CN116162099A - Heterocyclic derivative and preparation method and application thereof - Google Patents

Heterocyclic derivative and preparation method and application thereof Download PDF

Info

Publication number
CN116162099A
CN116162099A CN202211465937.7A CN202211465937A CN116162099A CN 116162099 A CN116162099 A CN 116162099A CN 202211465937 A CN202211465937 A CN 202211465937A CN 116162099 A CN116162099 A CN 116162099A
Authority
CN
China
Prior art keywords
alkyl
group
cycloalkyl
heterocyclyl
halogen
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
CN202211465937.7A
Other languages
Chinese (zh)
Inventor
黄贤贵
晏青燕
陈友喜
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 CN116162099A publication Critical patent/CN116162099A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/08Bridged systems

Abstract

The invention relates to heterocyclic derivatives, a preparation method and medical application thereof. In particular, the invention relates to heterocyclic derivatives shown in a general formula (I), a preparation method and pharmaceutically acceptable salts thereof, and application of the heterocyclic derivatives as therapeutic agents, particularly as KRAS G12D inhibitors, wherein each substituent in the general formula (I) or the general formula (I) has the same definition as in the specification.

Description

Heterocyclic derivative and preparation method and application thereof
Technical Field
The invention relates to a heterocyclic derivative, a preparation method thereof, a pharmaceutical composition containing the derivative and application of the heterocyclic derivative as a therapeutic agent, in particular to an inhibitor of K-Ras GTPase.
Background
RAS represents a closely related group of monomeric globular proteins (21 kDa molecular weight) with 189 amino acids and which are associated with the plasma membrane and bind GDP or GTP. Under normal developmental or physiological conditions, the RAS is activated by receiving 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, the on/off state of the RAS protein is determined by nucleotide binding, the active signaling conformation binds GTP, and the inactive conformation binds GDP. When the RAS contains bound GDP, it is in a dormant or quiescent or off state and is "inactive". When cells are exposed to certain growth promoting stimuli in response, the RAS is induced to convert the bound GDP to GTP. As GTP is bound, the RAS is "on" and is able to interact with and activate other proteins (its "downstream targets"). RAS proteins themselves have a very low inherent ability to hydrolyze GTP back to GDP and thereby turn themselves into an off state. Conversion of the RAS to shut down requires exogenous proteins called Gtpase Activating Proteins (GAPs) that interact with the RAS and greatly promote the conversion of GTP to GDP. Any mutation in the RAS that affects its ability to interact with GAP or convert GTP back to GDP will result in prolonged activation of the protein and thus produce a prolonged signal to the cell that signals it to continue growth and division. These signals may therefore cause cell growth and division, and overactivated RAS signaling may ultimately lead to cancer.
Structurally, RAS proteins contain a G domain responsible for enzymatic activity of the ras— guanine nucleotide binding and hydrolysis (gtpase reaction). It also includes a C-terminal extension region, called CAAX box, which can be post-translationally modified and targets the protein to a membrane. The G domain is approximately 21-25kDa in size and contains a phosphate binding loop (P-loop). The P-loop represents the pocket in the protein that binds the nucleotide and is a rigid part of the domain with conserved amino acid residues that are necessary for nucleotide binding and hydrolysis (glycine 12, threonine 26 and lysine 16). The G domain also contains so-called switch I regions (residues 30-40) and switch II regions (residues 60-76), which are both dynamic parts of the protein, often denoted as "spring-loaded" mechanisms due to the ability of the dynamic parts to switch between resting and loaded states. The main interaction is the hydrogen bond formed by threonine-35 and glycine-60 with the gamma-phosphate of GTP, which maintains their active conformation in switch I and switch II, respectively. After hydrolysis of GTP and release of phosphate, both relax to an inactive GDP conformation.
Among RAS family members, oncogenic mutations are most common in KRAS (85%), while NRAS (12%) and HRAS (3%) are less common. KRAS mutations are prevalent in three major deadly cancer types in the united states: pancreatic cancer (95%), colorectal cancer (45%) and lung cancer (25%), KRAS mutations are also found in other cancer types including multiple myeloma, uterine cancer, cholangiocarcinoma, gastric cancer, bladder cancer, diffuse large B-cell lymphoma, rhabdomyosarcoma, cutaneous squamous cell carcinoma, cervical cancer, testicular germ cell carcinoma, cardiac myxoma, large intestine tumor, prostate cancer, hepatocellular carcinoma, chondrosarcoma, multiple myeloma, seminoma, malignant melanoma, adrenal neuroblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, etc., while rarely found (< 2%) in breast cancer, ovarian cancer and brain cancer. 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 the frequency of specific allelic mutations comes mostly from classical smoking-induced classical 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 driving oncogenic mutations in NSCLC, including EGFR, ALK, ROS, RET, and BRAF, indicating the uniqueness of KRAS mutations in lung cancer. While at the same time KRAS mutations often coincide with certain co-mutations, such as STK11, KEAP1 and TP53, which in cooperation with the mutated RAS transform the cells into highly malignant and invasive tumor cells.
Three RAS oncogenes constitute the most frequently mutated gene family in human cancers. It is disappointing that despite thirty years of research efforts, there is still no clinically effective anti-RAS therapy, and targeting the gene using small molecules is a challenge. Accordingly, there is an urgent need in the art for small molecules for targeting the RAS (e.g., K-RAS, H-RAS, and/or N-RAS) and using the same to treat a variety of diseases, such as cancer.
At present, the clinical development of KRAS G12D inhibitor is in vigorous competition at home and abroad, wherein KRAS G12D inhibitor MRTX-1133 developed by Mirati Therapeutics Inc company already enters a preclinical stage and is used for treating diseases such as large intestine tumor, non-small cell lung cancer, pancreatic cancer and the like. There are a few published KRAS G12D inhibitor patent applications including WO2021041671 by Mirati Therapeutics Inc. Although research and use of KRas G12D inhibitors has advanced somewhat, there is still a tremendous space for improvement and there is still a need to continue to research and develop new KRas G12D inhibitors.
Disclosure of Invention
The invention aims to provide a heterocyclic derivative shown in a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof:
Figure BDA0003956217760000021
wherein:
Figure BDA0003956217760000022
selected from single bonds or double bonds as needed so that each atom thereof assumes a normal valence state; ring a is selected from cycloalkyl, 5-6 membered monocyclic heterocyclyl, 8-10 membered saturated bicyclic heterocyclyl, 8-10 membered saturated tricyclic heterocyclyl, aryl, heteroaryl, or fused rings;
ring B is selected from aryl, heteroaryl or fused ring;
ring C is selected from 4-8 membered monocyclic heterocyclic groups, wherein the heterocyclic groups contain at least 2 nitrogen atoms;
the conditions are as follows: ring C is not selected from piperazinyl;
Q 1 selected from N or CR a
Q 2 Selected from N, C or CR a
Y is selected from the group consisting of bond, -O-or-NR b
X 1 、X 2 Each independently selected from N, CR c Or CR (CR) d R e
R a The same or different are each independently selected from hydrogen atom, halogen, alkyl, alkoxy or cyano; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
R b selected from hydrogen atoms or alkyl groups;
R c 、R d and R is e The same or different, each independently selected from hydrogen, halogen, cyano, alkyl or alkoxy; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
R 1 Absence or two R 1 Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group; wherein said cycloalkyl or heterocyclyl is optionally further substituted with one or more R B Substitution;
R B the same or different, each independently selected from hydrogen, halogen, cyano, alkyl or alkoxy; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy
Alternatively, two R B Together with the same carbon atom to which it is attached, form-C (=o) -;
R 4 the same or different are each independently selected from a hydrogen atom, halogen, hydroxy, alkyl or alkoxy, preferably a hydrogen atom or alkyl;
R A the same or different, each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
alternatively, two R A Together with the attached carbon atom, form a cycloalkyl group; preferably cyclopropyl;
with the proviso that when ring A is selected from cycloalkyl or 5-6 membered monocyclic heterocyclyl, at least one R A Not selected as hydrogen atoms;
R 2 identical OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, alkenyl, alkynyl, 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 The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted with one OR more substituents selected from alkyl, halo, 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 Is substituted by a substituent of (2);
R 3 identical OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, alkenyl, alkynyl, 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 The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted with one OR more substituents selected from alkyl, halo, 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 Is substituted by a substituent of (2);
R 5 each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group, wherein the alkyl group, cycloalkyl group, heterocyclic group, aryl group, or heteroaryl group is optionally further substituted with one or more groups selected from hydroxy, halogen, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl group, heterocyclic group, aryl group, 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 Is substituted by a substituent of (2);
R 6 and R is 7 Each independently selected from the group consisting of hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein said alkyl, alkoxy, cycloalkyl,the heterocyclyl, aryl or heteroaryl is optionally further substituted with one or more groups selected from hydroxy, halo, 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 Is substituted by a substituent of (2);
alternatively, R 6 And R is 7 Together with the atoms to which they are attached form a 4-8 membered heterocyclic group, wherein the 4-8 membered heterocyclic group contains one or more of N, O or S (O) r And said 4-8 membered heterocyclyl is optionally further substituted with one or more substituents selected from 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 Is substituted by a substituent of (2);
R 8 、R 9 and R is 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, carboxyl, or carboxylate;
m is selected from 0, 1 or 2;
n is selected from 0, 1, 2 or 3;
p is selected from 0, 1, 2 or 3;
r is 0, 1 or 2.
In a preferred embodiment of the present invention, a compound of formula (I) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof is a compound of formula (II) or (III) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof:
Figure BDA0003956217760000041
wherein: r is R c The same or different, each independently selected from hydrogen, halogen, cyano or cyanoalkyl; wherein the halogen is preferably fluorine or chlorine;
ring a, ring B, ring C, Q 1 、Y、R 1 ~R 4 、R A The definitions of m, n and p are as described in the general formula (I).
In a preferred embodiment of the present invention, a compound of formula (I) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof is a compound of formula (IV):
Figure BDA0003956217760000051
wherein: ring a, ring B, ring C, Q 1 、Y、R 1 ~R 4 、R A The definitions of m, n and p are as described in the general formula (I).
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:
Figure BDA0003956217760000052
selected from->
Figure BDA0003956217760000053
Wherein R is B The definition of (a) is as described in a general formula (I);
in a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:
Figure BDA0003956217760000054
Selected from the following groups:
Figure BDA0003956217760000055
in a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein Q 1 Is N.
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein Y is-O-.
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein ring A is selected from 5-6 membered monocyclic heterocyclyl or 8-10 membered saturated bicyclic heterocyclyl.
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein two R A Together with the attached carbon atom, form a cycloalkyl group; cyclopropyl is preferred.
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein R A Each independently selected from a hydrogen atom, a halogen, a hydroxy group, or a hydroxymethyl group.
The conditions are as follows: when ring A is selected from cycloalkyl or 5-6 membered monocyclic heterocyclyl, at least one R A Not selected as hydrogen atoms;
in a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein R 2 Selected from alkyl, halogen, alkoxy or = O, wherein the alkyl is preferably methyl and the halogen is preferably fluoro.
In a preferred embodiment of the invention, a compound of the general formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein
Figure BDA0003956217760000061
Selected from: />
Figure BDA0003956217760000062
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:
ring B is selected from phenyl, naphthyl, pyridyl, quinolinyl, isoquinolinyl, indolyl, indazolyl, benzothiazolyl, tetrahydronaphthyl,
Figure BDA0003956217760000063
Ring B is preferably naphthyl.
In a preferred embodiment of the invention, a compound of formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:
R 3 the same or different, each independently selected from hydrogen atom, alkyl, halogen, alkoxy, alkynyl, hydroxy, amino, hydroxyalkyl, haloalkyl or haloalkoxy;
R 3 Preferably a hydrogen atom, methyl, fluorine, chlorine, bromine, iodine, hydroxyl, amino, hydroxymethyl or ethynyl.
In a preferred embodiment of the invention, a compound of the general formula (I), (II), (III) or (IV) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein
Figure BDA0003956217760000064
Selected from the following groups:
Figure BDA0003956217760000071
typical compounds of the present invention include, but are not limited to:
Figure BDA0003956217760000072
or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
Note that: if there is a difference between the drawn structure and the name given to the structure, the drawn structure will be given greater weight.
In another aspect, the present invention provides a pharmaceutical composition comprising an effective amount of a compound of formula (I), (II), (III) or (IV), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
In another aspect, the invention provides a method of inhibiting KRas G12D enzyme, wherein the method comprises administering to a patient a pharmaceutical composition comprising an effective amount of a compound of formula (I), (II), (III) or (IV), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
The invention also provides the use of a compound of formula (I), (II), (III) or (IV), or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the manufacture of a medicament for the treatment of a disease mediated by a KRas G12D mutation, wherein the disease mediated by a KRas G12D mutation is selected from cancer, wherein the cancer is preferably selected from cardiac myxoma, lung cancer, stomach cancer, large intestine tumour, rectal cancer, pancreatic cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, cholangiocarcinoma, chondrosarcoma, multiple myeloma, uterine cancer, cervical cancer, seminoma, malignant melanoma, cutaneous squamous cell carcinoma, adrenoneuroblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, more preferably pancreatic cancer, large intestine tumour, rectal cancer and lung cancer; wherein the lung cancer is selected from non-small cell lung cancer or small cell lung cancer.
In another aspect, the invention provides the use of a compound of formula (I), (II), (III) or (IV), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the preparation of a KRas G12D inhibitor.
Another aspect of the invention relates to a method for preventing and/or treating KRas G12D mutation mediated diseases comprising administering to a patient a therapeutically effective dose of a compound of formula (I), (II), (III) or (IV) or a tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The invention also provides the use of a compound of general formula (I), (II), (III) or (IV), or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the manufacture of a medicament for the treatment of cancer, wherein the cancer is preferably selected from the group consisting of cardiac myxoma, lung cancer, stomach cancer, large intestine tumor, rectal cancer, pancreatic cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, cholangiocarcinoma, chondrosarcoma, multiple myeloma, uterine cancer, cervical cancer, seminoma, malignant melanoma, cutaneous squamous cell carcinoma, adrenoneuroblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, more preferably pancreatic cancer, large intestine tumor, rectal 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, intraocular, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intradermal, intraperitoneal, subcutaneous, subcuticular or by inhalation. Pharmaceutical compositions containing the active ingredient may be in a form suitable for oral administration, for example, as tablets, troches, 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-dose form and may be prepared by any method well known in the pharmaceutical arts. The amount of active ingredient that can be combined with the carrier material to produce a single dosage form can 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 is capable of producing 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 admixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers or propellants which may be required.
When the compounds of the invention are administered to humans and animals in the form of a medicament, the compounds may be provided alone or in the form of a pharmaceutical composition containing the active ingredient in combination with a pharmaceutically acceptable carrier, for example 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 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; (19) ethanol; (20) phosphate buffer solution; (21) Cyclodextrins, e.g., targeting ligands attached to nanoparticles, e.g., accursinTM; and (22) other non-toxic compatible substances 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, cysteamine 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 chelators such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid and the like. Solid dosage forms (e.g., capsules, dragees, powders, granules and the like) may 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) Binders, such as carboxymethyl cellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerin; (4) Disintegrants, for example agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) dissolution retarders, such as paraffin; (6) an absorption accelerator, such as a quaternary ammonium compound; (7) Humectants, such as cetyl alcohol and glycerol monostearate; (8) absorbents such as kaolin and bentonite; (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 form may contain inert diluents commonly used in the art, such as water or other solvents; solubilizing agents and emulsifiers, for example, ethanol, isopropanol, 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 also contain suspending agents, such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum hydroxide oxide, bentonite, agar-agar, and tragacanth, and mixtures thereof.
In addition to the active compounds, ointments, pastes, creams and gels may contain excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
In addition to the active compounds, the powders and sprays can also contain 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:
"bond" means that the indicated substituent is absent and that the two end portions of the substituent are directly linked to form a bond.
"alkyl" when taken as a group or part of a group is meant to include 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.
"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, vinyl, 1-propenyl, 2-propenyl, 1-, 2-or 3-butenyl, and the like. Alkenyl groups may be optionally substituted or unsubstituted.
"alkynyl" refers to an aliphatic hydrocarbon group containing one carbon-carbon triple bond, which may be straight or branched. Preferably is C 2 -C 10 More preferably C 2 -C 6 Alkynyl, most preferably C 2 -C 4 Alkynyl groups. 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 carbocycles. Preferably C 3 -C 12 Cycloalkyl, more preferably C 3 -C 8 Cycloalkyl, most preferably C 3 -C 6 Cycloalkyl groups. Examples of monocyclic cycloalkyl groups 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, two or more cyclic structure, and monocyclic polycyclic groups sharing one carbon atom (called 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 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spiro group, a double spiro group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, preferably single spiro group and double spirocycloalkyl group, 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 an all-carbon polycyclic group having 5 to 18 members, two or more cyclic structures sharing a pair of carbon atoms with each other, one or more of the 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 members, more preferably 7 to 10 members. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. 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 tetradecahydrophenanthryl.
"bridged cycloalkyl" means an aromatic system having 5 to 18 members, containing two or more cyclic structures, sharing two all-carbon polycyclic groups with one another that are not directly attached to a carbon atom, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron, preferably 6 to 12 members, more preferably 7 to 10 members. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged cycloalkyl" include, but are not limited to: (1 s,4 s) -bicyclo [2.2.1] heptyl, bicyclo [3.2.1] octyl, (1 s,5 s) -bicyclo [3.3.1] nonyl, bicyclo [2.2.2] octyl, and (1 r,5 r) -bicyclo [3.3.2] decyl.
"heterocyclyl", "heterocycle" or "heterocyclic" are used interchangeably herein to refer to a saturated or partially unsaturated monocyclic or polycyclic non-aromatic cyclic hydrocarbon substituent in which one or more of the ring-forming atoms is a heteroatom, such as an oxygen, nitrogen, sulfur atom, or the like. Preferably having a 5 to 7 membered single 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 "monocyclic 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. Polycyclic heterocyclyl groups include fused, bridged and spiro heterocyclic groups. The heterocyclic group may be substituted or unsubstituted.
"spiroheterocyclyl" refers to a 5-to 18-membered, two or more cyclic structure, polycyclic group having single rings sharing one atom with each other, containing 1 or more double bonds in the ring, but no ring having a completely conjugated pi-electron aromatic system in which 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 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spiro heterocyclic group, a double spiro heterocyclic group or a multiple spiro heterocyclic group according to the number of common spiro atoms between rings, and preferably a single spiro heterocyclic group and a double spiro heterocyclic group. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered single spiro heterocyclic 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 the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system in which 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 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of "fused heterocyclyl" include, but are not limited to: octahydropyrrolo [3,4-c ] ]Pyrrolyl, octahydro-1H-isoindolyl, 3-azabicyclo [3.1.0 ]]Hexyl, octahydrobenzo [ b ]][1,4]Dioxin (dioxane) or
Figure BDA0003956217760000111
"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 of the rings may contain one or more double bonds, but none of the rings has a complete ring structureConjugated pi-electron aromatic systems in which 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 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged heterocyclyl" include, but are not limited to: 2-azabicyclo [2.2.1]Heptyl, 2-azabicyclo [2.2.2]Octyl and 2-azabicyclo [3.3.2]And (3) a decyl group.
"aryl" refers to a carbocyclic aromatic system containing one or two rings, wherein the rings may be linked together in a fused manner. 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, most preferably naphthyl. Aryl groups may be substituted or unsubstituted.
"heteroaryl" refers to an aromatic 5-to 6-membered monocyclic or 8-to 10-membered bicyclic ring, which may contain 1 to 4 atoms selected from nitrogen, oxygen and/or sulfur. Preferred bicyclic heteroaryl groups, examples of "heteroaryl" 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, benzothienyl, benzimidazolyl, indolyl, isoindolyl, 1, 3-dioxo-isoindolyl, quinolinyl, indazolyl, benzisothiazolyl, benzoxazolyl, benzisoxazolyl, benzoisoxazolyl, benzothiophenyl, benzofuranyl, and the like,
Figure BDA0003956217760000121
/>
Heteroaryl groups may be substituted or unsubstituted.
"fused ring" means a polycyclic group having two or more cyclic structures sharing a pair of atoms with each other, one or more rings being capable ofAromatic systems containing one or more double bonds, but at least one ring not having fully conjugated pi electrons, wherein the ring atoms are selected from 0, one or more of 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 double-or triple-ring fused ring, wherein the double-ring fused ring is preferably a fused ring of an aryl or heteroaryl group and a monocyclic heterocyclic group or a monocyclic cycloalkyl group. Preferably 7 to 14 membered, more preferably 8 to 10 membered. Examples of "fused rings" include, but are not limited to:
Figure BDA0003956217760000122
Figure BDA0003956217760000131
"alkoxy" refers to a group of (alkyl-O-). Wherein alkyl is as defined herein. C (C) 1 -C 6 Is preferably selected. Examples include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy and the like.
"haloalkyl" refers to a group wherein the alkyl is optionally further substituted with one or more halogens, where 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, where alkyl is as defined herein.
"hydroxymethyl" refers to a group that is optionally further substituted with one or more hydroxyl groups.
"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" refers to-CN.
"nitro" means-NO 2
"benzyl" means-CH 2 -phenyl.
"carboxy" means-C (O) OH.
"carboxylate" refers to-C (O) O-alkyl or-C (O) O-cycloalkyl, wherein alkyl, cycloalkyl are as defined above.
"DMSO" refers to dimethyl sulfoxide.
"BOC" refers to t-butoxycarbonyl.
"MOM" refers to methoxymethyl.
"Ts" refers to p-toluenesulfonyl.
"T3P" refers to propyl phosphoric anhydride.
"DPPA" refers to diphenyl azide phosphate.
"DEA" refers to diethylamine.
"X-PHOS Pd G2" refers to chloro (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II).
"RuPhos Pd G3" refers to sulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropyloxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II).
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"substituted" or "substituted" as used herein, unless otherwise indicated, means that the group may be substituted with one or more groups selected from the group consisting of: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, alkenyl, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio,Amino, haloalkyl, hydroxyalkyl, carboxyl, 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 Is substituted by a substituent of (2);
R 5 selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group or a heteroaryl group, wherein the alkyl group, the cycloalkyl group, the heterocyclic group, the aryl group or the heteroaryl group is optionally further substituted with one or more groups selected from a hydroxyl group, a halogen group, a nitro group, a cyano group, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a cycloalkyl group, a heterocyclic group, an aryl group, a heteroaryl group, =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 Is substituted by a substituent of (2);
R 6 and R is 7 Each independently selected from a hydrogen atom, 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 groups selected from 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 Is substituted by a substituent of (2);
alternatively, R 6 And R is 7 Together with the atoms to which they are attached form a 4-8 membered heterocyclic group, wherein the 4-8 membered heterocyclic group contains one or more of N, O or S (O) r And said 4-8 membered heterocyclyl is optionally further substituted with one or more substituents selected from 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 Is substituted by a substituent of (2);
R 8 、R 9 and R is 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, carboxyl, 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 (attopiomers) 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 stereoisomers (e.g., diastereomers, enantiomers and atropisomers and geometric (conformational) isomeric forms of such structures, e.g., the R and S configurations of each asymmetric center, (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 the original biological activity and are suitable for pharmaceutical use. The pharmaceutically acceptable salts of the compounds represented by formula (I) may be metal salts, amine salts with suitable acids.
"pharmaceutical composition" means a mixture comprising one or more of the compounds described herein or a physiologically acceptable salt or prodrug thereof, and other chemical components, such as physiologically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
Synthesis method of compound of the invention
In order to achieve the purpose of the invention, the invention adopts the following technical scheme:
the preparation method of the compound shown in the general formula (I) or the stereoisomer, the tautomer or the pharmaceutically acceptable salt thereof comprises the following steps:
Figure BDA0003956217760000151
carrying out Suzuki coupling reaction on the compound shown in the general formula (IA) and the compound shown in the general formula (IB) under the action of a palladium catalyst and an alkaline reagent to obtain a compound shown in the general formula (IC); further deprotecting the compound of formula (IC), optionally further deprotecting the protecting group on ring B, to give a compound of formula (I);
wherein:
x is a leaving group, preferably chlorine;
PG is a protecting group, preferably t-butoxycarbonyl;
m is selected from-B (OH) 2 、-BF 3 K or
Figure BDA0003956217760000152
Ring a, ring B, R 1 ~R 3 、X 1 、X 2 、Q 1 、Q 2 、Y、R A The definitions of m and n are as described in the general formula (I).
Detailed Description
The invention will be further described with reference to the following examples, which are not intended to limit the scope of the invention.
Examples
The preparation of representative compounds represented by formula (I) and related structural identification data are presented in the examples. It should be noted that the following examples are for illustrationThe invention is not limited thereto. 1 HNMR spectra were determined using a Bruker instrument (400 MHz) and chemical shifts were expressed in ppm. Tetramethylsilane internal standard (0.00 ppm) was used. 1 HNMR representation method: s=singlet, d=doublet, t=triplet, m=multiplet, br=broadened, dd=doublet of doublet, dt=doublet of triplet. If 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.
Thin Layer Chromatography (TLC) using tobacco stage yellow sea HSGF254 or Qingdao GF254 silica gel plate
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.
Column chromatography generally uses tobacco stand yellow sea silica gel 200-300 mesh silica gel as a carrier.
In the following examples, unless otherwise indicated, all temperatures are in degrees celsius and unless otherwise indicated, various starting materials and reagents are either commercially available or synthesized according to known methods, all of which are used without further purification and unless otherwise indicated, commercially available manufacturers include, but are not limited to, shanghai Haohong biological medicine technologies, shanghai Shaoshao reagent, shanghai Pico medicine, saen chemical technologies (Shanghai) and Shanghai Ling Kai medicine technologies, and the like.
CD 3 OD: deuterated methanol.
CDCl 3 : deuterated chloroform.
DMSO-d 6 : deuterated dimethyl sulfoxide.
The examples are not particularly described, and the solution in the reaction is an aqueous solution.
Purifying the compound using an eluent system 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: methylene chloride and methanol systems; c: dichloromethane and ethyl acetate system, D: dichloromethane and ethanol, wherein the volume ratio of the solvent is different according to the polarity of the compound, and small amount of acidic or alkaline reagent can be added for the conditions such as acetic acid or triethylamine.
Room temperature: 20-30 ℃.
Example 1
4-(4-(3,7-diazabicyclo[3.3.1]nonan-3-yl)-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-7-yl)-5-fluoronaphthalen-2-ol
4- (4- (3, 7-diazabicyclo [3.3.1] nonan-3-yl) -8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Figure BDA0003956217760000171
First step
tert-butyl 7-(2,7-dichloro-8-fluoropyrido[4,3-d]pyrimidin-4-yl)-3,7-diazabicyclo[3.3.1]nonane-3-
carboxylate
7- (2, 7-dichloro-8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester
2,4, 7-trichloro-8-fluoropyrido [4,3-d ] pyrimidine 1a (223.11 mg, 883.73. Mu. Mol, prepared according to WO 2020146613) and 3, 7-diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester 1b (200 mg, 883.73. Mu. Mol, commercially available) were added to dichloromethane (5 mL), cooled to-40 ℃, N-diisopropylethylamine (571.06 mg,4.42mmol, 793.14. Mu. L) was added dropwise, and the reaction was allowed to react at-40℃for 20 minutes, and LCMS detection was complete. The reaction mixture was quenched with water (10 mL), extracted with methylene chloride (10 mL. Times.3), 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: A system) to give tert-butyl 7- (2, 7-dichloro-8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylate 1c (390 mg, 881.73. Mu. Mol) in 99.77% yield.
MS m/z(ESI):442.0[M+H] +
Second step
tert-butyl 7-(7-chloro-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-
yl) methoxy) pyrido [4,3-d ] pyrimid-4-yl) -3,7-diazabicyclo [3.3.1] non-3-carboxylate 7- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3,7 ]
Diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester
7- (2, 7-dichloro-8-fluoropyrido [4, 3-d)]Pyrimidin-4-yl) -3,7-diazabicyclo [3.3.1]Nonane-3-carboxylic acid tert-butyl ester 1c (397.80 mg, 899.37. Mu. Mol), ((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methanol 1d (214.77 mg,1.35mmol, prepared in accordance with published patent WO 2020146613), N-diisopropylethylamine (348.70 mg,2.70mmol, 484.31. Mu.L) and molecular sieves
Figure BDA0003956217760000181
Form (500 mg, 899.37. Mu. Mol) was added sequentially to 1, 4-dioxane (4.52 mL), the temperature was raised to 100℃under the protection of argon, the reaction was allowed to proceed for 18 hours, ((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methanol 1d (214.77 mg,1.35 mmol) was added to the reaction mixture, the reaction was continued at 100℃for 5 hours, and the completion of the reaction was detected by LCMS. The reaction solution was cooled to room temperature, the molecular sieve was removed by filtration, the molecular sieve was eluted with ethyl acetate (50 mL), the filtrate was collected, the organic phase was washed with water (20 mL), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (eluent: B system) to give 7- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4, 3-d) ]Pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1]Nonane-3-carboxylic acid tert-butyl ester 1e (400 mg, 707.90. Mu. Mol) yield 78.71%.
MS m/z(ESI):565.0[M+H] +
Third step
tert-butyl 7-(8-fluoro-7-(8-fluoro-3-(methoxymethoxy)naphthalen-1-yl)-2-(((2R,7aS)-2-
fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,7-
diazabicyclo[3.3.1]nonane-3-carboxylate
7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester
7- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester 1e (204.13 mg, 361.26. Mu. Mol), 2- (8-fluoro-3-methoxymethoxy) naphthalen-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 1f (150 mg, 451.57. Mu. Mol, prepared according to published patent WO2021041671A 1), potassium phosphate (287.20 mg,1.35 mmol) and methanesulfonic acid [ n-butylbis (1-adamantyl) phosphine ] (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (98.80 mg, 135.47. Mu.) were added to tetrahydrofuran (8 mL), and the reaction was allowed to stand for 3 times at a temperature of 8℃until 10% of S was not completed when the reaction was detected. The reaction mixture was directly dried by spin-drying, and the obtained residue was separated and purified by silica gel column chromatography (eluent: system B) to give 1g (230 mg, 313.01. Mu. Mol) of tert-butyl 7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylate, yield 86.64%.
MS m/z(ESI):735.0[M+H] +
Fourth step
4-(4-(3,7-diazabicyclo[3.3.1]nonan-3-yl)-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-7-yl)-5-fluoronaphthalen-2-ol
4- (4- (3, 7-diazabicyclo [3.3.1] nonan-3-yl) -8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 7-diazabicyclo [3.3.1] nonane-3-carboxylic acid tert-butyl ester 1g (230 mg, 313.01. Mu. Mol) was dissolved in acetonitrile (2 mL), cooled in an ice water bath, and an ethyl acetate solution of hydrogen chloride (4M, 0.5 mL) was added thereto, stirred under ice water bath for 1 hour, and a solid was precipitated in the reaction solution to complete the LCMS detection reaction. Filtration, rinsing with ethyl acetate (10 mL), collecting the solid, placing it in a 25mL reaction flask, adding saturated aqueous sodium bicarbonate to adjust ph=8, stirring for 0.5 hours, filtering again, rinsing with water (10 mL), collecting the solid, freeze drying to give 4- (4- (3, 7-diazabicyclo [3.3.1] nonan-3-yl) -8-fluoro-2- (((2 r,7 as) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol 1 (107 mg,181.16 μmol), yield 57.88%.
MS m/z(ESI):591.0[M+H] +
Example 2
4-(4-(3,6-diazabicyclo[3.2.2]nonan-3-yl)-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-7-yl)-5-fluoronaphthalen-2-ol
4- (4- (3, 6-diazabicyclo [3.2.2] nonan-3-yl) -8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Figure BDA0003956217760000191
First step
tert-butyl 3-(2,7-dichloro-8-fluoropyrido[4,3-d]pyrimidin-4-yl)-3,6-diazabicyclo[3.2.2]nonane-6-
carboxylate
3- (2, 7-dichloro-8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3,6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester
2,4, 7-trichloro-8-fluoropyrido [4,3-d ] pyrimidine 1a (524.30 mg,2.08 mmol) and 3,6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2a (470 mg,2.08mmol, prepared in accordance with published patent WO 2010028011) were added to dichloromethane (2 mL), cooled to-40℃and N, N-diisopropylethylamine (1.34 g,10.38 mmol) was added dropwise thereto, reacted at-40℃for 30 minutes, warmed to room temperature and stirred for 1 hour, and LCMS detection reaction was complete. The reaction solution was quenched with water (10 mL), extracted with methylene chloride (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the obtained residue was separated and purified by silica gel column chromatography (eluent: B system) to give 3- (2, 7-dichloro-8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3,6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2B (400 mg, 904.34. Mu. Mol) in 43.55% yield.
MS m/z(ESI):442.0[M+H] +
Second step
tert-butyl 3-(7-chloro-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-
yl) methoxy) pyrido [4,3-d ] pyrimid-4-yl) -3,6-diazabicyclo [3.2.2] non-6-carboxylate 3- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3,6 ]
Diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester
((2R, 7 aS) -2-Fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methanol 1d (107.98 mg, 678.25. Mu. Mol) was dissolved in tetrahydrofuran (6 mL), protected by argon, cooled to 0℃and sodium hydride (81.39 mg,2.03mmol,60% oil dispersion) was added thereto, stirred at 0℃for 0.5 hours, 3- (2, 7-dichloro-8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2b (300 mg, 678.25. Mu. Mol) was added dropwise thereto, and the reaction was allowed to proceed to room temperature for 2 hours. The reaction solution was quenched with ice water (10 mL), extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the resulting residue was separated and purified by silica gel column chromatography (eluent: B system) to give 3- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2c (300 mg, 530.92. Mu. Mol), yield 78.28%.
MS m/z(ESI):565.0[M+H] +
Third step
tert-butyl 3-(8-fluoro-7-(8-fluoro-3-(methoxymethoxy)naphthalen-1-yl)-2-(((2R,7aS)-2-
fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,6-
diazabicyclo[3.2.2]nonane-6-carboxylate
3- (8-fluoro-7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester
3- (7-chloro-8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2c (200 mg, 353.95. Mu. Mol) was added to a mixed solvent of water (0.2 mL) and 1, 4-dioxane (1 mL), 2- (8-fluoro-3-methoxymethoxy) naphthalen-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 1f (141.09 mg, 424.74. Mu. Mol), sodium carbonate (112.56 mg,1.06 mmol) and tetrakis (triphenylphosphine) palladium (40.90 mg, 35.39. Mu. Mol) were sequentially added, argon was replaced 3 times, reacted at 80℃for 2 hours, the reaction solution was added to ethyl acetate (10 mL), and the aqueous solution was concentrated by a silica gel column chromatography (10 mL), and the aqueous phase was concentrated by a dry column chromatography method was eluted with sulfuric acid to give a residual sodium solution: b system) to give 3- (8-fluoro-7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2d (100 mg, 136.09. Mu. Mol) in 38.44% yield.
MS m/z(ESI):735.0[M+H] +
Fourth step
4-(4-(3,6-diazabicyclo[3.2.2]nonan-3-yl)-8-fluoro-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-7-yl)-5-fluoronaphthalen-2-ol
4- (4- (3, 6-diazabicyclo [3.2.2] nonan-3-yl) -8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
3- (8-fluoro-7- (8-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-4-yl) -3, 6-diazabicyclo [3.2.2] nonane-6-carboxylic acid tert-butyl ester 2d (100 mg, 136.09. Mu. Mol) was dissolved in acetonitrile (1 mL), and an ethyl acetate solution of hydrogen chloride (4M, 0.5 mL) was added and stirred at room temperature for 1 hour, and the LCMS detection reaction was complete. The reaction solution was concentrated under reduced pressure, and the resulting residue was purified by preparative liquid phase separation (separation column AKZONOBEL Kromasil; 250X 21.2mm I.D.;5 μm,20mL/min; mobile phase A:0.05% TFA+H2O; mobile phase B: CH3 CN) to give 4- (4- (3, 6-diazabicyclo [3.2.2] nonan-3-yl) -8-fluoro-2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) pyrido [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol 2 (2.24 mg, 3.02. Mu. Mol) in 2.22% yield.
MS m/z(ESI):591.0[M+H] +
Biological evaluation
Test example 1 determination of the inhibitory Activity of the Compounds of the invention on p-ERK1/2 in AGS cells
The following methods were used to determine the p-ERK1/2 inhibitory activity of the compounds of the invention on AGS cells. The method uses an Advanced phospho-ERK1/2 (Thr 202/tyr 204) kit (cat No. 64 AERPEH) from Cisbio, and the detailed experimental procedure is referred to the kit instructions. AGS cells (containing KRAS G12D mutation) were purchased from the national academy of sciences of life sciences cell resource center.
The experimental procedure is briefly described as follows: AGS cells were cultured in F12K complete medium containing 10% fetal bovine serum, 100U penicillin and 100. Mu.g/mL streptomycin. AGS cells were plated in 96-well plates 40000 per well, with medium being complete medium, and incubated overnight in a 5% co2 incubator at 37 ℃. Test compounds were dissolved in DMSO to prepare 10mM stock solution, then diluted with F12K complete medium, 100uL of F12K complete medium containing the corresponding concentration of test compound was added to each well, the final concentration of test compound in the reaction system ranged from 1000nM to 0.015nM, the cells were discarded after 3 hours of incubation in a cell incubator, the cells were washed with ice-bath PBS, then 50 uL of 1 Xcell phospho/total protein lysis buffer (Advanced phospho-ERK1/2 kit component) was added to each well for lysis, and the 96 well plate was placed on ice for half an hour, followed by detection of the lysate with reference to the Advanced phospho-ERK1/2 (Thr 202/tyr 204) kit instructions. Finally, the fluorescence intensities of the wells at excitation wavelengths of 304nM, at which the emission wavelengths of 620nM and 665nM are measured on an microplate reader in TF-FRET mode, and the fluorescence intensity ratio of the wells 665/620 is calculated. By passing throughComparing the fluorescence intensity ratio with that of control group (0.1% DMSO), calculating the percent inhibition rate of the tested compound at each concentration, and performing nonlinear regression analysis on the numerical value-inhibition rate of the tested compound at the concentration by GraphPad Prism 5 software to obtain the IC of the compound 50 Values.
Preferred compounds of the invention have a pronounced inhibitory effect on the p-ERK1/2 activity in AGS cells, preferably IC of the compounds 50 <500nM, more optimized IC of the compound 50 <200nM。
Test example 2 assay of the Compounds of the invention for the inhibition of AsPC-1 cell proliferation
The following method was used to determine the effect of the compounds of the invention on AsPC-1 cell proliferation. AsPC-1 cells (containing KRAS G12D mutation) were purchased from Shanghai institute of life sciences cell resource center, academy of sciences of China 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 BDA0003956217760000221
Luminescent Cell Viability Assay kit (Promega, cat# G7573).
The experimental method is operated according to the steps of the instruction book of the kit, and is briefly described as follows: test compounds were prepared by first dissolving the test compounds in DMSO to prepare a 10mM stock solution, and then diluting the stock solution with medium to prepare test samples, wherein the final concentration of the compounds ranged from 1000nM to 0.015nM. Cells in the logarithmic growth phase were seeded at a density of 800 cells per well in 96-well cell culture plates and incubated overnight at 37℃in a 5% CO2 incubator, followed by additional incubation for 120 hours after the addition of the test compound. After the incubation was completed, a 50uL volume of CellTiter-Glo assay was added to each well, and after shaking for 5 minutes, the wells were allowed to stand for 10 minutes, followed by reading the Luminescence values of each well of the sample on a microplate reader using the Luminescence mode. The percent inhibition of compounds at each concentration point was calculated by comparison with the values of the control group (0.3% dmso), followed by nonlinear regression analysis of the compound concentration log-inhibition in GraphPad Prism 5 software to obtain IC compounds that inhibited cell proliferation 50 Values.
Preferred compounds of the invention have a pronounced inhibitory effect on AsPC-1 cell proliferation, preferably IC of the compounds 50 <500nM, more optimized IC of the compound 50 <200nM。

Claims (21)

1. A compound of formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
Figure FDA0003956217750000011
wherein:
Figure FDA0003956217750000012
selected from single bonds or double bonds as needed so that each atom thereof assumes a normal valence state; ring a is selected from cycloalkyl, 5-6 membered monocyclic heterocyclyl, 8-10 membered saturated bicyclic heterocyclyl, 8-10 membered saturated tricyclic heterocyclyl, aryl, heteroaryl, or fused rings;
ring B is selected from aryl, heteroaryl or fused ring;
ring C is selected from 4-8 membered monocyclic heterocyclic groups, wherein the heterocyclic groups contain at least 2 nitrogen atoms;
the conditions are as follows: ring C is not selected from piperazinyl;
Q 1 selected from N or CR a
Q 2 Selected from N, C or CR a
Y is selected from the group consisting of bond, -O-or-NR b
X 1 、X 2 Each independently selected from N, CR c Or CR (CR) d R e
R a The same or different are each independently selected from hydrogen atom, halogen, alkyl, alkoxy or cyano; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
R b selected from hydrogen atoms or alkyl groups;
R c 、R d and R is e The same or different, each independently selected from hydrogen, halogen, cyano, alkyl or alkoxy; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
R 1 Absence or two R 1 Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group; wherein said cycloalkyl or heterocyclyl is optionally further substituted with one or more R B Substitution;
R B the same or different, each independently selected from hydrogen, halogen, cyano, alkyl or alkoxy; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy or two R B Together with the same carbon atom to which it is attached, form-C (=o) -;
R 4 the same or different are each independently selected from a hydrogen atom, halogen, hydroxy, alkyl or alkoxy, preferably a hydrogen atom or alkyl;
R A the same or different, each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
alternatively, two R A Together with the attached carbon atom, form a cycloalkyl group; preferably cyclopropyl;
with the proviso that when ring A is selected from cycloalkyl or 5-6 membered monocyclic heterocyclyl, at least one R A Not selected as hydrogen atoms;
R 2 identical OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, alkenyl, alkynyl, 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 The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, arylOR heteroaryl optionally further substituted with one OR more substituents 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-S (O) r R 5 Is substituted by a substituent of (2);
R 3 identical OR different, each independently selected from hydrogen atom, alkyl, halogen, nitro, cyano, alkenyl, alkynyl, 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 The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl OR heteroaryl is optionally further substituted with one OR more substituents selected from alkyl, halo, 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 Is substituted by a substituent of (2);
R 5 each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group, wherein the alkyl group, cycloalkyl group, heterocyclic group, aryl group, or heteroaryl group is optionally further substituted with one or more groups selected from hydroxy, halogen, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl group, heterocyclic group, aryl group, 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 Is substituted by a substituent of (2);
R 6 and R is 7 Each independently selected from a hydrogen atom, 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 groups selected from 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 Is substituted by a substituent of (2);
alternatively, R 6 And R is 7 Together with the atoms to which they are attached form a 4-8 membered heterocyclic group, wherein the 4-8 membered heterocyclic group contains one or more of N, O or S (O) r And said 4-8 membered heterocyclyl is optionally further substituted with one or more substituents selected from 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 Is substituted by a substituent of (2);
R 8 、R 9 and R is 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, carboxyl, or carboxylate;
m is selected from 0, 1 or 2;
n is selected from 0, 1, 2 or 3;
p is selected from 0, 1, 2 or 3;
r is 0, 1 or 2.
2. The compound according to claim 1, which is a compound represented by the general formula (II) or (III) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
Figure FDA0003956217750000031
/>
wherein: r is R c The same or different, each independently selected from hydrogen, halogen, cyano or cyanoalkyl; wherein the halogen is preferably fluorine or chlorine;
ring a, ring B, ring C, Q 1 、Y、R 1 ~R 4 、R A The definitions of m, n and p are as defined in claim 1.
3. A compound according to claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, which is a compound represented by the general formula (IV):
Figure FDA0003956217750000032
wherein: ring a, ring B, ring C, Q 1 、Y、R 1 ~R 4 、R A The definitions of m, n and p are as defined in claim 1.
4. A compound according to any one of claims 1 to 3, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
Figure FDA0003956217750000033
selected from->
Figure FDA0003956217750000034
Wherein R is B Is defined asThe method of claim 1.
5. The compound of claim 4, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
Figure FDA0003956217750000035
Selected from the following groups:
Figure FDA0003956217750000041
6. a compound according to any one of claims 1 to 5, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein Q 1 Is N.
7. A compound according to any one of claims 1 to 6, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein Y is-O-.
8. A compound according to any one of claims 1 to 7, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein ring a is selected from 5-6 membered monocyclic heterocyclyl or 8-10 membered saturated bicyclic heterocyclyl.
9. A compound according to any one of claims 1 to 8, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein two R A Together with the attached carbon atom, form a cycloalkyl group; cyclopropyl is preferred.
10. A compound according to any one of claims 1 to 8, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R A Each independently selected from a hydrogen atom, a halogen, a hydroxy group, or a hydroxymethyl group.
The conditions are as follows: when ring A is selected from cycloalkyl or 5-6 membered monocyclic heterocyclyl, at least one R A Not selected as hydrogen atoms.
11. A compound according to any one of claims 1 to 10, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 Selected from alkyl, halogen, alkoxy or = O, wherein the alkyl is preferably methyl and the halogen is preferably fluoro.
12. A compound according to claims 8-11, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein
Figure FDA0003956217750000042
Selected from:
Figure FDA0003956217750000043
13. a compound according to any one of claims 1 to 12, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
ring B is selected from phenyl, naphthyl, pyridyl, quinolinyl, isoquinolinyl, indolyl, indazolyl, benzothiazolyl, tetrahydronaphthyl,
Figure FDA0003956217750000044
Ring B is preferably naphthyl.
14. A compound according to any one of claims 1 to 13, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
R 3 the same or different, each independently selected from hydrogen atom, alkyl, halogen, alkoxy, alkynyl, hydroxy, amino, hydroxyalkyl, haloalkyl or haloalkoxy;
R 3 preferably a hydrogen atom, methyl, fluorine, chlorine, bromine, iodine, hydroxyl, amino, hydroxymethyl or ethynyl.
15. A compound according to any one of claims 13 or 14, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein
Figure FDA0003956217750000051
Selected from the following groups: / >
Figure FDA0003956217750000052
16. The compound according to claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0003956217750000053
Figure FDA0003956217750000061
17. a pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 16, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
18. Use of a compound according to any one of claims 1 to 16, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 17, for the preparation of a KRas G12D inhibitor.
19. Use of a compound according to any one of claims 1 to 16, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 17, for the manufacture of a medicament for the treatment of a disease mediated by a KRas G12D mutation, wherein the disease mediated by a KRas G12D mutation is preferably selected from cancer, wherein the cancer is preferably selected from cardiac myxoma, lung cancer, stomach cancer, large intestine tumour, rectal cancer, pancreatic cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, cholangiocarcinoma, chondrosarcoma, multiple myeloma, uterine cancer, cervical cancer, seminoma, malignant melanoma, cutaneous squamous cell carcinoma, adrenoneuroblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, more preferably pancreatic cancer, large intestine tumour, rectal cancer and lung cancer.
20. Use of a compound according to any one of claims 1 to 16, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 17, for the manufacture of a medicament for the treatment of cancer, wherein the cancer is preferably selected from the group consisting of cardiac myxoma, lung cancer, stomach cancer, large intestine tumor, rectal cancer, pancreatic cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, cholangiocarcinoma, chondrosarcoma, multiple myeloma, uterine cancer, cervical cancer, seminoma, malignant melanoma, cutaneous squamous cell carcinoma, adrenoneuroblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, more preferably pancreatic cancer, large intestine tumor, rectal cancer and lung cancer.
21. The use according to claim 19 or 20, wherein the lung cancer is selected from non-small cell lung cancer or small cell lung cancer.
CN202211465937.7A 2021-11-24 2022-11-22 Heterocyclic derivative and preparation method and application thereof Pending CN116162099A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2021114026416 2021-11-24
CN202111402641 2021-11-24
CN202111569361 2021-12-21
CN2021115693614 2021-12-21

Publications (1)

Publication Number Publication Date
CN116162099A true CN116162099A (en) 2023-05-26

Family

ID=86420760

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211465937.7A Pending CN116162099A (en) 2021-11-24 2022-11-22 Heterocyclic derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116162099A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116157400A (en) * 2021-03-30 2023-05-23 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116157400A (en) * 2021-03-30 2023-05-23 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof

Similar Documents

Publication Publication Date Title
JP7394074B2 (en) therapeutic compounds
CN108699055B (en) Heterocyclic compounds as anti-cancer agents
CN116113632A (en) Heterocyclic derivative, preparation method and medical application thereof
CN116323625A (en) Heterocyclic derivative, preparation method and medical application thereof
AU2016360481A1 (en) Novel biphenyl compound or salt thereof
WO2012019427A1 (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
WO2015077193A1 (en) Inhibitors of lysine methyl transferase
CN116249683A (en) Deuteromethyl substituted pyrazinopyrazinoquinolinone derivative, preparation method and application thereof in medicine
CN112745335A (en) Tri-heterocyclic compound and application thereof
CN116546985A (en) Pyridopyrimidine derivative and preparation method and application thereof
WO2023001229A1 (en) Pyrimidocyclic derivative, preparation method therefor, and use thereof
CN116162099A (en) Heterocyclic derivative and preparation method and application thereof
CN116406363A (en) Tetracyclic derivative, preparation method and medical application thereof
CN113929681A (en) Tetracyclic derivative and preparation method and application thereof
CN116514846A (en) Heterocyclic derivative, preparation method and medical application thereof
CN113929676A (en) Pyridino-heterocyclic derivative and preparation method and application thereof
CN116390923A (en) Heterocyclic derivative and preparation method and application thereof
CN113166148B (en) Heterocyclic compounds as CDK-HDAC dual pathway inhibitors
WO2022037631A1 (en) Heterocyclic derivative and preparation method therefor and use thereof
CN116157400A (en) Heterocyclic derivative and preparation method and application thereof
CN116332938A (en) Fused tricyclic derivative and preparation method and application thereof
CN116157401A (en) Heterocyclic derivative and preparation method and application thereof
CN116113416A (en) Tetracyclic derivative, preparation method and medical application thereof
CN117466917A (en) Heterocyclic 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