OXAAZAQUINAZOLINE-7(8H)-KETONE COMPOUND, PREPARATION METHOD THEREFOR AND PHARMACEUTICAL APPLICATION THEREOF

Information

  • Patent Application
  • 20220251109
  • Publication Number
    20220251109
  • Date Filed
    April 28, 2020
    4 years ago
  • Date Published
    August 11, 2022
    a year ago
Abstract
An oxaazaquinazolin-7(8H)-ketone compound with a selective inhibition effect on KRAS gene mutation and pharmaceutically acceptable salts thereof, stereoisomers, solvent compounds or prodrugs (as shown in formula I or formula II, see the details of the definition to each group in the formulas in the specification), as well as the pharmaceutical composition containing the compound, and the application thereof in preparation of cancer medicine.
Description
TECHNICAL FIELD

The present invention relates to the technical field of medicine, in particular, to a oxaazaquinazolin-7 (8H)-one compound, and its use as a selective inhibitor of KRAS gene mutation as well as a pharmaceutical composition prepared therefrom.


BACKGROUND OF THE INVENTION

Lung cancer is the cancer with the highest incidence in the world. The incidence of lung cancer ranks first among all cancers in China. It is also the cancer with the highest incidence and mortality in China. According to data released by the American Cancer Society in 2016, approximately 1.8 million people suffer from lung cancer, and nearly 80% of lung cancers are non-small cell lung cancer (NSCLC).


RAS is a group of closely related monomeric globular proteins (21 kDa molecular weight), which have 188-189 amino acids and bind to guanosine diphosphate GDP or guanosine triphosphate GTP. Members of the RAS subfamily include HRAS, KRAS and NRAS. RAS acts as a molecular switch, and when RAS contains bound GDP, it is in a dormant or closed position and is “inactive”. When cells are exposed to certain growth-promoting stimuli, RAS is induced to convert the bound GDP into GTP. When combined with GTP, RAS is “on” and can interact with other downstream target proteins and activate these proteins. RAS protein itself has a very low inherent ability to hydrolyze GTP and restore it to GDP (thus turning itself into a closed state). The exogenous protein GTPase Activated Protein (GAP) is required to restore it to the closed state. The interaction between GAP and RAS greatly accelerates the conversion of GTP to GDP.


Any mutation in RAS will affect the interaction between RAS and GAP and the ability of the conversion of GTP to GDP. Such mutation will lead to prolonged protein activation time, thereby prolonging cell signaling, which in turn will cause cells to continue to grow and divide. Since such signaling causes cell growth and division, over-activated RAS signaling can eventually lead to cancer.


Among lung cancers, mutations in the RAS genes are confirmed in about 32% of lung cancers. Any one mutation in the three main subtypes of the RAS (HRAS, NRAS, or KRAS) genes can lead to the occurrence of human tumors. It has been reported that the KRAS gene has the highest mutation frequency in the RAS genes, and KRAS mutations are detected in 25-30% of tumors. In comparison, the rates of oncogenic mutations in NRAS and HRAS family members are much lower (8% and 3%, respectively). The most common KRAS mutations are found in residues G12 and G13 as well as residue Q61 in the P loop. The G12C mutation is a frequent mutation of the KRAS gene (mutation of glycine-12 to cysteine). This mutation has been found in about 13% of cancers, about 43% of lung cancers, and almost 100% of MYH-related polyposis (familial colon cancer syndrome).


Therefore, it is a better direction to develop inhibitors that selectively inhibit KRAS mutations. In order to increase the inhibitory activity against KRAS mutations while reducing the inhibitory activity against wild-type KRAS, it has great significance to develop novel selective inhibitors of RAS mutants with higher activity, better selectivity, and lower toxicity.


SUMMARY OF THE INVENTION

The present invention provides a oxaazaquinazolin-7 (8H)-one compound, as a selective inhibitor of KRAS mutation, which is advantageous for its high activity, high selectivity and low toxic/side effect and the like.


In one respect, the present invention provides a oxaazaquinazolin-7 (8H)-one compound, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, the compound has a structure as represented by formula (I):




embedded image


wherein,


R1, R2 are each independently hydrogen, cyano, C1-3 alkyl, or —C1-3 alkyl-NRaRb;


R01, R02, R03, R04, R05, R06 are each independently hydrogen, C1-6 alkyl, —C1-4 alkyl-hydroxy, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, or —C1-4 alkyl-halo C1-6 alkoxy;


or R01, R02 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R03, R04 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R05, R06 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


L is a bond, (CRL1RL2)n, C(O), C(O)C(RL1RL2), or C(RL1RL2)C(O); wherein RL1, RL2 are each independently hydrogen, halo, or C1-6 alkyl;


n is 1 or 2;


X1 is NRx1, O, or CRx2Rx3; wherein Rx1 is hydrogen, or C1-6 alkyl; Rx2, Rx3 are each independently hydrogen, halo, cyano, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, NRgRh, —C1-4 alkyl-hydroxy, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, or —C1-4 alkyl-halo C1-6 alkoxy;


X2 is N, or CRx4; wherein Rx4 is hydrogen, halo, cyano, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, NRgRh, —C1-4 alkyl-hydroxy, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, or —C1-4 alkyl-halo C1-6 alkoxy;


Ra is hydrogen, halo, cyano, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, Nine, C2-4 alkenyl, C2-4 alkynyl, —C1-4 alkyl-hydroxy, alkyl-cyano, alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, or —C1-4 alkyl-halo C1-6 alkoxy;


Rb is C6-10 aryl, or C5-10 heteroaryl; the C6-10 aryl, C5-10 heteroaryl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S1, the substituents of the group S1 are halo, cyano, nitro, hydroxy, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, NRiRj, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-4 alkyl-hydroxy, —C1-4 alkyl-cyano, alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, —C1-4 alkyl-halo C1-6 alkoxy, —C1-4 alkyl-C3-6 monocyclic heterocyclyl, —C1-4 alkyl-NReRf, —C1-4 alkyl-C(O)NReRf, —C1-4 alkyl-SO2C1-3 alkyl, or C2-4 alkynyl;


Rc is C1-6 alkyl, C6-10 aryl, C5-10 heteroaryl, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, 7- to 11-membered spirocycloalkyl, alkyl-C6-10 aryl, —C1-4 alkyl-C5-10 heteroaryl, —NRe—C6-10 aryl, —O—C6-10 aryl, —C1-4 alkyl-C3-6 monocyclic heterocyclyl, —C1-4 alkyl-C3-6 monocyclic cycloalkyl; wherein


the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione;


the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, azetidin-2-one, oxetane, oxetan-2-one, oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 1,2-dihydroazacyclobutadiene, 1,2-dihydrooxetadiene, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one;


the —C1-4 alkyl- is unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from C1-3 alkyl;


the C1-6 alkyl, C6-10 aryl, C5-10 heteroaryl, 7- to 11-membered spirocycloalkyl, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S2, the substituents of the group S2 are halo, cyano, hydroxy, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, NRiRj, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-4 alkyl-hydroxy, —C1-4 alkyl-C2-4 alkynyl, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, —C1-4 alkyl-halo C1-6 alkoxy, alkyl-C3-6 monocyclic heterocyclyl, alkyl-C3-6 monocyclic cycloalkyl, —C1-4 alkyl-NReRf, —C1-4 alkyl-C(O)NReRf, —C1-4 alkyl-SO2C1-3 alkyl, or C2-4 alkynyl; wherein the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran; and the C1-6 alkyl, C1-6 alkoxy, —C1-4 alkyl-, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl in the substituents of the group S2 are optionally substituted by 1, 2, or 3 substituent(s) independently selected from halo, methyl, ethyl, propyl, isopropyl, trifluoromethyl, amino, N(CH3)2, hydroxy, carboxyl; wherein the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran;


Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently hydrogen, or C1-3 alkyl;


Ri, Rj are each independently hydrogen, C1-3 alkyl, —C(O)C1-3 alkyl, —CO2C1-3 alkyl.


In one embodiment of the present invention, the compound of formula (I) has a structure as shown in formula (IA) or formula (IB):




embedded image


wherein each group is as defined above.


In another aspect, the present invention provides an oxaazaquinazolin-7 (8H)-one compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, the compound has a structure as represented by formula (II):




embedded image


wherein Rb′ is C6-10 aryl, C5-10 heteroaryl, C3-6 monocyclic heterocyclyl, pyrimidinonyl, or pyridonyl; the C6-10 aryl, C5-10 heteroaryl, C3-6 monocyclic heterocyclyl, pyrimidinonyl, and pyridonyl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S1, or substituted by 1, 2, 3, or 4 substituent(s) independently selected from deuterated C1-6 alkyl and deuterated C1-6 alkoxy; the substituents of the group S1 are halo, cyano, nitro, hydroxy, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, NRiRj, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-4 alkyl-hydroxy, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, —C1-4 alkyl-halo C1-6 alkoxy, —C1-4 alkyl-C3-6 monocyclic heterocyclyl, alkyl-C(O)NReRf, —C1-4 alkyl-SO2C1-3 alkyl, or C2-4 alkynyl;


Rc′ is C1-6 alkyl, C6-10 aryl, C5-10 heteroaryl, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, 7- to 11-membered spirocycloalkyl, —C1-4 alkyl-C6-10 aryl, —C1-4 alkyl-C5-10 heteroaryl, —NRe—C6-10 aryl, —O—C6-10 aryl, —C1-4 alkyl-C3-6 monocyclic heterocyclyl, —C1-4 alkyl-C3-6 monocyclic cycloalkyl, pyrimidinonyl, or pyridonyl;


wherein,


the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione;


the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, azetidin-2-one, oxetane, oxetan-2-one, oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 1,2-dihydroazacyclobutadiene, 1,2-dihydrooxetadiene, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one;


the —C1-4 alkyl- is unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from C1-3 alkyl;


the C1-6 alkyl, C6-10 aryl, C5-10 heteroaryl, 7- to 11-membered spirocycloalkyl, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, pyrimidinonyl, and pyridonyl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S2, or substituted by 1, 2, 3, or 4 substituent(s) independently selected from deuterated C1-6 alkyl and deuterated C1-6 alkoxy; the substituents of the group S2 are halo, cyano, hydroxy, C1-6 alkyl, C1-6 alkoxy, halo C1-6 alkyl, halo C1-6 alkoxy, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, NRiRj, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-4 alkyl-hydroxy, —C1-4 alkyl-C2-4 alkynyl, —C1-4 alkyl-cyano, —C1-4 alkyl-C1-6 alkoxy, —C1-4 alkyl-halo C1-6 alkyl, —C1-4 alkyl-halo C1-6 alkoxy, alkyl-C3-6 monocyclic heterocyclyl, alkyl-C3-6 monocyclic cycloalkyl, —C1-4 alkyl-NReRf, —C1-4 alkyl-C(O)NReRf, —C1-4 alkyl-SO2C1-3 alkyl, or C2-4 alkynyl; wherein the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran; and the C1-6 alkyl, C1-6 alkoxy, —C1-4 alkyl-, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl in the substituents of the group S2 are optionally substituted by 1, 2, or 3 substituent(s) independently selected from halo, methyl, ethyl, propyl, isopropyl, trifluoromethyl, amino, N(CH3)2, hydroxy, carboxyl; wherein the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran;


Re, Rf are each independently hydrogen, or C1-3 alkyl;


Ri, Rj are each independently hydrogen, C1-3 alkyl, —C(O)C1-3 alkyl, —CO2C1-3 alkyl;


R1, R2, R01, R02, R03, R04, R05, R06, L, X1, X2, Ra are as defined above.


In one embodiment of the present invention, the compound of formula (II) has a structure as shown in formula (IIA) or formula (IIB):




embedded image


wherein each group is as defined above.


In one embodiment of the present invention, pyridonyl in Rb′, is pyridinyl-2 (1H)-one.


In one embodiment of the present invention, pyrimidinonyl in Rb′, is pyrimidinyl-4 (3H)-one.


In one embodiment of the present invention, the substituents of the group S1 are halo, cyano, nitro, hydroxy, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRiRj, —C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-2 alkyl-hydroxy, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, —C1-2 alkyl-halo C1-3 alkoxy, —C1-2 alkyl-C3-6 monocyclic heterocyclyl, —C1-2 alkyl-NReRf, —C1-2 alkyl-C(O)NReRf, —C1-2 alkyl-SO2C1-3 alkyl, or C2-4 alkynyl.


In one embodiment of the present invention, the substituents of the group S1 are halo, cyano, nitro, hydroxy, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRiRj, —C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —CH2-hydroxy, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, —CH2-halo C1-3 alkoxy, —CH2—C3-6 monocyclic heterocyclyl, —CH2—NReRf, —CH2—C(O)NReRf, —CH2—SO2C1-3 alkyl, or C2-4 alkynyl; wherein Ri is hydrogen, C1-3 alkyl, —C(O)CH3, or —CO2CH3; Re, Rf, Rj are each independently hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, the substituents of the group S1 are halo, cyano, nitro, hydroxy, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRiRj, —C(O)NReRf, —CH2-hydroxy, —CH2-cyano; wherein Ri is hydrogen, —C(O)CH3, or —CO2CH3; Re, Rf, Rj are each independently hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, the substituents of the group S2 are halo, cyano, hydroxy, C1-6 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, amino, NHCH3, N(CH3)2, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —C1-2 alkyl-hydroxy, —C1-2 alkyl-ethynyl, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, —C1-2 alkyl-halo C1-3 alkoxy, —C1-2 alkyl-C3-6 monocyclic heterocyclyl, —C1-2 alkyl-C3-6 monocyclic cycloalkyl, —C1-2 alkyl-NReRf, —C1-2 alkyl-C(O)NReRf, —C1-2 alkyl-SO2C1-3 alkyl, or ethynyl; wherein the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran; and C1-6 alkyl, C1-3 alkoxy, —C1-2 alkyl-, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl in the substituents of the group S2 are optionally substituted by 1, 2, or 3 substituent(s) independently selected from halo, methyl, ethyl, propyl, isopropyl, trifluoromethyl, amino, N(CH3)2, hydroxy, carboxyl; wherein the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran.


In one embodiment of the present invention, the substituents of the group S2 are halo, cyano, hydroxy, C1-4 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, amino, NHCH3, N(CH3)2, C(O)NReRf, —SO2C1-3 alkyl, —SO2halo C1-3 alkyl, —SO2NReRf, —CH2-hydroxy, —CH2-ethynyl, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, —CH2-halo C1-3 alkoxy, —CH2—C3-6 monocyclic heterocyclyl, —CH2—C3-6 monocyclic cycloalkyl, —CH2—NReRf, —CH2—C(O)NReRf, —CH2—SO2C1-3 alkyl, or ethynyl; wherein the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran; and the C1-4 alkyl, C1-3 alkoxy, —CH2—, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl in the substituents of the group S2 are optionally substituted by 1, 2, or 3 substituent(s) independently selected from halo, methyl, ethyl, propyl, isopropyl, trifluoromethyl, amino, N(CH3)2, hydroxy, carboxyl; wherein the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran.


In one embodiment of the present invention, the substituents of the group S2 are halo, cyano, hydroxy, C1-4 alkyl, C1-3 alkoxy, halo C1-3 alkyl, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl, amino, NHCH3, N(CH3)2, —CH2-hydroxy, —CH2-ethynyl; wherein the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran; and C1-4 alkyl, C1-3 alkoxy, —CH2—, C3-6 monocyclic cycloalkyl, C3-6 monocyclic heterocyclyl in the substituents of the group S2 are optionally substituted by 1, 2, or 3 substituent(s) independently selected from halo, methyl, ethyl, propyl, isopropyl, trifluoromethyl, amino, N(CH3)2, hydroxy, carboxyl; wherein the C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the C3-6 monocyclic heterocyclyl is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran.


In one embodiment of the present invention, the C3-6 monocyclic cycloalkyl in the substituents of the group S1 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, the C3-6 monocyclic heterocyclyl in the substituents of the group S1 is selected from the group consisting of: aziridine, oxirane, azetidine, azetidin-2-one, oxetane, oxetan-2-one, oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 1,2-dihydroazacyclobutadiene, 1,2-dihydrooxetadiene, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one.


In one embodiment of the present invention, the C3-6 monocyclic cycloalkyl in the substituents of the group S2 is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.


In one embodiment of the present invention, the C3-6 monocyclic heterocyclyl in the substituents of the group S2 is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran.


In one embodiment of the present invention, R1, R2 are each independently hydrogen, cyano, C1-3 alkyl, —CH2NH2, —CH2NHCH3, or —CH2N(CH3)2.


In one embodiment of the present invention, R1 is hydrogen; R2 is hydrogen, cyano, C1-3 alkyl, —CH2NH2, —CH2NHCH3, or —CH2N(CH3)2.


In one embodiment of the present invention, R1, R2 are hydrogen.


In one embodiment of the present invention, R01, R02, R03, R04, R05, R06 are each independently hydrogen, C1-3 alkyl, —C1-2 alkyl-hydroxy, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, —C1-2 alkyl-halo C1-3 alkoxy;


or R01, R02 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R03, R04 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R05, R06 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl.


In one embodiment of the present invention, R01, R02, R03, R04, R05, R06 are each independently hydrogen, C1-3 alkyl, —CH2-hydroxy, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, —CH2-halo C1-3 alkoxy;


or R01, R02 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R03, R04 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R05, R06 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl.


In one embodiment of the present invention, R05, R06 are hydrogen; R01, R02, R03, R04 are each independently hydrogen, C1-3 alkyl, —CH2-hydroxy, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, —CH2-halo C1-3 alkoxy;


or R01, R02 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;


or R03, R04 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl.


In one embodiment of the present invention, R01, R02 together with the carbon atom attached thereto formed C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, R03, R04 together with the carbon atom attached thereto formed C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, R05, R06 together with the carbon atom attached thereto formed C3-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, R02, R04 are each independently hydrogen, C1-3 alkyl, —CH2-hydroxy, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, —CH2-halo C1-3 alkoxy; R01, R03, R05, R06 are hydrogen.


In one embodiment of the present invention, R02, R04 are each independently hydrogen, CH3, —CH2-hydroxy, or —CH2-cyano; R01, R03, R05, R06 are hydrogen.


In one embodiment of the present invention, R02, R04 are each independently hydrogen, or CH3; R01, R03, R05, R06 are hydrogen.


In one embodiment of the present invention, R01, R02, R03, R04, R05, R06 are hydrogen.


In one embodiment of the present invention, L is a bond, or (CRL1RL2)n; wherein RL1, RL2 are each independently hydrogen, halo, or C1-6 alkyl; n is 1 or 2.


In one embodiment of the present invention, L is a bond, or (CRL1RL2)n; wherein RL1, RL2 are each independently hydrogen, halo, or C1-3 alkyl; n is 1 or 2.


In one embodiment of the present invention, L is a bond, or (CH2)n; n is 1 or 2.


In one embodiment of the present invention, L is (CRL1RL2)n; wherein RL1, RL2 are each independently hydrogen, halo, or C1-3 alkyl; n is 1 or 2.


In one embodiment of the present invention, L is CH2 or CH2CH2.


In one embodiment of the present invention, L is CH2.


In one embodiment of the present invention, X1 is NRx1 or O; wherein Rx1 is hydrogen, or C1-6 alkyl.


In one embodiment of the present invention, X1 is NRx1 or O; wherein Rx1 is hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, X1 is O.


In one embodiment of the present invention, L is CH2; X1 is O.


In one embodiment of the present invention, L is CH2CH2; X1 is O.


In one embodiment of the present invention, X2 is N or CRx4; wherein Rx4 is hydrogen, halo, C1-6 alkyl, C1-6 alkoxy, or halo C1-6 alkyl.


In one embodiment of the present invention, X2 is N or CRx4; wherein Rx4 is hydrogen, halo, C1-4 alkyl, C1-4 alkoxy, or halo C1-3 alkyl.


In one embodiment of the present invention, X2 is N.


In one embodiment of the present invention, X2 is CRx4; wherein Rx4 is hydrogen, halo, C1-4 alkyl, C1-4 alkoxy, or halo C1-3 alkyl.


In one embodiment of the present invention, X2 is CRx4; wherein Rx4 is hydrogen, fluorine, chlorine, C1-4 alkyl.


In one embodiment of the present invention, Ra is hydrogen, halo, cyano, C1-4 alkyl, C1-4 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRcRd, C2-4 alkenyl, C2-4 alkynyl, —C1-2 alkyl-hydroxy, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, or —C1-2 alkyl-halo C1-3 alkoxy; wherein Rc, Rd are each independently hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, Ra is hydrogen, halo, cyano, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRcRd, vinyl, ethynyl, —CH2-hydroxy, —CH2-cyano, —CH2—C1-3 alkoxy, —CH2-halo C1-3 alkyl, or —CH2-halo C1-3 alkoxy; wherein Rc, Rd are each independently hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, the C3-6 monocyclic cycloalkyl in Ra is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.


In one embodiment of the present invention, Ra is hydrogen, halo, cyano, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, NRcRd, ethynyl, —CH2-hydroxy, —CH2—C1-3 alkoxy; wherein Rc, Rd are each independently hydrogen, or C1-3 alkyl.


In one embodiment of the present invention, Ra is hydrogen, halo, cyano, C1-3 alkyl, C1-3 alkoxy, halo C1-3 alkyl, amino, NHCH3, N(CH3)2, ethynyl, —CH2-hydroxy, or —CH2—C1-3 alkoxy.


In one embodiment of the present invention, Ra is hydrogen, halo, cyano, or C1-3 alkyl.


In one embodiment of the present invention, Ra is hydrogen, fluorine, chlorine, cyano, methyl, ethyl, propyl, or isopropyl.


In one embodiment of the present invention, the 7- to 11-membered spirocycloalkyl in Rc, Rc′ is a monospirocycloalkyl containing one spiro atom formed by any two monocyclic cycloalkyl rings selected from cyclopropyl ring, cyclobutyl ring, cyclopentyl ring, and cyclohexyl ring.


In one embodiment of the present invention, the C6-10 aryl in Rb, Rc, Rb′, Rc′ are each independently phenyl, naphthyl, a 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl, or a 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic cycloalkyl.


In one embodiment of the present invention, the C5-6 monocyclic heterocyclyl in the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl is selected from the group consisting of: oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one.


In one embodiment of the present invention, the C5-6 monocyclic cycloalkyl in the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, the C6-10 aryl in Rb, Rc, Rb′, Rc′ is phenyl.


In one embodiment of the present invention, the C5-10 heteroaryl in Rb, Rc, Rb′, Rc′ are each independently a 5- or 6-membered monoheteroaryl, a 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl, a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl, a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic heterocyclyl, or a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic cycloalkyl.


In one embodiment of the present invention, when the C5-10 heteroaryl in Rb, Rc, Rb′, Rc′ are a 5- or 6-membered monoheteroaryl, the 5- or 6-membered monoheteroaryl are each independently selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.


In one embodiment of the present invention, when the C5-10 heteroaryl in Rb, Rc, Rb′, Rc′ are a 5- or 6-membered monoheteroaryl, the 5- or 6-membered monoheteroaryl are each independently has a structure selected from the group consisting of:




embedded image


embedded image


In one embodiment of the present invention, the 5- or 6-membered monoheteroaryl in the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.


In one embodiment of the present invention, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.


In one embodiment of the present invention, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic heterocyclyl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine;


the C5-6 monocyclic heterocyclyl is selected from the group consisting of: oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one.


In one embodiment of the present invention, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic cycloalkyl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine;


the C5-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl has a structure as shown in formula (a) or formula (b):




embedded image


wherein, C ring is a 5- or 6-membered monoheteroaryl; wherein the 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.


In one embodiment of the present invention, the C ring has a structure selected from the group consisting of:




embedded image


embedded image


wherein the attached two carbon atoms represented by “custom-character” are a pair of adjacent carbon atoms shared as fused to another ring.


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: benzoxazole, benzisoxazole, benzimidazole, benzothiazole, benzisothiazole, benzotriazole, benzofuran, benzothiophene, indole, indazole, isoindole, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline.


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: benzo[d]isoxazole, 1H-indole, isoindole, 1H-benzo[d]imidazole, benzo[d]isothiazole, 1H-benzo[d][1,2,3]triazole, benzo[d]oxazole, benzo[d]thiazole, indazole, benzofuran, benzo[b]thiophene, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline.


In one embodiment of the present invention, the 8- to 10-membered biheteroaryl has a structure as shown in formula (d) or formula (e):




embedded image


Wherein, D ring, E ring are a 5- or 6-membered monoheteroaryl; wherein the 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.


In one embodiment of the present invention, the D ring, E ring have a structure each independently selected from the group consisting of:




embedded image


wherein the attached two carbon atoms represented by “custom-character” are a pair of adjacent carbon atoms shared as fused to another ring.


In one embodiment of the present invention, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: pyridopyrimidine and naphthyridine.


In one embodiment of the present invention, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: pyrido[3,2-d]pyrimidine, pyrido[2,3-d]pyrimidine, pyrido[3,4-d]pyrimidine, pyrido[4,3-d]pyrimidine, 1,8-naphthyridine, 1,7-naphthyridine, 1,6-naphthyridine, 1,5-naphthyridine.


In one embodiment of the present invention, the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, Rb, Rb′ are independently phenyl, naphthyl, a 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl, a 5- or 6-membered monoheteroaryl, a 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl, a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl; the phenyl, naphthyl, 9- or 10-membered aromatic fused bicyclic ring, 5- or 6-membered monoheteroaryl, 9- or 10-membered biheteroaryl, 8- to 10-membered biheteroaryl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S1.


In one embodiment of the present invention, Rb, Rb′ are independently phenyl, or a 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl; the phenyl, 9- or 10-membered biheteroaryl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S1.


In one embodiment of the present invention, Rb, Rb′ are independently phenyl, or a 9-membered biheteroaryl formed by fusing a phenyl to a 5-membered monoheteroaryl; the phenyl, 9-membered biheteroaryl are unsubstituted or substituted by 1, 2, 3, or 4 substituent(s) independently selected from the group S1.


In one embodiment of the present invention, when Rb, Rb′ are independently phenyl, Rb, Rb′ have a structure selected from the group consisting of:




embedded image


wherein Rs1, Rs2 are each independently selected from the substituent of group S1.


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting




embedded image


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl or the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl has a structure selected from the group consisting of:




embedded image


In one embodiment of the present invention, Rb, Rb′ have a structure independently selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment of the present invention, Rb, Rb′ have a structure each independently selected from the group consisting of:




embedded image


embedded image


In one embodiment of the present invention, Rc, Rc′ have a structure independently selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment of the present invention, Rc, Rc′ have a structure each independently selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


In one embodiment of the present invention, the R1, R2, R01, R02, R03, R04, R05, R06, L, X1, X2, Ra, Rb, Rc, Rb′, Rc′ are each independently the corresponding groups in respective specific compounds in the Examples.


In one embodiment of the present invention, the C3-6 cycloalkyl in any group is selected from the group consisting of: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.


In one embodiment of the present invention, the 3- to 6-membered heterocycloalkyl in any group is selected from the group consisting of: aziridine, oxirane, azetidine, oxetane, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran.


In one embodiment of the present invention, In any group the 5- or 6-membered monocycloheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine.


In one embodiment of the present invention, the 8- to 10-membered bicycloheteroaryl in any group is selected from the group consisting of: benzoxazole, benzisoxazole, benzimidazole, benzothiazole, benzisothiazole, benzotriazole, benzofuran, benzothiophene, indole, indazole, isoindole, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline, pyridopyrimidine, naphthyridine.


In one embodiment of the present invention, the compound of formula (I) is selected from the group consisting of respective specific compounds as noted in the Examples, especially, any compound of Z1 to Z14.


In one embodiment of the present invention, the compound of formula (I) is selected from the group consisting of the compounds as prepared in the Examples of the present application.


In one embodiment of the present invention, the compound of formula (I) is selected from the Table A-1.


In one embodiment of the present invention, the compound of formula (I) is selected from the Table A-2.


In one embodiment of the present invention, the representative compounds of formula (I) and formula (II) include the compounds listed in Table A-1 below, or pharmaceutically acceptable salts, stereoisomers, solvents or prodrugs thereof:









TABLE A-1









embedded image







Z1







embedded image







Z2







embedded image







Z3







embedded image







Z4







embedded image







Z5







embedded image







Z6







embedded image







Z7







embedded image







Z8







embedded image







Z9







embedded image







Z10







embedded image







Z11







embedded image







Z12







embedded image







Z13







embedded image







Z14







embedded image







Z15







embedded image







Z16







embedded image







Z17







embedded image







Z18







embedded image







Z19







embedded image







Z20







embedded image







Z21







embedded image







Z22







embedded image







Z23







embedded image







Z24







embedded image







Z25







embedded image







Z26







embedded image







Z27







embedded image







Z28







embedded image







Z29







embedded image







Z30







embedded image







Z31







embedded image







Z32







embedded image







Z33







embedded image







Z34







embedded image







Z35







embedded image







Z36







embedded image







Z37







embedded image







Z38







embedded image







Z39







embedded image







Z40







embedded image







Z41







embedded image







Z42







embedded image







Z43







embedded image







Z44







embedded image









embedded image







Z46







embedded image







Z47







embedded image







Z48







embedded image







Z49







embedded image







Z50







embedded image







Z51







embedded image







Z52







embedded image







Z53







embedded image







Z54







embedded image







Z55







embedded image







Z56







embedded image







Z57







embedded image







Z58







embedded image







Z59







embedded image







Z60







embedded image







Z61







embedded image







Z62







embedded image







Z63







embedded image







Z64







embedded image







Z65







embedded image







Z66







embedded image







Z67







embedded image







Z68







embedded image







Z69







embedded image







Z70







embedded image







Z71







embedded image







Z72







embedded image







Z73







embedded image







Z74







embedded image







Z75







embedded image







Z76







embedded image







Z77







embedded image







Z78







embedded image







Z79







embedded image







Z80







embedded image







Z81







embedded image







Z82







embedded image







Z83







embedded image







Z84







embedded image







Z85







embedded image







Z86







embedded image







Z87







embedded image







Z88







embedded image







Z89







embedded image







Z90







embedded image







Z91







embedded image







Z92







embedded image







Z93







embedded image







Z94







embedded image







Z95







embedded image







Z96







embedded image







Z97







embedded image







Z98







embedded image







Z99







embedded image







Z100







embedded image







Z101







embedded image







Z102







embedded image







Z103







embedded image







Z104







embedded image







Z105







embedded image







Z106







embedded image







Z107







embedded image







Z108







embedded image







Z109







embedded image







Z110







embedded image







Z111







embedded image







Z112







embedded image







Z113







embedded image







Z114







embedded image







Z115







embedded image







Z116







embedded image







Z117







embedded image







Z118







embedded image







Z119







embedded image







Z120







embedded image







Z121







embedded image







Z122







embedded image







Z123







embedded image







Z124







embedded image







Z125







embedded image







Z126







embedded image







Z127







embedded image







Z128







embedded image







Z129







embedded image







Z130







embedded image







Z131







embedded image







Z133







embedded image







Z134







embedded image







Z135







embedded image







Z136







embedded image







Z137







embedded image







Z138







embedded image







Z139







embedded image







Z140







embedded image







Z141







embedded image







Z142







embedded image







Z143







embedded image







Z144







embedded image







Z145







embedded image







Z146







embedded image







Z147







embedded image







Z148







embedded image







Z149







embedded image







Z150







embedded image







Z151







embedded image







Z152







embedded image







Z153







embedded image







Z154







embedded image







Z155







embedded image







Z156







embedded image







Z157







embedded image







Z158







embedded image







Z159







embedded image







Z160







embedded image







Z161







embedded image







Z162







embedded image







Z163







embedded image







Z164







embedded image







Z165







embedded image







Z166







embedded image







Z167







embedded image







Z168







embedded image







Z169







embedded image







Z170







embedded image







Z171







embedded image







Z172







embedded image







Z173







embedded image







Z174







embedded image







Z175







embedded image







Z176







embedded image







Z177







embedded image







Z178







embedded image







Z179







embedded image







Z180







embedded image







Z181







embedded image







Z182







embedded image







Z183







embedded image







Z184







embedded image







Z185







embedded image







Z186







embedded image







Z187







embedded image







Z188







embedded image







Z189







embedded image







Z190







embedded image







Z191







embedded image







Z192







embedded image







Z193







embedded image







Z194







embedded image







Z195







embedded image







Z196







embedded image







Z197







embedded image







Z198







embedded image







Z199







embedded image







Z200







embedded image







Z201







embedded image







Z202







embedded image







Z203







embedded image







Z204







embedded image







Z205







embedded image







Z206







embedded image







Z207







embedded image







Z208







embedded image







Z209







embedded image







Z210







embedded image







Z211







embedded image







Z212







embedded image







Z213







embedded image







Z214







embedded image







Z215







embedded image







Z216







embedded image







Z217







embedded image







Z218







embedded image







Z219







embedded image







Z220







embedded image







Z221







embedded image







Z222







embedded image







Z223







embedded image







Z224







embedded image







Z225







embedded image







Z226







embedded image







Z227







embedded image







Z228







embedded image







Z229







embedded image







Z230







embedded image







Z231







embedded image







Z232







embedded image







Z233







embedded image







Z234







embedded image







Z235







embedded image







Z236







embedded image







Z236







embedded image







Z237









In one embodiment of the present invention, the representative compounds of formula (I) and formula (II) include the compounds listed in Table A-2 below, or pharmaceutically acceptable salts, stereoisomers, solvents or prodrugs thereof:









TABLE A-2









embedded image







Z15-1







embedded image







Z15-2







embedded image







Z17-1







embedded image







Z17-2







embedded image







Z19-1







embedded image







Z19-2







embedded image







Z20-1







embedded image







Z20-2







embedded image







Z20-3







embedded image







Z20-4







embedded image







Z23-1







embedded image







Z23-2







embedded image







Z24-1







embedded image







Z24-2







embedded image







Z25-1







embedded image







Z25-2







embedded image







Z26-2-1







embedded image







Z26-2-2







embedded image







Z29-1







embedded image







Z29-2







embedded image







Z29-3







embedded image







Z29-4







embedded image







Z30-2-1







embedded image







Z30-2-2







embedded image







Z36-1







embedded image







Z36-2







embedded image







Z37-1







embedded image







Z37-2







embedded image







Z45-1







embedded image







Z45-2







embedded image







Z45-3







embedded image







Z45-4







embedded image







Z46-1







embedded image







Z46-2







embedded image







Z47-1







embedded image







Z47-2







embedded image







Z49-1







embedded image







Z49-2







embedded image







Z57-1







embedded image







Z57-2







embedded image







Z73-1







embedded image







Z73-2







embedded image







Z76-1







embedded image







Z76-2







embedded image







Z77-1







embedded image







Z77-2







embedded image







Z82-1







embedded image







Z82-2







embedded image







Z83-1







embedded image







Z83-2







embedded image







Z87-1







embedded image







Z87-2







embedded image







Z88-1







embedded image







Z88-2







embedded image







Z91-1







embedded image







Z91-2







embedded image







Z95-1







embedded image







Z95-2







embedded image







Z96-1







embedded image







Z96-2







embedded image







Z100-1







embedded image







Z100-2







embedded image







Z101-1







embedded image







Z101-2







embedded image







Z102-1







embedded image







Z102-2







embedded image







Z104-1







embedded image







Z104-2







embedded image







Z105-1







embedded image







Z105-2







embedded image







Z107-1







embedded image







Z107-2







embedded image







Z109-1







embedded image







Z109-2







embedded image







Z111-1







embedded image







Z111-2







embedded image







Z112-1







embedded image







Z112-2







embedded image







Z121-1







embedded image







Z121-2







embedded image







Z122-1







embedded image







Z122-2







embedded image







Z125-1







embedded image







Z125-2







embedded image







Z126-1







embedded image







Z126-2







embedded image







Z127-1







embedded image







Z127-2







embedded image







Z128-1







embedded image







Z128-2







embedded image







Z129-1







embedded image







Z129-2







embedded image







Z130-1







embedded image







Z130-2







embedded image







Z131-1







embedded image







Z131-2







embedded image







Z133-1







embedded image







Z133-2







embedded image







Z134-1







embedded image







Z134-2







embedded image







Z137-1







embedded image







Z137-2







embedded image







Z138-1







embedded image







Z138-2







embedded image







Z139-1







embedded image







Z139-2







embedded image







Z143-1







embedded image







Z143-2







embedded image







Z144-1







embedded image







Z144-2







embedded image







Z147-1







embedded image







Z147-2







embedded image







Z148-1







embedded image







Z148-2







embedded image







Z149-1







embedded image







Z149-2







embedded image







Z150-1







embedded image







Z150-2







embedded image







Z151-1







embedded image







Z151-2







embedded image







Z152-1







embedded image







Z152-2







embedded image







Z155-1







embedded image







Z155-2







embedded image







Z156-1







embedded image







Z156-2







embedded image







Z157-1







embedded image







Z157-2







embedded image







Z158-1







embedded image







Z158-2







embedded image







Z159-1







embedded image







Z159-2







embedded image







Z160-1







embedded image







Z160-2







embedded image







Z161-1







embedded image







Z161-2







embedded image







Z162-1







embedded image







Z162-2







embedded image







Z163-1







embedded image







Z163-2







embedded image







Z164-1







embedded image







Z164-2







embedded image







Z168-1







embedded image







Z168-2







embedded image







Z169-1







embedded image







Z169-2







embedded image







Z172-1







embedded image







Z172-2







embedded image







Z173-1







embedded image







Z173-2







embedded image







Z174-1







embedded image







Z174-2







embedded image







Z175-1







embedded image







Z175-2







embedded image







Z176-1







embedded image







Z176-2







embedded image







Z177-1







embedded image







Z177-2







embedded image







Z180-1







embedded image







Z180-2







embedded image







Z181-1







embedded image







Z181-2







embedded image







Z182-1







embedded image







Z182-2







embedded image







Z183-1







embedded image







Z183-2







embedded image







Z184-1







embedded image







Z184-2







embedded image







Z185-1







embedded image







Z185-2







embedded image







Z186-1







embedded image







Z186-2







embedded image







Z199-1







embedded image







Z199-2







embedded image







Z200-1







embedded image







Z200-2







embedded image







Z201-1







embedded image







Z201-2







embedded image







Z205-1







embedded image







Z205-2







embedded image







Z208-1







embedded image







Z208-2







embedded image







Z209-1







embedded image







Z209-2







embedded image







Z210-1







embedded image







Z210-2







embedded image







Z211-1







embedded image







Z211-2







embedded image







Z212-1







embedded image







Z212-2







embedded image







Z213-1







embedded image







Z213-2







embedded image







Z216-1







embedded image







Z216-2







embedded image







Z217-1







embedded image







Z217-2







embedded image







Z218-1







embedded image







Z218-2







embedded image







Z219-1







embedded image







Z219-2







embedded image







Z220-1







embedded image







Z220-2







embedded image







Z221-1







embedded image







Z221-2







embedded image







Z222-1







embedded image







Z222-2







embedded image







Z231-1







embedded image







Z231-2







embedded image







Z232-1







embedded image







Z232-2







embedded image







Z233-1







embedded image







Z233-2







embedded image







Z234-1







embedded image







Z234-2







embedded image







Z235-1







embedded image







Z235-2







embedded image







Z236-1







embedded image







Z236-2







embedded image







Z237-1







embedded image







Z237-2









In another respect, the present invention provides a pharmaceutical composition, comprising the compound as described above or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof; and a pharmaceutically acceptable carrier.


As used herein, the term “pharmaceutically acceptable carrier” means any formulation or carrier medium capable of delivering an effective amount of the active substance of the invention without interfering with the biological activity of the active substance and without causing adverse effects to the host or subject. It is a non-toxic, inert, solid, semi-solid substance or liquid filling agent, diluent, packaging material or auxiliary preparation or any type of excipient. Representative carriers include water, oil, vegetables and minerals, cream base, lotion base, ointment base and the like. These bases include suspension agents, viscosifiers, transdermal promoters and the like. Their formulations are known to those skilled in the field of cosmetic or topical medicine.


In an embodiment of the present invention, the pharmaceutical composition may be administered in any form of oral, spray inhalation, rectal administration, nasal administration, buccal administration, topical administration, parenteral administration, such as, subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal and intracranial injection or infusion, or administered by means of an explanted reservoir. When administered orally, the compound of the present invention may be formulated into any orally acceptable dosage form, including but not limited to tablets, capsules, aqueous solutions or aqueous suspensions. Carriers used in tablets typically include lactose and cornstarch. Lubricants such as magnesium stearate may also be added. Diluents used in capsules typically include lactose and dried cornstarch. Aqueous suspensions are typically formulated by mixing an active ingredient with appropriate emulsifiers and suspension agents. Sweeteners, fragrances or colorants may be added to the oral dosage form as required. When topically administered, especially to the affected surface or organ readily accessible by topical application, such as eye, skin, or lower intestinal neuropathy, the compound of the present invention may be formulated into different topical dosage forms depending on the surface or organs. When topically administered to eyes, the compound of the present invention may be formulated into a dosage form of micronized suspension or solution using an isotonic sterile saline of a certain pH as the carrier, in which preservatives such as benzyl alkoxide chloride may or may not be added. For ocular administration, the compound may be formulated into a form of cream, such as, Vaseline cream. When administered topically to skin, the compound of the present invention may be formulated into a suitable dosage form of ointment, lotion or cream, in which an active ingredient is suspended or dissolved in one or more carriers. The carriers useful in an ointment formulation include but not limited to: mineral oils, liquid vaseline, white vaseline, propylene glycol, polyoxyethylene, polypropylene oxide, emulsified wax and water. The carriers useful in a lotion or cream include but not limited to: mineral oils, sorbitan monostearate, Tween 60, Cetyl ester wax, hexadecenyl aryl alcohol, 2-octyldodecanol, benzyl alcohol and water. The compound of the present invention may be administered in a dosage form of sterile injections, including sterial aqueous injection or oil suspension or sterile injection solution. Useful carriers and solvents include water, Ringer's solution and isotonic sodium chloride solution. Further, sterilized non-volatile oils can also be used as solvents or suspension media, such as monotriglycerides or diglycerides.


In another respect, the present invention provides use of the above oxaazaquinazolin-7 (8H)-one compound, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof in the preparation of a medicament for preventing and/or treating cancer.


In one embodiment of the present invention, the cancer is pancreatic ductal cancer, colorectal cancer, multiple myeloma, lung cancer, skin melanoma, endometrioid carcinoma, uterine carcinosarcoma, thyroid cancer, acute myeloid leukemia, bladder urothelial cancer, stomach cancer, cervical cancer, head and neck squamous cell carcinoma, diffuse large B-cell lymphoma, esophageal cancer, chronic lymphocytic leukemia, lung squamous cell carcinoma, small cell lung cancer, renal papillary cell carcinoma, adenoid cystic carcinoma, chromophobe renal cell carcinoma, liver cancer, breast invasive carcinoma, cervical squamous cell carcinoma, ovarian serous adenocarcinoma, adrenal cortex carcinoma, prostate cancer, neuroblastoma, brain low-grade glioma, glue Plasmoblastoma, medulloblastoma, esophageal squamous cell carcinoma, renal clear cell carcinoma, osteosarcoma, ovarian small cell carcinoma, rhabdoid tumor, sarcoma, small intestinal neuroendocrine tumor, T-cell prolymphocytic leukemia.


In one embodiment of the present invention, the cancer is lung cancer, preferably non-small cell lung cancer.


In another respect, the present invention provides use of the above oxaazaquinazolin-7 (8H)-one compound, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof in the preparation of an inhibitor of KRAS mutation (preferably, the KRAS mutation is KRAS G12C mutation).


In another respect, the present invention provides a method for treating cancer, comprising the step of administering to a patient in need thereof a therapeutically effective amount of compound, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, or any combination thereof, or the above pharmaceutical composition.


As used herein, the term “subject” refers to an aminal, especially a mammal, preferably a human being.


As used herein, the term “effective amount” or “therapeutically effective amount” refers to the sufficient amount of a drug or agent that is non-toxic but has the desired effect. In an embodiment of the present invention, when treating a patient in accordance with the present invention, the amount of a given drug depends on a number of factors, such as the particular dosage regimen, the type of disease or disorder and its severity, and the uniqueness of the subject or the host in need of treatment (e.g., body weight), however, depending on the particular circumstances, including, for example, the particular drug that has been employed, the route of administration, the condition being treated, and the subject or host being treated, the dosage administered can be decided by methods routinely known in the art. Generally, for use in the treatment for an adult, the dosage administered will typically range from 0.02 to 5000 mg/day, for example from about 1 to 1500 mg/day. The desired dose may conveniently be presented as a single dose, or concurrently (or in a short period of time) or in divided doses at appropriate intervals, such as two, three, four or more divided doses per day. It will be understood by those skilled in the art that although the above dosage ranges are given, the specific effective amount can be appropriately adjusted depending on the condition of the patient and in connection with the diagnosis of the physician.


As used therein, the term “pharmaceutically acceptable salts” refers to salts of the compound of the present invention which are pharmaceutically acceptable, and can retain the biological effectiveness of the free base without other side effects. The type of pharmaceutical acceptable salts includes: acid addition salts formed with inorganic acids (such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like) or organic acids (such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, trifluoroacetic acid, formic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, naphthalene sulfonic acid, camphor sulfonic acid, gluconic acid, glutamic acid, hydroxynaphthalamic acid, salicylic acid, stearic acid, muconic acid and the like); or salts formed when an acidic proton present in the parent compound either is replaced by a metal ion such as an alkali metal ion or alkaline earth ion, such as sodium salt, potassium salt, calcium salt and magnesium salt, and the like; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, and the like. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing acid radicals or base radicals by conventional chemical methods. In general, such salts are prepared by the reaction of these compounds in a form of free acid or base with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of the both. In general, non-aqueous media such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile are preferred. In addition to salt forms, the compounds provided herein also exist in prodrug forms. The prodrugs of the compounds described herein are readily chemically altered under physiological conditions to be converted into the compounds of the invention. In addition, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an in vivo environment.


As used herein, the term “solvate” refers to a substance formed by combining the compound of the invention with a pharmaceutically acceptable solvent. Pharmaceutical acceptable solvates include water, ethanol, acetic acid and the like. The solvates include stoichiometric solvates and non-stoichiometric solvates, preferably hydrates. Certain compounds of the present invention may be present in unsolvated or solvated forms, including hydrated forms. In general, solvated forms are equivalent to unsolvated forms and both are included within the scope of the present invention.


As used herein, the compound as represented by formula (I) or formula (II) of the present invention may contain one or more chiral centers and exist in different optically active forms. When the compound contains one chiral center, the compound contain enantiomers. The present invention includes these two isomers and mixtures of isomers, such as racemic mixtures. Enantiomers can be resolved by methods known in the art, such as crystallization and chiral chromatography and other methods. When the compound of formula (I) or formula (II) contains more than one chiral center, diastereomers may exist. The present invention includes resolved optically pure specific isomers and mixtures of diastereomers. Diastereomers can be resolved by methods known in the art, such as crystallization and preparative chromatography. As used herein, the term “stereoisomers” include both conformational and configurational isomers, wherein configurational isomers mainly include cis-trans isomers and optical isomers. The compound of the present invention may be present in a stereoisomeric form, and thereby cover all possible stereoisomeric forms, including but not limited to cis-trans isomers, tautomers, enantiomers, diastereomers, atropisomers, and the like. The compound of the present invention can also be present in forms such as any combination or any mixture of the aforementioned stereoisomers, such as a mixture of mesoisomer, racemate, atropisomer in equal amounts, and the like. For example, a single enantiomer, a single non-enantiomer, or a mixture thereof, or a single atropisomer, or a mixture thereof. Where the compound of the invention contains an olefinic double bond, it includes a cis-isomer and trans-isomer, and any combination thereof, unless otherwise specified. The atropisomers of the present invention are stereoisomers with axial or planar chirality based on the restriction of intramolecular rotation. The compound of the present invention have two atropisomers derived from axial asymmetry, which is produced by restricting the rotation of bond when the substituents Rb or Rc, Rb′ or Rc′ are cyclic groups such as C6-10 aryl, a 5- or 6-membered monocycloheteroaryl, a 8- to 10-membered bicyclo heteroaryl or pyridonyl (especially when the adjacent positions of two ends of the bond have substituents at the ortho positions) connected to the rings such as substituted naphthalidone, and forming a steric hindrance. Regarding the atropisomer of the present invention, wherein the compound has a structure of formula (I) or formula (II), or the compound of formula (I) or formula (II) has an isomer produced by asymmetric carbon, and the like, it represents any one of a pair of atropisomers present in each isomeric compound. And as a medicine, an atropisomer with excellent activity is preferred. The compound of formula (I) or formula (II) has optical isomers derived from asymmetric carbon, axial asymmetry, and the like, if necessary, a single isomer can be obtained by methods known in the art, such as crystallization or chromatography (e.g., chiral chromatography) and other methods. The atropisomers of the compound of the present invention can be expressed in P or M configuration, and can also be labeled in other commonly used ways known in the art.


As mentioned above, the present invention provides compounds shown in the above-mentioned various structures, or tautomers, cis-trans isomers, mesoisomers, racemates, enantiomers, diastereomers, atropisomers thereof, or the form of a mixture thereof, wherein “the form of a mixture thereof” includes any of the aforementioned stereoisomers (e.g., tautomers, cis-trans isomers, enantiomers, diastereomers, atropisomers) and/or mixtures (mesoisomers, racemates) in any form, such as mixtures of cis-trans isomers, mixtures of enantiomers and diastereomers, mixtures of diastereomers, mixtures of atropisomers, or mixtures of cis-trans isomers and racemates, mixtures of enantiomers and diastereomers, mixtures of atropisomers and diastereomers mixtures, and the like.


As used herein, the term “alkyl” refers to a liner or branched aliphatic hydrocarbon group having 1 to 20 carbon atoms. The term “C1-10 alkyl” refers to a liner or branched alkyl group having 1 to 10 carbon atoms, more preferably 1, 2, 3, 4, 5 or 6 carbon atoms, i.e., C1-6 alkyl, more preferably, C1-4 alkyl, the most preferably, C1-3 alkyl. Specific examples include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl and various branched isomers thereof.


As used herein, the term “alkoxy” refers to a group having a structure of —O-alkyl, wherein the alkyl is as defined above. The term “C1-10 alkoxy” refers to an alkoxy group having 1 to 10 carbon atoms, preferably, C1-6 alkoxy, more preferably, C1-4 alkoxy, the most preferably, C1-3 alkoxy. Specific examples include, but are not limited to, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy and the like.


As used herein, the term “alkenyl” refers to an alkyl as defined above having one or more carbon-carbon double bond at any position of the chain. The term “C2-8 alkenyl” refers to an alkenyl group having 2 to 8 carbon atoms and at least one carbon-carbon double bond, preferably, an alkenyl group having 2 to 6 carbon atoms and 1 to 2 carbon-carbon double bond, i.e., C2-6 alkenyl, more preferably, an alkenyl group having 2 to 4 carbon atoms and 1 to 2 carbon-carbon double bond, i.e., C2-4 alkenyl. Specific examples include, but are not limited to, vinyl, 1-propenyl, 2-propenyl, 1-, 2- or 3-butenyl, pentenyl, hexenyl, butadienyl, and the like.


As used herein, the term “alkynyl” refers to an alkyl as defined above having one or more carbon-carbon triple bond at any position of the chain. The term “C2-8 alkynyl” refers to an alkynyl group having 2 to 8 carbon atoms and at least one carbon-carbon triple bond, preferably, an alkynyl group having 2 to 6 carbon atoms and 1 to 2 carbon-carbon triple bond, i.e., C2-6 alkynyl, more preferably, an alkynyl group having 2 to 4 carbon atoms and 1 to 2 carbon-carbon triple bond, i.e., C2-4 alkynyl. Specific examples include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 1-, 2- or 3-butynyl, and the like.


As used herein, the term “halogen” refers to fluoro, chloro, bromo and iodine.


As used herein, the term “haloalkyl” refers to an alkyl as defined above which is substituted by one or more (1, 2, 3, 4 or 5) halogens. The term “halo C1-10 alkyl” refers to a haloalkyl having 1 to 10 carbon atoms, preferably, halo C1-6 alkyl, more preferably, halo C1-4 alkyl, most preferably, halo C1-3 alkyl. Specific examples include, but are not limited to, chloromethyl, dichloromethyl, trichloromethyl, chloroethyl, 1,2-dichloroethyl, trichloroethyl, bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, and the like.


As used herein, the term “haloalkoxy” refers to an alkoxy as defined above which is substituted by one or more (1, 2, 3, 4 or 5) halogens. The term “halo C1-10 alkoxy” refers to a haloalkoxy having 1 to 10 carbon atoms, preferably, halo C1-6 alkoxy, more preferably, halo C1-4 alkoxy, most preferably, halo C1-3 alkoxy. Specific examples include, but are not limited to, trifluoromethoxy, trifluoroethoxy, fluoromethoxy, fluoroethoxy, difluoromethoxy, difluoroethoxy, and the like.


As used herein, the term “deuterated alkyl” refers to an alkyl group substituted with one or more (e.g., 1, 2, 3, 4, or 5) deuterium atoms, wherein the definition of the alkyl group is as described above. The term “deuterated C1-10 alkyl” refers to a deuterated alkyl having 1 to 10 carbon atoms. It is preferably a deuterated C1-6 alkyl, more preferably a deuterated C1-4 alkyl, and more preferably a deuterated C1-3 alkyl. Specific examples include, but are not limited to, mono-deuterated methyl, di-deuterated methyl, tri-deuterated methyl, mono-deuterated ethyl, 1,2-di-deuterated ethyl, tri-deuterated ethyl, and the like.


As used herein, the term “deuterated alkoxy” refers to an alkoxy group substituted with one or more (e.g., 1, 2, 3, 4, or 5) deuterium atoms, wherein the alkoxy group is as defined above. The term “deuterated C1-10 alkoxy” refers to a deuterated alkoxy group having 1 to 10 carbon atoms. It is preferably a deuterated C1-6 alkoxy, more preferably a deuterated C1-4 alkoxy, and more preferably a deuterated C1-3 alkoxy. Specific examples include, but are not limited to, tri-deuterated methoxy, tri-deuterated ethoxy, mono-deuterated methoxy, mono-deuterated ethoxy, di-deuterated methoxy, di-deuterated ethoxy, and the like.


As used herein, the terms “cycloalkyl” and “cycloalkyl ring” refer to saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbyl substituents. The cycloalkyl ring contains 3 to 20 carbon atoms (C3-20), preferably contains 3 to 12 carbon atoms (C3-12), more preferably contains 3 to 10 carbon atoms (C3-10), most preferably contains 3 to 6 carbon atoms (C3-6). A ring carbon atom in a cycloalkyl may be optionally substituted by 1, 2 or 3 oxo group(s) to form a structure of cyclic ketone. When it is a monocyclic cycloalkyl, the monocyclic cycloalkyl is saturated or partially unsaturated, preferably a monocyclic cycloalkane containing 3 to 8 ring carbon atoms (i.e., 3- to 8-membered or C3-8), more preferably containing 3 to 6 ring carbon atoms. Non-limiting examples of the monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, cyclobutanone, cyclobutan-1,2-dione, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione, and the like; the polycyclic cycloalkyl includes spirocycloalkyl, fused cycloalkyl, and bridged cycloalkyl.


The term “spirocycloalkyl” refers to a polycyclic group containing 5 to 20 ring carbon atoms (i.e., 5- to 20-membered or C5-20), wherein 3- to 8-membered (i.e., containing 3 to 8 ring carbon atoms or C3-8) single rings share one carbon atom (referred to as spiro atom). Each single ring can contain one or more double bonds, but none of the rings has a fully conjugated π-electron system. It is preferably a 6- to 14-membered (i.e., containing 6 to 14 ring carbon atoms or C6-14) spirocycloalkyl, and more preferably a 7- to 11-membered spirocycloalkyl. Depending on the number of the spiro atoms shared between the rings, the spirocycloalkyls are divided into monospirocycloalkyl, bispirocycloalkyl, or polyspirocycloalkyl, preferably a monospirocycloalkyl and a bispirocycloalkyl, more preferably a 7-membered (4-membered monocyclic/4-membered monocyclic), 8-membered (4-membered monocyclic/5-membered monocyclic), 9-membered (4-membered monocyclic/6-membered monocyclic, 5-membered monocyclic/5-membered monocyclic), 10-membered (5-membered monocyclic/6-membered monocyclic) or 11-membered (6-membered monocyclic/6-membered monocyclic) monospirocycloalkyl. Non-limiting examples of spirocycloalkyl include:




embedded image


The cycloalkyl ring may be fused to an aryl ring, a heteroaryl ring or a heterocyclyl ring, wherein the ring attached to the parent structure is the cycloalkyl ring. Non-limiting examples include indanyl, tetralyl, benzocycloheptyl, and the like. In the present invention, each of the above types of cycloalkyl may be optionally substituted, where the substituent(s) are preferably one or more substituents as described in the present disclosure.


As used herein, the term “halocycloalkyl” refers to a cycloalkyl as defined above which is substituted by one or more (1, 2, 3, 4 or 5) halogens. The term “halo C3-8 cycloalkyl” refers to a halocycloalkyl having 3 to 8 carbon atoms, preferably, halo C3-6 cycloalkyl, more preferably, halo C3, halo C4, halo C5, or halo C6 cycloalkyl. Specific examples include, but are not limited to, trifluorocyclopropyl, fluorocyclopropyl, fluorocyclohexyl, difluorocyclopropyl, difluorocyclohexyl, and the like.


As used herein, the term “heterocyclyl” and “heterocyclyl ring” are used exchangeably to refer to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbyl, containing 3 to 20 ring atoms (i.e., 3- to 20-membered or C3-20), wherein one or more (preferably 1 to 4) ring atoms are heteroatoms selected from the group consisting of nitrogen, oxygen or S(O)m, (wherein m is an integer from 0 to 2), but not contain a cyclic moiety of —O—O—, —O—S— or —S—S—, and the remaining ring atoms are each carbon. The nitrogen atom may be substituted or unsubstituted (i.e., N or NR, R is hydrogen, or other substituents as defined herein). The ring carbon atoms of the heterocyclyl may be optionally substituted by 1, 2 or 3 oxo group(s) to form a structure of cyclic ketone, cyclic lactone or cyclic lactam. It preferably contains 3 to 12 ring atoms, more preferably 3 to 10 ring atoms, wherein 1 to 4 are heteroatoms.


In certain embodiments of the present invention, “heterocyclyl” refers to a monocyclic heterocyclyl, which is saturated or partially unsaturated, and preferably a monocyclic heterocyclyl containing 3 to 8 ring atoms (i.e., 3- to 8-membered or C3-8), wherein 1 to 3 are heteroatoms, more preferably a monocyclic heterocyclyl group containing 3 to 6 ring atoms, wherein 1 to 2 are heteroatoms, the most preferably a monocyclic heterocyclyl group containing 5 to 6 ring atoms, wherein 1 to 2 are heteroatoms. If the heteroatom is a nitrogen atom, the nitrogen atom may be substituted or unsubstituted (i.e., N or NR, R is hydrogen or other substituents as defined herein). If the heteroatom is a sulfur atom, the sulfur atom may be optionally oxidized (i.e., S(O)m, m is an integer from 0 to 2). The ring carbon atom in the monocyclic heterocyclyl may be optionally substituted by 1, 2 or 3 oxo group(s) to form a structure of cyclic ketone, cyclic lactone or cyclic lactam. Specific examples of the monocyclic heterocyclyl includes, but are not limited to, aziridine, oxirane, azetidine, azetidin-2-one, oxetane, oxetan-2-one, oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 1,2-dihydroazacyclobutadiene, 1,2-dihydrooxetadiene, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one, and the like.


The adjacent two ring atoms in the above monocyclic heterocyclyl, including C—C, N—C, may be optionally fused to the cycloalkyl, heterocyclyl, aryl or heteroaryl as defined herein, such as, monocyclic cycloalkyl ring, monocyclic heterocyclyl ring, monoaryl ring, 5- or 6-membered monoheteroaryl ring and the like, to form a fused polycyclyl. The adjacent two ring atoms in the above monocyclic heterocyclyl fused to another ring is preferably C—C.


In the present invention, each of the above types of heterocyclyl may be optionally substituted. If substituted, the substituent(s) are preferably one or more substituents as described in the present disclosure.


As used herein, the term “aryl” refers to an all-carbon monocyclyl, all-carbon polycyclyl (a ring is linked to another by a covalent bond, non-fused) or all-carbon fused polycyclyl (i.e., a pair of adjacent carbon atoms are shared between the ring) groups containing 6 to 14 ring atoms (i.e., 6- to 14-membered or C6-14), and at least one ring in the ring system is aromatic, that is, has a n electron conjugated system. It is preferably an aryl containing 6 to 10 ring atoms (i.e., 6- to 10-membered or C6-10). Each ring in the ring system contains 5 or 6 ring atoms. The terms “aryl” and “aryl ring” are used interchangeably.


In certain embodiments of the present invention, “aryl” refers to a monoaryl or polyaryl ring, and non-limiting examples thereof include: phenyl, biphenyl, and the like.


In certain embodiments of the present invention, “aryl” refers to an aromatic fused polycyclyl, the aromatic fused polycyclyl maybe a polycyclyl group formed by fusing a monoaryl ring to one or more monoaryl rings. Non-limiting examples include naphthyl, anthryl, and the like.


In certain embodiments of the present invention, “aryl” refers to an aromatic fused polycyclyl (preferably a 9- or 10-membered aromatic fused polycyclyl), the aromatic fused polycyclyl may be a polycyclyl group formed by fusing a monoaryl ring (preferably phenyl) to one or more non-aromatic rings, wherein the ring attached to the parent structure is an aromatic or non-aromatic ring. The non-aromatic ring includes, but is not limited to, a 3- to 6-membered monocyclic heterocyclyl ring, preferably a 5- or 6-membered monocyclic heterocyclyl ring (the ring carbon atom in the monocyclic heterocyclyl may be substituted by 1 or 2 oxo group(s) to form a structure of cyclic lactam or cyclic lactone), a 3- to 6-membered monocyclic cycloalkyl ring, preferably a 5- or 6-membered monocyclic cycloalkyl ring (the ring carbon atom in the monocyclic cycloalkyl may be substituted by 1 or 2 oxo group(s) to form a structure of cyclic ketone), and the like.


The above polycyclyl group formed by fusing a monoaryl ring to one or more non-aromatic rings may be linked to other moiety or the parent structure through a nitrogen atom or carbon atom. The ring attached together to the parent structure is an aromatic or non-aromatic ring, and non-limiting examples include:




embedded image


In the present invention, each of the above types of aryl may be substituted or unsubstituted. If substituted, the substituent(s) are preferably one or more groups as described in the present disclosure.


As used herein, the term “heteroaryl” refers to a monocyclic or fused polycyclic (that is, rings sharing adjacent carbon atoms or pairs of heteroatoms) groups containing 1 to 4 heteroatoms, having 5 to 14 ring atoms (i.e., 5- to 14-membered or C5-14), preferably 5 to 10 ring atoms (i.e., 5- to 10-membered or C5-10), more preferably 5, 6, 8, 9 or 10 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen, wherein the nitrogen and sulfur atoms may be optionally oxidized, and the nitrogen atom may be optionally quaternized. The heteroaryl in the ring system has 6, 10 or 14 π electrons as shared. At least one ring in the ring system is aromatic. The terms “heteroaryl” and “heteroaryl ring” are used interchangeably.


In certain embodiments of the present invention, “heteroaryl” refers to a monoheteroaryl ring (preferably a 5- or 6-membered monoheteroaryl ring), and non-limiting examples of the monoheteroaryl include: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine, and the like.


In certain embodiments of the present invention, “heteroaryl” refers to a fused polyheteroaryl ring (preferably a 8- to 10-membered biheteroaryl ring). The fused polyheteroaryl ring either includes a polycyclic group (preferably a 9- or 10-membered biheteroaryl ring) formed by fusing a monoaryl ring (preferably, phenyl) and a monoheteroaryl ring (preferably a 5- or 6-membered monoheteroaryl ring), or include a polycyclic group (preferably a 8- to 10-membered biheteroaryl ring) formed by fusing a monoheteroaryl (preferably a 5- or 6-membered monoheteroaryl) and a monoheteroaryl (preferably a 5- or 6-membered monoheteroaryl).


Any 2 adjacent ring atoms in the above monoheteroaryl ring, including C—C, N—C, N—N, may be fused to the cycloalkyl, heterocyclyl, aryl or heteroaryl such as the monocyclic cycloalkyl ring, monocyclic heterocyclyl ring, monoaryl ring, 5- or 6-membered monoheteroaryl ring and the like as define in the present disclosure, to form a fused polycyclyl. The 2 adjacent ring atoms in the monoheteroaryl ring, which are fused to another ring to form a fused ring, are preferably C—C, and include in a non-limiting way the forms of:




embedded image


Non-limiting examples of the fused polyheteroaryl ring include: benzo[d]isoxazole, 1H-indole, isoindole, 1H-benzo[d]imidazole, benzo[d]isothiazole, 1H-benzo[d][1,2,3]triazole, benzo[d]oxazole, benzo[d]thiazole, indazole, benzofuran, benzo[b]thiophene, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline, pyrido[3,2-d]pyrimidine, pyrido[2,3-d]pyrimidine, pyrido[3,4-d]pyrimidine, pyrido[4,3-d]pyrimidine, 1,8-naphthyridine, 1,7-naphthyridine, 1,6-naphthyridine, 1,5-naphthyridine, and the like.


The above monoheteroaryl, or polycyclyl group formed by fusing a monoaryl ring and a monoheteroaryl ring, or polycyclyl group formed by fusing a monoheteroaryl and a monoheteroaryl may be linked to other moiety or the parent structure through a nitrogen atom or carbon atom. If it is a polycyclyl group, the ring attached together to the parent structure is a heteroaryl ring, an aryl ring, a monocyclic cycloalkyl ring or a monocyclic heterocyclyl ring, and non-limiting examples thereof include:




embedded image


embedded image


In certain embodiments of the present invention, “heteroaryl” refers to a fused polyheteroaryl ring (preferably a 8- to 10-membered biheteroaryl ring). The fused polyheteroaryl ring is a polycyclyl group formed by fusing a monoheteroaryl ring (preferably a 5- or 6-membered monoheteroaryl ring) and one or more non-aromatic ring(s), wherein the ring attached together to the parent structure is a heteroaryl ring or a non-aromatic ring, the non-aromatic ring includes, but not limited to: a 3- to 6-membered (preferably a 5- or 6-membered) monocyclic heterocyclyl ring (the ring carbon atoms in the monocyclic heterocyclyl ring may be substituted by 1 or 2 oxo group(s) to form a structure of cyclic lactam or cyclic lactone), a 3- to 6-membered (preferably a 5- or 6-membered) monocyclic cycloalkyl ring (the ring carbon atoms in the monocyclic cycloalkyl ring may be substituted by 1 or 2 oxo group(s) to form a structure of cyclic ketone), and the like.


The above polycyclyl group formed by fusing a monoheteroaryl ring and one or more non-aromatic ring(s) may be linked to other moiety or the parent structure through a nitrogen atom or carbon atom, the ring attached together to the parent structure is a heteroaryl ring or a non-aromatic ring, and non-limiting examples thereof include:




embedded image


In the present invention, each of the above types of heteroaryl may be substituted or unsubstituted. When substituted, the substituent(s) are preferably one or more substituents as described in the present disclosure.


As used herein, the term “hydroxyl” refers to —OH.


As used herein, the term “hydroxylmethyl” refers to —CH2OH, and “hydroxyethyl” refers to —CH2CH2OH or —CH(OH)CH3.


As used herein, the term “cyanomethyl” refers to —CH2CN, and “cyanoethyl” refers to —CH2CH2CN or —CHCNCH3.


As used herein, the term “amino” refers to —NH2.


As used herein, the term “cyano” refers to —CN.


As used herein, the term “nitro” refers to —NO2.


As used herein, the term “benzyl” refers to —CH2-phenyl.


As used herein, the term “oxo group” refers to ═O.


As used herein, the term “carboxyl” refers to —C(O)OH.


As used herein, the term “carboxylic ester group” refers to —C(O)O(alkyl) or —C(O)O(cycloalkyl).


As used herein, the term “acetyl” refers to —COCH3.


As used herein, the term “—C1-4 alkyl-C6-10 aryl” refers to C1-4 alkyl is substituted by C6-10 aryl, preferably substituted by one C6-10 aryl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-C5-10 heteroaryl” refers to C1-4 alkyl is substituted by C5-10 heteroaryl, preferably substituted by one C5-10 heteroaryl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-C3-6 monocyclic heterocyclyl” refers to C1-4 alkyl is substituted by C3-6 monocyclic heterocyclyl, preferably substituted by one C3-6 monocyclic heterocyclyl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-C3-6 monocyclic cycloalkyl” refers to C1-4 alkyl is substituted by C3-6 monocyclic cycloalkyl, preferably substituted by one C3-6 monocyclic cycloalkyl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-hydroxy” refers to C1-4 alkyl is substituted by hydroxy, preferably substituted by one hydroxy, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—, and the examples of —C1-4 alkyl-hydroxy include hydroxymethyl, hydroxyethyl.


As used herein, the term “—C1-4 alkyl-cyano” refers to C1-4 alkyl is substituted by cyano, preferably substituted by one cyano, wherein —C1-4 alkyl-represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—, and the examples of —C1-4 alkyl-cyano include cyanomethyl, cyanoethyl.


As used herein, the term “—C1-4 alkyl-C1-6 alkoxy” refers to C1-4 alkyl is substituted by C1-6 alkoxy, preferably substituted by one C1-6 alkoxy, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-halo C1-6 alkyl” refers to C1-4 alkyl is substituted by halo C1-6 alkyl, preferably substituted by one halo C1-6 alkyl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-halo C1-6 alkoxy” refers to C1-4 alkyl is substituted by halo C1-6 alkoxy, preferably substituted by one halo C1-6 alkoxy, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-NReRf” refers to C1-4 alkyl is substituted by NReRf, preferably substituted by one NReRf, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-C(O)NReRf” refers to C1-4 alkyl is substituted by C(O)NReRf, preferably substituted by one C(O)NReRf, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-SO2C1-3 alkyl” refers to C1-4 alkyl is substituted by SO2C1-3 alkyl, preferably substituted by one SO2C1-3 alkyl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “—C1-4 alkyl-C2-4 alkynyl” refers to C1-4 alkyl is substituted by C2-4 alkynyl, preferably substituted by one C2-4 alkynyl, wherein —C1-4 alkyl- represents an alkylene or alkylidene radical formed by substitution, and is preferably —C1-4 alkyl-, more preferably —C1-3 alkyl-, more preferably —C1-2 alkyl-, such as —CH2—CH2—, the most preferably —CH2—.


As used herein, the term “substituted” means that any one or more hydrogen atoms on a particular atom are replaced with substituents, including deuterium and hydrogen variants, as long as the valence of a particular atom is normal and the substituted compound is stable. When the substituent is an oxo group (i.e., ═O), it means that two hydrogen atoms are replaced. Replacement of an oxo group does not occur on aromatic groups. The term “optionally substituted” means that it may or may not be substituted. Unless otherwise specified, the type and number of substituents may be arbitrary on the basis of being chemically achievable.


When any variant (e.g., R) occurs more than once in the constitution or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted by 0-2 R, the group may optionally be substituted with up to two R, and R in each case has an independent option. In addition, combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.


The compound represented by the formula (I) or formula (II) of the present invention may be prepared by using a synthetic method known in the art or using a method known in the art in combination with the method described in the present invention. The solvent, temperature and other reaction conditions provided by the present invention are exemplary and may be changed according to methods well known in the art. The compounds described in the Examples of the present invention may be synthesized by using appropriate starting materials according to the methods described in the Examples based on their specific structures. It may be synthesized using a method similar to that described in the Examples. The starting materials used to synthesize the compounds in the Examples of the present invention may be prepared by known synthetic methods or similar methods described in the literature or obtained from commercial sources. If necessary, the compounds in the Examples may be further resolved to obtain its stereoisomers by methods well known in the art, such as crystallization, chromatography, and the like, and the resolution conditions are easily obtained by those skilled in the art through conventional means or limited experiments.


As a further illustration, the compound of formula ((I-a) of the present invention may be synthesized by the following method, wherein the solvent, temperature and other reaction conditions in each step may be the same as or similar to those described in the following Examples, or the reaction conditions known in the art are used;




text missing or illegible when filed


wherein, Rlev is a well known leaving group in the art, such as trifluoromethanesulfonate; chlorine, bromine, iodine; sulfonate group, such as mesylate, tosylate, p-toluenesulfonate, and the like; acyloxy group, such as acetoxy, trifluoroacetoxy, and the like. Rp is an amino protecting group well known in the art, such as formyl; acyl, such as alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethyloxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1,1-di-(4′-methoxyphenyl)methyl; silyl, such as trimethylsilyl (TMS) and tert-butyldimethylsilyl (TBS), and the like. R1, R2, R01, R02, R03, R04, R05, R06, n, X1, X2, Ra, Rb, Rc are as defined above.


As a further illustration, the compound of formula ((II-a) of the present invention may be synthesized by the following method, wherein the solvent, temperature and other reaction conditions in each step may be the same as or similar to those described in the following Examples, or the reaction conditions known in the art are used;




text missing or illegible when filed


wherein, Rlev is a well known leaving group in the art, such as trifluoromethanesulfonate; chlorine, bromine, iodine; sulfonate group, such as mesylate, tosylate, p-toluenesulfonate, and the like; acyloxy group, such as acetoxy, trifluoroacetoxy, and the like. Rp is an amino protecting group well known in the art, such as formyl; acyl, such as alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethyloxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1,1-di-(4′-methoxyphenyl)methyl; silyl, such as trimethylsilyl (TMS) and tert-butyldimethylsilyl (TBS), and the like. R1, R2, R01, R02, R03, R04, R05, R06, n, X1, X2, Ra, Rb′, Rc′ are as defined above.







DETAILED DESCRIPTION OF THE INVENTION

The compound of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments obtained by combining those listed below with other chemical synthesis methods, and the equivalent alternatives well known to those skilled in the art. Preferred embodiments include, but are not limited to, the Examples of the present invention.


It should be understood that the absolute configuration of the stereoisomer separated in the following Examples is arbitrarily specified, and the separation method may be chromatographic separation, for example, including but not limited to column chromatography, thin layer chromatography, liquid chromatography, and the like.


The present invention is illustrated in details by means of the Examples, but is not meant to be unfavorably limited thereto. In the present disclosure, the present invention has been described in details, wherein specific embodiments thereof are also disclosed. It will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the present invention without departing from the spirit and scope of the present invention. If specific conditions are not indicated in the Examples, it shall be carried out in accordance with conventional conditions or those recommended by the manufacturer. The reagents or instruments without their manufacturers indicated are all conventional products that can be purchased commercially.


The abbreviations of reagents used in the following Examples are as follows: CDI is N,N′-carbonyl diimidazole, DBU is 1,8-diazabicyclo[5.4.0]undec-7-ene, PyBop is 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate, SPhos is 2-dicyclohexylphosphino-2′,6′-dimethoxybiphenyl, SPhos-Pd-G2 is chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl) [2-(2′-amino-1,1′-biphenyl)]palladium(II), LiHMDS is lithium bis(trimethylsilyl)amide, Selectfluor is 1-chloromethyl-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate), XantPhos-Pd-G2 is chloro[(4,5-bis(diphenylphosphino)-9,9-dimethylxanthene)-2-(2′-amino-1,1′-bi phenyl)]palladium(II), xantphos is 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene, Pd(dppf)Cl2 is [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloride, NCS is N-chlorosuccinimide, NaHMDS is sodium bis(trimethylsilyl)amide, T3P is 1-propylphosphonic anhydride, DIEA is N,N-diisopropylethylamine.


Preparation of Intermediate a



embedded image


Step 1: 2-isopropyl-4-methylpyridine-3-amine (50 g, 333.3 mmol) was dissolved in tetrahydrofuran (500 mL) and the reaction solution was cooled to −65° C., then NaHMDS (333.3 mL, 2M, 666.6 mmol) was slowly dropped, and the reaction solution was kept lower than −60° C. during the dropping process. After the completion of the dropping, the reaction proceeded at −60° C. for 30 minutes, then 4-bromo-2,6-difluorobenzonitrile in tetrahydrofuran (500 mL) was slowly dropped to the reaction solution, and the reaction solution was kept lower than −60° C. during the dropping process. After the completion of the dropping, the reaction solution returned to room temperature and the reaction proceeded overnight. After the completion of the reaction, the saturated ammonium chloride solution was added to quench the reaction. The reaction solution was extracted with ethyl acetate, the organic phase was washed with brine, dried with anhydrous sodium sulfate, and concentrated to dryness under reduced pressure, to obtain a crude product of 4-bromo-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzonitrile (133 g, yield: 100%). ES-API: [M+H]+=348.1.


Step 2: 4-bromo-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzonitrile (13 3 g, the crude product, 333.3 mmol) was dissolved in acetonitrile (1400 mL), and NCS (89.1 g, 666.6 mmol) was added. The reaction solution was heated to 80° C. to react for 2 hours, cooled to room temperature, and water was added to quench the reaction. The reaction solution was extracted with ethyl acetate, the organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, concentrated to dryness under reduced pressure, and purified by column to obtain 4-bromo-3-chloro-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzo nitrile (13.91 g, yield of two steps: 11%). ES-API: [M+H]+=382.1.


Step 3: 4-bromo-3-chloro-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzo nitrile (13.91 g, 36.5 mmol) was dissolved in DMSO (130 mL), then potassium carbonate (10.0 g, 73.2 mmol) was added, hydrogen peroxide (14.91 g, 131.41 mmol) was dropped under the ice bath, and the reaction proceeded at room temperature for 2 hours. After the completion of the reaction, 10% sodium sulfite solution (100 mL) was slowly added under the ice bath to quench the reaction. The reaction solution was extracted with ethyl acetate, the organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, and concentrated to dryness under reduced pressure to obtain the product 4-bromo-3-chloro-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (12.5 g, yield: 85.8%). ES-API: [M+H]+=400.1.


Step 4: 4-bromo-3-chloro-2-fluoro-6-((2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (12.5 g, 31.33 mmol) in tetrahydrofuran (150 ml) was dropped in a solution of sodium hydride (12.5 g, 156.66 mmol) in tetrahydrofuran (40 mL) under a dry ice ethanol bath to control the temperature below 0° C., the reaction proceeded for 15 minutes under a dry ice ethanol bath, and then carbonyl diimidazole (7.6 g, 47.1 mmol) in tetrahydrofuran (100 mL) was dropped. After the completion of the dropping, dry ice reacted with ethanol for 15 minutes. After the completion of the reaction, the saturated ammonium chloride solution was added to quench the reaction. The reaction solution was extracted with ethyl acetate, the organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, concentrated to dryness under reduced pressure, and the crude product was slurried with (petroleum ether/ethyl acetate=10:1) to obtain the target product: 7-bromo-6-chloro-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4 (1H,3H)-dione (10.8 g, yield: 81.1%), ES-API: [M+H]+=426.1.


Step 5: tert-butyl (R) 3-(hydroxymethyl)piperazin-1-carboxylate (3.0 g, 14.07 mmol) was dissolved in THF (60 mL) at 0° C., sodium hydride (1.88 g, 46.9 mmol) was added in batches, the reaction was continued at 0° C. for 30 minutes, then 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl) quinazolin-2-one (4.0 g, 9.38 mmol) was added in batches, and the reaction solution reacted at 0° C.˜room temperature for 1.5 hours. After the completion of the reaction, the reaction solution was diluted with ethyl acetate (100 mL) at 0° C., then the ice water was added, the product was extracted with ethyl acetate (3*100 mL), washed with the saturated brine (3*80 mL), dried with anhydrous sodium sulfate, and concentrated to obtain the crude product. The crude product was purified by a silica gel column to obtain tert-butyl (R)-3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (a brown oil, 4.5 g, yield: 77.59%). ES-API: [M+H]+=622, 624.


Step 6: tert-butyl (R)-3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (4.0 g, 6.42 mmol) was dissolved in DMF (100 mL) at 0° C., PyBOP (8.35 g, 16.05 mmol) was added in batches, and then DBU (4.88 g, 32.10 mmol) was dropped. After addition was completed, the reaction solution reacted at room temperature for 2 hours. After the completion of the reaction, the reaction solution was diluted with ethyl acetate (100 mL) at 0° C., then the ice water was added, the product was extracted with ethyl acetate (3*100 mL), washed with the saturated brine (3*100 mL), dried with anhydrous sodium sulfate, and concentrated to obtain the crude product. The crude product was purified by a silica gel column to obtain Intermediate a (a brown oil, 3.6 g, yield: 92.78%). ES-API: [M+H]+=603, 605.


Preparation of Intermediate b



embedded image


Step 1: 6-bromopyridin-2-ol (866 mg, 4.98 mmol) was dissolved in 5 mL acetonitrile, potassium carbonate (2.06 g, 14.93 mmol) and methyl iodide (1.06 g, 7.47 mmol) were added, and stirred at 60° C. for 2 hours. The reaction solution was extracted with ethyl acetate, and concentrated under reduced pressure. The crude product was purified by column chromatography (petroleum ether/ethyl acetate=1/1) to obtain 6-bromo-1-methylpyridin-2 (1H)-one (a white solid, 630 mg, yield: 73%). ES-API: [M+H]+=188.0.


Step 2: 6-bromo-1-methylpyridin-2 (1H)-one (2 g, 10.6 mmol) was dissolved in tetrahydrofuran (20 mL), and cooled to −78° C. under nitrogen protection, n-butyl lithium (5.1 mL, 12.7 mmol, 2.5 M in tetrahydrofuran) was dropped. Under stirring, the reaction proceeded for 15 minutes at −78° C., and then tributyltin chloride (5.19 g, 16.0 mmol) was dropped, and the reaction slowly dried to room temperature. The reaction solution was quenched with the saturated ammonium chloride solution, extracted with ethyl acetate, concentrated under reduced pressure. The crude product was purified by column chromatography (petroleum ether/ethyl acetate=10/1-2/1) to obtain 1-methyl-6-(tributyltinalkyl)pyridin-2 (1H)-one (light yellow liquid, 1.6 g, yield: 38%). ES-API: [M+H]+=400.2.


Preparation of Intermediate c



embedded image


Step 1: 2,4-dichloro-6-methyl-5-nitropyrimidine (20 g, 97.08 mmol) was dissolved in dioxane (400 ml), isopropylalkenyl potassium trifluoroborate (14.98 g, 101.9 mmol), bis(triphenylphosphine)palladium dichloride (3.4 g, 4.85 mmol) were added in sequence, sodium carbonate (20 g) dissolved in 100 mL of water was dropped thereto. After addition was completed, the nitrogen was used for replacement for three times, and the reaction proceeded for 2 hours at 80° C. After the completion of the reaction, the reaction system was cooled to room temperature, added into 500 ml ice water, extracted with ethyl acetate for three times, the organic phase was washed with brine once, dried with anhydrous sodium sulfate, and purified by manual column chromatography (pure petroleum ether) to obtain 2-chloro-4-methyl-5-nitro-6-(prop-1-en-2-yl)pyrimidine (8 g, yield: 38%). ES-API: [M+1]+=214.02.


Step 2: 2-chloro-4-methyl-5-nitro-6-(prop-1-en-2-yl)pyrimidine (8 g, 37.55 mmol) was dissolved in methanol (50 ml), sodium acetate (6.15 g, 75.11 mmol) and 800 mg palladium on carbon were added in sequence. The reaction was connected to hydrogen balloon and the hydrogen was used for replacement for three times. The reaction was stirred at room temperature for 24 hours. The point board was found that the raw materials disappeared, there were products and by-products that had not removed Cl. The reaction solution was filtered, the sample was mixed and passed the column machine (120 g column) to purify (petroleum ether/ethyl acetate=3/1) and obtain 4-isopropyl-6-methylpyrimidin-5-amine (4 g, yield: 71%). ES-API: [M+1]+=152.1.


Example 1: Preparation of Compound Z1



embedded image


embedded image


embedded image


Step 1: methyl 2-amino-4-bromo-6-fluorobenzoate (2.6 g, 10.48 mmol) and 50 mL isopropanol were added to a 250 mL round bottom flask. After the temperature of the system raising to 60° C., N-chlorosuccinimide (1.7 g, 12.7 mmol) was added to the reaction solution. The temperature of the system rised to 80° C. and reacted at this temperature for 20 minutes. After the completion of the reaction, the reaction solution was concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-8%) to obtain the target product: methyl 6-amino-4-bromo-3-chloro-2-fluorobenzoate 1.2 g, yield of 39%. ES-API: [M+H]+=282.0. HNMR (400 MHz, DMSO-d6): 7.05 (s, 1H), 6.87 (s, 2H), 3.82 (s, 3H).


Step 2: methyl 6-amino-4-bromo-3-chloro-2-fluorobenzoate (1.1 g, 3.89 mmol), 1-iodo-2-isopropylbenzene (1.44 g, 5.84 mmol), Pd2(dba)3 (0.36 g, 0.39 mmol), xantphos (0.56 g, 0.97 mmol), cesium carbonate (2.55 g, 7.78 mmol) and 20 mL of dioxane were added to a 100 mL round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction was stirred at 70° C. for 16 hours. The completion of the reaction was detected by LC-MS. 30 mL of water was added to the reaction. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-5%) to obtain the target product: methyl 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzoate 380 mg, yield of 24%. ES-API: [M+H]+=400.1.


Step 3: methyl 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzoate (380 mg, 0.95 mmol), sodium hydroxide (114 mg, 2.84 mmol), methanol (5 mL), tetrahydrofuran (10 mL) and water (5 mL) were added to a 100 mL round bottom flask. The reaction was stirred at room temperature for 3 hours. The completion of the reaction was detected by LC-MS. 30 mL of water was added to the reaction, and the pH of the reaction was adjusted to 7 with 1M aqueous hydrochloric acid. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated to obtain the crude product: 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzoic acid 367 mg. ES-API: [M+H]+=386.1.


Step 4: 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzoic acid (280 mg, 0.72 mmol), thionyl chloride (862 mg, 7.2 mmol) and toluene (5 mL) were added to a 100 mL round bottom flask. The reaction was stirred at 100° C. for 1 hour. The reaction solution was concentrated and dried to obtain a yellow oil. The yellow oil was dissolved in tetrahydrofuran, and then it was dropped to 28% ammonia water (5 mL). The reaction solution was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. 30 mL of water was added to the reaction. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-40%) to obtain the target product: 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzamide (300 mg), purity of 89%, yield of 96%. ES-API: [M+H]+=385.3.


Step 5: 60% NaH (146 mg, 3.65 mmol), tetrahydrofuran (6 mL) and DMF (2 mL) were added to a 100 mL round bottom flask. The reaction was cooled to 0° C., and 4-bromo-3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)benzamide (280 mg, 0.73 mmol) in tetrahydrofuran (2 mL) was dropped thereto. The reaction was stirred at 0° C. for 5 minutes, and then N,N′-carbonyl diimidazole (141 mg, 0.87 mmol) in tetrahydrofuran (1 mL) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 15 minutes. The completion of the reaction was detected by LC-MS. 30 mL of the saturated NH4Cl aqueous solution was added into the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-25%) to obtain the target product: 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylphenyl)quinazolin-2 (1H)-one (150 mg), a yellow solid, yield of 50%. ES-API: [M+H]+=411.2.


Step 6: 60% NaH (24 mg, 0.6 mmol), tert-butyl 3-(hydroxymethyl)piperazin-1-carboxylate (67 mg, 0.31 mmol), tetrahydrofuran (6 mL) were added to a 100 mL round bottom flask. The reaction was cooled to 0° C., and 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylphenyl)quinazolin-2 (1H)-one (100 mg, 0.24 mmol) in tetrahydrofuran (2 mL) was dropped thereto. The reaction was stirred at 0° C. for 5 minutes, and then the reaction was stirred under a 65° C. oil bath for 1 hour. The completion of the reaction was detected by LC-MS. 30 mL of the saturated NH4Cl aqueous solution was added into the reaction solution. The reaction solution was extracted with 40 m ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-5%) to obtain the target product: tert-butyl 3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropylphenyl)-2-oxo-1,2-dihydro quinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (130 mg), a yellow solid, purity of 84%, yield of 75%. ES-API: [M+H]+=607.1.


Step 7: tert-butyl 3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropylphenyl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (100 mg, 0.16 mmol), N,N-dimethylformamide (1 mL) and 50% T3P in ethyl acetate (523 mg, 0.82 mmol) were added to a 50 mL round bottom flask. DBU (146 mg, 0.96 mmol) was added all at once while stirring. The reaction was stirred at room temperature for 16 hours. The completion of the reaction was detected by LC-MS. The reaction was directly purified by a C18 reversed-phase column (acetonitrile/water (1% trifluoroacetic acid): 0-70%) to obtain the target product: tert-butyl 10-bromo-11-chloro-8-(2-isopropylphenyl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazine[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (50 mg), a yellow solid, purity of 90%, yield of 53%. ES-API: [M+H]+=589.1.


Step 8: tert-butyl 10-bromo-11-chloro-8-(2-isopropylphenyl)-7-oxo-3,4,7,8,13,13a-hexahydropyr azine[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (40 mg, 0.068 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (18 mg, 0.1 mmol), SPhos (7 mg, 0.017 mmol), Pd2(dba)3 (12 mg, 0.014 mmol), potassium phosphate (43 mg, 0.2 mmol), 2 mL of dioxane and 0.2 mL of water were added to a 10 mL microwave reaction tube. The microwave reaction tube was placed in a microwave reactor at 115° C. and the reaction proceeded for 1 hour, and the reaction stopped. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the target product: tert-butyl 11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazine[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (40 mg), a yellow solid, purity of 70%, the crude product yield of 64%. ES-API: [M+H]+=641.2.


Step 9: tert-butyl 11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazine[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (40 mg, 0.062 mmol), 2 mL dichloromethane and 2 mL of trifluoroacetic acid were added to a 25 mL round bottom flask. The reaction was stirred at room temperature for 0.5 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated at 40° C. to obtain the crude product: 11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (39 mg), a yellow solid, purity of 72%. ES-API: [M+H]+=541.2.


Step 10: 11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (39 mg, 0.06 mmol), 2 mL dichloromethane and triethylamine (30 mg, 0.3 mmol) were added to a 25 mL round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (5 mg, 0.04 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 20 minutes. 10 mL of the saturated NaHCO3 aqueous solution was added to the reaction solution, extracted with 10 mL dichloromethane for 3 times, the organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: Z1 (4.44 mg), a white solid, the reaction yield of 12%. ES-API: [M+H]+=595.2. 1HNMR (400 MHz, CDCl3): 7.52-7.34 (m, 4H), 7.28-7.26 (m, 2H), 7.11-7.09 (m, 1H), 6.68-6.61 (m, 1H), 6.44-6.40 (m, 1H), 6.21-6.20 (m, 1H), 5.83 (d, J=10 Hz, 1H), 5.05-5.01 (m, 1H), 4.72-4.65 (m, 3H), 4.08-3.98 (m, 2H), 3.62-3.58 (m, 1H), 3.21-3.12 (m, 2H), 2.76-2.71 (m, 1H), 2.12-2.10 (m, 3H), 1.22-1.20 (m, 3H), 1.05-1.01 (m, 3H).


Example 2-14

Compounds Z2 to Z14 were prepared by referring to the similar method of Compound Z1, wherein the starting materials of each Compound can be prepared by commercially available or by referring to existing methods well known to one skilled in the art, and similar synthetic methods of Intermediates are easily available for one skilled in the art by referring existing methods.














Example

MS


No.
Compound Structure
[M + H]+

















2


embedded image


575.2






Z2






3


embedded image


609.2






Z3






4


embedded image


613.2






Z4






5


embedded image


579.2






Z5






6


embedded image


575.2






Z6






7


embedded image


596.2






Z7






8


embedded image


609.2






Z8






9


embedded image


602.1






Z9






10


embedded image


599.2






Z10






11


embedded image


594.2






Z11






12


embedded image


575.2






Z12






13


embedded image


591.2






Z13






14


embedded image


557.2






Z14









Example 15: Preparation of Compound Z15, Z15-1 and Z15-2



embedded image


embedded image


embedded image


Step 1: 2-isopropyl-4-methylpyridin-3-amine (1.5 g, 9.99 mmol) and 50 mL of tetrahydrofuran were added to a 250 mL round bottom flask. After the system cooling to 0° C., sodium bis(trimethylsilyl)amide (12.5 mL, 2.5 M in tetrahydrofuran, 25 mmol) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at −65° C. for 30 minutes, and then 4-bromo-2,6-difluorobenzonitrile (2.39 g, 10.98 mmol) in tetrahydrofuran (50 mL) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. overnight. After the completion of the reaction, the reaction solution was poured into ice water, and extracted with ethyl acetate. The organic phase was washed with 1M hydrochloric acid, the saturated aqueous sodium bicarbonate solution, and the saturated brine. The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated and dried to obtain the product: 4-bromo-2-fluoro-6-(((2-isopropyl-4-methylpyridin-3-yl)amino)benzonitrile (1.7 g, 49%), a yellow solid. ES-API: [M+H]+=349.3.


Step 2: 4-bromo-2-fluoro-6-(((2-isopropyl-4-methylpyridin-3-yl)amino)benzonitrile (1.7 g, 4.88 mmol) and 50 mL of dimethyl sulfoxide were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, potassium carbonate (1.34 g, 9.76 mmol) and hydrogen peroxide (6.57 g, 30% aqueous solution, 58 mmol) were added. The reaction proceeded overnight at this temperature. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 150 mL of ice water, and a solid precipitated out. It was filtered to obtain filter cake. The filter cake was dried to obtain 4-bromo-2-fluoro-6-(((2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (1.8 g, the crude product), a white solid, the crude product was directly used in the next step. ES-API: [M+H]+=367.2.


Step 3: 4-bromo-2-fluoro-6-(((2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (1.8 g, 4.9 mmol) and 80 mL of dry tetrahydrofuran were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, sodium hydride was added in batches (980 mg, 24.5 mmol), and the reaction proceeded at this temperature for 10 minutes. A suspension of CDI (1.59 g, 9.8 mmol) in tetrahydrofuran (40 mL) was dropped to the above solution, and then the reaction proceeded at this temperature for 20 minutes. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 150 mL of ice water, and the pH was adjusted to about 4 with 3 M hydrochloric acid. The reaction solution was extracted with ethyl acetate. The organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, filtered. The filtrate was concentrated and dried to obtain 7-bromo-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4(1H, 3H)-dione (1.46 g, 76%), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=393.3.


Step 4: tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (1.04 g, 4.81 mmol) was added to a suspension of 60% sodium hydride (1.11 g, 18.5 mmol) in tetrahydrofuran (50 mL) at 0° C., reacted at 0° C. for 30 minutes, and 7-bromo-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4(1H, 3H)-dione (1.46 g, 3.7 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 30 minutes. The completion of the reaction was detected by LC-MS. The reaction solution was poured into 100 mL of ice water. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 20-100%) to obtain tert-butyl (S)-3-((((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetra hydroquinazolin-5-yl)oxy)methyl)piperazin-1-formate (1.34 g, 62%), a yellow solid. ES-API: [M+H]+=589.1.


Step 5: tert-butyl (S)-3-((((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetra hydroquinazolin-5-yl)oxy)methyl)piperazin-1-formate (1.34 g, 2.29 mmol), DBU (2.01 g, 13.74 mmol), PyBop (3.57 g, 6.87 mmol) dichloromethane (100 mL) were added to a round bottom flask. The reaction was stirred at room temperature for 30 minutes. The completion of the reaction was detected by LC-MS. 100 mL of dichloromethane was added to the reaction, and the organic phase was washed with 30 mL of hydrochloric acid (1M) and 100 mL of the saturated sodium bicarbonate aqueous solution in sequence. The organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain tert-butyl (S)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.04 g, 80%). ES-API: [M+H]+=571.4.


Step 6: tert-butyl (S)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.04 g, 1.83 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (429 mg, 2.75 mmol), SPhos (73 mg, 0.18 mmol), SPhos-Pd-G2 (129 mg, 0.18 mmol), potassium phosphate (1.16 g, 5.49 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, the organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (S)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (560 mg, 51%), a yellow solid. ES-API: [M+H]+=602.1.


Step 7: tert-butyl (S)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (560 mg, 0.93 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (S)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (582 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=502.1.


Step 8: (S)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (582 mg, 1.16 mmol), 6 mL of dichloromethane and triethylamine (1.17 g, 11.6 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (117 mg, 0.93 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: (S)-2-acryloyl-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one Z15 (257 mg, 40%). ES-API: [M+H]+=556.1. 1H NMR (400 MHz, DMSO-d6) δ 10.08 (d, J=1.8 Hz, 1H), 8.50 (d, J=5.0 Hz, 1H), 7.28 (d, J=5.0 Hz, 1H), 7.16 (dd, J=15.0, 8.2 Hz, 1H), 6.84 (s, 2H), 6.72 (d, J=8.2 Hz, 1H), 6.69-6.60 (m, 1H), 6.20 (d, J=16.4 Hz, 1H), 5.98 (s, 1H), 5.76 (s, 1H), 4.75 (s, 1H), 4.61 (s, 2H), 4.52-4.26 (m, 2H), 4.17-3.97 (m, 2H), 2.82-2.68 (m, 1H), 1.95 (d, J=3.4 Hz, 4H), 1.24 (s, 3H), 1.11-1.06 (m, 4H), 1.05-1.00 (m, 4H).


Step 9: Compound Z15 (257 mg, 0.46 mmol) was resolved chirally (mobile phase: methanol (0.2% ammonia methanol)); column type: Cellulose-SC (4.6*100*5 um); flow rate: 1.8 ml/min; column temperature: 40.2° C.) to obtain:


Compound Z15-1 (112 mg, retention time: 1.6 min, purity: 100%, de value: 99%). ES-API: [M+H]+=556.2.


and Compound Z15-2 (121 mg, retention time: 2.81 min, purity: 100%, de value: 99%). ES-API: [M+H]+=556.2.


Example 16: Preparation of Compound Z16



embedded image


embedded image


Step 1: sodium hydride (0.81 g, 20.25 mmol, 60%) was suspended in 15 ml of tetrahydrofuran solution and cooled to 0° C., and 7-bromo-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4(1H, 3H)-dione (1.44 g, 6.658 mmol), 10 ml of tetrahydrofuran solution was slowly dropped to the solution. After stirring for 5 minutes, tert-butyl (R)-3-(hydroxymethyl)piperazin-1-carboxylate (1.8 g, 4.589 mmol), 15 ml of tetrahydrofuran solution was dropped. After heating to 65° C., the reaction was stirred for 1 hour. 30 mL of ammonium chloride solution was added to the reaction solution. The reaction solution was extracted twice with 50 ml of ethyl acetate, dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-35%) to obtain the target product: tert-butyl (R)-3-((((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetra hydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.5 g, yield: 75%), a while-like solid. ES-API: [M+H]+=588.


Step 2: tert-butyl (R)-3-((((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetra hydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.5 g, 2.55 mmol) was dissolved in N,N-dimethylformamide (50 mL), benzotriazole-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (3.97 g, 7.64 mmol), 1,8-diazabicycloundec-7-ene (2.33 g, 15.29 mmol) were added in sequence. The reaction was stirred at room temperature for 1 hour. 150 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-20%) to obtain the target product: tert-butyl (R)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.0 g, yield: 69%), a light yellow solid. ES-API: [M+H]+=571.


Step 3: tert-butyl (R)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (200 mg, 0.35 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (82 mg, 0.526 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (25.24 mg, 0.035 mmol), 2-dicyclohexylphosphino-2′,6′-dimethoxy-biphenyl (14.37 mg, 0.035 mmol), potassium phosphate (223 mg, 0.105 mmol), 5 ml of 1,4-dioxane and 1 mL of water were added to a 100 ml three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction was stirred at 110° C. for 1 hour. 150 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-50%) to obtain the target product: tert-butyl (R)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-ox o-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (160 mg, yield: 76%), a light yellow solid. ES-API: [M+H]+=602.


Step 4: tert-butyl (R)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-ox o-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (160 mg, 0.266 mmol) was dissolved in dichloromethane (5 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours, and the reaction solution was concentrated to obtain the target product: (R)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (175 mg). The crude product was directly used in the next step. ES-API: [M+H]+=502.


Step 5: (R)-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (175 mg, 0.266 mmol) was dissolved in dichloromethane (15 mL), and triethylamine (1.0 mL, 7.21 mmol) was added. The reaction was cooled to 0° C., and acrylic anhydride (26.83 mg, 0.213 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 80 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 100 mL of the saturated NaHCO3 aqueous solution, 80 mL of the saturated brine, dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-60%) to obtain the target product: (R)-2-acryloyl-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one Z16 (60 mg, yield: 67%), a yellow solid. ES-API: [M+H]+=556. 1H-NMR (500 MHz, DMSO-d6) δ 10.05 (d, J=92.9 Hz, 1H), 8.50 (d, J=4.9 Hz, 1H), 7.35-7.24 (m, 1H), 7.16 (td, J=8.3, 6.7 Hz, 1H), 6.90 (t, J=13.6 Hz, 1H), 6.84 (d, J=2.9 Hz, 1H), 6.71 (d, J=8.3 Hz, 1H), 6.65 (dd, J=9.9, 8.4 Hz, 1H), 6.20 (d, J=16.7 Hz, 1H), 5.98 (dt, J=4.7, 1.4 Hz, 1H), 5.77 (dd, J=10.4, 2.4 Hz, 1H), 4.76 (d, J=18.0 Hz, 1H), 4.67-4.54 (m, 2H), 4.34 (d, J=17.2 Hz, 1H), 4.05 (s, 1H), 3.51 (dd, J=20.8, 11.2 Hz, 1H), 3.29-2.98 (m, 2H), 2.76 (dp, J=26.7, 6.7 Hz, 1H), 1.95 (d, J=4.5 Hz, 3H), 1.08 (t, J=6.7 Hz, 3H), 1.03 (dd, J=6.7, 4.2 Hz, 3H).


Example 17: Preparation of Compound Z17, Z17-1 and Z17-2



embedded image


embedded image


Step 1: 4-bromo-2,6-difluorobenzonitrile (10 g, 45.87 mmol), 40 mL of isopropanol and cyclopropylamine (13 g, 229 mmol) were added to the sealed tube. The reaction was heated under a 70° C. oil bath for 2 hours. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated. The crude product was washed with petroleum ether to obtain the product: 4-bromo-2-(cyclopropylamino)-6-fluorobenzonitrile (11 g, 94%), a white solid. The crude product was directly used in the next step. ES-API: [M+H]+=255.1.


Step 2: 4-bromo-2-(cyclopropylamino)-6-fluorobenzonitrile (10 g, 39.21 mmol), N-chlorosuccinimide (5.7 g, 43.13 mmol) and acetonitrile (150 mL) were added to a round bottom flask. The reaction was stirred at 65° C. for 0.5 hour. sodium thiosulfate aqueous solution was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-5%) to obtain 4-bromo-3-chloro-6-(cyclopropylamino)-2-fluorobenzonitrile (3.97 g, 35%), a white solid. HNMR (400 MHz, CDCl3) 7.17 (s, 1H), 5.13 (s, 1H), 2.50-2.48 (m, 1H), 0.92-0.90 (m, 2H), 0.62-0.60 (m, 2H). ES-API: [M+H]+=289.1.


Step 3: 4-bromo-3-chloro-6-(cyclopropylamino)-2-fluorobenzonitrile (5 g, 17.30 mmol), potassium carbonate (4.77 g, 34.60 mmol) and dimethyl sulfoxide (30 mL) were added to a round bottom flask. 30% hydrogen peroxide (3.47 mL, 34.60 mmol) was added under stirring. The reaction was stirred at room temperature for 3 hours. Ice water was added to the reaction solution, and a solid precipitated out. It was filtered and the filter cake was vacuum-dried to obtain 4-bromo-3-chloro-6-(cyclopropylamino)-2-fluorobenzamide (4.7 g, 89%), a white solid. HNMR (400 MHz, DMSO-d6) 7.91 (s, 1H), 7.85 (s, 1H), 7.15 (s, 1H), 6.96 (s, 1H), 2.502-2.45 (m, 1H), 0.80-0.75 (m, 2H), 0.47-0.43 (m, 2H). ES-API: [M+H]+=306.9.


Step 4: 4-bromo-3-chloro-6-(cyclopropylamino)-2-fluorobenzamide (4.7 g, 16.7 mmol) and 80 mL of dry tetrahydrofuran were added to a round bottom flask, cooled to 0-5° C. under the ice bath, sodium hydride (3.3 g, 83.5 mmol) was added in batches, the reaction proceeded at this temperature for 5 minutes. A suspension of CDI (3.2 g, 20.04 mmol) in tetrahydrofuran (40 mL) was dropped to the above solution, and then the reaction proceeded at this temperature for 15 minutes. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 150 mL of ice water, and the pH was adjusted to about 3 with 3 M hydrochloric acid. The reaction solution was extracted with ethyl acetate. The organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, and filtered. The filtrate was concentrated and dried to obtain 7-bromo-6-chloro-1-cyclopropyl-5-fluoro-4-hydroxyquinazolin-2 (1H)-one (3 g, 55%), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=333.1.


Step 5: 60% NaH (180 mg, 4.5 mmol), tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (291 mg, 1.35 mmol), tetrahydrofuran (12 mL) were added to a round bottom flask. The reaction was cooled to 0° C., and a solution of 7-bromo-6-chloro-1-cyclopropyl-5-fluoro-4-hydroxyquinazolin-2 (1H)-one (300 mg, 0.90 mmol) in tetrahydrofuran (3 mL) was dropped thereto. The reaction was stirred at 0° C. for 5 minutes, and then the reaction was stirred under a 60° C. oil bath for 1 hour. The completion of the reaction was detected by LC-MS. 30 mL of the saturated NH4Cl aqueous solution was added into the reaction solution. The reaction solution was extracted with 40 mL of ethyl acetate for 3 times. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-5%) to obtain the target product: tert-butyl (S)-3-((((7-bromo-6-chloro-1-cyclopropylmethyl-4-hydroxy-2-oxo-1,2-dihydro quinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (470 mg), a yellow solid, yield of 98%. ES-API: [M+H]+=529.1.


Step 6: tert-butyl (S)-3-((((7-bromo-6-chloro-1-cyclopropylmethyl-4-hydroxy-2-oxo-1,2-dihydro quinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (450 mg, 0.85 mmol), 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (1.3 g, 2.55 mmol) N,N-dimethylformamide (8 mL) were added to a round bottom flask. DBU (646 mg, 4.25 mmol) was added under stirring. The reaction was stirred at room temperature for 2 hours. The completion of the reaction was detected by LC-MS. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times. The organic phase was washed with the saturated brine for three times, dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the target product: tert-butyl (S)-10-bromo-11-chloro-8-cyclopropyl-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (420 mg), a yellow solid, yield of 96%. ES-API: [M+H]+=511.0.


Step 7: tert-butyl (S)-10-bromo-11-chloro-8-cyclopropyl-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (370 mg, 0.72 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (190 mg, 1.08 mmol), SPhos (29 mg, 0.072 mmol), SPhos-Pd-G2 (52 mg, 0.072 mmol), potassium phosphate (457 mg, 2.16 mmol), 8 mL of dioxane and 2 mL of water were added to a round bottom flask. The round bottom flask was placed in a microwave reactor at 105° C. and the reaction proceeded for 2 hours, and the reaction stopped. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-2%) to obtain the target product: tert-butyl (13aS)-11-chloro-8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (400 mg), a yellow solid, purity of 60%, yield of 59%. ES-API: [M+H]+=563 0.2.


Step 8: tert-butyl (13aS)-11-chloro-8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (400 mg, 0.44 mmol), 3 mL of dichloromethane and 3 mL of trifluoroacetic acid were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain the crude product: (13aS)-11-chloro-8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (203 mg), directly used in the next step. ES-API: [M+H]+=463.2.


Step 9: (13aS)-11-chloro-8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (203 mg, 0.44 mmol), 5 mL of dichloromethane and triethylamine (222 mg, 2.2 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., acrylic anhydride in dichloromethane (39 mg, 0.3 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 10 mL of the saturated NaHCO3 aqueous solution was added to the reaction solution, and the reaction solution was extracted with 10 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: Z17 (55.94 mg, 24%), a white solid. 1HNMR (400 MHz, CDCl3) 7.62 (s, 1H), 7.53-7.35 (m, 3H), 6.60-6.56 (m, 1H), 6.42-6.38 (m, 1H), 5.83-5.80 (m, 1H), 4.90-4.87 (m, 1H), 4.60-4.55 (m, 3H), 4.10-3.90 (m, 2H), 3.56-3.26 (m, 3H), 2.82-2.81 (m, 1H), 2.26-2.25 (m, 3H), 1.22-1.15 (m, 2H), 0.88-0.80 (m, 2H). ES-API: [M+H]+=517.0.


Step 10: Compound Z17 (0.3 g) was resolved chirally (column type: Chiralpak IH 250 mm*4.6 mm*5 um; mobile phase: acetonitrile:isopropanol:ammonia methanol=90:10:0.2; flow rate: 1 ml/min; column temperature=30° C.) to obtain:


Compound Z17-1 (80 mg, retention time 5.666 min; peak 1), a white solid. 1HNMR (400 MHz, CDCl3) 7.68 (s, 1H), 7.58 (d, J=4 Hz, 1H), 7.43-7.41 (m, 2H), 6.62-6.56 (m, 1H), 6.42-6.39 (m, 1H), 5.82-5.80 (m, 1H), 4.90-4.87 (m, 1H), 4.60-4.51 (m, 3H), 4.10-3.91 (m, 2H), 3.60-3.29 (m, 3H), 2.83-2.82 (m, 1H), 2.26 (s, 3H), 1.22-1.20 (m, 2H), 0.90-0.82 (m, 2H). ES-API: [M+H]+=517.1.


and Compound Z17-2 (85 mg, retention time 6.661 min; peak 2), a white solid. 1HNMR (400 MHz, CDCl3) 7.74-7.41 (m, 4H), 6.60-6.58 (m, 1H), 6.42-6.39 (m, 1H), 5.82-5.80 (m, 1H), 4.89-4.88 (m, 1H), 4.60-4.57 (m, 3H), 4.06-3.96 (m, 2H), 3.60-3.30 (m, 3H), 2.83-2.82 (m, 1H), 2.26 (s, 3H), 1.20-1.16 (m, 2H), 0.92-0.82 (m, 2H). ES-API: [M+H]+=517.1.


Example 18: Preparation of Compound Z18



embedded image


Step 1: tert-butyl (S)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (500 mg, 0.9 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (238 mg, 1.35 mmol), SPhos (37 mg, 0.09 mmol), SPhos-Pd-G2 (65 mg, 0.09 mmol), potassium phosphate (600 mg, 2.25 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (R)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (346 mg, 62%), a yellow solid. ES-API: [M+H]+=622.1.


Step 2: tert-butyl (R)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (346 mg, 0.56 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (R)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (361 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=522.1.


Step 3: (R)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (361 mg, 0.69 mmol), 10 mL of dichloromethane and triethylamine (700 mg, 6.9 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (69 mg, 0.55 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: (R)-2-acryloyl-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one Z18 (134 mg, 35%). ES-API: [M+H]+=576.1. 1H NMR (400 MHz, DMSO-d6) δ 13.12 (s, 1H), 8.44 (d, J=4.8 Hz, 1H), 7.55-7.39 (m, 2H), 7.25 (dd, J=19.8, 6.0 Hz, 2H), 6.87 (s, 2H), 6.22 (d, J=16.6 Hz, 1H), 5.85 (s, 1H), 5.78 (d, J=10.4 Hz, 1H), 4.70 (d, J=12.0 Hz, 3H), 4.53-4.28 (m, 2H), 4.13 (s, 2H), 3.54 (s, 2H), 2.93-2.72 (m, 1H), 2.13 (s, 3H), 2.01 (d, J=5.6 Hz, 3H), 1.08 (d, J=6.1 Hz, 3H), 0.97 (dd, J=12.3, 6.7 Hz, 4H).


Example 19: Preparation of Compound Z19, Z19-1 and Z19-2



embedded image


Step 1: tert-butyl (S)-10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (500 mg, 0.9 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (238 mg, 1.35 mmol), SPhos (37 mg, 0.09 mmol), SPhos-Pd-G2 (65 mg, 0.09 mmol), potassium phosphate (600 mg, 2.25 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (S)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (381 mg, 68%), a yellow solid. ES-API: [M+H]+=622.1.


Step 2: tert-butyl (S)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (381 mg, 0.61 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (S)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8 H)-one (401 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=522.1.


Step 3: (S)-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (401 mg, 0.96 mmol), 10 mL of dichloromethane and triethylamine (971 mg, 9.6 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and a solution of acrylic anhydride in dichloromethane (97 mg, 0.77 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: (S)-2-acryloyl-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one Z19 (248 mg, 45%). ES-API: [M+H]+=576.1.


Step 4: the above-obtained Compound Z19 (248 mg, 0.43 mmol) was resolved chirally (mobile phase: n-hexane (0.1% ammonia methanol):ethanol (0.1% ammonia methanol)=50:50); column type: AY-H (250*4.6 mm 5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain:


Compound Z19-1 (105 mg, retention time: 8.821 min, purity: 99%, de value: 99%). ES-API: [M+H]+=576.3.


and Compound Z19-2 (115 mg, retention time: 11.79 min, purity: 99%, de value: 99%). ES-API: [M+H]+=576.2. 1H NMR (400 MHz, DMSO-d6) δ 13.12 (s, 1H), 8.44 (d, J=4.8 Hz, 1H), 7.45 (dd, J=16.7, 6.5 Hz, 2H), 7.27 (d, J=3.2 Hz, 1H), 7.22 (d, J=8.6 Hz, 1H), 6.88 (t, J=10.8 Hz, 2H), 6.21 (d, J=16.3 Hz, 1H), 5.84 (d, J=1.7 Hz, 1H), 5.78 (dd, J=10.5, 2.4 Hz, 1H), 4.85-4.60 (m, 3H), 4.53-4.29 (m, 2H), 4.24-3.95 (m, 2H), 2.79 (ddd, J=20.1, 13.5, 6.8 Hz, 1H), 2.12 (s, 3H), 2.00 (d, J=5.8 Hz, 4H), 1.09-1.05 (m, 4H), 0.99-0.91 (dd, J=12.0, 6.8 Hz, 3H).


Example 20: Preparation of Compound Z20, Z20-1, Z20-2, Z20-3 and Z20-4



embedded image


embedded image


Step 1: methyl 6-amino-4-bromo-3-chloro-2-fluorobenzoate (6.3 g, 22.35 mmol), (5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)boronic acid (9.3 g, 35.77 mmol), 2-bicyclohexylphosphin-2′,6′-dimethoxybiphenyl (734 mg, 1.78 mmol), SPhos-Pd-G2 (1.27 g, 1.78 mmol), potassium phosphate (14.2 g, 67.05 mmol), 100 mL of dioxane and 20 mL of water were added to a reaction flask. The reaction proceeded under a 90° C. oil bath for 2 hours under nitrogen protection, and the reaction stopped. 100 mL of water was added to the reaction solution. The reaction solution was extracted with 100 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-25%) to obtain the target product: methyl 6-amino-3-chloro-2-fluoro-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoate (6.7 g), a yellow solid, purity of 58%. ES-API: [M+H]+=418.0.


Step 2: methyl 6-amino-3-chloro-2-fluoro-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoate (6.2 g, 14.8 mmol), 1-iodo-2-isopropylbenzene (5.4 g, 22.2 mmol), XantPhos-Pd-G2 (0.92 g, 1.03 mmol), 4,5-bisdiphenylphosphino-9,9-dimethylxanthene (0.60 g, 1.03 mmol), cesium carbonate (9.7 g, 29.6 mmol), and 120 mL of toluene were added to a round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction was stirred at 90° C. for 21 hours. The completion of the reaction was detected by LC-MS. Water was added to the reaction solution. The reaction solution was extracted with ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-10%) to obtain the target product: methyl 3-chloro-2-fluoro-6-(((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoate (5.55 g), yield of 70%. ES-API: [M+H]+=536.1.


Step 3: methyl 3-chloro-2-fluoro-6-(((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoate (6.5 g, 12.1 mmol), sodium hydroxide (1.5 g, 36.4 mmol), methanol (50 mL), tetrahydrofuran (50 mL) and water (20 mL) were added to a round bottom flask. The reaction was stirred at 45° C. for 1 hour. The completion of the reaction was detected by LC-MS. The pH of the reaction was adjusted to 8 with 6M aqueous hydrochloric acid, and then the organic solvent was concentrated and removed. Water was added, and the reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated to obtain 3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoic acid (5.9 g, 94%). ES-API: [M+H]+=522.1.


Step 4: 3-chloro-2-fluoro-6-((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzoic acid (6.3 g, 12.1 mmol), ammonium chloride (1.9 g, 36.2 mmol), 2-(7-azobenzotriazole)-N,N,N′,N′-tetramethylurea hexafluorophosphate (6.9 g, 18.2 mmol), dichloromethane (40 mL), N,N-dimethylformamide (20 mL) and triethylamine (6.1 g, 60.5 mmol) were added to the round bottom flask in sequence at 0° C. The reaction was stirred at room temperature for 4 hours. Water was added to the reaction solution. The reaction solution was extracted with ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by reversed-phase column (acetonitrile/water: 20%-100%) to obtain the target product: 3-chloro-2-fluoro-6-(((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzamide (6.1 g, 95%), a yellow solid. ES-API: [M+H]+=521.2.


Step 5: 60% NaH (2.3 g, 58.5 mmol) and tetrahydrofuran (60 mL) were added to a round bottom flask. The reaction was cooled to 0° C., and a solution of 3-chloro-2-fluoro-6-(((2-isopropylphenyl)amino)-4-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)benzamide (6.1 g, 11.7 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 5 minutes, and then a solution of N,N′-carbonyl diimidazole (2.8 g, 17.5 mmol) in tetrahydrofuran (40 mL) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 30 minutes. The completion of the reaction was detected by LC-MS. The reaction solution was poured into icy saturated sodium bicarbonate aqueous solution. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated to obtain the target product: 6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylphenyl)-7-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)quinazolin-2 (1H)-one (6.2 g, 98%), a yellow solid. ES-API: [M+H]+=547.2.


Step 6: 60% NaH (2.3 g, 57.5 mmol), tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (4 g, 18.4 mmol), tetrahydrofuran (100 mL) were added to a round bottom flask. The reaction was cooled to 0° C., and a solution of 6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylphenyl)-7-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)quinazolin-2 (1H)-one (6.3 g, 11.5 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 5 minutes, and then the reaction was stirred under a 65° C. oil bath for 0.5 hour. The completion of the reaction was detected by LC-MS. The reaction solution was poured into icy saturated sodium bicarbonate aqueous solution. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-7%) to obtain the target product: tert-butyl (3S)-3-((((6-chloro-4-hydroxy-1-(2-isopropylphenyl)-7-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)meth ylpiperazin-1-formate (6.7 g, 79%), a yellow solid. ES-API: [M+H]+=743.2.


Step 7: tert-butyl (3S)-3-((((6-chloro-4-hydroxy-1-(2-isopropylphenyl)-7-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)meth ylpiperazin-1-formate (6 g, 8.1 mmol), N,N-dimethylformamide (40 mL) and 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (10.5 g, 20.2 mmol) were added to a round bottom flask. DBU was added all at once while stirring (6.1 g, 40.4 mmol). The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction was directly purified by a C18 reversed-phase column (acetonitrile/water (0.5% trifluoroacetic acid): 0-80%) to obtain the target product: tert-butyl (13aS)-11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)-7 oxo 3,4,7,8,13,13a hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxy late (4.1 g, 70%), a yellow solid. ES-API: [M+H]+=589.1. ES-API: [M+H]+=725 0.2.


Step 8: tert-butyl (13aS)-11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-4-yl)-7 oxo 3,4,7,8,13,13a hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxy late (4.1 g, 5.6 mmol), 5 mL of dichloromethane and 20 mL of trifluoroacetic acid were added to a round bottom flask. The reaction was stirred at room temperature for 2 hours. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated at 40° C. to obtain the crude product: (13aS)-11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazol in-7 (8H)-one (3 g), a yellow solid, directly used in the next step. ES-API: [M+H]+=541.2.


Step 9: (13aS)-11-chloro-8-(2-isopropylphenyl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (3 g, 5.6 mmol), 20 mL of dichloromethane and triethylamine (2.8 g, 28 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride (635 mg, 5.04 mmol) was added to the reaction solution. The reaction was stirred at 0° C. for 20 minutes. 100 mL of the saturated NaHCO3 aqueous solution was added to the reaction solution, and the reaction solution was extracted with dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: Z20 (1.1 g, 33%), a white solid. 1HNMR (400 MHz, CDCl3): 7.50-7.30 (m, 4H), 7.28-7.26 (m, 2H), 7.11-7.09 (m, 1H), 6.68-6.61 (m, 1H), 6.44-6.40 (m, 1H), 6.21-6.20 (m, 1H), 5.84-5.81 (m, 1H), 5.05-5.01 (m, 1H), 4.72-4.65 (m, 3H), 4.08-3.98 (m, 2H), 3.62-3.58 (m, 1H), 3.21-3.12 (m, 2H), 2.76-2.70 (m, 1H), 2.12-2.10 (m, 3H), 1.22-1.20 (m, 3H), 0.88-0.86 (m, 3H). ES-API: [M+H]+=595.2.


Step 10: Compound Z20 (1.1 g) was resolved chirally (column type: Chiralpak IA 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol:ammonia methanol=50:50:0.2; flow rate: 1 ml/min; column temperature=30° C.) to obtain:


Compound Z20-1 (151.17 mg, retention time 6.439 min; peak 1), a white solid. 1HNMR (400 MHz, CDCl3) 7.42-7.30 (m, 4H), 7.22-7.16 (m, 2H), 7.10-7.00 (m, 1H), 6.56-6.53 (m, 1H), 6.33-6.32 (m, 1H), 6.06 (s, 1H), 5.74-5.71 (m, 1H), 4.94-4.91 (m, 1H), 4.67-4.57 (m, 3H), 4.00-3.92 (m, 2H), 3.54-3.22 (m, 3H), 2.66-2.63 (m, 1H), 2.03 (s, 3H), 0.94 (d, J=4 Hz, 3H), 0.88 (d, J=4 Hz, 3H). ES-API: [M+H]+=595.2.


Compound Z20-2 (110.33 mg; peak 4, retention time: 10.952 min), a white solid. 1HNMR (400 MHz, CDCl3) 7.38-7.21 (m, 4H), 7.20-7.19 (m, 2H), 7.10-7.00 (m, 1H), 6.56-6.53 (m, 1H), 6.33-6.32 (m, 1H), 6.13 (s, 1H), 5.74-5.71 (m, 1H), 4.95-4.92 (m, 1H), 4.67-4.57 (m, 3H), 4.00-3.92 (m, 2H), 3.54-3.22 (m, 3H), 2.66-2.63 (m, 1H), 2.03 (s, 3H), 1.13 (d, J=4 Hz, 3H), 0.95 (d, J=4 Hz, 3H). ES-API: [M+H]+=595.2.


Compound Z20-3 (232.96 mg; peak 2, retention time: 7.026 min), a white solid. 1HNMR (400 MHz, CDCl3) 7.47-7.32 (m, 4H), 7.20-7.19 (m, 2H), 7.10-7.05 (m, 1H), 6.66-6.60 (m, 1H), 6.40-6.38 (m, 1H), 6.20 (s, 1H), 5.84-5.81 (m, 1H), 5.00-4.90 (m, 1H), 4.69-4.57 (m, 3H), 4.00-3.92 (m, 2H), 3.73-3.32 (m, 3H), 2.71-2.68 (m, 1H), 2.10 (s, 3H), 1.22 (d, J=4 Hz, 3H), 1.01 (d, J=4 Hz, 3H). ES-API: [M+H]+=595.2.


and Compound Z20-4 (196.33 mg; peak 3, retention time: 8.800 min), a white solid. 1HNMR (400 MHz, CDCl3) 7.64-7.24 (m, 6H), 7.14-7.05 (m, 1H), 6.66-6.62 (m, 1H), 6.40-6.38 (m, 1H), 6.21 (s, 1H), 5.86-5.81 (m, 1H), 5.03-4.92 (m, 1H), 4.75-4.57 (m, 3H), 4.10-4.00 (m, 2H), 3.70-3.25 (m, 3H), 2.71-2.68 (m, 1H), 2.10 (s, 3H), 1.22 (d, J=4 Hz, 3H), 1.05 (d, J=4 Hz, 3H). ES-API: [M+H]+=595.2.


Example 21: Preparation of Compound Z21



embedded image


embedded image


Step 1: 4-bromo-2,6-difluorobenzonitrile (50.0 g, 0.229 mol), cyclopropylamine (17.1 g, 1.146 mol) and 500 mL of isopropanol were added to a 1000 mL three-necked round bottom flask, and the reaction was stirred at 70° C. for 2 hours. After the completion of the reaction, the reaction solution was cooled to room temperature, a solid precipitated out, and filtered to obtain the target product: 4-bromo-2-(cyclopropylamino)-6-fluorobenzonitrile (53 g, yield: 91%) ES-API: [M+H]+=255.0.


Step 2: 4-bromo-2-(cyclopropylamino)-6-fluorobenzonitrile (52.0 g, 0.204 mol), potassium carbonate (55.5 g, 0.408 mol) and 500 mL of dimethyl sulfoxide were added to a 1000 mL three-necked round bottom flask. 30% hydrogen peroxide (93.0 g, 0.734 mol) was slowly dropped at room temperature, The reaction solution was stirred for 3 hours at room temperature. 500 mL of water was added to the reaction solution, a large amount of solid precipitated out, filtered to obtain the target product: 4-bromo-2-(cyclopropylamino)-6-fluorobenzamide (55 g, yield: 97%) ES-API: [M+H]+=273 0.0.


Step 3: 4-bromo-2-(cyclopropylamino)-6-fluorobenzonitrile (13.2 g, 0.048 mol) and 150 mL of anhydrous tetrahydrofuran were added to a 500 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. sodium hydride (9.62 g, 0.240 mol) was added at 0° C., the reaction proceeded for 0.5 hour keeping this temperature, and a solution of N,N′-carbonyl diimidazole in anhydrous tetrahydrofuran (1.44 mol/L, 50 mL) was quickly added. After 5 min for the completion of the reaction, the reaction solution was added to ice water, extracted with ethyl acetate for three times, the organic phase was dried with anhydrous sodium sulfate and filtered, and the filtrate was concentrated to obtain the crude product. The crude product was slurried with 100 mL of methyl tert-butyl ether overnight and filtered to obtain the target product: 7-bromo-1-cyclopropyl-5-fluoro-4-hydroxyquinolin-2 (1H)-one (9 g, yield: 63%) ES-API: [M+H]+=299.0 s.


Step 4: tert-butyl 3-(2-hydroxyethyl)piperazin-1-formate (6.1 g, 0.026 mol) and 80 mL of anhydrous tetrahydrofuran were added to a 250 mL three-necked round bottom flask, the system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. Sodium hydride (4.5 g, 0.115 mol) was added at 0° C., the reaction proceeded for 0.5 hour keeping this temperature, a solution of 7-bromo-1-cyclopropyl-5-fluoro-4-hydroxyquinolin-2 (1H)-one in anhydrous tetrahydrofuran (1.12 mol/L, 20 mL) was added, and the reaction proceeded at room temperature for 1.5 hours. The completion of the reaction was detected by mass spectrometry. The reaction solution was added to ice water, extracted with ethyl acetate for three times, the organic phase was dried with anhydrous sodium sulfate and filtered, and the filtrate was concentrated to obtain the target product: 3-(2-(((7-bromo-1-cyclopropyl-4-hydroxy-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)ethyl)piperazine-1-formate tert-butyl (9 g, yield: 78%) ES-API: [M+H]+=509.1.


Step 5: tert-butyl 3-(2-(((7-bromo-1-cyclopropyl-4-hydroxy-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)ethyl)piperazine-1-formate (3.0 g, 0.006 mol), 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (9.3 g, 0.018 mol), 1,8-diazabicyclo[5.4.0]undec-7-ene (4.56 g, 0.03 mol) and 50 mL of N,N-dimethylformamide were added to a 100 mL three-necked round bottom flask, and stirred at room temperature for 2 hours. 80 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 80 mL of the saturated brine for three times. The phase in ethyl acetate was dried and concentrated, and the crude product was purified by a fast silica gel column to obtain the target product: tert-butyl 10-bromo-8-cyclopropyl-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazoline[4,3,2-de]quinazolin-2-carboxylate (0.65 g, yield: 23%). ES-API: [M+H]+=490.3.


Step 6: tert-butyl 10-bromo-8-cyclopropyl-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazoline[4,3,2-de]quinazolin-2-carboxylate (0.49 g, 0.001 mol), (5-methyl-1H-indazol-4-yl)boronic acid (0.260 g, 0.0015 mol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (0.072 g, 0.0001 mol), SPhos (0.041 g, 0.0001 mol), potassium phosphate (0.64 g, 0.003 mol), 10 mL of dioxane and 2 mL of water were added to a 100 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction proceeded at 110° C. for 2 hours. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert butyl 8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylate (550 mg, yield: 83%). ES-API: [M+H]+=543.3.


Step 7: tert-butyl 8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylate (550 mg, 0.001 mol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours, and the reaction solution was concentrated to obtain the target product: 8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one (500 mg), the crude product was directly used in the next step. ES-API: [M+H]+=443.3.


Step 8: 8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one (500 mg, 0.0011 mol) was dissolved in dichloromethane (15 mL), and triethylamine (0.034 g, 0.0003 mol) was added. The reaction was cooled to 0° C., and acrylic chloride (138 mg, 0.0011 mol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 80 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 100 mL of the saturated NaHCO3 aqueous solution, 80 mL of the saturated brine, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: 2-acryloyl-8-cyclopropyl-10-(5-methyl-1H-indazol-4-yl)-1,3,4,13,14,14a-hexa hydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one Z21 (80 mg, yield: 14%). ES-API: [M+H]+=497.2. 1H NMR (500 MHz, DMSO-d6) δ 13.13 (s, 1H), 7.81 (s, 1H), 7.50 (d, J=8.3 Hz, 1H), 7.28 (s, 2H), 6.78 (s, 2H), 6.19 (t, J=14.8 Hz, 1H), 5.75 (d, J=8.9 Hz, 1H), 4.85-4.58 (m, 1H), 4.48-3.97 (m, 5H), 3.60 (d, J=13.2 Hz, 1H), 3.05 (s, 2H), 2.70 (s, 1H), 2.37 (s, 3H), 2.01 (dd, J=81.0, 33.5 Hz, 2H), 1.06 (d, J=56.8 Hz, 2H), 0.67 (d, J=72.2 Hz, 2H). ES-API: [M+H]+=497.2.


Example 22: Preparation of Compound Z22



embedded image


embedded image


Step 1: Tert-butyl 3-(2-hydroxyethyl)piperazin-1-formate (1.76 g, 7.65 mmol) was added to a suspension of sodium hydride (404 mg, 10.20 mmol) in tetrahydrofuran (40 mL) under the ice bath, and stirred for 5 minutes. Then 7-bromo-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4(1H, 3H)-dione (2 g, 5.10 mmol) was added to the reaction solution, heated to 60° C., and stirred for 2 minutes. After cooling to room temperature, the reaction solution was quenched with the saturated ammonium chloride solution (40 mL), extracted with ethyl acetate (50 mL*3), the organic phases were combined and dried, concentrated and purified by a fast silica gel column. (0-10% methanol/dichloromethane) to obtain a white solid of tert-butyl 3-(2-((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydroquinazolin-5-yl)oxy)ethyl)piperazin-1-carboxylate (1.5 g, yield: 44%). ES-API: [M+H]+=602.0.


Step 2: tert-butyl 3-(2-((7-bromo-1-(2-isopropyl-4-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydroquinazolin-5-yl)oxy)ethyl)piperazin-1-carboxylate (1.5 g, 2.49 mmol) was dissolved in N,N-dimethylformamide (50 mL), 1,8-diazabicycloundec-7-ene (1.89 g, 12.45 mmol) and 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (3.89 g, 7.47 mmol) were added in sequence, and stirred for 2 hours at room temperature. The reaction solution was poured into the saturated brine (50 mL), extracted with ethyl acetate (50 mL*3), the organic phases were combined and dried, concentrated and purified by normal phase column (0-100% acetonitrile/water (1%0 trifluoroacetic acid)) to obtain a yellow oily compound of tert-butyl 10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-1,3,4,7,8,13,14,14a-octa hydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylat e (800 mg, yield: 55%). ES-API: [M+H]+=584.0.


Step 3: tert-butyl 10-bromo-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-1,3,4,7,8,13,14,14a-octa hydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylat e (200 mg, 0.34 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (90 mg, 0.51 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (24 mg, 0.03 mmol), 2-bicyclohexylphosphin-2′,6′-dimethoxybiphenyl (14 mg, 0.03 mmol) and potassium phosphate (218 mg, 1.03 mmol) in a mixture of 1,4-dioxane (4 mL) and water (1 mL) was stirred for 1 hour at 120° C. under nitrogen protection. the reaction solution was dissolved in ethyl acetate (20 mL), washed with the saturated brine (10 mL*3), the obtained organic phase was dried and concentrated, and purified by a fast silica column (0-10% methanol/dichloromethane) to obtain a white solid of tert-butyl 8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidino[4,3,2-de]quinazolin-2-carboxylate (120 mg, purity: 44%). ES-API: [M+H]+=636.3.


Step 4: trifluoroacetic acid (0.5 mL) was added to a solution of tert-butyl 8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidino[4,3,2-de]quinazolin-2-carboxylate (120 mg, 0.19 mmol) in dichloromethane (2 mL) under the ice bath, stirred for 2 hours at room temperature, and concentrated to obtain a yellow oily compound of 8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′: 5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one, directly used in the next step. ES-API: [M+H]+=536.2.


Step 5: under the ice bath, triethylamine (191 mg, 1.89 mmol) was added to the above solution of the compound 8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′: 5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one in dichloromethane (2 mL). After the reaction solution was clarified, acrylic anhydride (22 mg, 0.18 mmol) was dropped, and stirred for 5 minutes. The reaction solution was washed with the saturated sodium bicarbonate solution (5 mL), the organic phase was dried and concentrated, and then purified by Preparative HPLC (ammonium bicarbonate system) to obtain a white solid of Z22 (43.39 mg, purity: 100%, yield: 39%). 1H NMR (500 MHz, DMSO-d6) δ 13.10 (s, 1H), 8.42 (dd, J=11.3, 4.8 Hz, 1H), 7.50 (s, 1H), 7.42 (d, J=8.6 Hz, 1H), 7.26 (t, J=5.5 Hz, 1H), 7.21 (d, J=8.4 Hz, 1H), 6.91-6.76 (m, 2H), 6.26-6.17 (m, 1H), 5.82-5.71 (m, 2H), 4.94-4.76 (m, 1H), 4.48 (s, 1H), 4.42-4.29 (m, 1H), 4.27-4.05 (m, 3H), 3.84-3.67 (m, 1H), 3.67-3.42 (m, 3H), 2.96-2.82 (m, 1H), 2.67-2.52 (m, 1H), 2.14 (d, J=2.5 Hz, 3H), 2.08 (s, 1H), 1.90 (s, 2H), 1.07 (d, J=6.6 Hz, 3H), 1.02 (d, J=6.6 Hz, 2H), 0.85 (d, J=6.7 Hz, 1H). ES-API: [M+H]+=590.3.


Example 23: Preparation of Compound Z23, Z23-1 and Z23-2



embedded image


embedded image


Step 1: tert-butyl (R)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.900 mg, 1.607 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (752 mg, 4.821 mmol), SPhos-Pd-G2 (115.7 mg, 0.1607 mmol), SPhos (66.0 mg, 0.1607 mmol), potassium phosphate (1.02 g, 4.821 mmol), dioxane (30 mL) and water (5.0 mL) were added to a single-neck flask, nitrogen was used for replacement for 3 times, and the reaction proceeded at 75° C. for 0.5-1 hour. After the completion of the reaction, 100 mL of the saturated brine was added to the reaction solution, extracted with ethyl acetate twice (100 mL*2). The phase in ethyl acetate was dried with anhydrous sodium sulfate, filtered, spin-dried and purified by column[dichloromethane:methanol=100:0˜80:20, (V/V)] to obtain tert-butyl (5 aR)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.660 g, yield: 58.1%). API: [M+H]+=637.2.


Step 2: tert-butyl (5 aR)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.660 g, 1.037 mmol) and methanol (5 mL) were added to a 100 mL single-neck round bottom flask, then dioxane hydrochloride solution (3.0 mL, 4M, 12.0 mmol) was added, and the reaction proceeded at room temperature for 2 hours. After the completion of the reaction, the solvent was spin-dried under reduced pressure to obtain the crude product: (5 aR)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (0.720 g, the crude product), API: [M+H]+=537.2. The crude product was directly used in the next step.


Step 3: (5 aR)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (0.720 g, 1.037 mmol) was dissolved in dichloromethane (20 mL), and triethylamine (1.04 g, 10.37 mmol) was added. The reaction was cooled to 0° C., acrylic anhydride (117 mg, 0.933 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 80 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 100 mL of the saturated NaHCO3 aqueous solution, 80 mL of the saturated brine, dried and concentrated, and prepared to obtain (5aR)-7-acryloyl-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-11 (12H)-one Z23 (395 mg, yield: 65%). ES-API: [M+H]+=591.1. 1H-NMR (500 MHz, DMSO-d6) δ 10.05 (t, J=13.1 Hz, 1H), 9.60 (s, 1H), 8.35 (d, J=4.8 Hz, 1H), 7.28-7.14 (m, 2H), 6.94-6.75 (m, 1H), 6.72-6.58 (m, 2H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.84-5.74 (m, 1H), 4.87 (dt, J=11.8, 10.7 Hz, 2H), 4.56 (s, 1H), 4.42 (d, J=51.3 Hz, 1H), 4.28 (s, 1H), 3.65 (d, J=13.9 Hz, 2H), 2.75 (dd, J=25.2, 18.5 Hz, 1H), 1.92 (dd, J=12.4, 7.7 Hz, 3H), 1.19 (dd, J=13.7, 7.1 Hz, 8H). Step 4: Compound Z23 (395 mg, 0.6695 mmol) was resolved (column type: IF, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=50:50, flow rate: 1 ml/min, column temperature=30° C.) to obtain:


Compound Z23-1 (106 mg, peak 1, retention time: 6.792 min, yield: 27%), a light yellow solid, ES-API: [M+H]+=591.1. 1H NMR (500 MHz, DMSO-d6) δ 10.03 (s, 1H), 8.35 (d, J=4.8 Hz, 1H), 7.27-7.12 (m, 2H), 6.95-6.77 (m, 1H), 6.72-6.59 (m, 2H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.87 (t, J=20.3 Hz, 2H), 4.42 (dd, J=99.0, 41.8 Hz, 3H), 4.08 (d, J=41.4 Hz, 1H), 3.70 (d, J=52.0 Hz, 2H), 3.36 (d, J=14.7 Hz, 1H), 2.77 (dt, J=13.3, 6.7 Hz, 1H), 1.91 (d, J=9.3 Hz, 3H), 1.07 (d, J=6.9 Hz, 3H), 0.93 (dd, J=6.5, 3.5 Hz, 3H).


and Z23-2 (102.7 mg, peak 2, retention time: 11.512 min, yield: 26%), a light yellow solid, ES-API: [M+H]+=591.1. 1H NMR (500 MHz, DMSO-d6) δ 10.02 (d, J=30.9 Hz, 1H), 8.35 (d, J=4.7 Hz, 1H), 7.28-7.11 (m, 2H), 6.95-6.79 (m, 1H), 6.66 (ddd, J=24.8, 12.3, 5.9 Hz, 2H), 6.21 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.94-4.83 (m, 2H), 4.42 (dd, J=102.4, 44.1 Hz, 3H), 4.04 (s, 1H), 3.70 (d, J=35.8 Hz, 2H), 3.36 (d, J=9.8 Hz, 1H), 2.70 (dt, J=13.6, 6.8 Hz, 1H), 1.93 (d, J=5.8 Hz, 3H), 1.07 (d, J=6.9 Hz, 3H), 0.93 (dd, J=6.5, 3.8 Hz, 3H).


Example 24: Preparation of Compound Z24, Z24-1 and Z24-2



embedded image


embedded image


Step 1: tert-butyl (S)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (500 mg, 0.95 mmol), N-chlorosuccinimide (255 mg, 1.9 mmol) and acetonitrile (50 mL) were added to a round bottom flask. The reaction was stirred at 80° C. for 2 hours. Sodium thiosulfate aqueous solution was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain the product: tert-butyl (S)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthale n-7 (5H)-carboxylate (533 mg, the crude product), a yellow solid. ES-API: [M+H]+=562.2.


Step 2: tert-butyl (S)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthale n-7 (5H)-carboxylate (533 mg, 0.95 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (222 mg, 1.43 mmol), tetratriphenylphosphine palladium (109 mg, 0.095 mmol), sodium carbonate (302 mg, 2.85 mmol), 30 mL of dioxane and 6 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 90° C. for 2 hours under nitrogen protection, and the reaction stopped. 30 mL of water was added to the reaction solution. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, the organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (5 aS)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (309 mg, 51%), a yellow solid. ES-API: [M+H]+=638.2.


Step 3: tert-butyl (5 aS)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (309 mg, 0.48 mmol), 1 mL of methanol and 3 mL of hydrogen chloride/dioxane solution (4 M) were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (5 aS)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (317 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=538.2.


Step 4: (5 aS)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (317 mg, 0.59 mmol), 10 mL of dichloromethane and triethylamine (597 mg, 5.9 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (59 mg, 0.47 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: (5 aS)-7-acryloyl-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-11 (12H)-one Z24 (108 mg, 40%). ES-API: [M+H]+=592.2.


Step 5: the above-obtained Compound Z24 (108 mg, 0.18 mmol) was resolved chirally (mobile phase: n-hexane-ethanol=40-60); column type: IC (250*4.6 mm 5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain:


Compound Z24-1 (45 mg, retention time: 6.91 min, purity: 99%, de value: 99%). ES-API: [M+H]+=592.3;


and Compound Z24-2 (55 mg, retention time: 11.77 min, purity: 99%, de value: 99%). ES-API: [M+H]+=592.2.


Example 25: Preparation of Compound Z25, Z25-1 and Z25-2



embedded image


embedded image


Step 1: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (200 mg, 0.3800 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (90.0 mg, 0.5769 mmol), SPhos-Pd-G2 (30 mg, 0.04167 mmol), SPhos (20 mg, 0.0487 mmol), potassium phosphate (0.240 g, 01.132 mmol), dioxane (10 mL) and water (2.0 mL) were added to a single-neck flask, nitrogen was used for replacement for 3 times, and the reaction proceeded at 120° C. for 0.5-1 hour. After the completion of the reaction, 100 mL of the saturated brine was added to the reaction solution. The reaction solution was extracted with ethyl acetate twice (100 mL*2). The phase in ethyl acetate was dried with anhydrous sodium sulfate, filtered, spin-dried and purified by column[dichloromethane:methanol=100:0˜80:20, (V/V)] to obtain tert-butyl (R)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.210 g, yield: 92%). API: [M+H]+=603.2.


Step 2: Tert-butyl (R)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.210 g, 0.3487 mmol) and methanol (5 mL) were added to a 100 mL single-neck round bottom flask, then dioxane hydrochloride solution (3.0 mL, 4M, 12.0 mmol) was added, and the reaction proceeded at room temperature for 2 hours. After the completion of the reaction, the solvent was spin-dried under reduced pressure to obtain the crude product: (R)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (0.234 g, the crude product), API: [M+H]+=503.2. The crude product was directly used in the next step.


Step 3: (R)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a, 6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (0.234 g, 0.3487 mmol) was dissolved in dichloromethane (15 mL), triethylamine (3.0 mL, 21.62 mmol) was added. The reaction was cooled to 0° C., and acrylic anhydride (39.5 mg, 0.3138 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 80 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 100 mL of the saturated NaHCO3 aqueous solution, 80 mL of the saturated brine, dried and concentrated, and prepared to obtain (R)-7-acryloyl-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one Z25 (152 mg, yield: 78%). ES-API: [M+H]+=557 0.2.


Step 4: Compound Z25 (152 mg, 0.2733 mmol) was resolved chirally (column type: Chiralpak IC 250 mm*4.6 mm 5 um, mobile phase: acetonitrile:isopropanol:ammonia methanol=60:40:0.2, flow rate: 1 ml/min, column temperature=30° C.) to obtain:


Compound Z25-1 (21 mg, yield: 13.8%), 1H NMR (500 MHz, DMSO-d6) δ 11.72 (s, 1H), 8.51 (d, J=4.8 Hz, 1H), 7.37-7.20 (m, 3H), 6.95-6.81 (m, 1H), 6.72 (dd, J=11.9, 8.4 Hz, 1H), 6.60 (d, J=8.3 Hz, 1H), 6.21 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.91-4.69 (m, 2H), 4.60 (s, 1H), 4.48-4.33 (m, 1H), 4.27 (s, 1H), 4.04 (s, 1H), 3.91-3.62 (m, 2H), 3.40 (d, J=10.6 Hz, 1H), 2.80 (dt, J=13.4, 6.7 Hz, 1H), 1.95 (s, 3H), 1.09 (d, J=6.7 Hz, 3H), 0.96 (d, J=6.7 Hz, 3H).


and Compound Z25-2 (19 mg, 12.5%), 1H NMR (500 MHz, DMSO-d6) δ 11.76 (s, 1H), 8.52 (d, J=4.9 Hz, 1H), 7.35-7.21 (m, 3H), 6.85 (ddd, J=27.3, 16.4, 10.3 Hz, 1H), 6.72 (dd, J=12.2, 8.2 Hz, 1H), 6.60 (d, J=8.4 Hz, 1H), 6.21 (dd, J=16.7, 2.1 Hz, 1H), 5.81-5.73 (m, 1H), 4.86-4.71 (m, 2H), 4.59 (s, 1H), 4.39 (dd, J=53.3, 12.2 Hz, 1H), 4.25 (s, 1H), 4.14-3.98 (m, 1H), 3.91-3.58 (m, 2H), 3.45-3.37 (m, 2H), 2.71 (dt, J=13.3, 6.6 Hz, 1H), 1.99 (s, 3H), 1.09 (d, J=6.6 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


Example 26: Preparation of Compound Z26, Z26-1, Z26-2, Z26-2-1 and Z26-2-2



embedded image


embedded image


Step 1: Compound 4-bromo-2-fluoro-6-(((2-isopropyl-4-methylpyridin-3-yl)amino)benzonitrile (2.0 g, 5.6 mmol) was dissolved in acetonitrile (20 ml), then NCS (1.5 g, 2.0 eq) was added, the reaction proceeded at 80° C. for four hours and completed, returned to room temperature, washed with water, extracted with ethyl acetate, and separated by column chromatography (PE:EA=10:1) to obtain the product: 4-bromo-3-chloro-2-fluoro-64 (2-isopropyl-4-methylpyridin-3-yl)amino)benzo nitrile (880 mg, yield: 40%). ES-API: [M+H]+=383.2.


Step 2: 4-bromo-3-chloro-2-fluoro-64 (2-isopropyl-4-methylpyridin-3-yl)amino)benzo nitrile (880 mg, 2.3 mmol) and 50 mL of dimethyl sulfoxide were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, potassium carbonate (634 mg, 4.6 mmol) and hydrogen peroxide (938 mg, 30% aqueous solution, 8.28 mmol) were added, and the reaction proceeded overnight at this temperature. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 15 mL of ice water, and a solid precipitated out. It was filtered to obtain filter cake. The filter cake was dried to obtain 4-bromo-3-chloro-2-fluoro-64 (2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (1.01 g, the crude product), a white solid, the crude product was directly used in the next step. ES-API: [M+H]+=401.2.


Step 3: 4-bromo-3-chloro-2-fluoro-64 (2-isopropyl-4-methylpyridin-3-yl)amino)benzamide (1.01 g, 2.49 mmol) and 20 mL of dry tetrahydrofuran were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, sodium hydride (500 mg, 12.45 mmol) was added in batches, and the reaction proceeded at this temperature for 10 minutes. A suspension of CDI (806 mg, 4.98 mmol) in tetrahydrofuran (20 mL) was dropped to the above solution, and then the reaction proceeded at this temperature for 20 minutes. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 50 mL of ice water, and the pH was adjusted to about 4 with 3 M hydrochloric acid. The reaction solution was extracted with ethyl acetate. The organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, filtered. The filtrate was concentrated and dried to obtain 7-bromo-6-chloro-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4 (1H,3H)-dione (1.1 g, the crude product), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=427.3.


Step 4: tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (557 mg, 2.58 mmol) was added to a suspension of 60% sodium hydride (412 mg, 10.32 mmol) in tetrahydrofuran (50 mL) at 0° C., the reaction proceeded at 0° C. for 30 minutes, a solution of 7-bromo-6-chloro-5-fluoro-1-(2-isopropyl-4-methylpyridin-3-yl)quinazolin-2,4 (1.1 g, 2.58 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 30 minutes. The completion of the reaction was detected by LC-MS. 50 mL of ice water was added into the reaction solution. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 20-100%) to obtain tert-butyl (S)-3-((((7-bromo-6-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.04 g, 64%), a yellow solid. ES-API: [M+H]+=623.3.


Step 5: tert-butyl (S)-3-((((7-bromo-6-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.04 g, 1.67 mmol), DBU (1.46 g, 10.02 mmol), PyBop (2.59 g, 6.87 mmol) dichloromethane (50 mL) were added to a round bottom flask. The reaction was stirred at room temperature for 30 minutes. The completion of the reaction was detected by LC-MS. 100 mL of dichloromethane was added to the reaction, and the organic phase was washed with 30 mL of hydrochloric acid (1M) and 100 mL of the saturated sodium bicarbonate aqueous solution in sequence. The organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (806 mg, 80%). ES-API: [M+H]+=605.4.


Step 6: tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (806 mg, 1.34 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (353 mg, 2.01 mmol), SPhos (55 mg, 0.134 mmol), SPhos-Pd-G2 (96 mg, 0.134 mmol), potassium phosphate (852 mg, 4.02 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, the organic phase was dried and concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (13aS)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (395 mg, 45%), a yellow solid. ES-API: [M+H]+=657.1.


Step 7: tert-butyl (13aS)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (395 mg, 0.6 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (13aS)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (401 mg), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=557.1.


Step 8: (13aS)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (401 mg, 0.71 mmol), 6 mL of dichloromethane and triethylamine (718 mg, 7.1 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (72 mg, 0.57 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated to obtain Compound Z26.


Step 9: The crude product of Compound Z26 was purified by Preparative HPLC to obtain: Compound Z26-1 (108 mg, 24%), HPLC:retention time: 6.13/6.19 min, ES-API: [M+H]+=611.2. and Compound Z26-2 (104 mg, 24%), HPLC:retention time: 6.26/6.31 min, ES-API: [M+H]+=611.2.


Step 10: Compound Z26-2 (106 mg, 0.18 mmol) was resolved chirally (mobile phase: n-hexane-ethanol-40-60); column type: IB (250 mm*4.6 mm 5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z26-2-1 (35 mg, retention time: 6.824 min, purity: 100%, de value: 100%). ES-API: [M+H]+=611.2. and Compound Z26-2-2 ((31 mg, retention time: 9.384 min, purity: 100%, de value: 99%). ES-API: [M+H]+=611.2.


Example 27: Preparation of Compound Z27



embedded image


embedded image


Step 1: 2,4,6-trichloronicotinic acid (8 g, 35.5 mmoL) was dissolved in 150 mL of dichloromethane and cooled to 0° C., and oxalyl chloride (9.3 ml, 106.6 mmol) was added. After 30 minutes for the completion of the dropping, DMF (0.5 ml) was dropped, the reaction rised to room temperature and proceeded for 1 hour. After the completion of the reaction, the reaction solution was concentrated and diluted with 150 mL of dichloromethane, cooled to 0° C., 8 mL of ammonia water was slowly dropped, and the reaction was stirred at room temperature for 2 h. After the completion of the reaction, the reaction solution was concentrated and slurried with ethyl acetate, filtered to obtain the crude product: 2,4,6-Trichloronicotinamide 6 g (6.5 g, yield 82%). ES-API: [M+H]+=224.9.


Step 2: 2-isopropyl-4-methylpyridin-3-amine (4.4 g, 29.1 mmol) was dissolved in 80 mL of tetrahydrofuran, LiHMDS (106.4 ml, 1M) was dropped under the ice water bath under nitrogen protection, stirred for 30 minutes, 2,4,6-Trichloronicotinamide (6.5 g, 29.1 mmol) dissolved in 20 mL of tetrahydrofuran, was added to the above reaction solution, slowly rised to room temperature, and reacted for 2 hours, cooled to room temperature, and dilute hydrochloric acid was dropped to pH of 7-8. The reaction solution was extracted with ethyl acetate, and concentrated to obtain a gray solid, slurried with ethyl acetate, and filtered to obtain the crude product 4,6-dichloro-2-((2-isopropyl-4-methylpyridin-3-yl)amino)nicotinamide (1.77 g, yield 8%). ES-API: [M+H]+=339.0.


Step 3: 4,6-dichloro-2-((2-isopropyl-4-methylpyridin-3-yl)amino)nicotinamide (1.77 g, 5.24 mmoL) was dissolved in 30 ml of tetrahydrofuran, 60% NaH (3 g, 15.72 mmol) was added under the ice water bath, stirred for 15 minutes, CDI (0.63 g, 7.86 mmol) was added, the reaction proceeded under the ice water bath for 1 hour, raw material disappeared, the reaction solution was poured into ice water, pH was adjusted to 5-6 with dilute hydrochloric acid. The reaction solution was extracted with ethyl acetate, washed with water and the saturated brine in sequence, concentrated under reduced pressure to obtain 5,7-dichloro-1-(2-isopropyl-4-methylpyridin-3-yl)pyridine[2,3-d]pyrimidin-2,4 (1H,3H)-dione (1.72 g, yield 90%). ES-API: [MA-]E=365.0.


Step 4: tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (1.02 g, 4.72 mmoL) was dissolved in tetrahydrofuran, 60% NaH (0.567 g, 14.2 mm1) was added under the ice water bath, stirred for 10 minutes, 5,7-dichloro-1-(2-isopropyl-4-methylpyridin-3-yl)pyridine[2,3-d]pyrimidin-2,4 (1H,3H)-dione (1.72 g, 4.72 mmoL) was added, and stirred for 20 minutes. After the completion of the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate for 3 times, the organic phases was combined, dried and concentrated under reduced pressure, purified by column chromatography (methanol/dichloromethane: 0-10%) to obtain tert-butyl (S)-3-((((7-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydropyrido[2,3-d]pyrimidin-5-yl)oxy)methyl)piperazin-1-formate (1.93 g, yield 75%). ES-API: [M+1-1]+=545.2.


Step 5: tert-butyl (S)-3-((((7-chloro-4-hydroxy-1-(2-isopropyl-4-methylpyridin-3-yl)-2-oxo-1,2-dihydropyrido[2,3-d]pyrimidin-5-yl)oxy)methyl)piperazin-1-formate (1.93 g, 3.54 mmoL) was dissolved in DMF, PyBOP (9.2 g, 17.7 mmoL) and DBU (2.69 g, 17.7 mmoL) was dropped under the ice water bath, and the reaction proceeded at room temperature for 2 hours. The completion of the reaction was detected. The reaction solution was poured into ice water, extracted with ethyl acetate for 3 times, the organic phases was combined, dried and concentrated under reduced pressure, purified by column chromatography (methanol/dichloromethane: 0-10%) to obtain tert-butyl (S)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-3,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.745 g, yield 40%). ES-API: [M+H]+=527.2.


Step 6: tert-butyl (S)-2-chloro-12-(2-ethyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.745 g, 1.42 mmoL), (5-methyl-1H-indazol-4-yl)boronic acid (500 mg, 2.84 mmol), Pd(PPh3)4 (164 mg, 0.142 mmol) and potassium carbonate (588 mg, 4.26 mmol) was dissolved in 8 mL of dioxane and 2 mL of water, nitrogen was used for replacement, the reaction proceeded at 120° C. for 1.5 hours, cooled to room temperature, filtered, washed with water and the saturated brine, concentrated, column chromatography to obtain tert-butyl (S)-12-(2-isopropyl-4-methylpyridin-3-yl)-2-(5-methyl-1H-indazol-4-yl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (371 mg, yield 42%). ES-API: [M+H]+=623.3.


Step 7: tert-butyl (S)-12-(2-isopropyl-4-methylpyridin-3-yl)-2-(5-methyl-1H-indazol-4-yl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (371 mg, 0.6 mmoL) was dissolved in 10 mL of dichloromethane, 4 mL of trifluoroacetic acid was slowly dropped at room temperature, reacted for 5 hours, 10 mL of dichloromethane was added, concentrated under reduced pressure to obtain (S)-12-(2-isopropyl-4-methylpyridin-3-yl)-2-(5-methyl-1H-indazol-4-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (313 mg, the crude product), directly used in the next step. ES-API: [M+H]+=523.3


Step 8: (S)-12-(2-isopropyl-4-methylpyridin-3-yl)-2-(5-methyl-1H-indazol-4-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (313 mg, 0.6 mmol) was dissolved in dichloromethane, triethylamine (303 mg, 3 mmol) and acrylic anhydride (60 mg, 0.48 mmol) was dropped under the ice water bath, and stirred for 10 minutes under the ice bath. after the completion of the reaction, the reaction solution was concentrated at room temperature under reduced pressure, and purified by preparative HPLC to obtain Compound Z27 (6.05 mg, yield 1.3%), ES-API: [M+H]+=577.2.


Example 28: Preparation of Compound Z28



embedded image


Step 1: tert-butyl 10-bromo-8-cyclopropyl-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylate (100 mg, 0.2 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (48 mg, 0.3 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (15 mg, 0.02 mmol), SPhos (8.4 mg, 0.02 mmol), potassium phosphate (130 mg, 0.6 mol), 10 mL of dioxane and 2 mL of water were added to a 100 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction proceeded at 110° C. for 2 hours. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl 8-cyclopropyl-10-(2-fluoro-6-hydroxyphenyl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylate (100 mg, yield: 93%). ES-API: [M+H]+=523.2.


Step 2: tert-butyl 8-cyclopropyl-10-(2-fluoro-6-hydroxyphenyl)-7-oxo-1,3,4,7,8,13,14,14a-octahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-2-carboxylate (100 mg, 0.19 mol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction solution was concentrated to obtain the target product: 8-cyclopropyl-10-(2-fluoro-6-hydroxyphenyl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one (70 mg, the crude product), was directly used in the next step. ES-API: [M+H]+=443.3.


Step 3: 8-cyclopropyl-10-(2-fluoro-6-hydroxyphenyl)-1,3,4,13,14,14a-hexahydro-2H-pyrazino[1′,2′:5,6][1,5]oxazolidine[4,3,2-de]quinazolin-7 (8H)-one (70 mg, 0.165 mmol) was dissolved in dichloromethane (15 mL), triethylamine (50 mg, 0.5 mmol) was added. The reaction was cooled to 0° C., and acrylic chloride (21 mg, 0.016 mol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 40 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 50 mL of the saturated NaHCO3 aqueous solution, 40 mL of the saturated brine, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: Z28 (30 mg, yield: 38%). ES-API: [M+H]+=477.1. 1H NMR (500 MHz, DMSO-d6) δ 10.07 (s, 1H), 7.36-7.12 (m, 2H), 6.98-6.63 (m, 4H), 6.17 (m, J=14.3 Hz, 1H), 5.74 (d, J=9.9 Hz, 1H), 4.71 (d, J=33.4 Hz, 1H), 4.41-3.89 (m, 5H), 3.57 (d, J=13.6 Hz, 0.5H), 3.31-3.19 (m, 2H), 3.03 (s, 0.5H), 2.64 (s, 1H), 2.13-1.80 (m, 2H), 1.18-0.99 (m, 2H), 0.78-0.44 (m, 2H). ES-API: [M+H]+=477.1.


Example 29: Preparation of Compound Z29, Z29-1, Z29-2, Z29-3 and Z29-4



embedded image


embedded image


embedded image


Step 1: tert-butyl (R)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (149 mg, 0.25 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (59 mg, 0.38 mmol), SPhos (10 mg, 0.025 mmol), SPhos-Pd-G2 (18 mg, 0.025 mmol), potassium phosphate (159 mg, 0.75 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (13aR)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (111 mg, 70%), a yellow solid. ES-API: [M+H]+=637.1.


Step 2: Tert-butyl (13aR)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (111 mg, 0.18 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (13aR)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4 -methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (121 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=537.1.


Step 3: (13aR)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (121 mg, 0.21 mmol), 6 mL of dichloromethane and triethylamine (212 mg, 2.1 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (21 mg, 0.168 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated. The crude product was purified by Preparative HPLC to obtain the target product: Z29 (28 mg, 23%). ES-API: [M+H]+=591.1.


Step 4: the above obtained Compound Z29 (28 mg, 0.31 mmol) was resolved chirally (mobile phase: n-hexane:ethanol:ammonia methanol=60:40:0.2; column type: IE (250 mm*4.6 mm*5 um); flow rate: 1 ml/min; column temperature: 30° C.) to obtain:


Compound Z29-1 (6 mg, retention time: 9.598 min, purity: 100%, de value: 98%). ES-API: [M+H]+=591.1.


Compound Z29-2 (5.5 mg, retention time: 10.377 min, purity: 100.0%, de value: 97.1%). ES-API: [M+H]+=591.1.


Compound Z29-3 (4.3 mg, retention time: 12.512 min, purity: 99.1%, de value: 97.5%). ES-API: [M+H]+=591.1.


Compound Z29-4 (4.2 mg, retention time: 13.741 min, purity: 98.6%, de value: 98.2%). ES-API: [M+H]+=591.1.


Example 30: Preparation of Compound Z30, Z30-1, Z30-2, Z30-2-1 and Z30-2-2



embedded image


embedded image


Step 1: tert-butyl (R)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (500 mg, 0.83 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (224 mg, 1.27 mmol), SPhos (34 mg, 0.084 mmol), SPhos-Pd-G2 (60 mg, 0.084 mmol), potassium phosphate (535 mg, 2.52 mmol), 60 mL of dioxane and 12 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 115° C. for 2 hours under nitrogen protection, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (13aR)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (418 mg, 76%), a yellow solid. ES-API: [M+H]+=656.1.


Step 2: tert-butyl (13aR)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (418 mg, 0.64 mmol), 3 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (13aR)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (501 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=556.1.


Step 3: (13aR)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (501 mg, 0.9 mmol), 6 mL of dichloromethane and triethylamine (450 mg, 4.5 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (91 mg, 0.72 mmol, 1 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 5 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain the target product: Z30 (188 mg, 34%). ES-API: [M+H]+=611.2.


Step 4: Compound Z30 was purified by Preparative HPLC to obtain: Compound Z30-1 (188 mg, 34%), HPLC:retention time: 7.36/7.41 min, ES-API: [M+H]+=611.2. and Compound Z30-2 (88 mg, 16%), HPLC:retention time: 7.45/7.51 min, ES-API: [M+H]+=611.2.


Step 5: Compound Z30-2 (88 mg, 0.14 mmol) was resolved chirally (mobile phase: n-heptane:ethanol (0.1% ammonia methanol)=70-30); column type: AS-H (inner diameter: 0.46 cm, length: 15 cm); flow rate: 0.5 ml/min; column temperature: 25° C.) to obtain: Compound Z30-2-1 (36 mg, retention time: 4.612 min, purity: 100%, de value: 98%). ES-API: [M+H]+=611.2; and Compound Z30-2-2 ((31 mg, retention time: 4.861 min, purity: 100%, de value: 98%). ES-API: [M+H]+=611.2.


Example 31: Preparation of Compound Z31



embedded image


Compound Z31 was prepared with tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate as the raw material according to the method of Example 29. ES-API: [M+H]+=591.1. 1H NMR (500 MHz, DMSO-d6), δ 10.02 (d, J=2.2 Hz, 1H), 8.48 (dd, J=4.8, 2.5 Hz, 1H), 7.30-7.26 (m, 1H), 7.23-7.16 (m, 1H), 6.97-6.81 (m, 1H), 6.75-6.55 (m, 2H), 6.21 (d, J=16.1 Hz, 1H), 5.83 (dd, J=7.4, 4.1 Hz, 1H), 5.77 (d, J=12.5 Hz, 1H), 4.75 (s, 3H), 4.55-4.00 (m, 3H), 3.54 (d, J=12.4 Hz, 1H), 3.14 (s, 1H), 2.88-2.65 (m, 1H), 1.96 (dd, J=14.7, 8.8 Hz, 3H), 1.14-0.90 (m, 6H).


Example 32: Preparation of Compound Z32



embedded image


embedded image


Step 1: 4-bromo-2,6-difluorobenzonitrile (10 g, 45.87 mmol) was added to isopropanol (200 mL), then 2-methylprop-1-amine (5 g, 68.8 mmol) was added, and the reaction solution was heated to 50° C. to react for two hours. The completion of the reaction was detected by LCMS, iPrOH was spin-dried, 100 mL of water was added. The reaction solution was extracted with ethyl acetate (100 mL*3), and the extracted organic phase was spin-dried to obtain the crude product, and the crude product was passed through a fast column (120 g silica gel column, ethyl acetate/petroleum ether=20/1-10/1) to obtain the product: 4-bromo-2-fluoro-6-(isobutylamino)benzonitrile (12.7 g, yield: 96.7%). ES-API: [M+1]+=271.0, 273.0.


Step 2: 4-bromo-2-fluoro-6-(isobutylamino)benzonitrile (10 g, 37 mmol), was dissolved in isopropanol (100 mL), NCS (5.4 g, 40.7 mmol) was added, the reaction solution was heated to 50° C. to react for 3 hours. The completion of the reaction was detected by LCMS, iPrOH was spin-dried, 100 mL of water was added, then the reaction solution was extracted with ethyl acetate (100 mL*3), and the extracted organic phase was spin-dried to obtain the crude product, and the crude product was passed through a fast column (120 g silica gel column, ethyl acetate/petroleum ether=20/110/1) to obtain the product: 4-bromo-3-chloro-2-fluoro-6-(isobutylamino)benzonitrile (3.3 g, yield: 33%). ES-API: [M+1]+=304.9, 306.9.


Step 3: 4-bromo-3-chloro-2-fluoro-6-(isobutylamino)benzonitrile (3.3 g, 10.8 mmol) was dissolved in DMSO (50 mL), then potassium carbonate (2.8 g, 20.4 mmol) was added, hydrogen peroxide (1.3 g, 37 mmol) was slowly dropped at room temperature, the reaction solution was stirred for 3 hours at room temperature. The completion of the reaction was detected by LCMS. Sodium sulfite (5 g, 39.7 mmol) was added and stirred for 30 minutes, and then water (100 mL) was added. The reaction solution was extracted with ethyl acetate (100 mL*3), the extracted organic phase was spin-dried to obtain the crude product, and the crude product was passed through a fast column (40 g silica gel column, ethyl acetate/petroleum ether=10/1-5/1) to obtain the product: 4-bromo-3-chloro-2-fluoro-6-(isobutylamino)benzamide (3.3 g, yield: 73%). ES-API: [M+1]+=322.9, 324.9.


Step 4: NaH (1.6 g, 66 mmol) was dissolved in tetrahydrofuran (40 mL) and cooled to 0° C., then 4-bromo-3-chloro-2-fluoro-6-(isobutylamino)benzamide (2.1 g, 6.6 mmol) dissolved in 10 mL of tetrahydrofuran was slowly dropped to the reaction solution, stirred for 1 hour at 0° C., then N,N′-carbonyl diimidazole (1.4 g, 8.58 mmol) dissolved in 10 mL of tetrahydrofuran was slowly dropped to the reaction solution at 0° C., then naturally warmed up to room temperature, stirred for 30 minutes. The detection of LCMS showed that the product and raw materials are about 1/1, and the detection of LCMS showed no obvious change after 30 minutes. 5 mL of the saturated ammonium chloride solution was added to quench excess NaH, then 20 mL of water was added. The reaction solution was extracted with ethyl acetate (50 mL*3), the extracted organic phase was spin-dried to obtain the crude product, and the crude product (ethyl acetate/petroleum ether=3/1) was slurried to obtain the products: 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-isobutylquinazolin-2 (1H)-one (1.3 g, yield: 92.5%) and 4-bromo-3-chloro-2-fluoro-6-(isobutylamino)benzamide (900 mg). ES-API: [M+1]+=348.9, 350.9.


Step 5: 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-isobutylquinazolin-2 (1H)-one (1.2 g, 3.4 mmol), tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (1.1 g, 5.1 mmol) was dissolved in tetrahydrofuran (20 mL), then NaH (410 mg, 17 mmol) was slowly added to the reaction solution, stirred at room temperature for 30 minutes, the detection of LC-MS showed the completion of the reaction, 5 mL of the saturated ammonium chloride solution was added to quench the reaction, then 20 mL of water was added, extracted with ethyl acetate (30 mL*3), the extracted organic phase was spin-dried to obtain the crude product, and the crude product was passed through a fast column (20 g silica gel column, dichloromethane/methanol=100/1) to obtain the product: tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-1-isobutyl-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.3 g, yield: 72%). ES-API: [M+1]+=545.1, 547.1.


Step 6: Tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-1-isobutyl-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (1.2 g, 2.2 mmol) was dissolved in N,N-dimethylformamide (20 mL), 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (3.45 g, 6.6 mmol) was added, 1,8-diazabicyclo[5.4.0]undec-7-ene (1.67 g, 11 mmol) was slowly dropped under the ice bath, then heated to room temperature and stirred for 2 hours, the detection of LC-MS showed the completion of the reaction, 30 mL of water was added, extracted with ethyl acetate (30 mL*3), the extracted organic phase was spin-dried to obtain the crude product, and the crude product was passed through a fast column (20 g silica gel column, dichloromethane/methanol=20/1) to obtain the product: tert-butyl (S)-10-bromo-11-chloro-8-isobutyl-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.2 g, yield: 96.7%). ES-API: [M+1]+=527.1, 529.1.


Step 7: tert-butyl (S)-10-bromo-11-chloro-8-isobutyl-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (530 mg, 1.0 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (312 mg, 2.0 mmol), tripotassium phosphate (636 mg, 6.0 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (72 mg, 0.1 mmol), 2-bicyclohexylphosphin-2′,4′,6′-triisopropylbiphenyl(40 mg, 0.1 mmol) were added to dioxane (8 mL) and water (2 mL) in sequence, nitrogen was used for replacement for three times, heated to 100° C. to react for 16 hours. After cooling to room temperature, the reaction solution was poured into ethyl acetate (30 mL), washed once with brine and purified by a silica gel column (12 g silica gel column, methanol:dichloromethane=0-1:20) to obtain a yellow foaming solid: tert-butyl (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-isobutyl-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2(1H)-carboxylate (370 mg, yield 60%). ES-API: [M+1]+=559.2.


Step 8: tert-butyl (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-isobutyl-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (370 mg, 0.67 mmol) was dissolved in dichloromethane (5 mL), trifluoroacetic acid (2 mL) was added, and reacted at room temperature for 1 hour, spin-dried to obtain a yellow oily crude product: (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-isobutyl-1,2,3,4,13,13a hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (300 mg, yield 100%).


Step 9: (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-8-isobutyl1,2,3,4,13,13a hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (300 mg, 0.655 mmol), and triethylamine (2 mL) was dissolved in dichloromethane (5 mL), cooled to 0° C., acrylic anhydride (75 mg, 0.59 mmol) was added, and reacted at 0° C. for 0.5 hour. The reaction solution was extracted with 20 mL of water and dichloromethane (20 mL*3), the extracted organic phase was spin-dried to obtain the crude product, and the crude product was prepared and purified (Ultimate XB-C18,50*250 mm, 10 um, acetonitrile/water=10%/90%˜90%/10%, 40 minutes) and freeze-dried to obtain the product: Z32 (102 mg, yield 38%). ES-API: [M+1]+=513.1. 1H NMR (400 MHz, DMSO-d6) δ 10.07 (s, 1H), 7.26 (d, J=8.1 Hz, 1H), 7.05 (s, 1H), 6.88-6.71 (m, 3H), 6.17 (d, J=16.6 Hz, 1H), 5.73 (d, J=10.5 Hz, 1H), 4.60 (s, 2H), 4.52-4.23 (m, 3H), 4.10 (d, J=12.4 Hz, 1H), 3.94 (s, 3H), 3.42 (s, 1H), 3.16 (s, 1H), 3.00 (s, 1H), 2.03 (s, 1H), 0.84 (q, J=7.8 Hz, 6H).


Example 33: Preparation of Compound Z33



embedded image


embedded image


Step 1: 4-bromo-2,6-difluorobenzonitrile (10 g, 45.87 mmol) and tetrahydro-2H-pyran-4-amine (13.9 g, 137.6 mmol) was dissolved in i-PrOH (150 mL), and reacted at 70° C. for 16 hours. The solvent was concentrated to obtain a crude product, the crude product was washed with water (3×100 mL) to obtain 4-bromo-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzonitrile (13 g, 43.48 mmol, yield: 94.9%), a white solid. ES-API: [M+H]+=299.0.


Step 2: 4-bromo-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzonitrile (9 g, 30.1 mmol) was dissolved in DMF (80 mL), 1-chloropyrrolidin-2,5-dione (4.82 g, 31.62 mmol) was added in batches at 70° C. The resulting mixture reacted at 80° C. for 1 hour. After cooling to room temperature, water (50 mL) was added to form a solid. The solid was separated, and washed with water (3×30 mL) to obtain 4-bromo-3-chloro-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzonitrile (7.05 g, the crude product) a white solid. ES-API: [M+H]+=333.1.


Step 3: 4-bromo-3-chloro-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzonitrile (7 g, 20.99 mmol) was dissolved in DMSO (100 mL), then potassium carbonate (5.82 g, 42.2 mmol) was added, and finally hydrogen peroxide (8.61 g, 75.95 mmol) was dropped at 0° C. Then, the resulting mixture was stirred at room temperature for 2 hours. The mixture was poured into ice water (50 mL), and extracted with ethyl acetate (3×200 mL). The organic layer was washed with brine (3×200 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and the crude product was purified by a silica gel column (petroleum ether/ethyl acetate=4:10) to obtain 4-bromo-3-chloro-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzamide (3.6 g, yield: 48.78%), a white solid. ES-API: [M+H]+=351.0.


Step 4: 4-bromo-3-chloro-2-fluoro-6-((tetrahydro-2H-pyran-4-yl)amino)benzamide (3.6 g, 10.24 mmol) was dissolved in tetrahydrofuran (50 mL), NaH (2.5 g, 61.44 mmol) was added at 0° C. in batches, and reacted at 0° C. for 0.5 hour. Then a solution of CDI (2.5 g, 15.36 mmol) in tetrahydrofuran (10 mL) was dropped at 0° C., and finally the resulting mixture was stirred at 0° C. for 2 hours. Ethyl acetate (100 mL) was added to the reaction to dilute, and the product was extracted with ethyl acetate (3×200 mL). The organic layer was washed with brine (2×100 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and the crude product was washed (petroleum ether/ethyl acetate=10:1) to obtain 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(tetrahydro-2H-pyran-4-yl)quinazolin-2 (1H)-one (3.5 g, yield: 90%), a white solid. ES-API: [M+H]+=377.0.


Step 5: NaH (1.8 g, 45 mmol) was added to a solution of tert-butyl (S)-3-(hydroxymethyl)piperazin-1-formate (2.92 g, 13.5 mmol) in THF (60 mL) at 0° C. in batches, the resulting mixture was stirred at 0° C. for 0.5 hour. Then, 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(tetrahydro-2H-pyran-4-yl)quinazolin-2 (1H)-one (3.4 g, 9 mmol) was added at 0° C., and the resulting mixture was stirred at 0° C. to room temperature for 2 hours. Ethyl acetate (100 mL) was added to the reaction, and the product was extracted with ethyl acetate (3×200 mL). The organic layer was washed with brine (2×100 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and the crude product was washed (ethyl acetate/petroleum ether=1:3) to obtain tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-2-oxo-1-(tetrahydro-2H-pyran-4-yl)-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (4.2 g, yield=81%), a white solid. ES-API: [M+H]+=573.2.


Step 6: tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-2-oxo-1-(tetrahydro-2H-pyran-4-yl)-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (2 g, 3.48 mmol) was dissolved in (30 mL), then carter condensing agent (3.86 g, 8.71 mmol) was added. 1,8-diazabicycloundec-7-ene (2.65 g, 17.42 mmol) was dropped at 0° C., and the resulting mixture was stirred at 0° C. to room temperature for 2 hours. Ethyl acetate (100 mL) was added to the reaction, and the product was extracted with ethyl acetate (3×100 mL). The organic layer was washed with brine (2×100 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and the crude product was purified by a silica gel column (ethyl acetate/petroleum ether=7:3) to obtain tert-butyl (S)-10-bromo-11-chloro-7-oxo-8-(tetrahydro-2H-pyran-4-yl)-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.68 g, yield: 86.6%), a white solid. ES-API: [M+H]+=555.0.


Step 7: tert-butyl (S)-10-bromo-11-chloro-7-oxo-8-(tetrahydro-2H-pyran-4-yl)-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.3 g, 2.34 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (548 mg, 3.51 mmol), potassium phosphate (1.5 g, 7.02 mmol), SPhos (192 mg, 0.468 mmol) and SPhos-Pd-G2 (166 mg, 0.234 mmol) was dissolved in dioxane/water (20 mL, 5:1) under nitrogen protection, and reacted at 110° C. for 2 hours. After cooling to room temperature, the product was extracted with ethyl acetate (3×80 mL). The organic layer was washed with brine (3×30 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and crude product was purified by rapid elution (dichloromethane/methanol=5:1) to obtain tert-butyl (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-7-oxo-8-(tetrahydro-2H-pyran-4-yl)-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.37 g, yield: 99%), a white solid. ES-API: [M+H]+=587.2.


Step 8: tert-butyl (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-7-oxo-8-(tetrahydro-2H-pyran-4-yl)-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (500 mg, 0.85 mmol) was dissolved in dichloromethane (6.0 mL) and trifluoroacetic acid (2.0 mL), and reacted at room temperature for 1 hour. The solvent was concentrated to obtain a crude product (611 mg, a brown oil), it can be used in the next step without further purification. ES-API: [M+H]+=487.1.


Step 9: tert-butyl (13aS)-11-chloro-10-(2-fluoro-6-hydroxyphenyl)-7-oxo-8-(tetrahydro-2H-pyran-4-yl)-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (414 mg, 0.85 mmol) and triethylamine (430 mg, 4.25 mmol) was dissolved in dichloromethane (10 mL). Acrylic anhydride (107 mg, 0.85 mmol) in dichloromethane (1 mL) was dropped at 0° C., then the resulting mixture was stirred at 0° C. for 0.5 hour. 10 mL of sodium bicarbonate was added, and the product was extracted with dichloromethane (3×30 mL). The organic layer was washed with sodium bicarbonate (3×10 mL), dried with anhydrous sodium sulfate and concentrated to obtain a crude product, and the crude product was purified by a reverse column (C18 spherical 20-35 um 100 A) with rapid elution of acetonitrile and water (0-0, 5 min; 0-45%, 20 min; 45%-45%, 7 min) to obtain Compound Z33 (127.4 mg, yield: 27%), a white solid. ES-API: [M+H]+=541.0. 1H NMR (400 MHz, DMSO-d6) δ 2.64-2.74 (m, 3H), 3.11 (s, 1H), 3.40-3.50 (m, 4H), 3.85-4.04 (m, 4H), 4.18-4.55 (m, 6H), 5.73 (d, J=10.2 Hz, 1H), 6.16 (d, J=16.6 Hz, 1H), 6.69-6.83 (m, 3H), 7.22-7.33 (m, 2H), 10.06 (s, 1H).


Example 34: Preparation of Compound Z34



embedded image


Step 1: tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.1 g, 1.82 mmol), 3-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyrid-2-amine (600 mg, 2.73 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloride (133 mg, 0.182 mmol), potassium acetate (535 mg, 5.46 mmol) was dissolved in dioxane/water (15/3 mL), heated to 100° C. and reacted for 2 hours under nitrogen protection. The reaction solution was cooled to room temperature, water, ethyl acetate were added and separated, the organic phase was washed with brine, dried with anhydrous sodium sulfate, and spin-dried to obtain the crude product: tert-butyl (S)-10-(2-aminopyridin-3-yl)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxheptidine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.5 g, yield: 100%). ES-API: [M+H]+=618.2.


Step 2: tert-butyl (S)-10-(2-aminopyridin-3-yl)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxheptidine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.5 g, 1.82 mmol) was dissolved in dichloromethane (10 mL), trifluoroacetic acid (5 mL) was added, the reaction solution was stirred at room temperature for 0.5 hour, and the solvent was concentrated to dryness under reduced pressure to obtain the crude product: (S)-10-(2-aminopyrid-3-yl)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (1.5 g, yield: 100%). ES-API: [M+H]+=518.2.


Step 3: (S)-10-(2-aminopyridin-3-yl)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (1.5 g, 1.82 mmol) was dissolved in dichloromethane (100 mL), the reaction solution was cooled to 0° C., triethylamine (552 mg, 5.46 mmol) was added, acrylic anhydride (183 mg, 1.46 mmol) was dropped, and reacted at 0° C. for 0.5 hour. Water was added to the reaction solution to quench the reaction, and the organic phase was washed with the saturated brine, dried with anhydrous magnesium sulfate, spin-dried, prepared by chromatography and purified, chromatographic column: Ultimate XB-C18,50*250 mm, 10 um, mobile phase: acetonitrile/water=10/90-90/10, 40 min to obtain Compound Z34 (218 mg, yield: 21.0%). ES-API: [M+H]+=572.2. 1H NMR (400 MHz, DMSO-d6) δ 8.46 (d, J=4.7 Hz, 1H), 7.90 (s, 1H), 7.26 (s, 1H), 6.87 (s, 1H), 6.51 (s, 1H), 6.19 (d, J=17.1 Hz, 1H), 5.76 (d, J=11.8 Hz, 2H), 5.60 (s, 1H), 5.53 (d, J=10.0 Hz, 1H), 4.69 (s, 3H), 4.55-4.24 (m, 1H), 4.08 (d, J=43.0 Hz, 1H), 3.15 (s, 1H), 2.76 (s, 1H), 1.96 (d, J=12.4 Hz, 3H), 1.06 (d, J=8.8 Hz, 3H), 0.98 (d, J=6.5 Hz, 3H).


Example 35: Preparation of Compound Z35



embedded image


Step 1: tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (60 mg, 0.1 mmol), 3-fluoro-2-(tributyltinalkyl)pyridine (58 mg, 0.15 mmol), tetra(triphenylphosphine)palladium (12 mg, 0.01 mmol), lithium chloride (13 mg, 0.3 mmol), cuprous iodide (2 mg, 0.01 mmol) was dissolved in dioxane (1.5 mL), heated to 100° C. and reacted for 5 hours under nitrogen protection. The reaction solution was cooled to room temperature, water, ethyl acetate was added and separated, the organic phase was washed with brine, dried with anhydrous sodium sulfate, spin-dried, purified by thin layer chromatography (ethyl acetate: 100%) to obtain tert-butyl (S)-11-chloro-10-(3-fluoropyridin-2-yl)-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (30 mg, yield: 48.4%). ES-API: [M+H]+=621.2.


Steps 2-3: Compound Z35 was prepared according to the corresponding steps of Example 34. ES-API: [M+H]+=575.2. 1H NMR (400 MHz, DMSO-d6) δ 8.45 (d, J=14.2 Hz, 2H), 7.81 (s, 1H), 7.54 (s, 1H), 7.27 (s, 1H), 6.86 (s, OH), 6.19 (d, J=16.5 Hz, 1H), 5.99 (s, 1H), 5.75 (d, J=10.7 Hz, 1H), 4.74 (s, 2H), 4.33 (s, OH), 4.12 (s, 1H), 3.54 (s, 2H), 3.14 (s, 1H), 2.76 (s, 1H), 1.95 (d, J=10.5 Hz, 3H), 1.05 (d, J=6.6 Hz, 3H), 0.99 (s, 3H).


Example 36: Preparation of Compound Z36, Z36-1 and Z36-2



embedded image


embedded image


Step 1: tert-butyl (S)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (900 mg, 1.7 mmol), 2-fluoro-6-hydroxyphenylboronic acid (400 mg, 2.6 mmol), SPhos (53 mg, 0.13 mmol), SPhos-Pd-G2 (98 mg, 0.13 mmol), potassium phosphate (1.1 g, 5.1 mmol), 12 mL of dioxane and 3 mL of water were added to a reaction flask. The reaction was stirred under a 120° C. oil bath for 1 hour under nitrogen protection. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-2%) to obtain tert-butyl (S)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, purity 80%). ES-API: [M+H]+=603 0.2.


Step 2: tert-butyl (S)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, 1.66 mmol), 5 mL of methanol and 5 mL of hydrogen chloride/dioxane solution (4 M) were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain the crude product: (S)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a, 6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (835 mg), a yellow solid. The crude product was directly used in the next step. ES-API: [M+H]+=503.2.


Step 3: (S)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a, 6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (835 mg, 1.66 mmol), 15 mL of dichloromethane and triethylamine (545 mg, 5.4 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (188 mg, 1.49 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 30 mL of dichloromethane twice. The organic phase was dried and concentrated, and the crude product was purified by Preparative HPLC to obtain Z36 (350 mg, 35%), a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.88-11.85 (m, 1H), 8.70 (s, 1H), 7.58-7.57 (m, 1H), 7.26-7.25 (m, 1H), 7.18-7.17 (m, 1H), 6.61-6.57 (m, 3H), 6.46-6.42 (m, 1H), 5.86-5.84 (m, 1H), 5.07-5.05 (m, 1H), 4.58-4.50 (m, 3H), 4.11-3.40 (m, 5H), 2.78-2.77 (m, 1H), 2.16-2.15 (m, 3H), 1.92-1.90 (m, 3H), 1.13-1.08 (m, 6H). ES-API: [M+H]+=557 0.2.


Step 4: Compound Z36 (350 mg) was resolved by a chiral column (column type: Chiralpak IB 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol:ammonia methanol=50:50:0.2; flow rate: 1 ml/min; time=20 min) to obtain: Compound Z36-1 (121.5 mg, retention time: 8.77 min, peak 1), a white solid. HNMR (400 MHz, CDCl3) 11.84 (s, 1H), 8.72-8.71 (m, 1H), 7.57 (s, 1H), 7.27-7.20 (m, 2H), 6.69-6.43 (m, 4H), 5.87-5.84 (m, 1H), 5.02-5.00 (m, 1H), 4.57-4.50 (m, 3H), 4.13-3.67 (m, 4H), 3.40-3.38 (m, 1H), 2.85-2.83 (m, 1H), 2.16 (s, 3H), 1.28-1.17 (m, 6H). ES-API: [M+H]+=557.2; and Compound Z36-2 (130 mg; retention time: 11.81 min, peak 2), a white solid. HNMR (400 MHz, CDCl3) 11.89 (s, 1H), 8.70-8.69 (m, 1H), 7.57 (s, 1H), 7.27-7.20 (m, 2H), 6.69-6.43 (m, 4H), 5.87-5.84 (m, 1H), 5.08-5.05 (m, 1H), 4.57-4.50 (m, 3H), 4.08-3.65 (m, 4H), 3.40-3.39 (m, 1H), 2.78-2.76 (m, 1H), 2.15 (s, 3H), 1.28-1.10 (m, 6H). ES-API: [M+H]+=557.2.


Example 37: Preparation of Compound Z37, Z37-1 and Z37-2



embedded image


Compound Z37 was prepared with 2-cyclopropylmethyl-1-amine as the raw material according to the method of Example 32. The crude product of Compound Z37 was separated by a silica gel column (the eluent is petroleum ether/ethyl acetate/methanol=10/20/1-5/10/1), the less polar Compound Z37-1 was collected first (TLC Rf value 0.32 (developing agent is petroleum ether/ethyl acetate/methanol=5/10/1), HPLC:retention time 9.686 min, 17 mg, yield 8%), ES-API: [M-FH]+=511.1. 1H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 7.27 (d, J=8.1 Hz, 1H), 7.15 (s, 1H), 6.92-6.67 (m, 3H), 6.16 (d, J=16.8 Hz, 1H), 5.74 (s, 1H), 4.61 (s, 2H), 4.52-4.16 (m, 3H), 4.16-3.73 (m, 4H), 3.52-3.38 (m, 2H), 1.13 (d, J=18.8 Hz, 1H), 0.38 (d, J=7.4 Hz, 4H).


The more polar Compound Z37-2 was collected later (TLC Rf value 0.36 (developing agent is petroleum ether/ethyl acetate/methanol=5/10/1), HPLC:retention time 9.767 min, 18 mg, yield 8%), ES-API: [M-FH]+=511.1. 1H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 7.27 (d, J=8.3 Hz, 1H), 7.15 (s, 1H), 6.93-6.68 (m, 3H), 6.17 (d, J=16.4 Hz, 1H), 5.73 (d, J=9.9 Hz, 1H), 4.74-4.22 (m, 5H), 4.18-3.92 (m, 4H), 3.25-2.97 (m, 2H), 1.10 (s, 1H), 0.38 (d, J=7.8 Hz, 4H).


Example 38: Preparation of Compound Z38



embedded image


Step 1: tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.1 g, 1.82 mmol), (1-methyl-2-oxo-1,2-dihydropyridin-3-yl)boronic acid (418 g, 2.73 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (133 mg, 0.182 mmol), potassium acetate (535 mg, 5.46 mmol) was dissolved in dioxane/water (15/5 mL), heated to 100° C. and reacted for 2 hours under nitrogen protection. The reaction solution was cooled to room temperature, water, ethyl acetate were added and separated, the organic phase was washed with brine, dried with anhydrous sodium sulfate, spin-dried to obtain the crude product: tert-butyl (S)-11-chloro-8-(2-isopropyl-4-methylpyrid-3-yl)-10-(1-methyl-2-oxo-1,2-dihydropyrid-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.5 g, yield: 100%). ES-API: [M+H]+=633.2.


Steps 2-3: Compound Z38 was prepared according to the corresponding steps of Example 34. A chromatographic column (Ultimate XB-C18,50*250 mm, 10 um, mobile phase: acetonitrile/water=10/90-90/10.40 min) was used for purification. ES-API: [M+H]+=587.2. 1H NMR (400 MHz, DMSO-d6) δ8.50 (d, J=4.8 Hz, 1H), 7.69 (d, J=6.8 Hz, 1H), 7.29 (d, J=4.7 Hz, 1H), 6.87 (q, J=15.2 Hz, 1H), 6.18 (d, J=16.7 Hz, 1H), 6.13-6.02 (m, 2H), 5.81-5.71 (m, 2H), 4.72 (s, 3H), 4.34 (s, 1H), 4.08 (d, J=11.1 Hz, 1H), 3.50 (s, 1H), 3.37 (s, 3H), 3.08 (d, J=11.8 Hz, 2H), 2.70 (dt, J=31.2, 6.9 Hz, 1H), 1.96 (d, J=9.0 Hz, 3H), 1.12-0.90 (m, 6H).


Example 39: Preparation of Compound Z39



embedded image


embedded image


embedded image


Step 1: sodium bis(trimethylsilyl)amide (42 mL) was dropped to a mixed solution of 4-bromo-2,6-difluorobenzonitrile (8.0 g, 36.70 mmol) and 2-isopropylpyridin-3-amine (5.0 g, 36.70 mmol) in tetrahydrofuran (50 mL) at −65° C., and maintained for 0.5 hour at −60° C. Tetrahydrofuran (100 mL) was added when the temperature rised to −50° C. Then the mixture slowly rised to room temperature and stirred for 3 hours. The reaction mixture was poured into ice water (20 mL) and extracted with dichloromethane (3×100 mL). the organic phases was combined, washed with the saturated brine (3×50 mL), and dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 80 g silica gel column, petroleum ether/ethyl acetate=9% to obtain 4-bromo-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzonitrile (8.0 g, yield: 65.23%), a white solid. ES-API: [M+1]+=334.0.


Step 2: acetonitrile (75 mL) and deionized water (225 mL) were added to a 500 mL round bottom flask containing 4-bromo-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzonitrile (10.0 g, 29.92 mmol), sodium chloride (3.5 g, 59.85 mmol), potassium peroxymonosulfonate (13.79 g, 22.44 mmol). The reaction mixture was heated to 70° C. in an air atmosphere overnight, potassium peroxymonosulfonate (10.0 g, 16.27 mmol) was additional added, then heated to 80° C. and continued to react for 6 hours. The reaction solution was extracted with dichloromethane (3×150 mL). The organic phases was combined, washed with the saturated brine (3×50 mL), and dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 120 g silica gel column, petroleum ether and ethyl acetate (3%) to obtain 4-bromo-3-chloro-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzonitrile (2.0 g, yield: 18.13%), a white solid. ES-API: [M+1]+=368.0.


Step 3: hydrogen peroxide solution (2.3 g, ω=30%) was dropped to a mixed solution of 4-bromo-3-chloro-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzonitrile (2.0 g, 5.43 mmol) and potassium carbonate (1.5 g, 10.85 mmol) in dimethyl sulfoxide (20 mL) at 0° C., and after the completion of the dropping, the reaction proceeded at room temperature for 2 hours. 10% sodium thiosulfate solution (35 mL) was added at 0° C., then the mixture was poured into ice water (200 mL) and filtered with suction to obtain a filter cake, and vacuum dried to obtain a white solid of 4-bromo-3-chloro-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzamide (1.5 g, yield: 71.51%). ES-API: [M+1]+=386.0.


Step 4: sodium hydride (0.685 g, ω=60%) was added to a mixed solution of 4-bromo-3-chloro-2-fluoro-6-((2-isopropylpyridin-3-yl)amino)benzamide (1.32 5 g, 3.43 mmol) in tetrahydrofuran (10 mL) at 0° C., and after the completion of the addition, reacted at 0° C. for 1 hour. Carbonyl diimidazole (0.834 g, 5.14 mmol) dissolved in tetrahydrofuran (10 mL) was dropped to the above mixture, and maintained 0° C. After the completion of the dropping, the reaction mixture rised to room temperature and reacted for 2 hours. The mixture was poured into ice water (150 mL) and extracted with dichloromethane (3×100 mL). The organic phases was combined, washed with the saturated ammonium chloride (3×50 mL) and the saturated brine (3×50 mL), dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 40 g silica gel column, dichloromethane and methanol (3%) to obtain 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylpyridin-3-yl)quinazolin-2(1H)-one (700 mg, 49.50%), a white solid. ES-API: [M+1]+=412.0.


Step 5: sodium hydride (0.34 g, ω=60%) was added to a mixed solution of 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(2-isopropylpyridin-3-yl)quinazolin-2(1H)-one (0.7 g, 1.7 mmol) and tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (0.4 g, 1.87 mmol) in tetrahydrofuran (10 mL) at 0° C., and after the completion of the addition, reacted at 0° C. for 1 hour. The mixture was poured into ice water (100 mL) and extracted with dichloromethane (3×100 mL). The organic phases was combined, washed with the saturated ammonium chloride (3×30 mL) and the saturated brine (3×50 mL), dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 12 g silica gel column, dichloromethane and methanol (3%) to obtain tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (0.75 g, yield: 72.61%), a white solid. ES-API: [M+1]+=608.1.


Step 6: 1H-benzotriazol-1-yloxytripyrrolidinyl hexafluorophosphate (1.54 g, 2.96 mmol) was added to a mixed solution of tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-1-(2-isopropylpyridin-3-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (0.6 g, 0.985 mmol in N,N′-dimethylformamide (10 mL) at 0° C., and then 1,8-diazabicyclo[5.4.0]undec-7-ene (0.75 g, 4.93 mmol) was dropped, and after addition was completed, transferred to room temperature immediately and reacted for 1 hour. The mixture was poured into ice water (100 mL) and extracted with ethyl acetate (3×30 mL). The organic phases was combined, washed with the saturated ammonium chloride (3×30 mL) and the saturated brine (3×50 mL), dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 12 g silica gel column, dichloromethane and methanol (3%) to obtain tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (0.4 g, yield: 68.70%), a white solid. ES-API: [M+1]+=590.1.


Step 7: tetratriphenylphosphine palladium (0.051 g, 0.044 mmol) was added to a mixed solution of tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (0.26 g, 0.44 mmol), (5-methyl-1H-indazol-4-yl)boronic acid (0.155 g, 0.88 mmol) and sodium carbonate (0.117 g, 1.1 mmol) in 1,4-dioxane (3 mL) and water (0.75 mL). The resulting mixture was heated to 110° C. and reacted for 1.5 hours under argon atmosphere. After cooling to room temperature, The reaction mixture was extracted with dichloromethane (3×150 mL). The organic phases was combined, washed with water (1×50 mL) and the saturated brine (3×50 mL), dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed. The residue was purified by flash chromatography using silica gel, and eluted with a 12 g silica gel column, dichloromethane and ethyl acetate (65%) to obtain tert-butyl (13aS)-11-chloro-8-(2-isopropylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (200 mg, yield: 70.78%), a yellow oil. ES-API: [M+1]+=608.2.


Step 8: trifluoroacetic acid (1 mL) was added to a solution of tert-butyl (13aS)-11-chloro-8-(2-isopropylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (40 mg, 0.062 mmol) in dichloromethane (2 mL). The resulting mixture was stirred at room temperature for 30 minutes. The pH of the reaction mixture was adjusted to 8 with 1M sodium bicarbonate. The reaction mixture was extracted with dichloromethane (3×20 mL). The organic phases was combined, washed with the saturated brine (3×20 mL), dried with anhydrous sodium sulfate. The solvent was spin-evaporated to be removed to obtain (13aS)-11-chloro-8-(2-isopropylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (30 mg, yield: 88.85%), a yellow oil, directly used in the next step without further purification. ES-API: [M+1]+=508.1.


Step 9: triethylamine (0.056 g, 0.55 mmol) was added to a solution of (13aS)-11-chloro-8-(2-isopropylpyridin-3-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-one (30 mg, 0.055 mmol) in dichloromethane (2 mL). The mixture was cooled to 0° C. Acrylic anhydride (6.98 mg, 0.055 mmol) was added at 0° C. and stirred for 2 hours. Then the solvent was removed under reduced pressure. The residue was purified by a TLC plate to obtain Compound Z39 (2.8 mg, yield: 22.71%), a white solid. ES-API: [M+1]+=561.0. 1H NMR (400 MHz, CDCl3) δ 8.63 (s, 1H), 7.45 (d, J=8.8 Hz, 3H), 6.63 (t, J=13.9 Hz, 1H), 6.42 (d, J=16.9 Hz, 1H), 6.12 (s, 1H), 5.83 (d, J=10.8 Hz, 1H), 5.03 (d, J=13.3 Hz, 1H), 4.69 (t, J=17.4 Hz, 4H), 4.06 (s, 2H), 3.66-3.20 (m, 2H), 2.10 (d, J=6.1 Hz, 3H), 1.09 (d, J=6.6 Hz, 6H).


Example 40: Preparation of Compound Z40



embedded image


Step 1: tert-butyl (R)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.0 g, 1.65 mmol), (1-methyl-2-oxo-1,2-dihydropyridin-3-yl)boronic acid (379 mg, 2.48 mmol), potassium acetate (485 mg, 4.95 mmol) and Pd (dppf)Cl2 (121 mg, 0.165 mmol) dissolved in 1,4-dioxane/water (15 mL, 5:1) was added in sequence to a 100 mL round bottom flask, and reacted at 110° C. for 1.5 hours under nitrogen protection. The product was extracted with ethyl acetate (3*50 mL), washed with the saturated brine (2*20 mL), dried with anhydrous sodium sulfate, and concentrated to obtain the crude product. The crude product was purified by a silica gel column to obtain tert-butyl (R)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(1-methyl-2-oxo-1,2-di hydropyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (a brown oil, 1.0 g, yield: 94.34%). ES-API: [M+H]+=633, 635.


Step 2: tert-butyl (R)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(1-methyl-2-oxo-1,2-di hydropyridin-3-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (1.0 g, 1.58 mmol was dissolved in dichloromethane (12.0 mL), then trifluoroacetic acid (4.0 mL) was added, and reacted at room temperature for 1 hour. The reaction solution was concentrated to obtain (R)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(1-methyl-2-oxo-1,2-di hydropyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′: 3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-1-one (a brown oil, 921 mg, the crude product). ES-API: [M+H]+=533.


Step 3: (R)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(1-methyl-2-oxo-1,2-di hydropyridin-3-yl)-1,2,3,4,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-1-one (842 mg, 1.58 mmol) was dissolved in dichloromethane (10 mL), and triethylamine (479 mg, 4.74 mmol) was added at 0° C. After stirring for 2 min, acrylic anhydride (199 mg, 1.58 mmol, dissolved in 2.0 mL of dichloromethane) was dropped. After the dropping, the reaction proceeded at 0° C. for 1 hour. The saturated sodium bicarbonate (10 mL) was added to the reaction solution. The reaction solution was extracted with dichloromethane (3*20 mL), washed with the saturated sodium bicarbonate (2*20 mL) and the saturated brine (2*20 mL), dried with anhydrous sodium sulfate, and concentrated to obtain the crude product. Product Z40 was purified by a reversed-phase column (a 20 G C18 reversed-phase column, acetonitrile/water=36/100) (a white solid, 122.5 mg, yield: 13.21%). ES-API: [M+H]+=587. 1H NMR (400 MHz, DMSO-d6) δ 0.96-1.08 (m, 6H), 1.96 (d, J=9.0 Hz, 3H), 2.70-2.78 (m, 1H), 3.09 (d, J=13.2 Hz, 1H), 3.23 (d, J=18.6 Hz, 1H), 3.37 (s, 3H), 3.50 (s, 1H), 4.06-4.12 (m, 2H), 4.34 (d, J=12.9 Hz, 1H), 4.45 (d, J=13.3 Hz, 1H), 4.71 (s, 3H), 5.76 (d, J=21.1 Hz, 2H), 6.02-6.13 (m, 2H), 6.19 (d, J=16.6 Hz, 1H), 6.79-6.90 (m, 1H), 7.29 (d, J=4.7 Hz, 1H), 7.68 (d, J=6.9 Hz, 1H), 8.50 (d, J=4.6 Hz, 1H).


Example 41: Preparation of Compound Z41



embedded image


Compound Z41 was prepared with Intermediate a as the raw material according to the method of Example 34. ES-API: [M+H]+=572. 1H-NMR (400 MHz, DMSO-d6) δ 1.02 (dd, J=26.5, 6.3 Hz, 6H), 1.96 (d, J=11.4 Hz, 3H), 2.76 (s, 1H), 3.17 (s, 1H), 3.25 (s, 1H), 3.39-3.49 (m, 2H), 4.03 (s, 1H), 4.14 (s, 1H), 4.32-4.48 (m, 2H), 4.69 (s, 3H), 5.53 (d, J=9.5 Hz, 1H), 5.60 (s, 1H), 5.74 (d, J=10.9 Hz, 2H), 6.19 (d, J=16.7 Hz, 1H), 6.51 (t, J=6.0, 6.0 Hz, 1H), 6.87-7.00 (m, 1H), 7.09 (s, 1H), 7.26 (s, 1H), 7.90 (s, 1H), 8.46 (d, J=4.5 Hz, 1H).


Example 42: Preparation of Compound Z42



embedded image


Compound Z42 was prepared with tetrahydrofuran-3-amine as the raw material according to the method of Example 33. ES-API: [M/2+1]+=527.2. 1H NMR (400 MHz, CDCl3) δ 7.43 (s, 1H), 6.84 (s, 1H), 6.75 (s, 1H), 6.57 (s, 1H), 6.42 (d, J=16.3 Hz, 1H), 5.95 (s, 1H), 5.82 (s, 1H), 4.73 (s, 1H), 4.47 (d, J=21.3 Hz, 4H), 4.29 (s, 1H), 4.08 (s, 1H), 3.93 (d, J=8.5 Hz, 3H), 3.71 (s, 1H), 3.61-3.15 (m, 3H), 2.26 (s, 2H).


Example 43: Preparation of Compound Z43



embedded image


Step 1: tert-butyl (S)-10-bromo-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-3,4,7,8,13,13 a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (800 mg, 1.32 mmol) was dissolved in toluene (10 mL), 1-methyl-6-(tributyltinalkyl)pyridin-2 (1H)-one (631 mg, 1.59 mmol) and tetra(triphenylphosphine)palladium (114 mg, 0.132 mmol) were added, and reacted at 100° C. for 16 hours under nitrogen protection. The reaction solution was extracted with ethyl acetate, concentrated under reduced pressure, and the crude product was purified by column chromatography (dichloromethane/methanol=20/1) to obtain tert-butyl (S)-11-chloro-8-(2-isopropyl-4-methylpyridin-3-yl)-10-(1-methyl-6-oxo-1,6-di hydropyridin-2-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (brown liquid, 630 mg, yield: 75%). ES-API: [M+H]+=633.3.


Steps 2-3: Compound Z43 was prepared according to the corresponding steps of Example 34. ES-API: [M+H]+=587.3. 1H NMR (400 MHz, DMSO-d6) δ 8.48 (d, J=4.7 Hz, 1H), 7.47-7.19 (m, 2H), 6.85 (s, 1H), 6.40 (d, J=9.2 Hz, 1H), 6.19 (d, J=16.7 Hz, 1H), 6.07 (d, J=6.1 Hz, 1H), 5.93 (d, J=52.9 Hz, 1H), 5.75 (d, J=10.5 Hz, 1H), 4.74 (s, 3H), 4.52-3.98 (m, 3H), 3.52 (d, J=15.5 Hz, 2H), 3.25-3.11 (m, 1H), 3.02 (d, J=9.9 Hz, 3H), 2.73 (dd, J=25.7, 17.4 Hz, 1H), 1.96 (dd, J=10.5, 6.8 Hz, 3H), 1.13-0.93 (m, 6H).


Example 44: Preparation of Compound Z44



embedded image


Compound Z44 was prepared with 1-methylpiperidine-4-amine as the raw material according to the method of Example 33. ES-API: [M+1]+=554.2. 1H NMR (400 MHz, DMSO-d6) δ 10.13 (s, 1H), 7.27 (d, J=7.5 Hz, 2H), 6.91-6.68 (m, 3H), 6.16 (d, J=16.7 Hz, 1H), 5.73 (d, J=10.2 Hz, 1H), 4.56 (s, 4H), 4.28-3.93 (m, 3H), 3.44 (d, J=52.7 Hz, 3H), 3.15-2.77 (m, 6H), 2.56 (s, 3H), 1.71 (d, J=12.2 Hz, 2H).


Example 45: Preparation of Compound Z45-1, Z45-2, Z45-3 and Z45-4



embedded image


Step 1: 4-bromo-2,6-difluorobenzonitrile (540 mg, 2.48 mmol) was dissolved in tetrahydrofuran (4 mL), and cooled to −70° C., sodium bis(trimethylsilyl)amide (2.3 mL, 4.5 mmol) was dropped, and after stirring for 30 minutes at −70° C., a solution of 4-isopropyl-6-methylpyrimidin-5-amine in tetrahydrofuran (4 mL) was dropped. The reaction solution gradually rised to room temperature, and stirred overnight. The reaction was quenched with ammonium chloride aqueous solution. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, and passed the column machine (12 g column) to purify (petroleum ether/ethyl acetate=3/1) to obtain the product (380 mg, yield: 44%). ES-API: [M+1]+=349.0.


Step 2: 4-bromo-2-fluoro-6-((4-isopropyl-6-methylpyrimidin-5-yl)amino)benzonitrile (1.1 g, 3.15 mmol) was dissolved in acetonitrile (20 mL), NCS (419 mg, 3.15 mmol) was added, heated to 80° C., and stirred for 3 hours. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, and passed the column machine (12 g column) to purify (petroleum ether/ethyl acetate=4/1) to obtain the product (670 mg, yield: 65%). ES-API: [M+1]+=385.0.


Step 3: 4-bromo-3-chloro-2-fluoro-6-((4-isopropyl-6-methylpyrimidin-5-yl)amino)benzonitrile (620 mg, 1.61 mmol) was dissolved in dimethyl sulfoxide (8 mL), potassium carbonate (1.1 g, 8.07 mmol) was added, cooled to 0° C., hydrogen peroxide was dropped (659 mg, 5.81 mmol). The reaction rised to room temperature, and stirred for 5 hours. The reaction solution was quenched with sodium sulfite aqueous solution. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, and passed the column machine (12 g column) to purify (petroleum ether/ethyl acetate=2/1) to obtain the product (400 mg, yield: 55%). ES-API: [M+1]+=403.0.


Step 4: sodium hydride (497 mg, 12.4 mmol) was suspended in tetrahydrofuran (5 mL), cooled to −10° C., a solution of 4-bromo-3-chloro-2-fluoro-6-((4-isopropyl-6-methylpyrimidin-5-yl)amino)benzamide (500 mg, 1.24 mmol) in tetrahydrofuran (5 mL) was dropped, and after stirring for 20 minutes, a solution of carbonyl diimidazole in tetrahydrofuran (5 mL) was dropped. The reaction solution was stirred at −10° C. for 1 hour. The reaction solution cooled to −30° C., and the pH was adjusted to 2-3 with 0.5M hydrochloric acid solution. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, purified by reversed-phase column chromatography (40 g column) (water/acetonitrile=1/1) to obtain the product (240 mg, yield: 45%). ES-API: [M+1]+=429.0.


Step 5: 7-bromo-6-chloro-5-fluoro-4-hydroxy-1-(4-isopropyl-6-methylpyrimidin-5-yl) quinazolin-2 (1H)-one (240 mg, 0.56 mmol) and tert-butyl (S)-3-(hydroxymethyl)piperazin-1-carboxylate (182 mg, 0.84 mmol) was dissolved in tetrahydrofuran (4 mL), cooled to 0° C., sodium hydride (112 mg, 2.8 mmol) was added, rised to room temperature, and stirred for 1 hour. The reaction solution was quenched with water. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, and passed the column machine (12 g column) to purify (dichloromethane/methanol=10/1) to obtain the product (260 mg, yield: 74%). ES-API: [M+1]+=625.2.


Step 6: tert-butyl (S)-3-(((7-bromo-6-chloro-4-hydroxy-1-(4-isopropyl-6-methylpyrimidin-5-yl)-2-oxo-1,2-dihydroquinazolin-5-yl)oxy)methyl)piperazin-1-carboxylate (180 mg, 0.29 mmol) was dissolved in acetonitrile (9 mL), N,N-diisopropylethylamine (112 mg, 0.86 mmol) and phosphorus oxychloride (66 mg, 0.43 mmol) were added, heated to 80° C., and stirred for 2 hours. The reaction solution was quenched with sodium bicarbonate aqueous solution. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, passed the column machine (12 g column) (ethyl acetate) to obtain the product (80 mg, yield: 45%). ES-API: [M+1]+=607.1.


Step 7: tert-butyl (S)-10-bromo-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate (150 mg, 0.25 mmol) was dissolved in dioxane (2 mL) and water (0.4 mL), (5-methyl-1H-indazol-4-yl)boronic acid (87 mg, 0.49 mmol), 2-dicyclohexylphosphino-2′,6′-dimethoxy-biphenyl (10 mg, 0.025 mmol), tris(dibenzylideneacetone)dipalladium (23 mg, 0.025 mmol) and potassium phosphate (157 mg, 0.74 mmol) were added, and reacted under a 110° C. microwave for 1.33 hours. The reaction solution was extracted with ethyl acetate, the organic phase was dried, spin-dried, column chromatography (ethyl acetate/petroleum ether=0-100%, in the order of polarity from small to large) to obtain the following products, respectively:


tert-butyl (13aS)-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate Z45-a (19 mg), TLC Rf value: 0.30 (ethyl acetate), ES-API: [M+1]+=657.3;


tert-butyl (13aS)-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate Z45-b (30 mg), TLC Rf value: 0.25 (ethyl acetate), ES-API: [M+1]+=657.3;


tert-butyl (13aS)-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-10-(5-methyl-1H-indazol-4-yl)-7-oxo-3,4,7,8,13,13a-hexahydropyrazino[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-2 (1H)-carboxylate Z45-c (20 mg), TLC Rf value: 0.10 (ethyl acetate), ES-API: [M+1]+=657.3.


Step 8: Compound Z45-c (20 mg, 0.03 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (0.5 mL) was added and reacted at room temperature for 1 hour, spin-dried to obtain a yellow oily crude product: (13aS)-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazine[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-on e, a crude product (17 mg, yield: 100%).


Step 9: (13aS)-11-chloro-8-(4-isopropyl-6-methylpyrimidin-5-yl)-10-(5-methyl-1H-indazol-4-yl)-1,2,3,4,13,13a-hexahydropyrazine[2′,1′:3,4][1,4]oxazepine[5,6,7-de]quinazolin-7 (8H)-on e (17 mg, 0.03 mmol), and triethylamine (0.5 mL) was dissolved in dichloromethane (2 mL), cooled to 0° C., acrylic anhydride (3 mg, 0.027 mmol) was added, and reacted at 0° C. for 0.5 hour. The reaction solution was extracted with 10 mL of water and dichloromethane (10 mL*3), the extracted organic phase was spin-dried to obtain the crude product, the crude product was prepared and purified (chromatographic column: Ultimate XB-C18,50*250 mm, 10 um; elution system: acetonitrile/water, 40 minutes from 10% to 90%; monitoring wavelength 210 nm; flow rate 80 mL/min), freeze-dried to obtain Compound Z45-1 (4 mg, yield: 20%). ES-API: [M+1]+=611.2. 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.45 (d, J=9.5 Hz, 1H), 6.70-6.56 (m, 1H), 6.42 (d, J=16.6 Hz, 1H), 6.04 (s, 1H), 5.84 (d, J=10.4 Hz, 1H), 5.08 (d, J=13.2 Hz, 1H), 4.69 (s, 2H), 4.12 (d, J=7.2 Hz, 1H), 3.69 (d, J=43.3 Hz, 1H), 3.51-3.28 (m, 2H), 2.84-2.77 (m, 1H), 2.31 (s, 3H), 2.11 (s, 3H), 2.04 (s, 1H), 1.41 (s, 1H), 1.33 (s, 2H), 1.25 (s, 6H), 1.06 (d, J=6.7 Hz, 2H).


Steps 10-11: With Compound Z45-b as raw material, the method of steps 8-9 in Example 45 was referred to finally purify by column chromatography (methanol/dichloromethane=0-10%)(polarity from small to large) to obtain:


Compound Z45-2 (a white solid, 3.8 mg); LC-MS: retention time: 1.597 min; ES-API: [M+1]+=611.2. 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.45 (d, J=9.5 Hz, 1H), 6.70-6.56 (m, 1H), 6.42 (d, J=16.6 Hz, 1H), 6.04 (s, 1H), 5.84 (d, J=10.4 Hz, 1H), 5.08 (d, J=13.2 Hz, 1H), 4.69 (s, 2H), 4.12 (d, J=7.2 Hz, 1H), 3.69 (d, J=43.3 Hz, 1H), 3.51-3.28 (m, 2H), 2.84-2.77 (m, 1H), 2.31 (s, 3H), 2.11 (s, 3H), 2.04 (s, 1H), 1.41 (s, 1H), 1.33 (s, 2H), 1.25 (s, 6H), 1.06 (d, J=6.7 Hz, 2H).


and Compound Z45-3 (a white solid, 4.0 mg); LC-MS: retention time: 1.648 min; ES-API: [M+1]+=611.2. 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.32 (s, 1H), 6.64 (dd, J=16.7, 10.5 Hz, 1H), 6.43 (d, J=16.7 Hz, 1H), 6.03 (s, 1H), 5.84 (d, J=10.4 Hz, 1H), 5.08 (d, J=13.1 Hz, 1H), 4.69 (d, J=14.7 Hz, 3H), 4.08 (s, 2H), 3.72-3.30 (m, 3H), 2.87-2.76 (m, 1H), 2.34 (s, 3H), 2.09 (s, 3H), 1.43-1.37 (m, 1H), 1.26 (d, J=8.1 Hz, 6H), 1.05 (d, J=6.6 Hz, 2H).


Steps 12-13: With Compound Z45-a as raw material, the method of steps 8-9 in Example 45 was referred to obtain Compound Z45-4 (a white solid, 4.2 mg). ES-API: [M+1]+=611.2. 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.45 (d, J=9.5 Hz, 1H), 6.70-6.56 (m, 1H), 6.42 (d, J=16.6 Hz, 1H), 6.04 (s, 1H), 5.84 (d, J=10.4 Hz, 1H), 5.08 (d, J=13.2 Hz, 1H), 4.69 (s, 2H), 4.12 (d, J=7.2 Hz, 1H), 3.69 (d, J=43.3 Hz, 1H), 3.51-3.28 (m, 2H), 2.84-2.77 (m, 1H), 2.31 (s, 3H), 2.11 (s, 3H), 2.04 (s, 1H), 1.41 (s, 1H), 1.33 (s, 2H), 1.25 (s, 6H), 1.06 (d, J=6.7 Hz, 2H).


Example 46: Preparation of Compound Z46, Z46-1 and Z46-2



embedded image


embedded image


Step 1: tert-butyl (S)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5α,6,8,9,11,12hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthale n-7 (5H)-carboxylate (300 mg, 0.54 mmol), (2-fluorophenyl)boronic acid (227 mg, 1.62 mmol), tetratriphenylphosphine palladium (62 mg, 0.054 mmol), sodium carbonate (172 mg, 1.62 mmol), 50 mL of dioxane and 10 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 65° C. for 3 hours under nitrogen protection, and the reaction stopped. 30 mL of water was added to the reaction solution. The reaction solution was extracted with 30 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (S)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (211 mg, 63%), a yellow solid. ES-API: [M+H]+=622.3.


Steps 2-3: Compound Z46 was prepared according to the corresponding steps of Example 34. ES-API: [M+H]+=576.2. 1H NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=4.8 Hz, 1H), 7.47 (dd, J=13.4, 6.2 Hz, 1H), 7.31-7.21 (m, 2H), 7.15 (d, J=4.8 Hz, 2H), 6.95-6.75 (m, 1H), 6.21 (dd, J=16.6, 2.2 Hz, 1H), 5.77 (dd, J=10.4, 2.2 Hz, 1H), 4.98-4.78 (m, 2H), 4.65-4.53 (m, 1H), 4.51-4.32 (m, 1H), 4.26 (d, J=20.2 Hz, 1H), 4.18-3.94 (m, 1H), 3.82-3.56 (m, 2H), 2.83-2.66 (m, 1H), 1.94 (d, J=11.5 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 0.93 (d, J=6.6 Hz, 3H).


Step 4: Compound Z46 was prepared and resolved chirally (column type: IG, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=40:60, flow rate: 1 ml/min, column temperature=30° C.) to obtain:


Compound Z46-1 (79.8 mg, peak 1, retention time 8.093 min, yield: 30.2%), a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.48 (ddd, J=15.6, 5.4, 1.8 Hz, 1H), 7.35-7.07 (m, 4H), 6.96-6.77 (m, 1H), 6.00 (ddd, J=13.5, 12.7, 2.3 Hz, 2H), 5.01-4.79 (m, 2H), 4.69-4.25 (m, 3H), 4.07 (d, J=31.0 Hz, 1H), 3.82-3.57 (m, 2H), 3.38 (s, 1H), 2.78 (dt, J=13.4, 6.7 Hz, 1H), 1.94 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


and Compound Z46-2 (84.48 mg, peak 2, retention time 11.618 min, yield: 31.9%), a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.48 (dd, J=13.3, 6.1 Hz, 1H), 7.34-7.10 (m, 4H), 6.95-6.77 (m, 1H), 6.22 (d, J=16.7 Hz, 1H), 5.78 (d, J=12.2 Hz, 1H), 4.99-4.81 (m, 2H), 4.68-4.22 (m, 3H), 4.07 (d, J=14.4 Hz, 1H), 3.83-3.58 (m, 2H), 3.37 (s, 1H), 2.78-2.62 (m, 1H), 1.96 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.7 Hz, 3H).


Example 47: Preparation of Compound Z47, Z47-1 and Z47-2



embedded image


Compound Z47 was prepared with (2,3-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=594.2. Compound Z47 was resolved chirally (mobile phase: acetonitrile:isopropanol=60:40); column type: IC (250 mm*4.6 mm*5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z47-1 (23 mg, retention time: 5.176 min, purity: 100%, de value: 100%). ES-API: [M+H]+=594.2;


and Compound Z47-2 (29 mg, retention time: 9.285 min, purity: 100%, de value: 99%). ES-API: [M+H]+=594.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.52 (dd, J=18.0, 8.6 Hz, 1H), 7.26 (dd, J=12.8, 8.0 Hz, 1H), 7.20-7.13 (m, 1H), 6.99 (d, J=6.4 Hz, 1H), 6.94-6.75 (m, 1H), 6.21 (dd, J=16.8, 2.0 Hz, 1H), 5.78 (dd, J=10.4, 2.0 Hz, 1H), 5.04-4.81 (m, 2H), 4.56 (s, 1H), 4.42 (dd, J=53.8, 12.1 Hz, 1H), 4.30 (s, 1H), 4.06 (d, J=34.1 Hz, 1H), 3.85-3.61 (m, 2H), 3.41-3.30 (m, 1H), 2.78-2.63 (m, 1H), 1.94 (d, J=12.3 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H).


Example 48: Preparation of Compound Z48



embedded image


Compound Z48 was prepared with (3-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=576.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.42 (d, J=4.8 Hz, 1H), 7.45 (dd, J=14.0, 8.0 Hz, 1H), 7.40-7.27 (m, 2H), 7.25-7.08 (m, 2H), 7.00-6.72 (m, 1H), 6.22 (d, J=16.6 Hz, 1H), 5.78 (d, J=11.6 Hz, 1H), 4.87 (ddd, J=19.0, 17.8, 8.2 Hz, 2H), 4.59 (s, 1H), 4.52-4.34 (m, 1H), 4.27 (s, 1H), 4.08 (d, J=40.8 Hz, 1H), 3.71 (d, J=44.8 Hz, 2H), 3.40-3.25 (m, 1H), 2.87-2.67 (m, 1H), 1.95 (d, J=10.1 Hz, 3H), 1.08 (d, J=6.8 Hz, 3H), 0.95 (d, J=6.4 Hz, 3H).


Example 49: Preparation of Compound Z49, Z49-1 and Z49-2



embedded image


Compound Z49 was prepared with (2,4-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=594.2. Compound Z49 (75 mg, 0.12 mmol) was resolved chirally (mobile phase: acetonitrile:isopropanol=60:40; column type: IC (250 mm*4.6 mm 5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z49-1 (27 mg, retention time: 5.318 min, purity: 100%, de value: 100%). ES-API: [M+H]+=594.1.


and Compound Z49-2 (28 mg, retention time: 8.840 min, purity: 99%, de value: 99%). ES-API: [M+H]+=594.1. 1H-NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.35 (t, J=9.8 Hz, 1H), 7.29-7.10 (m, 3H), 6.98-6.73 (m, 1H), 6.22 (dd, J=16.8, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 5.05-4.80 (m, 2H), 4.57 (s, 1H), 4.42 (dd, J=54.5, 12.0 Hz, 1H), 4.29 (s, 1H), 4.06 (d, J=33.4 Hz, 1H), 3.73 (d, J=54.0 Hz, 2H), 2.87-2.65 (m, 1H), 1.94 (d, J=12.0 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.93 (d, J=6.8 Hz, 3H).


Example 50: Preparation of Compound Z50



embedded image


Compound Z50 was prepared with (4-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=576.2. 1H NMR (500 MHz, DMSO-d6) δ 8.42 (d, J=5.0 Hz, 1H), 7.48 (dd, J=8.8, 5.6 Hz, 2H), 7.24 (t, J=8.8 Hz, 2H), 7.20 (t, J=4.2 Hz, 1H), 6.86 (m, 1H), 6.21 (m, 1H), 5.78 (d, J=12.0 Hz, 1H), 4.87 (m, 2H), 4.60 (s, 1H), 4.43 (dd, J=54.2, 12.8 Hz, 1H), 4.26 (s, 1H), 4.09 (d, J=43.2 Hz, 1H), 3.70 (d, J=52.2 Hz, 2H), 2.74 (m, 1H), 1.95 (d, J=9.8 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H).


Example 51: Preparation of Compound Z51



embedded image


(5 aS)-7-acryloyl-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalene11 (12H)-one (200 mg, 0.34 mmol), N-chlorosuccinimide (137 mg, 1.02 mmol) and acetic acid (30 mL) were added to a round bottom flask. The reaction was stirred at 75° C. for 2 hours. concentrated, and the crude product was separated by Preparative HPLC to obtain the product: (5 aS)-7-acryloyl-3-chloro-2-(3,5-dichloro-2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (176 mg, 56%), a yellow solid. ES-API: [M+H]+=659.6. 1H-NMR (500 MHz, DMSO-d6) δ 10.25 (dd, J=23.0, 10.0 Hz, 1H), 8.29 (d, J=4.8 Hz, 1H), 7.70 (d, J=7.8 Hz, 1H), 7.10 (s, 1H), 6.78 (s, 1H), 6.14 (d, J=16.5 Hz, 1H), 5.71 (d, J=10.2 Hz, 1H), 4.93-4.72 (m, 2H), 4.59-4.35 (m, 2H), 4.28-4.11 (m, 1H), 4.10-3.89 (m, 1H), 3.77-3.48 (m, 3H), 2.81-2.56 (m, 1H), 1.85 (dd, J=19.6, 12.4 Hz, 3H), 0.99 (d, J=6.2 Hz, 3H), 0.89-0.81 (m, 3H).


Example 52: Preparation of Compound Z52



embedded image


(5aS)-7-acryloyl-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (300 mg, 0.5085 mmol) was added to 30 mL of acetonitrile, finally N-chlorosuccinimide (135 mg, 1.0169 mmol) was added, and reacted at 75° C. for 1 hour. 80 mL of ethyl acetate was added, the reaction solution was washed with the saturated brine for 3 times (3*60 mL). The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was spin-dried and prepared to obtain (5 aS)-7-acryloyl-3-chloro-2-(3-chloro-6-fluoro-2-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (9.2 mg, yield: 3%). ES-API: [M+H]+=625.2.


Example 53: Preparation of Compound Z53



embedded image


Step 1: tert-butyl (S)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (200 mg, 0.36 mmol), (2,3-difluoro-6-methoxyphenyl)boronic acid (200 mg, 1.07 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (28 mg, 0.04 mmol), 2-bicyclohexylphosphin-2′,6′-dimethoxybiphenyl (16 mg, 0.04 mmol), a mixed solution of potassium phosphate (227 mg, 1.07 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) reacted under a 100° C. microwave for 1 hour under nitrogen protection. The reaction solution was filtered, washed with ethyl acetate (30 mL), the filtrate was washed with the saturated brine (10 mL*3), the obtained organic phase was dried and concentrated, purified with a fast silica column (0-10% methanol/dichloromethane) to obtain a yellow solid: tert-butyl (S)-3-chloro-2-(2,3-difluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (100 mg, yield: 23%). ES-API: [M+H]+=669.1.


Step 2: 17% boron tribromide in dichloromethane solution (2 mL) was added to a solution of tert-butyl (S)-3-chloro-2-(2,3-difluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (100 mg, 0.15 mmol) in dichloromethane (2 mL) solution under the ice bath, and stirred at room temperature for 3 hours. The reaction solution was quenched with the saturated sodium bicarbonate solution (20 mL), extracted with dichloromethane, and concentrated to obtain a yellow solid: (S)-3-chloro-2-(2,3-difluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (83 mg). ES-API: [M+H]+=555.1.


Step 3: N,N-diisopropylethylamine (58 mg, 0.45 mmol) was added to a solution of (S)-3-chloro-2-(2,3-difluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (83 mg, 0.15 mmol) in dichloromethane (2 mL) under the ice bath. After the reaction solution was clarified, acrylic anhydride (11 mg, 0.09 mmol) was added, and stirred for 5 minutes. The reaction solution was concentrated, and then purified by Preparative HPLC (ammonium bicarbonate system) to obtain a white solid: Z53 (7.76 mg, purity: 90%, yield: 9%). 1H NMR (500 MHz, DMSO-d6) δ 10.04 (t, J=15.7 Hz, 1H), 8.37 (d, J=4.7 Hz, 1H), 7.32-7.23 (m, 1H), 7.21-7.14 (m 1H), 6.92-6.78 (m, 1H), 6.67-6.61 (m, 1H), 6.21 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.5, 2.1 Hz, 1H), 4.96-4.82 (m, 2H), 4.63-4.52 (m, 1H), 4.49-4.34 (m, 1H), 4.29 (s, 1H), 4.11-3.99 (m, 1H), 3.73-3.59 (m, 1H), 3.56-3.47 (m, 1H), 2.80-2.62 (m, 1H), 1.93 (d, J=12.2 Hz, 3H), 1.08 (d, J=6.7 Hz, 3H), 0.94 (d, J=4.7 Hz, 3H). ES-API: [M+H]+=609.1.


Example 54: Preparation of Compound Z54



embedded image


Compound Z54 was prepared with (3,5-difluoro-2-methoxyphenyl)boronic acid as the raw material according to the method of Example 53. 1H NMR (500 MHz, DMSO-d6) δ 9.83 (d, J=5.1 Hz, 1H), 8.37 (d, J=4.8 Hz, 1H), 7.35-7.24 (m, 1H), 7.17 (d, J=4.6 Hz, 1H), 6.95-6.78 (m, 1H), 6.61 (d, J=8.5 Hz, 1H), 6.22 (d, J=16.6 Hz, 1H), 5.78 (d, J=12.2 Hz, 1H), 4.99-4.81 (m, 2H), 4.60 (d, J=16.8 Hz, 1H), 4.43 (dd, J=55.1, 12.0 Hz, 1H), 4.27 (s, 1H), 4.08 (d, J=42.2 Hz, 1H), 3.69 (d, J=41.3 Hz, 2H), 3.37 (d, J=11.3 Hz, 1H), 2.74 (ddd, J=26.9, 13.3, 6.7 Hz, 1H), 1.94 (d, J=10.1 Hz, 3H), 1.07 (d, J=6.7 Hz, 3H), 0.96 (dd, J=6.6, 1.7 Hz, 3H). ES-API: [M+H]+=609.1.


Example 55: Preparation of Compound Z55



embedded image


Compound Z55 was prepared with (5-fluoro-2-hydroxyphenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=591.1. 1H-NMR (500 MHz, DMSO-d6) δ 9.72 (s, 1H), 8.37 (d, J=4.8 Hz, 1H), 7.17 (d, J=3.8 Hz, 1H), 7.05 (td, J=8.7, 3.2 Hz, 1H), 6.86 (ddd, J=13.7, 12.7, 7.5 Hz, 2H), 6.75-6.66 (m, 1H), 6.22 (dd, J=16.7, 2.1 Hz, 1H), 5.78 (dd, J=10.5, 2.0 Hz, 1H), 5.04-4.81 (m, 2H), 4.58 (s, 1H), 4.43 (dd, J=56.0, 12.8 Hz, 1H), 4.25 (s, 1H), 4.10 (d, J=52.6 Hz, 1H), 3.77-3.57 (m, 2H), 3.38 (s, 1H), 2.74 (ddt, J=33.6, 13.1, 6.6 Hz, 1H), 1.94 (d, J=9.7 Hz, 3H), 1.07 (d, J=6.7 Hz, 3H), 0.97 (dd, J=6.7, 3.5 Hz, 3H).


Example 56: Preparation of Compound Z56



embedded image


Compound Z56 was prepared with (2,4,6-trifluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=612.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.32 (s, 2H), 7.16 (d, J=4.8 Hz, 1H), 6.96-6.69 (m, 1H), 6.21 (dd, J=16.8, 2.2 Hz, 1H), 5.77 (dd, J=10.4, 2.2 Hz, 1H), 5.08-4.80 (m, 2H), 4.63-4.27 (m, 3H), 4.03 (s, 1H), 3.91-3.58 (m, 2H), 3.54-3.41 (m, 1H), 2.83-2.67 (m, 1H), 1.91 (d, J=17.8 Hz, 3H), 1.05 (d, J=6.8 Hz, 3H), 0.91 (t, J=6.3 Hz, 3H).


Example 57: Preparation of Compound Z57, Z57-1 and Z57-2



embedded image


Compound Z57 was prepared with (2-fluoro-3-(trifluoromethyl)phenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=644.2. Compound Z57 (40 mg) was resolved chirally (mobile phase: n-hexane:ethanol=50:50; column type: IG (250 mm*4.6 mm*5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z57-1 (16 mg, retention time: 7.222 min, purity: 100%, de value: 99.5%). ES-API: [M+H]+=644.2.


and Compound Z57-2 (12 mg, retention time: 10.152 min, purity: 100%, de value: 99.3%). ES-API: [M+H]+=644.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.88 (t, J=7.1 Hz, 1H), 7.56-7.43 (m, 2H), 7.16 (d, J=2.9 Hz, 1H), 6.95-6.74 (m, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 5.02-4.82 (m, 2H), 4.64-4.54 (m, 1H), 4.43 (dd, J=52.2, 12.1 Hz, 1H), 4.31 (s, 1H), 4.07 (d, J=27.8 Hz, 1H), 3.87-3.57 (m, 2H), 2.83-2.66 (m, 1H), 2.53-2.46 (m, 1H), 1.95 (d, J=11.2 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H).


Example 58: Preparation of Compound Z58



embedded image


Compound Z58 was prepared with 2-chlorophenylboronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=591.1. 1H NMR (500 MHz, DMSO-d6) δ 8.34 (d, J=4.8 Hz, 1H), 7.50 (d, J=7.9 Hz, 1H), 7.43 (td, J=7.7, 1.8 Hz, 1H), 7.41-7.36 (m, 1H), 7.16 (dd, J=13.4, 3.5 Hz, 2H), 6.86 (m, 1H), 6.22 (dd, J=16.7, 2.4 Hz, 1H), 5.78 (dd, J=10.4, 2.4 Hz, 1H), 4.90 (dd, J=26.6, 14.0 Hz, 2H), 4.57 (d, J=28.2 Hz, 1H), 4.42 (dd, J=52.6, 13.4 Hz, 1H), 4.30 (s, 1H), 4.07 (d, J=30.3 Hz, 1H), 3.67 (s, 2H), 3.38 (dd, J=10.2, 5.2 Hz, 1H), 2.84-2.63 (m, 1H), 1.94 (d, J=10.6 Hz, 3H), 1.00 (m, 6.6 Hz, 6H).


Example 59: Preparation of Compound Z59



embedded image


Compound Z59 was prepared with (5-chloro-2-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=609.0. 1H NMR (500 MHz, DMSO-d6) δ 8.38 (d, J=4.8 Hz, 1H), 7.60-7.53 (m, 1H), 7.41-7.33 (m, 1H), 7.23 (td, J=5.9, 2.7 Hz, 1H), 7.18 (dd, J=5.0, 2.1 Hz, 1H), 6.94-6.78 (m, 1H), 6.22 (dd, J=16.6, 2.4 Hz, 1H), 5.78 (dd, J=10.4, 2.3 Hz, 1H), 4.99-4.82 (m, 2H), 4.59 (s, 1H), 4.42 (dd, J=52.5, 13.2 Hz, 1H), 4.29 (s, 1H), 4.07 (d, J=32.5 Hz, 1H), 3.73 (d, J=55.8 Hz, 2H), 3.38 (s, 1H), 2.82-2.68 (m, 1H), 1.95 (d, J=11.7 Hz, 3H), 1.13-0.90 (m, 6H).


Example 60: Preparation of Compound Z60



embedded image


Compound Z60 was prepared with (2,5-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=593.2. 1H NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.9 Hz, 1H), 7.42-7.31 (m, 2H), 7.18 (dd, J=5.0, 2.6 Hz, 1H), 7.03 (q, J=7.1 Hz, 1H), 6.86 (m, 1H), 6.22 (m, 1H), 5.78 (dd, J=10.4, 2.4 Hz, 1H), 5.02-4.80 (m, 2H), 4.66-4.52 (m, 1H), 4.43 (m, H), 4.29 (s, 1H), 4.19-4.00 (m, 1H), 3.74 (d, J=60.6 Hz, 2H), 3.37 (s, 1H), 2.74 (m, 1H), 1.95 (d, J=12.1 Hz, 3H), 1.00 (dd, J=64.3, 6.7 Hz, 6H).


Example 61: Preparation of Compound Z61



embedded image


Compound Z61 was prepared with (3-chloro-2-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=609.1. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.9 Hz, 1H), 7.74-7.63 (m, 1H), 7.29 (t, J=7.9 Hz, 1H), 7.20-7.11 (m, 2H), 6.86 (m, 1H), 6.22 (dd, J=16.7, 2.4 Hz, 1H), 5.78 (dd, J=10.3, 2.4 Hz, 1H), 5.02-4.79 (m, 2H), 4.66-4.52 (m, 1H), 4.50-4.34 (m, 1H), 4.30 (s, 1H), 4.07 (d, J=32.6 Hz, 1H), 3.74 (d, J=56.3 Hz, 2H), 3.39 (s, 1H), 2.74 (m, 1H), 1.95 (d, J=12.1 Hz, 3H), 1.01 (dd, J=62.4, 6.7 Hz, 6H).


Example 62: Preparation of Compound Z62



embedded image


Compound Z62 was prepared with 2-isopropylbenzeneboronic acid as the raw material according to the method of Example 46. 1H-NMR (500 MHz, DMSO-d6) δ 8.32 (d, J=4.2, 1H), 7.34 (d, J=3.1 Hz, 2H), 7.24-7.15 (m, 1H), 7.15-7.08 (m, 1H), 6.96-6.69 (m, 2H), 6.22 (dd, J=16.7, 2.1 Hz, 1H), 5.78 (d, J=12.5 Hz, 1H), 4.98-4.80 (m, 2H), 4.67-4.52 (m, 1H), 4.51-4.34 (m, 1H), 4.29 (s, 1H), 4.19-3.99 (m, 1H), 3.88-3.62 (m, 2H), 3.42-3.25 (m, 1H), 2.79-2.64 (m, 1H), 1.92 (d, J=11.6 Hz, 3H), 1.06 (s, 6H), 0.96-0.86 (m, 3H), 0.83-0.70 (s, 3H). ES-API: [M+H]+=599.2.


Example 63: Preparation of Compound Z63



embedded image


Compound Z63 was prepared with 3-isopropylbenzeneboronic acid as the raw material according to the method of Example 46. 1H NMR (500 MHz, DMSO-d6) δ 8.41 (dd, J=4.7, 2.5 Hz, 1H), 7.44 (d, J=7.1 Hz, 1H), 7.32-7.26 (m, 2H), 7.24 (s, 1H), 7.18 (t, J=4.7 Hz, 1H), 6.95-6.77 (m, 1H), 6.22 (d, J=17.0 Hz, 1H), 5.78 (d, J=11.7 Hz, 1H), 4.95-4.81 (m, 2H), 4.70-4.55 (m, 1H), 4.50-4.35 (m, 1H), 4.31-4.22 (m, 1H), 4.17-4.00 (m, 1H), 3.79-3.57 (m, 2H), 3.37-3.28 (m, 1H), 2.85-2.71 (m, 2H), 1.94 (d, J=7.5 Hz, 3H), 1.12-1.05 (m, 9H), 1.00 (t, J=7.1 Hz, 3H). ES-API: [M+H]+=599.2.


Example 64: Preparation of Compound Z64



embedded image


Compound Z64 was prepared with (2-(trifluoromethoxy)phenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=641.1. 1H NMR (500 MHz, DMSO-d6) δ 8.34 (d, J=4.8 Hz, 1H), 7.59-7.54 (m, 1H), 7.44 (t, J=7.4 Hz, 2H), 7.27-7.12 (m, 2H), 6.87 (d, J=10.2 Hz, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.88 (d, J=21.2 Hz, 2H), 4.37 (dd, J=53.4, 41.9 Hz, 3H), 4.05 (s, 1H), 3.69 (s, 2H), 3.37 (d, J=5.7 Hz, 1H), 2.72 (s, 1H), 2.05-1.83 (m, 3H), 1.07 (d, J=6.0 Hz, 3H), 0.99-0.78 (m, 3H).


Example 65: Preparation of Compound Z65



embedded image


Compound Z65 was prepared with (4-chloro-2-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=609.1. 1H-NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.55 (d, J=9.8 Hz, 1H), 7.36 (d, J=8.2 Hz, 1H), 7.25-7.12 (m, 2H), 6.94-6.79 (m, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.1 Hz, 1H), 4.99-4.82 (m, 2H), 4.58 (d, J=15.7 Hz, 1H), 4.42 (dd, J=54.0, 13.6 Hz, 1H), 4.29 (s, 1H), 4.06 (d, J=29.2 Hz, 1H), 3.72 (d, J=48.9 Hz, 2H), 3.41-3.34 (m, 1H), 2.74 (ddd, J=30.8, 13.4, 6.7 Hz, 1H), 1.94 (d, J=12.5 Hz, 3H), 1.07 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.6 Hz, 3H).


Example 66: Preparation of Compound Z66



embedded image


Compound Z66 was prepared with (2-(trifluoromethyl)phenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=625.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.31 (d, J=4.6 Hz, 1H), 7.75 (dd, J=27.0, 7.7 Hz, 2H), 7.64 (t, J=7.2 Hz, 1H), 7.29 (dd, J=24.8, 7.5 Hz, 1H), 7.16-7.11 (m, 1H), 6.92-6.79 (m, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.97-4.78 (m, 2H), 4.63-4.28 (m, 3H), 4.05 (s, 1H), 3.75 (d, J=63.0 Hz, 2H), 3.37 (s, 1H), 2.78-2.66 (m, 1H), 1.91 (t, J=10.0 Hz, 3H), 1.06 (dd, J=11.8, 5.4 Hz, 3H), 0.96-0.84 (m, 3H).


Example 67: Preparation of Compound Z67



embedded image


Compound Z67 was prepared with 2-chloro-6-fluorophenylboronic acid as the raw material according to the method of Example 46. 1H-NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=5.0 Hz, 1H), 7.55-7.47 (m, 1H), 7.44-7.39 (m, 1H), 7.37-7.29 (m, 1H), 7.19-7.14 (m, 1H), 6.93-6.77 (m, 1H), 6.22 (d, J=16.6 Hz, 1H), 5.78 (d, J=10.9 Hz, 1H), 5.02-4.83 (m, 2H), 4.65-4.38 (m, 2H), 4.37-4.28 (m, 1H), 4.14-3.99 (m, 1H), 3.90-3.65 (m, 2H), 2.81-2.62 (m, 2H), 1.92 (t, J=14.8 Hz, 3H), 1.08 (d, J=6.6 Hz, 3H), 0.98-0.87 (m, 3H). ES-API: [M+H]+=609.1.


Example 68: Preparation of Compound Z68



embedded image


Compound Z68 was prepared with (2,6-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=594.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=4.8 Hz, 1H), 7.60-7.49 (m, 1H), 7.30-7.11 (m, 3H), 6.96-6.73 (m, 1H), 6.21 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 5.01-4.80 (m, 2H), 4.66-4.24 (m, 3H), 4.33-4.25 (m, 1H), 4.15-3.95 (m, 1H), 3.81-3.62 (m, 2H), 3.42-3.16 (m, 1H), 2.85-2.62 (m, 1H), 1.92 (d, J=16.8 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.92 (t, J=6.0 Hz, 3H).


Example 69: Preparation of Compound Z69



embedded image


Compound Z69 was prepared with (3-fluoropyrid-4-yl)boronic acid as the raw material according to the method of Example 46. 1H NMR (500 MHz, DMSO-d6) δ 8.62 (s, 1H), 8.44 (d, J=4.8 Hz, 1H), 8.29 (d, J=4.9 Hz, 1H), 7.24-7.05 (m, 2H), 6.88-6.69 (m, 1H), 6.14 (dd, J=16.7, 2.3 Hz, 1H), 5.71 (dd, J=10.4, 2.3 Hz, 1H), 4.95-4.83 (m, 2H), 4.59-4.18 (m, 3H), 3.98 (d, J=28.4 Hz, 1H), 3.67 (d, J=56.4 Hz, 2H), 3.30 (s, 1H), 2.75-2.59 (m, 1H), 1.88 (d, J=13.2 Hz, 3H), 0.99 (d, J=6.7 Hz, 3H), 0.86 (d, J=6.7 Hz, 3H). ES-API: [M+H]+=643.1.


Example 70: Preparation of Compound Z70



embedded image


Compound Z70 was prepared with (2-fluoro-6-(trifluoromethyl)phenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=643.1. 1H NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.82 (d, J=9.7 Hz, 1H), 7.67 (d, J=8.0 Hz, 1H), 7.43 (dd, J=14.0, 7.0 Hz, 1H), 7.17 (s, 1H), 6.84 (dd, J=25.9, 14.1 Hz, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.5, 2.2 Hz, 1H), 5.01-4.82 (m, 2H), 4.64-4.25 (m, 3H), 4.07 (d, J=29.3 Hz, 1H), 3.74 (d, J=56.6 Hz, 2H), 3.37 (s, 1H), 2.83-2.66 (m, 1H), 1.95 (d, J=12.1 Hz, 3H), 1.07 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H).


Example 71: Preparation of Compound Z71



embedded image


Compound Z71 was prepared with (2,6-dimethylpyridin-4-yl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=586.2. 1H NMR (500 MHz, DMSO-d6) δ 8.41 (d, J=4.5 Hz, 1H), 7.20 (s, 1H), 7.00 (s, 2H), 6.84 (dd, J=27.0, 14.4 Hz, 1H), 6.22 (d, J=16.8 Hz, 1H), 5.78 (d, J=10.3 Hz, 1H), 5.01-4.77 (m, 2H), 4.66-4.24 (m, 3H), 4.07 (d, J=33.5 Hz, 1H), 3.72 (d, J=53.7 Hz, 2H), 3.37 (s, 1H), 2.87-2.67 (m, 1H), 2.38 (s, 6H), 1.93 (d, J=9.7 Hz, 3H), 1.12-0.91 (m, 6H).


Example 72: Preparation of Compound Z72



embedded image


embedded image


Step 1: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo5α,6,8,9,11,12hexa hydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.43 g, 0.82 mmol), N-bromosuccinimide (292 mg, 1.64 mmol) and acetonitrile (5 mL) were added to a round bottom flask. The reaction was stirred at 70° C. for 2 hours. Sodium thiosulfate aqueous solution was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-50%) to obtain tert-butyl (R)-3-bromo-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (440 mg), yield of 88%, a yellow solid. ES-API: [M+H]+=605 0.2.


Step 2: tert-butyl (R)-3-bromo-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (180 mg, 0.30 mmol), 2,4,6-trimethyl-1,3,5,2,4,6-trioxytriborane (113 mg, 0.90 mmol), Pd(dppf)Cl2 (22 mg, 0.03 mmol), potassium phosphate (191 mg, 0.9 mmol), 5 mL of dioxane and 1 mL of water were added to a reaction flask. The reaction was stirred under a 70° C. oil bath for 12 hours, and the reaction stopped. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-60%) to obtain the target product: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (110 mg, 50% purity), a yellow solid. ES-API: [M+H]+=541.2.


Step 3: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (110 mg, 0.20 mmol), 2-fluoro-6-hydroxyphenylboronic acid (156 mg, 1.0 mmol), SPhos (8 mg, 0.02 mmol), SPhos-Pd-G2 (14 mg, 0.02 mmol), potassium phosphate (127 mg, 0.6 mmol), 5 mL of dioxane and 1 mL of water were added to a reaction flask. The reaction was stirred under a 80° C. oil bath for 6 hours, and the reaction stopped. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-60%) to obtain the target product: tert-butyl (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-11-oxo5α,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (55 mg, yield of two steps: 30%), a yellow solid. ES-API: [M+H]+=617.2.


Step 4: tert-butyl (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-11-oxo5α,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (55 mg, 0.089 mmol), 1 mL of methanol and 4 M hydrogen chloride/dioxane solution (3 mL) were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohept an [1,2,3-de]naphthalen-11 (12H)-one (46 mg, the crude product), a yellow solid. ES-API: [M+H]+=517.2.


Step 5: (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-methyl-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]naphthalen-11 (12H)-one (46 mg, 0.089 mmol), 3 mL of dichloromethane and triethylamine (27 mg, 0.27 mmol) were added to a 50 mL round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (31 mg, 0.25 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 20 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 10 mL of dichloromethane for 3 times. The organic phase was dried and concentrated. The crude product was purified by Preparative HPLC to obtain Z72 (17.88 mg, yield of two steps: 35%), a white solid. 1H-NMR (500 MHz, CDCl3) 8.69 (brs, 1H), 8.50-8.49 (m, 1H), 7.19-7.13 (m, 2H), 6.61-6.53 (m, 3H), 6.35-6.31 (m, 1H), 5.78 (d, J=8 Hz, 1H), 5.01-4.51 (m, 4H), 4.02-3.31 (m, 5H), 2.78-2.76 (m, 1H), 2.05-2.04 (m, 6H), 1.22-1.20 (m, 3H), 1.03-1.00 (m, 3H). ES-API: [M+H]+=571.2.


Example 73: Preparation of Compound Z73, Z73-1 and Z73-2



embedded image


embedded image


Step 1: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo5α,6,8,9,11,12hexa hydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1.1 g, 2.09 mmol), Selectfluor (3.69 g, 10.4 mmol) and 50 mL of acetonitrile were added to a round bottom flask. The reaction was stirred at 80° C. for 2 hours. The reaction solution was poured into ice water. The reaction solution was extracted with ethyl acetate. The organic phase was dried with sodium sulfate and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain tert-butyl (R)-2-chloro-3-fluoro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (0.6 g, purity 8%). The crude product was directly used in the next step. ES-API: [M+H]+=545.2.


Steps 2-4: Compound Z73 was prepared according to the method of steps 3-5 in Example 72. 1H-NMR (400 MHz, DMSO-d6) δ 10.25 (s, 1H), 8.36 (d, J=3.8 Hz, 1H), 7.23-7.22 (m, 1H), 7.15 (d, J=3.8 Hz, 1H), 6.82-6.70 (m, 1H), 6.63-6.61 (m, 2H), 6.22-6.17 (m, 1H), 5.78-5.77 (m, 1H), 4.88-4.80 (m, 2H), 4.46-4.02 (m, 4H), 3.82-3.72 (m, 2H), 3.34-3.32 (m, 1H), 2.77-2.66 (m, 1H), 1.92-1.90 (m, 3H), 1.07-1.06 (m, 3H), 0.94-0.92 (m, 3H). ES-API: [M+H]+=575 0.2.


Step 5: Compound Z73 (15 mg) was resolved chirally (mobile phase: n-hexane:ethanol=60:40; column type: IG (250 mm*4.6 mm*5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z73-1 (5.6 mg, retention time: 10.149 min, purity: 100%, de value: 100%). ES-API: [M+H]+=575.2.


and Compound Z73-2 (6.5 mg, retention time: 13.445 min, purity: 100%, de value: 99%). ES-API: [M+H]+=575.2.


Example 74: Preparation of Compound Z74



embedded image


embedded image


Step 1: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, 1.9 mmol) and a solution of N-chlorosuccinimide (507 mg, 3.8 mmol) in acetonitrile (20 mL) were stirred at 80° C. for 3 hours under nitrogen protection. The reaction solution was filtered, concentrated and purified by a fast silica gel column (0-10% methanol/dichloromethane) to obtain a white solid: tert-butyl (R)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, yield: 94%). ES-API: [M+H]+=561.1.


Step 2: tert-butyl (R)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (300 mg, 0.53 mmol), 2-fluoro-6-methoxyphenylboronic acid (270 mg, 1.60 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (39 mg, 0.05 mmol) and a mixed solution of potassium carbonate (220 mg, 1.60 mmol) in 1,4-dioxane (5 mL) and water (1 mL) reacted under a 120° C. microwave for 2 hours under nitrogen protection. The reaction solution was filtered, washed with ethyl acetate (30 mL), the filtrate was washed with the saturated brine (10 mL*3), the obtained organic phase was dried and concentrated, purified with a fast silica column (0-10% methanol/dichloromethane) to obtain a yellow solid: tert-butyl (5aR)-3-chloro-2-(2-fluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (220 mg, purity: 63%). ES-API: [M+H]+=651.2.


Step 3: tert-butyl (5aR)-3-chloro-2-(2-fluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (200 mg, 0.3 mmol), potassium vinyltrifluoroborate (120 mg, 0.9 mmol), chloro(2-dicyclohexylphosphino-2′,6′-dimethoxy-1,1′-biphenyl)(2′-amino-1,1′-biphenyl-2-yl)palladium(II) (20 mg, 0.03 mmol), 2-bicyclohexylphosphin-2′,6′-dimethoxybiphenyl (12 mg, 0.03 mmol) and a mixed solution of potassium phosphate (190 mg, 0.9 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) reacted under a 150° C. microwave for 2 hours under nitrogen protection. The reaction solution was filtered, washed with ethyl acetate (30 mL), the filtrate was washed with the saturated brine (10 mL*3), the obtained organic phase was dried and concentrated, purified with a fast silica column (0-10% methanol/dichloromethane) to obtain a yellow solid: tert-butyl (5aR)-2-(2-fluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-vinyl-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (120 mg, purity: 35%). ES-API: [M+H]+=643 0.3.


Step 4: 17% boron tribromide in dichloromethane solution (2 mL) was added to a solution of tert-butyl (5aR)-2-(2-fluoro-6-methoxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-vinyl-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (120 mg, 0.19 mmol) in dichloromethane (2 mL) under the ice bath, and stirred at room temperature for 3 hours. The reaction solution was quenched with methanol (5 mL), poured into the saturated sodium bicarbonate solution (20 mL), and extracted with dichloromethane (20 mL*3), and the organic phase was dried and concentrated to obtain a yellow solid: (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-vinyl-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (100 mg). ES-API: [M+H]+=529.2.


Step 5: N,N-diisopropylethylamine (74 mg, 0.57 mmol) was added to a solution of (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-vinyl-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (100 mg, 0.19 mmol) in dichloromethane (2 mL) under the ice bath. After the reaction solution was clarified, acrylic anhydride (17 mg, 0.19 mmol) was dropped, and stirred for 5 minutes. The reaction solution was washed with the saturated sodium bicarbonate solution (5 mL). The organic phase was dried and concentrated, then purified by Preparative HPLC (ammonium bicarbonate system) to obtain a white solid: Z74 (2.4 mg, purity: 94%, yield: 10%). ES-API: [M+H]+=583.2.


Example 75: Preparation of Compound Z75



embedded image


Compound Z75 was prepared according to the method of steps 2-3 in Example 34. ES-API: [M+H]+=605.2.


Example 76: Preparation of Compound Z76, Z76-1 and Z76-2



embedded image


embedded image


Compound Z76 was prepared with (2,4-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=593.1.


Compound Z76 was prepared and resolved chirally (column type: IG, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=40:60, flow rate: 1 ml/min, column temperature=30° C.) to obtain: Compound Z76-1 (50.7 mg, peak 1, retention time 8.084 min, yield: 18%), a white solid. ES-API: [M+H]+=593.1. 1H NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.35 (td, J=10.1, 2.4 Hz, 1H), 7.28-7.11 (m, 3H), 6.96-6.76 (m, 1H), 6.22 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.97-4.82 (m, 2H), 4.65-4.23 (m, 3H), 4.07 (d, J=20.5 Hz, 1H), 3.71 (dd, J=35.3, 22.8 Hz, 2H), 3.37 (s, 1H), 2.70 (dt, J=13.4, 6.7 Hz, 1H), 1.96 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


and Compound Z76-2 (53.4 mg, peak 2, retention time 11.484 min, yield: 19%), a white solid. ES-API: [M-FH]+=593.1. 1H NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.35 (td, J=10.2, 2.4 Hz, 1H), 7.16 (dd, J=9.7, 3.5 Hz, 3H), 6.95-6.76 (m, 1H), 6.22 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.3 Hz, 1H), 5.00-4.79 (m, 2H), 4.64-4.26 (m, 3H), 4.07 (d, J=31.9 Hz, 1H), 3.70 (dd, J=40.4, 28.8 Hz, 2H), 3.38 (s, 1H), 2.77 (dt, J=13.5, 6.8 Hz, 1H), 1.93 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


Example 77: Preparation of Compound Z77, Z77-1 and Z77-2



embedded image


embedded image


Compound Z77 was prepared with (2,3-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=593.1. Compound Z77 was prepared and resolved chirally (column type: IG, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=40:60, flow rate: 1 ml/min, column temperature=30° C.) to obtain: Compound Z77-1 (45.3 mg, peak 1, retention time 7.568 min, yield: 24%), a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.54 (dt, J=17.2, 5.6 Hz, 1H), 7.27 (dd, J=12.5, 7.7 Hz, 1H), 7.17 (d, J=4.9 Hz, 1H), 7.05-6.77 (m, 2H), 6.24 (s, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.90 (d, J=4.6 Hz, 2H), 4.65-4.26 (m, 3H), 4.06 (d, J=29.1 Hz, 1H), 3.72 (dd, J=36.9, 24.2 Hz, 2H), 3.41-3.36 (m, 1H), 2.70 (dt, J=13.4, 6.6 Hz, 1H), 1.96 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.7 Hz, 3H).


and Compound Z77-2 (37.8 mg, peak 2, retention time 9.781 min, yield: 20%), a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.52 (dd, J=17.1, 8.3 Hz, 1H), 7.17 (d, J=5.2 Hz, 2H), 7.07-6.75 (m, 2H), 6.22 (dd, J=16.7, 2.3 Hz, 1H), 5.85-5.72 (m, 1H), 5.03-4.81 (m, 2H), 4.69-4.26 (m, 3H), 4.07 (d, J=32.9 Hz, 1H), 3.71 (dd, J=40.6, 28.9 Hz, 2H), 3.36 (d, J=10.8 Hz, 1H), 2.78 (dt, J=13.3, 6.6 Hz, 1H), 1.93 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


Example 78: Preparation of Compound Z78



embedded image


Compound Z78 was prepared with 2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyridine as the raw material according to the method of Example 46. ES-API: [M+H]+=572.2. 1H NMR (500 MHz, DMSO-d6) δ 8.46 (d, J=3.6 Hz, 1H), 8.35 (d, J=4.8 Hz, 1H), 7.53-7.46 (m, 1H), 7.27 (dd, J=7.7, 4.9 Hz, 1H), 7.17 (d, J=4.9 Hz, 1H), 6.92-6.80 (m, 1H), 6.22 (dd, J=16.7, 2.3 Hz, 1H), 5.79 (dd, J=10.4, 2.3 Hz, 1H), 4.99-4.79 (m, 2H), 4.71-4.24 (m, 3H), 4.08 (d, J=32.5 Hz, 1H), 3.88-3.62 (m, 2H), 3.33-3.29 (m, 1H), 2.78-2.65 (m, 1H), 2.10 (s, 3H), 1.95 (d, J=12.3 Hz, 3H), 1.10-1.00 (m, 3H), 0.89 (dd, J=6.6, 3.6 Hz, 3H).


Example 79: Preparation of Compound Z79



embedded image


Compound Z79 was prepared with 2-chloro-phenylboronic acid as the raw material according to the method of steps 3-5 in Example 72. ES-API: [M+H]+=576.3. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.54 (d, J=8.0 Hz, 1H), 7.47 (td, J=7.8, 1.6 Hz, 1H), 7.40 (dd, J=7.5, 6.6 Hz, 1H), 7.27-7.21 (m, 1H), 7.17 (d, J=4.8 Hz, 1H), 6.92-6.77 (m, 1H), 6.22 (dd, J=16.7, 2.1 Hz, 1H), 5.78 (dd, J=10.4, 2.3 Hz, 1H), 4.97-4.79 (m, 2H), 4.58 (s, 1H), 4.51-4.26 (m, 2H), 4.05 (s, 1H), 3.93-3.66 (m, 2H), 2.78 (dt, J=13.6, 6.8 Hz, 1H), 2.73-2.60 (m, 1H), 1.95 (d, J=17.0 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.94 (dd, J=6.4, 4.8 Hz, 3H).


Example 80: Preparation of Compound Z80



embedded image


Compound Z80 was prepared according to the method of Example 46. ES-API: [MA-]+=594.2. 1H NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=4.8 Hz, 1H), 7.62-7.47 (m, 1H), 7.30-7.08 (m, 3H), 6.94-6.75 (m, 1H), 6.22 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.99-4.78 (m, 2H), 4.49-4.43 (m, 2H), 4.33-4.26 (m, 2H), 4.16-3.95 (m, 1H), 3.82-3.61 (m, 2H), 2.88-2.63 (m, 1H), 1.92 (d, J=17.0 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H), 0.91 (t, J=6.0 Hz, 3H).


Example 81: Preparation of Compound Z81



embedded image


Compound Z81 was prepared with 4-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyridine as the raw material according to the method of Example 46. ES-API: [M+H]+=572.2.


Example 82: Preparation of Compound Z82, Z82-1 and Z82-2



embedded image


Compound Z82 was prepared with (3-chloro-2-fluorophenyl)boronic acid as the raw material according to the method of Example 46, resolved chirally (column type: Chiralpak IG 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol=50:50; flow rate: 1 ml/min; column temperature=30° C.) to obtain: Compound Z82-1 (13 mg, retention time: 9.805 min; peak 1, yield 4.1%). 1H NMR (400 MHz, DMSO-d6) δ 8.37 (d, J=4.8 Hz, 1H), 7.68 (t, J=6.8 Hz, 1H), 7.28 (d, J=8.2 Hz, 1H), 7.16 (dd, J=13.4, 6.5 Hz, 2H), 6.84 (d, J=14.1 Hz, 1H), 6.22 (dd, J=16.4, 2.1 Hz, 1H), 5.80-5.77 (m, 1H), 4.99-4.81 (m, 2H), 4.65-4.25 (m, 3H), 4.15-3.97 (m, 1H), 3.75-3.48 (m, 2H), 3.32 (s, 1H), 2.82-2.75 (m, 1H), 1.93 (s, 3H) 1.07 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.7 Hz, 3H).


and Compound Z82-2 (8 mg, retention time: 13.724 min; peak 2, yield 2.5%).


Example 83: Preparation of Compound Z83, Z83-1 and Z83-2



embedded image


embedded image


Compound Z83 was prepared with (2-fluoro-6-methoxyphenyl)boronic acid as the raw material according to the method of steps 3-5 in Example 72, resolved chirally (column type: Chiralpak IC 250 mm*4.6 mm*5 um mobile phase: acetonitrile:isopropanol=60:40; flow rate: 1 ml/min; column temperature=30° C.) to obtain Compound Z83-1 (8 mg, retention time: 5.005 min; peak 1, yield 2.6%) and Compound Z83-2 (8 mg, retention time: 6.658 min; peak 2, yield 2.6%).


Example 84: Preparation of Compound Z84



embedded image


Compound Z84 was prepared according to the method of steps 3-5 in Example 72. ES-API: [M+H]+=559.2.


Example 85: Preparation of Compound Z85



embedded image


Compound Z85 was prepared according to the method of steps 3-5 in Example 72. ES-API: [M+H]+=575.2. 1H-NMR (500 MHz, DMSO-d6) δ 10.16 (s, 1H), 8.36 (d, J=4.8 Hz, 1H), 7.25 (dd, J=15.4, 8.2 Hz, 1H), 7.16 (d, J=4.9 Hz, 1H), 6.99-6.78 (m, 1H), 6.74-6.63 (m, 2H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.3 Hz, 1H), 4.96-4.77 (m, 2H), 4.60 (s, 1H), 4.39 (dd, J=50.2, 19.6 Hz, 2H), 4.05 (s, 1H), 3.75 (dd, J=38.5, 27.0 Hz, 2H), 3.38 (s, 1H), 2.83-2.63 (m, 1H), 1.92 (d, J=14.2 Hz, 3H), 1.07 (d, J=6.7 Hz, 3H), 0.94 (t, J=7.1 Hz, 3H).


Example 86: Preparation of Compound Z86



embedded image


Compound Z86 was prepared with (5-methyl-1H-indazol-4-yl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=611.2. 1HNMR (400 MHz, DMSO-d6) δ 13.08 (s, 1H), 8.28 (t, J=4.5 Hz, 1H), 7.45 (t, J=7.8 Hz, 2H), 7.25-7.01 (m, 2H), 6.90 (d, J=26.1 Hz, 1H), 6.23 (dd, J=16.7, 2.1 Hz, 1H), 5.79 (dd, J=10.5, 2.2 Hz, 1H), 5.00-4.81 (m, 2H), 4.67-4.29 (m, 3H), 4.08 (s, 1H), 3.71 (s, 2H), 3.38 (d, J=10.1 Hz, 1H), 2.84-2.70 (m, 1H), 1.96 (dd, J=17.1, 12.1 Hz, 6H), 1.10-0.72 (m, 6H).


Example 87: Preparation of Compound Z87, Z87-1 and Z87-2



embedded image


embedded image


Compound Z87 was prepared with 2-fluorophenylboronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=575.2. 290 mg Compound Z87 was prepared and resolved chirally (column type: IG, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=40:60, flow rate: 1 ml/min, column temperature=30° C.) to obtain: Compound Z87-1 (93.98 mg, peak 1, retention time 8.384 min, yield: 32.4%), a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.9 Hz, 1H), 7.70-7.42 (m, 2H), 7.37-7.12 (m, 2H), 7.05-6.76 (m, 2H), 6.21 (dd, J=16.6, 2.4 Hz, 1H), 5.77 (dd, J=10.4, 2.4 Hz, 1H), 5.03-4.83 (m, 2H), 4.69-4.22 (m, 3H), 4.05 (d, J=29.7 Hz, 1H), 3.87-3.60 (m, 2H), 3.39-3.34 (m, 1H), 2.70 (p, J=6.7 Hz, 1H), 1.95 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.7 Hz, 3H).


and Compound Z87-2 (95.37 mg, peak 2, retention time 11.618 min, yield: 32.7%), a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J=4.8 Hz, 1H), 7.46-7.35 (m, 1H), 7.26-7.04 (m, 4H), 6.89-6.69 (m, 1H), 6.14 (dd, J=16.7, 2.3 Hz, 1H), 5.71 (dd, J=10.4, 2.2 Hz, 1H), 4.94-4.73 (m, 2H), 4.60-4.15 (m, 3H), 4.11-3.92 (m, 1H), 3.76-3.50 (m, 2H), 3.39-3.34 (m, 1H), 2.71 (dt, J=13.5, 6.8 Hz, 1H), 1.86 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H).


Example 88: Preparation of Compound Z88, Z88-1 and Z88-2



embedded image


embedded image


Compound Z88 was prepared with (2-fluoro-6-hydroxyphenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=592.2. Compound Z88 was prepared and resolved chirally (column type: IG, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=65:35, flow rate: 1 ml/min, column temperature=30° C.) to obtain Compound Z88-1 (4.83 mg, peak 1, retention time 10.605 min, yield: 10.7%), a white solid. and Compound Z88-2 (13.64 mg, peak 2, retention time 14.649 min, yield: 30.3%), a white solid.


Example 89: Preparation of Compound Z89



embedded image


Step 1: morpholine (17.4 mg, 0.2 mmol) and 60% NaH (12 mg, 0.3 mmol) was dissolved in dry DMF (3 ml) under the ice water bath, stirred for 10 minutes, tert-butyl (R)-2,3-dichloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (56 mg, 0.1 mmoL) was slowly added, the reaction rised to room temperature and proceeded for 1 hour. The reaction solution was poured into ice water, washed with water and the saturated brine, concentrated, and purified by column chromatography to obtain tert-butyl (R)3-chloro-2-(morpholin)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (30 mg, yield 49%). ES-API: [M+H]+=612.3.


Steps 2-3: Compound Z89 was prepared according to the corresponding steps of Example 34. ES-API: [M+H]+=566.2.


Example 90: Preparation of Compound Z90



embedded image


Compound Z90 was prepared with (2-chlorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=592.2. 1H NMR (500 MHz, DMSO-d6) δ 8.27 (d, J=4.8 Hz, 1H), 7.44 (s, OH), 7.41-7.26 (m, 2H), 7.09 (t, J=6.7 Hz, 2H), 6.89-6.64 (m, 1H), 6.14 (dd, J=16.8, 2.3 Hz, 1H), 5.71 (dd, J=10.4, 2.3 Hz, 1H), 4.83 (dd, J=23.8, 12.6 Hz, 2H), 4.61-4.35 (m, 2H), 4.33-4.13 (m, 1H), 3.97 (s, 1H), 3.59 (s, 2H), 3.34-3.21 (m, 1H), 2.67 (d, J=31.0 Hz, 1H), 1.87 (d, J=10.4 Hz, 3H), 1.00 (d, J=6.6 Hz, 3H), 0.86 (d, J=6.4 Hz, 3H).


Example 91: Preparation of Compound Z91, Z91-1 and Z91-2



embedded image


embedded image


Compound Z91 was prepared with (3-chloro-2-fluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=610.3. Compound Z91 (274 mg, 0.45 mmol) was resolved chirally (mobile phase: n-hexane-ethanol-40-60); column type: IG (250 mm*4.6 mm 5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z91-1 (103 mg, retention time: 8.228 min, purity: 99%, de value: 99%). ES-API: [M+H]+=610.3.


and Compound Z91-2 (109 mg, retention time: 10.820 min, purity: 99%, de value: 99%). ES-API: [M+H]+=610.3. 1H-NMR (500 MHz, DMSO-d6) δ 8.36 (d, J=4.8 Hz, 1H), 7.68 (t, J=7.4 Hz, 1H), 7.29 (t, J=8.0 Hz, 1H), 7.24-7.10 (m, 2H), 7.06-6.71 (m, 1H), 6.22 (dd, J=16.8, 2.0 Hz, 1H), 5.78 (dd, J=10.4, 2.0 Hz, 1H), 5.07-4.75 (m, 2H), 4.70-4.22 (m, 3H), 4.16-3.98 (m, 1H), 3.84-3.57 (m, 2H), 3.40-3.29 (m, 1H), 2.89-2.63 (m, 1H), 1.94 (d, J=12.0 Hz, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.94 (d, J=6.6 Hz, 3H).


Example 92: Preparation of Compound Z92



embedded image


embedded image


Step 1: 4-isopropyl-2-methylpyridin-3-amine (1 g, 6.7 mmol) and 30 mL of tetrahydrofuran were added to a 250 mL round bottom flask. After the system cooling to 0° C., sodium bis(trimethylsilyl)amide (8.3 mL, 2 M in tetrahydrofuran, 16.5 mmol) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 10 minutes. and then a solution of 4,6-dichloro-2-fluoronicotinamide (1.4 g, 6.7 mmol) in tetrahydrofuran (30 mL) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 20 minutes. After the completion of the reaction, the reaction solution was poured into ice water. The reaction solution was extracted with ethyl acetate. The organic phase was washed with 1M hydrochloric acid, saturated aqueous sodium bicarbonate solution, and saturated brine. The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated and dried to obtain the product: 4,6-dichloro-2-(((4-isopropyl-2-methylpyridin-3-yl)amino)nicotinamide (2.1 g, 90%), a yellow solid. ES-API: [M+H]+=340.1.


Step 2: 4,6-dichloro-2-(((4-isopropyl-2-methylpyridin-3-yl)amino)nicotinamide (2.1 g, 6.0 mmol) and 80 mL of dry tetrahydrofuran were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, sodium hydride (1.2 g, 30 mmol) was added in batches, and the reaction proceeded at this temperature for 10 minutes. A suspension of CDI (1.9 g, 12 mmol) in tetrahydrofuran (40 mL) was dropped to the above solution, and then the reaction proceeded at this temperature for 20 minutes. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 150 mL of ice water, and the pH was adjusted to about 4 with 3 M hydrochloric acid. The reaction solution was extracted with ethyl acetate. The organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, filtered. The filtrate was concentrated and dried to obtain 5,7-dichloro-1-(4-isopropyl-2-methylpyridin-3-yl)pyridine[2,3-d]pyrimidin-2,4 (1H,3H)-dione (1.49 g), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=366.2.


Step 3: tert-butyl (R)-3-(hydroxymethyl)piperazin-1-carboxylate (885 mg, 4.1 mmol) was added to a suspension of 60% sodium hydride (676 mg, 16.9 mmol) in tetrahydrofuran (20 mL) at 0° C. a solution of 5,7-dichloro-1-(4-isopropyl-2-methylpyridin-3-yl)pyridine[2,3-d]pyrimidin-2,4 (1H,3H)-dione (1.49 g, 4.1 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 30 minutes. The completion of the reaction was detected by LC-MS. The reaction solution was poured into 100 mL of ice water. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 20-100%) to obtain tert-butyl (S)-3-(((7-chloro-1-(4-isopropyl-2-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (1.34 g, 60%), a yellow solid. ES-API: [M+H]+=546.3.


Step 4: tert-butyl (S)-3-(((7-chloro-1-(4-isopropyl-2-methylpyridin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (1.34 g, 2.45 mmol), diisopropylethylamine (20 mL), dichloromethane (30 mL) were added to a round bottom flask. propylphosphonic anhydride solution (20 mL, 50% w/w ethyl acetate solution was dropped thereto. The reaction was stirred at room temperature for 30 minutes. The completion of the reaction was detected by LC-MS. 50 mL of dichloromethane was added to the reaction, and the organic phase was washed with 30 mL of hydrochloric acid (1M) and 100 mL of the saturated sodium bicarbonate aqueous solution. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain tert-butyl (R)-2-chloro-12-(4-isopropyl-2-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (903 mg, 70%). ES-API: [M+H]+=527.3.


Steps 5-8: Compound Z92 was prepared according to the method of Example 73. ES-API: [M+H]+=575.2.


Example 93: Preparation of Compound Z93



embedded image


embedded image


embedded image


Step 1: 2,4-dichloro-6-fluorobenzoic acid (15 g, 71.4 mmoL) was dissolved in 200 mL of dichloromethane and cooled to 0° C., oxalyl chloride (12 ml, 142.9 mmol) was added. After 30 minutes for the completion of the dropping, DMF (0.5 ml) was dropped, the reaction rised to room temperature and proceeded for 1 hour. After the completion of the reaction, the reaction solution was concentrated and diluted with 150 mL of dichloromethane, cooled to 0° C., slowly dropped to 20 mL of ammonia water, and stirred for 2 hours. After the completion of the reaction, the reaction solution was concentrated and slurried with ethyl acetate, filtered to obtain the crude product: 2,4-dichloro-6-fluorobenzamide (13.5 g, yield 90%). ES-API: [M+H]+=208.9.


Step 2: 4,6-dicyclopropylpyrimidin-5-amine (3.56 g, 20.3 mmol) was dissolved in 60 mL of tetrahydrofuran, LiHMDS (38.2 ml, 38.2 mmol) was dropped under the ice water bath under nitrogen protection, stirred for 30 minutes, 2,4,6-nicotinamide (4 g, 19.1 mmol) dissolved in 30 mL of tetrahydrofuran was added to the above reaction solution, slowly rised to room temperature, reacted for 2 hours, cooled under the ice water bath, and dilute hydrochloric acid was dropped to pH of 7-8. The reaction solution was extracted with ethyl acetate, concentrated to obtain a gray solid, slurried with ethyl acetate, and filtered to obtain the crude product: 4,6-dichloro-2-(((4,6-dicyclopropylpyrimidin-5-yl)amino)nicotinamide (6.5 g, yield 85%). ES-API: [M+H]+=364.0.


Step 3: 4,6-dichloro-2-(((4,6-dicyclopropylpyrimidin-5-yl)amino)nicotinamide (6.5 g, 17.9 mmoL) was dissolved in 120 ml of tetrahydrofuran, 60% NaH (3.6 g, 89.5 mmol) was added under the ice water bath, stirred for 15 minutes, then CDI (4.35 g, 26.6 mmol) was added, the reaction proceeded under the ice water bath for 1 hour, raw material disappeared, the reaction solution was poured into ice water, and the pH was adjusted to 7-8 with dilute hydrochloric acid. The reaction solution was extracted with ethyl acetate, washed with water and the saturated brine in sequence, concentrated under reduced pressure to obtain 5,7-dichloro-1-(4,6-dicyclopropylpyrimidin-5-yl)pyrido[2,3-d]pyrimidin-2,4(1H, 3H)-dione (5.2 g, yield 74%). ES-API: [M+H]+=390.0.


Step 4: tert-butyl (R)-3-(hydroxymethyl)piperazin-1-carboxylate (2.83 g, 8.48 mmoL) was dissolved in tetrahydrofuran, 60% NaH (1.02 g, 25.4 mm1) was added under the ice water bath, and stirred for 10 minutes. 5,7-dichloro-1-(4,6-dicyclopropylpyrimidin-5-yl)pyrido[2,3-d]pyrimidin-2,4(1H, 3H)-dione (4.3 g, 8.48 mmoL) was added, and stirred for 20 minutes. After the completion of the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate for 3 times, the organic phases was combined, dried and concentrated under reduced pressure, purified by column chromatography (methanol/dichloromethane: 0-10%) to obtain tert-butyl (R)-3-((((7-chloro-1-(4,6-dicyclopropylpyrimidin-5-yl)-2,4-dioxo-1,2,3,4-tetrahydropyridine[2,3-d]pyrimidin-5-yl)oxy)methyl)piperazin-1-formate (5.7 g, yield 92%). ES-API: [M+H]+=570.2.


Step 5: tert-butyl (R)-3-((((7-chloro-1-(4,6-dicyclopropylpyrimidin-5-yl)-2,4-dioxo-1,2,3,4-tetrahydropyridine[2,3-d]pyrimidin-5-yl)oxy)methyl)piperazin-1-formate (5.7 g, 10.0 mmoL) was dissolved in dichloromethane, diisopropylethylamine (15.5 g, 120.0 mm1) was added at room temperature, and stirred for 5 minutes. 1-propylphosphonic anhydride (19 g, 30.0 mmoL) was added, stirred for 20 minutes. After the completion of the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate for 3 times, the organic phases was combined, washed with the saturated brine, dried and concentrated under reduced pressure, and purified by column chromatography (methanol/dichloromethane: 0-10%) to obtain tert-butyl (R)2-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (3.8 g, yield 69%). ES-API: [M+H]+=552.2.


Step 6: tert-butyl (R)-2-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexa hydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7(5H)-carboxylate (550 mg, 1.0 mmoL) was dissolved in 10 mL of acetonitrile, N-chlorosuccinimide (266 mg, 2.0 mmol) was added, heated to 80° C. and reacted for 1 hour, cooled to room temperature, filtered, washed with water and the saturated brine, concentrated, and purified by column chromatography to obtain tert-butyl (R)-2,3-dichloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthale n-7 (5H)-carboxylate (530 mg, yield 90%). ES-API: [M+H]+=586.2.


Step 7: tert-butyl (R)-2,3-dichloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (180 mg, 0.307 mmoL), (2-fluorophenyl)boronic acid (215 mg, 1.54 mmol), SPhos-Pd-G2 (82 mg, 0.114 mmol) and potassium carbonate (472 mg, 3.42 mmol) was dissolved in 8 mL of dioxane and 2 mL of water, nitrogen was used for replacement, the reaction proceeded at 80° C. for 1 hour, cooled to room temperature, filtered, diluted with ethyl acetate, washed with water and the saturated brine, concentrated, and purified by column chromatography to obtain tert-butyl (R)3-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-2-(2-fluorophenyl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (120 mg, yield: 60%). ES-API: [M+H]+=646.2.


Step 8: tert-butyl (R)-3-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-2-(2-fluorophenyl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (120 mg, 0.186 mmoL) was dissolved in 10 mL of dichloromethane, 5 mL of trifluoroacetic acid was slowly dropped at room temperature, reacted for 5 hours, then 10 mL of dichloromethane was added. The reaction solution was concentrated under reduced pressure to obtain (R)-3-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-2-(2-fluorophenyl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-1-one (180 mg, the crude product), directly used in the next step. ES-API: [M+H]+=546.2


Step 9: (R)-3-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-2-(2-fluorophenyl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-1-one (180 mg, the crude product) was dissolved in dichloromethane, diisopropylethylamine (255 mg, 1.98 mmol) and acrylic anhydride (83 mg, 0.66 mmol) was dropped under the ice water bath, and reacted under the ice bath for 10 minutes. After the completion of the reaction, the reaction solution was concentrated at room temperature under reduced pressure, and purified by preparative HPLC to obtain Compound Z93 (40 mg, yield 27%), ES-API: [M+H]+=600.2. 1H NMR (500 MHz, DMSO-d6) δ 8.62 (s, 1H), 7.48-7.41 (m, 1H), 7.27-7.19 (m, 2H), 7.16 (m, 1H), 6.79 (m, 1H), 6.14 (dd, J=16.7, 2.4 Hz, 1H), 5.71 (dd, J=10.4, 2.3 Hz, 1H), 4.85 (t, J=13.3 Hz, 1H), 4.77 (dd, J=12.8, 5.2 Hz, 1H), 4.52 (d, J=23.8 Hz, 1H), 4.42 (d, J=13.1 Hz, 1H), 4.22 (s, 1H), 3.99 (d, J=31.2 Hz, 1H), 3.78-3.62 (m, 1H), 3.62-3.52 (m, 1H), 3.25 (d, J=5.8 Hz, 1H), 1.73 (m, 1H), 1.65 (s, 1H), 1.05-0.65 (m, 8H).


Example 94: Preparation of Compound Z94



embedded image


Compound Z94 was prepared with (2-hydroxy-6-fluorophenyl)boronic acid as the raw material according to the method of steps 7-9 in Example 93. ES-API: [M+H]+=617.1. 1H NMR (500 MHz, DMSO-d6) δ 10.06 (d, J=4.7 Hz, 1H), 8.67 (s, 1H), 7.25-7.21 (m, 1H), 6.92-6.80 (m, 1H), 6.71 (dd, J=8.3, 4.3 Hz, 1H), 6.66 (m, 1H), 6.21 (dd, J=16.7, 2.4 Hz, 1H), 5.78 (dd, J=10.3, 2.4 Hz, 1H), 4.96-4.80 (m, 2H), 4.60 (d, J=20.4 Hz, 1H), 4.43 (d, J=59.7 Hz, 1H), 4.27 (s, 1H), 4.04 (s, 1H), 3.83-3.59 (m, 2H), 3.33-3.31 (m, 1H), 1.73 (d, J=48.5 Hz, 2H), 1.02-0.73 (m, 8H).


Example 95: Preparation of Compound Z95, Z95-1 and Z95-2



embedded image


embedded image


embedded image


Step 1: 4-isopropyl-6-methylpyrimidin-5-amine (2.8 g, 19.1 mmol) and 45 mL of tetrahydrofuran were added to a 250 mL round bottom flask. After the system cooling to 0° C., sodium bis(trimethylsilyl)amide (28.5 mL, 2 M in tetrahydrofuran, 57.3 mmol) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 10 minutes. and then 4,6-dichloro-2-fluoronicotinamide (4 g, 19.1 mmol) in tetrahydrofuran (15 mL) was dropped to the reaction solution. After the completion of the dropping, the reaction was stirred at 0° C. for 20 minutes. After the completion of the reaction, the reaction solution was poured into ice water. The reaction solution was extracted with ethyl acetate. The organic phase was washed with 1M hydrochloric acid, saturated aqueous sodium bicarbonate solution, and saturated brine. The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated and dried to obtain the product: 4,6-dichloro-2-(((4-isopropyl-6-methylpyrimidin-5-yl)amino)nicotinamide (5.7 g, 87%), a yellow solid. ES-API: [M+H]+=340.1.


Step 2: 4,6-dichloro-2-(((4-isopropyl-6-methylpyrimidin-5-yl)amino)nicotinamide (5.7 g, 16.7 mmol) and 80 mL of dry tetrahydrofuran were added to a 250 mL three-necked round bottom flask, cooled to 0-5° C. under the ice bath, sodium hydride (3 g, 75.1 mmol) was added in batches, and the reaction proceeded at this temperature for 5 minutes. A suspension of CDI (4 g, 24.7 mmol) in tetrahydrofuran (40 mL) was dropped to the above solution, and then the reaction proceeded at this temperature for 10 minutes. The completion of the reaction was detected by LCMS. The reaction solution was poured into about 150 mL of ice water, and the pH was adjusted to about 4 with 3 M hydrochloric acid. The reaction solution was extracted with ethyl acetate. The organic phase was washed with the saturated brine, dried with anhydrous sodium sulfate, filtered. The filtrate was concentrated and dried to obtain 5,7-dichloro-1-(4-isopropyl-6-methylpyrimidin-5-yl)pyrido[2,3-d]pyrimidin-2,4(1H,3H)-dione (4.5 g, purity 70%), a yellow solid, the crude product was directly used in the next step. ES-API: [M+H]+=366.1


Step 3: tert-butyl (R)-3-(hydroxymethyl)piperazin-1-carboxylate (3.2 g, 14.8 mmol) was added to a suspension of 60% sodium hydride (1.5 g, 36.9 mmol) in tetrahydrofuran (60 mL) at 0° C. A solution of 5,7-dichloro-1-(4-isopropyl-6-methylpyrimidin-5-yl)pyrido[2,3-d]pyrimidin-2,4 (1H,3H)-dione (4.5 g, 12.3 mmol) in tetrahydrofuran (20 mL) was dropped thereto. The reaction was stirred at 0° C. for 30 minutes. The completion of the reaction was detected by LC-MS. The reaction solution was poured into 100 mL of ice water. The reaction solution was extracted with ethyl acetate for 3 times. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 20-100%) to obtain tert-butyl (R)-3-((((7-chloro-1-(4-isopropyl-6-methylpyrimidin-5-yl)-2,4-dioxo-1,2,3,4-tetrahydropyridinyl[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (3 g, 44%), a yellow solid. ES-API: [M+H]+=546.3


Step 4: tert-butyl (R)-3-((((7-chloro-1-(4-isopropyl-6-methylpyrimidin-5-yl)-2,4-dioxo-1,2,3,4-tetrahydropyridinyl[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (3 g, 5.5 mmol), diisopropylethylamine (10 mL), dichloromethane (30 mL) were added to a round bottom flask. 1-propylphosphonic anhydride solution (10 mL, 50% w/w ethyl acetate solution) was dropped thereto. The reaction was stirred at room temperature for 30 minutes. The completion of the reaction was detected by LC-MS. 50 mL of dichloromethane was added to the reaction, and the organic phase was washed with 30 mL of hydrochloric acid (1M) and 100 mL of the saturated sodium bicarbonate aqueous solution. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain tert-butyl (R)-2-chloro-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]-naphthalen-7 (5H)-carboxylate (2.3 g, 79%). ES-API: [M+H]+=528.3.


Step 5: tert-butyl (R)-2-chloro-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]-naphthalen-7 (5H)-carboxylate (500 mg, 0.9 mmol), N-chlorosuccinimide (252 mg, 1.9 mmol) and acetonitrile (10 mL) were added to a round bottom flask. The reaction was stirred at 80° C. for 2 hours. Sodium thiosulfate aqueous solution was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (methanol/dichloromethane: 0-3%) to obtain the product: tert-butyl (R)-2,3-dichloro-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (470 mg, 88%), a yellow solid. ES-API: [M+H]+=562.2.


Step 6: tert-butyl (R)-2,3-dichloro-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (200 mg, 0.36 mmol), 2-fluoro-phenylboronic acid (124 mg, 0.89 mmol), Pd(PPh3)4 (42 mg, 0.036 mmol), sodium carbonate (114 mg, 1.08 mmol), 18 mL of dioxane and 2 mL of water were added to a 100 mL reaction flask. The reaction was stirred at 80° C. for 2.5 hours under nitrogen protection, and the reaction stopped. 20 mL of water was added to the reaction solution. The reaction solution was extracted with 20 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (200 mg, 89%), a yellow solid. ES-API: [M+H]+=622.2.


Step 7: tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-isopropyl-6-methylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (170 mg, 0.27 mmol), 1 mL of methanol and 3 mL of hydrogen chloride/dioxane solution (4 M) were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain (R)-3-chloro-2-(2-fluorophenyl)-12-(4-isopropyl-6-methylpyrimidin-5-yl)-5,5a, 6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]-naphthalen-11 (12H)-one (167 mg), a yellow solid. the crude product was directly used in the next step. ES-API: [M+H]+=522.2.


Step 8: (R)-3-chloro-2-(2-fluorophenyl)-12-(4-isopropyl-6-methylpyrimidin-5-yl)-5,5a, 6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]-naphthalen-11 (12H)-one (167 mg, 0.32 mmol), 3 mL of dichloromethane and triethylamine (97 mg, 0.96 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (40 mg, 0.32 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 40 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated. The crude product was purified by Preparative HPLC to obtain a racemate Z95.


Step 9: Compound Z95 was resolved chirally (column type: Chiralpak IF 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol=40:60; flow rate: 1 ml/min) to obtain: Compound Z95-1 (22.3 mg, retention time: 12.6 min; peak 1), a white solid. ES-API: [M+H]+=576.2.


and Compound Z95-2 (31.2 mg, retention time: 17.3 min; peak 2), a white solid. ES-API: [M+H]+=576.2. 1HNMR (500 MHz, DMSO-d6) 8.93 (s, 1H), 7.51-7.50 (m, 1H), 7.48-7.17 (m, 3H), 6.88-6.80 (m, 1H), 6.23-6.20 (m, 1H), 5.77 (dd, J=10.5, 2 Hz, 1H), 4.87-4.84 (m, 2H), 4.47-4.31 (m, 3H), 4.05-4.02 (m, 1H), 3.68-3.36 (m, 3H), 2.82-2.80 (m, 1H), 2.13 (s, 3H), 1.08 (d, J=6.5 Hz, 3H), 0.95 (d, J=6.5 Hz, 3H).


Example 96: Preparation of Compound Z96, Z96-1 and Z96-2



embedded image


embedded image


Compound Z96 was prepared with (2,3-difluorophenyl)boronic acid as the raw material according to the method of steps 6-8 in Example 95, resolved chirally (column type: Chiralpak IF 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol=40:60; flow rate: 1 ml/min) to obtain: Compound Z96-1 (25 mg, retention time: 12.02 min; peak 1), a white solid. ES-API: [M+H]+=594.1.


and Compound Z96-2 (37 mg, retention time: 17.03 min; peak 2), a white solid. ES-API: [M+H]+=594.1. 1HNMR (500 MHz, DMSO-d6) 8.94 (s, 1H), 7.56-7.54 (m, 1H), 7.53-7.52 (m, 1H), 7.26-7.24 (m, 1H), 6.86-6.80 (m, 1H), 6.23-6.20 (m, 1H), 5.79-5.77 (m, 1H), 4.88-4.84 (m, 2H), 4.47-4.32 (m, 3H), 4.02-4.01 (m, 1H), 3.72-3.66 (m, 2H), 3.34-3.33 (m, 1H), 2.82-2.80 (m, 1H), 2.12 (s, 3H), 1.07 (d, J=6.5 Hz, 3H), 0.95 (d, J=6.5 Hz, 3H).


Example 97: Preparation of Compound Z97



embedded image


Step 1: tert-butyl (R)-2-chloro-12-(4,6-diisopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (450 mg, 0.81 mmol) and a solution of N-chlorosuccinimide (216 mg, 1.62 mmol) in acetonitrile (20 mL) was stirred at 80° C. for 2 hours under nitrogen protection. The reaction solution was dissolved in ethyl acetate (50 mL), washed with water (30 mL*2). The organic phase was dried and concentrated, and purified by a fast silica column (0-10% methanol/dichloromethane) to obtain a yellow solid: tert-butyl (R)-2,3-dichloro-12-(4,6-diisopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (350 mg, yield: 73%). ES-API: [M+H]+=590.2.


Steps 2-4: Compound Z97 was prepared according to the method of Example 46. 1H NMR (500 MHz, DMSO-d6) δ 10.04 (s, 1H), 9.01 (s, 1H), 7.21 (dd, J=15.6, 8.1 Hz, 1H), 6.99-6.78 (m, 1H), 6.68 (dd, J=8.3, 3.3 Hz, 1H), 6.66-6.61 (m, 1H), 6.22 (dd, J=16.7, 2.1 Hz, 1H), 5.78 (dd, J=10.5, 2.2 Hz, 1H), 4.96-4.85 (m, 2H), 4.66-4.44 (m, 2H), 4.41-4.25 (m, 2H), 4.13-3.99 (m, 1H), 3.84-3.65 (m, 2H), 2.83-2.74 (m, 1H), 2.73-2.63 (m, 1H), 1.08 (d, J=6.7 Hz, 6H), 0.97-0.78 (m, 6H). ES-API: [M+H]+=620.2.


Example 98: Preparation of Compound Z98



embedded image


Compound Z98 was prepared with 4,6-diisopropylpyrimidin-5-amine as the raw material according to the method of Example 92. 1H-NMR (500 MHz, DMSO-d6) δ 10.15 (s, 1H), 9.02 (s, 1H), 7.24 (dd, J=15.3, 8.2 Hz, 1H), 6.94-6.76 (m, 1H), 6.71 (d, J=8.3 Hz, 1H), 6.66 (t, J=8.9 Hz, 1H), 6.22 (d, J=16.8 Hz, 1H), 5.83-5.75 (m, 1H), 4.91 (t, J=11.0 Hz, 1H), 4.84 (dd, J=12.7, 5.2 Hz, 1H), 4.57 (s, 1H), 4.49-4.28 (m, 2H), 4.08-3.98 (m, 1H), 3.94-3.66 (m, 2H), 3.50-3.38 (m, 1H), 2.78 (dt, J=13.4, 6.9 Hz, 1H), 2.67 (dd, J=13.4, 6.6 Hz, 1H), 1.11-1.05 (m, 6H), 0.94 (dd, J=18.8, 6.7 Hz, 6H). ES-API: [M+H]+=604.2.


Example 99: Preparation of Compound Z99



embedded image


Step 1: tert-butyl (R)2-chloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (2.0 g, 3.63 mmoL) was dissolved in 100 mL of acetonitrile, Selectfluor (6.64 g, 18.2 mmol) was added, heated to 55° C. and reacted for 18 hours, then cooled to room temperature, filtered, washed with water and the saturated brine, concentrated, and purified by column chromatography to obtain tert-butyl (R)-2,3-dichloro-12-(4,6-dicyclopropylpyrimidin-5-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthale n-7 (5H)-carboxylate (2.0 g, the crude product). ES-API: [M+H]+=570.2.


Steps 2-4: Compound Z99 was prepared according to the method of steps 7-9 in Example 93. ES-API: [M+H]+=600.1. 1H NMR (500 MHz, DMSO-d6) δ 10.17 (s, 1H), 8.69 (s, 1H), 7.27 (m, 1H), 6.85 (m, 1H), 6.77-6.65 (m, 2H), 6.21 (dd, J=16.7, 2.4 Hz, 1H), 5.78 (dd, J=10.3, 2.4 Hz, 1H), 4.89 (t, J=12.2 Hz, 1H), 4.82 (dd, J=12.8, 5.2 Hz, 1H), 4.59 (s, 1H), 4.41 (dd, J=56.1, 13.1 Hz, 1H), 4.29 (s, 1H), 4.02 (s, 1H), 3.83 (d, J=41.6 Hz, 1H), 3.69 (dd, J=25.9, 13.6 Hz, 1H), 3.42 (d, J=12.9 Hz, 1H), 1.77 (m, 1H), 1.70-1.62 (m, 1H), 1.04-0.84 (m, 6H), 0.79 (q, J=8.1 Hz, 2H).


Example 100: Preparation of Compound Z100, Z100-1 and Z100-2



embedded image


Step 1: tert-butyl (R)-2-chloro-3-fluoro-12-(2-isopropyl-4-methylpyrid-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (3.2 g, 5.87 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (5.5 g, 35.25 mmol), SPhos-pd-G2 (1.0 g, 1.1 mmol), potassium phosphate (2.5 g, 11.79 mmol), 100 mL of dioxane and 20 mL of water were added to a 250 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction proceeded at 120° C. for 3 hours. 100 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (5 aR)-3-fluoro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (350 mg, yield: 3%). ES-API: [M+H]+=621.3.


Step 2: tert-butyl (5 aR)-3-fluoro-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (150 mg, 0.24 mol) was dissolved in acetonitrile (8 mL), deuterated methyl iodide (300 mg, 2.11 mmol) was added, and the reaction proceeded at 80° C. for 1 hour. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (5 aR)-3-fluoro-2-(2-fluoro-6-(methoxy-d3)phenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (120 mg, yield: 78%) ES-API: [M+H]+=638.3.


Step 3: tert-butyl (5 aR)-3-fluoro-2-(2-fluoro-6-(methoxy-d3)phenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction solution was concentrated to obtain the target intermediate.


Step 4: the intermediate was dissolved in dichloromethane (15 mL), and triethylamine (1 g, 10 mmol) was added. The reaction was cooled to 0° C., and acrylic anhydride (120 mg, 0.95 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 40 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 50 mL of the saturated NaHCO3 aqueous solution, 40 mL of the saturated brine, dried and concentrated, the crude product was purified by preparative liquid chromatography to obtain the target product: Z100 (55 mg, yield: 48%). ES-API: [M+H]+=592.2.


Step 5: Compound Z100 was prepared and resolved chirally (column type: IC, 250 mm*4.6 mm*5 um, mobile phase: acetonitrile:isopropanol=60:40, flow rate: 1 ml/min, column temperature=30° C.) to obtain: Compound Z100-1 (13 mg, peak 1, retention time 5.065 min, yield: 23.6%), a white solid; and Compound Z100-2 (7 mg, peak 2, retention time 6.804 min, yield: 12.7%), a white solid.


Example 101: Preparation of Compound Z101, Z101-1 and Z101-2



embedded image


Compound Z101 was prepared according to the method of Example 99, and resolved chirally (column type: Chiralpak IF 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol=40:60; flow rate: 1 ml/min; column temperature=30° C.) to obtain: Compound Z101-1 (8.7 mg, retention time: 7.10 min; peak 1), a white solid. ES-API: [M+H]+=576.2.


and Compound Z101-2 (15.2 mg, retention time: 9.68 min; peak 2), a white solid. ES-API: [M+H]+=576.2.


Example 102: Preparation of Compound Z102, Z102-1 and Z102-2



embedded image


Compound Z102 was prepared with 2-cyclopropyl-4-methylpyridin-3-amine as the raw material according to the method of Example 95. was purified by Preparative HPLC (ammonium bicarbonate system, column type: Chiralpak IC 250 mm*4.6 mm*5 um; mobile phase: acetonitrile:isopropanol=70:30; flow rate: 1 ml/min; column temperature=30° C.) to obtain: Compound Z102-1 (85 mg, retention time: 8.467 min; peak 1, yield: 27%). 1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J=4.9 Hz, 1H), 7.49 (td, J=7.3, 1.8 Hz, 1H), 7.31-7.19 (m, 3H), 7.09 (d, J=5.0 Hz, 1H), 6.93-6.79 (m, 1H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.96-4.83 (m, 2H), 4.66-4.54 (m, 1H), 4.50-4.26 (m, 2H), 4.07 (d, J=40.0 Hz, 1H), 3.79-3.62 (m, 2H), 3.31 (d, J=9.5 Hz, 1H), 1.96 (s, 3H), 1.75-1.68 (m, 1H), 0.89-0.66 (m, 4H).


and Compound Z102-2 (80 mg, retention time: 9.990 min; peak 2, yield: 25%). 1H-NMR (500 MHz, DMSO-d6) δ 8.21 (d, J=4.9 Hz, 1H), 7.50 (dd, J=13.6, 5.5 Hz, 1H), 7.31-7.19 (m, 3H), 7.08 (d, J=4.9 Hz, 1H), 6.93-6.79 (m, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.5, 2.1 Hz, 1H), 4.96-4.81 (m, 2H), 4.58 (d, J=13.5 Hz, 1H), 4.49-4.23 (m, 2H), 4.06 (d, J=29.1 Hz, 1H), 3.69 (dd, J=41.5, 29.9 Hz, 2H), 3.32 (s, 1H), 1.98 (s, 3H), 1.65 (s, 1H), 0.92 (dd, J=11.5, 5.9 Hz, 1H), 0.77-0.67 (m, 3H).


Example 103: Preparation of Compound Z103



embedded image


Compound Z103 was prepared with (2-fluoro-6-hydroxyphenyl)boronic acid as the raw material according to the method of Example 53. ES-API: [M+H]+=590.2. 1H-NMR (500 MHz, DMSO-d6) δ 10.04 (dd, J=12.5, 4.9 Hz, 1H), 8.68 (s, 1H), 7.16 (dd, J=15.4, 7.3 Hz, 1H), 6.80 (dd, J=27.5, 16.5 Hz, 1H), 6.69-6.53 (m, 2H), 6.14 (dd, J=16.7, 2.2 Hz, 1H), 5.71 (dd, J=10.5, 2.1 Hz, 1H), 4.96-4.73 (m, 2H), 4.59-4.26 (m, 2H), 4.21 (s, 1H), 3.97 (s, 1H), 3.57 (s, 2H), 2.05 (t, J=15.6 Hz, 3H), 1.68 (d, J=48.3 Hz, 1H), 0.95-0.69 (m, 4H).


Example 104: Preparation of Compound Z104, Z104-1 and Z104-2



embedded image


Compound Z104 was prepared with 3-isopropylpyrazine-2-amine as the raw material according to the method of Example 93. ES-API: [M+H]+=562.2. Compound Z104 (191 mg, 0.34 mmol) was resolved chirally (mobile phase: n-hexane-ethanol-40-60; column type: IC (250 mm*4.6 mm*5 um); flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z104-1 (86 mg, retention time: 8.554 min, purity: 99.2%, de value: 99%). ES-API: [M+H]+=562.2.


and Compound Z104-2 (81 mg, retention time: 15.317 min, purity: 100%, de value: 99%). ES-API: [M+H]+=562.2.


Example 105: Preparation of Compound Z105, Z105-1 and Z105-2



embedded image


Compound Z105 was prepared with (2-fluoro-6-hydroxyphenyl)boronic acid as the raw material according to the method of steps 6-8 in Example 104. ES-API: [M+H]+=579.2. Compound Z105 (182 mg, 0.31 mmol) was resolved chirally (mobile phase: n-hexane-ethanol-40-60); column type: IC (250 mm*4.6 mm*5 um; flow rate: 1.0 ml/min; T: 30° C.) to obtain: Compound Z105-1 (78 mg, retention time 9.559 min, purity: 99.5%, de value: 99%). ES-API: [M+H]+=579.2.


and Compound Z105-2 (81 mg, retention time 16.267 min, purity: 100%, de value: 99%). ES-API: [M+H]+=579.2.


Example 106: Preparation of Compound Z106



embedded image


Compound Z106 was prepared with (2,3-difluorophenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=592.2. 1H NMR (500 MHz, DMSO-d6).δ 8.77 (s, 1H), 7.56 (dd, J=17.7, 8.8 Hz, 1H), 7.31 (dd, J=12.8, 8.1 Hz, 1H), 7.07 (t, J=6.7 Hz, 1H), 6.86 (ddd, J=33.1, 16.8, 10.3 Hz, 1H), 6.22 (dd, J=16.7, 2.2 Hz, 1H), 5.78 (dd, J=10.4, 2.1 Hz, 1H), 5.01-4.81 (m, 2H), 4.67-4.37 (m, 2H), 4.31 (s, 1H), 4.05 (d, J=27.3 Hz, 1H), 3.89-3.62 (m, 2H), 2.15 (d, J=12.2 Hz, 3H), 1.87-1.71 (m, 1H), 1.08-0.76 (m, 4H).


Example 107: Preparation of Compound Z107, Z107-1 and Z107-2



embedded image


Compound Z107 was prepared according to the method of Example 99. ES-API: [M+H]+=562.1. Compound Z107 (207 mg, 0.37 mmol) was resolved chirally (mobile phase: n-hexane-ethanol-40-60; column type: IB (250 mm*4.6 mm*5 um; flow rate: 1.0 ml/min; column temperature: 30° C.) to obtain: Compound Z107-1 (98 mg, retention time 9.02 min, purity: 99.2%, de value: 99%). ES-API: [M+H]+=562.1.


and Compound Z107-2 (95 mg, retention time 14.76 min, purity: 100%, de value: 99%). ES-API: [M+H]+=562.1.


Example 108: Preparation of Compound Z108



embedded image


Compound Z108 was prepared according to the method of Example 99. ES-API: [M+H]+=574.2. 1H NMR (500 MHz, DMSO-d6) δ 10.21 (s, 1H), 8.77 (s, 1H), 7.27 (dd, J=15.4, 8.0 Hz, 1H), 6.84 (ddd, J=27.0, 17.0, 10.6 Hz, 1H), 6.77-6.65 (m, 2H), 6.21 (dd, J=16.7, 1.9 Hz, 1H), 5.80-5.73 (m, 1H), 4.85 (ddd, J=18.0, 15.2, 5.7 Hz, 2H), 4.57 (s, 1H), 4.51-4.24 (m, 2H), 4.02 (s, 1H), 3.88 (d, J=39.9 Hz, 1H), 3.70 (d, J=38.6 Hz, 1H), 3.49-3.35 (m, 1H), 2.12 (d, J=18.0 Hz, 3H), 1.83-1.67 (m, 1H), 1.02-0.87 (m, 3H), 0.86-0.76 (m, 1H).


Example 109: Preparation of Compound Z109, Z109-1 and Z109-2



embedded image


Compound Z109 was prepared with 5-amino-1-(tert-butyl)-1H-pyrazole-4-nitrile as the raw material according to the method of Example 93. ES-API: [M+H]+=589.1, resolved chirally (column type: Chiralpak IA 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol=60:40; flow rate: 1 ml/min; column temperature=30° C.) to obtain: Compound Z109-1 (17 mg, retention time 7.25 min, peak 1), a yellow solid, ES-API: [M+H]+=589.1;


and Compound Z109-2 (17 mg, retention time 9.22 min, peak 2), a yellow solid. ES-API: [M+H]+=589.2. 1HNMR (500 MHz, DMSO-d6): 7.87 (s, 1H), 7.26-7.25 (m, 1H), 7.20-7.23 (m, 2H), 7.14-7.10 (m, 1H), 6.62-6.59 (m, 1H), 6.48-6.45 (m, 1H), 5.89-5.87 (m, 1H), 4.94-4.71 (m, 4H), 4.15-4.12 (m, 1H), 4.02-3.44 (m, 4H), 1.31 (s, 9H).


Example 110: Preparation of Compound Z110



embedded image


Compound Z110 was prepared with 4-isopropyl-6-methoxypyrimidin-5-amine as the raw material according to the method of Example 93. ES-API: [M+H]+=592.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.72 (s, 1H), 7.57-7.46 (m, 1H), 7.37-7.23 (m, 2H), 7.18 (t, J=7.4 Hz, 1H), 6.84 (dd, J=30.7, 13.7 Hz, 1H), 6.21 (d, J=16.7 Hz, 1H), 5.78 (d, J=12.2 Hz, 1H), 5.01-4.80 (m, 2H), 4.68-4.22 (m, 3H), 4.06 (d, J=32.5 Hz, 1H), 3.68 (s, 5H), 3.38 (d, J=11.8 Hz, 1H), 2.83 (dd, J=27.6, 21.0 Hz, 1H), 1.13-0.88 (m, 7H).


Example 111: Preparation of Compound Z111, Z111-1 and Z111-2



embedded image


Compound Z111 was prepared according to the method of Example 99. ES-API: [M+H]+=592.2. 1H NMR (500 MHz, DMSO-d6) δ 10.18 (s, 1H), 8.71 (s, 1H), 7.26 (dd, J=15.3, 8.2 Hz, 1H), 6.95-6.77 (m, 1H), 6.75-6.62 (m, 2H), 6.21 (d, J=15.5 Hz, 1H), 5.84-5.69 (m, 1H), 4.84 (ddd, J=19.9, 18.1, 8.9 Hz, 2H), 4.68-4.22 (m, 3H), 4.02 (s, 1H), 3.94-3.62 (m, 5H), 2.79 (ddd, J=45.6, 13.2, 6.6 Hz, 1H), 1.11-0.95 (m, 6H). Compound Z111 (75 mg, 0.1268 mmol) was resolved chirally (column type: IF, 250 mm*4.6 mm*5 um, mobile phase: n-hexane:ethanol=50:50, flow rate: 1 ml/min, column temperature=30° C.) to obtain: Compound Z111-1 (25 mg, peak 1, retention time: 7.33 min, yield: 34%), a light yellow solid. and Compound Z111-2 (26 mg, peak 2, retention time: 1.42 min, yield: 34.6%), a light yellow solid. ES-API: [M+H]+=592.2.


Example 112: Preparation of Compound Z112, Z112-1 and Z112-2



embedded image


embedded image


embedded image


Compound Z112 was prepared with 2-cyclopropyl-4-methylpyridin-3-amine as the raw material according to the method of Example 92. was purified by Preparative HPLC (ammonium bicarbonate system, column type: Chiralpak IC 250 mm*4.6 mm*5 um; mobile phase: acetonitrile:isopropanol:ammonia methanol=85:15:0.2; flow rate: 1 ml/min; column temperature=30° C.) to obtain a yellow solid: Compound Z112-1 (60 mg, retention time: 9.201 min; peak 1, yield: 22%). 1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J=4.9 Hz, 1H), 7.23 (dd, J=15.2, 7.8 Hz, 1H), 7.08 (d, J=5.2 Hz, 1H), 6.91-6.79 (m, 1H), 6.73-6.59 (m, 2H), 6.21 (d, J=17.2 Hz, 1H), 5.78 (dd, J=10.4, 2.3 Hz, 1H), 4.91-4.80 (m, 2H), 4.55 (s, 1H), 4.47-4.34 (m, 1H), 4.28 (s, 1H), 4.03 (d, J=29.0 Hz, 1H), 3.74 (dd, J=55.0, 44.2 Hz, 4H), 1.93 (s, 3H), 1.70 (td, J=8.1, 4.1 Hz, 1H), 0.82 (d, J=4.0 Hz, 2H), 0.77-0.71 (m, 1H), 0.65 (d, J=7.9 Hz, 1H).


and Compound Z112-2 (12 mg, retention time: 12.307 min; peak 2, yield: 4.4%). 1H NMR (500 MHz, DMSO-d6) δ 10.15 (s, 1H), 8.21 (d, J=4.9 Hz, 1H), 7.25 (dd, J=15.3, 8.3 Hz, 1H), 7.08 (d, J=4.9 Hz, 1H), 6.85 (s, 1H), 6.73-6.65 (m, 2H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.77 (dd, J=10.4, 2.2 Hz, 1H), 4.90-4.79 (m, 2H), 4.59 (s, 1H), 4.49-4.27 (m, 2H), 4.02 (s, 1H), 3.79-3.58 (m, 2H), 3.30 (s, 1H), 1.96 (s, 3H), 1.61 (d, J=4.6 Hz, 1H), 0.84 (s, 1H), 0.75 (dd, J=15.9, 7.2 Hz, 2H), 0.61 (d, J=8.3 Hz, 1H).


Example 113: Preparation of Compound Z113



embedded image


embedded image


Step 1: 4,6-dichloro-2-fluoronicotinamide (10.5 g, 50 mmol), (S)-(1-ethylpyrrolidin-2-yl)(7.68 g, 60 mmol), potassium carbonate (20.7 g, 150 mmol) and 200 mL of dimethylformamide were added to a 500 mL three-necked round bottom flask, the reaction proceeded at 90° C. for 2 hours. 300 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 300 mL of the saturated brine for 3 times, dried and concentrated to obtain the target crude product: (S)-4,6-dichloro-2-(((1-ethylpyrrolidin-2-yl)methyl)amino)nicotinamide (12.3 g, yield: 77%). ES-API: [M+H]+=317.1.


Step 2: (S)-4,6-dichloro-2-(((1-ethylpyrrolidin-2-yl)methyl)amino)nicotinamide (5.5 g, 17.3 mmol) and 100 mL of anhydrous dimethylformamide were added to a 250 mL three-necked round bottom flask, NaH (7.0 g, 17.5 mmol) was added under the ice water bath, stirred for 30 minutes with heat preserved, N,N-carbonyldiimidazole (5.2 g, 32 mmol) was added to the solution, and after five minutes, tert-butyl (R)-3-(hydroxymethyl)piperazin-1-carboxylate (3.8 g, 17.6 mmol) was added under the ice water bath, reacted for 20 minutes with heat preserved. 300 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 300 mL of the saturated brine for 3 times, dried, the filtrate was concentrated and passed through the column to obtain the target product: tert-butyl (R)-3-(((7-chloro-1-(((S)-1-ethylpyrrolidin-2-yl)methyl)-2,4-dioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (4.5 g, yield: 49%). ES-API: [M+H]+=523.3.


Step 3: tert-butyl (R)-3-(((7-chloro-1-(((S)-1-ethylpyrrolidin-2-yl)methyl)-2,4-dioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidin-5-yl)oxy)methylpiperazin-1-formate (5.2 g, 9.9 mmol), DIEA (23 g, 178 mmol), T3P (23 g, 36 mmol) and 50 mL of dichloromethane were added to a 100 mL three-necked round bottom flask, and stirred at room temperature for 2 hours. 80 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 80 mL of the saturated brine for three times. The phase in ethyl acetate was dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (R)-2-chloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-11-oxo5α,6,8,9,11,12hexa hydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (750 mg, yield: 14.7%). ES-API: [M+H]+=505.3.


Step 4: tert-butyl (R)-2-chloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-11-oxo5α,6,8,9,11,12hexa hydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (400 mg, 0.79 mmol), NCS (300 mg, 2.25 mmol) were added to a 100 mL three-necked round bottom flask, and the reaction proceeded at 80° C. for 2 hours. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (R)-2,3-dichloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-11-oxo-5α,6,8,9,11,12 hexahydro 4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (400 mg, yield: 94%). ES-API: [M+H]+=539.2.


Step 5: tert-butyl (R)-2,3-dichloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-11-oxo-5α,6,8,9,11,12 hexahydro 4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (370 mg, 0.68 mmol), (2-fluorophenyl)boronic acid (380 mg, 2.7 mmol), tetratriphenylphosphine palladium (150 mg, 0.13 mmol), sodium carbonate (209 mg, 1.97 mmol), 10 mL of dioxane and 2 mL of water were added to a 100 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction proceeded at 120° C. for 1 hour. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (R)-3-chloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-2-(2-fluorophenyl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (199 mg, yield: 96%). ES-API: [M+H]+=599.2.


Step 6: tert-butyl (R)-3-chloro-12-(((S)-1-ethylpyrrolidin-2-yl)methyl)-2-(2-fluorophenyl)-11-ox o-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (199 mg, 0.33 mmol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction solution was concentrated to obtain the target intermediate. The intermediate was dissolved in dichloromethane (15 mL), and triethylamine (1 g, 10 mmol) was added. The reaction was cooled to 0° C., and acrylic anhydride (50 mg, 0.39 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 40 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 50 mL of the saturated NaHCO3 aqueous solution, 40 mL of the saturated brine, dried and concentrated. The crude product was purified by preparative liquid chromatography to obtain the target product: Z113 (52 mg, yield: 28%). ES-API: [M+H]+=553.2. 1H NMR (500 MHz, DMSO-d6) δ 7.65-7.50 (m, 2H), 7.38 (dd, J=12.9, 5.9 Hz, 2H), 6.93-6.76 (m, 1H), 6.28-6.16 (m, 1H), 5.76 (d, J=12.0 Hz, 1H), 4.88-4.73 (m, 2H), 4.53-3.98 (m, 6H), 3.55 (t, J=19.2 Hz, 2H), 3.20 (s, 1H), 2.99-2.71 (m, 3H), 2.22-2.06 (m, 2H), 1.75-1.52 (m, 4H), 0.84 (t, J=7.1 Hz, 3H).


Example 114: Preparation of Compound Z114



embedded image


embedded image


Compound Z114 was prepared with 3-chloro-5-isopropylpyridin-4-amine as the raw material according to the method of Example 93. 1H-NMR (500 MHz, DMSO-d6) δ 8.65 (d, J=4.2 Hz, 1H), 8.59 (d, J=3.4 Hz, 1H), 7.52-7.46 (m, 1H), 7.30-7.23 (m, 2H), 7.15 (t, J=7.3 Hz, 1H), 6.90-6.78 (m, 1H), 6.21 (d, J=16.5 Hz, 1H), 5.80-5.75 (m, 1H), 4.89 (dd, J=19.2, 13.8 Hz, 2H), 4.51 (d, J=32.0 Hz, 2H), 4.32 (d, J=28.9 Hz, 2H), 4.03 (s, 1H), 3.75 (dd, J=41.6, 27.4 Hz, 2H), 3.36 (s, 1H), 3.30 (s, 1H), 2.78-2.65 (m, 1H), 1.16 (d, J=6.8 Hz, 3H), 1.07-1.02 (m, 3H).


Example 115: Preparation of Compound Z115



embedded image


embedded image


embedded image


Compound Z115 was prepared with 4-isopropyl-2-methoxy-6-methylpyrimidin-5-amine as the raw material according to the method of Example 92. ES-API: [M+H]+=606.2. 1H NMR (500 MHz, DMSO-d6) δ 10.19 (d, J=4.2 Hz, 1H), 7.27 (dd, J=15.5, 8.0 Hz, 1H), 6.98-6.78 (m, 1H), 6.76-6.56 (m, 2H), 6.21 (d, J=16.6 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.98-4.74 (m, 2H), 4.57 (s, 1H), 4.46 (d, J=12.8 Hz, 1H), 4.33 (d, J=4.4 Hz, 2H), 4.04 (s, 1H), 3.75 (s, 2H), 2.86-2.57 (m, 1H), 2.04 (d, J=18.0 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 0.93 (dd, J=10.0, 6.8 Hz, 3H).


Example 116: Preparation of Compound Z116



embedded image


embedded image


Compound Z116 was prepared according to the method of steps 6-9 in Example 93. ES-API: [M+H]+=606.2. 1H NMR (500 MHz, DMSO-d6) δ 7.51 (dd, J=13.6, 5.7 Hz, 1H), 7.29 (dd, J=18.2, 9.0 Hz, 2H), 7.25-7.17 (m, 1H), 6.97-6.72 (m, 1H), 6.21 (dd, J=16.7, 2.3 Hz, 1H), 5.78 (dd, J=10.4, 2.2 Hz, 1H), 4.88 (ddd, J=19.6, 17.8, 8.6 Hz, 2H), 4.68-4.52 (m, 1H), 4.43 (dd, J=54.5, 13.2 Hz, 1H), 4.30 (s, 1H), 4.06 (d, J=27.0 Hz, 1H), 3.89 (s, 3H), 3.84-3.58 (m, 2H), 2.82-2.60 (m, 1H), 2.06 (d, J=13.0 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 0.93 (d, J=6.8 Hz, 3H).


Example 117: Preparation of Compound Z117



embedded image


Compound Z117 was prepared with (1-methyl-1H-pyrazole-4-yl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=561.2. 1H NMR (500 MHz, DMSO-d6) δ 8.44 (dd, J=69.0, 4.5 Hz, 2H), 7.34-7.19 (m, 2H), 6.95-6.76 (m, 1H), 6.21 (d, J=16.6 Hz, 1H), 5.77 (d, J=10.4 Hz, 1H), 4.83 (s, 2H), 4.70-4.01 (m, 4H), 3.83 (s, 3H), 3.64 (t, J=20.8 Hz, 2H), 3.32 (s, 1H), 2.82-2.63 (m, 1H), 1.93 (d, J=16.8 Hz, 3H), 1.00 (ddd, J=17.9, 8.9, 4.8 Hz, 6H).


Example 118: Preparation of Compound Z118



embedded image


Compound Z118 was prepared with (2-fluoro-5-hydroxyphenyl)boronic acid as the raw material according to the method of Example 46. ES-API: [M+H]+=591.2. 1H NMR (500 MHz, DMSO-d6) δ 9.62 (s, 1H), 8.37 (d, J=4.8 Hz, 1H), 7.20-7.01 (m, 2H), 6.79 (d, J=8.6 Hz, 2H), 6.48-6.39 (m, 1H), 6.21 (dd, J=16.7, 2.0 Hz, 1H), 5.78 (d, J=12.4 Hz, 1H), 4.86 (dd, J=20.3, 9.2 Hz, 2H), 4.28 (s, 3H), 4.07 (d, J=37.9 Hz, 1H), 3.66 (s, 2H), 3.34 (s, 1H), 2.79-2.65 (m, 1H), 1.93 (d, J=12.4 Hz, 3H), 1.00 (dd, J=59.9, 6.6 Hz, 6H).


Example 119: Preparation of Compound Z119



embedded image


Compound Z119 was prepared with 3-chloro-1-isopropyl-4-methyl-1H-pyrazole-5-amine and 4,6-dichloro-2-fluoronicotinamide as the raw material according to the method of Example 93.


Example 120: Preparation of Compound Z120



embedded image


Step 1: tert-butyl (R)-12-(1-(tert-butyl)-4-cyano-1H-pyrazole-5-yl)-2,3-dichloro-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (550 mg, 0.95 mmol), (2-fluoro-6-hydroxyphenyl)boronic acid (447 mg, 2.86 mmol), SPhos-Pd-G2 (102 mg, 0.14 mmol), sodium carbonate (304 mg, 2.87 mmol), 10 mL of dioxane and 2 mL of water were added to a 100 mL three-necked round bottom flask. The system was replaced with the nitrogen for three times, and then protected with a nitrogen ball. The reaction proceeded at 110° C. for 2 hours. 30 mL of ethyl acetate was added to the reaction solution, the reaction solution was washed with 30 mL of the saturated brine for 3 times, dried and concentrated. The crude product was purified by a fast silica gel column to obtain the target product: tert-butyl (5 aR)-12-(1-(tert-butyl)-4-cyano-1H-pyrazole-5-yl)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (350 mg, yield: 56%). ES-API: [M+H]+=651.2.


Step 2: tert-butyl (5 aR)-12-(1-(tert-butyl)-4-cyano-1H-pyrazole-5-yl)-3-chloro-2-(2-fluoro-6-hydroxyphenyl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (350 mg, 0.53 mmol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction solution was concentrated to obtain the target intermediate.


Step 3: the intermediate was dissolved in dichloromethane (15 mL), and triethylamine (1 g, 9.9 mmol) was added. The reaction was cooled to 0° C., and acrylic anhydride (14 mg, 0.11 mmol) was dropped to the reaction solution. The reaction was stirred at 0° C. for 15 minutes. 40 mL of dichloromethane was added to the reaction solution, the reaction solution was washed with 50 mL of the saturated NaHCO3 aqueous solution, 40 mL of the saturated brine, dried and concentrated. The crude product was purified by preparative liquid chromatography to obtain Compound Z120 (64 mg, yield: 44%). ES-API: [M+H]+=605.1. 1H NMR (400 MHz, DMSO-d6) δ 10.17 (s, 1H), 8.16-8.09 (m, 1H), 7.26 (d, J=7.2 Hz, 1H), 6.90-6.66 (m, 3H), 6.20 (dd, J=16.7, 2.2 Hz, 1H), 5.77 (dd, J=10.4, 2.2 Hz, 1H), 4.91 (s, 2H), 4.63-4.27 (m, 4H), 3.86 (d, J=102.5 Hz, 3H), 3.55-3.37 (m, 1H), 1.40 (dd, J=5.0, 3.2 Hz, 9H).


Example 121: Preparation of Compound Z121, Z121-1 and Z121-2



embedded image


embedded image


embedded image


Step 1: m-chloroperoxybenzoic acid (2.78 g, 16.10 mmol) was added to a solution of tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (5 g, 8.05 mmol) in dichloromethane (70 mL). The reaction solution was stirred at room temperature overnight. After the completion of the reaction, the reaction solution was diluted with dichloromethane (100 mL), washed with water (50 mL*3), the organic phase was dried and concentrated, and purified by a fast silica column (0-10% methanol/dichloromethane) to obtain a yellow solid: (R)-3-(7-(tert-butoxycarbonyl)-3-chloro-2-(2-fluorophenyl)-11-oxo-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-12 (11H)-yl)-2-isopropyl-4-methylpyridine1-oxide (2.5 g, yield=49%). ES-API: [M+H]+=637.2.


Step 2: A solution of Compound (R)-3-(7-(tert-butoxycarbonyl)-3-chloro-2-(2-fluorophenyl)-11-oxo-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-12 (11H)-yl)-2-isopropyl-4-methylpyridine1-oxide (2.5 g, 3.92 mmol) in acetic anhydride (25 mL) was stirred at 80° C. for 1 hour. When the raw material was completely converted to the intermediate state, it was concentrated in vacuum to remove acetic anhydride, re-dissolved in methanol (12 mL), and potassium carbonate was added (8 g). The mixture was stirred at room temperature for 30 minutes. After concentrating to remove methanol, 30 mL of water and 30 mL of dichloromethane were added, separated. The organic phase was dried and concentrated to obtain a mixture of tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methyl-6-oxo-1,6-dihydropyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate and tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (2.3 g). ES-API: [M+H]+=637 0.2.


Step 3: methyl iodide (5.12 g, 36.10 mmol) and potassium carbonate (1.50 g, 10.83 mmol) was added to a solution of the mixture of tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methyl-6-oxo-1,6-dihydropyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate and tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (2.3 g, 3.61 mmol) in acetonitrile (20 mL), the tube was sealed, and stirred at 65° C. for 6 hours. The reaction solution was concentrated, and purified by a fast silica column (0-5% methanol/dichloromethane) to obtain two compounds: tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-1,4-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (600 mg, yield=15%). ES-API: [M+H]+=651.2. Compound tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (750 mg, yield=31%). ES-API: [M+H]+=637 0.2.


Step 4: tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-1,4-dimethyl-6-oxo-1,6-dihyd ropyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (600 mg, 0.92 mmol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 1 hour. The reaction solution was concentrated to obtain the crude product: (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-1,4-dimethyl-6-oxo-1,6-dihyd ropyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (508 mg), directly used in the next step. ES-API: [M+H]+=551.2.


Step 5: (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-1,4-dimethyl-6-oxo-1,6-dihyd ropyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (508 mg, 0.92 mmol) was dissolved in dichloromethane (6 mL), and N,N-diisopropylethylamine (596 mg, 4.61 mmol) was added. The reaction solution was cooled to 0° C., and acrylic chloride (167 mg, 1.84 mmol) was dropped. The reaction solution was stirred at 0° C. for 10 minutes, concentrated, and the crude product was purified by Preparative HPLC to obtain Compound Z121 (160 mg, purity: 100%, yield: 29%), a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.22 (s, 1H), 7.53-7.44 (m, 1H), 7.31-7.22 (m, 2H), 7.18-7.11 (m, 1H), 6.94-6.75 (m, 1H), 6.21 (dd, J=16.5, 2.0 Hz, 1H), 5.77 (dd, J=10.5, 2.0 Hz, 1H), 4.94-4.83 (m, 2H), 4.70-4.53 (m, 1H), 4.50-4.33 (m, 1H), 4.28 (s, 1H), 4.13-4.01 (m, 1H), 3.88 (d, J=1.5 Hz, 3H), 3.77-3.60 (m, 2H), 3.29 (s, 1H), 2.70-2.58 (m, 1H), 1.78 (d, J=13.5 Hz, 3H), 1.04 (d, J=6.5 Hz, 3H), 0.95-0.90 (m, 3H). ES-API: [M+H]+=605 0.2.


Step 6: Compound Z121 (160 mg, 0.26 mmol) was resolved by a chiral chromatographic column (column type: IC 250 mm*4.6 mm*5 um; mobile phase: acetonitrile:isopropanol=70:30; flow rate:1 mL/min; column temperature 30° C.) to obtain: Compound Z121-1 (72 mg, peak 1, retention time 4.07 min, purity: 100%, de value: 100%). 1H NMR (500 MHz, DMSO-d6) δ 8.22 (s, 1H), 7.53-7.44 (m, 1H), 7.32-7.22 (m, 2H), 7.17-7.12 (m, 1H), 6.93-6.78 (m, 1H), 6.21 (dd, J=16.5, 2.0 Hz, 1H), 5.81-5.75 (m, 1H), 4.95-4.84 (m, 2H), 4.62-4.51 (m, 1H), 4.42 (dd, J=56.6, 11.7 Hz, 1H), 4.27 (s, 1H), 4.05 (d, J=23.5 Hz, 1H), 3.88 (s, 3H), 3.80-3.63 (m, 2H), 3.33 (s, 1H), 2.65-2.59 (m, 1H), 1.80 (s, 3H), 1.04 (d, J=7.0 Hz, 3H), 0.94 (d, J=6.5 Hz, 3H). ES-API: [M+H]+=605 0.2.


and Compound Z121-2 (46 mg, peak 2, retention time 5.51 min, purity: 96%, de value: 97.3%). 1H-NMR (500 MHz, DMSO-d6) δ8.23 (s, 1H), 7.53-7.45 (m, 1H), 7.32-7.22 (m, 2H), 7.19-7.13 (m, 1H), 6.95-6.78 (m, 1H), 6.21 (dd, J=17.0, 2.0 Hz, 1H), 5.78 (dd, J=10.5, 2 Hz, 1H), 4.94 (t, J=12.0 Hz, 1H), 4.88-4.81 (m, 1H), 4.67-4.53 (m, 1H), 4.52-4.34 (m, 1H), 4.28 (s, 1H), 4.18-4.00 (m, 1H), 3.89 (s, 3H), 3.77-3.59 (m, 2H), 3.31 (s, 1H), 2.75-2.66 (m, 1H), 1.78 (s, 3H), 1.06-1.02 (m, 3H), 0.94 (d, J=6.5 Hz, 3H). ES-API: [M+H]+=605.2.


Example 122: Preparation of Compound Z122, Z122-1 and Z122-2



text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


Step 1, Step 2 and Step 3 were the same as Step 1, Step 2 and Step 3 of Example 121;


Step 4: tert-butyl (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (750 mg, 1.18 mmol) was dissolved in dichloromethane (8 mL), and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 1 hour. The reaction solution was concentrated to obtain the crude product: (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (632 mg), directly used in the next step. ES-API: [M+H]+=537.2.


Step 5: (R)-3-chloro-2-(2-fluorophenyl)-12-(4-(hydroxymethyl)-2-isopropylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (632 mg, 1.18 mmol) was dissolved in dichloromethane (6 mL), and N,N-diisopropylethylamine (760 mg, 5.88 mmol) was added. The reaction solution was cooled to 0° C., and acrylic chloride (85 mg, 0.94 mmol) was dropped. The reaction solution was stirred at 0° C. for 10 minutes, concentrated, and the crude product was purified by Preparative HPLC to obtain Compound Z122 (470 mg, purity: 100%, yield: 67%), a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.49 (d, J=5.0 Hz, 1H), 7.53-7.43 (m, 1H), 7.39-7.34 (m, 1H), 7.30-7.20 (m, 2H), 7.17-7.11 (m, 1H), 6.96-6.76 (m, 1H), 6.21 (dd, J=16.5, 2.5 Hz, 1H), 5.78 (dd, J=10.5, 2.0 Hz, 1H), 5.37-5.29 (m, 1H), 4.97-4.80 (m, 2H), 4.63-4.50 (m, 1H), 4.49-4.34 (m, 1H), 4.32-4.21 (m, 2H), 4.18-3.99 (m, 2H), 3.82-3.63 (m, 2H), 3.29 (s, 1H), 2.81-2.62 (m, 1H), 1.06 (d, J=6.5 Hz, 3H), 0.95 (dd, J=6.5, 2.5 Hz, 3H). ES-API: [M+H]+=591.2.


Step 6: Compound Z122 (470 mg, 0.79 mmol) was resolved by a chiral chromatographic column (column type: IC 250 mm*4.6 mm*5 um; mobile phase: acetonitrile:isopropanol=70:30; flow rate: 1 mL/min; column temperature=30° C.) to obtain: Compound Z122-1 (177 mg, peak 1, retention time 4.34 min, purity: 100%, de value: 100%). 1H NMR (500 MHz, DMSO-d6) δ 8.50 (d, J=5.0 Hz, 1H), 7.51-7.44 (m, 1H), 7.38 (d, J=4.5 Hz, 1H), 7.33-7.20 (m, 2H), 7.18-7.11 (m, 1H), 6.94-6.78 (m, 1H), 6.21 (dd, J=16.5, 2.5 Hz, 1H), 5.78 (dd, J=10.5, 2.0 Hz, 1H), 5.35 (s, 1H), 4.93 (s, 1H), 4.84 (dd, J=13.0, 5.0 Hz, 1H), 4.68-4.52 (m, 1H), 4.51-4.33 (m, 1H), 4.33-4.22 (m, 2H), 4.16-3.99 (m, 2H), 3.81-3.59 (m, 2H), 3.33 (s, 1H), 2.84-2.71 (m, 1H), 1.07 (d, J=6.5 Hz, 3H), 0.95 (d, J=6.5 Hz, 3H). ES-API: [M+H]+=591.2.


and Compound Z122-2 (266 mg, peak 2, retention time 7.32 min, purity: 97.55%, de value: 100%). 1H NMR (500 MHz, DMSO-d6) δ 8.50 (d, J=5.0 Hz, 1H), 7.51-7.44 (m, 1H), 7.38 (d, J=4.0 Hz, 1H), 7.32-7.19 (m, 2H), 7.16-7.11 (m, 1H), 6.96-6.76 (m, 1H), 6.21 (dd, J=17.0, 2.5 Hz, 1H), 5.78 (dd, J=10.5, 2.5 Hz, 1H), 5.36 (s, 1H), 5.00-4.79 (m, 2H), 4.63-4.50 (m, 1H), 4.49-4.34 (m, 1H), 4.32-4.21 (m, 2H), 4.18-3.99 (m, 2H), 3.82-3.63 (m, 2H), 3.36 (s, 1H), 2.75-2.65 (m, 1H), 1.07 (d, J=6.7 Hz, 3H), 0.96 (d, J=6.7 Hz, 3H). ES-API: [M+H]+=591.2.


Example 123: Preparation of Compound Z123



embedded image


Compound Z123 was prepared with (2-(difluoromethyl)-6-fluorophenyl)boronic acid as the raw material according to the method of steps 3-5 in Example 72. ES-API: [M+H]+=607.2. 1H-NMR (500 MHz, DMSO-d6) δ 8.22 (d, J=4.8 Hz, 1H), 7.70 (dd, J=13.6, 8.0 Hz, 1H), 7.55 (dd, J=18.2, 8.6 Hz, 2H), 7.07 (s, 1H), 6.92-6.60 (m, 1H), 6.58 (d, J=47.9 Hz, 1H), 6.21 (d, J=17.0 Hz, 1H), 5.78 (dd, J=10.4, 2.1 Hz, 1H), 4.71-4.03 (m, 2H), 4.46 (s, 2H), 4.35 (s, 2H), 4.01 (s, 1H), 3.73 (s, 1H), 3.43 (s, 1H), 1.94 (s, 3H), 1.73 (s, 1H), 1.02-0.78 (m, 4H).


Example 124: Preparation of Compound Z124



embedded image


embedded image


embedded image


Step 1: tert-butyl (R)-2-chloro-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-de]-naphthalen-7 (5H)-carboxylate (1.26 g, 2.4 mmol), benzyl alcohol (2.6 g, 24.0 mmol), XantPhos-Pd-G2 (192 mg, 0.22 mmol), xantphos (127 mg, 0.22 mmol), cesium carbonate (1.56 g, 4.8 mmol) and dioxane (10 mL) were added to the microwave tube. After nitrogen bubbling for 5 minutes in the system, the reaction proceeded under a 80° C. microwave for 3.5 hours. After the completion of the reaction, filtered. The filtrate was concentrated, and the crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the target product: tert-butyl (R)-2-(benzyloxy)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (866 mg, 59%), a yellow solid. ES-API: [M+H]+=599.3.


Step 2: tert-butyl (R)-2-(benzyloxy)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (2.6 g, 4.3 mmol), N-iodosuccinimide (2.4 g, 10.8 mmol) and acetonitrile (30 mL) were added to a round bottom flask. The reaction was stirred at 80° C. for 16 hours. Sodium thiosulfate aqueous solution was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain tert-butyl (R)-2-(benzyloxy)-3-iodo-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (2.2 g), yield of 70%. ES-API: [M+H]+=725.2.


Step 3: tert-butyl (R)-2-(benzyloxy)-3-iodo-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-5a,6,8,9,11,12-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5] cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1.9 g, 2.62 mmol), methyl 2,2-difluoro-2-(fluorosulfonyl)acetate (1.5 g, 7.86 mmol), cuprous iodide (1.5 g, 7.86 mmol), N,N-dimethylformamide (15 mL) were added to the microwave tube. The system was heated in a sealed tube under a 90° C. oil bath for 2 hours. Water was added to the reaction solution. The reaction solution was extracted with ethyl acetate. The organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the product: tert-butyl (R)-2-(benzyloxy)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo-[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, 57%), a yellow solid. ES-API: [M+H]+=667.2.


Step 4: tert-butyl (R)-2-(benzyloxy)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo-[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (1 g, 1.5 mmol), Pd/C (1 g, 10% purity) and methanol (10 mL) were added to a round bottom flask. The reaction was stirred at room temperature for 16 hours. The completion of the reaction was detected by LC-MS. The reaction was filtered with diatomaceous earth, and the filtrate was concentrated to obtain the target product: tert-butyl (R)-2-hydroxy-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (800 mg, 93%). ES-API: [M+H]+=577 0.2.


Step 5: tert-butyl (R)-2-hydroxy-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (800 mg, 1.4 mmol), triethylamine (1.1 g, 11.2 mmol) and 10 mL of dichloromethane were added to a reaction flask. The reaction solution was cooled to 0° C., trifluoromethanesulfonic anhydride (1.56 g, 5.5 mmol) was dropped. The reaction was stirred at 0° C. for 10 minutes. The saturated sodium bicarbonate aqueous solution was added to the reaction solution, the reaction solution was extracted with dichloromethane for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-80%) to obtain the target product: tert-butyl (R)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-2-(((trifluoromethyl)sulfonyl)oxy)-5a,6,8,9,11,12hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (500 mg), a yellow solid, yield of 50%. ES-API: [M+H]+=709.2.


Step 6: tert-butyl (R)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-2-(((trifluoromethyl)sulfonyl)oxy)-5a,6,8,9,11,12hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-7 (5H)-carboxylate (500 mg, 0.70 mmol), 2-fluoro-6-hydroxyphenylboronic acid (330 mg, 2.11 mmol), tetratriphenylphosphine palladium (81 mg, 0.07 mmol), sodium phosphate (224 mg, 2.11 mmol), 10 mL of dioxane and 2 mL of water were added to a round bottom flask. The reaction was stirred under a 100° C. oil bath for 1 hour, and the reaction stopped. 50 mL of water was added to the reaction solution. The reaction solution was extracted with 50 mL of ethyl acetate for 3 times, and the organic phase was dried and concentrated. The crude product was purified by a fast silica gel column (ethyl acetate/petroleum ether: 0-100%) to obtain the target product: tert-butyl (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]-naphthalene7 (5H)-carboxylate (150 mg), a yellow solid, yield of 31%. ES-API: [M+H]+=671.2.


Step 7: tert-butyl (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-11-oxo-3-(trifluoromethyl)-5a,6,8,9,11,12hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]-naphthalene7 (5H)-carboxylate (150 mg, 0.22 mmol), 1 mL of methanol and 4 M hydrogen chloride/dioxane solution (3 mL) were added to a round bottom flask. The reaction was stirred at room temperature for 1 hour. The completion of the reaction was detected by LC-MS. The reaction solution was concentrated to obtain the crude product: (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-(trifluoromethyl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (125 mg), a yellow solid. ES-API: [M+H]+=571.2.


Step 8: (5 aR)-2-(2-fluoro-6-hydroxyphenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-3-(trifluoromethyl)-5,5a,6,7,8,9-hexahydro-4-oxo-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (125 mg, 0.22 mmol), 5 mL of dichloromethane and triethylamine (111 mg, 1.1 mmol) were added to a round bottom flask. The reaction was cooled to 0° C., and acrylic anhydride in dichloromethane (23 mg, 0.18 mmol, 0.5 mL) was dropped to the reaction solution. The reaction was stirred at 0° C. for 10 minutes. 20 mL of the saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was extracted with 20 mL of dichloromethane for 3 times. The organic phase was dried and concentrated. The crude product was purified by Preparative HPLC to obtain Z124 (46 mg), a white solid, yield of two steps is 33%. 1HNMR (500 MHz, CDCl3) 8.57-8.54 (m, 1H), 8.53-8.41 (m, 1H), 7.26-7.12 (m, 2H), 6.61-6.55 (m, 3H), 6.41-6.40 (m, 1H), 5.86-5.84 (m, 1H), 5.19-4.63 (m, 4H), 4.29-3.29 (m, 5H), 2.77-2.69 (m, 1H), 2.05-2.01 (m, 3H), 1.21-1.19 (m, 3H), 0.94-0.88 (m, 3H). ES-API: [M+H]+=625.1.


Example 125 to Example 236

Compound Z125-236 was prepared according to the synthesis methods of the above Examples.














Ex-
Compound



ample
Structure and
ES-API:


No.
No.
[M + H]+







125


embedded image


576.2






Z125






126


embedded image


605.2






Z126






127


embedded image


629.2






Z127






128


embedded image


585.2






Z128






129


embedded image


625.2






Z129






130


embedded image


578.2






Z130






131


embedded image


574.2






Z131






133


embedded image


586.2






Z133






134


embedded image


593.2






Z134






135


embedded image


607.2






Z135






136


embedded image


584.2





137


embedded image


589.2






Z137






138


embedded image


589.2






Z138






139


embedded image


618.2






Z139






140


embedded image


619.2






Z140






141


embedded image


618.2






Z141






142


embedded image


647.2






Z142






143


embedded image


576.2






Z143






144


embedded image


616.2






Z144






145


embedded image


631.2






Z145






146


embedded image


647.2






Z146






147


embedded image


590.2






Z147






148


embedded image


590.2






Z148






149


embedded image


575.2






Z149






150


embedded image


603.2






Z150






151


embedded image


572.2






Z151






152


embedded image


567.2






Z152






153


embedded image


550.2






Z153






154


embedded image


548.2






Z154






155


embedded image


605.2






Z155






156


embedded image


575.2






Z156






157


embedded image


578.2






Z157






158


embedded image


567.2






Z158






159


embedded image


550.2






Z159






160


embedded image


565.2






Z160






161


embedded image


572.2






Z161






162


embedded image


605.2






Z162






163


embedded image


605.2






Z163






164


embedded image


634.2






Z164






165


embedded image


635.2






Z165






166


embedded image


634.2






Z166






167


embedded image


663.2






Z167






168


embedded image


592.2






Z168






169


embedded image


632.2






Z169






170


embedded image


647.2






Z170






171


embedded image


663.2





172


embedded image


606.2





173


embedded image


606.2






Z173






174


embedded image


591.2






Z174






175


embedded image


619.2






Z175






176


embedded image


588.2






Z176






177


embedded image


583.1






Z177






178


embedded image


566.2






Z178






179


embedded image


564.2






Z179






180


embedded image


621.2






Z180






181


embedded image


591.2






Z181






182


embedded image


594.2






Z182






183


embedded image


583.2






Z183






184


embedded image


566.2






Z184






185


embedded image


581.2






Z185






186


embedded image


588.2






Z186






187


embedded image


570.2






Z187






188


embedded image


570.2






Z188






189


embedded image


554.2






Z189






190


embedded image


554.2






Z190






191


embedded image


570.2






Z191






192


embedded image


570.2






Z192






193


embedded image


554.2






Z193






194


embedded image


554.2






Z194






195


embedded image


556.2






Z195






196


embedded image


556.2






Z196






197


embedded image


540.2






Z197






198


embedded image


540.2






Z198






199


embedded image


589.2






Z199






200


embedded image


589.2






Z200






201


embedded image


618.2






Z201






202


embedded image


619.2






Z202






203


embedded image


618.2






Z203






204


embedded image


647.2






Z204






205


embedded image


576.2






Z205






206


embedded image


631.2






Z206






207


embedded image


647.2






Z207






208


embedded image


590.2






Z208






209


embedded image


590.2






Z209






210


embedded image


575.2






Z210






211


embedded image


603.2






Z211






212


embedded image


572.2






Z212






213


embedded image


567.2






Z213






214


embedded image


550.2






Z214






215


embedded image


548.2






Z215






216


embedded image


605.2






Z216






217


embedded image


575.2






Z217






218


embedded image


578.2






Z218






219


embedded image


567.2






Z219






220


embedded image


550.2






Z220






221


embedded image


565.2






Z221






222


embedded image


572.2






Z222






223


embedded image


554.2






Z223






224


embedded image


554.2






Z224






225


embedded image


554.2






Z225






226


embedded image


554.2






Z226






227


embedded image


540.2






Z227






228


embedded image


540.2






Z228






229


embedded image


484.1






Z229






230


embedded image


498.2






Z230






231


embedded image


605.2






Z231






232


embedded image


592.2






Z232






233


embedded image


591.2






Z233






234


embedded image


591.2






Z234






235


embedded image


591.2






Z235






236


embedded image


589.2






Z236









Example 237 Preparation of Compound Z237-1 and Z237-2



embedded image


embedded image


Step 1: N,N-diisopropylethylamine (182 mg, 1.44 mmol) was added to a solution of (R)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cycloheptan[1,2,3-dinaphthalene]11 (12H)-one (250 mg, 0.48 mmol) in dichloromethane (4 mL) under the ice bath, then (E)-4-bromobut-2-enoyl chloride (88 mg, 0.48 mmol) was added, and stirred for 10 minutes. The reaction solution was dissolved in 10 mL of water, extracted with 10 mL of dichloromethane, the organic phase was dried and concentrated, and purified by a fast silica column (0-10% methanol/dichloromethane) to obtain a white solid: (R,E)-7-(4-bromobut-2-enoyl)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (300 mg), yield 93%.


Step 2: 1 M dimethylamine in tetrahydrofuran solution (1.35 mL, 1.35 mmol) was dropped into a solution of (R,E)-7-(4-bromobut-2-enoyl)-3-chloro-2-(2-fluorophenyl)-12-(2-isopropyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (300 mg, 0.45 mmol) in tetrahydrofuran under the ice bath, and stirred for 1 hour. The reaction solution was concentrated, and purified by Preparative HPLC (ammonium bicarbonate) to obtain (R,E)-3-chloro-7-(4-(dimethylamino)but-2-enoyl)-2-(2-fluorophenyl)-12-(2-iso propyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (120 mg), yield 42%.


Step 3: the above-obtained Compound (R,E)-3-chloro-7-(4-(dimethylamino)but-2-enoyl)-2-(2-fluorophenyl)-12-(2-iso propyl-4-methylpyridin-3-yl)-5,5a,6,7,8,9-hexahydro-4-oxa-1,7,9a,10,12-pentazabenzo[4,5]cyclohepta[1,2,3-de]naphthalen-11 (12H)-one (120 mg, 0.19 mmol) was resolved chirally (column type: IG 250 mm*4.6 mm*5 um; mobile phase: n-hexane:ethanol (0.2% ammonia methanol)=40:60; flow rate: 1 mL/min; column temperature 30° C.) to obtain: Z237-1 (65 mg, peak 1, retention time 11.060 min, purity: 100%, de value: 100%). 1H NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=5 Hz, 1H), 7.52-7.44 (m, 1H), 7.30-7.21 (m, 2H), 7.17-7.11 (m, 2H), 6.75-6.58 (m, 2H), 4.95-4.83 (m, 2H), 4.64-4.51 (m, 1H), 4.51-4.32 (m, 1H), 4.32-4.23 (m, 1H), 4.13-3.96 (m, 1H), 3.84-3.58 (m, 2H), 3.41-3.36 (m, 1H), 3.07 (d, J=4.9 Hz, 2H), 2.75-2.63 (m, 1H), 2.18 (s, 6H), 1.95 (s, 3H), 1.05 (d, J=6.5 Hz, 3H), 0.94 (d, J=7.0 Hz, 3H). ES-API: [M+H]+=632.2; and Z237-2 (45 mg, peak 2, retention time 14.48 min, purity: 100%, de value: 99.68%). 1H NMR (500 MHz, DMSO-d6) δ 8.35 (d, J=5 Hz, 1H), 7.51-7.44 (m, 1H), 7.31-7.20 (m, 2H), 7.18-7.12 (m, 2H), 6.74-6.58 (m, 2H), 4.98-4.80 (m, 2H), 4.63-4.51 (m, 1H), 4.50-4.33 (m, 1H), 4.32-4.24 (m, 1H), 4.14-3.97 (m, 1H), 3.84-3.56 (m, 2H), 3.41-3.36 (m, 1H), 3.08 (d, J=4.6 Hz, 2H), 2.84-2.69 (m, 1H), 2.18 (s, 6H), 1.93 (s, 3H), 1.06 (d, J=6.7 Hz, 3H), 0.93 (d, J=6.7 Hz, 3H). ES-API: [M+H]+=632.2.


Test Example 1. Cell Proliferation Inhibition Experiment

NCI-H358 was a human non-small cell lung cancer cell line with Kras G12C mutation, cultured in 10% FBS RPMI-1640 medium; A549 was a human lung adenocarcinoma cell line with Kras G12S mutation, cultured in 10% FBS F-12K medium. The cells in the logarithmic growth phase was taken, digested with trypsin EDTA, collected and counted, and H358 was adjusted to 1.8E4 cells/ml using 2% FBS RPMI-1640 medium, and A549 was adjusted to 8.9E3 cells/ml using 2% FBS F-12K medium; 800 (45 μl) H358 or 400 (45 μl) A549 cells were seeded in a 384-well sphere plate respectively, cultured overnight to establish a 3D cell model. DMSO was used to prepare 1000× compound 3.16-fold gradient concentration stock solution, and 2% FBS medium was used to dilute 100-fold to 10× compound stock solution. On the second day after cell seeding, 5 μl 10× compound stock solution was added to each cell culture well, the final concentration was 1×, and DMSO content was 0.1%. DMSO was used as the experimental control (control) and 2% FBS medium as the blank control (blank). After the compound and cells were added and cultured for 5 days, 25 μl CellTiter-Glo working solution was added to each well, mixed at 400 rpm and incubated for 30 minutes. After 30 minutes at room temperature, 40 μl of the mixture was transferred to a 384-well plate with a non-transparent white bottom, the luminescence chemiluminescence value was read, and the cells proliferation inhibition rate IR (%)=(RLU control-RLU compound)/(RLU control-RLU blank)×100% was calculated, Prism 6 four-parameter method was used to fit the compound's gradient dilution concentration and the corresponding cell proliferation inhibition rate, and the IC50 value was calculate.


It can be seen from the results that the exemplary compounds of the present invention had high inhibitory activity against Kras G12C mutant NCI-H358 cells, with IC50 of less than 500 nM, or less than 100 nM, or less than 10 nM, while their inhibitory activity against A549 cells was low, with both IC50 more than 1000 nM, having obvious selective inhibitory activity.









TABLE 1







Inhibitory activity of compounds on


H358 and A549 cells











Compound
H358
A549



No.
IC50(μM)
IC50(μM)















Z1
0.002
7.448



Z15
0.036
>10



Z15-1
0.017
22.189



Z15-2
0.149
29.763



Z16
0.041
>10



Z17
0.063
22.931



Z17-2
0.037
12.833



Z18
0.107
12.414



Z19
0.068
46.373



Z19-1
0.321
6.986



Z19-2
0.025
12.027



Z20
0.005
3.760



Z20-1
0.010
12.213



Z20-2
0.046
5.952



Z20-3
0.0006
>10



Z20-4
0.026
2.670



Z21
0.283
>10



Z22
0.118
>10



Z23
0.002
>10



Z23-1
0.001
>10



Z23-2
0.021
>10



Z24
0.008
10.437



Z24-1
0.016
>10



Z24-2
0.003
>10



Z25
0.094
24.199



Z25-1
0.046
>10



Z25-2
0.281
>10



Z26-1
0.031
13.826



Z26-2
0.001
>10



Z26-2-2
0.002
>10



Z28
0.184
>10



Z29
0.007
12.572



Z29-1
0.003
>10



Z29-2
0.005
>10



Z29-3
0.165
>10



Z29-4
0.044
>10



Z30-1
0.065
25.009



Z30-2-2
0.003
>10



Z30-2
0.002
20.726



Z31
0.047
12.311



Z32
0.308
23.532



Z35
0.164
>10



Z36-2
0.068
14.393



Z37-2
0.276
29.490



Z41
0.318
>10



Z45-1
0.003
>10



Z45-4
0.065
>10



Z46
0.005
>10



Z46-1
0.089
8.365



Z46-2
0.003
5.476



Z47
0.003
>10



Z47-1
0.001
>10



Z47-2
0.042
>10



Z48
0.022
>10



Z49
0.012
>10



Z49-1
0.006
>10



Z49-2
0.113
>10



Z50
0.050
>10



Z52
0.004
>10



Z53
0.004
>10



Z54
0.066
>10



Z55
0.009
>10



Z56
0.011
>10



Z57
0.005
>10



Z57-1
0.111
>10



Z57-2
0.003
>10



Z58
0.001
>10



Z59
0.006
>10



Z60
0.007
>10



Z61
0.001
>10



Z62
0.006
>10



Z64
0.007
>10



Z65
0.009
>10



Z66
0.003
>10



Z67
0.003
>10



Z68
0.003
>10



Z69
0.095
>10



Z70
0.042
>10



Z72
0.005
>10



Z73
0.002
>10



Z73-1
0.009
>10



Z73-2
0.002
>10



Z74
0.016
>10



Z75
0.002
>10



Z76-1
0.040
>10



Z76-2
0.001
>10



Z77-1
0.026
>10



Z77-2
0.001
>10



Z78
0.254
>10



Z79
0.004
>10



Z80
0.003
>10



Z81
0.204
>10



Z82-1
0.023
3.565



Z82-2
0.002
8.298



Z83-1
0.001
12.081



Z83-2
0.015
9.771



Z84
0.013
>10



Z85
0.003
18.709



Z86
0.013
>10



Z87-1
0.047
10.222



Z87-2
0.001
5.555



Z88
0.004
>10



Z88-1
0.100
>10



Z88-2
0.003
>10



Z90
0.004
7.775



Z91
0.002
4.222



Z91-1
0.030
10.147



Z91-2
0.003
>30



Z92
0.003
>10



Z93
0.001
12.777



Z94
0.002
>10



Z95-1
0.052
11.283



Z95-2
0.001
>10



Z96-1
0.059
>10



Z96-2
0.002
>10



Z97
0.001
26.840



Z98
0.001
>10



Z99
0.002
>10



Z100-1
0.002
19.664



Z100-2
0.031
>10



Z101-1
0.042
>10



Z101-2
0.003
>10



Z102-1
0.004
13.252



Z102-2
0.027
12.181



Z103
0.007
>30



Z104-1
0.008
11.257



Z104-2
0.028
>10



Z105-1
0.038
>30



Z105-2
0.005
>30



Z106
0.004
>30



Z107-1
0.019
>30



Z107-2
0.007
>30



Z108
0.009
>30



Z109-1
0.005
8.479



Z109-2
0.106
9.775



Z110
0.004
14.689



Z111
0.004
>30



Z111-1
0.034
>30



Z111-2
0.003
>30



Z112-1
0.003
>30



Z112-2
0.094
>30



Z114
0.008
4.629



Z115
0.005
18.748



Z116
0.003
8.891



Z118
0.019
>30



Z119
0.012
7.903



Z120
0.017
27.662



Z121
0.003
>10



Z122
0.007
>30



Z237-2
0.047
>30










It can be seen from Table 1 that the exemplary compounds of the present invention had higher inhibitory activity against Kras G12C mutant NCI-H358 cells, but had lower inhibitory activity against A549 cells, and had obvious selective inhibitory activity.


Test Example 2 Cell p-ERK Detection Experiment

MIA PaCa2 was a human pancreatic cancer cell line with Kras G12C mutation, cultured in 10% FBS+2.5% Horse serum DMEM medium. The cells in the logarithmic growth phase were taken, digested with enzyme EDTA, collected, counted and 2.5E4 cells were seeded in a 96-well cell culture plate, cultured overnight. DMSO was used to prepare 1000× compound 3.16-fold gradient concentration stock solution, and medium was used to dilute 200-fold to 5× compound stock solution. On the second day after cell seeding, 5× compound stock solution was added to each cell culture well, the final concentration was 1×, and DMSO content was 0.1%. DMSO was used as an experimental control (control). After the compound was added and cultured for two hours, the remaining medium was removed. 50 ul cell lysis buffer was added to each well, mixed and incubated for 30 minutes, then 16 ul of the mixture was transferred to a 96-well plate with a non-transparent white bottom, and 16 ul cell lysis buffer was added to blank wells. After the completion of the transfer, 4 ul p-ERK HTRF antibody mixture was added to each well, incubated for 4 hours and the fluorescence value was read. The compound inhibition rate IR (%)=(RLU control-RLU compound)/(RLU control-RLU blank)×100% was calculated, Prism 8 four-parameter method was used to fit the compound's gradient dilution concentration and the corresponding cell proliferation inhibition rate, and the IC50 value was calculate. It can be seen from the results that the exemplary compounds of the present invention had a good inhibitory activity on the level of phosphorylated ERK downstream of the cell pathway with Kras G12C protein mutation, with IC50 lower than 10 μM; or lower than 1000 nM, or lower than 500 nM, or lower than 100 nM. The results of the example compounds were shown in Table 2 below.









TABLE 2







Inhibitory activity of


compounds on p-ERK










Compound
p-ERK



No.
IC50(μM)














Z23-1
0.013



Z24-1
0.053



Z24-2
0.025



Z46-2
0.020



Z47-1
0.015



Z47-2
0.314



Z49
0.128



Z49-1
0.064



Z53
0.048



Z57-2
0.027



Z58
0.014



Z60
0.047



Z61
0.050



Z65
0.182



Z66
0.039



Z67
0.021



Z72
0.063



Z73
0.022



Z73-1
0.116



Z73-2
0.020



Z74
0.128



Z75
0.112



Z76-1
0.109



Z76-2
0.008



Z77-1
0.058



Z77-2
0.008



Z79
0.045



Z80
0.019



Z82-1
0.407



Z82-2
0.023



Z83-1
0.009



Z83-2
0.067



Z84
0.105



Z85
0.054



Z86
0.101



Z87-1
0.158



Z87-2
0.009



Z88
0.086



Z88-1
0.356



Z88-2
0.027



Z90
0.019



Z91
0.016



Z91-1
0.093



Z91-2
0.018



Z92
0.025



Z93
0.009



Z94
0.007



Z95-1
0.248



Z95-2
0.005



Z96-1
0.195



Z96-2
0.007



Z97
0.007



Z99
0.012



Z104-1
0.014



Z104-2
0.185



Z105-1
0.395



Z105-2
0.020



Z107-1
0.226



Z107-2
0.035



Z108
0.049



Z109-1
0.018



Z110
0.043



Z111
0.047



Z111-1
0.186



Z111-2
0.034



Z112-1
0.024



Z112-2
0.502



Z114
0.039



Z115
0.031



Z116
0.026



Z118
0.166



Z119
0.059



Z120
0.193



Z121
0.016



Z122
0.101










Test Example 3 Cell Proliferation Inhibition Experiment

MIA PaCa2 was a human pancreatic cancer cell line with Kras G12C mutation, cultured in 10% FBS+2.5% Horse serum DMEM medium; A549 was a human lung adenocarcinoma cell line with Kras G12S mutation, cultured in 10% FBS F-12K medium. The cells in the logarithmic growth phase were taken, digested with enzyme EDTA, collected, counted and 200 MIA PaCa-2 or 400 A549 cells were seeded in a 384-well sphere plate respectively, cultured overnight to establish a 3D cell model. DMSO was used to prepare 1000× compound 3.16-fold gradient concentration stock solution, and medium was used to dilute 100-fold to 10× compound stock solution. On the second day after cell seeding, 10× compound stock solution was added to each cell culture well, the final concentration was 1×, and DMSO content was 0.1%. DMSO was used as an experimental control (control), medium as the blank control (blank). After the compound and cells were added and cultured for 5 days, 30 μl CellTiter-Glo working solution was added to each well, mixed and incubated for 30 minutes. After 30 minutes at room temperature, 40 μl of the mixture was transferred to a 384-well plate with a non-transparent white bottom, the luminescence chemiluminescence value was read, and the cells proliferation inhibition rate IR (%)=(RLU control-RLU compound)/(RLU control-RLU blank)×100% was calculated, XLFit four-parameter method was used to fit the compound's gradient dilution concentration and the corresponding cell proliferation inhibition rate, and the IC50 value was calculate. It can be seen from the results that the exemplary compounds of the present invention had high inhibitory activity against Kras G12C mutant MIA PaCa-2 cells, with IC50 of less than 1000 nM, or less than 500 nM, or less than 100 nM, or less than 10 nM. The results of the example compounds were shown in Table 3 below.









TABLE 3







Inhibitory activity of


compounds on MIA-PaCa2










Compound
MIA-PaCa2



No.
IC50(μM)














Z15-1
0.020



Z15-2
0.261



Z16
0.029



Z17
0.055



Z17-2
0.033



Z19-2
0.024



Z20
0.015



Z20-1
0.017



Z20-2
0.173



Z20-3
0.0005



Z20-4
0.051



Z23-1
0.003



Z24
0.009



Z24-2
0.008



Z26-1
0.074



Z26-2
0.004



Z26-2-2
0.003



Z29
0.014



Z29-1
0.008



Z29-2
0.006



Z29-4
0.358



Z30-1
0.078



Z30-2-2
0.004



Z30-2
0.005



Z31
0.025



Z36-2
0.121



Z39
0.444



Z41
0.344



Z46
0.020



Z46-2
0.008



Z47
0.009



Z47-1
0.003



Z49
0.022



Z49-1
0.010



Z52
0.023



Z53
0.010



Z55
0.026



Z57
0.022



Z58
0.003



Z59
0.026



Z60
0.019



Z61
0.008



Z64
0.009



Z65
0.034



Z66
0.007



Z67
0.006



Z68
0.009



Z73
0.003



Z73-2
0.005



Z76-2
0.002



Z77-2
0.002



Z79
0.006



Z84
0.022



Z85
0.006



Z86
0.024



Z87-2
0.002



Z91-2
0.015



Z92
0.002



Z93
0.002



Z94
0.002



Z95-2
0.001



Z96-2
0.002



Z97
0.001



Z98
0.002



Z99
0.003



Z100-1
0.004



Z101-1
0.056



Z101-2
0.003



Z102-1
0.004



Z102-2
0.083



Z103
0.018



Z104-1
0.012



Z105-2
0.011



Z106
0.007



Z107-2
0.019



Z108
0.017



Z109-1
0.012



Z110
0.011



Z111
0.007



Z111-2
0.013



Z112-1
0.004



Z112-2
0.126



Z114
0.074



Z115
0.021



Z116
0.026



Z119
0.113



Z120
0.085



Z121
0.005



Z122
0.011



Z237-2
0.048










All documents mentioned in the present invention are cited as references in this application, as if each document is individually cited as a reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art may make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of the present application.

Claims
  • 1. A compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, the compound has a structure as represented by formula (I),
  • 2. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, R1, R2 are each independently hydrogen, cyano, C1-3 alkyl, —CH2NH2, —CH2NHCH3, or —CH2N(CH3)2.
  • 3. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, R01, R02, R03, R04, R05, R06 are each independently hydrogen, C1-3 alkyl, —C1-2 alkyl-hydroxy, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, —C1-2 alkyl-halo C1-3 alkoxy; or R01, R02 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;or R03, R04 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl;or R05, R06 together with the carbon atom attached thereto form C3-6 monocyclic cycloalkyl.
  • 4. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, R02, R04 are each independently hydrogen, CH3, —CH2-hydroxy, or —CH2-cyano; R01, R03, R05, R06 are hydrogen.
  • 5. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, L is a bond, or (CRL1RL2)n; wherein RL1, RL2 are each independently hydrogen, halo, or C1-6 alkyl; n is 1 or 2.
  • 6. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, X1 is NRx1 or O; wherein Rx1 is hydrogen, or C1-6 alkyl.
  • 7. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, X2 is N or CRx4; wherein Rx4 is hydrogen, halo, C1-6 alkyl, C1-6 alkoxy, or halo C1-6 alkyl.
  • 8. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, Ra is hydrogen, halo, cyano, C1-4 alkyl, C1-4 alkoxy, halo C1-3 alkyl, halo C1-3 alkoxy, C3-6 monocyclic cycloalkyl, NRcRd, C2-4 alkenyl, C2-4 alkynyl, —C1-2 alkyl-hydroxy, —C1-2 alkyl-cyano, —C1-2 alkyl-C1-3 alkoxy, —C1-2 alkyl-halo C1-3 alkyl, or —C1-2 alkyl-halo C1-3 alkoxy; wherein Rc, Rd are each independently hydrogen, or C1-3 alkyl.
  • 9. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the 7- to 11-membered spirocycloalkyl in Rc is a monospirocycloalkyl containing one spiro atom formed by any two monocyclic cycloalkyl rings selected from cyclopropyl ring, cyclobutyl ring, cyclopentyl ring, and cyclohexyl ring.
  • 10. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the C6-10 aryl in Rb, Re are each independently phenyl, naphthyl, a 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl, or a 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic cycloalkyl.
  • 11. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 10, wherein, the C5-6 monocyclic heterocyclyl in the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl is selected from the group consisting of: oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one.
  • 12. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 10, wherein, the C5-6 monocyclic cycloalkyl in the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.
  • 13. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the C5-10 heteroaryl in Rb, Rc are each independently a 5- or 6-membered monoheteroaryl, a 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl, a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl, a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic heterocyclyl, or a 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic cycloalkyl.
  • 14. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, when the C5-10 heteroaryl in Rb, Rc are 5- or 6-membered monoheteroaryl, the 5- or 6-membered monoheteroaryl are each independently selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.
  • 15. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein the 5- or 6-membered monoheteroaryl in the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.
  • 16. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine.
  • 17. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic heterocyclyl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine; the C5-6 monocyclic heterocyclyl is selected from the group consisting of: oxazolidine, pyrrolidin-2-one, pyrrolidin-2,5-dione, 1,3-dioxolane, dihydrofuran-2 (3H)-one, dihydrofuran-2,5-dione, piperidin-2-one, piperidin-2,6-dione, tetrahydro-2H-pyran-2-one, imidazolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyrrole, 1,3-dioxolan-2-one, oxazolidin-2-one, imidazolidine-2-one, piperidine, piperazine, piperazin-2-one, morpholine, morpholin-3-one, morpholin-2-one, thiomorpholin-3-one 1,1-dioxide, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydropyran, 2,5-dihydro-1H-pyrrole, 2,5-dihydrofuran, 2,3-dihydrofuran, 2,3-dihydro-1H-pyrrole, 3,4-dihydro-2H-pyran, 1,2,3,4-tetrahydropyridine, 3,6-dihydro-2H-pyran, 1,2,3,6-tetrahydropyridine, 1,3-oxazinane, hexahydropyrimidine, 1,4-dioxane, tetrahydropyrimidin-2 (1H)-one, 1,4-dioxan-2-one, 5,6-dihydro-2H-pyran-2-one, 5,6-dihydropyrimidin-4 (3H)-one, 3,4-dihydropyridin-2 (1H)-one, 5,6-dihydropyridin-2 (1H)-one.
  • 18. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 5- or 6-membered monoheteroaryl in the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to one C5-6 monocyclic cycloalkyl is selected from the group consisting of: thiophene, N-alkylcyclopyrrole, furan, thiazole, isothiazole, imidazole, oxazole, pyrrole, pyrazole, triazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,5-triazole, 1,3,4-triazole, tetrazole, isoxazole, oxadiazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, 1,3,4-oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, or pyrazine; the C5-6 monocyclic cycloalkyl is selected from the group consisting of: cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexdienyl, cyclopentanone, cyclopentan-1,3-dione, cyclohexanone, cyclohexan-1,3-dione.
  • 19. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 10, wherein, the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl has a structure selected from the group consisting of:
  • 20. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:
  • 21. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:
  • 22. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:
  • 23. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:
  • 24. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 13, wherein, the 9- or 10-membered biheteroaryl formed by fusing a phenyl to a 5- or 6-membered monoheteroaryl or the 8- to 10-membered biheteroaryl formed by fusing a 5- or 6-membered monoheteroaryl to a 5- or 6-membered monoheteroaryl has a structure selected from the group consisting of:
  • 25. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 10, wherein, the 9- or 10-membered aromatic fused bicyclic ring formed by fusing a phenyl to one C5-6 monocyclic heterocyclyl has a structure selected from the group consisting of:
  • 26. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the Rb has a structure selected from the group consisting of:
  • 27. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the R, has a structure selected from the group consisting of:
  • 28. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the compound of formula (I) is selected from each specific compound noted in the Examples.
  • 29. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, wherein, the compound of formula (I) has a structure as shown in formula (IA) or formula (IB):
  • 30. A pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1; and a pharmaceutically acceptable carrier.
  • 31. A method for preventing and/or treating cancer in a subject, comprising administering an effective amount of the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, or the pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, and a pharmaceutically acceptable carrier to the subject.
  • 32. A method for inhibiting of KRAS mutation in a subject, comprising administering an effective amount of the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1, or the pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 1 and a pharmaceutically acceptable carrier to the subject.
  • 33. An oxaazaquinazolin-7 (8H)-one compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, the compound has a structure as represented by formula (II):
  • 34. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33, wherein, the compound of formula (II) has a structure as shown in formula (IIA) or formula (IIB):
  • 35. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33, wherein, the compound of formula (II) is selected from the Table A-1.
  • 36. The compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33, wherein, the compound of formula (II) is selected from the Table A-2.
  • 37. A pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33 and a pharmaceutically acceptable carrier.
  • 38. A method for preventing and/or treating cancer in a subject, comprising administering an effective amount of the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33, or the pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33 and a pharmaceutically acceptable carrier to the subject.
  • 39. A method for inhibiting of KRAS mutation in a subject, comprising administering an effective amount of the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33, or the pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof according to claim 33 and a pharmaceutically acceptable carrier to the subject.
Priority Claims (1)
Number Date Country Kind
2019 10349813.4 Apr 2019 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2020/087428 4/28/2020 WO