CN114907267A - Pharmaceutical composition for resisting tumor - Google Patents

Pharmaceutical composition for resisting tumor Download PDF

Info

Publication number
CN114907267A
CN114907267A CN202110184609.9A CN202110184609A CN114907267A CN 114907267 A CN114907267 A CN 114907267A CN 202110184609 A CN202110184609 A CN 202110184609A CN 114907267 A CN114907267 A CN 114907267A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
group
alkyl
amino
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
CN202110184609.9A
Other languages
Chinese (zh)
Inventor
杨财广
董泽
徐洪蛟
王传辉
黄悦
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 CN202110184609.9A priority Critical patent/CN114907267A/en
Priority to PCT/CN2022/075561 priority patent/WO2022166990A1/en
Publication of CN114907267A publication Critical patent/CN114907267A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4409Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 4, e.g. isoniazid, iproniazid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • 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
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/38Radicals substituted by singly-bound nitrogen atoms having only hydrogen or hydrocarbon radicals attached to the substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings

Abstract

The invention provides a 2- (substituted phenyl hetero) aromatic formic acid FTO inhibitor, a preparation method and application thereof, and particularly discloses a compound shown as the following formula (I), and application of a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof. The compounds can be used in combination with immune checkpoint inhibitors for the treatment of solid tumors, such as melanoma, lung, colon, renal, pancreatic, lung, osteosarcoma, and like cancers.

Description

Anti-tumor pharmaceutical composition
Technical Field
The invention relates to the field of pharmaceutical compounds, and particularly provides application of an FTO inhibitor shown as a formula (I) and an immune checkpoint inhibitor in combination to treatment of solid tumors.
Background introduction
One of the reasons for cancer development is that tumors can weaken and evade immune T cells, e.g., tumors can express aggressive molecules on the cell surface, binding T cells and thereby inhibiting the killing activity of T cells, these molecules being referred to as immune checkpoints, e.g., PD-1 and CTLA-4. The inhibition of immune checkpoint molecules and thus the modulation of the body's immune system is central to tumor immunotherapy.
In recent years, tumor immunotherapy has made a breakthrough. Tumor immunotherapy stimulates the recognition of the immune system by modulating T cell receptor signaling or using monoclonal antibodies to natural biomolecules and associated tumor antigens. For example, the anti-PD-1/PD-L1 antibody, an immune checkpoint inhibitor, shows better efficacy in a variety of advanced solid tumors, such as melanoma, renal carcinoma, and the like, with an objective effective rate (ORR) of about 10% to 40% among different solid tumors, with the highest (about 36% to 53%) among malignant melanoma. However, many cancer patients do not achieve the expected therapeutic effect with anti-PD-1/PD-L1 antibody alone, especially for patients with advanced colorectal cancer. It is therefore imperative to find small molecule compounds that can be used in combination with the PD-1 or PD-L1 antibodies and enhance their therapeutic efficacy.
Targeting m 6 A modified small molecule inhibitors provide a new idea for these troubles. m is a unit of 6 The a modification affects the T cell immune response specific for tumor antigens by regulating the lysosomal cathepsin translation efficiency of dendritic cells. As m 6 A is the most important demethylase FTO, and its inhibitors can change m in tumor cells 6 The abundance of A, thereby achieving the purpose of improving the T cell immune response efficiency of tumor antigen specificity.
The invention provides an FTO inhibitor, which is combined with an immune checkpoint inhibitor PD-1 or PD-L1 antibody to achieve the aim of resisting tumors.
Disclosure of Invention
The invention aims to provide a medicine combination containing FTO inhibitor, which is suitable for treating tumors.
In a first aspect of the invention, there is provided a compound of formula (I), and the use of a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof,
Figure BDA0002942540170000021
wherein the content of the first and second substances,
A 1 、A 2 、A 3 、A 4 each independently is CR' or N;
m is selected from the group consisting of: CR' 2 NH, O or S;
r' is selected from the group consisting of: H. halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, and substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl, substituted or unsubstituted C 1 -C 6 Amido, substituted or unsubstituted C 2 -C 12 Ester group, or substituted or unsubstituted C 1 -C 10 Alkyl, or substituted or unsubstituted C3-C10 enamide;
x has a structure represented by the following formula: carboxy, O-substituted or unsubstituted hydroxamic acid group, substituted or unsubstituted C 2 -C 12 Ester group, substituted or unsubstituted amide group (C (O) NH 2 ) Substituted or unsubstituted 3-12 membered heterocyclyl;
y is selected from the group consisting of: substituted or unsubstituted C 6 -C 12 Substituted or unsubstituted 3-12 membered heterocyclyl;
R a 、R b 、R c 、R d each independently selected from the group consisting of: H. halogen, -OH, CN, NO 2 、NH 2 Substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 An alkoxy group;
the substituent means that one or more hydrogen atoms on the group are substituted with a substituent selected from the group consisting of: halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxy radicalCarbonyl group, C 1 -C 6 Amido, C 2 -C 12 Ester group, substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 2 -C 10 Alkenyl, substituted or unsubstituted C 2 -C 10 Alkynyl, substituted or unsubstituted C 6 -C 10 Aryl, or substituted or unsubstituted five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or C 1 -C 6 An alkylamino group; wherein, said substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 6 -C 10 The substituents of the aryl or five-or six-membered heteroaryl group are selected from the group consisting of: halogen atom, carbonyl (═ O), hydroxyl, carboxyl, C 1 -C 6 Alkoxycarbonyl, amino, C 1 -C 6 Amido, nitro, cyano, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 6 -C 10 Aryl or five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably a halogen atom, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or phenyl, 5-6 membered heterocyclyl;
wherein, the compound of formula (I) and an immune checkpoint inhibitor are used together for preparing a pharmaceutical composition for treating or preventing solid tumors.
In another preferred embodiment, the compound of formula (I) has the structure shown in formula (II) below:
Figure BDA0002942540170000031
wherein the content of the first and second substances,
A 1 、A 2 、A 3 、A 4 each independently is CR' or N;
m is selected from the group consisting of: NH or S;
r' is selected from the group consisting of: H. halogen, hydroxy, amino, nitro, cyano, substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 Alkylamino, substituted or unsubstituted C 1 -C 6 An amide;
R 0 selected from the group consisting of: hydrogen, hydroxy, substituted or unsubstituted C 1 -C 10 Alkyl, aryl, heteroaryl, and heteroaryl,
Figure BDA0002942540170000032
Wherein R is a ,R b Each independently selected from the group consisting of: hydrogen, substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 3 -C 10 Cycloalkyl (including monocyclic, polycyclic, bridged ring structures), substituted or unsubstituted C 2 -C 6 Alkenyl, substituted or unsubstituted C 2 -C 6 An alkynyl group;
m is selected from the group consisting of: 0.1, 2, 3 or 4;
R x 、R y each independently selected from the group consisting of: halogen, substituted or unsubstituted C 1 -C 4 An alkyl group;
het is selected from the group consisting of: substituted or unsubstituted C 6 -C 10 Substituted or unsubstituted 4-7 membered saturated heterocyclic group, substituted or unsubstituted 3-12 membered heterocyclic group;
the substituent means that one or more hydrogen atoms on the group are substituted with a substituent selected from the group consisting of: halogen, oxygen atom (═ O), carboxyl, hydroxyl, amino, nitro, cyano, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Amido, C 2 -C 12 An ester group; or unsubstituted or substituted by one or more halogens or C 1 -C 6 Alkyl-substituted groups selected from the group consisting of: c 1 -C 10 Alkyl radical, C 2 -C 10 Alkenyl radical, C 2 -C 10 Alkynyl, C 6 -C 10 Aryl, 5-6 membered heteroaryl, 3-12 membered heterocyclyl, 3-12 membered cycloalkyl.
In another preferred embodiment, R is 0 Has the structure shown as the following formula:
Figure BDA0002942540170000041
wherein the content of the first and second substances,
R a ,R b each independently selected from the group consisting of: hydrogen, substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 3 -C 10 Cycloalkyl (including monocyclic, polycyclic, bridged ring structures), substituted or unsubstituted C 2 -C 6 Alkenyl, substituted or unsubstituted C 2 -C 6 Alkynyl;
m is selected from the group consisting of: 1.2 or 3.
In another preferred embodiment, the Het is selected from the following group: substituted or unsubstituted pyridine, substituted or unsubstituted tetrazole, substituted or unsubstituted triazole, substituted or unsubstituted pyrimidine, substituted or unsubstituted pyrazole, substituted or unsubstituted isoxazole, substituted or unsubstituted morpholine, substituted or unsubstituted thiomorpholine, substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, substituted or unsubstituted oxetane, substituted or unsubstituted thietane, substituted or unsubstituted azetidine.
In another preferred embodiment, Het is selected from the following group:
Figure BDA0002942540170000042
wherein each R is 1 、R 2 、R 3 Or R 4 Each independently selected from the group consisting of: hydrogen, halogen, hydroxyl, amino, oxygen atom (═ O), Carboxyl (COOH), substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 6 -C 10 Aryl radicalsSubstituted or unsubstituted 5-6 membered heteroaryl, substituted or unsubstituted C 1 -C 6 Alkylamino, substituted or unsubstituted C 1 -C 6 Alkoxycarbonyl, substituted or unsubstituted C 1 -C 6 An amido group; wherein the substituents are selected from the group consisting of: F. cl, C 1 -C 6 An alkyl group.
In another preferred embodiment, each R 1 ,R 2 ,R 3 ,R 4 Each independently selected from the group consisting of: H. substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 An alkylamino group.
In another preferred embodiment, A is 2 、A 3 Each independently is CR'.
In another preferred embodiment, R' has a structure represented by the following formula:
Figure BDA0002942540170000051
wherein R' is H or substituted or unsubstituted C1-C6 alkyl.
In another preferred embodiment, the compound of formula (I) is selected from the group consisting of:
Figure BDA0002942540170000052
in another preferred embodiment, the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-L1 antibody, an anti-PD-1 antibody, or a combination thereof.
In another preferred embodiment, the solid tumor is selected from the group consisting of: melanoma, lung cancer, colon cancer, renal cancer, pancreatic cancer, lung cancer, osteosarcoma.
In a second aspect of the present invention, there is provided a compound, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, selected from the group consisting of:
Figure BDA0002942540170000061
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 repeated herein, depending on the space.
Drawings
FIG. 1 shows the results of treatment of tumor cells with compounds Dac51, Dac258, Dam62 and Dam128 of the present invention 6 The A abundance is changed in a concentration-dependent manner by the FTO inhibitor.
FIG. 2 shows the improvement of tumor growth and the change of body weight of tumor-bearing mice by the combination of the compound Dam59 of the present invention and an anti-PD-L1 antibody.
FIG. 3 shows the improvement of tumor growth and the change of body weight of tumor-bearing mice by the combination of the compound Dam159 of the present invention and an anti-PD-L1 antibody.
Detailed Description
Through long-term and intensive research, the inventor finds that the pharmaceutical composition of the compound shown as the formula (I) and the immune checkpoint inhibitor can act on solid tumors synergistically and effectively inhibit the growth of the tumors. Based on the above findings, the inventors have completed the present invention.
Term(s) for
Unless otherwise specified, the term "substituted" herein means that one or more hydrogen atoms on the group are replaced with a substituent selected from the group consisting of: c 1 ~C 10 Alkyl radical, C 3 ~C 10 Cycloalkyl radical, C 1 ~C 10 Alkoxy, halogen, hydroxy (-COOH), carboxyl (-COOH), C 1 ~C 10 Aldehyde group, C 2 ~C 10 Acyl radical, C 2 ~C 10 Ester group, amino group, phenyl group; the phenyl group includes an unsubstituted phenyl group or a substituted phenyl group having 1 to 3 substituents selected from: halogen, C 1 -C 10 Alkyl, cyano, OH, nitro, C 3 ~C 10 Cycloalkyl radical, C 1 ~C 10 Alkoxy, amino.
The term "C 1 ~C 6 The "alkyl group" means a straight or branched alkyl group having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, or the like.
The term "C 1 ~C 6 Alkoxy "means a straight or branched chain alkoxy group having 1 to 6 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy, or the like.
The term "carbonyl C 1 ~C 6 Alkyl "means a group such as" -COO-C1-C6 straight or branched chain alkyl ", for example, carbonyl-methyl, carbonyl-ethyl, carbonyl-propyl, carbonyl-isopropyl, carbonyl-butyl, carbonyl-isobutyl, carbonyl-sec-butyl, carbonyl-tert-butyl, or the like.
The term "C 6 ~C 12 The aryl group "means an aryl group having 6 to 12 carbon atoms, and includes a monocyclic or bicyclic aryl group such as phenyl, naphthyl, or the like.
The term "3-12 membered heterocyclyl" refers to a saturated or unsaturated (including aromatic) ring system substituent having one or more heteroatoms selected from O, S, N or P in a 3-12 membered ring system, such as pyridyl, thienyl, piperidinyl, or the like, preferably a 4-9 membered heterocyclyl.
The term "halogen" refers to F, Cl, Br and I.
As used herein, the terms "comprising," "including," or "including" mean that the various ingredients may be used together in a mixture or composition of the invention. Thus, the terms "consisting essentially of and" consisting of are encompassed by the term "comprising.
In the present invention, the term "pharmaceutically acceptable" ingredient refers to a substance that is suitable for use in humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response), i.e., at a reasonable benefit/risk ratio.
In the present invention, the term "effective amount" refers to an amount of a therapeutic agent that treats, ameliorates, or prevents a target disease or condition, or an amount that exhibits a detectable therapeutic or prophylactic effect. The precise effective amount for a subject will depend upon the size and health of the subject, the nature and extent of the disorder, and the therapeutic agent and/or combination of therapeutic agents selected for administration. Therefore, it is not useful to specify an exact effective amount in advance. However, for a given condition, the effective amount can be determined by routine experimentation and can be determined by a clinician.
Herein, unless otherwise specified, the term "substituted" means that one or more hydrogen atoms on a group are replaced with a substituent selected from the group consisting of: halogen, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C2-C6 acyl, unsubstituted or halogenated C1-C6 alkyl-hydroxy.
Unless otherwise specified, all occurrences of a compound in the present invention are intended to include all possible optical isomers, such as a single chiral compound, or a mixture (i.e., a racemate) of various chiral compounds. In all compounds of the present invention, each chiral carbon atom may optionally be in the R configuration or the S configuration, or a mixture of the R configuration and the S configuration.
As used herein, the term "compounds of the invention" refers to compounds of formula I. The term also includes various crystalline forms, pharmaceutically acceptable salts, hydrates or solvates of the compounds of formula (I).
In the present application, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention, which can be converted under physiological conditions or by solvolysis to a compound which is a biologically active compound of the invention. Prodrugs may not be active when administered to a subject in need thereof, but are converted in vivo to the active compounds of the invention. Prodrugs are generally rapidly converted in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. Prodrug compounds generally provide solubility, histocompatibility, or sustained release advantages in mammalian organisms.
Compounds of formula I as FTO inhibitors
In the invention, the FTO inhibitor compound and the immune checkpoint inhibitor are combined, so that a synergistic effect is achieved. The FTO inhibitor is a compound shown as the following formula (I), and pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof:
Figure BDA0002942540170000091
wherein, the first and the second end of the pipe are connected with each other,
A 1 、A 2 、A 3 、A 4 each independently is CR' or N;
m is selected from the group consisting of: CR' 2 NH, O or S;
r' is selected from the group consisting of: H. halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, and substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl, substituted or unsubstituted C 1 -C 6 Amido, substituted or unsubstituted C 2 -C 12 Ester group, or substituted or unsubstituted C 1 -C 10 Alkyl, or substituted or unsubstituted C3-C10 enamide;
x has a structure represented by the following formula: carboxy, hydroxamic acid, substituted or unsubstituted C 2 -C 12 Ester group, substituted or unsubstituted amide group (C (O) NH 2 ) Substituted or unsubstituted 3-12 membered heterocyclyl;
y is selected from the group consisting of: substituted or unsubstituted C 6 -C 12 Substituted or unsubstituted 3-12 membered heterocyclyl;
R a 、R b 、R c 、R d each independently selected from the group consisting of: H. halogen, -OH, CN, NO 2 、NH 2 Substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 An alkoxy group;
the substitution means that one or more hydrogen atoms on the group are substituted with a substituent selected from the group consisting of: halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Amido, C 2 -C 12 Ester group, substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 2 -C 10 Alkenyl, substituted or unsubstituted C 2 -C 10 Alkynyl, substituted or unsubstituted C 6 -C 10 Aryl, or substituted or unsubstituted five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or C 1 -C 6 An alkylamino group; wherein, said substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 6 -C 10 The substituents of the aryl or five or six membered heteroaryl group are selected from the group consisting of: halogen atom, carbonyl (═ O), hydroxyl group, carboxyl group, and C 1 -C 6 Alkoxycarbonyl, amino, C 1 -C 6 Amido, nitro, cyano, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 6 -C 10 Aryl or five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably halogen atom, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or phenyl, 5-6 membered heterocyclyl.
Preferred compounds have the structure as shown in the examples of the present application.
Pharmaceutical compositions and methods of administration
Because the pharmaceutical composition has excellent inhibitory activity on tumor cell proliferation, the pharmaceutical composition of the compound shown in the formula (I) and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof and the immune checkpoint inhibitor can be used for treating, preventing and relieving diseases caused by tumor cell proliferation. According to the prior art, the compounds of the invention are useful for the treatment of the following diseases: melanoma, lung cancer, colon cancer, etc.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof and a pharmacologically acceptable excipient or carrier in a safe and effective amount range. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 5-200mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"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 cellulose and its derivatives (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
Figure BDA0002942540170000101
Wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
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 a composition 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 used 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 of the compounds of the present invention for topical administration 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 compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
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 5 to 500 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.
The main advantages of the invention include:
the invention provides the use of a compound of formula (I) in combination with an immune checkpoint inhibitor for treating tumors, wherein the method can effectively inhibit the proliferation of tumor cells at a very low dosage and generates an obvious synergistic effect compared with the single use of the compound and the immune checkpoint inhibitor.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Unless otherwise indicated, percentages and parts are by weight.
Chemical example Synthesis and characterization of FTO inhibitors
EXAMPLE 1 Synthesis of Compound Dac51
Figure BDA0002942540170000121
The first step is as follows: 30g (120mmol,1.2eq) of o-iodobenzoic acid, 24g (100mmol,1.0eq) of 2, 6-dichloro-4-bromoaniline, 150mmol,1.5eq) of triethylamine and 9g (5.0mmol,0.5eq) of anhydrous copper acetate are dissolved in 500mL of DMF, and the mixture is heated to 120 ℃ under the protection of argon to react for 24 hours, and then the reaction is finished. The temperature was reduced to room temperature, an equal volume of water was added, and 300mL × 3 was extracted with ethyl acetate, washed with water, the organic phase was spin-dried, and separated by silica gel column chromatography (petroleum ether: ethyl acetate ═ 20:1 to 1:1) to obtain 9.8g of the objective product, 2- (4-bromo-2, 6-dichlorophenyl) amino) benzoic acid, which was a yellow solid.
The second step is that: 3.6g of 2- (4-bromo-2, 6-dichlorophenyl) amino) benzoic acid was dissolved in 200mL of absolute ethanol, and 20mL of concentrated sulfuric acid was added thereto under cooling in an ice water bath, followed by heating to 100 ℃ and reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, and was concentrated by rotary evaporation to remove ethanol, 100mL of water was added to the system until no bubbles were generated during neutralization with saturated sodium carbonate. The organic phase was extracted with ethyl acetate at 50 mL. times.3, and the organic phases were combined and washed with saturated brine at 10 mL. times.3. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel chromatography (petroleum ether: ethyl acetate 99:1) to obtain the objective product ethyl 2- ((4-bromo-2, 6-dichlorophenyl) amino) benzoate as a white solid (3.1 g).
The third step: 3.0g (8.0mmol,1.0eq) of ethyl 2- ((4-bromo-2, 6-dichlorophenyl) amino) benzoate, 2.13g (9.6mmol,1.2eq) of 3, 5-dimethylpyrazole-4-boronic acid pinacol ester, Pd (dppf) Cl 2 584mg (0.8mmol,0.1eq) of potassium carbonate 1.68g (12.0mmol,1.5eq) was dissolved in 160mL of a mixed solvent of 1, 4-dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, concentrated to dryness, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) to obtain 1.10g of ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoate as a target product as a white solid.
The fourth step: ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoate 1.10g (2.7mmol,1.0eq) was dissolved in a mixed solvent of 13.5mL of tetrahydrofuran and 27mL of anhydrous ethanol, and a solution of 540mg (13.5mmol,5.0eq) of sodium hydroxide in 7mL of water was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 by using 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. And (4) performing suction filtration, and washing the solid with water to obtain 930mg of the target product, namely white solid, namely 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoic acid.
The fifth step: 71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in 4.0mL of anhydrous DMF at room temperature, was added to a 10mL round bottom flask, and DIEA 77mg (0.6mmol,3.0eq) and hydroxylamine hydrochloride 17mg (0.24mmol,1.2eq) were added to the above system in this order with stirring for 5min, and the resulting mixture was reacted at 45 ℃. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly prepared and separated in reverse phase, and the separated solution was lyophilized to give the desired product, white solid Dac51(22 mg). 1 H NMR(500MHz,DMSO)δ11.31(s,1H),9.43(s,1H),7.50(d,J=7.7Hz,1H),7.45(s,2H),7.24(dd,J=11.4,4.2Hz,1H),6.76(t,J=7.5Hz,1H),6.30(d,J=8.3Hz,1H),2.23(s,6H).
EXAMPLE 2 Synthesis of Compound Dac50
Figure BDA0002942540170000141
The first and second reactions were prepared according to the synthetic procedure of Dac 51.
The third step: 3.0g (8.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-dichlorophenyl) amino) benzoate, 2.13g (9.6mmol,1.2eq) of 3-methylpyrazole-4-boronic acid pinacol ester, and Pd (dppf) Cl as a catalyst 2 584mg (0.8mmol,0.1eq) of potassium carbonate 1.68g (12.0mmol,1.5eq) was dissolved in 160mL of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL X3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL X3 of saturated brine. Anhydrous sodium sulfateThe organic phase was dried, filtered, the filtrate was concentrated, and silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) was performed to obtain 1.10g of ethyl 2- ((2, 6-dichloro-4- (3-methylpyrazole) phenyl) amino) benzoate as a white solid.
The fourth step: ethyl 2- ((2, 6-dichloro-4- (3-methylpyrazole) phenyl) amino) benzoate 1.10g (2.7mmol,1.0eq) was dissolved in a mixed solvent of tetrahydrofuran 13.5mL and absolute ethanol 27mL, and a 7mL solution of sodium hydroxide 540mg (13.5mmol,5.0eq) in water was slowly added dropwise thereto with cooling in an ice water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. And (4) carrying out suction filtration, and washing the solid with water to obtain 930mg of the target product, namely white solid 2- ((2, 6-dichloro-4- (3-methylpyrazole) phenyl) amino) benzoic acid.
The fifth step: 71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (3-methylpyrazole) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, dissolved in 4.0mL of anhydrous DMF at room temperature, stirred for 5min, and to the above system were added 77mg (0.6mmol,3.0eq) of DIEA and 17mg (0.24mmol,1.2eq) of hydroxylamine hydrochloride in this order, and the resulting mixed system was reacted at 45 ℃. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction was prepared and separated directly in reverse phase and the separated liquid was lyophilized to give Dac50(17mg) as a white solid. 1 H NMR(500MHz,DMSO)δ11.31(s,1H),9.43(s,1H),7.50(d,J=7.7Hz,1H),7.45(s,2H),7.24(dd,J=11.4,4.2Hz,1H),6.76(t,J=7.5Hz,1H),6.30(d,J=8.3Hz,1H),2.23(s,3H).
EXAMPLE 3 Synthesis of Compound Dac52
Figure BDA0002942540170000151
The first and second reactions were prepared according to the synthetic procedure of Dac 51.
The third step: 3.0g (8.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-dichlorophenyl) amino) benzoate, 2.13g (9.6mmol,1.2eq) of pyrazole-4-boronic acid pinacol ester and a catalyst Pd (dppf) Cl 2 584mg (0.8mmol,0.1eq) of potassium carbonate 1.68g (12.0mmol,1.5eq) are dissolved in diethyl ether160mL (4:1/v: v) of a mixed solvent of oxahexacyclic compound and water was heated to 100 ℃ to react for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL X3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL X3 of saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel column chromatography (petroleum ether: ethyl acetate 20:1) to obtain 1.10g of ethyl 2- ((2, 6-dichloro-4- (pyrazole) phenyl) amino) benzoate as a white solid.
The fourth step: ethyl 2- ((2, 6-dichloro-4- (pyrazole) phenyl) amino) benzoate 1.10g (2.7mmol,1.0eq) was dissolved in a mixed solvent of 13.5mL of tetrahydrofuran and 27mL of absolute ethanol, and a 7mL solution of sodium hydroxide 540mg (13.5mmol,5.0eq) in water was slowly added dropwise thereto with cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration is carried out, and the solid is washed by water to obtain 930mg of white solid 2- ((2, 6-dichloro-4- (pyrazole) phenyl) amino) benzoic acid.
The fifth step: 71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (pyrazole) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 17mg (0.24mmol,1.2eq) of hydroxylamine hydrochloride were added to the above system in this order, and the resulting system was reacted at 45 ℃. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation, and the separated solution was lyophilized to give Dac52(18mg) as a white solid. 1 H NMR(500MHz,DMSO)δ11.31(s,1H),9.43(s,1H),7.50(d,J=7.7Hz,1H),7.45(s,2H),7.24(dd,J=11.4,4.2Hz,1H),6.76(t,J=7.5Hz,1H),6.30(d,J=8.3Hz,1H),2.23(s,6H).
EXAMPLE 4 Synthesis of Compound Dam62
Figure BDA0002942540170000161
The first, second, third and fourth reactions were carried out according to the synthesis procedure of Dac 51.
The fifth step: in a 10mL circle71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoic acid and 114mg (0.3mmol,1.5eq) of HATU were added to a bottom flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, and stirred for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 14mg (0.24mmol,1.2eq) of ammonium chloride were added to the above system in this order, and the resulting mixed system was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation, and the separated solution was lyophilized to give Dam62(46mg) as a white solid. 1 H NMR(400MHz,DMSO)δ12.47(s,1H),10.13(s,1H),8.12(s,1H),7.74(d,J=7.8Hz,1H),7.51(s,1H),7.48(s,2H),7.28(t,J=7.7Hz,1H),6.78(t,J=7.5Hz,1H),6.31(d,J=8.2Hz,1H),2.29(s,3H),2.23(s,3H).
EXAMPLE 5 Synthesis of Compound Dam59
Figure BDA0002942540170000171
The first, second, third and fourth reactions were prepared according to the synthesis procedure of Dac 51.
The fifth step: 71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (3, 5-dimethylpyrazole) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in 4.0mL of anhydrous DMF at room temperature, was added to a 10mL round-bottomed flask, and DIEA 77mg (0.6mmol,3.0eq) and 3-aminopropanol 18mg (0.24mmol,1.2eq) were added to the above system in this order with stirring for 5min, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation, and the separated solution was lyophilized to give Dam59(48mg) as a white solid. 1 H NMR(400MHz,MeOD)δ7.67–7.54(m,1H),7.46–7.34(m,2H),7.25(t,J=7.8Hz,1H),6.83(t,J=7.5Hz,1H),6.41(d,J=8.3Hz,1H),3.71(t,J=6.2Hz,2H),3.51(t,J=6.9Hz,2H),2.31(s,6H),1.98–1.81(m,2H).
EXAMPLE 6 Synthesis of the Compound Dam253
Figure BDA0002942540170000172
The first and second reactions were prepared according to the synthesis procedure of Dac 51.
The third step: 3.0g (8.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-dichlorophenyl) amino) benzoate, 1.36g (9.6mmol,1.2eq) of 2, 5-dimethylisoxazole-4-boronic acid, and Pd (dppf) Cl as a catalyst 2 584mg (0.8mmol,0.1eq) of potassium carbonate 1.68g (12.0mmol,1.5eq) was dissolved in 160mL of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, concentrated to dryness, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) to obtain 1.98g of ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoate as a white solid.
The fourth step: ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoate 1.98g (4.4mmol,1.0eq) was dissolved in a mixed solvent of 36mL of tetrahydrofuran and 72mL of anhydrous ethanol, and a solution of 880mg (22mmol,5.0eq) of sodium hydroxide in 9mL of water was slowly added dropwise thereto with cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration is carried out, and the solid is washed with water, so as to obtain 1.6g of white solid 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoic acid.
The fifth step: 71mg (0.2mmol,1.0eq) of 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazol) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in 4.0mL of anhydrous DMF at room temperature, was added to a 10mL round-bottomed flask, and DIEA 77mg (0.6mmol,3.0eq) and N, N-diethylbutanediamine, 35mg (0.24mmol,1.2eq) were added to the above system in this order with stirring for 5min, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation, and the separated solution was lyophilized to give Dam253(66mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.86(s,1H),8.62(s,1H),7.66(d,J=7.6Hz,1H),7.63(s,2H),7.27(t,J=7.8Hz,1H),6.83(t,J=7.4Hz,1H),6.36(d,J=8.2Hz,1H),3.30(d,J=6.3Hz,3H),2.46(s,6H),2.42(d,J=8.4Hz,2H),2.28(s,3H),1.55(dd,J=13.8,6.8Hz,2H),1.46(d,J=6.8Hz,2H),0.94(t,J=7.1Hz,6H).
EXAMPLE 7 Synthesis of the Compound Dam128
Figure BDA0002942540170000191
The first step is as follows: 29.8g (120mmol,1.2eq) of o-iodobenzoic acid, 22.8g (100mmol,1.0eq) of 2, 6-diethyl-4-bromoaniline, 150mmol,1.5eq) of triethylamine and 9.1g (5.0mmol,0.5eq) of anhydrous copper acetate are dissolved in 360mL of DMF, and the mixture is heated to 120 ℃ under the protection of argon for 24 hours of reaction, and the reaction is finished. The temperature was reduced to room temperature, an equal volume of water was added, the mother liquor was extracted with DCM 300mL × 3, DMF was washed with water, the organic phase was spun dry and separated by silica gel column chromatography (petroleum ether: ethyl acetate: 20:1 to 1:1) to give the title product as a yellow solid 13.6 g.
The second step is that: 10.4g of 2- (4-bromo-2, 6-diethylphenyl) amino) benzoic acid was dissolved in 300mL of absolute ethanol, and 30mL of concentrated sulfuric acid was added thereto under cooling in an ice-water bath, followed by heating to 100 ℃ and reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, and was concentrated by rotary evaporation to remove ethanol, 100mL of water was added to the system until no bubbles were generated during neutralization with saturated sodium carbonate. The organic phase was extracted with ethyl acetate (50 mL. times.3), and the organic phases were combined and washed with saturated brine (10 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and subjected to silica gel column chromatography to obtain 10.2g of ethyl 2- (4-bromo-2, 6-diethylphenyl) amino) benzoate as a white solid.
The third step: 1.13g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-diethylphenyl) amino) benzoate, 508mg (3.6mmol,1.2eq) of 3, 5-dimethylisoxazole-4-boronic acid, and Pd (dppf) Cl as a catalyst 2 220mg (0.3mmol,0.1eq), 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 160mL of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, concentrated to dryness, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) to give ethyl 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoate as a white solid (650 mg).
The fourth step: 650mg (1.7mmol,1.0eq) of ethyl 2- ((2, 6-diethyl-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoate was dissolved in a mixed solvent of 4mL of tetrahydrofuran and 8mL of absolute ethanol, and a 2mL solution of 199mg (8.3mmol,5.0eq) of lithium hydroxide in water was slowly added dropwise thereto under cooling with an ice water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration was carried out, and the solid was washed with water to give 620mg of 2- ((2, 6-diethyl-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoic acid as a white solid.
The fifth step: 73mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4- (3, 5-dimethylisoxazole) phenyl) amino) benzoic acid, 73mg (0.3mmol,1.5eq) of HATU 114mg (0.3mmol,1.5eq) was added to a 10mL round bottom flask, dissolved in anhydrous DMF at room temperature was dissolved and stirred for 5min, DIEA 77mg (0.6mmol,3.0eq) and ammonium chloride 14mg (0.24mmol,1.2eq) were added to the above system in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation in an alkaline system, and the separated solution was lyophilized to obtain Dam128(52mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.84(s,1H),8.05(s,1H),7.72(dd,J=7.9,1.3Hz,1H),7.39(s,1H),7.24–7.12(m,3H),6.61(dd,J=11.0,4.0Hz,1H),6.12(d,J=8.3Hz,1H),2.57–2.51(m,3H),2.45(s,3H),2.28(s,3H),1.09(t,J=7.5Hz,6H).
EXAMPLE 8 Synthesis of Compound Dam159
Figure BDA0002942540170000201
The first and second reactions were prepared according to Dam128 synthesis procedure.
The third step: 1.13g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-diethylphenyl) amino) benzoate, 443mg (3.6mmol,1.2eq) of pyridine-4-boronic acid, and the catalyst Pd (dppf) Cl 2 220mg (0.3mmol,0.1eq), 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 160mL (4:1/v: v) of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL × 3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL × 3 saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated and separated by silica gel chromatography (petroleum ether: ethyl acetate: 20:1) to give 775mg of white solid ethyl 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoate.
The fourth step: 775mg (2.1mmol,1.0eq) of ethyl 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoate was dissolved in 4mL of a mixed solvent of tetrahydrofuran and 8mL of absolute ethanol, and 2mL of a solution of 249mg (10.5mmol,5.0eq) of lithium hydroxide in water was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 10mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration is carried out, and the solid is washed by water to obtain 693mg of white solid 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid.
The fifth step: 69mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid and 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 14mg (0.24mmol,1.2eq) of ammonium chloride were added to the above system in this order, and the resulting system was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation in an alkaline system, and the separated solution was lyophilized to obtain Dam159(52mg) as a white solid. 1 H NMR(600MHz,DMSO)δ9.87(s,1H),8.63(d,J=5.7Hz,2H),8.05(s,1H),7.75(d,J=6.0Hz,2H),7.72(d,J=7.9Hz,1H),7.62(s,2H),7.39(s,1H),7.17(t,J=7.7Hz,1H),6.63(t,J=7.5Hz,1H),6.12(d,J=8.3Hz,1H),2.55(q,J=7.5Hz,4H),1.12(t,J=7.5Hz,6H).
EXAMPLE 9 Synthesis of the Compound Dam281
Figure BDA0002942540170000211
The first, second, third and fourth reactions were carried out according to Dam 159.
The fifth step: to a 10mL round bottom flask, 69mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in 4.0mL of anhydrous DMF at room temperature, was added, followed by stirring for 5min, to the above system, 77mg (0.6mmol,3.0eq) of DIEA, 16mg (0.24mmol,1.2eq) of methylamine hydrochloride, and the resulting mixture system was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished.The reaction solution was directly subjected to reverse phase preparative separation in an alkaline system, and the separated solution was lyophilized to give Dam281(58mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.65(s,1H),8.64(dd,J=4.6,1.5Hz,2H),8.51(d,J=4.5Hz,1H),7.76(dd,J=4.6,1.6Hz,2H),7.65(dd,J=7.9,1.3Hz,1H),7.63(s,2H),7.16(dd,J=11.3,4.2Hz,1H),6.66(dd,J=11.0,4.0Hz,1H),6.13(d,J=8.3Hz,1H),2.82(d,J=4.5Hz,3H),2.56(q,J=7.5Hz,4H),1.13(t,J=7.5Hz,6H).
EXAMPLE 10 Synthesis of Compound Dam156
Figure BDA0002942540170000221
The first, second, third and fourth reactions were carried out according to the synthetic procedure of Dam 159.
The fifth step: in a 10mL round bottom flask, 69mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in 4.0mL of anhydrous DMF at room temperature, was stirred for 5min, and to the above system were added 77mg (0.6mmol,3.0eq) of DIEA, 28mg (0.24mmol,1.2eq) of N, N-diethylaminoethylenediamine in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation and freeze drying of the separated liquid to give Dam156(58mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.49(s,1H),8.63(d,J=5.6Hz,2H),8.44(t,J=5.7Hz,1H),7.76(dd,J=4.6,1.5Hz,2H),7.67–7.59(m,3H),7.22–7.13(m,1H),6.72–6.62(m,1H),6.16–6.09(m,1H),3.35(d,J=7.2Hz,4H),2.57(ddd,J=11.9,9.7,5.8Hz,8H),1.12(t,J=7.5Hz,6H),0.98(t,J=7.1Hz,6H).
Example 11 Synthesis of Compound Dam254
Figure BDA0002942540170000231
The first, second, third and fourth reactions were carried out according to Dam 159.
The fifth step: in a 10mL round bottom flask, 69mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid, HATU114mg (0.3mmol,1.5eq) was dissolved in 4.0mL of anhydrous DMF at room temperature, stirred for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 21mg (0.24mmol,1.2eq) of N, N-dimethylaminoethylenediamine were added to the above system in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation and freeze drying of the separated liquid to obtain Dam254(58mg) as white solid. 1 H NMR(400MHz,DMSO)δ9.47(s,1H),8.64(s,2H),8.44(s,1H),7.76(s,2H),7.62(s,3H),7.17(s,1H),6.67(s,1H),6.12(d,J=7.8Hz,1H),2.55(d,J=7.2Hz,5H),2.20(s,6H),1.12(s,6H).
EXAMPLE 12 Synthesis of Compound Dam158
Figure BDA0002942540170000232
The first, second, third and fourth reactions were carried out according to the synthetic procedure of Dam 159.
The fifth step: 69mg (0.2mmol,1.0eq) of 2- ((2, 6-diethyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, was added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 15mg (0.24mmol,1.2eq) of 2-aminoethanol were added to the above system in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed-phase preparative separation, and the separated liquid was lyophilized to give Dam158(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.57(s,1H),8.63(dd,J=4.6,1.5Hz,2H),8.50(t,J=5.5Hz,1H),7.76(dd,J=4.6,1.6Hz,2H),7.69(dd,J=7.9,1.3Hz,1H),7.62(s,2H),7.21–7.15(m,1H),6.66(dd,J=11.0,4.0Hz,1H),6.18–6.10(m,1H),4.80(t,J=5.6Hz,1H),3.56(q,J=6.1Hz,2H),3.36(d,J=3.2Hz,3H),2.55(q,J=7.5Hz,4H),1.12(t,J=7.5Hz,6H).
EXAMPLE 13 Synthesis of Compound Dac406
Figure BDA0002942540170000241
The first step is as follows: 30.7g (124mmol,1.2eq) of o-iodobenzoic acid, 22.7g (103mmol,1.0eq) of 2-chloro-6-methyl-4-bromoaniline, 15.7g (155mmol,1.5eq) of triethylamine and 9.3g (51.5mmol,0.5eq) of anhydrous copper acetate are dissolved in 372mL of DMF, and the mixture is heated to 120 ℃ under the protection of argon to react for 24 hours, and then the reaction is finished. The temperature was reduced to room temperature, an equal volume of water was added, the mother liquor was extracted with DCM (300 mL × 3), washed with water, saturated brine in that order, the organic phase was spin-dried, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1 to 1:1) to give 32g of a yellow solid.
The second step is that: 32g of 2- (4-bromo-2-chloro-6-methylphenyl) amino) benzoic acid was dissolved in 300mL of absolute ethanol, 50mL of concentrated sulfuric acid was added thereto under cooling in an ice water bath, and the mixture was heated to 100 ℃ for reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, and was concentrated by rotary evaporation to remove ethanol, 100mL of water was added to the system until no bubbles were generated during neutralization with saturated sodium carbonate. The organic phase was extracted with ethyl acetate at 50 mL. times.3, and the organic phases were combined and washed with saturated brine at 20 mL. times.3. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel column chromatography to obtain 18g of ethyl 2- (4-bromo-2-chloro-6-methylphenyl) amino) benzoate as a white solid.
The third step: 2.7g (7.3mmol,1.0eq) of ethyl 2- (4-bromo-2-chloro-6-methylphenyl) amino) benzoate, 1.2g (8.8mmol,1.2eq) of 3, 5-dimethylisoxazole-4-boronic acid, and Pd (dppf) Cl as a catalyst 2 534mg (0.73mmol,0.1eq) of potassium carbonate and 2.1g (14.6mmol,2.0eq) of potassium carbonate were dissolved in 37mL (4:1/v: v) of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL × 3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL × 3 saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) to obtain 1.86g of ethyl 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazole)) phenyl) amino) benzoate as a white solid.
The fourth step: 911mg (2.37mmol,1.0eq) of ethyl 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazole)) phenyl) amino) benzoate was dissolved in a mixed solvent of 8mL of tetrahydrofuran and 16mL of anhydrous ethanol, and a solution of 284mg (11.9mmol,5.0eq) of lithium hydroxide in 4mL of water was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 10mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. After suction filtration, the solid was washed with water to give 819mg of 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazole)) phenyl) amino) benzoic acid as a white solid.
The fifth step: 72mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazole)) phenyl) amino) benzoic acid, 72mg (0.2mmol,1.0eq) of HATU, 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, and dissolved in anhydrous DMF at room temperature, 4.0mL was stirred for 5min, and DIEA 77mg (0.6mmol,3.0eq) and hydroxylamine hydrochloride 17mg (0.24mmol,1.2eq) were added to the above system in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation, and the separated liquid was lyophilized to give Dam406(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ11.31(s,1H),9.31(s,1H),9.16(s,1H),7.52(d,J=7.6Hz,1H),7.47(s,1H),7.34(s,1H),7.24(t,J=7.6Hz,1H),6.74(t,J=7.3Hz,1H),6.23(d,J=8.2Hz,1H),2.45(s,3H),2.27(s,3H),2.21(s,3H).
EXAMPLE 14 Synthesis of Compound Dam210
Figure BDA0002942540170000261
The first, second, third and fourth reactions were prepared according to the synthetic procedure of Dac 406.
The fifth step: 72mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazol)) phenyl) amino) benzoic acid and 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, dissolved in 4.0mL of anhydrous DMF at room temperature, stirred for 5min, and to the above system were added 77mg (0.6mmol,3.0eq) of DIEA and 18mg (0.24mmol,1.2eq) of 3-aminopropanol in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation, and the separated liquid was lyophilized to give Dam210(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.65(s,1H),8.55(t,J=5.5Hz,1H),7.76–7.60(m,1H),7.46(d,J=1.8Hz,1H),7.33(d,J=1.5Hz,1H),7.24(t,J=7.8Hz,1H),6.76(t,J=7.5Hz,1H),6.23(d,J=8.2Hz,1H),4.53(t,J=5.1Hz,1H),3.50(dd,J=11.3,6.1Hz,2H),3.33(d,J=5.8Hz,2H),2.45(s,3H),2.27(s,3H),2.21(s,3H),1.72(p,J=6.6Hz,2H).
EXAMPLE 15 Synthesis of Compound Dam242
Figure BDA0002942540170000262
The first and second reactions were prepared according to the synthetic procedure of Dac 406.
The fifth step: 72mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4- (3, 5-dimethylisoxazol)) phenyl) amino) benzoic acid, 72mg (0.3mmol,1.5eq) of HATU 114mg (0.3mmol,1.5eq) was added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and to the above system were added 77mg (0.6mmol,3.0eq) of DIEA, 18mg (0.24mmol,1.2eq) of N, N-diethylaminoethylenediamine in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation, and the separated liquid was lyophilized to give Dam242(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.59(s,1H),8.48(t,J=5.5Hz,1H),7.63(d,J=7.8Hz,1H),7.45(d,J=1.5Hz,1H),7.33(s,1H),7.23(t,J=7.7Hz,1H),6.76(t,J=7.5Hz,1H),6.22(d,J=8.3Hz,1H),3.36–3.30(m,3H),2.59(t,J=7.0Hz,2H),2.54(d,J=7.1Hz,3H),2.44(s,3H),2.27(s,3H),2.21(s,3H),0.98(t,J=7.1Hz,6H).
EXAMPLE 16 Synthesis of Compound Dam418
Figure BDA0002942540170000271
The first and second reactions were carried out according to the Dam106 synthesis procedure.
The third step: 1.1g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2-chloro-6-methylphenyl) amino) benzoate, 443mg (3.6mmol,1.2eq) of pyridine-4-boronic acid, and Pd (dppf) Cl as a catalyst 2 220mg (0.3mmol,0.1eq), 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 15mL (4:1/v: v) of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, concentrated to dryness, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) to give 916mg of ethyl 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoate as a white solid.
The fourth step: 734mg (2.0mmol,1.0eq) of ethyl 2- ((2-chloro-6-methyl-4-pyridyl) phenyl) amino) benzoate was dissolved in 8mL of a mixed solvent of tetrahydrofuran and 16mL of absolute ethanol, and a solution of 240mg (10mmol,5.0eq) of lithium hydroxide in 4mL of water was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 10mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. And (4) carrying out suction filtration, and washing the solid with water to obtain 725mg of white solid 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoic acid.
The fifth step: 68mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 17mg (0.24mmol,1.2eq) of hydroxylamine hydrochloride were added to the above system in this order, and the resulting mixture system was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed-phase preparative separation, and the separated liquid was lyophilized to give Dam418(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ11.36(s,1H),9.44(s,1H),8.87(d,J=5.9Hz,2H),8.22(d,J=6.2Hz,2H),8.09(s,1H),7.96(s,1H),7.53(d,J=7.7Hz,1H),7.26(t,J=7.7Hz,1H),6.80(t,J=7.4Hz,1H),6.28(d,J=8.2Hz,1H),2.26(s,3H).
EXAMPLE 17 Synthesis of the Compound Dam226
Figure BDA0002942540170000281
The first, second, third and fourth reactions were prepared according to the synthetic procedure of Dac 406.
The fifth step: 68mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoic acid and 114mg (0.3mmol,1.5eq) of HATU were added to a 10mL round-bottomed flask, and dissolved in 4.0mL of anhydrous DMF at room temperature, followed by stirring for 5min, and 77mg (0.6mmol,3.0eq) of DIEA and 14mg (0.24mmol,1.2eq) of ammonium chloride were added to the above system in this order, and the resulting mixture system was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation and freeze drying of the separated liquid gave Dam226(47mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.99(s,1H),8.65(dd,J=4.6,1.5Hz,2H),8.11(s,1H),7.90(d,J=2.0Hz,1H),7.83–7.77(m,3H),7.74(d,J=7.9Hz,1H),7.49(s,1H),7.24(t,J=7.7Hz,1H),6.74(t,J=7.5Hz,1H),6.21(d,J=8.0Hz,1H),2.25(s,3H).
EXAMPLE 18 Synthesis of Compound Dam230
Figure BDA0002942540170000291
The first, second, third and fourth reactions were prepared according to the synthetic procedure of Dac 406.
The fifth step: 68mg (0.2mmol,1.0eq) of 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, dissolved in anhydrous DMF at room temperature, 4.0mL, were added to a 10mL round-bottomed flask, stirred for 5min, DIEA 77mg (0.6mmol,3.0eq) and 3-aminopropanol 18mg (0.24mmol,1.2eq) were added sequentially to the above system, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. Reversed phase preparative separation and freeze drying of the separated liquid to give Dam230(41mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.70(s,1H),8.65(d,J=6.1Hz,2H),8.57(t,J=5.4Hz,1H),7.90(d,J=1.8Hz,1H),7.79(d,J=6.2Hz,3H),7.66(d,J=7.8Hz,1H),7.24(t,J=7.8Hz,1H),6.78(t,J=7.5Hz,1H),6.23(d,J=8.3Hz,1H),4.53(t,J=5.1Hz,1H),3.50(q,J=6.1Hz,2H),3.33(s,1H),2.25(s,3H),1.72(p,J=6.6Hz,2H).
EXAMPLE 19 Synthesis of the Compound Dam264
Figure BDA0002942540170000301
The first step is as follows: 30g (120mmol,1.2eq) of o-iodobenzoic acid, 25.6g (100mmol,1.0eq) of 2, 6-diisopropyl-4-bromoaniline, 150mmol,1.5eq) and 9g (5.0mmol,0.5eq) of anhydrous copper acetate are dissolved in 500mL of DMF, and the mixture is heated to 120 ℃ under the protection of argon to react for 24h after the reaction is finished. The temperature was lowered to room temperature, equal volume of water was added, the mother liquor was extracted 300mL × 3 with DCM, the DMF was washed with water, the organic phase was spin dried and the column ratio was changed from PE: EA 20:1 to PE: EA 1:1 to give 14.6g of a yellow solid.
The second step is that: 3.8g of 2- (4-bromo-2, 6-diisopropylphenyl) amino) benzoic acid was dissolved in 200mL of absolute ethanol, and 20mL of concentrated sulfuric acid was added thereto under cooling in an ice-water bath, followed by heating to 100 ℃ and reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, and was concentrated by rotary evaporation to remove ethanol, 100mL of water was added to the system until no bubbles were generated during neutralization with saturated sodium carbonate. The organic phase was extracted with ethyl acetate at 50 mL. times.3, and the organic phases were combined and washed with saturated brine at 10 mL. times.3. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel column chromatography to give 3.0g of ethyl 2- (4-bromo-2, 6-diisopropylphenyl) amino) benzoate as a white solid.
The third step: 1.2g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-diisopropylphenyl) amino) benzoate, 443mg (3.6mmol,1.2eq) of pyridine-4-boronic acid, Pd (dppf) as a catalyst, Cl 2 220mg (0.3mmol,0.1eq), 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 15mL (4:1/v: v) of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL × 3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL × 3 saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel chromatography (petroleum ether: ethyl acetate ═ 20:1) to give ethyl 2- (2, 6-diisopropyl-4-pyridylphenyl) amino) benzoate as a white solid (913 mg).
The fourth step: 748mg (2.0mmol,1.0eq) of ethyl 2- ((2, 6-diisopropyl-4-pyridylphenyl) amino) benzoate was dissolved in 4mL of a mixed solvent of tetrahydrofuran and 8mL of absolute ethanol, and a solution of 240mg (10mmol,5.0eq) of lithium hydroxide in 2mL of water was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 20mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration is carried out, and the solid is washed by water, thus obtaining 693mg of white solid 2- ((2, 6-diisopropyl-4-pyridylphenyl) amino) benzoic acid.
The fifth step: in a 10mL round-bottom flask, 75mg (0.2mmol,1.0eq) of 2- ((2, 6-diisopropyl-4-pyridylphenyl) amino) benzoic acid, 114mg (0.3 mmo) of HATU were addedl,1.5eq), dissolved in 4.0mL of anhydrous DMF at room temperature, stirred for 5min, and to the above system were added 77mg (0.6mmol,3.0eq) of DIEA, 14mg (0.24mmol,1.2eq) of ammonium chloride in that order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation in an alkaline system, and the separated solution was lyophilized to obtain Dam264(58mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.84(s,1H),8.65(d,J=6.1Hz,2H),8.06(s,1H),7.82–7.76(m,2H),7.72(d,J=7.9Hz,1H),7.63(s,2H),7.38(s,1H),7.17(t,J=7.8Hz,1H),6.62(t,J=7.4Hz,1H),6.12(d,J=8.4Hz,1H),3.19–3.00(m,2H),1.16(t,J=6.6Hz,12H).
EXAMPLE 20 Synthesis of the Compound Dam185
Figure BDA0002942540170000311
The first step is as follows: 29.8g (120mmol,1.2eq) of o-iodobenzoic acid, 20.4g (100mmol,1.0eq) of 2-fluoro-6-methyl-4-bromoaniline, 15.2g (150mmol,1.5eq) of triethylamine and 9.1g (50mmol,0.5eq) of anhydrous copper acetate are dissolved in 360mL of DMF, and the mixture is heated to 120 ℃ under the protection of argon to react for 24 hours after the reaction is finished. The temperature was lowered to room temperature, equal volume of water was added, the mother liquor was extracted 300mL × 3 with DCM, the DMF was washed with water, the organic phase was spin dried and the column ratio was changed from PE: EA ═ 20:1 to PE: EA ═ 1:1 to give 33g of yellow solid.
The second step is that: 33g of 2- (4-bromo-2-fluoro-6-methylphenyl) amino) benzoic acid is dissolved in 300mL of absolute ethanol, 50mL of concentrated sulfuric acid is added to the absolute ethanol under cooling of an ice water bath, and the mixture is heated to 100 ℃ for reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, and was concentrated by rotary evaporation to remove ethanol, 100mL of water was added to the system until no bubbles were generated during neutralization with saturated sodium carbonate. The organic phase was extracted with ethyl acetate (50 mL. times.3), and the organic phases were combined and washed with saturated brine (20 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel column chromatography to obtain 13.6g of ethyl 2- (4-bromo-2-fluoro-6-methylphenyl) amino) benzoate as a white solid.
The third step: 1.1g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2-fluoro-6-methylphenyl) amino) benzoate, 443mg (3.6mmol,1.2eq) of pyridine-4-boronic acid, and a catalystPd(dppf)Cl 2 220mg (0.3mmol,0.1eq), 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 15mL (4:1/v: v) of a mixed solvent of dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, the solvent 1/2 was removed by rotary evaporation, 50mL X3 was extracted with ethyl acetate, and the organic phases were combined and washed with 10mL X3 of saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate 20:1) to give 801mg of ethyl 2- ((2-fluoro-6-methyl-4-pyridine) amino) benzoate as a white solid.
The fourth step: ethyl 2- ((2-fluoro-6-methyl-4-pyridyl) phenyl) amino) benzoate 741mg (2.12mmol,1.0eq) was dissolved in a mixed solvent of tetrahydrofuran 8mL and absolute ethanol 16mL, and a solution of lithium hydroxide 255mg (10.6mmol,5.0eq) in water 4mL was slowly added dropwise thereto under cooling in an ice-water bath. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding 10mL of water, placing in an ice water bath, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration is carried out, and the solid is washed by water, thus obtaining 723mg of white solid 2- ((2-chloro-6-methyl-4-pyridine) phenyl) amino) benzoic acid.
The fifth step: 64mg (0.2mmol,1.0eq) of 2- ((2-fluoro-6-methyl-4-pyridine) phenyl) amino) benzoic acid, 114mg (0.3mmol,1.5eq) of HATU, was added to a 10mL round-bottomed flask, dissolved in anhydrous DMF (4.0mL) at room temperature, stirred for 5min, and 77mg (0.6mmol,3.0eq) of DIEA, 18mg (0.24mmol,1.2eq) of 3-aminopropanol, were added to the above system in this order, and the resulting mixture was reacted at 45 ℃ for 2 hours. And (5) TLC detection, and the consumption of the raw materials is finished. The reaction solution was directly subjected to reverse phase preparative separation in an alkaline system, and the separated solution was lyophilized to obtain Dam185(26mg) as a white solid. 1 H NMR(400MHz,DMSO)δ9.61(s,1H),8.64(d,J=5.6Hz,2H),8.60(t,J=5.4Hz,1H),7.78(d,J=6.1Hz,2H),7.71–7.63(m,3H),7.27(t,J=7.7Hz,1H),6.79(t,J=7.6Hz,1H),6.42(dd,J=8.3,3.8Hz,1H),4.54(t,J=5.1Hz,1H),3.50(dd,J=11.6,6.1Hz,2H),3.33(d,J=6.8Hz,2H),2.30(s,3H),1.81–1.61(m,2H).
EXAMPLE 21 Synthesis of Compounds Dac258 and Dac58
Figure BDA0002942540170000331
The first step is as follows: 20.2g (100mmol,1.0eq) of 2-chloro-5-nitrobenzoic acid, 26.5g (110mmol,1.1eq) of 2, 6-dichloro-4-bromoaniline and 28.8g (300mmol,3.0eq) of sodium tert-butoxide are dissolved in 500mL of DMF, heated to 80 ℃ under the protection of argon and reacted for 24 hours, and then the reaction is finished. And cooling to room temperature, adjusting the pH value with 2M dilute hydrochloric acid to separate out yellow solid, performing suction filtration, and washing with water to obtain 28.0g of target product yellow solid.
The second step: 28.0g of 2- (4-bromo-2, 6-dichlorophenyl) amino) -5-nitrobenzoic acid is dissolved in 500mL of absolute ethanol, 50mL of concentrated sulfuric acid is added thereto under cooling in an ice-water bath, and the mixture is heated to 100 ℃ for reflux reaction for 12 hours. After the reaction, the reaction system was cooled to room temperature, ethanol was removed by rotary evaporation and concentration, 50mL of water was added to the system, and saturated sodium carbonate was used for neutralization until no bubbles were formed. The organic phases were extracted with ethyl acetate (20 mL. times.3), and the organic phases were combined and washed with saturated brine (10 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and separated by silica gel chromatography (petroleum ether: ethyl acetate 99:1) to obtain 21.0g of an aimed product as a white solid.
The third step: 1.3g (3.0mmol,1.0eq) of ethyl 2- (4-bromo-2, 6-dichlorophenyl) amino) -5-nitrobenzoate, 508mg (3.6mmol,1.2eq) of 3, 5-dimethylisoxazole-4-boronic acid and Pd (dppf) Cl as a catalyst 2 220mg (0.3mmol,0.1eq) and 828mg (6.0mmol,1.5eq) of potassium carbonate were dissolved in 30mL of a mixed solvent of 1, 4-dioxane and water, and the mixture was heated to 100 ℃ for reaction for 24 hours. Cooled to room temperature, concentrated to dryness, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate: 10:1) to obtain the target product, ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) -5-nitrobenzoate (1.3 g), as a white solid.
The fourth step: dissolving 1.3g (2.9mmol,1.0eq) of ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) -5-nitrobenzoate, 943mg (14.5mmol,5.0eq) of zinc powder, 1.8g (29.0mmol,10.0eq) of ammonium formate in 58mL of DMF, heating to 45 ℃, reacting overnight, adding water into the system, performing EA extraction, and performing silica gel column chromatography to obtain 1.22g of yellow target product ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) -5-aminobenzoate.
The fifth step: 79mg (0.55mmol,1.1eq) of monoethyl fumarate, 228mg (0.6mmol,1.2eq) of HATU were dissolved in DMF (4mL) and stirred for 5min, and to the above system were added sequentially, 194mg (1.5mmol,3.0eq) of DIEA, 210mg (0.5mmol,1.0eq) of ethyl 2- ((2, 6-dichloro-4- (3, 5-dimethylisoxazole) phenyl) amino) -5-aminobenzoate and the resulting mixture was reacted at 45 ℃. And (5) TLC detection, and the consumption of the raw materials is finished. And adding an ice-water mixture into the system to separate out a yellow solid, and performing suction filtration and water washing to obtain a target product, namely yellow solid Dac258(180 mg).
The fourth step: compound Dac 25855 mg (0.1mmol,1.0eq) was dissolved in a mixed solvent of tetrahydrofuran (2mL) and absolute ethanol (4mL), and a solution of 24mg (1.0mmol,10.0eq) of lithium hydroxide in water (1mL) was slowly added dropwise thereto. Heating to 45 ℃ for reaction overnight, after the reaction is finished, cooling the system to room temperature, concentrating to remove the organic solvent, adding water, adjusting the pH to 3 with 2M dilute hydrochloric acid, and continuously stirring the obtained suspension at room temperature for 30 min. Suction filtration and washing of the solid with water gave the desired product Dac58(46 mg).
Biological example 1: FTO inhibitory Activity assay
And purifying by nickel column affinity chromatography to obtain the high-purity FTO protein.
The reaction system for inhibiting the FTO enzyme activity is as follows: 50mM Tris-HCl, pH 7.5, 0.3. mu.M FTO, 1. mu.M 39nt-M 6 A modified double-stranded DNA, 300. mu.M 2OG, 280. mu.M (NH) 4 ) 2 Fe(SO 4 ) 2 2mM L-Ascorbic Acid and compounds of different concentrations, incubated at room temperature for 2h, slowly heated at 65 ℃ for inactivation, and annealed to double strand by adding 1. mu.M of 39nt antisense strand DNA. Taking 8ul reaction solution, carrying out enzyme digestion on a double-stranded substrate by using methylation sensitive enzyme DpnII, carrying out 15% non-denaturing polyacrylamide electrophoresis detection on a sample subjected to enzyme digestion, taking a picture under a Gel imaging system after Gel-Red dyeing, carrying out gray scale reading inhibition rate on the obtained strip, and evaluating IC (integrated circuit) for inhibiting FTO (fluorine-doped tin oxide) demethylation activity aiming at a compound with better inhibition rate 50 The results are given in the table below.
TABLE 1 inhibition of FTO enzymatic Activity by Compounds IC 50
Figure BDA0002942540170000341
Figure BDA0002942540170000351
The results show that the above compounds have certain activity, IC thereof 50 Less than 10. mu.M, and mostly less than 1. mu.M.
Biological example 2: evaluation of tumor cell proliferation inhibitory Activity
Respectively culturing solid tumor cell lines such as melanoma cell line B16-OVA, lung cancer cell line LLC, colon cancer cell line MC38, etc., seeding cells with a density of 1000 per well in 96-well plate, and placing at 37 deg.C CO 2 An incubator, culturing cells to be adherent, adding different compounds to continue culturing for 72h, adding 10uL of MTT solution into each hole, continuing incubating for 4h, detecting the absorbance value at 490nm, calculating the inhibition rate by taking a DMSO group as a control, and evaluating the cell proliferation inhibition activity IC aiming at the compound with better inhibition rate 50 The results are given in the table below.
TABLE 2 proliferation inhibitory Activity of FTO inhibitors on tumor cells IC 50
Figure BDA0002942540170000352
Figure BDA0002942540170000361
Biological example 3: dot-blot hybridization detection of m in tumor cells 6 Altered A abundance
Tumor cells were plated at a certain density in 6cm dishes and the following day the compound was added (concentration setting depending on IC of MTT) 50 Value), removing cell culture medium after 48-72hr treatment, adding 1mL TRIzol reagent, collecting cells, lysing, sucking into RNase free tube, and incubating on ice for 5min to completely separate nucleoprotein complex; adding 200 mu L chloroform for cracking, vortexing for 15s, and incubating; and (4) centrifuging and layering. Adding the aqueous phase containing RNAThe layer was transferred to a new 1.5mL RNase free tube and the solution was removed; adding 500 μ L of pre-cooled isopropanol, reversing, mixing, and incubating for 10 min; centrifuging at 4 ℃ for 10min, removing the supernatant, adding 1mL of DEPC & H 2 Resuspending the precipitate in O-ethanol solution, and centrifuging at 4 deg.C for 5 min; the supernatant was aspirated off, the RNA pellet was dried on ice for 10min, and 20-50. mu.L of DEPC. H was added 2 Dissolving the precipitate with O, dissolving with vortex, centrifuging, repeating for 3 times, and determining the concentration. After the concentration is measured, denaturation is carried out for 3min at 95 ℃, the mixture is immediately placed on ice for 5min, and then the mixture is centrifuged for a short time, the tube bottom is flicked, and the mixture is centrifuged again and placed on ice; diluting the RNA concentration into concentration gradient, and spotting; drying with warm light for 20-30min, crosslinking with 12000W light for 3min, sealing with 5% skimmed milk prepared from PBST at room temperature for 1 hr, m 6 Blocking the primary antibody overnight, washing the membrane with PBST, incubating the secondary antibody, washing the membrane with PBST for three times, and developing the color. The results are shown in FIG. 1, where FTO inhibitors are capable of concentration-dependent alteration of m in tumor cells 6 Abundance of A.
Biological example 4: evaluation of animal pharmacodynamics
A5-week-old C57BL/6 strain black mouse with a weight of 18-20g is selected. Simultaneously culturing the tumor cell MC 38. Injection of 2.5X 10 in the right axilla of mice 5 Cells (100uL, cell density 2.5X 10 6 cells/mL). After 6 days of tumor implantation, the tumor Length and Width were measured and the tumor Volume was calculated (Volume ═ 0.5 × Length × Width) 2 ) Tumor volume of 60mm was selected on day 9 3 -100mm 3 Range tumor-bearing mice, 4 per group.
Candidate compounds Dam59 and Dam159 were dissolved in DMSO, respectively, to prepare a 20mM stock solution, and diluted with PBS to finally obtain a dosing solution with a DMSO content of 20% for use.
Candidate compounds Dam59(2mg/kg) and Dam159(0.5mg/kg) were injected intraperitoneally from days 10, 11, and 12, respectively. On days 13 and 16, 100mg/kg of antibody PD-L1 and antibody negative control IgG were injected by intraperitoneal administration. Tumor volume and mouse body weight were measured every two days. Results as shown in fig. 2 and 3, the compounds of the present invention exhibited a significant synergistic effect when used in combination with the antibody PD-L1.
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 or 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 appended claims of the present application.

Claims (10)

1. A compound shown as the following formula (I), and the application of the pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof,
Figure FDA0002942540160000011
wherein, the first and the second end of the pipe are connected with each other,
A 1 、A 2 、A 3 、A 4 each independently is CR' or N;
m is selected from the group consisting of: CR' 2 NH, O or S;
r' is selected from the group consisting of: H. halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, and substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl, substituted or unsubstituted C 1 -C 6 Amido, substituted or unsubstituted C 2 -C 12 Ester group, or substituted or unsubstituted C 1 -C 10 Alkyl, or substituted or unsubstituted C3-C10 enamide;
x has a structure represented by the following formula: carboxy, O-substituted or unsubstituted hydroxamic acid group, substituted or unsubstituted C 2 -C 12 Ester group, substituted or unsubstituted amide group (C (O) NH 2 ) Substituted or unsubstituted 3-12 membered heterocyclyl;
y is selected from the group consisting of: substituted or unsubstituted C 6 -C 12 Substituted or unsubstituted 3-12 membered heterocyclyl;
R a 、R b 、R c 、R d each independently selected from the group consisting of: H. halogen, -OH, CN, NO 2 、NH 2 Substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 An alkoxy group;
the substitution means that one or more hydrogen atoms on the group are substituted with a substituent selected from the group consisting of: halogen atom, carbonyl (═ O), carboxyl, hydroxyl, amino, nitro, cyano, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Amido, C 2 -C 12 Ester group, substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 2 -C 10 Alkenyl, substituted or unsubstituted C 2 -C 10 Alkynyl, substituted or unsubstituted C 6 -C 10 Aryl, or substituted or unsubstituted five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or C 1 -C 6 An alkylamino group; wherein, said substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 6 -C 10 The substituents of the aryl or five-or six-membered heteroaryl group are selected from the group consisting of: halogen atom, carbonyl (═ O), hydroxyl, carboxyl, C 1 -C 6 Alkoxycarbonyl, amino, C 1 -C 6 Amido, nitro, cyano, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 6 -C 10 Aryl or five-or six-membered heteroaryl, 3-to 12-membered heterocyclyl, 3-to 12-membered cycloalkyl, preferably halogen atom, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy or phenyl, 5-6 membered heterocyclyl;
wherein, the compound of formula (I) and an immune checkpoint inhibitor are used together for preparing a pharmaceutical composition for treating or preventing solid tumors.
2. The compound of claim 1, and the use of a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, wherein the compound of formula (I) has the structure shown in formula (II):
Figure FDA0002942540160000021
wherein the content of the first and second substances,
A 1 、A 2 、A 3 、A 4 each independently CR' or N;
m is selected from the group consisting of: NH or S;
r' is selected from the group consisting of: H. halogen, hydroxy, amino, nitro, cyano, substituted or unsubstituted C 1 -C 10 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 Alkylamino, substituted or unsubstituted C 1 -C 6 An amide;
R 0 selected from the group consisting of: hydrogen, hydroxy, substituted or unsubstituted C 1 -C 10 An alkyl group,
Figure FDA0002942540160000022
Wherein R is a ,R b Each independently selected from the group consisting of: hydrogen, substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 3 -C 10 Cycloalkyl (including monocyclic, polycyclic, bridged ring structures), substituted or unsubstituted C 2 -C 6 Alkenyl, substituted or unsubstituted C 2 -C 6 An alkynyl group;
m is selected from the group consisting of: 0.1, 2, 3 or 4;
R x 、R y each independently selected from the group consisting of: halogen, substituted or unsubstituted C 1 -C 4 An alkyl group;
het is selected from the group consisting of: substituted or unsubstituted C 6 -C 10 Substituted or unsubstituted 4-7 membered saturated heterocyclic group, substituted or unsubstituted 3-12 membered heterocyclic group;
the substituent means that one or more hydrogen atoms on the group are substituted with a substituent selected from the group consisting of: halogen, oxygen atom (═ O), carboxyl, hydroxyl, amino, nitro, cyano, C 1 -C 6 Alkoxy radical, C 1 -C 6 Alkylamino radical, C 1 -C 6 Alkoxycarbonyl group, C 1 -C 6 Amido, C 2 -C 12 An ester group; or unsubstituted or substituted by one or more halogens or C 1 -C 6 Alkyl-substituted groups selected from the group consisting of: c 1 -C 10 Alkyl radical, C 2 -C 10 Alkenyl radical, C 2 -C 10 Alkynyl, C 6 -C 10 Aryl, 5-6 membered heteroaryl, 3-12 membered heterocyclyl, 3-12 membered cycloalkyl.
3. The use according to claim 1, wherein R is 0 Has a structure represented by the following formula:
Figure FDA0002942540160000031
wherein the content of the first and second substances,
R a ,R b each independently selected from the group consisting of: hydrogen, substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 3 -C 10 Cycloalkyl (including monocyclic, polycyclic, bridged ring structures), substituted or unsubstituted C 2 -C 6 Alkenyl, substituted or unsubstituted C 2 -C 6 An alkynyl group;
m is selected from the group consisting of: 1.2 or 3.
4. Use according to claim 1, characterised in that said Het is selected from the group consisting of: substituted or unsubstituted pyridine, substituted or unsubstituted tetrazole, substituted or unsubstituted triazole, substituted or unsubstituted pyrimidine, substituted or unsubstituted pyrazole, substituted or unsubstituted isoxazole, substituted or unsubstituted morpholine, substituted or unsubstituted thiomorpholine, substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, substituted or unsubstituted oxetane, substituted or unsubstituted thietane, substituted or unsubstituted azetidine.
5. Use according to claim 1, characterised in that said Het is selected from the group consisting of:
Figure FDA0002942540160000032
wherein each R is 1 、R 2 、R 3 Or R 4 Each independently selected from the group consisting of: hydrogen, halogen, hydroxy, amino, oxygen atom (═ O), Carboxy (COOH), substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 6 -C 10 Aryl, substituted or unsubstituted 5-6 membered heteroaryl, substituted or unsubstituted C 1 -C 6 Alkylamino, substituted or unsubstituted C 1 -C 6 Alkoxycarbonyl, substituted or unsubstituted C 1 -C 6 An amido group; wherein the substituents are selected from the group consisting of: F. cl, C 1 -C 6 An alkyl group.
6. The use according to claim 5, wherein each R is 1 ,R 2 ,R 3 ,R 4 Each independently selected from the group consisting of: H. substituted or unsubstituted C 1 -C 6 Alkyl, substituted or unsubstituted C 1 -C 6 Alkoxy, substituted or unsubstituted C 1 -C 6 An alkylamino group.
7. The use according to claim 1, wherein the compound of formula (I) is selected from the group consisting of:
Figure FDA0002942540160000041
8. the use of claim 1, wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-L1 antibody, an anti-PD-1 antibody, or a combination thereof.
9. The use according to claim 1, wherein the solid tumor is selected from the group consisting of: melanoma, lung cancer, colon cancer, renal cancer, pancreatic cancer, lung cancer, osteosarcoma.
10. A compound, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, wherein the compound is selected from the group consisting of:
Figure FDA0002942540160000051
CN202110184609.9A 2021-02-08 2021-02-08 Pharmaceutical composition for resisting tumor Pending CN114907267A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110184609.9A CN114907267A (en) 2021-02-08 2021-02-08 Pharmaceutical composition for resisting tumor
PCT/CN2022/075561 WO2022166990A1 (en) 2021-02-08 2022-02-08 Anti-tumor pharmaceutical combination

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110184609.9A CN114907267A (en) 2021-02-08 2021-02-08 Pharmaceutical composition for resisting tumor

Publications (1)

Publication Number Publication Date
CN114907267A true CN114907267A (en) 2022-08-16

Family

ID=82742006

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110184609.9A Pending CN114907267A (en) 2021-02-08 2021-02-08 Pharmaceutical composition for resisting tumor

Country Status (2)

Country Link
CN (1) CN114907267A (en)
WO (1) WO2022166990A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104069092A (en) * 2014-07-14 2014-10-01 中国科学院上海药物研究所 Use of 2-(substituted phenylamino) benzoic acid and ester compound thereof in preparation of FTO (Fat Mass and Obesity-Associated Protein) inhibitor
CN107567336A (en) * 2014-10-29 2018-01-09 戊瑞治疗有限公司 The therapeutic alliance of cancer
WO2018157843A1 (en) * 2017-03-02 2018-09-07 中国科学院上海药物研究所 2-(substituted benzene matrix) aromatic formate fto inhibitor, preparation method therefor, and applications thereof
CN110573500A (en) * 2017-05-24 2019-12-13 上海和誉生物医药科技有限公司 N- (nitrogen heterocyclic aryl) lactam-1-formamide derivative and preparation method and application thereof
CN111000859A (en) * 2018-10-05 2020-04-14 澳门科技大学 Methods and kits for treating cancer by combination therapy
CN111712242A (en) * 2017-09-25 2020-09-25 凯莫森特里克斯股份有限公司 Combination therapy using chemokine receptor 2(CCR2) antagonists and PD-1/PD-L1 inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000041505A2 (en) * 1999-01-13 2000-07-20 Warner-Lambert Company Anthranilic acid derivatives
EP2228367A1 (en) * 2009-03-13 2010-09-15 Almirall, S.A. Addition salts of amines containing hydroxyl and/or carboxylic groups with amino nicotinic acid derivatives as DHODH inhibitors
EP3781162A1 (en) * 2018-04-16 2021-02-24 Arrys Therapeutics, Inc. Ep4 inhibitors and use thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104069092A (en) * 2014-07-14 2014-10-01 中国科学院上海药物研究所 Use of 2-(substituted phenylamino) benzoic acid and ester compound thereof in preparation of FTO (Fat Mass and Obesity-Associated Protein) inhibitor
CN107567336A (en) * 2014-10-29 2018-01-09 戊瑞治疗有限公司 The therapeutic alliance of cancer
WO2018157843A1 (en) * 2017-03-02 2018-09-07 中国科学院上海药物研究所 2-(substituted benzene matrix) aromatic formate fto inhibitor, preparation method therefor, and applications thereof
CN110573500A (en) * 2017-05-24 2019-12-13 上海和誉生物医药科技有限公司 N- (nitrogen heterocyclic aryl) lactam-1-formamide derivative and preparation method and application thereof
CN111712242A (en) * 2017-09-25 2020-09-25 凯莫森特里克斯股份有限公司 Combination therapy using chemokine receptor 2(CCR2) antagonists and PD-1/PD-L1 inhibitors
CN111000859A (en) * 2018-10-05 2020-04-14 澳门科技大学 Methods and kits for treating cancer by combination therapy

Also Published As

Publication number Publication date
WO2022166990A1 (en) 2022-08-11

Similar Documents

Publication Publication Date Title
CN108524482B (en) Use of 2- (substituted phenylamino) benzoic acid FTO inhibitors for treating leukemia
CN111484477B (en) Benzopyridone heterocyclic compound and application thereof
CN106928206B (en) Aldehyde compound and its preparation method and use
KR20200105878A (en) Aromatic vinyl or aromatic ethyl derivative, its preparation method, intermediate, drug composition and use
CN103946231B (en) Oleanolic acid amidated derivative, and its preparation method and application
CN111393404B (en) Benzothiophene compounds, and pharmaceutical composition and application thereof
WO2012031563A1 (en) Heterocyclic amino berbamine derivatives, preparation method and use thereof
CN109748911B (en) Triazole-containing IDO inhibitor, and preparation method and medical application thereof
CN109111426B (en) Fused bicyclic heteroaryl or aryl compound and application thereof
CN110003204B (en) BET protein inhibitor, preparation method and application thereof
CN110240587B (en) Aryl difluoro benzyl ether compounds, preparation method and application
CN101805338B (en) Oxadiazole-based piperazine derivative and application thereof
CN112174940A (en) 3- (6, 7-bis (2-methoxyethoxy) -quinazoline-4-amido) phenyl-1H-triazole derivative
CN110577526A (en) Salt of bromodomain structural protein inhibitor and preparation method and application thereof
CN114907267A (en) Pharmaceutical composition for resisting tumor
CN111606890A (en) Acryloyl group-containing nuclear transport modulators and uses thereof
CN113527275A (en) SKLB1039 compound and preparation method and application thereof
WO2019029554A1 (en) Sulfonamide derivative, preparation method thereof, and use of same in medicine
CN114901639B (en) HBV inhibitors and uses thereof
CN115160277A (en) Apigenin derivative and application thereof
CN111393405B (en) Fluorine-containing substituted benzothiophene compounds, and pharmaceutical composition and application thereof
CN107513089B (en) Novel cytidine derivative dimer and application thereof
CN109879887B (en) Thieno [3,2-d ] pyrimidine derivative containing indole structure and application thereof
CN106810553B (en) 3- (4, 5-substituted aminopyrimidine) phenyl derivative and application thereof
CN110590807B (en) Thieno [3,2-d ] pyrimidine derivative 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