CN116143806A - Nitrogen-containing heterocyclic compound, preparation method and application - Google Patents

Nitrogen-containing heterocyclic compound, preparation method and application Download PDF

Info

Publication number
CN116143806A
CN116143806A CN202210940525.8A CN202210940525A CN116143806A CN 116143806 A CN116143806 A CN 116143806A CN 202210940525 A CN202210940525 A CN 202210940525A CN 116143806 A CN116143806 A CN 116143806A
Authority
CN
China
Prior art keywords
alkyl
membered
group
compound
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210940525.8A
Other languages
Chinese (zh)
Inventor
万惠新
王亚周
马金贵
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Lingda Biomedical Co ltd
Original Assignee
Shanghai Lingda Biomedical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Lingda Biomedical Co ltd filed Critical Shanghai Lingda Biomedical Co ltd
Publication of CN116143806A publication Critical patent/CN116143806A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The invention discloses a nitrogen-containing heterocyclic compound, a preparation method and application thereof, in particular to a nitrogen-containing heterocyclic compound shown in a general formula (I), or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, a preparation method thereof and application thereof in pharmacy, wherein the definition of each group is as described in the specification.

Description

Nitrogen-containing heterocyclic compound, preparation method and application
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and in particular relates to a nitrogen-containing heterocyclic compound, a compound for inhibiting activity of Ras mutein, a preparation method and application thereof.
Background
Ras is the first oncogene identified in human tumors, and was first discovered in two murine sarcoma viruses. There are three members of the Ras gene family, HRas, KRas, NRas respectively. In humansKRas mutations are most common in tumors, accounting for about 85%. Previous studies have shown that KRas mutations are oncogenic because of missense mutations at codon 12, altering the structure of the KRas protein and allowing it to remain active. Ras plays a major role in signaling pathway in activating kinases that control gene transcription, thereby regulating cell differentiation and proliferation, and is intimately involved in tumor cell survival, proliferation, migration, metastasis, and angiogenesis. It is counted that a high proportion of KRas mutations occur in malignant tumors such as pancreatic cancer, colorectal cancer, ovarian cancer, cholangiocarcinoma and the like. However, targeting drugs for common proto-oncogenes such as EGFR, BCL, etc. have been developed for several generations since the first discovery of KRas oncogenes, but targeting drugs for KRas have not been successfully developed all the time. Targeting drugs against KRas pathway mutant tumors have been mainly focused on farnesyl transferase inhibitors and Raf-MEK pathway inhibitors, but have had little effect. In recent years, inhibitors against KRAS-specific gene mutations have been developed as hot spots, although some inhibitors have gradually moved from preclinical hatching to clinical studies, such as KRAS G12C AMG510, MRTX1257, etc., and shows a certain efficacy in early clinical trials. Global first money KRAS G12C The first clinical data of the inhibitor AMG510 was formally published by the american clinical oncology institute held at month 6 of 2019, in which the drug AMG510 was installed to be shown to prevent tumor growth in most patients with non-small cell lung and colorectal cancer with KRas mutations.
However, the current KRas inhibitors are also limited to only KRas G12C Patients with mutations and a large number of KRAS G12C Other than mutation inhibitor drug development has not been broken through, such as highly-developed KRas G12D 、KRas G12V 、KRas G13D And the like. Thus, highly specific and excellent drug-specific KRAS-directed drugs were found and sought G12C Targeted drugs of specific mutant genes other than those have become a great hotspot in the industry. Through the long-term efforts of the inventor, a class of KRAS mutation inhibitors with novel structure and action mechanism, especially KRAS, is discovered G12D The mutation has better activity and drug-forming property.
Disclosure of Invention
The invention aims to solve the technical problems of KRAS in the prior art G12D The problem of lack of inhibitors provides a class of nitrogen-containing heterocyclic compounds, a preparation method and application. The nitrogen-containing heterocyclic compounds provided by the invention are brand new KRAS G12D Inhibitors, which exhibit good inhibitory activity; the preparation has good inhibitory activity on tumor cells and good drug-forming property, and has wide drug development prospect.
The invention solves the technical problems by the following technical proposal:
the invention provides a nitrogen-containing heterocyclic compound shown in a general formula (I), or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
Figure BDA0003785353610000021
wherein, the liquid crystal display device comprises a liquid crystal display device,
R 1 selected from substituted or unsubstituted piperazine, substituted or unsubstituted piperidine, or substituted or unsubstituted 5-10 membered heterocycloalkyl, said substituents being selected from one or more of the following groups: halogen, cyano, hydroxy, amino, - (CO) -NR 11 NR 12 、C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkylcyano, C 1 -C 6 Alkyl hydroxy, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, or a 3-10 membered carbocyclic or heterocyclic ring system between each of the foregoing substituents, said ring system comprising spiro, bridged, fused or fused rings; r is R 11 、R 12 Independently selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or R 11 、R 12 Together with the attached N atom, form a 3-8 membered heterocycloalkyl;
R 2 selected from substituted or unsubstituted C 1 -C 6 Alkyl, 3-12 membered cycloalkyl,3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl; the substituent is selected from one or more of the following groups: halogen, cyano, hydroxy, amino, monoalkylamino, dialkylamino, monoalkylaminoalkyl, dialkylaminoalkyl, C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl, 6-12 membered saturated or partially unsaturated spiro, bridged, fused, ring containing 0 to 3 heteroatoms, and the like;
R 4 a substituent selected from one or more of the following groups: hydrogen, deuterium, halogen, hydroxy, amino, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, etc., or between the two substituents forming a 3-to 10-membered saturated or partially unsaturated or aromatic ring system through a carbon atom or heteroatom;
l is selected from chemical bond, CHR 5 、O、S、NR 5 ;R 5 Selected from H, C 1 -C 6 Alkyl of (a);
L 1 selected from direct bond, CHR 5 、O、S、NR 5 ;R 5 Selected from H, C 1 -C 6 Alkyl of (a);
ar is selected from benzene ring, naphthalene ring or benzothiazole ring, and the Ar can be one or more different R 3 Substitution, said R 3 Selected from halogen, hydroxy, amino, cyano, C 1 -C 6 Alkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 1 -C 6 Alkoxy groups, and the like;
m is N or CR 6 ;R 6 Selected from hydrogen, halogen, CN, C 1 -C 3 Alkyl groups, etc.;
cy is selected from the following group of ring systems:
Figure BDA0003785353610000031
the hetero atoms in the heterocyclic ring system, the heterocycloalkyl group and the heteroaryl group are independently selected from N, O, S, P, and the number of the hetero atoms is 1-3.
In one embodiment of the present invention, the nitrogen-containing heterocyclic compound represented by the formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, is characterized in that,
m is N, C-H, C-F, C-CN;
cy is preferably self-contained
Figure BDA0003785353610000032
R 1 Selected from:
Figure BDA0003785353610000033
/>
Figure BDA0003785353610000034
r is as described above 1 Hydrogen on any C of the radicals being halogen, hydroxy, C 1 -C 6 Alkyl, or = O substitution;
ar is selected from benzene ring, naphthalene ring or benzothiazole ring, and the Ar can be one or more different R 3 Substitution, said R 3 Selected from halogen, hydroxy, amino, cyano, C 1 -C 6 Alkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 1 -C 6 Alkoxy groups, and the like; r3 is preferably F, hydroxy, methyl, ethyl, ethynyl, amino;
l is selected from: CH (CH) 2 O, NH or S;
L 1 selected from: direct bond, CH 2 O, NH or S;
R 2 selected from substituted or unsubstituted C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted C 1 -C 3 An alkyl group; the substituent is selected from the following groups: halogen, cyanoRadical, hydroxy, amino, mono C 1 -C 6 Alkylamino, bis C 1 -C 6 Alkylamino, mono C 1 -C 6 alkylamino-C 1 -C 6 Alkyl, bis C 1 -C 6 alkylamino-C 1 -C 6 Alkyl, C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted C 1 -C 3 Alkyl, 6-12 membered saturated or partially unsaturated spiro ring containing 0 to 3 hetero atoms, bridged ring, parallel ring, condensed ring, etc.; the substituents are more preferably F, methyl,
Figure BDA0003785353610000035
In one embodiment of the present invention, the nitrogen-containing heterocyclic compound represented by the formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, is characterized in that,
R 1 preferably selected from
Figure BDA0003785353610000041
Cy is preferably self-contained
Figure BDA0003785353610000042
R 3 Ar is preferably selected from
Figure BDA0003785353610000043
L is preferably selected from-O-; l (L) 1 Preferably a direct bond, CH2;
R 2 selected from the group consisting of
Figure BDA0003785353610000044
Wherein m and n are each selected from integers from 1 to 3; ry is selected from alkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl, 5-10 membered aryl, heteroaryl A base; r is R p And R is q Selected from hydrogen, halogen, C 1 -C 6 Alkyl or alkoxy, hydroxy, amino, or R p And R is q Forming a 3-to 10-membered carbocyclic or heterocyclic ring system;
R 4 preferably selected from hydrogen, deuterium, halogen, methyl, trifluoromethyl, cyano, and the like.
In some embodiments of the present invention, the nitrogen-containing heterocyclic compound represented by formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, stereoisomer, solvate, polymorph or prodrug thereof, wherein,
m is preferably N, C-H, C-F, C-CN;
alternatively, cy is selected from
Figure BDA0003785353610000045
Alternatively, R 11 、R 12 Independently selected from hydrogen, halogen, methyl;
alternatively, when R 11 、R 12 When taken together with the attached N atom form a 5-8 membered heterocycloalkyl, the 3-8 membered heterocycloalkyl is preferably
Figure BDA0003785353610000046
Alternatively, R 1 Selected from substituted or unsubstituted piperazine, substituted or unsubstituted piperidine, or substituted or unsubstituted 5-10 membered heterocycloalkyl, said substituents being selected from one or more of the following groups: halogen, hydroxy, C 1 -C 6 Alkyl group,
Figure BDA0003785353610000051
Figure BDA0003785353610000052
Or substituents form a 3-to 10-membered carbocyclic or heterocyclic ring system between them, said ring system comprising spiro, bridged, fused or fused rings; r is R 1 Preferably selected from: />
Figure BDA0003785353610000053
Figure BDA0003785353610000054
R is as described above 1 The hydrogen on any C of the radicals being halogen, hydroxy, C 1 -C 6 Alkyl, or = O substitution;
alternatively, R 3 Ar is selected from
Figure BDA0003785353610000055
Alternatively, L is selected from-O-;
alternatively, L 1 Selected from direct bond, CH 2
Alternatively, R 2 Selected from the group consisting of
Figure BDA0003785353610000056
Wherein m and n are each selected from integers from 1 to 3; ry is selected from alkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl, 5-10 membered aryl, heteroaryl; r is R p And R is q Selected from hydrogen, halogen, C 1 -C 6 Alkyl or alkoxy, hydroxy, amino, or R p And R is q Forming a 3-to 10-membered carbocyclic or heterocyclic ring system; />
Figure BDA0003785353610000057
Preferably +.>
Figure BDA0003785353610000058
Alternatively, R 4 Selected from hydrogen, deuterium, halogen, methyl, trifluoromethyl, cyano.
In some preferred embodiments, the above-mentioned monoalkylamino group, dialkylamino group, monoalkylaminoalkyl group, dialkylaminoalkyl group are each preferably (C) 1 -C 6 Alkyl) NH- (C 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-, C 1 -C 6 alkyl-NH-C 1 -C 6 Alkyl-, (C) 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-C 1 -C 6 Alkyl-;
in some preferred embodiments, the 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl is preferably (3-12 membered cycloalkyl or heterocycloalkyl) -C 1 -C 6 Alkyl-; the alkyl-substituted amino group is preferably (C) 1 -C 6 Alkyl) NH-or (C) 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-;
in some preferred embodiments, R 11 、R 12 Independently selected from hydrogen, halogen, methyl;
in some preferred embodiments, when R 11 、R 12 When taken together with the attached N atom form a 5-8 membered heterocycloalkyl, the 3-8 membered heterocycloalkyl is preferably
Figure BDA0003785353610000061
In some preferred embodiments, R p And R is R q Forming a three-membered carbocyclic ring with the attached carbon atom;
in some preferred embodiments, the C 1 -C 6 Preferably HO-CH 2 -、HO-CH 2 -CH 2 -;
In one embodiment of the present invention, the pharmaceutically acceptable salt, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, represented by formula (I), is characterized in that the compound has the following structure:
Figure BDA0003785353610000062
/>
Figure BDA0003785353610000071
/>
Figure BDA0003785353610000081
Figure BDA0003785353610000091
thus, throughout this specification, one skilled in the art may select the groups and substituents thereof in the nitrogen-containing heterocyclic compound of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, stereoisomer, solvate, polymorph or prodrug thereof, to provide a stable nitrogen-containing heterocyclic compound of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, stereoisomer, solvate, polymorph or prodrug thereof, including but not limited to the compounds described in the examples of the invention.
The nitrogen-containing heterocyclic compounds of formula I, or pharmaceutically acceptable salts thereof, or enantiomers, diastereomers, tautomers, torsional isomers, solvates, polymorphs, or prodrugs thereof, according to the invention, may be synthesized by methods that include methods similar to those known in the chemical arts, the steps and conditions of which may be referred to in the art as procedures and conditions of similar reactions, particularly in light of the description herein. The starting materials are typically from commercial sources, such as Aldrich or can be readily prepared using methods well known to those skilled in the art (available via SciFinder, reaxys on-line databases).
In the invention, the nitrogen-containing heterocyclic compound shown in the formula I or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof can also be prepared into the nitrogen-containing heterocyclic compound shown in the formula I or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, and other nitrogen-containing heterocyclic compounds shown in the formula I or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof can be obtained by peripheral modification by adopting a conventional method in the art.
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined in formula I, unless otherwise indicated. The following reaction schemes and examples are provided to further illustrate the present invention.
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-1, which comprises the following steps:
a) Reacting a compound of formula (a) with a haloaryl aldehyde and an aryl hydrazine to produce a compound of formula (B);
b) Combining a compound of the formula (B) with a nitrogen-containing spiro or bridged ring radical R 1 -H under alkaline conditions, to produce a compound of formula (C);
c) Reacting a compound of the general formula (C) with a transition metal reagent to generate a compound of the general formula (D) through an insertion carbonyl reaction;
d) Closing ring of the compound of the general formula (D) under the condition of acid catalysis to generate a compound of the general formula (E);
e) Oxidizing a compound of the general formula (E) in the presence of an oxidizing agent to produce a compound of the general formula (F);
f) Reacting a compound of the general formula (F) with a substituted alkylhydroxy or amino compound H-L-R under the action of a base 2 Substitution reaction occurs to produce the compound of the general formula (I-1).
Figure BDA0003785353610000101
X is halogen, and the definition of each other group is as described above;
preferably, the steps a), b), c), d), e), f) are each performed in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, hydrofluoric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid or combinations thereof.
Preferably, the transition metal palladium catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) Tetrakis (triphenylphosphine) palladium (Pd (PPh) 3 ) 4 ) Palladium acetate, palladium chloride, dichloro-bis (triphenylphosphine) palladium, trifluoropalladium acetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (triphenylphosphine) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-benzylphosphine, or a combination thereof.
Preferably, the peroxide is selected from the group consisting of: hydrogen peroxide, sodium peroxide, ammonium persulfate, m-chloroperoxybenzoic acid, peracetic acid, t-butyl ether peroxide, potassium peroxymonosulfonate, dibenzoyl peroxide, t-butyl hydroperoxide, and the like, or combinations thereof.
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-2, which comprises the following steps of g-l:
g) The compound of the general formula (G) is a nitrogenous spiro or bridged ring compound R 1 -H under alkaline conditions, substitution reaction to form a compound of formula (H);
h) Reacting a compound of the general formula (H) through an insertion carbonyl reaction under the catalysis of transition metal to generate a compound of the general formula (I);
i) Oxidizing a compound of the general formula (I) in the presence of an oxidizing agent to generate a compound of the general formula (J);
j) Reacting a compound of the general formula (J) with an alkyl-substituted hydroxy or amino compound H-L-R under the action of a base 2 Generating a compound of a general formula (K) through substitution reaction;
k) Reacting a compound of formula (K) with hydrazine hydrate to produce a compound of formula (L);
l) coupling the compound of formula (L) with a substituted arylboronic acid (ester) or a substituted aryl halide, etc. under the catalysis of a transition metal to produce the compound of formula (I-2).
Figure BDA0003785353610000111
The definition of each group is as above;
preferably, the steps g), h), i), j), k), l) are each carried out in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, hydrofluoric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid or combinations thereof.
Preferably, the transition metal palladium catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) Tetrakis (triphenylphosphine) palladium (Pd (PPh) 3 ) 4 ) Palladium acetate, palladium chloride, dichloro-bis (triphenylphosphine) palladium, trifluoropalladium acetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (triphenylphosphine) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-benzylphosphine, or a combination thereof.
Preferably, the peroxide is selected from the group consisting of: hydrogen peroxide, sodium peroxide, ammonium persulfate, m-chloroperoxybenzoic acid, peroxyacetic acid, t-butyl ether peroxide, potassium peroxymonosulfonate, dibenzoyl peroxide, t-butyl hydroperoxide, and the like, or combinations thereof.
Preferably, the transition metal copper catalyst is selected from the group consisting of: copper acetate, copper trifluoroacetate, copper sulfate, copper bromide, copper chloride, copper iodide, or a combination thereof.
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-3, which comprises the following steps of m-p:
Figure BDA0003785353610000121
X is halogen, pg is benzyl or Boc protecting group, and the definition of other groups is as described above;
m) reacting a compound of the general formula (M) with a nitrogen-containing spiro or bridged ring compound R 1 -H under basic conditions, to produce a compound of formula (N);
n) reacting a compound of the formula (N) with an alkyl-substituted hydroxy or amino compound H-L-R under the action of a base 2 Generating a compound of a general formula (O) through substitution reaction;
o) deprotecting the compound of formula (O) by acid-catalyzed or transition metal-catalyzed hydrogenation to form a compound of formula (P);
p) carrying out coupling reaction on the compound of the general formula (P) and substituted aryl halide or aryl boric acid under the catalysis of transition metal to generate a compound of the general formula (I-3);
preferably, the steps m), n), o), p) are each carried out in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, hydrofluoric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid or combinations thereof.
Preferably, the transition metal palladium catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) Tetrakis (triphenylphosphine) palladium (Pd (PPh) 3 ) 4 ) Palladium acetate, palladium chloride, dichloro-bis (triphenylphosphine) palladium, trifluoropalladium acetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (triphenylphosphine) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-benzylphosphine, or a combination thereof.
Preferably, the transition metal copper catalyst is selected from the group consisting of: copper acetate, copper trifluoroacetate, copper sulfate, copper bromide, copper chloride, copper iodide, or a combination thereof.
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-4, which comprises the following steps:
Figure BDA0003785353610000131
R 4 Is methyl or trifluoromethyl, X is halogen, and the definition of other groups is as described above;
q) reacting the compound of formula (Q) with a suitable reagent (e.g., CDI reagent, etc.), to produce a compound of formula (R);
r) reacting the compound of formula (R) with a halogenating agent to produce a compound of formula (S);
s) reacting a compound of the general formula (S) with a nitrogen-containing spiro or bridged ring compound R 1 -H under basic conditions, to produce a compound of formula (T);
t) reacting a compound of the formula (T) with an alkyl-substituted hydroxy or amino compound H-L-R under the action of a base 2 Generating a compound of a general formula (I-4) through substitution reaction;
preferably, the steps q), r), s), t) are each carried out in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, hydrofluoric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid or combinations thereof.
Preferably, the halogenating agent is selected from the group consisting of: phosphorus oxychloride, phosphorus oxybromide, oxalyl chloride, thionyl chloride, carbon tetrachloride/triphenylphosphine, carbon tetrabromide/triphenylphosphine, or combinations thereof.
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-5, which comprises the following steps:
Figure BDA0003785353610000141
R a is alkyl, and the definition of each other group is as above;
u) reacting a compound of the general formula (U) with a nitrogen-containing spiro or bridged ring compound R 1 -H under basic conditions, to produce a compound of formula (V);
v) reacting a compound of the formula (V) with an alkyl-substituted hydroxy or amino compound H-L-R under the action of a base 2 Generating a compound of a general formula (W) through substitution reaction;
w) subjecting the compound of formula (W) to hydrolysis under the action of a base to produce a compound of formula (X);
x) reacting the compound of formula (X) in a suitable reagent (e.g., a condensing reagent, etc.) to produce a compound of formula (Y);
y) reducing the compound of the general formula (Y) under the action of a reducing agent to generate a compound of the general formula (Z);
Z) ring-closing reaction of the compound of the general formula (Z) with anhydride of R4 or alkyl triethyl of R4 to generate a compound of the general formula (I-5);
preferably, the steps u), v), w), x), y), z) are each performed in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, hydrofluoric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid or combinations thereof.
Preferably, the condensing agent is selected from the group consisting of HATU, HBTU, HCTU, TSTU, HOBT, EDCI, DCC, CDI, BOP, pyBOP and the like or combinations thereof
The invention also provides a preparation method of the nitrogen-containing heterocyclic compound shown in the formula I-6, which comprises the following steps of:
Figure BDA0003785353610000151
x is halogen, and the definition of each other group is as described above;
1) The compound of the general formula (A-6) is a nitrogenous spiro or bridged ring compound R 1 -H under alkaline conditions, carrying out substitution reaction to produce a compound of formula (B-6);
2) Reacting a compound of the general formula (B-6) with an alkyl-substituted hydroxy or amino compound H-L-R under the action of a base 2 Generating a compound of a general formula (C-6) by substitution reaction;
3) Reacting a compound of formula (C-6) with a suitable halogenating reagent to produce a compound of formula (D-6);
4) The compound of the general formula (D-6) and arylboric acid or boric acid ester are subjected to coupling reaction under the catalysis of transition metal palladium to generate the compound of the general formula (I-6).
Preferably, the steps q), r), s), t) are each carried out in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the halogenating agent is selected from the group consisting of: chlorosuccinimide, bromosuccinimide, iodosuccinimide, bromine, iodine, or combinations thereof;
preferably, the transition metal palladium catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) Tetrakis (triphenylphosphine) palladium (Pd (PPh) 3 ) 4 ) Palladium acetate, palladium chloride, dichloro-bis (triphenylphosphine) palladium, trifluoropalladium acetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (triphenylphosphine) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-benzylphosphine, or a combination thereof.
The invention also provides a preparation method of the nitrogen-containing heterocyclic biaryl compound shown in the formula I-7, which comprises the following steps:
g) Combining a compound of the formula (A-7) with R 1 -H substitution occurs under alkaline conditionsReacting to produce a compound of formula (B-7);
h) Combining a compound of the formula (B-7) with R 2 -L-H is subjected to substitution reaction or metal catalytic coupling reaction under alkaline condition to generate a compound of the general formula (C-7);
i) The compound of the general formula (C-7) and arylboric acid or arylboric acid ester or arylmetal reagent (Ar-M) are subjected to a transition metal catalytic coupling reaction to generate the general formula (I-7).
Figure BDA0003785353610000161
X is halogen, and the definition of each other group is as described above;
preferably, each of said steps a), b), c) is performed in a solvent, and said solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methylpyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, methylene chloride, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium t-butoxide, sodium t-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or a combination thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or combinations thereof.
Preferably, the transition metal catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) Tetrakis (triphenylphosphine) palladium (Pd (PPh) 3 ) 4 ) Palladium acetate, palladium chloride, dichloro-bis (triphenylphosphine) palladium, trifluoropalladium acetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (triphenylphosphine) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: three kinds ofTertiary butyl phosphine, tri-tertiary butyl phosphine tetrafluoroborate, tri-n-butyl phosphine, triphenylphosphine, tri-p-benzyl phosphine, tricyclohexyl phosphine, tri-o-benzyl phosphine, or a combination thereof.
The necessary starting materials or reagents for preparing the compounds of formula I may be obtained by commercial reagents or prepared by synthetic methods known in the art. The compounds of the invention may be prepared as free bases or as salts thereof with acids, as described in the experimental section below. The term pharmaceutically acceptable salt refers to a pharmaceutically acceptable salt as defined herein, and has all of the pharmaceutical activity of the parent compound. Pharmaceutically acceptable salts can be prepared by adding the corresponding acid to a suitable organic solvent for the organic base, and processing according to conventional methods.
Examples of salification include: salifying with inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid; and salts formed with organic acids, such as acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, eurynic acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, glutamic acid, glycolic acid, hydroxynaphthoic acid, 2-hydroxyethanesulfonic acid, lactic acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, muconic acid, 2-naphthalenesulfonic acid, propionic acid, salicylic acid, succinic acid, tartaric acid, p-toluenesulfonic acid or trimethylacetic acid.
The nitrogen-containing heterocyclic compounds of formula I, or pharmaceutically acceptable salts thereof, or enantiomers, diastereomers, tautomers, torsional isomers, solvates, polymorphs, or prodrugs thereof, may have one or more chiral carbon atoms, and thus may be isolated as optically pure isomers, e.g., pure enantiomers, or racemates, or mixed isomers. Pure single isomers may be obtained by separation methods in the art, such as chiral crystallization to form salts, or chiral preparative column separation.
The chemicals used in the synthetic routes described in this patent include solvents, reagents, catalysts, and protecting groups, deprotecting groups, including t-butoxycarbonyl (Boc). The above-described methods may additionally include steps prior to or subsequent to the steps specifically described herein, and suitable protecting groups may be added or removed to provide the subject compounds. In addition, the various synthetic steps may be performed alternately or sequentially to obtain the final target product.
It is another object of the present invention to provide a medicament for treating or preventing tumors and a composition thereof. The technical scheme for achieving the purpose is as follows:
the invention provides a pharmaceutical composition comprising an effective amount of a nitrogen-containing heterocyclic compound shown as the formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, and (one or more) pharmaceutically acceptable carrier (pharmaceutical auxiliary materials). For example, such pharmaceutical compositions may comprise one or more additional nitrogen-containing heterocyclic biaryl compounds of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof. In the pharmaceutical composition, the nitrogen-containing heterocyclic biaryl compound shown in the formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof can be used in an amount effective for treatment.
The invention provides a pharmaceutical composition for treating tumors, which consists of a nitrogenous heterocyclic compound shown in the general formula I, or pharmaceutically acceptable salts thereof, or enantiomers, diastereomers, tautomers, torsional isomers, solvates, polymorphs or prodrugs thereof and a pharmaceutically acceptable carrier.
It is a further object of the present invention to provide the use of the above compounds. The technical scheme for achieving the purpose is as follows:
the invention also provides a nitrogenous heterocyclic compound shown as a formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, which are used for preparing Ras mutant protein inhibitorIs used in the application of (a); in the application, the Ras mutant protein can be KRAS G12D The method comprises the steps of carrying out a first treatment on the surface of the The Ras mutein inhibitors are useful in mammalian organisms; it is also useful in vitro, mainly as an experimental use, for example: provides comparison as a standard sample or a control sample, or is prepared into a kit according to a conventional method in the field, and provides rapid detection for the inhibition effect of Ras mutant protein.
The invention also provides an application of the nitrogen-containing heterocyclic compound shown in the formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof in preparing medicines; the medicine can be used for treating diseases related to activity or expression quantity of Ras mutant protein; alternatively, the drug may be a therapeutic drug for tumors. The Ras mutant protein can be KRAS G12D . The tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, bile duct cancer, brain cancer, leukemia, lymphoma, fibroma, sarcoma, basal cell carcinoma, glioma, renal cancer, melanoma, bone cancer, thyroid cancer, nasopharyngeal cancer, pancreatic cancer, etc.
Another aspect of the invention relates to a method for preventing and/or treating a disease associated with activity or expression of a Ras mutein, comprising administering to a patient a therapeutically effective amount of the nitrogen-containing heterocyclic compound of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof.
Another aspect of the present invention relates to a method for preventing and/or treating tumors, which comprises administering to a patient a therapeutically effective amount of the nitrogen-containing heterocyclic compound represented by formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof.
Another aspect of the invention relates to a medicament for preventing and/or treating a disease or tumor associated with activity or expression of a Ras mutein, which comprises the nitrogen-containing heterocyclic compound of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof.
The nitrogen-containing heterocyclic compound shown in the general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof is used for preparing medicaments for treating diseases related to activity or expression quantity of Ras mutant protein, in particular to medicaments for treating tumors. The tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, bile duct cancer, brain cancer, leukemia, lymphoma, fibroma, sarcoma, basal cell carcinoma, glioma, renal cancer, melanoma, bone cancer, thyroid cancer, nasopharyngeal cancer, pancreatic cancer, etc.
The invention relates to a compound with the structural characteristics of a general formula I, which can inhibit various tumor cells, in particular can kill KRAS with high efficiency G12D The tumor related to abnormal mutein signal pathway is a new therapeutic drug with a novel action mechanism.
The pharmaceutical excipients can be those which are widely used in the field of pharmaceutical production. Adjuvants are used primarily to provide a safe, stable and functional pharmaceutical composition, and may also provide means for allowing the subject to dissolve at a desired rate after administration, or for promoting effective absorption of the active ingredient after administration of the composition. The pharmaceutical excipients may be inert fillers or provide a function such as stabilizing the overall pH of the composition or preventing degradation of the active ingredients of the composition. The pharmaceutical excipients can comprise one or more of the following excipients: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, sizing agents, disintegrants, lubricants, anti-adherents, glidants, wetting agents, gelling agents, absorption retarders, dissolution inhibitors, enhancing agents, adsorbents, buffering agents, chelating agents, preservatives, colorants, flavoring agents, and sweeteners.
Substances that may be pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, aluminum stearate, lecithin, serum proteins, such as human serum proteins, buffer substances, such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silicon, magnesium trisilicate, polyvinylpyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, lanolin, sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; a gum powder; malt; gelatin; talc powder; adjuvants such as cocoa butter and suppository waxes; oils such as peanut oil, cotton seed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycol compounds such as propylene glycol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic salt; ringer's solution; ethanol, phosphate buffer, and other non-toxic suitable lubricants such as sodium lauryl sulfate and magnesium stearate, coloring agents, releasing agents, coating materials, sweetening, flavoring and perfuming agents, preserving and antioxidant agents.
The pharmaceutical compositions of the present invention may be prepared in accordance with the disclosure using any method known to those of skill in the art. For example, conventional mixing, dissolving, granulating, emulsifying, levigating, encapsulating, entrapping or lyophilizing processes.
The pharmaceutical dosage forms of the compounds of the present invention may be provided in the form of immediate release, controlled release, sustained release or target drug release systems. For example, common dosage forms include solutions and suspensions, (micro) emulsions, ointments, gels and patches, liposomes, tablets, dragees, soft or hard shell capsules, suppositories, ovules, implants, amorphous or crystalline powders, aerosols and freeze-dried formulations. Depending on the route of administration used, special devices may be required to administer or administer the drug, such as syringes and needles, inhalers, pumps, injection pens, applicators, or Special bottles (Special flash). Pharmaceutical dosage forms often consist of a drug, excipients and a container/sealing system. One or more excipients (also known as inactive ingredients) may be added to the compounds of the present invention to improve or promote the manufacture, stability, administration and safety of the drug, and may provide a means to achieve a desired drug release profile. Thus, the type of excipient added to a drug may depend on various factors, such as the physical and chemical characteristics of the drug, the route of administration, and the manufacturing steps. Pharmaceutically acceptable excipients are present in this field and include those listed in the various pharmacopoeias. (see U.S. Pharmacopeia (U.S.Pharmacopeia, USP), japanese Pharmacopeia (Japanese Pharmacopoeia, JP), european Pharmacopeia (European Pharmacopoeia, EP) and British Pharmacopeia (British pharmacopoeia, BP); U.S. food and drug administration (the U.S. food and Drug Administration, www.fda.gov) drug evaluation and research center (Centerfor Drug Evaluation and Research, CEDR) publications, for example, inactive ingredient guide (Inactive Ingredient Guide, 1996); pharmaceutical additives handbook written by Ash and Ash (Hand book of Pharmaceutical Additives,2002, incorporated information resources, inc. (Synapse Information Resources, inc., endiott NY; etc.).
Pharmaceutical dosage forms of the compounds of the present invention may be manufactured by any of the methods well known in the art, for example by conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tabletting, suspending, extruding, spray-drying, grinding, emulsifying, (nano/micro) encapsulating, packaging, or lyophilizing processes. As noted above, the compositions of the present invention may include one or more physiologically acceptable inactive ingredients that facilitate processing of the active molecule into a formulation for pharmaceutical use.
The pharmaceutical compositions of the invention may be administered topically or systemically, e.g. for enteral, such as rectal or oral administration, or for parenteral administration to a mammal (especially a human), and comprise a therapeutically effective amount of a compound according to the invention, a stereoisomer or a pharmaceutically acceptable salt thereof as active ingredient, together with a pharmaceutically acceptable excipient, such as a pharmaceutically acceptable carrier. A therapeutically effective amount of the active ingredient is defined as above and below and depending on the species, weight, age, individual condition, individual pharmacokinetic parameters, disease to be treated and mode of administration of the mammal, for enteral administration, such as oral administration, the compounds of the invention can be formulated in a wide variety of dosage forms.
The pharmaceutical compositions and dosage forms may comprise one or more compounds of the present invention, stereoisomers thereof, or one or more pharmaceutically acceptable salts thereof, as an active ingredient. The pharmaceutically acceptable carrier may be a solid or a liquid. Solid forms of preparation include powders, tablets, pills, troches, capsules, cachets, suppositories, and dispersible granules. The solid carrier may also be one or more substances that act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. In powders, the carrier is usually a finely divided solid, which is a mixture with the finely divided active component. In tablets, the active ingredient is typically mixed with a carrier having the necessary binding capacity in a suitable ratio and compacted in the shape and size desired. Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, methylcellulose, sodium carboxymethylcellulose, low melting waxes, cocoa butter and the like. Formulations of the active compounds may include an encapsulating material as a carrier providing a capsule in which the active ingredient with or without the carrier is surrounded by a carrier to which it is bound.
Other forms suitable for oral administration include liquid form preparations, including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions, or solid form preparations intended to be converted to liquid form preparations shortly before use. The emulsion may be prepared in solution, for example in an aqueous propylene glycol solution, or may contain an emulsifier such as lecithin, sorbitan monooleate, or acacia. Aqueous solutions can be prepared by dissolving the active ingredient in water and adding suitable colorants, fragrances, stabilizers and thickeners. Aqueous suspensions may be prepared by dispersing the finely divided active component in water with binders such as natural or synthetic gums, resins, methylcellulose, carboxymethylcellulose and other commonly used suspending agents. Solid form preparations include solutions, suspensions and emulsions which may contain, in addition to the active ingredient, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
Exemplary combinations for rectal administration include suppositories, which may contain, for example, suitable non-irritating excipients such as cocoa butter, synthetic glycerides or polyethylene glycols, which are solid at ordinary temperatures, but melt and/or dissolve in the rectal cavity to release the drug.
The compounds of the invention may also be administered parenterally, for example, by inhalation, injection or infusion, such as by intravenous, intra-arterial, intra-osseous, intramuscular, intra-cerebral, extra-cerebral, intra-synovial, intra-sternal, intrathecal, intralesional, intracranial, intratumoral, intradermal, and subcutaneous injection or infusion.
Thus, for parenteral administration, the pharmaceutical compositions of the invention may be in the form of sterile injectable or infusible preparations, e.g., as sterile aqueous or oleaginous suspensions. The suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (e.g., tween 80) and suspending agents. The sterile injectable or infusible formulation may also be a sterile injectable or infusible solution or suspension in a non-toxic parenterally acceptable diluent or solvent. For example, the pharmaceutical composition may be a solution in 1, 3-butanediol. Other examples of acceptable vehicles and solvents that may be used in the pharmaceutical compositions of the present invention include, but are not limited to, mannitol, water, ringer's solution, and isotonic sodium chloride solution. In addition, sterile, non-volatile oils are conventionally employed as a solvent or suspending medium. Any bland fixed oil may be employed for this purpose including synthetic mono-or diglycerides. Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant. Solutions for parenteral use may also include suitable stabilizers and, if desired, buffer substances. Suitable stabilizers include antioxidants such as sodium bisulfate, sodium sulfite or ascorbic acid, citric acid and salts thereof and sodium EDTA alone or in combination. The parenteral solution may also contain preservatives such as benzalkonium chloride, parahydroxybenzoic acid or propyl parahydroxybenzoate and chlorobutanol.
For inhalation or nasal administration, suitable pharmaceutical formulation chamber particles, aerosols, powders, mists or droplets, for example, have an average size of about 10 microns or less in diameter. For example, compositions for inhalation may be prepared in saline as solutions, using benzyl alcohol or other suitable preservatives, absorption promoters for improving bioavailability, fluorocarbon and/or other solubilizing or dispersing agents known in the art.
The pharmaceutical compositions of the present invention may also be administered topically to the skin or mucosa. For topical application, the pharmaceutical composition may be, for example, a lotion, gel, paste, tincture, transdermal patch, gel for transmucosal delivery.
The pharmaceutical compositions may be formulated in a suitable ointment comprising the active ingredient suspended or dissolved in a carrier. Carriers for topical administration of the compounds of the invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene, polyoxypropylene compounds, emulsifying waxes and water. Alternatively, the pharmaceutical compositions may be formulated as suitable lotions or emulsions comprising the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The pharmaceutical compositions of the present invention may also be administered topically to the lower intestinal tract in rectal suppository formulations or in suitable enema formulations. Suitable pharmaceutical excipients (e.g. carriers) and methods for preparing pharmaceutical dosage forms are described in standard reference textbooks in the pharmaceutical formulation arts (Remington's Pharmaceutical Sciences, mack Publishing Company)
It is understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute new or preferred technical solutions. The limited space is not described in any more detail herein.
Terminology
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are hereby incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the inventive subject matter. In this application, the singular is used to include the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or" means "and/or" unless stated otherwise. Furthermore, the terms "include," as well as other forms, such as "comprising," "including," and "containing," are not limiting.
The definition of standard chemical terms can be found in references (including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY TH ED." vols. A (2000) and B (2001), plenum Press, new York). Conventional methods within the skill of the art, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods are employed unless otherwise indicated. Unless specifically defined otherwise, the terms used herein in the description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques may be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the manufacturer's instructions for the kit, or in a manner well known in the art or in accordance with the teachings of the present invention. The techniques and methods described above may generally be practiced according to conventional methods well known in the art, based on a number of general and more specific descriptions in the literature cited and discussed in this specification. In this specification, groups and substituents thereof can be selected by one skilled in the art to provide stable moieties and compounds.
When substituents are described by conventional formulas written from left to right, the substituents also include chemically equivalent substituents obtained when writing formulas from right to left. For example, -CH 2 O-is equivalent to-OCH 2 -。
The section headings used herein are for purposes of organizing articles only and should not be construed as limiting the subject matter. All documents or portions of documents cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a simplified symbol to indicate the total number of carbon atoms present in the group. For example, C1-6 alkyl, C 1-6 Alkyl or C 1- C 6 Alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the reduced notation does not include carbon that may be present in a substituent of the group.
Certain chemical groups defined herein are preceded by a simplified symbol to indicate the total number of carbon atoms present in the group. For example, C1-C6 alkyl refers to an alkyl group as defined below having a total of 1, 2, 3, 4, 5 or 6 carbon atoms. The total number of carbon atoms in the reduced notation does not include carbon that may be present in a substituent of the group.
In this context, a numerical range as defined in substituents, such as 0 to 4, 1-4, 1 to 3, etc., indicates an integer within the range, such as 1-6 is 1, 2, 3, 4, 5, 6.
In the various parts of the invention, linking substituents are described. When the structure clearly requires a linking group, the markush variables recited for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for that variable enumerates an "alkyl" or "aryl" group, it will be understood that the "alkyl" or "aryl" represents a linked alkylene group or arylene group, respectively.
In some specific structures, when an alkyl groupWhen explicitly indicated as a linking group, then the alkyl group represents a linked alkylene group, e.g., the group "halo-C 1 -C 6 C in alkyl' 1 -C 6 Alkyl is understood to mean C 1 -C 6 Alkylene (e.g., methylene, ethylene, propylene, butylene, pentylene, isopropylene, isobutylene, sec-butylene, tert-butylene, isopentylene, 2-methylbutylene, 1-ethylpropylene, 1, 2-dimethylpropylene, neopentylene or 1, 1-dimethylpropylene, etc.).
In addition to the foregoing, when used in the specification and claims of this application, the following terms have the meanings indicated below, unless specifically indicated otherwise.
The term "comprising" is an open-ended expression, i.e. including what is indicated by the invention, but not excluding other aspects.
The term "substituted" refers to any one or more hydrogen atoms on a particular atom being substituted with a substituent, including heavy hydrogen and variants of hydrogen, so long as the valence of the particular atom is normal and the substituted compound is stable.
In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a specific substituent. Further, when the group is substituted with 1 or more of the substituents, the substituents are independent of each other, that is, the 1 or more substituents may be different from each other or the same. Unless otherwise indicated, a substituent group may be substituted at each substitutable position of the substituted group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, then the substituents may be the same or different at each position.
In the various parts of the present specification, substituents of the presently disclosed compounds are disclosed in terms of the type or scope of groups. It is specifically noted that the present invention includes each individual subcombination of the individual members of these group classes and ranges. For example, the term "C 1 ~C 6 Alkyl "or" C 1-6 Alkyl "means in particular methyl, ethyl, C independently disclosed 3 Alkyl, C 4 Alkyl, C 5 Alkyl and C 6 An alkyl group; "C 1-4 Alkyl "refers specifically to independently disclosed methyl, ethyl, C 3 Alkyl (i.e. propyl, including n-propyl and isopropyl), C 4 Alkyl (i.e., butyl, including n-butyl, isobutyl, sec-butyl, and tert-butyl).
In the present application, the term "halogen" refers to fluorine, chlorine, bromine or iodine; "hydroxy" refers to an-OH group; "hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxy (-OH); "carbonyl" refers to a-C (=o) -group; "nitro" means-NO 2 The method comprises the steps of carrying out a first treatment on the surface of the "cyano" refers to-CN; "amino" means-NH 2 The method comprises the steps of carrying out a first treatment on the surface of the "substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., mono-, di-, alkylamido, aralkylamino, heteroaralkylamino; "carboxy" means-COOH; "acyl" refers to a-C (=o) H group; "sulfone group" means-S (=O) 2 -a group; "sulfoxide" refers to the-S (=o) -group; "sulfonyl" means-S (=o) 2 An H group; "urea" means-NH-C (=o) -NH 2 The method comprises the steps of carrying out a first treatment on the surface of the "sulfonylurea" means-S (=o) 2 -NH-C(=O)-NH 2 A group; "alkoxy" refers to an alkyl-O-group as defined below.
In the present application, as part of a group or other group (e.g. as used in halogen substituted alkyl groups or the like), the term "alkyl" means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, free of unsaturated bonds, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms and being linked to the remainder of the molecule by a single bond. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl and the like.
The term "alkylene" as used herein refers to the number of carbon atoms (typically1-6 carbon atoms) and having two monovalent radical centers derived from the removal of two hydrogen atoms from the same or two different carbon atoms of the parent alkane. Typical alkylene groups include, but are not limited to, methylene (-CH) 2 (-) ethylene { including 1, 2-ethylene (-CH) 2 CH 2 (-), 2-dimethylene (-CH (CH) 3 ) (-) } propylene { including 2-methylpropylene (-CH (CH) 3 )CH 2 (-), isopropylidene (-C (CH) 3 ) 2 (-), 1, 3-propylene (-CH) 2 CH 2 CH 2 (-) } butylene { including 1, 4-butylene (-CH) 2 CH 2 CH 2 CH 2 -)}。
In the present application, the term "alkenyl" as part of a group or other group means a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being linked to the rest of the molecule by a single bond, such as, but not limited to, ethenyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In this application, the term "alkynyl" as part of a group or other group means a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, containing at least one triple bond and optionally one or more double bonds, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being attached to the remainder of the molecule by a single bond, such as, but not limited to, ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl, and the like.
In this application, as part of a group or other group, the term "cycloalkyl" means a stable, non-aromatic, mono-or polycyclic hydrocarbon group consisting of only carbon and hydrogen atoms, which may include fused ring systems, bridged ring systems, or spiro ring systems, having from 3 to 15 carbon atoms, preferably from 3 to 10 carbon atoms, more preferably from 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the remainder of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in cycloalkyl groups may optionally be oxidized. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexanyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7-dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [ 2.2.2.2 ] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, bicyclo [ 2.1.1 ] octadienyl, adamantylene, and the like.
In the present application, the term "heterocyclyl" as part of a group or other group means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen and sulfur. Unless specifically indicated otherwise in the present specification, a heterocyclyl group may be a monocyclic, bicyclic, tricyclic or more cyclic ring system, which may include fused, bridged or spiro ring systems; the nitrogen, carbon or sulfur atoms in the heterocyclyl may optionally be oxidized; the nitrogen atom may optionally be quaternized; and the heterocyclyl may be partially or fully saturated. The heterocyclic group may be attached to the remainder of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclyl groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the remainder of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl groups are preferably stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro groups comprising 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro groups comprising 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxacyclopentyl, tetrahydroisoquinolyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indolinyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimidyl, and the like.
In the present application, the term "heterocycloalkyl" means a stable 3-to 20-membered saturated cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen and sulfur, as part of a group or other groups. Unless otherwise specifically indicated in the present specification, a heterocycloalkyl group may be either a monocyclic ("monocyclic heterocycloalkyl") or a bicyclic, tricyclic or higher ring system, which may include fused, bridged or spiro ring systems (e.g., bicyclic systems ("bicyclic heterocycloalkyl"). The heterocycloalkyl bicyclic ring system may include one or more heteroatoms in one or both rings, and is saturated for purposes of the present invention, a heterocycloalkyl group is preferably a stable 4-to 12-membered saturated monocyclic, bicyclic, bridged or spiro ring group containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 7-membered saturated monocyclic, bicyclic, bridged or spiro ring group containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, in particular, the 4-to 7-membered heterocycloalkyl group may contain 3, 4, 5 or 6 carbon atoms and one or two of the heteroatoms or heteroatom-containing groups, provided that the total number of ring atoms is not greater than 7, in particular the heterocycloalkyl group may contain 3, 5 or 6 carbon atoms and the total number of heteroatoms or heteroatom-containing groups ("the total number of heteroatoms may be 3, 5 or 6 heteroatoms").
In this application, the term "aryl" as part of a group or other group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, aryl groups may be monocyclic, bicyclic, tricyclic or more ring systems, and may also be fused to cycloalkyl or heterocyclyl groups as defined above, provided that the aryl groups are linked to the remainder of the molecule by single bonds via atoms on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthryl, phenanthryl, fluorenyl, 2, 3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, as part of a group or other group, the term "heteroaryl" means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur within the ring. Unless otherwise specifically indicated in the present specification, heteroaryl groups may be monocyclic, bicyclic, tricyclic or more ring systems, and may also be fused to cycloalkyl or heterocyclyl groups as defined above, provided that heteroaryl groups are attached to the remainder of the molecule via an atom on an aromatic ring by a single bond. The nitrogen, carbon, or sulfur atoms in the heteroaryl group may optionally be oxidized; the nitrogen atom may optionally be quaternized. For the purposes of the present invention, heteroaryl groups are preferably stable 5-to 12-membered aromatic groups comprising 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably stable 5-to 10-membered aromatic groups comprising 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or 5-to 6-membered aromatic groups comprising 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furanyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindazolyl, purinyl, quinolinyl, isoquinolinyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxatriazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, phenanthroline, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthyridinyl, [1,2,4] triazolo [4, 3-triazolo [1, 4] pyridazine, 3-1, 4-imidazo [1, 4] triazolo [1, 4, 3-triazolo [1, 4] pyridazine, 3-1, 4-imidazo [ 2,4] a ] 1, 4-imidazo [ 2, 4-a ] and the like.
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above.
As used herein, the singular forms "a", "an", and "the" are understood to include plural referents unless the context clearly dictates otherwise. Furthermore, the term "comprising" is an open-ended limitation and does not exclude other aspects, i.e. it includes the content indicated by the invention.
Unless otherwise indicated, the present invention employs conventional methods of mass spectrometry, elemental analysis, and the various steps and conditions are referred to in the art by conventional procedures and conditions.
The present invention employs, unless otherwise indicated, standard nomenclature for analytical chemistry, organic synthetic chemistry and optics, and standard laboratory procedures and techniques. In some cases, standard techniques are used for chemical synthesis, chemical analysis, and light emitting device performance detection.
In addition, unless explicitly indicated otherwise, the description of the invention as "…" independently is to be understood broadly as meaning that each individual described may be independent of the other, and may be the same or different. In more detail, the description "… is independently" may mean that specific options expressed between the same symbols in different groups do not affect each other; it may also be expressed that specific options expressed between the same symbols in the same group do not affect each other.
Those skilled in the art will appreciate that, in accordance with convention used in the art, the present application describes the structural formula of a group as used in
Figure BDA0003785353610000271
Refers to the corresponding groupThrough which it is linked to other fragments, groups in the compound.
In this application, "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted aryl groups and unsubstituted aryl groups.
The terms "moiety", "structural moiety", "chemical moiety", "group", "chemical group" as used herein refer to a particular fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded or attached to a molecule.
"stereoisomers" refer to compounds that consist of the same atoms, are bonded by the same bonds, but have different three-dimensional structures. The present invention is intended to cover various stereoisomers and mixtures thereof.
When an olefinic double bond is contained in the compounds of the present invention, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention.
The compounds of the invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and thus may be produced in enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting racemates, diastereomers or enantiomers as starting materials or intermediates. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as crystallization and chiral chromatography.
Conventional techniques for preparing/separating individual isomers include chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, chiral high performance liquid chromatography, see, for example, gerald gabiz and Martin g.schmid (eds.), chiral Separations, methods and Protocols, methods in Molecular Biology, vol.243,2004; m.stalcup, chiral Separations, annu.rev.animal.chem.3:341-63, 2010; fumigs et al (EDs.), VOGEL' S ENCYCLOPEDIA OF PRACTICAL ORGANIC CHEMISTRY.sup.TH ED, longman Scientific and Technical Ltd., essex,1991,809-816; heller, acc.chem.Res.1990,23,128.
In the present application, the term "pharmaceutically acceptable salt" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
By "pharmaceutically acceptable acid addition salt" is meant a salt with an inorganic or organic acid that retains the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formate, acetate, 2-dichloroacetate, trifluoroacetate, propionate, hexanoate, octanoate, decanoate, undecylenate, glycolate, gluconate, lactate, sebacate, adipate, glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate, cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, alginate, ascorbate, salicylate, 4-aminosalicylate, naphthalenedisulfonate, and the like. These salts can be prepared by methods known in the art.
By "pharmaceutically acceptable base addition salt" is meant a salt formed with an inorganic or organic base that is capable of maintaining the bioavailability of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including natural substituted amines, cyclic amines and basic ion exchange resins such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
"polymorphs" refer to the different solid crystalline phases of certain compounds of the present invention in the solid state due to the presence of two or more different molecular arrangements. Certain compounds of the present invention may exist in more than one crystal form, and the present invention is intended to include various crystal forms and mixtures thereof.
In general, crystallization will produce solvates of the compounds of the present invention. The term "solvate" as used herein refers to an aggregate comprising one or more molecules of a compound of the invention and one or more solvent molecules. The solvent may be water, in which case the solvate is a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as hydrates, including monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may form true solvates, but in some cases may also retain only adventitious water or a mixture of water plus a portion of the adventitious solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized from a solvent. Solvates of the compounds of the present invention are also included within the scope of the present invention.
The invention also includes prodrugs of the above compounds. In this application, the term "prodrug" means a compound that can be converted into a biologically active compound of the invention under physiological conditions or by solvolysis. Thus, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention. Prodrugs may not be active when administered to an individual in need thereof, but are converted in vivo to the active compounds of the present invention. Prodrugs are typically rapidly converted in vivo to the parent compounds of the present invention, for example, by hydrolysis in blood. Prodrug compounds generally provide solubility, histocompatibility, or sustained release advantages in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups. Specific methods of prodrug preparation can be found in saunnier, m.g., et al, biorg. Med. Chem. Lett.1994,4,1985-1990; greenwald, r.b., et al, j.med.chem.2000,43,475.
In this application, "pharmaceutical composition" refers to a formulation of a compound of the invention with a medium commonly accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to promote the administration of organisms, facilitate the absorption of active ingredients and further exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a material (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention, and is relatively non-toxic, i.e., the material can be administered to an individual without causing an adverse biological reaction or interacting in an adverse manner with any of the components contained in the composition.
In this application, "pharmaceutically acceptable carrier" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonizing agent, solvent, or emulsifying agent that is approved by the relevant government regulatory agency as acceptable for human or livestock use.
The "tumor", "cell proliferation abnormality related disease", and the like of the present invention include, but are not limited to, leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, lung squamous carcinoma, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, and the like.
The terms "prevent", "preventing" and "preventing" as used herein include reducing the likelihood of a patient from developing or worsening a disease or condition.
The term "treatment" and other similar synonyms as used herein include the following meanings:
(i) Preventing the occurrence of a disease or disorder in a mammal, particularly when such mammal is susceptible to the disease or disorder, but has not been diagnosed as having the disease or disorder;
(ii) Inhibiting the disease or disorder, i.e., inhibiting its progression;
(iii) Alleviating a disease or condition, i.e., causing the state of the disease or condition to subside; or alternatively
(iv) Alleviating symptoms caused by the disease or condition.
The term "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein refers to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is required to provide clinically significant relief from a disorder. Effective amounts suitable in any individual case can be determined using techniques such as a dose escalation test.
The terms "administering," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, duodenal routes, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Application techniques useful in the compounds and methods described herein are well known to those skilled in the art, for example, at Goodman and Gilman, the Pharmacological Basis of Therapeutics, current ed.; pergamon; and Remington's, pharmaceutical Sciences (current edition), mack Publishing co., easton, pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "pharmaceutical combination", "co-administration", "administration of other treatments", "administration of other therapeutic agents" and the like as used herein refer to a pharmaceutical treatment obtained by mixing or combining more than one active ingredient, which includes both fixed and non-fixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one synergistic agent to a patient in the form of a single entity or single dosage form. The term "ambulatory combination" refers to the simultaneous administration, co-administration, or sequential administration of at least one compound described herein and at least one synergistic formulation as separate entities to a patient at variable intervals. These also apply to cocktail therapies, for example, administration of three or more active ingredients.
It will also be appreciated by those skilled in the art that in the methods described below, the intermediate compound functional groups may need to be protected by appropriate protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g., t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino groups include t-butoxycarbonyl, benzyloxycarbonyl and the like. Suitable mercapto protecting groups include-C (O) -R "(wherein" R "is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl, and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting groups is described in detail in Greene, t.w. and p.g.m. wuts, protective Groups in Organic Synthesis, (1999), 4th Ed. The protecting group may also be a polymeric resin.
The above preferred conditions can be arbitrarily combined on the basis of not deviating from the common knowledge in the art, and thus, each preferred embodiment of the present invention can be obtained.
The reagents and materials used in the present invention are commercially available.
The invention has the positive progress effects that: provides a nitrogen-containing heterocyclic compound which can be used as KRAS G12D An inhibitor; it can be used for preparing antitumor drugs, and preventing and/or treating tumor.
Detailed Description
The inventor prepares a class of nitrogen-containing heterocyclic compounds with novel structure shown in formula I through long-term and intensive research, and discovers that the nitrogen-containing heterocyclic compounds have better KRAS inhibition effect G12D Protein inhibitory Activity and said Compounds are useful in lower concentrations (as low as less than 500 nM), i.e. against KRAS G12D The associated cell proliferation and downstream signal pERK inhibitory activity were quite excellent (IC 50 Even less than 100 nM), and thus can be used in therapy with KRAS G12D Related diseases such as tumor caused by mutation or abnormal expression. Based on the above findings, the inventors have completed the present invention.
The invention is further illustrated by means of the following examples, which are not intended to limit the scope of the invention. The experimental methods, in which specific conditions are not noted in the following examples, were selected according to conventional methods and conditions, or according to the commercial specifications. The experimental methods in the following examples, in which specific conditions are not noted, are generally in accordance with conventional conditions, or in accordance with the conditions recommended by the manufacturer. Percentages and parts are weight percentages and parts unless otherwise indicated.
Preparation of intermediate A1: 4-hydrazine-2-naphthol pivalate
Figure BDA0003785353610000311
Step one: 4-bromo-2-naphthol (4.8 g,21.5 mmol) and triethylamine (2.6 g,25.8 mmol) were dissolved in dichloromethane (80 mL) and cooled in an ice-water bathNext, pivaloyl chloride (2.8 g,23.8 mmol) was added. The mixture was stirred at room temperature for 2 hours, the reaction mixture was poured into water (50 mL), the pH was adjusted to about 5, extracted twice with dichloromethane (50 mL), and the combined organic phases were concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=10:1) to give intermediate product (6.4 g) as colourless oil. LC-MS [ M-H ]] - :m/z 305.1。
Step two: the above intermediate compound (4.0 g,13.1 mmol), tert-butyl carbamate (1.8 g,15.7 mmol), cesium carbonate (6.0 g,18.3 mmol), palladium acetate (2918 mg,1.3 mmol), 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl (1.2 g,2.6 mmol) was dissolved in 1, 4-dioxane (30 mL) under nitrogen. The reaction was heated to 110℃and stirred overnight, concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=10:1) to give intermediate product (3.7 g) as a yellow solid. LC-MS [ M+H ]] + :m/z 288.1。
Step three: 1M hydrochloric acid/1, 4-dioxane solution (20 mL) in 1, 4-dioxane (20 mL) of the above intermediate compound (3.7 g,10.8 mmol). The reaction was stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to give a crude yellow solid (2.6 g). LC-MS [ M+H ] ] + :m/z 244.1。
Step four: the intermediate compound (2.6 g,10.8 mmol) was added to concentrated hydrochloric acid/water (8 mL/48 mL). Sodium nitrite (1.5 mg,21.6 mmol) was added to the reaction solution under ice-bath cooling. After the completion of the dropwise addition, the reaction was continued for 30 minutes. Tin dichloride (6.1 g,32.4 mmol) was then added under ice bath and the reaction stirred for an additional 4 hours. LC-MS detection reaction was complete, the reaction solution was concentrated under reduced pressure, and the obtained crude product was prepared by reverse HPLC to obtain a yellow target product (580 mg). LC-MS [ M+H ]] + :m/z 259.3。
Referring to the synthetic route and method of patent WO2021041671A1, the following intermediate compounds B1-B6 were prepared;
Figure BDA0003785353610000321
Figure BDA0003785353610000331
referring to the synthetic route and process of patent WO2021106231A1, the following intermediate compounds A2-A4 were prepared;
Figure BDA0003785353610000332
example general preparation method 1
Figure BDA0003785353610000333
The first step: intermediate compound a (1 eq.) was dissolved in an appropriate solvent, and a substituted aryl or heteroaryl hydrazine (1 eq.) was added and the reaction stirred at room temperature for several hours. After the LC-MS detection reaction is completed, purifying and drying after post-treatment to obtain a target product, and confirming a structure by adopting nuclear magnetism and mass spectrum.
And a second step of: intermediate compound B (1 eq.) is dissolved in a suitable solvent and amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (2.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a third step of: intermediate compound C (1 eq.) was dissolved in an appropriate solvent, organic base (1.5 eq.) and palladium transition metal catalyst (0.15 eq.) were added and reacted for several hours with heating in a carbon monoxide closed reactor at 20 atmospheres. The LC-MS detection reaction was substantially complete. The reaction liquid is filtered by diatomite, washed by organic solvent, and after the filtrate is decompressed and concentrated, the crude product is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
Fourth step: intermediate compound D (1 eq.) was dissolved in an organic acid and reacted for several minutes with heating. The LC-MS detection reaction was essentially complete. The reaction solution was concentrated under reduced pressure, the residue was adjusted to pH with aqueous sodium carbonate to basicity, and extracted with ethyl acetate. The combined organic phases are concentrated under reduced pressure, the obtained crude product is separated and purified to obtain the target product, and the structure is confirmed by adopting nuclear magnetism and mass spectrum.
Fifth step: intermediate compound E (1 eq.) was dissolved in an appropriate solvent, peroxide (1.5 eq.) was added and stirred at room temperature for several hours. After the LC-MS detection reaction was substantially complete, the reaction was quenched by addition of an aqueous solution of sodium thiosulfate. Extracting the reaction liquid with ethyl acetate, concentrating the combined organic phases under reduced pressure, separating and purifying the obtained crude product to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
Sixth step: by reacting amines or alcohols as intermediates R 2 L-H (1 eq.) is dissolved in a suitable solvent, an inorganic base (2 eq.) is added with low temperature cooling, and after stirring for half an hour, the intermediate of the above first step product formula (F) is added. The reaction was then stirred at room temperature overnight. TLC monitors the reaction completely, after adding water for quenching, the reaction is concentrated under reduced pressure, the remainder is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound, and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Example general preparation method 2
Figure BDA0003785353610000341
The first step: the intermediate compound A (1 eq.) is dissolved in a suitable solvent and an amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (2.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a second step of: intermediate compound B (1 eq.) was dissolved in an appropriate solvent, and an organic base (1.5 eq.) and a palladium transition metal catalyst (0.2 eq.) were added and reacted for several hours at room temperature in a carbon monoxide closed reactor at 20 atmospheres. The LC-MS detection reaction was substantially complete. The reaction liquid is filtered by diatomite, washed by organic solvent, and after the filtrate is decompressed and concentrated, the crude product is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a third step of: intermediate compound C (1 eq.) was dissolved in an appropriate solvent, peroxide (1.5 eq.) was added and stirred at room temperature for several hours. After the LC-MS detection reaction was substantially complete, the reaction was quenched by addition of an aqueous solution of sodium thiosulfate. Extracting the reaction liquid with ethyl acetate, concentrating the combined organic phases under reduced pressure, separating and purifying the obtained crude product to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
Fourth step: by reacting amines or alcohols as intermediates R 2 -L-H (1 eq.) is dissolved in a suitable solvent, the intermediate of general formula (D) of the product of the previous step (1.2 eq.) and the organic base (3 eq.) are added and heated with stirring for several hours. TLC monitoring reaction is complete, decompression concentration is carried out, the residue is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound, and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Fifth step: the intermediate compound E (1 eq.) was dissolved in an organic acid and reacted by heating for several minutes. The LC-MS detection reaction was essentially complete. The reaction solution was concentrated under reduced pressure, the residue was adjusted to pH with aqueous sodium carbonate to basicity, and extracted with ethyl acetate. The combined organic phases are concentrated under reduced pressure, the obtained crude product is separated and purified to obtain the target product, and the structure is confirmed by adopting nuclear magnetism and mass spectrum.
Sixth step: the above intermediate compound F (1 eq.) was dissolved in an appropriate solvent, and a substituted arylboronic acid (ester) (1.2 eq.) was added and the ketone acetate (0.5 eq.) was reacted at room temperature overnight. Or the above intermediate compound F (1 eq.) is dissolved in an appropriate solvent, and the substituted aryl halide (1 eq.) and the transition metal Pd complex (0.2 eq.) are added and reacted under nitrogen with heating for several hours. The LC-MS detection reaction was essentially complete. Concentrating the reaction liquid under reduced pressure, separating and purifying the obtained crude product to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
Example general preparation method 3
Figure BDA0003785353610000351
/>
The first step: intermediate compound a (1 eq.) is dissolved in a suitable solvent and amine is addedOr an alcohol intermediate R 1 -H (1.05 eq.) and organic base (2 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a second step of: intermediate compound B (1 eq.) was dissolved in an appropriate solvent, and an inorganic base (1.5 eq.) and a palladium transition metal catalyst (0.2 eq.) were added, and the phosphine ligand (0.2 eq.) was heated for several hours. The LC-MS detection reaction was substantially complete. The reaction liquid is filtered by diatomite, washed by organic solvent, and after the filtrate is decompressed and concentrated, the crude product is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a third step of: intermediate compound C (1 eq.) was dissolved in an appropriate solvent and palladium on carbon (0.1 eq.) was stirred at room temperature under hydrogen atmosphere for several hours. After the LC-MS detection reaction was substantially complete, the celite was filtered. Washing with organic solvent, concentrating the combined organic phases under reduced pressure, separating and purifying the obtained crude product to obtain the target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
Fourth step: intermediate compound D (1 eq.) was dissolved in an appropriate solvent, substituted aryl boronic acid (ester) (1.2 eq.) was added and the ketone acetate (0.5 eq.) was reacted overnight at room temperature. Or the above intermediate compound F (1 eq.) is dissolved in an appropriate solvent, and the substituted aryl halide (1 eq.) and the transition metal Pd complex (0.2 eq.) are added and reacted under nitrogen with heating for several hours. The LC-MS detection reaction was essentially complete. Concentrating the reaction liquid under reduced pressure, separating and purifying the obtained crude product to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
Example general preparation method 4
Figure BDA0003785353610000361
The first step: intermediate compound a (1 eq.) was dissolved in an appropriate solvent, sodium hydrogen (2 eq.) was added to the reaction solution with cooling in an ice water bath, and the reaction was stirred for one hour. CDI (1.5 eq.) was added to the reaction solution, which was slowly raised to 80 degrees and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a second step of: the intermediate compound B (1 eq.) was dissolved in an appropriate solvent, and phosphorus oxychloride (10 eq.) was added to the reaction solution. The reaction solution was heated to 80℃and stirred for four hours. After completion of the LC-MS detection reaction, the solvent was removed by concentration under reduced pressure, and the crude product was used directly in the next step.
And a third step of: intermediate compound C (1 eq.) is dissolved in a suitable solvent and amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (2.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
Fourth step: by reacting amines or alcohols as intermediates R 2 -L-H (1 eq.) is dissolved in a suitable solvent, the intermediate of general formula (D) of the product of the previous step (1.2 eq.) and the organic base (3 eq.) are added and heated with stirring for several hours. TLC monitoring reaction is complete, decompression concentration is carried out, the residue is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound (I), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Example general preparation method 5
Figure BDA0003785353610000362
The first step: intermediate compound A (1.0 eq.) is dissolved in a suitable solvent and amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (2.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitoring reaction completion, adding water into the reaction solution, extracting the water phase with ethyl acetate for three times, drying the extract with anhydrous sodium sulfate, concentrating under reduced pressure, separating and purifying the residue to obtain the target product, and confirming the structure by adopting nuclear magnetism and mass spectrum 。
And a second step of: by reacting amines or alcohols as intermediates R 2 -L-H (1 eq.) is dissolved in a suitable solvent, the intermediate of general formula (B) of the product of the previous step (1.2 eq.) and the organic base (3 eq.) are added and heated with stirring for several hours. TLC monitoring reaction is complete, decompression concentration is carried out, the residue is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound (C), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
And a third step of: the intermediate of general formula (C) (1.0 eq.) is dissolved in a suitable solvent, and an inorganic base (5 eq.) is added and stirred for several hours. TLC monitoring reaction is complete, the reaction solution is concentrated under reduced pressure, the residue is prepared and purified by silica gel column chromatography or HPLC to obtain the target compound (D), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Fourth step: the intermediate of general formula (C) (1.0 eq.) is dissolved in a suitable solvent, and an inorganic base (5 eq.) is added and stirred for several hours. TLC monitors that the reaction is complete, dilute acid adjusts PH to neutral, then decompresses and concentrates the reaction liquid, the remainder is prepared and purified by silica gel column chromatography or HPLC to obtain the target compound (D), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Fifth step: the intermediate of general formula (D) (1.0 eq.) is dissolved in a suitable solvent, an organic base (2 eq.) and a suitable condensing agent (1.5 eq.) are added and the reaction is stirred for several hours. TLC monitoring reaction is complete, the reaction solution is concentrated under reduced pressure, the residue is prepared and purified by silica gel column chromatography or HPLC to obtain the target compound (E), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Sixth step: the intermediate of general formula (E) (1.0 eq.) is dissolved in a suitable solvent, a reducing agent (e.g. tin dichloride, palladium on carbon/hydrogen etc.) (3 eq.) is added and the reaction is heated and stirred for several hours. TLC monitoring reaction is complete, the reaction solution is filtered, the filtrate is concentrated under reduced pressure, and the residue is prepared and purified by silica gel column chromatography or HPLC to obtain the target compound (F), and the structure is confirmed by nuclear magnetism and mass spectrum.
Sixth step: the intermediate of general formula (F) (1.0 eq.) is dissolved in a suitable solvent, trimethyl orthoacetate or trifluoroacetic anhydride (50 eq.) and methanesulfonic acid (2 eq.) are added and the reaction is heated and stirred for several hours. TLC monitoring reaction is complete, the reaction solution is filtered, the filtrate is concentrated under reduced pressure, and the residue is prepared and purified by silica gel column chromatography or HPLC to obtain the target compound (I), and the structure is confirmed by nuclear magnetism and mass spectrum.
Example general preparation method 6
Figure BDA0003785353610000371
The first step: intermediate compound A (1.0 eq.) is dissolved in a suitable solvent and amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (2.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a second step of: by reacting amines or alcohols as intermediates R 2 -L-H (1 eq.) is dissolved in a suitable solvent, the intermediate of general formula (B) of the product of the previous step (1.2 eq.) and the organic base (3 eq.) are added and heated with stirring for several hours. TLC monitoring reaction is complete, decompression concentration is carried out, the residue is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound (C), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
And a third step of: the intermediate of general formula (C) (1 eq.) of the product of the previous step was dissolved in a suitable solvent, and after addition of NBS (1.1 eq.) the reaction was heated and stirred for several hours. LC-MS detection reaction is complete, the reaction solution is concentrated under reduced pressure, the remainder is prepared, separated and purified by silica gel column chromatography or HPLC to obtain a target product (D), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Fourth step: to a suitable solvent for the intermediate of general formula (D) and the aromatic borate ester (1 eq.) of the previous step product under nitrogen protection, an inorganic base (2 eq.) and a transition metal palladium catalyst (0.3 eq.) were added. The reaction mixture was heated and stirred for several hours. TLC monitoring reaction is complete, decompression concentration is carried out, the residue is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound (I), and nuclear magnetism and mass spectrum are adopted to confirm the structure.
Example general preparation method 7
Figure BDA0003785353610000381
The first step: the intermediate compound A (1 eq.) is dissolved in a suitable solvent and an amine or alcohol intermediate R is added 1 -H (1.05 eq.) and organic base (3.5 eq.). The reaction was slowly warmed to room temperature and stirred overnight. LC-MS monitors the reaction completely, water is added into the reaction solution, the water phase is extracted three times by ethyl acetate, the extract is dried by anhydrous sodium sulfate, the concentration is reduced, the remainder is separated and purified to obtain the target product, and the structure is confirmed by nuclear magnetism and mass spectrum.
And a second step of: by reacting amines or alcohols as intermediates R 2 Y-H (1 eq.) is dissolved in a suitable solvent, an inorganic base (2 eq.) is added with low temperature cooling, and after stirring for half an hour, the intermediate of the above-mentioned first step product formula (B) is added. The reaction was then stirred at room temperature overnight. TLC monitors the reaction completely, after adding water for quenching, the reaction is concentrated under reduced pressure, the remainder is prepared and separated and purified by silica gel column chromatography or HPLC to obtain the target compound, and nuclear magnetism and mass spectrum are adopted to confirm the structure.
And a third step of: under nitrogen protection, the above general formula product C (1 eq.) and arylboronic acid (ester) or arylmetal reagent (1.2 eq.) are dissolved in a suitable solvent, a transition metal catalyst (0.1 eq.) and an inorganic base (2 eq.) are added, heated to 80-100 degrees for several hours of reaction, cooled to room temperature, the reaction solution is poured into water and extracted with ethyl acetate. The organic phase is washed by saturated saline, and after concentration, the crude product is prepared, separated and purified by silica gel column chromatography or HPLC to obtain the target compound, and the structure is confirmed by nuclear magnetism and/or mass spectrum.
Example 1:4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (3-hydroxynaphthalen-1-yl) -2- ((hexahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) pyrimidine [4,5-d ] pyridazin-8 (7H) -one
Figure BDA0003785353610000391
The first step: 4-hydrazinonaphthalene-2-pivaloyl phenol ester (580 mg,2.2 mmol) was dissolved in DMF (3 mL) under nitrogen, 4, 6-dichloro-2- (methylthio) pyrimidine-5-carbonyl (501 mg,2.2 mmol) was stirred at room temperatureOvernight. To the reaction mixture was added a saturated sodium bicarbonate solution (10 mL), and a solid was precipitated. The solid was filtered off, washed and dried to give the intermediate product as a yellow solid (710 mg). LC-MS [ M+H ]] + :m/z 463.2。
And a second step of: the intermediate compound (710 mg,1.5 mmol) was reacted under nitrogen with 3, 8-diazabicyclo [3.2.1]Tert-butyl octane-8-carboxylate (326 mg,1.5 mmol), N, N-diisopropylethylamine (400 mg,3.1 mmol) was dissolved in DMF (3 mL) and stirred overnight at room temperature. The reaction mixture was diluted with water (30 mL), extracted three times with ethyl acetate (30 mL), and the combined organic phases were dried and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=5:1) to give intermediate product (875 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 639.3。
And a third step of: the above intermediate compound (875 mg,1.3 mmol) was reacted with [1,1' -bis (diphenylphosphino) ferrocene ]Palladium dichloride (73 mg,0.1 mmol), triethylamine (400 mg,4.0 mmol) were dissolved in methanol (10 mL) and heated to 100℃overnight under carbon monoxide (1 MPa). The reaction solution was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=4:1) to obtain an intermediate product (250 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 663.3。
Fourth step: the intermediate compound (250 mg,0.4 mmol) was dissolved in acetic acid (10 mL) and reacted at 110℃for 30 minutes. The solution was neutralized with saturated sodium bicarbonate, adjusted to pH 8, extracted three times with ethyl acetate (30 mL) and the combined organic phases dried and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=2:1) to give intermediate product (100 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 631.3。
Fifth step: the above intermediate compound (100 mg,0.16 mmol) was dissolved in methylene chloride (20 mL), and potassium peroxomonosulphonate (74 mg,0.24 mmol) was added thereto under ice-bath cooling, and the reaction was carried out at room temperature for 30 minutes, whereby the reaction of the starting materials was examined to be substantially complete. Saturated aqueous sodium thiosulfate (10 mL) was added and stirred for 10 min. After separation of the organic phase, the aqueous phase was extracted twice with dichloromethane (20 mL). The combined organic phases were dried and the crude product was concentrated under reduced pressure to give a yellow solid intermediate product (30) by HPLC mg)。LC-MS[M+H] + :m/z 647.3。
Sixth step: the intermediate product (40 mg,0.06 mmol) and DIEA were added to 1, 4-dioxane (10 mL), and after addition of (hexahydro-1H-pyrrolin-7A-yl) methanol (71 mg,0.50 mmol), heated to 80℃and stirred for 5 hours. After completion of the detection reaction, the reaction mixture was extracted three times with ethyl acetate (50 mL), and the combined organic phases were dried over anhydrous magnesium sulfate and filtered. The crude product after concentration of the filtrate under reduced pressure was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30:1) to give a yellow solid product (30 mg). LC-MS [ M+H ]] + :m/z 724.4。
Seventh step: to a solution of the intermediate (30 mg,0.04 mmol) in Dichloromethane (DCM) (6 mL) was added trifluoroacetic acid (3 mL) under ice-bath cooling, and the reaction was warmed to room temperature and stirred for 4 hours. After concentrating under reduced pressure to remove most of the solvent, tetrahydrofuran (3 mL) was added. Then, ammonia (3 mL) was added thereto while cooling in an ice bath, and the mixture was stirred at room temperature for 10 minutes. The mixture was concentrated under reduced pressure, and the residue was purified by HPLC to give the product (5 mg) as a pale yellow solid. LC-MS m/z 540.3[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δ8.35(s,1H),7.77(d,J=8.4Hz,1H),7.53(d,J=8.4Hz,1H),7.46-7.42(m,1H),7.31(d,J=2.0Hz,1H),7.22-7.26(m,2H),4.91(m,2H),4.70(s,2H),4.27(s,2H),3.98(m,2H),3.73-3.63(m,2H),2.37-1.89(m,13H).
Starting with intermediates A1, A2 and commercially available piperazine derivatives, the following example compounds 2-5 were obtained by analogy with reference to example 1 using the general preparation of the examples:
Figure BDA0003785353610000401
Figure BDA0003785353610000411
Example 6:4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-ethynyl-3-hydroxynaphthalen-1-yl) -2- ((hexahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) pyrimidine [4,5-d ] pyridazin-8 (7H) -one
Figure BDA0003785353610000412
The first step: 4, 6-dichloro-2- (methylthio) pyrimidine-5-carbonyl (850 mg,3.8 mmol) was dissolved in DMF (10 mL) under nitrogen, 3, 8-diazabicyclo [3.2.1] was added]Tert-butyl octane-8-carboxylate (720 mg,3.8 mmol) and N, N-diisopropylethylamine (600 mg,4.6 mmol) were stirred overnight at room temperature. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed three times with ethyl acetate (30 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=4:1) to give intermediate product (1.3 g) as a white solid. LC-MS [ M+H ]] + :m/z 495.3。
And a second step of: the above intermediate compound (450 mg,0.9 mmol) was reacted with [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (146 mg,0.2 mmol), triethylamine (400 mg,4.0 mmol) were dissolved in methanol (10 mL), and stirred overnight at room temperature under carbon monoxide (1 MPa). The reaction solution was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=4:1) to obtain an intermediate product (240 mg) as a yellow solid. LC-MS [ M+H ] ] + :m/z 423.2。
And a third step of: the above intermediate compound (200 mg,0.47 mmol) was dissolved in dichloromethane (20 mL) and potassium peroxymonosulphonate (204 mg,0.66 mmol) was added under ice-bath cooling. The reaction was carried out at room temperature for 40 minutes, and the detection of the starting material was substantially complete. Saturated aqueous sodium thiosulfate (10 mL) was added and stirred for 20 minutes. After separation of the organic phase, the aqueous phase was extracted twice with dichloromethane (50 mL). The combined organic phases were dried and the resulting crude product was concentrated under reduced pressure to afford the intermediate product (70 mg) as a yellow solid by HPLC. LC-MS [ M+H ]] + :m/z 439.2。
Fourth step: the intermediate product (70 mg,0.16 mmol) and DIEA were added to 1, 4-dioxane (10 mL), and after addition of (hexahydro-1H-pyrrolin-7A-yl) methanol (45 mg,0.32 mmol), heated to 80℃and stirred for 5 hours. After completion of the detection reaction, the reaction mixture was extracted three times with ethyl acetate (50 mL) and the combined organic phases were taken to be freeDrying with magnesium sulfate and filtering. The crude product after concentration of the filtrate under reduced pressure was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30:1) to give a yellow solid product (80 mg). LC-MS [ M+H ]] + :m/z 516.3。
Fifth step: the intermediate compound (80 mg,0.15 mmol) was dissolved in ethanol (20 mL), and hydrazine hydrate (2 mL) and glacial acetic acid (1 mL) were added and heated to 80℃to react for 3 hours. After the reaction solution was concentrated under reduced pressure to remove most of the solvent, 30mL of water was added, followed by extraction with ethyl acetate (30 mL) three times. The combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by HPLC to give the intermediate product as a yellow solid (40 mg). LC-MS [ M+H ] ] + :m/z 498.3。
Sixth step: to a solution of the above intermediate (40 mg,0.08 mmol) in 1, 4-dioxane/water (10 mL/2 mL) was added at room temperature boric acid ester B2 (40 mg,0.08 mmol), copper acetate (15 mg,0.08 mmol) and pyridine (8 mg,0.1 mmol), and the reaction mixture was stirred at room temperature overnight. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the crude product was purified by flash column chromatography on silica gel to give a yellow solid product (30 mg). LC-MS [ M+H ]] + :m/z 864.5。
Seventh step: to dichloromethane (10 mL) of the above intermediate (35 mg,0.04 mmol) was added trifluoroacetic acid (2 mL) at room temperature, and the reaction mixture was stirred at room temperature for 30 minutes. The reaction solution was concentrated under reduced pressure, and the concentrated crude product was dissolved in tetrahydrofuran (5 mL), cesium fluoride CsF (30 mg,0.2 mmol) was added to the reaction solution, and stirring was continued for 2 hours. Concentrated under reduced pressure, and the crude product was purified by HPLC to give a yellow solid product (7 mg). LC-MS [ M+H ]] + :m/z 564.3。 1 H NMR(400MHz,CD 3 OD)δ8.42(s,1H),7.85(m,1H),7.51(m,1H),7.42-7.34(m,2H),7.16(m,1H),4.72-4.61(m,4H),4.06(s,2H),3.90-3.65(m,4H),3.26-3.02(m,3H),2.33-1.85(m,12H).
The following example compounds were obtained by following similar procedures with reference to example 6 using the general preparation methods of the examples, starting from intermediates B1 to B6, A2 to A4 and commercially available piperazine derivatives and other reagents:
Figure BDA0003785353610000421
/>
Figure BDA0003785353610000431
/>
Figure BDA0003785353610000441
/>
Figure BDA0003785353610000451
example 26:4- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) naphthalen-2-ol
Figure BDA0003785353610000452
The first step: under the protection of nitrogen, 7-benzyl-2, 4-dichloro-5, 6,7, 8-tetrahydropyrido [3,4-d ]]Pyrimidine (2.0 g,6.8 mmol) was dissolved in N, N-dimethylformamide DMF (20 mL) and 3, 8-diazabicyclo [3.2.1 ] was added]Tert-butyl octane-8-carboxylate (2.4 g,6.8 mmol) and N, N-diisopropylethylamine (2.6 g,20.4 mmol) were stirred overnight at room temperature. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=10:1) to give intermediate product (3.5 g) as a white solid. LC-MS [ M+H ]] + :m/z 470.2。
And a second step of: the intermediate compound (1.0 g,2.1 mmol) and (hexahydro-1H-pyrrolin-7A-yl) methanol (750 mg,4.3 mmol) were dissolved in toluene (30 mL) under nitrogen, cesium carbonate (2.08 g,9.6 mmol) was added, pd 2 (dba) 3 (195 mg,0.21 mmol) and Ruphos (199mg, 0.43 mmol). The reaction was heated to 110℃and stirred overnight. LC-MS detectionThe reaction was found to be substantially complete, water (30 mL) was added to the reaction mixture, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:20) to give intermediate product (800 mg) as a yellow solid. LC-MS [ M+H ] ] + :m/z 575.3。
And a third step of: to t-butanol (15 mL) of the above intermediate compound (189 mg,0.33 mmol) was added wet palladium on carbon (30 mg) under a hydrogen atmosphere. The reaction was stirred at room temperature for 48 hours. The LC-MS detection reaction is basically complete, and the reaction solution is filtered through kieselguhr and washed by ethanol. The filtrate was concentrated under reduced pressure to give an intermediate product (150 mg) as a pale yellow solid. LC-MS [ M+H ]] + :m/z 485.3。
Fourth step: the intermediate compound (120 mg,0.25 mmol) and 1-bromo-3- ((4-methoxybenzyl) oxy) naphthalene (120 mg,0.35 mmol) were dissolved in toluene (30 mL) under nitrogen, cesium carbonate (163 mg,0.5 mmol) and Pd were added 2 (dba) 3 (34 mg,0.04 mmol) and 2-dicyclohexylphosphine-2 ',6' -diisopropyloxybiphenyl Ruphos (23 mg,0.05 mmol). The reaction was heated to 80℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (21 mg). LC-MS [ M+H ]] + :m/z 671.1。
Fifth step: the intermediate compound (21 mg,0.03 mmol) was dissolved in THF (10 mL) under nitrogen and TFA (1 mL) was added. Stirring for 30 minutes at room temperature. The LC-MS detection reaction was substantially complete, and to the reaction mixture was added saturated aqueous sodium bicarbonate (10 mL) and extracted twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (5 mg). LC-MS [ M+H ] ] + :m/z 527.3。 1 H NMR(400MHz,DMSO-d 6 ):δ10.05(s,1H),9.68(s,1H),8.96-8.93(m,1H),8.83-8.80(m,1H),7.98(d,J=8.4Hz,1H),7.68(d,J=8.0Hz,1H),7.42-7.38(m,1H),7.29-7.25(m,1H),6.87(s,1H),6.76(s,1H),4.40(s,2H),4.15-4.04(m,6H),3.46-3.27(m,6H),2.97-2.93(m,2H),2.04-1.98(m,12H)。
Starting with intermediates B1 to B5 and commercially available reagents, the following example compounds were obtained by the general preparation method of the examples, in analogy with reference to example 26:
Figure BDA0003785353610000471
/>
Figure BDA0003785353610000481
example 34:4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-ethyl-3-hydroxynaphthalen-1-yl) -2- ((tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridine-3-cyano
Figure BDA0003785353610000482
The first step: sodium metal (Na) beads (3.2 g,139.1 mmol) were added to EtOH (250 mL) in portions and stirred at room temperature until the beads completely disappeared, ethyl 5-amino-1-benzyl-1, 2,3, 6-tetrahydropyridine-4-carboxylate (18.0 g,69.2 mmol) and ethyl cyanoacetate (15.7 g,138.9 mmol) were added and the pot heated to 120℃for three days. After cooling, filtration was carried out, and the cake was purified by beating with absolute ethanol, and after concentration and drying, a yellow solid intermediate compound (15.4 g) was obtained. LC-MS ESI [ M+H ]] + =282.3。
And a second step of: POCl3 (5 mL) was added to a solution of the above intermediate compound (5.0 g,17.8 mmol) in acetonitrile (20 mL), the reaction mixture was heated to 60℃and stirred for 2 hours, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 6:1) to give a yellow solid intermediate compound (3.1 g). LC-MS ESI [ M+H ] ] + =318.2。
And a third step of: the above intermediate compound (1.05 g,3.3 mmol) was dissolved in 1, 4-dioxane (30 mL) and DIPEA (0.97 g,7.6 mmol) and 3, 8-diazabicyclo were added[3.2.1]Octane-8-carboxylic acid tert-butyl ester (0.7 g,3.3 mmol). The mixture was warmed to 80℃and stirred overnight, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2:1) to give the intermediate compound (1.2 g) as a yellow solid. LC-MS ESI [ M+H ]] + =494.2。
Fourth step: naH (1.04 g,4.8 mmol) was added to a solution of (hexahydro-1H-pyrrolin-7A-yl) methanol (250 mg,1.4 mmol) in DMF (20 mL) at room temperature under nitrogen, and after stirring for 10 min the intermediate compound of the previous step (0.6 g,1.2 mmol) was added. The reaction was heated to 110℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:20) to give an intermediate product (300 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 599.4。
Fifth step: to t-butanol (15 mL) of the above intermediate compound (300 mg,0.5 mmol) was added wet palladium on carbon (30 mg) under a hydrogen atmosphere. The reaction was stirred at room temperature for 48 hours. The LC-MS detection reaction is basically complete, and the reaction solution is filtered through kieselguhr and washed by ethanol. The filtrate was concentrated under reduced pressure to give an intermediate product (190 mg) as a pale yellow solid. LC-MS [ M+H ] ] + :m/z 509.3。
Sixth step: to t-butanol (15 mL) of the above intermediate compound (300 mg,0.5 mmol) was added wet palladium on carbon (30 mg) under a hydrogen atmosphere. The reaction was stirred at room temperature for 48 hours. The LC-MS detection reaction is basically complete, and the reaction solution is filtered through kieselguhr and washed by ethanol. The filtrate was concentrated under reduced pressure to give an intermediate product (190 mg) as a pale yellow solid. LC-MS [ M+H ]] + :m/z 509.3。
Seventh step: the above intermediate compound (180 mg,0.35 mmol) and 8-ethyl-3- (methoxymethoxy) naphthalen-1-yl) -trifluoromethanesulfonate (180 mg,0.5 mmol) were dissolved in toluene (30 mL) under nitrogen, cesium carbonate (326 mg,1.0 mmol), pd was added 2 (dba) 3 (70 mg,0.1 mmol) and 2-dicyclohexylphosphine-2 ',6' -diisopropyloxybiphenyl Ruphos (47 mg,0.1 mmol). The reaction was heated to 80℃and stirred overnight. The LC-MS detection reaction is substantially complete, directed toWater (30 mL) was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (32 mg). LC-MS [ M+H ]] + :m/z 723.4。
Eighth step: the intermediate compound (32 mg,0.04 mmol) was dissolved in dichloromethane (10 mL) under nitrogen and TFA (1 mL) was added. Stirring for 30 minutes at room temperature. The LC-MS detection reaction was substantially complete, and to the reaction mixture was added saturated aqueous sodium bicarbonate (10 mL) and extracted twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (11 mg). LC-MS [ M+H ] ] + :m/z 579.3。 1 H NMR(400MHz,DMSO-d 6 ):δ10.3(s,1H),9.67(s,1H),8.96-8.93(m,1H),8.83-8.80(m,1H),7.98(d,J=8.4Hz,1H),7.68(d,J=8.0Hz,1H),7.43-7.32(m,1H),6.93(s,1H),6.79(s,1H),4.40(s,2H),4.19-3.93(m,6H),3.46-2.97(m,8H),2.04-1.98(m,12H)1.35-1.31(m,3H)。
Starting with intermediates B1 to B5 and commercially available reagents, the following example compounds were obtained in analogy to example 34 using the general preparation methods of the examples:
Figure BDA0003785353610000501
/>
Figure BDA0003785353610000511
example 42:4- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -7-fluorobenzothiazol-2-amine
Figure BDA0003785353610000512
To (4-bromo-7-fluorobenzothiazide)Azole-2-amino) formyl tert-butyl ester was used as a starting material in place of 1-bromo-3- ((4-methoxybenzyl) oxy) naphthalene, and the compound of example 42 was obtained as a white solid by the method of reference example 26. LC-MS [ M+H ]] + :m/z 551.7。
Example 43:4- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2 r,7 as) -2-fluoro-tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -7-fluorobenzothiazol-2-amine
Figure BDA0003785353610000513
The procedure of example 26 was followed using (4-bromo-7-fluorobenzothiazol-2-ylamino) formyl tert-butyl ester as starting material in place of 1-bromo-3- ((4-methoxybenzyl) oxy) naphthalene to give the compound of example 43 as a white solid. LC-MS [ M+H ]] + :m/z 569.7。
Example 44:7- (2-amino-7-fluorobenzothiazol-4-yl) -4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((hexahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) pyrimidine [4,5-d ] pyridazin-8 (7H) -one
Figure BDA0003785353610000514
The procedure of example 6 was referenced to give the compound of example 44 as a white solid, starting from (2- ((tert-butyloxycarbonyl) amino) -7-fluorobenzothiazol-4-yl) boronic acid instead of the boronic ester B2. LC-MS [ M+H ]] + :m/z 564.7。
Example 45:7- (2-amino-7-fluorobenzothiazol-4-yl) -4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -6- (trifluoromethyl) pyrido [3,4-d ] pyrimidin-8 (7H) -one
Figure BDA0003785353610000521
The first step: tert-butyl (4-amino-7-fluorobenzothiazol-2-yl) carbonate (2.83 g,1 mmol) and carbonic acid were added to the mixture3.45g,2.5 mmol) was dissolved in acetonitrile (50 mL) and monomethyl malonate acyl chloride (2.0 g,1.5 mmol) was added at 0 ℃. The reaction mixture was stirred at room temperature for 12 hours. LCMS showed complete reaction. The reaction was quenched by addition of water (100 mL), extracted three times with ethyl acetate (100 mL), the organic phases combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. And (5) pulping and purifying the concentrated crude product, filtering, and drying a filter cake under reduced pressure. Crude intermediate compound (2.6 g) was obtained as white. LCMS (ESI) m/z 384.4[ M+H ]] -
And a second step of: the above intermediate compound (2.6 g,6.78 mmol) was dissolved in methanol (80 mL), and 4-ethyl-1, 1-trifluoro-3-buten-2-one (1.63 g,9.7 mmol) and sodium methoxide (0.54 g,10 mmol) were added. The reaction was stirred at 90 degrees for 40 hours. LCMS showed complete reaction and product formation was detected. The reaction solution was concentrated under reduced pressure, a saturated aqueous ammonium chloride solution (100 mL) was added, and extraction was performed twice with ethyl acetate (100 mL). The organic phases were combined and concentrated under reduced pressure to give crude compound (2.9 g) as a white solid. LCMS (ESI) m/z 488.4[ M+1 ] ] +
And a third step of: the above intermediate compound (2.7 g,5.54 mmol) was dissolved in tetrahydrofuran (80 mL), and lithium hydroxide monohydrate (0.42 g,10 mmol) was added. The reaction was stirred at room temperature for 12 hours. LCMS showed complete reaction and product formation was detected. The reaction was concentrated under reduced pressure, and then pH was adjusted to 2-3 by adding 1M dilute aqueous hydrochloric acid, followed by extraction three times with ethyl acetate (100 mL). The organic phases were combined and concentrated under reduced pressure to give crude compound (2.5 g) as a white solid. LCMS (ESI) m/z 474.4[ M+1 ]] +
Fourth step: the above intermediate compound (2.5 g,5.28 mmol) was dissolved in toluene (50 mL), and triethylamine (1.12 g,11 mmol), benzyl alcohol (1 mL,10 mmol) and DPPA (1.49 g,5.4 mmol) were added. The reaction was stirred at 90 degrees for 3 hours. LCMS showed complete reaction and product formation was detected. The reaction solution was concentrated under reduced pressure, a saturated aqueous ammonium chloride solution (30 mL) was added, and extraction was performed 2 times with ethyl acetate (50 mL). The organic phases were combined and concentrated under reduced pressure to give crude compound (1.8 g) as a white solid. LCMS (ESI) m/z 579.5[ M+1 ]] +
Fifth step: the intermediate compound (1.8 g,3.11 mmol) was dissolved in methanol (30 mL) and Pd/C (5% wt,0.75g,0.35 mmol) was addedStirring was carried out at room temperature under a hydrogen (1 atm) atmosphere for 3 hours. LCMS showed complete reaction and product formation was detected. The reaction mixture was concentrated under reduced pressure to give crude compound (1.3 g) as a yellow solid. LCMS (ESI) m/z 445.4[ M+1 ] ] +
Sixth step: the intermediate compound (1.3 g,2.93 mmol) was dissolved in methylene chloride (30 mL), NBS (0.49 g,2.75 mmol) was added, and the mixture was stirred at room temperature for 1 hour. LCMS showed complete reaction. To the reaction mixture was added saturated brine (50 mL), and the separated organic phase was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 3:1) to obtain crude compound (1.4 g) as a yellow solid. LCMS (ESI) m/z 524.3[ M+1 ]] +
Seventh step: the above intermediate compound (1.4 g,2.67 mmol) was dissolved in N, N-dimethylacetamide (30 mL) under nitrogen, and bis-dibenzylideneacetone palladium (0.43 g,0.75 mmol), 1' -bis-diphenylphosphino ferrocene (0.69 g,1.24 mmol) and zinc cyanide (0.92 g,7.83 mmol) were added. The reaction mixture was heated to 125℃and reacted for 15 hours. LCMS showed complete reaction. The reaction solution was filtered, and to the filtrate, saturated brine (100 mL) was added, followed by extraction with ethyl acetate (50 mL) three times, and the combined organic phases were concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 4:1) to give a crude compound (920 mg) as a yellow solid. LCMS (ESI) m/z 470.4[ M+1 ]] +
Eighth step: to a solution of the intermediate compound (0.92 g,1.96 mmol) and potassium carbonate (1.26 g,9.15 mmol) in dimethyl sulfoxide (30 mL) at room temperature was added dropwise an aqueous solution of 35% hydrogen peroxide (4.8 mL,55 mmol). The reaction mixture was stirred at room temperature for 5 hours. LCMS showed complete reaction. The reaction solution was extracted three times with ethyl acetate (100 mL), the combined organic phases were washed with aqueous sodium thiosulfate solution, then with saturated brine, and the crude product obtained after concentration under reduced pressure was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2:1) to give a crude compound (500 mg) as a yellow solid. LCMS (ESI) m/z 488.4[ M+1 ] ] +
Ninth step: the above intermediate compound (400 mg,0.82 mmol) and potassium carbonate (566 mg,4.1 mmol) were dissolved in N, N-dimethylformamide (20 mL) at room temperature, and addedN, N' -carbonyldiimidazole (4.8 mL,55 mmol) was added. The reaction mixture was heated to 100℃and stirred for 5 hours. LCMS showed complete reaction. The reaction mixture was extracted three times with ethyl acetate (100 mL), and the combined organic phases were washed with saturated brine and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2:1) to give crude compound as a yellow solid (300 mg). LCMS (ESI) m/z 514.4[ M+1 ]] +
Tenth step: the above intermediate compound (300 mg,0.58 mmol) was dissolved in acetonitrile (20 mL), and phosphorus oxychloride (1.5 g,9.78 mmol) and dimethylaniline (72 mg,0.59 mmol) were added. The reaction mixture was heated to 100℃and stirred for 8 hours. LCMS showed the reaction was essentially complete. The reaction solution was concentrated under reduced pressure, and the residual reaction solution was diluted with ethyl acetate (50 mL), washed twice with saturated aqueous sodium bicarbonate solution, and washed with saturated brine. The separated organic phase was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 4:1) to give crude compound as a white solid (180 mg). LCMS (ESI) m/z 451.2[ M+1 ] ] +
Eleventh step: the above intermediate compound (180 mg,0.58 mmol) was dissolved in tetrahydrofuran (30 mL), and triethylamine (293 mg,2.9 mmol) and di-tert-butyl dicarbonate (262 mg,1.2 mmol) were added. The reaction mixture was stirred at room temperature for 4 hours. LCMS showed the reaction was essentially complete. To the reaction solution was added a saturated aqueous sodium carbonate solution (30 mL), and most of the organic solvent was removed by concentration under reduced pressure, followed by extraction twice with ethyl acetate (50 mL). The combined organic phases were concentrated under reduced pressure and the crude product was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2:1) to give crude compound as a white solid (210 mg). LCMS (ESI) m/z 551.3[ M+1 ]] +
Twelfth step: the above intermediate compound (210 mg,0.38 mmol) was dissolved in 1, 4-dioxane (30 mL) and DIPEA (195 mg,1.5 mmol) and 3, 8-diazabicyclo [3.2.1 were added]Octane-8-carboxylic acid tert-butyl ester (89 mg,0.42 mmol). The mixture was warmed to 50℃and stirred overnight, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2:1) to give an intermediate compound (225 mg) as a yellow solid. LCMS (ESI) m/z 727.1[ M+1 ]] +
Thirteenth step: naH (40 mg,1.67 mmol) was added to a solution of (hexahydro-1H-pyrrolin-7A-yl) methanol (66 mg,0.46 mmol) in DMF (20 mL) at room temperature under nitrogen, and after stirring for 10 min the intermediate compound of the previous step (218 mg,0.30 mmol) was added. The reaction was heated to 110℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:20) to give an intermediate product (70 mg) as a yellow solid. LC-MS [ M+H ] ] + :m/z 832.0。
Fourteenth step: the intermediate compound (70 mg,0.08 mmol) was dissolved in dichloromethane (10 mL) under nitrogen and TFA (1 mL) was added. Stirring for 30 minutes at room temperature. The LC-MS detection reaction was substantially complete, and to the reaction mixture was added saturated aqueous sodium bicarbonate (10 mL) and extracted twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by HPLC to give a pale yellow solid intermediate product (24 mg). LC-MS [ M+H ]] + :m/z 631.7。 1 H NMR(400MHz,MeOD-d 4 ):δ7.24-7.20(m,1H),7.02-6.97(m,1H),6.53(s,1H),4.54-4.48(m,1H),3.98-3.86(m,4H),3.74-3.62(m,4H),3.34-3.25(m,2H),2.75-2.65(m,3H),2.25-1.65(m,10H).
Starting with intermediates B1 to B5 and commercially available reagents, the following example compounds were obtained by the general preparation method of the examples, in analogy with reference to example 45:
Figure BDA0003785353610000541
/>
Figure BDA0003785353610000551
example 51:8- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -3- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -6- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -2-methylpyrimido [5,4-d ] pyrimidin-4 (3H) -one
Figure BDA0003785353610000561
The first step: 2, 6-dichloro-5-nitropyrimidine-4-carboxylic acid methyl ester (502 mg,2 mmol) was dissolved in 1, 4-dioxane (30 mL) and DIPEA (775 mg,6 mmol) and 3, 8-diazabicyclo [3.2.1 were added]Octane-8-carboxylic acid tert-butyl ester (450 mg,2.1 mmol). The mixture was warmed to 50℃and stirred overnight, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2:1) to give the intermediate compound (735 mg) as a yellow solid. LCMS (ESI) m/z 428.1[ M+1 ] ] +
And a second step of: naH (160 mg,6.7 mmol) was added to a solution of ((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methanol (293 mg,1.84 mmol) in DMF (30 mL) at room temperature under nitrogen, and after stirring for 20 min the intermediate compound of the previous step (730 mg,1.71 mmol) was added. The reaction was heated to 110℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (50 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (100 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:20) to give intermediate product (720 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 551.3。
And a third step of: to a solution of the above intermediate compound (720 mg,1.31 mmol) in methanol (10 mL) was added lithium hydroxide monohydrate (300 mg,7.3 mmol), and the reaction mixture was stirred overnight at room temperature. After the LC-MS detection reaction was substantially complete, the diluted acid adjusted the pH of the reaction mixture solution to neutral. The reaction solution was concentrated under reduced pressure, the residue was slurried with methanol, the solid was filtered off, and the filtrate was concentrated under reduced pressure to give a crude white solid intermediate product (550 mg). LC-MS [ M+H ]] + :m/z 537.2。
Fourth step: to a solution of the above intermediate compound (540 mg,1.01 mmol) in N, N-dimethylformamide (20 mL) was added HATU (570 mg,1.5 mmol) and DIPEA (400 mg,3.1 mmol) at room temperature. The reaction mixture was stirred at room temperature for 5 hours. LCMS showed complete reaction . The reaction mixture was extracted three times with ethyl acetate (100 mL), and the combined organic phases were washed with saturated brine and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=1:1) to give crude compound as a white solid (680 mg). LCMS (ESI) m/z 920.4[ M+1 ]] +
Fifth step: to a solution of the intermediate compound (640 mg,0.74 mmol) in tetrahydrofuran (20 mL) was added 5% Pd/C (50 mg) at room temperature. The reaction mixture was placed in a hydrogen atmosphere (1 atm) and stirred at room temperature for 1 hour. LCMS showed complete reaction, the reaction was filtered through celite and the filtrate concentrated under reduced pressure. The crude product obtained was dried to give crude compound (480 mg) as a grey solid. LCMS (ESI) m/z 890.5[ M+1 ]] +
Sixth step: trimethyl orthoacetate (2 mL) and methanesulfonic acid (78 mg,0.81 mmol) were added to a toluene solution (20 mL) of the above intermediate compound (450 mg,0.51 mmol) at room temperature. The reaction mixture was heated to 60 degrees and stirred for 5 hours. LCMS showed complete reaction and the reaction concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=1:1) to give crude compound as a white solid (120 mg). LCMS (ESI) m/z 914.4[ M+1 ]] +
Seventh step: to dichloromethane (10 mL) of the above intermediate (120 mg,0.13 mmol) was added trifluoroacetic acid (2 mL) at room temperature, and the reaction mixture was stirred at room temperature for 30 minutes. The reaction solution was concentrated under reduced pressure, and the concentrated crude product was dissolved in tetrahydrofuran (5 mL), cesium fluoride CsF (90 mg,0.6 mmol) was added to the reaction solution, and stirring was continued for 3 hours. Concentrated under reduced pressure, and the crude product was purified by HPLC to give a yellow solid product (23 mg). LC-MS [ M+H ] ] + :m/z 614.3。 1 H NMR(400MHz,CD 3 OD)δ7.85(m,1H),7.51(m,1H),7.32-7.16(m,2H),4.72-4.61(m,4H),4.07(s,2H),3.90-3.35(m,5H),3.26-3.02(m,3H),2.62(s,3H),2.53-1.85(m,11H)。
Starting with intermediates B1 to B5 and commercially available reagents, the following example compounds were obtained in analogy to example 51 using the general preparation procedure of the examples:
Figure BDA0003785353610000571
example 56:4- ((4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2 r,7 as) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5-methyl-5H-pyrrolo [3,2-d ] pyrimidin-7-yl) methyl-7-fluorobenzo [ d ] thiazol-2-amine
Figure BDA0003785353610000581
The first step: 2, 4-dichloro-5-methyl-5H-pyrrolo [3,2-d]Pyrimidine (402 mg,2.0 mmol) was dissolved in 1, 4-dioxane (30 mL) and DIPEA (775 mg,6 mmol) and 3, 8-diazabicyclo [3.2.1 were added]Tert-butyl octane-8-carboxylate (425 mg,2.0 mmol). Stirred overnight at room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2:1) to give intermediate compound (635 mg) as a yellow solid. LCMS (ESI) m/z 378.2[ M+1 ]] +
And a second step of: naH (160 mg,6.7 mmol) was added to a solution of ((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methanol (293 mg,1.84 mmol) in DMF (30 mL) at room temperature under nitrogen, and after stirring for 20 min the intermediate compound of the previous step (630 mg,1.67 mmol) was added. The reaction was heated to 110℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (50 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (100 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:10) to give intermediate product (480 mg) as a yellow solid. LC-MS [ M+H ] ] + :m/z 501.6。
And a third step of: the intermediate compound (480 mg,0.96 mmol) was dissolved in methylene chloride (30 mL), NBS (251 mg,1.41 mmol) was added, and the mixture was stirred at room temperature for 1 hour. LCMS showed complete reaction. To the reaction mixture was added saturated brine (50 mL), and the separated organic phase was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 3:1) to obtain a yellow solid crude compound (590 mg). LCMS (ESI) m/z 580.5[ M+1 ]] +
Fourth step: a tetrahydrofuran solution (20 mL) of the intermediate compound (284 mg,0.49 mmol) was reduced to-78℃under nitrogen, and then a tetrahydrofuran solution (1M, 0.6mL,0.60 mmol) of n-butyllithium was slowly added dropwise. The reaction mixture was stirred at-78℃for 10 minutes, and then (7-fluoro-4-carbaldehyde benzo [ d ] was added dropwise]Thiazole-2-amino) carboxylic acid tert-butyl ester (178 mg,0.60 mmol). After stirring the reaction mixture at-78 ℃ for 30 minutes, it was slowly warmed to room temperature. LCMS showed complete reaction. The reaction mixture was quenched by addition of saturated aqueous ammonium chloride (50 mL) and extracted three times with ethyl acetate (40 mL). The separated organic phase was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 1:1) to give crude compound (120 mg) as a yellow solid. LCMS (ESI) m/z 797.6[ M+1 ] ] +
Fifth step: to a solution of the intermediate (120 mg,0.15 mmol) in dichloromethane (30 mL) under nitrogen was added trifluoroacetic acid (50 mg,0.44 mmol) and triethylsilane (52 mg,0.45 mmol). The reaction mixture was stirred at room temperature overnight. The LC-MS detection reaction was substantially complete, water (50 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (100 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: methanol/dichloromethane=1:5) to give intermediate product (50 mg) as a yellow solid. LC-MS [ M+H ]] + :m/z 781.8。
Sixth step: to dichloromethane (10 mL) of the above intermediate (50 mg,0.06 mmol) was added trifluoroacetic acid (2 mL) at room temperature, and the reaction mixture was stirred at room temperature for 30 minutes. The reaction solution was concentrated under reduced pressure, and the crude product was purified by HPLC to give a yellow solid product (18 mg). LC-MS [ M+H ]] + :m/z 581.5。 1 H NMR(400MHz,CD 3 OD)δ7.23-6.95(m,2H),6.65(s,1H),4.72-4.35(m,6H),4.27(s,2H),3.90-3.35(m,8H),3.26-3.02(m,3H),2.67(s,3H),2.53-1.85(m,11H)。
Example 57:4- ((4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2 r,7 as) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5-methyl-5H-pyrrolo [3,2-d ] pyrimidin-7-yl) methyl-5-ethyne-6-fluoronaphthalen-2-ol
Figure BDA0003785353610000591
Referring to the synthesis of examples 56 and 51, the compound of example 57 was prepared as a white solid. LC-MS [ M+H ] ] + :m/z 599.3。 1 H NMR(400MHz,CD 3 OD)δ7.85(m,1H),7.51(m,1H),7.32-7.16(m,2H),6.75(s,1H),4.72-4.51(m,6H),4.37(s,2H),3.90-3.25(m,8H),3.26-3.02(m,3H),2.62(s,3H),2.53-1.85(m,11H)。
Example 58:4- ((4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5-methyl-5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -5-ethyne-6-fluoronaphthalen-2-ol
Figure BDA0003785353610000601
The first step: 3- (7-bromo-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5-methyl-5H-pyrrolo [3, 2-d)]Pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1]To a solution of tert-butyl octane-8-carboxylate (120 mg,0.21 mmol) in 1, 4-dioxane/water (12 mL/4 mL) was added borate B3 (98 mg,0.21 mmol), tetrakis (triphenylphosphine) palladium (48 mg,0.04 mmol) and sodium carbonate powder (Na 2 CO 3 ) (56 mg,0.53 mmol) and the reaction mixture was stirred overnight at 100deg.C under argon. After completion of the reaction, ethyl acetate was used for extraction, and the organic phase was washed with saturated brine and anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification by flash column chromatography on silica gel afforded the product as a yellow solid (100 mg). LC-MS m/z 839.7[ M+H ]] +
And a second step of: to dichloromethane (10 mL) of the above intermediate (100 mg,0.12 mmol) was added trifluoroacetic acid (2 mL) at room temperature, and the reaction mixture was stirred at room temperature for 30 minutes. The reaction solution was concentrated under reduced pressure, and the concentrated crude product was dissolved in tetrahydrofuran (5 mL), cesium fluoride CsF (90 mg,0.6 mmol) was added to the reaction solution, and stirring was continued for 3 hours. Concentrated under reduced pressure, and the crude product was purified by HPLC to give a yellow solid product (25 mg). L (L) C-MS[M+H] + :m/z 614.3。 1 H NMR(400MHz,CD 3 OD)δ7.85(m,1H),7.51(m,1H),7.32-7.16(m,2H),6.70(s,1H),4.72-4.61(m,4H),4.07(s,2H),3.83(s,3H),3.79-3.35(m,5H),3.26-3.02(m,3H),2.53-1.85(m,11H)。
Referring to the synthetic procedure of example 58, the following compounds of examples 59,60 were prepared using commercially available borates instead of alkynylnaphthol borates:
Figure BDA0003785353610000602
starting with intermediates B3 and B4 and commercially available reagents such as piperidine derivatives, the following example compounds were obtained by following similar procedures with reference to example 6 using the general preparation method of the examples:
Figure BDA0003785353610000611
starting with intermediate B4 and commercially available reagents, the following example compounds were obtained in analogy to the example general preparation methods, with reference to example 34:
Figure BDA0003785353610000612
/>
Figure BDA0003785353610000621
/>
Figure BDA0003785353610000631
starting with intermediates B3 and B4 and commercially available reagents, the following example compounds were obtained in analogy to example 45 using the general preparation methods of the examples:
Figure BDA0003785353610000632
starting with intermediate B4 and commercially available reagents, the following example compounds were obtained in analogy to the example general preparation methods, with reference to example 51:
Figure BDA0003785353610000633
/>
Figure BDA0003785353610000641
starting with intermediates B3, B4 and commercially available reagents, the following compounds of examples 86-90 were obtained in analogy to the examples general preparation and with reference to examples 57 and 58:
Figure BDA0003785353610000642
/>
Figure BDA0003785353610000651
example 91: (R) -1- (7- (8-ethyl-7-fluoro-3-hydroxynaphthalen-1-yl) -2- (((2R, 7 aS) -2-fluoro-tetrahydro-1H-pyrrolin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) -3-methylpiperidin-3-ol
Figure BDA0003785353610000652
The first step: to a methanol solution (20 mL) of boric acid ester B4 (500 mg,1.39 mmol) was added 30% hydrogen peroxide (20 mL) under ice-bath cooling. The reaction mixture was stirred at room temperature for 3 hours. After 50mL of water was added to the reaction mixture, ethyl acetate (30 mL) was extracted twice, and the combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was dissolved in methylene chloride (30 mL) and cooled in an ice bath to give a reaction solutionTo this mixture were added trifluoromethanesulfonic anhydride (290 mg,1.03 mmol) and triethylamine (0.5 mL). After the reaction mixture was stirred at room temperature for 2 hours, it was washed twice with saturated brine (20 mL), and the separated organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude intermediate naphthol triflate (270 mg). LC-MS m/z 383.0[ M+H ]] + .
And a second step of: to a solution of 7-benzyl-2, 4-dichloro-5, 6,7, 8-tetrahydro-1, 7-naphthyridine (588 mg,2.01mmol, preparation method reference CN112094269 a) in N, N-dimethylformamide (30 mL) was added N, N-diisopropylethylamine (650 mg,5.03 mmol) and (R) -3-methylpiperidin-3-ol (232 mg,2.02 mmol), and the reaction mixture was stirred overnight under argon at 80 ℃. Water (50 mL) was added to the reaction solution, and the mixture was extracted twice with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine, anhydrous Na 2 SO 4 Drying and filtering. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by silica gel flash column chromatography to give a yellow solid product (540 mg). LC-MS m/z 372.0/374.0[ M+H ]] +
And a third step of: to a solution (20 mL) of the above intermediate (540 mg,1.45 mmol) in 1, 4-dioxane was added (2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolin-7 a (5H) -yl) methanol (320 mg,2.01 mmol), xantphos (150 mg,0.26 mmol), potassium phosphate (K) 3 PO 4 ) (860 mg,4.05 mmol) and palladium acetate (49 mg,0.22 mmol). The reaction mixture was heated to 100 ℃ with sealing and stirred overnight. Water (100 mL) was added to the reaction mixture, and the mixture was extracted three times with ethyl acetate (40 mL). The combined organic phases were washed with saturated brine, anhydrous Na 2 SO 4 Drying and filtering. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by silica gel flash column chromatography to give a yellow solid product (340 mg). LC-MS m/z 495.3[ M+H ]] +
Fourth step: to t-butanol (15 mL) of the intermediate compound (340 mg,0.69 mmol) was added wet palladium on carbon (30 mg) under a hydrogen atmosphere. The reaction was stirred at room temperature for 48 hours. The LC-MS detection reaction is basically complete, and the reaction solution is filtered through kieselguhr and washed by ethanol. The filtrate was concentrated under reduced pressure to give an intermediate product (180 mg) as a pale yellow solid. LC-MS [ M+H ] ] + :m/z 405.2。
Fifth step: under nitrogen gasThe above intermediate compound (180 mg,0.44 mmol) and naphthol triflate (270 mg,0.71 mmol) prepared in the first step were dissolved in toluene (30 mL) under protection, cesium carbonate (163 mg,0.5 mmol) and Pd were added 2 (dba) 3 (42 mg,0.05 mmol) and 2-dicyclohexylphosphine-2 ',6' -diisopropyloxybiphenyl Ruphos (23 mg,0.05 mmol). The reaction was heated to 80℃and stirred overnight. The LC-MS detection reaction was substantially complete, water (30 mL) was added to the reaction solution, and extraction was performed twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (32 mg). LC-MS [ M+H ]] + :m/z 637.3。
Sixth step: the intermediate compound (32 mg,0.05 mmol) was dissolved in THF (10 mL) under nitrogen and TFA (1 mL) was added. Stirring for 30 minutes at room temperature. The LC-MS detection reaction was substantially complete, and to the reaction mixture was added saturated aqueous sodium bicarbonate (10 mL) and extracted twice with ethyl acetate (50 mL). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the obtained crude product was purified by HPLC to give a pale yellow solid intermediate product (12 mg). LC-MS [ M+H ]] + :m/z 593.3。 1 H NMR(400MHz,CD 3 OD)δ7.65-7.22(m,2H),7.05-6.95(m,2H),6.13(s,1H),5.31(m,1H),4.52-4.15(m,3H),4.04-3.47(m,8H),3.24-3.03(m,6H),2.67-2.35(m,3H),2.27-1.65(m,8H),1.32(t,J=6.4Hz,3H),1.29(s,3H).
Starting with intermediate B4 and commercially available reagents, the following compounds of examples 92-100 were obtained in a similar manner to the general preparation of the examples, with reference to example 91:
Figure BDA0003785353610000671
/>
Figure BDA0003785353610000681
The following compounds of examples 101-110 were obtained in analogy to reference example 91 starting from 7-benzyl-2, 4-dichloro-3-fluoro-5, 6,7, 8-tetrahydro-1, 7-naphthyridine instead of 7-benzyl-2, 4-dichloro-5, 6,7, 8-tetrahydro-1, 7-naphthyridine:
Figure BDA0003785353610000682
/>
Figure BDA0003785353610000691
test example 1 pair KRAS of the inventive Compound G12D Impact test of mediated ERK phosphorylation Capacity
The testing method comprises the following steps: 1) Expression of KRAS G12D AGS cells (ATCC) were cultured in DMEM medium containing 10% fetal bovine serum, 10mM HEPES and penicillin/streptomycin, inoculated in 96-well plates at a density of 40,000 cells/well, and attached for 12-14 hours. 2) After 3 hours, the medium was removed, 150. Mu.L of 4.0% formaldehyde was added, and the plate was incubated at room temperature for 20 minutes. 3) The plates were washed with PBS and infiltrated with 150. Mu.L ice-cold methanol for 10 min. 4) Binding of non-specific antibodies to the plates was blocked with 100 μl of blocking buffer for 1 hour at room temperature. Phosphorylation of ERK was detected using p-ERK specific antibodies, GAPDH as an internal standard. The information and experimental conditions of the antibody are as follows: p-ERK (Cell signaling) was diluted 1:500 in blocking buffer+0.05% tween 20; GAPDH was diluted 1:500 in blocking buffer+0.05% tween 20. The antibody was incubated for 2h at room temperature, PBS+0.05% Tween20, and the plate was washed. The secondary antibodies used to display primary antibodies were added as follows: anti-rabbit-680 was diluted 1:1000 in blocking buffer+0.05% tween20, anti-mouse-800 was diluted 1:1000 in blocking buffer+0.05% tween20 and incubated for 1 hour at room temperature. 5) Plates were washed with PBS +0.05% tween20, 100 μl PBS was added to each well and the plates were read. 6) The phosphorylated ERK (Thr 202/Tyr 204) signal was normalized to GAPDH signal for each well and the percentage of DMSO control value was calculated. Calculation of IC by four-parameter dose-response curve fitting 50 Values.
Results: most of the implementations provided by the inventionThe compounds of examples have a pronounced inhibitory effect on phosphorylated ERK levels in AGS cells, IC of all examples 50 All less than 3000nM, and some examples even less than 100nM. Specific activity results are shown in the table. (A represents 1000 nM.ltoreq.IC 50 <3000nM, B represents 100 nM.ltoreq.IC 50 <1000nM, C represents IC 50 <100nM)
Figure BDA0003785353610000701
/>
Figure BDA0003785353610000711
Test example two, test example Compounds for BaF3-KRAS-G12D cell proliferation inhibition Activity
The test adopts CellTiter-Glo (CTG) kit provided by Promega company, which is a method for detecting cell viability by a homogenization method, and the cell viability of the cultured cells is determined by quantifying ATP.
1. Experimental reagent consumable: RPMI1640 (Hyclone, SH 30809.01), fetal Bovine Serum (FBS, gibco, 10099-141), phosphate Buffered Saline (PBS, solarbio, P1020-500), celltiter Gloassay kit (Promega, G7573), blank 96-cell culture plate (Thermo, 165305).
2. Experimental instrument equipment: CO2 incubator (Thermo Scientific, model 3100 Series), microscope (OLYMPUS, CKX41 SF), multifunctional microplate reader (BMG,
Figure BDA0003785353610000722
plus), biosafety cabinet (Thermo, model 1300 Series A2)
3. Cell proliferation assay: all cell lines were cultured in complete medium at 37℃under 5% CO 2. Cells in the logarithmic growth phase were harvested and counted using a platelet counter. Cell viability was checked by trypan blue exclusion, ensuring that cell viability was above 90%. The cell density was adjusted using complete medium and then seeded into 96 well cell culture plates with 90 μl total of 3000 cells per well. Cells in 96-well plates were incubated at 37℃under 5% CO 2. A 10-fold drug solution was prepared and then transferred from 10 μl of each serial diluted compound to the corresponding experimental well of a 96-well cell plate, the initial concentration of compound test 10um, 3-fold dilution, 9 concentrations, three multiplex wells were set for each drug concentration. Cells in the dosed 96-well plates were incubated at 37℃under 5% CO2 for a further 72 hours before CTG analysis. The CTG reagent was thawed and the cell plates equilibrated to room temperature for 30 minutes. An equal volume of CTG solution was added to each well. Cells were lysed by shaking on an orbital shaker for 5 minutes. The cell plates were left at room temperature for 20 minutes to stabilize the luminescence signal. Using Luminescence Read Mode, luminescence values (luminescences) were read and data collected.
4. Data analysis: the data were analyzed using GraphPad Prism 7.0 software, and non-linear S-curve regression was used to fit the data to yield the dose-response curve, and IC50 values were calculated therefrom. Cell viability (%) = (Lum) Drug to be tested -Lum Culture broth control )/(Lum Cell control -Lum Culture broth control ) X 100%. (A represents IC 50 Value of<1000nM, B representing 1000 nM.ltoreq.IC 50 Value of<10000nM, C represents IC 50 ≥10000nM)
Figure BDA0003785353610000721
/>
Figure BDA0003785353610000731
All documents mentioned in this application are incorporated by reference as if each were individually incorporated by reference. Further, it will be appreciated that various changes and modifications may be made by those skilled in the art after reading the above teachings, and such equivalents are intended to fall within the scope of the claims appended hereto.

Claims (8)

1. A nitrogen-containing heterocyclic compound represented by the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, stereoisomer, solvate, polymorph or prodrug thereof,
Figure FDA0003785353600000011
wherein, the liquid crystal display device comprises a liquid crystal display device,
R 1 selected from substituted or unsubstituted piperazine, substituted or unsubstituted piperidine, or substituted or unsubstituted 5-10 membered heterocycloalkyl, said substituents being selected from one or more of the following groups: halogen, cyano, hydroxy, amino, - (CO) -NR 11 NR 12 、C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkylcyano, C 1 -C 6 Alkyl hydroxy, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, or a 3-10 membered carbocyclic or heterocyclic ring system between each of the foregoing substituents, said ring system comprising spiro, bridged, fused or fused rings; r is R 11 、R 12 Independently selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or R 11 、R 12 Together with the attached N atom, form a 3-8 membered heterocycloalkyl;
R 2 selected from substituted or unsubstituted C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl; the substituent is selected from the following groups: halogen, cyano, hydroxy, amino, monoalkylamino, dialkylamino, monoalkylaminoalkyl, dialkylaminoalkyl, C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl, 6-12 membered "saturated or partially unsaturated spiro, bridged, fused or fused ring containing 0 to 3 heteroatoms independently selected from N, O, S, P";
R 4 a substituent selected from one or more of the following groups: hydrogen, deuterium, halogenHydroxy, amino, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, or a 3-to 10-membered saturated or partially unsaturated or aromatic ring system formed between the two substituents via a carbon atom or heteroatom;
l is selected from chemical bond, CHR 5 、O、S、NR 5 ;R 5 Selected from H, C 1 -C 6 Alkyl of (a);
L 1 selected from direct bond, CHR 5 、O、OR 5 、S、NR 5 ;R 5 Selected from H, C 1 -C 6 Alkyl of (a);
ar is selected from benzene ring, naphthalene ring or benzothiazole ring, and the Ar can be one or more different R 3 Substitution, said R 3 Independently selected from halogen, hydroxy, amino, cyano, C 1 -C 6 Alkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 1 -C 6 An alkoxy group;
m is N or CR 6 ;R 6 Selected from hydrogen, halogen, CN, C 1 -C 3 An alkyl group;
cy is selected from the following group of ring systems:
Figure FDA0003785353600000012
wherein, the hetero atoms in the heterocyclic ring system, the heterocyclic alkyl and the heteroaryl are independently selected from N, O, S, P, and the number of the hetero atoms is 1-3.
2. A nitrogen-containing heterocyclic compound of the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, as claimed in claim 1, wherein:
m is N, C-H, C-F, C-CN;
cy is selected from
Figure FDA0003785353600000021
/>
R 1 Selected from:
Figure FDA0003785353600000022
Figure FDA0003785353600000023
r is as described above 1 The hydrogen on any C of the radicals being halogen, hydroxy, C 1 -C 6 Alkyl, or = O substitution;
ar is selected from benzene ring, naphthalene ring or benzothiazole ring, and the Ar can be one or more different R 3 Substitution, said R 3 Selected from halogen, hydroxy, amino, cyano, C 1 -C 6 Alkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 1 -C 6 Alkoxy groups, and the like; r is R 3 Preferably F, hydroxy, methyl, ethyl, ethynyl, amino;
l is selected from: CH (CH) 2 O, NH or S;
L 1 selected from: direct bond, CH 2 O, NH or S;
R 2 selected from substituted or unsubstituted C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted C 1 -C 3 An alkyl group; the substituent is selected from the following groups: halogen, cyano, hydroxy, amino, mono C 1 -C 6 Alkylamino, bis C 1 -C 6 Alkylamino, mono C 1 -C 6 alkylamino-C 1 -C 6 Alkyl, bis C 1 -C 6 alkylamino-C 1 -C 6 Alkyl, C 1 -C 6 Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 5-12 membered aryl or heteroaryl, 3-12 membered cycloalkyl or heterocycloalkyl substituted C 1 -C 3 Alkyl, 6-12 membered saturated or partially unsaturated spiro, bridged, or parallel rings containing 0-3 heteroatomsCondensed rings, etc.; the substituents are more preferably F, methyl,
Figure FDA0003785353600000024
3. The nitrogen-containing heterocyclic compound as described in claim 1 or 2, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
R 1 Selected from the group consisting of
Figure FDA0003785353600000025
Figure FDA0003785353600000031
Cy is selected from
Figure FDA0003785353600000032
R 3 Ar is selected from
Figure FDA0003785353600000033
L is selected from-O-;
L 1 selected from direct bond, CH 2
R 2 Selected from the group consisting of
Figure FDA0003785353600000034
Wherein m and n are each selected from integers from 1 to 3; ry is selected from alkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl, 5-10 membered aryl, heteroaryl; r is R p And R is q Selected from hydrogen, halogen, C 1 -C 6 Alkyl or alkoxy, hydroxy, amino, or R p And R is q Forming a 3-to 10-membered carbocyclic or heterocyclic ring system;
R 4 selected from hydrogen, deuterium, halogen, methyl, trifluoromethyl, cyano.
4. A nitrogen-containing heterocyclic compound of the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, as claimed in claim 1, wherein:
m is preferably N, C-H, C-F, C-CN;
alternatively, cy is selected from
Figure FDA0003785353600000035
Alternatively, R 11 、R 12 Independently selected from hydrogen, halogen, methyl;
alternatively, when R 11 、R 12 When taken together with the attached N atom form a 5-8 membered heterocycloalkyl, the 3-8 membered heterocycloalkyl is preferably
Figure FDA0003785353600000036
Alternatively, R 1 Selected from substituted or unsubstituted piperazine, substituted or unsubstituted piperidine, or substituted or unsubstituted 5-10 membered heterocycloalkyl, said substituents being selected from one or more of the following groups: halogen, hydroxy, C 1 -C 6 Alkyl group,
Figure FDA0003785353600000037
Figure FDA0003785353600000038
Or substituents form a 3-to 10-membered carbocyclic or heterocyclic ring system between them, said ring system comprising spiro, bridged, fused or fused rings; r is R 1 Preferably selected from: />
Figure FDA0003785353600000041
Figure FDA0003785353600000042
R is as described above 1 The hydrogen on any C of the radicals being halogen, hydroxy, C 1 -C 6 Alkyl, or = O substitution;
alternatively, R 3 Ar is selected from
Figure FDA0003785353600000043
Alternatively, L is selected from-O-;
alternatively, L 1 Selected from direct bond, CH 2
Alternatively, R 2 Selected from the group consisting of
Figure FDA0003785353600000044
Wherein m and n are each selected from integers from 1 to 3; ry is selected from alkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl, 5-10 membered aryl, heteroaryl; r is R p And R is q Selected from hydrogen, halogen, C 1 -C 6 Alkyl or alkoxy, hydroxy, amino, or R p And R is q Forming a 3-to 10-membered carbocyclic or heterocyclic ring system; />
Figure FDA0003785353600000045
Preferably +.>
Figure FDA0003785353600000046
Alternatively, R 4 Selected from hydrogen, deuterium, halogen, methyl, trifluoromethyl, cyano.
5. A nitrogen-containing heterocyclic compound of the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, as claimed in claim 1, wherein:
the monoalkylamino group, dialkylamino group, monoalkylaminoalkyl group and dialkylaminoalkyl group are each preferably (C) 1 -C 6 Alkyl) NH- (C 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-, C 1 -C 6 alkyl-NH-C 1 -C 6 Alkyl-, (C) 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-C 1 -C 6 Alkyl-;
alternatively, the 3-12 membered cycloalkyl or heterocycloalkyl substituted alkyl is preferably (3-12 membered cycloalkyl or heterocycloalkyl) -C 1 -C 6 Alkyl-; the alkyl-substituted amino group is preferably (C) 1 -C 6 Alkyl) NH-or (C) 1 -C 6 Alkyl) (C) 1 -C 6 Alkyl) N-;
alternatively, the R p And R is R q Forming a three-membered carbocyclic ring with the attached carbon atom;
alternatively, the C 1 -C 6 Preferably HO-CH 2 -、HO-CH 2 -CH 2 -。
6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, wherein the compound has the structure:
Figure FDA0003785353600000051
/>
Figure FDA0003785353600000061
/>
Figure FDA0003785353600000071
7. a pharmaceutical composition comprising an effective amount of the nitrogen-containing heterocyclic compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, and a pharmaceutically acceptable carrier.
8. Use of a nitrogen-containing heterocyclic compound as described in any one of claims 1-6, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, or a pharmaceutical composition as described in claim 7, for the preparation of a Ras mutein inhibitor or medicament; the Ras mutant protein can be KRAS G12D The method comprises the steps of carrying out a first treatment on the surface of the The medicine can be used for treating diseases related to activity or expression quantity of Ras mutant protein; alternatively, the drug may be a therapeutic drug for a tumor; the tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, bile duct cancer, brain cancer, leukemia, lymphoma, fibroma, sarcoma, basal cell carcinoma, glioma, renal cancer, melanoma, bone cancer, thyroid cancer, nasopharyngeal cancer, and pancreatic cancer.
CN202210940525.8A 2021-08-08 2022-08-06 Nitrogen-containing heterocyclic compound, preparation method and application Pending CN116143806A (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN202110905164 2021-08-08
CN2021109051649 2021-08-08
CN2021110953899 2021-09-17
CN202111095389 2021-09-17
CN2022100395072 2022-01-13
CN202210039507 2022-01-13
CN202210293818 2022-03-23
CN2022102938181 2022-03-23
CN2022107538310 2022-06-28
CN202210753831 2022-06-28

Publications (1)

Publication Number Publication Date
CN116143806A true CN116143806A (en) 2023-05-23

Family

ID=86339488

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210940525.8A Pending CN116143806A (en) 2021-08-08 2022-08-06 Nitrogen-containing heterocyclic compound, preparation method and application

Country Status (1)

Country Link
CN (1) CN116143806A (en)

Similar Documents

Publication Publication Date Title
CN113563323B (en) Benzothiazolyl biaryl compound, preparation method and application
JP7335882B2 (en) Pyrimidine-condensed ring compound, method for producing the same, and use
CN115192577B (en) KRAS mutein inhibitors
CN115448923B (en) Pyrimidine-fused ring compound, preparation method and application thereof
EP3112364B1 (en) 2,4-disubstituted phenylene-1,5-diamine derivatives and applications thereof, and pharmaceutical compositions and pharmaceutically acceptable compositions prepared therefrom
CA2984586C (en) Fused-ring or tricyclic aryl pyrimidine compound used as kinase inhibitor
CN106749233B (en) Sulfonamide derivatives and application thereof
CN112142735A (en) Condensed cyanopyridine compound, preparation method and application
TW201018696A (en) Fused bicyclic and tricyclic pyrimidine compounds as tyrosine kinase inhibitors
CN113135910A (en) Pyrimidine-4 (3H) -ketone heterocyclic compound, preparation method and pharmaceutical application thereof
CN116648452A (en) Preparation of heterocyclic compounds as KRAS inhibitors and methods of use thereof
CN113061132B (en) Condensed ring lactam compound, preparation method and application
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
WO2023061294A1 (en) Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
CN114656482A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN107207504B (en) Phthalazinone derivatives, preparation method and use thereof
JP2021501215A (en) Amino-substituted nitrogen-containing condensed ring compound, its preparation method and use
AU2022214080A1 (en) Phosphoinositide 3 kinase beta inhibitors and compositions and methods thereof
CN116801883A (en) Heteroaromatic ring compounds, preparation method and application thereof
CN116323562A (en) Compounds with kinase inhibitory activity
CN116143806A (en) Nitrogen-containing heterocyclic compound, preparation method and application
JP2022554385A (en) WDR5 inhibitors and modulators
CN117800976A (en) Nitrogen-containing heterocyclic compound, preparation method and application
CN116143805A (en) Nitrogen-containing heterocyclic biaryl compounds, preparation method and application

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication