CN115403575A - Heteroaromatic ring derivative and preparation method and application thereof - Google Patents

Heteroaromatic ring derivative and preparation method and application thereof Download PDF

Info

Publication number
CN115403575A
CN115403575A CN202210100843.3A CN202210100843A CN115403575A CN 115403575 A CN115403575 A CN 115403575A CN 202210100843 A CN202210100843 A CN 202210100843A CN 115403575 A CN115403575 A CN 115403575A
Authority
CN
China
Prior art keywords
alkyl
group
heteroaryl
cancer
aryl
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
CN202210100843.3A
Other languages
Chinese (zh)
Inventor
陈友喜
赵雯雯
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 CN115403575A publication Critical patent/CN115403575A/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
    • C07D471/02Heterocyclic 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

Abstract

The invention relates to a heteroaromatic ring derivative, a preparation method thereof and application thereof in medicines. Specifically, the invention relates to heteroaromatic ring derivatives shown in a general formula (I), a preparation method thereof, pharmaceutically acceptable salts thereof, and application thereof as a therapeutic agent, particularly 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

Heteroaromatic ring derivative and preparation method and application thereof
Technical Field
The invention relates to a heteroaromatic ring 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, the RAS is activated upon reception 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 a 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. As GTP is 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 overactivated RAS signal transduction may ultimately lead to cancer.
Structurally, RAS proteins contain a G domain responsible for the 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 major interaction is the hydrogen bond formed by threonine-35 and glycine-60 with the gamma-phosphate of GTP, which maintains the switch I and switch II regions in their active conformations, respectively. 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 carcinoma, etc., while rarely (< 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 indicate that lung cancer KRAS mutations, including G12C, are mutually exclusive from other known driver oncogenic mutations in NSCLC, including EGFR, ALK, ROS1, RET, and BRAF, indicating the uniqueness of KRAS mutations in lung cancer. While, 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 for the clinical development of the KRAS inhibitor is intense at home and abroad, wherein the KRAS enzyme inhibitor MRTX-849 developed by Mirati Therapeutics Inc. enters the second clinical stage and is used for preventing and treating diseases such as advanced solid tumors, metastatic colorectal cancer and metastatic non-small cell lung cancer. There are also other KRAS inhibitors in the research including AMG-510 (Amgen Inc, phase 2) and MRTX1257 (Mirati Therapeutics Inc, found). 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, 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 heteroaromatic ring derivative shown as a general formula (I), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof:
Figure BDA0003492412210000021
wherein:
ring B is selected from 5-6 membered heteroaryl or 5-6 membered heterocyclyl;
x and Y are each independently selected from N or CR 4
L is selected from C 1 -C 6 Alkylene, wherein said alkylene is optionally further substituted with one OR more groups selected from alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, cyano, -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-S (O) r R 5 Substituted with a substituent of (a);
e is selected from
Figure BDA0003492412210000022
G is selected from a 4-12 membered heterocyclic group containing 1-2 nitrogen atoms, wherein said heterocyclic group is optionally further substituted with one or more R c Substituted;
R a selected from a hydrogen atom or fluorine;
R b selected from the group consisting of hydrogen atoms, -CH 2 F、-CHF 2 or-CH 2 -NR 6 R 7
R c The same OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclic group, 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-S (O) r R 5 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-S (O) r R 5 Substituted with the substituent(s);
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 groups selected from halogen, hydroxy, cyano, alkylOr substituted with a substituent of alkoxy; r 1 Preferably a hydrogen atom;
R 2 selected from aryl OR heteroaryl, wherein said 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-S (O) r 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-S (O) r R 5 Substituted with the substituent(s);
R 3 selected from absent, hydrogen atom, halogen, alkyl, alkoxy, cyano, haloalkyl or haloalkoxy, preferably absent or a hydrogen atom;
R 4 identical or different, each independently 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 4 Preferably halogen, more preferably fluorine or chlorine;
R 5 selected from hydrogen atom, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted by one or more groups selected from 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 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 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;
n is selected from 0, 1 or 2;
r is 0, 1 or 2.
The invention provides a compound shown in a general formula (I) or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, wherein the compound is a compound shown in a general formula (II) or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof:
Figure BDA0003492412210000041
wherein:
ring B, R 1 ~R 4 L, G, E and n are as defined in formula (I).
The present invention provides a compound of general formula (I) or (II) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
-G-E is selected from:
Figure BDA0003492412210000042
R c identical or different, each independently selected from a hydrogen atom, a halogen, an alkyl group or an alkoxy group, preferably an alkyl group, more preferably a methyl group;
m is selected from 0, 1,2,3 or 4;
e is as defined in formula (I).
In a preferred embodiment of the present invention, the present invention provides a compound of formula (I) or (II) or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
-G-E is selected from:
Figure BDA0003492412210000051
R c identical or different, each independently selected from a hydrogen atom, a halogen, an alkyl group or an alkoxy group, preferably an alkyl group, more preferably a methyl group; m is selected from 0, 1,2,3 or 4;
e is selected from:
Figure BDA0003492412210000052
in a preferred embodiment, the present invention provides a compound of formula (I) or (II) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R is 1 Selected from hydrogen, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy, R 1 Preferably a hydrogen atom.
In a preferred embodiment of the present invention, the present invention provides a compound of formula (I) or (II) or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from phenyl, naphthyl, pyridyl, benzothiazolyl or benzopyrazolyl, wherein said phenyl, naphthyl, pyridyl, benzothiazolyl or benzopyrazolyl is optionally further substituted with one or more substituents selected from halogen, hydroxy, alkyl, alkoxy, cycloalkyl or-NR 6 R 7 Wherein said alkyl or alkoxy is optionally further substituted by one or more substituents selected from halogen or-NR 6 R 7 Substituted with the substituent(s); wherein said halogen is preferably fluorine;
R 6 and R 7 Each independently selected from a hydrogen atom or an alkyl group, wherein the alkyl group is preferably a methyl group.
In a preferred embodiment of the present invention, the present invention provides a compound of formula (I) or (II) or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from:
Figure BDA0003492412210000053
Figure BDA0003492412210000061
in a preferred embodiment, the present invention provides a compound of formula (I) or (II) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein R 3 Selected from absent or hydrogen atoms.
In a preferred embodiment of the present invention, the present invention provides a compound of formula (I) or (II) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein ring B is selected from:
Figure BDA0003492412210000062
R 3 the definition of (A) is described in the general formula (I).
In a preferred embodiment, the present invention provides a compound of formula (I) or (II) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R is 4 Selected from hydrogen, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy, R 4 Preferably halogen, more preferably fluorine or chlorine.
In a preferred embodiment of the present invention, the present invention provides a compound of formula (I) or (II) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein L is selected from the group consisting of-CH 2 -、-CH 2 CH 2 -or-CH (CH) 3 )-。
Typical compounds of the invention include, but are not limited to:
Figure BDA0003492412210000063
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.
Further, the present invention provides a process for the preparation of a compound of general formula (I) or a stereoisomer, tautomer or a pharmaceutically acceptable salt thereof, which process comprises:
Figure BDA0003492412210000071
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 B, R 1 ~R 3 X, Y, L, G, E and n are as defined in formula (I).
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.
The invention also provides 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 diseases mediated by KRAS mutation, wherein the diseases mediated by KRAS mutation are 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 and testicular germ cell cancer, preferably pancreatic cancer, colorectal cancer and lung cancer; wherein the lung cancer is preferably non-small cell lung cancer.
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, or diastereomer thereof, or a mixture thereof, or a 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, and 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 it may be mixed 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 nanoparticles, 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, for example, starches, 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 hydroxide oxide, 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:
"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. Alkyl groups may be substituted or unsubstituted.
An "alkylene" is a divalent alkyl group. Preferably C 1 -C 10 Alkylene, more preferably C 1 -C 6 Alkylene, particularly preferably C 1 -C 4 An alkylene group. Examples of alkylene groups include, but are not limited to, methylene, ethylene, and,
Figure BDA0003492412210000091
N-propylene, and the like. The alkylene group may be substituted or unsubstituted.
"alkenyl" refers to 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 groups 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 a single carbon atom (called the spiro atom) with each other, containing 1 or more double bonds within the ring, but no ring has a completely 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-ring 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 preferably 7 to 10. 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]]And a heptyl radical.
"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 ring atoms are 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),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] rings]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. Examples of preferably bicyclic heteroaryl groups, heteroaryl groups include, but are not limited to, furyl, pyridyl, 2-oxo-1, 2-dihydropyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, isoxazolyl, oxazolyl, oxadiazolyl, imidazolyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, 1,2, 3-thiadiazolyl, benzodioxolyl, benzothiophenyl, benzimidazolyl, indolyl, isoindolyl, 1, 3-dioxo-isoindolyl, quinolinyl, indazolyl, benzisothiazolyl, benzoxazolyl, benzisoxazolyl, benz,
Figure BDA0003492412210000121
Heteroaryl groups may be substituted or unsubstituted.
"alkoxy" refers to a radical of (alkyl-O-). Wherein alkyl is as defined herein. C 1 -C 6 Alkoxy groups of (4) are preferred. Examples 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.
"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" means 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" refers to-NH 2
"cyano" means-CN.
"nitro" means-NO 2
"benzyl" means-CH 2 -phenyl.
"carboxy" means-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 propyl phosphoric anhydride.
"DPPA" refers to diphenylphosphoryl azide.
"DEA" refers to diethylamine.
"X-PHOS Pd G2" chloro (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II).
"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.
The term "substituted" or "substituted" as used herein, 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 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-S (O) r R 5 Substituted with a substituent of (a);
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 containing 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 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;
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, the structures described herein also include all isomers (e.g., diastereomers, enantiomers, and atropisomers and geometric (conformational) isomeric forms) of such structures, e.g., the R and S configurations of the various asymmetric centers, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers.
"pharmaceutically acceptable salts" refers to certain salts of the above compounds which retain their biological activity and are suitable for pharmaceutical use. 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 BDA0003492412210000141
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 B, R 1 ~R 3 X, Y, L, G, E and n 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. Must be provided withThe following examples are illustrative of the invention and are not intended to be limiting thereof. 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 doublets, dt = doublet of triplets. When coupling constants are provided, they are in Hz.
The mass spectrum is measured by an LC/MS instrument, and the ionization mode can be ESI or APCI.
The thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) 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 are used without further purification, unless otherwise indicated, commercially available manufacturers include, but are not limited to, shanghai haohnhong biomedical science and technology limited, shanghai shaoshimo reagents limited, shanghai beide medical science and technology limited, saen chemical technology (shanghai) limited, shanghai ling medical science and technology limited, and the like.
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 for 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: ethyl acetate and tetrahydrofuran, 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.
Room temperature: 20-30 ℃.
Example 1
1-(3-(1-(7-(2-amino-5-fluorobenzo[d]thiazol-4-yl)-8-chloro-6-fluoro-1H-[1,2,3]triazolo[4,5-c]quinolin-1-yl)ethyl)azetidin-1-yl)prop-2-en-1-one
1- (3- (1- (7- (2-amino-5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidin-1-yl) prop-2-en-1-one
Figure BDA0003492412210000151
Figure BDA0003492412210000161
First step of
1-(3-fluoro-2-methoxyphenyl)thiourea
1- (3-fluoro-2-methoxyphenyl) thiourea
3-fluoro-2-methoxyaniline 1a (20g, 141.70mmol) is added into tetrahydrofuran (500 mL), a tetrahydrofuran solution (100 mL) of benzoyl isothiocyanate 1b (23.13g, 141.70mmol) is slowly added dropwise, reaction is continued for 3 hours at room temperature after the dropwise addition is finished, LCMS monitors that all raw materials are converted into intermediates, water (80 mL) and sodium hydroxide (6.80g, 170.04mmol) are added, and the mixture is heated to 80 ℃ for reaction overnight. Cooled to room temperature, extracted with ethyl acetate (100 mL × 1), the organic phase washed with saturated brine (100 mL × 1), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the obtained residue was separated and purified by silica gel column chromatography (eluent: system a) to obtain the product 1- (3-fluoro-2-methoxyphenyl) thiourea 1c (22g, 109.87mmol), yield: 77.55 percent. MS m/z (ESI) 201.1[ 2], [ M ] +1] +
Second step of
5-fluoro-4-methoxybenzo[d]thiazol-2-amine
5-fluoro-4-methoxybenzo [ d ] thiazol-2-amine
1- (3-fluoro-2-methoxyphenyl) thiourea 1c (7.09g, 35.41mmol) was added to acetic acid (200 mL), lithium bromide (4.61g, 53.11mmol) was added, liquid bromine (5.77g, 36.12mmol) was slowly added dropwise, maintaining the temperature below 30 ℃. After the dropwise addition, the reaction solution was heated to 40 ℃ to react overnight. The reaction mixture was cooled, poured into water (500 mL), made basic with a saturated sodium carbonate solution, extracted with ethyl acetate (300 mL × 1), the organic phase was washed with saturated brine (100 mL × 1), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue obtained was separated and purified by silica gel column chromatography (eluent: system a) to give the product 5-fluoro-4-methoxybenzo [ d ] thiazol-2-amine 1d (7 g, 35.31mmol), yield: 99.73 percent.
MS m/z(ESI):199.1[M+1] +
The third step
2-amino-5-fluorobenzo[d]thiazol-4-ol
2-amino-5-fluorobenzo [ d ] thiazol-4-ol
5-fluoro-4-methoxybenzo [ d ] thiazol-2-amine 1d (7g, 35.31mmol) was added to methylene chloride (70 mL), cooled to 0 deg.C, boron tribromide (22.12g, 88.29mmol, 8.51mL) was added dropwise, and after completion of the addition, the mixture was allowed to warm to room temperature for reaction overnight. The reaction solution was poured into ice water (300 mL), made alkaline with a saturated sodium carbonate solution, extracted with ethyl acetate (500 mL. Times.1), the organic phase was washed with saturated brine (100 mL. Times.1), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product 2-amino-5-fluorobenzo [ d ] thiazol-4-ol 1e (5.2g, 28.23mmol), yield: 79.94 percent.
MS m/z(ESI):185.1[M+1] +
The fourth step
tert-butyl(4-((tert-butoxycarbonyl)oxy)-5-fluorobenzo[d]thiazol-2-yl)carbamate
(4- ((tert-Butoxycarbonyl) oxy) -5-fluorobenzo [ d ] thiazol-2-yl) carbamic acid tert-butyl ester
2-amino-5-fluorobenzo [ d ] thiazol-4-ol 1e (10g, 54.29mmol), dimethylaminopyridine (1.33g, 10.86mmol), triethylamine (10.99g, 108.58mmol, 15.13mL), and di-tert-butyl dicarbonate (23.70g, 108.58mmol) were added to dichloromethane (80 mL) and reacted at room temperature for 4 hours. The reaction mixture was concentrated under reduced pressure, ethyl acetate (100 mL) and water (50 mL) were added, the mixture was separated, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give crude product tert-butyl (4- ((tert-butoxycarbonyl) oxy) -5-fluorobenzo [ d ] thiazol-2-yl) carbamate 1f (20g, 52.03mmol), yield: 95.83 percent.
MS m/z(ESI):385.1[M+1] +
The fifth step
tert-butyl(5-fluoro-4-hydroxybenzo[d]thiazol-2-yl)carbamate
(5-fluoro-4-hydroxybenzo [ d ] thiazol-2-yl) carbamic acid tert-butyl ester
Tert-butyl (4- ((tert-butoxycarbonyl) oxy) -5-fluorobenzo [ d ] thiazol-2-yl) carbamate 1f (20g, 52.03mmol) was added to a mixed solvent of tetrahydrofuran (80 mL) and water (20 mL), cooled to 0 ℃, lithium hydroxide monohydrate (10.92g, 260.13mmol) was added, and the mixture was allowed to react overnight at room temperature. The reaction mixture was added with ethyl acetate (100 mL) and water (50 mL), extracted with ethyl acetate (100 mL. Times.2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product tert-butyl (5-fluoro-4-hydroxybenzo [ d ] thiazol-2-yl) carbamate 1g (14.45g, 50.83mmol), yield: 97.69 percent.
MS m/z(ESI):228.9[M+1-56] +
The sixth step
2-((tert-butoxycarbonyl)amino)-5-fluorobenzo[d]thiazol-4-yl trifluoromethanesulfonate
2- ((tert-Butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl trifluoromethanesulfonate
Under ice bath, (5-fluoro-4-hydroxybenzo [ d ]]Thiazol-2-yl) carbamic acid tert-butyl ester 1g (20g, 70.35mmol) was dissolved in methylene chloride (80 mL), and pyridine (11.13g, 140.69mmol, 11.36mL), trifluoromethanesulfonic anhydride (23.82g, 84.42mmol, 14.26mL) were added in this order and stirred for 30 minutes. Adding water (150 mL) into the reaction liquid, extracting with dichloromethane (150 mL multiplied by 3), combining organic phases, washing with citric acid monohydrate aqueous solution (50 mL) and saturated saline (50 mL) in sequence, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the obtained residue by silica gel column chromatography (eluent: A system) to obtain the product 2- ((tert-butyloxycarbonyl radical)) Amino) -5-fluorobenzo [ d]Thiazol-4-yl triflate for 1h (13.6 g, 32.66mmol), yield: 46.43 percent. MS m/z (ESI) 361.0[ m ] +1-56 ]] +
Seventh step
(2-((tert-butoxycarbonyl)amino)-5-fluorobenzo[d]thiazol-4-yl)boronic acid
(2- ((tert-Butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) boronic acid
2- ((tert-Butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl trifluoromethanesulfonate (5 g, 12.01mmol) was mixed with pinacol diboron (24.40g, 96.07mmol) in 1, 4-dioxane (80 mL), potassium acetate (3.54g, 36.03mmol) was added, argon was substituted, stirring was carried out for 10 minutes, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (2.64g, 3.60mmol) was added, argon was used for protection, and stirring was carried out at 100 ℃ for 8 hours. And adding pinacol diboron (24.40g, 96.07mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (2.64g, 3.60mmol), stirring for 8 hours at 100 ℃ under the protection of argon. 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 obtain 1i (2g, 6.41mmol) of the product (2- ((t-butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) boronic acid, yield: 53.36 percent.
MS m/z(ESI):312.9[M+1] +
Eighth step
tert-butyl
3- (1- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylate3- (1- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylic acid tert-butyl ester
7-bromo-4,6-dichloro-8-fluoro-3-nitroquinoline 1j (527.47mg, 1.55mmol, prepared according to published patent WO 2019110751) and tert-butyl 3- (1-aminoethyl) azetidine-1-carboxylate 1k (404mg, 2.02mmol) were dissolved in acetonitrile (15 mL), cooled to 0 deg.C, N-diisopropylethylamine (601.62mg, 4.66mmol) was added dropwise, turned to room temperature, and the reaction was continued for 6 hours. Concentration under reduced pressure, and separation and purification of the obtained residue by silica gel column chromatography (eluent: system a) to give tert-butyl 3- (1- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylate 1m (650mg, 1.29mmol), yield: 83.16 percent.
MS m/z(ESI):504.8[M+1] +
The ninth step
tert-butyl
3-(1-((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl)amino)ethyl)azetidine-1-carboxylate
3- (1- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylic acid tert-butyl ester
Tert-butyl 3- (1- ((7-bromo-6-chloro-8-fluoro-3-nitroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylate 1m (650 mg, 1.29mmol), ammonium chloride (344.84mg, 6.45mmol) and iron powder (360.32mg, 6.45mmol) were dissolved in a mixed solvent of methanol (20 mL) and water (5 mL), and heated to 90 ℃ for 5 hours. After the reaction, the reaction mixture was filtered while it was hot, and the filtrate was concentrated under reduced pressure to remove methanol. The system was extracted with ethyl acetate (100 mL. Times.2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the resulting residue was separated and purified by silica gel column chromatography (eluent: system A) to give the product tert-butyl 3- (1- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylate 1n (500mg, 1.06mmol), yield: 81.79 percent.
MS m/z(ESI):474.8[M+1] +
The tenth step
tert-butyl3-(1-(7-bromo-8-chloro-6-fluoro-1H-[1,2,3]triazolo[4,5-c]quinolin-1-yl)ethyl)azetidine-1-carboxyla
te
3- (1- (7-bromo-8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylic acid tert-butyl ester
Tert-butyl 3- (1- ((3-amino-7-bromo-6-chloro-8-fluoroquinolin-4-yl) amino) ethyl) azetidine-1-carboxylate 1n (320mg, 675.44. Mu. Mol) was dissolved in a mixed solvent of acetic acid (6 mL) and water (2 mL), cooled to 0 ℃, added with sodium nitrite (69.91mg, 1.01mmol), reacted at 0 ℃ for 0.5 hour, and warmed to room temperature for 2 hours. After the reaction, the system was made basic with a saturated sodium carbonate solution, extracted with ethyl acetate (100 mL. Times.2), the organic phases were combined, washed with a saturated brine (100 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the obtained residue was separated and purified by silica gel column chromatography (eluent: system A) to give tert-butyl 3- (1- (7-bromo-8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylate 1p (300mg, 618.88. Mu. Mol), yield: 91.63 percent.
MS m/z(ESI):484.1[M+1] +
The eleventh step
tert-butyl3-(1-(7-(2-((tert-butoxycarbonyl)amino)-5-fluorobenzo[d]thiazol-4-yl)-8-chloro-6-fluoro-1H-[1,2,3]triazolo[4,5-c]quinolin-1-yl)ethyl)azetidine-1-carboxylate
3- (1- (7- (2- ((tert-butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylic acid tert-butyl ester
3- (1- (7-bromo-8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylic acid tert-butyl ester 1p (200mg, 412.58. Mu. Mol), (2- ((tert-butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) boronic acid 1i (154.53mg, 495.10. Mu. Mol), potassium phosphate (175.15mg, 825.17. Mu. Mol) and X-PHOS Pd G2 (64.84mg, 82.52. Mu. Mol) were dissolved in tetrahydrofuran (5 mL), protected with argon, heated to 50 ℃ and reacted overnight. After completion of the reaction, filtration was carried out, the filtrate was concentrated under reduced pressure, and the obtained residue was separated and purified by preparative HPLC (separation column: AKZONOBEL Kromasil; 250X 21.2mm I.D.;5 μm; mobile phase A:0.05% TFA + H2O, mobile phase B: acetonitrile; flow rate: 20 mL/min) to obtain tert-butyl 3- (1- (7- (2- ((tert-butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylate 1q (50mg, 74.39 μmol), yield: 18.03 percent.
MS m/z(ESI):671.8[M+1] +
The twelfth step
4-(1-(1-(azetidin-3-yl)ethyl)-8-chloro-6-fluoro-1H-[1,2,3]triazolo[4,5-c]quinolin-7-yl)-5-fluorobenzo[d]thiazol-2-amine
4- (1- (1- (azetidin-3-yl) ethyl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-7-yl) -5-fluorobenzo [ d ] thiazol-2-amine
Tert-butyl 3- (1- (7- (2- ((tert-butoxycarbonyl) amino) -5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidine-1-carboxylate 1q (49.12mg, 73.08. Mu. Mol) was dissolved in dichloromethane (3 mL), and a1, 4-dioxane solution of hydrochloric acid (4M, 2mL) was added and reacted at room temperature for 2 hours. After the reaction was completed, concentration under reduced pressure gave crude 4- (1- (1- (azetidin-3-yl) ethyl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-7-yl) -5-fluorobenzo [ d ] thiazol-2-amine 1r (34.49mg, 73.08. Mu. Mol), yield: 100 percent.
MS m/z(ESI):471.8[M+1] +
Thirteenth step
1-(3-(1-(7-(2-amino-5-fluorobenzo[d]thiazol-4-yl)-8-chloro-6-fluoro-1H-[1,2,3]triazolo[4,5-c]quinolin-1-yl)ethyl)azetidin-1-yl)prop-2-en-1-one
1- (3- (1- (7- (2-amino-5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidin-1-yl) prop-2-en-1-one
4- (1- (1- (azetidin-3-yl) ethyl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-7-yl) -5-fluorobenzo [ d ] thiazol-2-amine 1r (34.13mg, 72.33. Mu. Mol) was dissolved in dichloromethane (3 mL), triethylamine (21.96mg, 216.98. Mu. Mol) was added, cooling to 0 ℃ was performed, a solution of acryloyl chloride (7.86mg, 86.79. Mu. Mol) in dichloromethane (1 mL) was added dropwise, and the reaction was continued at 0 ℃ for 1 hour. The reaction liquid was concentrated under reduced pressure, and the obtained residue was separated and purified by preparative HPLC (separation column: AKZONOBEL Kromasil; 250X 21.2mm I.D.;5 μm; mobile phase A:0.05% TFA + H2O, mobile phase B: acetonitrile; flow rate: 20 mL/min) to give 1- (3- (1- (7- (2-amino-5-fluorobenzo [ d ] thiazol-4-yl) -8-chloro-6-fluoro-1H- [1,2,3] triazolo [4,5-c ] quinolin-1-yl) ethyl) azetidin-1-yl) propan-2-en-1-one 1 (5mg, 3.43 μmol), yield: 11.86 percent.
MS m/z(ESI):526.1[M+1] +
1 H NMR(400MHz,Methanol-d4)δ9.55(s,1H),8.67(t,J=2.2Hz,1H),7.79(dd,J=8.7,5.1Hz,1H),7.09(t,J=9.1Hz,1H),6.41(dd,J=17.0,10.3Hz,1H),6.29–6.20(m,1H),5.90(t,J=7.5Hz,1H),5.72(ddd,J=30.2,9.9,2.4Hz,1H),4.44(dt,J=35.2,9.4Hz,2H),4.28–4.05(m,2H),3.97–3.87(m,1H),1.77(dd,J=6.5,2.4Hz,3H).
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: recombinant human KRAS G12C protein (aa 1-169) was prepared at a concentration of 4uM using reaction buffer (20mM HEPES,150mM NaCl,1mM MgCl2, and 1mM DTT). Test compounds were dissolved in DMSO to prepare 10mM stock solutions, which were then diluted with reaction buffer for use. First, 1.5uL of test compound diluted with reaction buffer (final concentration of reaction system is 3. Mu.M or 10. Mu.M) was added to the well, then 23.5uL of reaction buffer was added and mixed, then 25 uL of 4uM recombinant human KRAS G12C protein was added, after incubation for 5 or 15 minutes at room temperature, 5uL of acetic acid was added to terminate the reaction, and the sample was transferred to a sample introduction bottle. The covalent binding rate of the test compound to the KRAS G12C Protein was measured using an Agilent 1290/6530 instrument, and the sample was purified on a liquid chromatography column (XBridge Protein BEH C4,
Figure BDA0003492412210000212
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 are analyzed by using MassHunter Workstation Software bioconjugate Version B.08.00 Software, the covalent Binding Rate (Binding Rate) of the tested compound and KRAS G12C protein is obtained under the condition of incubation for 5min, wherein the concentration of the tested compound is 3 mu M.
Compound number Protein covalent binding Rate (%)
1 47.6
The conclusion is that the compound 1 has better covalent binding rate with KRAS G12C protein.
Test example 2 measurement of inhibition of NCI-H358 cell proliferation by the Compound of the present invention
The following methods were used to determine the effect of the compounds of the invention on proliferation of NCI-H358 cells. 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 BDA0003492412210000211
The luminesent Cell Viability Assay kit (Promega, cat # G7573).
The experimental method is operated according to the steps of the kit specification, and is briefly 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 in the incubator is carried out overnight, followed by a further 120 hours after addition of the test compound. 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. Percent inhibition of compounds at each concentration point was calculated by comparison with the values of the control (0.3% DMSO), and then in GraphPad Prism 5 softwarePerforming nonlinear regression analysis on the logarithmic inhibition rate of the compound concentration to obtain the IC of the compound for inhibiting cell proliferation 50 The value is obtained.
Compound number IC 50 (nM)
1 41
The conclusion is that the compound 1 has better proliferation inhibition effect on NCI-H358 (human non-small cell lung cancer) cells.
Test example 3 determination of p-ERK1/2 inhibitory Activity of Compounds of the present invention in NCI-H358 cells
The following methods were used to determine the inhibitory activity of the compounds of the present invention on p-ERK1/2 in NCI-H358 cells. The method uses Advanced phosphor-ERK 1/2 (Thr 202/tyr 204) kit (cat. 64 AERPEH) of Cisbio company, and the detailed experimental operation can refer to the kit instruction. 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 a 10mM stock solution, which was then diluted with RPMI 1640 basic medium, to which 90. Mu.L of RPMI 1640 basic medium containing the test compound at the corresponding concentration was added per well, the final concentration of the test compound in the reaction system ranged from 1000nM to 0.015nM, and the cells were cultured in a cell culture chamber for 3 hours and 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. Finally, the fluorescence intensity of each well with the emission wavelength of 620nM and 665nM under the excitation wavelength of 304nM is measured in a microplate reader in TF-FRET mode, and the ratio of the fluorescence intensity of 665/620 of each well is calculated. 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 numerical-inhibition was subjected to nonlinear regression analysis by GraphPad Prism 5 software at the test compound concentration to obtain the IC of the compound 50 The value is obtained.
The conclusion is that the compound has better proliferation inhibition effect on p-ERK1/2 in NCI-H358 cells, and the IC of the compound is optimized 50 <500nM, more preferably IC of the compound 50 <200nM。

Claims (17)

1. A compound of formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
Figure FDA0003492412200000011
wherein:
ring B is selected from 5-6 membered heteroaryl or 5-6 membered heterocyclyl;
x and Y are each independently selected from N or CR 4
L is selected from C 1 -C 6 Alkylene, wherein said alkylene is optionally further substituted with one OR more groups selected from alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, cyano, -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-S (O) r R 5 Substituted with a substituent of (a);
e is selected from
Figure FDA0003492412200000012
G is selected from a 4-12 membered heterocyclic group containing 1-2 nitrogen atoms, wherein said heterocyclic group is optionally further substituted with one or more R c Substituted;
R a selected from hydrogen atoms or fluorine;
R b selected from the group consisting of hydrogen atoms, -CH 2 F、-CHF 2 or-CH 2 -NR 6 R 7
R c The same OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclic group, 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-S (O) r R 5 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-S (O) r R 5 Substituted with the substituent(s);
R 1 selected from hydrogen, halogen, alkyl or alkoxy; 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 a hydrogen atom;
R 2 selected from aryl OR heteroaryl, wherein said 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-S (O) r 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-S (O) r R 5 Substituted with the substituent(s);
R 3 selected from absent, hydrogen atom, halogen, alkyl, alkoxy, cyano, haloalkyl or haloalkoxy, preferably absent or a hydrogen atom;
R 4 identical or different, each independently 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 4 Preferably halogen, more preferably fluorine or chlorine;
R 5 selected from hydrogen atom, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further substituted by one or more groups selected from 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 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 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;
n is selected from 0, 1 or 2;
r is 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 FDA0003492412200000021
wherein:
ring B, R 1 ~R 4 L, G, E and n are as defined in claim 1.
3. A compound according to claim 1 or 2, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
-G-E is selected from:
Figure FDA0003492412200000031
R c identical or different, each independently selected from a hydrogen atom, a halogen, an alkyl or an alkoxy group, preferably an alkyl group, more preferably a methyl group;
m is selected from 0, 1,2,3 or 4;
e is as defined in claim 1.
4. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein E is selected from:
Figure FDA0003492412200000032
5. the compound of 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 a hydrogen atom.
6. A compound according to claim 1 or 2, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from phenyl, naphthyl, pyridyl, benzothiazolyl or benzopyrazolyl, wherein said phenyl, naphthyl, pyridyl, benzothiazolyl or benzopyrazolyl is optionally further substituted with one or more substituents selected from halogen, hydroxy, alkyl, alkoxy, cycloalkyl or-NR 6 R 7 Wherein said alkyl or alkoxy is optionally further substituted by one or more substituents selected from halogen or-NR 6 R 7 Substituted with the substituent(s); wherein said halogen is preferably fluorine;
R 6 and R 7 Each independently selected from a hydrogen atom or an alkyl group, wherein the alkyl group is preferably a methyl group.
7. The compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 Selected from:
Figure FDA0003492412200000033
Figure FDA0003492412200000041
8. the compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 3 Selected from absent or hydrogen atoms.
9. A compound according to claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein ring B is selected from:
Figure FDA0003492412200000042
R 3 is defined as in claim 1.
10. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 4 Selected from hydrogen, halogen, alkyl, alkoxy, haloalkyl or haloalkoxy, R 4 Preferably halogen, more preferably fluorine or chlorine.
11. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein L is selected from-CH 2 -、-CH 2 CH 2 -or-CH (CH) 3 )-。
12. A compound according to claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein said compound is:
Figure FDA0003492412200000043
13. a process for the preparation of a compound of general formula (I) according to claim 1, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, which process comprises:
Figure FDA0003492412200000051
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 B, R 1 ~R 3 X, Y, L, G, E and n are as defined in claim 1The above-mentioned processes are described.
14. A pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 12, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
15. Use of a compound according to any one of claims 1 to 12, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, for the preparation of a K-Ras gtpase inhibitor, wherein the K-Ras gtpase inhibitor is preferably a KRAS G12C inhibitor.
16. Use of a compound according to any one of claims 1 to 12, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, for the manufacture of a medicament for the treatment of a disease mediated by KRAS mutation, wherein the disease mediated by KRAS mutation is selected from cancer, wherein the cancer is 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, cutaneous squamous cell carcinoma, cervical cancer, testicular germ cell cancer, preferably pancreatic cancer, colorectal cancer and lung cancer, wherein the KRAS mutation is preferably KRAS G12C mutation.
17. Use of a compound according to any one of claims 1 to 12, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, 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 cancer, preferably pancreatic cancer, colorectal cancer and lung cancer; wherein the lung cancer is preferably non-small cell lung cancer.
CN202210100843.3A 2021-05-27 2022-01-27 Heteroaromatic ring derivative and preparation method and application thereof Pending CN115403575A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021105870082 2021-05-27
CN202110587008 2021-05-27

Publications (1)

Publication Number Publication Date
CN115403575A true CN115403575A (en) 2022-11-29

Family

ID=84157790

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210100843.3A Pending CN115403575A (en) 2021-05-27 2022-01-27 Heteroaromatic ring derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN115403575A (en)

Similar Documents

Publication Publication Date Title
RU2726115C1 (en) Pyridopyrimidine inhibitors cdk2/4/6
CN113286794A (en) KRAS mutein inhibitors
WO2022206723A1 (en) Heterocyclic derivative, and preparation method therefor and use thereof in medicine
CN112552295A (en) KRAS mutein inhibitors
WO2015077193A1 (en) Inhibitors of lysine methyl transferase
CN112300153B (en) Heterocyclic compound, pharmaceutical composition and application
TW201607930A (en) 5-amino-4-carbamoyl-pyrazole compounds as selective and irreversible T790M over WT EGFR kinase inhibitors and the use thereof
CN116249683A (en) Deuteromethyl substituted pyrazinopyrazinoquinolinone derivative, preparation method and application thereof in medicine
CN113939518A (en) Fused tricyclic compounds as kinase inhibitors
WO2023025116A1 (en) Heterocyclic derivative, preparation method therefor and use thereof in medicine
CN112839944B (en) Compounds and methods for treating rabies
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
CN116390923A (en) Heterocyclic derivative and preparation method and application thereof
WO2022037631A1 (en) Heterocyclic derivative and preparation method therefor and use thereof
CN116514846A (en) Heterocyclic derivative, preparation method and medical application thereof
CN115403575A (en) Heteroaromatic ring derivative and preparation method and application thereof
CN113166148B (en) Heterocyclic compounds as CDK-HDAC dual pathway inhibitors
WO2022198904A1 (en) Key intermediate of kras inhibitor and preparation method therefor
CN106349180B (en) 4, 5-diphenyl isoxazole derivative and preparation method and application thereof
CN115557949A (en) Tetracyclic derivative, preparation method and medical application thereof
CN115611898A (en) Tetracyclic derivative, preparation method and medical application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication