CN115557949A - Tetracyclic derivative, preparation method and medical application thereof - Google Patents

Tetracyclic derivative, preparation method and medical application thereof Download PDF

Info

Publication number
CN115557949A
CN115557949A CN202210319842.8A CN202210319842A CN115557949A CN 115557949 A CN115557949 A CN 115557949A CN 202210319842 A CN202210319842 A CN 202210319842A CN 115557949 A CN115557949 A CN 115557949A
Authority
CN
China
Prior art keywords
alkyl
group
heterocyclyl
aryl
heteroaryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210319842.8A
Other languages
Chinese (zh)
Inventor
陈友喜
程超英
向清
龚亮
毛文涛
和燕玲
叶成
钱文建
陈磊
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.)
Zhejiang Hisun Pharmaceutical Co Ltd
Shanghai Aryl Pharmtech Co Ltd
Original Assignee
Zhejiang Hisun Pharmaceutical Co Ltd
Shanghai Aryl Pharmtech Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zhejiang Hisun Pharmaceutical Co Ltd, Shanghai Aryl Pharmtech Co Ltd filed Critical Zhejiang Hisun Pharmaceutical Co Ltd
Publication of CN115557949A publication Critical patent/CN115557949A/en
Pending legal-status Critical Current

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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

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

Abstract

The invention relates to tetracyclic derivatives, a preparation method thereof and application thereof in medicines. Specifically, the invention relates to tetracyclic derivatives shown in a general formula (I), a preparation method thereof, pharmaceutically acceptable salts thereof, and application thereof as a therapeutic agent, in particular as a K-Ras GTP enzyme inhibitor, wherein the definition of each substituent in the general formula (I) is the same as that in the specification.

Description

Tetracyclic derivative, preparation method and medical application thereof
Technical Field
The invention relates to a tetracyclic derivative, a preparation method thereof, a pharmaceutical composition containing the derivative and application of the derivative as a therapeutic agent, in particular as a K-Ras GTP enzyme inhibitor.
Background
RAS represents a group of closely related monomeric globular proteins (21 kDa molecular weight) of 189 amino acids that are associated with the plasma membrane and bind GDP or GTP. Under normal developmental or physiological conditions, RAS is activated upon receipt of growth factors and various other extracellular signals, and is responsible for regulating functions such as cell growth, survival, migration and differentiation. RAS functions as a molecular switch, with the on/off state of the RAS protein determined by nucleotide binding, the active signaling conformation binding GTP, and the inactive conformation binding GDP. When the RAS comprises bound GDP, it is in a dormant or quiescent or off state and is "inactive". RAS is induced to convert bound GDP to GTP when cells are exposed to certain growth-promoting stimuli in response. With GTP bound, RAS is "on" and is able to interact with and activate other proteins (their "downstream targets"). The RAS protein itself has a very low intrinsic ability to hydrolyze GTP back to GDP and thereby turn itself off. Switching RAS off requires exogenous proteins called Gtpase Activating Proteins (GAPs), which interact with RAS and greatly facilitate conversion of GTP to GDP. Any mutation in the RAS that affects its ability to interact with GAPs or convert GTP back to GDP will result in prolonged activation of the protein and thus produce a prolonged signal to the cell that tells it to continue growing and dividing. These signals can therefore allow cells to grow and divide, and overactive RAS signaling may ultimately lead to cancer.
Structurally, the RAS protein contains a G domain responsible for enzymatic activity of RAS-guanine nucleotide binding and hydrolysis (gtpase reaction). It also includes a C-terminal extension containing a so-called CAAX box, which can be post-translationally modified and targets the protein to the membrane. The G domain is approximately 21-25kDa in size and contains a phosphate binding loop (P-loop). The P-loop represents the capsular bag of bound nucleotides in the protein, and this is a rigid part of the domain with conserved amino acid residues that are essential for nucleotide binding and hydrolysis (glycine 12, threonine 26 and lysine 16). The G domain also contains the so-called switch I region (residues 30-40) and switch II region (residues 60-76), which are both dynamic parts of the protein, often denoted as "spring-loaded" mechanisms due to the ability of the dynamic part to switch between resting and loaded states. The primary interaction is a hydrogen bond formed by threonine-35 and glycine-60 with the gamma-phosphate of GTP, which maintains switch I and switch II regions, respectively, in their active conformations. After hydrolysis of GTP and release of phosphate, both relax into the inactive GDP conformation.
Among RAS family members, oncogenic mutations are most common in KRAS (85%), whereas NRAS (12%) and HRAS (3%) are less common. KRAS mutations are prevalent in three major cancer types in the united states: pancreatic (95%), colorectal (45%), and lung (25%), KRAS mutations were also found in other cancer types including multiple myeloma, uterine, cholangiocarcinoma, gastric, bladder, diffuse large B-cell lymphoma, rhabdomyosarcoma, squamous cell carcinoma of the skin, cervical, testicular germ cell, etc., while rarely found (< 2%) in breast, ovarian, and brain cancers. In non-small cell lung cancer (NSCLC), KRAS G12C is the most common mutation, accounting for nearly half of all KRAS mutations, followed by G12V and G12D. In non-small cell lung cancer, the increase in frequency of specific allelic mutations is mostly due to classical smoking-induced canonical mutations (G: C to T: A substitutions), resulting in KRAS G12C (GGT to TGT) and G12V (GGT to GTT) mutations.
Large genomics studies have shown that lung cancer KRAS mutations, including G12C, are mutually exclusive of other known driver oncogenic mutations in NSCLC, including EGFR, ALK, ROS1, RET, and BRAF, indicating the uniqueness of KRAS mutations in lung cancer. At the same time, KRAS mutations often occur simultaneously with certain co-mutations, such as STK11, KEAP1 and TP53, which cooperate with the mutated RAS to transform cells into highly malignant and aggressive tumor cells.
The three RAS oncogenes constitute the most frequently mutated gene family in human cancers. Disappointingly, despite over thirty years of research efforts, there is still no clinically effective anti-RAS therapy, and the use of small molecules to target this gene is a challenge. Thus, there is an urgent need in the art for small molecules for targeting and utilizing the RAS (e.g., K-RAS, H-RAS and/or N-RAS) to treat a variety of diseases, such as cancer.
At present, the competition of clinical development of KRAS inhibitors is intense at home and abroad, wherein the KRAS enzyme inhibitor MRTX-849 developed by Mirati Therapeutics Inc. enters the second stage of clinic and is used for preventing and treating diseases such as advanced solid tumors, metastatic colorectal cancer, metastatic non-small cell lung cancer and the like. There are also other KRAS inhibitors in development, including AMG-510 (Amgen Inc, phase 3). Early clinical studies show that KRAS inhibitors significantly control and alleviate disease progression in patients with non-small cell lung cancer and significantly reduce tumor size in patients with advanced lung cancer and colorectal cancer. A series of KRAS inhibitor patent applications have been published so far, including WO2020047192, WO2019099524 and WO2018217651, etc., and research and application of KRAS inhibitors have made some progress, but the increased space is still huge, and there is still a need to continue research and development of new KRAS inhibitors.
Disclosure of Invention
The invention aims to provide a tetracyclic derivative shown as a general formula (I), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof:
Figure BDA0003570065910000021
wherein:
Figure BDA0003570065910000022
optionally a single or double bond, such that each atom thereof assumes a normal valence state;
e is selected from
Figure BDA0003570065910000023
Ring A is selected from a 4-to 12-membered heterocyclic group containing at least 2 nitrogen atoms;
x is selected from N or CR c
R a Selected from hydrogen atoms or fluorine;
R b selected from the group consisting of hydrogen atoms, -CH 2 F、-CHF 2
Figure BDA0003570065910000031
R c Selected from hydrogen, halogen, alkyl or alkoxy; wherein said alkyl or alkoxy is optionally further substituted by one or more substituents selected from the group consisting of halogen, hydroxy, cyano, alkyl or alkoxy; r c Preferably halogen, more preferably fluorine or chlorine;
R 1 selected from hydrogen atoms, halogens, alkyl groups or alkoxy groups; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from the group consisting of halo, hydroxy, cyano, alkyl or alkoxy; r 1 Preferably halogen, more preferably chlorine;
R 2 is selected from aryl OR heteroaryl, wherein said aryl OR heteroaryl is optionally further substituted by one OR more groups selected from alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with a substituent of (a); wherein said alkyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted by one OR more substituents selected from the group consisting of alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with the substituent(s);
R 3 selected from absent, hydrogen atom, halogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more R A Substituted; r 3 Preferably absent or a hydrogen atom;
R A each independently selected from alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 (ii) a Wherein said alkyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted by one OR more substituents selected from the group consisting of alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with a substituent of (a);
R 4 identical or different, each independently selected from the group consisting of hydrogen, halogen, hydroxyl, alkyl or alkoxy, R 4 Preferably a hydrogen atom or an alkyl group;
R 5 selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with the substituent(s);
R 6 and R 7 Each independently selected from the group consisting of hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with the substituent(s);
or, R 6 And R 7 Together with the atoms to which they are attached form a 4-to 8-membered heterocyclic group in which the 4-to 8-membered heterocyclic group contains one or more of N, O, S or SO 2 And said 4-to 8-membered heterocyclyl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with the substituent(s);
R 8 、R 9 and R 10 Each independently selected from the group consisting of hydrogen, alkyl, amino, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, amino, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxy or carboxylate;
m is selected from 0,1 or 2;
in a preferred embodiment of the present invention, the tetracyclic derivative represented by the general formula (I), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, is a compound represented by the general formula (II), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof:
Figure BDA0003570065910000041
wherein: ring A, X, R 1 、R 2 、R 4 E and m are as defined in formula (I).
In a preferred embodiment of the present invention, the tetracyclic derivative represented by the general formula (I) or (II), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein E is selected from:
Figure BDA0003570065910000042
in a preferred embodiment of the present invention, the tetracyclic derivative represented by the general formula (I) or (II), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from a 4-to 12-membered heterocyclic group containing 2 nitrogen atoms, preferably a 6-to 8-membered heterocyclic group.
In a preferred embodiment of the present invention, the tetracyclic derivative represented by general formula (I) or (II), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
x is selected from CR c
R c Selected from hydrogen atom, halogen, alkyl and alkaneOxy, haloalkyl or haloalkoxy; r c Preferably halogen, more preferably fluorine or chlorine.
In a preferred embodiment of the present invention, the tetracyclic derivative represented by the general formula (I) or (II), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, wherein R is 1 Selected from a hydrogen atom, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy; r is 1 Preferably halogen, more preferably chlorine;
in a preferred embodiment of the present invention, the tetracyclic derivative represented by the general formula (I) or (II), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof is provided, wherein R is 2 Selected from the group consisting of:
Figure BDA0003570065910000051
in a preferred embodiment of the present invention, the tetracyclic derivative represented by general formula (I) or (II), or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein: r is 4 Selected from a hydrogen atom or an alkyl group; wherein said alkyl group is preferably methyl.
Typical compounds of the invention include, but are not limited to:
Figure BDA0003570065910000052
Figure BDA0003570065910000061
Figure BDA0003570065910000071
or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
Note: if there is a difference between the drawn structure and the name given for that structure, the drawn structure will be given more weight.
The present invention provides a process for the preparation of a compound of general formula (I) according to claim 1 or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, which process comprises:
Figure BDA0003570065910000081
reacting the compound of the general formula (IA) with the compound of the general formula (IB) under alkaline conditions, and optionally further removing a protecting group to obtain a compound of the general formula (I);
wherein:
X 1 is a leaving group, preferably chloro;
ring A, R 1 ~R 4 X, E and m are as defined in formula (I).
The invention provides a compound shown in a general formula (IA) or a stereoisomer, a tautomer and a pharmaceutically acceptable salt thereof,
Figure BDA0003570065910000082
wherein: ring A, R 1 ~R 4 X and m are as defined in formula (I).
Typical compounds of formula (IA) include, but are not limited to:
Figure BDA0003570065910000083
Figure BDA0003570065910000091
Figure BDA0003570065910000101
or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
In another aspect, the present invention provides a pharmaceutical composition comprising an effective amount of a compound of formula (I) or (II) or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
In another aspect, the present invention provides a method of inhibiting K-Ras GTPase, wherein said method comprises administering to a patient a pharmaceutical composition comprising an effective amount of a compound of formula (I) or (II), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof, wherein the K-Ras GTPase is preferably KRAS G12C enzyme.
The invention also provides a compound of the general formula (I) or (II) or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, wherein the KRAS mutation-mediated disease is selected from cancers, wherein the cancers are selected from pancreatic cancer, colorectal cancer, lung cancer, multiple myeloma, uterine cancer, bile duct cancer, gastric cancer, bladder cancer, diffuse large B-cell lymphoma, rhabdomyosarcoma, squamous cell carcinoma of skin, cervical cancer, testicular germ cell carcinoma, preferably pancreatic cancer, colorectal cancer and lung cancer; wherein said lung cancer is preferably non-small cell lung cancer, and wherein said KRAS mutation is preferably a KRAS G12C mutation.
In another aspect, the present invention provides the use of a compound of formula (I) or (II) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or pharmaceutical composition thereof, for the preparation of a K-Ras GTPase inhibitor, wherein the K-Ras GTPase inhibitor is preferably a KRAS G12C inhibitor.
Another aspect of the present invention relates to a method for preventing and/or treating a KRAS mutation-mediated disease, comprising administering to a patient a therapeutically effective amount of a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer or mixture thereof, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, wherein the KRAS mutation is preferably a KRAS G12C mutation.
The invention also provides an application of the compound shown in the general formula (I) or (II) or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof in preparing a medicament for treating cancers, wherein the cancers are selected from pancreatic cancer, colorectal cancer, lung cancer, multiple myeloma, uterine cancer, bile duct cancer, gastric cancer, bladder cancer, diffuse large B-cell lymphoma, rhabdomyosarcoma, skin squamous cell carcinoma, cervical cancer and testicular germ cell cancer, preferably pancreatic cancer, colorectal cancer and lung cancer; wherein the lung cancer is preferably non-small cell lung cancer.
The pharmaceutical formulations of the present invention may be administered topically, orally, transdermally, rectally, vaginally, parenterally, intranasally, intrapulmonary, intraocularly, intravenously, intramuscularly, intraarterially, intrathecally, intracapsularly, intradermally, intraperitoneally, subcutaneously, subcortically, or by inhalation. The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
The formulations of the present invention are suitably presented in unit dosage form and may be prepared by any of the methods well known in the pharmaceutical art. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form generally refers to the amount of compound that produces a therapeutic effect.
Dosage forms for topical or transdermal administration of the compounds of the present invention may include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
When the compounds of the present invention are administered to humans and animals in the form of drugs, the compounds may be provided alone or in the form of pharmaceutical compositions containing the active ingredient in combination with a pharmaceutically acceptable carrier, e.g., 0.1% to 99.5% (more preferably, 0.5% to 90%) of the active ingredient.
Examples of pharmaceutically acceptable carriers include, but are not limited to: (1) sugars such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) Cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered gum tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients such as cocoa butter and suppository waxes; (9) Oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) Polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution (Ringer's solution); (19) ethanol; (20) phosphate buffer solution; (21) Cyclodextrins, e.g., targeting ligands attached to the nanoparticle, e.g., accurins (tm); and (22) other non-toxic compatible materials used in pharmaceutical formulations, such as polymer-based compositions.
Examples of pharmaceutically acceptable antioxidants include, but are not limited to: (1) Water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) Oil-soluble antioxidants, such as ascorbyl palmitate, butylated Hydroxyanisole (BHA), butylated Hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents such as citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Solid dosage forms (e.g., capsules, dragee pills, dragees, powders, granules, and the like) can include one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) Fillers or extenders, such as starch, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) Binding agents, such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) Disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; (5) dissolution retarders, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) Humectants, such as cetyl alcohol and glycerol monostearate; (8) absorbents such as kaolin and bentonite clay; (9) Lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof; and (10) a colorant. Liquid dosage forms may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents; solubilizers and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum oxyhydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Ointments, pastes, creams and gels may also contain, in addition to the active compound, excipients, for example animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can also contain, in addition to the active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances. The spray may contain other conventional propellants such as chlorofluorohydrocarbons, and volatile unsubstituted hydrocarbons such as butane and propane.
Detailed description of the invention
Unless stated to the contrary, some of the terms used in the specification and claims of the present invention are defined as follows:
"chemical bond" means that the indicated substituent is absent and the two end portions of the substituent are directly connected to form a bond.
"alkyl" when taken as a group or part of a group means including C 1 -C 20 Straight-chain or branched aliphatic hydrocarbon groups. Preferably C 1 -C 10 Alkyl, more preferably C 1 -C 6 An alkyl group. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be substituted or unsubstituted.
"alkenyl" means an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, representative examples include but are not limited to ethenyl, 1-propenyl, 2-propenyl, 1-, 2-or 3-butenyl, and the like. The alkenyl group may be optionally substituted or unsubstituted.
"alkynyl" refers to an aliphatic hydrocarbon group containing a carbon-carbon triple bond, and can be straight or branched. Preferred is that C 2 -C 10 Alkynyl of (2), more preferably C 2 -C 6 Alkynyl, most preferably C 2 -C 4 Alkynyl. Examples of alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 1-,2-, or 3-butynyl, and the like. Alkynyl may be substituted bySubstituted or unsubstituted.
An "alkylene" is a divalent alkyl group. Preferably C 1 -C 10 Alkylene, more preferably C1-C6 alkylene, particularly preferably C 1 -C 4 An alkylene group. Examples of alkylene groups include, but are not limited to, methylene, ethylene, -CH (CH) 3 ) 2 N-propylene, and the like. The alkylene group may be substituted or unsubstituted.
"cycloalkyl" refers to saturated or partially saturated monocyclic, fused, bridged, and spiro carbocyclic rings. Preferably C 3 -C 12 Cycloalkyl, more preferably C 3 -C 8 Cycloalkyl, most preferably C 3 -C 6 A cycloalkyl group. Examples of monocyclic cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like, with cyclopropyl, cyclohexenyl being preferred. Cycloalkyl groups may be optionally substituted or unsubstituted.
"spirocycloalkyl" refers to a 5-to 18-membered polycyclic group having two or more cyclic structures with single rings sharing one carbon atom (called the spiro atom) with each other, containing 1 or more double bonds within the ring, but with no ring having a fully conjugated pi-electron aromatic system. Preferably 6 to 14, more preferably 7 to 10. Spirocycloalkyl groups are classified according to the number of spiro atoms shared between rings into mono-spiro, di-spiro, or multi-spiro cycloalkyl groups, preferably mono-spiro and di-spiro cycloalkyl groups, preferably 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered. Non-limiting examples of "spirocycloalkyl" include, but are not limited to: spiro [4.5] decyl, spiro [4.4] nonyl, spiro [3.5] nonyl, spiro [2.4] heptyl.
"fused cyclic alkyl" refers to a 5 to 18 membered all carbon polycyclic group containing two or more cyclic structures sharing a pair of carbon atoms with each other, one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system, preferably 6 to 12, more preferably 7 to 10. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyls according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-or 6-membered bicycloalkyl. Non-limiting examples of "fused ring alkyl" include, but are not limited to: bicyclo [3.1.0] hexyl, bicyclo [3.2.0] hept-1-enyl, bicyclo [3.2.0] heptyl, decalinyl or tetradecaphenanthryl.
"bridged cycloalkyl" means a 5 to 18 membered all carbon polycyclic group containing two or more cyclic structures sharing two non-directly attached carbon atoms with each other, one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system, preferably 6 to 12, more preferably 7 to 10. Preferably 6 to 14, more preferably 7 to 10. They may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic, depending on the number of constituent rings. Non-limiting examples of "bridged cycloalkyl" groups include, but are not limited to: (1s, 4s) -bicyclo [2.2.1] heptyl, bicyclo [3.2.1] octyl, (1s, 5s) -bicyclo [3.3.1] nonyl, bicyclo [2.2.2] octyl, and (1r, 5r) -bicyclo [3.3.2] decyl.
"Heterocyclyl," "heterocycle," or "heterocyclic" are used interchangeably herein and all refer to non-aromatic heterocyclic groups in which one or more of the ring-forming atoms is a heteroatom, such as oxygen, nitrogen, sulfur, and the like, including monocyclic, fused, bridged, and spiro rings. Preferably having a 5 to 7 membered monocyclic ring or a 7 to 10 membered bi-or tricyclic ring, which may contain 1,2 or 3 atoms selected from nitrogen, oxygen and/or sulfur. Examples of "heterocyclyl" include, but are not limited to, morpholinyl, oxetanyl, thiomorpholinyl, tetrahydropyranyl, 1-dioxothiomorpholinyl, piperidinyl, 2-oxopiperidinyl, pyrrolidinyl, 2-oxopyrrolidinyl, piperazin-2-one, 8-oxa-3-aza-bicyclo [3.2.1] octyl, and piperazinyl. The heterocyclic group may be substituted or unsubstituted.
"spiroheterocyclyl" refers to a 5 to 18 membered polycyclic group having two or more cyclic structures wherein the individual rings share an atom with one another and wherein 1 or more double bonds are present in the ring, but none of the rings has a fully conjugated pi-electron aromatic system wherein one or more of the ring atoms is selected from nitrogen, oxygen or S (O) r (wherein r is selected from 0,1 or 2) and the remaining ring atoms are carbon. Preferably 6 to 14, more preferablyIs 7 to 10 yuan. The spirocycloalkyl group is classified into a single spiroheterocyclic group, a double spiroheterocyclic group or a multiple spiroheterocyclic group, preferably a single spiroheterocyclic group and a double spiroheterocyclic group, according to the number of spiro atoms shared between rings. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospiroheterocyclyl group. Non-limiting examples of "spiroheterocyclyl" include, but are not limited to: 1, 7-dioxaspiro [4.5]]Decyl, 2-oxa-7-azaspiro [4.4]Nonyl, 7-oxaspiro [3.5]]Nonyl and 5-oxaspiro [2.4]]A heptyl group.
"fused heterocyclyl" refers to an all-carbon polycyclic group containing two or more cyclic structures sharing a pair of atoms with each other, one or more of which rings may contain one or more double bonds, but none of which rings has a fully conjugated pi-electron aromatic system wherein one or more of the ring atoms is selected from nitrogen, oxygen, or S (O) r (wherein r is selected from 0,1 or 2) and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-or 6-membered bicyclic fused heterocyclic groups. Non-limiting examples of "fused heterocyclic groups" include, but are not limited to: octahydropyrrolo [3,4-c ] s]Pyrrolyl, octahydro-1H-isoindolyl, 3-azabicyclo [3.1.0]Hexyl, octahydrobenzo [ b ]][1,4]Dioxins (dioxines).
"bridged heterocyclyl" means a 5 to 14 membered, 5 to 18 membered polycyclic group containing two or more cyclic structures sharing two atoms not directly attached to each other, one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron aromatic system wherein one or more ring atoms is selected from nitrogen, oxygen or S (O) r (wherein r is selected from 0,1 or 2) and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclic groups according to the number of constituent rings, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged heterocyclic groups" include, but are not limited to: 2-azabicyclo [2.2.1]Heptyl, 2-azabicyclo [2.2.2]Octyl and 2-azabicyclo [3.3.2]A decyl group.
"aryl" refers to a carbocyclic aromatic system containing one or two rings, wherein the rings may be joined together in a fused fashion. The term "aryl" includes monocyclic or bicyclic aryl groups such as phenyl, naphthyl, tetrahydronaphthyl aromatic groups. Preferably aryl is C 6 -C 10 Aryl, more preferably aryl is phenyl and naphthyl. The aryl group may be substituted or unsubstituted.
"heteroaryl" refers to an aromatic 5-to 6-membered monocyclic or 8-to 10-membered bicyclic ring, which can contain 1 to 4 atoms selected from nitrogen, oxygen and/or sulfur. <xnotran> , " " , ,2- -1,2- , , , , , , , , , , , , , , ,1,2,3- , , , , , ,1,3- - , , , , , , </xnotran>
Figure BDA0003570065910000151
Heteroaryl groups may be substituted or unsubstituted.
"fused ring" refers to a polycyclic group in which two or more cyclic structures share a pair of atoms with each other, one or more of the rings may contain one or more double bonds, but at least one of the rings does not have a completely conjugated pi-electron aromatic system, wherein the ring atoms are selected from 0, one or more selected from nitrogen, oxygen, or S (O) r (wherein r is selected from 0,1 or 2) and the remaining ring atoms are carbon. The fused ring preferably includes a bicyclic or tricyclic fused ring, wherein the bicyclic fused ring is preferably a fused ring of an aryl or heteroaryl group with a monocyclic heterocyclic group or a monocyclic cycloalkyl group. Preferably 7 to 14, more preferably 8 to 10. Examples of "fused rings" include, but are not limited to:
Figure BDA0003570065910000161
"alkoxy" refers to a radical of (alkyl-O-). Wherein alkyl is as defined herein. C 1 -C 6 Alkoxy groups of (2) are preferred. Examples thereof include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy and the like.
"haloalkyl" refers to an alkyl group optionally further substituted with one or more halogens, wherein alkyl is as defined herein.
"deuterated alkyl" refers to an alkyl group optionally further substituted with one or more deuterium atoms, wherein alkyl is as defined herein. "deuterated alkyl" is preferably deuterated methyl and includes: mono-, di-and tri-deuterated methyl, preferably tri-deuterated methyl.
"hydroxyalkyl" refers to a group in which the alkyl group is optionally further substituted with one or more hydroxyl groups, wherein alkyl is as defined herein.
"haloalkoxy" refers to a group in which the alkyl group of (alkyl-O-) is optionally further substituted with one or more halogens, wherein alkoxy is as defined herein.
"hydroxy" refers to an-OH group.
"halogen" refers to fluorine, chlorine, bromine and iodine.
"amino" means-NH 2
"cyano" means-CN.
"nitro" means-NO 2
"benzyl" means-CH 2 -phenyl.
"carboxy" refers to-C (O) OH.
"carboxylate" means-C (O) O-alkyl or-C (O) O-cycloalkyl, wherein alkyl and cycloalkyl are as defined above.
"DMSO" refers to dimethyl sulfoxide.
"BOC" refers to tert-butoxycarbonyl.
"Ts" refers to p-toluenesulfonyl.
"T3P" refers to propylphosphoric anhydride.
"DPPA" refers to diphenylphosphoryl azide.
"DEA" refers to diethylamine.
"TFA" refers to trifluoroacetic acid.
“CaCl 2 "refers to calcium chloride.
“MgCl 2 "refers to magnesium chloride.
"KCl" refers to potassium chloride.
"NaCl" refers to sodium chloride.
"Glucose" refers to Glucose.
"HEPES" means N-2-hydroxyethylpiperazine-N' -2-ethanesulfonic acid.
"EGTA" refers to ethylene glycol bis (2-aminoethyl ether) tetraacetic acid.
"substituted" means that one or more, preferably up to 5, more preferably 1 to 3, hydrogen atoms in a group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
As used herein, "substituted" or "substituted," unless otherwise specified, means that the group may be substituted with one or more groups selected from: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxy, carboxylate, = O, -C (O) R 7 、-C(O)OR 7 、-NHC(O)R 7 、-NHC(O)OR 7 、-NR 8 R 9 、-C(O)NR 8 R 9 、-CH 2 NHC(O)OR 7 、-CH 2 NR 8 R 9 or-S (O) r R 7 Substituted with a substituent of (a);
R 7 selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 10 、-C(O)OR 10 、-OC(O)R 10 、-NR 11 R 12 、-C(O)NR 11 R 12 、-SO 2 NR 11 R 12 or-NR 11 C(O)R 12 Substituted with the substituent(s);
R 8 and R 9 Each independently selected from the group consisting of hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 10 、-C(O)OR 10 、-OC(O)R 10 、-NR 11 R 12 、-C(O)NR 11 R 12 、-SO 2 NR 11 R 12 or-NR 11 C(O)R 12 Substituted with the substituent(s);
or, R 8 And R 9 Together with the atoms to which they are attached form a 4-to 8-membered heterocyclic group in which the 4-to 8-membered heterocyclic group contains one or more of N, O or S (O) r And said 4-to 8-membered heterocyclyl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 10 、-C(O)OR 10 、-OC(O)R 10 、-NR 11 R 12 、-C(O)NR 11 R 12 、-SO 2 NR 11 R 12 or-NR 11 C(O)R 12 Substituted with a substituent of (a);
R 10 、R 11 and R 12 Each independently selected from hydrogen, alkyl, amino, cycloalkyl, heterocyclyl, aryl or heteroAryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, amino, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxy or carboxylate;
r is 0,1 or 2.
The compounds of the invention may contain asymmetric or chiral centers and thus exist in different stereoisomeric forms. It is contemplated that all stereoisomeric forms of the compounds of the present invention, including but not limited to diastereomers, enantiomers and atropisomers (atropisomers) and geometric (conformational) isomers and mixtures thereof, such as racemic mixtures, are within the scope of the present invention.
Unless otherwise indicated, all isomers (e.g., diastereomers, enantiomers, and atropisomers and geometric (conformational) isomeric forms; e.g., R and S configurations at each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers) of the structures are also encompassed by the structures described herein.
"pharmaceutically acceptable salts" refers to certain salts of the above compounds which retain their biological activity and are suitable for pharmaceutical use. The pharmaceutically acceptable salts of the compounds of formula (I) may be metal salts, amine salts with suitable acids.
"pharmaceutical composition" means a mixture containing one or more compounds described herein, or a physiologically acceptable salt or prodrug thereof, in admixture with other chemical components, as well as other components such as physiologically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
Synthesis of the Compounds of the invention
In order to achieve the purpose of the invention, the invention adopts the following technical scheme:
the invention relates to a preparation method of a compound shown in a general formula (I) or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, which comprises the following steps:
Figure BDA0003570065910000181
reacting a compound of general formula (IA) with a compound of general formula (IB) under basic conditions, optionally further deprotecting to give a compound of general formula (I);
wherein:
X 1 is a leaving group, preferably chloro;
ring A, R 1 ~R 4 X, E and m are as defined in formula (I).
Detailed Description
The present invention will be further described with reference to the following examples, which are not intended to limit the scope of the present invention.
Examples
The examples show the preparation of representative compounds represented by formula (I) and the associated structural identification data. It must be noted that the following examples are intended to illustrate the invention and not to limit it. 1 HNMR spectra were obtained using a Bruker instrument (400 MHz) and chemical shifts are expressed in ppm. Tetramethylsilane internal standard (0.00 ppm) was used. 1 Representation method of HNMR: s = singlet, d = doublet, t = triplet, m = multiplet, br = broadened, dd = doublet of doublet, dt = doublet of triplet. If a coupling constant is provided, it is in Hz.
The mass spectrum is measured by an LC/MS instrument, and the ionization mode can be ESI or APCI.
Thin layer chromatography silica gel plate is prepared from HSGF254 of tobacco yellow sea or GF254 of Qingdao by Thin Layer Chromatography (TLC)
The specification of the silica gel plate is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
The column chromatography generally uses 200-300 mesh silica gel of the Tibet Huanghai silica gel as a carrier.
In the following examples, unless otherwise indicated, all temperatures are in degrees celsius and unless otherwise indicated, the starting materials and reagents are commercially available or synthesized according to known methods, and none of the commercially available materials or reagents are used without further purification, unless otherwise indicated, and commercially available manufacturers include, but are not limited to, shanghai haohnhong reagent limited, shanghai beth medicine technology limited, shanghai kaider chemical technology (shanghai) limited, and shanghai ling kaiki medicine technology limited.
CD 3 OD: deuterated methanol.
CDCl 3 : deuterated chloroform.
DMSO-d 6 : deuterated dimethyl sulfoxide.
In the examples, the solution in the reaction is an aqueous solution unless otherwise specified.
Purifying the compound using an eluent system selected from the group consisting of column chromatography and thin layer chromatography, wherein the system is selected from the group consisting of: a: petroleum ether and ethyl acetate systems; b: dichloromethane and methanol systems; c: dichloromethane and ethyl acetate system, D: dichloromethane and ethanol system, E: tetrahydrofuran/petroleum ether system, F: tetrahydrofuran and methanol, wherein the volume ratio of the solvent is different according to the polarity of the compound, or a small amount of acidic or basic reagent such as acetic acid or triethylamine can be added for carrying out the conditions.
Example 1
1-(3-(2-amino-5,7-difluorobenzo[d]thiazol-4-yl)-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinolin-9(8H)-yl)prop-2-en-1-one
1- (3- (2-amino-5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-9 (8H) -yl) prop-2-en-1-one
Figure BDA0003570065910000201
First step of
tert-butyl tert-benzoate (2- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) carbamate
7-bromo-4,6-dichloro-8-fluoro-3-nitroquinoline 1a (500mg, 1.47mmol, prepared according to the method disclosed in patent WO 2019110751A 1) and tert-butyl (2-aminoethyl) carbamate 1b (282.79mg, 1.77mmol) were added to acetonitrile (8.54 mL), cooled to 0 deg.C, N-diisopropylethylamine (570.29mg, 4.41mmol, 729.27. Mu.L) was added, and the mixture was allowed to warm to room temperature for overnight. The reaction mixture was extracted with ethyl acetate (100 mL. Times.1), the organic phase was washed with saturated brine (100 mL. Times.1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give (2- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) carbamic acid tert-butyl ester 1c (680 mg, 1.47mmol), yield 99.70%, and the product was directly subjected to the next reaction without purification.
LCMS:463.0[M+1] +
Second step of
tert-butyl tert-benzoate (2- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) carbamate
Tert-butyl (2- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) carbamate 1c (680 mg, 1.47mmol), iron powder (409.52mg, 7.33mmol), and ammonium chloride (392.22mg, 7.33mmol) were added to a mixed solvent of methanol (10 mL) and water (2 mL), and heated under reflux for 4 hours. The reaction solution was filtered while hot, the filter cake was washed with methanol (20 mL), the filtrate was extracted with ethyl acetate (100 mL × 1) after removing methanol by rotation, the organic phase was washed with saturated brine (100 mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give (2- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) carbamic acid tert-butyl ester 1d (636 mg, 1.47mmol) in a yield of 100.00%, which was directly subjected to the next reaction without purification.
LCMS:433.0[M+1] +
The third step
tert-butyl(2-(7-bromo-8-chloro-2-(chloromethyl)-6-fluoro-1H-imidazo[4,5-c]quinolin-1-yl)ethyl)carbamate
(2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethyl) carbamic acid tert-butyl ester
Tert-butyl (2- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) carbamate 1d (300mg, 691.72 μmol) was added to dichloromethane (5 mL), triethylamine (139.99mg, 1.38mmol) was added, a dichloromethane solution (2 mL) of 2-chloroacetyl chloride (156.25mg, 1.38mmol) was added dropwise at room temperature, the reaction was continued overnight at room temperature, many intermediates were detected by LCMS without regard to rings, the reaction solution was extracted with dichloromethane (50 mL × 1), the organic phase was washed with saturated brine (100 mL × 2), dried over anhydrous sodium sulfate and concentrated, the resulting residue was added to acetic acid (5 mL), heated to 50 ℃ for 2 hours, the reaction solution was cooled to room temperature, the system was made basic with a saturated sodium carbonate solution, extracted with ethyl acetate (100 mL × 1), the organic phase was washed with saturated brine (100 mL × 1), dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain a residue, which was subjected to silica gel chromatography (eluent: system a) was separated and purified to give the product tert-butyl 2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethyl) carbamate 1e (190mg, 386.05 μmol), yield 57.58%.
LCMS:492.7[M+1] +
The fourth step
2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethan-1-amine
Tert-butyl (2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethyl) carbamate 1e (190mg, 386.05. Mu. Mol) was added to dichloromethane (4 mL), and a dioxane solution of hydrogen chloride (4M, 2mL) was added and reacted at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give 2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethan-1-amine 1f (151mg, 385.15. Mu. Mol), in 99.77% yield, which was directly subjected to the next reaction without purification.
LCMS:393.0[M+1] +
The fifth step
3-bromo-2-chloro-4-fluoro-8,9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinone 3-bromo-2-chloro-4-fluoro-8,9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline
2- (7-bromo-8-chloro-2- (chloromethyl) -6-fluoro-1H-imidazo [4,5-c ] quinolin-1-yl) ethan-1-amine 1f (151mg, 385.15. Mu. Mol), potassium iodide (6.39mg, 38.52. Mu. Mol) and potassium carbonate (106.46mg, 770.30. Mu. Mol) were added to acetonitrile (5 mL) and heated under reflux for 3 hours. The reaction mixture was cooled to room temperature, extracted with ethyl acetate (100 mL. Times.1), and the organic phase was washed with saturated brine (100 mL. Times.1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 1g (136mg, 382.46. Mu. Mol) of 3-bromo-2-chloro-4-fluoro-8,9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline in 99.30% yield, which was directly subjected to the next reaction without purification.
LCMS:355.0[M+1] +
The sixth step
tert-butyl 3-bromo-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinoline-9(8H)-carboxylate
3-bromo-2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylic acid tert-butyl ester
3-bromo-2-chloro-4-fluoro-8,9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline 1g (136mg, 382.46. Mu. Mol), triethylamine (77.40mg, 764.92. Mu. Mol) and di-tert-butyl dicarbonate (100.17mg, 458.95. Mu. Mol) were added to dichloromethane (5 mL) and reacted at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was separated and purified by silica gel column chromatography (eluent: system A) to give the product, tert-butyl 3-bromo-2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylate, in 1H (140mg, 307.21. Mu. Mol), 80.33% yield.
LCMS:456.8[M+1] +
Step seven
tert-butyl 3-(2-((tert-butoxycarbonyl)amino)-5,7-difluorobenzo[d]thiazol-4-yl)-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinoline-9(8H)-carboxylate
3- (2- ((tert-Butoxycarbonyl) amino) -5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylic acid tert-butyl ester
1i (152.12mg, 460.82. Mu. Mol, prepared according to published patent U.S. Pat. No. 5,00115375A 1), 3-bromo-2-chloro-4-fluoro-10,11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylic acid tert-butyl ester 1H (140mg, 307.21. Mu. Mol), sodium carbonate (65.13mg, 614.43. Mu. Mol) and tetratriphenylphosphine palladium (35.50mg, 30.72. Mu. Mol) were added to a mixed solution of dioxane (4 mL) and water (1 mL), argon protected, and heated to 110 ℃ for overnight reaction. The reaction solution was cooled, extracted with ethyl acetate (100 mL. Times.1), the organic phase was washed with saturated brine (100 mL. Times.1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure, and the obtained residue was purified by preparative liquid phase separation (separation column AKZONOBEL Kromasil; 250X 21.2mm I.D.; 5. Mu.m, 20mL/min; mobile phase A:0.05% TFA H2O; mobile phase B: CH3 CN) to give the product, tert-butyl 3- (2- ((tert-butoxycarbonyl) amino) -5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylate 1j (100mg, 151.26. Mu. Mol), yield 49.24%.
LCMS:660.8[M+1] +
Eighth step
4-(2-chloro-4-fluoro-8,9,10,11-tetrahydropyrazino[1',2':1,2]imidazo[4,5-c]quinolin-3-yl)-5,7-difluorobenzo[d]thiazol-2-amine
4- (2-chloro-4-fluoro-8, 9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-3-yl) -5, 7-difluorobenzo [ d ] thiazol-2-amine
3- (2- ((tert-Butoxycarbonyl) amino) -5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinoline-9 (8H) -carboxylic acid tert-butyl ester 1j (100mg, 151.26. Mu. Mol) was added to dichloromethane (5 mL), a solution of hydrogen chloride in 1, 4-dioxane (4M, 3 mL) was added and the reaction was carried out at room temperature for 3 hours. The reaction mixture was concentrated under reduced pressure, the system was made alkaline with a saturated sodium carbonate solution, extracted with ethyl acetate (100 mL. Times.1), the organic phase was washed with saturated brine (100 mL. Times.1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the product 4- (2-chloro-4-fluoro-8, 9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-3-yl) -5, 7-difluorobenzo [ d ] thiazol-2-amine 1k (45mg, 88.66. Mu. Mol), yield 58.61%.
LCMS:461.0[M+1]
The ninth step
1-(3-(2-amino-5,7-difluorobenzo[d]thiazol-4-yl)-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinolin-9(8H)-yl)prop-2-en-1-one
1- (3- (2-amino-5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-9 (8H) -yl) prop-2-en-1-one
4- (2-chloro-4-fluoro-8, 9,10,11-tetrahydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-3-yl) -5, 7-difluorobenzo [ d ] thiazol-2-amine 1k (45mg, 97.64. Mu. Mol) was added to dichloromethane (5 mL), triethylamine (29.64mg, 292.93. Mu. Mol) was added, cooled to 0 ℃, acryloyl chloride (10.60mg, 117.17. Mu. Mol) was added dropwise, and the mixture was allowed to warm to room temperature for 2h. Dichloromethane extraction (100 mL. Times.1), washing of the organic phase with saturated brine (100 mL. Times.1), drying over anhydrous sodium sulfate, and purification by column chromatography on silica gel (eluent: B system) to give the product 1- (3- (2-amino-5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-9 (8H) -yl) prop-2-en-1-one 1 (30mg, 52.44. Mu. Mol), 53.70% yield.
LCMS:517.0[M+1]+
Example 2 and example 3
(R)-1-(3-(2-amino-5,7-difluorobenzo[d]thiazol-4-yl)-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinolin-9(8H)-yl)prop-2-en-1-one
(S)-1-(3-(2-amino-5,7-difluorobenzo[d]thiazol-4-yl)-2-chloro-4-fluoro-10,11-dihydropyrazino[1',2':1,2]imidazo[4,5-c]quinolin-9(8H)-yl)prop-2-en-1-one
(R) -1- (3- (2-amino-5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-9 (8H) -yl) prop-2-en-1-one 2
(S) -1- (3- (2-amino-5, 7-difluorobenzo [ d ] thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2] imidazo [4,5-c ] quinolin-9 (8H) -yl) prop-2-en-1-one 3
Figure BDA0003570065910000241
1- (3- (2-amino-5, 7-difluorobenzo [ d ]]Thiazol-4-yl) -2-chloro-4-fluoro-10, 11-dihydropyrazino [1',2':1,2]Imidazo [4, 5-c)]Quinolin-9 (8H) -yl) prop-2-en-1-one 1 (26mg, 45.53mmol) was resolved by SFC chiral resolution (column No.: chiralPak AD,250 × 30mm i.d.,10 μm; mobile phase: a for CO 2 and B for Isopropanol (0.1% NH3H2O); column pressure: 100bar; flow rate: 80mL/min; detection wavelength: 220nm; column temperature: after purification at 38 ℃ C.), a single configuration compound (shorter retention time) and a single configuration compound (longer retention time) are obtained.
Single configuration compound (shorter retention time):
MS m/z(ESI):515.0[M+1] +
13mg; retention time 1.690 min, chiral purity 100% ee.
Single configuration compound (longer retention time):
MS m/z(ESI):515.0[M+1] +
10mg; retention time 2.028 min, chiral purity 99.26% ee.
Biological evaluation
Test example 1 determination of the ability of the Compounds of the invention to covalently bind to KRAS G12C protein
The following method was used to determine the ability of the compounds of the invention to covalently bind to recombinant human KRAS G12C protein under in vitro conditions.
The experimental procedure is briefly described as follows: reaction buffers (20mM HEPES,150mM NaCl,1mM MgCl) were used 2 1mM DTT) was prepared at a concentration of 4. Mu.M for use as recombinant human KRAS G12C protein (aa 1-169). Test compounds were prepared as 10mM stock solutions dissolved in DMSO and subsequently diluted with reaction buffer for use. First, 1.5. Mu.L of the test compound diluted with the reaction buffer (final concentration of the reaction system is 3. Mu.M) was added to the well, then 23.5. Mu.L of the reaction buffer was added and mixed, then 25. Mu.L of 4. Mu.M recombinant human KRAS G12C protein was added, after incubation at room temperature for 5 minutes, 5. Mu.L of acetic acid was added to terminate the reaction, and the sample was transferred to a sample bottle. Detection of the receptor by Agilent 1290/6530 InstrumentThe ratio of covalent binding of the test compound to KRAS G12C Protein, the sample was purified on a liquid chromatography column (XBridge Protein BEH C4,
Figure BDA0003570065910000242
3.5 μm,2.1mm × 50 mm), mobile phase a is 0.1% formic acid in water, mobile phase B is acetonitrile, mobile phase elution procedure is: 0-0.5 min, keeping mobile phase A: at 95%, at 2.5 minutes, mobile phase a became 30% and held for 0.5 minutes, 3.1 minutes, mobile phase a became 95% and held for 1.9 minutes; flow rate: 0.5mL/min; finally, the data were analyzed by using MassHunter Workstation Software bioconjugate Version B.08.00 Software, the concentration of the test compound was 3. Mu.M, and the covalent Binding Rate (Binding Rate) of the KRAS G12C protein under the incubation condition of 5min is shown in Table 1.
TABLE 1 covalent binding Rate of Compounds of the invention to KRAS G12C protein
Figure BDA0003570065910000243
Figure BDA0003570065910000251
The conclusion is that the compound has better covalent binding rate with KRAS G12C protein.
Test example 2 assay for inhibition of NCI-H358 cell proliferation by Compounds of the present invention
The following method was used to determine the effect of the compounds of the invention on NCI-H358 cell proliferation. NCI-H358 cells (containing KRAS G12C mutation) were purchased from the cell resource center of Shanghai Life sciences institute of Chinese academy of sciences, and cultured in RPMI 1640 medium containing 10% fetal bovine serum, 100U penicillin, 100. Mu.g/mL streptomycin and 1mM Sodium Pyruvate. Cell viability by
Figure BDA0003570065910000252
The Luminescent Cell Viability Assay kit (Promega, cat # G7573) was used for the Assay.
Experimental methodsThe procedures according to the kit instructions are as follows: test compounds were first prepared as 10mM stock solutions dissolved in DMSO and then diluted in culture medium to prepare test samples with compound concentrations ranging from 1000nM to 0.015nM. Cells in logarithmic growth phase were seeded at a density of 800 cells per well in 96-well cell culture plates and at 37 ℃,5% CO 2 The incubation was carried out overnight in an incubator, followed by addition of test compounds and further incubation for 120 hours. After the incubation was completed, 50. Mu.L of CellTiter-Glo detection solution was added to each well, shaken for 5 minutes and then allowed to stand for 10 minutes, and then Luminescence values of each well of the sample were read on a microplate reader using a Luminescence mode. The percentage inhibition of the compounds at each concentration point was calculated by comparison with the value of the control group (0.3% DMSO), after which non-linear regression analysis was performed in GraphPad Prism 5 software at the compound concentration log-inhibition to obtain the IC of the compounds inhibiting cell proliferation 50 The value is obtained.
The conclusion is that the compound of the invention has better proliferation inhibition effect on NCI-H358 (human non-small cell lung cancer) cells, and the preferable compound IC 50 <200nM, more preferably compound IC 50 <100nM。
Test example 3 measurement of p-ERK1/2 inhibitory Activity of the Compound of the present invention in NCI-H358 cells
The following method was used to determine the p-ERK1/2 inhibitory activity of the compounds of the present invention in NCI-H358 cells. The method uses an Advanced phosphor-ERK 1/2 (Thr 202/tyr 204) kit (cat 64 AERPEH) from Cisbio company, and the detailed experimental procedures can be referred to the kit instructions. NCI-H358 cells (containing KRAS G12C mutation) were purchased from the Shanghai Life sciences research institute cell resource center, chinese academy of sciences.
The experimental procedure is briefly described as follows: NCI-H358 cells were cultured in RPMI 1640 complete medium containing 10% fetal bovine serum, 100U penicillin, 100. Mu.g/mL streptomycin and 1mM Sodium Pyruvate. 30000 cells per well of NCI-H358 were plated in 96-well plates in complete medium at 37 ℃ 5% CO 2 The culture was carried out overnight in an incubator. Test compounds were dissolved in DMSO to prepare 10mM stock solutions, which were subsequently diluted using RPMI 1640 basal medium, with additions per wellPut into 90 μ L RPMI 1640 basic culture medium containing the corresponding concentration of the test compound, the final concentration range of the test compound in the reaction system is 1000nM-0.015nM, and put into a cell culture box to culture for 3 hours 40 minutes. Then 10. Mu.L of hEGF (purchased from Roche under the trademark 11376454001) prepared in RPMI 1640 basic medium was added to a final concentration of 5nM and incubated in an incubator for 20 minutes. Cell supernatants were discarded, cells were washed with ice-bath PBS, after which 45. Mu.L of 1 xcell phosphate/total protein lysis buffer (Advanced phosphate-ERK 1/2 kit component) was added per well for lysis, 96-well plates were placed on ice for half an hour for lysis, and lysates were then assayed according to the instructions of the Advanced phosphate-ERK 1/2 (Thr 202/tyr 204) kit. And finally, measuring the fluorescence intensity of each well with the emission wavelength of 620nm and 665nm under the excitation wavelength of 304nm in a TF-FRET mode by using a microplate reader, and calculating the ratio of the fluorescence intensity of 665/620 of each well. The percent inhibition of the test compound at each concentration was calculated by comparison with the fluorescence intensity ratio of the control group (0.1% DMSO), and the IC of the compound was obtained by nonlinear regression analysis of the value-inhibition at the test compound concentration using GraphPad Prism 5 software 50 Values, see table 2.
TABLE 2 inhibition of p-ERK1/2 in NCI-H358 cells by the compounds of the invention
Figure BDA0003570065910000253
Figure BDA0003570065910000261
The conclusion is that the compound of the invention has better proliferation inhibition effect on p-ERK1/2 in NCI-H358 cells, and the preferable compound IC 50 <200nM, more preferably compound IC 50 <100nM。
Test example 4 study of metabolic stability of the Compound of the present invention in human liver microsomes
1. Purpose of experiment
The purpose of this experimental study was to investigate the metabolic stability of the compounds of the invention in human liver microsomes.
2. Reagent information (see Table 3)
TABLE 3 reagent information for the experiments
Name(s) Batch number Suppliers of goods
Human mouse liver microsome 5118007 Corning Inc. of USA
Midazolam maleate 171265-201402 China Institute for food and drug control
NADPH 20595626 Roche Switzerland
Potassium dihydrogen phosphate 20150428 SINOPHARM CHEMICAL REAGENT Co.,Ltd.
Dipotassium hydrogen phosphate 20150312 SINOPHARM CHEMICAL REAGENT Co.,Ltd.
Magnesium chloride (MgCl) 2 ) F20090916 SINOPHARM CHEMICAL REAGENT Co.,Ltd.
Verapamil hydrochloride 100223-200102 China Institute for food and drug control
Glibenclamide 100135-201105 China Institute for food and drug control
DMSO 1427C108 Amresco Inc. of USA
Methanol QADG3H Honeywell Corp, USA
Acetonitrile S13A1H Honeywell Corp, USA
Formic acid A1819048 SHANGHAI ALADDIN BIOCHEMICAL TECHNOLOGY Co.,Ltd.
3. Experimental protocol
The test compound was incubated with human liver microsomes and the reaction was initiated by addition of the coenzyme NADPH. The reaction was stopped at 0, 5, 15, 30 and 60 minutes by removing 20. Mu.L of the incubation and transferring to 200. Mu.L of acetonitrile containing an internal standard. After protein precipitation, the mixture was centrifuged at 3,700rpm for 10 minutes, and the supernatant was collected. The supernatant was diluted with water 1. Intrinsic clearance in vitro was calculated from the clearance half-life of the test compound in the incubation system. Midazolam was used as an internal reference compound and 2 portions were incubated in parallel. The incubation conditions are summarized in table 4 below:
table 4 incubation conditions for compounds of the examples of the invention
Figure BDA0003570065910000262
Figure BDA0003570065910000271
4. Data analysis
Area ratio of analyte/internal standard peak (A) analyte /A IS ) Will be obtained from the instrument, the percentage remaining (% Control) is determined from A in the non-zero time point sample and the zero time sample analyte /A IS The ratio of the two is calculated. Ln (% Control) was plotted against incubation time and a linear fit was made. Test Compound clearance constant (k, min) -1 ) Clearance half-life (T) 1/2 Min) and in vitro intrinsic Clearance (CL) int ,mL·min -1 ·mg -1 proteins) was calculated from the following equation.
k=-slope
T 1/2 =0.693/k
CL int =k/C protein
C protein (mg·mL -1 ) Refers to the microsomal protein concentration in the incubation system.
5. Results of the experiment
TABLE 5 relevant parameters for human liver microsome stability of Compound 1 of the present example
Compound (I) Half life/(T) 1/2 ,min)
Example 1 40
And (4) conclusion: the compound of the embodiment of the invention has long half-life period and high stability of human liver microsomes.

Claims (17)

1. A compound of formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
Figure FDA0003570065900000011
wherein:
Figure FDA0003570065900000012
optionally a single or double bond, such that each atom thereof assumes a normal valence state;
e is selected from
Figure FDA0003570065900000013
Ring A is selected from a 4-to 12-membered heterocyclic group containing at least 2 nitrogen atoms;
x is selected from N or CR c
R a Selected from a hydrogen atom or fluorine;
R b selected from the group consisting of hydrogen atoms, -CH 2 F、-CHF 2
Figure FDA0003570065900000014
R c Selected from hydrogen atoms, halogens, alkyl groups or alkoxy groups; wherein said alkyl or alkoxy is optionally further substituted by one or more groups selected from halogen, hydroxyCyano, alkyl or alkoxy; r is c Preferably halogen, more preferably fluorine or chlorine;
R 1 selected from hydrogen atoms, halogens, alkyl groups or alkoxy groups; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from the group consisting of halo, hydroxy, cyano, alkyl or alkoxy; r is 1 Preferably halogen, more preferably chlorine;
R 2 is selected from aryl OR heteroaryl, wherein said aryl OR heteroaryl is optionally further substituted by one OR more groups selected from alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with the substituent(s); wherein said alkyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted by one OR more substituents selected from the group consisting of alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with the substituent(s);
R 3 selected from absent, hydrogen atom, halogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more R A Substituted; r 3 Preferably absent or a hydrogen atom;
R A each independently selected from alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 (ii) a Wherein said alkyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted by one OR more substituents selected from the group consisting of alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-SO 2 R 5 Substituted with the substituent(s);
R 4 identical or different, each independently selected from hydrogen atom, halogen, hydroxyl, alkyl or alkoxy, R 4 Preferably a hydrogen atom or an alkyl group;
R 5 selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with the substituent(s);
R 6 and R 7 Each independently selected from the group consisting of hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with a substituent of (a);
or, R 6 And R 7 Together with the atoms to which they are attached form a 4-to 8-membered heterocyclic group containing one or more of N, O, S or SO within the 4-to 8-membered heterocyclic group 2 And said 4-to 8-membered heterocyclyl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, = O, -C (O) R 8 、-C(O)OR 8 、-OC(O)R 8 、-NR 9 R 10 、-C(O)NR 9 R 10 、-SO 2 NR 9 R 10 or-NR 9 C(O)R 10 Substituted with a substituent of (a);
R 8 、R 9 and R 10 Each independently selected from the group consisting of hydrogen, alkyl, amino, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of hydroxy, halogen, nitro, amino, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxy or carboxylate;
m is selected from 0,1 or 2.
2. The compound according to claim 1, which is a compound represented by the general formula (II) or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof:
Figure FDA0003570065900000021
wherein: ring A, X, R 1 、R 2 、R 4 E and m are as defined in claim 1.
3. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein E is selected from:
Figure FDA0003570065900000031
4. a compound according to claim 1 or 2, or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, wherein ring a is selected from a 4-to 12-membered heterocyclyl group containing 2 nitrogen atoms, preferably a 6-to 8-membered heterocyclyl group.
5. A compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
x is selected from CR c
R c Selected from a hydrogen atom, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy; r is c Preferably halogen, more preferably fluorine or chlorine.
6. The compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 1 Selected from hydrogen, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy; r 1 Preferably halogen, more preferably chlorine.
7. The compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 Selected from:
Figure FDA0003570065900000032
8. a compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein: r is 4 Selected from a hydrogen atom or an alkyl group; wherein said alkyl is preferably methyl.
9. A compound according to claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein said compound is:
Figure FDA0003570065900000041
10. a process for the preparation of a compound of general formula (I) according to claim 1 or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, which comprises:
Figure FDA0003570065900000042
reacting the compound of the general formula (IA) with the compound of the general formula (IB) under alkaline conditions, and optionally further removing a protecting group to obtain a compound of the general formula (I);
wherein:
X 1 is a leaving group, preferably chloro;
ring A, R 1 ~R 4 X, E and m are as defined in claim 1.
11. A compound of formula (IA) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof,
Figure FDA0003570065900000051
wherein: ring A, R 1 ~R 4 X and m are as defined in claim 1.
12. A compound according to claim 11, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein said compound is:
Figure FDA0003570065900000052
13. a pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 9, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
14. Use of a compound according to any one of claims 1 to 9, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the preparation of a K-Ras gtpase inhibitor, wherein the K-Ras gtpase inhibitor is preferably a KRAS G12C inhibitor.
15. Use of a compound according to any one of claims 1 to 9, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the manufacture of a medicament for the treatment of a disease mediated by a KRAS mutation, wherein the disease mediated by a KRAS mutation is selected from the group consisting of cancer, wherein the cancer is selected from the group consisting of pancreatic cancer, colorectal cancer, lung cancer, multiple myeloma, uterine cancer, bile duct cancer, gastric cancer, bladder cancer, diffuse large B-cell lymphoma, rhabdomyosarcoma, cutaneous squamous cell carcinoma, cervical cancer, testicular germ cell carcinoma, preferably pancreatic cancer, colorectal cancer and lung cancer, wherein the KRAS mutation is preferably a KRAS G12C mutation.
16. Use of a compound according to any one of claims 1 to 9, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the manufacture of a medicament for the treatment of a cancer selected from the group consisting of pancreatic cancer, colorectal cancer, lung cancer, multiple myeloma, uterine cancer, bile duct cancer, gastric cancer, bladder cancer, diffuse large B-cell lymphoma, rhabdomyosarcoma, cutaneous squamous cell carcinoma, cervical cancer, testicular germ cell carcinoma, preferably pancreatic cancer, colorectal cancer and lung cancer.
17. The use according to claim 15 or 16, wherein the lung cancer is non-small cell lung cancer.
CN202210319842.8A 2021-07-01 2022-03-29 Tetracyclic derivative, preparation method and medical application thereof Pending CN115557949A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021107447980 2021-07-01
CN202110744798 2021-07-01

Publications (1)

Publication Number Publication Date
CN115557949A true CN115557949A (en) 2023-01-03

Family

ID=84738064

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210319842.8A Pending CN115557949A (en) 2021-07-01 2022-03-29 Tetracyclic derivative, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN115557949A (en)

Similar Documents

Publication Publication Date Title
KR102604975B1 (en) (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo [1,5-A] Preparation of pyrimidine-3-carboxamide and its uses
EP3386981B1 (en) Heterocycles useful as anti-cancer agents
WO2022206723A1 (en) Heterocyclic derivative, and preparation method therefor and use thereof in medicine
AU2017302019A1 (en) Macrocycle kinase inhibitors
WO2015077193A1 (en) Inhibitors of lysine methyl transferase
WO2020188467A1 (en) Condensed tricyclic compound used as kinase inhibitor
WO2023025116A1 (en) Heterocyclic derivative, preparation method therefor and use thereof in medicine
CN116535401A (en) Novel PARP1 inhibitors and uses thereof
CN116249683A (en) Deuteromethyl substituted pyrazinopyrazinoquinolinone derivative, preparation method and application thereof in medicine
EP3498707A1 (en) Fgfr4 inhibitor and preparation method and use thereof
WO2023001229A1 (en) Pyrimidocyclic derivative, preparation method therefor, and use thereof
WO2022037630A1 (en) Tetracyclic derivative, method for preparing same and use thereof in medicine
CN113929681A (en) Tetracyclic derivative and preparation method and application thereof
CN113929676A (en) Pyridino-heterocyclic derivative and preparation method and application thereof
CN116162099A (en) Heterocyclic derivative and preparation method and application thereof
CN116514846A (en) Heterocyclic derivative, preparation method and medical application thereof
CN116390923A (en) Heterocyclic derivative and preparation method and application thereof
WO2022037631A1 (en) Heterocyclic derivative and preparation method therefor and use thereof
CN115557949A (en) Tetracyclic derivative, preparation method and medical application thereof
CN116102533A (en) Aromatic heterocyclic compound and application thereof
WO2017049711A1 (en) Quinoline derivative, and pharmaceutical composition, preparation method and use thereof
WO2022206724A1 (en) Heterocyclic derivative, and preparation method therefor and use thereof
CN115403575A (en) Heteroaromatic ring derivative and preparation method and application thereof
CN115611898A (en) Tetracyclic derivative, preparation method and medical application thereof
CN115304602A (en) Pyrazinopyrazinonaphthyridinedione derivatives, preparation method and medical application thereof

Legal Events

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