SULFUR/PHOSPHORUS-CONTAINING ARYL COMPOUND AND APPLICATION THEREOF

Abstract
Provided are a sulfur/phosphorus-containing aryl compound and an application thereof, and specifically disclosed are a compound represented by formula (V) and a pharmaceutically acceptable salt thereof.
Description

The application claims the right of the following priorities: CN202110801875.1, application date: Jul. 15, 2021; CN202110875486.3, application date: Jul. 30, 2021; CN202111162655.5, application date: Sep. 30, 2021; CN202111273381.7, application date: Oct. 29, 2021; CN202210039225.2, application date: Jan. 13, 2022; CN202210260637.9, application date: Mar. 16, 2022; CN202210731484.1, application date: Jun. 24, 2022.


TECHNICAL FIELD

The present disclosure relates to a new sulfur/phosphorus-containing aryl compound and a use thereof, and specifically relates to a compound of formula (V) and a pharmaceutically acceptable salt thereof.


BACKGROUND

JAK is a category of non-receptor tyrosine kinases, comprising four subtypes: JAK1, JAK2, JAK3, and TYK2. The JAK-STAT signaling pathway mediated by them is related to cell proliferation, differentiation, apoptosis, and immune regulation. The JAK-STAT pathway is essential for immune responses, and during inflammation, the overactivation of JAK conversely promotes the progression of the disease. As research into the mechanism of the JAK-STAT pathway in various autoimmune diseases deepens, JAK inhibitors, as the latest type of targeted autoimmune drugs, have gradually received marketing approval for rheumatoid arthritis (RA), psoriatic arthritis (PsA), and atopic dermatitis, with more indications in late-stage clinical development, including ankylosing spondylitis (AS), ulcerative colitis (UC), Crohn's disease, and others. Research has revealed that TYK2 mediates the signaling of IL-6, IL-10, IL-12, IL-23, and type I interferon, encompassing the key cytokines IL-12 and IL-23, which are currently considered critical in the progression of psoriasis. Consequently, TYK2 inhibitors are considered to be important targets for treating a substantial population with another autoimmune disease—psoriasis. However, as of now, no TYK2 inhibitors have been approved.


BMS-986165, a selective allosteric TYK2 inhibitor, is the most advanced candidate compound in this field. Its phase III trials have demonstrated clinical efficacy comparable to first-line biologics, significantly superior to the oral standard therapy apremilast (a PDE4 inhibitor). Moreover, it has a low incidence of adverse reactions, with a discontinuation rate due to adverse events lower than that of both the apremilast group and the placebo group. The mechanism of action of BMS-986165 is unique. Unlike other JAK inhibitors, it targets the JH2 pseudokinase domain of TYK2. However, similar to orthosteric inhibitors, it suppresses the kinase activity of TYK2, thereby exerting its effect. Notably, it achieves high kinase selectivity (>1000-fold). This demonstrates the substantial clinical application potential of high-selectivity TYK2 inhibitors in the treatment of psoriasis and other targets, signifying considerable clinical application value.




embedded image


CONTENT OF THE PRESENT INVENTION

The present disclosure provides a compound of formula (V) or a pharmaceutically acceptable salt thereof,




embedded image




    • wherein

    • L is selected from —NH—, —NHC(═O)—, —NHC(═O)O—, and —NHC(═O)NH—;

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from H and C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from phenyl and 6-membered heteroaryl;

    • R3 is selected from —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-4 alkyl, —S(═O)nC1-3 alkylamino, —S(═O)n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-4 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,







embedded image


and the —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-4 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-4 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,




embedded image


are each independently and optionally substituted by 1, 2, or 3 halogens;

    • R5 is selected from C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl, and the C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl are each independently and optionally substituted by 1, 2, or 3 Rc;
    • R4 and R6 are each independently selected from H, F, Cl, Br, and I;
    • alternatively, R3, R4 together with the carbon atom to which they are attached form




embedded image




    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • Re is selected from F, Cl, Br, I, and C1-3 alkyl;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2;

    • provided that

    • when T is N, R3 is not —S(═O)nC1-4 alkyl;

    • “hetero” in the 4- to 5-membered heterocycloalkyl and 5- to 6-membered heteroaryl represents 1, 2, or 3 heteroatoms or heteroatom groups independently selected from —O—, —NH—, —S—, and —N—.





The present disclosure provides a compound of formula (V) or a pharmaceutically acceptable salt thereof,




embedded image




    • wherein

    • L is selected from —NH—, —NHC(═O)—, —NHC(═O)O—, and —NHC(═O)NH—;

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from H and C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from 5- to 6-membered heteroaryl;

    • R3 is selected from —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)˜ C1-4 alkyl, —S(═O)nC1-3 alkylamino, —S(═O)n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-4 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,







embedded image


and the —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-4 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-4 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,




embedded image


are each independently and optionally substituted by 1, 2, or 3 halogens;

    • R5 is selected from C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl, and the C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl are each independently and optionally substituted by 1, 2, or 3 Re;
    • R4 and R6 are each independently selected from H, F, Cl, Br, and I;
    • alternatively, R3, R4 together with the carbon atom to which they are attached form




embedded image




    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • Re is selected from F, Cl, Br, I, and C1-3 alkyl;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2;

    • provided that

    • when T is N, R3 is not —S(═O)nC1-4 alkyl;

    • “hetero” in the 4- to 5-membered heterocycloalkyl and 5- to 6-membered heteroaryl represents 1, 2, or 3 heteroatoms or heteroatom groups independently selected from —O—, —NH—, —S—, and —N—.





In some embodiments of the present disclosure, the L is selected from —NH— and —NHC(═O)—, and other variables are as defined in the present disclosure. In some embodiments of the present disclosure, the R1 is selected from OCH3, and the OCH3 is optionally substituted by 1, 2, or 3 Ra, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R1 is selected from OCH3 and OCF3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R1 is selected from OCH3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H and CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H, CH3, and CD3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R3 is selected from




embedded image


and other variables are as defined in the present disclosure. In some embodiments of the present disclosure, the ring C is selected from phenyl, pyridyl, pyrimidinyl, pyridazinyl, and pyrazinyl, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the Re is selected from F and CH3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R5 is selected from CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl, and the CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl are each independently and optionally substituted by 1, 2, or 3 Re, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R5 is selected from CH3,




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the compound or the pharmaceutically acceptable salt thereof is provided, and the compound is selected from:




embedded image




    • wherein

    • L is selected from —NH—, —NHC(═O)—, —NHC(═O)O—, and —NHC(═O)NH—;

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from H and C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from phenyl, pyridyl, pyrimidinyl, pyridazinyl, and pyrazinyl;

    • R3 is selected from —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-3 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-3 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,







embedded image


and the —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-3 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocycloalkyl, —S(═O))nNH2, —S(═O)(═NR)C1-3 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,




embedded image


are each independently and optionally substituted by 1, 2, or 3 halogens;

    • R5 is selected from C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl, and the C1-3 alkyl, C3-5 cycloalkyl, and 5- to 6-membered heteroaryl are each independently and optionally substituted by 1, 2, or 3 Re;
    • R4 and R6 are each independently selected from H, F, Cl, and Br;
    • alternatively, R3, R4 together with the carbon atom to which they are attached form




embedded image




    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • Re is selected from F, Cl, Br, I, and C1-3 alkyl;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2;

    • “hetero” in the 4- to 5-membered heterocycloalkyl and 5- to 6-membered heteroaryl represents 1, 2, or 3 heteroatoms or heteroatom groups independently selected from —O—, —NH—, —S—, and —N—.





In some embodiments of the present disclosure, the R1 is selected from OCH3, and the OCH3 is optionally substituted by 1, 2, or 3 Ra, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R1 is selected from OCH3 and OCF3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H and CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H, CH3, and CD3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R3 is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R3 is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R5 is selected from CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl, and the CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl are each independently and optionally substituted by 1, 2, or 3 Re, and other variables are as defined in the present disclosure. In some embodiments of the present disclosure, the R5 is selected from CH3,




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the compound or the pharmaceutically acceptable salt thereof is provided, and the compound is selected from




embedded image




    • wherein

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from H and C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from phenyl, pyridyl, and pyrimidinyl;

    • R3 is selected from —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-3 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocyclyl, —S(═O))nNH2, —S(═O)(═NR)C1-3 alkyl, —S(═O)(═NR)C1-3 alkylamino, —S(═O)(═NR)C3-5 cycloalkyl,







embedded image




    • R4 is selected from H, F, Cl, and Br;

    • alternatively, R3, R4 together with the carbon atom to which they are attached form







embedded image




    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2;

    • “hetero” in the 4- to 5-membered heterocyclyl represents 1, 2, or 3 heteroatoms or heteroatom groups independently selected from —O—, —NH—, —S—, and —N—.





In some embodiments of the present disclosure, the compound or the pharmaceutically acceptable salt thereof is provided, and the compound is selected from




embedded image




    • wherein

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from phenyl, pyridyl, and pyrimidinyl;

    • R3 is selected from —P(═O)(C1-3 alkyl)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)nC1-3 alkyl, —S(═O)nC1-3 alkylamino, —S(═O))n-4- to 5-membered heterocyclyl, —S(═O))nNH2, —S(═O)(═NR)C1-3 alkyl, —S(═O)(═NR)C1-3 alkylamino, and —S(═O)(═NR)C3-5 cycloalkyl;

    • R4 is selected from H, F, Cl, and Br;

    • alternatively, R3, R4 together with the carbon atom to which they are attached form







embedded image




    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2.





In some embodiments of the present disclosure, the R1 is selected from OCH3, and the OCH3 is optionally substituted by 1, 2, or 3 Ra, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R1 is selected from OCH3 and OCF3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H and CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from H, CH3, and CD3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from CH3 and CD3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R3 is independently selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R3 is independently selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


embedded image


embedded image


and other variables are as defined in the present disclosure.


In some embodiment of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


The present disclosure provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,




embedded image




    • wherein

    • T, T1, and T2 are each independently selected from N and CH, and the CH is optionally substituted by 1 halogen;

    • R1 is selected from C1-3 alkoxy, and the C1-3 alkoxy is optionally substituted by 1, 2, or 3 Ra;

    • R2 is selected from C1-3 alkyl, and the C1-3 alkyl is optionally substituted by 1, 2, or 3 Rb;

    • ring C is selected from phenyl, pyridine, pyrimidine, and 2,3-dihydrobenzo[b]thiophene 1,1-dioxide, and the phenyl, pyridine, pyrimidine, and 2,3-dihydrobenzo[b]thiophene 1,1-dioxide are each independently and optionally substituted by 1, 2, or 3 Re;

    • Ra and Rb are each independently selected from H, D, F, Cl, Br, and I;

    • each Re is independently selected from —P(═O)(CH3)2, —P(═O)(C3-5 cycloalkyl)2, —S(═O)1C1-3 alkyl, —S(═O)1C1-3 alkylamino, —S(═O)n-5-membered heterocyclyl, —S(═O))nNH2, and —S(═O)(═NR)C1-3 alkyl;

    • R is selected from H and C1-3 alkyl;

    • n is 1 or 2.





In some embodiments of the present disclosure, the R1 is selected from OCH3, and the OCH3 is optionally substituted by 1, 2, or 3 Ra, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R1 is selected from OCH3 and OCF3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the R2 is selected from CH3 and CD3, and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the ring C is selected from




embedded image


and other variables are as defined in the present disclosure.


In some embodiments of the present disclosure, the ring C is selected from




embedded image


and other variables are as defined in the present disclosure. In some embodiments of the present disclosure, the structural moiety




embedded image


is selected from




embedded image


and other variables are as defined in the present disclosure.


There are still some embodiments of the present disclosure which are obtained by any combination of the above variables.


In some embodiments of the present disclosure, the compound or the pharmaceutically acceptable salt thereof is selected from:




embedded image


embedded image




    • wherein

    • R1, R2, R3, R4 and T are as defined in the present disclosure.





The present disclosure provides a compound of the following formula or a pharmaceutically acceptable salt thereof,




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In some embodiments of the present disclosure, the compound or the pharmaceutically acceptable salt thereof is selected from:




embedded image


embedded image


Technical Effect

The compound of the present disclosure acts on the JH2 pseudokinase domain of TYK2 and exhibits a good inhibitory effect on TYK2 kinase. The compound of the present disclosure shows high inhibitory activity against cell proliferation in the Ba/F3-FL-TYK2-E957D cell line, which has a point mutation in the TYK2 gene. In human PBMC cells, the compound of the present disclosure demonstrates strong inhibitory activity against the TYK2 signaling pathway activated by IFN-α stimulation, and also exhibits weak inhibitory activity against the JAK1/2 signaling pathway activated by IL-6 stimulation, the JAK2/2 signaling pathway activated by GM-CSF stimulation, and the JAK1/3 signaling pathway activated by IL-2 stimulation, thereby showing high selectivity. The compound of the present disclosure possesses excellent pharmacokinetic properties. The compound of the present disclosure has a significant dose-dependent inhibitory effect on the release of IFNγ in mice induced by IL-12/IL-18. Additionally, the compound of the present disclosure has a significant alleviating effect on colitis and psoriasis.


Definition and Description

Unless otherwise specified, the following terms and phrases when used herein have the following meanings. A specific term or phrase should not be considered indefinite or unclear in the absence of a particular definition, but should be understood in the ordinary sense. When a trading name appears herein, it is intended to refer to its corresponding commodity or active ingredient thereof.


The term “pharmaceutically acceptable” is used herein in terms of those compounds, materials, compositions, and/or dosage forms, which are suitable for use in contact with human and animal tissues within the scope of reliable medical judgment, with no excessive toxicity, irritation, an allergic reaction, or other problems or complications, commensurate with a reasonable benefit/risk ratio.


The term “pharmaceutically acceptable salt” refers to a salt of the compound of the present disclosure that is prepared by reacting the compound having a specific substituent of the present disclosure with a relatively non-toxic acid or base. When the compound of the present disclosure contains a relatively acidic functional group, a base addition salt can be obtained by contacting the compound with a sufficient amount of base in a pure solution or a suitable inert solvent. When the compound of the present disclosure contains a relatively basic functional group, an acid addition salt can be obtained by contacting the compound with a sufficient amount of acid in a pure solution or a suitable inert solvent. Certain specific compounds of the present disclosure contain both basic and acidic functional groups, thus can be converted to any base or acid addition salt.


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


Unless otherwise specified, the term “isomer” is intended to include a geometric isomer, a cis-trans isomer, a stereoisomer, an enantiomer, an optical isomer, a diastereoisomer, and a tautomeric isomer.


The compounds of the present disclosure may exist in specific geometric or stereoisomeric forms. The present disclosure contemplates all such compounds, including cis and trans isomers, (−)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereoisomers, (D)-isomers, (L)-isomers, racemic mixtures, and other mixtures thereof, such as enantiomers or diastereomer enriched mixtures, all of which are within the scope of the present disclosure. Additional asymmetric carbon atoms may be present in substituents such as alkyl. All these isomers and their mixtures are included within the scope of the present disclosure.


Unless otherwise specified, the term “enantiomer” or “optical isomer” refers to stereoisomers that are mirror images of each other.


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


Unless otherwise specified, the term “diastereomer” refers to a stereoisomer in which a molecule has two or more chiral centers and the relationship between the molecules is not mirror images.


Unless otherwise specified, “(+)” refers to dextrorotation, “(−)” refers to levorotation, and “(±)” refers to racemic.


Unless otherwise specified, the absolute configuration of a stereogenic center is represented by a wedged solid bond (custom-character) and a wedged dashed bond (custom-character), and the relative configuration of a stereogenic center is represented by a straight solid bond (custom-character) and a straight dashed bond (custom-character), a wave line (custom-character) is used to represent a wedged solid bond (custom-character) or a wedged dashed bond (custom-character), or the wave line (custom-character) is used to represent a straight solid bond (custom-character) or a straight dashed bond (custom-character).


Unless otherwise specified, the terms “enriched in one isomer”, “enriched in isomers”, “enriched in one enantiomer”, or “enriched in enantiomers” refer to the content of one of the isomers or enantiomers is less than 100%, and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.


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


Optically active (R)- and (S)-isomers, or D and L isomers can be prepared using chiral synthesis, chiral reagents, or other conventional techniques. If one kind of enantiomer of certain compound of the present disclosure is to be obtained, it can be obtained by asymmetric synthesis or derivative action of chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (such as amino) or an acidic functional group (such as carboxyl), a salt of a diastereoisomer is formed with an appropriate optically active acid or base, and then diastereomeric resolution is performed by conventional methods known in the art, and then the pure enantiomer is recovered. In addition, the enantiomer and the diastereoisomer are generally separated through chromatography which uses a chiral stationary phase and optionally combines with a chemical derivative method (such as carbamate generated from amine).


The compound of the present disclosure may contain an unnatural proportion of atomic isotopes at one or more than one atom that constitutes the compound. For example, the compound can be radiolabeled with a radioactive isotope, such as tritium (3H), iodine-125 (125), or C-14 (14C). For another example, deuterated drugs can be formed by replacing hydrogen with deuterium, the bond formed by deuterium and carbon is stronger than that of ordinary hydrogen and carbon, compared with non-deuterated drugs, deuterated drugs have the advantages of reduced toxic and side effects, increased drug stability, enhanced efficacy, extended biological half-life of drugs, etc. All isotopic variations of the compound of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.


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


The term “substituted” means one or more than one hydrogen atom on a specific atom is substituted by the substituent, including deuterium and hydrogen variables, as long as the valence of the specific atom is normal and the substituted compound is stable. When the substituent is an oxygen (i.e., ═O), it means two hydrogen atoms are substituted. Positions on an aromatic ring cannot be substituted with a ketone.


The term “optionally substituted” means an atom can be substituted by a substituent or not, unless otherwise specified, the type and number of the substituent may be arbitrary as long as being chemically achievable.


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


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


When a substituent is 0 in number, it means that the substituent is absent. In the case of -A-(R)0, the structure is actually -A.


When a substituent is vacant, it means that the substituent is absent, for example, when X is vacant in A-X, the structure of A-X is actually A.


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


When the bond of a substituent can be cross-linked to two or more atoms on a ring, this substituent can be bonded to any atom on this ring. For example, the structural moiety




embedded image


indicates that the substituent R can be substituted at any position on cyclohexyl or cyclohexadiene. When the enumerative substituent does not indicate by which atom it is linked to the group to be substituted, such substituent can be bonded by any atom thereof. For example, when pyridyl acts as a substituent, it can be linked to the group to be substituted by any carbon atom on the pyridine ring.


When the enumerative linking group does not indicate the direction for linking, the direction for linking is arbitrary, for example, the linking group L contained in




embedded image


is -M-W—, then -M-W— can link ring A and ring B to form




embedded image


in the direction same as left-to-right reading order, and form




embedded image


in the direction contrary to left-to-right reading order. A combination of the linking groups, substituents and/or variables thereof is allowed only when such combination can result in a stable compound.


Unless otherwise specified, when a group has one or more than one linkable site, any one or more than one site of the group can be linked to other groups through chemical bonds. When the linking site of the chemical bond is not positioned, and there is an H atom at the linkable site, then the number of H atoms at the site will decrease correspondingly with the number of the chemical bonds linking thereto so as to meet the corresponding valence. The chemical bond between the site and other groups can be represented by a straight solid bond (custom-character), a straight dashed bond (custom-character), or a wavy line




embedded image


For example, the straight solid bond in —OCH3 means that it is linked to other groups through the oxygen atom in the group; the straight dashed bond in




embedded image


means that it is linked to other groups through the two ends of the nitrogen atom in the group; the wave line in




embedded image


means that the phenyl group is linked to other groups through carbon atoms at position 1 and position 2;




embedded image


means that it can be linked to other groups through any linkable sites on the piperidinyl by one chemical bond, including at least four types of linkage, including




embedded image


Even though the H atom is drawn on the —N—,




embedded image


still includes the linkage of




embedded image


merely when one chemical bond was connected, the H of this site will be reduced by one to the corresponding monovalent piperidinyl.


Unless otherwise specified, the number of atoms in a ring is usually defined as the number of ring members, for example, “5- to 7-membered ring” refers to a “ring” in which 5 to 7 atoms are arranged around.


Unless otherwise specified, the term “C1-4 alkyl” refers to a linear or branched saturated hydrocarbon group consisting of 1 to 4 carbon atoms. The C1-4 alkyl includes C1-2, C1-3, C2-3 alkyl, etc.; it can be monovalent (such as methyl), divalent (such as methylene), or multivalent (such as methine). Examples of C1-4 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, s-butyl, and t-butyl), etc.


Unless otherwise specified, the term “C1-3 alkyl” refers to a linear or branched saturated hydrocarbon group consisting of 1 to 3 carbon atoms. The C1-3 alkyl includes C1-2, C2-3 alkyl, etc.; it can be monovalent (such as methyl), divalent (such as methylene), or multivalent (such as methine). Examples of C1-3 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), etc.


Unless otherwise specified, the term “C1-3 alkoxy” refers to an alkyl group containing 1 to 3 carbon atoms that are connected to the rest of the molecule through an oxygen atom. The C1-3 alkoxy includes C1-2, C2-3, C3, C2 alkoxy, etc. Examples of C1-3 alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), etc.


Unless otherwise specified, the term “C1-3 alkylamino” refers to an alkyl group containing 1 to 3 carbon atoms that are connected to the rest of the molecule through an amino group. The C1-3 alkylamino includes C1-2, C3, C2 alkylamino, etc. Examples of C1-3 alkylamino include, but are not limited to, —NHCH3, —N(CH3)2, —NHCH2CH3, —N(CH3)CH2CH3, —NHCH2CH2CH3, —NHCH2(CH3)2, etc.


Unless otherwise specified, “C3-5 cycloalkyl” refers to a saturated cyclic hydrocarbon group consisting of 3 to 5 carbon atoms, which is a monocyclic system, and the C3-5 cycloalkyl includes C3-4, C4-5 cycloalkyl, etc.; it can be monovalent, divalent, or multivalent. Examples of C3-5 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, etc.


Unless otherwise specified, the term “4- to 5-membered heterocycloalkyl” by itself or in combination with other terms refers to a saturated monocyclic group consisting of 4 to 5 ring atoms, wherein 1, 2, 3, or 4 ring atoms are heteroatoms independently selected from O, S, and N, and the rest are carbon atoms, wherein nitrogen atoms are optionally quaternized, and nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2). In addition, with regard to the “4- to 5-membered heterocycloalkyl”, a heteroatom may occupy the connection position of the heterocycloalkyl with the rest of the molecule. The 4- to 5-membered heterocycloalkyl includes 4-membered and 5-membered heterocycloalkyl. Examples of 4- to 5-membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothiophenyl (including tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, etc.), tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), etc.


Unless otherwise specified, the terms “5- to 6-membered heteroaryl ring” and “5- to 6-membered heteroaryl” in the present disclosure can be used interchangeably, and the term “5- to 6-membered heteroaryl” refers to a monocyclic group consisting of 5 to 6 ring atoms with a conjugated π-electron system, wherein 1, 2, 3, or 4 ring atoms are heteroatoms independently selected from O, S, and N, and the rest are carbon atoms, wherein nitrogen atoms are optionally quaternized, and nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2). The 5- to 6-membered heteroaryl can be linked to the rest of the molecule through a heteroatom or a carbon atom. The 5- to 6-membered heteroaryl includes 5-membered and 6-membered heteroaryl. Examples of the 5- to 6-membered heteroaryl include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl, 3-pyrazolyl, etc.), imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, etc.), triazolyl (including 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, 4H-1,2,4-triazolyl, etc.), tetrazolyl, isoxazolyl (including 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, etc.), thiazolyl (including 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, etc.), furyl (including 2-furyl, 3-furyl, etc.), thienyl (including 2-thienyl, 3-thienyl, etc.), pyridyl (including 2-pyridyl, 3-pyridyl, 4-pyridyl, etc.), pyrazinyl, or pyrimidinyl (including 2-pyrimidinyl, 4-pyrimidinyl, etc.).


The structure of the compounds of the present disclosure can be confirmed by conventional methods known to those skilled in the art, and if the present disclosure involves an absolute configuration of a compound, then the absolute configuration can be confirmed by means of conventional techniques in the art. For example, in the case of single crystal X-ray diffraction (SXRD), diffraction intensity data are collected from the cultured single crystal using a Bruker D8 venture diffractometer with CuKα radiation as the light source and scanning mode: φ/ω scan, and after collecting the relevant data, the crystal structure is further analyzed by direct method (Shelxs97), so that the absolute configuration can be confirmed.


The compounds of the present disclosure can be prepared by a variety of synthetic methods known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by their combination with other chemical synthesis methods, and equivalent alternatives known to those skilled in the art, with preferred embodiments including, but not limited to, the examples of the present disclosure.


The solvent used in the present disclosure is commercially available.


In the present disclosure, the following abbreviations are used: aq stands for water; eq stands for equivalent; DCM stands for dichloromethane; PE stands for petroleum ether; DMSO stands for dimethyl sulfoxide; EtOAc stands for ethyl acetate; EtOH stands for ethanol; MeOH stands for methanol; DMF stands for N,N-dimethylformamide; Cbz stands for benzyloxycarbonyl, which is an amine protecting group; Boc stands for tert-butoxycarbonyl, which is an amine protecting group; r.t. stands for room temperature; O/N stands for overnight; THE stands for tetrahydrofuran; Boc2O stands for di-tert-butyl dicarbonate; TFA stands for trifluoroacetic acid; HCl stands for hydrochloric acid; mp stands for melting point; Pd(dppf)Cl2 stands for [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II); Pd(dppf)Cl2·CH2Cl2 stands for [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane complex; TEA stands for triethylamine; Xantphos stands for 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene; Pd2(dba)3 stands for tris(dibenzylideneacetone)dipalladium(0); EDCI stands for 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride; HOBt stands for 1-hydroxybenzotriazole; NMP stands for N-methyl-2-pyrrolidone; DIPEA stands for N,N-diisopropylethylamine; LiHMDS stands for lithium bis(trimethylsilyl)amide; Pd2(dba)3·CHCl3 stands for tris(dibenzylideneacetone)dipalladium(0)-chloroform adduct.


The compounds of the present disclosure are named according to the conventional naming principles in the art or by ChemDraw® software, and the commercially available compounds use the supplier catalog names.







DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT

The present disclosure is described in detail by the examples below, but it does not mean that there are any adverse restrictions on the present disclosure. The present disclosure has been described in detail herein, and its specific examples have also been disclosed; for one skilled in the art, it is obvious to make various modifications and improvements to the examples of the present disclosure without departing from the spirit and scope of the present disclosure.


Reference Example 1



embedded image


Step 1: Synthesis of Compound A-1

Compound A-1-1 (10 g, 49.49 mmol, 1 eq), bis(pinacolato)diboron (18.85 g, 74.24 mmol, 1.5 eq), and potassium acetate (14.57 g, 148.48 mmol, 3 eq) were dissolved in 1,4-dioxane (200 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (2.02 g, 2.47 mmol, 0.05 eq) was added, and the mixture was stirred at 100° C. for 2 hours. Water (200 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (100 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (V/V, ethyl acetate/petroleum ether=0 to 25%) to obtain compound A-1. 1HNMR (400 MHz, CDCl3) δ: 7.13-7.11 (m, 1H), 6.93 (t, J=15.2, 7.6 Hz, 1H), 6.88-6.85 (m, 1H), 3.81 (s, 3H), 1.36 (s, 12H); LCMS m/z=250.1 [M+H]+.


Reference Example 2



embedded image


Step 1: Synthesis of Compound A-2-2

Compound A-2-1 (18.5 g, 96.35 mmol, 1 eq) was dissolved in DCM (200 mL). Oxalyl chloride (15.90 g, 125.26 mmol, 10.96 mL, 1.3 eq) was then added to the mixture, and DMF (352.15 mg, 4.82 mmol, 370.68 μL, 0.05 eq) was added thereto. The reaction was carried out at 25° C. for 16 hours. The reaction system was then concentrated, added with dichloromethane (50 mL), concentrated again, and directly used in the next step to obtain compound A-2-2.


Step 2: Synthesis of Compound A-2

Compound A-2-2 (20 g, 95.04 mmol, 1 eq) was added to DCM (300 mL). Deuterated methylamine hydrochloride (5.36 g, 76.03 mmol, 0.8 eq) was then added. After cooling the mixture to 0° C., DIPEA (36.85 g, 285.11 mmol, 49.66 mL, 3 eq) was added, and the reaction was carried out at 20° C. for 16 hours. After the reaction was completed, a saturated ammonium chloride aqueous solution (100 mL) was added to the system for extraction and phase separation. The aqueous phase was then extracted once with dichloromethane (60 mL). The organic phases were combined, washed once with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain a crude product. tert-Butyl methyl ether (100 mL) was added to the crude product. The mixture was stirred for 2 hours, filtered, and the filter cake was concentrated under reduced pressure to obtain compound A-2. 1H NMR (400 MHz, DMSO-d6) δ: 8.59 (s, 1H), 8.48 (s, 1H), 7.91 (s, 1H); LCMS m/z=208.1 [M+1]+.


Reference Example 3



embedded image


Step 1: Synthesis of Compound A-3-2

Compound A-3-1 (20 g, 96.61 mmol, 1 eq) was dissolved in acetonitrile (100 mL) and water (15 mL). Lithium bromide (25.17 g, 289.84 mmol, 7.28 mL, 3 eq) and DIPEA (37.46 g, 289.84 mmol, 50.48 mL, 3 eq) were added thereto. The mixture was stirred and reacted at 20° C. for 3 hours. The system was filtered, and the filter cake was rinsed with acetonitrile (50 mL). The filter cake was collected to obtain compound A-3-2. LCMS m/z=192.9 [COOH+1]+.


Step 2: Synthesis of Compound A-3

Compound A-3-2 (10 g, 50.27 mmol, 1 eq) was dissolved in DCM (150 mL). Oxalyl chloride (8.93 g, 70.38 mmol, 6.16 mL, 1.4 eq) was then added to the mixture, and DMF (183.72 mg, 2.51 mmol, 193.39 μL, 0.05 eq) was added thereto. The reaction was carried out at 20° C. for 4 hours. The reaction system was concentrated, then dichloromethane (30 mL) was added, and the mixture was further concentrated. The concentrated mixture was added to dichloromethane (150 mL), and deuterated methylamine hydrochloride (3.37 g, 47.76 mmol, 0.95 eq) was added. After cooling to 0° C., DIPEA (19.49 g, 150.83 mmol, 26.27 mL, 3 eq) was added. The reaction was carried out at 20° C. for 16 hours. After the reaction was completed, water (150 mL) was added to the system for extraction and phase separation. The aqueous phase was then extracted once with dichloromethane (150 mL). The organic phases were combined, washed once with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 20%) to obtain compound A-3. LCMS m/z=209.0 [M+1]+.


Reference Example 4



embedded image


Step 1: Synthesis of Compound A-4

Compound A-4-1 (3 g, 12.48 mmol, 1 eq), dimethylphosphine oxide (1.02 g, 13.10 mmol, 1.05 eq), potassium phosphate tribasic (3.97 g, 18.72 mmol, 1.5 eq), and Xantphos (721.98 mg, 1.25 mmol, 0.1 eq) were dissolved in 1 4-dioxane (30 mL). After replacing with nitrogen three times, palladium acetate (280.13 mg, 1.25 mmol, 0.1 eq) was added. The mixture was stirred at 120° C. for 2 hours. The reaction mixture was filtered, and the filter cake was rinsed with ethyl acetate (30 mL*2). The filtrate was extracted with water (30 mL), and the organic phase was dried over anhydrous sodium sulfate and then concentrated. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 20%) to obtain compound A-4. LCMS m/z=191.0 [M+H]+.


Reference Example 5



embedded image


Step 1: Synthesis of Compound A-5

Compound A-5-1 (10 g, 35.35 mmol, 1 eq), dimethylphosphine oxide (2.76 g, 35.35 mmol, 1 eq), triethylamine (4.18 g, 41.36 mmol, 5.76 mL, 1.17 eq), and Xantphos (204.53 mg, 353.48 μmol, 0.01 eq) were dissolved in 1 4-dioxane (50 mL) and THF (50 mL). After replacing with nitrogen three times, tris(dibenzylideneacetone)dipalladium(0) (161.84 mg, 176.74 μmol, 0.005 eq) was added. The mixture was stirred at 15° C. for 2 hours. The reaction mixture was added with water (50 mL) and extracted with ethyl acetate (100 mL*2). The organic phase was dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 9%) to obtain compound A-5. LCMS m/z=232.8 [M+H]+.


To synthesize reference examples 6 and 7 listed in Table 1, follow the synthetic steps of reference example 5, replacing dimethylphosphine oxide in step 1 with fragment 2, and replacing A-5-1 with fragment 1.














TABLE 1





Ref-







er-







ence







ex-







am-
Com-
Fragment
Fragment
Structural
Spec-


ple
pound
1
2
formula
trum







6
A-6


embedded image




embedded image




embedded image


LCMS m/z = 260.8 [M + 1]+.





7
A-7


embedded image




embedded image




embedded image


LCMS m/z = 262.0 [M + 1]+.









Reference Example 8



embedded image


Step 1: Synthesis of Compound A-8-2

A-8-1 (1.38 g, 10.00 mmol, 1.29 mL, 1 eq) was dissolved in THE (30 mL). The mixture was cooled to −70° C., and a solution of cyclopropylmagnesium bromide in tetrahydrofuran (0.5 M, 3.09 g, 30 mmol, 60 mL, 3 eq) was added dropwise thereto. The mixture was stirred for 2 hours, then slowly warmed to 25° C., and stirred for 3 hours. The mixture was then cooled to 0° C., added with 0.5 M hydrochloric acid aqueous solution (40 mL) to quench the reaction, and then extracted with ethyl acetate (30 mL*3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound A-8-2. LCMS m/z=131.2 [M+H]+.


Step 2: Synthesis of Compound A-8

2-Bromo-5-iodopyridine (800 mg, 2.82 mmol, 1 eq), A-8-2 (366.69 mg, 2.82 mmol, 1 eq), and Xantphos (163.05 mg, 281.80 μmol, 0.1 eq) were dissolved in 1,4-dioxane (14 mL). Triethylamine (5.64 mmol, 5.64 mL, 2 eq) was added, followed by replacement with nitrogen. Finally, the catalyst Pd2(dba)3 (258.05 mg, 281.80 μmol, 0.1 eq) was added, followed by replacement with nitrogen. The reaction mixture was heated to 75° C. and stirred for 12 hours. After the reaction was completed, the reaction mixture was cooled, added with 20 mL of water, and extracted with dichloromethane (30 mL*2). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:0 to 50:1) to obtain compound A-8. LCMS m/z=285.9 [M+1]+.


To synthesize reference examples 9 and 10 listed in Table 2, follow the synthetic steps of reference example 8, replacing cyclopropylmagnesium bromide in step 1 with fragment 1, and replacing 2-bromo-5-iodopyridine in step 2 with fragment 2.














TABLE 2





Reference

Fragment
Fragment
Structural



example
Compound
1
2
formula
Spectrum




















9
A-9 


embedded image




embedded image




embedded image


LCMS m/z = 285.1 [M + H]+.





10
A-10


embedded image




embedded image




embedded image


LCMS m/z = 290.7 [M + H]+.









Reference Example 11



embedded image


Step 1: Synthesis of Compound A-11

Compound A-3-2 (7 g, 35.19 mmol, 1 eq) was dissolved in DCM (105 mL). Oxalyl chloride (6.25 g, 49.27 mmol, 4.31 mL, 1.4 eq) was then added to the mixture, and DMF (128.60 mg, 1.76 mmol, 135.37 μL, 0.05 eq) was added thereto. The reaction was carried out at 20° C. for 3 hours. The reaction system was concentrated, then dichloromethane (80 mL) was added, and the mixture was further concentrated. The concentrated mixture was added to dichloromethane (150 mL), and methylamine hydrochloride (2.26 g, 33.43 mmol, 0.95 eq) was added. After cooling to −60° C., DIPEA (13.64 g, 105.57 mmol, 18.39 mL, 3 eq) was added. The reaction was carried out at 20° C. for 10 hours. After the reaction was completed, water (80 mL) was added to the system for extraction and phase separation. The aqueous phase was then extracted with dichloromethane (100 mL). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 20%) to obtain compound A-11. LCMS m/z=206.0 [M+1]+.


Reference Example 12



embedded image


Step 1: Synthesis of Compound A-12

Compound A-2-2 (8.77 g, 41.67 mmol, 1 eq) was dissolved in DCM (100 mL). Methylamine hydrochloride (2.81 g, 41.67 mmol, 1 eq) was then added to the mixture. After the mixture was cooled to 0° C., DIPEA (16.16 g, 125.01 mmol, 21.78 mL, 3 eq) was added thereto. The reaction was carried out at 20° C. for 12 hours. After the reaction was completed, water (50 mL) was added to the system for extraction and phase separation. The aqueous phase was then extracted with dichloromethane (50 mL). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound A-12. LCMS m/z=205.0 [M+1]+.


Reference Example 13



embedded image


Step 1: Synthesis of Compound A-13-2

Compound A-13-1 (6 g, 24.00 mmol, 1 eq) and stannous chloride dihydrate (21.66 g, 95.99 mmol, 4 eq) were dissolved in ethyl acetate (200 mL). The mixture was stirred at 70° C. for 1 hour. After cooling to room temperature, the reaction mixture was added with water (200 mL), and extracted with ethyl acetate (100 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (V/V, ethyl acetate/petroleum ether=0 to 10%) to obtain compound A-13-2. LCMS m/z=219.7 [M+H]+.


Step 2: Synthesis of Compound A-13

Compound A-13-2 (2 g, 9.09 mmol, 1 eq), bis(pinacolato)diboron (3.46 g, 13.63 mmol, 1.5 eq), and potassium acetate (2.68 g, 27.27 mmol, 3 eq) were dissolved in 1,4-dioxane (20 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (371.13 mg, 0.45 mmol, 0.05 eq) was added, and the mixture was stirred at 100° C. for 18 hours. After cooling to room temperature, the reaction mixture was added with water (200 mL) and extracted with ethyl acetate (100 mL*2). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (V/V, ethyl acetate/petroleum ether=0 to 25%) to obtain compound A-13. LCMS m/z=267.9 [M+H]+.


Reference Example 14



embedded image


Step 1: Synthesis of Compound A-14

Compound A-14-1 (1 g, 6.71 mmol, 1 eq) was dissolved in N,N-dimethylformamide (10 mL), and then sodium 2-propanethiolate (724.39 mg, 7.38 mmol, 1.1 eq) was added thereto. The mixture was stirred at 15° C. for 1 hour. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (10 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound A-14. LCMS m/z=189.2 [M+H]+.


Reference Example 15



embedded image


Step 1: Synthesis of Compound A-15

Compound A-15-1 (1 g, 5.20 mmol, 1 eq) was dissolved in N,N-dimethylformamide (20 mL), and then sodium 2-propanethiolate (2.91 g, 29.62 mmol, 5.7 eq) was added thereto. The mixture was stirred at 15° C. for 1 hour. The reaction mixture was added with water (20 mL) and extracted with ethyl acetate (20 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound A-15. LCMS m/z=232.0 [M+H]+.




embedded image


embedded image


Step 1: Synthesis of Compound 1-2

Compound 1-1 (280 mg, 637.15 μmol, 1 eq) was dissolved in DMF (7 mL), and then sodium thiomethoxide (178.63 mg, 2.55 mmol, 162.39 μL, 4 eq) was added thereto. The mixture was stirred at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 1-2. LCMS m/z=468.1 [M+H]+.


Step 2: Synthesis of Hydrochloride of Compound WX-001 and Hydrochloride of Compound WX-002

Compound 1-2 (40 mg, 85.55 μmol, 1 eq) was dissolved in DCM (2 mL). The mixture was cooled to 0° C. and added with 3-chloroperoxybenzoic acid (26.05 mg, 128.33 μmol, purity of 85%, 1.5 eq). The reaction was carried out at 20° C. for 2 hours. 3-chloroperoxybenzoic acid was then added thereto (8.68 mg, 42.78 μmol, purity of 85%, 0.5 eq). The reaction was carried out for another 2 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 5% to 35%, 8 minutes) to obtain hydrochloride of compound WX-001 and hydrochloride of compound WX-002.


Hydrochloride of WX-001: 1H NMR (400 MHz, CD3OD) δ: 9.40 (s, 2H), 8.34 (s, 1H), 7.99-7.96 (m, 1H), 7.67-7.66 (m, 1H), 7.46 (t, J=8.0 Hz, 1H), 6.54 (s, 1H), 3.82 (s, 3H), 3.35 (s, 3H), 1.81-1.70 (m, 1H), 1.11-1.01 (m, 4H); LCMS m/z=500.1 [M+H]+.


Hydrochloride of WX-002: 1H NMR (400 MHz, CD3OD) δ: 9.21 (s, 2H), 8.36 (s, 1H), 7.93-7.91 (m, 1H), 7.66-7.64 (m, 1H), 7.44 (t, J=8.0 Hz, 1H), 6.56 (s, 1H), 3.78 (s, 3H), 3.06 (s, 3H), 1.81-1.72 (m, 1H), 1.11-1.01 (m, 4H); LCMS m/z=484.1 [M+H]+.


Example 3



embedded image


Step 1: Synthesis of Compound WX-003

Compound 1-2 (25 mg, 53.47 μmol, 1 eq) was dissolved in MeOH (0.2 mL), and then (diacetoxyiodo)benzene (51.67 mg, 160.41 μmol, 3 eq) and ammonium acetate (16.49 mg, 213.88 μmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 2 hours. The system was added with water (5 mL) and ethyl acetate (5 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (5 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel thin-layer chromatography (V:V:V, EtOAc:DCM:MeOH=5:5:1) to obtain compound WX-003. 1H NMR (400 MHz, CDCl3) δ: 10.37 (s, 1H), 9.34 (s, 2H), 9.13 (s, 1H), 8.25 (s, 1H), 8.05 (s, 1H), 7.66 (d, J=8.0 Hz, 2H), 7.25 (t, J=8.0 Hz, 1H), 6.65 (s, 1H), 3.85 (s, 3H), 3.25 (s, 3H), 1.60-1.56 (m, 1H), 1.06-1.02 (m, 2H), 0.88-0.83 (m, 2H); LCMS m/z=499.2 [M+H]+.


Example 4



embedded image


Step 1: Synthesis of Compound 4-2

Compound 4-1 (200 mg, 847.15 μmol, 1 eq), compound A-1 (211.04 mg, 847.15 μmol, 1 eq), and sodium carbonate (269.37 mg, 2.54 mmol, 3 eq) were dissolved in DMF (4 mL) and water (0.8 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (69.18 mg, 84.71 μmol, 0.1 eq) was added thereto. The reaction was carried out at 80° C. for 12 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain compound 4-2. LCMS m/z=279.0 [M+H]+.


Step 2: Synthesis of Compound 4-3

Compound 4-2 (180 mg, 646.72 μmol, 1 eq) and compound A-2 (161.47 mg, 776.07 mol, 1.2 eq) were added to THE (4.5 mL). After cooling to 0° C., the mixture was added with LiHMDS (1 M, 1.94 mL, 3.0 eq). The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (20 mL) and ethyl acetate (20 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (20 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain compound 4-3. LCMS m/z=450.1 [M+H]+.


Step 3: Synthesis of Compound WX-004

Compound 4-3 (270 mg, 600.10 μmol, 1 eq) and cyclopropanecarboxamide (1.28 g, 15.00 mmol, 25 eq) were added to 1,4-dioxane (20 mL) and NMP (4 mL). Cesium carbonate (586.57 mg, 1.80 mmol, 3 eq) and Xantphos (52.08 mg, 90.01 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (82.43 mg, 90.01 μmol, 0.15 eq) was added. The reaction was carried out at 120° C. for 18 hours. After the reaction was completed, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (30 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 100%) to obtain compound WX-004. 1H NMR (400 MHz, CDCl3) δ: 10.55 (s, 1H), 9.22 (s, 1H), 8.74 (s, 1H), 8.30 (s, 1H), 8.23-8.18 (m, 2H), 8.10 (s, 1H), 7.60 (d, J=8.0 Hz, 2H), 7.30 (t, J=8.0 Hz, 1H), 6.55 (s, 1H), 3.57 (s, 3H), 3.17 (s, 3H), 1.59-1.55 (m, 1H), 1.09-1.05 (m, 2H), 0.91-0.86 (m, 2H); LCMS m/z=499.1 [M+H]+.


To synthesize example 7 listed in Table 3, follow the synthesis steps of example 4, replacing 4-1 in step 1 with fragment 7-1 in the table below.













TABLE 3





Example
Compound
Fragment
Structural formula
Spectrum







7
Hydrochloride of WX-007


embedded image




embedded image



1H NMR (400 MHz, MeOD) δ: 8.34 (s, 1H), 8.06 (d, J = 8.0 Hz, 2H), 7.88 (d, J = 8.0 Hz, 2H), 7.56-7.53 (m, 1H), 7.49-7.46 (m, 1H), 7.41 (t, J = 8.0 Hz, 1H), 6.68 (s, 1H), 3.47 (s, 3H), 3.19 (s, 3H), 1.80-1.76 (m, 1H), 1.12- 1.09 (m, 2H), 1.06-1.02 (m, 2H); LCMS m/z = 498.1 [M + H]+. Purification method: preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80 * 40 mm * 3 μm; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 23% to 42%, 6 minutes).








7-1
WX-007









To synthesize example 26 listed in Table 4, follow the synthesis steps of example 4, replacing 4-1 in step 1 with fragment 7-1 in the table below, replacing A-1 with A-13, and replacing A-2 in step 2 with A-12.













TABLE 4





Example
Compound
Fragment
Structural formula
Spectrum







26
Trifluoroacetate of WX-026


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 12.45 (s, 1H), 11.32 (s, 1H), 8.27 (s, 1H), 8.19 (s, 1H), 7.97 (d, J = 8.4 Hz, 2H), 7.75 (d, J = 8.4 Hz, 2H), 7.17 (dd, J = 3.0, 8.6 Hz, 1H), 6.94 (dd, J = 3.0, 8.4 Hz, 2H), 3.31 (s, 3H), 3.06 (s, 3H), 3.06 (d, J = 4.8 Hz, 3H), 1.91-1.87 (m, 1H), 1.06-0.99 (m, 2H), 0.96-0.89 (m, 2H); LCMS m/z = 513.1 [M + 1]+. Purification method: preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100 * 40 mm * 3 μm; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 26% to 56%, 8 minutes).








7-1
WX-026









Example 5



embedded image


embedded image


Step 1: Synthesis of Compound 5-2

Compound 1-1 (0.45 g, 1.02 mmol, 1 eq) was dissolved in DMF (5 mL). Cesium carbonate (500.45 mg, 1.54 mmol, 1.5 eq) and benzyl mercaptan (254.37 mg, 2.05 mmol, 239.97 μL, 2 eq) were added thereto, and the mixture was stirred at 40° C. for 1 hour. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 5-2. LCMS m/z=544.3 [M+H]+.


Step 2: Synthesis of Compound 5-3

Compound 5-2 (200 mg, 367.88 μmol, 1 eq) was dissolved in acetic acid (2 mL) and water (0.6 mL). The mixture was then cooled to 0° C., and N-chlorosuccinimide (221.06 mg, 1.66 mmol, 4.5 eq) was added thereto. The mixture was stirred at 20° C. for 2 hours. Sodium sulfate was added to the system for drying, and the system was directly used in the next step. Compound 5-3 was obtained. LCMS m/z=520.0 [M+H]+.


Step 3: Synthesis of Compound WX-005

Compound 5-3 (50 mg, 96.16 μmol, 1 eq) was dissolved in THF (2 mL). The mixture was then cooled to −20° C., and dimethylamine hydrochloride (392.06 mg, 4.81 mmol, 440.52 L, 50 eq) and triethylamine (1.46 g, 14.42 mmol, 2.01 mL, 150 eq) were added thereto. The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Waters Xbridge BEH C18 100*30 mm*10 m; mobile phase: A (water containing 10 mM ammonium bicarbonate) and B (acetonitrile); gradient: B %: 30% to 65%, 10 minutes) to obtain compound WX-005. 1H NMR (400 MHz, CDCl3) δ:10.44 (s, 1H), 9.18 (s, 2H), 8.30 (s, 1H), 8.26 (s, 1H), 8.12 (s, 1H), 7.75-7.69 (m, 2H), 7.33 (t, J=8.0 Hz, 1H), 6.20 (s, 1H), 3.87 (s, 3H), 2.87 (s, 6H), 1.54-1.51 (m, 1H), 1.11-1.08 (m, 2H), 0.92-0.87 (m, 2H); LCMS m/z=529.2 [M+H]+.


Example 6



embedded image


Step 1: Synthesis of Hydrochloride of Compound WX-006

Compound 5-3 (95.6 mg, 183.85 μmol, 1 eq) was dissolved in THE (2 mL). The mixture was then cooled to −20° C., and tetrahydropyrrole (13.08 mg, 183.85 μmol, 15.35 μL, 1 eq) and triethylamine (2.79 g, 27.58 mmol, 3.84 mL, 150 eq) were added thereto. The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 15% to 45%, 8 minutes) to obtain hydrochloride of compound WX-006. 1H NMR (400 MHz, CD3OD) δ: 9.30 (s, 2H), 8.35 (s, 1H), 7.98-7.95 (m, 1H), 7.68-7.65 (m, 1H), 7.47-7.43 (m, 1H), 6.58 (s, 1H), 3.81 (s, 3H), 3.39-3.32 (m, 4H), 1.88-1.86 (m, 4H), 1.85-1.70 (m, 1H), 1.12-1.08 (m, 2H), 1.04-1.02 (m, 2H); LCMS m/z=555.2 [M+H]+.


Example 8



embedded image


Step 1: Synthesis of Compound 8-1

Compound A-5 (1 g, 4.29 mmol, 1 eq), compound A-1 (1.07 g, 4.29 mmol, 1 eq), and potassium phosphate tribasic (1.82 g, 8.58 mmol, 2 eq) were dissolved in water (4 mL) and 1,4-dioxane (20 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (350.43 mg, 429.11 μmol, 0.1 eq) was added. The mixture was stirred at 80° C. for 2 hours. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (20 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 8-1. 1HNMR (400 MHz, CDCl3) δ: 7.81-7.71 (m, 4H), 7.02-6.98 (m, 1H), 6.81-6.72 (m, 2H), 3.95 (s, 2H), 3.40 (s, 3H), 1.81 (s, 3H), 1.78 (s, 3H); LCMS m/z=276.1 [M+H]+.


Step 2: Synthesis of Compound 8-2

Compound 8-1 (200 mg, 726.53 μmol, 1 eq) and compound A-3 (151.88 mg, 726.53 mol, 1 eq) were dissolved in THE (10 mL). LiHMDS (1 M, 2.91 mL, 4 eq) was added. The mixture was stirred at 15° C. for 1 hour. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (20 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 8-2. LCMS m/z=448.1 [M+H]+.


Step 3: Synthesis of Hydrochloride of Compound WX-008

Compound 8-2 (0.1 g, 223.28 μmol, 1 eq), cyclopropanecarboxamide (475.05 mg, 5.58 mmol, 25 eq), cesium carbonate (218.25 mg, 669.84 μmol, 3 eq), and Xantphos (12.92 mg, 22.33 μmol, 0.1 eq) were dissolved in dioxane (5 mL). After replacing with nitrogen three times, Pd2(dba)3 (20.45 mg, 22.33 μmol, 0.1 eq) was added. The mixture was stirred at 120° C. for 4 hours. The reaction mixture was added with water (5 mL) and extracted with ethyl acetate (10 mL*3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*40 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 24% to 46%, 7 minutes) to obtain hydrochloride of compound WX-008. 1HNMR (400 MHz, CD3OD) δ: 7.94-7.88 (m, 2H), 7.84-7.81 (m, 2H), 7.58-7.55 (m, 1H), 7.51-7.48 (m, 1H), 7.43-7.39 (m, 1H), 6.95 (s, 1H), 3.47 (s, 3H), 1.86 (s, 3H), 1.83 (s, 3H), 1.68-1.61 (m, 1H), 1.18-1.06 (m, 4H); LCMS m/z=497.1 [M+H]+.


To synthesize example 27 listed in Table 5, follow the synthesis steps of example 8, replacing A-1 in step 1 with A-13.













TABLE 5





Example
Compound
Fragment
Structural formula
Spectrum







27
Hydrochloride of WX-027


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 11.20 (s, 1H), 8.91 (br s, 1H), 8.28 (s, 1H), 8.07 (s, 1H), 7.77-7.65 (m, 4H), 7.16 (dd, J = 2.9, 9.2 Hz, 1H), 6.80 (dd, J = 2.9, 8.7 Hz, 1H), 3.33 (s, 3H), 1.73 (d, J = 12.8 Hz, 6H), 1.66- 1.60 (m, 1H), 1.11- 1.02 (m, 2H), 0.93-0.87 (m, 2H); LCMS m/z = 537.1 [M + Na]+. Purification method: preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80 * 40 mm * 3 μm; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 24% to 46%, 7 minutes).








A-13
WX-027









Example 9



embedded image


Step 1: Synthesis of Compound 9-1

Compound 1-1 (50 mg, 113.78 μmol, 1 eq) was dissolved in DMF (1 mL), and then sodium 2-propanethiolate (44.67 mg, 455.11 μmol, 4 eq) was added thereto. The reaction was carried out at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate:petroleum ether=10% to 30%) to obtain compound 9-1. LCMS m/z=496.2 [M+H]+.


Step 2: Synthesis of Trifluoroacetate of Compound WX-009

Compound 9-1 (56 mg, 112.99 μmol, 1 eq) was added to MeOH (1 mL), and (diacetoxyiodo)benzene (109.18 mg, 338.98 μmol, 3 eq) and ammonium acetate (34.84 mg, 451.97 μmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 2 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (column: Welch Xtimate C18 100*40 mm*3 m; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 23 to 63%, 8 minutes) to obtain trifluoroacetate of compound WX-009. 1H NMR (400 MHz, CDCl3) δ: 11.97-11.77 (m, 1H), 11.60-11.42 (m, 1H), 9.33 (s, 2H), 8.73-8.65 (m, 1H), 8.73-8.65 (m, 1H), 8.01-7.93 (m, 1H), 7.67-7.62 (m, 1H), 7.46-7.40 (m, 1H), 3.90-3.83 (m, 3H), 3.52-3.41 (m, 3H), 1.93-1.83 (m, 1H), 1.52-1.38 (m, 6H), 1.13-1.07 (m, 2H), 1.05-0.95 (m, 2H); LCMS m/z=527.2 [M+H]+.


Example 10



embedded image


embedded image


Step 1: Synthesis of Compound 10-2

Compound 10-1 (997.91 mg, 5.94 mmol, 2 eq), compound A-1-1 (600 mg, 2.97 mmol, 1 eq), and potassium phosphate tribasic (1.89 g, 8.91 mmol, 3 eq) were added to 1,4-dioxane (24 mL). After replacing with nitrogen, Pd(dppf)Cl2·CH2Cl2 (242.51 mg, 296.96 μmol, 0.1 eq) was added. The reaction was carried out at 100° C. for 3 hours. The reaction mixture was added with water (40 mL) and ethyl acetate (40 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (40 mL). The organic phases were combined, washed twice with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 20%) to obtain compound 10-2. LCMS m/z=246.1 [M+H]+.


Step 2: Synthesis of Compound 10-3

Compound 10-2 (300 mg, 1.22 mmol, 1 eq) and compound A-2 (279.86 mg, 1.35 mmol, 1.1 eq) were added to THE (6 mL). After cooling to 0° C., the mixture was added with LiHMDS (1 M, 4.89 mL, 4 eq). The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction was quenched by methanol. The reaction mixture was added with water (20 mL) and ethyl acetate (20 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (20 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain compound 10-3. LCMS m/z=417.1 [M+H]+.


Step 3: Synthesis of Compound 10-4

Compound 10-3 (300 mg, 719.53 μmol, 1 eq) and cyclopropanecarboxamide (1.53 g, 17.99 mmol, 25 eq) were added to 1,4-dioxane (2 mL), NMP (0.4 mL), and water (0.4 mL). Cesium carbonate (703.31 mg, 2.16 mmol, 3 eq) and Xantphos (62.45 mg, 107.93 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen, Pd2(dba)3 (98.83 mg, 107.93 μmol, 0.15 eq) was added. After replacing with nitrogen, the reaction was carried out at 120° C. for 3 hours. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed twice with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 100%) to obtain compound 10-4. LCMS m/z=466.1 [M+H]+.


Step 4: Synthesis of Compound 10-5

Compound 10-4 (200 mg, 429.57 μmol, 1 eq) was added to methanol (4 mL), and then (diacetoxyiodo)benzene (415.09 mg, 1.29 mmol, 3 eq) and ammonium acetate (132.45 mg, 1.72 mmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 3 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by preparative thin-layer chromatography on a silica gel plate (ethyl acetate:dichloromethane :methanol=5:5:1) to obtain compound 10-5. 1H NMR (400 MHz, CDCl3) δ: 10.47 (s, 1H), 8.27 (s, 1H), 8.18 (s, 2H), 8.06 (d, J=8.0 Hz, 2H), 7.81 (d, J=8.0 Hz, 2H), 7.56 (d, J=8.0 Hz, 1H), 7.26 (t, J=8.0 Hz, 1H), 7.11-7.08 (m, 1H), 6.20 (s, 1H), 3.45 (s, 3H), 3.18 (s, 3H), 2.76 (s, 1H), 1.57-1.52 (m, 1H), 1.11-1.08 (m, 2H), 0.92-0.87 (m, 2H); LCMS m/z=497.2 [M+H]+.


Step 5: Synthesis of Hydrochloride of Compound WX-010

Compound 10-5 (40 mg, 80.55 μmol, 1 eq) was dissolved in 1,4-dioxane (3 mL), and then Cu(OAc)2 (21.95 mg, 120.82 μmol, 1.5 eq) and pyridine (15.29 mg, 193.32 μmol, 15.60 L, 2.4 eq) were added thereto. The mixture was stirred in an open container for 10 minutes, and then added with methylboronic acid (9.64 mg, 161.10 μmol, 2 eq). The reaction was carried out at 100° C. for 12 hours. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 10% to 30%, 8 minutes) to obtain hydrochloride of compound WX-010. 1H NMR (400 MHz, CDCl3) δ: 10.47 (s, 1H), 8.27-8.24 (m, 2H), 8.19 (s, 1H), 7.94 (d, J=8.0 Hz, 2H), 7.82 (d, J=8.0 Hz, 2H), 7.57-7.55 (m, 1H), 7.28-7.24 (m, 1H), 7.12-7.10 (m, 1H), 6.19 (s, 1H), 3.47 (s, 3H), 3.14 (s, 3H), 2.72 (s, 3H), 1.56-1.53 (m, 1H), 1.12-1.08 (m, 2H), 0.92-0.87 (m, 2H); LCMS m/z=511.2 [M+H]+.


Example 11



embedded image


embedded image


embedded image


Step 1: Synthesis of Compound 11-2

Compound 11-1 (3.83 g, 28.90 mmol, 3.58 mL, 0.9 eq), compound A-1 (8 g, 32.11 mmol, 1 eq), and potassium phosphate tribasic (20.45 g, 96.34 mmol, 3 eq) were added to 1,4-dioxane (120 mL) and water (24 mL). After replacing with nitrogen, Pd(dppf)Cl2·CH2Cl2 (1.57 g, 1.93 mmol, 0.06 eq) was added. The reaction was carried out at 100° C. for 6 hours. The reaction mixture was added with water (100 mL) and ethyl acetate (100 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (100 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 20%) to obtain compound 11-2. LCMS m/z=220.0 [M+H]+.


Step 2: Synthesis of Compound 11-3

Compound 11-2 (3.0 g, 13.69 mmol, 1 eq) and compound A-11 (3.67 g, 17.79 mmol, 1.3 eq) were added to THE (30 mL). After cooling to −60° C., the mixture was added with LiHMDS (1 M, 41.06 mL, 3 eq). The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction was quenched by methanol (10 mL). The reaction mixture was added with water (50 mL) and ethyl acetate (50 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (50 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain compound 11-3. LCMS m/z=389.0 [M+H]+.


Step 3: Synthesis of Compound 11-4

Compound 11-3 (4.2 g, 10.80 mmol, 1 eq) and cyclopropanecarboxamide (9.19 g, 108.03 mmol, 10 eq) were added to NMP (80 mL). Cesium carbonate (10.56 g, 32.41 mmol, 3 eq) and Xantphos (937.62 mg, 1.62 mmol, 0.15 eq) were then added thereto. After replacing with nitrogen, Pd2(dba)3 (1.48 g, 1.62 mmol, 0.15 eq) was added. After replacing with nitrogen, the reaction was carried out at 140° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (100 mL) and ethyl acetate (100 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (100 mL). The organic phases were combined, washed twice with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 11-4. LCMS m/z=438.1 [M+H]+.


Step 4: Synthesis of Compound 11-5

Compound 11-4 (300 mg, 685.83 μmol, 1 eq) was dissolved in DMF (5 mL), and then sodium 2-propanethiolate (100.96 mg, 1.03 mmol, 1.5 eq) was added thereto. The reaction was carried out at 60° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 11-5. LCMS m/z=494.1 [M+H]+.


Step 5: Synthesis of Compound WX-011

Compound 11-5 (400 mg, 810.41 μmol, 1 eq) was added to MeOH (30 mL), and then (diacetoxyiodo)benzene (783.08 mg, 2.43 mmol, 3 eq) and ammonium acetate (249.86 mg, 3.24 mmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 2 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 45% to 45%, 8 minutes). The obtained solution was concentrated under vacuum at 40° C. to remove acetonitrile, then adjusted to alkalinity (pH=8) with saturated sodium bicarbonate, and extracted with dichloromethane (30 mL*3). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under vacuum and dried to obtain compound WX-011. LCMS m/z=525.2 [M+H]+.


Step 6: Synthesis of Compounds WX-011A and WX-011B

WX-011 was subjected to resolution by SFC (column: DAICEL CHIRALCEL OD-H (250 mm*30 mm, 5 m); mobile phase: A (CO2) and B (ethanol containing 0.1% ammonia water); gradient: B %=65% to 65%, 15 minutes) to obtain WX-011A and WX-011B.


WX-011A: 1H NMR (400 MHz, CDCl3) δ: 11.07 (s, 1H), 10.05 (s, 1H), 9.28 (s, 2H), 8.25 (s, 1H), 8.13-8.12 (m, 1H), 7.80 (d, J=8.0 Hz, 1H), 7.64 (d, J=8.0 Hz, 1H), 7.33 (t, J=8.0 Hz, 1H), 3.83 (s, 3H), 3.38-3.33 (m, 1H), 3.03 (s, 3H), 1.91-1.88 (m, 1H), 1.41 (t, J=8.0 Hz, 6H), 1.10-1.08 (m, 2H), 0.92-0.89 (m, 2H); LCMS m/z=525.2 [M+H]+. SFC detection method: (column: Chiralpak AD-3, 3 μm, 0.46 cm id×5 cm L; mobile phase: A (CO2) and B (EtOH containing 0.1% isopropylamine); gradient: B %=50 to 50%, 5 minutes; flow rate: 4.0 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 2.538 min, and a chiral isomer excess of 100%.


WX-011B: 1H NMR (400 MHz, CDCl3) δ: 11.07 (s, 1H), 9.88 (s, 1H), 9.28 (s, 2H), 8.25 (s, 1H), 8.10-8.09 (m, 1H), 7.80 (d, J=8.0 Hz, 1H), 7.64 (d, J=8.0 Hz, 1H), 7.33 (t, J=8.0 Hz, 1H), 3.84 (s, 3H), 3.36-3.33 (m, 1H), 3.03 (s, 3H), 1.85-1.82 (m, 1H), 1.41 (t, J=8.0 Hz, 6H), 1.11-1.09 (m, 2H), 0.93-0.91 (m, 2H); LCMS m/z=525.2 [M+H]+. SFC detection method: (column: Chiralpak AD-3, 3 μm, 0.46 cm id×5 cm L; mobile phase: A (CO2) and B (EtOH containing 0.1% isopropylamine); gradient: B %=50 to 50%, 5 minutes; flow rate: 4.0 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 3.011 min, and a chiral isomer excess of 98.60%.


Step 7: Synthesis of Hydrochloride of Compound WX-011A

Compound WX-011A (790 mg, 1.51 mmol, 1 eq) was dissolved in MeCN (6 mL), and HCl/EtOAc (4 M, 1.13 mL, 3 eq) was added thereto. The mixture was stirred until clear. Water (50 mL) was then added to the system to obtain hydrochloride of compound WX-011A. 1H NMR (400 MHz, CD3OD) δ: 9.42 (s, 2H), 8.08 (dd, J=1.6, 8.0 Hz, 1H), 7.78 (dd, J=8.0 Hz, 1H), 7.51 (t, J=8.0 Hz, 1H), 6.95 (s, 1H), 3.88 (s, 3H), 3.86-3.77 (m, 1H), 3.02 (s, 3H), 1.91-1.78 (m, 1H), 1.47 (dd, J=6.8, 12.4 Hz, 6H), 1.19-1.04 (m, 4H); LCMS m/z=525.0 [M+H]+.


Example 12



embedded image


embedded image


embedded image


Step 1: Synthesis of Compound 12-2

Compound 12-1 (3 g, 17.05 mmol, 4.97 mL, 1 eq), A-1 (5.10 g, 20.46 mmol, 1.2 eq), and potassium carbonate (4.71 g, 34.09 mmol, 2 eq) were dissolved in 1,4-dioxane (50 mL) and water (10 mL). After replacing with nitrogen three times, Pd(dppf)Cl2 (1.25 g, 1.70 mmol, 0.1 eq) was added thereto. After replacing with nitrogen once more, the mixture was heated to 80° C. and stirred for 4 hours. The reaction mixture was added with water (50 mL) and extracted with ethyl acetate (50 mL*2). The organic phases were combined and washed with saturated brine (30 mL*2). The organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 30%) to obtain compound 12-2. LCMS m/z=219.1 [M+1]+.


Step 2: Synthesis of Compound 12-3

Under a nitrogen atmosphere, compound 12-2 (2 g, 9.16 mmol, 1 eq) was added to THE (5 mL). A-11 (1.89 g, 9.16 mmol, 1 eq) was added thereto, and the mixture was stirred at 25° C. (room temperature) until dissolved. LiHMDS (1 M, 22.91 mL, 2.5 eq) was slowly added dropwise thereto, and the mixture was stirred at 25° C. (room temperature) for another 1 hour. The reaction mixture was added with saturated ammonium chloride aqueous solution (20 mL) and water (50 mL), and extracted with ethyl acetate (50 mL*2). The organic phases were combined and washed with saturated brine (50 mL*2). The organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 12-3. LCMS m/z=387.9 [M+1]+.


Step 3: Synthesis of Compound 12-4

Compound 12-3 (800 mg, 2.06 mmol, 1 eq) and cyclopropanecarboxamide (1.76 g, 20.63 mmol, 10 eq) were added to 1,4-dioxane (3 mL) and NMP (0.3 mL). Cesium carbonate (2.69 g, 8.25 mmol, 4 eq) and Xantphos (358.10 mg, 618.88 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (320.30 mg, 309.44 μmol, 0.15 eq) was added. The reaction was carried out at 130° C. for 18 hours. After cooling to room temperature, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (30 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 12-4. LCMS m/z=437.3 [M+1]+.


Step 4: Synthesis of Compound 12-5

Compound 12-4 (600 mg, 1.37 mmol, 1 eq) was dissolved in DMF (1 mL), and then sodium 2-propanethiolate (539.69 mg, 5.50 mmol, 4 eq) was added thereto. The reaction was carried out at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 12-5. The crude product was directly used in the next step without purification. LCMS m/z=493.2 [M+1+.


Step 5: Synthesis of Compound WX-012

12-5 (350 mg, 710.53 μmol, 1 eq) was added to MeOH (20 mL), and (diacetoxyiodo)benzene (686.57 mg, 2.13 mmol, 3 eq) and ammonium acetate (219.08 mg, 2.84 mmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 2 hours. The reaction mixture was added with water (20 mL) and ethyl acetate (20 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (20 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution (20 mL), dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 3%) to obtain compound WX-012. 1H NMR (400 MHz, CDCl3) δ: 11.16 (s, 1H), 9.21 (d, J=2.4 Hz, 1H), 8.53 (br dd, J=2.5, 6.5 Hz, 1H), 8.29-8.14 (m, 4H), 7.69 (dd, J=1.5, 7.8 Hz, 1H), 7.57 (dd, J=1.4, 7.9 Hz, 1H), 7.38-7.32 (m, 1H), 3.59-3.52 (m, 1H), 3.56 (s, 3H), 3.39-3.29 (m, 1H), 3.07 (d, J=5.0 Hz, 3H), 2.82 (br s, 1H), 1.69-1.61 (m, 1H), 1.39 (dd, J=6.8, 13.3 Hz, 6H), 1.18-1.07 (m, 2H), 1.00-0.93 (m, 2H); LCMS m/z=524.1 [M+1]+.


Step 6: Synthesis of Compounds WX-012A and WX-012B

WX-012 was subjected to resolution by SFC (column: DAICEL CHIRALPAK AD (250 mm*30 mm, 10 m); mobile phase: A (CO2) and B (isopropanol containing 0.1% ammonia water); gradient: B %=50% to 50%, 15 minutes) to obtain WX-012A and WX-012B.


WX-012A: 1H NMR (400 MHz, CDCl3) δ: 11.16 (s, 1H), 9.22 (d, 2.4 Hz, 1H), 8.54 (br s, 1H), 8.30-8.15 (m, 4H), 7.70 (dd, J=1.5, 7.8 Hz, 1H), 7.58 (dd, J=1.5, 8.0 Hz, 1H), 7.42-7.32 (m, 1H), 3.57 (s, 3H), 3.40-3.30 (m, 1H), 3.08 (d, J=5.2 Hz, 3H), 2.84 (br s, 1H), 1.70-1.62 (m, 1H), 1.40 (dd, J=6.8, 13.3 Hz, 6H), 1.17-1.12 (m, 2H), 1.02-0.95 (m, 2H); LCMS m/z=524.0 [M+H]+. SFC detection method: (column: Chiralpak AD-3, 3 μm, 0.46 cm id×15 cm L; mobile phase: A (CO2) and B (isopropanol containing 0.05% diethylamine); gradient: B %=40%, 8 minutes; flow rate: 2.5 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 4.941 min, and a chiral isomer excess of 99.66%.


WX-012B: 1H NMR (400 MHz, CDCl3) δ: 11.15 (s, 1H), 9.22 (d, J=2.4 Hz, 1H), 8.61 (s, 1H), 8.28-8.19 (m, 2H), 8.21-8.13 (m, 2H), 7.70 (dd, J=1.4, 7.7 Hz, 1H), 7.58 (dd, J=1.6, 7.9 Hz, 1H), 7.36 (t, J=7.9 Hz, 1H), 3.57 (s, 3H), 3.39-3.29 (m, 1H), 3.08 (d, J=5.0 Hz, 3H), 2.84 (br s, 1H), 1.66 (br d, J=4.0 Hz, 1H), 1.40 (dd, J=6.8, 13.3 Hz, 6H), 1.17-1.11 (m, 2H), 1.01-0.94 (m, 2H); LCMS m/z=524.0 [M+H]+. SFC detection method: (column: Chiralpak AD-3, 3 μm, 0.46 cm id×15 cm L; mobile phase: A (CO2) and B (Isopropanol containing 0.05% diethylamine); gradient: B %=40%, 8 minutes; flow rate: 2.5 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 6.054 min, and a chiral isomer excess of 98.66%.


Example 13



embedded image


Step 1: Synthesis of Compound WX-013

Compound WX-011 (20 mg, 38.12 μmol, 1 eq) was added to 1,4-dioxane (2 mL), and then Cu(OAc)2 (20.77 mg, 114.37 μmol, 3 eq) and pyridine (7.24 mg, 91.50 μmol, 7.39 μL, 2.4 eq) were added thereto. The mixture was stirred in an open container for 10 minutes, and then added with methylboronic acid (6.85 mg, 114.37 μmol, 3 eq). The reaction was carried out at 100° C. for 3 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.0400 hydrochloric acid) and B (acetonitrile); gradient: B %0: 15% to 30%, 8 minutes) to obtain a crude product. The crude product was further purified by preparative thin-layer chromatography on a silica gel plate (ethyl acetate:dichloromethane:methanol=5:5:1) to obtain compound WX-013. 1HNMR (400 MHz, CD3CD) δ: 9.31 (s, 2H), 8.10-8.06 (m, 1H), 7.79-7.73 (m, 1H), 7.50 (t, J=8.0 Hz, 2H), 3.83 (s, 3H), 3.03 (s, 3H), 2.92-2.90 (m, 1H), 2.81 (s, 3H), 1.83-1.82 (i, 1H), 1.50 (d, J=8.0 Hz, 3H), 1.40 (d, J=8.0 Hz, 3H), 1.14-1.08 (in, 4H); LCMS m/z=539.2 [M+H]+.


To synthesize example 14 listed in Table 6, follow the synthesis steps of example 13, replacing WX-011 in step 1 with WX-012.












TABLE 6





Example
Compound
Structural formula
Spectrum







14
Trifluoroacetate of WX-014


embedded image



1H NMR (400 MHz, CDCl3) δ: 13.31 (br s, 1H), 11.90 (s, 1H), 9.21 (s, 1H), 8.58-8.39 (m, 2H), 8.31 (d, J = 8.5 Hz, 1H), 7.92 (dd, J = 1.4, 7.9 Hz, 1H), 7.88-7.81 (m, 1H), 7.58 (dd, J = 1.3, 7.8 Hz, 1H), 7.49-7.42 (m, 1H), 4.31-4.19 (m, 1H), 3.60 (s, 3H), 3.08 (d, J = 5.3 Hz, 3H), 2.82 (s, 3H), 2.11-2.01 (m, 1H), 1.62 (d, J = 6.4 Hz, 3H), 1.33 (d, J = 6.4 Hz, 3H), 1.15-1.09 (m, 2H), 1.08-1.02 (m, 2H); LCMS m/z = 538.1 [M + 1]+. Purification method: preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100 * 40 mm * 3 μm; mobile phase: A (water containing 0.04% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 13% to 43%, 8 minutes).








WX-014









Example 15



embedded image


Step 1: Synthesis of Hydrochloride of Compound WX-015

Compound WX-011 (100 mg, 190.62 μmol, 1 eq) was added to 1,4-dioxane (2 mL), and then Cu(OAc)2 (103.87 mg, 571.87 μmol, 3 eq) and pyridine (36.19 mg, 457.50 μmol, 36.93 μL, 2.4 eq) were added thereto. The mixture was stirred in an open container for 10 minutes, and then added with ethylboronic acid (21.13 mg, 285.94 μmol, 1.5 eq). The reaction was carried out at 100° C. for 4 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 20% to 40%, 8 minutes) to obtain hydrochloride of compound WX-015. 1H NMR (400 MHz, CDCl3) δ: 11.07 (s, 1H), 9.16 (s, 2H), 9.04 (s, 1H), 8.21 (s, 1H), 8.14-8.12 (m, 1H), 7.84 (d, J=8.0 Hz, 1H), 7.64 (d, J=8.0 Hz, 1H), 7.36 (t, J=8.0 Hz, 1H), 3.86 (s, 3H), 3.48-3.14 (m, 2H), 3.03 (s, 3H), 1.57-1.54 (m, 1H), 1.44 (d, J=8.0 Hz, 3H), 1.34 (d, J=8.0 Hz, 3H), 1.26-1.24 (m, 3H), 1.10-0.93 (m, 4H); LCMS m/z=553.2 [M+H]+.


To synthesize example 16 listed in Table 7, follow the synthesis steps of example 15, replacing WX-011 in step 1 with WX-012.












TABLE 7





Example
Compound
Structural formula
Spectrum







16
Hydrochloride of WX-016


embedded image


LCMS m/z = 552.2 [M + H]+. Purification method: preparative high performance liquid chromatography (chromatographic column: Xtimate C18 150 * 40 mm * 5 μm; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 20% to 40%, 10 minutes).







WX-016









Example 17



embedded image


embedded image


Step 1: Synthesis of Compound 17-1

Compound 11-2 (2.0 g, 9.12 mmol, 1 eq) and compound A-12 (2.06 g, 10.04 mmol, 1.1 eq) were added to THE (20 mL). After cooling to −60° C., the mixture was added with LiHMDS (1 M, 27.37 mL, 3 eq). The reaction was carried out at 20° C. for 2 hours. After the reaction was completed, the reaction was quenched by methanol (10 mL). The reaction mixture was added with water (50 mL) and ethyl acetate (50 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (50 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain compound 17-1. LCMS m/z=388.0 [M+H]+.


Step 2: Synthesis of Compound 17-2

Compound 17-1 (0.52 g, 1.34 mmol, 1 eq) and cyclopropanecarboxamide (1.14 g, 13.41 mmol, 10 eq) were added to NMP (7 mL). Cesium carbonate (1.31 g, 4.02 mmol, 3 eq) and Xantphos (116.38 mg, 201.14 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen, Pd2(dba)3 (184.19 mg, 201.14 μmol, 0.15 eq) was added. After replacing with nitrogen, the reaction was carried out at 140° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (30 mL). The organic phases were combined, washed twice with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 17-2. LCMS m/z=437.1 [M+H]+.


Step 3: Synthesis of Compound 17-3

Compound 17-2 (500 mg, 1.15 mmol, 1 eq) was dissolved in DMF (5 mL). Cesium carbonate (559.91 mg, 1.72 mmol, 1.5 eq) and benzyl mercaptan (284.58 mg, 2.29 mmol, 268.48 μL, 2 eq) were added. The mixture was stirred at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 17-3. LCMS m/z=541.2 [M+H]+.


Step 4: Synthesis of Compound 17-4

Compound 17-3 (400 mg, 739.87 μmol, 1 eq) was dissolved in acetic acid (4 mL) and water (0.5 mL). The mixture was then cooled to 0° C., and N-chlorosuccinimide (395.18 mg, 2.96 mmol, 4 eq) was added thereto. The mixture was stirred at 25° C. for 4 hours. Sodium sulfate was added to the system for drying, and the system was directly used in the next step. Compound 17-4 was obtained. LCMS m/z=517.0 [M+H]+.


Step 5: Synthesis of Hydrochloride of Compound WX-017

Compound 17-4 (190 mg, 367.54 μmol, 1 eq) was added to THE (2 mL). The mixture was then cooled to −60° C., and dimethylamine hydrochloride (1.50 g, 18.38 mmol, 50 eq) and triethylamine (5.58 g, 55.13 mmol, 7.67 mL, 150 eq) were added. The reaction was carried out at 20° C. for 12 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 17% to 37%, 7 minutes) to obtain hydrochloride of compound WX-017. 1H NMR (400 MHz, CDCl3) δ: 10.35 (s, 1H), 9.17 (s, 2H), 8.27 (s, 2H), 8.12 (s, 1H), 7.72-7.68 (m, J=8.0 Hz, 1H), 7.33 (t, J=8.0 Hz, 1H), 6.20 (s, 1H), 3.87 (s, 3H), 3.03 (s, 3H), 2.86 (s, 6H), 1.68-1.53 (m, 1H), 1.20-1.18 (m, 2H), 0.90-0.86 (m, 2H); LCMS m/z=526.1 [M+H]+.


Example 18



embedded image


Step 1: Synthesis of Hydrochloride of Compound WX-018

Compound 17-4 (190 mg, 367.54 μmol, 1 eq) was added to THE (2 mL). The mixture was then cooled to −60° C., and azetidine hydrochloride (1.72 g, 18.38 mmol, 50 eq) and triethylamine (5.58 g, 55.13 mmol, 7.67 mL, 150 eq) were added. The reaction was carried out at 20° C. for 12 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna C18 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 17% to 37%, 7 minutes) to obtain hydrochloride of compound WX-018. 1H NMR (400 MHz, CDCl3) δ 10.35 (s, 1H), 9.19 (s, 2H), 8.27 (s, 2H), 8.09 (s, 1H), 7.71-7.67 (m, 2H), 7.33 (t, J=8.0 Hz, 1H), 6.22 (s, 1H), 3.93-3.91 (m, 4H), 3.85 (s, 3H), 2.98 (s, 3H), 2.22-2.19 (s, 2H), 1.51-1.49 (m, 1H), 1.05-1.03 (m, 2H), 0.90-0.86 (m, 2H); LCMS m/z=538.2 [M+H]+.


Example 19



embedded image


Step 1: Synthesis of Compound 19-1

Compound 17-2 (200 mg, 458.26 μmol, 1 eq) was dissolved in DMF (7 mL), and then sodium 2-propanethiolate (179.90 mg, 1.83 mmol, 4 eq) was added thereto. The reaction was carried out at 40° C. for 16 hours. After cooling to room temperature, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product of compound 19-1. The crude product was directly used in the next step without purification. 1H NMR (400 MHz, CDCl3) δ: 10.30 (s, 1H), 8.85 (s, 2H), 8.36-8.25 (m, 2H), 8.12 (s, 1H), 7.63 (dd, J=7.8, 15.6 Hz, 2H), 6.24 (br s, 1H), 3.80 (s, 3H), 3.44 (quin, J=6.7 Hz, 1H), 3.01 (d, J=4.4 Hz, 3H), 1.58-1.50 (m, 1H), 1.38 (d, J=6.5 Hz, 6H), 1.13-1.05 (m, 2H), 0.92-0.82 (m, 2H); LCMS m/z=493.1 [M+H]+.


Step 2: Synthesis of Compound WX-019

Compound 19-1 (100 mg, 203.01 μmol, 1 eq) was dissolved in DCM (2 mL). The mixture was then cooled to 0° C. and added with 3-chloroperoxybenzoic acid (52.55 mg, 304.51 mol, purity of 85%, 1.5 eq). The reaction was carried out at 20° C. for 16 hours. The system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (10 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna 80*40 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 5% to 35%, 8 minutes). The obtained solution was concentrated under vacuum at 40° C. to remove acetonitrile, then adjusted to alkalinity (pH=8) with saturated sodium bicarbonate, and extracted with dichloromethane (30 mL*3). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under vacuum and dried to obtain compound WX-019. 1H NMR (400 MHz, CDCl3) δ: 10.39 (s, 1H), 9.26 (s, 2H), 8.31-8.24 (m, 1H), 8.17-8.11 (m, 1H), 7.80-7.70 (m, 2H), 7.39-7.31 (m, 1H), 6.14-6.07 (m, 1H), 3.89 (s, 3H), 3.37-3.25 (m, 1H), 3.04 (d, J=4.8 Hz, 2H), 1.59-1.52 (m, 1H), 1.43 (d, 6H), 1.32-1.27 (m, 2H), 1.12-1.07 (m, 2H). LCMS m/z=525.1 [M+H]+.


Example 20



embedded image


embedded image


Step 1: Synthesis of Compound 20-1

Under a nitrogen atmosphere, compound 12-1 (1 g, 4.58 mmol, 1.35 eq) and compound A-12 (695.62 mg, 3.39 mmol, 1 eq) were dissolved in THE (5 mL). The mixture was stirred at 25° C. (room temperature). LiHMDS (1 M, 8.48 mL, 2.5 eq) was slowly added dropwise thereto. The mixture was stirred at 25° C. (room temperature) for another 1 hour. The reaction mixture was added with saturated ammonium chloride aqueous solution (20 mL) and water (50 mL), and extracted with ethyl acetate (50 mL*2). The organic phases were combined and washed with saturated brine (50 mL*2). The organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 20-1. LCMS m/z=387.0 [M+1]+.


Step 2: Synthesis of Compound 20-2

Compound 20-1 (1.27 g, 3.28 mmol, 1 eq) and cyclopropanecarboxamide (2.79 g, 32.83 mmol, 10 eq) were added to a mixed solvent of 1,4-dioxane (2 mL) and NMP (0.5 mL). Cesium carbonate (3.21 g, 9.85 mmol, 3 eq) and Xantphos (284.97 mg, 492.49 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (450.99 mg, 492.49 μmol, 0.15 eq) was added. The reaction was carried out at 130° C. for 18 hours. After cooling to room temperature, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (30 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 20-2. LCMS m/z=436.1 [M+1]+.


Step 3: Synthesis of Compound 20-3

Compound 20-2 (500 mg, 1.15 mmol, 1 eq) was dissolved in DMF (5 mL), and then sodium 2-propanethiolate (450.76 mg, 4.59 mmol, 4 eq) was added thereto. The reaction was carried out at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (10 mL*2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 20-3. LCMS m/z=492.1 [M+1]+.


Step 4: Synthesis of Hydrochloride of Compound WX-020

Compound 20-3 (100.20 mg, 203.83 μmol, 1 eq) was dissolved in DCM (10 mL). The mixture was then cooled to 0° C. and added with 3-chloroperoxybenzoic acid (82.76 mg, 407.65 mol, purity of 85%, 2 eq). The reaction was carried out at 25° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted with ethyl acetate (30 mL). The organic phases were combined, washed with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated under vacuum to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex C18 150*40 mm*5 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 15% to 45%, 10 minutes) to obtain hydrochloride of compound WX-020. 1H NMR (400 MHz, DMSO-d6) δ: 11.85 (br s, 1H), 11.03 (s, 1H), 9.12 (d, J=1.6 Hz, 1H), 9.06 (br s, 1H), 8.51 (s, 1H), 8.36 (dd, J=2.3, 8.4 Hz, 1H), 8.19 (d, J=8.4 Hz, 1H), 7.73 (br d, J=7.8 Hz, 1H), 7.62 (br d, J=7.5 Hz, 1H), 7.41 (t, J=7.9 Hz, 1H), 7.31 (br s, 1H), 3.53 (s, 3H), 3.46-3.30 (m, 1H), 2.82 (d, J=4.4 Hz, 3H), 1.99-1.89 (m, 1H), 1.24 (d, J=6.8 Hz, 6H), 0.99-0.85 (m, 4H); LCMS m/z=524.1 [M+1]+.


Step 5: Synthesis of Compound WX-020

Compound 20-3 (1.8 g, 3.66 mmol, 1 eq) was dissolved in ethanol (18 mL). At 0° C., a solution of potassium hydrogenperoxomonosulphate (3.38 g, 5.49 mmol, 1.5 eq) in water (18 mL) was added thereto. The mixture was then gradually returned to 25° C., and stirred and reacted for 16 hours. After the reaction was completed, the reaction mixture was added with saturated sodium bicarbonate (15 mL) and sodium sulfite solution (15 mL) to quench, and extracted with ethyl acetate (30 mL*2). The organic phases were combined, washed with saturated sodium chloride solution, filtered, and the filtrate was dried over anhydrous sodium sulfate. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:0 to 100:2) to obtain a crude product. The crude product was stirred in methanol (5 mL) at 60° C. for 1 hour, and filtered. The filter cake was stirred in acetone (5 mL) at 60° C. for another 1 hour, and filtered. The filter cake was dried under vacuum to obtain compound WX-020. 1H NMR (400 MHz, DMSO-d6) δ: 10.79 (brs, 1H), 10.73 (brs, 1H), 9.09 (d, J=1.8 Hz, 1H), 8.64 (br d, J=4.5 Hz, 1H), 8.53 (s, 1H), 8.33 (dd, J=2.4, 8.4 Hz, 1H), 8.17 (d, J=8.4 Hz, 1H), 8.07 (s, 1H), 7.60-7.51 (m, 2H), 7.38-7.21 (m, 1H), 3.69-3.57 (m, 1H), 3.51 (s, 3H), 2.80 (d, J=4.4 Hz, 3H), 2.03-1.93 (m, 1H), 1.23 (d, J=6.8 Hz, 6H), 0.83-0.74 (m, 4H); LCMS m/z=524.0 [M+1]+.


Step 6: Synthesis of Sulfate of Compound WX-020

Compound WX-020 (0.4 g, 763.94 μmol, 1 eq) was added to acetone (4 mL), and the reaction mixture was stirred. Dilute sulfuric acid aqueous solution (0.25 M, 7.64 mL, 2.5 eq) was then added thereto. The reaction mixture was heated to 55° C., stirred for 30 minutes, and then naturally cooled to room temperature. The reaction mixture was filtered. The filter cake was washed with a small amount of ethyl acetate (10 mL), and dried under vacuum to obtain sulfate of WX-020. 1H NMR (400 MHz, DMSO-d6) δ: 11.33 (brs, 1H), 10.93 (brs, 1H), 9.11 (d, J=1.8 Hz, 1H), 8.87 (brs, 1H), 8.48 (s, 1H), 8.36 (dd, J=2.4, 8.4 Hz, 1H), 8.17 (d, J=8.4 Hz, 1H), 7.70 (br d, J=7.8 Hz, 1H), 7.61 (dd, J=1.2, 8.0 Hz, 1H), 7.39 (t, J=7.9 Hz, 1H), 3.61 (quin, J=6.8 Hz, 1H), 3.54 (s, 3H), 2.82 (d, J=4.4 Hz, 3H), 1.93-1.87 (m, 1H), 1.24 (d, J=6.8 Hz, 6H), 0.92-0.86 (m, 4H); LCMS m/z=523.9 [M+1]+.


Example 21



embedded image


embedded image


Step 1: Synthesis of Compound 21-1

Compound A-8 (300 mg, 1.05 mmol, 1 eq) and A-1 (261.21 mg, 1.05 mmol, 1 eq) were added to a mixed solvent of 1,4-dioxane (16 mL) and water (1 mL). Cesium carbonate (434.76 mg, 3.15 mmol, 3 eq) was then added thereto. After replacing with nitrogen, Pd(dppf)Cl2 (76.72 mg, 104.86 μmol, 0.1 eq) was finally added thereto. Under a nitrogen atmosphere, the reaction mixture was stirred at 90° C. for 6 hours. After cooling to room temperature, the reaction mixture was added with water (25 mL) and extracted with ethyl acetate (35 mL*2). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol:dichloromethane=0 to 30%) to obtain compound 21-1. LCMS m/z=329.1 [M+1]+.


Step 2: Synthesis of Compound 21-2

Compound 21-1 (90 mg, 274.10 μmol, 1 eq) and compound A-11 (56.47 mg, 274.10 mol, 1 eq) were dissolved in THE (10 mL). Under a nitrogen atmosphere, LiHMDS (1 M, 1.10 mL, 4 eq) was slowly added thereto at 0° C. The mixture was then warmed to 25° C. and stirred for 2 hours. After the reaction was completed, the reaction mixture was added with methanol (8 mL) and then concentrated under reduced pressure. The reaction mixture was then added with water (20 mL) and extracted with dichloromethane (30 mL*2). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol:dichloromethane=0 to 40%) to obtain compound 21-2. LCMS m/z=498.1 [M+1]+


Step 4: Synthesis of Compound WX-021

Compound 21-2 (50 mg, 100.42 μmol, 1 eq), cyclopropanecarboxamide (213.65 mg, 2.51 mmol, 25 eq), and Xantphos (8.72 mg, 15.06 μmol, 0.15 eq) were dissolved in a mixed solvent of 1.4-dioxane (6 mL) and NMP (1 mL). Cesium carbonate (98.16 mg, 301.26 μmol, 3 eq) was added thereto. The system was replaced with nitrogen. Pd2(dba)3 (13.79 mg, 15.06 mol, 0.15 eq) was finally added thereto. Under a nitrogen atmosphere, the mixture was stirred at 130° C. for 12 hours. After cooling to room temperature, the reaction mixture was added with water (10 mL) and extracted with dichloromethane (20 mL*2). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:0 to 30:1) to obtain compound WX-021. 1H NMR (400 MHz, CDCl3) δ: 11.15 (s, 1H), 9.13 (dd, J=1.4, 5.4 Hz, 1H), 8.89 (br s, 1H), 8.28 (s, 1H), 8.23-8.16 (m, 2H), 8.10 (br d, J=7.3 Hz, 1H), 7.66 (dd, J=1.4, 8.0 Hz, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.37-7.32 (m, 1H), 3.58 (s, 3H), 3.08 (d, J=5.0 Hz, 3H), 1.74-1.65 (m, 1H), 1.20-0.95 (m, 14H); LCMS m/z=547.1 [M+1]+.


To synthesize examples 22, 23, and 24, follow the synthesis steps of example 21, replacing A-8 in step 1 with fragments in the table below.













TABLE 8





Example
Compound
Fragment
Structural formula
Spectrum







22
WX-022


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 11.06 (s, 1H), 8.84 (dd, J = 1.1, 4.4 Hz, 1H), 8.73-8.63 (m, 1H), 8.17 (s, 1H), 8.11- 8.02 (m, 3H), 7.60-7.57 (m, 1H), 7.48-7.45 (m, 1H), 7.25 (t, J = 7.9 Hz, 1H), 3.48 (s, 3H), 2.99 (d, J = 5.0 Hz, 3H), 2.07-1.86 (m, 4H), 1.63-1.59 (m, 1H), 1.18-1.09 (m, 6H), 1.07- 1.03 (m, 2H), 0.89-0.84 (m, 2H); LCMS m/z = 523.2 [M + H]+.








A-7
WX-022






23
WX-023


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 11.14 (br s, 1H), 9.23 (br s, 1H), 8.34 (s, 1H), 8.21-8.11 (m, 1H), 7.79-7.73 (m, 4H), 7.54-7.48 (m, 1H), 7.31-7.23 (m, 1H), 7.22-7.18 (m, 1H), 3.43 (s, 3H), 3.07 (d, J = 5.0 Hz, 3H), 2.44- 2.31 (m, 2H), 1.81-1.64 (m, 1H), 1.30- 1.24 (m, 6H), 1.18-1.08 (m, 8H), 0.98-0.92 (m, 2H); LCMS m/z = 550.1 [M + H]+.








A-10
WX-023






24
WX-024


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 11.14 (s, 1H), 9.02 (br s, 1H), 8.32 (s, 1H), 8.16 (br d, J = 4.8 Hz, 1H), 7.93-7.87 (m, 2H), 7.74 (dd, J = 2.5, 8.3 Hz, 2H), 7.51 (dd, J = 1.4, 7.9 Hz, 1H), 7.31- 7.25 (m, 1H), 7.19 (dd, J = 1.5, 7.8 Hz, 1H), 3.45 (s, 3H), 3.07 (d, J = 5.0 Hz, 3H), 1.75-1.65 (m, 1H), 1.17-0.87 (m, 14H); LCMS m/z = 546.2 [M + H]+.








A-9
WX-024









To synthesize example 25 listed in Table 9, follow the synthesis steps of example 21, replacing A-8 in step 1 with A-6, and A-11 in step 2 with A-12.













TABLE 9







25
WX- 025


embedded image




embedded image



1H NMR (400 MHz, CDCl3) δ: 10.47 (s, 1H), 8.30-8.17 (m, 3H), 7.76 (d, J = 6.0 Hz, 4H), 7.59-7.53 (m, 1H), 7.26 (s, 1H), 7.13 (s, 1H), 6.34-6.21 (m, 1H), 3.45 (s, 3H), 3.04 (d, J = 4.8 Hz, 3H), 1.96 (br d, J = 9.8 Hz, 4H), 1.59-1.55 (m, 1H), 1.23-1.14 (m, 6H), 1.13-1.06 (m, 2H), 0.95-0.86 (m, 2H); LCMS m/z = 521.1 [M + H]+.








A-6
WX-025









Example 28



embedded image


embedded image


embedded image


Step 1: Synthesis of Compound 28-1

Compound 11-4 (100 mg, 228.61 μmol, 1 eq) was added to DMSO (1 mL), and then sodium sulfide (35.68 mg, 457.22 μmol, 19.18 μL, 2 eq) was added thereto. The reaction was carried out at 70° C. for 2 hours, and the mixture was cooled to room temperature to obtain a crude solution of compound 28-1. The crude solution was directly used in the next step without purification. LCMS m/z=452.1 [M+H]+.


Step 2: Synthesis of Compound 28-2

The crude DMSO solution of compound 28-1 (theoretical content of 108 mg, 228.10 mol, 1 eq) obtained in the previous step was dissolved in DMF (1 mL). Potassium carbonate (45.67 mg, 330.44 μmol, 1.45 eq) and 1-bromo-4-chloropropane (52.31 mg, 305.08 μmol, 35.11 μL, 1.34 eq) were added thereto, and the mixture was stirred at 25° C. for 10 minutes. The reaction mixture was added with water (3 mL) and extracted twice with ethyl acetate (6 mL). The organic phase was washed once with saturated brine (5 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 28-2. LCMS m/z=528.2 [M+H]+.


Step 3: Synthesis of Compound 28-3

Compound 28-2 (0.86 g, 1.63 mmol, 1 eq) was dissolved in dichloromethane (20 mL). 3-Chloroperoxybenzoic acid (330.66 mg, 1.63 mmol, purity of 85%, 1 eq) was added thereto at 0° C. The reaction was carried out at 25° C. for 0.5 hours. The reaction mixture was washed once with 10% sodium thiosulfate aqueous solution (20 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 28-3. LCMS m/z=544.2 [M+H]+.


Step 4: Synthesis of Compound 28-4

Compound 28-3 (0.27 g, 496.30 μmol, 1 eq) was dissolved in DMF (9 mL), and potassium tert-butoxide (111.38 mg, 992.60 μmol, 2 eq) was added thereto at 25° C. The mixture was stirred for 1 hour. The reaction mixture was added with saturated ammonium chloride aqueous solution (9 mL) and extracted with dichloromethane (15 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 28-4. LCMS m/z=508.1 [M+H]+.


Step 5: Synthesis of Compound 28-5

Compound 28-4 (0.15 g, 295.53 μmol, 1 eq) and trifluoroacetamide (33.41 mg, 295.53 mol, 1 eq) were dissolved in dichloromethane (7.5 mL). (Diacetoxyiodo)benzene (142.78 mg, 443.29 μmol, 1.5 eq), magnesium oxide (47.64 mg, 1.18 mmol, 13.31 μL, 4 eq), and rhodium acetate (26.12 mg, 59.11 μmol, 0.2 eq) were added thereto. The mixture was stirred at 40° C. for 1 hour. The reaction mixture was added with water (5 mL), and settled for phase separation. The organic phase was washed with 10% sodium thiosulfate aqueous solution (10 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by preparative thin-layer chromatography on a silica gel plate (dichloromethane:methanol=10:1) to obtain compound 28-5. LCMS m/z=619.1 [M+H]+.


Step 6: Synthesis of Compound WX-028

Compound 28-5 (10 mg, 16.17 μmol, 1 eq) was dissolved in methanol (1 mL), and potassium carbonate (2.23 mg, 16.17 μmol, 1 eq) was added thereto. The mixture was stirred at 25° C. for 10 minutes. The reaction mixture was added with water (1 mL) and extracted with dichloromethane (2 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by preparative thin-layer chromatography on a silica gel plate (dichloromethane:methanol=10:1) to obtain compound WX-028. 1H NMR (400 MHz, CDCl3) δ: 11.19 (s, 1H), 9.34 (s, 2H), 8.54-8.24 (m, 2H), 7.84-7.82 (m, 1H), 7.72 (d, J=7.2 Hz, 1H), 7.41-7.37 (m, 1H), 3.90 (s, 3H), 3.06 (d, J=4.8 Hz, 3H), 2.71-2.65 (m, 1H), 2.41-2.24 (m, 1H), 1.14-1.06 (m, 8H); LCMS m/z=523.2 [M+H]+.


Example 29



embedded image


embedded image


Step 1: Synthesis of Compound 29-1

The crude DMSO solution of compound 28-1 (theoretical content of 108 mg, 228.10 mol, 1 eq) was dissolved in DMF (1 mL). Potassium carbonate (45.67 mg, 330.44 μmol, 1.45 eq) and 1-bromo-4-chlorobutane (52.31 mg, 305.08 μmol, 35.11 μL, 1.34 eq) were added thereto, and the mixture was stirred at 25° C. for 10 minutes. The reaction mixture was added with water (3 mL) and extracted with ethyl acetate (6 mL*2). The organic phase was washed with saturated brine (5 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 29-1. LCMS m/z=542.2 [M+H]+.


Step 2: Synthesis of Compound 29-2

Compound 29-1 (120 mg, 221.38 μmol, 1 eq) was dissolved in methanol (5 mL), and (diacetoxyiodo)benzene (213.92 mg, 664.14 μmol, 3 eq) and ammonium acetate (51.19 mg, 664.14 μmol, 3 eq) were added thereto. The mixture was stirred at 25° C. for 10 minutes. The reaction mixture was extracted with dichloromethane (10 mL) and water (5 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 29-2. LCMS m/z=573.2 [M+H]+.


Step 5: Synthesis of Hydrochloride of Compound WX-29

Compound 29-2 (35 mg, 61.08 μmol, 1 eq) was dissolved in DMF (1 mL), and sodium hydride (9.77 mg, 244.30 μmol, purity of 60%, 4 eq) was added thereto at 0° C. The mixture was stirred at 25° C. for 0.5 hours. The reaction mixture was added with saturated ammonium chloride aqueous solution (2 mL), then added with water (1 mL), and extracted with dichloromethane (3 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 10% to 40%, 8 minutes) to obtain hydrochloride of compound WX-29. 1HNMR (400 MHz, CD3OD) δ: 9.64 (s, 2H), 8.12-8.09 (m, 1H), 7.83-7.81 (m, 1H), 7.51 (t, J=8.0 Hz, 1H), 7.10 (s, 1H), 4.31-4.21 (m, 1H), 4.12-4.08 (m, 1H), 3.89 (s, 3H), 3.85-3.73 (m, 2H), 3.00 (s, 3H), 2.66-2.55 (m, 2H), 2.26-2.15 (m, 1H), 2.09-2.02 (m, 1H), 1.92-1.84 (m, 1H), 1.16-1.05 (m, 4H); LCMS m/z=537.3 [M+H]+.


Example 30



embedded image


embedded image


Step 1: Synthesis of Compound 30-1

Compound 11-1 (1.24 g, 9.36 mmol, 1.16 mL, 1 eq), A-13 (2.5 g, 9.36 mmol, 1 eq), and potassium carbonate (2.59 g, 18.72 mmol, 2 eq) were dissolved in a mixed solvent of 1,4-dioxane (80 mL) and water (15 mL). After replacing with nitrogen three times, Pd(dppf)Cl2 (684.86 mg, 936.00 μmol, 0.1 eq) was added thereto. Under a nitrogen atmosphere, the mixture was heated to 80° C. and stirred for 4 hours. After cooling to room temperature, the reaction mixture was added with saturated ammonium chloride aqueous solution (100 mL) and water (100 mL), and then extracted with ethyl acetate (100 mL*2). The organic phase was washed with saturated brine (100 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=10 to 30%) to obtain compound 30-1. LCMS m/z=238.0 [M+1]+.


Step 2: Synthesis of Compound 30-2

Under a nitrogen atmosphere, compound 30-1 (1.8 g, 3.79 mmol, purity of 50%, 1 eq) and A-12 (777.96 mg, 3.79 mmol, 1 eq) were added to THE (10 mL), and the mixture was stirred until dissolved. The mixture was cooled to 0° C., and LiHMDS (1 M, 9.49 mL, 2.5 eq) was added dropwise thereto. The mixture was then returned to 25° C. and stirred for another 2 hours. After the reaction was completed, the reaction mixture was added with water (100 mL) and extracted with ethyl acetate (50 mL*2). The organic phases were combined, washed with saturated brine (50 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=3%) to obtain compound 30-2. LCMS m/z=406.0 [M+1]+.


Step 3: Synthesis of Compound 30-3

Compound 30-2 (1.18 g, 2.91 mmol, 1 eq) and cyclopropanecarboxamide (6.19 g, 72.70 mmol, 25 eq) were added to 1,4-dioxane (80 mL) and NMP (2 mL). Cesium carbonate (2.84 g, 8.72 mmol, 3 eq) and Xantphos (252.39 mg, 436.19 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (399.43 mg, 436.19 μmol, 0.15 eq) was added. The mixture was refluxed and reacted at 130° C. for 18 hours. After the reaction was completed, the reaction mixture was added with water (100) and extracted with ethyl acetate (50 mL*2). The organic phases were combined, washed with saturated sodium chloride aqueous solution (50 mL*2), dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=3%) to obtain compound 30-3. LCMS m/z=455.1 [M+1]+.


Step 4: Synthesis of Compound 30-4

30-3 (200 mg, 440.11 μmol, 1 eq) was dissolved in DMF (10 mL), and sodium 2-propanethiolate (86.39 mg, 880.23 μmol, 2 eq) was added thereto. The reaction was carried out at 40° C. for 16 hours. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, and concentrated under vacuum to obtain a crude product of compound 30-4. The crude product was directly used in the next step without purification. LCMS m/z=511.2 [M+1]+.


Step 5: Synthesis of Compound WX-030

Compound 30-4 (200 mg, 391.71 μmol, 1 eq) was added to a mixed solution of ethanol (10 mL) and water (5 mL), and then potassium hydrogenperoxomonosulphate (361.21 mg, 587.56 μmol, 1.5 eq) was added thereto. The reaction was carried out at 25° C. for 2 hours. After the reaction was completed, the reaction mixture was added with saturated sodium bisulfite aqueous solution (10 mL), and extracted with ethyl acetate (20 mL*2). The organic phase was washed with saturated brine (10 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100*40 mm*3 m; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 13% to 43%, 8 minutes). The obtained solution was concentrated under vacuum at 40° C. to remove acetonitrile, then adjusted to alkalinity (pH=8) with saturated sodium bicarbonate, and extracted with dichloromethane (30 mL*3). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain compound WX-030. 1H NMR (400 MHz, CDCl3) δ: 10.47 (s, 1H), 9.17 (s, 2H), 8.53 (br s, 1H), 8.22 (s, 1H), 8.12 (s, 1H), 7.38 (d, J=9.0 Hz, 2H), 6.31-6.23 (m, 1H), 3.80 (s, 3H), 3.28-3.18 (m, 1H), 2.94 (d, J=4.8 Hz, 3H), 1.51-1.44 (m, 1H), 1.34 (d, J=6.8 Hz, 6H), 1.12-0.96 (m, 2H), 0.87-0.80 (m, 2H); LCMS m/z=543.2 [M+1]+.


Example 31



embedded image


embedded image


Step 1: Synthesis of Compound 31-1

Compound 11-2 (2 g, 9.12 mmol, 1 eq) and sodium 2-propanethiolate (906.59 mg, 9.24 mmol, 1.5 eq) were dissolved in DMF (20 mL), and the mixture was reacted at 40° C. for 1 hour. The reaction mixture was added with ethyl acetate (20 mL) and water (20 mL). The aqueous phase was then extracted once with ethyl acetate (20 mL), and the organic phase was washed three times with saturated sodium chloride aqueous solution (20 mL), dried over anhydrous sodium chloride, and concentrated under reduced pressure to obtain compound 31-1. LCMS m/z=276.1 [M+H]+.


Step 2: Synthesis of Compound 31-2

Compound 31-1 (0.8 g, 2.91 mmol, 1 eq) and compound A-3-2 (577.90 mg, 2.91 mmol, 1 eq) were dissolved in THE (16 mL), and LiHMDS (1 mol/L, 8.72 mL, 3 eq) was added thereto at −60° C. The mixture was warmed to 25° C. and reacted for 3 hours. The reaction mixture was added with methanol (5 mL) to quench, then added with 2 mol/L hydrochloric acid solution to adjust the pH to 7 to 8, and added with ethyl acetate (20 mL) and water (20 mL). The aqueous phase was then extracted once with ethyl acetate (20 mL). The organic phase was washed once with saturated sodium chloride aqueous solution (20 mL), dried over anhydrous sodium sulfate, and concentrated. The obtained crude product was subjected to silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 31-2. LCMS m/z=432.1 [M+H]+.


Step 3: Synthesis of Compound 31-3

Compound 31-2 (0.46 g, 1.07 mmol, 1 eq) and cyclopropanecarboxamide (906.42 mg, 10.65 mmol, 10 eq) were dissolved in 1,4-dioxane (16 mL). Cesium carbonate (867.55 mg, 2.66 mmol, 2.5 eq) was then added thereto. Under a nitrogen atmosphere, Pd2(dba)3 (30.53 mg, 33.34 μmol, 0.1 eq) and Xantphos (38.58 mg, 66.68 μmol, 0.2 eq) were added thereto. The reaction was carried out at 120° C. for 4 hours. The reaction system was cooled to 20 to 30° C. The reaction mixture was added with 2 mol/L hydrochloric acid solution to adjust the pH to 3, and added with dichloromethane (20 mL) and water (20 mL). The aqueous phase was then extracted once with dichloromethane (20 mL). The organic phase was washed once with saturated sodium chloride aqueous solution (20 mL), dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. The obtained crude product was subjected to silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 31-3. LCMS m/z=481.0 [M+H]+.


Step 4: Synthesis of Compound 31-4

Compound 30-3 (0.42 g, 524.41 μmol, purity of 60%, 1 eq) was dissolved in MeOH (10 mL). Ammonium acetate (160.41 mg, 2.08 mmol, 4 eq) and (diacetoxyiodo)benzene (502.71 mg, 1.56 mmol, 3 eq) were then added thereto, and the reaction was carried out at 25° C. for 4 hours. The reaction mixture was added with ethyl acetate (10 mL) and water (10 mL). The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phase was washed with saturated sodium chloride aqueous solution (10 mL*3), dried over anhydrous sodium sulfate, and then concentrated. Compound 31-4 was obtained. LCMS m/z=512.2 [M+H]+. The crude product was directly used in the next step without purification.


Step 5: Synthesis of Hydrochloride of Compound WX-031

Compound 31-4 (450 mg, 175.94 μmol, purity of 20%, 1 eq) and ammonium chloride (470.55 mg, 8.80 mmol, 50 eq) were dissolved in DMF (10 mL). HATU (200.69 mg, 527.81 mol, 3 eq) and DIPEA (27.29 mg, 211.12 μmol, 36.77 μL, 1.2 eq) were then added thereto. The reaction was carried out at 25° C. for 3 hours. The reaction mixture was added with ethyl acetate (10 mL) and water (10 mL). The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phase was washed with saturated sodium chloride aqueous solution (10 mL*3), dried over anhydrous sodium sulfate, and then concentrated. The obtained crude product was subjected to silica gel column chromatography (methanol/dichloromethane=0 to 10%), and then separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Luna 80*30 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 10% to 35%) to obtain hydrochloride of compound WX-031. 1H NMR (400 MHz, CD3OD) δ: 9.47 (s, 2H), 8.10 (dd, J=0.8, 8.0 Hz, 1H), 7.78 (dd, J=0.9, 7.9 Hz, 1H), 7.50 (t, J=8.0 Hz, 1H), 6.91 (s, 1H), 4.12-4.04 (m, 1H), 3.88 (s, 3H), 1.86-1.80 (m, 1H), 1.52 (dd, J=8.0, 18.8 Hz, 6H), 1.18-1.08 (m, 4H); LCMS m/z=511.1 [M+H]+.


Example 32



embedded image


embedded image


Step 1: Synthesis of Compound 32-1

Compound 12-2 (1.12 g, 5.13 mmol, 1 eq) was dissolved in DMF (10 mL), and then sodium 2-propanethiolate (2.01 g, 20.53 mmol, 4 eq) was added thereto. The reaction was carried out at 40° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=10%) to obtain compound 32-1. LCMS m/z=275.1 [M+1]+.


Step 2: Synthesis of Compound 32-2

Under a nitrogen atmosphere, compound 32-1 (250 mg, 911.14 μmol, 1 eq) and A-3-2 (181.25 mg, 911.14 μmol, 1 eq) were added to THF (10 mL). The mixture was stirred at 25° C., and LiHMDS(1 M, 2.28 mL, 2.5 eq) was slowly added dropwise thereto. The mixture was stirred at 25° C. for another 2 hours. The reaction mixture was added with saturated ammonium chloride aqueous solution (20 mL) and water (50 mL), and extracted with ethyl acetate (50 mL*2). The organic phases were combined, washed with saturated brine (50 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=10%) to obtain compound 32-2. LCMS m/z=431.0 [M+1]+.


Step 3: Synthesis of Compound 32-3

Compound 32-2 (240 mg, 556.96 μmol, 1 eq) and cyclopropanecarboxamide (1.19 g, 13.92 mmol, 25 eq) were added to a mixed solvent of 1,4-dioxane (10 mL) and NMP (1 mL). Cesium carbonate (544.41 mg, 1.67 mmol, 3 eq) and Xantphos (48.34 mg, 83.54 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (76.50 mg, 83.54 μmol, 0.15 eq) was added. The mixture was stirred and reacted at 130° C. for 18 hours. After cooling to room temperature, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (30 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=3%) to obtain compound 32-3. LCMS m/z=480.1 [M+1]+.


Step 4: Synthesis of Compound 32-4

Compound 32-3 (187 mg, 389.95 μmol, 1 eq) was added to methanol (1 mL), and then (diacetoxyiodo)benzene (376.80 mg, 1.17 mmol, 3 eq) and ammonium acetate (120.23 mg, 1.56 mmol, 4 eq) were added thereto. The reaction was carried out at 25° C. for 2 hours. The reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution (10 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product of 32-4. LCMS m/z=511.1 [M+1]+. The crude product was directly used in the next step without purification.


Step 5: Synthesis of Hydrochloride of Compound WX-032

Compound 32-4 (199 mg, 389.76 μmol, 1 eq) was dissolved in DMF (10 mL). Ammonium chloride (1.04 g, 19.49 mmol, 50 eq), HATU (222.30 mg, 584.65 μmol, 1.5 eq), and DIPEA (151.12 mg, 1.17 mmol, 203.67 μL, 3 eq) were then sequentially added thereto. The reaction was carried out at 25° C. for 1 hour. The reaction system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution (10 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Xtimate C18, 150*40 mm*3 m; mobile phase: A (water containing 0.04% hydrochloric acid) and B (acetonitrile); gradient: B %: 15% to 45%, 10 minutes) to obtain hydrochloride of compound WX-032. 1H NMR (400 MHz, DMSO-d6) δ: 11.49 (brs, 1H), 11.12 (brs, 1H), 9.27-9.08 (m, 1H), 8.67-8.52 (m, 1H), 8.48-8.36 (m, 1H), 8.28-8.17 (m, 1H), 8.12-8.08 (m, 1H), 7.98-7.92 (m, 1H), 7.73-7.59 (m, 2H), 7.44-7.36 (m, 1H),), 3.97-3.84 (m, 1H), 3.54-3.52 (m, 3H), 2.16-2.00 (m, 1H), 1.41-1.25 (m, 6H), 0.93-0.78 (m, 4H); LCMS m/z=510.1[M+1]+.


Example 33



embedded image


embedded image


Step 1: Synthesis of Compound 33-1

Under a nitrogen atmosphere, compound A-2-1 (627.52 mg, 3.27 mmol, 1 eq) and 31-1 (900 mg, 3.27 mmol, 1 eq) were added to tetrahydrofuran (10 mL), and the mixture was stirred until dissolved. LiHMDS (1 M, 8.17 mL, 2.5 eq) was slowly added dropwise at 25° C., and then the mixture was stirred at 25° C. for another 1 hour. After the reaction was completed, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (30 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 33-1. 1H NMR (400 MHz, DMSO-d6) δ: 11.39 (br s, 1H), 8.94 (s, 2H), 8.65 (s, 1H), 7.563-7.55 (m, 2H), 7.31 (t, J=8.0 Hz, 1H), 6.85 (s, 1H), 3.77-3.69 (m, 1H), 3.66 (s, 3H), 1.31 (d, J=6.8 Hz, 6H); LCMS m/z=431.1 [M+1]+.


Step 2: Synthesis of Compound 33-2

Compound 33-1 (500 mg, 1.16 mmol, 1 eq), cyclopropanecarboxamide (2.47 g, 29.01 mmol, 25 eq), and Xantphos (201.42 mg, 348.10 μmol, 0.3 eq) were sequentially added to a mixed solvent of NMP (1 mL) and 1,4-dioxane (10 mL). The reaction mixture was replaced with nitrogen three times, and cesium carbonate (1.51 g, 4.64 mmol, 4 eq) and Pd2(dba)3·CHCl3 (180.16 mg, 174.05 μmol, 0.15 eq) were added thereto. After replacing with nitrogen three times, the reaction mixture was heated to reflux (external temperature of 130° C.) and reacted for 16 hours. After cooling to room temperature, the reaction mixture was added with water (20 mL) and ethyl acetate (20 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=0 to 10%) to obtain compound 33-2. LCMS m/z=480.1 [M+1]+.


Step 3: Synthesis of Compound 33-3

Compound 33-2 (150 mg, 312.79 μmol, 1 eq) was dissolved in DCM (2 mL). 3-Chloroperoxybenzoic acid (80.97 mg, 469.19 μmol, 1.5 eq) was added thereto. The reaction was carried out at 20° C. for 16 hours. After the reaction was completed, the reaction mixture was added with water (20 mL) and ethyl acetate (20 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product of 33-3. LCMS m/z=512.1 [M+1]+. The crude product was directly used in the next step without purification.


Step 4: Synthesis of Compound WX-033

Compound 33-3 (140 mg, 273.68 μmol, 1 eq) and ammonium chloride (731.97 mg, 13.68 mmol, 50 eq) were added to DMF (10 mL). HATU (312.18 mg, 821.04 μmol, 3 eq) and DIPEA (212.23 mg, 1.64 mmol, 286.02 μL, 6 eq) were sequentially added thereto. The mixture was stirred and reacted at 25° C. for 1 hour. The reaction system was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100*40 mm*3 m; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 10% to 40%, 8 minutes). The obtained solution was concentrated under vacuum at 40° C. to remove acetonitrile, then adjusted to alkalinity (pH=8) with saturated sodium bicarbonate, and extracted with dichloromethane (30 mL*3). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain compound WX-033. 1H NMR (400 MHz, CDCl3) δ: 10.67 (br s, 1H), 9.27 (s, 2H), 8.38 (s, 1H), 8.16-8.04 (m, 1H), 7.82 (d, J=8.4 Hz, 1H), 7.73 (d, J=6.8 Hz, 1H), 7.40-7.34 (m 1H), 5.83 (brs, 2H), 3.85 (s, 2H), 3.38-3.24 (m, 1H), 1.74-1.65 (m, 1H), 1.44 (d, J=6.8 Hz, 6H), 1.13-1.06 (m, 2H), 0.95-0.89 (m, 2H); LCMS m/z=511.2 [M+1]+.


Example 34



embedded image


embedded image


Step 1: Synthesis of Compound 34-1

Under a nitrogen atmosphere, 32-1 (250 mg, 911.14 μmol, 1 eq) was added to THF (10 mL), then A-2-1 (174.94 mg, 911.14 μmol, 1 eq) was added thereto, and the mixture was stirred until dissolved. LiHMDS (1 M, 2.28 mL, 2.5 eq) was added dropwise thereto at 25° C., and the mixture was stirred for another 2 hours. After the reaction was completed, the reaction mixture was added with saturated ammonium chloride aqueous solution (20 mL) and water (50 mL), and extracted with ethyl acetate (50 mL*2). The organic phases were combined, washed with saturated brine (50 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=10%) to obtain compound 34-1. LCMS m/z=430.0 [M+1]+.


Step 2: Synthesis of Compound 34-2

Compound 34-1 (550 mg, 1.28 mmol, 1 eq) and cyclopropanecarboxamide (2.72 g, 31.98 mmol, 25 eq) were added to a mixed solvent of 1,4-dioxane (20 mL) and NMP (2 mL). Cesium carbonate (1.25 g, 3.84 mmol, 3 eq) and Xantphos (111.04 mg, 191.90 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (175.72 mg, 191.90 μmol, 0.15 eq) was added. The mixture was reacted at 130° C. for 18 hours. After the reaction was cooled to room temperature, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (30 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/dichloromethane=3%) to obtain compound 34-2. LCMS m/z=479.2 [M+1]+.


Step 3: Synthesis of Compound 34-3

Compound 34-2 (430 mg, 898.52 μmol, 1 eq) was dissolved in dichloromethane (10 mL). The mixture was then cooled to 0° C. and added with 3-chloroperoxybenzoic acid (364.84 mg, 1.80 mmol, purity of 85%, 2 eq). The reaction was then carried out at 25° C. for 2 hours. After the reaction was completed, the reaction mixture was added with water (30 mL) and ethyl acetate (30 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (30 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product of 34-3. LCMS m/z=511.1 [M+1]+. The crude product was directly used in the next step without further purification.


Step 4: Synthesis of Trifluoroacetate of Compound WX-034

Compound 34-3 (458 mg, 897.05 μmol, 1 eq) was added to DMF (10 mL). Ammonium chloride (2.40 g, 44.85 mmol, 50 eq), HATU (511.63 mg, 1.35 mmol, 1.5 eq), and DIPEA (347.81 mg, 2.69 mmol, 468.74 μL, 3 eq) were then added thereto. The reaction was carried out at 25° C. for 1 hour. After the reaction was completed, the reaction mixture was added with water (10 mL) and ethyl acetate (10 mL) for extraction and phase separation. The aqueous phase was then extracted once with ethyl acetate (10 mL). The organic phases were combined, washed once with saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100*40 mm*3 m; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 15 to 45%, 8 minutes) to obtain trifluoroacetate of compound WX-034. 1H NMR (400 MHz, DMSO-d6) δ: 11.34-10.93 (m, 2H), 9.22-9.00 (m, 1H), 8.59 (s, 1H), 8.50-8.23 (m, 2H), 8.17 (d, J=8.4 Hz, 1H), 7.77-7.56 (m, 4H), 7.42-7.30 (m, 1H), 3.65-3.58 (m, 1H), 3.52 (s, 3H), 2.01-1.87 (m, 1H), 1.24 (d, J=6.8 Hz, 6H), 0.93-0.79 (m, 4H); LCMS m/z=510.1 [M+1]+.


Example 35



embedded image


embedded image


Step 1: Synthesis of Compound 35-1

Compound 21-1 (100 mg, 304.56 μmol, 1 eq) and A-3-2 (60.58 mg, 304.56 μmol, 1 eq) were dissolved in THE (10 mL). LiHMDS (1 M, 1.22 mL, 4 eq) was added dropwise thereto at 0° C., and then the mixture was stirred and reacted at 25° C. for 2 hours. The reaction mixture was added with 8 mL of methanol, concentrated under reduced pressure, then added with water (20 mL), and extracted with dichloromethane (30 mL*2). The organic phase was washed with saturated sodium chloride solution (25 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:0 to 100:5) to obtain compound 35-1. LCMS m/z=485.0 [M+1]+.


Step 4: Synthesis of Compound 35-2

Compound 35-1 (50 mg, 103.12 μmol, 1 eq), cyclopropanecarboxamide (219.40 mg, 2.58 mmol, 25 eq), and Xantphos (8.95 mg, 15.47 μmol, 0.15 eq) were dissolved in a mixed solvent of 1,4-dioxane (1 mL) and NMP (0.5 mL). Cesium carbonate (100.80 mg, 309.36 μmol, 3 eq) was then added thereto. After replacing with nitrogen, Pd2(dba)3 (14.16 mg, 15.47 μmol, 0.15 eq) was added thereto. After replacing with nitrogen, the reaction mixture was heated to reflux and reacted for 12 hours. After the reaction was completed, the reaction mixture was cooled to room temperature, and concentrated under reduced pressure to remove the organic solvent. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:0 to 1:1) to obtain compound 35-2. LCMS m/z=534.1 [M+1]+.


Step 5: Synthesis of Trifluoroacetate of Compound WX-035

Compound 35-2 (30 mg, 56.23 μmol, 1 eq), ammonium chloride (150.39 mg, 2.81 mmol, 50 eq), and HATU (32.07 mg, 84.35 μmol, 1.5 eq) were dissolved in DMF (2 mL). DIPEA (21.80 mg, 168.69 μmol, 29.38 μL, 3 eq) was then added thereto. The mixture was stirred and reacted at 25° C. for 12 hours. After the reaction was completed, the reaction mixture was added with water (6 mL) and extracted with ethyl acetate (10 mL*2). The organic phase was washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (chromatographic column: Welch Xtimate C18 100*40 mm*3 m; mobile phase: A (water containing 0.075% trifluoroacetic acid) and B (acetonitrile); gradient: B %: 8 to 38%, 8 minutes) to obtain trifluoroacetate of compound WX-035. 1H NMR (400 MHz, CDCl3) δ: 13.05 (br s, 1H), 11.53 (s, 1H), 9.06 (d, J=4.4 Hz, 1H), 8.40 (s, 1H), 8.19-8.13 (m, 1H), 7.96 (br d, J=8.0 Hz, 1H), 7.71 (d, J=7.8 Hz, 1H), 7.61 (br s, 1H), 7.45 (d, J=6.8 Hz, 1H), 7.35-7.30 (m, 1H), 5.81 (br s, 1H), 3.48 (s, 3H), 2.00-1.93 (m, 1H), 1.06-0.87 (m, 14H); LCMS m/z=533.2 [M+1]+.


Example 36



embedded image


embedded image


embedded image


Step 1: Synthesis of Compound 36-1

Compound A-1 (2 g, 8.03 mmol, 1 eq), compound A-14 (1.36 g, 7.23 mmol, 0.9 eq), and potassium phosphate tribasic (5.11 g, 24.09 mmol, 3 eq) were dissolved in 1,4-dioxane (20 mL) and water (10 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (393.38 mg, 481.71 μmol, 0.06 eq) was added. The mixture was stirred at 100° C. for 2 hours. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (20 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 36-1. 1HNMR (400 MHz, CDCl3) δ: 7.83-7.80 (m, 1H), 7.30-7.29 (m, 1H), 7.28-7.27 (m, 1H), 7.08-7.04 (m, 1H), 6.87 (dd, J=8.0, 1.6 Hz, 1H), 4.39-4.33 (m, 1H), 3.53 (s, 3H), 1.51 (d, J=6.8 Hz, 6H); LCMS m/z=276.1 [M+H]+.


Step 2: Synthesis of Compound 36-2

Compound 36-1 (1 g, 3.63 mmol, 1 eq) and compound A-3-2 (830.74 mg, 4.18 mmol, 1.15 eq) were dissolved in isopropanol (10 mL) and water (3 mL). Zinc acetate (799.56 mg, 4.36 mmol, 1.2 eq) was added thereto. The mixture was stirred at 80° C. for 16 hours. The reaction mixture was added with water (30 mL), filtered, and the filter cake was collected to obtain compound 36-2. LCMS m/z=432.2 [M+H]+.


Step 3: Synthesis of Compound 36-3

Compound 36-2 (1 g, 2.32 mmol, 1 eq), cyclopropanecarboxamide (788.19 mg, 9.26 mmol, 4 eq), cesium carbonate (1.51 g, 4.63 mmol, 2 eq), and Xantphos (133.97 mg, 231.54 mol, 0.1 eq) were dissolved in dioxane (10 mL). After replacing with nitrogen three times, Pd2(dba)3 (212.02 mg, 231.54 μmol, 0.1 eq) was added. The mixture was stirred at 120° C. for 3 hours. The reaction mixture was added with water (10 mL) and extracted with dichloromethane (10 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was added with tert-butyl methyl ether (20 mL), stirred at 20° C. for 1 hour, and then filtered. The filter cake was collected to obtain compound 36-3. LCMS m/z=481.3 [M+H]+.


Step 4: Synthesis of Compound 36-4

Compound 36-3 (1.42 g, 2.96 mmol, 1 eq) was dissolved in NMP (10 mL) and acetonitrile (5 mL). 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (793.08 mg, 4.14 mmol, 1.4 eq), 1-hydroxybenzotriazole (199.65 mg, 1.48 mmol, 0.5 eq), methylamine hydrochloride (199.52 mg, 2.96 mmol, 1 eq), and methylimidazole (727.85 mg, 8.87 mmol, 706.65 μL, 3 eq) were then added thereto. The mixture was stirred at 65° C. for 1 hour. The reaction mixture was added with water (10 mL) and extracted twice with dichloromethane (10 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 36-4. LCMS m/z=494.2 [M+H]+.


Step 5: Synthesis of Compounds WX-036A and WX-036B

Compound 36-4 (0.7 g, 1.42 mmol, 1 eq) was dissolved in methanol (14 mL). (Diacetoxyiodo)benzene (1.14 g, 3.55 mmol, 2.5 eq) and ammonium acetate (273.30 mg, 3.55 mmol, 2.5 eq) were added thereto, and the mixture was stirred at 15° C. for 2 hours. The reaction mixture was added with water (15 mL) and extracted with dichloromethane (15 mL*2). The organic phase was washed with 5% sodium thiosulfate aqueous solution (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/ethyl acetate=0 to 10%) to obtain compound WX-036. Compound WX-036 was subjected to resolution by SFC (column: Chiralpak UH (250 mm*30 mm, 10 m); mobile phase: A (CO2) and B (ethanol containing 0.1% ammonia water); gradient: B %=50% to 50%, 15 minutes) to obtain compound WX-036A and compound WX-036B.


WX-036A: 1HNMR (400 MHz, CDCl3) δ: 11.14 (s, 1H), 8.94 (s, 1H), 8.40-8.38 (m, 1H), 8.27-8.23 (m, 2H), 8.19-8.14 (m, 1H), 7.85-7.83 (m, 1H), 7.62 (dd, J=8.0, 1.6 Hz, 1H), 7.42-7.38 (m, 1H), 4.08-4.01 (m, 2H), 3.56 (s, 3H), 3.07 (d, J=5.2 Hz, 3H), 1.75-1.69 (m, 1H), 1.48 (d, J=7.0 Hz, 3H), 1.42 (d, J=6.8 Hz, 3H), 1.15-1.11 (m, 2H), 0.98-0.93 (m, 2H); LCMS m/z=525.2 [M+H]+. SFC (column: Chiralpak IH-3, 3 μm, 0.46 cm id×5 cm L; mobile phase: A (CO2) and B (EtOH containing 0.1% isopropylamine); gradient: B %=5 to 50%, 3 minutes; flow rate: 3.4 mL/min; wavelength: 220 nm; pressure: 100 bar, Rt=1.615 min) with a chiral isomer excess of 100%.


WX-036B: 1HNMR (400 MHz, CDCl3) δ: 11.14 (s, 1H), 8.92 (s, 1H), 8.40-8.38 (m, 1H), 8.27-8.22 (m, 2H), 8.19-8.17 (m, 1H), 7.84 (dd, J=8.0, 1.6 Hz, 1H), 7.62 (dd, J=8.0, 1.6 Hz, 1H), 7.42-7.38 (m, 1H), 4.08-4.01 (m, 1H), 3.56 (s, 3H), 3.07 (d, J=5.2 Hz, 3H), 1.74-1.69 (m, 2H), 1.48 (d, J=6.8 Hz, 3H), 1.42 (d, J=7.2 Hz, 3H), 1.15-1.11 (m, 2H), 0.98-0.94 (m, 2H); LCMS m/z=525.2 [M+H]+. SFC (column: Chiralpak IH-3, 3 μm, 0.46 cm id×5 cm L; mobile phase: A (CO2) and B (EtOH containing 0.1% isopropylamine); gradient: B %=5 to 50%, 3 minutes; flow rate: 3.4 mL/min; wavelength: 220 nm; pressure: 100 bar, Rt=1.771 min) with a chiral isomer excess of 99.38%.


Example 37



embedded image


embedded image


embedded image


Step 1: Synthesis of Compound 37-1

Compound A-1 (2 g, 8.03 mmol, 1 eq), compound A-15 (1.12 g, 4.82 mmol, 0.6 eq), and potassium phosphate tribasic (5.11 g, 24.09 mmol, 3 eq) were dissolved in dioxane (20 mL) and water (10 mL). After replacing with nitrogen three times, Pd(dppf)Cl2·CH2Cl2 (393.38 mg, 481.71 μmol, 0.06 eq) was added. The mixture was stirred at 100° C. for 1.5 hours. The reaction mixture was added with water (10 mL) and extracted twice with ethyl acetate (20 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 37-1. 1HNMR (400 MHz, CDCl3) δ: 8.67-8.65 (m, 1H), 7.76 (dd, J=8.4, 2.4 Hz, 1H), 7.23-7.21 (m, 1H), 7.02-6.98 (m, 1H), 6.79-6.71 (m, 2H), 4.08-3.98 (m, 3H), 3.44 (s, 3H), 1.45 (d, J=6.8 Hz, 6H); LCMS m/z=275.0 [M+H]+.


Step 2: Synthesis of Compound 37-2

Compound 37-1 (950 mg, 3.46 mmol, 1 eq) and compound A-3-2 (757.60 mg, 3.81 mmol, 1.1 eq) were dissolved in isopropanol (9.5 mL) and water (3.2 mL). Zinc acetate (762.32 mg, 4.15 mmol, 1.2 eq) was then added thereto. The mixture was stirred at 80° C. for 16 hours. The reaction mixture was added with water (30 mL), filtered, and the filter cake was collected to obtain compound 37-2. LCMS m/z=431.0 [M+H]+.


Step 3: Synthesis of Compound 37-3

Compound 37-2 (1 g, 2.32 mmol, 1 eq), cyclopropanecarboxamide (790.00 mg, 9.28 mmol, 4 eq), and cesium carbonate (1.51 g, 4.64 mmol, 2 eq) were dissolved in dioxane (10 mL). After replacing with nitrogen three times, Xantphos (134.28 mg, 232.07 μmol, 0.1 eq) and Pd2(dba)3 (212.51 mg, 232.07 μmol, 0.1 eq) were added. The mixture was stirred at 120° C. for 3 hours. The reaction mixture was added with water (10 mL) and extracted with dichloromethane (10 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was added with tert-butyl methyl ether (20 mL), stirred at 20° C. for 1 hour, and then filtered. The filter cake was collected to obtain compound 37-3. LCMS m/z=480.3 [M+H]+.


Step 4: Synthesis of Compound 37-4

Compound 37-3 (1.06 g, 2.21 mmol, 1 eq) was dissolved in NMP (10 mL) and acetonitrile (5 mL). 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (593.23 mg, 3.09 mmol, 1.4 eq), 1-hydroxybenzotriazole (149.33 mg, 1.11 mmol, 0.5 eq), methylamine hydrochloride (149.24 mg, 2.21 mmol, 1 eq), and N-methylimidazole (544.42 mg, 6.63 mmol, 528.57 μL, 3 eq) were then added thereto. The mixture was stirred at 65° C. for 1 hour. The reaction mixture was added with water (10 mL) and extracted with dichloromethane (10 mL*2). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0 to 50%) to obtain compound 37-4. LCMS m/z=493.2 [M+H]+.


Step 5: Synthesis of Compounds WX-037A and WX-037B

Compound 37-4 (0.9 g, 1.83 mmol, 1 eq) was dissolved in methanol (18 mL). (Diacetoxyiodo)benzene (1.47 g, 4.57 mmol, 2.5 eq) and ammonium acetate (352.08 mg, 4.57 mmol, 2.5 eq) were added thereto, and the mixture was stirred at 15° C. for 2 hours. The reaction mixture was added with water (15 mL) and extracted with dichloromethane (15 mL*2). The organic phase was washed with 5% sodium thiosulfate aqueous solution (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (methanol/ethyl acetate=0 to 10%) to obtain compound WX-037. Compound WX-037 was subjected to resolution by SFC (column: Chiralpak UH (250 mm*30 mm, 10 m); mobile phase: A (CO2) and B (methanol containing 0.1% ammonia water); gradient: B %=45% to 45%, 10 minutes) to obtain WX-037A and WX-037B.


WX-037A: 1HNMR (400 MHz, CDCl3) δ: 11.14 (s, 1H), 8.97-8.96 (m, 1H), 8.84-8.75 (m, 1H), 8.26 (s, 1H), 8.22-8.16 (m, 3H), 7.55 (dd, J=8.0, 1.6 Hz, 1H), 7.35-7.31 (m, 1H), 7.23-7.21 (m, 1H), 3.83-3.76 (m, 1H), 3.46 (s, 3H), 3.07 (d, J=5.2 Hz, 3H), 1.73-1.69 (m, 1H), 1.42 (d, J=7.2 Hz, 3H), 1.36 (d, J=6.8 Hz, 3H), 1.15-1.12 (m, 2H), 0.99-0.94 (m, 2H); LCMS m/z=524.2 [M+H]+. SFC (column: Chiralpak IH-3, 3 μm, 0.46 cm id×10 cm L; mobile phase: A (CO2) and B (MeOH containing 0.1% isopropylamine); gradient: B %=10% to 50%, 4 minutes; flow rate: 3.4 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 2.795 min, and a chiral isomer excess of 100%.


WX-037B: 1HNMR (400 MHz, CDCl3) δ: 11.15 (s, 1H), 9.03-8.78 (m, 2H), 8.38-8.08 (m, 4H), 7.56-7.53 (m, 1H), 7.35-7.31 (m, 1H), 7.22-7.21 (m, 1H), 3.85-3.76 (m, 1H), 3.46 (s, 3H), 3.07 (d, J=4.4 Hz, 3H), 1.74-1.69 (m, 1H), 1.42 (d, J=6.0 Hz, 3H), 1.42 (d, J=6.4 Hz, 3H), 1.14-1.13 (m, 2H), 0.99-0.94 (m, 2H); LCMS m/z=524.2 [M+H]+. SFC (column: Chiralpak IH-3, 3 μm, 0.46 cm id×10 cm L; mobile phase: A (CO2) and B (MeOH containing 0.1% isopropylamine); gradient: B %=10% to 50%, 4 minutes; flow rate: 3.4 mL/min; wavelength: 220 nm; pressure: 100 bar), with Rt of 2.983 min, and a chiral isomer excess of 98.50%.


Example 38



embedded image


embedded image


Step 1: Synthesis of Compound 38-1

Under a nitrogen atmosphere, compound 36-1 (1.82 g, 6.61 mmol, 1 eq) and A-12 (1.36 g, 6.61 mmol, 1 eq) were sequentially added to tetrahydrofuran (50 mL), and the mixture was stirred until dissolved. LiHMDS (1 M, 16.52 mL, 2.5 eq) was slowly added dropwise thereto at 0° C., and the mixture was stirred at 25° C. for another 2 hours. After the reaction was completed, the reaction mixture was added with water (100 mL) and extracted with ethyl acetate (100 mL*2). The organic phases were combined, washed with saturated brine (100 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:3) to obtain compound 38-1. LCMS m/z=444.0 [M+1]+.


Step 2: Synthesis of Compound 38-2

Compound 38-1 (2.1 g, 4.73 mmol, 1 eq) and cyclopropanecarboxamide (4.03 g, 47.30 mmol, 10 eq) were dissolved in dioxane (40 mL) and NMP (2 mL). Cesium carbonate (4.62 g, 14.19 mmol, 3 eq) and Xantphos (410.55 mg, 709.54 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (649.74 mg, 709.54 μmol, 0.15 eq) was added. The mixture was stirred at 130° C. for 16 hours. After the reaction was completed, the reaction mixture was cooled to room temperature, then added with water (40 mL), and extracted with ethyl acetate (40 mL*2). The organic phase was washed with saturated brine (40 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=10:1) to obtain compound 38-2. LCMS m/z=493.1 [M+1]+.


Step 3: Synthesis of Compound WX-038

Compound 38-2 (200 mg, 406.02 μmol, 1 eq) was added to a mixed solution of ethanol (10 mL) and water (5 mL), and potassium hydrogenperoxomonosulphate (374.41 mg, 609.02 mol, 1.5 eq) was added thereto. The reaction was carried out at 25° C. for 2 hours. The reaction mixture was directly filtered to obtain compound WX-038. 1H NMR (400 MHz, CDCl3) δ: 11.97 (br s, 1H), 9.39 (br s, 1H), 9.27 (s, 1H), 8.39 (d, J=8.8 Hz, 1H), 8.27-8.16 (m, 2H), 7.97 (dd, J=1.4, 7.9 Hz, 1H), 7.62 (d, J=6.8 Hz, 1H), 7.44 (t, J=7.9 Hz, 1H), 4.19-4.09 (m, 1H), 3.55 (s, 3H), 3.06 (d, J=4.3 Hz, 3H), 2.09-1.97 (m, 1H), 1.48 (d, J=7.0 Hz, 6H), 1.05-0.96 (m, 2H), 0.94-0.85 (m, 2H); LCMS m/z=525.1 [M+1]+.


Example 39



embedded image


embedded image


Step 1: Synthesis of Compound 39-1

Under a nitrogen atmosphere, compound 37-1 (532 mg, 1.94 mmol, 1 eq) and A-12 (397.56 mg, 1.94 mmol, 1 eq) were sequentially added to tetrahydrofuran (20 mL), and the mixture was stirred until dissolved. LiHMDS (1 M, 4.85 mL, 2.5 eq) was slowly added dropwise thereto at 0° C., and the mixture was stirred at 25° C. for another 2 hours. After the reaction was completed, the reaction mixture was added with water (50 mL) and extracted with ethyl acetate (50 mL*2). The organic phases were combined, washed with saturated brine (100 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=100:3) to obtain compound 39-1. LCMS m/z=443.0 [M+1]+.


Step 2: Synthesis of Compound 39-2

Compound 39-1 (786 mg, 1.77 mmol, 1 eq) and cyclopropanecarboxamide (1.51 g, 17.74 mmol, 10 eq) were dissolved in dioxane (20 mL) and NMP (0.5 mL). Cesium carbonate (1.73 g, 5.32 mmol, 3 eq) and Xantphos (154.01 mg, 266.16 μmol, 0.15 eq) were then added thereto. After replacing with nitrogen three times, Pd2(dba)3 (243.73 mg, 266.16 μmol, 0.15 eq) was added. The mixture was stirred at 130° C. for 16 hours. After the reaction was completed, the reaction mixture was cooled to room temperature, then added with water (20 mL), and extracted with ethyl acetate (20 mL*2). The organic phase was washed with saturated brine (20 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (dichloromethane:methanol=10:1) to obtain compound 38-2. LCMS m/z=492.2 [M+1]+.


Step 3: Synthesis of WX-039

Compound 39-2 (200 mg, 406.83 μmol, 1 eq) was added to a mixed solution of ethanol (10 mL) and water (5 mL), and potassium hydrogenperoxomonosulphate (375.16 mg, 610.25 mol, 1.5 eq) was added thereto. The reaction was carried out at 25° C. for 2 hours. After the reaction was completed, the reaction mixture was added with saturated sodium bisulfite aqueous solution (10 mL), and extracted with ethyl acetate (20 mL*2). The organic phase was washed with saturated brine (10 mL*2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product was purified by silica gel column chromatography (DCM:MeOH=10:1) to obtain compound WX-039. 1H NMR (400 MHz, CDCl3) δ: 10.58 (s, 1H), 9.10-8.90 (m, 1H), 8.35 (s, 2H), 8.28-8.21 (m, 1H), 8.20-8.14 (m, 2H), 7.62 (dd, J=1.3, 8.0 Hz, 1H), 7.33 (t, J=7.9 Hz, 1H), 7.16 (dd, J=1.5, 7.8 Hz, 1H), 6.35 (br s, 1H), 3.89-3.79 (m, 1H), 3.49 (s, 3H), 3.05 (d, J=4.8 Hz, 3H), 1.61-1.53 (m, 1H), 1.40 (d, J=7.0 Hz, 6H), 1.15-1.07 (m, 2H), 0.96-0.88 (m, 2H); LCMS m/z=524.1 [M+1]+.


Biological Test Data
Experimental Example 1: Inhibitory Activity of Compounds on TYK2 JH2 Pseudokinase

In this experiment, the inhibitory effect of compounds on TYK2 JH2 pseudokinase was tested using the method of time-resolved fluorescence resonance energy transfer (TR-FRET). In the experiment, TYK2 JH2 or JAK1 JH2 pseudokinase can simultaneously bind to fluorescently labeled Tracer and Tb antibody to form a “sandwich structure”. Tb antibody, functioning as a fluorescent donor, produces fluorescence at a wavelength of 495 nm under the excitation of light at a specific wavelength. Tracer, functioning as a fluorescent acceptor, can receive fluorescence at a wavelength of 495 nm and thus produce fluorescence at a wavelength of 520 nm, i.e., a time-resolved fluorescence resonance energy transfer (TR-FRET) signal, only when it is within the “sandwich structure”, i.e., when it is sufficiently close to Tb antibody. When the compound is added to compete with Tracer for binding to the pseudokinase, the binding of Tracer decreases, leading to a weakened TR-FRET signal. The inhibitory activity of the compound binding to the pseudokinase can be reflected by the ratio of signal values at 520 nm/495 nm.


1 Experimental Reagents: See Table 10









TABLE 10







Information on experimental reagents











Reagent name
Supplier
Storage conditions







TYK2/JAK1
Bioduro
−80° C.



Tracer
Bioduro
−80° C.



Tb antibody
Cisbio
−80º C.



HEPES
Invitrogen
 4º C.



MgCl2 1 M
Sigma
Room temperature



Brij L23 solution
Sigma
Room temperature



(Brij-35)





DTT
Sigma
−20° C.



BSA
Sigma
 4° C.










2 Experimental Methods
1) Preparation of 1× Experimental Working Solution

The working solution consists of the following components: HEPES (pH 7.5) at a final concentration of 20 mM; MgCl2 at a final concentration of 10 mM; Brij-35 at a final concentration of 0.015%; DTT at a final concentration of 2 mM; and BSA at a final concentration of 50 μg/mL.


2) Experimental Steps:





    • a) The compound was dissolved in DMSO to achieve a storage concentration of 10 mM.

    • b) Various concentrations of the compound, 200 times the final concentration, were prepared in a compound dilution plate and transferred to an Echo plate.

    • c) The Echo instrument was used to transfer 150 nL of the compound from the Echo plate to a 384-well experimental plate.

    • d) 5 μL of TYK2 JH2 kinase, at three times the final concentration, was added to the 384-well experimental plate.

    • e) 5 μL of Tb, at three times the final concentration, was added to the 384-well experimental plate.

    • f) 5 μL of Tracer, at three times the final concentration, was added to the 384-well experimental plate.

    • g) The plate was centrifuged for 30 seconds and then incubated at room temperature for 60 minutes.

    • h) The fluorescence signal values at 520 nm/495 nm were measured using an Envision plate reader (PerkinElmer).





3) Data Analysis

Data analysis was performed using the XL-Fit software to determine the IC50 of the compounds. The results are shown in Table 11:









TABLE 11







Kinase half maximal inhibitory


concentration IC50 (nM)










Test samples
TYK2 JH2







Hydrochloride of WX-001
0.27



Hydrochloride of WX-002
0.19



WX-003
0.34



WX-004
0.20



WX-011A
0.10



WX-011B
0.08



WX-012A
0.08



WX-012B
0.08



Hydrochloride of WX-018
0.13



WX-019
0.09



Hydrochloride of WX-020
0.08



WX-021
0.08










Conclusion: The compounds of the present disclosure exhibit good inhibitory effect on TYK2 JH2 pseudokinase.


Experimental Example 2: Inhibitory Activity of Compounds on the Proliferation of Ba/F3-FL-TYK2-E957D and Ba/F3-TEL-TYK2 Cells

Adenosine triphosphate (ATP) serves as a universal energy carrier in various life processes, representing the smallest unit of energy storage and transfer. The CellTiter-Glo™ Luminescent Cell Viability Assay Kit utilizes luciferase as a detection agent. During the luminescent process, luciferase requires the participation of ATP. By adding CellTiter-Glo™ reagent to the cell culture medium and measuring the luminescence, the light signal was directly proportional to the amount of ATP in the system, and ATP was positively correlated with the number of viable cells. Therefore, cell proliferation can be detected by measuring ATP content using the CellTiter-Glo kit. In this test, the cell lines used were Ba/F3-FL-TYK2-E957D and Ba/F3-TEL-TYK2. Ba/F3-FL-TYK2-E957D cells can stably express the exogenously introduced human TYK2-E957D gene, and the TYK2-E957D gene sequence contains both JH1 and JH2 domains. On the other hand, Ba/F3-TEL-TYK2 cells can stably express the exogenously introduced human TEL-TYK2 gene, and the TEL-TYK2 gene sequence contains only the JH1 domain of TYK2.


IC50 Measurement Process:
1) Cell Culture

The cell lines were cultured in an incubator at 37° C. with 5% CO2. Regular passaging was performed, and cells in the logarithmic growth phase were selected for plating.


2) Preparation of Compound Storage Plate





    • a) The test compounds were prepared into a 10 mM solution with DMSO, and then diluted to 0.3 or 1 mM with DMSO.

    • b) Preparation of 1000× compound storage plate (tube): The compounds were 3-fold serially diluted from the highest concentration to the lowest concentration, creating 9 different concentrations.

    • c) Preparation of 20× compound working solution: 49 μL of cell culture medium was added to a flat-bottomed 96-well transparent drug plate. From the 1000× compound storage plate, 1 μL of the compound was pipetted into the cell culture medium in the 96-well transparent drug plate. In the solvent control, 1 μL of DMSO was added. After the compound or DMSO was added, the mixture was homogenized using a multi-channel pipette.





3) Cell Plating and Drug Administration





    • a) Cells were stained with trypan blue and viable cells were counted to ensure a cell viability of 90% or more.

    • b) 95 μL of cell suspension (2000 cells/well) was added to each well of the compound assay cell plate, and cell-free culture medium containing 0.1% DMSO was added to the Min control wells.

    • c) Administration in the compound assay cell plate: 5 μL of 20× compound working solution was added to the cell culture plate. 5 μL of DMSO-cell culture medium mixture was added to the Max control wells. The final concentration of DMSO was 0.1%.

    • d) The culture plate was then incubated in an incubator at 37° C. with 5% CO2 for 72 hours.





4) Cell Viability Assay Using CellTiter-Glo Luminescence Method

The following steps were conducted in accordance with the instructions of the Promega CellTiter-Glo Luminescent Cell Viability Assay Kit (Promega-G7573).

    • a) CellTiter-Glo buffer was thawed and brought to room temperature.
    • b) CellTiter-Glo substrate was also brought to room temperature.
    • c) CellTiter-Glo buffer was added to a bottle of CellTiter-Glo substrate to dissolve the substrate, thereby preparing CellTiter-Glo working solution.
    • d) The mixture was slowly vortexed and shaken to be fully dissolved.
    • e) The cell culture plate was removed and allowed to equilibrate to room temperature for 10 minutes.
    • f) 50 μL (equal to half the volume of cell culture medium in each well) of CellTiter-Glo working solution was added to each well.
    • g) The culture plate was shaken on an orbital shaker for 2 minutes to induce cell lysis.
    • h) The plate was then left at room temperature for 10 minutes to stabilize the luminescent signal.
    • i) The luminescent signal was detected using a SpectraMax Paradigm plate reader.


5) Data Processing

Data analysis was conducted using GraphPad Prism 5.0 software. Nonlinear S-curve regression was employed to fit the data and generate dose-response curves. From these curves, the IC50 values were calculated. The data are shown in Table 12.









TABLE 12







Half maximal inhibitory concentration IC50 (nM) for cells












Ba/F3-FL-
Ba/F3-



Compound
TYK2-E957D
TEL-TYK2















Hydrochloride of WX-001
13.7




Hydrochloride of WX-002
19.2




WX-004
5.5




WX-005
2.2




Hydrochloride of WX-006
10.6




Hydrochloride of WX-007
9.5




Hydrochloride of WX-008
16.0




Trifluoroacetate of WX-009
12.1




Hydrochloride of WX-010
12.6




WX-011A
5.3
>10000



WX-011B
3.3
>10000



WX-012A
3.3
>10000



WX-012B
2.4
>10000



WX-013
12.1




Trifluoroacetate of WX-014
10.9




Hydrochloride of WX-015
13.0




Hydrochloride of WX-016
11.7




Hydrochloride of WX-017
4.0




Hydrochloride of WX-018
4.2




WX-019
2.0
>10000



Hydrochloride of WX-020
1.5
>10000



WX-021
2.5
>10000



WX-022
13.7




WX-023
13.8




WX-024
10.0




WX-025
12.7




Trifluoroacetate of WX-026
18.8




Hydrochloride of WX-027
18.3




WX-028
4.0




Hydrochloride of WX-029
3.8




WX-030
2.0




Hydrochloride of WX-031
2.6




Hydrochloride of WX-032
2.9




WX-033
0.1
>10000



Trifluoroacetate of WX-034
0.1
>10000



Trifluoroacetate of WX-035
6.7




WX-036A
8.3




WX-036B
17.3




WX-037A
15.0




WX-037B
17.0




WX-038
3.0




WX-039
4.7








“—” indicates that the measurement was not conducted.






Conclusion: The compounds of the present disclosure exhibit strong inhibitory activity against the proliferation of Ba/F3 cells transfected with the human TYK2-E957D gene (containing both JH1 and JH2 domains of TYK2). However, they have no inhibitory activity against the proliferation of Ba/F3 cells transfected with the human TEL-TYK2 gene (which contains only the JH1 domain of TYK2). This suggests that the compounds of the present disclosure are highly selective allosteric inhibitors of TYK2 JH12.


Experimental Example 3: Inhibitory Activity of Compounds on TYK2, JAK1/2, JAK2/2, and JAK1/3 Signaling Pathways in Human PBMC Cells

The purpose of this experiment was to detect the inhibitory effect of the compounds on cytokine-activated JAK-STAT signaling pathways in human peripheral blood mononuclear cells (PBMC).


1 Main Reagents and Instruments:





    • 1) Cells: Human peripheral blood mononuclear cells (PBMC) (Supplier: Sailybio)

    • 2) Reagents: see Table 13












TABLE 13







Information on reagents










Name
Supplier







1640 culture medium
Gibco



Non-essential amino acids
Gibco



Fetal bovine serum
ExCell Bio



Double antibiotic
Millipore



(Penicillin, Streptomycin)




Human interleukin 6 (IL-6)
Absin



Human interferon α (IFN-α)
PBL Assay Science



Human interleukin 2 (IL-2)
Absin



Human granulocyte-macrophage
Peprotech



colony-stimulating factor (GM-CSF)




Human CD4 antibody
Biolegend



Human CD33 antibody
Biolegend



Human pSTAT1 antibody
BD



Human pSTAT5 antibody
BD



Staining solution
Biolegend



Fixation buffer
BD



Permeabilization buffer
BD



Dulbecco's phosphate buffered
Corning



saline (DPBS)










3) Instruments

Flow cytometer: Brand: BD, model: Fortessa


2 Reagent Preparation

Culture medium: 1640 culture medium+10% fetal bovine serum+1% double antibiotic+1% non-essential amino acids (all percentages are volume ratios)


3 Experimental Steps





    • a) PBMCs frozen in liquid nitrogen were thawed in a 37° C. water bath, and culture medium was added. The mixture was then centrifuged at 320 g for 3 minutes.

    • b) Cells were resuspended in culture medium, counted, and the cell concentration was adjusted to 5×105 cells/mL with culture medium. Subsequently, the cell suspension was inoculated into two 96-well round-bottom plates, with 200 μL per well. The plates were incubated at 37° C. with 5% CO2 for 90 minutes.

    • c) Different concentrations of the test compound (starting at 2 μM, 5-fold serial dilution, a total of 8 concentrations) were added, followed by incubation at 37° C. with 5% CO2 for 30 minutes.

    • d) IL-6 (final concentration at 20 ng/mL) was added to the first round-bottom plate and incubated at 37° C. with 5% CO2 for 15 minutes.

    • e) IFNα (final concentration at 1000 U/mL) was added to the second round-bottom plate and incubated at 37° C. with 5% CO2 for 15 minutes.

    • f) IL-2 (final concentration at 4 ng/mL) was added to the third round-bottom plate and incubated at 37° C. with 5% CO2 for 15 minutes.

    • g) GM-CSF (final concentration at 20 μg/mL) was added to the fourth round-bottom plate and incubated at 37° C. with 5% CO2 for 15 minutes.

    • h) Cells were centrifuged at 320 g for 3 minutes, and each well was washed once with 200 μL of staining solution.

    • i) 50 μL of staining solution containing human CD33 antibody was added to each well of the GM-CSF-stimulated culture plate, and 50 μL of staining solution containing CD4 antibody was added to the remaining three culture plates. The staining was performed at 4° C. for 30 minutes. The cells were washed twice with the staining solution.

    • j) 100 μL of fixation buffer was added to each well, and the cells were fixed at 4° C. for 15 minutes. The cells were washed once with the staining solution.

    • k) 100 μL of permeabilization buffer was added to each well, and the cells were permeabilized at 4° C. for 20 minutes. The cells were washed twice with the staining solution.

    • l) 50 μL of staining solution containing human pSTAT1 antibody was added to each well of the IL-6 and IFN-α-stimulated culture plates, and 50 μL of staining solution containing human pSTAT5 antibody was added to each well of the IL-2 and GM-CSF-stimulated culture plates. The staining was performed at room temperature for 15 minutes. The cells were washed twice with the staining solution.

    • m) The cells were resuspended in 150 μL of staining solution.

    • n) The fluorescence intensity of pSTAT1 (stimulated by IL-6 and IFN-α) or pSTAT5 (stimulated by IL-2) in CD4-positive cells was detected using flow cytometer. The fluorescence intensity of pSTAT5 (stimulated by GM-CSF) in CD33-positive cells was also detected.





4 Data Analysis

Data analysis was performed using Flowjo software to determine the IC50 of the compounds. The results are shown in Table 14:









TABLE 14







Half maximal inhibitory concentration IC50 (nM) for cells












IFN-α/
IL-6/
GM-CSF/
IL-2/



pSTAT1
pSTAT1
pSTAT5
pSTAT5


Compound
(TYK2)
(JAK1/2)
(JAK2/2)
(JAK1/3)





WX-011A
2.6
 87.0
1656
1311


WX-011B
2.4
 72.5
1348
 773


WX-012A
2.0
 52.4




WX-012B
1.5
 32.5




WX-019
1.5
 99.7
1438
1576


Hydrochloride
1.3
163.9
1072
1396


of WX-020






WX-021
2.1
 63.4
 580
 540





“—” indicates that the measurement was not conducted.






Conclusion: The compound of the present disclosure exhibits high inhibitory activity against the TYK2 signaling pathway activated by IFN-α stimulation in human PBMC cells. Meanwhile, it exhibits weak inhibitory activity against the JAK1/2 signaling pathway activated by IL-6 stimulation, the JAK2/2 signaling pathway activated by GM-CSF stimulation, and the JAK1/3 signaling pathway activated by IL-2 stimulation, thereby showing high selectivity.


Experimental Example 4: Pharmacokinetic Testing of Compounds in Mice
Experimental Purpose:

The objective was to study the pharmacokinetic behavior of the compound of the present disclosure in mice and evaluate the pharmacokinetic characteristics, using male CD-1 mice aged 7 to 9 weeks as test animals. The drug concentrations in plasma at different time points after a single intravenous injection (IV) and oral administration (PO) of the compound were determined using LC/MS/MS.


Experimental Operation:

The pharmacokinetic characteristics of the compound in rodents following intravenous injection and oral administration were tested using a standard protocol. Test animals were fasted for 10 to 14 hours before administration and allowed to feed 4 hours post-administration. The compound was prepared into a clear solution with the appropriate solvent for both IV (intravenous injection) and PO (oral gavage) administration. The solvent was 10% DMSO+10% solutol+80% (10% HP-β-CD aqueous solution). Whole blood samples were collected within 24 hours, centrifuged at 6000 g for 3 minutes, and the supernatant was separated to obtain plasma samples. Protein precipitation was performed by adding four times the volume of acetonitrile solution containing internal standard to the plasma samples. After centrifugation, the supernatant was added with an equal volume of water and then centrifuged before being subjected to LC-MS/MS analysis for quantifying plasma drug concentration. Pharmacokinetic parameters such as peak concentration, time to peak concentration, clearance, half-life, area under the drug-time curve, and bioavailability were calculated.


The results of the pharmacokinetic parameters are shown in Table 15.









TABLE 15







Results of pharmacokinetic testing in mice




















WX-020





WX-011
WX-011A
WX-012
WX-019
(Hydrochloride)
WX-021

















IV
Dose (mg/kg)
1
3
1
3
3
3



Starting concentration
5422
9915
2747
16325
8045
9889



C0 (nM)









Half-life T1/2 (h)
3.2
4.2
4.4
1.1
2.3
2.9



Apparent volume of
0.7
0.8
2.9
0.4
1.1
0.9



distribution









Vd (L/kg)









Apparent clearance
6.6
14.1
12.8
4.7
7.0
8.5



Cl (mL/Kg/min)









Area under the curve
4791
6908
2462
21042
14130
11869



AUC0-last (nM · hr)








PO
Dose (mg/kg)
10
10
10
10
10
10



Peak concentration
10169
16814
12436
17335
15539
11337



Cmax (nM)









Time to peak
0.5
0.3
0.3
0.8
0.5
0.3



concentration









Tmax (h)









Area under the curve
24983
26715
16661
61215
56784
20720



AUC0-last (nM · hr)









Bioavailability F %
52%
51%
68%
87%
121%
52%









Conclusion: The compounds of the present disclosure exhibit excellent pharmacokinetic properties in mice.


Experimental Example 5: Pharmacokinetic Testing of Compounds in Rats
Experimental Purpose:

The objective was to study the pharmacokinetic behavior of the compound of the present disclosure in rats and evaluate the pharmacokinetic characteristics, using male SD rats aged 7 to 9 weeks as test animals. The drug concentrations in plasma at different time points after a single intravenous injection (IV) and oral administration (P0) of the compound were determined using LC/MS/MS.


Experimental Operation:

The pharmacokinetic characteristics of the compound in rodents following intravenous injection and oral administration were tested using a standard protocol. Test animals were fasted for 10 to 14 hours before administration and allowed to feed 4 hours post-administration. The compound was prepared into a clear solution with the appropriate solvent for both IV (intravenous injection) and PO (oral gavage) administration. The solvent was 1000 DMSO+10% solutol+80% (10% HP-β-CD aqueous solution). Whole blood samples were collected within 24 hours, centrifuged at 6000 g for 3 minutes, and the supernatant was separated to obtain plasma samples. Protein precipitation was performed by adding four times the volume of acetonitrile solution containing internal standard to the plasma samples. After centrifugation, the supernatant was added with an equal volume of water and then centrifuged before being subjected to LC-MS/MS analysis for quantifying plasma drug concentration. Pharmacokinetic parameters such as peak concentration, time to peak concentration, clearance, half-life, area under the drug-time curve, and bioavailability were calculated.


The results of the pharmacokinetic parameters are shown in Table 16.









TABLE 16







Results of pharmacokinetic testing in rats












Hydrochloride





of WX-011A
WX-020













IV
Dose (mg/kg)
1
3



Starting concentration C0 (nM)
4545
3026



Half-life T1/2 (h)
0.6
0.9



Apparent volume of distribution
0.5
0.9



Vd (L/kg)





Apparent clearance Cl
16.8
13.1



(mL/Kg/min)





Area under the curve
2075
2606



AUC0-last (nM.hr)




PO
Dose (mg/kg)
10
10



Peak concentration Cmax (nM)
4394
3377



Time to peak concentration
0.5
0.8



Tmax (h)





Area under the curve
8202
9762



AUC0-last (nM.hr)





Bioavailability F%
41%
39%









Conclusion: The compounds of the present disclosure exhibit excellent pharmacokinetic properties in rats.


Experimental Example 6: Inhibition Experiment of Compounds on IFN-α-Induced STAT1 Phosphorylation in Mouse Whole Blood

The purpose of this experiment was to detect the inhibitory effect of the compound on the JAK-STAT signaling pathway activated by IFN-α in mouse whole blood. Fresh mouse whole blood was collected and placed in a 96-well plate. The test compound was added and incubated for 1 hour, followed by stimulation with IFN-α. The corresponding STAT1 phosphorylation level in the CD3+ cell population was analyzed using flow cytometer by means of surface antibody staining and intracellular phosphorylated antibody staining. The IFN-α-induced STAT1 phosphorylation in mouse whole blood is dependent on TYK2 activity. By detecting the inhibitory activity of the compound on downstream STAT5 phosphorylation, the half maximal inhibitory concentration (IC50) of the compound on TYK2 signaling pathway activity can be determined.


1 Main Reagents and Instruments
1) Main Reagents: See Table 17









TABLE 17







Information on reagents










Name
Supplier







Dimethyl sulfoxide
Sigma



Perm buffer III
BD Biosciences



(permeabilization buffer)




Lyse/Fix buffer
BD Biosciences



(lysis/fixation buffer)




EDTA
Invitrogen



(ethylenediaminetetraacetic acid)




PBS (phosphate buffered saline)
BI



Brilliant Violet 421 anti-mouse
Biolegend



CD3 antibody




Alexa Fluor647 anti-STAT1
Biolegend



phospho (Ser727) antibody




Recombinant mouse IFNalpha
Miltenyi










2) Experimental Consumables

96-well V-bottom microplate, Greiner; 96 square-well deep well plate, Thermo; 96-well flat-bottom microplate, Corning


3) Instruments

CO2 incubator: MCO-15AC (Thermo);


Single-channel pipettes: 0.2 to 10 μL, 20 to 200 μL, 200 to 1000 μL (Thermo);


Multi-channel pipettes: 0.2 to 10 μL, 5 to 50 μL, 20 to 300 μL (Raining);


Centrifuges: Thermo Centrifuge ST 40R; Thermo LEGEND Micro 21R;


Water purification system: Millipore Milli-Q Reference system;


Vortex mixer: EARTH REQUIRED;


Shaker: QI LIN BEI ER; MH-2;


Flow cytometer: Beckman CytoFlex.


2 Experimental Steps
1) Compound Dilution





    • a) The compound was prepared into a 10 mM solution with dimethyl sulfoxide (DMSO), and diluted in DMSO to a solution with various concentrations, 500 times the final concentration.

    • b) 5 μL of the diluted compound was transferred to 120 μL of phosphate buffered saline (PBS) containing 0.1% bovine serum albumin (BSA).

    • c) Positive and negative control groups were set up, with both finally containing 0.2% DMSO.





2) Experimental Process





    • a) 67.5 μL of mouse whole blood was added to each well of a 96-well cell culture plate.

    • b) 3.5 μL of the diluted compound was added thereto and mixed thoroughly.

    • c) The plate was incubated in a 37° C. incubator for 60 minutes.

    • d) Mouse interferon recombinant protein was diluted 125-fold in PBS containing 0.1% BSA, and anti-mouse CD3 antibody was diluted 5-fold in PBS containing 0.1% BSA. 5 μL of diluted anti-mouse CD3 antibody and 4 μL of diluted mouse interferon recombinant protein were added to each well.

    • e) The plate was incubated in a 37° C. incubator for 30 minutes.

    • f) All cells were transferred to a 96-well deep well plate, and 1 mL of 37° C. pre-warmed 1× lysis/fixation buffer was added thereto.

    • g) The cells were incubated in the dark at 37° C. for 10 minutes.

    • h) After centrifugation at 600 g for 5 minutes, the supernatant was discarded, and the cells were washed twice with 1 mL of PBS by centrifugation.

    • i) 0.4 mL per well of permeabilization buffer was added to the cell pellet and mixed thoroughly. The cells were incubated in the dark at 4° C. for 30 minutes.

    • j) After centrifugation at 600 g for 5 minutes, the supernatant was discarded, and the cells were washed twice with 1 mL of flow cytometry staining buffer (PBS+0.2% BSA+1 mM EDTA) by centrifugation.

    • k) Anti-STAT1 antibody was diluted 65-fold in flow cytometry staining buffer, and added to cell-containing wells at 100 μL per well and mixed thoroughly.

    • l) The cells were incubated at room temperature for 40 minutes.

    • m) 1 mL per well of flow cytometry staining buffer was added, and the cells were washed twice by centrifugation at 600 g for 5 minutes.

    • n) The supernatant was discarded, and the cell pellet was resuspended in 300 μL of flow cytometry staining buffer.

    • o) The samples were loaded for analysis using the Beckman CytoFlex flow cytometer.





3 Data Analysis

Data were analyzed using FlowJo software. Curve fitting was performed and the IC50 was calculated using GraphPad Prism 8 software. The results are shown in Table 18.









TABLE 18







Half maximal inhibitory


concentration IC50 (nM)











IFN-α/pSTAT1



Compound
(TYK2)














WX-011A
148.2



WX-011B
375.0



WX-012B
418.6



WX-019
427.2



WX-020
369.3



WX-021
452.0



WX-033
315.9



Trifluoroacetate
93.2



of WX-034











Conclusion: In mouse whole blood, the compound of the present disclosure exhibits high inhibitory activity against the TYK2 signaling pathway activated by IFNα stimulation.


Experimental Example 7: Inhibition Experiment of Compounds on Cytokine-Induced STAT Phosphorylation in Human Whole Blood/Platelets

The purpose of this experiment was to detect the inhibitory effect of the compound on the JAK-STAT signaling pathway activated by cytokines in human whole blood or platelet-rich plasma. Fresh human whole blood/platelets were placed in a 96-well plate. The test compound was added and incubated for 1 hour, followed by stimulation with various cytokines. The corresponding STAT phosphorylation levels in different cell populations were analyzed using flow cytometer by means of surface antibody staining and intracellular phosphorylated antibody staining. The experiments using the cytokines IFN-α, TL-6, and TL-2 as stimulants were conducted in human whole blood, whereas the experiment using the cytokine TPO as a stimulant was conducted in platelet-rich plasma.


1 Main Reagents: See Table 19









TABLE 19







Information on reagents








Name
Supplier





1640 culture medium
BI


Alexa Fluor 647 Mouse Anti-Stat5 (pY694)
BD


(AF647-labeled mouse anti-STAT5 (pY694) antibody)



FITC Mouse Anti-Human CD3
BD


(FITC-labeled mouse anti-human CD3 antibody)



PE anti-human CD61 Antibody
Biolegend


(PE-labeled mouse anti-human CD61 antibody)



Alexa Fluor 647 mouse IgG1, k Isotype Ctrl antibody
Biolegend


(AF647-labeled mouse IgG1 isotype control antibody)



Recombinant human IL-2
PEPROTECH


Universal Type I IFN (1MU) (IFN-α)
R&D


Recombinant Human TPO (thrombopoietin)
Stem cell


96 Well Microplate
Beaver


2 mL 96 Well Standard Certified RNase/DNase Free
Costar


Non-Sterile (2 mL 96-well deep well plate)



Phosflow Lyse/Fix Buffer 5X (5X concentrated
BD


fixation buffer for lysing red blood cells)



Perm Buffer III (permeabilization buffer III)
BD









2 Experimental Steps
1) Cytokine-Induced Phosphorylation Experiment in Human Whole Blood





    • a) Fresh blood from volunteers was collected using EDTA anticoagulant tubes.

    • b) 90 μL per well of whole blood was rapidly seeded into a 96-well plate and incubated in a cell culture incubator for 15 minutes. 10 μL of the test compound was added thereto, with each assay performed in single wells. Both the negative control group and the positive control group were added with an equal volume of 1640 culture medium, equivalent to the DMSO content in the experimental group, with each assay performed in triplicate. The plate was then incubated in the incubator for 60 minutes.

    • c) After incubation, 25 μL of IL-2 at a final concentration of 20 ng/mL, IL-6 at a final concentration of 50 ng/mL, or IFN-α at a final concentration of 1000 U/mL was added to each well to induce stimulation. The negative control group was added with an equal volume of 1640 culture medium. The plate was then incubated in the incubator for 15 minutes.

    • d) 100 μL of 1× Lyse/Fix buffer was added to each well, and the plate was placed on ice to stop the reaction. The samples were then transferred to a 96-well deep well plate containing 10 times the volume of the blood sample of 37° C. pre-warmed 1× Lyse/Fix buffer, mixed thoroughly, and left to stand at 37° C. for 15 minutes.

    • e) 500 μL of ice-cold PBS was added to each well, followed by centrifugation at 500×g for 8 minutes. The supernatant was then discarded.

    • f) Cells were resuspended in 250 μL of pre-cooled PBS and transferred to a 96-well shallow well plate. The plate was centrifuged at 500×g for 8 minutes, and the supernatant was discarded.

    • g) The cell pellet was washed once with pre-cooled PBS.

    • h) After discarding the supernatant, anti-human CD3 antibody was added thereto, followed by incubation at room temperature for 40 minutes.

    • i) The plate was centrifuged at 500×g for 8 minutes, then the supernatant was discarded, and the cells were washed twice with 200 μL of pre-cooled PBS.

    • j) The cells were resuspended in 200 μL of pre-cooled Perm Buffer III per well and permeabilized on ice for 60 minutes.

    • k) The plate was centrifuged at 600×g for 8 minutes, then the supernatant was discarded, and the cells were washed twice with pre-cooled PBS.

    • l) Intracellular staining with anti-pSTAT5 antibody was carried out, followed by incubation at room temperature for 60 minutes.

    • m) The cells were centrifuged at 600×g for 8 minutes, washed twice with PBS, resuspended in 200 μL of PBS, and detected using the CytoFlex S flow cytometer.

    • n) Data analysis was conducted using FlowJo and GraphPad Prism 8 software.





2) Cytokine-Induced Phosphorylation Experiment in Human Platelet





    • a) Fresh blood from volunteers was collected using EDTA anticoagulant tubes and centrifuged at 200 g for 20 minutes to prepare platelet-rich plasma (PRP).

    • b) 90 μL per well of PRP was rapidly seeded into a 96-well plate and incubated in a cell culture incubator for 15 minutes. 10 μL of the test compound was added thereto, with each assay performed in single wells. Both the negative control group and the positive control group were added with an equal volume of 1640 culture medium, equivalent to the DMSO content in the experimental group, with each assay performed in triplicate. The plate was then incubated in the incubator for 60 minutes.

    • c) 25 μL of TPO at a final concentration of 200 ng/mL was added to induce stimulation. The negative control group was added with an equal volume of 1640 culture medium. The plate was then incubated in the incubator for 15 minutes.

    • d) After incubation, the plate was centrifuged at 1000×g for 10 minutes, then the supernatant was discarded, and the cells were washed twice with 200 μL of PBS.

    • e) PE-anti-human CD61 antibody was added to each well, followed by incubation at room temperature for 40 minutes.

    • f) After incubation, the plate was centrifuged at 1000×g for 10 minutes, then the supernatant was discarded, and the cells were washed twice with 200 μL of PBS.

    • g) The cells were resuspended in 200 μL of pre-cooled Perm Buffer III and permeabilized on ice for 60 minutes.

    • h) The plate was centrifuged at 1000×g for 10 minutes, then the supernatant was discarded, and the cells were washed twice with pre-cooled PBS.

    • i) Intracellular anti-pSTAT5 phosphorylated antibody was added thereto, followed by incubation at room temperature for 60 minutes.

    • j) The cells were washed twice with PBS and resuspended in 200 μL of PBS, and then detected using the CytoFlex S flow cytometer.





3 Data Analysis

Data were analyzed using FlowJo software. Curve fitting was performed and the IC50 was calculated using GraphPad Prism 8 software. Data were presented as mean and standard deviation (SD). The inhibition rate of the compound was defined as follows: Inhibition %=(1−(A−B)/(C−B))*100. Where: A is the MFI (mean fluorescence intensity) for experimental wells containing both the compound and the cytokine; B is the MFI (minimum mean fluorescence intensity) for control wells without the cytokine; C is the MFI (maximum mean fluorescence intensity) for control wells containing only the cytokine. The results are shown in Table 20.









TABLE 20







Half maximal inhibitory concentration IC50 (nM)












IFN-α/pSTAT5
IL-6/pSTAT3
TPO/pSTAT5
IL-2/pSTAT5


Compound
(TYK2)
(JAK1/2)
(JAK2/2)
(JAK1/3)














WX-011A
23.1
10754
>100000
11188


WX-019
29.7
33851
>100000
33754


WX-020
40.3
>100000
>100000
>100000









Conclusion: In human whole blood or platelets, the compound of the present disclosure exhibits high inhibitory activity against the TYK2 signaling pathway activated by IFNα stimulation. Meanwhile, it exhibits weak inhibitory activity against the JAK1/2 signaling pathway activated by IL-6 stimulation, the JAK2/2 signaling pathway activated by TPO stimulation, and the JAK1/3 signaling pathway activated by IL-2 stimulation, thereby showing high selectivity.


Experimental Example 8: Pharmacodynamic Study of Compounds on IL-12/IL-18-Induced IFN-γ Secretion by Mouse Immune Cells in an In Vivo Model

The purpose of this experiment was to evaluate the inhibitory effect of the compound on IL-12/IL-18-induced IFN-γ secretion by immune cells in mice. The IFN-γ protein level in serum samples of mice treated with the compound was detected using the enzyme-linked immunosorbent assay (ELISA) method.


1 Experimental Materials
a) Experimental Animals

C57BL/6J mice, aged 9 to 10 weeks, weighing 17.82 to 21.42 g, female, supplied by Shanghai Lingchang Biotechnology Co., Ltd.


b) Information on Recombinant Proteins: See Table 21









TABLE 21







Information on recombinant proteins










Product name
Supplier







Recombinant Mouse IL-12 Protein
R&D



Recombinant Mouse IL-18/IL-1F4 Protein
R&D



Mouse IFN-gamma Quantikine ELISA Kit
R&D










2 Experimental Methods and Procedures
1) Experimental Design and Drug Preparation

This experiment was conducted twice. The specific grouping of experimental animals and administration regimen are shown in Tables 22 and 23, respectively.


The compounds were all prepared into a clear solution with 10% DMSO+10% solutol+80% (10% HP-β-CD) as a solvent for PO (oral gavage) administration. The frequency of administration was once daily (QD).


Preparation of recombinant mouse IL-12 protein: An appropriate amount of antibody was transferred to a centrifuge tube, and a certain volume of PBS buffer was added to prepare a solution with a concentration of 0.1 μg/mL. The solution was gently shaken to mix and used immediately.


Preparation of recombinant mouse IL-18/IL-1F4 protein: An appropriate amount of antibody was transferred to a centrifuge tube, and a certain volume of PBS buffer was added to prepare a solution with a concentration of 10 μg/mL. The solution was gently shaken to mix and used immediately.


For the Normal group, the administration consisted of PO (oral gavage) with blank solvent and intraperitoneal injection of blank PBS solution. For the blank control group, the administration consisted of PO (oral gavage) with blank solvent and intraperitoneal injection of inducer IL-12/IL-18.









TABLE 22







Grouping of experimental animals and


administration regimen (first experiment)














Dose
Administration
Frequency of



Group
Drug
(mg/kg)
volume (mL/kg)
administration
N





1
Normal group
/
10
QD
3


2
Blank control
/
10
QD
6



group






3
WX-011A
 3
10
QD
6


4
WX-011A
10
10
QD
6
















TABLE 23







Grouping of experimental animals and


administration regimen (second experiment)














Dose
Administration
Frequency of



Group
Drug
(mg/kg)
volume (mL/kg)
administration
N





1
Normal group
/
10
QD
3


2
Blank control
/
10
QD
6



group






3
WX-020
 3
10
QD
6


4
WX-020
10
10
QD
6









2) Administration and Sample Collection





    • a) According to the grouping, the mice were administered by gavage (P0) with either the compound formulation or solvent control.

    • b) One hour later, each mouse received an intraperitoneal injection of IL-12 (0.01 μg per mouse) or PBS buffer.

    • c) One hour after IL-12 administration, each mouse received an intraperitoneal injection of IL-18 (1 μg per mouse) or PBS buffer.

    • d) Blood was collected three hours after IL-18 administration, and serum was separated.





3) ELISA Testing of Serum Samples





    • a) Prior to use, all reagents were mixed thoroughly to avoid foaming.

    • b) The number of strips required was determined based on the number of experimental wells (blanks and standards). Duplicate tests were performed for samples (including standards) and blanks.

    • c) Sample addition: 100 μL/well of the diluted Cytokine standard was added to the standard wells, 100 μL/well of the sample was added to the sample wells, and 100 μL/well of Dilution buffer R (1×) was added to the blank control wells.

    • d) Detection antibody addition: 50 μL/well of Biotinylated antibody working solution was added. After mixing, the plate was covered with a sealing film and incubated at 37° C. for 90 minutes.

    • e) Plate washing: The liquid was discarded from the wells, then 300 μL/well of 1× Washing buffer working solution was added, and allowed to stand for 1 minute before the liquid was discarded from the wells. This process was repeated 4 times, with drying on filter paper each time.

    • f) Enzyme addition: 100 μL/well of Streptavidin-HRP working solution was added.





The plate was covered with a sealing film and incubated at 37° C. for 30 minutes.

    • g) Plate washing: Step 5 was repeated.
    • h) Color development: 100 μL/well of TMB was added, then the plate was incubated at 37° C. in the dark for 5 to 30 minutes, and the reaction was terminated depending on the intensity of the color (dark blue) in the well. Typically, a color development period of 10 to 20 minutes was found to yield optimal results.
    • i) Reaction termination: 100 μL/well of Stop solution was quickly added to terminate the reaction.
    • j) Plate reading: Within 10 minutes after termination, the values were read at a measurement wavelength of 450 nm. Dual wavelengths were recommended for reading the plate, specifically measuring at a measurement wavelength of 450 nm and a reference wavelength of 610 to 630 nm simultaneously.


4) Data Analysis

The expression level of each sample was calculated based on the standard curve, and statistical analysis of the results for each group was performed, including mean and standard error of the mean (SEM). The signal values for each group were normalized against the blank control group. Statistical analysis was then conducted to assess differences between groups based on this data. A T-test was used for analysis, and for comparisons among three or more groups, one-way ANOVA was employed. All data were analyzed using GraphPad Prism 6.02. The inhibition rate of serum IFN-γ following a single administration of the compound is shown in Table 24.









TABLE 24







Inhibition rate of serum IFN-γ following single


administration of the compound











Serum IFN-γ


Compound
Dose
inhibition rate (%)












WX-011A
 3 mg/kg
64%


WX-011A
10 mg/kg
96%


WX-020
 3 mg/kg
40%


WX-020
10 mg/kg
86%









Conclusion: The compound of the present disclosure exhibits a significant dose-dependent inhibitory effect on IL-12/IL-18-induced IFNγ release in mice.


Experimental Example 9: Pharmacodynamic Experiment in a Mouse Model of Acute Colitis Induced by CD40 Antibody

The purpose of this experiment was to evaluate the alleviating effect of the compound on colitis in a mouse model induced by CD40 antibody.


1 Experimental Materials
1) Experimental Animals

CB-17 SCID mice, 8 weeks old, weighing 18 to 20 g, female, supplied by Beijing Vital River Laboratory Animal Technology Co., Ltd.


2) Reagents: See Table 25









TABLE 25







Information on reagents










Reagent
Source







Anti-CD40 antibody
BioXCell



IL-12 P40 antibody
BioXCell



Occult blood test reagent
Baso Diagnostic



(Piramidon method)
Inc



Saline
Shandong Kelun




Pharmaceutical



Dulbecco's phosphate
Corning



buffered saline (DPBS)










2 Experimental Methods
1) Experimental Design

CB-17 SCID mice were randomly divided into groups. Each group consisted of 6 mice, which received an intraperitoneal injection (IP) of CD40 antibody (80 μg per mouse) on Day 0. The positive control drug IL-12 P40 antibody and the test compound were administered from Day-1 to Day 4, with Day 5 being the endpoint of the experiment. The specific grouping of animals is shown in Table 26. The positive control drug IL-12 P40 antibody was administered via intraperitoneal injection (IP) once every three days; the test compound was administered by gavage (PO) twice a day (BID).









TABLE 26







Animal grouping and administration regimen
















Administration







Dose
volume
Route of
Frequency of
Number of


Group
Drug
(mg/kg)
(mL/kg)
administration
administration
animals





1
Blank control

10
PO
BID
6



group







2
IL-12 P40
20
10
IP
Once/3 days
6



antibody







3
Hydrochloride
30
10
PO
BID
6



of WX-011A







4
Hydrochloride
50
10
PO
BID
6



of WX-011A







5
Sulfate of
50
10
PO
BID
6



WX-020







6
Sulfate of
100
10
PO
BID
6



WX-020









2) Drug Preparation

The compounds were all prepared into a clear solution with 10% DMSO+10% solutol+80% (10% HP-3-CD) as a solvent for PO (oral gavage) administration. Administration volume parameters: 10 mL/kg based on the body weight of the mice.


Preparation of IL-12 P40 antibody: An appropriate amount of IL-12 P40 antibody stock solution was transferred to a 15 mL centrifuge tube, and a certain volume of DPBS was added to prepare a working solution with a concentration of 2 mg/mL. The solution was gently shaken to mix and used immediately.


3) Model Construction

IL-12 P40 antibody and the test compound were administered from Day-1 to Day 4. On Day 0, 30 minutes after administration, 200 μL of CD40 antibody (4 mg/kg) was intraperitoneally injected, while mice in the blank control group were intraperitoneally injected with 200 μL of DPBS. Body weight was recorded and feces were scored during the experiment.


4) Description of Fecal Observation Process in Animals

Fecal scoring involved placing each mouse individually in a specially designed cage for about 10 minutes for scoring. The fecal scoring criteria were as per Table 27.


5) Methodology Description for Fecal Occult Blood Testing

If blood is observed in the feces or around the anus, occult blood testing is not conducted. For the remaining mice without visible bloody stools, feces are collected for occult blood testing. Occult blood scoring is based on the assumption that the daily test results for the mice in the Normal group are 0. Each day, the fecal samples of four mice from the blank control group are observed for the time taken to develop color on the occult blood test strips. The shortest time required for color development in the feces of these four mice is set as the daily threshold. If the feces develop color before this time point and the color intensifies within 1 to 2 minutes, a score of 2 is assigned. If no visible color or only a weak color is seen within this threshold time, and later a color develops but is significantly less intense than that of the feces scoring 2, a score of 1 is assigned.


4 Data Acquisition

The body weight and Disease Activity Index (DAI) scores of the animals were recorded daily to assess the disease progression in each group and the effect of the test compound on the disease. The daily Disease Activity Index (DAI) score consists of three parts, with specific criteria referenced in Table 27. The results are shown in Table 28. DAI scores are shown in Table 29.


Note: Mean refers to the average value.









TABLE 27







DAI scoring criteria











Weight
Stool
Occult blood or bloody


Score
loss %
consistency
stools





0
   0
Normal
Occult blood negative


1
1 to 5
Soft stool
Occult blood weak positive


2
 6 to 10
Loose stool
Occult blood positive


3
11 to 20
Watery stool
Small amount of blood


4
>20
Watery diarrhea
Large amount of blood
















TABLE 28







Mouse weight change rate (%)











Weight change rate (%)















Group
Day
−1
0
1
2
3
4
5


















Blank control
Mean
100.00
97.58
98.63
89.07
86.11
92.24
93.88


group










IL-12 P40
Mean
100.00
100.28
101.07
98.73
102.65
104.08
108.98


antibody,










20 mg/kg










Hydrochloride of
Mean
100.00
100.63
99.54
97.69
100.43
100.96
99.34


WX-011A










50 mg/kg, BID










Hydrochloride of
Mean
100.00
100.60
100.01
95.93
98.85
99.69
97.07


WX-011A










30 mg/kg, BID










Sulfate of
Mean
100.00
100.78
99.88
97.14
99.65
101.16
100.90


WX-020










100 mg/kg, BID










Sulfate of
Mean
100.00
100.48
98.37
95.22
100.73
100.81
100.44


WX-020










50 mg/kg, BID
















TABLE 29







DAI scores











DAI score















Group
Day
−1
0
1
2
3
4
5





Blank control group
Mean
0.00
0.67
1.33
4.33
5.17
3.17
2.67


IL-12 P40 antibody,
Mean
0.00
0.33
0.33
1.17
0.67
0.00
0.00


20 mg/kg










Hydrochloride of
Mean
0.00
0.33
1.00
1.83
0.67
0.50
1.17


WX-011A










50 mg/kg, BID










Hydrochloride of
Mean
0.00
0.17
1.00
2.17
0.83
0.50
1.00


WX-011A










30 mg/kg, BID










Sulfate of WX-020
Mean
0.00
0.33
1.33
1.67
0.50
0.33
0.33


100 mg/kg, BID










Sulfate of WX-020
Mean
0.00
0.33
1.17
2.67
0.83
0.33
0.50


50 mg/kg, BID









Conclusion: In the mouse model of colitis induced by CD40 antibody, the compound of the present disclosure significantly outperformed the blank control group in controlling weight loss and reducing DAI scores at various doses. The compound of the present disclosure exhibits a significant alleviating effect on mouse colitis induced by CD40 antibody.


Experimental Example 10: Pharmacodynamic Experiment in a Mouse Psoriasis-Like Model Induced by IL-23

The purpose of this experiment was to evaluate the prophylactic and therapeutic effect of the test substance on IL-23-induced psoriasis-like skin lesions in mice by intradermal injection of IL-23 into the ear pinnae in a psoriasis-like skin lesion model.


1 Experimental Materials
1) Experimental Animals

C57BL/6 mice, SPF grade, weighing 19±2 g, female, sourced from the Laboratory Animal Business Department of Shanghai Institute of Planned Parenthood Research. The acclimatization period is 5 to 7 days.


2) Reagents: See Table 30









TABLE 30







Information on reagents










Reagent
Source







Recombinant human
Novoprotein



interleukin-23 (IL-23)




Ustekinumab
Selleck



(anti-IL-12/IL-23)




Sterile water for
Guangdong Aixida



injection
Pharmaceutical Co., Ltd.



Saline
Chimin Health




Management Co., Ltd.










3 Experimental Methods
1) Experimental Design

Female C57BL/6 mice of qualified body weight were randomly divided into groups, with 6 mice per group. The grouping and administration information was as follows. The groups were: blank control group, model group, positive control Ustekinumab (5 mg/kg) group, WX-011A hydrochloride (30, 50 mg/kg) group, and WX-020 sulfate (30, 50 mg/kg) group. Except for the blank control group, the animals in the other groups received an intradermal injection of IL-23 (3 μg/10 μL/mouse/day, QD) in the right ear for 8 consecutive days (Day 0 to Day 7). The blank control group received an intradermal injection of an equal volume of saline solution (10 μL/mouse/day, QD) in the right ear for 8 consecutive days (Day 0 to Day 7). Concurrent with model induction, the solvent or test drug was orally administered twice a day (Bid) at 6-hour intervals for 8 consecutive days (Day 0 to Day 7). The positive control was subcutaneously injected on Day 0 and Day 3 (a total of 2 doses). During the administration period, the body weight of the animals was monitored every two days. On Day 0, Day 2, Day 4, Day 6 (all before IL-23 injection), and Day 8, the thickness of the right ear of the mice was measured, and the appearance of the ear pinnae was observed and scored. Animal grouping and administration regimen are shown in Table 31.









TABLE 31







Animal grouping and administration regimen













Single
Administration
Route and




dose
volume
frequency of


Group
Drug
(mg/kg)
(mL/kg)
administration





1
Blank control
Solvent
10
PO/BID



group


(Administered at


2
Model group
Solvent
10
6-hour intervals)


3
Ustekinumab
 5
10
SC



(anti-IL-12/


(Administered on



IL-23)


Day 0 and Day 3)


4
Hydrochloride
30
10
PO/BID


5
of WX-011A
50
10
(Administered at


6
Sulfate of
30
10
6-hour intervals)


7
WX-020
50
10





*SC: subcutaneous injection;


PO: oral administration;


BID: twice a day






2) Instruments: See Table 32









TABLE 32







Instrument information









Instruments
Model
Manufacturer





Electronic digital
0-10X30
Zhejiang Deqing Shengtaixin


micrometer

Electronic Technology Co., Ltd.


Electric ear
YLS-25A
Jinan Yiyan Technology


edema piercer

Development Co., Ltd.









3) Drug Preparation

The compounds were all prepared into a clear solution with 10% DMSO+1000 solutol+80% (1000 HP-β-CD) as a solvent for PO (oral gavage) administration. Administration volume parameters: 10 mL/kg based on the body weight of the mice.


For the preparation of inducer IL-23, 3 vials were taken. 1.667 mL of sterile water for injection was added to each vial (500 μg), shaken and mixed, then transferred to a 10 mL EP tube, and shaken and mixed to obtain IL-23 solution at a concentration of 0.3 mg/mL. The solution was divided into 8 portions, aliquoted, and stored in a −80° C. freezer.


Ustekinumab (anti-IL-12/IL-23) preparation: 0.15 mL of Ustekinumab (anti-IL-12/IL-23) with a concentration of 5 mg/mL was pipetted and diluted with 1.35 mL of PBS (pH 7.2) solution to obtain Ustekinumab (anti-IL-12/IL-23) solution at a concentration of 0.5 mg/mL.


3) Model Construction and Administration

After grouping, IL-23 (3 μg/10 μL/mouse/day, QD) was injected intradermally into the right ears of the mice for 8 consecutive days (Day 0 to Day 7). Measurements of the thickness of the right ear and observations and scoring of the appearance of the ear pinnae were performed on Day 0, Day 2, Day 4, Day 6 (all before IL-23 injection), and Day 8. At the end of the experiment (Day 8), the left ear tissues of the mice from groups G4 to G10 (3 animals per group at each time point) were collected. The right ear, which had been modeled, was excised, an 8 mm ear punch was taken along the edge of the pinna, and its weight was measured. The excised right ear tissues were fixed in formalin for histopathological examination (H&E staining).


Concurrent with model induction, corresponding drug treatment was given for administration. Except for the positive control drug (Ustekinumab), which was subcutaneously injected on Day 0 and Day 3 (once a day), control and test drugs for each dose group were orally administered by gavage twice a day at 6-hour intervals for 8 consecutive days (Day 0 to Day 7).


The pathological scoring of mouse ear tissue sections was conducted according to the criteria in Table 33.









TABLE 33







Pathological scoring criteria












Degree of
Epidermal
Dermal
Inflammatory


Score
keratinization
thickness
thickness
cell infiltration





0
Relatively
Relatively
Relatively
Relatively



normal
normal
normal
normal


1
Mild
Mild
Mild
Mild


2
Moderate
Moderate
Moderate
Moderate


3
Moderate to
Moderate to
Moderate to
Moderate to



significant
significant
significant
significant


4
Significant
Significant
Significant
Significant









4) Data Statistics

The experimental data are represented as Mean±SD; statistical analysis was performed using IBM SPSS Statistics 21, with p<0.05 considered to indicate a statistically significant difference between the two groups. The effect of the test substance on the thickness of mouse ears in the IL-23 induced mouse pinna epidermal hyperplasia model is shown in Table 34. The effect of the test substance on the total score and AUC for TL-23 induced mouse pinna epidermal hyperplasia is shown in Table 35. The effect of the test substance on the pathological scoring of right ear tissue sections stained with H&E is shown in Table 36.









TABLE 34







Effect of the test substance on ear thickness in IL-23 induced


mouse pinna epidermal hyperplasia model (x)









Ear thickness (mm)/Day












Group
Day 0
Day 2
Day 4
Day 6
Day 8





Blank control group
0.196
0.211
0.213
0.213
0.217


Model group
0.198
0.297
0.354
0.446
0.482


Ustekinumab
0.196
0.277*
0.288**
0.316**
0.329*


5 mg/kg, SC







Hydrochloride of
0.199
0.274*
0.299*
0.297**
0.301**


WX-011A







30 mg/kg, BID







Hydrochloride of
0.197
0.279p=0.081
0.291 *
0.299**
0.300**


WX-011A







50 mg/kg, BID







Sulfate of WX-020
0.197
0.283
0.311
0.351**
0.343p=0.082


30 mg/kg, BID







Sulfate of WX-020
0.195
0.289
0.303*
0.311**
0.317**


50 mg/kg, BID






##P < 0.01 vs. blank control group



*P < 0.05 vs. model group


**P < 0.01 vs. model group













TABLE 35







Effect of the test substance on the total score and AUC for epidermal


hyperplasia of mouse pinnae induced by IL-23 (x)










Total score for epidermal hyperplasia of ear




pinnae/Day














Group
Day 0
Day 2
Day 4
Day 6
Day 8
AUC
















Blank control group
0.00
0.00
0.00
0.00
0.00
0.00


Model group
0.00
0.00
2.33##
5.83##
7.83##
24.17##


Ustekinumab
0.00
0.00
0.33*
0.67**
0.83**
2.83**


5 mg/kg, SC








Hydrochloride of
0.00
0.00
0.33*
0.67**
1.17**
3.17**


WX-011A








30 mg/kg, BID








Hydrochloride
0.00
0.00
0.17**
0.67**
1.00*
2.67**


of WX-011A








50 mg/kg, BID








Sulfate of WX-020
0.00
0.00
0.50*
2.5
1.67*
7.67


30 mg/kg, BID








Sulfate of WX-020
0.00
0.00
0.50*
1.33*
1.33**
5.00*


50 mg/kg, BID






##P < 0.01 vs. blank control group



*P < 0.05 vs. model group


**P < 0.01 vs. model group













TABLE 36







Effect of the test substance on pathological scoring of right ear tissue


sections stained with H&E (x)









Pathological score













Degree of
Epidermal
Dermal
Inflammatory
Comprehensive


Group
keratinization
thickness
thickness
cell infiltration
pathological score





Blank control
0.00
0.17
0.00
0.33
0.50


group







Model group
1.67##
2.50##
2.17##
3.00##
9.33##


Ustekinumab
0.50**
1.17**
0.67
1.17**
3.50**


5 mg/kg, SC







Hydrochloride
0.17**
0.67**
0.17
0.17**
1.17**


of WX-011A







30 mg/kg, BID







Hydrochloride
0.33**
0.50**
0.50
0.50**
1.83**


of WX-011A







50 mg/kg, BID







Sulfate of
0.83**
0.83**
1.33
1.17**
4.17**


WX-020







30 mg/kg, BID







Sulfate of
0.33**
0.67**
0.50
0.67**
2.17**


WX-020







50 mg/kg, BID






##P < 0.01 vs. blank control group



*P < 0.05 vs. model group


**P < 0.01 vs. model group






Conclusion: Compared to the model group, the compound of the present disclosure, at various doses, exhibits a significant alleviating effect on the IL-23 induced mouse psoriasis-like condition. This was evidenced in the increase thickness of the modeled side ear, the total score of the modeled side ear, and the comprehensive pathological score of the ear tissue sections, demonstrating significant pharmacological efficacy.

Claims
  • 1. A compound of formula (V) or a pharmaceutically acceptable salt thereof,
  • 2. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R1 is selected from OCH3, and the OCH3 is optionally substituted by 1, 2, or 3 Ra.
  • 3. The compound or the pharmaceutically acceptable salt thereof according to claim 2, wherein R1 is selected from OCH3 and OCF3.
  • 4. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R2 is selected from H and CH3, and the CH3 is optionally substituted by 1, 2, or 3 Rb.
  • 5. The compound or the pharmaceutically acceptable salt thereof according to claim 4, wherein R2 is selected from H, CH3, and CD3.
  • 6. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R3 is selected from
  • 7. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein the structural moiety
  • 8. The compound or the pharmaceutically acceptable salt thereof according to claim 6, wherein the structural moiety
  • 9. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R5 is selected from CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl, and the CH3, cyclopropyl, imidazolyl, pyrazolyl, and pyridyl are each independently and optionally substituted by 1, 2, or 3 Re.
  • 10. The compound or the pharmaceutically acceptable salt thereof according to claim 9, wherein R5 is selected from CH3,
  • 11. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein the structural moiety
  • 12. The compound or the pharmaceutically acceptable salt thereof according to claim 1, selected from:
  • 13. A compound of the following formula or a pharmaceutically acceptable salt thereof, selected from:
  • 14. The compound or the pharmaceutically acceptable salt thereof according to claim 13, selected from:
  • 15. A method for inhibiting TYK2 kinase in a subject in need thereof, comprising: administering the compound or the pharmaceutically acceptable salt thereof according to claim 1 to the subject.
  • 16. A method for treating or alleviating colitis or psoriasis in a subject in need thereof, comprising: administering the compound or the pharmaceutically acceptable salt thereof according to claim 1 to the subject.
Priority Claims (7)
Number Date Country Kind
202110801875.1 Jul 2021 CN national
202110875486.3 Jul 2021 CN national
202111162655.5 Sep 2021 CN national
202111273381.7 Oct 2021 CN national
202210039225.2 Jan 2022 CN national
202210260637.9 Mar 2022 CN national
202210731484.1 Jun 2022 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2022/106053 7/15/2022 WO