CN113788827B - Phthalazinone compound, preparation method and medical application thereof - Google Patents

Phthalazinone compound, preparation method and medical application thereof Download PDF

Info

Publication number
CN113788827B
CN113788827B CN202111212595.3A CN202111212595A CN113788827B CN 113788827 B CN113788827 B CN 113788827B CN 202111212595 A CN202111212595 A CN 202111212595A CN 113788827 B CN113788827 B CN 113788827B
Authority
CN
China
Prior art keywords
nmr
compound
cancer
preparation
chloroform
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.)
Active
Application number
CN202111212595.3A
Other languages
Chinese (zh)
Other versions
CN113788827A (en
Inventor
蒋晟
张阔军
廖金标
王德祥
倪勇
王天雨
章翔宇
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 CN202111212595.3A priority Critical patent/CN113788827B/en
Publication of CN113788827A publication Critical patent/CN113788827A/en
Application granted granted Critical
Publication of CN113788827B publication Critical patent/CN113788827B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • C07D401/02Heterocyclic 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 two hetero rings
    • C07D401/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a phenolphthalein ketone compound with a structure shown in a general formula I, a pharmaceutical composition, a preparation method and application thereof. The phenolphthalein ketone compound has remarkable inhibitory activity on NAMPT and PARP1 and has good therapeutic effect on tumors.

Description

Phthalazinone compound, preparation method and medical application thereof
Technical Field
The invention belongs to the field of innovative pharmaceutical chemistry, and in particular relates to a phenolphthalein ketone compound shown in a formula I or pharmaceutically acceptable salts, solvates, prodrugs, isomers or metabolites thereof, a preparation method, a pharmaceutical composition containing the compound, and application of the compound or the pharmaceutical composition in preparing antitumor drugs.
Background
Cancer is a serious disease which afflicts human health, and research and development of novel antitumor drugs are important subjects and long-term tasks in the field of biological medicine. Poly ADP-ribose polymerase, PARP, is a multifunctional protein post-translational modification enzyme that is widely found in eukaryotic cells. In mammalian cells, the PARP family has 17 members, whereas the members involved in DNA damage repair processes are only three of PARP1, PARP2, and PARP3, wherein PARP1 is a major member involved in DNA repair processes, when DNA damage is single-stranded broken, cells initiate DNA damage response procedures, PARP1 can sense DNA single-stranded damage gaps, and Nicotinamide Adenine Dinucleotide (NAD) is catalyzed by self-glycosylation of its zinc finger DNA binding domain located at the site of DNA damage + ) Decomposing into nicotinamide and ADP ribose, and using ADP ribose as substrate to make receptor protein and PARP1 form poly ADP-ribose polymer to form PARP-ADP ribose branched chain, avoiding recombination of DNA molecule around damaged part and damaged DNA, and simultaneously attracting DNA repair protein, histone H1, a series of transcription factors and other DNA repair enzymes such as X-ray repair cross complementary gene 1, DNA ligase 3, DNA guide polymerase beta and the like to be combined at the damaged part to repair the damaged part. When PARP function is impaired or inhibited, single strand breaks persist, which can easily lead to stalling of replication forks and DNA double strand breaks, and thus impaired DNA replicas appear and accumulate gradually, eventually leading to replication fork breakdown. In cells with normal functional homologous recombination repair pathways, the DNA double strand breaks are immediately repaired, which can compensate for the loss of function in the base excision repair pathway; however, in tumor cells lacking homologous recombination repair, such as breast cancer carrying BRCA1/2 mutations, there is a lack of compensatory mechanisms, and the tumor cells fail to repair DNA damage, ultimately leading to cell death.
Nicotinamide adenine dinucleotide (nicotinamide adenine dinucleotide, NAD for short) + ) Is an indispensable substance in the electron transfer process and plays an important role in the energy metabolism and signal transduction processes. In addition, NAD + NADH in the form of its reduced form is important for maintaining a reducing environment within the cell and thereby protecting the cell from oxidative stress. Thus, intracellular NAD + Is a critical factor in determining cell fate. Tumor cells have uncontrolled proliferation rates, rapid metabolic rates, and significantly increased oxidative stress levels relative to normal cells, and thus tumor cells are resistant to NAD + The change in level is sensitive. NAD (NAD) + Key enzymes of the in vivo biosynthetic pathway have become important targets for anticancer drugs. NAD (NAD) + In vivo synthetic pathwayIncluding de novo and salvage synthetic pathways.
NAD + The salvage synthesis route is to use nicotinic acid, nicotinamide or nicotinamide ribose as a starting material. Due to a plurality of NADs + Consumable enzymes such as poly (adenosine diphosphate) ribose polymerase (PARP) and deacetylase (Sirtuis, SIRT) can rapidly re-release nicotinamide, thereby NAD starting from nicotinamide + The salvage pathway is the most economical and dominant NAD + Biosynthetic pathways (gali U.S. et al J.Med. Chem.2013, 22, 56 (16): 6279-96). Whereas nicotinamide riboside transferase (nicotinamide phosphoribosyltransferase, NAMPT) controls nicotinamide to NAD + And (3) a speed-limiting enzyme for transformation. In view of the abnormal energy metabolism of tumor cells, NAD is more required + Rapid and timely supplementation, tumor cells are more dependent on NAMPT and are more sensitive to NAMPT inhibition. Therefore, NAMPT is considered as an effective anticancer target, and NAMPT inhibitors have become a hotspot for extensive research. Currently, two NAMPT inhibitors CHS-828 and FK866 have entered the clinical trial phase (Montecocco Fet al. Curr. Drug targets.2013,1, 14 (6): 637-43). However, the clinical trial results of these two compounds were not ideal and did not show significant therapeutic effect after administration to patients. Furthermore, these two compounds have poor pharmacokinetic properties and also show dose-dependent toxicity such as thrombocytopenia and gastrointestinal toxic side effects. Therefore, NAMPT inhibitors with novel structure and good pharmaceutical property are required to be further researched and developed.
Figure BDA0003308477660000021
Disclosure of Invention
The invention aims to: the invention aims to provide a phenolphthalein ketone compound and a pharmaceutical composition thereof, which have the advantages of good curative effect of anticancer drugs, overcoming drug resistance and the like.
The invention obtains PARP/NAMPT double-target inhibitor, and can reduce NAD through inhibiting nicotinamide phosphoribosyl transferase + To increase the level of chemical combinationThe competitive inhibition of poly (adenosine diphosphate) ribose polymerase by the compound is expected to solve the current drug resistance problem of PARP inhibitors.
The invention also aims to provide a preparation method and application of the phenolphthalein ketone compound with the advantages of good curative effect of the anticancer drug, overcoming drug resistance and the like, and a pharmaceutical composition thereof.
The invention provides phthalazinone compounds with a structure shown in a general formula I, or pharmaceutically acceptable salts, solvates, prodrugs, isomers or metabolites thereof,
Figure BDA0003308477660000022
in formula I:
l is-CH 2 OCH 2 CH 2 OCH 2 -, or, unsubstituted or R 1 Substituted C 2-10 An alkyl group;
R 1 independently hydroxy, amino, halogen, C 1-6 Alkyl, C of (2) 1-6 Alkoxy, C 1-6 A haloalkyl group;
e is O, S or N-C.ident.N;
x is a single bond, C 2-4 Olefinic bond, C 1-4 Alkyl, cyclopropyl, -NHCH 2 -;
R is unsubstituted or R 2 Substituted C 6-10 Aryl, unsubstituted or R 3 The substituted hetero atom is selected from one or more of N, O and S, and the hetero atom number is 1-3, 5-10 membered hetero aryl, unsubstituted or R 4 The substituted hetero atom is selected from one or more of N, O and S, and the hetero atom number is 5-10 membered heterocyclic alkyl of 1-3;
R 2 、R 3 and R is 4 Independently deuterium, halogen, hydroxy, amino, C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 A haloalkyl group.
Further, when L is unsubstituted or R 1 Substituted C 2-10 In the case of alkyl, said R 1 Is one or more, when there are a plurality of R 1 In the time-course of which the first and second contact surfaces,said R is 1 May be the same or different;
and/or when L is unsubstituted or R 1 Substituted C 2-10 In the case of alkyl, said C 2-10 Alkyl is C 2-8 An alkyl group;
and/or when R 1 Is C 1-6 When alkyl is said C 1-6 Alkyl is C 1-3 An alkyl group;
and/or when R 1 When halogen, the halogen is fluorine, chlorine, bromine or iodine;
and/or, when E is O, X is a single bond, C 1-4 Alkyl, C 2-4 Alkenyl, cyclopropyl or-NHCH 2 -, or, when E is S, X is-NHCH 2 -or, when E is N-c≡n, X is a single bond;
and/or when R is unsubstituted or R 3 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 1-3, the hetero atom is 5-10 membered heteroaryl, the R 3 Is one or more, when there are a plurality of R 3 When said R is 3 May be the same or different;
and/or when R is unsubstituted or R 3 When the heteroatom is selected from one or more of N, O and S and the heteroatom number is 1-3, the heteroaryl is one or more of N, O and S, the heteroatom number is 1-3 and the heteroatom number is at least 1, and the heteroatom number is 5-10;
and/or when R is unsubstituted or R 4 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 1-3, the hetero atom is 5-10 membered heterocyclic alkyl, the R 4 Is one or more, when there are a plurality of R 4 When said R is 4 May be the same or different;
and/or when R is unsubstituted or R 4 When the heteroatom is selected from one or more of N, O and S and the 5-to 10-membered heterocycloalkyl group with 1-3 heteroatoms, the heterocycloalkyl group is one or more of N, O and S, the heteroatom is 1-3, and the 5-to 10-membered heterocycloalkyl group with at least 1N atom;
and/or when R 3 Halogen, wherein the halogen is fluorine, chlorine, bromine or iodine;
and/or when R 3 Is C 1-6 In the case of alkyl, said C 1-6 Alkyl is C 1-3 An alkyl group;
and/or when R 3 Is C 1-6 In the case of alkoxy, said C 1-6 Alkoxy is C 1-3 An alkoxy group;
and/or when R 3 Is C 1-6 When haloalkyl, said C 1-6 Haloalkyl is C 1-3 A haloalkyl group;
and/or when R 4 Is C 1-6 In the case of alkyl, said C 1-6 Alkyl is C 1-3 An alkyl group;
and/or when R 4 Is C 1-6 When haloalkyl, said C 1-6 Haloalkyl is C 1-3 A haloalkyl group.
Further, when L is unsubstituted or R 1 Substituted C 2-10 In the case of alkyl, said R 1 Is 1,2 or 3;
and/or when L is unsubstituted or R 1 Substituted C 2-10 In the case of alkyl, said unsubstituted or R 1 Substituted C 2-10 Alkyl is C 2-6 An alkyl group;
and/or when R is unsubstituted or R 3 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 1-3, the hetero atom is 5-10 membered heteroaryl, the R 3 Is 1,2 or 3;
and/or when R is unsubstituted or R 3 When the heteroatom is selected from one or more of N, O and S and the heteroatom is 5-10 membered heteroaryl with 1-3 heteroatoms, the heteroaryl is pyridyl, pyridopyrrolyl, pyridoimidazolyl, pyridofuryl, pyrazolothienyl or pyrazolopyrazolyl;
and/or when R is unsubstituted or R 4 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 5-10 membered heterocycloalkyl, the R is 1-3 4 The number of (2) is 1,2 or 3;
And/or when R is unsubstituted or R 4 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 5-10 membered heterocycloalkyl of 1-3, the heterocycloalkyl is
Figure BDA0003308477660000041
And/or when R 3 Halogen, wherein the halogen is fluorine;
and/or when R 3 Is C 1-6 When alkyl is said C 1-6 Alkyl of (2) is methyl, ethyl, propyl or isopropyl, preferably methyl;
and/or when R 3 Is C 1-6 When alkoxy is said C 1-6 Alkoxy of (a) is methoxy, ethoxy or propoxy, preferably methoxy;
and/or when R 3 Is C 1-6 When haloalkyl, said C 1-6 Haloalkyl is-CF 3
And/or when R 4 Is C 1-6 In the case of alkyl, said C 1-6 Alkyl is methyl, ethyl or propyl, preferably methyl;
and/or when R 4 Is C 1-6 When haloalkyl, said C 1-6 Haloalkyl is-CF 3
Further, when R is unsubstituted or R 3 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 5-10 membered heteroaryl of 1-3, the unsubstituted or R 3 Substituted heteroaryl groups are:
Figure BDA0003308477660000042
and/or when R is unsubstituted or R 4 The substituted hetero atom is one or more of N, O and S, and when the hetero atom number is 1-3' 5-10 membered heterocyclic alkyl group, the unsubstituted or R 4 Substituted heterocycloalkyl is
Figure BDA0003308477660000051
And/or when E is O, X is a single bond, R is
Figure BDA0003308477660000052
Alternatively, when E is O, X is-NHCH 2 -、C 2-4 Alkenyl or cyclopropyl, R is
Figure BDA0003308477660000053
Alternatively, when E is S, X is-NHCH 2 -, R is
Figure BDA0003308477660000054
Alternatively, when E is N-C≡N, X is a single bond and R is
Figure BDA0003308477660000055
Further, L is unsubstituted C 2-10 An alkyl group;
and/or E is O, S or N-C≡N;
and/or X is a single bond, C 2-4 Olefinic bond, C 1.4 Alkyl, cyclopropyl, -NHCH 2 -;
And/or R is unsubstituted or R 3 The substituted hetero atom is selected from one or more of N, O and S, the hetero atom number is 1-3 '5-10 membered heteroaryl, or the unsubstituted or R4 substituted hetero atom is selected from one or more of N, O and S, and the hetero atom number is 1-3' 5-10 membered heterocycloalkyl;
and/or R 3 Is halogen, amino or C 1-6 An alkyl group;
and/or R 4 Is amino or C 1-6 An alkyl group.
Further, the method comprises the steps of,
the phenolphthalein ketone compound or pharmaceutically acceptable salts, solvates, prodrugs, isomers or metabolites thereof is any one of the following:
Figure BDA0003308477660000061
/>
Figure BDA0003308477660000071
/>
Figure BDA0003308477660000081
/>
Figure BDA0003308477660000091
/>
Figure BDA0003308477660000101
Figure BDA0003308477660000111
/>
further, the preparation method of the phenolphthalein ketone compound is characterized by comprising the following steps of: the method comprises the following steps:
Figure BDA0003308477660000112
and (3) in a solvent, under the action of alkali and a condensing agent, carrying out condensation reaction on the compound shown in the formula II and the compound shown in the formula III, wherein L, E, X and R are defined as above.
A pharmaceutical composition comprising an effective amount of a compound of formula I according to any one of claims 1 to 6, or a pharmaceutically acceptable salt, solvate, prodrug, isomer or metabolite thereof, and a pharmaceutically acceptable carrier. The invention also provides application of the pharmaceutical composition in preparing medicines for preventing and/or treating tumors, including but not limited to breast cancer, ovarian cancer, prostatic cancer, colon cancer, gastric cancer, non-small cell lung cancer, glioma, kidney cancer, pancreatic cancer, liver cancer, melanoma, leukemia and cervical cancer, preferably breast cancer, ovarian cancer, prostate cancer, colon cancer, gastric cancer, non-small cell lung cancer, melanoma, leukemia and cervical cancer.
The application of phenolphthalone compounds with the structure shown in the general formula I or pharmaceutically acceptable salts, solvates, prodrugs, isomers or metabolites thereof in preparing medicines for preventing and/or treating tumors or NAMPT and PARP1 inhibitors.
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, prostate, colon, non-small cell lung, leukemia and kidney cancers.
The pharmaceutically acceptable carrier can be those excipients widely used in the field of pharmaceutical production. Adjuvants are used primarily to provide a safe, stable and functional pharmaceutical composition, and may also provide means for allowing the subject to dissolve at a desired rate after administration, or for promoting effective absorption of the active ingredient after administration of the composition. The pharmaceutical excipients may be inert fillers or provide a function such as stabilizing the overall pH of the composition or preventing degradation of the active ingredients of the composition. The pharmaceutical excipients can comprise one or more of the following excipients: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, sizing agents, disintegrants, lubricants, anti-adherents, glidants, wetting agents, gelling agents, absorption retarders, dissolution inhibitors, enhancing agents, adsorbents, buffering agents, chelating agents, preservatives, colorants, flavoring agents, and sweeteners.
The pharmaceutical compositions of the present invention may be prepared in accordance with the disclosure using any method known to those of skill in the art. For example, conventional mixing, dissolving, granulating, emulsifying, levigating, encapsulating, entrapping or lyophilizing processes.
The pharmaceutical 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, implantation, subcutaneous, intravenous, intra-arterial, intramuscular). The pharmaceutical compositions of the invention may also be in controlled or delayed release dosage forms (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 formulations which may 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; a liquid dosage form suitable for parenteral administration; suppositories and lozenges.
The term "pharmaceutically acceptable salt" refers to salts of the compounds of the present invention prepared from the compounds of the present invention which have the specified substituents found herein with relatively non-toxic acids or bases. When the compounds of the present invention contain relatively acidic functional groups, base addition salts may be obtained by contacting neutral forms of such compounds with a sufficient amount of a base in pure 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 the compounds of the present invention contain relatively basic functional groups, the acid addition salts may be obtained by contacting the neutral form of such compounds with a sufficient amount of an acid in pure 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 organic acid salts including acids such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and methanesulfonic acid; also included are salts of amino acids (e.g., arginine, etc.), and salts of organic acids such as glucuronic acid. Certain specific compounds of the invention contain basic and acidic functionalities that can be converted to either base or acid addition salts. Preferably, the salt is contacted with a base or acid in a conventional manner to isolate the parent compound, thereby regenerating the neutral form of the compound. The parent form of a compound differs from its various salt forms in certain physical properties, such as solubility in polar solvents.
The term "isomer" refers to two or more compounds that have the same molecular composition but different structures and properties.
The term "solvate" refers to a mixture of a compound and a solvent, e.g., crystalline is a solvate.
The term "metabolite" refers to a pharmaceutically active product of a compound of formula I or a salt thereof produced by in vivo metabolism. Such products may result from, for example, oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, glucuronidation (glucorination), enzymatic cleavage, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds produced by a method of contacting a compound of the present invention with a mammal for a period of time sufficient to obtain the metabolites thereof.
The identification of metabolites is typically accomplished by preparing radiolabeled compounds of the invention (e.g., 14 c or 3 H) Isotope, parenteral administration thereof to an animal, such as a rat, mouse, guinea pig, monkey, or human, at a detectable dose (e.g., greater than about 0.5 mg/kg), allows forAllowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and for its conversion products to be isolated from urine, blood or other biological samples. These products are easy to isolate because they are labeled (others are isolated by using antibodies that are capable of binding to epitopes present in the metabolite). The metabolite structures are determined in a conventional manner, for example by MS, LC/MS or NMR analysis. In general, the analysis of metabolites is performed in the same manner as conventional drug metabolism studies known to those skilled in the art. So long as the metabolite products are not otherwise undetectable in vivo, they are useful in assays for therapeutic dosing of the compounds of the invention. The compounds of the present invention may contain non-natural proportions of atomic isotopes on one or more of the atoms comprising the compounds. For example, compounds can be labeled with radioisotopes, such as tritium @, for example 3 H) Iodine-125% 125 I) Or C-14% 14 C) A. The invention relates to a method for producing a fibre-reinforced plastic composite 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 exist in prodrug forms. Prodrugs of the compounds described herein readily undergo chemical changes under physiological conditions to convert to the compounds of the 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 invention. For example, compounds containing a carboxyl group can form a physiologically hydrolyzable ester that acts as a prodrug by hydrolyzing in vivo to give the compound of formula I itself. The prodrugs are preferably administered orally, as hydrolysis occurs in many cases primarily under the influence of digestive enzymes. Parenteral administration may be used when the ester itself is active or hydrolysis occurs in the blood.
"substitution" in the present invention may be one or more, and when there are plural "substitution", the "substitution" may be the same or different.
The term "plurality" refers to 2,3, 4, or 5. For example, the term "alkyl" refers to a straight or branched saturated hydrocarbon group having the indicated number of carbon atoms. Representative saturated hydrocarbon groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, n-pentyl, t-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 means only the number of carbon atoms of the alkyl moiety indicated 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-R Y Wherein R is Y Are alkyl groups 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 may be a bicyclic or tricyclic cycloalkyl having 4 to 10 carbon atoms.
In the present invention, "heterocycloalkyl" means a saturated monocyclic ring having a heteroatom or a bicyclic ring formed by fusing a saturated monocyclic ring having a heteroatom with a heteroaryl, and when "heterocycloalkyl" is a bicyclic ring formed by fusing a saturated monocyclic ring having a heteroatom with a heteroaryl, the "heterocycloalkyl" is linked to other fragments or groups in the compound represented by formula I through a saturated monocyclic ring having 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, and when bicyclic or tricyclic, each ring satisfies the shock rule. C of the invention 6-10 Aryl of (a) refers to an aromatic group containing 6 to 10 carbon atoms, such as phenyl or naphthyl.
The term "heteroaryl" refers to an aromatic group containing heteroatoms, preferably an aromatic 5-6 membered monocyclic ring or a 9-10 membered bicyclic ring containing 1,2 or 3 atoms independently selected from nitrogen, oxygen and sulfur. The 5-to 6-membered monocyclic ring includes, but is not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, furazanyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, 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 9-10 membered bicyclic ring includes, but is not limited to, benzimidazolyl, benzisothiazolyl, benzoxazolyl, imidazopyridine, thiazolopyridine, furopyridine, tetrahydropyrrolopyridine.
The beneficial effects are that: compared with the prior art, the invention has the following advantages:
1. the compounds of the invention have good inhibitory activity on VEGFR, especially VEGFR 2.
2. The compound has good inhibitory activity on NAMPT.
3. The compound has dual inhibition activities of VEGFR and NAMPT, has good treatment effect on tumors, has fewer toxic and side effects and can solve the problem of drug resistance compared with a single-target inhibitor.
Detailed Description
Example 1: synthetic route of the compound of formula I (Synthesis of A series of compounds)
Figure BDA0003308477660000141
Preparation of 4- (4-aminoethyl) -1-t-butoxycarbonylpiperazine
Figure BDA0003308477660000142
Into a 25ml round bottom flask was charged compound 1 (2.0 g,10.8 mmol), compound 2 (3.0 g,10.8 mmol), triethylamine (3.0 ml,21.6 mmol), o-xylene (70 ml) and LThe reaction was carried out at 120℃for 24 hours. After the reaction, the mixture was filtered, the filtrate was concentrated, and column chromatography (EA/pe=1:4 to 2:1) was performed to obtain compound 3. To a 250ml round bottom flask was added compound 3 (3.7 g,9.6 mmol), hydrazine hydrate (1.0 ml,19.2 mmol), ethanol (80 ml), and the mixture was refluxed at 80℃for 10 hours. After completion of the reaction, the mixture was filtered, the filtrate was concentrated, and the mixture was chromatographed (DCM/MeOH/Et 3 N=90:8:2) to give 4- (4-aminoethyl) -1-t-butoxycarbonylpiperazine (1.9 g, 80%).
Preparation of 4- (4- (imidazo [1,2-a ] pyridine-7-carboxamide) n-butyl) piperazine-1-t-butoxycarbonyl piperazine
Figure BDA0003308477660000151
In a 25ml round bottom flask was added compound 4 (1.5 g,5.3 mmol), compound 5 (850 mg,5.3 mmol), HBTU (3.0 g,8.0 mmol), DIPEA (2.7 ml,16.0 mmol), DMF (25 ml) and reacted at room temperature for 14 h. After the reaction was completed, DMF was dried by spin-drying, and column chromatography (MeOH/DCM=1:30.about.1:10) gave 4- (4- (imidazo [1,2-a ] pyridine-7-carboxamide) n-butyl) piperazine-1-t-butoxycarbonyl piperazine (1.0 g, 47%).
Preparation of N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) imidazo [1,2-a ] pyridine-7-carboxamide
Figure BDA0003308477660000152
A25 ml round bottom flask was charged with Compound 6 (1.0 g,2.5 mmol), ethyl acetate (10 ml), and a saturated solution of ethyl acetate in hydrogen chloride (4 ml) was prepared and reacted at room temperature for 4 hours. After the reaction is finished, filtering to obtain solid, and drying for later use. A25 ml round bottom flask was charged with the solid from the previous step, compound 7 (1.1 g,3.8 mmol), HATU (1.4 g,3.8 mmol), DIPEA (2.1 ml,12.5 mmol), DMF (20 ml) and reacted at room temperature for 10 hours. After the reaction, spin-drying DMF, column chromatography (MeOH/DCM=1:20-1:8) gives N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazinyl-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) imidazo [1,2-a]pyridine-7-carboxamide (500 mg, 43%). 1 H NMR(500MHz,Chloroform-d)δ8.83(d,J=9.0Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),8.08(t,J=4.4Hz,1H),8.02(d,J=1.6Hz,1H),7.80(ddt,J=5.0,2.0,1.1Hz,1H),7.78-7.72(m,1H),7.72-7.65(m,3H),7.65(s,1H),7.44(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.36(dd,J=9.0,1.5Hz,1H),7.21(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.74(dt,J=11.7,5.3Hz,2H),3.45-3.34(m,4H),2.58(t,J=5.4Hz,4H),2.46(t,J=6.3Hz,2H),1.69-1.60(m,2H),1.56(pd,J=6.3,1.0Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ166.97,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,142.28,132.79-132.48(m),132.42-132.03(m),130.76,129.93-129.68(m),129.24,127.35,125.74,125.58,123.91,123.33,115.59,115.43,112.00,111.06,109.28,56.03,53.41,45.21,39.75,36.72,26.61,24.99.
Example 2: preparation of N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) ethyl) imidazo [1,2-a ] pyridine-7-carboxamide
Figure BDA0003308477660000153
The synthesis was as in example 1, with only the corresponding starting materials being replaced. 1 H NMR(500MHz,Chloroform-d)δ8.79(d,J=8.9Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),8.00(d,J=1.5Hz,1H),7.86(t,J=4.4Hz,1H),7.78-7.62(m,6H),7.49(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.39(dd,J=9.0,1.5Hz,1H),7.24(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.76-3.68(m,2H),3.52(td,J=5.6,4.4Hz,2H),3.44-3.35(m,2H),2.68(t,J=5.6Hz,2H),2.62-2.51(m,4H). 13 C NMR(125MHz,DMSO-d6)δ166.51,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.85,142.29,132.79-132.03(m),130.75,130.00-129.70(m),129.24,127.35,125.74,125.58,123.91,123.33,115.59,115.43,112.22,111.06,109.27,55.86,52.46,45.21,37.56,36.72.
Example 3: preparation of N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) propyl) imidazo [1,2-a ] pyridine-7-carboxamide
Figure BDA0003308477660000161
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.68(d,J=8.9Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),8.17(t,J=4.4Hz,1H),8.06(d,J=1.6Hz,1H),7.78-7.64(m,5H),7.61(ddt,J=4.9,1.9,1.0Hz,1H),7.50(dddt,J=8.1,5.1,2.0,1.1Hz,1H),7.35(dd,J=9.0,1.5Hz,1H),7.25(t,J=8.2Hz,1H),4.30(t,J=1.0Hz,2H),3.73(dt,J=11.7,5.3Hz,2H),3.39-3.28(m,4H),2.64-2.55(m,4H),2.46(t,J=6.4Hz,2H),1.78(p,J=6.4Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ166.63,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.85,142.28,132.79-132.03(m),130.75,129.81(d,J=8.2Hz),129.24,127.35,125.74,125.58,123.91,123.33,115.59,115.43,112.00,111.06,109.27,53.26(d,J=6.9Hz),45.21,38.13,36.72,25.71.
Example 4: preparation of N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) imidazo [1,2-a ] pyridine-7-carboxamide
Figure BDA0003308477660000162
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.79(d,J=8.9Hz,1H),8.26-8.18(m,2H),8.01(d,J=1.5Hz,1H),7.78-7.72(m,1H),7.72-7.67(m,3H),7.67-7.63(m,2H),7.52(dddt,J=7.9,5.1,2.0,1.1Hz,1H),7.33(dd,J=9.0,1.5Hz,1H),7.24(t,J=8.2Hz,1H),4.25(t,J=1.0Hz,2H),3.81-3.72(m,2H),3.27(dtd,J=11.9,6.0,4.6Hz,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.63(dp,J=22.2,6.5Hz,4H),1.44-1.34(m,2H). 13 C NMR(125MHz,Chloroforrm-d)δ166.75,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,142.28,132.82-132.48(m),132.42-132.03(m),130.76,129.93-129.59(m),129.33,127.35,125.74,125.58,123.91,123.33,115.59,115.43,112.00,111.06,109.28,56.16,53.41,45.21,39.86,36.72,28.56,26.50,25.02.
Example 5: preparation of N- (4- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) imidazo [1,2-a ] pyridine-7-carboxamide
Figure BDA0003308477660000171
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.79(d,J=8.9Hz,2H),8.29-8.23(m,2H),8.15(t,J=4.4Hz,2H),8.02(d,J=1.5Hz,2H),7.76(d,J=1.8Hz,1H),7.75-7.67(m,7H),7.65(s,2H),7.56(ddt,J=5.0,2.0,1.0Hz,2H),7.48(dddt,J=8.6,4.9,1.9,0.9Hz,2H),7.33(dd,J=9.0,1.5Hz,2H),7.23(t,J=8.2Hz,2H),4.27(t,J=1.0Hz,4H),3.74(ddd,J=11.7,6.0,4.6Hz,4H),3.32(ddd,J=11.7,6.0,4.5Hz,4H),3.24(td,J=6.3,4.3Hz,4H),2.63-2.51(m,8H),2.37(t,J=6.4Hz,4H),1.64(p,J=6.6Hz,4H),1.45(p,J=6.6Hz,4H),1.37-1.23(m,8H). 13 C NMR(125MHz,Chloroform-d)δ166.75,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,142.28,132.82-132.48(m),132.42-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.91,123.33,115.59,115.43,112.00,111.06,109.28,56.16,53.41,45.21,39.77,36.72,28.48,27.33,27.11,26.71.
Example 6: (E) Preparation of (E) -N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) ethyl) -3- (pyridin-3-yl) acrylamide
Figure BDA0003308477660000172
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.65(t,J=1.9Hz,1H),8.58(dt,J=3.7,1.8Hz,1H),8.29-8.23(m,1H),7.97(dt,J=7.8,1.9Hz,1H),7.83(t,J=4.4Hz,1H),7.82-7.75(m,2H),7.75-7.64(m,2H),7.62-7.50(m,3H),7.25(t,J=8.2Hz,1H),6.59(d,J=15.9Hz,1H),4.24(t,J=1.0Hz,2H),3.83-3.75(m,2H),3.43-3.38(m,2H),3.38-3.30(m,2H),2.68(t,J=5.6Hz,2H),2.61-2.52(m,4H). 13 C NMR(125MHz,Chloroform-d)δ167.27,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.85,150.23,149.57,138.69,134.34,132.71,132.37-132.03(m),130.75,129.83(dd,J=18.6,6.7Hz),129.16,127.27,125.74,125.58,123.91,123.33,120.46,115.59,115.43,55.78,52.46,45.21,38.41.36.72.
Example 7: (E) Preparation of (E) -N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) propyl) -3- (pyridin-3-yl) acrylamide
Figure BDA0003308477660000181
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.90(t,J=1.8Hz,1H),8.58(dt,J=3.6,1.8Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),7.98(dt,J=7.9,1.8Hz,1H),7.78-7.72(m,2H),7.68(dtd,J=14.8,7.5,1.7Hz,2H),7.64-7.56(m,3H),7.49(dddt,J=8.7,4.9,1.9,0.9Hz,1H),7.24(t,J=8.2Hz,1H),6.59(d,J=15.9Hz,1H),4.28(t,J=1.0Hz,2H),3.76(dt,J=11.7,5.3Hz,2H),3.38-3.29(m,2H),3.17(td,J=6.3,4.3Hz,2H),2.64-2.55(m,4H),2.45(t,J=6.4Hz,2H),1.72(p,J=6.4Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ167.67,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.85,150.23,149.56,138.69,134.33,132.71,132.37-132.03(m),130.75,130.00-129.66(m),129.16,127.35,125.74,125.58,123.91,123.33,120.74,115.59,115.43,53.33(d,J=11.0Hz),45.16,39.19,36.72,25.95.
Example 8: (E) Preparation of (E) -N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) -3- (pyridin-3-yl) acrylamide
Figure BDA0003308477660000182
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.88(t,J=1.9Hz,1H),8.64(dt,J=3.6,1.8Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),7.96(dt,J=7.8,1.8Hz,1H),7.78-7.45(m,8H),7.24(t,J=8.2Hz,1H),6.59(d,J=15.9Hz,1H),4.26(t,J=1.0Hz,2H),3.60(dt,J=11.7,5.3Hz,2H),3.26(dt,J=11.7,5.3Hz,2H),3.20(td,J=6.3,4.4Hz,2H),2.59(t,J=5.3Hz,4H),2.46(t,J=6.3Hz,2H),1.68-1.59(m,2H),1.56-1.46(m,2H). 13 C NMR(125MHz,Chloroform-d)δ167.37,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.85,150.23,149.56,138.69,134.33,132.71,132.42-132.03(m),130.75,129.94-129.66(m),129.24,127.35,125.74,125.58,123.91,123.33,120.74,115.59,115.43,56.03,53.37,45.21,39.60,36.72,26.67,24.95.
Example 9: (E) Preparation of (E) -N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -3- (pyridin-3-yl) acrylamide
Figure BDA0003308477660000191
Synthetic prescriptionThe procedure is as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.93(t,J=1.9Hz,1H),8.64(dt,J=3.6,1.8Hz,1H),8.29-8.23(m,1H),8.03(dt,J=7.9,1.8Hz,1H),7.79-7.64(m,4H),7.63-7.54(m,3H),7.50(dddt,J=8.2,5.1,2.0,1.0Hz,1H),7.24(t,J=8.2Hz,1H),6.60(d,J=15.9Hz,1H),4.28(t,J=1.0Hz,2H),3.67-3.59(m,2H),3.26-3.15(m,4H),2.58(t,J=5.3Hz,4H),2.38(t,J=6.3Hz,2H),1.59-1.47(m,4H),1.47-1.44(m,1H),1.44-1.38(m,1H). 13 C NMR(125MHz,Chloroform-d)δ167.37,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,150.23,149.58,138.69,134.33,132.74,132.42-132.03(m),130.76,129.93-129.64(m),129.24,127.35,125.74,125.58,123.91,123.26,120.72,115.59,115.43,56.16,53.33,45.21,39.73,36.72,28.70,26.50,25.03.
Example 10: (E) Preparation of (E) -N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -3- (pyridin-3-yl) acrylamide
Figure BDA0003308477660000192
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.98(t,J=1.9Hz,1H),8.64(dt,J=3.6,1.8Hz,1H),8.29-8.23(m,1H),8.13(dt,J=7.9,1.9Hz,1H),7.79(t,J=4.4Hz,1H),7.76-7.64(m,3H),7.61(dd,J=7.9,3.5Hz,1H),7.54-7.45(m,2H),7.30(d,J=15.9Hz,1H),7.23(t,J=8.1Hz,1H),6.60(d,J=15.9Hz,1H),4.29(t,J=1.0Hz,2H),3.66(ddd,J=11.7,6.0,4.6Hz,2H),3.27-3.16(m,4H),2.63-2.51(m,4H),2.37(t,J=6.4Hz,2H),1.57(p,J=6.5Hz,2H),1.46(p,J=6.6Hz,2H),1.38-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ167.37,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,150.23,149.58,138.69,134.33,132.74,132.42-132.03(m),130.76,129.76(dd,J=13.6,8.1Hz),129.33,127.35,125.74,125.58,123.91,123.26,120.72,115.59,115.43,56.16,53.41,45.21,39.72,36.72,28.51,27.30,27.11,26.71.
Example 11: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) -1H-pyrrolo [3,2-c ] pyridine-3-carboxamide
Figure BDA0003308477660000193
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.31(d,J=1.8Hz,1H),8.59(t,J=4.4Hz,1H),8.34(dd,J=5.7,1.5Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),8.10(d,J=2.6Hz,1H),7.787.65(m,3H),7.62(ddt,J=5.0,2.0,1.0Hz,1H),7.52-7.42(m,2H),7.26-7.19(m,1H),4.27(t,J=1.0Hz,2H),3.68-3.59(m,2H),3.38(td,J=6.2,4.4Hz,2H),3.30-3.22(m,2H),2.63-2.53(m,4H),2.46(t,J=6.3Hz,2H),1.68-1.60(m,2H),1.55(pd,J=6.3,1.1Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),165.54,160.15,158.42,158.13,151.88,143.76(d,J=8.1Hz),138.80,132.71,132.42-132.03(m),130.76,130.10,129.93-129.68(m),129.24,127.35,125.74,125.58,123.91,122.11,115.59,115.43,108.41,108.07,56.03,53.41,45.21,39.81,36.72,26.78,24.99.
Example 12: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -1H-pyrrolo [3,2-c ] pyridine-3-carboxamide
Figure BDA0003308477660000201
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.43(d,J=1.6Hz,1H),8.51(t,J=4.4Hz,1H),8.34(dd,J=5.7,1.6Hz,1H),8.29-8.23(m,1H),8.08(d,J=2.6Hz,1H),7.78-7.64(m,3H),7.61(ddt,J=5.0,2.0,1.0Hz,1H),7.51-7.42(m,2H),7.24(t,J=8.2Hz,1H),4.19(t,J=1.0Hz,2H),3.86-3.78(m,2H),3.36-3.24(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.56(dp,J=34.0,6.5Hz,4H),1.44-1.34(m,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),165.61,160.15,158.42,158.13,151.88,143.80(d,J=3.3Hz),138.80,132.74,132.42-132.03(m),130.76,130.15,129.93-129.59(m),129.33,127.35,125.74,125.58,123.91,122.11,115.59,115.43,108.43,108.07,56.16,53.41,45.21,39.99,36.72,28.70,26.50,25.02.
Example 13: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -1H-pyrrolo [3,2-c ] pyridine-3-carboxamide
Figure BDA0003308477660000202
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.43(d,J=1.8Hz,1H),8.55(t,J=4.3Hz,1H),8.34(dd,J=5.7,1.6Hz,1H),8.29-8.23(m,1H),7.78-7.68(m,4H),7.67(dd,J=7.4,1.7Hz,1H),7.56-7.45(m,2H),7.45(dd,J=5.6,2.1Hz,1H),7.24(t,J=8.2Hz,1H),4.29(t,J=1.0Hz,2H),3.67-3.58(m,2H),3.34-3.25(m,4H),2.58(dd,J=5.7,5.0Hz,4H),2.37(t,J=6.4Hz,2H),1.62(p,J=6.6Hz,2H),1.45(p,J=6.6Hz,2H),1.38-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),165.63,160.15,158.42,158.13,151.88,144.05,143.79,138.80,132.74,132.42-132.03(m),130.76,130.15,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.91,122.11,115.59,115.43,108.43,108.07,56.16,53.41,45.21,39.94,36.72,28.60,27.33,27.11,26.75.
Example 14: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) ethyl) -1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000211
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.61(d,J=1.8Hz,1H),8.40(dd,J=5.6,1.7Hz,1H),8.26(dd,J=7.7,1.8Hz,1H),7.72(dtd,J=22.5,7.4,1.8Hz,2H),7.68-7.61(m,2H),7.49(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.37(d,J=5.7Hz,1H),7.24(t,J=8.2Hz,1H),6.48(t,J=4.4Hz,1H),4.86(s,2H),4.30(s,2H),4.26(t,J=1.0Hz,2H),3.69(ddd,J=11.7,5.9,4.7Hz,2H),3.48-3.41(m,2H),3.41-3.35(m,2H),2.67(t,J=5.6Hz,2H),2.59-2.48(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,157.77,151.85,146.68,145.95,143.08,132.71,132.57-132.03(m),130.75,130.00-129.70(m),129.24,127.35,125.74,125.58,123.91,116.96,115.59,115.43,55.79,52.71,52.49,52.15,45.21,38.03,36.72.
Example 15: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) propyl) -1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000212
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.62(d,J=1.8Hz,1H),8.40(dd,J=5.7,1.8Hz,1H),8.26(dd,J=7.7,1.8Hz,1H),7.78-7.63(m,3H),7.55(ddt,J=5.0,2.0,1.0Hz,1H),7.50(dddt,J=8.7,4.9,1.9,0.9Hz,1H),7.36(d,J=5.6Hz,1H),7.24(t,J=8.2Hz,1H),6.68(t,J=4.4Hz,1H),4.86(s,2H),4.40(d,J=13.4Hz,1H),4.33-4.26(m,3H),3.65(dt,J=11.7,5.2Hz,2H),3.35(td,J=6.3,4.4Hz,2H),3.26(dt,J=11.7,5.3Hz,2H),2.59(t,J=5.3Hz,4H),2.45(t,J=6.4Hz,2H),1.76(p,J=6.4Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,157.90,151.85,146.68,145.95,143.08,132.71,132.48-132.03(m),130.75,129.81(d,J=8.2Hz),129.24,127.35,125.74,125.58,123.91,117.04,115.59,115.43,53.26(d,J=7.9Hz),52.49,52.15,45.21,38.86,36.72,25.79.
Example 16: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) -1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000221
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.60(d,J=1.8Hz,1H),8.40(dd,J=5.7,1.8Hz,1H),8.29-8.23(m,1H),7.78-7.64(m,3H),7.63(ddt,J=5.0,2.0,1.0Hz,1H),7.50(dddt,J=8.0,5.1,2.0,1.1Hz,1H),7.37(d,J=5.7Hz,1H),7.24(t,J=8.2Hz,1H),6.46(t,J=4.4Hz,1H),4.86(s,2H),4.31(s,2H),4.26(t,J=1.0Hz,2H),3.73(dt,J=11.7,5.3Hz,2H),3.34(dt,J=11.7,5.3Hz,2H),3.18(td,J=6.2,4.4Hz,2H),2.59(t,J=5.3Hz,4H),2.46(t,J=6.4Hz,2H),1.64(p,J=6.7Hz,2H),1.55-1.46(m,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.11(d,J=4.5Hz),151.88,146.68,145.95,143.08,132.71(d,J=7.2Hz),132.42-132.03(m),130.76,129.93-129.68(m),129.24,127.35,125.74,125.58,123.91,117.04,115.59,115.43,56.03,53.41,52.49,52.18,45.21,39.43,36.72,26.62,24.99.
Example 17: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -1, 3-dihydro-2H-pyrrole [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000222
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.60(d,J=1.8Hz,1H),8.40(dd,J=5.5,1.8Hz,1H),8.29-8.23(m,1H),7.78-7.61(m,4H),7.50(dddt,J=8.2,5.1,2.0,1.0Hz,1H),7.37(d,J=5.7Hz,1H),7.24(t,J=8.2Hz,1H),6.70(t,J=4.4Hz,1H),4.86(s,2H),4.31(s,2H),4.26(t,J=1.0Hz,2H),3.78-3.69(m,2H),3.38-3.29(m,2H),3.16(td,J=6.3,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.59-1.44(m,4H),1.43-1.33(m,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.11(d,J=4.5Hz),151.88,146.68,145.95,143.08,132.74,132.50-132.03(m),130.76,129.93-129.59(m),129.33,127.35,125.74,125.58,123.91,117.04,115.59,115.43,56.16,53.41,52.49,52.18,45.21,39.50,36.72,28.39,26.50,25.01.
Example 18: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000231
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.61(d,J=1.8Hz,1H),8.40(dd,J=5.7,1.8Hz,1H),8.29-8.23(m,1H),7.78-7.65(m,3H),7.62(ddt,J=4.9,2.0,1.0Hz,1H),7.49(dddt,J=8.8,5.1,2.0,1.0Hz,1H),7.36(d,J=5.6Hz,1H),7.24(t,J=8.2Hz,1H),6.58(t,J=4.4Hz,1H),4.86(s,2H),4.30(s,2H),4.26(t,J=1.0Hz,2H),3.71(ddd,J=11.7,5.9,4.8Hz,2H),3.37(ddd,J=11.7,5.9,4.8Hz,2H),3.16(td,J=6.3,4.4Hz,2H),2.63-2.52(m,4H),2.37(t,J=6.4Hz,2H),1.52-1.40(m,4H),1.37-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.11(d,J=4.5Hz),151.88,146.70,145.95,143.08,132.74,132.50-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.91,117.04,115.59,115.43,56.16,53.41,52.49,52.18,45.21,39.56,36.72,28.76,27.33,27.11,26.60.
Example 19: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) imidazo [1,2-a ] pyrimidine-7-carboxamide
Figure BDA0003308477660000232
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.38(d,J=8.9Hz,1H),8.34(t,J=4.4Hz,1H),8.29-8.22(m,1H),8.08(s,1H),7.79-7.63(m,4H),7.47(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.36(s,1H),7.28-7.15(m,2H),4.26(t,J=1.0Hz,2H),3.78-3.68(m,2H),3.42-3.31(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.58(dp,J=14.9,6.6Hz,4H),1.46-1.34(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.73(d,J=7.9Hz),163.51,158.23,151.89,145.53,143.19,132.54-132.28(m),132.07(d,J=2.9Hz),130.82,129.87(d,J=7.9Hz),129.54(d,J=3.1Hz),129.32,129.14,127.36,123.89,115.58,115.42,113.57,111.37,56.23,53.50,45.22,40.13,36.72,28.57,26.49,25.00.
Example 20: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -3a,7 a-dihydro-1H-pyrazolo [3,4-b ] pyridine-6-carboxamide
Figure BDA0003308477660000233
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.28-8.23(m,1H),7.80-7.72(m,2H),7.72-7.64(m,2H),7.52(t,J=4.3Hz,1H),7.46(dddt,J=8.6,5.0,1.9,0.9Hz,1H),7.36(d,J=5.5Hz,1H),7.22(t,J=8.2Hz,1H),6.97(d,J=6.6Hz,1H),6.89(dd,J=9.3,1.0Hz,1H),6.08(dd,J=9.2,5.7Hz,1H),5.26(dd,J=6.6,5.1Hz,1H),4.40(dt,J=11.4,1.0Hz,1H),4.11(dt,J=11.3,1.0Hz,1H),3.76-3.68(m,2H),3.49(qd,J=5.6,1.1Hz,1H),3.41-3.33(m,2H),3.27(dtdJ=14.3,6.2,4.3Hz,1H),3.12(dtd,J=14.4,6.2,4.4Hz,1H),2.58(t,J=5.4Hz,4H),2.43(dt,J=11.9,6.4Hz,1H),2.32(dt,J=12.0,6.3Hz,1H),1.64-1.28(m,6H). 13 C NMR(125MHz,Common NMR Solvents)δ166.64,165.76,165.70,159.83,158.23,157.81,153.23,151.89,134.56,132.46,132.42,132.36,132.09,132.06,130.82,129.90,129.84,129.52,129.14,128.57,127.48,127.36,125.65,125.49,123.89,115.58,115l42,74.73,56.23,53.50,45.22,43.28,40.29,36.72,28.65,26.49.25.00.
Example 21: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) furo [2,3-c ] pyridine-2-carboxamide
Figure BDA0003308477660000241
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.85(d,J=1.7Hz,1H),8.72(dd,J=5.7,1.6Hz,1H),8.30-8.23(m,2H),7.91(d,J=2.1Hz,1H),7.79(ddt,J=5.0,2.0,1.0Hz,1H),7.78-7.65(m,4H),7.46(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.22(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.72(dt,J=11.7,5.2Hz,2H),3.42-3.31(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.57(dp,J=19.4,6.5Hz,4H),1.44-1.35(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.76,165.70,161.31,159.83,158.23,157.81,151.89,151.31,151.18,144.55,134.40,132.69,132.46,132.42,132.36,132.09,132.06,130.82,129.90,129.84,129.52,129.14,127.36,125.65,125.49,123.89,117.65,115.58,115.42,110.56,56.23,53.50,45.22,39.93,36.72,28.69,26.49,25.00.
Example 22: preparation of 4-fluoro-N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) nicotinamide
Figure BDA0003308477660000242
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.92(dd,J=5.0,1.8Hz,1H),8.65-8.59(m,1H),8.26(dd,J=7.6,1.7Hz,1H),8.10(t,J=4.4Hz,1H),7.78-7.64(m,3H),7.63(ddt,J=5.0,2.0,1.0Hz,1H),7.50(dddt,J=8.0,5.0,2.0,1.1Hz,1H),7.28-7.20(m,2H),4.26(t,J=1.0Hz,2H),3.71(dt,J=11.7,5.2Hz,2H),3.42-3.33(m,2H),3.19(td,J=6.2,4.4Hz,2H),2.58(t,J=5.3Hz,4H),2.38(t,J=6.4Hz,2H),1.57(dp,J=11.1,6.5Hz,4H),1.41(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.73(dJ=7.9Hz),164.60-164.34(m),162.40,159.83,158.23,157.81,152.01-151.61(m),132.57-132.30(m),132.07(d,J=2.9Hz),130.82,129.93(d,J=8.1Hz),129.59,129.06,127.36,125.65,125.49,123.89,120.91,120.75,115.58,115.42,110.08,109.92,56.23,53.46,45.22,40.10,36.72,28.79,26.50,25.00.
Example 23: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -1H-pyrrolo [3,2-c ] pyridine-2-carboxamide
Figure BDA0003308477660000251
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.92(s,1H),9.01(t,J=1.7Hz,1H),8.40-8.31(m,2H),8.29-8.23(m,1H),7.78-7.64(m,3H),7.63(ddt,J=4.9,2.0,0.9Hz,1H),7.54-7.46(m,2H),7.39(d,J=1.4Hz,1H),7.24(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.76-3.68(m,2H),3.44-3.32(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.58(dp,J=22.7,6.5Hz,4H),1.45-1.35(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.73(d,J=7.9Hz),162.74,159.83,158.23,157.81,151.89,143.59,142.86,140.02,132.54-132.28(m),132.07(d,J=2.9Hz),130.82,129.87(d,J=7.9Hz),129.52,129.14,127.36,125.65,125.49,124.82,123.89,115.58,115.42,108.41,104.28,56.23,53.50,45.22,40.26,36.72,28.69,26.49,25.00.
Example 24: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) thieno [2,3-c ] pyridine-2-carboxamide
Figure BDA0003308477660000252
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.03(d,J=1.6Hz,1H),8.76(dd,J=5.6,1.7Hz,1H),8.44(t,J=4.4Hz,1H),8.29-8.23(m,1H),8.17-8.07(m,2H),7.80(ddt,J=4.9,2.0,1.0Hz,1H),7.77-7.64(m,3H),7.44(dddt,J=8.0,5.1,2.0,1.0Hz,1H),7.21(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.78-3.70(m,2H),3.44-3.33(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.58(dp,J=19.5,6.5Hz,4H),1.41(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.76,165.70,164.56,159.83,158.23,157.81,151.89,146.13,144.48,143.16,139.02,136.76,132.53,132.42,132.36,132.09,132.06,130.82,129.99,129.93,129.52,129.14,127.36,125.65,125.49,124.49,123.89,119.96,115.58,115.42,56.23,53.50,45.22,40.08,36.72,28.70,26.49,25.00.
Example 25: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -6-methylnicotinamide
Figure BDA0003308477660000253
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.01(d,J=1.9Hz,1H),8.29-8.18(m,2H),7.89(ddJ=8.4,1.8Hz,1H),7.81-7.65(m,4H),7.46(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.30(dq,J=8.4,0.8Hz,1H),7.22(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.76-3.67(m,2H),3.42-3.34(m,2H),3.24(td,J=6.2,4.3Hz,2H),2.62-2.53(m,7H),2.38(t,J=6.4Hz,2H),1.56(dp,J=10.3,6.5Hz,4H),1.40(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ166.86,165.76,165.70,159.83,158.23,157.81,156.77,151.89,148.29,134.96,132.49,132.45,132.38,132.09,132.06,130.82,129.97,129.90,129.59,129.06,128.32,127.36,125.65,125.49,123.89,122.76,115.58,115.42,56.23,53.46,45.22,39.79,36.72,28.75,26.50,25.00.23.95.
Example 26: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) nicotinamide
Figure BDA0003308477660000261
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.14(t,J=1.8Hz,1H),8.77(ddd,J=4.6,2.6,1.8Hz,1H),8.29-8.21(m,2H),8.11(t,J=4.4Hz,1H),7.78-7.63(m,4H),7.50(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.43(dd,J=7.9,4.8Hz,1H),7.24(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.75-3.66(m,2H),3.42-3.33(m,2H),3.25(td,J=6.2,4.4Hz,2H),2.62-2.53(m,4H),2.38(t,J=6.4Hz,2H),1.57(dp,J=11.3,6.6Hz,4H),1.41(q,J=7.0Hz,2H).13C NMR(125MHz,Common NMR Solvents)δ166.61,165.76,165.70,159.83,158.23,157.81,151.89,150.73,149.53,135.48,132.51,132.42,132.35,132.11,132.08,130.82,130.25,129.99,129.93,129.59,129.07,127.36,125.65,125.49,123.89,123.62,115.58,115.42,56.23,53.56,45.19,39.79,36.72,28.78,26.54,25.00.
Example 27: preparation of 6-amino-N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) nicotinamide
Figure BDA0003308477660000262
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.83(d,J=1.9Hz,1H),8.29-8.21(m,2H),7.78-7.63(m,5H),7.48(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.23(t,J=8.2Hz,1H),6.32(d,J=8.4Hz,1H),5.92(d,J=6.2Hz,1H),5.80(d,J=6.2Hz,1H),4.26(t,J=1.0Hz,2H),3.71(dt,J=11.7,5.2Hz,2H),3.42-3.33(m,2H),3.30-3.13(m,2H),2.58(t,J=5.3Hz,4H),2.38(t,J=6.4Hz,2H),1.58(dp,J=28.9,6.5Hz,4H),1.44-1.34(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ166.76,165.76,165.70,160.35,159.83,158.23,157.81,151.89,149.18,136.09,132.49,132.45,132.38,132.09,132.06,130.82,129.97,129.90,129.59,129.06,127.36,125.65,125.49,123.89,122.06,115.58,115.42,107.59,56.23,53.46,45.22,39.79,36.72,28.75,26.50,25.00.
Example 28: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) isonicotinamide
Figure BDA0003308477660000263
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.73-8.62(m,2H),8.26(dd J=7.6,1.7Hz,1H),7.89(t,J=4.4Hz,1H),7.78-7.62(m,6H),7.50(dddt,J=8.0,4.9,2.0,1.1Hz,1H),7.24(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.70(dt,J=11.7,5.3Hz,2H),3.42-3.33(m,4H),2.62-2.53(m,4H),2.38(t,J=6l3Hz,2H),1.57(dp,J=13.1,6.5Hz,4H),1.41(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ167.71,165.76,165.70,159.83,158.23,157.81,151.89,150.28,139.57,132.51,132l42,132l35,132.11,132.08,130.82,129.99,129.93,129.59,129.07,127l36,125.65,125.49,123.89,121.22,115.58,115.42,56.23,53.56,45.19,39.70,36.72,28.75,26.54,25.00.
Example 29: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -6- (trifluoromethyl) nicotinamide
Figure BDA0003308477660000271
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ9.07(d,J=1.9Hz,1H),8.29-8.23(m,1H),8.15(t,J=4.3Hz,1H),8.00(dd,J=8.4,1.8Hz,1H),7.78-7.64(m,4H),7.63(ddt,J=5.0,2.0,1.1Hz,1H),7.50(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.24(t,J=8.2Hz,1H),4.26(t,J=0.9Hz,2H),3.72(dt,J=11.7,5.3Hz,2H),3.41-3.32(m,2H),3.15(td,J=6.3,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.57(dp,J=13.0,6.5Hz,4H),1.40(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ166.69,165.76,165.70,159.83,158.23,157.81,151.89,149.48,149.22,148.77,148.75,148.74,148.72,136.06,136.04,136.02,136.01,132.53,132.42,132.36,132.09,132.06,130.82,130.08,129.99,129.93,129.52,129.07,127.36,125.65,125.49,123.91,122.76,120.61,119.84,119.81,119.78,119.75,115.58,115.42,56.23,53.50,45.22,39.79,36.72,28.72,26.49,24.98.
Example 30: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -2-methyl isonicotinamide
Figure BDA0003308477660000272
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.65(d,J=5.7Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),7.96(t,J=4.3Hz,1H),7.83-7.64(m,6H),7.44(dddt,J=8.6,5.0,1.8,0.9Hz,1H),7.21(t,J=8.2Hz,1H),4.26(t,J=0.9Hz,2H),3.77-3.69(m,2H),3.45-3.36(m,2H),3.28(td,J=6.3,4.4Hz,2H),2.62-2.53(m,7H),2.38(t,J=6.3Hz,2H),1.57(pd,J=6.6,3.0Hz,4H),1.45-1.36(m,2H). 13 C NMR(125MHz,CommonNMR Solvents)δ167.42,165.76,165.70,159.83,159.35,158.23,157.81,151.89,146.12,138.74,132.49,132.45,132.38,132.09,132.06,130.82,129.97,129.90,129.59,129.06,127.36,125.65,125.49,123.89,120.45,119.13,115.58,115.42,56.23,53.46,45.22,39.70,36.72,28.73,26.50,25.00,23.75.
Example 31: preparation of 2-fluoro N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) isonicotinamide
Figure BDA0003308477660000281
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.52(d,J=5.6Hz,1H),8.26(dd,J=7.6,1.7Hz,1H),8.03(t,J=4.3Hz,1H),7.80(ddt,J=5.0,2.0,1.0Hz,1H),7.77-7.63(m,4H),7.50(dd,J=5.7,1.8Hz,1H),7.44(dddt,J=8.0,4.9,2.0,1.1Hz,1H),7.21(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.73(dt,J=11.7,5.2Hz,2H),3.45-3.36(m,2H),3.27(td,J=6.3,4.4Hz,2H),2.58(t,J=5.3Hz,4H),2.38(t,J=6.3Hz,2H),1.57(dp,J=8.2,6.6Hz,4H),1.45-1.36(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ167.83,167.80,165.76,165.70,163.29,161.27,159.83,158.23,157.81,151.89,147.68,147.57,141.24,141.18,132.49,132.45,132.38,132.09,132.06,130.82,129.97,129.90,129.59,129.06,127.36,125.65,125.49,123.89,118.66,118.64,115.58,115.42,106.51,106.35,56.23,53.46,45.22,39.74,36.72,28.75,26.50,25.00.
Example 32: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -2- (trifluoromethyl) isonicotinamide
Figure BDA0003308477660000282
/>
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.80(d,J=5.6Hz,1H),8.29-8.23(m,1H),8.12-8.03(m,2H),7.80(ddt,J=5.0,2.0,0.9Hz,1H),7.77-7.64(m,3H),7.48-7.40(m,2H),7.21(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.74(dt,J=11.7,5.2Hz,2H),3.44-3.36(m,2H),3.23(td,J=6.3,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.65(p,J=6.5Hz,2H),1.56(p,J=6.6Hz,2H),1.45-1.36(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ168.03,165.76,165.70,159.83,158.23,157.81,151.89,149.23,148.98,147.33,147.31,147.30,147.28,140.03,140.02,132.53,132.42,132.36,132.09,132.06,130.82,129.99,129.93,129.52,129.07,127.36,125.65,125.49,123.91,123.20,122.11,121.06,117.61,117.58,117.55,117.51,115.58,115.42,56.23,53.50,45.22,39.71,36.72,28.72,26.49,24.98.
Example 33: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) imidazo [1,2-a ] pyridine-6-carboxamide
Figure BDA0003308477660000283
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.65(d,J=1.4Hz,1H),8.33-8.23(m,2H),7.95(dd,J=9.1,1.6Hz,1H),7.83-7.63(m,7H),7.44(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.21(t,J=8.2Hz,1H),4.26(t,J=1.0Hz,2H),3.74(dt,J=11.7,5.3Hz,2H),3.44-3.36(m,2H),3.20(td,J=6.3,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.58(dp,J=19.8,6.6Hz,4H),1.41(q,J=7.0Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ166.74,165.76,165.70,159.83,158.23,157.81,151.89,140.68,134.40,132.46,132.42,132.36,132.09,132.06,130.90,130.82,129.90,129.84,129.52,129.14,127.36,125.65,125.49,123.89,122.17,118.43,115.58,115.42,113.21,112.79,56.23,53.50,45.22,40.07,36.72,28.58,26.49,25.00.
Example 34: preparation of 6-amino-N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -1H-pyrrolo [3,2-c ] pyridine-2-carboxamide
Figure BDA0003308477660000291
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ10.00(s,1H),8.69(d,J=1.5Hz,1H),8.53(t,J=4.4Hz,1H),8.29-8.23(m,1H),7.78-7.64(m,4H),7.46(dddt,J=7.9,4.9,1.9,1.0Hz,1H),7.28-7.20(m,2H),6.68(s,1H),5.84(d,J=6.0Hz,1H),5.77(d,J=6.0Hz,1H),4.21(t,J=1.0Hz,2H),3.77-3.68(m,2H),3.44-3.35(m,4H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.59(dp,J=17.5,6.6Hz,4H),1.41(q.J=6.9Hz,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.76,165.70,162.72,159.83,158.23,157.81,154.72,151.89,144.53,143.14,132.51,132.42,132.36,132.16,132.09,132.06,130.82,129.99,129.93,129.52,129.13,127.36,125.65,125.49,123.89,115.64,115.58,115.42,104.21,89.88,56.23,53.50,45.22,40.26,36.72,28.69,26.49,24.98.
Example 35: preparation of 5-amino N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) furo [2,3-c ] pyridine-2-carboxamide
Figure BDA0003308477660000292
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.49(s,1H),8.35(t,J=4.4Hz,1H),8.29-8.23(m,1H),7.80(ddt,J=5.0,2.0,1.0Hz,1H),7.77-7.64(m,3H),7.50-7.40(m,2H),7.21(t,J=8.2Hz,1H),7.03(d,J=2.3Hz,1H),5.35(d,J=6.2Hz,1H),5.23(d,J=6.2Hz,1H),4.26(t,J=1.0Hz,2H),3.79-3.70(m,2H),3.45-3.37(m,2H),3.34(td,J=6.3,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.3Hz,2H),1.58(dp,J=15.0,6.6Hz,4H),1.45-1.36(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.76,165.70,161.27,159.83,158.23,157.81,152.75,151.89,150.20,143.41,133.99,133.20,132.51,132.42,132.36,132.09,132.06,130.82,129.99,129.93,129.52,129.13,127.36,125.65,125.49,123.89,115.58,115.42,109.00,98.52,56.23,53.50,45.22,39.93,36.72,28.69,26.49,24.98.
Example 36: preparation of 6-amino N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000293
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.37-8.17(m,1H),8.11(s,1H),7.79-7.64(m,4H),7.47(dddt,J=8.7,4.9,1.9,0.9Hz,1H),7.22(t,J=8.2Hz,1H),6.74(t,J=4.4Hz,1H),6.52(s,1H),5.32(d,J=6.0Hz,1H),5.22(d,J=6.0Hz,1H),4.33-4.23(m,6H),3.76-3.67(m,2H),3.42-3.33(m,2H),3.14(td,J=6.2,4.4Hz,2H),2.58(t,J=5.4Hz,4H),2.38(t,J=6.4Hz,2H),1.59-1.53(m,2H),1.53-1.45(m,3H),1.45-1.35(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.73(d,J=7.9Hz),159.83,158.42,158.23(d,J=1.1Hz),157.81,151.89,148.75,140.58,132.59-132.28(m),132.07(d,J=2.9Hz),130.82,129.96(d,J=7.9Hz),129.52,129.13,127.36,125.65,125.49,123.89,121.12,115.58,115.42,101.83,56.23,53.50,52.11,51.90,45.22,39.63,36.72,28.35,26.49,24.97.
Example 37: preparation of N- (5- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-phthalazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -6-methyl-1, 3-dihydro-2H-pyrrolo [3,4-c ] pyridine-2-carboxamide
Figure BDA0003308477660000301
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.49(s,1H),8.29-8.23(m,1H),7.78-7.61(m,4H),7.49(dddt,J=8.2,5.1,2.0,1.0Hz,1H),7.29(s,1H),7.24(t,J=8.2Hz,1H),6.74(t,J=4.4Hz,1H),4.82(s,2H),4.42(d, J=13.5Hz,1H),4.35-4.24(m,3H),3.76-3.67(m,2H),3.42-3.33(m,2H),3.16(td,J=6.2,4.4Hz,2H),2.62-2.53(m,7H),2.38(t,J=6.3Hz,2H),1.59-1.45(m,4H),1.45-1.36(m,2H). 13 C NMR(125MHz,Common NMR Solvents)δ165.76,165.70,159.83,158.23,158.12,157.81,157.66,151.89,147.88,142.02,132.51,132.42,132.36,132.09,132.06,130.82,130.18,129.99,129.93,129.52,129.13,127.36,125.65,125.49,123.89,117.17,115.58,115.42,56.23,53.50,52.12,52.07,45.22,39.63,36.72,28.35,26.49,24.97,24.47.
Example 38: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -2- (pyridin-3-yl) cyclopropyl-1-carboxamide
Figure BDA0003308477660000302
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.56(dt,J=4.8,1.8Hz,1H),8.33-8.23(m,2H),7.78-7.64(m,3H),7.59(ddt,J=5.0,2.0,1.0Hz,1H),7.56-7.46(m,2H),7.31(dd,J=7.8,4.7Hz,1H),7.24(t,J=8.2HZ,1H),7.03(t,J=4.4Hz,1H),4.40(dt,J=11.5,1.1Hz,1H),4.13(dt,J=11.3,0.9Hz,1H),3.85-3.77(m,2H),3.42-3.33(m,2H),3.19(dtd,J=14.3,6.2,4.3Hz,1H),2.98(dtd,J=14.3,6.2,4.3Hz,1H),2.63-2.52(m,5H),2.47-2.38(m,1H),2.37-2.24(m,2H),1.66-1.21(m,8H). 13 C NMR(125MHz,Chloroform-d)δ173.48,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,149.37,148.19,135.03,132.74,132.53-132.03(m),130.76,129.93-129.59(m),129.33,127.35,125.74,125.58,123.87(d,J=11.0Hz),115.59,115.43,56.16,53.41,45.21,39.56,36.72,28.70,27.33,26.53,25.02,23.95,17.27.
Example 39: preparation of N- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -2- (pyridin-3-yl) cyclopropyl-1-carboxamide
Figure BDA0003308477660000312
The synthesis was as in example 1. 1 H NMR(500MHz,Chloroform-d)δ8.56(dt,J=4.8,1.8Hz,2H),8.32(t,J=1.9Hz,2H),8.29-8.23(m,2H),7.78-7.68(m,6H),7.67(dd,J=7.4,1.7Hz,1H),7.52-7.44(m,4H),7.30-7.19(m,4H),7.06(t,J=4.3Hz,2H),4.41(dt,J=11.3,1.0Hz,2H),4.11(dt,J=11.3,0.9Hz,2H),3.72-3.63(m,4H),3.39-3.31(m,4H),3.23(dtd,J=14.3,6.2,4.2Hz,2H),2.97(dtd,J=14.3,6.4,4.4Hz,2H),2.63-2.52(m,9H),2.43(dt,J=11.9,6.3Hz,2H),2.38-2.27(m,4H),1.66-1.58(m,2H),1.58-1.31(m,10H),1.31-1.16(m,6H). 13 C NMR(125MHz,Chloroform-d)δ173.48,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,149.37,148.19,135.13,132.74,132.53-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.87(d,J=11.0Hz),115.59,115.43,56.16,53.41,45.21,39.46,36.72,28.57,27.33,27.11,26.71,23.95,17.27.
Example 40: synthetic route of the compound of formula I (Synthesis of B series Compound)
Figure BDA0003308477660000311
Preparation of methyl-N' -cyano-N- (pyridin-4-yl) amidinothio
Figure BDA0003308477660000321
NaH (60%, 5g,127.4 mmol), DMF (100 ml) and Compound 8 (10.0g,106.2mmol,in 50ml DMF) were added dropwise at 0deg.C, after the completion of the dropwise addition, the mixture was stirred at 0deg.C for half an hour, compound 9 (15.6g,106.2mmol,in 50ml DMF) was added dropwise, after the completion of the dropwise addition, the reaction solution was allowed to stand at room temperature and reacted for 16 hours. After the reaction, the reaction mixture was washed three times with diethyl ether/petroleum ether=1:5, and the pH was adjusted to 8 with glacial acetic acid, and the reaction mixture was suction-filtered to give methyl-N' -cyano-N- (pyridin-4-yl) carbamimidothioide (14.1 g, 69%).
Preparation of tert-butyl-4- (2- (2-cyano-3- (pyridin-4-yl) guanidino) ethyl) piperazine-1-carboxylate
Figure BDA0003308477660000322
Into a 50ml round bottom flask was charged compound 10 (1.0 g,5.2 mmol), 4- (2-aminoethyl) -1-t-butoxycarbonylpiperazine (1.2 g,5.2 mmol), triethylamine (1.4 ml,10.4 mmol), DMAP (122 mg,1.0 mmol), pyridine 20ml, and reacted at 50℃for 12 hours. After the reaction, the pyridine was dried by spin, water was added, ethyl acetate was added, the organic phase was separated, the aqueous phase was extracted twice with an appropriate amount of ethyl acetate, the organic phases were combined, washed with saturated saline, dried over anhydrous sodium sulfate, concentrated, and column-chromatographed to give tert-butyl-4- (2- (2-cyano-3- (pyridin-4-yl) guanidino) ethyl) piperazine-1-carboxylate (1.0 g, 52%).
Preparation of 2-cyano-1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) ethyl) -3- (piperidin-4-yl) guanidine
Figure BDA0003308477660000323
1 H NMR(500MHz,Chloroform-d)δ9.10(s,1H),8.40-8.35(m,2H),8.26(dd,J=7.6,1.7Hz,1H),8.02(t,J=4.4Hz,1H),7.96(ddt,J=4.9,2.0,1.1Hz,1H),7.78-7.63(m,3H),7.47(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.21(t,J=8.2Hz,1H),7.03-6.98(m,2H),4.25(t,J=1.0Hz,2H),3.76(dt,J=11.7,5.3Hz,2H),3.60(td,J=5.5,4.2Hz,2H),3.38-3.29(m,2H),2.58(dt,J=14.2,5.5Hz,6H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,154.49,151.85,149.78,144.41,132.71,132.37-132.03(m),130.75,130.00-129.70(m),129.16,127.35,125.74,125.58,123.91,118.00,115.59,115.43,112.92,55.70,52.46,45.21,40.41,36.72.
Example 41: preparation of 2-cyano-1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) propyl) -3- (piperidin-4-yl) guanidine
Figure BDA0003308477660000331
The synthesis was as in example 40. 1 H NMR(500MHz,Chloroform-d)δ8.81(s,1H),8.40-8.35(m,2H),8.26(dd,J=7.6,1.7Hz,1H),7.74(ddd,J=15.3,7.6,1.8Hz,2H),7.71-7.63(m,3H),7.59(ddt,J=4.9,2.0,1.0Hz,1H),7.50(dddt,J=8.0,4.9,2.0,1.1Hz,1H),7.24(t,J=8.2Hz,1H),7.03-6.98(m,2H),4.28(t,J=1.0Hz,2H),3.69(ddd,J=11.7,6.0,4.7Hz,2H),3.39(td,J=6.3,4.3Hz,2H),3.22(ddd,J=11.7,6.0,4.7Hz,2H),2.64-2.53(m,4H),2.45(t,J=6.4Hz,2H),1.77(p,J=6.3Hz,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,154.86,151.85,149.80,144.65,132.71,132.42-132.03(m),130.75,129.94-129.70(m),129.24,127.35,125.74,125.58,123.91,118.00,115.59,115.43,112.92,53.44,53.23,45.21,39.91,36.72,26.24.
Example 42: preparation of 2-cyano-1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) butyl) -3- (piperidin-4-yl) guanidine
Figure BDA0003308477660000332
The synthesis was as in example 40. 1 H NMR(500MHz,Chloroform-d)δ9.29(s,1H),8.40-8.35(m,2H),8.26(dd,J=7.6,1.7Hz,1H),7.80(t,J=4.4Hz,1H),7.78-7.68(m,3H),7.68-7.63(m,1H),7.49(dddt,J=8.6,5.0,1.8,0.9Hz,1H),7.23(t,J=8.2Hz,1H),7.03-6.98(m,2H),4.26(t,J=1.0Hz,2H),3.69(dt,J=11.5,5.3Hz,2H),3.51(td,J=6.2,4.4Hz,2H),3.37(dt,J=11.7,5.3Hz,2H),2.58(t,J=5.4Hz,4H),2.46(t,J=6.2Hz,2H),1.68-1.52(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,154.85,151.88,149.96,144.65,132.71,132.42-132.03(m),130.76,129.93-129.68(m),129.24,127.35,125.74,125.58,123.91,118.00,115.59,115.43,112.92,56.06,53.33,45.21,41.39,36.72,26.79,24.97.
Example 43: preparation of 2-cyano-1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -3- (piperidin-4-yl) guanidine
Figure BDA0003308477660000341
The synthesis was as in example 40. 1 H NMR(500MHz,Chloroform-d)δ8.95(s,1H),8.40-8.35(m,2H),8.29-8.23(m,1H),7.78-7.64(m,3H),7.64-7.58(m,2H),7.49(dddt,J=7.9,5.1,2.0,1.1Hz,1H),7.27-7.19(m,1H),7.03-6.98(m,2H),4.27(t,J=1.0Hz,2H),3.71(ddd,J=11.5,6.0,4.5Hz,2H),3.45(td,J=6.2,4.4Hz,2H),3.27(ddd,J=11.7,6.0,4.5Hz,2H),2.63-2.51(m,4H),2.38(t,J=6.3Hz,2H),1.56-1.46(m,4H),1.45-1.35(m,2H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,154.85,151.88,149.96,144.65,132.74,132.42-132.03(m),130.76,129.93-129.59(m),129.33,127.35,125.74,125.58,123.91,118.00,115.59,115.43,112.92,56.16,53.41,45.21,41.56,36.72,27.88,26.54,25.02.
Example 44: preparation of 2-cyano-1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-oxazin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -3- (piperidin-4-yl) guanidine
Figure BDA0003308477660000342
The synthesis was as in example 40. 1 H NMR(500MHz,Chloroform-d)δ8.89(s,1H),8.40-8.35(m,2H),8.29-8.23(m,1H),7.80(t,J=4.4Hz,1H),7.78-7.68(m,3H),7.68-7.64(m,1H),7.48(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.22(t,J=8.2Hz,1H),7.03-6.98(m,2H),4.25(t,J=1.0Hz,2H),3.77-3.69(m,2H),3.45(td,J=6.3,4.4Hz,2H),3.41-3.33(m,2H),2.58(dd,J=5.7,4.9Hz,4H),2.37(t,J=6.4Hz,2H),1.48(dp,J=33.0,6.5Hz,4H),1.37-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.42,158.13,154.85,151.88,149.96,144.25,132.74,132.42-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.91,118.00,115.59,115.43,112.92,56.16,53.41,45.21,41.56,36.72,28.56,27.30,27.11,26.63.
Example 45: synthetic route of the compound of formula I (Synthesis of C series Compound)
Figure BDA0003308477660000351
Preparation of tert-butyl 4- (2- (3- (pyridin-4-methyl) ureido) ethyl) piperazine-1-carboxylate
Figure BDA0003308477660000352
BCT (294 mg,1.0 mmol), nitrogen-protected, anhydrous tetrahydrofuran (10 ml) was added to a 25ml round bottom flask, compound 12 (216 mg,2.0mmol, dissolved in 5ml THF) was added at 0deg.C, after the addition was completed, the reaction was carried out at 70deg.C for 12 hours, after the reaction was completed, compound 4 (4- (2-aminoethyl) -1-t-butoxycarbonylpiperazine) (230 mg,1.0mmol, dissolved in 5ml THF) was added at 0deg.C, after the addition was completed, the reaction was carried out at 70deg.C for 12 hours. After the reaction, water was added to spin-dry tetrahydrofuran, ethyl acetate was added, the organic phase was separated, the aqueous phase was extracted twice with an appropriate amount of ethyl acetate, the organic phases were combined, washed with saturated saline water three times, dried over anhydrous sodium sulfate, concentrated, and column chromatographed to give tert-butyl 4- (2- (3- (pyridin-4-methyl) ureido) ethyl) piperazine-1-carboxylate (127 mg, 35%).
Preparation of 1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -3- (pyridin-3-methyl) urea
Figure BDA0003308477660000353
The synthesis was as in example 16. 1 H NMR(500MHz,Chloroform-d)δ8.61(t,J=1.7Hz,1H),8.49(ddd,J=3.6,2.2,1.5Hz,1H),8.29-8.23(m,1H),7.78-7.64(m,4H),7.63(ddt,J=4.9,2.0,1.1Hz,1H),7.50(dddt,J=8.6,4.9,1.9,0.9Hz,1H),7.31-7.20(m,2H),6.37(t,J=6.1Hz,1H),5.97(t,J=4.4Hz,1H),4.53(d,J=6.2Hz,2H),4.26(t,J=1.0Hz,2H),3.76-3.68(m,2H),3.41-3.33(m,2H),3.09(td,J=6.2,4.4Hz,2H),2.58(t,J=5.3Hz,4H),2.38(t,J=6.3Hz,2H),1.57-1.38(m,6H).13C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.86,158.42,158.13,151.88,148.44(d,J=11.2Hz),135.20,133.78,132.74,132.42-132.03(m),130.76,129.93-129.68(m),129.32,127.35,125.74,125.58,123.93(d,J=5.7Hz),115.59,115.43,56.16,53.33,45.21,42.02,39.06,36.72,28.33,26.50,25.02.
Example 46: preparation of 1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -3- (pyridin-3-methyl) urea
Figure BDA0003308477660000361
The synthesis was as in example 45. 1 H NMR(500MHz,Chloroform-d)δ8.51-8.46(m,2H),8.29-8.23(m,1H),7.78-7.64(m,4H),7.53-7.44(m,2H),7.30-7.22(m,2H),6.39(t,J=6.1Hz,1H),6.08(t,J=4.3Hz,1H),4.54(d,J=6.0Hz,2H),4.09(t,J=1.0Hz,2H),3.68(ddd,J=11.7,5.8,4.7Hz,2H),3.28(ddd,J=11.7,5.9,4.8Hz,2H),3.14(td,J=6.3,4.3Hz,2H),2.63-2.52(m,4H),2.37(t,J=6.4Hz,2H),1.50-1.40(m,4H),1.37-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ166.29(d,J=8.1Hz),160.15,158.86,158.42,158.13,151.88,148.44(d,J=11.4Hz),135.20,133.78,132.74,132.42-132.03(m),130.76,129.93-129.59(m),129.33,127.35,125.74,125.58,123.93(d,J=5.7Hz),115.59,115.43,56.16,53.41,45.21,42.02,38.85,36.72,28.73,27.30,27.11,26.60.
Example 47: synthetic route of the compound of formula I (Synthesis of D series Compound)
Figure BDA0003308477660000362
Preparation of tert-butyl-4- (2- (3- (pyridin-3-methyl) thiourea) ethyl) piperazine-1-carboxylate
Figure BDA0003308477660000363
Into a 25ml round bottom flask was charged thiophosgene (115 mg,1.0 mmol), nitrogen gas was used to protect, anhydrous toluene (10 ml) was added at 0℃and after the completion of the dropwise addition, compound 12 (108 mg,1.0mmol, dissolved in 5ml THF) was heated to 70℃for 5 hours, after the completion of the reaction, compound 4 (4- (2-aminoethyl) -1-t-butoxycarbonylpiperazine) (230 mg,1.0mmol, dissolved in 5ml THF) was added at 0℃without any post-treatment, and after the completion of the dropwise addition, the temperature was raised to 70℃for 5 hours. After the completion of the reaction, toluene was dried by spin-drying, water was added, ethyl acetate was added, the organic phase was separated, the aqueous phase was extracted twice with an appropriate amount of ethyl acetate, the organic phases were combined, washed with saturated saline water three times, dried over anhydrous sodium sulfate, concentrated, and column-chromatographed to give tert-butyl-4- (2- (3- (pyridin-3-methyl) thiourea) ethyl) piperazine-1-carboxylate (135 mg, 33%).
Preparation of 1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) pentyl) -3- (piperazine-3-methyl) thiourea
Figure BDA0003308477660000371
1 H NMR(500MHz,Chloroform-d)δ8.49-8.43(m,4H),8.29-8.23(m,2H),7.78-7.75(m,1H),7.75-7.64(m,9H),7.53(dddt,J=8.2,5.1,2.0,1.0Hz,2H),7.33-7.21(m,6H),7.06(t,J=4.4Hz,2H),4.86(d,J=6.2Hz,4H),4.25(t,J=1.0Hz,4H),3.71(dt,J=11.7,5.3Hz,4H),3.38-3.29(m,8H),2.58(t,J=5.3Hz,8H),2.38(t,J=6.4Hz,4H),1.52(dp,J=25.5,6.5Hz,8H),1.34(q,J=7.0Hz,4H). 13 C NMR(125MHz,Chloroform-d)δ180.76,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,148.43(d,J=8.3Hz),135.55,135.20,132.74,132.42-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.93(d,J=5.7Hz),115.59,115.43,56.16,53.33,45.21,44.84,42.88,36.72,28.12,26.55,25.03.
Example 48: preparation of 1- (2- (4- (2-fluoro-5- ((4-oxo-3, 4-dihydro-azin-1-yl) methyl) benzoyl) piperazin-1-yl) hexyl) -3- (piperazin-3-methyl) thiourea
Figure BDA0003308477660000372
The synthesis was as in example 47. 1 H NMR(500MHz,Chloroform-d)δ8.56(t,J=1.8Hz,1H),8.46(dt,J=3.7,1.9Hz,1H),8.29-8.23(m,1H),7.77(dt,J=7.8,2.0Hz,1H),7.75-7.65(m,3H),7.62(ddt,J=5.0,2.0,1.0Hz,1H),7.31-7.23(m,3H),7.23-7.16(m,1H),7.03(t,J=4.4Hz,1H),4.89(d,J=6.2Hz,2H),4.16(t,J=1.0Hz,2H),3.84-3.75(m,2H),3.46-3.37(m,2H),3.32(td,J=6.3,4.4Hz,2H),2.58(dd,J=5.7,4.9Hz,4H),2.39(t,J=6.4Hz,2H),1.48(dp,J=22.7,6.5Hz,4H),1.38-1.22(m,4H). 13 C NMR(125MHz,Chloroform-d)δ180.76,166.29(d,J=8.1Hz),160.15,158.42,158.13,151.88,148.43(d,J=8.3Hz),135.55,135.20,132.74,132.42-132.03(m),130.76,129.88(d,J=7.9Hz),129.67,129.32,127.35,125.74,125.58,123.94(d,J=4.8Hz),115.59,115.43,56.15,53.41,45.21,44.84,42.70,36.72,28.36,27.30,27.11,26.63.
Example 49: determination of PARP and NAMPT enzyme inhibitory Activity
PARP1 enzyme inhibition activity test method
PARP1 enzyme inhibition activity adopts a PARP homogeneous assay kit. The test system included 1 ng/. Mu.L PARP-1, 20 ng/. Mu.L activated DNA (Sigma), 500nM NAD dissolved in the following reaction buffer: 100mM Tris-HCl (pH 8.0), 100mM NaCl and 20mM MgCl 2 . PARP-1 protein (1 ng/. Mu.L) was pre-incubated with various concentrations of test compound for 30min at room temperature, followed by addition of 500nM NAD to initiate the PARylation reaction. The remaining unreacted NAD was incubated with the circulating test solution (1% ethanol, 25. Mu.M resazurin, 0.30U/mL alcohol dehydrogenase and 0.25U/mL diaphorase) and the concentration of NAD was proportional to the fluorescent signal of Ex 540nm/Em 590 nm. Assessment of the rate of decrease in NAD under the action of different concentrations of Compounds IC for PARP1 inhibitory Activity 50 Is a numerical value of (2).
NAMPT inhibition activity assays were performed according to the procedure provided by the CycLex NAMPT colorimetric assay kit (MBL International Corp.) manufacturer. 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 under the action of alcohol dehydrogenase (alcohol dehydrogenase, ADH) produces NADH, which returns to NAD under the action of diaphorase. WST-1 forms orange-yellow formazan in the enzymatic cycling of NAD/NADH, and the effect of a compound on NAMPT enzyme activity can be detected by detecting a change in absorbance at OD 450 nM.
The specific experimental operation steps are as follows:
(1) preparing a compound solution to be tested: all compounds were dissolved in DMSO at a stock concentration of 10mM, formulated to the required concentration as required for the test, and diluted 2-fold from the initial concentration, 8 concentration gradients were set, each concentration repeated three times.
(2) Taking ddH 2 O (10. Mu.L) and NAM (5. Mu.L) were added to 96-well assay plates;
(3) adding 5 mu L of a compound to be tested or DMSO into a 96-well detection plate;
(4) preparing a mixture I, including: 20 XNAMPT assay buffer (5. Mu.L), PRPP (5. Mu.L), ATP (5. Mu.L), recombinant NMNAT1 (5. Mu.L), ddH 2 O (35. Mu.L) and NAMPT (5. Mu.L) for a total of 60. Mu.L. Adding the mixture I into a 96-well detection plate;
(5) placing the 96-well detection plate into a 30 ℃ incubator for incubation for 60min;
(6) configuring the texture I includes: WST-1 (5. Mu.L), ADH (5. Mu.L), diaphorase (5. Mu.L), ethanol (5. Mu.L), and a total of 20. Mu.L. After incubation, the 96-well assay plate was removed and the mix II was added to the 96-well assay plate;
(7) dynamically monitoring the absorbance value of each hole at the OD 450nm in 30min on an enzyme-labeled instrument, and detecting every 5 min;
(8) and selecting a time period in which the absorbance and the time are linearly changed, and calculating the reaction rate. Treatment with GraphPad Prism5 software. To compensate for NAMPT inhibitory activity of DMSO, values were corrected using DMSO as a solvent control.
The specific experimental results are shown in table 1: the compound cells of the examples have better inhibitory activity on NAMPT and PARP 1.
Table 1: NAMPT and PARP1 inhibitory Activity
Figure BDA0003308477660000381
/>
Figure BDA0003308477660000391
/>
Figure BDA0003308477660000401
Example 50: antitumor Activity test
The data of the inhibiting activity of the compound cancer cells are detected by an MTT method, which is also called MTT colorimetric method, and is a method for detecting the survival and growth of the cells. MTT (yellow thiazole blue) can penetrate through cell membranes and enter cells, amber dehydrogenase in mitochondria of living cells can reduce exogenous MTT into bluish purple needle-shaped Formazan crystals which are difficult to dissolve in water and deposit in the cells, the crystals can be dissolved by dimethyl sulfoxide (DMSO), and the light absorption value of the crystals can be detected by an enzyme-linked immunosorbent assay (ELISA) at the wavelength of 490nm/570nm, so that the number of the living cells can be indirectly reflected. The cancer cell lines used were MCF-7 (human breast cancer cells), K562 (human chronic granulocytic leukemia cells), HT29 (human prostate cancer cells), A549 (human non-small cell lung cancer cells) and ACHN (renal cell carcinoma cells)
The specific experimental results are shown in table 1:
(1) collecting cells in logarithmic growth phase, regulating the concentration of the cell suspension, and adding 100 mu L of the cell suspension into each well of a 96-well plate; the number of cells per well was about 7000, at 5% CO 2 Incubating overnight at 37 ℃ until the cells are fully adherent;
(2) setting medicine concentration gradients, setting 3 compound holes for each concentration gradient, diluting medicine into corresponding culture medium to required final concentration, sucking out original culture medium in 96-well plate, adding 100 μl of prepared culture medium containing medicine with required final concentration, adding 5% CO 2 Incubating at 37 ℃; and a blank group (containing only 100. Mu.L of culture medium, no cells, and the subsequent treatment is the same as that of other wells) and a control group (containing cells and culture medium) are simultaneously set;
(3) the drug treatment was continued for 4 hours by adding 10. Mu. LMTT solution (5 mg/ml) per well at 44 hours (drug treated cells for 48 hours);
(4) the culture medium in the wells was blotted off (if the cells were suspended, the medium was aspirated after centrifugation at 2500rpm for 5 min). 150 μl of dimethyl sulfoxide was added to each well, and the mixture was shaken until the crystals were sufficiently dissolved. Detecting the absorbance value of each hole at the OD490nm on an enzyme-labeled instrument;
(5) calculating the inhibition rate: inhibition ratio = 1- (dosing OD value-blank OD value)/(control OD value-blank OD value) = (control OD value-dosing OD value)/(control OD value-blank OD value);
(6) repeating the above experimental steps for three times to obtain average value of three inhibition rates, and using IC 50 IC for calculating medicine by calculator 50 Values.
The specific experimental results are shown in table 2 below: the compound of the embodiment has better antiproliferative activity on various tumor cells.
Table 2: anti-cell proliferation Activity (IC) of the Compounds of examples 50 nM)
Figure BDA0003308477660000411
/>
Figure BDA0003308477660000421
/>
Figure BDA0003308477660000431
/>

Claims (7)

1. A phthalazinone compound having a structure represented by formula i, or a pharmaceutically acceptable salt thereof:
Figure FDA0004146582260000011
in formula I:
l is C 2-8 An alkyl group;
e is O or S;
x isSingle bond, vinyl bond, cyclopropyl or-NHCH 2 -;
R is
Figure FDA0004146582260000012
When E is O, X is a single bond, R is
Figure FDA0004146582260000013
Figure FDA0004146582260000021
When E is O, X is a vinyl bond, cyclopropyl, -NH-CH 2 -, R is
Figure FDA0004146582260000022
When E is S, X is-NH-CH 2 -, R is
Figure FDA0004146582260000023
2. The phthalazinone compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein when L is C 2-8 In the case of alkyl, said C 2-8 Alkyl is C 2-6 An alkyl group.
3. The phthalazinone compound of claim 1, or a pharmaceutically acceptable salt thereof, being any one of the following:
Figure FDA0004146582260000024
/>
Figure FDA0004146582260000031
/>
Figure FDA0004146582260000041
/>
Figure FDA0004146582260000051
/>
Figure FDA0004146582260000061
4. a process for the preparation of phthalazinone compounds in accordance with any one of claims 1 to 3, characterised in that: the method comprises the following steps:
Figure FDA0004146582260000062
in a solvent, under the action of alkali and condensing agent, performing condensation reaction on a compound shown in a formula II and a compound shown in a formula III, wherein L, E, X and R are defined as above; the solvent is DMF, the base is DIPEA, and the condensing agent is HATU.
5. A pharmaceutical composition comprising an effective amount of a phthalazinone compound of formula I, or a pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 3, and a pharmaceutically acceptable carrier.
6. The use of a phthalazinone compound of the structure shown in the general formula i or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3 in the preparation of a medicament for preventing and/or treating tumors or NAMPT and PARP1 inhibitor.
7. The use according to claim 6, 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.
CN202111212595.3A 2021-10-18 2021-10-18 Phthalazinone compound, preparation method and medical application thereof Active CN113788827B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111212595.3A CN113788827B (en) 2021-10-18 2021-10-18 Phthalazinone compound, preparation method and medical application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111212595.3A CN113788827B (en) 2021-10-18 2021-10-18 Phthalazinone compound, preparation method and medical application thereof

Publications (2)

Publication Number Publication Date
CN113788827A CN113788827A (en) 2021-12-14
CN113788827B true CN113788827B (en) 2023-04-25

Family

ID=78878159

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111212595.3A Active CN113788827B (en) 2021-10-18 2021-10-18 Phthalazinone compound, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN113788827B (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0305681D0 (en) * 2003-03-12 2003-04-16 Kudos Pharm Ltd Phthalazinone derivatives
US9649394B2 (en) * 2010-11-22 2017-05-16 The General Hospital Corporation Compositions and methods for in vivo imaging
CN102898377B (en) * 2012-02-14 2016-01-20 南京圣和药业股份有限公司 One class phthalazinone derivatives and uses thereof
WO2016025480A1 (en) * 2014-08-11 2016-02-18 The General Hospital Corporation Cyclooctenes for bioorthogonol reactions
CN110194762B (en) * 2018-02-27 2021-09-10 中国科学院上海药物研究所 Phthalazinone derivatives, preparation method and application thereof
US11826430B2 (en) * 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds

Also Published As

Publication number Publication date
CN113788827A (en) 2021-12-14

Similar Documents

Publication Publication Date Title
TWI748539B (en) Cot modulators and methods of use thereof
CN105960405B (en) Glutaminase inhibitors
EP3753937B1 (en) Atr inhibitor and application thereof
TW202144345A (en) Kras mutant protein inhibitors
TWI627174B (en) P2x7 modulators
HU228195B1 (en) Lactam derivatives having a pyrazolo-pyridine ring and pharmaceutical compositions thereof as factor xa inhibitors
BR112017003312B1 (en) INDAZOLE COMPOUNDS AS FGFR KINASE INHIBITORS, PREPARATION AND USE THEREOF
EP3313825A1 (en) Glucose uptake inhibitors
BR112015015477B1 (en) 2,3-DI-HYDRO-ISOINDOL-1-ONA DERIVATIVES AND METHODS OF USING THEM AS BTK INHIBITORS
CN103224496B (en) Tricyclic antidepressants PI3K and/or mTOR inhibitors
AU2014367283B2 (en) Maleimide derivatives as modulators of WNT pathway
EP3845527A1 (en) Multi-substituted pyridone derivatives and medical use thereof
WO2021115401A1 (en) Fluorine-containing heterocyclic derivatives with macrocyclic structure and use thereof
CN108530444A (en) A kind of novel NAMPT and IDO double inhibitors and preparation method thereof and medical usage
JP2016530304A (en) Thienopiperidine derivatives and uses thereof
WO2005058315A1 (en) Novel heterocyclic compounds as ikk2 inhibitors with anti-hbv activity
TW202227443A (en) Tricyclic compound and use thereof
CN113788827B (en) Phthalazinone compound, preparation method and medical application thereof
TWI546304B (en) Protein tyrosine kinase inhibitors and their use
TW202334169A (en) Six-membered cyclothiazole compound and use thereof
CN115417867A (en) Substituted naphthalimide derivative and medical application thereof
WO2022052924A1 (en) Preparation method for class of nitrogen-containing fused ring compounds and use thereof
EP2716639A1 (en) Inhibitors of viral replication, their process of preparation and their therapeutical uses
CN102827160B (en) PI3K or PI3K/m-TOR pathway inhibitor and the purposes in pharmacy thereof
Tang et al. Design, development and evaluation of a prodrug-type TrkA-selective inhibitor with antinociceptive effects in vivo

Legal Events

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