CN116157400A - Heterocyclic derivative and preparation method and application thereof - Google Patents

Heterocyclic derivative and preparation method and application thereof Download PDF

Info

Publication number
CN116157400A
CN116157400A CN202280006425.6A CN202280006425A CN116157400A CN 116157400 A CN116157400 A CN 116157400A CN 202280006425 A CN202280006425 A CN 202280006425A CN 116157400 A CN116157400 A CN 116157400A
Authority
CN
China
Prior art keywords
group
alkyl
cancer
heterocyclyl
cycloalkyl
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
CN202280006425.6A
Other languages
Chinese (zh)
Inventor
陈友喜
龚亮
向清
毛文涛
赵雯雯
赵伟峰
程超英
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 CN116157400A publication Critical patent/CN116157400A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Abstract

The invention relates to heterocyclic derivatives, a preparation method and medical application thereof. In particular to heterocyclic derivatives shown in a general formula (I), a preparation method thereof and application thereof as therapeutic agents, particularly as KRAS G12D inhibitors, wherein the definition of each substituent in the general formula (I) is the same as that 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), which also includes a C-terminal extension containing a so-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 this 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), 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, etc., whereas KRAS mutations are rarely found (< 2%) in breast, ovarian and brain cancers. In non-small cell lung cancer (NSCLC), KRAS G12C is the most common mutation, accounting for nearly half of all KRAS mutations, followed by G12V and G12D. In non-small cell lung cancer, the increase in 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 compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof:
Figure PCTCN2022083547-APPB-000001
wherein:
Figure PCTCN2022083547-APPB-000002
selected from single or double bonds as required so that each atom to which it is attached assumes a normal valence state;
ring B is selected from aryl, heteroaryl or fused ring;
Q 1 selected from N or CR a
Q 2 Selected from N, C or CR a
Y is selected from bond, O or NR b
X 1 、X 2 Each independently selected from N, C = O, CR c Or CR (CR) d R e
Provided that when X 2 When selected from N, X 1 Selected from N, C =o or CR d R e
R a The same or different are each independently selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, or a cyano group; 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 selected from hydrogen atomsHalogen, 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 is R c Preferably halogen, more preferably fluorine or chlorine;
R d and R is e The same or different are each independently selected from a hydrogen atom, a halogen, an alkyl group or an alkoxy group; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
Alternatively, R d And R is e Together with the carbon atom to which it is attached, form a cycloalkyl or heterocyclyl group; preferably cyclopropyl;
R 1 selected from-L-cycloalkyl, -L- (6-to 9-membered) monocyclic heterocyclyl, -L-bicyclic heterocyclyl, -L-tricyclic heterocyclyl, -L-aryl, -L-heteroaryl or-L-fused ring; wherein said cycloalkyl, (6-to 9-membered) monocyclic heterocyclyl, bicyclic heterocyclyl, tricyclic heterocyclyl, aryl, heteroaryl OR fused ring is optionally further substituted with one OR more substituents selected from alkyl, halogen, haloalkyl, hydroxyalkyl, benzyl, 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);
l are each independently selected from a bond or C 1 -C 6 An alkylene group, wherein said alkylene group is optionally further substituted with one or more R D Substituted;
R D each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
alternatively, two R's attached to the same carbon atom D And the institute are connected withThe attached carbon atoms together form a cycloalkyl group; preferably cyclopropyl;
R 2 the same or different are each independently selected from a hydrogen atom, halogen, hydroxy, alkyl or alkoxy, preferably a hydrogen atom or alkyl;
two R 3 Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group;
R 4 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, and 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, and aryl, and heteroaryl,Cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl, or carboxylate substituents;
m is selected from 0, 1, 2, 3 or 4;
n is selected from 0, 1, 2 or 3;
r is selected from 0, 1 or 2.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
Figure PCTCN2022083547-APPB-000003
are each selected from double bonds;
Q 1 selected from N or CR a
X 1 Selected from N;
X 2 selected from CR c
R a Selected from cyano;
R c Selected from halogen, preferably fluorine or chlorine, more preferably fluorine.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
Figure PCTCN2022083547-APPB-000004
are each selected from double bonds;
Q 1 selected from N or CR a
X 1 Selected from CR c
X 2 Selected from CR c
R a Selected from cyano;
R c selected from halogen, preferably fluorine or chlorine, more preferably fluorine.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
Figure PCTCN2022083547-APPB-000005
are all selected from single bonds;
X 1 、X 2 each independently selected from CR d R e
R d And R is e Selected from hydrogen atoms;
alternatively, R d And R is e Together with the attached carbon atom, form a 3-5 membered monocyclic cycloalkyl or 3-5 membered monocyclic heterocyclyl; cyclopropyl is preferred.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
X 2 connected to
Figure PCTCN2022083547-APPB-000006
Selected from double bonds;
X 1 and Q 2 Between which are located
Figure PCTCN2022083547-APPB-000007
Selected from single bonds;
X 1 selected from c=o;
X 2 selected from N;
Q 2 selected from N.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
R 1 a self-L-bicyclic heterocyclyl; wherein said bicyclic heterocyclic group is optionally further substituted with one or more groups selected from alkyl, halogen, alkane Oxy or = O; wherein said halogen is preferably fluorine;
l is selected from a bond or C 1 -C 3 An alkylene group, wherein said alkylene group is optionally further substituted with one or more R D Substituted;
R D each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
alternatively, two R's attached to the same carbon atom D Together with the attached carbon atom, form a cycloalkyl group; cyclopropyl is preferred.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
R 1 a self-L-bicyclic heterocyclyl; wherein said bicyclic heterocyclyl is optionally further substituted with one or more substituents selected from alkyl, halogen, alkoxy or =o; wherein said halogen is preferably fluorine;
l is selected from bond, -CH 2 -、-CH 2 CH 2 -or
Figure PCTCN2022083547-APPB-000008
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
R 1 selected from:
Figure PCTCN2022083547-APPB-000009
the invention provides a compound shown in a general formula (I), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, wherein two R 3 Together with the atoms to which they are attached form a 6-to 8-membered cycloalkyl or 6-to 8-membered heterocyclyl.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
R 4 the same or different, each independently selected from hydrogen atom, alkyl, halogen, alkoxy, alkynyl, hydroxy, amino, hydroxyalkyl, haloalkyl or haloalkoxy;
R 4 preferably a hydrogen atom, methyl, fluorine, chlorine, hydroxyl, amino, hydroxymethyl or ethynyl.
The invention provides a compound shown as a general formula (I), or a stereoisomer, a tautomer or pharmaceutically acceptable salt thereof, wherein:
ring B is selected from phenyl, naphthyl, pyridyl, quinolinyl, isoquinolinyl, indolyl, indazolyl, benzothiazolyl, tetrahydronaphthyl,
Figure PCTCN2022083547-APPB-000010
Ring B is preferably naphthyl.
The invention provides a compound shown in a general formula (I), or stereoisomers, tautomers or pharmaceutically acceptable salts thereof, wherein
Figure PCTCN2022083547-APPB-000011
Selected from the following groups:
Figure PCTCN2022083547-APPB-000012
the present invention provides a compound represented by the general formula (I), or a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2022083547-APPB-000013
Selected from the following groups:
Figure PCTCN2022083547-APPB-000014
typical compounds of the present invention include, but are not limited to:
Figure PCTCN2022083547-APPB-000015
Figure PCTCN2022083547-APPB-000016
Figure PCTCN2022083547-APPB-000017
or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
And (3) injection: 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 according to formula (I) 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) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
The invention also provides a use of a compound according to formula (I) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof (e.g. a pharmaceutical composition according to the foregoing technical scheme) 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 selected from cardiac myxoma, lung cancer, stomach cancer, colorectal cancer, 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, preferably pancreatic cancer, colorectal cancer, 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 present invention provides a compound according to formula (I), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof (e.g. a pharmaceutical composition according to the foregoing technical scheme) for use in the preparation of a KRas G12D inhibitor.
Another aspect of the invention relates to a method for preventing and/or treating a disease mediated by KRas G12D mutation, comprising administering to a patient a therapeutically effective dose of a compound according to formula (I) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same (e.g. a pharmaceutical composition according to the foregoing technical scheme). Wherein the disease mediated by the KRas G12D mutation is selected from cancer, wherein the cancer is selected from cardiac myxoma, lung cancer, stomach cancer, colorectal cancer, 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, preferably pancreatic cancer, colorectal cancer, rectal cancer and lung cancer; wherein the lung cancer is selected from non-small cell lung cancer or small cell lung cancer.
The invention also provides the use of a compound of formula (I) or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof (e.g. as described in the preceding claims) for the manufacture of a medicament for the treatment of cancer selected from the group consisting of heart myxoma, lung cancer, stomach cancer, colorectal cancer, 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, preferably pancreatic cancer, colorectal cancer, rectal cancer and lung cancer; wherein the lung cancer is preferably non-small cell lung cancer.
The present invention also provides a method for preventing and/or treating cancer, comprising administering to a patient a therapeutically effective amount of a compound according to formula (I), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof (e.g., a pharmaceutical composition according to the foregoing technical scheme). Wherein the cancer is selected from the group consisting of cardiac myxoma, lung cancer, stomach cancer, colorectal cancer, 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, preferably pancreatic cancer, colorectal cancer, 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-dimethylbutylA group, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, etc. 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 non-aromatic heterocyclic group in which one or more of the ring-forming atoms are heteroatoms, such as oxygen, nitrogen, sulfur atoms, and the like, including monocyclic, fused, bridged and spiro rings. 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 "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, 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 PCTCN2022083547-APPB-000018
"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 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. 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. Examples of "heteroaryl" groups, which are preferably bicyclic 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, benzothienyl, benzimidazolyl, indolyl, isoindolyl, 1, 3-dioxo-isoindolyl, quinolinyl, indazolyl, benzisothiazolyl, benzoxazolyl, benzisoxazolyl,
Figure PCTCN2022083547-APPB-000019
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 of the rings may contain one or more double bonds, but at least one of the rings does not have a fully conjugated pi-electron aromatic system, wherein the ring atoms are selected from 0, one or more of the 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. 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 PCTCN2022083547-APPB-000020
Figure PCTCN2022083547-APPB-000021
"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.
"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, halo, 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 the general 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 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 PCTCN2022083547-APPB-000022
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 PCTCN2022083547-APPB-000023
Ring B, R 1 ~R 4 、X 1 、X 2 、Q 1 、Q 2 The definitions of Y, 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 must be noted that the following examples are given by way of illustration and not by way of limitation. 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.
The thin layer chromatography silica gel plate uses a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification of the silica gel plate used by the 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.
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
5-(4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,8-dihydropyrido[3,4-d]pyrimidin-7(6H)-yl)-4-fluoronaphthalen-2-ol
5- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-fluoronaphthalen-2-ol
Figure PCTCN2022083547-APPB-000024
First step
tert-butyl 4-(8-((benzyloxy)carbonyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)-2-chloro-5,8-dihydropyrido[3,4-d]pyrimidine-7(6H)-carboxylate
4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2-chloro-5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester
Tert-butyl 2, 4-dichloro-5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylate 1a (802.71 mg,2.64 mmol), benzyl 3, 8-diazabicyclo [3.2.1] octane-8-carboxylate 1b (650.00 mg,2.64 mmol) and N, N-diisopropylethylamine (1.02 g,7.92 mmol) were dissolved in sequence in N, N-dimethylacetamide (25 mL) and reacted at 85℃for 2 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, 20mL of water was added, extraction was performed with ethyl acetate (25 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), 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: B system) to give 4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2-chloro-5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester 1c (1 g,1.95 mmol), yield 73.72%.
MS m/z(ESI):513.9[M+1]+
Second step
tert-butyl 4-(8-((benzyloxy)carbonyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,8-dihydropyrido[3,4-d]pyrimidine-7(6H)-carboxylate
4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester
((2R, 7 aS) -2-Fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methanol 1d (281.23 mg,1.77 mmol) was dissolved in tetrahydrofuran (5 mL), sodium hydride (95.72 mg,2.21mmol,60% purity) was added at 0℃and reacted at 0℃for 0.5 hours, 4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2-chloro-5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester 1c (454 mg, 883.24. Mu. Mol) was added and reacted at 70℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, diluted with ethyl acetate (20 mL), washed successively with water (10 mL) and saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the resulting residue was separated and purified by silica gel column chromatography (eluent: B system) to give 4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester 1e (450 mg, 706.71. Mu. Mol) in 80.01% yield.
MS m/z(ESI):637.4[M+1]+
Third step
benzyl 3-(2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
3- (2- (((2R, 7 aS) -2-Fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid benzyl ester
4- (8- ((benzyloxy) carbonyl) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester 1e (450 mg, 706.71. Mu. Mol) was dissolved in dichloromethane (4 mL), trifluoroacetic acid (1 mL) was added, and the reaction was carried out at room temperature for 2 hours. After the completion of the reaction, a saturated sodium hydrogencarbonate solution was added to adjust the pH to 7-8, extracted with methylene chloride (10 mL. Times.3), the organic phases were combined, washed with saturated brine (15 mL), 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 benzyl 3- (2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate 1f (250 mg, 465.86. Mu. Mol), yield 65.92%.
MS m/z(ESI):537.3[M+1]+
Fourth step
benzyl 3-(7-(8-fluoro-6-(methoxymethoxy)naphthalen-1-yl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
3- (7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid benzyl ester
3- (2- (((2R, 7 aS) -2-Fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid benzyl ester 1f (100 mg, 186.34. Mu. Mol), 8-bromo-1-fluoro-3- (methoxymethoxy) naphthalene 1G (79.69 mg, 279.52. Mu. Mol), cesium carbonate (182.14 mg, 559.03. Mu. Mol) and RuPhos Pd G3 (15.60 mg, 18.63. Mu. Mol) were dissolved in sequence in 1, 4-dioxane (5 mL) and reacted at 80℃for 5 hours under the protection of argon. After the completion of the reaction, the reaction mixture was cooled to room temperature, filtered through celite, the cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (eluent: B system) to give benzyl 3- (7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate 1H (73 mg, 98.54. Mu. Mol), in 52.88% yield.
MS m/z(ESI):741.4[M+1]+
Fifth step
4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-fluoro-6-(methoxymethoxy)naphthalen-1-yl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidine
4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2 r,7 as) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidine
3- (7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate benzyl ester 1H (73 mg, 98.54. Mu. Mol) was dissolved in methanol (2 mL), 10% palladium on charcoal (20.97 mg) was added and reacted overnight at room temperature under hydrogen. After the reaction, the reaction mixture was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated under reduced pressure to give the crude product 4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidine 1i (60 mg, 98.89. Mu. Mol) which was directly used for the next reaction.
MS m/z(ESI):607.0[M+1]+
Sixth step
5-(4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-5,8-dihydropyrido[3,4-d]pyrimidin-7(6H)-yl)-4-fluoronaphthalen-2-ol
5- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-fluoronaphthalen-2-ol
4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-fluoro-6- (methoxymethoxy) naphthalen-1-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizine-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidine 1i (60 mg, 98.89. Mu. Mol) was dissolved in 1, 4-dioxane solution (1 mL, 4M) of HCl at 0℃and reacted at room temperature for 2 hours. 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 5- (4- (3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7 aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-fluoronaphthalen-2-ol 1 (5 mg, 8.13. Mu. Mol) in 8.22% yield.
MS m/z(ESI):563.0[M+1]+
1 H NMR(400MHz,CD 3 OD)δ7.44(d,J=8.3Hz,1H),7.30(m,1H),6.94-6.84(m,2H),6.78(d,J=2.2Hz,1H),4.55(d,J=4.7Hz,2H),4.19(m,3H),4.03(m,2H),3.89(dd,J=10.6,7.1Hz,4H),3.46(dq,J=10.9,5.7Hz,2H),2.75-2.56(m,4H),2.34-2.31(m,6H),2.17-2.02(m,6H).
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 (human gastric adenocarcinoma) 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, at 37℃with 5% CO 2 The cells were incubated overnight in an incubator. Test compounds were dissolved in DMSO to prepare 10mM stock solution, then diluted with F12K complete medium, 100. Mu.L 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. Mu.L 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 the excitation wavelength of 304nm were measured on a microplate reader in TF-FRET mode, with emission wavelengths of 620nm and 665nm, and the fluorescence intensity ratio of each well 665/620 was calculated. The percent inhibition of the test compounds at each concentration was calculated by comparison with the fluorescence intensity ratio of the control group (0.1% dmso) and nonlinear regression analysis was performed by GraphPad Prism 5 software with the test compound concentration log-inhibition to obtain compound IC 50 Values.
The 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 preferably IC of compound 50 <200nM。
Test example 2 determination of the inhibition of AsPC-1 cell proliferation by the Compounds of the invention
The following method was used to determine the effect of the compounds of the invention on the proliferation of AsPC-1 (human metastatic pancreatic adenocarcinoma) cells. AsPC-1 cells (containing KRAS G12D mutation) were purchased from Shanghai national academy of sciences of life sciencesCell resource center, in RPMI 1640 medium containing 10% fetal bovine serum, 100U penicillin, 100 μg/mL streptomycin and 1mM Sodium Pyruvate. Cell viability by
Figure PCTCN2022083547-APPB-000025
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 at 37℃with 5% CO 2 The culture was continued overnight in the incubator, followed by the addition of the test compound and continued for 120 hours. After the incubation was completed, a volume of 50. Mu.L of CellTiter-Glo assay solution was added to each well, and after shaking for 5 minutes, the mixture was 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.
The compounds of the invention have a significant inhibitory effect on AsPC-1 cell proliferation, preferably IC of the compounds 50 <500nM, more preferably IC of compound 50 <200nM。
Test example 3 inhibition ability assay of KRAS G12D and RAF1 protein interactions by Compounds of the present invention
The following method was used to determine that the compounds of the invention block KRAS G12D under in vitro conditions: the ability of RAF1 proteins to interact. The method uses KRAS-G12C/SOS1BINDING ASSAY KITS kit (63 ADK000CB21 PEG) from Cisbio, and the detailed experimental operation is referred to the kit instruction.
The experimental procedure is briefly described as follows: working solution concentrations of 5X for Tag1-RAF1 and Tag2-KRAS-G12D proteins were prepared using a current buffer (cat No. 62 DLBDDF). Test compounds were dissolved in DMSO to prepare 10mM stock solutions, which were then diluted using a diluet buffer for use. Firstly, adding 2 mu L of a tested compound (the final concentration of a reaction system is 10000nM-0.1 nM) into a hole, then adding 4 mu L of a Tag1-RAF1 5X working solution and 4 mu L of a Tag2-KRAS-G12D 5X working solution, centrifuging and mixing uniformly, and standing for 15 minutes; then 10 mu L of pre-mixed anti-Tag1-Eu is added 3+ And anti-Tag2-XL665, incubated for 4 hours at room temperature; finally, the fluorescence intensities of the wells at the excitation wavelength of 304nm were measured on a TF-FRET model using a microplate reader, the emission wavelengths of which were 620nm and 665nm, and the fluorescence intensity ratio of each well 665/620 was calculated. The percent inhibition of the test compounds at each concentration was calculated by comparison with the fluorescence intensity ratio of the control group (0.1% dmso) and nonlinear regression analysis was performed by GraphPad Prism 5 software with the test compound concentration log-inhibition to obtain compound IC 50 The values are given in Table 1 below.
TABLE 1 IC for the inhibitory potency of the compounds of the invention on KRAS G12D interactions with RAF1 proteins 50 Data
Examples numbering IC 50 (nM)
1 269
Conclusion: the compound has better inhibition capability on the interaction of KRAS G12D and RAF1 protein.
Test example 4 determination of inhibition of AGS cell proliferation by Compounds of the invention
The following methods were used to determine the effect of the compounds of the invention on AGS cell proliferation. AGS cells (containing KRAS G12D mutation) were purchased from the national academy of sciences of life sciences cell resource center and cultured in F12K medium containing 10% fetal bovine serum, 100U penicillin and 100 μg/mL streptomycin. Cell viability by
Figure PCTCN2022083547-APPB-000026
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 500 cells per well in 96-well cell culture plates at 37℃with 5% CO 2 The culture was continued overnight in the incubator, followed by the addition of the test compound and continued for 72 hours. After the incubation was completed, a volume of 50. Mu.L of CellTiter-Glo assay solution was added to each well, and after shaking for 5 minutes, the mixture was 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 The values are shown in Table 2.
TABLE 2 IC of the compounds of the invention for inhibition of AGS cell proliferation 50 Data
Examples numbering IC 50 (nM)
1 208
Conclusion: the compound has better proliferation inhibition effect on AGS cells.
Unless otherwise defined, all terms used herein are intended to have the meanings commonly understood by those skilled in the art.
The described embodiments of the present invention are intended to be illustrative only and not to limit the scope of the invention, and various other alternatives, modifications, and improvements may be made by those skilled in the art within the scope of the invention, and therefore the invention is not limited to the above embodiments but only by the claims.

Claims (19)

  1. A compound of the general formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
    Figure PCTCN2022083547-APPB-100001
    wherein:
    Figure PCTCN2022083547-APPB-100002
    selected from single or double bonds as required so that each atom to which it is attached assumes a normal valence state;
    ring B is selected from aryl, heteroaryl or fused ring;
    Q 1 selected from N or CR a
    Q 2 Selected from N, C or CR a
    Y is selected from bond, O or NR b
    X 1 、X 2 Each independently selected from N, C = O, CR c Or CR (CR) d R e
    Provided that when X 2 When selected from N, X 1 Selected from N, C =o or CR d R e
    R a The same or different are each independently selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, or a cyano group; 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 selected from the group consisting of 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 is R c Preferably halogen, more preferably fluorine or chlorine;
    R d and R is e The same or different are each independently selected from a hydrogen atom, a halogen, an alkyl group or an alkoxy group; wherein said alkyl or alkoxy is optionally further substituted with one or more substituents selected from halogen, hydroxy, cyano, alkyl or alkoxy;
    alternatively, R d And R is e Together with the carbon atom to which it is attached, form a cycloalkyl or heterocyclyl group; preferably cyclopropyl;
    R 1 selected from-L-cycloalkyl, -L- (6-to 9-membered) monocyclic heterocyclyl, -L-bicyclic heterocyclyl, -L-tricyclic heterocyclyl, -L-aryl, -L-heteroaryl or-L-fused ring; wherein said cycloalkyl, (6-to 9-membered) monocyclic heterocyclyl, bicyclic heterocyclyl, tricyclic heterocyclyl, aryl, heteroaryl or fused ring is optionally further substituted with one or more groups selected from alkyl, halogen, haloalkyl, hydroxyalkyl, benzyl, 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);
    l are each independently selected from a bond or C 1 -C 6 An alkylene group, wherein said alkylene group is optionally further substituted with one or more R D Substituted;
    R D each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
    alternatively, two R's attached to the same carbon atom D Together with the attached carbon atom, form a cycloalkyl group; preferably cyclopropyl;
    R 2 the same or different are each independently selected from a hydrogen atom, halogen, hydroxy, alkyl or alkoxy, preferably a hydrogen atom or alkyl;
    two R 3 Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group;
    R 4 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 oneOne OR more groups selected from alkyl, halogen, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, =o, -OR 5 、-C(O)R 5 、-C(O)OR 5 、-NHC(O)R 5 、-NHC(O)OR 5 、-NR 6 R 7 、-C(O)NR 6 R 7 、-CH 2 NHC(O)OR 5 、-CH 2 NR 6 R 7 or-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, 2, 3 or 4;
    n is selected from 0, 1, 2 or 3;
    r is selected from 0, 1 or 2.
  2. The compound of claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    Figure PCTCN2022083547-APPB-100003
    are each selected from double bonds;
    Q 1 selected from N or CR a
    X 1 Selected from N;
    X 2 selected from CR c
    R a Selected from cyano;
    R c selected from halogen, preferably fluorine or chlorine, more preferably fluorine.
  3. The compound of claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    Figure PCTCN2022083547-APPB-100004
    are each selected from double bonds;
    Q 1 selected from N or CR a
    X 1 Selected from CR c
    X 2 Selected from CR c
    R a Selected from cyano;
    R c selected from halogen, preferably fluorine or chlorine, more preferably fluorine.
  4. The compound of claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    Figure PCTCN2022083547-APPB-100005
    are all selected from single bonds;
    X 1 、X 2 each independently selected from CR d R e
    R d And R is e Selected from hydrogen atoms;
    alternatively, R d And R is e Together with the attached carbon atom, form a 3-5 membered monocyclic cycloalkyl or 3-5 membered monocyclic heterocyclyl; cyclopropyl is preferred.
  5. The compound of claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    X 2 connected to
    Figure PCTCN2022083547-APPB-100006
    Selected from double bonds;
    X 1 and Q 2 Between which are located
    Figure PCTCN2022083547-APPB-100007
    Selected from single bonds;
    X 1 selected from c=o;
    X 2 selected from N;
    Q 2 Selected from N.
  6. A compound according to any one of claims 1 to 5, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    R 1 a self-L-bicyclic heterocyclyl; wherein said bicyclic heterocyclyl is optionally further substituted with one or more substituents selected from alkyl, halogen, alkoxy or =o; wherein said halogen is preferably fluorine;
    l is selected from a bond or C 1 -C 3 An alkylene group, wherein said alkylene group is optionally further substituted with one or more R D Substituted;
    R D each independently selected from a hydrogen atom, halogen, hydroxy or hydroxymethyl;
    alternatively, two R's attached to the same carbon atom D Together with the attached carbon atom, form a cycloalkyl group; cyclopropyl is preferred.
  7. The compound according to claim 6 or a salt thereofStereoisomers, tautomers or pharmaceutically acceptable salts thereof, wherein L is selected from the group consisting of a bond, -CH 2 -、-CH 2 CH 2 -or
    Figure PCTCN2022083547-APPB-100008
  8. The compound according to claim 6, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 1 Selected from:
    Figure PCTCN2022083547-APPB-100009
  9. a compound according to any one of claims 1 to 8, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein two R 3 Together with the atoms to which they are attached form a 6-to 8-membered cycloalkyl or 6-to 8-membered heterocyclyl.
  10. A compound according to any one of claims 1 to 9, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein:
    R 4 the same or different, each independently selected from hydrogen atom, alkyl, halogen, alkoxy, alkynyl, hydroxy, amino, hydroxyalkyl, haloalkyl or haloalkoxy;
    R 4 preferably a hydrogen atom, methyl, fluorine, chlorine, hydroxyl, amino, hydroxymethyl or ethynyl.
  11. A compound according to any one of claims 1 to 10, 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 PCTCN2022083547-APPB-100010
    Ring B is preferably naphthyl.
  12. A compound according to any one of claims 1 to 11, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein
    Figure PCTCN2022083547-APPB-100011
    Selected from the following groups:
    Figure PCTCN2022083547-APPB-100012
  13. a compound according to any one of claims 1 to 12, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein
    Figure PCTCN2022083547-APPB-100013
    Selected from the following groups:
    Figure PCTCN2022083547-APPB-100014
  14. the compound according to claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein the compound is:
    Figure PCTCN2022083547-APPB-100015
    Figure PCTCN2022083547-APPB-100016
  15. A pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 14, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient, or combination thereof.
  16. Use of a compound according to any one of claims 1 to 14, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 15, for the preparation of a KRas G12D inhibitor.
  17. Use of a compound according to any one of claims 1 to 14, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 15, 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 selected from cardiac myxoma, lung cancer, stomach cancer, colorectal cancer, 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, adrenoblastoma, myelogenous leukemia, acute lymphoblastic leukemia or glioblastoma, preferably pancreatic cancer, colorectal cancer, rectal cancer or lung cancer.
  18. Use of a compound according to any one of claims 1 to 14, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 15, for the manufacture of a medicament for the treatment of cancer, wherein the cancer is selected from the group consisting of cardiac myxoma, lung cancer, stomach cancer, colorectal cancer, 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, preferably pancreatic cancer, colorectal cancer, rectal cancer or lung cancer.
  19. The use according to claim 17 or 18, wherein the lung cancer is selected from non-small cell lung cancer or small cell lung cancer.
CN202280006425.6A 2021-03-30 2022-03-29 Heterocyclic derivative and preparation method and application thereof Pending CN116157400A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN2021103388095 2021-03-30
CN202110338809 2021-03-30
CN202110404515 2021-04-15
CN2021104045158 2021-04-15
PCT/CN2022/083547 WO2022206724A1 (en) 2021-03-30 2022-03-29 Heterocyclic derivative, and preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN116157400A true CN116157400A (en) 2023-05-23

Family

ID=83457954

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280006425.6A Pending CN116157400A (en) 2021-03-30 2022-03-29 Heterocyclic derivative and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN116157400A (en)
WO (1) WO2022206724A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020055760A1 (en) * 2018-09-10 2020-03-19 Mirati Therapeutics, Inc. Combination therapies
WO2020239077A1 (en) * 2019-05-29 2020-12-03 上海翰森生物医药科技有限公司 Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
CN116162099A (en) * 2021-11-24 2023-05-26 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof
CN116390923A (en) * 2020-08-21 2023-07-04 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10689377B2 (en) * 2017-11-15 2020-06-23 Mirati Therapeutics, Inc. KRas G12C inhibitors
CN114222743A (en) * 2019-08-16 2022-03-22 劲方医药科技(上海)有限公司 Oxoaxamembered ring pyrimidine compounds, preparation method and medical application thereof
WO2021106231A1 (en) * 2019-11-29 2021-06-03 Taiho Pharmaceutical Co., Ltd. A compound having inhibitory activity against kras g12d mutation
EP4182313A1 (en) * 2020-07-16 2023-05-24 Mirati Therapeutics, Inc. Kras g12d inhibitors
CN115968286A (en) * 2020-08-26 2023-04-14 益方生物科技(上海)股份有限公司 Heteroaryl compounds, their preparation and use
US20230081426A1 (en) * 2020-09-18 2023-03-16 Plexxikon Inc. Compounds and methods for kras modulation and indications therefor
CN114057776A (en) * 2021-10-31 2022-02-18 南京碳硅人工智能生物医药技术研究院有限公司 Novel synthesis method of pyrimidopiperidine derivative with anticancer activity
CN114031562A (en) * 2021-10-31 2022-02-11 南京碳硅人工智能生物医药技术研究院有限公司 Process optimization of pyridine derivative with anti-tumor effect

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020055760A1 (en) * 2018-09-10 2020-03-19 Mirati Therapeutics, Inc. Combination therapies
WO2020239077A1 (en) * 2019-05-29 2020-12-03 上海翰森生物医药科技有限公司 Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
CN116390923A (en) * 2020-08-21 2023-07-04 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof
CN116162099A (en) * 2021-11-24 2023-05-26 浙江海正药业股份有限公司 Heterocyclic derivative and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ELENA CORRAL DE LA FUENTE ET AL.: ""Targeting KRAS in Non-Small Cell Lung Cancer"", 《FRONTIERS IN ONCOLOGY》, vol. 11, 31 January 2022 (2022-01-31), pages 1 - 11 *

Also Published As

Publication number Publication date
WO2022206724A1 (en) 2022-10-06

Similar Documents

Publication Publication Date Title
JP7394074B2 (en) therapeutic compounds
EP3386981B1 (en) Heterocycles useful as anti-cancer agents
CN116113632A (en) Heterocyclic derivative, preparation method and medical application thereof
CN116323625A (en) Heterocyclic derivative, preparation method and medical application 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
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN117769556A (en) Pyrimidine ring derivative and preparation method and application thereof
CN116406363A (en) Tetracyclic derivative, preparation method and medical application thereof
CN116162099A (en) Heterocyclic derivative and preparation method and application thereof
CN116514846A (en) Heterocyclic derivative, preparation method and medical application thereof
CN113929681A (en) Tetracyclic derivative and preparation method and 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
CN116157401A (en) Heterocyclic derivative and preparation method and application thereof
CN116332938A (en) Fused tricyclic derivative and preparation method and application thereof
CN116113416A (en) Tetracyclic derivative, preparation method and medical application thereof
CN115304602A (en) Pyrazinopyrazinonaphthyridinedione derivatives, preparation method and medical application thereof
CN117466917A (en) Heterocyclic derivative, preparation method and medical application thereof
CN115611898A (en) Tetracyclic derivative, preparation method and medical application thereof
CN115557949A (en) Tetracyclic derivative, preparation method and medical application thereof

Legal Events

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