CN116693518A - Synthesis and application of EGFR inhibitor - Google Patents

Synthesis and application of EGFR inhibitor Download PDF

Info

Publication number
CN116693518A
CN116693518A CN202310398689.7A CN202310398689A CN116693518A CN 116693518 A CN116693518 A CN 116693518A CN 202310398689 A CN202310398689 A CN 202310398689A CN 116693518 A CN116693518 A CN 116693518A
Authority
CN
China
Prior art keywords
pharmaceutically acceptable
cancer
compound
acceptable salt
ring
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.)
Pending
Application number
CN202310398689.7A
Other languages
Chinese (zh)
Inventor
唐春雷
袁昕
范为正
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.)
Jiangnan University
Original Assignee
Jiangnan University
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 Jiangnan University filed Critical Jiangnan University
Priority to CN202310398689.7A priority Critical patent/CN116693518A/en
Publication of CN116693518A publication Critical patent/CN116693518A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

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

Abstract

The invention discloses synthesis and application of an EGFR inhibitor, and belongs to the technical field of medicines. The invention relates to synthesis and application of oxo-isoindole compounds and pharmaceutically acceptable salts thereof, which can selectively inhibit EGFR (epidermal growth factor receptor), so that the oxo-isoindole compounds and the pharmaceutically acceptable salts thereof can be used for treating or improving abnormal cell proliferation diseases.

Description

Synthesis and application of EGFR inhibitor
Technical Field
The invention belongs to the field of medicines, and particularly relates to synthesis and application of an EGFR inhibitor.
Background
Epidermal Growth Factor Receptor (EGFR) is one of the transmembrane protein tyrosine kinases of the ErbB receptor family, which when bound to a growth factor ligand (e.g., epidermal Growth Factor (EGF)), can homodimerize with additional EGFR molecules or heterodimerize with another family member (e.g., erbB2 (HER 2), erbB3 (HER 3), or ErbB4 (HER 4)), homodimerization and/or heterodimerization of ErbB receptors resulting in phosphorylation of intracellular critical tyrosine residues and stimulation of many intracellular signaling pathways involved in cell proliferation and survival. Deregulation of ErbB family signaling promotes proliferation, invasion, metastasis, angiogenesis and survival of tumor cells, and is closely related to human cancers such as lung cancer, head and neck cancer, colon cancer, breast cancer, and the like.
The small molecule kinase inhibitor comprises first generation EGFR-TKI targeting drugs erlotinib, gefitinib and the like, and has good treatment effect on gold mutation of EGFR, namely 19 exon deletion mutation (delE 746-A750) and 21 exon point mutation (L858R) of EGFR genes. The second-generation EGFR-TKI targeting drugs such as dacatinib and afatinib have the advantages that the curative effect is not obviously improved compared with the first-generation drugs, and the side effect is larger, so that the clinical application is not wide. The third generation EGFR-TKI targeting drugs of Ornitinib, eveltinib and the like have been used as the first choice for the treatment of EGFR sensitive mutation and T790M resistant mutation NSCLC. They are Adenosine Triphosphate (ATP) analogues that block activation of signal pathways by blocking phosphorylation of intracellular domains.
In recent decades, molecular targeted therapies represented by epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKI) have brought tremendous revolution to cancer treatment, but EGFR-TKI resistance has always been a pending problem. For example, EGFR-TKIs were initially effective in non-small cell lung cancer (NSCLC) patients carrying EGFR mutations, but often evolved to acquired resistance and lost therapeutic effects, such as T790M mutation resulting in first/second generation EGFR-TKI resistance and C797S mutation resulting in third generation EGFR-TKI resistance, and the like, especially tumors containing both L858R/T790M/C797S mutation, for which currently existing EGFR-TKIs are not of interest. However, all current EGFR TKIs target the ATP site, although third generation irreversible inhibitors (e.g., octenib) are able to overcome T790M, drug resistance occurs due to the C797S mutation that has occurred in the treated patient. Cetuximab, an anti-EGFR antibody, which blocks receptor dimerization, is ineffective in EGFR-mutant NSCLC because mutant activation of the kinase is effectively "downstream" of receptor dimerization. Thus, an alternative strategy is to inhibit EGFR. Currently, suitable compounds with alternative mechanisms of action that target mutant EGFR are not available.
Recent studies have shown that targeted targeting of allosteric sites can lead to the fact that mutant selective inhibitors do have a need to generate selective molecules that specifically inhibit EGFR mutants containing T790M/L858R, T M/L858R/C797S, L858R, L858R/C797S, in particular EGFR mutants containing T790M and C797S, which can be used for the therapeutic and/or prophylactic treatment of cancer. Thus, there is an urgent need for new and effective small molecule EGFR inhibitors with alternative mechanisms of action that target mutant EGFR. EGFR allosteric inhibitors have been found to be able to bind to different EGFR sites with existing ATP-competitive EGFR TKIs, and are considered potential therapeutic strategies to overcome the therapeutic resistance of EGFR mutant patients.
In 2019, michael J.Eck, nathanael S.Gray, pasi A, by the Husky medical institute Dana-Faber cancer research center.A mutant selective EGFR allosteric inhibitor, JBJ-04-125-02, was reported by the Time team. Studies have shown that it is effective against EGFR L858R/T790M/C797S mutation both in vitro and in vivo, but unfortunately, the single treatment of JBJ-04-125-02 was ineffective in patient-derived cell lines or xenograft models, and then the team improved on JBJ-04-125-02, and found a new and more potent EGFR allosteric inhibitor, JBJ-09-063, with better pharmacokinetics and efficacy than JBJ-04-125-02, and good therapeutic efficacy against models that result in EGFR-TKI resistance, such as EGFR T790M mutation and C797S mutation. However, how to further improve the EGFR inhibition effect is still a problem.
Disclosure of Invention
In order to solve the problems, the inventor provides a novel oxo-isoindole compound which has more excellent pharmacokinetic properties and EGFR inhibition effect than JBJ-09-063 and JBJ-04-125-02 and has good development prospect.
The present invention provides a compound of the following general formula (I):
ring A is a substituted or unsubstituted benzene ring, and ring B is optionally self-substituted
Alternatively, ring A is a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted C36 cycloalkyl, and ring B is optionally self-substituted
Wherein the substituents on the benzene ring, heterocyclyl or C36 cycloalkyl are selected from: c1-4 alkyl; n is 1-4;
R 1 、R 2 、R 3 、R 1 ’、R 2 ’、R 3 ’、R 4 ' are each independently selected from H, C1-4 alkyl, deuterated C1-4 alkyl; r is R 4 H, C2-4 alkyl, deuterated C2-4 alkyl; r is R 1 、R 2 、R 3 、R 4 、R 1 ’、R 2 ’、R 3 ’、R 4 ' when substituted, includes mono-, di-, tri-, or tetra-substituted;
in one embodiment of the present invention, the heterocyclic group includes:
in one embodiment of the inventionWherein the C36 cycloalkyl group comprises:
in one embodiment of the present invention, when ring a is a benzene ring, ring B is specifically selected from:
in one embodiment of the present invention, when ring a is a benzene ring, ring B is further preferred:
In one embodiment of the present invention, when ring a is a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted C36 cycloalkyl, ring B is specifically selected from:
in the present invention, further particularly preferred compounds of formula (I) or pharmaceutically acceptable salts thereof are selected from:
the compound shown in the general formula (I) or pharmaceutically acceptable salt thereof is inorganic salt or organic salt, wherein the inorganic salt comprises hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate and acid phosphate; the organic salt is selected from acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate, maleate, lactate, malate, citrate, tartrate, methanesulfonate, sulfonate, benzenesulfonate, salicylate.
It is a further object of the present invention to provide pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as described above in accordance with the present invention, together with a pharmaceutically acceptable carrier, excipient or diluent.
In another aspect, the present invention provides the use of a compound of formula (I) above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in an EGFR inhibitor.
In another aspect, the present invention provides the use of a compound of formula (I) above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
The cancers referred to in the present invention may be selected from: breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, hematological cancer, gastrointestinal cancer (e.g., gastric and colorectal cancer), or lung cancer, and the like.
The cancers referred to in the present invention may be selected from: lung cancer, bone cancer, membranous adenocarcinoma, skin cancer, head or neck cancer, skin melanin cancer or intraocular melanin cancer, uterine cancer, ovarian cancer, rectal cancer, gall bladder cancer, stomach cancer, colon cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, hodgkin's disease, esophageal cancer, small intestine cancer, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal gland cancer, soft tissue meat cancer, urinary tract cancer, penile cancer, prostate cancer, chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, renal cancer or ureter cancer, renal cell carcinoma, renal Monascus cancer, central Nervous System (CNS) cancer, primary central nervous system lymphoma, spinal cord axis cancer, brain stem glioma, brain vertical adenoma, and the like.
The invention has the beneficial effects of designing a compound with biological functions of inhibiting EGFR, thereby providing a new means for searching new treatment methods of cancers, metabolic and immune diseases, cardiovascular diseases, neurological diseases and the like.
The beneficial effects are that:
the invention provides a class of compounds having a structure according to formula (I) useful in the therapeutic and/or prophylactic treatment of patients suffering from cancer, in particular non-small cell lung cancer, having EGFR mutations T790M/L858RT790M/L858R/C797S, L858R and/or L858R/C797S, said treatment comprising determining the EGFR activating mutation status of said patient, and then administering to said patient a compound of formula I as described herein or a pharmaceutically acceptable salt thereof.
Detailed Description
The technical scheme of the present invention will be described in detail with reference to examples.
In the present invention, "C 1 -C 6 Alkyl "refers to a saturated, straight or branched, monovalent hydrocarbon radical of 1 to 6 carbon atoms, respectively. Examples include, but are not limited to, methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2-methyl-1-propyl, 2-butyl, and 2-methyl-2-propyl.
In the present invention, "C 3 -C 6 Cycloalkyl "refers to cycloalkyl groups of 3 to 6 carbon atoms, respectively.
"heteroaryl" in the context of the present invention, unless otherwise indicated, refers to an unsubstituted or substituted stable 5-or 6-membered monocyclic aromatic ring system or an unsubstituted or substituted 9-or 10-membered benzofused heteroaromatic ring system or bicyclic heteroaromatic ring system consisting of carbon atoms and 1 to 4 heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may be optionally oxidized and the nitrogen heteroatoms may be optionally quaternized.
"substituted" in the context of the present invention means that one or more hydrogen atoms in the group are each replaced by the same or different substituents.
In the present invention, "administering" or "administering" a compound to an individual refers to providing a compound of the present invention to an individual in need of treatment.
< Compound or pharmaceutically acceptable salt thereof >
The invention provides a novel oxo-isoindole compound or pharmaceutically acceptable salt thereof, which can be used as an inhibitor for inhibiting Epidermal Growth Factor Receptor (EGFR), and has a structural formula shown in a general formula (I):
< pharmaceutical composition >
The invention also provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof according to the invention as described above, and a pharmaceutically acceptable carrier, excipient or diluent.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated as solid formulations for oral administration, including, but not limited to, capsules, tablets, pills, powders, granules, and the like. In these solid dosage forms, the compounds of formula (I) according to the invention are mixed as active ingredient with at least one conventional inert excipient (or carrier), for example with sodium citrate or dicalcium phosphate. Or with the following components: (1) Fillers or solubilisers, for example starch, lactose, sucrose, glucose, mannitol, silicic acid and the like; (2) Binders, for example, hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose, acacia, and the like; (3) humectants, for example, glycerin, etc.; (4) Disintegrants, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium carbonate, and the like; (5) a slow solvent such as paraffin wax or the like; (6) absorption accelerators such as quaternary ammonium compounds and the like; (7) Wetting agents such as cetyl alcohol and glycerol monostearate, and the like; (8) adsorbents such as kaolin and the like; (9) Lubricants, for example, talc, calcium stearate, solid polyethylene glycol, sodium lauryl sulfate, and the like, or mixtures thereof. Buffers may also be included in capsules, tablets, pills.
The solid dosage forms, such as tablets, dragees, capsules, pills and granules, may be provided with coatings and shell materials such as enteric coatings and other materials known in the art in the form of crystalline coatings or microencapsulations. They may contain opacifying agents and the release of the active ingredient in such a composition may be released in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxes. The active ingredient may also be in the form of microcapsules with one or more of the above excipients, if desired.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated into liquid dosage forms for oral administration, including, but not limited to, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, tinctures, and the like. In addition to the compounds of formula (I) or pharmaceutically acceptable salts thereof as active ingredients, liquid dosage forms may contain inert diluents commonly used in the art such as water and other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular cottonseed, groundnut, corn, olive, castor, sesame oils and the like or mixtures of these substances and the like. In addition to these inert diluents, the liquid dosage forms of the present invention can also include conventional adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents and the like.
Such suspending agents include, for example, ethoxylated stearyl alcohol, polyoxyethylene sorbitol, and sorbitan, microcrystalline cellulose, agar-agar, and the like, or mixtures of these.
The compounds of the present invention and pharmaceutically acceptable salts thereof may be formulated in dosage forms for parenteral injection, including, but not limited to, physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions and dispersions. Suitable carriers, diluents, solvents, excipients include water, ethanol, polyols and suitable mixtures thereof.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated into dosage forms for topical administration, including, for example, ointments, powders, suppositories, drops, sprays, inhalants and the like. The compounds of the general formula (I) according to the invention or their pharmaceutically acceptable salts as active ingredients are mixed under sterile conditions with physiologically acceptable carriers and optionally with preservatives, buffers and, if appropriate, propellants.
The pharmaceutical composition of the invention comprises a compound of a general formula (I) or pharmaceutically acceptable salt thereof as an active ingredient, and pharmaceutically acceptable carriers, excipients and diluents. In preparing pharmaceutical compositions, a compound of formula (I) or a pharmaceutically acceptable salt thereof of the present invention is typically admixed with a pharmaceutically acceptable carrier, excipient or diluent. Wherein the content of the compound of the general formula (I) or a pharmaceutically acceptable salt thereof may be 0.01 to 1000mg, for example, 0.05 to 800mg, 0.1 to 500mg, 0.01 to 300mg, 0.01 to 200mg, 0.05 to 150mg, 0.05 to 50mg, etc.
< use >
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 a mammal.
The compounds of formula (I) or pharmaceutically acceptable salts thereof, which inhibit the activity of Epidermal Growth Factor Receptor (EGFR)), are useful in the treatment of mammals, including humans, for the therapeutic and/or prophylactic treatment of patients having EGFR activating mutations, particularly non-small cell lung cancer, which treatment comprises determining the status of EGFR activating mutations in said patients, and thereafter administering to said patients a compound of formula I as described herein, or a pharmaceutically acceptable salt thereof.
A "therapeutically effective amount" is an amount of a compound of the invention effective to produce a biological or medical response (e.g., reduce or inhibit the activity of an enzymatically active protein, or ameliorate symptoms, alleviate a condition, slow or delay the progression of a disease, or prevent a disease) in an individual.
Cancers as referred to herein include lung cancer, bone cancer, membranous adenocarcinoma, skin cancer, head or neck cancer, skin melanin cancer or intraocular melanin cancer, uterine cancer, ovarian cancer, rectal cancer, gall bladder cancer, stomach cancer, colon cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulval cancer, hodgkin's disease, esophageal cancer, small intestine cancer, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue meat cancer, urinary tract cancer, penile cancer, prostate cancer, chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, renal cancer or ureter cancer, renal cell carcinoma, renal Monday cancer, central Nervous System (CNS) cancer, primary central nervous system lymphoma, spinal cord shaft cancer, brain stem glioma, brain vertical adenoma, and the like.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be administered to mammals, including humans, orally, rectally, parenterally (intravenous, intramuscular or subcutaneous), topically (powders, ointments, drops) or intratumorally.
The compounds of the present invention, or pharmaceutically acceptable salts thereof, may be administered alone or in combination with other pharmaceutically acceptable therapeutic agents, in combination with other antitumor agents. The combination therapy may be achieved by the simultaneous, sequential or separate use of the individual components of the therapy. Such therapeutic agents include, but are not limited to: antitumor drugs acting on DNA chemical structures such as cisplatin, antitumor drugs affecting nucleotide synthesis such as methotrexate, 5-fluorouracil and the like, antitumor drugs affecting nucleic acid transcription such as doxorubicin, epirubicin, aclacinomycin and the like, antitumor drugs acting on tubulin synthesis such as paclitaxel, vinorelbine and the like, aromatase inhibitors such as aminoglutethimide, letrozole, ryanodine and the like, cell signaling pathway inhibitors such as ALK inhibitors such as crizotinib, ceritinib, ai Leti ni, lazotinib and the like. Anti-tumor monoclonal antibodies, immunosuppressants PD-1, PD-L1, etc., and the individual components to be combined may be administered simultaneously or sequentially, in a single formulation or in different formulations. The combinations include not only combinations of one or more other active agents of the compounds of the present invention, but also combinations of two or more other active agents of the compounds of the present invention.
The invention will be further illustrated with reference to specific examples. It should be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental procedure, in which specific conditions are not noted in the examples below, is generally followed by conventional conditions.
Example 1:
2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-hydroxyphenyl) -N- (thiazol-2-yl) acetamide
Preparation of 2-amino-2- (5-fluoro-2-methoxyphenyl) acetonitrile (intermediate 1):
in a 100mL thick-wall pressure-resistant bottle, 5-fluoro-2-methylDissolving oxybenzaldehyde (10 g,65 mmol), anhydrous zinc iodide (0.5 g) and 4A molecular sieve (1 g) in 100mL anhydrous acetonitrile, cooling to below-15deg.C, and introducing NH 3 To saturation TMSCN (6.4 g,65 mmol) was added and the mixture was sealed and reacted overnight at 65 ℃. After the completion of the reaction, the reaction mixture was filtered, concentrated and recrystallized from diethyl ether-petroleum ether to give 2-amino-2- (5-fluoro-2-methoxyphenyl) acetonitrile (intermediate 1) (8 g, 68%) as a brown oil. ESI-MS m/z 181.2[ M+H ]] +
Preparation of 2-amino-2- (5-fluoro-2-methoxyphenyl) acetic acid hydrochloride (intermediate 2):
2-amino-2- (5-fluoro-2-methoxyphenyl) acetonitrile (intermediate 1) (8 g,44.4 mmol) was dissolved in concentrated hydrochloric acid (100 mL), heated to 80℃and reacted overnight, after the completion of the reaction, the impurities were removed by extraction with ethyl acetate (50 mL). The aqueous phase was concentrated under reduced pressure to give 2-amino-2- (5-fluoro-2-methoxyphenyl) acetate (intermediate 2) (6.2 g, 60%) as a white solid. ESI-MS m/z 200.1[ M+H ] ] +
Preparation of 2- ((tert-butoxycarbonyl) amino) -2- (5-fluoro-2-methoxyphenyl) acetic acid (intermediate 3):
2-amino-2- (5-fluoro-2-methoxyphenyl) acetate (intermediate 2) (6 g,25.5 mmol) was dissolved in THF (30 mL) and 1N NaOH (30 mL) and Boc was added thereto 2 O (6.67 g,30.6 mmol), stirring at room temperature for 18 h, after completion of the reaction, THF was removed by concentration under reduced pressure, the aqueous phase was washed once with DCM (50 mL), then adjusted to pH 2 with 1N HCl, DCM (100 mL. Times.3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 2- ((tert-butoxycarbonyl) amino) -2- (5-fluoro-2-methoxyphenyl) acetic acid (intermediate 3) (5.8 g, 76%) as a colorless oil. ESI-MS m/z 300.3[ M+H ]] +
Preparation of tert-butyl (1- (5-fluoro-2-methoxyphenyl) -2-oxo-2- (thiazol-2-ylamino) ethyl) carbamate (intermediate 4):
2- ((tert-Butoxycarbonyl) amino) -2- (5-fluoro-2-methoxyphenyl) acetic acid (intermediate 3) (5 g,16.7 mmol) and thiazol-2-amine hydrochloride (2.7 g,20.0 mmol) were dissolved in DMF (50 mL), DIPEA (7 mL,41.7 mmol) and HATU (9.5 g,25.0 mmol) were added and the reaction mixture was stirred at room temperature for 18 hours after the end of the reaction. The reaction was slowly poured into water, extracted with ethyl acetate (100 ml x 3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give tert-butyl (1- (5-fluoro-2-methoxyphenyl) -2-oxo-2- (thiazol-2-ylamino) ethyl) carbamate (intermediate 4) (4.5 g, 70%) as a white solid. ESI-MS m/z 382.2[ M+H ] ] +
Preparation of 2-amino-2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 5):
tert-butyl (1- (5-fluoro-2-methoxyphenyl) -2-oxo-2- (thiazol-2-ylamino) ethyl) carbamate (intermediate 4) (4 g,10.5 mmol) was dissolved in methanol (30 mL), HCl (4M in dioxane) (20 mL) was added, the reaction mixture was stirred at room temperature for 18 hours, after the completion of the reaction, 2-amino-2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 5) (2.9 g, 98%) was obtained as a white solid by concentration under reduced pressure. ESI-MS m/z 282.1[ M+H ]] +
Preparation of 2- (6-bromo-1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 6):
2-amino-2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 5) (2.5 g,8.9 mmol) and methyl 5-bromo-2- (bromomethyl) benzoate (2.7 g,8.9 mmol) were dissolved in DMF (40 mL), DIPEA (6 mL,35.6 mmol) was added, heated to 70℃for 2 hours, after completion of the reaction, cooled to room temperature, the reaction was slowly poured into water, extracted with ethyl acetate (100 mL 3), the organic phases combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give 2- (6-bromo-1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 6) (3 g, 71%) as a white solid. ESI-MS m/z 476.4[ M+H ] ] +
Preparation of 2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 7):
2- (6-bromo-1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 6) (100 mg,0.21 mmol) and 2- (4-cyclohexylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborane (72 mg,0.25 mmol) were dissolved in THF (2 mL)) In which sodium carbonate (44 mg,0.42 mmol) and PdCl were added sequentially 2 (dppf) (15 mg,0.02 mmol) and water (0.5 mL), and the reaction solution was heated to 70℃under nitrogen atmosphere and stirred for 16 hours. After the reaction was completed, ethyl acetate (15 ml x 3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 7) (60 mg, 64%) as a white solid. ESI-MS m/z 556.3[ M+H ]] +
Preparation of 2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-hydroxyphenyl) -N- (thiazol-2-yl) acetamide (example 1):
2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-methoxyphenyl) -N- (thiazol-2-yl) acetamide (intermediate 7) (60 mg,0.11 mmol) was dissolved in DCM (2 mL), boron tribromide (55 mg,0.22 mmol) was added and stirred at room temperature for 3 hours, after completion of the reaction, water was added to quench, DCM (5 mL x 3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and isolated as plates to give 2- (6- (4-cyclohexylphenyl) -1-oxoisoindol-2-yl) -2- (5-fluoro-2-hydroxyphenyl) -N- (thiazol-2-yl) acetamide (example 1) (30 mg, 51%) as a white solid. ESI-MS m/z 542.1[ M+H ] ] +
1 H NMR(400MHz,DMSO-d 6 )δ7.90(d,J=0.8Hz,1H),7.74-7.68(m,1H),7.62-7.55(m,2H),7.54-7.48(m,2H),7.47-7.40(m,2H),7.12(d,J=6.0Hz,1H),7.01-6.94(m,1H),6.88-6.78(m,2H),5.85(s,1H),5.82(s,1H),5.11(s,1H),4.92(s,1H),2.58(dq,J=15.0,7.5Hz,1H),2.25-2.00(m,2H),1.85-1.66(m,3H),1.60-1.47(m,2H),1.46-1.35(m,3H).
Preparation of 4 '-bromo-4, 4-dimethyl-2, 3,4, 5-tetrahydro-1, 1' -biphenyl (intermediate 8):
p-bromophenyl boric acid (1 g,5 mmol) and 4, 4-dimethylcyclohex-1-en-1-yl triflate (1.9 g,7.5 mmol) were dissolved in THF (10 mL) and water (1 mL), triethylamine (1.4 mL,10 mmol) and tetrakis triphenylphosphine palladium (289 mg,0.25 mmol) were added, and the reaction solution was heated to 70℃under nitrogen atmosphere and stirred for 16 hours. After the reaction was completed, water (50 mL) was added, extraction was performed with ethyl acetate (50 ml×3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 4 '-bromo-4, 4-dimethyl-2, 3,4, 5-tetrahydro-1, 1' -biphenyl (intermediate 8) (800 mg, 61%) as a colorless oil.
Preparation of 1-bromo-4- (4, 4-dimethylcyclohexyl) benzene (intermediate 9):
4 '-bromo-4, 4-dimethyl-2, 3,4, 5-tetrahydro-1, 1' -biphenyl (intermediate 8) (800 mg,3.0 mmol) was dissolved in methanol (5 mL) and 10% Pd-C (100 mg), H was added 2 After the reaction was completed, the mixture was filtered through celite and the filtrate was concentrated under reduced pressure to give 1-bromo-4- (4, 4-dimethylcyclohexyl) benzene (intermediate 9) (0.8 g, 100%) as a colorless oil.
Preparation of 2- (4, 4-dimethylcyclohexyl) phenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborane (intermediate 10):
1-bromo-4- (4, 4-dimethylcyclohexyl) benzene (intermediate 9) (800 mg,3.0 mmol) and pinacol biborate (1100 mg,4.5 mmol) were dissolved in dioxane (10 mL), and potassium acetate (588 mg,6.0 mmol) and PdCl were added 2 (dppf) (22 mg,0.03 mmol) the reaction was heated to 80℃under nitrogen and stirred for 16 hours. After the reaction was completed, water (30 mL) was added, ethyl acetate (50 ml×3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 2- (4, 4-dimethylcyclohexyl) phenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborane (intermediate 10) (400 mg, 42%) as a colorless oil.
Preparation of 1-bromo-4- (4-methylcyclohexyl) benzene (intermediate 11):
methyl triphenylphosphine bromide (5.9 g,16.5 mmol) and potassium tert-butoxide (1.8 g,16.5 mmol) were mixed in diethyl ether (100 mL), stirred at room temperature for 1 hour, then a solution of 4- (4-bromophenyl) cyclohexane-1-one (3 g,11.8 mmol) in diethyl ether (20 mL) was added dropwise, stirred at room temperature overnight, after the reaction was completed, water (50 mL) was added, diethyl ether (50 mL. Times.3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give 1-bromo-4- (4-methylcyclohexyl) benzene (intermediate 11) (700 mg, 23%) as a white solid.
Preparation of 6- (4-bromophenyl) spiro [2.5] octane (intermediate 12):
1-bromo-4- (4-methylcyclohexyl) benzene (intermediate 11) (700 mg,2.8 mmol) and iodomethylboronic acid pinacol ester (1.5 g,5.6 mmol) were dissolved in toluene (20 mL) and BEt was added 3 (5.6 mL,5.6mmol, 1M) at room temperature for 16 hr, adding water (20 mL), extracting with ethyl acetate (30 mL. Times.3), mixing the organic phases, washing with saturated sodium chloride, drying with anhydrous sodium sulfate, concentrating under reduced pressure, and purifying with column to obtain 6- (4-bromophenyl) spiro [2.5]]Octane (intermediate 12) (300 mg, 40%) as a white solid.
Preparation of 4, 5-tetramethyl-2- (4- (spiro [2.5] octane-6-yl) phenyl) -1,3, 2-dioxaborane (intermediate 13):
synthetic method reference is made to the preparation of intermediate 10.
Preparation of 1- (4-bromophenyl) -4, 4-dimethylpiperidine (intermediate 14):
1, 4-Dibromobenzene (1 g,4.2 mmol) and 4, 4-dimethylpiperidine (470 mg,4.2 mmol) were dissolved in toluene (20 mL), and BINAP (522 mg,0.84 mmol), pd, was added sequentially 2 (dba) 3 (384 mg,0.42 mmol), sodium t-butoxide (806 mg,8.4 mmol), under nitrogen atmosphere, is heated to 100deg.C for 6 hours,
after the reaction, water (20 mL) was added, ethyl acetate (20 ml×3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 1- (4-bromophenyl) -4, 4-dimethylpiperidine (intermediate 14) (400 mg, 35%) as a white solid. ESI-MS m/z 268.1[ M+H ] ] +
Preparation of 4, 4-dimethyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) phenyl) piperidine (intermediate 15):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 316[ M+H ]] +
Preparation of synthetic reference intermediate 15 of 6-6- (4-bromophenyl) -6-azaspiro [2.5] octane (intermediate 16) and 6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) phenyl) -6-azaspiro [2.5] octane (intermediate 17), wherein 4, 4-dimethylpiperidine is replaced with 6-azaspiro [2.5] octane.
Preparation of 4- (4-bromophenyl) -1-cyclopropylpiperidine (intermediate 18):
4 (4 bromophenyl) piperidine (2 g,8.3 mmol) was dissolved in THF (20 mL) and MeOH (20 mL), and (1 ethoxy cyclopropyloxy) trimethylsilane (2.9 g,16.6 mmol), sodium cyanoborohydride (782 mg,12.4 mmol) and acetic acid (744 mg,12.4 mmol) were added in this order, and the reaction was heated to 50℃and stirred for 16 hours. After the reaction was completed, water (50 mL), ethyl acetate (50 ml×3) was added to extract, and the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 4- (4-bromophenyl) -1-cyclopropylpiperidine (intermediate 18) (1.2 g, 51%) as a white solid. ESI-MS m/z 280.2[ M+H ]] +
Preparation of 1-cyclopropyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) phenyl) piperidine (intermediate 19):
Synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 328.1[ M+H ]] +
Examples 2-6 (see Table 1) were synthesized in a similar manner to example 1 using the corresponding intermediate boron ester in place of 2- (4-cyclohexylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborane in example 1 to give the desired product.
TABLE 1
Example 7:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
Preparation of 4- (4-bromophenyl) -1-methylpiperidin-4-ol (intermediate 20):
1, 4-dibromobenzene (5 g,21.2 mmol) was dissolved in THF (100 mL) at-78deg.C, n-BuLi (28.5 mL,1.2mmol, 2.5M) was slowly added dropwise, after completion of the dropwise addition, 1-methylpiperidin-4-one (2.4 g,21.2 mmol) was then added, warmed to room temperature, stirred for 1 hour, quenched with water after completion of the reaction, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give 4- (4-bromophenyl) -1-methylpiperidin-4-ol (intermediate 20) (3.2 g, 56%) as a colorless oil. ESI-MS m/z 270.2[ M+H ]] +
Preparation of 4- (4-bromophenyl) -1-methyl-1, 2,3, 6-tetrahydropyridine (intermediate 21):
4- (4-bromophenyl) -1-methylpiperidin-4-ol (intermediate 20) (3 g,11.1 mmol) was added to hydrochloric acid (50 mL,6N, heated to 100℃for 16 hours, and after completion of the reaction, na was added 2 CO 3 Neutralization, extraction with ethyl acetate (100 ml x 3), combined organic phases, washing with saturated sodium chloride, drying over anhydrous sodium sulfate, concentration under reduced pressure and column purification gave 4- (4-bromophenyl) -1-methyl-1, 2,3, 6-tetrahydropyridine (intermediate 21) (1.6 g, 57%),. White solid. ESI-MS m/z 252.1[ M+H ]] +
Preparation of 1-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) phenyl) -1,2,3, 6-tetrahydropyridine (intermediate 22):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 300.1[ M+H ]] +
Preparation of 2- (5-fluoro-2-methoxyphenyl) -2- (6- (4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (intermediate 23):
synthetic method reference is made to the preparation of intermediate 7. ESI-MS m/z 569.3[ M+H ]] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 7):
synthetic method reference is made to the preparation of example 1. White solid. ESI-MS m/z 555.2[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ7.93(d,J=1.2Hz,1H),7.83-7.75(m,1H),7.62-7.56(m,1H),7.55-7.45(m,5H),7.05(d,J=5Hz,1H),7.03-6.97(m,1H),6.84(s,1H),6.77-6.68(m,1H),6.31(q,J=6.1Hz,1H),5.89-5.73(m,2H),4.82-4.62(m,1H),4.34-4.11(m,1H),3.65-3.44(m,2H),2.74(q,J=5.8Hz,1H),2.56-2.51(m,1H),2.46(q,J=5.5Hz,2H),2.36(s,3H).
Example 8:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1- (methyl-d 3) piperidin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
Preparation of 4- (4-bromophenyl) -1- (methyl-d 3) piperidine (intermediate 24):
4- (4-bromophenyl) piperidine (1 g,4.2 mmol) was dissolved in DCM (20 mL), DIPEA (1.1 mL,6.3 mmol) and deuterated iodomethane (0.26 mL,4.2 mmol) were added under ice, after the addition was complete, reacted under ice for 1 hour, water (20 mL) was added after the reaction was complete, DCM (20 mL) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give 4- (4-bromophenyl) -1- (methyl-d 3) piperidine (intermediate 24) (500 mg, 46%) as a white solid. ESI-MS m/z 257.1[ M+H ]] +
Preparation of 1- (methyl-d 3) -4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) piperidine (intermediate 25):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 305[ M+H ]] +
Preparation of 2- (5-fluoro-2-methoxyphenyl) -2- (6- (4- (1- (methyl-d 3) piperidin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (intermediate 26):
synthetic method reference is made to the preparation of intermediate 7. ESI-MS m/z 574.3[ M+H ]] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1- (methyl-d 3) piperidin-4-yl) phenyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 8):
Synthetic method reference is made to the preparation of example 1. White solid. ESI-MS m/z 560.2[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ7.93(d,J=1.2Hz,1H),7.80-7.75(m,1H),7.63-7.55(m,3H),7.53-7.48(m,1H),7.47-7.44(m,2H),7.20(d,J=4Hz,1H),7.03(s,1H),6.95-6.90(m,1H),6.90–6.84(m,1H),5.82(s,1H),5.78(s,1H),4.93(s,1H),4.57(s,1H),2.73(ddd,J=11.5,11.0,5.5Hz,2H),2.70-2.60(m,1H),2.41(ddd,J=12.0,11.0,5.5Hz,2H),2.23-2.14(m,2H),2.01-1.87(m,2H).
Example 9:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohex-1-en-1-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
Preparation of 4- (1, 4-dioxopyridin [4.5] dec-7-en-8-yl) pyridine (intermediate 27):
4, 5-tetramethyl-2- (1, 4-dioxapyrimidine [4.5]]Dec-7-en-8-yl) -1,3, 2-dioxaborane (5 g,18.8 mmol) and 4-bromopyridine (3 g,18.8 mmol) were dissolved in 1, 4-dioxane (50 mL) and water (5 mL) and Na was added sequentially 2 CO 3 (4 g,37.6 mmol) and Pd (PPh) 3 ) 4 (1.1 g,0.94 mmol) and the reaction was heated to 100deg.C under nitrogen and stirred for 16 hours. After the reaction is finished, the mixture is cooled to room temperature,water (100 mL) was added, ethyl acetate (100 mL. Times.3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 4- (1, 4-dioxopyridine [ 4.5)]Dec-7-en-8-yl) pyridine (intermediate 27) (2.3 g, 57%) as a white solid. ESI-MS m/z 218.1[ M+H ]] +
Preparation of 4- (1, 4-Dioxyacetylpyridinium [4.5] dec-8-yl) piperidine (intermediate 28):
4- (1, 4-Dioxypyridine [4.5]]Dec-7-en-8-yl) pyridine (intermediate 27) (2.2 g,10.1 mmol) was dissolved in methanol (30 mL) and acetic acid (15 mL), platinum dioxide (300 mg) was added, the mixture was stirred at room temperature for 48 hours under a hydrogen atmosphere (6 MPa), after the reaction was completed, the mixture was filtered, the filtrate was neutralized with saturated sodium bicarbonate, and the filtrate was extracted with ethyl acetate (100 mL. Times.3), and the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 4- (1, 4-dioxoacetylpyridine [ 4.5% ]Dec-8-yl) piperidine (intermediate 28) (1.5 g, 65%) as a colourless oil. ESI-MS m/z 226.2[ M+H ]] +
Preparation of tert-butyl 4- (4-oxocyclohexyl) piperidine-1-carboxylate (intermediate 29):
4- (1, 4-Dioxyacetylpyridine [4.5 ]]Dec-8-yl) piperidine (intermediate 28) (1.5 g,6.6 mmol) was dissolved in THF (30 mL) and NaHCO was added sequentially 3 (8 mL, 1M) and Boc 2 O (1.7 g,7.9 mmol), stirred at room temperature for 6 hours, after completion of the reaction, water (50 mL), ethyl acetate (50 mL. Times.3) were added, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give tert-butyl 4- (4-oxocyclohexyl) piperidine-1-carboxylate (intermediate 29) (1.5 g, 80%) as a colorless oil. ESI-MS m/z 282.2[ M+H ]] +
Preparation of tert-butyl 4- (4- (((trifluoromethyl) sulfonyl) oxy) cyclohex-3-en-1-yl) piperidine-1-carboxylate (intermediate 30):
tert-butyl 4- (4-oxocyclohexyl) piperidine-1-carboxylate (intermediate 29) (1.5 g,5.3 mmol) was dissolved in THF (15 mL), -dropwise added to a solution of NaHMDS (5.3 mL,5.3mmol, 1M) in THF (15 mL) at 78deg.C, stirred for 30 min with heat preservation, then a solution of N-phenylbis (trifluoromethanesulfonyl) imide (1.9 g,5.3 mmol) in THF (5 mL) was slowly added dropwise, and after completion of the dropwise addition, the reaction was continued at-78deg.C for 1.5 h. Naturally raise the temperature to 0 ℃, After the reaction was completed, saturated ammonium chloride was added to quench (50 mL), extracted with ethyl acetate (50 mL x 3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give tert-butyl 4- (4- (((trifluoromethyl) sulfonyl) oxy) cyclohex-3-en-1-yl) piperidine-1-carboxylate (intermediate 30) (1.7 g, 77%) as a colorless oil. ESI-MS m/z 414.2[ M+H ]] +
Preparation of tert-butyl 4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) cyclohex-3-en-1-yl) piperidine-1-carboxylate (intermediate 31):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 392.2[ M+H ]] +
Synthesis of tert-butyl 4- (4- (2- (1- (5-fluoro-2-methoxyphenyl) -2-oxo-2- (thiazol-2-ylamino) ethyl) -3-oxoisoindol-5-yl) cyclohex-3-en-1-yl) piperidine-1-carboxylate (intermediate 32):
synthetic method reference is made to the preparation of intermediate 7. ESI-MS m/z 661.1[ M+H ]] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (1-oxo-6- (4- (piperidin-4-yl) cyclohex-1-en-1-yl) isoindol-2-yl) -N- (thiazol-2-yl) acetamide (intermediate 33):
synthetic method reference is made to the preparation of example 1. ESI-MS m/z 547.1[ M+H ]] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohex-1-en-1-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 9):
4- (4- (2- (1- (5-fluoro-2-methoxyphenyl) -2-oxo-2- (thiazol-2-ylamino) ethyl) -3-oxoisoindol-5-yl) cyclohex-3-en-1-yl) piperidine-1-carboxylic acid tert-butyl ester (intermediate 33) (200 mg,0.36 mmol) was dissolved in methanol (5 mL), paraformaldehyde (49 mg,0.54 mmol) and acetic acid (22 mg,0.36 mmol) were sequentially added, the reaction mixture was stirred at room temperature for 2 hours, then sodium cyanoborohydride (23 mg,0.36 mmol) was added and stirred at room temperature for 16 hours. After the reaction was completed, the mixture was concentrated under reduced pressure, and 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohex-1-en-1-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 9) (30 mg, 15%) was isolated as a white solid. ESI-MS m/z 561.2[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ7.65(s,1H),7.30(d,J=4.0Hz,1H),7.27-7.24(m,1H),7.20-7.16(m,1H),7.11(d,J=4.0Hz,1H),7.02-6.97(m,1H),6.82(s,1H),6.80-6.77(m,1H),6.15(q,J=6.2Hz,1H),5.83-5.80(m,1H),5.78-5.71(m,1H),4.63-4.56(m,1H),3.96-3.87(m,1H),2.77-2.68(m,2H),2.56-2.36(m,5H),2.25(s,3H),2.18 -2.09(m,1H),2.02-1.85(m,3H),1.50-1.41(m,1H),1.39-1.26(m,3H),1.26-1.17(m,1H).
Example 10:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohexyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohex-1-en-1-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 9) (100 mg,0.18 mmol) was dissolved in methanol (5 mL), pd-C (50 mg) was added and stirred at room temperature under hydrogen atmosphere for 16 hours, after completion of the reaction, the filtrate was filtered through celite and concentrated under pressure to give 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (4- (1-methylpiperidin-4-yl) cyclohexyl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 10)) (55 mg, 55%) as a white solid by plate separation. ESI-MS m/z 563.2[ M+H ] ] +
1 H NMR(400MHz,DMSO-d 6 )δ7.86(s,1H),7.59(d,J=4.0Hz,1H),7.40-7.34(m,1H),7.34(s,1H),7.12(d,J=4.0Hz,1H),7.01-6.92(m,1H),6.89-6.76(m,2H),5.85(s,2H),5.08(s,1H),4.80-4.45(m,1H),2.92-2.58(m,3H),2.45-2.33(m,2H),2.25(s,3H),2.16 -2.08(m,2H),2.00-1.93(m,2H),1.87-1.76(m,2H),1.66-1.55(m,2H),1.39-1.20(m,4H),1.10-0.96(m,2H).
Preparation of 4- (4-bromo-2-oxopyridin-1 (2H) -yl) piperidine-1-carboxylic acid tert-butyl ester (intermediate 34):
4- ((methylsulfonyl) oxy) piperidine-1-carboxylic acid tert-butyl ester (2 g,7.2 mmol) was dissolved in DMF (20 mL), 4-bromopyridine-2 (1.2 g,7.2 mmol) and cesium carbonate (3.5 g,10.8 mmol) were added sequentially, then heated to 100deg.C for 3 hours, cooled to room temperature after reaction, water (100 mL) was added, ethyl acetate (100 mL. Times.3) was used to extract, the organic phases combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified via column to give 4- (4-bromo-2-oxopyridin-1 (2H) -yl) piperidine-1-carboxylic acid tert-butyl ester (intermediate 34) (1.3 g, 52%) as a white solid. ESI-MS m/z 357.1[ M+H ]] +
Preparation of tert-butyl 4- (2-oxo-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) pyridin-1 (2H) -yl) piperidine-1-carboxylate (intermediate 35):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 405.2[ M+H ]] +
Example 11:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (1- (1-methylpiperidin-4-yl) -2-oxo-1, 2-dihydropyridin-4-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
/>
Example 11 was synthesized following the procedure for the preparation of example 9, wherein intermediate 35 was substituted for intermediate 31 as a white solid. ESI-MS m/z 574.2[ M+H ] ] +
1 H NMR(400MHz,DMSO-d 6 )δ7.81(s,1H),7.68(d,J=4.0Hz,1H),7.65-7.61(m,1H),7.60-7.58(m,1H),7.57(s,1H),7.15-7.10(m,1H),7.00-6.94(m,1H),6.89-6.82(m,2H),6.01-5.85(m,2H),5.82(s,2H),5.02-4.90(m,2H),3.87(tt,J=14.5,5.7Hz,1H),2.90-2.68(m,4H),2.42-2.33(m,2H),2.31(s,3H),1.68-1.20(m,2H).
Example 12:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (1- (1-methylpiperidin-4-yl) -1,2,3, 6-tetrahydropyridin-4-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
Preparation of 8- (1-methylpiperidin-4-yl) -1, 4-dioxa-8-azaspiro [4.5] decane (intermediate 36):
1-methylpiperidin-4-one (2 g,17.7 mmol) and 1, 4-dioxa-8-azaspiro [4.5]]Decane (2.5 g,17.7 mmol) was dissolved in 1, 2-dichloroethane (50 mL), acetic acid (1.1 g,17.7 mmol) and sodium triacetoxyborohydride (5.6 g,26.5 mmol) were added. The reaction solution is stirred for 16 hours at room temperature, after the reaction is finished, the solution is directly concentrated under reduced pressure and purified by a column to obtain 8- (1-methylpiperidin-4-yl) -1, 4-dioxa-8-azaspiro [4.5]]Decane (intermediate 36) (2.5 g, 59%) was a pale yellow liquid. ESI-MS m/z 241.1[ M+H ]] +
Preparation of 1 '-methyl- [1,4' -piperidin-4-one (intermediate 37):
8- (1-methylpiperidin-4-yl) -1, 4-dioxa-8-azaspiro [4.5]Decane (intermediate 36) (2.5 g,10.4 mmol) was added to concentrated hydrochloric acid (30 mL), stirred at room temperature for 2 hours, after completion of the reaction, ph=14 was adjusted with sodium hydroxide, ethyl acetate (100 ml×3) was extracted, the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column to give 1 '-methyl- [1,4' -piperidin-4-one (intermediate 37) (1.4 g, 70%) as a colorless liquid. ESI-MS m/z 197.2[ M+H ] ] +
Synthesis of 1- (1-methylpiperidin-4-yl) -1,2,3, 6-tetrahydropyridin-4-yl triflate (intermediate 38):
synthetic method reference is made to the preparation of intermediate 30. 329.3[ M+H ] ESI-MS m/z] +
Preparation of 1- (1-methylpiperidin-4-yl) -4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -1,2,3, 6-tetrahydropyridine (intermediate 39):
synthetic method reference is made to the preparation of intermediate 10. ESI-MS m/z 307.2[ M+H ]] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (1- (1-methylpiperidin-4-yl) -1,2,3, 6-tetrahydropyridin-4-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (intermediate 40):
preparation of synthetic method reference intermediate 7。ESI-MS m/z:562.2[M+H] +
Preparation of 2- (5-fluoro-2-hydroxyphenyl) -2- (6- (1- (1-methylpiperidin-4-yl) -1,2,3, 6-tetrahydropyridin-4-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide (example 12):
synthetic method reference is made to the preparation of example 1. White solid, ESI-MS m/z 562.2[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ7.62(s,1H),7.38-7.29(m,1H),7.23-7.19(m,1H),7.19-7.11(m,2H),7.04-6.93(m,1H),6.84(s,1H),6.81-6.77(m,1H),6.32(q,J=6.2Hz,1H),5.97-5.62(m,2H),4.67-4.43(m,1H),4.14-3.92(m,1H),3.70-3.39(m,2H),2.90-2.80(m,1H),2.72(ddd,J=11.5,11.0,5.5Hz,2H),2.58-2.49(m,3H),2.39(ddd,J=12.0,11.0,5.5Hz,2H),2.25(s,3H),2.13(dt,J=15.2,7.7Hz,1H),1.99-1.87(m,2H),1.80-1.63(m,2H).
Example 13:
2- (5-fluoro-2-hydroxyphenyl) -2- (6- (1 '-methyl- [1,4' -piperidinyl ] -4-yl) -1-oxoisoindol-2-yl) -N- (thiazol-2-yl) acetamide
Synthetic method reference is made to the preparation of example 10. White solid, ESI-MS m/z 564.2[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ7.81(s,1H),7.61-7.54(m,1H),7.39(s,1H),7.35-7.31(m,1H),7.13(s,1H),6.97(d,J=4.2Hz,1H),6.86-6.76(m,2H),5.83(s,2H),5.13-4.97(m,1H),4.84-4.59(m,1H),2.87-2.77(m,3H),2.71(ddd,J=11.5,11.0,5.5Hz,2H),2.47-2.32(m,4H),2.26(s,3H),2.19-2.06(m,3H),2.00-1.89(m,2H),1.88-1.78(m,2H),1.73(ddt,J=16.6,7.8,5.6Hz,2H).
Example 14: biological assay analysis
1. Evaluation of the inhibition of EGFR kinase by the compounds of the present invention:
The measuring process comprises the following steps:
the compound kinase IC50 was measured using a mobility change method with JBJ-04-125-02 and JBJ-09-063 as positive references. The experimental reagents are shown in Table 2, and the experimental instruments are shown in Table 3.
Table 2 experimental reagents
Reagent name Goods number Suppliers of goods
EGFR L858R/T790M/C797S 08-115 Carna
Caliper substrate 18 114202 GL
384-well plate 3573 Corning
Dimethyl sulfoxide D8418-1L Sigma
Table 3 laboratory apparatus
/>
The experimental steps are as follows:
1) Preparation of compound solution: the weighed compounds were prepared in EP tubes using a dimethylsulfoxide solvent to give a 10mM stock solution, which was stored in a-20℃refrigerator in the absence of light for further use. The mother liquor was diluted to the concentration required for the experiment and transferred to 250nL to 384 well plates using Echo 550.
2) Preparation of kinase buffer and kinase solution: preparing kinase buffer solution, and preparing kinase solution by using the kinase buffer solution. The kinase solution is added to the experimental hole, the positive control hole and the blank control hole, the kinase buffer is added to the negative control hole, and the mixture is centrifuged and incubated for 10 minutes at room temperature.
3) The reaction is initiated: preparing a mixed solution of ATP and a kinase substrate by using a kinase buffer solution, adding the mixed solution to initiate a reaction, centrifuging after the addition, and incubating for 40 minutes at room temperature.
4) Terminating the reaction: the reaction was terminated by adding a stop solution and the conversion was measured.
5) Inhibition and IC50 calculation: data were processed with GraphPad Prism 5 and inhibition and IC50 values were calculated.
TABLE 4 test compound kinase inhibitory Activity
Test article Test object IC 50 (nmol/L)
JBJ-04-125-02 1.2
JBJ-09-063 0.87
Example 1 0.65
Example 2 0.71
Example 3 0.35
Example 4 0.72
Example 5 0.67
Example 6 0.59
Example 7 0.81
Example 8 0.85
Example 9 0.54
Example 10 0.63
Example 11 0.58
Example 12 0.51
Example 13 0.44
2. Cancer cell model assay
HTRF Phospho EGFR assay (cellular) cell lines and media: baF3 cell line (EGFRL 858R/T790M/C797S mutant, a type of mutation common in non-small cell lung cancer). At 37℃with 5% CO 2 Cells were kept in RPMI ATCC+2mM glutamine+0.5 μg/ml puromycin supplemented with 10% Fetal Bovine Serum (FBS) (Gibco).
The operation flow is as follows:
after pre-filling the Greiner bione, nr.78408 microtiter plate with 12.5nl of DMSO solution of the compound to be tested (dose response) or DMSO alone, cells were transferred to the plate at 20000 cells/well and 12.5 μl growth medium/well. After 30 seconds of plate rotation at 300Xg, the cells were incubated at 37℃with 5% CO 2 Incubate for 4 hours at 95% humidity. Cell lysis was performed by adding 4. Mu.l/well of supplemental lysis buffer (Cisbio, phospho EGFR HTRF kit, 64EG1 PEH) to the compound mixture, followed by incubation at room temperature for 30 minutes with shaking (400 rpm). The plates were then frozen and stored overnight at 80 ℃. The next day and after thawing the plates, 4 μl of a mixture solution of anti-Phospho EGFR compound and anti-Phospho EGFR d2 antibody prepared in the supplied assay buffer was added to each well. The capped plates were then incubated at room temperature for 4 hours, after which the fluorescent emissions at 616 and 665nm were read using an Envision plate reader (Perkin Elmer). The data was analyzed in a similar manner as described above using a normalized ratio of 665 to 616 signals multiplied by 10000.
The results are shown in Table 5.
Table 5H1975, baF3 cell HTRF Phospho EGFR TMLRCS assay data
Test article BaF3IC 50 (nmol/L)
JBJ-04-125-02 50
JBJ-09-063 15
Example 1 10
Example 2 9.5
Example 3 5.3
Example 4 11
Example 5 8.2
Example 6 12
Example 7 10.5
Example 8 12
Example 9 7
Example 10 10
Example 11 9.6
Example 12 7.2
Example 13 6.5
The results show that all compounds of the invention are EGFR inhibitors.
For compounds of the general formula (I), the linking groups and the substituents have an important influence on the pharmacodynamic properties of the compounds. While the invention has been illustrated by the foregoing specific examples, it should not be construed as being limited thereto; but rather the invention encompasses the generic aspects previously disclosed. Various modifications and embodiments can be made without departing from the spirit and scope of the invention.

Claims (10)

1. A compound having a structure represented by the following general formula (I):
ring A is a substituted or unsubstituted benzene ring, and ring B is optionally self-substituted
Alternatively, ring A is a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted C36 cycloalkyl, and ring B is optionally self-substituted
Wherein the substituents on the benzene ring, heterocyclyl or C36 cycloalkyl are selected from: c1-4 alkyl; n is 1-4;
R 1 、R 2 、R 3 、R 1 ’、R 2 ’、R 3 ’、R 4 ' are each independently selected from H, C1-4 alkyl, deuterated C1-4 alkyl; r is R 4 H, C2-4 alkyl, deuterated C2-4 alkyl; r is R 1 、R 2 、R 3 、R 4 、R 1 ’、R 2 ’、R 3 ’、R 4 The term "substituent" includes mono-, di-, tri-, or tetra-substitution.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the heterocyclyl comprises:
3. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the C36 cycloalkyl comprises:
4. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein when ring a is a benzene ring, ring B is selected from the group consisting of:
5.the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein when ring a is a substituted or unsubstituted heterocyclyl, a substituted or unsubstituted C36 cycloalkyl, ring B is
6. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein the structure of the compound is specifically selected from:
7. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the pharmaceutically acceptable salt is an inorganic salt or an organic salt; inorganic salts include hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate, acid phosphate; the organic salt is selected from acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate, maleate, lactate, malate, citrate, tartrate, methanesulfonate, sulfonate, benzenesulfonate, salicylate.
8. A pharmaceutical composition comprising a compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient or diluent.
9. Use of a compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for use as an EGFR inhibitor.
10. Use of a compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
CN202310398689.7A 2023-04-14 2023-04-14 Synthesis and application of EGFR inhibitor Pending CN116693518A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310398689.7A CN116693518A (en) 2023-04-14 2023-04-14 Synthesis and application of EGFR inhibitor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310398689.7A CN116693518A (en) 2023-04-14 2023-04-14 Synthesis and application of EGFR inhibitor

Publications (1)

Publication Number Publication Date
CN116693518A true CN116693518A (en) 2023-09-05

Family

ID=87836333

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310398689.7A Pending CN116693518A (en) 2023-04-14 2023-04-14 Synthesis and application of EGFR inhibitor

Country Status (1)

Country Link
CN (1) CN116693518A (en)

Similar Documents

Publication Publication Date Title
JP6783663B2 (en) New glutaminase inhibitor
US6624174B2 (en) 2-amino-nicotinamide derivatives and their use as VEGF-receptor tyrosine kinase inhibitors
EP3181560B1 (en) Pyridine amidopyrimidine derivative, preparation method and use thereof
KR101749192B1 (en) Triazine, pyrimidine and pyridine analogs and their use as therapeutic agents and diagnostic probes
RU2638540C1 (en) Dna-pk inhibitors
TW201838966A (en) Substituted dihydroindene-4-carboxamides and analogs thereof, and methods using same
CN111362967B (en) Benzoxadiazatetetradecene derivatives and use thereof
CN113195471B (en) Polysubstituted pyridone derivative and application thereof in medicine
KR20090090365A (en) Pyridine or pyrimidine derivative having excellent cell growth inhibition effect and excellent anti-tumor effect on cell strain having amplification of hgfr gene
WO2011035540A1 (en) 4-(substituted anilino)quinazoline derivatives as tyrosine kinase inhibitors
KR20080006614A (en) 2-amino-quinazolin-5-ones as hsp90 inhibitors useful in treating proliferation diseases
US20060281763A1 (en) Carboxamide inhibitors of TGFbeta
JP7254094B2 (en) Substituted imidazolidin-2-one derivatives as PRMT5 inhibitors
EP4165035A1 (en) Heteroaryl alkylene substituted 2-oxoquinazoline derivatives as methionine adenosyltransferase 2a inhibitors
CN113166119A (en) Substituted aryl compound and preparation method and application thereof
WO2019134573A1 (en) Method for preparing deuterated diphenylaminopyrimidine compound and crystal form thereof
CN116283953B (en) Indoline compound containing thiazole structure, and preparation method and application thereof
CN112745298A (en) Polysubstituted isoindoline compound, preparation method, pharmaceutical composition and application thereof
CN110684016A (en) Fluorine-containing AZD9291 derivative and preparation method and application thereof
CN116693518A (en) Synthesis and application of EGFR inhibitor
KR20120060871A (en) 8-oxodihydropurine derivative
CN111362924B (en) Deuterated pyrimidine derivatives and uses thereof
WO2022194265A1 (en) Quinazoline-based compound, composition, and application of quinazoline-based compound
CN115960117B (en) Sulfur-containing fused ring derivative inhibitor, preparation method and application thereof
CN115894486B (en) Hydrogenated pyridoquinazoline compound, composition and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination