The present disclosure relates to a compound with Menin-MLL protein-protein interaction inhibitory activity and cell proliferation inhibitory activity, and use thereof in treating cancer and other diseases mediated by Menin-MLL interaction.
Mixed-lineage leukemia (MLL) protein is a histone methyltransferase that may mutate clinically and in biologically distinct subtypes of acute leukemia. The MLL gene family includes five members, MLL1-5, and is closely associated with the development, progression, and metastasis of various tumors. The multiple endocrine oncoprotein (Menin protein) is encoded by the multiple endocrine neoplasia type 1 (MEN1) gene, which acts as a tumor suppressor gene in endocrine organs. Menin protein interacts with many other proteins, forming a complex interaction network. Studies have demonstrated that the direct interaction of Menin with MLL1 and MLL2 proteins is indispensable for the enzymatic activity of the complex histone methylation modification (H3K4), the transcriptional regulation of target genes, and their corresponding functions. Menin interacts with the amide terminus of MLL1 and acts as an oncogenic cofactor that increases the transcription of gene clusters such as HOX and MEIS1. The interaction between Menin and MLL fusion proteins is indispensible for the abnormal activation of a series of gene clusters and the onset of leukemia caused by MLL fusion proteins. Furthermore, Menin, as a nuclear protein widely expressed in tissues, participates in the formation of a multitude of important transcriptional regulatory complexes and exhibits a variety of important biological functions in the body. In addition to its role in the formation of MLL1 and MLL2 epigenetic regulatory complexes, the Menin protein has been reported to interact with a variety of transcription factors, including JunD, NFKB, and SMAD3, to regulate the activation or inhibition on the transcription of target genes.
The use of small molecules to target the Menin-MLL interaction represents an attractive strategy for the development of new therapies for MLL leukemia. Moreover, inhibiting the interaction of Menin with wild-type MLL1 and MLL2 may have therapeutic potential for numerous solid cancers, including liver, brain, colon, and breast cancers.
Therefore, inhibitors of Menin-MLL1 protein-protein interaction can be considered as prospective therapeutic compounds with a broad range of applications in the treatment of tumors. The selective targeting of this interaction interface is conducive to the development of new drugs related thereto.
The present disclosure provides a compound represented by Formula I, or a deuterated compound, a stereoisomer or a pharmaceutically acceptable salt thereof:
In some embodiments of the present disclosure, when R1, R2, R3, R4, R5, R1′, R2′, R3′ and R4′ are all hydrogen, X is O, and Y1 is CH, m is not 1.
In some embodiments of the present disclosure, preferably, n1 and n2 are 1, n3 and n4 are 2; alternatively, n1 and n2 are 2, n3 and n4 are 1. Preferably, Y1 is N, and Y2 is N; alternatively, Y1 is CH, and Y2 is N. Preferably, m is 1 or 2; more preferably, m is 1.
In some embodiments of the present disclosure, preferably, W is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, methyl, ethyl, n-propyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, hydroxy, methoxy, ethoxy, methoxymethyl, amino, methylamino and dimethylamino.
In some embodiments of the present disclosure, preferably, X is selected from the group consisting of CRaRb, NRa and O; more preferably, X is CRaRb or NRa.
In some embodiments of the present disclosure, when X is CRaRb, preferably, Ra and Rb are independently selected from the group consisting of hydrogen, fluorine, chlorine, cyano, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, —C0 alkylene-ORA1, —C1 alkylene-ORA1, —C2 alkylene-ORA1 and —C3 alkylene-ORA1; RA1 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and isopropyl.
In some embodiments of the present disclosure, when X is CRaRb, preferably, Ra is hydrogen, Rb is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, —C0 alkylene-ORA1, —C1 alkylene-ORA1, —C2 alkylene-ORA1 and —C3 alkylene-ORA1; RA1 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and isopropyl.
In some embodiments of the present disclosure, when X is CRaRb, preferably, Ra and Rb are the same and are selected from the group consisting of hydrogen, fluorine, chlorine, cyano, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, —C0 alkylene-ORA1, —C1 alkylene-ORA1, —C2 alkylene-ORAL and —C3 alkylene-ORA1; RA1 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and isopropyl.
In some embodiments of the present disclosure, when X is CRaRb, preferably, Ra, Rb and the atom connecting therewith together form
3-membered carbocyclyl, 4-membered carbocyclyl, 5-membered carbocyclyl, 6-membered carbocyclyl, 4-membered heterocycloalkyl, 5-membered heterocycloalkyl or 6-membered heterocycloalkyl; each RB1 is independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and isopropyl.
In some embodiments of the present disclosure, when X is NRa, preferably, Ra is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, —C1 alkylene-ORA1, —C2 alkylene-ORA1 and —C3 alkylene-ORA1; RA1 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and isopropyl.
In some embodiments of the present disclosure, preferably, R1, R2, R3, R4, R5, R1′, R2′, R3′ and R4′ are independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl and trifluoromethyl.
In some embodiments of the present disclosure, preferably, R1, R2′, R3′ and R4+ are independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl and trifluoromethyl; two non-adjacent groups of R1, R2, R3, R4 and R5 are interconnected, and together with the ring where the atom connecting therewith is located, form 7-membered bridged cycloalkyl, 8-membered bridged cycloalkyl or 9-membered bridged cycloalkyl; wherein two non-adjacent groups of R1, R2, R3, R4 and R5 are interconnected into a connecting chain selected from the group consisting of —CH2—, —CH2CH2— and —CH2CH2CH2—. More preferably, R2 and R4 are interconnected, or R2 and R3 are interconnected, or R1 and R4 are interconnected.
In some embodiments of the present disclosure, preferably, R6 is —C(O)NRE1RE2; RE1 and RE2 are independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, —C4 alkyl, —C5 alkyl, —C6 alkyl, 3-membered carbocyclyl, 4-membered carbocyclyl, 5-membered carbocyclyl, 6-membered carbocyclyl, —C1 alkylene-(3-membered carbocyclyl), —C1 alkylene-(4-membered carbocyclyl), —C1 alkylene-(5-membered carbocyclyl), and —C1 alkylene-(6-membered carbocyclyl).
In some embodiments of the present disclosure, preferably, R6 is selected from the group consisting of 5-membered heteroaromatic ring, 6-membered heteroaromatic ring, —C1 alkylene-(5-membered heteroaromatic ring) and —C1 alkylene-(6-membered heteroaromatic ring); wherein the heteroaromatic group is further optionally substituted by one, two, three or four independent RE5; each RE5 is independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, 3-membered carbocyclyl, 4-membered carbocyclyl, 5-membered carbocyclyl and 6-membered carbocyclyl.
Further,
Further,
Further,
In some embodiments of the present disclosure, Ra is hydrogen and Rb is selected from the group consisting of hydrogen, fluorine, methyl,
ethyl, isopropyl,
In some embodiments of the present disclosure, Ra and Rb are the same and are selected from the group consisting of hydrogen, fluorine, methyl,
ethyl, isopropyl,
In some embodiments of the present disclosure, further,
In some embodiments of the present disclosure, further,
In some embodiments of the present disclosure, further,
In some embodiments of the present disclosure, further,
Further specifically,
In some embodiments of the present disclosure, further,
Further specifically,
In some embodiments of the present disclosure, further,
Preferably, R6 is selected from the group consisting of —C(O)NRE1RE2, —NRE1C(O)RE2, —NRE1S(O)2RE2, —NRE1S(O)RE2, 6-membered aromatic ring, -5-membered heteroaromatic ring, -6-membered heteroaromatic ring, 3-membered carbocyclyl, 4-membered carbocyclyl, 5-membered carbocyclyl, 6-membered carbocyclyl, -4-membered heterocycloalkyl, -5-membered heterocycloalkyl, -6-membered heterocycloalkyl, —S(O)RE1, —S(O)2RE1, —S(O)2NRE1RE2, —S(O)(NH)RE1, —S(O)(NH)NRE1RE2, —ORE1, —OC(O)RE1 and —SRE1; wherein the carbocyclyl, heterocycloalkyl, aryl or heteroaryl is further optionally substituted by one, two, three or four independent RE5.
Preferably, RE1 and RE2 are independently selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, monodeuteriomethyl, dideuteriomethyl, trideuteriomethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, —C4 alkyl, —C5 alkyl, —C6 alkyl, 3-membered carbocyclyl, 4-membered carbocyclyl, 5-membered carbocyclyl, 6-membered carbocyclyl, —C1 alkylene-(3-membered carbocyclyl), —C1 alkylene-(4-membered carbocyclyl), —C1 alkylene-(5-membered carbocyclyl) and —C1 alkylene-(6-membered carbocyclyl).
Further specifically, R6 is selected from the group consisting of
In some embodiments of the present disclosure, the general formula shown in Formula I may be as follows:
In some embodiments of the present disclosure, the general formula shown in Formula I may be as follows:
In some embodiments of the present disclosure, further, the general formula shown in Formula I may be as follows:
In some embodiments of the present disclosure, the compound is represented by Formula II:
In some embodiments of the present disclosure, the compound is represented by Formula IIa or Formula IIb:
In some embodiments of the present disclosure, the compound is represented by Formula IIIa or Formula IIIb:
In some embodiments of the present disclosure, the compound is represented by Formula IVa or Formula IVb:
In some embodiments of the present disclosure, the compound is represented by formula Va:
In some specific embodiments of the present disclosure, the compound is specifically selected from the group consisting of:
In another specific embodiments of the present disclosure, the compound is specifically selected from the group consisting of:
The present disclosure further provides use of the above-mentioned compound, or the deuterated compound, stereoisomer thereof or pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a disease associated with abnormal Menin activity.
The present disclosure further provides use of the above-mentioned compound, or the deuterated compound, stereoisomer thereof or pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer.
The present disclosure further provides a pharmaceutical composition, comprising a preparation prepared from any of the above-mentioned compounds, or the deuterated compound, stereoisomer thereof or pharmaceutically acceptable salt thereof.
The pharmaceutical composition further comprises a pharmaceutically acceptable carrier, excipient or vehicle.
The compound of the present disclosure has excellent inhibitory activity against Menin-MLL protein-protein interaction and cell proliferation, and has excellent performance in terms of safety, bioavailability and animal efficacy.
The diseases related to Menin-MLL interaction or Menin-MLL fusion protein interaction as defined in the present disclosure include one or more of cancer or malignant tumors, diabetes and other Menin-related diseases. “Cancer” or “malignant tumor” are used as synonymous terms and refer to any of a number of diseases that are characterized by uncontrolled, abnormal proliferation of cells, the ability of affected cells to spread locally or through the bloodstream and lymphatic system to other parts of the body (i.e., metastasize) as well as any of a number of characteristic structural and/or molecular features. “Cancer cell” refers to a cell that undergoes an early, intermediate or advanced stage of multi-step tumor progression. The “cancer” or “malignant tumor” refers to leukemia, lymphoma, sarcoma, lung cancer, esophageal cancer, gastric cancer, liver cancer, brain tumor, myeloma, pancreatic cancer, breast cancer, colon cancer, prostate cancer, bladder cancer, multiple myeloma, brain tumor, or multiple endocrine cancer.
In particular embodiments, the compound of the present disclosure is used to treat leukemia associated with MLL rearrangement, acute lymphocytic leukemia associated with MLL rearrangement, acute lymphoblastic leukemia associated with MLL rearrangement, acute lymphoid leukemia associated with MLL rearrangement, acute myeloid leukemia associated with MLL rearrangement, or acute myeloblastic leukemia associated with MLL rearrangement. As used herein, “MLL rearrangement” means rearrangement of the MLL gene. In certain embodiments, diseases or conditions treatable with the compound of the present disclosure include insulin resistance, prediabetes, diabetes (such as type II diabetes or type I diabetes), risk of diabetes, and hyperglycemia.
Combination therapies: The present disclosure also relates to combination therapies for treating the diseases or disorders described herein. In certain embodiments, combination therapies include administering at least one compound of the present disclosure in combination with one or more other pharmaceutically active agents for treating cancer or other diseases mediated by Menin/MLL. The pharmaceutically active agent can be combined with the compound of the present disclosure in a single dosage form, or can be administered simultaneously or sequentially as separate dosage forms. The compound of the present disclosure can also be used in combination with immunotherapies, including but not limited to cell-based therapies, antibody therapies, and cytokine therapies, for treating the diseases or disorders disclosed herein.
The compounds and derivatives provided in the present disclosure can be named according to the IUPAC (International Union of Pure and Applied Chemistry) or CAS (Chemical Abstracts Service, Columbus, OH) nomenclature system.
Definitions of terms used in the present disclosure: Unless otherwise stated, the initial definitions provided for groups or terms herein apply to the groups or terms throughout the specification; for terms that are not specifically defined herein, the meaning that a person skilled in the art can give them should be given based on the disclosure and context.
“Substitution” means that the hydrogen atom in the molecule is replaced by other different atoms or groups; or the lone pair of electrons of an atom in the molecule is replaced by other atoms or groups, for example, the lone pair of electrons on the S atom can be replaced by an O atom to form
“Further optionally substituted”, “further substituted” or “optionally substituted” means that “substitution” may but need not occur, and the expression includes situations where substitution occurs or does not occur.
The minimum and maximum carbon atom number of the hydrocarbon group is indicated by a prefix. For example, the prefix Ca-b alkyl indicates any alkyl group containing “a” to “b” carbon atoms. Thus, for example, C1-6 alkyl refers to an alkyl group containing 1 to 6 carbon atoms.
“Alkyl” refers to a saturated hydrocarbon chain with a specified number of member atoms. Alkyl groups can be linear or branched. Representative branched alkyl groups have one, two or three branches. Alkyl groups may be optionally substituted with one or more substituents as defined herein. Alkyl groups include methyl, ethyl, propyl (n-propyl and isopropyl), butyl (n-butyl, isobutyl and tert-butyl), pentyl (n-pentyl, isopentyl and neopentyl) and hexyl. Alkyl groups may also be part of other groups, such as —O(C1-6 alkyl).
“Alkylene” refers to a divalent saturated aliphatic hydrocarbon radical having a specified number of member atoms. Cab alkylene refers to an alkylene group having a to b carbon atoms. Alkylene groups include branched and linear hydrocarbon groups. For example, the term “propylene” can be exemplified by the following structure:
Similarly, the term “dimethylbutylene” can be exemplified, for example, by any of the following structures:
The —C0-4 alkylene of the present disclosure can be C0 alkylene, C1 alkylene (such as —CH2—), C2 alkylene (such as —CH2CH2—), C3 alkylene or C4 alkylene; C0 alkylene means that the group here does not exist and is connected in the form of a chemical bond, and A-C0 alkylene-B means A-B, that is, the A group and the B group are directly connected by a chemical bond. For example, —C0 alkylene-(3-membered cycloalkyl) refers to cyclopropyl.
“Unsaturation” described herein refers to the presence of carbon-carbon double bond, carbon-carbon triple bond, carbon-oxygen double bond, carbon-sulfur double bond, carbon-nitrogen triple bond, etc. in the groups or molecules.
“Alkenyl” refers to a linear or branched hydrocarbon group having at least one site of vinyl unsaturation (>C═C<). For example, Ca-b alkenyl refers to an alkenyl group having a to b carbon atoms and is intended to include, for example, ethenyl, propenyl, isopropenyl, 1,3-butadienyl, and the like.
“Alkynyl” refers to a linear monovalent hydrocarbon group or a branched monovalent hydrocarbon group containing at least one triple bond. The term “alkynyl” is also intended to include those hydrocarbon groups having one triple bond and one double bond. For example, C2-6 alkynyl is intended to include ethynyl, propynyl, and the like.
“Carbocyclyl” described herein refers to a saturated or non-aromatic partially saturated cyclic group with a single ring or multiple rings (fused, bridged, or spiro) having multiple carbon atoms and no ring heteroatoms. The term “carbocyclyl” includes cycloalkenyl groups, such as cyclohexenyl. Examples of monocarbocyclyl groups include, for example, cyclopropyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclooctyl, cyclopentenyl and cyclohexenyl. Examples of carbocyclyl groups of fused carbocyclyl systems include dicyclohexyl, dicyclopentyl, dicyclooctyl, etc., and two such dicycloalkyl polycyclic structures are exemplified and named below:
dicyclohexyl and
dicyclohexyl. Examples of carbocyclyl groups of bridged carbocyclyl systems include
adamantyl, etc. Examples of carbocyclyl groups of spiral carbocyclyl systems include
etc. The term “carbocyclyl” also includes the case where an aromatic ring is fused with a non-aromatic ring to form a partially saturated cyclic group, and the attachment point can be located at a non-aromatic carbon atom or an aromatic carbon atom, examples of which include 1,2,3,4-tetrahydronaphthalen-5-yl and 5,6,7,8-tetrahydronaphthalen-5-yl.
The “bridged ring” or “bridged cycloalkyl” described herein refers to a saturated or non-aromatic partially saturated cyclic group formed by bridging multiple rings having multiple carbon atoms and no ring heteroatoms. Examples of this term include, but are not limited to,
adamantyl, etc.
The “heterocycloalkyl” described herein refers to a saturated ring or a non-aromatic partially saturated ring with a single ring or multiple rings (fused, bridged, spiro) containing at least one heteroatom; wherein the heteroatom refers to a nitrogen atom, an oxygen atom, a sulfur atom, etc. It usually refers to a monovalent saturated or partially unsaturated monocyclic or polycyclic ring system with multiple ring atoms, which contains 1, 2 and 3 ring heteroatoms selected from the group consisting of N, O and S, and the remaining ring atoms are carbon. Examples of heterocycloalkyl groups of monoheterocycloalkyl systems include oxetanyl, azetidinyl, pyrrolidinyl, 2-oxo-pyrrolidin-3-yl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl, imidazolidinyl, thiazolidinyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperazinyl, morpholinyl, thiomorpholinyl, 1,1-dioxo-thiomorpholin-4-yl, azepanyl, diazepanyl, homopiperazinyl, oxaazepanyl, etc. Examples of heterocycloalkyl groups of fused heterocycloalkyl systems include 8-aza-bicyclo[3.2.1]octyl, quinuclidine, 8-oxa-3-aza-bicyclo[3.2.1]octyl, 9-aza-bicyclo[3.3.1]nonyl, etc. Examples of heterocycloalkyl groups of bridged heterocycloalkyl systems include
etc. Examples of heterocycloalkyl groups of spiral heterocycloalkyl systems include
etc. Examples of partially saturated heterocycloalkyl groups include dihydrofuranyl, imidazolinyl, tetrahydropyridyl, dihydropyranyl, etc. The term “heterocycloalkyl” also includes the case where an aromatic ring containing at least one heteroatom is fused to a non-aromatic ring to form a partially saturated cyclic group, and the attachment site can be located at a non-aromatic carbon atom, an aromatic carbon atom or a heteroatom, and examples include
The “bridged heterocycle” or “bridged heterocycloalkyl” described herein refers to a saturated ring or a non-aromatic partially saturated ring formed by bridging multiple rings containing at least one heteroatom; wherein the heteroatom refers to a nitrogen atom, an oxygen atom, a sulfur atom, etc. Examples of this term include, but are not limited to,
etc.
The “aromatic ring” described herein refers to an aromatic hydrocarbon group having multiple carbon atoms. Aryl is generally a monocyclic, bicyclic or tricyclic aromatic group having multiple carbon atoms. In addition, the term “aryl” used herein refers to an aromatic substituent that may be a single aromatic ring or multiple aromatic rings fused together. Non-limiting examples include phenyl, naphthyl or tetrahydronaphthyl.
The “aromatic heterocycle” described herein refers to an aromatic unsaturated ring containing at least one heteroatom, wherein the heteroatom refers to a nitrogen atom, an oxygen atom, a sulfur atom, etc; and generally refers to aromatic monocyclic or bicyclic hydrocarbons that contain multiple ring atoms, one or more of which are heteroatoms selected from the group consisting of O, N, and S, preferably one to three heteroatoms. Heterocyclic aryl represents, for example, pyridyl, indolyl, quinoxalinyl, quinolyl, isoquinolinyl, benzothienyl, benzofuranyl, benzothienyl, benzopyranyl, benzothiopyranyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazolyl, imidazolyl, thienyl, oxadiazolyl, benzimidazolyl, benzothiazolyl, or benzoxazolyl.
The “halogen” described herein refers to fluorine, chlorine, bromine or iodine.
The “halogen-substituted alkyl” described herein refers to an alkyl group in which one or more hydrogen atoms are replaced by halogen; for example, halogen-substituted C1-4 alkyl refers to an alkyl group containing 1 to 4 carbon atoms in which hydrogen atoms are replaced by one or more halogen atoms; examples include monofluoromethyl, difluoromethyl, and trifluoromethyl.
The “deuterium-substituted alkyl” described herein refers to an alkyl group in which one or more hydrogen atoms are replaced by deuterium; for example, deuterium-substituted C1-4 alkyl refers to an alkyl group containing 1 to 4 carbon atoms in which a hydrogen atom is replaced by one or more deuterium atoms; examples include monodeuteriomethyl, dideuteriomethyl and trideuteriomethyl.
The “—OR”, “—NRR” and the like described herein means that the R group is connected to the oxygen atom or the nitrogen atom by a single bond.
The oxygen atom in “—C(O)R”, “—S(O)2R” and the like described herein is connected to the carbon atom or the sulfur atom by a double bond, and the R group is connected to the oxygen atom or the sulfur atom by a single bond. For another example, “—S(O)(NH)R” means that the oxygen atom and the nitrogen atom are connected to the sulfur atom by a double bond, and the R group is connected to the sulfur atom by a single bond.
The “oxo” described herein refers to ═O, that is, an oxygen atom replaces two hydrogen atoms or lone pairs of electrons through a double bond.
In the description of the groups of the present disclosure, “ - - -” and
are used to describe the position of the group substitution. For example,
means that the tetrahydropyrrole ring forms a spiro ring with other rings in the structure through the position of
The “deuterated compound” described herein refers to a molecule or group in which one or more hydrogen atoms are replaced by deuterium atoms, wherein the proportion of deuterium atoms is greater than the abundance of deuterium in nature.
The “stereoisomer” described herein refers to a compound composed of the same atoms, bonded by the same chemical bonds, but with different three-dimensional structures. The stereoisomers of the present disclosure cover each single stereoisomer and its compounds, including but not limited to enantiomers and diastereomers. The compounds of the present disclosure or their pharmaceutically acceptable salts may contain one or more chiral carbon atoms, and thus may produce enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom can be defined as (R)- or (S)-based on stereochemistry. The present disclosure is intended to include all possible isomers, as well as their racemates and optically pure forms. The preparation of the compounds of the present disclosure can select racemates, diastereomers or enantiomers as raw materials or intermediates. Optically active isomers can be prepared using chiral synthons or chiral reagents, or separated using conventional techniques, such as crystallization and chiral chromatography.
When the compounds of the present disclosure contain olefinic double bonds, unless otherwise specified, the compounds of the present disclosure include cis-trans isomers of E-form and Z-form. All tautomeric forms of the compounds of the present disclosure are also included within the scope of the present disclosure.
The term “pharmaceutically acceptable” means that a carrier, vehicle, diluent, excipient, and/or formed salt is generally chemically or physically compatible with the other ingredients that constitute a pharmaceutical dosage form and physiologically compatible with the receptor.
The terms “salts” and “pharmaceutically acceptable salts” refer to acidic and/or basic salts of the above compounds or their stereoisomers, formed with inorganic and/or organic acids and bases, and also include zwitterionic salts (inner salts) as well as quaternary ammonium salts such as alkylammonium salts. These salts can be obtained directly during the final separation and purification of the compounds. They can also be obtained by mixing the above compounds, or their stereoisomers, with a certain amount of acid or base appropriately (e.g., equivalent amounts). These salts may be precipitated in the solution and collected by filtration, or recovered after evaporation of the solvent, or obtained by freeze-drying after reaction in an aqueous medium.
In certain embodiments, one or more compounds of the present disclosure may be used in combination with each other. It is also possible to select the compounds of the present disclosure and any other active agent for use in combination to prepare a drug or pharmaceutical composition for regulating cell function or treating a disease. If a group of compounds is used, these compounds may be administered to a subject simultaneously, separately or sequentially.
Obviously, according to the above contents of the present disclosure, in accordance with common technical knowledge and customary means in the art, other various forms of modification, replacement or change may be made without departing from the above basic technical ideas of the present disclosure.
The above contents of the present disclosure are further described in detail below through specific embodiments in the form of examples. However, this should not be understood as the scope of the above subject matter of the present disclosure being limited to the following examples. All technologies realized based on the above contents of the present disclosure belong to the scope of the present disclosure.
The known starting materials of the present disclosure can be synthesized by methods known in the art, or can be purchased from companies such as Energy Chemical, Chengdu Chron Chemicals, Accela ChemBio, and J&K Scientific.
The reagents described in the examples are abbreviated as follows: UHP: urea peroxide; DBU: 1,8-diazabicycloundec-7-ene; TCCA: trichloroisocyanuric acid; KOAc: potassium acetate; Na2HPO4: disodium hydrogen phosphate; TFAA: trifluoroacetic anhydride; DIPEA: N,N-diisopropylethylamine; n-BuLi: n-butyllithium; NH2OH·HCl: hydroxylamine hydrochloride; NiCl2·6H2O: nickel chloride hexahydrate; NaBH4: sodium borohydride; NaBH3CN: sodium cyanoborohydride; NFSI: N-fluorobisbenzenesulfonamide; TEA: triethylamine; PySO3: pyridine sulfur trioxide; HATU: 2-(7-azobenzotriazole)-N,N,N′,N′-tetramethyluronium hexafluorophosphate; DMF: N,N-dimethylformamide; DCM: dichloromethane; DCE: 1,2-dichloroethane; TFA: trifluoroacetic acid; MeCN: acetonitrile; EtOH: ethanol; MeOH: methanol; NMP: N-methylpyrrolidone.
Unless otherwise specified in the examples, the reaction is carried out in a nitrogen atmosphere. Unless otherwise specified in the examples, the solution refers to an aqueous solution. Unless otherwise specified in the examples, the reaction temperature is room temperature. Room temperature is the most suitable reaction temperature, which is 20° C.-30° C. Unless otherwise specified in the examples, M is mole per liter.
The structures of the compounds were determined by nuclear magnetic resonance (NMR) and mass spectrometry (MS). NMR shifts (8) are given in units of 10-6 (ppm). NMR measurements were performed using (Bruker Avance III 400 MHz and Bruker Avance NEO 600 MHz) NMR spectrometers, and the solvents used were deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDCl3) and deuterated methanol (CD3OD), and the internal standard was tetramethylsilane (TMS). LC-MS measurements were performed using Shimadzu's liquid chromatography-mass spectrometry (Shimadzu LC-MS 2020 (ESI)). HPLC measurements were performed using Shimadzu's high pressure liquid chromatograph (Shimadzu LC-20A). MPLC (medium pressure liquid chromatography) was performed using a Gilson GX-281 reverse phase preparative chromatograph. The silica gel plate used for thin layer chromatography was Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate, and the specifications of thin layer chromatography used for separating and purifying products were 0.4 mm-0.5 mm. For column chromatography, Yantai Huanghai silica gel 200-300 mesh silica gel was generally used as a carrier. The instrument used for supercritical fluid chromatography (SFC) analysis and preparation was SHIMADZU SFC-30A.
To a solution of 5-fluoro-2-methoxybenzoic acid (45 g, 264.49 mmol) and ethylisopropylamine (23.21 g, 317.39 mmol) in DCM (880 mL), HATU (120.61 g, 317.39 mmol) and DIPEA (102.55 g, 793.48 mmol, 138.21 mL) were added at 0° C. The reaction mixture was slowly warmed to room temperature and stirred for 17 h. After the reaction was completed, the reaction mixture was diluted with water, extracted three times with EA, and the organic phases were combined. The crude product obtained by concentration was separated and purified by silica gel column to obtain intermediate 1-2 (60 g, 250.75 mmol, yield 94.8%). MS m/z=240 [M+H]+.
To a solution of 1-2 (63 g, 263.28 mmol) in DCM (200.00 mL), BBr3 (131.64 g, 526.57 mmol) was added dropwise at −70° C. After the addition was completed, the reaction mixture was slowly warmed to room temperature and stirred for 17 h. After the reaction was completed, the mixture was cooled to −70° C., quenched with MeOH, and extracted with EA. The organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product 1-3 (49 g, 217.53 mmol, yield 82.6%), which was used directly in the next step without further purification. MS m/z=226 [M+H]+.
Referring to the synthetic route of intermediate 1-3, diisopropylamine was used instead of ethylisopropylamine to obtain intermediate 1-5. MS m/z=240 [M+H]+.
Referring to the synthetic route of intermediate 1-3, dicyclopropylamine was used instead of ethylisopropylamine to obtain intermediate 1-7. MS m/z=236 [M+H]+.
At room temperature, 1-8 (3 g, 52.54 mmol) was dissolved in anhydrous methanol (40 mL), then added with 4 N HCl/MeOH (46 mL), acetone (7.63 g, 131.37 mmol) and NaBH3CN (4.97 g, 78.81 mmol), and stirred at room temperature for 2 h. After the reaction was completed, the reaction mixture was concentrated directly. The concentrated crude product was dissolved in 10% NaOH aqueous solution and extracted three times with MTBE. The organic phases were combined, acidified with HCl/MeOH, and concentrated to obtain a crude product (6.5 g, 47.92 mmol, yield 91.2%), which was directly used in the next step. MS m/z=100 [M+H]+.
Referring to the synthetic route of intermediate 1-3, 1-9 was used instead of ethylisopropylamine to obtain target intermediate 1-11. MS m/z=238 [M+H]+.
At room temperature, 1-12 (3.56 g, 50.06 mmol) was dissolved in anhydrous methanol (40 mL), then added with acetone (7.27 g, 125.14 mmol) and NaBH3CN (4.73 g, 75.08 mmol), and stirred at room temperature for 2 h. After the reaction was completed, the mixture was concentrated directly. The concentrated crude product was dissolved in 10% NaOH aqueous solution and extracted three times with MTBE. The organic phases were combined, acidified with HCl/MeOH, and concentrated to obtain a crude product (6.5 g, 43.43 mmol, yield 86.7%), which was directly used in the next step. MS m/z=114 [M+H]+.
Referring to the synthetic route of intermediate 1-3, 1-13 was used instead of ethylisopropylamine to obtain target intermediate 1-15. MS m/z=252 [M+H]+.
Referring to the synthetic route of intermediate 1-3, isopropylmethylamine was used instead of ethylisopropylamine to obtain intermediate 1-17. MS m/z=212 [M+H]+.
Under ice bath, 1-8 (3.0 g, 52.54 mmol) was dissolved in DCM (50 mL), then added with (Boc)2O (12.04 g, 55.17 mmol) and triethylamine (6.37 g, 63.05 mmol), slowly warmed to room temperature and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude 1-18 was directly used in the next step without further purification. MS m/z=158 [M+H]+.
At 0° C., 1-18 (8.33 g, 52.99 mmol) was dissolved in anhydrous DMF, and NaH (1.34 g, 55.64 mmol) was added under nitrogen protection. The reaction was carried out at 0° C. for 30 min. Ethyl iodide (9.9 g, 58.29 mmol) was slowly added dropwise. After the addition was completed, the mixture was warmed to room temperature and stirred. After the reaction monitored by LCMS was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain 1-19 (5.6 g, 30.23 mmol, yield 57%). MS m/z=186 [M+H]+.
1-19 (5.6 g, 30.23 mmol) was dissolved in MTBE, slowly added with HCl/EA, stirred at room temperature until solid no longer precipitated, and subjected to suction filtration. The filter cake was washed with a small amount of EA to obtain intermediate 1-20 (2.74 g, 22.53 mmol, yield 74.52%). MS m/z=86 [M+H]+.
Referring to the synthetic route of intermediate 1-3, 1-20 was used instead of ethylisopropylamine to obtain target intermediate 1-22. MS m/z=224 [M+H]+.
1-23 (680 mg, 3.56 mmol) was dissolved in a mixed solvent of dioxane (15 mL) and water (3 mL). 1-24 (1.01 g, 4.27 mmol), Na2CO3 (754.77 mg, 7.12 mmol), and Pd(dppf)Cl2 (130.13 mg, 0.178 mmol) were added in sequence, and stirred at 90° C. overnight. After the reaction monitored by LCMS was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain 1-25 (700 mg, 3.18 mmol, yield 89%). MS m/z=221 [M+H]+.
Referring to the synthesis method of 1-25, 1-26 was used instead of 1-24 to obtain 1-27 (232 mg, 1.0 mmol, yield 80%). MS m/z=233 [M+H]+.
At 0° C., 2-1 (4.9 g, 26.57 mmol) and TEA (5.38 g, 53.14 mmol) were dissolved in DCM, and a solution of tert-butyl 2,7-diazaspiro[3.5]nonane-7-carboxylate (4.81 g, 21.26 mmol) in DCM was slowly added dropwise. The mixture was stirred at this temperature for 2 h. After the reaction was completed, the mixture was diluted with water and extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1-1:1, v/v) to obtain 2-2 (6.5 g, 17.37 mmol, yield 65.37%). MS m/z=374 [M+H]+.
At 0° C., 2-2 (1.0 g, 2.67 mmol) and DBU (488.13 mg, 3.21 mmol) were dissolved in THE, then added with 1-3 (601 mg, 2.67 mmol), warmed to room temperature and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=10:1-5:1, v/v) to obtain 2-3 (1.04 g, 1.84 mmol, yield 68.9%). MS m/z=563 [M+H]+.
At room temperature, 2-3 (1.04 g, 1.84 mmol) was dissolved in anhydrous methanol. Pd/C (250 mg, 0.6 mmol) and triethylamine (558 mg, 5.52 mmol) were added. The reaction was carried out at room temperature after replacing with hydrogen. After the reaction was completed, the mixture was filtered. The filtrate was concentrated and purified by silica gel column (PE/EA=5:1-1:1, v/v) to obtain 2-4 (625 mg, 1.18 mmol, yield 64.13%). MS m/z=529 [M+H]+.
At 0° C., 2-4 (40 g, 75.67 mmol) was dissolved in DCM (70 mL), and TFA (30 mL) was slowly added dropwise. The mixture was warmed to room temperature and stirred for 0.5 h. After the reaction was completed, the mixture was concentrated, adjusted to a weak alkaline state with a NaHCO3 aqueous solution, extracted with DCM and concentrated to obtain a crude product M1, which was directly used in the next reaction. MS m/z=429 [M+H]+.
At 0° C., M1 intermediate 2-2 (1.0 g, 2.67 mmol) and DBU (488.13 mg, 3.21 mmol) were dissolved in THF. After adding 1-5 (601 mg, 2.67 mmol), the reaction mixture was warmed to room temperature and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=10:1-5:1, v/v) to obtain 2-5 (1.0 g, 1.73 mmol, yield 64.9%). MS m/z=577 [M+H]+.
At room temperature, 2-5 (1.0 g, 1.73 mmol) was dissolved in anhydrous methanol. Pd/C (250 mg, 0.6 mmol) and triethylamine (558 mg, 5.52 mmol) were added. The reaction was carried out at room temperature after replacing with hydrogen. h. After the reaction was completed, the reaction mixture was filtered. The filtrate was concentrated and purified by silica gel column (PE/EA=5:1-1:1, v/v) to obtain 2-6 (720 mg, 1.33 mmol, yield 76.79%). MS m/z=543 [M+H]+.
At 0° C., 2-6 (4.0 g, 7.38 mmol) was dissolved in DCM (7 mL), and TFA (3 mL) was slowly added dropwise. The mixture was warmed to room temperature and stirred for 0.5 h. After the reaction was completed, the mixture was concentrated, adjusted to a weak alkaline state with a NaHCO3 aqueous solution, extracted with DCM and concentrated to obtain a crude product M2, which was directly used in the next reaction. MS m/z=443 [M+H]+.
Referring to the synthesis method of intermediate M1, 1-7, 1-11, 1-15, 1-17, 1-22, and 1-27 were used instead of 1-3, respectively, to obtain the corresponding intermediates M3-M8.
At room temperature, 1-3 (10 g, 44.39 mmol) and 5-bromopyrimidine (21.17 g, 133.18 mmol) were dissolved in DMF, added with cesium carbonate (43.39 g, 133.18 mmol), heated to 130° C. and stirred overnight. After the reaction was completed, the reaction mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=4:1, v/v) to obtain 3-1 (11 g, 36.26 mmol, yield 81.68%). MS m/z=304 [M+H]+.
3-1 (8.0 g, 26.37 mmol) was dissolved in DCM, and m-CPBA (13.61 g, 79.12 mmol) was slowly added at 0° C. The mixture was warmed to room temperature and stirred for 24 h. After the reaction was completed, the mixture was cooled to 0° C., quenched with saturated aqueous sodium thiosulfate solution, and extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain 3-2 (5.6 g, 17.54 mmol, yield 66.51%). MS m/z=320 [M+H]+.
Triethylamine (3.55 g, 35.07 mmol) was dissolved in DCM, and POCl3 (4.03 g, 26.31 mmol) was slowly added dropwise at 0° C. Then a solution of 3-2 (5.6 g, 17.54 mmol) in DCM was added dropwise to the reaction solution. After the addition was completed, the reaction solution was warmed to room temperature and stirred overnight. After the reaction was completed, the reaction solution was slowly poured into ice water, stirred for half an hour, and then extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified with silica gel column to obtain 3-3 (1.7 g, 5.03 mmol, yield 28.67%). MS m/z=338 [M+H]+.
3-3 (1.0 g, 2.96 mmol) was dissolved in isopropanol, and tert-butyl
2,7-diazaspiro[3.5]nonan-7-carboxylate (871 mg, 3.85 mmol) and DIPEA (1.15 g, 8.88 mmol) were added at room temperature. The mixture was heated to 80° C. and stirred for 2 h. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=2:1, v/v) to obtain 3-4 (1.54 g, 2.92 mmol, yield 98.6%). MS m/z=528 [M+H]+.
At 0° C., 3-4 (1.54 g, 2.92 mmol) was dissolved in DCM (20 mL), and TFA (10 mL) was slowly added dropwise. The mixture was warmed to room temperature and stirred for 0.5 h. After the reaction was completed, the mixture was concentrated, adjusted to a weak alkaline state with a NaHCO3 aqueous solution, extracted with DCM and concentrated to obtain a crude product, which was directly used in the next reaction. MS m/z=428 [M+H]+.
Referring to the synthesis method of intermediate N1, 1-5, 1-7, 1-11, 1-15, and 1-25 were used instead of 1-3 to obtain the corresponding intermediates N2-N6.
At room temperature, hydroxylamine hydrochloride (3.34 g, 48.06 mmol) and KOAc (4.72 g, 48.08 mmol) were added to a solution of ethyl p-cyclohexanonecarboxylate (6.8 g, 40 mmol) in EtOH (80 mL). The reaction mixture was heated to 80° C. and stirred for 4 h. After the reaction was completed, the organic phase was removed by rotary evaporation to obtain a crude product, which was diluted with water and extracted with ethyl acetate. The organic phases were combined and concentrated under reduced pressure to obtain a crude product, which was purified by silica gel column (PE/EA=5:1, v/v) to obtain 4-2 (4.08 g, 22.02 mmol, yield 55.05%). MS m/z=186 [M+H]+.
4-2 (372 mg, 2.01 mmol) and sodium bicarbonate (4.22 g, 50.21 mmol) were dissolved in ethyl acetate (60 mL) and water (60 mL), respectively. TCCA (2.33 g, 10.04 mmol) was added in portions while mixing the above two phases. The organic phase turned blue. After stirring at room temperature for 9 h, the organic phase changed from blue to colorless. After the reaction was completed, the reaction solution was placed in a separatory funnel and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=10:1, v/v) to obtain 4-3 (320 mg, 1.36 mmol, yield 67.66%). MS m/z=236 [M+H]+.
At −20° C., NaBH4 (4.05 g, 106.93 mmol) was added in portions to a solution of 4-3 (18 g, 76.38 mmol) and Pd/C (1.27 g, 10.49 mmol) in EtOH (200 mL). The reaction was completed when the addition was completed. After the reaction was completed, the mixture was filtered through diatomaceous earth. The filtrate was concentrated, dissolved in EA, washed with water followed by saturated NaCl, dried over anhydrous Na2SO4, filtered and dried to obtain a concentrated crude product, which was purified by silica gel column (PE/EA=10:1, v/v) to obtain 4-4 (8.4 g, 41.75 mmol, yield 54.67%). MS m/z=202 [M+H]+.
At 0° C., DBU (1.97 g, 12.94 mmol) was added dropwise to a solution of 4-4 (2.17 g, 10.78 mmol) in MeCN (30 mL), and methyl acrylate (1.11 g, 12.94 mmol) was then added dropwise thereto to perform a reaction at 0° C. for 1 h. After the reaction was completed, the mixture was added with water, extracted with EA, washed with water followed by saturated NaCl, dried over anhydrous Na2SO4, filtered, dried, separated and purified by silica gel column chromatography (PE/EA=40/1→10/1) to obtain products 4-5a (1.035 g, 3.6 mmol, yield 33.39%, obtained by PE/EA=30/1, MS m/z=288 [M+H]+) and 4-5b (1.424 g, 4.96 mmol, yield 46%, obtained by PE/EA=10/1, MS m/z=288 [M+H]+). TLC developing solvent PE/EA=5/1:4-5a, Rf=0.5; 4-5b, Rf=0.3.
At −10° C., NaBH4 (217.26 mg, 5.74 mmol) was added in portions to 4-5a (330 mg, 1.15 mmol) and NiCl2·6H2O (272.88 mg, 1.15 mmol) in MeOH (3 mL), and the mixture was reacted at −10° C. for 2 h. Then, an aqueous solution (1 mL) of K2CO3 (634.94 mg, 4.59 mmol) was added dropwise, and the mixture was reacted at 0° C. for 2 h. After the reaction was completed, the mixture was filtered through diatomaceous earth. The filtrate was adjusted to neutral or acidic with 1N HCl, concentrated, and extracted with EA. The organic layers were combined, washed with water followed by saturated NaCl, dried over anhydrous Na2SO4, filtered and dried, and separated and purified by silica gel column chromatography (DCM/MeOH=40:1) to obtain the product 4-6a (197 mg, 0.874 mmol, yield 7.6%). MS m/z=226 [M+H]+. The synthesis method of 4-6b was the same as above.
Under ice bath, LiAlH4 (120.88 mg, 3.19 mmol) was added to a solution of 4-6a (598 mg, 2.65 mmol) in THF (10 mL). The mixture was stirred 0° C. for 1 h, quenched with water (100 μL), then added with aq. NaOH (15% wt, 100 μL) and H2O (300 μL), stirred at room temperature for 10 min, and filtered. The filtrate was concentrated to obtain the crude product 4-7a (457 mg, 2.49 mmol, yield 93.95%), which was used directly in the next step without further purification. MS m/z=184 [M+H]+. The synthesis method of 4-7b was the same as above.
To a solution of 4-7a (457 mg, 2.49 mmol) in DCM (12 mL)/DMSO (4 mL), DIPEA (1.29 g, 9.98 mmol, 1.74 mL) was added, and then a suspension of Py·SO3 (1.57 g, 9.98 mmol) in DMSO (4 mL) was added dropwise. The reaction mixture was stirred at room temperature for 10 min, then cooled to 0° C., and added with DCM (10 mL)/1N aq HCl (10 mL). The separated aqueous phase was extracted with EA. The combined organic phases were washed with 1 N HCl followed by saturated brine and dried over anhydrous sodium sulfate. The crude product was concentrated under reduced pressure, separated and purified by silica gel column to obtain A1-a (279 mg, 1.54 mmol, yield 61.73%), 1H NMR (400 MHz, Chloroform-d) δ 9.68 (s, 1H), 7.11 (s, 1H), 2.40 (t, J=8.1 Hz, 2H), 2.35-2.24 (m, 1H), 2.00-1.87 (m, 2H), 1.78-1.70 (m, 2H), 1.68-1.53 (m, 4H). MS m/z=184 [M+H]+. The synthesis method of A1-b was the same as above.
At −78° C., n-BuLi (11 mL, 27.5 mmol, 2.5 M in hexane) was added dropwise to a solution of diisopropylamine (2.78 g, 27.46 mmol) in diethyl ether (20 mL). The mixture was gradually warmed to −11° C., and a solution of 2-cyclohexene-1-one (2.4 g, 24.97 mmol) in diethyl ether (20 mL) was added dropwise to the mixture. The temperature of the reaction solution was kept between −11° C. and −3° C. during the addition process. The reaction solution was stirred for 25 min, and then a solution of methyl acrylate (2.15 g, 24.97 mmol) in THF (20 mL) was added dropwise. Afterwards, the mixture was stirred at −10° C. for 1 h. After the reaction was completed, the reaction solution was poured into a saturated ammonium chloride solution (200 mL) and stirred for 15 min. The mixed solution was extracted with EA, and the combined organic phase was dried over anhydrous sodium sulfate and concentrated to obtain a crude product which was separated and purified by a silica gel column (PE/EA=5:1, v/v) to obtain 4-9 (2.1 g, 11.52 mmol, yield 46.16%).
At room temperature, hydroxylamine hydrochloride (2.21 g, 31.8 mmol) and NaOAc (2.61 g, 31.8 mmol) were added to a solution of 4-9 (4.83 g, 26.5 mmol) in EtOH (25 mL). The reaction mixture was heated to 80° C. and stirred for 4 h. After the reaction was completed, the organic phase was removed by rotary evaporation to obtain a crude product, which was diluted with water and extracted with ethyl acetate. The combined organic phases were concentrated under reduced pressure to obtain a crude product, which was separated and purified by silica gel column to obtain 4-10 (4.45 g, 22.47 mmol, yield 84.7%). MS m/z=198 [M+H]+.
Trichloroisocyanuric acid (26.2 g, 112.81 mmol) was added dropwise to a turbid solution of 4-10 (4.45 g, 22.56 mmol) and NaHCO3 (37.6 g, 564 mmol) in EA (500 mL)/H2O (500 mL). After stirring at room temperature for 20 min, the reaction solution turned blue and stirred continuously for 9 h until the organic phase turned into a colorless solution. After the reaction was completed, the mixed solution was extracted with EA. The organic phases were combined, washed with water followed by saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was separated and purified by silica gel column to obtain 4-11 (4.48 g, 18.13 mmol, 80.36% yield).
At 0° C., NaBH4 (950 mg, 24.92 mmol) was added in portions to a solution of 4-11 (4.4 g, 17.8 mmol) and Pd/C (300 mg) in EtOH (80 mL), and the mixture was stirred at room temperature for 1 h. After the reaction was completed, Pd/C was filtered off, and the filtrate was concentrated and purified by silica gel column to obtain 4-12 (3.04 g, 14.27 mmol, 80% yield).
At 0° C., DBU (2.56 mL, 17.11 mmol) was added to a solution of 4-12 (3.04 g, 14.26 mmol) in MeCN (30 mL), and then methyl acrylate (1.54 mL, 17.11 mmol) was added dropwise. The reaction mixture was stirred at 0° C. for 1 h. After the reaction was completed, water was added to quench the reaction, and EA was added for extraction. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was separated and purified by silica gel column (PE/EA=10:1, v/v) to obtain 4-13a (1.66 g, 5.55 mmol, yield 38.92%) and 4-13b (1.55 g, 5.18 mmol, yield 36.32%). TLC developing agent PE/EA=5/1:4-13a, Rf=0.40; 4-13b, Rf=0.35).
Referring to the method of steps 5 to 7 in the synthetic route of A1-a, intermediates B1-a and B1-b can be obtained, wherein B1-a and B1-b were not further resolved but directly used in the next step. MS m/z=208 [M+H]+.
5-1 (hydrochloride, 193 g, 1.0 mol) was placed in a 5 L beaker, to which 400 mL of a mixed solvent (DCM/IPA=3/1) was added, and potassium carbonate (1.10 g, 0.8 mol) aqueous solution was slowly added under ice bath, and the temperature was controlled below 20° C. The mixed solvent was repeatedly extracted 3-4 times, and the organic phases were combined and concentrated to obtain a crude product for direct reaction in the next step.
The crude product was dissolved in DCE (2.5 L), and cooled to 10° C. m-CPBA (6.88 g, 4 mol) was added in portions, and the temperature was controlled below 35° C. After the addition was completed, the reaction solution was heated to reflux and stirred for 3 h. After the reaction was completed, the reaction solution was cooled to below 10° C., stirred for 20 min and filtered. The reaction bottle was rinsed with the filtrate, and the filter cake was rinsed with an appropriate amount of DCE and dried. The filtrate was carefully quenched with Na2SO3 aqueous solution and detected with starch potassium iodide test paper. The filtrate was extracted three times with DCM, concentrated, and purified by silica gel column chromatography (PE/EA=80/1->30/1) to obtain product 5-2 (138 g, 0.737 mol, yield 73.7%). MS m/z=188 [M+H]+.
Referring to the synthesis method of 4-5a and 4-5b, the target intermediate 5-3a (PE/EA=30/1, MS m/z=274 [M+H]+) and 5-3b (PE/EA=20/1, MS m/z=274 [M+H]+) were obtained by silica gel column purification. TLC developing agent PE/EA=5/1:5-3a, Rf=0.3; 5-3b, Rf=0.2.
Referring to the synthesis method of 4-6a, 5-3a and 5-3b were used as raw materials to obtain 5-4a (MS m/z=212 [M+H]+) and 5-4b (MS m/z=212 [M+H]+).
5-4a (2.5 g, 118.34 mmol), (Boc)2O (103.31 g, 473.36 mmol) and DMAP (5.78 g, 47.34 mmol) were dissolved in acetonitrile (250 mL) and reacted at 60° C. overnight under nitrogen protection. After the reaction was completed, the mixture was directly concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1-3:1, v/v) to obtain 6-1a (34.28 g, 110.09 mmol, yield 93.02%). MS m/z=312 [M+H]+.
6-1a (100 mg, 0.307 mmol) was dissolved in THF (20 mL), cooled to −78° C., added with LiHMDS (0.614 mmol, 2.0 eq) dropwise under nitrogen protection, and stirred at this temperature for 1 h. Then the mixture was slowly added with a solution of MOMBr (96 mg, 0.768 mmol) in THE dropwise, heated to room temperature, and stirred for 2 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction, and the mixture was extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by MPLC to obtain 6-2a (99 mg, 0.239 mmol, yield 77.85%). MS m/z=400 [M+H]+.
6-2a (244 mg, 0.59 mmol) was dissolved in anhydrous DCM (7 mL), added with TFA (3 mL) dropwise, and stirred at room temperature for 1 h. After the reaction was completed, the mixture was concentrated to obtain a crude product (114 mg, 0.363 mmol) which was directly used for the next step. MS m/z=300 [M+H]+.
6-3a (114 mg, 0.363 mmol) was dissolved in THF (10 mL), added with LiAlH4 (16.57 mg, 0.436 mmol) at 0° C., and stirred at this temperature for 1 h. After the reaction was completed, sodium sulfate decahydrate was added in portions under ice bath until no bubbles were generated in the system. After filtration, the filtrate was concentrated to obtain a crude product, which was directly used for the next step. MS m/z=272 [M+H]+.
6-4a (100 mg, 0.368 mmol) was dissolved in a mixed solvent of DCM/DMSO=6 mL/2 mL and cooled to 0° C., and DIPEA (190.51 mg, 1.47 mmol) and sulfur trioxide pyridine (234.62 mg, 1.47 mmol) in DMSO (2 mL) were added dropwise in sequence. The mixture was warmed to room temperature and stirred for 30 min. After the reaction was completed, the system was moved to an ice bath, quenched with 1 N HCl, diluted with water, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain C1 (75 mg, 0.278 mmol, yield 75.54%). MS m/z=270 [M+H]+.
6-1a (2.8 g, 8.7 mmol) was dissolved in THE, cooled to −78° C., added with LiHMDS (15.96 ml, 2.4 eq) dropwise under nitrogen protection, stirred at this temperature for 1 h, then slowly added with a solution of iodomethane (2.93 g, 20.65 mmol) in THE dropwise, heated to room temperature and stirred for 2 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction, and the mixture was extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1, v/v) to obtain 6-5a (2.4 g, 6.85 mmol, yield 78.73%). 1H NMR (400 MHZ, Chloroform-d) δ 9.67 (s, 1H), 6.51 (s, 1H), 2.32-2.22 (m, 1H), 2.00-1.90 (m, 2H), 1.85 (s, 2H), 1.79-1.66 (m, 2H), 1.63-1.50 (m, 4H), 1.23 (s, 6H). MS m/z=340 [M+H]+.
Referring to the synthesis method of C1, intermediate C2 was obtained. MS m/z=210 [M+H]+.
4-4 (100 mg, 0.496 mmol) was dissolved in acetonitrile, added with DBU (150 mg, 0.596 mmol) and methyl 2-(bromomethyl) acrylate (106 mg, 0.596 mmol) at 0° C., warmed to room temperature, and stirred for 2 h. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1, v/v) to obtain 6-8a (52 mg, 0.173 mmol, yield 34.8%, containing two isomers). MS m/z=300 [M+H]+.
Trimethylsulfoxide iodide (19.12 g, 86.86 mmol) was dissolved in DMSO, added with NaH (2.08 g, 86.86 mmol) at 0° C., slowly warmed to room temperature, stirred for 1 h and then added with 6-8a (20 g, 66.82 mmol) to perform a reaction at room temperature for 17 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction and the mixture was extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=4:1, v/v) to obtain 6-9a (8.5 g, 27.13 mmol, yield 40.6%, containing two isomers). MS m/z=314 [M+H]+.
Referring to the synthesis method of C1, intermediate C3 (containing two isomers) was obtained. MS m/z=208 [M+H]+.
Referring to the synthesis method of C1, deuterated iodomethane was used instead of iodomethane to obtain intermediate C4. MS m/z=216 [M+H]+.
10-1a (1.7 g, 6.0 mmol) was dissolved in DCM (40 mL), and TEA (2.43 g, 24 mmol), TsCl (465.45 mg, 6.6 mmol) and DMAP (73.29 mg, 0.60 mmol) were added in sequence. The mixture was stirred at 0° C. for 2 h under nitrogen protection. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=3:1, v/v) to obtain 6-15a (2.1 g, 4.8 mmol, yield 80%). MS m/z=438 [M+H]+.
6-15a (200 mg, 0.45 mmol) was dissolved in anhydrous THF (3 mL), cooled to −78° C., slowly added with LiHMDS (1 mL, 1.28 mmol) under nitrogen protection, stirred at this temperature for 1 h, then slowly added with NFSI (248.28 mg, 1.28 mmol) in THF (3 mL), and stirred at room temperature for 4 h. The mixture was cooled again to −78° C., slowly added with LiHMDS (1 mL, 1.28 mmol), stirred for 1 h, then slowly added with NFSI (248.28 mg, 1.28 mmol) in THF (3 mL), and stirred at room temperature for 16 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction and the mixture was extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by MPLC to obtain C5 (90 mg, 0.19 mmol, yield 42.2%). MS m/z=474 [M+H]+.
6-15a (500 mg, 1.14 mmol) was dissolved in anhydrous THF (10 mL), cooled to −30° C., and LDA (183.61 mg, 1.71 mmol) and methyl methylthiosulfonate (216.3 mg, 1.17 mmol) were slowly added dropwise under nitrogen protection. The mixture was stirred at room temperature for 20 h. After the reaction was completed, saturated ammonium chloride solution was added to quench the reaction and the mixture was extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1, v/v) to obtain 6-16a (303 mg, 0.57 mmol, yield 50%). MS m/z=530 [M+H]+.
6-16a (303 mg, 0.57 mmol) was dissolved in a mixed solvent of acetonitrile and water (5 mL: 0.5 mL), cooled to 0° C., added with PIFA (757.93 mg, 1.76 mmol) under nitrogen protection, and then stirred at the same temperature for 3 h. After the reaction was completed, saturated sodium bicarbonate solution was added to quench the reaction and the mixture was extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (DCM/MeOH=100:1, v/v) to obtain 6-17a (170 mg, 0.37 mmol, yield 64.91%). MS m/z=452 [M+H]+.
At 0° C., 6-17a (170 mg, 0.376 mmol) was dissolved in DCM (2 mL), slowly added with TFA (1 mL) dropwise, warmed to room temperature, and stirred for 0.5 h. After the reaction was completed, the mixture was concentrated to obtain a crude product which was used directly in the next step without further purification. MS m/z=352 [M+H]+.
4-6a (12.5 g, 55.5 mmol), (Boc)2O (51.6 g, 237 mmol) and DMAP (2.89 g, 23.7 mmol) were dissolved in acetonitrile (250 mL) and reacted at 60° C. overnight under nitrogen protection. After the reaction was completed, the mixture was directly concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=5:1-3:1, v/v) to obtain 6-18a (17 g, 52 mmol, yield 93.69%). MS m/z=326 [M+H]+.
6-18a (500 mg, 1.54 mmol) was dissolved in THF (1.5 mL), cooled to −78° C., added
with LiHMDS (3 ml, 3.84 mmol) dropwise under nitrogen protection, and stirred at this temperature for 1 h. Then a solution of 2-iodoethyl ether (751 mg, 2.3 mmol) in THF (1.5 mL) was slowly added dropwise, warmed to room temperature and stirred for 2 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction and the mixture was extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by MPLC to obtain 6-19a (62 mg, 0.156 mmol, yield 10.12%). MS m/z=396 [M+H]+.
Referring to the synthesis method of C1, the intermediate C7 (45 mg, 0.179 mmol) was obtained by the same experimental operation. MS m/z=252 [M+H]+.
6-1a (34.28 g, 110.09 mmol) was dissolved in THE, cooled to −78° C., added with LiHMDS (93 ml, 1.1 eq) dropwise under nitrogen protection, and stirred at this temperature for 1 h. Then a solution of MOMBr (17.89 g, 143.12 mmol) in THF was slowly added, warmed to room temperature and stirred for 2 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction and the mixture was extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by MPLC to obtain 7-1a (25.6 g, 72.03 mmol, yield 65.43%). MS m/z=356 [M+H]+.
7-1a (25.6 g, 72.03 mmol) was dissolved in anhydrous DCM (250 mL), added with TFA (40 mL) dropwise, and stirred at room temperature for 1 h. After the reaction was completed, the concentrated crude product was directly used in the next step. MS m/z=256 [M+H]+.
7-2a (18.34 g, 71.83 mmol) was dissolved in THF (400 mL), added with LiAlH4 (3.27 g, 86.20 mmol) at 0° C., and stirred at this temperature for 1 h. After the reaction was completed, sodium sulfate decahydrate was added in portions under ice bath until no bubbles were generated in the system. After filtration, the filtrate was concentrated to obtain the crude product, which was directly used in the next step. MS m/z=228 [M+H]+.
7-3a (16.16 g, 71.10 mmol) was dissolved in anhydrous DMF (160 mL), added with imidazole (7.26 g, 106.64 mmol) and TBDPS-Cl (29.31 g, 106.64 mmol), and stirred at room temperature for 1 h. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=1:1, v/v) to obtain 7-4a (33 g, 70.86 mmol, yield 99.7%). MS m/z=466 [M+H]+.
7-4a was resolved by SFC into isomers 7-4a1 (SFC peak retention time: 2.986 min) and 7-4a2 (SFC peak retention time: 4.269 min).
SFC method of intermediates 7-4a1 and 7-4a2: chiral column model: CHIRALPAK AS; specification: 3 um, 150 mm*3 mm; mobile phase A-CO2, mobile phase B-ethanol, A/B=70/30: flow rate: 1 mL/min; column temperature: 40° C.
7-4a1 (654 mg, 1.40 mmol) was dissolved in THF (10 mL), added with TBAF (1.47 g, 5.62 mmol), and stirred at room temperature overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified with a silica gel column (PE/EA=1:1, v/v) to obtain 7-5a1 (300 mg, 1.32 mmol, yield 94.28%). MS m/z=228 [M+H]+. (The synthesis method of 7-5a2 was the same as above)
7-5a1 (300 mg, 1.32 mmol) was dissolved in a mixed solvent of DCM/DMSO=24 mL/4 mL, cooled to 0° C., added with solutions of DIPEA (682.31 mg, 5.28 mmol) and sulfur trioxide pyridine (840.27 mg, 5.28 mmol) in DMSO (2 mL) dropwise sequentially, warmed to room temperature and stirred for 30 min. After the reaction was completed, the system was moved to an ice bath, quenched with 1 N HCl, diluted with water, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain D1-a1 (130 mg, 1.02 mmol, yield 77.27%) of single configuration. 1H NMR (400 MHZ, Chloroform-d) δ 9.67 (s, 1H), 7.01 (s, 1H), 3.59 (d, J=5.0 Hz, 2H), 3.36 (s, 3H), 2.76 (tt, J=9.5, 4.9 Hz, 1H), 2.33-2.21 (m, 1H), 2.16 (dd, J=13.0, 9.5 Hz, 1H), 2.00-1.90 (m, 2H), 1.85-1.69 (m, 3H), 1.67-1.48 (m, 4H). MS m/z=226 [M+H]+. The synthesis method of another single configuration D1-a2 was the same as above.
Referring to the synthesis methods of D1-a1 and D1-a2, iodomethane was used instead of MOMBr to obtain intermediates D2-a1 (MS m/z=196 [M+H]+) and D2-a2 (MS m/z=196 [M+H]+) of single configuration, respectively.
SFC method of intermediates 8-4a1 and 8-4a2: chiral column model: CHIRAL ART Cellulose-SC; specification: 3 um, 150 mm*3 mm; mobile phase A-CO2, mobile phase B-isopropanol, A/B=70/30: flow rate: 1 mL/min; column temperature: 40° C.; SFC peak times of 8-4a1 and 8-4a2 were 3.809 min and 4.479 min respectively.
Referring to the synthesis methods of D1-a1 and D1-a2, deuterated iodomethane was used instead of MOMBr to obtain intermediates D3-a1 (MS m/z=199 [M+H]+) and D3-a2 (MS m/z=199 [M+H]+) of single configuration, respectively.
SFC method of intermediates 9-4a1 and 9-4a2: chiral column model: CHIRAL ART Cellulose-SC; specification: 3 um, 150 mm*3 mm; mobile phase A-CO2, mobile phase B-isopropanol, A/B=65/35: flow rate: 1 mL/min; column temperature: 40° C.; SFC peak times of 9-4a1 and 9-4a2 were 2.936 min and 3.386 min respectively.
4-7a (1.83 g, 10 mmol), (Boc)2O (8.72 g, 40 mmol) and DMAP (0.48 mg, 4 mmol) were dissolved in acetonitrile (20 mL) and reacted at 60° C. overnight under nitrogen protection. After the reaction was completed, the mixture was directly concentrated to obtain a crude product which was purified by silica gel column (PE/EA=5:1-3:1, v/v) to obtain 10-1a (2.9 g, 9.3 mmol, yield 93%). MS m/z=284 [M+H]+.
10-1a (2.9 g, 9.3 mmol) was dissolved in anhydrous DMF (10 mL), added with imidazole (0.95 g, 13.88 mmol) and TBDPS-Cl (3.81 g, 13.88 mmol), and stirred at room temperature for 1 h. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=1:1, v/v) to obtain 10-2a (4.83 g, 9.26 mmol, yield 99.57%). MS m/z=522 [M+H]+.
10-2a (916 mg, 1.76 mmol) was dissolved in anhydrous THF (10 mL), slowly added with LiHMDS (1.93 mmol, 1.1 eq) dropwise at −70° C., and stirred at this temperature for 1 h. Subsequently, the mixture was added with acetone (112.16 mg, 1.93 mmol) and boron trifluoride etherate (274.22 mg, 1.93 mmol) dropwise at −70° C., warmed to room temperature, and stirred for 2.5 h. After the reaction was completed, saturated ammonium chloride was added to quench the reaction and the mixture was extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by MPLC to obtain 10-3a (883 mg, 1.52 mmol, yield 86.36%). MS m/z=580 [M+H]+.
10-3a (883 mg, 1.52 mmol) was dissolved in THF (10 mL), added with TBAF (1.59 g, 6.08 mmol) and stirred at room temperature overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=1:1, v/v) to obtain 10-4a (488 mg, 1.43 mmol, yield 94.08%). MS m/z=342 [M+H]+.
10-4a (488 mg, 1.43 mmol) was dissolved in a mixed solvent of DCM/DMSO=24 mL/4 mL, cooled to 0° C., added dropwise with solutions of DIPEA (784.65 mg, 6.07 mmol) and sulfur trioxide pyridine (966.31 mg, 6.07 mmol) in DMSO (2 mL) sequentially, warmed to room temperature and stirred for 30 min. After the reaction was completed, the system was moved to an ice bath, quenched with 1 N HCl, diluted with water, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain D4-a (397.88 mg, 1.17 mmol, yield 81.8%, containing 2 isomers, which were not resolved and used directly in the next step). MS m/z=340 [M+H]+.
10-3a (499 mg, 0.86 mmol) was dissolved in DCM (8 mL), added with TEA (870.82 mg, 8.61 mmol) and MsCl (98.58 mg, 0.86 mmol) dropwise at 0° C., warmed to room temperature and stirred for 2 days. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain 10-5a (263 mg, 0.47 mmol, yield 54.7%). MS m/z=562 [M+H]+.
Referring to the synthesis method of D4-a, intermediate D5-a was obtained by TBDPS deprotection and oxidation. MS m/z=322 [M+H]+.
4-1 (1.65 g, 8.20 mmol) was dissolved in acetonitrile (7.37 mL), added with TEA (912.74 mg, 9.02 mmol) and formaldehyde aqueous solution (492.43 mg, 16.2 mmol) dropwise at 0° C., warmed to room temperature and stirred overnight. After the reaction was completed, the mixture was directly concentrated to obtain a crude product which was purified by silica gel column (PE/EA=4:1, v/v) to obtain stereoisomer 12-1a (764 mg, 3.3 mmol, obtained by PE/EA=4/1, MS m/z=232 [M+H]+) and another stereoisomer 12-1b (1.09 g, 4.7 mmol, yield 57.3%, obtained by PE/EA=2/1, MS m/z=232 [M+H]+). TLC developing agent PE/EA=1/1:12-1a, Rf=0.3; 12-1b, Rf=0.2.
12-1a (746 mg, 3.63 mmol) was dissolved in isopropanol (20 mL), added with Raney Ni (70 mg, 3.23 mmol), replaced with hydrogen, heated to 70° C. and stirred overnight. After the reaction was completed, the mixture was filtered. The filtrate was concentrated to obtain the crude product (640 mg, 3.18 mmol, yield 87.6%), which was directly used for the next step without purification. MS m/z=202 [M+H]+.
12-2a (640 mg, 3.18 mmol) was dissolved in THF (25 mL), added with TEA (386.13 mg, 3.82) dropwise, cooled to −10° C., and slowly added with a THF solution of triphosgene (350 mg, 1.18) dropwise. The reaction solution was stirred at this temperature for 30 min, then moved to room temperature and stirred for 2 h. After the reaction was completed, saturated sodium bicarbonate and ethyl acetate were added for extraction for three times. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=1:1, v/v) to obtain 12-3a (380 mg, 1.67 mmol, yield 52.5%). MS m/z=228 [M+H]+.
12-3a (380 mg, 1.67 mmol) was dissolved in THF (10 mL), added with LiAlH4 (76.15 mg, 2.01 mmol) at 0° C., and stirred at this temperature for 1 h. After the reaction was completed, sodium sulfate decahydrate was added in portions under ice bath until no bubbles were generated in the system. After filtration, the filtrate was concentrated to obtain a crude product (304 mg, 1.64 mmol, yield 98.2%), which was directly used for the next step. MS m/z=186 [M+H]+.
12-4a (304 mg, 1.64 mmol) was dissolved in a mixed solvent of DCM/DMSO=12 mL/2 mL, cooled to 0° C., added with solutions of DIPEA (848.48 mg, 6.57 mmol) and sulfur trioxide pyridine (1.04 g, 6.57 mmol) in DMSO (2 mL) dropwise sequentially, warmed to room temperature and stirred for 30 min. After the reaction was completed, the system was moved to an ice bath, quenched with 1 N HCl, diluted with water, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain E2-a (single configuration, 167 mg, 0.91 mmol, yield 55.5%). 1H NMR (400 MHZ, Chloroform-d) δ 9.67 (s, 1H), 6.42 (s, 1H), 4.11 (s, 2H), 2.40-2.30 (m, 1H), 2.03-1.94 (m, 2H), 1.87-1.79 (m, 2H), 1.73-1.56 (m, 4H). MS m/z=184 [M+H]+.
Referring to the synthesis method of E2-a, another intermediate E2-b of single configuration can be obtained by the same experimental operation. MS m/z=184 [M+H]+.
Referring to the synthesis method of E2-a, intermediates E3-a (obtained by PE/EA=5/1, MS m/z=210 [M+H]+, used directly in the next step without resolution) and E3-b (obtained by PE/EA=3/1, MS m/z=210 [M+H]+, used directly in the next step without resolution) were obtained by the same experimental operation. TLC developing solvent PE/EA=5/1: E3-a, Rf=0.4; E3-b, Rf=0.3.
14-1a (referring to the synthesis of 12-2a, obtained from raw material 5-2, 720 mg, 3.85 mmol, the isomer with less polarity during isomer resolution), (Boc)2O (1.01 g, 4.61 mmol) and TEA (778 mg, 7.67 mmol) were dissolved in acetonitrile (20 mL) and reacted at 60° C. overnight under nitrogen protection. After the reaction was completed, the mixture was directly concentrated to obtain a crude product which was purified by silica gel column (PE/EA=5:1-3:1, v/v) to obtain 14-2a (650 mg, 2.26 mmol, yield 58.7%). MS m/z=288 [M+H]+.
14-2a (650 mg, 2.26 mmol) was dissolved in DCM (15 mL), cooled to 0° C., added with Dess-Martin oxidant (1.92 g, 4.52 mmol) in portions under nitrogen protection, warmed to room temperature and stirred for 2 h. After the reaction was completed, water was added to quench the solution. After filtration, the filtrate was extracted three times with DCM. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=10:1-8:1, v/v) to obtain 14-3a (537 mg, 1.88 mmol, yield 83.18%). MS m/z=286 [M+H]+.
14-3a (537 mg, 1.88 mmol) and methylamine hydrochloride (190.60 mg, 2.82 mmol) were dissolved in anhydrous methanol (10 mL), and then TEA was added to adjust the pH to about 8. The reaction mixture was stirred at room temperature for 0.5 h. Then, acetic acid was added to adjust the pH to 5, and sodium cyanoborohydride (237.13 mg, 3.76 mmol) was added and stirred at room temperature for 1.5 h. After the reaction was completed, saturated sodium bicarbonate solution was added to quench the solution, and ethyl acetate was added for extraction for three times. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (DCM/MeOH=50:1, v/v) to obtain 14-4a (397 mg, 1.32 mmol, yield 70.21%). MS m/z=301 [M+H]+.
At 0° C., 14-4a (397 mg, 1.32 mmol) was dissolved in DCM (4 mL), slowly added with TFA (2 mL) dropwise, warmed to room temperature and stirred for 0.5 h. After the reaction was completed, the mixture was concentrated to obtain a crude product which was used directly in the next step without further purification. MS m/z=201 [M+H]+.
The crude product obtained in the previous step was dissolved in THF (10 mL), cooled to 0° C., added with CDI (285.37 mg, 1.98 mmol) under nitrogen protection, then warmed to room temperature and stirred for 1 h. After the reaction was completed, saturated sodium bicarbonate solution was added to quench the solution, and ethyl acetate was added for extraction for three times. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (DCM/MeOH=60:1, v/v) to obtain 14-6a (190 mg, 0.84 mmol, yield 63.6%). MS m/z=227 [M+H]+.
14-6a (190 mg, 0.84 mmol) was dissolved in THF (5 mL), added with LiAlH4 (41.43 mg, 1.09 mmol) at 0° C., and stirred at this temperature for 1 h. After the reaction was completed, sodium sulfate decahydrate was added in portions under ice bath until no bubbles were generated in the system. After filtration, the filtrate was concentrated to obtain a crude product (120 mg, 0.6 mmol), which was directly used for the next step. MS m/z=199 [M+H]+.
14-7a (120 mg, 0.6 mmol) was dissolved in a mixed solvent of DCM/DMSO=4 mL/0.5 mL, cooled to 0° C., added with solutions of DIPEA (312.9 mg, 2.24 mmol) and pyridine sulfur trioxide (385.43 mg, 2.42 mmol) in DMSO (0.5 mL) dropwise sequentially, warmed to room temperature and stirred for 30 min. After the reaction was completed, the system was moved to an ice bath, quenched with 1 N HCl, diluted with water, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain one of the single isomers E4-a (35 mg, 0.178 mmol, yield 29.7%). MS m/z=197 [M+H]+.
Similarly, referring to the synthesis method of E4-a, another intermediate E4-b of single configuration can be obtained by the same experimental operation. MS m/z=197 [M+H]+.
N1 (268.92 mg, 629.03 μmol) was added to a solution of A1-a (95 mg, 524.19 μmol) in MeOH (10 mL), and the mixture was stirred at room temperature for 1 h. AcOH was added to adjust the pH to 4-5, and then NaBH3CN (49.54 mg, 786.29 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 1 compound (133.16 mg, 220.16 μmol, 42.00% yield, 98% purity) as a white solid. MS m/z=593 [M+H]+. NMR spectrum 1H NMR (600 MHz, Methanol-d4) δ 8.23 (d, J=9.6 Hz, 1H), 7.72 (d, J=24.6 Hz, 1H), 7.23-7.14 (m, 2H), 6.99 (dd, J=9.0, 4.2 Hz, 1H), 4.04-3.88 (m, 4H), 3.52 (dd, J=13.7, 7.0 Hz, 1H), 3.36 (dd, J=13.7, 7.0 Hz, 1H), 3.26 (q, J=7.1 Hz, 1H), 2.35 (dd, J=8.6, 7.5 Hz, 5H), 2.16 (d, J=6.8 Hz, 2H), 1.96 (t, J=8.1 Hz, 2H), 1.84-1.76 (m, 6H), 1.70 (d, J=12.9 Hz, 2H), 1.54 (td, J=13.3, 3.9 Hz, 3H), 1.31 (d, J=6.5 Hz, 2H), 1.23 (t, J=7.0 Hz, 3H), 1.18 (d, J=6.7 Hz, 2H), 1.15 (d, J=6.7 Hz, 2H), 1.13-1.05 (m, 3H).
N2 (3.95 g, 8.94 mmol) was added to a solution of A1-a (1.62 g, 8.94 mmol) in DCM (30 mL), and the mixture was stirred at room temperature for 1 h. AcOH was added to adjust the pH to 4-5, and then NaBH(OAc)3 (5.68 g, 26.82 m mol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 5 compound (2.14 g, 3.53 mmol, 39.46% yield, 98% purity) as a white solid. MS m/z=607 [M+H]+. NMR spectrum 1H NMR (600 MHZ, CDCl3) δ 8.38 (s, 1H), 7.77 (s, 1H), 7.00 (d, J=7.6 Hz, 2H), 6.87-6.70 (m, 1H), 5.78 (s, 1H), 3.97 (s, 2H), 3.88 (d, J=9.2 Hz, 2H), 3.84-3.76 (m, 1H), 3.56-3.45 (m, 1H), 2.39 (t, J=8.1 Hz, 3H), 2.28 (d, J=15.3 Hz, 3H), 2.11 (d, J=6.7 Hz, 2H), 1.94 (t, J=8.1 Hz, 2H), 1.83 (s, 1H), 1.76 (s, 9H), 1.56 (s, 3H), 1.50 (d, J=6.8 Hz, 3H), 1.45 (td, J=12.8, 3.2 Hz, 3H), 1.15 (d, J=6.6 Hz, 3H), 1.11 (d, J=6.7 Hz, 3H), 1.04 (dd, J=24.3, 11.4 Hz, 2H).
M1 (40 mg, 93.35 μmol) was added to a solution of A1-a (16.92 mg, 93.35 μmol) in MeOH (5 mL), and the mixture was stirred at room temperature for 1 h. AcOH was added to adjust the pH to 4-5, and then NaBH3CN (8.82 mg, 140.02 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 7 compound (40.42 mg, 66.55 μmol, 71% yield, 97% purity) as a white solid. MS m/z=594 [M+H]+. NMR spectrum 1H NMR (600 MHZ, Methanol-d4) δ 8.38 (d, J=1.8 Hz, 1H), 7.44-7.38 (m, 1H), 7.27 (tt, J=8.5, 4.2 Hz, 1H), 7.21 (ddd, J=14.9, 8.0, 3.1 Hz, 1H), 4.36 (d, J=24.3 Hz, 2H), 3.98-3.86 (m, 2H), 3.81 (p, J=6.6 Hz, 1H), 3.53-3.46 (m, 1H), 3.26-3.20 (m, 1H), 2.36 (t, J=8.1 Hz, 5H), 2.18 (d, J=6.8 Hz, 2H), 1.97 (t, J=8.1 Hz, 2H), 1.87 (d, J=5.2 Hz, 4H), 1.83-1.76 (m, 2H), 1.75-1.68 (m, 2H), 1.54 (td, J=13.3, 3.8 Hz, 3H), 1.21 (d, J=6.8 Hz, 2H), 1.15 (d, J=7.0 Hz, 5H), 1.12-1.04 (m, 3H), 0.86-0.75 (m, 2H).
M1 (26.2 mg, 61.26 μmol) was added to a solution of C1 (15 mg, 55.59 μmol) in MeOH (3 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (38.3 mg, 610.1 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 20 compound (200 mg, 287.4 μmol, 70.66% yield, 98% purity) as a white solid. MS m/z=696 [M+H]+.
M2 (180 mg, 406.8 μmol) was added to a solution of C1 (98.6 mg, 366.1 μmol) in MeOH (5 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (38.3 mg, 610.1 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 20 compound (200 mg, 287.4 μmol, 70.66% yield, 98% purity) as a white solid. MS m/z=696 [M+H]+. NMR spectrum 1H NMR (400 MHZ, Chloroform-d) § 8.46 (s, 1H), 7.26-7.23 (m, 1H), 7.10 (ddd, J=9.1, 7.9, 3.0 Hz, 1H), 6.96 (dd, J=7.8, 3.0 Hz, 1H), 5.75 (s, 1H), 4.47 (d, J=10.3 Hz, 1H), 4.28 (d, J=10.3 Hz, 1H), 3.94-3.83 (m, 2H), 3.79 (p, J=6.6 Hz, 1H), 3.46 (d, J=8.9 Hz, 2H), 3.43-3.36 (m, 1H), 3.33 (s, 7H), 3.30 (s, 1H), 2.32 (s, 3H), 2.13-2.02 (m, 4H), 1.90-1.64 (m, 9H), 1.50 (d, J=6.8 Hz, 3H), 1.37 (dd, J=12.9, 8.7 Hz, 6H), 1.16-0.95 (m, 5H), 0.72 (d, J=6.6 Hz, 3H).
N4 (46 mg, 104.6 μmol) was added to a solution of C2 (21.9 mg, 104.6 μmol) in MeOH (5 mL), and the mixture was stirred at room temperature for 1 h. Then AcOH was added to adjust the pH to 4-5, and NaBH3CN (9.85 mg, 156.9 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 23 (29 mg, 45.83 μmol, 43% yield, 98% purity) as a white solid. MS m/z=633 [M+H]+. NMR spectrum 1H NMR (600 MHZ, CDCl3) δ 8.38 (s, 1H), 7.80 (s, 1H), 7.00 (s, 2H), 6.73 (d, J=5.1 Hz, 1H), 5.43 (s, 1H), 4.53 (s, 1H), 3.86 (s, 4H), 2.56 (s, 1H), 2.25 (s, 3H), 2.07 (d, J=4.8 Hz, 2H), 1.86 (s, 2H), 1.73 (s, 11H), 1.43 (s, 3H), 1.35 (d, J=6.4 Hz, 6H), 1.21 (s, 7H), 1.01 (d, J=11.6 Hz, 2H), 0.56 (s, 4H).
M1 (43 mg, 100.34 μmol) was added to a solution of C2 (21 mg, 100.34 μmol) in MeOH (3 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (9.46 mg, 150.51 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 26 (12.63 mg, 20.29 μmol, 20.22% yield, 99% purity) as a white solid. MS m/z=622 [M+H]+. NMR spectrum 1H NMR (400 MHZ, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.51-4.14 (m, 2H), 4.02-3.70 (m, 3H), 3.61-3.39 (m, 1H), 3.28-3.18 (m, 1H), 2.59-2.28 (m, 4H), 2.17 (d, J=6.8 Hz, 2H), 1.93-1.83 (m, 6H), 1.83-1.75 (m, 2H), 1.71-1.63 (m, 2H), 1.63-1.44 (m, 3H), 1.25-1.01 (m, 15H), 0.88-0.76 (m, 2H).
M2 (66.38 mg, 150 μmol) was added to a solution of D1-a2 (33.79 mg, 150 μmol) in MeOH (2 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (14.14 mg, 225 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 38-2 (42.76 mg, 63.63 μmol, 42.42% yield, 97% purity) as a white solid. MS m/z=652 [M+H]+. NMR spectrum 1H NMR (400 MHZ, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J=9.1, 4.5 Hz, 1H), 7.25 (ddd, J=9.1, 8.1, 3.0 Hz, 1H), 7.16 (dd, J=8.0, 3.0 Hz, 1H), 4.48-4.31 (m, 2H), 3.92 (q, J=10.5 Hz, 2H), 3.75 (p, J=6.6 Hz, 1H), 3.63-3.47 (m, 3H), 3.329 (s, 3H), 2.78-2.69 (m, 1H), 2.58-2.11 (m, 7H), 1.92-1.64 (m, 9H), 1.63-1.35 (m, 9H), 1.21-1.00 (m, 5H), 0.75 (d, J=6.6 Hz, 3H).
M2 (44.40 mg, 100.34 μmol) was added to a solution of D2-a1 (19.59 mg, 100.34 μmol) in MeOH (3 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (9.46 mg, 150.51 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 41-1 (25.06 mg, 40.26 μmol, 40.13% yield, 99% purity) as a white solid. MS m/z=622 [M+H]+. NMR spectrum 1H NMR (400 MHZ, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J=9.0, 4.5 Hz, 1H), 7.25 (ddd, J=9.0, 8.0, 3.0 Hz, 1H), 7.16 (dd, J=8.0, 3.0 Hz, 1H), 4.49-4.29 (m, 2H), 3.92 (q, J=10.5 Hz, 2H), 3.75 (hept, J=6.6 Hz, 1H), 3.54 (hept, J=6.9 Hz, 1H), 2.65-2.22 (m, 6H), 2.17 (d, J=6.9 Hz, 2H), 1.91-1.69 (m, 7H), 1.71-1.58 (m, 2H), 1.58-1.31 (m, 9H), 1.24-0.99 (m, 8H), 0.75 (d, J=6.6 Hz, 3H).
M2 (44.40 mg, 100.34 μmol) was added to a solution of D2-a2 (19.59 mg, 100.34 μmol) in MeOH (3 mL), and the mixture was stirred at room temperature for 1 h. AcOH was then added to adjust the pH to 4-5, and NaBH3CN (9.46 mg, 150.51 μmol) was added. The mixture was stirred at room temperature for 16 h. After the reaction was completed, the mixture was separated and purified by Pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 41-2 (20.57 mg, 33.05 μmol, 32.94% yield, 99% purity) as a white solid. MS m/z=622 [M+H]+. NMR spectrum 1H NMR (400 MHZ, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J=9.0, 4.5 Hz, 1H), 7.25 (ddd, J=9.0, 8.0, 3.0 Hz, 1H), 7.16 (dd, J=8.0, 3.0 Hz, 1H), 4.49-4.29 (m, 2H), 3.92 (q, J=10.5 Hz, 2H), 3.75 (hept, J=6.6 Hz, 1H), 3.54 (hept, J=6.9 Hz, 1H), 2.65-2.22 (m, 2H), 2.17 (d, J=6.9 Hz, 2H), 1.91-1.69 (m, 7H), 1.63 (dd, J=11.9, 3.4 Hz, 2H), 1.58-1.31 (m, 9H), 1.24-0.99 (m, 8H), 0.75 (d, J=6.6 Hz, 3H).
Referring to the synthesis method of Example 1, the following example compounds were obtained by reacting raw material 1 and raw material 2:
1H NMR (600 MHz, Methanol-d4) δ 8.52 (d, J = 7.3 Hz, 1H), 7.86 (s, 1H), 7.29 (tdd, J = 13.0, 6.5, 2.6 Hz, 3H), 4.24 (d, J = 69.8 Hz, 4H), 3.99-3.80 (m, 1H), 3.59 (d, J = 12.5 Hz, 2H), 3.51-3.38 (m, 2H), 3.01 (d, J = 7.0 Hz, 4H), 2.37 (t, J = 8.1 Hz, 2H), 2.26 (d, J = 14.0 Hz, 2H), 2.11 (s, 2H), 1.92 (q, J = 8.4 Hz, 3H), 1.77 (q, J = 9.6, 6.9 Hz, 4H), 1.58 (td, J = 14.3, 13.6, 4.1 Hz, 2H), 1.38-1.07 (m, 11H). MS m/z = 593 [M + H]+
1H NMR (600 MHz, Methanol-d4) δ 8.26-8.21 (m, 1H), 7.77-7.69 (m, 1H), 7.24-7.14 (m, 2H), 7.05- 6.95 (m, 1H), 4.10-3.83 (m, 5H), 3.52 (dq, J = 13.9, 7.0 Hz, 1H), 3.36 (dq, J = 14.0, 7.0 Hz, 1H), 2.52-2.12 (m, 9H), 1.93- 1.71 (m, 9H), 1.68-1.46 (m, 6H), 1.37-1.06 (m, 10H). MS m/z = 619 [M + H]+
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Methanol-d4)
1H NMR (600 MHz, Methanol-d4) δ 8.26-8.19 (m, 1H), 7.77-7.69 (m, 1H), 7.23-7.15 (m, 2H), 7.05- 6.94 (m, 1H), 4.08-3.83 (m, 5H), 3.52 (dq, J = 13.9, 7.0 Hz, 1H), 3.36 (dq, J = 13.9, 7.0 Hz, 1H), 2.54-2.15 (m, 9H), 1.96- 1.62 (m, 11H), 1.56-1.43 (m, 4H), 1.38-1.05 (m, 10H). MS m/z = 619 [M + H]+
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Methanol-d4)
1H NMR (600 MHz, CDCl3) δ 8.38 (s, 1H), 7.77 (s, 1H), 7.00 (d, J = 7.6 Hz, 2H), 6.87-6.70 (m, 1H), 5.78 (s, 1H), 3.97 (s, 2H), 3.88 (d, J = 9.2 Hz, 2H), 3.84-3.76 (m, 1H), 3.56-3.45 (m, 1H), 2.39 (t, J = 8.1 Hz, 3H), 2.28 (d, J = 15.3 Hz, 3H), 2.11 (d, J = 6.7 Hz, 2H), 1.94 (t, J = 8.1 Hz, 2H), 1.83 (s, 1H), 1.76 (s, 9H), 1.56 (s, 3H), 1.50 (d, J = 6.8 Hz, 3H), 1.45 (td, J = 12.8, 3.2 Hz, 3H), 1.15 (d, J = 6.6 Hz, 3H), 1.11 (d, J = 6.7 Hz, 3H), 1.04 (dd, J = 24.3, 11.4 Hz, 2H). MS m/z = 607 [M + H]+ ∘
1H NMR (400 MHz, Methanol-d4) δ 8.24 (s, 1H), 7.69 (s, 1H), 7.25- 7.15 (m, 2H), 6.95 (dd, J = 8.9, 4.2 Hz, 1H), 3.93 (s, 4H), 2.70 (s, 2H), 2.49-2.26 (m, 6H), 2.16 (d, J = 6.9 Hz, 2H), 1.96 (t, J = 8.1 Hz, 2H), 1.85-1.74 (m, 6H), 1.74- 1.66 (m, 2H), 1.60-1.42 (m, 3H), 1.16-1.02 (m, 2H), 0.96-0.84 (m, 2H), 0.84-0.69 (m, 4H), 0.69- 0.57 (m, 2H) MS m/z = 603 [M + H]+
1H NMR (600 MHz, Methanol-d4) δ 8.38 (d, J = 1.8 Hz, 1H), 7.44- 7.38 (m, 1H), 7.27 (tt, J = 8.5, 4.2 Hz, 1H), 7.21 (ddd, J = 14.9, 8.0, 3.1 Hz, 1H), 4.36 (d, J = 24.3 Hz, 2H), 3.98-3.86 (m, 2H), 3.81 (p, J = 6.6 Hz, 1H), 3.53-3.46 (m, 1H), 3.26-3.20 (m, 1H), 2.36 (t, J = 8.1 Hz, 5H), 2.18 (d, J = 6.8 Hz, 2H), 1.97 (t, J = 8.1 Hz, 2H), 1.87 (d, J = 5.2 Hz, 4H), 1.83-1.76 (m, 2H), 1.75-1.68 (m, 2H), 1.54 (td, J = 13.3, 3.8 Hz, 3H), 1.21 (d, J = 6.8 Hz, 2H), 1.15 (d, J = 7.0 Hz, 5H), 1.12-1.04 (m, 3H), 0.86-0.75 (m, 2H).MS m/z = 594 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.1, 8.1, 3.1 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.50-4.28 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (p, J = 6.6 Hz, 1H), 3.55 (h, J = 7.1 Hz, 1H), 2.36 (dd, J = 8.7, 7.5 Hz, 6H), 2.17 (d, J = 6.9 Hz, 2H), 1.97 (t, J = 8.1 Hz, 2H), 1.91-1.75 (m, 6H), 1.71 (d, J = 12.9 Hz, 2H), 1.61-1.45 (m, 6H), 1.39 (d, J = 6.7 Hz, 3H), 1.20- 1.01 (m, 5H), 0.75 (d, J = 6.6 Hz, 3H).MS m/z = 608 [M + H]+
1H NMR (600 MHz, CDCl3) δ 8.45 (d, J = 15.3 Hz, 1H), 7.29 (dd, J = 9.0, 4.4 Hz, 1H), 7.23 (d, J = 4.4 Hz, 1H), 7.13 (s, 1H), 7.00 (d, J = 1.9 Hz, 1H), 5.56 (s, 1H), 4.91- 4.58 (m, 1H), 4.52-4.15 (m, 2H), 3.87 (d, J = 6.6 Hz, 3H), 2.85 (s, 2H), 2.65 (s, 2H), 2.37 (s, 3H), 2.32 (s, 2H), 2.10 (d, J = 4.6 Hz, 2H), 1.93 (t, J = 8.0 Hz, 2H), 1.86- 1.70 (m, 8H), 1.65 (s, 1H), 1.44 (t, J = 11.4 Hz, 3H), 1.04 (dd, J = 35.3, 23.2 Hz, 7H), 0.80 (s, 2H) MS m/z = 580 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.36 (s, 1H), 7.40 (dd, J = 9.0, 4.6 Hz, 1H), 7.28 (ddt, J = 17.1, 8.6, 4.4 Hz, 2H), 4.39-4.27 (m, 2H), 3.92 (s, 2H), 3.54-3.42 (m, 2H), 2.74 (td, J = 6.8, 3.5 Hz, 1H), 2.36 (dd, J = 8.7, 7.4 Hz, 6H), 2.18 (d, J = 6.9 Hz, 2H), 1.97 (t, J = 8.1 Hz, 2H), 1.92-1.65 (m, 9H), 1.54 (td, J = 13.1, 3.8 Hz, 3H), 1.11 (dt, J = 11.6, 4.6 Hz, 5H), 0.63 (dd, J = 26.7, 5.9 Hz, 3H). MS m/z = 592 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.27 (s, 1H), 7.13-7.04 (m, 2H), 5.53 (s, 1H), 4.42 (s, 1H), 4.30 (s, 2H), 3.87 (s, 2H), 2.37 (t, J = 8.0 Hz, 4H), 2.10 (d, J = 7.0 Hz, 2H), 1.93 (t, J = 8.1 Hz, 2H), 1.86-1.70 (m, 9H), 1.64 (s, 2H), 1.43 (td, J = 13.1, 3.7 Hz, 3H), 1.24 (s, 6H), 1.03 (d, J = 12.7 Hz, 2H), 0.62 (d, J = 87.6 Hz, 4H). MS m/z = 606 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.38 (dd, J = 9.0, 4.5 Hz, 1H), 7.33-7.22 (m, 2H), 4.30 (s, 2H), 3.93 (s, 2H), 2.62 (s, 2H), 2.50-2.28 (m, 6H), 2.19 (d, J = 6.9 Hz, 2H), 1.97 (t, J = 8.1 Hz, 2H), 1.93-1.85 (m, 4H), 1.84- 1.76 (m, 2H), 1.75-1.67 (m, 2H), 1.61-1.48 (m, 3H), 1.17- 1.02 (m, 2H), 0.96-0.55 (m, 8H). MS m/z = 604 [M + H]+
1H NMR (600 MHz, CDCl3) δ 8.39 (s, 1H), 7.80 (s, 1H), 7.03-6.94 (m, 2H), 6.76 (s, 1H), 3.92 (s, 3H), 3.78 (s, 1H), 3.50 (s, 2H), 2.39 (s, 2H), 2.32-2.24 (m, 2H), 2.13 (s, 2H), 1.78 (s, 7H), 1.62 (s, 10H), 1.53 (s, 6H), 1.47 (s, 6H), 1.25 (s, 12H), 1.14 (d, J = 6.5 Hz, 4H), 1.10 (s, 4H), 0.88 (s, 5H) MS m/z = 633 [M + H]+
1H NMR (600 MHz, CDCl3) δ 8.37
1H NMR (600 MHz, CDCl3) δ 8.37
1H NMR (600 MHz, CDCl3) δ 8.38
1H NMR (600 MHz, CDCl3) δ 8.37
1H NMR (400 MHz,
1H NMR (400 MHz,
1H NMR (400 MHz,
1H NMR (400 MHz,
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Methanol-d4)
1H NMR (400 MHz, Chloroform-d) δ 8.37 (d, J = 3.5 Hz, 1H), 7.77 (s, 1H), 7.02 (ddt, J = 7.5, 5.2, 2.8 Hz, 2H), 6.76 (dt, J = 9.8, 5.2 Hz, 1H), 5.45 (s, 1H), 4.06-3.77 (m, 5H), 3.54-3.42 (m, 3H), 3.33 (s, 9H), 2.27 (s, 3H), 2.06 (d, J = 8.4 Hz, 4H), 1.76 (d, J = 17.4 Hz, 8H), 1.67 (s, 3H), 1.42- 1.32 (m, 3H), 1.25 (t, J = 7.0 Hz, 4H), 1.14 (dd, J = 6.7, 3.3 Hz, 5H). MS m/z = 681 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.23 (s, 1H), 7.74 (s, 1H), 7.21- 7.10 (m, 2H), 7.00 (dd, J = 9.0, 4.2 Hz, 1H), 4.11-3.89 (m, 4H), 3.83 (p, J = 6.6 Hz, 1H), 3.61 (h, J = 6.7 Hz, 1H), 3.39 (d, J = 8.8 Hz, 2H), 3.30 (s, 6H), 3.23 (d, J = 8.8 Hz, 2H), 2.38 (s, 4H), 2.17 (d, J = 6.9 Hz, 2H), 2.04 (s, 2H), 1.85-1.72 (m, 8H), 1.53 (d, J = 6.8 Hz, 3H), 1.47 (t, J = 7.9 Hz, 6H), 1.18 (d, J = 6.6 Hz, 3H), 1.08 (dd, J = 16.4, 9.3 Hz, 5H). MS m/z = 695 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.46 (d, J = 2.8 Hz, 1H), 7.25 (d, J = 4.4 Hz, 1H), 7.17-7.08 (m, 1H), 7.02 (ddd, J = 20.2, 7.8, 3.0 Hz, 1H), 5.42 (s, 1H), 4.33 (d, J = 66.0 Hz, 2H), 3.85 (dt, J = 13.2, 6.5 Hz, 3H), 3.46 (d, J = 8.9 Hz, 2H), 3.33 (s, 8H), 3.11 (dd, J = 13.3, 6.8 Hz, 1H), 2.32 (s, 3H), 2.08 (d, J = 18.2 Hz, 4H), 1.88- 1.73 (m, 9H), 1.60 (s, 1H), 1.37 (ddd, J = 15.6, 12.5, 3.4 Hz, 3H), 1.18-1.01 (m, 9H), 0.77 (d, J = 6.6 Hz, 2H). MS m/z = 682 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.26- 7.23 (m, 1H), 7.10 (ddd, J = 9.1, 7.9, 3.0 Hz, 1H), 6.96 (dd, J = 7.8, 3.0 Hz, 1H), 5.75 (s, 1H), 4.47 (d, J = 10.3 Hz, 1H), 4.28 (d, J = 10.3 Hz, 1H), 3.94-3.83 (m, 2H), 3.79 (p, J = 6.6 Hz, 1H), 3.46 (d, J = 8.9 Hz, 2H), 3.43-3.36 (m, 1H), 3.33 (s, 7H), 3.30 (s, 1H), 2.32 (s, 3H), 2.13-2.02 (m, 4H), 1.90-1.64 (m, 9H), 1.50 (d, J = 6.8 Hz, 3H), 1.37 (dd, J = 12.9, 8.7 Hz, 6H), 1.16-0.95 (m, 5H), 0.72 (d, J = 6.6 Hz, 3H). MS m/z = 696 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.26-8.19 (m, 1H), 7.77-7.67 (m, 1H), 7.25-7.13 (m, 2H), 7.06- 6.94 (m, 1H), 4.09-3.82 (m, 5H), 3.52 (dq, J = 14.0, 7.0 Hz, 1H), 3.36 (dt, J = 14.1, 6.9 Hz, 1H), 2.35 (s, 4H), 2.15 (d, J = 6.8 Hz, 2H), 1.90 (s, 2H), 1.85-1.72 (m, 6H), 1.71-1.62 (m, 2H), 1.62- 1.43 (m, 3H), 1.38-1.00 (m, 17H) MS m/z = 621 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.23 (s, 1H), 7.74 (s, 1H), 7.23- 7.09 (m, 2H), 6.99 (dd, J = 9.0, 4.2 Hz, 1H), 4.09-3.88 (m, 4H), 3.83 (p, J = 6.7 Hz, 1H), 3.62 (p, J = 6.8 Hz, 1H), 2.39 (s, 4H), 2.18 (d, J = 6.7 Hz, 2H), 1.90 (s, 2H), 1.87- 1.73 (m, 6H), 1.71-1.62 (m, 2H), 1.62-1.42 (m, 9H), 1.22-1.14 (m, 9H), 1.16-1.00 (m, 5H) MS m/z = 635 [M + H]+
1H NMR (600 MHz, CDCl3) δ 8.38 (s, 1H), 7.80 (s, 1H), 7.00 (s, 2H), 6.73 (d, J = 5.1 Hz, 1H), 5.43 (s, 1H), 4.53 (s, 1H), 3.86 (s, 4H), 2.56 (s, 1H), 2.25 (s, 3H), 2.07 (d, J = 4.8 Hz, 2H), 1.86 (s, 2H), 1.73 (s, 11H), 1.43 (s, 3H), 1.35 (d, J = 6.4 Hz, 6H), 1.21 (s, 7H), 1.01 (d, J = 11.6 Hz, 2H), 0.56 (s, 4H) MS m/z = 633 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.24 (s, 1H), 7.69 (s, 1H), 7.26- 7.15 (m, 2H), 6.96 (dd, J = 8.9, 4.2 Hz, 1H), 3.93 (s, 4H), 2.70 (s, 2H), 2.35 (s, 4H), 2.15 (d, J = 6.9 Hz, 2H), 1.90 (s, 2H), 1.85-1.74 (m, 6H), 1.71-1.62 (m, 2H), 1.61- 1.43 (m, 3H), 1.18 (s, 6H), 1.08 (qd, J = 13.4, 3.5 Hz, 2H), 0.94- 0.87 (m, 2H), 0.83-0.70 (m, 4H), 0.70-0.60 (m, 2H) MS m/z = 631 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.35 (d, J = 9.5 Hz, 1H), 7.75 (d, J = 14.2 Hz, 1H), 6.98 (dd, J = 24.2, 7.9 Hz, 2H), 6.87-6.72 (m, 1H), 5.37 (s, 1H), 3.92 (d, J = 32.6 Hz, 5H), 3.23- 3.02 (m, 1H), 2.28 (s, 3H), 2.13- 2.05 (m, 3H), 1.86 (s, 3H), 1.75 (s, 7H), 1.67 (d, J = 15.8 Hz, 5H), 1.49-1.36 (m,6H), 1.22 (s, 7H), 1.05 (d, J = 37.9 Hz, 5H). MS m/z = 647 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.51-4.14 (m, 2H), 4.02-3.70 (m, 3H), 3.61- 3.39 (m, 1H), 3.28-3.18 (m, 1H), 2.59-2.28 (m, 4H), 2.17 (d, J = 6.8 Hz, 2H), 1.93-1.83 (m, 6H), 1.83-1.75 (m, 2H), 1.71- 1.63 (m, 2H), 1.63-1.44 (m, 3H), 1.25-1.01 (m, 15H), 0.88-0.76 (m, 2H) MS m/z = 622 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.0, 8.1, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.47-4.33 (m, 2H), 3.92 (q, J = 10.7 Hz, 2H), 3.75 (p, J = 6.6 Hz, 1H), 3.54 (p, J = 6.8 Hz, 1H), 2.54-2.29 (m, 4H), 2.20 (d, J = 6.8 Hz, 2H), 1.95-1.74 (m, 8H), 1.72-1.63 (m, 2H), 1.63- 1.43 (m, 6H), 1.39 (d, J = 6.7 Hz, 3H), 1.18 (s, 6H), 1.17-1.01 (m, 5H), 0.75 (d, J = 6.6 Hz, 3H) MS m/z = 636 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.40-8.33 (m, 1H), 7.41(ddd, J = 20.0, 9.1, 4.5 Hz, 1H), 7.28(ddd, J = 9.1, 8.0, 3.0 Hz, 1H), 7.21(dd, J = 8.0, 3.0 Hz, 1H), 4.65(h, J = 6.9 Hz, 1H), 4.4(s, 1H), 4.36-4.27 (m, 1H), 3.95- 3.90 (m, 2H), 2.91-2.67 (m, 3H), 2.61-2.29 (m, 4H), 2.21 (d, J = 6.8 Hz, 2H), 1.94-1.85 (m, 6H), 1.84- 1.74 (m, 2H), 1.73-1.63 (m, 2H), 1.63-1.46 (m, 3H), 1.23- 0.94 (m, 12H), 0.93-0.77 (m, 2H) MS m/z = 608 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.36 (s, 1H), 7.40 (dd, J = 9.0, 4.5 Hz, 1H), 7.34-7.19 (m, 2H), 4.31 (s, 2H), 3.92 (s, 2H), 3.48 (q, J = 7.3 Hz, 2H), 2.74 (tt, J = 7.1, 4.0 Hz, 1H), 2.42 (s, 4H), 2.18 (d, J = 6.9 Hz, 2H), 1.96-1.83 (m, 6H), 1.79 (dd, J = 14.1, 3.5 Hz, 2H), 1.67 (d, J = 12.8 Hz, 2H), 1.62- 1.44 (m, 3H), 1.18 (s, 6H), 1.15- 1.09 (m, 4H), 1.09-1.02 (m, 1H), 0.63 (dd, J = 26.5, 5.8 Hz, 4H). MS m/z = 620 [M + H]+
1H NMR (400 MHz, CDCl3) δ 8.46 (s, 1H), 7.27 (s, 1H), 7.10 (dd, J = 13.9, 5.7 Hz, 2H), 5.36 (s, 1H), 4.36 (d, J = 46.5 Hz, 3H), 3.87 (s, 2H), 2.32 (s, 4H), 2.09 (d, J = 6.9 Hz, 2H), 1.87 (s, 2H), 1.81 (s, 5H), 1.73 (s, 2H), 1.70 (s, 1H), 1.60 (s, 3H), 1.44 (t, J = 11.4 Hz, 3H), 1.22 (s, 8H), 1.09-0.96 (m, 2H), 0.86 (d, J = 11.5 Hz, 1H), 0.70 (s, 2H), 0.56 (s, 2H). MS m/z = 634 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.38 (dd, J = 9.0, 4.5 Hz, 1H), 7.34-7.21 (m, 2H), 4.30 (s, 2H), 3.93 (s, 2H), 2.62 (s, 2H), 2.54-2.29 (m, 4H), 2.17 (d, J = 6.9 Hz, 2H), 1.95-1.84 (m, 6H), 1.84-1.74 (m, 2H), 1.72- 1.62 (m, 2H), 1.63-1.43 (m, 3H), 1.18 (s, 6H), 1.09 (qd, J = 13.8, 3.7 Hz, 2H), 0.92-0.56 (m, 8H) MS m/z = 632 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.44 (s, 1H), 7.24 (s, 1H), 7.17-7.06 (m, 1H), 6.93 (dd, J = 44.6, 7.3 Hz, 1H), 5.42 (s, 1H), 4.60-4.03 (m, 3H), 3.87 (s, 2H), 3.71 (d, J = 8.6 Hz, 1H), 2.32 (s, 3H), 2.07 (d, J = 15.9 Hz, 3H), 1.87 (s, 3H), 1.82 (s,6H), 1.69 (t, J = 15.8 Hz, 6H), 1.54-1.36 (m, 6H), 1.22 (s, 9H), 1.03 (d, J = 12.8 Hz, 3H). MS m/z = 648 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.31-7.23 (m, 1H), 7.24-7.16 (m, 1H), 4.44-4.28 (m, 2H), 3.99- 3.86 (m, 2H), 3.81 (p, J = 6.6 Hz, 1H), 3.56-3.43 (m, 1H), 3.28- 3.17 (m, 1H), 2.55-2.25 (m, 4H), 2.17 (d, J = 6.8 Hz, 2H), 1.91- 1.83 (m, 6H), 1.83-1.75 (m, 2H), 1.71-1.63 (m, 2H), 1.63-1.43 (m, 3H), 1.24-1.02 (m, 9H), 0.81 (d, J = 6.6 Hz, 2H) MS m/z = 628 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.0, 8.0, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.48-4.31 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (hept, J = 6.6 Hz, 1H), 3.54 (hept, J = 6.9 Hz, 1H), 2.60-2.23 (m, 4H), 2.17 (d, J = 6.9 Hz, 2H), 1.93-1.74 (m, 8H), 1.71-1.62 (m, 2H), 1.62-1.46 (m, 6H), 1.39 (d, J = 6.7 Hz, 3H), 1.18-1.01 (m, 5H), 0.74 (d, J = 6.5 Hz, 3H) MS m/z = 642 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.24 (s, 1H), 7.74 (s, 1H), 7.24- 7.14 (m, 2H), 7.00 (td, J = 8.8, 4.2 Hz, 1H), 4.06-3.84 (m, 5H), 3.63- 3.46 (m, 3H), 3.42-3.30 (m, 4H), 2.73 (tdd, J = 9.0, 5.2, 3.6 Hz, 1H), 2.38 (s, 4H), 2.27-2.13 (m, 3H), 1.87-1.64 (m, 9H), 1.64- 1.41 (m, 3H), 1.36-0.98 (m, 11H) MS m/z = 637 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.26 (s, 1H), 7.78 (s, 1H), 7.22- 7.11 (m, 2H), 6.98 (dd, J = 8.9, 4.2 Hz, 1H), 4.03 (s, 2H), 3.97-3.90 (m, 2H), 3.83 (p, J = 6.6 Hz, 1H), 3.67-3.55 (m, 2H), 3.54-3.46 (m, 1H), 3.33 (s, 3H), 2.74 (tdd, J = 8.8, 5.2, 3.5 Hz, 1H), 2.60 (s, 4H), 2.39 (s, 2H), 2.23 (dd, J = 13.0, 9.8 Hz, 1H), 1.95-1.65 (m, 9H), 1.65-1.42 (m, 9H), 1.21- 1.04 (m, 8H) MS m/z = 651 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.43-4.30 (m, 2H), 4.00-3.87 (m, 2H), 3.81 (hept, J = 6.9 Hz, 1H), 3.63-3.45 (m, 3H), 3.33 (s, 3H), 3.28-3.18 (m, 1H), 2.80-2.67 (m, 1H), 2.58- 2.12 (m, 7H), 1.93-1.64 (m, 9H), 1.64-1.41 (m, 3H), 1.26- 0.99 (m, 9H), 0.81 (d, J = 6.6 Hz, 2H) MS m/z = 638 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.43-4.30 (m, 2H), 4.00-3.87 (m, 2H), 3.81 (hept, J = 6.9 Hz, 1H), 3.63-3.45 (m, 3H), 3.33 (s, 3H), 3.28-3.18 (m, 1H), 2.80-2.67 (m, 1H), 2.58- 2.12 (m, 7H), 1.93-1.64 (m, 9H), 1.64-1.41 (m, 3H), 1.26- 0.99 (m, 9H), 0.81 (d, J = 6.6 Hz, 2H) MS m/z = 638 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.1, 8.1, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.48-4.31 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (p, J = 6.6 Hz, 1H), 3.63-3.47 (m, 3H), 2.78-2.69 (m, 1H), 2.58- 2.11 (m, 7H), 1.92-1.64 (m, 9H), 1.63-1.35 (m, 9H), 1.21-1.00 (m, 5H), 0.75 (d, J = 6.6 Hz, 3H) MS m/z = 652 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.1, 8.1, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.48-4.31 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (p, J = 6.6 Hz, 1H), 3.63-3.47 (m, 3H), 3.329 (s, 3H), 2.78-2.69 (m, 1H), 2.58-2.11 (m, 7H), 1.92- 1.64 (m, 9H), 1.63-1.35 (m, 9H), 1.21-1.00 (m, 5H), 0.75 (d, J = 6.6 Hz, 3H). MS m/z = 652 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.37 (s, 1H), 7.76 (s, 1H), 6.98 (dd, J = 7.7, 2.6 Hz, 2H), 6.81-6.73 (m, 1H), 5.41 (s, 1H), 3.96 (s, 2H), 3.87 (d, J = 9.2 Hz, 2H), 3.78 (h, J = 6.6 Hz, 1H), 3.49 (p, J = 6.8 Hz, 1H), 2.55 (ddt, J = 16.4, 9.2, 7.2 Hz, 1H), 2.44- 2.19 (m, 5H), 2.09 (s, 2H), 1.75 (s, 8H), 1.67 (s, 4H), 1.54 (d, J = 6.8 Hz, 4H), 1.48 (d, J = 6.8 Hz, 4H), 1.20 (d, J = 7.1 Hz, 3H), 1.13 (d, J = 6.6 Hz, 3H), 1.09 (d, J = 6.7 Hz, 3H). MS m/z = 621 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.47-4.28 (m, 2H), 4.01-3.86 (m, 2H), 3.81 (p, J = 6.6 Hz, 1H), 3.57-3.41 (m, 1H), 3.28-3.17 (m, 1H), 2.64- 2.23 (m, 6H), 2.17 (d, J = 6.9 Hz, 2H), 1.93-1.69 (m, 7H), 1.71- 1.57 (m, 2H), 1.58-1.36 (m, 3H), 1.24-1.01 (m, 12H), 0.81 (d, J = 6.6 Hz, 2H) MS m/z = 608 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.45-7.36 (m, 1H), 7.33-7.16 (m, 2H), 4.47-4.28 (m, 2H), 4.01-3.86 (m, 2H), 3.81 (p, J = 6.6 Hz, 1H), 3.57-3.41 (m, 1H), 3.28-3.17 (m, 1H), 2.64- 2.23 (m, 6H), 2.17 (d, J = 6.9 Hz, 2H), 1.93-1.69 (m, 7H), 1.71- 1.57 (m, 2H), 1.58-1.36 (m, 3H), 1.24-1.01 (m, 12H), 0.81 (d, J = 6.6 Hz, 2H) MS m/z = 608 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.0, 4.5 Hz, 1H), 7.25 (ddd, J = 9.0, 8.0, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.49-4.29 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (hept, J = 6.6 Hz, 1H), 3.54 (hept, J = 6.9 Hz, 1H), 2.65-2.22 (m, 6H), 2.17 (d, J = 6.9 Hz, 2H), 1.91-1.69 (m, 7H), 1.71-1.58 (m, 2H), 1.58-1.31 (m, 9H), 1.24- 0.99 (m, 8H), 0.75 (d, J = 6.6 Hz, 3H) MS m/z = 622 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.0, 4.5 Hz, 1H), 7.25 (ddd, J = 9.0, 8.0, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.49-4.29 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.75 (hept, J = 6.6 Hz, 1H), 3.54 (hept, J = 6.9 Hz, 1H), 2.65-2.22 (m, 2H), 2.17 (d, J = 6.9 Hz, 2H), 1.91-1.69 (m, 7H), 1.63 (dd, J = 11.9, 3.4 Hz, 2H), 1.58-1.31 (m, 9H), 1.24-0.99 (m, 8H), 0.75 (d, J = 6.6 Hz, 3H) MS m/z = 622 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (d, J = 1.2 Hz, 1H), 7.40 (dt, J = 9.3, 4.7 Hz, 1H), 7.32-7.24 (m, 1H), 7.20 (dd, J = 8.0, 3.0 Hz, 1H), 4.36 (d, J = 13.0 Hz, 2H), 4.02-3.74 (m, 3H), 3.57-3.43 (m, 1H), 3.27-3.06 (m, 1H), 2.61- 2.26 (m, 6H), 2.18 (d, J = 6.9 Hz, 2H), 1.87 (t, J = 5.4 Hz, 4H), 1.83- 1.70 (m, 3H), 1.63 (dd, J = 11.9, 3.5 Hz, 2H), 1.55-1.37 (m, 3H), 1.25-1.01 (m, 9H), 0.81 (d, J = 6.6 Hz, 2H). MS m/z = 611 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 8.38 (s, 1H), 7.39 (dd, J = 9.1, 4.5 Hz, 1H), 7.25 (ddd, J = 9.0, 8.1, 3.0 Hz, 1H), 7.16 (dd, J = 8.0, 3.0 Hz, 1H), 4.49-4.30 (m, 2H), 3.92 (q, J = 10.5 Hz, 2H), 3.76 (h, J = 6.6 Hz, 1H), 3.55 (h, J = 6.8 Hz, 1H), 2.55 (t, J = 9.1 Hz, 1H), 2.40 (dd, J = 12.7, 9.1 Hz, 2H), 2.18 (d, J = 6.8 Hz, 2H), 1.86 (d, J = 5.6 Hz, 8H), 1.63 (dd, J = 11.9, 3.4 Hz, 2H), 1.50 (d, J = 6.8 Hz, 5H), 1.44- 1.35 (m, 5H), 1.20-0.99 (m, 6H), 0.75 (d, J = 6.6 Hz, 3H). MS m/z = 625 [M + H]+
1H NMR (400 MHz, MeOD) δ
1H NMR (400 MHz, MeOD) δ
1H NMR (400 MHz, Chloroform-d) δ 8.40 (s, 1H), 7.18 (d, J = 4.4 Hz, 1H), 7.10-7.02 (m, 1H), 6.99-6.91 (m, 1H), 5.53 (s, 1H), 4.41-4.18 (m, 2H), 3.95- 3.75 (m, 5H), 3.44 (td, J = 11.1, 2.2 Hz, 3H), 3.12-2.98 (m, 1H), 2.26 (s, 3H), 2.12-1.95 (m, 5H), 1.88 (s, 2H), 1.60 (d, J = 12.6 Hz, 3H), 1.41 (t, J = 12.6 Hz, 3H), 1.33 1.24 (m, 3H), 1.21-1.18 (m, 1H), 1.12-0.94 (m, 9H), 0.70 (d, J = 6.6 Hz, 2H) MS m/z = 664 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.39 (d, J = 1.6 Hz, 1H), 7.18 (d, J = 4.5 Hz, 1H), 7.09-7.02 (m, 1H), 6.94 (dd, J = 7.8, 3.1 Hz, 1H), 4.38-4.18 (m, 2H), 4.14 (s, 1H), 3.85-3.74 (m, 3H), 3.44 (s, 1H), 3.12 (s, 2H), 3.08-3.00 (m, 1H), 2.71 (s, 3H), 2.26 (s, 4H), 2.03 (s, 2H), 1.62 (s, 6H), 1.33 (td, J = 11.6, 9.9, 2.6 Hz, 3H), 1.22-1.16 (m, 1H), 1.12- 0.83 (m, 10H), 0.70 (d, J = 6.6 Hz, 2H) MS m/z = 609 [M + H]+
1H NMR (400 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.26- 7.23 (m, 1H), 7.11 (ddd, J = 9.1, 7.9, 3.1 Hz, 1H), 6.96 (dd, J = 7.8, 3.0 Hz, 1H), 5.20 (s, 1H), 4.48 (s, 1H), 4.29 (d, J = 10.4 Hz, 1H), 4.14 (s, 2H), 3.88 (s, 2H), 3.79 (p, J = 6.6 Hz, 1H), 3.39 (p, J = 6.8 Hz, 1H), 2.32 (s, 3H), 2.09 (s, 2H), 1.96-1.74 (m, 7H), 1.67 (s, 2H), 1.44 (s, 6H), 1.38 (d, J = 6.7 Hz, 3H), 1.08 (d, J = 6.6 Hz, 3H), 0.91 (d, J = 14.2 Hz, 2H), 0.71 (d, J = 6.5 Hz, 3H). MS m/z = 610 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.32 (s, 1H), 7.72 (s, 1H), 6.99-6.90 (m, 2H), 6.67 (dd, J = 10.0, 4.4 Hz, 1H), 5.89 (s, 1H), 4.30 (d, J = 8.5 Hz, 1H), 4.02-3.68 (m, 6H), 3.47-3.10 (m, 2H), 2.48-1.97 (m, 5H), 1.79- 1.62 (m, 11H), 1.56-1.35 (m, 4H), 1.18 (t, J = 7.0 Hz, 4H), 1.04 (dt, J = 34.9, 6.7 Hz, 6H) MS m/z = 621 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.31 (q, J = 6.1 Hz, 1H), 7.72 (d, J = 19.8 Hz, 1H), 6.95 (qd, J = 6.3, 5.8, 2.7 Hz, 2H), 6.69 (dd, J = 9.9, 4.3 Hz, 1H), 4.28 (dd, J = 11.7, 8.5 Hz, 1H), 4.01- 3.74 (m, 6H), 3.52-3.40 (m, 1H), 3.29-3.09 (m, 1H), 2.27 (d, J = 72.6 Hz, 5H), 1.75 (d, J = 34.3 Hz, 7H), 1.62 (dd, J = 13.6, 8.2 Hz, 3H), 1.55-1.41 (m, 3H), 1.32- 1.16 (m, 6H), 1.13-1.00 (m, 6H). MS m/z = 621 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.37 (s, 1H), 7.77 (s, 1H), 6.99 (t, J = 8.0 Hz, 2H), 6.78-6.74 (m, 1H), 4.37 (d, J = 8.5 Hz, 1H), 4.07-3.92 (m, 3H), 3.87 (d, J = 9.2 Hz, 2H), 3.79 (p, J = 6.6 Hz, 1H), 3.49 (p, J = 6.8 Hz, 1H), 2.30 (s, 2H), 2.10 (s, 2H), 1.66 (s, 15H), 1.54 (d, J = 6.8 Hz, 4H), 1.48 (d, J = 6.7 Hz, 4H), 1.13 (d, J = 6.7 Hz, 3H), 1.10 (d, J = 6.7 Hz, 3H) MS m/z = 635 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.37 (s, 1H), 7.77 (s, 1H), 6.98 (d, J = 7.9 Hz, 2H), 6.77 (dd, J = 8.8, 4.1 Hz, 1H), 4.34 (d, J = 8.5 Hz, 1H), 4.06-3.92 (m, 3H), 3.89 (d, J = 9.1 Hz, 2H), 3.78 (p, J = 6.6 Hz, 1H), 3.49 (p, J = 6.8 Hz, 1H), 2.38 (s, 4H), 1.91-1.67 (m, 12H), 1.65-1.57 (m, 2H), 1.54 (d, J = 7.0 Hz, 4H), 1.48 (d, J = 6.8 Hz, 4H), 1.36 (ddd, J = 14.6, 7.5, 3.5 Hz, 1H), 1.13 (d, J = 6.7 Hz, 3H), 1.10 (d, J = 6.7 Hz, 3H) MS m/z = 635 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.26- 7.24 (m, 1H), 7.11 (td, J = 8.6, 3.1 Hz, 1H), 6.96 (dd, J = 7.8, 3.1 Hz, 1H), 4.48 (s, 1H), 4.37 (d, J = 8.5 Hz, 1H), 4.29 (s, 1H), 4.02 (d, J = 8.5 Hz, 1H), 3.92-3.84 (m, 2H), 3.79 (p, J = 6.6 Hz, 1H), 3.39 (p, J = 6.7 Hz, 1H), 2.34 (s, 4H), 2.12 (s, 3H), 1.83-1.60 (m, 14H), 1.50 (d, J = 6.8 Hz, 3H), 1.38 (d, J = 6.7 Hz, 3H), 1.08 (d, J = 6.7 Hz, 3H), 0.71 (d, J = 6.6 Hz, 3H). MS m/z = 636 [M + H]+
1H NMR (600 MHz, Chloroform-d) δ 8.46 (s, 1H), 7.25 (d, J = 4.3 Hz, 1H), 7.11 (dt, J = 11.5, 5.8 Hz, 1H), 6.96 (dd, J = 7.8, 3.1 Hz, 1H), 4.50 (s, 1H), 4.33 (d, J = 8.7 Hz, 2H), 4.04 (dd, J = 16.0, 8.4 Hz, 1H), 3.93-3.86 (m, 2H), 3.79 (p, J = 6.7 Hz, 1H), 3.39 (p, J = 6.9 Hz, 1H), 2.23 (s, 7H), 1.92- 1.65 (m, 14H), 1.50 (d, J = 6.7 Hz, 3H), 1.38 (d, J = 6.9 Hz, 3H), 1.08 (d, J = 6.6 Hz, 3H), 0.72 (d, J = 6.5 Hz, 3H). MS m/z = 636 [M + H]+
C6 (132 mg, 0.284 mmol, TF) was dissolved in NMP (3 mL), added with KI (94.79 mg, 0.571 mmol), and stirred at room temperature for 0.5 h. Then M1 (177 mg, 0.38 mmol, Cl) and K2CO3 (525 mg, 3.81 mmol) were added. After replacement with nitrogen, the mixture was heated to 70° C. and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 54 (1.55 mg, 0.002 mmol, 5.9%). 1H NMR (400 MHZ, Chloroform-d) δ 8.40 (d, J=1.5 Hz, 1H), 7.21 (d, J=4.5 Hz, OH), 7.18 (s, OH), 7.06 (ddd, J=7.9, 6.4, 3.1 Hz, 1H), 6.95 (ddd, J=13.5, 7.9, 3.0 Hz, 1H), 5.91 (d, J=1.9 Hz, 1H), 5.84 (s, 1H), 4.46-4.21 (m, 2H), 3.92-3.74 (m, 3H), 3.48 (d, J=6.1 Hz, 3H), 3.09 (d, J=54.9 Hz, 5H), 1.89-1.82 (m, 4H), 1.77 (d, J=14.0 Hz, 2H), 1.62 (dd, J=12.8, 3.6 Hz, 2H), 1.58-1.55 (m, 2H), 1.50 (d, J=18.5 Hz, 2H), 1.26-1.13 (m, 3H), 1.11-0.94 (m, 7H), 0.70 (d, J=6.6 Hz, 2H). MS m/z=608 [M+H]+.
C5 (45 mg, 95.3 μmol) was dissolved in NMP (1 mL), added with KI (19.39 mg, 116.8 μmol), and stirred at room temperature for 0.5 h. Then M2 (44 mg, 77.87 μmol) and K2CO3 (59.51 mg, 431.22 μmol) were added. After replacement with nitrogen, the mixture was heated to 70° C. and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Intermediate-1 (15 mg, 20.55 μmol, yield 26.39%).
Intermediate-1 (15 mg, 20.55 μmol) was dissolved in DCM (0.5 mL), added with TFA (0.5 mL) dropwise, and stirred at room temperature for 1 h. After the reaction was completed, the mixture was concentrated to obtain a crude product which was purified by pre-HPLC to obtain Example 56 (1.16 mg, 1.84 μmol, yield 8.9%). MS m/z=630 [M+H]+.
Referring to the synthesis method of Example 56, the following example compounds were obtained:
1H NMR (400 MHz, CDCl3) δ 8.45
1H NMR (400 MHz, CDCl3) δ 8.46
1H NMR (400 MHz, CDCl3) δ 8.45
1H NMR (400 MHz, CDCl3) δ 8.45
1H NMR (400 MHz, CDCl3) δ 8.46 (s, 1H), 7.27 (s, 1H), 7.25 (d, J = 4.6 Hz, 1H), 7.16-7.08 (m, 1H), 7.00 (dd, J = 7.9, 3.0 Hz, 1H), 5.44 (s, 1H), 4.34 (dd, J = 23.1, 16.7 Hz, 2H), 3.86 (d, J = 6.9 Hz, 3H), 3.51 (s, 1H), 3.10 (s, 1H), 2.50 (s, 2H), 2.32 (s, 3H), 2.24 (s, 3H), 2.11 (s, 2H), 1.79 (d, J = 22.4 Hz, 8H), 1.66 (s, 7H), 1.39 (t, J = 11.5 Hz, 3H), 1.09 (ddd, J = 23.9, 14.0, 7.0 Hz, 9H), 0.76 (d, J = 6.0 Hz, 2H). MS m/z = 634 [M + H]+
1H NMR (400 MHz, CDCl3) δ 8.45 (s, 1H), 7.26-7.22 (m, 1H), 7.13- 7.06 (m, 1H), 6.95 (dd, J = 7.8, 3.0 Hz, 1H), 5.48 (s, 1H), 4.47 (d, J = 10.1 Hz, 1H), 4.28 (d, J = 10.2 Hz, 1H), 3.97-3.71 (m, 3H), 3.49- 3.31 (m, 1H), 2.50 (s, 2H), 2.28 (d, J = 33.5 Hz, 7H), 2.11 (d, J = 5.2 Hz, 2H), 1.76 (s, 10H), 1.67 (d, J = 12.9 Hz, 2H), 1.50 (d, J = 6.7 Hz, 4H), 1.38 (d, J = 6.5 Hz, 6H), 1.07 (d, J = 6.6 Hz, 4H), 0.71 (d, J = 6.5 Hz, 3H). MS m/z = 648 [M + H]+
Example 5 (230 mg, 0.38 mmol), (Boc)2O (124 mg, 0.57 mmol), DMAP (9 mg, 0.076 mmol) and TEA (78 mg, 0.76 mmol) were dissolved in DCM (3 mL) and reacted at 60° C. overnight under nitrogen protection. After the reaction was completed, the mixture was directly concentrated to obtain a crude product which was purified by silica gel column (PE/EA=5:1-3:1, v/v) to obtain Intermediate-2 (190 mg, 0.236 mmol, yield 62.1%). MS m/z=707 [M+H]+.
Intermediate-2 (95 mg, 134.39 μmol) was dissolved in THF (2 mL), cooled to −78° C., slowly added with LiHMDS (0.2 mL, 268.78 μmol) dropwise, and stirred at this temperature for 1 h. Then ethyl trifluoroacetate (81.86 mg, 416.62 μmol) was added, warmed to room temperature and stirred overnight. After the reaction was completed, the system was moved to an ice bath, quenched with saturated aqueous ammonium chloride, and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column to obtain the crude Intermediate-3. MS m/z=803 [M+H]+.
Intermediate-3 (60 mg, 74.73 μmol) and paraformaldehyde (78.53 mg, 2.62 mmol) were dissolved in toluene (2 mL), then added with potassium carbonate (32.02 mg, 231.66 μmol) and 1,8-crown ether-6 (5.93 mg, 22.42 μmol), heated to 110° C. and stirred overnight. After the reaction was completed, the mixture was diluted with water and extracted three times with EA. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain a crude product, which was purified by silica gel column (PE/EA=1:1, v/v) to obtain Intermediate-4 (20 mg, 27.82 μmol, yield 37.23%). MS m/z=719 [M+H]+.
Intermediate-4 (4 mg, 5.48 μmol) was dissolved in DCM (0.5 mL), added with TFA (0.5 mL) dropwise, and stirred at room temperature for 1 h. After the reaction was completed, the mixture was concentrated to obtain a crude product which was purified by pre-HPLC to obtain Example 62 (1.16 mg, 1.84 μmol, yield 33.58%). MS m/z=619 [M+H]+.
Example 20 (70 mg, 100 μmol) was dissolved in anhydrous DCM (1 mL), cooled to −70° C., and BBr3 solution (0.6 mL, 600 μmol) was slowly added dropwise. After the addition was completed, the mixture was naturally heated to room temperature and stirred overnight. After the reaction was completed, the mixture was concentrated to obtain a crude product which was purified by pre-HPLC (10 mM NH4HCO3 aqueous solution/acetonitrile system) to obtain Example 63 (10 mg, 14.9 μmol, yield 14.9%). 1H NMR (400 MHZ, Chloroform-d) δ 8.45 (s, 1H), 7.24 (d, J=4.5 Hz, 1H), 7.10 (ddd, J=9.0, 7.8, 3.0 Hz, 1H), 6.95 (dd, J=7.8, 3.0 Hz, 1H), 5.75 (s, 1H), 4.47 (d, J=10.4 Hz, 1H), 4.28 (d, J=10.3 Hz, 1H), 3.87 (s, 2H), 3.81-3.68 (m, 5H), 3.39 (p, J=6.7 Hz, 1H), 2.33 (s, 3H), 2.10 (d, J=5.6 Hz, 2H), 1.85-1.74 (m, 11H), 1.57-1.31 (m, 10H), 1.08 (d, J=6.5 Hz, 4H), 0.71 (d, J=6.4 Hz, 3H).
Referring to the synthesis method of Example 1, the following example compounds were obtained by reacting raw material 1 and raw material 2:
1H NMR (600 MHz, Methanol-d4) δ 8.20 (s, 1H), 7.69 (s, 1H), 7.57 (d, J = 1.8 Hz, 1H), 7.28 (ddd, J = 9.0, 7.8, 3.1 Hz, 1H), 7.22 (dd, J = 8.4, 3.1 Hz, 1H), 7.08 (dd, J = 9.1, 4.5 Hz, 1H), 6.27 (d, J = 1.9 Hz, 1H), 4.41 (p, J = 6.6 Hz, 1H), 3.77 (s, 4H), 2.35 (dd, J = 8.6, 7.5 Hz, 5H), 2.16 (d, J = 6.9 Hz, 2H), 1.96 (t, J = 8.1 Hz, 2H), 1.82-1.67 (m, 9H), 1.53 (qd, J = 12.3, 11.5, 5.7 Hz, 3H), 1.39 (d, J = 6.6 Hz, 6H), 1.14-1.04 (m, 2H). MS m/z = 636 [M + H]+
1H NMR (400 MHz, Methanol-d4) δ 9.12 (s, 1H), 8.59 (s, 1H), 8.38 (s, 1H), 7.45-7.40 (m, 1H), 7.38 (d, J = 8.9 Hz, 1H), 7.34 (dd, J = 6.1, 2.8 Hz, 1H), 4.43 (s, 2H), 3.95 (s, 2H), 3.14-3.02 (m, 1H), 2.54- 2.31 (m, 6H), 2.19 (d, J = 6.9 Hz, 2H), 1.97 (t, J = 8.1 Hz, 2H), 1.90 (t, J = 5.4 Hz, 4H), 1.85-1.76 (m, 2H), 1.71 (d, J = 12.8 Hz, 2H), 1.54 (td, J = 13.0, 3.8 Hz, 3H), 1.23 (d, J = 6.7 Hz, 6H), 1.17- 1.03 (m, 2H). MS m/z = 601 [M + H]+
The SFC chiral resolution peak retention times and conditions of some of the example compounds in the above table are shown in the table below.
In order to illustrate the absolute configuration of the compounds of the present disclosure, crystals of intermediates A1-a (
The inhibition of Menin/MLL-4-43 peptide interaction by small molecule inhibitors was quantitatively detected by fluorescence polarization competition assay. The experiment was performed in a 384-well plate (Corning, Cat #3575) using a reaction buffer consisting of 50 mM Tris, pH 7.5, 50 mM NaCl, and 1 mM DTT. A 40 μL reaction system included 10 μL of 8 nM Menin recombinant protein and 10 μL of the test compound at different concentrations. The compound was pre-incubated with Menin protein for 15 min, and then 20 μL of 10 nM FITC-MLL4-43-peptide was added. After incubation on a shaker at 25° C. for 60 min, the fluorescence polarization signal (FP 485 520 520) was read using BMG PHERAStar detection. The experimental data was analyzed and processed by GraphPad Prism 6 software to obtain the IC50 value. The control compound 1 was prepared by referring to the method of Example 64 in WO2017214367.
The inhibitory ability of the compounds of the present disclosure on the proliferation of tumor cell lines (including cell lines containing MLL fusion protein such as MV4-11, MOLM-13, THP-1 and NOMO-1, control cell lines without MLL fusion protein such as HL-60, K562 and MOLM-16, and cell lines containing NPM1 mutation such as OCI-AML3) was evaluated by using the cell viability analysis method. The cells were plated in 96-well plates at a certain concentration (e.g., 5,000-20,000 cells/well), and then an equal volume of culture medium containing 2 times the final concentration of the test compound (the final concentration ranging 10 from 1 nM to 10 μM) was added. The plates were placed in an incubator and cultured at 37° C. and 5% CO2 for 72-168 h. Before the test, an equal volume of CellTiter-Glo® Luminescent reagent was added, and the plate was incubated at room temperature for 10 min before detection using an ELISA reader (BMG LABTECH). The data were analyzed using GraphPad Prism software to obtain the IC50 value and compound fitting curve. The control compound 1 was prepared by referring to the method of Example 64 in WO2017214367.
Test purpose: To determine the stability of some of the compounds of the present disclosure in mouse, dog and human liver microsomes by LC-MS/MS method.
Test materials: The test drugs were in-house prepared example compounds of the present disclosure; the positive reference compound SNDX-5613 was prepared by the method of Example 253 in patent WO2017214367; the control compound 1 was prepared by the method of Example 64 in WO2017214367; liver microsomes were purchased from CORNING.
The total volume of each incubation system was approximately 45 μL, and the medium was 100 mM phosphate buffer (PBS, pH 7.4), including liver microsomal protein at a final concentration of 0.5 mg/mL, 1.00 μM compound and 2.00 mM NADPH, and the organic phase content was <1%. Incubation was performed in a 37° C. incubator, and 135 μL of ice-cold acetonitrile was added after 0, 5, 15, 30 and 60 min of reaction to terminate the reaction. The positive control group was incubated with 1.00 μM Ketanserin, 0.5 mg/mL liver microsomal protein and 2.00 mM NADPH in a 37° C. incubator for 0, 5, 15, 30 and 60 min respectively, and then 135 μL of ice-cold acetonitrile was added to terminate the reaction. The 96-well plates were shaken at 600 rpm for 10 min and then centrifuged at 4700 rpm at 4° C. for 15 min. 80 μL of supernatant was mixed with 320 μL of pure water, and the remaining amount of the compound was detected by LC-MS/MS method. The in vitro half life (T½) and intrinsic clearance (CLint) were calculated according to the following formula:
Intrinsic clearance (Clint)=(0.693/T1/2)×(1/liver microsome concentration)×conversion factor. k is the linear regression slope of the remaining percentage of the ln compound-incubation time. The conversion factor is as follows:
The specific test results are shown in Table 3.
Compared with the positive compound SNDX-5613, some example compounds of the present disclosure have better metabolic stability to liver microsomes, and especially have significant advantages in the stability in human liver microsomes.
Test purpose: To determine the pharmacokinetic properties of some of the example compounds of the present disclosure in mice, rats and dogs after single intravenous injection (i.v.) and oral gavage (i.g.) administration by LC-MS/MS method.
Research methods: An appropriate amount of compound was weighed and prepared into a transparent clear solution of a certain concentration using 0.9% sodium chloride injection and 1.5 equivalents of 1M HCl aqueous solution based on the molar number of the compound. After SPF male ICR mice, SPF male SD rats, and male beagle dogs were fasted overnight, the test compound solution was given by intravenous injection and oral gavage at the corresponding dose. Anticoagulated whole blood of the test animals was collected at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, and 24 h after administration, and plasma was separated. Plasma samples at each time point were detected by LC-MS/MS, and the plasma concentration of the compound was determined by the standard curve correction method. Phoenix WinNonlin 5.2 non-compartmental model was used to calculate the pharmacokinetic parameters of terminal elimination half-life (t½), blood peak concentration (Cmax), area under the concentration-time curve (AUC), clearance (CL), and bioavailability (F %), which was directly obtained from the serum concentration results. Blood drug concentration and pharmacokinetic parameters are expressed as mean±standard deviation (X±SD). The specific test protocols for each species are as follows.
Test animals: SPF male ICR mice, weighing 25-30 g, 12 mice/compound, purchased from Chengdu Dossy Experimental Animal Co., Ltd.
Test design: On the day of the test, ICR mice were randomly divided into groups according to body weight, fasted but with free water for 12-14 h one day before administration, and fed 2 h after administration.
Preparation of test compound solution: The concentration of the test compound for oral gavage was 1 mg/mL. An appropriate amount of compound was weighed and prepared into a 1 mg/mL transparent clear solution using 0.9% sodium chloride injection and 1.5 equivalents of 1M HCl aqueous solution. The concentration of the test compound for intravenous injection was 0.2 mg/mL. The 1 mg/mL transparent clear solution was diluted to a concentration of 0.2 mg/mL using 0.9% sodium chloride injection. The volume of 1 M HCl solution added was calculated as: volume of 1 M HCl solution added (mL)=weighed amount of compound (mg)/molecular weight×1.5×1000. The amount added during preparation must not exceed the calculated value.
The test substance was administered as follows: intravenous injection: 1 mg/kg of dose, 5 mL/kg of volume, 6 mice; oral gavage: 10 mg/kg of dose, 10 mL/kg of volume, 6 mice.
Sample collection: Blood (40-50 μl) was collected in anticoagulant tubes sprayed with EDTA-K2 through the orbital venous plexus at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, and 24 h after administration, and centrifuged at 10,000 rpm for 20 min within 1 h (the blood was saved on wet ice before and after centrifugation). The supernatant, which was plasma, was collected and frozen in a refrigerator at −20° C. or below for LC-MS/MS analysis of Full PK of male ICR mice (2 mice per group, 4 blood collection time points per mouse, crossover).
a1 mg/kg for intravenous injection
Compared with the positive compound SNDX-5613, some example compounds of the present disclosure have superior pharmacokinetic properties and significant advantages especially in terms of exposure amount and oral bioavailability.
Test animals: SPF male SD rats, weighing 180-220 g, 6 rats/compound, purchased from Chengdu Dossy Experimental Animal Co., Ltd.
Test design: On the day of the test, SD rats were randomly divided into groups according to body weight, fasted but with free water for 12-14 h one day before administration, and fed 2 h after administration.
Preparation of test compound solution: The concentration of the test compound for oral gavage was 1 mg/mL. An appropriate amount of compound was weighed and prepared into a 1 mg/mL transparent clear solution using 0.9% sodium chloride injection and 1.5 equivalents of 1M HCl aqueous solution. The concentration of the test compound for intravenous injection was 0.5 mg/mL. The 1 mg/mL transparent clear solution was diluted to a concentration of 0.5 mg/ml using 0.9% sodium chloride injection. The volume of 1 M HCl solution added was calculated as: volume of 1 M HCl solution added (mL)=weighed amount of compound (mg)/molecular weight×1.5×1000. The amount added during preparation must not exceed the calculated value.
The test substance was administered as follows: intravenous injection: 1 mg/kg of dose, 2 mL/kg of volume, 3 rats; oral gavage: 10 mg/kg of dose, 10 mL/kg of volume, 3 rats.
Sample collection: Blood (40-50 μl) was collected in anticoagulant tubes sprayed with EDTA-K2 through the orbital venous plexus at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, and 24 h after administration, and centrifuged at 10,000 rpm for 20 min within 1 h (the blood was saved on wet ice before and after centrifugation). The supernatant, which was plasma, was collected and froze in a refrigerator at −20° C. or below for LC-MS/MS analysis of Full PK of male SD rats (1 rat per group, 8 blood collection time points per rat).
Some of the example compounds of the present disclosure have excellent pharmacokinetic properties in rats, especially in terms of exposure amount and oral bioavailability.
Test animals: Male beagle dogs, weighing 8-10 kg, 6 dogs/compound, purchased from Beijing Marshall Biotechnology Co., Ltd.
Test design: On the day of the test, beagle dogs were randomly divided into groups according to their body weight, fasted but with free water for 12-14 h one day before administration, and fed 4 h after administration.
Preparation of test compound solution: The concentration of the test compound for oral gavage was 5 mg/mL. An appropriate amount of compound was weighed and prepared into a 1 mg/mL transparent clear solution using 0.9% sodium chloride injection and 1.5 equivalents of 1M HCl aqueous solution. The concentration of the test compound for intravenous injection was 1 mg/mL. The 5 mg/mL transparent clear solution was diluted to a concentration of 1 mg/ml using 0.9% sodium chloride injection. The volume of 1 M HCl solution added was calculated as: volume of 1 M HCl solution added (mL)=weighed amount of compound (mg)/molecular weight×1.5×1000. The amount added during preparation must not exceed the calculated value.
The test substance was administered as follows: intravenous injection: 1 mg/kg of dose, 1 mL/kg of volume, 3 dogs; oral gavage: 5 mg/kg of dose, 5 mL/kg of volume, 3 dogs.
Sample collection: Blood (500 μl) was collected in anticoagulant tubes sprayed with EDTA-K2 through the orbital venous plexus at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, and 24 h after administration, and centrifuged within 1 hour (3000 g, 4° C., 5 min). The supernatant, which was plasma, was collected and frozen in a refrigerator at −20° C. or below for LC-MS/MS analysis.
Some of the example compounds of the present disclosure have excellent oral bioavailability in beagle dogs.
In this test, human CYP enzyme recombinant protein was incubated with different concentrations (1 and 10 μM) of test compounds and corresponding probe drugs, and the changes in CYP enzyme activity were measured to evaluate the inhibitory ability of the test compounds on each CYP subtype.
The test results show that some of the example compounds of the present disclosure have basically no inhibitory effect on CYP3A4, CYP2C9, CYP2C8, CYP2C19, CYP2D6, and CYP1A2 at a concentration of 10 μM, with an IC50 greater than 10 μM.
The hERG inhibition test was performed on some of the example compounds of the present disclosure using the manual electrophysiological patch clamp method. The cell line was derived from HEK293 cells that overexpressed hERG potassium ion channels. The specific experimental protocol was developed by PharmaCore Labs with reference to literature published in peer-reviewed journals and was performed by PharmaCore Labs in accordance with its standard experimental operating procedures. The highest test concentration was 30 μM. The results are shown in Table 5.
The results show that some of the example compounds of the present disclosure have almost no obvious blocking effect on hERG potassium ion channels within the test concentration range, which is significantly better than the reported IC50 value of the positive reference compound SNDX-5613 of 5-15 μM.
Eight-week-old female BALB/c nude mice were purchased from Zhejiang Vital River Laboratory Animal Technology Co., Ltd. MV-4-11 cells were purchased from Nanjing Cobioer Biotechnology Co., Ltd., and cultured in the culture medium of IMDM plus 10% FBS serum at 37° C. and 5% CO2. MV-4-11 cells were cultured in vitro (suspension cells), and cells in the logarithmic growth phase were collected. The cells were gently rinsed twice with PBS, and the cell pellet was gently blown with PBS to resuspend the cells to prepare a single cell suspension. After counting, the final cell concentration was adjusted to 1×107 cells/100 μL, added with completely liquefied Matrigel matrix gel at a ratio of 1:1, and mixed evenly. Each mouse was inoculated with 1×107 cells in the right axilla, and the inoculation volume was 100 μL/mouse. Tumor growth was observed regularly. On the 8th day after inoculation, when the tumor grew to an average of 131 mm3, the mice were randomly divided and administered according to tumor size and mouse weight (n=8). The example compound was administered by gavage at 15 mg/kg and 30 mg/kg twice a day for 32 consecutive days. A clear solution of the example compound was prepared by mixing with an appropriate amount of 0.9% sodium chloride injection by vortex, and then adding with 1.5 equivalents of 1N HCl hydrochloric acid based on the molar number of the compound to dissolve. During the experiment, animal activity was observed once a day, each animal was weighed once before administration, and the long and short diameters of the tumor were measured with a vernier caliper twice a week. After 32 days of administration, the tumor volume was measured to evaluate the antitumor efficacy of the example compound. At the end of the experiment, all surviving experimental animals were sacrificed. The calculation formula for tumor volume is: V=0.5 (a×b2), where a and b represent the long and short diameters of the tumor, respectively.
The results show that some of the example compounds of the present disclosure show good in vivo efficacy at doses of 15 mg/kg and 30 mg/kg.
In summary, the compounds provided by the present disclosure have excellent inhibitory activity on Menin-MLL protein-protein interaction and cell proliferation, and have more significant advantages in safety and bioavailability. These compounds may become new clinical drugs for the treatment of cancer or other diseases mediated by Menin-MLL interactions
Number | Date | Country | Kind |
---|---|---|---|
202111650341.X | Dec 2021 | CN | national |
202210948453.1 | Aug 2022 | CN | national |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2022/143908 | 12/30/2022 | WO |