CN113717159A - Indolone compounds and pharmaceutical composition, preparation method and application thereof - Google Patents

Indolone compounds and pharmaceutical composition, preparation method and application thereof Download PDF

Info

Publication number
CN113717159A
CN113717159A CN202111089653.8A CN202111089653A CN113717159A CN 113717159 A CN113717159 A CN 113717159A CN 202111089653 A CN202111089653 A CN 202111089653A CN 113717159 A CN113717159 A CN 113717159A
Authority
CN
China
Prior art keywords
alkyl
unsubstituted
heteroatom
substituted
cancer
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
CN202111089653.8A
Other languages
Chinese (zh)
Inventor
蒋晟
张阔军
朱琳瑜
章翔宇
王天雨
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical University
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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202111089653.8A priority Critical patent/CN113717159A/en
Publication of CN113717159A publication Critical patent/CN113717159A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Abstract

The invention discloses indolone compounds with a structure shown in a general formula I, and a pharmaceutical composition, a preparation method and application thereof. The benzindolone compounds of the invention have good inhibitory activity to VEGFRs and NAMPT, and have good effect on tumorsHas therapeutic effect.

Description

Indolone compounds and pharmaceutical composition, preparation method and application thereof
Technical Field
The invention relates to innovative medicaments, a preparation method and application, in particular to indolone compounds and a pharmaceutical composition, a preparation method and application thereof.
Background
Angiogenesis, i.e., the formation of new blood vessels by sprouting or the division of blood vessels from already existing ones for cell proliferation, migration and survival, plays a crucial role in normal embryonic development and adult growth. However, abnormal angiogenesis is closely related to the occurrence and development of many diseases, such as cancer, psoriasis, rheumatoid arthritis, inflammation, and retinal complications. In particular, in cancer, cancer angiogenesis produces new blood vessels, ensures the supply of oxygen and nutrients required for tumor cell proliferation, and is an important process for maintaining tumor cell growth and metastasis. The Vascular Endothelial Growth Factor (VEGF) family, including VEGF-A, VEGF-B, VEGF-C, VEGF-D and placental growth factor (PlGF), and its receptor, Vascular Endothelial Growth Factor Receptor (VEGFR), including VEGFR-1(Flt-1), VEGFR-2(Flk-1/KDR) and VEGFR-3, are highly expressed in a variety of tumors and contribute to the development and maintenance of the tumor vascular network, which promotes the growth and metastasis of tumor cells. Therefore, inhibition of VEGF and its receptor VEGFR-mediated signaling pathways is a promising anti-tumor strategy. The research of the VEGFR small molecule inhibitor is concerned, 9 VEGFR small molecule inhibitors are on the market at present, and a plurality of VEGFR small molecule inhibitors with different structures are in different research stages before clinic or in clinic. Compared with monoclonal antibodies, small molecule inhibitors have the advantages of non-single action target and wider inhibition activity, but have the problems of larger toxic and side effects, drug resistance and the like caused by poor selectivity.
Nicotinamide adenine dinucleotide (NAD for short)+) Is an indispensable substance in the process of electron transfer and plays an important role in the processes of energy metabolism and signal transduction. In addition, NAD+And the reduced NADH thereof have important significance for maintaining the reducing environment in the cells so as to protect the cells from being damaged by oxidative stress. Thus, intracellular NAD+Is a critical factor in determining cell fate. Tumor cells have an uncontrolled proliferation rate, a rapid metabolic rate and a significant increase over normal cellsSo that the tumor cells are towards NAD+The level change is more sensitive. NAD (nicotinamide adenine dinucleotide)+Key enzymes of the in vivo biosynthetic pathway have become important targets for anticancer drugs. NAD (nicotinamide adenine dinucleotide)+In vivo synthetic pathways include de novo and salvage synthetic pathways.
NAD+The salvage synthesis route is to use nicotinic acid, nicotinamide or nicotinamide ribose as starting materials. Due to much NAD+Nicotinamide can be quickly released again by consuming enzymes such as poly (adenosine diphosphate ribose) polymerase (PARP) and Sirtuins (SIRT), and thus NAD from nicotinamide+The salvage synthesis pathway is the most economical, predominant NAD+The biosynthetic pathway (Galli U.et al.J.Med.chem.2013, 22, 56 (16): 6279-96). Nicotinamide phosphoribosyltransferase (NAMPT) controls nicotinamide to NAD+The rate-limiting enzyme of transformation. In view of the abnormal exuberance of energy metabolism by tumor cells, NAD is more desirable+Rapid and timely recruitment, tumor cells are more dependent on NAMPT and are more sensitive to NAMPT inhibition. Therefore, NAMPT is considered as a potent anti-cancer target and NAMPT inhibitors have been the focus of extensive research. Currently, two NAMPT inhibitors CHS-828 and FK866 have entered the clinical trial phase (Montecucco F. et al. curr. drug targets.2013, 1, 14 (6): 637-43). However, the results of clinical trials of these two compounds are not ideal and patients do not show significant therapeutic effects after administration.
Figure BDA0003265645630000021
The multi-target medicine can reduce toxicity, increase curative effect and is expected to solve the problem of drug resistance. In addition, multi-target drugs can avoid the defects of drug combination, such as drug-drug interaction, complex pharmacokinetic properties, incapability of ensuring that two (more) drugs reach treatment targets, poor patient compliance and the like.
Disclosure of Invention
The purpose of the invention is as follows: the invention aims to provide an indolone compound and a pharmaceutical composition thereof, wherein the indolone compound has the advantages of good curative effect, good drug resistance and the like of an anticancer drug.
The invention also aims to provide the indolone compounds with the advantages of good curative effect, good drug resistance and the like of the anticancer drugs, and a preparation method and application of the pharmaceutical composition thereof.
The technical scheme is as follows: the invention provides indolone compounds with a structure shown in a general formula I or pharmaceutically acceptable salts, solvates, prodrugs, stereoisomers, tautomers or metabolites thereof,
Figure BDA0003265645630000022
in the formula:
l is-CH2OCH2CH2OCH2-, or unsubstituted or R1Substituted C2-10An alkyl group;
R1is hydroxy, amino, cyano, halogen or C1-6Alkyl groups of (a);
e is O, S or N-C ≡ N;
x is a single bond, C2-4Olefinic bond, C1-4Alkyl, cyclopropyl, -NHCH2-;
R is unsubstituted or R2Substituted C6-10Aryl, unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, 5-10 membered heteroaryl with 1-3 heteroatoms, or unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and the heteroatom number is 1-3, namely 5-10 membered heterocycloalkyl;
R2、R3and R4Independently is deuterium, halogen, hydroxy, amino, C1-6Alkyl radical, C1-6Alkoxy or C1-6A haloalkyl group.
Further, when L is unsubstituted or R1Substituted C1-10When it is alkyl, said R1Is one or more, when there are more than one R1When R is said1Can be the sameOr different;
and/or, when L is unsubstituted or R1Substituted C2-10When alkyl, said C2-10Alkyl is C2-8An alkyl group;
and/or when R1Is C1-6When there is an alkyl group, said C1-6Alkyl is C1-3An alkyl group;
and/or when R1When the halogen is fluorine, chlorine, bromine or iodine;
and/or, when E is O, X is a single bond, C1-4Alkyl radical, C2-4Alkenyl, cyclopropyl or-NHCH2-, or, when E is S, X is-NHCH2-or, alternatively, when E is N-C ≡ N, X is a single bond;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, R is3Is one or more, when there are more than one R3When R is said3May be the same or different;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, when the heteroatom is 5-10-membered heteroaryl with 1-3 heteroatoms, the heteroaryl is one or more of N, O and S, the heteroatom is 1-3, and the number of N atoms is at least 1, namely the 5-10-membered heteroaryl;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heterocycloalkyl with 1-3 heteroatoms, R is4Is one or more, when there are more than one R4When R is said4May be the same or different;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, when the heteroatom is 5-10 membered heterocycloalkyl with 1-3 heteroatoms, the heterocycloalkyl is one or more of N, O and S, the heteroatom is 1-3, and the number of N atoms is at least 1, namely 5-10 membered heterocycloalkyl;
and/or when R3Is halogen, and the halogen is fluorine, chlorine, bromine or iodine;
and/or when R3Is C1-6When alkyl, said C1-6Alkyl is C1-3An alkyl group;
and/or when R3Is C1-6At alkoxy, said C1-6Alkoxy is C1-3An alkoxy group;
and/or when R3Is C1-6When halogenated alkyl, said C1-6Haloalkyl being C1-3A haloalkyl group;
and/or when R4Is C1-6When alkyl, said C1-6Alkyl is C1-3An alkyl group;
and/or when R4Is C1-6When halogenated alkyl, said C1-6Haloalkyl being C1-3A haloalkyl group.
Further, when L is unsubstituted or R1Substituted C1-10When it is alkyl, said R1The number of (a) is 1, 2 or 3;
and/or, when L is unsubstituted or R1Substituted C2-10When alkyl, said is unsubstituted or R1Substituted C2-10Alkyl is C4-7An alkyl group;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, R is3The number of (a) is 1, 2 or 3;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the number of the heteroatoms is 1-3, the heteroaryl is 5-10-membered heteroaryl, the heteroaryl is pyridyl, pyridopyrrolyl, pyridoimidazolyl, pyridofuryl, pyrazolothiophenyl or pyrazolopyrazolyl;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3, the R is 5-10 membered heterocycloalkyl4The number of (a) is 1, 2 or 3;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3, the heterocyclic alkyl is 5-10 membered heterocyclic alkyl
Figure BDA0003265645630000041
And/or when R3Is halogen, and the halogen is fluorine;
and/or when R3Is C1-6When there is an alkyl group, said C1-6Alkyl of (a) is methyl, ethyl, propyl or isopropyl, preferably methyl;
and/or when R3Is C1-6Alkoxy of (2), said C1-6Alkoxy of (a) is methoxy, ethoxy or propoxy, preferably methoxy;
and/or when R3Is C1-6When halogenated alkyl, said C1-6Haloalkyl is-CF3
And/or when R4Is C1-6When alkyl, said C1-6Alkyl is methyl, ethyl or propyl, preferably methyl;
and/or when R4Is C1-6When halogenated alkyl, said C1-6Haloalkyl is-CF3
Further, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, the substituent is unsubstituted or R3Substituted heteroaryl groups are:
Figure BDA0003265645630000051
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3 and is '5-10 membered heterocycloalkyl', the substituent is unsubstituted or R4Substituted heterocycloalkyl is
Figure BDA0003265645630000052
And/or, when E is O, X is a single bond, R is
Figure BDA0003265645630000053
Or, when E is O, X is-NHCH2-、C2-4Alkenyl or cyclopropyl, R is
Figure BDA0003265645630000054
Or, when E is S, X is-NHCH2-, R is
Figure BDA0003265645630000061
Or, when E is N-C [ identical to ] N, X is a single bond and R is
Figure BDA0003265645630000062
Further, L is unsubstituted C2-10An alkyl group;
and/or E is O, S or N-C ≡ N;
and/or X is a single bond, C2-4Olefinic bond, C1-4Alkyl, cyclopropyl, -NHCH2-;
And/or R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, 5-10 membered heteroaryl with 1-3 heteroatoms, or unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and the heteroatom number is 1-3, namely 5-10 membered heterocycloalkyl;
and/or, R3Is halogen, amino or C1-6An alkyl group;
and/or, R4Is amino or C1-6An alkyl group.
Further, the air conditioner is provided with a fan,
the indolone compound or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer or metabolite thereof is any one of the following compounds:
Figure BDA0003265645630000071
Figure BDA0003265645630000081
Figure BDA0003265645630000091
Figure BDA0003265645630000101
further, the preparation method of the indolone compounds is characterized by comprising the following steps: the method comprises the following steps:
Figure BDA0003265645630000111
in a solvent, under the action of alkali and a condensing agent, carrying out condensation reaction on a compound shown as a formula II and a compound shown as a formula III, wherein L, E, X and R are defined as before.
A pharmaceutical composition comprising an effective amount of a compound of formula I as described in any one of claims 1-6, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, and a pharmaceutically acceptable carrier. The invention also provides application of the pharmaceutical composition in preparing a medicament for preventing and/or treating tumors, wherein the tumors comprise but are not limited to breast cancer, ovarian cancer, prostate cancer, colon cancer, gastric cancer, non-small cell lung cancer, glioma, renal cancer, pancreatic cancer, liver cancer, melanoma, leukemia and cervical cancer, and preferably breast cancer, ovarian cancer, prostate cancer, colon cancer, gastric cancer, non-small cell lung cancer, melanoma, leukemia and cervical cancer.
An indolone compound with a structure shown in a general formula I or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer or metabolite thereof in the preparation of a medicament for preventing and/or treating tumors.
Further, the tumor is one or more of breast cancer, ovarian cancer, prostate cancer, colon cancer, gastric cancer, non-small cell lung cancer, glioma, renal cancer, pancreatic cancer, liver cancer, melanoma, leukemia and cervical cancer. Preferred are breast cancer, prostate cancer, colon cancer, non-small cell lung cancer, leukemia and renal cancer.
The pharmaceutically acceptable carrier may be those adjuvants which are widely used in the field of pharmaceutical production. The excipients are used primarily to provide a safe, stable and functional pharmaceutical composition and may also provide methods for dissolving the active ingredient at a desired rate or for promoting the effective absorption of the active ingredient after administration of the composition by a subject. The pharmaceutical excipients may be inert fillers or provide a function such as stabilizing the overall pH of the composition or preventing degradation of the active ingredients of the composition. The pharmaceutical excipients may include one or more of the following excipients: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, adhesives, disintegrating agents, lubricants, antiadherents, glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, reinforcing agents, adsorbents, buffering agents, chelating agents, preservatives, colorants, flavoring agents and sweeteners.
The pharmaceutical compositions of the present invention may be prepared according to the disclosure using any method known to those skilled in the art. For example, conventional mixing, dissolving, granulating, emulsifying, levigating, encapsulating, entrapping or lyophilizing processes.
The pharmaceutical compositions of the present invention may be administered in any form, including injection (intravenous), mucosal, oral (solid and liquid formulations), inhalation, ocular, rectal, topical or parenteral (infusion, injection, implant, subcutaneous, intravenous, intraarterial, intramuscular) administration. The pharmaceutical compositions of the present invention may also be in a controlled release or delayed release dosage form (e.g., liposomes or microspheres). Examples of solid oral formulations include, but are not limited to, powders, capsules, caplets, soft capsules, and tablets. Examples of liquid formulations for oral or mucosal administration include, but are not limited to, suspensions, emulsions, elixirs and solutions. Examples of topical formulations include, but are not limited to, emulsions, gels, ointments, creams, patches, pastes, foams, lotions, drops or serum formulations. Examples of formulations for parenteral administration include, but are not limited to, solutions for injection, dry preparations which can be dissolved or suspended in a pharmaceutically acceptable carrier, suspensions for injection, and emulsions for injection. Examples of other suitable formulations of the pharmaceutical composition include, but are not limited to, eye drops and other ophthalmic formulations; aerosol: such as nasal sprays or inhalants; liquid dosage forms suitable for parenteral administration; suppositories and lozenges.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are salts of amino acids such as arginine and the like, and salts of organic acids such as glucuronic acid and the like. Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt. Preferably, the neutral form of the compound is regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound differs from the various salt forms by certain physical properties, such as solubility in polar solvents.
The term "isomer" refers to two or more compounds that are identical in molecular composition but differ in structure and properties.
The term "racemate" refers to an equimolar mixture of a chiral molecule and its enantiomer having optical activity, which is formed by mixing equal amounts of molecules having opposite optical rotation directions and the same optical activity, and whose optical activity is offset by the interaction between these molecules, and thus is optically inactive.
The term "solvate" refers to a mixture of a compound and a solvent, e.g., a crystal is a solvate.
The term "metabolite" refers to a pharmaceutically active product produced by the in vivo metabolism of a compound of formula I or a salt thereof. Such products may result from, for example, oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, glucuronidation (glucuronidation), enzymatic cleavage, etc. of the administered compound. Accordingly, the invention includes metabolites of the compounds of the invention, including compounds produced by a method comprising contacting a compound of the invention with a mammal for a period of time sufficient to obtain a metabolite thereof.
Identification of metabolites is typically accomplished by making a radiolabel of a compound of the invention (e.g.,14c or3H) Isotopes, which are administered parenterally to an animal, such as a rat, mouse, guinea pig, monkey, or human, in detectable doses (e.g., greater than about 0.5mg/kg), allowing sufficient time for metabolism to occur (typically about 30 seconds)To 30 hours) and isolating its conversion products from urine, blood or other biological samples. These products are easy to isolate because they are labelled (others are isolated by using antibodies capable of binding to epitopes present in the metabolite). Metabolite structure is determined in a conventional manner, e.g., by MS, LC/MS or NMR analysis. Typically, analysis of metabolites is performed in the same manner as conventional drug metabolism studies well known to those skilled in the art. Metabolite products are useful in assays for the administration of therapeutic doses of the compounds of the invention, provided that they are not otherwise detectable in vivo. The compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be labelled with radioactive isotopes, such as tritium (A), (B), (C) and C)3H) Iodine-125 (125I) Or C-14(14C) In that respect All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
In addition to salt forms, the compounds provided herein also exist in prodrug forms. Prodrugs of the compounds described herein readily undergo chemical changes under physiological conditions to convert to the compounds of the present invention. Any compound that can be converted in vivo to provide a biologically active substance (i.e., a compound of formula I) is a prodrug within the scope and spirit of the present invention. For example, compounds containing a carboxyl group may form physiologically hydrolyzable esters that act as prodrugs by hydrolyzing in vivo to give the compounds of formula I themselves. The prodrugs are preferably administered orally, since hydrolysis in many cases takes place mainly under the influence of digestive enzymes. Parenteral administration may be used when the ester itself is active or hydrolysis occurs in the blood.
The "substitution" in the present invention may be one or more, and when there are a plurality of "substitutions", the "substitutions" may be the same or different.
The term "plurality" means 2, 3, 4 or 5. For example, the term "alkyl" refers to a straight or branched chain saturated hydrocarbon group having the specified number of carbon atoms. Representative saturated hydrocarbon groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, n-pentyl, tert-butyl, neopentyl, n-hexyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 4-methyl-2-pentyl. It should be noted that, when the number of carbon atoms thereof is not particularly limited, it refers only to the number of carbon atoms of the alkyl moiety specified therein, and does not include the number of carbon atoms on the substituent of the alkyl group.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "alkoxy" refers to the group-O-RYWherein R isYIs an alkyl group as defined above.
The term "cycloalkyl" refers to a saturated monocyclic or polycyclic alkyl group. The monocyclic cycloalkyl group is preferably a monovalent saturated cyclic alkyl group having 3 to 7 ring carbon atoms, more preferably 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. Each ring of the polycyclic cycloalkyl is saturated and can be a bicyclic or tricyclic cycloalkyl having 4 to 10 carbon atoms.
The term "heterocycloalkyl" as used herein means a bicyclic ring formed by fusing a saturated monocyclic ring having a heteroatom or a saturated monocyclic ring containing a heteroatom with a heteroaryl, and when the term "heterocycloalkyl" is a bicyclic ring formed by fusing a saturated monocyclic ring containing a heteroatom with a heteroaryl, the "heterocycloalkyl" is linked to other fragments or groups in the compound of formula I through a saturated monocyclic ring containing a heteroatom.
The term "aryl" refers to an aromatic group having the indicated number of carbon atoms, preferably a monocyclic, bicyclic or tricyclic aromatic group, each of which, when bicyclic or tricyclic, satisfies the huckel rule. C of the invention6-10The aryl group of (b) means an aromatic group having 6 to 10 carbon atoms, such as phenyl or naphthyl.
The term "heteroaryl" refers to an aromatic group containing a heteroatom, preferably an aromatic 5-6 membered monocyclic or 9-10 membered bicyclic ring containing 1, 2 or 3 members independently selected from nitrogen, oxygen and sulfur. The 5-to 6-membered monocyclic ring includes, but is not limited to, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, 1, 2, 3-triazolyl, 1, 2, 4-triazolyl, furazanyl, 1, 2, 3-oxadiazolyl, 1, 2, 4-oxadiazole, 1, 2, 5-oxadiazole, 1, 3, 4-oxadiazole, thiadiazolyl, dithiazolyl, tetrazolyl, pyridyl, pyranyl, thiopyranyl, diazinyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazinyl, thiazinyl, dioxinyl, dithiinyl, 1, 2, 3-triazinyl, 1, 2, 4-triazinyl, 1, 3, 5-triazinyl, or tetrazinyl. The 9-to 10-membered bicyclic ring comprises, but is not limited to, benzimidazolyl, benzisothiazolyl, benzoxazolyl, imidazopyridine, thiazolopyridine, furopyridine and tetrahydropyrrolopyridine.
Has the advantages that: compared with the prior art, the invention has the following advantages:
1. the compounds of the present invention have good inhibitory activity against VEGFR, especially VEGFR 2.
2. Meanwhile, the compound has good inhibitory activity on NAMPT.
3. The compound has VEGFR and NAMPT dual inhibitory activity, has a good treatment effect on tumors compared with a single-target inhibitor, has fewer toxic and side effects, and can solve the problem of drug resistance.
Detailed Description
Example 1: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (2- ((E) -3- (pyridin-3-yl) acrylamido) ethyl) -1H-pyrrole-3-carboxamide (S1)
Figure BDA0003265645630000141
The method comprises the following steps: (E) synthesis of (E) -N- (2-aminoethyl) -3- (pyridin-3-yl) acrylamide hydrochloride (4a)
(E) -3- (3-pyridine) acrylic acid 1a (500mg, 3.35mmol) was weighed, dissolved in anhydrous DMF (10mL), EDCI (1.1g, 5.7mmol), HOBt (595mg, 4.4mmol), TEA (0.92mL, 6.7mmol) and compound 2a (536.8mg, 3.35mmol) were added in this order, the reaction was stirred at room temperature for 5h, stopped, and dried under reduced pressureDMF. 5ml of saturated NaHCO was added to the reaction residue3Solution, DCM (20 ml. times.3), combined organic phases, saturated NH4Cl (10ml) was washed 1 time, saturated brine (10ml) was washed 1 time, dried over anhydrous sodium sulfate, filtered, the solvent was evaporated to dryness, and subjected to sand making and column chromatography separation and purification (DCM: MeOH ═ 60: 1 to 20: 1) to give white solid 3a (800mg, 82%).
1H NMR(500MHz,Chloroform-d)δ9.03(t,J=1.9Hz,1H),8.63(dt,J=2.0,3.8Hz,1H),7.99(dt,J=2.0,7.8Hz,1H),7.85(t,J=4.3Hz,1H),7.62-7.54(m,2H),6.59(d,J=15.9Hz,1H),6.01(t,J=4.4Hz,1H),3.45(m,2H),3.37(m,2H),1.43(s,9H).13C NMR(125MHz,Chloroform-d)δ167.88,156.13,150.24,149.69,138.64,134.35,129.74,123.33,121.23,79.58,39.47,39.02,28.32.
Weighing 300mg of the white solid, adding 2mL of ethyl acetate, adding 2mL of HCl/ethyl acetate solution (3M) into the suspension, stirring at room temperature for 3h, directly filtering after complete reaction, washing a filter cake with ethyl acetate, and drying in vacuum to obtain hydrochloride 4a, wherein the hydrochloride is directly reacted in the next step without purification.
Step two: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (2- ((E) -3- (pyridin-3-yl) acrylamido) ethyl) -1H-pyrrole-3-carboxamide (S1)
To hydrochloride salt 4a (80mg, 0.30mmol) was added anhydrous DMF (2mL) and TEA (0.21mL, 1.5mmol), EDCI (98mg, 0.51mmol), HOBt (53mg, 0.39mmol), and Compound 5a (90mg, 0.3mmol) were added sequentially, the reaction was stirred at room temperature for 10h, the reaction was stopped, and the DMF was dried under reduced pressure. To the reaction residue was added 2ml of saturated NaHCO3 solution, DCM (5ml × 3), and the organic phases were combined, washed 1 time with saturated NH4Cl (2ml), 1 time with saturated brine (2ml), dried over anhydrous sodium sulfate, filtered, the solvent was evaporated to dryness, granulated, and purified by column chromatography (DCM: MeOH ═ 60: 1 to 20: 1) to give S1(92mg, 65%) as a pale yellow solid.
Figure BDA0003265645630000151
1H NMR(500MHz,DMSO-d6)δ13.71(s,1H),10.88(s,1H),8.79(d,J=2.3Hz,1H),8.58(dd,J=1.6,4.8Hz,1H),8.30(d,J=6.3Hz,1H),8.02(dt,J=2.0,8.1Hz,1H),7.77(dd,J=2.5,9.4Hz,1H),7.73(s,1H),7.68(s,1H),7.51(d,J=15.9Hz,lH),7.47(dd,J=4.8,8.0Hz,1H),6.95(td,J=2.6,9.1Hz,1H),6.87(dd,J=4.5,8.4Hz,1H),6.77(d,J=15.9Hz,1H),3.41(q,J=3.1Hz,4H),2.47(s,3H),2.45(s,3H).13C NMR(125MHz,DMSO-d6)δ169.85,167.61,164.85,159.10,150.15,149.62,138.64,137.01,136.60,134.33,129.93,129.70,127.81,124.87,124.73,123.93,123.34,120.28,116.72,116.52,110.08,107.45,40.17,39.40,13.95,10.75.
Example 2: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (5- (3- (pyridin-3-ylmethyl) ureido) pentyl) -1H-pyrrole-3-carboxamide (S11)
Figure BDA0003265645630000161
The method comprises the following steps: synthesis of 3- (isocyanatomethyl) -pyridine (7a)
Compound 6a (300mg, 2.86mmol) was weighed out and dissolved in dry THF (5mL) under nitrogen and TEA (0.8mL, 5.72mmol) was added. Triphosgene (339mg, 1.14mmol) was dissolved in 3mL of anhydrous THF at 0 deg.C, added dropwise slowly to the above solution, transferred to room temperature, heated to 50 deg.C, and reacted overnight. After the reaction is completed, the solvent is removed by rotation under reduced pressure, and the mixture is directly put into the next reaction.
Step two: synthesis of tert-butyl (Z) - (5- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide) pentyl) carbamate (9a)
Compound 5a (900mg, 3.00mmol) was weighed out, dissolved in anhydrous DMF (10mL), EDCI (978mg, 5.1mmol), HOBt (527mg, 3.9mmol), TEA (0.83mL, 6mmol) and compound 8a (604mg, 3.00mmol) were added in that order, and the reaction was stirred at room temperature for 6h, stopStopping the reaction, and spin-drying DMF under reduced pressure. 5ml of saturated NaHCO was added to the reaction residue3Solution, DCM (20 ml. times.3), combined organic phases, saturated NH4Cl (10ml) is washed for 1 time, saturated saline (10ml) is washed for 1 time, anhydrous sodium sulfate is dried, filtered, the solvent is evaporated to dryness, sand is prepared, and column chromatography separation and purification (DCM: MeOH is 60: 1-20: 1) are carried out to obtain 9a1.23g (85%) of white solid.
1H NMR(500MHz,Chloroform-d)δ13.34(s,1H),9.92(s,1H),8.51(t,J=4.3Hz,1H),7.72(dd,J=2.6,8.0Hz,1H),7.43(s,1H),6.95(td,J=2.6,8.2Hz,1H),6.85(dd,J=4.9,8.4Hz,1H),5.42(t,J=4.4Hz,1H),3.10(m,4H),2.37(s,3H),2.27(s,3H),1.63-1.54(m,2H),1.55-1.37(m,4H),1.41(s,9H).13C NMR(125MHz,Chloroform-d)δ169.85,164.93,160.08,157.14,136.94,136.58,129.98,127.84,124.73,123.94,122.27,116.83,116.51,110.06,107.52,79.70,40.35,40.02,28.64,28.60,28.33,24.69,13.94,10.75.
Step three: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (5- (3- (pyridin-3-ylmethyl) ureido) pentyl) -1H-pyrrole-3-carboxamide (S11)
Weighing 9a (242mg, 0.5mmol), adding 1mL ethyl acetate, adding 1mL HCl/ethyl acetate solution (3M) into the suspension, stirring at room temperature for 6h, directly filtering after the reaction is completed, washing the filter cake with ethyl acetate, and drying in vacuum to obtain hydrochloride, wherein the hydrochloride is directly reacted in the next step without purification. To the hydrochloride was added 5mL of anhydrous THF and TEA (0.42mL, 3.00mmol) was added. The prepared isocyanate 7a was dissolved in anhydrous THF (2mL), added dropwise to the above solution slowly at 0 ℃ and transferred to room temperature for overnight reaction. The solvent is removed under reduced pressure, DCM (10mL) is added to dissolve the solvent, the mixture is washed once with water (10mL), saturated brine is once with water (10mL), the mixture is dried by anhydrous sodium sulfate, the filtration is carried out, the solvent is evaporated to dryness, sand is prepared, and the white solid S11 is obtained after column chromatography separation and purification (DCM: MeOH is 60: 1-20: 1).
Figure BDA0003265645630000171
1H NMR(500MHz,DMSO-d6)δ12.87(s,1H),9.92(s,1H),8.62(t,J=1.9Hz,1H),8.54-8.45(m,2H),7.75-7.66(m,2H),7.43(s,1H),7.27(dd,J=3.5,7.9Hz,1H),6.94(td,J=2.7,8.1Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),6.36(t,J=6.1Hz,1H),5.97(t,J=4.4Hz,1H),4.50(d,J=6.2Hz,2H),3.10(m,4H),2.37(s,3H),2.26(s,3H),1.62-1.48(m,4H),1.46-1.36(m,2H).13C NMR(125MHz,DMSO-d6)δ170.24,165.10,160.07,158.82,148.59,148.42,137.01,136.61,135.15,133.74,129.98,127.84,124.73,123.94,123.81,122.27,116.86,116.51,110.06,107.62,42.01,40.05,38.56,28.68,28.21,24.53,13.94,10.75.
Example 3: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (5- (3- (pyridin-3-ylmethyl) thioureido) pentyl) -1H-pyrrole-3-carboxamide (S13)
Figure BDA0003265645630000181
Weighing 9a (242mg, 0.5mmol), adding 1mL ethyl acetate, adding lmL HCl/ethyl acetate solution (3M) into the suspension, stirring at room temperature for 6h, directly filtering after the reaction is completed, washing a filter cake with ethyl acetate, and drying in vacuum to obtain hydrochloride, wherein the hydrochloride is directly reacted in the next step without purification. To the hydrochloride was added 5mL of anhydrous THF and TEA (0.42mL, 3.00mmol) was added. A solution of thiocarbonyldiimidazole 10a (134mg, 0.75mmol) in anhydrous THF (2mL) was added slowly dropwise to the solution at 0 deg.C over 15 minutes while protected with N2 and stirred for 2 hours. A solution of 6a (108mg, 1.00mmol) in anhydrous THF (2mL) was slowly added dropwise to the solution, and the temperature was raised to 60 ℃ for 4 h. The reaction was cooled to room temperature, the solvent was spun off under reduced pressure, DCM (10mL) was added to dissolve, water washed once (10mL), saturated brine washed once (10mL), dried over anhydrous sodium sulfate, filtered, the solvent was evaporated to dryness, sand was prepared, and column chromatography separation and purification (DCM: MeOH ═ 60: 1-20: 1) gave S13 as a white solid (197mg, 73%).
Figure BDA0003265645630000182
1H NMR(500MHz,DMSO-d6)δ13.01(s,1H),9.92(s,1H),8.59(t,J=1.9Hz,1H),8.54-8.43(m,2H),7.72(dd,J=2.6,7.9Hz,1H),7.65(dt,J=1.8,7.9Hz,1H),7.43(s,1H),7.29-7.22(m,2H),6.98-6.90(m,2H),6.84(dd,J=5.0,8.4Hz,1H),4.89(d,J=6.2Hz,2H),3.32(td,J=4.4,6.3Hz,2H),3.12(td,J=4.4,6.3Hz,2H),2.37(s,3H),2.26(s,3H),1.70-1.55(m,4H),1.48-1.38(m,2H).13C NMR(125MHz,DMSO-d6)δ180.50,170.24,165.10,160.07,148.56,148.42,137.01,136.58,135.51,135.14,129.98,127.85,124.73,124.04,123.94,122.27,116.91,116.53,110.00,107.62,45.03,42.84,40.05,28.67,28.07,24.58,13.94,10.75.
Example 4: synthesis of N- (5- ((E) -2-cyano-3- (pyridin-3-yl) guanidino) pentyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S15)
Figure BDA0003265645630000191
The method comprises the following steps: synthesis of methyl (Z) -N' -cyano-N- (pyridin-3-yl) aminomethylthiomethyl ester (13a)
NaH (500g, 1.3mmol) was weighed, 10mL DMF was added, the suspension was stirred at 0 ℃ for 15min, then Compound 11a (1g, 1.1mmol) was slowly added dropwise to the suspension, and the reaction was stirred at room temperature for 30 min. Compound 12a (1.6g, 1.06mmol) was dissolved in 5mL DMF and added to the suspension and reacted overnight at room temperature. Washed 3 times with diethyl ether/petroleum ether (5: 1). Glacial acetic acid was added to adjust the pH to 8 at 0 deg.C, filtered and the filter cake was dried under vacuum to give 13a as a pale yellow solid (1.4g, 68%).
1H NMR(500MHz,Chloroform-d)δ8.67(t,J=1.8Hz,1H),8.14(dt,J=1.9,3.7Hz,1H),7.52(dt,J=2.0,7.9Hz,1H),7.42(dd,J=3.5,7.8Hz,1H),2.45(s,3H).13C NMR(125MHz,Chloroform-d)δ165.73,144.50,144.15,135.90,127.58,124.81,116.10,13.22.
Step two: synthesis of tert-butyl (Z) - (5- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide) pentyl) carbamate (9a)
See example 2, step two.
Step three: synthesis of N- (5- ((E) -2-cyano-3- (pyridin-3-yl) guanidino) pentyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S15)
Weighing 9a (242mg, 0.5mmol), adding 1mL ethyl acetate, adding 0.5mL HCl/ethyl acetate solution (3M) into the suspension, stirring at room temperature for 3h, directly filtering after the reaction is completed, washing a filter cake with ethyl acetate, and drying in vacuum to obtain a hydrochloride, wherein the hydrochloride is directly reacted in the next step without purification. The hydrochloride was dissolved in 10mL of pyridine, DMAP (61mg, 0.5mmol) and TEA (0.28mL, 2.0mmol) were added in this order, and the mixture was heated to 50 ℃ for overnight reaction. After the reaction is finished, cooling to room temperature, removing pyridine by spinning under reduced pressure, adding EA (20mL) for dissolving, washing once (10mL) with water, washing once (10mL) with saturated salt solution, drying with anhydrous sodium sulfate, filtering, evaporating the solvent to dryness, preparing sand, and performing column chromatography separation and purification (DCM: MeOH is 60: 1-20: 1) to obtain a white solid S15.
Figure BDA0003265645630000201
1H NMR(500MHz,DMSO-d6)δ12.99(s,1H),9.93(s,1H),9.55(s,1H),8.68(t,J=1.6Hz,1H),8.51(t,J=4.4Hz,1H),8.19-8.13(m,1H),7.72(dd,J=2.6,7.9Hz,1H),7.50-7.40(m,3H),7.23(t,J=4.4Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.54(td,J=4.3,6.3Hz,2H),3.11(td,J=4.4,6.4Hz,2H),2.37(s,3H),2.25(s,3H),1.60(p,J=6.6Hz,2H),1.56-1.47(m,2H),1.43-1.33(m,2H).13C NMR(125MHz,DMSO-d6)δ170.24,165.10,160.07,154.43,144.49,142.36,137.21,136.95,136.58,129.98,127.78,125.98,124.73,124.61,123.94,122.27,118.00,116.70,116.53,110.00,107.62,41.36,40.05,28.69,27.89,24.36,13.94,10.76.
Example 5: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- (2- (pyridin-3-yl) cyclopropyl-1-carboxamido) hexyl) -1H-pyrrole-3-carboxamide (S29)
Figure BDA0003265645630000211
The method comprises the following steps: (E) synthesis of (E) -N-methoxy-N-methyl-3- (pyridin-3-yl) acrylamide (15a)
1a (1.0g, 6.71mmol) and 14a (1.3g, 13.22mmol) were dissolved in DCM (10mL), EDCI (1.3g, 6.71mmol) and DMAP (820mg, 6.71mmol) were added to the above solution in order, and the reaction was carried out at room temperature for 2 h. After the reaction is finished, 5mL of EDCM is added to dilute the reaction solution, washed once with water (5mL), washed once with saturated salt (10mL), dried with anhydrous sodium sulfate, filtered, and the solvent is evaporated to obtain a yellow oily substance 15a, which is directly put into the next reaction.
Step two: synthesis of N-methoxy-N-methyl 2- (pyridin-3-yl) cyclopropyl-1-carboxamide (17a)
NaH (640mg, 16mmol) was added to a solution of trimethyl sulfoxide iodide (2.2g, 10mmol) in DMSO (10mL) at 0 deg.C, then transferred to room temperature and stirred for 1 h. Feed 15a (960mg, 5mmol) was added to the above solution and stirred for 1 h. After the reaction is completed, saturated NH is added into the reaction4Cl (8mL), ethyl acetate extraction (10mL × 3), combined organic phases, washed with water (10mL × 3), dried over anhydrous sodium sulfate, filtered, solvent evaporated, sand dried, and column chromatography purification (DCM: MeOH ═ 5: 1) afforded 17a (824mg, 80%) as a yellow oil.
Step three: synthesis of 2- (pyridin-3-yl) cyclopropyl-1-carboxylic acid (18a)
KOH (647mg, 11.6mmol) was dissolved in 10mL of water, and then aqueous KOH was added to a solution of 17a (800g, 3.88mmol) in ethanol (15mL) and stirred at room temperature for 24 h. After the reaction was complete, 10mL of water was added, DCM (3X 5mL) was extracted, the pH of the aqueous phase was adjusted to 6.0 with 12M HCl solution, then water was spun off under reduced pressure, the solid obtained by vacuum drying was slurried with methanol (20mL), the insoluble solid was removed by filtration to give a suction filtration, and the filtrate was concentrated to give a pale yellow solid. Recrystallization from ethyl acetate/methanol (5: 1) gave pure 18a (341mg, 54%).
Step four: synthesis of 2- (pyridin-3-yl) cyclopropyl-1-carbonyl chloride (19a)
To a solution of 18a (341mg, 2.1mmol) in DCM was added thionyl chloride (0.35mL, 4.8mmol) and the temperature was raised to 40 ℃. And reacting for 5 hours. After the reaction is completed, the solvent is removed by spinning off to obtain an off-white crude product 19a, which is directly put into the next reaction.
Step five: synthesis of tert-butyl (Z) - (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide) hexyl) carbamate (21a)
See step two of example 2, only need to change the corresponding raw materials.
Step six: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- (2- (pyridin-3-yl) cyclopropyl-1-carboxamido) hexyl) -1H-pyrrole-3-carboxamide (S29)
21a (215mg, 0.43mmol) was weighed, 1mL ethyl acetate was added, 1mL HCl/ethyl acetate solution (3M) was added to the suspension, stirred at room temperature for 3h, after the reaction was complete, filtered directly with suction, the filter cake was washed with ethyl acetate and dried in vacuo to give the hydrochloride, which was reacted in the next step without purification. To the hydrochloride was added 5mL of anhydrous DCM and TEA (0.36mL, 2.58 mmol). The above solution was added dropwise to the prepared anhydrous DCM solution of acid chloride 19a at 0 ℃ and the reaction was stirred at room temperature for 3 h. After completion of the reaction, DCM (10mL) was added to dilute the reaction mixture, washed once with water (10mL), washed once with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered and the solvent was evaporated to dryness to give S29(154mg, 66%) as a white solid.
Figure BDA0003265645630000221
1H NMR(500MHz,DMSO-d6)δ13.39(s,1H),9.91(s,1H),8.56(dt,J=1.6,4.8Hz,1H),8.54-8.46(m,2H),7.71(dd,J=2.7,8.0Hz,1H),7.50(dt,J=1.8,7.9Hz,1H),7.41(s,1H),7.31(dd,J=4.7,7.8Hz,1H),7.22(t,J=4.3Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.85(dd,J=5.0,8.4Hz,1H),3.35-3.13(m,3H),3.07(m,1H),2.72(dt,J=6.4,7.9Hz,1H),2.38(s,3H),2.25(s,3H),2.25(dt,J=7.1,7.9Hz,1H),1.70-1.58(m,2H),1.59-1.45(m,2H),1.46-1.19(m,6H).13C NMR(125MHz,DMSO-d6)δ173.63,170.24,165.25,160.08,149.32,148.19,136.95,136.59,134.79,132.59,130.01,127.79,124.73,123.94,123.82,122.26,116.94,116.53,110.03,107.44,40.06,39.71,28.61,28.38,27.33,26.51,26.47,23.95,17.23,13.94,10.76.
Example 6: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (3- ((E) -3- (pyridin-3-yl) acrylamido) propyl) -1H-pyrrole-3-carboxamide (S2)
Figure BDA0003265645630000231
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.70(s,1H),10.88(s,1H),8.79(d,J=2.3Hz,1H),8.57(d,J=5.1Hz,1H),8.25(t,J=5.7Hz,1H),8.01(dt,J=2.0,8.1Hz,1H),7.76(dd,J=2.6,9.4Hz,1H),7.73(s,1H),7.67(t,J=5.8Hz,1H),7.53-7.43(m,2H),6.95(td,J=2.6,9.0Hz,1H),6.88(dd,J=4.5,8.4Hz,1H),6.77(d,J=15.9Hz,1H),3.31(q,J=6.6Hz,4H),2.48(s,3H),2.46(s,3H),1.75(p,J=7.0Hz,2H).13C NMR(125MHz,DMSO-d6)δ170.10,167.65,165.19,159.14,150.15,149.62,138.72,136.94,136.60,134.35,129.98,129.71,127.81,124.73,123.94,123.34,122.27,120.54,116.77,116.52,110.03,107.45,38.49,37.38,27.97,13.95,10.75.
Example 7: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (4- ((E) -3- (pyridin-3-yl) acrylamido) butyl) -1H-pyrrole-3-carboxamide (S3)
Figure BDA0003265645630000232
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced. .1H NMR(500MHz,DMSO-d6)δ13.68(s,1H),11.00(s,1H),8.76(d,J=2.2Hz,1H),8.56(dd,J=1.6,4.8Hz,1H),8.43(d,J=6.2Hz,1H),8.00(d,J=8.1Hz,1H),7.78-7.70(m,3H),7.50-7.42(m,2H),6.92(m,2H),6.86(d,J=15.9Hz,1H),3.25(q,J=6.6Hz,4H),2.44(d,J=10.6Hz,6H),1.62-1.52(m,4H).13C NMR(125MHz,DMSO-d6)δ170.10,167.52,165.10,160.14,150.15,149.62,138.73,137.01,136.61,134.35,129.98,129.71,127.84,124.73,123.94,123.34,122.27,120.63,116.86,116.53,110.00,107.45,39.96,39.45,26.63,26.53,13.95,10.75.
Example 8: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (5- ((E) -3- (pyridin-3-yl) acrylamido) pentyl) -1H-pyrrole-3-carboxamide (S4)
Figure BDA0003265645630000241
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.67(s,1H),10.87(s,1H),8.77(d,J=2.3Hz,1H),8.56(dd,J=1.7,4.7Hz,1H),8.17(t,J=5.7Hz,1H),7.99(dt,J=2.1,8.1Hz,1H),7.79-7.68(m,2H),7.63(t,J=5.8Hz,1H),7.52-7.42(m,2H),6.95(td,J=2.5,9.1,9.5Hz,1H),6.87(dd,J=4.5,8.5Hz,1H),6.75(d,J=15.9Hz,1H),3.24(dt,J=6.5,12.9Hz,4H),2.44(d,J=9.9Hz,6H),1.56(m,4H),1.40(q,J=7.0,7.5Hz,2H).13C NMR(125MHz,DMSO-d6)δ170.24,167.57,165.10,160.07,150.15,149.62,138.73,137.01,136.58,134.28,129.98,129.71,127.78,124.73,123.94,123.34,122.27,120.60,116.86,116.53,110.00,107.46,40.03,39.98,28.68,28.44,24.58,13.94,10.75.
Example 9: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- ((E) -3- (pyridin-3-yl) acrylamido) hexyl) -1H-pyrrole-3-carboxamide (S5)
Figure BDA0003265645630000242
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced. .1H NMR(500MHz,DMSO-d6)δ13.68(s,1H),10.87(s,lH),8.77(d,J=2.2Hz,1H),8.58-8.53(m,1H),8.17(t,J=5.6Hz,lH),7.99(dt,J=2.0,8.0Hz,1H),7.79-7.69(m,2H),7.64(t,J=5.7Hz,lH),7.50-7.42(m,2H),6.94(td,J=2.5,9.1Hz,1H),6.87(dd,J=4.5,8.5Hz,1H),6.75(d,J=15.9Hz,1H),3.24(m,4H),2.45(s,3H),2.43(s,3H),1.59-1.49(m,4H),1.42-1.35(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.57,165.10,160.07,150.15,149.64,138.73,136.95,136.61,134.35,129.98,129.72,127.84,124.73,123.94,123.34,122.27,120.60,116.86,116.53,110.00,107.62,40.06,39.73,28.49,28.41,26.51,26.50,13.94,10.76.
Example 10: synthesis of (Z) -N- (2- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) ethyl) imidazo [1, 2-a ] pyridine-6-carboxamide (S6)
Figure BDA0003265645630000251
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ12.98(s,1H),9.91(s,1H),8.65(d,J=1.8Hz,1H),8.27(m,1H),8.04-7.98(m,1H),7.95(dd,J=1.5,9.1Hz,1H),7.79-7.67(m,3H),7.65(s,1H),7.44(s,1H),6.95(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.1,8.3Hz,1H),3.53-3.42(m,2H),3.42-3.31(m,2H),2.28(s,3H),2.24(s,3H).13C NMR(125MHz,DMSO-d6)δ170.10,166.79,164.98,159.12,140.73,137.01,136.61,134.38,130.90,129.93,127.84,124.87,124.73,123.94,122.15,118.9l,116.85,116.53,113.16,112.73,110.06,107.53,40.13,40.09,13.95,10.75.
Example 11: synthesis of (Z) -N- (3- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) propyl) imidazo [1, 2-a ] pyridine-6-carboxamide (S7)
Figure BDA0003265645630000252
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ12.99(s,1H),9.88(s,1H),8.65(d,J=1.3Hz,1H),8.34(t,J=4.4Hz,1H),8.24(t,J=4.4Hz,1H),7.96(dd,J=1.4,9.1Hz,lH),7.79-7.69(m,2H),7.66(d,J=10.3Hz,2H),7.39(s,lH),6.94(td,J=2.7,8.1Hz,1H),6.84(dd,J=5.0,8.4Hz,lH),3.29(m,4H),2.37(s,3H),2.27(s,3H),1.79(p,J=6.3Hz,2H).13C NMR(125MHz,DMSO-d6)δ170.10,166.96,165.19,160.14,140.73,137.01,136.61,134.38,130.90,129.98,127.78,124.73,123.94,122.27,122.16,118.75,116.70,116.53,113.21,112.73,110.00,107.45,37.43,37.40,27.90,13.94,10.75.
Example 12: synthesis of (Z) -N- (4- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) butyl) imidazo [1, 2-a ] pyridine-6-carboxamide (S8)
Figure BDA0003265645630000261
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.01(s,1H),9.89(s,1H),8.65(d,J=1.4Hz,1H),8.32(t,J=4.4Hz,1H),8.17(t,J=4.3Hz,lH),7.95(dd,J=1.4,9.1Hz,1H),7.79-7.69(m,2H),7.66(d,J=7.9Hz,2H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,lH),3.42-3.33(m,4H),2.30(s,3H),2.25(s,3H),1.68-1.57(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,166.72,165.10,160.07,140.73,136.95,136.58,134.38,130.90,129.98,127.78,124.73,123.94,122.27,122.16,118.86,116.86,116.51,113.21,112.73,110.00,107.62,39.96,39.94,26.96,26.50,13.94,10.75.
Example 13: synthesis of (Z) -N- (5- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) pentyl) imidazo [1, 2-a ] pyridine-6-carboxamide (S9)
Figure BDA0003265645630000262
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.03(s,1H),9.89(s,1H),8.65(d,J=1.3Hz,1H),8.51(t,J=4.4Hz,1H),8.15(t,J=4.4Hz,1H),7.95(dd,J=1.6,9.1Hz,1H),7.78-7.68(m,3H),7.65(s,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.1,8.4Hz,1H),3.14(m,4H),2.31(s,3H),2.25(s,3H),1.64-1.55(m,4H),1.46-1.37(m,2H).13C NMR(125MHz,DMSO-d6)δ170.24,166.74,165.10,158.06,140.73,136.95,136.61,134.38,130.90,129.98,127.84,124.73,123.94,122.27,122.16,118.86,116.86,116.51,113.21,112.73,110.06,107.54,40.11,40.05,28.69,28.58,24.58,13.94,10.76.
Example 14: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) imidazo [1, 2-a ] pyridine-6-carboxamide (S10)
Figure BDA0003265645630000271
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.05(s,1H),9.89(s,1H),8.64(d,J=1.6Hz,1H),8.49(t,J=4.4Hz,1H),8.24(t,J=4.3Hz,1H),7.96(dd,J=1.5,9.2Hz,1H),7.78-7.68(m,2H),7.66(d,J=7.7Hz,2H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.37(td,J=4.4,6.3Hz,2H),3.23(td,J=4.3,6.3Hz,2H),2.28(s,6H),1.70-1.52(m,4H),1.38-1.27(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,166.74,165.25,158.06,140.68,136.95,136.61,134.38,130.90,130.01,127.79,124.73,123.94,122.26,122.16,118.86,116.94,116.53,113.21,112.79,110.09,107.60,40.12,40.06,28.61,28.49,26.52,26.51,13.94,10.76.
Example 15: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- (3- (pyridin-3-ylmethyl) ureido) hexyl) -1H-pyrrole-3-carboxamide (S12)
Figure BDA0003265645630000272
The synthesis method is as in example 2, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ12.88(s,1H),9.93(s,1H),8.64(t,J=1.9Hz,1H),8.50-8.41(m,2H),7.75-7.66(m,2H),7.43(s,1H),7.27(dd,J=3.5,7.9Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),6.24(t,J=6.1Hz,1H),6.06(t,J=4.4Hz,1H),4.53(d,J=6.1Hz,2H),3.23(td,J=4.3,6.3Hz,2H),3.08(td,J=4.3,6.3Hz,2H),2.30(s,3H),2.24(s,3H),1.57(p,J=6.5Hz,2H),1.47-1.38(m,2H),1.39-1.24(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.10,160.07,158.92,148.59,148.38,136.95,136.61,135.14,133.72,129.98,127.85,124.73,123.94,123.81,122.27,116.91,116.53,110.06,107.46,42.01,40.06,38.54,28.62,28.49,26.47,26.43,13.94,10.76.
Example 16: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- (3- (pyridin-3-ylmethyl) thioureido) hexyl) -1H-pyrrole-3-carboxamide (S14)
Figure BDA0003265645630000281
The synthesis method is as in example 3, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.02(s1H),9.93(s,1H),8.60(t,J=1.9Hz,1H),8.48-8.41(m,2H),7.77-7.66(m,2H),7.43(s1H),7.25(dd,J=3.5,7.9Hz,lH),7.15-7.08(m,2H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.3Hz,1H),4.90(d,J=6.1Hz,2H),3.36(td,J=4.3,6.3Hz,2H),3.23(td,J=4.4,6.3Hz,2H),2.35(s,3H),2.25(s,3H),1.57(p,J=6.5Hz,2H),1.53-1.43(m,2H),1.38-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ180.50,170.24,165.25,160.16,148.56148.34,136.95,136.58,135.51,135.14,129.98,127.79,124.73,124.04,123.94,122.26116.95,116.53,110.03,107.52,45.03,42.69,40.06,28.49,28.18,26.53,26.51,13.94,10.76.
Example 17: synthesis of N- (6- ((E) -2-cyano-3- (pyridin-3-yl) guanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S16)
Figure BDA0003265645630000282
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.00(s,1H),9.93(s,1H),9.63(s,1H),8.68(t,J=1.8Hz,1H),8.50(t,J=4.4Hz,1H),8.21(dt,J=1.8,3.5Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.50-7.40(m,3H),7.15(t,J=4.4Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.1,8.4Hz,lH),3.48(td,J=4.4,6.3Hz,2H),3.23(td,J=4.4,6.3Hz,2H),2.38(s,3H),2.26(s,3H),1.52(m,4H),1.32(q,J=3.7Hz,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.25,158.06,154.43,144.49,142.36,137.26,136.95,136.58,130.01,127.79,125.98,124.73,124.61,123.94,122.26,118.00,116.78,116.53,110.03,107.60,41.36,40.06,28.65,28.50,26.46,26.40,13.94,10.76.
Example 18: synthesis of N- (5- ((E) -2-cyano-3- (pyridin-4-yl) guanidino) pentyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S17)
Figure BDA0003265645630000291
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ12.98(s,1H),9.91(s,1H),9.05(s,1H),8.51(t,J=4.4Hz,1H),8.40-8.35(m,2H),7.72(dd,J=2.6,7.9Hz,1H),7.43(s,1H),7.23(t,J=4.4Hz,1H),7.03-6.98(m,2H),6.94(td,J=2.6,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.55(td,J=4.4,6.3Hz,2H),3.11(td,J=4.4,6.4Hz,2H),2.36(s,3H),2.25(s,3H),1.62-1.48(m,4H),1.44-1.34(m,2H).13C NMR(125MHz,DMSO-d6)δ170.24,165.10,160.07,154.85,150.01,149.88,145.16,136.95,136.58,129.98,127.84,124.73,123.94,122.27,118.00,116.70,116.53,112.91,112.87,110.00,107.46,41.36,40.05,28.69,27.89,24.36,13.94,10.76.
Example 19: synthesis of N- (6- ((E) -2-cyano-3- (pyridin-4-yl) guanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S18)
Figure BDA0003265645630000292
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.00(s,1H),9.93(s,1H),9.06(s,1H),8.49(t,J=4.4Hz,1H),8.40-8.35(m,2H),7.71(dd,J=2.7,8.1Hz,1H),7.43(s,1H),7.15(t,J=4.4Hz,1H),7.03-6.98(m,2H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.48(td,J=4.4,6.3Hz,2H),3.23(td,J=4.4,6.3Hz,2H),2.35(s,3H),2.26(s,3H),1.53(m,4H),1.39-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.25,160.08,154.85,150.01,149.88,145.16,136.95,136.58,130.01,127.79,124.73,123.94,122.26,118.00,116.78,116.51,112.91,112.87,110.03,107.44,41.36,40.06,28.65,28.50,26.46,26.40,13.94,10.76.
Example 20: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -1H-pyrrolo [3, 2-c ] pyridine-3-carboxamide (S19)
Figure BDA0003265645630000301
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ12.90(s,1H),9.89(s,1H),9.42(d,J=1.7Hz,1H),9.05(s,1H),8.49(t,J=4.4Hz,1H),8.41(t,J=4.4Hz,1H),8.34(dd,J=1.8,5.7Hz,1H),8.08(d,J=2.6Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.47-7.41(m,2H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.28-3.19(m,4H),2.36(s,3H),2.26(s,3H),1.59(m,4H),1.38-1.29(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.66,165.25,160.08,143.77,143.44,138.85,136.95,136.59,130.07,130.01,127.85,124.73,123.94,122.26,122.16,116.94,116.51,110.03,108.53,108.27,107.60,40.11,40.06,28.65,28.61,26.52,26.51,13.94,10.76.
Example 21: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -3a, 7 a-dihydro-1H-pyrrolo [3, 4-b ] pyridine-6-carboxamide (S20)
Figure BDA0003265645630000302
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.22(s,1H),9.92(s,1H),8.49(t,J=4.4Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.50(t,J=4.4Hz,1H),7.43(s,1H),7.36(d,J=5.5Hz,1H),7.15(d,J=6.6Hz,1H),6.98-6.88(m,2H),6.84(dd,J=5.0,8.4Hz,1H),6.09(dd,J=5.7,9.3Hz,1H),5.26(dd,J=5.1,6.6Hz,1H),3.50(m,1H),3.35-3.06(m,4H),2.37(s,3H),2.25(s,3H),1.70-1.21(m,8H).13C NMR(125MHz,DMSO-d6)δ170.24,166.61,165.25,160.08,153.05,136.95,136.61,134.56,130.01,128.57,127.79,127.44,124.73,123.94,122.26,116.78,116.51,110.03,107.60,74.73,43.28,40.06,40.01,28.61,28.38,26.51,26.48,13.94,10.76.
Example 22: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) furo [2, 3-c ] pyridine-2-carboxamide (S21)
Figure BDA0003265645630000311
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.34(s,1H),9.89(s,1H),8.82(d,J=1.9Hz,1H),8.72(dd,J=1.6,5.5Hz,1H),8.48(t,J=4.3Hz,1H),8.25(t,J=4.4Hz,1H),7.96(d,J=2.2Hz,1H),7.87(dd,J=1.9,5.6Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),3.36(td,J=4.3,6.3Hz,2H),3.24(td,J=4.3,6.3Hz,2H),2.38(s,3H),2.24(s,3H),1.60(m,4H),1.40-1.28(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.25,161.31,158.06,151.19,151.11,144.34,136.95,136.59,134.40,132.74,130.01,127.79,124.73,123.94,122.26,117.52,116.78,116.51,110.56,110.09,107.44,40.06,39.93,28.71,28.61,26.51,26.48,13.94,10.76.
Example 23: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -1H-pyrrolo [2, 3-c ] pyridine-2-carboxamide (S22)
Figure BDA0003265645630000312
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.30(s,1H),9.89(s,1H),9.06(s,1H),8.92(d,J=1.9Hz,1H),8.73(dd,J=1.6,5.5Hz,1H),8.51(dt,J=4.4,24.0Hz,2H),7.79(dd,J=2.0,5.6Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.43(s,1H),7.25(d,J=2.1Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,lH),3.42(td,J=4.4,6.3Hz,2H),3.23(td,J=4.3,6.3Hz,2H),2.36(s,3H),2.24(s,3H),1.60(m,4H),1.40-1.28(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.25,162.74,160.08,139.84,136.95,136.59,135.06,132.37,131.95,131.91,130.01,127.79,124.73,123.94,122.26,116.88,116.78,116.53,110.03,107.68,107.44,40.30,40.06,28.70,28.61,26.51,26.48,13.94,10.76.
Example 24: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -1, 3-dihydro-2H-pyrrolo [3, 4-c ] pyridine-2-carboxamide (S23)
Figure BDA0003265645630000321
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.89(s,1H),8.65(d,J=1.8Hz,1H),8.48(t,J=4.4Hz,1H),8.42(dd,J=1.8,5.7Hz,1H),7.71(dd,J=2.7,8.0Hz,1H),7.43(s,1H),7.32(d,J=5.7Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),6.71(t,J=4.4Hz,1H),4.86(s,2H),4.31(s,2H),3.23(td,J=4.3,6.3Hz,2H),3.16(td,J=4.3,6.3Hz,2H),2.38(s,3H),2.25(s,3H),1.58(p,J=6.5Hz,2H),1.44(p,J=6.5Hz,2H),1.38-1.24(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.25,160.08,158.12,145.95,145.69,143.11,136.95,136.59,132.62,130.01,127.79,124.73,123.94,122.26,116.94,116.78,116.53,110.03,107.44,52.44,52.17,40.06,39.66,28.67,28.61,26.47,26.41,13.94,10.76.
Example 25: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -6-methyl-1, 3-dihydro-2H-pyrrolo [3, 4-c ] pyridine-2-carboxamide (S24)
Figure BDA0003265645630000322
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.34(s,1H),9.89(s,1H),8.52(t,J=4.4Hz,1H),8.42(s,1H),7.73(dd,J=2.6,8.0Hz,1H),7.43(s,1H),7.25(d,J=0.7Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,lH),6.71(t,J=4.4Hz,1H),4.81(s,2H),4.32(s,2H),3.26(td,J=4.4,6.3Hz,2H),3.16(td,J=4.4,6.4Hz,2H),2.58(s,3H),2.37(s,3H),2.26(s,3H),1.60(p,J=6.5Hz,2H),1.49-1.40(m,2H),1.38-1.24(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,160.22,158.12,157.66,147.88,142.02,136.61,136.59,130.33,130.01,127.83,124.73,123.94,122.26,117.25,116.53,116.51,110.05,107.44,52.12,52.07,40.06,39.66,28.68,28.61,26.49,26.41,24.47,13.94,10.76.
Example 26: synthesis of (Z) -6-amino-N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -1, 3-dihydro-2H-pyrrolo [3, 4-c ] pyridine-2-carboxamide (S25)
Figure BDA0003265645630000331
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.36(s,1H),9.89(s,1H),9.06(s,1H),8.52(t,J=4.4Hz,1H),8.17(s,1H),7.73(dd,J=2.6,8.0Hz,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),6.71(t,J=4.4Hz,1H),6.51(s,1H),5.32(d,J=6.0Hz,1H),5.22(d,J=6.0Hz,1H),4.31(d,J=10.2Hz,3H),3.26(td,J=4.4,6.3Hz,2H),3.14(td,J=4.4,6.3Hz,2H),2.36(s,3H),2.25(s,3H),1.60(m,2H),1.49-1.40(m,2H),1.38-1.24(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,160.22,158.42,158.23,148.75,140.58,136.95,136.61,130.01,127.83,124.73,123.94,122.26,121.09,116.95,116.53,110.05,107.44,101.84,52.11,51.90,40.06,39.66,28.68,28.61,26.49,26.41,13.94,10.76.
Example 27: synthesis of (Z) -5-amino-N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) furo [2, 3-c ] pyridine-2-carboxamide (S26)
Figure BDA0003265645630000332
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.89(s,1H),8.55-8.45(m,2H),8.06(t,J=4.4Hz,1H),7.73(dd,J=2.6,8.0Hz,1H),7.47-7.41(m,2H),7.03(d,J=2.2Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.0,8.4Hz,1H),5.34(d,J=6.0Hz,1H),5.22(d,J=6.2Hz,1H),3.35(td,J=4.3,6.3Hz,2H),3.27(td,J=4.4,6.4Hz,2H),2.35(s,3H),2.26(s,3H),1.62(m,4H),1.40-1.28(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,161.27,160.22,152.74,150.07,143.72,136.95,136.59,133.97,133.20,130.01,127.83,124.73,123.94,122.26,116.95,116.53,110.05,108.91,107.60,98.52,40.06,39.93,28.74,28.61,26.52,26.50,13.94,10.76..
Example 28: synthesis of (Z) -N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) thiazolo [2, 3-c ] pyridine-2-carboxamide (S27)
Figure BDA0003265645630000341
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.37(s,1H),9.89(s,1H),9.00(d,J=1.3Hz,1H),8.76(dd,J=1.6,5.6Hz,lH),8.52(t,J=4.4Hz,1H),8.42(t,J=4.4Hz,1H),8.11(dd,J=2.1,5.6Hz,1H),7.91(d,J=1.7Hz,1H),7.73(dd,J=2.6,8.0Hz,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),3.38(td,J=4.3,6.3Hz,2H),3.23(td,J=4.3,6.3Hz,2H),2.38(s,3H),2.25(s,3H),1.60(m,4H),1.34(q,J=3.8Hz,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,164.52,158.20,146.13,144.51,142.88,139.04,136.95,136.92,136.59,130.01,127.83,124.73,124.49,123.94,122.26,119.93,116.79,116.53,110.05,107.44,40.06,40.03,28.61,28.58,26.53,26.52,13.94,10.76.
Example 29: synthesis of (Z) -6-amino-N- (6- (5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamido) hexyl) -1H-pyrrolo [3, 2-c ] pyridine-2-carboxamide (S28)
Figure BDA0003265645630000342
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.37(s,1H),9.99(s,1H),9.89(s,1H),8.72(d,J=1.5Hz,1H),8.52(t,J=4.3Hz,1H),8.24(t,J=4.4Hz,1H),7.73(dd,J=2.7,8.0Hz,1H),7.43(s,1H),7.21(d,J=1.4Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),6.68(s,1H),5.83(d,J=6.0Hz,1H),5.75(d,J=6.2Hz,1H),3.42(td,J=4.4,6.4Hz,2H),3.26(td,J=4.4,6.4Hz,2H),2.37(s,3H),2.25(s,3H),1.70-1.58(m,4H),1.40-1.28(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,162.72,158.20,154.66,144.53,143.15,136.95,136.59,132.14,130.01,127.83,124.73,123.94,122.26,116.95,116.51,115.60,110.05,107.44,104.21,89.88,40.30,40.06,28.70,28.61,26.52,26.50,13.94,10.76.
Example 30: synthesis of N- (6- ((E) -3- (6-aminopyridin-3-yl) acrylamido) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S30)
Figure BDA0003265645630000351
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.39(s,1H),9.93(s,1H),8.51-8.44(m,2H),7.74-7.67(m,2H),7.57(dd,J=1.8,8.4Hz,1H),7.52(d,J=15.9Hz,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.1,8.4Hz,1H),6.58(d,J=15.9Hz,1H),6.19(d,J=8.4Hz,1H),5.93(d,J=6.0Hz,1H),5.85(d,J=6.0Hz,1H),3.21(m,4H),2.37(s,3H),2.26(s,3H),1.58(p,J=6.5Hz,2H),1.50(p,J=6.6Hz,2H),1.38-1.25(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.49,165.25,160.08,159.04,149.26,137.95,136.95,136.61,135.30,130.01,127.79,124.73,123.94,122.26,120.42,118.90,116.94,116.53,110.03,108.72,107.44,40.06,39.74,28.59,28.50,26.51,26.50,13.94,10.76.
Example 31: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- ((E) -3- (6- (trifluoromethyl) pyridin-3-yl) acrylamido) hexyl) -1H-pyrrole-3-carboxamide (S31)
Figure BDA0003265645630000352
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced. 1H NMR (500MHz, DMSO-d6) δ 13.32(s, 1H), 9.93(s, 1H), 8.88(d, J ═ 1.9Hz, 1H), 8.56(t, J ═ 4.3Hz, 1H), 7.94(dd, J ═ 1.9, 8.4Hz, 1H), 7.77-7.69(m, 2H), 7.62-7.55(m, 2H), 7.43(s, 1H), 6.94(td, J ═ 2.7, 8.2Hz, 1H), 6.84(dd, J ═ 4.9, 8.4Hz, 1H), 6.59(d, J ═ 15.7Hz, 1H), 3.22(m, 4H), 2.38(s, 3H), 2.26(s, 3H), 56.56 (s, 1H), 56.59 (d, J ═ 15.7Hz, 1H), 3.22(m, 4H), 2.38(s, 3H), 2.26(s, 3H), 2H), 19.56.56, 56.38 (d, 1H), 38(s, 38H), 366338 (m, 38, 3H), 19, 38, 18H), 19, 18H), 19 (m, 18H), 3H), 18H), 19, 18H), 3H, 18H), 3H, 18H, 3H, 18H, 3H, 18H, 3H, 18H, 3m, 3H, 18H, 3m, 18H, 3m, 3H, 18H, 3H, 18H, 3m, 3H, 18 m, 3H, 3m, 3H, 120.34, 116.79, 116.67, 110.05, 107.44, 40.06, 39.74, 28.58, 28.54, 26.53, 13.92, 10.78.
Example 32: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- ((E) -3- (2-methylpyridin-4-yl) acrylamido) hexyl) -1H-pyrrole-3-carboxamide (S32)
Figure BDA0003265645630000361
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.32(s,1H),9.92(s,1H),8.51-8.45(m,2H),7.78-7.68(m,2H),7.56(d,J=15.9Hz,1H),7.43(s,1H),7.31(dd,J=2.0,5.7Hz,1H),7.18(d,J=2.1Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=5.1,8.4Hz,1H),6.60(d,J=15.9Hz,1H),3.21(m,4H),2.59(s,3H),2.36(s,3H),2.25(s,3H),1.54(m,4H),1.39-1.25(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.38,165.25,160.08,158.06,147.20,141.52,140.70,136.61,136.58,130.01,127.79,124.73,123.94,122.26,121.88,121.36,119.84,116.94,116.51,110.03,107.44,40.06,39.85,28.59,28.50,26.51,26.50,24.21,13.94,10.76.
Example 33: synthesis of (Z) -5- ((5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- (2- (6- (trifluoromethyl) pyridin-3-yl) cyclopropyl-1-carboxamido) hexyl) -1H-pyrrole-3-carboxamide (S33)
Figure BDA0003265645630000362
The synthesis method is as in example 5, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.34(s,1H),9.92(s,lH),8.59-8.50(m,2H),7.73(dd,J=2.7,8.0Hz,1H),7.57-7.47(m,2H),7.43(s,1H),6.98-6.89(m,2H),6.85(dd,J=5.1,8.4Hz,1H),3.31(m,1H),3.19(m,2H),3.05(m,1H),2.59(dt,J=6.4,7.9Hz,1H),2.38(s,3H),2.25(s,3H),1.71-1.61(m,2H),1.64-1.59(m,1H),1.63-1.52(m,1H),1.55-1.45(m,1H),1.48-1.32(m,2H),1.36-1.20(m,4H).13C NMR(125MHz,DMSO-d6)δ173.62,170.24,165.29,160.22,148.81,147.09,136.95,136.63,134.34,134.31,130.01,127.79,124.19,123.94,122.26,120.35,120.18,116.95,116.67,110.05,107.44,40.06,39.70,28.56,28.38,27.33,26.52,26.46,23.99,17.10,13.92,10.78.
Example 34: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -N- (6- ((E) -3- (4-fluoropyridin-3-yl) acrylamido) hexyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S34)
Figure BDA0003265645630000371
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.93(s,1H),8.87(dd,J=1.8,4.9Hz,1H),8.57(m,1H),8.49(t,J=4.4Hz,1H),7.83(t,J=4.3Hz,1H),7.73(dd,J=2.7,8.0Hz,1H),7.64(d,J=15.9Hz,1H),7.43(s,1H),7.12(dd,J=5.6,8.0Hz,lH),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.3Hz,1H),6.54(d,J=15.7Hz,1H),3.20(m,4H),2.36(s,3H),2.25(s,3H),1.58(p,J=6.5Hz,2H),1.49(p,J=6.6Hz,2H),1.38-1.25(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.48,165.25,161.05,158.06,150.56,149.67,136.95,136.58,130.01,128.81,127.85,124.73,123.94,122.68,122.26,117.15,116.79,116.53,110.09,109.77,107.60,40.06,39.74,28.59,28.50,26.51,26.50,13.94,10.76.
Example 35: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-N- (6- ((E) -3- (2- (trifluoromethyl) pyridin-4-yl) acrylamido) hexyl) -1H-pyrrole-3-carboxamide (S35)
Figure BDA0003265645630000372
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.34(s,1H),9.93(s,1H),8.62(d,J=5.6Hz,1H),8.56(t,J=4.3Hz,1H),7.77-7.70(m,2H),7.55(dd,J=6.9,8.9Hz,2H),7.45-7.39(m,2H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),6.52(d,J=15.9Hz,1H),3.22(m,4H),2.38(s,3H),2.25(s,3H),1.70-1.52(m,4H),1.39-1.25(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.57,165.29,158.20,149.11,147.46,141.71,141.48,136.95,136.65,130.01,127.89,124.19,123.94,123.85,122.26,120.31,119.81,119.00,116.95,116.67,110.05,107.44,40.06,39.85,28.58,28.54,26.55,26.53,13.92,10.78.
Example 36: synthesis of 5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -N- (6- ((E) -3- (2-fluoropyridin-4-yl) acrylamido) hexyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S36)
Figure BDA0003265645630000381
The synthesis method is as in example 1, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.93(s,1H),8.51-8.44(m,2H),7.78-7.70(m,2H),7.61-7.53(m,2H),7.43(s,1H),7.35(dd,J=1.9,8.0Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.3Hz,1H),6.61(d,J=15.9Hz,1H),3.20(m,4H),2.36(s,3H),2.25(s,3H),1.58(p,J=6.5Hz,2H),1.49(p,J=6.6Hz,2H),1.39-1.25(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,167.60,165.25,161.18,158.06,147.77,146.43,141.54,136.95,136.58,130.01,127.85,124.73,123.94,122.26,120.81,119.76,116.95,116.51,110.03,107.60,107.16,40.06,39.85,28.59,28.50,26.51,26.50,13.94,10.76.
Example 37: synthesis of N- (6- ((Z) -3- (6-aminopyridin-3-yl) -2-cyanoguanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S37)
Figure BDA0003265645630000382
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.34(s,1H),9.92(s,1H),9.06(s,1H),8.55-8.47(m,2H),7.79(t,J=4.4Hz,1H),7.76-7.70(m,2H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),6.43(d,J=8.4Hz,1H),5.86(d,J=6.0Hz,1H),5.79(d,J=6.0Hz,1H),3.52(td,J=4.3,6.3Hz,2H),3.23(td,J=4.4,6.3Hz,2H),2.38(s,3H),2.25(s,3H),1.53(m,4H),1.39-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,160.22,155.69,154.43,142.00,136.95,136.59,130.01,129.60,127.83,126.19,124.73,123.94,122.26,118.00,116.95,116.51,110.12,108.81,107.60,41.36,40.06,28.65,28.61,26.39,26.38,13.94,10.76.
Example 38: synthesis of N- (6- ((Z) -3- (6- (trifluoromethyl) pyridin-3-yl) -2-cyanoguanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S38)
Figure BDA0003265645630000391
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.96(s,1H),9.06-8.98(m,2H),8.56(t,J=4.3Hz,1H),7.80(t,J=4.4Hz,1H),7.73(dd,J=2.7,8.0Hz,1H),7.59(d,J=8.4Hz,1H),7.43(s,1H),7.10(dd,J=2.0,8.4Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.85(dd,J=5.1,8.3Hz,1H),3.44(td,J=4.4,6.3Hz,2H),3.25(td,J=4.3,6.3Hz,2H),2.37(s,3H),2.25(s,3H),1.55(m,4H),1.39-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.29,160.22,154.42,144.05,141.56,138.18,136.95,136.66,130.01,127.85,125.08,124.19,123.94,122.26,120.10,119.79,118.00,116.95,116.67,110.05,107.44,41.36,40.06,28.61,28.58,26.41,26.40,13.92,10.78.
Example 39: synthesis of N- (6- ((Z) -2-cyano-3- (2-methylpyridin-4-yl) guanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S39)
Figure BDA0003265645630000392
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.36(s,1H),9.92(s,1H),8.91(s,1H),8.52(t,J=4.4Hz,1H),8.26(d,J=5.6Hz,1H),7.76-7.67(m,2H),7.45-7.38(m,2H),7.23(d,J=1.7Hz,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.3Hz,1H),3.48(td,J=4.4,6.4Hz,2H),3.23(td,J=4.4,6.3Hz,2H),2.37(s,3H),2.28(s,3H),2.25(s,3H),1.52(m,4H),1.37-1.27(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,160.22,158.20,154.75,145.68,145.21,136.61,136.59,130.01,127.85,124.73,123.94,122.26,118.00,116.95,116.51,113.68,110.17,110.05,107.60,41.36,40.06,28.65,28.61,26.40,26.38,24.47,13.94,10.76.
Example 40: synthesis of N- (6- ((Z) -2-cyano-3- (2-fluoropyridin-4-yl) guanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S40)
Figure BDA0003265645630000401
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.38(s,1H),9.93(s,1H),9.18(s,1H),8.52(t,J=4.4Hz,1H),8.30(d,J=5.7Hz,1H),7.76-7.69(m,2H),7.48-7.41(m,2H),6.94(td,J=2.7,8.2Hz,1H),6.89(dd,J=1.9,8.0Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),3.47(td,J=4.3,6.3Hz,2H),3.28(td,J=4.4,6.3Hz,2H),2.37(s,3H),2.28(s,3H),1.53(m,4H),1.39-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.49,161.53,158.20,154.76,148.53,147.10,136.95,136.59,130.01,127.83,124.73,123.94,122.26,118.00,116.79,116.51,110.19,110.05,107.44,100.38,41.36,40.06,28.65,28.61,26.40,26.38,13.94,10.76.
Example 41: synthesis of N- (6- ((Z) -2-cyano-3- (2- (trifluoromethyl) pyridin-4-yl) guanidino) hexyl) -5- (((Z) -5-fluoro-2-oxoindolin-3-ylidene) methyl) -2, 4-dimethyl-1H-pyrrole-3-carboxamide (S41)
Figure BDA0003265645630000402
The synthesis method is as in example 4, and only the corresponding raw materials need to be replaced.1H NMR(500MHz,DMSO-d6)δ13.35(s,1H),9.93(s,1H),8.91(s,1H),8.56(t,J=4.4Hz,1H),8.43(d,J=5.7Hz,1H),8.04(d,J=2.0Hz,1H),7.80(t,J=4.4Hz,1H),7.73(dd,J=2.7,8.0Hz,1H),7.49(dd,J=1.9,5.6Hz,1H),7.43(s,1H),6.94(td,J=2.7,8.2Hz,1H),6.84(dd,J=4.9,8.4Hz,1H),3.46(td,J=4.3,6.3Hz,2H),3.25(td,J=4.3,6.3Hz,2H),2.38(s,3H),2.28(s,3H),1.59(m,2H),1.49(m,2H),1.39-1.26(m,4H).13C NMR(125MHz,DMSO-d6)δ170.24,165.29,160.22,154.75,148.79,146.71,146.14,136.95,136.63,130.01,127.79,124.19,123.94,122.26,120.48,118.00,116.95,116.67,112.93,110.05,108.45,107.44,41.36,40.06,28.61,28.58,26.42,26.41,13.92,10.78.
Example 42: assay for NAMPT and VEGFR2 enzyme inhibitory Activity
NAMPT inhibitory activity testing was performed according to the protocol provided by the CycLex NAMPT colorimetric kit (MBL International Corp.). The basic principle is as follows: NAM and PRPP produce NMN under the action of NAMPT, NMN and ATP produce NAD under the action of NMNAT1, NAD produces NADH under the action of Alcohol Dehydrogenase (ADH), and NADH returns to NAD under the action of diaphorase. WST-1 forms orange-yellow tympany in the enzymatic catalysis cycle of NAD/NADH, and the influence of the compound on the activity of NAMPT enzyme can be detected by detecting the change of absorbance at OD 450 nM.
The specific experimental operating steps are as follows:
preparing a solution of a compound to be detected: all compounds were dissolved in DMSO at a stock concentration of 10mM, formulated to the required concentration as required for the assay, starting from the starting concentration, diluted in 2-fold gradients, set up 8 concentration gradients, each concentration repeated three times.
② take ddH2O (10. mu.L) and NAM (5. mu.L) were added to a 96-well assay plate;
③ adding 5 mu L of compound to be detected or DMSO into a 96-hole detection plate;
fourthly, preparing the texture I, which comprises the following steps: 20 XNAMPT assay buffer (5. mu.L), PRPP (5. mu.L), ATP (5. mu.L), recombinant NMNAT1 (5. mu.L), ddH2O (35. mu.L) and NAMPT (5. mu.L), amounting to 60. mu.L. Addition of mix I to 96 well assayIn the plate;
fifthly, placing the 96-hole detection plate in an incubator at 30 ℃ for incubation for 60 min;
sixthly, configuring the texture I, which comprises the following steps: WST-1 (5. mu.L), ADH (5. mu.L), diaphorase (5. mu.L), ethanol (5. mu.L), amounting to 20. mu.L. After incubation, the 96-well assay plate was removed and mix II was added to the 96-well assay plate;
seventhly, dynamically monitoring the light absorption value of each hole at OD 450nm within 30min on a microplate reader, and detecting once every 5 min;
selecting time periods with linearly changing absorbance and time, and calculating the reaction rate. Processed with GraphPad Prism5 software. To compensate for the NAMPT inhibitory activity of DMSO, values were corrected with DMSO as a solvent control.
Method for testing VEGFR2 Activity
VEGFR2 is a receptor tyrosine kinase that consumes ATP, and the kinase activity is quantitatively determined by measuring the amount of ATP remaining in the solution after the kinase reaction by chemiluminescence. According to Kinase-LumiTMThe instructions of the chemiluminescence method kinase activity detection kit are operated. Briefly, the kinase buffer, the kinase, the reaction substrate and the test compound were added to a 96-well plate, respectively, and the 96-well plate was coated at room temperature and incubated for 40 min. Then, Kinase-Glo reagent was added to the 96-well plate and incubated for another 15 min. After the end, the microplate reader records luminescence. IC calculation Using GraphPad software50
The specific experimental results are shown in table 1:
table 1: NAMPT and VEGFR2 inhibitory Activity
Figure BDA0003265645630000421
Figure BDA0003265645630000431
Example 43: antitumor Activity test
The data of the cancer cell inhibitory activity of the compound are detected by an MTT method, which is also called an MTT colorimetric method and is a method for detecting the survival and growth of cells. MTT (yellow thiazole blue) can penetrate through cell membranes to enter cells, amber dehydrogenase in mitochondria of living cells can enable exogenous MTT to be reduced into water-insoluble needle-shaped Formazan crystals and deposited in the cells, the crystals can be dissolved by dimethyl sulfoxide (DMSO), an enzyme linked immunosorbent detector is used for detecting the light absorption value at the wavelength of 490nm/570nm, and the quantity of the living cells can be indirectly reflected. The cancer cell lines used were MCF-7 (human breast cancer cell), K562 (human chronic myelocytic leukemia cell), HT29 (human prostate cancer cell), A549 (human non-small cell lung cancer cell) and ACHN (renal cell carcinoma cell)
The specific experimental results are shown in table 1:
collecting cells in logarithmic growth phase, adjusting the concentration of cell suspension, and adding 100 mu L of cell suspension into each hole of a 96-hole plate; the number of cells per well was about 7000 cells at 5% CO2Incubating overnight at 37 ℃ until the cells are completely attached to the wall;
setting medicine concentration gradient, setting 3 multiple holes for each concentration gradient, diluting medicine into corresponding culture medium to required final concentration, sucking out original culture medium from 96-well plate, adding prepared culture medium containing medicine with required final concentration 100 μ L in 5% CO2Incubating at 37 ℃; simultaneously setting a blank group (only containing 100 mu L of culture medium, no cells and the same subsequent treatment as other wells) and a control group (containing cells and culture medium);
③ when the drug is treated for 44 hours, 10 mu of LMTT solution (5mg/ml) is added into each hole, and the culture is continued for 4 hours (48 hours for drug-treated cells);
fourthly, the culture solution in the hole is completely sucked (if the cells are suspended, the cell is firstly centrifuged at 2500rpm for 5min and then the culture medium is sucked out). Add 150. mu.L of DMSO to each well and shake until the crystals are fully dissolved. Detecting the light absorption value of each hole at OD490nm on a microplate reader;
calculating the inhibition rate: the inhibition ratio is 1- (the OD value of the drug addition agent-the OD value of the blank group)/(the OD value of the control group-the OD value of the blank group) — (the OD value of the control group-the OD value of the drug addition agent)/(the OD value of the control group-the OD value of the blank group);
sixthly, repeating the experimental steps for three times to obtain the average of the inhibition rate of the three timesValue, using IC50Calculator calculating IC of medicine50The value is obtained.
The specific experimental results are shown in table 2 below:
table 2: anti-cell proliferation Activity (IC) of the Compounds of the examples50μM)
Figure BDA0003265645630000441
Figure BDA0003265645630000451
Figure BDA0003265645630000461

Claims (10)

1. An indolone compound with a structure shown as a general formula I or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer or metabolite thereof,
Figure FDA0003265645620000011
in the formula:
l is-CH2OCH2CH2OCH2-, or unsubstituted or R1Substituted C2-10An alkyl group;
R1is hydroxy, amino, cyano, halogen or C1-6Alkyl groups of (a);
e is O, S or N-C ≡ N;
x is a single bond, C2-4Olefinic bond, C1-4Alkyl, cyclopropyl, -NHCH2-;
R is unsubstituted or R2Substituted C6-10Aryl, unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, 5-10 membered heteroaryl with 1-3 heteroatoms, or unsubstituted or R4SubstitutionThe heteroatom (S) is selected from N, O and S, and the heteroatom number is 1-3, namely, 5-10 membered heterocycloalkyl;
R2、R3and R4Independently is deuterium, halogen, hydroxy, amino, C1-6Alkyl radical, C1-6Alkoxy or C1-6A haloalkyl group.
2. The indolone compound of claim 1 or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, wherein:
when L is unsubstituted or R1Substituted C1-10When it is alkyl, said R1Is one or more, when there are more than one R1When R is said1May be the same or different;
and/or, when L is unsubstituted or R1Substituted C2-10When alkyl, said C2-10Alkyl is C2-8An alkyl group;
and/or when R1Is C1-6When there is an alkyl group, said C1-6Alkyl is C1-3An alkyl group;
and/or when R1When the halogen is fluorine, chlorine, bromine or iodine;
and/or, when E is O, X is a single bond, C1-4Alkyl radical, C2-4Alkenyl, cyclopropyl or-NHCH2-, or, when E is S, X is-NHCH2-or, alternatively, when E is N-C ≡ N, X is a single bond;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, R is3Is one or more, when there are more than one R3When R is said3May be the same or different;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, when the heteroatom is 5-10-membered heteroaryl with 1-3 heteroatoms, the heteroaryl is one or more of N, O and S, the heteroatom isA 5-to 10-membered heteroaryl group having 1 to 3N atoms, the number of N atoms being at least 1;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heterocycloalkyl with 1-3 heteroatoms, R is4Is one or more, when there are more than one R4When R is said4May be the same or different;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, when the heteroatom is 5-10 membered heterocycloalkyl with 1-3 heteroatoms, the heterocycloalkyl is one or more of N, O and S, the heteroatom is 1-3, and the number of N atoms is at least 1, namely 5-10 membered heterocycloalkyl;
and/or when R3Is halogen, and the halogen is fluorine, chlorine, bromine or iodine;
and/or when R3Is C1-6When alkyl, said C1-6Alkyl is C1-3An alkyl group;
and/or when R3Is C1-6At alkoxy, said C1-6Alkoxy is C1-3An alkoxy group;
and/or when R3Is C1-6When halogenated alkyl, said C1-6Haloalkyl being C1-3A haloalkyl group;
and/or when R4Is C1-6When alkyl, said C1-6Alkyl is C1-3An alkyl group;
and/or when R4Is C1-6When halogenated alkyl, said C1-6Haloalkyl being C1-3A haloalkyl group.
3. The indolone compound of claim 1 or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, wherein:
when L is unsubstituted or R1Substituted C1-10When it is alkyl, said R1The number of (a) is 1, 2 or 3;
and/or, when L is unsubstituted or R1Substituted C2-10When alkyl, said is unsubstituted or R1Substituted C2-10Alkyl is C4-7An alkyl group;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, R is3The number of (a) is 1, 2 or 3;
and/or, when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the number of the heteroatoms is 1-3, the heteroaryl is 5-10-membered heteroaryl, the heteroaryl is pyridyl, pyridopyrrolyl, pyridoimidazolyl, pyridofuryl, pyrazolothiophenyl or pyrazolopyrazolyl;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3, the R is 5-10 membered heterocycloalkyl4The number of (a) is 1, 2 or 3;
and/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3, the heterocyclic alkyl is 5-10 membered heterocyclic alkyl
Figure FDA0003265645620000021
And/or when R3Is halogen, and the halogen is fluorine;
and/or when R3Is C1-6When there is an alkyl group, said C1-6Alkyl of (a) is methyl, ethyl, propyl or isopropyl, preferably methyl;
and/or when R3Is C1-6Alkoxy of (2), said C1-6Alkoxy of (a) is methoxy, ethoxy or propoxy, preferably methoxy;
and/or when R3Is C1-6When halogenated alkyl, said C1-6Haloalkyl is-CF3
And/or when R4Is C1-6When alkyl, said C1-6Alkyl is methyl, ethyl or propyl;
and/or when R4Is C1-6When halogenated alkyl, said C1-6Haloalkyl is-CF3
4. The indolone compound of claim 1 or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, wherein:
when R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, the substituent is unsubstituted or R3Substituted heteroaryl is
Figure FDA0003265645620000031
And/or, when R is unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and when the heteroatom is 1-3 and is '5-10 membered heterocycloalkyl', the substituent is unsubstituted or R4Substituted heterocycloalkyl is
Figure FDA0003265645620000032
And/or, when E is O, X is a single bond, R is
Figure FDA0003265645620000033
Or, when E is O, X is-NHCH2-、C2-4Alkenyl or cyclopropyl, R is
Figure FDA0003265645620000041
Or, when E is S, X is-NHCH2-, R is
Figure FDA0003265645620000042
Or, when E is N-C [ identical to ] N, X is a single bond and R is
Figure FDA0003265645620000043
5. The indolone compound of claim 1 or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, wherein:
l is unsubstituted C2-10An alkyl group;
and/or E is O, S or N-C ≡ N;
and/or X is a single bond, C2-4Olefinic bond, C1-4Alkyl, cyclopropyl, -NHCH2-;
And/or R is unsubstituted or R3The substituted heteroatom is selected from one or more of N, O and S, 5-10 membered heteroaryl with 1-3 heteroatoms, or unsubstituted or R4The substituted heteroatom is selected from one or more of N, O and S, and the heteroatom number is 1-3, namely 5-10 membered heterocycloalkyl;
and/or, R3Is halogen, amino or C1-6An alkyl group;
and/or, R4Is amino or C1-6An alkyl group.
6. The indolone compound of claim 1, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, being any one of:
Figure FDA0003265645620000051
Figure FDA0003265645620000061
Figure FDA0003265645620000071
Figure FDA0003265645620000081
7. the process for producing an indolone compound according to any one of claims 1 to 6, wherein: the method comprises the following steps:
Figure FDA0003265645620000091
in a solvent, under the action of alkali and a condensing agent, carrying out condensation reaction on a compound shown as a formula II and a compound shown as a formula III, wherein L, E, X and R are defined as before.
8. A pharmaceutical composition comprising an effective amount of a compound of formula I as described in any one of claims 1-6, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer, or metabolite thereof, and a pharmaceutically acceptable carrier.
9. Use of an indolone compound of the structure shown in any one of claims 1-6 or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, tautomer or metabolite thereof in the preparation of a medicament for preventing and/or treating tumors.
10. The use according to claim 9, wherein the tumor is one or more of breast cancer, ovarian cancer, prostate cancer, colon cancer, gastric cancer, non-small cell lung cancer, glioma, renal cancer, pancreatic cancer, liver cancer, melanoma, leukemia and cervical cancer.
CN202111089653.8A 2021-09-16 2021-09-16 Indolone compounds and pharmaceutical composition, preparation method and application thereof Pending CN113717159A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111089653.8A CN113717159A (en) 2021-09-16 2021-09-16 Indolone compounds and pharmaceutical composition, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111089653.8A CN113717159A (en) 2021-09-16 2021-09-16 Indolone compounds and pharmaceutical composition, preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113717159A true CN113717159A (en) 2021-11-30

Family

ID=78684129

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111089653.8A Pending CN113717159A (en) 2021-09-16 2021-09-16 Indolone compounds and pharmaceutical composition, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113717159A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1439005A (en) * 2000-02-15 2003-08-27 苏根公司 Pyrrole substituted 2-indolinone protein kinase inhibitors
CN101389331A (en) * 2005-12-29 2009-03-18 斯克里普斯研究学院 Amino acid derivatives of indolinone based protein kinase inhibitors
CN102250069A (en) * 2010-05-17 2011-11-23 苏州波锐生物医药科技有限公司 Pyrrolic amide compound and purpose thereof in preparation of anti-malignant tumor medicament
US20190263823A1 (en) * 2016-11-22 2019-08-29 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with e3 ligase ligand and methods of use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1439005A (en) * 2000-02-15 2003-08-27 苏根公司 Pyrrole substituted 2-indolinone protein kinase inhibitors
CN101389331A (en) * 2005-12-29 2009-03-18 斯克里普斯研究学院 Amino acid derivatives of indolinone based protein kinase inhibitors
CN102250069A (en) * 2010-05-17 2011-11-23 苏州波锐生物医药科技有限公司 Pyrrolic amide compound and purpose thereof in preparation of anti-malignant tumor medicament
US20190263823A1 (en) * 2016-11-22 2019-08-29 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with e3 ligase ligand and methods of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BAOHUA ZOU等: "Design, syntheses and biological evaluation of 5-fluoroindolin-2-one derivatives with urea linkage", 《ADVANCED MATERIALS RESEARCH》 *

Similar Documents

Publication Publication Date Title
RU2728829C1 (en) New isoindoline derivative, pharmaceutical composition including thereof and use thereof
CN107709320B (en) Pyrido-nitrogen heterocyclic compound and preparation method and application thereof
CN105308033B (en) JAK2 and ALK2 inhibitor and its application method
CN109810041B (en) Halogenated allylamine SSAO/VAP-1 inhibitor and application thereof
JP2022518860A (en) Immunomodulators, compositions and their uses
EP2751108B1 (en) Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
CN102099036A (en) Compounds and methods for treating inflammatory and fibrotic disorders
KR20150087271A (en) Pyrimidine-2,4-diamine derivatives for treatment of cancer
CN103130792B (en) A kind of thiazolamine compounds
CN108530444B (en) Novel NAMPT and IDO dual inhibitor and preparation method and medical application thereof
CA2756099A1 (en) Nicotinamide derivatives, preparation thereof, and therapeutic use thereof as anticancer drugs
WO2021115401A1 (en) Fluorine-containing heterocyclic derivatives with macrocyclic structure and use thereof
US20220233513A1 (en) Compound used as ret kinase inhibitor and application thereof
EP3845527A1 (en) Multi-substituted pyridone derivatives and medical use thereof
CN114656482A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN106660970A (en) Quinazoline derivative
CN110938109B (en) O-quinone compound, preparation method and medical application thereof
CN111718332B (en) 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof
CN112898286A (en) Benzothiophene compound or pharmaceutically acceptable salt and isomer thereof, and preparation method, pharmaceutical composition and application thereof
CN114380823B (en) Imidazole-2-methylamine derivative and medical application thereof
CN111848506B (en) Biphenyl urea compound and pharmaceutical composition, preparation method and application thereof
CN113717159A (en) Indolone compounds and pharmaceutical composition, preparation method and application thereof
CN100572381C (en) The pyrazolo naphthyridines derivatives
JP2021533085A (en) 1,2,4-Triazole derivative as a tankylase inhibitor
CN111170995A (en) ASK1 inhibitor 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
RJ01 Rejection of invention patent application after publication

Application publication date: 20211130

RJ01 Rejection of invention patent application after publication