The present application relates to a compound for treating cancer, in particular to a methionine adenosyltransferase 2A inhibitor, a preparation method thereof, a pharmaceutical composition comprising the same, and pharmaceutical use thereof.
Methionine adenosyltransferase (MAT), also known as S-adenosylmethionine synthetase, has the major biological function of catalyzing the reaction of methionine with ATP to produce a biological methyl donor S-adenosylmethionine (SAM, AdoMet, or SAMe). The MAT-catalyzed reaction is considered to be an important rate-limiting step in the methionine metabolic cycle.
The MAT family includes three family members, MAT1A, MAT2A, and MAT2B. MAT1A is specifically expressed in human liver, while MAT2A and MAT2B are widely distributed in multiple human tissues. In normal liver cells, there is a dynamic equilibrium between MAT 1A and MAT2A, which together maintain intracellular SAM homeostasis, whereas in hepatocellular carcinoma (HCC), MAT1A expression is abnormally down-regulated, and MAT2A expression is abnormally up-regulated. This gene expression change from MAT1A to MAT2A promotes cancer invasion and metastasis, and is closely related to increased metastasis and relatively poor recurrence-free survival (RFS) of HCC patients. See, e.g., Ruizhi Wang et al., Molecular Carcinogenesis 57 (9), (2018) 1201-1212. In addition to hepatocellular carcinoma, the abnormally increased expression level of MAT2A is also present in various tumors such as gastric cancer, colorectal cancer, and pancreatic cancer, and is closely related to the development and progression of tumors. MAT2A plays an important role in the pathogenesis of various tumors, and gene silencing of MAT2A can inhibit the proliferation of cancer cells such as liver cancer cells or gastric cancer cells and induce apoptosis. Therefore, MAT2A has become a target for drugs of antitumor therapy. See, e.g., T. Li et al., J. Cancer 7 (10) (2016) 1317-1327 and T. Zhang et al., Acta Histochemica 115 (2013) 48-55.
Further, it has been reported that MAT2A is a synthetic lethal target in MTAP-deficient cancer. MTAP, i.e., S-methyl-5′-thioadenosine phosphorylase, is widely distributed in normal tissues and cells, and mainly catalyzes the conversion of methylthioadenosine (MTA) to 5′-methylthioribose-1-phosphate (MTRP) and adenine. The process is an important procedure in the methionine salvage pathway in the human body. The gene encoding human MTAP is located in the chromosome 9p21 region. Studies have shown that the chromosome 9p21 region has a homozygous deletion frequency in all tumors of about 15%, and the deletion frequency varies among different tumors. The cancer with relatively high MTAP deletion frequency includes glioma, mesothelioma, esophageal cancer, bladder cancer, pancreatic cancer, melanoma, non-small cell lung cancer, head and neck cancer, cholangiocarcinoma, esophagogastric cancer, osteosarcoma, brain diffuse glioma, gastric cancer, adrenocortical carcinoma, breast cancer, thymus cancer, hepatocellular carcinoma, ovarian cancer, renal cell carcinoma, and the like. Further, K Marjon et al. found that S-methyl-5′-thioadenosine phosphorylase (MTAP)-deficient cancer was more sensitive to signaling pathways targeting MAT2A/PRMT5/RIOK1 (Cell Reports Volume 15, Issue 3, 2016, 574-587). When MTAP is deleted, the metabolic pathway of MTA is inhibited, resulting in the accumulation of MTA in vivo in a large amount. The accumulation of MTA in a large amount will partially inhibit the activity of PRMT5.
MAT2A is a key enzyme for SAM production in cells. Inhibition of MAT2A activity results in SAM reduction, causing PRMT5 to lack the corresponding substrate. Therefore, in MTAP-deficient tumor cells, inhibition of MAT2A further inhibits the methylation function of PRMT5, which in turn causes defects in RNA cleavage, gene expression, and chromosomal integrity, thereby inhibiting tumor cell growth.
Given that MAT2A is abnormally expressed in various human tumors (such as liver cancer, pancreatic cancer, gastric cancer, and colon cancer), and inhibition of MAT2A can selectively reduce the proliferative capacity of MTAP-deficient tumor cells and has weak effect on normal cells with MTAP normally expressed, the development of MAT2A inhibitors can be used as an effective tumor treatment method.
The present application provides an effective methionine adenosyltransferase 2A inhibitor, a preparation method thereof, a pharmaceutical composition comprising the same, and pharmaceutical use thereof, in particular for treating MTAP-deficient cancer.
In one aspect, the present application provides a compound of formula (I) or a stereoisomer, a tautomer, a solvate, a hydrate, a prodrug, a stable isotopic derivative, or a pharmaceutically acceptable salt thereof,
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof has a structure of formula (IA):
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof has a structure of formula (IB1), (IB2), (IB3), (IB4), or (IB5):
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof has a structure of formula (IC1), (IC2), (IC3), (IC4), or (IC5):
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof has optionally the following structures:
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof has optionally the following structures
In another aspect, the present application provides a preparation method for the compound of formula (I) described above.
In another aspect, the present application provides a composition comprising a therapeutically effective amount of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in inhibiting methionine adenosyltransferase 2A (MAT2A), wherein the inhibitory effect may be in vitro inhibition or in vivo inhibition.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in preparing a drug for inhibiting methionine adenosyltransferase 2A (MAT2A).
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in treating and/or preventing cancer, in particular cancer in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in preparing a drug for treating and/or preventing cancer, in particular cancer in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional.
In another aspect, the present application provides a method for treating and/or preventing cancer, comprising administering to a patient a therapeutically effective amount of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above, wherein the cancer includes those in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional.
The methionine adenosyltransferase 2A inhibitor with a novel structure provided by the present application has excellent inhibitory activity.
In order to better understand the content of the present application, the terms in the present application are explained as follows.
“Alkyl” refers to a saturated aliphatic hydrocarbon group that is a linear or branched group containing 1 to 20 carbon atoms, preferably alkyl containing 1 to 12 carbon atoms, and more preferably alkyl containing 1 to 6 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,2-dimethylpentyl, 3,3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2,3-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 2,2-dimethylhexyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2,2-diethylpentyl, n-decyl, 3,3-diethylhexyl, 2,2-diethylhexyl, and various branched-chain isomers thereof, and the like. The alkyl may be optionally independently substituted with one or more of the substituents described herein.
“Alkylene” refers to a saturated linear or branched aliphatic hydrocarbon group having 2 residues derived from the parent alkane by removal of two hydrogen atoms from the same carbon atom or two different carbon atoms. It is a linear or branched group containing 1 to 20 carbon atoms, preferably containing 1 to 12 carbon atoms, and more preferably alkylene containing 1 to 6 carbon atoms. Non-limiting examples of alkylene include, but are not limited to, methylene (—CH2—), 1,1-ethylene (—CH(CH3)—), 1,2-ethylene (—CH2CH2—), 1,1-propylene (—CH(CH2CH3)—), 1,2-propylene (—CH2CH(CH3)—), 1,3-propylene (—CH2CH2CH2—), 1,4-butylene (—CH2CH2CH2CH2—), and the like. The alkylene may be optionally independently substituted with one or more of the substituents described herein.
“Alkenyl” refers to a linear or branched monovalent hydrocarbon group containing 2 to 8 carbon atoms and having at least one site of unsaturation (i.e., a carbon-carbon sp2 double bond), wherein the alkenyl may be optionally independently substituted with one or more substituents described herein, and includes groups having “cis” and “trans” orientations (or “E” and “Z” orientations). Examples include, but are not limited to, vinyl (—CH═CH2), allyl (—CH2CH═CH2), and the like.
“Alkynyl” refers to a linear or branched monovalent hydrocarbon group containing 2 to 8 carbon atoms and having at least one site of unsaturation (i.e., a carbon-carbon sp triple bond), wherein the alkynyl may be optionally independently substituted with one or more substituents described herein. Examples include, but are not limited to, ethynyl (—C≡CH), propynyl (propargyl and —CH2C≡CH), and the like.
“Carbocyclic ring” or “cycloalkyl” that may be used interchangeably herein refers to a saturated or partially unsaturated monocyclic or polycyclic hydrocarbon substituent. The cycloalkyl ring contains 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 8 carbon atoms, and most preferably 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like. Polycyclic cycloalkyl includes spirocycloalkyl, fused cycloalkyl, and bridged cycloalkyl. The cycloalkyl may be optionally independently substituted with one or more of the substituents described herein.
“Spirocycloalkyl” refers to a 5- to 20-membered polycyclic group in which monocyclic rings share one carbon atom (referred to as a spiro atom). It may contain one or more double bonds, but none of the rings has a fully conjugated π-electron system. It is preferably 6- to 14-membered, and is more preferably 7- to 10-membered. According to the number of spiro atoms shared among rings, spirocycloalkyl may be monospirocycloalkyl, bispirocycloalkyl or polyspirocycloalkyl, preferably monospirocycloalkyl and bispirocycloalkyl. Non-limiting examples of spirocycloalkyl include;
“Fused cycloalkyl” refers to a 5- to 20-membered all-carbon polycyclic group in which each ring shares a pair of adjacent carbon atoms with the other rings in the system, wherein one or more of the rings may contain one or more double bonds. It is preferably 6- to 14-membered, and is more preferably 7- to 10-membered. According to the number of the formed rings, fused cycloalkyl may be bicyclic, tricyclic, tetracyclic, or polycyclic fused cycloalkyl, and is preferably bicyclic or tricyclic. Non-limiting examples off used cycloalkyl include:
“Bridged cycloalkyl” refers to a 5- to 20-membered all-carbon polycyclic group in which any two rings share two carbon atoms that are not directly connected, and it may contain one or more double bonds. It is preferably 6- to 14-membered, and is more preferably 7- to 10-membered.
According to the number of the formed rings, bridged cycloalkyl may be bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, preferably bicyclic, tricyclic or tetracyclic, and more preferably bicyclic or tricyclic. Non-limiting examples of bridge cycloalkyl include:
The cycloalkyl ring includes those in which the cycloalkyl described above (including monocyclic, spiro, fused and bridged rings) is fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring connected to the parent structure is cycloalkyl. Non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptanyl, and the like.
“Alkoxy” refers to (alkyl)-O—, wherein the alkyl is as defined above. Non-limiting examples of alkoxy include methoxy, ethoxy, propoxy, and butoxy. The alkoxy may be optionally independently substituted with one or more of the substituents described herein.
“Heterocyclic ring” or “heterocyclyl” that may be used interchangeably herein refers to a saturated or partially unsaturated monocyclic or polycyclic substituent containing 3 to 20 ring atoms, wherein one or more of the ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, the sulfur optionally being substituted with oxo (i.e., to form sulfoxide or sulfone), but excluding a cyclic portion of —O—O—, —O—S— or —S—S—; the remaining ring atoms are carbon. Preferably, it contains 3 to 12 (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12) ring atoms, wherein 1 to 4 (e.g., 1, 2, 3, and 4) are heteroatoms; more preferably, it contains 3 to 8 ring atoms (e.g., 3, 4, 5, 6, 7, and 8), wherein 1-3 (e.g., 1, 2, and 3) are heteroatoms. Non-limiting examples of monocyclic heterocyclyl include pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. Polycyclic heterocyclyl includes spiroheterocyclyl, fused heterocyclyl, and bridged heterocyclyl. The heterocyclyl may be optionally independently substituted with one or more of the substituents described herein.
“Spiroheterocyclyl” refers to a 5- to 20-membered polycyclic heterocyclyl group in which monocyclic rings share one atom (referred to as a spiro atom), wherein one or more of the ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, the sulfur optionally being substituted with oxo (i.e., to form sulfoxide or sulfone); the remaining ring atoms are carbon. It may contain one or more double bonds. It is preferably 6- to 14-membered, and more preferably 7- to 10-membered (e.g., 7-membered, 8-membered, 9-membered or 10-membered). According to the number of spiro atoms shared among rings, spiroheterocyclyl may be monospiroheterocyclyl, bispiroheterocyclyl or polyspiroheterocyclyl, preferably, monospiroheterocyclyl and bispiroheterocyclyl. Non-limiting examples of spiroheterocyclyl include:
“Fused heterocyclyl” refers to a 5- to 20-membered polycyclic heterocyclyl group in which each ring shares a pair of adjacent atoms with the other rings in the system, wherein one or more of the rings may contain one or more double bonds, wherein one or more of the ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, the sulfur optionally being substituted with oxo (i.e., to form sulfoxide or sulfone); the remaining ring atoms are carbon. It is preferably 6- to 14-membered, and more preferably 7- to 10-membered (e.g., 7-membered, 8-membered, 9-membered or 10-membered). According to the number of the formed rings, fused heterocyclyl may be bicyclic, tricyclic, tetracyclic, or polycyclic fused heterocyclyl, and is preferably bicyclic or tricyclic. Non-limiting examples of fused heterocyclyl include:
“Bridged heterocyclyl” refers to a 5- to 14-membered polycyclic heterocyclyl group in which any two rings share two atoms that are not directly connected, and it may contain one or more double bonds, wherein one or more ring atoms are heteroatoms selected from nitrogen, oxygen, and sulfur, the sulfur optionally being substituted with oxo (i.e., to form sulfoxide or sulfone); the remaining ring atoms are carbon. It is preferably 6- to 14-membered, and more preferably 7- to 10-membered (e.g., 7-membered, 8-membered, 9-membered or 10-membered). According to the number of the formed rings, bridged heterocyclyl may be bicyclic, tricyclic, tetracyclic, or polycyclic bridged heterocyclyl, and is preferably bicyclic, tricyclic, or tetracyclic. Non-limiting examples of bridged heterocyclyl include:
The heterocyclyl ring includes those in which the heterocyclyl described above (including monocyclic, spiro heterocyclic, fused heterocyclic and bridged heterocyclic rings) is fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring connected to the parent structure is heterocyclyl; its non-limiting examples include:
“Aromatic ring” or “aryl” that may be used interchangeably herein refers to a 6- to 14-membered all-carbon monocyclic or fused polycyclic (fused polycyclic rings are rings that share a pair of adjacent carbon atoms) group with a conjugated π-electron system, which is preferably 6- to 10-membered, e.g., phenyl and naphthyl. The aryl ring includes those in which the aryl ring described above is fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring connected to the parent structure is an aryl ring; its non-limiting examples include:
The aryl may be optionally independently substituted with one or more of the substituents described herein.
“Heteroaromatic ring” and “heteroaryl” that may be used interchangeably herein refers to a heteroaromatic system containing 1 to 4 (e.g., 1, 2, 3 and 4) heteroatoms and 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. The heteroaryl is preferably 5- to 10-membered (e.g., 5-, 6-, 7-, 8-, 9- or 10-membered), and is more preferably 5- or 6-membered, e.g., furanyl, thienyl, pyridinyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, etc. The heteroaryl ring includes those in which the heteroaryl described above is fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring connected to the parent structure is a heteroaryl ring; its non-limiting examples include.
The heteroaryl may be optionally independently substituted with one or more of the substituents described herein, which when substituted, may be substituted at any available linking site.
The cycloalkyl, heterocyclyl, aryl and heteroaryl described above include residues derived from the parent ring by removal of one hydrogen atom from a ring atom, or residues derived from the parent ring by removal of two hydrogen atoms from the same ring atom or two different ring atoms, i.e., “divalent cycloalkyl”, “divalent heterocyclyl”, “arylene” and “heteroarylene”.
“Cycloalkyloxy” refers to cycloalkyl-O—, wherein the cycloalkyl is as defined above. The cycloalkyloxy may be optionally independently substituted with one or more of the substituents described herein.
“Heterocyclyloxy” refers to heterocyclyl-O—, wherein the heterocyclyl is as defined above. The heterocyclyloxy may be optionally independently substituted with one or more of the substituents described herein.
“Aryloxy” refers to aryl-O—, wherein the aryl is as defined above. The aryloxy may be optionally independently substituted with one or more of the substituents described herein.
“Heteroaryloxy” refers to heteroaryl-O—, wherein the heteroaryl is as defined above. The heteroaryloxy may be optionally independently substituted with one or more of the substituents described herein.
“Sulfonyl” refers to —SO2—. The sulfonyl may be optionally substituted with the substituent described herein, such as alkylsulfonyl, cycloalkylsulfonyl, heterocyclylsulfonyl, arylsulfonyl, wherein the alkyl, cycloalkyl, heterocyclyl, and aryl are as defined above.
“Alkylthio” refers to alkyl-S—, wherein the alkyl is as defined above.
“Halogen” refers to fluorine, chlorine, bromine, or iodine.
“Cyano” refers to —CN.
“Hydroxyl” refers to —OH.
“Oxo” refers to “═O”.
“Carboxyl” refers to —C(═O)OH.
“Amino” refers to —NH2, which is optionally substituted with one or more of the substituents described herein, to form, for example, alkylamino, dialkylamino, cycloalkylamino, heterocyclylamino, and arylamino, wherein the alkyl, cycloalkyl, heterocyclyl, and aryl are as defined above.
“Alkylamino” refers to the RNH— group, wherein R is the alkyl as defined above. For example, C1-C6 alkylamino may be methylamino, ethylamino, and the like.
“Dialkylamino” refers to the RR′N— group, wherein R and R′ are each the alkyl as defined above. For example, di(C1-C6)alkylamino may be dimethylamino, methylethylamino, and the like.
“Ureido” means the —NH—C(═O)—NH2 group, which is optionally substituted with one or more of the substituents described herein.
“Acyl” refers to the “—C(═O)—” group, which is optionally substituted with any substituent to form, for example, alkylacyl, alkoxyacyl, cycloalkylacyl, heterocyclylacyl, arylacyl, aminoacyl, etc., e.g., C1-C6 alkylacyl, C1-C6 alkoxyacyl, cycloalkylacyl having 3-10 ring atoms, heterocyclylacyl having 4-10 ring atoms, arylacyl having 6-10 ring atoms, etc., wherein the alkyl, alkoxy, cycloalkyl, heterocyclyl, and aryl are as defined above. For example, non-limiting examples of C1-C6 alkylacyl include formyl, acetyl, 3-methylpentanoyl, and the like.
The compounds disclosed herein include isotopic derivatives thereof. The term “isotopic derivative” refers to compounds that differ in structure only by having one or more enriched isotopic atoms. For example, compounds with the structure disclosed herein having “deuterium” or “tritium” in place of hydrogen, or 18F-fluorine labeling (18F isotope) in place of fluorine, or 11C—, 13C- or 14C-enriched carbon (11C—, 13C- or 14C-carbon labeling; 11C—, 13C— or 14C-isotope) in place of a carbon atom are within the scope of the present disclosure. Such a compound can be used as an analytical tool or a probe in, for example, a biological assay, or may be used as a tracer for in vivo diagnostic imaging of disease, or as a tracer in a pharmacodynamic, pharmacokinetic, or receptor study. The various deuterated forms of the compound of the present disclosure mean that each available hydrogen atom connected to a carbon atom may be independently replaced with a deuterium atom. Those skilled in the art can synthesize the compounds in deuterated form by reference to the relevant literature. Commercially available deuterated starting materials can be used in preparing the deuterated compounds, or they can be synthesized using conventional techniques with deuterated reagents including, but not limited to, deuterated borane, tri-deuterated borane in tetrahydrofuran, deuterated lithium aluminum hydride, deuterated iodoethane, deuterated iodomethane, and the like. Deuterides can generally retain comparable activity to non-deuterated compounds and can achieve better metabolic stability when deuterated at certain specific sites, thereby achieving certain therapeutic advantages.
“Optional” or “optionally” means that the event or circumstance subsequently described may, but does not necessarily, occur, and that the description includes instances where the event or circumstance occurs or does not occur. For example, “heterocyclyl group optionally substituted with alkyl” means that alkyl may be, but not necessarily, present, and that the description includes instances where the heterocyclyl group is or is not substituted with alkyl.
“Each independently” means that at least two groups (or ring systems) with the same or similar value ranges in the structure may have the same or different meanings under a certain circumstance. For example, X and Y are each independently hydrogen, halogen, hydroxyl, cyano, alkyl or aryl, meaning that when X is hydrogen, Y may be hydrogen or halogen, hydroxyl, cyano, alkyl or aryl; similarly, when Y is hydrogen, X may be hydrogen or halogen, hydroxyl, cyano, alkyl or aryl.
“Substituted” means that one or more, preferably 1-5, more preferably 1-3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. Those skilled in the art are able to determine (experimentally or theoretically) possible or impossible substitution without undue effort. For example, it may be unstable when amino or hydroxyl having a free hydrogen is bound to a carbon atom having an unsaturated (such as olefin) bond.
For drugs or pharmacologically active agents, the term “therapeutically effective amount” refers to an amount of a medicament or an agent that is sufficient to provide the desired effect but is non-toxic. The determination of the effective amount varies from person to person. It depends on the age and general condition of a subject, as well as the particular active substance used. The appropriate effective amount in a case may be determined by those skilled in the art in the light of routine tests.
“Pharmaceutical composition” refers to a mixture containing one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or pro-drug thereof, and other chemical components, for example, physiologically/pharmaceutically acceptable carriers and excipients. The pharmaceutical composition is intended to promote administration to an organism and facilitate the absorption of the active ingredient so that it can exert its biological activity.
“Pharmaceutically acceptable salt” refers to the salts of the compound of the present disclosure, which are safe and effective for use in the body of a mammal and possess the requisite biological activities. The salts may be prepared separately during the final separation and purification of the compound, or by reacting an appropriate group with an appropriate base or acid. Bases commonly used to form pharmaceutically acceptable salts include inorganic bases such as sodium hydroxide and potassium hydroxide, and organic bases such as ammonia. Acids commonly used to form pharmaceutically acceptable salts include inorganic acids and organic acids.
“Solvate” used herein refers to a substance formed by the physical association of the compound of the present disclosure to one or more, preferably 1 to 3, solvent molecules, whether organic or inorganic. The physical association includes hydrogen bonding. In certain cases, e.g., when one or more, preferably 1 to 3, solvent molecules are incorporated in the crystal lattice of the crystalline solid, the solvate will be isolated. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
“Hydrate” refers to a substance formed by the binding of the compounds of the present invention or the pharmaceutically acceptable salt thereof to water through non-covalent intermolecular forces. Common hydrates include, but are not limited to, hemihydrate, monohydrate, dihydrate, trihydrate, etc.
“Prodrug” refers to a substance that can be converted in vivo under physiological conditions, e.g., by hydrolysis in blood, to generate the active parent drug compound.
“Pharmaceutically acceptable” means that those compounds, materials, compositions, and/or dosage forms that are, within the scope of reasonable medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic reaction, or other problems or complications, and are commensurate with a reasonable benefit/risk ratio and effective for the intended use.
The “stereoisomer” refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereoisomers, conformers (rotamers), geometric isomers (cis/trans isomers), atropisomers, and the like.
“Tautomer” refers to isomers of a compound that differ from each other in proton position and/or electron distribution. Examples of tautomers include, but are not limited to, enol-keto tautomers, imine-enamine tautomers, amide-imidic acid tautomers, amine-imine tautomers, and tautomeric forms of heteroaryl, wherein the heteroaryl comprises a ring atom connected to an —NH-moiety of the ring and an ═N— moiety of the ring, such as pyrazole, imidazole, benzimidazole, triazole, pyridotriazole, piperidinotriazole, and tetrazole.
“In vitro” refers to a biological entity, a biological process, or a biological reaction under artificial conditions outside the body. By way of example, cells grown in vitro are understood to be cells grown in an environment outside the body, for example in test tubes, culture plates or microtiter plates.
In one aspect, the present application provides a compound of formula (I) or a stereoisomer, a tautomer, a solvate, a hydrate, a prodrug, a stable isotopic derivative, or a pharmaceutically acceptable salt thereof,
In some embodiments, B is 6-membered heteroaryl.
In some embodiments, B is selected from:
wherein the dashed line indicates where B is connected to the parent structure.
In some embodiments, B is 5-membered aryl or heteroaryl.
In some embodiments, B is 5-membered heteroaryl comprising at least two heteroatoms selected from N, O, and S.
In some embodiments, B is optionally substituted with one or more R2, wherein each R2 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; preferably, R2 is selected from hydrogen, halogen, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C2-C4 alkynyl, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; more preferably, R2 is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyano, methoxy, and oxo.
In some embodiments, two adjacent R2 form a 5-6 membered carbocyclic ring, heterocyclic ring, or heteroaromatic ring, preferably 5-6 membered heterocyclic ring or heteroaromatic ring, and more preferably
wherein the dashed line indicates where the ring is connected to the parent structure.
In some embodiments, R2 or the ring structure formed by two adjacent R2 is further substituted with one or more R2a, wherein each R2a is independently selected from halogen, C1-C6 alkyl, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, amino, heteroaryl having 5-10 ring atoms, cycloalkyl having 3-10 ring atoms, aryl having 6-10 ring atoms, C1-C6 alkylamino, di(C1-C6)alkylamino, hydroxyl, sulfonyl, C1-C6 alkoxy, cyano, C2-C6 alkenyl, C2-C6 alkynyl, and oxo; preferably, each R2a is independently selected from halogen, C1-C4 alkyl, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, amino, heteroaryl having 5-6 ring atoms, cycloalkyl having 3-6 ring atoms, aryl having 6-10 ring atoms, C1-C3 alkylamino, di(C1-C3)alkylamino, hydroxyl, sulfonyl, C1-C4 alkoxy, cyano, C2-C4 alkenyl, C2-C4 alkynyl, and oxo; more preferably, each R2a is independently selected from methyl, chlorine, fluorine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, pyrazolyl, and pyrimidinyl; most preferably, each R2a is independently selected from methyl, chlorine, fluorine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, and pyrazolyl.
In some embodiments, R2a is further substituted with one or more R2b, wherein each R2b is independently selected from C1-C3 alkyl substituted with 0-3 halogens, C1-C3 alkoxy substituted with 0-3 halogens, halogen, hydroxyl, unsubstituted 5-6 membered heterocyclyl or 5-6 membered heterocyclyl substituted with C1-C3 alkyl, cyano, and oxo, preferably C1-C3 alkyl, C1-C3 alkoxy, halogen, hydroxyl, and oxo; more preferably, each R2b is independently selected from methoxy and methyl.
In some embodiments, each R2b is selected from piperazinyl substituted with alkyl or unsubstituted piperazinyl, preferably piperazinyl substituted with C1-C3 alkyl.
In some embodiments, each R2b is independently selected from methoxy, methyl, trifluoromethyl, difluoromethyl, difluoromethoxy, N-methylpiperazinyl, and cyano.
In some embodiments, R3 is selected from hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, sulfonyl, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, heterocyclyl having 5-10 ring atoms, heterocyclyloxy having 5-10 ring atoms, and cycloalkyloxy having 3-10 ring atoms; preferably, R3 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, halogen, cycloalkyl having 3-6 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, and cycloalkyloxy having 3-6 ring atoms; more preferably, R3 is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, propoxy, and piperidinyloxy.
In some embodiments, R3 is further substituted with one or more R3a, wherein each R3a is independently selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 alkylamino, di(C1-C6 alkyl)amino, halogen, sulfonyl, cycloalkyl having 3-10 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 5-10 ring atoms, and heterocyclyloxy having 5-10 ring atoms; preferably, R3a is each independently selected from C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C3 alkoxy, C1-C3 alkylamino, di(C1-C3 alkyl)amino, halogen, sulfonyl, cycloalkyl having 3-6 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 5-6 ring atoms, and heterocyclyloxy having 5-6 ring atoms; more preferably, R3a is each independently selected from methyl and dimethylamino.
In some embodiments, R3a is further substituted with one or more R3b, wherein each R3b is independently selected from C1-C3 alkyl, halogen, hydroxyl, C1-C3 alkoxy, and amino.
In some embodiments, R4 is selected from hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 alkylthio, sulfonyl, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, and acyl; preferably, R4 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, C1-C4 alkylthio, sulfonyl, halogen, cycloalkyl having 3-6 ring atoms, cyano, amino, and acyl; more preferably, R4 is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, fluorine, chlorine, bromine, and cyano.
In some embodiments, R4 is further substituted with one or more R4a, wherein each R4a is independently selected from C1-C6 alkyl, halogen, amino, C1-C6 alkylamino, and di(C1-C6)alkylamino; preferably, R4a is each independently selected from C1-C3 alkyl, halogen, amino, C1-C3 alkylamino, and di(C1-C3)alkylamino.
In some embodiments, R5 is selected from halogen, cyano, amino, hydroxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, and heteroaryl having 5-10 ring atoms; preferably, R5 is selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 4-6 ring atoms, and heteroaryl having 5-6 ring atoms; more preferably, R5 is selected from chlorine, fluorine, bromine, iodine, cyano, ethyl, n-propyl, isopropyl, methyl, cyclopropyl, methoxy, and cyclopropyloxy.
In some embodiments, R5 is further substituted with one or more R5a, wherein each R5a is independently selected from C1-C6 alkyl, C1-C6 alkoxy, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, and hydroxyl; preferably, R5a is each independently selected from C1-C6 alkyl, halogen, cyano, and cycloalkyl having 3-6 ring atoms; more preferably, R5a is each independently selected from fluorine and cyano.
In some embodiments, R6 is selected from hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 alkylthio, sulfonyl, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, and acyl; preferably, R6 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, C1-C4 alkylthio, sulfonyl, halogen, cycloalkyl having 3-6 ring atoms, cyano, amino, and acyl; more preferably, R6 is selected from hydrogen, fluorine, chlorine, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, methoxy, cyclopropyloxy, and cyano.
In some embodiments, R6 is further substituted with one or more R6a, wherein each R6a is independently selected from C1-C6 alkyl, halogen, amino, C1-C6 alkylamino, and di(C1-C6)alkylamino; preferably, R6a is each independently selected from C1-C3 alkyl, halogen, amino, C1-C3 alkylamino, and di(C1-C3)alkylamino.
In some embodiments, R1 is —X1—R7, wherein X1 is a bond or methylene.
In some embodiments, X1 is substituted with one or more X1a, wherein each X1a is independently selected from halogen and C1-C3 alkyl, preferably methyl.
In some embodiments, R7 is selected from cycloalkyl having 4-10 ring atoms, aryl having 6-12 ring atoms, heteroaryl having 5-12 ring atoms, and heterocyclyl having 3-12 ring atoms; preferably, R7 is selected from the following groups:
In some embodiments, R7 is optionally substituted with one or more R7a, wherein each R7a is independently selected from oxo, halogen, C1-C6 alkyl, cycloalkyl having 3-10 ring atoms, C1-C6 alkoxy, cycloalkyloxy having 3-6 ring atoms, cyano, amino, hydroxyl, C2-C6 alkenyl, C2-C6 alkynyl, sulfonyl, acyl, carboxyl, heterocyclyl having 3-12 ring atoms, heteroaryl having 5-10 ring atoms, and ureido; preferably, R7a is each independently selected from oxo, halogen, C1-C4 alkyl, cycloalkyl having 3-6 ring atoms, C1-C4 alkoxy, cycloalkyloxy having 3-4 ring atoms, cyano, amino, hydroxyl, C2-C4 alkenyl, C2-C4 alkynyl, sulfonyl, acyl, carboxyl, heterocyclyl having 3-6 ring atoms, heteroaryl having 5-6 ring atoms, and ureido; more preferably, R7a is each independently selected from fluorine, chlorine, bromine, hydroxyl, cyano, methoxy, methyl, cyclopropyloxy, and cyclopropyl; most preferably, R7a is each independently selected from fluorine, chlorine, bromine, hydroxyl, cyano, methoxy, methyl, and cyclopropyloxy.
In some embodiments, R7a is further substituted with one or more R7b, wherein R7b is independently selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyl, amino, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkylamino, di(C1-C6)alkylamino, sulfonyl, heterocyclyl having 3-6 ring atoms, and heteroaryl having 5-6 ring atoms; preferably, each R7, is independently selected from halogen, C1-C3 alkyl, C1-C3 alkoxy, hydroxyl, amino, C2-C4 alkenyl, C2-C4 alkynyl, C1-C3 alkylamino, di(C1-C3)alkylamino, sulfonyl, heterocyclyl having 3-6 ring atoms, and heteroaryl having 5-6 ring atoms; more preferably, each R7b is independently selected from fluorine, chlorine, and bromine.
In some embodiments, when X1 is methylene, R7 is not substituted or unsubstituted aryl.
In some embodiments, R1 is selected from the following structures, wherein the dashed line indicates where R1 is connected to the parent structure:
In some embodiments w is CR3, x is CR4, and y is CR5 wherein R3, R4, and R5 are as defined in any one of the embodiments.
In some embodiments, when y is CR5, R5 is not hydrogen.
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, wherein the compound has a structure of formula (IA):
In some embodiments, at least two of m, t, q, v, and u are substituted or unsubstituted heteroatoms; preferably, the heteroatom is N, O, or S.
In some embodiments, when X1 is methylene, R7 is not substituted or unsubstituted aryl.
In some embodiments, w is CR3, x is CR4, and y is CR5, wherein R3, R4, and R5 are as defined in any one of the above embodiments.
In some embodiments, when y is CR5, R5 is not hydrogen.
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, wherein the compound has a structure of formula (IB1), (IB2), (IB3), (IB4), or (IB5):
In some embodiments, in formula (IB1), (IB2), (IB4), or (IB5), at least one of v, u, m, and t is a substituted or unsubstituted heteroatom; preferably, the heteroatom is N, O, or S.
In some embodiments, in formula (IB3), at least two of v, u, m, and t are substituted or unsubstituted heteroatoms; preferably, the heteroatom is N, O, or S.
In some embodiments, when X1 is methylene, R7 is not substituted or unsubstituted aryl.
In some embodiments, w is CR3, x is CR4, and y is CR5, wherein R3, R4, and R5 are as defined in any one of the above embodiments.
In some embodiments, when y is CR5, R5 is not hydrogen.
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, wherein the compound has a structure of formula (IC1), (IC2), (IC3), (IC4), or (IC5):
In some embodiments, in formula (IC1), (IC2), (IC4), or (IC5), at least one of v, u, m, and t is a substituted or unsubstituted heteroatom; preferably, the heteroatom is N, O, or S.
In some embodiments, in formula (IC3), at least two of v, u, m, and t are substituted or unsubstituted heteroatoms; preferably, the heteroatom is N, O, or S.
In some embodiments, when X1 is methylene, R7 is not substituted or unsubstituted aryl.
In some embodiments, w is CR3, and x is CR4, wherein R3 and R4 are as defined in any one of the above embodiments.
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, wherein ring B of the compound is selected from the following structures:
In some embodiments, the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, wherein the compound is selected from the following structures:
In some embodiments, when X1 is methylene, R7 is not substituted or unsubstituted aryl.
In some embodiments, w is CR3, and x is CR4, wherein R3 and R4 are as defined in any one of the above embodiments.
In some embodiments, R2, R8, and R9 according to any one of the above are each independently selected from hydrogen, halogen, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkyloxy, heterocyclyl, heterocyclyloxy, aryl, heteroaryl, heteroaryloxy, cyano, amino, acyl, sulfonyl, and oxo.
In some embodiments, R2 according to any one of the above is selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; preferably, R2 is selected from hydrogen, halogen, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C2-C4 alkynyl, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; more preferably, R2 is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyano, methoxy, and oxo.
In some embodiments, R8 and R9 according to any one of the above are each independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; preferably, R8 and R9 are each independently selected from hydrogen, halogen, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C2-C4 alkynyl, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, aryl having 6 ring atoms, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, cyano, amino, acyl, sulfonyl, and oxo; more preferably, R8 and R9 are each independently selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyano, methoxy, oxo, cyclopropyl, tetrahydropyrrolyl, tetrahydrofuranyl, phenyl, and propenyl.
In some embodiments, adjacent R8 and R2 form a 3-7 membered carbocyclic ring, heterocyclic ring, aromatic ring, or heteroaromatic ring, preferably 5-6 membered carbocyclic ring, heterocyclic ring, or heteroaromatic ring, and more preferably 5-6 membered heterocyclic ring or heteroaromatic ring.
In some embodiments, adjacent R8 and R9 form a 3-7 membered carbocyclic ring, heterocyclic ring, aromatic ring, or heteroaromatic ring, preferably 5-6 membered carbocyclic ring, heterocyclic ring, or heteroaromatic ring, and more preferably 5-6 membered heterocyclic ring or heteroaromatic ring, for example
wherein the dashed line indicates where the ring is connected to the parent structure.
In some embodiments, R2 is further substituted with one or more R2a.
In some embodiments, R8 is further substituted with one or more R8a.
In some embodiments, R9 is further substituted with one or more R9a.
In some embodiments, the ring formed by adjacent R8 and R2 is further substituted with one or more R2a or R8a.
In some embodiments, the ring formed by adjacent R8 and R9 is further substituted with one or more R9a or R8a.
In some embodiments, R2a, R9a, or R8a according to any one of the above is each independently selected from halogen, alkyl, heterocyclyl, heterocyclyloxy, amino, heteroaryl, acyl, cycloalkyl, aryl, alkylamino, dialkylamino, hydroxyl, sulfonyl, alkoxy, cyano, alkenyl, alkynyl, and oxo.
In some embodiments, each R2a is independently selected from halogen, C1-C6 alkyl, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, amino, heteroaryl having 5-10 ring atoms, cycloalkyl having 3-10 ring atoms, aryl having 6-10 ring atoms, C1-C6 alkylamino, di(C1-C6)alkylamino, hydroxyl, sulfonyl, C1-C6 alkoxy, cyano, C2-C6 alkenyl, C2-C6 alkynyl, and oxo; preferably, each R2a is independently selected from halogen, C1-C4 alkyl, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, amino, heteroaryl having 5-6 ring atoms, cycloalkyl having 3-6 ring atoms, aryl having 6-10 ring atoms, C1-C3 alkylamino, di(C1-C3)alkylamino, hydroxyl, sulfonyl, C1-C4 alkoxy, cyano, C2-C4 alkenyl, C2-C4 alkynyl, and oxo; more preferably, each R2a is independently selected from methyl, chlorine, fluorine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, pyrazolyl, and pyrimidinyl; most preferably, each R2a is independently selected from methyl, chlorine, fluorine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, and pyrazolyl.
In some embodiments, R8a or R9a is each independently selected from halogen, C1-C6 alkyl, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, amino, heteroaryl having 5-10 ring atoms, cycloalkyl having 3-10 ring atoms, aryl having 6-10 ring atoms, C1-C6 alkylamino, di(C1-C6)alkylamino, hydroxyl, sulfonyl, C1-C6 alkoxy, cyano, C2-C6 alkenyl, C2-C6 alkynyl, and oxo; preferably, each R8a or each R9a is independently selected from halogen, C1-C4 alkyl, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, amino, heteroaryl having 5-6 ring atoms, cycloalkyl having 3-6 ring atoms, aryl having 6 ring atoms, C1-C3 alkylamino, di(C1-C3)alkylamino, hydroxyl, sulfonyl, C1-C4 alkoxy, cyano, C2-C4 alkenyl, C2-C4 alkynyl, and oxo; more preferably, each R8a or each R9a is independently selected from methyl, chlorine, fluorine, bromine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, pyrazolyl, sulfonyl, morpholinyl, tetrahydrofuranyl, isoxazolyl, and pyrimidinyl; most preferably, each R8a or each R9a is independently selected from methyl, chlorine, fluorine, bromine, phenyl, cyclopropyl, oxo, amino, methylamino, dimethylamino, methoxy, hydroxyl, pyridinyl, pyrazolyl, sulfonyl, morpholinyl, tetrahydrofuranyl, and isoxazolyl.
In some embodiments, R2a is further substituted with one or more R2b.
In some embodiments, R8a is further substituted with one or more R8b.
In some embodiments, R9a is further substituted with one or more R9b.
In some embodiments, each R2b, each R8b, or each R9b is independently selected from alkyl substituted with 0-3 halogens, alkoxy substituted with 0-3 halogens, halogen, hydroxyl, unsubstituted heterocyclyl or heterocyclyl substituted with alkyl, cyano, and oxo, preferably selected from alkyl, alkoxy, halogen, hydroxyl, and oxo.
In some embodiments, each R2b is independently selected from C1-C3 alkyl substituted with 0-3 halogens, C1-C3 alkoxy substituted with 0-3 halogens, halogen, hydroxyl, unsubstituted 5-6 membered heterocyclyl or 5-6 membered heterocyclyl substituted with C1-C3 alkyl, cyano, and oxo, preferably C1-C3 alkyl, C1-C3 alkoxy, halogen, hydroxyl, and oxo; preferably, each R2b is independently selected from methoxy and methyl.
In some embodiments, R8b or R9b is each independently selected from C1-C3 alkyl substituted with 0-3 halogens, C1-C3 alkoxy substituted with 0-3 halogens, halogen, hydroxyl, unsubstituted 5-6 membered heterocyclyl or 5-6 membered heterocyclyl substituted with C1-C3 alkyl, cyano, and oxo, preferably C1-C3 alkyl, C1-C3 alkoxy, halogen, hydroxyl, and oxo; preferably, R8b or R9b is each independently selected from methoxy and methyl.
In some embodiments, R8b or R9b is each independently selected from methoxy, methyl, trifluoromethyl, difluoromethyl, difluoromethoxy, N-methylpiperazinyl, and cyano.
It should be understood that the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof described herein includes, for example, formula (IA), (IB1), (IB2), (IB3), (IB4), (IB5), (IC1), (IC2), (IC3), (IC4), or (IC5), or includes the structure of the following formula:
wherein each substituent definition thereof may optionally be derived from any one of the above embodiments or a combination of the embodiments.
For example, in one embodiment, the compound or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof described herein, wherein:
In one embodiment, the compound or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof described herein, wherein:
In one embodiment, the compound or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof described herein, wherein:
The present application provides a compound of formula (I) or a stereoisomer, a tautomer, a solvate, a hydrate, a prodrug, a stable isotopic derivative, or a pharmaceutically acceptable salt thereof, wherein the compound has optionally the following structures:
In some embodiments, w is CR3, and R3 is selected from hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, sulfonyl, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, heterocyclyl having 5-10 ring atoms, heterocyclyloxy having 5-10 ring atoms, and cycloalkyloxy having 3-10 ring atoms; preferably, R3 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, halogen, cycloalkyl having 3-6 ring atoms, cyano, amino, acyl, hydroxyl, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, and cycloalkyloxy having 3-6 ring atoms; more preferably, R3 is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, propoxy, and piperidinyloxy;
In some embodiments, x is CR4, and R4 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 alkylthio, sulfonyl, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, and acyl; preferably, R4 is selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, C1-C4 alkylthio, sulfonyl, halogen, cycloalkyl having 3-6 ring atoms, cyano, amino, and acyl; more preferably, R4 is selected from methyl, ethyl, n-propyl, isopropyl, fluorine, chlorine, bromine, and cyano;
In some embodiments, R5 is selected from halogen, cyano, amino, hydroxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, and heteroaryl having 5-10 ring atoms; preferably, R5 is selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 4-6 ring atoms, and heteroaryl having 5-6 ring atoms; more preferably, R5 is selected from chlorine, fluorine, bromine, iodine, cyano, ethyl, n-propyl, isopropyl, methyl, cyclopropyl, methoxy, and cyclopropyloxy; optionally, R5 is further substituted with one or more R5a, wherein each R5a is independently selected from C1-C6 alkyl, C1-C6 alkoxy, halogen, cycloalkyl having 3-10 ring atoms, cyano, amino, and hydroxyl; preferably, R5a is each independently selected from C1-C6 alkyl, halogen, cyano, and cycloalkyl having 3-6 ring atoms; more preferably, R5a is each independently selected from fluorine and cyano.
In some embodiments, z is CR6 or N;
In some embodiments, R1 is —X1—R7, wherein X1 is a bond or methylene; optionally, X1 is substituted with one or more X1a, wherein each X1a is independently selected from halogen and C1-C3 alkyl, preferably methyl;
R7 is optionally substituted with one or more R7a, wherein each R7a is independently selected from oxo, halogen, C1-C6 alkyl, cycloalkyl having 3-10 ring atoms, C1-C6 alkoxy, cycloalkyloxy having 3-6 ring atoms, cyano, amino, hydroxyl, C2-C6 alkenyl, C2-C6 alkynyl, sulfonyl, acyl, carboxyl, heterocyclyl having 3-12 ring atoms, heteroaryl having 5-10 ring atoms, and ureido; preferably, R7a is each independently selected from oxo, halogen, C1-C4 alkyl, cycloalkyl having 3-6 ring atoms, C1-C4 alkoxy, cycloalkyloxy having 3-4 ring atoms, cyano, amino, hydroxyl, C2-C4 alkenyl, C2-C4 alkynyl, sulfonyl, acyl, carboxyl, heterocyclyl having 3-6 ring atoms, heteroaryl having 5-6 ring atoms, and ureido; more preferably, R7a is each independently selected from fluorine, chlorine, bromine, hydroxyl, cyano, methoxy, methyl, cyclopropyloxy, and cyclopropyl;
In some embodiments, R8 and R9 are each independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, cycloalkyl having 3-10 ring atoms, cycloalkyloxy having 3-10 ring atoms, heterocyclyl having 4-10 ring atoms, heterocyclyloxy having 4-10 ring atoms, aryl having 6-10 ring atoms, heteroaryl having 5-10 ring atoms, heteroaryloxy having 5-10 ring atoms, cyano, amino, acyl, sulfonyl, and oxo, or adjacent R8 and R9 form a 5-6 membered carbocyclic ring, heterocyclic ring, or heteroaromatic ring; preferably, R8 and R9 are each independently selected from hydrogen, halogen, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C2-C4 alkynyl, cycloalkyl having 3-6 ring atoms, cycloalkyloxy having 3-6 ring atoms, heterocyclyl having 5-6 ring atoms, heterocyclyloxy having 5-6 ring atoms, aryl having 6 ring atoms, heteroaryl having 5-6 ring atoms, heteroaryloxy having 5-6 ring atoms, cyano, amino, acyl, sulfonyl, and oxo, or adjacent R8 and R9 form a 5-6 membered heterocyclic ring or heteroaromatic ring; more preferably, R8 and R9 are each independently selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyano, methoxy, oxo, cyclopropyl, tetrahydropyrrolyl, tetrahydrofuranyl, phenyl, and propenyl, or adjacent R8 and R9 form
In another aspect, the present application provides a compound of formula (I) or a stereoisomer, a tautomer, a solvate, a hydrate, a prodrug, a stable isotopic derivative, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structures:
In another aspect, the present application provides a preparation method for an exemplary compound, comprising the following routes or steps:
In another aspect, the present application provides a composition comprising a therapeutically effective amount of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in inhibiting methionine adenosyltransferase 2A (MAT2A), wherein the inhibitory effect may be in vitro inhibition or in vivo inhibition.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in preparing a drug for inhibiting methionine adenosyltransferase 2A (MAT2A).
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in treating and/or preventing cancer, in particular cancer in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional, including but not limited to colon cancer.
In another aspect, the present application provides use of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above in preparing a drug for treating and/or preventing cancer, in particular cancer in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional.
In another aspect, the present application provides a method for treating and/or preventing cancer, comprising administering to a patient a therapeutically effective amount of the compound of formula (I) or the stereoisomer, the tautomer, the solvate, the hydrate, the prodrug, the stable isotopic derivative, or the pharmaceutically acceptable salt thereof according to any one of the above, or the composition according to any one of the above, wherein the cancer includes those in which a gene encoding methylthioadenosine phosphorylase (MTAP) is deleted and/or is not fully functional.
B2-1 (2 g, 13.19 mmol) and pyridine (1.36 g, 17.15 mmol) were dissolved in diethyl ether (40 mL) under nitrogen atmosphere, ethyl chloroglyoxylate (2.16 g, 15.83 mmol) was added at 0° C., and the mixture was stirred for 2 h. The formed yellow precipitate was filtered off, washed with water, and dried to give a yellow solid (1.5 g, 45%).
B2-2 (1.0 g, 3.97 mmol), ethylhydrazine hydrochloride (0.50 g, 5.16 mmol), and acetic acid (2.38 g, 39.7 mmol) were dispersed in ethanol (20 mL) under nitrogen atmosphere, and the mixture was heated to 100° C. and stirred for reaction for 24 h. The mixture was cooled to room temperature, concentrated under reduced pressure, and separated by column chromatography to give B2-3 (0.3 g, 30.5%) as a brown solid.
B2-3 (0.2 g, 0.81 mmol), iodobenzene (0.20 g, 0.97 mmol), cuprous iodide (0.031 g, 0.16 mmol), 8-hydroxyquinoline (0.047 g, 0.32 mmol), and potassium phosphate (0.34 g, 1.62 mmol) were dispersed in DMF (10 mL) under nitrogen atmosphere, and the mixture was heated to 110° C. and reacted for 18 h. The mixture was cooled to room temperature, diluted with water, and extracted with ethyl acetate. The organic phase was washed twice with saturated ammonium chloride, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, and purified by column chromatography and preparative thin-layer chromatography to give B2 (0.07 g, 27%) as a pale pink solid.
Compound B3 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 3-iodopyridine and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B4 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 3-fluoroiodobenzene and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B5 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 4-fluoroiodobenzene and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B6 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 2-iodopyrazine and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B7 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 2-iodopyrimidine and potassium phosphate replaced by potassium tert-butoxide in step 3.
B2-3 (100 mg, 0.40 mmol), 3-methoxyiodobenzene (377.9 mg, 1.61 mmol), cuprous iodide (76.71 mg, 0.40 mmol), 8-hydroxyquinoline (29.03 mg, 0.20 mmol), and potassium tert-butoxide (112.21 mg, 1.01 mmol) were dispersed in DMSO (15 mL) under nitrogen atmosphere, and the mixture was heated to 135° C. and reacted overnight. The mixture was cooled to room temperature, diluted with water, and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, and purified by column chromatography and preparative thin-layer chromatography to give B8-1 (96.5 mg, 68%) as a brown solid.
B8-1 (60 mg, 0.17 mmol) was dissolved in dichloromethane (10 mL), a 1.0 M solution of boron tribromide in dichloromethane (0.5 mL, 0.5 mmol) was added dropwise under nitrogen atmosphere at −78° C., and the mixture was warmed to room temperature and reacted for 3 h. The mixture was quenched with 1 mL of methanol, extracted 3 times with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, purified by preparative HPLC, and separated to give B8 (32.4 mg, 56%) as a white solid.
Compound B9 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 3-iodobenzonitrile and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B10 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 4-iodopyridine and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B11 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 4-iodoanisole and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B12 was synthesized according to a synthetic route similar to that of Example 1 with ethylhydrazine hydrochloride replaced by methylhydrazine sulfate in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
Compound B28 was synthesized according to a synthetic route similar to that of Example 1 with ethylhydrazine hydrochloride replaced by p-methoxybenzylhydrazine in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
Compound B28 (1.0 g, 2.4 mmol) was mixed with trifluoroacetic acid (20 mL), and the mixture was stirred for reaction at 40° C. for 16 h. After the reaction was completed, the reaction solution was concentrated under reduced pressure, adjusted to pH=9 with a saturated sodium bicarbonate solution, and extracted with ethyl acetate (50 mL×3). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane:methanol=20:1) to give compound B13 (600 mg, 65%) as a yellow solid.
Compound B14 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 5-fluoro-3-iodopyridine and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B15 was synthesized according to a synthetic route similar to that of Example 1 with iodobenzene replaced by 3-iodotrifluorotoluene and potassium phosphate replaced by potassium tert-butoxide in step 3.
Compound B16 was synthesized according to a synthetic route similar to that of Example 1 with B2-1 replaced by
in step 1, ethylhydrazine hydrochloride replaced by methylhydrazine sulfate in step 2, and iodobenzene replaced by 3-iodopyridine in step 3.
Compound B17 was synthesized according to a synthetic route similar to that of Example 1 with B2-1 replaced by
in step 1 and iodobenzene replaced by 3-iodopyridine in step 3.
B18-1 (0.2 g, 1.31 mmol) was dissolved in DMF (2 mL) under an ice-water bath under nitrogen atmosphere, a solution of triethylamine (0.15 g, 1.44 mmol) in DMF (1.0 mL) and a solution of di-tert-butyl dicarbonate (0.31 g, 1.44 mmol) in THF (1.5 mL) were slowly added, and the mixture was warmed to room temperature and reacted overnight. The mixture was diluted with water, and extracted twice with ethyl acetate. The organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, and purified by column chromatography to separately give compounds B18-2A (110 mg, 39%) and B18-2B (90 mg, 32%). B18-2A: 1H NMR (400 MHz, Chloroform-d) δ 8.11 (s, 1H), 7.45 (s, 1H), 4.61 (s, 2H), 1.68 (s, 9H). B18-2B 1H NMR (400 MHz, Chloroform-d) δ 8.11 (s, 1H), 7.45 (s, 1H), 4.61 (s, 2H), 1.68 (s, 9H).
B2-3 (100 mg, 0.40 mmol) was dissolved in DMF (3 mL) under an ice-water bath under nitrogen atmosphere, NaH (20.8 mg, 0.52 mmol, 60% purity) was added, and the mixture was stirred at room temperature for 30 min. B18-2A (87 mg, 0.40 mmol) was dissolved in DMF (2 mL), and the mixture was stirred at room temperature overnight. The mixture was diluted with water, and extracted with ethyl acetate. The organic phase was washed with saturated ammonium chloride, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, and purified by column chromatography to give a pale yellow solid (125 mg, 73%). The resulting solid (100 mg, 0.23 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (1.0 mL, 6.71 mmol) was added under an ice-water bath, and the mixture was stirred at room temperature for 4 h. The mixture was concentrated under reduced pressure and purified by preparative HPLC to give a white solid (43 mg, 57%).
Compound B19 was synthesized according to a synthetic route similar to that of Example 1 with B2-1 replaced by
in step 1 and iodobenzene replaced by 3-iodopyridine in step 3.
Compound B3 (100 mg, 0.31 mmol) and N-chlorosuccinimide (41.04 mg, 0.31 mmol) were separately added to N—N dimethylformamide (6 mL), and the mixture was heated to 60° C. and stirred for reaction overnight. The reaction solution was cooled to room temperature, poured into water, extracted with ethyl acetate, and washed with saturated brine. The separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated. The concentration residue was purified by preparative high performance liquid chromatography to give compound B20 (57.6 mg, 52%) as an off-white solid.
Potassium carbonate (465 mg, 3.37 mmol) was added to compound B13 (400 mg, 1.35 mmol) and iodoacetonitrile (270 mg, 1.62 mmol) in N,N-dimethylformamide (10 mL) at room temperature, and the mixture was stirred at room temperature for reaction overnight. The reaction solution was quenched with water (20 mL) and extracted with ethyl acetate (30 mL×3). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by preparative high performance liquid chromatography to separately give compound B21 (4.1 mg, 0.8%) as a white solid and compound B22 (84.5 mg, 17%) as a white solid.
B23-1 (5 g, 32.15 mmol) and 3-aminopyridine (4.13 g, 32.15 mmol) were dissolved in N,N-dimethylformamide (50 mL), potassium tert-butoxide (7.55 g, 67.5 mmol) was added, and the mixture was reacted at room temperature for 30 min. Water was added, and a solid was precipitated. The mixture was filtered under vacuum. The filter cake was sequentially washed with water, isopropanol, and isopropyl ether, and dried to give a black solid (4.8 g, 60%).
Reduced iron powder (5.4 g, 96.4 mmol) was added to a solution of B23-2 (4.8 g, 19.3 mmol) in acetic acid (50 mL). The mixture was warmed to 80° C. and stirred for 2 h. The mixture was cooled to room temperature, adjusted to pH=8 with saturated NaHCO3, and extracted 2 times with ethyl acetate. The organic phases were combined, dried, and concentrated under reduced pressure to give B23-3 (4 g, 96%).
B23-3 (4.0 g, 18.2 mmol) and diethyl oxalate (20 mL) were mixed, heated, and refluxed overnight. The reaction was completed as monitored by thin-layer chromatography. The mixture was cooled to room temperature and filtered. The filter cake was washed with ethanol and dried to give B23-4 (3 g, 60%).
B23-4 (1 g, 3.65 mmol) was dissolved in DMF (50 mL), DIPEA (1 g, 7.3 mmol) and POCl3 (1.8 g, 11.68 mmol) were added, and the mixture was warmed to 95° C. and reacted for 1 h. The mixture was cooled to room temperature, poured slowly into ice water, and extracted twice with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation to give B23-5 (800 mg, 74.7%) as a yellow solid.
B23-5 (0.2 g, 0.68 mmol) was dissolved in n-butanol (10 mL), acethydrazide (0.06 g, 0.81 mmol) was added, and the mixture was warmed to 120° C. and reacted for 2 h. The mixture was cooled to room temperature, diluted with water, and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness by rotary evaporation, and purified by preparative thin-layer chromatography to give B23 (31 mg, 15%) as a white solid.
1) Synthesis of intermediate B29-2:
B29-1 (500 mg, 2.2 mmol) and 3-aminopyridine (244 mg, 2.6 mmol) were dissolved in N,N-dimethylformamide (10 mL), diisopropylethylamine (568 mg, 4.4 mmol) and HATU (1.2 g, 3.3 mmol) were added, and the mixture was reacted at room temperature for 30 min. The mixture was directly concentrated to dryness by rotary evaporation, and purified by column chromatography to give B29-2 (450 mg, 70%).
B29-2 (450 mg, 1.5 mmol) and 2-chloro-6-trifluoromethylpyridine-3-boronic acid (360 mg, 1.6 mmol) were dissolved in 1,4-dioxane (10 mL), and potassium carbonate (621 mg, 4.5 mmol) and Pd(dppf)Cl2 (110 mg, 0.15 mmol) were added. The mixture was reacted at 100° C. for 2 h under nitrogen atmosphere. The mixture was directly concentrated to dryness by rotary evaporation, and purified by column chromatography to give B29-3 (150 mg, 25%) as a yellow solid.
B29-3 (100 mg, 0.25 mmol) was dissolved in dimethyl sulfoxide (5 mL), DBU (76 mg, 0.05 mmol) was added, and the mixture was warmed to 140° C. and reacted overnight. The reaction solution was separated by reversed-phase preparative chromatography to give B29 (23 mg, 25%) as a brown solid.
Compound B30 was obtained according to a synthetic route similar to that of Example 23 with 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 1 and 2-chloro-6-trifluoromethylpyridine-3-boronic acid replaced by 2-fluoro-4-chlorophenylboronic acid in step 2.
Compound B47 was obtained according to a synthetic route similar to that of Example 1 with ethylhydrazine replaced by 3-pyridinylmethylhydrazine in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
Compound B60 was obtained according to a synthetic route similar to that of Example 23 carboxylic acid and 3-aminopyridine replaced by
in step 1 and 2-chloro-6-trifluoromethylpyridine-3-phenylboronic acid replaced by 2-fluoro-4-chlorophenylboronic acid in step 2 and Boc simultaneously left during the last ring-closing step.
Compound B72 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazol-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1.
Compound B139-1 was obtained according to the preparation method for B29-2 in Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid.
B139-1 (200 mg, 0.7 mmol) and bis(pinacolato)diboron (220 mg, 0.9 mmol) were dissolved in 1,4-dioxane (2 mL), and potassium acetate (210 mg, 2.1 mmol) and Pd(dppf)Cl2 (52 mg, 0.1 mmol) were added. The mixture was stirred at 100° C. overnight under nitrogen atmosphere. The mixture was directly concentrated to dryness by rotary evaporation, and purified by column chromatography to give brown oily B139-2 (230 mg, 86%).
B139-2 (270 mg, 0.82 mmol) and 1-bromo-4-chloro-2,5-difluorobenzene (220 mg, 0.88 mmol) were dissolved in 1,4-dioxane (5 mL) and water (0.5 mL), and potassium carbonate (350 mg, 2.53 mmol) and Pd(dppf)Cl2 (90 mg, 0.12 mmol) were added. The mixture was stirred at 100° C. overnight under nitrogen atmosphere. The mixture was directly concentrated to dryness by rotary evaporation, and purified by column chromatography to give orange oily B139-3 (140 mg, 98%).
B139-3 (140 mg, 0.40 mmol) was dissolved in N,N-dimethylformamide (5 mL), cesium carbonate (390 mg, 1.20 mmol) was added, and the mixture was warmed to 60° C. and stirred for two days. The reaction solution was separated by reversed-phase preparative chromatography to give B139 (46 mg, 34%) as a white solid.
Compound B148 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-aminophenol in step 1.
Compound B171 was obtained according to a synthetic route similar to that of Example 28 with 1-bromo-4-chloro-2,5-difluorobenzene replaced by 1-bromo-4-chloro-2-fluoro-5-methoxybenzene in step 2.
Compound B172 was obtained according to a synthetic route similar to that of Example 28 with 1-bromo-4-chloro-2,5-difluorobenzene replaced by 2-bromo-5-chloro-1,3-difluorobenzene in step 2.
Compound B176 was obtained according to a synthetic route similar to that of Example 28 with 1-bromo-4-chloro-2,5-difluorobenzene replaced by 2-bromo-5-chloro-1-fluoro-3-methoxybenzene in step 2.
Intermediate B178-1 was obtained according to the preparation method for B29 in Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-p-methoxybenzylpyrazole-3-carboxylic acid in step 1.
Trifluoroacetic acid (1.5 mL) was slowly added dropwise to a solution of B178-1 (90 mg, 0.18 mmol) in dichloromethane (5 mL), and the mixture was stirred at room temperature overnight. The reaction solution was separated by reversed-phase preparative chromatography to give B178 (14 mg, 20%) as a white solid.
Compound B76 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-2-methylpyridine in step 1.
B76 was subjected to chiral resolution (IC-3.0 cm, n-hexane:ethanol=30:70, 25 mL/min, 230 nm) to give B179 (retention time: 8.11 min) and B180 (retention time: 14.98 min).
Compound B181 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1 and Boc simultaneously left during the last ring-closing step.
Compound B239 was obtained according to a synthetic route similar to that of Example 33 with 3-aminopyridine replaced by
in step 1.
Compound B245 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-difluoromethylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1 and Boc simultaneously left during the last ring-closing step.
Compound B250 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1 and Boc simultaneously left during the last ring-closing step.
Compound B254 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-methyl-4-bromo-1-difluoromethylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by Boc in step 1
and Boc simultaneously left during the last ring-closing step.
Compound B256 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-trideuteromethylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1 and Boc simultaneously left during the last ring-closing step.
Compound B257 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by
in step 1 and Boc simultaneously left during the last ring-closing step.
Compound B182 was obtained according to a synthetic route similar to that of Example 23 with 3-amino-1-methylpyridine replaced by o-chloroaniline and 4-bromo-1-ethyl-3-carboxypyrazole replaced by 4-bromo-1-methyl-3-carboxypyrazole in step 1.
Compound B183 was obtained according to a synthetic route similar to that of Example 23 with 3-amino-1-methylpyridine replaced by 2-cyclopropyl-3-aminopyridine and 4-bromo-1-ethyl-3-carboxypyrazole replaced by 4-bromo-1-methyl-3-carboxypyrazole in step 1.
B183 was subjected to chiral resolution (REGIS(S,S)WHELK-O1 (250 mm×25 mm, 10 m), methanol=45%, 70 g/min, 4 min) to give B212 (retention time: 1.97 min) and B213 (retention time: 2.35 min).
N-Bromosuccinimide (3.92 g, 22.04 mmol) was added to a solution of B258-1 (3.0 g, 22.04 mmol) in acetic acid (20 mL). The mixture was warmed to 75° C. and stirred for 5 h. The reaction solution was cooled to room temperature and concentrated, and the crude product was separated by column chromatography to give B258-2 (3.5 g, 74%) as a white solid.
B258-2 (500 mg, 2.33 mmol), cuprous bromide (280 mg, 2.80 mmol), and acetonitrile (5 mL) were added to a reaction flask. tert-Butyl nitrite (720 mg, 6.99 mmol) was added at 0° C., and the mixture was stirred for 4 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with water (50 mL). The organic phase was concentrated, and the crude product was purified by column chromatography to give B258-3 (150 mg, 28%) as a white solid product.
Compound B258-4 was obtained according to a synthetic route similar to that for B139-2 in Example 28 with 3-aminopyridine replaced by 1-Boc-4-aminoindazole.
B258-3 (100 mg, 0.43 mmol), B258-4 (150 mg, 0.52 mmol), 1,1′-bis(diphenylphosphino)ferrocenepalladium dichloride (31 mg, 0.04 mmol), potassium carbonate (180 mg, 3.90 mmol), 1,4-dioxane (2 mL), and water (0.2 mL) were added to a reaction flask. The mixture was stirred at 100° C. for 16 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with water (50 mL). The organic phase was concentrated, and the crude product was purified by column chromatography to give B258-5 (100 mg, 59%) as a white solid product.
B258-5 (100 mg, 0.25 mmol), potassium carbonate (96 mg, 0.63 mmol), and N,N-dimethylformamide (1 mL) were added to a reaction flask. The mixture was stirred at 60° C. for 16 h. The reaction solution was diluted with dichloromethane (20 mL) and washed with water (10 mL). The organic phase was concentrated to give B258-6 (70 mg, 74%) as a brown oily substance.
B258-6 (70 mg, 0.064 mmol), trifluoroacetic acid (0.5 mL), and dichloromethane (1 mL) were added to a reaction flask. The mixture was stirred at room temperature for 2 h. The reaction solution was diluted with dichloromethane (20 mL) and washed with water (10 mL). The organic phase was concentrated, and the crude product was purified by reversed-phase column chromatography to give B258 (12 mg, 20%) as a white solid.
Compound B260 was obtained according to a synthetic route similar to that of Example 28 with 3-amino-1-methylpyridine replaced by N-Boc-4-aminoindazole and 4-bromo-1-ethyl-3-carboxypyrazole replaced by 4-bromo-1-difluoromethyl-3-carboxypyrazole in step 1.
Compound B273 was obtained according to a synthetic route similar to that of Example 45 with B258-3 replaced by 2-bromo-5-chloro-3-fluoropyridine.
Lithium diisopropylamide/tetrahydrofuran (0.5 mL, 1 mmol, 2 mol/L) was added to a solution of B274-1 (100 mg, 0.5 mmol) in tetrahydrofuran (3 mL) at −78° C., meanwhile the solution being stirred. After the mixture was reacted at −78° C. for 10 min, iodine (150 mg, 0.6 mmol) was added, and the mixture was warmed to room temperature and reacted for 2 h. Water (10 mL) was added, and the mixture was extracted twice with dichloromethane (10 mL). The organic phases were combined and concentrated to dryness by rotary evaporation to remove the solvent, thus giving crude B274-2 (150 mg, yield: 100%), which was directly used in the next step.
Compound B274 was obtained according to a synthetic route similar to that of Example 45 with B258-3 replaced by B274-2.
1) Synthesis of product B332:
NBS (30 mg, 1.68 mmol) was added to a solution of B60 (50 mg, 0.14 mmol) in DMF (1 mL), meanwhile the solution being stirred. The reaction system was reacted at room temperature for 1 h. The reaction solution was directly purified by reversed-phase chromatographic column to give B332 (13 mg, 22%) as a white solid.
[1,1′-bis(Diphenylphosphino)ferrocene]palladium dichloride (8 mg, 0.01 mmol) and a dimethylzinc/toluene solution (0.2 mL, 0.2 mmol, 1 mol/L) were added to a solution of B332 (50 mg, 0.117 mmol) in 1,4-dioxane (1 mL), meanwhile the solution being stirred. The mixture was warmed to 100° C. and reacted at room temperature for 3 h. The reaction was quenched with water. The mixture was concentrated to dryness by rotary evaporation to remove the solvent and purified by thin layer chromatography to give B227 as a white solid (6.2 mg, 11%).
Sodium trifluoromethanesulfonate (274 mg, 2 mmol) was added to a solution of B3 (310 mg, 1 mmol) and TBHP (180 mg, 2 mmol) in DMSO (5 mL). The mixture was heated to 80° C. and stirred for 72 h under nitrogen atmosphere. The reaction solution was cooled to room temperature and directly separated and purified by reversed-phase preparative chromatography to give B24 (35 mg, 9%) as a white solid.
Sodium difluoromethanesulfonate (274 mg, 2 mmol) was added to a solution of B3 (310 mg, 1 mmol) and TBHP (180 mg, 2 mmol) in DMSO (5 mL). The mixture was heated to 80° C. and stirred for 72 h under nitrogen atmosphere. The reaction solution was cooled to room temperature and directly separated and purified by reversed-phase preparative chromatography to give B25 (15 mg, 4%) as a white solid.
Compound B26 was obtained according to a synthetic route similar to that of Example 50 with B3 replaced by B12 in step 1.
Compound B27 was obtained according to a synthetic route similar to that of Example 51 with B3 replaced by B12 in step 1.
Compound B339 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 2-trifluoromethyl-3-aminopyridine and 4-bromo-1-methyl-3-carboxypyrazole replaced by 4-bromo-1-difluoromethyl-3-carboxypyrazole in step 1.
Compound B337 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 2-cyclopropyl-3-aminopyridine and 4-bromo-1-methyl-3-carboxypyrazole replaced by 4-bromo-1-difluoromethyl-3-carboxypyrazole in step 1.
B337 was subjected to chiral resolution (IB-3.0 cm, n-hexane:ethanol=40:60, 25 mL/min, 214 nm) to give B398 (retention time: 10.2 min) and B399 (retention time: 13.9 min).
A solution of B31-1 (2.32 g, 10 mmol), B31-2 (1.74 g, 10 mmol), potassium carbonate (4.14 g, 30 mmol), and Pd(dppf)Cl2 (0.73 g, 1 mmol) in dioxane (50 mL) was heated and stirred at 100° C. for 16 h under nitrogen atmosphere. The reaction solution was cooled to room temperature and directly purified by column chromatography to give B31-3 (1.2 g, 42%) as a brown solid.
Lithium hydroxide (115 mg, 48 mmol) was added to a mixture of B31-3 (1.13 g, 40 mmol) in tetrahydrofuran (15 mL)/water (20 mL). The mixture was stirred at room temperature for 0.5 h. The reaction solution was concentrated to dryness, thus giving a crude product B31-4, which was directly used in the next step.
Diisopropylethylamine (387 mg, 3 mmol) and HATU (456 mg, 1.2 mmol) were added to a solution of B31-4 (268 mg, 1 mmol) and B31-5 (122 mg, 1 mmol) in DMF (5 mL). The mixture was stirred at 40° C. for 3 h. The reaction solution was directly purified by column chromatography to give B31-6 (130 mg, 36%) as a brown oily substance.
A mixture of B31-6 (71 mg, 0.2 mmol) and K2CO3 (27 mg, 0.6 mmol) in DMF (3 mL) was stirred at 60° C. for 16 h. The reaction solution was cooled to room temperature and directly separated by column chromatography to give B31 (50 mg, 73%) as a white solid.
Compound B32 was obtained according to a synthetic route similar to that of Example 56 with 2,4-dimethyl-3-aminopyridine replaced by 2-trifluoromethyl-3-aminopyridine in step 3.
A solution of B31-1 (5.0 g, 21.4 mmol) and Select-F (30.3 g, 85.8 mmol) in acetonitrile (100 mL) was heated to 80° C. and stirred for reaction for 4 days under nitrogen atmosphere. The reaction solution was washed with a saturated aqueous sodium bicarbonate solution and extracted with ethyl acetate. The organic phase was concentrated, and separated and purified by column chromatography to give B33-1 (1.73 g, 32%) as a white solid.
Compound B33 was obtained according to a synthetic route similar to that of Example 56 with B31-1 replaced by B33-1 and 2,4-dimethyl-3-aminopyridine replaced by 3-aminopyridine.
Compound B34 was obtained according to a synthetic route similar to that of Example 56 with B31-1 replaced by methyl 4-bromo-2-methyloxazole-5-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 3-aminopyridine in step 3.
Compound B35 was obtained according to a synthetic route similar to that of Example 56 with B31-1 replaced by methyl 4-bromo-2-cyclopropylmethyl-5-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 3-aminopyridine in step 3.
Potassium carbonate (836 mg, 6.06 mmol) was added to a solution of B13 (600 mg, 2.02 mmol) and ethyl bromoacetate (405 mg, 2.42 mmol) in DMF (15 mL), and the mixture was stirred at room temperature for 16 h. The mixture was washed with water, extracted with ethyl acetate, concentrated, and purified by reversed-phase preparative chromatography to give B36-1 (200 mg, 19%) as a yellow solid.
Lithium hydroxide monohydrate (69 mg, 1.65 mmol) was added to a mixture of B36-1 (210 mg, 0.55 mmol) in tetrahydrofuran (15 mL)/water (2 mL), and the mixture was stirred at room temperature for 16 h. The mixture was neutralized to weak acidity with diluted hydrochloric acid (1 M) and extracted with dichloromethane. The organic phase was concentrated to dryness, thus giving B36-2 (200 mg, 70%) as a yellow solid.
Potassium carbonate (58 mg, 0.42 mmol) and HATU (80 mg, 0.21 mmol) were added to a solution of B36-2 (50 mg, 0.14 mmol) and ammonium chloride (11 mg, 0.21 mmol) in DMF (5 mL). The mixture was stirred at room temperature for 16 h. The mixture was treated with water and extracted with ethyl acetate. The organic phase was concentrated, and purified by reversed-phase preparative chromatography to give B36 (4.0 mg, 6%) as a white solid. Compounds B37 and B38 were separately obtained according to a preparation method similar to that for B36 with ammonium chloride replaced by methylamine hydrochloride or dimethylamine hydrochloride.
Compound B39 was obtained according to a synthetic route similar to that of Example 1 with ethylhydrazine replaced by isopropylhydrazine in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
Potassium carbonate (279 mg, 2.02 mmol) was added to a solution of B13 (200 mg, 0.67 mmol) and 1,1-difluoro-2-iodoethane in DMF (8 mL). The reaction solution was stirred at room temperature for 16 h, treated with water, and extracted with ethyl acetate. The organic phase was concentrated, and separated by reversed-phase preparative chromatography to give B40 (18 mg, 7%) and B170 (69 mg, 27%) as white solids.
Compound B41 was obtained according to a synthetic route similar to that of Example 1 with ethylhydrazine replaced by trifluoroethylhydrazine in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
Compounds B42 and B167 were obtained according to a synthetic route similar to that of Example 63 with 1,1-difluoro-2-iodoethane replaced by N,N-dimethyl-3-bromopropylamine in step 2.
Compound B43 was obtained according to a synthetic route similar to that of Example 63 with 1,1-difluoro-2-iodoethane replaced by N,N-dimethyl-2-bromoethylamine in step 2.
Compound B46 was obtained according to a synthetic route similar to that of Example 1 with ethylhydrazine replaced by hydroxyethylhydrazine in step 2 and iodobenzene replaced by 3-iodopyridine in step 3.
NaH (12 mg, 60%, 0.3 mmol) was added to a solution of B46 (100 mg, 0.3 mmol) in DMF (15 mL) at 0° C., and the mixture was stirred at room temperature for 20 min. Iodomethane (43 mg, 0.3 mmol) was added, and the mixture was stirred at room temperature for 2 h. The mixture was treated with saturated brine and extracted with ethyl acetate. The organic phase was concentrated and separated by column chromatography to give B45 (38 mg, 30%).
Compound B48 and isomer B168 were obtained according to a synthetic route similar to that of Example 63 with 1,1-difluoro-2-iodoethane replaced by (1-methyl-1H-pyrazol-3-yl)methyl methanesulfonate in step 2.
Palladium acetate (80 mg, 0.36 mmol) and 2-dicyclohexylphosphino-2′,6′-dimethoxybiphenyl (29 mg, 0.72 mmol) were added to B50-1 (1 g, 7.17 mmol), B31-2 (1.38 g, 7.89 mmol), and sodium carbonate (3 g, 28.68 mmol) in a mixed solvent of ethylene glycol dimethyl ether (8 mL) and water (4 mL). The reaction solution was stirred at 80° C. for 1 h under nitrogen atmosphere. Water was added, and the mixture was extracted with ethyl acetate. The organic phase was concentrated and separated by column chromatography to give B50-2 (1.2 g, 72%) as a yellow solid.
Potassium hydroxide (0.6 g, 10.7 mmol) was added to a suspension of B50-2 (500 mg, 2.14 mmol) in tert-butanol (20 mL). The mixture was stirred at 120° C. for 1.5 h. After the reaction solution was cooled to room temperature, 20 mL of water was added, and the mixture was acidified with 6 N hydrochloric acid to give a suspension, which was filtered and dried under vacuum to give B50-3 (420 mg, 85%) as a pale yellow solid.
Copper acetate (0.39 g, 2.16 mmol) was added to a solution of B50-3 (250 mg, 1.08 mmol) and pyridine-3-boronic acid (660 mg, 5.4 mmol) in pyridine (15 mL). The reaction solution was stirred at 100° C. for 16 h open-to-air. Water was added, and the mixture was extracted with ethyl acetate. The organic phase was concentrated and separated by column chromatography to give B50 (7 mg, 2%) as a white solid.
Compound B51 was obtained according to a synthetic route similar to that of Example 56 with 2,4-dimethyl-3-aminopyridine replaced by 4-methyl-3-aminopyridine in step 3.
Potassium fluoride (78 mg, 1.35 mmol) was added to a mixture of B13 (200 mg, 0.67 mmol) and diethyl monobromodifluoromethylphosphate (180 mg, 0.67 mmol) in acetonitrile (10 mL), and the mixture was stirred at room temperature for 16 h. The reaction solution was treated with water and extracted with ethyl acetate. The organic phase was concentrated and separated by reversed-phase preparative chromatography to give B55 (46 mg, 18%) as a yellow solid.
Compound B57 was obtained according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 5-aminoindazole in step 3.
Compound B58 was obtained according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 1-methyl-5-aminopyrazole in step 3.
Compound B65 was obtained according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 3.
Compound B68 was obtained according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 2-trifluoromethyl-3-aminopyridine in step 3.
Compound B69 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2-chloro-6-trifluoromethylpyridine-3-boronic acid replaced by 2-fluoro-4-trifluoromethylphenylboronic acid in step 2.
Compound B70-1 was obtained according to the preparation method for compound B31-3 in Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate.
Water (0.3 mL), potassium phosphate (1.05 g, 4.97 mmol), tricyclohexylphosphorus (0.48 M toluene solution, 0.295 mL, 0.142 mmol), and palladium acetate (16 mg, 0.071 mmol) were sequentially added to a solution of B70-1 (300 mg, 1.42 mmol) in toluene (6 mL). The mixture was stirred at 100° C. for 4 h under nitrogen atmosphere. The mixture was cooled. Water was added. The mixture was extracted with ethyl acetate. The organic phase was concentrated and separated by column chromatography to give B70-2 (211 mg, 86%) as a yellow solid.
Compound B70 was obtained according to a synthetic route similar to that of Example 56 with B31-3 replaced by B70-2 and B31-5 replaced by 3-aminopyridine.
A mixture of B71-1 (5.0 g, 21 mmol), cyclopropylboronic acid (1.97 g, 23 mmol), Pd(dppf)Cl2 (1.54 g, 2.1 mmol), potassium phosphate (11.1 g, 53 mmol), and tricyclohexylphosphorus (1.18 g, 4.2 mmol) in dioxane (100 mL) was heated to 100° C. and stirred for 17 h under nitrogen atmosphere. The mixture was treated with water and extracted with ethyl acetate. The organic phase was concentrated and separated by column chromatography to give B71-2 (2.52 g, 60%) as a yellow solid.
m-Chloroperoxybenzoic acid (5.16 g, 30 mmol) was added to a solution of B71-2 (2.97 g, 15 mmol) in dichloromethane (60 mL), and the mixture was stirred for 17 h. The reaction solution was washed with a saturated aqueous sodium sulfite solution, concentrated, and separated and purified by column chromatography to give B71-3 (1.65 g, 51%) as a yellow solid.
B71-3 (1.65 g, 7.7 mmol) was heated to 90° C. in phosphorus oxychloride (20 mL), and the mixture was stirred for 3 h. The phosphorus oxychloride was removed by distillation under reduced pressure. The residue was poured into ice water, treated with a saturated aqueous sodium bicarbonate solution to pH ≈8, and extracted with dichloromethane. The organic phase was dried, concentrated, and separated and purified by column chromatography to give B71-4 (1.2 g, 67%) as a yellow solid.
A mixture of B71-4 (276 mg, 1.2 mmol), B71-5 (480 mg, 1.8 mmol), Pd(dppf)Cl2 (90 mg, 0.12 mmol), and potassium carbonate (498 mg, 3.6 mmol) in dioxane (10 mL)/water (1 mL) was heated to 90° C. and stirred for 2 h under nitrogen atmosphere. The reaction solution was treated with water, extracted with ethyl acetate, concentrated, and separated by column chromatography to give B71-6 (260 mg, 74%) as a yellow solid.
Compound B71 was obtained according to a synthetic route similar to that of Example 56 with B31-3 replaced by B71-6 and B31-5 replaced by 3-aminopyridine.
Compound B74 was obtained according to a synthetic route similar to that of Example 78 with 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 3.
Compound B75 was obtained according to a synthetic route similar to that of Example 79 with 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 6.
Compound B77 was synthesized according to a synthetic route similar to that of Example 79 with B71-4 replaced by 1-bromo-2-fluoro-4-trifluoromethoxybenzene in step 4 and 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 6.
Compound B78 was synthesized according to a synthetic route similar to that of Example 79 with B71-4 replaced by 1-bromo-2-fluoro-4-trifluoromethoxybenzene in step 4.
Compound B80 was synthesized according to a synthetic route similar to that of Example 23 with B29-1 replaced by 4-bromo-1-methyl-pyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-trifluoromethyl-3-aminopyridine in step 1.
Intermediate B95-1 was obtained according to the synthetic route of Example 79 with methyl 4-boronic acid-1-methylpyrazole-3-carboxylate replaced by methyl 4-boronic acid-1-p-methoxybenzylpyrazole-3-carboxylate in step 4 and 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 6.
B95 was obtained according to the synthetic route of Example 33 with B178-1 replaced by B95-1.
Compound B96 was synthesized according to a synthetic route similar to that of Example 33 with 3-aminopyridine replaced by 2-methyl-3-aminopyridine.
Compound B99 was synthesized according to a synthetic route similar to that of Example 85 with 2-methyl-3-aminopyridine replaced by 3-aminopyridine.
Compound B101-1 was synthesized according to a synthetic route similar to that of Example 79 with B71-4 replaced by 1-bromo-2-fluoro-4-trifluoromethoxybenzene and B71-5 replaced by methyl 4-boronic acid-1-p-methoxybenzylpyrazole-3-carboxylate in step 4.
B101 was obtained according to the synthetic route of Example 33 with B178-1 replaced by B101-1.
Compound B103 was synthesized according to a synthetic route similar to that of Example 13 with 3-aminopyridine replaced by 2-methyl-3-aminopyridine.
Compound B109 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by trans-4-amino-cyclohexanol in step 3.
Compound B117 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 3-aminomethylpyrazole in step 3.
Compound B122 was synthesized according to a synthetic route similar to that of Example 23 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate and 3-aminopyridine replaced by 3-aminomethylimidazole in step 1.
Compound B123 was synthesized according to a synthetic route similar to that of Example 23 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate and 3-aminopyridine replaced by 3-aminomethylpyrazole in step 1.
Compound B150 was synthesized according to a synthetic route similar to that of Example 23 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate and 3-aminopyridine replaced by 3-hydroxyaniline in step 1.
Compound B152 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 3-hydroxyaniline in step 3.
Compound B169 was synthesized according to a synthetic route similar to that of Example 79 with B71-4 replaced by 1-bromo-2-fluoro-4-trifluoromethoxybenzene in step 4 and 3-aminopyridine replaced by 3-hydroxyaniline in step 6.
Compound B173 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by (S)-3-aminotetrahydrofuran in step 3.
Compound B174 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by (R)-3-aminotetrahydrofuran in step 3.
Compound B175 was synthesized according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 1,3-dimethyl-5-aminopyrazole in step 3.
Compound B177 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by aniline in step 1.
Compound B184 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-methoxyaniline in step 1.
Compound B185 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 4-methoxyaniline in step 3.
Compound B186 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 4-difluoromethoxyaniline in step 3.
Compound B187 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 4-methoxyaniline in step 1.
Compound B188 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 4-difluoromethoxyaniline in step 1.
Compound B189 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1,5-dimethylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 1.
Compound B190 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-difluoromethoxyaniline in step 1.
Compound B191 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylmethylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 1.
Compound B192 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-cyanoaniline in step 1.
Compound B193 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-fluoroaniline in step 1.
Compound B194 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-difluoromethylaniline in step 1.
Compound B195 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2,6-difluoroaniline in step 1.
Compound B196 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-chloro-3-aminopyridine in step 1.
Compound B197 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 4-aminomethylpyrazole in step 1.
Compound B198 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1,5-dimethylpyrazole-3-carboxylic acid in step 1.
Compound B199 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylmethylpyrazole-3-carboxylic acid in step 1.
Compound B200 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3,5-difluoroaniline in step 1.
Compound B201 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3,5-difluoro-4-hydroxyaniline in step 1.
Compound B202 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-5-fluoropyridine in step 1.
Compound B203 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-methoxy-3-aminopyridine in step 1.
Compound B204 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-(3-tetrahydrofuran)pyrazole-3-carboxylic acid in step 1.
Compound B205 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by
in step 3.
Compound B206 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by
in step 3.
Compound B207 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-cyano-3-aminopyridine in step 1.
Compound B208 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by
in step 3.
Compound B209 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1.
Compound B210 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-(4-tetrahydropyran)pyrazole-3-carboxylic acid in step 1.
Compound B211 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 1.
Compound B214 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by
in step 1.
Compound B215 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylpyrazole-3-carboxylic acid in step 1.
Compound B216-1 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-p-methoxybenzyl-5-methylpyrazole-3-carboxylic acid in step 1.
B216 was obtained according to the synthetic route of Example 33 with B178-1 replaced by B216-1.
Compound B333-1 (2 g, 11.59 mmol) and triethylamine (2.35 g, 23.18 mmol) were added to a solution of ethanol (10 mL), N-methylpiperazine (1.2 g, 11.94 mmol) was added with stirring, and the reaction solution was warmed to 80° C. and stirred overnight. Water was added to the reaction solution, and the mixture was extracted with dichloromethane. The organic phase was washed with saturated brine, dried over sodium sulfate, filtered, and concentrated to give B333-2 (2 g, 73%) as a yellow oily product.
Compound B333-2 (2 g, 7.99 mmol) was added to a solution of ethanol (20 mL), sodium borohydride (1 g, 26.43 mmol) was added with stirring, and the reaction was stirred at room temperature overnight. The reaction solution was filtered and concentrated to give B333-3 (1.5 g, 90%) as an off-white solid product.
Compound B333-3 (1.5 g, 7.20 mmol) was added to a solution of phosphorus oxychloride (6 mL), and the mixture was warmed to 80° C. and stirred for 4 h. Water and ethyl acetate were added to the reaction solution, and the pH was adjusted to 7-9 with a saturated aqueous sodium carbonate solution. The phases were separated and extracted with ethyl acetate. The organic phase was washed with water, washed with saturated brine, concentrated, and separated by column chromatography to give B333-4 (0.7 g, 42%) as a pale yellow oily product.
Compounds methyl 4-bromopyrazole-3-carboxylate (450 mg, 2.05 mmol) and potassium carbonate (850 mg, 6.15 mmol) were added to a solution of N,N-dimethylformamide (5 mL). Compound B333-4 (700 mg, 2.16 mmol) was added with stirring, and the mixture was stirred at room temperature overnight. Water was added to the reaction solution, and the mixture was extracted with ethyl acetate. The organic phase was washed with water, washed with saturated brine, concentrated, and separated by column chromatography to give B333-5 (0.7 g, 86%) as a pale yellow oily product.
Compound B333-5 (700 mg, 1.77 mmol), 2,5-difluoro-3-chlorophenylboronic acid (700 mg, 2.55 mmol), potassium carbonate (730 mg, 5.28 mmol), and 1,1′-bis(diphenylphosphino)ferrocene dichloropalladium(II) (120 mg, 0.16 mmol) were added to a mixed solution of dioxane (10 mL) and water (1 mL), and the mixture was purged three times with nitrogen. The mixture was warmed to 100° C. and stirred overnight. The reaction solution was concentrated and separated by column chromatography to give B333-6 (600 mg, 73%) as a pale yellow oily product.
Compound B333 was obtained according to a synthetic route similar to that of Example 56 with B31-3 replaced by B333-6, 2,4-dimethyl-3-aminopyridine replaced by
and a deprotection reaction finally performed.
Methyl 4-bromopyrazole-3-carboxylate (1 g, 4.88 mmol) and potassium carbonate (2.02 g, 5.37 mmol) were added to a solution of N,N-dimethylformamide (10 mL). 2-Fluoro-5-pyridinylmethyl chloride (780 mg, 2.16 mmol) was added with stirring, and the mixture was stirred at room temperature overnight. Water was added to the reaction solution, and the mixture was extracted with dichloromethane. The organic phase was concentrated, and separated by column chromatography to give B268-1 (0.5 g, 65%) as a pale yellow oily product.
B268-1 (0.5 g, 1.59 mmol) was added to a solution of N,N-dimethylformamide (5 mL). Cesium carbonate (1.55 g, 4.77 mmol) and methylpiperazine (290 mg, 2.86 mmol) were added with stirring, and the mixture was stirred at 80° C. for reaction overnight. The reaction solution was directly separated by column chromatography to give B268-2 (0.5 g, 79%) as a colorless oily substance.
Compound B268 was obtained according to a synthetic route similar to that of Example 132 with B333-5 replaced by B268-2.
Compound B336 was obtained according to a synthetic route similar to that of Example 28 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 2-cyclopropyl-3-aminopyridine in step 1.
Compound B376 was obtained according to a synthetic route similar to that of Example 28 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 1-aminonaphthalene in step 1.
Compound B340 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 2-cyclopropyl-3-aminopyridine in step 1.
Compound B370 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 1-aminonaphthalene in step 1.
Compound B371 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 8-aminoquinoline in step 1.
Compound B373 was obtained according to a synthetic route similar to that of Example 45 with 1-Boc-4-aminoindazole replaced by 1-aminonaphthalene.
Compound B217 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 5-methyl-3-aminopyridine in step 1.
Methyl 4-bromopyrazole-3-carboxylate (2 g, 9.76 mmol), 6-methoxy-3-boronic acid (2.5 g, 16.45 mmol), copper acetate (2 g, 10 mmol), 2,2-bipyridine (1.5 g, 9.6 mmol), sodium carbonate (2.5 g, 23.59 mmol), and dichloroethane (10 mL) were added to a reaction flask. The mixture was stirred at 70° C. for 16 h. The reaction solution was filtered, and the organic phase was concentrated. The crude product was separated and purified by column chromatography to give B218-1 (0.5 g, 16%) as a white solid.
B218-1 (0.5 g, 1.61 mmol), lithium hydroxide (0.2 g, 8.35 mmol), methanol (5 mL), and water (5 mL) were added to a reaction flask. The mixture was stirred at 60° C. for 16 h. The reaction solution was concentrated, adjusted to pH=1, extracted with ethyl acetate, and washed with saturated brine. The organic phase was concentrated to give B218-2 (0.5 g, 79%) as a brown oily product.
Compound B218 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by B218-2 and 3-aminopyridine replaced by aniline in step 1.
Compound B219 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-aminobenzamide in step 1.
Compound B220 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-amino-6-methylpyridine in step 1.
Compound B221 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-5-hydroxypyridine in step 1.
Compound B222 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-5-cyanopyridine in step 1.
Compound B223 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-6-methylpyridine in step 1.
Compound B224 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by methyl 3-aminobenzoate in step 1.
B224 (100 mg, 0.3 mmol), lithium hydroxide (18 mg, 0.8 mmol), methanol (2 mL), and water (2 mL) were added to a reaction flask, and the mixture was stirred at 60° C. for 16 h. The reaction solution was acidified with a solution (4 M) of hydrogen chloride in dioxane, diluted with dichloromethane (50 mL), and washed with water. The organic phase was concentrated and purified by reversed-phase column chromatography to give B225 (8 mg, 8%) as a white solid.
Compound B227 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 2-aminopyrazine in step 1.
Compound B226 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 2-methyl-3-aminopyridine in step 3.
Compound B228 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-hydroxyl-4-fluoro-aniline in step 1.
Compound B229 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by (S)-3-aminotetrahydropyran in step 3.
Compound B230 was obtained according to a synthetic route similar to that of Example 56 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid in step 1 and 2,4-dimethyl-3-aminopyridine replaced by (R)-3-aminotetrahydropyran in step 3.
Methyl 4-bromopyrazole-3-carboxylate (2 g, 9.76 mmol), 6-methoxy-3-pyridinylboronic acid (1.49 g, 2.44 mmol), copper acetate (1.21 g, 3.17 mmol), 2,2-bipyridine (1.68 g, 10.74 mmol), sodium carbonate (2.07 g, 19.52 mmol), and dichloroethane (10 mL) were added to a reaction flask. The mixture was stirred at 70° C. for 16 h. The reaction solution was filtered, and the organic phase was concentrated. The crude product was purified by column chromatography to give B231-1 (1.2 g, 78%) as a white solid product.
Compound B231 was obtained according to a synthetic route similar to that of Example 23 with B29-1 replaced by B231-1 and 3-aminopyridine replaced by aniline in step 1.
Compound B234 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-aminobenzamide in step 1.
Compound B235 was obtained according to a synthetic route similar to that of Example 56 with methyl 4-bromo-1-ethylpyrazole-3-carboxylate replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and 2,4-dimethyl-3-aminopyridine replaced by 4-aminomethylpyrazole in step 3.
B177 (400 mg, 1.16 mmol), N-bromosuccinimide (310 mg, 1.74 mmol), and N,N-dimethylformamide (5 mL) were added to a reaction flask. The mixture was stirred at 60° C. for 16 h. The reaction solution was poured into water and extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried, and purified by column chromatography to give B237 (300 mg, 61%) as a white solid product.
B237 (150 mg, 0.35 mmol), p-methoxyphenylboronic acid (80 mg, 0.52 mmol), [1,1′-bis(di-tert-butylphosphino)ferrocene]palladium dichloride (23 mg, 0.035 mmol), potassium fluoride (61 mg, 1.05 mmol), 1,4-dioxane (3 mL), and water (0.6 mL) were added to a reaction flask. The mixture was stirred at 80° C. for 16 h. The reaction solution was diluted with ethyl acetate and washed with water. The organic phase was concentrated, and the crude product was purified by column chromatography to give B236 (50 mg, 31%) as a white solid product.
Compound B238 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 1-(3-pyrazolyl)-ethylamine in step 1.
Compound B240 was obtained according to a synthetic route similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-1-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 3-amino-5-difluoromethylpyridine in step 1.
2-(7-Azabenzotriazol)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (4.15 g, 10.93 mmol) and N,N-diisopropylethylamine (1.83 g, 14.14 mmol) were added to a solution of compound B244-1 (2 g, 9.76 mmol) in DMF (10 mL), meanwhile the solution being stirred, and the mixture was stirred at room temperature for 0.5 h. 3-Aminopyridine (1.5 g, 15.9 mmol) was then added to the mixed solution, and the mixture was stirred at room temperature overnight. Water was added to the reaction solution, and the mixture was extracted with ethyl acetate. The organic phases were combined, washed with water, washed with saturated brine, dried over sodium sulfate, filtered, and concentrated to give B244-2 (2.2 g, 80%) as an orange solid product.
Compound B244-2 (2 g, 7.11 mmol), 2,6-dichloro-3-pyridineboronic acid (1.7 g, 8.86 mmol), potassium fluoride (1.25 g, 21.51 mmol), and dichloro[1,1′-bis(di-tert-butylphosphino)ferrocene]palladium(II) (400 mg, 0.086 mmol) were added to a mixed solution of dioxane (10 mL) and water (1 mL), and the mixture was purged three times with nitrogen. The mixture was heated to 80° C. and stirred overnight. The reaction solution was directly concentrated and separated by column chromatography to give B244-3 (2 g, 80%) as a black oily product.
1,8-Diazabicyclo[5.4.0]undec-7-ene (2.1 g, 13.79 mmol) was added to a solution of compound B244-3 (1.6 g, 4.6 mmol) in dimethyl sulfoxide (8 mL), meanwhile the solution being stirred. The mixture was purged three times with nitrogen. The mixture was warmed to 120° C. and stirred overnight. Water was added to the reaction solution, and the mixture was extracted with dichloromethane. The organic phases were combined, washed with water, washed with saturated brine, dried over sodium sulfate, concentrated, and separated by column chromatography to give B244 (500 mg, 35%) as a white solid.
Compound B244 (40 mg, 0.13 mmol), 2,2-difluoroethylamine (11 mg, 0.13 mmol), potassium carbonate (50 mg, 0.36 mmol), tris(dibenzylideneacetone)dipalladium (10 mg, 0.09 mmol), and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (40 mg, 0.08 mmol) were added to DMF (2 mL), and the mixture was purged three times with nitrogen. The mixture was heated to 80° C. and stirred overnight. The reaction solution was directly separated by column chromatography to give B241 (24 mg, 54%) as an off-white solid.
Compound B244 (80 mg, 0.26 mmol), potassium carbonate (120 mg, 0.86 mmol), tris(dibenzylideneacetone)dipalladium (24 mg, 0.026 mmol), and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (82 mg, 0.17 mmol) were added to a mixed solution of ethanol (1 mL), and the mixture was purged three times with nitrogen. The mixture was heated to 80° C. and stirred overnight. The reaction solution was separated by reversed-phase column chromatography to give B242 (28 mg, 33%) as a white solid.
Compound B243-1 was obtained according to a synthetic route similar to that for intermediate B33-4 in Example 58 with B31-1 replaced by methyl 4-bromo-1-methylpyrazole-3-carboxylate in step 1 and B31-2 replaced by 2-chloro-6-trifluoromethylpyridine-3-boronic acid in step 2.
1,8-Diazabicyclo[5.4.0]undec-7-ene (170 mg, 1.12 mmol) was added to a solution of B243-1 (150 mg, 0.38 mmol) in DMSO (3 mL), meanwhile the solution being stirred. The mixture was warmed to 120° C. and stirred overnight. Water was added to the reaction solution, and the mixture was extracted with dichloromethane. The organic phase was concentrated and separated by column chromatography to give B243 (23 mg, 12%) as a white solid product.
Compound B251 was obtained according to a synthetic route similar to that of Example 159 with 2,2-difluoroethylamine replaced by ethylamine in step 4.
Compound B252 was obtained according to a synthetic route similar to that of Example 23 with B29-1 replaced by 4-bromo-1-difluoromethyl-5-methylpyrazole-3-carboxylic acid in step 1.
B244 (40 mg, 0.13 mmol), 2,2-difluoroethanol (30 mg, 0.37 mmol), potassium carbonate (60 mg, 0.43 mmol), tris(dibenzylideneacetone)dipalladium (20 mg, 0.022 mmol), and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (60 mg, 0.13 mmol) were added to a mixed solution of DMF (1 mL). The mixture was purged three times with nitrogen, heated to 80° C., and stirred overnight. The reaction solution was directly separated by column chromatography to give B253 (35 mg, 76%) as an off-white solid product.
Compound B255 was obtained according to a synthetic route similar to that of Example 23 with B29-1 replaced by 4-bromo-1-difluoromethylpyrazole-3-carboxylic acid in step 1.
Intermediate B263-1 was synthesized according to a synthetic route similar to that of Example 28 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-p-methoxybenzylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by 1-Boc-4-aminoindazole.
B263 was synthesized according to a preparation method similar to that for B178 in Example 33.
Compound B267 was obtained according to a synthetic route similar to that of Example 133 with 3-chloromethyl-6-fluoropyridine replaced by 2-chloromethyl-6-fluoropyridine in step 1.
Compound B342 was obtained according to a synthetic route similar to that of Example 28 with 3-aminopyridine replaced by 2-trifluoromethyl-3-aminopyridine.
B342 was subjected to chiral resolution (IA-3.0 cm, n-hexane/ethanol=50:50, 25 mL/min, 230 nm) to give B388 (retention time: 9.79 min) and B389 (retention time: 14.33 min).
Compound B348 was obtained according to a synthetic route similar to that of Example 45 with 1-Boc-3-aminoindazole replaced by 2-cyclopropyl-3-aminopyridine.
Compound B372-1 was obtained according to a preparation method similar to that for B29-2 in Example 23 with B29-1 replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 1-Boc-3-aminoindazole.
B372-1 (0.5 g, 1.19 mmol), (4-chloro-2,5-difluorophenyl)boronic acid (0.34 g, 1.78 mmol), [1,1′-bis(di-tert-butylphosphino)ferrocene]palladium dichloride (87 mg, 0.12 mmol), potassium fluoride (490 mg, 3.57 mmol), 1,4-dioxane (2.5 mL), and water (0.5 mL) were added to a reaction flask. The mixture was stirred at 80° C. for 16 h under nitrogen atmosphere. The reaction solution was diluted with ethyl acetate and washed with water. The organic phase was concentrated, and the crude product was purified by column chromatography to give B372-2 (70 mg, 8%) as a white solid product.
B372-2 (70 mg, 0.14 mmol), cesium fluoride (110 mg, 0.70 mmol), and N,N-dimethylformamide (1 mL) were added to a reaction flask. The mixture was stirred at 80° C. for 16 h. The reaction solution was diluted with dichloromethane and washed with water. The organic phase was concentrated, and the crude product was purified by column chromatography to give B372-3 (60 mg, 89%) as a yellow solid.
B372-3 (60 mg, 0.13 mmol), trifluoroacetic acid (0.5 mL), and dichloromethane (0.5 mL) were added to a reaction flask. The mixture was stirred at 80° C. for 16 h. The reaction solution was concentrated and purified by reversed-phase column chromatography to give B372 (22 mg, 43%) as a white solid.
Compound B351 was obtained according to a synthetic route similar to that of Example 170 with 1-Boc-3-aminoindazole replaced by 2-trifluoromethyl-3-aminopyridine.
Compound B374 was obtained according to a synthetic route similar to that of Example 45 with 1-Boc-3-aminoindazole replaced by 8-aminoquinoline.
Compound B375 was obtained according to a synthetic route similar to that of Example 170 with 4-chloro-2,5-difluorophenylboronic acid replaced by 2-fluoro-4-chloro-5-cyanophenylboronic acid pinacol ester in step 1.
Compound B377 was obtained according to a synthetic route similar to that of Example 170 with 1-Boc-3-aminoindazole replaced by 8-aminoquinoline.
Compound B378 was obtained according to a synthetic route similar to that of Example 170 with 4-chloro-2,5-difluorophenylboronic acid replaced by 2-fluoro-4-chloro-5-cyanophenylboronic acid pinacol ester and 1-Boc-3-aminoindazole replaced by 1-aminonaphthalene.
Compound B379 was obtained according to a synthetic route similar to that of Example 170 with 4-chloro-2,5-difluorophenylboronic acid replaced by 2-fluoro-4-chloro-5-cyanophenylboronic acid pinacol ester and 1-Boc-3-aminoindazole replaced by 8-aminoquinoline.
Compound B381 was obtained according to a synthetic route similar to that of Example 28 with 2-methyl-3-aminopyridine replaced by 2-methyl-4-aminoindazole.
Compound B382 was obtained according to a synthetic route similar to that of Example 28 with 2-methyl-3-aminopyridine replaced by 2-methyl-7-aminoindazole.
Intermediate B394-1 was obtained according to a synthetic route similar to that of Example 45 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-benzylpyrazole-3-carboxylic acid.
Compound B394 was obtained according to the conditions similar to those for the preparation of B178 in Example 33.
Compound B400 was obtained according to a synthetic method similar to that for B253 in Example 164 with B244 replaced by B186.
Compound B401 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 6-aminoquinoline in step 1.
Compound B402 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 2-aminoquinoline in step 1.
Compound B403 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 3-aminoquinoline in step 1.
Compound B404 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 2-aminonaphthalene in step 1.
Compound B405 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 2-amino-1,5-dinaphthyridine in step 1.
Compound B408 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 5-aminoquinoline in step 1.
Compound B407 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 8-aminoisoquinoline in step 1.
Compound B409 was obtained according to a synthetic method similar to that of Example 33 with 3-aminopyridine replaced by o-chloroaniline.
Compound B410 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 4-bromo-2-methylpyrazole-3-carboxylic acid and 3-aminopyridine replaced by o-chloroaniline in step 1.
Compound B406 was obtained according to a synthetic method similar to that of Example 23 with 4-bromo-1-ethylpyrazole-3-carboxylic acid replaced by 5-bromo-1-methylimidazole-4-carboxylic acid and 3-aminopyridine replaced by 4-aminoisoquinoline in step 1.
Compound B286 was obtained according to a synthetic method similar to that of Example with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-p-methoxybenzylpyrazole-3-carboxylic acid.
Compound B293 was obtained according to a synthetic method similar to that of Example 45 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-(1-methylpyrazole-3-methyl)pyrazole-3-carboxylic acid.
Compound B393 was obtained according to a synthetic method similar to that of Example 45 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1,5-dimethylpyrazole-3-carboxylic acid.
Compound B396 was obtained according to a synthetic method similar to that of Example with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylmethylpyrazole-3-carboxylic acid.
B257 (0.15 g, 0.41 mmol) was dissolved in a solution of N,N-dimethylformamide (2 mL), N-chlorosuccinimide (0.11 g, 0.82 mmol) was added with stirring, and the mixture was stirred at 25° C. for reaction for 16 h. When LCMS indicated the completion of the reaction, the reaction solution was filtered to give a filtrate, which was purified by reversed-phase column chromatography (formic acid 0.05% aqueous solution/acetonitrile) to give B412 (7 mg, 4.22%) as a white product.
B257 (0.15 g, 0.41 mmol) was dissolved in a solution of N,N-dimethylformamide (2 mL), N-bromosuccinimide (0.15 g, 0.82 mmol) was added with stirring, and the mixture was stirred at 25° C. for reaction for 16 h. When LCMS indicated the completion of the reaction, the reaction solution was filtered to give a filtrate, which was purified by reversed-phase column chromatography (formic acid 0.05% aqueous solution/acetonitrile) to give B413 (33 mg, 18%) as a white product.
B414-1 (5.00 g, 19.62 mmol), cyclopropylboronic acid (1.85 g, 21.58 mmol), and potassium phosphate (10.41 g, 49.05 mmol) were added to a solution of 1,4-dioxane (50 mL). After the mixture was purged three times with nitrogen, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (1.44 g, 1.96 mmol) and tricyclohexylphosphine (1.11 g, 3.92 mmol) were added, and the mixture was stirred at 90° C. and reacted for 6 h. When LCMS indicated the completion of the reaction, the reaction solution was poured into water, and the mixture was extracted with dichloromethane (50 mL×2), concentrated, and purified by column chromatography (petroleum ether/dichloromethane) to give B414-2 (2.3 g, 39%) as a white oily substance. m/z [M+H]+=215.9.
B414-2 (2 g, 9.26 mmol) was added to a solution of dichloromethane (40 mL), m-chloroperoxybenzoic acid (3.2 g, 18.52 mmol) was added in portions with stirring at 0° C., and then the mixture was reacted at 25° C. for 16 h. LCMS indicated the completion of the reaction. The reaction solution was poured into water, extracted with ethyl acetate (25 mL×2), and concentrated to give B414-3 (0.53 g, 22%) as a white oily substance. m/z [M+H]+=231.9.
B414-3 (0.53 g, 2.28 mmol) was added to a solution of phosphorus oxychloride (15 mL), and the mixture was stirred at 80° C. and reacted for 12 h. When LCMS indicated the completion of the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate (10 mL×2), and concentrated to give B414-4 (0.11 g, yield: 16.53%) as a colorless oily substance. m/z [M+H]+=249.9.
4-Bromo-1-methylpyrazole-3-carboxylic acid (0.3 g, 1.46 mmol) was added to a solution of N,N-dimethylformamide (5 mL). 1-Naphthylamine (0.23 g, 1.61 mmol), 2-(7-azabenzotriazole)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (0.61 g, 1.61 mmol) and N,N-diisopropylethylamine (0.57 g, 4.38 mmol) were sequentially added with stirring, and the mixture was reacted at 25° C. overnight. When LCMS indicated the completion of the reaction, the reaction solution was poured into water, and extracted with ethyl acetate (20 mL×2). The organic phases were combined and concentrated to give B414-5 (0.21 g, 43%) as a brown solid. m/z [M+H]+=330.1.
B414-5 (0.21 g, 0.64 mmol) was dissolved in a solution of 1,4-dioxane (4 mL). bis(Pinacolato)diboron (0.24 g, 0.96 mmol) and potassium acetate (0.19 g, 1.92 mmol) were added with stirring. After the mixture was purged three times with nitrogen, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (0.05 g, 0.06 mmol) was added, and the mixture was stirred at 100° C. and reacted for 6 h. When LCMS indicated the completion of the reaction, the mixture was purified by column chromatography (ethyl acetate/petroleum ether) to give B414-6 (0.15 g, 63%) as a white solid.
B414-4 (0.1 g, 0.40 mmol), B414-6 (0.15 g, 0.40 mmol), and potassium fluoride (0.07 g, 1.2 mmol) were added to a mixed solution of 1,4-dioxane (20 mL) and water (0.5 mL). After the mixture was purged three times with nitrogen, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (0.026 g, 0.04 mmol) was added, and the mixture was stirred at 90° C. and reacted for 12 h. When LCMS indicated the completion of the reaction, the reaction solution was poured into water, extracted with dichloromethane (50 mL×2), concentrated, and purified by column chromatography (petroleum ether/dichloromethane) to give B414-7 (40 mg, 24%) as a white oily substance. m/z [M+H]+=421.1.
B414-7 (40 mg, 0.09 mmol) was dissolved in a solution of N,N-dimethylformamide (2 mL), potassium fluoride (17 mg, 0.29 mmol) was added with stirring, and the mixture was stirred at 80° C. for reaction for 16 h. When LCMS indicated the completion of the reaction, the reaction solution was filtered to give a filtrate, which was purified by reversed-phase column chromatography (formic acid 0.05% aqueous solution/acetonitrile) to give B414 (3 mg, 8%) as a white product.
B391 was obtained according to a synthetic method similar to that for compound B258 in Example 45 with B258-1 replaced by 4-amino-2-trifluoromethylbenzonitrile in step 1.
Compound B283 was obtained according to a synthetic method similar to that for compound B258 in Example 45 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-difluoromethylpyrazole-3-carboxylic acid.
Compound B281 was obtained according to a synthetic method similar to that of Example 48 with B274-1 replaced by 6-chloro-2-Trifluoromethylnicotinonitrile in step 1.
Compound B284 was obtained according to a synthetic method similar to that of Example 45 with 4-bromo-1-methylpyrazole-3-carboxylic acid replaced by 4-bromo-1-cyclopropylpyrazole-3-carboxylic acid.
B257 (0.15 g, 0.41 mmol) was dissolved in acetonitrile (2 mL) solution, 1-chloromethyl-4-fluoro-1,4-diazobicyclo[2.2.2]octane bis(tetrafluoroborate) (0.23 g, 0.66 mmol) was added with stirring, and the mixture was stirred at 25° C. for reaction for 36 h. The reaction solution was filtered, and the filtrate was separated by reversed-phase column chromatography to give B411 (10 mg, 6%) as a white product.
Compound B359 was obtained according to a synthetic method similar to that of Example 197 with
replaced by
Compound B361 was obtained according to a synthetic method similar to that of Example 203 with
replaced by
B425-1 was obtained according to a synthetic method similar to that of Example 136 with
replaced by
Compound B425 was obtained according to a synthetic method similar to that of Example 13 with compound B28 replaced by B425-1.
Nuclear magnetic resonance and mass spectrometry analysis data of the exemplary compounds of the present application are shown below:
1H NMR (400 MHz, DMSO-d6) δ 8.86 (s, 1H), 8.05 (d, J = 8.3 Hz, 1H), 7.69 (dd, J = 8.3, 6.7 Hz, 2H), 7.66-7.58 (m, 1H), 7.45-7.38 (m, 2H), 7.34 (dd, J = 8.4, 2.0 Hz, 1H), 6.39 (d, J = 2.0 Hz, 1H), 4.48 (q, J = 7.3 Hz, 2H), 1.55 (t, J = 7.3 Hz, 3H). LC-MS. [M + H]+ = 324.1
1H NMR (400 MHz, DMSO-d6) δ 8.85 (s, 1H), 8.79 (dd, J = 4.8, 1.5 Hz, 1H), 8.63 (d, J = 2.4 Hz, 1H), 8.05 (d, J = 8.4 Hz, 1H), 7.94 (ddd, J = 8.1, 2.5, 1.6 Hz, 1H), 7.72 (dd, J = 8.1, 4.8 Hz, 1H), 7.35 (dd, J = 8.3, 2.0 Hz, 1H), 6.35 (d, J = 2.0 Hz, 1H), 4.46 (q, J = 7.3 Hz, 2H), 1.53 (t, J = 7.3 Hz, 3H). LC-MS. [M + H]+ = 325.1
1H NMR (400 MHz, DMSO-d6): δ 8.84 (s, 1H), 8.04-8.02 (m, 1H), 7.73- 7.67 (m, 1 H), 7.48-7.40 (m, 2H), 7.32-7.32 (m, 1H), 7.27-7.26 (m, 1H), 6.39 (s, 1H), 4.48-4.42 (m, 2H), 1.53-1.50 (m, 3 H). LC-MS. [M + H]+ = 342.1
1H NMR (400 MHz, DMSO-d6): δ 8.83 (s, 1H), 8.02 (d, J = 8 Hz, 1H), 7.53- 7.43 (m, 4H), 7.34-7.30 (m, 1H), 6.40 (d, J = 1.6 Hz, 1H), 4.49-4.40 (m, 2H), 1.56-1.48 (m, 3H). LC-MS [M + H]+ = 342.1
1H NMR (400 MHz, DMSO-d6): δ 8.94-8.87 (m, 4H), 8.06 (d, J = 8.4 Hz, 1H), 7.39-7.36 (m, 1H), 6.46 (s, 1H), 4.49-4.44 (m, 2H), 1.53 (t, 3H). LC-MS [M + H]+ = 326.1
1H NMR (400 MHz, DMSO-d6): δ 9.15 (d, J = 4.4 Hz, 2H), 8.88 (s, 1H), 8.07 (d, J = 8.8 Hz, 1H), 7.85-7.77 (m, 1H), 7.37 (d, J = 8.6 Hz, 1H), 6.28 (s, 1H), 4.56-4.41 (m, 2H), 1.58-1.48 (m, 3H). LC-MS [M + H]+ = 326.1
1H NMR (400 MHz, DMSO-d6): δ 9.87 (brs, 1H), 8.88 (s, 1H), 8.02 (d, J = 8.4 Hz, 1H), 7.46-7.42 (m, 1H), 7.30 (dd, J1 = 2.0 Hz, J2 = 8.4 Hz, 1H), 6.99-6.96 (m, 1H), 6.77-6.71 (m, 2H), 6.45 (d, J = 2.0 Hz, 1H), 4.44 (q, J = 7.2 Hz, 2H), 1.52 (t, J = 7.2 Hz, 3H). LC-MS [M + H]+ = 340.0
1H NMR (400 MHz, DMSO-d6): δ 8.85 (s, 1H), 8.08-8.04 (m, 3H), 7.89-7.79 (m, 2H), 7.34 (dd, J1 = 2.0 Hz, J2 = 8.4 Hz, 1H), 6.38 (d, J = 2.0 Hz, 1H), 4.45 (q, J = 7.2 Hz, 2H), 1.52 (t, J = 7.2 Hz, 3H). LC-MS [M + H]+ = 349.0
1H NMR (400 MHz, DMSO-d6): δ 8.89 (d, J = 5.2 Hz, 2H), 8.85 (s, 1H), 8.05 (d, J = 7.6 Hz, 1H), 7.53 (d, J = 2.4 Hz, 2H), 7.37-7.34 (m, 1H), 6.40 (s, 1H), 4.48- 4.43 (m, 2H), 1.52 (t, 3H). LC-MS [M + H]+ = 325.1
1H NMR (400 MHz, DMSO-d6): δ 8.83 (s, 1H), 8.01 (d, J = 8.4 Hz, 1H), 7.32- 7.29 (m, 3H), 7.18-7.16 (m, 2H), 7.44 (d, J = 2.0 Hz, 1H), 4.47-4.41 (m, 2H), 3.87 (s, 3H), 1.53-1.50 (m, 3H). LC-MS [M + H]+ = 354.1
1H NMR (400 MHz, DMSO-d6): δ 8.78 (d, J = 4.0 Hz, 2H), 8.62 (d, J = 0.8 Hz, 1H), 8.05 (d, J = 8.0 Hz, 1H), 7.96-7.93 (m, 1H), 7.72-7.69 (m, 1H), 7.36- 7.33 (m, 1H), 6.38 (d, J = 2 Hz, 1H), 4.16 (s, 3H). LC-MS [M + H]+ = 311.1
1H NMR (400 MHz DMSO_d6): δ 10.08-9.26 (m, 1H), 8.80 (dd, J1 = 4.8 Hz, J2 = 1.2 Hz, 1H), 8.69-8.64 (m, 2 H), 8.18 (d, J = 8.4 Hz, 1H), 8.00-7.95 (m, 1 H), 7.72 (dd, J1 = 8.0, J2 = 4.8 Hz, 1H), 7.39 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, 1H), 6.43 (d, J = 1.6 Hz, 1H). LC-MS [M + H]+ = 297.0
1H NMR (400 MHz, DMSO-d6): δ 8.86 (s, 1H), 8.03 (d, J = 2.4 Hz, 1H), 8.54 (s, 1H), 8.10-8.04 (m, 2H), 7.38-7.35 (m, 1H), 6.51 (d, J = 2.0 Hz, 1H), 4.49- 4.43 (m, 2H), 1.54-1.50 (m, 3H). LC-MS [M + H]+ = 343.0
1H NMR (400 MHz, DMSO-d6): δ 8.78 (d, J = 3.6 Hz, 1H), 8.60 (d, J = 2.0 Hz, 1H), 8.07 (d, J = 8.4 Hz, 1H), 7.94-7.91 (m, 1H), 7.72-7.69 (m, 1H), 7.34- 7.31 (m, 1H), 6.38 (d, J = 2 Hz, 1H), 4.07 (s, 3H), 2.78 (s, 3H). LC-MS [M + H]+ = 325.0
1H NMR (400 MHz, DMSO-d6): δ 8.78-8.79 (m, 1H), 8.60 (d, J = 2.4 Hz, 1H), 8.08 (d, J = 8.8 Hz, 1H), 7.91-7.94 (m, 1H), 7.70-7.73 (m, 1H), 7.32-7.35 (m, 1H), 6.38 (d, J = 2.0 Hz, 1H), 4.44 (q, J = 8.0 Hz, 2H), 2.82 (s, 3H), 1.42 (t, J = 6.0 Hz, 3H). LC-MS [M + H]+ = 339.0
1H NMR (400 MHz, DMSO-d6) δ 12.00 (s, 1H), 8.78 (s, 1H), 7.97 (d, J = 8.3 Hz, 1H), 7.87 (s, 1H), 7.59 (s, 1H), 7.32 (dd, J = 8.3, 1.8 Hz, 1H), 6.97 (s, 1H), 5.41 (s, 2H), 4.45 (q, J = 7.3 Hz, 2H), 1.54 (t, J = 7.3 Hz, 3H). LC-MS. [M + H]+ = 328.1
1H NMR (400 MHz, DMSO-d6): δ 8.88 (s, 1H), 8.78-8.80 (m, 1H), 8.64 (d, J = 2.2 Hz, 1H), 8.15 (d, J = 8.6 Hz, 1H), 7.94-7.97 (m, 1H), 7.70-7.73 (m, 1H), 7.31 (d, J = 9.6 Hz, 1H), 6.29 (s, 1H), 4.47 (q, J = 7.3 Hz, 2H), 1.53 (t, J = 7.3 Hz, 3H). LC-MS [M + H]+ = 375.0
1H NMR (400 MHz, CDCl3): δ 8.83 (d, J = 3.6 Hz, 1H), 8.63-8.58 (m, 1H), 8.32 (s, 1H), 7.77-7.68 (m, 2H), 7.64-7.58 (m, 1H), 7.25-7.22 (m, 1H), 6.58 (d, J = 1.6 Hz, 1H), 5.42 (s, 2H). LC-MS [M + H]+ = 336.0
1H NMR (400 MHz, CDCl3): δ 8.87 (dd, J1 = 4.4 Hz, J2 = 1.2 Hz, 1H), 8.61 (d, J = 2.4 Hz, 1H), 8.30 (s, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.75-7.70 (m, 1 H), 7.67-7.61 (m, 1 H), 7.34 (dd, J1 = 8.0, J2 = 1.6 Hz, 1H), 6.66 (d, J = 1.6 Hz, 1H), 5.83-5.69 (m, 2 H). LC-MS [M + H]+ = 336.0
1H NMR (400 MHz, d6-DMSO) δ 8.88-8.75 (m, 1H), 8.67 (t, J = 11.2 Hz, 1H), 8.22 (d, J = 8.9 Hz, 1H), 7.99 (d, J = 8.1 Hz, 1H), 7.76 (dd, J = 8.0, 4.8 Hz, 1H), 7.47 (dd, J = 8.8, 2.2 Hz, 1H), 6.54 (d, J = 2.2 Hz, 1H), 3.06 (s, 3H). LCMS [M + H]+ = 311.9
1H NMR (400 MHz, DMSO) δ 8.81 (d, J = 4.8 Hz, 1H), 8.66 (d, J = 2.2 Hz, 1H), 7.98 (dd, J = 8.3, 4.3 Hz, 2H), 7.73 (dd, J = 8.0, 4.8 Hz, 1H), 7.47 (dd, J = 8.7, 1.9 Hz, 1H), 6.49 (d, J = 1.9 Hz, 1H), 4.37 (d, J = 1.9 Hz, 3H). LC-MS [M + H]+ = 378.9
1H NMR (400 MHz, DMSO-d6): δ 9.00 (s, 1H), 8.66 (d, J = 7.6 Hz, 2H), 8.55 (d, J = 2.0 Hz, 1H), 7.86-7.83 (m, 1H), 7.80-7.78 (m, 1H), 7.62-7.59 (m, 1H), 4.52-4.47 (m, 2H), 1.55 (t, 3H). LC-MS [M + H]+ = 360.0
1H NMR (400 MHz, DMSO-d6): δ 8.87 (s, 1H), 8.68-8.67 (m, 1H), 8.06 (d, J = 8.0 Hz, 1H), 7.83-7.80 (m, 1H), 7.54-7.51 (m, 1H), 7.38-7.35 (m, 1H), 6.30 (d, J = 2.0 Hz, 1 H), 4.52-4.47 (m, 2H), 2.16 (s, 3H), 1.56-1.52 (m, 3H). LC-MS [M + H]+ = 339.0
1H NMR (400 MHz, DMSO-d6): δ 8.99 (d, J = 4.0 Hz, 1H), 8.87 (s, 1H), 8.25 (d, J = 8.0 Hz, 1H), 8.08-8.05 (m, 2H), 7.38-7.36 (m, 1H), 6.38 (d, J = 1.6 Hz, 1H), 4.49-4.36 (m, 2H), 1.56-1.52 (t, 3H). LC-MS [M + H]+ = 393.0
1H NMR (400 MHz, DMSO_d6): δ 8.79 (d, J = 4.8 Hz, 1 H), 8.63 (s, 1H), 7.96-7.93 (m, 1 H), 7.86 (d, J = 8.0 Hz, 1 H), 7.73-7.70 (m, 1 H), 7.38 (d, J = 8.0 Hz, 1 H), 6.39 (s, 1 H), 4.42-4.37 (m, 2 H), 1.48 (t, J = 7.6 Hz, 3 H). LC-MS [M + H]+ = 342.9
1H NMR (400 MHz, DMSO_d6): δ 8.81 (d, J = 4.0 Hz, 1 H), 8.66 (d, J = 1.6 Hz, 1 H), 8.14 (d, J = 8.4 Hz, 1 H), 7.97 (d, J = 8.0 Hz, 1 H), 7.75-7.72 (m, 1 H), 7.50-7.48 (m, 1 H), 6.60 (S, 1 H), 2.75 (S, 3H). LC-MS [M + H]+ = 312.0
1H NMR (400 MHz, DMSO_d6): δ 8.81-8.76 (m, 2H), 8.63 (d, J = 2.4 Hz, 1H), 8.11 (d, J = 8.0 Hz, 1H), 7.97-7.92 (m, 1 H), 7.76-7.69 (m, 2 H), 7.44- 7.40 (m, 1H), 7.35 (dd, J1 = 8.0 Hz, J2 = 2.0 Hz, 1H), 6.38 (d, J = 2.0 Hz, 1H), 5.12 (s, 2H). LC-MS [M + H]+ = 353.9
1H NMR (400 MHz, DMSO_d6): δ 8.82-8.77 (m, 2H), 8.63 (d, J = 2.0 Hz, 1H), 8.29-8.23 (m, 1H), 8.11 (d, J = 8.4 Hz, 1H), 7.97-7.92 (m, 1 H), 7.72 (dd, J1 = 7.6 Hz, J2 = 4.8 Hz, 1H), 7.35 (dd, J1 = 8.0 Hz, J2 = 2.0 Hz, 1H), 6.38 (d, J = 2.0 Hz, 1H), 5.12 (s, 2H), 2.70-2.63 (m, 3H). LC-MS [M + H]+ = 368.0
1H NMR (400 MHz, DMSO_d6): δ 8.85-8.81 (m, 1H), 8.74-8.69 (m, 2 H), 8.11 (d, J = 8.4 Hz, 1H), 8.07-8.03 (m, 1 H), 7.78 (dd, J1 = 8.0 Hz, J2 = 4.8 Hz, 1H), 7.35 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, 1H), 6.43 (d, J = 1.6 Hz, 1H), 5.51 (s, 2H), 3.10 (s, 3H), 2.89 (s, 3H). LC-MS [M + H]+ = 382.0
1H NMR (400 MHz, CDCl3): δ 8.82 (dd, J1 = 4.8 Hz, J2 = 1.2 Hz, 1H), 8.60 (d, J = 2.0 Hz, 1H), 8.18 (s, 1H), 7.77-7.69 (m, 2 H), 7.60 (dd, J1 = 8.0 Hz, J2 = 4.8 Hz, 1H), 7.23 (dd, J1 = 8.0 Hz, J2 = 1.6 Hz, 1H), 6.56 (d, J = 2.0 Hz, 1H), 6.42- 6.11 (m, 1H), 4.81-4.72 (m, 2H). LC-MS [M + H]+ = 361.0
1H NMR (400 MHz, CDCl3): δ 8.81 (dd, J1 = 4.8 Hz, J2 = 1.6 Hz, 1H), 8.60 (d, J = 2.4 Hz, 1H), 8.13 (s, 1H), 7.73-7.69 (m, 2 H), 7.62-7.57 (m, 1 H), 7.20 (dd, J1 = 8.4, J2 = 1.6 Hz, 1H), 6.55 (d, J = 2.0 Hz, 1H), 4.50 (t, J = 6.8 Hz, 2H), 2.31-2.25 (m, 2 H), 2.24 (s, 6 H), 2.24-2.14 (m, 2H). LC-MS [M + H]+ = 382.0
1H NMR (400 MHz, DMSO_d6): δ 8.82 (s, 1H), 8.78 (dd, J1 = 4.8 Hz, J2 = 1.6 Hz, 1H), 8.63 (d, J = 2.4 Hz, 1H), 8.06 (d, J = 8.8 Hz, 1H), 7.97-7.93 (m, 1H), 7.71 (dd, J1 = 8.0 Hz, J2 = 4.8 Hz, 1H), 7.34 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, 1H), 6.37 (d, J = 2.0 Hz, 1H), 4.51 (t, J = 6.4 Hz, 2H), 2.78 (t, J = 6.4 Hz, 2H), 2.20 (s, 6 H). LC-MS [M + H]+ = 368.0.
1H NMR (400 MHz, CDCl3): δ 8.80 (d, J = 4.4 Hz, 1H), 8.58 (d, J = 2.0 Hz, 1H), 8.13 (s, 1H), 7.73-7.65 (m, 2 H), 7.61-7.57 (m, 1 H), 7.35 (d, J = 2.4 Hz, 1H), 7.16 (dd, J1 = 8.8 Hz, J2 = 2.0 Hz, 1H), 6.52 (d, J = 2.0 Hz, 1H), 6.32 (d, J = 2.4 Hz, 1H), 5.60 (s, 2 H), 3.93 (s, 3 H). LC-MS [M + H]+ = 391.0
1H NMR (400 MHz, DMSO) δ 9.22 (d, J = 2.1 Hz, 1H), 9.09 (d, J = 2.1 Hz, 1H), 8.86 (dd, J = 4.8, 1.5 Hz, 1H), 8.79 (d, J = 8.6 Hz, 1H), 8.74 (d, J = 1.9 Hz, 1H), 8.04 (ddd, J = 8.1, 2.4, 1.6 Hz, 1H), 7.79 (ddd, J = 8.2, 4.8, 0.7 Hz, 1H), 7.54 (dd, J = 8.5, 1.9 Hz, 1H), 6.56 (d, J = 1.9 Hz, 1H). LC-MS [M + H]+ = 309.1
1H NMR (400 MHz, DMSO_d6): δ 10.00 (s, 1H), 9.73 (d, J = 6.0 Hz, 1H), 9.21 (d, J = 8.4 Hz, 1H), 8.76-8.68 (m, 2 H), 8.50-8.20 (m, 2 H), 7.46-7.44 (m, 1 H), 7.06 (s, 1H). LC-MS [M + H]+ = 346.9
1H NMR (400 MHz, DMSO-d6) δ 13.42 (s, 1H), 8.79 (s, 1H), 8.22 (s, 1H), 8.05 (d, J = 8.3 Hz, 1H), 7.87-7.78 (m, 2H), 7.31 (ddd, J = 14.2, 8.5, 2.0 Hz, 2H), 6.42 (d, J = 2.0 Hz, 1H), 4.19 (s, 3H). LC-MS [M + H]+ = 350.1
1H NMR (400 MHz, DMSO-d6) δ 8.82 (s, 1H), 8.09 (d, J = 8.3 Hz, 1H), 7.74 (d, J = 2.0 Hz, 1H), 7.43 (dd, J = 8.4, 2.0 Hz, 1H), 6.51 (dd, J = 8.8, 2.0 Hz, 2H), 4.20 (s, 3H), 3.56 (s, 3H). LC-MS [M + H]+ = 314.1
1H NMR (400 MHz, DMSO-d6) δ 8.83 (s, 1H), 8.70 (dd, J = 4.8, 1.6 Hz, 1H), 8.10 (d, J = 8.4 Hz, 1H), 7.85 (dd, J = 7.9, 1.6 Hz, 1H), 7.56 (dd, J = 7.9, 4.8 Hz, 1H), 7.39 (dd, J = 8.3, 2.0 Hz, 1H), 6.33 (d, J = 1.9 Hz, 1H), 4.20 (s, 3H), 2.16 (s, 3H). LC-MS [M + H]+ = 325.1
1H NMR (400 MHz, DMSO_d6): δ 8.99 (d, J = 4.8 Hz, 1 H), 8.80 (s, 1 H), 8.25 (d, J = 7.6 Hz, 1 H), 8.08-8.04 (m, 2 H), 7.38-7.35 (m, 1 H), 6.38 (d, J = 2.0 Hz, 1 H), 4.17 (s, 3H), LC-MS [M + H]+ = 378.9
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.72 (d, J = 7.9 Hz, 1H), 8.59 (dd, J = 4.9, 1.6 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.76 (dd, J = 7.9, 1.6 Hz, 1H), 7.46 (dd, J = 7.9, 4.8 Hz, 1H), 4.24 (s, 3H), 2.13 (s, 3H). LC-MS [M + H]+ = 360.1
1H NMR (400 MHz, DMSO_d6): δ 8.82 (s, 1H), 8.69-8.67 (m, 1H), 8.19 (d, J = 8.8 Hz, 1H), 7.85-7.82 (m, 1H), 7.55-7.52 (m, 1H), 7.31-7.29 (m, 1H), 6.26 (s, 1H), 4.18 (s, 3H), 2.13 (s, 3H). LC-MS [M + H]+ = 375.0
1H NMR (400 MHz, DMSO-d6): δ = 8.80-8.78 (m, 2H), 8.63 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.97-7.94 (m, 1H), 7.72-7.69 (m, 1H), 7.31-7.29 (m, 1H), 6.29 (d, J = 1.2 Hz, 1H), 4.17 (s, 3H). LC-MS [M + H]+ = 414.1
1H NMR (400 MHz, DMSO-d6) δ 8.99 (s, 1H), 8.94 (dd, J = 4.8, 1.4 Hz, 1H), 8.75 (d, J = 7.7 Hz, 1H), 8.23 (dd, J = 8.2, 1.4 Hz, 1H), 8.02 (dd, J = 8.1, 4.7 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 4.24 (s, 3H). LC-MS [M + H]+ = 414.1
1H NMR (400 MHz, DMSO-d6) δ 14.79 (s, 1H), 8.85 (s, 2H), 8.61 (d, J = 4.8 Hz, 1H), 7.84 (dd, J = 38.5, 8.0 Hz, 2H), 7.47 (dd, J = 7.9, 4.8 Hz, 1H), 2.14 (s, 3H). LC-MS [M + H]+ = 346.1
1H NMR (400 MHz, Chloroform-d) δ 8.82 (dd, J = 4.9, 1.6 Hz, 1H), 8.40 (s, 1H), 7.98 (d, J = 8.4 Hz, 1H), 7.64 (dd, J = 7.9, 1.6 Hz, 1H), 7.50 (dd, J = 7.9, 4.8 Hz, 1H), 7.37 (dd, J = 8.4, 1.9 Hz, 1H), 6.60 (d, J = 1.9 Hz, 1H), 2.36 (s, 3H). [M + H]+ = 311.1
1H NMR (400 MHz, DMSO-d6) δ 12.76 (s, 1H), 8.72 (s, 1H), 7.98 (d, J = 8.4 Hz, 1H), 7.70 (d, J = 30.5 Hz, 2H), 7.32 (d, J = 8.4 Hz, 1H), 6.11 (s, 1H), 5.52 (s, 2H), 4.17 (s, 3H). LC-MS [M + H]+ = 314.1
1H NMR (400 MHz, DMSO-d6) δ 8.86 (s, 1H), 8.61 (d, J = 7.9 Hz, 1H), 8.18 (s, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.47 (s, 1H), 6.85 (s, 1H), 5.59 (s, 2H), 4.19 (s, 3H). LC-MS [M + H]+ = 349.1
1H NMR (400 MHz, DMSO-d6) δ 12.47 (s, 1H), 8.86 (s, 1H), 8.62 (d, J = 7.9 Hz, 1H), 7.79 (d, J = 7.9 Hz, 1H), 7.53 (s, 1H), 6.04 (d, J = 2.2 Hz, 1H), 5.64 (s, 2H), 4.17 (s, 3H). LC-MS [M + H]+ = 349.1
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.84 (dd, J = 4.8, 1.5 Hz, 1H), 8.70 (d, J = 2.4 Hz, 1H), 8.29 (d, J = 8.1 Hz, 1H), 8.02 (dt, J = 8.1, 2.0 Hz, 1H), 7.80-7.74 (m, 1H), 7.67 (dd, J = 8.2, 1.6 Hz, 1H), 6.64 (d, J = 1.6 Hz, 1H), 4.23 (s, 3H). LC-MS [M + H]+ = 360.1
1H NMR (400 MHz, DMSO-d6) δ 8.78 (s, 1H), 8.02 (d, J = 8.3 Hz, 1H), 7.37- 7.29 (m, 2H), 6.88 (d, J = 8.3 Hz, 1H), 6.62-6.51 (m, 3H), 4.18 (s, 3H). LC-MS [M + H]+ = 326.1
1H NMR (400 MHz, CDCl3): δ 8.84 (dd, J1 = 4.8 Hz, J2 = 1.6 Hz, 1H), 8.60 (d, J = 2.4 Hz, 1H), 8.17 (s, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.74-7.69 (m, 1H), 764-7.59 (m, 1 H), 7.30-7.26 (m, 1 H), 6.60 (d, J = 2.0 Hz, 1H), 4.79 (t, J = 7.2 Hz, 2H), 2.37-2.20 (m, 2 H), 2.20 (s, 6 H), 2.14-2.05 (m, 2 H), LC-MS [M + H]+ = 382.0
1H NMR (400 MHz, CDCl3): δ 8.84 (d, J = 4.0 Hz, 1H), 8.61 (d, J = 2.4 Hz, 1H), 8.21 (s, 1H), 7.86 (d, J = 8.4 Hz, 1H), 7.74-7.70 (m, 1 H), 7.64- 7.59 (m, 1 H), 7.29-7.23 (m, 2 H), 6.60 (d, J = 1.6 Hz, 1H), 6.22 (d, J = 2.0 Hz, 1H), 6.02-5.93 (m, 2 H), 3.85 (s, 3 H). LC-MS [M + H]+ = 391.0
1H NMR (400 MHz, DMSO_d6): δ 9.82 (s, 1H), 8.77 (s, 1H), 8.10 (d, J = 8.4 Hz, 1 H), 7.43 (t, J = 8.0 Hz, 1 H), 7.25 (d, J = 8.4 Hz, 1 H), 6.97 (d, J = 8.0 Hz, 1 H), 6.76 (d, J = 8.0 Hz, 1 H), 6.72-6.71 (m, 1 H), 6.36 (s, 1H), 4.16 (s, 3H). LC-MS [M + H]+ = 376.0
1H NMR (400 MHz, CDCl3): δ 8.86 (dd, J1 = 4.8 Hz, J2 = 1.6 Hz, 1H), 8.61 (d, J = 2.0 Hz, 1H), 8.26 (s, 1H), 7.89 (d, J = 8.4 Hz, 1H), 7.74-7.69 (m, 1 H), 7.62 (dd, J1 = 8.0 Hz, J2 = 4.8 Hz, 1H), 7.31 (dd, J1 = 8.4, J2 = 2.0 Hz, 1H), 6.65 (d, J = 2.0 Hz, 1H), 6.41-6.09 (m, 1H), 5.20-5.07 (m, 2 H). LC-MS [M + H]+ = 360.9
1H NMR (400 MHz, DMSO-d6) δ 8.86 (s, 1H), 8.80 (dd, J = 4.8, 1.5 Hz, 1H), 8.63 (d, J = 2.4 Hz, 1H), 7.95 (dt, J = 8.1, 1.8 Hz, 1H), 7.84 (s, 1H), 7.73 (dd, J = 8.0, 4.8 Hz, 1H), 6.43 (s, 1H), 4.20 (s, 3H), 3.98 (s, 3H). LC-MS [M + H]+ = 341.1
1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J = 4.7 Hz, 1H), 8.69 (dd, J = 18.1, 2.9 Hz, 2H), 7.99 (d, J = 7.6 Hz, 1H), 7.75 (dd, J = 8.2, 4.7 Hz, 1H), 7.56- 7.47 (m, 1H), 6.28 (d, J = 1.7 Hz, 1H), 4.22 (d, J = 3.4 Hz, 3H). LC-MS [M + H]+ = 329.0
1H NMR (400 MHz, DMSO-d6) δ 8.69 (s, 1H), 8.00 (d, J = 8.3 Hz, 1H), 7.95 (d, J = 1.9 Hz, 1H), 7.37 (dd, J = 8.3, 1.8 Hz, 1H), 6.23 (dt, J = 13.5, 6.8 Hz, 1H), 4.37 (dt, J = 8.3, 5.3 Hz, 1H), 4.15 (s, 3H), 4.05 (dd, J = 9.6, 4.8 Hz, 1H), 3.96 (t, J = 9.5 Hz, 1H), 3.78 (q, J = 8.4 Hz, 1H), 2.26 (m, 2H). LCMS [M + H]+: 304.1.
1H NMR (400 MHz, Methanol-d4) δ 8.47 (s, 1H), 8.07 (d, J = 1.8 Hz, 1H), 7.95 (d, J = 8.3 Hz, 1H), 7.33 (dd, J = 8.4, 1.9 Hz, 1H), 6.47-6.35 (m, 1H), 4.52 (td, J = 8.5, 3.2 Hz, 1H), 4.21 (s, 4H), 4.07 (t, J = 9.7 Hz, 1H), 3.90 (td, J = 9.3, 6.9 Hz, 1H), 2.50-2.31 (m, 2H). LC-MS [M + H]+ = 340.1
1H NMR (400 MHz, DMSO-d6) δ 8.82 (s, 1H), 8.08 (d, J = 8.3 Hz, 1H), 7.42 (dd, J = 8.4, 2.0 Hz, 1H), 6.56 (d, J = 2.0 Hz, 1H), 6.29 (s, 1H), 4.19 (s, 3H), 3.46 (s, 3H), 2.30 (s, 3H). LC-MS [M + H]+ = 328.1
1H NMR (400 MHz, DMSO-d6) δ 8.81 (d, J = 4.8 Hz, 1H), 8.67 (s, 1H), 8.63 (d, J = 2.5 Hz, 1H), 7.96 (d, J = 8.3 Hz, 1H), 7.73 (dd, J = 8.1, 4.7 Hz, 1H), 7.10 (d, J = 1.8 Hz, 1H), 6.02 (d, J = 1.7 Hz, 1H), 4.20 (s, 3H), 4.10 (s, 3H). LC-MS [M + H]+ = 341.1.
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.72 (d, J = 7.9 Hz, 1H), 8.59 (dd, J = 4.9, 1.6 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.76 (dd, J = 7.9, 1.6 Hz, 1H), 7.46 (dd, J = 7.9, 4.8 Hz, 1H), 4.24 (s, 3H). LCMS m/z [M + H]+: 360.1.
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.72 (d, J = 7.9 Hz, 1H), 8.59 (dd, J = 4.9, 1.6 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.76 (dd, J = 7.9, 1.6 Hz, 1H), 7.46 (dd, J = 7.9, 4.8 Hz, 1H), 4.24 (s, 3H). LCMS m/z [M + H]+: 360.1.
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.72 (dd, J = 7.9, 0.8 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.74-7.69 (m, 1H), 7.56 (q, J = 2.2 Hz, 3H), 4.23 (s, 3H). LC-MS [M + H]+ = 379.1.
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.71 (dd, J = 8.0, 0.8 Hz, 1H), 8.54 (dd, J = 4.7, 1.6 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.72 (dd, J = 7.9, 1.6 Hz, 1H), 7.35 (dd, J = 7.9, 4.7 Hz, 1H), 4.23 (s, 3H), 1.57 (m, 1H), 1.08- 0.98 (m, 1H), 0.82-0.65 (m, 2H), 0.63-0.53 (m, 1H). LC-MS [M + H]+ = 386.1.
1H NMR (400 MHz, DMSO-d6) δ 8.93 (s, 1H), 8.67 (d, J = 7.9 Hz, 1H), 7.78 (d, J = 7.9 Hz, 1H), 7.46 (t, J = 8.0 Hz, 1H), 7.07 (dd, J = 8.3, 2.5 Hz, 1H), 6.99-6.86 (m, 2H), 4.22 (s, 3H), 3.79 (s, 3H). LC-MS [M + H]+ = 375.1
1H NMR (400 MHz, DMSO-d6) δ 8.77 (d, J = 3.0 Hz, 1H), 8.04 (dd, J = 8.4, 3.0 Hz, 1H), 7.32 (ddd, J = 10.7, 6.2, 2.5 Hz, 3H), 7.21 (dd, J = 9.0, 2.9 Hz, 2H), 6.51-6.47 (m, 1H), 4.18 (d, J = 2.9 Hz, 3H), 3.90 (d, J = 3.1 Hz, 3H). LC-MS [M + H]+ = 340.1
1H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H), 8.06 (d, J = 8.3 Hz, 1H), 7.52-7.44 (m, 5H), 7.35 (dd, J = 8.4, 2.0 Hz, 1H), 6.44 (d, J = 2.0 Hz, 1H), 4.19 (s, 3H). LC-MS [M + H]+ = 376.1
1H NMR (400 MHz, DMSO-d6) δ 8.94 (s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.62 (t, J = 8.4 Hz, 1H), 7.36-7.30 (m, 2H), 7.29-7.24 (m, 2H), 4.23 (s, 3H). LC-MS [M + H]+ = 411.1
1H NMR (400 MHz, Methanol-d4) δ 8.90 (s, 1H), 8.67 (d, J = 7.9 Hz, 1H), 8.62 (dd, J = 5.0, 1.6 Hz, 1H), 7.80 (dd, J = 8.0, 1.6 Hz, 1H), 7.74 (d, J = 7.9 Hz, 1H), 7.54 (dd, J = 7.9, 4.9 Hz, 1H), 4.40 (d, J = 7.3 Hz, 2H), 2.26 (s, 3H), 1.53 (tt, J = 7.7, 4.8 Hz, 1H), 0.82-0.71 (m, 2H), 0.60 (dt, J = 6.5, 4.7 Hz, 2H). LC-MS [M + H]+ = 400.2.
1H NMR (400 MHz, DMSO-d6) δ 8.94 (s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 7.86- 7.69 (m, 3H), 7.58 (d, J = 19.4 Hz, 2H), 7.21 (d, J = 55.6 Hz, 1H), 4.23 (s, 3H). LC-MS [M + H]+ = 395.2.
1H NMR (400 MHz, DMSO-d6) δ 8.77 (s, 1H), 8.64 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.66 (tt, J = 8.5, 6.3 Hz, 1H), 7.28 (t, J = 8.3 Hz, 2H), 4.32 (s, 3H). LC-MS [M + H]+ = 381.2
1H NMR (400 MHz, DMSO-d6) δ 8.99 (s, 1H), 8.75 (d, J = 7.9 Hz, 1H), 8.60 (dd, J = 4.8, 1.8 Hz, 1H), 8.14 (dd, J = 7.7, 1.8 Hz, 1H), 7.87 (d, J = 7.9 Hz, 1H), 7.72 (dd, J = 7.8, 4.8 Hz, 1H), 4.25 (s, 3H). LC-MS [M + H]+ = 380.1.
1H NMR (400 MHz, DMSO-d6) δ 9.07 (s, 1H), 8.73 (d, J = 7.9 Hz, 1H), 8.69 (dd, J = 4.9, 1.5 Hz, 1H), 8.60 (d, J = 2.4 Hz, 1H), 7.89 (dt, J = 8.2, 1.8 Hz, 1H), 7.83 (d, J = 7.9 Hz, 1H), 7.64 (dd, J = 8.1, 4.8 Hz, 1H), 4.37 (d, J = 7.3 Hz, 2H), 1.44 (ddt, J = 10.6, 7.6, 3.8 Hz, 1H), 0.66 (dt, J = 7.8, 3.0 Hz, 2H), 0.59-0.49 (m, 2H). LC-MS [M + H]+ = 386.2.
1H NMR δ 8.95 (s, 1H), 8.69 (d, J = 7.9 Hz, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.48-7.39 (m, 1H), 7.27 (dd, J = 7.7, 2.4 Hz, 2H), 4.23 (s, 3H). LC-MS [M + H]+ = 381.1.
1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 4.9 Hz, 1H), 8.63 (d, J = 7.9 Hz, 1H), 7.77 (d, J = 7.8 Hz, 1H), 7.13 (d, J = 7.9 Hz, 2H), 4.19 (s, 3H). LC- MS [M + H]+ = 397.1.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.69 (d, J = 7.9 Hz, 1H), 8.33 (dd, J = 5.0, 1.8 Hz, 1H), 7.89-7.75 (m, 2H), 7.22 (dd, J = 7.5, 5.0 Hz, 1H), 4.23 (s, 3H), 3.75 (s, 3H). LC-MS [M + H]+ = 376.1
1H NMR (400 MHz, DMSO-d6) δ 9.10 (s, 1H), 8.74 (d, J = 7.9 Hz, 1H), 8.69 (dd, J = 4.8, 1.5 Hz, 1H), 8.58 (d, J = 2.4 Hz, 1H), 7.95-7.80 (m, 2H), 7.70- 7.58 (m, 1H), 5.51-5.37 (m, 1H), 4.23-4.06 (m, 3H), 3.93 (td, J = 8.4, 5.4 Hz, 1H), 2.68-2.56 (m, 1H), 2.49-2.39 (m, 1H). LC-MS [M + H]+ = 402.2.
1H NMR (400 MHz, DMSO-d6) δ 8.80 (d, J = 1.8 Hz, 2H), 8.15 (t, J = 1.0 Hz, 1H), 8.07 (d, J = 8.3 Hz, 1H), 7.81-7.72 (m, 2H), 7.37 (dd, J = 8.4, 2.0 Hz, 1H), 6.94 (dd, J = 7.1, 2.0 Hz, 1H), 6.73 (d, J = 2.0 Hz, 1H), 4.19 (s, 3H). LC-MS [M + H]+ = 350.1
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.81 (s, 1H), 8.08 (dd, J = 8.3, 3.2 Hz, 2H), 8.00 (d, J = 1.8 Hz, 1H), 7.44 (dd, J = 8.4, 1.9 Hz, 1H), 7.35 (dd, J = 8.4, 2.0 Hz, 1H), 6.44 (d, J = 2.0 Hz, 1H), 4.20 (s, 3H). LC-MS [M + H]+ = 351.0
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.89 (s, 1H), 8.71 (d, J = 7.9 Hz, 1H), 7.98 (d, J = 8.4 Hz, 1H), 7.91 (d, J = 1.8 Hz, 1H), 7.81 (d, J = 7.9 Hz, 1H), 7.41 (dd, J = 8.4, 1.9 Hz, 1H), 4.25 (s, 3H). LC-MS [M + H]+ = 386.1
1H NMR (400 MHz, DMSO-d6) δ 9.14 (s, 1H), 8.73-8.66 (m, 2H), 8.58 (dd, J = 2.4, 0.8 Hz, 1H), 7.88 (ddd, J = 8.1, 2.5, 1.6 Hz, 1H), 7.84 (d, J = 7.9 Hz, 1H), 7.64 (ddd, J = 8.0, 0.8 Hz, 1H), 4.92-4.74 (m, 1H), 4.13-3.98 (m, 2H), 3.59 (td, J = 11.6, 2.4 Hz, 2H), 2.22-2.05 (m, 4H). LC-MS [M + H]+ = 416.2
1H NMR (400 MHz, DMSO-d6) δ 9.13 (d, J = 1.6 Hz, 1H), 8.73-8.67 (m, 1H), 8.62-8.57 (m, 1H), 7.84 (dd, J = 7.9, 1.7 Hz, 1H), 7.74 (dd, J = 7.9, 1.7 Hz, 1H), 7.45 (ddd, J = 7.6, 4.8, 1.5 Hz, 1H), 4.27-4.15 (m, 1H), 2.13 (d, J = 1.6 Hz, 3H), 1.38-1.31 (m, 2H), 1.26-1.18 (m, 2H). LC-MS [M + H]+ = 386.2.
1H NMR (400 MHz, DMSO-d6) δ 8.96 (s, 1H), 8.74-8.68 (m, 1H), 8.54 (dd, J = 4.7, 1.6 Hz, 1H), 7.82 (d, J = 7.9 Hz, 1H), 7.71 (dd, J = 7.9, 1.6 Hz, 1H), 7.35 (dd, J = 7.9, 4.7 Hz, 1H), 4.23 (s, 3H), 1.57 (tt, J = 8.4, 4.7 Hz, 1H), 1.10-0.97 (m, 1H), 0.84-0.64 (m, 2H), 0.65-0.48 (m, 1H). LCMS [M + H]+ = 386.2
1H NMR (400 MHz, DMSO-d6) δ 8.97 (s, 1H), 8.71 (d, J = 7.9 Hz, 1H), 8.54 (dd, J = 4.8, 1.6 Hz, 1H), 7.82 (d, J = 7.9 Hz, 1H), 7.71 (dd, J = 7.9, 1.7 Hz, 1H), 7.35 (dd, J = 7.9, 4.8 Hz, 1H), 4.24 (s, 3H), 1.56 (td, J = 8.1, 4.1 Hz, 1H), 1.10-0.95 (m, 1H), 0.84-0.65 (m, 2H), 0.66-0.51 (m, 1H). LCMS [M + H]+ = 386.2
1H NMR (400 MHz, DMSO-d6) δ 9.92 (s, 1H), 8.94 (s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.53 (t, J = 8.0 Hz, 1H), 7.37-7.28 (m, 1H), 7.18 (t, J = 2.0 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 4.23 (s, 3H), 3.00 (s, 3H). LC-MS [M + H]+ = 438.1.
1H NMR (400 MHz, DMSO-d6) δ 9.12 (d, J = 3.7 Hz, 1H), 8.69 (dt, J = 6.1, 3.2 Hz, 2H), 8.57 (d, J = 2.6 Hz, 1H), 7.90-7.78 (m, 2H), 7.64 (dd, J = 8.1, 4.7 Hz, 1H), 4.21 (dq, J = 7.6, 3.9 Hz, 1H), 1.31 (q, J = 4.1, 3.2 Hz, 2H), 1.22 (s, 2H). LC-MS [M + H]+ = 372.1.
1H NMR (400 MHz, DMSO-d6) δ 14.42 (s, 1H), 8.70 (dd, J = 4.9, 1.5 Hz, 1H), 8.66 (d, J = 8.0 Hz, 1H), 8.60 (d, J = 2.4 Hz, 1H), 7.90 (dt, J = 8.0, 2.0 Hz, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.65 (dd, J = 8.0, 4.8 Hz, 1H), 2.82 (s, 3H). LC-MS [M + H]+ = 346.1.
1H NMR (400 MHz, DMSO-d6) δ 8.93 (s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 8.53 (s, 1H), 8.36 (d, J = 2.2 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.69 (d, J = 2.2 Hz, 1H), 4.22 (s, 3H), 2.41 (s, 3H). LC-MS [M + H]+ = 360.1
1HNMR (400 MHz, DMSO-d6) δ 9.67 (s, 1H), 8.71 (d, J = 7.9 Hz, 1H), 8.07-7.92 (m, 2H), 7.86 (d, J = 7.9 Hz, 1H), 7.59 (dd, J = 8.2, 6.7 Hz, 2H), 7.52 (d, J = 7.3 Hz, 1H), 7.41-7.35 (m, 2H), 7.28-7.19 (m, 2H), 3.89 (s, 3H). LC-MS [M + H]+ = 437.2.
1H NMR (400 MHz, DMSO-d6) δ 8.96 (s, 1H), 8.70 (d, J = 7.8 Hz, 1H), 8.54 (d, J = 4.7 Hz, 1H), 8.06-7.94 (m, 1H), 7.87-7.76 (m, 2H), 7.66 (t, J = 7.8 Hz, 1H), 7.57-7.48 (m, 1H), 4.23 (s, 3H), 2.82 (d, J = 4.5 Hz, 3H). LC-MS [M + H]+ = 402.1.
1H NMR (400 MHz, DMSO-d6) δ 8.96 (s, 1H), 8.69 (dd, J = 8.0, 0.9 Hz, 1H), 7.95 (d, J = 7.7 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.43 (d, J = 7.6 Hz, 1H), 7.34 (d, J = 7.7 Hz, 1H), 4.23 (s, 3H), 2.54 (d, J = 2.0 Hz, 3H). LC-MS [M + H]+ = 360.1.
1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 1H), 8.94 (d, J = 3.4 Hz, 1H), 8.73-8.63 (m, 1H), 8.25 (d, J = 2.7 Hz, 1H), 8.01 (d, J = 2.1 Hz, 1H), 7.81 (dd, J = 8.0, 3.3 Hz, 1H), 7.24 (q, J = 2.6 Hz, 1H), 4.23 (d, J = 3.3 Hz, 3H). LC-MS [M + H]+ = 362.0.
1H NMR (400 MHz, DMSO-d6) δ 9.19 (s, 1H), 8.98 (s, 2H), 8.73 (d, J = 7.9 Hz, 1H), 8.58 (s, 1H), 7.87 (d, J = 7.9 Hz, 1H), 4.24 (s, 3H). LC-MS [M + H]+ = 371.1.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.72-8.67 (m, 1H), 8.44- 8.40 (m, 1H), 7.82 (d, J = 7.9 Hz, 1H), 7.73 (dd, J = 8.2, 2.5 Hz, 1H), 7.47 (d, J = 8.2 Hz, 1H), 4.23 (s, 3H), 2.61 (s, 3H). LC-MS [M + H]+ = 360.1.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (d, J = 2.4 Hz, 1H), 8.69 (dt, J = 7.7, 3.4 Hz, 1H), 8.14-8.07 (m, 1H), 7.98 (m, 1H), 7.80 (dd, J = 7.9, 2.5 Hz, 1H), 7.74 (td, J = 7.7, 2.6 Hz, 1H), 7.70-7.65 (m, 1H), 4.24 (t, J = 3.3 Hz, 3H), 3.90 (s, 3H). LC-MS [M + H]+ = 403.1.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 8.02 (d, J = 7.8 Hz 1H), 7.78 (d, J = 8.0 Hz, 2H), 7.53 (t, J = 7.8 Hz, 1H), 7.37 (d, J = 7.6 Hz, 1H), 4.23 (s, 3H). LC-MS [M + H]+ = 389.1.
1H NMR (400 MHz, DMSO_d6): δ 8.95 (s, 1H), 8.67 (d, J = 4.8 Hz, 1 H), 8.07 (d, J = 8.0 Hz, 1 H), 7.80 (d, J = 8.0 Hz, 1 H), 7.56-7.51 (m, 1 H), 7.37 (d, J = 8.0 Hz, 1 H), 6.29 (s, 1 H), 4.16-4.11 (m, 1 H), 2.14 (s, 3H), 1.30-1.26 (m, 2 H), 1.19-1.14 (m, 2H). LC-MS [M + H]+ = 351.0
1H NMR (400 MHz, DMSO-d6) δ 9.00 (s, 1H), 8.91 (d, J = 1.1 Hz, 1H), 8.85 (d, J = 1.4 Hz, 2H), 8.74 (d, J = 7.9 Hz, 1H), 7.86 (d, J = 8.0 Hz, 1H), 4.25 (s, 3H). LC-MS [M + H]+ = 347.1.
1H NMR (400 MHz, DMSO-d6) δ 10.07 (s, 1H), 8.93 (s, 1H), 8.76-8.60 (m, 1H), 7.79 (d, J = 7.9 Hz, 1H), 7.30 (dd, J = 11.2, 8.6 Hz, 1H), 6.92 (dd, J = 8.0, 2.5 Hz, 1H), 6.76 (ddd, J = 8.5, 4.0, 2.5 Hz, 1H), 4.22 (s, 3H). LC-MS [M + H]+ = 379.1.
1H NMR (400 MHz, DMSO-d6) δ 9.67 (d, J = 0.9 Hz, 1H), 8.86 (d, J = 2.8 Hz, 1H), 8.69 (d, J = 7.9 Hz, 1H), 8.38 (dd, J = 9.0, 2.9 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 7.59 (dd, J = 8.3, 6.7 Hz, 2H), 7.56-7.49 (m, 1H), 7.42- 7.36 (m, 2H), 7.18-7.12 (m, 1H), 3.99 (d, J = 0.8 Hz, 3H). LC-MS [M + H]+ = 438.4.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.69 (d, J = 8.0 Hz, 1H), 8.07- 7.99 (m, 2H), 7.85 (t, J = 1.9 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.65 (t, J = 7.8 Hz, 1H), 7.53 (ddd, J = 7.8, 2.0, 1.1 Hz, 1H), 7.46 (s, 1H), 4.23 (s, 3H). LC-MS [M + H]+ = 388.3
1H NMR (400 MHz, DMSO-d6) δ 12.75 (s, 1H), 8.71 (s, 1H), 7.98 (d, J = 8.3 Hz, 1H), 7.75-7.64 (m, 2H), 7.44 (s, 1H), 7.33 (dd, J = 8.3, 1.8 Hz, 1H), 5.42 (s, 2H), 4.16 (s, 3H). LC-MS [M + H]+ = 314.1.
1H NMR (400 MHz, DMSO-d6) δ 7.87 (d, J = 8.0 Hz, 1H), 7.70-7.54 (m, 5H), 7.54-7.47 (m, 1H), 7.38-7.32 (m, 2H), 7.32-7.25 (m, 2H), 3.97 (s, 3H), 3.93 (s, 3H). LC-MS [M + H]+ = 451.3.
1H NMR (400 MHz, DMSO-d6) δ 8.94 (d, J = 8.0 Hz, 1H), 7.87 (d, J = 8.1 Hz, 1H), 7.57 (t, J = 7.5 Hz, 2H), 7.50 (t, J = 7.3 Hz, 1H), 7.35 (d, J = 7.5 Hz, 2H), 4.21 (s, 3H). LC-MS [M + H]+ = 423.2
1H NMR (400 MHz, DMSO-d6) δ 8.85 (s, 1H), 8.60 (d, J = 7.9 Hz, 1H), 7.63 (d, J = 96.0 Hz, 2H), 6.88 (s, 1H), 6.16 (d, J = 2.0 Hz, 1H), 4.19 (s, 3H), 3.98 (s, 1H), 1.97 (d, J = 7.1 Hz, 3H). LC-MS [M + H]+ = 363.2.
1H NMR (400 MHz, DMSO-d6) δ 13.22 (s, 1H), 8.73 (d, J = 12.8 Hz, 2H), 7.76 (d, J = 7.9 Hz, 1H), 7.74-7.66 (m, 2H), 7.56-7.49 (m, 1H), 7.11 (d, J = 7.2 Hz, 1H). LC-MS m/z [M + H]+ = 371.2.
1H NMR (400 MHz, DMSO-d6) δ 8.93 (d, J = 9.8 Hz, 2H), 8.80 (s, 1H), 8.69 (d, J = 7.9 Hz, 1H), 8.19 (s, 1H), 7.82 (d, J = 7.9 Hz, 1H), 7.30 (t, J = 55.1 Hz, 1H), 4.23 (s, 3H). LC-MS [M + H]+ = 396.3.
1H NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 3.3 Hz, 1H), 8.56-8.42 (m, 2H), 8.05 (d, J = 8.4 Hz, 1H), 7.88-7.73 (m, 1H), 7.59 (ddd, J = 8.0, 4.8, 0.8 Hz, 1H), 7.32 (t, J = 5.8 Hz, 1H), 6.53 (d, J = 8.5 Hz, 1H), 5.58 (tt, J = 57.1, 4.4 Hz, 1H), 4.16 (s, 3H), 3.27-3.05 (m, 2H). LC-MS [M + H]+ = 357.3.
1H NMR (400 MHz, DMSO-d6) δ 8.73-8.62 (m, 2H), 8.57 (dd, J = 2.4, 0.8 Hz, 1H), 8.33 (d, J = 8.4 Hz, 1H), 7.87 (ddd, J = 8.0, 2.5, 1.6 Hz, 1H), 7.62 (ddd, J = 8.0, 4.8, 0.8 Hz, 1H), 6.74 (d, J = 8.3 Hz, 1H), 4.19 (s, 3H), 3.85 (q, J = 7.0 Hz, 2H), 1.06 (t, J = 7.0 Hz, 3H). LC-MS [M + H]+ = 322.2.
1H NMR (400 MHz, DMSO-d6) δ 8.68 (dd, J = 4.8, 1.5 Hz, 1H), 8.60 (d, J = 8.0 Hz, 1H), 8.57-8.52 (m, 1H), 7.85 (ddd, J = 8.1, 2.5, 1.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (ddd, J = 8.1, 4.8, 0.8 Hz, 1H), 5.73 (t, J = 6.7 Hz, 1H), 4.04 (s, 3H), 3.42 (t, J = 6.9 Hz, 2H), 3.38 (s, 2H), 3.16 (q, J = 6.9 Hz, 2H), 2.49-2.41 (m, 2H), 1.79 (p, J = 7.0 Hz, 2H), 1.70-1.59 (m, 2H), 1.52 (d, J = 5.4 Hz, 4H). LC-MS [M + H]+ = 514.4.
1H NMR (400 MHz, DMSO-d6) δ 8.84 (s, 1H), 8.69 (dd, J = 4.8, 1.5 Hz, 1H), 8.57 (d, J = 2.5 Hz, 1H), 8.50 (d, J = 8.1 Hz, 1H), 7.87 (ddd, J = 8.1, 2.5, 1.6 Hz, 1H), 7.69-7.57 (m, 1H), 7.44 (d, J = 8.1 Hz, 1H), 4.21 (s, 3H). LC-MS [M + H]+ = 312.1
1H NMR (400 MHz, DMSO-d6) δ 13.24 (s, 1H), 9.61 (s, 1H), 8.86 (d, J = 7.9 Hz, 1H), 8.28 (t, J = 58.8 Hz, 1H), 7.91-7.81 (m, 2H), 7.69 (d, J = 8.4 Hz, 1H), 7.52 (dd, J = 8.4, 7.2 Hz, 1H), 7.14 (d, J = 7.2 Hz, 1H). LC-MS [M + H]+ = 421.2.
1H NMR (400 MHz, DMSO-d6) δ 13.23 (s, 1H), 9.14 (s, 1H), 8.69 (d, J = 7.9 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.74 (s, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.52 (t, J = 7.8 Hz, 1H), 7.09 (d, J = 7.2 Hz, 1H), 4.22 (tt, J = 7.5, 3.9 Hz, 1H), 1.33 (q, J = 4.2 Hz, 2H), 1.23 (dd, J = 7.5, 5.0 Hz, 2H). LC-MS [M + H]+ = 411.3
1H NMR (400 MHz, DMSO-d6) δ 8.63 (d, J = 4.9 Hz, 1H), 8.50-8.45 (m, 2H), 7.97-7.92 (m, 1H), 7.78 (dt, J = 8.0, 2.0 Hz, 1H), 6.84 (t, J = 5.5 Hz, 1H), 6.39 (d, J = 8.7 Hz, 1H), 4.14 (s, 3H), 2.88-2.78 (m, 2H), 0.84 (t, J = 7.1 Hz, 3H). LC-MS [M + H]+ = 321.2
1H NMR (400 MHz, DMSO-d6) δ 8.76 (d, J = 8.0 Hz, 1H), 8.70 (dd, J = 4.8, 1.5 Hz, 1H), 8.60 (d, J = 2.5 Hz, 1H), 8.32 (t, J = 57.0 Hz, 1H), 7.89 (dt, J = 8.2, 1.8 Hz, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.65 (dd, J = 8.1, 4.8 Hz, 1H), 3.06 (s, 3H). LC-MS [M + H]+ = 396.2.
1H NMR (400 MHz, DMSO-d6) δ 8.72 (s, 1H), 8.69 (dd, J = 4.8, 1.5 Hz, 1H), 8.59 (dd, J = 2.5, 0.8 Hz, 1H), 8.43 (d, J = 8.4 Hz, 1H), 7.89 (ddd, J = 8.0, 2.5, 1.5 Hz, 1H), 7.64 (ddd, J = 8.1, 4.8, 0.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.06 (tt, J = 55.2, 3.9 Hz, 1H), 4.20 (s, 3H), 4.06 (td, J = 14.5, 3.8 Hz, 3H). LC-MS [M + H]+ = 358.3.
1H NMR (400 MHz, DMSO-d6) δ 13.28 (s, 1H), 8.82 (d, J = 8.0 Hz, 1H), 8.37 (t, J = 56.9 Hz, 1H), 7.88-7.80 (m, 2H), 7.73 (d, J = 8.5 Hz, 1H), 7.57 (t, J = 7.9 Hz, 1H), 7.17 (d, J = 7.3 Hz, 1H), 3.13 (s, 3H). LC-MS [M + H]+ = 435.3.
1H NMR (400 MHz, DMSO-d6) δ 9.59 (s, 1H), 8.85 (d, J = 7.9 Hz, 1H), 8.71 (dd, J = 4.8, 1.5 Hz, 1H), 8.61 (dd, J = 2.4, 0.8 Hz, 1H), 8.28 (t, J = 58.8 Hz, 1H), 7.92-7.88 (m, 2H), 7.65 (ddd, J = 8.1, 4.8, 0.8 Hz, 1H). LC-MS [M + H]+ = 382.3.
1H NMR (400 MHz, DMSO-d6) δ 13.28-13.13 (m, 1H), 8.97 (s, 1H), 8.71 (d, J = 7.9 Hz, 1H), 7.78 (d, J = 7.9 Hz, 1H), 7.72 (t, J = 1.2 Hz, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.52 (dd, J = 8.4, 7.2 Hz, 1H), 7.11 (d, J = 7.2 Hz, 1H). LC- MS m/z [M + H]+ = 388.3.
1H NMR (400 MHz, DMSO-d6) δ 13.49 (s, 1H), 8.82 (s, 1H), 8.22 (d, J = 9.6 Hz, 1H), 7.80 (d, J = 9.2 Hz, 2H), 7.62 (t, J = 7.8 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 6.40 (d, J = 6.5 Hz, 1H), 4.21 (s, 3H). LC-MS m/z [M + H]+: 368.2.
1H NMR (400 MHz, DMSO-d6) δ 13.50 (s, 1H), 8.83 (d, J = 20.9 Hz, 2H), 7.86-7.75 (m, 2H), 7.62 (t, J = 7.8 Hz, 1H), 7.22 (d, J = 7.2 Hz, 1H), 6.49 (s, 1H), 4.23 (s, 3H). LC-MS [M + H]+: 375.3.
1H NMR (400 MHz, DMSO-d6) δ 13.49 (s, 1H), 9.45 (s, 1H), 8.44-8.11 (m, 2H), 7.92 (s, 1H), 7.82 (d, J = 8.4 Hz, 1H), 7.63 (dd, J = 8.4, 7.2 Hz, 1H), 7.23 (d, J = 7.2 Hz, 1H), 6.42 (d, J = 6.4 Hz, 1H). LC-MS [M + H]+: 404.1.
1H NMR (400 MHz, DMSO-d6) δ 13.47 (s, 1H), 8.90 (s, 1H), 8.30 (d, J = 2.4 Hz, 1H), 8.23 (d, J = 9.6 Hz, 1H), 7.85-7.73 (m, 2H), 7.67 (dd, J = 8.8, 2.5 Hz, 1H), 7.63-7.58 (m, 1H), 7.17 (d, J = 7.2 Hz, 1H), 6.92 (d, J = 8.8 Hz, 1H), 6.36 (d, J = 6.5 Hz, 1H), 5.57 (s, 2H), 3.56 (s, 4H), 2.55 (s, 3H), 2.35 (d, J = 5.8 Hz, 4H). [M + H]+: 543.2
1H NMR (400 MHz, DMSO-d6) δ 13.52 (s, 1H), 8.85 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 7.86 (s, 1H), 7.82 (d, J = 8.4 Hz, 1H), 7.62 (dd, J = 8.4, 7.2 Hz, 1H), 7.23 (d, J = 7.2 Hz, 1H), 6.69 (d, J = 2.1 Hz, 1H), 4.24 (s, 3H). LC-MS [M + H]+ = 351.1.
1H NMR (400 MHz, DMSO-d6) δ 13.24 (s, 1H), 8.93 (s, 1H), 8.83 (d, J = 10.3 Hz, 1H), 7.76 (s, 1H), 7.68 (d, J = 8.3 Hz, 1H), 7.57-7.47 (m, 1H), 7.12 (d, J = 7.2 Hz, 1H), 4.26 (s, 3H). LC-MS [M + H]+ = 403.1.
1H NMR (400 MHz, DMSO-d6) δ 13.06 (s, 1H), 8.83 (s, 1H), 8.10 (d, J = 8.4 Hz, 1H), 7.72 (d, J = 8.4 Hz, 1H), 7.58 (dd, J = 8.4, 7.2 Hz, 1H), 7.36 (dd, J = 8.4, 2.0 Hz, 1H), 7.12 (d, J = 7.1 Hz, 1H), 6.29 (d, J = 2.0 Hz, 1H), 4.21 (s, 3H), 1.80 (s, 3H). LC-MS [M + H]+ = 364.2.
1H NMR (400 MHz, DMSO-d6) δ 9.06 (s, 1H), 8.82 (s, 1H), 7.78-7.73 (m, 2H), 7.64-7.55 (m, 2H), 7.17 (d, J = 7.2 Hz, 1H), 4.31 (s, 3H). LCMS m/z [M + H]+: 410.1.
1H NMR (400 MHz, DMSO-d6) δ 13.53 (s, 1H), 9.05 (s, 1H), 8.78 (s, 1H), 7.87 (s, 1H), 7.82 (d, J = 8.4 Hz, 1H), 7.65-7.59 (m, 1H), 7.21 (d, J = 7.2 Hz, 1H), 6.47 (s, 1H), 4.22 (dq, J = 7.6, 3.8 Hz, 1H), 1.31-1.28 (m, 2H), 1.24-1.20 (m, 2H). LCMS m/z [M + H]+ = 401.1.
1H NMR (400 MHz, DMSO-d6) δ 13.52 (s, 1H), 8.95 (s, 1H), 8.83 (s, 1H), 7.89 (s, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.61 (t, J = 7.8 Hz, 1H), 7.46-7.40 (m, 2H), 7.21 (d, J = 7.1 Hz, 1H), 7.04-6.99 (m, 2H), 6.45 (s, 1H), 5.64 (s, 2H), 3.78 (s, 3H). LCMS [M + H]+ = 481.1.
1H NMR (400 MHz, DMSO-d6) δ 13.52 (s, 1H), 8.93 (s, 1H), 8.87 (s, 1H), 7.88 (s, 1H), 7.82 (d, J = 8.4 Hz, 1H), 7.73 (d, J = 2.2 Hz, 1H), 7.62 (t, J = 7.8 Hz, 1H), 7.21 (d, J = 7.2 Hz, 1H), 6.46 (s, 1H), 6.35 (d, J = 2.2 Hz, 1H), 5.64 (s, 2H), 3.87 (s, 3H). LCMS [M + H]+ = 455.1
1H NMR (400 MHz, DMSO-d6) δ 13.88 (s, 1H), 8.82 (s, 1H), 8.09 (d, J = 8.3 Hz, 1H), 7.86 (d, J = 8.5 Hz, 1H), 7.70 (t, J = 7.9 Hz, 1H), 7.34 (dd, J = 8.3, 2.1 Hz, 1H), 7.27 (d, J = 7.2 Hz, 1H), 6.26 (d, J = 2.0 Hz, 1H), 4.20 (s, 3H). LCMS [M + H]+ = 428.2.
1H NMR (400 MHz, DMSO-d6) δ 13.49 (s, 1H), 9.00 (s, 1H), 8.39 (d, J = 4.9 Hz, 1H), 8.30 (d, J = 9.5 Hz, 1H), 7.89-7.76 (m, 2H), 7.62 (t, J = 7.9 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 6.41 (dd, J = 10.2, 5.7 Hz, 2H), 5.65 (s, 2H), 3.75 (t, J = 4.9 Hz, 4H), 2.37 (t, J = 4.9 Hz, 4H), 2.23 (s, 3H). LCMS [M + H]+ = 544.1
1H NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 4.7 Hz, 1H), 8.28 (t, J = 5.0 Hz, 2H), 7.77 (d, J = 7.9 Hz, 1H), 7.46 (dd, J = 7.9, 4.8 Hz, 1H), 6.62 (d, J = 6.5 Hz, 1H), 4.27 (s, 3H), 1.51 (dt, J = 8.2, 3.7 Hz, 1H), 1.07 (d, J = 6.5 Hz, 1H), 0.88 (s, 1H), 0.80 (d, J = 8.3 Hz, 1H), 0.69 (d, J = 8.1 Hz, 1H). LCMS [M + H]+ = 369.1
1H NMR (400 MHz, DMSO-d6) δ 9.43 (s, 1H), 8.67 (dd, J = 4.8, 1.6 Hz, 1H), 8.44-8.07 (m, 2H), 7.84 (dd, J = 7.9, 1.6 Hz, 1H), 7.46 (dd, J = 7.9, 4.7 Hz, 1H), 6.51 (d, J = 6.4 Hz, 1H), 1.73 (dt, J = 8.1, 3.6 Hz, 1H), 1.11-0.87 (m, 2H), 0.87-0.66 (m, 2H). LCMS [M + H]+ = 405.1
1H NMR (400 MHz, DMSO-d6) δ 9.45 (s, 1H), 9.03 (d, J = 4.6 Hz, 1H), 8.41-8.07 (m, 4H), 6.72 (d, J = 6.1 Hz, 1H). LCMS [M + H]+ = 433.1
1H NMR (400 MHz, DMSO-d6) δ 8.78 (s, 1H), 8.62 (dd, J = 4.8, 1.7 Hz, 1H), 8.21 (d, J = 9.6 Hz, 1H), 7.77 (dd, J = 7.9, 1.7 Hz, 1H), 7.42 (dd, J = 7.9, 4.7 Hz, 1H), 6.44 (d, J = 6.4 Hz, 1H), 4.18 (s, 3H), 1.56 (d, J = 4.8 Hz, 1H), 1.02 (d, J = 4.6 Hz, 1H), 0.86 (d, J = 3.7 Hz, 1H), 0.82-0.72 (m, 1H), 0.69 (d, J = 7.9 Hz, 1H). LCMS [M + H]+ = 369.1
1H NMR (400 MHz, DMSO-d6) δ 8.86 (s, 1H), 8.25 (dd, J = 14.8, 8.9 Hz, 2H), 8.18 (d, J = 8.2 Hz, 1H), 7.80 (s, 1H), 7.65 (dd, J = 14.5, 7.4 Hz, 2H), 7.49 (t, J = 7.6 Hz, 1H), 7.34 (d, J = 8.5 Hz, 1H), 6.28 (d, J = 6.5 Hz, 1H), 4.23 (s, 3H). LCMS [M + H]+ = 378.1
1H NMR (400 MHz, DMSO-d6) δ 8.82 (s, 1H), 8.78 (dd, J = 4.2, 1.7 Hz, 1H), 8.60 (dd, J = 8.4, 1.7 Hz, 1H), 8.29 (dd, J = 8.2, 1.5 Hz, 1H), 8.21 (d, J = 9.6 Hz, 1H), 7.96 (dd, J = 7.3, 1.5 Hz, 1H), 7.92-7.86 (m, 1H), 7.65 (dd, J = 8.3, 4.2 Hz, 1H), 6.32 (d, J = 6.5 Hz, 1H), 4.22 (s, 3H). LCMS [M + H]+ = 379.1
1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 16.8 Hz, 2H), 8.22 (dd, J = 24.4, 8.3 Hz, 2H), 7.80 (t, J = 7.8 Hz, 1H), 7.73-7.61 (m, 2H), 7.50 (t, J = 7.7 Hz, 1H), 7.38 (d, J = 8.4 Hz, 1H), 6.38 (s, 1H), 4.25 (s, 3H). LCMS [M + H]+ = 385.1
1H NMR (400 MHz, DMSO-d6) δ 8.35-8.28 (m, 2H), 8.24 (d, J = 8.3 Hz, 1H), 8.18 (d, J = 8.3 Hz, 1H), 7.81 (t, J = 7.8 Hz, 1H), 7.65 (t, J = 6.9 Hz, 2H), 7.49 (t, J = 7.6 Hz, 1H), 7.25 (d, J = 8.5 Hz, 1H), 6.44 (d, J = 6.6 Hz, 1H), 4.31 (s, 3H). LCMS [M + H]+ = 378.1
1H NMR (400 MHz, DMSO-d6) δ 8.99 (dd, J = 4.8, 1.4 Hz, 1H), 8.79 (s, 1H), 8.24 (dd, J = 8.2, 1.4 Hz, 1H), 8.20 (d, J = 9.5 Hz, 1H), 8.05 (dd, J = 8.1, 4.7 Hz, 1H), 6.60 (d, J = 6.3 Hz, 1H), 4.18 (s, 3H). LCMS [M + H]+ = 397.0
1H NMR (400 MHz, DMSO-d6) δ 8.99 (dd, J = 4.8, 1.4 Hz, 1H), 8.79 (s, 1H), 8.24 (dd, J = 8.2, 1.4 Hz, 1H), 8.20 (d, J = 9.5 Hz, 1H), 8.05 (dd, J = 8.1, 4.7 Hz, 1H), 6.60 (d, J = 6.3 Hz, 1H), 4.18 (s, 3H). LCMS [M + H]+ = 397.0
1H NMR (400 MHz, DMSO-d6) δ 13.52 (s, 1H), 8.59 (s, 1H), 7.82 (d, J = 6.7 Hz, 2H), 7.63 (t, J = 7.8 Hz, 1H), 7.22 (d, J = 7.2 Hz, 1H), 6.51 (s, 1H), 4.14 (s, 3H), 2.91 (s, 3H). LCMS [M + H]+ = 389.1
1H NMR (400 MHz, DMSO-d6) δ 14.69 (s, 1H), 13.52 (s, 1H), 8.94 (s, 1H), 8.80 (s, 1H), 7.90 (s, 1H), 7.83 (d, J = 8.4 Hz, 1H), 7.63 (dd, J = 8.4, 7.3 Hz, 1H), 7.25 (d, J = 7.2 Hz, 1H), 6.53 (s, 1H). LCMS [M + H]+ = 361.3
1H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.59 (s, 1H), 7.87 (d, J = 8.4 Hz, 1H), 7.80 (s, 1H), 7.70 (dd, J = 8.5, 7.2 Hz, 1H), 7.25 (d, J = 7.1 Hz, 1H), 6.67 (s, 1H), 4.38 (d, J = 7.3 Hz, 2H), 1.59-1.46 (m, 1H), 0.98-0.86 (m, 1H), 0.82-0.71 (m, 2H), 0.65-0.55 (m, 2H). LCMS [M + H]+ = 415.1
1H NMR (400 MHz, DMSO-d6) δ 9.41 (s, 1H), 8.64 (dd, J = 4.8, 1.6 Hz, 1H), 8.42-8.08 (m, 2H), 7.81 (dd, J = 7.9, 1.6 Hz, 1H), 7.44 (dd, J = 7.9, 4.7 Hz, 1H), 6.48 (d, J = 6.4 Hz, 1H), 1.76-1.64 (m, 1H), 1.10-0.99 (m, 1H), 0.94- 0.65 (m, 3H). LCMS [M + H]+ = 405.1
1H NMR (400 MHz, DMSO-d6) δ 9.41 (s, 1H), 8.64 (dd, J = 4.6, 1.6 Hz, 1H), 8.42-8.02 (m, 2H), 7.81 (dd, J = 7.9, 1.6 Hz, 1H), 7.44 (dd, J = 7.9, 4.7 Hz, 1H), 6.48 (d, J = 6.4 Hz, 1H), 1.70 (tt, J = 8.4, 4.6 Hz, 1H), 1.09-0.99 (m, 1H), 0.88 (d, J = 4.8 Hz, 1H), 0.84-0.75 (m, 1H), 0.76-0.65 (m, 1H). LCMS [M + H]+ = 405.1
1H NMR (400 MHz, DMSO-d6) δ 8.66 (s, 1H), 8.31 (d, J = 8.4 Hz, 1H), 7.62- 7.14 (m, 5H), 6.72 (d, J = 8.4 Hz, 1H), 4.18 (s, 3H), 3.88 (q, J = 7.0 Hz, 2H), 1.06 (t, J = 7.0 Hz, 3H). LCMS [M + H]+ = 387.3
1H 1H NMR (400 MHz, DMSO-d6) δ 9.04 (dd, J = 4.3, 1.7 Hz, 1H), 8.90 (d, J = 8.2 Hz, 1H), 8.46 (dd, J = 8.4, 1.7 Hz, 1H), 8.37 (s, 1H), 8.19 (d, J = 8.9 Hz, 1H), 8.03 (d, J = 2.3 Hz, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.71 (dd, J = 8.9, 2.3 Hz, 1H), 7.64 (dd, J = 8.3, 4.2 Hz, 1H), 4.31 (s, 3H). LCMS [M + H]+: 396.2.
1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 8.3 Hz, 1H), 8.65 (d, J = 8.5 Hz, 1H), 8.40 (s, 1H), 8.24-8.16 (m, 1H), 8.05 (d, J = 8.4 Hz, 1H), 7.92- 7.83 (m, 2H), 7.78 (ddd, J = 8.1, 6.9, 1.2 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 4.32 (s, 3H). LCMS [M + H]+: 396.2.
1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 8.2 Hz, 1H), 8.85 (d, J = 2.4 Hz, 1H), 8.48 (d, J = 2.4 Hz, 1H), 8.39 (s, 1H), 8.18 (d, J = 8.4 Hz, 1H), 8.10 (d, J = 8.2 Hz, 1H), 7.95-7.84 (m, 2H), 7.74 (t, J = 7.5 Hz, 1H), 4.32 (s, 3H). LCMS [M + H]+ = 396.2
1H NMR (400 MHz, DMSO-d6) δ 8.89 (d, J = 8.2 Hz, 1H), 8.37 (s, 1H), 8.09 (t, J = 8.4 Hz, 2H), 8.02-7.98 (m, 1H), 7.93 (d, J = 2.0 Hz, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.63 (pd, J = 6.9, 1.5 Hz, 2H), 7.44 (dd, J = 8.6, 2.0 Hz, 1H), 4.31 (s, 3H). LCMS [M + H]+: 395.2.
1H NMR (400 MHz, DMSO-d6) δ 9.17 (dd, J = 4.2, 1.6 Hz, 1H), 8.93 (d, J = 8.2 Hz, 1H), 8.72 (d, J = 8.7 Hz, 1H), 8.50 (d, J = 8.3 Hz, 1H), 8.40 (s, 1H), 7.94 (d, J = 8.7 Hz, 1H), 7.91 (dd, J = 8.5, 4.1 Hz, 1H), 7.87 (d, J = 8.2 Hz, 1H), 4.33 (s, 3H). LCMS [M + H]+: 397.1.
1H NMR (400 MHz, DMSO-d6) δ 9.51 (d, J = 0.8 Hz, 1H), 8.94 (d, J = 8.2 Hz, 1H), 8.54 (s, 1H), 8.41 (s, 1H), 8.38-8.29 (m, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.73 (dddd, J = 25.2, 8.2, 6.9, 1.2 Hz, 2H), 7.44-7.37 (m, 1H), 4.34 (s, 3H). LCMS [M + H]+: 396.1.
1H NMR (400 MHz, DMSO-d6) δ 8.94 (d, J = 8.3 Hz, 1H), 8.79 (d, J = 0.9 Hz, 1H), 8.57 (d, J = 5.7 Hz, 1H), 8.41 (s, 1H), 8.18 (d, J = 8.3 Hz, 1H), 8.04- 7.96 (m, 2H), 7.84 (d, J = 8.2 Hz, 1H), 7.69 (dd, J = 7.3, 1.0 Hz, 1H), 4.34 (s, 3H). LCMS [M + H]+: 396.2.
1H NMR (400 MHz, DMSO-d6) δ 8.98 (dd, J = 4.2, 1.6 Hz, 1H), 8.93 (d, J = 8.2 Hz, 1H), 8.41 (s, 1H), 8.22 (d, J = 8.5 Hz, 1H), 7.97 (dd, J = 8.5, 7.4 Hz, 1H), 7.82 (dd, J = 9.7, 7.8 Hz, 2H), 7.65 (dd, J = 7.4, 1.1 Hz, 1H), 7.44 (dd, J = 8.6, 4.1 Hz, 1H), 4.34 (s, 3H). LCMS [M + H]+ = 396.2.
1H NMR (400 MHz, DMSO-d6) δ 8.89 (s, 1H), 8.83 (d, J = 7.9 Hz, 1H), 7.87 (d, J = 7.9 Hz, 1H), 7.76-7.71 (m, 1H), 7.58 (m, 4H). LCMS [M + H]+: 365.1.
1H NMR (400 MHz, DMSO-d6) δ 8.88 (d, J = 8.0 Hz, 1H), 8.72 (s, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.82-7.70 (m, 1H), 7.58 (q, J = 2.6, 1.8 Hz, 3H), 4.36 (s, 3H). LCMS [M + H]+: 379.2.
1H NMR (400 MHz, DMSO-d6) δ 13.01 (s, -1H), 8.83 (s, 1H), 8.24 (d, J = 9.5 Hz, 1H), 7.75 (dd, J = 8.2, 2.2 Hz, 1H), 7.72-7.69 (m, 1H), 7.35-7.25 (m, 1H), 6.54 (d, J = 6.5 Hz, 1H), 4.21 (s, 3H). LCMS m/z [M + H]+ = 386.0.
1H NMR (400 MHz, DMSO-d6) δ 13.73 (s, 1H), 8.82 (s, 1H), 8.23 (d, J = 9.5 Hz, 1H), 7.84 (d, J = 8.5 Hz, 1H), 7.70 (dd, J = 8.6, 7.2 Hz, 1H), 7.30 (d, J = 7.2 Hz, 1H), 6.41 (d, J = 6.4 Hz, 1H), 4.21 (s, 3H). LCMS [M + H]+: 402.0.
1H NMR (400 MHz, DMSO-d6) δ 13.86 (s, 1H), 8.81 (s, 1H), 8.22 (d, J = 9.5 Hz, 1H), 7.86 (d, J = 8.5 Hz, 1H), 7.69 (dd, J = 8.6, 7.2 Hz, 1H), 7.28 (d, J = 7.1 Hz, 1H), 6.36 (d, J = 6.5 Hz, 1H), 4.21 (s, 3H). LCMS [M + H]+ = 446.0
1H NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 33.0 Hz, 2H), 8.12 (d, J = 7.8 Hz, 2H), 7.70-7.66 (m, 2H), 7.60 (d, J = 7.7 Hz, 1H), 7.55 (d, J = 8.2 Hz, 1H), 7.50 (d, J = 7.2 Hz, 1H), 4.22 (s, 3H), 1.98 (s, 1H), 1.02-0.79 (m, 4H). LCMS [M + H]+ = 385.1
1H NMR (400 MHz, DMSO-d6) δ 14.60 (s, 1H), 8.73 (s, 1H), 8.67 (dd, J = 4.8, 1.6 Hz, 1H), 8.36 (d, J = 9.5 Hz, 1H), 7.84 (dd, J = 7.9, 1.6 Hz, 1H), 7.46 (dd, J = 7.9, 4.7 Hz, 1H), 6.53 (d, J = 6.4 Hz, 1H), 1.61 (tt, J = 8.4, 4.7 Hz, 1H), 1.06 (ddt, J = 9.1, 6.2, 3.3 Hz, 1H), 0.90 (ddq, J = 7.1, 4.8, 3.0 Hz, 1H), 0.86-0.76 (m, 1H), 0.72 (tdd, J = 8.9, 6.4, 2.8 Hz, 1H). LCMS m/z [M + H]+ = 355.1
MAT2A catalyzes the reaction of L-methionine and ATP to generate S-adenosylmethionine (SAM), inorganic phosphonic acid, and inorganic diphosphonic acid. Malachite green reacts with inorganic phosphonic acid to generate a green product, and the enzyme activity of MAT2A can be indirectly reflected by detecting the content of the inorganic phosphonic acid in a sample.
The test compounds were formulated using DMSO and diluted 1:3 diluted through 10 concentration points to give a final starting concentration in the reaction system of 1000 nM. The test compounds at different concentrations described above were transferred to a 384-well plate. Experimental buffer (50 mM HEPES pH 7.5, 50 mM KCl, 10 mM MgCl2, 10 mM DTT) was prepared. 2×MAT2A recombinant protein was prepared using the buffer and added to a 384-well plate. A mixed solution of 2×L-methionine and ATP was prepared using the buffer, and added to a 384-well plate. The enzymatic reaction was started, and the reaction was carried out at room temperature for 60 min. 20 μL of a colorimetric detection reagent was added to the above system, and incubated at room temperature for 15 min. The absorbance at 630 nm was measured. The inhibition rate of the compound on the enzyme activity was calculated, and the inhibition rate value and the logarithm value of the compound concentration were fitted using non-linear regression (dose response-variable slope) to give the IC50 value of the test compound.
In the present application, the test compounds were tested using the above assay method, and the test compounds were scored according to the IC50 value: (A) less than 100 nM; (B) between 100 nM and 1 μM; (C) greater than 1 μM, and (NT) IC50 value was not tested, as shown in Table 1.
MAT2A catalyzes the reaction of L-methionine and ATP to generate SAM, inorganic phosphonic acid, and inorganic diphosphonic acid. SAM is an important methyl donor, and is a substrate of arginine methyltransferase 5 (PRMT5). PRMT5 catalyzes SDMA modification of multiple proteins within a cell. The intracellular SDMA modification level was detected by ICW (In-cell Western), and the inhibition of the test compound on the enzymatic activity of intracellular MAT2A was indirectly reflected.
In the present application, the human colon cancer cell line HCT116 and the corresponding HCT116-MTAP-knockout cell line were seeded into a 96-well cell culture plate at an appropriate cell density. After 24 h, the cells were treated using the test compounds diluted 1:4 fold at the highest concentration of 6 μM for 6 gradients, and a DMSO treatment group was set up separately. The cells were cultured in a 37° C./5% CO2 incubator for 4 days. To test the inhibitory activity of the test compounds against tumor cell SDMA, the medium was discarded, and the plate was washed 3 times with PBST. 4% paraformaldehyde was then used at room temperature for fixation for 20 min. The fixation solution was discarded, the plate was washed 3 times with PBST, and a special blocking solution was added to each well for blocking at room temperature for 2 h. The blocking solution was discarded. After the plate was washed 3 times with PBST, the plate was incubated with SDMA antibody at 4° C. overnight. The next day, the plate was washed 3 times with PBST. The secondary antibody and DRAQ5 were diluted to an appropriate concentration with the blocking solution, and incubated at room temperature for 2 h; the plate was washed 3 times with PBST, and the fluorescence signals at 700 nm and 800 nm were detected using a Li—COR Odyssey two-color near-infrared laser imager.
The data was then processed. After the corresponding background value was subtracted from each well, statistical analysis was performed on the 800 nm/700 nm ratio for each well, a sigmoidal dose-inhibition curve was plotted using a non-linear regression model, and IC50 values were fitted and calculated. Inhibition Rate (%)=(1−SDMA dose/SDMA DMSO control)*100. SDMA dose is the value of the drug treatment group, and SDMA DMSO control is the value of the DMSO control group.
The exemplary compounds disclosed herein were tested in the above assay and determined to inhibit SDMA with IC50 based on the following scores: (A) less than 200 nM, (B) between 200 nM and 1 μM, (C) between 1 μM and 3 μM, (D) greater than 3 μM, and (NT) not tested, as shown in Table 1 below.
As can be seen from Table 1, most of the compounds had good inhibitory activity against MAT2A protein, good SDMA inhibitory activity against MATP-knockout HCT116 cells, and a certain selectivity against MATP wild-type HCT116 cells.
After the HCT116-MTAP-knockout (−/−) cell strain, the HCT116-MTAP wild-type(+/+)-cell strain, the DOHH-2 cell strain, or the Hut-78 cell strain was treated with the test compound for 5 days, the ATP level was measured using a CellTiter-Glo kit of Promega to evaluate the inhibition of the test compound against the growth of the tumor cell strain.
In the present application, the HCT116-MTAP-knockout (−/−) cell strain, the HCT116-MTAP wild-type (+/+)-cell strain, the DOHH-2 cell strain, or the Hut-78 cell strain was seeded into a 96-well cell culture plate at an appropriate cell density. After 24 h, the cells were treated using the test compounds diluted 1:3 fold at the highest concentration of 10 μM for 10 gradients, and a DMSO treatment group was set up separately. The cells were cultured in a 37° C./5% CO2 incubator for 5 days. To test the proliferation inhibition of the test compound on tumor cells, the cells were equilibrated at room temperature for 10 m e, followed by addition of 30 μL of cell proliferation assay reagent CellTiter-Glo (CTG) per well. The plate was shaken for 2 mi, and incubated in the dark for 15 min Chemiluminescence values were read using a Thermo Varioskan LUX-3020 multifunctional microplate reader for conversion to proliferation indices to calculate the inhibition rate of the compound on tumor cell proliferation, and the inhibition rate value and the logarithm value of the compound concentration were fitted using non-linear regression (dose response-variable slope) to give the IC50 value of the compound.
The exemplary compounds disclosed herein were tested in the above assay and determined to inhibit cell proliferation with IC50 based on the following scores: (A) less than 500 nM, (B) between 500 nM and 2 μM, (C) greater than or equal to 2 μM, and (NT) not tested, as shown in Table 2.
As can be seen from Table 2, most of the compounds had good proliferation inhibitory activity against MATP-knockout HCT116 cells and a certain selectivity against MATP wild-type HCT116 cells. They also had good anti-proliferative activity on MATP-deficient tumor cells DOHH2 and Hut-78.
Phase I metabolic stability of the test compounds in liver microsomes of CD-1 mice, Sprague-Dawley rats, beagle dogs, cynomolgus monkeys, and humans was assessed.
The animal and human liver microsomes used in the test system were purchased from Xenotech, Corning, or other qualified suppliers and stored in a freezer at a temperature below −60° C. prior to use.
The test samples and control compounds were separately incubated with animal and human liver microsomes at 37±1° C. for a period of incubation time up to 60 min. The samples were removed at the indicated time points, and the reaction was terminated with acetonitrile or other organic solvents containing an internal standard. After centrifugation, the resulting supernatant was assayed by liquid chromatography-tandem mass spectrometry (LC-MS/MS).
73.21 g of dipotassium hydrogenphosphate trihydrate and 10.78 g of potassium dihydrogenphosphate were dissolved in 4000 mL of ultrapure water. The pH value of the solution was adjusted to 7.40±0.10 using 10% phosphoric acid or 1 M potassium hydroxide to a final concentration of 100 mM.
The test sample powder was prepared into a stock solution with a certain concentration using DMSO or other organic solvents, which was then further diluted using a proper organic solvent. Control compounds testosterone, diclofenac, and propafenone were prepared into a 10 mM stock solution using DMSO, which was then further diluted using a proper organic solvent.
Microsomes of each species were diluted to a 2× working solution using a 100 mM potassium phosphate buffer. The final concentration of the microsomes in the reaction system was 0.5 mg/mL.
An appropriate amount of nicotinamide adenine dinucleotide phosphate (NADP) and isocitric acid (ISO) powder was weighed out, dissolved in a magnesium chloride solution, and shaken and mixed well. An appropriate amount of isocitrate dehydrogenase (IDH) was added, and the mixture was mixed well by gently turning upside down. The final concentrations in the reaction system were: 1 mM NADP, 1 mM magnesium chloride, 6 mM ISO, and 1 unit/mL IDH.
The stop solution was prepared using acetonitrile or other organic solvent containing an internal standard (tolbutamide or other suitable compounds). The prepared stop solution was stored in a freezer at a temperature of 2-8° C.
Incubation was performed in a 96-well plate. 8 incubation plates were prepared and designated T0, T5, T15, T30, T45, T60, Blank60, and NCF60, respectively. The first 6 plates corresponded to reaction time points of 0, 5, 15, 30, 45, and 60 min, respectively. No test sample or control compound was added to the Blank60 plate, and samples were taken after 60 min of incubation. In the NCF60 plate, incubation was performed for 60 min using the potassium phosphate buffer instead of the NADPH regeneration system solution. Three replicates were made for samples in all conditions.
The microsomes and the test sample or control compound were mixed, and then the incubation plates Blank60, T5, T15, T30, T45, and T60 except for TO and NCF60 were placed in a 37° C. water bath for pre-incubation for about 10 min. In the incubation plate TO, a stop solution was first added, followed by addition of the NADPH regeneration system working solution, and 98 μL of the potassium phosphate buffer was added to each sample well of the incubation plate NCF60 to initiate the reaction. After the pre-incubations of the incubation plates Blank60, T5, T15, T30, T45, and T60 were completed, 98 μL of the NADPH regeneration system working solution was added to each sample well to initiate the reaction. The reaction temperature was 37±1° C., the final volume of the reaction was 200 μL, and the reaction system included 0.5 mg/mL microsomes, 1.0 μM substrate, 1 mM NADP, 6 mM ISO, and 1 unit/mL IDH.
At 5, 15, 30, 45, and 60 min, a cold stop solution containing an internal standard was separately added to the reaction plate to stop the reaction.
All the reaction plates after termination were shaken up and centrifuged for 20 min at 3220×g at 4° C. After the supernatant was diluted according to a certain proportion, LC-MS/MS analysis was performed.
Sample analysis was carried out by the liquid chromatography-tandem mass spectrometry (LC-MS/MS) method, and the standard curve and quality control sample were not included. Semi-quantitative determinations were made using the ratio of the analyte peak area to the internal standard peak area. The retention times of the analyte and internal standard, the chromatogram acquisitions and the integrals of the chromatograms were processed with software Analyst (Sciex, Framingham, Massachusetts, USA).
The CV for the internal standard peak area in each matrix in each analysis batch should be within 20%.
The in vitro elimination rate constant ke of the compound was obtained by converting the ratio of the peak area of the compound to the internal standard peak area in the following formula into the remaining rate:
CL
int (mic)=0.693/T1/2/microsome protein content (microsome concentration mg/mL during incubation)
CL
int (liver)
=CL
int (mic)×microsome protein amount in liver (mg/g)×liver-to-body weight ratio
According to the well stirred model, the hepatic intrinsic clearance and hepatic clearance can be converted through the following formula.
Parameters in the formula are shown in Table 3.
The liver microsome stability data for some of the compounds are shown in Table 4
This experimental example was used to test the metabolic stability of the compounds in hepatocytes.
A suspension of hepatocytes was prepared at 0.5×106/mL with a pre-heated medium, then 198 μL of the pre-heated cell suspension was added to a 96-well plate. To each well of the 96-well plate, 2 μL of the test compound was added so that the final concentration was 1 μM, and 2 replicate wells were set. For the T=0 min sample, the compound was mixed well with the cells for 1 min, then L of the sample was immediately added to 125 μL of a stop solution (an acetonitrile solution containing 200 ng/mL tolbutamide and 200 ng/mL labetalol) under an ice bath and mixed well. At the same time, all the plates were placed in an incubator at 37° C. with 5% CO2, with the shaker set at 600 rpm. The sample was mixed well at 15, 30, 60, and 90 min of incubation, respectively, and 25 μL of the sample was added to 125 μL of a stop solution (an acetonitrile solution containing 200 ng/mL tolbutamide and 200 ng/mL labetalol) under an ice bath, mixed well, and shaken at 500 rpm for 10 min. Subsequently, the plate was centrifuged for 20 min at 3220×g at 4° C. After the centrifugation was completed, 80 μL of the supernatant from each well was transferred to another 96-well plate containing 240 μL of ultrapure water. Subsequently, analysis was carried out using LC-MS/MS, and intrinsic clearance (CLint) and half-life (T½) were calculated.
The metabolic stability data in hepatocytes for some of the compounds are shown in Table 5:
In this example, the pharmacokinetic behavior of the compound after intravenous injection (IV) and intragastric (PO) administration in SD rats was tested.
On the day of administration, rats were weighed for actual body weight and the administration volume was calculated. Each compound was tested in two groups with 3 rats in each group, one group administered by single intravenous injection and the other by single intragastric administration. Whole blood samples were collected via the jugular vein at prescribed time points (0.25, 0.5, 1, 2, 4, 8, and 24 h after administration). After blood sample collection, the samples were transferred to labeled commercial sample tubes containing K2-EDTA (0.85-1.15 mg), followed by centrifugation (3200×g, 4° C., 10 min) and plasma collection. The plasma was transferred to a pre-cooled centrifuge tube, frozen in dry ice, and stored in an ultra-low temperature freezer at −60° C. or lower until the LC-MS/MS analysis.
Plasma concentrations were determined using LC-MS/MS method. Plasma drug concentration data for the compounds were processed in a non-compartmental model using WinNonlin Version 6.3 (Pharsight, Mountain View, CA) pharmacokinetic software. The relevant pharmacokinetic parameters were calculated using linear logarithmic trapezoid method.
The pharmacokinetic results in rats for some of the compounds are shown in Table 6:
As can be seen from the results in Table 6, the compounds of the present invention had relatively good oral exposure and oral bioavailability in rats.
In this example, the pharmacokinetic behavior of the compound after intravenous injection (IV) and intragastric (PO) administration in BALB/c mice was tested.
On the day of administration, mice were weighed for actual body weight and the administration volume was calculated. Each compound was tested in two groups with 9 mice in each group, one group administered by single intravenous injection and the other group of mice by single intragastric administration. Whole blood samples were collected via the orbit at prescribed time points (0.25, 0.5, 1, 2, 4, 8, and 24 h after administration). After blood sample collection, the samples were transferred to labeled commercial sample tubes containing K2-EDTA (0.85-1.15 mg), followed by centrifugation (3200×g, 4° C., 10 min) and plasma collection. The plasma was transferred to a pre-cooled centrifuge tube, frozen in dry ice, and stored in an ultra-low temperature freezer at −60° C. or lower until the LC-MS/MS analysis.
Plasma concentrations were determined using LC-MS/MS method. Plasma drug concentration data for the compounds were processed in a non-compartmental model using WinNonlin Version 6.3 (Pharsight, Mountain View, CA) pharmacokinetic software. The relevant pharmacokinetic parameters were calculated using linear logarithmic trapezoid method.
The pharmacokinetic results in mice for some of the compounds are shown in Table 7:
As can be seen from the results in Table 7, the compounds of the present invention had relatively good oral exposure and oral bioavailability in mice, wherein B60, B 181, B245, B254, B256, B257, and B258 had a relatively high AUC and a relatively long half-life (T½); B258 had a relatively high Vz value and good tissue distribution.
The BEK293 cells were cultured in a DiVEM medium containing 1000 fetal bovine serum and 0.8 mg/mL G418 at 37° C. with 5% o CO2. The cells were digested by TrypLE™ Express and centrifuged. The cell density was adjusted to 2×106 cells/mL. The cells were gently mixed for 15-20 min using a shaker equilibrated at room temperature, and subjected to patch clamping on a machine. The medium of the prepared cells was replaced with extracellular fluid. The intracellular fluid and the extracellular fluid were taken from the fluid pool, and respectively added to the intracellular fluid pool and the cell and the test substance pool of the QPlate chip. The voltage stimulation of the whole-cell hERG potassium current was recorded by the whole-cell patch clamp, and the test data was collected and stored by Qpatch. The compound was diluted 3-fold at an initial concentration of 30 μM, and 6 concentration points were set, with each drug concentration being set for two administrations over a period of at least 5 min. The current detected in compound-free extracellular fluid for each cell was served as its own control group, and the detection was repeated at least twice independently using two cells per concentration. All electrophysiological experiments were performed at room temperature.
For data analysis, the current acted for each drug concentration and the current of the blank control were standardized
and then the inhibition rate corresponding to each drug concentration was calculated
The mean and standard error were calculated for each concentration, and the half maximal inhibitory concentration of each compound was calculated:
The dose-dependent effect was fitted non-linearly using the above equation, wherein Y represents the inhibition rate, C represents the concentration of the test substance, IC50 is the half maximal inhibitory concentration, and HillSlope represents the Hill coefficient. Curve fitting and calculation of IC50 were performed by Graphpad software.
The hERG inhibitory activity data for some of the compounds are shown in Table 8:
As can be seen from the results in Table 8, the compounds of the present invention had no hERG inhibitory activity.
100 mM K-Buffer: 9.5 mL of stock solution A was mixed with 40.5 mL of stock solution B, the total volume was adjusted to 500 mL with ultrapure water, and the buffer was titrated to pH 7.4 with KOH or H3PO4.
The test substance powder was prepared into a stock solution with a certain concentration using DMSO or other organic solvents, which was then further diluted using a proper organic solvent.
The extracorporeal incubation system for CYP450 enzyme metabolism phenotype study was biochemical reaction of a prepared liver microsome, a redox coenzyme, and an enzyme-specific selective inhibitor in simulated physiological temperature and environment.
The concentration of the parent drug or a metabolite thereof in the incubation solution was determined by LC-MS/MS.
The CYP inhibitory activity data for some of the compounds are shown in Table 9:
As can be seen from the results in Table 9, the compounds of the present invention had no CYP liver enzyme inhibitory activity.
In this example, the compound was tested for PXR activation.
100 μL of stably transfected DPX2 cells were seeded per well of a 96-well cell culture plate (4.5×105 cells/mL in Puracyp medium), followed by placing the 96-well plate in a 37° C. incubator. After 24 h, the 96-well plate was taken out from the incubator, the medium was replaced with 100 μL of the test compound (at a final concentration of 10 μM and 30 μM), positive control compound rifampicin (at a final concentration of 10 μM), and the DMSO treatment group (at a final concentration of 0.1%), and 3 replicate wells were set. The test plate was placed back into the incubator. After 24 h, the medium was replaced with the freshly prepared test compound and rifampicin solution, and the test plate was again placed back into the incubator. After the compound was treated for 48 h, the test plate was taken out from the incubator. The medium in the wells was discarded, the plate was washed twice with PBS, 50 μL of medium containing 1×CellTiter-Fluor™ Cell Viability Assay reagent was added, and the mixture was incubated at 37° C. for 30 min. The test plate was taken out and equilibrated to room temperature, and the fluorescence signal under the conditions of 400 nm excitation light/505 nm emission light was detected by a microplate reader.
Subsequently, 50 μL of medium containing ONE-Glo Assay reagent was added to each well, and the plate was shaken, mixed well, and incubated at room temperature for 5 min. Chemiluminescence was detected by a microplate reader. The data was analyzed to evaluate the PXR activation of the test compound.
The PXR activation data for some of the compounds are shown in Table 10:
As can be seen from the results in Table 10, the compound o the present invention ad relatively weak activation activity on PXR at 10 μM and medium activation activity on PXR at 30 μM. The activation activity of B181 and B257 on PXR at 30 μM was also very weak, much lower than the other compounds.
In this example, the in vivo efficacy of the compound in a mouse xenograft tumor model after intragastric (PO) administration was tested.
Human colorectal adenocarcinoma cells HCT116-MTAP-KO were cultured in monolayer in vitro under the condition of McCoy's 5A medium containing 10% fetal bovine serum and 1% penicillin and streptomycin, and the culture was carried out at 37° C. with 5% CO2. The cells were digested with trypsin three times a week for passaging. When the saturation degree of the cells was 80-90%, the cells were collected, counted and inoculated. 0.1 mL (5×106 cells) of HCT116-MTAP-KO cells were subcutaneously inoculated on the right back of each mouse. On day 16 after inoculation of the cells, the mean tumor volume reached 154 mm3. The mice were randomly grouped for administration and intragastrically dosed once daily. Changes in body weight and tumor volume were recorded. After a certain number of days of administration, the experiment was ended. Tumor volume changes and mouse body weight changes were counted and analyzed. AG270 (CAS: 2201056-66-6) was the positive control compound.
Human pancreatic cancer cells KP-4 were cultured in monolayer in vitro under the condition of 1640 medium containing 100. fetal bovine serum and 10 penicillin and streptomycin, and the culture was carried out at 37° C. with 5% CO2. The cells were digested with trypsin three times a week for passaging. When the saturation degree of the cells was 80-90%, the cells were collected, counted and inoculated. 0.1 mL (5×106 cells) of KP-4 cells were subcutaneously inoculated on the right back of each mouse. On day 34 after inoculation of the cells, the mean tumor volume reached 154 mm3. The mice were randomly grouped for administration and intragastrically dosed once daily. Changes in body weight and tumor volume were recorded. After 29 days of administration, the experiment was ended. Tumor volume changes and mouse body weight changes were counted and analyzed. AG270 (CAS: 2201056-66-6) was the positive control compound.
As can be seen from the results in Table 11 and Table 12, the compound of the present invention had relatively good in vivo efficacy.
About 0.21 g of sodium hydroxide, about 2.24 g of sodium dihydrogen phosphate dihydrate, and 3.09 g of sodium chloride were weighed out and dissolved in 500 mL of water, and then the pH was adjusted to 6.5 with 1 N sodium hydroxide or 1 N hydrochloric acid. 112 mg of FaSSIF solid was weighed out and placed in a 50 mL volumetric flask, and the above buffer was added for dissolution, diluted to volume, mixed well, and left to stand at room temperature for 2 h or more.
The peak of the main component of the control substance and the peak of the test substance consistent with the retention time of the peak of the main component of the control substance were integrated, and the concentration thereof in the FaSSIF solution was calculated according to external standard method. The test results are shown in Table 14.
As can be seen from the results in Table 13, the compound of the present application had relatively good solubility.
It should be understood that the above examples are exemplary and are not intended to encompass all possible implementations encompassed by the claims. Various modifications and changes may also be made on the above examples without departing from the scope of the present disclosure. Likewise, various technical features of the above examples may also be combined in any combination to form additional examples of the present application that may not be explicitly described. Therefore, the above examples only represent several embodiments of the present application, and do not limit the scope of the present application.
Number | Date | Country | Kind |
---|---|---|---|
202210097016.3 | Jan 2022 | CN | national |
202210965087.0 | Aug 2022 | CN | national |
202211486374.X | Nov 2022 | CN | national |
202310030336.1 | Jan 2023 | CN | national |
The present disclosure is a Continuation application of International Application No. PCT/CN2023/073092, filed on Jan. 19, 2023, which claims priority to the Chinese Patent Application No. 202210097016.3 filed on Jan. 26, 2022, the Chinese Patent Application No. 202210965087.0 filed on Aug. 12, 2022, the Chinese Patent Application No. 202211486374.X filed on Nov. 24, 2022, and the Chinese Patent Application No. 202310030336.1 filed on Jan. 9, 2023, the entire contents of which are incorporated herein by reference.
Number | Date | Country | |
---|---|---|---|
Parent | PCT/CN2023/073092 | Jan 2023 | WO |
Child | 18784600 | US |