CA3203276A1 - Five-membered ring derivative and medical use thereof - Google Patents

Five-membered ring derivative and medical use thereof

Info

Publication number
CA3203276A1
CA3203276A1 CA3203276A CA3203276A CA3203276A1 CA 3203276 A1 CA3203276 A1 CA 3203276A1 CA 3203276 A CA3203276 A CA 3203276A CA 3203276 A CA3203276 A CA 3203276A CA 3203276 A1 CA3203276 A1 CA 3203276A1
Authority
CA
Canada
Prior art keywords
ring
alkyl
substituted
alkoxy
optionally further
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CA3203276A
Other languages
French (fr)
Inventor
Chen Zhang
Ming Lei
Mingliang ZHAO
Yan Yu
Pingming TANG
Guanglin WENG
Tao MOU
Yao Li
Jia NI
Pangke YAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xizang Haisco Pharmaceutical Co Ltd
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority claimed from PCT/CN2021/141243 external-priority patent/WO2022135572A1/en
Publication of CA3203276A1 publication Critical patent/CA3203276A1/en
Pending legal-status Critical Current

Links

Abstract

A compound represented by general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, an intermediate thereof and a preparation method therefor, as well as an application in preparation of a drug for treating diabetes.

Description

FIVE-MEMBERED RING DERIVATIVE AND MEDICAL USE THEREOF
Technical Field [0001] The present invention relates to a compound represented by general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, an intermediate thereof, and a preparation method therefor, as well as an application in the preparation of a drug for treating diabetes.
Background Art
[0002] Diabetes is a group of metabolic diseases characterized by hyperglycemia.
Hyperglycemia is caused by defective insulin secretion or impaired insulin biological action, or both. The long-term high blood sugar in diabetes leads to chronic damage and dysfunction of various tissues, especially eyes, kidneys, heart, blood vessels, and nerves.
Diabetes is mainly divided into two types. Type 1 diabetes: destruction of islet B cells leads to absolute insulin deficiency. Type 2 diabetes: insulin resistance is dominant with insulin relative deficiency or impaired insulin secretion is dominant with insulin resistance.
[0003] Drugs for type 2 diabetes can be divided into six major classes (insulin, insulin secretagogues, biguanides, glucosidase inhibitors, thiazolidinediones, SGLT2 inhibitors), each of which works through a different primary mechanism. However, with the exception of GLP-1 receptor agonists and SGLT2 inhibitors, these drugs have limited efficacy and cannot address the most important problems, namely decreased cellular function and associated obesity.
[0004] GLP-1 is a 30-amino acid long intestinal insulin stimulating hormone secreted by L cells in the intestine. GLP-1 stimulates insulin secretion, reduces glucagon secretion, inhibits gastric emptying, reduces appetite, and stimulates 13-cell proliferation in a physiological and glucose-dependent manner. In nonclinical experiments, GLP-1 promotes the sustainability of 13 cells by stimulating the transcription of genes important for glucose-dependent insulin secretion and by promoting (3 cell regeneration. In healthy individuals, GLP- 1 plays an important role in the regulation of postprandial blood, leading to increased peripheral glucose uptake by stimulating glucose-dependent insulin secretion from the pancreas. GLP-1 also inhibits glucagon secretion, resulting in decreased hepatic glucose output. In addition, GLP-1 delays gastric emptying, slows small bowel motility, and delays food absorption.
[0005] GLP-1 receptor agonists, such as GLP-1, liraglutide and exendin-4, are polypeptide drugs and are mostly used for injection. Small molecule GLP-1 receptor agonists have become a hot spot in drug development in recent years due to their high oral bi availability potential.
Summary of the Invention
[0006] The object of the present invention is to provide a compound capable of stimulating a GLP-1 receptor or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and an intermediate and a preparation method thereof, as well as an application in the preparation of a drug for treating diabetes.
[0007] The compound of the present invention has good activity of stimulating a GLP-1 receptor, good pharmacokinetic performance and bioavailability, oral performance and good safety.
[0008] The present invention provides a compound or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and the compound is selected from a compound represented by general formula (I), wherein, R2 R2 iR1 DI ______________________________ Le /
N ' COOH
(I)
[0009] In some embodiments, ring B is selected from a 4- to 12-membered heterocyclic ring, C5-12 carbocyclic ring, C6-10 aromatic ring or a 5- to 12-membered heteroaromatic ring. The carbocyclic ring, heterocyclic ring, aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RB, the heterocyclyl or heteroaromatic ring contains 1 to 3 (for example 1, 2 or 3) heteroatoms selected from 0, S and N.
[0010]
In some embodiments, ring B is selected from a benzene ring, a 4- to 8-membered heterocyclyl, C5-8 carbocyclyl or a 5- to 6-membered heteroaromatic ring. The benzene ring, carbocyclyl, heterocyclyl or heteroaromatic ring is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) RB, the heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0011]
In some embodiments, ring B is selected from one of the following substituted or unsubstituted groups: cyclohexyl, cyclohexenyl, azacyclohexenyl, piperidinyl, phenyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RB.
[0012]
In some embodiments, ring B is selected from one of the following substituted or unsubstituted groups: pyrimidin-4(3H)-one (that is, the ortho position of the pyrimidine N atom is substituted with a hydroxyl group, and pyrimidin-4(3H)-one is formed through tautomerism), pyridazine-3(2H)-one (that is, the ortho position of the pyridazine N atom is substituted with a hydroxyl group, and pyridazine-3(2H)-one is formed through tautomerism), pyridine-2(/H)-one (that is, the ortho position of the pyridine N atom is substituted with a hydroxyl group, and pyridin-2(/H)-one is formed through tautomerism).
[0013]
In some embodiments, ring B is selected from one of the following substituted or unsubstituted groups: piperazinyl, tetrahydropyrrolyl or 1,4-diazepanyl, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RB.
[0014]
In some embodiments, ring B is selected from one of the following substituted r')C
A4lNj ANI
N
or unsubstituted groups:

C)I\JC /\)c N) ?NO'k -css:N/k ?Nk nN
`?-=
, the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 Rs.
[0015]
In some embodiments, ring B is selected from one of the following substituted C)I\JC
µ7õN '7õNJ

or unsubstituted groups:

N
or the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.
[0016]
In some embodiments, ring B is selected from one of the following substituted 1-/Nrµ
or unsubstituted groups: -z C or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.
[0017]
In some embodiments, ring B is selected from one of the following substituted NC ,0)?
or unsubstituted groups: '1/2-) or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 R.
[0018] In some embodiments, ring B is selected from one of the following substituted N
or unsubstituted groups: or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 Rs.

In some embodiments, ring B is selected from one of the following substituted N-\ N-\ Ni N
or unsubstituted groups:
NN
, N
or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 Rs.

[0020] In some embodiments, is selected from or N
, the left side of which is directly connected to ring C, and which, when the 6-membered ring in the spiro ring is substituted, is optionally further substituted with 1, 2 or 3 Rs.
[0021] In some embodiments, ring B is selected from one of the following substituted N
or unsubstituted groups: V--) , or -, the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.
[0022]
In some embodiments, ring C is selected from C6_io carbocyclic ring, a 5-to 10-membered heterocyclic ring, C6-10 aromatic ring or a 5- to 10-membered heteroaromatic ring. The carbocyclic ring, heterocyclic ring, aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc, the heterocyclic ring or heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0023] In some embodiments, ring C is selected from one of the following substituted or unsubstituted groups: A benzene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a tetrazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc.
[0024]
In some embodiments, ring C is selected from a benzene ring, a 5- to 6-memberedmonocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6-membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring. The benzene ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc, the heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0025]
In some embodiments, ring C is selected from one of the following substituted Nk !\lk Nk or unsubstituted groups: a benzene ring, ¨
N77"Nk N
N N
N N or , which = , when substituted, is optionally further substituted with 1, 2 or 3 Rc, and the left side of which is connected to L.
[0026]
In some embodiments, ring C is selected from one of the following substituted \
or unsubstituted groups: or , which, when substituted, is optionally further substituted with 1, 2 or 3 Rc, and the left side of which is connected to L.

L
[0027] In some embodiments, is selected from , the right side of which is connected to ring B, and in which the benzene ring is optionally further substituted with 1, 2 or 3 Rc.

1100281 In some embodiments, L is selected from a bond, 0, S, -NRL-, -C(R02-, -C(RL)2-C(RL)2-, -Y-C(RL)2-, -C(RL)2-Y-, -Y-C(RL)2-C(RL)2-, -C(RL)2-C(RL)2.-Y-or -C(RL)2-Y-C(R02-.
[0029] In some embodiments, L is selected from a bond, 0, S, -NRL-, -CHRL-, -CHRL-CHRL-, -Y-CHRL-, -CHRL-Y-, -Y-CHRL-CHRL-, -CHRL-CHRL-Y- or -CHRL-Y-CHRL-.
[0030] In some embodiments, L is selected from -Y-CHRL-, -CHRL-Y-, -CHRL-CURL-, -CHRL-CHRL-Y- or -Y-CHRL-CHRL-.
[0031] In some embodiments, Y is selected from 0, S or -NRL-.
[0032] In some embodiments, L is selected from -CH20-, -OCH2-, -CH2NH-, -NIICH2-, -CII2C112-, -OCH2C112-, -CH2C1120-, -NIICH2C112- Or -CII2C112N11-, and the right side of L is connected with ring C.
[0033] In some embodiments, L is selected from -CH20-, and the right side of L is connected to ring C.
[0034] In some embodiments, ring D is selected from a 6- to 10-membered aromatic ring or a 5- to 12-membered heteroaromatic ring. The aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 5 RD. The heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0035] In some embodiments, ring D is selected from a benzene ring, a naphthalene ring, a 5- to 6-membered monocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6-membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring. The benzene ring, naphthalene ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RD, the heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0036] In some embodiments, ring D is selected from a benzene ring, a naphthalene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RD.

1100371 In some embodiments, ring D is selected from a benzene ring or a pyridine ring, and the benzene ring or pyridine ring is optionally further substituted with 1, 2 or 3 RD.
[0038] In some embodiments, ring D is selected from a substituted or unsubstituted benzene ring, and when substituted, is optionally further substituted with 1, 2 or 3 RD.
[0039] In some embodiments, ring E is selected from a 5-membered heterocyclic ring, and the heterocyclic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NH2, C1-6 alkyl, C1-6 alkoxy or C3-6 cycloalkyl. The heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[00401 In some embodiments, r is selected from 0 or 1.
N ' N-' [0041] In some embodiments, r=1, f is selected from N
N ' N
[0042] In some embodiments, r=0, l: is selected from N N N N
N
-Th N
N

,)1, S H 0 , S
, , , or .
N N
/ / S
N N
[0043] In some embodiments, 1=0, I' is selected from , S , or S .
[0044] In some embodiments, ring E is selected from a 5-membered non-aromatic heterocyclic ring or a 5-membered heteroaromatic ring, and the heterocyclic ring or heteroaromatic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NH2, C1-4 alkyl, C14 alkoxy or C3-6 cycloalkyl, the heterocyclic ring or heteroaromatic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[0045] In some embodiments, R1 is selected from H, halogen, OH, -SH, CF3, CN, Ci-6 alkyl, C1-6 alkoxy, -Ci_3 alkylene-Z-00_3 alkylene-Ria, -00-4 alkylene-Ria.
The alkyl, alkoxy, and alkylene are optionally further substituted with 0 to 4 (such as 0, 1 , 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, C1-6 alkyl, halogen-substituted C1-6 alkyl, hydroxy-substituted C1-6 alkyl, cyano-substituted C1-6 alkyl, C1-6 alkoxy, C3-6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl, a 5- to 10-membered heteroaryl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0046] In some embodiments, R1 is selected from H, halogen, OH, -SH, CF3, CN, Ci-4 alkyl, C14 alkoxy, -Ci_2 alkylene-Z-00_2 alkylene-Ria, -00_4 alkylene-Ria.
The alkyl, alkoxy, and alkylene are optionally further substituted with 0 to 4 (such as 0, 1 , 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, C1_4 alkyl, halogen-substituted C1_4 alkyl, hydroxy-substituted Ci_4 alkyl, cyano-substituted C1_4 alkyl, C1-4 alkoxy, C3-6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl, a 5- to 6-membered heteroaryl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0047] In some embodiments, R1 is selected from H, F, Cl, OH, CN, CF3, methyl, ethyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, -ethylene-Z-methylene-Ri a, -ethylene-Z-ethylene-Ri a, -ethylene-Z-Ri a, -R1 a, -methylene-Ri a, -ethylene-Ri a. The methyl, ethyl, propyl, methoxy, ethoxy, methylene, ethylene are optionally further substituted with 0 to 3 (for example 0, 1, 2 or 3) substituents selected from H, F, Cl, =0, CN, 01-1, -N(Rib)2, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyrimidinyl, pyrazinyl or pyridazinyl.
[0048] In some embodiments, Ri is selected from H, CN, CF3, CHF2, CH2F, -CH2OH, -CH(OH)CH3, methyl, ethyl, methoxy, ethoxy, propoxy, isopropoxy, methoxymethyl, methoxyethyl, ethoxym ethyl , i sopropoxym ethyl , -ethyl en e-Z-m ethyl en e-Ri a, -ethyl en e-Z-ethylene-Ri a, -ethylene-Z-Ri a, Ria- or -methylene-R1 a-o [0049] In some embodiments, Ri is selected from -CH7CH7OCH3, </N

(crk.k. 0 N 0 0 or C
CC-kk ga\____µ, 0 [0050] In some embodiments, Ri is selected from or [0051] In some embodiments, Z is selected from a bond, N(Rib), 0 or S.
[0052] In some embodiments, Z is selected from NH, N(CH3) or 0.
[0053] In some embodiments, Ria is selected from C1_6 alkyl, C1-6 alkoxy, Cs cycloalkyl, a 3- to 8-membered heterocycloalkyl, a 6- to 10-membered aryl or a 5- to 12-membered heteroaryl. The cycloalkyl, heterocycloalkyl, aryl or heteroaryl is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, C1-6 alkyl, C1_6 alkoxy or -N(R1b)2. The alkyl or alkoxy is optionally further substituted with 0 to 4 (such as 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, C1-6 alkyl, C1_6 alkoxy or C3-6 cycloalkyl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0054] In some embodiments, Ria is selected from C1-4 alkyl, C1-4 alkoxy, C3-6 cycloalkyl, a 3- to 6-membered heterocycloalkyl, phenyl or a 5- to 6-membered heteroaryl.
The cycloalkyl, heterocycloalkyl, phenyl or heteroaryl is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, CI_ 4 alkyl, Ci_4 alkoxy or -N(R1b)2. The alkyl or alkoxy is optionally further substituted with 0 to 4 (such as 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, CN, 01-1, -N(Rib)2, C14 alkyl, C14 alkoxy or C3-6 cycloalkyl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0055] In some embodiments, Ria is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyrimidinyl, pyrazinyl or phenyl. The Ria is optionally further substituted with 0 to 3 substituents selected from H, F, =0, OH, CN, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy or -N(Rib)2.
[0056] In some embodiments, Ria is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, pyrrolidinyl, piperidinyl, pyrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, 1,2,4-oxadiazolyl, isothiazolyl, pyrimidyl, pyridine or phenyl. The Ria is optionally further substituted with 0 to 3 (such as 0, 1, 2 or 3 ) substituents selected from H, F, CN, methyl, ethyl, propyl, CF3, methoxy or ethoxy.
[0057] In some embodiments, each Rib is independently selected from H or C1-6 alkyl;
the alkyl is optionally further substituted with 0 to 3 (such as 0, 1, 2 or 3) substituents selected from H, halogen, =0, CN, OH, NH2, Ci-6alkyl, C1-6 alkoxy or C3-6 cycloalkyl.
[0058] In some embodiments, each R11) is independently selected from H or C1_4 alkyl which alkyl is optionally further substituted with 0 to 3 (such as 0, 1, 2 or 3) substituents selected from H, halogen, =0, CN, OH, NH2, C1_4 alkyl, C1_4 alkoxy or C3-6 cycloalkyl.
[0059] In some embodiments, each Rib is independently selected from H, methyl or ethyl.
[0060] In some embodiments, each RL is independently selected from H, halogen, -SH, CN, OH, CF3, NH2, C1-6 alkyl, C1-6 alkoxy, C3-6 cycloalkyl. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy.
[0061] In some embodiments, each RL is independently selected from H, halogen, CN, OH, -SH, CF3, NH2, C1_4 alkyl, C1_4 alkoxy, C3-6 cycloalkyl. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C14 alkyl, C36 cycloalkyl or C14 alkoxy.
[0062] In some embodiments, each RL is independently selected from H, F, Cl, CN, CF3, OH, NH?, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl or cyclopentyl.
[0063] In some embodiments, each RL is independently selected from H, F, methyl, ethyl or propyl.
[0064] In some embodiments, each RL is independently selected from H or methyl.
[0065] In some embodiments, R2 is selected from H, halogen or C1_6alkyl.
[0066] In some embodiments, R2 is selected from H, halogen or C1_4alkyl.
[0067] In some embodiments, R2 is selected from H.
[0068] In some embodiments, R2 is selected from H, F, methyl or ethyl.
[0069] In some embodiments, each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, Ci_6 alkyl, C3_6 cycloalkyl or C1-6 alkoxy.
[0070] In some embodiments, each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, C14 alkyl, C3-6 cycloalkyl or C1_4 alkoxy.
The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C14 alkoxy.
[0071] In some embodiments, each RB, Rc or RD is independently selected from H, halogen, =0, CN, OH, -SH, NH2, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, tert-butyloxy, butoxy, cyclopropyl or cyclobutyl. The methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, tert-butyloxy, butoxy, cyclopropyl or cyclobutyl is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-cycloalkyl or C1_4 alkoxy substituents.
[0072] In some embodiments, each RB, Rc or RD is independently selected from H, F, Cl, =0, CN, CF3, OH, NH2, methyl, ethyl, propyl, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, propoxy, isopropoxy, tert-butyloxy, trifluoromethoxy, cyclopropyl, cyclobutyl or cyclopentyl.
[0073] In some embodiments, each RE is independently selected from H, =0, F, OH, CN, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy.
[0074] In some embodiments, each Rc is independently selected from H, F, Cl, OH, CN, NH2, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy.
[0075] In some embodiments, each RE is independently selected from H, =0, F, CN, CF3, methyl, ethyl or cyclopropyl.
[0076] In some embodiments, each RE is independently selected from H, F, =0 or methyl.
[0077] In some embodiments, each Rc is independently selected from H, F, CF3 or methyl.
[0078] In some embodiments, each Rc is independently selected from H, F, Cl, CN, CF3, methyl, ethyl, methoxy or ethoxy.
[0079] In some embodiments, each RD is independently selected from H, F, Cl, CN, CF3, CHF2, methyl, ethyl, methoxy, ethoxy, tert-butyloxy or trifluoromethoxy.
[0080] In some embodiments, each RE is independently selected from H, =0, F, CN, CF3, methyl, ethyl or cyclopropyl.
[0081] In some embodiments, each Rc is independently selected from H, F, Cl, CN, CF3, methyl, ethyl, methoxy or ethoxy.
[0082] In some embodiments, each RD is independently selected from H, F, Cl, CN, CF3, CHF2, methyl or ethyl.
[0083] In some embodiments, two RB, two Rc, two RD, Rc and RL, or RD and RL
and the atoms attached thereto together form a C3_iocarbocyclic ring or a 3- to 10-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy.
The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S
and N.
[0084] Tn some embodiments, two RB, two Rc, two RD, Rc and RL, or RB and R2 and the atoms attached thereto together form a C3_7carbocyclic ring or a 4- to 7-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C1_4 alkoxy.
The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S
and N.
[0085] In some embodiments, Rc and RL together with the atoms attached thereto form a C3-6 carbocyclic ring or a 4- to 7-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0086] In some embodiments, two RB, or RL and Rc are directly connected (between two RB, or RL and Rc) to form C4_6carbocyclic ring or a 4- to 7-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, 01-1, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0087] In some embodiments, RL and Rc are directly connected to form a 4, 5, 6 or 7-membered heterocyclic ring. The heterocyclic ring is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, F, =0, OH, CN, NH2, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl or methoxy. The heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[0088] Tn some embodiments, Rc and RL together with the atoms attached thereto form a 4- to 7-membered heterocyclic ring. The heterocyclic ring is further substituted with 0, 1, 2, 3 or 4 substituents selected from H, F, methyl or ethyl. The heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[0089] In some embodiments, RB and R2 together with the atoms attached thereto form a C3-6 carbocyclic ring or a 4- to 7-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NI-12, C1_4 alkyl, C3-6 cycloalkyl or C14 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0090] As the first embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0091] Ring B is selected from a 4- to 12-membered heterocyclic ring, C5-12 carbocyclic ring, C6-10 aromatic ring or a 5- to 12-membered heteroaromatic ring. The carbocyclic ring, heterocyclic ring, aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) R. The heterocyclyl or heteroaromatic ring contains 1 to 3 (for example 1, 2 or 3) heteroatoms selected from 0, S
and N.
[0092] Ring C is selected from C6-10 carbocyclic ring, a 5- to 10-membered heterocyclic ring, C6_10 aromatic ring or a 5- to 10-membered heteroaromatic ring. The carbocyclic ring, heterocyclic ring, aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc. The heterocyclic ring or heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0093] L is selected from a bond, 0, S, -NRL-, -C(RL)2-, -C(RL)2-C(RL)2-, -Y-C(RL)2-, -C(RL)2-Y-, -Y-C(RL)2-C(RL)2-, -C(RL)2-C(RL)2.-Y- or -C(RL)2-Y-C(RL)2.-.
[0094] Y is selected from 0, S or -NRL-.
[0095] Ring D is selected from a 6- to 10-membered aromatic ring or a 5- to 12-membered heteroaromatic ring. The aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 5 RD. The heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0096] Ring E is selected from a 5-membered heterocyclic ring, and the heterocyclic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NI-12, C1_6 alkyl, C1_6 alkoxy or C3-6 cycloalkyl. The heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[0097] R1 is selected from H, halogen, OH, -SH, CF3, CN, C1_6 alkyl, C1_6 alkoxy, -C1-3 alkylene-Z-Co_3 alkylene-Ri a, -00_4 alkylene-Ria or -N(R1b)2. The alkyl, alkoxy, and alkylene are optionally further substituted with 0 to 4 (such as 0, 1 , 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(R16)2, C1-6 alkyl, halogen-substituted C1-6 alkyl, hydroxy-substituted C1_6 alkyl, cyano-substituted C1_6 alkyl, C1_6 alkoxy, C3_6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl, a 5- to 10-membered heteroaryl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0098] Z is selected from a bond, N(Rib), 0 or S.
[0099] R a is selected from C1_6 alkyl, C1_6 alkoxy, C3_8 cycloalkyl, a 3- to 8-membered heterocycloalkyl, a 6- to 10-membered aryl or a 5- to 12-membered heteroaryl.
The cycloalkyl, heterocycloalkyl, aryl or heteroaryl is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, C1-6 alkyl, C1_6 alkoxy or -N(R16)2. The alkyl or alkoxy is optionally further substituted with 0 to 4 (such as 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, C1-6 alkyl, Ci-6alkoxy or C3-6 cycloalkyl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.

[0100] Each R11) is independently selected from H or C1-6 alkyl;
the alkyl is optionally further substituted with 0 to 3 substituents selected from H, halogen, =0, CN, OH, NI-12, C1-6 alkyl, C1-6 alkoxy or C3-6 cycloalkyl.
[0101] R2 is selected from H, halogen or Ci_6alkyl.
[0102] Each RL is independently selected from H, halogen, -SH, CN, OH, CF3, NH2, C1 6 alkyl, Ci 6 alkoxy, C36 cycloalkyl. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy.
[0103] Each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, Ci _6 alkyl, C3_6 cycloalkyl or Ci _6 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy.
[0104] Alternatively, two RB, two Rc, two RD, Rc and RL, RD and RL, or RB and R2 and the atoms attached thereto together form a C3_iocarbocyclic ring or a 3-to 10-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or Ci_6 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, Ci-6alkyl, C3-6 cycloalkyl or C1-6 alkoxy. The said heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0105] r is selected from 0 or 1.
[0106] As the second embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0107] Ri is selected from H, halogen, OH, -SH, CF3, CN, C14 alkyl, C14 alkoxy, -C1-2 alkylene-Z-Co_2 alkylene-Ria, -Co-4 alkylene-Ria. The alkyl, alkoxy, and alkylene are optionally further substituted with 0 to 4 (such as 0, 1 , 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(Rib)2, Ci_4 alkyl, halogen-substituted C14 alkyl, hydroxy-substituted C14 alkyl, cyano-substituted C14 alkyl, C14 alkoxy, C3-6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl, a 5- to 6-membered heteroaryl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0108] Ri a is selected from C14 alkyl, Ci_4 alkoxy, C3-6 cycloalkyl, a 3- to 6-membered heterocycloalkyl, phenyl or a 5- to 6-membered heteroaryl. The cycloalkyl, heterocycloalkyl, phenyl or heteroaryl is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, Ci4 alkyl, Cl 4 alkoxy or -N(Rib)2. The alkyl or alkoxy is optionally further substituted with 0 to 4 (such as 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, CN, OH, -N(Rib)2, C14 alkyl, C14 alkoxy or C3-6 cycloalkyl. The heterocycloalkyl or heteroaryl contains 1 to 3 (for 1() example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0109] Each Rib is independently selected from H or C1_4 alkyl;
the alkyl is optionally further substituted with 0 to 3 substituents selected from H, halogen, =0, CN, OH, NH2, C14 alkyl, C1_4 alkoxy or C3-6 cycloalkyl.
[0110] Each RL is independently selected from H, halogen, CN, OH, -SH, CF3, NH2, Ci_4 alkyl, C1_4 alkoxy, C3-6 cycloalkyl. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C1_4 alkoxy.
[0111] Each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, C1_4 alkyl, C3_6 cycloalkyl or C14 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or Ci _4 alkoxy.
[0112] Alternatively, two RB, two Rc, two RD, Rc and RL or RB
and R2 and the atoms attached thereto together form a C3_7carbocyclic ring or a 4- to 7-membered heterocyclic ring. The carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C14 alkyl, C3-6 cycloalkyl or C14 alkoxy. The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C14 alkyl, C3-6 cycloalkyl or C14 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0113] The definitions of other groups are the same as those in the first embodiment.
-19-1101141 As the third embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0115] Ring B is selected from a benzene ring, a 4- to 8-membered heterocyclyl, C5-8 carbocyclyl or a 5- to 6-membered heteroaromatic ring. The benzene ring, carbocyclyl, heterocyclyl or heteroaromatic ring is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) RB, the heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0116] Ring C is selected from a benzene ring, a 5- to 6-membered monocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6-membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring. The benzene ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc, the heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0117] Ring D is selected from a benzene ring, a naphthalene ring, a 5- to membered monocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6-membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring. The benzene ring, naphthalene ring or heteroaromatic ring is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RD, the heteroaromatic ring contains 1 to 5 (for example, 1, 2, 3, 4 or 5) heteroatoms selected from 0, S and N.
[0118] Ring E is selected from a 5-membered non-aromatic heterocyclic ring or a 5-membered heteroaromatic ring, and the heterocyclic ring or heteroaromatic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NH2, C14 alkyl, C14 alkoxy or C3-6 cycloalkyl, the heterocyclic ring or heteroaromatic ring contains 1 to 2 heteroatoms selected from 0, S and N.
[0119] R1 is selected from H, F, Cl, OH, CN, CF3, methyl, ethyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, -ethylene-Z-methylene-Ri a, -ethylene-Z-ethylene-Ri a, -ethylene-Z-Ri a, -RI a, -methylene-R 1 a, -ethylene-RI a. The methyl, ethyl, propyl, methoxy, ethoxy, methylene, ethylene are optionally further substituted with 0 to 3 substituents selected from H, F, Cl, =0, CN, OH, -N(R1b)2, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1
- 20 -to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyri midi nyl , pyrazinyl or pyridazinyl [0120] Z is selected from 0, S or N(R 10-[0121] Ri a is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyrimidinyl, pyrazinyl or phenyl. The Ri a is optionally further substituted with 0 to 3 substituents selected from H, F, =0, OH, CN, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy or -N(Rib)2.
[0122] Each Rib is independently selected from H, methyl or ethyl.
[0123] R2 is selected from H, F, methyl or ethyl.
[0124] Each RL is independently selected from H, F, Cl, CN, CF3, OH, NH2, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl or cyclopentyl.
[0125] Each RB, Rc or RD is independently selected from H, F, Cl, =0, CN, CF3, OH, NH2, methyl, ethyl, propyl, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, propoxy, isopropoxy, tert-butyloxy, trifluoromethoxy, cyclopropyl, cyclobutyl or cyclopentyl.
[0126] Alternatively, RB and R2 together with the atoms attached thereto form a C3-6 carbocyclic ring (for example, C3, C4, C5 or C6 carbocyclic ring) or a 4- to 7-membered
- 21 -(for example, 4, 5, 6 or 7-membered) heterocyclic ring. The carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or C1_4 alkoxy.
The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH), C1_4 alkyl, C3-6 cycloalkyl or C14 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0127] Alternatively, Rc and RL together with the atoms attached thereto form a C3-6 carbocyclic ring (for example, C3, C4, C5 or C6 carbocyclic ring) or a 4- to 7-membered (for example, 4, 5, 6 or 7-membered) heterocyclic ring. The carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH2, C1_4 alkyl, C3-6 cycloalkyl or Ci_4 alkoxy.
The alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 (for example 0, 1, 2, 3 or 4) substituents selected from H, halogen, =0, OH, CN, NH), C1_4 alkyl, C3-6 cycloalkyl or C14 alkoxy. The heterocyclic ring contains 1 to 3 (for example, 1, 2 or 3) heteroatoms selected from 0, S and N.
[0128] The definitions of other groups are the same as those in any one of the first and second embodiments.
[0129] As the fourth embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0130] L is selected from a bond, 0, S, -NRL-, -CHRL-, -CHRL-CHRL-, -Y-CHRL-, -CHRL-Y-, -Y-CHRL-CHRL-, -CHRL-CHRL-Y- or -CHRL-Y-CHRL-.
[0131] Each RL is independently selected from H, F, methyl, ethyl or propyl.
[0132] Y is selected from 0, S or -NRL-.
[0133] Ring B is selected from one of the following substituted or unsubstituted groups: cyclohexyl, cyclohexenyl, azacyclohexenyl, piperidinyl, phenyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RB.
- 22 -[0134] Or ring B is selected from one of the following substituted or unsubstituted groups: piperazinyl, tetrahydropyrrolyl or 1,4-diazepanyl, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RE.
[0135] Ring C is selected from one of the following substituted or unsubstituted groups: A benzene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a tetrazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) Rc.
[0136] Ring D is selected from one of the following substituted or unsubstituted groups: A benzene ring, a naphthalene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 (for example, 0, 1, 2, 3 or 4) RD.
N

N /
110137] When r=1, ( is selected from N
N N N
[0138] When r=0, ( is selected from , `1"--N N N
, , NI-1?'= H 0 , , , --N N N
N \ N N
S jl ,,i,1 H 0 S
, , or .
[0139] The definitions of other groups are the same as those in any one of the first, second and third embodiments.
- 23 -[0140]
As the fifth embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0141]
L is selected from -Y-CHRL-, -CHRL-Y-, -CHRL-CHRL-, -CHRL-CHRL-Y- or -Y-CHRL-CHRL-.
[0142] Y is selected from 0, S or -NRL-.
[0143] Each RL is independently selected from H or methyl.
[0144]
Ring B is selected from one of the following substituted or unsubstituted N AN N
groups: µV-) , , , , , or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.
[0145]
Or ring B is selected from one of the following substituted or unsubstituted '?,õN_, ) 1--N¨e_ / i¨N
groups: , C \.,,s_v j or , the left side of which is directly --connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.
[0146]
Or ring B is selected from one of the following substituted or unsubstituted -LI\J)c N

N_,c, I\ j -- N
groups: N
, , , , , , I I
,-, or ''l , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB.

[0147] Or 0 is selected from or kN
, the left side of which is directly connected to ring C, and which, when the 6-membered ring in the spiro ring is substituted, is optionally further substituted with 1, 2 or 3 RB.
[0148]
Ring C is selected from one of the following substituted or unsubstituted -1`1=Nik (x`1\1)4- eCNA- NO$-groups: a benzene ring, ¨/ , \--/
N
or N
, which, when substituted, is optionally further substituted with 1, 2 or 3 Rc, and the left side of which is connected to L.
[0149]
Ring D is selected from a benzene ring or a pyridine ring, and the benzene ring or pyridine ring is optionally further substituted with 1, 2 or 3 R.
[0150]
Each RB is independently selected from H, =0, F, OH, CN, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy.
[0151]
Each Rc is independently selected from H, F, Cl, OH, CN, NH2, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy.
[0152] Each RD is independently selected from H, F, Cl, CN, CF3, CHF2, methyl, ethyl, methoxy, ethoxy, tert-butyloxy or trifluoromethoxy.
[0153]
Alternatively, Rc and RL together with the atoms attached thereto form a 4-to 7-membered heterocyclic ring. The heterocyclic ring is further substituted with 0, 1, 2, 3 or 4 substituents selected from H, F, methyl or ethyl. The heterocyclic ring contains 1 to 2 heteroatoins selected from 0, S and N.
[0154]
The definitions of other groups are the same as those in any one of the first, second, third and fourth embodiments.

[0155]
As the sixth embodiment of the present invention, the compound represented by the above general formula (I) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, [0156] L is selected from -CH20-, and the right side of L is connected to ring C.
[0157] R1 is selected from H, CN, CF3, CHF2, CH2F, -CH2OH, -CH(OH)CH3, methyl, ethyl, methoxy, ethoxy, propoxy, isopropoxy, methoxymethyl, methoxyethyl, ethoxymethyl, isopropoxymethyl, -ethylene-Z-methylene-R I a, -ethylene-Z-ethylene-R I a, -ethylene-Z-R 1 a, -R1 a or -methylene-R1 a-[0158] Z is selected from NH, N(C113) or 0.
[01591 Ri a is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, pyrrolidinyl, piperidinyl, pyrazolyl, 1,2,3-triazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, 1,2,4-oxadiazolyl, isothiazolyl, pyrimidyl, pyridine or phenyl. The Ri a is optionally further substituted with 0, 1, 2 or 3 substituents selected from F, CN, methyl, ethyl, propyl, CF3, methoxy or ethoxy.
[0160]
Ring C is selected from one of the following substituted or unsubstituted N \
groups: or , which, when substituted, is optionally further substituted with 1, 2 or 3 Rc, and the left side of which is connected to L.
[0161] Ring D is selected from a benzene ring or pyridine, and the benzene ring or pyridine is optionally further substituted with 1, 2 or 3 RD.

L
[0162] Or is selected from , the right side of which is connected to ring B, and in which the benzene ring is optionally further substituted with 1, 2 or 3 Rc.
[0163] Each RB is independently selected from H, =0, F, CN, CF3, methyl, ethyl or cyclopropyl.

[0164] Each Rc is independently selected from H, F, Cl, CN, CF3, methyl, ethyl, methoxy or ethoxy.
[0165] Each RD is independently selected from H, F, Cl, CN, CF3, CHF?, methyl or ethyl.
[0166] The definitions of other groups are the same as those in any one of the first, second, third, fourth and fifth embodiments.
[0167] As the seventh embodiment of the present invention, the compound represented by the above general formula (1) or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, wherein, -kk \..k 0 [0168] R1 is selected from (c gi or [0169] R2 is selected from H;
[0170] Ring B is selected from one of the following substituted or unsubstituted r-N )r( is+j L1/2N.
groups: or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB;
R2 ,R2 B
[0171] Or is selected from , the left side of which is directly connected to ring C, and which, when the 6-membered ring in the Spiro ring is substituted, is optionally further substituted with 1, 2 or 3 RB;
N N N
/ II / S II /
N
[0172] r=0, is selected from or N
[0173] The definitions of other groups are the same as those in any one of the first, second, third, fourth, fifth and sixth embodiments.

[0174] The present invention provides the following compound or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, C)--NC. ,--,N,---,,N NC, --, ,,N
,)..._s I [I, \ _, N õCOON
F y (:) N) N---{ 1 .- OH
II

O,-- 0--NC NC--,0õN -,, ) 14-4 N(/ OH
F
-, =Ns----H.,i 0 H
F ,--,-.- S f ('=), NC 'Isl-')--N \. NC N.
N
'I 1 ,,Z) N ,N) \ ---c-N
1 ' 11, /'-\'_ 7 I s'y OH õ, F F N--.F ,N F N, NC- --;(- \ \, ir--S NC 7 ,.0_1:7N-- N--=( 0 \ 7reN-Z N---'k . -0 OH t_z OH

,F iz---,f1õ,,,.Nh NC, . r ,0 , ._- ON õ -i J N /\s ', '1 1 OH
7 OH F J. 0 C)-- 0-F F

N

-_, NC N =-''") OH
N
S \S--o cr:1 oTh F
NC, F )___N

/
-1- 0 N, y r IV-------li N 11 OH
N
S
- ---s-- -r0 NC

-I
F

, C1,,,_ N T\
CI NII*1/

(:),INI. ) N--- \ OH
N

OH

1 t F0 , 2----, 0 NN r '14-' _ NC z--- 7F 1 f----11_1-11. 1 j N-j<
- N ¨ \S )21 NC 1-s OH
If ) 7) OH F -04N 10 ----...=
0 --\
F
F Cci-/
NC F
-N
N )1-N '-''''j NOH
r NC
S

OH

F
NC),_ F OTh NC _ F /
N
1 11 )------N---< -K C:$
L
S"---) S
r r O OH H

NC- F
N
N
0 N'll / 1 0 N

N / \
S

S
OH
OH
(:)-- NC F
NC Ai F
1\1 N

\-\'' /
S

S
OH
OH

CI F

--õ, ---- /LO r'il''N
' 0 'S S
---, .-.:"- OH
OH

0---) CI N
-Nil- 1;

OH OH
CI F Cr,- 1:1J--1, I\10õ ,.-------....-Nz 0 rN7C N 1 1 r N,,) \

OH

OH
0--a ON
/
NC,, ''N" _ N, NC, .-7 0 N, ,-, ) IV----=c' OH r',i /
' 1 11 'I S ; :r s iOH
F

0--J\
I NC, F /
NC
r N--;-N,,,_ 1 J 1:1 -..<' J. ,0 N õN T ---`-,10 N,,, -J N /
\ OH T s If' 'S 0 F

OH
I
NCF (c-aN F 0--\
i NC F
..-- ---_-- :Is /
11 /µ>-----, A 0 N -L 3 0 ) N-Z( 11 - ---- --.Tr 7.
µS"
OH
OH
.-, I N ' N----,µ
NC ) N
F - N NC / r , , ,,.----' F -J ---- N
-..N ,----i: - ni...'1, . 1 ,::: NJ ) N-I S --1:) ( 1 .

OH ) \S
r OH

NN---- (-0 N=--/
\
N / NC _F 7 NC
(--Isr--1-N\
--y..,---c , li- ' ¨
N -,õ_rk_,0 J N /
I
- _L
0 S- y r , OH
OH
,------NN' 0 0\ 7 \ ---( \?
1 , ,-: F i NC ,F ^ N
NC
JIIIIIIN'ThiNs r -.- 7 -N _ ON N-2-N N, 1 N = ri 1 s , \S

OH
OH
,.. 7-0\
( 0--) F

NC
N.N __- ,-r-N1---N
--L 0N N4 \ XI, N
0 - ---, =,--.1.--- /-- -s. y0 'S-I
OH
OH
\, 0¨,/ OTh CI
NC F F
=NI N
L i 0 N CNII N / I .
ON)N / II
- -..---- -_.- ..õ===õ,.7- --õ,---I s -, --:-OH
OH
0-a 0¨

NC F /
N
NC , ,,, F
'Y' r ( ICI N
--. N-I
ji:>'1--.1%1 \-1 il sS

j S 1%0 OH F' OH
I I
Om OTh NC -_, c N
NC F .N ^ N 'r ,r-F
-'rslI .; r -',--:, i ON, (J r',i / 11 .ON N---I
')-' .--0 S y-r U -, F-OH - "F
OH
I I

0 \
--\
/ NC F / NC F
. -'? 11-\ ,O, N
N-- -;
OH 1-' OH
-/ ''' NC,. F N--- _______________________________________ 0 i--N---T,-N
0õN_ N.,) N- )---1, NC ,_,,r F /---N, N¨S OH
,10 --..zi0 N.r ,- OH J
, o crD-3 \
b2----_,\ , N_ _ 0 NC, F ,)-L. , .N
NC F ,-- t / , I r N -'''' N N" S OH ' -I o N.
.-- ---,--- - - - -- ----- \
\
-C a r-- 1 'T s----r-_. y_,N.. 0, ,._ OH
N_J--0---J) Oa \ 0 i NC F )t NC .r F )1 . ----N
,, N N
Nil - ')1 I ) /
0õ, NN N \
'--j-N,0 ,N N_____c 3 õ, 1 s 0 , O
OH H

CC-J) N
j NC F _, N ..,,---.õ,,,õ N
NC ,- F N ,_,, õN I 11 /
0,,_, N,,,,,,,,.. N N \ 0 - ...0õ ,N, s_ 0 I s OH
OH
CrD-N
[-_;\ i / NC .,_ , F N.,..
NC ,-,..., F _N..----T-1,:q 'r T \ ---, -),-- ir N As! 0 0 - = ...---------r / OH -. OH
\
ga, OA) /
NC,,,,r- F N.. ---,_ õN
NC F N ,-- ,N 11 r - iS
'r: =----- N' 1- '')'' s;,N N -4. n ---,1- N \ µ-'' -c-- õ,-- - ------E, -,-, r OH
OH
j ----1 ) , _N NC ,c NC , I 0 N, --, - N -----< \ 0 ----, -,,, --,_, 1 .7- N µs- ---t-0 N. ,,,- N-----C
F ---.--- OH
F OH

CL , _F

0 I Iii____ F
NC , )1- N 0 --1 Nil 1-1-1 ,0 N - N N-J, -1 0 \
S COOH ) r N-CI, 0 CI , F
p...../2 1 0 -CI 6--tejµ I / µ= 1 11 /)--F 0, ), '..-, , N--_,/ ,Ai I 'S '0 ,--\S- 'CO2H
OH
CI F F CI ,_ F
I
0 r_00 r N
;I

-\S- 'CO2H
CI -1----,-;" -----I I i 01) !--0 ^-7.N___ F 0 \ L N--/C), F

-I
OH OH
CI ,.,-, Oq CI, I /
F N i0 0, ',.-U
i 'S -' OH OH
CI ,,, 01-i NC 011--1, , /
FO \ I F 0 N
N \ ' N-__ \

I S
OH

CN
0 ----.
/ \\ I > <, F 1 ( N
õ,j0õN1,,,,N, _I Y, I I N T
%1 z F
Oa\ 1:1-.N
/ F i NC ).N NC, N
0 N, N---- I
----- 0 ,r=-0,..,N1,---, N 8,,,\ 17) I , i 1 r F F OH
OH

C)-- C)--F F
NC ,N NC , .., ,I,c r=lz_ - (:) fq,, (:)N,---.j N--_< \ ,õ
F
F '-' OH F OH
F O
i F
NC N NC

N
, I ' s F OH
OH
0-1 F NC y, F
F
rel NC 1., ,?.

lj ON, N- =< I , N.,.y..- ry ___<, , i =.- r- ,u1 F

OH
J ] 0 IN NC F 0 \ NC F
F F
N N
C:) N N --{ \ ()N
' I sS--- CO2H 5- CO2H
F F F
F
NC
F ,F 0--7 NC 0---, N
0 N N ---/0 ,)_ 0 N N---c __1 S¨ CO2H ,--- T.
's CO2H
F F F

CI
0-1) 1 Th ,rJN N-0 0 N} T N
s co2H s y õ F , OH
CI
CF)-- CI

N

Nli.-- 0 N N"10 N NMI' /
O\1J N 0 / \ 0 N \

S S
OH
OH

o z 1µ171 z OH OH
N7fl N--\
CI O\ ¨
, / NC
r 0 rF
J N /

OH
OH
[0175] The present invention relates to a pharmaceutical composition comprising the above-mentioned compound or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and a pharmaceutically acceptable carrier.
[0176] The present invention relates to the above-mentioned compound or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, or an application of the above-mentioned pharmaceutical composition in the preparation of a drug for treating diabetes.
[0177] A compound has a structure as shown below, on X N
N

0 Rm [0178] X is selected from H or Br;
[0179] Rm is selected from H or C1_4alky1; the alkyl is optionally further substituted with 0 to 4 substituents selected from halogen, C1-4 alkyl, C1-4 alkoxy or C6-10 carbocyclic ring.
[0180] Synthetic method I:

R1111 Ri NH2 Rml HN
E COORm2 __ 02N / COO IRm2 __ (1-3) 02N)COORrn2 (I-1) (1-2) (1-4) V / OL C OH
HN
_____________________________________ 1Rm3 H2N E COORrr2 (1-7) N E
COORm2 (1-5) (1-6) I / /
COORm2 N E COOH
[0181] The compound of general formula (I-1) is nitrated to obtain the corresponding compound of general formula (I-2).
[0182] The compound of general formula (I-2) and the compound of general formula (I-3) are subjected to a substitution reaction to obtain the corresponding compound of general formula (I-4).
[0183] The compound of general formula (I-4) is reduced to obtain the corresponding compound of general formula (I-5).
[0184] The compound of general formula (I-5) is subjected to a ring-closing reaction to obtain the corresponding compound of general formula (I-6).
[0185] The compound of general formula (I-6) and the compound of general formula (I-7) are subjected to a substitution reaction to obtain the corresponding compound of general formula (I-8).
[0186] The compound of the general formula (I-8) is subjected to a decarboxylation protecting group reaction to obtain the corresponding compound of the general formula (I).
[0187] RI and R1113 are selected from F, Cl, Br, I, OTf, etc.
[0188] Rm2 is selected from carboxyl protecting groups, preferably Ci_6alkyl or benzyl.
[0189] The definitions of other groups are the same as those in any embodiment of the above general formula (I).
[0190] Unless stated to the contrary, the terms used in the description and claims have the following meanings.

[0191] The carbon, hydrogen, oxygen, sulfur, nitrogen or F, Cl, Br, I involved in the groups and compounds of the present invention all comprise their isotopes, and the carbon, hydrogen, oxygen, sulfur or nitrogen involved in the groups and compounds of the present invention is optionally further substituted with one or more of their corresponding isotopes, wherein the isotopes of carbon comprise 12C, '3C and 14C, the isotopes of hydrogen comprise protium (H), deuterium (D, also known as heavy hydrogen), tritium (T, also known as superheavy hydrogen), the isotopes of oxygen comprise 160, 170 and 180, the isotopes of sulfur comprise 32S, 33S, 34S and 36S, the isotopes of nitrogen comprise 14N and 15N, the isotopes of fluorine comprise 17F and 19F, the isotopes of chlorine comprise 35C1 and 37C1, and the isotopes of bromine comprise 79Br and 81Br.
[0192] "Halogen" means F, Cl, Br or I.
[0193] "Halogen-substituted" refers to F, Cl, Br or I
substitution, including but not limited to a substitution with 1 to 10 substituents selected from F, Cl, Br or I, a substitution with 1 to 6 substituents selected from F, Cl, Br or I, a substitution with 1 to 4 substituents selected from F, Cl, Br or I. "Halogen-substituted" is referred to simply as "halo".
[0194] "Alkyl" means a substituted or unsubstituted linear or branched saturated aliphatic hydrocarbon group, including but not limited to an alkyl group of 1 to 20 carbon atoms, an alkyl group of 1 to 8 carbon atoms, an alkyl group of 1 to 6 carbon atoms, or an alkyl group of 1 to 4 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neobutyl, tert-butyl, n-pentyl, isoamyl, neopentyl, n-hexyl and various branched isomers thereof; The definition of the alkyl described herein is consistent with this definition. Alkyl group can be monovalent, divalent, trivalent or tetravalent.
[0195] "Heteroalkyl" refers to a substituted or unsubstituted alkyl group in which one or more (including but not limited to 2, 3, 4, 5 or 6) carbon atoms are replaced by heteroatoms (including but not limited to N, 0 or S). Non-limiting examples include -X(CH2)v-X(CH2)v-X(CH2)v-H (v is an integer from 1 to 5, X is independently selected from bonds or heteroatoms, heteroatoms include but not limited to N, 0 or S, and at least one X is selected from heteroatoms, and N or S in heteroatoms can be oxidized to various oxidation states). Heteroalkyl group can be monovalent, divalent, trivalent or tetravalent.

[0196] An "alkylene" means a substituted or unsubstituted straight or branched chain divalent saturated hydrocarbon group, including -(CH2),- (v is an integer from 1 to 10), and examples of alkylene include, but are not limited to, methylene, ethylene, propylene, butylene, etc.
[0197] "Heteroalkylene" means a substituted or unsubstituted alkylene group in which one or more (including but not limited to 2, 3, 4, 5 or 6) carbon atoms are replaced by heteroatoms (including but not limited to N, 0 or S). Non-limiting examples include -X(CH2)v-X(CH2)v-X(CH9)v-, wherein v is an integer from 1 to 5, each X is independently selected from a bond, N, 0 or S, and at least one X is selected from N, 0 or S.
[0198] "Cycloalkyl" means a substituted or unsubstituted saturated carbocyclic hydrocarbon group, usually having from 3 to 10 carbon atoms, and non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, etc.
The "cycloalkyl" herein is as defined above. Cycloalkyl group can be monovalent, divalent, trivalent or tetravalent.
[0199] "Heterocycloalkyl" means a substituted or unsubstituted saturated heteroatom-containing cyclic hydrocarbon group including but not limited to 3 to 10 atoms, 3 to 8 atoms, and 1 to 3 atoms selected from N, 0 or S. N and S selectively substituted in heterocycloalkyl ring can be oxidized to various oxidation states. The heterocycloalkyl group can be connected to a heteroatom or a carbon atom, the heterocycloalkyl group can be connected to an aromatic ring or a non-aromatic ring, and the heterocycloalkyl group can be connected to a bridged ring or a spiro ring. Non-limiting examples include oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl or piperazinyl. Heterocycloalkyl group can be monovalent, divalent, trivalent or tetravalent.
[0200] "Alkenyl" means a substituted or unsubstituted straight and branched unsaturated hydrocarbon group having at least 1, usually 1, 2 or 3 carbon-carbon double bonds, with a main chain including but not limited to 2 to 10, 2 to 6, or 2 to 4 carbon atoms. Examples of alkenyl include, but are not limited to vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-1-butenyl, 2-methyl-1-butenyl, 2-methyl-3-butenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-1 -pentenyl, 2-methyl-1 -pentenyl, 1-heptenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 1-octenyl, 3-octenyl, 1-nonenyl, 3-nonenyl, 1-decenyl, 4-decenyl, 1,3-butadiene, 1,3-pentadiene, 1,4-pentadiene, 1,4-hexadiene, etc.
The definition of the alkenyl described herein is consistent with this definition. Alkenyl group can be monovalent, divalent, trivalent or tetravalent.
[0201]
"Alkynyl" means a substituted or unsubstituted straight and branched monovalent unsaturated hydrocarbon group having at least 1, usually 1, 2 or 3 carbon-carbon triple bonds, with a main chain including but not limited to 2 to 10 carbon atoms, 2 to 6 carbon atoms or 2 to 4 carbon atoms. Examples of alkynyl include but are not limited to ethynyl, propargyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-1-butynyl, 2-methyl-1-butynyl, 2-methy1-3-butynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5- hexynyl, 1-methyl- 1 -pentynyl, 2-methyl-1 -pentynyl, 1-heptynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 1-octynyl, 3-octynyl, 1-nonynyl, 3-nonynyl, 1-decynyl, 4-decynyl, etc. Alkynyl group can be monovalent, divalent, trivalent or tetravalent.
[0202]
"Alkoxy" means a substituted or unsubstituted -0-alkyl group. Non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexyloxy, cyclopropoxy and cyclobutoxy.
[0203]
"Carbocycly1" or "carbocyclic ring" means a substituted or unsubstituted saturated or unsaturated aromatic ring or non-aromatic ring, and the aromatic ring or non-aromatic ring can be 3- to 8-membered monocyclic ring, 4- to 12-membered bicyclic ring or 10- to 15-membered tricyclic ring system. The carbocyclyl can be connected to an aromatic ring or a nonaromatic ring, and the aromatic ring or nonaromatic ring is optionally a monocyclic ring, a bridged ring or a spirocyclic ring. Non-limiting examples include cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, 1-cyclopentyl-1-enyl , 1-cyclopenty1-2-enyl, 1-cyclopenty1-3-enyl, cyclohexyl, 1-cyclohexyl-d2-enyl, 1-cyclohexy1-3-enyl, cyclohexenyl, benzene ring, naphthalene ringõ
Cl>, or . A "carbocycly1" or "carbocyclic ring" can be , monovalent, divalent, trivalent or tetravalent.

[0204]
"Heterocycly1" or "heterocyclic ring" refers to a substituted or unsubstituted saturated or unsaturated aromatic ring or non-aromatic ring, and the aromatic ring or non-aromatic ring can be 3- to 8-membered monocyclic ring, 4- to 12-membered bicyclic ring or 10- to 15-membered tricyclic ring system, and contains one or more (including but not limited to 2, 3, 4 or 5) heteroatoms selected from N, 0 or S, and the optionally substituted N or S in the heterocyclyl ring can be oxidized into various oxidation states.
Heterocyclyl can be connected to other groups through its heteroatoms or carbon atoms, heterocyclyl can be connected to other groups through its aromatic ring or nonaromatic ring, and heterocyclyl can be connected to a bridged ring or a Spiro ring (when the ring of the heterocyclyl is a bridged ring or a spiro ring, the site where the heterocyclyl is connected to other groups is on the bridged ring or Spiro ring). Non-limiting examples include oxiranyl, azacyclopropyl, oxetanyl, azetidinyl, 1,3-dioxolane, 1,4-dioxolane, 1,3-dioxane, azacycloheptyl, pyridyl, furanyl, thienyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, piperidyl, morpholinyl, thiomorpholinyl, 1,3-dithianyl, dihydrofuranyl, dihydropyranyl, dithiolanyl, tetrahydrofuranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydropyranyl, benzimidazolyl, benzopyridyl, pyrrolopyridyl, benzodihydrofuranyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, pyrazinyl, indazolyl, benzothiophenyl, benzofuranyl, benzopyrrolyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzopyridyl, benzopyrimidinyl, benzopyrazinyl, piperazinyl, azabicyclo[3.2.11octyl, azabicyclo [5 .2.0]nonalkyl , oxatricyclo[5.3.1.11dodecyl, azaadamantyl, oxaspiro[3.31heptyl, 0 0,7 FC1 , Nr>
N .ccN
----- (ON,N 0 N
N N or . A "heterocyclyl" or "heterocyclic ring" can be monovalent, divalent, trivalent or tetravalent.
[0205] "Spiro ring" or "Spiro ring group" means a polycyclic group that shares one atom (called Spiro atom) between substituted or unsubstituted inonocyclic rings. The number of ring atoms in the Spiro ring system includes but is not limited to 5 to 20. 6 to 14, 6 to 12, or 6 to 10, wherein one or more rings may contain 0 or more (including but not limited to 1, 2, 3 or 4) double bonds, and can optionally contain 0 to 5 heteroatoms selected from N, 0 or S(=0), /---\\_.,;
i--I X/ X x ,0 xr3 x \I
\ __ /
., NH
/ ________________________________________________________ ' -0 , /---- , ( Y > ><
' \/ ----, s-,--- '---¨0\ \
/ x ----il I
---- / \ ---- v 9, C / \ 7---- NH , 1 - , / ' \
i L) /NH / j / __ \
\
\ \ \ /X\ HN7\>(' X \ NH
\ / / \ /NH
HN ,NH
\/ - - .
[0206] "Spiro ring" or "spiro ring group" can be monovalent, divalent, trivalent or tetravalent.
[0207] "Fused ring" or "fused ring group" refers to a polycyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, wherein one or more rings may contain 0 or more (including but not limited to 1, 2, 3 or 4) double bonds, and may be substituted or unsubstituted, and each ring in the fused ring system may contain 0 to 5 heteroatoms or groups containing heteroatoms (including but not limited to N, S(=0)n or 0, n is 0, 1 or 2). The number of the ring atoms in the fused ring system includes but is not limited to 5 to 20, 5 to 14, 5 to 12, or 5 to 10. Non-limiting examples HN-----\NH HN/--------- NH
i I nclude: CL7N1H , ------./ \---------/ .
,0-, 0 0, <__>, i\C>, -"'"----II , N--- ( \
N , ( ':- -------, ---\
-------- S , ----- , <-1-> , ail , C- ''> \
_ _ ,-- ,s¨

(1___);, o- -----\ o ;------\
----/., --< I
) _______________________________ / L __ NH 1 /
------ -----N1 , '------ N
, ---H __s or =
[0208] "Fused ring" or "fused ring group" may be monovalent, divalent, trivalent or tetravalent.
[0209] "Bridged ring" or "bridged ring group" means a substituted or unsubstituted polycyclic group containing any two atoms that are not directly connected, and may contain 0 or more double bonds. Any ring in the bridged ring system may contain 0 to 5 groups selected from heteroatoms or groups containing heteroatoms (including but not limited to N, S(=0)n or 0, wherein n is 0, 1 or 2). The number of ring atoms includes, but is not limited to, 5 to 20, 5 to 14, 5 to 12 or 5 to 10. Non-limiting examples include zr-Ity o' / FIN

HN
, cub ane or adatnantane. "Bridged ring" or "bridged ring group" may be monovalent, divalent, trivalent or tetravalent.
[0210]
"Carbospiro ring", "Spiro ring carbocyclyl", "spirocarbocyclyr or "carbospiro ring group" means a "Spiro ring" with a ring system consisting only of carbon atoms. The definitions of "carbospiro ring", "Spiro ring carbocycly1", "spirocarbocyclyr or "carbospiro ring group" used herein is consistent with that of spiro ring.
[0211]
"Carbo-fused ring", "fused ring carbocycly1", "fused carbocyclyr or "carbo-fused ring group" means a "fused ring" with a ring system consisting only of carbon atoms. The definition of "carbo-fused ring", "fused ring carbocycly1", "fused carbocyclyr or "carbo-fused ring group" used herein is consistent with that of fused ring.
[0212]
"Carbo-bridged ring", "bridged ring carbocycly1", "bridged carbocyclyr or "carbo-bridged ring group" means a "bridged ring" with a ring system consisting only of carbon atoms. The definitions of "carbo-bridged ring", "bridged ring carbocycly1", "bridged carbocyclyr or "carbo-bridged ring group" used herein is consistent with that of bridged ring.
[0213]
"Heteromonocyclic ring", "monocyclic heterocyclyr or "heteromonocycly1"
means "heterocycly1" or "heterocyclic ring" with a monocyclic system. The definition of the heterocyclyl, "monocyclic heterocycly1" or "heteromonocycly1" used herein is consistent with that of heterocyclic ring.

[0214]
"Hetero-fused ring", "hetero-fused ring group", "fused ring heterocycly1"
or "hetero-fused ring group" means a "fused ring" containing a heteroatom. The definition of hetero-fused ring, "hetero-fused ring group", "fused ring heterocycly1" or "hetero-fused ring group" used herein is consistent with that of fused ring.
[0215] "Hetero-spiro ring", "hetero-spiro ring group", "spiro ring heterocycly1" or "hetero-spiro ring group" means a "Spiro ring" containing a heteroatom. The definition of hetero-spiro ring, "hetero-spiro ring group", "Spiro ring heterocycly1" or "hetero-spiro ring group" used herein is consistent with that of Spiro ring.
[0216]
"Hetero-bridged ring", "hetero-bridged ring group", "bridged ring heterocycly1" or "hetero-bridged ring group" means a "bridged ring" containing a heteroatom. The definition of hetero-bridged ring, "hetero-bridged ring group", "bridged ring heterocycly1" or "hetero-bridged ring group" used herein is consistent with that of bridged ring.
[0217]
"Aryl" or "aromatic ring" means a substituted or unsubstituted aromatic hydrocarbon group with a single ring or a fused ring, and the number of ring atoms in the aromatic ring includes but not limited to 6 to 18, 6 to 12 or 6 to 10 carbon atoms. The aryl ring may be fused to a saturated or unsaturated carbocyclic ring or heterocyclic ring, wherein the ring connected to the parent structure is an aryl ring. Non-limiting examples include a benzene ring, a naphthalene ring Or . "Aryl" or "aromatic ring"
may be monovalent, divalent, trivalent or tetravalent. When divalent, trivalent or tetravalent, the point of connection is on the aryl ring.
[0218]
"Heteroaryl" or "heteroaromatic ring" means a substituted or unsubstituted aromatic hydrocarbon group containing 1 to 5 heteroatoms or groups containing heteroatoms (including but not limited to N, 0 or S(=0 )n, n is 0, 1 or 2), and the number of ring atoms in the heteroarotnatic ring includes but not limited to 5-15, 5-10 or 5-6. Non-limiting examples of heteroaryl include, but are not limited to pyridyl, furanyl, thienyl, pyridinyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, benzopyrazole, benzimidazolyl, benzopyridine, pyrrolopyridine and the like.
The heteroaryl ring may be fused to a saturated or unsaturated carbocyclic ring or heterocyclic ring, wherein the ring connected to the parent structure is an heteroaryl ring. Non-limiting examples include fN-s N 0 >
N N N. N.
and . The definition of the heteroaryl used herein is consistent with this definition.
Heteroaryl group can be monovalent, divalent, trivalent or tetravalent. When divalent, trivalent or tetravalent, the point of connection is on the heteroaryl ring.
[0219]
"5-membered ring fused 5-membered heteroaromatic ring" means a 5 fused 5-membered fused heteroaromatic ring, wherein at least one of the two fused rings contains more than one heteroatom (including but not limited to 0, S or N), and the entire group is aromatic. Non-limiting examples include pyrrolopyrrole ring, pyrazolopyrrole ring, pyrazolopyrazole ring, pyrrolofuran ring, pyrazolofuran ring, pyrrolothiophene ring and pyrazolothiophene ring.
[0220]
"5 fused 6-membered heteroaromatic ring" means a 5 fused 6-membered fused heteroaromatic ring, at least one of the two fused rings contains more than one heteroatom (including but not limited to 0, S or N), and the entire group is aromatic.
Non-limiting examples include benzo 5-membered heteroaryl and 6-membered heteroaromatic ring fused 5-membered heteroaromatic ring.
[0221]
"Substitution" or "substituted" means a substitution with 1 or more (including but not limited to 2, 3, 4 or 5) substituents including but not limited to H, F, Cl, Br, I, alkyl, cycloalkyl, alkoxy, haloalkyl, mercaptan, hydroxyl, nitro, sulfhydryl, amino, cyano, isocyano, aryl, heteroaryl, heterocyclyl, bridged ring group, Spiro ring group, fused ring group, hydroxyalkyl, =0, carbonyl, aldehyde, carboxylic acid, formate, -(CH2).-C(=0)-Ra, -0-(CH2)m-C(=0)-R', -(CH2)m-C(=0)-NRbRc, -(CH2)mS(=0)nRa, -(CH2)m-alkenyl-Ra, ORE' or -(CH2)m-alkynyl-Ra (wherein m and n are 0, 1 or 2), arylthio, thiocarbonyl, silyl or -NRbRc and the like, wherein Rb and RC are independently selected from H, hydroxyl, amino, carbonyl, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, sulfonyl, trifluoromethylsulfonyl. Alternatively, Rb and RC may form a five- or six-membered cycloalkyl or heterocyclyl. Ra and Rd are each independently selected from aryl, heteroaryl, alkyl, alkoxy, cycloalkyl, heterocyclyl, carbonyl, ester group, bridged ring group, spiro ring group or bicyclic ring group.
[0222]
"Containing 1 to 5 heteroatoms selected from 0, S or N" means containing 1, 2, 3, 4 or 5 heteroatoms selected from 0, S or N.
[0223]
"Substituted with 0 to X substituents" refers to substituted with 0, 1, 2, 3 ... X
substituents, wherein X is selected from any integer between 1 and 10. For example, "substituted with 0 to 4 substituents" refers to substituted with 0, 1, 2, 3 or 4 substituents.
For example, "substituted with 0 to 5 substituents" refers to substituted with 0, 1, 2, 3, 4 or 5 substituents. For example, "bridged-heterocyclic ring is optionally further substituted with 0 to 4 substituents selected from H or F" means that the bridged-heterocyclic ring is optionally further substituted with 0, 1, 2, 3 or 4 substituents selected from H or F.
[0224]
An X- to Y-membered ring (X is selected from an integer less than Y and greater than or equal to 3, and Y is selected from any integer between 4 and 12) includes X-, X+1-, X+2-, X+3-, X+4-, Y-membered rings. Rings include a heterocyclic ring, a carbocyclic ring, an aromatic ring, aryl, heteroaryl, cycloalkyl, a mono-heterocyclic ring, a fused heterocyclic ring, a spiro-heterocyclic ring or a bridged-heterocyclic ring. For example, "a 4- to 7-membered mono-heterocyclic ring" refers to a 4-membered, 5-membered, 6-membered or 7-membered mono-heterocyclic ring, and "a 5- to 10-membered fused heterocyclic ring" refers to a 5-membered, 6-membered, 7-membered, 8-membered, 9-membered or 10-membered fused heterocyclic ring.
[0225]
"Pharmaceutically acceptable salt" or "pharmaceutically acceptable salt thereof" refers to a salt of the compound of the present invention maintaining the biological effectiveness and characteristics of the free acid or free base, and obtained by reacting the free acid with a non- toxic inorganic base or organic base, reacting the free base with a non- toxic inorganic acid or organic acid.
[0226]
"Pharmaceutical composition" refers to a mixture of one or more compounds of the present invention, or stereoisomers, tautomers, deuterated substances, solvates, prodrugs, metabolites, pharmaceutically acceptable salts or co-crystals thereof and other chemical components, wherein "other chemical components" refer to pharmaceutically acceptable carriers, excipients and/or one or more other therapeutic agents.

[0227] "Carrier" refers to a material that does not cause significant irritation to the organism and does not eliminate the biological activity and characteristics of the administered compound.
[0228] "Excipient" refers to an inert substance added to a pharmaceutical composition to facilitate the administration of a compound. Non-limiting examples include calcium carbonate, calcium phosphate, sugar, starch, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, adhesives and disintegrants.
[0229] "Prodrug" refers to a substance that can be converted into a biologically active compound of the present invention through in vivo metabolism. The prodrug of the present invention is prepared by modifying the amino or carboxyl group in the compound of the present invention, and the modification can be removed by conventional operations or in vivo to obtain the parent compound. When the prodrug of the present invention is administered to a mammalian individual, the prodrug is split to form free amino or carboxyl group.
[0230] The term "co-crystal" refers to a crystal formed by the combination of active pharmaceutical ingredient (API) and co-crystal former (CCF) under the action of hydrogen bonds or other non-covalent bonds. The pure state of API and CCF are both solid at room temperature, and there is a fixed stoichiometric ratio between various components. The co-crystal is a multi-component crystal, which includes both a binary co-crystal formed between two neutral solids and a multi-element co-crystal formed between a neutral solid and a salt or solvate.
[0231] "Animal" is meant to include mammals, such as humans, companion animals, zoo animals, and domestic animals, preferably humans, horses, or dogs.
[0232] The term "stereoisomer" refers to an isomer produced as a result of different spatial arrangement of atoms in molecules, including cis-trans isomers, enantiomers and conformational isomers.
[0233] "Tautomer" refers to a functional group isomer produced by the rapid movement of an atom in two positions in a molecule, such as keto-enol isomerization, amide-imino alcohol isomerization, etc.

[0234] "Optional" or "optionally" or "selective" or "selectively" refers to that the events or conditions subsequently described may but not necessarily occur, and the description includes the case where the events or conditions occur and do not occur. For example, "heterocyclyl optionally substituted with alkyl" refers to that the alkyl may but not necessarily exist, and the description includes the case where the heterocyclyl is substituted with an alkyl group and the case where the heterocyclyl is not substituted with alkyl.
[0235] "TC50" refers to the concentration of a drug or inhibitor required to inhibit half of a given biological process (or a component of the process such as an enzyme, a receptor 1(3 and a cell).
Detailed Description of Embodiments [0236] The technical solutions of the present invention will be described in detail by the following examples, but the scope of protection of the present invention includes but is not limited thereto.
[0237] The compounds used in the reactions described herein are prepared according to organic synthesis techniques known to those skilled in the art, and starting from commercially available chemicals and(or) compounds described in chemical documents.
"Commercially available chemicals" are obtained from regular commercial sources, and suppliers include: Titan Technology Co., Ltd., Energy Chemical Co., Ltd., Shanghai Demo Co., Ltd., Chengdu Kelong Chemical Co., Ltd., Accela ChemBio Co., Ltd., PharmaBlock Sciences (Nanjing), Inc., WuXi Apptec Co., Ltd., J&K Scientific and the like.
[0238] The structures of the compounds are determined by nuclear magnetic resonance (NMR) or (and) mass spectrometry (MS). The NMR shift (6) is given in the unit of 10-6 (ppm). NMR is measured with (Bruker Avance III 400 and Bruker Avance 300) NMR instrument, and the solvent for determination is deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDC13), deuterated methanol (CD30D), and the internal standard is tetramethylsilane (TMS);
[0239] MS is measured with (Agilent 6120B(ESI) and Agilent 6120B(APCI));
[0240] HPLC is determined with Agilent 1260DAD high pressure liquid chromatograph (Zorbax SB-C18 100 x 4.6 mm, 3.5 pM);

[0241] Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate is used as a thin layer chromatography silica plate, and the silica gel plate for the thin layer chromatography (TLC) is of the specification of 0.15 mm-0.20 mm, and the specification when separating and purifying a product by thin layer chromatography is 0.4 mm - 0.5 mm.
[0242] For the column chromatography, Yantai Huanghai silica gel of 200-300 mesh silica gel is generally used as a carrier.
Example 1 (S)-2-((4-(6-((4-cyano-2-fluoroben7y1)oxy)pyridin-2-yl)piperidin-l-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 1) ci--1 NC
N N

F -/-OH
Compound 1 01\.)NH2 F-A H \----A H
Br) 0 Br__, p lc-1 S 0¨ 02N S ¨
Step 1 Step 2 02N'-s 0-Step 3 H2Nlz -S ()¨

la lb lc Id NC, 0 , NH
N (5--NC N, ''NT\
Step 4 CI N S 0¨
Step 5 F I _, le (=) if H
)1\111;N 1g NC N\
N
I].
OH
Compound 1 Step 1: methyl 4-bromo-5-nitrothiophene-2-carboxylate (lc) Br 0 02N /"----S ¨
lb [0243] Under ice-salt bath conditions, la (2.2 g, 10.0 mmol) was added to concentrated sulfuric acid (10 mL), and fuming nitric acid (945.0 mg, 15.0 mmol) was dissolved in concentrated sulfuric acid (5 mL) and slowly added dropwise into the reaction liquid; after the dropwise addition, the reaction was continued for 30 minutes in the ice-salt bath. The reaction liquid was slowly poured into an ice-water solution, and a large amount of white solids precipitated out. The filter cake was collected by filtration, washed with water (10 mL x 2), and dried to obtain lb (2.6 g, yield 99.3%).
[0244] LCMS m/z = 265.9 [M+1]+.
[0245] 'H NMR (400 MHz, CDC13) 6 7.71 (s, 1H), 3.96 (s, 311).
Step 2: methyl (S)-5-nitro-4-((oxetan-2-ylmethyl)amino)thiophene-2-carboxylate (1c) H

-lc [0246] At room temperature, lb (L3 g, 5.0 mmol) was dissolved in acetonitrile (30 mL), then lc-1 (436.0 mg, 5.0 mmol) and potassium carbonate (2A g, 15.0 mmol) were added (100 mL) successively, heated to 60 C, and continued to react for 18 h.
The reaction liquid was cooled to room temperature, filtered, and the filtrate was collected and concentrated. The obtained crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) = 2:1), and concentrated to obtain lc (854.0 mg, yield 62.7%).
[0247] LCMS m/z = 2711 [M+1]+.
[0248] 11-1 NMR (400 MHz, CDCb) 6 8A2 ¨ 8.03 (m, 1H), 7.35 (s, 1H), 5.10 ¨ 5.00 (in, 1H), 4.74 ¨ 4.66 (in, 1H), 4.58 ¨ 4.50 (in, 1H), 192 (s, 3H), 3.71 ¨ 3.57 (in, 2H), 2.80 ¨ 2.68 (m, 1H), 2.60 ¨ 2.48 (in, 1H).
Step 3: methyl (S)-5-amino-4-((oxetan-2-ylmethyl)amino)thiophene-2-carboxylate (1d) H

Id [0249] At room temperature, lc (854M mg, 3.1 mmol) was dissolved in methanol (30 mL), then palladium on carbon (170.0 mg, L6 inmol) was added, and the reaction was continued for 18 h under hydrogen atmosphere. The reaction liquid was filtered to remove palladium on carbon, the filtrate was collected and concentrated to obtain a crude product, and the obtained crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=1:1), and concentrated to obtain 1 d (410.0 mg, yield 53.9%).
[0250] LCMS m/z = 243.1 [M+1] .
[0251] 111 NMR (400 MHz, DMSO - d6) 6 7.15 (s, 111), 5.89 (s, 211), 4.83 -4.74 (m, 1H), 4.55 - 4.47 (m, 1H), 4.47 - 4.39 (m, 1H), 4.23 - 4.15 (m, 1H), 3.66 (s, 3H), 3.25 -3.11 (m, 214), 2.64- 2.55 (m, 111), 2.46 - 2.36 (m, 111).
Step 4: methyl (S)-2-(chloromethyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylate ( 1 e) /

1e [0252] At room temperature, ld (410.0 mg, 1.7 mmol) was dissolved in acetonitrile (10 mL), then le-1 (391.0 mg, 2.5 mmol) and p-toluenesulfonic acid (15.0 mg, 0.1 mmol) were added successively, and finally the reaction liquid was heated to 60 C
for 5 h under the protection of nitrogen. The reaction liquid was cooled to room temperature, saturated sodium bicarbonate solution was added to quench the reaction, extracted with dichloromethane (25 mL x 3), and the organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was directly used in the next reaction without further purification.
[0253] LCMS m/z = 301.0 [M+11+.
Step 5: methyl (S)-24(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-yppiperidin-1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d]imidazole-5-carboxyl ate (10 NC
N Tr=Lch if [0254] At room temperature, if-1 (527.0 mg, 1.7 mmol) was dissolved in acetonitrile (15 mL) solution, and then crude product le (508.0 mg, 1.7 mmol) and potassium carbonate (702.0 mg, 5.1 mmol) were added to the reaction system successively, and heated to 60 C for 18 h. The reaction liquid was cooled to room temperature, concentrated to remove the solvent. The crude product was dissolved in water (20 mL), and then extracted with ethyl acetate (20 mL x 4). The organic phase was washed with aqueous saturated NaC1 solution, dried over anhydrous sodium sulfate, and concentrated by filtration. The obtained crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=1:3), and concentrated to obtain 1 f (135.0 mg, yield 13.8%).
[0255] LCMS m/7 = 576.2 [M+1] .
Step 6: (S)-2-((4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-yppiperidin-1-yOmethyl)-1-1() (oxetan-2-ylmethyl)-1H-thieno [2,3-dlimidazole-5-carboxylic acid (Compound 1) om NC N N

OH
Compound 1 [0256] At room temperature, if (137.0 mg, 0.24 inmol) was dissolved in (6 mL) and water (3 InL) solution, then 1 g (CAS: 5807-14-7) (166.0 mg, L20 mmol) was added, and the reaction was continued for 24 h. 1N hydrochloric acid was used to adjust pH to 6, then dichloromethaneanethanol = 10:1 (20 tiaL x 3) was used for extraction. The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated.
The obtained crude product was subjected to preparative liquid chromatography to obtain (S)-2-((4-(6-((4-cyano-2-fluorobenzypoxy)pyridin-2-yepiperidin-l-yemethyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (compound 1) trifluoroacetic acid salt (60 mg, yield 44.9%). Compound 1 was obtained by preparative thin layer chromatography (dichloromethanehnethanol (v/v) = 10:1).
[0257] Conditions for preparative liquid chromatography:
[0258] Instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0259] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 in filter membrane to prepare into a sample liquid.

1102601 Mobile phase system: acetonitrile/water (containing 1%
TFA). Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 mL/min, elution time 18 min.
[0261] Compound 1:
[0262] LCMS m/z = 562.2 [M+1] .
[0263] 1H NMR (400 MHz, CD30D) 6 7.79 (s, 1H), 7.70-7.61 (m, 2H), 7.60-7.53 (m, 2H), 6.90 (d, 111), 6.75 (d, 114), 5.53 (s, 211), 5.26-5.17 (m, 114), 4.74-4.65 (m, 214), 4.62-4.55 (ni, 1H), 4.52-4.40 (ni, 3H), 3.67-3.55 (ni, 2H), 3.15-3.03 (ni, 2H), 2.98-2.87 (m, 1H), 2.85 ¨ 2.75 (m, 1H), 2.56-2.45 (m, 1H), 2.11 ¨ 1.99 (m, 411).
Example 2 (S)-2-((64(4-cyano-2-fluorobenzypoxy)-3',6'-dihydro-[2,4'-bipyridin1-1.(21-1)-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 2) OTh NC

OH
Compound 2 NH HCI
gil 11/11: ONJ
OMe 0 F
0 2a-1 2c-1 H2Nz 0- Step 1 N S 0- Step 2 Br N S 0-.. Step 3 1d 2a 2b N N
NC
11,õ,0¨N,XnN-t-11 0 j 1 g NCNTN
=Sy I S) Step 4 2c OH
Compound 2 Step 1: methyl (S)-2-methy1-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylate (2a) 2a [0264] At room temperature, ld (1.2 g, 5.1 mmol) was added to glacial acetic acid (50 mL), 2a-1 (926.0 mg, 7.7 mmol) was added to the reaction liquid, and the temperature was raised to 70 C for 1 hour. The glacial acetic acid was distilled off under reduced pressure, and the obtained crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=1:4), and concentrated to obtain 2a (680.0 mg, yield 50.1%).
[0265] LCMS m/z = 267.1 [M+1]+.
[0266] 'H NMR (400 MHz, CDC13) 6 7.68 (s, 111), 5.19 ¨ 5.11 (m, 114), 4.66 ¨ 4.58 (in, 1H), 4.36¨ 4.29 (in, 1H), 4.26 (d, 2H), 3.89 (s, 3H), 2.79 ¨ 2.68 (m, 1H), 2.63 (s, 3H), 2.44 ¨ 2.34 (m, 1H).
Step 2: methyl (S)-2-(bromomethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3 -d]imi dazol e-5 -carboxylate (2b) \) ________________________________________________ =/
Br 0-2b [0267] At room temperature, 2a (680.0 mg, 2.6 mmol) was dissolved in 1,2-dichloroethane (50 'EL) solution, then AIBN (84.0 mg, 0.5 inmol) was added, and NB S(1.1 g, 6.4 mmol) was added in portions in the process of slowly raising the temperature to 50 C. After the addition, the reaction liquid was heated to 70 C and continued to react for 8 h. The reaction liquid was cooled to room temperature, saturated sodium thiosulfate solution (30 mL) was added to quench the reaction and extracted with dichloromethane (30 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v)=1:1), and concentrated to obtain 2b (440.0 mg, yield 51.5%).
[0268] LCMS in/z = 344.9 [M+1]+.
Step 3: methyl (S)-2-((64(4-cyano-2-fluorobenzypoxy)-3',6'-dihydro-[2,4'-bipyridin1-1'(2'H)-yetnethyl)-1-(oxetan-2-ylinethyl)-1H-thieno 112,3 -d] imidazole-5-c arboxylate (2c) NC
N

2c [0269] At room temperature, 2c-1 (69.0 mg, 0.2 mmol) was dissolved in acetonitrile (5 mL) solution, then potassium carbonate (83.0 mg, 0.6 mmol) and 2b (69.0 mgõ
0.2 mmol) were added, and the temperature was raised to 40 C for 4 h. The reaction liquid was filtered to remove the inorganic base, the filtrate was collected and concentrated to obtain a crude product, and the obtained crude product was separated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) = 20:1), and concentrated to obtain 2c (75.0 mg, yield 66.0%).
[0270] LCMS in/z = 574.2 [M+1] .
Step 4: (S)-2((64(4-cyano-2-fluorobenzypoxy)-3',6'-dihydro- [2,4'-bipyridin] - (2'H)-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylic acid (Compound 2) NC
N

OH
Compound 2 [0271] At room temperature, 2c (75.0 mg, 0.13 mmol) was dissolved in acetonitrile (5 in-L) and water (1 naL), then 1 g (CAS: 5807-14-7) (91.0 mg, 0.66 iirmol) was added, and the reaction was continued for 24 h. 1N hydrochloric acid was used to adjust pH to 6, then dichloromethane:methanol = 10:1 (20 inL x 3) was used for extraction. The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product. The obtained crude product was subjected to prep-HPLC to obtain (S)-2-((4-(6((4-cyano -2-fluorobenzypoxy)pyridin-2-yppiperidin- 1 -yemethyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (compound 2) trifluoroacetic acid salt and then preparative thin-layer chromatography (dichlorotnethane:
methanol (v/v)=10:1) to obtain the compound 2 (16.0 mg, yield 21.9%).

[0272] Preparation conditions:
[0273] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0274] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 !..tm filter membrane to prepare into a sample liquid.
[0275] Mobile phase system: acetonitrile/water (containing 1%
TFA). Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 mUmin, elution time 18 min.
[0276] Compound 2:
[0277] LCMS m/z = 560.2 1M+11+.
[0278] 111 NMR (400 MHz, CD30D) 6 7.73 (s, 111), 7.69 ¨ 7.60 (m, 211), 7.59 ¨ 7.51 (m, 2H), 7.07 (d, 1H), 6.73 (d, 1H), 6.70 ¨ 6.65 (m, 1H), 5.53 (s, 2H), 5.22 ¨
5.14 (m, 1H), 4.72 ¨ 4.53 (m, 311), 4.45 ¨4.37 (m, 114), 4.10¨ 3.99 (m, 211), 3.37-3.33 (m, 211), 2.95 ¨
2.87 (m, 2H), 2.77 ¨ 2.69 (m, 1H), 2.66¨ 2.57 (m, 2H),2.49 ¨ 2.42 (m, 1H).
Example 3 2-(((S)-4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-methylpiperazin-l-ypmethyl)-1-(((S)-oxetan-2-y1)methyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 3) NC
0 )V r\l) N

OH
Compound 3 NBoc NfO
NC .F HN.õ.) NC - F NC FBV N S 0¨
.0 ,N_CI 3b 0 m t(j.NBoc r - NH
N,õ,) 2b Step 1 Step 2 UN
Step 3 3a 3c 3d rp7 CN:YN 0 NC 1 gH
, NC -F N
k,1õ0õ N
s 0 Step 4 0, OH
3f Compound 3 Step 1: tert-butyl (S)-4-(6-((4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-2-methylpiperazine-l-carboxylate (3c) NC
NBoc 3c [0279] 3a (524 mg, 2.0 minol), 3b (400 mg, 2.0 minol), Pd2(dba)3 (192 mg, 0.2 mmol), RuPhos (2-bicyclohexylphosphine-2',6'-diisopropoxybiphenyl) (184 mg, 0.4 mmol) and cesium carbonate (L3 g, 4M mmol) were added to 1,4-dioxane (20 mL).
The reaction flask was placed at 90 C and stirred for 6 hours after nitrogen replacement for protection. Then the reaction liquid was cooled to room temperature and quenched by pouring into aqueous saturated ammonium chloride solution (100 mL), then extracted with ethyl acetate (30 naL x 3), and the organic phase was dried over anhydrous sodium sulfate, filtered and spin-dried. The crude product was separated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) = 5:1), and concentrated to obtain 3c (700 mg, yield 82%).
[0280] LCMS m/z = 427.3[M+ 1 r.
Step 2: (S)-3-fluoro-4-(46-(3-methylpiperazin-1-yl)pyridin-2-yl)oxy)methyl) benzonitrile (3d) NC
r'NH

3d [0281] 3c (213 mg, 0.5 mmol) was added into a mixed solvent of dichloromethane (6 inL) and trifluoroacetic acid (1 inL), and stirred at 15 C for 1 hour. The reaction liquid was directly concentrated under reduced pressure to obtain 3d trifluoroacetic acid salt (230 mg, crude product), which was directly used in the next reaction without further purification.
[0282] LCMS tn/z = 327A [1\4+1] +.
Step 3: methyl 2-(((S)-4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-tnethylpiperazin-1-ypmethyl)-1-4(S)-oxetan-2-ypinethyl)-1H-thieno [2,3-d]imidazole-5-carboxylate (3t) NC
N

3f [0283] 3d (40 mg, 0.11 minol) and 2b (3g mg, 0.11 mmol) and potassium carbonate (47 mg, 0.33 trump were added into acetonitrile (3 mL), and stirred at 12 C
for 7 hours.
The reaction system was quenched by adding into an aqueous saturated ammonium chloride solution (20 mL), then extracted with ethyl acetate (30 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered and the filtrate was spin-dried. Separation and purification by silica gel column chromatography (dichloromethane/methanol (v/v) = 20:1) was performed to give 3f (40 mg, yield 60%).
[0284] LCMS m/z = 591.2 [M+1r.
Step 4: 2-(((S)-4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-2-methylpiperazin-l-yOmethyl)-1-(((S)-oxetan-2-yemethyl)-1H-thieno [2,3 -d] imidazole-5-c arb oxylic acid (Compound 3) NC

OH
Compound 3 [0285] 3f (40 mg, 0.067 inmol) and 1 g (47 mg, 0.033 mmol) were added to a mixed solution of acetonitrile (3 mL) and water (0.6 mL), and stirred at 12 C for 10 hours. The reaction liquid was then adjusted to pH = 6 with 1N hydrochloric acid, then extracted with ethyl acetate (10 mL x 3). The organic phase was combined and dried over anhydrous sodium sulfate, filtered and spin-dried, and prep-HPLC was performed to give 2-((S)-4-(6-((4-cy ano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-methylpiperazin- 1-yl)methyl)-1-((S)-oxetan-2-yenriethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (compound 3) trifluoroacetate salt. After separation by silica gel column chromatography (dichloromethane/methanol (v/v) = 20:1), compound 3 (10 mg, yield 25.6%) was obtained.
[0286] Preparation conditions:

[0287] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0288] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 tm filter membrane to prepare into a sample liquid.
[0289] Mobile phase system: acetonitrile/water (containing 1%
TFA). Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 mL/min, elution time 18 min.
[0290] LCMS m/z = 577.2 [M+1] .
[0291] 111 NMR (400 MHz, CD30D) 6 7.80 (s, 111), 7.61 (t, 111), 7.53 (t, 211), 7.44 (t, 1H), 6.27 (d, 1H), 6.13 (d, 1H), 5.43 (s, 2H), 5.27 ¨ 5.21 (m, 1H), 4.75 ¨
4.68 (m, 1H), 4.65 ¨ 4.57 (m, 211), 4.43 ¨ 4.32 (m, 211), 3.85 ¨ 3.79 (m, 111), 3.76¨ 3.70 (m, 111), 3.61 ¨
3.56 (m, 1H), 3.12¨ 3.03 (m, 1H), 2.93 ¨2.85 (m, 1H), 2.80¨ 2.71 (m, 2H), 2.63 ¨ 2.57 (m, 1H), 2.51 ¨ 2.43 (m, 1H), 2.40 ¨ 2.32 (m, 1H), 1.18 (d, 311).
Example 4
24(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d][1,3]dioxo1-4-yppiperidin-1-y1)methyl)-1-(((S)-oxetan-2-y1)methyl)-1H-thieno[2,3-dlimidazole-5-carboxylic acid (Compound 4) ci s OH
Compound 4 Br BOO
Bac OH
E C)13 CNBoc 4b F
^
Cr Step 3 F Step 0 RP
Step 2 111 4a 4c 4e CI 4164' 4f =pTSA N
Step 4 Br N S 0¨ 1 0 2b F 9 ,C rjj'I;_rsii?"1 Cirrf1V
õO>S0 Step 5 \S% Step 6 t CI
4h Compound 4 Step 1: 4-Bromo-2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d][1,3]dioxole (4c) Br CI
4c [0292] Substrates 4a (4L0 g, 0.2 mol) and 4b (45.0 g, 0.237 mol), and p-toluenesulfonic acid monohydrate (900 mg, 4.75 mmol) were added to toluene (200 mL), subjected to nitrogen replacement for protection, and placed at 140 C for reflux and water separation for 60 hours. The reaction liquid was cooled to room temperature, spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 4c (18.0 g, yield 22%).
Step 2: tert-butyl 4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo [d] [1,3]
dioxo1-4-y1)-3,6-dihydropyridine-1(2H)-carboxylate (4e) Boc CI
4e [0293] 4b (2.0 g, 5.82 mime and 4c (1.62 g, 5.24 mime, Pd(dpp0C12 (470 mg, 0.58 minol) and sodium carbonate (3.08 g, 29.1 ininol) were added to a mixed solvent of 1,4-dioxane (30 mL) and water (S mL), subjected to nitrogen replacement for protection, stirred at 90 C for 20 hours, cooled to room temperature, and extracted with ethyl acetate (80 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered, and spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 4e (1.6 g, yield 62%).
[0294] LCMS in/z = 390.0[M-55]t Step 3: tert-butyl 44244 -chloro-2-fluoropheny1)-2-methylbenzo [d] [1,3] dioxo1-4-yl)piperidine-l-carboxylate (40 Boc CI
4f [0295] 4e (L6 g, 3.59 mmol) and Raney nickel (160 mg) were added to a mixed solvent of methanol (15 mL) and tetrahydrofuran (10 mL), subjected to hydrogen replacement for protection, and stirred at room temperature for 8 hours. The reaction liquid was filtered and spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 4f (1.4 g, yield 87%).
[0296] LCMS in/z = 392.2[M-55] .
Step 4: 4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo [d] [1,3] dioxo1-4-yl)piperidine P-toluenesulfonate (4g) .pTSA

CI
4g [0297] 4f (0.8 g, L79 mmol) and p-toluenesulfonic acid monohydrate (410 mg, 2J5 mmol) were added to ethyl acetate (20 mL), subjected to nitrogen replacement for protection, and stirred at 45 C for 12 hours. The reaction liquid was cooled to room temperature, filtered, and washed with ethyl acetate to obtain the p-toluenesulfonate 4g (620 mg, yield 99%) as a solid.
[0298] LCMS m/z = 348.0 [M+1]+.
Step 5:
methyl 24(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo [d] [1, 3] dioxo1-4-yl)piperidin-l-y1) methyl)-1-(((S)-oxetan-2-yl)tnethyl)-1H-thieno [2,3 -d] imidazole-5-carboxylate (41) CI
0 1\17 4i [0299] 4g (150 mg, 0.3 mmol) and 2b (103 mg, 0.3 mmol), and potassium carbonate (210 mg, 1.5 inmol) were added into acetonitrile (5 mL) and stirred at room temperature for 5 hours. The reaction liquid was directly mixed with silica gel, and separated and purified by silica gel column chromatography to obtain 4i (200 mg, yield 76%).
[0300] LCMS m/z = 612.3 [M+1]+.
Step 6: 2-44-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo [d] [1,3] dioxo1-4-yepiperidin-1-yOmethyl)-1-4(S)-oxetan-2-yl)methyl)-1H-thieno [2,3 -d] imidazole-5-c arb oxylic acid (Compound 4) ci om OH
Compound 4 [0301] 4i (200 mg, 0.33 intnol) and lithium hydroxide monohydrate (140 mg, 3.3 inmol) were added to a mixed solvent of acetonitrile (4 inL) and water (1 mL), and stirred at 50 C for 8 hours. The reaction liquid was cooled to room temperature, adjusted with 1N
hydrochloric acid to pH=6, and extracted with ethyl acetate. The organic phase was dried with anhydrous sodium sulfate, filtered and spin-dried. The residue was separated and purified by silica gel column chromatography to obtain the product (24(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d] [1,3] dioxo1-4-yepiperidine-1 -yemethyl)-14(S)-oxetan-2-yOmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (compound 4) (50 ing, yield
25%).
[0302] LCMS in/z = 598.1 [M+1]+.
[0303] 11-1 NMR (400 MHz, DMS0-(16) 8 7.73 (s, 1H),7.63 ¨ 7.51 (in, 2H), 7.34 (d, 1H), 6.83¨ 630 (in, 3H), 5.15 ¨ 5.04 (in, 1H), 4.67 ¨ 4.58 (in, 1H), 4.55 ¨
4.43 (in, 2H), 4.41 ¨ 4.33(m, 1H), 3.80 (d, 1H), 3.69 (d, 1H), 2.97 (d, 1H), 2.87 (d, 1H), 2.73¨ 2.59 (m, 214), 2.46 ¨ 2.37 (m, 114), 2.23 ¨2.09 (m, 214), 2.02 (s, 311), 1.83¨ 1.61 (m, 411).
Example 5 24(4-(2-(4-cyano-2-fluoropheny1)-2-methylbenzo[d] [1,3] dioxo1-4-yppiperidin-1-yl)methyl)-1-(((S)-oxetan-2-yl)methyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 5) NC
0 N z OH
Compound 5 (:)D
Boc Boc =pTSA I \
Br N S 0-jo(0 F 0 F 2b b Step 1 NC 5g NC 5 Step 2 Step 3 CI-- 4f NC la NC
0 F õ5.1 011µ1' F a ), N--cs1 0 Step 4 \s 0 5j Compound 5 Step 1: tert-butyl 4-(2-(4-cyano-2-fluoropheny1)-2-methylbenzordi 1-1,31dioxo1-4-yl)piperidine-l-carboxylate (5g) Boc NC
5g [0304] 4f (600 mg, L34 mmol) and Pd2(dba)3 (65 mg, 0.07 mmol), dppf (84 mg, 0.15 mmol), Zn(CN)2 (264 mg, 2.25 inmol), and zinc powder (24 mg, 0.37 minol) were added to DMA (15 inL), subjected to nitrogen replacement for protection, and stirred at 120 C
for 20 hours. The reaction liquid was cooled to room temperature, filtered and spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain g (150 mg, yield 23%).
[0305] LCMS m/z = 383.0[M-55]t Step 2: 4-(2-(4-cyano-2-fluoropheny1)-2-methylbenzo[d][1,3]dioxol-4-ylipiperidine P-5 toluenesulfonate (5h) =pTSA
xl NC
5h [0306] 5g (200 mg, 0.46 mmol) and p-toluenesulfonic acid monohydrate (130 mg, 0.69 mmol) were added to ethyl acetate (8 tnL), subjected to nitrogen replacement for protection, and stirred at 45 C for 12 hours. The reaction liquid was cooled to room temperature, filtered, and washed with ethyl acetate to obtain the p-toluenesulfonate 5h (250 mg, yield 90%) as a solid.
[0307] LCMS in/z = 339.2 [M+1] .
Step 3: methyl 2-((4-(2-(4-cy ano-2-fluoropheny1)-2-methylbenzo [d] [1,3] dioxo1-4-yl)piperidin-l-y1) methyl)-1-(((S)-oxetan-2-yl)tnethyl)-1H-thieno [2,3 -d] imidazole-5-carboxylate (5j) NC

5j [0308] 5h (120 mg, 0.2 mmol) and 2b (821ng, 0.22mm01), and potassium carbonate (160 mg, 1.1 mmol) were added into acetonitrile (5 mL) and stirred at room temperature for 5 hours. The reaction liquid was directly mixed with silica gel, and separated and purified by silica gel column chromatography to obtain 5j (100 mg, yield 83%).
[0309] LCMS tn/z = 603.2 [M+1] .

Step 4: 24(4-(2-(4-cyano-2-fluoropheny1)-2-methylbenzo[d] [1,3] di oxo1-4-yl)piperi din-1-yOmethyl)-1-(((S)-oxetan-2-yOmethyl)-1H-thi en o [2,3-dlimidazole-5-carboxylic acid (Compound 5) NC
(c-A

S
OH
Compound 5 [0310] 5j (100ing, 0Ø17 inmol) and lithium hydroxide monohydrate (115 ing, 2.73 mmol) were added to a mixed solvent of acetonitrile (4 inL) and water (1 inL), and stirred at 50 C for 8 hours. The reaction liquid was cooled to room temperature, adjusted with 1N
hydrochloric acid to pH=6, and extracted with ethyl acetate. The organic phase was dried with anhydrous sodium sulfate, filtered and spin-dried. The residue was separated and purified by silica gel column chromatography to obtain (24(4-(2-(4-cyano-2-fluoropheny1)-2-methylbenzo[d] [1,3]dioxo1-4-yepiperidine-1-yemethyl)-1-((S)-oxoalkane-2-y1)methyl)-1H-thieno 112,3 -d] imidazole-5-c arboxylic acid (compound 5) (50 ing, 48% yield).
[0311] LCMS m/z = 589.2 [M+1] .
[0312] 1H NMR (400 MHz. DMSO-c/o) 6 12.94 (hr, 1H), 7.97 (d, 1H), 7.81 (s, 1H), 7.77 ¨ 7.69 (in, 2H), 6.85¨ 6.70 (in, 3H), 5.14 ¨ 5.05 (in, 1H), 4.69 ¨ 4.61 (in, 1H), 4.57 ¨
4.44 (in, 2H), 4.40 ¨ 4.32(in, 1H), 3.82 (d, 1H), 3.72 (d, 1H), 2.98 (d, 1H), 2.88 (d, 1H), 2.73¨ 2.60 (in, 2H), 2.45¨ 2.36 (in. 1H), 2.24 ¨2.10 (in, 2H), 2.05 (s, 3H), 1.79 ¨ 1.63 (in, 4H).
NCO NC

OH OH
Compound 5-a Compound 5-b [0313] 2g compound 5 was prepared by SFC and dried to obtain compound 5-1 (600 ingõ yield 30%) as a white solid with a retention time of 6.441 minutes.
compound 5-2 (500 mg, yield 25%) as a white solid with a retention time of 7.104 minutes.

[0314] Preparation conditions:
[0315] instrument and preparative column: Waters 150 MGM chiral liquid phase chromatographic instrument was used, and the preparative column model was DAICEL
CHIRALPAK AD, I.D., 5 m, inner diameter x length = 250 x 30 mm.
[0316] Preparation method: The crude product was dissolved in ethanol and filtered with a 0.45 !..im filter membrane to prepare into a sample liquid.
[0317] Mobile phase system: carbon dioxide/ethanol (0.1% NH3 H20). Gradient elution: the ethanol content was 25%, and the elution time was 11 minutes.
[0318] Compound 5-1:
[0319] LCMS m/z = 589.2 [M+11+.
[0320] 1H NMR (400 MHz, DMSO-d6) 6 8.01-7.93 (m, 111), 7.79(s, 1H),7.77-7.70 (m, 2H), 6.84-6.69 (m, 3H), 5.15-5.06 (m, 111), 4.69-4.60 (m, 111), 4.57-4.44 (m, 2H), 4.42-4.34(m, 1H), 3.85-3.78 (m, 1H), 3.76-3.68 (m, 111), 3.02-2.93 (m, 111), 2.92-2.84 (m, 111), 2.74-2.58 (m, 2H), 2.46-2.36 (m, 111), 2.25-2.10(m, 2H), 2.05 (s, 311), 1.81-1.64 (m, 411).
[0321] The structure of compound 5-1 is one of the above formulas 5-a and 5-b;
[0322] Compound 5-2:
[0323] LCMS m/z = 589.2 [M+1]+.
[0324] 1H NMR (400 MHz, DMSO-d6) 6 8.00-7.93 (m, 1H), 7.77-7.67 (m, 3H), 6.84-6.72 (m, 3H), 5.13-5.04 (m, 1H), 4.67-4.58 (m, 111), 4.56-4.44 (m, 2H), 4.41-4.33(m, 1H), 3.83-3.77 (m, 1H), 3.74-3.67 (m, 1H), 3.01-2.93 (m, 1H), 2.91-2.83 (m, 1H), 2.72-2.59 (m, 211), 2.46-2.35 (m, 111), 2.23-2.10 (m, 211), 2.05 (s, 311), 1.79-1.61 (m, 411).
[0325] The structure of compound 5-2 is one of the above formulas 5-a and 5-b; and compound 5-2 and compound 5-1 were isomers for each other, that is, when the structure of compound 5-1 was the structure of formula 5-a, the structure of compound 5-2 was the structure of formula 5-b; when the structure of compound 5-1 was the structure of formula 5-b, the structure of compound 5-2 was the structure of formula 5-a.
Example 6 (S)-2-(4-(64(4-cyano-2-fluorobenzypoxy)pyridi n-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-111-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 6) Fr.------1 NC N
N

S
Compound 6 r)o_3 HNjp Rip 0 B F H2 N - ,L4 - '0- Br 0- \
--t.-I 9 ;.N ___________________________________________________________________ -1 .. 40 0 r 1--Step 1 NH=FICI Step 2 step 4 N
u Step 3 F(. s IN S U-rsirsA
6a 6b 6c 6f 6e 0,CN 0, N N Fi ir,o,, F 6NC , F _ 14--,/
lg H NC
Step 5 n_o N, a- ----Kõ __ Step 6 1---3 o OH 6h Compound 6 Step 1: ethyl 2-(4-bromo-2-fluorophenypacetimidate hydrochloride (6b) Br F

N H +I CI
6b [0326] 6a (5.0 g, 26.04 minol) was added to anhydrous ethanol (3.0 mL), placed at 0 C and fully stirred, and at the same time, dry hydrogen chloride gas was introduced into the reaction liquid until it was saturated, and the reaction liquid was continuously stirred at this temperature for 10 hours. Diethyl ether (40 inL) was then added to the reaction liquid, filtered to obtain a solid that was further washed with diethyl ether (100 mL
x 2 ), and dried to obtain 6b (5.0 g, yield 72%).
[0327] II-1 NMR (400 MHz, DMSO-do) 6 12.04 (s, 2H), 7.61 (d, 1H), 7.51 ¨ 7A3 (in, 2H), 446 (q, 2H), 4A4 (s, 2H), 1.26 (t, 3H).
[0328] 19F NMR (400 MHz, DMSO-do) 6 111.30.
Step 2: 4-broino-2-fluoro-1-(2,2,2-triethoxyethyl)benzene (6c) Br F

0, 6c [0329] 6b (5.0 g, 19.30 ininol) was added into anhydrous ethanol (40 itiL), and stirred at room temperature (15 C) for 48 hours. Then the reaction liquid was directly filtered to obtain a filtrate, and the filtrate was spin-dried to make a slurry with a mixed solvent of petroleum ether and ethyl acetate (100 inL) (petroleum ether/ethyl acetate (v/v) = 10:1) and filtered, and the filtrate was spin-dried to obtain 6c (4M g, yield 66%).
[0330] 11-1 NMR (400 MHz, CDC13) 7.45 (t, 1H), 7.23 ¨ 7.14 (in, 2H), 3.50 (q, 6H), 3.07 (s, 2H), 1.16 (t, 9H).
Step 3: methyl (S)-2-(4-bromo-2-fluorobenzy1)- 1-(oxetan-2-ylinethyl)- 1H-thieno [2,3-d]imidazole-5-carboxylate (6e) Br ;):_)]

N¨S 0-6e [0331] 6d (500 mg, 2.06 mmol) and 6c (1.0 g, 3.19 mmol) were added to glacial acetic acid (20 inL), and stirred at 60 C for 10 hours. The reaction system was cooled to room temperature, then was slowly added to an aqueous saturated sodium bicarbonate solution (200 mL) to quench the reaction, and then extracted with ethyl acetate (50 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered.
The filtrate was spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 6e (430 mg, yield 47%).
[0332] LCMS m/z = 439.0 [M+1]+.
Step 4: methyl (S)-2-(2-fluoro-4-(4,4,5 ,5-tetramethy1-1,3 ,2-dioxaborolan-2-yl)benzy1)-1-(oxetan-2-yltnethyl)-1H-thieno [2,3 -d] imidazole-5-c arboxylate (61) oo 6f [0333] 6e (400 mg, 0.98 mmol) and pinacol biborate (262 mg, 1.0 mmol), Pd(dppf)C12.DCM (81 mg, OA mmol), and potassium acetate (196 mg, 2.0 mmol) were added to 1,4-dioxane (20 mL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (100 mL) was added and extracted with ethyl acetate (50 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered, the filtrate was spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 6f (340 mg, yield 71%).
[0334] LCMS in/z = 487.2 [M+1]+.
Step 5: methyl (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylinethyl)-1H-thieno 112,3 -d] imidazole-5-c arboxylate (6h) NC

6h [0335] 6f (340 mg, 030 mmol) and 6g (220 mg, 0.84 mmol), Pd(dppf)C12.DCM
(83 mg, OA minol) and potassium carbonate (290 mg, 2A ininol) were added to a mixed solution of 1,4-dioxane (10 mL) and water (2 mL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (50 inL) was added, and extracted with ethyl acetate (30 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 6h (200 mg, yield 48%).

[0336] LCMS m/z = 587.1 [M+1]+.
Step 6: (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5 -carboxylic acid (Compound 6) ,70 NC
, OH
Compound 6 [0337] 6h (100 mg, 0.17 mmol) and 1 g (120 mg, 0.85 inmol) were added to a mixed solution of acetonitrile (5 mL) and water (1 inL), and stirred at room temperature (12 C) for 10 hours. Then the reaction liquid was adjusted to pH=6 with 1M
hydrochloric acid, continuously stirred until a white solid precipitated, and filtered and the solid was washed with water. The solid was dissolved in dichloromethane, dried over anhydrous sodium sulfate and filtered. The organic phase was spin-dried. The residue was separated by preparative HPLC to give (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-fluorobenzy1)-1-(oxoalkane-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-c arboxylic acid (compound 6) tritluoroacetate salt, which was separated and purified by silica gel column chromatography to obtain compound 6 (40 mg, yield 41%).
[0338] LCMS in/z = 573.1 [M+1] .
[0339] 114 NIVIR (400 MHz, CD30D) 6 12.90 (br, 1H), 7.91 (d, 1H), 7.88 ¨ 7.80 (in, 4H), 7.79 ¨ 7.68 (in, 2H) 7.64 (d, 1H), 7.39 (t, 1H), 6.92 (d, 1H), 5.61 (s, 2H), 5.06 ¨ 4.98 (in, 1H), 4.60(dd, 1H), 4.54 ¨ 4.44 (in, 2H), 4.43 ¨ 4.28 (in, 3H), 2.73 ¨
2.61 (in, 1H), 2.40¨ 2.27 (in, 1H).
Example 7:
(S)-2-(4-(64(4-cy ano-2-fluorobenzypoxy)pyridin-2-yl)benzyl)- 1 -(oxetan-2-ylmethyl)- 1H-thieno[2,3-d[imidazole-5-carboxylic acid (Compound 7) o NC
OH
Compound 7 `ci.;.A
OEt r- rCN _________________________ H2N S 0¨ ld Br N11-1.HCI 13r' 1 0 ___ Step 1 Br' Step 2 7a 7b 7c 0, Step 3 \L) 7d 6 NCyp-6]) T F Step 4 NC Step -N
3a __________________________________________________________ NcçON N-tro OH
7e Compound 7 Step 1: ethyl 2-(4-bromophenyl)acetimidate hydrochloride (7b) OEt NH.HCI
Br 7b [0340] Under the condition of ice bath, the substrate 7a (19.6 g, 100.0 mmol) was dissolved in a solution of 33% hydrochloric acid in ethanol (45 mL). After the addition, the temperature was naturally raised to room temperature for overnight reaction.
At room temperature, anhydrous diethyl ether (100 mL) was added to the reaction system, stirred for 5 minutes and filtered, and the filter cake was washed with diethyl ether (20 mL x 2), collected and dried to obtain the product 7b of interest (22.0 g , yield 79.4%) as a white solid.
[0341] LCMS m/z = 242.1 [M+11 .
Step 2: methyl (S)-2-(4-bromobenzy1)-1-(ox etan-2-ylmethyl)-1H-thi eno [2,3-dlimidazole-5-carboxylate (7c) on N
Br 0 7c [0342] At room temperature, the substrate 7b (1.4 g, 5.0 mmol) was dissolved in glacial acetic acid (50 mL), and ld (1.2 g, 5.0 mmol) was added. After the addition, the reaction system was heated to 45 C for overnight reaction under nitrogen protection. The reaction solvent was distilled off under reduced pressure to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 7c (740.0 mg, yield 35.2%) as a white solid.
[0343] LCMS m/z = 421.0 [M+1]+.

Step 3: methyl (S)-1-(oxetan-2-ylmethyl)-2-(4-(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-y1) benzy1)-1H-thieno[2,3-dlimidazole-5-carboxylate (7d) cr) /

7d N
[0344] At room temperature, 7c (740.0 mg. 1.8 mmol) was dissolved in 1.4-dioxane (30 inL), and then pinacol biborate (820.0 mg, 2.7 mmol), Pd(dppf)C12 (147.0 mg, 0.2 mmol) and potassium acetate (519.0 mg. 5.3 mmol) were added successively.
After the addition, the reaction system was heated to 100 C for overnight reaction under nitrogen protection. The reaction liquid was cooled to room temperature and filtered.
The filter cake was washed with dichloromethane (10 mLx2). The filtrate was collected and concentrated to obtain a crude product, which was purified by silica gel column chromatography to obtain 7d (680.0 mg, yield 82.4%) as a yellow solid.
[0345] LCMS m/z = 469.2 [M+1]+.
Step 4: methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yebenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5 -c arboxy late (7e) o NC
I17,_N,, = N
7e [0346] At room temperature, 3a (130.0 mg, 0.5 mmol) was dissolved in a mixed solvent of 1,4-dioxane (10 mL) and water (1 mL), and then 7d (193.0 mg, 0.4 mmol) and Pd(dppf)C12 (34.0 mg, 0.04 mmol) and cesium carbonate (261.0 mg, 0.8 niftiol) were added successively. After the addition, the reaction system was heated to 100 C for overnight reaction under nitrogen protection. The reaction liquid was cooled to room temperature and filtered. The filter cake was washed with dichloromethane (10 mL x 2).
The filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 7e (109.0 mg, yield 46.6% ) as a white solid.

1103471 LCMS m/z = 569.3 [M+1]+.
Step 5: (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-yObenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 7) NC
' \S¨ o OH
Compound 7 [0348] At room temperature, 7e (109.0 mg, 0.2 mmol) was dissolved in a mixed solvent of acetonitrile (5 mL) and water (1 mL), and then 1 g (CAS: 5807-14-7) (80.0 mg, 0.6 mmol) was added. After the addition, the reaction was carried out at room temperature for 24 hours. Acetonitrile was distilled off under reduced pressure. The crude product was dissolved in water (5 mL), adjusted to pH=6 with 1N hydrochloric acid. The aqueous phase was extracted with ethyl acetate (15 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain compound 7 (26.0 mg, yield 24.5%) as a white solid.
[0349] Compound 7:
[0350] LCMS m/z = 555.2 [M+1] .
[0351] 111 NMR (400 MHz, DMSO-d6) 6 7.98 (d, 2H), 7.93 ¨ 7.88 (m, 111), 7.82 (t, 1H), 7.78 ¨ 7.68 (m, 3H), 7.57 (d, 1H), 7.38 (d, 2H), 6.87 (d, 1H), 5.60 (s, 2H), 4.94 ¨ 4.86 (m, 1H), 4.54 ¨ 4.30 (m, 6H), 2.65 ¨2.56 (m, 111), 2.33 ¨ 2.24 (m, 111).
Example 8 (S)-2-(4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-2-fluoro-5-methylbenzy1)-(oxetan-2-ylmethyl)-1H-thieno[2,3-dlimidazole-5-carboxylic acid (Compound 8) r/1 NC , N
T
)- )0, N 0 T
OH
Compound 8 FO F F F F

_-OH _____________________________________ ib OMs ____ 6 'CN __ Br ' Br"'r NI-1.1-1C1 Step 1 Step 2 Br ' Step 3 Br Step 4 Br 8a 8 b 8c 8d 8e r---1 HN 0 NC,,,,, Q rrC7 --.
HN C' ri) ' F
2 0- F if-P-7 I d _\p-B 0 A--1\s-iro 3a Step 5 Br B--Yo Step 6 ))s0 (:) Step 7 , 8f 0 8g 0- r In F N N F
NC
NI) NC ,-...,., -T... H

-,1)L,0 0 N ep 8 St F OH
0, 8h Compound 8 Step 1: (4-bromo-2-fluoro-5-methylphenyl)methanol (8b) F
OH
Br 8b [0352] 8a (10.0 g, 43 mmol) and a 2M solution of borane in tetrahydrofuran (26 tnL, 51.6 minol) were added to tetrahydrofuran (40 inL) and stirred at room temperature for 30 hours. Then the reaction liquid was slowly added to 0.5 N hydrochloric acid (100 mL) to quench the reaction, and then extracted with ethyl acetate (80 mL x 3). The organic phase was dried over anhydrous sodium sulfate and filtered, and the liquid was concentrated to dryness. The residue was separated and purified by silica gel column chromatography to obtain 8b (9.4 g, yield 99%).
[0353] LCMS m/z = 201.0[M-17]t Step 2: 4-bromo-2-fluoro-5-methylbenzyl tnethanesulfonate (8c) F
0Ms Br 8c [0354] 8b (2.18 g, 10 minol) and triethylamine (2.0 g, 20 minol) were added to dry dichloromethane (20 mL) and stirred at 0 C for 10 minutes, and then methyl sulfonyl chloride (1.38 g, 12 mmol), was slowly added to the reaction system and the reaction was stirred at 0 C for further 30 minutes. Water (20 mL) was added to the reaction liquid, extracted with dichloromethane (50 mL x 3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was directly concentrated under reduced pressure to obtain 8c (3.0 g, yield 99%), which was directly used in the next reaction without further purification.
Step 3: 2-(4-bromo-2-fluoro-5-methylphenyl)acetonitrile (8d) CN
Br 8d [0355] 8c (3.0 g, 10 inmol) and sodium cyanide (440 mg, 9 inmol) were added to DIVIF (15 mL), and stirred at room temperature for 40 hours. Ethyl acetate (300 inL) was added to the reaction system, washed with water (50 mL x 3). The aqueous phase was added to an aqueous sodium hypochlorite solution (50 mL) and stirred for 5 hours. The organic phase was combined, dried over anhydrous sodium sulfate and filtered.
The filtrate was concentrated to dryness and purified by silica gel column chromatography to give 8d (L5 g, yield 66%).
Step 4: ethyl 2-(4-bromo-2-fluoro-5-methylphenyl)acetimidate hydrochloride (8e) Br NH=FICI
8e [0356] 8d (1.5g, 6.6 ininol) was added into ethanol (5 inL) and stirred at room temperature. Then, dry hydrogen chloride gas was introduced into the reaction liquid for 1 hour, and then continually stirred for 10 hours. Diethyl ether (10 inL) was added to the reaction liquid, stirred until a solid precipitated, and filtered. The filter cake was washed with diethyl ether and dried to obtain 8e (L5 g, yield 73%).
Step 5: methyl (S)-2-(4-bromo-2-fluoro-5-methylbenzy1)-1-(oxetan-2-ylmethyl)-thieno 112,3 -d] imidazole -5 -c arboxylate (8f) Br 0 8f [0357] 8e (300 mg, Idi.96 mmol) and Id (250 mg, LO mmol) were added to glacial acetic acid (10 mL), subjected to nitrogen replacement for protection and then stirred at 40 C for 10 hours. The reaction liquid was added to a aqueous saturated sodium bicarbonate solution to quench the reaction, and extracted with ethyl acetate (30 mL x 3).
The organic phase was dried over anhydrous sodium sulfate and filtered. The liquid was concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain 8f (100 mg, yield 25%).
[0358] LCMS m/z = 453.0[M+ lr.
Step 6: methyl (S)-2-(2-fluoro-5-methy1-4 -(4,4,5 ,5-tetramethy1-1,3,2-dio xaborolan-2-y1) benzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3 -d] imidazole-5-c arboxylat e (8g) 8g [0359] 8f (100 mg, 0.22 mmol), pinacol biborate (69 mg, 0.26 mmol), Pd(dppf)C12 (18 mg, 0.022 mmol) and potassium acetate (45 mg, 0.44 mmol) were added to 1,4-dioxane (8 mL), subjected to nitrogen replacement for protection and then stirred at 100 C
for 12 hours. Then the reaction liquid was cooled to room temperature, concentrated to dryness, and separated and purified by silica gel column chromatography to obtain 8g (70 mg, yield 63%).
[0360] LCMS tn/z = 501.2[M+ 1 r.
Step 7: methyl (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2-fluoro-methylbenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylate (8h) õ70 NC
I /

8h [0361] 8g (70 ing, 0.14 minol), 3a (50 mg, 0.2 ininol), Pd(dppt)C12dichloromethane complex (18 mg, 0.02inino1) and potassium carbonate (55 mg , 0.4 intnol) were added to a mixed solvent of 1,4-dioxane (5 mL) and water (1 mL), subjected to nitrogen replacement for protection and then stirred at 100 C for 6 hours. The reaction liquid was cooled to room temperature, quenched with an aqueous saturated ammonium chloride solution (20 inL), and extracted with ethyl acetate (20 mL x 3). The organic phase was dried over anhydrous sodium sulfate, filtered, spin-dried, and separated and purified by silica gel column chromatography to obtain 8h (60 mg, yield 71%).
[0362] LCMS in/z = 601.3 [M+1[+.
Step 8: (S)-2-(4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-2-fluoro-5-methylbenzy1)-1-(oxetan-2-ylinethyl)-1H-thieno [2, 3-d] imidazole-5-c arboxylic acid (Compound 8) ,70 NC
/

OH
Compound 8 [0363] 8h (60 mg, 0.1 mmol) and lg (CAS: 5807-14-7) (70 mg, 0.5 mmol) were added into a mixed solvent of acetonitrile (5 inL) and water (1.0 inL), and stirred at 50 C
for 5 hours. The reaction liquid was cooled to room temperature, adjusted to pH=6 with 1N
hydrochloric acid, then extracted with ethyl acetate (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered and the liquid was concentrated to dryness under reduced pressure. The residue was separated and purified by silica gel plate chromatography to obtain compound 8 (10 mg, yield 17%).
[0364] LCMS in/z = 587.3 [M+1[+.
[0365] 11-1 NMR (400 MHz, DMSO-d6) 6 12.89(s, 1H), 7.95 ¨ 7.80 (in, 3H), 7.77 ¨
7.66 (in, 2H), 7.26 ¨ 7.16 (in, 3H), 6.94(d, 1H), 6.51 (s, 2H), 5.06¨ 4.97 (in, 1H), 4.61 (in, 1H), 4.54 ¨ 4.45 (m, 2H), 4.39 ¨ 4.22 (m, 3H), 2.74 ¨ 2.65 (m, 1H), 2.38¨ 2.30 (m, 1H), 2.22(s, 3H).
Example 9:
(S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-6-oxopyridazin-1(61-1)-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thienor,3-dlimidazole-5-carboxylic acid (Compound 9) icr:
o NC _ -N TI-NI>r____ - 1 0. .1µ1 N N----/ \1 \
I S---COOH
F-. ,---.--Compound 9 NC
Br 6 INI__ B (:j 0 6--I1, 0 -- F N I
OH
' u, _ N
, NH 2b 'S -0O2Me e- 1\1-N, 9c F
CI
I N ' CIN N 7 \ F N,)LIN
9a Step 1 9b S CO2Me Step 2 1 9d S CO2Me Step 3 0 01- 0o I
NC. Step 4 NC N
0 1 j ¨?1,`----..
N,. ' - NN I
I S'cO2Me F I
F
9h Compound 9 Step 1: methyl (S)-2-((4-chloro-6-oxopyridazin-1(611)-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thienor,3-dlimidazole-5-carboxylate (9b) (c-A

Am N

S co2me 9b [0366] The raw materials 9a (120 mg, 0.87 mmol) and 2b (300 mg, 0.87 mmol) were added into a reaction flask and dissolved in acetonitrile (10inL). Anhydrous potassium carbonate (250 mg, 1.80 mmol) was added, reacted at 40 C for 4h and cooled to room temperature. The solid was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to obtain product 9b (247 mg, yield 71.8%) as a yellow solid.
[0367] LCMS m/z = 395.0 [M+Hr.

Step 2: methyl (S)-2-((4-(6-fluoropyridin-2-y1)-6-oxopyridazin-1(6H)-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d]imidazol e-5-carboxylate (9d) /
N N
S 002me 9d [0368] 9b (40 mg, 0.10 mmol), 9c (30 mg, 0.20 mmol), anhydrous potassium carbonate (40 mg, 0.20 mmol), and Pd(dppt)C12 (15 mg, 0.01 mmol) were added to a reaction flask successively, dissolved in a mixed solution of 1,4-dioxane (2 mL)/water (0.4 mL), subjected to nitrogen replacement, stirred at 90 C for 5 h, cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to obtain product 9d (22 mg, yield 48.3%) as a yellow solid.
[0369] LCMS in/z = 456.1 [M+Hr.
Step 3: methyl (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-6-oxopyridazin-1(6H)-ye methyl)-1-(oxetan-2-ylinethyl)-1H-thieno [2,3 -di imidazole-5-c arboxylate (9h) NC
CO2me 9h [0370] 9d (22 mg, 0.05 mmol) and 9f (15 mg, 0.09 mmol) were added into a reaction flask and dissolved in tetrahydrofuran solution (1 mL). Potassium tert-butoxide (10 mg, 0.07 mmol) was added under nitrogen, and stirred at room temperature for 1 h.
The reaction was quenched with aqueous ammonium chloride solution (1 mL) and extracted with ethyl acetate 2 mL x 3. The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was separated and purified by silica gel column chromatography to obtain 9h (20 mg, yield 71.1 %) as a yellow solid.
[0371] LCMS m/z = 587.1 1M+Hr.
Step 4: (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-6-oxopyridazin-1(611)-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 9) O----NC.,õ )N--\ N\
1 ' 1\1, N-J-C1 T s COON
F
Compound 9 [0372] At room temperature, 9h (20 mg, 0.03 mmol) was dissolved in a solution of acetonitrile (2 mL) and water (0.5 mL), then 1 g (CAS: 5807-14-7) (40 mg, 0.28 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with dichloromethane:methanol = 10:1 (2 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was separated and purified by silica gel plate chromatography to obtain compound 9 (5 mg, yield 25.7%).
[0373] LCMS m/z = 573.1 [M+H]t [0374] 111 NMR (400 MHz, DMSO-d6) 6 13.14¨ 12.78 (m, 111), 8.62 (d, 111), 7.98 ¨
7.68 (m, 6H), 7.57 (d, 1H), 7.08 (d, 1H), 5.68 ¨ 5.50 (m, 4H), 5.04 (d, 1H), 4.70 (dd, 1H), 4.58 (d, 111), 4.48 (dd, 111), 4.33 (dd, 1H), 2.74 ¨ 2.63 (m, 1H), 2.33 (dd, 111).
Example 10:
2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo [d] [1,3]dioxo1-4-y1)-2-fluorobenzy1)-1-(((S)-oxetan-2-yl)methyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 10) a ---- I F 0, /
/3, cl , IT¨Ii", --- N.--_.
k. ) -s - 'co2H
Compound 10 i:
HN---, 1d :D_J
I-12N-// )õ, Br - F
BrF__cN
_ I
--- NH HCI F
OEt Step 2 )\_"1 Step 3 Step 1 CO2Me 10a 10b Br 10c S
CL F
Br a ah, F
F F
"N -0 Step 5 0 IN. --- \ --\

--\
s CO2Me C)-13 "10d Step 4 6 CO2Me S cO2H
10e glip Compound 10 Step 1: ethyl 2-(4-bromo-2-fluorophenyl)acetimidate hydrochloride (10b) Br NH HCI
OEt 10b [0375] 10a (10.0 g, 52.1 mmol) was added into a reaction flask, and 33% hydrogen chloride-ethanol solution was introduced under ice bath, and then the temperature was gradually raised to room temperature for overnight reaction, and a large number of white solids were precipitated. Diethyl ether was added and stirred for 0.5h, the solid was filtered, washed with diethyl ether, and dried to obtain 10b (9.3 g, yield 60.6%) as a white solid.
Step 2: methyl (R)-2- (4-bromo-2-fluorobenzy1)- 1-(oxetan-2-ylmethyl)- 1H-thieno [2,3-d]imidazole-5-carboxylate (10c) Ot]
/ \
s CO2Me Br 10c [0376] 10b (500 mg, 2.06 inmol) and ld (620 mg, 2.10 mmol) were dissolved in glacial acetic acid (20 mL), subjected to nitrogen replacement, stirred at 45 C for 12 h, and cooled to room temperature. The reaction liquid was concentrated under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain the product 10c of interest (350 mg, yield 39.9%) as a yellow solid.
[0377] LCMS in/z = 439.0 [M+Hr.
Step 3: methyl 2-(2-fluoro-4- (4,4,5 -trimethyl-1,3 ,2-dioxaborolan-2-yl)benzy1)-1- (((S )-oxetan-2-y1) methyl)-1H-thieno[2,3-dlimidazole-5-carboxylate (10d) 0,B N
S CO2Me 10d [0378] 10c (350 mg, 0.79 inmol) and pinacol diboronate (305 mg, 1.20 inmol), potassium acetate (235 mg, 2.40 mmol) and Pd(dppt)C12 (65 mg, 0.08 mmol) were added to a reaction flask successively, dissolved in 1,4-dioxane solution (10 mL), subjected to nitrogen replacement, stirred at 90 C for 5 h, cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure to obtain crude product 10d as a yellow oil, which was directly used in the next reaction without further purification.
Step 4: methyl 2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d][1,31dioxo1-4-y1)-2-fluorobenzyl)-1-(((S)-oxetan-2-yOmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylate (10e) CI

J
SCOMe 10e [0379]
The crude products 10d and 4c (344 mg, 1.01 mmol), anhydrous potassium carbonate (230 mg, 1.60 mmol) and Pd(dppf)C12 (65 mg, 0.08 mmol) were added to a reaction flask successively, dissolved in a mixed solution of 1,4-dioxane (10 mL)/water (2 mL), subjected to nitrogen replacement, stirred at 90 C for 5 h and cooled to room temperature. the solid was filtered, the filtrate was concentrated under reduced pressure, and purified on a silica gel column to obtain the product 10e (350 mg, yield 71.2% over two steps) as a yellow solid.
[0380] LCMS m/z = 623.1 [M+H]t Step 5:
2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzokfl[1,31dioxo1-4-y1)-2-fluorobenzy1)-1-(((S)-oxetan-2-yOmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 10) (N 1 \_J, I
N--(( Compound 10S-- COOH
[0381] At room temperature, 10e (350 mg, 0.70 mmol) was dissolved in acetonitrile (5 mL) and water (1 mL), then lithium hydroxide (126.0 mg, 3.01 mmol) was added and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N
hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, concentrated by filtration, and concentrated under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain compound 10 (100.0 mg, yield 29.3%) as a yellow solid.
[0382] LCMS m/z = 609.2 [M+H]+.
[0383] 111 NMR (400 MHz, DMSO-d6) 6 7.73 (br, 111), 7.62-7.54 (m, 411), 7.42-7.33 (m, 2H), 7.22-7.18 (m, 2H), 7.01-6.94 (m, 2H), 5.04-4.96 (m, 1H), 4.62-4.53 (m, 1H), 4.52-4.42 (m, 211), 4.40-4.27 (m, 314), 2.73-2.61 (m, 114), 2.39-2.27 (m, 111), 2.09 (s, 311).
Example 11:
2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylben zo [d] [1,3] di oxo1-4-yOben zy1)-1-4(S)-oxetan-2-yl)methyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 11) ci oTh OH
Compound 11 1(;-CI-0-\--Ck Br Cl 'n 0 F ) 4c 0 Step 2 CI O

7d Step 1 F 0, ha ,0 r Compound 11 11 OH
Step 1: methyl 2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d] [1,3] di oxo1-4-yl)benzy1)-1-(((S)-oxetan-2-yl)methyl)-1H-thi eno [2,3-d]imidazole-5 -carboxylate (11a) cion lia 0, [0384] At room temperature, substrate 4c (91.0 mg, 0.26 mmol) was dissolved in a mixed solvent of 1,4-dioxane (5 mL) and water (0.5 mL), and then 7d (103.0 mg, 0.22 mmol) and Pd(dppt)C12 (18.0 mg, 0.02 minol) and cesium carbonate (215.0 mg, 0.66 minol) were added successively. After the addition, the reaction system was heated to 100 C for overnight reaction under nitrogen protection. The reaction liquid was cooled to room temperature and filtered. The filter cake was washed with dichloromethane (10 mL x 2). The filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain product lla of interest (110.0 mg, yield 82.7% ) as a white solid.

[0385] LCMS m/z = 605.3 [M+1]+.
Step 2: 2-(4-(2-(4-chloro-2-fluoropheny1)-2-methylbenzo[d][1,3]dioxo1-4-yObenzy1)-1-(((S)-oxetan-2-yOmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 11) cI
o --F O. \
S"-yu OH
Compound 11 [0386] At room temperature, substrate ha (110.0 mg, 0.18 mmol) was dissolved in a mixed solvent of acetonitrile (5 mL) and water (1 mL), and then 1 g (CAS: 5807-14-7) (4.4.0 ) Dec-5-ene (23.0 mg, 0.54 mmol) was added. After the addition, the reaction was carried out at room temperature for 24 hours. Acetonitrile was distilled off under reduced pressure. The crude product was dissolved in water (5 mL), adjusted to pH=6 with 1N
hydrochloric acid. The aqueous phase was extracted with ethyl acetate (15 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain compound 11(25.0 mg, yield 23.4%) as a white solid.
[0387] LCMS m/z = 591.2 [M+1] .
[0388] 1H NMR (400 MHz, DMSO-d6) 6 12.90 (brs, 1H), 7.82 (s, 1H), 7.74 -7.67 (m, 214), 7.62 - 7.52 (m, 214), 7.42 - 7.37 (m, 211), 7.36 - 7.32 (dd, 114), 7.16 - 7.10 (m, 1H), 6.95 (d, 2H), 4.97 -4.87 (m, 1H), 4.60- 4.52 (m, 1H), 4.51 - 4.40 (m, 2H), 4.38 -4.28 (m, 314), 2.68 - 2.56 (m, 111), 2.35 - 2.23 (m, 111), 2.06 (s, 314).
Example 12 (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yppiperidin-1-y1)methyl)-1-((tetrahydrofuran-2-yl)m ethyl)-1H-thi eno [2,3 -d] imi dazol e-5-carboxylic acid (Compound 12) NC
OH
Compound 12 NI-12 EN1 0\
BI-N{/(0 12a 1, 0 0 _____ I \ \
0 12b Step 3 N 0 02N S Step 1 02N S Step 2 H2Iµr \---S 0-lb 12c 12d CNN
Cr\, 0N
C\j 1 f-1 ____________________ Br 4\10_40 _______________ NC
Step 4 N
) N--2<sc, Step 6 Step 5 S 0¨
12e 0, 12f NC-, -N
N, No `s-OH
Compound 12 Step 1: methyl (S)-5-nitro-4-(((tetrahydrofuran-2-yl)methyl)amino)thiophene-2-carboxylate (12b) H

02N z---S
12 b [0389] lb (2 g, 7.55 minol), 12a (838 mg, 8.31 mmol), Pd2(dba)3 (346 ing, 0A2 mmol), t-BuBrettphos (366 mg, 036 minol) and cesium carbonate (4.92 g, 15.1 minol) were added into a round bottom flask, and 1,4-dioxane (10 mL) was added. The reaction liquid was stirred at a constant temperature of 100 C for 18 hours after the reaction flask was subjected to nitrogen replacement for protection. After the reaction was completed, the reaction liquid was cooled to room temperature. 20 mL of water was added, then extracted with ethyl acetate (20 mL x 3). The organic phase was washed with aqueous saturated sodium chloride solution, then dried over anhydrous sodium sulfate, filtered and spin-dried. The residue was separated and purified by silica gel column chromatography to obtain 12b (L6 g. yield 74%) as a light yellow solid.
[0390] LCMS in/z = 287.0 [M+1]+.
Step 2: methyl (S )-5-amino-4-(((tetrahydrofuran-2-yl)methyeamino)thiophene-2-carboxylate (12c) H

12c [0391] Raney nickel (60 mg) was added to Me0H (2 mL) and THF (2 mL) containing 12b (200 mg, 0.70 mmol). The reaction liquid was stirred at room temperature for 1 h after the reaction flask was subjected to hydrogen replacement. After the reaction was completed, the reaction liquid was filtered over celite and spin-dried, and the obtained crude product 12c was directly used in the next step.
Step 3: methyl (S)-2-methy1-1-((tetrahydrofuran-2-ypmethyl)-1H-thieno [2, 3-d]
imidazole--carboxylate (12d) /<0 12d [0392] At room temperature, 12c was added to acetonitrile (5 mL), and ytterbium trifluoromethanesulfonate (11 mg, 0.02 mmol) and trimethyl orthoacetate (63 mg, 0.52 mmol) were added to the reaction liquid successively, and the temperature was raised to 70 C for reaction for 2 hours. The reaction liquid was cooled to room temperature. Glacial acetic acid was distilled off under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain 12d (50 mg, yield 26%
over two step).
[0393] LCMS tn/z = 281.1 [M+1]+.
Step 4: methyl (S)-2-(bromomethyl)-1-((tetrahydrofuran-2-y1) methyl)-1H-thieno [2,3-d]imidazole-5-carboxylate (12e) Br N
(\ \>
0 ¨
12e [0394] At room temperature, 12d (50 mg, 0.18 mmol) was dissolved in 1,2-dichloroethane (2 mL) solution, then AIBN (3.2 mg, 0.02 mmol) was added, and NBS(80.1 g, 0.45 mmol) was added in portions in the process of slowly raising the temperature to 50 C. After the addition, the reaction liquid was heated to 70 C and continued to react for 8 h. The reaction liquid was cooled to room temperature, saturated sodium thiosulfate solution (5 mL) was added to quench the reaction and extracted with dichloromethane (5 niL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product. The residue was separated and purified by silica gel column chromatography to obtain 12e (26 mg, yield 40%) as a dark yellow solid.
[0395] LCMS m/z = 359.0 [M+1[+.
Step 5: methyl (S)-24(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-yppiperidin-1-y1)methyl)-1-((tetrahydrofuran-2-y1)methyl)-1H-thieno[2,3-d]imidazole-5-carboxylate (120 NC
/

[0396] At room temperature, 12e (26 ing, 0.07 tntnol) was dissolved in acetonitrile (1 mL) solution, then potassium carbonate (29.0 mg, .21 mmol) and lf-1 (24.8 mg, 0.08 mmol) were added, stirred at room temperature for 18 h. The reaction liquid was filtered to remove the inorganic base, and the filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 12f (20 mg, yield 49%) as a yellow solid.
[0397] LCMS ink = 590.3 1M+11h.
Step 6: (S)-24(4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-yppiperidin-l-yOmethyl)-1-((tetrahydrofuran-2-yemethyl)-1H-thieno[2.3-d]imidazole-5-carboxylic acid (Compound 12) (OD) NC

OH
Compound 12 [0398] At room temperature, 12f (20ing, 0.03 minol) was dissolved in a solution of acetonitrile (1 inL) and water (0.2 inL), then 1 g (CAS: 5807-14-7) (20.9 mg, 0.15 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated. The obtained crude product was subjected to prep-HPLC to obtain compound 12 (5.0 mg, yield 29 %) as a white solid.
[0399] Preparation conditions:
[0400] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0401] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 pin filter membrane to prepare into a sample liquid.
[0402] Mobile phase system: acetonitrile/water (containing 1% TFA).
Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 inUmin, elution time 18 min.
[0403] LCMS in/z = 576.3 [M+1]+.
[0404] 114 NMR (400 MHz, CD30D) 8 7.87 (s, 1H), 7.68 (t, 2H), 7.57 (t, 2H), 6.93 (d, 1H), 6.78 (d, 1H), 5.54 (s, 2H), 4.76-4.62 (in, 2H), 4.62-4.56 (m, 1H), 4.40-4.33 (in, 1H), 4.24-4.15 (in, 1H), 3.95-3.73 (in, 4H), 3.42-3.36 (in, 1H), 3.09-2.98 (in, 1H), 2.27-2.09 (in, 5H), 2.03-1.81 (in, 3H), 1.69-1.58 (in, 1H).
Example 13 (S)-2-44-(64(4-cy ano-2-fluorobenzypoxy)pyridin-2-yepiperazin-l-yl)methyl)- 1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-c arboxylic acid F 0 N71\1) N OH

Compound 13 HINnN
1`3, Boc rro N CI ____________________________ frO N NON _______________________________________________________ Cy N' -) NCF3 Step 1 NCF Boc Step 2 NC-F
KNH
13b 13c C1-i) BrNjz 2bS On) NC
Step 3 ouN, \ 0- step 4 F
N \ OH

13d Compound 13 0 Step 1: tert-butyl 4-(6-((4-cyan o-2-fluorob en zyl)oxy)pyri din-2-yl)pi perazi n e-1-carboxyl ate (13b) NCF
NBoc 13b [0405] 3a (1 g, 3.82 mmol), 13a (781 mg, 4.20 mmol), Pd2(dba)3 (174 mg, 0.19 mmol), RuPhos (178 mg, 0.38 mmol) and cesium carbonate (2.48 g, 7.64 mmol) were added into a round bottom flask, and 1,4-dioxane (10 mL) was added. The reaction liquid was stirred at a constant temperature of 100 C for 18 hours after the reaction flask was subjected to nitrogen replacement for protection. After the reaction was completed, the reaction liquid was cooled to room temperature. 20 mL of water was added, then extracted with ethyl acetate (20 mL x 3). The organic phase was washed with aqueous saturated NaC1 solution, then dried over anhydrous sodium sulfate, filtered and spin-dried. The obtained crude product was separated and purified by silica gel column chromatography to obtain 13b (700 mg, yield 44%) as a light yellow solid.
[0406] LCMS m/z = 357.1 [M-55] .
Step 2: 3-fluoro-4-(((6-(piperazin-1-yl)pyridin-2-ypoxy)methypbenzonitrile (13c) N
NC H
13c [0407] 13b (100 mg, 0.24 mmol) was added into a round bottom flask, then tritluoroacetic acid (0.5 inL) and dichloromethane (1 inL) were added and stirred at room temperature for 5 h. After the reaction was completed, the reaction liquid was directly concentrated under reduced pressure to obtain 13c (90 mg, yield 92%) as a colorless oil. It was directly used in the next step without further purification.
[0408] LCMS m/z = 313.1 [M+1]+.
Step 3: methyl (S)-24(4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-yepiperazin-l-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylate (13d) NC
11\11\-N
F 0 N,N) N

13d [0409] At room temperature, 13c (50 mg, 0.16 mmol) was dissolved in acetonitrile (2 mL) solution, then potassium carbonate (66.3 mg, 0.48 mmol) and id-1 (55.0 mg, 0.16 mmol) were added, stirred at room temperature for 2 h. The reaction liquid was filtered to remove the inorganic base, and the filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 13d (50 mg, yield 54%) as a yellow solid.
[0410] LCMS in/z = 577.2 [M+1] .
Step 4: (S)-24(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-yl)piperazin-l-yOmethyl)-1-(oxetan-2-ylinethyl)-1H-thieno [2,3 -d] imidazole-5-c arboxylic acid (Compound 13) NC
F 0 1µ1,7N1) N
OH

Compound 13 [0411] At room temperature, 13d (50 mg, 0.09 mmol) was dissolved in a solution of acetonitrile (1 mL) and water (0.2 mL), then 1 g (CAS: 5807-14-7) (62.6 mg, 0.45 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated. The obtained crude product was subjected to prep-HPLC to obtain compound 13 (30 mg, yield 59 %) as a white solid.
[0412] Preparation conditions:
[0413] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0414] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 !..tm filter membrane to prepare into a sample liquid.
[0415] Mobile phase system: acetonitrile/water (containing 1%
TFA). Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 mL/min, elution time 18 min.
[0416] LCMS m/z = 563.2 [M+11+.
[0417] 111 NMR (400 MHz, DMSO-d6) 6 13.17 (brs, 114), 7.96 (s, 111), 7.88 (d, 111), 7.74-7.65 (m, 2H), 7.56 (t, 1H), 6.45 (d, 1H), 6.23 (d, 1H), 5.42 (s, 2H), 5.07-4.97 (m, 1H), 4.75-4.66 (m, 111), 4.63-4.46 (m, 411), 4.38-4.31 (m, 111), 4.06-3.52 (m, 411), 3.43-3.15 (m, 4H), 2.79-2.64 (m, 1H), 2.37-2.26 (m, 1H).
Example 14 (S)-24(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-1,4-diazepan-1-yOmethyl)-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 14) N-_¨\ 0 / \\
NCT NS
OH
Compound 14 O
NC
_NBoc Boc alb NH
NC Br N S 0¨ 2b ' a N CI [11(1 14a I 0 N 2 _____________________ IMF 0INTN, j 3a 14b 14c /--\\
NC
(¨N N S 0¨

/¨N N S OH
0 N, ,NO
NN
1 j 14d Compound 14 Step 1: tert-butyl 4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-1,4-diazepane-carboxylate (14b) ,Bob NC

14b [0418] At room temperature, substrate 3a (393.0 mg, 1.5 mmol) was dissolved in 1,4-dioxane (25 inL), and then 14a (301.0 mg, 1.5 mmol), Pd2(dba)3 (138.0 mg, 0.15 mmol), Ru-Phos (140.0 mg, 0.3 mmol) and cesium carbonate (1.5 g, 4.5 minol) were added successively. After addition, the reaction system was heated to 100 C under the protection of nitrogen for overnight reaction. The reaction liquid was cooled to room temperature, filtered to remove inorganic salts, and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain the compound 14b of interest (462.0 ing, yield 72.3%).
[0419] LCMS in/z = 427.2 [M+1]+.
Step 2: 4-(((6-(1,4 -Diazep an-l-yppyridin-2-ypoxy)methyl)- 3 -fluorob enzonitrile (14c) NC /NH

14c [0420] At room temperature, the substrate 14b (462.0 mg, 1.1 mmol) was dissolved in dichloromethane (20 inL), and then trifluoroacetic acid (4 inL) was added dropwise to the reaction system. After the dropwise addition, the reaction was carried out at room temperature for 2 hours. The reaction liquid was concentrated to obtain the crude product 14c as a brown viscous liquid, which was directly used in the next reaction without any purification.
[0421] LCMS m/z = 327.1 [M+1] .
Step 3: methyl (S)-2-((4-(64(4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-1,4-diazepan-1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d]imidazole-5-carboxyl ate (14d) \>
NC N"----S 0-0 N, 14d [0422] At room temperature, the substrate 14c (115.0 mg, 0.35 mmol) was dissolved in acetonitrile (5 inL), then 2b (121.0 mg, 0.35 mmol) and potassium carbonate (244.0 mg, 1.8 mmol) were added successively. After the addition, the reaction system was reacted at room temperature for 4 hours. The reaction liquid was filtered, and the filter cake was washed with dichloromethane (10 mL x 2). The filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 3d (138.0 mg, yield 66.5%) as a white solid.
[0423] LCMS m/z = 591.3 [M+1]+.
Step 4: (S)-2-44-(6-((4-cy ano-2-fluorobenzypoxy)pyridin-2-y1)-1,4-diazep an-1-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylic acid (Compound 14) (c>"1 NC
N
K
N -------S OH
0õNõNIJõ
Compound 14 [0424] At room temperature, substrate 14d (138.0 mg, 0.2 mmol) was dissolved in a mixed solvent of acetonitrile (5 mL) and water (1 mL), and then 1 g (CAS: 5807-14-7) (98.0 mg, 0.7 mmol) was added. After the addition, the reaction was carried out at room temperature for 24 hours. Acetonitrile was distilled off under reduced pressure. The crude product was dissolved in water (5 mL), adjusted to pH=6 with 1N hydrochloric acid. The aqueous phase was extracted with ethyl acetate (15 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain compound 14 (66.0 mg, yield 48.9%) as a white solid.
[0425] LCMS m/z = 577.2 [M+1]+.
[0426] 11-1 NMR (400 MHz, DMSO-d6) 6 12.94 (brs, 114), 7.87 ¨
7.80 (m, 111), 7.77 (s, 1H), 7.68¨ 7.63 (m, 1H), 7.62 ¨ 7.56 (m, 1H), 7.43 (t, 1H), 6.16 (d, 1H), 6.05 (d, 1H), 5.36 (s, 2H), 4.99 ¨ 4.92 (m, 1H), 4.53 ¨ 4.40 (m, 2H), 4.38 ¨ 4.28 (m, 2H), 3.87-3.74 (m, 21-1), 3.61 (t, 211), 3.51 (t, 21-1), 2.70¨ 2.62 (m, 211), 2.60¨ 2.52 (m, 311), 2.34 ¨ 2.20 (m, 1H), 1.75¨ 1.66 (m, 2H).
Example 15 (S)-24(4-(24(4-cyano-2-fluorobenzypoxy)pyridin-3-yppiperidin-l-ypmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 15) CN
FH

N
N OH

Compound 15 0 / Boc Boc Boc-N1BL N N¨\

N A 15a-1 1_0\yc N
F \ ___________ 0)0 Br ¨ Step 1 NC N¨ Step 2 Njc__/ 'NI¨
Step 3 15a 15b 15c N CN
HN¨ Br , CN
¨1 N S \ I

2b 0 F "IN
0¨) ) \
NC 41, \N¨ Step 4 Step 5 Nb7C
N4s0H
15d 0 15e 0 Compound 15 Step 1: tert-butyl 24(4-cyano-2-fluorobenzypoxy)-3',6'-dihydro-[3,4'-bipyridine]-1'(2'14)-carboxylate (15b) Boo, ¨
F

NC N-15b [0427] 15a-1 (504.9 mg, L63 tnmol), 15a (500 mg, 1.63 mmol), Pd(dppf)C12 (133 mg, 016 mmol) and cesium carbonate (1.06 g, 126 mmol) were added into a round bottom flask, and 1,4-dioxane (10 mL) was added. The reaction liquid was stirred at a constant temperature of 100 C for 18 hours after the reaction flask was subjected to nitrogen replacement for protection. After the reaction was completed, the reaction liquid was cooled to room temperature. 10 inL of water was added, then extracted with ethyl acetate (6 inL x 3). The organic phase was washed with aqueous saturated NaCl solution, then dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to dryness. The obtained crude product was separated and purified by silica gel column chromatography to obtain 15b (600 mg, yield 90%) as a light yellow solid.
[0428] LCMS tn/z = 410.2 [M+1] .
Step 2: tert-butyl 4-(24(4-cyano-2-fluorobenzypoxy)pyridin-3-yl)piperidine-1-c arboxylate (15c) Boc NCN-15c [0429] Palladium on carbon (60 mg) was added to tetrahydrofuran (10 inL) containing 15b (600 mg, 1.47 mmol). The reaction liquid was stirred at room temperature for 18 h after the reaction flask was subjected to hydrogen replacement. After the reaction was completed, the reaction liquid was filtered over celite, and the liquid was concentrated to dryness. The crude product was separated and purified by silica gel column chromatography to obtain 15c (450 mg, yield 74%) as a light yellow solid.
[0430] LCMS in/z = 412.1 [M+1]+.
Step 3: 3-fluoro-4-(((3-(piperidin-4-yl)pyridin-2-yl)oxy)methyl)benzonitrile (15d) HN_ NC N-15d [0431] 15c (100 mg, 0.24 mmol) was added into a round bottom flask, then tritluoroacetic acid (0.5 inL) and dichloromethane (1 inL) were added and stirred at room temperature for 5 h. After the reaction was completed, the reaction liquid was directly concentrated under reduced pressure to obtain 15d (90 mg, yield 92%) as a colorless oil. It was directly used in the next step without further purification.
[0432] LCMS in/z = 312.1 [M+1]+.
Step 4: methyl (S)-24(4-(24(4-cyano-2-fluorobenzyl)oxy)pyridin-3-yepiperidin-1-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylate (15e) CN
FH
N \ 0 N

15e [0433] At room temperature, 15d (25 mg, 0.08 minol) was dissolved in acetonitrile (1 inL) solution, then potassium carbonate (33.2 mg, 0.24 mmol) and 2b (27.5 mg, 0.08 inmol) were added, stirred at room temperature for 5 h. The reaction liquid was filtered to remove the inorganic base, and the filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 15e (20 mg, yield 43%) as a brown solid.
[0434] LCMS in/z = 576.2 [M+1]+.
Step 5: (S)-24(4-(24(4-cyano-2-fluorobenzypoxy)pyridin-3-yppiperidin-l-ypinethyl)-1-(oxetan-2-ylinethyl)-1H-thieno [2,3 -d] imid azole-5-c arboxylic acid (Compound 15) CN

N
N OH
I z 0 Compound 15 [0435] At room temperature, 15e (20ing, 0.03 mmol) was dissolved in a solution of acetonitrile (1 mL) and water (0.2 naL), then 1 g (CAS: 5807-14-7) (20.9 mg, 0.15 minol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethaneanethano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated. The obtained crude product was subjected to prep-HPLC to obtain compound 15 (6.0 mg, yield 32%) as a white solid.
[0436] Preparation conditions:
[0437] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0438] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 [fin filter membrane to prepare into a sample liquid.
[0439] Mobile phase system: acetonitrile/water (containing 1% TFA).
Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 inL/min, elution time 18 min.
[0440] Compound 15:
[0441] LCMS in/z = 562.2 [M+1]+.
[0442] 11-1 NMR (400 MHz, DMSO-d6) 8 13.22 (brs, 1H), 8.07-8.04 (in, 1H), 7.98 (s, 1H), 7.91 (d, 1H), 7.74-7.67 (in, 2H), 7.63-7.56 (in, 1H), 7.08-7.02 (in, 1H), 5.53 (s, 2H), 5.06-4.98 (in, 1H), 4.77-4.63 (in, 3H), 4.63-4.56 (in, 1H), 4.54-4.47 (in, 1H), 4.38-4.31 (in, 1H), 3.74-3.64 (in, 2H), 3.36-3.23 (in, 2H), 3.15-3.04 (in, 1H), 2.77-2.66 (in, 1H), 2.37-2.26 (in, 1H), 2.09-1.99 (in, 2H), 1.96-1.84 (in, 2H).
Example 16 (S)-2-44-(64(4-cy ano-2-fluorobenzypoxy)pyridin-2-yepiperidin- 1-yl)methyl)-3-(oxetan-2-ylinethyl)-3H- thieno [2,3-d] imidazole-5-c arboxylic acid C;,--NC N

F -OH
Compound 16 CI'C)Me rOMe \--\ OMe 16a 01NH2 02N, 0 H2N 0 le-1 , 0 lc-0¨ Step 2 f____("N s o¨
Step 3 CIXSO¨ Step 1 L-0 H 16b Lo H 16c NC-NH
N S 0 F lf-1 NC,2 1 /¨K Isl-A Io , N, N ' ' Step 5 CI N Step 4 --1,f0 16d F -- 0-16e 0--i NO IN( ,01'S
--C .--- 0 F 1;

Compound 16 Step 1: methyl (S)-4-nitro-5-((oxetan-2-ylmethyl)amino)thiophene-2-carboxylate (16b) 02N , o 1 ________________________________________________ F---N---s O¨

H

16b [0443] At room temperature, 16a (2 g, 9.0 mmol) was dissolved in acetonitrile (50 triL) solution, then potassium carbonate (3.73 g, 27.0 inmol) and lc-1 (783 mg, 9.0 mmol) were added, and the temperature was raised to 70 C for 18 h. The reaction liquid was filtered to remove solids, and the filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 16b (2 g, yield 83%).
[0444] LCMS in/z = 273.1 [M+1]+.
Step 2: methyl (S)-4-amino-5-((oxetan-2-ylmethyl)amino)thiophene-2-carboxylate (16c) 16c [0445] Zinc powder (L2 g, 18.4 inmol) was added to acetic acid (20 inL) containing 16b (1.0 g, 3.68 mmol), and the reaction liquid was stirred at room temperature for 1 h.
After the reaction was completed, the reaction liquid was filtered over celite, and the filtrate was concentrated to dryness. The residue was separated and purified by silica gel column chromatography to obtain 16c (700 mg, yield 79%) as a light yellow solid.
[0446] LCMS in/z = 243.1 [M+1]+.
Step 3: methyl (S)-2-(chloroinethyl)-3 -(oxetan-2-ylinethyl)-3H-thieno [2,3 -(1] imidazole-5-carboxylate (16d) CI Nj)_ 0¨
16d [0447] At room temperature, 16c (700 mg, 2.89 ininol) was added to acetonitrile (20 inL), and le-1 (668.2 ing, 4.34 -mime and p-toluenesulfonic acid (24.9 ing, 0.14 ininol) were added to the reaction liquid successively, heated to 60 C for 5 hours.
Acetonitrile was distilled off under reduced pressure, and the obtained crude product was purified by silica gel column chromatography to obtain 16d (45 ing, yield 5.2%) as a yellow solid.
[0448] LCMS in/z = 301.1 [M+1]+.
Step 4: methyl (S)-24(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-yepiperidin-l-yl)methyl)-3-(oxetan-2-ylmethyl)-3H-thieno[2,3-(11imidazole-5-carboxylate (16e) NC
/ S


16e [0449] At room temperature, 16d (45 ing, 0.15 minol) was dissolved in acetonitrile (2 inL) solution, then potassium carbonate (62.1 mg, 0.45 mmol) and if-1 (46.5 mg, 0.15 mmol) were added, stirred at room temperature for 18 h. The reaction liquid was filtered to remove solids, and the filtrate was collected and concentrated to obtain a crude product, which was separated and purified by silica gel column chromatography to obtain 16e (50 mg, yield 58%) as a yellow solid.
[0450] LCMS m/z = 576.2 1M+11+.
Step 5: (S)-2-((4-(64(4-cyano-2-fluorobenzypoxy)pyri din-2-y] ipiperi din -1-yl)methyl)-3-(oxetan-2-y1 methyl)-3H-thi eno 12,3-dlimidazole-5-carboxylic acid (Compound 16) o NC
s 0 N ' ---' 0 OH
Compound 16 [0451] At room temperature, 16e (40 mg, 0.07 mmol) was dissolved in a solution of acetonitrile (1 inL) and water (0.2 inL), then 1 g (CAS: 5807-14-7) (483 mg, 0.35 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated. The obtained crude product was subjected to prep-HPLC to obtain compound 16 (20 mg, yield 51 %) as a white solid.
[0452] Preparation conditions:
[0453] instrument and preparative column: waters 2767 (preparative liquid phase chromatographic instrument) was used; the preparative column model was SunFire@Prep C18 (19 mm x 250 mm).
[0454] Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 [lin filter membrane to prepare into a sample liquid.
[0455] Mobile phase system: acetonitrile/water (containing 1%
TFA). Gradient elution: acetonitrile content 20-65%, elution flow rate: 12 inUmin, elution time 18 min [0456] LCMS in/z = 562.3 [M+1]+.
[0457] 114 NMR (400 MHz, DMSO-d6) 8 13A5 (brs, 1H), 7.89 (d, 1H), 7.86 (s, 1H), 7.76-7.69 (in, 3H), 6.98-6.91 (in, 1H), 6.79 (d, 1H), 5A9 (s, 2H), 5.14-5.06 (in, 1H), 4.69 (s, 2H), 4.66-4.60 (m, 1H), 4.57-4.49 (m, 2H), 4.40-4.33 (m, 1H), 3.80-3.65 (m, 2H), 3.32-3.19 (m, 214), 2.96-2.86 (m, 111), 2.80-2.70 (m, 114), 2.42-2.31 (m, 111), 2.12-1.93 (m, 411).
Example 17 (S)-2-((4-(6-((4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-yppiperidin-1-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 17) O¨L7 NC

OH
Compound 17 ,N Boc ^N Bac Boc step 3 Boc __________________________________________ HO
Step 1 Step 2 N N, N, 1_J
F F
17a 17b 17c 17d o (5])N
NC
, H Step 5 Step 6 NC
_,N,Tv ___________________ WI 0 N
Step 4 F F F- OH
17e 17f Compound 17 Step 1: 2-(1-(tert-butoxycarbonyl)piperidin-4-y1)-5-fluoropyridine1-oxide (17b) Boc 17b [0458] At room temperature, in-chloroperoxybenzoic acid (3.08 g, 17.82 minol) was added to a solution of 17a (2.00 g, 7.13 inmol) in dichloromethane (20 ml), and then stirred and reacted at room temperature for 14 hours. The insolubles were removed by filtration, the liquid was washed with an equal volume of water, dried over anhydrous sodium sulfate and concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain the product 17b of interest (1.20 g, yield:
56.8%).
Step 2: tert-buty14-(5-fluoro-6-hydroxypyridin-2-yppiperidine-1-carboxylate (17c) N,Boc HO N
17c [0459] Under ice-water bath, triethylamine (0.82 g, 8.10 mmol) was added to a solution of 17b (L20 g, 4.05 mmol) and trifluoroacetic anhydride (8.51 g, 40.5 mmol) in tetrahydrofuran (20 ml). After one hour, the temperature was raised to room temperature to react for 14 hours. The reaction liquid was concentrated to dryness under reduced pressure, and the residue was separated and purified by column chromatography to obtainl7c (0.8 g, yield: 66.6%).
[0460] LCMS in/z=241.1 [M-55] +.
Step 3: tert-butyl 4-(6-((4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-yl)piperidine-1-carboxylate (17d) NC -1\1,Boc , 17d [0461] At room temperature, tri-n-butylphosphine (513 mg, 2.54 mmol), diisopropyl azodicarboxylate (513 mg, 2.54 mmol) were added to a solution of 17c (0.5 g, 1.69 minol) and 3-Fluoro-4-(hydroxymethyebenzonitrile (383 mg, 2.54 mmol) in toluene (5 me. The temperature was raised to 80 C to react for 14 hours. The reaction liquid was concentrated under reduced pressure. The residue was separated and purified by silica gel column chromatography to obtain 17d (0.2 g, yield: 27.6%).
[0462] LCMS in/z=374.1[M-55] .
Step 4: 3-fluoro-4-(43-fluoro-6-(piperidin-4-yl)pyridin-2-yl)oxy)tnethyl)benzonitrile (17e) NC
NH
F
17e [0463] At room temperature, trifluoroacetic acid (2 ml) was added to a solution of 17d (0.2 g, 0.47 mmol) in dichloromethane (2 ml), and then reacted at room temperature for 3 hours. The reaction liquid was concentrated to dryness under reduced pressure, and the residue was separated and purified by column chromatography to obtain 17e (0.1 g, yield: 64.60%).
Step 5: methyl (S)-24(4-(6-((4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-yl)piperidin-l-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-dlimidazole-5-carboxylate (170 0 m NC

17f [0464] At room temperature, potassium carbonate (82.93 mg, 0.60 mmol) was added to a solution of 17e (0.1 g, 0.30 mmol) and 2b (103.56 mg, 0.30 mmol) in acetonitrile (10 ml), and then reacted at room temperature for 4 hours. The solid was removed by filtration, and the liquid was concentrated to dryness to obtain 17f. The crude product yield was calculated as 100% and the next step was directly carried out.
[0465] LCMS iniz=594.2(M+1) .
Step 6: (S)-24(4-(6-((4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-yepiperidin-l-yOmethyl)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylic acid (Compound 17) on NC

F OH
Compound 17 [0466] At room temperature, lg (CAS: 5807-14-7) (139.2 mg, 1.00 mmol) was added to a mixed solution of 17f (0.15 g, 0.25 mmol) in acetonitrile (5 ml) and water (5 ml) and then reacted at room temperature for 14 hours. The reaction liquid was concentrated to obtain a crude product. The crude product was purified by Pre-HPLC (instrument and preparative column: Waters 2767 (preparative liquid phase chromatographic instrument) was used, and the preparative column model was Sunfire@PrepC18, 5 [im, inner diameter x length = 19 mm x 250 mm). Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 [tm filter membrane to prepare into a sample liquid. Mobile phase system: acetonitrile/water (containing 0.5% ammonium bicarbonate).
Gradient elution method: gradient elution with acetonitrile from 5% to 50% (flow rate:
15 mL/min;
elution time 15 min), compound 17 (0.05 g, yield: 34.51%) was obtained.
[0467] LCMS m/7 = 580.2[M+1] .
[0468] 111 NMR (400 MHz,CD30D) 6 7.69 (t, 111), 7.65 (s, 111), 7.63-7.53 (m, 211), 7.45-7.38 (m, 1H), 6.87-6.83(m, 1H), 5.59 (s, 2H), 5.26-5.18 (m, 1H), 4.72-4.54(m, 3H), 4.47-4.39 (m, 111), 4.12-4.00 (m, 211), 3.28-3.16 (m, 211), 2.82-2.69 (m, 211), 2.59-2.44 (m, 3H), 1.95-1.82 (m, 4H).
Example 18 (S)-2-(4-(644-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 18) OraN
NC

F OH
Compound 18 F
NC'C T: OH
002Me 18b NC 0.B1 NICS-0 10d F
CI, Br _______________________________________________________ oth 111-P 0 N Br ___________________________________________ Step 1 F Fp' Step 2 18a 18c 18d Step 3 NC 0 N
F 'T-C) OH
Compound 18 Step 1: 44(6-bromo-3-fluoropyridin-2-yl)oxy)methyl)-3-fluorobenzonitrile (18c) NC
0 N Br F
18c [0469] Under ice bath, 18a (278 mg, L32 mmol) was added to a solution of 18b (200 mg, L32 mmol) and potassium tert-butoxide (22L8 mg, L98 mmol) in tetrahydrofuran (6 ml) and reacted at room temperature for 3 hours. The insolubles were removed by filtration, the liquid was washed once with aqueous saturated ammonium chloride solution (6 ml), dried over anhydrous sodium sulfate, concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain the product 18c of interest (250 mg, yield: 58.5%).
Step 2: methyl (S)-2-(4-(64(4-cy ano -2-fluorobenzyl)oxy)-5-fluoropyridin-2-y1)-2-fluorobenzy1)- 1-(oxetan-2-ylmethyl)- 1H-thieno [2,3-d] imidazole-5 -c arboxy late (18d) FO
NC
/

18d [0470] The crude products 10d and 18c (178 mg, 0.55 minol), anhydrous potassium carbonate (115 ingõ 0.80 mmol) and Pd(dppf)C12 (32.5 mg, 0.04 mmol) were added to a reaction flask successively, dissolved in a mixed solution of 1,4-dioxane (5mL)/water (1mL), subjected to nitrogen replacement, stirred at 90 C for 5 h and filtered to remove solids. The filtrate was concentrated under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain the product 18b (140 mg, yield 42.1% over two steps) as a yellow solid.
[0471] LCMS in/z = 605.2 [M+Hr.
Step 3: (S)-2-(4-(64(4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-y1)-2-f1uorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-c arboxylic acid (Compound 18) F
NC
/

OH
Compound 18 [0472] At room temperature, 18b (80 mg, 0.13 mmol) was dissolved in a solution of acetonitrile (3 tnL) and water (0.6 mL), then 1 g (CAS: 5807-14-7) (54.2 mg, 0.39 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, concentrated by filtration, and concentrated under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain compound 18 (7 mg, yield 9.1%) as a white solid.
[0473] LCMS tn/z = 591.1 [M+Hr.
[0474] 11-1 NMR (400 MHz, DMSO-do) 6 7.96-7.91 (m, 1H), 7.86-7.72 (m, 6H), 7.70-7.65 (m, 1H), 7.29 (t, 1H), 5.70 (s, 2H), 5.05-4.97 (m, 1H), 4.64-4.56 (m, 1H), 4.52-4.45 (in, 2H), 4.41-4.28 (in, 3H), 2.70-2.64 (in, 1H), 2.35-2.29 (in, 1H).
Example 19 (S)-2-(4-(6-((4-cyanobenzypoxy)-5-fluoropyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno12,3-dlimidazole-5-carboxylic acid (Compound 19) FO
NC
/

OH
Compound 19 F
NC P-13 S CO2Me CI Br (r3-19b --\r--No 10d N o .N_Br N
18a Step 1 F'' Step 2 19c 19d Step 3 Ne....rni ^ N
N---(s it) -F OH
Compound 19 Step 1: 4-(((6-bromo-3-fluoropyridin-2-yl)oxy)methylibenzonitrile (19c) NC
0 N Br F
19c [0475] Under ice bath, 18a (300 mg, L43 mmol) was added to a solution of 19b (285.7 mg, 1.43 mmol) and potassium tert-butoxide (240 mg, 2.15 mmol) in tetrahydrofuran (10 ml) and stirred and reacted at room temperature for 3 hours. The insolubles were removed by filtration, the liquid was washed with an equal volume of water, dried over anhydrous sodium sulfate and concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain the product 19c of interest (250 mg, yield: 57.1%).
Step 2: methyl(S)-2-(4-(64(4-cyanobenzypoxy)-5-fluoropyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylinethyl)-1H-thieno [2,3 -d] imidazole-5-c arboxylate (19d) NC
I /

19d [0476] The crude products 10d and 19c (168.3 ing, 0.55 intnol), anhydrous potassium carbonate (115 mg, 0.80 ininol) and Pd(dppf)C12 (32.5 mg, 0.04 inmol) were added to a reaction flask successively, dissolved in a mixed solution of 1,4-dioxane (5 mL)/water (1 mL), subjected to nitrogen replacement, stirred at 90 C for 5 h and cooled to room temperature. the solid was filtered, the filtrate was concentrated under reduced pressure, and purified on a silica gel column to obtain the product 19d (180 mg, yield 67 % over two steps) as a yellow solid.
[0477] LCMS m/z = 587.1 [M+H]t Step 3: (S)-2-(4-(6-((4-cyanobenzyl)oxy)-5-fluoropyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-114-thieno[2,3-d]imidazole-5 -carboxylic acid (Compound 19) FO
NC
/

OH
Compound 19 [0478] At room temperature, 19d (80 mg, 0.14 mmol) was dissolved in a solution of acetonitrile (3 inL) and water (0.6 inL), then 1 g (CAS: 5807-14-7) (56.9 mg, 0.41 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with IN hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, concentrated by filtration, and concentrated under reduced pressure, and the residue was separated and purified by silica gel column chromatography to obtain compound 19 (40 mg, yield 50%) as a white solid.
[0479] LCMS in/z = 573.1 [M+Hr.
[0480] 11-1 NIVIR (400 MHz, DMSO-do) 6 7.90-7.77 (in, 6H), 7.71 (d, 2H), 7.68-7.64 (in, 1H). 7.38 (t, 1H), 5.67 (s, 2H), 5.04-4.96 (in, 1H), 4.63-4.55 (in, 1H), 4.53-4.44 (in, 2H), 4.42-4.28 (m, 3H), 2.72-2.62 (in, 1H), 2.38-2.27 (in, 1H).
Example 20 (S)-2-(4-(64(4-cyanobenzypoxy)pyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3 -di imidazole-5-c arboxylic acid (Compound 20) FO
NC
/

OH
Compound 20 -]
0-B 10 \I--(sCO2Me CI k ________________________ 19b NC. 0 10d NC
CI k CI
rsk 7 A, Step 1 Step 2 20a 20b FO
Step 3 N
I
OH
Compound 20 Step 1: 4-(((6-chloropyridin-2-yeoxy)methyl)benzonitrile (20a) NC

20a [0481] Under ice bath, 2,6-dichloropyridine (5 g, 33.8 mmol) was added to a solution of 19b (4.4 g, 33.8 mmol) and potassium tert-butoxide (7.6 g, 67.6 mmol) in tetrahydrofuran (100 ml) and stirred and reacted at room temperature for 3 hours. The insolubles were removed by filtration, the liquid was washed with an equal volume of water, dried over anhydrous sodium sulfate and concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain the product 20a of interest (4.7 g, yield: 57.0%).
Step 2: nriethyl(S)-2-(4-(64(4-cyanobenzypoxy)pyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylate (20b) F
NC
I /

20b [0482] The crude products 10d and 20a (134.2 mg, 0.55 mmol), anhydrous potassium carbonate (115 mg, 0.80 mmol) and Pd(dppf)C12 (32.5 mg, 0.04 inmol) were added to a reaction flask successively, dissolved in a mixed solution of 1,4-dioxane (5 mL)/water (1 inL), subjected to nitrogen replacement, stirred at 90 C for 5 h and cooled to room temperature, the solid was filtered, the filtrate was concentrated under reduced pressure, and purified on a silica gel column to obtain the product 20b (160 mg, yield 61.5% over two steps) as a yellow solid.
[0483] LCMS in/z = 569.1 [1\4+Hr.
Step 3: (S)-2-(4-(64(4-cy anobenzyl)oxy)pyridin-2-y1)-2-fluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2, 3-d] itnid azole-5-c arboxylic acid (Compound 20) FO
NC
/

OH
Compound 20 [0484] At room temperature, 19d (80 mg, 0.14 mmol) was dissolved in a solution of acetonitrile (3 InL) and water (0.6 InL), then 1 g (CAS: 5807-14-7) (56.9 mg, 0.41 mmol) was added, and the reaction was continued for 24 h. The reaction liquid was adjusted with 1N hydrochloric acid to pH=6, and then extracted with a mixed solvent of dichloromethane:methano1=10:1 (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, concentrated by filtration, and concentrated under reduced pressure. The residue was purified by Pre-HPLC (instrument and preparative column:
Waters 2767 (preparative liquid phase chromatographic instrument) was used, and the preparative column model was Sunfire@PrepC18, 5 [tin, inner diameter x length = 19 mm x 250 mm). Preparation method: The crude product was dissolved in DMF and filtered with a 0.45 [tin filter membrane to prepare into a sample liquid. Mobile phase system:

acetonitrile/water (containing 0.5% ammonium bicarbonate). Gradient elution method:
acetonitrile content 5-50% (flow rate: 15 mL/min; elution time 15 min), compound 20 (20 mg, yield: 25.8%) was obtained.
[0485] LCMS m/z=555.1 [M+1]+.
[0486] 1H NMR (400 MHz, DMSO-do) 6 7.88-7.80 (m, 6H), 7.68 (d, 2H), 7.63 (d, 111), 7.38 (t, 114), 6.93 (d, 114), 5.69 (s, 211), 5.05-4.97 (m, 114), 4.63-4.56(m, 111), 4.52-4.44 (m, 2H), 4.42-4.29 (in, 3H), 2.72-2.62 (m, 1H), 2.37-2.28 (m, 1H).
Example 21 (S)-2-(4-(6-((4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-dlimidazole-5-carboxylic acid (Compound 21) NC F Cra) F
N
I /
0 1\1 N \

Compound 21 ly_3 Br,,---õrõF Br -,,, F Br ¨ F Q
i ---T NH HCI H2N 1d F-ii ,)--. ,CO2H Step 1 F CONH2 Step 2 F J1. ,--A. CN
Step 3 F'-'-- OEt Step 4 21a 21b 21c 21d F (-1- NC., (5]) F NC, F F
F 3a 0N) CO2Me ) HO d IR---i, 1 )' ¨ r.--- -NI
,,,, N--/c?-1 Br CO2Me CO2Me 1 I I
S
F Step 5 H6 F Step 6 21e 21 21g rr1L'i N, N O
H NC-F
-I" 7 F
1 ---TN__ Step 7 0 N, --, N---t 11 Compound 21 Step 1: 2-(4-bromo-2,5-difluorophenyl)acetic acid (21b) Br F

21b [0487] 21a (7.5 g, 29.88 mmol) was added into thionyl chloride (20.0 mL), and stirred at 70 C for 2 hours. Then the reaction liquid was cooled to room temperature and concentrated to dryness and the concentrated residue was added to dichloromethane (30 mL). The system was placed at 0 C and ammonia water (15 mL) was added, and a large amount of white solid was precipitated and filtered. The obtained solid was further washed with water (20 mL x 2), and the solid was dried to give 21b (7.3 g, yield 97%).
[0488] LCMS m/z = 250.1 [M+1]+.
Step 2: 2-(4-bromo-2,5-difluorophenypacetonitrile (21c) Br CN
21c [0489] 21b (7.3 g, 29.20 mmol) was added into phosphorus oxychloride (30.0 mL), and stirred at 100 C for 2 hours. Then the reaction liquid was cooled to room temperature, the system was poured into ice water (100 mL), and then extracted with ethyl acetate (100 inL x 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain 21c (6.0 g, yield 88%).
[0490] LCMS in/z = 232.1 [M+1] .
Step 3: ethyl 2-(4-bromo-2,5-difluorophenypacetimidate hydrochloride (21d) Br NH.HCI
OEt 21d [0491] 21c (6.0 g, 25.86 mmol) was added to anhydrous ethanol (3.0 mL) and fully stirred at 0 C. At the same time, 33% hydrochloric acid-ethanol solution (25 inL) was added. The temperature was gradually raised to room temperature and the reaction liquid was stirred for 10 hours. Diethyl ether (40 inL) was then added, filtered to obtain a solid that was further washed with diethyl ether (100 mL x 2 ), and dried to obtain 21d (5.8 g, yield 71%).
[0492] LCMS inh = 278.1 [M-C1r.
Step 4: methyl (S)-2-(4 -bromo-2,5-difluorobenzy1)- 1-(oxetan-2-ylinethyl)- 1H-thieno [2,3-d]imidazole-5-carboxylate (21e) OTh /
Br S CO2Me 21e [0493] 21d (3.0 g, 9.58 mmol) and id (2.5 g, 10.3 inmol) were added to glacial acetic acid (50 inL), and stirred at 60 C for 10 hours. The reaction system was cooled to room temperature, then was slowly added to an aqueous saturated sodium bicarbonate solution (200 inL) to quench the reaction, and then extracted with ethyl acetate (50 inL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered.
The filtrate was spin-dried, and the residue was separated and purified by silica gel column chromatography to obtain 21e (1.34 g, yield 30%).
[0494] LCMS in/z = 457.1 [M+1] .
Step 5: (S)-(2,5-difluoro-4((5-(methoxycarbony1)- 1-(oxet an-2-ylmethyl)- 1H-thieno [2,3 -d]imidazol-2-yl)methyl)phenyl)boronic acid (210 FO
HO,B
S CO2Me OH F
21f [0495] 21e (600 mg, 1.3 mmol) and pinacol biborate (519 mg, 1.9 mmol), Pd(dpp0C12=DCM (110 mg, 0.1 mmol), and potassium acetate (400 mg, 4.0 mmol) were added to 1,4-dioxane (10 inL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (50 inL) was added, and extracted with ethyl acetate (50 inL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness to obtain crude compound 21f (438 mg, yield 80%).
[0496] LCMS m/z = 423.1[M+1]+.
Step 6: methyl (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)pyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-c arboxylate (21g) CC-J) S CO2Me F
21g [0497] 21f (60 mg, 0.14 mmol) and 3a (34 mg, 0.13 mmol), Pd(dppf)C12.DCM (10 mg, 0.01 mmol) and potassium carbonate (36 mg, 0.26 mmol) were added to a mixed solution of 1,4-dioxane (2 mL) and water (0.5 mL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (2 mL) was added, and extracted with ethyl acetate (10 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain 21g (46 mg, yield 54%).
[0498] LCMS m/z = 605.2 [M+1]+.
Step 7: (S)-2-(4-(6-((4-cyano-2-fluorobenzypoxy)pyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid (Compound 21) NC

F
Compound 21 [0499] 21g (46 ing, 0.076 mmol) and 1 g (32 ing, 0.228 mmol) were added to a mixed solution of acetonitrile (3 inL) and water (0.5 mL), and stirred at room temperature for 16 hours. Then the reaction liquid was adjusted to pH=6 with 1N hydrochloric acid, continuously stirred until a white solid precipitated, and filtered and the solid was washed with water. The solid was dissolved in dichloromethane, dried over anhydrous sodium sulfate and filtered. The organic phase was concentrated to dryness, the residue was separated and purified by silica gel column chromatography to obtain compound 21 (10 mg, yield 22%).
[0500] LCMS in/z = 591.0 [M+1]+.

1105011 1H NMR (400 MHz, DMSO-d6) 6 12.91 (br, 1H), 7.93-7.83 (m, 3H), 7.78-7.67 (m, 3H), 7.53-7.48 (m, 1H), 7.37-7.30 (m, 1H), 7.01-6.95 (m, 1H), 5.59 (s, 214), 5.08-4.99 (m, 1H), 4.67-4.58 (m, 1H), 4.55-4.45 (m, 2H), 4.43-4.29 (m, 3H), 2.74-2.64 (m, 1H), 2.40-2.29 (m, 1H).
Example 22 (S)-2-(4-(6-((4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thienor,3-dlimidazole-5-carboxylic acid (Compound 22) oI
NC F
/

Compound 22 FO
co2NmCe.: N
n'n NC mr, HO g N NC F
NB 1114P 0 r OH F 21f = N, LT: 02Me Step 2 Step 1 F N_cCO2H
18c 22b Compound Step 1: methyl (S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thienor,3-dlimidazole-5-carboxylate (22b) NC
/

S CO2Me F
22b [0502] 21f (143 ing, 0.34mm01) and 18c (110 ing, 0.34mmo1), Pd(dppf)C12.DCM (40 mg, 0.04 mmol) and potassium carbonate (140 mg, 1.01 mmol) were added to a mixed solution of 1,4-dioxane (5 inL) and water (1 mL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (5 tuL) was added, and extracted with ethyl acetate (10 tuL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain 22b (170 nag, yield 80%).

1105031 LCMS m/z = 623.1 [M+1]+.
Step 2:
(S)-2-(4-(64(4-cyano-2-fluorobenzyl)oxy)-5-fluoropyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d]imidazole-5-carboxylic acid (Compound 22) NC
/

F
Compound 22 [0504] 22b (50 mg, 0.08 mmol) and 1 g (34 mg, 0.24 mmol) were added to a mixed solution of acetonitrile (5 inL) and water (1 inL), and stirred at room temperature for 16 hours. Then the reaction liquid was adjusted to pH=6 with 1N hydrochloric acid, continuously stirred until a white solid precipitated, and filtered and the solid was washed with water. The solid was dissolved in dichloromethane, dried over anhydrous sodium sulfate and filtered. The organic phase was concentrated to dryness, the residue was separated by preparative HPLC to obtain compound 22 (10 mg, yield 20%).
[0505] LCMS tn/z = 609.1 [M+1]+.
[0506] 114 NMR (400 MHz, DIVISO-d6) 6 7.95-7.90 (in, 1H), 7.88-7.82 (in, 2H), 7.80-7.67 (m, 3H), 7.55-7.50(m, 1H), 7.37-7.31 (m, 1H), 5.68 (s, 2H), 5.06-5.00 (in, 1H), 4.66-4.59 (m, 1H), 4.54-4.45 (m, 2H), 4.40-4.29 (m, 3H), 2.69-2.64 (m, 1H), 2.37-2.30 (m, 1H).
Example 23 (S)-2-(4-(64(4-cy anobenzypoxy)pyridin-2-y1)-2,5 -difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d] imidazole-5-carboxylic acid (Compound 23) NC
() N
N¨_/<

Compound 23 (r)NC , NC
N
HO B N r "0õ!11''i NC
N N 0]) 1-:õ.11-.. 0 N CI OH F 2if S CO2Me q1113-'' N, 1 g H
J=J
co2Nne Step 2 'IL; S CO2H Step 1 20b 23b Compound Step 1: methyl (S)-2-(4-(64(4-cyano-2-fluorobenzypoxy)-5-fluoropyridin-2-y1)-2,5-difluorobenzy1)-1-(oxetan-2-ylmethyl)-1H-thieno [2,3-d]imidazole-5-carboxylate (23b) NC s iCr) S CO2Me F
23b [0507] 21f (200 mg, 0.49mino1) and 20b (120 mg, 0.49mino1), Pd(dppf)C12=DCM (40 mg, 0.04 mmol) and potassium carbonate (140 mg, 1.01 mmol) were added to a mixed solution of 1,4-dioxane (5 inL) and water (1 inL), and the reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 100 C for 5 hours. The reaction liquid was cooled to room temperature, then water (5 inL) was added, and extracted with ethyl acetate (10 inL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated to dryness, and the residue was separated and purified by silica gel column chromatography to obtain 23b (143 mg, yield 50%).
[0508] LCMS m/z = 587.0 [M+1]+.
Step 2: (S)-2-(4-(64(4-cy anobenzyl)oxy)pyridin-2-y1)-2,5-difluorob enzy1)-1-(oxetan-2-ylinethyl)-1H-th ieno [2,3-d] im idazole-5-carboxyl ic acid (Compound 23) NC
/

F
Compound 23 [0509] 23b (50 mg, 0.08 mmol) and 1 g (34 mg, 0.24 mmol) were added to a mixed solution of acetonitrile (5 inL) and water (1 inL), and stirred at room temperature for 16 hours. Then the reaction liquid was adjusted to pH=6 with 1N hydrochloric acid, continuously stirred until a white solid precipitated, and filtered and the solid was washed with water. The solid was dissolved in dichloromethane, dried over anhydrous sodium sulfate and filtered. The organic phase was concentrated to dryness, the residue was separated and purified by silica gel column chromatography to obtain compound 23 (15 mg, yield 30%).

1105101 LCMS m/z = 573.1 [M+1]+.
[0511] 111 NMR (400 MHz, DMSO-d6) 6 7.92-7.83 (m, 411), 7.73-7.64 (m, 311), 7.53-7.47 (m, 1H), 7.37-7.29 (m, 1H), 7.02-6.96 (m, 1H), 5.57 (s, 2H), 5.08-5.00 (m, 1H), 4.66-4.59 (m, 111), 4.54-4.45 (m, 211), 4.39-4.29 (m, 311), 2.75-2.64 (m, 111), 2.39-2.29 (m, 111).
Example 24:
2-44-(2-(5-chloropyridin-2-y1)-2-methylbenzo[d] [1,3]dioxo1-4-ylipiperidin-1-ylimethyl)-1-4(S)-oxetan-2-ylimethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid -Th\-OH
Compound 24 Boc Br Boc N.
Bockp-B
I OH Br N - n 0 \
' OH 24c I

24a 24b CI 24e CI
24d ciz4NINIrso 0i) CI
-c?
le 0 N 0 0 CJI4--1 ______ 0 N-IP S
OH
CI 0- Compound 24f 24h Step 1: 2-(4-bromo-2-methylbenzo[d][1,3]dioxo1-2-y1)-5-chloropyridine (24b) Br TrO
24b [0512] 15a (276 mg, 2.0 mmol) and 3-bromocatechol (378 mg, 2.0 mmol) were dissolved in tetrahydrofuran (6 mL), and Ru3(C0)12 (128 mg, 0.2 minol) was added to the system. The reaction flask was subjected to nitrogen replacement for protection, and the reaction liquid in the reaction flask was stirred at 110 C for 10 hours. The reaction liquid was cooled to room temperature, then concentrated under reduced pressure and purified by silica gel column chromatography to obtain lb as a gray oily liquid (190 mg, yield 30%).

1105131 LCMS m/z = 326.0 [M+1]+.
Step 2: tert-butyl 4-(2-(5-chloropyridin-2-y1)-2-methylbenzo[d][1,31dioxo1-4-y1)-3,6-dihydropyridine-1(2H)-carboxylate (24d) Boc <0 CI N
[0514] lb (190 mg, 0.58 mmol), lc (180 mg, 0Ø58 mmol), cesium carbonate (945 mg, 2.9 mmol) and Pd(dppf)C12.DCM (47 mg, 0.058 trunol) were dissolved in a mixed solvent of 1,4-dioxane (15 inL) and water (3 mL), subjected to nitrogen replacement and then heated to 90 C for 3 hours. The reaction liquid was cooled to room temperature.
Water (30 inL) was added, extracted with ethyl acetate (30 inL x 3). The organic phase was combined and washed with saturated brine (30 inL x 1), and the obtained organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and purified by silica gel column chromatography to obtain ld (180 mg, yield 72%) as a colorless oily liquid.
[0515] LCMS tn/z = 373.1 [M-55]+.
Step 3: tert-butyl 44245 -chloropyridin-2-y1)-2-methylbenzo [d]
[1,3] dioxo1-4-y1) piperidine- 1-c arboxylate (24e) Boc CI N
[0516] ld (170 mg, 0.4 mmol) was dissolved in a mixed solvent of tetrahydrofuran (3 inL) and methanol (3 inL), and Raney-Ni (20 mg) was added. The reaction was carried out at room temperature for 10 h after hydrogen replacement for protection. The reaction liquid was filtered, the filter cake (20 inL x 3) was washed with tetrahydrofuran, the filtrate was concentrated under reduced pressure, and purified by silica gel column chromatography to obtain 24e as a colorless oily liquid (120 mg, yield 70%).

1105171 LCMS m/z = 431.1 [M+1].
Step 4: 5-chloro-2-(2-methyl-4-(piperidin-4-yObenzo[d] [1,3] di oxo1-2-yl)pyri dine p-toluenesulfonate (240 =pTSA N

CI N
[0518] 24e (120 mg, 0.28 mmol) was dissolved in ethyl acetate (5 inL), and p-toluenesulfonic acid monohydrate (80 mg, 0.42 minol) was added. The reaction was carried out at 50 C for 10 hours after hydrogen replacement for protection.
The reaction liquid was cooled to room temperature and then concentrated under reduced pressure to obtain the p-toluenesulfonate salt of crude product 24f, which was directly used in the next reaction without further purification.
[0519] LCMS in/z = 33L2 [M+1] .
Step 5: methyl 2-((4-(2-(5-chloropyridin-2-y1)-2-methylbenzo [d] [1,3] dioxo1-4-yl)piperidin-1-yl)methyl)-1-(((S)-oxetan-2-y1)methyl)-1H-thieno [2,3 -d]
imidazole-5 -carboxylate (24h) ci NCN
0 Ns 24h [0520] The crude product 24f obtained in the previous step was dissolved in acetonitrile (3 mL), then potassium carbonate (60 mg, 0.42 inmol) and intermediate le (42 mg, 0.14 mmol) were added. The reaction was carried out at 60 C for 6 hours after the reaction flask was subjected to nitrogen replacement for protection. The reaction liquid was cooled to room temperature, then concentrated under reduced pressure and purified by silica gel column chromatography to obtain 24h (80 mg, yield 96%) as a light yellow solid.
[0521] LCMS in/z = 595.2 [M+1] .

Step 6: 2-((4-(2-(5-chloropyridin-2-y1)-2-methylbenzo [d]
[1,3] di oxo1-4-yl)piperi din-1-yOmethyl)-1-(((S)-oxetan-2-yOmethyl)-1H-thieno [2,3-dlimidazole-5-carboxylic acid (Compound 24) N 'er\

S
OH
Compound 24 [0522] Compound 24h (80 mg, 0.134 mmol) and lithium hydroxide (56 ing, 1.34 ininol) were added to a mixed solvent of acetonitrile (3 inL) and water ((16 inL), and stirred at 50 C for 6 hours. 1N hydrochloric acid was used to adjust pH = 6, then water (15 inL) was added, extracted with ethyl acetate (20 inL x 3), washed with saturated brine (20 inL x 1). The organic phase was dried over anhydrous sodium sulfate and filtered. The solvent was removed from the filtrate under reduced pressure, and the residue was purified by column chromatography (dichloromethane/methano1=15:1) to obtain the compound 24 of interest (53 ing, yield 70%).
[0523] LCMS in/z = 58L2 [M+Hr.
[0524] 11-1 NMR (400 MHz, CD30D) 8 8.59 (s, 1H), 7.88 (dd, 1H), 7.73 ¨ 7.60 (in, 2H), 6.84 ¨ 6.76 (in, 1H), 6.72 (d, 2H), 5.24 ¨ 5.15 (in, 1H), 4.71 ¨ 4.60 (in, 2H), 4.60 ¨
4.51 (in, 1H), 4.47 ¨ 4.38 (in, 1H), 4.21 (s, 2H), 3.45 ¨ 3.32 (in, 2H), 2.90 ¨ 2.66 (in, 4H), 2.55 ¨ 2.42 (in, 1H), 2.01 (s, 3H), 1.99 ¨ 1.89 (in, 4H).
Biological test examples [0525] 1. HEK293/CRE-luc/GLP-1R cell viability [0526] Cell: HEK293/CRE-luc/GLP-1R
[0527] Cell culture medium: DMEM + 10% FBS + 400 ILighnl G418 +
100 vg/m1 Hygromycin B
[0528] Frozen stock solution: 90% FBS, 10% (V/V) DMSO
[0529] Detection buffer: DMEM + 1% FBS
[0530] Experimental procedure [0531] Cells were cultured in DMEM medium + 10% FBS + 400 Kg/m' G418 +

Hygromycin B in a CO2 incubator at 37 C, and passaged once every 3-4 days.

1105321 Cell plating: Trypsinization adjusted the cell density to 1.67x105 cells/mL; 60 tiL of cells per well (10000 cells/well) was inoculated in a 384-well plate containing compounds; A NC well (negative control) and a background well (no cells) were set.
Incubation was performed for about 18 2 h in the incubator.
[0533] Compounds were serially diluted with the detection buffer, and the detection concentration was 0.01 nM-1000 nM.
[0534] The cell culture plate was removed and all supernatant was aspirated from the cells. The cells were washed gently 2 times with 1X PBS.
[0535] The diluted compound was added to a 384-well plate (10 [iL/well), and three replicate wells were set for each concentration. 10 lit. of the detection buffer was added to the NC well, sealed and incubated at 37 C for 6 hours.
[0536] The plate was removed, the cells were allowed to equilibrate to room temperature (at least 15 min), and then all supernatant was aspirated from the cells.
[0537] Steady-Glo Reagent was added to the sample well at 10 L/well and incubated at room temperature for 5 min to lyse the cells.
[0538] The test results were read with a BMG microplate reader.
[0539] Data processing [0540] The mean background value was calculated.
[0541] The induction factor (Fold of induction, Fl) = (induction well value -background value) / (negative control well value - background value) was calculated.
[0542] The EC50 value of the sample was calculated using Graphpad Prim 8.0 software using four-parameter fitting analysis.
[0543] Statistic analysis [0544] Mean and standard error (Mean SEM) were calculated for all results and Graphpad Prism software was used for statistical analysis. Specific data were presented in the form of graphs. P<0.05 was considered to be statistically different.
[0545] Biological test results:
Compounds EC50 (nM) Trifluoroacetate salt of compound 1 A
Trifluoroacetate salt of compound 2 A
Trifluoroacetate salt of compound 3 A

Compound 4 A
Compound 5 A
Compound 5-2 A
Compound 6 A
Compound 7 A
Compound 8 A
Compound 9 A
Compound 10 A
Compound 11 A
Compound 12 A
Compound 13 A
Compound 15 A
Compound 16 A
Compound 17 A
Compound 18 A
Compound 20 A
Compound 21 A
Compound 22 A
Compound 23 A
Compound 24 A
[0546] A < 10 nM, 10 nM < B <50 nM, 50nM < C < 100 nM
[0547] Conclusion: The compound of the present invention has good agonistic effect on GLP-1 receptor, and has a EC50 value less than 100 nM, for example the EC50 values of the trifluoroacetate salt of compound 1, the trifluoroacetate salt of compound 2, the trifluoroacetate salt of compound 3, Compound 4, Compound 5, Compound 5-2, Compound 6, Compound 7, Compound 8, Compound 9, Compound 10, Compound 11, Compound 12, Compound 13, Compound 15, Compound 16, Compound 17, Compound 18, Compound 20, Compound 21, Compound 22, Compound 23, and Compound 24 were less than 10 nM.
[0548] 2. Effects of the compounds on hERG potassium channels [0549] Experimental platform: electrophysiological manual patch-clamp system 1105501 Cell line: Chinese hamster ovary (CHO) cells stably expressing hERG
potassium ion channel [0551] Experimental method: For CHO (Chinese Hamster Ovary) cells stably expressing hERG potassium channel, whole cell patch-clamp technique was used to recorded hERG potassium channel current at room temperature in this experiment. The glass microelectrode is made of a glass electrode blank (BF150-86-10, Sutter) by a puller.
The tip resistance after filling the liquid in the electrode is about 2-5 MEI
The glass microelectrode can be connected to the patch-clamp amplifier by inserting the glass microelectrode into an amplifier probe. The clamping voltage and data recording are controlled and recorded by the pClamp 10 software through a computer. The sampling frequency is 10 kHz, and the filtering frequency is 2 kHz. After the whole cell records were obtained, the cells were clamped at -80 mV, and the step voltage that induced the hERG potassium current (/ hERG) was depolarized from -80 mV to +20 mV for 2 s, then repolarized to -50 mV, and returned to -80 mV after 1 s. This voltage stimulation was given every 10 s, and the administration process was started after the hERG
potassium current was confirmed to be stable (at least 1 minute). The compound was administered for at least 1 minute at each test concentration, and at least 2 cells (n>2) were tested at each concentration.
[0552] Data processing:
[0553] Data analysis processing was carried out by using pClamp 10, GraphPad Prism 5 and Excel software. The inhibition degree of hERG potassium current (peak value of hERG tail current induced at -50 mV) at different compound concentrations was calculated by the following formula: Inhibition % = [1 ¨ (///o)]x100%.
[0554] Among the formula, Inhibition % represents the percentage of inhibition of hERG potassium current by the compound, and / and /o represent the amplitude of hERG
potassium current after and before dosing, respectively.
[0555] Compound IC50 was calculated using GraphPad Prism 5 software by fitting according to the following equation:
[0556] Y = Bottom + (Top-Bottom)/(1+10^((LogIC50-X) x HillSlope)) [0557] Among the equation, X represents the Log value of the tested concentration of the test sample, Y represents the inhibition percentage at the corresponding concentration, and Bottom and Top represent the minimum and maximum inhibition percentage, respectively.
[0558] Biological test results Compounds Structure ICso (1.11\4) O-1) NC
Compound 1 33.6 ON N¨ ):1 S
OH
N ,N
Compound 2 C 52.2 /s r 0 OH
NC
Compound 5-2 o > 40 OH

NC
Compound 6 >40 F
OH
Compound 6 [0559] Conclusion: The compounds of the present invention have less hERG
inhibitory effect.
[0560] 3. Stability test in mouse liver microsomes [0561] At 37 C, 1 ILEA/1 of the test compound was incubated with mouse liver microsomes (0.5 mg/mL) supplemented with NADPH regeneration system for 5, 10, 20, 30 and 60 minutes, and then LC-MS/MS method was used to detect the concentration of the test compound in the generated sample. The half-life (Ti/2) and inherent clearance rate (CLint(mic)) of the compound in mouse liver microsome solution were obtained by calculating at the corresponding time points and the remaining percentage of the compound.
[0562] Biological test results:

T1/2 CLint(mio Compounds Structure (min) (Ill./min/mg) Compound 2 NC

0.977 \S
OH
NC OrI
Compound 5-2 / 0 N \\ 5588 0.252 F 0, N 0 OH
NC
Compound 6 I 296 4.69 OH
Compound 6 Compound DZ-6 (See WO

NC os F1)J.N 130 10.6 ON
for Compound 84) [0563] Conclusion: The compound of the present invention has good stability in mouse liver microsomes. For example, compared with the control compound, the clearance rate of compound 6 in mouse liver microsomes was significantly slowed down, and the half-life was significantly prolonged. The Tin ratio of compound 6 to compound DZ-6 was greater than 2.
[0564] 4. Stability test in monkey liver microsomes [0565] At 37 C, 1 iuM of the test compound was incubated with monkey liver microsomes (0.5 mg/mL) supplemented with NADPH regeneration system for 5, 15, 30, 45 and 60 minutes, and then LC-MS/MS method was used to detect the concentration of the test compound in the generated sample. The half-life (Ti/2) and inherent clearance rate (CLint(mic)) of the compound in monkey liver microsome solution were obtained by calculating at the corresponding time points and the remaining percentage of the compound.
[0566] Biological test results:

Tin CLint(mic) Compounds Structure (min) (IL/min/mg) o Compound 2 NC 842 1.60 N
- s OH
[0567] Conclusion: The compound of the present invention has good stability in monkey liver microsomes.
[0568] 6. Stability test in canine liver microsomes [0569] At 37 C, 1 ILEM of the test compound was incubated with canine liver microsomes (0.5 mg/mL) supplemented with NADPH regeneration system for 5, 15, 30, 45 and 60 minutes, and then LC-MS/MS method was used to detect the concentration of the test compound in the generated sample. The half-life (Tin) and inherent clearance rate (CLint(mic)) of the compound in canine liver microsome solution were obtained by calculating at the corresponding time points and the remaining percentage of the compound.
[0570] Biological test results:

CLint(mic) Compounds Structure (min) (IL/min/mg) O
Compound 1 NC NMN
6116 0.23 OH
[0571] Conclusion: The compound of the present invention has good stability in canine liver microsomes.
[0572] 7. Stability test in human liver microsomes [0573] At 37 C, 1 tiM of the test compound was incubated with human liver microsomes (0.5 mg/mL) supplemented with NADPH regeneration system for 5, 10, 20, 30 and 60 minutes, and then LC-MS/MS method was used to detect the concentration of the test compound in the generated sample. The half-life (Tin) and inherent clearance rate (CLint(mic)) of the compound in human liver microsome solution were obtained by calculating at the corresponding time points and the remaining percentage of the compound.
[0574] Biological test results:

CLint(mic) Compounds Structure (min) (nUmin/mg) _70 NO
Compound 6 o N 554 2.50 sO
OH
Compound 6 Compound DZ-6 (See WO
1.1) NC 40 F 173 8.01 2020207474 *
N, OH
for Compound 84) [0575] Conclusion: The compound of the present invention has good stability in canine liver microsomes. For example, compared with the control compound, the clearance rate of compound 6 in monkey liver microsomes was significantly slowed down, and the half-life was significantly prolonged. The Tin ratio of compound 6 to compound DZ-6 was greater than 3.
[0576] 8. Activity of the compounds on PDE10A1 [0577] In this experiment, the effect of the compounds on the inhibition of activity was detected by the method of fluorescence polarization. PDE10A1 kit was used for detection (Biomol, BPS-60385). According to the kit instruction, FAM-Cyclic-3',5'-AMP (200 nM) dilution was added to wells other than the blank control well(Blank) at 12.5 l/well; PDE test buffer was added to the blank control well (Blank) at 22.5 [LI/well;
PDE test buffer was added to the Substrate control well at 10 p1/well; a compound working solution was added to the corresponding well of the test plate at 2.5 I/well;
PDE test buffer (10% DMSO) was added to the positive control well (Positive control), the Substrate control well, and the blank control (Blank) well at 2.5 [LI/well, respectively;
PDE10A1 enzyme was diluted to 1 pg/ 1 with PDE test buffer, and the enzyme solution was added to the Substrate control well and the positive control well (Positive control) at 10 l/well; the final concentration of the test compound was 300 [LM-15.24 nM, and the final concentration of the positive reference compound TAK063 was 100 nM-0.00508 nM;
incubation was performed at room temperature for 1 hour; the Binding Agent was diluted in a ratio of 1: 100 with Binding Agent Diluent (cAMP) for later use; after the incubation, the Binding Agent dilution was added to the test plate at 50 ill/well, and incubated at room temperature for 20 minutes with slow shaking; after the incubation, Envision was used for FP detection (Excitation 480 nm, Emission 535 nm); Data were normalized to %
inhibition and IC50 values were calculated using a Graphpad 4 parameter logistic equation.
[0578] Conclusion: The compounds of the present invention have a small inhibitory effect on PDE10A1, for example, for the inhibitory effect of Compound 2 on PDE10A1, IC50 (pM) > 500.
[0579] 8. Pharmacokinetic test Evaluation of pharmacokinetics in mice [0580] Male C57 mice (purchased from Hunan SJA Laboratory Animal Co., Ltd, license number: SCXK (Xiang) 2019-0004), 20-25g, were fasted overnight. On the day of the experiment, 18 male C57 mice were divided into 2 groups, with 9 mice in each group.
The test compound was administered intravenously and intragastrically at doses of 5 mg/kg and 20 mg/kg respectively. Before administration and 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after administration, 0.1 ml of blood was collected from the orbital venous plexus, placed in an EDTAK2 centrifuge tube, and centrifuged at 6000 rpm at 4 C
for 5 min, and then the plasma was collected. The plasma sample was added to an acetonitrile solution containing an internal standard (dexamethasone, 500 ng/mL), vortexed and centrifuged at 10,000 rpm for 10 min, and the supernatant was taken for LC-MS/MS
analysis. Pharmacokinetic parameters were calculated using the non-compartmental model in WinNonlin Version 8.0 (Pharsight, Mountain View, CA) pharmacokinetic software.
[0581] Conclusion: The compounds of the present invention have good oral performance, for example, the bioavailability of compound 6 in C57 mice is >25%.
Evaluation of pharmacokinetics in cynomolgus monkeys [0582] Male cynomolgus monkeys (raised in Suzhou LEOLAB Bio Tech. Co.,Ltd.), weighing 3-5 kg, were fasted overnight (14-18 h), 3-5 kg, were fasted overnight. On the day of the experiment, 6 male cynomolgus monkeys were divided into 2 groups, with 3 mice in each group. The test compound was administered intravenously and intragastrically at doses of 2 mg/kg and 10 mg/kg respectively. Before administration and min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h 10 h, 12 hand 24 h after administration, 1 ml of blood was collected via the limb vein, placed in an EDTAK2 centrifuge tube, and centrifuged at 6000 rpm at 4 C for 5 min, and then the plasma was collected.
The plasma 5 sample was added to an acetonitrile solution containing an internal standard (dexamethasone, 500 ng/mL), vortexed and centrifuged at 10,000 rpm for 10 min, and the supernatant was taken for LC-MS/MS analysis. Pharmacokinetic parameters were calculated using the non-compartmental model in WinNonlin Version 8.0 (Pharsight, Mountain View, CA) pharmacokinetic software. Conclusion: The compounds of the present invention have good metabolic properties. For example, compound 6 has a low clearance rate and a long half-life in monkeys, which has the advantages of longer drug action time and lower frequency of administration.
[0583] 9. Cytochrome P450 isoenzyme inhibition test [0584] Experimental objective: The inhibitory effect of the test compound on the activity of human liver microsomal cytochrome P450 isoenzyme (CYP1A2, CYP2C19, CYP2D6 and CYP3A4) was determined.
[0585] Experimental operation: The test compound at an incubation concentration of 0, 0.03, 0.1, 0.3, 1, 3, 10, 30 and 100 [xM was pre-incubated with the human liver microsome solutions containing 5 enzyme mixed substrates (liver microsome concentration: 0.1 mg/ mL) in a water bath at 37 C for 10 min, respectively, and then a coenzyme factor (NADPH) was added to start the reaction. After incubation in a water bath at 37 C for 10 min, the reaction was terminated by adding glacial acetonitrile solution containing an internal standard, shaken for 1 min, centrifuged at 4 C and 10,000 rpm for 10 min, and the supernatant was taken for LC/MS/MS analysis. The amount of metabolites produced by each CYP enzyme substrate was detected.
[0586] The activity of each CYP isoenzyme was reflected by the production rate of each metabolite. The activity of each isoenzyme in the solvent control incubation system without test substances was set as 100%, and the ratio of the production rate of metabolites in the case of different concentrations of test substances to the production rate of metabolites in the case of a solvent control sample was taken as the percentage of residual activity of each isoenzyme. With the residual activity percentage as the ordinate and the inhibitor concentration as the abscissa, Graphpad prism 5.0 was used to plot the data and calculate the IC50 value of the test substance for each CYP isoenzyme.
[0587] Conclusion: The compounds of the present invention have weak inhibitory activity on cytochrome P450 isoenzymes, for example, for the inhibitory activity of compound 6 on CYP2C19, CYP1A2, CYP2D6 and CYP3A4-M, IC50 > 50 M.

Claims (11)

    - 130 -WHAT IS CLAIMED IS:
    1. A compound or a stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, which compound is selected from a compound represented by general formula (I), wherein, ____________________ L __ /
    N ' COON
    (I) ring B is selected from a 4- to 12-membered heterocyclic ring, Cs_p carbocyclic ring, c6-10 aromatic ring or a 5- to 12-membered heteroaromatic ring; the carbocyclic ring, heterocyclic ring, .aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 RB; the heterocyclyl or heteroaromatic ring contains 1 to 3 heteroatoms selected from 0, S and N;
    ring C is selected from C6-10 carbocyclic ring, a 5- to 10-membered heterocyclic ring, C6-10 aromatic ring or a 5- to 10-membered heteroaromatic ring; the carbocyclic ring, heterocyclic ring, aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 4 Rc; the heterocyclic ring or heteroaromatic ring contains 1 to 5 heteroatoms selected from 0, S and N;
    L is selected from a bond, 0, S, -NRL-, -C(RL)2-, -C(RL)2-C(R02-, -Y-C(R02-, -C(RL)2-Y-, -Y-C(RL)2-C(RL)2-, -C(RL)2-C(RL)2-Y- or -C(RL)2-Y-C(R02-;
    Y is selected from 0, S or -NRL-;
    ring D is selected from a 6- to 10-membered aromatic ring or a 5- to 12-membered heteroaromatic ring; the aromatic ring or heteroaromatic ring is optionally further substituted with 0 to 5 RD; the heteroaromatic ring contains 1 to 5 heteroatoms selected from 0, S and N;
    ring E is selected from a 5-membered heterocyclic ring, and the heterocyclic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NH2, Ci_6 alkyl, Ci_6 alkoxy or C3-6 cycloalkyl; the heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N;
    Ri is selected from H, halogen, OH, -SH, CF3, CN, Ci_6 alkyl, C1-6 alkoxy, -C1-alkylene-Z-00_3 alkylene-Ri a, C0-4 alkylene-Ri a; the alkyl, alkoxy, and alkylene are optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, C1 6 alkyl, halogen-substituted C1 6 alkyl, hydroxy-substituted C1 6 alkyl, cyano-substituted C1-6 alkyl, C1-6 alkoxy, C3-6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl, a 5- to 10-membered heteroaryl; the heterocycloalkyl or heteroaryl contains 1 to 3 heteroatoms selected from 0, S and N;
    Z is selected from a bond, N(Rib), 0 or S;
    Ri a is selected from C1-6 alkyl, C1-6 alkoxy, C3-8 cycloalkyl, a 3- to 8-membered heterocycloalkyl, a 6- to 10-membered aryl or a 5- to 12-membered heteroaryl;
    the cycloalkyl, heterocycloalkyl, aryl or heteroaryl is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, Ci _6 alkyl, C1-6 alkoxy or -N(Rib)2; the alkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, CN, OH, -N(R1b)2, Ci_6 alkyl, C1-6 alkoxy or C3-6 cycloalkyl; the heterocycloalkyl or heteroaryl contains 1 to 3 heteroatoms selected from 0, S
    and N;
    each Rlb is independently selected from H or C1-6alkyl; the alkyl is optionally further substituted with 0 to 3 substituents selected from H, halogen, =0, CN, OH, NH,,, Ci_6 alkyl, C1-6 alkoxy or C3-6 cycloalkyl;
    R2 is selected from H, halogen or Ci_6a1ky1;
    each RL is independently selected from H, halogen, -SH, CN, OH, CF3, NH2, C1-6 alkyl, C1-6 alkoxy or C3-6 cycloalkyl; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy;
    each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, N112, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy;
    alternatively, two RB, two Rc, two RD, Rc and RL, RD and RL or RB and R, each independently form a C3_iocarbocyc1ic ring or a 3- to 10-membered heterocyclic ring together with the atoms attached thereto; the carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-6 alkyl, C3-6 cycloalkyl or C1-6 alkoxy; the heterocyclic ring contains 1 to 3 heteroatoms selected from 0, S and N;
    and r is selected from 0 or 1.
    2. The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to
  1. claim 1, wherein, Ri is selected from H, halogen, OH, -SH, CF3, CN, C1_4 alkyl, C1-4 alkoxy, -CI-alkylene-Z-00-2 alkylene-Ri a or -Co_4 alkylene-Ria; the alkyl, alkoxy or alkylene is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, CN, OH, -N(Rib)2, C1-4 alkyl, halogen-substituted C1-4 alkyl, hydroxy-substituted C1-4 alkyl, cyano-substituted C1-4 alkyl, C1-4 alkoxy, C3-6 cycloalkyl, a 3- to 6 -membered heterocycloalkyl or a 5- to 6-membered heteroaryl; the heterocycloalkyl or heteroaryl contains 1 to 3 heteroatoms selected from 0, S and N;
    Ri a is selected from Ci4 alkyl, Ci4 alkoxy, C3-6 cycloalkyl, a 3- to 6-membered heterocycloalkyl, phenyl or a 5- to 6-membered heteroaryl; the cycloalkyl, heterocycloalkyl, phenyl or heteroaryl is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, C1-4 alkyl, C1-4 alkoxy or -N(Rib)?; the alkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, CN, OH, -N(Rib)2, Ci4 alkyl, C1-4 alkoxy or C3-6 cycloalkyl; the heterocycloalkyl or heteroaryl contains 1 to 3 heteroatoms selected from 0, S
    and N;
    each 1216 is independently selected from H or Ci _4 alkyl; the alkyl is optionally further substituted with 0 to 3 substituents selected from H, halogen, =0, CN, OH, NH2, C1-4 alkyl, C1-4 alkoxy or C3-6 cycloalkyl;
    each RL is independently selected from H, halogen, CN, OH, -SH, CF3, NH2, C1-4 alkyl, C1_4 alkoxy or C3_6 cycloalkyl; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or Ci alkoxy;
    each RB, Rc or RD is independently selected from H, halogen, =0, CN, CF3, OH, -SH, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1 4 alkyl, C3 6 cycloalkyl or Ci 4 alkoxy;
    alternatively, two RB, two Rc, two RD, Rc and RL or RB and R2 each independently form a C3_7 carbocyclic ring or a 4- to 7-membered heterocyclic ring together with the atoms attached thereto; the carbocyclic ring or heterocyclic ring is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy; the heterocyclic ring contains 1 to 3 heteroatoms selected from 0, S and N.
    3.
    The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to
  2. claim 2, wherein, ring B is selected from a benzene ring, a 4- to 8-membered heterocyclyl, C5-8 carbocyclyl or a 5- to 6-membered heteroaromatic ring; the benzene ring, carbocyclyl, heterocyclyl or heteroaromatic ring is optionally further substituted with 0 to 4 RE, the heterocyclic ring contains 1 to 3 heteroatoms selected from 0, S and N;
    ring C is selected from a benzene ring, a 5- to 6-membered monocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6 membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring; the benzene ring or heteroaromatic ring is optionally further substituted with 0 to 4 Rc, the heteroaromatic ring contains 1 to 5 heteroatoms selected from 0, S and N;
    ring D is selected from a benzene ring, a naphthalene ring, a 5- to 6-membered monocyclic heteroaromatic ring, a 5-membered fused 5-membered heteroaromatic ring, a 5-membered fused 6-membered heteroaromatic ring or a 6-membered fused 6-membered heteroaromatic ring; the benzene ring, naphthalene ring or heteroaromatic ring is optionally further substituted with 0 to 4 RD, the heteroaromatic ring contains 1 to 5 heteroatoms selected from 0, S and N;
    ring E is selected from a 5-membered non-aromatic heterocyclic ring or a 5-membered heteroaromatic ring, and the heterocyclic ring or heteroaromatic ring is optionally further substituted with 0 or 1 substituent selected from H, halogen, CN, OH, NH2, Ci 4 alkyl, Ci 4 alkoxy or C3 6 cycloalkyl; the heterocyclic ring or heteroaromatic ring contains 1 to 2 heteroatoms selected front 0, S and N;
    Ri is selected from H, F, CI, OH, CN, CF3, methyl, ethyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, -ethylene-Z-methylene-R a, -ethylene-Z-ethylene-R a, -ethylene-Z-R1 a, -R1 a, -methylene-R1 a, -ethylene-R1 a; the methyl, ethyl, propyl, methoxy, ethoxy, methylene, ethylene are optionally further substituted with 0 to 3 substituents selected from H, F, CI, =0, CN, OH, -N(Rib)2, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to
  3. 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyrimidinyl, pyrazinyl or pyridazinyl;
    Z is selected from 0, S or N(Rib);
    Ri a is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidinyl, piperazinyl, pyrrolyl, pyridyl, pyrazolyl, triazolyl, tetrazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, pyrazinyl or phenyl, the Ria is optionally further substituted with 0 to 3 substituents selected from H, F, OH, CN, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy or -N(R102;
    each Rib is independently selected from H, methyl or ethyl;

    R2 is selected from H, F, methyl or ethyl;
    each RL is independently selected from H, F, CI, CN, CF3, OH, NH2, methyl, ethyl, propyl, CF3, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl or cyclopentyl;
    each RB, Rc or RD is independently selected from H, F, CI, =0, CN, CF3, OH, NH2, methyl, ethyl, propyl, -CH2F, -CHF2, 1 to 3 F-substituted ethyl, hydroxymethyl, hydroxyethyl, cyano-substituted methyl, cyano-substituted ethyl, methoxy, ethoxy, propoxy, isopropoxy, tert-butyloxy, trifluoromethoxy, cyclopropyl, cyclobutyl or cyclopentyl ;
    alternatively, RB and R2 together with the atoms attached thereto form a C3-6 carbocyclic ring or a 4- to 7-membered heterocyclic ring; the carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1_4 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C14 alkoxy; the heterocyclic ring contains 1 to 3 heteroatoms selected from 0, S and N;
    alternatively, Rc and RL together with the atoms attached thereto form a C3-6 carbocyclic ring or a 4- to 7-membered heterocyclic ring; the carbocyclic ring or heterocyclic ring is further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy; the alkyl, cycloalkyl or alkoxy is optionally further substituted with 0 to 4 substituents selected from H, halogen, =0, OH, CN, NH2, C1-4 alkyl, C3-6 cycloalkyl or C1-4 alkoxy; the heterocyclic ring contains 1 to 3 heteroatoms selected from 0, S and N.
  4. 4. The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to claim 3, wherein, L is selected from a bond, 0, S, -NRL-, -CHRL-, -CHRL-CHRL-, -Y-CHRL-CHRL-, -CHRL-CHRL-Y- or -CHRL-Y-CHRL-;
    each RL is independently selected from H, F, methyl, ethyl or propyl;

    Y is selected from 0, S or -NRL-;
    ring B is selected from one of the following substituted or unsubstituted groups:
    cyclohexyl, cyclohexenyl, azacyclohexenyl, piperidinyl, phenyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, which, when substituted, is optionally further substituted with 0 to 4 RB;
    or ring B is selected from one of the following substituted or unsubstituted groups:
    piperazinyl, tetrahydropyrrolyl or 1,4-diazepanyl, which, when substituted, is optionally further substituted with 0 to 4 RB;
    ring C is selected from one of the following substituted or unsubstituted groups: a benzene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a tetrazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 Rc;
    ring D is selected from one of the following substituted or unsubstituted groups: a benzene ring, a naphthalene ring, a pyrrole ring, a pyrazole ring, a pyridine ring, a furan ring, a thiophene ring, an imidazole ring, a thiazole ring, a oxazole ring, an isothiazole ring, an isoxazole ring, a triazole ring, a oxadiazole ring, a thiadiazole ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring or a triazine ring, which, when substituted, is optionally further substituted with 0 to 4 RD;
    N
    when r=1, 555- is selected from --- , --- ) when r=0, s'37 is selected from -h--I /
    N

    , , , , t-^-N
    N
    H 0 Or S
    9 .
  5. 5. The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to claim 4, wherein, L is selected from -Y-CHRL-, -CHRL-Y-, -CHRL-CHRL-, -CHRL-CHRL-Y- or -Y-CHRL-CHRL-;
    Y is selected from 0, S or -NRL-;
    each RL is independently selected from H or methyl;
    ring B is selected from one of the following substituted or unsubstituted groups:

    N AN N
    , , , , , or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB;
    or ring B is selected from one of the following substituted or unsubstituted groups:
    N ,) 1-C/N "µ , ._._..,.,,, j , or , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB;

    or ring B is selected from one of the following substituted or unsubstituted groups:

    N-\ N N a N
    -\ /-LN)c , ,-34.- _.----e....õ N .) I ) I ,, r - -,, i - ii I ,N --The N
    'µz-, -le -N
    , , , , ,\)>,1\1 N-N, or ''', ' , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB;

    .5 Or 0 is selected from Or k N
    , the left side of which is directly connected to ring C, and which, when the 6-membered ring in the spiro ring is substituted, is optionally further substituted with 1, 2 or 3 RB;
    ring C is selected from one of the following substituted or unsubstituted groups: a 1`1=1\A- --N!\Ik ,Cj\J (''N1\1A- 1\14- _4-N N e benzene ring, \ -/ , `=N -N , N-=-/ N=-/ \-/
    , -- N N
    -1 0 , , N , Or N
    , which, when substituted, is optionally further substituted with 1, 2 or 3 Rc, and the left side of which is connected to L;
    ring D is selected from a benzene ring or a pyridine ring, and the benzene ring or pyridine ring is optionally further substituted with 1, 2 or 3 RD;
    15 each RB is independently selected from H, F, =0, OH, CN, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy;
    each Rc is independently selected from H, F, Cl, OH, CN, NT1/, CF3, methyl, ethyl, propyl, cyclopropyl, cyclobutyl, cyclopentyl, methoxy or ethoxy;
    each RD is independently selected from H, F, CI, CN, CF3, CHF2, methyl, ethyl, 20 methoxy, ethoxy, tert-butyloxy or trifluoromethoxy;

    alternatively, Rc and RL together with the atoms attached thereto form a 4- to membered heterocyclic ring; the heterocyclic ring is further substituted with 0, 1, 2, 3 or 4 substituents selected from H, F, methyl or ethyl; the heterocyclic ring contains 1 to 2 heteroatoms selected from 0, S and N.
  6. 6.
    The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to claim 5, wherein, L is selected from -C1120-, and the right side of L is connected to ring C;
    Ri is selected from H, CN, CF3, CHF2, CH2F, -CH2OH, -CH(OH)CH3, methyl, ethyl, methoxy, ethoxy, propoxy, isopropoxy, methoxymethyl, methoxyethyl, ethoxymethyl, isopropoxymethyl, -ethyl ene-Z-methyl en e-R a, -ethyl ene-Z-ethyl ene-R a, -ethyl ene-Z-R a, -Ri a or -methylene-Ri a;
    Z is selected from NH, N(CH3) or 0;
    Ri a is selected from methyl, ethyl, isopropyl, propyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxiranyl, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, pyrrolidinyl, piperidinyl, pyrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, imidazolyl, thiazolyl, thienyl, furyl, oxazolyl, isoxazolyl, 1,2,4-oxadiazolyl, isothiazolyl, pyrimidyl, pyridine or phenyl, the Ri a is optionally further substituted with 0, 1, 2 or 3 substituents selected from H, F, CN, methyl, ethyl, propyl, CF3, methoxy or ethoxy;
    ring C is selected from one of the following substituted or unsubstituted groups:
    or " , which, when substituted, is optionally further substituted with 1. 2 or 3 Rc, and the left side of which is connected to L;

    L
    or is selected from , the right side of which is connected to ring B, and in which the benzene ring is optionally further substituted with 1, 2 or 3 Rc;
    ring D is selected from a benzene ring or pyridine, and the benzene ring or pyridine is optionally further substituted with 1, 2 or 3 RD;

    each RB is independently selected from H, =0, F, CN, CF3, methyl, ethyl or cyclopropyl ;
    each Rc is independently selected from H, F, CI, CN, CF3, methyl, ethyl, methoxy or ethoxy;
    each RD is independently selected from H, F, CI, CN, CF3, CHF2, methyl or ethyl.
  7. 7. The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to claim 6, wherein, R1 is selected from 0 or R2 is selected from H;
    ring B is selected from one of the following substituted or unsubstituted groups:
    r'1%?Cii kNj or )CN , the left side of which is directly connected to ring C, and which, when substituted, is optionally further substituted with 1, 2 or 3 RB ;

    B r-jj or is selected from N
    , the left side of which is directly connected to ring C, and which, when the 6-membered ring in the spiro ring is substituted, is optionally further substituted with 1, 2 or 3 RB ;
    N N N
    N
    r=0, is selected from or
  8. 8. The compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to claim 1, wherein the compound is selected from one of the following structures:

    C)-NC
    NC,- -`/4--)-IN___ t 1. .-1 ,0 N,----J .---'( N*,-IN COON N 0H

    r-- I
    F --. ,----õ- 0 F ,, n 0 m N ') l'i>.____, NC ,, NC _......----õN.---- ...,.1,1õNµ/ s 1;H-.01%1N..,õ N ----< N
    OH
    ---*N,-J,1r OH S -1-r NC
    NC ,----õNõ.---,,,TcNz,N
    --,0 N,,, K
    o0i fsl, N-sc \ OH
    N OH-- )--r S- )--r F ---õ,:,---- 0 I

    0,--F -\ i---S
    _ ,NN,_ N----z,C) NC,. ..--F r-)---\ N--S
    NC, N-- -( \ - O -,--- y 1 ,OyN1' N-- OH
    OH
    0--a 0--;
    . NN NC,,, '-`hl---''"1, N/_____ NC-,. ;r' F
    r- r\,i_o N¨cs OH
    s f'C' OH
    i F --, --,--"-- 0 U
    I
    F------r m F OTh NC ..-- F F 0-N

    -S
    NC
    OH
    ------,---OH
    N N

    0 0 m F C)--A
    ,I, NC F
    =---'''Ci ---1 N.---g N ' ----- 1 0 NC-''' N '\S
    zOH
    S -f% 1 ii - OH I
    F

    C)--C)--cl - ,,,N
    -N -T\ õ,_õ...-JJ, Ci. IN1,1 N4 1 Cl J,,(,..70f.1+.1,õ,..,-õ, OH
    yOH
    F -., ,--- µSM-------N NC., NCõ, \ -__ \V ,0 0.--,r"--f 's' ¨1' 1- I s OH
    - 7) OH F 0 I
    (1; F 0Th F

    NOF ,,--- -,õ,õ-----õN
    N

    N17) S N rOH S" y0 NC
    OH

    I I

    OTh NC,-F F

    -J- -__. N
    NC-r.
    N
    I z L ON1, 5 IV / 1 L S 0 y O
    OH H
    NC F
    N
    Cc- C CI F
    r)-N /

    0 N / \
    0 0 N \
    S

    S
    OH
    OH
    NC Ah F
    C:r) NC F (D--Iglõ
    '1-0 NN 0 NN/
    N \
    0 0 N / \ 0 S 0 S
    OH
    OH
    CI F

    Cr)-,.

    N N
    N I
    0 N \ 0 S 0 S
    OH
    OH

    a F
    Cr- CI F

    r--NN 0 N
    r -N
    0 N N / \
    , 0 0 N N /
    , S
    S
    OH
    OH
    C
    1 0 1Vr::
    CI F
    NC , ,--, 0 r-NN - 1 1 N -----, s-- .,i0F1 OH
    i=-,\ C---J r)-, ¨ rsi NC --INJ' N0,---- ---J N--<.¨I\
    --- - y ON ---,,------ y \
    ,OH \s, liOH
    F S' i-[ F

    I I
    0-iN F 0¨\
    ) NC . F I ) NC r N rµIµ
    õN
    j ,:i. -,0 N N N---/t1 -- J1::: N.
    T I , , s- OH
    T 's o OH
    NCç F
    N
    NC F I ) ----=`r N, 0 N / \ 0 S- j-) S
    OH
    OH
    NNN----\\
    O--\ \¨ci NC ) F N NC ,,...,_-õ, F
    1 T ii ; -- 1-jc r--Nr-C_____, 0 Ns N- / & 0, N -1,,) N /
    ,- ) -T- s-I
    yO
    OH OH
    NN¨

    \ 0 \-- N---K
    NC F ) N ) C r t F
    -0 N ) N
    ST- O
    O
    OH H

    o'->
    1\r''0 \--/
    \---/
    NC _TT F

    1 " N" ---II r ,-------N-r-L .0 N N.) N-n \ 1:307 7N,,r, N-_< 17 yO S ru OH
    OH
    --"-, 7 Om ) NC, F
    NC F
    O N"MIN' ----. 7-- N

    I i N N, 1-1_______ -7 0, Nõ N---{ \
    N /
    r OH
    OH
    \ \
    0 0-\
    -\

    r'NI7Th-IN_ N-Th-crIN\ 7-7)-7 (Z: TN. ) N
    L-77 i 0 N) N-\ 11 -- ---\
    )3 I S ro Q
    S r , O OH H

    0--J\ OTh r_.-NC F N. , N , , 0 N 71%/
    I S
    Tr T 'S }21 F 7' OH
    OH
    1 I j NC F
    0 \
    NC
    r F OTh ) , - N
    ; e N - J N / \
    N-,:< I

    'S--OH
    F OH F
    I I
    --\ o--\\ 0\
    , ) NC NC F
    N, F
    r'IN1-7r rµ=
    I 1 - , 1 r --N---N
    J ) ,, _. , )0õNõ,, J N- Ir 0 N. N- Ti µs--) F7 r O
    OH H

    Ic\02--7 OTh 7/---Nf-N I S :
    NC F
    r"Nrõ)N
    NC ,F

    -OH -7,--I
    o o ---\
    c:>----A
    i N 0 I\10 ..--- F
    T,- ).----- N-Ir'l NC F z n /
    .ik' ON N.,_,....-) N.-------N N-----, \S--(:) S OH
    OH
    I N
    ---0- \ 0 OTh ) NC F
    N
    NC _,., F
    )IN
    (N-fIIN
    C
    S
    O
    OH H

    0- \ OTh i NC F F NCN .õõ-,---N
    1\1 r.- r 1,1õN
    , \
    i 1 , UC,0 Niss _L N N ' OH

    0 --\ 0¨\ ) ) NCF
    N ^ N
    NC.,,,,.<:¨.. F
    ( 1::: _N. N-----ftl 'ft NõON ,N ,,y,,N N- -<ftftl --- ft -..------ ft----s r-0 OH i S `r-ID
    OH

    0--\
    0¨\
    ) ) NC F
    1\1 )-ir ,_,Thi,N,, NC .7 N F ,Nõ.___,N

    T. r --ft, II,NOftN
    IO N--2 N---- \ ,s 0 O OH H
    ( Op N r)-NCN,-,, õ...,._____ õT\ N NC

    I y 0 N õ,., N N---c,s-ly_O T---- ----- -1- -, s `rICI

    cl F
    (r\ 0 I NC ,A NI ------,,,,,, N F
    N
    ON, ---.)- N N---..c \ NI
    I
    ' S- 'CO01-I 1 F

    CI n F CI ON
    F r --> I
    N

    N \

    OH

    F F...... 0 r_QO
    )--- 0 F
    jN :

    N

    N

    NI 'e n CI (=r; \ ' CI =
    , --0 F d --, N--// \ :) \ F
    N \
    S
    OH OH
    CI CI
    Cra) , _ N ,, f N
    I

    S S
    OH OH
    CI
    (VD-- 0--NC
    _ _ N N . 0 I / I N \ NI----II )----- OH

    \

    S S-OH

    CN
    0' 7 N _-- 0 /___N_l____S)-- OH F 0-----NC , -.
    c ) T N \
    F N OH

    Oq) r]
    0 ) F
    NC ,,---.N.----,,TcN,)___s L: L O. N. ..----õõ ,--1 N----- ,C:1 NI, N---k\ I
    1, - -... ./..z..-- ----F
    OH F ,--F -OH
    1:1. 10--F F
    NC j-.,---.,--N, I - N
    NC
    L 1", 0 N , N--=<
    .------1,- ---- ---1- --..- ,-- [-.0õ Nõ ---=-- NA i SS yO ,10 F F-- 1 S r OH F''' OH
    F
    F

    NC .N NC.-----,õ
    \I /
    : 1 0 N NA \
    , -,.../
    S \s F OH
    OH
    F

    --- NC F CI-;
    NC N N
    ,Co ,O N.
    - N----_< I 0 N N\---?-1I
    \S- 'r / I ,.

    F
    OH
    _71 F
    NC u----\ NC F

    F
    /
    N

    C) N,-- , - NI---4 ON, N I
    ' I \
    S CO2H li S- 'CO2H
    F F
    F
    F
    NC F CF:a NC
    F
    CrD-N N

    I S CO2H l' S CO2H
    F F
    F

    NC _,--F 0a, ) CI
    I gi---,õ.õ,--;=--=,õi -Ly--,,,N
    I\17CI- N/
    CO2H 0 s 0 F
    OH
    CI

    g-N N
    N-j-- 0 N / 1\l' '7-- 0 N
    0 N \ 0 N \

    S S
    OH
    OH
    CI
    (:)-- CI ----g-I I
    1\- 0 N
    N v- / ...._;___ S S
    OH OH
    N N - \
    CI CFI -3) \=() N NC-),-- F
    /=_, -N

    r OH OH
  9. 9. A pharmaceutical composition, comprising the compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to any one of claims 1 to 8, and a pharmaceutically acceptable carrier.
  10. 10. Use of the compound or the stereoisomer, tautomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof according to any one of claims 1 to 8 or the pharmaceutical composition according to claim 9 in the preparation of a drug for treating diabetes.
  11. 11. A compound, comprising the following structure:

    Cr)-a X N
    N

    Rm wherein X is selected from H or Br;
    Rill is selected from H or C14 alkyl; the alkyl is optionally further substituted with 0 to 4 substituents selected from halogen, C1-4 alkyl, C1-4 alkoxy or C6-10 carbocyclic ring.
CA3203276A 2020-12-25 2021-12-24 Five-membered ring derivative and medical use thereof Pending CA3203276A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202011557297.3 2020-12-25
CN202011557297 2020-12-25
CN202110109531.4 2021-01-27
CN202110403365.9 2021-04-16
CN202111000175.9 2021-09-02
PCT/CN2021/141243 WO2022135572A1 (en) 2020-12-25 2021-12-24 Five-membered ring derivative and medical use thereof

Publications (1)

Publication Number Publication Date
CA3203276A1 true CA3203276A1 (en) 2022-06-30

Family

ID=87143084

Family Applications (1)

Application Number Title Priority Date Filing Date
CA3203276A Pending CA3203276A1 (en) 2020-12-25 2021-12-24 Five-membered ring derivative and medical use thereof

Country Status (1)

Country Link
CA (1) CA3203276A1 (en)

Similar Documents

Publication Publication Date Title
EP4269399A1 (en) Five-membered ring derivative and medical use thereof
JP5916700B2 (en) Amide compounds, compositions and uses thereof
CA2938294C (en) Substituted [1,2,4]triazolo[1,5-a]pyrimidin-7-yl compounds as pde2 inhibitors
AU2007292155B2 (en) Imidazole derivative
CN109810041B (en) Halogenated allylamine SSAO/VAP-1 inhibitor and application thereof
CN114591296A (en) GLP-1R agonist and application thereof in medicine
WO2011076744A1 (en) Disubstituted heteroaryl-fused pyridines
RU2684635C1 (en) Tetrahydroimidazopyridine derivatives as tnf activity modulators
JP2021535164A (en) Myocardial sarcomere inhibitor
WO2011142359A1 (en) Spiro compound and drug for activating adiponectin receptor
CA2983826A1 (en) Azabenzimidazoles and their use as ampa receptor modulators
US20190152992A1 (en) Benzodioxane derivatives and their pharmaceutical use
KR20240005838A (en) Phosphonyl derivatives and compositions thereof and pharmaceutical applications
AU2016214802B2 (en) Diaza-benzofluoranthrene compounds
EP2632924A1 (en) Diaza-spiro[5.5]undecanes useful as orexin receptor antagonists
CA3203276A1 (en) Five-membered ring derivative and medical use thereof
EP3782986A1 (en) Heterocyclic compound
TW202333721A (en) Certain octahydrofuro[3,4-b]pyrazines as glp-1 receptor modulators
TW202302602A (en) Fused-ring heterocycle derivative and medical application thereof
EP4015039A1 (en) Heterocyclic derivatives as p2x7 receptor antagonists
CA3219984A1 (en) Glp-1 receptor agonist and composition and use thereof
CA3206871A1 (en) Pyrimidine compound as wee-1 inhibitor
TW202346294A (en) Pyrazinone derivative and use thereof in medicine
AU2022289869A1 (en) Anticancer compounds
CN116075505A (en) Opioid receptor agonist, preparation method and application thereof