CN111393404A - Benzothiophene compounds, and pharmaceutical composition and application thereof - Google Patents

Benzothiophene compounds, and pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN111393404A
CN111393404A CN201910002427.8A CN201910002427A CN111393404A CN 111393404 A CN111393404 A CN 111393404A CN 201910002427 A CN201910002427 A CN 201910002427A CN 111393404 A CN111393404 A CN 111393404A
Authority
CN
China
Prior art keywords
compound
group
heteroatoms selected
formula
reaction
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.)
Granted
Application number
CN201910002427.8A
Other languages
Chinese (zh)
Other versions
CN111393404B (en
Inventor
张翱
耿美玉
丁春勇
谢作权
张燕
沈安成
王玺渊
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 Institute of Materia Medica of CAS
Original Assignee
Shanghai Institute of Materia Medica of CAS
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 Institute of Materia Medica of CAS filed Critical Shanghai Institute of Materia Medica of CAS
Priority to CN201910002427.8A priority Critical patent/CN111393404B/en
Publication of CN111393404A publication Critical patent/CN111393404A/en
Application granted granted Critical
Publication of CN111393404B publication Critical patent/CN111393404B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/54Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • C07D333/60Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to benzothiophene compounds, and a pharmaceutical composition and application thereof. Specifically, the compound has a structure shown in a formula I, wherein the definition of each group and substituent is described in the specification. The invention also discloses a preparation method of the compound and an application of the compound in the aspect of tumor resistance.

Description

Benzothiophene compounds, and pharmaceutical composition and application thereof
Technical Field
The invention relates to the field of medicines, in particular to benzothiophene compounds, and a pharmaceutical composition and application thereof.
Background
Interferons (IFNs) are a family of multifunctional cytokines with antiviral, antiproliferative, and immunomodulatory activities, whose chemical components are glycoproteins that can be classified into type i, type ii, and type iii, depending on their binding receptors, and are key components of the body's natural immune system, wherein type i interferons (mainly α/β) are closely related to the pathology and clinical manifestations of various diseases, including infectious diseases, viral infections, cancer, and autoimmune diseases.
The induction of type I interferon gene expression not only plays an important role in natural immune signal pathway, but also activates immune stimulating cells including dendritic cells, etc., changes the tumor microenvironment, and activates the adaptive immune system through a series of cascade reactions, induces and activates the generation of tumor specific T cells, and further kills tumor cells.
For example, the normal activation of cyclic guanosine monophosphate synthetase (cGAS) signal pathway can not only induce the production of type I interferon and other cytokines, activate T cells targeting tumors, but also trigger the immune response of the body to tumor cells, thereby enhancing the efficacy of radiotherapy of tumors, for example, cGAS can sense DNA released by killed tumor cells to activate interferon gene stimulating proteins, thereby inducing dendritic cells to produce type I interferon, thereby activating potential anti-tumor immune response, and enhancing the efficacy of radiotherapy.
Although two monoclonal antibody drugs against the immune checkpoint PD-1/PD-L1 have been successfully marketed, the overall effectiveness of these drugs is relatively low, only about 20-30%, whereas PD-1/L1 inhibitors can relieve the inhibition of T cell activation, but if T cells do not exist in/near the tumor, they are also poorly effective, which is partly responsible for the low overall effectiveness of these drugs.
In conclusion, activating the interferon gene to stimulate protein and inducing the generation of type I interferon becomes a potential research direction in the field of anti-tumor immunity. The interferon gene stimulating protein agonist which is reported to be effective to both human and murine sources at present is mainly Cyclic Dinucleotide (CDN) compounds, for example, ADU-S100 has entered into the first clinical research, but the compounds have complex structures and are not easy to synthesize, more importantly, the defects of unstable metabolism exist, and the like, so that the administration mode of the medicine is mainly intratumoral injection at present, and the clinical application of the medicine is greatly limited.
Therefore, there is a need for further development of novel small molecules with simple structure, convenient synthesis and stable metabolism, which activate interferon gene stimulating protein and induce the production of type I interferon IFN- β.
Disclosure of Invention
The invention aims to provide a compound shown in a formula I, a preparation method thereof and an application thereof in the aspect of tumor resistance.
In a first aspect of the invention, there is provided a compound of formula I, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof,
Figure BDA0001934211350000021
wherein the content of the first and second substances,
R1and R2Independently selected from the group consisting of substituted or unsubstituted: H. halogen, amino, hydroxyl, carboxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, O, S, 3-8 membered heterocycloalkoxy containing 1-3 heteroatoms selected from N, O, S, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S; or R1And R2Together with the carbon atom to which they are attached form a 5-14 membered heterocyclyl;
the R is1And R2The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: halogen, amino, hydroxyl, carboxyl, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S;
X1and X2Independently selected from the group consisting of: H. d, halogen;
X3selected from the group consisting of: NH, O, S, Se;
X4selected from the group consisting of: o, NR5、S;
n is an integer selected from 0, 1,2, 3, 4, 5, 6, 7, 8;
R3independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic acid group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-to 10-membered heteroaryl containing 1 to 3 heteroatoms selected from N, O, S, 3-to 8-membered heterocycloalkyl containing 1 to 3 heteroatoms selected from N, O, S;
R4and R5Independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic acid group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-to 10-membered heteroaryl containing 1 to 3 heteroatoms selected from N, O, S, 3-to 8-membered heterocycloalkyl containing 1 to 3 heteroatoms selected from N, O, S;
the R is3、R4And R5The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: hydroxyl, halogen, carboxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C12 alkoxy, C3-C8 cycloalkyl, C3-C8 cycloalkoxy, C6-C10 aryl, halogenated C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S.
In another preferred embodiment, the "5-14 membered heterocyclyl" is a heterocyclyl containing at least 2O.
In another preferred embodiment, the "5-14 membered heterocyclyl" is a heterocyclyl containing 2O's.
In another preferred embodiment, the "5-14 membered heterocyclyl" is cycloalkoxy.
In another preferred embodiment, X1And X2Independently selected from the group consisting of: H. d, fluorine;
X3selected from the group consisting of: o, S, respectively;
n is an integer selected from 1,2, 3;
R1and R2Independently selected from the group consisting of substituted or unsubstituted: H. halogen, C2-C6 alkenyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocycloalkoxy containing 1-3 heteroatoms selected from N, O, S; or R1And R2Together with the carbon atom to which they are attached form a 5-14 membered heterocycloalkoxy;
the R is1And R2The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: halogen, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S.
In another preferred embodiment, X4Selected from the group consisting of: o, NR5
n is an integer selected from 1,2, 3;
R3independently selected from the group consisting of substituted or unsubstituted: H. C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S, 3-8 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, O, S;
R4and R5Independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic acid group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-to 10-membered heteroaryl containing 1 to 3 heteroatoms selected from N, O, S, 3-to 8-membered heterocycloalkyl containing 1 to 3 heteroatoms selected from N, O, S;
the R is3、R4And R5The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: hydroxy, halogen, carboxy, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C12 alkoxy, C3-C8 cycloalkoxy, C2-C6 alkenyl, C6-C10 aryl, haloC 6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S.
In another preferred embodiment, X4Is O.
In another preferred embodiment, the compound is selected from the compounds listed in table 1.
In a second aspect of the present invention, there is provided a process for the preparation of a compound according to the first aspect of the present invention, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof, selected from the group consisting of:
the method one comprises the following steps:
Figure BDA0001934211350000041
a-1) reacting a compound of formula 1 with a compound of formula 2 or a hydrochloride thereof in the presence of a condensing agent to obtain a compound of formula 3;
Figure BDA0001934211350000051
a-2) optionally reacting the compound of formula 3 with a lawson's reagent to provide a compound of formula 4;
wherein R is1、R2、R3、R4、X1、X2、X4N is as defined in the first aspect of the invention;
the second method comprises the following steps:
Figure BDA0001934211350000052
b-1) reacting the compound of formula 5 with the compound of formula 6 or a hydrochloride thereof in the presence of a condensing agent to obtain a compound of formula 7;
Figure BDA0001934211350000053
b-2) optionally subjecting the compound of formula 7 to a coupling reaction to obtain a compound of formula 8;
wherein R is1、R2、R3、R4、X1、X2、X3、X4N is as defined in the first aspect of the invention, and R2Is not a halogen.
In another preferred embodiment, the condensing agent is selected from the group consisting of: EDCI, HBTU, TBTU, HATU.
In another preferred embodiment, the Lawson's reagent is 2, 4-bis (p-methoxyphenyl) -1, 3-dithiodiphosphetane-2, 4 sulfide.
In a third aspect of the present invention, there is provided a pharmaceutical composition comprising:
(i) one or more therapeutically effective amounts of a compound of the first aspect of the invention, or its isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof; and
(ii) a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition is an injection, a sachet, a tablet, a pill, a powder or a granule.
In another preferred embodiment, the pharmaceutical composition further comprises one or more second therapeutic agents, which are drugs for preventing and/or treating cancer.
In another preferred embodiment, the second therapeutic agent is a traditional cytotoxic chemotherapy drug or other anti-tumor immune drug.
In a fourth aspect of the present invention, there is provided a use of the compound of the first aspect of the present invention, or its isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of the third aspect of the present invention for the preparation of a formulation for the prevention and/or treatment of a disease associated with type I interferon.
In another preferred embodiment, the type I interferon is selected from the group consisting of IFN- α and IFN- β.
In another preferred embodiment, the type I interferon related diseases are selected from the group consisting of: breast cancer, ovarian cancer, liver cancer, melanoma, prostate cancer, colon cancer, and gastric cancer.
In a fifth aspect of the present invention, there is provided an interferon gene stimulating protein agonist comprising one or more compounds according to the first aspect of the present invention, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof.
In a sixth aspect of the present invention, there is provided a method for preventing and/or treating a disease associated with type I interferon, comprising the steps of: administering to a patient in need thereof a therapeutically effective amount of one or more compounds of the first aspect of the invention, or isomers, prodrugs, solvates, hydrates, or pharmaceutically acceptable salts thereof, or a pharmaceutical composition of the third aspect of the invention.
In another preferred embodiment, the method is non-diagnostic, non-therapeutic.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Detailed Description
The present inventors have conducted extensive and intensive studies for a long time and have unexpectedly prepared a compound represented by formula I, which has a simple structure, is convenient to synthesize, and is metabolically stable, and has an excellent activation property for interferon gene-stimulating protein, thereby activating T cells and significantly promoting the expression of interferon factor IFN- β, thereby achieving effective treatment of tumors and complications thereof.
Term(s) for
In the present invention, unless otherwise specified, the terms used have the ordinary meanings well known to those skilled in the art.
In the present invention, the term "halogen" means F, Cl, Br or I.
In the present invention, "C1-C6 alkyl" means a straight or branched chain alkyl group including 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, neopentyl, tert-pentyl, hexyl or the like.
In the present invention, the term "C3-C8 cycloalkyl" refers to a cyclic alkyl group having 3 to 8 carbon atoms in the ring, including, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like.
In the present invention, the term "aromatic ring" or "aryl" has the same meaning, and is preferably "C6-C10 aryl". The term "C6-C10 aryl" refers to an aromatic ring group having 6 to 10 carbon atoms, such as phenyl, naphthyl, and the like, that does not contain heteroatoms in the ring.
In the present invention, the term "heterocycloaryl" or "heteroaryl" has the same meaning and refers to a heteroaromatic group containing one to more heteroatoms. For example, "C3-C10 heteroaryl" refers to an aromatic heterocycle containing 1 to 4 heteroatoms selected from oxygen, sulfur and nitrogen and 3 to 10 carbon atoms. Non-limiting examples include: furyl, thienyl, pyridyl, pyrazolyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring joined to the parent structure is a heteroaryl ring. Heteroaryl groups may be optionally substituted or unsubstituted.
In the present invention, the term "halo" means substituted by halogen.
In the present invention, the term "C2-C6 alkenyl group" means a straight or branched chain alkenyl group having 2 to 6 carbon atoms containing one double bond, including, but not limited to, ethenyl, propenyl, butenyl, isobutenyl, pentenyl, hexenyl and the like.
In the present invention, the term "C2-C6 alkynyl" refers to a straight or branched alkynyl group having 2 to 6 carbon atoms containing one triple bond, including, but not limited to, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl, and the like.
In the present invention, the term "C1-C6 alkoxy" means a straight or branched chain alkoxy group having 1 to 6 carbon atoms, including, but not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy and the like. C1-C4 alkoxy is preferred.
In the present invention, the term "substituted" means that one or more hydrogen atoms on a specified group are replaced with a specified substituent. Particular substituents are those described correspondingly in the foregoing, or as appearing in the examples. Unless otherwise specified, a certain substituted group may have one substituent selected from a specific group at any substitutable site of the group, and the substituents may be the same or different at each position. It will be understood by those skilled in the art that the combinations of substituents contemplated by the present invention are those that are stable or chemically achievable. Such substituents are for example (but not limited to): halogen, hydroxyl, carboxyl (-COOH), C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-to 12-membered heterocyclyl, aryl, heteroaryl, C1-C8 aldehyde, C2-C10 acyl, C2-C10 ester, amino, C1-C6 alkoxy, C1-C10 sulfonyl and the like.
In the present invention, the term "sulfonic acid group" has the following structure:
Figure BDA0001934211350000081
in the present invention, the term "phosphoryl" has the following structure:
Figure BDA0001934211350000082
compound (I)
The invention provides a compound shown in formula I, or an isomer, a prodrug, a solvate, a hydrate or a pharmaceutically acceptable salt thereof,
Figure BDA0001934211350000083
wherein R is1、R2、R3、R4、X1、X2、X3、X4N is as defined above.
In another preferred embodiment, in the compound, R1、R2、R3、R4、R5、X1、X2、X3、X4And n are respectively the corresponding groups in the specific compounds in the table 1.
In another preferred embodiment, the compound is preferably the compound prepared in the examples.
In another preferred embodiment, the compound is selected from the compounds listed in table 1.
TABLE 1
Figure BDA0001934211350000084
Figure BDA0001934211350000091
Figure BDA0001934211350000101
Salt form
As used herein, the term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, and the like; organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and the like; and amino acids such as proline, phenylalanine, aspartic acid, glutamic acid, etc.
Another preferred class of salts are those of the compounds of the invention with bases, for example alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., magnesium or calcium salts), ammonium salts (e.g., lower alkanolammonium salts and other pharmaceutically acceptable amine salts), for example methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butylamine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, triethanolamine salts, and amine salts formed from morpholine, piperazine, lysine, respectively.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
The term "prodrug" includes a class of compounds which are biologically active or inactive in nature and which, when administered by an appropriate method, undergo a metabolic or chemical reaction in the body to convert the compound to formula I, or a salt or solution of a compound of formula I. The prodrugs include, but are not limited to, carboxylate, carbonate, phosphate, nitrate, sulfate, sulfone, sulfoxide, amide, carbamate, azo, phosphoramide, glucoside, ether, acetal forms of the compounds.
Preparation method
The process for the preparation of the compounds of formula I according to the invention is described in more detail below, but these particular processes do not limit the invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
Typically, the process for the preparation of the compounds of the present invention is as follows, wherein the starting materials and reagents used are commercially available without specific reference.
Typically, the compound is prepared by a method selected from the group consisting of:
the method comprises the following steps:
Figure BDA0001934211350000111
hydrolyzing the compound A under an alkaline condition to obtain B, and reacting the B with N, O-dimethylhydroxylamine hydrochloride under a condensing agent condition to obtain C; reacting C with methyl Grignard reagent to obtain D, further brominating to obtain E, reacting E with diethyl ester under alkaline condition to obtain F, further hydrolyzing and decarboxylating to obtain G, condensing G with substituted hydroxylamine or hydrazine to obtain H, and further reacting with Lawson reagent to obtain I. Wherein R is1、R2、R3、R4、R5、X1、X2、X4N is as defined above;
the second method comprises the following steps:
Figure BDA0001934211350000121
the compound C is reacted with Grignard reagent to obtain K, further brominated to obtain L, L is reacted with diethyl to obtain M, further decarboxylated to obtain N, the N is condensed with substituted hydroxylamine or hydrazine to obtain O, and further coupled to obtain P, wherein R is1、R2、R3、R4、R5、X1、X2、X3、X4N is as defined above;
the third method comprises the following steps:
Figure BDA0001934211350000122
demethylating the compound F to obtain Q, further cyclizing to obtain R, and decarboxylating to obtain S; condensing S and substituted hydroxylamine or hydrazine to obtain T; wherein the content of the first and second substances,R1、R2、R3、R4、R5、X1、X2、X3、X4n is as defined above.
Pharmaceutical compositions and methods of administration
The present invention also provides a pharmaceutical composition comprising:
(i) one or more therapeutically effective amounts of said compound, or its isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof; and
(ii) a pharmaceutically acceptable carrier.
The compound of the present invention and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof, and pharmaceutical compositions containing the compound as a main active ingredient can be used for treating, preventing and relieving diseases related to tumors because the compound of the present invention has excellent antitumor activity.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. The therapeutically effective amount is determined according to the age, condition, course of treatment, etc. of the subject to be treated. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 10-1000mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
The "pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are sugars (e.g., glucose, sucrose, lactose, etc.), starches (e.g., corn starch, potato starch, etc.), celluloses and derivatives thereof (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g., propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g., tween), wetting agents (e.g., sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The benzothiophene derivatives represented by the above general formula I and pharmaceutically acceptable salts thereof in the present invention may be administered alone or in combination with other pharmaceutically acceptable therapeutic agents, particularly in combination with conventional cytotoxic chemotherapy drugs, tumor immune checkpoint inhibitors, including, but not limited to, other acceptable therapeutic agents in combination with the benzothiophene derivatives represented by the general formula I, for example, the immune checkpoint inhibitor monoclonal antibody drugs Opdivo and Keytruda, which act directly on the PD-1 protein on the surface of T cells, interrupting the binding of PD-1/PD-L to each other, as well as conventional cytotoxic chemotherapy small molecule drugs, such as the antitumor drugs Methotrexate (MTX), 5-fluorouracil (5FU), etc., which affect nucleic acid synthesis, such as doxorubicin, epirubicin, aclarubicin, mithramycin, etc., which affect nucleic acid transcription, such as antitumor drugs acting on tubulin synthesis, such as paclitaxel, vinorelbine, etc., aromatase inhibitors such as aminoglutethimide, etc., cell signaling pathway inhibitors such as epidermal growth factor receptor inhibitor Imatinib, Gefitinib (Gefitinib), or other active combination thereof, including not only one or more of the present invention in combination with a single or a combination of the present invention.
The treatment methods of the present invention can be administered alone or in combination with other therapeutic means or agents.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 50 to 1000 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
Compared with the prior art, the invention has the following main advantages:
(1) the compound has the characteristics of simple structure, easy synthesis and various administration modes;
(2) the compound has excellent activation performance on interferon gene stimulating protein, so that T cells are activated, the expression of an interferon factor IFN- β is remarkably promoted, and effective treatment on tumors and complications thereof is realized;
(3) the agonistic activity of the compound on interferon gene stimulating protein is obviously better than that of TW201817723A disclosed compound 1 g.
The present invention is further illustrated below with reference to specific examples, which are not intended to limit the scope of the invention.
For the following examples, standard procedures and purification methods known to those skilled in the art may be used. Unless otherwise specified, starting materials are generally available from commercial sources, such as Aldrich Chemicals Co. and Across organics. Commercial solvents and reagents were generally used without further purification, anhydrous solvents were processed by standard methods, and other reagents were commercially available as analytical grade. Unless otherwise stated, all temperatures are expressed in degrees Celsius (C.) and room or ambient temperature means 20-25 ℃. The structure of the compound is determined by nuclear magnetic resonance spectroscopy (NMR). The nuclear magnetic resonance hydrogen spectral shift () is given in units of parts per million (ppm). NMR Hydrogen spectra were measured using a Mercury-300MHz NMR spectrometer, deuterated chloroform (CDCl)3) Deuterated methanol (CD)3OD) as solvent and Tetramethylsilane (TMS) as internal standard.
The chromatographic column generally uses 200-300 mesh silica gel as a carrier.
In the above discussion and in the examples below, the following abbreviations have the following meanings. An abbreviation has a generally accepted meaning if it is not defined.
RT is room temperature;
DIPEA is N, N-diisopropylethylamine;
THF is tetrahydrofuran.
TBTU is O-benzotriazole-N, N, N ', N' -tetramethylurea tetrafluoroboric acid;
selectfluor is 1-chloromethyl-4-fluoro-1, 4-diazotized bicyclo 2.2.2 octane bis (tetrafluoroborate) salt.
First, compound preparation example section
The following preparations exemplarily prepare a part of the compounds of formula I of the present invention, each of which is represented by S1 to S33, respectively.
1. Synthesis of Compound S1
Figure BDA0001934211350000161
Step 1: the compound 1a (1eq) was dissolved in anhydrous methanol/tetrahydrofuran (1: 1), followed by addition of an aqueous solution of KOH (10eq) and heating to 60 ℃ for 1 hour. Most of the organic solvent is removed by spinning, then the pH value is adjusted to 5-6 by using 1N HCl, and the compound 1b is obtained after filtration and drying.1H NMR(400MHz,DMSO)13.17(s,1H),7.94(s,1H),7.58(s,1H),7.48(s,1H),3.85(s,3H),3.82(s,3H)。
Figure BDA0001934211350000162
Step 2: the compound 1b (1eq) was suspended in dichloromethane, followed by the addition of N, O-dimethylhydroxylamine hydrochloride (1.1eq), EDCI (1.5eq), and triethylamine (2.5eq) in that order, and the reaction was carried out at room temperature for 18 hours. Quenching the reaction by saturated sodium bicarbonate water solution, extracting by ethyl acetate, concentrating the organic phase, and separating by column chromatography to obtain the compound 1 c.1H NMR(400MHz,CDCl3)8.09(s,1H),7.25(s,1H),7.24(s,1H),3.96(s,3H),3.95(s,3H),3.81(s,3H),3.40(s,3H)。
Figure BDA0001934211350000171
And step 3: compound 1c (1eq) was dissolved in tetrahydrofuran, and methylmagnesium bromide (1.0M in THF,3eq) was added slowly at-10 deg.C, followed by moving to room temperature for 3 hours. Quenching with 1N HCl, extracting with ethyl acetate, and separating by column chromatography to obtain compound 1 d.1H NMR(400MHz,CDCl3)7.83(s,1H),7.26(s,1H),7.25(s,1H),3.98(s,3H),3.96(s,3H),2.63(s,3H)。
Figure BDA0001934211350000172
And 4, step 4: compound 1d (1eq) was dissolved in chloroform, pyridine tribromide (1.1eq) was added in two portions at 0 ℃ and then allowed to warm to room temperature for reaction overnight. After the reaction is finished, the solvent is dried by spinning, and the obtained solid is stirred in waterAfter 10 minutes, filtration was carried out, and the obtained solid was recrystallized from ethyl acetate to obtain compound 1 e.1H NMR(400MHz,DMSO)8.29(s,1H),7.63(s,1H),7.48(s,1H),4.87(s,2H),3.87(s,3H),3.85(s,3H)。
Figure BDA0001934211350000173
And 5: potassium carbonate (0.65eq) was suspended in DMF, then diethyl malonate (1.05eq) was added at room temperature, compound 1e (1eq) was added after stirring for 3 hours, and the temperature was raised to 110 ℃ for reaction for 4 hours. After the reaction is finished, cooling the reaction liquid to room temperature, quenching the reaction liquid by using water, extracting the reaction liquid by using ethyl acetate, and separating the extraction liquid by using column chromatography to obtain a compound 1 f.1H NMR(400MHz,CDCl3)7.92(s,1H),7.25(s,1H),7.24(s,1H),4.30–4.19(m,4H),4.07(t,1H,J=7.2Hz),3.98(s,3H),3.95(s,3H),3.63(d,2H,J=7.2Hz),1.29(t,6H,J=7.1Hz)。
Figure BDA0001934211350000181
Step 6: compound 1f was dissolved in 1, 4-dioxane, followed by addition of an appropriate amount of 6N hydrochloric acid and refluxing overnight. After the reaction is finished, cooling the reaction liquid to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain 1g of the compound.1H NMR(400MHz,DMSO)12.19(s,1H),8.20(s,1H),7.60(s,1H),7.48(s,1H),3.86(s,3H),3.84(s,3H),3.26(t,2H,J=6.4Hz),2.60(t,2H,J=6.3Hz).
Figure BDA0001934211350000182
And 7: the compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then hydroxylamine hydrochloride (1.5eq), condensing agent TBTU (3eq), N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S1.1H NMR(400MHz,CDCl3+CD3OD)7.41(s,1H),7.22(s,1H),7.20(s,1H),3.94(s,3H),3.93(s,3H),3.11(t,2H,J=8.0Hz),2.68(t,2H,J=8.0Hz);MS(EI):309.
2. Synthesis of Compound S2
Figure BDA0001934211350000183
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of hydrazine hydrochloride (1.0eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S2.1H NMR(400MHz,DMSO)9.87(s,1H),8.19(s,1H),7.59(s,1H),7.48(s,1H),3.86(s,3H),3.83(s,3H),3.27(t,2H,J=8.0Hz),2.54(t,2H,J=8.0Hz);MS(EI):308.
3. Synthesis of Compound S3
Figure BDA0001934211350000191
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-methylhydrazine hydrochloride (1.0eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S3.1H NMR(400MHz,DMSO)9.80(s,1H),8.11(s,1H),7.53(s,1H),7.41(s,1H),3.90(s,3H),3.87(s,3H),3.22(t,2H,J=8.0Hz),3.04(s,3H),2.50(t,2H,J=8.0Hz);MS(EI):322.
4. Synthesis of Compound S4
Figure BDA0001934211350000192
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then N-methylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S4.1H NMR(400MHz,DMSO)7.99(s,1H),7.29(s,1H),7.28(s,1H),3.99(s,3H),3.96(s,3H),3.35(t,2H,J=8.0Hz),3.25(s,3H),2.93(t,2H,J=8.0Hz);MS(EI):323.
5. Synthesis of Compound S5
Figure BDA0001934211350000193
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then N-difluoromethylhydroxylamine (1.0eq), a condensing agent TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S5.1H NMR(400MHz,CDCl3)7.94(s,1H),7.25(s,1H),7.24(s,1H),7.20(m,1H),3.95(s,3H),3.93(s,3H),3.38(t,2H,J=8.0Hz),2.97(t,2H,J=8.0Hz);MS(EI):359.
6. Synthesis of Compound S6
Figure BDA0001934211350000201
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-ethylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) and reacted at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S6.1H NMR(400MHz,CDCl3)8.21(brs,1H),7.94(s,1H),7.27(s,1H),7.25(s,1H),3.99(s,3H),3.96(s,3H),3.84–3.68(m,2H),3.53–3.40(m,2H),2.98–2.70(m,2H),1.20-1.37(m,3H);MS(EI):337。
7. Synthesis of Compound S7
Figure BDA0001934211350000202
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-propylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate, and the organic phase was collectedAnd purifying by column chromatography to obtain compound S7.1H NMR(400MHz,CDCl3)8.19(brs,1H),7.91(s,1H),7.24(s,1H),7.20(s,1H),3.94(s,3H),3.90(s,3H),3.81–3.76(m,2H),3.49–3.45(m,2H),2.92–2.74(m,2H),1.35-1.29(m,2H),1.19(t,3H,J=8.0Hz);MS(EI):351.10。
8. Synthesis of Compound S8
Figure BDA0001934211350000203
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-allylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) and reacted at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S8.1H NMR(400MHz,CDCl3)8.23(brs,1H),8.01(s,1H),7.31(s,1H),7.23(s,1H),5.78(m,1H),5.23(m,2H),3.93(s,3H),3.89(s,3H),3.72(d,2H,J=6.2Hz),3.42(m,2H),2.48(m,2H);MS(EI):349.0。
9. Synthesis of Compound S9
Figure BDA0001934211350000211
Dissolving the compound S8(1eq) in 1, 4-dioxane, adding dihydroxylation mixture reagent AD-Mix- β (1.5eq) and reacting at room temperature overnight, pouring the reaction liquid into ice water after the reaction is finished, extracting with dichloromethane, collecting the organic phase and purifying by a column to obtain the compound S9.1H NMR(400MHz,CD3OD)8.31(brs,1H),8.04(s,1H),7.39(s,1H),7.25(s,1H),4.11(m,1H),3.95(s,3H),3.92(s,3H),3.86(m,2H),3.67(m,2H),3.54(m,2H),2.46(m,2H);MS(EI):383。
10. Synthesis of Compound S10
Figure BDA0001934211350000212
1g (1eq) of the compound was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-isopropylhydroxylamine hydrochloride (1.5eq)TBTU (3eq), N, N-diisopropylethylamine (5eq) at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S10.1H NMR(400MHz,CDCl3)7.89(s,1H),7.30(brs,1H),7.25(s,1H),7.23(s,1H),3.98(s,3H),3.96(s,3H),3.37(t,2H,J=6.8Hz),3.26(m,1H),2.83(t,2H,J=6.8Hz,),1.11(d,6H,J=4.0Hz);MS(EI):351.
11. Synthesis of Compound S11
Figure BDA0001934211350000221
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-cyclopropylhydroxylamine (1.0eq), the condensing agent TBTU (3eq), and N, N-diisopropylethylamine (5eq) and reacted at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S11.1H NMR(400MHz,CDCl3)7.97(s,1H),7.27(s,1H),7.26(s,1H),3.97(s,3H),3.94(s,3H),3.34(t,2H,J=8.0Hz),3.28(m,1H),2.97(t,2H,J=8.0Hz),0.94(m,4H).MS(EI):349.
12. Synthesis of Compound S12
Figure BDA0001934211350000222
1g (1eq) of the compound was dissolved in N, N-dimethylformamide, and then N-cyclohexylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S12.1H NMR(400MHz,CDCl3)7.89(s,1H),7.40(brs,1H),7.26(s,1H),7.25(s,1H),3.98(s,3H),3.96(s,3H),3.36(t,J=6.8Hz,2H),2.92(m,1H),2.82(t,J=6.8Hz,2H),1.87(d,J=12.2Hz,2H),1.75(d,J=12.6Hz,2H),1.20(m,6H).MS(EI):391.
13. Synthesis of Compound S13
Figure BDA0001934211350000223
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-benzylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) and reacted at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S13.1H NMR(400MHz,CDCl3)7.85(s,1H),7.70(brs,1H),7.39-7.30(m,5H),7.25(s,1H),7.24(s,1H),4.16(s,2H),3.98(s,3H),3.96(s,3H),3.30(t,2H,J=6.8Hz,),2.77(t,2H,J=6.8Hz),MS(EI):399.
14. Synthesis of Compound S14
Figure BDA0001934211350000231
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-difluorobenzylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S14.1H NMR(400MHz,CDCl3)7.94(s,1H),7.80(brs,1H),7.52-7.49(m,5H),7.34(s,1H),7.27(s,1H),3.96(s,3H),3.94(s,3H),3.30(t,2H,J=6.8Hz,),2.77(t,2H,J=6.8Hz),MS(EI):435.
15. Synthesis of Compound S15
Figure BDA0001934211350000232
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then fluorobenzylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S15.1H NMR(400MHz,CDCl3)7.73(s,1H),7.78(brs,1H),7.34(s,1H),7.27(s,1H),6.97(m,1H),9.89(m,1H),3.92(s,3H),3.89(s,3H),3.37(t,2H,J=6.8Hz,),2.89(t,2H,J=6.8Hz),MS(EI):453.
16. Synthesis of Compound S16
Figure BDA0001934211350000233
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by sequential addition of pyridine methylene hydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S16.1H NMR(400MHz,CDCl3)8.48(s,1H),8.14(d,1H,J=7.5Hz),7.80(s,1H),7.76(d,1H,J=7.5Hz),7.73(brs,1H),7.32(m,1H),7.28(s,1H),7.22(s,1H),4.23(s,2H),3.96(s,3H),3.94(s,3H),3.35(t,2H,J=6.8Hz,),2.79(t,2H,J=6.8Hz),MS(EI):400.
17. Synthesis of Compound S17
Figure BDA0001934211350000241
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then N-phenylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S17.1H NMR(400MHz,CDCl3)8.05(s,1H),7.85(brs,1H),7.52-7.40(m,5H),7.28(s,1H),7.23(s,1H),3.96(s,3H),3.91(s,3H),3.20(t,2H,J=6.8Hz,),2.67(t,2H,J=6.8Hz).MS(EI):385.
18. Synthesis of Compound S18
Figure BDA0001934211350000242
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then hydroxylamine sulfonate (1.0eq), condensing agent TBTU (3eq), N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction solution was poured into ice water, extracted with ethyl acetate, and collectedThe organic phase was collected and purified by column to give compound S18.1H NMR(400MHz,CDCl3)1H NMR(400MHz,CDCl3+CD3OD)12.04(brs,1H,),7.43(s,1H),7.29(s,1H),7.19(s,1H),3.96(s,3H),3.94(s,3H),3.10(t,2H,J=8.0Hz),2.69(t,2H,J=8.0Hz).MS(EI):389.
19. Synthesis of Compound S19
Figure BDA0001934211350000251
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by sequential addition of methoxyhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S19.1H NMR(400MHz,CDCl3)8.62(brs 1H),7.90(s,1H),7.24(s,2H),3.98(s,3H),3.95(s,3H),3.77(s,3H),3.40(t,J=6.5Hz,2H),2.53(brs,2H);MS(EI):323.0。
20. Synthesis of Compound S20
Figure BDA0001934211350000252
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of O-benzylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S20.1H NMR(400MHz,CDCl3)8.39(s,1H),7.89(s,1H),7.40–7.36(m,5H),7.25(s,2H),4.91(s,2H),3.98(s,3H),3.96(s,3H),3.38(brs,2H),2.50(brs,2H);MS(EI):399.0.
21. Synthesis of Compound S21
Figure BDA0001934211350000253
Dissolving the compound 1g (1eq) in N, N-dimethylformamide, and then adding N-methyl, O-Methylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N, N-diisopropylethylamine (5eq) at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S21.1H NMR(400MHz,CDCl3)7.92(s,1H),7.26(s,1H),7.25(s,1H),3.98(s,3H),3.95(s,3H),3.77(s,3H),3.35(t,2H,J=8.0Hz),3.21(s,3H),2.92(t,2H,J=8.0Hz).MS(EI):399.0.
22. Synthesis of Compound S22
Figure BDA0001934211350000261
Step 1: compound 1f (1eq) was dissolved in dichloromethane, 1.0M boron tribromide in dichloromethane (6eq) was slowly added dropwise at-78 ℃ and reacted at-78 ℃ for half an hour, followed by moving to room temperature for about half an hour. After the reaction, water is slowly added dropwise to quench the reaction product in an ice bath, and then the reaction product is extracted by dichloromethane and purified by a column to obtain the compound 22 a.1H NMR(400MHz,CDCl3)7.93(s,1H),7.25(s,1H),7.18(s,1H),4.71(t,J=7.3Hz,1H),4.10(dq,J=13.3,6.8Hz,4H),3.11(qd,J=17.3,7.0Hz,2H),1.29(t,J=7.1Hz,6H)。
Figure BDA0001934211350000262
Step 2: compound 22a (1eq) was dissolved in N, N-dimethylformamide, and triethylene glycol di-p-toluenesulfonate (1eq) and potassium carbonate (4.5eq) were added in this order, reacted at 90 ℃ overnight, cooled to room temperature, poured into ice water, extracted with ethyl acetate, and purified by column chromatography to give compound 22 b.1H NMR(400MHz,CDCl3)8.03(s,1H),7.46(s,1H),7.35(s,1H),4.77(t,J=7.3Hz,1H),4.23(dd,J=8.3,4.3Hz,4H),4.16(d,J=7.1Hz,2H),3.95–3.91(m,2H),3.90–3.86(m,2H),3.82(s,4H),3.08(d,J=7.2Hz,2H),1.22(t,J=7.1Hz,6H)。
Figure BDA0001934211350000263
And step 3: will combine withThe substance 22b was dissolved in 1, 4-dioxane, and then an appropriate amount of 6N hydrochloric acid was added thereto, followed by refluxing overnight. After the reaction is finished, cooling the reaction liquid to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain the compound 22 c.1H NMR(500MHz,MeOD)7.83(s,1H),7.37(s,1H),7.28(s,1H),4.15(dd,J=8.0,5.5Hz,4H),3.87–3.82(m,2H),3.81–3.78(m,2H),3.73(s,4H),3.23(t,J=6.3Hz,2H),2.69(t,J=6.4Hz,2H)。
Figure BDA0001934211350000271
And 4, step 4: compound 22c (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-isopropylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S22.1H NMR(400MHz,CDCl3)7.89(s,1H),7.35(brs,1H),7.29(s,1H),7.24(s,1H),4.17(dd,J=8.0,5.5Hz,4H),3.89–3.83(m,2H),3.85–3.80(m,2H),3.76(s,4H),3.35(t,2H,J=6.8Hz),3.21(m,1H),2.79(t,2H,J=6.8Hz,),1.13(d,6H,J=4.0Hz).MS(EI):437.
23. Synthesis of Compound S23
Figure BDA0001934211350000272
Step 1: compound 1f (1eq) was dissolved in dichloromethane; then, anhydrous aluminum chloride (5eq) was added thereto, and the mixture was reacted at 45 ℃ for 8 hours, cooled to room temperature, quenched with an appropriate amount of 1N hydrochloric acid, extracted with dichloromethane, and purified by column chromatography to give compound 23 a.1HNMR(400MHz,CDCl3)7.90(s,1H),7.32(s,1H),7.23(s,1H),6.09(s,1H),4.24(pd,J=7.7,3.6Hz,4H),4.07(t,J=7.2Hz,1H),3.98(s,3H),3.62(d,J=7.2Hz,2H),1.29(t,J=7.1Hz,6H)。
Figure BDA0001934211350000273
Step 2: compound 23a (1eq) was dissolved in N, N-dimethylformamide, andn- (3-chloropropyl) morpholine (1eq), potassium carbonate (3eq) and 4-dimethylaminopyridine (0.01eq) were added in this order and reacted at 80 ℃ for 3 hours, cooled to room temperature, poured into ice water, extracted with ethyl acetate and purified by column chromatography to give compound 23 b.1H NMR(400MHz,CDCl3)7.91(s,1H),7.29(s,1H),7.24(s,1H),4.29–4.21(m,4H),4.18(t,J=6.5Hz,2H),4.07(t,J=7.2Hz,1H),3.93(s,3H),3.75-3.73(m,4H),3.62(d,J=7.2Hz,2H),2.58-2.55(m,2H),2.58-2.47(m,4H),2.15–2.04(m,2H),1.29(t,J=7.1Hz,6H)。
Figure BDA0001934211350000281
And step 3: compound 23b was dissolved in 1, 4-dioxane, followed by the addition of the appropriate amount of 6N hydrochloric acid and refluxing overnight. After the reaction is finished, cooling the reaction solution to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain the compound 23 c.1H NMR(400MHz,DMSO)12.20(s,1H),8.21(s,1H),7.63(s,1H),7.51(s,1H),4.17(t,J=5.7Hz,2H),3.99(d,J=12.7Hz,2H),3.85(s,3H),3.75(t,J=12.2Hz,2H),3.50(d,J=12.0Hz,2H),3.29–3.23(m,4H),3.10(dd,J=21.6,9.7Hz,2H),2.60(t,J=6.2Hz,2H),2.28–2.20(m,2H)。
Figure BDA0001934211350000282
And 4, step 4: compound 23c (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-isopropylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S23.1H NMR(400MHz,CDCl3)8.27(s,1H),7.66(s,1H),7.53(s,1H),4.14(t,J=5.7Hz,2H),3.92(d,J=12.7Hz,2H),3.89(s,3H),3.77(t,J=12.2Hz,2H),3.56(d,J=12.0Hz,2H),3.34–3.26(m,4H),3.19(m,1H),3.12(dd,J=21.6,9.7Hz,2H),2.63(t,J=6.2Hz,2H),2.26–2.18(m,2H),1.10(d,6H,J=4.0Hz).MS(EI):464.
24. Synthesis of Compound S24
Figure BDA0001934211350000283
Step 1: compound 1f (1eq) was dissolved in acetonitrile and the selective fluoro reagent was added at room temperature and allowed to react overnight. After the reaction is finished, the mixture is quenched by saturated sodium bicarbonate solution, extracted by ethyl acetate, collected and purified by a column to obtain the compound 24 a.1H NMR(400MHz,CDCl3)8.03(s,1H),7.08(s,1H),4.25(ddq,J=14.8,7.5,3.7Hz,4H),4.07(t,1H,J=7.2Hz),3.99(s,3H),3.97(s,3H),3.64(d,2H,J=7.2Hz),1.30(t,6H,J=7.1Hz)。
Figure BDA0001934211350000291
Step 2: compound 24a was dissolved in 1, 4-dioxane, followed by addition of the appropriate amount of 6N hydrochloric acid and refluxing overnight. After the reaction is finished, cooling the reaction liquid to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain a compound 24 b.1H NMR(400MHz,DMSO)12.19(s,1H),8.32(s,1H),7.58(s,1H),3.92(s,3H),3.86(s,3H),3.32(t,2H,J=6.4Hz),2.59(t,2H,J=6.3Hz)。
Figure BDA0001934211350000292
And step 3: the compound 24b (1eq) was dissolved in N, N-dimethylformamide, and then hydroxylamine hydrochloride (1.0eq), a condensing agent TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S24.1H NMR(400MHz,DMSO)8.35(s,1H),7.56(s,1H),3.95(s,3H),3.89(s,3H),3.32(t,2H,J=6.4Hz),2.61(t,2H,J=6.3Hz).MS(EI):327.
25. Synthesis of Compound S25
Figure BDA0001934211350000293
Compound 24b (1eq) was dissolved in N, N-dimethylformamide, followed byN-isopropylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added and reacted at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S25.1H NMR(400MHz,CDCl3)8.37(s,1H),7.53(s,1H),3.94(s,3H),3.90(s,3H),3.32(t,2H,J=6.4Hz),3.24(m,1H),2.61(t,2H,J=6.3Hz),1.13(d,J=4.0Hz,6H).MS(EI):369.
26. Synthesis of Compound S26
Figure BDA0001934211350000301
Compound 24b (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-benzylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S26.1H NMR(400MHz,CDCl3)8.40(s,1H),7.57(s,1H),,7.40-7.35(m,5H),4.02(s,2H),3.99(s,3H),3.96(s,3H),3.24(t,2H,J=6.8Hz,),2.2(t,2H,J=6.8Hz).MS(EI):417.
27. Synthesis of Compound S27
Figure BDA0001934211350000302
Dissolving the compound S10(1eq) in toluene, adding Lawson reagent (1.5eq), heating to 120 ℃, refluxing for about 10 minutes, cooling to room temperature, quenching with saturated sodium bicarbonate solution under ice bath, extracting with ethyl acetate, and separating and purifying by column to obtain the compound S27.1H NMR(400MHz,CDCl3)7.8(s,1H),7.23(s,1H),7.19(s,1H),3.99(s,3H),3.98(s,3H),3.34(t,2H,J=6.8Hz),3.22(m,1H),2.87(t,2H,J=6.8Hz,),1.10(d,6H,J=4.0Hz);MS(EI):367.
28. Synthesis of Compound S28
Figure BDA0001934211350000311
Step 1, dissolving 1eq of compound 28a (prepared according to the synthesis method of 1C) in dichloromethane, adding NBS (1.1eq), continuously stirring the reaction solution for 6 hours, detecting by T L C that the reaction is finished, then removing the solvent by spinning, and purifying by a column to obtain compound 28 b;
Figure BDA0001934211350000312
step 2: compound 28b (1eq) was dissolved in tetrahydrofuran, and methylmagnesium bromide (1.0M in THF,3eq) was added slowly at-10 deg.C, followed by moving to room temperature for 3 hours. Quenching with 1N HCl, extracting with ethyl acetate, and separating by column chromatography to obtain compound 28 c;
Figure BDA0001934211350000313
and step 3: compound 28c (1eq) was dissolved in chloroform, pyridine tribromide (1.1eq) was added in two portions at 0 ℃ and then allowed to warm to room temperature for reaction overnight. After the reaction was complete, the solvent was spin dried, the resulting solid was stirred in water for about 10 minutes, filtered, and the resulting solid was recrystallized from ethyl acetate to give compound 28 d.
Figure BDA0001934211350000314
And 4, step 4: potassium carbonate (0.65eq) was suspended in DMF, then diethyl malonate (1.05eq) was added at room temperature, after stirring for 3 hours compound 28d (1eq) was added, and the temperature was raised to 110 ℃ for 4 hours reaction. After the reaction is finished, cooling the reaction liquid to room temperature, quenching the reaction liquid by using water, extracting the reaction liquid by using ethyl acetate, and separating the extraction liquid by using column chromatography to obtain a compound 28 e.1H NMR(400MHz,CDCl3)7.99(s,1H),7.30(s,1H),7.28(s,1H),4.32–4.20(m,4H),4.11(t,1H,J=7.2Hz),3.97(s,3H)3.66(d,2H,J=7.2Hz),1.32(t,6H,J=7.1Hz)。
Figure BDA0001934211350000315
And 5: the compound 28e is dissolved in 1 and,4-dioxane, then adding a proper amount of 6N hydrochloric acid, and refluxing overnight. After the reaction is finished, cooling the reaction liquid to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain a compound 28 f.1H NMR(400MHz,DMSO)12.24(s,1H),8.34(s,1H),7.68(s,1H),7.50(s,1H),3.90(s,3H),3.32(t,2H,J=6.4Hz),2.63(t,2H,J=6.3Hz).
Figure BDA0001934211350000321
Step 6: compound 28f (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of hydroxylamine hydrochloride (1.5eq), condensing agent TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S28.1H NMR(400MHz,CDCl3+CD3OD)7.50(s,1H),7.29(s,1H),7.22(s,1H),3.96(s,3H),3.28(m,1H),3.14(t,2H,J=8.0Hz),2.71(t,2H,J=8.0Hz),1.14(d,6H,J=4.0Hz);MS(EI):399.
29. Synthesis of Compound S29
Figure BDA0001934211350000322
S28(1eq), alkenyl borate (1.1eq) and Pd as a catalyst2(dba)3(0.05eq), ligand X-Phos (0.05eq) and Cs2CO3(1eq) in dry toluene, reaction at 110 ℃ for 12 hours, T L C detection reaction after completion, adding ethyl acetate dilution, saturated salt water washing, organic phase with anhydrous Na2SO4Drying, concentrating under reduced pressure and purifying by column chromatography to obtain compound S29.1H NMR(400MHz,CDCl3+CD3OD)8.04(s,1H),7.65(s,1H),7.36(s,1H),6.45(m,1H),5.28-5.43(m,2H),3.94(s,3H),3.23(m,1H),3.17(t,2H,J=8.0Hz),2.67(t,2H,J=8.0Hz),1.10(d,6H,J=4.0Hz);MS(EI):347.
30. Synthesis of Compound S30
Figure BDA0001934211350000331
Step 1: compound 22a (1eq) was dissolved in N, N-dimethylformamide, dibromomethane (1eq) and potassium carbonate (4.5eq) were added in sequence, reacted at 90 ℃ overnight, cooled to room temperature, poured into ice water, extracted with ethyl acetate and purified by column chromatography to give compound 30 a.
Figure BDA0001934211350000332
Step 2: compound 30a was dissolved in 1, 4-dioxane, followed by addition of the appropriate amount of 6N hydrochloric acid and refluxing overnight. After the reaction is finished, cooling the reaction liquid to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain a compound 30 b.
Figure BDA0001934211350000333
And step 3: compound 30b (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-isopropylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S30.1H NMR(400MHz,CDCl3)7.91(s,1H),7.43(brs,1H),7.30(s,1H),7.22(s,1H),6.01(s,2H),3.32(t,2H,J=6.8Hz),3.21(m,1H),2.75(t,2H,J=6.8Hz,),1.13(d,6H,J=4.0Hz).MS(EI):335.
31. Synthesis of Compound S31
Figure BDA0001934211350000334
Step 1: compound 22a (1eq) was dissolved in N, N-dimethylformamide, and 1, 2-dibromoethane (1eq) and potassium carbonate (4.5eq) were added in this order, reacted at 90 ℃ overnight, cooled to room temperature, poured into ice water, extracted with ethyl acetate, and purified by column chromatography to give compound 31 a.
Figure BDA0001934211350000341
Step 2: compound 31a is dissolved in 1, 4-dioxane, followed by addition of the appropriate amount of 6N hydrochloric acid and refluxing overnight. After the reaction is finished, cooling the reaction solution to room temperature, spin-drying the solvent, and recrystallizing with ethanol to obtain the compound 31 b.
Figure BDA0001934211350000342
And step 3: compound 31b (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-isopropylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S31.1H NMR(400MHz,CDCl3)7.90(s,1H),7.32(s,1H),7.23(s,1H),4.21(s,4H),3.34(t,2H,J=6.8Hz),3.22(m,1H),2.72(t,2H,J=6.8Hz,),1.10(d,6H,J=4.0Hz).MS(EI):349.
32. Synthesis of Compound S32
Figure BDA0001934211350000343
Compound 1g (1eq) was dissolved in N, N-dimethylformamide, followed by the sequential addition of N-methyl acetate hydroxylamine hydrochloride (1.5eq), TBTU (3eq), N-diisopropylethylamine (5eq) and reaction at room temperature overnight. After the reaction is finished, pouring the reaction solution into ice water, extracting with ethyl acetate, collecting an organic phase, and purifying by a column to obtain an intermediate; the intermediate (1eq) was dissolved in tetrahydrofuran: adding lithium hydroxide monohydrate (3eq) into a mixed solvent of water (1: 1), reacting at room temperature for half an hour, adjusting the pH to 5-6 with 1N hydrochloric acid, extracting with chloroform, collecting an organic phase, and purifying by a column to obtain a compound S32.1H NMR(400MHz,CDCl3)7.98(s,1H),7.32(s,1H),7.21(s,1H),4.10(s,2H),3.99(s,3H),3.97(s,3H),3.31(t,2H,J=8.0Hz),2.87(t,2H,J=8.0Hz).MS(EI):367.
33. Synthesis of Compound S33
Figure BDA0001934211350000351
The compound (1 g, 1eq) was dissolved in N, N-dimethylformamide, and then O-hydroxyethylhydroxylamine hydrochloride (1.5eq), TBTU (3eq), and N, N-diisopropylethylamine (5eq) were added in this order to react at room temperature overnight. After the reaction, the reaction mixture was poured into ice water, extracted with ethyl acetate, and the organic phase was collected and purified by column chromatography to obtain compound S33.1H NMR(400MHz,CDCl3)7.88(s,1H),7.32(s,1H),7.28(s,1H),3.97(s,3H),3.96(s,3H),3.72(t,2H,J=8.4Hz),3.54(t,2H,J=8.0Hz),3.30(t,2H,J=8.0Hz),2.64(t,2H,J=8.0Hz).MS(EI):353.
Cell screening experiment for activating interferon gene stimulating protein and promoting IFN- β expression by compound
The detection method and principle are that human THP1-Blue-ISG cell is transferred with a report system containing IFN- β, the report system can induce the expression of downstream alkaline phosphatase, when the alkaline phosphatase is secreted to the outside of the cell, OD650 can be measured by chromogenic reaction to reflect the content of the alkaline phosphatase, and after the cell is added with a compound, if interferon gene stimulation protein is activated, the expression of IFN- β can be promoted, and then the increase of downstream alkaline phosphorylation secretion and the increase of the absorbance of the chromogenic reaction are promoted.
The test method comprises the following steps:
1. add Compound 20. mu. L of a physiological saline diluted compound was added to each well of a 96-well cell culture plate
The concentration of the compound was 100. mu.M, 2 replicate wells were set, the positive control compound was ADU-S100, the concentration was 100. mu.M, and 20. mu. L was added to a normal saline solution containing 1% DMSO without drug addition.
2. Adding cells, THP1-Blue-ISG cell counting, adjusting cell concentration to 5 × 105and/M L, adding 180 mu l of cells into each well for incubation, so that the content of 200 mu L in each test well is 0.1%, the test concentration of the compound is 10 mu M, the final concentration of ADU-S100 as a positive control compound is 10 mu M, and incubating for 24 hours for detection, and adding 180 mu L culture solution into the blank group.
3. Detecting a color development reaction: after 24 hours, 20. mu.l of the culture medium was transferred to a new 96-well plate, 200. mu.l of color developing solution Quanti-Blue was added thereto, the plate was placed in an incubator at 37 ℃ and the OD650 value was measured after 0.5 to 2 hours.
4. Screening concentration of compound: 10 μ M.
5. And (4) analyzing results:
Figure BDA0001934211350000361
where CompundOD 650 is the OD650 value of the compound of the present invention, Blank OD650 is the OD650 value of the medium, and Control OD650 is the OD650 value of the Control group without the compound of the present invention (cells only and 0.1% DMSO).
6. And (4) evaluating the result: the activation multiple (Fold change) is more than or equal to 2.
The experimental results are as follows:
table 2 partial Compounds ability to activate human interferon gene stimulating protein in THP1 cells at 10. mu.M concentration to promote type I interferon expression
Figure BDA0001934211350000362
Where CON represents a control without compound (i.e. cells only and 0.1% DMSO).
The results show that the compound in the patent can activate interferon gene stimulating protein and promote the expression of interferon factor IFN- β at the concentration of 10uM, wherein the agonistic activity of S10, S11, S12, S13, S19, S25 and S29 is equivalent to that of the cyclic dinucleoside compound ADU-S100 in phase I clinical research, even better and has further application prospect.
Furthermore, the above compound has 2.2 to 2.5-fold enhanced agonistic activity relative to TW201817723A discloses compound 1g (i.e., compound 16 therein).
TABLE 3 ability of compounds S10 and S13 to activate human interferon gene stimulating protein in THP1-blue cells and promote type I interferon expression at different concentrations
Figure BDA0001934211350000371
Where CON represents a control without compound (i.e. cells only and 0.1% DMSO).
The above results indicate that the compounds S10 and S13 in this patent can activate interferon gene stimulating protein dose-dependently and promote the expression of interferon factor IFN- β.
TABLE 4 ability of 10. mu. M S10 and S13 to promote type I interferon expression in interferon gene-stimulated protein knock-out THP1-Dual cells
Figure BDA0001934211350000372
Where CON represents a control without compound (i.e. cells only and 0.1% DMSO).
The above results show that the compounds S10 and S13 in this patent can not promote the expression of type I interferon in THP1-Dual cells of interferon gene stimulating protein knockout, which indicates that the compounds can promote the expression of interferon factor IFN- β by activating interferon gene stimulating protein.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

1. A compound shown as a formula I, or an isomer, a prodrug, a solvate, a hydrate or a pharmaceutically acceptable salt thereof,
Figure FDA0001934211340000011
wherein the content of the first and second substances,
R1and R2Independently selected from the group consisting of substituted or unsubstituted: H. halogen, amino, hydroxyl, carboxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C1-C6 alkoxyA group, C3-C8 cycloalkoxy, 3-8 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, O, S, 3-8 membered heterocycloalkoxy containing 1-3 heteroatoms selected from N, O, S, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S; or R1And R2Together with the carbon atom to which they are attached form a 5-14 membered heterocyclyl;
the R is1And R2The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: halogen, amino, hydroxyl, carboxyl, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S;
X1and X2Independently selected from the group consisting of: H. d, halogen;
X3selected from the group consisting of: NH, O, S, Se;
X4selected from the group consisting of: o, NR5、S;
n is an integer selected from 0, 1,2, 3, 4, 5, 6, 7, 8;
R3independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic acid group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-to 10-membered heteroaryl containing 1 to 3 heteroatoms selected from N, O, S, 3-to 8-membered heterocycloalkyl containing 1 to 3 heteroatoms selected from N, O, S;
R4and R5Independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic acid group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-to 10-membered heteroaryl containing 1 to 3 heteroatoms selected from N, O, S, 3-to 8-membered heterocycloalkyl containing 1 to 3 heteroatoms selected from N, O, S;
the R is3、R4And R5The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: hydroxyl, halogen, carboxyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C12 alkoxy, C3-C8 cycloalkyl, C3-C8 cycloalkoxy, C6-C10 aryl, halogenated C6-C10 aryl, 1-3-10 membered heteroaryl of 3 heteroatoms selected from N, O, S.
2. The compound of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof,
X1and X2Independently selected from the group consisting of: H. d, fluorine;
X3selected from the group consisting of: o, S, respectively;
n is an integer selected from 1,2, 3;
R1and R2Independently selected from the group consisting of substituted or unsubstituted: H. halogen, C2-C6 alkenyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocycloalkoxy containing 1-3 heteroatoms selected from N, O, S; or R1And R2Together with the carbon atom to which they are attached form a 5-14 membered heterocycloalkoxy;
the R is1And R2The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: halogen, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S.
3. The compound of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof,
X4selected from the group consisting of: o, NR5
n is an integer selected from 1,2, 3;
R3independently selected from the group consisting of substituted or unsubstituted: H. C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S, 3-8 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, O, S;
R4and R5Independently selected from the group consisting of substituted or unsubstituted: H. carboxyl, sulfonic group, phosphoryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, containing 1-3 groups selected from N, O3-10 membered heteroaryl of a heteroatom of S, 3-8 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, O, S;
the R is3、R4And R5The substituent(s) in (b) means being independently substituted with one or more substituents selected from the group consisting of: hydroxy, halogen, carboxy, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C12 alkoxy, C3-C8 cycloalkoxy, C2-C6 alkenyl, C6-C10 aryl, haloC 6-C10 aryl, 3-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O, S.
4. The compound of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof, selected from the group consisting of:
Figure FDA0001934211340000031
Figure FDA0001934211340000041
5. a method of preparing a compound of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof, selected from the group consisting of:
the method one comprises the following steps:
Figure FDA0001934211340000042
a-1) reacting a compound of formula 1 with a compound of formula 2 or a hydrochloride thereof in the presence of a condensing agent to obtain a compound of formula 3;
Figure FDA0001934211340000043
a-2) optionally reacting the compound of formula 3 with a lawson's reagent to provide a compound of formula 4;
wherein,R1、R2、R3、R4、X1、X2、X4N is as defined in claim 1;
the second method comprises the following steps:
Figure FDA0001934211340000051
b-1) reacting the compound of formula 5 with the compound of formula 6 or a hydrochloride thereof in the presence of a condensing agent to obtain a compound of formula 7;
Figure FDA0001934211340000052
b-2) optionally subjecting the compound of formula 7 to a coupling reaction to obtain a compound of formula 8;
wherein R is1、R2、R3、R4、X1、X2、X3、X4N is as defined in claim 1, and R2Is not a halogen.
6. A pharmaceutical composition, comprising:
(i) one or more therapeutically effective amounts of a compound of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof; and
(ii) a pharmaceutically acceptable carrier.
7. Use of a compound according to claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 6 for the preparation of a formulation for the prophylaxis and/or treatment of a disease associated with type I interferon.
8. The use according to claim 7, wherein the type I interferon related diseases are selected from the group consisting of: breast cancer, ovarian cancer, liver cancer, melanoma, prostate cancer, colon cancer, and gastric cancer.
9. An interferon gene stimulating protein agonist comprising one or more compounds of claim 1, or an isomer, prodrug, solvate, hydrate or pharmaceutically acceptable salt thereof.
10. A method for preventing and/or treating a disease associated with type I interferon, comprising the steps of: administering to a patient in need thereof a therapeutically effective amount of one or more compounds of claim 1, or isomers, prodrugs, solvates, hydrates thereof, or pharmaceutically acceptable salts thereof, or a pharmaceutical composition of claim 6.
CN201910002427.8A 2019-01-02 2019-01-02 Benzothiophene compounds, and pharmaceutical composition and application thereof Active CN111393404B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910002427.8A CN111393404B (en) 2019-01-02 2019-01-02 Benzothiophene compounds, and pharmaceutical composition and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910002427.8A CN111393404B (en) 2019-01-02 2019-01-02 Benzothiophene compounds, and pharmaceutical composition and application thereof

Publications (2)

Publication Number Publication Date
CN111393404A true CN111393404A (en) 2020-07-10
CN111393404B CN111393404B (en) 2023-02-17

Family

ID=71418812

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910002427.8A Active CN111393404B (en) 2019-01-02 2019-01-02 Benzothiophene compounds, and pharmaceutical composition and application thereof

Country Status (1)

Country Link
CN (1) CN111393404B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112898286A (en) * 2021-01-28 2021-06-04 中国药科大学 Benzothiophene compound or pharmaceutically acceptable salt and isomer thereof, and preparation method, pharmaceutical composition and application thereof
CN115197207A (en) * 2021-04-09 2022-10-18 上海交通大学 Tetrahydro-gamma-carboline derivatives, and pharmaceutical composition and application thereof
CN115197206A (en) * 2021-04-09 2022-10-18 上海交通大学 Indole 3-substituted tetrahydro-gamma-carboline compound, and pharmaceutical composition and application thereof
CN115772154A (en) * 2021-09-08 2023-03-10 上海交通大学 Deuterium-substituted benzothiophene derivative and preparation and application thereof
US11939343B2 (en) 2019-08-02 2024-03-26 Mersana Therapeutics, Inc. Sting agonist compounds and methods of use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001070675A2 (en) * 2000-03-24 2001-09-27 Methylgene, Inc. Inhibitors of histone deacetylase
US20050119305A1 (en) * 2001-03-21 2005-06-02 Masao Naka Il-6 production inhibitors
WO2006117549A1 (en) * 2005-05-05 2006-11-09 Chroma Therapeutics Ltd Enzyme inhibitors
WO2015085860A1 (en) * 2013-12-10 2015-06-18 广州康缔安生物科技有限公司 Heterocyclic hydroxamic acid compound, and pharmaceutical composition and application thereof
US20180093964A1 (en) * 2016-10-04 2018-04-05 Merck Sharp & Dohme Corp. BENZO[b]THIOPHENE COMPOUNDS AS STING AGONISTS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001070675A2 (en) * 2000-03-24 2001-09-27 Methylgene, Inc. Inhibitors of histone deacetylase
US20050119305A1 (en) * 2001-03-21 2005-06-02 Masao Naka Il-6 production inhibitors
WO2006117549A1 (en) * 2005-05-05 2006-11-09 Chroma Therapeutics Ltd Enzyme inhibitors
WO2015085860A1 (en) * 2013-12-10 2015-06-18 广州康缔安生物科技有限公司 Heterocyclic hydroxamic acid compound, and pharmaceutical composition and application thereof
US20180093964A1 (en) * 2016-10-04 2018-04-05 Merck Sharp & Dohme Corp. BENZO[b]THIOPHENE COMPOUNDS AS STING AGONISTS

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
COLUMBUS, OHIO, US REGISTRY[ONLINE]: "STN检索报告", 《STN REGISTRY》 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11939343B2 (en) 2019-08-02 2024-03-26 Mersana Therapeutics, Inc. Sting agonist compounds and methods of use
CN112898286A (en) * 2021-01-28 2021-06-04 中国药科大学 Benzothiophene compound or pharmaceutically acceptable salt and isomer thereof, and preparation method, pharmaceutical composition and application thereof
CN115197207A (en) * 2021-04-09 2022-10-18 上海交通大学 Tetrahydro-gamma-carboline derivatives, and pharmaceutical composition and application thereof
CN115197206A (en) * 2021-04-09 2022-10-18 上海交通大学 Indole 3-substituted tetrahydro-gamma-carboline compound, and pharmaceutical composition and application thereof
CN115197206B (en) * 2021-04-09 2024-03-22 上海交通大学 Indole 3-substituted tetrahydro-gamma-carboline compound, pharmaceutical composition and application thereof
CN115197207B (en) * 2021-04-09 2024-03-22 上海交通大学 Tetrahydro-gamma-carboline derivatives, pharmaceutical composition and application thereof
CN115772154A (en) * 2021-09-08 2023-03-10 上海交通大学 Deuterium-substituted benzothiophene derivative and preparation and application thereof
WO2023036195A1 (en) * 2021-09-08 2023-03-16 上海交通大学 Deuterium-substituted benzothiophene derivative, and preparation and use thereof
CN115772154B (en) * 2021-09-08 2024-04-30 上海交通大学 Deuterium-substituted benzothiophene derivatives, preparation and application thereof

Also Published As

Publication number Publication date
CN111393404B (en) 2023-02-17

Similar Documents

Publication Publication Date Title
CN111393404B (en) Benzothiophene compounds, and pharmaceutical composition and application thereof
EP4265620A1 (en) Novel camptothecin derivative, composition containing same, and use thereof
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN112457308A (en) Novel tricyclic aromatic heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
CN107709312B (en) Novel antimalarial drug
JPH03505204A (en) Tricyclic 3-oxo-propanenitrile derivative and method for its preparation
CN111393405B (en) Fluorine-containing substituted benzothiophene compounds, and pharmaceutical composition and application thereof
JP7209723B2 (en) Nucleoside Cyclic Phosphate Ester Compounds and Applications of Gemcitabine Prodrugs Based on Hepatic Delivery
CN115197207B (en) Tetrahydro-gamma-carboline derivatives, pharmaceutical composition and application thereof
CN105985349B (en) Seven-membered ring berberine analogue, and pharmaceutical composition, preparation method and application thereof
EP3369740B1 (en) New cytidine derivative dimers and applications thereof
CN111689975B (en) Isoquinoline derivatives and preparation and application thereof
CN116848124A (en) Toxin molecules suitable for antibody-drug conjugates
CN115772154B (en) Deuterium-substituted benzothiophene derivatives, preparation and application thereof
TWI831325B (en) Naphthyridine derivatives as ATR inhibitors and preparation methods thereof
CN117800979A (en) Tetrahydrobenzofuran [2,3-c ] pyridine kinase inhibitor and preparation method and application thereof
CN117088887A (en) SHP2 inhibitors and uses thereof
CN116253747A (en) Novel homocamptothecin derivative, composition containing same and application thereof
CN116693515A (en) GST protein inhibitor, preparation method thereof, pharmaceutical composition and application thereof
CN115710255A (en) Benzo-heterocycle-substituted urea derivative and preparation and application thereof
CN117384162A (en) Selective HER2 inhibitors
CN115433175A (en) Quinoline and quinazoline compounds containing alkynyl substitution, preparation and application thereof
CN117222648A (en) Fused ring compounds as Wee-1 inhibitors, process for their preparation and their use
CN117658983A (en) Selective PARP1 inhibitors
CN111732585A (en) Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant