WO2023274088A1 - 二甲基取代的噻唑并内酰胺类化合物及其应用 - Google Patents

二甲基取代的噻唑并内酰胺类化合物及其应用 Download PDF

Info

Publication number
WO2023274088A1
WO2023274088A1 PCT/CN2022/101283 CN2022101283W WO2023274088A1 WO 2023274088 A1 WO2023274088 A1 WO 2023274088A1 CN 2022101283 W CN2022101283 W CN 2022101283W WO 2023274088 A1 WO2023274088 A1 WO 2023274088A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
reaction
dmso
pharmaceutically acceptable
solution
Prior art date
Application number
PCT/CN2022/101283
Other languages
English (en)
French (fr)
Inventor
李翼
于涛
刘宁
吴成德
陈曙辉
Original Assignee
南京明德新药研发有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to EP22831902.6A priority Critical patent/EP4353730A1/en
Priority to AU2022303895A priority patent/AU2022303895A1/en
Priority to KR1020247002353A priority patent/KR20240024961A/ko
Priority to BR112023027454A priority patent/BR112023027454A2/pt
Priority to CA3224314A priority patent/CA3224314A1/en
Priority to IL309706A priority patent/IL309706A/en
Priority to CN202280043903.0A priority patent/CN117561265A/zh
Publication of WO2023274088A1 publication Critical patent/WO2023274088A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • the invention relates to a class of dimethyl-substituted thiazololactam compounds and their application in the preparation of medicines for treating related diseases. It specifically relates to the compound represented by formula (I) and pharmaceutically acceptable salts thereof.
  • Ras/Raf/MEK/ERK pathway is a classic mitogen activated protein kinase (MAPK) signaling cascade pathway, which is involved in the activation of various growth factors, cytokines, mitogens and hormone receptors Transduction is one of the most important signal transduction pathways controlling cell growth, differentiation and survival.
  • MAPK mitogen activated protein kinase
  • Extracellular regulated protein kinases are the main players and downstream key nodes of the Ras/Raf/MEK/ERK pathway, and they can be found in many human cancers overactivation.
  • ERK as the terminal signaling kinase of this pathway, has not yet found drug-resistant mutations. Therefore, drugs targeting ERK kinase are expected to overcome the problem of drug resistance after treatment with upstream target inhibitors and become a more potential therapeutic strategy. But so far, research on ERK inhibitors is still in the clinical stage, and no ERK inhibitor has been approved as a drug for marketing.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof,
  • R and R are independently selected from H and C 1-3 alkyl, and the C 1-3 alkyl is optionally substituted by 1 , 2 or 3 R a ;
  • each R is independently selected from H, F, Cl, Br, I, and C 1-3 alkyl optionally substituted by 1 , 2 or 3 R;
  • n is selected from 1 and 2;
  • Ring A is selected from pyrazolyl and tetrahydropyranyl optionally substituted with 1, 2 or 3 R;
  • R and R are independently selected from D, F, Cl, Br and I;
  • R d is selected from F, Cl, Br, I, C 1-3 alkyl and C 1-3 alkoxy, and said C 1-3 alkyl and C 1-3 alkoxy are optionally replaced by 1, 2 or 3 R substitutions;
  • R is selected from F, Cl, Br and I.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof,
  • R and R are independently selected from H and C 1-3 alkyl, and the C 1-3 alkyl is optionally substituted by 1 , 2 or 3 R a ;
  • each R is independently selected from H, F, Cl, Br, I, and C 1-3 alkyl optionally substituted by 1 , 2 or 3 R;
  • n is selected from 1 and 2;
  • Ring A is selected from pyrazolyl and tetrahydropyranyl optionally substituted with 1, 2 or 3 R;
  • R and R are independently selected from D, F, Cl, Br and I;
  • R d is selected from F, Cl, Br, I, C 1-3 alkyl and C 1-3 alkoxy, and said C 1-3 alkyl and C 1-3 alkoxy are optionally replaced by 1, 2 or 3 R substitutions;
  • R is selected from F, Cl and Br.
  • R 1 and R 2 are independently selected from H, CH 3 and CH 2 CH 3 , and the CH 3 and CH 2 CH 3 are optionally substituted by 1, 2 or 3 R a , Other variables are as defined herein.
  • R 1 and R 2 are independently selected from H, CH 3 , CHF 2 , CD 3 and CH 2 CH 3 , and other variables are as defined in the present invention.
  • R 4 is independently selected from H, F, Cl, Br, I and CH 3 , and the CH 3 is optionally substituted by 1, 2 or 3 R c , and other variables are as described in the present invention definition.
  • R 4 is independently selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • R d is selected from F, Cl, Br, I, CH 3 and OCH 3 , and the CH 3 and OCH 3 are optionally substituted by 1, 2 or 3 R, and other variables are as in the present invention defined.
  • R d is selected from CH 3 and OCH 3 , and other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from said Optionally substituted with 1, 2 or 3 Rd , other variables are as defined herein.
  • the above-mentioned ring A is selected from Other variables are as defined herein.
  • the above compound or a pharmaceutically acceptable salt thereof is selected from:
  • R 2 and R 4 are as defined in the present invention.
  • the present invention also provides a compound represented by the following formula or a pharmaceutically acceptable salt thereof,
  • the present invention also provides the application of the above compound or a pharmaceutically acceptable salt thereof in the preparation of a medicament for treating solid tumors.
  • the compound of the present invention shows better inhibitory activity to ERK1 and ERK2 enzymes;
  • the compound of the present invention shows better inhibitory activity to HT29 cell proliferation;
  • the compound of the present invention has better solubility under different pH conditions;
  • the compound of the present invention has excellent Pharmacokinetic properties and anti-tumor effect;
  • the compound of the present invention has weak inhibitory effect on hERG potassium channel current, lower risk of cardiotoxicity, and higher safety; the compound of the present invention has moderate to high plasma protein binding.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms, which are suitable for use in contact with human and animal tissues within the scope of sound medical judgment , without undue toxicity, irritation, allergic reaction or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a salt of a compound of the present invention, which is prepared from a compound having a specific substituent found in the present invention and a relatively non-toxic acid or base.
  • base addition salts can be obtained by contacting such compounds with a sufficient amount of base, either neat solution or in a suitable inert solvent.
  • acid addition salts can be obtained by contacting such compounds with a sufficient amount of the acid, either neat solution or in a suitable inert solvent.
  • Certain specific compounds of the present invention contain basic and acidic functional groups and can thus be converted into either base or acid addition salts.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing acid groups or bases by conventional chemical methods.
  • such salts are prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
  • the compounds of the invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)-isomers, (L)-isomers, and their racemic and other mixtures, such as enantiomerically or diastereomerically enriched mixtures, all of which are subject to the present within the scope of the 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.
  • enantiomer or “optical isomer” refer to stereoisomers that are mirror images of each other.
  • cis-trans isomers or “geometric isomers” arise from the inability to rotate freely due to the double bond or the single bond of the carbon atoms forming the ring.
  • diastereoisomer refers to stereoisomers whose molecules have two or more chiral centers and which are not mirror images of the molecules.
  • keys with wedge-shaped solid lines and dotted wedge keys Indicates the absolute configuration of a stereocenter, with a straight solid line bond and straight dashed keys Indicates the relative configuration of the stereocenter, with a wavy line Indicates wedge-shaped solid-line bond or dotted wedge key or with tilde Indicates a straight solid line key or straight dotted key
  • tautomer or “tautomeric form” means that isomers with different functional groups are in dynamic equilibrium at room temperature and are rapidly interconvertible. If tautomerism is possible (eg, in solution), then chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also called prototropic tautomers
  • Valence isomers include interconversions by recombination of some bonding electrons.
  • keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • the terms “enriched in an isomer”, “enriched in an isomer”, “enriched in an enantiomer” or “enantiomerically enriched” refer to one of the isomers or enantiomers
  • the content of the enantiomer is 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%.
  • the terms “isomer excess” or “enantiomeric excess” refer to the difference between the relative percentages of two isomers or two enantiomers. For example, if the content of one isomer or enantiomer is 90% and the other isomer or enantiomer is 10%, then the isomer or enantiomeric 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 enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with chiral auxiliary agents, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide pure desired enantiomer.
  • a diastereoisomeric salt is formed with an appropriate optically active acid or base, and then a diastereomeric salt is formed by a conventional method known in the art. Diastereomeric resolution is performed and the pure enantiomers are recovered. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by the use of chromatography using chiral stationary phases, optionally in combination with chemical derivatization methods (e.g. amines to amino groups formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compounds.
  • compounds may be labeled with radioactive isotopes such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
  • radioactive isotopes such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
  • heavy hydrogen can be used to replace hydrogen to form deuterated drugs.
  • the bond formed by deuterium and carbon is stronger than the bond formed by ordinary hydrogen and carbon.
  • deuterated drugs can reduce toxic side effects and increase drug stability. , enhance the efficacy, prolong the biological half-life of drugs and other advantages. All changes in isotopic composition of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • substituted means that any one or more hydrogen atoms on a specified atom are replaced by a substituent, which may include deuterium and hydrogen variants, as long as the valence of the specified atom is normal and the substituted compound is stable.
  • Oxygen substitution does not occur on aromatic groups.
  • optionally substituted means that it may or may not be substituted, and unless otherwise specified, the type and number of substituents may be arbitrary on a chemically realizable basis.
  • any variable eg, R
  • its definition is independent at each occurrence.
  • said group may optionally be substituted with up to two R, with independent options for each occurrence of R.
  • substituents and/or variations thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of a linking group is 0, such as -(CRR) 0 -, it means that the linking group is a single bond.
  • a substituent can be bonded to any atom on a ring when the bond of a substituent can cross-link two or more atoms on the ring, e.g., structural unit It means that the substituent R can be substituted at any position on cyclohexyl or cyclohexadiene. When the enumerated substituent does not indicate which atom it is connected to the substituted group, this substituent can be bonded through any atom, for example, pyridyl as a substituent can be connected to any atom on the pyridine ring. The carbon atom is attached to the group being substituted.
  • linking group listed does not indicate its linking direction
  • its linking direction is arbitrary, for example,
  • the connecting group L in the middle is -MW-, at this time -MW- can connect ring A and ring B in the same direction as the reading order from left to right to form It can also be formed by connecting loop A and loop B in the opposite direction to the reading order from left to right
  • any one or more sites of the group can be linked to other groups through chemical bonds.
  • connection method of the chemical bond is not positioned, and there is an H atom at the connectable site, when the chemical bond is connected, the number of H atoms at the site will decrease correspondingly with the number of chemical bonds connected to become the corresponding valence group.
  • the chemical bonds that the site connects with other groups can use straight solid line bonds Straight dotted key or tilde express.
  • the straight-shaped solid-line bond in -OCH3 indicates that it is connected to other groups through the oxygen atom in the group;
  • the straight dotted line bond indicates that the two ends of the nitrogen atom in the group are connected to other groups;
  • the wavy lines in indicate that the 1 and 2 carbon atoms in the phenyl group are connected to other groups;
  • the number of atoms in a ring is generally defined as the number of ring members, eg, "5-7 membered ring” means a “ring” with 5-7 atoms arranged around it.
  • C 1-3 alkyl is used to denote a straight or branched chain saturated hydrocarbon group consisting of 1 to 3 carbon atoms.
  • the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups, etc.; it can be monovalent (such as methyl), divalent (such as methylene) or multivalent (such as methine) .
  • Examples of C1-3 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
  • C 1-3 alkoxy denotes those alkyl groups containing 1 to 3 carbon atoms attached to the rest of the molecule through an oxygen atom.
  • the C 1-3 alkoxy group includes C 1-2 , C 2-3 , C 3 and C 2 alkoxy groups and the like.
  • Examples of C 1-3 alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), and the like.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and the methods well known to those skilled in the art Equivalent alternatives, preferred embodiments include but are not limited to the examples of the present invention.
  • the solvent used in the present invention is commercially available.
  • aq stands for water; eq stands for equivalent, equivalent; DCM stands for dichloromethane; PE stands for petroleum ether; DMSO stands for dimethyl sulfoxide; EtOAc stands for ethyl acetate; EtOH stands for ethanol; MeOH stands for Methanol; Cbz stands for benzyloxycarbonyl, which is an amine protecting group; BOC stands for tert-butoxycarbonyl, which is a kind of amine protecting group; rt stands for room temperature; O/N stands for overnight; THF stands for tetrahydrofuran; Boc 2 O stands for di-tert-tert Butyl dicarbonate; TFA stands for trifluoroacetic acid; DIPEA stands for diisopropylethylamine; iPrOH stands for 2-propanol; mp stands for melting point.
  • Figure 1 Tumor growth curves of human melanoma A375 model animals after administration of solvent and WX001;
  • Figure 2 Body weight change rate of human melanoma A375 model animals during administration.
  • Step 1 Synthesis of Compound A-1-2.
  • reaction solution was quenched with saturated ammonium chloride aqueous solution (1750 mL), and adjusted to pH 3-4 with 4N hydrochloric acid (about 2 L).
  • the layers were separated, and the aqueous phase was extracted with ethyl acetate (3000 mL x 2).
  • the organic phase was washed with saturated brine (1500 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was collected, and the filtrate was concentrated under reduced pressure to dryness to obtain a crude product.
  • Step 4 Synthesis of Compound A-1.
  • A-1-4 150g, 481.93mmol, 92.9% purity, 1eq
  • ethanol 750mL
  • hydrobromic acid 1.07kg, 5.30mol, 719.67mL, 40% purity, 11eq
  • dichloromethane 1.5 L
  • ice water 500 mL
  • pH was adjusted to 7-8 with 4N aqueous sodium hydroxide solution (about 1500 mL).
  • reaction solution was quenched with saturated aqueous sodium sulfite (50mL), extracted with dichloromethane (50mLx3), the organic phases were combined, washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was used Concentrate under reduced pressure in a water pump to obtain D-1-2.
  • Step 1 Synthesis of Compound E-1-3.
  • A-1 (70 g, 283.27 mmol, 1 eq), dichloromethane (1400 mL), 4-dimethylaminopyridine (38.07 g, 311.60 mmol, 1.1 eq) and di-tert-butyl dicarbonate ( 123.65g, 566.54mmol, 130.15mL, 2eq).
  • the nitrogen gas was replaced, and the reaction was carried out at 20° C. for 12 hours.
  • water 300 mL was added to the reaction solution, and the organic phase and the aqueous phase were separated.
  • the aqueous phase was extracted 3 times with dichloromethane (400 mL).
  • A-1 500 mg, 2.02 mmol, 1 eq
  • N'N-dimethylformamide 5 mL
  • cesium carbonate 988.88 mg, 3.04 mmol, 1.5 eq
  • WX001-4 451.74 mg, 2.43 mmol, 1.2 eq
  • the nitrogen gas was replaced, and the reaction was carried out at 25° C. for 16 hours.
  • water (20 mL) was added to the reaction liquid, and filtered to obtain a filter cake.
  • the filter cake was dissolved in dichloromethane (10 mL), washed with saturated brine (15 mL x 3), and separated to obtain an organic phase.
  • the crude product was first purified by thin layer chromatography on a silica gel plate, and then subjected to high performance liquid chromatography (column: Waters Xbridge BEH C18 100*25mm*5 ⁇ m; mobile phase: [water (ammonium bicarbonate)-acetonitrile]; B (acetonitrile)% : 20%-50%, 10 minutes) separation and purification to obtain WX003.
  • the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • the crude product was first purified by thin layer chromatography silica gel plate, and then by high performance liquid chromatography (chromatographic column: Waters Xbridge BEH C18 100*30mm*10 ⁇ m; mobile phase: [water (ammonium bicarbonate)-acetonitrile]; B (acetonitrile)% : 25%-55%, 8 minutes) isolated and purified to obtain WX004.
  • Test compounds were dissolved in 100% DMSO to prepare stock solutions of specific concentrations. Compounds were serially diluted in DMSO solution using an Integra Viaflo Assist smart pipette.
  • ERK1 (or ERK2) kinase activity is the ratio of the remaining kinase activity in the test sample to the kinase activity of the control group (DMSO treatment). Curve fitting was performed using Prism (GraphPad software) and IC50 values were calculated.
  • the compound of the present invention shows better inhibitory activity on ERK1 enzyme.
  • the compound of the present invention shows better inhibitory activity on ERK2 enzyme.
  • Test compounds were dissolved in 100% DMSO to make 10 mM stock solutions.
  • the compound of the present invention exhibits better inhibitory activity on HT29 cell proliferation.
  • mice Female BALB/c mice were used as experimental animals, and the blood concentration of the compound was measured after a single administration and the pharmacokinetic behavior was evaluated.
  • the solvent of the intravenous injection group was 5% DMSO+95% (20% HP- ⁇ -CD)
  • the compound to be tested was mixed with an appropriate amount of intravenous injection solvent, vortexed and sonicated to prepare a 0.5mg/mL clear solution, and filtered through a microporous membrane For future use;
  • the vehicle for the oral group is 5% DMSO+95% (20% HP- ⁇ -CD). After mixing the compound to be tested with the vehicle, vortex and sonicate, a 0.3 mg/mL solution is prepared.
  • DMSO dimethyl sulfoxide
  • HP- ⁇ -CD hydroxypropyl- ⁇ -cyclodextrin.
  • C max is the maximum concentration
  • F% is the oral bioavailability
  • DNAUC AUC PO /Dose
  • AUC PO is the oral exposure
  • Dose is the drug dose
  • Vd ss is the volume of distribution
  • Cl is the clearance rate
  • T 1/2 is the half-life.
  • the compounds of the present invention exhibit excellent oral exposure and bioavailability.
  • UV standard solutions of 1 ⁇ M, 20 ⁇ M and 200 ⁇ M are used as standard solutions for solubility experiments.
  • the compound of the present invention has better solubility under different pH conditions.
  • the anti-tumor effect of WX001 was evaluated using human melanoma A375 cell subcutaneous xenograft tumor nude mouse model.
  • Cage made of polycarbonate, volume 375mm x 215mm x 180mm, bedding is corn cob, replaced once a week;
  • Experimental animals are free to eat during the whole experimental period (irradiation sterilization, dry granular food);
  • the animal information card for each cage should indicate the number of animals in the cage, sex, strain, date of receipt, test number of the dosing regimen, group and start date of the test;
  • Animal identification Experimental animals were identified with ear tags.
  • Tumor tissue inoculation and grouping 0.1 mL (5 ⁇ 10 5 cells) of A375 cells were inoculated subcutaneously in the right axilla of each mouse, and when the average volume of the tumor reached 170 mm 3 , the animals were randomly divided into 4 groups, and began to give medicine.
  • the experimental grouping and dosing regimen are shown in Table 7;
  • Vehicle group 5% DMSO+95% (20% HP- ⁇ -CD).
  • Test compound group Weigh the quantitative test compound in the dispensing bottle, add the corresponding volume of DMSO and vortex to obtain a clear solution, add the corresponding volume of 20% HP- ⁇ -CD and vortex to obtain a uniform suspension.
  • Tumor diameter was measured twice a week with a vernier caliper.
  • TGI (%) The antitumor efficacy of the compound is evaluated by TGI (%).
  • WX001 can inhibit tumor growth in a dose-dependent manner after oral administration until the 21st day. Under the three doses of 10mg/kg and 20mg/kg, TGI were 36%, 81% and 104% respectively.
  • the body weight of the experimental animals is used as a reference index for indirect determination of drug toxicity. As shown in Fig. 2, the body weight of all animals in the solvent control group and WX001 group did not decrease significantly, and there was no morbidity or death on the 21st day after administration.
  • WX001 can inhibit tumor growth in a dose-dependent manner at three doses of 5 mg/kg, 10 mg/kg and 20 mg/kg; the body weight of the animals did not decrease significantly during the administration process, and the tolerance was good.
  • the solvent of the intravenous injection group was 5% DMSO+95% (20% HP- ⁇ -CD).
  • the compound to be tested was mixed with an appropriate amount of intravenous injection solvent, vortexed and sonicated to prepare a 0.2 mg/mL clear solution, and filtered through a microporous membrane For future use; the vehicle for the oral group is 5% DMSO+95% (20% HP- ⁇ -CD). After mixing the compound to be tested with the vehicle, vortex and sonicate, a 1 mg/mL solution is prepared.
  • C max is the maximum concentration
  • F% is the oral bioavailability
  • DNAUC AUC PO /Dose
  • AUC PO is the oral exposure
  • Dose is the drug dose
  • Vd ss is the volume of distribution
  • Cl is the clearance rate
  • T 1/2 is the half-life.
  • the compounds of the present invention exhibit excellent oral exposure and bioavailability.
  • the solvent in the intravenous injection group was 5% DMSO+95% (20% HP- ⁇ -CD).
  • the compound to be tested was mixed with an appropriate amount of intravenous injection solvent, stirred and dissolved to prepare a 0.4 mg/mL clear solution, which was filtered through a microporous membrane for use
  • the vehicle of the oral group is 5% DMSO+95% (20% HP- ⁇ -CD). After the compound to be tested is mixed with the vehicle, it is stirred and dissolved to prepare a 0.3 mg/mL solution.
  • DMSO dimethyl sulfoxide
  • HP- ⁇ -CD hydroxypropyl- ⁇ -cyclodextrin.
  • C max is the maximum concentration
  • F% is the oral bioavailability
  • DNAUC AUC PO /Dose
  • AUC PO is the oral exposure
  • Dose is the drug dose
  • Vd ss is the volume of distribution
  • Cl is the clearance rate
  • T 1/2 is the half-life.
  • the compounds of the present invention exhibit excellent oral exposure and bioavailability.
  • the solvent in the intravenous injection group was 5% DMSO+95% (20% HP- ⁇ -CD).
  • the compound to be tested was mixed with an appropriate amount of intravenous injection solvent, stirred and dissolved to prepare a 0.4 mg/mL clear solution, which was filtered through a microporous membrane for use
  • the vehicle of the oral group is 5% DMSO+95% (20% HP- ⁇ -CD). After the compound to be tested is mixed with the vehicle, it is stirred and dissolved to prepare a 0.3 mg/mL solution.
  • DMSO dimethyl sulfoxide
  • HP- ⁇ -CD hydroxypropyl- ⁇ -cyclodextrin.
  • C max is the maximum concentration
  • F% is the oral bioavailability
  • DNAUC AUC PO /Dose
  • AUC PO is the oral exposure
  • Dose is the drug dose
  • Vd ss is the volume of distribution
  • Cl is the clearance rate
  • T 1/2 is the half-life.
  • the compounds of the present invention exhibit excellent oral exposure and bioavailability.
  • CHO-hERG cells were cultured in a 175cm 2 culture flask, and when the cell density grew to 60-80%, the culture medium was removed, washed with 7mL PBS (Phosphate Buffered Saline), and then digested by adding 3mL Detachin. After the digestion is complete, add 7 mL of culture medium to neutralize, then centrifuge, absorb the supernatant, and then add 5 mL of culture medium to resuspend to ensure that the cell density is 2-5 ⁇ 10 6 /mL.
  • PBS Phosphate Buffered Saline
  • Extracellular fluid formulation 140 NaCl, 5KCl, 1CaCl 2 , 1.25MgCl 2 , 10 HEPES and 10 Glucose, adjust the pH to 7.4 with NaOH.
  • Intracellular fluid formulation 140 KCl, 1MgCl 2 , 1CaCl 2 , 10 EGTA and 10 HEPES, adjust the pH to 7.2 with KOH.
  • the single-cell high-impedance sealing and whole-cell pattern formation processes are all automatically completed by the Qpatch instrument.
  • the cell After obtaining the whole-cell recording pattern, the cell is clamped at -80 mV, before giving a 5-second +40 mV depolarization stimulus , give a pre-voltage of -50 mV for 50 milliseconds, then repolarize to -50 mV for 5 seconds, and then return to -80 mV. Apply this voltage stimulus every 15 seconds, record for 2 minutes, give extracellular fluid for 5 minutes, and then start the administration process.
  • the compound concentration starts from the lowest test concentration, and each test concentration is given for 2.5 minutes. After all concentrations are given continuously, give Positive control compound 3M Cisapride. At least 3 cells (n ⁇ 3) were tested for each concentration.
  • the compound 20.00mM mother solution was diluted with DMSO, and 10 ⁇ L of the compound mother solution was added to 20 ⁇ L DMSO solution, and serially diluted 3 times to 6 DMSO concentrations. Take 4 ⁇ L of compounds with 6 DMSO concentrations, add them to 396 ⁇ L of extracellular fluid, dilute 100 times to 6 intermediate concentrations, then take 80 ⁇ L of compounds with 6 intermediate concentrations, add them to 320 ⁇ L of extracellular fluid, 5 times Dilute to the final concentration to be tested. The highest test concentration is 40 ⁇ M, and there are 6 concentrations in total of 40, 13.3, 4.4, 1.48, 0.494, and 0.165 ⁇ M. The DMSO content in the final test concentration did not exceed 0.2%, and this concentration of DMSO had no effect on the hERG potassium channel. Compound preparation is done by Bravo Instruments throughout the dilution process.
  • the experimental data were analyzed by GraphPad Prism 5.0 software.
  • the experimental reagents used were purchased from Sigma, with a purity of >98%
  • the compound of the present invention has weak inhibitory effect on hERG potassium channel current, lower risk of cardiotoxicity and higher safety.
  • test compound To study the binding degree of test compound to human/mouse/rat/dog/monkey plasma albumin.
  • test compound was dissolved in dimethyl sulfoxide (DMSO) to prepare stock solutions with concentrations of 10 mM and 2 mM, respectively.
  • DMSO dimethyl sulfoxide
  • a 40 ⁇ M working solution was prepared by diluting 2 ⁇ L of the stock solution (2 mM) with 98 ⁇ L of DMSO.
  • a 400 ⁇ M working solution of the control compound was prepared by diluting 10 ⁇ L of the stock solution with 240 ⁇ L DMSO.
  • the working solution of the compound (5 ⁇ L) was mixed with the blank matrix (995 ⁇ L) at a ratio of 1:200 to prepare the loaded matrix.
  • test samples an additional matrix-containing sample was transferred to a separate 96-well plate (sample incubation plate) and incubated at 37° C. for 4 h.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

提供一种二甲基取代的噻唑并内酰胺类化合物,及其在制备治疗相关疾病的药物中的应用,具体为式(I)所示化合物及其药学上可接受的盐。

Description

二甲基取代的噻唑并内酰胺类化合物及其应用
本申请主张如下优先权:
CN202110723288.5,申请日:2021年06月28日;
CN202111669920.9,申请日:2021年12月31日;
CN202210693547.9,申请日:2022年06月17日。
技术领域
本发明涉及一类二甲基取代的噻唑并内酰胺类化合物,及其在制备治疗相关疾病的药物中的应用。具体涉及式(I)所示化合物及其药学上可接受的盐。
背景技术
Ras/Raf/MEK/ERK通路是一条经典的有丝分裂原活化蛋白激酶(mitogen activated protein kinase,MAPK)信号级联通路,参与各种生长因子、细胞因子、丝裂原以及激素受体活化后的信号传导,是控制细胞生长、分化和存活最重要的信号传导途径之一。
研究表明,突变或扩增引起的Ras/Raf/MEK/ERK通路异常活化是多种癌症发生的决定因素。在人类肿瘤中,RAS突变发生率约为22%,BRAF突变发生率约为7%,MEK突变发生率约为1%,因此,该通路上的关键节点蛋白己成为癌症治疗的重要靶点(Cancer Discov.2019,9,329-341)。目前,已有多个BRAF抑制剂和MEK1/2抑制剂,以及它们的联用方案,被美国FDA批准用于黑色素瘤、BRAFV600E突变型非小细胞肺癌等癌症的治疗。然而,使用这些上游节点的BRAF和MEK抑制剂后,由于突变或通路重新激活,会快速导致耐药性问题,极大地限制了它们的临床应用。
细胞外调节蛋白激酶(extracellular regulated protein kinases,ERK),特别是ERK1和ERK2激酶,是Ras/Raf/MEK/ERK通路的主要参与者和下游关键节点,在许多人类的癌症中都可发现它们的过度激活。ERK作为该通路的末端信号激酶,目前尚未发现有耐药突变,因此,靶向ERK激酶的药物有望克服上游靶点抑制剂治疗后产生的耐药性问题,成为更具潜力的治疗策略。但迄今为止,关于ERK抑制剂的研究仍处于临床阶段,还没有ERK抑制剂作为药物批准上市。
综上所述,迫切需要研发出安全、有效的ERK抑制剂药物满足肿瘤治疗的需要。
发明内容
本发明提供了式(I)所示化合物或其药学上可接受的盐,
Figure PCTCN2022101283-appb-000001
其中,
R 1和R 2分别独立地选自H和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R a取代;
各R 4独立地选自H、F、Cl、Br、I和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R c取代;
n选自1和2;
环A选自吡唑基和四氢吡喃基,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
R a和R c分别独立地选自D、F、Cl、Br和I;
R d选自F、Cl、Br、I、C 1-3烷基和C 1-3烷氧基,所述C 1-3烷基和C 1-3烷氧基任选被1、2或3个R取代;
R选自F、Cl、Br和I。
本发明提供了式(I)所示化合物或其药学上可接受的盐,
Figure PCTCN2022101283-appb-000002
其中,
R 1和R 2分别独立地选自H和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R a取代;
各R 4独立地选自H、F、Cl、Br、I和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R c取代;
n选自1和2;
环A选自吡唑基和四氢吡喃基,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
R a和R c分别独立地选自D、F、Cl、Br和I;
R d选自F、Cl、Br、I、C 1-3烷基和C 1-3烷氧基,所述C 1-3烷基和C 1-3烷氧基任选被1、2或3个R取代;
R选自F、Cl和Br。
本发明的一些方案中,上述R 1和R 2分别独立地选自H、CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R a取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 1和R 2分别独立地选自H、CH 3、CHF 2、CD 3和CH 2CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 4独立地选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 4独立地选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R d选自F、Cl、Br、I、CH 3和OCH 3,所述CH 3和OCH 3任选被1、2或3个R取代,其他变量如本发明所定义。
本发明的一些方案中,上述R d选自CH 3和OCH 3,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2022101283-appb-000003
所述
Figure PCTCN2022101283-appb-000004
Figure PCTCN2022101283-appb-000005
任选被1、2或3个R d取代,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2022101283-appb-000006
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2022101283-appb-000007
选自
Figure PCTCN2022101283-appb-000008
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2022101283-appb-000009
选自
Figure PCTCN2022101283-appb-000010
其他变量如本发明所定义。
本发明还有一些方案由上述变量任意组合而来。
本发明的一些方案中,上述化合物或其药学上可接受的盐,其选自:
Figure PCTCN2022101283-appb-000011
其中,R 2和R 4如本发明所定义。
本发明还提供了下式所示化合物或其药学上可接受的盐,
Figure PCTCN2022101283-appb-000012
本发明还提供了上述化合物或其药学上可接受的盐在制备治疗实体瘤的药物中的应用。
技术效果
本发明化合物表现出较优的对ERK1和ERK2酶抑制活性;本发明化合物表现出较优的对HT29细胞增殖抑制活性;本发明化合物在不同pH条件下具有较好的溶解度;本发明化合物具有优异的药代动力学 性质和抑瘤效果;本发明化合物对hERG钾通道电流抑制作用弱,心脏毒性风险更低,安全性更高;本发明化合物具有中等到高血浆蛋白结合度。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物接触的方式获得碱加成盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物接触的方式获得酸加成盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
除非另有说明,术语“异构体”意在包括几何异构体、顺反异构体、立体异构体、对映异构体、旋光异构体、非对映异构体和互变异构体。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(+)”表示右旋,“(-)”表示左旋,“(±)”表示外消旋。
除非另有说明,用楔形实线键
Figure PCTCN2022101283-appb-000013
和楔形虚线键
Figure PCTCN2022101283-appb-000014
表示-个立体中心的绝对构型,用直形实 线键
Figure PCTCN2022101283-appb-000015
和直形虚线键
Figure PCTCN2022101283-appb-000016
表示立体中心的相对构型,用波浪线
Figure PCTCN2022101283-appb-000017
表示楔形实线键
Figure PCTCN2022101283-appb-000018
或楔形虚线键
Figure PCTCN2022101283-appb-000019
或用波浪线
Figure PCTCN2022101283-appb-000020
表示直形实线键
Figure PCTCN2022101283-appb-000021
或直形虚线键
Figure PCTCN2022101283-appb-000022
除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括-些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,取代基可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当一个取代基数量为0时,表示该取代基是不存在的,比如-A-(R) 0表示该结构实际上是-A。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基的键可以交叉连接到一个环上的两一个以上原子时,这种取代基可以与这个环上的任意原子相键合,例如,结构单元
Figure PCTCN2022101283-appb-000023
表示其取代基R可在环己基或者环己二烯上的任意一个位置发生取代。当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。
当所列举的连接基团没有指明其连接方向,其连接方向是任意的,例如,
Figure PCTCN2022101283-appb-000024
中连接基团L为-M-W-,此时-M-W-既可以按与从左往右的读取顺序相同的方向连接环A和环B构成
Figure PCTCN2022101283-appb-000025
也可以按照与从左往右的读取顺序相反的方向连接环A和环B构成
Figure PCTCN2022101283-appb-000026
所述连接基团、取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
除非另有规定,当某一基团具有一个或多个可连接位点时,该基团的任意一个或多个位点可以通过化学键与其他基团相连。当该化学键的连接方式是不定位的,且可连接位点存在H原子时,则连接化学键时,该位点的H原子的个数会随所连接化学键的个数而对应减少变成相应价数的基团。所述位点与其他基团连接的化学键可以用直形实线键
Figure PCTCN2022101283-appb-000027
直形虚线键
Figure PCTCN2022101283-appb-000028
或波浪线
Figure PCTCN2022101283-appb-000029
表示。例如-OCH 3中的直形 实线键表示通过该基团中的氧原子与其他基团相连;
Figure PCTCN2022101283-appb-000030
中的直形虚线键表示通过该基团中的氮原子的两端与其他基团相连;
Figure PCTCN2022101283-appb-000031
中的波浪线表示通过该苯基基团中的1和2位碳原子与其他基团相连;
Figure PCTCN2022101283-appb-000032
表示该哌啶基上的任意可连接位点可以通过1个化学键与其他基团相连,至少包括
Figure PCTCN2022101283-appb-000033
Figure PCTCN2022101283-appb-000034
这4种连接方式,即使-N-上画出了H原子,但是
Figure PCTCN2022101283-appb-000035
仍包括
Figure PCTCN2022101283-appb-000036
这种连接方式的基团,只是在连接1个化学键时,该位点的H会对应减少1个变成相应的一价哌啶基。
除非另有规定,环上原子的数目通常被定义为环的元数,例如,“5-7元环”是指环绕排列5-7个原子的“环”。
除非另有规定,术语“C 1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述C 1-3烷基包括C 1-2和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C1-3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
除非另有规定,术语“C 1-3烷氧基”表示通过一个氧原子连接到分子的其余部分的那些包含1至3个碳原子的烷基基团。所述C 1-3烷氧基包括C 1-2、C 2-3、C 3和C 2烷氧基等。C 1-3烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(包括正丙氧基和异丙氧基)等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明所使用的溶剂可经市售获得。
本发明采用下述缩略词:aq代表水;eq代表当量、等量;DCM代表二氯甲烷;PE代表石油醚;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;EtOH代表乙醇;MeOH代表甲醇;Cbz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁氧羰基是一种胺保护基团;r.t.代表室温;O/N代表过夜;THF代表四氢呋喃;Boc 2O代表二叔丁基二碳酸酯;TFA代表三氟乙酸;DIPEA代表二异丙基乙基胺;iPrOH代表2-丙醇;mp代表熔点。
化合物依据本领域常规命名原则或者使用
Figure PCTCN2022101283-appb-000037
软件命名,市售化合物采用供应商目录名称。
说明书附图
图1:人黑色素瘤A375模型动物在分别给予溶剂和WX001后的肿瘤生长曲线;
图2:人黑色素瘤A375模型动物在给药过程中的体重变化率。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
参考例1
Figure PCTCN2022101283-appb-000038
步骤1:化合物A-1-2的合成。
向反应瓶中加入A-1-1(500g,2.12mol,1eq)、水(1875mL)和四氢呋喃(1875mL),抽换氮气后加入氢氧化锂一水合物(97.76g,2.33mol,1.1eq),25℃下混合液反应3小时。反应完毕后,先浓缩去除有机溶剂,再加入冰水(2L),然后再缓慢加入浓度为4N的盐酸溶液(600mL),调节pH至2-3。搅拌20分钟后,过滤,滤饼用水(1L)和乙腈(500mL)洗涤,收集滤饼。滤饼加入乙腈(1L),搅拌0.5小时。过滤,滤饼用乙腈(500mL)洗涤,收集滤饼烘干得到A-1-2。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=13.32(br s,1H),8.46(s,1H)。
步骤2:化合物A-1-3的合成。
向反应瓶中加入A-1-2(175g,823.55mmol,97.9%纯度,1eq)和2-甲基四氢呋喃(1.75L),抽换氮气后降温至-30℃,缓慢滴入二异丙基胺基锂(2M,905.90mL,2.2eq),-30℃再搅拌1小时。随后缓慢滴入丙酮(95.66g,1.65mol,121.09mL,2eq)和2-甲基四氢呋喃(175mL)溶液,-30℃下混合液反应1小时。反应完毕后,反应液用饱和氯化铵水溶液(1750mL)淬灭,4N盐酸(大约2L)调节pH至3-4。分液,水相用乙酸乙酯(3000mL x 2)萃取。有机相用饱和食盐水(1500mL)洗涤,无水硫酸钠干燥,过滤,收集滤液,滤液减压浓缩干得粗品。向粗品中加入甲基叔丁基醚(3.5L),搅拌30分钟,再加入正己烷(3.5L),继续搅拌4小 时。过滤,收集滤饼得到A-1-3。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=13.28(br s,1H),6.67-5.90(br s,1H),1.62(s,6H)。
步骤3:化合物A-1-4的合成。
向反应瓶中加入A-1-3(200g,668.89mmol,89%纯度,1eq)和乙腈(2L),抽换氮气后加入三氟化硼乙醚溶液(265.82g,1.87mol,231.15mL,2.8eq),60℃下混合液反应8小时。反应完毕后,向反应液中加入乙醇(200mL),减压浓缩,得到粗品。随后将粗品缓慢倒入水(2000mL)中,搅拌30分钟,过滤,收集滤饼。滤饼中加入无水乙醇(600mL),搅拌30分钟,过滤,滤饼用乙醇(200mL)洗涤,收集滤饼干燥得A-1-4。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=2.49(s,3H),1.77(s,6H)。
步骤4:化合物A-1的合成。
向反应瓶中加入A-1-4(150g,481.93mmol,92.9%纯度,1eq)和乙醇(750mL),抽换氮气后缓慢滴入氢溴酸(1.07kg,5.30mol,719.67mL,40%纯度,11eq),50℃下混合液反应24小时。反应完毕后,向反应液中加入二氯甲烷(1.5L)、冰水(500mL),再用4N氢氧化钠水溶液(大约1500mL)调节pH至7-8。分液,水相用二氯甲烷(1000mL x 2)萃取得到有机相,有机相用饱和食盐水(1000mL)洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩得粗品。向粗品中加入乙酸乙酯(225mL)和正己烷(225mL),搅拌2小时。过滤,收集滤饼得到A-1。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.88(s,1H),1.51(s,6H)。
参考例2
Figure PCTCN2022101283-appb-000039
步骤1:化合物B-1-2的合成。
向反应瓶中加入氢氧化钠(590.8g,14.8mol,1.05eq)和水(20L)、B-1-1(2000.00g,14.07mol,1eq),然后加入碘甲烷(2495.80g,17.59mol,1.25eq),在25℃下反应2小时。反应完毕后,将6N的冰盐酸水溶液缓慢加入反应瓶中,调节pH至6~7,搅拌0.5小时。过滤,收集滤饼。向滤饼中加入乙腈(500mL),搅拌0.5小时,过滤,收集滤饼,烘干得到B-1-2。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=12.69(br s,1H),7.74(br s,1H),2.45(s,3H),1.86(s,3H)。
步骤2:化合物B-1-3的合成。
25℃下向反应瓶中加入乙腈(15L)、B-1-2(1500.00g,9.60mol,1eq),然后再加入三氯氧磷(1840.00g,12.0mol,1.25eq),缓慢升温至62℃,在62℃下反应12小时。将反应液倒入水(10.5L)中,加入固体碳酸氢钠调节pH至6-7。用乙酸乙酯(10.5L)萃取,分液得有机相。有机相用饱和食盐水(7.5L)洗涤、无水硫酸钠干燥。过滤,滤液减压浓缩得到B-1-3。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.54(s,1H),2.50(s,3H),2.22(s,3H)。
步骤3:化合物B-1的合成。
向反应瓶中加入B-1-3(100g,572.57mmol,1eq)、水(24.76g,1.37mol,24.76mL,2.4eq)和乙腈(1000mL),抽换氮气后依次加入碘化钠(571.59g,3.81mol,6.66eq)、三甲基氯硅烷(186.61g,1.72mol,218.00mL,3eq),20℃下混合液反应14小时。反应完毕后,反应液中依次加入二氯甲烷(800mL)和用水(12000mL)。然后加碳酸氢钠固体调pH至6-7,分液,水相用二氯甲烷(500mL)萃取一次,合并有机相,有机相依次用饱和亚硫酸钠水溶液(500mL)和饱和食盐水(500mL)洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩得粗品。粗品中加入正庚烷(0.5L),搅拌1小时,过滤,收集滤饼得到B-1。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.34(s,1H),2.48(s,3H),2.21(s,3H)。
参考例3
Figure PCTCN2022101283-appb-000040
步骤1:化合物D-1-2的合成。
向干燥的反应瓶中加入乙酸钠(4.54g,55.39mmol,5eq)、单过硫酸氢钾(13.62g,22.16mmol,2eq)和水(46mL)。降温到0℃,加入D-1-1(4.6g,11.08mmol,1eq)、甲醇(46mL)和四氢呋喃(46mL)的溶液,25℃下反应12小时。反应完毕后,反应液用饱和亚硫酸钠水溶液(50mL)淬灭,用二氯甲烷(50mLx3)萃取,合并有机相,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液用水泵减压浓缩得D-1-2。 1H NMR(400MHz,CDCl 3)δ(ppm)=8.66-8.67(d,J=4.60Hz,1H),7.64-7.65(d,J=4.82Hz,1H),3.37(s,3H),1.36-1.57(m,6H),1.33-1.35(m,6H),1.21-1.23(m,6H),0.88-0.95(m,9H)。
步骤2:化合物D-1的合成。
向干燥的反应瓶中加入D-1-2(4.68g,10.46mmol,1eq)、D-1-3(1.22g,12.56mmol,1.2eq)和四氢呋喃 (70mL)。抽换氮气,-30℃滴加六甲基二硅基胺基锂(1M,21.98mL,2.1eq),-30℃反应2小时。反应完毕后,将反应液倒入饱和氯化铵水溶液(100mL)中,用乙酸乙酯(100mL x 3)萃取,合并有机相。有机相用饱和食盐水(100mL)洗涤、无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品通过柱层析纯化得到D-1。 1H NMR(400MHz,CDCl 3)δ(ppm)=8.17(d,J=4.65Hz,1H),7.45(d,J=1.96Hz,1H),6.91(d,J=4.65Hz,1H),6.79(br s,1H),6.31(d,J=1.96Hz,1H),3.78(s,3H),1.43-1.64(m,6H),1.24-1.38(m,6H),1.07-1.14(m,6H),0.89(t,J=7.34Hz,9H)。
参考例4
Figure PCTCN2022101283-appb-000041
步骤1:化合物E-1-3的合成。
向反应瓶中加入叔丁醇钾(4.83g,43.04mmol,2.5eq)和四氢呋喃(16mL)。抽换氮气后缓慢滴入E-1-1(2g,17.22mmol,2.30mL,1eq)、E-1-1(2.55g,34.44mmol,2.77mL,2eq)的四氢呋喃(16mL)溶液,25℃下混合液反应3小时。反应完毕后,浓缩得到E-1-3。
步骤2:化合物E-1-5的合成。
向反应瓶中加入E-1-3(2.24g,17.21mmol,1eq)和异丙醇(140mL),抽换氮气后加入E-1-4(2.62g,34.42mmol,2eq),90℃下混合液反应12小时。反应完毕后,反应液减压浓缩得到粗品。向粗品中加入20mL水,用醋酸调节pH至4,过滤,收集滤饼。滤饼通过柱层析纯化得到E-1-5。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=12.25(br s,2H),7.22(s,1H),2.21(q,J=7.5Hz,2H),1.01(t,J=7.5Hz,3H)。
步骤3:化合物E-1-6的合成。
向反应瓶中加入氢氧化钠(1.34g,33.61mmol,1.05eq)和水(50mL),随后加入化合物E-1-5(5g,32.01 mmol,1eq)。抽换氮气后,降温至10℃,缓慢加入碘甲烷(5.68g,40.01mmol,2.49mL,1.25eq),10℃下混合液反应0.5小时,缓慢升温至25℃继续反应2.5小时。反应完毕后,将反应液降温至0~5℃,然后用6N的盐酸调节pH至7~8,过滤,收集滤饼,滤饼干燥得到E-1-6。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=12.64(br s,1H),7.70(br s,1H),2.45(s,3H),2.29(q,J=7.4Hz,2H),1.06(t,J=7.4Hz,3H)。
步骤4:化合物E-1-7的合成。
向反应瓶中加入E-1-6(7.4g,43.47mmol,1eq)和乙腈(75mL),抽换氮气后缓慢滴入三氯氧磷(8.33g,54.34mmol,5.05mL,1.25eq),62℃下混合液反应2.5小时。反应完毕后,将反应液倒入水(100mL)中,然后用固体碳酸钠调节pH至6~7。水相用乙酸乙酯(50mL x 2)萃取,合并有机相,有机相用饱和食盐水(50mL)洗涤、无水硫酸钠干燥。过滤,滤液减压浓缩得到E-1-7。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.58(s,1H),2.63(q,J=7.5Hz,2H),2.51(s,3H),1.17(t,J=7.5Hz,3H)。
步骤5:化合物E-1的合成。
向反应瓶中加入E-1-7(3.5g,18.55mmol,1eq)和乙腈(40mL),抽换氮气后依次加入碘化钠(18.52g,123.54mmol,6.66eq)、三甲基氯硅烷(6.71g,61.77mmol,7.84mL,3.33eq)和水(802.26mg,44.52mmol,802.26μL,2.4eq),25℃下混合液反应12小时。反应完毕后,反应液中依次加入二氯甲烷(50mL)和水(50mL),再加碳酸氢钠固体调pH至6-7。分液后,水相用二氯甲烷(50mL)萃取,合并有机相。有机相用分别用饱和亚硫酸钠水溶液(50mL)和饱和食盐水(50mL)洗涤、无水硫酸钠干燥。过滤,滤液减压浓缩得粗品。粗品用柱层析纯化得到E-1。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.32(s,1H),2.58-2.52(m,2H),2.48(s,3H),1.13(t,J=7.5Hz,3H)。
实施例1
Figure PCTCN2022101283-appb-000042
合成路线:
Figure PCTCN2022101283-appb-000043
步骤1:WX001-1的合成
向干燥的反应瓶中加入A-1(70g,283.27mmol,1eq)、二氯甲烷(1400mL)、4-二甲氨基吡啶(38.07g,311.60mmol,1.1eq)和二碳酸二叔丁酯(123.65g,566.54mmol,130.15mL,2eq)。抽换氮气,20℃下反应12小时。反应完毕后,向反应液加入水(300mL),分离有机相和水相。水相用二氯甲烷(400mL)萃取3次。合并有机相,用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品用正己烷(140mL)打浆,过滤,收集滤饼干燥得WX001-1。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=1.75(s,6H),1.52(s,9H)。
步骤2:WX001-2的合成
向干燥的反应瓶中加入WX001-1(10g,28.80mmol,1eq)、四氢呋喃(133mL)、氯化锌溶液(0.7M,41.14mL,1eq)和四甲基乙二胺(3.35g,28.80mmol,4.35mL,1eq)。降温至-78℃加入正丁基锂(2.5M,17.28mL,1.5eq),搅拌10分钟。补加正丁基锂(2.5M,5.76mL,0.5eq),继续搅拌10分钟。补加正丁基锂(2.5M,3.46mL,0.3eq),继续在20℃反应1小时,得到反应液1。
将B-1(7.66g,28.80mmol,1eq)、四三苯基膦钯(998.39mg,863.99μmol,0.03eq)和N’N-二甲基甲酰胺(67mL)的混合液升温至50℃,得到反应液2。将反应液1滴加到反应液2中,50℃反应40分钟。反应完 毕后,向反应液中加入0.1M的乙二胺四乙酸二钠水溶液(450mL),搅拌30分钟,过滤,收集滤饼得到粗品。粗品通过柱层析分离纯化得到WX001-2。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.72(s,1H),2.59(s,3H),2.58(s,3H),1.82(s,6H),1.54(s,9H)。
步骤3:WX001-3的合成
在干燥的反应瓶中加入WX001-2(10g,24.60mmol,1eq)、DCM(100mL)和三氟乙酸(36.49g,320.03mmol,23.69mL,13.01eq),20℃反应1小时。反应完毕后,将反应液先浓缩,然后用氯仿(30mL x 3)置换除去残余三氟乙酸,得到WX001-3。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.89(s,1H),8.72(s,1H),2.61(s,3H),2.59(s,3H),1.57(s,6H)。
步骤4:WX001-5的合成
向干燥的反应瓶中加入WX001-3(150mg,489.55μmol,1eq)、N’N-二甲基甲酰胺(1.5mL)、碳酸铯(239.26mg,734.32μmol,1.5eq)和WX001-4(109.30mg,587.46μmol,1.2eq)。抽换氮气,25℃下反应16小时。反应完毕后,向反应液中加入水(10mL),过滤得到滤饼。滤饼溶于二氯甲烷(10mL)中,并用饱和食盐水(15mL)洗涤,分液得有机相。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩干得WX001-5。 1H NMR(400MHz,DMSO-d 6)δ=8.74(s,1H),7.62(t,J=7.8Hz,1H),7.18-7.06(m,2H),4.72(s,2H),2.64(s,3H),2.59(s,3H),2.45(s,3H),1.54(s,6H)。
步骤5:WX001-6的合成
向干燥的反应瓶中加入WX001-5(130mg,315.88μmol,1eq)、乙腈(3mL)、水(1.5mL)和单过硫酸氢钾(388.39mg,631.77μmol,2eq)。抽换氮气,20℃下反应16小时。反应完毕后,向反应液中加入饱和硫代硫酸钠溶液(10mL),随后用二氯甲烷(10mL x 3)萃取。合并有机相,依次用饱和碳酸氢钠水溶液(20mL x2)和饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到WX001-6。 1H NMR(400MHz,DMSO-d 6)δ=9.18(s,1H),7.62(t,J=7.7Hz,1H),7.13(d,J=7.7Hz,2H),4.73(s,2H),3.48(s,3H),2.82(s,3H),2.45(s,3H),1.57(s,6H)。
步骤6:WX001的合成
向干燥的反应瓶中加入WX001-6(140mg,315.64μmol,1eq)、D-1-3(61.31mg,631.28μmol,2eq)、二氯甲烷(1mL)和四氢呋喃(1mL)。抽换氮气,将反应液降温至0℃下,滴加六甲基二硅基胺基锂(1M,599.72μL,1.9eq)。滴加完毕后0℃继续反应2小时。反应完毕后,向反应液中加水(10mL)淬灭反应,随后用二氯甲烷(10mL x 3)萃取。合并有机相,用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得粗品。粗品通过高效液相色谱(色谱柱:Waters Xbridge BEH C18 100*30mm*10μm;流动相:[水(10mM碳酸氢铵)-乙腈];B(乙腈)%:25%-55%,8分钟)纯化得到WX001。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=9.61(s,1H),8.60(s,1H),7.61(t,J=7.69Hz,1H),7.39(d,J=1.88Hz,1H),7.12(dd,J=7.57,3.56Hz,2H), 6.34(d,J=1.88Hz,1H),4.71(s,2H),3.72(s,3H),2.58(s,3H),2.45(s,3H),1.53(s,6H);LCMS m/z:461[M+H]+。
实施例2
Figure PCTCN2022101283-appb-000044
合成路线:
Figure PCTCN2022101283-appb-000045
步骤1:WX002的合成
向干燥的反应瓶中加入WX001-6(180mg,405.82μmol,1eq)、WX002-1(246.29mg,2.43mmol,6eq)和DMSO(1mL)。在100℃下反应12小时。反应完毕后,反应液直接通过高效液相色谱(色谱柱:Phenomenex C18 80*40mm*3μm;流动相:[水(碳酸氢铵)-乙腈];B(乙腈)%:25%-55%,8分钟)纯化得到WX002。 1H NMR(DMSO-d 6,400MHz)δ=8.40(s,1H),7.61(t,J=7.7Hz,1H),7.2-7.4(m,1H),7.12(dd,J=4.8,7.7Hz,2H),4.6-4.8(m,2H),3.8-4.0(m,3H),3.4-3.5(m,2H),2.52(s,3H),2.45(s,3H),1.89(dd,J=1.5,12.0Hz,2H),1.5-1.6(m,2H),1.52(s,6H);LCMS m/z:465[M+H]+。
实施例3
Figure PCTCN2022101283-appb-000046
合成路线
Figure PCTCN2022101283-appb-000047
步骤1:WX003-1的合成
向干燥的反应瓶中加入A-1(500mg,2.02mmol,1eq)、N’N-二甲基甲酰胺(5mL)、碳酸铯(988.88mg,3.04mmol,1.5eq)和WX001-4(451.74mg,2.43mmol,1.2eq)。抽换氮气,在25℃下反应16小时。反应完毕后,向反应液加入水(20mL),过滤得到滤饼。滤饼溶解于二氯甲烷(10mL)中,并用饱和食盐水(15mL x 3)洗涤,分液得有机相。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩干得WX003-1。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=7.60(t,J=7.7Hz,1H),7.09(dd,J=7.7,13.8Hz,2H),4.66(s,2H),2.44(s,3H),1.48(s,6H)。
步骤2:WX003的合成
向干燥的反应瓶中加入WX003-1(150mg,425.84μmol,1eq)、D-1(217.46mg,468.42μmol,1.1eq)和甲苯(3mL)。抽换氮气,然后加入四三苯基膦钯(98.42mg,85.17μmol,0.2eq),加热到110℃反应12小时。反应完毕后,反应液直接减压浓缩得粗品。粗品先通过薄层层析硅胶板纯化,再经高效液相色谱(色谱柱:Waters Xbridge BEH C18 100*25mm*5μm;流动相:[水(碳酸氢铵)-乙腈];B(乙腈)%:20%-50%,10分钟)分离纯化得到WX003。 1H NMR(400MHz,DMSO-d 6)δ=9.79(s,1H),8.69(d,J=5.0Hz,1H),7.61(t,J=7.7Hz,1H),7.54(d,J=5.0Hz,1H),7.40(d,J=1.9Hz,1H),7.11(t,J=6.8Hz,2H),6.33(d,J=1.6Hz,1H),4.71(s,2H),3.72(s,3H),2.45(s,3H),1.53(s,6H);LCMS m/z:447[M+H]+。
实施例4
Figure PCTCN2022101283-appb-000048
合成路线
Figure PCTCN2022101283-appb-000049
步骤1:WX004-2的合成
向干燥的反应瓶中加入WX001-1(500.00mg,1.44mmol,1eq)、四氢呋喃(6.5mL)、氯化锌溶液(0.7M,2.06mL,1eq)和四甲基乙二胺(167.33mg,1.44mmol,217.32μL,1eq)。置换氮气,降温至-78℃,滴加正丁基锂(2.5M,863.99μL,1.5eq)。搅拌10分钟,补加正丁基锂(2.5M,288.00μL,0.5eq)。搅拌10分钟,补加正丁基锂(2.5M,172.80μL,0.3eq)。滴加完毕后于20℃下反应1小时,得到反应液1。
氮气保护下,将E-1(403.38mg,1.44mmol,1eq)、四三苯基膦钯(49.92mg,43.20μmol,0.03eq)和N’N-二甲基甲酰胺(3.5mL)的混合液升温至50℃,随后加入上述反应液1,在50℃下继续反应40分钟。反应完毕后,反应液用饱和氯化铵水溶液(20mL)淬灭,乙酸乙酯(20mL x 3)萃取。合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩干得粗品。粗品通过柱层析分离纯化得到WX004-2。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.79(s,1H),3.14(d,J=7.34Hz,2H),2.60(s,3H),1.82(s,6H),1.54(s,9H), 1.22(t,J=7.40Hz,3H)。
步骤2:WX004-3的合成
向反应瓶中加入WX004-2(200mg,475.57μmol,1eq)和二氯甲烷(5mL)。降温至0℃后,将三氟乙酸(108.45mg,951.14μmol,70.42μL,2eq)加到反应瓶中,在25℃搅拌1小时。反应完毕后,将反应液缓慢倒入饱和碳酸氢钠水溶液(20mL)中,调节pH至7~8。用二氯甲烷(10mL x 2)萃取,合并有机相。有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩干得WX004-3。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.90(s,1H),8.75(s,1H),3.15(q,J=7.40Hz,2H),2.59(s,3H),1.58(s,6H),1.22(t,J=7.40Hz,3H)。
步骤3:WX004-4的合成
向反应瓶中加入WX004-3(225mg,702.18μmol,1eq)、碳酸铯(343.17mg,1.05mmol,1.5eq)和N’N-二甲基甲酰胺(5mL)。抽换氮气后加入WX001-4(156.77mg,842.61μmol,1.13mL,1.2eq),25℃下混合液反应2小时。反应完毕后,将反应液倒入冰水混合物(200mL)中,搅拌0.5小时,过滤得到滤饼。滤饼用甲苯(5mL x 3)共沸浓缩得到WX004-4。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.77(s,1H),7.60-7.64(t,J=7.67Hz,1H),7.11-7.13(d,J=7.67Hz,2H),4.72(s,2H),3.18(q,J=7.38Hz,2H),2.60(s,3H),2.45(s,3H),1.54(s,6H),1.24(t,J=7.45Hz,3H)。
步骤4:WX004-5的合成
向干燥的反应瓶中加入WX004-4(100mg,234.98μmol,1eq)、水(1.5mL)和乙腈(3mL)。抽换氮气后降温至0℃,分批加入单过硫酸氢钾(288.92mg,469.96μmol,2eq),然后25℃下反应12小时。反应完毕后,反应液中加入饱和亚硫酸钠水溶液(10mL)搅拌0.5小时,然后加入乙酸乙酯(10mL x 2)萃取。合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得WX004-5。 1H NMR(400MHz,DMSO-d 6)δ(ppm)=8.77(s,1H),7.62(t,J=7.7Hz,1H),7.12(d,J=7.7Hz,2H),4.72(s,2H),3.18(q,J=7.4Hz,2H),2.60(s,3H),2.45(s,3H),1.54(s,6H),1.24(t,J=7.5Hz,3H)。
步骤5:WX004的合成
将WX004-5(55mg,120.20μmol,1eq)、D-1-3(23.35mg,240.40μmol,2eq)、二氯甲烷(1mL)和四氢呋喃(1mL)加到反应瓶中。置换氮气,降温至0℃。将六甲基二硅基胺基锂(1M,228.38μL,1.9eq)滴加到反应瓶中,在0℃下搅拌1小时。反应完毕后,反应液中加入水(10mL)淬灭反应,然后用乙酸乙酯(10mL x 2)萃取。合并有机相,用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品先通过薄层层析硅胶板纯化,再通过高效液相色谱(色谱柱:Waters Xbridge BEH C18 100*30mm*10μm;流动相:[水(碳酸氢铵)-乙腈];B(乙腈)%:25%-55%,8分钟)分离纯化得到WX004。 1H NMR(DMSO-d 6,400MHz)δ(ppm)=9.64(s,1H),8.63(s,1H),7.61(t,J=7.7Hz,1H),7.39(d,J=1.8Hz,1H),7.12(d,J=7.8Hz,2H),6.34(d,J=1.8Hz,1H),4.71(s,2H),3.72(s,3H),3.12(q,J=7.3Hz,2H),2.45(s,3H),1.53(s,6H),1.22(t, J=7.4Hz,3H);LCMS m/z:475[M+H]+。
实验例一、体外酶活性测试
1.实验目的:
测量化合物抑制ERK1和ERK2激酶活性的能力。
2.实验缓冲液:
20mM Hepes(pH 7.5),10mM MgCl 2,1mM乙二醇双(2-氨基乙基醚)四乙酸(EGTA),0.02%Brij35,0.02mg/mL牛血清白蛋白(BSA),0.1mM Na 3VO 4,2mM二硫苏糖醇(DTT),1%DMSO。
3.化合物处理:
将测试化合物溶于100%DMSO中,配制成特定浓度的母液。利用Integra Viaflo Assist智能移液器将化合物连续稀释在DMSO溶液中。
4.实验方法:
a)在新制备的反应缓冲液中配置底物MBP;
b)将ERK1(或ERK2)激酶加入到上述MBP溶液中并轻轻混合;
c)运用超声技术(Echo550;纳升范围)将溶于100%DMSO中的化合物加入到激酶反应体系中,在室温下孵育20分钟;
d)将 33P-ATP(特定浓度10μCi/μL)加入到反应体系中,此时开始发生反应;
e)在室温下孵育2小时;
f)通过过滤--结合方法检测放射性的量;
g)ERK1(或ERK2)激酶活性计算方式为测试样品中剩余激酶活性占对照组(二甲基亚砜处理)激酶活性的比值。使用Prism(GraphPad软件)进行曲线拟合并计算IC 50值。
5.实验结果见表1和表2:
表1 ERK1酶活性测试结果
Figure PCTCN2022101283-appb-000050
结论:本发明化合物表现出较优的对ERK1酶抑制活性。
表2 ERK2酶活性测试结果
Figure PCTCN2022101283-appb-000051
结论:本发明化合物表现出较优的对ERK2酶抑制活性。
实验例二、体外细胞增殖抑制实验
1.实验目的:
测量化合物抑制HT29肿瘤细胞增值的能力。
2.化合物处理:
将测试化合物溶于100%DMSO中,配制成10mM的母液。
3.实验步骤与方法:
a)开启生物安全柜紫外灯,倒计时30分钟;
b)37℃水浴锅中,预热RPMI1640培养基和胰酶;
c)紫外照射完毕,开启生物安全柜,将预热培养基,胰酶,磷酸缓冲盐溶液(PBS)等用酒精擦拭并放入生物安全柜中;
d)将HT29细胞从培养箱中取出,在生物安全柜中去除旧培养基,加入10毫升PBS,轻轻摇晃,并去除PBS;
e)加入预热0.25%胰酶1.5毫升,水平晃动培养瓶,使其均匀覆盖到底部的细胞,置培养箱中2分钟;
f)用完全培养基终止细胞消化,并吹打至均匀的细胞悬液计数;
g)根据细胞计数结果,调整细胞悬液密度为1500细胞每孔,50微升每孔进行种板;
h)将化合物母液连续稀释在DMSO溶液中,并使用Tecan将化合物加入细胞板中;
i)将加过化合物的细胞板和CellTiterGlo放到室温平衡,后加CellTiterGlo 25微升至每孔,震荡1-2分钟,静置10分钟后检测信号值,并用XL-Fit分析数据,计算各化合物的IC 50
4.实验结果见表3:
表3体外细胞活性测试结果
Figure PCTCN2022101283-appb-000052
Figure PCTCN2022101283-appb-000053
结论:本发明化合物表现出较优的对HT29细胞增殖抑制活性。
实验例三、小鼠体内PK研究
1.实验目的:
以雌性BALB/c小鼠为受试动物,单次给药后测定化合物血药浓度并评估药代动力学行为。
2.实验操作:
选择健康成年雌性BALB/c小鼠4只,2只为静注组,2只为口服组。静注组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与适量静注溶媒混合,涡旋并超声,制备得到0.5mg/mL澄清溶液,微孔滤膜过滤后备用;口服组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与溶媒混合后,涡旋并超声,制备得到0.3mg/mL溶液。小鼠1mg/kg静脉给药或3mg/kg口服给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
注DMSO:二甲基亚砜;HP-β-CD:羟丙基-β-环糊精。
3.实验结果见表4:
表4化合物PK测试结果
Figure PCTCN2022101283-appb-000054
注:C max为最大浓度;F%为口服生物利用度;DNAUC=AUC PO/Dose,AUC PO为口服暴露量,Dose为药物剂量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期。
结论:本发明化合物展现了优良的口服暴露量和生物利用度。
实验例四、溶解度研究
1.实验目的:
测定化合物的溶解度,评估化合物的溶解性。
2.测试溶液:
1)缓冲液A(pH 2.0):50mM磷酸盐缓冲液,pH 2.0;缓冲液B(pH 6.5):50mM磷酸盐缓冲液,pH 6.5;缓冲液C液(pH 7.4):50mM磷酸盐缓冲液,pH 7.4;
2)标准溶液的制备:
a)将50%的乙腈溶液和50%的缓冲溶液混合,得到稀释液;
b)10mM(10μL/化合物)化合物储备液加入稀释液(490μL/化合物)混合为200μM的检测标准溶液;
c)以10倍和200倍量的稀释液稀释200μM的紫外检测标准液,得到20μM,1μM的紫外标准溶液;
d)1μM、20μM、200μM的紫外标准溶液作为溶解性实验的标准溶液。
3.实验方法:
a)DMSO溶解化合物配制成10mM的储配液。盐酸胺碘酮,卡马西平,氯霉素作为溶解度实验的对照;
b)被测试化合物和对照的储备液(各10μL)放入96孔板中,分别加入三种不同的溶解介质(缓冲液A,B,C)490μL,对应溶解度溶液的pH分别为2.0,6.5和7.4。实验化合物的理论最大浓度为200μM,含DMSO 2%;
c)在室温(25±2℃)下以每分钟600转的转速,在摇床中振荡24小时;
d)吸取200μL的溶液于96孔板真空抽滤装置中抽滤后取至新96孔板中作为测试样品;
e)用HPLC-UV测试化合物浓度,HPLC条件如表5:
表5 HPLC条件
Figure PCTCN2022101283-appb-000055
f)从低浓度到高浓度(1μM,20μM,200μM)注入3个紫外标准液至HPLC中,然后注入待测化合物的测试样品;
g)对紫外色谱峰进行积分,计算样品的溶解度。
4.实验结果见表6:
表6化合物溶解度测试结果
Figure PCTCN2022101283-appb-000056
结论:本发明化合物在不同pH条件下具有较好的溶解度。
实验例五、人黑色素瘤A375小鼠模型体内药效研究
1.实验目的:
使用人黑色素瘤A375细胞皮下异种移植肿瘤裸小鼠模型,评价WX001的抗肿瘤作用。
2.实验动物:
种属:小鼠
品系:BALB/c裸小鼠
周龄:6-8周龄
性别:雌性
体重:18-22克
供应商:维通利华实验动物技术有限公司
3.饲养环境:
动物在SPF级动物房以IVC(独立送风系统,恒温恒湿)笼具饲养(每笼3只),温度:20-26℃,湿度:40-70%;
笼具:以聚碳酸酯制成,体积375mm x 215mm x 180mm,垫料为玉米芯,每周更换一次;
食物:实验动物在整个实验阶段中可自由进食(照射灭菌,干颗粒状食物);
饮水:实验动物可自由饮用灭菌水;
笼具标识:每笼动物信息卡应注明笼内动物数目,性别,品系,接收日期,给药方案实验编号,组别以及实验开始日期;
动物标识:实验动物以耳标进行标识。
4.实验内容:
1)实验细胞及培养:人黑色素瘤A375细胞体外单层培养,培养条件为DMEM培养基中加10%胎牛血清,37℃5%CO 2孵箱培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数,接种;
2)肿瘤组织接种及分组:0.1mL(5×10 5个)A375细胞皮下接种于每只小鼠的右侧腋下,肿瘤平均体积达到170mm 3时,将动物随机分为4组,开始给药。实验分组和给药方案见表7;
表7实验动物分组及给药方案
Figure PCTCN2022101283-appb-000057
Figure PCTCN2022101283-appb-000058
3)实验动物日常观察:本实验方案的拟定及任何修改均通过了实验动物管理与使用委员会(IACUC)的评估核准。实验动物的使用及福利遵照国际实验动物评估和认可委员会(AAALAC)的规定执行。每天监测动物的健康状况及死亡情况,例行检查包括观察肿瘤生长和药物治疗对动物日常行为表现的影响如行为活动,摄食摄水量(仅目测),体重变化(每周测量两次体重),外观体征或其它不正常情况。基于各组动物数量记录了组内动物死亡数和副作用。
4)受试物的配制
a)溶媒组:5%DMSO+95%(20%HP-β-CD)。
b)待测化合物组:称量定量的受试化合物于配药瓶内,加入相应体积的DMSO后涡旋,得到澄清溶液,在加入相应体积的20%HP-β-CD后涡旋,得到均一混悬液。
5)肿瘤测量和实验指标:
a)每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:TV=1/2×a×b 2,a和b分别表示肿瘤的长径和短径;
b)化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)={[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)]/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)}×100%。
5.实验结果:
1)如表8和图1所示,在人黑色素瘤A375细胞皮下异种移植肿瘤裸小鼠模型上,口服给药至第21天,WX001能够剂量依赖性地抑制肿瘤生长,在5mg/kg、10mg/kg和20mg/kg三个剂量下,TGI分别为36%、81%和104%。
2)实验动物的体重作为间接测定药物毒性的参考指标。如图2所示,给药至第21天时,溶剂对照组和WX001组所有动物的体重均未有明显下降,无发病或死亡现象。
表8小鼠A375模型体内药效实验结果
药物 TGI
WX001(5mg/kg,PO,BID) 36%
WX001(10mg/kg,PO,BID) 81%
WX001(20mg/kg,PO,BID) 104%
实验结论:WX001在5mg/kg、10mg/kg和20mg/kg三个给药剂量下,能够剂量依赖性地抑制肿瘤生长;给药过程中动物的体重未见明显下降,耐受性好。
实验例六、SD大鼠体内PK研究
1.实验目的:
以雄性SD大鼠为受试动物,单次给药后测定化合物血药浓度并评估药代动力学行为。
2.实验操作:
选择健康成年雄性SD大鼠6只,3只为静注组,3只为口服组。静注组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与适量静注溶媒混合,涡旋并超声,制备得到0.2mg/mL澄清溶液,微孔滤膜过滤后备用;口服组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与溶媒混合后,涡旋并超声,制备得到1mg/mL溶液。SD大鼠1mg/kg静脉给药或10mg/kg口服给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。注DMSO:二甲基亚砜;HP-β-CD:羟丙基-β-环糊精。
3.实验结果见表9:
表9化合物PK测试结果
Figure PCTCN2022101283-appb-000059
注:C max为最大浓度;F%为口服生物利用度;DNAUC=AUC PO/Dose,AUC PO为口服暴露量,Dose为药物剂量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期。
结论:本发明化合物展现了优良的口服暴露量和生物利用度。
实验例七、食蟹猴体内PK研究
1.实验目的:
以雄性食蟹猴为受试动物,单次给药后测定化合物血药浓度并评估药代动力学行为。
2.实验操作:
选择健康成年雄性食蟹猴5只,2只为静注组,3只为口服组。静注组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与适量静注溶媒混合,搅拌溶解,制备得到0.4mg/mL澄清溶液,微孔滤膜过滤后备用;口服组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与溶媒混合后,搅拌溶解,制备得到0.3mg/mL溶液。食蟹猴1mg/kg静脉给药或3mg/kg口服给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
注DMSO:二甲基亚砜;HP-β-CD:羟丙基-β-环糊精。
3.实验结果见表10:
表10化合物PK测试结果
Figure PCTCN2022101283-appb-000060
注:C max为最大浓度;F%为口服生物利用度;DNAUC=AUC PO/Dose,AUC PO为口服暴露量,Dose为药物剂量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期。
结论:本发明化合物展现了优良的口服暴露量和生物利用度。
实验例八、比格犬体内PK研究
1.实验目的:
以雄性比格犬为受试动物,单次给药后测定化合物血药浓度并评估药代动力学行为。
2.实验操作:
选择健康成年雄性比格犬5只,2只为静注组,3只为口服组。静注组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与适量静注溶媒混合,搅拌溶解,制备得到0.4mg/mL澄清溶液,微孔滤膜过滤后备用;口服组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与溶媒混合后,搅拌溶解,制备得到0.3mg/mL溶液。食蟹猴1mg/kg静脉给药或3mg/kg口服给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
注DMSO:二甲基亚砜;HP-β-CD:羟丙基-β-环糊精。
3.实验结果见表11:
表11化合物PK测试结果
Figure PCTCN2022101283-appb-000061
注:C max为最大浓度;F%为口服生物利用度;DNAUC=AUC PO/Dose,AUC PO为口服暴露量,Dose为药物剂量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期。
结论:本发明化合物展现了优良的口服暴露量和生物利用度。
实验例九、hERG测试
1.实验目的:
使用全自动膜片钳方法测试化合物对hERG钾通道(human Ether-a-go-go Related Gene potassium channel)电流的影响。
2.实验方法:
2.1细胞准备
CHO-hERG细胞培养于175cm 2培养瓶中,待细胞密度生长到60~80%,移走培养液,用7mL PBS(Phosphate Buffered Saline磷酸盐缓冲液)洗一遍,然后加入3mL Detachin消化。待消化完全后加入7mL培养液中和,然后离心,吸走上清液,再加入5mL培养液重悬,以确保细胞密度为2~5×10 6/mL。
2.2溶液配制
细胞外液配方(mM):140 NaCl,5KCl,1CaCl 2,1.25MgCl 2,10 HEPES and 10 Glucose,用NaOH调节pH至7.4。
细胞内液配方(mM):140 KCl,1MgCl 2,1CaCl 2,10 EGTA and 10 HEPES,用KOH调节pH至7.2。
2.3电生理记录过程
单细胞高阻抗封接和全细胞模式形成过程全部由Qpatch仪器自动完成,在获得全细胞记录模式后,细胞钳制在-80毫伏,在给予一个5秒的+40毫伏去极化刺激前,先给予一个50毫秒的-50毫伏前置电压,然后复极化到-50毫伏维持5秒,再回到-80毫伏。每15秒施加此电压刺激,记录2分钟后给予细胞外液记录5分钟,然后开始给药过程,化合物浓度从最低测试浓度开始,每个测试浓度给予2.5分钟,连续给完所有浓度后,给予阳性对照化合物3M Cisapride。每个浓度至少测试3个细胞(n≥3)。
2.4化合物准备
将化合物20.00mM母液用DMSO进行稀释,取10μL化合物母液加入至20μL DMSO溶液中,3倍连续稀释至6个DMSO浓度。分别取4μL 6个DMSO浓度的化合物,加入至396μL的细胞外液中,100倍稀释至6个中间浓度,再分别取80μL的6个中间浓度化合物,加入至320μL的细胞外液中,5倍稀释至需要测试的最终浓度。最高测试浓度为40μM,依次分别为40,13.3,4.4,1.48,0.494,0.165μM共6个浓度。最终测试浓度中的DMSO含量不超过0.2%,此浓度的DMSO对hERG钾通道没有影响。化合物准备由Bravo仪器完成整个稀释过程。
2.5数据分析
实验数据由GraphPad Prism 5.0软件进行分析。
2.6质量控制
环境:湿度20~50%,温度22~25℃
试剂:所用实验试剂购买于Sigma公司,纯度>98%
报告中的实验数据必须满足以下标准:
全细胞封接阻抗>100MΩ
尾电流幅度>300pA
药理学参数:
多浓度Cisapride对hERG通道的抑制效应设为阳性对照。
3.实验结果见表12:
表12化合物hERG测试结果
化合物 IC50(μM)
WX001 >40
结论:本发明化合物对hERG钾通道电流抑制作用弱,心脏毒性风险更低,安全性更高。
实验例十、血浆蛋白结合度测试(PPB)
1.实验目的:
研究受试化合物与人/小鼠/大鼠/犬/猴血浆白蛋白的结合度。
2.实验操作:
1)基质准备:实验当天,将血浆在冷水中解冻,并以3220rpm的速度离心5min,以去除所有血块。测量得到的血浆的pH值,并根据需要使用1%的磷酸或1N的氢氧化钠将其pH调整到7.4±0.1。
2)测试化合物的稀释步骤:测试化合物溶解在二甲基亚砜(DMSO)中,以制备浓度分别为10mM和2mM的原液。用98μL DMSO稀释2μL原液(2mM),制得40μM的工作溶液。用240μL DMSO稀释10μL原液,制得400μM对照化合物的工作溶液。将化合物的工作溶液(5μL)与空白基质(995μL)按1∶200的比例混合均匀以制备负载基质。
3)分析步骤:
a)将等量的30μL负载基质(n=2)转移至样品采集板,制备待测时间0(T0)样品用于残留测定。样品立即与相应的空白缓冲液进行匹配,最终体积为60μL,每孔中血浆与缓冲液的体积比为1∶1。然后,测试化合物的T0样品分别加入60μL的4%H 3PO 4的H 2O和480μL含有内标物的终止液。然后将它们与其他样品一起储存在2-8℃下等待进一步处理。
b)将剩余的血浆样品放在37±1℃的二氧化碳培养箱预培养30min。准备无蛋白样品(F样品),负载基质的样品(230μL)都被转移到聚碳酸酯管(n=2)中,并在37℃和155000×g(35000rpm)条件下超速离心4h。
c)为了制备T样品(测试样品),额外一份含基质样品转移到单独的96孔板(样品培育板)上,并在37℃下培育4h。
d)离心结束后,从上清液第二层(上层以下)取30μL的无蛋白样品和30μL的T样品转移到新的样品收集板中。每个样品与相应的空白缓冲液或基质混合,最终体积为60μL,基质∶缓冲液体积比1∶1。在所有样品加入60μL 4%的H 3PO 4水溶液和480μL的终止溶液(含内标)。混合物在转速4000rpm下离心20min,取各样品上清液100μL进行LC-MS/MS分析。
3.实验结果见表13:
表13化合物血浆蛋白结合度测试结果
化合物 血浆蛋白结合率(unbound%)
  小鼠 大鼠
WX001 6.1% 7.2% 3.8% 4.1% 2.6%
结论:本发明化合物具有中等到高血浆蛋白结合度。

Claims (14)

  1. 式(I)所示化合物或其药学上可接受的盐,
    Figure PCTCN2022101283-appb-100001
    其中,
    R 1和R 2分别独立地选自H和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R a取代;
    各R 4独立地选自H、F、Cl、Br、I和C 1-3烷基,所述C 1-3烷基任选被1、2或3个R c取代;
    n选自1和2;
    环A选自吡唑基和四氢吡喃基,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
    R a和R c分别独立地选自D、F、Cl、Br和I;
    R d选自F、Cl、Br、I、C 1-3烷基和C 1-3烷氧基,所述C 1-3烷基和C 1-3烷氧基任选被1、2或3个R取代;
    R选自F、Cl、Br和I。
  2. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 1和R 2分别独立地选自H、CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R a取代。
  3. 根据权利要求2所述化合物或其药学上可接受的盐,其中,R 1和R 2分别独立地选自H、CH 3、CHF 2、CD 3和CH 2CH 3
  4. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 4独立地选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代。
  5. 根据权利要求4所述化合物或其药学上可接受的盐,其中,R 4独立地选自H、F、Cl、Br、I和CH 3
  6. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R d选自F、Cl、Br、I、CH 3和OCH 3,所述CH 3和OCH 3任选被1、2或3个R取代。
  7. 根据权利要求6所述化合物或其药学上可接受的盐,其中,R d选自CH 3和OCH 3
  8. 根据权利要求1所述化合物或其药学上可接受的盐,其中,环A选自
    Figure PCTCN2022101283-appb-100002
    所述
    Figure PCTCN2022101283-appb-100003
    任选被1、2或3个R d取代。
  9. 根据权利要求8所述化合物或其药学上可接受的盐,其中,环A选自
    Figure PCTCN2022101283-appb-100004
  10. 根据权利要求1所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2022101283-appb-100005
    选自
    Figure PCTCN2022101283-appb-100006
  11. 根据权利要求10所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2022101283-appb-100007
    选自
    Figure PCTCN2022101283-appb-100008
  12. 根据权利要求1~5任意一项所述化合物或其药学上可接受的盐,其选自
    Figure PCTCN2022101283-appb-100009
    其中,
    R 2如权利要求1~3任意一项所定义;
    R 4如权利要求1、4或5任意一项所定义。
  13. 下式所示化合物或其药学上可接受的盐,
    Figure PCTCN2022101283-appb-100010
  14. 根据权利要求1~13任意一项所述化合物或其药学上可接受的盐在制备治疗实体瘤的药物中的应用。
PCT/CN2022/101283 2021-06-28 2022-06-24 二甲基取代的噻唑并内酰胺类化合物及其应用 WO2023274088A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP22831902.6A EP4353730A1 (en) 2021-06-28 2022-06-24 Dimethyl-substituted thiazololactam compound and use thereof
AU2022303895A AU2022303895A1 (en) 2021-06-28 2022-06-24 Dimethyl-substituted thiazololactam compound and use thereof
KR1020247002353A KR20240024961A (ko) 2021-06-28 2022-06-24 디메틸-치환된 티아졸로락탐 화합물 및 이의 용도
BR112023027454A BR112023027454A2 (pt) 2021-06-28 2022-06-24 Composto, uso de um composto, medicamento, e, invenção de produto, processo, sistema, kit ou uso
CA3224314A CA3224314A1 (en) 2021-06-28 2022-06-24 Dimethyl-substituted thiazololactam compound and use thereof
IL309706A IL309706A (en) 2021-06-28 2022-06-24 Dimethyl-converted thiazololactam compound and its uses
CN202280043903.0A CN117561265A (zh) 2021-06-28 2022-06-24 二甲基取代的噻唑并内酰胺类化合物及其应用

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202110723288 2021-06-28
CN202110723288.5 2021-06-28
CN202111669920.9 2021-12-31
CN202111669920 2021-12-31
CN202210693547 2022-06-17
CN202210693547.9 2022-06-17

Publications (1)

Publication Number Publication Date
WO2023274088A1 true WO2023274088A1 (zh) 2023-01-05

Family

ID=84690065

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/101283 WO2023274088A1 (zh) 2021-06-28 2022-06-24 二甲基取代的噻唑并内酰胺类化合物及其应用

Country Status (9)

Country Link
EP (1) EP4353730A1 (zh)
KR (1) KR20240024961A (zh)
CN (1) CN117561265A (zh)
AU (1) AU2022303895A1 (zh)
BR (1) BR112023027454A2 (zh)
CA (1) CA3224314A1 (zh)
IL (1) IL309706A (zh)
TW (1) TWI836486B (zh)
WO (1) WO2023274088A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020228817A1 (zh) * 2019-05-15 2020-11-19 南京明德新药研发有限公司 Erk抑制剂及其应用
CN112457326A (zh) * 2019-09-06 2021-03-09 上海凌达生物医药有限公司 一类芳香杂环并内酰胺类化合物、制备方法和用途
CN112638917A (zh) * 2018-05-22 2021-04-09 捷思英达控股有限公司 作为激酶抑制剂的杂环化合物、包括该杂环化合物的组合物、及其使用方法
WO2021110169A1 (zh) * 2019-12-06 2021-06-10 南京明德新药研发有限公司 作为erk抑制剂的噻唑并内酰胺类化合物及其应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112638917A (zh) * 2018-05-22 2021-04-09 捷思英达控股有限公司 作为激酶抑制剂的杂环化合物、包括该杂环化合物的组合物、及其使用方法
WO2020228817A1 (zh) * 2019-05-15 2020-11-19 南京明德新药研发有限公司 Erk抑制剂及其应用
CN112457326A (zh) * 2019-09-06 2021-03-09 上海凌达生物医药有限公司 一类芳香杂环并内酰胺类化合物、制备方法和用途
WO2021110169A1 (zh) * 2019-12-06 2021-06-10 南京明德新药研发有限公司 作为erk抑制剂的噻唑并内酰胺类化合物及其应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CANCER DISCOV., vol. 9, 2019, pages 329 - 341

Also Published As

Publication number Publication date
BR112023027454A2 (pt) 2024-03-12
TW202309054A (zh) 2023-03-01
CA3224314A1 (en) 2023-01-05
KR20240024961A (ko) 2024-02-26
CN117561265A (zh) 2024-02-13
TWI836486B (zh) 2024-03-21
IL309706A (en) 2024-02-01
EP4353730A1 (en) 2024-04-17
AU2022303895A1 (en) 2024-02-15

Similar Documents

Publication Publication Date Title
CN109563047A (zh) 含有硅原子的依伐卡托类似物
WO2020228817A1 (zh) Erk抑制剂及其应用
TWI758999B (zh) 作為erk抑制劑的噻唑并內醯胺類化合物及其應用
CN114761411B (zh) 作为erk抑制剂的螺环类化合物及其应用
TWI789886B (zh) 一種作為可透腦的btk或her2抑制劑的化合物及其製備方法與用途
CN102307868A (zh) 作为抗癌剂的吲哚衍生物
WO2023274088A1 (zh) 二甲基取代的噻唑并内酰胺类化合物及其应用
KR20220024199A (ko) Ccr2/ccr5 길항제로서의 헤테로시클로알킬류 화합물
US20240217973A1 (en) Imidazopyridine compounds and use thereof
WO2023274256A1 (zh) 噻唑并内酰胺并螺杂环类化合物及其应用
JP2024526243A (ja) ジメチル置換チアゾロラクタム化合物及びその使用
US20220033376A1 (en) Estrogen receptor antagonist
RU2805569C1 (ru) Тиазололактамные соединения в качестве ингибиторов ERK и их применение
EP4183788A1 (en) Crystal of pyrimidine compound
CN116120315B (zh) 一种kras g12c抑制剂及其应用
WO2022143773A1 (zh) 苯并咪唑类化合物及其应用
JP2024526242A (ja) チアゾール-ラクタム-スピロ複素環化合物及びその適用
US20230203057A1 (en) Five-membered heteroaromatic imidazole compound and use thereof
CN115340527A (zh) 一种bcl-xl抑制剂及其制备方法和用途

Legal Events

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

Ref document number: 22831902

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 18571087

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 202280043903.0

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 309706

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2023580483

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 202393513

Country of ref document: EA

Ref document number: 3224314

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023027454

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: MX/A/2024/000084

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2022831902

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20247002353

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020247002353

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: AU2022303895

Country of ref document: AU

Ref document number: 807719

Country of ref document: NZ

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022831902

Country of ref document: EP

Effective date: 20240108

ENP Entry into the national phase

Ref document number: 2022303895

Country of ref document: AU

Date of ref document: 20220624

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112023027454

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20231226