent is the liquid or homogeneous mixture of liquids in which extractant(s) and possible modifier(s) may be dissolved to form the solvent phase.
  • the alcoholic solvent is for example ethylene glycol, methanol, iso-propanol, or butanol.
  • the alcoholic solvent is iso-propanol.
  • the condensing step of the present invention occurs at a temperature of 10-200° C. In a more preferred embodiment the temperature is 140° C. In another preferred embodiment the temperature is 80° C.
  • the catalyst is an alcohol. In another embodiment the catalyst is an amine base.
  • the peptide coupling reagent is water soluble.
  • a reaction flask was charged with dimethylformamide (DMF) (500 mL), 2,4-dichloro-5-methoxyaniline (100 g, 0.52 mol) and cyanoacetic acid (46.6 g, 0.55 mol). The mixture was cooled to 10° C. in an ice bath. To the cooled mixture was added, dropwise, a solution of N,N′ dicyclohexylcarbodiimide (119.1 g, 0.58 mol) in DMF (240 mL) so as to keep the temperature below 15° C. After the addition was completed, cooling was discontinued and the reaction was stirred for 2 hours. The urea by-product was then removed via filtration and the cake was washed twice with DMF.
  • DMF dimethylformamide
  • 2,4-dichloro-5-methoxyaniline 100 g, 0.52 mol
  • cyanoacetic acid 46.6 g, 0.55 mol
  • 3,4-Dimethoxyaniline (1.50 g, 0.010 mol), dimethylaminocyanoacetamide (1.50 g, 0.013 mol) and triethylorthoformate (2 mL, 0.012 mol) were mixed in ethylene glycol (10 mL) and the solution heated to reflux. The solution was stirred for 5 hours then allowed to cool to room temperature when precipitate was formed. Water (120 mL) was added and the suspension filtered.
  • the filter cake was washed with acetonitrile (3 ⁇ 0.63 kg) and the filter cake dried on the filter funnel for 2 hours.
  • a 50-L round-bottomed flask equipped with overhead stirrer, thermocouple, condenser and 1000 mL dropping funnel was charged with the filter cake from above.
  • THF (4.44 kg) was added and the suspension warmed to 35-40° C.
  • Ammonium hydroxide (0.63 kg) and water (2 kg) were mixed in a flask and transferred to the dropping funnel.
  • the ammonium hydroxide solution was added to the THF suspension over 30 minutes maintaining the temperature below 55° C. Water (10 kg) was added over 25 minutes then the resulting suspension was cooled to 15-20° C.
  • a 12-L round-bottomed flask equipped with overhead stirrer, thermocouple, condenser and 1000 mL dropping funnel was charged with the filter cake from above.
  • THF (2.22 kg) was added and the suspension warmed to 35-40° C. 28%
  • Ammonium hydroxide (0.63 kg) and water (1 kg) were mixed in a flask and transferred to the dropping funnel.
  • the ammonium hydroxide solution was added to the THF suspension over 30 minutes maintaining the temperature below 55° C.
  • Water (5 kg) was added over 25 minutes then the resulting suspension was cooled to 15-20° C.
  • reaction mixture was cooled in an ice-bath and the solid was collected by filtration, washed with cold acetonitrile (20 mL) and then suspended in tetrahydrofuran (50 mL). To both the acetonitrile filtrate and the tetrahydrofuran suspension were added concentrated ammonium hydroxide (2 ⁇ 25 mL) and the mixtures were stirred for 1 hour. Water (2 ⁇ 400 mL) was added and stirring was continued for 2 hours.
  • the mixture was filtered and rinsed and the salts collected with ethyl acetate.
  • the filtrate was washed once with 400 mL of 0.5 N NaOH.
  • the layers were separated and the organic layer was washed with water (3 ⁇ 390 mL).
  • the combined organic layers were concentrated under vacuum at 35-40° C.
  • the quench was very exothermic; the addition of sodium hydroxide was such that the temperature of the mixture was kept below 55° C.
  • the mixture was basified to pH 9-10. After cooling to ⁇ 30° C., 500 mL of ethyl acetate was added and the resulting mixture was stirred for 15 minutes. The solids were filtered and then rinsed with ethyl acetate. The filtrate layers were separated and the organic layer was concentrated in vacuo.
  • the crude oil was dissolved in 45 mL of methanol and stirred at room temperature for 1 hour. The crystalline solid that formed was filtered and rinsed with minimal cold methanol. The filtrate was concentrated and the procedure repeated with about half as much methanol.
  • the resultant intermediate boronic acid was dissolved in 20 mL of ethylene glycol dimethyl ether and 2-iodo-4-nitroanisole (1.06 mg, 4.25 mmol) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (1:1) (87 mg, 0.11 mol) were added.
  • a solution of 2.25 g of sodium carbonate in 6.5 mL of water was added and the resulting mixture was heated at reflux for 3 hours. The mixture was cooled to room temperature and partitioned between ethyl acetate and water.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Materials Engineering (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Quinoline Compounds (AREA)

Abstract

This invention discloses a process for the preparation of a 4-amino-3-quinolinecarbonitrile comprising combining an amine compound with a cyanoacetic acid and an acid catalyst to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline in an alcoholic solvent and a trialkylorthoformate to yield a 3-amino-2-cyanoacrylamide; combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 4-amino-3-quinolinecarbonitrile and also discloses a process for the preparation of a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile comprising combining a disubstituted 3-amino thiophene with a cyanoacetamide and trialkylorthoformate in an alcoholic solvent to obtain a 3-amino-2-cyanoacrylamide; and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride and acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile and also discloses a process for the preparation of a 4-amino-3-quinolinecarbonitrile by combining an amine compound with a cyanoacetic acid and a peptide coupling reagent to obtain a suspension; filtering the suspension to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline, an alcoholic solvent, and triethylorthoformate to yield a 3-amino-2-cyanoacrylamide; and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride to yield a 4-amino-3-quinolinecarbonitrile.

Description

  • This is a divisional of copending application(s) application Ser. No. 10/918,947 filed on Aug. 16, 2004, and application No. 60/496,191 filed on Aug. 19, 2003 the entire disclosure of which is hereby incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • This invention relates to the manufacture of 4-amino-3-quinolinecarbonitriles, which can serve as intermediates for the synthesis of additional 4-amino-3-quinolinecarbonitrile analogues. Such substituted quinolines, as well as the pharmaceutically acceptable salts thereof, inhibit the action of certain protein kinases (PK) thereby inhibiting the abnormal growth of certain cell types.
  • The compounds derived from this invention are for example, useful in the treatment of polycystic kidney disease, colonic polyps, cancer, and stroke in mammals.
  • Throughout this patent application the quinoline ring system will be numbered as indicated in the formula below:
    Figure US20080033175A1-20080207-C00001
  • This invention also relates to the manufacture of 7-amino-thieno[3,2-b]pyridine-6-carbonitriles useful as intermediates in the synthesis of 7-amino-thieno[3,2-b]pyridine-6-carbonitrile analogues. Throughout this patent application the thieno[3,2-b]pyridine ring system will be numbered as indicated in the formula below:
    Figure US20080033175A1-20080207-C00002
  • Historically there are several methods for the preparation of 4-aminosubstituted quinolines and the two most frequent methods involve intramolecular Friedel-Crafts reactions or electrocyclic ring closures of N-(2-carboxyvinyl)-aniline derivatives at elevated temperatures. Cyclodehydration of suitable amide substrates via Vilsmeier type intermediates appears attractive but the literature is scarce and in the example below, the desired amino-quinoline species is unstable under the reaction conditions and the chloro-quinoline is favored [Meth-Cohn, O., Taylor, D. L. (1995) Tetrahedron, 51, 12869].
  • The approach described herein is favorable due to the relative ease of preparation of the cyanoacetamide portion from cyanoacetic acid and the desired aniline and suitably substituted aniline that results in the carbocyclic ring. One example exists in the literature where 3-chloroaniline is reacted via a variety of cyanoacetamides to form enaminonitriles, which undergo cyclization to 4-aminoquinolines in some cases. The conditions used to prepare the enaminonitrile are much harsher than those outlined herein and cyclization conditions in some examples do not result in cyclodehydration [Price, C. C., Boekelheide, V. (1946) J. Am. Chem. Soc., 68, 1246]. In addition, the compounds described in this invention contain significantly different substitution patterns.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention relates to a process for the preparation of a 4-amino-3-quinolinecarbonitrile comprising combining an amine compound with cyanoacetic acid and an acid catalyst to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline in an alcoholic solvent and a trialkylorthoformate to yield a 3-amino-2-cyanoacrylamide, and then combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 4-amino-3-quinolinecarbonitrile.
  • The present invention also relates to a process for the preparation of a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile comprising combining an optionally up to disubstituted 3-amino thiophene with a cyanoacetamide and trialkylorthoformate in an alcoholic solvent to obtain a 3-amino-2-cyanoacrylamide and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile.
  • In addition, the present invention relates to a process for the preparation of a 4 amino-3-quinolinecarbonitrile by combining an amine compound with cyanoacetic acid and a peptide coupling reagent to obtain a solution, filtering the solution to yield a cyanoacetamide; condensing the cyanoacetamide with an optionally up to tetra-substituted aniline, an alcoholic solvent, and a trialkylorthoformate to yield a 3-amino-2-cyanoacrylamide; and combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride to yield a 4-amino-3-quinolinecarbonitrile.
  • The present invention includes a process for the preparation of a cyanoacetamide comprising combining dimethylformamide (DMF), an amine and cyanoacetic acid to yield a mixture; cooling the mixture; adding a solution of N,N′ dicyclohexylcarbodiimide in DMF so as to keep the temperature below 15° C. to yield a suspension; filtering the suspension and washing the resulting solid by-product to yield a filtrate; adding water to the filtrate to obtain a mixture; and filtering the mixture to yield a cyanoacetamide
  • The following experimental details are set forth to aid in an understanding of the invention, and are not intended, and should not be construed, to limit in any way the invention set forth in the claims that follow thereafter.
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention provides a high yielding, high purity, operationally simple process for the production of 4-amino-3-quinolinecarbonitriles optionally with substituents in 5, 6, 7, and 8 positions, comprising:
      • (a) preparation of a cyanoacetamide;
      • (b) preparation of a 3-amino-2-cyanoacrylamide of optionally substituted anilines using a cyanoacetamide and a trialkylorthoformate; and
      • (c) cyclodehydration of the 3-amino-2-cyanoacrylamide prepared as above with phosphorus oxychloride in a suitable solvent with or without additional tertiary amine bases or alcohols.
  • The methodology described herein constructs a functionalized aniline, containing substituents that permit intramolecular cyclization under dehydrating conditions.
  • The sequence illustrated below describes the invention claimed herein.
    Figure US20080033175A1-20080207-C00003
  • wherein:
  • R1 and R2 are each independently hydrogen, alkyl of 1-6 carbon atoms, substituted and unsubstituted aryl; and
  • R3, R4, R5 and R6 are each independently hydrogen, hydroxy, halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, alkoxy of 1-6 carbon atoms, -alkoxyalkoxy-, haloalkoxy-, -alkylheteroaryl, optionally substituted aryl, N-alkylpiperazino wherein the alkyl moiety is of 1-6 carbon atoms, pyrrolidino, morpholino, piperazino, -alkoxy[(N)-alkylpiperazino] wherein the alkyl and alkoxy moiety are of 1-6 carbon atoms, or optionally substituted heteroaryl.
  • The cyanoacetamide 1 can be prepared from the corresponding amine and cyanoacetic acid in refluxing toluene with an acid catalyst or in tetrahydrofuran (THF) with a peptide coupling reagent. The examples contained herein provide preferred conditions and include use of 1.03 equivalents of cyanoacetic acid relative to amine and 1,3-diisopropylcarbodiimide in THF at 77-80° C. The optimum procedure allows for filtration of the suspension then dilution with water to allow the product to precipitate and collecting by filtration. This procedure provides a near quantitative yield 1 of sufficient purity to be used in the subsequent step without further purification.
  • Use of a water-soluble peptide coupling reagent in THF permits direct isolation of the cyanoacetamide on treatment with water.
  • The condensation reaction of the cyanoacetamide with optionally up to tetra-substituted anilines can be performed with the hydrochloride salt or free base of the aniline with trialkylorthoformate using alcoholic solvents at 20-140° C. The optimum conditions for this transformation result from conducting the reaction in iso-propanol at 80° C. with the hydrochloride salt or free base of the aniline and 2.0-7.0 equivalents of triethylorthoformate. These conditions allow for precipitation of the product 2 that is isolated after filtration in good yield and high purity and can be used in the subsequent step without purification.
  • The cyclo-dehydration is accomplished with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene optionally with alcohols or amine bases as catalysts at 80-110° C. The transformation may be done by heating the substrate in acetonitrile or butyronitrile with methanol and phosphorus oxychloride. The product salt precipitates from the reaction and can be neutralized directly or isolated and neutralized in a separate vessel to produce 3 in good yield and high quality. Addition of base to the reaction, such as pyridine, triethylamine, or diisopropylamine can provide a significant rate acceleration.
  • In accordance with this invention a high yielding and operationally simple process for the production 7-amino-thieno[3,2-b]pyridine-6-carbonitriles in high purity is provided by a process, which comprises the sequence illustrated below:
    Figure US20080033175A1-20080207-C00004

    wherein:
  • R1 and R2 are each independently hydrogen, alkyl of 1-6 carbon atoms substituted and unsubstituted aryl; and
  • R3 and R4 are each independently hydrogen, halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, -alkylheteroaryl, aryl, or heteroaryl.
  • The condensation reaction of the cyanoacetamide 1 with 3-aminothiophene can be performed with the hydrochloride salt or free base of 3-aminothiophene with trialkylorthoformate using alcoholic solvents at 20-140° C. The optimum conditions for this transformation result from conducting the reaction in iso-propanol at 80° C. with 3-aminothiophene and 2.0-7.0 equivalents of triethylorthoformate. These conditions allow for precipitation of the product 4 isolated after filtration in good yield and high purity and can be used in the subsequent step without purification.
  • The cyclo-dehydration is accomplished with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene optionally with alcohols or amine bases as catalyst at 80-110° C. The optimum procedure for this transformation is by heating the substrate in acetonitrile with phosphorus oxychloride. The product salt precipitates from the reaction and can be neutralized directly or isolated and neutralized in a separate vessel to produce 7-amino-thieno[3,2-b]pyridine-6-carbonitriles of formula 5 in good yield and high quality.
  • The described route 1 is shown using the following examples 4, 6, 42, 43, 44 and 45.
  • The described route 2 is shown using the following examples 4, 46, 47, 48, 49, and 50.
    Figure US20080033175A1-20080207-C00005
    Figure US20080033175A1-20080207-C00006
  • In certain embodiments of the invention the cyanoacetic acid is used in an amount of 1-1.5 equivalents relative to the amine, with a preferred concentration of 1.03 equivalents.
  • In certain embodiments of the invention the optionally up to tetra-substituted aniline is a hydrochloride salt. In another embodiment the optionally up to tetra-substituted aniline is a free base.
  • DEFINITIONS
  • For convenience, certain terms employed in the specification, examples, and appended claims are collected here.
  • The term “alkyl” refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl(alicyclic)groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In a preferred embodiment, a straight chain or branched chain alkyl has 6 or fewer carbon atoms in its backbone. The term “alkyl” can be used alone or as part of a chemical name as in for example, “trialkylorthoformate”.
  • The terms “alkenyl” and “alkynyl” refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one double or triple carbon-carbon bond, respectively.
  • The term “alkoxy” as used herein refers to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. The term alkoxy can be used alone or as part of a chemical name as in for example, “alkoxy-enaminonitrile”.
  • Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, alkenylamines, alkynylamines, alkenylamides, alkynylamides, alkenylimines, alkynylimines, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls, alkenoxyls, alkynoxyls, metalloalkenyls and metalloalkynyls.
  • The term “aryl” as used herein includes 4-, 5-, 6-, 7- and 10-membered carbocyclic single ring or fused multiple ring aromatic groups, which may be substituted or unsubstituted.
  • The term “heteroaryl” refers to a 4 to 10 aromatic membered ring structure, which ring structure includes one to four heteroatoms. Heteroaryls include, but are not limited to, pyrrolidine, oxolane, thiolane, piperidine, piperazine, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and morpholine.
  • The term “heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur, phosphorus, and selenium.
  • The term “halogen” refers to an atom of fluorine, chlorine, bromine, or iodine.
  • As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents of organic compounds include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • As used herein the acid and base catalysts of this invention include a substance that increases the rate of a reaction without modifying the overall standard Gibbs energy change in the reaction. In a preferred embodiment the acid and base catalysts include for example, pyridine and 3-nitrophenylboronic acid.
  • As used herein the peptide coupling reagent includes for example, 1,3-diisopropylcarbodiimide, 1,3-[(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride, and dicyclohexylcarbodiimide (DCC).
  • As used herein the alcoholic solvent is the liquid or homogeneous mixture of liquids in which extractant(s) and possible modifier(s) may be dissolved to form the solvent phase. In an embodiment the alcoholic solvent is for example ethylene glycol, methanol, iso-propanol, or butanol. In a preferred embodiment the alcoholic solvent is iso-propanol.
  • As used herein the condensing step of the present invention occurs at a temperature of 10-200° C. In a more preferred embodiment the temperature is 140° C. In another preferred embodiment the temperature is 80° C.
  • In a certain embodiment of this invention the catalyst is an alcohol. In another embodiment the catalyst is an amine base.
  • In an embodiment of the invention the peptide coupling reagent is water soluble.
  • EXAMPLE 1 (3-Chloro-4-fluoro)-2-cyanoacetamide
  • A 5-L round-bottomed flask under N2 equipped with an overhead stirrer, a condenser, thermocouple and 500 mL dropping funnel was charged with cyanoacetic acid (150 g, 1.77 mol), 3-chloro-4-fluoroaniline (250 g, 1.72 mol) and tetrahydrofuran (THF) (750 mL). The mixture was heated to 75° C. A solution of 1,3-diisopropylcarbodiimide (221.6 g, 275 mL, 1.77 mol) in THF (50 mL) was added dropwise over 25 minutes at 75-79° C. At the end of the addition diisopropylurea precipitated from solution. The suspension was stirred for an hour then cooled to 13° C. The suspension was filtered and the filter cake washed with THF (2×250 mL). The filtrate was transferred to a 2 L dropping funnel and the solids discarded.
  • A 12-L round-bottomed flask equipped with overhead stirrer and thermocouple was charged with water (4250 mL) at 15° C. The filtrate from above was added slowly to the water over 25 minutes forming a white suspension. The suspension was stirred at room temperature overnight.
  • The suspension was filtered and the filter cake washed with water (2×250 mL) and dried under vacuum (50 mm Hg) at 45° C. to provide (3-chloro-4-fluoro)-2-cyanoacetamide as a white solid (294.5 g, 81% yield, >99% purity by HPLC, mp 155.7-157° C.). 1H NMR (300 MHz, DMSO-d6): 10.4 (s, 1H), 7.80 (d, J=8 Hz, 1H), 7.5-7.3 (m, 2H), 3.92 (s, 2H).
  • EXAMPLE 2 (3-Chloro-4-fluoro)-2-cyanoacetamide
  • A 500-mL round bottomed flask under N2 equipped with overhead stirrer, a condenser, thermocouple and 100 mL dropping funnel was charged with 1-[3-(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride (21.17 g, 0.11 ml) and THF (140 mL). A solution of cyanoacetic acid (10.03 g, 0.118 mol) and 3-chloro-4-fluoroaniline (15.44 g, 0.106 mol) in THF (60 mL) was added to the dropping funnel and added to the reaction flask at 25-45° C. At the end of the addition the reaction became a clear solution. The solution was stirred at ambient temperature for 1.5 hours then poured slowly into water (500 mL) forming a white suspension. The suspension was stirred at room temperature overnight then filtered and the filter cake washed with water and dried under vacuum (50 mm Hg) at 45° C. to provide (3-chloro-4-fluoro)-2-cyanoacetamide as a white solid (21.27 g, 94% yield, 97.5% by HPLC). 1H NMR (300 MHz, DMSO-d6): 10.4 (s, 1H), 7.80 (d, J=8 Hz, 1H), 7.5-7.3 (m, 2H), 3.92 (s, 2H).
  • EXAMPLE 3 (3-Chloro-4-fluoro)-2-cyanoacetamide
  • A 500-mL round bottomed flask under N2 equipped with overhead stirrer, a Dean-Stark trap and condenser, thermocouple was charged with cyanoacetic acid (28.04 g, 0.33 mol), 3-chloro-4-fluoroaniline (40.0 g, 0.28 mol), 3-nitrophenylboronic acid (2.28 g, 0.014 mol) and toluene (200 mL). The mixture was heated to reflux and water collected in the Dean-Stark trap. After 5.5 hours, thin layer chromatography (TLC) indicated complete consumption of the aniline. The reaction mixture was cooled to room temperature (RT) forming a pink suspension. The solids were isolated by filtration, washed with MTBE (2×200 mL) and dried to afford (3-chloro-4-fluoro)-2-cyanoacetamide as a pale pink solid (46.40 g, 79% yield, 96.7% by GC-MS). 1H NMR (300 MHz, DMSO-d6): 10.4 (s, 1H), 7.80 (d, J=8 Hz, 1H), 7.5-7.3 (m, 2H), 3.92 (s, 2H).
  • EXAMPLE 4 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide
  • 2,4-Dichloro-5-methoxyaniline (5.00 g, 26 mmol) and cyanoacetic acid (2.28 g, 26.8 mmol) were mixed in 50 mL of tetrahydrofuran until a solution formed. This solution was heated to reflux and 1,3-diisopropylcarbodiimide (4.2 mL, 26.8 mmol) was added dropwise. After 30 minutes the mixture was cooled to ˜15° C. in an ice-bath. The solid was collected by filtration and washed with tetrahydrofuran. The filtrate was slowly poured into water and stirred for 30 minutes. The white solid was collected by filtration, washed with water, and was then dissolved in 500 mL of ethyl acetate. The solution was dried over sodium sulfate and concentrated in vacuo to give 5.9 g (88%) of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide as a white solid, mp 180-181° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.84 (s, 3H), 4.02 (s, 2H), 7.58 (s, 1H), 7.66 (s, 1H), 10.00 (s, 1H); MS (ES) m/z 257.0, 259.0 (M−H)−
  • Analysis for C10H8Cl2N2O2
  • Calcd: C, 46.36; H, 3.11; N, 10.81.
  • Found: C, 46.25; H, 3.10; N, 10.85.
  • ALTERNATIVE EXAMPLE 4
  • A reaction flask was charged with dimethylformamide (DMF) (500 mL), 2,4-dichloro-5-methoxyaniline (100 g, 0.52 mol) and cyanoacetic acid (46.6 g, 0.55 mol). The mixture was cooled to 10° C. in an ice bath. To the cooled mixture was added, dropwise, a solution of N,N′ dicyclohexylcarbodiimide (119.1 g, 0.58 mol) in DMF (240 mL) so as to keep the temperature below 15° C. After the addition was completed, cooling was discontinued and the reaction was stirred for 2 hours. The urea by-product was then removed via filtration and the cake was washed twice with DMF. To the filtrate was added 700 mL of water. The solid product emerged from solution. The slurry was cooled to 5° C. and held for at least 30 minutes. The product was collected by filtration and washed with water and then dried in vacuo at 60° C. to give 127.08 g of light tan solid.
  • EXAMPLE 5 2-Cyano-N-(3,4,5-trimethoxyphenyl)acetamide
  • 3,4,5-Trimethoxyaniline (10.00 g, 54.6 mmol) and cyanoacetic acid (4.78 g, 56.19 mmol) were mixed in 100 mL of tetrahydrofuran and heated to reflux. To the resultant solution 1,3-diisopropylcarbodiimide (8.8 mL, 56.29 mmol) was added dropwise. The mixture was cooled in an ice-bath and the solid was collected by filtration washing with tetrahydrofuran. The filtrate was slowly poured into water and stirred for 30 minutes. The white solid was collected by filtration, washed with water and dried to give 2.58 g of 2-cyano-N-(3,4,5-trimethoxyphenyl)acetamide as a white solid, mp 146-147° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.61 (s, 3H), 3.74 (s, 6H), 3.86 (s, 2H), 6.90 (s, 2H), 10.23 (s, 1H); MS (ES) m/z 251.1 (M+H)+
  • Analysis for C12H14N2O4
  • Calcd: C, 57.59; H, 5.64; N, 11.19.
  • Found: C, 57.24; H, 5.67; N, 11.08.
  • EXAMPLE 6 2-(3-Chloropropoxy)-1-methoxy-4-nitrobenzene
  • A mixture of 2-methoxy-5-nitrophenol (16.90 g, 100 mmol), 3-chloropropyl p-toluenesulfonate (29.2 g, 120 mmol) and potassium carbonate (27.0 g, 195 mmol) in 160 mL of N,N-dimethylformamide was heated at 80° C. for 2 hours. The reaction mixture was cooled to room temperature and partitioned between water and ethyl acetate. The organic layer was washed with saturated sodium bicarbonate, dried over magnesium sulfate and filtered. The solution was concentrated in vacuo and recrystallized from hexane and ethyl acetate to give 4.68 g of 2-(3-chloropropoxy)-1-methoxy-4-nitrobenzene as a first crop of off-white crystals. A second crop of 11.10 g was obtained from the mother liquor. The remaining solution was concentrated in vacuo and purified by flash column chromatography, eluting with 4:1 hexane:ethyl acetate, to provide 1.31 g of 2-(3-chloropropoxy)-1-methoxy-4-nitrobenzene as off-white crystals, mp 85-87° C.; 1H NMR (400 MHz, DMSO-d6) δ 2.23 (m, 2H), 3.79 (t, J=6 Hz, 2H), 3.92 (s, 3H), 4.21 (t, J=6 Hz, 2H), 7.19 (d, J=9 Hz, 1H), 7.77 (d, J=2 Hz, 1H), 7.92 (dd, J=9.2 Hz, 1H); MS (ES) m/z 246.1, 248.1 (M+H)+
  • Analysis for C10H12CINO4
  • Calcd: C, 48.89; H, 4.92; N, 5.70.
  • Found: C, 49.09; H, 4.68; N, 5.62.
  • ALTERNATIVE EXAMPLE 6
  • 2-methoxy-5-nitrophenol (50 g, 0.30 mol) was combined with 250 mL isopropyl alcohol. 1N sodium hydroxide solution, 310 mL (0.31 mol), was added to the mixture at such a rate that the temperature remained below 40° C. After the addition was completed, the mixture was stirred for at least 30 minutes. The reaction was then charged with 1-bromo-3-chloropropane (93.1 g, 0.59 mol) in a single portion. The mixture was heated to 75° C. and monitored periodically by high performance liquid chromatography (HPLC). After 8 hours, heating was discontinued and the reaction mixture was allowed to cool to ambient temperature overnight. The product gradually precipitated from the mixture as a light yellow/tan solid. Water, 200 mL, was added in a single portion and the resulting slurry was stirred for at least 30 minutes. The slurry was filtered and washed with water. The damp crude product was slurried in 650 mL methanol and heated to reflux until the solids had dissolved or mostly dissolved. The solution was allowed to cool gradually with stirring to ambient temperature over 3-5 hours or longer. The mixture was then cooled to 10-15° C. and held for at least 30 minutes. The solids were filtered and washed with cold methanol. The product was then dried in vacuo to give 60.3 g of 2-(3-chloropropoxy)-1-methoxy-4-nitrobenzene.
  • EXAMPLE 7 3-(3-Chloropropoxy)-4-methoxyaniline
  • A mixture of 2-(3-chloropropoxy)-1-methoxy-4-nitrobenzene (2.15 g, 8.77 mmol) and tin(II)chloride dihydrate (6.1 g, 27.11 mmol) in 50 mL of ethyl acetate was heated at reflux for 6.5 hours. The reaction mixture was cooled to room temperature and poured into 250 mL of saturated aqueous sodium bicarbonate. After stirring for 40 minutes, additional ethyl acetate was added and the layers were separated. The organic layer was washed with saturated sodium bicarbonate and water then dried over magnesium sulfate and filtered. The solution was concentrated in vacuo and purified by flash column chromatography, eluting with 1:1 hexane:ethyl acetate, to provide 753 mg of 3-(3-chloropropoxy)-4-methoxyaniline as a dark brown oil; 1H NMR (400 MHz, DMSO-d6) δ 2.13 (m, 2H), 3.62 (s, 3H), 3.78 (t, J=6 Hz, 2H), 3.96 (t, J=6 Hz, 2H), 4.77 (br s, 2H), 6.10 (dd, J=8, 2 Hz, 1H), 6.29 (d, J=2 Hz, 1H), 6.66 (d, J=8 Hz, 1H); MS (ES) m/z 216.1, 218.1 (M+H)+.
  • EXAMPLE 8 1-Ethoxy-2-iodo-4-nitrobenzene
  • A suspension of 2-iodo-4-nitrophenol (21 g, 79.2 mmol) [ref: Kometani, T.; Watt, D. S.; Ji, T., Tetrahedron Lett. (1985), 26(17), 2043], ethyl iodide (9 mL, 0.48 mol) and potassium carbonate (40.7 g, 0.3 mol) in 100 mL of N,N-dimethylformamide was heated at 70° C. for 3 hours. The reaction was cooled to room temperature and ethyl acetate was added. The inorganic salts were filtered and washed with ethyl acetate. The organic material was washed with water (3×) and brine, dried over magnesium sulfate and filtered. Upon concentration of the filtrate a solid appeared. This solid was filtered and washed with hexanes to give 5.2 g of 1-ethoxy-2-iodo-4-nitrobenzene as white crystals. Concentration of the filtrate provided an additional 11.3 g of the desired product, mp 81-83° C.; 1H NMR (400 MHz, DMSO-d6) δ 1.42 (t, 3H), 4.26 (q, 2H), 7.18 (d, 1H), 8.26 (dd, 1H), 8.55 (d, 1H)
  • Analysis for C8H8INO3
  • Theory: C, 32.79; H, 2.75; N, 4.78.
  • Found: C, 32.71; H, 2.58; N, 4.53.
  • EXAMPLE 9 (4-Ethoxy-3-iodophenyl)amine
  • A suspension of iron (3.81 g, 70 mmol) and ammonium chloride (5.47 g, 102 mmol) in 80 mL of ethanol and 25 mL of water was heated to reflux. 1-Ethoxy-2-iodo-4-nitrobenzene (5.0 g, 20 mmol) was added in portions and the reaction was heated at reflux for 1 hour. The hot mixture was filtered through Celite, washed with hot ethanol. The filtrate was concentrated in vacuo and treated with ethyl acetate and water. The organic layer was extracted, washed with brine, dried over magnesium sulfate and filtered. Removal of the solvent in vacuo provided 5.1 g of (4-ethoxy-3-iodophenyl)amine as a light brown oil; 1H NMR (400 MHz, DMSO-d6) δ 1.30 (s, 3H), 3.89 (q, 2H), 4.82 (bs, 2H), 6.53 (dd, 1H), 6.72 (d, 1H), 7.11 (d, 1H); MS (ES) m/z 263.9 (M+H)+
  • Analysis for C8H10INO
  • Theory: C, 36.52; H, 3.83; N, 5.32.
  • Found: C, 36.84; H, 3.71; N, 4.96.
  • EXAMPLE 10 2-Iodo-1-(2-methoxyethoxy)-4-nitrobenzene
  • A suspension of 2-iodo-4-nitrophenol (15.5 g, 58.7 mmol) [ref: Kometani, T.; Watt, D. S.; Ji, T., Tetrahedron Lett. (1985), 26(17), 2043], 2-chloroethyl methyl ether (10.69 mL, 117 mmols), potassium carbonate (16.8 g, 117 mmol) and sodium iodide (100 mg) in 160 mL of N,N-dimethylformamide was heated at 70-80° C. for 4 hours. An additional 5.35 mL of 2-chloroethyl methyl ether was added and the reaction was heated until thin layer chromatography (TLC) showed the absence of the phenol. The mixture was filtered and the filtrate was concentrated in vacuo. The solid was partitioned between ethyl acetate and 1 N sodium hydroxide. The organic layer was dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography eluting with a gradient of hexane to 50% ethyl acetate in hexane. The resultant oil was recrystallized from ether to provide 7.5 g of 2-iodo-1-(2-methoxyethoxy)-4-nitrobenzene as a white solid, mp 35-36° C.; 1H NMR (300 MHz, DMSO-d6) δ 3.37 (s, 3H), 3.74 (t, 2H), 4.33 (t, 2H), 7.20 (d, 1H), 8.26 (d, 1H), 8.56 (s, 1H)
  • Analysis for C9H10INO4
  • Theory: C, 33.46; H, 3.12; N, 4.34.
  • Found: C, 34.39; H, 3.11; N, 4.41.
  • EXAMPLE 11 [3-Iodo-4-(2-methoxyethoxy)phenyl]amine
  • A suspension of iron (3.89 g, 69.7 mmol), ammonium chloride (5.0 g, 93.5 mmol), in 100 mL of ethanol and 28 mL of water was heated to reflux. 2-Iodo-1-(2-methoxyethoxy)-4-nitrobenzene (7.5 g, 23.2 mmol) was added and the reaction was heated at reflux for 3 hours. The mixture was cooled to room temperature and filtered through Celite, washed with ethanol. The filtrate was concentrated in vacuo until a precipitate appeared. The precipitate was removed by filtration through Magnesol and the filtrate was concentrated in vacuo to provide 4.0 g of [3-iodo-4-(2-methoxyethoxy)phenyl]amine as an oil; 1H NMR (300 MHz, DMSO-d6) δ 3.33 (s, 3H), 3.62 (t, 2H), 3.95 (t, 2H), 5.01 (d, 2H), 6.55 (d, 1H), 6.73 (d, 1H), 7.03 (s, 1H); MS (ES) m/z 294.0 (M+H)
  • Analysis for C9H12INO2
  • Theory: C, 36.88; H, 4.13; N, 4.78.
  • Found: C, 37.28; H, 4.29; N, 4.80.
  • EXAMPLE 12 3,4-Dimethoxyanilino-N-(dimethyl)-2-cyano-2-propenamide
  • 3,4-Dimethoxyaniline (1.50 g, 0.010 mol), dimethylaminocyanoacetamide (1.50 g, 0.013 mol) and triethylorthoformate (2 mL, 0.012 mol) were mixed in ethylene glycol (10 mL) and the solution heated to reflux. The solution was stirred for 5 hours then allowed to cool to room temperature when precipitate was formed. Water (120 mL) was added and the suspension filtered. The wet solids were suspended in dichloromethane (100 mL), stirred with magnesium sulfate, filtered and concentrated to provide 3,4-dimethoxyanilino-N-(dimethyl)-2-cyano-2-propenamide as a grey solid (1.54 g, 57% yield, 88% purity by GC). 1H NMR (300 MHz, DMSO-d6): 10.9 (d, J=21 Hz, 1H), 8.23 (d, J=21 Hz, 1H), 7.08 (s, 1H), 6.9-6.9 (m, 2H), 3.78 (s, 3H), 3.72 (s, 3H), 3.03 (bs, 6H).
  • EXAMPLE 13 3,4-Dimethoxyanilino-N-(3-chloro-4-fluorophenyl)-2-cyano-2-propenamide
  • 3,4-Dimethoxyaniline (0.30 g, 0.002 mol) and (3-chloro-4-fluoro)-2-cyanoacetamide (0.50 g, 0.0024 mol) and triethylorthoformate (2 mL, 0.012 mol) were mixed in ethylene glycol (4 ml) and heated to reflux. The solution was stirred for 1.5 hours then allowed to cool to −50° C. and water (15 mL) was added. The suspension was allowed to cool to room temperature and the solids isolated by filtration, washed with MTBE (2×15 mL) to provide 3,4-dimethoxyanilino-N-(3-chloro-4-fluorophenyl)-2-cyano-2-propenamide as a beige solid (0.55 g, 93% purity by LCMS): 1H NMR, DMSO-d6): 11.3 (d, J=15 Hz, 1H), 9.76 (s, 1H), 8.70 (d, J=15 Hz, 1H), 7.97 (d, J=8 Hz, 1H), 7.6-7.57 (m, 1H), 7.51 (d, J=9 Hz, 1H), 7.38 (t, J=9 Hz, 1H), 7.29 (s, 1H), 6.97 (d, J=8 Hz, 1H), 3.72 (s, 3H), 3.03 (bs, 6H).
  • EXAMPLE 14 4-Bromo-3-ethoxyanilino-(N-3-chloro-4-fluorophenyl)-2-cyano-2-propenamide
  • A 2-L round-bottomed flask under N2 equipped with an overhead stirrer, a condenser, thermocouple and 100 mL dropping funnel were charged with (3-chloro-4-fluoro)-2-cyanoacetamide (100 g, 0.47 mol), 4-bromo-3-ethoxyaniline hydrochloride (100.4 g, 0.40 mol) and iso-propanol (500 mL). Triethylorthoformate (64 mL, 57 g, 0.38 mol) was added and the suspension heated to 72° C. After 1.5 hours a further aliquot of triethylorthoformate (64 mL, 57 g, 0.38 mol) was added and the reflux continued. After a further 2 hours at reflux a third aliquot of triethylorthoformate (64 mL, 57 g, 0.38 mol) was added and heat continued for 10 hours. The reaction mixture was cooled to 20-25° C. and the precipitate collected by filtration. The filter cake was washed with iso-propanol (2×100 mL) and dried under vacuum (45° C., 50 mm Hg) to provide 4-bromo-3-ethoxyanilino-(N-3-chloro-4-fluorophenyl)-2-cyano-2-propenamide as an off-white solid (136.5 g, 70% yield, >97.5% purity by HPLC mp 226-228° C.). 1H NMR, DMSO-d6): 11.3 (d, J=15 Hz, 1H), 9.76 (s, 1H), 8.70 (d, J=15 Hz, 1H), 7.97 (d, J=8 Hz, 1H), 7.6-7.57 (m, 1H), 7.51 (d, J=9 Hz, 1H), 7.38 (t, J=9 Hz, 1H), 7.29 (s, 1H), 6.97 (d, J=8 Hz, 1H), 4.14 (q, J=9 Hz, 2H), 1.37 (t, J=9 Hz, 3H).
  • EXAMPLE 15 3-Fluoro-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (1.00 g, 3.86 mmol) in 200 mL of iso-propanol was added 3-fluoro-p-anisidine (0.60 g, 4.25 mmol). This mixture was heated to reflux to give a clear yellow solution. To this solution, triethylorthoformate (1.72 mL, 10.34 mmol) was added dropwise and the reaction mixture was heated at reflux overnight. An additional 2 mL of triethylorthoformate was added and the mixture was heated at reflux overnight. An additional 2 mL of triethylorthoformate was added and the mixture was heated at reflux overnight. The mixture was allowed to cool to room temperature and the white solid was collected by filtration, washed with iso-propanol, and dried overnight at ˜40° C. under reduced pressure. Purification by suspension in hot ethyl acetate followed by addition of cold hexanes gave 1.08 g (68%) of 3-fluoro-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide as a white solid, mp 275-276° C.; MS 408.1, 410.1 (M−H)−
  • Analysis for C18H14Cl2FN3O3
  • Calcd: C, 52.70; H, 3.44; N, 10.24.
  • Found: C, 52.44; H, 3.26; N, 10.14.
  • EXAMPLE 16 3-(3-Chloropropoxy)-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (738 mg, 2.84 mmol) in 100 mL of iso-propanol was added 3-(3-chloropropoxy)-4-methoxyaniline (611 mg, 2.84 mmol). This mixture was heated to reflux and triethylorthoformate (3.0 mL, 18.0 mmol) was added dropwise. The reaction mixture was heated at reflux overnight. The mixture was filtered while still warm and the collected white solid was washed with iso-propanol to give 610 mg of 3-(3-chloropropoxy)-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide, mp 185-192° C.; MS (ES) m/z 482.1, 484.2, 486.0 (M−H)−
  • Analysis for C21H20Cl3N3O4
  • Calcd: C, 52.03; H, 4.16; N, 8.67.
  • Found: C, 51.89; H, 4.11; N, 8.53.
  • EXAMPLE 17 3-Bromo-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (900 mg, 3.46 mmol) in 100 mL of iso-propanol was added 3-bromo-4-methoxyaniline (700 mg, 3.46 mmol). This mixture was heated to reflux and triethylorthoformate (3.3 mL, 19.8 mmol) was added dropwise. The reaction mixture was heated at reflux for 6 hours. The mixture was filtered while still warm and the collected white solid was washed with iso-propanol to give 376 mg of 3-bromo-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide, mp >250° C.; MS (ES) m/z 467.7, 469.9, 471.7 (M−H)−
  • Analysis for C18H14BrCl2N3O3
  • Calcd: C, 45.89; H, 3.00; N, 8.92.
  • Found: C, 45.75; H, 2.77; N, 8.78.
  • EXAMPLE 18 3-Bromo-4-ethoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (715 mg, 2.74 mmol) in 50 mL of iso-propanol was added 3-bromo-4-ethoxyaniline (586 mg, 2.71 mmol). This mixture was heated to reflux and triethylorthoformate (2.7 mL, 16.2 mmol) was added dropwise. The reaction mixture was heated at reflux overnight. The mixture was filtered while still warm and the collected white solid was washed with iso-propanol to give 666 mg of 3-bromo-4-ethoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide, mp >250° C.; MS (ES) m/z 482.0, 484.0, 486.0 (M−H)−
  • Analysis for C19H16BrCl2N3O3
  • Calcd: C, 47.04; H, 3.32; N, 8.66.
  • Found: C, 46.64; H, 3.40; N, 8.59.
  • EXAMPLE 19 4-Bromo-3-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (6.1 g, 23.5 mmol) in 500 mL of iso-propanol was added 5-amino-2-bromoanisole (5.0 g, 24.8 mmol). This mixture was heated to reflux to give a clear yellow solution. To this solution, triethylorthoformate (10.6 mL, 63.5 mmol) was added dropwise and the reaction mixture was heated at reflux for 5 hours. An additional 10.6 mL of triethylorthoformate was added and the mixture was heated at reflux overnight. The mixture was allowed to cool to room temperature. The solid was collected by filtration, washed with iso-propanol and ethyl acetate, and then dried overnight at ˜40° C. under reduced pressure to give 8.1 g (73%) of 4-bromo-3-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide as a yellow solid, mp >250° C.; MS (ES) m/z 470.0, 472.0 (M+H)+
  • Analysis for C18H14BrCl2N3O3
  • Calcd: C, 45.89; H, 3.00; N, 8.92.
  • EXAMPLE 20 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-iodo-4-methoxyphenyl)amino]prop-2-enamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (5.00 g, 19.30 mol) in 400 mL of iso-propanol, under N2, was added 3-iodo-p-anisidine (5.80 g, 23.16 mmol). This mixture was heated to reflux to give a clear yellow solution. To this solution, triethylorthoformate (8.60 mL, 52.11 mmol) was added dropwise and the reaction mixture was heated at reflux overnight. An additional 10 mL of triethylorthoformate was added and the mixture was heated at reflux overnight. The mixture was allowed to cool to room temperature and the white solid was collected by filtration, washed with iso-propanol, and dried overnight at ˜40° C. under reduced pressure. Purification by suspension in hot ethyl acetate followed by addition of cold hexanes gave 8.50 g (85%) of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-iodo-4-methoxyphenyl)amino]prop-2-enamide as a yellow solid, mp 289-290° C.; MS (ES) m/z 516.7 (M−H)−
  • Analysis for C18H14Cl2IN3O3
  • Calcd: C, 41.73; H, 2.72; N, 8.11.
  • Found: C, 40.66; H, 2.94; N, 7.90.
  • EXAMPLE 21 4-Ethoxy-3-fluoroanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (5.9 g, 22.7 mmol) in 400 mL of iso-propanol was added 4-ethoxy-3-fluoroaniline (3.7 g, 23.8 mmol). This mixture was heated to reflux to give a clear solution. To this solution, triethylorthoformate (11.6 mL, 69.6 mmol) was added dropwise and the reaction mixture was heated at reflux for 18 hours. Additional triethylorthoformate (11.6 mL, 69.6 mmol) was added dropwise and the reaction mixture was heated at reflux for 42 hours. The mixture was allowed to cool to room temperature and the solid was collected by filtration, washed with ethyl acetate, and dried in vacuo at 40° C. to give 6.4 g (67%) of 4-ethoxy-3-fluoroanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide as a grey solid, mp 245-247° C.; MS 424.1, 426.1 (M+H)+
  • Analysis for C19H16Cl2FN3O3
  • Calcd: C, 53.79; H, 3.80; N, 9.90.
  • Found: C, 53.39; H, 3.97; N, 9.69.
  • EXAMPLE 22 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-(thien-3-ylamino)prop-2-enamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (1.57 g, 6.06 mmol) in 200 mL of iso-propanol was added 3-aminothiophene (600 mg, 6.06 mmol). This mixture was heated to reflux and triethylorthoformate (6.5 mL, 39.1 mmol) was added dropwise. The reaction mixture was heated at reflux overnight. Additional 3-aminothiophene (214 mg, 2.16 mmol) was added and the reaction mixture was heated at reflux for 5 hours. Additional 3-aminothiophene (100 mg, 1.01 mmol) was added and the reaction mixture was heated at reflux for 2.5 hours. The mixture was allowed to cool to room temperature and the solid was collected by filtration, washed with iso-propanol to give 572 mg (26%) of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-(thien-3-ylamino)prop-2-enamide as a tan solid, mp >250° C.; MS 366.0, 368.2 (M−H)−
  • Analysis for C15H11Cl2N3O2S-1.0H2O
  • Calcd: C, 46.64; H, 3.39; N, 10.88.
  • Found: C, 46.37; H, 2.94; N, 10.60.
  • EXAMPLE 23 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-ethoxy-3-iodophenyl)amino]acrylamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (5.44 g, 21.0 mmol) in 350 mL of iso-propanol, under N2, was added (4-ethoxy-3-iodophenyl)amine (5.0 g, 19.30 mmol). This mixture was heated to reflux and triethylorthoformate (8.53 mL, 51.30 mmol) was added dropwise and the reaction mixture was heated at reflux overnight. The mixture was allowed to cool to room temperature and the yellow solid was collected by filtration, washed with iso-propanol, and dried overnight at ˜40° C. under reduced pressure to give 5.46 g (54%) of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-ethoxy-3-iodophenyl)amino]prop-2-enamide as a yellow solid, mp >245° C.; MS (EI) m/z 531.01 (M)+
  • Analysis for C19H16Cl2IN3O3
  • Calcd: C, 42.88; H, 3.03; N, 7.90.
  • Found: C, 42.99; H, 2.97; N, 7.74.
  • EXAMPLE 24 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{[3-iodo-4-(2-methoxyethoxy)phenyl]amino}acrylamide
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (3.2 g, 12.4 mmol) and [3-iodo-4-(2-methoxyethoxy)phenyl]amine (4.0 g, 13.7 mmol) in 350 mL of iso-propanol was heated to reflux and triethylorthoformate (5.6 mL, 33.7 mmol) was added dropwise. The reaction mixture was heated at reflux for 3 hours. Additional triethylorthoformate (2.0 mL, 12.0 mmol) was added dropwise and the reaction mixture was heated at reflux overnight. The mixture was allowed to cool to room temperature and the solid was collected by filtration, washed with iso-propanol, diethyl ether, and ethyl acetate and then dried in vacuo to give 5.5 g of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{[3-iodo-4-(2-methoxyethoxy)phenyl]amino}acrylamide as a yellow solid, mp 209-210° C.; MS (ES) 560.0, 562.1 (M−H)−
  • Analysis for C20H18Cl2IN3O4
  • Calcd: C, 42.73; H, 3.23; N, 7.47.
  • Found: C, 43.04; H, 3.07; N, 7.28.
  • EXAMPLE 25 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-bromo)phenyl)amino]acrylamide
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (3.0 g, 11.5 mmol) and 3-bromoaniline (1.3 mL, 11.9 mmol) in 250 mL of iso-propanol was heated to reflux and triethylorthoformate (12 mL, 72.2 mmol) was added dropwise. The reaction mixture was heated at reflux overnight. The mixture was allowed to cool slightly and the solid was collected by filtration while still warm, washed with iso-propanol and diethyl ether to give 2.05 g of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-bromo)phenyl)amino]acrylamide as a yellow solid, mp >250° C.; MS (ES) 437.8, 439.7, 441.8 (M−H)−
  • Analysis for C17H12BrCl2N3O2
  • Calcd: C, 46.29; H, 2.74; N, 9.53.
  • Found: C, 46.33; H, 2.73; N, 9.40.
  • EXAMPLE 26 6-Bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile
  • A 22-L round-bottomed flask under N2 equipped with overhead stirrer, thermocouple, condenser and 1000 mL dropping funnel was charged with 4-bromo-3-ethoxyanilino-(N-3-chloro-4-fluorophenyl)-2-cyano-2-propenamide (1000 g, 2.29 mol) methanol (40 mL) and acetonitrile (7.50 L). The suspension was heated to 80-82° C. Phosphorus oxychloride (740 g, 4.83 mol) was charged to the dropping funnel and added over 30 minutes. The suspension was refluxed for 24 hours until <3% starting material was present by HPLC. The mixture was cooled to 0-10° C. and filtered. The filter cake was washed with acetonitrile (3×0.63 kg) and the filter cake dried on the filter funnel for 2 hours. A 50-L round-bottomed flask equipped with overhead stirrer, thermocouple, condenser and 1000 mL dropping funnel was charged with the filter cake from above. THF (4.44 kg) was added and the suspension warmed to 35-40° C. 28% Ammonium hydroxide (0.63 kg) and water (2 kg) were mixed in a flask and transferred to the dropping funnel. The ammonium hydroxide solution was added to the THF suspension over 30 minutes maintaining the temperature below 55° C. Water (10 kg) was added over 25 minutes then the resulting suspension was cooled to 15-20° C. The precipitate was collected by filtration then washed with hot water (3×1 kg), acetonitrile (2×1 kg) and dried under vacuum (55 mm Hg, 65° C.) to provide 6-bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile as an orange-yellow solid (806 g, 84% yield, >96% purity by HPLC mp 213-215° C.). 1H NMR (300 MHz, CDCl3): 8.7 (s, 1H), 7.99 (s, 1H), 7.40-6.91 (m, 5H), 4.18 (q, J=7.5 Hz, 2H), 1.56 (t, J=7.5 Hz, 3H).
  • EXAMPLE 27 6-Bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile
  • A 250-mL round-bottomed flask under N2 equipped with overhead stirrer, thermocouple, condenser and 10 mL dropping funnel was charged with 4-bromo-3-ethoxyanilino-(N-3-chloro-4-fluorophenyl)-2-cyano-2-propenamide (10.0 g, 0.023 mol) pyridine (3.80 g, 0.048 mol) and toluene (100 ml). The suspension was heated to 110° C. Phosphorus oxychloride (4.4 mL, 0.047 mol) was charged to the dropping funnel and added over 3.75 hours. Heat was continued for 0.25 hours until <3% starting material was present by HPLC then the suspension allowed to cool to room temperature. Water (50 mL) was added to the reaction mixture as a slow stream then the mixture basified by the addition of 50% aq, sodium hydroxide solution. The suspension was cooled to 10-15° C. and the precipitate was collected by filtration then washed with water (2×10 mL), toluene (2×10 ml)) and dried under vacuum (55 mm Hg, 65° C.) to provide 6-bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile as an orange-yellow solid (6.90 g, 72% yield, >92.2% purity by HPLC). 1H NMR (300 MHz, CDCl3): 8.7 (s, 1H), 7.99 (s, 1H), 7.40-6.91 (m, 5H), 4.18 (q, J=7.5 Hz, 2H), 1.56 (t, J=7.5 Hz, 3H).
  • EXAMPLE 28 6-Bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile
  • A 5-L round-bottomed flask under N2 equipped with overhead stirrer, thermocouple, condenser and 500 mL dropping funnel was charged with 4-bromo-3-ethoxyanilino-(N-3-chloro-4-fluorophenyl)-2-cyano-2-propenamide (500 g, 1.14 mol) 1-butanol (15 g, 0.20 mol) and toluene (3.5 L). The suspension was heated to 105-108° C. Phosphorus oxychloride (370 g, 2.41 mol) was charged to the dropping funnel and added to the suspension over 10 hours. Heat was continued for further 16 hours until <3% starting material was present by HPLC. The mixture was cooled to 0-10° C. and filtered. The filter cake was washed with toluene (2×0.3 kg) and the filter cake dried on the filter funnel for 2 hours.
  • A 12-L round-bottomed flask equipped with overhead stirrer, thermocouple, condenser and 1000 mL dropping funnel was charged with the filter cake from above. THF (2.22 kg) was added and the suspension warmed to 35-40° C. 28% Ammonium hydroxide (0.63 kg) and water (1 kg) were mixed in a flask and transferred to the dropping funnel. The ammonium hydroxide solution was added to the THF suspension over 30 minutes maintaining the temperature below 55° C. Water (5 kg) was added over 25 minutes then the resulting suspension was cooled to 15-20° C. The precipitate was collected by filtration then washed with hot water (3×1 kg), acetonitrile (2×0.4 kg) and dried under vacuum (55 mm Hg, 65° C.) to provide 6-Bromo-[7-ethoxy-4-(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile as an orange-yellow solid (389.1 g, 81% yield, >97.5% purity by HPLC). 1H NMR (300 MHz, CDCl3): 8.7 (s, 1H), 7.99 (s, 1H), 7.40-6.91 (m, 5H), 4.18 (q, J=7.5 Hz, 2H), 1.56 (t, J=7.5 Hz, 3H)
  • Analysis for C18H12BrClFN3O
  • Calcd: C, 51.39; H, 2.88; N, 9.99.
  • Found: C, 51.50; H, 2.75; N, 10.09.
  • EXAMPLE 29 6,7-Dimethoxy-4-(N,N-dimethylamino)-3-quinolinecarbonitrile
  • A suspension of 3,4-dimethoxyanilino-N-(dimethyl)-2-cyano-2-propenamide (0.500 g, 1.82 mmol) in acetonitrile (10 mL) was treated with phosphorus oxychloride (0.17 mL, 0.28 g, 1.82 mmol) and heated to reflux. The mixture was stirred for 1 hour and an additional aliquot of phosphorus oxychloride (0.17 mL, 0.28 g, 1.82 mmol) added. The mixture was heated at reflux for 8 hours then stirred at room temperature for 24 hours. The resultant solution was concentrated in vacuo and stirred with aqueous sodium carbonate solution. The mixture was extracted with methylene chloride (3×30 mL), the combined extracts were dried over magnesium sulfate, filtered and concentrated in vacuo to produce 6,7-dimethoxy-4-(N,N-dimethylamino)-3-quinolinecarbonitrile as a tan solid (0.37 g, 79%, 89% purity by GC).
  • EXAMPLE 30 6,7-Dimethoxy-4-[(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile
  • A suspension of 3,4-dimethoxy-N-(3-chloro-4-fluorophenyl)-2-cyano-2-propenamide (0.84 g, 2.23 mmol) in acetonitrile (10 mL) was treated with phosphorus oxychloride (0.42 mL, 0.68 g, 4.50 mmol) and heated to reflux for 8 hours. The suspension was cooled to room temperature and stirred with saturated sodium carbonate solution (70 mL). The mixture was extracted with methylene chloride (3×70 mL), the combined extracts were dried over magnesium sulfate, filtered and concentrated in vacuo to produce 6,7-dimethoxy-4-[(3-chloro-4-fluorophenyl)amino]-3-quinolinecarbonitrile as brown solid (0.74 g, 93%, 95% by LC-MS 1.3% isomer by LC-MS). 1H NMR (300 MHz, DMSO-d6).
  • EXAMPLE 31 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-6-ethoxy-7-fluoro-3-quinolinecarbonitrile
  • A suspension of 4-ethoxy-3-fluoroanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide (6.28 g, 14.8 mmol) in 54 mL of acetonitrile and 2.0 mL of methanol was heated to reflux and phosphorous oxychloride (8.3 mL, 88.8 mmol) was added dropwise. The mixture was heated at reflux for 17 hours. The resultant solution was concentrated in vacuo and acetonitrile was added to the residue. The solid was collected by filtration, washed with acetonitrile. The solid was suspended in tetrahydrofuran and neutralized with concentrated ammonium hydroxide. After stirring for 30 minutes, water was added and the mixture was stirred for 1 hour. The solid was collected by filtration, washed with water, followed by 1:1 diethyl ether and hexane. The light yellow solid was dried in vacuo to provide 2.2 g (37%) of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-ethoxy-7-fluoro-3-quinolinecarbonitrile, mp 185-187° C.; 1H NMR (400 MHz, DMSO-d6) δ 1.95 (t, J=7 Hz, 3H), 3.87 (s, 3H), 4.29 (q, J=7 Hz, 2H), 7.39 (s, 1H), 7.66-7.81 (m, 2H), 8.09 (d, J=9 Hz, 1H), 8.49 (s, 1H), 9.80 (s, 1H); MS (ES) m/z 406.1, 408.2 (M+H)+
  • Analysis for C19H14Cl2FN3O2-0.4H2O
  • Calcd: C, 55.20; H, 3.61; N, 10.16.
  • Found: C, 55.25; H, 3.53; N, 10.15.
  • EXAMPLE 32 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-7-fluoro-6-methoxy-3-quinolinecarbonitrile
  • To a suspension of 3-fluoro-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide (360 mg, 0.88 mmol) in 40 mL of acetonitrile was added 0.1 mL of methanol. The reaction mixture was heated to reflux and phosphorous oxychloride (0.65 mL, 7.0 mmol) was added dropwise, via syringe. After 2 hours the mixture became a clear orange solution. This solution was heated at reflux overnight. After 24 hours, the reaction mixture was cooled in an ice-bath and the solid was collected by filtration, washed with cold acetonitrile (20 mL) and then suspended in tetrahydrofuran (50 mL). To both the acetonitrile filtrate and the tetrahydrofuran suspension were added concentrated ammonium hydroxide (2×25 mL) and the mixtures were stirred for 1 hour. Water (2×400 mL) was added and stirring was continued for 2 hours. The resulting solids were combined, washed with hot water and dried under reduced pressure at ˜40° C., overnight, to provide 189 mg (55%) of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-7-fluoro-6-methoxy-3-quinolinecarbonitrile as orange crystals, mp 219-221° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.87 (s, 3H), 4.02 (s, 3H), 7.36 (s, 1H), 7.65-7.80 (m, 2H), 8.08 (d, J=9 Hz, 1H), 8.48 (s, 1H), 9.85 (s, 1H); MS (ES) m/z 392.0, 394.0 (M+H)+
  • Analysis for C18H12Cl2FN3O2-0.5H2O
  • Calcd: C, 53.88; H, 3.27; N, 10.48.
  • Found: C, 54.09; H, 3.20; N, 10.24.
  • EXAMPLE 33 7-(3-Chloropropoxy)-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-3-quinolinecarbonitrile
  • A suspension of 3-(3-chloropropoxy)-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide (462 mg, 0.95 mmol) in 40 mL of acetonitrile was heated to reflux and phosphorous oxychloride (0.60 mL) was added dropwise, via syringe. The reaction mixture was heated at reflux overnight, then cooled to room temperature and concentrated in vacuo. The residue was cooled to 0° C. and saturated aqueous sodium bicarbonate was added. The mixture was stirred for 10 minutes and the solid was collected by filtration, washed with water. The solid was suspended in hot ethyl acetate and methanol and then filtered to provide 200 mg of 7-(3-chloropropoxy)-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-3-quinolinecarbonitrile as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 2.30 (m, 2H), 3.84 (t, J=6 Hz, 1H), 3.88 (s, 3H), 4.01 (s, 3H), 4.32 (t, J=6 Hz, 2H), 7.46 (s, 1H), 7.51 (s, 1H), 7.81 (s, 1H), 8.14 (s, 1H), 8.81 (s, 1H), 10.83 (br s, 1H); MS (ES) m/z 466.1, 468.1 (M+H)+.
  • EXAMPLE 34 7-Bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-3-quinolinecarbonitrile
  • A suspension of 3-bromo-4-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide (4.00 g, 8.50 mmol) in 200 mL of acetonitrile was heated to reflux and phosphorous oxychloride (5.0 mL) was added dropwise, via syringe. The reaction mixture was heated at reflux overnight, then cooled to room temperature and concentrated in vacuo. The residue was cooled to 0° C. and saturated aqueous sodium bicarbonate was added. The mixture was stirred for 1 hour and then extracted with a large volume of ethyl acetate. The organic layer was washed with saturated aqueous sodium bicarbonate, dried over magnesium sulfate and filtered. The filtrate was reduced in volume until solids appeared. The solids were collected by filtration, washed with diethyl ether and hexanes to provide 1.73 g of 7-bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-3-quinolinecarbonitrile as a light tan solid, mp >250° C. 1H NMR (400 MHz, DMSO-d6) δ 3.87 (s, 3H), 4.04 (s, 3H), 7.41 (s, 1H), 7.78 (s, 1H), 7.99 (s, 1H), 8.22 (s, 1H), 8.48 (s, 1H), 9.93 (s, 1H); MS (ES) m/z 449.9, 451.9, 453.9 (M−H)−
  • Analysis for C18H12BrCl2N3O2
  • Calcd: C, 47.71; H, 2.67; N, 9.27.
  • Found: C, 47.94; H, 2.74; N, 9.03.
  • EXAMPLE 35 7-Bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-ethoxy-3-quinolinecarbonitrile
  • A suspension of 3-bromo-4-ethoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide (630 mg, 1.29 mmol) in 35 mL of acetonitrile was heated to reflux and phosphorous oxychloride (0.80 mL) was added dropwise, via syringe. The reaction mixture was heated at reflux overnight, then cooled to room temperature. The solids were collected by filtration, washed with saturated aqueous sodium bicarbonate. The solids were stirred with saturated aqueous sodium bicarbonate for 1 hour and then collected by filtration, washed with saturated aqueous sodium bicarbonate and water. The solid was recrystallized from methanol and dried in vacuo to provide 225 mg of 7-bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-ethoxy-3-quinolinecarbonitrile as a yellow solid, mp >250° C. 1H NMR (400 MHz, DMSO-d6) δ 1.47 (t, J=7 Hz, 3H), 3.87 (s, 3H), 4.32 (q, J=7 Hz, 2H), 7.45 (s, 1H), 7.82 (s, 1H), 8.16 (s, 1H), 8.27 (s, 1H), 8.76 (s, 1H), 10.73 (s, 1H); MS (ES) m/z 466.1, 468.0, 470.1 (M+H)+
  • Analysis for C19H14BrCl2N3O2:2.0H2O
  • Calcd: C, 45.35; H, 3.61; N, 8.35.
  • Found: C, 44.97; H, 3.17; N, 8.07.
  • EXAMPLE 36 6-Bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-7-methoxy-3-quinolinecarbonitrile
  • To a suspension of (5.13 g, 11 mmol) of 4-bromo-3-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide in 400 mL of acetonitrile was added 1.5 mL of methanol. The mixture was heated to reflux and phosphorous oxychloride (4.10 mL, 44.0 mmol) was added dropwise. The resulting mixture was heated at reflux for 26 hours. Additional phosphorous oxychloride (2.1 mL) and methanol (0.75 mL) were added and the mixture was heated at reflux for 21 hours. After cooling to 0-5° C. in an ice-bath, the solid was collected by filtration, washed with cold acetonitrile (100 mL) and then suspended in tetrahydrofuran (100 mL). To the tetrahydrofuran suspension was added concentrated ammonium hydroxide and the mixtures were stirred for 1 hour. Water (300 mL) was added and stirring was continued for 1 hour. The solid was collected by filtration, washed with hot water (46° C.), acetonitrile and dried in vacuo overnight to recover 2.1 g of 4-bromo-3-methoxyanilino-N-(2,4-dichloro-5-methoxyphenyl)-2-cyano-2-propenamide. The acetonitrile filtrate and washing were combined and concentrated aqueous ammonium hydroxide was added. The resulting acetonitrile mixture was stirred for 1 hour. Water was added and stirring was continued for 1 hour. The solid was collected by filtration, washed with water, ethyl acetate and ether, then dried in vacuo to provide 1.92 g (39%) of 6-bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-7-methoxy-3-quinolinecarbonitrile as a yellow solid, mp 256-257° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.85 (s, 3H), 4.04 (s, 3H), 7.37 (s, 1H), 7.46 (s, 1H), 7.75 (s, 1H), 8.56 (s, 1H), 8.89 (s, 1H), 9.93 (s, 1H); MS (ES) m/z 451.9, 454.0 (M+H)+
  • Analysis for C18H12BrCl2N3O2
  • Calcd: C, 47.71; H, 2.67; N, 9.27.
  • Found: C, 47.41; H, 2.67; N, 9.56.
  • EXAMPLE 37 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-7-iodo-6-methoxy-3-quinolinecarbonitrile
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-iodo-4-methoxyphenyl)amino]prop-2-enamide (720 mg, 1.39 mmol) in 40 mL of acetonitrile was added 0.2 mL of methanol. The mixture was heated to reflux and phosphorous oxychloride (1.24 mL, 13.9 mmol) was added dropwise, via syringe. This solution was heated at reflux overnight. After 24 hours, the mixture was cooled in an ice-bath and the solid was collected by filtration, washed with cold acetonitrile (40 mL) and then suspended in tetrahydrofuran (100 mL). To both the acetonitrile filtrate and the tetrahydrofuran suspension were added concentrated ammonium hydroxide (2×50 mL) and the mixtures were stirred for 1 hour. Water (2×800 ml) was added and stirring was continued for 2 hours. The resulting solids were combined, washed with hot water and dried under reduced pressure at ˜40° C., overnight, to provide 200 mg (29%) of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-7-iodo-6-methoxy-3-quinolinecarbonitrile, as yellow solid, mp 253-254° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.86 (s, 3H), 4.00 (s, 3H), 7.33 (s, 1H), 7.74 (s, 1H), 7.86 (s, 1H), 8.39 (s, 1H), 8.43 (s, 1H), 9.61 (s, 1H); MS (ES) m/z 500.0, 502.1 (M+H)+
  • Analysis for C18H12Cl21N3O2—H2O
  • Theory: C, 41.72, H, 2.72; N, 8.11.
  • Found: C, 41.80; H, 2.52; N, 7.87.
  • EXAMPLE 38 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-6-ethoxy-7-iodo-3-quinolinecarbonitrile
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-ethoxy-3-iodophenyl)amino]prop-2-enamide (2.0 g, 3.76 mmol) in 100 mL of toluene was heated to reflux and phosphorous oxychloride (3.5 mL, 37.6 mmol) was added dropwise via syringe. This suspension was heated at reflux for 6 hours and additional phosphorous oxychloride (3.5 mL, 37.6 mmol) was added to slowly give a dark solution. After 72 hours the mixture was cooled to room temperature, the solid was filtered and washed with toluene and ether. The light brown solid was dried under reduced pressure at −40° C., overnight, to provide 1.47 g (76%) of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-ethoxy-7-iodo-3-quinolinecarbonitrile as a yellow solid, mp 213-215° C.; 1H NMR (300 MHz, DMSO-d6) δ 1.48 (t, 3H, J=6.9 Hz), 4.32 (q, 2H, J=6.9 Hz), 3.88 (s, 3H), 7.53 (s, 1H), 7.87 (s, 1H), 8.06 (s, 1H), 8.49 (s, 1H), 9.02 (s, 1H), 11.14 (bs, 1H); MS (ES) m/z 514.1 (M+H)+
  • Analysis for C19H14Cl2IN3O2-4.0HCl
  • Theory: C, 34.58, H, 2.75; N, 6.37.
  • Found: C, 34.79; H, 2.60; N, 6.13.
  • EXAMPLE 39 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-7-iodo-6-(2-methoxyethoxy)-3-quinolinecarbonitrile
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{[3-iodo-4-(2-methoxyethoxy)phenyl]amino}acrylamide (1.0 g, 1.80 mmol) in 35 mL of toluene was heated to reflux and phosphorous oxychloride (1.7 mL, 18.0 mmol) was added dropwise. The mixture was heated at reflux for 2 hours, and an additional 1.7 mL of phosphorous oxychloride was added. The mixture was continued to heat at reflux for 2 hours, and an additional 1.7 mL of phosphorous oxychloride was added and the reaction mixture was heated at reflux overnight. The reaction is then cooled to room temperature and water was added followed by 10 N sodium hydroxide. The mixture was stirred in an ice-bath for 30 minutes and the precipitate was collected by filtration, washed with methylene chloride to give 870 mg of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-7-iodo-6-(2-methoxyethoxy)-3-quinolinecarbonitrile as a yellow solid; 1H NMR (400 MHz, DMSO-d6) δ 3.41 (s, 3H), 3.81 (t, 2H), 3.87 (s, 3H), 4.34 (t, 2H), 7.40 (s, 1H), 7.78 (s, 1H), 7.88 (s, 1H), 8.43 (s, 1H), 8.45 (s, 1H), 9.85 (s, 1H); MS (ES) m/z 542.1, 544.1 (M−H)−
  • Analysis for C20H16Cl2IN3O3-4.0HCl
  • Calcd: C, 34.81; H, 2.92; N, 6.09.
  • Found: C, 35.04; H, 2.53; N, 5.95.
  • EXAMPLE 40 7-Bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-3-quinolinecarbonitrile
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(3-bromo)phenyl)amino]acrylamide (1.5 g, 3.40 mmol) in 50 mL of acetonitrile was heated to reflux and phosphorous oxychloride (4.0 mL, 42.4 mmol) was added dropwise. The mixture was heated at reflux overnight. The reaction was cooled to room temperature and the volatiles were removed in vacuo. Ice was added to the residue followed by saturated aqueous sodium bicarbonate. The mixture was stirred and then extracted with ethyl acetate. The organic layer was washed with water, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 310 mgs of 7-bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-3-quinolinecarbonitrile. The mother liquor was concentrated in vacuo and purified by flash column chromatography eluting with a gradient of 3:1 hexane:ethyl acetate to 1:1 hexane:ethyl acetate to give 310 mg of 7-bromo-4-[(2,4-dichloro-5-methoxyphenyl)amino]-3-quinolinecarbonitrile as a light orange solid; 1H NMR (400 MHz, TFA/DMSO-d6) δ 3.87 (s, 3H), 7.42 (s, 1H), 7.79 (s, 1H), 7.97 (s, 1H), 8.14 (s, 1H), 8.56 (s, 1H), 8.85 (s, 1H); MS (ES) m/z 422.0, 423.9, 426.0 (M+H)+
  • Analysis for C17H10BrCl2N3O
  • Calcd: C, 48.26; H, 2.38; N, 9.93.
  • Found: C, 48.47; H, 2.37; N, 9.70.
  • EXAMPLE 41 7-[(2,4-Dichloro-5-methoxyphenyl)amino]thieno[3,2-b]pyridine-6-carbonitrile
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-(thien-3-ylamino)prop-2-enamide (500 mg, 1.35 mmol) in 35 mL of acetonitrile was heated to reflux and phosphorous oxychloride (0.84 mL, 8.98 mmol) was added dropwise. The mixture was heated at reflux overnight. The resultant solution was concentrated in vacuo and the residue was cooled to 0° C. Ice water was added followed by the slow addition of saturated aqueous sodium bicarbonate. The mixture was stirred for 10 minutes then partitioned between ethyl acetate and additional saturated aqueous sodium bicarbonate. The organic layer was washed with water, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was triturated with ethyl acetate and hexane to provide 337 mg (71%) of 7-[(2,4-dichloro-5-methoxyphenyl)amino]thieno[3,2-b]pyridine-6-carbonitrile, mp 208-210° C.; 1H NMR (400 MHz, DMSO-d6) δ 3.85 (s, 3H), 7.39 (s, 1H), 7.46 (d, J=5 Hz, 1H), 7.76 (s, 1H), 8.11 (d, J=5 Hz, 1H), 8.61 (s, 1H), 9.74 (s, 1H); MS (ES) m/z 348.1, 350.0 (M−H)−
  • Analysis for C15H9Cl2N3O2S-0.5H2O
  • Calcd: C, 50.15; H, 2.81; N, 11.70.
  • Found: C, 50.32; H, 2.62; N, 11.54.
  • EXAMPLE 42 1-[3-(2-Methoxy-5-nitro-phenoxy)-propyl]-4-methyl-piperazine
  • 2(3-chloropropoxy)-1-methoxy-4-nitrobenzene, (100 g, 0.407 mol) and sodium iodide (9.2 g, 0.614 mol) was added to 520 mL of dry 1,2-dimethoxyethane. To the mixture was then added N-methylpiperazine (82 g, 0.814 mol). Stirred and heated the reaction to reflux. The reaction was monitored by HPLC. After 8 hours, the heating mantle was removed and the reaction was allowed to cool to ambient temperature. The reaction mixture was then drowned in 1 L of ethyl acetate and stirred for a minimum of 1 hour. Bright yellow solids precipitated. The mixture was filtered and rinsed and the salts collected with ethyl acetate. The filtrate was washed once with 400 mL of 0.5 N NaOH. The layers were separated and the organic layer was washed with water (3×390 mL). The combined organic layers were concentrated under vacuum at 35-40° C. to obtain 97.8 g (light orange oil) (77%); 1H NMR (300 MHz, CDCl3) δ 7.91 (dd, J=2.6, 9 Hz, 1H), 7.77 (d, J=2.6 Hz, 1H), 6.90 (d, J=9 Hz, 1H), 4.14 (t, J=7 Hz, 2H), 3.96 (s, 3H) 2.54 (t, J=7 Hz, 2H), 2.48 (m, 8H), 2.30 (s, 3H), 2.06 (m, 2H); MS (ES) m/z 310.1 (M+H).
  • EXAMPLE 43 4-Methoxy-3-[3-(4-methyl-piperazin-1-yl)-propoxy]-phenylamine
  • To a solution of 1-[3-(2-methoxy-5-nitro-phenoxy)-propyl]-4-methyl-piperazine (2.5 g, 8.1 mmol) in 25 mL of iso-propanol was added 10% Pd—C (0.25 g, 10 wt. %). This mixture was shaken under 35-40 psi of hydrogen in a Parr shaker until hydrogen uptake ceased. The reaction mixture was purged with nitrogen and filtered through celite. The resulting solution was concentrated in vacuo to give the crude 4-methoxy-3-[3-(4-methyl-piperazin-1-yl)-propoxy]-phenylamine as a yellow oil which was used directly in the coupling step described below. 1H NMR (300 MHz, CDCl3) δ 6.71 (d, J=8 Hz, 1H), 6.33 (d, J=2.4 Hz, 1H), 6.23 (dd, J=2.4, 8 Hz, 1H), 4.05 (t, J=8 Hz, 2H), 3.80 (s, 3H), 2.53 (m, 10H), 2.30 (s, 3H), 2.02 (m, 2H).
  • EXAMPLE 44 2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{4-methoxy-3-[3-(4-methylpiperazin-1-yl)-propoxy]-phenylamino}-acrylamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (1.75 g, 6.75 mmol) in 8.2 mL of iso-propanol was added triethylorthoformate (1.5 g, 10.1 mmol). The mixture was heated to reflux. After 20 minutes, 4-methoxy-3-[3-(4-methyl-piperazin-1-yl)-propoxy]-phenylamine in 20 mLs of iso-propanol was added dropwise so as to maintain reflux. After 26 hours, the reaction was cooled to room temperature and the suspended solids were filtered. The solids were rinsed with iso-propanol until effluent was colorless. The yellow solid was dried in vacuo to give 3.45 g (93%) of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{4-methoxy-3-[3-(4-methylpiperazin-1-yl)-propoxy]-phenylamino}-acrylamide as a mixture of isomers. 1H NMR (300 MHz, CDCl3) δ 11.35 (br d, 1H), 8.17 (m, 2H), 7.76 (d, 1H), 7.41 (s, 1H), 6.88 (m, 1H) 6.69 (m, 2H), 4.10 (t, J=7 Hz, 2H), 3.96 (s, 3H), 3.87 (s, 3H), 2.55 (t, J=7 Hz, 2H), 2.48 (m, 8H), 2.29 (s, 3H), 2.06 (m, 2H); MS (ES) m/z 548.1 (M+)
  • Analysis for C10H14CINO2
  • Calcd: C, 55.69; H, 6.54; N, 6.49.
  • Found: C, 55.49; H, 6.59; N, 6.32.
  • EXAMPLE 45 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methyl-piperazin-1-yl)-propoxy-3-quinolinecarbonitrile
  • A suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-{4-methoxy-3-[3-(4-methylpiperazin-1-yl)-propoxy]-phenylamino}-acrylamide (6 g, 10.9 mmol) in 9 mL of acetonitrile was heated to reflux and phosphorous oxychloride (21.8 g, 142 mmol) was added dropwise. The reaction mixture was heated at reflux for 40 hours, then cooled to room temperature. The mixture was cooled to 0° C. and 10N sodium hydroxide was added slowly and carefully to neutralize the reaction. The quench was very exothermic; the addition of sodium hydroxide was such that the temperature of the mixture was kept below 55° C. The mixture was basified to pH 9-10. After cooling to <30° C., 500 mL of ethyl acetate was added and the resulting mixture was stirred for 15 minutes. The solids were filtered and then rinsed with ethyl acetate. The filtrate layers were separated and the organic layer was concentrated in vacuo. The crude oil was dissolved in 45 mL of methanol and stirred at room temperature for 1 hour. The crystalline solid that formed was filtered and rinsed with minimal cold methanol. The filtrate was concentrated and the procedure repeated with about half as much methanol. The two crops were combined to give 4.3 g (75%) of crude 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methyl-piperazin-1-yl)-propoxy-3-quinoline-3-carbonitrile; 1H NMR (300 MHz) δ 8.71 (s, 1H), 7.50 (s, 1H), 7.43 (s, 1H), 6.90 (s, 1H), 6.46 (s, 1H), 4.26 (t, J=7 Hz, 2H), 3.78 (s, 3H), 3.67 (s, 3H), 2.57 (t, J=7 Hz, 2H), 2.48 (m, 8H), 2.29 (s, 3H), 2.12 (m, 2H); MS (ES) m/z 265.6, 530.1 (M+)
  • EXAMPLE 46 4-(2-Methoxy-5-nitrophenyl)-2-furaldehyde
  • To a mixture of 2-iodo-4-nitroanisole (565 mg, 2.02 mmol) and 2-formylfuran-4-boronic acid pinacol ester (670 mg, 3.03 mmol) in 20 mL of ethylene glycol dimethyl ether and 12 mL of aqueous saturated sodium bicarbonate was added 80 mg of tetrakis(triphenylphosphine)palladium (0). The reaction mixture was heated at reflux overnight then allowed to cool to room temperature and added to a biphasic mixture of 10% methanol in ethyl acetate and water. The solid was collected by filtration, washed with ethyl acetate and water to provide 259 mg (52%) of 4-(2-methoxy-5-nitrophenyl)-2-furaldehyde as a light brown solid, 1H NMR (DMSO-d6) d 4.07 (s, 3H), 7.37 (d, J=9 Hz, 1H), 1H), 8.20-8.29 (m, 2H), 8.55 (d, J=3 Hz, 1H), 8.69 (s, 1H), 9.68 (s, 1H); MS 247.1 (M−H).
  • EXAMPLE 47 1-{[4-(2-Methoxy-5-nitrophenyl)-2-furyl]methyl}-4-methylpiperazine
  • A mixture of 4-(2-methoxy-5-nitrophenyl)-2-furaldehyde (230 mg, 0.93 mmol) and N-methylpiperazine (0.90 mL, 8.1 mmol) in 20 mL of dichloromethane and 2 mL of 1-methylpyrrolidinone was cooled to 0° C. Sodium triacetoxyborohydride (1.21 g, 5.7 mmol) was added in portions followed by a few drops of acetic acid. The resulting mixture was stirred at 0° C. for 10 minutes then at room temperature for 1.5 hours. The mixture was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate. The organic phase was dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography eluting with a gradient of 20% methanol in ethyl acetate to 1% ammonium hydroxide in 20% methanol in ethyl acetate. Trituration with diethyl ether and hexane provided 57 mg (18%) of 1-{[4-(2-methoxy-5-nitrophenyl)-2-furyl]methyl}-4-methylpiperazine as a light yellow solid, 1H NMR (DMSO-d6) δ 2.14 (s, 3H), 2.25-2.36 (broad s, 4H), 2.38-2.46 (broad s, 4H), 3.52 (s, 2H), 4.04 (s, 3H), 6.96 (s, 1H), 7.31 (d, J=8 Hz, 1H), 8.14-8.20 (m, 2H), 8.38 (d, J=3 Hz, 1H); MS 332.1 (M+H)+.
  • Analysis for C17H21N3O4:
  • Calcd: C, 61.62; H, 6.39; N, 12.68.
  • Found: C, 61.46; H, 6.68; N, 12.54.
  • EXAMPLE 48 (4-Methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amine
  • To a solution of 1-{[4-(2-methoxy-5-nitrophenyl)-2-furyl]methyl}-4-methylpiperazine (396 mg, 1.19 mmol) in 20 mL of methanol was added 10% palladium on carbon (40 mg). The resulting mixture was shaken with hydrogen in a Parr shaker until hydrogen uptake ceased. The reaction mixture was filtered through Celite and the filtrate concentrated in vacuo to give 358 mg (100%) of (4-methoxy-3-[5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amine as a brown oil, 1H NMR (DMSO-d6) δ 2.14 (s, 3H), 2.29 (broad s, 4H), 2.40 (broad s, 4H), 3.49 (s, 2H), 3.72 (s, 3H), 4.61 (s, 2H), 6.47 (d, J=8 Hz, 1H), 6.58 (s, 1H), 6.78 (d, J=9 Hz, 1H), 6.80 (s, 1H), 7.91 (s, 1H); MS 302.1 (M+H)+.
  • ALTERNATIVE EXAMPLE 48 (4-Methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amine
  • To a solution of 1-[(4-bromo-2-furyl)methyl]-4-methylpiperazine (1.43 g, 5.52 mmol) and tri-isopropyl borate (1.76 g, 9.38 mmol) in 20 mL of tetrahydrofuran at −78° C. was added 1.6 M n-butyl lithium in hexane (5.38 mL, 8.61 mmol) over 10 minutes. After stirring at −78° C. for 10 minutes, the temperature of the reaction mixture was allowed to rise to room temperature. Water (1.0 mL) was added and the solvents were removed in vacuo. The resultant intermediate boronic acid was dissolved in 20 mL of ethylene glycol dimethyl ether and 2-iodo-4-nitroanisole (1.06 mg, 4.25 mmol) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (1:1) (87 mg, 0.11 mol) were added. A solution of 2.25 g of sodium carbonate in 6.5 mL of water was added and the resulting mixture was heated at reflux for 3 hours. The mixture was cooled to room temperature and partitioned between ethyl acetate and water. The aqueous layer was further extracted with ethyl acetate and the organic layers were combined, washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography, eluting with a gradient of 5% methanol in dichloromethane to 10% methanol in dichloromethane to give 900 mg (70%) of (4-methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amine as a red oil.
  • EXAMPLE 49 2E/Z)-2-Cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amino]acrylamide
  • To a suspension of 2-cyano-N-(2,4-dichloro-5-methoxyphenyl)acetamide (261 mg, 1.01 mmol) in 5 mL of iso-propanol was added triethylorthoformate (0.504 mL, 3.03 mmol). The mixture was heated to reflux and (4-methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amine (320 mg, 1.06 mmol) in 9 mL of iso-propanol was added dropwise. This mixture was heated at reflux for 25 hours. The mixture was allowed to cool to room temperature and the solid was collected by filtration, washed with iso-propanol, to provide 465 mg (81%) of (2E/Z)-2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amino]acrylamide as a gray solid, mp 198-199° C.; MS 570.1 (M+H)+.
  • Analysis for C28H29Cl2N5O4-0.2H2O:
  • Calcd: C, 58.58; H, 5.16; N, 12.20.
  • Found: C, 58.50; H, 5.07; N, 12.07.
  • EXAMPLE 50 4-[(2,4-Dichloro-5-methoxyphenyl)amino]-6-methoxy-[7-[5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}-3-quinolinecarbonitrile
  • A suspension of (2E/Z)-2-cyano-N-(2,4-dichloro-5-methoxyphenyl)-3-[(4-methoxy-3-{5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}phenyl)amino]acrylamide (409 mg, 0.72 mmol) in 20 mL of butyronitrile was heated to 105° C. and phosphorous oxychloride (0.856 mL, 9.36 mmol) was added dropwise. After 14 hours additional phosphorous oxychloride (0.430 mL) was added, and the reaction mixture was heated at 105° C. for an additional 8 hours. Additional phosphorous oxychloride (0.430 mL) was added and the reaction temperature was increased to 115° C. The reaction mixture was heated at 115° C. for 18 hours. Additional phosphorous oxychloride (0.856 mL) was added and the reaction mixture was heated at 115° C. for an additional 26 hours. The reaction mixture was concentrated in vacuo and the residue suspended in a 1:1 mixture of ethyl acetate and tetrahydrofuran and neutralized with concentrated ammonium hydroxide. The mixture was stirred at room temperature for 1 hour. The solvents were removed in vacuo and the residue was purified by flash column chromatography eluting with a gradient of dichloromethane to 20% methanol in dichloromethane. Subsequent preparative thin layer chromatography developing with 10% methanol in dichloromethane gives 61 mg (15%) of 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-{7-[5-[(4-methylpiperazin-1-yl)methyl]-3-furyl}-3-quinolinecarbonitrile as a yellow solid, mp 174-176° C.
  • Calc'd: 552.15638
  • Found: 552.15723
  • HPLC: 96.8%

Claims (24)

1. A process for the preparation of a 4-amino-3-quinolinecarbonitrile comprising:
a. combining an amine compound with cyanoacetic acid and an acid catalyst to yield a cyanoacetamide;
b. condensing the cyanoacetamide of step a with an aniline, an alcoholic solvent, and a trialkylorthoformate to yield 3-amino-2-cyanoacrylamide; and
c. combining the 3-amino-2-cyanoacrylamide with phosphorus oxychloride in acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 4-amino-3-quinolinecarbonitrile.
2. The process of claim 1 wherein the ratio of cyanoacetic acid to amine is 1 to 1.5 equivalents.
3. The process of claim 1 wherein the ratio of cyanoacetic acid to amine is 1.03 equivalents.
4. The process of claim 1 wherein the aniline is a hydrochloride salt.
5. The process of claim 1 wherein the aniline is a free base.
6. The process of claim 1 wherein the alcoholic solvent is iso-propanol.
7. The process of claim 1 wherein the condensing step occurs at a temperature of 10-200° C.
8. The process of claim 1 wherein the condensing step occurs at a temperature of 20-140° C.
9. The process of claim 1 wherein the condensing step occurs at a temperature of 80° C.
10. The process of claim 1 wherein the catalyst is an alcohol.
11. The process of claim 1 wherein the catalyst is an amine base.
12. The process of claim 1 wherein the combining of step c occurs at a temperature of 50-200° C.
13. The process of claim 1 wherein the combining of step c occurs at a temperature of 80-110° C.
14. A process for the preparation of a 4-amino-3-quinolinecarbonitrile comprising:
a. combining an amine compound with cyanoacetic acid, a peptide coupling reagent to obtain a suspension;
b. filtering the suspension in step a to yield a cyanoacetamide;
c. condensing the cyanoacetamide of step b with an aniline, an alcoholic solvent, and a trialkylorthoformate to yield 3-amino-2-cyanoacrylamide; and
d. combining the 3-amino-2-cyanoacrylamide in step c with phosphorus oxychloride to yield a 4-amino-3-quinolinecarbonitrile.
15. The process of claim 14 wherein the ratio of cyanoacetic acid to amine is 1 to 1.5 equivalents.
16. The process of claim 14 wherein the ratio of cyanoacetic acid to amine is 1.03 equivalents.
17. The process of claim 14 wherein the peptide coupling reagent is water soluble.
18. The process of claim 14 wherein the peptide coupling reagent is 1,3-diisopropylcarbodiimide.
19. The process of claim 14 wherein said process occurs at a temperature of 50-100° C.
20. The process of claim 14 wherein said process occurs at a temperature of 77-80° C.
21. The process of claim 14 wherein the amine is pyridine, triethylamine, and diisopropylamine.
22. A process for the preparation of a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile comprising:
a. combining a 3-amino thiophene with cyanoacetamide and a trialkylorthoformate in an alcoholic solvent to obtain a propenamide;
b. combining the propenamide with phosphorus oxychloride and acetonitrile, butyronitrile, toluene or xylene, optionally in the presence of a catalyst to yield a 7-amino-thieno[3,2-b]pyridine-6-carbonitrile.
23. A process for the preparation of a cyanoacetamide comprising:
a. combining DMF, an aniline and cyanoacetic acid to yield a mixture;
b. cooling the mixture;
c. adding a solution of N,N′ dicyclohexylcarbodiimide in DMF to keep the temperature below 15° C. and yield a suspension;
d. filtering the suspension and washing a resulting solid by-product to yield a filtrate;
e. adding water to the filtrate of step d to obtain a suspension, cooling to 5° C.; and
f. filtering the suspension to yield a cyanoacetamide.
24. The process of claim 23 wherein the by-product is urea.
US11/973,259 2003-08-19 2007-10-05 Process for preparation of 4-amino-3-quinolinecarbonitriles Abandoned US20080033175A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/973,259 US20080033175A1 (en) 2003-08-19 2007-10-05 Process for preparation of 4-amino-3-quinolinecarbonitriles

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US49619103P 2003-08-19 2003-08-19
US10/918,947 US7297795B2 (en) 2003-08-19 2004-08-16 Process for preparation of 4-amino-3-quinolinecarbonitriles
US11/973,259 US20080033175A1 (en) 2003-08-19 2007-10-05 Process for preparation of 4-amino-3-quinolinecarbonitriles

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/918,947 Division US7297795B2 (en) 2003-08-19 2004-08-16 Process for preparation of 4-amino-3-quinolinecarbonitriles

Publications (1)

Publication Number Publication Date
US20080033175A1 true US20080033175A1 (en) 2008-02-07

Family

ID=34215972

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/918,947 Expired - Fee Related US7297795B2 (en) 2003-08-19 2004-08-16 Process for preparation of 4-amino-3-quinolinecarbonitriles
US11/973,259 Abandoned US20080033175A1 (en) 2003-08-19 2007-10-05 Process for preparation of 4-amino-3-quinolinecarbonitriles

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/918,947 Expired - Fee Related US7297795B2 (en) 2003-08-19 2004-08-16 Process for preparation of 4-amino-3-quinolinecarbonitriles

Country Status (25)

Country Link
US (2) US7297795B2 (en)
EP (1) EP1660453A2 (en)
JP (1) JP2007502819A (en)
KR (1) KR20060066733A (en)
CN (1) CN1835923A (en)
AR (1) AR045277A1 (en)
AU (1) AU2004267061A1 (en)
BR (1) BRPI0413667A (en)
CA (1) CA2535385A1 (en)
CO (1) CO5670369A2 (en)
CR (1) CR8226A (en)
EC (1) ECSP066384A (en)
GT (1) GT200400165A (en)
IL (1) IL173456A0 (en)
MX (1) MXPA06001590A (en)
MY (1) MY140542A (en)
NO (1) NO20060539L (en)
PA (1) PA8609301A1 (en)
PE (1) PE20050344A1 (en)
RU (1) RU2345069C2 (en)
SA (1) SA04250258B1 (en)
SG (1) SG145744A1 (en)
SV (1) SV2005001855A (en)
TW (1) TW200517378A (en)
WO (1) WO2005019201A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9421203B2 (en) 2012-06-15 2016-08-23 Institute For Cancer Research Sensitization of cancer cells to DNA damage by inhibiting kinases essential for DNA damage checkpoint control
WO2019186429A1 (en) * 2018-03-30 2019-10-03 Sun Pharmaceutical Industries Limited A process for the preparation of bosutinib

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7479561B2 (en) * 2004-08-16 2009-01-20 Wyeth 4-(2,4-dichloro-5-methoxyphenyl)amino-6-methoxy-7-{[5-substituted-amino)methyl]-3-furyl}-3-quinolinecarbonitriles as kinase inhibitors
CN101155579B (en) 2005-02-03 2012-10-31 综合医院公司 Method for treating gefitinib resistant cancer
BRPI0609928A2 (en) * 2005-05-18 2010-05-11 Wyeth Corp 3-cyanoquinoline tpl-2 kinase inhibitors and methods of manufacture and use thereof
BRPI0610144A2 (en) * 2005-05-25 2010-06-01 Wyeth Corp method of preparing a substituted 3-cyanoquinoline; compound represented by formula i; and compound represented by formula ii
EP1883631A1 (en) * 2005-05-25 2008-02-06 Wyeth Methods of synthesizing substituted 3-cyanoquinolines and intermediates thereof
KR20080016604A (en) * 2005-05-25 2008-02-21 와이어쓰 Methods of synthesizing 6-alkylaminoquinoline derivatives
RU2007148072A (en) 2005-07-01 2009-08-10 Вайет (Us) CRYSTAL FORMS 4 - [(2,4-DICHLOR-5-METHOXYPHINYL) AMINO] -6-MEOXY-7- [3- (4-METHYL-1-PIPERAZINYL) PROPOXY] -3-HINALINCARBONITRIL AND METHODS
CN103110948A (en) 2005-11-04 2013-05-22 惠氏公司 Antineoplastic combinations with mTOR inhibitor,herceptin, and/or HKI-272
JP2009526048A (en) * 2006-02-08 2009-07-16 ワイス Preparation of 7-alkenyl-3quinolinecarbonitrile via palladium mediated coupling reaction
CN101195578B (en) * 2006-12-08 2012-04-25 上海睿智化学研究有限公司 Method for synthesizing 1-(halogen alkoxyl)-2-alkoxyl-5-nitrobenzene compounds
US8022216B2 (en) 2007-10-17 2011-09-20 Wyeth Llc Maleate salts of (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide and crystalline forms thereof
KR20130088908A (en) 2008-06-17 2013-08-08 와이어쓰 엘엘씨 Antineoplastic combinations containing hki-272 and vinorelbine
CN109464445A (en) * 2008-08-04 2019-03-15 惠氏有限责任公司 The antineoplastic combinations of 4- anilino- -3- cyano quinolines and capecitabine
WO2010030835A2 (en) * 2008-09-11 2010-03-18 Wyeth Llc Pharmaceutical compositions of an src kinase inhibitor and an aromatase inhibitor
ES2941894T3 (en) 2009-04-06 2023-05-26 Wyeth Llc Treatment regimen using neratinib for breast cancer
CN102470109A (en) 2009-07-02 2012-05-23 惠氏有限责任公司 3-cyanoquinoline tablet formulations and uses thereof
CN101792416B (en) * 2010-01-15 2011-10-05 南京医科大学 Process for preparing bosutinib
SG186378A1 (en) * 2010-06-23 2013-01-30 Hanmi Science Co Ltd Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
CN102952037A (en) * 2012-11-18 2013-03-06 大连九信生物化工科技有限公司 Synthesis method of 2-cyano-N-(2, 4-dichloro-5-methoxyphenyl) acetamide
IN2014CH00840A (en) * 2014-02-20 2015-09-18 Apotex Inc
CN104876865A (en) * 2014-02-27 2015-09-02 南京正荣医药化学有限公司 Preparation technology of bosutinib
KR102082331B1 (en) 2015-07-06 2020-02-27 길리애드 사이언시즈, 인코포레이티드 6-amino-quinoline-3-carbonitrile as Cot modifier
PE20180462A1 (en) 2015-07-06 2018-03-06 Gilead Sciences Inc TOC MODULATORS AND METHODS OF USING THEM
CN105085398A (en) * 2015-09-06 2015-11-25 合肥华方医药科技有限公司 Preparation method of bosutinib isomer impurities
CN105646345A (en) * 2016-03-16 2016-06-08 浙江海正药业股份有限公司 Novel crystal forms of bosutinib and preparation method thereof
ES2800339T3 (en) 2016-06-30 2020-12-29 Gilead Sciences Inc 4,6-diaminoquinazolines as cradle modulators and methods of using them
CN110770231B (en) * 2017-10-18 2022-11-22 江苏恒瑞医药股份有限公司 Preparation method of tyrosine kinase inhibitor and intermediate thereof
CN108084085B (en) * 2017-12-28 2019-11-26 山东铂源药业有限公司 A kind of preparation method of N- (the chloro- 4- of 3- (2- pyridomethoxy) phenyl) -2- cyanoacetamide
CN109180578B (en) * 2018-10-19 2020-06-26 山东创新药物研发有限公司 Preparation method of bosutinib
CN111646939A (en) * 2019-03-04 2020-09-11 鲁南制药集团股份有限公司 Preparation method of bosutinib
CN111646955B (en) * 2019-03-04 2024-05-24 鲁南制药集团股份有限公司 Method for preparing bosutinib intermediate
TWI770527B (en) 2019-06-14 2022-07-11 美商基利科學股份有限公司 Cot modulators and methods of use thereof
CN115397511A (en) 2020-03-30 2022-11-25 吉利德科学公司 Solid forms of the COT inhibitor compound (S) -6- (((1- (bicyclo [1.1.1] pent-1-yl) -1H-1,2, 3-triazol-4-yl) 2-methyl-1-oxo-1, 2-dihydroisoquinolin-5-yl) methyl))) amino 8-chloro- (neopentylamino) quinoline-3-carbonitrile
EP4126862A1 (en) 2020-04-02 2023-02-08 Gilead Sciences, Inc. Process for preparing a cot inhibitor compound

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA73073C2 (en) * 1997-04-03 2005-06-15 Уайт Холдінгз Корпорейшн Substituted 3-cyan chinolines

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9421203B2 (en) 2012-06-15 2016-08-23 Institute For Cancer Research Sensitization of cancer cells to DNA damage by inhibiting kinases essential for DNA damage checkpoint control
WO2019186429A1 (en) * 2018-03-30 2019-10-03 Sun Pharmaceutical Industries Limited A process for the preparation of bosutinib

Also Published As

Publication number Publication date
NO20060539L (en) 2006-02-13
SA04250258B1 (en) 2009-01-07
US7297795B2 (en) 2007-11-20
AR045277A1 (en) 2005-10-19
CN1835923A (en) 2006-09-20
EP1660453A2 (en) 2006-05-31
SV2005001855A (en) 2005-11-04
ECSP066384A (en) 2006-08-30
RU2345069C2 (en) 2009-01-27
IL173456A0 (en) 2006-06-11
JP2007502819A (en) 2007-02-15
US20050043537A1 (en) 2005-02-24
MY140542A (en) 2009-12-31
MXPA06001590A (en) 2006-05-19
SG145744A1 (en) 2008-09-29
RU2006108524A (en) 2007-09-27
CO5670369A2 (en) 2006-08-31
KR20060066733A (en) 2006-06-16
CR8226A (en) 2008-08-21
BRPI0413667A (en) 2006-10-24
WO2005019201A2 (en) 2005-03-03
TW200517378A (en) 2005-06-01
AU2004267061A1 (en) 2005-03-03
GT200400165A (en) 2005-04-12
PE20050344A1 (en) 2005-06-19
CA2535385A1 (en) 2005-03-03
PA8609301A1 (en) 2005-03-03
WO2005019201A3 (en) 2005-08-04

Similar Documents

Publication Publication Date Title
US7297795B2 (en) Process for preparation of 4-amino-3-quinolinecarbonitriles
US7977337B2 (en) Quinoline derivatives and their use as 5-HT6 ligands
RU2309149C2 (en) Method for production of 7-substituted-3-quinoline and 3-quinol-4-on carbonitryles
WO2010036831A1 (en) Preparation of a quinolinyloxydiphenylcyclopropanedicarboxamide
WO2011082098A1 (en) Lysine and arginine methyltransferase inhibitors for treating cancer
CN110498770B (en) Method for preparing intermediate of oxaagolide
KR101908333B1 (en) Naphthylamide compound, preparation method and use thereof
CA2060107A1 (en) Nitrogen heterocycles
IL182439A (en) Intermediates useful for the preparation of aripiprazole and methods for the preparation of the intermediates and aripiprazole
NO147838B (en) INTERMEDIATE FOR USE IN PREPARATION OF THE HYPOTENSIVE AGENT 2- (4- (2-FUROYL) PIPERAZIN-1-YL) -4-AMINO-6,7-DIMETOXYKINAZOLINE
KR101041277B1 (en) Process for producing quinolonecarboxylic acid derivative
AU2007212756A1 (en) Preparation of 7-alkenyl-3 quinolinecarbonitriles via a palladium mediated coupling reaction
SU999967A3 (en) Process for producing 6-n-substituted 6-amino-3-pyridazinyl hydrizines or their salts
CN109988172B (en) Synthesis method of pyrazolo [1,5-A ] pyrimidine heterocyclic compound and derivative
KR810000293B1 (en) Process for preparing substituted amino quinazoline derivatives
KR800001685B1 (en) Process for preparing pyrido (2,3-d)pyrimidine derivatives
JP6084569B2 (en) Process for producing 3,4-dihydroisoquinoline derivatives and production intermediates thereof
KR20110129622A (en) Preparation method of azosemide
CN111646939A (en) Preparation method of bosutinib
JPS628114B2 (en)
JPH01110676A (en) Production of quinazolinone compound
CZ281171B6 (en) Process for preparing spiroisoquinoline pyrrolidine derivatives and intermediate for making the same
CS202071B2 (en) Method of preparing 2-/4-substituted piperazin-1-yl/-4-amino-6,7-dimethoxyquinazolines
JPS6160831B2 (en)
FR2632305A1 (en) 4-Amino-3-aroylquinolines and -naphthyridines, process for their preparation and their application in therapeutics

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION