US20210230138A1 - 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine - Google Patents

1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine Download PDF

Info

Publication number
US20210230138A1
US20210230138A1 US17/223,736 US202117223736A US2021230138A1 US 20210230138 A1 US20210230138 A1 US 20210230138A1 US 202117223736 A US202117223736 A US 202117223736A US 2021230138 A1 US2021230138 A1 US 2021230138A1
Authority
US
United States
Prior art keywords
cancer
tumor
disease
mmol
compound
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.)
Abandoned
Application number
US17/223,736
Inventor
Biao Lu
Junzhen ZHANG
Fangfang JIN
Feng He
Weikang Tao
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.)
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical 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 Jiangsu Hengrui Medicine Co Ltd, Shanghai Hengrui Pharmaceutical Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Priority to US17/223,736 priority Critical patent/US20210230138A1/en
Publication of US20210230138A1 publication Critical patent/US20210230138A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention belongs to the field of medicine, and relates to a 1,2,4-triazin-3-amine derivative of formula (I), a method for preparing the same, a pharmaceutical composition comprising the same, a use thereof as a therapeutic agent, in particular as an A 2a receptor antagonist, and a use thereof in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A 2a receptor.
  • Adenosine is a naturally occurring purine nucleoside, and is an endogenous regulator of many physiological functions. It plays an important role in the regulation of the cardiovascular system, central nervous system, respiratory system, kidney, fat and platelets.
  • adenosine The action of adenosine is mediated by a family of G-protein coupled receptors. It is known currently that there are at least four subtypes of adenosine receptors, which are classified into A 1 , A 2a , A 2b and A 3 . Among them, the A 1 and A 3 receptors inhibit the activity of the enzyme adenylate cyclase, whereas the A 2a and A 2b receptors stimulate the activity of the same enzyme, thereby modulating the level of cyclic AMP in cells. Adenosine regulates a wide range of physiological functions through these receptors.
  • the A 2a receptor (A 2a R) is widely distributed in the body, and is mainly expressed in the striatum in the central nervous system, and is also expressed in tissues such as the periphery, heart, liver, lung and kidney.
  • adenosine A 2a receptor antagonists have surprising efficacy in the treatment of neurodegenerative diseases, primarily Parkinson's disease, Huntington's disease or Alzheimer's disease ( Trends in Neurosci. 2006, 29(11), 647-654 ; Expert Opinion on Therapeutic Patents, 2007, 17, 979-991 and the like).
  • adenosine A 2a receptor antagonists can also be used to treat other central nervous system (CNS) related diseases such as depression, restless syndrome, sleep disorders and anxiety disorders ( Clin. Neuropharmacol. 2010, 33, 55-60 ; J Neurosci. 2010, 30 (48), 16284-16292 ; Parkinsonisn Relat. Disord. 2010, 16 (6), 423-426; and references therein: Mov. Disorders, 2010, 25(2), S305).
  • CNS central nervous system
  • adenosine A 2a receptor antagonists also have therapeutic potential as neuroprotective agents (see Jenner P. J Neurol. 2000; 24 7Supp12: 1143-50).
  • the activation of the adenosine A 2a receptor can exert an important immunomodulatory effect in many pathological processes such as ischemia, hypoxia, inflammation, trauma, transplantation and the like, which may be related to the higher expression level of the A 2a receptor in various immune cells such as T cells, B cells, monocyte macrophages, neutrophils and the like.
  • the activation of the A 2a receptor can promote the body to generate immune tolerance, and closely participate in the formation of “immune escape” or “immunosuppression” of tumor cells, thereby creating a favorable condition for the occurrence and development of tumors. Lokshin and his colleagues ( Cancer Res. 2006, Aug.
  • a 2a receptor can promote the proliferation of tumor cells such as melanoma A375 cells, fibroblast NIH3T3 cells, pheochromocytoma PC12 cells and the like, which may be related to the fact that the activation of the A 2a receptor in T cells can inhibit T cell activation, proliferation, adhesion to tumor cells, and produce cytotoxic effect on tumor cells.
  • tumor cells such as melanoma A375 cells, fibroblast NIH3T3 cells, pheochromocytoma PC12 cells and the like, which may be related to the fact that the activation of the A 2a receptor in T cells can inhibit T cell activation, proliferation, adhesion to tumor cells, and produce cytotoxic effect on tumor cells.
  • the anti-tumor immunity of CD8 + T cells is enhanced, and the tumor proliferation is significantly inhibited.
  • a 2a receptor antagonists can be used in the treatment of tumor.
  • Deepak Mittal et al. find that the combination administration of A 2b receptor inhibitors with chemotherapeutic drugs or immunological checkpoint inhibitors can significantly reduce tumor metastasis in a mice triple negative breast cancer model; the knockout of the A 2b receptor in mice or human colon cancer cell line significantly reduces colon cancer metastasis and cell tumorigenicity; meanwhile, the study finds that the A 2b receptor is highly expressed in human triple negative breast cancer cell line, and the expression level of the A 2b receptor is closely related to tumor progression. These results show that inhibition of the A 2b receptor can inhibit tumor metastasis, and the A 2b receptor is thus expected to be an ideal target for the treatment of tumors (Cancer Res. 2016 Aug. 1; 76(15):4372-82). The development of dual inhibitors of the A 2a receptor and the A 2b receptor has also become a direction worth exploring.
  • adenosine receptors Although compounds having significant biological activity on a variety of subtypes of adenosine receptors can have a therapeutic effect, they can cause undesired side effects. For example, during tissue ischemia/hypoxia, when cells of central system, circulatory system, digestive system, and skeletal muscle are in an anoxic and hypoxic stress environment, extracellular aggregated adenosine initiates a corresponding protective mechanism by activating the adenosine A 1 receptor on the cell membrane, thereby increasing the tolerance of the cells to anoxia and hypoxia.
  • the A 1 receptor located on immune cells can promote cellular immune responses in a hypoxic environment. Moreover, the A 1 receptor can also reduce free fatty acids and triglycerides, and is involved in regulating blood glucose.
  • the continued blocking of the A 1 receptor can cause various adverse effects in the body tissues ( Chinese Pharmacological Bulletin, 2008, 24(5), 573-576). For example, it is reported that the blocking of the A 1 receptor will cause adverse effects such as anxiety, awakening and the like in animal models ( Basic & Clinical Pharmacology & Toxicology, 2011, 109 (3), 203-7).
  • the Adenosine Released by the adenosine A 3 receptor during myocardial ischemia exerts a strong protective effect in heart (as described by Gessi S et al, Pharmacol. Ther. 117 (1), 2008, 123-140).
  • the continued blocking of the A 3 receptor can increase the likelihood of complications caused by any pre-existing or developing ischemic heart disease such as angina or heart failure.
  • WO2011095625 discloses a 1,2,4-triazin-4-amine derivative of formula (A 1 ) and use thereof in the treatment of a disease or condition ameliorated by the inhibition of the A 1 receptor or the A 2a receptor.
  • This patent application discloses a total of more than 200 examples, among which there are only five examples in which the ring A is a fused aromatic ring.
  • the data in this patent application show that when ring A is a fused aromatic ring, the inhibition activity on the A 2a R is weak (see Table 1).
  • ring A of Example 1(lxxii) is naphthyl.
  • a derivative in which a nitrogen atom is introduced at the 5-position of the naphthyl shows a surprising activity, and the inhibition activity on the A 2a R is 30 to 1500 times or more than that of Example 1 (lxxii).
  • Such a strong inhibition activity cannot be expected when reading WO2011095625.
  • the present invention provides a novel structure of an adenosine A 2a receptor antagonist with a strong inhibition activity, and the compounds having such a structure also have a good inhibition effect on the adenosine A 2b receptor and a weak inhibition effect on the adenosine A 1 receptor and the adenosine A 3 receptor, exhibiting a good selectivity for the adenosine A 2a receptor. Meanwhile, the compounds having such a structure exhibit an excellent anti-tumor effect and pharmacokinetics activity.
  • the object of the present invention is to provide a compound of formula (I):
  • ring A is aryl or heteroaryl
  • G 1 , G 2 , G 3 and G 4 are identical or different and each are independently selected from the group consisting of C, CH and N;
  • R 1 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR 5 , —C(O)R 5 , —S(O) m R 5 , NH 2 S(O) m R 5 , —NR 6 R 7 , S(O) m NR 6 R 7 and —C(O)NR 6 R 7 , wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl
  • each R 2 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR 5 , —C(O)R 5 , —S(O) m R 5 , NH 2 S(O) m R 5 , —NR 6 R 7 , S(O) m NR 6 R 7 and —C(O)NR 6 R 7 , wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cyclo
  • each R 3 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, deuterated alkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR 5 , —C(O)R 5 , —S(O) m R 5 , NH 2 S(O) m R, —NR 6 R 7 , S(O) m NR 6 R 7 and —C(O)NR 6 R 7 , wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano
  • each R 4 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR 5 , —C(O)R 5 , —S(O) m R 5 , NH 2 S(O) m R 5 , —NR 6 R 7 , S(O) m NR 6 R 7 and —C(O)NR 6 R 7 , wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cyclo
  • R 5 is selected from the group consisting of hydrogen, alkyl, haloalkyl, amino, hydroxy, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 6 and R 7 are each independently selected from the group consisting of hydrogen, alkyl, haloalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a heterocyclyl, wherein the heterocyclyl contains one or two identical or different heteroatoms selected from the group consisting of N, O and S, and the heterocyclyl is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • n 0, 1 or 2;
  • r 0, 1, 2 or 3;
  • q 0, 1 or 2;
  • n 0, 1, 2, 3, 4 or 5.
  • the compound of formula (I) is a compound of formula (Iaa):
  • G 1 , G 2 , R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • the compound of formula (I) is a compound of formula (Ibb):
  • G 1 and G 2 are identical or different and are each independently CR a or N;
  • R a is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, deuterated alkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R c is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 1 , R 4 and n are as defined in formula (I).
  • the compound of formula (I) is a compound of formula (II):
  • ring A is selected from the group consisting of phenyl, pyridyl, thienyl and furanyl.
  • the compound of formula (I) is a compound of formula (III):
  • R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • R 1 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, cyano, cycloalkyl, haloalkyl, heterocyclyl and —C(O)NR 6 R;
  • R 6 and R 7 are as defined in formula (I);
  • the halogen is preferably fluorine, chlorine or bromine
  • the alkyl is preferably methyl, ethyl, isopropyl or n-butyl
  • the alkoxy is preferably methoxy or ethoxy
  • the cycloalkyl is preferably cyclopropyl, cyclopentyl or cyclohexyl.
  • each R 3 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, deuterated alkyl, alkoxy, cyano, cycloalkyl and heterocyclyl, wherein the alkyl and alkoxy are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, hydroxy, cyano, amino, nitro, cycloalkyl and heterocyclyl; the halogen is preferably fluorine, chlorine or bromine, the alkyl is preferably methyl, ethyl, isopropyl or n-butyl, the alkoxy is preferably methoxy or ethoxy, and the heterocyclyl is preferably piperidinyl, piperazinyl, morpholinyl or tetrahydropyranyl.
  • each R 4 is identical or different and each is independently selected from the group consisting of hydrogen, alkyl and halogen.
  • Typical compounds of the present invention include, but are not limited to:
  • the present invention relates to a method for preparing the compound of formula (I), comprising a step of:
  • X is halogen
  • G 1 -G 4 , R 1 -R 4 , r, q and n are as defined in formula (I).
  • the present invention relates to a method for preparing the compound of formula (Iaa), comprising a step of:
  • X is halogen
  • M is or
  • G 1 , G 2 , R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • the present invention relates to a method for preparing the compound of formula (II), comprising a step of:
  • X is halogen
  • R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • the present invention relates to a method for preparing the compound of formula (III), comprising a step of:
  • X is halogen
  • R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A 2a receptor.
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A 2b receptor.
  • the disease or condition ameliorated by the inhibition of the A 2a receptor or the A 2b receptor is selected from the group consisting of cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior; preferably cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer,
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain damage, neuroinflammation and addictive behavior, and preferably cancer.
  • a medicament for treating cancer depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirr
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endo
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating lung cancer, and preferably non-small cell lung cancer.
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for inhibiting the A 2a receptor.
  • the present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for inhibiting the A 2b receptor.
  • the present invention also relates to a method for inhibiting the A 2a receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • the present invention also relates to a method for inhibiting the A 2b receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • the present invention also relates to a method for treating a disease or condition ameliorated by the inhibition of the A 2a receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • the present invention also relates to a method for treating a disease or condition ameliorated by the inhibition of the A 2b receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, a diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • the present invention relates to a method for treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior, and preferably cancer, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • the present invention further relates to a method for treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer and the like), multiple myeloma, malignant lymphoma (reticular sarcoma, lympho
  • the present invention further relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as a medicament.
  • the present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as an A 2a receptor antagonist.
  • the present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as an A 2b receptor antagonist.
  • the present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating a disease or condition ameliorated by the inhibition of the A 2a receptor.
  • the present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating a disease or condition ameliorated by the inhibition of the A 2b receptor.
  • the present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior, and preferably cancer.
  • neurodegenerative disorder Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like
  • attention-related disorder extrapyramidal syndrome
  • abnormal movement disorder cirrhosis
  • liver fibrosis fatty liver
  • dermal fibrosis sleep
  • the present invention further relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head
  • compositions containing the active ingredient can be in a form suitable for oral administration, for example, a tablet, troche, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, syrup or elixir.
  • Oral compositions can be prepared according to any known method in the art for the preparation of pharmaceutical composition.
  • Such composition can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, colorants and preservatives, in order to provide a pleasing and palatable pharmaceutical preparation.
  • Tablets contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • An aqueous suspension contains the active ingredient in admixture with excipients suitable for the manufacture of an aqueous suspension.
  • the aqueous suspension can also contain one or more preservatives such as ethyl paraben or n-propyl paraben, one or more colorants, one or more flavoring agents, and one or more sweeteners.
  • An oil suspension can be formulated by suspending the active ingredient in a vegetable oil.
  • the oil suspension can contain a thickener.
  • the aforementioned sweeteners and flavoring agents can be added to provide a palatable formulation.
  • the active ingredient in admixture with the dispersants or wetting agents, suspending agent or one or more preservatives can be prepared as a dispersible powder or granule suitable for the preparation of an aqueous suspension by adding water.
  • Suitable dispersants or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, such as sweeteners, flavoring agents and colorants, can also be added. These compositions can be preserved by adding an antioxidant, such as ascorbic acid.
  • the pharmaceutical composition of the present invention can also be in the form of an oil-in-water emulsion.
  • the pharmaceutical composition can be in the form of a sterile injectable aqueous solution.
  • Acceptable vehicles or solvents that can be used are water, Ringer's solution or isotonic sodium chloride solution.
  • the sterile injectable formulation can be a sterile injectable oil-in-water micro-emulsion in which the active ingredient is dissolved in the oil phase.
  • the active ingredient is dissolved in a mixture of soybean oil and lecithin.
  • the oil solution is then added into a mixture of water and glycerol, and processed to form a micro-emulsion.
  • the injectable solution or micro-emulsion can be introduced into a patient's bloodstream by local bolus injection.
  • the solution and micro-emulsion are preferably administrated in a manner that maintains a constant circulating concentration of the compound of the present invention.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is Deltec CADD-PLUSTM 5400 intravenous injection pump.
  • the pharmaceutical composition can be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • a suspension can be formulated with suitable dispersants or wetting agents and suspending agents as described above according to known techniques.
  • the sterile injectable formulation can also be a sterile injectable solution or suspension prepared in a nontoxic parenterally acceptable diluent or solvent.
  • sterile fixed oils can easily be used as a solvent or suspending medium.
  • the compound of the present invention can be administrated in the form of a suppository for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures, but liquid in the rectum, thereby melting in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerin gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols with various molecular weights and fatty acid esters of polyethylene glycols.
  • the dosage of a drug depends on a variety of factors including but not limited to, the following factors: activity of a specific compound, age of the patient, weight of the patient, general health of the patient, behavior of the patient, diet of the patient, administration time, administration route, excretion rate, drug combination and the like.
  • the optimal treatment such as treatment mode, daily dose of the compound of formula (I) or the type of pharmaceutically acceptable salt thereof can be verified by traditional therapeutic regimens.
  • alkyl refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group comprising 1 to 20 carbon atoms, preferably an alkyl having 1 to 12 carbon atoms, and more preferably an alkyl having 1 to 6 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl,
  • an alkyl group is a lower alkyl having 1 to 6 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, and the like.
  • the alkyl group can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point.
  • the substituent group(s) is preferably one or more groups independently optionally selected from the group consisting of H atom, D atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkoxy refers to an —O-(alkyl) or an —O-(unsubstituted cycloalkyl) group, wherein the alkyl is as defined above.
  • alkoxy include methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy can be optionally substituted or unsubstituted.
  • the substituent group(s) is preferably one or more group(s) independently selected from the group consisting of H atom, D atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic hydrocarbon substituent group having 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 10 carbon atoms, and most preferably 3 to 6 carbon atoms.
  • monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl and the like.
  • Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring.
  • the cycloalkyl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point.
  • the substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heterocyclyl refers to a 3 to 20 membered saturated or partially unsaturated monocyclic or polycyclic hydrocarbon group, wherein one or more ring atoms are heteroatoms selected from the group consisting of N, O and S(O) m (wherein m is an integer of 0 to 2), but excluding —O—O—, —O—S— or —S—S— in the ring, with the remaining ring atoms being carbon atoms.
  • the heterocyclyl has 3 to 12 ring atoms wherein 1 to 4 atoms are heteroatoms, more preferably, 3 to 10 ring atoms wherein 1 to 4 atoms are heteroatoms, and more preferably 5 to 6 ring atoms wherein 1 to 3 atoms are heteroatoms.
  • monocyclic heterocyclyl include pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like.
  • Polycyclic heterocyclyl includes a heterocyclyl having a spiro ring, fused ring or bridged ring.
  • the ring of heterocyclyl can be fused to the ring of aryl, heteroaryl or cycloalkyl, wherein the ring bound to the parent structure is heterocyclyl.
  • Non-limiting examples thereof include:
  • the heterocyclyl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point.
  • the substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic ring or polycyclic fused ring (i.e., each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) having a conjugated 7-electron system, preferably 6 to 10 membered aryl, for example, phenyl and naphthyl.
  • the ring of aryl can be fused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to the parent structure is aryl ring.
  • Non-limiting examples thereof include:
  • the aryl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point.
  • the substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heteroaryl refers to a 5 to 14 membered heteroaromatic system having 1 to 4 heteroatoms selected from the group consisting of O, S and N.
  • the heteroaryl is preferably 5 to 10 membered heteroaryl, more preferably 5 or 6 membered heteroaryl, for example, furanyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, tetrazolyl and the like.
  • the ring of heteroaryl can be fused to the ring of aryl, heterocyclyl or cycloalkyl, wherein the ring bound to the parent structure is heteroaryl ring. Non-limiting examples thereof include:
  • the heteroaryl can be substituted or unsubstituted.
  • the substituent group(s) can be substituted at any available connection point.
  • the substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • haloalkyl refers to an alkyl group substituted by one or more halogens, wherein the alkyl is as defined above.
  • deuterated alkyl refers to an alkyl group substituted by one or more deuterium atoms, wherein the alkyl is as defined above.
  • hydroxy refers to an —OH group.
  • hydroxyalkyl refers to an alkyl group substituted by hydroxy(s), wherein the alkyl is as defined above.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • hydroxy refers to an —OH group.
  • amino refers to a —NH 2 group.
  • cyano refers to a —CN group.
  • nitro refers to a —NO 2 group.
  • carbonyl refers to a C ⁇ O group.
  • carboxylate group refers to a —C(O)O(alkyl) group or a —C(O)O(cycloalkyl) group, wherein the alkyl and cycloalkyl are as defined above.
  • acyl halide refers to a compound containing a —C(O)-halogen group.
  • the present invention also comprises the compounds of formula (I) in various deuterated forms.
  • Each of the available hydrogen atoms attached to the carbon atom can be independently replaced by a deuterium atom.
  • Those skilled in the art can synthesize a compound of formula (I) in a deuterated form with reference to the relevant literature.
  • Commercially available deuterated starting materials can be employed in the preparation of the compound of formula (I) in deuterated form, or they can be synthesized by conventional techniques with deuterated reagents including, but not limited to, deuterated borane, trideuterated borane in tetrahydrofuran, deuterated lithium aluminum hydride, deuterated iodoethane, deuterated iodomethane and the like.
  • “Optional” or “optionally” means that the event or circumstance described subsequently can, but need not, occur, and such a description includes the situation in which the event or circumstance does or does not occur.
  • the heterocyclyl optionally substituted by an alkyl means that an alkyl group can be, but need not be, present, and such a description includes the situation of the heterocyclyl being substituted by an alkyl and the heterocyclyl being not substituted by an alkyl.
  • “Substituted” refers to one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms, independently substituted by a corresponding number of substituents. It goes without saying that the substituents only exist in their possible chemical position. The person skilled in the art is able to determine whether the substitution is possible or impossible by experiments or theory without paying excessive efforts. For example, the combination of amino or hydroxy having free hydrogen and carbon atoms having unsaturated bonds (such as olefinic) may be unstable.
  • a “pharmaceutical composition” refers to a mixture of one or more of the compounds according to the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof with other chemical components, and other components such as physiologically/pharmaceutically acceptable carriers and excipients.
  • the purpose of the pharmaceutical composition is to facilitate administration of a compound to an organism, which is conducive to the absorption of the active ingredient so as to show biological activity.
  • a “pharmaceutically acceptable salt” refers to a salt of the compound of the present invention, which is safe and effective in mammals and has the desired biological activity.
  • a method for preparing the compound of formula (I) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof comprises the following steps of:
  • Step 1 a compound of formula A is reacted with boric aci or a orate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (I-A);
  • Step 2 the compound of formula (I-A) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (I).
  • the reagent that provides an alkaline condition includes organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide.
  • the inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • the catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • the above reactions are preferably carried out in a solvent.
  • the solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • X is halogen
  • G 1 -G 4 , R 1 -R 4 , r, q and n are as defined in formula (I).
  • a method for preparing the compound of formula (Iaa) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof comprises the following steps of
  • Step 1 a compound of formula (Iaa-2) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (Iaa-1);
  • Step 2 the compound of formula (Iaa-1) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (Iaa).
  • X is halogen
  • the reagent that provides an alkaline condition includes organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide.
  • the inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • the catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • the above reactions are preferably carried out in a solvent.
  • the solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • a method for preparing the compound of formula (Ibb) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof comprises the following steps of
  • Step 1 a compound of formula (Ibb-2) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (Ibb-1);
  • Step 2 the compound of formula (Ibb-1) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (Ibb).
  • X is halogen
  • G 1 , G 2 , R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • the reagent that provides an alkaline condition includes organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide.
  • the inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • the catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • the above reactions are preferably carried out in a solvent.
  • the solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • a method for preparing the compound of formula (II) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof comprises the following steps of
  • Step 1 a compound of formula (B) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (II-A);
  • Step 2 the compound of formula (II-A) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (II).
  • the reagent that provides an alkaline condition includes organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide.
  • the inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • the catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • the above reactions are preferably carried out in a solvent.
  • the solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • X is halogen
  • R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • a method for preparing the compound of formula (III) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof comprises the following steps of:
  • Step 1 a compound of formula (B) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (II-A);
  • Step 2 the compound of formula (II-A) is reacted with a compound of formula (III-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (III).
  • the reagent that provides an alkaline condition includes organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide.
  • the inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • the catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • the above reactions are preferably carried out in a solvent.
  • the solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • X is halogen
  • R 1 , R 3 , R 4 , r and n are as defined in formula (I).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • MS was determined by a FINNIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, type: Finnigan LCQ advantage MAX).
  • HPLC High performance liquid chromatography
  • Chiral HPLC was determined on an Agilent 1260 DAD high performance liquid chromatograph.
  • Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate was used as the thin-layer silica gel chromatography (TLC) plate.
  • TLC thin-layer silica gel chromatography
  • the dimension of the silica gel plate used in TLC was 0.15 mm to 0.2 mm, and the dimension of the silica gel plate used in product purification was 0.4 mm to 0.5 mm.
  • Yantai Huanghai 200 to 300 mesh silica gel was generally used as a carrier for silica gel column chromatography.
  • Prep Star SD-1 (Varian Instruments Inc.) or SFC-multigram (Berger Instruments Inc.) was used for chiral preparative column chromatography.
  • the average kinase inhibition rates and IC 50 values were determined by a NovoStar ELISA (BMG Co., Germany).
  • the known starting materials of the present invention can be prepared by the known methods in the art, or can be purchased from ABCR GmbH & Co. KG, Acros Organnics, Aldrich Chemical Company, Accela ChemBio Inc., or Dari chemical Company, etc.
  • argon atmosphere or “nitrogen atmosphere” means that a reaction flask is equipped with an argon or nitrogen balloon (about 1 L).
  • “Hydrogen atmosphere” means that a reaction flask is equipped with a hydrogen balloon (about 1 L).
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature from 20° C. to 30° C.
  • the reaction process in the examples was monitored by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the developing solvent used in the reactions, the eluent system in column chromatography and the developing solvent system in thin layer chromatography for purification of the compounds included: A: dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, and D: dichloromethane/ethyl acetate/methanol system.
  • the ratio of the volume of the solvent was adjusted according to the polarity of the compounds, and a small quantity of alkaline reagent such as triethylamine or acidic reagent such as acetic acid can also be added for adjustment.
  • 6-Bromo-8-methylquinoline 1a (444 mg, 2.00 mmol, prepared according to the known method disclosed in “ Journal of Organic Chemistry, 2014, 79(11), 5379-5385”), bis(pinacolato)diboron (508 mg, 2.00 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 10 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated.
  • 6-Bromo-8-fluoroquinoline 2a (226 mg, 1.00 mmol), bis(pinacolato)diboron (305 mg, 1.20 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (146 mg, 0.20 mmol) and potassium acetate (294 mg, 3.00 mmol) were dissolved successively in 10 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 2b (220 mg), yield: 80.1%.
  • 6-Bromoquinoline 3a (1.0 g, 4.80 mmol, Accela ChemBio Inc.), bis(pinacolato)diboron (1.46 g, 5.76 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (0.7 g, 0.96 mmol) and potassium acetate (1.4 g, 14.40 mmol) were dissolved successively in 20 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 3b (1.2 g), yield: 98.4%.
  • 6-Bromo-8-chloroquinoline 4a 300 mg, 1.24 mmol
  • bis(pinacolato)diboron 378 mg, 1.49 mmol
  • [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium 181 mg, 0.25 mmol
  • potassium acetate 364 mg, 3.72 mmol
  • the reaction solution was heated to 80° C., and stirred for 12 hours.
  • the reaction was stopped, and the reaction solution was cooled to room temperature and filtrated.
  • the filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 4b (260 mg), yield: 72.6%.
  • reaction was stopped, and the reaction solution was cooled to room temperature, added with 20 mL of ethyl acetate, washed with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 4 (30 mg), yield: 20.5%.
  • 6-Bromo-5-(5-methylfuran-2-yl)-1,2,4-triazin-3-amine 5c 6-Bromo-1,2,4-triazin-3-amine 5a (1.0 g, 5.72 mmol, prepared according to the known method disclosed in “ Journal of the American Chemical Society, 2015, 137(26), 8388-8391”), 6 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a reaction flask. The reaction solution was added with 2-methylfuran 5b (567 ⁇ L, 6.29 mmol), and stirred at room temperature for 17 hours. The reaction was stopped, and the reaction solution was added dropwise with saturated sodium bicarbonate solution to adjust the pH>7.
  • reaction solution was added with 30 mL of a pre-prepared aqueous solution of potassium hydroxide (962 mg, 17.14 mmol) and potassium hexacyanoferrate (5.65 g, 17.14 mmol), and stirred at room temperature for 1 hour.
  • the reaction was stopped, and the reaction solution was extracted with ethyl acetate (150 mL ⁇ 3).
  • the organic phases were combined, added with silica gel, and concentrated under reduced pressure.
  • the residue was purified by CombiFlash rapid preparation instrument with elution system C to obtain the title product 5c (450 mg), yield: 30.9%.
  • reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 8 (15 mg), yield: 12.2%.
  • 2-Cyclopropylaniline 12a (1.0 g, 7.52 mmol, prepared according to the method disclosed in the patent application “WO201314997”) was added to 100 mL of acetonitrile, followed by addition of N-bromosuccinimide (1.4 g, 7.89 mmol) and ammonium acetate (58 mg, 0.075 mmol). The reaction solution was stirred for 2 hours before the reaction was stopped. The reaction solution was added with 60 mL of water, and extracted with ethyl acetate (100 mL ⁇ 3). The organic phases were combined, washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 12b (0.85 g), yield: 53.5%.
  • 6-Bromo-4-methylquinoline 14a (444 mg, 2 mmol, prepared according to the known method disclosed in “ Tetrahedron, 2003, 59(6), 813-819”), bis(pinacolato)diboron (762 mg, 3 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 14b (460 mg), yield: 85.5%.
  • reaction solution was cooled to room temperature, and concentrated under reduced pressure.
  • the residue was added with water, added dropwise with saturated sodium bicarbonate solution until the pH of the mixture was 10, and extracted with ethyl acetate three times.
  • the organic phases were combined, washed once with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated.
  • the filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 16c (3.5 g), yield: 27.73%.
  • reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 21 (40 mg), yield: 50.99%.
  • 6-Bromo-8-methoxyquinoline 25a (530 mg, 2.2 mmol, prepared according to the known method disclosed in “ Journal of the American Chemical Society, 2005, 127(1), 74-75”), bis(pinacolato)diboron (845 mg, 3.3 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (162 mg, 0.22 mmol) and potassium acetate (652 mg, 6.65 mmol) were dissolved successively in 10 mL of 1,4-dioxane under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 3 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated through celite. The filter cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 25b (410 mg), yield: 65%
  • reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 26 (45 mg), yield: 36.11%.
  • reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 27 (25 mg), yield: 15.43%.
  • 6-Bromo-3-methylquinoline 28a 250 mg, 1.13 mmol, prepared according to the method disclosed in the patent application “WO2006132739A2”), bis(pinacolato)diboron (429 mg, 1.69 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (165 mg, 0.225 mmol) and potassium acetate (331 mg, 3.38 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 28b (240 mg), yield: 79.2%.
  • 6-Bromoquinazoline 29a (418 mg, 2 mmol, prepared according to the known method disclosed in “ Science of Synthesis, 2004, 16, 573-749”), bis(pinacolato)diboron (609 mg, 7.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 29b (450 mg), yield: 87.9%.
  • 6-Bromo-3-methoxyquinoline 34a 120 mg, 0.5 mmol, prepared according to the method disclosed in the patent application “WO2012009194A1”), bis(pinacolato)diboron (192 mg, 0.76 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (74 mg, 0.1 mmol) and potassium acetate (148 mg, 1.5 mmol) were dissolved successively in 15 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 34b (90 mg), yield: 62.9%.
  • the starting compound 23a in Step 1 was replaced with 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoxaline 36a (prepared according to the known method disclosed in “ Organic Letters, 2009, 11(13), 2860-2863”), accordingly, the title product 36 (20 mg) was prepared.
  • the starting compound 23a in Step 1 was replaced with 2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 37a (prepared according to the known method disclosed in “ Journal of the American Chemical Society, 2015, 137(4), 1593-1600”), accordingly, the title product 37 (20 mg) was prepared.
  • Methyl 6-bromoquinoline-8-carboxylate 38a (400 mg, 1.5 mmol, prepared according to the method disclosed in the patent application “WO2011020193A1”) was dissolved in 15 mL of methanol. The reaction solution was added dropwise with 5 mL of 40% aqueous ammonia, and stirred overnight. The reaction solution was added with water, and extracted with ethyl acetate three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with elution system B to obtain the title product 38b (280 mg), yield: 74%.
  • 3-Bromo-6-chloroquinoline 39a (266 mg, 1.1 mmol, prepared according to the known method disclosed in “ Journal of Heterocyclic Chemistry, 2015, 52(4), 1019-1025”), morpholine (87 mg, 1 mmol), palladium acetate (12 mg, 0.05 mmol), ( ⁇ )-2,2′-bis-(diphenylphosphino)-1,1′-binaphthalene (31 mg, 0.05 mmol) and cesium carbonate (652 mg, 2 mmol) were dissolved in 10 mL of tetrahydrofuran under an argon atmosphere. The reaction solution was heated to 70° C., and stirred overnight.
  • Lithium aluminum hydride (150.76 mg, 3.97 mmol) was added to 50 mL of tetrahydrofuran.
  • the reaction solution was added with 6-bromoquinoline-4-carboxylic acid 40a (1.0 g, 3.97 mmol, prepared according to the known method disclosed in “ Chinese Chemical Letters, 2010, 21(1), 35-38”) in batches at 0° C., and stirred for 2 hours.
  • the reaction solution was added with 5 mL of water, and filtrated through celite. The filter cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 40b (250 mg), yield: 26.5%.
  • Example 39 the starting compound 39a in Step 1 was replaced with 6-bromo-4-chloroquinoline 43a (prepared according to the known method disclosed in “ Journal of Medicinal Chemistry, 2015, 58(14), 5522-5537”), accordingly, the title compound 43 (50 mg) was prepared.
  • 2-Amino-3-fluorobenzonitrile 45a (1 g, 7.35 mmol, purchased from Shanghai Bide Pharmatech Ltd.) was dissolved in 50 mL of dichloromethane. The reaction solution was added with N-bromosuccinimide (1.37 g, 7.71 mmol), and stirred for 16 hours. The reaction solution was added with water, and extracted with dichloromethane (50 mL ⁇ 3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 45b (1.25 g), yield: 79.13%.
  • 1-(2-Methylpyridin-4-yl)ethan-1-one 48a (4.29 g, 31.74 mmol, prepared according to the known method disclosed in “ Journal of Medicinal Chemistry, 2015, 58(12), 5028-5037”) was dissolved in 35 mL of dimethyl sulfoxide. The reaction solution was then added with 35 mL of hydrobromic acid, and stirred at 55° C. overnight. The reaction solution was used directly in the next step.
  • the crude compound 48d (8.5 g, 33.96 mmol) was dissolved in 80 mL of dioxane.
  • the reaction solution was added with 20 mL of aqueous ammonia, and stirred for 1 hour.
  • the reaction solution was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 48e (1.6 g), yield: 25.17%.
  • 2-Amino-5-bromobenzonitrile 53a (500 mg, 2.54 mmol, prepared according to the known method disclosed in “ European Journal of Medicinal Chemistry, 2014, 76, 341-343”) was dissolved in 10 mL of tetrahydrofuran. The reaction solution was cooled in an ice bath, added dropwise with 12.69 mL of 1.0 M ethyl magnesium bromide under an argon atmosphere, and stirred for 2 hours. The reaction solution was added with 6 M hydrochloric acid, and stirred for 2 hours. The reaction solution was added with saturated sodium carbonate solution, and extracted with ethyl acetate (50 mL ⁇ 3).
  • the resulting intermediate was added with 6 mL of sulfuric acid, and stirred at 100° C. for 4 hours.
  • the reaction solution was cooled to room temperature, added dropwise with saturated sodium bicarbonate solution to adjust the pH to greater than 10, and extracted with ethyl acetate (30 mL ⁇ 3).
  • the organic phases were combined, dried over anhydrous sodium sulfate, and filtrated.
  • the filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 54b (1.5 g), yield: 63.70%.
  • 6-Bromo-4-chloroquinazoline 55a (1.0 g, 4.10 mmol) was dissolved in 80 mL of methanol. The reaction solution was added with sodium methoxide (2.21 g, 41.05 mmol), and stirred for 3 hours. The reaction solution was concentrated to dryness, added with water, and filtrated. The filter cake was dried to obtain the product 55b (0.55 g), yield: 56.01%.
  • Test Example 1 Determination of the inhibition activity of the compounds of the present invention on the adenosine A 2a receptor (A 2a R) cAMP signaling pathway, the adenosine A 2b receptor (A 2b R) cAMP signaling pathway, the adenosine A 1 receptor (AiR) cAMP signaling pathway and the adenosine A 3 receptor (A3R) cAMP signaling pathway.
  • the inhibition activity of the compounds of the present invention on the adenosine A 2a receptor (A 2a R) cAMP signaling pathway, the adenosine A 2b receptor cAMP signaling pathway, the adenosine A 1 receptor cAMP signaling pathway and the adenosine A 3 receptor cAMP signaling pathway was determined by the following method. The experimental method is briefly described as follows:
  • CHO-K1/A 2a R cells NM_000675.5
  • CHO-K1/A 2b R cells NM_000676.2
  • CHO-K1/A 1 R cells NM_000674.2
  • CHO-K1/A 3 R cells NM_000677.3
  • Adenosine deaminase (sigma, 10102105001)
  • PHERAstar multi-function microplate reader (Cisbio, 62AM4PEB)
  • CHO-K1/A 2a R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 800 g/ml bleomycin. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 10 6 cells/ml.
  • each well was added with 5 ⁇ l of cell suspension, and 2.5 ⁇ l of test compound (4 ⁇ concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes.
  • test compound 4 ⁇ concentration
  • the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase
  • the final concentrations of the compounds were: 10000, 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256, 0.00512, and 0.001024 nM.
  • the final concentration of ethyl carbazole was 20 nM.
  • Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit.
  • cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 5 ⁇ l of diluted cAMP-d2, followed by addition of 5 ⁇ l of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour.
  • the HTRF signal values were read by the PHERAstar multi-function microplate reader.
  • IC 50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 1.
  • CHO-K1/A 2b R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 1 mg/ml G418. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 10 6 cells/ml.
  • each well was added with 5 ⁇ l of cell suspension, and 2.5 ⁇ l of test compound (4 ⁇ concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes.
  • ethyl carbazole 4 ⁇ concentration
  • the final concentrations of the compounds were: 100000, 10000, 1000, 100, 10, 1, 0.1 and 0 nM.
  • the final concentration of ethyl carbazole was 1 ⁇ M. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit.
  • cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 5 ⁇ l of diluted cAMP-d2, followed by addition of 5 ⁇ l of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour.
  • the HTRF signal values were read by the PHERAstar multi-function microplate reader.
  • IC 50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 2.
  • CHO-K1/A 1R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 1 mg/ml G418. The cells were digested with the cell separation buffer during the experiment. The cells were then resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 5 ⁇ 10 5 cells/ml.
  • each well was added with 12.5 ⁇ l of cell suspension, and 6.25 ⁇ l of test compound (4 ⁇ concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes.
  • cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 12.5 ⁇ l of diluted cAMP-d2, followed by addition of 12.5 ⁇ l of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour.
  • the HTRF signal values were read by the PHERAstar multi-function microplate reader.
  • IC 50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 3.
  • Adenosine A 3 Receptor CHO-K/A3R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 10 ⁇ g/ml puromycin. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 5 ⁇ 10 5 cells/ml.
  • each well was added with 12.5 ⁇ l of cell suspension, and 6.25 ⁇ l of test compound (4 ⁇ concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes.
  • each well was then added with 6.25 ⁇ l of forskolin and 2C1-IB-MECA (4 ⁇ concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes.
  • the final concentrations of the compounds were: 100000, 10000, 1000, 100, 10, 1, 0.1 and 0 nM.
  • the final concentrations of forskolin was 10 ⁇ M.
  • the final concentration of 2Cl-IB-MECA was 5 nM. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit.
  • cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 12.5 ⁇ l of diluted cAMP-d2, followed by addition of 12.5 ⁇ l of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour.
  • the HTRF signal values were read by the PHERAstar multi-function microplate reader.
  • IC 50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 3.
  • IC 50 values of the inhibition activity of the compounds of the present invention on the adenosine A 2a receptor (A 2a R) cAMP signaling pathway Example No. IC 50 /nM (A 2a R) 1 0.5 2 0.6 3 0.7 4 0.7 5 0.4 6 1.3 7 3.1 8 3.6 14 0.1 15 0.1 16 0.2 17 0.5 18 0.2 19 0.4 20 1.5 22 0.3 23 0.4 24 0.9 25 1.5 26 1.8 27 2.2 28 3.0 29 3.5 30 3.7 31 3.8 44 0.8 45 1.2 46 1.6 47 1.6 48 2.4 49 2.5 50 3.2 51 3.5 52 0.9 13 (Example 1 (lxxii) 158.3 of WO2011095625)
  • the compounds of the present invention have a significant inhibition activity on the adenosine A 2a receptor.
  • the introduction of a nitrogen atom into the fused aryl moiety of the core structrue results in an unexpected inhibition activity of the compounds of the present invention on the adenosine A 2a receptor.
  • the structural difference between the compound of Comparative Example 1 and the compound of Example 3 is merely that the compound of Example 3 has the introduction of a nitrogen atom into the 5-position of the naphthyl of the compound of Comparative Example 1, but the inhibition activity on the adenosine A 2a receptor differs by 225 times.
  • IC 50 values of the inhibition activity of the compounds of the present invention on the adenosine A 2b receptor (A 2b R) cAMP signaling pathway Example No. IC 50 /nM (A 2b R) 3 47 4 4 5 46 14 3 15 7 16 18 17 22 19 25 22 7 23 17 25 4 45 25 46 18 52 14
  • the compounds of the present invention have a good inhibition activity on the adenosine A2 b receptor.
  • the compounds of the present invention have a weak inhibition activity on the adenosine A 1 receptor and the adenosine A 3 receptor, indicating that the compounds of the present invention are selective for the adenosine A 2a receptor and the adenosine A 2b receptor, particularly for the adenosine A 2a receptor.
  • Test Example 2 Pharmacokinetics assay of the compounds of the present invention in mice
  • mice were used as test animals.
  • the drug concentration in plasma at different time points was determined by LC/MS/MS method after intragastrical administration of the compounds of Examples 2, 3, 17, 18, 19, 20, 31 and 44 to mice.
  • the pharmacokinetic behavior of the compounds of the present invention was studied and evaluated in mice.
  • mice Seventy-two C57 mice (female) were purchased from Shanghai Jiesijie Laboratory Animal Co., LTD, with Certificate No.: SCXK (Shanghai) 2013-0006, and equally divided into 8 groups (9 mice per group).
  • test compound was weighed, and added with 5% by volume of DMSO, 5% by volume of tween 80 and 90% by volume of normal saline to prepare a 0.1 mg/mL colorless, clear and transparent solution.
  • mice were administered intragastrically the test compounds at an administration dosage of 2.0 mg/kg and an administration volume of 0.2 mL/10 g.
  • mice were intragastrically administered the compounds of Examples 2, 3, 17, 18, 19, 20, 31 and 44.
  • 0.1 ml of blood was taken before administration and at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0 and 24.0 hours after administration.
  • the samples were stored in heparinized tubes, and centrifuged for 10 minutes at 3,500 rpm to separate the blood plasma.
  • the plasma samples were stored at ⁇ 20° C.
  • the content of the test compounds in the plasma of mice after intragastrical administration of the test compounds at different concentrations was determined: 25 ⁇ L of mouse plasma at each time after administration were taken, added with 50 ⁇ L of the internal standard solution of camptothecin (100 ng/mL) and 200 ⁇ L of acetonitrile, vortex-mixed for 5 minutes, and centrifuged for 10 minutes (4000 rpm). 5 ⁇ L of the supernatant were taken from the plasma samples for LC/MS/MS analysis.
  • the compounds of the present invention are well absorbed, and have a pharmacokinetic advantage.

Landscapes

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

Abstract

A 1,2,4-triazine-3-amine derivative, a preparation therefor, and use thereof in medicine are provided. Specifically, a 1,2,4-triazine-3-amine derivative as represented by general formula (I), a preparation method therefor, a pharmaceutical composition containing the derivative, and use thereof as a therapeutic agent, in particular as an A2a or A2b receptor antagonist, and use thereof in the preparation of a medicament for treating a condition or disorder that is ameliorated by means of inhibition of the A2a or A2b receptor are provided. Each substituent in general formula (I) is defined in the description.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a continuation of U.S. patent application Ser. No. 16/477,011 filed Jul. 10, 2019, which is a Section 371 of International Application No. PCT/CN2018/072308 filed Jan. 12, 2018, which was published in the Chinese language on Jul. 19, 2018, under International Publication No. WO 2018/130184 A1, which claims priority under 35 U.S.C. § 119(b) to Chinese Application No. 201710023970.7, filed Jan. 13, 2017 and Chinese Application No. 201710874488.4, filed Sep. 25, 2017, the disclosures of which are incorporated herein by reference in its/their entirety.
  • FIELD OF THE INVENTION
  • The present invention belongs to the field of medicine, and relates to a 1,2,4-triazin-3-amine derivative of formula (I), a method for preparing the same, a pharmaceutical composition comprising the same, a use thereof as a therapeutic agent, in particular as an A2a receptor antagonist, and a use thereof in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A2a receptor.
  • BACKGROUND OF THE INVENTION
  • Adenosine is a naturally occurring purine nucleoside, and is an endogenous regulator of many physiological functions. It plays an important role in the regulation of the cardiovascular system, central nervous system, respiratory system, kidney, fat and platelets.
  • The action of adenosine is mediated by a family of G-protein coupled receptors. It is known currently that there are at least four subtypes of adenosine receptors, which are classified into A1, A2a, A2b and A3. Among them, the A1 and A3 receptors inhibit the activity of the enzyme adenylate cyclase, whereas the A2a and A2b receptors stimulate the activity of the same enzyme, thereby modulating the level of cyclic AMP in cells. Adenosine regulates a wide range of physiological functions through these receptors.
  • The A2a receptor (A2aR) is widely distributed in the body, and is mainly expressed in the striatum in the central nervous system, and is also expressed in tissues such as the periphery, heart, liver, lung and kidney. Several preclinical studies show that adenosine A2a receptor antagonists have surprising efficacy in the treatment of neurodegenerative diseases, primarily Parkinson's disease, Huntington's disease or Alzheimer's disease (Trends in Neurosci. 2006, 29(11), 647-654; Expert Opinion on Therapeutic Patents, 2007, 17, 979-991 and the like). Moreover, adenosine A2a receptor antagonists can also be used to treat other central nervous system (CNS) related diseases such as depression, restless syndrome, sleep disorders and anxiety disorders (Clin. Neuropharmacol. 2010, 33, 55-60; J Neurosci. 2010, 30 (48), 16284-16292; Parkinsonisn Relat. Disord. 2010, 16 (6), 423-426; and references therein: Mov. Disorders, 2010, 25(2), S305). In addition, adenosine A2a receptor antagonists also have therapeutic potential as neuroprotective agents (see Jenner P. J Neurol. 2000; 24 7Supp12: 1143-50).
  • Recent studies indicate that the activation of the adenosine A2a receptor can exert an important immunomodulatory effect in many pathological processes such as ischemia, hypoxia, inflammation, trauma, transplantation and the like, which may be related to the higher expression level of the A2a receptor in various immune cells such as T cells, B cells, monocyte macrophages, neutrophils and the like. Moreover, the activation of the A2a receptor can promote the body to generate immune tolerance, and closely participate in the formation of “immune escape” or “immunosuppression” of tumor cells, thereby creating a favorable condition for the occurrence and development of tumors. Lokshin and his colleagues (Cancer Res. 2006, Aug. 1; 66 (15):7758-65) demonstrate that the activation of A2aR in natural killer cells can inhibit the killing of tumor cells by natural killer cells through increasing cAMP and activating PKA. Studies also show that the activation of A2a receptor can promote the proliferation of tumor cells such as melanoma A375 cells, fibroblast NIH3T3 cells, pheochromocytoma PC12 cells and the like, which may be related to the fact that the activation of the A2a receptor in T cells can inhibit T cell activation, proliferation, adhesion to tumor cells, and produce cytotoxic effect on tumor cells. However, in the A2a receptor knockout mice, the anti-tumor immunity of CD8+T cells is enhanced, and the tumor proliferation is significantly inhibited. Therefore, A2a receptor antagonists can be used in the treatment of tumor. Deepak Mittal et al. find that the combination administration of A2b receptor inhibitors with chemotherapeutic drugs or immunological checkpoint inhibitors can significantly reduce tumor metastasis in a mice triple negative breast cancer model; the knockout of the A2b receptor in mice or human colon cancer cell line significantly reduces colon cancer metastasis and cell tumorigenicity; meanwhile, the study finds that the A2b receptor is highly expressed in human triple negative breast cancer cell line, and the expression level of the A2b receptor is closely related to tumor progression. These results show that inhibition of the A2b receptor can inhibit tumor metastasis, and the A2b receptor is thus expected to be an ideal target for the treatment of tumors (Cancer Res. 2016 Aug. 1; 76(15):4372-82). The development of dual inhibitors of the A2a receptor and the A2b receptor has also become a direction worth exploring.
  • Although compounds having significant biological activity on a variety of subtypes of adenosine receptors can have a therapeutic effect, they can cause undesired side effects. For example, during tissue ischemia/hypoxia, when cells of central system, circulatory system, digestive system, and skeletal muscle are in an anoxic and hypoxic stress environment, extracellular aggregated adenosine initiates a corresponding protective mechanism by activating the adenosine A1 receptor on the cell membrane, thereby increasing the tolerance of the cells to anoxia and hypoxia. The A1 receptor located on immune cells can promote cellular immune responses in a hypoxic environment. Moreover, the A1 receptor can also reduce free fatty acids and triglycerides, and is involved in regulating blood glucose. Therefore, the continued blocking of the A1 receptor can cause various adverse effects in the body tissues (Chinese Pharmacological Bulletin, 2008, 24(5), 573-576). For example, it is reported that the blocking of the A1 receptor will cause adverse effects such as anxiety, awakening and the like in animal models (Basic & Clinical Pharmacology & Toxicology, 2011, 109 (3), 203-7). The Adenosine Released by the adenosine A3 receptor during myocardial ischemia exerts a strong protective effect in heart (as described by Gessi S et al, Pharmacol. Ther. 117 (1), 2008, 123-140). The continued blocking of the A3 receptor can increase the likelihood of complications caused by any pre-existing or developing ischemic heart disease such as angina or heart failure.
  • Currently, many compounds have been developed as A2a receptor antagonists for the treatment of various diseases, as described in WO2007116106, WO2009080197, WO2011159302, WO2011095625, WO2014101373 and WO2015031221. However, there still exist problems such as low solubility, photosensitivity, low activity, low selectivity and low bioavailability.
  • WO2011095625 discloses a 1,2,4-triazin-4-amine derivative of formula (A1) and use thereof in the treatment of a disease or condition ameliorated by the inhibition of the A1 receptor or the A2a receptor.
  • Figure US20210230138A1-20210729-C00002
  • This patent application discloses a total of more than 200 examples, among which there are only five examples in which the ring A is a fused aromatic ring. The data in this patent application show that when ring A is a fused aromatic ring, the inhibition activity on the A2aR is weak (see Table 1).
  • TABLE 1
    Examples in the patent application WO2011095625
    pKi/A2aR
    (determined in this patent
    Example No. Structure application)
    1(lxxii) i.e., Example 13 of the present invention
    Figure US20210230138A1-20210729-C00003
    6.99
    1(xviii)
    Figure US20210230138A1-20210729-C00004
    6.75
    1(xxii)
    Figure US20210230138A1-20210729-C00005
    No data provided
    1(xxxv)
    Figure US20210230138A1-20210729-C00006
    7.19
    1(cxlii)
    Figure US20210230138A1-20210729-C00007
    No data provided
  • Among them, ring A of Example 1(lxxii) is naphthyl. We have now found that a derivative in which a nitrogen atom is introduced at the 5-position of the naphthyl shows a surprising activity, and the inhibition activity on the A2aR is 30 to 1500 times or more than that of Example 1 (lxxii). Such a strong inhibition activity cannot be expected when reading WO2011095625.
  • Therefore, the present invention provides a novel structure of an adenosine A2a receptor antagonist with a strong inhibition activity, and the compounds having such a structure also have a good inhibition effect on the adenosine A2b receptor and a weak inhibition effect on the adenosine A1 receptor and the adenosine A3 receptor, exhibiting a good selectivity for the adenosine A2a receptor. Meanwhile, the compounds having such a structure exhibit an excellent anti-tumor effect and pharmacokinetics activity.
  • SUMMARY OF THE INVENTION
  • The object of the present invention is to provide a compound of formula (I):
  • Figure US20210230138A1-20210729-C00008
  • or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • wherein:
  • ring A is aryl or heteroaryl;
  • G1, G2, G3 and G4 are identical or different and each are independently selected from the group consisting of C, CH and N;
  • R1 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR5, —C(O)R5, —S(O)mR5, NH2S(O)mR5, —NR6R7, S(O)mNR6R7 and —C(O)NR6R7, wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • each R2 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR5, —C(O)R5, —S(O)mR5, NH2S(O)mR5, —NR6R7, S(O)mNR6R7 and —C(O)NR6R7, wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • each R3 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, deuterated alkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR5, —C(O)R5, —S(O)mR5, NH2S(O)mR, —NR6R7, S(O)mNR6R7 and —C(O)NR6R7, wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • each R4 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, —OR5, —C(O)R5, —S(O)mR5, NH2S(O)mR5, —NR6R7, S(O)mNR6R7 and —C(O)NR6R7, wherein the alkyl, alkoxy, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R5 is selected from the group consisting of hydrogen, alkyl, haloalkyl, amino, hydroxy, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R6 and R7 are each independently selected from the group consisting of hydrogen, alkyl, haloalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • or, R6 and R7 together with the nitrogen atom to which they are attached form a heterocyclyl, wherein the heterocyclyl contains one or two identical or different heteroatoms selected from the group consisting of N, O and S, and the heterocyclyl is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • m is 0, 1 or 2;
  • r is 0, 1, 2 or 3;
  • q is 0, 1 or 2; and
  • n is 0, 1, 2, 3, 4 or 5.
  • In a preferred embodiment of the present invention, the compound of formula (I) is a compound of formula (Iaa):
  • Figure US20210230138A1-20210729-C00009
  • or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • wherein
  • ring A, G1, G2, R1, R3, R4, r and n are as defined in formula (I).
  • In a preferred embodiment of the present invention, the compound of formula (I) is a compound of formula (Ibb):
  • Figure US20210230138A1-20210729-C00010
  • or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • wherein
  • G1 and G2 are identical or different and are each independently CRa or N;
  • Ra is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, deuterated alkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Rc is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • ring A, R1, R4 and n are as defined in formula (I).
  • In a preferred embodiment of the present invention, the compound of formula (I) is a compound of formula (II):
  • Figure US20210230138A1-20210729-C00011
  • or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • wherein ring A, R1, R3, R4, r and n are as defined in formula (I).
  • In a preferred embodiment of the present invention, in the compound of formula (I), ring A is selected from the group consisting of phenyl, pyridyl, thienyl and furanyl.
  • In a preferred embodiment of the present invention, the compound of formula (I) is a compound of formula (III):
  • Figure US20210230138A1-20210729-C00012
  • or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • wherein R1, R3, R4, r and n are as defined in formula (I).
  • In a preferred embodiment of the present invention, in the compound of formula (I), R1 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, cyano, cycloalkyl, haloalkyl, heterocyclyl and —C(O)NR6R; R6 and R7 are as defined in formula (I); the halogen is preferably fluorine, chlorine or bromine, the alkyl is preferably methyl, ethyl, isopropyl or n-butyl, the alkoxy is preferably methoxy or ethoxy, and the cycloalkyl is preferably cyclopropyl, cyclopentyl or cyclohexyl.
  • In a preferred embodiment of the present invention, in the compound of formula (I), each R3 is identical or different and each is independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, deuterated alkyl, alkoxy, cyano, cycloalkyl and heterocyclyl, wherein the alkyl and alkoxy are each independently optionally substituted by one or more substituents selected from the group consisting of halogen, deuterium, hydroxy, cyano, amino, nitro, cycloalkyl and heterocyclyl; the halogen is preferably fluorine, chlorine or bromine, the alkyl is preferably methyl, ethyl, isopropyl or n-butyl, the alkoxy is preferably methoxy or ethoxy, and the heterocyclyl is preferably piperidinyl, piperazinyl, morpholinyl or tetrahydropyranyl.
  • In a preferred embodiment of the present invention, in the compound of formula (I), each R4 is identical or different and each is independently selected from the group consisting of hydrogen, alkyl and halogen.
  • Typical compounds of the present invention include, but are not limited to:
  • Example
    No. Structure and name of the compound
     1
    Figure US20210230138A1-20210729-C00013
    6-(8-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 1
     2
    Figure US20210230138A1-20210729-C00014
    6-(8-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 2
     3
    Figure US20210230138A1-20210729-C00015
    5-Phenyl-6-(quinolin-6-yl)-1,2,4-triazin-3-amine 3
     4
    Figure US20210230138A1-20210729-C00016
    6-(8-Chloroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 4
     5
    Figure US20210230138A1-20210729-C00017
    5-(5-Methylfuran-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine
    5
     6
    Figure US20210230138A1-20210729-C00018
    5-(Furan-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine 6
     7
    Figure US20210230138A1-20210729-C00019
    6-(8-Methylquinolin-6-yl)-5-(thiophen-2-yl)-1,2,4-triazin-3-amine 7
     8
    Figure US20210230138A1-20210729-C00020
    5-(4-Fluorophenyl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine 8
     9
    Figure US20210230138A1-20210729-C00021
    5-Phenyl-6-(8-(trifluoromethyl)quinolin-6-yl)-1,2,4-triazin-3-amine 9
    10
    Figure US20210230138A1-20210729-C00022
    6-(8-Isopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 10
    11
    Figure US20210230138A1-20210729-C00023
    6-(8-Ethylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 11
    12
    Figure US20210230138A1-20210729-C00024
    6-(8-Cyclopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 12
    13 (Comparative example 1)
    Figure US20210230138A1-20210729-C00025
    6-(Naphthalen-2-yl)-5-phenyl-1,2,4-triazin-3-amine 13
    14
    Figure US20210230138A1-20210729-C00026
    6-(4-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 14
    15
    Figure US20210230138A1-20210729-C00027
    6-(4-Methylquinazolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 15
    16
    Figure US20210230138A1-20210729-C00028
    6-(8-Fluoro-4-methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 16
    17
    Figure US20210230138A1-20210729-C00029
    5-(2-Fluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 17
    18
    Figure US20210230138A1-20210729-C00030
    5-(4-Fluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 18
    19
    Figure US20210230138A1-20210729-C00031
    5-(4-Fluorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine
    19
    20
    Figure US20210230138A1-20210729-C00032
    6-(8-Fluoro-4-methylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-
    amine 20
    21
    Figure US20210230138A1-20210729-C00033
    5-(2,4-Difluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine
    21
    22
    Figure US20210230138A1-20210729-C00034
    5-(4-Fluorophenyl)-6-[4-(trideuteromethyl)-6-quinolinyl]-1,2,4-triazin-
    3-amine 22
    23
    Figure US20210230138A1-20210729-C00035
    6-(4-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 23
    24
    Figure US20210230138A1-20210729-C00036
    6-(3-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 24
    25
    Figure US20210230138A1-20210729-C00037
    6-(8-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 25
    26
    Figure US20210230138A1-20210729-C00038
    5-(3-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 26
    27
    Figure US20210230138A1-20210729-C00039
    6-(4-Chloroquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 27
    28
    Figure US20210230138A1-20210729-C00040
    6-(3-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 28
    29
    Figure US20210230138A1-20210729-C00041
    5-Phenyl-6-(quinazolin-6-yl)-1,2,4-triazin-3-amine 29
    30
    Figure US20210230138A1-20210729-C00042
    5-(4-Fluorophenyl)-6-(quinolin-6-yl)-1,2,4-triazin-3-amine 30
    31
    Figure US20210230138A1-20210729-C00043
    6-(8-Fluoro-4-methylquinolin-6-yl)-5-(2-fluorophenyl)-1,2,4-triazin-3-
    amine 31
    32
    Figure US20210230138A1-20210729-C00044
    5-(4-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 32
    33
    Figure US20210230138A1-20210729-C00045
    5-(2,4-Difluorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine
    33
    34
    Figure US20210230138A1-20210729-C00046
    6-(3-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 34
    35
    Figure US20210230138A1-20210729-C00047
    5-(2-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 35
    36
    Figure US20210230138A1-20210729-C00048
    5-Phenyl-6-(quinoxalin-6-yl)-1,2,4-triazin-3-amine 36
    37
    Figure US20210230138A1-20210729-C00049
    6-(2-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 37
    38
    Figure US20210230138A1-20210729-C00050
    6-(3-Amino-5-phenyl-1,2,4-triazin-6-yl)quinoline-8-carbonitrile 38
    39
    Figure US20210230138A1-20210729-C00051
    6-(3-Morpholinoquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 39
    40
    Figure US20210230138A1-20210729-C00052
    6-(4-(Morpholinomethyl)quinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine
    40
    41
    Figure US20210230138A1-20210729-C00053
    6-(3-(Morpholinomethyl)quinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine
    41
    42
    Figure US20210230138A1-20210729-C00054
    6-(3-Amino-5-phenyl-1,2,4-triazin-6-yl)quinoline-8-carboxamide 42
    43
    Figure US20210230138A1-20210729-C00055
    6-(4-Morpholinoquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 43
    44
    Figure US20210230138A1-20210729-C00056
    6-(4-Ethylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 44
    45
    Figure US20210230138A1-20210729-C00057
    6-(8-Fluoro-4-methylquinazolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 45
    46
    Figure US20210230138A1-20210729-C00058
    5-(4-Chlorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine
    46
    47
    Figure US20210230138A1-20210729-C00059
    6-(4-Ethyl-8-fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 47
    48
    Figure US20210230138A1-20210729-C00060
    5-(2-Methylpyridin-4-yl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine
    48
    49
    Figure US20210230138A1-20210729-C00061
    6-(4-Cyclopropylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine
    49
    50
    Figure US20210230138A1-20210729-C00062
    6-(8-Fluoro-4-methylquinazolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-
    3-amine 50
    51
    Figure US20210230138A1-20210729-C00063
    6-(4-(Difluoromethyl)quinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-
    amine 51
    52
    Figure US20210230138A1-20210729-C00064
    5-(4-Fluorophenyl)-6-(4-(methyl-d3)quinazolin-6-yl)-1,2,4-triazin-3-
    amine 52
    53
    Figure US20210230138A1-20210729-C00065
    6-(4-Ethylquinazolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 53
    54
    Figure US20210230138A1-20210729-C00066
    5-(4-Fluorophenyl)-6-(4-(trifluoromethyl)quinolin-6-yl)-1,2,4-triazin-3-
    amine 54
    55
    Figure US20210230138A1-20210729-C00067
    5-(4-Fluorophenyl)-6-(4-methoxyquinazolin-6-yl)-1,2,4-triazin-3-amine
    55

    or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • In another aspect, the present invention relates to a method for preparing the compound of formula (I), comprising a step of:
  • Figure US20210230138A1-20210729-C00068
  • subjecting a compound of formula (I-A) and a compound of formula (I-B) to a coupling reaction to obtain the compound of formula (I),
  • wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00069
  • ring A, G1-G4, R1-R4, r, q and n are as defined in formula (I).
  • In another aspect, the present invention relates to a method for preparing the compound of formula (Iaa), comprising a step of:
  • Figure US20210230138A1-20210729-C00070
  • subjecting a compound of formula (Iaa-1) and a compound of formula (I-B) to a coupling reaction to obtain the compound of formula (Iaa),
  • wherein:
  • X is halogen;
  • M is or;
  • Figure US20210230138A1-20210729-C00071
  • ring A, G1, G2, R1, R3, R4, r and n are as defined in formula (I).
  • In another aspect, the present invention relates to a method for preparing the compound of formula (II), comprising a step of:
  • Figure US20210230138A1-20210729-C00072
  • subjecting a compound of formula (II-A) and a compound of formula (I-B) to a coupling reaction to obtain the compound of formula (II),
  • wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00073
  • ring A, R1, R3, R4, r and n are as defined in formula (I).
  • In another aspect, the present invention relates to a method for preparing the compound of formula (III), comprising a step of:
  • Figure US20210230138A1-20210729-C00074
  • subjecting a compound of formula (II-A) and a compound of formula (III-B) to a coupling reaction to obtain the compound of formula (III), wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00075
  • R1, R3, R4, r and n are as defined in formula (I).
  • In another aspect, the present invention relates to a pharmaceutical composition comprising a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A2a receptor.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating a disease or condition ameliorated by the inhibition of the A2b receptor.
  • In the context of the present invention, the disease or condition ameliorated by the inhibition of the A2a receptor or the A2b receptor is selected from the group consisting of cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior; preferably cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer and the like), multiple myeloma, malignant lymphoma (reticular sarcoma, lymphosarcoma, Hodgkin's lymphoma and the like), polycythemia vera, leukemia (acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia and the like), thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma, pediatric tumor (Ewing's familial sarcoma, Wilms' sarcoma, rhabdomyosarcoma, angiosarcoma, embryonic testicular cancer, neuroblastoma, retinoblastoma, hepatoblastoma, nephroblastoma and the like) and the like; and more preferably lung cancer.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain damage, neuroinflammation and addictive behavior, and preferably cancer.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer and the like), multiple myeloma, malignant lymphoma (reticular sarcoma, lymphosarcoma, Hodgkin's lymphoma and the like), polycythemia vera, leukemia (acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia and the like), thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma, pediatric tumor (Ewing's familial sarcoma, Wilms' sarcoma, rhabdomyosarcoma, angiosarcoma, embryonic testicular cancer, neuroblastoma, retinoblastoma, hepatoblastoma, nephroblastoma and the like) and the like.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for treating lung cancer, and preferably non-small cell lung cancer.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for inhibiting the A2a receptor.
  • The present invention further relates to a use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same in the preparation of a medicament for inhibiting the A2b receptor.
  • The present invention also relates to a method for inhibiting the A2a receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention also relates to a method for inhibiting the A2b receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention also relates to a method for treating a disease or condition ameliorated by the inhibition of the A2a receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention also relates to a method for treating a disease or condition ameliorated by the inhibition of the A2b receptor, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, a diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention relates to a method for treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior, and preferably cancer, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention further relates to a method for treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer and the like), multiple myeloma, malignant lymphoma (reticular sarcoma, lymphosarcoma, Hodgkin's lymphoma and the like), polycythemia vera, leukemia (acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia and the like), thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma, pediatric tumor (Ewing's familial sarcoma, Wilms' sarcoma, rhabdomyosarcoma, angiosarcoma, embryonic testicular cancer, neuroblastoma, retinoblastoma, hepatoblastoma, nephroblastoma and the like) and the like, comprising a step of administrating to a patient in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same.
  • The present invention further relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as a medicament.
  • The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as an A2a receptor antagonist.
  • The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use as an A2b receptor antagonist.
  • The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating a disease or condition ameliorated by the inhibition of the A2a receptor.
  • The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating a disease or condition ameliorated by the inhibition of the A2b receptor.
  • The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating cancer, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis and the like), attention-related disorder, extrapyramidal syndrome, abnormal movement disorder, cirrhosis, liver fibrosis, fatty liver, dermal fibrosis, sleep disorder, stroke, brain injury, neuroinflammation and addictive behavior, and preferably cancer.
  • The present invention further relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same, for use in treating cancer selected from the group consisting of melanoma, brain tumor (glioma with malignant astroglia and oligodendroglioma and the like), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer, rectal cancer and the like), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous carcinoma and the like), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer and the like), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer and the like), multiple myeloma, malignant lymphoma (reticular sarcoma, lymphosarcoma, Hodgkin's lymphoma and the like), polycythemia vera, leukemia (acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia and the like), thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma, pediatric tumor (Ewing's familial sarcoma, Wilms' sarcoma, rhabdomyosarcoma, angiosarcoma, embryonic testicular cancer, neuroblastoma, retinoblastoma, hepatoblastoma, nephroblastoma and the like) and the like.
  • Pharmaceutical compositions containing the active ingredient can be in a form suitable for oral administration, for example, a tablet, troche, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, syrup or elixir.
  • Oral compositions can be prepared according to any known method in the art for the preparation of pharmaceutical composition. Such composition can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, colorants and preservatives, in order to provide a pleasing and palatable pharmaceutical preparation. Tablets contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • An aqueous suspension contains the active ingredient in admixture with excipients suitable for the manufacture of an aqueous suspension. The aqueous suspension can also contain one or more preservatives such as ethyl paraben or n-propyl paraben, one or more colorants, one or more flavoring agents, and one or more sweeteners.
  • An oil suspension can be formulated by suspending the active ingredient in a vegetable oil. The oil suspension can contain a thickener. The aforementioned sweeteners and flavoring agents can be added to provide a palatable formulation.
  • The active ingredient in admixture with the dispersants or wetting agents, suspending agent or one or more preservatives can be prepared as a dispersible powder or granule suitable for the preparation of an aqueous suspension by adding water. Suitable dispersants or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, such as sweeteners, flavoring agents and colorants, can also be added. These compositions can be preserved by adding an antioxidant, such as ascorbic acid.
  • The pharmaceutical composition of the present invention can also be in the form of an oil-in-water emulsion.
  • The pharmaceutical composition can be in the form of a sterile injectable aqueous solution. Acceptable vehicles or solvents that can be used are water, Ringer's solution or isotonic sodium chloride solution. The sterile injectable formulation can be a sterile injectable oil-in-water micro-emulsion in which the active ingredient is dissolved in the oil phase. For example, the active ingredient is dissolved in a mixture of soybean oil and lecithin. The oil solution is then added into a mixture of water and glycerol, and processed to form a micro-emulsion. The injectable solution or micro-emulsion can be introduced into a patient's bloodstream by local bolus injection. Alternatively, the solution and micro-emulsion are preferably administrated in a manner that maintains a constant circulating concentration of the compound of the present invention. In order to maintain this constant concentration, a continuous intravenous delivery device can be used. An example of such a device is Deltec CADD-PLUS™ 5400 intravenous injection pump.
  • The pharmaceutical composition can be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration. Such a suspension can be formulated with suitable dispersants or wetting agents and suspending agents as described above according to known techniques. The sterile injectable formulation can also be a sterile injectable solution or suspension prepared in a nontoxic parenterally acceptable diluent or solvent. Moreover, sterile fixed oils can easily be used as a solvent or suspending medium.
  • The compound of the present invention can be administrated in the form of a suppository for rectal administration. These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures, but liquid in the rectum, thereby melting in the rectum to release the drug. Such materials include cocoa butter, glycerin gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols with various molecular weights and fatty acid esters of polyethylene glycols.
  • It is well known to those skilled in the art that the dosage of a drug depends on a variety of factors including but not limited to, the following factors: activity of a specific compound, age of the patient, weight of the patient, general health of the patient, behavior of the patient, diet of the patient, administration time, administration route, excretion rate, drug combination and the like. In addition, the optimal treatment, such as treatment mode, daily dose of the compound of formula (I) or the type of pharmaceutically acceptable salt thereof can be verified by traditional therapeutic regimens.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Unless otherwise stated, the terms used in the specification and claims have the meanings described below.
  • The term “alkyl” refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group comprising 1 to 20 carbon atoms, preferably an alkyl having 1 to 12 carbon atoms, and more preferably an alkyl having 1 to 6 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,2-dimethylpentyl, 3,3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2,3-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 2,2-dimethylhexyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2,2-diethylpentyl, n-decyl, 3,3-diethylhexyl, 2,2-diethylhexyl, and various branched isomers thereof. More preferably, an alkyl group is a lower alkyl having 1 to 6 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, and the like. The alkyl group can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point. The substituent group(s) is preferably one or more groups independently optionally selected from the group consisting of H atom, D atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “alkoxy” refers to an —O-(alkyl) or an —O-(unsubstituted cycloalkyl) group, wherein the alkyl is as defined above. Non-limiting examples of alkoxy include methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy. The alkoxy can be optionally substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more group(s) independently selected from the group consisting of H atom, D atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “cycloalkyl” refers to a saturated or partially unsaturated monocyclic or polycyclic hydrocarbon substituent group having 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 10 carbon atoms, and most preferably 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl and the like. Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring. The cycloalkyl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point. The substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “heterocyclyl” refers to a 3 to 20 membered saturated or partially unsaturated monocyclic or polycyclic hydrocarbon group, wherein one or more ring atoms are heteroatoms selected from the group consisting of N, O and S(O)m (wherein m is an integer of 0 to 2), but excluding —O—O—, —O—S— or —S—S— in the ring, with the remaining ring atoms being carbon atoms. Preferably, the heterocyclyl has 3 to 12 ring atoms wherein 1 to 4 atoms are heteroatoms, more preferably, 3 to 10 ring atoms wherein 1 to 4 atoms are heteroatoms, and more preferably 5 to 6 ring atoms wherein 1 to 3 atoms are heteroatoms. Non-limiting examples of monocyclic heterocyclyl include pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like. Polycyclic heterocyclyl includes a heterocyclyl having a spiro ring, fused ring or bridged ring.
  • The ring of heterocyclyl can be fused to the ring of aryl, heteroaryl or cycloalkyl, wherein the ring bound to the parent structure is heterocyclyl. Non-limiting examples thereof include:
  • Figure US20210230138A1-20210729-C00076
  • The heterocyclyl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point. The substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “aryl” refers to a 6 to 14 membered all-carbon monocyclic ring or polycyclic fused ring (i.e., each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) having a conjugated 7-electron system, preferably 6 to 10 membered aryl, for example, phenyl and naphthyl. The ring of aryl can be fused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to the parent structure is aryl ring. Non-limiting examples thereof include:
  • Figure US20210230138A1-20210729-C00077
  • The aryl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point. The substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “heteroaryl” refers to a 5 to 14 membered heteroaromatic system having 1 to 4 heteroatoms selected from the group consisting of O, S and N. The heteroaryl is preferably 5 to 10 membered heteroaryl, more preferably 5 or 6 membered heteroaryl, for example, furanyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, tetrazolyl and the like. The ring of heteroaryl can be fused to the ring of aryl, heterocyclyl or cycloalkyl, wherein the ring bound to the parent structure is heteroaryl ring. Non-limiting examples thereof include:
  • Figure US20210230138A1-20210729-C00078
  • The heteroaryl can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point. The substituent group(s) is preferably one or more group(s) independently optionally selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • The term “haloalkyl” refers to an alkyl group substituted by one or more halogens, wherein the alkyl is as defined above.
  • The term “deuterated alkyl” refers to an alkyl group substituted by one or more deuterium atoms, wherein the alkyl is as defined above.
  • The term “hydroxy” refers to an —OH group.
  • The term “hydroxyalkyl” refers to an alkyl group substituted by hydroxy(s), wherein the alkyl is as defined above.
  • The term “halogen” refers to fluorine, chlorine, bromine or iodine.
  • The term “hydroxy” refers to an —OH group.
  • The term “amino” refers to a —NH2 group.
  • The term “cyano” refers to a —CN group.
  • The term “nitro” refers to a —NO2 group.
  • The term “carbonyl” refers to a C═O group.
  • The term “carboxy” refers to a —C(O)OH group.
  • The term “carboxylate group” refers to a —C(O)O(alkyl) group or a —C(O)O(cycloalkyl) group, wherein the alkyl and cycloalkyl are as defined above.
  • The term “acyl halide” refers to a compound containing a —C(O)-halogen group.
  • The present invention also comprises the compounds of formula (I) in various deuterated forms. Each of the available hydrogen atoms attached to the carbon atom can be independently replaced by a deuterium atom. Those skilled in the art can synthesize a compound of formula (I) in a deuterated form with reference to the relevant literature. Commercially available deuterated starting materials can be employed in the preparation of the compound of formula (I) in deuterated form, or they can be synthesized by conventional techniques with deuterated reagents including, but not limited to, deuterated borane, trideuterated borane in tetrahydrofuran, deuterated lithium aluminum hydride, deuterated iodoethane, deuterated iodomethane and the like.
  • “Optional” or “optionally” means that the event or circumstance described subsequently can, but need not, occur, and such a description includes the situation in which the event or circumstance does or does not occur. For example, “the heterocyclyl optionally substituted by an alkyl” means that an alkyl group can be, but need not be, present, and such a description includes the situation of the heterocyclyl being substituted by an alkyl and the heterocyclyl being not substituted by an alkyl.
  • “Substituted” refers to one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms, independently substituted by a corresponding number of substituents. It goes without saying that the substituents only exist in their possible chemical position. The person skilled in the art is able to determine whether the substitution is possible or impossible by experiments or theory without paying excessive efforts. For example, the combination of amino or hydroxy having free hydrogen and carbon atoms having unsaturated bonds (such as olefinic) may be unstable.
  • A “pharmaceutical composition” refers to a mixture of one or more of the compounds according to the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof with other chemical components, and other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration of a compound to an organism, which is conducive to the absorption of the active ingredient so as to show biological activity.
  • A “pharmaceutically acceptable salt” refers to a salt of the compound of the present invention, which is safe and effective in mammals and has the desired biological activity.
  • Synthesis Method of the Compound of the Present Invention
  • In order to achieve the object of the present invention, the present invention applies the following technical solutions:
  • Figure US20210230138A1-20210729-C00079
  • A method for preparing the compound of formula (I) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprises the following steps of:
  • in Step 1, a compound of formula A is reacted with boric aci or a orate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (I-A);
  • in Step 2, the compound of formula (I-A) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (I).
  • The reagent that provides an alkaline condition includes organic bases and inorganic bases. The organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide. The inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • The catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • The above reactions are preferably carried out in a solvent. The solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • Wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00080
  • ring A, G1-G4, R1-R4, r, q and n are as defined in formula (I).
  • Figure US20210230138A1-20210729-C00081
  • A method for preparing the compound of formula (Iaa) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprises the following steps of
  • in Step 1, a compound of formula (Iaa-2) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (Iaa-1);
  • in Step 2, the compound of formula (Iaa-1) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (Iaa).
  • Wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00082
  • ring A, G, G2, R1, R3, R4, r and n are as defined in formula (I).
  • The reagent that provides an alkaline condition includes organic bases and inorganic bases. The organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide. The inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • The catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • The above reactions are preferably carried out in a solvent. The solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof. Scheme III
  • Figure US20210230138A1-20210729-C00083
  • A method for preparing the compound of formula (Ibb) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprises the following steps of
  • in Step 1, a compound of formula (Ibb-2) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (Ibb-1);
  • in Step 2, the compound of formula (Ibb-1) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (Ibb).
  • Wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00084
  • ring A, G1, G2, R1, R3, R4, r and n are as defined in formula (I).
  • The reagent that provides an alkaline condition includes organic bases and inorganic bases. The organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide. The inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • The catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • The above reactions are preferably carried out in a solvent. The solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • Figure US20210230138A1-20210729-C00085
  • A method for preparing the compound of formula (II) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprises the following steps of
  • in Step 1, a compound of formula (B) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (II-A);
  • in Step 2, the compound of formula (II-A) is reacted with a compound of formula (I-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (II).
  • The reagent that provides an alkaline condition includes organic bases and inorganic bases. The organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide. The inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • The catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • The above reactions are preferably carried out in a solvent. The solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • Wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00086
  • ring A, R1, R3, R4, r and n are as defined in formula (I).
  • Figure US20210230138A1-20210729-C00087
  • A method for preparing the compound of formula (III) of the present invention or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprises the following steps of:
  • in Step 1, a compound of formula (B) is reacted with boric acid or a borate compound in the presence of a catalyst under an alkaline condition to obtain a compound of formula (II-A);
  • in Step 2, the compound of formula (II-A) is reacted with a compound of formula (III-B) in the presence of a catalyst under an alkaline condition to obtain the compound of formula (III).
  • The reagent that provides an alkaline condition includes organic bases and inorganic bases. The organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amine, potassium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide and sodium n-butoxide. The inorganic bases include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide.
  • The catalyst includes, but is not limited to, Pd/C, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, bis(dibenzylideneacetone)palladium, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium, [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium, 1,1′-bis(dibenzylphosphoryl)ferrocene palladium dichloride and tris(dibenzylideneacetone)dipalladium, and preferably [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • The above reactions are preferably carried out in a solvent. The solvent used includes, but is not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, glycol dimethyl ether, water, N,N-dimethylformamide, and mixtures thereof.
  • Wherein:
  • X is halogen;
  • M is
  • Figure US20210230138A1-20210729-C00088
  • R1, R3, R4, r and n are as defined in formula (I).
  • PREFERRED EMBODIMENTS
  • The present invention will be further described with reference to the following examples, but the examples should not be considered as limiting the scope of the present invention.
  • EXAMPLES
  • The structures of the compounds were identified by nuclear magnetic resonance (NMR) and/or mass spectrometry (MS). NMR shifts (δ) are given in 10−6 (ppm). NMR was determined by a Bruker AVANCE-400 machine. The solvents for determination were deuterated-dimethyl sulfoxide (DMSO-d6), deuterated-chloroform (CDCl3) and deuterated-methanol (CD3OD), and the internal standard was tetramethylsilane (TMS).
  • MS was determined by a FINNIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, type: Finnigan LCQ advantage MAX).
  • High performance liquid chromatography (HPLC) was determined on an Agilent HPLC 1200DAD, Agilent HPLC 1200VWD and Waters HPLC e2695-2489 high pressure liquid chromatographs.
  • Chiral HPLC was determined on an Agilent 1260 DAD high performance liquid chromatograph.
  • High performance liquid preparation was carried out on Waters 2767, Waters 2767-SQ Detecor2, Shimadzu LC-20AP and Gilson-281 preparative chromatographs.
  • Chiral preparation was carried out on a Shimadzu LC-20AP preparative chromatograph.
  • CombiFlash rapid preparation instrument used was Combiflash Rf200 (TELEDYNE ISCO).
  • Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate was used as the thin-layer silica gel chromatography (TLC) plate. The dimension of the silica gel plate used in TLC was 0.15 mm to 0.2 mm, and the dimension of the silica gel plate used in product purification was 0.4 mm to 0.5 mm.
  • Yantai Huanghai 200 to 300 mesh silica gel was generally used as a carrier for silica gel column chromatography.
  • Prep Star SD-1 (Varian Instruments Inc.) or SFC-multigram (Berger Instruments Inc.) was used for chiral preparative column chromatography.
  • The average kinase inhibition rates and IC50 values were determined by a NovoStar ELISA (BMG Co., Germany).
  • The known starting materials of the present invention can be prepared by the known methods in the art, or can be purchased from ABCR GmbH & Co. KG, Acros Organnics, Aldrich Chemical Company, Accela ChemBio Inc., or Dari chemical Company, etc.
  • Unless otherwise stated, the reactions were carried out under argon atmosphere or nitrogen atmosphere.
  • “Argon atmosphere” or “nitrogen atmosphere” means that a reaction flask is equipped with an argon or nitrogen balloon (about 1 L).
  • “Hydrogen atmosphere” means that a reaction flask is equipped with a hydrogen balloon (about 1 L).
  • Pressurized hydrogenation reactions were performed on a Parr 3916EKX hydrogenation instrument and a Qinglan QL-500 hydrogen generator or HC2-SS hydrogenation instrument.
  • In hydrogenation reactions, the reaction system was generally vacuumed and filled with hydrogen, with the above operation was repeated three times.
  • CEM Discover-S 908860 type microwave reactor was used in microwave reactions.
  • Unless otherwise stated, the solution refers to an aqueous solution.
  • Unless otherwise stated, the reaction temperature is room temperature from 20° C. to 30° C.
  • The reaction process in the examples was monitored by thin layer chromatography (TLC). The developing solvent used in the reactions, the eluent system in column chromatography and the developing solvent system in thin layer chromatography for purification of the compounds included: A: dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, and D: dichloromethane/ethyl acetate/methanol system. The ratio of the volume of the solvent was adjusted according to the polarity of the compounds, and a small quantity of alkaline reagent such as triethylamine or acidic reagent such as acetic acid can also be added for adjustment.
  • Example 1 6-(8-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00089
  • Step 1 8-Methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 1b
  • 6-Bromo-8-methylquinoline 1a (444 mg, 2.00 mmol, prepared according to the known method disclosed in “Journal of Organic Chemistry, 2014, 79(11), 5379-5385”), bis(pinacolato)diboron (508 mg, 2.00 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 10 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was added with 20 mL of ethyl acetate, washed with water (10 mL) and saturated sodium chloride solution (10 mL) successively, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography with developing solvent system B to obtain the title product 1b (320 mg), yield: 59.5%.
  • MS m/z (ESI): 270.1 [M+1].
  • Step 2 6-(8-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 1
  • Compound 1b (54 mg, 0.20 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (50 mg, 0.20 mmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2012, 55(5), 1898-1903”), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (29 mg, 0.04 mmol) and potassium carbonate (82 mg, 0.60 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was added with 20 mL of ethyl acetate, washed with water (10 mL) and saturated sodium chloride solution (10 mL) successively, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography with developing solvent system A to obtain the title product 1 (20 mg), yield: 32.2%.
  • MS m/z (ESI): 314.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.92 (m, 1H), 8.22-8.24 (d, 1H), 7.80 (s, 1H), 7.57 (s, 1H), 7.50-7.51 (m, 3H), 7.43-7.45 (m, 3H), 7.33-7.35 (m, 2H), 2.62 (s, 3H).
  • Example 2 6-(8-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 2
  • Figure US20210230138A1-20210729-C00090
  • Step 1 8-Fluoro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 2b
  • 6-Bromo-8-fluoroquinoline 2a (226 mg, 1.00 mmol), bis(pinacolato)diboron (305 mg, 1.20 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (146 mg, 0.20 mmol) and potassium acetate (294 mg, 3.00 mmol) were dissolved successively in 10 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 2b (220 mg), yield: 80.1%.
  • MS m/z (ESI): 274.1 [M+1].
  • Step 2 6-(8-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 2
  • Compound 2b (109 mg, 0.40 mmol), compound 1c (100 mg, 0.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (156 mg, 1.20 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 2 (20 mg), yield: 15.9%.
  • MS m/z (ESI): 318.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.95 (m, 1H), 8.38-8.40 (d, 1H), 7.91 (s, 1H), 7.58-7.62 (m, 3H), 7.41-7.46 (m, 4H), 7.35-7.37 (m, 2H).
  • Example 3 5-Phenyl-6-(quinolin-6-yl)-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00091
  • Step 1 6-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 3b
  • 6-Bromoquinoline 3a (1.0 g, 4.80 mmol, Accela ChemBio Inc.), bis(pinacolato)diboron (1.46 g, 5.76 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (0.7 g, 0.96 mmol) and potassium acetate (1.4 g, 14.40 mmol) were dissolved successively in 20 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 3b (1.2 g), yield: 98.4%.
  • MS m/z (ESI): 256.1 [M+1].
  • Step 2 5-Phenyl-6-(quinolin-6-yl)-1,2,4-triazin-3-amine 3
  • Compound 3b (203 mg, 0.80 mmol), compound 1c (200 mg, 0.80 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (116 mg, 0.16 mmol) and potassium carbonate (330 mg, 2.40 mmol) were dissolved successively in 24 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 3 (100 mg), yield: 42.0%.
  • MS m/z (ESI): 300.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90-8.91 (m, 1H), 8.32-8.34 (d, 1H), 8.10 (s, 1H), 7.89-7.91 (d, 1H), 7.52-7.59 (m, 4H), 7.43-7.44 (m, 3H), 7.33-7.35 (m, 2H).
  • Example 4 6-(8-Chloroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00092
  • Step 1 8-Chloro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 4b
  • 6-Bromo-8-chloroquinoline 4a (300 mg, 1.24 mmol), bis(pinacolato)diboron (378 mg, 1.49 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (181 mg, 0.25 mmol) and potassium acetate (364 mg, 3.72 mmol) were dissolved successively in 50 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 4b (260 mg), yield: 72.6%.
  • MS m/z (ESI): 290.5 [M+1].
  • Step 2 6-(8-Chloroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 4
  • Compound 4b (127 mg, 0.44 mmol), compound 1c (100 mg, 0.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.20 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 20 mL of ethyl acetate, washed with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 4 (30 mg), yield: 20.5%.
  • MS m/z (ESI): 334.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.01-9.02 (m, 1H), 8.39-8.41 (d, 1H), 8.05 (s, 1H), 7.80 (s, 1H), 7.59-7.66 (m, 3H), 7.44-7.47 (m, 3H), 7.36-7.38 (m, 2H).
  • Example 5 5-(5-Methylfuran-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00093
  • Step 1
  • 6-Bromo-5-(5-methylfuran-2-yl)-1,2,4-triazin-3-amine 5c 6-Bromo-1,2,4-triazin-3-amine 5a (1.0 g, 5.72 mmol, prepared according to the known method disclosed in “Journal of the American Chemical Society, 2015, 137(26), 8388-8391”), 6 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a reaction flask. The reaction solution was added with 2-methylfuran 5b (567 μL, 6.29 mmol), and stirred at room temperature for 17 hours. The reaction was stopped, and the reaction solution was added dropwise with saturated sodium bicarbonate solution to adjust the pH>7. The reaction solution was added with 30 mL of a pre-prepared aqueous solution of potassium hydroxide (962 mg, 17.14 mmol) and potassium hexacyanoferrate (5.65 g, 17.14 mmol), and stirred at room temperature for 1 hour. The reaction was stopped, and the reaction solution was extracted with ethyl acetate (150 mL×3). The organic phases were combined, added with silica gel, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system C to obtain the title product 5c (450 mg), yield: 30.9%.
  • MS m/z (ESI): 257.3 [M+1].
  • Step 2 5-(5-Methylfuran-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine 5
  • Compound 5c (80 mg, 0.31 mmol), compound 1b (127 mg, 0.47 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (23 mg, 0.031 mmol) and potassium carbonate (173 mg, 1.25 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 90° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 50 mL of water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, and concentrated under reduced pressure. The residue was purified by thin layer chromatography with developing solvent system B to obtain the title product 5 (33 mg), yield: 33.0%.
  • MS m/z (ESI): 318.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.99-8.98 (m, 1H), 8.41-8.39 (m, 1H), 7.97 (s, 1H), 7.69 (s, 1H), 7.61-7.58 (m, 1H), 7.39 (s, 2H), 6.19-6.14 (m, 2H), 2.75 (s, 3H), 2.21 (s, 3H).
  • Example 6 5-(Furan-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00094
  • Step 1 6-Bromo-5-(5-methylfuran-2-yl)-1,2,4-triazin-3-amine 6a
  • Compound 5a (1.0 g, 5.72 mmol), 6 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a reaction flask. The reaction solution was added with furan (457 μL, 6.29 mmol), and stirred at room temperature for 17 hours. The reaction was stopped, and the reaction solution was added dropwise with saturated sodium bicarbonate solution to adjust the pH>7. The reaction solution was added with 20 mL of a pre-prepared aqueous solution of potassium hydroxide (962 mg, 17.14 mmol) and potassium hexacyanoferrate (5.65 g, 17.14 mmol), and stirred at room temperature for 1 hour. The reaction was stopped, and the reaction solution was extracted with dichloromethane (100 mL×4). The organic phases were combined, added with silica gel, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system C to obtain the title product 6a (222 mg), yield: 16.1%.
  • MS m/z (ESI): 243.3 [M+1].
  • Step 2 5-(Furan-2-yl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine 6
  • Compound 6a (70 mg, 0.29 mmol), compound 1b (86 mg, 0.32 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (21 mg, 0.029 mmol) and potassium carbonate (160 mg, 1.16 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 90° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 50 mL of water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, and concentrated under reduced pressure. The residue was purified by thin layer chromatography with developing solvent system B to obtain the title product 6 (35 mg), yield: 39.8%.
  • MS m/z (ESI): 304.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.04-9.02 (m, 1H), 8.21-8.19 (m, 1H), 7.91 (s, 1H), 7.68 (s, 1H), 7.56 (s, 1H), 7.50-7.47 (m, 1H), 6.44-6.43 (m, 1H), 6.38-6.36 (m, 1H), 5.50 (s, 2H), 2.86 (s, 3H).
  • Example 7 6-(8-Methylquinolin-6-yl)-5-(thiophen-2-yl)-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00095
  • Step 1 6-Bromo-5-(thiophen-2-yl)-1,2,4-triazin-3-amine 7a
  • Compound 5a (1.0 g, 5.72 mmol), 6 mL of trifluoroacetic acid and 6 mL of dichloromethane were added to a reaction flask. The reaction solution was added with thiophene (503 μL, 6.29 mmol), and stirred at room temperature for 17 hours. The reaction was stopped, and the reaction solution was added dropwise with saturated sodium bicarbonate solution to adjust the pH>7. The reaction solution was added with 30 mL of a pre-prepared aqueous solution of potassium hydroxide (962 mg, 17.14 mmol) and potassium hexacyanoferrate (5.65 g, 17.14 mmol), and stirred at room temperature for 2 hours. The reaction was stopped, and the reaction solution was extracted with ethyl acetate (50 mL×3). The organic phases were combined, added with silica gel, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system C to obtain the title product 7a (400 mg), yield: 27.2%.
  • MS m/z (ESI): 259.2 [M+1].
  • Step 2 6-(8-Methylquinolin-6-yl)-5-(thiophen-2-yl)-1,2,4-triazin-3-amine 7
  • Compound 7a (70 mg, 0.27 mmol), compound 1b (81 mg, 0.30 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (20 mg, 0.027 mmol) and potassium carbonate (150 mg, 1.09 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 90° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 50 mL of water, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, and concentrated under reduced pressure. The residue was purified by thin layer chromatography with developing solvent system B to obtain the title product 7 (40 mg), yield: 46.0%.
  • MS m/z (ESI): 320.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.00-8.99 (m, 1H), 8.42-8.40 (m, 1H), 8.02 (s, 1H), 7.77-7.76 (m, 1H), 7.72 (s, 1H), 7.62-7.59 (m, 1H), 7.41 (s, 2H), 6.94-6.92 (m, 1H), 6.82-6.81 (m, 1H), 2.75 (s, 3H).
  • Example 8 5-(4-Fluorophenyl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00096
  • Step 1 5-(4-Fluorophenyl)-6-(8-methylquinolin-6-yl)-1,2,4-triazin-3-amine 8
  • Compound 1b (100 mg, 0.37 mmol), 6-bromo-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 8a (100 mg, 0.37 mmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2012, 55(5), 1898-1903”), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54 mg, 0.074 mmol) and potassium carbonate (154 mg, 1.12 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 8 (15 mg), yield: 12.2%.
  • MS m/z (ESI): 332.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.92-8.93 (m, 1H), 8.25-8.27 (d, 1H), 7.80 (s, 1H), 7.59 (s, 1H), 7.48-7.55 (m, 5H), 7.17-7.21 (m, 2H), 2.64 (s, 3H).
  • Example 9 5-Phenyl-6-(8-(trifluoromethyl)quinolin-6-yl)-1,2,4-triazin-3-amine 9
  • Figure US20210230138A1-20210729-C00097
  • Step 1 6-Bromo-8-(trifluoromethyl)quinoline 9b
  • 2-Amino-5-bromobenzotrifluoride 9a (1.1 g, 4.58 mmol), 1,2,3-propanetriol (1.69 g, 18.3 mmol) and iron (II) sulfate heptahydrate (204 mg, 0.73 mmol) were added to a reaction flask. The reaction solution was cooled to 0° C., and added dropwise with 0.8 mL of sulfuric acid. After completion of the addition, the reaction solution was heated to 120° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 20 mL of water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 9b (0.9 g), yield: 71.4%.
  • MS m/z (ESI): 210.6 [M+1].
  • Step 2 6-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-8-(trifluoromethyl)quinoline 9c
  • Compound 9b (276 mg, 1.00 mmol), bis(pinacolato)diboron (381 mg, 1.50 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (146 mg, 0.20 mmol) and potassium acetate (294 mg, 3.00 mmol) were dissolved successively in 15 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 9c (250 mg), yield: 77.4%.
  • MS m/z (ESI): 324.1 [M+1].
  • Step 3 5-Phenyl-6-(8-(trifluoromethyl)quinolin-6-yl)-1,2,4-triazin-3-amine 9
  • Compound 9c (129 mg, 0.40 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (100 mg, 0.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (156 mg, 1.20 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was added with 20 mL of ethyl acetate, washed with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 9 (50 mg), yield: 34.2%.
  • MS m/z (ESI): 368.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.04-9.05 (m, 1H), 8.47-8.49 (d, 1H), 8.39 (s, 1H), 7.97 (s, 1H), 7.63-7.71 (m, 3H), 7.44-7.45 (m, 3H), 7.34-7.38 (m, 2H).
  • Example 10 6-(8-Isopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 10
  • Figure US20210230138A1-20210729-C00098
  • Step 1 6-Bromo-8-isopropylquinoline 10b
  • 4-Bromo-2-isopropylaniline 10a (1.0 g, 4.70 mmol, prepared according to the known method disclosed in “Synthesis, 2013, 45(17), 2474-2480”), 1,2,3-propanetriol (2.1 g, 23.40 mmol) and iron (II) sulfate heptahydrate (0.2 g, 0.75 mmol) were added to a reaction flask. The reaction solution was cooled to 0° C., and added dropwise with 0.9 mL of sulfuric acid. After completion of the addition, the reaction solution was heated to 120° C., and stirred for 3 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 20 mL of water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 10b (0.6 g), yield: 51.7%.
  • MS m/z (ESI): 251.1 [M+1].
  • Step 2 8-Isopropyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 10c
  • Compound 10b (600 mg, 2.40 mmol), bis(pinacolato)diboron (731 mg, 2.88 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (350 mg, 0.48 mmol) and potassium acetate (705 mg, 7.20 mmol) were dissolved successively in 40 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 10c (450 mg), yield: 63.1%.
  • MS m/z (ESI): 298.2 [M+1].
  • Step 3 6-(8-Isopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 10
  • Compound 10c (118 mg, 0.40 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (100 mg, 0.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.20 mmol) were dissolved successively in 24 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 10 (40 mg), yield: 29.3%.
  • MS m/z (ESI): 342.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90-8.92 (m, 1H), 8.36-8.38 (d, 1H), 8.11 (s, 1H), 7.51-7.53 (m, 3H), 7.40-7.41 (m, 3H), 7.33-7.35 (m, 2H), 7.25 (m, 1H), 4.10-4.12 (m, 1H), 0.99-1.01 (s, 6H).
  • Example 11 6-(8-Ethylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 11
  • Figure US20210230138A1-20210729-C00099
  • Step 1 6-Bromo-8-ethylquinoline 1b
  • 4-Bromo-2-ethylaniline 11a (1.0 g, 5.00 mmol, Alfa), 1,2,3-propanetriol (2.3 g, 25.00 mmol) and iron (II) sulfate heptahydrate (0.22 g, 0.80 mmol) were added to a reaction flask. The reaction solution was cooled to 0° C., and added dropwise with 0.9 mL of sulfuric acid. After completion of the addition, the reaction solution was heated to 120° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 20 mL of water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 1b (0.9 g), yield: 76.3%.
  • MS m/z (ESI): 237.1 [M+1].
  • Step 2 8-Ethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 11c
  • Compound 1b (472 mg, 2.00 mmol), bis(pinacolato)diboron (610 mg, 2.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 30 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 3 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with elution system B to obtain the title product 11c (400 mg), yield: 70.7%.
  • MS m/z (ESI): 284.1 [M+1].
  • Step 3 6-(8-Ethylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 11
  • Compound 1c (113 mg, 0.40 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (100 mg, 0.40 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.20 mmol) were dissolved successively in 24 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 11 (20 mg), yield: 15.3%.
  • MS m/z (ESI): 328.4 [M+1].
  • 1H NMR (400 MHz, Pyridine-d5) δ 8.93-8.95 (m, 1H), 8.52 (br, 2H), 8.10 (s, 1H), 8.06-8.10 (m, 1H), 7.72 (s, 1H), 7.65-7.67 (m, 2H), 7.32-7.35 (m, 1H), 7.26-7.28 (m, 3H), 3.30-3.32 (m, 2H), 1.22-1.25 (t, 3H).
  • Example 12 6-(8-Cyclopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 12
  • Figure US20210230138A1-20210729-C00100
  • Step 1 4-Bromo-2-cyclopropylaniline 12b
  • 2-Cyclopropylaniline 12a (1.0 g, 7.52 mmol, prepared according to the method disclosed in the patent application “WO201314997”) was added to 100 mL of acetonitrile, followed by addition of N-bromosuccinimide (1.4 g, 7.89 mmol) and ammonium acetate (58 mg, 0.075 mmol). The reaction solution was stirred for 2 hours before the reaction was stopped. The reaction solution was added with 60 mL of water, and extracted with ethyl acetate (100 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 12b (0.85 g), yield: 53.5%.
  • MS m/z (ESI): 213.1 [M+1].
  • Step 2 6-Bromo-8-cyclopropylquinoline 12c
  • 4-Bromo-2-ethylaniline 12b (500 mg, 2.36 mmol), 1,2,3-propanetriol (1.08 g, 11.80 mmol) and iron (II) sulfate heptahydrate (105 mg, 0.38 mmol) were added to a reaction flask. The reaction solution was cooled to 0° C., and added dropwise with 0.5 mL of sulfuric acid. After completion of the addition, the reaction solution was heated to 120° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with 50 mL of water, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 12c (0.4 g), yield: 68.5%.
  • MS m/z (ESI): 249.1 [M+1].
  • Step 3 8-Cyclopropyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 12d
  • Compound 12c (100 mg, 0.40 mmol), bis(pinacolato)diboron (123 mg, 0.48 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (59 mg, 0.08 mmol) and potassium acetate (118 mg, 1.20 mmol) were dissolved successively in 10 mL of glycol dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 12d (94 mg), yield: 79.0%.
  • MS m/z (ESI): 296.2 [M+1].
  • Step 4 6-(8-Cyclopropylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 12
  • Compound 12d (94 mg, 0.32 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (80 mg, 0.32 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (47 mg, 0.064 mmol) and potassium carbonate (132 mg, 0.96 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 12 (10 mg), yield: 9.2%.
  • MS m/z (ESI): 340.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.92-8.94 (m, 1H), 8.35-8.38 (dd, 1H), 8.02 (s, 1H), 7.43-7.57 (m, 3H), 7.37-7.39 (m, 1H), 7.34-7.36 (m, 4H), 6.79-6.80 (m, 1H), 3.06-3.08 (m, 1H), 0.91-0.93 (m, 2H), 0.29-0.30 (m, 2H).
  • Example 13 (Comparative Example 1) 6-(Naphthalen-2-yl)-5-phenyl-1,2,4-triazin-3-amine
  • Figure US20210230138A1-20210729-C00101
  • 6-(Naphthalen-2-yl)-5-phenyl-1,2,4-triazin-3-amine 13
  • 2-Naphthylboronic acid 13a (55 mg, 0.32 mmol), 6-bromo-5-phenyl-1,2,4-triazin-3-amine 1c (80 mg, 0.32 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (47 mg, 0.064 mmol) and potassium carbonate (132 mg, 0.96 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 13 (20 mg), yield: 21.3%.
  • MS m/z (ESI): 299.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 7.80-7.87 (m, 3H), 7.31-7.52 (m, 10H).
  • Example 14 6-(4-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 14
  • Figure US20210230138A1-20210729-C00102
  • Step 1 4-Methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 14b
  • 6-Bromo-4-methylquinoline 14a (444 mg, 2 mmol, prepared according to the known method disclosed in “Tetrahedron, 2003, 59(6), 813-819”), bis(pinacolato)diboron (762 mg, 3 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 12 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 14b (460 mg), yield: 85.5%.
  • MS m/z (ESI): 270.4 [M+1].
  • Step 2 6-(4-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 14
  • Compound 14b (107 mg, 0.4 mmol), compound 1c (100 mg, 0.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.20 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 14 (15 mg), yield: 12%.
  • MS m/z (ESI): 314.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.74-8.75 (d, 1H), 8.02 (m, 1H), 7.90-7.93 (d, 1H), 7.71-7.73 (d, 1H), 7.63 (m, 2H), 7.42-7.44 (m, 3H), 7.34-7.36 (m, 3H), 2.46 (s, 3H).
  • Example 15 6-(4-Methylquinazolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 15
  • Figure US20210230138A1-20210729-C00103
  • Step 1 6-Bromo-4-methylquinazoline 15b
  • 1-(2-Amino-5-bromophenyl)ethanone 15a (1 g, 4.67 mmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2015, 58(14), 5522-5537”), triethyl orthoformate (1.04 g, 7.01 mmol) and ammonium acetate (540.15 mg, 7.01 mmol) were added to a reaction flask. The reaction solution was stirred at 110° C. for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature. The reaction solution was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 15b (500 mg), yield: 47.98%.
  • Step 2 4-Methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinazoline 15c
  • Compound 15b (360 mg, 1.61 mmol), bis(pinacolato)diboron (409.82 mg, 1.61 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (236.17 mg, 322.77 μmol) and potassium acetate (475.16 mg, 4.84 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 15c (330 mg), yield: 75.7%.
  • MS m/z (ESI): 271.1[M+1].
  • Step 3 6-(4-Methylquinazolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 15
  • Compound 15c (108 mg, 399.80 μmol), compound 1c (100.38 mg, 399.80 μmmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58.51 mg, 79.96 μmol) and potassium carbonate (165 mg, 1.2 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=4:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 15 (52 mg), yield: 41.38%.
  • MS m/z (ESI): 315.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 8.22 (s, 1H), 7.89-7.90 (m, 2H), 7.59 (m, 2H), 7.43-7.45 (m, 3H), 7.34-7.38 (m, 2H), 2.73 (s, 3H).
  • Example 16 6-(8-Fluoro-4-methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 16
  • Figure US20210230138A1-20210729-C00104
  • Step 1 6-Bromo-8-fluoro-4-methylquinoline 16c
  • 4-Bromo-2-fluoro-aniline 16b (9.99 g, 52.58 mmol, prepared according to the known method disclosed in “Tetrahedron Letters, 2015, 56(41), 5646-5650”) was dissolved in 300 mL of 1,4-dioxane, followed by dropwise addition of 5 mL of sulfuric acid. The reaction solution was heated to reflux, and then slowly added dropwise with 20 mL of but-3-en-2-one 16a (7.37 g, 105.15 mmol, prepared according to the known method disclosed in “Tetrahedron Letters, 2006, 47(37), 6635-6636”) in 1,4-dioxane over 1.5 hours. After completion of the addition, the reaction solution was heated to reflux for 2 hours. The reaction solution was cooled to room temperature, and concentrated under reduced pressure. The residue was added with water, added dropwise with saturated sodium bicarbonate solution until the pH of the mixture was 10, and extracted with ethyl acetate three times. The organic phases were combined, washed once with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 16c (3.5 g), yield: 27.73%.
  • MS m/z (ESI):240.0 [M+1].
  • Step 2 8-Fluorine-4-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 16d
  • Compound 16c (480 mg, 2.00 mmol), bis(pinacolato)diboron (761.59 mg, 3.00 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292.60 mg, 399.88 μmol) and potassium acetate (588.68 mg, 6.00 mmol) were added to 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 3 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 16d (460 mg), yield: 80.12%.
  • MS m/z (ESI): 288.1 [M+1].
  • Step 3 6-(8-Fluoro-4-methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 16
  • Compound 16d (110 mg, 383.09 μmol), compound 1c (96 mg, 383.09 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (56 mg, 76.62 μmol) and potassium carbonate (158.60 mg, 1.15 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 16 (18 mg), yield: 14.2%.
  • MS m/z (ESI): 332.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.76 (m, 1H), 7.78 (s, 1H), 7.52-7.55 (m, 3H), 7.41-7.43 (m, 4H), 7.34-7.36 (m, 2H), 2.42 (s, 3H).
  • Example 17 5-(2-Fluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 17
  • Figure US20210230138A1-20210729-C00105
  • Compound 14b (100 mg, 371.55 μmol), 6-bromo-5-(2-fluorophenyl)-1,2,4-triazin-3-amine 17a (99.97 mg, 371.55 μmol, prepared according to the method disclosed in the patent application “WO2016102672A2”), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54 mg, 74.35 μmol) and potassium carbonate (153.82 mg, 1.11 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 17 (50 mg), yield: 40.6%.
  • MS m/z (ESI): 332.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.70-8.71 (m, 1H), 7.92-7.94 (m, 1H), 7.84-7.87 (m, 1H), 7.80 (s, 1H), 7.60-7.67 (m, 3H), 7.48-7.50 (m, 1H), 7.09-7.36 (m, 2H), 7.04-7.09 (m, 1H), 2.32 (s, 3H).
  • Example 18 5-(4-Fluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 18
  • Figure US20210230138A1-20210729-C00106
  • Compound 14b (100 mg, 371.55 μmol), compound 8a (99.97 mg, 371.55 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.37 mg, 74.31 μmol) and potassium carbonate (153.82 mg, 1.11 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 18 (15 mg), yield: 12.18%.
  • MS m/z (ESI): 332.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.76-8.77 (m, 1H), 8.09 (m, 1H), 7.92-7.95 (d, 1H), 7.70-7.71 (m, 1H), 7.53 (m, 2H), 7.47-7.50 (m, 2H), 7.39-7.40 (m, 1H), 7.18-7.22 (m, 2H), 2.52 (s, 3H).
  • Example 19 5-(4-Fluorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine 19
  • Figure US20210230138A1-20210729-C00107
  • Compound 15c (100 mg, 370.19 μmol), compound 8a (99.61 mg, 370.19 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.17 mg, 74.04 μmol) and potassium carbonate (153.26 mg, 1.11 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 19 (50 mg), yield: 40.64%.
  • MS m/z (ESI): 333.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.29 (s, 1H), 7.88-7.90 (m, 2H), 7.59 (m, 2H), 7.47-7.51 (m, 2H), 7.19-7.23 (m, 2H), 2.78 (s, 3H).
  • Example 20 6-(8-Fluoro-4-methylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 20
  • Figure US20210230138A1-20210729-C00108
  • Compound 8a (100 mg, 371.65 μmol), compound 16d (106.71 mg, 371.65 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.39 mg, 74.33 μmol) and potassium carbonate (153.86 mg, 1.11 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 20 (50 mg), yield: 38.51%.
  • MS m/z (ESI): 350.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.77-8.78 (m, 1H), 7.83 (s, 1H), 7.46-7.55 (m, 6H), 7.19-7.21 (m, 2H), 2.50 (s, 3H).
  • Example 21 5-(2,4-Difluorophenyl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 21
  • Figure US20210230138A1-20210729-C00109
  • Compound 14b (68 mg, 252.65 μmol), 6-bromo-5-(2,4-difluorophenyl)-1,2,4-triazin-3-amine 21a (72.53 mg, 252.65 μmol, prepared according to the method disclosed in the patent application “WO2011095625A1”), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (36.97 mg, 50.53 μmol) and potassium carbonate (104.60 mg, 757.95 μmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 21 (40 mg), yield: 50.99%.
  • MS m/z (ESI): 350.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.74-8.75 (m, 1H), 7.96-7.98 (d, 1H), 7.85-7.89 (m, 2H), 7.75-7.78 (m, 1H), 7.65 (m, 2H), 7.35-7.37 (m, 1H), 7.28-7.30 (m, 1H), 7.19-7.25 (m, 1H), 2.42 (s, 3H).
  • Example 22 5-(4-Fluorophenyl)-6-[4-(trideuteromethyl)-6-quinolinyl]-1,2,4-triazin-3-amine 22
  • Figure US20210230138A1-20210729-C00110
  • Step 1 6-Bromo-4-(trideuteromethyl)quinoline 22a
  • Compound 14a (222 mg, 999.64 μmol) and benzoic acid (12.21 mg, 99.96 μmol) were dissolved in 1 mL of deuteroxide. The reaction solution was stirred at 100° C. overnight, and then added with saturated sodium bicarbonate solution, and extracted with ethyl acetate three times. The organic phases were combined, and dried over anhydrous sodium sulfate. The residue was purified by CombiFlash rapid preparation instrument with elution system B. The purified solid and benzoic acid (12.21 mg, 99.96 μmol) were added successively to 1 mL of deuteroxide. The reaction solution was stirred at 100° C. overnight, and then added with saturated sodium bicarbonate solution, and extracted with ethyl acetate three times. The organic phases were combined, and dried over anhydrous sodium sulfate. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 22a (100 mg), yield: 44.44%.
  • MS m/z (ESI): 225.0 [M+1].
  • Step 2 6-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-4-(trideuteromethyl)quinoline 22b
  • Compound 22a (100 mg, 444.25 μmol), bis(pinacolato)diboron (169.22 mg, 666.37 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (65.01 mg, 88.85 μmol) and potassium acetate (130.80 mg, 1.33 mmol) were added to 10 mL of 1,4-dioxane under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 22b (70 mg), yield: 57.9%.
  • MS m/z (ESI): 273.1 [M+1].
  • Step 3 5-(4-Fluorophenyl)-6-[4-(trideuteromethyl)-6-quinolinyl]-1,2,4-triazin-3-amine 22
  • Compound 22b (70 mg, 257.20 μmol), compound 8a (69.20 mg, 257.20 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (37.64 mg, 51.44 μmol) and potassium carbonate (106.48 mg, 771.59 μmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, cooled and filtrated. The residue was purified by CombiFlash rapid preparation instrument with elution system A, and the resulting crude product was purified by thin layer chromatography with developing solvent system A to obtain the title product 22 (29 mg), yield: 29.07%.
  • MS m/z (ESI): 335.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.76 (m, 1H), 8.08 (s, 1H), 7.92-7.95 (d, 1H), 7.68-7.70 (d, 1H), 7.47-7.50 (m, 4H), 7.38-7.39 (m, 1H), 7.20-7.22 (m, 2H).
  • Example 23 6-(4-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 23
  • Figure US20210230138A1-20210729-C00111
  • 4-Methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 23a (114 mg, 0.4 mmol, prepared according to the method disclosed in the patent application “WO2011084402A1”), compound 1c (100 mg, 0.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.2 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 23 (20 mg), yield: 15.3%.
  • MS m/z (ESI): 330.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.74-8.75 (m, 1H), 8.30 (m, 1H), 7.79-7.81 (d, 1H), 7.48-7.51 (m, 3H), 7.40-7.44 (m, 3H), 7.33-7.35 (m, 2H), 7.03-7.05 (m, 1H), 4.02 (s, 3H).
  • Example 24 6-(3-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 24
  • Figure US20210230138A1-20210729-C00112
  • Step 1 3-Fluoroquinolin-6-yl trifluoromethanesulfonate 24b
  • 3-Fluoroquinolin-6-ol 24a (489 mg, 3 mmol, prepared according to the known method disclosed in “Synlett, 2014, 25(6), 858-862”) and pyridine (474 mg, 6 mmol) were dissolved in 10 mL of dichloromethane. The reaction solution was added dropwise with trifluoromethanesulfonic anhydride (0.55 mL, 3.3 mmol) at 0° C., and stirred for 2 hours. The reaction solution was added with water, and extracted with dichloromethane (20 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 24b (520 mg), yield: 58.7%.
  • MS m/z (ESI): 296.4 [M+1].
  • Step 2 3-Fluoro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 24c
  • Compound 24b (100 mg, 0.34 mmol), bis(pinacolato)diboron (103 mg, 0.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (50 mg, 0.068 mmol) and potassium acetate (100 mg, 1 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 24c (70 mg), yield: 76%.
  • MS m/z (ESI): 274.4 [M+1].
  • Step 3 6-(3-Fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 24
  • Compound 24c (70 mg, 0.26 mmol), compound 1c (64 mg, 0.26 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (37 mg, 0.05 mmol) and potassium carbonate (106 mg, 0.77 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 24 (20 mg), yield: 25%.
  • MS m/z (ESI): 318.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.94-8.95 (m, 1H), 8.26-8.29 (m, 1H), 8.17 (m, 1H), 7.92-7.95 (d, 1H), 7.51-7.55 (m, 3H), 7.41-7.44 (m, 3H), 7.34-7.36 (m, 2H).
  • Example 25 6-(8-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 25
  • Figure US20210230138A1-20210729-C00113
  • Step 1 8-Methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 25b
  • 6-Bromo-8-methoxyquinoline 25a (530 mg, 2.2 mmol, prepared according to the known method disclosed in “Journal of the American Chemical Society, 2005, 127(1), 74-75”), bis(pinacolato)diboron (845 mg, 3.3 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (162 mg, 0.22 mmol) and potassium acetate (652 mg, 6.65 mmol) were dissolved successively in 10 mL of 1,4-dioxane under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 3 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated through celite. The filter cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 25b (410 mg), yield: 65%.
  • MS m/z (ESI): 286.1 [M+1].
  • Step 2 6-(8-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 25
  • Compound 25b (100 mg, 0.37 mmol), compound 1c (93 mg, 0.37 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (27 mg, 0.037 mmol) and potassium carbonate (10 mg, 0.074 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature, added with water, and extracted with ethyl acetate three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by preparative high performance liquid chromatography (Waters 2767-SQ Detecor2, elution system: ammonium bicarbonate, water, acetonitrile) to obtain the title product 25 (25 mg), yield: 21%.
  • MS m/z (ESI): 330.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, 1H), 8.25 (d, 1H), 7.62 (s, 1H), 7.51-7.53 (m, 3H), 7.42-7.44 (m, 3H), 7.34-7.36 (m, 2H), 7.98 (s, 1H), 3.65 (s, 3H).
  • Example 26 5-(3-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 26
  • Figure US20210230138A1-20210729-C00114
  • Compound 2b (101.50 mg, 371.65 μmol), 6-bromo-5-(3-fluorophenyl)-1,2,4-triazin-3-amine 26a (100 mg, 71.65 μmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2012, 55(5), 1898-1903”), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.39 mg, 74.33 μmol) and potassium carbonate (153.86 mg, 1.11 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 26 (45 mg), yield: 36.11%.
  • MS m/z (ESI): 336.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.94 (m, 1H), 8.37-8.39 (m, 1H), 7.88 (s, 1H), 7.53-7.61 (m, 3H), 7.44-7.47 (m, 1H), 7.26-7.35 (m, 3H), 7.15-7.17 (m, 1H).
  • Example 27 6-(4-Chloroquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 27
  • Figure US20210230138A1-20210729-C00115
  • 4-Chloro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolone 27a (200 mg, 690.69 μmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2011, 54(13), 4735-4751”), compound 8a (123.90 mg, 460.46 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (67.38 mg, 92.09 μmol) and potassium carbonate (190.63 mg, 1.38 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 27 (25 mg), yield: 15.43%.
  • MS m/z (ESI): 352.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.85-8.86 (m, 1H), 8.26 (m, 1H), 8.02-8.04 (d, 1H), 7.76-7.79 (m, 2H), 7.60 (m, 2H), 7.48-7.52 (m, 2H), 7.19-7.23 (m, 2H).
  • Example 28 6-(3-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 28
  • Figure US20210230138A1-20210729-C00116
  • Step 1 3-Methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 28b
  • 6-Bromo-3-methylquinoline 28a (250 mg, 1.13 mmol, prepared according to the method disclosed in the patent application “WO2006132739A2”), bis(pinacolato)diboron (429 mg, 1.69 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (165 mg, 0.225 mmol) and potassium acetate (331 mg, 3.38 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 28b (240 mg), yield: 79.2%.
  • MS m/z (ESI): 270.1 [M+1].
  • Step 2 6-(3-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 28
  • Compound 28b (107 mg, 0.4 mmol), compound 1c (100 mg, 0.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (58 mg, 0.08 mmol) and potassium carbonate (165 mg, 1.20 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 28 (50 mg), yield: 40%.
  • MS m/z (ESI): 314.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.76-8.77 (m, 1H), 7.06 (s, 1H), 7.95-7.96 (m, 1H), 7.85-7.87 (d, 1H), 7.51-7.55 (m, 3H), 7.41-7.43 (m, 3H), 7.32-7.34 (m, 2H), 2.47 (s, 3H).
  • Example 29 5-Phenyl-6-(quinazolin-6-yl)-1,2,4-triazin-3-amine 29
  • Figure US20210230138A1-20210729-C00117
  • Step 1 6-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)quinazoline 29b
  • 6-Bromoquinazoline 29a (418 mg, 2 mmol, prepared according to the known method disclosed in “Science of Synthesis, 2004, 16, 573-749”), bis(pinacolato)diboron (609 mg, 7.4 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (292 mg, 0.40 mmol) and potassium acetate (588 mg, 6.00 mmol) were dissolved successively in 20 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 29b (450 mg), yield: 87.9%.
  • MS m/z (ESI): 257.1 [M+1].
  • Step 2 5-Phenyl-6-(quinazolin-6-yl)-1,2,4-triazin-3-amine 29
  • Compound 29b (81 mg, 0.32 mmol), compound 1c (80 mg, 0.32 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (47 mg, 0.064 mmol) and potassium carbonate (132 mg, 0.96 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 29 (10 mg), yield: 5.1%.
  • MS m/z (ESI): 301.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.60 (s, 1H), 9.30 (s, 1H), 8.31 (s, 1H), 7.89-7.91 (d, 1H), 7.80-7.82 (m, 1H), 7.60 (m, 2H), 7.42-7.44 (m, 3H), 7.34-7.36 (m, 2H).
  • Example 30 5-(4-Fluorophenyl)-6-(quinolin-6-yl)-1,2,4-triazin-3-amine 30
  • Figure US20210230138A1-20210729-C00118
  • Compound 3b (94.81 mg, 371.65 μmol), compound 8a (100 mg, 371.65 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.39 mg, 74.33 μmol) and potassium carbonate (153.86 mg, 1.11 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 30 (5 mg), yield: 4.24%.
  • MS m/z (ESI): 318.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.91-8.92 (m, 1H), 8.36-8.38 (d, 1H), 8.11-8.12 (m, 1H), 7.92-7.94 (d, 1H), 7.47-7.59 (m, 6H), 7.17-7.21 (m, 2H).
  • Example 31 6-(8-Fluoro-4-methylquinolin-6-yl)-5-(2-fluorophenyl)-1,2,4-triazin-3-amine 31
  • Figure US20210230138A1-20210729-C00119
  • Compound 17a (100 mg, 371.65 μmol), compound 16d (106.71 mg, 371.65 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.39 mg, 74.33 μmol) and potassium carbonate (153.86 mg, 1.11 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 31 (50 mg), yield: 38.51%.
  • MS m/z (ESI): 350.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.74-8.75 (m, 1H), 7.67-7.69 (m, 4H), 7.59 (s, 1H), 7.50-7.52 (m, 1H), 7.36-7.41 (m, 2H), 7.09-7.11 (m, 1H), 2.31 (s, 3H).
  • Example 32 5-(4-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 32
  • Figure US20210230138A1-20210729-C00120
  • Compound 8a (100 mg, 371.65 μmol), compound 2b (101.50 mg, 371.65 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (54.39 mg, 74.33 μmol) and potassium carbonate (153.86 mg, 1.11 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A. The resulting crude product was purified by thin layer chromatography with developing solvent system D to obtain the title product 32 (20 mg), yield: 16.05%.
  • MS m/z (ESI): 336.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.96-8.97 (m, 1H), 8.41-8.43 (d, 1H), 7.91 (s, 1H), 7.64-7.66 (m, 1H), 7.63 (m, 2H), 7.46-7.52 (m, 3H), 7.19-7.24 (m, 2H).
  • Example 33 5-(2,4-Difluorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine 33
  • Figure US20210230138A1-20210729-C00121
  • Compound 21a (100 mg, 349.56 μmol), compound 15c (94.43 mg, 349.56 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (51.16 mg, 69.91 μmol) and potassium carbonate (144.72 mg, 1.05 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 33 (55 mg), yield: 44.91%.
  • MS m/z (ESI): 351.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 8.10 (s, 1H), 8.02-8.04 (d, 1H), 7.94-7.96 (d, 1H), 7.72-7.79 (m, 3H), 7.25-7.35 (m, 1H), 7.15-7.25 (m, 1H), 2.68 (s, 3H).
  • Example 34 6-(3-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 34
  • Figure US20210230138A1-20210729-C00122
  • Step 1 3-Methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 34b
  • 6-Bromo-3-methoxyquinoline 34a (120 mg, 0.5 mmol, prepared according to the method disclosed in the patent application “WO2012009194A1”), bis(pinacolato)diboron (192 mg, 0.76 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (74 mg, 0.1 mmol) and potassium acetate (148 mg, 1.5 mmol) were dissolved successively in 15 mL of dimethyl ether under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 4 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 34b (90 mg), yield: 62.9%.
  • MS m/z (ESI): 286.1 [M+1].
  • Step 2 6-(3-Methoxyquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 34
  • Compound 34b (79 mg, 0.279 mmol), compound 1c (70 mg, 0.279 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (41 mg, 0.056 mmol) and potassium carbonate (115 mg, 0.84 mmol) were dissolved successively in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was heated to 80° C., and stirred for 2 hours. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 34 (30 mg), yield: 33%.
  • MS m/z (ESI): 330.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.64-8.65 (m, 1H), 8.03-8.04 (m, 1H), 7.81-7.83 (d, 1H), 7.75-7.76 (m, 1H), 7.51 (m, 2H), 7.42-7.45 (m, 3H), 7.33-7.35 (m, 3H), 3.91 (s, 3H).
  • Example 35 5-(2-Fluorophenyl)-6-(8-fluoroquinolin-6-yl)-1,2,4-triazin-3-amine 35
  • Figure US20210230138A1-20210729-C00123
  • In accordance with the synthetic route of Example 32, the starting compound 8a in Step 1 was replaced with compound 17a, accordingly, the title product 35 (61 mg) was prepared.
  • MS m/z (ESI): 336.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90-8.91 (m, 1H), 8.29-8.31 (d, 1H), 7.77 (s, 1H), 7.60-7.66 (m, 3H), 7.58-7.59 (m, 1H), 7.42-7.49 (m, 2H), 7.11-7.34 (m, 1H), 7.08-7.11 (m, 1H).
  • Example 36 5-Phenyl-6-(quinoxalin-6-yl)-1,2,4-triazin-3-amine 36
  • Figure US20210230138A1-20210729-C00124
  • In accordance with the synthetic route of Example 23, the starting compound 23a in Step 1 was replaced with 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoxaline 36a (prepared according to the known method disclosed in “Organic Letters, 2009, 11(13), 2860-2863”), accordingly, the title product 36 (20 mg) was prepared.
  • MS m/z (ESI): 301.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.92-8.94 (m, 2H), 8.03-8.05 (m, 2H), 7.82-7.85 (m, 1H), 7.60 (m, 2H), 7.43-7.46 (m, 3H), 7.34-7.36 (m, 2H).
  • Example 37 6-(2-Methylquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 37
  • Figure US20210230138A1-20210729-C00125
  • In accordance with the synthetic route of Example 23, the starting compound 23a in Step 1 was replaced with 2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline 37a (prepared according to the known method disclosed in “Journal of the American Chemical Society, 2015, 137(4), 1593-1600”), accordingly, the title product 37 (20 mg) was prepared.
  • MS m/z (ESI): 314.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.19-8.21 (d, 1H), 8.02 (s, 1H), 7.78-7.80 (d, 1H), 7.50-7.53 (m, 3H), 7.40-7.43 (m, 4H), 7.32-7.34 (m, 2H), 2.65 (s, 3H).
  • Example 38 6-(3-Amino-5-phenyl-1,2,4-triazin-6-yl)quinoline-8-carbonitrile 38
  • Figure US20210230138A1-20210729-C00126
  • 6-Bromoquinoline-8-carboxamide 38b
  • Methyl 6-bromoquinoline-8-carboxylate 38a (400 mg, 1.5 mmol, prepared according to the method disclosed in the patent application “WO2011020193A1”) was dissolved in 15 mL of methanol. The reaction solution was added dropwise with 5 mL of 40% aqueous ammonia, and stirred overnight. The reaction solution was added with water, and extracted with ethyl acetate three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with elution system B to obtain the title product 38b (280 mg), yield: 74%.
  • MS m/z (ESI): 251.0 [M+1].
  • Step 2 6-Bromoquinoline-8-carbonitrile 38c
  • Compound 38b (190 mg, 0.76 mmol) was dissolved in 20 mL of dichloromethane. The reaction solution was added successively with triethylamine (115 mg, 1.14 mmol) and trifluoroacetic anhydride (238 mg, 1.14 mmol), and stirred for 2 hours. The reaction solution was added with water, and extracted with dichloromethane three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was pulped in methanol and filtrated. The filter cake was collected to obtain the title compound (180 mg), yield: 100%.
  • MS m/z (ESI): 232.9 [M+1].
  • In accordance with the similar synthetic route of Example 11, the starting compound 1b in Step 2 was replaced with 38c, accordingly, the title product 38 (25 mg) was prepared.
  • MS m/z (ESI): 325.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.08 (d, 1H), 8.51 (d, 1H), 8.45 (s, 1H), 8.13 (s, 1H), 7.73 (d, 1H), 7.71 (brs, 2H), 7.43-7.45 (m, 3H), 7.36-7.38 (m, 2H).
  • Example 39 6-(3-Morpholinoquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 39
  • Figure US20210230138A1-20210729-C00127
  • Step 1 4-(6-Chloroquinolin-3-yl)morpholine 39b
  • 3-Bromo-6-chloroquinoline 39a (266 mg, 1.1 mmol, prepared according to the known method disclosed in “Journal of Heterocyclic Chemistry, 2015, 52(4), 1019-1025”), morpholine (87 mg, 1 mmol), palladium acetate (12 mg, 0.05 mmol), (±)-2,2′-bis-(diphenylphosphino)-1,1′-binaphthalene (31 mg, 0.05 mmol) and cesium carbonate (652 mg, 2 mmol) were dissolved in 10 mL of tetrahydrofuran under an argon atmosphere. The reaction solution was heated to 70° C., and stirred overnight. The reaction was stopped, and the reaction solution was cooled to room temperature and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 39b (180 mg), yield: 66.2%.
  • MS m/z (ESI): 249.7 [M+1].
  • In accordance with the similar synthetic route of Example 11, the starting compound 11b in Step 2 was replaced with 39b, accordingly, the title product 39 (25 mg) was prepared.
  • MS m/z (ESI): 385.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.88 (s, 1H), 7.93 (s, 1H), 7.73-7.76 (d, 1H), 7.40-7.52 (m, 6H), 7.26-7.33 (m, 3H), 3.80 (m, 4H), 3.28 (m, 4H).
  • Example 40 6-(4-(Morpholinomethyl)quinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 40
  • Figure US20210230138A1-20210729-C00128
  • Step 1 (6-Bromoquinolin-4-yl)methanol 40b
  • Lithium aluminum hydride (150.76 mg, 3.97 mmol) was added to 50 mL of tetrahydrofuran. The reaction solution was added with 6-bromoquinoline-4-carboxylic acid 40a (1.0 g, 3.97 mmol, prepared according to the known method disclosed in “Chinese Chemical Letters, 2010, 21(1), 35-38”) in batches at 0° C., and stirred for 2 hours. The reaction solution was added with 5 mL of water, and filtrated through celite. The filter cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 40b (250 mg), yield: 26.5%.
  • Step 2 6-Bromo-4-(chloromethyl)quinoline 40c
  • Compound 40b (250 mg, 1.05 mmol) was added to 10 mL dichloromethane. The reaction solution was added with 2 mL of thionyl chloride, and stirred for 3 hours. The reaction solution was concentrated to dryness under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 40c (180 mg), yield: 66.8%.
  • In accordance with the similar synthetic route of Example 39, the starting compound 39a in Step 1 was replaced with compound 40c, accordingly, the title product 40 (5 mg) was prepared.
  • MS m/z (ESI): 399.1 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.82-8.83 (m, 1H), 8.16 (s, 1H), 7.98-8.00 (d, 1H), 7.86-7.88 (d, 1H), 7.53 (m, 2H), 7.42-7.47 (m, 4H), 7.35-7.37 (m, 2H), 3.61 (s, 2H), 3.49 (m, 4H), 2.30 (m, 4H).
  • Example 41 6-(3-(Morpholinomethyl)quinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 41
  • Figure US20210230138A1-20210729-C00129
  • In accordance with the similar synthetic route of Example 40, the starting compound 40b in Step 2 was replaced with compound 28a, accordingly, the title product 41 (5 mg) was prepared.
  • MS m/z (ESI): 399.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.82-8.83 (m, 1H), 8.16 (s, 1H), 8.01-8.06 (m, 1H), 7.84-7.86 (d, 1H), 7.48-7.50 (m, 3H), 7.37-7.41 (m, 3H), 7.30-7.32 (m, 2H), 3.64 (s, 2H), 3.56 (m, 4H), 2.39 (m, 4H).
  • Example 42 6-(3-Amino-5-phenyl-1,2,4-triazin-6-yl)quinoline-8-carboxamide 42
  • Figure US20210230138A1-20210729-C00130
  • In accordance with the synthetic route of Example 14, the starting compound 14a in Step 1 was replaced with compound 38b, accordingly, the title product 42 (14 mg) was prepared.
  • MS m/z (ESI): 343.3 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 10.16 (s, 1H), 9.01 (s, 1H), 8.55 (s, 1H), 8.45 (d, 1H), 8.19 (s, 1H), 7.90 (s, 1H), 7.64 (d, 1H), 7.58 (brs, 2H), 7.41-7.44 (m, 3H), 7.33-7.35 (m, 2H).
  • Example 43 6-(4-Morpholinoquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 43
  • Figure US20210230138A1-20210729-C00131
  • In accordance with the similar synthetic route of Example 39, the starting compound 39a in Step 1 was replaced with 6-bromo-4-chloroquinoline 43a (prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2015, 58(14), 5522-5537”), accordingly, the title compound 43 (50 mg) was prepared.
  • MS m/z (ESI): 385.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.66-8.68 (m, 1H), 8.01-8.02 (m, 2H), 7.71 (s, 1H), 7.46-7.48 (m, 4H), 7.35-7.38 (m, 3H), 6.92-6.93 (m, 1H), 3.56-3.58 (m, 4H), 2.71-2.73 (m, 4H).
  • Example 44 6-(4-Ethylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 44
  • Figure US20210230138A1-20210729-C00132
  • Step 1 6-Bromo-4-ethylquinoline 44a
  • Compound 14a (500 mg, 2.25 mmol) was dissolved in 50 mL of tetrahydrofuran. The reaction solution was added dropwise with diisopropylamino lithium (723.54 mg, 6.75 mmol) at −78° C., and stirred for 1 hour. The reaction solution was added with methyl iodide (3.20 g, 22.51 mmol), gradually warmed up to room temperature, and stirred overnight. The reaction solution was added with saturated aqueous ammonium chloride solution, and extracted with ethyl acetate three times. The organic phases were combined, washed with saturated sodium chloride solution once, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 44a (480 mg), yield: 90.3%.
  • In accordance with the similar synthetic route of Example 25, the starting compound 25a in Step 1 was replaced with compound 44a, and the starting compound 1c in Step 2 was replaced with compound 8a, accordingly, the title compound 44 (40 mg) was prepared.
  • MS m/z (ESI): 346.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.78-8.79 (m, 1H), 7.98-8.03 (m, 2H), 7.81-7.83 (d, 1H), 7.47-7.51 (m, 4H), 7.36-7.37 (m, 1H), 7.17-7.22 (m, 2H), 2.88-2.90 (m, 2H), 1.05-1.09 (t, 3H).
  • Example 45 6-(8-Fluoro-4-methylquinazolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 45
  • Figure US20210230138A1-20210729-C00133
  • Step 1 2-Amino-5-bromo-3-fluorobenzonitrile 45b
  • 2-Amino-3-fluorobenzonitrile 45a (1 g, 7.35 mmol, purchased from Shanghai Bide Pharmatech Ltd.) was dissolved in 50 mL of dichloromethane. The reaction solution was added with N-bromosuccinimide (1.37 g, 7.71 mmol), and stirred for 16 hours. The reaction solution was added with water, and extracted with dichloromethane (50 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 45b (1.25 g), yield: 79.13%.
  • MS m/z (ESI):215.0 [M+1].
  • Step 2 1-(2-Amino-5-bromo-3-fluorophenyl)ethan-1-one 45c
  • Compound 45b (1 g, 4.65 mmol) was dissolved in 60 mL of tetrahydrofuran. The reaction solution was added dropwise with methylmagnesium bromide (2.77 g, 23.25 mmol) at −10° C., and stirred for 4 hours. The reaction solution was added with water, and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 45c (900 mg), yield: 83.41%.
  • Step 3 6-Bromo-8-fluoro-4-methylquinazoline 45d
  • Compound 45c (0.9 g, 3.88 mmol), triethyl orthoformate (862 mg, 5.82 mmol) and ammonium acetate (448 mg, 5.82 mmol) were added to a 250 mL reaction flask. The reaction mixture was stirred at 110° C. for 2 hours, and then cooled. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 45d (300 mg), yield: 32.09%.
  • MS m/z (ESI):241.0 [M+1].
  • In accordance with the synthetic route of Example 34, the starting compound 34a in Step 1 was replaced with compound 45d, accordingly, the title product 45 (40 mg, yield: 30.16%) was prepared.
  • MS m/z (ESI): 333.4 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.16 (s, 1H), 8.03 (s, 1H), 7.76-7.79 (m, 1H), 7.65 (br, 2H), 7.44-7.46 (m, 3H), 7.38-7.40 (m, 2H), 2.72 (s, 3H).
  • Example 46 5-(4-Chlorophenyl)-6-(4-methylquinazolin-6-yl)-1,2,4-triazin-3-amine 46
  • Figure US20210230138A1-20210729-C00134
  • In accordance with the synthetic route of Example 15, the starting compound 1c in Step 3 was replaced with 6-bromo-5-(4-chlorophenyl)-1,2,4-triazin-3-amine 46a (prepared according to the method disclosed in the patent application “WO0201195625A1”), accordingly, the title product 46 (42 mg, yield: 34.38%) was prepared.
  • MS m/z (ESI): 348.8 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.12 (s, 1H), 8.30 (s, 1H), 7.87-7.90 (m, 2H), 7.62 (br, 2H), 7.45 (m, 4H), 2.79 (s, 3H).
  • Example 47 6-(4-Ethyl-8-fluoroquinolin-6-yl)-5-phenyl-1,2,4-triazin-3-amine 47
  • Figure US20210230138A1-20210729-C00135
  • Step 1 6-Bromo-4-ethyl-8-fluoroquinoline 47a
  • Compound 16c (480 mg, 2.00 mmol) was dissolved in 10 mL of tetrahydrofuran under an argon atmosphere. The reaction solution was added dropwise with diisopropylamino lithium (257.02 mg, 2.40 mmol) at −78° C., and stirred for 1 hour. The reaction solution was then added with methyl iodide (297.98 mg, 2.10 mmol), and stirred for 2 hours. The reaction solution was added with water, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, washed with saturated sodium chloride solution (20 mL), and dried over anhydrous sodium sulfate. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 47a (120 mg), yield: 23.6%.
  • In accordance with the synthetic route of Example 14, the starting compound 14a in Step 1 was replaced with compound 47a, accordingly, the title product 47 (30 mg, yield: 37.37%) was prepared.
  • MS m/z (ESI): 346.5[M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.81-8.82 (m, 1H), 7.79 (s, 1H), 7.58-7.66 (m, 1H), 7.47 (br, 2H), 7.36-7.45 (m, 6H), 2.81-2.82 (m, 2H), 0.99-1.03 (t, 3H).
  • Example 48 5-(2-Methylpyridin-4-yl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 48
  • Figure US20210230138A1-20210729-C00136
  • Step 1 2-(2-Methylpyridin-4-yl)-2-oxoacetaldehyde 48b
  • 1-(2-Methylpyridin-4-yl)ethan-1-one 48a (4.29 g, 31.74 mmol, prepared according to the known method disclosed in “Journal of Medicinal Chemistry, 2015, 58(12), 5028-5037”) was dissolved in 35 mL of dimethyl sulfoxide. The reaction solution was then added with 35 mL of hydrobromic acid, and stirred at 55° C. overnight. The reaction solution was used directly in the next step.
  • Step 2 5-(2-Methylpyridin-4-yl)-3-(methylthio)-1,2,4-triazine 48c
  • S-Methylisothiosemicarbazide hydroiodide (8.87 g, 38.06 mmol) and sodium bicarbonate (28 g, 333.31 mmol) were added to 300 mL of ethanol, followed by slow addition of the reaction solution containing the crude compound 48b (4.73 g, 31.71 mmol). After completion of the addition, the reaction solution was stirred at 80° C. for 1 hour. The reaction solution was concentrated under reduced pressure, added with water, and extracted with ethyl acetate (100 mL×2). The organic phases were combined, washed with water (80 mL×3) and saturated sodium chloride solution (80 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 48c (5.6 g), yield: 80.90%.
  • MS m/z (ESI): 219.4[M+1].
  • Step 3 5-(2-Methylpyridin-4-yl)-3-methylsulfonyl-1,2,4-triazine 48d
  • Compound 48c (5.4 g, 24.74 mmol) was dissolved in 180 mL of dichloromethane, followed by addition of m-chloroperoxybenzoic acid (8 g, 46.36 mmol). The reaction solution was stirred for 3 hours. The reaction solution was filtrated, and the filtrate was concentrated under reduced pressure to obtain the crude title product 48d (8.5 g), which was used directly in the next step without purification.
  • Step 4 5-(2-Methylpyridin-4-yl)-1,2,4-triazin-3-amine 48e
  • The crude compound 48d (8.5 g, 33.96 mmol) was dissolved in 80 mL of dioxane. The reaction solution was added with 20 mL of aqueous ammonia, and stirred for 1 hour. The reaction solution was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 48e (1.6 g), yield: 25.17%.
  • MS m/z (ESI): 188.1[M+1].
  • Step 5 6-Bromo-5-(2-methylpyridin-4-yl)-1,2,4-triazin-3-amine 48f
  • Compound 48e (1.6 g, 8.55 mmol) was added to 200 mL of acetonitrile. The reaction solution was then added with N-bromosuccinimide (3.80 g, 21.37 mmol) and trifluoroacetic acid (1.95 g, 17.09 mmol), and stirred for 64 hours. The reaction solution was concentrated under reduced pressure, and added with water. The aqueous phase was extracted with ethyl acetate (100 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 48f (1.87 g), yield: 82.22%.
  • Step 6 5-(2-Methylpyridin-4-yl)-6-(4-methylquinolin-6-yl)-1,2,4-triazin-3-amine 48
  • Compound 14b (100 mg, 375 μmol), compound 48f (101 mg, 375 μmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (55 mg, 75 μmol) and potassium carbonate (155 mg, 1.13 mmol) were dissolved in 12 mL of a mixed solution of 1,4-dioxane and water (V/V=5:1) under an argon atmosphere. The reaction solution was stirred at 80° C. for 2 hours. The reaction solution was cooled, and filtrated through celite. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system A to obtain the title product 48 (30 mg), yield: 24.31%.
  • MS m/z (ESI): 329.5[M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.77 (m, 1H), 8.37-8.38 (m, 1H), 8.07 (s, 1H), 7.93-7.95 (m, 1H), 7.70-7.72 (m, 1H), 7.65 (br, 2H), 7.38-7.40 (m, 2H), 7.06-7.07 (m, 1H), 2.50 (s, 3H), 2.42 (s, 3H).
  • Example 49 6-(4-Cyclopropylquinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 49
  • Figure US20210230138A1-20210729-C00137
  • Step 1 6-Bromo-4-iodoquinoline 49b
  • 5 mL of 4M hydrogen chloride in 1,4-dioxane were added to 6-bromo-4-chloroquinoline 49a (1 g, 4.12 mmol). The reaction solution was stirred for 10 minutes, and concentrated under reduced pressure for following use. The above concentrated residue was added with 60 mL of acetonitrile, followed by addition of sodium iodide (6.18 g, 41.24 mmol). The reaction solution was stirred under reflux for 16 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with saturated sodium bicarbonate solution, and extracted with ethyl acetate (20 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 49b (850 mg), yield: 61.72%.
  • MS m/z (ESI): 333.9[M+1].
  • Step 2 6-Bromo-4-cyclopropylquinoline 49c
  • Compound 49b (350 mg, 1.05 mmol), cyclopropylboronic acid (99 mg, 1.15 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (153 mg, 209 μmol) and potassium carbonate (433 mg, 3.14 mmol) were added to 30 mL of 1,4-dioxane under an argon atmosphere. The reaction solution was stirred at 80° C. for 16 hours. The reaction solution was cooled and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 49c (110 mg), yield: 42.30%.
  • MS m/z (ESI): 250.1[M+1].
  • In accordance with the synthetic route of Example 22, the starting compound 22a in Step 2 was replaced with compound 49c, accordingly, the title product 49 (40 mg, yield: 27.53%) was prepared.
  • MS m/z (ESI): 358.5[M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.74-8.75 (m, 1H), 8.34 (s, 1H), 7.96-7.98 (m, 1H), 7.78-7.80 (m, 1H), 7.48-7.52 (m, 4H), 7.18-7.22 (m, 2H), 7.14-7.15 (m, 1H), 2.27-2.28 (m, 1H), 0.97-0.99 (m, 2H), 0.71-0.73 (m, 2H).
  • Example 50 6-(8-Fluoro-4-methylquinazolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 50
  • Figure US20210230138A1-20210729-C00138
  • In accordance with the synthetic route of Example 22, the starting compound 22a in Step 2 was replaced with compound 45d, accordingly, the title product 50 (54 mg, yield: 41.48%) was prepared.
  • MS m/z (ESI): 351.0 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.18 (s, 1H), 8.08 (s, 1H), 7.76-7.79 (m, 1H), 7.66 (br, 2H), 7.49-7.53 (m, 2H), 7.20-7.25 (m, 2H), 2.77 (s, 3H).
  • Example 51 6-(4-(Difluoromethyl)quinolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 51
  • Figure US20210230138A1-20210729-C00139
  • Step 1 6-Bromoquinoline-4-carbaldehyde 51a
  • Compound 14a (1.0 g, 4.50 mmol) was dissolved in 20 mL of 1,4-dioxane. The reaction solution was added with 2 mL of water, followed by addition of selenium dioxide (1.5 g, 13.51 mmol). The reaction solution was stirred at 80° C. for 16 hours, and then cooled to room temperature, added with saturated sodium bicarbonate solution, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with elution system B to obtain the title compound 51a (670 mg), yield: 63.0%.
  • Step 2 6-Bromo-4-(difluoromethyl)quinoline 51b
  • Compound 51a (670 mg, 2.84 mmol) was dissolved in 20 mL of dichloromethane. The reaction solution was added dropwise with diethylaminosulfur trifluoride (915 mg, 5.68 mmol), and stirred for 16 hours. The reaction solution was added with saturated sodium bicarbonate solution, and stirred for 30 minutes. The reaction solution was extracted with dichloromethane (20 mL×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with elution system B to obtain the title compound 51b (630 mg), yield: 86.0%.
  • In accordance with the similar synthetic route of Example 25, the starting compound 25a in Step 1 was replaced with compound 51b, and the starting compound 1c in Step 2 was replaced with compound 8a, accordingly, the title compound 51 (55 mg) was prepared.
  • MS m/z (ESI): 368.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.05 (d, 1H), 8.21 (s, 1H), 8.08 (d, 1H), 7.80 (d, 1H), 7.75 (d, 1H), 7.53 (brs, 2H), 7.48 (t, 1H), 7.47 (q, 2H), 7.19 (t, 2H).
  • Example 52 5-(4-Fluorophenyl)-6-(4-(methyl-d3)quinazolin-6-yl)-1,2,4-triazin-3-amine 52
  • Figure US20210230138A1-20210729-C00140
  • Step 1 6-Bromo-4-(methyl-d3)quinazoline 52a
  • Compound 15b (200 mg, 0.90 mmol) was suspended in 4 mL of deuteroxide. The reaction solution was added with benzoic acid (10.95 mg, 0.09 mmol), and stirred at 100° C. for 48 hours. The reaction solution was added with saturated sodium bicarbonate solution, and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title compound 52a (150 mg), yield: 74.0%.
  • In accordance with the similar synthetic route of Example 25, the starting compound 25a in Step 1 was replaced with compound 52a, and the starting compound 1c in Step 2 was replaced with compound 8a, accordingly, the title compound 52 (18 mg) was prepared.
  • MS m/z (ESI): 336.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.11 (s, 1H), 8.28 (s, 1H), 7.85-8.28 (m, 2H), 7.59 (brs, 2H), 7.46-7.50 (m, 2H), 7.20 (t, 2H).
  • Example 53 6-(4-Ethylquinazolin-6-yl)-5-(4-fluorophenyl)-1,2,4-triazin-3-amine 53
  • Figure US20210230138A1-20210729-C00141
  • Step 1 1-(2-Amino-5-bromophenyl)propan-1-one 53b
  • 2-Amino-5-bromobenzonitrile 53a (500 mg, 2.54 mmol, prepared according to the known method disclosed in “European Journal of Medicinal Chemistry, 2014, 76, 341-343”) was dissolved in 10 mL of tetrahydrofuran. The reaction solution was cooled in an ice bath, added dropwise with 12.69 mL of 1.0 M ethyl magnesium bromide under an argon atmosphere, and stirred for 2 hours. The reaction solution was added with 6 M hydrochloric acid, and stirred for 2 hours. The reaction solution was added with saturated sodium carbonate solution, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title compound 53b (440 mg), yield: 76.02%.
  • Step 2 6-Bromo-4-ethylquinazoline 53c
  • Compound 53b (440 mg, 1.93 mmol), triethyl orthoformate (857.68 mg, 5.79 mmol) and ammonium acetate (451.14 mg, 5.79 mmol) were mixed, heated to 110° C. and stirred for 16 hours. The reaction solution was added with saturated sodium carbonate solution, and extracted with ethyl acetate (20 mL×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title compound 53c (280 mg), yield: 61.22%.
  • In accordance with the similar synthetic route of Example 25, the starting compound 25a in Step 1 was replaced with compound 53c, and the starting compound 1c in Step 2 was replaced with compound 8a, accordingly, the title compound 53 (45 mg) was prepared.
  • MS m/z (ESI): 347.5 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.15 (s, 1H), 8.21 (s, 1H), 7.94-8.01 (m, 2H), 7.57 (brs, 2H), 7.47-7.50 (m, 2H), 7.20 (t, 2H), 3.10 (q, 2H), 1.16 (t, 3H).
  • Example 54 5-(4-Fluorophenyl)-6-[4-(trifluoromethyl)quinolin-6-yl]-1,2,4-triazin-3-amine 54
  • Figure US20210230138A1-20210729-C00142
  • Step 1 6-Bromo-4-trifluoromethylquinolin-2(1H)-one 54b
  • Ethyl trifluoroacetoacetate (1.24 g, 6.75 mmol) and triethylamine (1.65 g, 16.28 mmol) were added to 20 mL of toluene. The reaction solution was then added dropwise with 3 mL of 2.7 M 4-bromoaniline 54a in toluene, and stirred under reflux for 16 hours. The reaction solution was concentrated under reduced pressure. The residue was added with 30 mL of dichloromethane, washed with water (20 mL×2) and saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The resulting intermediate was added with 6 mL of sulfuric acid, and stirred at 100° C. for 4 hours. The reaction solution was cooled to room temperature, added dropwise with saturated sodium bicarbonate solution to adjust the pH to greater than 10, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and filtrated. The filtrate was concentrated under reduced pressure, and the residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 54b (1.5 g), yield: 63.70%.
  • MS m/z (ESI): 291.9[M+1].
  • Step 2 6-Bromo-2-chloro-4-(trifluoromethyl)quinoline 54c
  • Compound 54b (1.5 g, 5.14 mmol) was added with phosphorus oxychloride (4.73 g, 30.82 mmol), and stirred at 100° C. for 5 hours. The reaction solution was cooled, added to ice water, stirred for 30 minutes, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 54c (1.3 g), yield: 81.52%.
  • Step 3 6-Bromo-4-trifluoromethylquinoline 54d
  • Compound 54c (500 mg, 1.61 mmol) was dissolved in 8 mL of trifluoroacetic acid. The reaction solution was then added with zinc powder (842 mg, 12.88 mmol), and stirred for 16 hours. The reaction solution was filtrated and concentrated under reduced pressure. The residue was added dropwise with 1 M sodium hydroxide until the pH was greater than 10, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by CombiFlash rapid preparation instrument with elution system B to obtain the title product 54d (200 mg), yield: 44.99%.
  • MS m/z (ESI): 275.8 [M+1].
  • In accordance with the synthetic route of Example 22, the starting compound 22a in Step 2 was replaced with compound 54d, accordingly, the title product 54 (30 mg) was prepared.
  • MS m/z (ESI): 386.4[M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 9.11-9.12 (m, 1H), 8.19-8.21 (m, 1H), 7.99-8.03 (m, 2H), 7.93-7.94 (m, 1H), 7.62 (br, 2H), 7.47-7.50 (m, 2H), 7.18-7.23 (m, 2H).
  • Example 55 5-(4-Fluorophenyl)-6-(4-methoxyquinazolin-6-yl)-1,2,4-triazin-3-amine 55
  • Figure US20210230138A1-20210729-C00143
  • Step 1 6-Bromo-4-methoxyquinazoline 55b
  • 6-Bromo-4-chloroquinazoline 55a (1.0 g, 4.10 mmol) was dissolved in 80 mL of methanol. The reaction solution was added with sodium methoxide (2.21 g, 41.05 mmol), and stirred for 3 hours. The reaction solution was concentrated to dryness, added with water, and filtrated. The filter cake was dried to obtain the product 55b (0.55 g), yield: 56.01%.
  • MS m/z (ESI): 239.1[M+1].
  • In accordance with the synthetic route of Example 22, the starting compound 22a in Step 2 was replaced with compound 55b, accordingly, the title product 55 (40 mg) was prepared. Yield: 31.29%.
  • MS m/z (ESI): 349.2 [M+1].
  • 1H NMR (400 MHz, DMSO-d6) δ 8.82 (s, 1H), 8.28 (s, 1H), 7.81-7.83 (m, 1H), 7.72-7.74 (m, 1H), 7.56 (br, 2H), 7.47-7.50 (m, 2H), 7.18-7.22 (m, 2H), 4.12 (s, 3H).
  • Test Examples:
  • Biological Assay
  • Test Example 1. Determination of the inhibition activity of the compounds of the present invention on the adenosine A2a receptor (A2aR) cAMP signaling pathway, the adenosine A2b receptor (A2bR) cAMP signaling pathway, the adenosine A1 receptor (AiR) cAMP signaling pathway and the adenosine A3 receptor (A3R) cAMP signaling pathway.
  • The inhibition activity of the compounds of the present invention on the adenosine A2a receptor (A2aR) cAMP signaling pathway, the adenosine A2b receptor cAMP signaling pathway, the adenosine A1 receptor cAMP signaling pathway and the adenosine A3 receptor cAMP signaling pathway was determined by the following method. The experimental method is briefly described as follows:
  • I. Experimental Materials and Instruments
  • 1. CHO-K1/A2aR cells (NM_000675.5) or CHO-K1/A2bR cells (NM_000676.2) or CHO-K1/A1R cells (NM_000674.2) or CHO-K1/A3R cells (NM_000677.3)
  • 2. Fetal bovine serum (Gibco, 10099-141)
  • 3. Bleomycin (Thermo, R25001) or G418 (ENZO, ALX-380-013-G005) or puromycin (Thermo, 10687-010)
  • 4. DMEM/F12 medium (GE, SH30023.01)
  • 5. Cell separation buffer (Thermo Fisher, 13151014)
  • 6. HEPES (Gibco, 42360-099)
  • 7. Bovine serum albumin (MP Biomedicals, 219989725)
  • 8. Rolipram (sigma, R6520-10MG)
  • 9. Adenosine deaminase (sigma, 10102105001)
  • 10. Forskolin (sigma, F6886)
  • 11. 2C1-IB-MECA(Tocrics, 1104/10)
  • 12. N6-cyclopentyladenosine (Tocris, 1702/50)
  • 13. Balanced salt buffer (Thermo, 14025-092)
  • 14. cAMP dynamic 2 kit (Cisbio, 62AM4PEB)
  • 15. 384-well plate (Corning, 4514) or (Nunc, 267462#)
  • 16. Ethyl carbazole (Torcis, 1691/10)
  • 17. PHERAstar multi-function microplate reader (Cisbio, 62AM4PEB)
  • II. Experimental Procedures
  • 2.1 Adenosine A2a Receptor
  • CHO-K1/A2aR cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 800 g/ml bleomycin. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 106 cells/ml. In the 384-well plate, each well was added with 5 μl of cell suspension, and 2.5 μl of test compound (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. Each well was then added with 2.5 μl of ethyl carbazole (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. The final concentrations of the compounds were: 10000, 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256, 0.00512, and 0.001024 nM. The final concentration of ethyl carbazole was 20 nM. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit. cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 5 μl of diluted cAMP-d2, followed by addition of 5 μl of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour. The HTRF signal values were read by the PHERAstar multi-function microplate reader. IC50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 1.
  • 2.2 Adenosine A2b Receptor
  • CHO-K1/A2bR cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 1 mg/ml G418. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 106 cells/ml. In the 384-well plate, each well was added with 5 μl of cell suspension, and 2.5 μl of test compound (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. Each well was then added with 2.5 μl of ethyl carbazole (4× concentration) (Torcis, 1691/10) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. The final concentrations of the compounds were: 100000, 10000, 1000, 100, 10, 1, 0.1 and 0 nM. The final concentration of ethyl carbazole was 1 μM. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit. cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 5 μl of diluted cAMP-d2, followed by addition of 5 μl of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour. The HTRF signal values were read by the PHERAstar multi-function microplate reader. IC50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 2.
  • 2.3 Adenosine A1 Receptor
  • CHO-K1/A1R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 1 mg/ml G418. The cells were digested with the cell separation buffer during the experiment. The cells were then resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 5×105 cells/ml. In the 384-well plate, each well was added with 12.5 μl of cell suspension, and 6.25 μl of test compound (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. Each well was then added with 6.25 μl of forskolin and N6-cyclopentyladenosine (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. The final concentrations of the compounds were: 100000, 10000, 1000, 100, 10, 1, 0.1 and 0 nM. The final concentrations of forskolin was 10 μM. The final concentrations of CPA was 10 nM. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit. cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 12.5 μl of diluted cAMP-d2, followed by addition of 12.5 μl of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour. The HTRF signal values were read by the PHERAstar multi-function microplate reader. IC50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 3.
  • 2.4 Adenosine A3 Receptor CHO-K/A3R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 10 μg/ml puromycin. The cells were digested with the cell separation buffer during the experiment. The cells were resuspended in the balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin and counted, and the cell density was adjusted to 5×105 cells/ml. In the 384-well plate, each well was added with 12.5 μl of cell suspension, and 6.25 μl of test compound (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. Each well was then added with 6.25 μl of forskolin and 2C1-IB-MECA (4× concentration) formulated with the balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 μM rolipram and 2.7 U/ml adenosine deaminase, and the plate was incubated at room temperature for 30 minutes. The final concentrations of the compounds were: 100000, 10000, 1000, 100, 10, 1, 0.1 and 0 nM. The final concentrations of forskolin was 10 μM. The final concentration of 2Cl-IB-MECA was 5 nM. Intracellular cAMP concentration was detected with the cAMP dynamic 2 kit. cAMP-d2 and Anti-cAMP-Eu-Cryptate were diluted respectively with the cAMP lysis buffer at a ratio of 1:4. Each well was added with 12.5 μl of diluted cAMP-d2, followed by addition of 12.5 μl of diluted Anti-cAMP-Eu-Cryptate, and the plate was incubated at room temperature in the dark for 1 hour. The HTRF signal values were read by the PHERAstar multi-function microplate reader. IC50 values of inhibition activity of the compounds were calculated by Graphpad Prism software, and are shown in Table 3.
  • TABLE 1
    IC50 values of the inhibition activity of the
    compounds of the present invention on the adenosine
    A2a receptor (A2aR) cAMP signaling pathway.
    Example No. IC50/nM (A2aR)
    1 0.5
    2 0.6
    3 0.7
    4 0.7
    5 0.4
    6 1.3
    7 3.1
    8 3.6
    14 0.1
    15 0.1
    16 0.2
    17 0.5
    18 0.2
    19 0.4
    20 1.5
    22 0.3
    23 0.4
    24 0.9
    25 1.5
    26 1.8
    27 2.2
    28 3.0
    29 3.5
    30 3.7
    31 3.8
    44 0.8
    45 1.2
    46 1.6
    47 1.6
    48 2.4
    49 2.5
    50 3.2
    51 3.5
    52 0.9
    13 (Example 1 (lxxii) 158.3
    of WO2011095625)
  • Conclusion: The compounds of the present invention have a significant inhibition activity on the adenosine A2a receptor. Compared with Comparative Example 1, the introduction of a nitrogen atom into the fused aryl moiety of the core structrue results in an unexpected inhibition activity of the compounds of the present invention on the adenosine A2a receptor. The structural difference between the compound of Comparative Example 1 and the compound of Example 3 is merely that the compound of Example 3 has the introduction of a nitrogen atom into the 5-position of the naphthyl of the compound of Comparative Example 1, but the inhibition activity on the adenosine A2a receptor differs by 225 times.
  • TABLE 2
    IC50 values of the inhibition activity of the
    compounds of the present invention on the adenosine
    A2b receptor (A2bR) cAMP signaling pathway.
    Example No. IC50/nM (A2bR)
    3 47
    4 4
    5 46
    14 3
    15 7
    16 18
    17 22
    19 25
    22 7
    23 17
    25 4
    45 25
    46 18
    52 14
  • Conclusion: The compounds of the present invention have a good inhibition activity on the adenosine A2b receptor.
  • TABLE 3
    IC50 values of the inhibition activity of the compounds of the
    present invention on the adenosine A1 receptor (A1R) cAMP signaling
    pathway and the adenosine A3 receptor cAMP signaling pathway.
    Example IC50/nM IC50/nM IC50 ratio IC50/nM IC50 ratio
    No. (A2aR) (A1R) (A1R/A2aR) (A3R) (A3R/A2aR)
    1 0.5 121 242 >104 >104
    2 0.6 133 221 >104 >104
    3 0.7 97 139 >104 >104
    15 0.1 248 2484 >104 >104
    16 0.2 161 803 >104 >104
    17 0.5 1349 2698 >104 >104
    18 0.2 190 948 >104 >104
    19 0.4 1423 3557 >104 >104
    20 1.5 617 411 >104 >103
    22 0.3 100 334 >104 >104
    31 3.8 1639 431 >104 >103
    44 0.8 646 807 >104 >104
    45 1.2 930 775 >104 >103
    48 2.4 3276 1365 >104 >103
    50 3.2 3257 1018 >104 >103
    52 0.9 876 973 >104 >104
  • Conclusion: The compounds of the present invention have a weak inhibition activity on the adenosine A1 receptor and the adenosine A3 receptor, indicating that the compounds of the present invention are selective for the adenosine A2a receptor and the adenosine A2b receptor, particularly for the adenosine A2a receptor.
  • Pharmacokinetics Evaluation
  • Test Example 2. Pharmacokinetics assay of the compounds of the present invention in mice
  • 1. Abstract
  • Mice were used as test animals. The drug concentration in plasma at different time points was determined by LC/MS/MS method after intragastrical administration of the compounds of Examples 2, 3, 17, 18, 19, 20, 31 and 44 to mice. The pharmacokinetic behavior of the compounds of the present invention was studied and evaluated in mice.
  • 2. Test Protocol
  • 2.1 Test Compounds
  • Compounds of Examples 2, 3, 17, 18, 19, 20, 31 and 44.
  • 2.2 Test Animals
  • Seventy-two C57 mice (female) were purchased from Shanghai Jiesijie Laboratory Animal Co., LTD, with Certificate No.: SCXK (Shanghai) 2013-0006, and equally divided into 8 groups (9 mice per group).
  • 2.3 Preparation of the Test Compounds
  • A certain amount of the test compound was weighed, and added with 5% by volume of DMSO, 5% by volume of tween 80 and 90% by volume of normal saline to prepare a 0.1 mg/mL colorless, clear and transparent solution.
  • 2.4 Administration
  • After an overnight fast, C57 mice were administered intragastrically the test compounds at an administration dosage of 2.0 mg/kg and an administration volume of 0.2 mL/10 g.
  • 3. Process
  • The mice were intragastrically administered the compounds of Examples 2, 3, 17, 18, 19, 20, 31 and 44. 0.1 ml of blood was taken before administration and at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0 and 24.0 hours after administration. The samples were stored in heparinized tubes, and centrifuged for 10 minutes at 3,500 rpm to separate the blood plasma. The plasma samples were stored at −20° C.
  • The content of the test compounds in the plasma of mice after intragastrical administration of the test compounds at different concentrations was determined: 25 μL of mouse plasma at each time after administration were taken, added with 50 μL of the internal standard solution of camptothecin (100 ng/mL) and 200 μL of acetonitrile, vortex-mixed for 5 minutes, and centrifuged for 10 minutes (4000 rpm). 5 μL of the supernatant were taken from the plasma samples for LC/MS/MS analysis.
  • 4. Results of pharmacokinetic parameters
  • Pharmacokinetic parameters of the compounds of the present invention are shown below:
  • Pharmacokinetics assay in mice (2 mg/kg)
    Apparent
    Plasma Area under Residence distribution
    concentration curve Half-life time Clearance volume
    Cmax AUC MRT CLz/F Vz/F
    No. (ng /mL) (ng /mL*h) (h) (h) (ml/min/kg) (ml/kg)
    Example 2 441 412 2.17 1.79 78.3 14736
    Example 3 395 419 2.3 1.61 78.6 15676
    Example 17 265 204 1.09 1.17 162 15329
    Example 18 402 495 1.59 1.43 65 9012
    Example 19 1960 5030 2.74 3.13 6.61 1567
    Example 20 866 1173 1.24 1.41 28 3047
    Example 31 302 339 2.02 2.02 95 16716
    Example 44 720 501 0.85 0.85 65 4794
  • Conclusion: The compounds of the present invention are well absorbed, and have a pharmacokinetic advantage.

Claims (15)

What is claimed is:
1. A compound selected from the group consisting of
Figure US20210230138A1-20210729-C00144
Figure US20210230138A1-20210729-C00145
Figure US20210230138A1-20210729-C00146
Figure US20210230138A1-20210729-C00147
Figure US20210230138A1-20210729-C00148
Figure US20210230138A1-20210729-C00149
Figure US20210230138A1-20210729-C00150
Figure US20210230138A1-20210729-C00151
Figure US20210230138A1-20210729-C00152
Figure US20210230138A1-20210729-C00153
Figure US20210230138A1-20210729-C00154
or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1, selected from the group consisting of:
Figure US20210230138A1-20210729-C00155
or a pharmaceutically acceptable salt thereof.
3. A compound selected from the group consisting of:
Figure US20210230138A1-20210729-C00156
and 3 or a pharmaceutically acceptable salt thereof.
4. The compound according to claim 3, selected from the group consisting of:
Figure US20210230138A1-20210729-C00157
or a pharmaceutically acceptable salt thereof.
5. The compound according to claim 3, selected from the group consisting of
Figure US20210230138A1-20210729-C00158
or a pharmaceutically acceptable salt thereof.
6. A pharmaceutical composition, comprising the compound according to claim 1, and one or more pharmaceutically acceptable carriers, diluents or excipients.
7. A pharmaceutical composition, comprising the compound according to claim 3, and one or more pharmaceutically acceptable carriers, diluents or excipients.
8. A method for treating a disease or condition in a subject, the method comprising administering to the subject the compound according to claim 1, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
9. A method for treating a disease or condition in a subject, the method comprising administering to the subject the compound according to claim 3, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
10. A method for treating a disease or condition ameliorated by inhibition of A2a and/or A2b receptor in a subject, the method comprising administering to the subject the compound according to claim 1, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
11. A method for treating a disease or condition ameliorated by inhibition of A2a and/or A2b receptor in a subject, the method comprising administering to the subject the compound according to claim 3, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
12. A method for treating a disease or condition in a subject, the method comprising administering to the subject the pharmaceutical composition according to claim 6, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
13. A method for treating a disease or condition in a subject, the method comprising administering to the subject the pharmaceutical composition according to claim 7, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
14. A method for treating a disease or condition ameliorated by inhibition of A2a and/or A2b receptor in a subject, the method comprising administering to the subject the pharmaceutical composition according to claim 6, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
15. A method for treating a disease or condition ameliorated by inhibition of A2a and/or A2b receptor in a subject, the method comprising administering to the subject the pharmaceutical composition according to claim 7, wherein the disease or condition is selected from the group consisting of cancer, depression, Parkinson's disease, Huntington's disease, Alzheimer's disease, abnormal movement disorder, sleep disorder, wherein the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder tumor, gallbladder cancer, cholangiocarcinoma, chorionic epithelioma and pediatric tumor.
US17/223,736 2017-01-13 2021-04-06 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine Abandoned US20210230138A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/223,736 US20210230138A1 (en) 2017-01-13 2021-04-06 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
CN201710023970 2017-01-13
CN201710023970.7 2017-01-13
CN201710874488 2017-09-25
CN201710874488.4 2017-09-25
PCT/CN2018/072308 WO2018130184A1 (en) 2017-01-13 2018-01-12 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
US201916477011A 2019-07-10 2019-07-10
US17/223,736 US20210230138A1 (en) 2017-01-13 2021-04-06 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US16/477,011 Continuation US11014904B2 (en) 2017-01-13 2018-01-12 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
PCT/CN2018/072308 Continuation WO2018130184A1 (en) 2017-01-13 2018-01-12 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine

Publications (1)

Publication Number Publication Date
US20210230138A1 true US20210230138A1 (en) 2021-07-29

Family

ID=62839274

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/477,011 Active 2038-01-14 US11014904B2 (en) 2017-01-13 2018-01-12 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
US17/223,736 Abandoned US20210230138A1 (en) 2017-01-13 2021-04-06 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/477,011 Active 2038-01-14 US11014904B2 (en) 2017-01-13 2018-01-12 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine

Country Status (6)

Country Link
US (2) US11014904B2 (en)
EP (1) EP3569596A4 (en)
JP (1) JP2020506885A (en)
CN (1) CN108884061B (en)
TW (1) TW201827422A (en)
WO (1) WO2018130184A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110655509B (en) * 2018-06-29 2023-01-24 江苏恒瑞医药股份有限公司 2-aminopyrimidine derivatives, preparation method and medical application thereof
CN110684020B (en) * 2018-07-05 2023-01-20 江苏恒瑞医药股份有限公司 2-aminopyrimidine derivatives, preparation method and medical application thereof
CN111094254B (en) * 2018-07-12 2022-07-26 江苏恒瑞医药股份有限公司 Heteroaryl derivative, preparation method and application thereof in medicine
TWI728404B (en) * 2018-07-13 2021-05-21 大陸商江蘇恒瑞醫藥股份有限公司 Crystalline form of 1,2,4-triazin-3-amine derivative and preparation method thereof
JP2022509388A (en) 2018-10-24 2022-01-20 リードエックスプロ アーゲー Functionalized aminotriazine
US20220017483A1 (en) * 2018-12-28 2022-01-20 Sichuan-Kelun-Biotech Biopharmaceutical Co., Ltd Aminopyridine compound, preparation method therefor and use thereof
CN111377873B (en) * 2018-12-28 2023-03-28 四川科伦博泰生物医药股份有限公司 Aminopyrimidine compounds, their preparation and use
CN111377906B (en) * 2018-12-28 2022-09-02 四川科伦博泰生物医药股份有限公司 Substituted pyrazine compounds, their preparation and use
MX2021008094A (en) 2019-01-11 2021-09-21 Omeros Corp Methods and compositions for treating cancer.
KR20200100429A (en) * 2019-02-18 2020-08-26 한국과학기술연구원 NOVEL PYRIDO[3,4-d]PYRIMIDIN-8-ONE DERIVATIVES AS PROTEIN KINASE INHIBITORS AND COMPOSITIONS FOR PREVENTING, IMPROVING OR TREATING CANCER CONTAINING THE SAME
WO2021076886A1 (en) 2019-10-18 2021-04-22 The Regents Of The University Of California 3-phenylsulphonyl-quinoline derivatives as agents for treating pathogenic blood vessels disorders
WO2021249475A1 (en) * 2020-06-10 2021-12-16 江苏恒瑞医药股份有限公司 Fused quinazoline derivative, preparation method therefor and application thereof in medicine
GB2615307A (en) 2022-01-28 2023-08-09 Adorx Therapeutics Ltd Antagonist compounds

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200716132A (en) 2005-05-03 2007-05-01 Smithkline Beecham Corp Novel chemical compounds
WO2007116106A1 (en) 2006-04-12 2007-10-18 Palobiofarma, S.L. Novel compounds as antagonists of adenosine a1 receptors
DE102007061763A1 (en) 2007-12-20 2009-06-25 Bayer Healthcare Ag Substituted azabicyclic compounds and their use
EP2467385A4 (en) 2009-08-18 2013-01-16 Merck Canada Inc Renin inhibitors
AU2010340087A1 (en) 2009-12-21 2012-07-26 Merck Sharp & Dohme Corp. Tyrosine kinase inhibitors
PT2531492T (en) 2010-02-05 2016-07-07 Heptares Therapeutics Ltd 1,2,4-triazine-4-amine derivatives
WO2011159302A1 (en) 2010-06-17 2011-12-22 Janssen Pharmaceutica Nv Arylindenopyrimidines for treating neurodegenerative and movement disorders while minimizing cardiac toxicity
WO2012009194A1 (en) 2010-07-12 2012-01-19 Merck Sharp & Dohme Corp. Tyrosine kinase inhibitors
JP6017766B2 (en) 2011-07-26 2016-11-02 株式会社Ihi Novel anticancer agent of metal salen complex compound
WO2014101120A1 (en) 2012-12-28 2014-07-03 Merck Sharp & Dohme Corp. Heterobicyclo-substituted-7-methoxy-[1,2,4]triazolo[1,5-c]quinazolin-5-amine compounds with a2a antagonist properties
WO2015027431A1 (en) 2013-08-29 2015-03-05 Merck Sharp & Dohme Corp. 2,2-difluorodioxolo a2a receptor antagonists
AU2015370911B2 (en) 2014-12-23 2020-07-09 Bergenbio Asa Inhibitors of Akt kinase

Also Published As

Publication number Publication date
US20190352278A1 (en) 2019-11-21
EP3569596A4 (en) 2020-08-26
WO2018130184A1 (en) 2018-07-19
CN108884061A (en) 2018-11-23
CN108884061B (en) 2021-11-16
JP2020506885A (en) 2020-03-05
US11014904B2 (en) 2021-05-25
TW201827422A (en) 2018-08-01
EP3569596A1 (en) 2019-11-20

Similar Documents

Publication Publication Date Title
US11014904B2 (en) 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
US11312705B2 (en) Heteroaryl[4,3-C]pyrimidine-5-amine derivative, preparation method therefor, and medical uses thereof
TWI542590B (en) 1,2-disubstituted heterocyclic compounds
RU2637936C2 (en) Inhibitors of lrrk2 kinases activity
EP3004057B1 (en) Heterocyclic derivatives and their use as stat 3 inhibitors
AU2016228634B2 (en) Substituted pyrimidine compounds as phosphatidylinositol 3-kinase delta inhibitor and use thereof
JP2011526294A (en) Disubstituted phenyl compounds as phosphodiesterase 10 inhibitors
JPWO2005087749A1 (en) 2-Aminoquinazoline derivatives
WO2019154294A1 (en) Pyrazolo[1,5-a][1,3,5]triazine-2-amine derivative, preparation method therefor and medical use thereof
CN111094254B (en) Heteroaryl derivative, preparation method and application thereof in medicine
CA3228411A1 (en) Sulfonamide derivative, preparation method therefor and medical use thereof
CN108467386B (en) Fused heteroaryl substituted 1,2, 4-triazine-3-amine derivatives, preparation method and medical application thereof
KR20210061202A (en) Benzonitrile-substituted Fused pyrimidine derivatives and their pharmaceutical use
WO2020156505A1 (en) 2-amionpyrimidine derivative, preparation method therefor and application thereof in medicines
CN110655509B (en) 2-aminopyrimidine derivatives, preparation method and medical application thereof
CN110684020B (en) 2-aminopyrimidine derivatives, preparation method and medical application thereof
JP2000154139A (en) Condensed ring pyrazine derivative
KR20190128671A (en) Deuterated benzimidazole compounds and their medical uses
JP2018154620A (en) Novel medicine composed of benzimidazole compound

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION