CN113912591A - Biaryl compounds - Google Patents

Biaryl compounds Download PDF

Info

Publication number
CN113912591A
CN113912591A CN202110772717.8A CN202110772717A CN113912591A CN 113912591 A CN113912591 A CN 113912591A CN 202110772717 A CN202110772717 A CN 202110772717A CN 113912591 A CN113912591 A CN 113912591A
Authority
CN
China
Prior art keywords
compound
cell
experimental
added
tumor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202110772717.8A
Other languages
Chinese (zh)
Other versions
CN113912591B (en
Inventor
丁照中
刘希乐
陈曙辉
胡利红
周成亮
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Qilu Pharmaceutical Co Ltd
Original Assignee
Medshine Discovery Inc
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
Priority claimed from PCT/CN2020/133933 external-priority patent/WO2021110141A1/en
Application filed by Medshine Discovery Inc filed Critical Medshine Discovery Inc
Publication of CN113912591A publication Critical patent/CN113912591A/en
Application granted granted Critical
Publication of CN113912591B publication Critical patent/CN113912591B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Abstract

The invention discloses biaryl compounds, and particularly discloses a compound shown as a formula (I) and pharmaceutically acceptable salts thereof.

Description

Biaryl compounds
The following priority is claimed in the present application:
CN202010652149.3, filing date 2020, month 07, 08;
PCT/CN2020/133933, filing date 2020, 12/04.
Technical Field
The present invention discloses biaryl compounds, their pharmaceutically acceptable salts, which are inhibitors of Pan-RAF kinase and their use in the treatment of diseases associated with cancer.
Background
The mitogen-activated protein kinase (MAPK) pathway is an important signal transduction pathway in cells, and the pathway transfers signals from the outside of cells into cell nuclei through specific cascade phosphorylation of RAS/RAF/MEK/ERK, finally leads to activation of specific genes, and causes cell proliferation, apoptosis or differentiation. This pathway overactivation is closely related to the development of multiple tumors. RAF is a serine/threonine protein kinase that is important in the RAS/RAF/MEK/ERK signaling pathway, is located downstream of RAS, and can be activated by RAS. The RAF family includes three subtypes ARAF, BRAF and CRAF (RAF-1), and has high homology and similar structural domains. Wild-type RAF is capable of producing three subtypes of homo-or heterodimers. ARAF and CRAF mutations occur less frequently and BRAF mutation rates are higher. BRAF mutations are present in about 5% to 10% of malignant tumors, including 66% of melanoma patients. RAF kinase as a key signal protein downstream of RAS has important research significance in treating RAS mutant gene mutant tumors. Through inhibiting RAF kinase, MAPK signal transduction is regulated, so that the tumor cell proliferation promoter plays a role in RAS mutant tumor cell proliferation. Therefore, RAF kinase has become an important target for clinical treatment of tumors.
The first generation BRAF kinase inhibitors Vemurafenib and Dabrafinib have been FDA approved for the development of B-RafV600ETreatment of mutant cancers. Although Vemurafenib and Dabrafinib are in B-RafV600EThe treatment of mutant melanoma shows promising efficacy but still has certain limitations. Most patients using both drugs initially have a tumor that shrinks but recurs within a year (acquired resistance); the primary mechanism for developing this resistance is the reactivation of the MAPK signaling pathway. Research finds that on the basis of V600E mutation of BRAF, NRAS mutation can cause MAPK pathway activation in the presence of an inhibitor to generate drug resistance. Mutant N-Ras promotes B-RafV600EAnd C-Raf form homo-or heterodimers. The inhibitor binds to one monomer of the dimer, reducing the affinity of the drug for the other monomer, and promoting phosphorylation of the monomer without the effect of the inhibitor, resulting in activation of MEK. At present, the main method for clinically overcoming drug resistance caused by reactivation of MAPK pathway is to block 2 key sites of MAPK pathway by combined use of Raf and MEK inhibitors so as to delay drug resistance generation. In addition, development of a new generation of pan-Raf inhibitors, which can inhibit the activity of dimers and block abnormal activation of the pathway (paradoxcal activation), thereby reducing drug resistance, to overcome drug resistance and to expand the scope of clinical application is under development. The pan-Raf dimer inhibitors in clinical research mainly comprise HM95573, TAK-580, BGB-283, LXH254, LY3009120 and the like, and the development of the novel RAF inhibitors is expected to overcome the drug resistance of the first-generation inhibitors and further expand the clinical application.
Disclosure of Invention
The present invention provides a compound of formula (I), N- (3- (2- ((1R,5S) -3-oxabicyclo [3.1.0] hex-1-yl) -6- (2-hydroxyethoxy) pyridin-4-yl) -4-methylphenyl) -2- (trifluoromethyl) isonicotinamide or a pharmaceutically acceptable salt thereof,
Figure BDA0003154430400000021
the present invention also provides compounds of the formula:
Figure BDA0003154430400000022
the invention also provides
Figure BDA0003154430400000023
Figure BDA0003154430400000024
In the preparation of compounds of formula (I)
Figure BDA0003154430400000025
The use of (1).
The invention also provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a RAF kinase inhibitor.
The invention also provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cancer.
In some embodiments of the invention, the cancer is colon cancer or lung cancer.
Technical effects
The compound has good drug properties, good RAF enzyme inhibition activity and various cell antiproliferative activities, and simultaneously has good in-vivo drug effect and good safety. Is expected to solve the current BRAFV600EResistance to mutant cancer treatments, and provide effective treatments for RAS mutant-type cancers.
Definitions and explanations
As used herein, the following terms and phrases are intended to have the following meanings, unless otherwise indicated. A particular term or phrase, unless specifically defined, should not be considered as indefinite or unclear, but rather construed according to ordinary meaning. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The term "pharmaceutically acceptable" as used herein is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amines or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting such compounds with a sufficient amount of acid, either in neat solution or in 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, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are salts of amino acids such as arginine and the like, and salts of organic acids such as glucuronic acid and the like. Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains an acid or base, by conventional chemical methods. In general, such salts are prepared by the following method: prepared by reacting these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid, in water or an organic solvent or a mixture of the two.
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, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
Unless otherwise indicated, the terms "enantiomer" or "optical isomer" refer to stereoisomers that are mirror images of each other.
Unless otherwise indicated, the term "cis-trans isomer" or "geometric isomer" results from the inability of a double bond or a single bond to rotate freely within a ring-forming carbon atom.
Unless otherwise indicated, the term "diastereomer" refers to a stereoisomer in which the molecules have two or more chiral centers and a non-mirror image relationship between the molecules.
Unless otherwise indicated, "(+)" means dextrorotation, "(-) -means levorotation," (±) "means racemization.
Using solid wedge keys, unless otherwise indicated
Figure BDA0003154430400000041
And wedge dotted bond
Figure BDA0003154430400000042
Showing the absolute configuration of a solid centre, by means of straight solid keys
Figure BDA0003154430400000043
And straight dotted line bond
Figure BDA0003154430400000044
Showing the relative configuration of the centres of solids, by wavy lines
Figure BDA0003154430400000045
Shows a solid wedge lineKey with a key body
Figure BDA0003154430400000046
Or wedge dotted bond
Figure BDA0003154430400000047
Or by wavy lines
Figure BDA0003154430400000048
Indicating straight solid-line keys
Figure BDA0003154430400000049
And straight dotted line bond
Figure BDA00031544304000000410
Unless otherwise indicated, the term "tautomer" or "tautomeric form" means that at room temperature, the isomers of different functional groups are in dynamic equilibrium and can be rapidly interconverted. If tautomers are possible (e.g., in solution), then the chemical equilibrium of the tautomers can be reached. For example, proton tautomers (prototropic tautomers), also known as proton transfer tautomers (prototropic tautomers), include interconversions by proton transfer, such as keto-enol isomerization and imine-enamine isomerization. Valence isomers (valencetatomer) include interconversion by recombination of some of the bonding electrons. A specific example of where keto-enol tautomerism is the interconversion between two tautomers of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one.
Unless otherwise indicated, the terms "enriched in one isomer", "isomer enriched", "enantiomer enriched" or "enantiomeric enrichment" refer to a content of one isomer or enantiomer of less than 100%, and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.
Unless otherwise indicated, the term "isomeric excess" or "enantiomeric excess" refers to the difference between the relative percentages of two isomers or enantiomers. For example, if the content of one isomer or enantiomer is 90%, and the content of the other isomer or enantiomer is 10%, the isomer or enantiomer excess (ee value) is 80%.
Optically active (R) -and (S) -isomers as well as D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one of the enantiomers of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl), diastereomeric salts are formed with an appropriate optically active acid or base, followed by diastereomeric resolution by conventional methods known in the art, and the pure enantiomers are recovered. Furthermore, separation of enantiomers and diastereomers is typically accomplished by using chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (e.g., carbamate formation from amines).
The term "protecting group" includes, but is not limited to, "amino protecting group," hydroxyl protecting group, "or" thiol protecting group. The term "amino protecting group" refers to a protecting group suitable for use in preventing side reactions at the amino nitrogen position. Representative amino protecting groups include, but are not limited to: a formyl group; acyl, for example alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl groups such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl groups such as benzyl (Bn), trityl (Tr), 1-bis- (4' -methoxyphenyl) methyl; silyl groups, such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like. The term "hydroxy protecting group" refers to a protecting group suitable for use in preventing side reactions of a hydroxy group. Representative hydroxy protecting groups include, but are not limited to: alkyl groups such as methyl, ethyl and tert-butyl; acyl groups, such as alkanoyl (e.g., acetyl); arylmethyl groups such as benzyl (Bn), p-methoxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (benzhydryl, DPM); silyl groups, such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like.
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention.
The compounds of the present invention may be structurally confirmed by conventional methods well known to those skilled in the art, and if the present invention relates to the absolute configuration of the compound, the absolute configuration may be confirmed by means of conventional techniques in the art. For example, single crystal X-ray diffraction method (SXRD), diffraction intensity data of the cultured single crystal is collected by a Bruker D8 venture diffractometer, a light source is CuK alpha radiation, and a scanning mode is as follows:
Figure BDA0003154430400000051
after scanning and collecting relevant data, the crystal structure is further analyzed by a direct method (Shelxs97), so that the absolute configuration can be confirmed. The absolute configuration of the compound can also be confirmed by the chiral structure of the starting material and the reaction mechanism of the asymmetric synthesis.
The solvent used in the present invention can be commercially available.
The invention employs the following abbreviations: HATU stands for 2- (7-azobenzotriazol) -N, N' -tetramethyluronium hexafluorophosphate; HOBt represents 1-hydroxybenzotriazole; t is3P represents tri-n-propyl cyclic phosphoric anhydride; pd2dba3Represents tris-diphenylpropanone-dipalladium; ruphos represents 2-dicyclohexyl-phosphorus-2 ',6' -diisopropoxy-1, 1' -biphenyl; brettphos for (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl; TBAF for tetrabutylammonium fluoride, [ Ir (COD) OMe]2Represents cyclooctadiene methoxy iridium dimer; tmphen represents 3,4,7, 8-tetramethyl-1, 10 phenanthroline; dtbpy represents 4,4 '-di-tert-butyl-2, 2' -bipyridine. Pd (dppf) Cl2Represents diphenylphosphinoferrocene palladium dichloride; pd (dppf) Cl2DCM for diphenylphosphinoferrocene palladium dichloride complex with dichloromethane; DIAD represents diisopropyl azodicarboxylate; DIPEA or DIEA for diisopropylethylamine; PPh3Represents triphenylphosphine; TFA represents trifluoroacetic acid; THP represents 2-tetrahydropyran; OTHP represents 2-oxotetrahydropyran; TBSCl represents tert-butyldimethylsilyl chloride; TBS represents tert-butyldimethylsilyl group; OTBS stands for tert-butyl dimethylsiloxy; DMF represents N, N-dimethylformamide; DMSO represents dimethyl sulfoxide; EA represents ethyl acetate; PE represents petroleum ether; EtOH stands for ethanol; MeOH represents methanol; DME represents ethylene glycol dimethyl ether; DCM represents dichloromethane; THF represents tetrahydrofuran; MeCN represents acetonitrile; NMP stands for N-methylpyrrolidone; PE/EA represents the volume ratio of petroleum ether to ethyl acetate; DCM/MeOH represents the volume ratio of dichloromethane to methanol; HCOOH-MeCN-H2O represents a separation system of formic acid-acetonitrile-water; IPA stands for isopropanol; me represents methyl, OMs represents methanesulfonyloxy.
Drawings
FIG. 1 is a schematic representation of a crystal of a compound of formula (I).
FIG. 2 is the tumor growth curve of mouse with tumor of human colon cancer Colo-205 cell subcutaneous xenograft tumor model.
FIG. 3 is the body weight change curve of mouse with human colon cancer Colo-205 cell subcutaneous xenograft tumor model.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention. It will be apparent to those skilled in the art that various changes and modifications can be made in the specific embodiments of the invention without departing from the spirit and scope of the invention.
Example 1: preparation of Compounds of formula (I)
Scheme 1:
Figure BDA0003154430400000061
compound 2
To a solution of compound 1(10 g, 51.96 mmol) in dichloromethane (100mL) at-78 deg.C was added n-butyllithium (23mL, 2.5M), and the mixture was stirred for 30 minutes. To this mixture was added 1a (4.92 g, 57.16 mmol) at-78 ℃ and the mixture was stirred at 25 ℃ for 0.5 hour. To the reaction mixture was added a saturated ammonium chloride solution (40mL), and extraction was performed with dichloromethane (40mL × 2), and the combined extracts were dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography (PE/EA ═ 10/1 to 5/1, V/V) to obtain compound 2.1HNMR(400MHz,CDCl3)δ7.72(t,J=7.8Hz,1H),7.47(d,J=7.2Hz,1H),7.28(d,J=7.2Hz,1H),4.35(s,1H),4.24-4.14(m,2H),4.05-4.00(m,1H),3.96-3.92(m,1H),2.45(td,J=8.8,13.0Hz,1H),2.32-2.22(m,1H)。
Compound 3
To a solution of compound 2(6 g, 30.06 mmol) in toluene (80mL) was added p-toluenesulfonic acid (11.5 g, 60.46 mmol), and the mixture was stirred for 16 hours while warming to 110 ℃. To the reaction mixture were added a saturated sodium bicarbonate solution (40mL) and ethyl acetate (100mL), and the mixture was separated, washed with a saturated sodium bicarbonate solution (50mL × 2) with a camera, dried over anhydrous sodium sulfate, concentrated, and subjected to silica gel column chromatography (PE/EA ═ 50/1 to 20/1, V/V) to obtain compound 3.1HNMR(400MHz,CDCl3)δ7.64(t,J=7.8Hz,1H),7.23(dd,J=5.2,7.8Hz,2H),6.68(quin,J=2.0Hz,1H),5.08(dt,J=2.0,5.0Hz,2H),4.91(dt,J=2.0,5.0Hz,2H)。
Compound 4
To a mixture of potassium tert-butoxide (3.71 g, 33.04 mmol), compound 2a (7.3 g, 33.17 mmol) was added DMSO (50 mL). The reaction mixture was stirred at 25 ℃ under nitrogenStirring for 1 hour. To the reaction was added a solution of 3(1 g, 5.51 mmol) in DMSO (5mL), and the reaction mixture was heated to 70 ℃ and stirred for 6 hours. Water (180mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (30 × 3mL), and the organic phase was dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column chromatography (PE/EA ═ 20/1, V/V) to give compound 4.1HNMR(400MHz,CDCl3)δ7.46(t,J=7.8Hz,1H),7.05(d,J=7.8Hz,1H),6.88(d,J=7.6Hz,1H),4.11-4.04(m,2H),3.89-3.78(m,2H),2.08(ddd,J=2.8,5.2,8.0Hz,1H),1.32(dd,J=4.4,8.0Hz,1H),1.07(t,J=4.6Hz,1H)。
Compound 5
Compound 3a (1.08 g, 4.25 mmol), [ Ir (COD) OMe]2(70 mg, 105.60 micromoles) and tmphen (50 mg, 211.59 micromoles) were placed in methyl tertiary-butyl ether (20 mL). The reaction mixture was stirred under nitrogen with 4(770 mg, 3.94 mmol) added and heated to 80 ℃ for 3 hours. Filtering the reaction solution, and concentrating the filtrate to obtain a crude product of the compound 5. MS (ESI) M/z240.3[ M-84+2H]+
Compound 6
Compound 5(1.2 g, 3.73 mmol), 4a (1.4 g, 3.90 mmol) and Pd (dppf) Cl2DCM (300 mg, 367.36 micromoles) and sodium carbonate (800 mg, 7.55 mmol) in dioxane (50mL) and water (10 mL). The reaction mixture was heated to 100 ℃ and stirred for 3 hours under nitrogen. The reaction mixture was filtered, the filtrate was concentrated, ethyl acetate (20mL) and water (10mL) were added, the layers were separated, the aqueous layer was extracted with ethyl acetate (10mL × 3), the combined organic layers were dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column chromatography (PE/EA ═ 10/1 to 5/1) to give compound 6.1HNMR(400MHz,CDCl3)δ8.92(d,J=5.0Hz,1H),8.20(s,1H),8.11(s,1H),7.94(d,J=5.0Hz,1H),7.60-7.51(m,2H),7.32(d,J=7.8Hz,1H),7.10(d,J=1.2Hz,1H),6.90(d,J=1.2Hz,1H),4.19-4.14(m,2H),3.95-3.87(m,2H),2.27(s,3H),2.24-2.17(m,1H),1.48-1.41(m,1H),1.20-1.15(m,1H)。
Compound 7
To a solution of compound 6(900 mg, 1.90 mmol) and compound 5a (400 mg, 2.27 mmol) in toluene (30mL) was addedInto Pd2dba3(180 mg, 196.57 micromoles), Brettphos (200 mg, 372.60 micromoles) and cesium carbonate (1.26 g, 3.87 mmol). The reaction mixture was heated to 110 ℃ and stirred for 4 hours under nitrogen. The reaction solution was filtered, and the filtrate was concentrated and separated by silica gel column chromatography (PE/EA ═ 20/1 to 6/1) to obtain compound 7.1HNMR(400MHz,DMSO-d6)δ10.65(s,1H),8.95(d,J=5.0Hz,1H),8.31(s,1H),8.14(d,J=4.6Hz,1H),7.70(dd,J=2.0,8.2Hz,1H),7.59(d,J=2.2Hz,1H),7.29(d,J=8.4Hz,1H),6.68(s,1H),6.49(s,1H),4.31(br d,J=2.8Hz,1H),4.07-3.97(m,2H),3.90-3.86(m,2H),3.79-3.69(m,2H),2.17(s,3H),2.15-2.09(m,1H),1.33(dd,J=3.8,7.8Hz,1H),1.13(t,J=7.2Hz,1H),0.96-0.91(m,1H),0.80(s,9H),0.00(s,6H)。
Compound 8
To compound 7(300 mg, 488.81 micromoles) in tetrahydrofuran (10mL) was added hydrochloric acid (1mL, 4M), and the reaction was stirred at 25 ℃ for 1 hour. The reaction mixture was diluted with ethyl acetate (20mL), the pH was adjusted to 8 with a saturated solution of sodium hydrogencarbonate, and the mixture was extracted with ethyl acetate (10 × 3mL), and the organic phase was dried over anhydrous sodium sulfate and concentrated, followed by separation on a thin-layer silica gel plate (PE/EA 1/1, V/V) to obtain compound 8.1HNMR(400MHz,DMSO-d6)δ10.70(s,1H),9.06-8.95(d,J=5.0Hz,1H),8.37(s,1H),8.23-8.18(d,J=4.2Hz,1H),7.77-7.71(dd,J=2.2,8.4Hz,1H),7.68-7.64(d,J=2.2Hz,1H),7.39-7.31(d,J=8.4Hz,1H),6.73(s,1H),6.59-6.54(d,J=1.0Hz,1H),4.86-4.81(t,J=5.2Hz,1H),4.35-4.30(t,J=5.2Hz,2H),4.13-4.03(m,2H),3.83-3.70(m,4H),2.24(s,3H),2.22-2.08(ddd,J=2.4,5.0,7.8Hz,1H),1.40-1.32(dd,J=3.6,7.8Hz,1H),1.02-0.96(t,J=4.4Hz,1H)。MS(ESI)m/z:500.4[M+H]+
A compound of formula (I)
Compound 8 was separated chirally by SFC (chiral column DAICEL CHIRALCEL OJ-H (250mm 30mm,5 μm), mobile phase A: ethanol (containing 0.05% diisopropylethylamine), mobile phase B: carbon dioxide) to give 8A (retention time 1.487min) and the compound of formula (I) (retention time 1.590 min). 8A is an enantiomer of a compound of formula (I).
Compound 8A:1HNMR(400MHz,DMSO-d6)δ10.70(s,1H),9.06-8.95(d,J=5.0Hz,1H),8.37(s,1H),8.23-8.18(d,J=4.2Hz,1H),7.77-7.71(dd,J=2.2,8.4Hz,1H),7.68-7.64(d,J=2.2Hz,1H),7.39-7.31(d,J=8.4Hz,1H),6.73(s,1H),6.59-6.54(d,J=1.0Hz,1H),4.86-4.81(t,J=5.2Hz,1H),4.35-4.30(t,J=5.2Hz,2H),4.13-4.03(m,2H),3.83-3.70(m,4H),2.24(s,3H),2.22-2.08(ddd,J=2.4,5.0,7.8Hz,1H),1.40-1.32(dd,J=3.6,7.8Hz,1H),1.02-0.96(t,J=4.4Hz,1H)。MS(ESI)m/z:500.4[M+H]+,100%(ee%)。
a compound of formula (I):1HNMR(400MHz,DMSO-d6)δ10.70(s,1H),9.06-8.95(d,J=5.0Hz,1H),8.37(s,1H),8.23-8.18(d,J=4.2Hz,1H),7.77-7.71(dd,J=2.2,8.4Hz,1H),7.68-7.64(d,J=2.2Hz,1H),7.39-7.31(d,J=8.4Hz,1H),6.73(s,1H),6.59-6.54(d,J=1.0Hz,1H),4.86-4.81(t,J=5.2Hz,1H),4.35-4.30(t,J=5.2Hz,2H),4.13-4.03(m,2H),3.83-3.70(m,4H),2.24(s,3H),2.22-2.08(ddd,J=2.4,5.0,7.8Hz,1H),1.40-1.32(dd,J=3.6,7.8Hz,1H),1.02-0.96(t,J=4.4Hz,1H)。MS(ESI)m/z:500.4[M+H]+,99.7%(ee%)。
scheme 2:
Figure BDA0003154430400000081
step 1: compound I-2
N, N-dimethylformamide (12.5 l) was charged into a 50 l reaction vessel at room temperature, and Compound I-1(2.75 kg, 20.91 mol) and Compound I-1-1(3.55 kg, 31.36 mol) were added and stirred until dissolved and clarified. Then, potassium carbonate (7.22 kg, 52.27 mol) was added thereto, and the mixture was heated to 75 to 80 ℃ and stirred for 16 hours. After the reaction is finished, cooling the reaction liquid to 25 ℃; adding water (27 liters) into a reaction kettle, and transferring the reaction solution of about 27 liters into a white barrel of 30 liters for temporary storage; hydrochloric acid solution (6 mol/l, 8.5 l) was slowly added to the remaining 27 l of reaction solution in the kettle, 0.3 l each time, and the pH was adjusted to 3-4. A yellow solid precipitated with a large amount of gas generation and a small exotherm. The solid-liquid mixture was filtered off with suction under reduced pressure and the filter cake was washed three times with 2 liters of water each time. The remaining 27 liters of the reaction solution were treated in the same manner. After the filter cakes are combined, the mixture is dried in vacuum at 45 DEGDrying for 24 hours to obtain the compound I-2.1HNMR(400MHz,CDCl3)δ14.32(br s,1H),7.42(dd,J=7.4,9.0Hz,1H),7.12(d,J=9.0Hz,1H),6.53(dd,J=1.4,7.4Hz,1H),4.26-4.18(m,2H),1.28(t,J=7.2Hz,3H)。
Step 2: compound I-3
Dimethyl sulfoxide (12L) was charged to a 50L reactor at room temperature, compound I-2(3.6 kg, 16.03 mol) was added, and the internal temperature was raised to 70 ℃. After the reaction solution was clarified, a solution of sodium chloride (1.87 kg, 32.05 mol) in water (6 l) was added, the internal temperature was raised to 80 to 85 ℃ and stirred for 16 hours. After completion of the reaction, the temperature was lowered to 25 ℃, water (20.5 liters) was added to the kettle, stirring was carried out for 5 minutes, and the reaction solution was extracted with ethyl acetate 2 times by 10 liters each. The combined organic phases were washed twice with 10 l each time of saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure at 45 degrees to give the product, compound I-3.1HNMR(400MHz,CDCl3)δ7.74(t,J=7.8Hz,1H),7.43(d,J=7.6Hz,1H),7.34(d,J=8.0Hz,1H),3.94(s,2H)。
And step 3: compound I-4
Tetrahydrofuran (7.5 l) was charged to a 50 l reactor at room temperature, compound I-3(1.5 kg, 9.83 mol) and compound I-3-1(0.926 kg, 10 mol) were added and the internal temperature was lowered to-25 ℃. Hexamethylsilamidolithium (10 liters, 10 moles, tetrahydrofuran solution) was pumped into the kettle under nitrogen atmosphere, and the internal temperature was controlled not to exceed-15 ℃ for about 3 hours. The internal temperature rises between minus 8 ℃ and minus 5 ℃, and the reaction is carried out for 3 hours under the condition of heat preservation. The temperature was lowered to-25 ℃ and sodium hexamethyl-silazane (10 l, 10 mol, tetrahydrofuran solution) was added to the reactor by means of a peristaltic pump (addition was completed over 2 hours) with the internal temperature being controlled not to exceed-15 ℃. Raising the temperature to 20-25 ℃, and stirring for reacting for 16 hours. After completion of the reaction, water (0.883L, 49.1 mol) was slowly added to the reaction solution, and the mixture was stirred for 0.5 hour while controlling the internal temperature to be lower than 30 ℃. The reaction solution was distilled under reduced pressure at 40 ℃ in a kettle and concentrated to give a crude product solution (about 10 liters) of Compound I-4, which was used directly in the next reaction.
And 4, step 4: compound I-5
To a 50 liter reactor of crude compound I-4(10 liter solution) was added hydrogen hydroxide at room temperatureAdding an aqueous solution of potassium (9.6 liters, 19.2 moles) and ethanol (10 liters), raising the internal temperature to 75-80 ℃, and stirring for 5 hours. After the reaction is finished, cooling to 25-30 ℃, slowly adding a hydrochloric acid aqueous solution (8 liters and 48 moles) into the reaction solution, controlling the temperature not to exceed 40 ℃, heating the internal temperature to 60 ℃, and stirring for 1 hour. After completion of the reaction, concentration gave a crude product solution (about 2 l of the residue) which was extracted twice with 7.5 l each time of ethyl acetate. The extract was charged into a reaction vessel, silica gel (100 and 200 mesh, 750 g) was added, n-heptane (15L) was added, and stirring was carried out for 16 hours. Filtration was carried out, the filter cake was washed three times with 1 l each time of n-heptane/ethyl acetate (1:1), the organic phases were combined and concentrated under reduced pressure to give the crude product as a black solid. Heating the crude product with ethanol (3L) to an internal temperature of 70 deg.C, stirring to dissolve, cooling to 20 deg.C, precipitating a large amount of solid, adding water (9L) into the suspension, and stirring for 0.5 hr. Filtering, washing a filter cake twice with water, wherein each time is 1 liter; the filter cake was added to the mixed solution (n-heptane/ethanol, 4:1, 4.5 liters), stirred for 0.5 hour, filtered, washed 2 times with n-heptane/ethanol (4:1), 0.3 liters each, and dried under vacuum at 45 degrees for 16 hours to give compound I-5.1HNMR(400MHz,CDCl3)δ8.09(dd,J=0.8,7.8Hz,1H),7.64(t,J=7.8Hz,1H),7.18(dd,J=0.8,7.8Hz,1H),4.43(dd,J=4.6,9.2Hz,1H),4.31(d,J=9.2Hz,1H),3.04-2.89(m,1H),2.14(dd,J=4.4,7.8Hz,1H),1.56-1.44(m,1H)。
And 5: compound I-6
Tetrahydrofuran (16 l) was added to a 50 l kettle at room temperature, compound I-5(3.2 kg, 15.27 mol) was added, and stirred until dissolved and clear. Raising the internal temperature to 30 ℃, slowly adding a tetrahydrofuran solution of lithium borohydride (4.58 liters and 9.16 moles) into the kettle by using a peristaltic pump under the nitrogen atmosphere (after 1.5 hours of addition), controlling the temperature to be not more than 35 ℃, controlling the temperature of a jacket to be 10 ℃, controlling the internal temperature to be 25-35 ℃ after the feeding is finished, preserving the temperature for reaction for 2 hours, and stirring overnight for 16 hours. To the reaction solution, a saturated ammonium chloride (2.3 liters) solution was slowly added, and the mixture was stirred for 0.5 hour while controlling the internal temperature to be lower than 20 ℃. Adding n-heptane (8L) into the reaction solution, stirring for 0.5 hr, vacuum filtering the solid-liquid mixture, washing the filter cake with n-heptane/tetrahydrofuran (1:2) for 2 times (1.5L each time), filteringThe solution was concentrated under reduced pressure to about 8 liters, and 16 liters of ethyl acetate was added. The organic phase is washed 3 times with 3 liters each time and the aqueous phase is extracted 3 times with 2 liters each time with ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate (2 kg) and concentrated under reduced pressure. The crude product was slurried with n-heptane/ethyl acetate (10:1, 6.4 liters), stirred for 0.5 hour, filtered, the filter cake was washed 1 time with n-heptane/ethyl acetate (10:1, 3.2 liters) and the filter cake was dried under vacuum for 16 hours to give compound I-6.1HNMR(400MHz,DMSO-d6)δ7.76(t,J=7.8Hz,1H),7.59(d,J=7.8Hz,1H),7.25(d,J=7.8Hz,1H),4.84-4.62(m,2H),4.09(dd,J=6.2,12.4Hz,1H),3.88-3.71(m,2H),3.50(ddd,J=4.8,8.8,11.8Hz,1H),1.82-1.62(m,1H),1.29(dd,J=4.0,8.8Hz,1H),0.92(dd,J=4.0,6.2Hz,1H)。
Step 6: compound I-7
Tetrahydrofuran (22.5 l) was added to a 50 l kettle at room temperature, compound I-6(1.5 kg, 7.02 mol) was added, and stirred until dissolved and clear. Azodicarbonyldipiperidine (2.04 kg, 8.07 mol) was added, stirred until clear, and a solution of n-butylphosphine in tetrahydrofuran (1.63 kg, 8.07 mol) was added slowly to the kettle (over 1.5 hours) using a peristaltic pump under nitrogen, with the temperature being controlled to no more than 40 ℃. After the feeding is finished, controlling the internal temperature to be 15-25 ℃, reacting for 2 hours, and stirring overnight for 16 hours at room temperature. The reaction solution is filtered, the filter cake is washed with methyl tertiary butyl ether for 2 times, 3 liters each time, and the filtrate is concentrated to obtain a crude product. The crude product was dissolved with methyl tert-butyl ether (15 l), washed 3 times with hydrochloric acid (4 mol/l), 3 l each time, the aqueous phase was extracted 3 times with methyl tert-butyl ether, 3 l each time, the combined organic phases were washed 3 times with saturated brine, 5 l each time, dried over anhydrous sodium sulfate (2 kg), and concentrated under reduced pressure to give the crude product. The crude product was transferred to a kettle and n-heptane (7 liters) was added, warmed to an internal temperature of 70 ℃ and stirred for 0.5 hour to dissolve clear. The temperature is reduced to 15 ℃ of the internal temperature, and the mixture is stirred for 16 hours. Filtration, washing of the filter cake with n-heptane (1.4 l) 1 time, and vacuum drying of the filter cake for 16 h gave compound I-7.1HNMR(400MHz,CDCl3)δ7.46(t,J=7.8Hz,1H),7.04(dd,J=0.8,7.8Hz,1H),6.87(dd,J=0.8,7.7Hz,1H),4.10-4.04(m,2H),3.86-3.78(m,2H),2.07(ddd,J=2.8,5.2,8.0Hz,1H),1.32(dd,J=4.4,8.2Hz,1H),1.07(t,J=4.8Hz,1H)。
And 7: compound I-8
Dioxane (12L) was charged to a 50L reactor at room temperature, and Compound I-7-1(2.11 kg, 17.89 mol) and potassium tert-butoxide (2.51 kg, 22.36 mol) were added. The reaction solution was heated to 95 ℃ and a solution of compound I-7(1.75 kg, 8.94 mol) in dioxane (5.5 l) was added to the reaction solution via a constant pressure dropping funnel with an internal temperature of 90 to 100 ℃. After 30 minutes, the reaction was incubated for 30 minutes. After the reaction was complete, the temperature was reduced to 50 ℃, the pad was filtered through celite, the filter cake was washed 2 times with 2 liters each time with n-heptane, and the filtrates were combined and spin dried. The resulting yellow oil was dissolved in n-heptane (20 l) and washed 3 times with 15 l each time, and the dot plate showed no compound I-7-1 remaining. The organic phase was dried over anhydrous sodium sulfate (2.5 kg) and concentrated to give crude compound I-8.1HNMR(400MHz,CDCl3)δ7.45(t,J=7.8Hz,1H),6.66-6.48(m,2H),4.45-4.29(m,2H),4.21-4.07(m,2H),3.95-3.81(m,2H),3.75-3.64(m,2H),2.13-2.02(m,1H),1.42-1.34(m,1H),1.25-1.17(m,9H),1.10-1.02(m,1H)。
And 8: compound 4a
Dichloromethane (13 l) was added to a 50 l kettle at room temperature, and trifluoromethyl isonicotinic acid (2.6 kg, 13.6 mol) and N, N-dimethylformamide (105 ml, 1.36 mol) were added and stirred until clear. Oxalyl chloride (1.67 l, 19.05 mol) was added slowly from an isopiestic dropping funnel and the off-gas was absorbed with lye. Stirring was carried out at 20 ℃ under a nitrogen atmosphere for 16 hours. After the reaction is completed, the solvent in the reaction solution is removed by rotary evaporation, the obtained trifluoromethyl isonicotinoyl chloride is dissolved by N, N-dimethylformamide (12 liters), the solution is slowly added into a solution of 3-bromo-4-methylaniline (3.0 kg, 13.97 moles) and diisopropylethylamine (4.11 liters, 20.44 moles) in N, N-dimethylformamide (6 liters) by using a constant-pressure dropping funnel, and the internal temperature is controlled to be 15-25 ℃. After the addition was complete, the reaction was continued for 0.5 hour. And after the reaction is finished, adding the reaction solution into water (54 liters), separating out solids, controlling the temperature to be 20-30 ℃, continuing stirring for 10 minutes after the addition is finished, filtering the suspension by using a suction filtration funnel under reduced pressure, washing the filter cake once by using water (2 liters), and collecting the filter cake. The filter cake was transferred to a column filled with n-heptane (15 l)) The reaction kettle (2) is stirred for 16 hours at an internal temperature of 15-25 ℃. The reaction was filtered, the filter cake was washed 2 times with n-heptane (1 l), and the filter cake was dried under vacuum for 16 hours to give compound 4 a.1HNMR(400MHz,CDCl3)δ8.93(d,J=4.8Hz,1H),8.11(s,1H),8.01(br s,1H),7.96-7.86(m,2H),7.49(br d,J=8.2Hz,1H),7.26(d,J=8.2Hz,1H),2.41(s,3H)。
And step 9: compound I-9
At room temperature, tert-butyl methyl ether (18L) was charged into a 50L reactor, compound I-8(1.8 kg, 6.49 mol) and pinacol borate (1.73 kg, 6.81 mol) were added, the mixture was stirred to clarify, and the system was evacuated for three nitrogen replacements. Tmphen (30.67 g, 0.13 mol) and [ Ir (COD) (OMe) 2(40.02 g, 65 mmol) were added in turn to the reaction vessel, and the temperature was raised to 55-60 ℃ under the protection of nitrogen stream, and the mixture was stirred for 2 hours under heat. After the reaction was completed, the reaction solution was naturally cooled and stirred for 16 hours. And concentrating the reaction solution under reduced pressure to obtain a crude product, namely a compound I-9.
Step 10: compound I-10
At room temperature, dioxane (16L) was charged to a 50L reactor, compound 4a (2.2 kg, 6.12 mol) was added, stirring was continued until clear, and Pd (dppf) Cl was added2·CH2Cl2(263.22 g, 0.32 mol) was charged into the autoclave and purged with nitrogen 3 times. Sodium carbonate (1.37 kg, 12.89 mol) was dissolved in water (5.2 l) to clarify, added to the reaction kettle and heated to 70-75 ℃. N is a radical of2The solution of compound I-9(2.6 kg, 6.45 mol) was added to the reaction vessel (100 ml/min) with a peristaltic pump under protection and stirred for about 2 hours with internal temperature controlled at 70-75 ℃. After the reaction was completed, the reaction solution was cooled to room temperature, the reaction solution was filtered with celite under reduced pressure, and the filter cake was washed with ethyl acetate 2 times (2.5 liters each time). After the filtrate was concentrated under reduced pressure, ethyl acetate (25 l) and saturated brine (20 l) were added thereto, and suction filtration was performed under reduced pressure using celite, and after separating the filtrate, the organic phase was dried over anhydrous sodium sulfate (2.5 kg). The organic phase was transferred to a reaction kettle, to which was added modified silica gel (3.6 kg), heated to 60 ℃ and stirred for 16 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure. To the yellow viscous mass was added tert-butyl methyl ether (4).5 l), is dissolved at 50 ℃, is transferred to a 50 l reaction kettle while hot, and is added with n-heptane (13.5 l) and heated to 60 ℃. And (4) closing the heating, naturally cooling the pulping liquid to 20 ℃, and stirring for 16 hours. The mixture was filtered under reduced pressure, the filter cake was washed with (tert-butyl methyl ether/n-heptane-1/3, 2.5 l) and the filter cake was dried under vacuum for 16 hours to give the crude product. Ethanol/n-heptane (1/10, 12.5 liters) was added to a 50 liter kettle and the crude product was added thereto, warmed to 65 degrees and stirred for 1 hour with constant temperature. And (4) closing the heating, pulping, naturally cooling to 20 ℃, and stirring for 16 hours. Vacuum filtering, washing the filter cake with ethanol/n-heptane (1:10, 2L), and vacuum drying the filter cake for 16 hr to obtain compound I-10.1HNMR(400MHz,CDCl3)δ8.90(d,J=4.8Hz,1H),8.24(br s,1H),8.15(s,1H),7.95(br d,J=3.8Hz,1H),7.63(br d,J=8.2Hz,1H),7.44(s,1H),7.32-7.27(m,1H),6.53(s,1H),6.47(s,1H),4.41(t,J=5.2Hz,2H),4.20-4.07(m,2H),3.94-3.84(m,2H),3.74(t,J=5.2Hz,2H),2.25(s,3H),2.12(ddd,J=2.6,5.0,7.8Hz,1H),1.41(dd,J=4.2,8.0Hz,1H),1.24(s,9H),1.08(t,J=4.4Hz,1H)。
Step 11: compound I-11
At room temperature, adding formic acid (12L) into a 50L reaction kettle, adding the compound I-10(2.4 kg, 4.32 mol), raising the internal temperature to 60-70 ℃, and stirring for 4-5 hours. After completion of the reaction, heating was stopped, the reaction solution was cooled to room temperature, and the reaction solution was concentrated under reduced pressure (45 degrees). Ethyl acetate (20 l) and water (15 l) were added to the crude product, sodium bicarbonate solid was added to adjust the pH of the solution to 4-5, the solution was separated, the organic phase was washed once with saturated brine (10 l), dried over anhydrous sodium sulfate and spin dried. Transferring to an oven for vacuum drying for 16 hours to obtain the compound I-11.1HNMR(400MHz,CDCl3)δ8.87(d,J=4.8Hz,1H),8.41(s,1H),8.17-8.06(m,2H),7.93(br d,J=4.4Hz,1H),7.62-7.52(m,1H),7.46(s,1H),7.32-7.21(m,1H),6.56(s,1H),6.50(s,1H),4.64-4.45(m,4H),4.22-4.03(m,2H),3.94-3.83(m,2H),2.25(s,3H),2.12(ddd,J=2.6,5.0,7.8Hz,1H),1.40(dd,J=4.2,7.8Hz,1H),1.09(t,J=4.4Hz,1H)。
Step 12: a compound of formula (I)
Ethylene glycol dimethyl ether (10.5 liters) was charged to a 50 liter reactor at room temperature. Adding the compound I-11(2.1 kg, 3.98 mol) and stirred until the solution is clear. The temperature is reduced to 5-10 ℃, a pure water (2.1 liters) solution of sodium hydroxide (238.85 g, 5.97 mol) is slowly added, and the mixture is stirred for 0.5 hour. After completion of the reaction, ethyl acetate (20 l) and pure water (10 l) were added to the reaction solution, and the mixture was separated, and the organic phase was washed once with water (10 l) and dried over anhydrous sodium sulfate. The organic phase was rotary evaporated and the resulting solid transferred to an oven and dried in vacuo for 16 h to give the compound of formula (I) (crude).1HNMR(400MHz,CDCl3)δ8.93(d,J=4.8Hz,1H),8.18(s,1H),8.14(s,1H),7.96(br d,J=4.8Hz,1H),7.60(br d,J=8.2Hz,1H),7.52(s,1H),7.32(d,J=8.2Hz,1H),6.63(s,1H),6.57(s,1H),4.54-4.46(m,2H),4.20-4.11(m,2H),4.02-3.96(m,2H),3.96-3.86(m,2H),2.29(s,3H),2.16(ddd,J=2.6,5.0,7.8Hz,1H),1.41(dd,J=4.2,7.9Hz,1H),1.14(t,J=4.4Hz,1H)。
Example 2: preparation of a Single Crystal of the Compound of formula (I)
The experimental method comprises the following steps:
30mg of the compound of the formula (I) was dissolved in 2mL of acetone, and the dissolved solution was put into a centrifuge tube (capacity: 10 mL). 2mL of petroleum ether is slowly added into the solution, the two solvents are obviously layered up and down, the pipe orifice is sealed by a sealing film, and a small evaporation hole is formed. Standing at room temperature for 10 days until most of the solvent is volatilized and crystals are separated out, collecting the crystals, and detecting by single crystal X-ray diffraction (SC-XRD).
The results are shown in tables 1-4:
TABLE 1 Crystal Structure data and measurement parameters for the Compounds of formula (I)
Figure BDA0003154430400000121
Figure BDA0003154430400000131
TABLE 2 atomic coordinates (. times.10) of the crystals of the compound of formula (I)4) And equivalent isotropic shift parameter
Figure BDA0003154430400000132
Figure BDA0003154430400000133
Figure BDA0003154430400000141
TABLE 3 bond length of crystals of the compound of formula (I)
Figure BDA0003154430400000142
And bond angle [ deg ]]
Figure BDA0003154430400000143
Figure BDA0003154430400000151
Figure BDA0003154430400000161
Figure BDA0003154430400000171
TABLE 4 twist angle [ deg ] of crystals of the compound of formula (I)
Figure BDA0003154430400000172
Figure BDA0003154430400000181
Figure BDA0003154430400000191
Figure BDA0003154430400000201
And (4) conclusion: the crystal unit diagram is shown in figure 1 and the results indicate that the compound of formula (I) has the structure N- (3- (2- ((1R,5S) -3-oxabicyclo [3.1.0] hex-1-yl) -6- (2-hydroxyethoxy) pyridin-4-yl) -4-methylphenyl) -2- (trifluoromethyl) isonicotinamide.
Biological test:
experimental example 1: in vitro enzyme Activity assay
Purpose of the experiment:
the inhibitory effect of the compounds on RAF kinase activity is tested.
The experimental steps are as follows:
(1) preparation of compound:
the test compound and the reference compound were diluted to 1 μ M with DMSO and the compounds were diluted in 3-fold gradient to obtain 10 concentration-gradient target plates. ATP concentration was 10. mu.M.
(2) The experimental process comprises the following steps:
1) preparing a buffer solution: 20mM HEPES (pH 7.5),10mM MgCl2,1mM EGTA,0.01%Brij35,0.02mg/ml,BSA,0.1mM Na3VO4,2mM DTT,1%DMSO;
2) Preparing a specified substrate solution by using a newly prepared buffer solution, and adding a corresponding cofactor;
3) adding corresponding RAF kinase enzyme into the substrate solution, and uniformly mixing;
4) adding a DMSO solution of the compound to the above solution with Echo 550;
5) adding into33P-ATP (specific activity 0.01. mu. Ci/. mu.l) elicited reactions, incubated for 2 hours at room temperature;
6) the reaction was labeled with P81 ion exchange paper (Whatman # 3698-915);
7) the filter was washed thoroughly with 0.75% phosphoric acid;
8) the residual radiophosphorylated substrate on the filter paper is detected.
The experimental results are as follows: table 5 provides the inhibitory activity of the compounds of the present invention on RAF kinase.
In vitro enzymatic Activity of the Compounds of Table 5
Figure BDA0003154430400000202
And (4) conclusion: the compound of the invention has obvious inhibitory activity on RAF enzyme.
Experimental example 2: calu-6 (Kras)Q61K) Experiment for antiproliferative Activity
Experimental materials:
1) experimental reagent consumable
Figure BDA0003154430400000203
Figure BDA0003154430400000211
2) Laboratory apparatus
Name (R) Brand goods number
Cell counting plate Refinement of the original
Victor Nivo PerkinElmer
The experimental steps are as follows:
cell inoculation:
(1) cell culture medium: 89% EMEM, 10% fetal bovine serum and 1% penicillin-streptomycin;
(2) removing the original culture medium from the culture flask, digesting the cells with pancreatin, counting, diluting the cell suspension with the culture medium to the cell density of 3.75 × 104Per ml of individual cells;
(3) add 100. mu.L of medium to each well around the cell plate, add 80. mu.L of cell suspension to the other wells, and put 5% CO2And cultured overnight in a 37-degree incubator.
Adding medicine:
gradient dilution and dosing of the compounds with Echo were performed, and then the cell plates were returned to the incubator for three days;
reading the plate, analyzing data:
add CTG and read plate: add 20. mu.L of CellTiterGlo to each well of the cell plate, shake for 10min away from light, read the plate on Victor Nivo.
The experimental results are as follows: table 6 provides the antiproliferative activity of the compounds of the invention on Calu-6 cells.
In vitro Activity of the Compounds of Table 6
Type (B) A compound of formula (I)
Lung cancer Calu-6 cell antiproliferative activity IC50(nM) 600
Anti-proliferative Activity of Colon cancer HCT-116 cells IC50(nM) 1100
Lung cancer Calu-6 cell ERK phosphorylation inhibitory Activity IC50(nM) 490
ERK phosphorylation inhibitory Activity of Colon cancer HCT-116 cells IC50(nM) 180
Experimental example 3: HCT-116 (Kras)G13D) Experiment for antiproliferative Activity
Experimental materials:
1) experimental reagent consumable
Name (R) Brand goods number
Mc' Coy 5A Medium BI-01-075-1ACS
Fetal bovine serum Biosera-FB-1058/500
0.25% Trypsin Source culture-S310 KJ
Double resistance (penicillin, streptomycin) Procell-PB180120
CellTiter Glo Promega-G7573
Cell plate Corning-3610
2) Laboratory apparatus
Name (R) Brand goods number
Cell counting plate Refinement of the original
Victor Nivo PerkinElmer
The experimental steps are as follows:
cell inoculation:
(1) cell culture medium: 89% Mc' Coy 5A, 10% fetal bovine serum and 1% penicillin-streptomycin;
(2) removing the original culture medium from the culture flask, digesting the cells with pancreatin, counting, diluting the cell suspension with the culture medium to the cell density 2.5X 10 required for plating4Per ml of individual cells;
(3) add 100. mu.L of medium to each well around the cell plate, add 80. mu.L of cell suspension to the other wells, and put 5% CO2And cultured overnight in a 37-degree incubator.
Adding medicine:
carrying out gradient dilution and dosing on the compound, and then putting the cell plate back to an incubator for culturing for three days;
reading the plate, analyzing data:
add CTG and read plate: add 20. mu.L of CellTiterGlo to each well of the cell plate, shake for 10min away from light, read the plate on Victor Nivo.
The experimental results are as follows: table 6 provides the antiproliferative activity of the compounds of the invention on HCT-116 cells.
Examples of the experiments4:HCT116(KrasG13D) ERK phosphorylation inhibition assay
Experimental materials:
1. reagent consumable
Figure BDA0003154430400000221
Figure BDA0003154430400000231
2. Main instrument
Instrument for measuring the position of a moving object Manufacturer of the product Model number
Biological safety cabinet AIRTECH BSC-1304IIA2
Carbon dioxide incubator Thermo 311
Cell counter BECKMAN Vi-cellXR
Enzyme-linked immunosorbent assay (ELISA) instrument PerkinElmer Envision
Centrifugal machine Eppendorf Centrifuge 5810R
3. Cellular information
Cell name Source Goods number
HCT116 ATCC ATCC-HTB-132
Experimental procedures and methods:
1) the cells were revived and cultured to logarithmic phase, trypsinized, seeded in 96-well plates, and placed in an incubator for overnight incubation.
2) DMSO dissolved series of gradient compounds were added to 96-well plates and placed back into the incubator for 1 hour incubation.
3) The cell plates were removed, the supernatant removed, and cell lysates (containing 1% blocking peptide) added, incubated and lysed for 30min at room temperature.
4) Transfer 16 μ L of cell lysate to HTRF plate per well followed by 4 μ L of prepared antibody mix.
5) After overnight incubation, plates were read with Envision, fitted curves were obtained according to ratio (ratio of Ex665/Ex615 fluorescence intensity) and EC was calculated according to Graphpad's four-parameter fitting formula Y ═ Bottom + (Top-Bottom)/(1+10^ ((LogEC 50-X): HillSlope))50
The experimental results are as follows: table 6 provides the inhibitory activity of the compounds of the invention on HCT-116ERK phosphorylation.
Experimental example 5: calu-6 (Kras)Q61K) ERK phosphorylation inhibition assay
Experimental materials:
1) reagent consumable
Reagent Brand goods number
High-sensitivity detection kit for human ERK phosphorylated protein Cisbio-64AERPEH
RPMI1640 medium Gibco-22400089
Fetal bovine serum Hyclone-SV30087.03
96HTRF microplate Cisbio-66PL96025
96 micro-porous plate COSTAR-3599
DMSO Sigma-D2650-100mL
0.05%Trypsin-EDTA Gibco-25300-062
2) Main instrument
Instrument for measuring the position of a moving object Manufacturer of the product Model number
Biological safety cabinet AIRTECH BSC-1304IIA2
Carbon dioxide incubator Thermo 311
Cell counter BECKMAN Vi-cellXR
Enzyme-linked immunosorbent assay (ELISA) instrument PerkinElmer Envision
Centrifugal machine Eppendorf Centrifuge 5810R
3) Cellular information
Cell name Source Goods number
Calu6 ATCC ATCC-HTB-56
Experimental procedures and methods:
1) the cells were revived and cultured to logarithmic phase, trypsinized, seeded in 96-well plates, and placed in an incubator for overnight incubation.
2) DMSO dissolved series of gradient compounds were added to 96-well plates and placed back into the incubator for 1 hour incubation.
3) The cell plates were removed and lysed by adding cell lysate (containing 1% blocking peptide) for 30min at room temperature.
4) Transfer 16 μ L of cell lysate to HTRF plate per well followed by 4 μ L of prepared antibody mix.
5) After overnight incubation, plates were read with Envision, fitted curves were obtained according to ratio (ratio of Ex665/Ex615 fluorescence intensity) and EC was calculated according to Graphpad's four-parameter fitting formula Y ═ Bottom + (Top-Bottom)/(1+10^ ((LogEC 50-X): HillSlope))50
The experimental results are as follows: table 6 provides the inhibitory activity of the compounds of the present invention on Calu-6 ERK phosphorylation.
And (4) conclusion: the compound of the formula (I) has good antiproliferative activity and good ERK phosphorylation inhibition activity on Calu-6 cells and HCT-116 cells.
Experimental example 6: a375 (BRAF)V599E) Experiment for antiproliferative Activity
Experimental materials:
1) experimental reagent consumable
Name (R) Brand goods number
EMEM medium Vickers-320-005-CL
Fetal bovine serum Biosera-FB-1058/500
0.25% Trypsin Basal Media-S310KJ
Double resistance (penicillin, streptomycin) Basal Media-S110JV
CellTiter Glo Promega-G7573
Cell plate Corning-3610
2) Laboratory apparatus
Name (R) Brand goods number
EnVision PerkinElmer
The experimental steps are as follows:
cell inoculation:
(1) cell culture medium: 89% EMEM, 10% fetal bovine serum and 1% penicillin-streptomycin;
(2) removing the original culture medium in the cell culture bottle, digesting the cells with pancreatin, counting, and diluting the cell suspension with the culture medium to the cell density of 2000 cells per well required by the plate laying;
(3) add 80. mu.L of cell suspension to each well and put 5% CO2And cultured overnight in a 37-degree incubator.
Adding medicine:
add 20. mu.L of compound working solution to each well in cell plates at 37 ℃ in a 5% CO2 incubator and incubate for an additional 5 days.
Reading the plate, analyzing data:
add CTG and read plate: to each well of the cell plate, 50. mu.L CellTiterGlo was added, shaken for 10min, and detected using EnVision. The experimental results are as follows: table 7 provides the antiproliferative activity of compounds of the invention on a375 cells.
In vitro cell Activity of the Compounds of Table 7
Compound (I) A375 cells IC50(nM) Colo-205 cells IC50(nM)
A compound of formula (I) 269.5 619.5
Experimental example 7: colo-205 (BRAF)V599E) Experiment for antiproliferative Activity
Experimental materials:
1) experimental reagent consumable
Name (R) Brand goods number
RPMI-1640 medium Biological Industries-06-1055-57-1ACS
Fetal bovine serum Biosera-FB-1058/500
0.25% Trypsin Basal Media-S310KJ
Double resistance (penicillin, streptomycin) Basal Media-S110JV
CellTiter Glo Promega-G7573
Cell plate Corning-3610
2) Laboratory apparatus
Name (R) Brand goods number
EnVision PerkinElmer
The experimental steps are as follows:
cell inoculation:
(1) cell culture medium: 89% EMEM, 10% fetal bovine serum and 1% penicillin-streptomycin;
(2) removing the original culture medium in the cell culture bottle, digesting the cells with pancreatin, counting, and diluting the cell suspension with the culture medium to the cell density of 3000 cells per well required by the plate laying;
(3) add 80. mu.L of cell suspension to each well and put 5% CO2And cultured overnight in a 37-degree incubator.
Adding medicine:
add 20. mu.L of compound working solution to each well in cell plates at 37 ℃ in a 5% CO2 incubator and incubate for an additional 3 days.
Reading the plate, analyzing data:
add CTG and read plate: to each well of the cell plate, 50. mu.L CellTiterGlo was added, shaken for 10min, and detected using EnVision.
The experimental results are as follows: table 7 provides the anti-proliferative activity of compounds of the invention on Colo-205 cells.
And (4) conclusion: the compound of the invention has obvious antiproliferative activity on A375 cells and Colo-205 cells.
Experimental example 8: pharmacokinetics studies of single intravenous and oral dosing in mice
This experiment was conducted to investigate the Pharmacokinetic (PK) profile of the compounds of formula (I) in mice following a single intravenous and single oral administration of the compounds.
Sample collection and preparation:
after intravenous or oral administration, animal blood samples were collected and the actual blood sampling time was recorded. Immediately after blood collection, the cells were transferred to labeled centrifuge tubes containing K2-EDTA, and then centrifuged to obtain plasma. The plasma was transferred to a pre-chilled centrifuge tube, snap frozen in dry ice and stored in an ultra low temperature freezer at-70 ± 10 ℃ until LC-MS/MS analysis was performed.
Pharmacokinetic data analysis:
plasma drug concentration data for compounds were processed in a non-compartmental model using pharmacokinetic software. Peak concentration (C)max) And time to peak (T)max) And quantifiable end times, obtained directly from blood concentration-time profiles. The following pharmacokinetic parameters were calculated using the log linear trapezoidal method: half life (T)1/2) Apparent volume of distribution (V)dss) And clearance (Cl), area under the time-plasma concentration curve from point 0 to the end point (AUC)0-last) Initial concentration (C)0)。
The results are shown in Table 8:
TABLE 8 pharmacokinetic parameters of single intravenous and oral administration of the Compounds of the invention in mice
Figure BDA0003154430400000261
And (4) conclusion: the compound of formula (I) is better absorbed in mice by oral administration, has lower clearance rate, longer half-life period and better bioavailability.
Experimental example 9: in vivo pharmacodynamic experiment of human source lung cancer Calu-6 cell subcutaneous xenograft tumor BALB/c nude mouse model
Experimental materials:
1.1 Experimental animals and raising environments
1.1.1 Experimental animals
The species are as follows: mouse
Strain: BALB/c nude mice
Arrival at week: 6-8 weeks old
Sex: female
1.1.2 Breeding Environment
Animals are raised in SPF animal room in IVC (independent air supply system, constant temperature and humidity) cages (3-5 per cage)
Temperature: 20-26 deg.C
Humidity: 40-70 percent
1.2 information on Compounds
Figure BDA0003154430400000271
1.3 tumor tissue or cell information
Cell: human lung cancer Calu-6 cell in vitro culture, adding 0.2Units/mL bovine insulin, 10% fetal calf serum, 5% CO at 37 deg.C into EMEM culture medium2And (5) incubator culture. Passage was performed twice a week with conventional digestion treatment with pancreatin-EDTA. When the saturation degree of the cells is 80-90% and the quantity reaches the requirement, collecting the cells, counting and inoculating.
1.4 other reagent information
Name (R) Manufacturer of the product Goods number Storage conditions
Fetal bovine serum Hyclone SV30087.03 -20℃
Trypsin gibco 25200-072 -20℃
EMEM medium ATCC ATCC30-2003 2-8℃
1.5 Instrument information
Name (R) Manufacturer of the product Model number
Carbon dioxide incubator Saimo Feishale (Thermo Fisher) Heracell240i
Low-temperature high-speed centrifuge Aibende (Eppendorf) 5810R
Analytical balance Sadorius (Sartorius) SECURA225D-1CN
Ordinary balance Changzhou Tianping Instrument and Equipment Co Ltd EL-2KJ
Digital display vernier caliper Sanfeng tea 0~150mm
The experimental method and the steps are as follows:
2.1 tumor cell inoculation
Cell inoculation: 0.2mL of Calu-6 cells (1:1 in matrigel) were subcutaneously inoculated to the right hind-dorsal aspect of each mouse, and the mean tumor volume reached 173mm3The grouped administration is started.
2.2 Experimental animal grouping and dosing regimens
Figure BDA0003154430400000281
Note: 1, the number of mice in each group; 2, administration volume parameter: according to the weight of the mouse, 10 mu L/g. Stopping administration if the body weight is reduced by more than 15% until the body weight is recovered to be within 10%; 3: 0.5% MC (methyl cellulose).
2.3 preparation of the test substances
Figure BDA0003154430400000282
Note 1: the medicine is required to be gently and fully mixed before being administered to animals, wherein the solutol is polyethylene glycol-15-hydroxystearate, and the PEG is polyethylene glycol.
2.4 tumor measurement and Experimental indices
Tumor diameters were measured twice weekly using a vernier caliper. The formula for tumor volume is: v is 0.5a × b2And a and b represent the major and minor diameters of the tumor, respectively.
The tumor suppressor therapeutic effect of the compound was evaluated as TGI (%) or relative tumor proliferation rate T/C (%). Relative tumor proliferation rate T/C (%)TRTV/CRTV×100%(TRTV: treatment group RTV mean; cRTV: negative control RTV mean). Calculating Relative Tumor Volume (RTV) according to the tumor measurement result, wherein the calculation formula is that RTV is Vt/V0In which V is0The resulting tumor volume, V, was measured at the time of group administration (i.e., D0)tTumor volume at a certain measurement, TRTVAnd CRTVThe same day data was taken.
TGI (%), reflecting the rate of tumor growth inhibition. TGI (%) × 100% (1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of administration of the treatment group)/(average tumor volume at the end of treatment of the solvent control group-average tumor volume at the start of treatment of the solvent control group)).
2.5 statistical analysis
Statistical analysis was performed using SPSS software based on RTV data at the end of the experiment. Comparisons between groups were analyzed by one-way ANOVA, and variance (significant differences in F-values) was examined by the Games-Howell method. Significant differences were considered with p < 0.05.
3. Results of the experiment
3.1 inhibition of growth of subcutaneous transplanted tumor of human Lung cancer nude mouse
The experiment evaluates the efficacy of the test substance in a human lung cancer xenograft tumor model, with a solvent control group as a reference. The T/C of the compound (100mg/kg) of the formula (I) of the administration group is 11.4 percent, the TGI is 99.1 percent, and the compound has obvious tumor inhibition effect (P is less than 0.01).
3.2 weight Change
The weight and the state of the mice are not abnormal.
And (4) conclusion: the compound of the invention has obvious inhibition effect on the growth of a tumor-bearing mouse of a human lung cancer Calu-6 cell subcutaneous xenograft tumor model.
Experimental example 10: in vivo pharmacodynamic experiment of BALB/c nude mouse of human colon cancer Colo-205 cell subcutaneous xenograft tumor
Experimental Material
1.1 Experimental animals and raising environments
1.1.1 Experimental animals
The species are as follows: mouse
Strain: BALB/c nude mice
Arrival at week: 6-8 weeks old
Sex: female
1.1.2 Breeding Environment
Animals were kept in IVC (independent air supply system, constant temperature and humidity) cages (3 per cage) in SPF animal houses
Temperature: 20-26 deg.C
Humidity: 40-70 percent
1.2 information on Compounds
Figure BDA0003154430400000291
1.3 tumor tissue or cell information
Cell: human colon cancer Colo-205 cells (ATCC-CCL-222) are cultured in vitro adherent manner by adding 10% fetal bovine serum, 1% penicillin/streptomycin/amphotericin B into RPMI1640 culture medium and 5% CO at 37 DEG C2And (5) incubator culture. Passage was performed twice a week with conventional digestion treatment with pancreatin-EDTA. When the saturation degree of the cells is 80-90% and the quantity reaches the requirement, collecting the cells, counting and inoculating.
1.4 other reagent information
Name (R) Manufacturer of the product Goods number Storage conditions
Fetal bovine serum Cellmax SA211.02 -10℃
RPMI1640 medium gibco 22400-089 2℃-8℃
Anti-Anti Gibco 15240062 -20℃-5℃
Pancreatin Gibco 25200072 -20℃-5℃
PBS Hyclone SH30256.01 15℃-30℃
1.5 Instrument information
Figure BDA0003154430400000292
Figure BDA0003154430400000301
Experimental methods and procedures
2.1 tumor cell inoculation and grouping
Cell inoculation: 0.1mL (5X 10)6Seed) Colo-205 cells were inoculated subcutaneously into each litterThe average tumor volume of the right back of the mouse reaches about 150mm3The grouped administration is started.
Grouping and dosing regimen for laboratory animals
Figure BDA0003154430400000302
Note: n is the number of mice in each group; dosing volume parameters: according to the weight of the mouse, 10 mu L/g. If the weight loss exceeds 15%, stopping administration until the weight is recovered to within 10%.
2.3 preparation of the test substances
Figure BDA0003154430400000303
Note: the drug needs to be gently mixed well before administration to the animal.
2.4 tumor measurement and Experimental indices
Tumor diameters were measured twice weekly using a vernier caliper. The formula for tumor volume is: v is 0.5a × b2And a and b represent the major and minor diameters of the tumor, respectively.
The tumor suppressor therapeutic effect of the compound was evaluated as TGI (%) or relative tumor proliferation rate T/C (%). Relative tumor proliferation rate T/C (%) ═ TRTV/CRTV×100%(TRTV: treatment group RTV mean; cRTV: negative control RTV mean). Calculating Relative Tumor Volume (RTV) according to the tumor measurement result, wherein the calculation formula is that RTV is Vt/V0In which V is0The resulting tumor volume, V, was measured at the time of group administration (i.e., D0)tTumor volume at a certain measurement, TRTVAnd CRTVThe same day data was taken.
TGI (%), reflecting the rate of tumor growth inhibition. TGI (%) × 100% (1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of administration of the treatment group)/(average tumor volume at the end of treatment of the solvent control group-average tumor volume at the start of treatment of the solvent control group)).
2.5 statistical analysis
Statistical analysis was performed using SPSS software based on RTV data at the end of the experiment. Comparisons between groups were analyzed by one-way ANOVA, and variance (significant differences in F-values) was examined by the Games-Howell method. Significant differences were considered with p < 0.05.
3. Results of the experiment
3.1 inhibition of growth of subcutaneous transplanted tumors in nude mice with human Colon cancer
The experiment evaluates the efficacy of the test substance in a human lung cancer xenograft tumor model, with a solvent control group as a reference. The T/C of the compound (30mg/kg QD,100mg/kg QD) of the administration group formula (I) is 68.46% and 36.12% respectively, the TGI is 31.98% and 74.05% respectively, the compound has obvious tumor inhibition effect and better dose-effect relationship. Human colon carcinoma Colo-205 cell subcutaneous xenograft tumor model tumor growth curves of tumor-bearing mice after administration of the compound are shown in FIG. 2.
3.2 weight Change
The weight and the state of the mice are not abnormal. The effect of the test substance on the body weight of the mice is shown in FIG. 3.
And (4) conclusion: the compound has obvious inhibition effect on the growth of tumor-bearing mice of colon cancer Colo-205 cell subcutaneous xenograft tumor models.
Experimental example 11: SD rat oral gavage repeated administration for 4 weeks recovery 4 weeks toxicity test accompanied by pharmacokinetic test
Experimental materials:
1.1 Experimental animals and raising environments
1.1.1 Experimental animals
The species are as follows: rat
Strain: SD rat
Age of animal: about 7 to 9 weeks old
Sex: female and male
1.1.2 Breeding Environment
Temperature: 20-26 deg.C
Humidity: 40-70 percent
The cage is a suspension type PC transparent rat feeding box on a stainless steel feeding rack, male and female animals are separately fed, and each box is not more than 5 animals. The temperature of the animal raising room is controlled to be 20-26 ℃, and the relative humidity is controlled to be 40-70%. The light/dark is controlled for each 12h cycle (this requirement can be temporarily avoided if night operation is encountered).
1.2 information on Compounds
Figure BDA0003154430400000311
1.3 other reagent information
Figure BDA0003154430400000312
Figure BDA0003154430400000321
1.4 Instrument information
Figure BDA0003154430400000322
Experimental methods and procedures
2.1 preparation and preservation
2.1.1 solvent preparation preparing method
1) Measuring 1000mL of pure water into a clean and dry beaker;
2) accurately (not more than +/-1% of theoretical amount) weighing 0.5g of methylcellulose, adding into 1, and stirring on a magnetic stirrer until the methylcellulose is fully dissolved to obtain the required preparation;
3) sealed for 3 months at the temperature of 2-8 ℃.
2.1.2 preparation method of blank auxiliary material preparation
1) Measuring a solvent preparation with a target volume for later use;
2) accurately weighing (not more than +/-1% of theoretical amount) required blank auxiliary materials into a clean and dry mortar, adding a small amount of solvent preparation, grinding until no obvious particles appear on eyes, transferring the mixture into a proper container, rinsing the mortar with the rest standby solvent preparation, and transferring the rinsing solution into the container;
3) and (5) placing the mixture in a magnetic stirrer for stirring for at least 1h until the mixture is uniform visually, thus obtaining the eye-friendly eye-cream.
Theoretical concentration x volume/sample content x auxiliary material content in the sample is high concentration sample preparation theoretical concentration x volume/46.5% × 50%.
2.1.3 method for preparing test preparation
1) Measuring a volume solvent preparation for preparing a target for later use;
2) accurately weighing the required amount of the test sample (not more than +/-1.0% of theoretical amount), adding a small amount of solvent preparation into a clean and dry mortar, grinding until no obvious particles appear on the eye, transferring the mixture into a proper container, rinsing the mortar with the rest of the standby solvent preparation, and transferring the rinsed mortar and rinsing liquid into the container;
3) dispersing with a dispersing machine until the visual uniformity is achieved;
4) stirring in magnetic stirrer for at least 1 hr until it is uniform visually to obtain the desired preparation.
Note: theoretical concentration × volume/content ═ theoretical concentration × volume/46.5%.
2.2 animal grouping and administration
2.2.1 animal groups
Rats were divided into sex groups according to the principles of randomness and weight balance, and the rats were stratified into 8 groups using the Provantis system according to the recent weight of the animals.
2.2.2 animal administration
The administration route is as follows: performing oral gavage;
duration and frequency of administration: the medicine is taken 1 time a day for 28 days continuously, and the medicine is stopped and recovered for 28 days;
requirements before and during administration: the test preparation and the adjuvant preparation are stirred for at least 30min before administration, and are continuously stirred until the administration is finished.
Grouping and dosing regimen for laboratory animals
Figure BDA0003154430400000331
Figure BDA0003154430400000341
Note: 1. animals with the last two animal numbers of 11-15 enter the recovery period.
2. The animal number is 4, thousand represent sex, "1" is male, "2" is female; the last two bits represent the intra-group sequence number.
3. If the animals need to be raised separately due to trauma or other reasons, the records are recorded in original records, see a general evaluation table, and are only stored as the original records.
4. The dosages are based on the formula (I) compound dispersion API.
2.3 Experimental testing
2.3.1 clinical pathology sampling and testing
Blood: all animals were fasted overnight before blood sampling without restriction of drinking water. Blood was taken via the abdominal aorta using a disposable vacuum suction tube. The hematology adopts EDTA-K2An anticoagulation tube, with a blood collection volume of about 1.0 mL; separating gel/coagulant tube is adopted for serum biochemistry/electrolyte, and the blood collection amount is about 3.0 mL; the blood coagulation is conventionally performed by using a sodium citrate anticoagulation tube, and the blood collection amount is about 1.0 mL.
Urine: the rat (single rat) is placed in a clean metabolism cage, a syringe is used for sucking not less than 0.5mL of urine after a certain amount of urine is stored in a collection tube, the urine is transferred to a clean urine collection tube, and information such as collection time, volume and the like of the rat is recorded.
After treatment, the parameters of blood and urine are analyzed by an instrument.
2.3.2 pharmacokinetic sampling and evaluation
Blood samples were collected from animals at each time point on day 1 and day 28 of dosing at approximately 0.2 mL/time point, and 15. mu.L of EDTA-K was added to a 1.5mL centrifuge tube at a concentration of 30mg/mL2Anticoagulant (EDTA-K)2The preparation method comprises the following steps: to prepare 10mL of EDTA-K30 mg/mL2For example, 300mg of EDTA-K was accurately weighed2Put into a clean container, added with 10mL of ultrapure water, shaken up, kept at 2-8 ℃ and sealed for later use).
Measurement by LC-MS/MS methodThe concentration of the compound of formula (I) in plasma is determined and analyzed using Analyst 1.6.3 and Watson LIMS 7.4 software, after which concentration and time curves are drawn and at least the following parameters are calculated using winnonlin 8.1: area under concentration-time Curve (AUC)0-tOr AUC0-∞) Maximum peak concentration (C)max) Time to peak (T)max). Report major parameters (including plasma concentrations at different times, C)max、TmaxAnd AUC), each parameter was calculated for each sex.
2.4 data processing and analysis
Data collected directly by the Provantis 10.2.3 and data recorded back into the Provantis 10.2.3 are processed and analyzed statistically by SAS statistical software built in the Provantis 10.2.3. The viral raw data measured by LC-MS/MS were derived using Analyst 1.6.3 software and further statistically analyzed using Microsoft Excel. And acquiring and processing data by using Agilent OpenLAB CDS 2.4.
The experimental results are as follows:
no obvious abnormality is observed in clinical pathology and pharmacokinetic detection of female and male SD rats in each experimental group, and no abnormality is found in serum biochemical total bilirubin.
And (4) experimental conclusion:
after the compound of the formula (I) is orally administrated to SD rats by gavage, the compound is safe and good, the male MTD is more than or equal to 300mg per kilogram, and the female MTD is more than or equal to 100mg per kilogram. (MTD: maximum tolerated dose)
Experimental example 12: beagle dog oral gavage repeated administration for 4 weeks recovery 4 weeks toxicity test accompanied by pharmacokinetic test
Experimental materials:
1.1 Experimental animals and raising environments
1.1.1 Experimental animals
The species are as follows: dog
Strain: beagle dog
Age of animal: 7-8 months old
Sex: female and male
1.1.2 Breeding Environment
Temperature: 19-26 deg.C
Humidity: 40-70 percent
Animals were housed in a single cage using a stainless steel cage. The temperature of the breeding room is controlled at 19-26 ℃, and the relative humidity is controlled at 40-70%. The light/dark 12h cycle is controlled (this requirement can be temporarily waived if night operation is met).
1.2 information on Compounds
Figure BDA0003154430400000351
1.3 other reagent information
Figure BDA0003154430400000352
1.4 Instrument information
Name (R) Manufacturer of the product Model number
Blood analyzer Siemens medical diagnostic products (Shanghai) Co., Ltd ADVIA 2120i
Full-automatic integrated biochemical analyzer Siemens medical diagnostic products (Shanghai) Co., Ltd Dimension EXL 200
Electrolyte analyzer Madi card medical devices (Suzhou) Inc EasyLyte PLUS
Full-automatic blood coagulation analyzer Heatson Meikang Medical Electronics (Shanghai) Co.,Ltd. CA-1500
Urine analyzer URIT Medical Electronic Co.,Ltd. URIT-500B
Experimental methods and procedures
2.1 preparation and preservation
2.1.1 solvent preparation preparing method
1) Measuring 1000mL of pure water into a clean and dry beaker;
2) accurately (not more than +/-1% of theoretical amount) weighing 0.5g of methylcellulose, adding into 1, and stirring on a magnetic stirrer until the methylcellulose is fully dissolved to obtain the required preparation;
3) sealed for 3 months at the temperature of 2-8 ℃.
2.1.2 preparation method of blank auxiliary material preparation
1) Measuring a solvent preparation with a target volume for later use;
2) accurately weighing (not more than +/-1% of theoretical amount) required blank auxiliary materials into a clean and dry mortar, adding a small amount of solvent preparation, grinding until no obvious particles appear on eyes, transferring the mixture into a proper container, rinsing the mortar with the rest standby solvent preparation, and transferring the rinsing solution into the container;
3) and (5) placing the mixture in a magnetic stirrer for stirring for at least 1h until the mixture is uniform visually, thus obtaining the eye-friendly eye-cream.
Theoretical concentration x volume/sample content x auxiliary material content in the sample is high concentration sample preparation theoretical concentration x volume/46.5% × 50%.
2.1.3 method for preparing test preparation
1) Measuring a volume solvent preparation for preparing a target for later use;
2) accurately weighing the required amount of the test sample (not more than +/-1.0% of theoretical amount), adding a small amount of solvent preparation into a clean and dry mortar, grinding until no obvious particles appear on the eye, transferring the mixture into a proper container, rinsing the mortar with the rest of the standby solvent preparation, and transferring the rinsed mortar and rinsing liquid into the container;
3) dispersing with a dispersing machine until the visual uniformity is achieved;
4) stirring in magnetic stirrer for at least 1 hr until it is uniform visually to obtain the desired preparation.
Note: theoretical concentration × volume/content ═ theoretical concentration × volume/46.5%.
2.2 animal grouping and administration
2.2.1 animal groups
According to the principle of randomness and weight balance, dogs are classified into 4 groups according to the recent weight of animals by using a Provantis system.
2.2.2 animal administration
The administration route is as follows: performing oral gavage;
duration and frequency of administration: 1 dose daily for 4 weeks, 4 weeks after discontinuation of D29;
requirements before and during administration: animals were dosed about 1h after feeding. The preparation is stirred for at least 1h in a room temperature environment before administration, and the stirring is continued during the administration until the administration is finished.
Grouping and dosing
Figure BDA0003154430400000371
Note: 1. animals with last animal number 4, 5 entered recovery.
2. The animal number is 4, thousand represent sex, "1" is male, "2" is female; the last two bits represent the intra-group sequence number.
3. The dosages are based on the formula (I) compound dispersion API.
2.3 Experimental testing
2.3.1 clinical pathology sampling and testing
Blood: all animals were bled before feeding (except for dying animals), without restriction on drinking water. Blood was collected via the forelimb or hindlimb veins with a disposable vacuum suction tube. The hematology adopts EDTA-K2An anticoagulation tube, the blood collection volume is about 1.0 mL; separating gel/coagulant tube is adopted for serum biochemistry/electrolyte, and the blood collection amount is about 3.0 mL; the blood coagulation is conventionally performed by using a sodium citrate anticoagulation tube, and the blood collection amount is about 1.0 mL.
Urine: after collecting fresh urine by using a clean stainless steel urine collecting disc, about 1.0mL of urine is extracted by using a disposable sterile syringe, the urine is transferred into a clean urine collecting tube (identified according to a corresponding label template in the mechanism), and information such as collecting time, volume and the like of the urine is recorded.
After treatment, the parameters of blood and urine are analyzed by an instrument.
2.3.2 pharmacokinetic sampling and evaluation
Blood samples of approximately 1mL EDTA-K were collected from the animals at each time point on day 1 and day 28 of the administration2Anticoagulant blood collection tube.
After determining the concentration of the compound of formula (I) in plasma by analysis using Analyst 1.6.3 and Watson LIMS 7.4 software, concentration and time curves are plotted and at least the following parameters are calculated using WinNonlin 8.1: area under concentration-time Curve (AUC)0-tOr AUC0-∞) Maximum peak concentration (C)max) Time to peak (T)max). Report major parameters (including plasma concentrations at different times, C)max、TmaxAnd AUC), the mean and standard deviation values. The respective parameters were calculated for different sexes.
2.4 data processing and analysis
Data collected directly by the Provantis 10.2.3 and data recorded back into the Provantis 10.2.3 are processed and analyzed statistically by SAS statistical software built in the Provantis 10.2.3. The viral raw data measured by LC-MS/MS were derived using Analyst 1.6.3 software and further statistically analyzed using Microsoft Excel. And acquiring and processing data by using Agilent OpenLAB CDS 2.4.
Results of the experiment
No obvious abnormality is observed in clinical pathology and pharmacokinetic detection of female and male Beagle dogs in each experimental group, and no abnormality is found in serum biochemical total bilirubin.
And (4) experimental conclusion:
after the compound shown as the formula (I) is orally administrated to Beagle dogs by gastric lavage, the compound is safe and good, and the MTD is more than or equal to 100 milligrams per kilogram.

Claims (5)

1. A compound of formula (I) N- (3- (2- ((1R,5S) -3-oxabicyclo [3.1.0] hex-1-yl) -6- (2-hydroxyethoxy) pyridin-4-yl) -4-methylphenyl) -2- (trifluoromethyl) isonicotinamide or a pharmaceutically acceptable salt thereof,
Figure FDA0003154430390000011
2. a pharmaceutical composition comprising the compound N- (3- (2- ((1R,5S) -3-oxabicyclo [3.1.0] hex-1-yl) -6- (2-hydroxyethoxy) pyridin-4-yl) -4-methylphenyl) -2- (trifluoromethyl) isonicotinamide of formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
3. Use of a compound of formula (I) and salts thereof according to claim 1 or a pharmaceutical composition according to claim 2 in the preparation of a RAF kinase inhibitor.
4. A compound of the formula:
Figure FDA0003154430390000012
5. use of a compound according to claim 4 in the preparation of a compound of formula (I)
Figure FDA0003154430390000013
The use of (1).
CN202110772717.8A 2020-07-08 2021-07-08 Biaryl compounds Active CN113912591B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2020106521493 2020-07-08
CN202010652149 2020-07-08
PCT/CN2020/133933 WO2021110141A1 (en) 2019-12-06 2020-12-04 Biaryl compound as pan-raf kinase inhibitor
CNPCT/CN2020/133933 2020-12-04

Publications (2)

Publication Number Publication Date
CN113912591A true CN113912591A (en) 2022-01-11
CN113912591B CN113912591B (en) 2023-10-20

Family

ID=79232910

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110772717.8A Active CN113912591B (en) 2020-07-08 2021-07-08 Biaryl compounds

Country Status (1)

Country Link
CN (1) CN113912591B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114746417A (en) * 2019-12-06 2022-07-12 南京明德新药研发有限公司 Biaryl compounds as inhibitors of Pan-RAF kinase
WO2022253334A1 (en) * 2021-06-04 2022-12-08 南京明德新药研发有限公司 Crystal form of raf kinase inhibitor and preparation method therefor

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104822658A (en) * 2013-06-28 2015-08-05 百济神州有限公司 Fused tricyclic amide compounds as multiple kinase inhibitors
CN105143209A (en) * 2013-03-14 2015-12-09 诺华股份有限公司 Biaryl amide compounds as kinase inhibitors
CN109310761A (en) * 2016-06-10 2019-02-05 诺华股份有限公司 The therapeutical uses of C-RAF inhibitor
WO2019185026A1 (en) * 2018-03-30 2019-10-03 南京明德新药研发有限公司 Glucoside derivatives acting as inhibitors of sglts, and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105143209A (en) * 2013-03-14 2015-12-09 诺华股份有限公司 Biaryl amide compounds as kinase inhibitors
CN104822658A (en) * 2013-06-28 2015-08-05 百济神州有限公司 Fused tricyclic amide compounds as multiple kinase inhibitors
CN109310761A (en) * 2016-06-10 2019-02-05 诺华股份有限公司 The therapeutical uses of C-RAF inhibitor
WO2019185026A1 (en) * 2018-03-30 2019-10-03 南京明德新药研发有限公司 Glucoside derivatives acting as inhibitors of sglts, and use thereof

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114746417A (en) * 2019-12-06 2022-07-12 南京明德新药研发有限公司 Biaryl compounds as inhibitors of Pan-RAF kinase
CN114746417B (en) * 2019-12-06 2023-12-08 齐鲁制药有限公司 Biaryl compounds as Pan-RAF kinase inhibitors
WO2022253334A1 (en) * 2021-06-04 2022-12-08 南京明德新药研发有限公司 Crystal form of raf kinase inhibitor and preparation method therefor

Also Published As

Publication number Publication date
CN113912591B (en) 2023-10-20

Similar Documents

Publication Publication Date Title
JP6215938B2 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
EP2308877B1 (en) Imidazopyridin-2-one derivatives
AU2014356583B2 (en) New octahydro-cyclobuta (1,2-c;3,4-c&#39;)dipyrrol-2-yl
CN113912591B (en) Biaryl compounds
JP7088906B2 (en) FGFR4 inhibitor and its production method and use
JP2023535746A (en) Compounds used as CDK7 kinase inhibitors and their applications
KR20180073682A (en) Pyrimidine Derivatives and Their Use
EP4119559A1 (en) Maleate salts of a b-raf kinase inhibitor, crystalline forms, methods of preparation, and uses therefore
CN113527335A (en) Macrocyclic compound as EGFR inhibitor and application thereof
CN107427523A (en) The targeted therapies of leiomyosarcoma
US20180179189A1 (en) Adipate forms and compositions of biaryl inhibitors of bruton&#39;s tyrosine kinase
CN111788197B (en) Substituted diamino heterocyclic carboxamide compounds, compositions containing the same and uses thereof
EP4269403A1 (en) Aromatic heterocyclic compound, and pharmaceutical composition and application thereof
EP2382214A1 (en) Method of preparing (+)-1,4-dihydro-7-ý(3s,4s)-3methoxy-4-(methylamino)-1-pyrrolidinyl¨-4-oxo-1-(2-thiazolyl)-1,8-naphthyridine-3-carboxylic acid
CN114746417B (en) Biaryl compounds as Pan-RAF kinase inhibitors
CN113717245B (en) EGFR degradation agent containing 2,8, 9-trisubstituted-9H-purine structural fragment, salt and application thereof
WO2021218912A1 (en) Compounds containing benzosultam
JP2023543080A (en) Crystal of pyrrolo heterocyclic derivative and method for producing the same
JP2024501658A (en) Polycyclic IRAK and FLT3 inhibitory compounds and uses thereof
WO2021088839A1 (en) Imidazolidinone compound, preparation method therefor and use thereof
CN106029664A (en) Crystalline form of malate of tyrosine kinase inhibitor and preparation method therefor
CN110256408A (en) A kind of diaryl pyrazole azole compounds and the composition comprising the compound and application thereof
CN115109055B (en) Difunctional compound for EGFR degradation and application thereof
TWI788444B (en) Crystal form of fgfr4 inhibitor and its preparation method thereof
CN110234649B (en) Salts and crystals of FGFR4 inhibitor, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40062726

Country of ref document: HK

TA01 Transfer of patent application right

Effective date of registration: 20230315

Address after: 250100 Xinlu Street 317, Jinan High-tech Zone, Shandong Province

Applicant after: Qilu Pharmaceutical Co.,Ltd.

Address before: 210032 room 218, business office building, No.9 Gaoxin Road, Jiangbei new district, Nanjing City, Jiangsu Province

Applicant before: Nanjing Mingde New Drug Development Co.,Ltd.

TA01 Transfer of patent application right
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant