CN116888100A - High-activity HPK1 kinase inhibitor - Google Patents

High-activity HPK1 kinase inhibitor Download PDF

Info

Publication number
CN116888100A
CN116888100A CN202280013620.1A CN202280013620A CN116888100A CN 116888100 A CN116888100 A CN 116888100A CN 202280013620 A CN202280013620 A CN 202280013620A CN 116888100 A CN116888100 A CN 116888100A
Authority
CN
China
Prior art keywords
alkyl
compound
pharmaceutically acceptable
represents hydrogen
stereoisomer
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
CN202280013620.1A
Other languages
Chinese (zh)
Inventor
陈宇锋
时永强
武朋
温俏冬
杨寒
吕萌
刘灿丰
陈凯旋
王友平
何南海
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.)
Hangzhou Arnold Biomedical Technology Co ltd
Original Assignee
Hangzhou Arnold Biomedical Technology 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 Hangzhou Arnold Biomedical Technology Co ltd filed Critical Hangzhou Arnold Biomedical Technology Co ltd
Publication of CN116888100A publication Critical patent/CN116888100A/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
    • A61K31/495Heterocyclic 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms

Abstract

The invention provides a compound with a structure shown in a formula (I) and capable of inhibiting HPK1 kinase activity and a pharmaceutical composition containing the compound. The invention also provides the use of the compounds in the prevention and/or treatment of cancer, tumour, inflammatory diseases, autoimmune diseases or immune mediated diseases.

Description

High-activity HPK1 kinase inhibitor
The present application claims priority to chinese patent application 202110378424.1 entitled "a highly active HPK1 kinase inhibitor" filed on 4/8 of 2021 to the national intellectual property agency of china, the contents of which are incorporated herein by reference in their entirety.
Technical Field
The application relates to a heterocyclic compound, in particular to a high-activity HPK1 kinase inhibitor and application thereof.
Background
HPK1 is one of the members of the MAP4K family, is mainly expressed in hematopoietic cells, and acts as an intracellular negative regulator of T cell proliferation and signaling. Antigen stimulation of T cells causes recruitment of cytoplasmic linker protein SLP-76 to the lipid membrane TCR complex, providing binding sites for signal transduction-related kinases to effect TCR-mediated signaling to induce T cell activation. During this process HPK1 is activated by phosphorylation of tyrosine kinases Lck and Zap70, involved in regulating T cell receptor protein interactions. HPK1 blocks TCR signaling by phosphorylating the Ser376 site of the linker protein SLP-76, allowing SLP-76 to bind to the scaffold protein 14-3-3 epsilon and be degraded by the proteasome, and this effect allows SLP-76 to bind less to signal transduction-related kinases, blocking T cell activation and proliferation. On the other hand, HPK1 is also involved in regulating the maturation and activation of Dendritic Cells (DCs), particularly in inhibiting the expression of proteins involved in the activation of helper T cells in DC cells, such as CD80, CD86 and MHC complexes, thereby affecting the effect of DCs in regulating T cell activation; and the presentation of tumor antigens by activated DCs and the cooperation of DCs and T cells are one of the most important links in the anti-tumor immune system. Furthermore, there are a large number of immunosuppressive molecules such as PGE2 and TGF- β in the tumor microenvironment, and these factor-mediated immunosuppression effects are also important in connection with HPK 1. In general, small molecule compounds that specifically target to inhibit HPK1 can improve T cell function, enhance DC cell function and simultaneously reverse tumor immunosuppressive microenvironment, exert an enhanced anti-tumor immune effect through multiple pathways, thereby achieving the effect of inhibiting tumor growth.
Accordingly, there remains a strong need in the art for potent inhibitors of HPK1 kinase activity in order to provide more effective options for anti-tumor.
Disclosure of Invention
The present invention unexpectedly provides a compound of formula (I) and pharmaceutically acceptable salts or stereoisomers thereof which inhibit HPK1 kinase activity. Thus, in a first aspect, the present invention provides a compound having the structure of formula (I):
wherein the method comprises the steps of
X represents N or CR 10
R 1 Represents hydrogen, C 1 -C 6 Alkyl, halogenated C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, halo C 3 -C 6 Cycloalkyl, halogen, cyano or-C (O) NH 2
R 2 Represents hydrogen, halogen, hydroxy, (C) 1 -C 6 ) Alkyl, (C) 2 -C 6 ) Alkenyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) (4-8 membered) heterocycloalkyl, (C) 1 -C 6 ) Alkoxy, (C) 3 -C 8 ) Cycloalkyl (C) 0 -C 6 Alkylene) oxy, (4-8 membered) heterocycloalkyl (C 0 -C 6 Alkylene) oxy;
R 3 represents hydrogen or C 1 -C 6 Alkyl, C 3 -C 6 Alkenyl, C 3 -C 6 Cycloalkyl, hydroxy (C) 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl, 4-8 membered heterocycloalkyl;
R 4 represents hydrogen or C 1 -C 6 An alkyl group; and said C 1 -C 6 The alkyl group may be optionally substituted with a substituent selected from the group consisting of: halogen, -OR a 、-SR a 、-P(O)R a R b 、-S(O) 2 R a 、-S(O)(NH)R a 、-S(O)R a 、-S(O) 2 NR a R b 、-C(O)NR a R b 、-C(O)OH、-OC(O)NR a R b 、-NR a C(O)R b 、-NR a S(O) 2 R b
R 5 And R is 5’ Each independently represents hydrogen, C 1 -C 6 Alkyl, (C) 2 -C 6 ) Alkenyl, halogen, halo C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 An alkyl group);
or R is 5 And R is R 5’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
R 6 and R is 6’ Each independently represents hydrogen, C 1 -C 6 Alkyl, (C) 2 -C 6 ) Alkenyl, halogen, halo C 1 -C 6 Alkyl, hydroxy (C) 1 -C 6 An alkyl group);
or R is 6 And R is R 6’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
p represents 1 or 2;
R 7 and R is 7’ Each independently represents hydrogen, C 1 -C 6 Alkyl, (C) 2 -C 6 ) Alkenyl, halogen, halo C 1 -C 6 Alkyl, hydroxy (C) 1 -C 6 An alkyl group);
or R is 7 And R is R 7’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
w represents
Wherein A represents
Represents that the ring contains a single bond or a double bond;
when the bond between A and A 'is a double bond, A' represents CR a Or N, B represents CR b
When the bond between A and A 'is a single bond, A' represents CHR a B represents CHR b Or is absent;
R 8 and R is 9 Each independently represents hydrogen or C 1 -C 6 An alkyl group; or R is 8 And R is R 9 Can be combined with W 1 And W is 2 Together forming a 3-6 membered ring;
W 1 represents C, W 2 Represents CH or N;
R M And R is N Each independently represents hydrogen or C 1 -C 6 An alkyl group;
R 10 represents hydrogen, halogen, (C) 1 -C 6 ) Alkyl, (C) 2 -C 6 ) Alkenyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) 4-10 membered heterocycloalkyl, - (C) 0 -C 6 Alkylene group) (C) 6 -C 10 ) Aryl, - (C) 0 -C 6 Alkylene) a 5-to 10-membered heteroaryl,
alternatively, when X represents CR 10 When R is 10 Can be adjacent to R 2 Together forming a (5-10 membered) cycloalkyl or a 5-10 membered heterocycloalkyl;
for cycloalkyl, heterocycloalkyl, aryl, heteroaryl groups as defined above, they may be optionally substituted with 0, 1, 2 or 3 substituents selected from: (C) 1 -C 6 ) Alkyl, (C) 2 -C 6 ) Alkenyl, halo (C) 1 -C 6 ) Alkyl, halo (C) 1 -C 6 ) Alkoxy, - (C) 1 -C 6 Alkylene) -O- (C 1 -C 6 ) Alkyl, (C) 3 -C 8 ) Cycloalkyl, halo (C) 3 -C 8 ) Cycloalkyl, halogen, -CN, oxo, -NR a R b 、-OR a 、-SR a 、-(C 1 -C 6 Alkylene) hydroxy, -C (O) R a 、-N(R a )C(O)R a 、-NR a C(O)OR a 、-NR a SO 2 R a 、-C(O)OR a 、-C(O)N R a R b 、-S(O) 2 N R a R b 、-S(O)R a 、-S(O) 2 R a 、-P(O)R a R b
R a 、R b Each independently represents hydrogen, C 1 -C 6 Alkyl, hydroxy (C) 1 -C 6 Alkyl, halo C 1 -C 6 An alkyl group;
m and n represent 0, 1 or 2, respectively.
In one embodiment of the present invention, W represents
Wherein A represents
Represents that the ring contains a single bond or a double bond;
each o independently represents 0 or 1;
R 8 represents hydrogen or C 1 -C 6 An alkyl group;
R 9 each independently represents hydrogen or C 1 -C 6 An alkyl group;
or R is 8 With adjacent R 9 Together with the carbon atoms to which they are attached, form a 3-6 membered ring;
W 2 、R M 、R N As previously described.
In one embodiment of the present invention, W representsOr alternatively
Represents that the ring contains a single bond or a double bond;
each o independently represents 0 or 1;
R 8 represents hydrogen or C 1 -C 6 An alkyl group;
R 9 each independently represents hydrogen or C 1 -C 6 An alkyl group;
or R is 8 With adjacent R 9 Together with the carbon atoms to which they are attached, form a 3-6 membered ring;
W 2 、R M 、R N the method of claim 1.
In one embodiment of the invention, W is an aromatic heterocycle, preferably an aromatic lactam.
In one embodiment of the invention, W is a saturated heterocycle, preferably a saturated lactam.
In a preferred embodiment of the invention, wherein ring W represents Or alternatively
In a further preferred embodiment of the invention, ring W represents
In a further preferred embodiment of the present invention, the ring W represents W
In a preferred embodiment of the present invention, wherein R 1 Represents hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl, cyano or-C (O) NH 2 The method comprises the steps of carrying out a first treatment on the surface of the More preferably, R 1 Represents hydrogen, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
In a preferred embodiment of the present invention, wherein R 2 Represents hydroxy, (C) 1 -C 6 ) Alkyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) (4-8 membered) heterocycloalkyl, (C) 1 -C 6 ) An alkoxy group; more preferably, R 2 Represent C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyl groups.
In a preferred embodiment of the present invention, wherein R 3 Represents hydrogen or C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, halo C 1 -C 6 An alkyl group; more preferably, R 3 Represents hydrogen or C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 4 Represents hydrogen or C 1 -C 6 An alkyl group; and said C 1 -C 6 The alkyl group may be optionally substituted with a substituent selected from the group consisting of: halogen, -OR a 、-SR a 、-P(O)R a R b 、-S(O) 2 R a 、-S(O)(NH)R a 、-S(O)R a 、-S(O) 2 NR a R b 、-C(O)NR a R b -C (O) OH; more preferably, R 4 Represents hydrogen or C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 5 、R 5’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 6 、R 6’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 7 、R 7’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl, halogenated C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 Alkyl).
In a preferred embodiment of the present invention, wherein R 8 Represents hydrogen or C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 9 Represents hydrogen or C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R 10 Represents hydrogen, halogen, (C) 1 -C 6 ) Alkyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) 4-10 membered heterocycloalkyl; more preferably, R 10 Represents hydrogen, halogen or (C) 1 -C 6 ) An alkyl group.
In a preferred embodiment of the present invention, wherein R M Represents hydrogen or C 1 -C 6 An alkyl group.
In a preferred embodiment of the present invention, wherein R a 、R b Each independently represents hydrogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
Preferably, the compounds of the present invention have the following structure:
in addition, the invention also provides a pharmaceutical composition which comprises the compound and a pharmaceutically acceptable carrier.
In addition, the invention also provides application of the compound or the pharmaceutical composition in preparing medicines for preventing and/or treating cancers, tumors, inflammatory diseases, autoimmune diseases or immune mediated diseases.
In addition, the invention also provides the use of the compound of the invention for preventing and/or treating cancer, tumor, inflammatory diseases, autoimmune diseases or immune-mediated diseases.
It is particularly noted that, in this context, references to "compounds" of a particular structural formula are also generally intended to encompass stereoisomers, diastereomers, enantiomers, racemic mixtures, and isotopic derivatives thereof.
It is well known to those skilled in the art that salts, solvates, hydrates of a compound are alternative forms of a compound, all of which can be converted to the compound under certain conditions, and therefore, it is of particular note herein that when referring to a compound, generally also pharmaceutically acceptable salts thereof, and further solvates and hydrates thereof, are included.
Similarly, when a compound is referred to herein, prodrugs, metabolites, and nitrogen oxides thereof are also generally included.
Pharmaceutically acceptable salts according to the invention may be formed using, for example, the following mineral or organic acids: by "pharmaceutically acceptable salt" is meant a salt which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like commensurate with a reasonable benefit/risk ratio. The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by reacting the free base or free acid with a suitable reagent alone, as outlined below. For example, the free base functionality may be reacted with a suitable acid. Furthermore, when the compounds of the present invention bear an acidic moiety, suitable pharmaceutically acceptable salts thereof may include metal salts, such as alkali metal salts (e.g., sodium or potassium salts); and alkaline earth metal salts (such as calcium or magnesium salts). Examples of pharmaceutically acceptable non-toxic acid addition salts are salts of amino groups with inorganic acids (e.g., hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric) or organic acids (e.g., acetic, oxalic, maleic, tartaric, citric, succinic or malonic) or by using other methods in the art such as ion exchange. Other pharmaceutically acceptable salts include adipic acid salts, sodium alginate, ascorbate, aspartic acid salts, benzenesulfonate salts, benzoate salts, bisulfate salts, borate salts, butyric acid salts, camphoric acid salts, citric acid salts, cyclopentanepropionate salts, digluconate salts, dodecylsulfate salts, ethanesulfonate salts, formate salts, fumaric acid salts, glucoheptonate salts, glycerophosphate salts, gluconate salts, southern sulfate salts, heptanoate salts, caproate salts, hydroiodic acid salts, 2-hydroxy-ethanesulfonate salts, lactobionate salts, lactate salts, laurate salts, lauryl sulfate salts, malate salts, maleate salts, malonate salts, methanesulfonate salts, 2-naphthalenesulfonate salts, nicotinate salts, nitrate salts, oleate salts, oxalate salts, palmitate salts, pamoate salts, pectate salts, persulfates, 3-phenylpropionate salts, phosphate salts, bitter salts, pivalate salts, propionate salts, stearate salts, succinate salts, sulfate salts, tartrate salts, thiocyanate salts, p-toluenesulfonate salts, undecanoate salts, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Other pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts, and ammonium cations formed with counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkyl sulfonates, and aryl sulfonates, as appropriate.
The pharmaceutically acceptable salts of the invention may be prepared by conventional methods, for example by dissolving the compounds of the invention in a water miscible organic solvent (e.g. acetone, methanol, ethanol and acetonitrile), adding thereto an excess of an organic or inorganic acid aqueous solution to precipitate the salt from the resulting mixture, removing the solvent and the remaining free acid therefrom, and then isolating the precipitated salt.
The precursor or metabolite of the present invention may be a precursor or metabolite well known in the art, as long as the precursor or metabolite is convertible by in vivo metabolism to form the target compound. For example, "prodrugs" refer to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like commensurate with a reasonable benefit/risk ratio, and are effective for their intended use. The term "prodrug" refers to a compound that is rapidly transformed in vivo to produce the parent compound of the formula described above, for example by metabolism in vivo, or N-demethylation of a compound of the invention.
"solvate" as used herein means a physical association of a compound of the invention with one or more solvent molecules (whether organic or inorganic). The physical association includes hydrogen bonding. In some cases, for example when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, the solvate will be able to be isolated. The solvent molecules in the solvate may be present in a regular arrangement and/or in a disordered arrangement. The solvate may comprise a stoichiometric or non-stoichiometric solvent molecule. "solvate" encompasses both solution phases and separable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolamides. Solvation methods are well known in the art.
The term "stereoisomers" as used herein is divided into conformational isomerism and configurational isomerism, which may be also divided into cis-trans isomerism and optical isomerism (i.e. optical isomerism), and conformational isomerism refers to a stereoisomerism phenomenon that an organic molecule with a certain configuration makes each atom or group of molecules generate different arrangement modes in space due to rotation or twisting of carbon and carbon single bonds, and commonly includes structures of alkane and cycloalkane compounds, such as chair-type conformations and boat-type conformations, which occur in cyclohexane structures. "stereoisomers" means that when a compound of the invention contains one or more asymmetric centers, it is useful as racemate and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The compounds of the invention have asymmetric centers, each of which produces two optical isomers, and the scope of the invention includes all possible optical isomers and diastereomeric mixtures and pure or partially pure compounds. The compounds described herein may exist in tautomeric forms having different points of attachment of hydrogen through displacement of one or more double bonds. For example, the ketone and its enol form are keto-enol tautomers. Each tautomer and mixtures thereof are included in the compounds of the present invention. All enantiomers, diastereomers, racemates, meso, cis-trans isomers, tautomers, geometric isomers, epimers, mixtures thereof and the like of the compounds of formula (I) are included within the scope of the present invention.
The term "isotopically-labeled" as used herein refers to molecules wherein the compound is isotopically labeled. Isotopes commonly used as isotopic labels are: the hydrogen isotope is selected from the group consisting of, 2 h and 3 h is formed; carbon isotopes: 11 C、 13 c and C 14 C, performing operation; chlorine isotopes: 35 cl and Cl 37 Cl; fluorine isotopes: 18 f, performing the process; iodine isotopes: 123 i and 125 i, a step of I; nitrogen isotopes: 13 n and 15 n; oxygen isotopes: 15 O、 17 o and 18 isotopes of O and sulfur 35 S, S. These isotopically-labeled compounds can be used to study the distribution of a pharmaceutical molecule in a tissue. In particular deuterium 3 H and carbon 13 C, because they are easily labeled and conveniently detected, the application is wider. Certain heavy isotopes, such as heavy hydrogen @, for example 2 H) The substitution can enhance the metabolic stability and prolong the half-life period, thereby achieving the aim of reducing the dosage and providing curative effect advantages. Isotopically-labeled compounds are generally synthesized starting from a starting material that has been labeled, using known synthetic techniques as if it were a non-isotopically-labeled compound.
The invention also provides the use of the compounds of the invention in the manufacture of a medicament for the prophylaxis and/or treatment of cancer, tumour, inflammatory disease, autoimmune disease or immune mediated disease.
Furthermore, the present invention provides a pharmaceutical composition for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, comprising the compound of the present invention as an active ingredient.
Furthermore, the present invention provides a method for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease comprising administering to a mammal in need thereof a compound of the present invention.
Representative examples of inflammatory, autoimmune and immune-mediated diseases may include but are not limited to, arthritis, rheumatoid arthritis, spinal arthritis, gouty arthritis, osteoarthritis, juvenile arthritis, other arthritic conditions, lupus, systemic Lupus Erythematosus (SLE), skin-related diseases, psoriasis, eczema, dermatitis, allergic dermatitis, pain, lung disease, pulmonary inflammation, adult Respiratory Distress Syndrome (ARDS), pulmonary sarcoidosis, chronic pulmonary inflammatory diseases, chronic Obstructive Pulmonary Disease (COPD), cardiovascular diseases, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia reperfusion injury, inflammatory bowel disease, crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, sjogren's syndrome, autoimmune thyroid disease urticaria (rubella), multiple sclerosis, scleroderma, organ transplant rejection, xenograft, idiopathic Thrombocytopenic Purpura (ITP), parkinson's disease, alzheimer's disease, diabetes-related diseases, inflammation, pelvic inflammatory disease, allergic rhinitis, allergic bronchitis, allergic sinusitis, leukemia, lymphoma, B-cell lymphoma, T-cell lymphoma, myeloma, acute Lymphoblastic Leukemia (ALL), chronic Lymphoblastic Leukemia (CLL), acute Myelogenous Leukemia (AML), chronic Myelogenous Leukemia (CML), hairy cell leukemia, hodgkin's disease, non-hodgkin's lymphoma, multiple myeloma, myelodysplastic syndrome (MDS), myeloproliferative neoplasm (MPN), diffuse large B-cell lymphoma and follicular lymphoma.
Representative examples of cancers or tumors may include but are not limited to, skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neuroblastoma, rectal cancer, colon cancer, familial adenomatous polyposis, hereditary non-polyposis colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, hypopharynx cancer, tongue cancer, salivary gland cancer, stomach cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, endometrial cancer, choriocarcinoma, pancreatic cancer, prostate cancer, testicular cancer, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma, and peripheral nerve ectodermal tumors hodgkin's lymphoma, non-hodgkin's lymphoma, burkitt's lymphoma, acute Lymphoblastic Leukemia (ALL), chronic Lymphocytic Leukemia (CLL), acute Myelogenous Leukemia (AML), chronic Myelogenous Leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchogenic carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basal cell carcinoma, teratoma, retinoblastoma, choriocarcinoma, seminoma, rhabdomyosarcoma, craniopharyngeal pipe carcinoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, ewing's sarcoma, or plasmacytoma.
Representative examples of therapeutic agents for the treatment of inflammatory, autoimmune, and immune-mediated diseases may include, but are not limited to, steroidal drugs (e.g., prednisone, hydroprednisone, methyl hydroprednisone, cortisone, hydroxy cortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-tnfα agents (e.g., etanercept, infliximab, ada Li Shan resistance, etc.), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, etc.), and antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebastine, ketotifen, cetirizine, levocetirizine, fexofenadine, etc.), and at least one therapeutic agent selected therefrom may be included in the pharmaceutical compositions of the present invention.
The compound of the present invention or a pharmaceutically acceptable salt thereof may be administered orally or parenterally as an active ingredient in an effective amount ranging from 0.1 to 2,000mg/kg body weight/day, preferably 1 to 1,000mg/kg body weight/day in the case of mammals including humans (body weight of about 70 kg), and administered in divided doses, single or 4 times daily, or with/without following a predetermined time. The dosage of the active ingredient may be adjusted according to a number of relevant factors, such as the condition of the subject to be treated, the type and severity of the disease, the rate of administration and the opinion of the physician. In some cases, amounts less than the above dosages may be suitable. An amount greater than the above dosage may be used if it does not cause deleterious side effects and may be administered in divided doses per day.
In addition, the present invention provides a method for preventing and/or treating a tumor, cancer, viral infection, organ transplant rejection, neurodegenerative disease, attention-related disease or autoimmune disease, comprising administering to a mammal in need thereof a compound of the present invention or a pharmaceutical composition of the present invention.
The pharmaceutical compositions of the present invention may be formulated according to any of the conventional methods into dosage forms for oral administration or parenteral administration (including intramuscular, intravenous and subcutaneous routes, intratumoral injection), such as tablets, granules, powders, capsules, syrups, emulsions, microemulsions, solutions or suspensions.
Other features of the present invention will become apparent in the course of describing exemplary embodiments of the invention, which are presented to illustrate the invention and are not intended to be limiting thereof, the following examples being prepared, isolated and characterized using the methods disclosed herein.
The compounds of the present invention may be prepared in a variety of ways known to those skilled in the art of organic synthesis, and may be synthesized using the methods described below as well as synthetic methods known in the art of organic synthetic chemistry or by variations thereof as will be appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below. The reaction is carried out in a solvent or solvent mixture suitable for the kit materials used and for the transformation to be effected. Those skilled in the art of organic synthesis will understand that the functionalities present on the molecule are consistent with the proposed transformations. This sometimes requires judgment to change the order or starting materials of the synthesis steps to obtain the desired compounds of the invention.
Detailed Description
Terminology
The terms used in the present application, including the specification and claims, are defined as follows, unless otherwise indicated. It must be noted that, in the specification and the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Conventional methods of mass spectrometry, nuclear magnetism, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are used, if not otherwise indicated. In the present application, the use of "or" and "means" and/or "unless otherwise indicated.
In the description and claims, a given formula or name shall encompass all stereoisomers and optical isomers and racemates in which the isomers exist. Unless otherwise indicated, all chiral (enantiomers and diastereomers) and racemic forms are within the scope of the present application. Many geometric isomers of c=c double bonds, c=n double bonds, ring systems, etc. may also be present in the compounds, and all such stable isomers are contemplated within the present application. The present application describes cis-and trans- (or E-and Z-) geometric isomers of the compounds of the present application, and which may be separated into mixtures of isomers or separate isomeric forms. The compounds of the application may be isolated in optically active or racemic forms. All processes for preparing the compounds of the application and intermediates prepared therein are considered part of the present application. When preparing the enantiomeric or diastereomeric products, they can be separated by conventional methods, for example by chromatography or fractional crystallization. Depending on the process conditions, the end products of the application are obtained in free (neutral) or salt form. Both the free form and the salt of these end products are within the scope of the application. If desired, one form of the compound may be converted to another form. The free base or acid may be converted to a salt; the salt may be converted to the free compound or another salt; mixtures of the isomeric compounds of the application may be separated into the individual isomers. The compounds of the application, free forms and salts thereof, may exist in various tautomeric forms in which hydrogen atoms are transposed to other parts of the molecule and thereby the chemical bonds between the atoms of the molecule are rearranged. It is to be understood that all tautomeric forms that may exist are included within the application.
Unless otherwise defined, the definition of substituents of the invention are each independent of, and not interrelated with, each other, e.g. for R in a substituent a (or R) a ') which are independent of each other in the definition of the different substituents. Specifically, for R a (or R) a ' when a definition is selected in a substituent, it does not mean that R a (or R) a ') have the same definition in all other substituents. More specifically, for example (by way of non-exhaustive list) for NR a R a In' when R a (or R) a Where the definition of') is selected from hydrogen, it is not meant to be in-C (O) -NR a R a In' R a (or R) a ') is necessarily hydrogen.
Unless otherwise defined, when a substituent is noted as "optionally substituted", the substituent is selected from, for example, substituents such as alkyl, cycloalkyl, aryl, heterocyclyl, halogen, hydroxy, alkoxy, oxo, alkanoyl, aryloxy, alkanoyloxy, amino, alkylamino, arylamino, arylalkylamino, disubstituted amino groups (wherein 2 amino substituents are selected from alkyl, aryl or arylalkyl), alkanoylamino, aroylamino, aralkylamino, substituted alkanoylamino, substituted arylamino, substituted aralkylamino, thio, alkylthio, arylthio, arylalkylthio, arylthiocarbonyl, arylalkylthiocarbonyl, alkylsulfonyl, arylsulfonyl, arylalkylsulfonyl, sulfonylamino, e.g., -SO 2 NH 2 Substituted sulfonylamino, nitro, cyano, carboxyl, carbamoylFor example-CONH 2 Substituted carbamoyl such as-CONH alkyl, -CONH aryl, -CONH arylalkyl or where there are two substituents on the nitrogen selected from alkyl, aryl or arylalkyl, alkoxycarbonyl, aryl, substituted aryl, guanidino, heterocyclyl such as indolyl, imidazolyl, furanyl, thienyl, thiazolyl, pyrrolidinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, homopiperazinyl and the like and substituted heterocyclyl.
The term "alkyl" or "alkylene" as used herein is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having the indicated number of carbon atoms. For example, "C 1 -C 6 Alkyl "means an alkyl group having 1 to 6 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, tert-butyl), and pentyl (e.g., n-pentyl, isopentyl, neopentyl).
The term "alkenyl" denotes a straight or branched hydrocarbon radical containing one or more double bonds and typically having a length of 2 to 20 carbon atoms. For example, "C2-C6 alkenyl" contains two to six carbon atoms. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, and the like.
The term "alkynyl" denotes a straight or branched hydrocarbon radical containing one or more triple bonds and typically ranging in length from 2 to 20 carbon atoms. For example, "C 2 -C 6 Alkynyl "contains two to six carbon atoms. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, and the like.
The term "alkoxy" or "alkyloxy" refers to an-O-alkyl group. "C 1 -C 6 Alkoxy "(or alkyloxy) is intended to include C 1 、C 2 、C 3 、C 4 、C 5 、C 6 An alkoxy group. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), and t-butoxy. Similarly, "alkylthio" or "thioalkanesOxy "represents a sulfur-bridged alkyl group as defined above having the indicated number of carbon atoms; such as methyl-S-and ethyl-S-.
The term "carbonyl" refers to an organofunctional group (c=o) formed by the double bond connection of two atoms of carbon and oxygen.
The term "aryl", alone or as part of a larger moiety such as "aralkyl", "aralkoxy" or "aryloxyalkyl", refers to a monocyclic, bicyclic or tricyclic ring system having a total of 5 to 12 ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. In certain embodiments of the present invention, "aryl" refers to an aromatic ring system including, but not limited to, phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl, and tetrahydronaphthyl. The term "aralkyl" or "arylalkyl" refers to an alkyl residue attached to an aryl ring. Non-limiting examples include benzyl, phenethyl, and the like. The fused aryl group may be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring. Examples dashed lines drawn from the ring system indicate that the bond may be attached to any suitable ring atom.
The term "cycloalkyl" refers to a monocyclic or bicyclic cyclic alkyl group, preferably having 3 to 8 ring members. Monocyclic cyclic alkyl means C 3 -C 8 Including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbornyl. Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included in the definition of "cycloalkyl". Bicyclic cyclic alkyl groups include bridged, spiro, or fused cyclic cycloalkyl groups.
The term "cycloalkenyl" refers to a monocyclic or bicyclic cyclic alkenyl, preferably having 3 to 8 ring members. Monocyclic cyclic alkenyl means C 3 -C 8 Cyclic alkenyl groups of (c) including, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, and norbornenyl. Branched cycloalkenyl groups such as 1-methylcyclopropenyl and 2-methylcyclopropenyl are included in the definition of "cycloalkenyl". Bicyclic cycloalkenyl groups include bridged, spiro, or fused cyclic alkenyl groups.
"halo" or "halogen" includes fluoro, chloro, bromo and iodo. "haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the indicated number of carbon atoms and substituted with 1 or more halogens, preferably 1, 2 or 3 halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl. Examples of haloalkyl groups also include "fluoroalkyl groups" intended to include branched and straight-chain saturated aliphatic hydrocarbon groups having the indicated number of carbon atoms (preferably 1 to 6 carbon atoms) and substituted with 1 or more fluorine atoms.
"haloalkoxy" or "haloalkyloxy" means an oxygen-bridged haloalkyl as defined above having the indicated number of carbon atoms, preferably 1 to 6 carbon atoms. For example, "halo C 1 -C 6 Alkoxy "is intended to include C 1 、C 2 、C 3 、C 4 、C 5 、C 6 Haloalkoxy groups. Examples of haloalkoxy groups include, but are not limited to, trifluoromethoxy, 2-trifluoroethoxy, and pentafluoroethoxy. Similarly, "haloalkylthio" or "thiohaloalkoxy" means a thio-bridged haloalkyl as defined above having the indicated number of carbon atoms (preferably 1 to 6 carbon atoms); such as trifluoromethyl-S-and pentafluoroethyl-S-.
In the present disclosure, C is used when referring to some substituents x1 -C x2 This means that the number of carbon atoms in the substituent group may be x1 to x 2. For example, C 0 -C 8 Represents that the radical contains 0, 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms, C 1 -C 8 Representing that the radicals contain 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms, C 2 -C 8 Representing that the radicals contain 2, 3, 4, 5, 6, 7 or 8 carbon atoms, C 3 -C 8 Representing that the radicals contain 3, 4, 5, 6, 7 or 8 carbon atoms, C 4 -C 8 Indicating that the radicals containHaving 4, 5, 6, 7 or 8 carbon atoms, C 0 -C 6 Represents that the radical contains 0, 1, 2, 3, 4, 5 or 6 carbon atoms, C 1 -C 6 Representing that the radicals contain 1, 2, 3, 4, 5 or 6 carbon atoms, C 2 -C 6 Representing that the radicals contain 2, 3, 4, 5 or 6 carbon atoms, C 3 -C 6 Meaning that the group contains 3, 4, 5 or 6 carbon atoms.
In the present disclosure, the expression "x1-x2 membered ring" is used when referring to a cyclic group (e.g., aryl, heteroaryl, cycloalkyl, and heterocycloalkyl), which means that the number of ring atoms of the group can be x1 to x 2. For example, the 3-12 membered cyclic group may be a 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 membered ring, the number of ring atoms of which may be 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; the 3-6 membered ring represents that the cyclic group may be a 3, 4, 5 or 6 membered ring, and the number of ring atoms may be 3, 4, 5 or 6; the 3-8 membered ring represents that the cyclic group may be a 3, 4, 5, 6, 7 or 8 membered ring, and the number of ring atoms may be 3, 4, 5, 6, 7 or 8; the 3-9 membered ring represents that the cyclic group may be a 3, 4, 5, 6, 7, 8 or 9 membered ring, and the number of ring atoms may be 3, 4, 5, 6, 7, 8 or 9; the 4-7 membered ring represents that the cyclic group may be a 4, 5, 6 or 7 membered ring, and the number of ring atoms may be 4, 5, 6 or 7; the 5-8 membered ring represents that the cyclic group may be a 5, 6, 7 or 8 membered ring, and the number of ring atoms may be 5, 6, 7 or 8; the 5-12 membered ring represents that the cyclic group may be a 5, 6, 7, 8, 9, 10, 11 or 12 membered ring, and the number of ring atoms may be 5, 6, 7, 8, 9, 10, 11 or 12; the 6-12 membered ring means that the cyclic group may be a 6, 7, 8, 9, 10, 11 or 12 membered ring, and the number of ring atoms may be 6, 7, 8, 9, 10, 11 or 12. The ring atom may be a carbon atom or a heteroatom, for example a heteroatom selected from N, O and S. When the ring is a heterocyclic ring, the heterocyclic ring may contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more ring heteroatoms, for example heteroatoms selected from N, O and S.
In the present disclosure, the one or more halogens may each be independently selected from fluorine, chlorine, bromine, and iodine.
The term "heteroaryl" means a stable 3-, 4-, 5-, 6-, or 7-membered aromatic monocyclic or aromatic bicyclic or 7-, 8-, 9-, 10-, 11-, 12-membered aromatic polycyclic heterocycle which is fully unsaturated, partially unsaturated and which contains carbon atoms and 1,2,3 or 4 heteroatoms independently selected from N, O and S; and includes any of the following polycyclic groups wherein any of the heterocycles defined above is fused to a benzene ring. The nitrogen and sulfur heteroatoms may optionally be oxidized. The nitrogen atom is substituted or unsubstituted (i.e., N or NR, where R is H or another substituent if defined). The heterocycle may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. If the resulting compound is stable, the heterocyclyl groups described herein may be substituted on a carbon or nitrogen atom. The nitrogen in the heterocycle may optionally be quaternized. Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to each other. Preferably, the total number of S and O atoms in the heterocycle is no greater than 1. When the term "heterocycle" is used, it is intended to include heteroaryl. Examples of aryl radicals include, but are not limited to, acridinyl, azetidinyl, azepinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothienyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzotriazole, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4 aH-carbazolyl, carbolinyl, chromanyl, chromen, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro [2,3-b ] tetrahydrofuranyl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, imidazopyridinyl, indolyl (indolenyl), indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinyl (atinoyl), isobenzofuranyl, isochromanyl isoindazolyl, isoindolinyl, isoindolyl, isoquinolyl, isothiazolyl, isothiazolopyridinyl, isoxazolyl, isoxazolopyridinyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolyl, oxadiazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolopyridinyl, oxazolidinyl, naphthyridinyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidinonyl, 2H-pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-1,2, 5-thiadiazinyl, 1,2, 3-thiadiazinyl, 1,2, 4-thiadiazinyl, 1,2, 5-thiadiazinyl, 1,3, 4-thiadiazinyl, thianthrenyl, thiazolyl, thienyl, thiazolopyridinyl thienothiazolyl, thienooxazolyl, thienoimidazolyl, thienyl, triazinyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 5-triazolyl, 1,3, 4-triazolyl and xanthenyl, quinolinyl, isoquinolinyl, phthalazinyl, quinazolinyl, indolyl, isoindolyl, indolinyl, 1H-indazolyl, benzimidazolyl, 1,2,3, 4-tetrahydroquinolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 5,6,7, 8-tetrahydro-quinolinyl, 2, 3-dihydro-benzofuranyl, chromanyl, 1,2,3, 4-tetrahydro-quinoxalinyl and 1,2,3, 4-tetrahydro-quinazolinyl. The term "heteroaryl" may also include biaryl structures formed from "aryl" and monocyclic "heteroaryl" as defined above, such as, but not limited to "-phenyl bipyridyl-", "-phenyl bipyrimidinyl", "-pyridinyl biphenyl", "-pyridinyl bipyrimidinyl-", "-pyrimidinyl biphenyl-"; wherein the invention also includes fused and spiro compounds containing, for example, the above-described heterocycles.
The term "heterocycloalkyl" as used herein refers to a monocyclic heterocycloalkyl system, or a bicyclic heterocycloalkyl system, and also includes spiroheterocycles or bridged heterocycloalkyl groups. A monocyclic heterocycloalkyl group refers to a 3-8 membered, and contains at least one saturated or unsaturated, but not aromatic, cyclic alkyl system selected from O, N, S, P. Bicyclic heterocycloalkyl system refers to a heterocycloalkyl fused to a phenyl, or a cycloalkyl, or a cycloalkenyl, or a heterocycloalkyl, or a heteroaryl.
The term "bridged cycloalkyl" as used herein refers to polycyclic compounds sharing two or more carbon atoms. Can be classified into bicyclic bridged ring hydrocarbons and polycyclic bridged ring hydrocarbons. The former is composed of two alicyclic rings sharing more than two carbon atoms; the latter is a bridged cyclic hydrocarbon consisting of three or more rings.
The term "spirocycloalkyl" as used herein refers to a polycyclic hydrocarbon having a single ring of carbon atoms in common with each other (referred to as spiro atoms).
The term "bridged cyclohexyl" as used herein refers to a polycyclic compound having a common use of two or more carbon atoms, at least one of the rings containing a member selected from the group consisting of O, N, S atoms. Can be divided into two-ring bridged heterocyclic rings and multiple-ring bridged heterocyclic rings.
The term "heterospirocyclic" as used herein refers to a polycyclic hydrocarbon having a single ring with at least one atom selected from O, N, S which shares a single carbon atom (referred to as the spiro atom).
The term "substituted" as used herein means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that the normal valence is maintained and that the substitution results in a stable compound. As used herein, a ring double bond is a double bond formed between two adjacent ring atoms (e.g., c= C, C =n or n=n).
In the case where nitrogen atoms (e.g., amines) are present on the compounds of the present invention, these nitrogen atoms may be converted to N-oxides by treatment with an oxidizing agent (e.g., mCPBA and/or hydrogen peroxide) to obtain other compounds of the present invention. Thus, the nitrogen atoms shown and claimed are considered to both encompass the nitrogen shown and its N-oxides to obtain the derivatives of the invention.
When any variable occurs more than one time in any composition or formula of a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-3R, then the group may optionally be substituted with up to three R groups, and R is independently selected at each occurrence from the definition of R. Furthermore, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "patient" as used herein refers to an organism treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murine, simian/monkey, equine, bovine, porcine, canine, feline, etc.) and most preferably refer to humans.
The term "effective amount" as used herein means the amount of a drug or pharmaceutical agent (i.e., a compound of the present invention) that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means an amount of: such amounts result in improved treatment, cure, prevention, or alleviation of a disease, disorder, or side effect, or a reduction in the rate of progression of a disease or disorder, as compared to a corresponding subject not receiving such amounts. An effective amount may be administered in one or more administrations, or dosages and is not intended to be limited to a particular formulation or route of administration. The term also includes within its scope an effective amount to enhance normal physiological function.
The term "treatment" as used herein includes any effect that results in an improvement in a condition, disease, disorder, etc., such as a reduction, decrease, modulation, improvement or elimination, or improvement of symptoms thereof.
The term "pharmaceutically acceptable" is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are: it is suitable for use in contact with human and animal tissue without undue toxicity, irritation, allergic response, and/or other problems or complications commensurate with a reasonable benefit/risk ratio, within the scope of sound medical judgment.
The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutical substance, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc, magnesium stearate, calcium or zinc stearate, or stearic acid), or solvent encapsulating material, which involves carrying or transporting the subject compound from one organ or body part to another organ or body part. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the patient.
The term "pharmaceutical composition" means a composition comprising a compound of the invention and at least one other pharmaceutically acceptable carrier. "pharmaceutically acceptable carrier" refers to a medium commonly accepted in the art for delivery of biologically active agents to animals, particularly mammals, and includes (i.e., adjuvants, excipients or vehicles such as diluents, preservatives, fillers, flow control agents, disintegrants, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, antibacterial agents, antifungal agents, lubricants, and dispersing agents, depending upon the mode of administration and the nature of the dosage form.
Specific pharmaceutical and medical terminology
The term "acceptable" as used herein, means that a prescription component or active ingredient does not unduly adversely affect the health of the general therapeutic objective.
The term "cancer", as used herein, refers to an abnormal growth of cells that is not controllable and is capable of metastasis (transmission) under certain conditions. Cancers of this type include, but are not limited to, solid tumors (e.g., bladder, intestine, brain, chest, uterus, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas, or other endocrinologic organ (e.g., thyroid), prostate, skin (melanoma), or hematological tumors (e.g., non-leukemia).
The term "co-administration" or similar terms, as used herein, refers to administration of several selected therapeutic agents to a patient, administered at the same or different times, in the same or different modes of administration.
The term "enhance" or "potentiating," as used herein, means that the intended result can be increased or prolonged in either potency or duration. Thus, in enhancing the therapeutic effect of a drug, the term "enhancing" refers to the ability of a drug to increase or prolong the potency or duration of the drug in the system. As used herein, "potentiating value" refers to the ability of an additional therapeutic agent to be maximally enhanced in an ideal system.
The term "immunological disorder" refers to a disease or condition that produces an adverse or detrimental response to an endogenous or exogenous antigen. As a result, the cells are often dysfunctional, or thus destroyed and dysfunctional, or destroy organs or tissues that may develop immune symptoms.
The term "kit" is synonymous with "product package".
The term "subject" or "patient" includes mammals and non-mammals. Mammals include, but are not limited to, mammals: humans, non-human primates such as gorillas, apes, and monkeys; agricultural animals such as cattle, horses, goats, sheep, pigs; domestic animals such as rabbits and dogs; laboratory animals include rodents such as rats, mice, guinea pigs, and the like. Non-mammalian animals include, but are not limited to, birds, fish, and the like. In a preferred embodiment, the mammal selected is a human.
The terms "treat," "course of treatment," or "therapy" as used herein include alleviation, inhibition, or amelioration of symptoms or conditions of a disease; inhibit the occurrence of complications; improving or preventing underlying metabolic syndrome; inhibiting the occurrence of a disease or condition, such as controlling the progression of a disease or condition; alleviating a disease or symptom; causing the disease or symptom to subside; alleviating complications caused by diseases or symptoms, or preventing and/or treating signs caused by diseases or symptoms.
As used herein, a compound or pharmaceutical composition, upon administration, may result in an improvement in a disease, symptom, or condition, particularly an improvement in severity, delay of onset, slow progression, or decrease in duration. Whether stationary or temporary, continuous or intermittent, may be due to or associated with administration.
Route of administration
Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ocular, pulmonary, transdermal, vaginal, auditory canal, nasal, and topical. Further, by way of example only, parenteral administration includes intramuscular, subcutaneous, intravenous, intramedullary, ventricular, intraperitoneal, intralymphatic, and intranasal.
In one aspect, the administration of the compounds described herein is topical rather than systemic. In certain embodiments, the depot is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Furthermore, in another specific embodiment, the drug is administered by a targeted drug delivery system. For example, liposomes encapsulated by organ-specific antibodies. In this particular embodiment, the liposomes are selectively targeted to a specific organ and absorbed.
Pharmaceutical composition and dosage
The invention also provides pharmaceutical compositions comprising a therapeutically effective amount of one or more compounds of the invention, and optionally one or more other therapeutic agents described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The compounds of the invention may be administered by any suitable means for any of the above uses, for example, orally, such as tablets, pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups and emulsions; sublingual delivery; is taken orally; parenteral, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., in the form of sterile injectable aqueous or nonaqueous solutions or suspensions); transnasally, including administration to the nasal membrane, such as by inhalation spray; topical, such as in the form of a cream or ointment; or rectally, such as in the form of suppositories; or intratumoral injection. They may be administered alone, but are typically administered using a drug carrier selected based on the chosen route of administration and standard pharmaceutical practice.
Pharmaceutical carriers are formulated according to a number of factors within the purview of one skilled in the art. These factors include, but are not limited to: the type and nature of the active agent formulated; a subject to whom the active agent-containing composition is to be administered; the intended route of administration of the composition; and targeted therapeutic indications. Pharmaceutically acceptable carriers include aqueous and nonaqueous liquid media and various solid and semi-solid dosage forms.
The carrier may include a number of different ingredients and additives in addition to the active agent, which other ingredients are included in the formulation for a variety of reasons known to those skilled in the art, such as stabilizing the active agent, binder, etc. For a description of suitable pharmaceutical carriers and the factors involved in carrier selection, see a number of readily available sources, for example, allen l.v. jr.et al remington: the Science and Practice of Pharmacy (2 Volumes), 22nd Edition (2012), pharmaceutical Press.
Of course, the dosage regimen of the compounds of the invention will vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health condition, medical condition and weight of the recipient; the nature and extent of the symptoms; the type of concurrent treatment; treatment frequency; the route of administration, the renal and hepatic function of the patient, and the desired effect. According to general guidelines, when used for the indicated effects, the daily oral dosage of each active ingredient should be from about 0.001 mg/day to about 10-5000 mg/day, preferably from about 0.01 mg/day to about 1000 mg/day, and most preferably from about 0.1 mg/day to about 250 mg/day. During constant infusion, the most preferred dosage for intravenous administration should be about 0.01 mg/kg/min to about 10 mg/kg/min. The compounds of the present invention may be administered in a single daily dose, or the total daily dose may be administered in divided doses of two, three or four times daily.
The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration (e.g., oral tablets, capsules, elixirs and syrups) and consistent with conventional pharmaceutical practices.
Dosage forms suitable for administration (pharmaceutical compositions) may contain from about 1 mg to about 2000 mg of active ingredient per dosage unit. In these pharmaceutical compositions, the active ingredient will typically be present in an amount of about 0.1 to 95% by weight, based on the total weight of the composition.
Typical capsules for oral administration contain at least one compound of the invention (250 mg), lactose (75 mg) and magnesium stearate (15 mg). The mixture was passed through a 60 mesh screen and packaged into size 1 gelatin capsules.
Typical injectable formulations can be prepared as follows: at least one compound of the invention (250 mg) is placed in a bottle in a sterile manner, lyophilized in a sterile manner and sealed. For use, the vial contents were mixed with 2mL of physiological saline to produce an injectable formulation.
The scope of the present invention includes pharmaceutical compositions (alone or in combination with a pharmaceutical carrier) comprising a therapeutically effective amount of at least one compound of the present invention as an active ingredient. Optionally, the compounds of the present invention may be used alone, in combination with other compounds of the present invention, or in combination with one or more other therapeutic agents (e.g., anticancer agents or other pharmaceutically active substances).
Regardless of the route of administration selected, the compounds of the invention (which may be used in a suitable hydrated form) and/or the pharmaceutical compositions of the invention are formulated into pharmaceutical dosage forms by conventional methods known to those skilled in the art.
The actual dosage level of the active ingredient in the pharmaceutical compositions of the present invention may be varied to achieve amounts of the active ingredient that are effective to achieve the desired therapeutic response, composition and mode of administration for a particular patient, but which are non-toxic to the patient.
The selected dosage level will depend on a variety of factors including the activity of the particular compound of the invention or an ester, salt or amide thereof employed; a route of administration; administration time; the rate of excretion of the particular compound being used; the rate and extent of absorption; duration of treatment; other drugs, compounds and/or substances used in combination with the particular compound used; the age, sex, weight, condition, general health and previous medical history of the patient being treated.
A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, to achieve a desired therapeutic effect, a physician or veterinarian may begin the relative amounts of the compound of the invention used in the pharmaceutical composition at a level less than that required and step up the dosage until the desired effect is achieved. In general, a suitable daily dose of a compound of the invention will be the amount of the compound at the lowest dose effective to produce a therapeutic effect. Such effective dosages will generally depend on the factors described above. Generally, oral, intravenous, intraventricular and subcutaneous dosages of the compounds of the invention for patients range from about 0.01 to about 50mg/kg body weight/day. If desired, an effective daily dose of the active compound may be administered separately at appropriate intervals throughout the day in two, three, four, five, six or more sub-doses, optionally in unit dosage form. In certain aspects of the invention, the administration is once daily.
Although the compounds of the present invention may be administered alone, it is preferable to administer the compounds in the form of a pharmaceutical formulation (composition). Kit/product package
For use in the treatment of the above indications, the kit/product package is also described herein. These kits may consist of a conveyor, a pack or a container box which may be divided into multiple compartments to hold one or more containers, such as vials, tubes and the like, each of which contains a separate one of the components of the method. Suitable containers include bottles, vials, syringes, test tubes, and the like. The container is made of acceptable glass or plastic materials.
For example, the container may contain one or more compounds described herein, either in the form of pharmaceutical compositions or as a mixture with other ingredients described herein. The container may have a sterile outlet (e.g., the container may be an iv bag or vial, and the vial stopper may be pierced by a hypodermic needle). Such kits may carry a compound, and instructions, tags, or instructions for use as described herein.
A typical kit may include one or more containers, each containing one or more materials (e.g., reagents, or concentrated mother liquor, and/or equipment) to accommodate commercial popularization and use of the compound by the user. Such materials include, but are not limited to, buffers, diluents, filters, needles, syringes, conveyors, bags, containers, bottles and/or tubes with a content list and/or instructions for use, and with instructions for packaging. The complete set of instructions is included.
The label may be displayed on or closely associated with the container. The appearance of a label on a container means that the label letters, numbers or other features are affixed, molded, engraved on the container; the label may also be present in a container box or shipping box containing a variety of containers, such as in a product insert. A label may be used to indicate a particular therapeutic use of the contents. The label may also indicate instructions for use of the content, such as described in the methods above.
All of the features described in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so described, may be present in any combination, unless certain features or steps are mutually exclusive in the same combination.
The above-mentioned features of the invention, or of the embodiments, may be combined in any desired manner. All of the features disclosed in this specification may be combined with any combination of the features disclosed in this specification, and the various features disclosed in this specification may be substituted for any alternative feature serving the same, equivalent or similar purpose. Thus, unless expressly stated otherwise, the disclosed features are merely general examples of equivalent or similar features.
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental procedures, which do not address the specific conditions in the examples below, are generally carried out under conventional conditions or under conditions recommended by the manufacturer. All percentages, ratios, proportions, or parts are by weight unless otherwise indicated.
The units in weight volume percent are well known to those skilled in the art and refer, for example, to the weight of solute in 100 milliliters of solution. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred methods and materials described herein are presented for illustrative purposes only.
Examples
Universal procedure
When the preparation route is not included, the raw materials and reagents used in the present invention are known products, and can be synthesized according to the methods known in the art, or can be obtained by purchasing commercial products. The commercial reagents used were all used without further purification.
Room temperature refers to 20-30 ℃.
Unless otherwise specified in the reaction examples, the reactions were all carried out under nitrogen atmosphere. The nitrogen atmosphere is defined as the reaction flask being attached to a balloon of about 1L of nitrogen.
The hydrogenation reaction is usually vacuumized, filled with hydrogen and repeatedly operated for 3 times. The hydrogen atmosphere is defined as the reaction flask being connected to a balloon of hydrogen gas of about 1L.
Microwave reaction is usedInitiator + microwave reactor.
The structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) and Mass Spectrometry (MS). NMR shift (. Delta.) of 10 -6 Units of (ppm) are given. NMR was determined using (Bruker Assetnd TM 500) nuclear magnetic resonance apparatus, the measuring solvent is deuterated dimethyl sulfoxide (DMSO-d 6), deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard is Tetramethylsilane (TMS). The following abbreviations are used for multiplicity of NMR signals: s=singlet, brs=broad, d=doublet, t=triplet, m=multiplet. Coupling constants are listed as J values, measured in Hz.
LC-MS was determined using a Thermo liquid chromatography apparatus (UltiMate 3000+MSQ PLUS). HPLC was determined using a Thermo high pressure liquid chromatograph (UltiMate 3000). Reverse phase preparative chromatography a Thermo (UltiMate 3000) reverse phase preparative chromatograph was used. Quick column chromatography using Ai Jieer (FS-9200T) automatic column passing machine, silica gel pre-packed column using SantaiAnd (5) preassembling the column. The specification of the thin layer chromatography separation and purification product adopted by the smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate is 0.4 mm-0.5 mm.
The synthesis method of some intermediates in the invention is as follows:
intermediate 1
Intermediate 1 was prepared by the following steps:
the first step: 1-methyl-3, 5-dinitropyridin-2-one Int-1a (1.0 g,5.02 mmol) was dissolved in methanol (50 mL) and methanolic ammonia solution (7 mol/L,8.61mL,60.27 mmol) and 1-methylpiperidin-4-one Int-1b (625 mg,5.52 mmol) were added sequentially. The reaction mixture was heated to 50 ℃ and stirred for 5 hours. After cooling to room temperature, the reaction mixture was allowed to stand for 48 hours, concentrated under reduced pressure, and the residue was added to ethyl acetate (50 mL) and filtered. The filtrate was concentrated under reduced pressure to give Int-1c (1.0 g) as a red solid, which was used directly in the next reaction. ESI-MS (m/z): 194.4[ M+H ] ] +1 H NMR(500MHz,DMSO-d 6 )δ9.14(d,J=2.5Hz,1H),8.36(d,J=2.5Hz,1H),3.64(s,2H),3.02(t,J=6.0Hz,2H),2.74(t,J=6.0Hz,2H),2.39(s,3H)。
And a second step of: the compound Int-1C (1.0 g) obtained in the previous step was dissolved in methanol (30 mL), 10% Pd-C (400 mg) was added thereto, and the mixture was reacted at room temperature under a hydrogen atmosphere for 6 hours. Palladium on carbon was removed by filtration, and the filtrate was concentrated to give Int-1d (800 mg, yield 94.70%) as a yellow solid. ESI-MS (m/z): 164.2[ M+H ]] +
And a third step of: compound Int-1d (100 mg,0.61 mmol) was dissolved in acetic acid (3 mL), N-bromosuccinimide (109 mg,0.61 mmol) was added, and the reaction mixture was stirred at room temperature for 1 hour. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate until no bubbles were generated, the aqueous phase was extracted with methanol/dichloromethane (1/20, 50 mL. Times.2), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated by filtration to give compound Int-1e (38 mg, yield 25%). ESI-MS (m/z): 242.3[ M+H ]] +1 H NMR(500MHz,DMSO-d 6 )δ6.77(s,1H),5.25(s,2H),3.37(s,2H),2.69(t,J=6.0Hz,2H),2.60(t,J=6.0Hz,2H),2.32(s,3H)。
Fourth step: compound Int-1e (37 mg,0.15 mmol) was dissolved in methanol (1 mL), and cuprous iodide (3 mg,0.015 mmol), 1, 10-phenanthroline (3 mg,0.03 mmol) and cesium carbonate (99 mg,0.30 mmol) were added. The reaction mixture was heated to 100 ℃ with microwaves after nitrogen substitution and stirred for 2 hours. The reaction was cooled to room temperature, the reaction mixture was concentrated, and the residue was purified by preparative thin layer chromatography (methanol/dichloromethane/triethylamine=1/10/0.1) to give Int-1 (20 mg, yield 67%) as a yellow solid. ESI-MS (m/z): 194.5[ M+H ] ] +1 H NMR(500MHz,DMSO-d 6 )δ6.54(s,1H),4.68(s,2H),3.80(s,3H),3.30(s,2H),2.64(t,J=5.6Hz,2H),2.59(t,J=5.7Hz,2H),2.31(s,3H)。
Intermediate 2
Intermediate 2 was prepared by the following steps:
the first step: compound Int-1e (230 mg,0.94 mmol) was dissolved in ethanol (2 mL) and cuprous iodide (18 mg,0.095 mmol), 1, 10-phenanthroline (34 mg,0.18 mmol) and cesium carbonate (612 mg,1.90 mmol) were added. The reaction mixture was heated to 100 ℃ with microwaves after nitrogen substitution and stirred for 5 hours. The reaction was cooled to room temperature, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography (methanol/dichloromethane/triethylamine=1/50/0.1) to give Int-2 (113 mg, yield 57%) as a yellow solid. ESI-MS (m/z): 208.5[ M+H ]] +
Intermediate 3
Intermediate 3 was prepared by the following steps:
the first step: compound Int-1e (100 mg,0.41 mmol) and trimethylcyclotriboroxane (148 mg,1.19 mmol) were dissolved in dioxane (1.5 mL) and water (0.15 mL), potassium carbonate (171 mg,1.24 mmol), pd (dppf) Cl were added 2 (30 mg,0.041 mmol). After nitrogen was replaced by the reaction system, the reaction system was heated to 140℃with microwaves and stirred for 1 hour. The reaction was cooled to room temperature, the reaction mixture was filtered through celite, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (methanol/dichloromethane=1/20) to give Int-3 (50 mg, yield 68%) as a yellow solid. ESI-MS (m/z): 178.6[ M+H ]] +
Intermediate 4
Intermediate 4 was prepared by the following steps:
The first step: compound Int-1e (350 mg,1.45 mmol) and potassium vinyltrifluoroborate (387 mg,2.89 mmol) were dissolved in 1, 4-dioxane (1.5 mL) and water (0.15 mL), and potassium carbonate (399 mg,2.89 mmol) and Pd (dppf) Cl were added 2 (105 mg,0.14 mmol). After nitrogen was replaced by the reaction system, the reaction system was heated to 120℃with a microwave reactor and stirred for 1 hour. After the reaction was cooled to room temperature, it was filtered through celite, the filtrate was concentrated, and the residue was separated by column chromatography (methanol/dichloromethane=1/20) to give Int-4a as a yellow solid (136 mg, yield 49%). ESI-MS (m/z): 190.7[ M+H ]] +1 H NMR(500MHz,CDCl 3 )δ6.85(dd,J=17.2,11.0Hz,1H),6.66(s,1H),6.16(dd,J=17.3,1.9Hz,1H),5.49(dd,J=11.0,1.9Hz,1H),3.67-3.63(m,2H),3.55(s,2H),3.00(t,J=6.1Hz,2H),2.80(t,J=6.1Hz,2H),2.49(s,3H)。
And a second step of: compound Int-4a (60 mg,0.31 mmol) was dissolved in methanol (5 mL), 10% palladium on carbon (20 mg) was added, and the mixture was stirred at room temperature under a hydrogen atmosphere for 1 hour. The reaction solution was filtered through celite, and the filtrate was concentrated to give intermediate 4 (37 mg, yield 61%). ESI-MS (m/z): 192.7[ M+H ]] +
Intermediate 5
Intermediate 5 was prepared by the following steps:
the first step: compound Int-1e (100 mg,0.41 mmol) was dissolved in a mixed solvent of toluene (3 mL) and water (0.3 mL), and cyclopropylboric acid (42 mg,0.49 mmol), potassium phosphate (306 mg,1.45 mmol), tricyclohexylphosphine (23 mg,0.082 mmol) and palladium acetate (9 mg,0.041 mmol) were added. The reaction system was heated to 100℃after nitrogen substitution and stirred for 18 hours. After the reaction was cooled to room temperature, the reaction solution was filtered through celite, the filtrate was concentrated, and the residue was separated by column chromatography (methanol/dichloromethane=1/20) to give Int-5 (61 mg, yield 72%) as a yellow solid. ESI-MS (m/z): 204.2[ M+H ] ] +
Intermediate 6
Intermediate 6 was prepared by the following steps:
the first step: N-Boc-4-piperidone Int-6a (4.4 g,22.1 mmol) and 1-methyl-3, 5-dinitro-2-pyridone Int-1a (4.0 g,20.1 mmol) were dissolved in methanol (150 mL) and methanolic ammonia solution (7N, 34.4mL,240.8 mmol) was added. Stirring at 60℃for 6 hours under nitrogen. The reaction solution was cooled to room temperature and stirred for 2 days. LCMS monitored the end of the reaction, the reaction was concentrated, ethyl acetate (150 mL) was added, stirred for half an hour, filtered, and the filtrate was concentrated to give Int-6b (5.1 g, 91% yield) as a yellow solid. ESI-MS (m/z): 280.1[ M+H ]] +
And a second step of: compound Int-6b (5.0 g,17.9 mmol) was dissolved in methanol (50 mL) and 10% palladium on carbon (500 mg) was added. The mixture was stirred at room temperature under a hydrogen atmosphere (hydrogen balloon) for 16 hours. After the completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated to give Int-6c (3.7 g, yield 84%) as a pale yellow solid. ESI-MS (m/z): 250.2[ M+H ]] +
And a third step of: compound Int-6c (3.7 g,14.8 mmol) was dissolved in DMF (20 mL) and N-bromosuccinimide (2.78 g,15.6 mmol) and acetic acid (370 mg) were added. The reaction mixture was stirred at room temperature for 2 hours and LCMS monitored the reaction to end. Water (100 mL) was added, the aqueous phase (150 mL. 3) was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was separated by silica gel column chromatography to give Int-6d (3.6 g, 74% yield) as a yellow solid. ESI-MS (m/z): 328.2[ M+H ] ] +
Fourth step: compound Int-6d (500 mg,1.53 mmol) was dissolved in methanol (5 mL) and sodium methoxide methanol solution (5N, 0.33mL,1.65 mmol) was added. The reaction mixture was heated to 100 ℃ with microwaves and stirred for 3 hours. The reaction was cooled to room temperature, the reaction solution was concentrated, and the residue was separated by silica gel column chromatography to give Int-6 (330 mg, yield 77%) as a yellow solid. ESI-MS (m/z): 280.2[ M+H ]] +
Example 1
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 1 was prepared by the following steps:
the first step: 2,4, 5-Trichloropyrimidine 1a (57 mg,0.31 mmol) and 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b (50 mg,0.28 mmol) were dissolved in isopropanol (2 mL) and N, N-diisopropylethylamine (85 mg,0.65mmol,0.11 mL) was added. The reaction mixture was stirred at room temperature for 18 hours. The reaction solution was filtered, and the solid was washed with isopropyl alcohol and dried to give white solid 1c (62 mg, yield 75%). ESI-MS (m/z): 285.2[ M+H ]] +
And a second step of: compound 1c (62 mg,0.21 mmol) and Int-1 (42 mg,0.21 mmol) were dissolved in 1, 4-dioxane (5 mL), brettPhos Pd G3 (19 mg,0.021 mmol), brettPhos (23 mg,0.043 mmol) and cesium carbonate (141 mg,0.43 mmol) were added, and the reaction system was heated to 100℃with nitrogen substitution and stirred for 18 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (methanol/dichloromethane=1/30) to give a crude product, which was separated by reverse phase preparative HPLC to give white solid 1 (25 mg, yield 26%). ESI-MS (m/z): 442.3[ M+H ] ] +1 H NMR(500MHz,DMSO-d6)δ8.00(s,1H),7.86(s,1H),7.69-7.61(m,2H),7.56(s,1H),7.08(d,J=6.8Hz,1H),6.19(t,J=6.8Hz,1H),4.37(d,J=5.9Hz,2H),3.86(s,3H),3.50(s,3H),3.12(s,2H),2.70(t,J=6.0Hz,2H),2.60(t,J=5.9Hz,2H),2.33(s,3H)。
Example 2
3- (((5-chloro-2- ((2-ethoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Substitution of Int-2 for Int-1 in the second step of example 1, in a similar manner and reaction procedure, gives compound 2.ESI-MS (m/z): 456.1[ M+H ]] +1 HNMR(500MHz,DMSO-d6)δ8.00(s,1H), 7.87(s,1H),7.69(t,J=5.9Hz,1H),7.64(dd,J=6.7,2.0Hz,1H),7.48(s,1H),7.12-7.06(m,1H),6.18(t,J=6.8Hz,1H),4.38(d,J=5.9Hz,2H),4.30(q,J=7.0Hz,2H),3.50(s,3H),3.09(s,2H),2.67(d,J=5.9Hz,2H),2.59(d,J=5.7Hz,2H),2.32(s,3H),1.32(t,J=7.0Hz,3H)。
Example 3
3- (((2- ((2-ethoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 3 can be obtained by a similar method and reaction procedure, substituting 2, 4-dichloro-5-trifluoromethylpyrimidine for 2,4, 5-trichloropyrimidine 1a in the first step of example 1, and then substituting Int-2 for Int-1 in the second step of example 1. ESI-MS (m/z): 490.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.23(s,1H),7.95(s,1H),7.76(s,1H),7.64(dd,J=6.7,2.0Hz,1H),7.59(br s,1H),7.02(d,J=7.0Hz,1H),6.19(t,J=6.8Hz,1H),4.40(d,J=5.6Hz,2H),4.30(q,J=7.0Hz,2H),3.50(s,3H),3.10(br s,2H),2.69(t,J=5.9Hz,2H),2.59(t,J=5.9Hz,2H),2.33(s,3H),1.31(t,J=7.0Hz,3H)。
Example 4
5- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) pyridin-2 (1H) -one
Compound 4 can be obtained by a similar method and reaction steps using 5- (aminomethyl) -1, 2-dihydropyridin-2-one instead of 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 428.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ7.99(s,1H),7.96(s,1H),7.74(s,1H),7.68(t,J=5.9Hz,1H),7.40(dd,J=9.4,2.6Hz,1H),7.20(br s,1H),6.27(d,J=9.4Hz,1H),4.29(d,J=5.6Hz,2H),3.88(s,3H),3.34-3.30(m,2H),2.74(t,J=5.8Hz,2H),2.63(t,J=5.9Hz,2H),2.33(s,3H)。
Example 5
3- (((5-chloro-2- ((2, 6-dimethyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Substitution of Int-3 for Int-1 in the second step of example 1, in a similar manner and reaction procedure, gives compound 5.ESI-MS (m/z): 426.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.36(s,1H),7.91(s,1H),7.63(dd,J=6.8,2.0Hz,1H),7.50-7.40(m,2H),7.08-6.96(m,1H),6.18(t,J=6.8Hz,1H),4.29(d,J=5.9Hz,2H),3.47(s,3H),3.19(s,2H),2.76(t,J=6.0Hz,2H),2.62(t,J=6.0Hz,2H),2.31(s,3H),2.30(s,3H)。
Example 6
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1,4, 6-trimethylpyridin-2 (1H) -one
Compound 6 was prepared by the following steps:
the first step: to a suspension of sodium hydride (202 mg,5.06mmol, 60% strength) in DMF (5 mL) was added a solution of compound 6a (500 mg,3.37 mmol) in DMF (5 mL) under an ice bath at 0deg.C and nitrogen atmosphere. After stirring for 30 minutes, methyl iodide (428 mg,5.06mmol,0.31 mL) was added and the mixture was stirred at room temperature for 16 hours. The reaction solution was diluted with ethyl acetate, and washed with water and then with saturated brine. The organic phase was dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 6b (230 mg, yield 42%) as a yellow solid. ESI-MS (m/z): 163.1[ M+H ]] +
And a second step of: compound 6b (230 mg,1.42 mmol) was dissolved in methanol (5 mL) and Raney Ni (0.5 mL, aqueous suspension) and aqueous ammonia (0.5 mL) were added. The mixture was stirred at room temperature under an atmosphere of hydrogen (hydrogen balloon) for 16 hours. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give compound 6c (200 mg) which was directly used in the next reaction. ESI-MS (m/z): 167.1[ M+H ] ] +
And a third step of: compound 6c (135 mg) was dissolved in isopropanol (2 mL), N-diisopropylethylamine (159 mg,1.23mmol,0.21 mL) and 2,4, 5-trichloropyrimidine 1a (150 mg,0.81 mmol) were added, and the mixture was stirred at room temperature for 16 hours. The reaction solution was concentrated, and the residue was separated by silica gel column chromatography (dichloromethane/methanol=10/1) to give 6d (130 mg) as a white solid. ESI-MS (m/z): 313.2[ M+H ]] +
Fourth step: compound 6d (71 mg,0.22 mmol) and Int-1 (40 mg,0.20 mmol) were dissolved in 1, 4-dioxane (4 mL), cesium carbonate (202 mg,0.62 mmol), brettPhos (22 mg, 0.041 mmol) and BrettPhos Pd G3 (18 mg, 0.020mmol) were added. The reaction system was heated to 100℃after nitrogen substitution and stirred for 16 hours. After the reaction solution was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by preparative thin layer chromatography (dichloroMethane/methanol=10/1) to give a crude product, which was separated by reverse phase preparative HPLC to give compound 6 (10 mg, yield 10%). ESI-MS (m/z): 470.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.07(s,1H),7.94(s,1H),7.69(s,1H),7.07(br s,1H),6.05(s,1H),4.44(d,J=5.5Hz,2H),3.88(s,3H),3.46-3.40(m,5H),2.75(br s,2H),2.65(t,J=5.5Hz,2H),2.34(s,3H),2.30(s,3H),2.12(s,3H)。
Example 7
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -4, 6-dimethylpyridin-2 (1H) -one
Compound 7 can be obtained by a similar method and reaction steps using 3- (aminomethyl) -4, 6-dimethyl-1, 2-dihydropyridin-2-one instead of 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 456.0[ M+H ] ] +1 HNMR(500MHz,DMSO-d6)δ11.59(br s,1H),8.09(s,1H),7.94(s,1H),7.69(s,1H),7.20(br s,1H),5.89(s,1H),4.42(br s,2H),3.89(s,3H),3.42(s,2H),2.75(br s,2H),2.65(br s,2H),2.34(s,3H),2.12(s,6H)。
Example 8
4- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) pyridin-2 (1H) -one
Compound 8 was obtained by a similar procedure and reaction steps using 4- (aminomethyl) -1, 2-dihydropyridin-2-one instead of 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 428.1[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.01(s,1H),7.90(s,1H),7.83(t,J=6.1Hz,1H),7.62(s,1H),7.31(d,J=6.7Hz,1H),6.11(dd,J=6.8,1.7Hz,1H),6.08(s,1H),4.41(d,J=5.8Hz,2H),3.86(s,3H),3.22(s,2H),2.71(t,J=5.9Hz,2H),2.61(t,J=5.9Hz,2H),2.33(s,3H)。
Example 9
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-ethylpyridin-2 (1H) -one
Compound 9 was prepared by the following steps:
the first step: compound 9a (700 mg,5.83 mmol) was dissolved in DMF (10 mL), cesium carbonate (2.28 g,6.99 mmol) was added, and then ethyl iodide (1.36 g,8.74mmol,0.70 mL) was added dropwise and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water, and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 9b (730 mg, yield 84%) as a white solid. ESI-MS (m/z): 149.4[ M+H ]] +1 HNMR(500MHz,Chloroform-d)δ7.80(dd,J=7.0,2.0Hz,1H),7.57(dd,J=7.0,2.0Hz,1H),6.28(t,J=7.0Hz,1H),4.05(q,J=7.0Hz,2H),1.40(t,J=7.0Hz,3H)。
And a second step of: compound 9b (100 mg,0.67 mmol) was dissolved in methanol (5 mL) and Raney Ni (0.3 mL, aqueous suspension) and aqueous ammonia (1 mL) were added. The mixture was stirred overnight at room temperature under a hydrogen atmosphere (hydrogen balloon). The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give compound 9c (100 mg) which was directly used for the next reaction.
And a third step of: compound 9c(100 mg) was dissolved in isopropyl alcohol (5 mL), N-diisopropylethylamine (169 mg,1.31mmol,0.23 mL) and 2,4, 5-trichloropyrimidine 1a (180 mg,0.98 mmol) were added, and the reaction mixture was stirred at room temperature overnight. The reaction solution was filtered, and the cake was washed with isopropyl alcohol and dried to give 9d (100 mg) of a white solid. ESI-MS (m/z): 299.2[ M+H ]] +
Fourth step: compound 9d (50 mg,0.16 mmol) and Int-1 (32 mg,0.16 mmol) were dissolved in 1, 4-dioxane (5 mL), cesium carbonate (108 mg,0.33 mmol), brettPhos (9 mg,0.016 mmol) and BrettPhos Pd G3 (15 mg,0.016 mmol) were added. The reaction system was heated to 100 ℃ after nitrogen substitution and stirred overnight. After the reaction solution was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by preparative thin layer chromatography (dichloromethane/methanol=10/1) to give a crude product, which was separated by reverse phase preparative HPLC to give compound 9 (8 mg, yield 11%). ESI-MS (m/z): 456.1[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.00(s,1H),7.91(s,1H),7.66-7.62(m,2H),7.56(s,1H),7.08(dd,J=7.0,2.0Hz,1H),6.20(t,J=7.0Hz,1H),4.39(d,J=6.0Hz,2H),3.98(q,J=7.0Hz,2H),3.85(s,3H),3.18(s,2H),2.70(t,J=6.0Hz,2H),2.60(t,J=6.0Hz,2H),2.32(s,3H),1.25(t,J=7.0Hz,3H)。
Example 10
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1,5, 6-trimethylpyridin-2 (1H) -one
Compound 10 was obtained by a similar procedure and reaction steps using 2-hydroxy-3-cyano-5, 6-dimethylpyridine in place of 3-cyano-2-hydroxy-4, 6-dimethylpyridine in the first step of example 9. ESI-MS (m/z): 470.1[ M+H ] ] +1 HNMR(500MHz,DMSO-d6)δ7.98(s,1H),7.93(s,1H),7.58(t,J=6.0Hz,1H),7.56(s,1H),6.94(s,1H),4.36(d,J=6.0Hz,2H),3.86(s,3H),3.52(s,3H),3.19(s,2H),2.71(t,J=6.0Hz,2H),2.60(t,J=6.0Hz,2H),2.33(s,3H),2.27(s,3H),1.99(s,3H)。
Example 11
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) pyridin-2 (1H) -one
Compound 11 can be obtained by a similar method and reaction procedure substituting 3- (aminomethyl) -pyridin-2 (1H) -one for 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1.ESI-MS (m/z): 428.3[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ11.68(br s,1H),8.00(s,1H),7.90(s,1H),7.65-7.62(m,1H),7.56(s,1H),7.33-7.28(m,1H),7.13-7.09(m,1H),6.15(t,J=6.6Hz,1H),4.35(d,J=5.9Hz,2H),3.86(s,3H),3.17(s,2H),2.69(d,J=5.9Hz,2H),2.60(d,J=5.7Hz,2H),2.33(s,3H)。
Example 12
3- (((5-chloro-2- ((2-cyclopropyl-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Substitution of Int-5 for Int-1 in the second step of example 1, followed by a similar procedure and reaction procedure, provided compound 12.ESI-MS (m/z): 452.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.55(s,1H),7.90(s,1H),7.62(dd,J=6.7,2.0Hz,1H),7.36(t,J=6.0Hz,1H),7.34(s,1H),6.97(d,J=6.9Hz,1H),6.15(t,J=6.8Hz,1H),4.29(d,J=6.0Hz,2H),3.46(s,3H),3.20(s,2H),2.71(t,J=6.0Hz,2H),2.61(t,J=6.0Hz,2H),2.31(s,3H),2.23-2.12(m,1H),0.85-.075(m,4H)。
Example 13
3- (((5-chloro-2- ((2-ethyl-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Substitution of Int-4 for Int-1 in the second step of example 1, in a similar manner and reaction procedure, compound 13 was obtained. ESI-MS (m/z): 440.2[ M+H ]] +1 HNMR(500MHz,DMSO-d6)δ8.36(s,1H),7.89(s,1H),7.62(dd,J=6.8,2.0Hz,1H),7.37(d,J=6.0Hz,1H),7.36(s,1H),6.98(d,J=6.9Hz,1H),6.16(t,J=6.8Hz,1H),4.27(d,J=6.0Hz,2H),3.46(s,3H),3.22(s,2H),2.78(t,J=6.0Hz,2H),2.69-2.61(m,4H),2.32(s,3H),1.07(t,J=7.5Hz,3H)。
Example 14
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyrrolidin-2-one
Compound 14 can be obtained by a similar method and reaction steps substituting 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1 with 3- (aminomethyl) -1-methylpyrrolidin-2-one. ESI-MS (m/z): 432.3[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.13(s,1H),7.96(s,1H),7.61(s,1H),7.38-7.33(m,1H),3.88(s,3H),3.65-3.58(m,1H),3.50-3.40(m,3H),3.27-3.24(m,2H),2.79-2.73(m,6H),2.66-2.62(m,2H),2.34(s,3H),2.08-2.02(m,1H),1.78-1.72(m,1H)。
Example 15
5- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) pyrrolidin-2-one
Compound 15 was obtained by a similar procedure and reaction steps substituting 5-aminomethyl-2-pyrrolidone for 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 418.4[ M+H ]] +1 HNMR(500MHz,DMSO-d6)δ8.10(s,1H),7.95(s,1H),7.75(br s,1H),7.67(s,1H),7.27(t,J=5.9Hz,1H),3.89(s,3H),3.82-3.77(m,1H),3.53-3.47(m,1H),3.41(s,2H),3.36-3.30(m,1H),2.75(t,J=5.9Hz,2H),2.64(t,J=6.3Hz,2H),2.34(s,3H),2.13-2.03(m,3H),1.78-1.71(m,1H)。
Example 16
3- (((2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) -5-methylpyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 16 was obtained by a similar procedure and reaction steps using 2, 4-dichloro-5-methylpyrimidine instead of 2,4, 5-trichloropyrimidine 1a in the first step of example 1. ESI-MS (m/z): 422.3[ M+H ]] +1 HNMR(500MHz,DMSO-d6)δ8.19(s,1H),7.74(s,1H),7.63(dd,J=6.8,1.9Hz,1H),7.35(br s,1H),7.31(br s,1H),7.15(d,J=6.8Hz,1H),6.19(t,J=6.8Hz,1H),4.41(d,J=5.9Hz,2H),3.96-3.85(m,5H),3.52(s,3H),3.30-3.22(m,2H),2.91(br s,2H),2.74(br s,2H),2.49(s,3H),2.02(s,3H)。
Example 17
3- (((5-fluoro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 17 can be obtained by a similar method and reaction procedure using 2, 4-dichloro-5-fluoropyrimidine instead of 2,4, 5-trichloropyrimidine 1a in the first step of example 1. ESI-MS (m/z): 426.3[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.03(s,1H),8.00-7.91(m,2H),7.64(dd,J=6.7,2.0Hz,1H),7.43(s,1H),7.19(dd,J=7.0,1.8Hz,1H),6.19(t,J=6.8Hz,1H),4.37(d,J=5.9Hz,2H),3.87(s,3H),3.50(s,3H),3.47(br s,2H),2.88(br s,2H),2.79(t,J=5.8Hz,2H),2.49(s,3H)。
Example 18
4- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 33 was obtained by a similar procedure and reaction steps substituting 4- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one for 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 442.3[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.01(s,1H),7.89(s,1H),7.84(t,J=5.5Hz,1H),7.66-7.60(m,2H),6.17-6.13(m,2H),4.41(d,J=5.9Hz,2H),3.85(s,3H),3.37(s,3H),3.21(br s,2H),2.71(t,J=5.3Hz,2H),2.61(t,J=5.7Hz,2H),2.33(s,3H)。
Example 19
3- (((5-chloro-2- ((2-methoxy-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 19 was prepared by the following steps:
the first step: compound 1b (244 mg,0.85 mmol) and Int-7 (200 mg,0.71 mmol) were dissolved in 1, 4-dioxane (5 mL), cesium carbonate (700 mg,2.15 mmol), brettPhos (76 mg,0.14 mmol) and BrettPhos Pd G3 (64 mg,0.071 mmol) were added. The reaction system was heated to 120℃after nitrogen substitution and stirred for 16 hours. After the reaction solution was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to give a crude product (300 mg) of compound 19a, which was directly used for the next reaction. ESI-MS (m/z): 528.1[ M+H ] ] +
And a second step of: the crude 19a product (300 mg) obtained in the previous step was dissolved in acetonitrile (5 mL), and p-toluenesulfonic acid monohydrate (323 mg,1.70 mmol) was added. The reaction mixture was warmed to 70 ℃ and stirred for 2 hours. The reaction solution was concentrated to obtain a crude product (240 mg) of compound 19, 20mg of which was purified by reverse phase preparative HPLC to obtain compound 19 (1 mg). ESI-MS (m/z): 428.3[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.00(s,1H),7.84(s,1H),7.69-7.60(m,2H),7.53(s,1H),7.09(d,J=6.9Hz,1H),6.18(t,J=6.8Hz,1H),4.37(d,J=5.9Hz,2H),3.85(s,3H),3.52(s,2H),3.49(s,3H),2.96(t,J=5.8Hz,2H),2.59(t,J=5.8Hz,2H)。
Example 20
2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) -4- (((1-methyl-2-carbonyl-1, 2-dihydropyridin-3-yl) methyl) amino) pyrimidine-5-carbonitrile
Compound 20 was obtained by a similar procedure and reaction steps substituting 2, 4-dichloro-5-cyanopyrimidine for 2,4, 5-trichloropyrimidine 1a in the first step of example 1. ESI-MS (m/z): 433.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.38(s,1H),8.24-8.19(m,1H),8.16(s,1H),7.71(s,1H),7.63-7.60(m,1H),6.98-6.94(m,1H),6.15-6.12(m,1H),4.16(d,J=6.1Hz,2H),3.85(s,3H),3.47(s,3H),3.14(br s,2H),2.73(d,J=6.1Hz,2H),2.63-2.60(m,2H),2.33(s,3H)。
Example 21
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1, 5-dimethylpyridin-2 (1H) -one
Compound 21 was prepared by the following steps:
the first step: 3-bromo-2-hydroxy-5-methylpyridine 21a (200 mg,1.06 mmol) was dissolved in tetrahydrofuran (5 mL), naH (50 mg,1.28mmol, content 60%) was added at 0deg.C, and the reaction was stirred for 30 minutes. Methyl iodide (227 mg,1.60 mmol) was added dropwise to the reaction solution, and the reaction solution was stirred at 0℃for 2 hours. The reaction mixture was quenched with saturated aqueous ammonium chloride and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/3) to give compound 21b (200 mg, yield 93%). ESI-MS (m/z): 202.5[ M+H ] ] +
And a second step of: compound 21b (180 mg,0.89 mmol) was dissolved in DMF (5 mL) and Pd (PPh) was added 3 ) 4 (103 mg,0.09 mmol) and Zn (CN) 2 (105 mg,0.89 mmol) and the reaction system was replaced with nitrogen, and then heated to 130℃with microwaves for 4 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/4) to give 21c (122 mg, yield 92%) as a white solid. ESI-MS (m/z): 149.4[ M+H ]] +
And a third step of: compound 21c (120 mg,0.81 mmol) was dissolvedTo methanol (5 mL) and aqueous ammonia (0.5 mL) were added Raney Nickel (0.3 mL, aqueous suspension) and BOC anhydride (212 mg,0.97 mmol), and the mixture was stirred overnight at room temperature under a hydrogen atmosphere (hydrogen balloon). The reaction solution was filtered through celite, and the filtrate was concentrated. The residue was dissolved in 4N dioxane hydrochloride solution (5 mL), and the reaction solution was stirred at room temperature for 1 hour. The reaction solution was concentrated to obtain a crude product of compound 21d, which was directly used for the next reaction. ESI-MS (m/z): 153.8[ M+H ]] +
Fourth step: the crude compound 21d obtained in the previous step and 2,4, 5-trichloropyrimidine 1a (169 mg,0.92 mmol) were dissolved in i-PrOH (5 mL), N-diisopropylethylamine (356 mg,2.76 mmol) was added, and the reaction was stirred at 90℃overnight. After the reaction solution was cooled to room temperature, the reaction solution was concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/4) to give 21e (98 mg, yield 36%) as a white solid. ESI-MS (m/z): 300.2[ M+H ] ] +
Fifth step: compound 21e (50 mg,0.17 mmol) and Int-1 (36 mg,0.18 mmol) were dissolved in 1, 4-dioxane (5 mL), and BrettPhos G3Pd (15 mg,17 umol), brettPhos (9 mg,17 umol) and cesium carbonate (109 mg,0.34 mmol) were added. The reaction system was heated to 100 ℃ after nitrogen substitution and stirred overnight. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin layer chromatography (petroleum ether/ethyl acetate=1/1) to give crude product of compound 21, which was purified by reverse phase preparative HPLC to give compound 21 (6 mg, yield 8%). ESI-MS (m/z): 456.2[ M+H ]] +1 H NMR(500MHz,DMSO-d 6 )δ7.99(s,1H),7.89(s,1H),7.62(t,J=6.0Hz,1H),7.57(s,1H),7.43(s,1H),6.97(s,1H),4.37(d,J=5.8Hz,2H),3.86(s,3H),3.45(s,3H),3.16(s,2H),2.70(t,J=5.7Hz,2H),2.60(t,J=5.9Hz,2H),2.33(s,3H),1.96(s,3H)。
Example 22
3- (((2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 22 was obtained by a similar procedure and reaction steps, substituting 2, 4-dichloropyrimidine for 2,4, 5-trichloropyrimidine 1a in the first step of example 1. ESI-MS (m/z): 408.3[ M+H ]] +1 HNMR(500MHz,DMSO-d6)δ8.08(br s,1H),7.83(d,J=5.8Hz,1H),7.68(br s,1H),7.63(dd,J=6.7,1.9Hz,1H),7.28(s,1H),7.19(br s,1H),6.19(t,J=6.8Hz,1H),6.10(br s,1H),4.30(br s,2H),3.88(s,3H),3.49(s,3H),3.19(br s,2H),2.70(t,J=6.6Hz,2H),2.62(t,J=5.7Hz,2H),2.33(s,3H)。
Example 23
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) -1, 4-dimethylpyridin-2 (1H) -one
Compound 23 was prepared by the following steps:
the first step: 3-bromo-2-hydroxy-4-methylpyridine 23a (200 mg,1.06 mmol) was dissolved in tetrahydrofuran (5 mL), naH (50 mg,1.28mmol, content 60%) was added at 0deg.C, and the reaction was stirred for 30 minutes. Methyl iodide (227 mg,1.60 mmol) was added dropwise to the reaction solution, and the reaction solution was stirred at 0℃for 2 hours. The reaction mixture was quenched with saturated aqueous ammonium chloride and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/3) to give 23b (210 mg, yield 98%) as a yellow oil. ESI-MS (m/z): 202.5[ M+H ] ] +
And a second step of: compound 23b (210 mg,1.04 mmol) was dissolved in DMF (5 mL) and addedPd (PPh) 3 ) 4 (120 mg,0.11 mmol) and Zn (CN) 2 (122 mg,1.04 mmol) and the reaction system replaced nitrogen and was then heated to 130℃with microwaves for 4 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/4) to give 23c (143 mg, yield 92%) as a white solid. ESI-MS (m/z): 149.4[ M+H ]] +
And a third step of: compound 23c (143 mg,0.97 mmol) was dissolved in methanol (5 mL) and aqueous ammonia (0.5 mL), raney Nickel (0.3 mL, aqueous suspension) and BOC anhydride (255 mg,1.16 mmol) were added and the mixture was stirred overnight at room temperature under a hydrogen atmosphere (hydrogen balloon). The reaction solution was filtered through celite, and the filtrate was concentrated. The residue was dissolved in 4N dioxane hydrochloride solution (5 mL), and the reaction solution was stirred at room temperature for 1 hour. The reaction solution was concentrated to give a crude product of compound 23d, which was directly used for the next reaction. ESI-MS (m/z): 153.5[ M+H ]] +
Fourth step: the crude compound 23d obtained in the previous step and 2,4, 5-trichloropyrimidine (176 mg,0.96 mmol) were dissolved in i-PrOH (5 mL), DIEA (372 mg,2.88 mmol) was added and the reaction stirred at 90℃overnight. After the reaction solution was cooled to room temperature, the reaction solution was concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/4) to give 23e (161 mg, yield 56%) as a white solid. ESI-MS (m/z): 300.2[ M+H ] ] +
Fifth step: compound 23e (50 mg,0.17 mmol) and Int-1 (32 mg,0.17 mmol) were dissolved in 1, 4-dioxane (5 mL), and BrettPhos G3Pd (15 mg,17 umol), brettPhos (9 mg,17 umol) and cesium carbonate (109 mg,0.34 mmol) were added. The reaction system was heated to 100 ℃ after nitrogen substitution and stirred overnight. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin layer chromatography (petroleum ether/ethyl acetate=1/1) to give crude compound 23, which was purified by reverse phase preparative HPLC to give compound 23 (7 mg, yield 9%). ESI-MS (m/z): 456.1[ M+H ]] +1 H NMR(500MHz,DMSO-d 6 )δ8.07(s,1H),7.95(s,1H),7.71(s,1H),7.56(d,J=6.9Hz,1H),7.14(t,J=5.4Hz,1H),6.12(d,J=7.0Hz,1H),4.47(d,J=5.4Hz,2H),3.88(s,3H),3.43(s,5H),2.76(t,J=5.8Hz,2H),2.65(t,J=5.9Hz, 2H),2.34(s,3H),2.16(s,3H)。
Example 24
3- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) pyrrolidin-2-one
Compound 24 was prepared by the following steps:
the first step: compound 3- (hydroxymethyl) pyrrolidin-2-one 24a (258 mg,2.24 mmol), phthalimide (362 mg2.4,7 mmol) and triphenylphosphine (646 mg,2.47 mmol) were dissolved in THF (5 mL) and diisopropyl azodicarboxylate (498 mg,2.47mmol,0.48 mL) was slowly added dropwise at 0deg.C. The reaction mixture was allowed to warm to room temperature and stirred for 18 hours, LCMS detected reaction complete. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography to give 24b (335 mg, yield 61%) as a white solid. ESI-MS (m/z): 245.5[ M+H ] ] +
And a second step of: compound 24b (335 mg,1.37 mmol) was dissolved in ethanol (5 mL), hydrazine hydrate ((171 mg,2.74mmol,0.17mL, content 80%) was added, the reaction mixture was stirred at 85℃for 2 hours and then cooled to room temperature, the reaction solution was filtered, and the filtrate was concentrated to give crude compound 24c (156 mg) which was used directly in the next reaction.
And a third step of: crude compound 24c (156 mg) obtained in the previous step and 2,4, 5-trichloropyrimidine 1a (501 mg,2.73 mmol) were dissolved in i-PrOH (5 mL), and DIPEA (529 mg,4.10 mmol) was added. The reaction mixture was stirred at room temperature for 18 hours. The reaction solution was concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 24d (300 mg, yield 84%) as a white solid. ESI-MSm/z):261.2[M+H] +
Fourth step: compound 24d (54 mg,0.20 mmol) and Int-1 (40 mg,0.20 mmol) were dissolved in 1, 4-dioxane (4 mL), cesium carbonate (134 mg,0.41 mmol), brettPhos (22 mg,0.041 mmol) and BrettPhos Pd G3 (18 mg, 0.020mmol) were added. The reaction system was heated to 110℃after nitrogen substitution and stirred for 16 hours. After the reaction solution was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by preparative thin layer chromatography to give a crude product, which was separated by reverse phase preparative HPLC to give compound 24 (17 mg, yield 20%). ESI-MS (m/z): 418.2[ M+H ] ] +1 HNMR(500MHz,DMSO-d6)δ8.15(s,1H),7.96(s,1H),7.74(br s,1H),7.61(s,1H),7.35-7.31(m,1H),3.88(s,3H),3.65-3.59(m,1H),3.48-3.37(m,3H),3.21-3.11(m,2H),2.74(t,J=5.9Hz,2H),2.69-2.61(m,3H),2.34(s,3H),2.14-2.06(m,1H),1.85-1.76(m,1H)。
Example 25
4- (((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) methyl) piperidin-2-one
Compound 25 was obtained by a similar procedure and reaction steps substituting 4- (aminomethyl) piperidin-2-one for 3- (aminomethyl) -1-methyl-1, 2-dihydropyridin-2-one hydrochloride 1b in the first step of example 1. ESI-MS (m/z): 432.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.07(s,1H),7.94(s,1H),7.64(s,1H),7.43(s,1H),7.39(t,J=5.9Hz,1H),3.88(s,3H),3.41(s,2H),3.31-3.27(m,2H),3.21-3.14(m,1H),3.10-3.03(m,1H),2.75(t,J=5.9Hz,2H),2.64(t,J=5.9Hz,2H),2.34(s,3H),2.23-2.15(m,2H),1.94-1.83(m,1H),1.81-1.74(m,1H),1.36-1.27(m,1H)。
Example 26
3- (((2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) methyl) -1-methylpyridin-2 (1H) -one
Compound 26 was obtained by a similar method and reaction procedure using 2, 4-dichloro-5-trifluoromethylpyrimidine instead of 2,4, 5-trichloropyrimidine 1a in the first step of example 1. ESI-MS (m/z): 476.2[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.23(s,1H),8.06(s,1H),7.74(s,1H),7.64(d,J=6.5Hz,1H),7.57(br s,1H),7.01(d,J=6.8Hz,1H),6.19(t,J=6.8Hz,1H),4.39(d,J=5.2Hz,2H),3.84(s,3H),3.50(s,3H),3.12(s,2H),2.71(d,J=6.9Hz,2H),2.60(t,J=5.9Hz,2H),2.33(s,3H)。
Example 27
(R) -3- (1- ((5-chloro-2- ((2-methoxy-6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) amino) pyrimidin-4-yl) amino) ethyl) -1-methylpyridin-2 (1H) -one
Compound 27 was prepared by the following steps:
the first step: compound 27a (500 mg,4.06 mmol) was dissolved in DMF (5 mL) and cesium carbonate (1.59 g,4.87 mmol) and methyl iodide (692 mg,4.87 mmol) were added at 0deg.C in ice and the reaction was warmed to room temperature and stirred for 16 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (40 mL x 2). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give compound 27b (290 mg, yield 52%) as a white solid. 1 H NMR(500MHz,CDCl 3 )δ10.37(s,1H),8.04(dd,J=7.0.4.5Hz,1H),7.63(dd,J=6.5,2,0Hz,1H),6.32(t,J=7.0Hz,1H),3.64(s,3H)。
And a second step of: compound 27b (200 mg,1.46 mmol) was dissolved in DCE (5 mL), cesium carbonate (950 mg,2.92 mmol) and S-tert-butylsulfinamide (212 mg,1.75 mmol) were added and the mixture was stirred at 65℃for 16 h. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (30 mL x 2). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give compound 27c (330 mg, yield 94%) as a pale yellow oil. ESI-MS (m/z): 241.4[ M+H ]] +1 H NMR(500MHz,DMSO-d6)δ8.71(s,1H),8.15(dd,J=6.0.2.0Hz,1H),8.05(dd,J=6.5,2,5Hz,1H),6.41(t,J=7.0Hz,1H),3.52(s,3H),1.15(s,9H)。
And a third step of: compound 27c (35 mg,0.14 mmol) was dissolved in DCM (3 mL) and methyl magnesium bromide (1 mol/L in THF,0.29 mL) was added under nitrogen at-78deg.C and the mixture stirred at-78deg.C for 2 h. After completion of the reaction, the reaction was quenched by addition of saturated ammonium chloride solution (20 mL) and the mixture was extracted with ethyl acetate (20 mL x 2). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give compound 27d (30 mg, yield 80%) as a pale yellow oil. ESI-MS (m/z): 257.5[ M+H ]] +1 H NMR (500 mhz, dmso-d 6) delta 7.61 (dd, j=7.0.2.0 hz, 1H), 7.46 (dd, j=6.5, 2,0hz, 1H), 6.25-6.20 (m, 1H), 5.68 (d, j=10.5 hz, 1H), 4.42-4.37 (m, 1H), 3.44 (s, 3H), 1.29 (d, j=6.5 hz, 3H), 1.10 (s, 9H). The steric configuration of the newly generated chiral center of compound 27d is assumed to be R-type.
Fourth step: compound 27d (30 mg,0.11 mmol) was dissolved in MeOH (2 mL) and dioxane hydrochloride solution (4 mol/L,0.12 mL) was added and the reaction mixture was stirred at room temperature for 2 hours. After the reaction was completed, the reaction solution was concentrated to give compound 27e (22 mg, yield 99%) as a white solid. ESI-MS (m/z): 153.1[ M+H ]] +
Fifth step: compound 27e (22 mg,0.11 mmol) was dissolved in isopropanol (2 mL) and 2,4, 5-trichloropyrimidine 1a (32 mg,0.17 mmol) and N, N-diisopropylethylamine (30 mg,0.23 mmol) were added. The reaction mixture was stirred at room temperature for 16 hours. To be reactedAfter completion, the reaction solution was concentrated, and the residue was purified by preparative thin layer chromatography (petroleum ether/ethyl acetate=1/1) to give compound 27f (31 mg, yield 88%) as a white solid. ESI-MS (m/z): 299.1[ M+H ]] +
Sixth step: compound Int-1 (20 mg,0.10 mmol) and compound 27f (31 mg,0.10 mmol) were dissolved in 1, 4-dioxane (3 mL), cesium carbonate (67 mg,0.20 mmol), brettPhos (11 mg, 0.020mmol) and BrettPhos Pd G3 (9 mg, 0.010mmol) were added. The reaction mixture was heated to 100 ℃ after nitrogen substitution and stirred for 18 hours. The reaction solution was filtered through celite, the filtrate was concentrated, and the residue was purified by preparative thin layer chromatography (dichloromethane/methanol=10/1) to give a crude product, which was separated by reverse phase preparative HPLC to give compound 27 (3 mg, yield 6%). ESI-MS (m/z): 299.1[ M+H ] ] +1 H NMR(400MHz,DMSO-d6)δ8.00(s,1H),7.99(s,1H),7.67-7.63(m,1H),7.57(s,1H),7.53(d,J=8.4Hz,1H),7.37-7.33(m,1H),6.26-6.19(m,1H),5.35-5.26(m,1H),3.87(s,3H),3.49(s,3H),3.45-3.43(m,2H),2.77-2.71(m,2H),2.68-2.60(m,2H),2.37(s,3H),1.44(d,J=6.8Hz,3H)。
The following examples were prepared according to the synthetic routes and synthetic methods of intermediates described in the examples above.
Biological screening and results for HPK1 inhibitors
Test example 1: detection of the ability of Compounds to inhibit the Activity of HPK1 kinase (method 1)
The required reagents are as follows
Experimental procedure
The specific operation is as follows: preparing an enzymatic reaction system buffer (10mM MOPS,pH 7.2,5mM beta-glycol-phosphate, 10mM MgCl2,0.8mM EDTA,2mM EGTA,0.1mM DTT); the compounds tested (1 mM stock of compound in DMSO) were diluted with buffer to a maximum concentration of 60uM (containing 6% DMSO) and a gradient of 8 spots in total of 5-fold dilution of the compound with buffer containing 6% DMSO was prepared starting at a concentration of 60 uM; the HPK1 kinase was then diluted to 30nM using buffer. Mu.l of HPK1 kinase diluent was added to each well of Greiner 384 well microplates (cat# 784075), and 2. Mu.l of buffer was added to control wells; after brief centrifugation, 1. Mu.l of the diluted compound was added to the reaction wells and 1. Mu.l of buffer containing 6% DMSO was added to the control wells; after brief centrifugation, the mixture was placed in a constant temperature incubator (Shanghai-Heng scientific instruments Co., ltd., product number: LRH-150) at 25℃for 20min. 3. Mu.l of the reaction substrate (10. Mu.M MBP and 20. Mu.M ATP in distilled water) was added to each well, centrifuged briefly and incubated in a constant temperature incubator at 25℃for 60min, and the enzymatic activity was detected by ADP-Glo Kinase Assay Kit, with ADP-Glo Kinase Assay Kit all according to the instructions of the kit. Data are described using half inhibition concentration IC50 of the compound.
Numbering of compounds IC50(nM) Numbering of compounds IC50(nM)
1 <0.1 2 88
3 45.73 5 18.52
6 2.6 7 <0.1
8 5.43 9 16.12
10 98.54 11 4.98
13 82.59 14 18.99
16 34.42 17 82.82
19 8.52 21 94.75
23 0.16 24 0.71
25 33.99 26 1.35
27 1.92 28 18.35
29 19.43 30 29.78
31 11.80 32 0.85
33 13.56 34 43.51
35 42.28 36 3.44
37 8.03 38 12.82
39 19.01 40 2.06
42 81.06 43 4.26
44 0.48 45 29.40
46 44.97 47 20.97
48 17.94 49 5.78
50 9.67 51 14.61
52 9.00 53 2.69
Test example 2: detection of the agonistic Capacity of Jurkat cells to secrete the cytokine interleukin-2 (IL-2) (method 2)
The reagents and cells to be used are as follows
Experimental reagent:
reagent(s) Branding Goods number Preservation conditions
RPMI1640 BI 01-100-1ACS 4℃
FBS BI 04-002-1A -80℃
BSA Amresco 0332-1KG 4℃
Phosphate buffer solution BI 02-024-1ACS 4℃
Tween-20 Solarbio T8220 RT
Anti-human CD3 Antibody invitrogen 16-0037-85 4℃
Anti-human CD28 Antibody invitrogen 16-0289-85 4℃
Human IL-2 DuoSet ELISA R&D DY202 4℃
Experimental cells:
cells Cell type Branding
Jurkat E6-1 Human T lymphocyte leukemia cells Cell bank of Chinese academy
Experimental procedure
The specific operation is as follows: compound powder was dissolved in DMSO to 10mM, 2 μl of compound was added to 998 μl RPMI 1640 medium (10% fbs in each of the experiments) and vortexed to the highest concentration point. The compound solution was gradually diluted 3-fold with 0.2% dmso medium for a total of 8 concentration points. As a control, RPMI 1640 medium solution containing dmso at a concentration of 0.1% was used. 1X 105Jurkat E6-1 cells were added to each well of a Corning 96 well cell culture plate (cat# 3599), followed by an equal volume of compound dilution, control addition of RPMI 1640 medium containing 0.2% DMSO, and incubation in a 37℃cell incubator (Thermo Fisher Scientific, model: 3111) for 1h. Then adding Anti-human CD3 Anti-body and Anti-human CD28 Anti-body antibodies with the final concentration of 1 mug/ml, and placing the mixture in a cell culture incubator at 37 ℃ for incubation for 24 hours. IL-2 content in cell supernatants was detected using a Human IL-2 DuoSet ELISA KIT ELISA assay performed according to the instructions of the kit. Data are described as the highest fold ratio of the stimulation signal of the compound to the signal of 0.1% dmso.
Test example 3: detection of the agonistic Capacity of the Compounds to secrete the cytokine interleukin-2 (IL-2) by human PBMC cells (method 3)
The required reagents are as follows
Reagent(s) Branding Goods number Preservation conditions
RPMI1640 BI 01-100-1ACS 4℃
FBS BI 04-002-1A -80℃
BSA Amresco 0332-1KG 4℃
Phosphate buffer solution BI 02-024-1ACS 4℃
Tween-20 Solarbio T8220 RT
Anti-human CD3 Antibody invitrogen 16-0037-85 4℃
Anti-human CD28 Antibody invitrogen 16-0289-85 4℃
Human IL-2 DuoSet ELISA R&D DY202 4℃
Experimental cell source information:
experimental procedure
The specific operation is as follows: human PBMC were removed from liquid nitrogen according to standard procedures, thawed and resuscitated in a 37℃water bath, and cells were resuspended in RPMI 1640 medium (10% FBS in each case), washed twice by centrifugation; human PBMC cells were then resuspended in RPMI 1640 medium for use. The compound powder was dissolved to 10mM with DMSO, 2. Mu.l of the compound was added to 998. Mu.l of RPMI 1640 medium, and the mixture was vortexed to the highest concentration point. The compound solution was gradually diluted 3-fold with 0.2% dmso medium for a total of 8 concentration points. As a control, RPMI 1640 medium solution containing dmso at a concentration of 0.1% was used. 1X 105 human PBMC cells were added to each well of a Corning 96 well cell culture plate (cat# 3599), followed by an equal volume of compound dilution, control group addition of RPMI 1640 medium containing 0.2% DMSO, and incubation in a 37℃cell incubator (Thermo Fisher Scientific, model: 3111) for 1h. Then adding Anti-human CD3 Anti-body Antibody with the final concentration of 0.01 mug/ml and Anti-human CD28 Anti-body Antibody with the final concentration of 1 mug/ml, and placing the mixture in a cell incubator at 37 ℃ for incubation for 24 hours. IL-2 content in cell supernatants was detected using a Human IL-2 DuoSet ELISA KIT ELISA assay performed according to the instructions of the kit. Data are described as the highest fold ratio of the stimulation signal of the compound to the signal of 0.1% dmso.
The above results indicate that the compounds of the present invention can significantly increase IL-2.

Claims (27)

  1. A compound having the structure of formula (I) or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof:
    wherein the method comprises the steps of
    X represents N or CR 10
    R 1 Represents hydrogen, C 1 -C 6 Alkyl, halogenated C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, halo C 3 -C 6 Cycloalkyl, halogen, cyano or-C (O) NH 2
    R 2 Represents hydrogen, halogen, hydroxy, (C) 1 -C 6 ) Alkyl, (C) 2 -C 6 ) Alkenyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) (4-8 membered) heterocycloalkyl, (C) 1 -C 6 ) Alkoxy, (C) 3 -C 8 ) Cycloalkyl (C) 0 -C 6 Alkylene) oxy or (4-8 membered heterocycloalkyl) (C 0 -C 6 Alkylene) oxy;
    R 3 represents hydrogen or C 1 -C 6 Alkyl, C 3 -C 6 Alkenyl, C 3 -C 6 Cycloalkyl, hydroxy (C) 1 -C 6 Alkyl, halo C 1 -C 6 Alkyl, 4-8 membered heterocycloalkyl;
    R 4 represents hydrogen or C 1 -C 6 An alkyl group; and said C 1 -C 6 The alkyl group may be optionally substituted with a substituent selected from the group consisting of: halogen, -OR a 、-SR a 、-P(O)R a R b 、-S(O) 2 R a 、-S(O)(NH)R a 、-S(O)R a 、-S(O) 2 NR a R b 、-C(O)NR a R b 、-C(O)OH、-OC(O)NR a R b 、-NR a C(O)R b 、-NR a S(O) 2 R b
    R 5 And R is 5’ Each independently represents hydrogen, C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, halogen, halo C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 An alkyl group);
    or R is 5 And R is R 5’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
    R 6 and R is 6’ Each independently represents hydrogen, C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, halogen, halo C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 An alkyl group);
    or R is 6 And R is R 6’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
    p represents 1 or 2;
    R 7 and R is 7’ Each independently represents hydrogen, C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, halogen, halo C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 An alkyl group);
    or R is 7 And R is R 7’ Together with the carbon atoms to which they are attached form a 3-6 membered ring, which ring may optionally contain 0, 1 or 2 heteroatoms selected from N, O, S;
    w represents
    Wherein A represents
    Represents that the ring contains a single bond or a double bond;
    when the bond between A and A 'is a double bond, A' represents CR a Or N, B represents CR b
    When the bond between A and A 'is a single bond, A' represents CHR a B represents CHR b Or is absent;
    R 8 and R is 9 Each independently represents hydrogen or C 1 -C 6 An alkyl group; or R is 8 And R is R 9 Can be combined with W 1 And W is 2 Together forming a 3-6 membered ring;
    W 1 represents C, W 2 Represents CH or N;
    R M and R is N Each independently represents hydrogen or C 1 -C 6 An alkyl group;
    R 10 represents hydrogen, halogen, (C) 1 -C 6 ) Alkyl, (C) 2 -C 6 ) Alkenyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) 4-10 membered heterocycloalkyl, - (C) 0 -C 6 Alkylene group) (C) 6 -C 10 ) Aryl or- (C) 0 -C 6 Alkylene) a 5-to 10-membered heteroaryl,
    alternatively, when X represents CR 10 When R is 10 Can be adjacent to R 2 Together forming a 5-10 membered cycloalkyl or 5-10 membered heterocycloalkyl;
    wherein R is a 、R b Each independently represents hydrogen, C 1 -C 6 Alkyl, hydroxy (C) 1 -C 6 Alkyl) or halogenated C 1 -C 6 An alkyl group;
    m and n represent 0, 1 or 2, respectively.
  2. The compound of claim 1, or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein,
    w represents
    Wherein A represents
    Represents that the ring contains a single bond or a double bond;
    each o independently represents 0 or 1;
    R 8 represents hydrogen or C 1 -C 6 An alkyl group;
    R 9 each independently represents hydrogen or C 1 -C 6 An alkyl group;
    or R is 8 With adjacent R 9 Together with the carbon atoms to which they are attached, form a 3-6 membered ring;
    W 2 、R M 、R N the method of claim 1.
  3. The compound of claim 2, or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein,
    w representsOr alternatively
    Represents that the ring contains a single bond or a double bond;
    each o independently represents 0 or 1;
    R 8 represents hydrogen or C 1 -C 6 An alkyl group;
    R 9 each independently represents hydrogen or C 1 -C 6 An alkyl group;
    or R is 8 With adjacent R 9 Together with the carbon atoms to which they are attached, form a 3-6 membered ring;
    W 2 、R M 、R N the method of claim 1.
  4. A compound according to claim 3, or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, wherein W is an aromatic heterocycle.
  5. A compound according to claim 3, or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, wherein W is a saturated heterocycle.
  6. A compound according to claim 1, or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, wherein ring W represents Or alternatively
  7. A compound according to claim 6, wherein ring W represents
  8. The compound according to claim 7, wherein ring W represents
  9. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, wherein R 1 Represents hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl, cyano or-C (O) NH 2
  10. The compound of claim 9, or a pharmaceutically acceptable salt isotope derivative thereof, or a stereoisomer thereof, wherein R 1 Represents hydrogen, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
  11. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 2 Represents hydroxy, (C) 1 -C 6 ) Alkyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) (4-8 membered) heterocycloalkyl, (C) 1 -C 6 ) An alkoxy group.
  12. As claimed inThe compound of claim 11, or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R 2 Represent C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyl groups.
  13. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 3 Represents hydrogen or C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, halo C 1 -C 6 An alkyl group;
  14. the compound of claim 13, or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R 3 Represents hydrogen or C 1 -C 6 An alkyl group.
  15. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 4 Represents hydrogen or C 1 -C 6 An alkyl group; and said C 1 -C 6 The alkyl group may be optionally substituted with a substituent selected from the group consisting of: halogen, -OR a 、-SR a 、-P(O)R a R b 、-S(O) 2 R a 、-S(O)(NH)R a 、-S(O)R a 、-S(O) 2 NR a R b 、-C(O)NR a R b 、-C(O)OH。
  16. The compound of claim 15, or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R 4 Represents hydrogen or C 1 -C 6 An alkyl group. 17. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 5 、R 5’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
  17. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 6 、R 6’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
  18. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 7 、R 7’ Each independently represents hydrogen, halogen, C 1 -C 6 Alkyl, halogenated C 1 -C 6 Alkyl or hydroxy (C) 1 -C 6 Alkyl). 20. A compound according to any one of the preceding claims or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 8 Represents hydrogen or C 1 -C 6 An alkyl group.
  19. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 9 Represents hydrogen or C 1 -C 6 An alkyl group.
  20. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R 10 Represents hydrogen, halogen, (C) 1 -C 6 ) Alkyl, - (C) 0 -C 6 Alkylene group) (C) 3 -C 8 ) Cycloalkyl, - (C) 0 -C 6 Alkylene) 4-10 membered heterocycloalkyl.
  21. The compound of claim 22, or a pharmaceutically acceptable salt isotope derivative thereof, or a stereoisomer thereof, wherein R 10 Represents hydrogen, halogen or (C) 1 -C 6 ) An alkyl group.
  22. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R M Represents hydrogen or C 1 -C 6 An alkyl group.
  23. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt isotopic derivative or stereoisomer thereof, wherein R a 、R b Each independently represents hydrogen, C 1 -C 6 Alkyl or halo C 1 -C 6 An alkyl group.
  24. A compound having the structure:
  25. a pharmaceutical composition comprising a compound according to any one of the preceding claims or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, and a pharmaceutically acceptable carrier.
  26. Use of a compound according to any one of claims 1 to 26 or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof or a pharmaceutical composition according to claim 27 for the manufacture of a medicament for the prevention and/or treatment of cancer, tumour, inflammatory disease, autoimmune disease or immune mediated disease.
  27. Use of a compound according to claims 1-26, or a pharmaceutically acceptable salt, isotopic derivative or stereoisomer thereof, for the prevention and/or treatment of cancer, tumor, inflammatory disease, autoimmune disease or immune mediated disease.
CN202280013620.1A 2021-04-08 2022-04-07 High-activity HPK1 kinase inhibitor Pending CN116888100A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2021103784241 2021-04-08
CN202110378424 2021-04-08
PCT/CN2022/085446 WO2022214009A1 (en) 2021-04-08 2022-04-07 High-activity hpk1 kinase inhibitor

Publications (1)

Publication Number Publication Date
CN116888100A true CN116888100A (en) 2023-10-13

Family

ID=83546012

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280013620.1A Pending CN116888100A (en) 2021-04-08 2022-04-07 High-activity HPK1 kinase inhibitor

Country Status (3)

Country Link
CN (1) CN116888100A (en)
TW (1) TW202304434A (en)
WO (1) WO2022214009A1 (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE530530T1 (en) * 2002-06-28 2011-11-15 Astellas Pharma Inc DIAMINOPYRIMIDINE CARBONIC ACID AMIDE DERIVATIVE
CA2529611C (en) * 2002-12-20 2009-12-15 Pfizer Products Inc. Pyrimidine derivatives for the treatment of abnormal cell growth
MXPA06013165A (en) * 2004-05-14 2007-02-13 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth.
MX2019011511A (en) * 2017-03-30 2019-11-18 Hoffmann La Roche Naphthyridines as inhibitors of hpk1.
CN112243439A (en) * 2018-06-13 2021-01-19 百济神州有限公司 Pyrrolo [2,3-B ] pyridines or pyrrolo [2,3-B ] pyrazines as HPK1 inhibitors and uses thereof

Also Published As

Publication number Publication date
WO2022214009A1 (en) 2022-10-13
TW202304434A (en) 2023-02-01

Similar Documents

Publication Publication Date Title
US6653471B2 (en) Heterocyclic compounds as ligands of the GABAA receptor
WO2018045956A1 (en) Benzimidazole compound kinase inhibitor, preparation method therefor and application thereof
AU2017302182A1 (en) Substituted thiazolo-pyridine compounds as MALT1 inhibitors
JP5824068B2 (en) Novel triazolylphenylsulfonamides as serine / threonine kinase inhibitors
KR20130041068A (en) Tetrahydro-pyrido-pyrimidine derivatives
JP6322275B2 (en) N- (2-cyanoheterocyclyl) pyrazolopyridone as Janus kinase inhibitor
EP4074710A1 (en) Pyrazolo[1,5-a]pyridine compound, preparation method therefor and use thereof
CN116348469A (en) High-activity Wnt pathway inhibitor compound
CN116685585A (en) High-activity HPK1 kinase inhibitor
JP2020537645A (en) Amine-substituted heterocyclic compounds as EHMT2 inhibitors and their derivatives
CN116655602A (en) PI3K alpha allosteric inhibitors
CN112313220B (en) PD-L1 antagonist compounds
JP7349750B2 (en) Aromatic heterocyclic compounds with kinase inhibitory activity
CN116964053A (en) Wnt pathway inhibitor compound
BR112020026337A2 (en) TRICYCLIC COMPOUNDS
TWI768781B (en) TRANSFORMING GROWTH FACTOR-β RECEPTOR INHIBITOR
CN116888100A (en) High-activity HPK1 kinase inhibitor
TW202222797A (en) Pyrazolo[1,5-a]pyridine compound, preparation method therefor and use thereof
CA3151039A1 (en) Substituted benzimidazole carboxamides and their use in the treatment of medical disorders
CN115066423A (en) PD-L1 antagonist compounds
CN116848103A (en) High activity HPK1 kinase inhibitors
CN116348117A (en) HPK1 kinase inhibitor compounds
CN117903169A (en) Pan-KRAS inhibitor compound
CN116848115A (en) Wnt pathway inhibitor compounds
CN117396482A (en) Wnt pathway inhibitor compound

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