WO2020147842A1 - 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用 - Google Patents

吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用 Download PDF

Info

Publication number
WO2020147842A1
WO2020147842A1 PCT/CN2020/072792 CN2020072792W WO2020147842A1 WO 2020147842 A1 WO2020147842 A1 WO 2020147842A1 CN 2020072792 W CN2020072792 W CN 2020072792W WO 2020147842 A1 WO2020147842 A1 WO 2020147842A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
reaction
preparation
drug
acid
Prior art date
Application number
PCT/CN2020/072792
Other languages
English (en)
French (fr)
Inventor
魏霞蔚
陈新海
陈兆国
张丽
于衍新
周凯
胡伯羽
Original Assignee
南京明德新药研发有限公司
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 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to CN202080008916.5A priority Critical patent/CN113286594B/zh
Publication of WO2020147842A1 publication Critical patent/WO2020147842A1/zh

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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention relates to the application of a class of pyridopyrimidine compounds in the preparation of drugs for treating nasopharyngeal carcinoma. Specifically, it relates to the application of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicine for treating nasopharyngeal carcinoma.
  • the present invention relates to a signal pathway that plays a key role in tumor proliferation, invasion and metastasis and anti-apoptosis, that is, phosphatidylinositol 3-kinase (PI3K)-AKT-mTOR signal pathway of mammalian rapamycin protease.
  • PI3K phosphatidylinositol 3-kinase
  • PI3K vascular endothelial growth factor
  • HGF human growth factor
  • Ang1 angiogenic protein I
  • FGF fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • HGF human growth factor
  • Ang1 angiogenic protein I
  • RTK receptor tyrosine kinase
  • PIP3 binds to the signal protein AKT and PDK1 (phosphoinositide dependent kinase-1) containing the PH domain in the cell, and promotes the phosphorylation of Ser308 of the AKT protein by PDK1 to activate AKT.
  • Other PDK1 substrates also include PKC (protein kinase C), S6K (p70S6) and SGK (serum/glucocorticoid regulated kinases).
  • AKT also known as protein kinase B (PKB)
  • PKT protein kinase B
  • Activated AKT phosphorylates downstream factors such as a variety of enzymes, kinases and transcription factors, thereby regulating cell functions.
  • AKT phosphorylates target proteins through various downstream pathways to exert anti-apoptotic effects.
  • PTEN phosphatase and tensin homology deleted on chromosome 10
  • PTEN is a tumor suppressor gene, gene mutation or deletion occurs in a wide range of human tumors.
  • PTEN is a PIP3-phosphatase, contrary to the function of PI3K, it can convert PIP3 to PIP2 by dephosphorylation.
  • PTEN can reduce the activation of AKT and prevent all downstream signaling events regulated by AKT.
  • mTOR is relatively conservative in evolution.
  • mTOR phosphorylates the downstream target protein S40S ribosomal S6 Protein kinases, such as S6K1 and 4EBP1, regulate downstream protein translation.
  • mTOR binds to eIF3, phosphorylates S6K1, and then releases S6K1 from eIF3 to be activated, and further phosphorylates cell substrates, such as p70S6 to promote protein translation and expression.
  • 4EBP1 binds to the eukaryotic transcription initiation factor 4E and inhibits its activity. When mtor phosphorylates 4E-BP1, it activates and separates it from eif-4e to realize eukaryotic transcription.
  • mTORC2 can phosphorylate AKT, thereby up-regulating its kinase activity.
  • this signaling pathway is used in a variety of solid tumors, such as nasopharyngeal cancer, breast cancer, prostate cancer, lung cancer, colon cancer, pancreatic cancer, liver cancer, gastric cancer, colorectal cancer, kidney cancer, thyroid cancer, meningitis cancer and acute cancer. Chronic lymphocytic leukemia, Merkel cell tumor, etc. And it is closely related to treatment tolerance and poor prognosis. It can be seen that the development of fine molecular compounds to achieve inhibition of the PI3K/AKT/MTOR signaling pathway has a good development prospect.
  • the purpose of the present invention is to discover a double mTOR small molecule compound targeted drug, which has good activity and exhibits excellent effects and functions.
  • US20170281637 discloses compound AZD2014, which belongs to mTORC1&mTORC2 kinase inhibitor, and its structural formula is as follows:
  • the present invention provides the use of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicine for treating nasopharyngeal carcinoma,
  • R 1 is selected C 1-3 alkyl, said C 1-3 alkyl optionally substituted with 1, 2 or 3 R a;
  • R 2 is selected from C 1-3 alkyl, said C 1-3 alkyl optionally substituted with 1, 2 or 3 R b;
  • R 3 is selected from H and C 1-3 alkyl, the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R c ;
  • R 4 is selected from H, F, Cl, Br, I and C 1-3 alkyl, the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R d ;
  • R a, R b, R c and R d are each independently selected from H, F, Cl, Br and I.
  • the aforementioned R 1 is selected from CH 3 , CF 3 , CH 2 CH 3 , CF 2 CH 3 , CHFCH 2 F, and CF 2 CH 2 F, and other variables are as defined in the present invention.
  • R 1 is selected from CH 3 , and other variables are as defined in the present invention.
  • the aforementioned R 2 is selected from CH 3 , CF 3 , CH 2 CH 3 , CF 2 CH 3 , CHFCH 2 F, and CF 2 CH 2 F, and other variables are as defined in the present invention.
  • R 2 is selected from CH 3 , and other variables are as defined in the present invention.
  • R 3 is selected from CH 3 , CF 3 , CH 2 CH 3 , CF 2 CH 3 , CHFCH 2 F, and CF 2 CH 2 F, and other variables are as defined in the present invention.
  • R 3 is selected from CH 3 , and other variables are as defined in the present invention.
  • R 4 is independently selected from H, F, Cl, Br, I, CH 3 , CF 3 , CH 2 CH 3 , CF 2 CH 3 , CHFCH 2 F, and CF 2 CH 2 F , Other variables are as defined in the present invention.
  • R 4 is independently selected from H and F, and other variables are as defined in the present invention.
  • the present invention also provides the use of the compound represented by the following formula or a pharmaceutically acceptable salt thereof in the preparation of a medicine for treating nasopharyngeal cancer,
  • the compounds of the present invention have significant or unexpected mTOR kinase inhibitory activity.
  • the compound of the present invention has obvious proliferation inhibitory activity on MCF-7, N87 and OE-21 cells, and a certain proliferation inhibitory activity on HT-29 cells.
  • Compound 1 showed the same or even better pharmacokinetic properties as the reference compound.
  • the efficacy of the patented compound in the subcutaneous xenograft tumor model of human nasopharyngeal carcinoma C666-1 cells was evaluated in vivo. Compared with the solvent control group, the 30 mg/kg of the treatment group of Example 2 is equivalent to the reference compound AZD2014 15 mg/kg, and both exhibited significant tumor inhibition effects.
  • pharmaceutically acceptable salt refers to a salt of the compound of the present invention, which is prepared from a compound with specific substituents discovered in the present invention and a relatively non-toxic acid or base.
  • the base addition salt can be obtained by contacting the neutral form of the compound with a sufficient amount of base in a pure solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salt or similar salts.
  • the acid addition salt can be obtained by contacting the neutral form of the compound with a sufficient amount of acid in a pure solution or a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogen carbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and organic acid salts, the organic acids include such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid and methanesulfonic acid; also include salts of amino acids (such as arginine, etc.) , And salts of organic acids such as glucuronic acid. Certain specific compounds of the present invention contain basic and acidic
  • the pharmaceutically acceptable salt of the present invention can be synthesized from the parent compound containing an acid radical or a base by conventional chemical methods. Generally, such salts are prepared by reacting these compounds in free acid or base form with a stoichiometric amount of appropriate base or acid in water or an organic solvent or a mixture of both.
  • the compounds of the present invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, and diastereomers Conformers, (D)-isomers, (L)-isomers, and their racemic mixtures and other mixtures, such as enantiomers or diastereomeric enriched mixtures, all of these mixtures belong to Within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All these isomers and their mixtures are included in the scope of the present invention.
  • enantiomer or “optical isomer” refers to stereoisomers that are mirror images of each other.
  • cis-trans isomer or “geometric isomer” is caused by the inability to rotate freely because of double bonds or single bonds of ring-forming carbon atoms.
  • diastereomer refers to a stereoisomer in which a molecule has two or more chiral centers and the relationship between the molecules is not mirror images.
  • wedge-shaped solid line keys And wedge-shaped dashed key Represents the absolute configuration of a three-dimensional center, with a straight solid line key And straight dashed key Indicates the relative configuration of the three-dimensional center, using wavy lines Represents a wedge-shaped solid line key Or wedge-shaped dotted key Or use wavy lines Represents a straight solid line key And straight dashed key
  • the compound of the present invention may be specific.
  • tautomer or “tautomeric form” means that at room temperature, the isomers of different functional groups are in dynamic equilibrium and can be transformed into each other quickly. If tautomers are possible (such as in solution), the chemical equilibrium of tautomers can be reached.
  • proton tautomer also called prototropic tautomer
  • proton migration such as keto-enol isomerization and imine-ene Amine isomerization.
  • Valence isomers include some recombination of bonding electrons to carry out mutual transformation.
  • keto-enol tautomerization is the tautomerization between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • optically active (R)- and (S)-isomers and D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If you want to obtain an enantiomer of a compound of the present invention, it can be prepared by asymmetric synthesis or derivatization with chiral auxiliary agents, in which the resulting diastereomeric mixture is separated, and the auxiliary group is cleaved to provide pure The desired enantiomer.
  • the molecule when the molecule contains a basic functional group (such as an amino group) or an acidic functional group (such as a carboxyl group), it forms a diastereomeric salt with an appropriate optically active acid or base, and then passes through a conventional method known in the art The diastereoisomers are resolved, and then the pure enantiomers are recovered.
  • the separation of enantiomers and diastereomers is usually accomplished through the use of chromatography, which employs a chiral stationary phase and is optionally combined with chemical derivatization (for example, the formation of amino groups from amines). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms constituting the compound.
  • compounds can be labeled with radioisotopes, such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
  • radioisotopes such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
  • heavy hydrogen can be used to replace hydrogen to form deuterated drugs.
  • the bond formed by deuterium and carbon is stronger than the bond formed by ordinary hydrogen and carbon.
  • deuterated drugs have reduced toxic side effects and increased drug stability. , Enhance the efficacy, extend the biological half-life of drugs and other advantages.
  • the conversion of all isotopic compositions of the compounds of the present invention, whether radioactive or not, is included in the scope of the present invention.
  • pharmaceutically acceptable carrier refers to any preparation or carrier medium that can deliver an effective amount of the active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects on the host or patient.
  • Representative carriers include water, oil, Vegetables and minerals, cream base, lotion base, ointment base, etc. These bases include suspending agents, tackifiers, penetration enhancers and the like. Their formulations are well known to those skilled in the field of cosmetics or topical medicine.
  • substituted means that any one or more hydrogen atoms on a specific atom are replaced by substituents, and may include deuterium and hydrogen variants, as long as the valence of the specific atom is normal and the substituted compound is stable of.
  • oxygen it means that two hydrogen atoms are replaced. Oxygen substitution does not occur on aromatic groups.
  • optionally substituted means that it can be substituted or unsubstituted. Unless otherwise specified, the type and number of substituents can be arbitrary on the basis that they can be chemically realized.
  • any variable such as R
  • its definition in each case is independent.
  • the group can optionally be substituted with up to two Rs, and R has independent options in each case.
  • combinations of substituents and/or variants thereof are only permitted if such combinations result in stable compounds.
  • linking group When the number of a linking group is 0, such as -(CRR) 0 -, it means that the linking group is a single bond.
  • alkyl is used to indicate a linear or branched saturated hydrocarbon group, which can be mono-substituted (such as -CH 2 F) or multi-substituted (such as -CF 3 ), and can be monovalent (such as Methyl), divalent (such as methylene) or multivalent (such as methine).
  • alkyl groups examples include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl) , T-butyl), pentyl (eg, n-pentyl, isopentyl, neopentyl) and the like.
  • C 1-3 alkyl is used to indicate a linear or branched saturated hydrocarbon group composed of 1 to 3 carbon atoms.
  • the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups, etc.; it can be monovalent (such as methyl), divalent (such as methylene) or multivalent (such as methine) .
  • Examples of C 1-3 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
  • halogen or “halogen” by itself or as part of another substituent represents a fluorine, chlorine, bromine or iodine atom.
  • haloalkyl is intended to include monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, etc. Wait.
  • examples of haloalkyl include, but are not limited to: trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • C n-n+m or C n -C n+m includes any specific case of n to n+m carbons, for example, C 1-12 includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 , and C 12 , including any range from n to n+m, for example, C 1-12 includes C 1-3 , C 1-6 , C 1-9 , C 3-6 , C 3-9 , C 3-12 , C 6-9 , C 6-12 , and C 9-12, etc.; in the same way, from n to n +m means the number of atoms in the ring is n to n+m, for example, 3-12 membered ring includes 3-membered ring, 4-membered ring, 5-membered ring, 6-membered ring, 7-membered ring, 8-membered ring, 9-membered
  • the solvent used in the present invention is commercially available.
  • aq stands for water
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • EDC stands for N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride
  • m-CPBA stands for 3-chloroperoxybenzoic acid
  • eq stands for equivalent, equivalent amount
  • CDI stands for Carbonyl diimidazole
  • DCM stands for dichloromethane
  • PE stands for petroleum ether
  • DIAD diisopropyl azodicarboxylate
  • DMF stands for N,N-dimethylformamide
  • DMSO stands for dimethyl sulfoxide
  • EtOAc stands for ethyl acetate Esters
  • EtOH stands for ethanol
  • MeOH stands for methanol
  • CBz stands for benzyloxycarbonyl, which is an
  • the compounds of the present invention are based on conventional naming principles in the field or The software is named, and the commercially available compounds use the supplier catalog name.
  • Dissolve compound 1a (23.0g, 328mmol, 24.5mL, 1.0eq) and zinc diiodide (5.20g, 16.4mmol, 0.05eq) in 200ml of dichloromethane, reduce the internal temperature to 0°C, add trimethyl Silica cyanide (39.1g, 393mmol, 49mL, 1.2eq), the reaction solution was reacted at 25°C for 18 hours, the reaction was complete, the reaction solution was concentrated and added 50ml of acetonitrile and 50ml of aqueous hydrochloric acid (1N), stirred at 25°C for 5 minutes .
  • 1b (16.0g, 164mmol, 1eq) was added to a tetrahydrofuran solution (300mL) containing lithium tetrahydroaluminum (9.38g, 247mmol, 1.5eq), and reacted at 20°C for 18 hours. After the reaction was complete, water (9.38 mL), 15% sodium hydroxide (9.38 mL) and water (28.1 mL) were sequentially added to the reaction solution, stirred for 15 minutes, filtered, and concentrated to obtain 1c.
  • triphenylmethylphosphonium bromide 25.9g, 72.6mmol, 1.6eq
  • potassium tert-butoxide 1M, 81.7mL, 1.8eq
  • the reaction solution was The reaction was carried out at 20°C for 3 hours, and 3a (8.0 g, 45.4 mmol, 1 eq) was added to the reaction solution and reacted for 18 hours.
  • the labeled mTOR kinase was stably expressed in HEK-293 cells.
  • Ligand beads were blocked with excess biotin and washed with buffer (1% bovine serum albumin, 0.05% Tween 20 ml, 1 ml dithiothreitol) to wash away unbound ligands and non-specific binding ⁇ ligand;
  • buffer 1% bovine serum albumin, 0.05% Tween 20 ml, 1 ml dithiothreitol
  • test compound is dissolved in dimethyl sulfoxide
  • the 10 affinity beads were resuspended in buffer (1x PBS, 0.05% Tween 20, 0.5 ⁇ m non-biotin affinity ligand) and incubated at room temperature for 30 minutes.
  • the compound of the present invention has significant or unexpected mTOR kinase inhibitory activity.
  • Experimental purpose to detect the inhibitory activity of the test compound on cell proliferation.
  • Luciferase in Cell-Titer-Glo reagent uses luciferin, oxygen and ATP as reaction substrates to produce oxyluciferin and release energy in the form of light. Since the luciferase reaction requires ATP, the total amount of light produced by the reaction is directly proportional to the total amount of ATP that reflects cell viability.
  • MCF-7 cell line ATCC-CRL-22
  • HT-29 cell line ATCC-HTB-38
  • OE21 ECACC-96062201
  • NCI-N87 cell line ATCC-CRL-5822
  • Cell culture medium (RPMI 1640 medium (Invitrogen#1868546; 10% serum Invitrogen#1804958; L-glutamine 1 spin, Invitrogen#1830863; double antibody Hyclone#J170012))
  • the compound stock solution is 10 mM, and the compound is diluted with DMSO to make the initial concentration 4 mM. Add the compound to the compound mother liquid plate, 9 ⁇ L per well.
  • Test result The test result is shown in Table 2
  • the compound of the present invention has obvious proliferation inhibitory activity on MCF-7, N87 and OE-21 cells, and has a certain proliferation inhibitory activity on HT-29 cells.
  • test compound was mixed with 5% DMSO/95% 10% polyoxyethylene castor oil (Cremophor EL), vortexed and sonicated to prepare an approximately clear solution of 1 mg/mL, which was filtered with a microporous membrane for use.
  • Cremophor EL polyoxyethylene castor oil
  • the test compound was mixed with 1% Tween 80, 9% polyethylene glycol 400, 90% aqueous solution, vortexed and sonicated to prepare a 1 mg/mL approximately clear solution, which was filtered through a microporous membrane for use.
  • mice Female BALB/c nude mice, 6-8 weeks old, weighing 18-22 grams; Supplier: Nanda model animals
  • Human nasopharyngeal carcinoma C666-1 cells were cultured in a monolayer in vitro, and the culture conditions were DMEM medium with 10% fetal bovine serum, 100U/mL penicillin, 100U/mL streptomycin, 37°C, 5% CO 2 culture. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90%, the cells are collected, counted, and seeded.
  • test compound was formulated into clear solutions of 5 mg/mL and 5 mg/mL, and the solvent was 5% DMSO + 30% PEG400 + 65% water
  • the experimental index is to investigate whether the tumor growth is inhibited, delayed or cured.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI (%) reflects the tumor growth inhibition rate.
  • TGI(%) [1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group)/(Average tumor volume at the end of treatment in the solvent control group- The average tumor volume at the start of treatment in the solvent control group)] ⁇ 100%.
  • T/C (%) average tumor volume at the end of a certain treatment group/average tumor volume at the end of the solvent control group ⁇ 100%.
  • the body weight of experimental animals is used as a reference index for indirect determination of drug toxicity.
  • the body weight of the mice has a tendency to decrease, one in the solvent group died early, and the stocking density was suspected to be too high, and no abnormalities were found in the anatomy; in the example 2 group, one died on the 28th day with too low body weight and too much toxicity. No abnormalities were found during anatomy. (One in the AZD2014 group and the Example 2 group weighed less than 17g)
  • c.p value is calculated based on tumor volume.

Landscapes

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

Abstract

本发明公开了一类吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用。具体公开了式(I)所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用。

Description

吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用
本申请主张如下优先权:
CN201910049704.0,申请日2019年1月18日。
技术领域
本发明涉及一类吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用。具体涉及式(I)所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用。
技术背景
肿瘤,特别是恶性肿瘤是目前危害人类健康的最严重一大类疾病之一,随着科技的进步和人们对肿瘤治疗研究的越来越深入,在肿瘤的发生,发展机制上和肿瘤的治疗方面取得了飞速的进展。很多新的机制和生物标着物被发现。本发明涉及到一条对肿瘤的增殖、侵润转移和抗凋亡起关键作用的信号通路,即磷脂酰肌醇3激酶(PI3K)-AKT-哺乳动物雷帕霉素蛋白酶mTOR信号通路。
PI3K的活化很大程度上参与到靠近其质膜内侧的底物。多种生长因子和信号传导复合物,包括成纤维细胞生长因子(FGF)、血管内皮生长因子(VEGF)、人生长因子(HGF)、血管位蛋白I(Ang1)和胰岛素都能启始PI3K的激活过程。这些因子激活受体酪氨酸激酶(RTK),从而引起自磷酸化。PI3K激活的结果是在质膜上产生第二信使PIP3,PIP3与细胞内含有PH结构域的信号蛋白AKT和PDK1(phosphoinositide dependent kinase-1)结合,促使PDK1磷酸化AKT蛋白的Ser308导致AKT活化。其它PDK1的底物还包括PKC(蛋白激酶C)、S6K(p70S6)和SGK(serum/glucocorticoid regulated kinases)。AKT,亦称为蛋白激酶B(PKB),是PI3K下游主要的效应物。活化的AKT通过磷酸化多种酶、激酶和转录因子等下游因子,进而调节细胞的功能。AKT通过下游多种途径对靶蛋白进行磷酸化而发挥抗凋亡作用。PTEN(phosphatase and tensin homology deleted on chromosome 10),是一种抑癌基因,在广泛的人类肿瘤中发生基因突变或缺失。PTEN是一个PIP3-磷酸酶,与PI3K的功能相反,它可以通过去磷酸化将PIP3转变为PIP2。PTEN可减少AKT的活化而阻止所有由AKT调控的下游信号传导事件。mTOR作为AKT下游底物,进化上相对保守,可整合营养、能量及生长因子的多种信号,参与基因的转录、蛋白质翻译、核糖体合成和细胞凋亡等生物过程,在细胞生长中发挥极为重要的作用。其有两种高度同源的复合物,Tor与KOG01结合形成mTORC1,mTOR与AVO1/AVO2/AVO3/和LST8形成对雷帕霉素不敏感的mTORC2.mTOR通过磷酸化下游靶蛋白S40S核糖体S6蛋白激酶,比如S6K1和4EBP1来调节下游蛋白翻译。mTOR与eIF3结合,磷酸化S6K1,使S6K1从eIF3上释放而被活化,进一步磷酸化细胞底物,如p70S6促进蛋白质翻译及表达。4EBP1与真核转录启动因子4E结合并抑制其活性,当mtor磷酸化4E-BP1后,使其活化与eif-4e分离,实现真核细胞转录。mTORC2能磷酸化AKT,从而上调其激酶活性。
由上可见,PI3K/AKT/mTOR信号通路上游出现任何的突变或过度表达,都会导致下游一系列级联反应,最终导致肿瘤的发生,发展和转移。而mTOR处于信号通路的枢纽,对mTORC1和mTORC2的抑制能很好的阻断信号的传递,从而达到控制肿瘤的发展。
研究发现,此信号通路在多种实体瘤,如鼻咽癌、乳腺癌、前列腺癌、肺癌、结肠癌、胰腺癌、肝癌、胃癌、结直肠癌、肾癌、甲状腺癌、脑膜炎癌和急慢性淋巴细胞白血病,梅克尔细胞瘤等。并且与 治疗耐受和不良预后紧密相关。由此可见,通过开发细分子化合物实现对PI3K/AKT/MTOR的信号通路的抑制,具有良好的开发前景。
本发明旨在发现一种双mTOR小分子化合物靶向药物,此类化合物具有良好的活性,并表现出了优异的效果和作用。
US20170281637公开化合物AZD2014,属于mTORC1&mTORC2激酶抑制剂,其结构式如下所示:
Figure PCTCN2020072792-appb-000001
发明内容
本发明提供式(I)所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,
Figure PCTCN2020072792-appb-000002
其中
R 1选自C 1-3烷基,所述C 1-3烷基任选被1、2或3个R a取代;
R 2选自C 1-3烷基,所述C 1-3烷基任选被1、2或3个R b取代;
T选自O、S、S(=O) 2、-N(R 3)-和-C(R 4) 2-;
R 3选自H和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R c取代;
R 4选自H、F、Cl、Br、I和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R d取代;
R a、R b、R c和R d分别独立地选自H、F、Cl、Br和I。
本发明的一些方案中,上述R 1选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F,其它变量如本发明所定义。
本发明的一些方案中,上述R 1选自CH 3,其它变量如本发明所定义。
本发明的一些方案中,上述R 2选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F,其它变量如本发明所定义。
本发明的一些方案中,上述R 2选自CH 3,其它变量如本发明所定义。
本发明的一些方案中,上述R 3选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F,其它变 量如本发明所定义。
本发明的一些方案中,上述R 3选自CH 3,其它变量如本发明所定义。
本发明的一些方案中,上述R 4分别独立地选自H、F、Cl、Br、I、CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F,其它变量如本发明所定义。
本发明的一些方案中,上述R 4分别独立地选自H、F,其它变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020072792-appb-000003
选自
Figure PCTCN2020072792-appb-000004
Figure PCTCN2020072792-appb-000005
其它变量如本发明所定义。
本发明还有一些方案是上述变量任意组合而来。
本发明还提供下式所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,
Figure PCTCN2020072792-appb-000006
技术效果:
本发明化合物具有显著甚至意料不到的mTOR激酶抑制活性。本发明化合物对MCF-7,N87和OE-21细胞增殖抑制活性明显,对HT-29细胞具有一定的增殖抑制活性。化合物1展现了与参比化合物相同甚至更优的药代动力学性质。在本实验评价了本专利化合物在人鼻咽癌C666-1细胞皮下异种移植瘤模型中的体内药效。与溶剂对照组相比,治疗组实施例2 30mg/kg与参照化合物AZD2014 15mg/kg药效相当,都展现了显著的抑瘤作用。
相关定义
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,用楔形实线键
Figure PCTCN2020072792-appb-000007
和楔形虚线键
Figure PCTCN2020072792-appb-000008
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2020072792-appb-000009
和直形虚线键
Figure PCTCN2020072792-appb-000010
表示立体中心的相对构型,用波浪线
Figure PCTCN2020072792-appb-000011
表示楔形实线键
Figure PCTCN2020072792-appb-000012
或楔形虚线键
Figure PCTCN2020072792-appb-000013
或用波浪线
Figure PCTCN2020072792-appb-000014
表示直形实线键
Figure PCTCN2020072792-appb-000015
和直形虚线键
Figure PCTCN2020072792-appb-000016
本发明的化合物可以存在特定的。除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来 制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。
“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
除非另有规定,术语“烷基”用于表示直链或支链的饱和烃基,可以是单取代(如-CH 2F)或多取代的(如-CF 3),可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。烷基的例子包括甲基(Me),乙基(Et),丙基(如,n-丙基和异丙基),丁基(如,n-丁基,异丁基,s-丁基,t-丁基),戊基(如,n-戊基,异戊基,新戊基)等。
除非另有规定,术语“C 1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述C 1-3烷基包括C 1-2和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C 1-3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C 1-C 4)烷基”意在包括但 不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。除非另有规定,卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。
除非另有规定,C n-n+m或C n-C n+m包括n至n+m个碳的任何一种具体情况,例如C 1-12包括C 1、C 2、C 3、C 4、C 5、C 6、C 7、C 8、C 9、C 10、C 11、和C 12,也包括n至n+m中的任何一个范围,例如C 1-12包括C 1-3、C 1-6、C 1-9、C 3-6、C 3-9、C 3-12、C 6-9、C 6-12、和C 9-12等;同理,n元至n+m元表示环上原子数为n至n+m个,例如3-12元环包括3元环、4元环、5元环、6元环、7元环、8元环、9元环、10元环、11元环、和12元环,也包括n至n+m中的任何一个范围,例如3-12元环包括3-6元环、3-9元环、5-6元环、5-7元环、6-7元环、6-8元环、和6-10元环等。
本发明所使用的溶剂可经市售获得。
本发明各实施例涉及高效液相色谱分离的均采用中性分离。
本发明采用下述缩略词:aq代表水;HATU代表O-(7-氮杂苯并三唑-1-基)-N,N,N',N'-四甲基脲六氟磷酸盐;EDC代表N-(3-二甲基氨基丙基)-N'-乙基碳二亚胺盐酸盐;m-CPBA代表3-氯过氧苯甲酸;eq代表当量、等量;CDI代表羰基二咪唑;DCM代表二氯甲烷;PE代表石油醚;DIAD代表偶氮二羧酸二异丙酯;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;EtOH代表乙醇;MeOH代表甲醇;CBz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁基羰基是一种胺保护基团;HOAc代表乙酸;NaCNBH 3代表氰基硼氢化钠;r.t.代表室温;O/N代表过夜;THF代表四氢呋喃;Boc 2O代表二-叔丁基二碳酸酯;TFA代表三氟乙酸;DIPEA代表二异丙基乙基胺;SOCl 2代表氯化亚砜;CS 2代表二硫化碳;TsOH代表对甲苯磺酸;NFSI代表N-氟-N-(苯磺酰基)苯磺酰胺;n-Bu 4NF代表氟化四丁基铵;iPrOH代表2-丙醇;mp代表熔点;LDA代表二异丙基胺基锂;Pd(PPh 3) 4代表四(三苯基膦)钯;IV代表静脉注射;PO代表口服。
本发明化合物依据本领域常规命名原则或者
Figure PCTCN2020072792-appb-000017
软件命名,市售化合物采用供应商目录名称。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。
参考例1
Figure PCTCN2020072792-appb-000018
Figure PCTCN2020072792-appb-000019
第一步
将化合物1-1(20.0g,104mmol,1.00eq)和浓氨水(200mL,1.45mol,14.0eq)密闭于高压釜中,130℃下搅拌24小时,压力约为0.9MPa。将反应液浓缩,得到化合物1-2。
MS-ESI计算值[M+H] +173和175,实测值173和175。
1H NMR(400MHz,DMSO-d 6)δ:8.03(d,J=8.0Hz,1H),7.56(br s,2H),6.61(d,J=8.0Hz,1H)。
第二步
将化合物1-2(17.0g,98.5mmol,1.00eq)、氯化铵(10.5g,197mmol,2.00eq)、1-羟基苯并三唑(13.3g,98.5mmol,1.00eq)、1-(3-二甲胺丙基)-3-乙基碳二亚胺盐酸盐(18.9g,98.5mmol,1.00eq)和二异丙基乙胺(38.2g,296mmol,3.00eq)溶于N,N-二甲基甲酰胺(200.0mL)中。混合物在20℃下搅拌16小时。反应完成后,溶剂减压旋干,加水(200mL),乙酸乙酯萃取(200mL×3),合并的有机相用无水硫酸钠干燥,过滤,柱层析(1:1石油醚/乙酸乙酯,R f=0.4),得到化合物,乙酸乙酯(50mL)打浆十分钟,得到化合物1-3。
1H NMR(400MHz,DMSO-d 6)δ:7.96(d,J=8.0Hz,2H),7.62(br s,2H),7.40(br s,1H),6.61(d,J=8.0Hz,1H)。
第三步
将化合物1-3(8.00g,46.6mmol,1.00eq)和草酰氯(7.1g,56.0mmol,4.9mL,1.00eq)依次加入甲苯(200mL)中。混合物在110℃下搅拌15小时。冷却至室温,过滤,干燥。得到化合物1-4。
1H NMR(400MHz,DMSO-d 6)δ:8.24(d,J=8.0Hz,1H),7.30(d,J=8.0Hz,1H)。
第四步
将化合物1-4(6.00g,30.4mmol,1.00eq)和二异丙基乙胺(11.8g,91.1mmol,15.9mL,3.00eq)依次加入甲苯(100mL)中。混合物在70℃下搅拌半小时。冷却至室温,将三氯氧磷(14.0g,91.1mmol,8.5mL,3.00eq)滴入混合物中。混合物在100℃下搅拌2小时。冷却至室温,浓缩,柱层析(3:1石油醚/乙酸乙酯,Rf=0.4),得到化合物1-5。
1H NMR(400MHz,DMSO-d 6)δ:8.45(d,J=8.0Hz,1H),7.63(d,J=8.3Hz,1H)。
第五步
将化合物1-5(1.90g,8.10mmol,1.00eq)、(S)-2-甲基吗啡啉(819mg,8.10mmol,1.00eq)和二异丙基乙胺(2.09g,16.2mmol,2.83mL,2.00eq)溶于二氯甲烷(50mL)中,所得溶液在25℃反应2小时。反应完成后,浓缩,柱层析(3:1石油醚/乙酸乙酯),得到化合物1-6。
1H NMR(400MHz,DMSO-d 6)δ:8.47(d,J=8.8Hz,1H),7.55(d,J=8.8Hz,1H),4.71-4.72(m,1H),4.12-4.09(m,1H),3.92-3.91(m,1H),3.84-3.74(m,1H),3.73-3.64(m,2H),3.54-3.53(m,1H),1.46(d,J=6.8Hz,3H)。
第六步
将化合物1-6(1.2g,4.01mmol,1.00eq)、化合物1-7(1.15g,4.41mmol,1.10eq)、四三苯基膦钯(232mg,200μmol,0.05eq)和碳酸钾(1.66g,12.0mmol,3.00eq)溶于水(24mL)和1,4-二氧六环(120mL)中,在氮气保护下,60℃反应5小时,反应完成后,浓缩掉溶剂,加水(30mL)稀释后用乙酸乙酯萃取(50mL×2),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯)得化合物1h。
1H NMR(400MHz,DMSO-d 6)δ:8.71(s,1H),8.67(d,J=4.8Hz,1H),8.55(d,J=8.8Hz,1H),8.39(d,J=8.0Hz,1H),8.14(d,J=8.8Hz,1H),8.01(d,J=8.0Hz,1H),7.68(t,J=7.6Hz,1H),4.75(d,J=6.4Hz,1H),4.17-4.15(m,1H),3.94-3.92(m,1H),3.87-3.77(m,1H),3.72(s,2H),3.59-3.57(m,1H),2.86-2.84(m,3H),1.49(d,J=6.8Hz,3H)。
实施例1
Figure PCTCN2020072792-appb-000020
第一步
将化合物1a(23.0g,328mmol,24.5mL,1.0eq)和二碘化锌(5.20g,16.4mmol,0.05eq)溶解于200 毫升二氯甲烷中,降内温至0℃,加入三甲基硅氰(39.1g,393mmol,49mL,1.2eq),反应液于25℃下反应18小时,反应完全,浓缩反应液并加入50毫升乙腈和50毫升盐酸水溶液(1N),于25℃搅拌5分钟。乙酸乙酯萃取,干燥有机相,过滤,浓缩,蒸发残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得1b。
1H NMR(400MHz,CDCl 3)δ:4.30(s,1H),2.62-2.59(m,2H),2.33-2.30(m,2H),1.95-1.79(m,2H)。
第二步
0℃条件下,向装有四氢铝锂(9.38g,247mmol,1.5eq)的四氢呋喃溶液(300mL)中加入1b(16.0g,164mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(9.38mL)、15%氢氧化钠(9.38mL)和水(28.1mL),搅拌15分钟,过滤,浓缩得1c。
1H NMR(400MHz,CDCl 3)δ:2.72(s,2H),2.12-1.76(m,7H),1.7-1.63(m,1H),1.49-1.33(m,1H)。
第三步
0℃条件下,向装有1c(2.0g,19.8mmol,1eq)和二异丙基乙基胺(4.0g,31.0mmol,5.4mL,1.6eq)的二氯甲烷(20.0mL)溶液中加入氯乙酰氯(2.23g,19.8mmol,1.6mL,1.0eq)。反应液于20℃下反应2小时。反应完全,浓缩反应液,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得1d。
1H NMR(400MHz,CDCl 3)δ:6.99(s,1H),4.17-4.02(m,2H),3.50(d,J=6.0Hz,2H),2.71(s,1H),2.14-1.98(m,4H),1.81-1.71(m,1H),1.64-1.49(m,1H)。
第四步
将化合物1d(2.2g,12.4mmol,1eq)加入无水四氢呋喃(100mL)中,降内温至0℃,加入氢化钠(1.49g,37.2mmol,60%纯度,3eq)。反应液于20℃下反应18小时,反应完全,向反应中加入水(15.0mL),用乙酸乙酯(20mL×3)萃取,合并有机相,干燥,过滤,浓缩得1f。
1H NMR(400MHz,CDCl 3)δ:6.96(s,1H),4.15(s,2H),3.44-3.35(m,2H),2.27-2.16(m,2H),2.09-2.01(m,2H),1.95-1.86(m,1H),1.74-1.63(m,1H)。
第五步
0℃条件下,向装有四氢铝锂(645mg,17mmol,2eq)的四氢呋喃溶液(30mL)中加入1f(1.2g,8.5mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(0.7mL)、15%氢氧化钠(0.7mL)和水(2.1mL),搅拌15分钟,过滤,浓缩得1g。
1H NMR(400MHz,CDCl 3)δ:3.61-3.51(m,2H),2.87-2.76(m,4H),2.03-1.97(m,4H),1.86-1.82(m,1H),1.62-1.54(m,1H)。
第六步
将化合物1g(53mg,414μmol,1.1eq),1h(150mg,377μmol,1eq)和DIPEA(48mg,377μmol,66μL,1eq)溶于DMSO(4mL),使混合溶液70℃反应18小时。反应完全,反应液经高效液相色谱法纯化得化合物1。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.22(d,J=7.6Hz,1H),8.05(d,J=8.4Hz,1H),7.97(d,J=7.6Hz,1H),7.65-7.41(m,2H),6.53(br s,1H),4.40(d,J=6.8Hz,1H),4.12-3.95(m,3H),3.93-3.83(m,4H),3.83-3.68(m,5H),3.06(d,J=4.8Hz,3H),2.07-2.04(m,,4H),1.91-1.80(m,1H),1.77-1.70(m,1H),1.50(d,J=6.8Hz, 3H)。
实施例2
Figure PCTCN2020072792-appb-000021
第一步
将化合物1h(70m g,176μmol,1eq)、2a(40.2mg,176μmol,1eq)和二异丙基乙胺(22.7mg,176μmol,30.7μL,1eq)溶于二甲基亚砜(5mL),使混合溶液在70℃反应17小时。反应完全,反应冷却后,向反应液中加入10mL水和30mL的乙酸乙酯进行萃取。然后向有机相中加入水萃取多余的二甲基亚砜,有机相用无水硫酸钠干燥,浓缩。板层析(0/1石油醚/乙酸乙酯),得到化合物2b。
MS-ESI计算值[M+H] +590,实测值590。
第二步
将化合物2b(100mg,169μmol,1eq)溶于乙酸乙酯(3mL),然后向上述溶液中加入盐酸/乙酸乙酯(4M,3mL,70.8eq),使反应液于20℃下反应3小时。反应完全后将反应液浓缩,然后加入水10mL和乙酸乙酯45mL(15mL×3)进行萃取,有机相用无水硫酸钠干燥,浓缩。取少量反应液经高效液相色谱法纯化得化合物2c。
MS-ESI计算值[M+H] +490,实测值490。
1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.36-8.29(m,2H),7.96(d,J=7.8Hz,1H),7.69(d,J=8.4Hz,1H),7.64(t,J=7.8Hz,1H),4.62(br d,J=6.4Hz,1H),4.19-3.86(m,7H),3.81-3.72(m,5H),3.65(br d,J=9.2Hz,2H),3.53(br d,J=8.0Hz,2H),2.99(s,3H),1.51(d,J=6.8Hz,3H)。
第三步
将化合物2c(150mg,306μmol,1eq)和甲醛(11.96mg,398μmol,11.0μL,1.3eq)溶于二氯乙烷(10mL)和醋酸(2mL),然后加入氰基硼氢化钠(38.5mg,613μmol,2eq),使混合溶液20℃反应18小时。反应完全,反应冷却后,将反应液减压浓缩残留物经高效液相色谱法纯化得化合物2。
MS-ESI计算值[M+H] +504,实测值504。
1H NMR(400MHz,CD 3OD)δ:8.63(s,1H),8.36-8.30(m,2H),7.96(d,J=8.0Hz,1H),7.71(d,J=8.6Hz,1H),7.65(t,J=7.8Hz,1H),4.63(br s,2H),4.17-3.85(m,7H),3.83-3.67(m,8H),2.99(s,3H),2.61(s,3H),1.51(d,J=6.8Hz,3H)。
实施例3
Figure PCTCN2020072792-appb-000022
第一步
将化合物三苯基甲基溴化膦(25.9g,72.6mmol,1.6eq)溶于350毫升四氢呋喃中,20℃条件下,加入叔丁醇钾(1M,81.7mL,1.8eq),反应液于20℃下反应3小时,向反应液中加入3a(8.0g,45.4mmol,1eq),反应18小时。反应完全,向其中加入水(200mL)和乙酸乙酯(300mL)萃取,有机相用饱和食盐水(100mL x 3)洗涤,干燥有机相,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得3b。
1H NMR(400MHz,CDCl 3)δ:7.36-7.31(m,5H),4.87-4.85(m,2H),4.46(s,2H),,4.45-4.08(m,1H),2.89-2.86(m,2H),2.78-2.73(m,2H)。
第二步
向装有3b(1.5g,8.61mmol,640μL,1eq)和N-(2-羟乙基)甲酸叔丁酯(1.67g,10.3mmol,1.60mL,1.2eq)的乙腈溶液(14mL)中加入NIS(2.32g,10.3mmol,1.2eq),并在20℃下反应4小时。反应完全,依次向反应液中加入水(20mL)和乙酸乙酯(30mL)萃取,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)3c。
1H NMR(400MHz,CDCl 3)δ:7.33-7.30(m,5H),5.01-4.97(m,1H),4.44-4.43(m,2H),3.79-3.71(m,1H),3.33-3.32(m,3H),2.45-2.40(m,5H),2.03-2.02(m,1H),1.46(s,9H)。
第三步
将化合物3c(1.8g,3.90mmol,1eq)加入无水四氢呋喃(50mL)中,降内温至0℃,加入氢化钠(312mg,7.80mmol,60%,2eq)。反应液于20℃下反应18小时,反应完全,向反应中加入水(50.0mL),用乙酸乙酯(50mL)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得3d。
1H NMR(400MHz,CDCl 3)δ:7.33-7.20(m,5H),4.63-4.09(m,3H),3.52-3.50(m,2H),3.37(s,1H),3.30(s,2H),3.18(s,1H),2.35(s,1H),2.28-2.15(m,1H),1.97-1.85(m,2H),1.43-1.28(m,9H)。
第四步
将化合物3d(2.6g,13.6mmol,1eq)加入乙酸乙酯(15mL)中,向其中加入湿钯碳(0.1g,10%)。反应液用氢气置换三次并在此氛围(15psi)下于25℃下反应2小时,反应完全,过滤,浓缩得3e,粗品直接用于下一步。
第五步
向装有3e(360mg,1.48mmol,1eq)和乙酸乙酯(4mL)的反应瓶中加入HCl/EtOAc(4M,10mL,27eq),并在20℃下反应2小时。反应完全,浓缩得粗品3f,直接用于下一步。
第六步
将化合物3f(210mg,1.47mmol,1eq)和DIPEA(381mg,2.95mmol,513μL,2eq)溶于二氯甲烷(5mL)中,向反应液中加入氯甲酸苄酯(302mg,1.77mmol,251μL,1.2eq)。反应于20℃反应18小时。反应完全,向反应中加入水(30.0mL),用乙酸乙酯(20mL x 3)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得3g。
1H NMR(400MHz,CDCl 3)δ:7.43-7.29(m,5H),5.23-5.10(m,2H),4.55-4.10(m,1H),3.61(s,2H),3.55(s,1H),3.47-3.45(m,2H),3.34(s,1H),2.60-2.30(m,2H),1.95-1.93(m,2H)。
第七步
将化合物3g(320mg,1.15mmol,1eq)溶于二氯甲烷(5mL)中,向反应液中加入DMP(636mg,1.50mmol,1.3eq)。反应于20℃反应2小时。反应完全后浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得3h。
1H NMR(400MHz,CDCl 3)δ:7.46-7.30(m,5H),5.16(s,2H),3.71(s,2H),3.62(s,2H),3.58-3.52(m,2H),3.15-3.07(m,2H),2.98(s,2H)。
第八步
将化合物3h(200mg,727μmol,1eq)溶于二氯甲烷(1mL)中,向反应液中加入N,N-二乙基三氟化硫(703mg,4.36mmol,576μL,6eq),反应于20℃反应18小时。反应完全,向反应中加入水(40.0mL),用二氯甲烷(30mL×3)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶柱色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得3i。
第九步
将化合物3i(180mg,606μmol,1eq)加入甲醇(5mL)中,向其中加入湿钯碳(0.01g,10%)。反应液用氢气置换三次并在此氛围(15psi)下于20℃下反应2小时,反应完全,过滤,浓缩得3j,粗品直接用于下一步。
第十步
将化合物3j(42mg,257μmol,1eq)、1h(102mg,257μmol,1eq)和N,N-二异丙基乙基胺(66.5mg,514μmol,89.7μL,2eq)溶于DMSO(1mL),使混合溶液在70℃反应18小时。反应完全,反应液经高效液相色谱法纯化得3。
MS-ESI计算值[M+H] +525,实测值525。
1H NMR(400MHz,CDCl 3)δ:8.54(s,1H),8.15(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.89(d,J=8.0Hz,1H),7.54-7.41(m,2H),6.38(br s,1H),4.36(br s,1H),4.06-3.84(m,6H),3.79-3.53(m,6H),2.99(d,J=4.8Hz,3H),2.73-2.46(m,4H),1.44(d,J=6.8Hz,3H)。
实施例4
Figure PCTCN2020072792-appb-000023
Figure PCTCN2020072792-appb-000024
第一步
将化合物4a(0.28g,1.13mmol,1eq)溶于三氟乙酸(5.00mL)和二氯甲烷(10.0mL),室温搅拌反应2小时,反应完成后,减压旋干,得4b。
MS-ESI计算值[M+H] +148,实测值148。
第二步
将化合物4b(300mg,1.15mmol,1eq,TFA)、化合物1h(320mg,804μmol,0.7eq)、DIPEA(445mg,3.45mmol,600μL,4eq)溶于二甲基亚砜(5.00mL),70℃反应16小时,反应完成后,经高效液相色谱法提纯得4c。
MS-ESI计算值[M+H] +509,实测值509。
第三步
将化合物4c(100mg,197μmol,1eq)、对甲苯磺酰氯(37.5mg,197μmol,1eq)和氢化钠(15.7mg,393μmol,60%,2eq)溶于DMF(10.0mL),室温反应16小时,反应完成后,经高效液相色谱法提纯得4。MS-ESI计算值[M+H] +491,实测值491。
1H NMR(400MHz,CDCl 3)δ:8.65(s,1H),8.23(br d,J=7.8Hz,1H),8.09(d,J=8.4Hz,1H),7.99(br d,J=7.8Hz,1H),7.62-7.53(m,2H),6.60(br s,1H),4.66(d,J=6.4Hz,2H),4.57-4.41(m,3H),4.32-4.14(m,2H),4.08-3.85(m,5H),3.83-3.75(m,5H),3.08(d,J=4.8Hz,3H),1.52(d,J=6.8Hz,3H)。
实施例5
Figure PCTCN2020072792-appb-000025
第一步
将化合物4c(70.0mg,138umol,1eq)、甲烷磺酰氯(0.8g,6.98mmol,541μL,50.7eq)和三乙胺(27.9mg,275μmol,38.3μL,2eq)溶于二氯甲烷(10.0mL)中,室温反应16小时,反应完成后,减压旋干,得化合物5a。
MS-ESI计算值[M+H] +665,实测值665。
第二步
将化合物5a(59.9mg,90.26μmol,1eq)、四正丁基碘化铵(3.33mg,9.03μmol,0.1eq)、硫化钠(21.1mg,271μmol,11.4μL,3eq)溶于N,N’-二甲基甲酰胺(5.00mL),在氮气保护下,70℃反应18小时,反应完成后,水洗(50.0mL×3),经高效液相色谱法提纯得5。
MS-ESI计算值[M+H] +507,实测值507。
1H NMR(400MHz,CDCl 3)δ:8.67(br s,1H),8.20(br d,J=8.0Hz,1H),8.10-8.03(m,1H),7.99(br d,J=8.0Hz,1H),7.60-7.52(m,2H),6.64(br s,1H),4.46(br d,J=5.6Hz,1H),4.42-4.33(m,1H),4.19(br d,J=13.2Hz,1H),4.09-3.91(m,3H),3.91-3.69(m,7H),3.42(br d,J=10.0Hz,2H),3.06(d,J=4.8Hz,3H),3.00(br d,J=8.0Hz,2H),1.53(br d,J=6.8Hz,3H)。
实施例6
Figure PCTCN2020072792-appb-000026
将化合物5(120mg,237μmol,1eq)溶于甲醇(5.00mL)中,滴加单过硫酸氢钾(291mg,474μmol,2eq)的水溶液(5.00mL),室温反应30小时,反应完成后,经高效液相色谱法提纯得6。
MS-ESI计算值[M+H] +539,实测值539。
1H NMR(400MHz,CDCl 3)δ:8.58(br s,1H),8.14(br d,J=8.0Hz,1H),8.02(d,J=8.4Hz,1H),7.91(br d,J=7.6Hz,1H),7.60-7.44(m,2H),6.57(br s,1H),4.42(br d,J=6.0Hz,1H),4.27-3.85(m,10H),3.82-3.60(m,6H),2.99(d,J=4.8Hz,3H),1.46(d,J=6.8Hz,3H)。
实验例1:体外评价mTOR激酶抑制活性
实验材料:
本实验测试于DiscoverX,所有材料和方法均来自DiscoverX。
实验操作:
激酶活性分析。
1将标记的mTOR激酶稳定的表达于HEK-293细胞中。
2室温条件下,用生物素化小分子配体处理链霉亲和素磁珠30分钟,产生用于激酶分析的亲和树脂;
3配体珠用过量的生物素阻断并用缓冲液(1%牛血清白蛋白,0.05%吐温20毫升,1毫升二硫苏糖醇)洗涤,清洗掉未结合的配体和非特异性结合的配体;
4激酶配体亲和珠,组装,测试化合物三者的结合反应均在缓冲液(20%封闭缓冲液,0.17x磷酸盐缓冲液,0.05%吐温20,6毫升二硫苏糖醇)中实现;
5测试化合物溶于二甲基亚砜中;
6所有用于测量的化合物都是溶解于DMSO,然后将化合物直接稀释到浓度为0.9%。
7溶液置于384孔聚丙烯板,每个体积为0.02毫升;
8室温条件下,摇晃1小时;
9用缓冲液洗(1x PBS,0.05%吐温20)洗涤
10亲和珠再次悬浮于缓冲液(1x PBS,0.05%吐温20,0.5μm的非生物素亲和配体)并在室温孵化30分钟。
11用qPCR测量洗脱液中激酶的浓度。
实验结果:
表1 mTORC1和mTORC2激酶复合物活性测试结果
Figure PCTCN2020072792-appb-000027
结论:本发明化合物具有显著甚至意料不到的mTOR激酶抑制活性。
实验例2细胞增殖抑制活性评价:
实验目的:检测待测化合物对细胞增殖抑制活性。
实验原理:Cell-Titer-Glo试剂中的荧光素酶利用荧光素、氧和ATP作为反应底物,产生氧化荧光素,并以光的形式释放能量。由于荧光素酶反应需要ATP,因而反应产生的光的总量和反应细胞活力的ATP数总量成正比。
实验材料:
细胞系:MCF-7细胞系(ATCC-CRL-22),HT-29细胞系(ATCC-HTB-38),OE21(ECACC-96062201),NCI-N87细胞系(ATCC-CRL-5822)
细胞培养基:(RPMI 1640培养基(Invitrogen#1868546;10%血清Invitrogen#1804958;左旋谷酰胺1旋,Invitrogen#1830863;双抗Hyclone#J170012))
Cell Titer-Glo J发光法细胞活力检测试剂盒(Promega#G7573)
384孔细胞培养板(Greiner#E15103MA)
化合物板(LABCYTE#0006346665)
CO 2培养箱(Thermo#371)
Vi-cell细胞计数仪(Beckman Coulter)
移液器(Eppendorf)
移液管(Greiner)
移液枪(Eppendorf)
多功能酶标仪(Envision Reader)
ECHO Liquid-handling workstation(Labcyte-ECHO555)
实验步骤和方法:
2.1第1天:
按照细胞种板示意图在384或96孔板中分别按每孔1000个细胞,25μL每孔的密度种板,边缘孔不种细胞补25μL PBS。
2.2第1天:
(1)化合物母液为10mM,用DMSO稀释化合物使其初始浓度为4mM。在化合物母液板上加入化合物,每孔9μL。
(2)用ECHO液体工作站做化合物稀释并向细胞板每孔加入125nL化合物,第2列和23列细胞孔每孔加125nL DMSO,第1列和24列PBS孔每孔加125nL DMSO。
(3)细胞板每孔补加25μL培养基,最终细胞板每孔为50μL,化合物浓度为1μM,3倍稀释,10个浓度,左右复孔,DMSO终浓度为0.25%。
2.3加好化合物后,1000rpm离心1min,将细胞板放置于37℃、5%CO 2培养箱中培养3天。
2.4第三天:
从培养箱中取出细胞板,在室温下平衡30分钟。向每孔加入25μL Cell-Titer-Glo试剂,振摇一分钟使它被充分混匀,1000rpm离心1分钟。10分钟后,在PerkinElmer Envision上读板,设置荧光读取时间0.2秒。试验结果:试验结果如表2
表2:本发明化合物体外细胞增殖抑制活性筛选试验结果
Figure PCTCN2020072792-appb-000028
结论:本发明化合物对MCF-7,N87和OE-21细胞增殖抑制活性明显,对HT-29细胞具有一定的增殖抑制活性。
实验例3:药代动力学评价
实验方法
受试化合物与5%DMSO/95%10%聚氧乙烯蓖麻油(Cremophor EL)混合,涡旋并超声,制备得到 1mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,静脉注射给予候选化合物溶液,剂量为1或2mg/kg。受试化合物与1%吐温80,9%聚乙二醇400,90%水溶液混合,涡旋并超声,制备得到1mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,口服给予候选化合物溶液,剂量为2或10mg/kg。收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
测试结果:
测试结果见表3。
表3实施例化合物血浆中的药物代谢动力学(PK)参数
Figure PCTCN2020072792-appb-000029
表5实施例化合物血浆中的PK参数
Figure PCTCN2020072792-appb-000030
“--”是指未测试或未获得数据。
试验结论:化合物1展现了与参比化合物相同甚至更优的药代动力学性质。
实验例4人鼻咽癌C666-1细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究:
实验目的:研究本专利待测化合物对人鼻咽癌C666-1细胞皮下异种移植瘤在BALB/c裸小鼠模型体内药效进行评估
实验动物:雌性BALB/c裸小鼠,6-8周龄,体重18-22克;供应商:南大模式动物
实验方法与步骤:
4.1细胞培养
人鼻咽癌C666-1细胞,体外单层培养,培养条件为DMEM培养基中加10%胎牛血清,100U/mL青霉素,100U/mL链霉素,37℃,5%CO 2培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
4.2肿瘤细胞接种(肿瘤接种)
将0.1ml(1×10 7)C666-1细胞(DMEM双无)皮下接种于每只小鼠的右后背,肿瘤平均体积达到200mm 3时开始分组给药
4.3受试物的配制:
受试化合物配制成5mg/mL和5mg/mL的澄清溶液,溶媒为5%DMSO+30%PEG400+65%水
4.4肿瘤测量和实验指标
实验指标是考察肿瘤生长是否被抑制、延缓或治愈。每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计 算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)或肿瘤增殖率T/C(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。
肿瘤增殖率T/C(%):计算公式如下:T/C(%)=某处理组给药结束时平均瘤体积/溶剂对照组治疗结束时平均瘤体积×100%。
4.5统计分析
统计分析,包括每个组的每个时间点的肿瘤体积的平均值和标准误(SEM)(具体数据见表5-1)。治疗组在试验结束时给药后第28天表现出最好的治疗效果,因此基于此数据进行统计学分析评估组间差异。两组间比较用T-test进行分析,三组或多组间比较用one-way ANOVA进行分析,经检验,F值有显著性差异,应用Games-Howell法进行检验。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
4.6试验结果
4.6.1死亡率、发病率及体重变化情况
实验动物的体重作为间接测定药物毒性的参考指标。在此模型治疗组小鼠体重有下降趋势,溶剂组早期死亡一只,疑饲养密度过高,解剖未发现异常;实施例2组第28天死亡一只,体重过低、毒性太大死亡,解剖未发现异常。(AZD2014组及实施例2组均有一只体重低于17g)
4.6.2抗肿瘤药效评价指标
表4本发明化合物对人鼻咽癌C666-1细胞皮下异种移植瘤模型的抑瘤药效评价
(基于给药后第28天肿瘤体积计算得出)
Figure PCTCN2020072792-appb-000031
注:
a.平均值±SEM。
b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T 28-T 0)/(V 28-V0)]×100)计算。
c.p值根据肿瘤体积计算。
4.7试验结论和讨论
在本实验评价了本专利化合物在人鼻咽癌666-1细胞皮下异种移植瘤模型中的体内药效。与溶剂对照组相比,治疗组实施例2 30mg/kg与参照化合物AZD2014 15mg/kg药效相当,都展现了显著的抑瘤作用。

Claims (11)

  1. 式(I)所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用:
    Figure PCTCN2020072792-appb-100001
    其中,
    R 1选自C 1-3烷基,所述C 1-3烷基任选被1、2或3个R a取代;
    R 2选自C 1-3烷基,所述C 1-3烷基任选被1、2或3个R b取代;
    T选自O、S、S(=O) 2、-N(R 3)-和-C(R 4) 2-;
    R 3选自H和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R c取代;
    R 4选自H、F、Cl、Br、I和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R d取代;
    R a、R b、R c和R d分别独立地选自H、F、Cl、Br和I。
  2. 根据权利要求1所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 1选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F。
  3. 根据权利要求1或2所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 1选自CH 3
  4. 根据权利要求1所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 2选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F。
  5. 根据权利要求1或4所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 2选自CH 3
  6. 根据权利要求1所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 3选自CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F。
  7. 根据权利要求1或6所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 3选自CH 3
  8. 根据权利要求1所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 4分别独立地选自H、F、Cl、Br、I、CH 3、CF 3、CH 2CH 3、CF 2CH 3、CHFCH 2F和CF 2CH 2F。
  9. 根据权利要求1或8所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,R 4分别独立地选自H和F。
  10. 根据权利要求1所述化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,其中,结构单 元
    Figure PCTCN2020072792-appb-100002
    选自
    Figure PCTCN2020072792-appb-100003
    Figure PCTCN2020072792-appb-100004
  11. 下式所示化合物或其药学上可接受的盐在制备治疗鼻咽癌药物中的应用,
    Figure PCTCN2020072792-appb-100005
PCT/CN2020/072792 2019-01-18 2020-01-17 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用 WO2020147842A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080008916.5A CN113286594B (zh) 2019-01-18 2020-01-17 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910049704.0 2019-01-18
CN201910049704 2019-01-18

Publications (1)

Publication Number Publication Date
WO2020147842A1 true WO2020147842A1 (zh) 2020-07-23

Family

ID=71614276

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/072792 WO2020147842A1 (zh) 2019-01-18 2020-01-17 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用

Country Status (2)

Country Link
CN (1) CN113286594B (zh)
WO (1) WO2020147842A1 (zh)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101558067A (zh) * 2006-08-23 2009-10-14 库多斯药物有限公司 作为mtor抑制剂的2-甲基吗啉吡啶并、吡唑并和嘧啶并-嘧啶衍生物
CN105530937A (zh) * 2013-03-13 2016-04-27 昂克希尔迪克斯有限公司 使用7-苄基-10-(2-甲基苄基)-2,6,7,8,9,10-六氢咪唑并[1,2-a]吡啶并[4,3-d]嘧啶-5(3h)-酮的联合疗法
WO2019085996A1 (zh) * 2017-11-06 2019-05-09 南京明德新药研发股份有限公司 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5227321B2 (ja) * 2006-08-23 2013-07-03 クドス ファーマシューティカルズ リミテッド Mtor阻害剤としての2−メチルモルホリンピリド−、ピラゾ−及びピリミド−ピリミジン誘導体
CN103012399B (zh) * 2012-11-22 2015-06-17 中国科学院广州生物医药与健康研究院 7-氧代吡啶并嘧啶类化合物及其药用组合物和应用
CN104829609B (zh) * 2014-02-11 2016-08-03 北大方正集团有限公司 取代的吡啶并嘧啶化合物及其制备方法和应用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101558067A (zh) * 2006-08-23 2009-10-14 库多斯药物有限公司 作为mtor抑制剂的2-甲基吗啉吡啶并、吡唑并和嘧啶并-嘧啶衍生物
CN105530937A (zh) * 2013-03-13 2016-04-27 昂克希尔迪克斯有限公司 使用7-苄基-10-(2-甲基苄基)-2,6,7,8,9,10-六氢咪唑并[1,2-a]吡啶并[4,3-d]嘧啶-5(3h)-酮的联合疗法
WO2019085996A1 (zh) * 2017-11-06 2019-05-09 南京明德新药研发股份有限公司 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物

Also Published As

Publication number Publication date
CN113286594A (zh) 2021-08-20
CN113286594B (zh) 2023-12-15

Similar Documents

Publication Publication Date Title
ES2670416T3 (es) Compuestos de imidazo[4,5-c]quinolin-2-ona y su uso en el tratamiento de cáncer
CN113801114B (zh) 稠合二环杂芳基类衍生物、其制备方法及其在医药上的应用
RU2605096C2 (ru) Пиразолохинолиновое производное
JP2021525717A (ja) 治療用化合物
JP6948659B1 (ja) ピリダジニルチアアゾールカルボキシアミド化合物
TWI810550B (zh) 3-氮雜二環烷基衍生物及包含其之醫藥組成物
WO2020035065A1 (zh) 作为ret抑制剂的吡唑衍生物
WO2017088746A1 (zh) 新的表皮生长因子受体抑制剂及其应用
WO2015058661A1 (zh) Bcr-abl激酶抑制剂及其应用
WO2020259626A1 (zh) 作为irak4抑制剂的咪唑并吡啶类化合物
TW202220995A (zh) 一種作為btk抑制劑的化合物及其製備方法與用途
BR112021005750A2 (pt) inibidor de fgfr4 e uso do mesmo
WO2021244634A1 (zh) 咪唑并吡啶类化合物及其用途
TW202110848A (zh) 取代的稠合雙環類衍生物、其製備方法及其在醫藥上的應用
CN113874379B (zh) 作为Cdc7抑制剂的四并环类化合物
WO2020259620A1 (zh) 作为ccr2/ccr5拮抗剂的杂环烷基类化合物
CN111315750B (zh) 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物
JP7296017B2 (ja) ベンゾスルタムを含む化合物
WO2020147842A1 (zh) 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用
WO2020156568A1 (zh) 作为pd-l1免疫调节剂的氟乙烯基苯甲酰胺基化合物
CN114072396B (zh) 作为dna-pk抑制剂的喹啉和噌啉衍生物
CN110256408B (zh) 一种二芳基吡唑化合物及包含该化合物的组合物及其用途
WO2023207991A1 (zh) 稠合喹唑啉类化合物及其应用
CN111718351B (zh) 含氧取代吡唑并嘧啶化合物和药物组合物及其应用
WO2024017381A1 (zh) 一种抑制irak4活性的化合物及其应用

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: 20740938

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: 20740938

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 20740938

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 21.01.2022)

122 Ep: pct application non-entry in european phase

Ref document number: 20740938

Country of ref document: EP

Kind code of ref document: A1