CYANOQUINOLINE DERIVATIVES

Abstract
Disclosed is a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,
Description
FIELD

The present application is directed to organic chemistry and pharmaceutical chemistry.


BACKGROUND

In worldwide, the tumor, including cancer, is one of the major factors leading to death. Although there is a notable development in the discovery of novel methods for treating tumor, the major selections for therapy are still the surgery operation, chemotherapy and radiotherapy. The three therapeutic treatment methods can be used alone or in combination. However, the surgery operation and radiotherapy generally are useful for patients, of which the tumor type has been identified. There are limits of the surgery operation and radiotherapy for treating patients, of which the tumor has spread. The chemotherapy is generally useful for treating patients having metastatic cancer or diffuse carcinoma, such as leukemia. Although the chemotherapy has therapeutic values, it usually does not cure the diseases because cancer cells of patients are tolerant to the drugs of chemotherapy.


Therefore, there is a need for novel chemotherapeutants to treat tumor. In this regard, different researchers are doing continuous efforts to discover novel potential effective chemotherapeutic drugs.


SUMMARY

In one aspect, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In another aspect, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, C2-C6 alkynylacylamino, C6-C18 arylacylamino, C1-C6 alkyl-substituted amino, C1-C6 alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, C7-C24 aralkyl, substituted or unsubstituted C6-C15 aryl, substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, C6-C18 aryloxy, and C5-C18 heteroaryloxy, and a substituent on the heteroaryl is selected from C7-C24 aralkyl, C6-C18 arylacylamino, C6-C18 arylsulfonylamino, C5-C18 heteroarylacylamino, C3-C10 cycloalkylacylamino, C6-C18 arylaminoacyl, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl; and


R4 is C3-C18 heterocyclyl or C5-C18 heteroaryl.


In another aspect, the present application is directed to a pharmaceutical composition, comprising a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or prodrug thereof, and a pharmaceutically acceptable carrier.


In yet another aspect, the present application is directed to a method for treating and/or preventing a tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof.


In another aspect, the present application is directed to a method for inhibiting growth of tumor cells, comprising contacting the tumor cells with a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof.


In still another aspect, the present application is directed to a method for inhibiting overexpression or mutantion of a receptor tyrosine kinase in a mammal, comprising contacting the receptor tyrosine kinase with a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof.


In still yet another aspect, the present application is directed to a method for treating and/or preventing physiological abnormity caused by overexpression or mutation of a receptor tyrosine kinase in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof.







DETAILED DESCRIPTION
Definitions

Certain chemical groups named herein are preceded by a shorthand notation indicating the total number of carbon atoms that are to be found in the indicated chemical group. For example, C7-C12 alkyl describes an alkyl group, as defined below, having a total of 7 to 12 carbon atoms, and C3-C10 cycloalkyl describes a cycloaklyl group, as defined below, having a total of 3 to 10 carbon atoms. The total number of carbon atoms in the shorthand notation does not include the carbons that may exist in the substituents of the groups described.


Furthermore, as used in the specification and appended claims of the present application, unless specified to the contrary, the following terms have the meanings indicated:


“Amino” refers to the —NH2 group. The amino group may be substituted with a group selected from alkyl, alkenyl, alkynyl, haloalkyl, haloalkenyl, haloalkynyl, hydroxyalkyl, aralkyl, cycloalkyl, cycloalkylalkyl, and the like.


“Cyano” refers to the —CN group.


“Hydroxy” refers to the —OH group.


“Imino” refers to ═NH substituent.


“Nitro” refers to the —NO2 group.


“Oxo” refers to ═O substituent.


“Thio” refers to ═S substituent.


“Trifluoromethyl” refers to the —CF3 group.


“Alkyl” refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen, containing no unsaturation, having from one to twelve carbon atoms, preferably one to eight or one to six carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (tert-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted by one or more groups independently selected from the group consisting of alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilyl, —OR14, —OC(O)—R14, —N(R14)2, —C(O)R14, —C(O)OR14, —C(O)N(R14)2, —N(R14)C(O)OR16, —N(R14)C(O)R16, —N(R14)S(O)tR16 (wherein t is 1 to 2), —S(O)tOR16 (wherein t is 1 to 2), —S(O)tR16 (wherein t is 0 to 2), and —S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the alkyl group is C1-C12 alkyl.


In some embodiments, the alkyl group is C1-C5 alkyl.


In some embodiments, the alkyl group is C1-C6 alkyl.


“Alkenyl” refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having two to twelve carbon atoms, preferably two to six carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group may be optionally substituted with one or more groups independently selected from the group consisting of alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilyl, —OR14, —OC(O)—R14, —N(R14)2, —C(O)R14, —C(O)OR14, —C(O)N(R14)2, —N(R14)C(O)OR16, —N(R14)C(O)R16, —N(R14)S(O)tR16 (wherein t is 1 to 2), —S(O)tOR16 (wherein t is 1 to 2), —S(O)tR16 (wherein t is 0 to 2), and —S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the alkenyl group is C2-C12 alkenyl.


In some embodiments, the alkenyl group is C2-C8 alkenyl.


In some embodiments, the alkenyl group is C2-C6 alkenyl.


“Alkynyl” refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having two to twelve carbon atoms, preferably two to six carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group may be optionally substituted with one or more groups independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroaralkyl, —OR″, —OC(O)—R14, —N(R14)2, —C(O)R14, —C(O)OR14, —C(O)N(R14)2, —N(R14)C(O)OR16, —N(R14)C(O)R16, —N(R14)S(O)tR16 (wherein t is 1 to 2), —S(O)tOR16 (wherein t is 1 to 2), —S(O)tR16 (wherein t is 0 to 2), and —S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the alkynyl group is C2-C12 alkynyl.


In some embodiments, the alkynyl group is C2-C8 alkynyl.


In some embodiments, the alkynyl group is C2-C6 alkynyl.


“Alkylene” or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain, linking the rest of the molecule of a group, consisting solely of carbon and hydrogen, containing no unsaturation and having one to twelve carbon atoms, e.g., methylene, ethylidene, propylidene, n-butylidene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and other goups through a single bond. The alkylene chain may be attached to the rest of the molecule and to the group through one carbon within the chain or through any two carbons within the chain. Unless stated otherwise specifically in the specification, the alkylidene chain may be optionally substituted with one or more groups independently selected from the group consisting of alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilyl, —OR14, —OC(O)—R14, —N(R14)2, —C(O)R14, —C(O)OR14, —C(O)N(R14)2, —N(R14)C(O)OR16, —N(R14)C(O)R16, —N(R14)S(O)tR16 (wherein t is 1 to 2), —S(O)tOR16 (wherein t is 1 to 2), —S(O)tR16 (wherein t is 0 to 2), and —S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the alkylene group is C1-C12 alkylidene.


In some embodiments, the alkylene group is C1-C8 alkylidene.


In some embodiments, the alkylene group is C1-C6 alkylidene.


“Alkenylene” or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a group, consisting solely of carbon and hydrogen, containing at least one double bond and having two to twelve carbon atoms, e.g., such as ethenylene, propenylene, n-butenylene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to other groups through a double bond or a single bond. The points of attachments of the alkenylene to the rest of the molecule and to other groups can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, the alkenylene may be optionally substituted with one or more groups independently selected from the group consisting of alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilyl, —OR″, —OC(O)—R14, —N(R14)2, —C(O)R14, —C(O)OR14, —C(O)N(R14)2, —N(R14)C(O)OR16, —N(R14)C(O)R16 (O)tR16 (wherein t is 1 to 2), —S(O)tOR16 (wherein t is 1 to 2), —S(O)tR16 (wherein t is 0 to 2), and —S(O)N(R14)2 (wherein t is 1 to 2).


In some embodiments, the alkenylene group is C2-C12 alkenylidene.


In some embodiments, the alkenylene group is C2-C8 alkenylidene.


In some embodiments, the alkenylene group is C2-C6 alkenylidene.


“Alkoxy” refers to a group of the formula —ORa, where Ra is an alkyl group as defined above. The alkoxy group contains one to twelve carbon atoms, preferably one to six carbon atoms. The alkyl part of the alkoxy group may be optionally substituted as defined above for an alkyl group.


“Alkoxylalkyl” refers to a group of the formula —Ra—O—Ra, where each Ra is independently an alkyl group as defined above. The oxygen atom may be bonded to any carbon in either alkyl group. Each alkyl part of the alkoxylalkyl group may be optionally substituted as defined above for an alkyl group.


“Aryl” refers to aromatic monocyclic or multicyclic hydrocarbon ring system consisting solely of hydrogen and carbon and containing from six to eighteen carbon atoms, where the ring system may be partially saturated. Aryl groups include, but are not limited to the groups such as phenyl, naphthyl and fluorenyl. Unless stated otherwise specifically in the specification, the term “aryl” or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl groups optionally substituted with one or more substituents independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, heteroaryl, heteroaralkyl, —R15—OR14, —R15—OC(O)—R14, —R15—N(R14)2, —R15—C(O)R14, —R15—C(O)OR14, —R15—C(O)N(R14)2, —R15—N(R14)C(O)OR16, —R15—N(R14)C(O)R16, —R15—N(R14)S(O)tR16 (wherein t is 1 to 2), —R15—S(O)tOR16 (wherein t is 1 to 2), —R15—S(O)tR16 (wherein t is 0 to 2), and —R15—S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the aryl group is C6-C18 aryl.


In some embodiments, the aryl group is C6-C12 aryl.


In some embodiments, the aryl group is C5-C10 aryl.


“Aralkyl” refers to a group of the formula —RaRb, where Ra is an alkyl group as defined above and Rb is one or more aryl groups as defined above, e.g., benzyl, diphenylmethyl, and the like. The aryl part may be optionally substituted as described above.


“Aryloxy” refers to a group of the formula —ORb, where Rb is an aryl group as defined above. The aryl part of the aryloxy group may be optionally substituted as defined above.


“Aralkyloxy” refers to a group of the formula —ORe, where Re is an aralkyl group as defined above. The aralkyl part of the aralkyloxy group may be optionally substituted as defined above.


“Cycloalkyl” refers to a stable non-aromatic monocyclic or multicyclic hydrocarbon group consisting solely of carbon and hydrogen atoms, including fused or bridged ring systems and having three to eighteen carbon atoms, preferably three to fifteen carbon atoms, preferably three to ten carbon atoms, and which is saturated or unsaturated and attatched to the rest of the molecule by a single bond. The monocyclic group comprises, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. The multicyclic group comprises, e.g., adamantanyl, norcamphanyl, decalinyl, 7,7-dimethyl-biscyclo[2.2.1]heptyl, and the like. Unless stated otherwise specifically in the specification, the term “cycloalkyl” is meant to include cycloalkyl groups which are optionally substituted with one or more substituents independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, oxo, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycylylalkyl, heteroaryl, heteroaralkyl, —R15—OR14, —R15—OC(O)R14, —R15—N(R14)2, —R15—C(O)R14, —R15—C(O)OR14, —R15—C(O)N(R14)2, —R15—N(R14)C(O)OR16, —R15—N(R14)C(O)R16, —R15—N(R14)S(O)tR16 (wherein t is 1 to 2), —R15—S(O)tR16 (wherein t is 1 to 2), —R15—S(O)tR16 (wherein t is 0 to 2), and —R15—S(O)N(R14)2 (wherein t is 1 to 2).


In some embodiments, the cycloalkyl group is C3-C18 cycloalkyl.


In some embodiments, the cycloalkyl group is C3-C15 cycloalkyl.


In some embodiments, the cycloalkyl group is C3-C10 cycloalkyl.


“Cycloalkylalkyl” refers to a group of the formula-RaRd, where Ra is an alkyl group as defined above, and Rd is a cycloalkyl group as defined above. The alkyl part and cycloalkyl part may be optionally substituted as defined above.


“Halo” refers to bromo, chloro, fluoro or iodo.


“Haloalkyl” refers to an alkyl group, as defined above, that is substituted by one or more halo groups, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like. The alkyl part of the haloalkyl may be optionally substituted as defined above for an alkyl group.


“Heterocyclyl” refers to a stable 3- to 18-membered non-aromatic ring group, which contains three to eighteen carbon atoms and one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulphur. Unless stated otherwise specifically in the specification, the heterocyclyl group may be monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Moreover, the nitrogen, carbon or sulphur atom in the heterocyclyl group may be optionally oxidized, and the nitrogen atom may be optionally quaternized. The heterocyclyl group may be partially or fully saturated. Examples of such heteroyclyl groups include, but are not limited to, dioxolanyl, thiophene[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, the term “heterocyclyl” is meant to include the heterocyclyl groups which may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, alkoxy, cyano, oxo, thio, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycylylalkyl, heteroaryl, heteroaralkyl, —R15—OR14, —R15—OC(O)—R14, —R15—N(R14)2, —R15—C(O)R14, —R15—C(O)OR14, —R15—C(O)N(R14)2, —R15—N(R14)C(O)OR16, —R15—N(R14)C(O)R16 (wherein t is 1 to 2), —R15—S(O)tOR16 (wherein t is 1 to 2), —R15—S(O)tR16 (wherein t is 0 to 2), and —R15—S(O)N(R14)2 (wherein t is 1 to 2).


In some embodiments, the heterocyclyl group is C3-C18 heterocyclyl.


In some embodiments, the heterocyclyl group is C3-C12 heterocyclyl.


In some embodiments, the heterocyclyl group is C3-C10 heterocyclyl.


“Heterocyclylalkyl” refers to a group of the formula-RaRe, where Re is an alkyl group as defined above, and Re is a heterocyclyl group as defined above. Moreover, if the heterocyclyl is a nitrogen-containing heterocyclyl, then the heterocyclyl may be attached to the alkyl group at the nitrogen atom. The alkyl part of the heterocyclylalkyl group may be optionally substituted as defined above for an alkyl group. The heterocyclyl part of the heterocyclylalkyl group may be optionally substituted as defined above for a heterocyclyl group.


“Heteroaryl” refers to a 5- to 18-membered aromatic ring group, which contains one to seventeen carbon atoms and one to ten heteroatoms selected from the group consisting of nitrogen, oxygen and sulphur. For the purpose of the present invention, the heteroaryl group may be monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Moreover, the nitrogen, carbon or sulphur atom in the heteroaryl group may be optionally oxidized, and the nitrogen atom may be optionally quaternized. The examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzoindolyl, benzodioxolanyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepanyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzodioxolanyl, benzodioxadienyl, benzopyranyl, benzopyronyl, benzofuranyl, benzofuranonyl, benzothienyl, benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothienyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, 2,3-naphthyridinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolyl, quinuclidinyl, isoquinolyl, tetrahydroquinolyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl and thiophenyl. Unless stated otherwise specifically in the specification, the term “heteroaryl” is meant to include the heteroaryl groups which may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl, alkenyl, alkoxy, halo, haloalkyl, haloalkenyl, cyano, oxo, thio, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycylylalkyl, heteroaryl, heteroaralkyl, —R15—OR14, —R15—OC(O)—R14, —R15—N(R14)2, —R15—C(O)R14, —R15—C(O)OR14, —R15—C(O)N(R14)2, —R15—N(R14)C(O)OR16, —R15—N(R14)C(O)R16, —R15—N(R14)S(O)tR16 (wherein t is 1 to 2), —R15—S(O)tOR16 (wherein t is 1 to 2), —R15—S(O)tR16 (wherein t is 0 to 2), and —R15—S(O)tN(R14)2 (wherein t is 1 to 2).


In some embodiments, the heteroaryl group is C5-C18 heteroaryl.


In some embodiments, the heteroaryl group is C5-C12 heteroaryl.


In some embodiments, the heteroaryl group is C5-C10 heteroaryl.


“Heteroarylalkyl” refers to a group of the formula —RaRf, where Ra is an alkyl group as defined above, and Rf is a heteroaryl group as defined above. The heteroaryl part of the heteroarylalkyl group may be optionally substituted as defined above for a heteroaryl group. The alkyl part of the heteroarylalkyl may be optionally substituted as defined above for an alkyl group.


In definition of groups as described above, each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl optionally substituted with one or more halogens, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroaralkyl; each R15 is independently a direct bond or straight or branched alkylene or alkenylene chain; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteraryl, or heteroaralkyl.


“Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the specification includes instances where said event or circumstance occurs and instances in which it does not. For example, “optionally substituted aryl” means that the aryl may or may not be substituted and that the specification includes the substituted aryl and the aryl which is not substituted.


“Stable compound” and “stable structure” are meant to indicate a compound that is sufficient robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an effecacious therapeutic agent.


“Pharmaceutically acceptable salt” includes both acid and base addition salt.


“Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphanic acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleinic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid and the like.


“Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base into the free acid. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum slats, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, slats of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucosamine, theobromine, triethanolamine, trometamol, purine, piperazine, piperidine, N-ethyl piperidine, polyamine resin and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.


Often crystallization produces a solvate of the compound of the invention. As used herein, the term “solvate” refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be hydrate. Alternatively, the solvent may be an organic solvent. Therefore, the compounds of the present invention may exist as a hydrate, including monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.


An intermediate of a compound of the formula I and all polymorphs and crystal habits of the foregoing species are also included whitin the scope of the present invention.


The compounds of the invention or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may therefore produce enantiomers, diastereoisomers and other stereoisomeric forms, that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or, as (D)- or (L)-for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (−), (R)- and (S)-, or (D)- and (L)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as chromatography and fractional crystallization. The conventional techniques for preparing/isolating a single enantiomer include chiral synthesis from a suitable optically pure precursor, or resolution of a racemate (or the racemate of a salt or derivative) by using such as chiral High Pressure Liquid Chromatography (HPLC). When the compounds described herein comprise olefinic double bonds or other centers of geometric asymmetry, unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.


A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomer”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror image of one another.


A “cis-trans-isomer” refers to a molecule having the molecular formula, in which a spacial configuration of different relative distance between the adjacent atoms or radicals exists due to the factors such as the presence of a double bond or a ring, which block the free rotation of a bond.


A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tautomers of any said compounds.


The term “prodrug” is meant to indicate a compound that may be converted into a biologically active compound of the invention under physiological conditions or by solvolysis. Therefore, the term “prodrug” refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administrated to a subject in need thereof, but is converted in vivo into an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the present invention, for example, by hydrolysis in blood. The prodrug compound often provides advantages of solubility, tissue compatibility or controlled-release in organism of mammals (see Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)). A discussion of prodrugs is provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems”, A.C.S. Symposium Series, Vol. 14 and Bioreversible Carriers in Drug Design, Ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.


The term “prodrug” is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is administrated to a mammal subject Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino, or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammal subject, cleaves to form a free hydroxy, free amino or free mecapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate of alcohol functional group in the compounds of the present invention and the like.


The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidization, reduction, hydrolysis, amidation, esterification, and the like of the administered compounds, primarily due to enzyme processes. Therefore, the invention includes compounds produced by a process comprising contacting a compound of the invention with a mammal for a period time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabelled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time to occur, and isolating its conversion products from urine, blood or other biological samples.


“Mammal” includes humans and both domestic animals, such as laboratory animals and household pets (e.g. cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.


The term “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally acceptable in the art for the delivery of the biologically active compound to a mammal e.g. humans. Such a medium includes all pharmaceutically acceptable carriers for use. The pharmaceutical composition is conducive to administration of a compound to an organism. There are various routes of administration of a compound in the art including, but not limited to oral administration, injection administration, aerosol administration, parenteral administration and topical administration. The pharmaceutical compositions can also be obtained by reacting a compound with an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like, or with an organic acid.


The term “carrier” defines a compound that facilitates the incorporation of a compound into cells or tissues. For example, dimethylsulfoxide (DMSO) is generally used as a carrier, as it facilitates the uptake of many organic compounds into cells or tissues of an organism.


“Pharmaceutically acceptable carrier” includes without limitation to any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isosmotic agent, solvent, or emulsifier which has been approved by the national drug regulatory authorities as being acceptable for use in humans or domestic animals.


The term “physiologically acceptable” defines a carrier or a diluent that does not abrogate the biological activities and properties of a compound.


“Therapeutically effective amount” refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of the tumor in the mammal, preferably a human. The amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the condition and its severity, the administration mode and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.


“Treating” or “treatment” as used herein covers the treatment of the tumor in a mammal, preferably a human, having the tumor, and includes:


(i) preventing the tumor from occurring in a mammal, in particular, when such mammal is predisposed to the tumor but has not yet been diagnosed as having it;


(ii) inhibiting the tumor, i.e. arresting its development; or


(iii) relieving the tumor, i.e. causing regression of the tumor; or


(iv) relieving the symptoms caused by the tumor.


Specific Embodiments

In one aspect, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In another aspect, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, C2-C6 alkynylacylamino, C6-C18 arylacylamino, C1-C5 alkyl-substituted amino, and C1-C6 alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, C7-C24 aralkyl, substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, C6-C18 aryloxy, and C5-C18 heteroaryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyl, C6-C18 arylacylamino, C6-C18 arylsulfonylamino, C5-C18 heteroarylacylamino, C3-C10 cycloalkylacylamino, C6-C18 arylaminoacyl, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl; and


R4 is C3-C18 heterocyclyl or C5-C18 heteroaryl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or prodrug thereof,


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, C2-C6 alkynylacylamino, C8-C18 arylacylamino, C1-C6 alkyl-substituted amino, and C1-C6 alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, C7-C24 aralkyl, substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, C6-C18 aryloxy, and C5-C18 heteroaryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyl, C6-C18 arylacylamino, C6-C18 arylsulfonylamino, C5-C18 heteroarylacylamino, C3-C10 cycloalkylacylamino, C6-C18 arylaminoacyl, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, C2-C6 alkynylacylamino, C6-C18 arylacylamino, C1-C6 alkyl-substituted amino, and C1-C6 alkoxy;


one of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, C6-C18 aryloxy, and C5-C18 heteroaryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyl, C6-C18 arylacylamino, C6-C18 arylsulfonylamino, C5-C18 heteroarylacylamino, C3-C10 cycloalkylacylamino, C6-C18 arylaminoacyl, C7-C24 aralkyloxy, C6-C18 heteroaralkyloxy, and C6-C18 aryloxy; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, and C6-C18 arylacylamino;


one of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, and substituted or unsubstituted C6-C18 aryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C18 aryloxy, C6-C24 heteroaryloxy, and C6-C24 heteroaralkyloxy; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is C2-C6 alkenylacylamino;


one of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, and substituted or unsubstituted C6-C15 aryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C24 heteroaralkyloxy; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is selected from the group consisting of 4-(dimethylamino)-but-2-enamido, 4-(diethylamino)-but-2-enamido, 4-(piperidin-1-yl)-but-2-enamido, 4-(morpholin-4-yl)-but-2-enamido, 4-(tert-butylamino)-but-2-enamido, 4-(benzylamino)-but-2-enamido, 4-(6-hydroxyhexylamino)-but-2-enamido, 4-(2-methoxylethylamino)-but-2-enamido, 2-(piperidin-4-ylidene)acetamido, 2-(1-methylpiperidin-4-ylidene)acetamido, 4-(diethanolamino)-but-2-enamido, 4-(N-methylmethoxylethylamino)-but-2-enamido, 4-(N-methylethanolamino)-but-2-enamido, 4-(dimethoxylethylamino)-but-2-enamido, 4-(N-methyl-6-amino-1-hexanolyl)-but-2-enamido, 4-(N-methylbenzylamino)-but-2-enamido, 2-(1-ethylpiperidin-4-ylidene)acetamido, 2-(1-(2-methoxylethyl)piperidin-4-ylidene)acetamido, acrylamido, but-2-enamido, 3-methyl-but-2-enamido, and 2-(pyrrolidin-3-ylidene)acetamido;


one of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, and substituted or unsubstituted C6-C18 aryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C24 heteroaralkyloxy; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is selected from the group consisting of C1-C6 alkylacylamino, C2-C6 alkenylacylamino, and C6-C18 arylacylamino;


one of R2 and R3 is H, while the other one is substituted or unsubstituted C6-C18 aryl, wherein a substituent on the aryl is selected from the group consisting of halogen, C6-C24 heteroaralkyloxy, C2-C6 alkynyl, and C7-C24 aralkyloxy; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is C2-C6 alkenylacylamino;


R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is 2-(piperidin-4-ylidene)acetamido;


R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl; and


R4 is tetrahydrofuranyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is C2-C6 alkenylacylamino;


one of R2 and R3 is H, while the other one is selected from the group consisting of substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C18 aryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyloxy, and C6-C15 aryloxy; and


R4 is hexahydropyridinyl optionally substituted with C1-C6 alkyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof, wherein:


R1 is 2-(piperidin-4-ylidene)acetamido;


one of R2 and R3 is H, while the other one is selected from the group consisting of substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C18 aryloxy, and a substituent on the heteroaryl is C7-C24 aralkyloxy; and


R4 is hexahydropyridinyl optionally substituted with C1-C6 alkyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof, wherein:


R1 is 2-(pyrrolidin-3-ylidene)acetamido;


one of R2 and R3 is H, while the other one is C5-C18 heteroaryl substituted with aryloxy; and


R4 is hexahydropyridyl optionally substituted with C1-C6 alkyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is C2-C6 alkenylacylamino;


one of R2 and R3 is H, while the other one is selected from the group consisting of substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyloxy, and C6-C18 aryloxy; and


R4 is pyridinyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,


wherein:


R1 is 2-(piperidin-4-ylidene)acetamido;


one of R2 and R3 is H, while the other one is selected from the group consisting of substituted or unsubstituted C6-C18 aryl, and substituted or unsubstituted C5-C18 heteroaryl, wherein a substituent on the aryl is selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy, and a substituent on the heteroaryl is selected from the group consisting of C7-C24 aralkyloxy, and C6-C18 aryloxy; and


R4 is pyridinyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof, wherein:


R1 is 2-(pyrrolidin-3-ylidene)acetamido;


one of R2 and R3 is H, while the other one is C5-C18 heteroaryl substituted with C6-C18 aryloxy; and


R4 is pyridinyl.


In some embodiments, the present application is directed to a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof, wherein:


R1 is 4-(diethylamino)-but-2-enamido;


one of R2 and R3 is H, while the other one is C6-C18 aryl substituted with halogen; and


R4 is pyridinyl.


The specific embodiments of a compound of formula I are more detailedly described in the following preparations of the compound of the present invention.


Methods of Use

The present application provides a method for treating a patient having a tumor or protecting a patient from development of a tumor, comprising administering to an animal in need thereof, such as a mammal, especially a human patient, a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the invention.


The compounds of the invention can be used to treat and/or prevent tumor. Therefore, for example, the compounds of the invention can be used to treat, prevent development of a tumor, relieve growth of tumor cells, or kill tumor cells. In some embodiments, the compounds of the invention are administered to a subject having a tumor. In one embodiment, the subject is human. In some embodiments, the tumor cells are contacted with one or more compounds of the invention.


As will be understood by a person having ordinary skill in the art, “need” is not an absolute term and merely implies that the patient can benefit from the treatment of the anti-tumor agent. By “patient” what is meant is an organism which benefits by the use of the anti-tumor agent. For example, any organism with the cancer, such as a colorectal carcinoma, a prostate carcinoma, a breast adenocarcinoma, a non-small cell lung carcinoma, an ovarian carcinoma, multiple myelomas, a melanoma, and the like, may benefit from the application of the anti-tumor agent that may in turn reduce the amount of the cancer present in the patient. In one embodiment, the patient's health may not require that an anti-tumor agent be administered, but the patient may still obtain some benefit by the reduction of the level of the tumor cells present in the patient, and thus be in need. In some embodiment, the anti-tumor agent is effective against one type of the tumor, but not against other types, therefore, allowing a high degree of selectivity in the treatment of the patient. In choosing such an anti-tumor agent, the methods and results disclosed in the examples can be useful.


The term “anti-tumor agent” as used herein refers to a compound or composition including the compound that reduces the likelihood of survival of a tumor cell. In one embodiment, the likelihood of survival is determined as a function of an individual tumor cell. Therefore, the anti-tumor agent will increase the chance that an individual tumor cell will die. In one embodiment, the likelihood of survival is determined as a function of a polulation of tumor cells. Therefore, the anti-tumor agent will increase the chances that there will be a decrease in the population of tumor cells. In one embodiment, the anti-tumor agent means the chemotherapeutant (chemotherapeutic agent) and other similar terms.


The term “chemotherapeutic agent” as used herein refers to a compound useful in the treatment of the neoplastic disease, such as cancer. Examples of the chemotherapeutic agents include alkylating agent, such as a nitrogen mustard, an ethyleneimine and a methylmelamine, an alkyl sulfonate, a nitrosourea and a triazene, folic acid antagonists, antimetabolites of nucleic acid metabolism, antibiotics, pyrimidine analogs, 5-fluorouracil, cisplatin, purine nucleosides, amines, amino acids, triazole nucleosides, corticosteroids, a natural product such as a vinca alkaloid, an epipodophyllotoxin, an antibiotic, an enzyme, a taxane, and a biological response modifier; miscellaneous reagents, such as a platinum coordination complex, an anthraquinone, an anthracycline, a substituted urea, a methyl hydrazine derivative, or an adrenocortical suppressant; or a hormone or an antagonist such as an adrenocorticosteroid, a progesterone, an estrogen, an antiestrogen, an androgen, an antiandrogen, or a gouadotropin-releasing hormone analog. Specific examples include doxorubicin, 14-hydroxy daunorubicin, 5-fluorouracil, cytosine arabinoside (“Ara-C”), cyclophosphamide, thiotepa, busulfan, cytotoxin, taxol, Toxotere, methotrexate, cisplatin, melphalan, vinblastine, bleomycin, etoposide, ifosfamide, mitomycin C, mitoxantrone, vincristine, vinorelbine, carboplatin, teniposide, daunorubicin, 10-demethylated daunorubicin, aminopterin, dactinomycin, mitomycin, esperamicin, melphalan, and other related nitrogen mustards. Also included in this definition are hormone reagents that act to regulate or inhibit hormone action on tumors, such as tamoxifen and onapristone.


The anti-tumor agent may act directly on a tumor cell to kill the cell, induce death of the cell, to prevent division of the cell, and the like. Alternatively, the anti-tumor agent may act indirectly on a tumor cell, such as by limiting nutrient or blood supply to the cell. Such anti-tumor agents are capable of destroying or suppressing the growth or reproduction of the tumor cells, such as a colon carcinoma, a prostate carcinoma, a breast adenocarcinoma, a non-small cell lung carcinoma, an ovarian carcinoma, multiple myelomas, a melanoma, and the like.


The term “neoplastic disease” or “neoplasm” as used herein refers to a cell or a population of cells, including a tumor or tissue (including cell suspensions such as bone marrow cell and fluids such as blood or serum), that exhibits abnormal growth by cellular proliferation greater than normal tissue. Neoplasms can be benign or malignant.


The methods of treatment disclosed herein can be used for any patient suspectable of having benign or malignant tumor growth, cancer or other tumorigenesis growth (the “tumor” or “tumors” as used herein covers tumor, solid tumor, cancer, disseminated tumor formative cell and topical tumorigenesis growth). Examples of the growth include, but are not limited to, breast cancer; osteosarcoma, angiosarcoma, fibrosarcoma, and other sarcoma; leukemia; sinus tumor; cancers of ovarian, ureter, bladder, prostate and other urogenital system; cancers of colon, esophageal and gastric and other gastrointestinal cancer; lung cancer; lymphoma; myeloma; pancreatic cancer; liver cancer; kidney cancer; endocrine cancer; skin cancer; melanoma; hemangioma; and brain or central nervous system (CNS; glioma) cancer. Generally, the tumor or growth to be treated can be any primary or secondary tumor or cancer.


In some embodiments, the cancer may be, for example, breast cancer, sarcoma, leukemia, ovarian cancer, ureter cancer, bladder cancer, prostate cancer, colon cancer, rectal cancer, stomach cancer, lung cancer, lymphoma, multiple myeloma, pancreatic cancer, liver cancer, kidney cancer, endocrine cancer, skin cancer, melanoma, hemangioma, and brain or central nervous system (CNS) cancer.


In some aspects, the cancer may be drug-resistant cancer. The drug-resistant cancer may exhibit, for example, one of: Bcl-2-overexpression, boosted level of the P-glycoprotein efflux pump, an increased expression of the multidrug-resistance related protein 1 encoded by MRP1, reduced drug intakes, the change of the drug target, or increased repairing of the drug induced DNA damage, the change of the apoptosis pathway, or activation of the cytochrome P450 enzyme. The drug-resistant cancer may be, for example, multiple myeloma, sarcoma, lymphoma (including non-Hodgkin's lymphoma), leukemia or any other drug-resistant cancer. The cancer may be, for example, naturally drug-resistant, or resistant to chemotherapy, biological therapy, radiotherapy, or immunotherapy. The cancer may be resistant to rituximab monoclonal antibody, Gleevac, velcade, Gleveec, Revlimid, Avastin, Tarceva, Erbitux, bortezomib, thalidomide and the like. Other specific examples of the drug-resistant cell line include MES-SA cell line, and multidrug-resistant derivative thereof, such as MES-SA/Dx5, HL-60 and HL-60/MX2.


Pharmaceutical Compositions

In one aspect, the present application is directed to a pharmaceutical composition, comprising a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof, and a pharmaceutically acceptable carrier,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In some embodiments, the pharmaceutical composition of the present application comprises a physiologically acceptable surfactants, carriers, diluents, excipients, smoothing agents, suspending agents, film forming substances, and coating assistants, or a combination thereof, and a compound of the invention. Acceptable carriers or diluents for therapeutic use are well-known in the art, and are described, for example, in Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, Pa. (1990), which is incorporated herein by reference in its entirety.


Preservatives, stabilizers, dyes, sweeteners, flavoring agents, fragrances, and the like, may be provided in the pharmaceutical composition. For example, sodium benzoate, ascorbic acid and esters of p-hydroxybenzoic acid may be added as preservatives. Furthermore, antioxidants and suspending agents may be used.


In various embodiments, alcohols, esters, sulfating aliphatic alcohols, and the like may be used as surfactants; sucrose, glucose, lactose, starch, crystalline cellulose, mannitol, light anhydrous silicate, magnesium aluminate, methyl magnesium silicate aluminate, synthetic aluminum silicate, calcium carbonate, calcium bicarbonate, calcium hydrogenphosphate, calcium hydroxymethyl cellulose and the like may be used as excipients; magnesium stearate, talc, hardened oil may be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soybean may be used as suspending agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or methylacetate-metharylate copolymer as a derivative of polyethylene may be used as suspending agents; and plasticizers such as ester phthalates and the like may be used as suspending agents.


Suitable routes of administration may, for example, include oral, rectal, transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal or intraocular injections. The compound can be administered in sustained or controlled release dosage froms, including depot injections, osmotic pumps, pills, transdermal (including electromigrating) patches, and the like for prolonged and/or timed, pulsed administration at a predetermined rate.


Pharmaceutical compositions of the present application may be manufacture in manner that is itself known, for example, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or tabletting processes.


Pharmaceutical compositions for use in accordance with the present application thus may be formulated by a conventional manner using one or more physiologically acceptable carriers comprising excipienst and auxiliaries which facilitate processing the active compounds into preparation which can be used pharmaceutically. Proper formulation is dependent on the route of administration chosen. Any of the well-known techniques, carriers and excipients may be used as suitable and as understood in the art.


Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, glucose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like. Furthermore, if desired, the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like. Physiologically compatible buffers include, but are not limited to, Hank's solution, Ringer's solution or physiological saline buffer. If desired, absorption enhancing preparations (such as liposomes) may be used.


For transmucosal administration, penetrants suitable for the barrier to be permeated may be used in the formulation.


Pharmaceutical formulations for parenteral administration, e.g., by bolus injection or continuous infusion, include aqueous solution of the active compounds in water-soluable form. Furthermore, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipotropic solvents or vehicles inculde fatty oil such as sesame oil, or other organic oils such as soybean oil, grapefruit oil or almond oil, or synthetic fatty acid esters, such as ethyl oleate or triglyceride, or liposomes. Aqueous injection suspension may contain substances which increase the viscosity of the suspension, such as sodium carboxymethylcellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Formations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending agents, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable carrier, e.g., sterile pyrogen-free water, before use.


For oral administration, the compound can be formulated readily by combining the active compound with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compound of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, ointments, suspensions, and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparation for oral use can be obtained by combining the active compound with solid excipient, optionally grinding a resultant mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, saccharose, mannitol or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, hydroxypropyl methylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the crosslinked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solution, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added into the tablets or dagree coatings for identification or to characterizing different combinations of active compound doses. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solution, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added into the tablets or dagree coatings for identification or to characterizing different combinations of active compound doses.


Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain active ingredients in admixture with filler such as sugar, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oil, liquid paraffin, or liquid polyethylene glycols. Furthermore, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.


For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.


For administration by inhalation, the compound for the invention is conveniently delivered in the form of an aerosol spray presentation from the pressurized packs or a nebulizer, with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.


Further disclosed herein are various pharmaceutical compositions well known in the pharmaceutical art for uses that include intraocular, intranasal, and intraauricular delivery. Suitable penetrants for these uses are generally known in the art. Pharmaceutical compositions for intraocular delivery include aqueous ophthalmic solution of the active compounds in water-soluble form, such as eyedrops, or in gellan gum or hydrogels; ophthalmic ointments; ophthalmic suspensions, such as microparticulates, drug-containing small polymeric particles that are suspended in a liquid carrier medium, lipid-soluble formulations, and microspheres; and ocular inserts. Suitable pharmaceutical formulations are most often and preferably formulated to be sterile, isotonic and buffered for stability and comfort. Pharmaceutical compositions for intranasal delivery may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action. As well known to a person having ordinary skill in the art, suitable formulations are most often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers. Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for such aural formulations include glycerin and water.


The compound may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., including conventional suppository bases such as cocoa butter or other glycerides.


In addition to the formulations described previously, the compound may also be formulated as a depot preparation. Such long acting formulations may be administrated by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Therefore, for example, the compound may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or s sparingly soluble derivatives, for example, as a sparingly soluble salt.


For hydrophobic compounds, a suitable pharmaceutical carrier may be a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. A common cosolvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant POLYSORBATE 80™, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity. Furthermore, the identity of the co-solvent may be varied, for example, other low-toxicity nonpolar surfactants may be used instead of POLYSORBATE 80™; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyethylene pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.


Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethyl sulfoxide also may be employed, although usually at the cost of greater toxicity. Furthermore, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by a person having ordinary skill in the art. Sustained-release capsules may, depending on their chemical nature, release the compound fore a few weeks up to over 100 days.


Agents intended to be administered intracellularly may be administered using techniques well known to a person having ordinary skill in the art. For example, such agents may be encapsulated in liposomes. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposome contents are both protected from the external micro-environment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm. The liposome may be coated with a tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the desirable organ. Alternatively, small hydrophobic organic molecules may be directly administered intracellularly.


Methods of Treatment and Use

In one aspect, the present application is directed to a method for treating and/or preventing tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In some embodiments, the mammal in the method for treating and/or preventing tumor is a human.


In some embodiment, the receptor tyrosine kinase in the method for treating and/or preventing tumor in a mammal occurs overexpression or mutation.


In some embodiments, the receptor tyrosine kinase in the method for treating and/or preventing tumor in a mammal is erbB family.


In some embodiments, the erbB family in the method for treating and/or preventing tumor in a mammal is selected from EGFR and/or Her2.


In another aspect, the present application is directed to a method for inhibiting growth of tumor cells, comprising contacting the tumor cells with a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In some embodiment, the receptor tyrosine kinase in the method for inhibiting growth of tumor cells occurs overexpression or mutation.


In some embodiments, the receptor tyrosine kinase in the method for inhibiting growth of tumor cells is erbB family.


In some embodiments, the erbB family in the method for inhibiting growth of tumor cell is selected from EGFR and/or Her2.


In yet another aspect, the present application is directed to a method for inhibiting overexpression or mutantion of a receptor tyrosine kinase in a mammal, comprising contacting the receptor tyrosine kinase with a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In some embodiments, the receptor tyrosine kinase in the method for inhibiting overexpression or mutation of a receptor tyrosine kinase in a mammal is erbB family.


In some embodiments, the erbB family in the method for inhibiting overexpression or mutation of a receptor tyrosine kinase in a mammal is selected from EGFR and/or Her2.


In yet another aspect, the present application is directed to a method for treating and/or preventing physiological abnormality caused by overexpression or mutation of a receptor tyrosine kinase in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein:


R1 is selected from the group consisting of substituted or unsubstituted alkylacylamino, substituted or unsubstituted alkenylacylamino, substituted or unsubstituted alkynylacylamino, substituted or unsubstituted arylacylamino, substituted or unsubstituted amino, and substituted or unsubstituted alkoxy;


R2 and R3 are each independently selected from the group consisting of hydrogen, substituted or unsubstituted aralkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;


or R2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted heterocyclyl; and


R4 is substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl.


In some embodiments, the receptor tyrosine kinase in the method for treating and/or preventing physiological abnormality caused by overexpression or mutation of a receptor tyrosine kinase in a mammal is erbB family.


In some embodiments, the erbB family in the method for treating and/or preventing physiological abnormality caused by overexpression or mutation of a receptor tyrosine kinase in a mammal is selected from EGFR and/or Her2.


In some embodiments, the physiological abnormality in the method for treating and/or preventing physiological abnormality caused by overexpression or mutation of a receptor tyrosine kinase in a mammal is tumor.


Methods of Administration

The compound or pharmaceutical compositions may be administered to the patient in any suitable means. Non-limiting examples of methods of administration include, among other, (a) administration through oral pathways, which include administration in capsule, tablet, granule, spray, syrup, or other such forms; (b) administration through non-oral pathways, such as rectal, vaginal, intraurethral, intraocular, intranasal, or intraauricular, which include administration as an aqueous suspension, an oily preparation or the like or as a drip, spray, suppository, salve, ointment or the lise; (c) administration via injection, subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, intraorbitally, intracapsularly, intraspinally, intrasternally, or the like, including infusion pump delivery; (d) administration locally such as by injection directly in the renal or cardiac area, e.g., by depot implantation; as well as (e) administration topically; as deemed appropriate by a person having ordinary skill in the art for bringing the compound of the invention into contact with living tissue.


The most suitable route depends on the nature and severity of the condition to be treated. A person having ordinary skill in the art also knows determination of methods of administration (buccal, intravenous, inhalation subcutaneous, rectal and the like), dosage form, suitable pharmaceutical excipients and other events regarding delivering the compound to a subject in need thereof.


Pharmaceutical compositions suitable for administration include the compositions where the active ingredient is contained in an effective amount to achieve its intended purpose. The therapeutically effective amount of the pharmaceutical composition disclosed herein required as a dose depends on the route of administration, the type of the animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The does can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize. More specifically, a therapeutically effective amount means an amount of the compound effective to prevent, alleviate or ameliorate symptoms of disease or prolongs the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capacity of a person having ordinary skill in the art, especially in light of the detailed disclosure provided herein.


As will be readily apparent to one skilled in the art, the useful in vivo dosage to be administered and particular mode of administration will vary depending on the age, weight and f mammal species treated, and the specific use for which the compound are employed. The determination of effective amount level, that is, the dosage levels necessary to achieve the desired results, can be accomplished by a person having ordinary skill in the art using routine pharmacological methods. Typically, human clinical applications of the compound are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro study can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods.


In non-human animal studies, applications of potential compounds are commenced at higher dosage levels, with dosage being decreased until the desired effect is no longer achieved or the adverse side effects disappear. The dosage may range broadly, depending on the desired effects and the therapeutic indication. Typically, the dosage may be between about 10 μg/kg and 500 mg/kg body weight, preferably between about 100 μg/kg and 200 mg/kg body weight. Alternatively, dosages may be based and calculated upon the surface area of the patient, as understood by a person having ordinary skill in the art.


The exact formulation, route of administration and dosage of the pharmaceutical compositions of the invention can be chosed by the individual physician in view of the patient's condition. Typically, the dose range of the composition administered to the patient can be from about 0.5 mg/kg to 1000 mg/kg of the patient's body weight. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. In instances where human dosages for compounds have been established for at least some condition, the present invention will use those same dosages, or dosages that are between about 0.1% and about 500%, more preferred between 25% to 250% of the established human dosage. Where no human dosage is established, as well be the case for newly-discovered pharmaceutical compounds, a suitable human dosage can be inferred from ED50 or ID50 values, or other appropriate values derived from in vitro or in vivo studies, as quantified by toxicity studies and efficacy study in animals.


It should be noted that the attending physician would know how to and when to terminate, interrupt, or adjust the administration due to toxicity and organs dysfunction. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.


Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg to 2000 mg of each active ingredient, preferably between 1 mg to 1000 mg of each active ingredient, e.g., 5 mg to 500 mg of each active ingredient. In other embodiments, an intravenous, subcutaneous or intramuscular dosage of each active ingredient of between 0.01 mg and 1000 mg, preferably between 0.1 mg and 800 mg, e.g. 1 mg to 200 mg is used. In cases of administration of a pharmaceutically acceptable salt, dosages may be calculated as the free base. In some embodiments, the compound is administered one to four times per day. Alternatively, the compositions of the present invention may be administered by continuous intravenous infusion, preferably at a dose of each active ingredient up to 1000 mg per day. As will be understood by a person having ordinary skill in the art, in certain situations, it may be necessary to administer the compound disclosed in the present invention in amounts that exceed, or even far exceed, the above preferred dosage range in order to effectively and aggressively treat particularly aggressive diseases or infections. In some embodiments, the compound will be administered for a period of continuous therapy, for example a week or more, or for months or years.


Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound, but the MEC can be estimated from in vitro data. Dosage necessary to achieve the MEC will depend on individual characteristics of route of administration. However, HPLC assays or biological assay can be used to determine plasma concentrations.


Dosage intervals can be also determined using MEC value. Compositions can be administered using regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably for 30-90% of the time and more preferably for 50-90% of the time.


In case of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.


The amount of the composition administered may be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgement of the prescribing physician.


Compounds disclosed in the present application can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties, can be established by determining in vitro toxicity towards a cell line, such as a mammalian cell line, and preferably a human cell line. The results of such studies are often predicative of toxicity in animals, such as mammals, or more specifically, humans. Alternatively, the toxicity of particular compounds in an animal model, such as mice, rats, rabbits or monkeys and the like can be determined using known methods. The efficacy of a particular compound can be established using several art recognized methods, such as in vitro methods, animal models, or human clinical trials. Art-recognized in vitro models exist for nearly every class of condition, including but not limited to cancer, cardiovascular disease, and various immune dysfunctions. Similarly, acceptable animal models can be used to establish efficacy of chemicals to treat such conditions. When selecting a model to determine efficacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, and route of administration, and regimen. Of course, human clinical trials can also be used to determine the efficacy of a compound in humans.


The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser may be accompanied with instructions for administration. The pack or dispenser may be also accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for of human or veterinary administration. Such notice, for example, may be the labeling approved by the United State Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising the compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.


Preparation of Compounds

The process for preparing a compound of the present invention is exemplarily illustrated in the following reaction scheme. The compound is a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, a solvate thereof or a prodrug thereof,




embedded image


wherein R1, R2, R3 and R4 are each as defined in the present application.


It is understood that the following description, combinations of substituents and/or the variables of the depicted formulae are permissible only if such contributions result in stable compounds.


It will also be appreciated by a person having ordinary skill in the art that in the process described below the functional groups of intermediate compounds may be needed to be protected by suitable protecting group. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxyl include trialkylsilyl or diarylalkylsilyl (such as tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include tert-butylcarbonyl, carboxybenzyl, fluorenylmethoxy carbony, and the like. Suitable protecting groups for mercapto include —C(O)—R″ (where R″ is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or aralkyl esters.


Protecting group may be added or removed in accordance with the standard techniques, which are well-known by a person having ordinary skill in the art and as described herein.


The use of protecting groups is described in detail in Green, T. W. and P. G. M. Wuts, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. The protecting group may be polymer resin such as Wang resin or 2-chlorotrityl-chloride resin.


It will also appreciated by a person having ordinary skill in the art, although such protected derivatives of compound of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as “prodrug”. All prodrugs of compounds of this invention are included within the scope of the invention.


The following Reaction Scheme illustrates methods to make compounds of this invention. It is understood that a person having ordinary skill in the art would be able to make these compounds by similar methods or by methods known to the one skilled in the art. It is also understood that a person having ordinary skill in the art would use suitable starting components in the similar way as described below, and modify the synthesis parameters as required, in order to manufacture other compounds of formula I which are not explicitly illustrated hereinafter. In general, starting components may be obtained from the common commercial sources or synthesized according to sources known to a person having ordinary skill in the art or prepared as described in this invention.


R1, R2, R3 and R4 are defined in the following reaction scheme as in the Specification.


In general, the compounds of formula I can be synthesized following the gernal procedure as described in the Reaction Scheme 1.




embedded image


In Reaction Scheme 1, R′ represents C1-C6 alkylacyl, C2-C6 alkenylacyl, C2-C6 alkynylacyl, C6-C18 arylacylamino, or C1-C6 alkyl. All the compounds may be present as a stereoisomer, a cis-trans-isomer, a tautomer or a mixture thereof


As described below, the compound of formula (101) is subject to nitration to obtain the nitrated derivative of formula (102). Nitrating agent may be a mixed acid of nitric acid and sulfuric acid, concentrated nitric acid, and preferably a mixed acid. It is well-known for a person having ordinary skill in the art how to select conditions of a nitration reaction.


The compound of formula (102) reacts with alcohol of formula R4OH in the presence of a base. The resulting compound is treated with concentrated sulfuric acid, concentrated hydrochloric acid, concentrated phosphoric acid, and the like, to obtain the compound of formula (103). A base that can be used in the present invention includes, but is not limited to LiOH, NaOH, KOH, sodium ethoxide, potassium tert-butoxide, and the like.


The compound of formula (103) reacts with ethyl 2-cyano-3-ethoxy acrylate in a solvent to obtain the compound of formula (104). The solvent that can be used in this reaction includes, but is not limited to dichloromethane, toluene, and the like.


The compound of formula (104) is heated in a solvent in the presence of inert gas to obtain the compound of formula (105). The solvent that can be used in this reaction includes, but is not limited to Dowtherm A, and the like.


The compound of formula (105) reacts with a chlorinating agent to obtain the compound of formula (106). The chlorinating agent that can be used in the present invention includes, but is not limited to HCl, SOCl2, PCl3, PCl5, POCl3, COCl2, and the like.


The compound of formula (106) reacts with an amine of formula HNR2R3 to obtain the compound of formula (107). The nitro group in the compound of formula (107) is reduced to an amino group with a reduction reaction to obtain the compound of formula (108). The reducing agent that can be used in this reaction includes, but is not limited to hydrogen, Zn/CH3COOH, SnCl2, Na2Sx, NaSO3, hydrazine and the like. Alternatively, electrochemical reduction process may be used.


The compound of formula (108) reacts with R′X, in which X represents Cl, Br, F, OMs or OTs to obtain the compound of formula I of the present invention.


In the following preparations for preparing intermediates of a compound of general formula I and the following examples regarding a compound of formula I, the used serial numbers of the compounds do not correspond to the serial numbers of the compounds described in the above Reaction Scheme.


Abbreviations in the following description of the preparation methods are as follows: Dowtherm A: a mixture of biphenyl and biphenyl ether; DCM: dichloromethane; THF: tetrahydrofuran; DIEA: diisopropylethylamine; DMF: N,N-dimethyl carboxamide; NBS: bromosuccinimide; DMAP: 4-dimethylaminopyridine; DCC: dicyclohexylcarbodiimide; Boc: tert-butoxycarbonyl; Fmoc: 9-fluorenylmethoxycarbonyl; Ms: methanesulfonyl; Ts: p-toluene sulfonyl; Su: succinylimide; At: 7-azobenzotriazol-1-yl; Bt: benzotriazol-1-yl; CBZ: benzyloxycarbonyl; Tyr: tyrosine; Glu: glutamate.


Unless specifically indicated, all H1-NMR spectrum measuring instruments used in the exmples in the present application are 400 MHz nuclear magnetic resonance spectrometer.


Preparation 1
Methyl 2-Acetylamino-4-Fluoro-5-Nitro-Benzoate



embedded image


To a three-neck flask (2,000 ml) was added concentrated nitric acid (500 ml). The mixture was cooled in an ice-water bath. Concentrated sulfuric acid (500 ml) was dropwise added to the mixture under mechanical stirring. After addition, the temperature of the resulting mixture was maintained below 15° C. Methyl 2-acetylamino-4-fluoro-benzoate (105.5 g, 0.5 mol) was slowly added. The resultant mixture was kept in an ice-water bath for 40 min with stirring. Then the mixture was poured into ice water (8 L). A large number of yellow solids precipitated. The mixture was stood for 10 min and filtered in vacuo. The filter cake was washed with lots of water and then transferred in water (2 L). The pH of the solution was adjusted to 7 with ammonia liquor under stirring and filtered in vacuo. The filter cake was dried by baking and recrystalized with ethyl acetate.


Preparation 2
4-Nitro-3-(Tetrahydrofuran-3-yl-Oxy)-Benzenamine



embedded image


To a single-neck reaction flask (100 ml) was added 3-hydroxytetrahydrofuran (27 ml, 400 mmol) under argon atmosphere. To the flask was added potassium tert-butoxide (15.9 g, 150 mmol) in batch under stirring. The resulting mixture was warmed to the temperature of 75° C. and stirred. After 15 min, the white solid substantially disappeared. Methyl 2-acetylamino-4-fluoro-5-nitro-benzoate (25.6 g, 100 mmol) was slowly added in batch to the solution. After addition, the resultant mixture was heated and stirred for further 50 min. The reaction was stopped. The mixture was slightly cooled and poured into water (500 ml). A large number of red solids precipitated. The pH of the solution was adjusted to 3 with hydrochloric acid (2N). The solids became yellow. The solution was extracted with ethyl acetate three times (total 800 mL). The aqueous layer was discarded. The layers of ethyl acetate were combined and washed once with water (300 mL) and once with saturated NaCl solution (300 mL). The organic layer was dried over MgSO4 for half-hour, filtered and rotary-evaporated to dryness. The resultant substance was dried with an oil pump to give a tawny foam.


Concentrated sulfuric acid (200 mL) was dropwise added into water (300 mL) in an ice-water bath. The resultant acid was poured into the tawny foam. The mixture was stirred for 4.5 h in an oil bath at the temperature of 110° C. The reaction was stopped. The reaction solution was poured into ice water (2 L). The mixture was stirred in an ice water bath. The pH of the resulting solution was adjusted to 9 with ammonia liquor. The resultant mixture was extracted with ethyl acetate three times. The ethyl acetate layers were combined and washed with water (500 mL), saturated NaHCO3 solution (500 mL), and saturated NaCl solution (500 mL), successively. The resultant organic layer was dried over MgSO4 for half-hour. The mixture was filtered and rotary-evaporated to dryness. The resultant substance was purified with column chromatography (eluent: dichloromethane:ethyl acetate=2:5) to give a yellow solid. Yield: 11.5 g, 51%.


Preparation 3
(E/Z)-Ethyl 2-Cyano-3-(4-Nitro-3-(Tetrahydrofuran-3-yl-Oxy)-Phenylamino)-Acrylate



embedded image


To a single-neck reaction flask (250 mL) were added 4-nitro-3-(tetrahydrofuran-3-yl-oxy)-benzenamine (7.966 g, 35.722 mmol), (E/Z)-ethyl 2-cyano-3-ethoxy acrylate (8.452 g, 50.0 mmol) and toluene (146 mL). The mixture was stirred and refluxed for 16 hr in an oil bath at the temperature of 115° C. The reaction was stopped. The resultant mixture was cooled in an ice-water bath and filtered in vacuo. The filter cake was washed with mother liquor, toluene (200 mL) and anhydrous ethyl ether, successively. The filter cake was pumped to dryness to give a yellow solid (11.5 g). The resultant substance was recrystalized with ethylene glycol monomethyl ether to give a yellow crystal (9.95 g). Yield: 80.5%.


Preparation 4a
4-Hydroxy-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


To a three-neck flask (2000 mL) under argon atmosphere was added (E/Z)-ethyl 2-cyano-3-(4-nitro-3-(tetrahydrofuran-3-yl-oxy)phenylamino)-acrylate (17.09 g, 49.251 mmol). To the flask was added Dowtherm A (600 mL) under argon atmosphere. After 10 min, the mixture was heated. The mixture was stirred at the temperature of about 256° C. for 2 hr. The resultant mixture was stood to cool to the room temperature. Yellow solids precipitated. Anhydrous ethyl ether (360 mL) was added to the mixture. The resulting mixture was stirred for 15 min at the room temperature and filtered in vacuo. The filter cake was washed with anhydrous ethyl ether and dried in the air. The filter cake was dissolved in THF (40 mL). The solution was stirred and refluxed at the termperature of 85° C. for 1 hr, cooled to the room temperature and filtered in vacuo. The resultant filter cake was washed with mother liquor and dried in the air. The resultant substance was recrystalized with ethylene glycol monomethyl ether to give a gray solid (6.485 g). Yield: 43.7%.


Preparation 4b
7-Fluoro-4-Hydroxy-6-Nitroquinoline-3-Carbonitrile



embedded image


The titled compound was prepared with 3-fluoro-4-nitrobenzenamine as starting material according to the processes of the Preparation 3 and Preparation 4a.


Preparation 4c
(S)-4-Hydroxy-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


The titled compound was prepared with (S)-4-hydroxy-6-nitro-7-(tetrahydrofuran-3-yl-oxy)quinoline-3-carbonitrile as starting material according to the processes of the Preparation 3 and Preparation 4a.


Preparation 4d
(R)-4-Hydroxy-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


The titled compound was prepared with (R)-4-hydroxy-6-nitro-7-(tetrahydrofuran-3-yl-oxy)quinoline-3-carbonitile as starting material according to the processes of the Preparation 3 and Preparation 4a.


Preparation 5a
4-Chloro-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


To a single-neck reaction flask (100 ml) were added 4-hydroxy-6-nitro-7-(tetrahydrofuran-3-yl-oxy)quinoline-3-carbonitrile (1.8 g, 3 mmol) and POCl3 (10 mL) under argon atmosphere. The mixture was warmed to the temperature of 105° C. The reaction mixture was stirred for 2.5 hr. Then the reaction was stopped. The resultant mixture was rotary-evaporated to dryness. DCM (20 mL) was added to dissolve the resultant mixture. The solution was poured into a cooled mixture solution of DCM (200 mL) and saturated K2CO3 solution (60 mL). The resultant mixture was stirred for 10 min. The solution was extracted and the aqueous layer was discarded. The DCM layer was washed with water (150 mL) and staturated NaCl solution (150 mL), successively. The resultant substance was dried over MgSO4 for half-hour, filtered, rotary-evaporated to dryness, and dried in vacuo to give a yellow solid (1.771 g). Yield: 92.7%.


The compounds of Preparation 5b and Preparation 5c were prepared with the compounds obtained from Preparation 4c and Preparation 4d as starting materials, respectively, according to the process of Preparation 5a.


Preparation 5b
(S)-4-Chloro-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 5c
(R)-4-Chloro-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 6a
3-Chloro-4-(Pyridin-2-yl-Methoxy)Benzenamine



embedded image


To a reaction flask were added 2-chloro-4-tert-butylcarbonylamino-phenol (24.35 g, 100 mmol), 2-chloromethylpyridine hydrochloride (32.8 g, 200 mmol), potassium carbonate (41.4 g, 300 mmol) and tetrabutyl ammonium iodide (1.107 g, 3 mmol). To the flask was added DMF (100 ml). The reaction mixture was stirred. After the reaction finished, the reaction solution was poured into water (1 L). The mixture was extracted with ethyl acetate three times. The ethyl acetate layers were combined. The resultant organic layer was washed with water (500 ml) and staturated saline solution (500 ml), successively. The resulting mixture was dried over anhydrous magnesium sulfate for 30 min, filtered and rotary-evaporated to dryness. The resultant substance was purified with column chromatography (eluent:ethyl acetate:petroleum ether=1:) to give N-(3-chloro-4-(pyridin-2-yl-methoxy)phenyl)-tert-butoxyacylamine.


To a reaction flask was added N-(3-chloro-4-(pyridin-2-yl-methoxy) phenyl)-tert-butoxyacylamine. N-(3-chloro-4-(pyridin-2-yl-methoxy)phenyl)-tert-butoxy acylamine was dissolved with 20% TFA in DCM (50 ml). The mixture was stirred at the room temperature. After the reaction finished, the solvent was rotary-evaporated. The residue was dissolved in DCM (200 ml). The resulting solution was washed with saturated sodium carbonate three times (200 ml×3), with water (200 ml) once, with saturated saline solution (200 ml) once, successively. The resultant substance was dried over anhydrous magnesium sulfate and filtered. The solvent was rotary-evaporated to give the target product.


The compounds of Preparations 6b to Preparation 6g were prepared with different starting materials according to a process similar to that of Preparation 6a.


Preparation 6b
1-Benzyl-1H-Indole-5-Amine



embedded image


Preparation 6c
3-((5-Amino-1H-Indol-1-yl)Methyl)Benzonitrile



embedded image


Preparation 6d
1-(3-Methoxybenzyl)-1H-Indol-5-Amine



embedded image


Preparation 6e
1-(3-Chlorobenzyl)-1H-Indol-5-Amine



embedded image


Preparation 6f
6-(Benzyloxy)-Indoline



embedded image


Preparation 6g
6-(Benzyloxy)Pyridin-3-Amine



embedded image


Preparation 6h
4-(2-Chlorobenzyloxy)-3-Chlorobenzenamine



embedded image


To a reaction flask were added 2-chloro-4-nitrophenol (5.205 g, 30 mmol), 2-chlorobenzyl chloride (5.313 g, 33 mmol), potassium carbonate (4.554 g, 33 mmol) and DMF (40 ml). The reaction mixture was heated to the temperature of 100° C. After the reaction finished, the reaction solution was poured into water (400 ml). The mixture was extracted with ethyl acetate (300 ml). The ethyl acetate layer was washed with saturated saline solution (200 ml) once and dried over anhydrous magnesium sulfate. The organic layer was filtered and the solvent was rotary-evaporated to dryness. The resultant substance was recrystalized with ethyl acetate to give 1-(2-chlorobenzyloxy)-2-chloro-4-nitrobenzene (8.567 g).


To a three-neck flask (500 ml) was added 1-(2-chlorobenzyloxy)-2-chloro-4-nitrobenzene (8.567 g). To the flask were added THF (100 ml) and methanol (50 ml). The solution was mechanically stirred and heated under reflux. Glacial acetic acid (17 ml) and reduced iron powders (16.8 g) were added into the solution. The mixture reacted for 1 hr. After the reaction finished, the resultant substance was filtered in vacuo and the filtrate was rotary-evaporated to dryness. To the resultant crude product was added HCl (4N, 200 ml). The mixture was sufficiently vibrated and then filtered in vacuo. To the resultant filter cake was added water (100 ml). The pH of the mixture was adjusted to 12 with 5% sodium hydroxide. Ethyl acetate (700 ml) was added. The solution was sufficiently vibrated and separated. The ethyl acetate layer was washed with saturated sodium carbonate (200 ml), water (200 ml) and saturated saline solution (200 ml), successively. The organic layer was dried over anhydrous magnesium sulfate and filtered. The solvent was rotary-evaporated to dryness. The resultant substance was recrystalized with ethyl acetate to give the target compound.


The compounds of Preparation 6i to Preparation 6aa were prepared according to the process of Preparation 6h.


Preparation 6i
4-(2-Fluorobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6j
4-(3-Fluorobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6k
4-(4-Fluorobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6l
4-(3-Chlorobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6m
4-(4-Chlorobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6n
4-(2-Methylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6o
4-(3-Methylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6p
4-(4-Methylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6q
4-(2-Methoxybenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6r
4-(3-Methoxybenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6s
4-(4-Methoxylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6t
2-((4-Amino-2-Chlorophenoxy)Methyl)Benzonitrile



embedded image


Preparation 6u
3-((4-Amino-2-Chlorophenoxy)Methyl)Benzonitrile



embedded image


Preparation 6v
4-((4-Amino-2-Chlorophenoxy)Methyl)Benzonitrile



embedded image


Preparation 6w
4-(4-Tert-Butylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6x
4-(Benzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6y
4-(2-Chlorobenzyloxy)-3-Fluorobenzenamine



embedded image


Preparation 6z
4-(3-Chlorobenzyloxy)-3-Fluorobenzenamine



embedded image


Preparation 6aa
4-(4-Chlorobenzyloxy)-3-Fluorobenzenamine



embedded image


Preparation 6ab
4-(4-Bromobenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6ac
4-(4-Ethylbenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6ad
4-(4-Ethoxybenzyloxy)-3-Chlorobenzenamine



embedded image


Preparation 6ah
6-(3-Chlorobenzyloxy)Pyridine-3-Amine



embedded image


Preparation 6ai
4-(3-Methoxybenzyloxy)Benzenamine



embedded image


Preparation 6aj
4-Benzyloxybenzenamine



embedded image


Preparation 6ak
2-Amino-5-(N-Benzoyl)-Amino-Pyrimidine



embedded image


To a three-neck flask were added 2-amino-5-nitropyrimidine (1.4 g, 10 mmol), (Boc)2O (2.18 g, 10 mmol), sodium bicarbonate (1.4 g), THF (15 ml) and water (15 ml). The mixture was stirred at the room temperature for 4 hr. After the reaction finished, the resultant mixture was exacted with chloroform three times (20 ml*3). The organic phases were combined and washed once with saturated sodium chloride solution (20 ml). The organic phase was dried over anhydrous magnesium sulfate. After half-hour, the drying agent was discarded and the filtrate was concentrated in vacuo to give 5-nitro-2-(N-tert-butoxycarbonyl)-aminopyrimidine as a solid (2.3 g).


5-nitro-2-(N-tert-butoxycarbonyl)-aminopyrimidine (2.3 g) was dissolved in THF solution (20 ml). 10% palladium on carbon (230 mg) was added to the solution. The mixture was hydrogenated for 6 hr. The solution was filtered with silica gel-sand panel funnel. The filtrate was retained and the solvent was evaporated in vacuo. The resultant substance was purified by flash chromatography with silica gel column and eluted with chloroform:methanol=9:1 to give 5-amino-2-(N-tert-butoxycarbonyl)-aminopyrimidine (1.5 g).


5-amino-2-(N-tert-butoxycarbonyl)-aminopyrimidine (1.5 g) was dissolved in redistilled DCM (15 ml). Redistilled triethylamine (1.2 ml, 8.6 mmol) was added. The solution was cooled in an ice water bath under nitrogen atmosphere. After 20 min, a solution of benzoyl chloride (1 ml) in DCM (5 ml) was dropwise added into the solution. The ice water bath was removed after the addition finished. The solution was stirred overnight. To the solution was added water (100 ml). The resultant solution was extracted and separated. The organic phase was retained and washed once with of saturated sodium chloride (20 ml) and dried over anhydrous magnesium sulfate. After 0.5 hr, the solution was filtered. The filtrate was concentrated in vacuo. The resulting substance was purified by flash chromatography with silica gel column and eluted with chloroform:methanol=95:5 to give 2-(N-tert-butoxycarbonyl)-amino-5-(N-benzoyl)-aminopyrimidine (1.6 g).


2-(N-tert-butoxycarbonyl)-amino-5-(N-benzoyl)-aminopyrimidine (1.6 g) was dissolved in DCM (20 ml). Trifluoroacetic acid (1.5 ml, 5.8 mmol) was added under stirring at the room temperature. The mixture was stirred at the room temperature. After 2 hr, the reaction was stopped. The solvent was rotary-evaporated to dryness in vacuo. To the resultant substance was added ethyl acetate (20 ml) and 2N HCl (20 ml). The mixture was extracted and separated. The aqueous phase was retained and the pH was adjusted to 10 with 10% sodium hydroxide solution. The resulting mixture was extracted three times with ethyl acetate (25 ml*3) and the ethyl acetate layers were combined. The resultant organic phase was washed with saturated sodium chloride (20 ml) once and dried over anhydrous magnesium sulfate. After 0.5 hr, the solution was filtered and the filtrate was concentrated in vacuo to give 2-amino-5-(N-benzoyl)-amino-pyrimidine (0.7 g).


The compounds of Preparation 6al to Preparation 6an were prepared according to the process of Preparation 6ak.


Preparation 6al
2-Amino-5-(N-4-Dimethylamino-Benzoyl)-Amino-Pyrimidine



embedded image


Preparation 6am
2-Amino-5-(N-4-Methoxy-Benzoyl)-Amino-Pyrimidine



embedded image


Preparation 6an
2-Amino-5-(N-Benzenesulfonyl)-Amino-Pyrimidine



embedded image


Preparation 6ao
2-(N-Benzoyl)-Amino-5-Amino-Pyrimidine



embedded image


To stirred solution of 2-amino-5-nitropyrimidine (1.0 g, 7.14 mmol) in pyridine (20 ml) was added benzoyl chloride (0.92 ml, 7.93 mmol). The mixture was refluxed for 4 hr under nitrogen atmosphere. The reactant mixture was cooled to the room temperature and poured into 200 ml water. The resulting mixture was stirred overnight and then filtered in vacuo. The filter cake was washed with water (20 ml*3) and dried in vacuo to give 2-(N-benzoyl)amino-5-nitropyrimidine as a white solid (790 mg).


2-(N-benzoyl)amino-5-nitropyrimidine (790 mg) was added into ethanol (100 ml). 80 mg of 10% palladium on carbon was added to the solution under stirring at the room temperature. The reactant was hydrogenated and stirred for 4 hr. The solution was filtered through silica gel-sand panel funnel. The filtrate was retained and the solvent was evaporated in vacuo. The resultant substance was purified by flash chromatography with silica gel column and eluted with chloroform:methanol=95:5 to give 2-(N-benzoyl)amino-5-aminopyrimidine (200 mg).


The compounds of Preparation 6ap to Preparation 6as were prepared according to the process of Preparation 6ao.


Preparation 6ap
2-(N-4-Methoxy-Benzoyl)-Amino-5-Amino-Pyrimidine



embedded image


Preparation 6aq
2-(N-Cyclohexyl-1-Benzoyl)-Amino-5-Amino-Pyrimidine



embedded image


Preparation 6ar
2-(N-2-Formyl-Thiophenyl)-Amino-5-Amino-Pyrimidine



embedded image


Preparation 6as
2-(N-2-Formyl-Furanyl)-Amino-5-Amino-Pyrimidine



embedded image


Preparation 7a
4-(3-Chloro-4(Pyridin-2-yl-Methoxy)Phenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


To a single-neck reaction flask (500 ml) were added 4-chloro-6-nitro-7-(tetrahydrofuran-3-yl-oxy)quinoline-3-carbonitrile (5 g, 15.649 mmol), 3-chloro-4-(pyridin-2-yl-methoxy)benzenamine (4.037 g, 17.214 mmol) and pyridine hydrochloride (421 mg). To the flask was added isopropanol (150 ml). The mixture was stirred and refluxed for 2 hr at the temperature of 85° C. The reaction was stopped and cooled to the room temperature. The resultant mixture was filtered in vacuo. The filter cake was washed with mother liquor and dried in vacuo to give a yellow solid (5.988 g). Yield: 73.9%.


The compounds of Preparation 7b to Preparation 7bh were prepared with different starting materials according to the process of Preparation 7a.


Preparation 7b
4-(3-Chloro-4-Fluorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7c
4-(3-Alkynylphenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7d
4-(3-Bromophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7e
6-Nitro-4-(1H-Indol-5-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7f
4-(4-(2-Fluorobenzyloxy)-3-Fhlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7g
4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7h
4-(4-(4-Fluorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7i
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7j
4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7k
4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7l
4-(4-(2-Methylbenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7m
4-(4-(3-Methylbenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7n
4-(4-(4-Nethylbenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7O
4-(4-(2-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7p
4-(4-(3-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7q
4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7r
4-(4-(2-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7s
4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7t
4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7u
4-(4-(4-Tert-Butylbenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7v
4-(4-Benzyloxy-3-Chlorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7x
4-(4-(2-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7y
4-(4-(3-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7z
4-(4-(4-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7aa
6-Nitro-4-((S)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7ab
6-Nitro-4-((R)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7ac
4-(1-Benzyl-1H-Indol-5-Ylamine)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ad
4-(1-(3-Cyanobenzyl)-1H-Indol-5-yl-Amino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ae
4-(1-(3-Methoxybenzyl)-1H-Indol-5-yl-Amino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7af
4-(1-(3-Chlorobenzyl)-1H-Indol-5-yl-Amino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ag
4-(Indolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ah
4-(6-Chloroindolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7ai
4-(6-Fluoroindolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7aj
4-(4-Chloroindolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7ak
4-(3,4-Dihydroquinolin-1(2H)-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7al
4-(6-Methyl-3,4-Dihydroquinolin-1(2H)-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7am
6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)-4-(7-(Trifluoromethyl)-3,4-Dihydroquinolin-1(2H)-yl)Quinoline-3-Carbonitrile



embedded image


Preparation 7an
4-((6-(Benzyloxy)Indolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ao
Methyl 1-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4yl) Indoline-2-Carboxylate



embedded image


Preparation 7ap
4-(2-(Hydroxymethyl)Indolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7aq
4-(6-(1H-Pyrrol-1-yl)Indolin-1-yl)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ar
6-Nitro-4-(Octahydroindol-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7as
6-Nitro-4-(Pyrimidin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7at
N-(2-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)Benzamide



embedded image


Preparation 7au
N-(2-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)-4-(Dimethylamino)Benzamide



embedded image


Preparation 7av
N-(2-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)Benzenesulfonamide



embedded image


Preparation 7aw
N-(5-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Benzamide



embedded image


Preparation 7ax
N-(5-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Furan-2-Carboxamide



embedded image


Preparation 7ay
N-(5-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Thiophene-2-Carboxamide



embedded image


Preparation 7az
N-(5-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Cyclohexylcarboxamide



embedded image


Preparation 7ba
5-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Pyrimidine-2-Carboxamide



embedded image


Preparation 7bb
6-Nitro-4-(Pyridin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7bc
6-(3-Cyano-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Nicotinamide



embedded image


Preparation 7bd
6-Nitro-4-(Pyridin-3-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7be
6-Nitro-4-(Pyridin-4-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7bf
4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-6-Nitro-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7bg
6-Nitro-4-(Pyrazin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7bh
4-(3-Chloro-4-Fluorophenylamino)-7-Fluoro-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bi
4-(3-Chloro-4-Fluorophenylamino)-7-(1 Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


The compound was obtained according to the process of Preparation 2 with 4-(3-chloro-4-fluorophenylamino)-7-fluoro-6-nitroquinoline-3-carbonitrile and 4-hydroxy-1-methylpiperidine as starting materials.


The compounds of Preparation 7bj to Preparation 7by were prepared according to the process of Preparation 7bi.


Preparation 7bj
4-(3-Ethynylphenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bk
4-(4-Benzyloxy-3-Chlorophenylamino)-7-(1 Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bl
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bm
4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bn
4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bo
4-(4-(4-Bromobenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bp
4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bq
4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7br
4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bs
4-(4-(4-Ethylbenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bt
4-(4-(4-Ethoxybenzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bu
4-(3-Chloro-4-Phenoxyphenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bv
7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitro-4-(Pyridin-2-yl-Amino) Quinoline-3-Carbonitrile



embedded image


Preparation 7bw
7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitro-4-(Pyridin-3-yl-Amino) Quinoline-3-Carbonitrile



embedded image


Preparation 7bx
7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitro-4-(Pyridin-4-yl-Amino) Quinoline-3-Carbonitrile



embedded image


Preparation 7by
4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-7-(1-Methylpiperidin-4-yl-Oxy)-6-Nitroquinoline-3-Carbonitrile



embedded image


Preparation 7bz
4-(3-Chloro-4-Fluorophenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


The compound was prepared according to the process of Preparation 2 and with 4-(3-chloro-4-fluorophenylamino)-7-fluoro-6-nitroquinoline-3-carbonitrile and 4-hydroxypyridine as starting materials.


The compounds of Preparation 7ca to Preparation 7cl were prepared according to the process of Preparation 7bz.


Preparation 7ca
4-(3-Ethynylphenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7cb
6-Nitro-4-(4-Phenoxyphenylamino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7cc
4-(4-(Benzyloxy)Phenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7cd
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ce
4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7cf
4-(4-(4-Methoxybenzyloxy)Phenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7cg
4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ch
6-Nitro-4-(Pyridin-2-yl-Amino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7ci
6-Nitro-7-(Pyridin-4-yl-Oxy)-4-(Pyrimidin-2-yl-Amino)Quinoline-3-Carbonitrile



embedded image


Preparation 7cj
4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-6-Nitro-7-(Pyridin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 7ck
4-(6-(3-Chlorobenzyloxy)Pyridin-3-yl-Amino)-6-Nitro-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 7cl
6-Nitro-4-(6-Phenoxypyridin-3-yl-Amino)-7-(Pyridin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8a
6-Amino-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


To a single-neck flask (500 mL) were added 4-(3-chloro-4-(pyridin-2-yl-methoxy)benzenamine)-6-nitro-7-(tetrahydrofuran-3-yl-oxy) quinoline-3-carbonitrile (5.950 g, 11.498 mmol) and SnCl2.2H2O (12.935 g). To the flask was added anhydrous ethanol (250 mL). The mixture was stirred under reflux at the temperature of 85° C. After 1.5 hr, the reaction finished. The resultant mixture was rotary-evaporated to dryness. To the resulting mixture was added water (150 mL). The resultant mixture was ultrasonically treated and rotary-evaporated to dryness. The resulting product was poured into water (300 mL). The pH of the mixture was adjusted to 8 with saturated NaHCO3 solution. The solution was extracted with chloroform three times (700 mL in total) and then extracted with ethyl acetate three times. The ethyl acetate layers were combined and washed with saturated NaCl solution (300 mL). The resultant organic phase was dried over anhydrous MgSO4 for half-hour, filtered and rotary-evaporated to dryness. The resultant products were combined and purified by column chromatography (chloroform:methanol=95:5) to give a yellow solid (2.881 g). Yield: 51.4%.


The compounds of Preparation 8b to Preparation 8ck were prepared according to the process of Preparation 8a.


Preparation 8b
6-Amino-4-(3-Chloro-4-Fluorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8c
6-Amino-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8d
6-Amino-4-(3-Bromophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8e
6-Amino-4-(1H-Indol-5-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8f
4-(4-(2-Fluorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8g
4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8h
4-(4-(4-Fluorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8i
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8j
4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8k
4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8l
4-(4-(2-Methylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8m
4-(4-(3-Methylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8n
4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8o
4-(4-(2-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8p
4-(4-(3-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8q
4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8r
4-(4-(2-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8s
4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8t
4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8u
4-(4-(4-Tert-Butylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8v
6-Amino-4-(4-(Benzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8x
4-(4-(2-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8y
4-(4-(3-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8z
4-(4-(4-Chlorobenzyloxy)-3-Fluorophenylamino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8aa
6-Amino-4-((S)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8ab
6-Amino-4-((R)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8ac
6-Amino-4-(1-Benzyl-1H-Indol-5-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ad
4-(1-(3-Cyanobenzyl)-1H-Indol-5-yl-Amino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ae
4-(1-(3-Methoxybenzyl)-1H-Indol-5-yl-Amino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8af
4-(1-(3-Chlorobenzyl)-1H-Indol-5-yl-Amino)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ag
6-Amino-4-(Indolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ah
6-Amino-4-(6-Chloroindolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8ai
6-Amino-4-(6-Fluoroindolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8aj
6-Amino-4-(4-Chloroindolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8ak
6-Amino-4-(3,4-Dihydroquinolin-1(2H)-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8al
6-Amino-4-(6-Methyl-3,4-Dihydroquinolin-1(2H)-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8am
6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)-4-(7-(Trifluoromethyl)-3,4-Dihydroquinolin-1(2H)-yl)Quinoline-3-Carbonitrile



embedded image


Preparation 8an
6-Amino-4-(6-(Benzyloxy)Indolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ao
Methyl 1-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-4-yl)Indoline-2-Carboxylate



embedded image


Preparation 8ap
6-Amino-4-(2-(Hydroxymethyl)Indolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8aq
4-(6-(1H-Pyrrol-1-yl)Indolin-1-yl)-6-Amino-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ar
6-Amino-4-(Octahydroindol-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8as
6-Amino-4-(Pyrimidin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8at
N-(2-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)Benzamide



embedded image


Preparation 8au
N-(2-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)-4-(Dimethylamino)Benzamide



embedded image


Preparation 8av
N-(2-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)Benzenesulfonamide



embedded image


Preparation 8aw
N-(5-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Benzamide



embedded image


Preparation 8ax
N-(5-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Furan-2-Carboxamide



embedded image


Preparation 8ay
N-(5-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Thiophene-2-Carboxamide



embedded image


Preparation 8az
N-(5-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl)Cyclohexylcarboxamide



embedded image


Preparation 8ba
5-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Pyrimidin-2-yl-Carboxamide



embedded image


Preparation 8bb
6-Amino-4-(Pyridin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8bc
6-(6-Amino-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Nicotinamide



embedded image


Preparation 8bd
6-Amino-4-(Pyridin-3-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8be
6-Amino-4-(Pyridin-4-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bf
6-Amino-4-(6 (Benzyloxy)Pyridin-3-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bg
6-Amino-4-(Pyrazin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bh
6-Amino-4-(3-Chloro-4-Fluorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)-Quinoline-3-Carbonitrile



embedded image


Preparation 8bi
6-Amino-4-(3 Ethynylphenylamino)-7-(1-Methylpiperidin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8bj
6-Amino-4-(4-(Benzyloxy)-3-Chlorophenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bk
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bl
4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bm
4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bn
4-(4-(4-Bromobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bo
4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bp
4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bq
4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8br
4-(4-(4-Ethylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bs
4-(4-(4-Ethoxybenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bt
6-Amino-4-(3-Chloro-4-Phenoxyphenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bu
6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-2-yl-Amino)Quinoline-3-Carbonitrile



embedded image


Preparation 8bv
6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-3-yl-Amino)Quinoline-3-Carbonitrile



embedded image


Preparation 8bw
6-Amino-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-4-yl-Amino)Quinoline-3-Carbonitrile



embedded image


Preparation 8bx
6-Amino-4-(6 (Benzyloxy)Pyridin-3-yl-Amino)-7-(1-Methylpiperidin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8by
6-Amino-4-(3-Chloro-4-Fluorophenylamino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8bz
6-Amino-4-(3-Ethynylphenylamino)-7-(Pyridin-4-yl-Oxy) Quinoline-3-Carbonitrile



embedded image


Preparation 8ca
6-Amino-4-(4-Phenoxyphenylamino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cb
6-Amino-4-(4-(Benzyloxy)Phenylamino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cc
4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cd
4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ce
4-(4-(4-Methoxybenzyloxy)Phenylamino)-6-Amino-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cf
4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-6-Amino-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cg
6-Amino-4-(Pyridin-2-yl-Amino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ch
6-Amino-7-(Pyridin-4-yl-Oxy)-4-(Pyrimidin-2-yl-Amino)Quinoline-3-Carbonitrile



embedded image


Preparation 8ci
6-Amino-4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8cj
4-(6-(3-Chlorobenzyloxy)Pyridin-3-yl-Amino)-6-Amino-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 8ck
6-Amino-4-(6-Phenoxypyridin-3-yl-Amino)-7-(Pyridin-4-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


Preparation 9
(E)-Trimethylsilyl 4-Bromo-2-Butenoate



embedded image


To a single-neck reaction flask (250 mL) were added of crotonic acid (10.750 g, 0.125 mol), anhydrous ethyl ether (125 mL) and trimethyl chlorosilane (19 mL, 0.150 mol). The mixture was stirred for 5 min. Pyridine (12 mL, 0.150 mol) was dropwise added into the resulting mixture under stirring at the temperature of 35° C. After the addition finished, the mixture was warmed up to the temperature of 38° C. and stirred under relux for 3 hr. The white solid was filtered in vacuo. Ethyl ether was evaporated under atmospheric pressure. The resultant was distilled under reduced pressure to collect the fraction under 10 mmHg at the temperature of 46° C.-48° C. to give (E)-trimethylsilyl 2-butenoate as a colorless liquid (12.242 g). Yield: 62.0%.


To a single-neck reaction flask (250 mL) were added (E)-trimethylsilyl 2-butenoate (12.242 g, 77.481 mmol), NBS (16.550 g, 92.977 mmol), (PhCO2)2 (310 mg) and carbon tetrachloride (100 mL). The mixture was stirred under reflux for 3.5 hr. The resulting mixture was cooled in an ice-water bath. The insoluble substance was filtered. The residue was rotatory-evaporated and concentrated until no liquid dropped. The concentrate was distilled under reduced pressure to collect the fraction under 9 mmHg at the temperature of 100° C.-102° C. to give the target compound as a colorless transparent liquid.


Preparation 10
(E/Z)-Trimethylsilyl 4-Bromo-2-Butenoate

The compound was prepared according to the process of Preparation 9 with 2-butenoic acid as starting material.


The compounds of Preparations 11, 12 and 13 were prepared according to the preparation process disclosed in Chinese patent No. CN 200610138377.9.


Preparation 11
2-(1-(Tert-Butoxycarbonyl)Piperidin-4-Ylidene)-Acetic Acid



embedded image


Preparation 12
2-(1-(2-Methoxyethyl)-Piperidin-4-Ylidene)-Acetic Acid



embedded image


Preparation 13
(E) 2-(1-(Tert-Butoxycarbonyl)Pyrrolin-3-Ylidene)Acetic Acid
Example 1
(E)-4-Bromo-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-but-2-Enamide



embedded image


a) After a single-neck reaction flask (100 mL) was blown with argon for 5 min, to the reaction flask was added (E)-trimethylsilyl 4-bromo-2-butenoate (1.363 g, 5.75 mmol). DCM (treated with molecular sieves) (3 mL) was added under argon atmosphere. To the mixture were added oxalyl chloride (0.55 mL, 6.325 mmol) and one drop of DMF (chromatographically pure) under stirring. Lots of bubbles produced. After 1 min, the bubbles reduced. The resulting mixture was continuously stirred for 2 hr at the room temperature and then rotary-evaporated to dryness.


b) 6-amino-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydro furan-3-yl-oxy)quinolin-3-carbonitrile (2.44 g, 5 mmol) was dissolved in THF (redistilled) (36 mL) under argon atmosphere. The solution was cooled in an ice-water bath. DIEA (1.73 mL, 10 mmol) was added. The resulting solution was stirred for 10 min. The rotary-evaporated product from a) was dissolved in THF (redistilled) (14 mL). The resultant solution was dropwise added into the reaction solution. The mixture was stirred in an ice-water bath overnight. The resultant mixture was rotary-evaporated to dryness. Ethyl acetate (200 mL) and saturated Na2CO3 solution (150 mL) were added. The solution was stirred for 10 min at the room temperature. After separation, the aqueous layer was extracted once with ethyl acetate (100 mL). The ethyl acetate layers were combined. The organic phase was washed with water (200 mL) and saturated NaCl solution (200 mL), dried over MgSO4 for half-hour, filtered and rotary-evaporated to dryness. The resultant substance was purified with column chromatography (eluent:chloroform:methanol=9:1) to give a yellow solid (2.381 g). Yield: 75.0%.


MS (M+1): 634.


H1-NMR (CDCl3): δ 2.222-2.270 (m, 1H); 2.346-2.476 (m, 1H); 3.431-3.458 (m, 1H); 3.925-3.955 (m, 1H); 4.012-4.247 (m, 4H); 5.158 (br, 1H); 5.281 (d, 2H, J=4.8); 6.114-6.422 (m, 2H); 6.905-7.198 (m, 3H); 7.535 (br, 1H); 7.648 (d, 1H, J=6.6); 7.765 (t, 1H, J=6.8); 8.154 (d, 1H, J=76.9); 8.485 (s, 1H); 8.596 (d, 2H, J=3.8); 9.109 (d, 1H, J=42.4).


Example 2
(E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


(E)-4-bromo-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2 butenamide (2.35 g, 3.704 mmol) was dissolved in DMF (chromatographically pure) (40 mL). The solution was cooled in an ice-water bath under argon atmosphere. To the solution was added NaI (278 mg, 1.852 mmol). The mixture was stirred for 30 min. To the mixture were added dimethylamine hydrochloride (604 mg, 7.408 mmol), potassium carbonate (2.556 g, 18.519 mmol) and tetrabutyl ammonium iodide (59 mg, 0.185 mmol). The resultant mixture was stirred at the room temperature overnight. After the reaction finished, the reaction solution was poured into staturated NaHCO3 solution (400 mL) to precipitate a yellow solid. The solid was extracted with ethyl acetate three times (550 mL in total). The ethyl acetate layers were combined. The organic phase was washed with water (300 mL) and saturated NaCl solution (300 mL), successively. The resultant organic phase was dried over anhydrous MgSO4 for half-hour, filtered and rotary-evaporated to dryness. The resultant substance was purified with column chromatography (eluent:chloroform:methanol=9:1) to give a yellow solid (1.375 g). Yield: 62.0%.


MS (M+1): 599.


H1-NMR (DMSO-d6, CH3OH-d4): δ 2.167-2.219 (m, 1H); 2.354-2.452 (m, 1H); 2.782 (s, 6H); 3.794 (dd, 1H, J1=8.0; J2=14.0); 3.949-4.074 (m, 5H); 5.258 (br, 1H); 5.546 (s, 2H); 6.795-6.914 (m, 2H); 7.387 (d, 1H, J=8.8); 7.551 (s, 1H); 7.674 (s, 1H); 7.845 (t, 1H, J=6.4); 8.000 (d, 1H, J=7.8); 8.406 (t, 1H, J=7.6); 8.851 (d, 1H, J=5.2); 9.043 (s, 1H); 9.231 (s, 1H).


Example 3
(E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide Hydrochloride



embedded image


(E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahy drofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-2-butenamide was dissolved in redistilled methanol (10 mL). A solution of HCl in ethyl ether (5 mL, 1.8 N) was dropwise added into the resultant solution under stirring. The resulting mixture was stirred for 10 min and rotary-evaporated to dryness. The resultant substance was recrystalized with a mixed solution of anhydrous methanol and hydrochloric acid (2 N) to give a crystal of (E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-2-butenamide as hydrochloride (1.163 g).


MS (M+1): 599.


The compounds of Examples 4-21 were prepared according to the process for preparing the compound of Example 2.


Example 4
(E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 510




embedded image


Example 5
(E)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 482


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.173-2.218 (m, 1H); 2.405-2.468 (m, 1H); 2.746 (s, 3H); 2.755 (s, 3H); 3.774-3.831 (m, 1H); 3.956-4.103 (m, 5H); 4.315 (s, 1H); 5.270 (br, 1H); 6.826-6.968 (m, 2H); 7.500 (s, 3H); 7.559 (s, 1H); 7.768 (s, 1H); 9.074 (s, 1H); 9.219 (s, 1H); 10.037 (s, 1H); 11.340 (br, 2H).


Example 6
(E)-N-(4-(4-(Benzyloxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 564




embedded image


Example 7
(E)-N-(4-(3-Bromophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 536


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.157-2.216 (m, 1H); 2.385-2.456 (m, 1H); 2.741 (s, 3H); 2.753 (s, 3H); 3.803 (dd, 1H, J1=8.0, J2=13.6); 3.939-4.103 (m, 5H); 5.266 (br, 1H); 6.826-6.968 (m, 2H); 7.431-7.466 (m, 2H); 7.587-7.608 (m, 1H); 7.718 (s, 1H); 7.786 (s, 1H); 9.110 (s, 1H); 9.222 (s, 1H); 10.052 (s, 1H); 11.345 (br, 2H).


Example 8
(E)-N-(4-(4-(2-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 616


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.148-2.211 (m, 1H); 2.380-2.452 (m, 1H); 2.744 (s, 3H); 2.755 (s, 3H); 3.803 (dd, 1H, J1=8.0, J2=13.6); 3.943-4.098 (m, 5H); 5.256 (br, 1H); 5.310 (s, 2H); 6.816-6.971 (m, 2H); 7.258-7.321 (m, 2H); 7.417-7.473 (m, 3H); 7.607-7.652 (m, 2H); 7.750 (s, 1H); 9.082 (s, 1H); 9.203 (s, 1H); 10.056 (s, 1H); 11.312 (br, 2H).


Example 9
(E)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 616


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (CDCl3, CH3OH-d4): δ 2.168-2.202 (m, 1H); 2.425-2.482 (m, 1H); 2.747 (s, 3H); 2.750 (s, 3H); 3.798-3.835 (m, 1H); 3.960-4.121 (m, 5H); 5.250 (br, 1H); 5.325 (s, 2H); 6.834-6.967 (m, 2H); 7.174-7.219 (m, 1H); 7.311-7.361 (m, 2H); 7.461 (q, 2H, J=7.2); 7.674 (s, 1H); 7.822 (s, 1H); 9.090 (s, 1H); 9.212 (s, 1H); 10.085 (s, 1H); 11.387 (s, 1H); 11.441 (s, 1H).


Example 10
(E)-N-(4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 632


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.148-2.213 (m, 1H); 2.399-2.452 (m, 1H); 2.746 (s, 3H); 2.757 (s, 3H); 3.804 (dd, 1H, J1=8.0, J2=13.6); 3.955-4.097 (m, 5H); 5.262 (br, 1H); 5.327 (s, 2H); 6.816-6.968 (m, 2H); 7.394-7.465 (m, 3H); 7.545-7.568 (m, 1H); 7.664-7.686 (m, 2H); 7.741 (s, 1H); 9.078 (s, 1H); 9.205 (s, 1H); 10.054 (s, 1H); 11.316 (br, 2H).


Example 11
(E)-N-(4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 632


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.150-2.211 (m, 1H); 2.385-2.454 (m, 1H); 2.743 (s, 3H); 2.751 (s, 3H); 3.801 (dd, 1H, J1=8.0, J2=13.2); 3.954-4.097 (m, 5H); 5.249 (br, 1H); 5.314 (s, 2H); 6.824-6.977 (m, 2H); 7.347 (d, 1H, J=8.8); 7.426-7.473 (m, 4H); 7.568 (s, 1H); 7.668 (d, 1H, J=2.4); 7.790 (s, 1H); 9.080 (s, 1H); 9.203 (s, 1H); 10.066 (s, 1H); 11.334 (s, 1H); 11.413 (br, 1H).


Example 12
(E)-N-(4-(4-(Benzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 598


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.152-2.214 (m, 1H); 2.382-5.452 (m, 1H); 2.741 (s, 3H); 2.752 (s, 3H); 3.801 (dd, 1H, J1=7.6, J2=13.6); 3.936-4.091 (m, 5H); 5.254 (br, 1H); 5.282 (s, 2H); 6.818-6.975 (m, 2H); 7.345-7.379 (m, 2H); 7.408-7.446 (m, 3H); 7.495-7.513 (m, 2H); 7.642 (d, 1H, J=2.4); 7.778 (s, 1H); 9.055 (s, 1H); 9.195 (s, 1H); 10.028 (s, 1H); 11.292 (s, 1H); 11.418 (br, 1H).


Example 13
(E)-N-(4-(4-(2 Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 623


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.150-2.211 (m, 1H); 2.374-2.443 (m, 1H); 2.749 (s, 3H); 2.759 (s, 3H); 3.805 (dd, 1H, J1=7.6, J2=13.2); 3.938-4.089 (m, 5H); 5.271 (br, 1H); 5.420 (s, 2H); 6.802-6.955 (m, 2H); 7.433 (s, 2H); 7.601-7.678 (m, 3H); 7.790 (s, 1H); 7.800 (s, 1H); 7.953 (d, 1H, J=8.0); 9.024 (s, 1H); 9.178 (s, 1H); 9.987 (s, 1H); 11.166 (br, 2H).


Example 14
(E)-N-(4-(4-(4-Tert-Butylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 654


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 1.268 (s, 9H); 2.121-2.181 (m, 1H), 2.353-2.422 (m, 1H); 2.713 (s, 3H); 2.724 (s, 3H); 3.771 (dd, 1H, J1=8.0, J2=13.6); 3.913-4.067 (m, 5H); 5.198 (s, 2H); 5.271 (br, 1H); 6.787-6.938 (m, 2H); 7.327-7.432 (s, 6H); 7.619 (d, 1H, J=2.8); 7.725 (s, 1H); 9.057 (s, 1H); 9.175 (s, 1H); 10.036 (s, 1H); 11.309 (br, 2H).


Example 15
(E)-N-(4-(4-(3 Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 623


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.150-2.197 (m, 1H); 2.380-2.451 (m, 1H); 2.748 (s, 3H); 2.758 (s, 3H); 3.803 (dd, 1H, J1=8.0, J2=13.6); 3.957-4.098 (m, 5H); 5.259 (br, 1H); 5.365 (s, 2H); 6.821-6.968 (m, 2H); 7.360 (d, 1H, J=8.8); 7.440 (d, 1H, J=8.8); 7.641-7.673 (m, 2H); 7.744 (br, 1H); 7.840 (s, 1H); 7.859 (s, 1H); 7.939 (s, 1H); 9.070 (s, 1H); 9.201 (s, 1H); 10.062 (s, 1H); 11.317 (br, 2H).


Example 16
(E)-N-(4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 632


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.147-2.209 (m, 1H); 2.381-2.452 (m, 1H); 2.742 (s, 3H); 2.753 (s, 3H); 3.801 (dd, 1H, J1=8.0, J2=13.6); 3.942-4.096 (m, 5H); 5.253 (br, 1H); 5.290 (s, 2H); 6.818-6.972 (m, 2H); 7.345 (d, 1H, J=8.8); 7.429 (dd, 1H, J1=2.4, J2=8.8); 7.512 (dd, 1H, J1=8.4, J2=14.8); 7.655 (d, 1H, J=2.4); 7.763 (s, 1H); 9.074 (s, 1H); 9.198 (s, 1H); 10.059 (s, 1H); 11.305 (s, 1H); 11.360 (br, 1H).


Example 17
(E)-N-(4-(4-(2-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 612


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.164-2.195 (m, 1H); 2.368 (s, 3H); 2.403-2.454 (m, 1H); 2.744 (s, 6H); 3.774-3.809 (m, 1H); 3.955-4.097 (m, 5H); 5.260 (s, 3H); 6.824-6.975 (m, 2H); 7.236-7.283 (m, 3H); 7.413-7.498 (m, 3H); 7.651 (s, 1H); 7.780 (s, 1H); 9.082 (s, 1H); 9.208 (s, 1H); 10.069 (s, 1H); 11.348 (br, 2H).


Example 18
(E)-N-(4-(4-(3-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 612




embedded image


Example 19
(E)-N-(4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 612


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.148-2.194 (m, 1H); 2.325 (s, 3H); 2.396-2.448 (m, 1H); 2.743 (s, 3H); 2.754 (s, 3H); 3.801 (dd, 1H, J1=8.0, J2=13.6); 3.953-4.094 (m, 5H); 5.221 (s, 2H); 5.256 (br, 1H); 6.815-6.968 (m, 2H); 7.220 (s, 1H); 7.239 (s, 1H); 7.331-7.423 (m, 4H); 7.632 (d, 1H, J=2.4); 7.739 (s, 1H); 9.059 (s, 1H); 9.190 (s, 1H); 10.049 (s, 1H); 11.259 (br, 1H); 11.335 (br, 1H).


Example 20
(E)-N-(3-Cyano-4-((R)-1-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 486




embedded image


Example 21
(E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 599




embedded image


Example 22
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Acrylamide



embedded image


To a reaction flask were added 6-amino-4-(3-chloro-4-(pyridin-2-yl-methoxy) phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-3-carbonitrile (100 mg, 0.205 mmol), pyridine (0.3 ml), DMAP (20 mg) and THF (10 ml). The mixture was cooled to the temperature of 0° C. To the mixture was added acryloyl chloride (20 mg, 0.22 mmol). The resultant mixture was stirred for 30 min at the temperature of 0° C. Then the mixture was warmed to the room temperature and stirred for 5 hr. After the reaction finished, the resultant mixture was filtered and the filtrate was rotary-evaporated to dryness to give a solid. The solid was dissolved in ethyl acetate. The solution was washed with saturated sodium carbonate once, acetic acid solutioni (10%) once and saturated saline once, successively. The resulting solution was dried over anhydrous magnesium sulfate, filtered and rotary-evaporated to dryness to give a crude product. The product was purified with thin layer chromatography.


MS (M+1): 542.


The compounds of Examples 23-28 were prepared according to the process of Example 22.


Example 23
(E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)but-2-Enamide

MS (M+1): 556




embedded image


Example 24
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-3-Methylbut-2-Enamide

MS (M+1): 570




embedded image


Example 25
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Propiolamide

MS (M+1): 540




embedded image


Example 26
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Propiolamide

MS (M+1): 544




embedded image


Example 27
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Acetamide



embedded image


MS (M+1): 530


H1-NMR (DMSO-d9): δ 2.181 (br, 4H), 62.326-2.393 (m, 1H); 3.805 (dd, 1H, J1=7.2, J2=12.8); 3.955 (dd, 1H, J1=7.6, J2=14.4); 4.017-4.058 (m, 2H); 5.279 (br, 1H); 5.319 (s, 2H); 7.303 (s, 1H); 7.375-7.444 (m, 2H); 7.521 (s, 1H); 7.593 (d, 1H, J=8.4); 7.893 (t, 1H, J=8.0); 8.617 (s, 1H); 8.752 (s, 1H); 8.967 (s, 1H); 9.442 (s, 1H).


Example 28
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Benzamide



embedded image


MS (M+1): 592


H1-NMR (DMSO-d6): δ 2.087-2.132 (m, 1H); 2.287-2.376 (m, 1H); 3.769-3.820 (m, 1H); 3.859-3.948 (m, 2H); 4.019 (dd, 1H, J1=4.4; J2=10.0); 5.298 (s, 2H); 5.348 (br, 1H); 7.234-7.267 (m, 2H); 7.360-7.390 (m, 1H); 7.438 (s, 1H) 7.449 (s, 1H); 7.556-7.650 (m, 4H); 7.877 (t, 1H, J=8.0); 7.889 (d, 2H, J=7.2); 8.324 (s, 1H); 8.527 (s, 1H); 8.603 (d, 1H, J=4.4); 8.821 (s, 1H); 9.757 (s, 1H); 9.808 (s, 1H).


Example 29
Tert-Butyl 4-(2-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl-Amino)-2-Oxo Ethylidene)Piperidine-1-Carboxylate



embedded image


To a single-neck reaction flask (100 ml) were added 2-(1-(tert-butyloxycarbonyl)piperidin-4-ylidene)acetic acid (1 g) and anhydrous THF (20 ml). The mixture was stirred to dissolve and cooled to the temperature of −5° C. To the mixture were added isobutyl chloroformate (0.6 ml) and N-methylmorphine (0.5 ml). The resultant mixture was stirred for 20 min. 6-amino-4-(3-chloro-4-(pyridin-2-yl-methoxy) phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-3-carbonitrile (2 g, 4.1 mmol) was dissolved in anhydrous pyridine (20 ml). The resulting solution was added into the reaction flask in an ice-water bath. After the reaction finished, the solvent was rotary-evaporated to dryness. To the residue were added chloroform and water. The solution was stood for separation. The chloroform layer was wash once with saturated saline solution, dried over anhydrous magnesium sulfate, filtered and rotary-evaporated to dryness to give a crude product. The product was recrystalized with ethanol.


MS (M+1): 711.


Example 30
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide



embedded image


The product of Example 29 (100 mg) was dissolved in 20% TFA/DCM solution (20 ml). The solution was stirred for 2 hr at the room temperature. Then the solvent was rotary-evaporated to dryness. The residue was dissolved in chloroform. The organic phase was washed with saturated sodium carbonate once and saline solution once, successively. The resultant substance was dried over anhydrous magnesium sulfate and filtered. Then the solvent was rotary-evaporated to dryness to give the target compound.


MS (M+1): 611.


The compound was prepared as hydrochloride according to the process of Example 3 to determine, H1-NMR (DMSO-d6): δ 2.168-2.209 (m, 1H); 82.408-2.451 (m, 1H); 2.576 (bs, 2H); 3.133 (bs, 2H); 3.211 (s, 2H); 3.259 (s, 2H); 3.805-3.830 (m, 1H); 3.939-4.114 (m, 5H); 5.258 (br, 1H); 5.497 (s, 2H); 6.374 (s, 1H); 7.401 (d, 2H, J=8.4); 7.401 (d, 1H, J=8.4); 7.464 (d, 1H, J=8.4); 7.678 (br, 1H); 7.705 (s, 1H); 7.783-7.852 (m, 2H); 8.173-8.214 (m, 1H); 8.785 (s, 1H); 9.090 (s, 1H); 9.213 (s, 1H); 9.468 (br, 1H); 9.710 (s, 1H); 11.342 (br, 2H).


Example 31
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-Methylpiperidin-4-Ylidene)Acetamide



embedded image


To a single-neck reaction flask (50 ml) was added the product of Example 30 (20 mg). The product was dissolved in acetonitrile (10 ml). To the resultant solution were added anhydrous potassium carbonate powders (20 mg) and methyl iodide (5 mg). The resulting mixture was stirred at the room temperature. After the reaction finished, the resultant substance was filtered. The filtrate was rotary-evaporated to dryness to give a crude product. The crude product was purified with thin layer chromatography (developing solvent: chloroform:methanol=92:8).


MS (M+1): 625.


The compounds of Examples 32-37 were prepared according to the process of Example 31.


Example 32
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-Ethylpiperidin-4-Ylidene)Acetamide

MS (M+1): 639




embedded image


Example 33
2-(1-Benzylpiperidin-4-Ylidene)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)Acetamide

MS (M+1): 701




embedded image


Example 34
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-(2-Methoxyethyl) Piperidin-4-Ylidene)Acetamide

MS (M+1): 669




embedded image


Example 35
Methyl 2-(4-(2-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl-Amino)-2-Oxoethylidene)Piperidin-1-yl)Acetate

MS (M+1): 683




embedded image


Example 36
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-Isopropylpiperidin-4-Ylidene)Acetamide

MS (M+1): 653




embedded image


Example 37
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-(2-Hydroxyethyl) Piperidin-4-Ylidene)Acetamide

MS (M+1): 655




embedded image


Example 38
(E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Pyrrolidin-3-Ylidene)Acetamide

MS (M+1): 597




embedded image


In accordance with the process of Examples 29 and 30, the target compound was prepared by replacing 2-(1-(tert-butoxycarbonyl)piperidin-4-ylidene)acetic acid with (E/Z)-2-(1-(tert-butoxycarbonyl)pyrrolin-3-ylidene)acetic acid.


Example 39
N1-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-N4-(2-(2-(Dimethylamino)Ethoxy)Ethyl)Fumaramide



embedded image


To a single-neck reaction flask (50 ml) were added 6-amino-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7 (tetrahydrofuran-3-yl-oxy)quinoline-3-carbonitrile (20 mg, 0.041 mmol), maleic acid (4 mg), pyridine (0.1 ml) and THF (5 ml), The mixture was stirred at the room temperature. After the reaction finished, the solvent was rotary-evaporated to dryness. The residue was purified with thin layer chromatography (developing solvent: chloroform:methanol:acetic acid=450:50:2). The resulting pure product was dissolved in anhydrous THF. Aminoethoxyethanol was added into the resultant solution. The mixture was cooled in an ice bath. DCC (8 mg) was dissolved in anhydrous THF (5 ml). The resulting solution was dropwise added into the reaction flask under continuously stirring. After the reaction finished, the solvent was rotary-evaporated to dryness to give a crude product. The crude product was dissolved in pyridine (5 ml). p-toluene sulfonyl chloride (7 mg) was added into the resultant solution. The mixture was stirred at the room temperature. After the reaction finished, the solvent was rotary-evaporated to dryness. The resulting crude product was dissolved in chloroform. The organic phase was washed with saturated sodium carbonate once, HCl (1 N) once and saturated saline solution once, successively. The resultant substance was dried over anhydrous magnesium sulfate, filtered and rotary-evaporated to dryness to give a prodcut. The above resulting product was dissolved in pyridine (10 ml). Dimethylamine was added into the solution. The resultant mixture was stirred at the room temperature. After the reaction finished, the solvent was rotary-evaporated to dryness. The resultant substance was purified with thin layer chromatography, (developing solvent: chloroform:methanol=9:1) to give the targent compound.


MS (M+1): 700.


Example 40
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-(2-(2-(2-Hydroxyethoxy)Ethylamino)Acetyl)Piperidin-4-Ylidene) Acetamide



embedded image


To a single-neck reaction flask (50 ml) were added N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide (20 mg, 0.041 mmol) and anhydrous THF (10 ml). The solution was cooled in an ice bath. Chloroacetyl chloride (4.5 mg) and anhydrous triethylamine (0.02 ml) were added into the solution. The resultant mixture was continuously stirred. After the reaction finished, the solvent was rotary-evaporated to dryness. The residue was dissolved in chloroform. The solution was washed with water three times and with saturated saline solution once. The resultant solution was dried over anhydrous magnesium sulfate, filtered and rotary-evaporated to dryness. The above resulting crude product was dissolved in acetonitrile (10 ml). To the solution were added aminoethoxyethanol (4 mg) and triethylamine (0.2 ml). The mixture was stirred at the room temperature. After the reaction finished, the solvent was rotary-evaporated to dryness. The resultant substance was purified with thin layer chromatography (developing solvent: chloroform methanol=9:1.


MS (M+1): 756.


Example 41
N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(1-(Methylsulfonyl) Piperidin-4-Ylidene)Acetamide



embedded image


In accordance with the process of Example 22, the compound was prepared by replacing acryloyl chloride with methylsulfonyl chloride.


MS (M+1): 689.


The compounds of Examples 42-132 were prepared according to the processes of Examples 29 and 30.


Example 42
N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide



embedded image


MS (M+1): 522


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.143-2.214 (m, 1H); 52.372-2.443 (m, 1H); 2.561 (bs, 2H); 3.123 (bs, 2H); 3.169-3.240 (m, 4H); 3.803 (dd, 1H, J1=8.0, J2=13.6); 3.927-4.099 (m, 5H); 5.258 (br, 1H); 6.357 (s, 1H); 7.507 (br, 1H); 7.567 (t, 1H, J=9.0); 7.744-7.794 (m, 2H); 9.069 (s, 1H); 9.170 (s, 1H); 9.409 (br, 2H); 9.651 (s, 1H); 11.248 (s, 1H).


Example 43
N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 494




embedded image


Example 44
N-(4-(3-Bromophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 548




embedded image


Example 45
N-(4-(4-(2-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 46
N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 47
N-(4-(4-(4-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 48
N-(4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 644




embedded image


Example 49
N-(4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 644




embedded image


Example 50
N-(4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 644




embedded image


Example 51
N-(4-(4-(2-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 624




embedded image


Example 52
N-(4-(4-(3-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 624




embedded image


Example 53
N-(4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 624




embedded image


Example 54
N-(4-(4-(2-Methoxybenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 640




embedded image


Example 55
N-(4-(4-(3-Methoxybenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 640




embedded image


Example 56
N-(4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 640




embedded image


Example 57
N-(4-(4-(2-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 635




embedded image


Example 58
N-(4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 635




embedded image


Example 59
N-(4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 635




embedded image


Example 60
N-(4-(4-(4-Tert-Butylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 666




embedded image


Example 61
N-(4-(4-(Benzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 610




embedded image


Example 62
N-(4-(1H-Indol-5-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 509




embedded image


Example 63
N-(4-(4-(2-Chlorobenzyloxy)-3-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 64
N-(4-(4-(3-Chlorobenzyloxy)-3-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 65
N-(4-(4-(4-Chlorobenzyloxy)-3-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 628




embedded image


Example 66
N-(3-Cyano-4-((S)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 498




embedded image


Example 67
N-(3-Cyano-4-((R)-1-Phenylethylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 498




embedded image


Example 68
N-(4-(1-Benzyl-1H-Indol-5-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 599




embedded image


Example 69
N-(4-(1-(3-Cyanobenzyl)-1H-Indol-5-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 624




embedded image


Example 70
N-(4-(1-(3-Methoxybenzyl)-1H-Indol-5-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 629




embedded image


Example 71
N-(4-(1-(3-Chlorobenzyl)-1H-Indol-5-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 633




embedded image


Example 72
N-(3-Cyano-4-(Indolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 496




embedded image


Example 73
N-(4-(6-Chloroindolin-1-yl)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 530




embedded image


Example 74
N-(3-Cyano-4-(6-Fluoroindolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 514




embedded image


Example 75
N-(4-(4-Chloroindolin-1-yl)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 530




embedded image


Example 76
N-(3-Cyano-4-(3,4-Dihydroquinolin-1(2H)-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 510




embedded image


Example 77
N-(3-Cyano-4-(6-Methyl-3,4-Dihydroquinolin-1(2H)-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 524




embedded image


Example 78
N-(3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)-4-(7-(Trifluoromethyl)-3,4-Dihydroquinolin-1(2H)-yl)-Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 578




embedded image


Example 79
N-(4-(6-(Benzyloxy)Indolin-1-yl)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 602




embedded image


Example 80
Methyl 1-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl)-Indoline-2-Carboxylate

MS (M+1): 554




embedded image


Example 81
N-(3-Cyano-4-(2-(Hydroxymethyl)Indolin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 526




embedded image


Example 82
N-(4-(6-(1HH-Pyrrol-1-yl)Indolin-1-yl)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 561




embedded image


Example 83
N-(3-Cyano-4-(Octahydroindol-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 502




embedded image


Example 84
N-(3-Cyano-4-(Pyrimidin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 472




embedded image


Example 85
N-(2-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl) Benzamide

MS (M+1): 591




embedded image


Example 86
N-(2-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-5-yl)-4-(Dimethylamino)Benzamide

MS (M+1): 634




embedded image


Example 87
N-(3-Cyano-4-(5 (Phenylsulfonamido)Pyrimidin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 627




embedded image


Example 88
N-(5-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl) Benzamide

MS (M+1): 591




embedded image


Example 89
N-(5-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl) Furan-2-Carboxamide

MS (M+1): 581




embedded image


Example 90
N-(5-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl) Thiophene-2-Carboxamide

MS (M+1): 597




embedded image


Example 91
N-(5-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)Pyrimidin-2-yl) Cyclohexylcarboxamide

MS (M+1): 597




embedded image


Example 92
5-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Pyrimidin-2-Carboxamide

MS (M+1): 621




embedded image


Example 93
N-(3-Cyano-4-(Pyridin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 471




embedded image


Example 94
6-(3-Cyano-6-(2-(Piperidin-4-Ylidene)Acetamido)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-4-yl-Amino)-N-(4-Methoxyphenyl)Nicotinamide

MS (M+1): 620




embedded image


Example 95
N-(3-Cyano-4-(Pyridin-3-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 471




embedded image


Example 96
N-(3-Cyano-4-(Pyridin-4-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 471




embedded image


Example 97
N-(4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 577




embedded image


Example 98
N-(3-Cyano-4-(Pyrazin-2-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 472




embedded image


Example 99
N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 549




embedded image


Example 100
N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 521




embedded image


Example 101
N-(4-(4-(Benzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 637




embedded image


Example 102
N-(4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 671




embedded image


Example 103
N-(4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 671




embedded image


Example 104
N-(4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 671




embedded image


Example 105
N-(4-(4-(4-Bromobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 715




embedded image


Example 106
N-(4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 651




embedded image


Example 107
N-(4-(4-(4-Methoxybenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 667




embedded image


Example 108
N-(4-(4-(4-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 662




embedded image


Example 109
N-(4-(4-(4-Ethylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 665




embedded image


Example 110
N-(4-(4-(4-Ethoxybenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 681




embedded image


Example 111
N-(4-(3-Chloro-4-Phenoxyphenylamino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 623




embedded image


Example 112
N-(3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-2-yl-Amino) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 498




embedded image


Example 113
N-(3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-3-yl-Amino) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 498




embedded image


Example 114
N-(3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)-4-(Pyridin-4-yl-Amino) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 498




embedded image


Example 115
N-(4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene) Acetamide

MS (M+1): 604




embedded image


Example 116
N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 529




embedded image


Example 117
N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 501




embedded image


Example 118
N-(3-Cyano-4-(4 Phenoxyphenylamino)-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 569




embedded image


Example 119
N-(4-(4-(Benzyloxy)Phenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 583




embedded image


Example 120
N-(4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 651




embedded image


Example 121
N-(4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 631




embedded image


Example 122
N-(4-(4-(4-Methoxybenzyloxy)Phenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 613




embedded image


Example 123
N-(4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 642




embedded image


Example 124
N-(3-Cyano-4-(Pyridine-2-yl-Amino)-7-(Pyridin-4-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 478




embedded image


Example 125
N-(3-Cyano-7-(Pyridin-4-yl-Oxy)-4-(Pyrimidin-2-yl-Amino) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 479




embedded image


Example 126
N-(4-(6-(Benzyloxy)Pyridin-3-yl-Amino)-3-Cyano-7-(Pyridin-4-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 584




embedded image


Example 127
N-(4-(6-(3-Chlorobenzyloxy)Pyridin-3-yl-Amino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 618




embedded image


Example 128
N-(3-Cyano-4-(6 (Phenoxypyridin-3-yl-Amino)-7-(Pyridin-4-yl-Oxy) Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 570




embedded image


Example 129
N-(4-(3-Chloro-4-(Pyridine-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Piperidin-4-Ylidene)Acetamide

MS (M+1): 611




embedded image


Example 130
(E/Z)-N-(3-Cyano-4-(6-(Phenoxypyridin-3-yl-Amino)-7-(Pyridin-4-yl-Oxy)Quinolin-6-yl)-2-(Pyrrolidin-3-Ylidene)Acetamide

MS (M+1): 556




embedded image


Example 131
(E/Z)-N-(3-Cyano-7-(1-Methylpiperidin-4-yl-Oxy)-4-(6-(Phenoxypyridin-3-yl-Amino)Quinolin-6-yl)-2-(Pyrrolidin-3-Ylidene)Acetamide

MS (M+1): 576




embedded image


Example 132
(E/Z)-N-(3-Cyano-4-(6-(Phenoxypyridin-3-yl-Amino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-2-(Pyrrolidin-3-Ylidene)Acetamide

MS (M+1): 549




embedded image


Example 133
4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-6-Methoxy-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


The target compound was prepared according to the process disclosed in Chinese patent No. ZL 03815201.0.


MS (M+1): 503.


Example 134
4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-6-(Piperidin-1-yl)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile



embedded image


To a single-neck reaction flask (50 ml) were added 6-amino-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-3-carbonitrile (20 mg, 0.041 mmol), anhydrous potassium carbonate powder (10 mg) and chloroform (5 ml). To the flask was added 1,5-dibromopentane (10 mg). The mixture was stirred at the room temperature. After the reaction finished, the mixture was filtered. The filtrate was rotary-evaporated to dryness. The residue was purified with thin layer chromatography (developing solvent: chloroform:methanol=9:1) to give a pure product.


MS (M+1): 556.


The compounds of Examples 135-137 were prepared according to the process disclosed in Chinese patent No. ZL 03815201.0.


Example 135
4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-6-(2-Methoxyethoxy)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile

MS (M+1): 547




embedded image


Example 136
4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-6-(2-(2-Methoxyethoxy)Ethoxy)-7-(Tetrahydrofuran-3-yl-Oxy) Quinoline-3-Carbonitrile

MS (M+1): 591




embedded image


Example 137
4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-6-(2-Morpholinoethoxy)-7-(Tetrahydrofuran-3-yl-Oxy)Quinoline-3-Carbonitrile

MS (M+1): 602




embedded image


The compounds of Examples 138-206 were prepared according to the process of Example 2.


Example 138
(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 599


The compound was prepared as hydrochloride according to the process of Example 3 so as to determine the specific rotation (the measurement conditions: 20° C., sodium lamp, D light, 589 nm): +51.4°


Example 139
(R,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 599




embedded image


Example 140
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 482


The compound was prepared as hydrochloride according to the process of Example 3 to determine H1-NMR (DMSO-d6): δ 2.144-2.232 (m, 1H); 2.375-2.469 (m, 1H); 2.741 (s, 3H); 2.756 (s, 3H); 3.765-3.837 (dd, 1H, J1=10.8; J2=16.0); 3.930-4.109 (m, 5H); 4.327 (s, 1H); 5.246 (br, 1H); 6.839-6.997 (m, 2H); 7.508-7.511 (m, 3H); 7.576 (s, 1H); 7.848 (s, 1H); 9.080 (s, 1H); 9.226 (s, 1H); 10.081 (s, 1H); 11.356 (br, 1H); 11.505 (br, 1H).


Specific rotation (the measurement conditions: 20° C., sodium lamp, D light, 589 nm): +10.3°


Example 141
(R,E)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 482




embedded image


Example 142
(S,E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 510


The compound was prepared as hydrochloride according to the process of Example 3 so as to determine the specific rotation (the measurement conditions: 20° C., sodium lamp, D light, 589 nm): +21.2°


Example 143
(S,E)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 510




embedded image


Example 144
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide

MS (M+1): 522




embedded image


Example 145
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 524




embedded image


Example 146
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Tert-Butylaimino)-but-2-Enamide

MS (M+1): 510




embedded image


Example 147
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Benzylamino)-but-2-Enamide

MS (M+1): 544




embedded image


Example 148
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(6-Hydroxyhexylaimino)-but-2-Enamide

MS (M+1): 552




embedded image


Example 149
(S,E)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylbenzylamino)-but-2-Enamide

MS (M+1): 558




embedded image


Example 150
(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 627




embedded image


Example 151

(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide


MS (M+1): 639




embedded image


Example 152
(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 641




embedded image


Example 153
(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Tert-Butylamino)-but-2-Enamide

MS (M+1): 627




embedded image


Example 154
(S,E)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(6-Hydroxyhexyl Amino)-but-2-Enamide

MS (M+1): 671




embedded image


Example 155
(S,E)-N-(4-(4-(Benzyloxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 564




embedded image


Example 156
(S,E)-N-(4-(3-Bromophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 536




embedded image


Example 157
(S,E)-N-(4-(4-(2-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 616




embedded image


Example 158
(S,E)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 616




embedded image


Example 159
(S,E)-N-(4-(4-(2-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 632




embedded image


Example 160
(S,E)-N-(4-(4-(3-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 632




embedded image


Example 161
(S,E)-N-(4-(4 (Benzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 598




embedded image


Example 162
(S,E)-N-(4-(4-(2-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 623




embedded image


Example 163
(S,E)-N-(4-(4-(4-Tert-Butylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 654




embedded image


Example 164
(S,E)-N-(4-(4-(3-Cyanobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 623




embedded image


Example 165
(S,E)-N-(4-(4-(4-Chlorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 632




embedded image


Example 166
(S,E)-N-(4-(4-(2-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 612




embedded image


Example 167
(S,E)-N-(4-(4-(4-Methylbenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 612




embedded image


Example 168
(S,E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 538




embedded image


Example 169
(S,E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide

MS (M+1): 550




embedded image


Example 170
(S,E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 552




embedded image


Example 171
(S,E)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 644




embedded image


Example 172
(S,E)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide

MS (M+1): 656




embedded image


Example 173
(S,E)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 658




embedded image


Example 174
(E)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Pyridin-3-yl-Oxy)Quinolin-6-yl)4-(Dimethylamino)-but-2-Enamide



embedded image


MS (M+1): 517.


The compound was prepared as hydrochloride according to the process of Example 3 to determine, H1-NMR (DMSO-d6): δ 2.732 (s, 3H); 2.744 (s, 3H); 3.945 (t, 2H, J1=6.0); 3.955-4.097 (m, 5H); 6.784-6.989 (m, 2H); 7.544-7.621 (m, 3H); 7.781-7.853 (m, 2H); 8.029 (dd, 1H, J1=1.6, J2=8.8); 8.695 (d, 1H, J=4.8); 8.773 (d, 1H, J=2.8); 9.079 (s, 1H); 9.346 (s, 1H); 10.739 (s, 1H); 11.428 (br, 1H).


Example 175
(E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 600




embedded image


Example 176
(E/Z)-N-(4-(3-Chloro-4-Fluorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 510




embedded image


Example 177
(E/Z)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 482




embedded image


Example 178
(E/Z)-N-(4-(4-(Benzyloxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 564




embedded image


Example 179
(E/Z)-N-(4-(3-Bromophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 536




embedded image


Example 180
(E/Z)-N-(4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethylamino)-but-2-Enamide

MS (M+1): 616




embedded image


Example 181
(S,E/Z)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 510




embedded image


Example 182
(S,E/Z)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide

MS (M+1): 522




embedded image


Example 183
(S,E/Z)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 524




embedded image


Example 184
(S,E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethylamino)-but-2-Enamide

MS (M+1): 627




embedded image


Example 185
(S,E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Piperidin-1-yl)-but-2-Enamide

MS (M+1): 639




embedded image


Example 186
(S,E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Morpholin-4-yl)-but-2-Enamide

MS (M+1): 641




embedded image


Example 187
(S,E/Z)-N-(4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-3-Cyano-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(2-Methoxyethylamino)-but-2-Enamide

MS (M+1): 629




embedded image


Example 188
(S,E)-N-(3-Cyano-4-(3 Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethanolamino)-but-2-Enamide

MS (M+1): 542




embedded image


Example 189
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylmethoxyethylamino)-but-2-Enamide

MS (M+1): 526




embedded image


Example 190
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylethanolamino)-but-2-Enamide

MS (M+1): 512




embedded image


Example 191
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethoxyethylamino)-but-2-Enamide

MS (M+1): 570




embedded image


Example 192
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethanolamino)-but-2-Enamide

MS (M+1): 570




embedded image


Example 193
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylmethoxyethylamino)-but-2-Enamide

MS (M+1): 554




embedded image


Example 194
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylethanolamino)-but-2-Enamide

MS (M+1): 540




embedded image


Example 195
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethoxyethylamino)-but-2-Enamide

MS (M+1): 598




embedded image


Example 196
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-4-(Diethanolamino)-but-2-Enamide

MS (M+1): 676




embedded image


Example 197
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-4-(N-Methylmethoxyethylamino)-but-2-Enamide

MS (M+1): 660




embedded image


Example 198
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-4-(N-Methylethanolamino)-but-2-Enamide

MS (M+1): 646




embedded image


Example 199
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy) Quinolin-6-yl)-4-(Dimethoxyethylamino)-but-2-Enamide

MS (M+1): 704




embedded image


Example 200
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Diethanolamino)-but-2-Enamide

MS (M+1): 589




embedded image


Example 201
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylmethoxy Ethylamino)-but-2-Enamide

MS (M+1): 643




embedded image


Example 202
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methylethanol Amino)-but-2-Enamide

MS (M+1): 639




embedded image


Example 203
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(Dimethoxyethyl Amino)-but-2-Enamide

MS (M+1): 687




embedded image


Example 204
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(6-Hydroxyhexyl Amino)-but-2-Enamide

MS (M+1): 671




embedded image


Example 205
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(6-Hydroxyhexylamino)-but-2-Enamide

MS (M+1): 582




embedded image


Example 206
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(6-Hydroxyhexylamino)-but-2-Enamide

MS (M+1): 688




embedded image


Example 207
(S,E)-N-(3-Cyano-4-(3-Ethynylphenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methyl-6-Amino-1-Hexanolyl)-but-2-Enamide

MS (M+1): 566




embedded image


(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(6-amino-1-hexanolyl)-but-2-enamide (552 mg, 0.001 mol) was dissolved in DMF (15 ml). The solution was stirred homogeneously. To the solution were added methyl iodide (156 mg (1.1 mmol), anhydrous potassium carbonate (276 mg, 2 mmol) and tetrabutyl ammonium iodide (11 mg, 0.03 mmol). The resulting mixture was stirred in the dark at the room temperature. After 48 hr, the reaction was stopped. The reaction solution was added into saturated sodium bicarbonate (150 ml). The mixture was extracted with ethyl acetate (150 ml) once. The organic phase was retained. Anhydrous magnesium sulfate was added into the organic phase for half-hour. After half-hour, the drying agent was removed. The organic phase was concentrated in vacuo to give a yellow solid. The solid was purified with column chromatography (eluent:chloroform:methanol=9:1) to give a yellow solid (342 mg). Yield: 60.5%.


The compounds of Examples 208-210 were prepared according to the process of Example 207.


Example 208
(S,E)-N-(3-Cyano-4-(3-Chloro-4-Fluoro-Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methyl-6-Amino-1-Hexanolyl)-but-2-Enamide

MS (M+1): 596




embedded image


Example 209
(S,E)-N-(3-Cyano-4-(4-(3-Fluorobenzyloxy)-3-Chlorophenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methyl-6-Amino-1-Hexanolyl)-but-2-Enamide

MS (M+1): 702




embedded image


Example 210
(S,E)-N-(3-Cyano-4-(3-Chloro-4-(Pyridin-2-yl-Methoxy)Phenylamino)-7-(Tetrahydrofuran-3-yl-Oxy)Quinolin-6-yl)-4-(N-Methyl-6-Amino-1-Hexanolyl)-but-2-Enamide

MS (M+1): 485




embedded image


BIOLOGICAL EXAMPLES

Abbreviations used in the following Biological Examples are as follows: EGFR-TK: intracellular epidermal growth factor receptor phosphorylase; A431 (human epithelial gland cancer cell strain); A549: human lung cancer cell strain; LoVo: human intestinal cancer cell strain; NCI-H460: human large cell lung cancer cell strain; NCI-N87: human gastric cancer cell strain; Sk-Br-3: human breast cancer cell strain; SW620: human colorectal cancer cell strain; BT-474: human breast cancer cell strain; CCRF-CEM/T: human acute lymphocytic cell leukemia cell resistant to paclitaxel; Fadu: human head and neck cancer cell strain; BxPC-3: human pancreatic cancer cell strain; AsPC-1: human pancreatic cancer cell strain; SK-OV-3: human ovarian cancer cell strain; NCI-H358: human non-small cell lung cancer cell strain; NCI-H1650: human non-small cell lung cancer cell strain; MDA-MB-453: human breast cancer cell strain; PGT: polyglutamic acid tyrosine; PBS: phosphate buffer, pH 7.4; ATP: triphosadenine; TKB: tyrosine kinase reaction buffer; SDS: sodium dodecyl sulfate; PBST: PBS containing 0.05% Tween 20; BSA: bovine serum albumin; HRP: horseradish peroxidase; TMB: 3,3′,5,5′-tetramethyl benzidine; DTT: dithiothreitol; ddH2O: double distilled water; MTT: tetrazole; DMEM: Dulbecco's Modified Eagle's Medium; F12: F-12 Nutrient Mixture (Ham); EDTA: ethylene diamine tetraacetic acid; RPMI-1640: RPMI-1640 Medium; FBS: fetal bovine serum; SRB: sulforhodamine; Tris: trihydroxy methyl amino methane; EMEM: Minimum Essential Medium with Earle's salts; NEAA: non-essential amino acid; McCoy's 5A: McCoy's 5A Medium; HEPES: hydroxyethylpiperazine ethanesulfonic acids; DMSO: dimethyl sulfoxide.


Biological Example 1
A431 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

1. Cell strains: A431 (human epithelial adenocarcinoma cell strains);


2. MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 45% DMEM, 45% F12+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out, and 2 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out, and the culture plate was placed in an incubator. Digestion was carried out for about 15 min at 37° C.;


5. 4 ml of complete culture solution was added in the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 2500 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising the compound was added in each well, and further incubated for 68 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 4 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added into each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cells after growth in test wells with a compound;


NC: Background OD value of blank wells without a compound and cells;


IC50: concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 1







Growth Inhibition Ratio of Some Compounds (1 μM) in Examples on A431 Cells













Growth

Growth

Growth



Inhibition

Inhibition

Inhibition


Compound
Ratio (%)
Compound
Ratio (%)
Compound
Ratio (%)















Example 3
85
Example 4
80
Example 5
85


Example 8
91
Example 9
90
Example 13
89


Example 15
91
Example 138
89
Example 139
90


Example 140
87
Example 141
80
Example 142
75


Example 143
70
Example 150
84
Example 155
82


Example 157
72
Example 158
77
Example 166
69


Example 167
75
Example 176
79
Example 177
81


Example 180
73
Example 185
70
Example 190
79
















TABLE 2







Growth Inhibition Activity (IC50) of Some


Compounds in Examples on A431 Cells













IC50

IC50

IC50


Compounds
(μM)
Compounds
(μM)
Compounds
(μM)















Example 4
0.068
Example 5
0.024
Example 7
0.160


Example 8
0.145
Example 9
0.135
Example 10
0.222


Example 11
0.184
Example 12
0.147
Example 13
0.158


Example 174
0.95
Example 15
0.37
Example 16
0.358


Example 17
0.194
Example 19
0.39
Example 21
0.081


Example 138
0.062
Example 139
0.093
Example 140
0.026


Example 141
0.050
Example 142
0.045
Example 143
0.059


Example 144
0.026
Example 145
0.071
Example 146
0.053


Example 149
0.089
Example 150
0.027
Example 151
0.061


Example 152
0.062
Example 153
0.053
Example 154
0.162









Biological Example 2
BT-474 Cell Growth Inhibition Test (SRB Assay)
I. Assay Materials

1. Cell strains: BT-474 (human breast tumor cell strains);


2. SRB: available from Sigmaaldrich, goods number: 59012, lot number: 047K3751. SRB was reserved at the room temperature. 0.4% (w/v) of working solution was formulated with 1% of glacial acetic acid. The solution was reserved at the temperature of 4° C.; anti-tumor compounds; DMSO.


II. Reagents and Consumable Materials

Culture medium (90% EMEM+10% FBS+0.1 mM NEAA, reserved at 4° C.);


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


FBS (fetal bovine serum);


Tris;


Glacial acetic acid;


96-well cell culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected. The culture solution was sucked out. The plate was washed with 5 mL of PBS 1-2 times;


2. PBS was sucked out, and 1.5 m of 0.25% pancreatin was added to infiltrate the cells for 30 s;


3. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 5 min at 37° C.;


4. 3 mL of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform single cell suspension;


5. Cell suspension was counted. The suspension was diluted to 1×105/ml. The resulting suspension was developed uniformly on a plate. The plate was incubated under 5% CO2 at 37° C. overnight. In day 2, 80 μL of complete culture medium was added in each well and then 20 μL of culture solution comprising a compound was added. The mixture was incubated for 70 h under 5% CO2 at 37° C.


6. The culture solution was sucked out. 100 uL of TCA fixed cells which were diluted to 10% were added to each well. The plate was kept in a refrigerator for 1 h at 4° C.


7. TCA stationary liquid was sucked out. Each well was washed with 150 μL of ddH2O five times;


8. After the stationary liquid was cleansed, the plate was dried in the air at the room temperature;


9. 60 μL of SRB staining solution was added in each well. The well was stained for 15 min at the room temperature;


10. The SRB staining solution was sucked out. Each well was washed with 150 μL of 1% glacial acetic acid five times;


11. After the SRB staining solution was cleansed, the plate was dried in the air at the room temperature;


12. 100 μL of 10 mM Tris was added in each well. The plate was vibrated to dissolve out SRB;


13. OD values were determined at 570 nM.


IV. Assay Results

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 3







Growth Inhibition Ratio of Some Compounds


(1 μM) in Examples on BT-474 Cells













Growth

Growth

Growth



Inhibition

Inhibition

Inhibition


Compounds
Ratio (%)
Compounds
Ratio (%)
Compounds
Ratio (%)















Example 4
79
Example 5
85
Example 8
79


Example 9
78
Example 13
75
Example 15
71


Example 16
72
Example 138
82
Example 139
81


Example 140
85
Example 141
89
Example 142
89


Example 143
87
Example 145
75
Example 147
75


Example 149
79
Example 150
82
Example 151
80


Example 152
78
Example 156
70
Example 158
82


Example 162
80
Example 166
69
Example 167
75


Example 168
86
Example 169
80
Example 171
84


Example 172
62
Example 173
67
Example 176
89


Example 177
87
Example 180
78
Example 181
87


Example 183
75
Example 184
83
Example 186
80


Example 189
85
Example 192
85
Example 198
78


Example 201
80
Example 203
82
Example 205
75
















TABLE 4







Growth Inhibition Activity (IC50) of Some


Compounds in Examples on BT-474 Cells













IC50

IC50

IC50


Compounds
(μM)
Compounds
(μM)
Compounds
(μM)















Example 4
0.140
Example 5
0.041
Example 7
0.309


Example 9
0.0052
Example 10
0.0099
Example 11
0.0079


Example 12
0.008
Example 13
0.011
Example 189
0.108


Example 15
0.0081
Example 16
0.035
Example 17
0.017


Example 19
0.032
Example 190
0.267
Example 21
0.0036


Example 138
0.003
Example 139
0.0039
Example 140
0.033


Example 141
0.096
Example 142
0.133
Example 143
0.149


Example 144
0.129
Example 145
0.654
Example 146
0.236


Example 149
0.212
Example 150
0.0049
Example 151
0.0065


Example 152
0.0104
Example 153
0.0072
Example 154
0.012


Example 191
0.390









Biological Example 3
A549 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

1. Cell strains: A4549 (human lung caner cell strains);


2. MTT; antitumor compounds; DMSO.


II. Reagents and Consumable Materials

Culture medium: 90% F12K+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 2 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 2 min at 37° C.;


5. 4 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 2500 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cells after growth in test wells with s compound;


NC: Background OD value of blank wells without s compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 5







Growth Inhibition Activity (IC50) of Some


Compounds in Examples on A549 Cells













IC50

IC50

IC50


Compounds
(μM)
Compounds
(μM)
Compounds
(μM)















Example 4
1.25
Example 5
1.3
Example 7
3


Example 8
1.7
Example 9
1.7
Example 10
2.3


Example 11
1.6
Example 12
2
Example 13
1.1


Example 14
1.5
Example 15
3.9
Example 16
3.4


Example 17
2.4
Example 19
2
Example 21
0.9


Example 153
6.4
Example 138
1.5
Example 139
0.6


Example 140
1.6
Example 141
0.8
Example 142
1.5


Example 143
1.5
Example 144
1
Example 145
6.7


Example 146
7
Example 150
3.8
Example 151
3.7


Example 174
3.7









Biological Example 4
LoVo Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

1. Cell strains: LoVo (human intestinal cancer cell strains);


2. MTT; antitumor compounds; DMSO.


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, and 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 2 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 4 min at 37° C.;


5. 4 ml of complete culture solution was added in the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 3000-4000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added into each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 4 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cells after growth in test wells with a compound;


NC: Background OD value of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 6







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on LoVo Cells

















IC50


Compounds
IC50 (μM)
Compounds
IC50 (μM)
Compounds
(μM)















Example 4
8.0
Example 5
8.57
Example 7
7.0


Example 8
1.7
Example 9
1.58
Example 10
2.0


Example 11
1.6
Example 12
2.0
Example 13
2.0


Example 14
1.3
Example 15
2.2
Example 16
1.8


Example 17
2.0
Example 19
2.0
Example 21
2.0


Example 138
2.1
Example 139
2.2
Example 140
8.8


Example 141
7.63
Example 142
8.83
Example 143
7.5


Example 144
3.3
Example 153
5.6
Example 149
2.3


Example 150
2.2
Example 151
1.9
Example 152
6.6









Biological Example 5
NCI-H460 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

1. Cell strains: NCI-H460 (human large cell lung cancer cell strains);


2. MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, and 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 2 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 2 min at 37° C.;


5. 4 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 2500 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cell after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 7







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on NCI-H460 Cells

















IC50


Compounds
IC50 (μM)
Compounds
IC50 (μM)
Compounds
(μM)















Example 4
1.6
Example 5
1.2
Example 7
0.94


Example 8
1.2
Example 9
1.7
Example 10
1.7


Example 11
1.5
Example 12
0.97
Example 13
1.8


Example 14
0.94
Example 15
1.4
Example 16
1.1


Example 17
1.5
Example 19
1.1
Example 21
2.7


Example 174
3.0
Example 46
4.0
Example 138
2.2


Example 140
1.1
Example 141
2.9
Example 142
1.04









Biological Example 6
NCI-N87 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

1. Cell strains: NCI-N87 (human gastric carcinoma cell strains);


MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, and 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 12 min at 37° C.;


5. 4.5 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture disc by 17000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD value of cells after normal growth in control wells without a compound;


n: OD value of cells after growth in test wellswith a compound;


NC: Background OD value of blank wellswithout a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 8







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on NCI-N87 Cells















IC50

IC50


Compounds
IC50 (μM)
Compounds
(μM)
Compounds
(μM)















Example 4
0.079
Example 5
0.022
Example 7
0.113


Example 8
0.0056
Example 9
0.0086
Example 10
0.011


Example 11
0.011
Example 12
0.0061
Example 13
0.0044


Example 153
0.015
Example 15
0.0071
Example 16
0.026


Example 17
0.021
Example 19
0.032
Example 154
0.041


Example 21
0.0068
Example 138
0.0044
Example 139
0.0072


Example 140
0.015
Example 141
0.032
Example 142
0.075


Example 143
0.089
Example 144
0.098
Example 145
0.346


Example 146
0.189
Example 149
0.111
Example 150
0.0066


Example 151
0.016
Example 152
0.020









Biological Example 7
Sk-Br-3 Cell Growth Inhibition Test (SRB Assay)
I. Assay Materials

1. Cell strains: Sk-Br-3 (human breast cancer cell strains);


2. SRB: available from Sigmaaldrich, goods number: 59012, lot number: 047K3751. SRB was reserved at the room temperature. 0.4% (w/v) of working solution was formulated with 1% of glacial acetic acid. The solution was reserved at 4° C.; anti-tumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% DMEM+10% FBS;


Pancreatin (0.25% (w/v) solution was formulatedwith PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


Tris;


Glacial acetic acid;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected. The culture solution was sucked out. The plate was washed with 5 mL of PBS 1-2 times;


2. PBS was sucked out. 1.5 m of 0.25% pancreatin was added to infiltrate the cells for 30 s;


3. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 1.5-2 min at 37° C.;


4. 4 mL of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform single cell suspension;


5. Cell suspension was counted. The suspension was diluted to 1×105/ml. The resulting suspension was developed uniformly on a plate by 10000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 91 h under 5% CO2 at 37° C.


6. The culture solution was sucked out. 100 uL of TCA fixed cells which were diluted to 10% were added to each well. The plate was kept in a refrigerator for 1 h at 4° C.


7. TCA stationary liquid was sucked out. Each well was washed with 150 μL of ddH2O five times;


8. After the stationary liquid was cleansed, the plate was dried in the air at the room temperature;


9. 60 μL of SRB staining solution was added in each well. The well was stained for 15 min at the room temperature;


10. The SRB staining solution was sucked out. Each well was washed with 150 μL of 1% glacial acetic acid five times;


11. After the SRB staining solution was cleansed, the plate was dried in the air at the room temperature;


12. 100 μL of 10 mM Tris was added in each well. The plate was vibrated to dissolve out SRB;


13. OD values were determined at 570 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio


To: OD values of the starting content of cells when the cells were developed on a plate;


PC: OD values of cells after normal growth in control wells without a compound;


Ti: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank well without a compound and cells


(1) If Ti>To, it shows that cells still grow after a compound was added. Therefore:





Ratio of cells in test wells to normal wells (% of control cell growth)=(Ti−To)/(PC−To)×100%


Ratio G150 was concentration of a compound at the point of 50%.


(2) If Ti<To, it shows that cells gradually die after a compound was added.


Therefore:





Ratio of killed cells to inoculated cells (% of killed cells)=(Ti−To)/(To−NC)×100%


LC50 was concentration point of a compound when half of the starting inoculated concentration was reached.


(3) If Ti=To, it shows that in the presence of a compound, both growth and death of cells tended to balance. Concentration of a compound at this point is defined as TGI (total growth inhibition).









TABLE 9







Growth Inhibition Activity (GI50) of Some Compounds


in Examples on Sk-Br-3 Cells















GI50

GI50


Compounds
GI50 (μM)
Compounds
(μM)
Compounds
(μM)















Example 4
0.148
Example 5
0.024
Example 7
0.213


Example 9
0.015
Example 10
0.006
Example 14
0.924


Example 16
0.036
Example 17
0.007
Example 19
0.063


Example 21
0.0041
Example 154
0.038
Example 138
0.0038


Example 139
0.0052
Example 140
0.022
Example 141
0.039


Example 142
0.117
Example 143
0.231
Example 144
0.202


Example 145
0.831
Example 146
0.823
Example 149
0.644


Example 150
0.011
Example 151
0.017
Example 152
0.019


Example 153
0.0068









Biological Example 8
SW620 Cell Growth Inhibition Test (SRB Assay)
I. Assay Materials

Cell strains: SW620 (human colorectal cancer cell strains);


SRB: available from Sigmaaldrich, goods number: 59012, lot number: 047K3751. RT was reserved at the room temperature. 0.4% (w/v) of working solution was formulated with 1% of glacial acetic acid. The solution was reserved at 4° C.; anti-tumor compounds; DMSO.


II. Reagents and Consumable Materials

Culture medium: 90% L15+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation); PBS;


Tris;


Glacial acetic acid;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected. The culture solution was sucked out. The plate was washed with 5 mL of PBS 1-2 times;


2. PBS was sucked out. 1.5 m of 0.25% pancreatin was added to infiltrate the cells for 30 s;


3. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 1.5-2 min at 37° C.;


4. 4 mL of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform single cell suspension;


5. Cell suspension was counted. The suspension was diluted to 1.5×105/ml. The resulting suspension was developed uniformly on a plate by 15000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 91 h under 5% CO2 at 37° C.;


6. The culture solution was sucked out. 100 uL of TCA fixed cells which were diluted to 10% were added to each well. The plate was kept in a refrigerator for 1 h at 4° C.


7. TCA stationary liquid was sucked out. Each well was washed with 150 μL of ddH2O five times;


8. After the stationary liquid was cleansed, the plate was dried in the air at the room temperature;


9. 60 μL of SRB staining solution was added in each well. The well was stained for 15 min at the room temperature;


10. The SRB staining solution was sucked out. Each well was washed with 150 of 1% glacial acetic acid five times;


11. After the SRB staining solution was cleansed, the plate was dried in the air at the room temperature;


12. 100 μL of 10 mM Tris was added in each well, the plate was vibrated to dissolve out SRB;


13. OD values were determined at 570 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio


To: OD values of the starting content of cells when the cells were developed on a plate;


PC: OD values of cells after normal growth in control wells without a compound;


Ti: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells


(1) If Ti>To, it shows that cells still grow after a compound was added. Therefore:





Ratio of cells in test wells to normal wells (% of control cell growth)=(Ti−To)/(PC−To)×100%


Ratio GI50 was concentration of a compound at the point of 50%.


(2) If Ti<To, it shows that cells gradually die after a compound was added.


Therefore:





Ratio of killed cells to inoculated cells (% of killed cell)=(Ti−To)/(To−NC)×100%


LC50 was concentration point of a compound when half of the starting inoculated concentration was reached.


(3) If Ti=To, it shows that in the presence of a compound, both growth and death of cells tended to balance. Concentration of a compound at this point is defined as TGI (total growth inhibition).









TABLE 10







Growth Inhibition Activity (GI50) of Some Compounds


in Examples on SW620 Cells

















GI50


Compounds
GI50 (μM)
Compounds
GI50 (μM)
Compounds
(μM)















Example 4
3.5
Example 5
1.68
Example 9
2.1


Example 21
1.78
Example 138
1.78
Example 139
1.9


Example 140
2.3
Example 141
1.4
Example 142
2.3









Biological Example 9
CCRF-CEM/T Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: CCRF-CEM/T (human acute lymphocytic cell leukemia cells); MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


PBS;


96-well cell culture plate;


triple lysate solution: 10% SDS, 5% isobutanol, 0.012M HCl.


III. Assay Process

1. Cells were cultured to exponential phase. The cells were suspension cells;


2. The cells were collected by centrifuge. The resulting cells were resuspended in a complete culture medium to a desired concentration. The cells suspension was implanted into a 96-well cell culture plate by 20000 cells per well. A compound was added in the well such that the final volume of the system was 120 μl. The system was further incubated for 72 h under 5% CO2 at 37° C.


7. 30 μl of serum-free culture solution containing 2.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. 150 μl of triple lysate solution was added in each well and placed to dissolve at the room temperature;


9. OD values were determined at 570 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 11







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on CCRF-CEM/T Cells

















IC50


Compounds
IC50 (μM)
Compounds
IC50 (μM)
Compounds
(μM)















Example 2
1.8
Example 4
5.8
Example 5
9


Example 158
1.1
Example 138
5.6
Example 139
1.4


Example 140
7.9
Example 141
9.1
Example 142
6.6


Example 150
3.76
Example 151
3.6









Biological Example 10
Fadu Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: Fudu (human head and neck cancer cell strains);


MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% EMEM+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 5 min at 37° C.;


5. 4.5 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 6000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 it of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve; 10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without s compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 12







Growth Inhibition Activity (IC50) of Somes Compounds


in Examples on Fadu Cells

















IC50


Compounds
IC50 (nM)
Compounds
IC50 (nM)
Compounds
(nM)















Example 2
149
Example 4
331
Example 143
127


Example 7
474
Example 8
188
Example 9
219


Example 11
187
Example 12
202
Example 13
300


Example 15
260
Example 138
101
Example 139
201


Example 144
103
Example 145
194
Example 146
200


Example 149
163
Example 150
207
Example 151
244


Example 152
170
Example 153
231
Example 154
324


Example 142
115
Example 158
119









Biological Example 11
BxPc-3 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: BxPC-3 (human pancreatic cancer cell strains);


MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. was added into The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 5 min at 37° C.;


5. 4.5 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 5000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 13







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on BxPc-3 Cells

















IC50


Compounds
IC50 (nM)
Compounds
IC50 (nM)
Compounds
(nM)





Example 131
306
Example 138
337
Example 139
188


Example 142
416









Biological Example 12
AsPC-1 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: AsPC-1 (human pancreatic cancer cell strains);


MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 7.5 min at 37° C.;


5. 4.5 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 8000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 14







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on AsPC-1 Cells

















IC50


Compounds
IC50 (μM)
Compounds
IC50 (μM)
Compounds
(μM)















Example 2
0.77
Example 4
1
Example 142
1.5


Example 150
0.895
Example 8
0.878
Example 9
0.458


Example 11
1.2
Example 12
1.2
Example 13
0.922


Example 14
1.4
Example 15
1.1
Example 16
0.81


Example 17
1
Example 19
0.621
Example 10
1


Example 138
1
Example 139
0.422


Example 151
0.81
Example 158
0.44









Biological Example 13
SK-OV-3 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: SK-OV-3 (human ovarian cancer cell strains);


MTT; antitumor compounds; DMSO.


II. Reagents and Consumable Materials

Culture medium: 90% McCoy's 5A+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 5 min at 37° C.;


5. 4.5 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture disc by 4000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 it of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 μl of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 15







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on SK-OV-3 Cells

















IC50


Compounds
IC50 (nM)
Compounds
IC50 (nM)
Compounds
(nM)





Example 2
331
Example 4
409
Example 5
195


Example 9
469
Example 138
457
Example 139
371


Example 140
286
Example 141
283
Example 142
553


Example 189
312









Biological Example 14
NCI-H358 Cell Growth Inhibition Test (MTT Assay)
I. Assay Materials

Cell strains: NCI-H358 (human non-small cell lung cancer cell strains);


MTT; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI-1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 ml of PBS 1-2 times;


3. PBS was sucked out. 1.5 ml of 0.25% pancreatin was added to infiltrate the cells for 1 min;


4. The culture plate was placed in an incubator. Digestion was carried out for about 5 min at 37° C.;


5. 3 ml of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform cell suspension. The suspension was implanted into a 96-well cell culture plate by 10000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.


6. The culture solution was sucked out;


7. 100 μl of serum-free culture solution containing 0.5 mg/ml MTT was added in each well, and incubated for 3 h;


8. The culture solution was carefully sucked out;


9. 100 id of DMSO was added in each well and vibrated to dissolve;


10. OD values were determined at 490 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 16







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on NCI-H358 Cells

















IC50


Compounds
IC50 (nM)
Compounds
IC50 (nM)
Compounds
(nM)





Example 138
54
Example 153
78
Example 188
110









Biological Example 15
NCI-H1650 Cell Growth Inhibition Test (SRB Assay)
I. Assay Materials

1. Cell strains: NCI-H1650 (human non-small cell lung cancer cell strains);


2. SRB, 0.4% (w/v) working solution was formulated with 1% glacial acetic acid and reserved at 4° C.; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% RPMI1640+10% FBS;


Pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


Tris;


Glacial acetic acid;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 mL of PBS 1-2 times;


3. 1.5 mL of 0.25% pancreatin was added to the plate to infiltrate the cells;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 3 min at 37° C.;


5. 4 mL of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform single cell suspension. The suspensin was implanted into a 96-well cell culture plate by 6000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μlof culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.;


6. The culture solution was sucked out 100 uL of TCA fixed cells which were diluted to 10% were added to each well. The plate was kept in a refrigerator for 1 h at 4° C.


7. TCA stationary liquid was sucked out. Each well was washed with 150 μL of ddH2O five times;


8. After the stationary liquid was cleansed, the plate was dried in the air at the room temperature;


9. 60 μL of SRB staining solution was added in each well. The well was stained for 15 min at the room temperature;


10. The SRB staining solution was sucked out. Each well was washed with 150 μL of 1% glacial acetic acid five times;


11. After the SRB staining solution was cleansed, the plate was dried in the air at the room temperature;


12. 100 μL of 10 mM Tris was added in each well, the plate was vibrated to dissolve out SRB;


13. OD values were determined at 570 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 17







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on NCI-H1650 Cells

















IC50


Compounds
IC50 (nM)
Compounds
IC50 (nM)
Compounds
(nM)















Example 9
636
Example 138
1100
Example 139
1130


Example 3
819
Example 158
760









Biological Example 16
MDA-MB-453 Cell Growth Inhibition Test (SRB Assay)
I. Assay Materials

Cell strains: MDA-MB-453 (human breast cancer cell strains);


SRB: 0.4% (w/v) working solution was formulated with 1% glacial acetic acid, reserved at 4° C.; antitumor compounds; DMSO


II. Reagents and Consumable Materials

Culture medium: 90% L15+10% FBS;


pancreatin (0.25% (w/v) solution was formulated with PBS, 0.53 mM of EDTA was added in the formulation);


PBS;


96-well culture plate


III. Assay Process

1. A plate (10 cm) of cells in logarithmic growth phase normally cultured was collected;


2. The culture solution was sucked out. The plate was washed with 5 mL of PBS 1-2 times;


3. 1.5 mL of 0.25% pancreatin was added to the plate to infiltrate the cells;


4. The pancreatin was sucked out. The culture plate was placed in an incubator. Digestion was carried out for about 3 min at 37° C.;


5. 4 mL of complete culture solution was added to the culture plate to stop the digestion. The cells were carefully scoured with micropipette (1 ml) to give a uniform single cell suspension. The suspension was implanted into a 96-well cell culture plate by 7000 cells/100 μl per well. The culture plate was incubated overnight under 5% CO2 at 37° C. In day 2, 100 μl of culture solution comprising a compound was added in each well, and further incubated for 72 h under 5% CO2 at 37° C.;


6. The culture solution was sucked out. 100 uL of TCA fixed cells which were diluted to 10% were added to each well. The plate was kept in a refrigerator for 1 h at 4° C.


7. TCA stationary liquid was sucked out. Each well was washed with 150 μL of ddH2O five times;


8. After the stationary liquid was cleansed, the plate was dried in the air at the room temperature;


9. 60 μL of SRB staining solution was added in each well. The well was stained for 15 min at the room temperature;


10. The SRB staining solution was sucked out. Each well was washed with 150 μL of 1% glacial acetic acid five times;


11. After the SRB staining solution was cleansed, the plate was dried in the air at the room temperature;


12. 100 μL of 10 mM Tris was added in each well. The plate was vibrated to dissolve out SRB;


13. OD values were determined at 570 nM.


IV. Results and Treatments

1. Calculation of Relative Inhibition Ratio





The inhibition ratio of a compound on cell growth=(PC−n)/(PC−NC)×100%


wherein:


PC: OD values of cells after normal growth in control wells without a compound;


n: OD values of cells after growth in test wells with a compound;


NC: Background OD values of blank wells without a compound and cells;


IC50: Concentration of a compound where inhibition ratio was 50%. IC50 values were fitted with Origin7.5.









TABLE 18







Growth Inhibition Activity (IC50) of Some Compounds


in Examples on MDA-MB-453 Cells












Compounds
IC50 (nM)
Compounds
IC50 (nM)







Example 5
505
Example 189
523










Biological Example 17
Inhibition Test on EGFR-TK
I. Operation Procedures

1. 2 mg/ml of PGT was diluted with PBS to 0.2 mg/ml. The resulting solution was added into elisa plate by 50 μl per well. The plate was placed in a refrigerator at 4° C. and coated overnight.


2. The coating solution was discarded to stop coating. The plate was washed three times with PBS, and then beaten to dryness.


3. The elisa plate was placed in a refrigerator at 4° C., drained out and stood for 2 h.


4. The mother liquid of aqueous solution of each compound was diluted with double distilled water to four folds, i.e. a desirable concentration. Then, the compound was diluted and added in the corresponding elisa well by 25 μl/well.


5. 4 nM of ATP solution was diluted with double distilled water to 50 folds and then sufficiently mixed. The resulting solution was separately added in each elisa well by 25 μl per well (except for the negative control).


6. EGFR kinase was diluted with 2×TKB to 800 folds. The solution was added in elisa well with 50 μl per well. Then, the enzyme reaction began. The elisa plate was immediately placed on a micro-oscillator and shaked to react for 20 min at the room temperature.


7. 2% of SDS was added in the elisa well to stop the reaction by adding 100 μl of SDS into each well. The solution was sufficiently mixed on the micro-oscillator to sufficiently mix for about 5 min.


8. The reaction solution was sucked out. The plate was washed four times with PBST. After the plate was beaten to dryness, 0.25 μg/ml of mice anti-phosphorylation tyrosine-HRP antibody which was diluted with blocking solution (3% solution of BSA in PBST) was added in the plate by 100 μl per well. The mixture was reacted for 30 min at the room temperature.


9. The antibody reaction solution was sucked out. The plate was washed four times with PBST. After the plate was beated to dryness, TMB peroxidase substrate was added to the plate by 100 μl/well. The mixture reacted in the dark for 15 min at the room temperature.


10. 100 μl of 2N H2SO4 was added in the plate to stop the chromogenic reaction. After the bubbles were cleansed, OD values were determined in ELIASA at 450 nm.


11. Formula for calculating inhibition percentage:







Inhibition





%

=

100
-



Reading





of





Infrared






Reader


(
Drug
)




Reading





of





Infrared






Reader


(
Blank
)




×
100






EC50 values of some compounds were given in Table 1.









TABLE 19







Inhibition Activity (EC50) of Some Compounds in


Examples on EGFR-TK phosphorylation

















EC50


Compounds
EC50 (nM)
Compounds
EC50 (nM)
Compounds
(nM)















Example 4
42
Example 5
44
Example 9
135


Example 46
25
Example 138
110
Example 21
154









Biological Example 18
Inhibition Activity Assay on Her2 Enzyme
I. Assay Materials

1. Her2 kinase, Cell Signaling Tech, #7382, Lot. 2, reserved at −80° C.;


2. Elisa plate, Nunc Maxisorp, 442404;


3. Test compounds and control compounds were prepared by applicants;


II. Assay Reagemts

1. PBS: 8 g/L NaCl, 0.2 g/L KCl, 2.9 g/L Na2HPO4.12H2O, 0.2 g/L KH2PO4;


2. PBST: PBS+0.05% (v/v) Tween 20;


3. Confining liquid: 3% of BSA in PBS;


4. HEPEs buffer (2×): 50 mM HEPEs, 20 mM MgCl2, 0.1 mM MnCl2, 0.2 mM Na3VO4, adjusted to pH of 7.4 with NaOH;


5. PGT, Sigma, Cat#P0275): 2 mg/ml of stock solution was prepared by dissolving PBS, reserved at −20° C.


6. ATP: 4 nM of stock solution was prepared by dissolving ddH2O, reserved at −20° C.;


7. Mice anti-phosphorylation tyrosine-HRP: Invitrogen-037720, 0.5 mg/ml, reserved at 4° C.;


8. TMB: Cell Signaling Tech, Cat #7004L;


9. DTT: 2.5 M of stock solution was prepared with ddH2O, reserved at −20° C. Before use, 2×HEPEs buffer was added in the solution to the final concentration of 1.25 mM.


III. Assay Equipment

1. ELIASA, Bio-Rad, Model-680;


2. Plate washer, Bio-Rad, Model-1575;


3. 4° C. refrigerator, Frestech, Model-BCD-213KC;


4. Micro oscillator, Shanghai Yarong Biochemisty Instrument Plant, Model MM-I;


5. −80° C. refrigerator, Haier, Model DW-86L386.


IV. Assay Process

1. 2 mg/ml of PGT was diluted with PBS to 0.2 mg/ml. 60 μl of the resulting solution was added in each well. The elisa plate was placed in a refrigerator at 4° C. and coated overnight;


2. The coating solution was discarded to stop the coating. The plate was washed four times with PBS, and then beaten to dryness;


3. The elisa plate was placed in a refrigerator at 4° C., drained out and stood for 3 h.


4. 2 nM of aqueous solution of each compound was diluted with ddH2O to a desirable concentration. Then, the solution was added in the corresponding elisa well by 25 μl/well;


5. 2 nM of ATP solution was diluted with ddH2O to 25 folds and then sufficiently mixed. The resulting solution was separately added in each elisa well by 25 μl per well (25 μl ddH2O was added in the negative control);


6. Her2 kinase was diluted with 2×TKB to 800 folds. The resulting solution was added in the elisa well by 50 μl per well. Then, the enzyme reaction began. Moreover, the elisa plate was immediately placed in a micro-oscillator. The reaction is carried out for 8 min at the temperature of 4° C.;


7. 2% of SDS was added in the elisa well to stop the reaction by adding 100 μl of SDS in each well. The solution was sufficiently mixed on the micro-oscillator to sufficiently mix for about 5 min;


8. The reaction solution was sucked out. The plate was washed four times with PBST. After the plate was beaten to dryness, 0.25 μg/ml of mice anti-phosphorylation tyrosine-HRP which was diluted with confining liquid was added in the plate by 100 μl per well, and then reacted for 30 min at the room temperature.


9. The antibody reaction solution was sucked out. The plate was washed six times with PBST. After the plate was beaten to dryness, TMB peroxidase substrate was added in the plate with 100 μl/well, and then reacted in the dark for 15 min at the room temperature;


10. 100 μl of 2N H2SO4 was added in the solution to stop the chromogenic reaction. After the bubbles were cleansed, the solution was measured in elisa at 450 nm.


V. Data Processing Method

OD values were converted in relative inhibition ratios according to following formula:





Relative inhibition ratio=[1−(experiment values−NC mean values)/(PC mean values−NC mean values)]λ100%


wherein:


PC: Group of cells that grow normally in control wells without a compound;


NC: Blank group without a compound and cells.


The inhibition activity of some compounds on Her-2 were measured as given in Table 2.









TABLE 20







Inhibition Activity (EC50) of Some Compounds in Examples on Her-2

















EC50


Compounds
EC50 (nM)
Compounds
EC50 (nM)
Compounds
(nM)















Example 4
161
Example 5
96
Example 9
221


Example 21
234
Example 42
291
Example 138
244


Example 140
110









In view of the above, the compounds of the invention have excellent inhibition activity on receptor tyrosine kinase, especially erbB family, more specially EGFR and Her 2.


All the patents, patent application publications, patent applications and non-patent publications cited in the present description are incorporated herein by reference in their entirety.


From the forging, it will be appreciated, although specific embodiments of the present invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the present invention. Therefore, the scope of the present invention is only defined by the pending claims.

Claims
  • 1.-35. (canceled)
  • 36. A method for treating and/or preventing tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, a stereoisomer thereof, a cis-trans-isomer thereof, a tautomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • 37. The method of claim 36, wherein the tumor is selected from the group consisting of epithelial adenocarcinoma, breast cancer, lung cancer, intestinal cancer, gastric carcinoma, colorectal cancer, acute lymphocytic cell leukemia, head and neck cancer, pancreatic cancer and ovarian cancer.
  • 38. The method of claim 37, wherein the lung cancer is large cell lung cancer or non-small cell lung cancer.
  • 39. The method of claim 36, wherein R4 is tetrahydrofuranyl.
  • 40. The method of claim 36, wherein:R1 is selected from the group consisting of C2-C6 alkenylacylamino and C2-C6 alkynylacylamino; andone of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, C6-C18 aryl, and C5-C18 heteroaryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, C6-C18 aryloxy and C5-C18 heteroaryloxy, and wherein the heteroaryl is optionally substituted with a substituent selected from the group consisting of C7-C24 aralkyl, C6-C18 arylacylamino, C6-C18 arylsulfonylamino, C5-C18 heteroarylacylamino, C3-C10 cycloalkylacylamino, C6-C18 arylaminoacyl, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy and C6-C18 aryloxy.
  • 41. The method of claim 36, wherein:R1 is C2-C6 alkenylacylamino; andone of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, and C6-C18 aryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C18 aryloxy, C5-C18 heteroaryloxy, and C6-C24 heteroaralkyloxy.
  • 42. The method of claim 36, wherein:R1 is C2-C6 alkenylacylamino; andone of R2 and R3 is H, while the other one is selected from the group consisting of C7-C24 aralkyl, and C6-C18 aryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C24 heteroaralkyloxy.
  • 43. The method of claim 36, wherein:R1 is selected from the group consisting of 4-(dimethylamino)-but-2-enamido, 4-(diethylamino)-but-2-enamido, 4-(piperidin-1-yl)-but-2-enamido, 4-(morpholin-4-yl)-but-2-enamido, 4-(tert-butylamino)-but-2-enamido, 4-(benzylamino)-but-2-enamido, 4-(N-methylbenzylamino)-but-2-enamido, 4-(6-hydroxyhexylamino)-but-2-enamido, 4-(2-methoxylethylamino)-but-2-enamido, 2-(piperidin-4-ylidene)acetamido, 2-(1-methylpiperidin-4-ylidene)acetamido, 2-(1-ethylpiperidin-4-ylidene)acetamide, 2-(1-(2-methoxyehtyl)piperdin-4-ylidene)acetamido, 4-(diethanolamino)-but-2-enamido, 4-(N-methylmethoxylethylamino)-but-2-enamido, 4-(N-methylethanolamino)-but-2-enamido, 4-(dimethoxylethylamino)-but-2-enamido, 4-(N-methyl-6-amino-1-hexanolyl)-but-2-enamido, acrylamido, but-2-enamido, 3-methyl-but-2-enamido, and 2-(pyrrolidin-3-ylidene)acetamido.
  • 44. The method of claim 36, wherein:R1 is C2-C6 alkenylacylamino; andone of R2 and R3 is H, while the other one is C6-C18 aryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of halogen, C6-C24 heteroaralkyloxy, C2-C6 alkynyl, and C7-C24 aralkyloxy.
  • 45. The method of claim 36, wherein:R1 is C2-C6 alkenylacylamino; andR2 and R3 together with nitrogen atom to which they are attached form substituted or unsubstituted C3-C18 heterocyclyl.
  • 46. The method of claim 44, wherein:R1 is 2-(piperidin-4-ylidene)acetamide.
  • 47. A method for treating and/or preventing tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof,
  • 48. The method of claim 47, wherein the tumor is selected from the group consisting of epithelial adenocarcinoma, breast cancer, lung cancer, intestinal cancer, gastric carcinoma, colorectal cancer, acute lymphocytic cell leukemia, head and neck cancer, pancreatic cancer and ovarian cancer.
  • 49. The method of claim 48, wherein the lung cancer is large cell lung cancer or non-small cell lung cancer.
  • 50. The method of claim 47, wherein: R1 is 2-(piperidin-4-ylidene)acetamide; andone of R2 and R3 is H, while the other one is selected from the group consisting of C6-C18 aryl, and C5-C18 heteroaryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C18 aryloxy, and wherein the heteroaryl is optionally substituted with C7-C24 aralkyloxy.
  • 51. The method of claim 47, wherein: R1 is 2-(piperidin-4-ylidene)acetamide; andone of R2 and R3 is H, while the other one is selected from the group consisting of C6-C18 aryl, and C5-C18 heteroaryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, and C6-C18 aryloxy, and wherein the heteroaryl is optionally substituted with C7-C24 aralkyloxy.
  • 52. A method for treating and/or preventing tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof,
  • 53. The method of claim 52, wherein the tumor is selected from the group consisting of epithelial adenocarcinoma, breast cancer, lung cancer, intestinal cancer, gastric carcinoma, colorectal cancer, acute lymphocytic cell leukemia, head and neck cancer, pancreatic cancer and ovarian cancer.
  • 54. The method of claim 53, wherein the lung cancer is large cell lung cancer or non-small cell lung cancer.
  • 55. The method of claim 52, wherein: R1 is 2-(piperidin-4-ylidene)acetamido; andone of R2 and R3 is H, while the other one is selected from the group consisting of C6-C18 aryl, and C5-C18 heteroaryl, wherein the aryl is optionally substituted with a substituent selected from the group consisting of C2-C6 alkynyl, halogen, C7-C24 aralkyloxy, C6-C24 heteroaralkyloxy, and C6-C18 aryloxy, and wherein the heteroaryl is optionally substituted with a substituent selected from the group consisting of C7-C24 aralkyloxy, and C6-C18 aryloxy.
  • 56. The compound of claim 52, wherein: R1 is 2-(pyrrolidin-3-ylidene)acetamido; andone of R2 and R3 is H, while the other one is C5-C18 heteroaryl substituted with C6-C18 aryloxy.
  • 57. The compound of claim 52, wherein: R1 is 4-(diethylamino)-but-2-eneamido; andone of R2 and R3 is H, while the other one is C6-C18 aryl substituted with halogen.
  • 58. A method for treating and/or preventing tumor in a mammal, comprising administering to the mammal in need thereof a therapeutically effective amount of a compound or a pharmaceutically acceptable salt thereof, selected from the group consisting of: (E)-4-bromo-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)but-2-enamide;(E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide hydrochloride;(E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(benzyloxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(3-bromophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(2-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(3-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(benzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(2-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(4-tert-butylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(3-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(4-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(2-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(3-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(4-(4-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-3-cyano-4-((R)-1-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)acrylamide;(E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)but-2-enamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-3-methylbut-2-enamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)propiolamide;tert-butyl 4-(2-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl-amino)-2-oxoethylidene)piperidine-1-carboxylate;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-methylpiperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-ethylpiperidin-4-ylidene)acetamide;2-(1-benzylpiperidin-4-ylidene)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy) phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)acetamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-(2-methoxyethyl)piperidin-4-ylidene)acetamide;methyl 2-(4-(2-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl-amino)-2-oxoethylidene)piperidin-1-yl)acetate;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-isopropylpiperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-(2-hydroxyethyl)piperidin-4-ylidene) acetamide;(E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(pyrrolidin-3-ylidene)acetamide;N1-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-N4-(2-(2-(dimethylamino)ethoxy)ethyl) fumaramide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-(2-(2-(2-hydroxyethoxy)ethylamino)acetyl) piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(1-(methylsulfonyl)piperidin-4-ylidene) acetamide;N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-bromophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-methoxybenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-methoxybenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene) acetamide;N-(4-(4-(4-methoxybenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-tert-butylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(benzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-((S)-1-phenylethylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-((R)-1-phenylethylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(1-benzyl-1H-indol-5-yl-amino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(1-(3-cyanobenzyl)-1H-indol-5-yl-amino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(1-(3-methoxybenzyl)-1H-indol-5-yl-amino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(1-(3-chlorobenzyl)-1H-indol-5-yl-amino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(indolin-1-yl)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-chloroindolin-1-yl)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(6-fluoroindolin-1-yl)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-chloroindolin-1-yl)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(3,4-dihydroquinolin-1(2H)-yl)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(6-methyl-3,4-dihydroquinolin-1 (2H)-yl)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene) acetamide;N-(3-cyano-7-(tetrahydrofuran-3-yl-oxy)-4-(7-(trifluoromethyl)-3,4-dihydroquinolin-1 (2H)-yl)-quinolin-6-yl)-2-(piperidin-4-ylidene) acetamide;N-(4-(6-(benzyloxy)indolin-1-yl)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;methyl 1-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl)-indoline-2-carboxylate;N-(3-cyano-4-(2-(hydroxymethyl)indolin-1-yl)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-(1H-pyrrol-1-yl)indolin-1-yl)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(octahydroindol-1-yl)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(pyrimidin-2-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(2-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-5-yl)benzamide;N-(2-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-5-yl)-4-(dimethylamino)benzamide;N-(3-cyano-4-(5-(phenylsulfonamido)pyrimidin-2-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(5-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-2-yl)benzamide;N-(5-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-2-yl)furan-2-carboxamide;N-(5-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-2-yl)thiophene-2-carboxamide;N-(5-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)pyrimidin-2-yl)cyclohexylcarboxamide;5-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)-N-(4-methoxyphenyl)pyrimidin-2-carboxamide;N-(3-cyano-4-(pyridin-2-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;6-(3-cyano-6-(2-(piperidin-4-ylidene)acetamido)-7-(tetrahydrofuran-3-yl-oxy)quinolin-4-yl-amino)-N-(4-methoxyphenyl)nicotinamide;N-(3-cyano-4-(pyridin-3-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(pyridin-4-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-(benzyloxy)pyridin-3-yl-amino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(pyrazin-2-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(3-ethynylphenylamino)-7-(1-methylpiperidin-4-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(benzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-bromobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-methoxybenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-ethylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-ethoxybenzyloxy)-3-chlorophenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-phenoxyphenylamino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-7-(1-methylpiperidin-4-yl-oxy)-4-(pyridin-2-yl-amino)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-7-(1-methylpiperidin-4-yl-oxy)-4-(pyridin-3-yl-amino)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-7-(1-methylpiperidin-4-yl-oxy)-4-(pyridin-4-yl-amino)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-(benzyloxy)pyridin-3-yl-amino)-3-cyano-7-(1-methylpiperidin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(3-ethynylphenylamino)-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(4-phenoxyphenylamino)-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(benzyloxy)phenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(4-methoxybenzyloxy)phenylamino)-3-cyano-7-(pyridin-4-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(4-(3-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(pyridine-2-yl-amino)-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-7-(pyridin-4-yl-oxy)-4-(pyrimidin-2-yl-amino)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-(benzyloxy)pyridin-3-yl-amino)-3-cyano-7-(pyridin-4-yl-oxy) quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(6-(3-chlorobenzyloxy)pyridin-3-yl-amino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(3-cyano-4-(6-(phenoxypyridin-3-yl-amino)-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;N-(4-(3-chloro-4-(pyridine-2-yl-methoxy)phenylamino)-3-cyano-7-(pyridin-4-yl-oxy)quinolin-6-yl)-2-(piperidin-4-ylidene)acetamide;(E/Z)-N-(3-cyano-4-(6-(phenoxypyridin-3-yl-amino)-7-(pyridin-4-yl-oxy) quinolin-6-yl)-2-(pyrrolidin-3-ylidene)acetamide;(E/Z)-N-(3-cyano-7-(1-methylpiperidin-4-yl-oxy)-4-(6-(phenoxypyridin-3-yl-amino)quinolin-6-yl)-2-(pyrrolidin-3-ylidene)acetamide;(E/Z)-N-(3-cyano-4-(6-(phenoxypyridin-3-yl-amino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-2-(pyrrolidin-3-ylidene)acetamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(R,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(R,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(tert-butylaimino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(benzylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(6-hydroxyhexylaimino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(N-methylbenzylamino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(tert-butylamino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(6-hydroxyhexylamino)-but-2-enamide;(S,E)-N-(4-(4-(benzyloxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(3-bromophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(2-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(2-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(S,E)-N-(4-(4-(3-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(S,E)-N-(4-(4-(benzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(2-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(4-tert-butylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(3-cyanobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(4-chlorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(S,E)-N-(4-(4-(2-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(4-(4-methylbenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(S,E)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(E)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(pyridin-3-yl-oxy) quinolin-6-yl)4-(dimethylamino)-but-2-enamide;(E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(E/Z)-N-(4-(3-chloro-4-fluorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E/Z)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(E/Z)-N-(4-(4-(benzyloxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylaimino)-but-2-enamide;(E/Z)-N-(4-(3-bromophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(E/Z)-N-(4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethylamino)-but-2-enamide;(S,E/Z)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E/Z)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E/Z)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(S,E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethylamino)-but-2-enamide;(S,E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(piperidin-1-yl)-but-2-enamide;(S,E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(morpholin-4-yl)-but-2-enamide;(S,E/Z)-N-(4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-3-cyano-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(2-methoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(diethanolamino)-but-2-enamide(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(N-methylmethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(N-methylethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(dimethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-ethynylphenylamino)-7-(tetrahydrofuran-3-yl-oxy) quinolin-6-yl)-4-(N-methyl-6-amino-1-hexanolyl)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-fluoro-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-fluoro-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylmethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-fluoro-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-fluoro-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-fluoro-phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methyl-6-amino-1-hexanolyl)-but-2-enamide;(S,E)-N-(3-cyano-4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylmethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(4-(3-fluorobenzyloxy)-3-chlorophenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methyl-6-amino-1-hexanolyl)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(diethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylmethoxyethylamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methylethanolamino)-but-2-enamide;(S,E)-N-(3-cyano-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(dimethoxyethylamino)-but-2-enamide; and(S,E)-N-(3-cyano-4-(3-chloro-4-(pyridin-2-yl-methoxy)phenylamino)-7-(tetrahydrofuran-3-yl-oxy)quinolin-6-yl)-4-(N-methyl-6-amino-1-hexanol)-but-2-enamide.
  • 59. The method of claim 58, wherein the tumor is selected from the group consisting of epithelial adenocarcinoma, breast cancer, lung cancer, intestinal cancer, gastric carcinoma, colorectal cancer, acute lymphocytic cell leukemia, head and neck cancer, pancreatic cancer and ovarian cancer.
  • 60. The method of claim 59, wherein the lung cancer is large cell lung cancer or non-small cell lung cancer.
Divisions (1)
Number Date Country
Parent 13702987 May 2013 US
Child 14881085 US