QUINAZOLINE DERIVATIVE AND USE THEREOF

Abstract
The present invention relates to a series of quinazoline compounds, especially compounds as represented by formula (I), isomers thereof or pharmaceutically acceptable salts thereof, pharmaceutical compositions thereof, and use thereof as Pan-HER tyrosine kinase inhibitors.
Description
TECHNICAL FIELD

The present disclosure relates to a series of quinazoline compounds, especially compounds of Formula (I), isomers thereof or pharmaceutically acceptable salts thereof, the pharmaceutical composition containing the same and their use as Pan-HER tyrosine kinase inhibitors.


BACKGROUND OF THE INVENTION

Human epidermal growth factor receptor (HER, EGFR) is a member of the protein tyrosine kinase family. It is widely distributed on the cell membrane of various human tissues, and can regulate the proliferation, growth, metastasis and apoptosis of cells. Its structure is composed of three parts: extracellular ligand binding region, transmembrane region and intracellular tyrosine kinase region. According to the structural differences of receptors, HER can be divided into four isoforms, i.e., HER1 (EGFR, ErbB-1), HER2 (ErbB-2), HER3 (ErbB-3) and HER4 (ErbB-4). Research disclosed that there is over expression or abnormal activation of HER in various tumor cells such as breast cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, ovarian cancer, colorectal cancer, head and neck squamous cell carcinoma, malignant glioma and prostate cancer. In addition, research shows that the over expression or abnormal activation of HER is closely associated with the degree of tumor differentiation, the degree of malignancy and prognosis (Baselga. J., Oncologist 2002, 7, 2-8). Therefore, the inhibition of HER has become a hot topic in the research of anti-tumor drugs.


At present, targeted HER inhibitors on the market include Gefitinib, Erlotinib and Lapatinib, etc. However, these marketed drugs have a low effective response rate, are susceptiple to drug resistance, and have some toxic and side effects. Therefore, there is an urgent need to develop other anti-tumor drugs that have excellent anti-tumor effects, can overcome drug resistance, and are well tolerated.


Irreversible inhibitors of Pan-HER tyrosine kinase simultaneously inhibit HER1, HER2 and HER4. Research reveals that this irreversible inhibition of HER family receptors can not only enhance the activity of the drug, but also reduce the occurrence of drug resistance. At the same time, it has a significant inhibitory effect on some tumor cell lines with drug resistance, such as the H1975 cell line that is resistant to Erlotinib. At present, the marketed irreversible inhibitors of Pan-HER tyrosine kinase are only Afatinib and Neratinib. Many inhibitors are in clinical research, such as Poziotinib, Dacomitinib and Canertinib, and there are still unmet market demands.


Therefore, it is necessary to further develop irreversible inhibitors of Pan-HER tyrosine kinase for the treatment of cancer.


Poziotinib (control compound 1) is Pan-HER inhibitor developed by WO2008150118, with the following structure:




embedded image


The patent application WO2015043515 discloses control compound 2, which has inhibitory effects on EGFR and HER2, and has the following structure:




embedded image


SUMMARY OF THE INVENTION

The present disclosure provides compounds of Formula (I), isomers thereof or pharmaceutically acceptable salts thereof,




embedded image


wherein,


T is selected from the group consisting of N and CR′;


each R1 is independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, C1-3 alkyl and C1-3 alkoxy, wherein the C1-3 alkyl or C1-3 alkoxy is optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and CN;


R2 and R3 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2 and C1-3 alkyl, wherein the C1-3 alkyl is optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br and I;


R4 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, 3- to 7-membered heterocycloalkyl-O—, 3- to 7-membered heterocycloalkyl-C1-6 alkyl-O—, C3-6 cycloalkyl-O— and C3-6 cycloalkyl-C1-6 alkyl-O—, wherein the C1-6 alkyl, C1-6 alkoxy, 3- to 7-membered heterocycloalkyl-O—, 3- to 7-membered heterocycloalkyl-C1-6 alkyl-O—, C3-6 cycloalkyl-O— and C3-6 cycloalkyl-C1-6 alkyl-O— are each independently optionally substituted with 1, 2, or 3 R;


R5 and R6 are each independently selected from the group consisting of H, C1-3 alkyl and 3- to 7-membered heterocycloalkyl, wherein the C1-3 alkyl and 3- to 7-membered heterocycloalkyl are optionally substituted with 1, 2, or 3 R;


L is selected from the group consisting of —O—, —NRa—, —CRb1Rb2—, —CRb1Rb2—O—, and —CRb1Rb2—NH—;


Ra is H;


alternatively, R4 is connected to Ra to form a 5- to 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3 or 4 R′;


ring A is selected from the group consisting of phenyl and 5- to 10-membered heteroaryl;


ring B is selected from the group consisting of




embedded image


n is selected from the group consisting of 1, 2, 3, 4 and 5;


each R is independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN, NRc1Rc2, C1-3 alkyl, C1-3 alkoxy, C1-3 alkyl-NH—C(═O)—, C1-3 alkylthio and 3- to 7-membered heterocycloalkyl, wherein the C1-3 alkyl, C1-3 alkoxy, C1-3 alkyl-NH—C(═O)—, C1-3 alkylthio and 3- to 7-membered heterocycloalkyl are optionally substituted with 1, 2, or 3 R′;


R′ is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN and C1-3 alkyl;


Rb1 and Rb2 are each independently selected from the group consisting of H, F, Cl, Br, I and C1-3 alkyl;


Rc1 and Rc2 are each independently selected from the group consisting of H and C1-3 alkyl;


the 5- to 7-membered heterocycloalkyl, 5- to 10-membered heteroaryl and 3- to 7-membered heterocycloalkyl groups each contain 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from the group consisting of N, —O—, —S— and —NH—.


In some embodiments disclosed herein, the above R is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN,




embedded image


Me, Et,



embedded image


piperidinyl, pyrrolidinyl, azetidinyl, morpholinyl and oxetanyl, wherein the Me, Et,




embedded image


piperidinyl, pyrrolidinyl, azetidinyl, morpholinyl and oxetanyl are optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and C1-3 alkyl, and other variables are as defined herein.


In some embodiments disclosed herein, the above R is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN,




embedded image


Me, Et,



embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R1 is each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, Me and




embedded image


wherein the Me and




embedded image


are optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and CN, and other variables are as defined herein.


In some embodiments disclosed herein, the above R1 is each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, Me, CH2F, CHF2, CF3,




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R2 and R3 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, Me and CF3, and other variables are as defined herein.


In some embodiments disclosed herein, the above R4 is selected from the group consisting of




embedded image


oxetanyl-O—, piperidinyl-C1-3 alkyl-O—, morpholinyl-C1-3 alkyl-O—, azetidinyl-O—, tetrahydrofuranyl-O—, cyclopropyl-O—, pyrrolidinyl-C1-3 alkyl-O— and azetidinyl-C1-3 alkyl-O—, wherein the




embedded image


oxetanyl-O—, piperidinyl-C1-3 alkyl-O—, morpholinyl-C1-3 alkyl-O—, azetidinyl-O—, tetrahydrofuranyl-O—, cyclopropyl-O—, pyrrolidinyl-C1-3 alkyl-O— and azetidinyl-C1-3 alkyl-O— are optionally substituted with 1, 2 or 3 R, and other variables are as defined herein.


In some embodiments disclosed herein, the above R4 is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R5 and R6 are each independently selected from the group consisting of H, Me, Et, and




embedded image


wherein the Me, Et and




embedded image


are optionally substituted with 1, 2 or 3 R, and other variables are as defined herein.


In some embodiments disclosed herein, the above R5 and R6 are each independently selected from the group consisting of H,




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above T is selected from the group consisting of N and C (CN), and other variables are as defined herein.


In some embodiments disclosed herein, the above L is selected from the group consisting of —O—, —NH—, —CH2—, —CH2—O— and —CH2—NH—, and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above ring A is selected from the group consisting of phenyl, azaindenyl, benzo[b]thienyl, imidazo[1,2-a]pyridyl and benzo[d]isoxazolyl, and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above ring B is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


The present disclosure also provides compounds of Formula (I), isomers thereof or pharmaceutically acceptable salts thereof,




embedded image


wherein,


T is selected from the group consisting of N and CR′;


R1 is each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, C1-3 alkyl and C1-3 alkoxy, wherein the C1-3 alkyl or C1-3 alkoxy is optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and CN;


R2 and R3 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2 and C1-3 alkyl, wherein the C1-3 alkyl is optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, and I;


R4 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, 3- to 7-membered heterocycloalkyl-O—, 3- to 7-membered heterocycloalkyl-C1-6 alkyl-O—, C3-6 cycloalkyl-O— and C3-6 cycloalkyl-C1-6 alkyl-O—, wherein the C1-6 alkyl, C1-6 alkoxy, 3- to 7-membered heterocycloalkyl-O—, 3- to 7-membered heterocycloalkyl-C1-6 alkyl-O—, C3-6 cycloalkyl-O— and C3-6 cycloalkyl-C1-6 alkyl-O— are each independently optionally substituted with 1, 2, or 3 R;


R5 and R6 are each independently selected from the group consisting of H, C1-3 alkyl and 3- to 7-membered heterocycloalkyl, wherein the C1-3 alkyl and 3- to 7-membered heterocycloalkyl are optionally substituted with 1, 2, or 3 R;


L is selected from the group consisting of —O—, —NRa—, —CRb1Rb2—, —CRb1Rb2—O—, and —CRb1Rb2—NH—;


Ra is H;


alternatively, R4 is connected to Ra to form a 5- to 7-membered heterocycloalkyl optionally substituted with 1, 2, 3 or 4 R′;


ring A is selected from the group consisting of phenyl and 5- to 10-membered heteroaryl;


ring B is selected from the group consisting of




embedded image


n is selected from the group consisting of 1, 2, 3, 4 and 5;


each R is independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN, NRc1R2, C1-3 alkyl, C1-3 alkoxy, C1-3 alkylthio and 3- to 7-membered heterocycloalkyl, wherein the C1-3 alkyl, C1-3 alkoxy, C1-3 alkylthio and 3- to 7-membered heterocycloalkyl are optionally substituted with 1, 2, or 3 R′;


R′ is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN and C1-3 alkyl;


Rb1 and Rb2 are each independently selected from the group consisting of H, F, Cl, Br, I and C1-3 alkyl;


Rc1 and Rc2 are each independently selected from the group consisting of H and C1-3 alkyl;


the 5- to 7-membered heterocycloalkyl, 5- to 10-membered heteroaryl, and 3- to 7-membered heterocycloalkyl each contains 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from the group consisting of N, —O—, —S— and —NH—.


In some embodiments disclosed herein, the above R is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN,




embedded image


Me, Et,



embedded image


piperidinyl, pyrrolidinyl, azetidinyl, morpholinyl and oxetanyl, wherein the Me, Et,




embedded image


piperidinyl, pyrrolidinyl, azetidinyl, morpholinyl and oxetanyl are optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and C1-3 alkyl, and other variables are as defined herein.


In some embodiments disclosed herein, the above R is each independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN,




embedded image


Me,



embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R1 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, Me and




embedded image


wherein the Me and




embedded image


are optionally substituted with 1, 2 or 3 substituents independently selected from the group consisting of F, Cl, Br, I, OH, NH2 and CN, and other variables are as defined herein.


In some embodiments disclosed herein, the above R1 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, Me,




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R2 and R3 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, Me and CF3, and other variables are as defined herein.


In some embodiments disclosed herein, the above R4 is selected from the group consisting of




embedded image


oxetanyl-O—, morpholinyl-C1-3 alkyl-O—, azetidinyl-O—, tetrahydrofuranyl-O—, cyclopropyl-O—, pyrrolidinyl-C1-3 alkyl-O— and azetidinyl-C1-3 alkyl-O—, wherein the




embedded image


oxetanyl-O—, morpholinyl-C1-3 alkyl-O—, azetidinyl-O—, tetrahydrofuranyl-O—, cyclopropyl-O—, pyrrolidinyl-C1-3 alkyl-O— and azetidinyl-C1-3 alkyl-O— are optionally substituted with 1, 2, or 3 R, and other variables are as defined herein.


In some embodiments disclosed herein, the above R4 is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above R5 and R6 are each independently selected from the group consisting of H, Me, Et and




embedded image


wherein the Me, Et and




embedded image


are optionally substituted with 1, 2, or 3 R, and other variables are as defined herein.


In some embodiments disclosed herein, the above R5 and R6 are each independently selected from the group consisting of H,




embedded image


and N and other variables are as defined herein.


In some embodiments disclosed herein, the above T is selected from the group consisting of N and C(CN), and other variables are as defined herein.


In some embodiments disclosed herein, the above L is selected from the group consisting of —O—, —NH—, —CH2—CH2—O— and —CH2—NH—, and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is


and other variables are as defined herein.


In some embodiments disclosed herein, the above ring A is selected from the group consisting of phenyl, azaindenyl, benzo[b]thienyl and benzo[d]isoxazolyl, and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above ring Bis selected from the group consisting of




embedded image


and other variables are as defined herein.


In some embodiments disclosed herein, the above moiety




embedded image


is selected from the group consisting of




embedded image


and other variables are as defined herein.


The present disclosure also includes some embodiments that are obtained by combination of any above definitions of the above variables.


In some embodiments disclosed herein, the above compounds, isomers thereof or pharmaceutically acceptable salts thereof are selected from the group consisting of:




embedded image


embedded image


embedded image


wherein T, L, R1, R2, R3, R4, R5, and R6 are as defined herein.


In some embodiments disclosed herein, the above compounds, isomers thereof or pharmaceutically acceptable salts thereof are selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


wherein T, L, R1, R2, R3, R4, R5 and R6 are as defined herein.


The present disclosure also provides the following compounds, isomers thereof or pharmaceutically acceptable salts thereof:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In some embodiments disclosed herein, the above compounds are selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


The present disclosure also provides a pharmaceutical composition, comprising a therapeutically effective amount of the above compound or a pharmaceutically acceptable salt thereof as an active ingredient, and pharmaceutically acceptable carrier(s).


The present disclosure also provides use of the above compound or a pharmaceutically acceptable salt thereof or the above composition in the manufacture of a medicament associated with a Pan-HER tyrosine kinase inhibitor.


Definitions and Terms

Unless otherwise specified, the following terms and phrases used herein are intended to have the following meanings. A specific term or phrase should not be considered indefinite or unclear in the absence of a particular definition, but should be understood in the conventional sense. When a trade name appears herein, it is intended to refer to its corresponding commodity or active ingredient thereof. The term “pharmaceutically acceptable” is used herein in terms of those compounds, materials, compositions, and/or dosage forms, which are suitable for use in contact with human and animal tissues within the scope of reliable medical judgment, with no excessive toxicity, irritation, allergic reaction or other problems or complications, commensurate with a reasonable benefit/risk ratio.


The term “pharmaceutically acceptable salt” refers to a salt of the compound disclosed herein that is prepared by reacting the compound having a specific substituent disclosed herein with a relatively non-toxic acid or base. When the compound disclosed herein contains a relatively acidic functional group, a base addition salt can be obtained by bringing the neutral form of the compound into contact with a sufficient amount of base in a pure solution or a suitable inert solvent. The pharmaceutically acceptable base addition salt includes a salt of sodium, potassium, calcium, ammonium, organic amine or magnesium or similar salts. When the compound disclosed herein contains a relatively basic functional group, an acid addition salt can be obtained by bringing the neutral form of the compound into contact with a sufficient amount of acid in a pure solution or a suitable inert solvent.


Examples of the pharmaceutically acceptable acid addition salt include an inorganic acid salt, wherein the inorganic acid includes, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and an organic acid salt, wherein the organic acid includes, for example, acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and methanesulfonic acid, and the like; and an salt of amino acid (such as arginine and the like), and a salt of an organic acid such as glucuronic acid and the like. Certain specific compounds disclosed herein contain both basic and acidic functional groups and can be converted to any base or acid addition salt.


The pharmaceutically acceptable salt disclosed herein can be prepared from the parent compound that contains an acidic or basic moiety by conventional chemical methods. Generally, such salt can be prepared by reacting the free acid or base form of the compound with a stoichiometric amount of an appropriate base or acid in water or an organic solvent or a mixture thereof.


The compound disclosed herein may be present in a specific geometric or stereoisomeric form. The present disclosure contemplates all such compounds, including cis and trans isomer, (−)- and (+)-enantiomer, (R)- and (S)-enantiomer, diastereoisomer, (D)-isomer, (L)-isomer, and racemic mixture and other mixtures, for example, an enantiomer or diastereoisomer enriched mixture, all of which are encompassed within the scope disclosed herein. The substituent such as alkyl may have an additional asymmetric carbon atom. All these isomers and mixtures thereof are encompassed within the scope disclosed herein.


Unless otherwise specified, the term “enantiomer” or “optical isomer” refers to stereoisomers that are in a mirrored relationship with each other.


Unless otherwise specified, the term “cis-trans isomer” or “geometric isomer” is produced by the inability of a double bond or a single bond between ring-forming carbon atoms to rotate freely.


Unless otherwise specified, the term “diastereomer” refers to a stereoisomer in which two or more chiral centers of are contained in a molecule and is in a non-mirrored relationship between molecules.


Unless otherwise specified, “(D)” or “(+)” means dextroisomer, “(L)” or “(−)” means levoisomer, and “(DL)” or “(±)” means racemate.


Unless otherwise specified, a wedged solid bond (custom-character) and a wedged dashed bond (custom-character) indicate the absolute configuration of a stereocenter; a straight solid bond (custom-character) and a straight dashed bond (custom-character) indicate the relative configuration, of a stereocenter; a wavy line (custom-character) indicates a wedged solid bond (custom-character) or a wedged dashed bond (custom-character); or a wavy line (custom-character) indicates a straight solid bond (custom-character) and a straight dashed bond (custom-character).


The compounds disclosed herein may be present in a particular form. Unless otherwise specified, the terms “tautomer” or “tautomeric form” means that different functional groups are in dynamic equilibrium at room temperature and can be rapidly converted into each other. If tautomers are possible (as in solution), a chemical equilibrium of tautomers can be achieved. For example, proton tautomers (also known as prototropic tautomers) include interconversions by proton transfer, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers include interconversions by recombination of some bonding electrons. A specific example of keto-enol tautomerization is interconversion between two tautomers pentane-2,4-dione and 4-hydroxypent-3-en-2-one.


Unless otherwise specified, the term “enriched in one isomer”, “isomer enriched”, “enriched in one enantiomer” or “enantiomeric enriched” means that the content of one isomer or enantiomer is less than 100%, and the content of the isomer or enantiomer is 60% or more, or 70% or more, or 80% or more, or 90% or more, or 95% or more, or 96% or more, or 97% or more, 98% or more, 99% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, or 99.9% or more.


Unless otherwise specified, the term “isomer excess” or “enantiomeric excess” refers to the difference between the relative percentages of two isomers or two enantiomers. For example, if one isomer or enantiomer is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomeric excess (ee value) is 80%.


Optically active (R)- and (S)-isomer, or D and L isomer can be prepared using chiral synthesis or chiral reagents or other conventional techniques. If one kind of enantiomer of certain compound disclosed herein is to be obtained, the pure desired enantiomer can be obtained by asymmetric synthesis or derivative action of chiral auxiliary followed by separating the resulting diastereomeric mixture and cleaving the auxiliary group.


Alternatively, when the molecule contains a basic functional group (such as amino) or an acidic functional group (such as carboxyl), the compound reacts with an appropriate optically active acid or base to form a salt of the diastereomeric isomer which is then subjected to diastereomeric resolution through the conventional method in the art to give the pure enantiomer. In addition, the enantiomer and the diastereoisomer are generally isolated through chromatography which uses a chiral stationary phase and optionally combines with a chemical derivative method (for example, carbamate generated from amine).


The compounds disclosed herein may contain an unnatural proportion of atomic isotopes at one or more of the atoms that make up the compounds. For example, a compound may be labeled with a radioisotope such as tritium (3H), iodine-125 (125I) or C-14(14C). For another example, hydrogen can be replaced by heavy hydrogen to form a deuterated drug.


The bond between deuterium and carbon is stronger than that between ordinary hydrogen and carbon. Compared with undeuterated drugs, deuterated drugs have advantages of reduced toxic side effects, increased drug stability, enhanced efficacy, and prolonged biological half-life of drugs. All changes in the isotopic composition of compounds disclosed herein, regardless of radioactivity, are included within the scope of the present disclosure.


The term “pharmaceutically acceptable carrier” refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance disclosed herein, which does not interfere with the biological activity of an active substance, and has no toxic side effects to the host or patient. Representative carriers include water, oil, vegetables, minerals, cream bases, lotion bases, ointment bases and the like. These bases include suspending agents, tackifiers, transdermal enhancers, etc. Their formulations are well known to those skilled in cosmetic or topical pharmaceutical arts.


“Optional” or “optionally” means that the subsequent event or condition may occur but not requisite, that the term includes the instance in which the event or condition occurs and the instance in which the event or condition does not occur.


The term “substituted” means one or more than one hydrogen atom(s) on a specific atom are substituted by a substituent, including deuterium and hydrogen variants, as long as the valence of the specific atom is normal and the substituted compound is stable. When the substituent is oxo (i.e. ═O), it means two hydrogen atoms are substituted. Positions on an aromatic ring cannot be substituted by oxo. The term “optionally substituted” means an atom can be substituted by a substituent or not, unless otherwise specified, the species and number of the substituent may be arbitrary as long as being chemically achievable.


When any variable (such as R) occurs in the constitution or structure of the compound more than once, the definition of the variable at each occurrence is independent. Thus, for example, if a group is substituted by 0-2 R, the group can be optionally substituted by up to two R, wherein the definition of R at each occurrence is independent. Moreover, a combination of the substituent and/or the variant thereof is allowed only when the combination results in a stable compound.


When the number of a linking group is 0, such as —(CRR)0—, it means that the linking group is a single bond.


When one of the variable is a single bond, it means that the two groups linked by the single bond are connected directly. For example, when L in A-L-Z represents a single bond, the structure of A-L-Z is actually A-Z.


When a substituent is vacant, it means that the substituent does not exist. For example, when X is vacant in A-X, the structure of A-X is actually A. When an enumerative substituent does not indicate through which atom it is linked to the substituted group, such substituent can be bonded through any of its atoms. For example, a pyridyl group as a substituent may be linked to the substituted group through any one of carbon atoms on the pyridine ring. When an enumerative linking group does not indicate its linking direction, its linking direction is arbitrary. For example, when the linking group L in




embedded image


is -M-W—, the -M-W— can be linked to the ring A and the ring B in the same direction as the reading order from left to right to constitute




embedded image


or can be linked to the ring A and the ring B in the reverse direction as the reading order from left to right to constitute




embedded image


A combination of the linking groups, substituents and/or variants thereof is allowed only when such combination can result in a stable compound.


Unless otherwise specified, the term “hetero” represents a heteroatom or a heteroatom group (e.g., an atom group containing a heteroatom), including the atom except carbon (C) and hydrogen (H) and the atom group containing the above heteroatom, for example, including oxygen (O), nitrogen (N), sulfur (S), silicon (Si), germanium (Ge), aluminum (Al), boron (B), —O—, —S—, ═O, ═S, —C(═O)O—, —C(═O)—, —C(═S)—, —S(═O), —S(═O)2—, and the group consisting of —C(═O)N(H)—, —N(H)—, —C(═NH)—, —S(═O)2N(H)— and —S(═O)N(H)—, each of which is optionally substituted.


Unless otherwise specified, the term “ring” refers to a substituted or unsubstituted cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, aryl or heteroaryl. The so-called ring includes a single ring, a ring assembly, a spiral ring, a fused ring or a bridged ring. The number of the atom on the ring is usually defined as the member number of the ring, for example, a “5-7 membered ring” means that 5 to 7 atoms are arranged on a ring. Unless otherwise specified, the ring optionally contains 1 to 3 heteroatoms. Therefore, a “5-7 membered ring” includes, for example, phenyl, pyridyl and piperidyl; on the other hand, the term “5-7 membered heterocycloalkyl ring” includes pyridyl and piperidyl, but excluding phenyl. The term “ring” also includes a ring system containing at least one ring, wherein each ring independently meets the above definition.


Unless otherwise specified, the term “heterocycle” or “heterocyclyl” refers to a stable monocyclic, bicyclic or tricyclic ring containing a heteroatom or a heteroatom group, which can be saturated, partially unsaturated or unsaturated (aromatic) and can contain carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from the group consisting of N, O and S, wherein any of the above heterocycle can be fused to a benzene ring to form a bicyclic ring. Nitrogen and sulfur heteroatoms can optionally be oxidized (i.e., NO and S(O)p, p is 1 or 2). Nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents already defined herein). The heterocycle can be attached to the pendant group of any heteroatom or carbon atom to form a stable structure. If the resulting compound is stable, the heterocycle described herein may have a substitution at a carbon or nitrogen position. Nitrogen atom on the heterocycle is optionally quaternized. In a preferred embodiment, when the total number of S and O atom of the heterocycle is more than 1, the heteroatom is not adjacent to each other. In another preferred embodiment, the total number of S and O atom of the heterocycle is not more than 1. As used herein, the term “aromatic heterocyclic group” or “heteroaryl” refers to a stable 5-, 6- or 7-membered monocyclic or bicyclic or 7-, 8-, 9- or 10-membered bicyclic heterocyclic aromatic ring which contains carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from the group consisting of N, O and S. Nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents already defined herein). Nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., NO and S(O)p, p is 1 or 2). It is worth noting that the total number of S and O atom of an aromatic heterocycle is not more than one. The bridged ring is also included in the definition of the heterocycle. A bridged ring is formed when one or more than one atom (i.e, C, O, N or S) link two non-adjacent carbon or nitrogen atoms. A preferred bridged ring includes, but not limited to one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms and one carbon-nitrogen group. It is worth noting that a bridge always converts a monocyclic ring to a tricyclic ring. In a bridged ring, the substituent on the ring may also be present on the bridge.


Examples of the heterocyclic compound include, but are not limited to: azetidinyl, azocinyl, benzimidazolyl, benzofuryl, benzomercaptofuryl, benzomercaptophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzoisoxazolyl, benzoisothiazolyl, benzoimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromene, cinnolinyl decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuryl, furyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isobenzofuryl, isoindolyl, isoindolinyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydro-isoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, hydroxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazine, phenothiazine, benzoxanthinyl, phenoloxazinyl, phthalazinyl, piperazinyl, piperidyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyrido-oxazolyl, pyrido-imidazolyl, pyrido-thiazolyl, pyridyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, isothiazolylthienyl, thieno-oxazolyl, thieno-thiazolyl, thieno-imidazolyl, thienyl, triazinyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, 4H-1,2,4-triazolyl and xanthenyl. Fused-ring compounds and spiro compounds are also included.


Unless otherwise specified, the term “hydrocarbyl” or its hyponyms (e.g. alkyl, alkenyl, alkynyl, and aryl, etc.), by itself or as part of another substituent, refers to a linear, branched chain or cyclic hydrocarbon radical or any combination thereof. They can be fully saturated (e.g. alkyl), mono- or polyunsaturated (e.g. alkenyl, alkynyl, and aryl), can be mono-, di- or poly-substituted, can be monovalent (e.g. methyl), divalent (e.g. methylene) or multivalent (e.g. methenyl), can also include a divalent or multivalent group, have a specified number of carbon atom (for example, C1-C12 indicates 1 to 12 carbon atoms, C1-12 is selected from the group consisting of C1, C2, C3, C4, C5, C6, C7, C8, C9, C10, C11 and C12; C3-12 is selected from the group consisting of C3, C4, C5, C6, C7, C8, C9, C10, C11 and C12). The term “hydrocarbyl” includes, but is not limited to aliphatic hydrocarbyl and aromatic hydrocarbyl. The aliphatic hydrocarbyl includes linear and cyclic hydrocarbyl, specifically includes but not limited to alkyl, alkenyl, and alkynyl. The aromatic hydrocarbyl includes but is not limited to 6-12 membered aromatic hydrocarbyl such as phenyl, naphthyl and the like. In some embodiments, the term “hydrocarbyl” refers to a linear or branched group or a combination thereof which can be fully saturated, mono- or polyunsaturated, and can include a divalent or multivalent group. Examples of the saturated hydrocarbyl group include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, sec-butyl, isobutyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, and the homolog or isomer of n-amyl, n-hexyl, n-heptyl, n-octyl and other atom groups. The unsaturated hydrocarbyl has one or more than one double or triple bonds. Examples of the unsaturated alkyl include but are not limited to, vinyl, 2-propenyl, butenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and more higher homologs and isomers.


Unless otherwise specified, the term “heterohydrocarbyl” or the specific term thereof (such as heteroalkyl, heteroalkenyl, heteroalkynyl, and heteroaryl, etc.), by itself or as part of another substituent, refers to a stable linear, branched or cyclic hydrocarbon radical or any combination thereof, which has a specified number of carbon atoms and at least one heteroatom. In some embodiments, the term “heteroalkyl” by itself or in combination with another term refers to a stable, linear or branched hydrocarbon radical or a combination thereof, which has a specified number of carbon atoms and at least one heteroatom. In a typical embodiment, a heteroatom is selected from the group consisting of B, O, N and S, wherein nitrogen and sulfur atoms are optionally oxidized and the nitrogen atom is optionally quaternized. The heteroatom or heteroatom group can be located at any interior position of a heterohydrocarbyl, including the position where the hydrocarbyl attaches to the rest part of the molecule. But the terms “alkoxy”, “alkylamino” and “alkylthio” are used by the conventional meaning and refer to an alkyl group connected to the rest part of the molecule via an oxygen atom, an amino or a sulfur atom respectively. Examples include, but are not limited to, —CH2—CH2—O—CH3, —CH2—CH2—NH—CH3, —CH2—CH2—N(CH3)—CH3, —CH2—S—CH2—CH3, —CH2—CH2, —S(O)—CH3, —CH2—CH2—S(O)2—CH3, —CH═CHO—CH3, —CH2—CH═N—OCH3 and —CH═CH—N(CH3)—CH3. Up to two consecutive heteroatoms can be present, such as, —CH2—NH—OCH3.


Unless otherwise specified, the term “cyclohydrocarbyl”, “heterocyclohydrocarbyl” or hyponyms thereof (such as aryl, heteroaryl, cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, etc.) by itself or in combination with another term refers to cyclized “hydrocarbyl” or “heterohydrocarbyl”. Furthermore, for heterohydrocarbyl or heterocyclohydrocarbyl (e.g. heteroalkyl, and heterocycloalkyl), one heteroatom can occupy the position where the heterocycle attaches to the remainder position of the molecule. Examples of the cyclohydrocarbyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl and the like. Non-limiting examples of heterocyclohydrocarbyl include 1-(1,2,5,6-tetrahydropyridyl), 1-piperidyl, 2-piperidyl, 3-piperidyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl and 2-piperazinyl.


Unless otherwise specified, the term “heterocycloalkyl” by itself or in combination with other terms means cyclized “heteroalkyl”. In addition, as far as the “heterocycloalkyl” is concerned, the heteroatom may occupy the connection position of the heterocycloalkyl to the rest of the molecule. In some embodiments, the heterocycloalkyl is a 4- to 6-membered heterocycloalkyl; in other embodiments, the heterocycloalkyl is a 5- to 6-membered heterocycloalkyl. Examples of heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothienyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, dioxanyl, dithianyl, isoxazolidinyl, isothiazolidinyl, 1,2-oxazinyl, 1,2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl or oxepanyl.


Unless otherwise specified, the term “alkyl” refers to a linear chain or branched saturated hydrocarbon group, can be mono-substituted (e.g. —CH2F) or poly-substituted (e.g. —CF3), can be monovalent (e.g. methyl), divalent (e.g. methylene) or multivalent (e.g. methenyl). Examples of alkyl include methyl (Me), ethyl (Et), propyl (such as n-propyl and isopropyl), butyl (such as n-butyl, isobutyl, s-butyl, t-butyl), pentyl (such as n-pentyl, isopentyl, neopentyl) and the like.


Unless otherwise specified, cycloalkyl includes any stable cyclic or polycyclic hydrocarbyl, and any carbon atom thereof is saturated. Cycloalkyl can be mono-substituted or poly-substituted, and can be monovalent, divalent or multivalent. Examples of cycloalkyl include, but are not limited to, cyclopropyl, norbornanyl, [2.2.2]bicyclooctane, [4.4.0]bicyclodecanyl and the like.


Unless otherwise specified, the term “halo” or “halogen” by itself or as part of another substituent refers to fluorine, chlorine, bromine or iodine atom. Furthermore, the term “haloalkyl” is meant to include monohaloalkyl and polyhaloalkyl. For example, the term “halo(C1-C4)alkyl” is meant to include, but not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl and the like. Examples of haloalkyl include, but not limited to trifluoromethyl, trichloromethyl, pentafluoroethyl and pentachloroethyl.


The term “alkoxy” represents any alkyl defined above having a specified number of carbon atoms attached by an oxygen bridge. Unless otherwise specified, C1-6 alkoxy includes C1, C2, C3, C4, C5 and C6 alkoxy. Examples of alkoxy include, but not limited to methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy and s-pentoxy.


Unless otherwise specified, the term “aryl” refers to a polyunsaturated aromatic substituent, can be mono-, di- or poly-substituted, can be a monovalent, divalent or multivalent, can be a single ring or a multiple ring (e.g. one to three rings; wherein at least one ring is aromatic), which are fused together or connected covalently. The term “heteroaryl” refers to an aryl (or ring) containing one to four heteroatoms. In an illustrative example, the heteroatom is selected from the group consisting of B, O, N and S, wherein nitrogen and sulfur atoms are optionally oxidized and nitrogen atom is optionally quaternized. A heteroaryl may attach to the rest part of a molecule via a heteroatom. Non-limiting examples of aryl or heteroaryl include phenyl, naphthyl, biphenyl, pyrrolyl, pyrazolyl, imidazolyl, pyrazinyl, oxazolyl, phenyl-oxazolyl, isoxazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrimidinyl, benzothiazolyl, purinyl, benzimidazolyl, indolyl, isoquinolinyl, quinoxalinyl, quinolinyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolinyl, 5-isoquinolinyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolinyl and 6-quinolinyl. The substituent of any of the above aryl and heteroaryl ring system is selected from the group consisting of the acceptable substituents described below.


Unless otherwise specified, when combined with other terms (such as aryloxy, arylthio, arylalkyl), the aryl includes the aryl and heteroaryl ring as defined above. Thus, the term “aralkyl” is meant to include the group (e.g. benzyl, phenethyl, pyridylmethyl, etc.) where an aryl is attached to an alkyl, including an alkyl where the carbon atom (e.g. methylene) has been replaced by an atom such as oxygen, for example, phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like.


The term “leaving group” refers to a functional group or atom which can be replaced by another functional group or atom through a substitution reaction (such as affinity substitution reaction). For example, representative leaving groups include triflate; chlorine, bromine and iodine; sulfonate group, such as mesylate, tosylate, p-bromobenzenesulfonate, p-toluenesulfonate and the like; acyloxy, such as acetoxy, trifluoroacetoxy and the like.


The term “protecting group” includes, but is not limited to “amino protecting group”, “hydroxy protecting group” or “thio protecting group”. The term “amino protecting group” refers to a protecting group suitable for blocking the side reaction on the nitrogen of an amino. Representative amino protecting groups include, but are not limited to: formyl; acyl, such as alkanoyl (e.g. acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl such as benzyl (Bn), trityl (Tr), 1,1-bis-(4′-methoxyphenyl)methyl; silyl such as trimethylsilyl (TMS) and tert-butyldimethylsilyl (TBS) and the like. The term “hydroxy protecting group” refers to a protecting group suitable for blocking the side reaction on hydroxy. Representative hydroxy protecting groups include, but are not limited to: alkyl such as methyl, ethyl and tert-butyl; acyl such as alkanoyl (e.g. acetyl); arylmethyl such as benzyl (Bn), p-methoxybenzyl (PMB), 9-fluorenylmethyl (Fm), and diphenylmethyl (benzhydryl, DPM); silyl such as trimethylsilyl (TMS) and tert-butyl dimethyl silyl (TBS) and the like.


The compound disclosed herein can be prepared by a variety of synthetic methods well known to the skilled in the art, including the following enumerative embodiment, the embodiment formed by the following enumerative embodiment in combination with other chemical synthesis methods and the equivalent replacement well known to the skilled in the art. The preferred embodiment includes, but is not limited to the embodiment disclosed herein.


The compounds disclosed herein may have various uses or indications, including but not limited to the specific uses or indications listed herein.


All of the solvents used in the present disclosure are commercially available. The present disclosure employs the following abbreviations: aq represents water; HATU represents O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate; EDC represents N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride; m-CPBA represents 3-chloroperoxybenzoic acid; eq represents equivalent or equivalence; CDI represents carbonyl diimidazole; DCM represents dichloromethane; PE represents petroleum ether; DIAD represents diisopropyl azodicarboxylate; DMF represents N,N-dimethylformamide; DMSO represents dimethyl sulfoxide; EtOAc represents ethyl acetate; EtOH represents ethanol; MeOH represents methanol; CBz represents benzyloxycarbonyl, which is an amino protecting group; BOC represents tert-butyloxycarbonyl, which is an amino protecting group; HOAc represents acetic acid; NaCNBH3 represents sodium cyanoborohydride; r.t. represents room temperature; O/N represents overnight; THF represents tetrahydrofuran; Boc2O represents di-tert-butyldicarbonate; TFA represents trifluoroacetic acid; DIPEA represents diisopropylethylamine; SOCl2 represents thionyl chloride; CS2 represents carbon disulfide; TsOH represents p-toluenesulfonic acid; NFSI represents N-fluoro-N-(phenylsulfonyl)benzenesulfonamide; NCS represents 1-chloropyrrolidine-2,5-dione; n-Bu4NF represents tetrabutylammonium fluoride; iPrOH represents 2-propanol; mp represents melting point; LDA represents lithium diisopropylamide; DEA represents diethylamine; ACN represents acetonitrile; HATU represents 2-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate; DIEA represents N,N-diisopropylethylamine; PBS represents phosphate buffer solution; and Matrigel represents matrix gelatin.


Compounds are named according to conventional naming principles in the art or using ChemDraw® software, and names in supplier catalog are used for commercially available compounds.


Technical effect: The compounds disclosed herein have obvious inhibitory activity against HER1, HER2 and HER4, and have obvious inhibitory activity against the proliferation of NCI-N87 cells and BT-474 cells. The results from the in vivo pharmacodynamics study in BALB/c nude mouse model with subcutaneous xenograft tumor of human gastric cancer NCI-N87 cells showed that the compounds disclosed herein have a significant effect of inhibiting tumor growth without significantly affecting the weight of mice at the effective dose, and thus they are safe.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1: relative weight change (%) of BALB/c nude mouse model with subcutaneous xenograft tumor of human esophageal cancer OE21 cells, wherein the relative weight change is calculated based on the animal's body weight at the start of dosing. Data points represent the percentage in average body weight change within the group, and error lines represent standard error (SEM).





PO: oral administration; QD: once a day; D1-D7: day 1 to day 7 of administration; D8-D21: day 8 to day 21 of administration; D1-D2: day 1 to day 2 of administration; D3-D21: day 3 to day 21 of administration; n=6:6 mice per group.


DETAILED DESCRIPTION OF THE EMBODIMENTS

The present disclosure is described in detail below by means of examples. However, it is not intended that these examples have any disadvantageous limitations to the present disclosure. The present disclosure has been described in detail herein, and the embodiments are also disclosed herein. It will be apparent to those skilled in the art that various changes and modifications may be made to the embodiments disclosed herein without departing from the spirit and scope disclosed herein.


Reference Example 1



embedded image


Synthetic Route:




embedded image


Compound A-1 (500 mg, 4.42 mmol) and triethylamine (1.12 g, 11.05 mmol) were dissolved in dichloromethane (20 mL), and to the mixture was added di-tert-butyl dicarbonate (1.01 g, 4.64 mmol). The reaction solution was stirred at 10° C. for 16 hours. The reaction solution was washed twice with saturated ammonium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was slurried with petroleum ether (3 mL) to afford compound A-2. 1H NMR (400 MHz, CDCl3) δ 4.05-4.15 (m, 1H), 3.50-3.62 (m, 4H), 2.40-2.60 (m, 2H), 2.04 (d, J=7.2 Hz, 1H), 1.52-1.60 (m, 1H), 1.47 (s, 9H), 1.30-1.40 (m, 1H).


Example 1



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Sodium metal (1.84 g, 80.15 mmol) was added to anhydrous methanol (100 mL) and stirred under nitrogen at 30° C. for 30 minutes. The reaction solution was cooled to 0° C., and compound 1-1 (10 g, 53.44 mmol) was added. The reaction solution was stirred at 70° C. for 16 hours, and then cooled to 0° C. To the mixture was added 1 N dilute hydrochloric acid (100 mL), diluted with water (200 mL), and extracted with dichloromethane (200 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:8) to afford compound 1-2. 1H NMR (400 MHz, CDCl3) δ 7.53 (d, J=12.4 Hz, 1H), 6.15 (d, J=6.8 Hz, 1H), 5.69 (brs, 2H), 3.87 (s, 3H), 3.84 (s, 3H). LC-MS: m/z=199.9 [M+H]+.


The Second Step

A solution of compound 1-2 (5.00 g, 25.10 mmol) in dichloroethane (100 mL) was slowly added to a solution of metachloroperbenzoic acid (85%, 20.39 g, 100.41 mmol) in dichloroethane (200 mL) dropwise at 90° C. The reaction solution was stirred at 90° C. for two hours, and then cooled to 0° C., and quenched with saturated aqueous sodium sulfite solution (300 mL). The mixture was extracted with dichloromethane (300 mL×2). The organic phases were combined, washed sequentially with saturated sodium bicarbonate solution (300 mL×2), water (300 mL×2) and saturated brine (300 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:8) to afford compound 1-3. 1H NMR (400 MHz, CDCl3) δ 7.47 (d, J=10.4 Hz, 1H), 7.43 (d, J=7.2 Hz, 1H), 3.98 (s, 3H), 3.89 (s, 3H).


The Third Step

Compound 1-3 (730.0 mg, 3.19 mmol) was dissolved in N,N-dimethylformamide (10 mL). To the mixture were added cesium carbonate (2.08 g, 6.37 mmol) and compound A-2 (679.0 mg, 3.19 mmol). The reaction solution was heated to 65° C., and stirred for 3 hours. The reaction solution was cooled to 20° C., diluted with water (30.0 mL) and extracted with ethyl acetate (30 mL×2). The organic phases were combined, washed sequentially with water (40 mL×2) and saturated brine (40 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (petroleum ether:ethyl acetate=8:1) to afford compound 1-4. 1H NMR (400 MHz, CDCl3) δ 7.44 (s, 1H), 7.03 (s, 1H), 4.54 (t, J=5.6 Hz, 1H), 3.92 (s, 3H), 3.89 (s, 3H), 3.89 (td, J1=12.0 Hz, J2=3.2 Hz, 2H), 3.51 (t, J=11.2 Hz, 2H), 2.80-2.95 (m, 2H), 1.75-1.85 (m, 1H), 1.55-1.65 (m, 1H), 1.47 (s, 9H). LC-MS: m/z=445.2 [M+Na]+.


The Fourth Step

Compound 1-4 (740.0 mg, 1.75 mmol) was dissolved in methanol (100 mL), and to the mixture was added wet palladium/carbon (10%, 150 mg). The reaction solution was stirred at 20° C. under hydrogen atmosphere (hydrogen balloon) for 0.5 hour. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure to afford compound 1-5. 1H NMR (400 MHz, CDCl3) δ 7.29 (s, 1H), 6.12 (s, 1H), 4.30-4.40 (m, 1H), 3.83 (s, 3H), 3.81 (s, 3H), 3.55-3.65 (m, 4H), 2.70-2.85 (m, 2H), 1.65-1.75 (m, 1H), 1.50-1.55 (m, 1H), 1.47 (s, 9H).


The Fifth Step

Compound 1-5 (687.0 mg, 1.75 mmol) and ammonium acetate (675.0 mg, 8.75 mmol) were added to trimethyl orthoformate (17.09 g, 161.02 mmol). The reaction solution was heated to 70° C. and stirred for 10 hours. The reaction solution was cooled to 30° C., and concentrated under reduced pressure. The residue was slurried with water (10 mL) and filtered. The filter cake was dried to afford compound 1-6. 1H NMR (400 MHz, DMSO-d6) δ 12.08 (brs, 1H), 7.99 (s, 1H), 7.47 (s, 1H), 7.15 (s, 1H), 4.65 (t, J=5.6 Hz, 1H), 3.89 (s, 3H), 3.45-3.60 (m, 2H), 3.10-3.20 (m, 2H), 2.75-2.90 (m, 2H), 1.75-1.85 (m, 1H), 1.40-1.50 (m, 1H), 1.40 (s, 9H). LC-MS: m/z=410.1 [M+Na]+.


The Sixth Step

Compound 1-6 (640.0 mg, 1.65 mmol) was added to a solution of hydrogen chloride in dioxane (4 N, 10 mL). The reaction solution was stirred at 0° C. for 0.4 hour, and then concentrated under reduced pressure to obtain the hydrochloride salt of compound 1-7. 1H NMR (400 MHz, DMSO-d6) δ 9.72 (brs, 1H), 9.02 (brs, 1H), 8.34 (s, 1H), 7.56 (s, 1H), 7.27 (s, 1H), 4.76 (t, J=6.0 Hz, 1H), 3.96 (s, 3H), 3.35-3.45 (m, 2H), 3.20-3.35 (m, 2H), 2.89-2.95 (m, 2H), 2.01-2.08 (m, 1H), 1.90-2.00 (m, 1H).


The Seventh Step

The hydrochloride salt of 1-7 (640.0 mg, 1.98 mmol) and triethylamine (2.00 g, 19.77 mmol) were dissolved in dichloromethane (10 mL) at 0° C., and trifluoroacetic anhydride (830.0 mg, 3.95 mmol) was added to the mixture dropwise. The reaction solution was stirred at 20° C. for 2 hours. The reaction solution was quenched by adding saturated aqueous sodium bicarbonate solution (10 mL), and extracted with dichloromethane (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound 1-8. 1H NMR (400 MHz, DMSO-d6) δ 12.10 (s, 1H), 8.00 (s, 1H), 7.52 (s, 1H), 7.15 (s, 1H), 4.84 (t, J=5.6 Hz, 1H), 3.90-4.00 (m, 1H), 3.87 (s, 3H), 3.65-3.85 (m, 2H), 3.45-3.55 (m, 1H), 2.90-3.00 (m, 2H), 1.90-2.00 (m, 1H), 1.50-1.60 (m, 1H).


The Eighth Step

Compound 1-8 (270.0 mg, 704.37 μmol) was dissolved in thionyl chloride (13.1 mL, 180.28 mmol), and N,N-dimethylformamide (26.0 μL, 338.10 μmol) was added to the mixture. The reaction solution was heated to 80° C. and stirred for 1 hour. The reaction solution was concentrated under reduced pressure. To the residue was added a solution of 3,4-dichloro-2-fluoroaniline (392.0 mg, 2.18 mmol) in isopropanol (10 mL) at room temperature. The mixture was heated to 90° C., and stirred for 1 hour. The reaction solution was cooled to 20° C., quenched by adding saturated sodium bicarbonate solution (10 mL), and extracted with ethyl acetate (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=2:1) to afford compound 1-9. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.41 (s, 1H), 7.80 (s, 1H), 7.60 (s, 2H), 7.24 (s, 1H), 4.82 (t, J=6.0 Hz, 1H), 3.95-4.05 (m, 1H), 3.91 (s, 3H), 3.70-3.95 (m, 2H), 3.50-3.60 (m, 1H), 3.05-3.15 (m, 2H), 1.85-1.95 (m, 1H), 1.55-1.65 (m, 1H). LC-MS: m/z=545.0 [M+H]+.


The Ninth Step

Compound 1-9 (300.0 mg, 551.14 umol) was dissolved in methanol (15 mL), and to the mixture was added potassium carbonate (380.9 mg, 2.76 mmol). The reaction solution was stirred at 40° C. for 14 hours. The mixture was concentrated under reduced pressure, diluted with water (10 mL), and extracted with ethyl acetate (40 mL×4). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound 1-10. 1H NMR (400 MHz, DMSO-d6) δ 9.81 (s, 1H), 8.41 (s, 2H), 7.76 (s, 1H), 7.59 (s, 2H), 7.29 (s, 1H), 4.73 (t, J=6.0 Hz, 1H), 3.99 (s, 3H), 3.20-3.30 (m, 4H), 2.95-3.05 (m, 2H), 1.95-2.05 (m, 1H), 1.75-1.85 (m, 1H). LC-MS: m/z=449.1 [M+H]+.


The Tenth Step

Compound 1-10 (240.0 mg, 534.16 μmol) was dissolved in a mixed solvent of tetrahydrofuran (6 mL) and water (6 mL) at 0° C., and to the mixture was added sodium bicarbonate (134.6 mg, 1.60 mmol). After the addition of sodium bicarbonate, acryloyl chloride (48.4 mg, 534.16 μmol) was added slowly dropwise to the mixture. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding methanol (1 mL) and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain the compound of example 1. 1H NMR (400 MHz, CDCl3) δ 8.68 (s, 1H), 8.43 (t, J=8.8 Hz, 1H), 7.34-7.45 (m, 2H), 7.10 (s, 1H), 6.60 (dd, J1=16.8 Hz, J2=9.6 Hz, 1H), 5.71 (dd, J1=16.8 Hz, J2=2.0, Hz, 1H), 6.60 (dd, J1=9.6 Hz, J2=2.0 Hz, 1H), 4.66 (t, J=5.6 Hz, 1H), 3.96 (s, 3H), 3.85-3.95 (m, 2H), 3.65-3.75 (m, 2H), 2.95-3.05 (m, 2H), 1.85-1.95 (m, 1H), 1.55-1.65 (m, 1H). LC-MS: m/z=503.1 [M+H]+.


Example 2



embedded image


Synthetic Route:




embedded image


The First Step

Compound 2-2 was obtained by referring to the eighth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 11.79 (s, 1H), 8.44 (s, 1H), 8.37 (s, 1H), 7.60-7.70 (m, 1H), 7.45-7.55 (m, 1H), 7.35 (s, 1H), 4.95 (t, J=5.6 Hz, 1H), 4.00-4.05 (m, 1H), 3.97 (s, 3H), 3.70-3.90 (m, 3H), 3.10-3.20 (m, 2H), 1.80-1.90 (m, 1H), 1.61 (d, J=10.4 Hz, 1H). LC-MS: m/z=529.1 [M+H]+.


The Second Step

Compound 2-2 (70.0 mg, 132.61 μmol) was dissolved in methanol (2 mL), and to the mixture was added potassium carbonate (91.6 mg, 663.04 μmol). The reaction solution was stirred at 50° C. for 14 hours. The reaction solution was concentrated under reduced pressure, diluted with water (10 mL) and extracted with ethyl acetate (20 mL×4). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound 2-3. 1H NMR (400 MHz, DMSO-d6) δ 9.78 (s, 1H), 8.39 (s, 2H), 7.79 (s, 1H), 7.50-7.60 (m, 1H), 7.38-7.48 (m, 1H), 7.29 (s, 1H), 4.74 (t, J=6.0 Hz, 1H), 3.99 (s, 3H), 3.40-3.50 (m, 4H), 2.95-3.05 (m, 2H), 1.98 (d, J=11.2 Hz, 1H), 1.80-1.90 (m, 1H). LC-MS: m/z=433.1 [M+H]+.


The Third Step

Compound of example 2 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.66 (s, 1H), 8.37 (s, 1H), 7.79 (s, 1H), 7.55-7.65 (m, 1H), 7.41 (t, J=8.4 Hz, 1H), 7.22 (s, 1H), 6.75 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.16 (d, J=16.8, Hz, 1H), 5.68 (d, J=9.6 Hz, 1H), 4.65-4.75 (m, 1H), 3.90-3.95 (m, 1H), 3.90 (s, 3H), 3.65-3.85 (m, 2H), 3.45-3.55 (m, 1H), 3.02-3.12 (m, 2H), 1.75-1.85 (m, 1H), 1.51 (d, J=10.0 Hz, 1H). LC-MS: m/z=487.0 [M+H]+.


Example 3



embedded image


Synthetic Route:




embedded image


The First Step

Compound 3-2 was obtained by referring to the eighth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 11.45 (s, 1H), 8.87 (s, 1H), 8.35 (s, 1H), 8.01-8.07 (m, 1H), 7.70-7.80 (m, 1H), 7.57 (t, J=8.8 Hz, 1H), 7.31 (s, 1H), 4.98 (t, J=5.6 Hz, 1H), 3.96-4.44 (m, 1H), 3.96 (s, 3H), 3.70-3.85 (m, 2H), 3.50-3.55 (m, 1H), 1.80-1.90 (m, 1H), 1.62 (d, J=10.4 Hz, 1H). LC-MS: m/z=511.1 [M+H]+.


The Second Step

Compound 3-3 was obtained by referring to the second step of Example 2. 1H NMR (400 MHz, DMSO-d6) δ 9.60 (s, 1H), 8.50 (s, 1H), 8.05-8.12 (m, 1H), 7.70-7.85 (m, 2H), 7.46 (t, J=9.2 Hz, 1H), 7.26 (s, 1H), 4.75 (t, J=5.6 Hz, 1H), 3.98 (s, 3H), 3.15-3.30 (m, 2H), 2.95-3.10 (m, 2H), 2.88-2.95 (m, 2H), 1.90-2.00 (m, 1H), 1.70-1.80 (m, 1H). LC-MS: m/z=415.1 [M+H]+.


The Third Step

Compound 3-3 (40.0 mg, 96.42 μmol) was dissolved in a mixed solvent of tetrahydrofuran (4 mL) and water (4 mL) at 0° C. To the mixture was added sodium bicarbonate (24.3 mg, 289.25 μmol), and added slowly acryloyl chloride (10.57 mg, 115.70 μmol) dropwise. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding methanol (1 mL), and extracted with ethyl acetate (20 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain the compound of example 3. 1H NMR (400 MHz, DMSO-d6) δ 9.59 (s, 1H), 8.49 (s, 1H), 8.08-8.12 (m, 1H), 7.75-7.85 (m, 2H), 7.46 (t, J=9.2 Hz, 1H), 7.22 (s, 1H), 6.74 (dd, J1=16.4 Hz, J2=10.0 Hz, 1H), 6.17 (d, J=12.0 Hz, 1H), 5.69 (d, J=10.0 Hz, 1H), 4.73 (t, J=5.6, Hz, 1H), 3.95-4.00 (m, 1H), 3.89 (s, 3H), 3.65-3.80 (m, 2H), 3.40-3.50 (m, 1H), 3.05-3.10 (m, 2H), 1.75-1.90 (m, 1H), 1.51 (d, J=10.0 Hz, 1H). LC-MS: m/z=469.1 [M+H]+.


Example 4



embedded image


Synthetic Route:




embedded image


The First Step

Compound 4-2 was obtained by referring to the eighth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 11.50 (s, 1H), 8.94 (s, 1H), 8.47 (s, 1H), 8.39 (d, J=2.0 Hz 1H), 8.10-8.15 (m, 1H), 8.05-8.10 (m, 1H), 7.35 (s, 1H), 5.06 (t, J=5.6 Hz, 1H), 3.97 (s, 3H), 3.70-3.90 (m, 4H), 3.10-3.20 (m, 2H), 1.80-1.90 (m, 1H), 1.61 (d, J=10.4 Hz, 1H). LC-MS: m/z=518.1 [M+H]+.


The Second Step

Compound 4-3 was obtained by referring to the ninth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.90 (s, 1H), 8.64 (s, 1H), 8.37 (s, 1H), 8.05-8.15 (m, 1H), 7.90-8.00 (m, 1H), 7.72 (s, 1H), 7.30 (s, 1H), 4.70-4.80 (m, 1H), 3.99 (s, 3H), 3.02-3.10 (m, 2H), 2.75-2.85 (m, 4H), 1.93 (d, J=9.6 Hz, 1H), 1.60-1.70 (m, 1H). LC-MS: m/z=422.1 [M+H]+.


The Third Step

Compound of example 4 was obtained by referring to the third step of Example 3. 1H NMR (400 MHz, DMSO-d6) δ 9.91 (s, 1H), 8.65 (s, 1H), 8.39 (s, 1H), 8.05-8.15 (m, 1H), 7.92-8.00 (m, 1H), 7.85 (s, 1H), 7.28 (s, 1H), 6.74 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.60 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.77 (t, J=5.6 Hz, 1H), 3.92 (s, 3H), 3.85-3.90 (m, 1H), 3.65-3.80 (m, 2H), 3.40-3.50 (m, 1H), 3.00-3.10 (m, 2H), 1.80-1.90 (m, 1H), 1.52 (d, J=10.0 Hz, 1H). LC-MS: m/z=476.0 [M+H]+.


Example 5



embedded image


Synthetic Route:




embedded image


The First Step

Compound 5-2 was obtained by referring to the eighth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.87 (s, 1H), 8.41 (s, 1H), 7.65-7.75 (m, 1H), 7.50-7.65 (m, 1H), 7.35-7.45 (m, 2H), 4.97 (t, J=5.6 Hz, 1H), 3.98-4.00 (m, 1H), 3.98 (s, 3H), 3.70-3.90 (m, 3H), 3.10-3.20 (m, 2H), 1.80-1.90 (m, 1H), 1.62 (d, J=10.0 Hz, 1H). LC-MS: m/z=511.2 [M+H]+.


The Second Step

Compound 5-3 was obtained by referring to the ninth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.87 (s, 1H), 8.40 (s, 1H), 7.85 (s, 1H), 7.45-7.60 (m, 1H), 7.20-7.40 (m, 2H), 4.60 (t, J=5.6 Hz1H), 3.99 (s, 3H), 3.30-3.50 (m, 4H), 3.00-3.15 (m, 2H), 2.00-2.10 (m, 1H), 1.80-1.90 (m, 1H). LC-MS: m/z=415.1 [M+H]+.


The Third Step

Compound of example 5 was obtained by referring to the third step of Example 3. 1H NMR (400 MHz, DMSO-d6) δ 9.67 (s, 1H), 8.38 (s, 1H), 7.76 (s, 1H), 7.50-7.60 (m, 2H), 7.25-7.35 (m, 1H), 7.22 (s, 1H), 6.75 (dd, J1=16.4, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.4 Hz, J2=2.0 Hz, 1H), 5.69 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.71 (t, J=5.6 Hz, 1H), 3.95-4.00 (m, 1H), 3.92 (s, 3H), 3.65-3.80 (m, 2H), 3.42-3.50 (m, 1H), 3.00-3.10 (m, 2H), 1.75-1.85 (m, 1H), 1.52 (d, J=10.0 Hz, 1H). LC-MS: m/z=469.0 [M+H]+.


Example 6



embedded image


Synthetic Route:




embedded image


The First Step

Compound 6-2 was obtained by referring to the eighth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.77 (s, 1H), 8.56 (s, 1H), 8.27 (s, 1H), 7.90-7.80 (m, 2H), 7.55-7.65 (m, 1H), 7.25 (s, 1H), 4.90 (t, J=5.6 Hz, 1H), 3.92-4.00 (m, 1H), 3.95 (s, 3H), 3.80-3.90 (m, 1H), 3.70-3.80 (m, 1H), 3.50-3.60 (m, 1H), 3.00-3.10 (m, 2H), 1.80-1.90 (m, 1H), 1.60 (d, J=10.0 Hz, 1H). LC-MS: m/z=577.1 [M+H]+.


The Second Step

Compound 6-3 was obtained by referring to the second step of Example 2. 1H NMR (400 MHz, DMSO-d6) δ 10.01 (s, 1H), 8.57 (s, 1H), 8.33 (s, 1H), 8.00-8.10 (m, 2H), 7.55-7.70 (m, 1H), 7.32 (s, 1H), 4.80-4.90 (m, 1H), 4.00 (s, 3H), 3.00-3.20 (m, 4H), 1.80-2.10 (m, 2H), 1.10-1.30 (m, 2H). LC-MS: m/z=481.1 [M+H]+.


The Third Step

Compound of example 6 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.54 (s, 1H), 8.23 (s, 1H), 7.90-8.00 (m, 1H), 7.81 (s, 1H), 7.55-7.65 (m, 1H), 7.24 (s, 1H), 6.65-6.80 (m, 1H), 6.17 (d, J=15.2 Hz, 1H), 5.68 (d, J=8.8 Hz, 1H), 4.70-4.80 (m, 1H), 3.90 (s, 3H), 3.80-3.90 (m, 1H), 3.50-3.80 (m, 3H), 3.00-3.10 (m, 2H), 1.80-1.90 (m, 1H), 1.45-1.55 (m, 1H). LC-MS: m/z=535.1 [M+H]+


Example 7



embedded image


Synthetic Route:




embedded image


embedded image


embedded image


The First Step

Compound B-1 (2.60 g, 11.54 mmol) was dissolved in ethanol (30 mL), and to the mixture was added sodium borohydride (1.09 g, 28.85 mmol). The reaction solution was stirred at 30° C. for 2 hours. The reaction solution was quenched by adding saturated ammonium chloride solution (30 mL) and extracted with dichloromethane (40 mL×2). The organic phases were combined, washed with saturated brine (40 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound B-2. 1H NMR (400 MHz, CDCl3) δ 4.00-4.32 (m, 3H), 2.10-2.20 (m, 1H), 1.90-2.00 (m, 3H), 1.60-1.90 (m, 4H), 1.40-1.50 (m, 9H).


The Second Step

Compound B-2 (2.60 g, 11.44 mmol), triethylamine (3.2 mL, 22.88 mmol), and 4-dimethylaminopyridine (1.68 g, 13.73 mmol) were dissolved in dichloromethane (60 mL), and to the mixture was added p-toluenesulfonyl chloride (2.62 g, 13.73 mmol). The reaction solution was stirred at 25° C. for 16 hours, and then quenched by adding saturated ammonium chloride aqueous solution (60 mL×3). The organic phase was washed with saturated brine (60 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound B-3. 1H NMR (400 MHz, CDCl3) δ 7.77 (d, J=8.0 Hz, 2H), 7.33 (d, J=8.0 Hz, 2H), 4.75-4.90 (m, 1H), 4.05-4.25 (m, 2H), 2.45 (s, 3H), 1.50-1.90 (m, 4H), 1.85-2.20 (m, 4H), 1.30-1.45 (s, 9H). LC-MS: m/z=404.1 [M+Na]+.


The Third Step

Compound 7-1 (2.00 g, 5.65 mmol) and potassium carbonate (1.56 g, 11.29 mmol) were added to N,N-dimethylacetamide (20 mL), and then to the mixture was added compound B-3 (4.20 g, 8.47 mmol). The reaction was heated to 70° C., and stirred for 16 hours. The reaction solution was cooled to 30° C., poured into water (30 mL), and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed sequentially with water (30 mL×3) and saturated brine (30 mL×3), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (petroleum ether:ethyl acetate=1:1) to afford compounds 7-2 and 8-1.


Compound 7-2: 1H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 8.42 (t, J=8.4 Hz, 1H), 7.26-7.40 (m, 3H), 6.96 (s, 1H), 4.80-4.90 (m, 1H), 4.15-4.35 (m, 2H), 4.00 (s, 3H), 2.10-2.35 (m, 4H), 1.90-2.10 (m, 4H), 1.48 (s, 9H). According to two-dimensional nuclear magnetic NOE identification: the single hydrogen on the carbon of the piperidine ring connected to the oxygen atom is not observed to be associated with the hydrogen of methylene on the bridge ring, and it is determined to be a cis structure. LC-MS: m/z 563.2 [M+H]+.


Compound 8-1: 1H NMR (400 MHz, CDCl3) δ 8.70 (s, 1H), 8.53 (t, J=8.4 Hz, 1H), 7.26-7.40 (m, 3H), 7.21 (s, 1H), 4.70-4.80 (m, 1H), 4.20-4.40 (m, 2H), 4.01 (s, 3H), 2.10-2.20 (m, 4H), 1.85-2.00 (m, 2H), 1.65-1.80 (m, 2H), 1.50 (s, 9H). According to two-dimensional nuclear magnetic NOE identification: the single hydrogen on the carbon of the piperidine ring connected to the oxygen atom is observed to be associated with the hydrogen of methylene on the bridge ring, and it is determined to be a trans structure. LC-MS: m/z 563.2 [M+H]+.


The Fourth Step

Compound 7-2 (400.0 mg, 709.92 μmol) was dissolved in a solution of hydrogen chloride in dioxane (4 N, 6 mL). The reaction solution was reacted at 0° C. for 0.3 hour. The reaction solution was concentrated under reduced pressure, diluted with water (10 mL), and washed with ethyl acetate (10 mL). The aqueous phase was adjusted to pH of 10 with the solid potassium carbonate and extracted with ethyl acetate (10 mL×3). The organic phases were combined, and dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (methanol:dichloromethane=1:10) to afford compound 7-3. 1H NMR (400 MHz, DMSO-d6) δ 9.96 (s, 1H), 8.39 (s, 1H), 7.96 (s, 1H), 7.55-7.65 (m, 2H), 7.26 (s, 1H), 4.95-5.00 (m, 1H), 3.90-4.05 (m, 5H), 2.45-2.50 (m, 2H), 2.25-2.35 (m, 2H), 2.10-2.20 (m, 2H), 1.90-2.05 (m, 2H).


The Fifth Step

Compound 7-3 (230.0 mg, 496.41 μmol) was dissolved in a mixed solvent of tetrahydrofuran (6 mL) and water (6 mL) at 0° C. To the mixture was added sodium bicarbonate (125.1 mg, 1.49 mmol), and then added slowly acryloyl chloride (35.9 mg, 397.12 μmol) dropwise. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding methanol (1 mL), and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain the compound of example 7. 1H NMR (400 MHz, DMSO-d6) δ 9.63 (s, 1H), 8.39 (s, 1H), 7.74 (s, 1H), 7.50-7.60 (m, 2H), 7.26 (s, 1H), 6.75 (dd, J1=16.4 Hz, J2=10.0 Hz, 1H), 6.19 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.70 (dd, J1=10.0 Hz, J2=2.4 Hz, 1H), 4.88-4.90 (m, 1H), 4.50-4.61 (m, 2H), 3.97 (s, 3H), 2.20-2.30 (m, 2H), 1.95-2.15 (m, 5H), 1.80-1.95 (m, 1H). LC-MS: m/z=517.1 [M+H]+.


Example 8



embedded image


Synthetic Route:




embedded image


The First Step

Compound 8-2 was obtained by referring to the fourth step of Example 7. 1H NMR (400 MHz, DMSO-d6) δ 10.13 (s, 1H), 8.39 (s, 1H), 8.10 (s, 1H), 7.55-7.70 (m, 2H), 7.23 (s, 1H), 4.90-5.00 (m, 1H), 3.90-4.05 (m, 2H), 3.94 (s, 3H), 2.30-2.40 (m, 2H), 2.10-2.20 (m, 2H), 1.80-2.00 (m, 4H).


The Second Step

Compound of example 8 was obtained by referring to the fifth step of Example 7. 1H NMR (400 MHz, DMSO-d6) δ 9.67 (s, 1H), 8.39 (s, 1H), 7.80 (s, 1H), 7.55-7.70 (m, 2H), 7.26 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.23 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.72 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.90-5.10 (m, 1H), 4.55-4.60 (m, 2H), 3.92 (s, 3H), 2.20-2.40 (m, 2H), 1.80-2.10 (m, 4H), 1.66 (t, J=10.4 Hz, 1H), 1.50 (t, J=10.4 Hz, 1H). LC-MS: m/z=517.1 [M+H]+.


Example 9



embedded image


Synthetic Route:




embedded image


embedded image


embedded image


embedded image


The First Step

Compound A-2 (2.00 g, 9.38 mmol) was dissolved in dichloromethane (50 mL), and to the mixture was added Dess-Martin periodinane (4.77 g, 11.25 mmol). The reaction solution was stirred at 25° C. for 2 hours. The reaction solution was quenched by adding saturated aqueous sodium bicarbonate solution (30 mL), and extracted with dichloromethane (30 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:4) to afford compound C-2. 1H NMR (400 MHz, CDCl3) δ 3.95-4.10 (m, 2H), 3.85-3.95 (m, 2H), 3.10-3.20 (m, 2H), 2.10-2.20 (m, 1H), 1.75-1.80 (m, 1H), 1.45 (s, 9H).


The Second Step

Compound C-2 (1.70 g, 8.05 mmol) was dissolved in methanol (20 mL), and to the mixture were added hydroxylamine hydrochloride (671.0 mg, 9.66 mmol) and sodium carbonate (511.7 mg, 4.83 mmol). The reaction solution was stirred at 25° C. for 16 hours. The reaction solution was concentrated under reduced pressure, diluted by adding dichloromethane (30 mL), washed with saturated aqueous sodium bicarbonate solution (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound C-3. 1H NMR (400 MHz, CDCl3) δ 7.40-7.50 (m, 1H), 3.70-3.90 (m, 4H), 3.45-3.60 (m, 1H), 3.05-3.20 (m, 1H), 2.10-2.20 (m, 1H), 1.60-1.65 (m, 1H), 1.46 (s, 9H)


The Third Step

Compound C-3 (2.00 g, 8.84 mmol) was dissolved in methanol (20 mL), and to the mixture was added Raney nickel (0.50 g). The reaction solution was stirred at 20° C. under hydrogen atmosphere (hydrogen balloon) for 16 hours, and then filtered through celite. The filtrate was concentrated under reduced pressure and dried to afford compound C-4. 1H NMR (400 MHz, CDCl3) δ 3.85-4.10 (m, 1H), 3.45-3.75 (m, 4H), 2.60-2.80 (m, 2H), 1.85-1.95 (m, 1H), 1.50-1.60 (m, 1H), 1.47 (s, 9H).


The Fourth Step

Compound 9-1 (900.0 mg, 3.93 mmol) was dissolved in N,N-dimethylformamide (20 mL), and to the mixture were added potassium carbonate (1.09 g, 7.85 mmol) and compound C-4 (1.50 g, 7.07 mmol). The reaction solution was stirred at 90° C. for 16 hours. The reaction solution was cooled to 20° C., diluted with water (30 mL) and extracted with ethyl acetate (30 mL×2). The organic phases were combined, washed sequentially with water (40 mL×2) and saturated brine (40 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (petroleum ether:ethyl acetate=3:1) to afford compounds 9-2 and 10-1.


Compound 10-1: 1H NMR (400 MHz, CDCl3) δ 7.51 (s, 1H), 6.27 (s, 1H), 5.36 (d, J=4 Hz, 1H), 3.97 (s, 3H), 3.93 (s, 3H), 3.80-3.90 (m, 1H), 3.55-3.70 (m, 2H), 3.45-3.53 (m, 1H), 3.35-3.45 (m, 1H), 2.65-2.85 (m, 2H), 1.90-2.00 (m, 1H), 1.45-1.55 (m, 1H), 1.51 (s, 9H). According to two-dimensional nuclear magnetic NOE identification: the single hydrogen on the carbon of the piperidine ring connected to the phenylamine is strongly associated with the methylene group next to the amino group of the piperidine ring, and not associated with the hydrogen of methylene on the bridge ring, and it is determined to be a cis structure. LC-MS: m/z=444.2 [M+Na]+.


Compound 9-2: 1H NMR (400 MHz, CDCl3) δ 7.47 (s, 1H), 6.56 (s, 1H), 5.15-5.25 (m, 1H), 3.97 (s, 3H), 3.91 (s, 3H), 3.65-3.80 (m, 4H), 3.40-3.50 (m, 1H), 2.45-2.60 (m, 2H), 2.40-2.45 (m, 1H), 1.50-1.55 (m, 1H), 1.51 (s, 9H). LC-MS: m/z=444.2 [M+Na]+. The structure of compound 9-2 was deduced by the results of structural identification of compound 10-2.


The Fifth Step

Compound 9-2 (580.0 mg, 1.38 mmol) was dissolved in methanol (100 mL), and to the mixture was added wet palladium/carbon (10%, 150 mg). The reaction solution was stirred at 20° C. under hydrogen atmosphere (hydrogen balloon) for 0.5 hour, and then filtered through celite. The filtrate was concentrated under reduced pressure, and dried to afford compound 9-3. 1H NMR (400 MHz, CDCl3) δ 7.04 (s, 1H), 6.10 (s, 1H), 3.83 (s, 3H), 3.82 (s, 3H), 3.70-3.80 (m, 1H), 3.40-3.60 (m, 4H), 2.60-2.70 (m, 2H), 1.85-1.95 (m, 1H), 1.45 (s, 9H), 1.40-1.45 (m, 1H).


The Sixth Step

Compound 9-3 (500.0 mg, 1.28 mmol) and ammonium acetate (984.6 mg, 12.77 mmol) were added to trimethyl orthoformate (12.47 g, 117.48 mmol). The reaction solution was stirred at 70° C. for 10 hours. The reaction solution was cooled to 20° C., and concentrated under reduced pressure. The residue was slurried with water (10 mL), filtered, and dried to afford compound 9-4. 1H NMR (400 MHz, DMSO-d6) δ 7.87 (s, 1H), 7.06 (s, 1H), 7.04 (s, 1H), 5.18 (d, J=5.2 Hz, 1H), 3.94 (s, 3H), 3.75-3.80 (m, 1H), 3.35-3.55 (m, 4H), 2.65-2.75 (m, 2H), 1.92-2.00 (m, 1H), 1.40 (s, 9H), 1.30-1.40 (m, 1H). LC-MS: m/z=387.2 [M+Na]+.


The Seventh Step

Compound 9-4 (400.0 mg, 1.04 mmol) was added to a solution of hydrogen chloride in dioxane (4 N, 10 mL). The reaction solution was stirred at 0° C. for 0.4 hour. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 9-5. LC-MS: m/z=287.0 [M+H]+.


The Eighth Step

At 0° C., the hydrochloride salt of 9-5 (340.0 mg, 1.19 mmol) and triethylamine (1.7 mL, 11.87 mmol) were dissolved in dichloromethane (10 mL), and to the mixture was added trifluoroacetic anhydride (498.8 mg, 2.37 mmol) dropwise. The reaction solution was stirred at 20° C. for 1 hour, and then concentrated under reduced pressure. The residue was slurried with water (40 mL), filtered, and dried to afford compound 9-6. 1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 7.87 (s, 1H), 7.02-7.88 (m, 2H), 4.95-5.05 (m, 1H), 3.98 (s, 3H), 3.80-3.95 (m, 3H), 3.60-3.70 (m, 1H), 3.51-3.60 (m, 1H), 2.80-2.90 (m, 2H), 2.00-2.10 (m, 1H), 1.52 (d, J=10.0 Hz, 1H). LC-MS: m/z=383.1 [M+H]+.


The Ninth Step

Compound 9-6 (280.0 mg, 732.34 μmol) was dissolved in phosphorus oxychloride (20 mL, 215.22 mmol), and to the mixture was added N,N-dimethylformamide (53.5 mg, 732.34 μmol). The reaction solution was stirred at 100° C. for 5 hours, and then concentrated under reduced pressure. To the residue was added a solution of 3,4-dichloro-2-fluoroaniline (263.65 mg, 1.46 mmol) in isopropanol (10 mL). The reaction solution was stirred at 90° C. for 1 hour. The reaction solution was cooled to 20° C., quenched by adding saturated aqueous sodium bicarbonate solution (10 mL), and extracted with ethyl acetate (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=2:1) to afford compound 9-7. 1H NMR (400 MHz, DMSO-d6) δ 9.46 (s, 1H), 8.29 (s, 1H), 7.52-7.65 (m, 2H), 7.12-7.20 (m, 2H), 5.64 (d, J=3.6 Hz, 1H), 3.98 (s, 3H), 3.82-3.98 (m, 3H), 3.65-3.75 (m, 1H), 3.55-3.60 (m, 1H), 2.95-3.05 (m, 2H), 2.00-2.10 (m, 1H), 1.56 (d, J=10.0 Hz, 1H). LC-MS: m/z=544.2 [M+H]+.


The Tenth Step

Compound 9-7 (290.0 mg, 533.74 μmol) was dissolved in methanol (15 mL), and to the mixture was added potassium carbonate (369.0 mg, 2.67 mmol). The reaction solution was stirred at 55° C. for 14 hours. The reaction solution was concentrated under reduced pressure, quenched by adding water (10 mL), and extracted with ethyl acetate (20 mL×4). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound 9-8. 1H NMR (400 MHz, DMSO-d6) δ 9.50 (s, 1H), 8.28 (s, 1H), 7.50-7.65 (m, 2H), 7.16 (s, 1H), 7.14 (s, 1H), 5.91 (d, J=4.8 Hz, 1H), 4.01 (s, 3H), 3.85-3.95 (m, 1H), 3.15-3.20 (m, 2H), 2.80-2.90 (m, 2H), 1.90-2.00 (m, 2H), 1.70-1.80 (m, 1H), 1.10 (d, J=6.4 Hz, 1H). LC-MS: m/z=448.2 [M+H]+.


The Eleventh Step

Compound 9-8 (200.0 mg, 446.11 μmol) was dissolved in a mixed solvent of tetrahydrofuran (6 mL) and water (6 mL) at 0° C. To the mixture was added sodium bicarbonate (112.4 mg, 1.34 mmol), and added slowly acryloyl chloride (36.3 mg, 401.50 μmol) dropwise. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding MeOH (1 mL), and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain the compound of example 9. 1H NMR (400 MHz, DMSO-d6) δ 9.45 (s, 1H), 8.28 (s, 1H), 7.55-7.65 (m, 2H), 7.17 (s, 1H), 7.12 (s, 1H), 6.75 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.68 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 5.40 (d, J=4.8 Hz, 1H), 3.96 (s, 3H), 3.80-3.90 (m, 2H), 3.65-3.75 (m, 2H), 3.45-3.52 (m, 1H), 2.90-3.05 (m, 2H), 1.90-2.00 (m, 1H), 1.47 (d, J=9.6 Hz, 1H). LC-MS: m/z=502.1 [M+H]+.


Example 10



embedded image


Synthetic Route:




embedded image


The First Step

Compound 10-2 was obtained by referring to the fifth step of Example 9. 1H NMR (400 MHz, CDCl3) δ 6.78 (s, 1H), 6.13 (s, 1H), 3.86 (s, 3H), 3.84 (s, 3H), 3.60-3.80 (m, 4H), 3.32-3.36 (m, 1H), 2.50-2.60 (m, 1H), 2.40-2.48 (m, 1H), 2.30-2.40 (m, 1H), 1.51 (s, 9H), 1.40-1.45 (m, 1H).


The Second Step

Compound 10-3 was obtained by referring to the sixth step of Example 9. 1H NMR (400 MHz, DMSO-d6) δ 7.85 (s, 1H), 7.05 (s, 1H), 6.72 (s, 1H), 5.95 (d, J=3.2 Hz, 1H), 3.97 (s, 3H), 3.55-3.80 (m, 4H), 3.25-3.30 (m, 1H), 2.51-2.60 (m, 2H), 1.75-1.80 (m, 1H), 1.46 (s, 9H), 1.35-1.40 (m, 1H). LC-MS: m/z=387.2 [M+Na]+.


The Third Step

The hydrochloride salt of compound 10-4 was obtained by referring to the seventh step of Example 9. LC-MS: m/z=287.0 [M+H]+.


The Fourth Step

Compound 10-5 was obtained by referring to the eighth step of Example 9. 1H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 7.85 (d, J=3.2 Hz, 1H), 7.06 (s, 1H), 6.76 (s, 1H), 6.02 (d, J=3.2 Hz, 1H), 4.05-4.15 (m, 2H), 3.98 (s, 3H), 3.85-3.95 (m, 1H), 3.75-3.80 (m, 1H), 3.45-3.50 (m, 1H), 2.55-2.70 (m, 3H), 1.50-1.60 (m, 1H). LC-MS: m/z=383.1 [M+H]+.


The Fifth Step

Compound 10-6 was obtained by referring to the ninth step of Example 9. 1H NMR (400 MHz, DMSO-d6) δ 9.35 (s, 1H), 8.29 (s, 1H), 7.60-7.70 (m, 1H), 7.55-7.60 (m, 1H), 7.15 (s, 1H), 6.81 (s, 1H), 6.18 (d, J=3.6 Hz, 1H), 4.10-4.25 (m, 2H), 4.03 (s, 3H), 3.80-4.00 (m, 2H), 3.50-3.63 (m, 1H), 2.65-2.75 (m, 2H), 2.55-2.60 (m, 1H), 1.50-1.56 (m, 1H). LC-MS: m/z=544.2 [M+H]+.


The Sixth Step

Compound 10-7 was obtained by referring to the tenth step of Example 9. 1H NMR (400 MHz, DMSO-d6) δ 9.69 (s, 1H), 8.26 (s, 1H), 7.50-7.65 (m, 2H), 7.12 (s, 1H), 6.95 (d, J=8.0 Hz, 1H), 4.10-4.20 (m, 1H), 4.01 (s, 3H), 3.60-3.70 (m, 2H), 3.20-3.30 (m, 2H), 2.40-2.50 (m, 3H), 1.60-1.70 (m, 1H). LC-MS: m/z=448.2 [M+H]+.


The Seventh Step

Compound of example 10 was obtained by referring to the eleventh step of Example 9. 1H NMR (400 MHz, DMSO-d6) δ 9.38 (s, 1H), 8.27 (s, 1H), 7.55-7.65 (m, 2H), 7.14 (s, 1H), 6.75-6.85 (m, 2H), 6.18 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 6.14 (d, J1=4.0 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.10-4.20 (m, 1H), 4.02 (s, 3H), 3.85-3.95 (m, 2H), 3.75-3.80 (m, 1H), 3.50-3.55 (m, 1H), 2.50-2.65 (m, 3H), 1.40-1.50 (m, 1H). LC-MS: m/z=502.1 [M+H]+.


Example 11



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Compound E-1 (500.0 mg, 2.79 mmol) was added to dichloromethane (6 mL), and compound E-2 (475.7 mg, 5.59 mmol) was added under nitrogen protection. The reaction solution was stirred at 25° C. for 2 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (petroleum ether:ethyl acetate=3:1) to afford compound E-3. 1H NMR (400 MHz, CDCl3) δ 7.03-6.95 (m, 1H), 5.89-6.03 (m, 1H), 3.72 (s, 3H), 3.09 (dd, J1=6.4 Hz, J2=1.6 Hz, 2H), 2.20-2.50 (m, 4H), 1.63-1.56 (m, 4H), 1.38-1.46 (m, 2H). LC-MS: m/z=184.0 [M+H]+.


The Second Step

To a solution of compound E-3 (367.0 mg, 2.00 mmol) dissolved in tetrahydrofuran (10 mL) and water (10 mL) was added lithium hydroxide monohydrate (168.1 mg, 4.01 mmol). The reaction solution was stirred at 25° C. for 1 hour, and then extracted with ethyl acetate (10 m). The aqueous phase was adjusted to pH of 3 with dilute hydrochloric acid and lyophilized to afford compound E-4. 1H NMR (400 MHz, DMSO-d6) δ 11.28 (brs, 1H), 7.10-6.86 (m, 1H), 6.17 (d, J=15.6 Hz, 1H), 3.85 (d, J=6.8 Hz, 2H), 3.31-3.20 (m, 4H), 1.61-1.89 (m, 6H). LC-MS: m/z=170.2 [M+H]+.


The Third Step

Compound 1-10 (50.0 mg, 111.28 μmol), HATU (63.5 mg, 166.92 μmol) and DIEA (36.0 mg, 278.21 μmol) were dissolved in N,N-dimethylformamide (8 mL), and to the mixture was added compound E-4 (56.5 mg, 333.85 μmol). The reaction solution was stirred at 25° C. for 16 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative high performance liquid chromatography (basic) to afford compound of example 11. 1H NMR (400 MHz, DMSO-d6) δ 9.69 (brs, 1H), 8.39 (s, 1H), 7.74 (s, 1H), 7.60 (s, 1H), 7.23 (s, 1H), 6.45-6.71 (m, 2H), 4.69 (s, 1H), 3.80-4.00 (m, 4H), 3.62-3.80 (m, 3H), 2.90-3.10 (m, 4H), 2.25-2.35 (m, 3H), 1.78-1.85 (m, 1H), 1.33-1.52 (m, 7H). LC-MS: m/z=600.2 [M+H]+.


Example 12



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Compound F-1 (1.00 g, 5.59 mmol) and DIEA (1.44 g, 11.17 mmol) were dissolved in tetrahydrofuran (10 mL), and to the mixture was added an aqueous solution of dimethylamine (33%, 1.53 g, 11.17 mmol) dropwise. The reaction solution was stirred at 70° C. for 1 hour. The reaction solution was cooled to room temperature, quenched with water (10 mL), and extracted with ethyl acetate (15 mL×2). The organic phases were combined and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (petroleum ether:ethyl acetate=0:1) to afford compound F-2. 1H NMR (400 MHz, CDCl3) δ 7.00-6.80 (m, 1H), 6.00-5.85 (m, 1H), 3.67 (s, 3H), 3.07-2.93 (m, 2H), 2.18 (s, 6H).


The Second Step

Compound F-3 was obtained by referring to the second step of Example 11. 1H NMR (400 MHz, DMSO-d6) δ 11.42 (brs, 1H), 6.78-6.94 (m, 1H), 6.19 (d, J=15.6 Hz, 1H), 3.87-3.91 (m, 2H), 2.73-2.60 (m, 6H).


The Third Step

Compound of example 12 was obtained by referring to the third step of Example 11. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (s, 1H), 7.73 (s, 1H), 7.51-7.63 (m, 2H), 7.20 (s, 1H), 6.59-6.69 (m, 1H), 6.47-6.56 (m, 1H), 4.68 (t, J=5.6 Hz, 1H), 3.84-3.94 (m, 4H), 3.64-3.78 (m, 2H), 3.50-3.45 (m, 1H), 2.98-3.10 (m, 4H), 2.14 (s, 6H), 1.74-1.91 (m, 1H), 1.50 (d, J=10.0 Hz, 1H).


Example 13



embedded image


Synthetic Route:




embedded image


The First Step

Compound 1-9 (6.80 g, 10.62 mmol) was added to pyridine hydrochloride (57.94 g, 501.35 mmol). The reaction solution was stirred at 180-185° C. for 1 hour. The reaction solution was cooled to room temperature, slurried with water (30 mL) and ethyl acetate (30 mL) at 20-25° C. for 1 hour, filtered, and dried to afford compound 13-2. LC-MS: m/z 530.9 [M+H]+.


The Second Step

Compound 13-2 (500.0 mg, 941.11 μmol) was dissolved in N,N-dimethylformamide (5 mL), and to the mixture were added 1,3-dioxolan-2-one (165.8 mg, 1.88 mmol) and potassium fluoride (54.7 mg, 941.11 μmol). The reaction solution was stirred at 110° C. for 18 hours, and potassium fluoride (164.0 mg, 2.82 mmol) and 1,3-dioxolan-2-one (414.4 mg, 4.71 mmol) were added again. The reaction solution was stirred at 110° C. for 3 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=15:1) to afford compound 13-3. LC-MS: m/z=575.2 [M+H]+.


The Third Step

Compound 13-3 (170.0 mg, 295.48 μmol) was dissolved in methanol (10 mL), and to the mixture was added potassium carbonate (163.4 mg, 1.18 mmol). The reaction solution was stirred at 80° C. for 3 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=15:1) to afford compound 13-4. LC-MS: m/z=479.2[M+H]+.


The Fourth Step

Compound of example 13 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.69 (s, 1H), 8.39 (s, 1H), 7.78 (s, 1H), 7.64-7.57 (m, 2H), 7.24 (s, 1H), 6.74 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.16 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.68 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.84 (t, J=5.2 Hz, 1H), 4.73-4.71 (m, 1H), 4.11 (t, J=5.2 Hz, 2H), 4.00-3.90 (m, 1H), 3.76-3.70 (m, 4H), 3.49 (d, J=13.2 Hz, 1H), 3.05 (brs, 2H), 1.86-1.77 (m, 1H), 1.51 (d, J=10.0 Hz, 1H). LC-MS: m/z=533.3 [M+H]+.


Example 14



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Compound G-1 (3.00 g, 40.50 mmol) was dissolved in dichloromethane (30 mL), and to the mixture were added 4-methylbenzenesulfonyl chloride (7.72 g, 40.50 mmol), triethylamine (10.24 g, 101.24 mmol) and 4-dimethylaminopyridine (247.4 mg, 2.02 mmol). The reaction solution was stirred at room temperature for 8 hours, concentrated under reduced pressure, slurried with water (150 mL), and filtered. The filter cake was washed with water and dried to afford compound G-2. 1H NMR (400 MHz, CDCl3) δ 7.79 (d, J=8.0 Hz, 2H), 7.37 (d, J=8.4 Hz, 2H), 5.38-5.23 (m, 1H), 4.80-4.60 (m, 4H), 2.47 (s, 3H).


The Second Step

Compound 13-2 (200.0 mg, 376.44 μmol) was dissolved in N,N-dimethylformamide (5 mL), and to the mixture were added potassium fluoride (65.6 mg, 1.13 mmol) and compound G-2 (257.8 mg, 1.13 mmol). The reaction solution was stirred at 110° C. for 18 hours. To the mixture were added potassium fluoride (65.6 mg, 1.13 mmol), potassium carbonate (52.0 mg, 376.44 μmol) and compound G-2 (257.8 mg, 1.13 mmol). The reaction solution was stirred at 110° C. for 5 hours. To the mixture were added compound G-2 (100.0 mg, 438.09 μmol) and potassium carbonate (30.0 mg, 217.07 μmol). The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=15:1) to afford compound 14-2. LC-MS: m/z=587.2 [M+H]+.


The Third Step

Compound 14-3 was obtained by referring to the third step of Example 13. LCMS: m/z=587.2 [M+H]+.


The Fourth Step

Compound of example 14 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 9.74 (s, 1H), 8.39 (s, 1H), 7.86 (s, 1H), 7.60 (s, 2H), 6.88 (s, 1H), 6.75 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.68 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 5.51-5.40 (m, 1H), 5.02-4.90 (m, 2H), 4.85-4.75 (m, 1H), 4.57-4.47 (m, 2H), 3.96-3.85 (m, 1H), 3.77-3.66 (m, 2H), 3.56-3.46 (m, 1H), 3.06 (m, 2H), 1.89-1.78 (m, 1H), 1.53 (d, J=10.4 Hz, 1H). LC=MS: m/z=545.3 [M+H]+.


Example 15



embedded image


Synthetic Route:




embedded image


The First Step

Compound 13-2 (200.0 mg, 376.44 μmol) was dissolved in N,N-dimethylformamide (2 mL), and then to the mixture were added sodium iodide (112.9 mg, 752.89 μmol), potassium fluoride (87.5 mg, 1.51 mmol) and 1-bromo-2-methoxyethane (130.8 mg, 941.11 μmol) at room temperature. The reaction solution was stirred at 110° C. for 5 hours. To the mixture were added potassium fluoride (87.5 mg, 1.51 mmol) and 1-bromo-2-methoxyethane (157.0 mg, 1.13 mmol) again, and the reaction solution was further stirred at 110° C. for 18 hours. The reaction solution was concentrated under reduced pressure, and isolated by preparative thin-layer chromatography (dichloromethane:methanol=7:1) to afford compound 15-2. LC-MS: m/z=589.3[M+H]+.


The Second Step

Compound 15-3 was obtained by referring to the third step of Example 13.


The Third Step



embedded image


Compound of example 15 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, DMSO-d6) δ 7.89 (s, 1H), 7.64 (s, 1H), 7.33 (d, J=8.4 Hz, 1H), 7.03 (s, 1H), 6.91 (s, 1H), 6.74 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.16 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.71-5.65 (m, 1H), 4.70-4.63 (m, 1H), 4.15-4.25 (m, 2H), 3.80-3.90 (m, 2H), 3.65-3.75 (m, 1H), 3.59 (t, J=4.8 Hz, 2H), 3.51-3.46 (m, 1H), 3.23 (s, 3H), 2.84-2.90 (m, 2H), 1.80-1.85 (m, 1H), 1.47-1.40 (m, 1H); LC-MS: m/z=547.2[M+H]+.


Example 16



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Compound 13-2 (1.00 g, 1.88 mmol) was dissolved in methanol (20 mL), and then to the mixture was added potassium carbonate (1.30 g, 9.41 mmol) in one portion at 20-25° C. The reaction solution was stirred at 20-25° C. for 18 hours, and then heated to and reacted at 50° C. for 5 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure. The residue was diluted with saturated aqueous citric acid solution (50 mL) and extracted with ethyl acetate (50 mL×3). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to afford compound 16-2.


The Second Step

Compound 16-2 (850.0 mg, 1.95 mmol) was dissolved in methanol (10 mL), and to the mixture were added triethylamine (988.0 mg, 9.76 mmol) and di-tert-butyl dicarbonate (1.28 g, 5.86 mmol) at 20° C. The reaction solution was stirred at 20-25° C. for 2 hours. The reaction solution was concentrated under reduced pressure, slurried with water (20 mL) for 0.5 hour, and filtered. The filter cake was dried to afford compound 16-3. LC-MS: m/z=635.3 [M+H]+.


The Third Step

Compound 16-3 (1.00 g, 1.57 mmol) was dissolved in methanol (15 mL), and then to the mixture was added potassium carbonate (1.09 g, 7.87 mmol) in one portion at 20-25° C. The reaction solution was stirred at 20-25° C. for 18 hours. The reaction solution was concentrated under reduced pressure, and the residue was adjusted to pH of 5 with saturated citric acid aqueous solution. The reaction solution was stirred at 20-25° C. for 0.5 hour and then filtered. The filter cake was dried to afford compound 16-4. LC-MS: m/z=535.3 [M+H]+.


The Fourth Step

Compound 16-4 (150.0 mg, 280.17 μmol) was dissolved in N,N-dimethylformamide (3 mL), and then to the mixture was added sodium hydride (60%, 28.0 mg, 700.42 μmol) in one portion at 0° C. The reaction solution was stirred at 0° C. for 0.5 hour, and then 2-bromo-N,N-dimethylethylamine (55.4 mg, 364.22 μmol) was added. The reaction solution was stirred at 20° C. for 2 hours, quenched by adding water (2 mL), and extracted with ethyl acetate (3 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=5:1) to afford compound 16-5. LC-MS: m/z=606.4 [M+H]+.


The Fifth Step

Compound 16-5 (60.0 mg, 98.93 μmol) was dissolved in a solution of hydrogen chloride in dioxane (4 N, 4 m), and stirred at 20-25° C. for 0.5 hour. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 16-6. LC-MS: m/z=506.3 [M+H]+.


The Sixth Step

The hydrochloride salt of compound 16-6 (50.0 mg, 92.10 μmol) was dissolved in tetrahydrofuran (0.5 mL) and water (0.5 mL). To the mixture was added sodium bicarbonate (38.7 mg, 460.52 μmol) at 0° C., and added slowly acryloyl chloride (3.3 mg, 36.84 μmol) dropwise. The reaction solution was stirred at 0° C. for 0.5 hour and at 20-25° C. for 18 hours. Acryloyl chloride (2.2 mg, 23.95 μmol) was added again, and the reaction solution was further stirred at 20-25° C. for 2 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=3:1) to afford compound of example 16. 1H NMR (400 MHz, CD3OD) δ 8.34 (s, 1H), 7.58 (s, 1H), 7.50-7.47 (m, 2H), 6.69 (s, 1H), 6.21-6.12 (m, 1H), 6.11-6.04 (m, 1H), 5.75 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.58-5.51 (m, 1H), 4.73-4.65 (m, 1H), 4.39 (t, J=6.2 Hz, 2H), 4.19 (d, J=10.8 Hz, 1H), 3.96-3.83 (m, 2H), 3.66-3.58 (m, 1H), 3.22-3.13 (m, 1H), 3.05-3.10 (m, 1H), 2.79 (t, J=6.2 Hz, 2H), 2.32 (s, 6H), 2.00-1.91 (m, 1H), 1.59 (d, J=10.0 Hz, 1H). LC-MS: m/z=560.4 [M+H]+.


Example 17



embedded image


Synthetic Route:




embedded image


The First Step

Compound 16-4 (150.0 mg, 280.17 μmol) was dissolved in N,N-dimethylformamide (3 mL), and to the mixture was added sodium hydride (60%, 44.82 mg, 1.12 mmol) in one portion at 0° C. The reaction solution was stirred at 0° C. for 0.5 hour, and 4-(2-bromoethyl)morpholine (163.1 mg, 840.50 μmol) was added. The reaction solution was further stirred at 0° C. for 2 hours. The reaction solution was quenched with water (20 mL) and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to afford compound 17-2.


LC-MS: m/z=648.4[M+H]+.


The Second Step

Compound 17-3 was obtained by referring to the fifth step of Example 16. LC-MS: m/z=548.3 [M+H]+.


The Third Step

Compound of example 17 was obtained by referring to the tenth step of Example 1.



1H NMR (400 MHz, CD3OD) δ 8.39 (s, 1H), 7.77 (s, 1H), 7.67-7.55 (m, 1H), 7.49-7.45 (m, 1H), 7.22 (s, 1H), 6.79 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.34 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.80 (dd, J=10.4 Hz, J2=2.4 Hz, 1H), 4.40-4.25 (m, 2H), 3.92-3.81 (m, 2H), 3.75-3.68 (m, 6H), 3.60-3.56 (m, 2H), 3.17-3.05 (m, 2H), 2.89 (t, J=5.2 Hz, 2H), 2.69-2.62 (m, 3H), 2.07-1.90 (m, 1H), 1.62 (d, J=10.4 Hz, 1H). LC-MS: m/z=602.4 [M+H]+.


Example 18



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

H-1 (2.00 g, 13.77 mmol) was dissolved in tetrahydrofuran (20 mL), and to the mixture was added triphenylphosphine (3.97 g, 15.15 mmol). The reaction solution was cooled to 0° C. Under nitrogen protection, carbon tetrabromide (5.02 g, 15.15 mmol) was added to the reaction solution in batches. The reaction solution was warmed to room temperature and stirred for 10 hours. The reaction solution was concentrated under reduced pressure, diluted with water (50 mL), and extracted with ethyl acetate (30 mL×3). The aqueous phase was adjusted to pH of 11 with 4N sodium hydroxide solution, and concentrated to remove water. Methanol (100 mL) was added to the solid. The mixture was stirred at room temperature for 15 minutes, and filtered. The filtrate was concentrated under reduced pressure, and to the residue was added dichloromethane (50 m). The mixture was further stirred for 15 minutes, and filtered. The filtrate was concentrated under reduced pressure to afford compound H-2. 1H NMR (400 MHz, CDCl3) δ 3.76-3.66 (m, 4H), 3.48 (t, J=6.4 Hz, 2H), 2.51-2.46 (m, 2H), 2.46-2.41 (m, 4H), 2.11-1.97 (m, 2H).


The Second Step

Compound 16-4 (200.0 mg, 373.56 μmol) was dissolved in N,N-dimethylformamide (3 mL), and to the mixture was added sodium hydride (60%, 44.8 mg, 1.12 mmol) in one portion at 0° C. The reaction solution was stirred for 0.5 hour, and then compound H-2 (155.5 mg, 747.11 μmol) was added. The reaction solution was stirred at 0° C. for 1.5 hours. The reaction solution was diluted with water (5 mL) and extracted with ethyl acetate (15 mL×3). The organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to afford compound 18-2. LC-MS: m/z=662.5 [M+H]


The Third Step

The hydrochloride salt of compound 18-3 was obtained by referring to the fifth step of Example 16. LC-MS: m/z=562.3 [M+H]


The Fourth Step



embedded image


Compound of example 18 was obtained by referring to the tenth step of Example 1. 1H NMR (400 MHz, CD3OD) δ 8.39 (s, 1H), 7.75 (s, 1H), 7.61-7.59 (m, 1H), 7.49-7.46 (m, 1H), 7.21 (s, 1H), 6.79 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.32 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.78 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 4.27-4.17 (m, 2H), 4.03-3.94 (m, 1H), 3.92-3.83 (m, 2H), 3.77-3.60 (m, 5H), 3.37-3.35 (m, 1H), 3.10 (t, J=5.4 Hz, 2H), 2.65-2.44 (m, 6H), 2.12-1.94 (m, 3H), 1.63 (d, J=10.0 Hz, 1H). LC-MS: m/z=616.4 [M+H]+.


Example 19



embedded image


Synthetic Route:




embedded image


embedded image


The First Step

Compound I-1 (300.0 mg, 2.60 mmol) was added to toluene (5 mL), and to the mixture was added thionyl chloride (309.8 mg, 2.60 mmol). The mixture was stirred at 70° C. for 3 hours. The reaction solution was concentrated under reduced pressure to afford compound I-2. 1H NMR (400 MHz, DMSO-d6) δ 4.16 (dd, J1=12.0 Hz, J2=6.0 Hz, 1H), 4.00 (dd, J1=12.0 Hz, J2=6.0 Hz, 1H), 3.70 (m, 1H), 3.56 (m, 1H), 3.09 (m, 1H), 2.87 (s, 3H), 2.36-2.21 (m, 1H), 2.07-1.72 (m, 4H).


The Second Step

Compound 16-4 (100.0 mg, 186.78 μmol) was added to N,N-dimethylformamide (1.0 mL). To the mixture was added sodium hydride (60%, 29.8 mg, 747.12 μmol) at 0° C. After stirring for 0.2 hour, the reaction solution was warmed to 16° C. Compound I-2 (49.9 mg, 373.56 μmol) was added and stirred at 40° C. for 0.5 hour. The reaction solution was quenched with water (10 mL) and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (10 mL) and saturated brine (10 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative high performance liquid chromatography (basic) to afford compound 19-2. LC-MS: m/z=632.5 [M+H]+.


The Third Step

Compound 19-2 (33.0 mg, 52.17 μmol) was added to a solution of hydrogen chloride in ethyl acetate (2.0 mL). The reaction solution was stirred at 16° C. for 0.5 hour. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 19-3. LC-MS: m/z=532.4 [M+H]+.


The Fourth Step

The hydrochloride salt of compound 19-3 (30.0 mg, 56.34 μmol) was added to a mixed solvent of anhydrous tetrahydrofuran (0.5 mL) and water (0.5 mL), and to the mixture was added sodium bicarbonate (14.2 mg, 169.03 μmol). The reaction solution was stirred at 0° C. for 0.05 hour, and acryloyl chloride (3.8 mg, 42.26 μmol) was added slowly. The reaction solution was stirred at 0° C. for 0.2 hour. The reaction solution was concentrated under reduced pressure, diluted with water (5 mL), and extracted with ethyl acetate (10 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative high performance liquid chromatography (basic) to obtain example 19. 1H NMR (400 MHz, CDCl3) δ 8.71-8.68 (m, 1H), 8.54-8.45 (m, 1H), 7.35 (m, 1H), 7.08-7.00 (m, 1H), 6.66-6.53 (m, 1H), 6.47-6.36 (m, 1H), 5.71 (m, 1H), 4.77-4.68 (m, 1H), 4.13 (m, 3H), 4.02-3.85 (m, 3H), 3.76-3.66 (m, 1H), 3.50 (s, 1H), 3.00 (m, 2H), 2.97 (s, 1H), 2.89 (s, 1H), 2.55 (m, 1H), 2.31 (s, 2H), 2.05 (s, 3H), 1.93 (m, 2H), 1.86 (m, 2H); LC-MS: m/z=586.4 [M+H]+.


Example 20



embedded image


Synthetic Route:




embedded image


embedded image


embedded image


The First Step

Compound J-1 (6.40 g, 28.16 mmol) was dissolved in pyridine (20 mL), and to the mixture was added p-toluenesulfonyl chloride (8.05 g, 42.24 mmol). The mixture was stirred at 10° C. for 16 hours. The reaction solution was concentrated under reduced pressure, diluted with ethyl acetate (300 mL), washed sequentially with 1 N dilute hydrochloric acid (100 mL×2) and saturated brine (100 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:8) to afford compound J-2. 1H NMR (400 MHz, CDCl3) δ 7.77 (d, J=8.4 Hz, 2H), 7.34 (d, J=8.4 Hz, 2H), 5.69 (t, J=4.8 Hz, 1H), 4.05-4.30 (m, 2H), 2.45 (s, 3H), 1.90-2.15 (m, 6H), 1.70-1.90 (m, 2H), 1.43 (s, 9H). LC-MS: m/z=282.0 [M+H-100]+.


The Second Step

Compound K-1 (1.00 g, 10.00 mmol) was dissolved in pyridine (5 mL) and to the mixture was added p-toluenesulfonyl chloride (2.86 g, 14.99 mmol). The reaction solution was stirred at 10° C. for 16 hours, and then concentrated under reduced pressure. The residue was dissolved in ethyl acetate (40 mL), washed successively with saturated ammonium chloride solution (60 mL×3), saturated brine (60 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:8) to afford compound K-2. 1H NMR (400 MHz, CDCl3) δ 7.82 (d, J=8.4 Hz, 2H), 7.38 (d, J=8.0 Hz, 2H), 4.34 (q, J=8.8 Hz, 2H), 2.47 (s, 3H).


The Third Step

Compound 7-1 (4.00 g, 11.29 mmol) was dissolved in N,N-dimethylacetamide (20 mL), and to the mixture were added potassium carbonate (2.34 g, 16.94 mmol) and compound J-2 (5.60 g, 14.68 mmol). The reaction solution was stirred at 75° C. for 16 hours. The reaction solution was cooled to 30° C., diluted with water (60 mL), and extracted with ethyl acetate (60 mL×3). The organic phases were combined, washed successively with aqueous solution (100 mL×3) and saturated brine (60 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:1) to afford compound 20-2. 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 8.49 (t, J=8.4 Hz, 1H), 7.26-7.40 (m, 3H), 7.13 (s, 1H), 4.60-4.75 (m, 1H), 4.15-4.40 (m, 2H), 3.94 (s, 3H), 1.75-2.20 (m, 6H), 1.55-1.70 (m, 2H), 1.43 (s, 9H). LC-MS: m/z=563.1 [M+H]+.


The Fourth Step

A mixture of compound 20-2 (2.70 g, 4.80 mmol) and pyridine hydrochloride (10.80 g, 93.46 mmol) was heated to 170° C. under nitrogen protection, and stirred for 3 hours to afford compound 20-3. LC-MS: m/z=449.0 [M+H]+.


The Fifth Step

Compound 20-3 (13.00 g, 4.63 mmol) and triethylamine (7.00 g, 69.18 mmol) were dissolved in methanol (20 mL), and to the mixture was added di-tert-butyl dicarbonate (9.63 g, 44.12 mmol). The reaction solution was stirred at 10° C. for 1 hour. The reaction solution was concentrated under reduced pressure, and the residue was slurried with water (20 mL) and filtered. The obtained solid was washed successively with saturated ammonium chloride solution (10 mL×3) and petroleum ether (10 mL), and dried to afford compound 20-4. LC-MS: m/z=649.1 [M+H]+.


The Sixth Step

Compound 20-4 (3.00 g, 4.62 mmol) was dissolved in methanol (50 mL), and to the mixture was added potassium carbonate (3.19 g, 23.09 mmol). The reaction solution was stirred at 40° C. for 14 hours, and then water (30 mL) was added. The mixture was adjusted to pH of 6 with acetic acid and filtered. The solid was washed with water (30 mL) and dried in the air to afford compound 20-5. 1H NMR (400 MHz, DMSO-d6) δ 9.54 (brs, 1H), 8.22 (s, 1H), 7.68 (s, 1H), 7.61 (t, J=8.4 Hz, 1H), 7.50-7.60 (m, 1H), 6.94 (s, 1H), 4.90-5.00 (m, 1H), 4.10-4.20 (m, 2H), 2.10-2.20 (m, 2H), 1.70-2.00 (m, 4H), 1.60-1.70 (m, 2H), 1.44 (s, 9H). LC-MS: m/z=549.1 [M+H]+.


The Seventh Step

Compound 20-5 (70.0 mg, 127.41 μmol) was dissolved in N,N-dimethylformamide (3 mL), and to the mixture were added potassium carbonate (35.2 mg, 254.81 μmol) and compound K-2 (64.78 mg, 254.81 μmol). The mixture was stirred at 70° C. for 16 hours. The reaction solution was cooled to 30° C., and water (10 mL) was added. The mixture was extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (30 mL×2) and saturated brine (30 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:1) to afford compound 20-6. 1H NMR (400 MHz, CDCl3) δ 9.77 (s, 1H), 8.43 (s, 1H), 7.97 (s, 1H), 7.55-7.65 (m, 2H), 7.43 (s, 1H), 4.95-5.05 (m, 2H), 4.75-4.90 (m, 1H), 4.10-4.20 (m, 2H), 2.10-2.20 (m, 2H), 1.90-2.20 (m, 2H), 1.70-1.80 (m, 2H), 1.55-1.70 (m, 2H), 1.44 (s, 9H). LC-MS: m/z=631.1 [M+H]+.


The Eighth Step

Compound 20-6 (35.0 mg, 55.43 μmol) was dissolved in a solution of hydrogen chloride in dioxane (4 N, 5 mL). The reaction solution was stirred at 10° C. for 0.3 hour. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 20-7. LC-MS: m/z=531.0 [M+H]+.


The Ninth Step

The hydrochloride salt of compound 20-7 (30.0 mg, 52.84 μmol) was dissolved in a mixed solvent of tetrahydrofuran (3 mL) and water (3 mL) at 0° C. To the mixture was added sodium bicarbonate (13.3 mg, 158.51 μmol), and added slowly acryloyl chloride (3.8 mg, 42.27 μmol) dropwise. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding methanol (1 mL) and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain example 20. 1H NMR (400 MHz, DMSO-d6) δ 9.78 (s, 1H), 8.42 (s, 1H), 7.93 (s, 1H), 7.60-7.70 (m, 2H), 7.43 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.19 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.70 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 4.95-5.05 (m, 2H), 4.85-4.95 (m, 1H), 4.55-4.65 (m, 2H), 2.20-2.35 (m, 2H), 1.80-2.10 (m, 4H), 1.60-1.75 (m, 1H), 1.50-1.60 (m, 1H). LC-MS: m/z=585.1 [M+H]+.


Example 21

Synthetic Route:




embedded image




embedded image


The First Step

Compound 20-5 (150.0 mg, 273.0 μmol) was added to N,N-dimethylformamide (3 mL), and to the mixture was added sodium hydride (60%, 32.8 mg, 819.1 μmol) under nitrogen protection. After stirring at 0° C. for 30 minutes, bromoethane (74.4 mg, 682.54 μmol) was added. The mixture was warmed to 15° C. and further stirred for 30 minutes. The reaction solution was quenched by adding water (5 mL) and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (10 mL×3) and saturated brine (10 mL×1), dried over sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:0) to afford compound 21-2. 1H NMR (400 MHz, DMSO-d6) δ 9.62 (br s, 1H), 8.39 (s, 1H), 7.82 (s, 1H), 7.68-7.55 (m, 1H), 7.21 (s, 1H), 4.82 (m, 1H), 4.25-4.19 (m, 2H), 4.18-4.13 (m, 2H), 2.19 (m, 2H), 1.92 (m, 2H), 1.79 (m, 2H), 1.65 (m, 2H), 1.45 (s, 9H), 1.43-1.41 (m, 3H). LC-MS: m/z=577.3 [M+H]+.


The Second Step

Compound 21-2 (50.0 mg, 86.6 μmol) was added to ethyl acetate (0.5 mL), and to the mixture was added a solution of hydrogen chloride in ethyl acetate (4 N, 1.7 mL) at room temperature. The mixture was stirred at 15° C. for 10 minutes. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 21-3. LC-MS: m/z=477.1 [M+H]+.


The Third Step

Compound of example 21 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, DMSO-d6) δ 9.65 (s, 1H), 8.39 (s, 1H), 7.80 (s, 1H), 7.67-7.55 (m, 2H), 7.21 (s, 1H), 6.78 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 5.00-4.85 (m, 1H), 4.62 (m, 2H), 4.20 (q, J=6.8 Hz, 2H), 3.17 (d, J=5.2 Hz, 3H), 2.39-2.30 (m, 1H), 2.25 (m, 1H), 2.06-1.93 (m, 2H), 1.89 (m, 2H), 1.67 (m, 1H), 1.52 (m, 1H). LC-MS: m/z=531.2 [M+H]+.


Example 22



embedded image


Synthetic Route:




embedded image


The First Step

Compound 22-2 was obtained by referring to the first step of Example 21. 1H NMR (400 MHz, DMSO-d6) δ 9.63 (s, 1H), 8.40 (s, 1H), 7.86 (s, 1H), 7.63-7.57 (m, 2H), 7.26 (s, 1H), 4.92-4.74 (m, 1H), 4.34-4.26 (m, 2H), 4.19-4.14 (m, 2H), 3.79-3.72 (m, 2H), 3.36 (s, 3H), 2.18 (m, 2H), 1.96-1.86 (m, 3H), 1.63 (m, 3H), 1.44 (s, 9H). LC-MS: m/z=607.3 [M+H]+.


The Second Step

The hydrochloride salt of compound 22-3 was obtained by referring to the first step of example 21. LC-MS: m/z=507.1 [M+H]+.


The Third Step

Compound of example 22 was obtained by referring to the ninth step of Example 20. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.40 (s, 1H), 7.85 (s, 1H), 7.69-7.55 (m, 2H), 7.26 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.80-5.68 (m, 1H), 5.00-4.84 (m, 1H), 4.61 (m, 2H), 4.34-4.23 (m, 2H), 3.79-3.70 (m, 2H), 3.35 (s, 3H), 2.36-2.15 (m, 2H), 2.04-1.82 (m, 4H), 1.68 (m, 1H), 1.52 (m, 1H). LC-MS: m/z=561.3 [M+H]+.


Example 23



embedded image


Synthetic Route:




embedded image


The First Step

Compound 20-5 (200.0 mg, 364.02 μmol) was added to N,N-dimethylformamide (5 mL) at room temperature, and then to the mixture were added cesium carbonate (474.4 mg, 1.46 mmol), 2-chloro-N-methylacetamide (47.0 mg, 436.82 μmol) and potassium iodide (60.4 mg, 364.02 μmol). The mixture was heated to 50° C. and stirred for 3 hours. The reaction solution was cooled to room temperature, quenched with water (10 mL), and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (5 mL×3) and saturated brine (5 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=20:1) to afford compound 23-2. LC-MS: m/z=620.3 [M+H]+.


The Second Step

The hydrochloride salt of compound 23-3 was obtained by referring to the first step of example 21. LC-MS: m/z=520.2 [M+H]+.


The Third Step

Compound of example 22 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, MeOD-d4) δ 8.38 (s, 1H), 7.82 (s, 1H), 7.61 (t, J=8.4 Hz, 1H), 7.47 (dd, J1=8.8 Hz, J2=1.6 Hz, 1H), 7.19 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.36 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.82 (dd, J1=10.8 Hz, J2=1.6 Hz, 1H), 5.22-5.09 (m, 1H), 4.79 (m, 1H), 4.73 (s, 2H), 4.69-4.64 (m, 1H), 2.85 (s, 3H), 2.53-2.31 (m, 2H), 2.20-2.08 (m, 2H), 2.07-2.01 (m, 2H), 1.96-1.86 (m, 1H), 1.80 (m, 1H). LC-MS: m/z=574.1 [M+H]+.


Example 24



embedded image


Synthetic Route:




embedded image


The First Step

Compound 24-2 was obtained by referring to the first step of Example 23. LC-MS: m/z=676.3 [M+H]+.


The Second Step

The hydrochloride salt of compound 24-3 was obtained by referring to the first step of Example 21. LC-MS: m/z=576.1 [M+H]+.


The Third Step

Compound of example 24 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, MeOD-d4) δ 8.38 (s, 1H), 7.79 (s, 1H), 7.62 (t, J=8.4 Hz, 1H), 7.46 (dd, J1=8.8 Hz, J2=1.6 Hz, 1H), 7.22 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.36 (dd, J1=16.4 Hz, 1.6 Hz, 1H), 5.82 (dd, J1=10.4 Hz, J2=1.6 Hz, 1H), 5.13-4.98 (m, 1H), 4.80-4.78 (m, 1H), 4.68-4.61 (m, 1H), 4.24 (t, J=6.0 Hz, 2H), 3.77-3.67 (m, 4H), 2.68-2.58 (m, 2H), 2.53 (m, 4H), 2.42 (m, 1H), 2.37-2.30 (m, 1H), 2.20-1.99 (m, 6H), 1.91-1.82 (m, 1H), 1.74 (m, 1H). LC-MS: m/z=630.2 [M+H]+.


Example 25



embedded image


Synthetic Route:




embedded image


The First Step

Compound 20-5 (150.0 mg, 273.02 μmol) was added to N,N-dimethylformamide (5 mL), and then to the mixture were added potassium fluoride (47.6 mg, 819.05 μmol) and ethylene carbonate (72.1 mg, 819.05 μmol). The reaction solution was heated to 110° C. and stirred for 3 hours. The reaction solution was cooled to room temperature, quenched with water (20 mL), and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (5 mL×3) and saturated brine (5 mL), dried over sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=20:1) to afford compound 25-2. LC-MS: m/z=593.2 [M+H]+.


The Second Step

The hydrochloride salt of compound 25-3 was obtained by referring to the first step of example 21. LC-MS: m/z=493.0 [M+H]+.


The Third Step

Compound of example 25 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, MeOD-d4) δ 8.27 (s, 1H), 7.69 (s, 1H), 7.50 (t, J=8.0 Hz, 1H), 7.35 (br d, J=8.8 Hz, 1H), 7.12 (s, 1H), 6.65 (dd, J1=16.8 Hz, J2=10.8 Hz, 1H), 6.24 (dd, J1=16.8 Hz, J2=1.6 Hz, 1H), 5.70 (dd, J1=10.4 Hz, J2=1.6 Hz, 1H), 5.03-4.90 (m, 1H), 4.66 (m, 1H), 4.52 (m, 1H), 4.14 (m, 2H), 3.94-3.83 (m, 2H), 2.38-2.27 (m, 1H), 2.23 (m, 1H), 2.07-1.93 (m, 2H), 1.89 (m, 2H), 1.77 (m, 1H), 1.66 (m, 1H). LC-MS: m/z=547.1 [M+H]+.


Example 26



embedded image


Synthetic Route:




embedded image


The First Step

Compound 20-5 (200.0 mg, 364.02 umol) was added to N,N-dimethylformamide (5 mL) at room temperature, and then to the mixture were added cesium carbonate (55.8 mg, 1.09 mmol) and compound L-1 (97.0 mg, 400.42 μmol). The reaction solution was heated to 100° C., and stirred for 3 hours. The reaction solution was cooled to room temperature, quenched with water (10 mL), and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (5 mL×3) and saturated brine (5 mL), dried over sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=20:1) to afford compound 26-2. LC-MS: m/z=619.2 [M+H]+.


The Second Step

The hydrochloride salt of compound 26-3 was obtained by referring to the first step of Example 21. LC-MS: m/z=519.0 [M+H]+.


The Third Step

Compound of example 26 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, MeOD-d4) δ 8.39 (s, 1H), 7.81 (s, 1H), 7.62 (t, J=8.4 Hz, 1H), 7.45 (dd, J1=9.2 Hz, J2=2.0 Hz, 1H), 7.17 (s, 1H), 6.76 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.36 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.81 (dd, J1=10.4 Hz, J2=1.6 Hz, 1H), 5.22 (m, 1H), 5.07-4.94 (m, 1H), 4.80-4.73 (m, 1H), 4.67-4.60 (m, 1H), 4.05 (s, 2H), 4.04-3.98 (m, 1H), 3.93 (m, 1H), 2.46-2.35 (m, 2H), 2.32 (m, 1H), 2.26-2.18 (m, 1H), 2.17-2.09 (m, 1H), 2.06-1.93 (m, 3H), 1.92-1.82 (m, 1H), 1.75 (m, 1H); LC-MS: m/z=573.3 [M+H]+.


Example 27



embedded image


Synthetic Route:




embedded image


The First Step

Compound 27-2 was obtained by referring to the first step of example 26. LC-MS: m/z=619.2 [M+H]+.


The Second Step

The hydrochloride salt of compound 27-3 was obtained by referring to the first step of example 21. LC-MS: m/z=519.0 [M+H]+.


The Third Step

Compound of example 27 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, MeOD-d4) δ 8.39 (s, 1H), 7.82 (s, 1H), 7.62 (t, J=8.4 Hz, 1H), 7.46 (dd, J1=8.8 Hz, J2=1.6 Hz, 1H), 7.19 (s, 1H), 6.76 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.36 (dd, J1=16.8 Hz, J2=1.6 Hz, 1H), 5.81 (dd, J1=10.4 Hz, J2=1.6 Hz, 1H), 5.23 (m, 1H), 5.08-4.94 (m, 1H), 4.78 (m, 1H), 4.68-4.61 (m, 1H), 4.06 (s, 2H), 4.04-3.99 (m, 1H), 3.93 (m, 1H), 2.46-2.36 (m, 2H), 2.32 (m, 1H), 2.26-2.19 (m, 1H), 2.18-2.10 (m, 1H), 2.06-1.94 (m, 3H), 1.87 (m, 1H), 1.76 (m, 1H). LC-MS: m/z=573.3 [M+H]+.


Example 28



embedded image


Synthetic Route:




embedded image


The First Step

Compound 8-2 (83.6 mg, 647.49 μmol) was dissolved in N,N-dimethylformamide (1 mL), and to the mixture was added HATU (123.1 mg, 323.74 μmol). The reaction solution was stirred at 16° C. for 0.5 hour, and DIEA (69.7 mg, 539.57 μmol) and compound N-1 (100.0 mg, 215.83 μmol) were added. The mixture was further stirred for 16 hours. The reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was isolated by preparative high-performance liquid chromatography (neutral) to obtain the compound of example 28. 1H NMR (400 MHz, MeOD-d4) δ 8.37 (s, 1H), 7.76 (s, 1H), 7.60 (t, J=8.3 Hz, 1H), 7.45 (dd, J1=8.8 Hz, J2=1.6 Hz, 1H), 7.20 (s, 1H), 6.90 (m, 1H), 6.60 (d, J=15.2 Hz, 1H), 5.11-5.01 (m, 1H), 4.75 (m, 1H), 4.63 (m, 1H), 3.99 (s, 3H), 3.19 (d, J=6.0 Hz, 2H), 2.44-2.37 (m, 1H), 2.33 (m, 1H), 2.30 (s, 6H), 2.19-2.10 (m, 1H), 2.09-1.96 (m, 3H), 1.85 (m, 1H), 1.75 (m, 1H). LC-MS: m/z=574.4 [M+H]+.


Example 29



embedded image


Synthetic Route:




embedded image


The First Step

Compound 13-2_(250.0 mg, 470.56 μmol) was dissolved in N,N-dimethylformamide (4 mL). To the reaction solution were added potassium fluoride (65.6 mg, 1.13 mmol), potassium carbonate (130.07 mg, 941.11 μmol) and compound L-1 (171.0 mg, 705.83 μmol). The reaction solution was stirred at 118° C. for 18 hours. The reaction solution was concentrated under reduced pressure to remove the solvent, and the residue was isolated by preparative thin-layer chromatography (ethyl acetate) to afford compound 29-2. LC-MS: m/z=601.3 [M+H]+.


The Second Step

Compound 29-2 (147.0 mg, 244.44 μmol) was dissolved in methanol (10 mL), and to the mixture was added potassium carbonate (168.9 mg, 1.22 mmol). The reaction solution was stirred at 50° C. for 5 hours. The reaction solution was concentrated under reduced pressure, and the residue was dissolved in methanol (5 mL). Potassium carbonate (33.8 mg, 244.44 μmol) was added, and the reaction solution was stirred at 50° C. for 5 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to afford compound 29-3. LC-MS: m/z=505.3 [M+H]+.


The Third Step

Compound of example 29 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, CDCl3) δ 8.68 (s, 1H), 8.39 (t, J=8.4 Hz, 1H), 7.33 (dd, J=8.8, 1.6 Hz, 1H), 7.19 (d, J=5.2 Hz, 1H), 7.16 (s, 1H), 6.61 (dd, J=16.8, 10.4 Hz, 1H), 6.42 (dt, J=16.4, 2.4 Hz, 1H), 5.72 (dd, J=10.4, 2.0 Hz, 1H), 5.07-5.02 (m, 1H), 4.70-4.65 (m, 1H), 4.09-4.03 (m, 1H), 4.02-3.95 (m, 3H), 3.94-3.83 (m, 3H), 3.70-3.65 (m, 1H), 3.00-2.97 (m, 2H), 2.36-2.23 (m, 1H), 2.18-2.15 (m, 1H), 1.92-1.87 (m, 2H); LC-MS: m/z=559.3 [M+H]+.


Example 30



embedded image


Synthetic Route:




embedded image


The First Step

Compound 0-1 (2.00 g, 22.44 mmol) was dissolved in pyridine (5 mL), and to the mixture was added p-toluenesulfonyl chloride (4.28 g, 22.44 mmol). The reaction solution was stirred at 10° C. for 16 hours. Ethyl acetate (50 mL) was added to the reaction solution and the mixture was filtered. The filtrate was concentrated to afford compound 0-2. 1H NMR (400 MHz, CDCl3) δ 7.49 (d, J=7.6 Hz, 2H), 7.12 (d, J=7.6 Hz, 2H), 3.98 (t, J=6.4 Hz, 2H), 3.49 (t, J=6.4 Hz, 2H), 2.76 (s, 3H), 2.30 (s, 6H). LC-MS: m/z=244.0 [M+H]+.


The Second Step

Compound 20-5 (200.0 mg, 364.02 μmol) was dissolved in N,N-dimethylformamide (3 mL), and to the mixture were added potassium carbonate (100.6 mg, 728.04 μmol) and compound 0-2 (177.2 mg, 728.04 μmol). The mixture was stirred at 70° C. for 16 hours. The reaction solution was cooled to 30° C., diluted with water (10 mL), and extracted with ethyl acetate (10 mL×3). The organic phases were combined, washed sequentially with water (30 mL×2) and saturated brine (30 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to afford compound 30-2. 1H NMR (400 MHz, CDCl3) δ 8.70 (s, 1H), 8.53 (t, J=8.4 Hz, 1H), 7.20-7.40 (m, 4H), 4.65-4.85 (m, 1H), 4.20-4.45 (m, 4H), 2.90-3.00 (m, 2H), 2.44 (s, 6H), 2.10-2.20 (m, 2H), 1.95-2.10 (m, 2H), 1.75-1.95 (m, 2H), 1.60-1.75 (m, 2H), 1.50 (s, 9H). LC-MS: m/z=620.1 [M+H]+.


The Third Step

The hydrochloride salt of compound 30-3 was obtained by referring to the eighth step of Example 20. LC-MS: m/z=520.1 [M+H]+.


The Fourth Step

Compound of example 30 was obtained by referring to the ninth step of Example 20. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.40 (s, 1H), 7.86 (s, 1H), 7.55-7.70 (m, 2H), 7.26 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (d, J=16.8 Hz 1H), 5.72 (d, J=10.4 Hz 1H), 4.90-5.00 (m, 1H), 4.55-4.60 (m, 2H), 4.20-4.30 (m, 2H), 2.70-2.80 (m, 2H), 2.31 (s, 6H), 1.80-2.10 (m, 6H), 1.60-1.70 (m, 1H), 1.45-1.60 (m, 1H). LC-MS: m/z=574.1 [M+H]+.


Example 31



embedded image


Synthetic Route:




embedded image


The First Step

Compound 31-1 (300.0 mg, 1.28 mmol) was dissolved in thionyl chloride (3.0 mL, 41.45 mmol), and to the mixture was added N,N-dimethylformamide (47.30 μL, 614.84 μmol). The reaction solution was heated to 80° C. and stirred for 1 hour. The reaction solution was concentrated under reduced pressure, and to the residue was added a solution of compound P-1 (171.8 mg, 1.28 mmol) in isopropanol (10 mL) at room temperature. The mixture was heated to 90° C. and stirred for 1 hour. The reaction solution was cooled to 20° C. and filtered. The solid was washed with isopropanol (10 mL) and dried in the air to afford compound 31-2. 1H NMR (400 MHz, DMSO-d6) δ 11.51 (s, 1H), 9.35 (s, 1H), 8.90 (s, 1H), 8.72 (s, 1H), 8.23 (s, 1H), 7.85-8.00 (m, 2H), 7.50 (s, 1H), 4.03 (s, 3H), 2.41 (s, 3H). LC-MS: m/z=351.0 [M+H]+.


The Second Step

Compound 31-2 (400.0 mg, 1.14 mmol) was dissolved in ethanol (10 mL), and to the mixture was added a solution of sodium hydroxide (228.4 mg, 5.71 mmol) in water (2 mL). The reaction solution was stirred at 10° C. for 1 hour. To the reaction solution was added acetic acid (0.5 mL) and water (10 mL). The reaction solution was further stirred for 0.5 hour, and then filtered. The solid was washed with water (10 mL) and dried under reduced pressure to afford compound 31-3. 1H NMR (400 MHz, DMSO-d6) δ 10.89 (s, 1H), 9.67 (s, 1H), 9.38 (s, 1H), 8.42 (s, 1H), 8.08 (d, J=2.8 Hz, 1H), 7.80-7.90 (m, 1H), 7.74 (s, 1H), 7.19 (s, 1H), 7.03 (d, J=8.8 Hz, 1H), 3.97 (s, 3H). LC-MS: m/z=309.0 [M+H]+.


The Third Step

Compound 31-3 (100.0 mg, 324.37 μmol) was dissolved in N,N-dimethylacetamide (5 mL), and to the mixture were added potassium carbonate (89.7 mg, 648.74 μmol) and compound P-2 (1034 mg, 308.15 μmol). The reaction solution was stirred at 70° C. for 16 hours. The reaction solution was cooled to 20° C., diluted with water (30 mL), and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed sequentially with water (30 mL×3) and saturated brine (30 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=15:1) to obtain example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 9.46 (s, 1H), 8.44 (s, 1H), 8.23 (d, J=2.4 Hz 1H), 8.00-8.10 (m, 1H), 7.76 (s, 1H), 7.50 (d, J=9.6 Hz 1H), 6.77 (dd, JJ=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.90-5.10 (m, 1H), 4.55-4.65 (m, 2H), 3.98 (s, 3H), 2.15-2.35 (m, 2H), 1.80-2.00 (m, 4H), 1.60-1.70 (m, 1H), 1.45-1.55 (m, 1H). LC-MS: m/z=472.1 [M+H]+.


Example 32



embedded image


Synthetic Route:




embedded image


The First Step

Compound 32-2 was obtained by referring to the first step of Example 31. 1H NMR (400 MHz, DMSO-d6) δ 11.21 (s, 1H), 8.97 (s, 1H), 8.71 (s, 1H), 8.18 (d, J=2.0 Hz, 1H), 7.71-7.81 (m, 2H), 7.49 (s, 1H), 4.02 (s, 3H), 2.40 (s, 3H). LC-MS: m/z=378.0 [M+H]+.


The Second Step

Compound 32-3 was obtained by referring to the second step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.77 (s, 1H), 9.61 (s, 1H), 8.54 (s, 1H), 8.34 (d, J=2.4 Hz, 1H), 7.80-7.90 (m, 1H), 7.81 (s, 1H), 7.61 (d, J=8.8 Hz, 1H), 7.24 (s, 1H), 3.99 (s, 3H). LC-MS: m/z=335.9 [M+H]+.


The Third Step

Compound of example 32 was obtained by referring to the third step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.62 (s, 1H), 8.58 (s, 1H), 8.32 (d, J=2.8 Hz 1H), 7.85-7.95 (m, 2H), 7.68 (d, J=8.8 Hz 1H), 7.24 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.4 Hz, 1H), 5.72 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.90-5.10 (m, 1H), 4.60-4.65 (m, 2H), 3.92 (s, 3H), 2.20-2.40 (m, 2H), 1.80-2.10 (m, 4H), 1.60-1.70 (m, 1H), 1.40-1.50 (m, 1H). LC-MS: m/z=499.1 [M+H]+.


Example 33



embedded image


Synthetic Route:




embedded image


The First Step

Compound 33-2 was obtained by referring to the first step of example 31. LC-MS: m/z=362.0 [M+H]+.


The Second Step

Compound 33-3 was obtained by referring to the second step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.76 (s, 1H), 9.50 (s, 1H), 8.35 (s, 1H), 7.66 (s, 1H), 7.42-7.60 (m, 2H), 7.20-7.30 (m, 1H), 7.21 (s, 1H), 3.98 (s, 3H). LC-MS: m/z=319.9 [M+H]+.


The Third Step

Compound of example 33 was obtained by referring to the third step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.57 (s, 1H), 8.38 (s, 1H), 7.78 (s, 1H), 7.50-7.65 (m, 2H), 7.25-7.35 (m, 1H), 7.21 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.77 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 4.90-5.10 (m, 1H), 4.55-4.65 (m, 2H), 3.98 (s, 3H), 2.20-2.40 (m, 2H), 1.85-2.10 (m, 4H), 1.60-1.70 (m, 1H), 1.45-1.55 (m, 1H). LC-MS: m/z=483.0 [M+H]+.


Example 34



embedded image


Synthetic Route:




embedded image


The First Step

Compound 34-2 was obtained by referring to the first step of example 31. LC-MS: m/z=380.0 [M+H]+.


The Second Step

Compound 34-3 was obtained by referring to the second step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.76 (s, 1H), 9.49 (s, 1H), 8.33 (s, 1H), 7.65 (s, 1H), 7.50-7.60 (m, 1H), 7.32-7.40 (m, 1H), 7.21 (s, 1H), 3.98 (s, 3H). LC-MS: m/z=337.9 [M+H]+.


The Third Step

Compound of example 34 was obtained by referring to the third step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.60 (s, 1H), 8.37 (s, 1H), 7.78 (s, 1H), 7.56-7.65 (m, 1H), 7.38-7.50 (m, 1H), 7.21 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.72 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 4.90-5.05 (m, 1H), 4.55-4.65 (m, 2H), 3.98 (s, 3H), 2.20-2.40 (m, 2H), 1.85-2.10 (m, 4H), 1.60-1.70 (m, 1H), 1.45-1.55 (m, 1H). LC-MS: m/z=501.1 [M+H]+.


Example 35



embedded image


Synthetic Route:




embedded image


The First Step

Compound 35-2 was obtained by referring to the first step of example 31. LC-MS: m/z=350.0 [M+H]+.


The Second Step

Compound 35-3 was obtained by referring to the second step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.74 (s, 1H), 9.51 (s, 1H), 9.37 (s, 1H), 8.49 (s, 1H), 8.07 (s, 1H), 7.91 (s, 1H), 7.60-7.70 (m, 3H), 7.22 (s, 1H), 3.98 (s, 3H). LC-MS: m/z=307.9 [M+H]+.


The Third Step

Compound of example 35 was obtained by referring to the third step of example 31. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 9.27 (s, 1H), 8.49 (s, 1H), 8.00-8.10 (m, 2H), 7.55-7.70 (m, 3H), 7.21 (s, 1H), 6.77 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.72 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 5.10-5.15 (m, 1H), 4.55-4.65 (m, 2H), 3.98 (s, 3H), 2.20-2.40 (m, 2H), 1.80-2.10 (m, 4H), 1.60-1.70 (m, 1H), 1.40-1.50 (m, 1H). LC-MS: m/z=471.2 [M+H]+.


Example 36



embedded image


Synthetic Route:




embedded image


The First Step

Compound 16-4 (100.0 mg, 470.56 μmol) was dissolved in N,N-dimethylformamide (2 mL), and to the reaction solution was added NaH (60%, 22.4 mg, 560.34 μmol). The reaction solution was stirred at 0° C. for 0.5 hour, and a solution of compound 1-(2-bromoethyl)piperidine (55.1 mg, 373.56 μmol) in N,N-dimethylformamide (0.5 mL) was added. The reaction solution was stirred at 15° C. for 5 hours, and then diluted with water (10 mL), and extracted with ethyl acetate (5 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=7:1) to afford compound 36-2.


The Second Step

The hydrochloride salt of compound 36-3 was obtained by referring to the second step of example 21.


The Third Step

Compound of example 36 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, CDCl3) δ 8.70 (s, 1H), 8.49 (t, J=8.4 Hz, 1H), 7.34 (d, J=8.8 Hz, 1H), 7.27-7.25 (m, 1H), 7.07 (s, 1H), 6.66-6.57 (m, 1H), 6.43 (d, J=16.8 Hz, 1H), 5.73 (d, J=10.0 Hz, 1H), 4.69-4.67 (m, 1H), 4.28-4.28 (m, 2H), 3.99-3.94 (m, 2H), 3.90-3.87 (m, 1H), 3.74-3.71 (m, 1H), 3.01-3.00 (m, 2H), 2.89 (m, 2H), 2.59 (m, 4H), 2.05-2.01 (m, 2H), 1.65-1.61 (m, 4H), 1.48-1.46 (m, 2H); LC-MS: m/z=600.3 [M+H]+.


Example 37



embedded image


Synthetic Route:




embedded image


The First Step

Compound 13-2 (250.0 mg, 470.56 μmol) was dissolved in N,N-dimethylformamide (4 mL), and to the reaction solution were added potassium carbonate (130.0 mg, 941.11 μmol) and compound M-1 (171.0 mg, 705.83 μmol). The reaction solution was stirred at 110° C. for 18 hours. The solvent was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=15:1) to afford compound 37-2. LC-MS: m/z=601.3 [M+H]+.


The Second Step

Compound 37-2 (100.0 mg, 166.29 μmol) was dissolved in methanol (5 mL), and to the mixture was added potassium carbonate (68.9 mg, 498.86 μmol). The reaction solution was stirred at 40-50° C. for 3 hours, and then further stirred at 60° C. for 12 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to afford compound 37-3. LC-MS: m/z=505.2 [M+H]+.


The Third Step

Compound of example 37 was obtained by referring to the ninth step of example 20. 1H NMR (400 MHz, CDCl3) δ 8.40 (s, 1H), 7.79 (s, 1H), 7.65-7.58 (m, 1H), 7.47 (d, J=10.8 Hz, 1H), 7.18 (s, 1H), 6.84-6.73 (m, 1H), 6.36-6.29 (m, 1H), 5.82-5.75 (m, 1H), 5.19 (s, 1H), 4.09-4.03 (m, 1H), 3.99-3.87 (m, 5H), 3.84-3.78 (m, 1H), 3.60-3.75 (m, 1H), 3.37 (s, 1H), 3.17-3.03 (m, 2H), 2.39-2.26 (m, 1H), 2.16 (s, 1H), 2.04-1.93 (s, 1H), 1.61 (d, J=10.4 Hz, 1H). LC-MS: m/z=559.3 [M+H]+.


Example 38

Synthetic Route:




embedded image




embedded image


embedded image


The First Step

Compound 38-1 (900.0 mg, 3.93 mmol) was dissolved in N,N-dimethylformamide (20 mL), and to the mixture were added potassium carbonate (1.09 g, 7.85 mmol) and compound Q-1 (1.60 g, 7.07 mmol). The reaction solution was stirred at 75° C. for 16 hours. The reaction solution was cooled to 20° C., diluted with water (30 mL), and extracted with ethyl acetate (30 mL×2). The organic phases were combined, washed sequentially with water (40 mL×2) and saturated brine (40 mL×2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=1:3) to afford compound 38-2. 1H NMR (400 MHz, CDCl3) δ 7.42 (s, 1H), 6.45 (s, 1H), 4.75 (d, J=8.4 Hz, 1H), 4.10-4.40 (m, 2H), 3.87 (s, 3H), 3.86 (s, 3H), 3.75-3.80 (m, 1H), 1.90-2.10 (m, 4H), 1.50-1.70 (m, 4H), 1.41 (s, 9H). LC-MS: m/z=458.1 [M+Na]+.


The Second Step

Compound 38-2 (1.50 g, 3.44 mmol) was dissolved in methanol (30 mL), and to the mixture was added wet palladium/carbon (10%, 50.0 mg). The reaction solution was stirred at 20° C. under hydrogen atmosphere for 0.5 hour. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure and dried to afford compound 38-3. 1H NMR (400 MHz, CDCl3) δ 7.09 (s, 1H), 6.10 (s, 1H), 5.36 (brs, 2H), 4.15-4.40 (m, 2H), 3.85 (s, 3H), 3.82 (s, 3H), 3.65-3.80 (m, 1H), 1.95-2.15 (m, 4H), 1.90-1.85 (m, 2H), 1.47 (s, 9H), 1.30-1.45 (m, 2H). LC-MS: m/z=406.2 [M+H]+.


The Third Step

Compound 38-3 (1.30 g, 3.21 mmol) and ammonium acetate (2.47 g, 32.06 mmol) were added to trimethyl orthoformate (31.29 g, 294.89 mmol). The reaction solution was stirred at 70° C. for 10 hours. The reaction solution was cooled to 20° C., and concentrated under reduced pressure. The residue was slurried with water (10 mL) and filtered. The solid was dried to afford compound 38-4. 1H NMR (400 MHz, DMSO-d6) δ 7.83 (s, 1H), 7.08 (s, 1H), 7.00 (s, 1H), 5.17 (d, J=8.4 Hz, 1H), 4.10-4.20 (m, 2H), 3.95-4.00 (m, 1H), 3.92 (s, 3H), 1.90-2.00 (m, 4H), 1.75-1.90 (m, 2H), 1.50-1.60 (m, 2H), 1.43 (s, 9H). LC-MS: m/z=401.1 [M+H]+.


The Fourth Step

Compound 38-4 (1.20 g, 3.00 mmol) was added to a solution of hydrogen chloride in dioxane (4 N, 10 mL). The reaction solution was stirred at 0° C. for 0.4 hour. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 38-5. LC-MS: m/z=301.0 [M+H]+.


The Fifth Step

The hydrochloride salt of 38-5 (1.10 g, 3.27 mmol) and triethylamine (4.6 mL, 32.66 mmol) were dissolved in dichloromethane (30 mL) at 0° C., and to the mixture was added trifluoroacetic anhydride (1.37 g, 6.53 mmol) dropwise. The reaction solution was stirred at 20° C. for 1 hour. The reaction solution was concentrated under reduced pressure, and the residue was slurried with water (40 mL) and filtered. The solid was dried to afford compound 38-6. 1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 7.80-7.90 (m, 1H), 7.13 (m, 1H), 7.00 (m, 1H), 5.28 (d, J=9.2 Hz, 1H), 4.60-4.70 (m, 1H), 4.40-4.50 (m, 1H), 4.00-4.10 (m, 1H), 3.91 (s, 3H), 1.90-2.20 (m, 6H), 1.55-1.75 (m, 2H). LC-MS: m/z=397.0 [M+H]+.


The Sixth Step

Compound 38-6 (500.0 mg, 1.26 mmol) was dissolved in phosphorus oxychloride (24.1 mL, 260.22 mmol), and to the mixture was added N,N-dimethylformamide (92.2 mg, 1.26 mmol). The reaction solution was stirred at 100° C. for 5 hours. The reaction solution was concentrated under reduced pressure, and to the residue was added a solution of 3,4-dichloro-2-fluoroaniline (340.6 mg, 1.89 mmol) in isopropanol (5 mL). The reaction solution was stirred at 90° C. for 1 hour, and then cooled to 20° C. The reaction solution was quenched by adding saturated sodium bicarbonate aqueous solution (10 mL), and extracted with ethyl acetate (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate:petroleum ether=2:1) to afford compound 38-7. 1H NMR (400 MHz, CDCl3) δ 8.50-8.65 (m, 2H), 7.40-7.50 (m, 1H), 7.15-7.25 (m, 2H), 6.59 (s, 1H), 4.75-4.85 (m, 1H), 4.55-4.65 (m, 2H), 4.05-4.10 (m, 1H), 3.98 (s, 3H), 2.10-2.40 (m, 4H), 1.90-2.00 (m, 2H), 1.60-1.75 (m, 2H). LC-MS: m/z=557.9 [M+H]+.


The Seventh Step

Compound 38-7 (305.0 mg, 547.2 μmol) was dissolved in methanol (15 mL), and to the mixture was added potassium carbonate (378.2 mg, 2.74 mmol). The reaction solution was stirred at 50° C. for 14 hours. The reaction solution was concentrated under reduced pressure, and the residue was isolated using a silica gel preparation plate (dichloromethane:methanol=10:1) to afford compound 38-8. 1H NMR (400 MHz, DMSO-d6) δ 9.45 (s, 1H), 8.28 (s, 1H), 7.67 (t, J=8.0 Hz, 1H), 7.58 (d, J=10.0 Hz, 1H), 7.20 (s, 1H), 7.14 (s, 1H), 5.17 (d, J=8.4 Hz, 1H), 4.05-4.10 (m, 2H), 3.98-4.05 (m, 1H), 3.93 (s, 3H), 2.15-2.30 (m, 4H), 1.95-2.10 (m, 2H), 1.70-1.80 (m, 2H). LC-MS: m/z=462.0 [M+H]+.


The Eighth Step

Compound 38-8 (220.0 mg, 475.8 μmol) was dissolved in a mixed solvent of tetrahydrofuran (6 mL) and water (6 mL) at 0° C. To the mixture was added sodium bicarbonate (119.9 mg, 1.43 mmol), and added slowly acryloyl chloride (38.8 mg, 428.3 μmol) dropwise. The reaction solution was further stirred at 0° C. for 0.5 hour. The reaction solution was quenched by adding MeOH (1 mL) and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (dichloromethane:methanol=10:1) to obtain the compound of example 38 (Rf=0.3). 1H NMR (400 MHz, DMSO-d6) δ 9.35 (s, 1H), 8.24 (s, 1H), 7.66 (t, J=8.0 Hz, 1H), 7.57 (d, J=8.8 Hz, 1H), 7.14 (s, 1H), 7.06 (s, 1H), 6.72 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.8 Hz, J2=1.6 Hz, 1H), 5.68 (dd, J1=10.4 Hz, J2=1.6 Hz, 1H), 5.35 (d, J=9.2 Hz, 1H), 4.50-4.60 (m, 2H), 4.05-4.15 (m, 1H), 3.92 (s, 3H), 1.85-2.20 (m, 6H), 1.60-1.70 (m, 1H), 1.50-1.60 (m, 1H). LC-MS: m/z=538.0 [M+Na]+.


Example 39



embedded image


Synthetic Route:




embedded image


The First Step

Compound of example 39 was obtained by referring to the eighth step of example 38 (Rf=0.4). 1H NMR (400 MHz, DMSO-d6) δ 9.38 (s, 1H), 8.27 (s, 1H), 7.50-7.60 (m, 2H), 7.14 (s, 1H), 7.05 (s, 1H), 6.74 (dd, J1=16.8 Hz, J2=10.4 Hz, 1H), 6.18 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.69 (dd, J1=10.4 Hz, J2=2.0 Hz, 1H), 5.47 (d, J=4.8 Hz, 1H), 4.50-4.60 (m, 2H), 4.01 (s, 3H), 3.80-3.90 (m, 1H), 2.20-2.30 (m, 1H), 1.85-2.15 (m, 7H). LC-MS: m/z=538.0 [M+Na]+.


Example 40



embedded image


Synthetic Route:




embedded image


The First Step

Compound 40-1 (200.0 mg, 853.94 μmol) was dissolved in thionyl chloride (3 mL), and to the mixture was added N,N-dimethylformamide (6 μL 85.39 μmol). The reaction solution was heated to 80° C. and stirred for 1 hour. The reaction solution was concentrated under reduced pressure, and to the residue was added a solution of 3-chloro-4-fluoroaniline (130.0 mg, 893.09 μmol) in isopropanol (5 mL) at room temperature. The mixture was heated to 90° C. and stirred for 3 hours. The reaction solution was concentrated under reduced pressure, and to the residue was added ethyl acetate (15 mL) at room temperature. The suspension was stirred at room temperature for 1 hour and then filtered. The filter cake was dried in vacuo to afford compound 40-2. LC-MS: m/z=320.1 [M+H]+.


The Second Step

Compound 40-2 (50.0 mg, 156.39 μmol) was dissolved in N,N-dimethylformamide (1 mL), and to the mixture were added cesium carbonate (100.0 mg, 306.92 μmol) and compound P-2 (50.0 mg, 149.07 μmol). The reaction solution was stirred at 90° C. for 45 minutes. The reaction solution was cooled to 15° C., poured into water (30 mL), and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed sequentially with water (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (ethyl acetate:tetrahydrofuran=4:1) to obtain the compound of example 40. 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.51 (s, 1H), 8.19 (dd, J1=6.8 Hz, J2=2.8 Hz, 1H), 7.95 (s, 1H), 7.78-7.84 (m, 1H), 7.46 (t, J=9.2 Hz, 1H), 7.21 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.97-5.11 (m, 1H), 4.58-4.63 (m, 2H), 3.92 (s, 3H), 2.30-2.38 (m, 1H), 2.20-2.27 (m, 1H), 2.00-2.09 (m, 2H), 1.84-1.97 (m, 2H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=483.1 [M+H]+.


Example 41



embedded image


Synthetic Route:




embedded image


The First Step

Compound 41-2 was obtained by referring to the first step of example 40. 1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.29 (s, 1H), 8.07 (s, 1H), 7.93-8.02 (m, 2H), 7.40 (s, 1H), 4.05 (s, 3H). LC-MS: m/z=370.0 [M+H]+.


The Second Step

Compound of example 41 was obtained by referring to the second step of example 40. 1H NMR (400 MHz, DMSO-d6) δ 9.82 (s, 1H), 8.65 (s, 1H), 8.44 (s, 1H), 8.04-8.09 (m, 1H), 7.87-7.94 (m, 2H), 7.28 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.96-5.08 (m, 1H), 4.58-4.63 (m, 2H), 3.94 (s, 3H), 2.23-2.37 (m, 2H), 1.80-2.12 (m, 4H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=532.9 [M+H]+.


Example 42



embedded image


Synthetic Route:




embedded image


The First Step

Compound 20-5 (200.0 mg, 364.02 μmol) was dissolved in N,N-dimethylformamide (2 mL), and to the mixture were added cesium carbonate (240.0 mg, 736.60 μmol), sodium iodide (4.0 mg, 26.69 μmol) and 3-iodooxetane (80.0 mg, 434.84 μmol). The reaction solution was stirred at 90° C. for 1 hour. The reaction solution was cooled to 15° C., poured into water (30 mL), and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed sequentially with water (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (ethyl acetate) to afford compound 42-2. 1H NMR (400 MHz, CDCl3) δ 8.63 (s, 1H), 8.48 (t, J=8.8 Hz, 1H), 7.27 (dd, J1=9.2 Hz, J2=2.0 Hz, 1H), 7.18 (s, 1H), 6.76 (s, 1H), 5.28-5.32 (m, 1H), 4.99-5.05 (m, 2H), 4.68-4.80 (m, 3H), 4.21-4.40 (m, 2H), 2.10-2.15 (m, 2H), 2.00-2.06 (m, 2H), 1.82-1.96 (m, 2H), 1.58-1.64 (m, 2H), 1.45 (s, 9H). LC-MS: m/z=605.3 [M+H]+.


The Second Step

Compound 42-2 (400.0 mg, 709.92 μmol) was dissolved in dichloromethane (2 mL), and to the mixture was added trifluoroacetic acid (1 mL). The reaction solution was reacted at 15° C. for 0.5 hour. The reaction solution was directly concentrated under reduced pressure to obtain the trifluoroacetate salt of compound 42-3. LC-MS: m/z=505.2 [M+H]+.


The Third Step

The trifluoroacetate salt of compound 42-3 (50.0 mg, 98.94 μmol) was dissolved in a mixed solvent of tetrahydrofuran (2 mL) and water (2 mL) at 0° C. To the mixture was added sodium bicarbonate (26.0 mg, 309.50 μmol), and added slowly acryloyl chloride (10.0 mg, 110.49 μmol) dropwise. The reaction solution was further stirred at 0° C. for 1 hour. The reaction solution was quenched by adding methanol (10 mL) and concentrated under reduced pressure. The residue was isolated by preparative thin-layer chromatography (ethyl acetate:tetrahydrofuran=4:1) to obtain the compound of example 42. 1H NMR (400 MHz, DMSO-d6) δ 9.72 (br s, 1H), 8.38 (s, 1H), 7.84 (s, 1H), 7.61 (s, 2H), 6.84-6.89 (m, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 5.40-5.52 (m, 1H), 4.95-5.08 (m, 3H), 4.55-4.69 (m, 4H), 2.25-2.43 (m, 2H), 1.86-2.05 (m, 4H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=559.0 [M+H]+.


Example 43



embedded image


Synthetic Route:




embedded image


The First Step

Compound 20-5 (300.0 mg, 546.03 μmol) was dissolved in N,N-dimethylformamide (3 mL), and then to the mixture was added sodium hydride (60%, 45 mg, 1.12 mmol) in one portion at 0° C. The reaction solution was stirred at 0° C. for 0.5 hour, and 1,2-dibromoethane (149.4 mg, 795.27 μmol) was added. The reaction solution was stirred at 15° C. for 11.5 hours, then quenched with water (50 mL) and extracted with ethyl acetate (50 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was isolated by preparative thin-layer chromatography (ethyl acetate) to afford compound 43-2. 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 8.46 (t, J=8.53 Hz, 1H), 7.20-7.29 (m, 4H), 4.66-4.77 (m, 1H), 4.40-4.45 (m, 2H), 4.18-4.32 (m, 2H), 3.70-3.75 (m, 2H), 2.03-2.15 (m, 2H), 1.90-1.95 (m, 2H), 1.72-1.82 (m, 2H), 1.58-1.65 (m, 2H), 1.45 (s, 9H). LC-MS: m/z=657.2 [M+H]+.


The Second Step

Compound 43-2 (170.0 mg, 259.00 μmol) was dissolved in tetrahydrofuran (3 mL), and to the mixture was added azetidine (42.3 mg, 740.88 μmol). The reaction solution was stirred at 50° C. for 1.5 hours, and then concentrated under reduced pressure to afford compound 43-3. LC-MS: m/z=632.2 [M+H]+.


The Third Step

The trifluoroacetate salt of compound 43-4 was obtained by referring to the second step of example 42. LC-MS: m/z=532.3 [M+H]+.


The Fourth Step

Compound of example 43 was obtained by referring to the third step of example 42. 1H NMR (400 MHz, DMSO-d6) δ 9.63 (s, 1H), 8.39 (s, 1H), 7.80 (s, 1H), 7.56-7.67 (m, 2H), 7.19 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.88-5.06 (m, 1H), 4.54-4.66 (m, 2H), 4.04-4.14 (m, 2H), 3.22-3.26 (m, 4H), 2.76-2.84 (m, 2H), 2.16-2.32 (m, 2H), 1.85-2.07 (m, 6H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=586.1 [M+H]+.


Example 44



embedded image


Synthetic Route:




embedded image


The First Step

Compound 44-2 was obtained by referring to the first step of example 43. LC-MS: m/z=607.3 [M+H]+.


The Second Step

Compound 44-2 (170.0 mg, 279.84 μmol) was dissolved in a solution of hydrogen chloride in ethyl acetate (4 N, 10 mL), and stirred at 20-25° C. for 15 minutes. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of compound 44-3. LC-MS: m/z=507.2 [M+H]+.


The Third Step

Compound of example 44 was obtained by referring to the third step of example 42. 1H NMR (400 MHz, DMSO-d6) δ 9.63 (s, 1H), 8.37 (s, 1H), 7.80 (s, 1H), 7.46-7.68 (m, 2H), 7.21 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.82-4.94 (m, 1H), 4.57-4.62 (m, 2H), 4.16-4.24 (m, 2H), 3.53-3.64 (m, 2H), 2.16-2.32 (m, 2H), 1.75-2.10 (m, 6H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=560.9 [M+H]+.


Example 45



embedded image


Synthetic Route:




embedded image


The First Step

Compound 45-2 was obtained by referring to the first step of example 42. 1H NMR (400 MHz, CDCl3) δ 8.71 (s, 1H), 8.53 (t, J=8.4 Hz, 1H), 7.27-7.37 (m, 3H), 7.17 (s, 1H), 4.71-4.84 (m, 1H), 4.28-4.44 (m, 3H), 2.14-2.22 (m, J=7.8 Hz, 2H), 1.54-2.01 (m, 10H), 1.51 (s, 9H), 1.33 (s, 6H). LC-MS: m/z=635.3 [M+H]+.


The Second Step

The hydrochloride salt of compound 45-3 was obtained by referring to the second step of example 44. LC-MS: m/z=535.3 [M+H]+.


The Third Step

Compound of example 45 was obtained by referring to the third step of example 42. 1H NMR (400 MHz, DMSO-d6) δ 9.75 (s, 1H), 8.37 (s, 1H), 7.84 (s, 1H), 7.49-7.66 (m, 2H), 7.22 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.88-5.03 (m, 1H), 4.55-4.64 (m, 2H), 4.18-4.27 (m, 2H), 2.17-2.39 (m, 2H), 1.82-2.07 (m, 6H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H), 1.17 (s, 6H). LC-MS: m/z=589.3 [M+H]+.


Example 46



embedded image


Synthetic Route:




embedded image


The First Step

Compound 46-2 was obtained by referring to the first step of example 40. 1H NMR (400 MHz, DMSO-d6) δ 11.43 (s, 1H), 8.90 (s, 2H), 8.34 (d, J=2.4 Hz, 1H), 8.19 (s, 1H), 8.15 (dd, J1=8.4 Hz, J2=2.4 Hz, 1H), 7.83 (d, J=8.4 Hz, 1H), 7.51 (s, 1H), 4.03 (s, 3H). LC-MS: m/z=370.1 [M+H]+.


The Second Step

Compound of example 46 was obtained by referring to the second step of example 40. 1H NMR (400 MHz, DMSO-d6) δ 10.16 (s, 1H), 8.50-8.62 (m, 2H), 8.30-8.38 (m, 1H), 8.17 (s, 1H), 7.73 (d, J=8.8 Hz, 1H), 7.23 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 5.09-5.21 (m, 1H), 4.54-4.65 (m, 2H), 3.93 (s, 3H), 2.20-2.34 (m, 2H), 1.87-2.10 (m, 4H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=533.1 [M+H]+.


Example 47



embedded image


Synthetic Route:




embedded image


The First Step

Compound 47-2 was obtained by referring to the first step of example 40. LC-MS: m/z=354.1 [M+H]+.


The Second Step

Compound of example 47 was obtained by referring to the second step of example 40. 1H NMR (400 MHz, DMSO-d6) δ 9.81 (s, 1H), 8.52 (s, 1H), 8.29-8.38 (m, 1H), 8.17-8.25 (m, 1H), 7.95 (s, 1H), 7.56 (t, J=9.54 Hz, 1H), 7.23 (s, 1H), 6.77 (dd, J1=16.4 Hz, J2=10.4 Hz, 1H), 6.22 (dd, J1=16.4 Hz, J2=2.4 Hz, 1H), 5.73 (dd, J1=10.4 Hz, J2=2.4 Hz, 1H), 4.97-5.12 (m, 1H), 4.57-4.69 (m, 2H), 3.93 (s, 3H), 2.19-2.31 (m, 2H), 1.88-2.03 (m, 4H), 1.65 (t, J=10.4 Hz, 1H), 1.49 (t, J=10.4 Hz, 1H). LC-MS: m/z=517.1 [M+H]+.


Example 48



embedded image


Synthetic Route:




embedded image


The First Step

Compound 48-2 (2.7 g) was obtained by referring to the sixth step of example 38. LC-MS: m/z=542.2 [M+H]+.


The Second Step

Compound 48-3 (0.9 g) was obtained by referring to the seventh step of example 38. LC-MS: m/z=446.1 [M+H]+.


The Third Step

Example 48 (160.0 mg) was obtained by referring to the eighth step of example 38. 1H NMR (400 MHz, CD3OD) D 8.23 (s, 1H), 7.61-7.52 (m, 1H), 7.27-7.17 (m, 2H), 7.09 (s, 1H), 6.75 (dd, J1=12.8 Hz, J2=10.4 Hz, 1H), 6.33 (dd, J1=16.8 Hz, J2=2.0 Hz, 1H), 5.79 (dd, J1=6.4 Hz, J2=2.0 Hz, 1H), 4.79-4.75 (m, 1H), 4.63-4.61 (m, 1H), 4.29-4.23 (m, 1H), 4.03 (s, 3H), 2.34-2.31 (m, 1H), 2.27-2.21 (m, 1H), 2.19-2.14 (m, 2H), 2.13-2.03 (m, 2H), 1.76-1.66 (m, 1H), 1.64-1.53 (m, 1H). LC-MS: m/z=500.2 [M+H]+.


Biochemical Assay: In Vitro Evaluation
Assay Example 1: Evaluation of Enzyme Activity

The purpose of this assay is to detect the in vitro inhibitory activity of compounds on HER1 (ErbB1), HER2 (ErbB2), and HER4 (ErbB4). The enzymes used in this assay are human ErbB1, ErbB2 and ErbB4. Eurofins Pharma Discovery Service provided the method for activity detection. The results of inhibitory activity of the test compounds on HER1, HER2, and HER4 are shown in Table 1.


Steps and Method of the Assay (96-Well Plate):


A buffer solution of the test compounds in a dilution of 5-fold (5 μL), polypeptide substrate poly(Glu, Tyr) (4:1) (2.5 μL), ErbB (4-20 ng, 2.5 μL), MnCl2 (50 mM, 1.25 μL), dH2O (3.75 μL), and [γ-33P]ATP (10 μL) were added, and incubated at 30° C. for 10 minutes. The reaction was terminated by adding 3% phosphoric acid. 10 μL of the sample was taken and transferred to Filtermate A. The filter disc was washed 3 times with 75 mM phosphoric acid and once with methanol. The filter disc was transferred to a sealed plastic bag and the scintillation mixture (4 mL) was added. The intensity of the emitted photon was detected by a scintillation luminescence counter. The CPM (times/min) of the enzyme sample was compared with the CPM of the internal control sample. The level of photon intensity reflected the degree of the tyrosine kinase activity.









TABLE 1







results of the in vitro enzyme activity screening


assay for compounds disclosed herein













HER1
HER2
HER4



Compound
IC50 (nM)
IC50 (nM)
IC50 (nM)
















Example 1
2
3
10



Example 3
2
5
17



Example 4
2
3
13



Example 5
9
24
157



Example 6
2
4
17



Example 7
40
69
61



Example 8
6.0b
5.5b
3.5b



Example 9
9
13
8



Example 13
5
8
7



Example 18
9
12
5



Example 20
18
11
10



Example 21
12
5
3



Example 22
12
7
3



Example 23
8
4
2



Example 24
10
6
3



Example 25
6
3
6



Example 26
8
5
4



Example 27
16
5
3



Example 32
15
5
3



Example 33
7
6
3



Example 34
5
4
2



Example 38
8
5
3



Example 40
11
5
2



Example 44
5
4
3



Example 45
8
7
10



Example 48
9
11
4








b: average of two tests.







Conclusion: the compounds disclosed herein have obvious inhibitory activity against HER1, HER2 and HER4.


Assay Example 2: Evaluation of Inhibitory Activity on Cell Proliferation

Purpose of the assay: to detect the inhibitory activity of the test compounds on cell proliferation.


Principle of the assay: the luciferase in a Cell-Titer-Glo reagent produces oxidized luciferin with luciferin, oxygen and ATP as reaction substrates, and releases energy in the form of light. Since the luciferase reaction requires ATP, the total amount of light generated by the reaction is proportional to the total amount of ATP that reflects cell viability.


Materials of the Assay:


Cell line: NCI-N87 cell line (ATCC-CRL-5822), BT-474 cell line (ATCC-HTB-20), OE21 (ECACC-96062201)


Cell culture medium: (RPMI 1640 medium (Invitrogen #22400-105; 10% serum Invitrogen #10090148; L-glutamine 1×, Gibco #25030-081; penicillin-streptomycin Hyclone #SV30010)


Cell Titer-Glo cell viability assay kit with luminescence (Promega #G7573)


384-well cell culture plate (Greiner #781090)


Compound plate (LABCYTE #LP-0200)


CO2 incubator (Thermo #371)


Vi-cell cell counter (Beckman Coulter)


Pipette (Eppendorf)


Pipetting tube (Greiner)


Pipetting gun (Eppendorf)


Multifunctional microplate reader (Envision Reader)


ECHO Liquid-handling workstation (Labcyte-ECHO555)


Steps and Method of the Assay:


2.1 Day 1:


According to the schematic diagram of the cell seed plate, cells were seeded in a 384- or 96-well plate at a density of 1000 cells per well and 25 μL per well, and 25 μL of PBS was added to the wells near edge of the place where no cells were seeded.


2.2 Day 0:


(1) The stock solution of the compounds has a concentration of 10 mM, and the compounds were diluted with DMSO to an initial concentration of 4 mM. The compounds were added to the plate with stock solution of compounds, 9 μL per well.


(2) The compounds were diluted with an ECHO liquid workstation, where 125 nL of compounds were added to each well of the cell plate, 125 nL of DMSO was added to each of the cell well in columns 2 and 23, and 125 nL of DMSO was added to each PBS well in columns 1 and 24.


(3) The cell plate was supplemented with 25 μL of medium per well, to a final volume of 50 μL per well; the compounds had a concentration of 1 M, and was serially 3-fold diluted to obtain 10 concentrations, and the left and right wells were used in duplicate; the final concentration of DMSO was 0.25%.


2.3 After addition of the compounds, the cell plate was centrifuged at 1000 rpm for 1 min, and then placed in a 37° C., 5% CO2 incubator for incubation for 3 days.


2.4 Day 3:


The cell plate was taken out from the incubator and equilibrated at room temperature for 30 minutes. 25 μL of Cell-Titer-Glo reagent was added to each well, shaken for one minute to allow well mixed, and centrifuged at 1000 rpm for 1 minute. After 10 minutes, the plate was read on PerkinElmer Envision, and the fluorescence reading time was set to 0.2 second.


Assay results: the assay results are shown in Table 2.


CTG assay of OE19 cells


OE19 cells with saturation of 80%-90% were digested with trypsin, centrifuged, resuspended and counted. The concentration of cells was adjusted to 90 ml/well. OE19 cells were added to a 96-well cell culture plate to 8000 OE19 cells per well. The cells were cultured in a cell incubator containing 5% CO2 at 37° C. overnight.


The stock solution of the test compounds was serially 3-fold diluted with DMSO to a total of 10 concentrations. Before the start of the assay, the serially diluted test compounds were further diluted to 10× compound solutions (the compound with a highest concentration contains 1% DMSO) with cell culture medium under sterile conditions. The prepared 10× compound solutions were added to the cell culture plate, and the 10 μL compound solution was added to each well. In this way, the final concentrations of staurosporine as the standard control and all the test compounds were obtained starting from 10 M as the initial concentration in a 3-fold serial dilution to get 10 test concentrations.


After 72 hours of cell culture, the 96-well cell culture plate was taken out. Cell Titer Glo reagent was added at 50 ml/well, mixed, centrifuged, and incubated at room temperature in the dark for 10 minutes. The cell plate was placed in Envision for reading.


The inhibition rate was calculated according to original data as follows:





Inhibition %=(ZPE−sample detection value)/(ZPE−HPE)×100%


The well containing 10 mM staurosporine on DAY3 (the first day of the assay is the day when compounds were added, and DAY3 is the day when reading is perfomed) was used as the HPE (100% inhibition control) well, and the well containing 0.1% DMSO on DAY3 was used as the ZPE (0% inhibition control) well. Each concentration of test compounds was tested in duplicate.


Non-linear regression analysis was performed using the processed data and GraphPad Prism 6 analysis software to obtain a dose-response curve, and the half-inhibitory concentration (IC50) of the test compounds on OE19 cells was calculated. The assay results are shown in Table 2.









TABLE 2







results of the screening assay of inhibitory activity of


compounds disclosed herein against cell proliferation in vitro












BT-474 cell
NCI-N87 cell
OE21 cellc
OE19 cellc


Compound
IC50 (nM)
IC50 (nM)
IC50 (nM)
IC50 (nM)














Example 1
ND
<1.5d
ND
ND


Example 7
ND
1.5d
ND
ND


Example 8
 0.39b
0.28b
0.47
0.48


Example 9
2.1d
0.9d
ND
ND


Example 13
2.0d
1.6d
ND
ND


Example 18
2.4d
1.3d
ND
ND


Example 20
ND
0.6
ND
ND


Example 21
0.46
0.35
ND
ND


Example 23
ND
0.14
ND
ND


Example 24
ND
0.17
ND
ND


Example 25
ND
0.19
ND
ND


Example 26
ND
0.24
ND
ND


Example 27
ND
0.31
ND
ND


Example 32
ND
0.29
ND
ND


Example 34
ND
0.42
0.27
 0.001


Example 38
ND
0.46
0.52
1.20


Example 40
ND
0.34
ND
ND


Example 42
ND
0.92
ND
ND


Example 44
ND
0.33
ND
ND


Example 45
ND
0.69
ND
ND


Example 48
ND
ND
0.17
0.24





ND: not tested;



b: average of two tests;




d: initial dilution concentration of compounds is 10 μM







Conclusion: the compounds disclosed herein have obvious inhibitory activity on the proliferation of NCI-N87, BT-474, OE21 and OE19 cells.


Assay Example 3: In Vivo Pharmacodynamics Study in BALB/c Nude Mouse Model with Subcutaneous Xenograft Tumor of Human Gastric Cancer NCI-N87 Cells

Purpose of the assay: to evaluate the in vivo efficacy of compounds to be tested on subcutaneous xenograft tumor of human gastric cancer NCI-N87 cells in a BALB/c nude mouse model.


Animals of the assay: female BALB/c nude mice, 6-8 weeks old, weighted 18-22 grams; supplier: shanghai sippr bk laboratory animals Ltd.


Method and Steps of the Assay:


3.1 Cell Culture

Human gastric cancer NCI-N87 cells were cultured in monolayer in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 100 U/mL penicillin, 100 μg/mL streptomycin and 2 mM glutamine, 37° C., 5% CO2. The passaging was performed by the conventional digestion with trypsin-EDTA twice a week. When the cell saturation was 80%-90%, the cells were collected, counted and inoculated.


3.2 Tumor Cell Inoculation (Tumor Inoculation)

0.2 mL (10×106 cells) of NCI-N87 cells were inoculated subcutaneously on the right back of each nude mouse (PBS+Matrigel, 1:1). When the average tumor volume reached 145 mm3, administration to each group was started.


3.3 Formulation of Test Compounds

The test compounds were formulated into a 0.3 mg/mL clear solution in the solvents of 10% NMP (N-methylpyrrolidone)+10% ethylene glycol stearate+80% water.


3.4 Tumor Measurement and Assay Index

The assay index is to investigate whether tumor growth is inhibited, delayed or cured. Tumor diameter was measured twice a week with a vernier caliper. The calculation formula of tumor volume is: V=0.5a×b2, wherein a and b represent the long and short diameters of tumor, respectively.


The antitumor efficacy of the compounds were evaluated by TGI (%) or relative tumor proliferation rate T/C (%). TGI (%) reflects the tumor growth inhibition rate. TGI (%) is calculated as follows: TGI (%)=[1−(average tumor volume at the end of administration for a certain treatment group−average tumor volume at the beginning of administration for this treatment group)/(average tumor volume at the end of treatment for a solvent control group−average tumor volume at the beginning of administration for this solvent control group)]×control group.


Relative tumor proliferation rate T/C (%): the calculation formula is as follows: T/C %=TRTV/CRTV (TRTV: RTV for the treatment group; CRTV: RTV for the negative control group). Relative tumor volume (RTV) is calculated based on tumor measurement results. The calculation formula is RTV=Vt/V0, wherein V0 is the average tumor volume measured at the beginning of administration (i.e., d0) to each group, Vt is the average tumor volume at a certain measurement, and the data for TRTV and CRTV were taken on the same day.


The tumor weight was measured after the assay is over and the T/C weight percentage was calculated. T weight and C weight represent the tumor weight of the administration group and the vehicle control group, respectively.


3.5 Statistical Analysis

Statistical analysis included the mean and standard error (SEM) of tumor volume at each time point in each group. The treatment group showed the best therapeutic effect at the end of the assay, i.e., on the twenty-first day after administration. Therefore, based on this data, statistical analysis was performed to assess the difference between the groups. The comparison between two groups was analyzed by T-test, and the comparison among three or more groups was analyzed by one-way ANOVA. If the F value shows a significant difference, a test should be carried out by Games-Howell method. If the F value does not show a significant difference, an analysis should be carried out by Dunnet (2-sided) method. All data analysis was carried out with SPSS 17.0. p<0.05 was considered a significant difference.


3.6 Assay Results

The body weight of test animals was used as a reference index for indirect determination of drug toxicity. In this model, all administration groups (except the Poziotinib group) did not show significant weight loss. In the Poziotinib group, on the 7th day after administration, 2 mice showed more than 20% of body weight loss, 3 mice showed more than 15% of body weight loss, and 1 mouse showed more than 10% of body weight loss. On the 8th day after administration, 2 mice were euthanized, and the remaining mice gradually recovered from lowering the dose of administration, and no morbidity or death was observed. The results of drug efficacy were shown in Table 3.


3.7 Conclusion of Assay and Discussion

Compared with the vehicle group, the control group Poziotinib and Example 1, Example 7 and Example 8 disclosed herein all exhibited excellent tumor growth inhibitory effects, in which TGI was 115.6%, 103.98%, 92.63% and 119.71%, respectively. Moreover, the safety of Example 1, Example 7 and Example 8 disclosed herein on the weight index of model animals is significantly better than that of the control compound Poziotinib.









TABLE 3







evaluation of the anti-tumor efficacy of compounds disclosed


herein on human gastric cancer NCI-N87 xenograft model


(calculated based on tumor volume on day 21 after administration)












Tumor volume
T/Cb
TGIb
P


Group
(mm3)a (Day 21)
(%)
(%)
value c





Vehicle group
727 ± 120





Example 1 (3 mg/kg)
122 ± 11 
16.62
103.98 
0.050


Example 7 (3 mg/kg)
188 ± 9 
26.10
92.63
0.079


Example 8
30 ± 7 
 4.19
119.71 
0.028


3 mg/kg (D0-7) + 1 mg/kg (D8-21)






Poziotinib
54 ± 12
 7.55
115.60 
0.032


3 mg/kg (D0-7) + 1 mg/kg (D8-21)





Note:


“—”means no calculation was required.



aMean ± SEM.




bTumor growth inhibition was calculated by T/C and TGI (TGI (%) = [1-(T21-T0)/(V21-V0)] × 100).




c The p value was based on tumor volume.







Assay Example 4: In Vivo Pharmacodynamics Study in BALB/c Nude Mouse Model with Subcutaneous Xenograft Tumor of Human Gastric Cancer NCI-N87 Cells

Purpose of the assay: to evaluate the in vivo efficacy of test compounds disclosed herein on subcutaneous xenograft tumor of human gastric cancer NCI-N87 cells in a BALB/c nude mouse model.


Animals of the assay: female BALB/c nude mice, 6-8 weeks old, weighted 18-22 grams; supplier: Shanghai Ling Chang Biotechnology Co., Ltd.


Method and Steps of the Assay:


4.1 Cell Culture

Human gastric cancer NCI-N87 cells were cultured in monolayer in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 100 U/mL penicillin, 100 U/mL streptomycin and 2 mM glutamine, 37° C., 5% CO2. The passaging was performed by conventional digestion with trypsin-EDTA twice a week. When the cell saturation was 80%-90%, the cells were collected, counted and inoculated.


4.2 Tumor Cell Inoculation (Tumor Inoculation)

0.2 mL (10×106) of NCI-N87 cells (PBS+Matrigel, 1:1) were inoculated subcutaneously on the right back of each mouse. When the average tumor volume reached 158 mm3, administration to each group was started.


4.3 Formulation of Test Compounds

The test compounds were formulated into a 0.04 mg/mL and 0.08 mg/mL clear solution in and the solvents of 10% NMP (N-methylpyrrolidone)+10% ethylene glycol stearate+80% water.


4.4 Tumor Measurement and Assay Index

The assay index is to investigate whether tumor growth is inhibited, delayed or cured. Tumor diameter was measured twice a week with a vernier caliper. The calculation formula of tumor volume is: V=0.5a×b2, wherein a and b represent the long and short diameters of tumor, respectively.


The antitumor efficacy of the compounds were evaluated by TGI (%) or tumor proliferation rate T/C (%). TGI (%) reflects the tumor growth inhibition rate. TGI (%) is calculated as follows: TGI (%)=[1−(average tumor volume at the end of administration for a certain treatment group−average tumor volume at the beginning of administration for this treatment group)/(average tumor volume at the end of treatment for a solvent control group−average tumor volume at the beginning of administration for this solvent control group)]×100%.


Tumor proliferation rate T/C (%): the calculation formula is as follows: T/C (%)=average tumor volume at the end of administration for a certain treatment group/average tumor volume at the end of treatment for the solvent control group×100%.


4.5 Statistical Analysis

Statistical analysis included the mean and standard error (SEM) of tumor volume at each time point in each group (See Table 5-1 for specific data). The treatment group showed the best therapeutic effect at the end of the assay, i.e., on the twenty-eight day after. Therefore, based on this data, statistical analysis was performed to assess the difference between the groups. The comparison between two groups was analyzed by T-test, and the comparison among three or more groups was analyzed by one-way ANOVA. After testing, F value showed significant difference, and thus a test was carried out by Games-Howell method. All data analysis was carried out with SPSS 17.0. p<0.05 was considered a significant difference.


4.6 Assay Results
4.6.1 Mortality, Morbidity and Body Weight Changes

The body weight of test animals was used as a reference index for indirect determination of drug toxicity. In this model, the body weight of mice in the treatment group has a downward trend, and there was no other morbidity or death.


4.6.2 Evaluation Index of Antitumor Efficacy









TABLE 4







evaluation of anti-tumor efficacy of compounds disclosed herein in


subcutaneous xenograft tumor model of human gastric cancer NCI-N87 cells


(calculated based on the tumor volume on the 28th day after administration)














Tumor volume
T/Cb
TGIb
P


Group
Dose
(mm3)a (Day 28)
(%)
(%)
valuec





Vehicle group

626 ± 60 





Poziotinib
0.8 mg/kg
28 ± 8 
 4.47
127.78
0.002


Poziotinib
0.4 mg/kg
84 ± 9 
13.42
115.81
0.003


Example 38
0.8 mg/kg
15 ± 3 
 2.40
130.56
0.002


Example 38
0.4 mg/kg
47 ± 13
 7.51
123.72
0.002





Note:


aMean + SEM.



bTumor growth inhibition was calculated by T/C and TGI (TGI (%) = [1-(T28-T0)/(V28-V0)] × 100).




cThe p value was calculated based on tumor volume.







4.7 Conclusion of Assay and Discussion

In the assay, in vivo efficacy of compounds disclosed herein in subcutaneous xenograft tumor model of human gastric cancer NCI-N87 cells was evaluated. Compared with the vehicle control group, example 38@0.8 mg/kg of the treatment group was comparable to the reference compound Poziotinib@0.8 mg/kg regarding the drug efficacy, and both exhibited significant tumor inhibiting effects; in the low-dose group, example 38 was comparable to or better than the reference compound Poziotinib regarding the drug efficacy.


Assay Example 5: In Vivo Pharmacodynamics Study in BALB/c Nude Mouse Model with Subcutaneous Xenograft Tumor of Human Gastric Cancer NCI-N87 Cells

Purpose of the assay: to evaluate the in vivo efficacy of test compounds disclosed herein on subcutaneous xenograft tumor of human gastric cancer NCI-N87 cells in a BALB/c nude mouse model.


Animals of the assay: female BALB/c nude mice, 6-8 weeks old, weighing 18-22 grams; supplier: shanghai sippr bk laboratory animals Ltd.


Method and Steps of the Assay:


5.1 Cell Culture

Human gastric cancer NCI-N87 cells were cultured in monolayer in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 100 U/mL penicillin, 100 μg/mL streptomycin, 37° C., 5% CO2. The passaging was performed by conventional digestion with trypsin-EDTA twice a week. When the cell saturation was 80%-90%, the cells were collected, counted and inoculated.


5.2 Tumor Cell Inoculation (Tumor Inoculation)

0.2 mL (10×106) of NCI-N87 cells (PBS+Matrigel, 1:1) were inoculated subcutaneously on the right back of each mouse. When the average tumor volume reached 156 mm3, administration to each group was started.


5.3 Formulation of Test Compounds

The test compounds were formulated into a 0.05 mg/mL clear solution in the solvents of 10% NMP (N-methylpyrrolidone)+10% ethylene glycol stearate+80% water.


5.4 Tumor Measurement and Assay Index

The assay index is to investigate whether tumor growth is inhibited, delayed or cured. Tumor diameter was measured twice a week with a vernier caliper. The calculation formula of tumor volume is: V=0.5a×b2, wherein a and b represent the long and short diameters of tumor, respectively.


The antitumor efficacy of the compounds were evaluated by TGI (%) or tumor proliferation rate T/C (%). TGI (%) reflects the tumor growth inhibition rate. TGI (%) is calculated as follows: TGI (%)=[1−(average tumor volume at the end of administration for a certain treatment group−average tumor volume at the beginning of administration for this treatment group)/(average tumor volume at the end of treatment for a solvent control group−average tumor volume at the beginning of administration for this solvent control group)]×100%.


Tumor proliferation rate T/C (%): the calculation formula is as follows: T/C (%)=average tumor volume at the end of administration for a certain treatment group/average tumor volume at the end of treatment for the solvent control group×100%.


5.5 Statistical Analysis

Statistical analysis included the mean and standard error (SEM) of tumor volume at each time point in each group (See Table 5-1 for specific data). The treatment group showed the best therapeutic effect on at the end of the assay, i.e., the twenty-eight day after administration. Therefore, based on this data, statistical analysis was performed to assess the difference between the groups. The comparison between two groups was analyzed by T-test, and the comparison among three or more groups was analyzed by one-way ANOVA. After testing, F value showed significant difference, and thus a test was carried out by Games-Howell method. All data analysis was carried out with SPSS 17.0. p<0.05 was considered a significant difference.


5.6 Assay Results
5.6.1 Mortality, Morbidity and Body Weight Changes

The body weight of test animals was used as a reference index for indirect determination of drug toxicity. In this model, the body weight of mice in the treatment group has a downward trend, and there was no other morbidity or death.


4.6.2 Evaluation Index of Antitumor Efficacy









TABLE 5







evaluation of anti-tumor efficacy of compounds disclosed herein in


subcutaneous xenograft tumor model of human gastric cancer NCI-N87 cells


(calculated based on the tumor volume on the 21th day after administration)














Tumor volume
T/Cb
TGIb
P


Group
Dose
(mm3)a (Day 21)
(%)
(%)
valuec





Vehicle group

631 ± 107





Example 38
0.5 mg/kg
63 ± 10
10.69
119.41 
0.007


Control
0.5 mg/kg
541 ± 88 
89.35
18.83
0.796


compound 2





Note:



aMean + SEM.




bTumor growth inhibition was calculated by T/C and TGI (TGI (%) = [1-(T21-T0)/(V21-V0)] × 100).




cThe p value was calculated based on tumor volume.







4.7 Conclusion of Assay and Discussion

In the assay, in vivo efficacy of the compounds disclosed herein and control compound 2 in subcutaneous xenograft tumor model of human gastric cancer NCI-N87 cells was evaluated. Compared with the solvent control group, Example 38@0.5 mg/kg of the treatment group exhibited significant tumor inhibiting effect, in which the tumor inhibiting rate was T/C=10.69%, TGI=119.41%, and p=0.007; at the same dose, the efficacy of control compound 2 was not obvious, in which T/C=89.35%, and TGI=18.83%; the efficacy of the compounds disclosed herein was significantly better than that of the control compound 2.


Assay Example 6: In Vivo Pharmacodynamics Study in BALB/c Nude Mouse Model with Subcutaneous Xenograft Tumor of Human Esophageal Carcinoma OE21 Cells

Purpose of the assay: to evaluate the in vivo efficacy of the compounds to be tested on subcutaneous xenograft tumor of human esophageal carcinoma OE21 cells in a BALB/c nude mouse model.


Animals of the assay: female BALB/c nude mice, 6-8 weeks old, weighing 17-23 grams; supplier: shanghai sippr bk laboratory animals Ltd.


Method and Steps of the Assay:


6.1 Cell Culture

Human esophageal carcinoma OE21 cells were cultured in monolayer in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 100 U/mL penicillin, 100 U/mL streptomycin and 2 mM glutamine, 37° C., 5% CO2. The passaging was performed by conventional digestion with trypsin-EDTA twice a week. When the cell saturation was 80%-90%, the cells were collected, counted and inoculated.


6.2 Tumor Cell Inoculation (Tumor Inoculation)

0.1 ml (20.1) OE21 cells (PBS) were inoculated subcutaneously on the right back of each mouse. When the average tumor volume reached 112 mm3, administration to each group was started.


6.3 Formulation of Test Compounds

The test compound was formulated into a 0.15 mg/mL to 0.3 mg/mL clear or suspension solution in the solvents of 10% NMP+10% ethylene glycol stearate+80% water. 6.4 Tumor measurement and assay index


The assay index is to investigate whether tumor growth is inhibited, delayed or cured. Tumor diameter was measured twice a week with vernier caliper. The calculation formula of tumor volume is: V=0.5a×b2, wherein a and b represent the long and short diameters of tumor, respectively.


The antitumor efficacy of the compounds were evaluated by TGI (%) or relative tumor proliferation rate T/C (%). TGI (%) reflects the tumor growth inhibition rate. TGI (%) is calculated as follows: TGI (%)=[(1−(average tumor volume at the end of administration for a certain treatment group−average tumor volume at the beginning of administration for this treatment group))/(average tumor volume at the end of treatment for a solvent control group−average tumor volume at the beginning of administration for this solvent control group)]×100%.


Relative tumor proliferation rate T/C (%): the calculation formula is as follows: T/C (%)=average tumor volume at the end of administration for a certain treatment group/average tumor volume at the end of treatment for the solvent control group×100%.


6.5 Statistical Analysis

Statistical analysis included the mean and standard error (SEM) of tumor volume at each time point in each group. One-way ANOVA was used for comparison among three or more groups. If the F value showed significant difference, multiple comparison should be made after ANOVA analysis. SPSS 17.0 was used for all data analysis. p<0.05 is considered a significant difference.


6.6 Assay Results
6.6.1 Mortality, Morbidity and Body Weight Changes

In this model, animals in the Poziotinib@2/1.5 mg/kg, Example 8/Example 38@2/0.5 mg/kg, and Example 38@2/1.5 mg/kg administration groups all showed varying degrees of body weight loss and dandruff; in the Poziotinib@2/1.5 mg/kg administration group, from the 6th and 7th day after the administration to each group, 1 mouse showed more than 15% of body weight loss and 3 mice showed more than 15% of body weight loss, and their administration was discontinued; moreover, mice in all groups had dark yellow urine, which suggested toxicity; starting from the 3rd day after administration, in the test group, Example 8@2 mg/kg was replaced with Example 38@0.5 mg/kg, the dose of Poziotinib@2 mg/kg was reduced to 1.5 mg/kg, and the dose of Example 38@2 mg/kg was reduced to 1.5 mg/kg. After the dose adjustment, the body weight of each group of animals recovered significantly (FIG. 1).


6.6.2 Evaluation Index of Antitumor Efficacy









TABLE 6







evaluation of anti-tumor efficacy of compounds disclosed


herein on OE21 xenograft tumor model


(calculated based on tumor volume on day 21 after administration)














Tumor volume
T/Cb
TGIb
P


Group
Dose
(mm3)a (Day 21)
(%)
(%)
valuec





Vehicle group

948 ± 124





Poziotinib
2/1.5 mg/kg
57 ± 7 
6.05
106.65
0.008


Example 8/
2/0.5 mg/kg
56 ± 15
5.89
106.87
0.008


Example 38







Example 38
2/1.5 mg/kg
55 ± 13
5.82
106.65
0.008





Note:


aMean + SEM.



bTumor growth inhibition was calculated by T/C and TGI by referring to Assay Example 4.




cThe p value was calculated based on the tumor volume on the 21st day after administration using One-Way ANOVA statistical method by comparing with the vehicle group.







6.7 Conclusion of Assay and Discussion

In this assay, in vivo efficacy of compounds disclosed herein and the reference compound Poziotinib in the OE21 xenograft tumor model was evaluated. On the 21st day after the administration, the tumor volume of tumor-bearing mice in the solvent control group reached 948 mm3. The test substance Poziotinib@2/1.5 mg/kg, Example 8/Example 38@2/0.5 mg/kg and Example 38@2/1.5 mg/kg group had a significant tumor inhibiting effect compared with the solvent control group. The tumor inhibition rates were: T/C=6.05%, TGI=106.65%, p=0.008 (Poziotinib@2/1.5 mg/kg group); T/C=5.89%, TGI=106.87%, p=0.008 (Example 8/Example 38@2/0.5 mg/kg group); T/C=5.82%, TGI=106.65%, p=0.008 (Example 38@2/1.5 mg/kg group), respectively. The efficacy of low-dose 0.5 mpk of Example 38 was comparable to that of high-dose 1.5 mpk of reference compound Poziotnib and Example 38.


Example 38 has significant tumor inhibiting effect, and its efficacy is comparable to that of the reference compound Poziotinib. However, in the high-dose group of the reference compound, the body weight loss of mice was significant. Three mice showed more than 15% of body weight loss, and their administration was discontinued. Moreover, mice in all groups had dark yellow urine, which suggested toxicity. Example 38 had little effect on the body weight of mice, without discontinuing the administration and without the toxicity suggestion of dark yellow urine, suggesting that its tolerability was significantly better than that of the reference compound. Compared with the reference compound Poziotinib, Example 38 had lower effective dose, better tolerability, and superior safety window.


Assay Example 7: Assay to Study Pharmacokinetics in Mice

Purpose of the assay: this assay aims to investigate in vivo plasma pharmacokinetics of the example compounds disclosed herein and the reference compound in female BALB/c mice after the single intravenous injection and intragastric administration thereof.


Animals of the assay: female BALB/c nude mice, 7-9 weeks old, weighted 17-23 grams; supplier: shanghai sippr bk laboratory animals Ltd.


Sample collection: at each time point, a 0.03 mL blood sample was collected from the saphenous vein of the test animal by puncture, and the actual blood collection time was recorded. All blood samples were added into commercial EDTA-K2 anticoagulation tubes with a specification of 1.5 mL (the supplier was Jiangsu KANGJIAN Medical Apparatus Co., Ltd.). Within half an hour after the blood sample was collected, the blood sample was centrifuged at 3000 g at 4° C. for 10 minutes, and the supernatant plasma was drawn, quickly placed in dry ice, and stored in a refrigerator at −80° C. for LC-MS/MS analysis.


Data analysis: Plasma concentrations were processed using a non-compartmental model of WinNonlin™ Version6.3 (Pharsight, MountainView, Calif.) pharmacokinetic software, and pharmacokinetic parameters Cmax, Tmax, T1/2, and AUC0-last were calculated using a linear log trapezoid method.









TABLE 7







Comparison of PK results of the compounds disclosed herein and the reference compound












Cmax (nM)
Tmax (h)
T1/2 (h)
AUC0-last (nM · h)





Example 38
10600
2.0
3.2
122154


Poziotinib
17267
 0.25
2.3
 97870


Control compound 2
15300
2.0
1.5
 82021





Cmax: peak concentration;


Tmax: time to reach peak concentration;


T1/2: the time required to clear half of the compound;


AUC0-last: integrated area of concentration in the time from 0 to the last sampling.






The results of pharmacokinetic studies in mice showed that the compounds disclosed herein had a longer half-life than that of the reference compound, and had significantly better oral plasma exposure than that of the reference compound Poziotinib and the control compound 2.


Assay Example 8: Evaluation of Inhibition of CYP Enzyme Activity

The results of the compounds disclosed herein and the reference compound Poziotinib in the assay of CYP enzyme activity in human liver microsome were shown in the following table (Table 8).









TABLE 8







comparison of results of the compounds disclosed herein and


the reference compound on the inhibition of CYP enzyme













CYP1A2
CYP2C9
CYP2C19
CYP2D6
CYP3A4





Example 38
>50
16
11
6
>50


Poziotinib
>50
 8
12
1
>50









The results of inhibition of CYP activity of human liver microsome by the compounds disclosed herein showed that example 38 and Poziotinib had no inhibitory activity on CYP1A2 and CYP3A4. The inhibitory activity of example 38 on CYP2C19 was comparable to that of the reference compound Poziotinib, and the activity of example 38 on CYP2C9 and CYP2D6 was improved, which was 2 times and 6 times better than that of the reference compound, respectively. In a comprehensive comparison, example 38 had improved activity on CYP and was superior to that of the reference compound, so the risk of drug-drug interaction was lower.

Claims
  • 1. A compound of Formula (I), an isomer thereof or a pharmaceutically acceptable salt thereof,
  • 2. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein each R is independently selected from the group consisting of F, Cl, Br, I, OH, NH2, CN,
  • 3. (canceled)
  • 4. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein each R1 is independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, CN, Me and
  • 5. (canceled)
  • 6. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein R2 and R3 are each independently selected from the group consisting of H, F, Cl, Br, I, OH, NH2, Me and CF3.
  • 7. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein R4 is selected from the group consisting of
  • 8. (canceled)
  • 9. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein R5 and R6 are each independently selected from the group consisting of H, Me, Et and
  • 10. (canceled)
  • 11. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein T is selected from the group consisting of N and C(CN).
  • 12. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein L is selected from the group consisting of —O—, —NH—, —CH2—, —CH2—O— and —CH2—NH—.
  • 13. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the moiety
  • 14. (canceled)
  • 15. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the ring A is selected from the group consisting of phenyl, azaindenyl, benzo[b]thienyl, imidazo[1,2-a]pyridyl and benzo[d]isoxazolyl.
  • 16. The compound according to claim 15, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the moiety
  • 17-18. (canceled)
  • 19. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the ring B is selected from the group consisting of
  • 20. The compound according to claim 19, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the moiety
  • 21. The compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  • 22. The compound according to claim 21, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  • 23. A compound, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  • 24. The compound according to claim 23, which is selected from the group consisting of:
  • 25. A pharmaceutical composition, comprising a therapeutically effective amount of the compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof, as an active ingredient, and pharmaceutically acceptable carrier(s).
  • 26. A method of treating a disease associated with Pan-HER tyrosine kinase in a subject in need thereof, comprising administering to the subject the compound according to claim 1, an isomer thereof or a pharmaceutically acceptable salt thereof.
  • 27. A compound, an isomer thereof or a pharmaceutically acceptable salt thereof, wherein the compound has the following formula:
Priority Claims (3)
Number Date Country Kind
201711376757.0 Dec 2017 CN national
201810225742.2 Mar 2018 CN national
201810576529.6 Jun 2018 CN national
REFERENCE TO RELATED APPLICATIONS

This application is a national stage filing under 35 U.S.C. § 371 of international application number PCT/CN2018/122016, filed Dec. 19, 2018, which claims the benefit of priority of Chinese Patent Application No. 201810576529.6, filed Jun. 6, 2018, Chinese Patent Application No. 201810225742.2, filed Mar. 19, 2018, and Chinese Patent Application No. 201711376757.0, filed Dec. 19, 2017, the entire contents of each of which are herein incorporated by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/CN2018/122016 12/19/2018 WO 00