WO2019157959A1 - Composé de pyrimidine, son procédé de préparation et son utilisation médicale - Google Patents

Composé de pyrimidine, son procédé de préparation et son utilisation médicale Download PDF

Info

Publication number
WO2019157959A1
WO2019157959A1 PCT/CN2019/073874 CN2019073874W WO2019157959A1 WO 2019157959 A1 WO2019157959 A1 WO 2019157959A1 CN 2019073874 W CN2019073874 W CN 2019073874W WO 2019157959 A1 WO2019157959 A1 WO 2019157959A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
cycloalkyl
alkoxy
cyano
Prior art date
Application number
PCT/CN2019/073874
Other languages
English (en)
Chinese (zh)
Inventor
司聚同
姜美锋
李加艳
张丽云
樊平平
Original Assignee
恩瑞生物医药科技(上海)有限公司
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 恩瑞生物医药科技(上海)有限公司 filed Critical 恩瑞生物医药科技(上海)有限公司
Priority to CN201980005206.4A priority Critical patent/CN111247137A/zh
Publication of WO2019157959A1 publication Critical patent/WO2019157959A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Definitions

  • the present invention belongs to the field of medicine, and relates to a novel pyrimidine compound, a preparation method thereof and a pharmaceutical composition containing the same, and as a selective cyclin-dependent kinase 7 (CDK7) inhibitor in the prevention and/or treatment of human Diseases include use in cancer.
  • CDK7 selective cyclin-dependent kinase 7
  • Tumors including leukemia, are one of the major causes of clinical death in humans, with millions of patients worldwide dying of cancer each year.
  • Malignant tumors such as lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer, ovarian cancer, cervical cancer, esophageal cancer, nasopharyngeal carcinoma, leukemia and malignant lymphoma have extremely high mortality rates.
  • genetic screening, molecular diagnosis, and precision medicine for cancer have improved clinical early detection, correct diagnosis, and treatment for cancer patients, most cancers, especially advanced, refractory, relapsed, and drug-resistant malignancies have not been effective so far.
  • the method and medicine can be completely eradicated or cured. There is an urgent need for high-quality anticancer drugs with good specificity, high activity, low toxicity, and no drug resistance.
  • the mammalian cell cycle is a highly organized, orderly and precisely regulated cell mitosis process in which the genetic material of the cell replicates and is equally distributed among the two proliferating daughter cells.
  • Cell growth factors and cell cycle regulators play an important role in the cell cycle.
  • Cell cycle regulators are a class of self-synthesized proteins in cells. Abnormal activities of various cell cycle regulators (proteins) often cause abnormalities in the normal cell cycle, leading to different types of diseases, such as when cell proliferation is not controlled. Cell transformation forms cancer cells.
  • Cyclin Dependent Kinase is a group of serine/threonine protein kinases that act synergistically with cyclin (Cyclin) and are key regulators of cell cycle progression and transcription. CDK can bind to cyclins to form heterodimers, in which CDK is a catalytic subunit, cyclin is a regulatory subunit, and different Cyclin-CDK complexes are phosphorylated to different substrates in cells. Advance and transformation of different phases of the cell cycle.
  • CDK1 to CDK20 in which CDK11 has two genes CDK11A and CDK11B
  • CDKL1 to CDKL5 CDK-type genes
  • Amino acid sequences are highly evolutionarily conserved.
  • CDK direct cell cycle regulation of CDK
  • CDK such as CDK1, CDK2, CDK3, CDK4 and CDK6
  • transcriptional function CDK such as CDK7, CDK 8, CDK 9, CDK 11, CDK 12 and CDK13.
  • Direct cell cycle regulation of CDK directly regulates the progression of the cell cycle, and its phosphorylation substrate is a cell cycle-associated protein.
  • Transcriptional Function CDK regulates gene transcription by phosphorylating the RNA polymerase II complex.
  • Clinical data found that in different types of malignant tumors and leukemia patient samples such as skin cancer, melanoma, lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer or colon cancer and acute myeloid leukemia, different CDK frequent gene mutations, expansion Increased overexpression, which is closely related to the occurrence, development, and/or maintenance of malignant cell phenotypes, as well as patient survival and drug resistance.
  • CDK Crohn's disease .
  • CDK4/6 selective inhibitors Palbociclib, Ribociclib and Abemaciclib Otto T et al. (2017) Nat Rev Cancer 17(2): 93-115; Kwapisz D (2017) Breast Cancer Res Treat. 166(1): 41-54; Vijayaraghavan S et al. (2017) Target Oncol.2017 Dec 7; Ingham M et al. (2017) J Clin Oncol. 35 (25): 2949-2959; Abou Zahr A et al.
  • CDK4/6 and CDK5 have tumor immune regulation function, and selective inhibition of CDK4/6 or CDK5 can enhance the effect of tumor immunotherapy, further demonstrating that CDK is an important target protein for tumor therapy (Dorand RD et al. (2016) Science .353(6297): 399-403; Goel S et al. (2017) Nature. 548 (7668): 471-475; Deng J et al. (2017) Cancer Discov. 8(2); 216–33; Zhang J et al. Person (2016) Nature. 553 (7686): 91-95).
  • CDK inhibitors Over the years, many different types of CDK inhibitors have undergone extensive preclinical and clinical studies, but to date only the CDK4/6 highly selective inhibitors Pabsini, Ribociclib and Abemaciclib have been successfully applied to hormonal receptor-positive, Clinical treatment of HER2-negative advanced or recurrent breast cancer, Pabsini and Ribociclib need to be combined with letrozole, Abemaciclib alone or in combination with Fulvestrant.
  • Pan-CDK inhibitors such as Alvocidib and Seliciclib are flavonoids, and Alvocidib and ATP competitively inhibit CDK1, CDK2, CDK4 and CDK6 with IC 50 values of approximately 40 nM; Seliciclib inhibits CDK5, Cdc2 and CDK2, IC 50 were 0.2 ⁇ M, 0.65 ⁇ M and 0.7 [mu; but did not show antitumor activity in clinical research and promising pre clinic.
  • Second-generation pan-CDK inhibitors such as Dinaciclib, AT7519, Milciclib, TG02, CYC065, and RGB-286638 are highly active and inhibit multiple CDKs, although they enter different phases of clinical trials, respectively, but these inhibitors alone do not show Good clinical effects and high clinical side effects.
  • CDK7 is unique in mammalian CDK and regulates cell cycle and gene transcription.
  • CDK7 is present as a CDK activating enzyme (CAK) heterotrimeric complex that is capable of fully activating CDK1/2 by phosphorylating the CDK1/2 activation domain conserved residue (T-loop), a process that is cell cycle Required for the process; in the nucleus, CDK7 forms a complex with RNA polymerase (RNAP) II, which is responsible for phosphorylating the C-terminal domain (CTD) of RNAP II, which is a necessary step in the initiation of gene transcription.
  • RNAP RNA polymerase
  • CTD C-terminal domain
  • CDK7 selective inhibitors are expected to be effective new targeted drugs for the treatment of cancer.
  • CDK7 high-selective inhibitor SY-1365 (THZ1) has entered human clinical trials.
  • the preclinical test of this compound shows good anti-tumor activity and is a potential new type of CDK7 anti-tumor inhibitor (Lücking U et al. (2017) Chem Med Chem. 12 (21): 1776-1793; Kwiatkowski N et al. (2014) Nature. 511 (7511): 616-20), but the stability of this compound in vivo (T1/2 in mouse plasma) It is limited to 45 minutes). Therefore, there is an urgent need for a more efficient and toxic side effect CDK7 inhibitor.
  • the present inventors discovered a novel pyrimidine compound in the development of a long-term selective inhibitor of CDK7, which can effectively inhibit the growth of CDK7-positive leukemia cell MOLM-13 in vitro, and its IC 50 value can reach Yana Moore. Concentration is expected to be developed as a new and effective anti-tumor drug.
  • the object of the present invention is to provide a novel orally administrable small molecule compound which is selective, high in activity and low in toxicity, and can be used as a cyclin-dependent kinase 7 (CDK7) inhibitor for prevention and/or prevention. Treatment of human diseases including cancer.
  • the present invention relates to a novel pyrimidine compound which is capable of covalently binding to CDK7 and inhibits the growth of CDK7-positive tumor cells in vitro, and has an IC 50 value of subnanomolar concentration.
  • the present invention provides a compound of the formula (I),
  • Each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, nitro, -N(R y )(R z ), alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl Base, heteroaryl, -NHC(O)R x , -C(O)N(R y )(R z ), -OR u OR x , -OR x or -OR u N(R y )(R z
  • said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl group is further optionally selected from the group consisting of halogen, alkyl, alkoxy, haloalkoxy, cyano, Substituted by one or more substituents of amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl,
  • R 2 and R 3 are each independently selected from hydrogen, halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, -NHC(O)R x or -C(O) N(R y )(R z ), wherein the alkyl, alkoxy, cycloalkyl and heterocyclic groups are optionally further selected from the group consisting of halogen, alkyl, alkoxy, haloalkoxy, cyano, amino Substituting one or more substituents of a nitro group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group;
  • R 4 and R 5 are each independently selected from hydrogen, halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, -NHC(O)R x or -C(O) N(R y )(R z ), wherein the alkyl, alkoxy, cycloalkyl and heterocyclic groups are optionally further selected from the group consisting of halogen, alkyl, alkoxy, haloalkoxy, cyano, amino Substituting one or more substituents of a nitro group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group;
  • Each R 6 is each independently selected from a Q group
  • Each R 7 is independently selected from the group consisting of -NHC(O)R and Q groups;
  • R 8 and R 9 are the same or different and are each independently selected from the group consisting of -NHC(O)R and Q groups;
  • Q is selected from the group consisting of hydrogen, halogen, hydroxy, cyano, nitro, -N(R y )(R z ), alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or a heteroaryl group, wherein the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl group is further optionally selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy Substituted by one or more substituents in the group, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
  • R is selected from alkenyl, alkynyl, cyano, optionally further selected from halo, alkyl, haloalkyl, alkoxy, haloalkoxy or -R u N(R y ) ( Substituting one or more groups of R z );
  • R u is selected from an alkylene group, an alkenylene group, or an alkynylene group
  • R x is selected from hydrogen, alkyl, cycloalkyl, aryl or heteroaryl, and the alkyl, cycloalkyl, aryl or heteroaryl group is optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkane Substituted with one or more substituents of oxy, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
  • R y and R z are each independently selected from hydrogen, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; the alkyl, alkoxy, alkene
  • the base, alkynyl, cycloalkyl, aryl or heteroaryl group is optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl Substituting one or more substituents of a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group; or
  • R y and R z together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl group, which is optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkyl Substituted by one or more substituents of oxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
  • R 8 is selected from -NHC(O)R, and R 9 is selected from a Q group; or,
  • R 9 is selected from -NHC(O)R, and R 8 is selected from the group consisting of Q;
  • R is selected from alkenyl, alkynyl, cyano, and the alkenyl and alkynyl are optionally further substituted with one or more groups selected from the group consisting of hydrogen, alkyl or R u N(R y )(R z ) ;
  • R u , R y , R z , Q groups are as defined in the general formula (I).
  • R 8 is selected from -NHC(O)R, and R 9 is selected from the Q group; or,
  • R 9 is selected from -NHC(O)R, and R 8 is selected from the group consisting of Q;
  • R is selected from alkenyl, alkynyl, cyano, and the alkenyl and alkynyl are optionally further substituted with one or more groups selected from the group consisting of hydrogen, alkyl or R u N(R y )(R z ) ;
  • R u is selected from a C 1 -C 6 alkylene group
  • R y and R z are each independently selected from hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl; or
  • R y and R z together with the nitrogen atom to which they are attached form a 5- to 7-membered nitrogen-containing heterocyclic group, preferably morpholinyl, piperidinyl, piperazinyl, azepanyl, tetrahydropyrrolyl,
  • the 5- to 7-membered nitrogen-containing heterocyclic group is optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy Substituted by one or more groups of a C 3 -C 7 cycloalkyl group;
  • the Q group is selected from the group consisting of hydrogen, halogen, hydroxy, cyano, nitro, amino, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 7 cycloalkyl.
  • Each R 7 is each independently selected from -NHC(O)R;
  • R is selected from alkenyl, alkynyl, cyano, and the alkenyl and alkynyl are optionally further substituted with one or more groups selected from the group consisting of hydrogen, alkyl or R u N(R y )(R z ) ;
  • R u is selected from a C 1 -C 6 alkylene group
  • R y and R z are each independently selected from hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl; or
  • R y and R z together with the nitrogen atom to which they are attached form a 5- to 7-membered nitrogen-containing heterocyclic group, preferably morpholinyl, piperidinyl, piperazinyl, azepanyl, tetrahydropyrrolyl,
  • the 5- to 7-membered nitrogen-containing heterocyclic group is optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy Substituted by one or more groups of a C 3 -C 7 cycloalkyl group.
  • Each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, nitro, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl, -NHC(O)R x , -C(O N(R y )(R z ), -OR u OR x , -OR x , wherein the C 1 -C 6 alkyl group, C 3 -C 7 cycloalkyl group is optionally further selected from halogen, cyano group Substituting one or more substituents of an amino group, a nitro group, a hydroxyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group;
  • R u is selected from a C 1 -C 6 alkylene group
  • R x is selected from hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl or 5- to 7-membered heteroaryl, said C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl or The 5- to 7-membered heteroaryl group is optionally further substituted with one or more groups of a halogen, a hydroxyl group, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group or a heteroaryl group.
  • R 2 and R 3 are each independently selected from hydrogen, halogen, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, -NHC(O)R x or -C(O)N(R y ) (R z ), wherein the alkyl group, alkoxy group, cycloalkyl group and heterocyclic group are optionally further selected from the group consisting of halogen, alkyl, alkoxy, haloalkoxy, cyano, amino, nitro, hydroxy Substituted by one or more substituents of a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, or a heteroaryl group;
  • R x , R y , R z are as defined in the general formula (I).
  • R 2 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, cyano, -NHC(O)R x or -C(O)N(R y )(R z );
  • R 3 is selected from hydrogen
  • R x is selected from the group consisting of hydrogen, alkyl, cycloalkyl, and the alkyl, cycloalkyl is optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, nitro Substituting one or more substituents of a hydroxyl group or a hydroxyalkyl group;
  • R y and R z are each independently selected from hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; the alkyl, cycloalkyl, aryl or heteroaryl group is optionally further selected Substituted by one or more substituents of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl; or
  • R y and R z together with the nitrogen atom to which they are attached form a nitrogen-containing heterocyclic group or heteroaryl group, which is optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy Substituting one or more substituents of a haloalkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, or a hydroxyalkyl group.
  • R 4 and R 5 are each independently selected from the group consisting of hydrogen, halogen, cyano, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy .
  • Each R 6 is each independently selected from a Q group
  • Q is selected from the group consisting of hydrogen, halogen, hydroxy, cyano, nitro, amino, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 7 cycloalkyl.
  • Exemplary compounds of the invention include, but are not limited to, the following compounds:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • the intermediate compound M3 and the aniline intermediate compound M4 are reacted under an acid catalysis at a suitable temperature and a suitable solvent to obtain a compound of the formula (I);
  • the solvent is preferably isopropanol, isoamyl alcohol, secondary pentanol or dioxane;
  • the acid is preferably hydrochloric acid, sulfuric acid, methanesulfonic acid, p-toluenesulfonic acid or benzenesulfonic acid;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , n, m, p are as defined in the formula (I).
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) according to the invention, together with a pharmaceutically acceptable carrier.
  • the invention further relates to the use of a compound of the formula (I) according to the invention or a pharmaceutical composition comprising the same, for the preparation of a medicament for the prevention and/or treatment of a cancer of a mammal, including a human, said cancer Including, but not limited to, non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • the invention further relates to a method of preventing and/or treating cancer in a mammal, including a human, comprising administering to a patient in need thereof a prophylactically or therapeutically effective amount of a compound of the formula (I) according to the invention or A pharmaceutical composition comprising the same, including, but not limited to, non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, gastric cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, gastric cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • the invention further relates to a compound of the formula (I) according to the invention or a pharmaceutical composition comprising the same, which is used as a medicament.
  • the invention further relates to a compound of the formula (I) according to the invention or a pharmaceutical composition comprising the same, which is useful as a medicament for the prevention and/or treatment of cancer, including but not limited to, non-solid tumors Such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • non-solid tumors Such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer.
  • alkyl refers to a saturated aliphatic hydrocarbon group, including straight chain and branched chain groups of 1 to 20 carbon atoms. It includes a linear or branched alkyl group having from 1 to 18 carbon atoms, preferably from 1 to 10 carbon atoms, more preferably from 1 to 6 carbon atoms, even more preferably from 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl, Isopyl, n-heptyl, isoheptyl, n-octyl, isooctyl, n-decyl, n-decyl and the like.
  • the "alkyl group” further includes a cyclic alkyl group having 3 to 10 carbon atoms, preferably 3 to 8 carbon atoms, more preferably 4 to 6 carbon atoms, such as a cyclopropyl group, a cyclobutyl group, a cyclopentyl group or a ring. Hexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclodecyl, decahydronaphthyl, norbornane, adamantyl.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups, independently selected from alkyl, alkenyl, Alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycle Alkoxy, cycloalkylthio, heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
  • alkylene refers to a saturated straight or branched aliphatic hydrocarbon radical having two residues derived from the removal of two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms.
  • Non-limiting examples of alkylene include, but are not limited to, methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2 ) CH 2 )-, 1,1-propylene (-CH(CH 2 CH 3 )-), 1,2-propylene (-CH 2 CH(CH 3 )-), 1,3-propylene (-CH 2 CH 2 CH 2 -), 1,4-butylene (-CH 2 CH 2 CH 2 CH 2 -) and 1,5-pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 - )Wait.
  • alkenyl refers to a straight or branched hydrocarbon chain radical containing at least one double bond consisting of carbon and hydrogen atoms and attached to the remainder of the molecule by a single or double bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethenyl, propenyl, butenyl, pentenyl, pentadienyl, hexenyl.
  • the alkenyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, hetero A cycloalkylthio group, an oxo group, an amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • alkenylene refers to a straight or branched hydrocarbon chain radical containing at least one double bond consisting of carbon and hydrogen atoms, having two substituents removed from the same carbon atom or two different carbon atoms of the parent. A residue derived from two hydrogen atoms which is attached to the remainder of the molecule by a single bond or a double bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethenylene, propenylene, butenylene, pentenylene, hexylene, and the like.
  • alkynyl refers to a straight or branched hydrocarbon chain radical containing at least one triple bond consisting of a carbon atom and a hydrogen atom and attached to the remainder of the molecule by a single or triple bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethynyl, propynyl, butynyl, pentynyl, hexynyl.
  • the alkynyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, hetero A cycloalkylthio group, an oxo group, an amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • alkynylene refers to a straight or branched hydrocarbon chain radical containing at least one triple bond consisting of a carbon atom and a hydrogen atom, having two identical carbon atoms or two different carbon atoms from the parent. Residues derived from two hydrogen atoms are removed which are attached to the remainder of the molecule by a single or triple bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethynylene, propynylene, butynylene, pentynylene, hexynylene, and the like.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably the cycloalkyl ring comprises from 3 to The 10 carbon atoms, most preferably the cycloalkyl ring contains 3 to 7 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptene
  • the alkenyl group, the cyclooctyl group and the like are preferably a cyclopropyl group or a cyclohexenyl group.
  • Polycyclic cycloalkyl groups include spiro, fused, and bridged cycloalkyl groups.
  • the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O)m A hetero atom (where m is an integer of 0 to 2), but does not include a ring moiety of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon. It preferably comprises from 3 to 12 ring atoms, wherein from 1 to 4 are heteroatoms, more preferably the heterocyclyl ring contains from 3 to 10 ring atoms, more preferably the heterocyclyl ring contains from 5 to 7 ring atoms.
  • Non-limiting examples of monocyclic heterocyclic groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, tetrahydrofuranyl, azepan Alkyl and the like.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cyclo
  • aryl refers to an all-carbon monocyclic or fused polycyclic ring (i.e., a ring that shares a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably from 5 to 10 members, more preferably from 5 to 7 members. Even more preferred are phenyl and naphthyl, most preferably phenyl.
  • the aryl group may be a completely aromatic group such as a phenyl group, a naphthyl group, an anthracenyl group, a phenanthryl group or the like.
  • the aryl group may also contain a combination of an aromatic ring and a non-aromatic ring, for example, ruthenium, osmium, and iridium.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples comprising:
  • the aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups, independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkane.
  • Base amino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkane A thio group, an amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, more preferably 5 to 7 members, even more preferably 5 or 6 members, such as thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, or the like.
  • heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples of which include:
  • the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • Alkoxy means -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl, cycloalkyl are as defined above. Non-limiting examples include methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy and the like.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, and an alkane group.
  • haloalkyl refers to an alkyl group wherein one or more hydrogen atoms are replaced by a halogen, wherein alkyl is as defined above.
  • Non-limiting examples include chloromethyl, trifluoromethyl, 1-chloro-2-fluoroethyl, 2,2-difluoroethyl, 2-fluoropropyl, 2-fluoropropan-2-yl, 2, 2,2-Trifluoroethyl, 1,1-difluoroethyl, 1,3-difluoro-2-methylpropyl, 2,2-difluorocyclopropyl, (trifluoromethyl)cyclopropane Base, 4,4-difluorocyclohexyl and 2,2,2-trifluoro-1,1-dimethyl-ethyl.
  • haloalkoxy refers to an alkoxy group wherein one or more hydrogen atoms are replaced by a halogen, wherein the alkoxy group is as defined above.
  • halogen includes fluoro, chloro, bromo and iodo.
  • amino means -NH 2.
  • nitro refers to -NO 2 .
  • cyano refers to -CN.
  • hydroxy refers to an -OH group.
  • hydroxyalkyl refers to an alkyl group substituted by a hydroxy group, wherein the alkyl group is as defined above.
  • hydroxyalkoxy refers to an alkoxy group substituted by a hydroxy group, wherein the alkoxy group is as defined above.
  • acyl refers to -C(O)R, wherein R refers to alkyl, cycloalkyl, alkenyl, alkynyl, wherein alkyl, cycloalkyl, alkenyl, alkynyl are as defined above.
  • R refers to alkyl, cycloalkyl, alkenyl, alkynyl, wherein alkyl, cycloalkyl, alkenyl, alkynyl are as defined above.
  • Non-limiting examples include acetyl, propionyl, butyryl, valeryl, hexanoyl, vinyl, acryloyl groups.
  • acylamino refers to -NHC (O) R, or -C (O) NH 2, wherein R denotes an alkyl group, an alkenyl group, an alkynyl group, where the definition of an alkyl group, an alkenyl group, an alkynyl group as described above.
  • R denotes an alkyl group, an alkenyl group, an alkynyl group, where the definition of an alkyl group, an alkenyl group, an alkynyl group as described above.
  • Non-limiting examples include formylamino, acetylamino, propionylamino, butyrylamino, pentanoylamino, hexanoylamino, vinylamido, acrylamido.
  • ester group refers to -C(O)OR, wherein R refers to alkyl or cycloalkyl, wherein alkyl, cycloalkyl are as defined above.
  • R refers to alkyl or cycloalkyl, wherein alkyl, cycloalkyl are as defined above.
  • Non-limiting examples include ethyl ester groups, propyl ester groups, butyl ester groups, amyl ester groups, cyclopropyl ester groups, cyclobutyl ester groups, cyclopentyl ester groups, cyclohexyl ester groups.
  • substituents are selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a halogenated alkyl group, an alkoxy group, an aryl group, a halogenated aryl group, an aryloxy group, an aralkyl group, an aralkyloxy group, and a hetero group.
  • substituents can also be further substituted.
  • the alkyl group as a substituent is also optionally selected from one or more groups selected from a halogen atom, a hydroxyl group, an alkoxy group, an alkylamino group, a pyrrolidinyl group, a phenyl group, a pyridyl group, or a halogenated phenyl group.
  • the heterocyclic group as a substituent is also optionally substituted with one or more groups selected from a halogen atom, an alkyl group, and an alkoxy group.
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • Drug Combination refers to the simultaneous or sequential application of two or more drugs for therapeutic purposes.
  • the present invention mainly employs the following synthetic routes and technical solutions.
  • the intermediate compound M3 is obtained by a substitution reaction of a hydrazine intermediate M1 and a pyrimidine intermediate M2 at a suitable temperature and a base under a catalyst catalyzed in a suitable solvent;
  • the base may be, for example, hydrogenated Sodium, potassium carbonate, cesium carbonate, etc.
  • the solvent may be, for example, DMF, NMP or the like, and the catalyst may be, for example, 1-hydroxybenzotriazole (HOBT).
  • Substituted pyrimidine intermediates M2 are generally commercially available.
  • Step 1 An aniline starting material M5 and a carboxylic acid starting material M6 are subjected to a condensation reaction to obtain an intermediate M7 under a suitable temperature and basic conditions in a suitable solvent;
  • the base may be, for example, triethyl An amine, N-methylmorpholine or the like
  • the solvent may be, for example, DCM, THF, etc.
  • the condensing agent may be, for example, HATU, HBTU, TBTU, or the like;
  • Step 2 reduction of the nitro group of intermediate M7 to an amino group to obtain intermediate M8; reduction of the nitro group can be achieved, for example, in an iron powder ammonium chloride system or a H 2 /palladium carbon system;
  • Step 3 The carboxylic acid starting material M9 is reacted in a suitable solvent under the action of a suitable chlorinating reagent and a catalyst to form an acid chloride
  • the solvent may be, for example, tetrahydrofuran, dichloromethane or the like
  • the chlorinating reagent may be
  • the catalyst may be, for example, DMF or the like;
  • the resulting acid chloride is then reacted with the intermediate M8 by nucleophilic substitution in a suitable solvent under appropriate conditions and under basic conditions to give the intermediate M10, which may be, for example, triethylamine, pyridine or N-methyl.
  • a suitable solvent for example, triethylamine, pyridine or N-methyl.
  • Morpholine or the like the solvent may be, for example, DCM, THF or the like;
  • Step 4 Intermediate M10 is deprotected under acidic conditions in a suitable solvent to give intermediate M4.
  • the acidic conditions may be, for example, trifluoroacetic acid, ethyl acetate, and the like, and the solvent may be, for example, DCM. , ethyl acetate, and the like.
  • Step 1 The intermediate carboxylic acid M11 is reacted in a suitable solvent under the action of a suitable chlorinating reagent and a catalyst to form an acid chloride, which may be, for example, tetrahydrofuran, dichloromethane or the like.
  • a suitable chlorinating reagent and a catalyst for example, oxalyl chloride, thionyl chloride, phosphorus oxychloride, etc.
  • the catalyst may be, for example, DMF or the like
  • the obtained acid chloride and intermediate M8 are passed through a suitable solvent under appropriate temperature and basic conditions.
  • the nucleophilic substitution reaction produces the intermediate M12, which may be, for example, triethylamine, pyridine, N-methylmorpholine or the like, and the solvent may be, for example, DCM, THF, DMF, or the like;
  • Step 2 Intermediate M12 is obtained by a substitution reaction in an appropriate solvent under appropriate temperature and basic conditions to obtain an intermediate product M10;
  • the base may be, for example, potassium carbonate, cesium carbonate or the like, and the solvent may be, for example, DMF, NMP, etc.
  • Step 3 Intermediate M10 is deprotected under acidic conditions in a suitable solvent to form intermediate M4.
  • the acidic conditions may be, for example, trifluoroacetic acid, ethyl acetate, and the like, and the solvent may be, for example, DCM. , ethyl acetate, and the like.
  • the intermediate M3 and the aniline intermediate M4 are reacted under acid catalysis at a suitable temperature and a suitable solvent to obtain a compound of the formula (I);
  • the solvent may be, for example, isopropanol, isoamyl alcohol, secondary pentanol, dioxane or the like
  • the acid may be, for example, hydrochloric acid, sulfuric acid, methanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid or the like.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , n, m, p, R u , R z , R y are as in the formula (I) Defined.
  • the pharmaceutically acceptable salt of the compound of the formula (I) of the present invention may be an acid addition salt or a base addition salt.
  • the acid may be a mineral acid including, but not limited to, hydrochloric acid, sulfuric acid, phosphoric acid, hydrobromic acid; or may be an organic acid including, but not limited to, citric acid, maleic acid, oxalic acid, formic acid, acetic acid, propionic acid, valeric acid.
  • glycolic acid glycolic acid, benzoic acid, fumaric acid, trifluoroacetic acid, succinic acid, tartaric acid, lactic acid, glutamic acid, aspartic acid, salicylic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid, p-benzenesulfonic acid .
  • the base may be an inorganic base including, but not limited to, sodium hydroxide, potassium hydroxide, magnesium hydroxide, calcium hydroxide; or may be an organic base including, but not limited to, ammonium hydroxide, triethylamine, N, N- Dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzyl phenyl An amine, arginine or lysine; or may be an alkali metal salt, including but not limited to: lithium, potassium and sodium salts; or may be an alkaline earth metal salt, including but not limited to: barium, calcium and magnesium salts;
  • the transition metal salt includes, but is not limited to, a zinc salt; or other metal salts including, but not limited to, sodium hydrogen phosphate and disodium hydrogen phosphate.
  • the compound of the formula (I) or a pharmaceutically acceptable salt is prepared into a clinically usable pharmaceutical composition.
  • the pharmaceutical preparations thereof include, but are not limited to, oral preparations such as tablets, gels, soft/hard capsules, emulsions, dispersible powders, granules, water/oil suspoemulsions; injections Including intravenous injection, intramuscular injection, intraperitoneal injection, rectal suppository, intracranial injection, these dosage forms may be aqueous solutions or oily solutions; topical preparations include creams, ointments, gels, water/oil solutions and packs Formulations; inhalation dosage forms include fine powders, liquid aerosols, and various dosage forms suitable for in vivo implantation.
  • the pharmaceutical composition of the present invention may be added with a pharmaceutically acceptable carrier, diluent or excipient as needed.
  • a pharmaceutically acceptable carrier diluent or excipient as needed.
  • Carriers for solid oral formulations include, but are not limited to, mannitol, lactose, starch, magnesium stearate, cellulose, glucose, sucrose, cyclodextrin, and the intestinal absorption molecular carrier vitamin E-PEG 1000.
  • Oral formulations may incorporate suitable colorants, sweeteners, flavoring agents, and preservatives.
  • the compound of the formula (I) or a pharmaceutically acceptable salt of the present invention is administered to a warm-blooded animal at a unit dose of 0.01 to 100 mg/kg.
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt of the present invention can be used alone or in combination with radiotherapy, chemotherapy, immunotherapy, tumor vaccine, and tumor which are conventionally used in clinical practice.
  • Combination therapy with one or more methods of viral, RNAi, cancer adjuvant therapy, and bone marrow transplantation and stem cell transplantation including but not limited to the following anti-tumor drugs and treatments:
  • alkylating agents such as cisplatin, cisplatin, oxaliplatin, chlorambucil, carbophosphoramide, nitrogen mustard, melphalan, temozolomide, busulfan, nitrosourea.
  • anti-tumor antibiotics such as doxorubicin, bleomycin, doxorubicin, daunorubicin, epirubicin, idarubicin, mitomycin C, actinomycin, genus Anti-mitotic drugs such as vincristine, vinblastine, vindesine, vinorelbine, paclitaxel, taxotere, Polo kinase inhibitors.
  • Antimetabolites and antifolates such as fluoropyrimidine, rametamine, cytarabine, azacitidine, decitabine, trebuta, hydroxyurea, IDH1/IDH2 mutant inhibitors.
  • Topoisomerase inhibitors such as epipodophyllotoxin, camptothecin, and irinotecan.
  • Cell growth inhibitors such as antiestrogens/antiandrogens, such as tamoxifen, fulvestrant, toremifene, raloxifene, ranoxifene, oxyxid, bicalutamide , flutamide, nilutamide, cyproterone acetate;
  • LHRH antagonists or LHRH agonists such as goserelin, leuprolide, and buserelin, progestogens such as megestrol acetate;
  • Aromatase inhibitors such as anastrozole, letrozole, vorozole, exemestane, 5a-reductase inhibitors such as finasteride.
  • Anti-invasive agents such as c-Src kinase family inhibitors, metalloproteinase inhibitors, inhibitors of urokinase plasminogen activator receptor function or heparanase-like antibodies.
  • inhibitors of growth function such as growth factor antibodies and growth factor receptor antibodies such as anti-HER2 antibody trastuzumab, anti-EGFR antibody panitumumab, anti-EGFR antibody cetuximab, etc.; Including other tyrosine kinase inhibitors and inhibitors of serine/threonine kinases such as Ras/Raf signaling inhibitors, cell signaling inhibitors of MEK and/or AKT kinase, c-kit inhibitors, abl kinase inhibitors , PI3 kinase inhibitors, JAKs and STAT3 inhibitors, FLT3 kinase inhibitors, CSF-1R kinase inhibitors, IGF receptor kinase inhibitors, Aurora kinase inhibitors, NTRKA/B/C kinase inhibitors.
  • growth factor antibodies and growth factor receptor antibodies such as anti-HER2 antibody trastuzumab, anti-EGFR antibody panitumumab, anti-EGFR antibody
  • An anti-angiogenic agent such as bevacizumab which inhibits the action of vascular endothelial growth factor and a VEGF receptor tyrosine kinase inhibitor.
  • HDACi histone deacetylase inhibitors
  • DNMTi DNA methyltransferase inhibitors
  • histone acetyltransferase inhibitors histone demethylases Inhibitors
  • histone methyltransferase inhibitors and the like.
  • PARPi Poly ADP-ribose polymerase inhibitors
  • Tumor immunotherapy includes any in vitro and in vivo methods that increase the immunogenicity of a patient's tumor cells.
  • cytokine IL-2, IL-4 or GM-CSF for transfection; methods for reducing T cell ineffectiveness such as anti-PD-1/PD-L mAb; transfected immune cells such as cytokine transfected trees Method of squamous cell; method of cytokine transfected tumor cell line; reduction of immunosuppressive cells such as regulatory T cells, myeloid suppressor cells, or dendrites expressing guanamine 2,3-deoxygenase Functional methods of cells; methods of agonists that increase the activity of immune cells, such as STING, and cancer vaccines composed of tumor-associated antigenic proteins or peptides.
  • Tumor gene therapy such as CRISPR-Cas 9, RNAi, gene transduction.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • the NMR was measured by a (Bruker AVANCE-400) nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), and the internal standard was Tetramethylsilane (TMS).
  • DMSO-d6 dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS Tetramethylsilane
  • the MS was measured by a liquid chromatography mass spectrometer (Thermo, Ultimate 3000/MSQ).
  • the HPLC was measured using a high pressure liquid chromatograph (Agilent 1260 Infinity, Gemini C18 250 x 4.6 mm, 5u column).
  • the silica gel plate HSGF245 used for thin layer chromatography has a specification of 0.15 mm to 0.2 mm, and the specification for separation and purification of thin layer chromatography is 0.9 mm to 1.0 mm (Yantai Yellow Sea).
  • the known starting materials of the present invention can be synthesized by or according to methods known in the art, or purchased from Shanghai Darui Fine Chemicals Co., Ltd., Shanghai Titan Technology Co., Ltd., Shanghai Runjie Chemical Reagent Co., Ltd., TCI, Aldrich Chemical Company.
  • the experimental methods in the examples which do not specify the specific conditions are usually carried out according to conventional conditions or according to the conditions recommended by the raw material or commodity manufacturer. Reagents without specific source are routine reagents purchased from the market.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • 2,4-Dichloropyrimidine (7.15 g, 0.048 mol) was dissolved in DMF (80 ml), HOBT (1.0 g, 8 mmol) and potassium carbonate (11 g, 0.08 mol) were added at room temperature, and the mixture was stirred at room temperature for 15 minutes.
  • the hydrazine (4.68 g, 0.04 mol) (dissolved in DMF) was slowly added dropwise to the reaction system, and the reaction was carried out by heating at 85 ° C for 6 hours, and the reaction was confirmed by TLC.
  • the mixture was poured into water (200 ml), EtOAc (EtOAc m. Purification by column chromatography (eluent: petroleum ether / ethyl acetate) afforded 5.3 g of 1-(2-chloro-pyrimidin-4-yl)-1H-indole as a yellow solid.
  • Step 2 Preparation of (3-nitro-phenyl)-carbamic acid tert-butyl ester
  • Step 3 Preparation of (3-amino-phenyl)-carbamic acid tert-butyl ester
  • Step 4 Preparation of [3-(3-nitro-benzoylamino)-phenyl]-carbamic acid tert-butyl ester
  • Step 5 Preparation of [3-(3-Amino-benzoylamino)-phenyl]-carbamic acid tert-butyl ester
  • Step 6 Preparation of (E)- ⁇ 3-[3-(4-dimethylamino-but-2-enoylamino)-benzoylamino]-phenyl ⁇ -carbamic acid tert-butyl ester
  • Trans-4-dimethylamino croton hydrochloride (6.1 g, 0.0367 mol) was added to a 250 ml three-necked flask under a nitrogen atmosphere, and the nitrogen balloon was ventilated three times.
  • Ultra dry (anhydrous) THF 150 ml
  • 6 drops of DMF were added by syringe and cooled to 0-5 ° C in an ice water bath.
  • Oxalyl chloride (3.9 g, 0.0306 mol) was slowly added dropwise to the system, and the mixture was added dropwise, and the ice water bath was removed, and the mixture was warmed to room temperature for 2 hours.
  • EtOAc EtOAc
  • EtOAc ⁇ 3-[3-(4-Dimethylamino-but-2-enoylamino)-benzoylamino]-phenyl ⁇ -carbamic acid tert-butyl ester.
  • Step 7 Preparation of (E)-N-(3-amino-phenyl)-3-(4-dimethylamino-but-2-enoylamino)-benzamide
  • Step 8 (E)-3-(4-Dimethylamino-but-2-enoylamino)-N-[3-(4-indol-1-yl-pyrimidin-2-ylamino)-benzene Preparation of bis-benzamide
  • Example 2 Same as the preparation method in Example 1, except that 4-methoxy hydrazine (TCI) was used instead of the hydrazine in the first step of Example 1, to obtain (E)-3-(4-dimethylamino-butyl- 2-enoylamino)-N- ⁇ 3-[4-(4-methoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -benzamide.
  • TCI 4-methoxy hydrazine
  • Methyl (E)-4-bromo-2-butenoate (3 g, 0.0168 mol) was dissolved in 30 ml of THF under nitrogen atmosphere, cooled to 0-5 ° C in an ice water bath, and 10 ml of aqueous lithium hydroxide solution was slowly added dropwise. (960 mg of lithium hydroxide), after completion of the dropwise addition, the temperature was maintained at 0 to 5 ° C for 3 hours. The reaction was completely detected by TLC, 50 ml of ice water was added, and petroleum ether was extracted (100 ml ⁇ 2), and the organic phase was discarded.
  • the aqueous phase was cooled to about 0 ° C, the pH was adjusted to about 1 with concentrated hydrochloric acid, extracted with dichloromethane (70 ml ⁇ 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 1.35 g of 4-bromo croton as a solid. acid.
  • Step 3 Preparation of (E)- ⁇ 3-[3-(4-bromo-but-2-enoylamino)-benzoylamino]-phenyl ⁇ -carbamic acid tert-butyl ester
  • Step 4 Preparation of (E)- ⁇ 3-[3-(4-morpholin-4-yl-but-2-enoylamino)-benzoylamino]-phenyl ⁇ -carbamic acid tert-butyl ester
  • Step 6 (E)-N- ⁇ 3-[4-(4-Methoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -3-(4-morpholine- Preparation of 4-yl-but-2-enoylamino)-benzamide
  • Step 3 (E)-4-(4-Dimethylamino-but-2-enoylamino)-N-[3-(4- ⁇ 4-[2-(6-methyl-pyridine-3- Preparation of methoxy)-ethoxy]-indol-1-yl ⁇ -pyrimidin-2-ylamino)-phenyl]-benzamide
  • Step 2 (E)-3-(4-Dimethylamino-but-2-enoylamino)-N- ⁇ 3-[4-(4-isopropoxy-indol-1-yl)- Preparation of pyrimidin-2-ylamino]-phenyl ⁇ -benzamide
  • Step 4 (E)-N- ⁇ 3-[4-(4-Difluoromethoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -3-(4-di Preparation of methylamino-but-2-enoylamino)-benzamide
  • Example 1 Same as the preparation method of Example 1, except that 4-difluoromethoxy-1H-indole was used instead of the hydrazine in the first step of Example 1, and p-nitrobenzoic acid (TCI) was used instead of the first step in Example 1.
  • TCI p-nitrobenzoic acid
  • M-nitrobenzoic acid to give (E)-N- ⁇ 3-[4-(4-difluoromethoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -3 -(4-Dimethylamino-but-2-enoylamino)-benzamide.
  • Example 2 The same procedure as in Example 1, except that 4-methoxyindole (Dari) was used instead of the hydrazine in Step 1 of Example 1, and 4-methoxy-3-nitroaniline was used instead of Example 1.
  • m-Nitroaniline in 2 gives (E)-3-(4-dimethylamino-but-2-enoylamino)-N- ⁇ 2-methoxy-5-[4-(4-A Oxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -benzamide.
  • Example 1 Same as the preparation method of Example 1, except that 4-methoxy hydrazine (Dare) was used instead of the hydrazine in the first step of Example 1, and 4-chloro-3-nitrobenzoic acid was used instead of Example 1 Step 4.
  • m-Nitrobenzoic acid to give (E)-4-chloro-3-(4-dimethylamino-but-2-enoylamino)-N- ⁇ 3-[4-(4-methoxy -Indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -benzamide.
  • 3-indolecarboxylic acid (30 g, 0.186 mol) was stirred at room temperature in 500 ml of dichloromethane and was not completely dissolved. 0.5 ml of DMF was added, and then oxalyl chloride (71.0 g, 0.56 mol) was added dropwise thereto at room temperature. After 30 minutes, the addition was completed, and the reaction was continued at room temperature for 2 hours. The reaction was completed by EtOAc (EtOAc:EtOAc) The product was used in the next step without purification.
  • 1H-indole-3-carbonyl chloride (0.186 mol, theoretical yield) obtained in the step 1 was added to 500 ml of DCM, stirred at room temperature for 30 minutes, and was not completely dissolved, and was a cloudy dispersion system.
  • 350 ml of ammonia water and 200 ml of DCM were added to a 2 L three-necked flask and stirred vigorously.
  • the dichloromethane turbid dispersion of 1H-indole-3-carbonyl chloride was slowly added dropwise to a 2 L three-necked flask at room temperature, and after 20 minutes, the reaction was continued at room temperature for 1 hour.
  • Step 3 (E)-1-(2- ⁇ 3-[3-(4-Dimethylamino-but-2-enoylamino)-benzoylamino]-phenylamino ⁇ -pyrimidine-4- Preparation of -1H-indole-3-carboxamide
  • Step 2 (E)-3-(4-Dimethylamino-but-2-enoylamino)-N- ⁇ 3-[5-fluoro-4-(4-methoxy-oxime-1- Of p-pyrimidin-2-ylamino]-phenyl ⁇ -benzamide
  • the reaction mixture was cooled to room temperature, 20 ml of methyl tert-butyl ether was added, and the solid was washed with a small amount of methyl t-butyl ether.
  • the obtained solid was dissolved in dichloromethane/methanol (50 ml/5 ml), adjusted to pH 9-10 by adding 20 ml of 0.2 mol/L dilute aqueous sodium hydroxide solution, extracted with dichloromethane (30 ml ⁇ 2), and the organic phase was saturated with NaCl solution. It was washed twice, dried over anhydrous sodium sulfate, filtered, evaporated, evaporated, evaporated.
  • Example 22 The preparation method of Example 22 was carried out except that 2,4-dichloro-5-fluoropyrimidine in Step 1 of Example 22 was replaced with 2,4,5-trichloropyrimidine (Dari) to obtain (E)-N. - ⁇ 3-[5-Chloro-4-(4-methoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -3-(4-dimethylamino-butyl- 2-enoylamino)-benzamide.
  • Example 22 The same procedure as in Example 22 was carried out except that 2,4-dichloro-5-methylpyrimidine (Dari) was used instead of 2,4-dichloro-5-fluoropyrimidine in Step 1 of Example 22 to give (E )-3-(4-Dimethylamino-but-2-enoylamino)-N- ⁇ 3-[4-(4-methoxy-indol-1-yl)-5-methyl-pyrimidine 2-ylamino]-phenyl ⁇ -benzamide.
  • Dari 2,4-dichloro-5-methylpyrimidine
  • Step 1 of Example 22 to give (E )-3-(4-Dimethylamino-but-2-enoylamino)-N- ⁇ 3-[4-(4-methoxy-indol-1-yl)-5-methyl-pyrimidine 2-ylamino]-phenyl ⁇ -benzamide.
  • Example 22 Same as the preparation method of Example 22 except that 4-isopropoxy hydrazine (prepared in Example 7) was used instead of 4-methoxyindole in Step 1 of Example 22, and 2,4,5-three was used. Chloropyrimidine (Dari) is substituted for the 2,4-dichloro-5-fluoropyrimidine in Step 1 of Example 22 to give (E)-N- ⁇ 3-[5-chloro-4-(4-isopropoxy) -Indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -3-(4-dimethylamino-but-2-enoylamino)-benzamide.
  • 4-isopropoxy hydrazine prepared in Example 7
  • 2,4,5-three was used.
  • Chloropyrimidine (Dari) is substituted for the 2,4-dichloro-5-fluoropyrimidine in Step 1 of Example 22 to give (E)-N- ⁇ 3-[5-chloro-4-(4
  • Example 22 The same procedure as in Example 22 was carried out except that 1H-indole-3-carboxamide (prepared in Step 2 of Example 21) was used instead of 4-methoxyindole in Step 1 of Example 22 to give (E)-1. -(2- ⁇ 3-[3-(4-Dimethylamino-but-2-enoylamino)-benzoylamino]-phenylamino ⁇ -5-fluoro-pyrimidin-4-yl)-1H- Indole-3-carboxamide.
  • the acid salt replaces the trans-4-dimethylamino crotonate salt of the first step of Example 1, to give 3-(4-dimethylamino-butyrylamino)-N- ⁇ 3-[4-( 4-methoxy-indol-1-yl)-pyrimidin-2-ylamino]-phenyl ⁇ -benzamide.
  • MOLM13 is a human acute myeloid leukemia cell line with positive expression of CDK7 and cell line derived from DMSZ. The cells were cultured in suspension with RPMI 1640 (Gibco) plus 10% fetal bovine serum (Gibco), 1% double antibody, and 2 mM glutamine medium.
  • the MOLM-13 suspension cells in logarithmic growth phase were collected by centrifugation (1700 rpm, 3 minutes), the supernatant was discarded, and the cells were counted.
  • the cell concentration was 2 x 10 5 cells per ml in RPMI1640 medium, inoculated into 96-well plates (Corning), 100 ⁇ l per well, and cultured overnight at 37 ° C, 5% CO 2 .
  • the final concentration of the organic solvent is not more than one thousandth, and the cells are further cultured for 3 to 6 days, and the MTT is measured.
  • the compound of the present invention and the control compounds THZ1 and THZ2 were each dissolved in DMSO (Sigma), and the purity of the compound was 98% or more.
  • the compound was stored at a concentration of 10 mM, stored at -20 ° C, and serially diluted 10 or 10 times before use.
  • the MTT detection reagent is the Dojindo CCK8 kit, and the enzyme labeling instrument is THERMO MULTISKAN FC instrument.
  • the CCK8 reagent was directly added to the drug-treated and solvent-controlled suspension cells MOLM-13, the final concentration of CCK8 was 10%, and the culture was continued for 1 to 4 hours.
  • the solvent control wells were dark yellow, the OD450nm light absorption value was measured (THERMO MULTISKAN) FC instrument), calculate the cell growth rate according to the following formula:
  • T drug treatment cell hole optical density value - blank control hole optical density value
  • T 0 cell hole optical density value before drug treatment - blank control hole optical density value
  • C solvent control cell hole optical density - blank control hole light Density value.
  • the drug concentration, i.e., IC50, at which 50 % inhibition of cell growth was calculated by GraphPad Prism7 software. The experiment was repeated 1-3 times and the data was subjected to biological statistical analysis.
  • Table 2 summarizes the results of the determination of the IC 50 concentration of the compound of the present invention for inhibiting the growth-inducing apoptosis of tumor cells MOLM-13 in vitro. The smaller the IC 50 value, the stronger the activity of the compound.
  • "*****” represents an IC 50 value of ⁇ 1 nM
  • "****” represents an IC 50 value in the range of 1 nM to ⁇ 10 nM
  • "***” represents an IC 50 value in the range of 10 nM to ⁇ 100 nM
  • **" represents an IC 50 value in the range of 100 nM to ⁇ 1000 nM
  • "*” represents an IC 50 value > 1000 nM
  • "-" represents no measurement.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (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)

Abstract

La présente invention concerne un composé de pyrimidine, son procédé de préparation et son utilisation médicale. En particulier, la présente invention concerne un composé de pyrimidine représenté par la formule générale (I), et son procédé de préparation, ainsi que l'utilisation de celui-ci en tant qu'inhibiteur de kinase cycline-dépendante 7 (CDK7), notamment l'utilisation dans la prévention et/ou le traitement de maladies humaines y compris des cancers. Les définitions de chaque groupe dans la formule générale (I) sont les mêmes que celles définies dans la description.
PCT/CN2019/073874 2018-02-13 2019-01-30 Composé de pyrimidine, son procédé de préparation et son utilisation médicale WO2019157959A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980005206.4A CN111247137A (zh) 2018-02-13 2019-01-30 一种嘧啶类化合物、其制备方法及其医药用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810150051.0 2018-02-13
CN201810150051 2018-02-13

Publications (1)

Publication Number Publication Date
WO2019157959A1 true WO2019157959A1 (fr) 2019-08-22

Family

ID=67619169

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/073874 WO2019157959A1 (fr) 2018-02-13 2019-01-30 Composé de pyrimidine, son procédé de préparation et son utilisation médicale

Country Status (3)

Country Link
CN (1) CN111247137A (fr)
TW (1) TW201934547A (fr)
WO (1) WO2019157959A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023227125A1 (fr) * 2022-05-26 2023-11-30 杭州德睿智药科技有限公司 Nouveau composé hétérocyclique fusionné en tant qu'inhibiteur de cdk et son utilisation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114133394B (zh) * 2020-08-12 2023-12-08 赛诺哈勃药业(成都)有限公司 一种选择性针对细胞周期依赖性激酶12活性的化合物、制备方法及医药用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014063068A1 (fr) * 2012-10-18 2014-04-24 Dana-Farber Cancer Institute, Inc. Inhibiteurs de cycline-dépendante kinase 7 (cdk7)
WO2015154039A2 (fr) * 2014-04-04 2015-10-08 Syros Pharmaceuticals, Inc. Inhibiteurs de la kinase cycline-dépendante 7 (cdk7)

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014063068A1 (fr) * 2012-10-18 2014-04-24 Dana-Farber Cancer Institute, Inc. Inhibiteurs de cycline-dépendante kinase 7 (cdk7)
WO2015154039A2 (fr) * 2014-04-04 2015-10-08 Syros Pharmaceuticals, Inc. Inhibiteurs de la kinase cycline-dépendante 7 (cdk7)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KWIATKOWSKI, NICHOLAS: "Targeting transcription regulation in cancer with a covalent CDK7 inhibitor", NATURE, vol. 511, no. 7511, 31 July 2014 (2014-07-31), pages 616 - 620, XP055193048 *
WANG, YUBAO: "CDK7-Dependent Transcriptional Addiction in Triple-Negative Breast Cancer", CELL ., vol. 163, no. 1, 24 September 2015 (2015-09-24), XP029280470, doi:10.1016/j.cell.2015.08.063 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023227125A1 (fr) * 2022-05-26 2023-11-30 杭州德睿智药科技有限公司 Nouveau composé hétérocyclique fusionné en tant qu'inhibiteur de cdk et son utilisation

Also Published As

Publication number Publication date
TW201934547A (zh) 2019-09-01
CN111247137A (zh) 2020-06-05

Similar Documents

Publication Publication Date Title
JP5670266B2 (ja) フラザノベンゾイミダゾール
WO2017101803A1 (fr) Nouveau double inhibiteur de egfr et de alk
TW200902010A (en) Anthranilamide inhibitors of aurora kinase
TW200808771A (en) Novel compounds II
JP2008524191A (ja) Cdk阻害剤としての4−(4−(イミダゾール−4−イル)ピリミジン−2−イルアミノ)ベンズアミド
TW200800900A (en) Novel pyridine derivatives and pyrimidine derivatives
CN111051300B (zh) 作为组蛋白脱乙酰基酶1和/或2(hdac1-2)的选择性抑制剂的新杂芳基酰胺衍生物
CN109661229B (zh) 用于治疗癌症的化合物和组合物
TW200528447A (en) Novel imidazole derivatives and their use as pharmaceutical agents
BRPI0911679B1 (pt) Composto aciltioureia ou um sal do mesmo, uso do dito composto para tratar câncer, bem como agente farmacêutico, agente antitumoral e composição farmacêutica compreendendo dito composto
JP2020511468A (ja) Mk2阻害剤の重水素化アナログおよびその使用
WO2012041524A1 (fr) Hydrazonopyrazolones comme inhibiteurs de la protéine tyrosine phosphatase
CA2601979A1 (fr) Pyrazine sulfonamides 2,3 substituees
JP2020512315A (ja) Mk2阻害剤として有用なヘテロアリール化合物
WO2018145621A1 (fr) Composé de quinoléine, procédé de préparation et utilisation médicale associés
BR112021014977A2 (pt) Derivados de acetamido como inibidores de dna polimerase teta
WO2017152874A1 (fr) Composé d'urée, son procédé de préparation et son utilisation médicale
WO2018184585A1 (fr) Composé pour l'inhibition de l'ido, son procédé de fabrication et son utilisation
WO2019154177A1 (fr) Composé de pyrimidine, son procédé de préparation et son utilisation médicale
Dou et al. Rational modification, synthesis and biological evaluation of 3, 4-dihydroquinoxalin-2 (1H)-one derivatives as potent and selective c-Jun N-terminal kinase 3 (JNK3) inhibitors
WO2019157959A1 (fr) Composé de pyrimidine, son procédé de préparation et son utilisation médicale
WO2018137639A1 (fr) Inhibiteur d'histone méthyltransférase ezh2, son procédé de préparation et son utilisation pharmaceutique
WO2019024876A1 (fr) Dérivé de formylpyridine ayant une activité inhibitrice de fgfr4, son procédé de préparation et son utilisation
WO2020172906A1 (fr) Inhibiteur de kinase pan-raf de type nouveau et son utilisation
CN115322158A (zh) 作为krasg12c蛋白抑制剂的取代喹唑啉类化合物

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19754917

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19754917

Country of ref document: EP

Kind code of ref document: A1