CN117865984A - Naphthalene compound and preparation method and medical application thereof - Google Patents

Naphthalene compound and preparation method and medical application thereof Download PDF

Info

Publication number
CN117865984A
CN117865984A CN202410068136.XA CN202410068136A CN117865984A CN 117865984 A CN117865984 A CN 117865984A CN 202410068136 A CN202410068136 A CN 202410068136A CN 117865984 A CN117865984 A CN 117865984A
Authority
CN
China
Prior art keywords
acid
pharmaceutically acceptable
compound
usp7
naphthalene
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
CN202410068136.XA
Other languages
Chinese (zh)
Inventor
温小安
朱华云
张凤仪
孙宏斌
陈彩萍
张弦
王大林
谭淑丹
陈迪
陈琪
张鸿瀚
梁子江
袁浩亮
许庆龙
刘鎏
戴量
冯志奇
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.)
Innovation Research Institute Of Chongqing China Pharmaceutical University
China Pharmaceutical University
Original Assignee
Innovation Research Institute Of Chongqing China Pharmaceutical University
China Pharmaceutical 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 Innovation Research Institute Of Chongqing China Pharmaceutical University, China Pharmaceutical University filed Critical Innovation Research Institute Of Chongqing China Pharmaceutical University
Priority to CN202410068136.XA priority Critical patent/CN117865984A/en
Publication of CN117865984A publication Critical patent/CN117865984A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Transplantation (AREA)

Abstract

The invention discloses naphthalene compoundsThe invention provides naphthalene compounds shown in a formula (I) or pharmaceutically acceptable salts, esters, stereoisomers, deuterated compounds or solvates thereof. The compound of the present invention or pharmaceutically acceptable salts, esters, deuterates, stereoisomers or solvates thereof inhibit the activity of USP7 enzyme and may be used in the manufacture of a medicament for the treatment of USP7 mediated diseases.

Description

Naphthalene compound and preparation method and medical application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to naphthalene compounds, a preparation method and medical application thereof.
Background
Ubiquitin Specific Proteases (USP) are the largest class of deubiquitinase families, with USP7 (also called HAUSP) being one of the most interesting deubiquitinating enzymes. USP7 cysteine proteases, which are located mainly in the nucleus, regulate the stability of proteins involved in various cellular processes such as DNA damage response, transcription, epigenetic gene expression, immune response and viral infection, and are closely related to the development and progression of many diseases. For example, USP7 is overexpressed in a variety of tumor cells, stabilizing the MDM2 protein by deubiquitination, whereas MDM2 as a major negative regulator of p53 protein not only directly inhibits the transcriptional activity of p53, but also reduces p53 levels by ubiquitination. Knocking out or inhibiting USP7 in tumor cells can reduce MDM2 protein level, raise p53 level and inhibit proliferation of tumor cells. Furthermore, USP7 can stabilize Foxp3 protein by deubiquitination, and Foxp3 is a key transcriptional regulatory protein that determines immunosuppressive regulatory T cell (Treg) differentiation and function. Knocking out or inhibiting USP7 can reduce Foxp3 protein level, reduce the number and activity of Treg cells in tumor microenvironment, thereby reducing the immune suppression of tumor cells mediated by Treg, and further increasing the anti-tumor immune response of the organism. In conclusion, inhibition of USP7 is expected to be a new approach for treating diseases such as tumors. In recent years, several USP7 small molecule inhibitors and their crystal structure with USP7 complexes have been sequentially published, and these structures provide guidance for obtaining structure-based small molecule inhibitors. However, these USP7 inhibitors have not been clinically tested at present because of unsatisfactory in vivo efficacy data. Therefore, there is a need for developing USP7 inhibitors with good in vivo activity for early use in the treatment of tumor patients with abnormal USP7 expression.
Disclosure of Invention
The invention aims to: aiming at the problems existing in the prior art, the invention provides naphthalene compounds, which can be used as USP7 inhibitors, have good inhibitory activity on USP7 enzyme and can be used as medicaments for treating USP7 mediated diseases, or pharmaceutically acceptable salts, esters, stereoisomers, deuterated compounds or solvates thereof.
The technical scheme is as follows: in order to achieve the above object, the present invention provides naphthalene compounds represented by formula (I) or pharmaceutically acceptable salts, esters, deuterides, stereoisomers or solvates thereof:
wherein L is 1 Selected from chemical bond, -CH 2 -, -O-, -S-or-NH-or- (CO) -;
ring a is selected from a 5-6 membered aryl or heteroaryl, a 3-6 membered cycloalkyl or heterocycloalkyl, which may optionally be substituted with (=o), halogen, -CN, -NO 2 、-CF 3 、-OR 3 、-SR 3 、-NR 3 R 4 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl;
R 1 selected from C 1-6 Alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, 5-6 membered aryl or heteroaryl, which alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl may optionally be substituted with (=o), halogen, -CN, -NO 2 、-CF 3 、-OR 3 、-SR 3 、-NR 3 R 4 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl;
R 2 selected from H, halogen, -CN, -CF 3 Or C 1-6 An alkyl group;
preferably selected from any one of the following structural fragments:
R 1 preferably selected from any one of the following structural fragments:
in some preferred embodiments, the compound of formula I, or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate, or solvate thereof, is selected from any one of the following tables, the structure of which is detailed in table 1:
TABLE 1
Wherein the salt may be an acid salt of at least one of galactose diacid, D-glucuronic acid, glycerophosphate, hippuric acid, isethionic acid, lactobionic acid, maleic acid, 1, 5-naphthalene disulfonic acid, naphthalene-2-sulfonic acid, pivalic acid, terephthalic acid, thiocyanic acid, cholic acid, n-dodecylsulfuric acid, benzenesulfonic acid, citric acid, D-glucose, glycolic acid, lactic acid, malic acid, malonic acid, mandelic acid, phosphoric acid, propionic acid, hydrochloric acid, sulfuric acid, tartaric acid, succinic acid, formic acid, hydroiodic acid, hydrobromic acid, methanesulfonic acid, nicotinic acid, nitric acid, orotic acid, oxalic acid, picric acid, L-pyroglutamic acid, saccharinic acid, salicylic acid, gentisic acid, p-toluenesulfonic acid, valeric acid, palmitic acid, stearic acid, lauric acid, acetic acid, adipic acid, carbonic acid, 4-benzenesulfonic acid, ethanedisulfonic acid, ethylsuccinic acid, fumaric acid, 3-hydroxynaphthalene-2-carboxylic acid, 1-hydroxynaphthalene-2-carboxylic acid, oleic acid, undecylenic acid, ascorbic acid, camphoric acid, dichloroacetic acid, ethanesulfonic acid.
Further, the salts are salts of the compounds of the present invention with metal (including sodium, potassium, calcium, etc.) ions or pharmaceutically acceptable amines (including ethylenediamine, tromethamine, etc.) or ammonium ions.
Use of the compound or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate or solvate thereof in the preparation of a USP7 inhibitor.
Use of the compound or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate or solvate thereof in the manufacture of a medicament for the prevention or treatment of a USP7 mediated disease.
Wherein the USP7 mediated disease is any one of a tumor, an infectious disease, an inflammatory disease, or an autoimmune disease.
Such neoplastic diseases include, but are not limited to: bone cancer, acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome, hodgkin's lymphoma, non-hodgkin's lymphoma, hemangioma, granuloma, xanthoma, meningioma, glioma, neuroblastoma, astrocytoma, neuroblastoma, ependymoma, germ cell tumor (pineal tumor), glioblastoma multiforme, oligodendroglioma, schwannoma, retinoblastoma, fibroneuroma, sarcoma, esophageal carcinoma, gastric carcinoma, pancreatic carcinoma, colorectal carcinoma, colon carcinoma, rectal carcinoma, renal carcinoma, prostate carcinoma, lymphoma, testicular carcinoma, interstitial cell carcinoma, lung carcinoma, liver carcinoma, skin carcinoma, malignant melanoma, basal cell carcinoma, and the like.
Such viral infectious diseases include, but are not limited to, infectious diseases caused by the following viruses: mycobacterium tuberculosis infection, chlamydia infection, herpes virus (herpes simplex virus) infection, adenovirus infection, hepatitis B virus infection, orthomyxovirus infection, coronavirus infection, bacterial infection, mycoplasma infection, etc.
The inflammation, autoimmune diseases include, but are not limited to, ulcerative colitis, crohn's disease, systemic lupus erythematosus, rheumatoid arthritis, psoriasis, multiple sclerosis, or Behcet's disease.
The invention provides a pharmaceutical composition, which comprises a compound shown in a general formula (I) or pharmaceutically acceptable salt, ester, stereoisomer, deuteride or solvate thereof and a pharmaceutically acceptable carrier.
Further, the carrier is any one of excipient, binder, disintegrating agent, lubricant, flavoring agent, coloring agent or sweetener.
Further, the pharmaceutical composition is a capsule, powder, tablet, granule, pill, injection, syrup, oral liquid, inhalant, ointment, suppository or patch.
Preferably, the pharmaceutical composition is an oral dosage form due to the high oral bioavailability of the compounds of the invention.
The deuterated forms of the compounds described herein, i.e., each hydrogen atom attached to a carbon atom, may be independently substituted with a deuterium atom.
The present invention includes various prodrug forms of the compounds of the present invention.
In certain embodiments, the compounds of the present invention may be used in combination with one or more other types of agents for the prevention or treatment of the above-described diseases, including but not limited to the following combinations:
other types of prophylactic or therapeutic agents that may be optionally used in combination with the compounds of the invention may be one or more anticancer agents including alkylating agents (e.g., cisplatin, cyclophosphamide, ifosfamide, melphalan, chlorambucil, bendamustine, estramustine, thiotepa, iminoquinone, busulfan, dibromomannitol, cyclohexen, carmustine, pyrimideimide, mechlorethamine, procarbazine, etc.), antimetabolites (e.g., fluorouracil, cytarabine, furfurfururacil, bifluoride, mercaptopurine sodium, azathioprine, thioguanine, methotrexate, aminopterin, etc.), antitumor antibiotics (e.g., mitomycin C, bleomycin, actinomycin D, mithramycin, doxorubicin, chromycin A3, neocarcinomycin, anticancer drugs, dactinomycin, etc.), natural drugs (e.g., vincristine, flunine, indomethacin, benzone, hydrobromide, benzodiazepine, etc.), antimuscarin, etc.), antimetabins (e, such as flunine, fludrozole, flupirone, etc.), drugs (e, such as fludrozole, flupirone, etc.; PD-L1 inhibitor atezolizumab, durvalumab and avelumab, etc., CTLA-4 inhibitor Ipilimumab, etc., other immune checkpoint inhibitors, immunotherapeutic agents such as cytotherapeutic agents, etc.), antibody drug conjugates (e.g., kadcyla, etc.), kinase inhibitors (e.g., SHP-2 inhibitors, B-RAF inhibitors, MEK inhibitors, BTK inhibitors, etc.), and the like, IDO inhibitors (e.g., epacoadostat), and the like.
The beneficial effects are that: compared with the prior art, the invention has the following remarkable advantages:
1. the naphthalene compound or pharmaceutically acceptable salts, esters, stereoisomers, deuterated compounds or solvates thereof provided by the invention have strong inhibitory activity on USP7 enzyme; can be used for preparing medicines for preventing or treating USP7 mediated diseases.
2. The naphthalene compound or pharmaceutically acceptable salt, ester, stereoisomer, deuteride or solvate thereof provided by the invention has the advantages of simple structure, ingenious design, cheap and easily obtained raw materials, safe and environment-friendly synthesis process and easiness in large-scale production.
Detailed Description
The technical scheme of the invention is further described below.
Materials, reagents and the like used in the examples described below are commercially available unless otherwise specified. The experimental methods for which specific conditions are not specified in the examples are generally conducted under conventional conditions or under conditions recommended by the manufacturer.
Example 1
3- ((7- (4-chloro-1- (piperidin-4-yloxy) naphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-1)
7-chlorothiophene [3,2-b ] pyridine (CAS number: 69627-03-8) (A1) (10 g,59 mmol) was dissolved in anhydrous tetrahydrofuran (100 mL), replaced with argon three times, cooled to-78 ℃, N-butyllithium (2.5M in hexane,30.6mL,76.63mmol) was slowly added dropwise with a syringe pump, the dropwise addition was stirred at-78 ℃ for 1h, then anhydrous N, N-dimethylformamide (15.3 mL,194.53 mmol) was slowly added dropwise, the reaction was slowly warmed to room temperature after the dropwise addition, TLC monitoring was completed, saturated ammonium chloride solution was added dropwise to the system for quenching, then the solvent was distilled off under reduced pressure, 150mL of water was added to pulp, suction filtration was performed, the filter cake was washed with a small amount of water, and a earthy yellow solid (11.5 g, crude product yield 99%) of compound A2 was obtained after drying, and the crude product was directly thrown into the next step without purification.
Compound A2 (11.5 g,59 mmol) was added to methanol (150 mL), sodium borohydride (4.46 g,118 mmol) was added in portions under ice-bath, then slowly warmed to room temperature for reaction, TLC monitored the completion of the reaction, quenched with saturated ammonium chloride solution, the solvent was distilled off under reduced pressure, the residue was slurried with 100mL of water, suction filtered, the filter cake was washed with a small amount of water, and dried to give compound A3 as a yellow solid (10.4 g, crude yield 89.6%) which was directly fed to the next reaction without purification.
Compound A3 (10.4 g,52.86 mmol) was added to 150mL of anhydrous dichloromethane, and thionyl chloride (11.5 mL,158.58 mmol) was slowly added dropwise under ice-bath and reacted overnight at room temperature after the dropwise addition. TLC monitored the reaction, quenched by slow addition of saturated sodium bicarbonate solution in ice bath, extracted with dichloromethane (80 ml×2), the combined organic phases were washed with saturated aqueous sodium chloride (100 ml×2), dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=15:1) to give compound A4 as a white solid (8.21 g, 72.2% yield).
Compound A4 (8.21 g,37.64 mmol) and 6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (CAS number: 69627-03-8) (A5) (5.5 g,39.52 mmol) were dissolved in 120mL of acetonitrile, potassium carbonate (10.39 g,75.28 mmol) was added, and the temperature was raised to 80℃for reaction. After the completion of the reaction, the reaction mixture was cooled to room temperature by TLC, 150mL of water was added, extraction was performed with methylene chloride (80 mL. Times.3), and the combined organic phases were washed with saturated aqueous sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to give compound A6 as a pink solid (11.34 g, yield 93.9%).
Compound A6 (11.34 g,35.35 mmol), pinacol ester of diboronic acid (A7) (8.98 g,35.35 mmol), [1,1' -bis (diphenylphosphine) ferrocene ] palladium dichloride (CAS number: 72287-26-4) (1.72 g,2.12 mmol) and potassium acetate (10.41 g,106.05 mmol) were added to 110mL of anhydrous dimethyl sulfoxide, replaced with argon three times, and the temperature was raised to 120℃for reaction. After the completion of the TLC monitoring, the reaction solution was cooled to room temperature, diluted with 100mL of ethyl acetate, suction-filtered with celite, 300mL of water was added to the filtrate, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with water (200 mL. Times.2) and saturated aqueous sodium chloride solution (200 mL. Times.2), respectively, and after the solvent was distilled off under reduced pressure, 150mL of n-hexane was added to pulp, suction-filtered, the filter cake was washed with a small amount of n-hexane, and dried to give a dark brown solid (11.45 g) of Compound A8, which was directly used for the next reaction without purification.
4-chloro-1-naphthol (B1) (800 mg,4.5 mmol) and tetrabutylammonium tribromide (2.3 g,4.7 mmol) were added to a dried three-necked flask, replaced with argon three times, and 45ml of anhydrous dichloromethane was added to react at room temperature. After the completion of the TLC monitoring, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether) to give compound B2 as a white powdery solid (1.03 g, yield 88.88%).
Compound B2 (150 mg,0.58 mmol), N-Boc-4-hydroxypiperidine (B3) (140 mg,0.70 mmol) and triphenylphosphine (199mg, 0.76 mmol) were added to a dry Schlenk tube, replaced three times with argon, 4mL of anhydrous tetrahydrofuran was added, diisopropyl azodicarboxylate (0.15 mL,0.76 mmol) was slowly added dropwise under ice bath, and the reaction was completed at room temperature. After TLC monitoring the reaction, the solvent was distilled off directly under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=50:1) to give compound B4 as a pale yellow oil (239 mg, yield 93.49%).
Compound B4 (158 mg,0.36 mmol), compound A8 (441 mg,1.07 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (CAS number: 72287-26-4) (14.7 mg,0.018 mmol) and potassium carbonate (99 mg,0.72 mmol) were added to a mixed solvent of 4mL of dioxane and 0.7mL of water, and the mixture was replaced with argon 3 times, and the reaction was heated at 100 ℃. After TLC monitoring the reaction, the reaction was cooled to room temperature, 10mL of water was added, extracted with ethyl acetate (10 ml×3), the combined organic phases were washed with saturated aqueous sodium chloride (10 ml×2), the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=1:1) to give compound B5 as a pale yellow oil (192 mg, crude yield 82.53%), which was directly used in the next reaction.
Compound B5 (192 mg,0.30 mmol) was dissolved in 3mL of dichloromethane, and a dioxane solution (1.1 mL,4.44 mmol) of hydrogen chloride was slowly added dropwise under ice-bath, and the reaction was completed at room temperature. TLC monitored the reaction, diluted with 5mL of dichloromethane, then neutralized with saturated aqueous sodium bicarbonate, extracted with dichloromethane (5 ml×3), the combined organic phases were washed with saturated aqueous sodium chloride (10 ml×1), the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol=15:1) to give compound I-1 as a white solid (60 mg, yield 36.6%). 1 H NMR(400MHz,DMSO-d 6 )δ8.79(d,J=4.8Hz,1H),8.29(t,J=7.4Hz,2H),7.87–7.78(m,1H),7.56(d,J=4.3Hz,2H),4.82(s,2H),3.95–3.87(m,1H),2.81–2.73(m,1H),2.60–2.53(m,2H),1.66–1.57(m,2H),1.46–1.37(m,6H),1.16(s,3H),1.00(s,3H)。
Example 2
3- ((7- (4-chloro-1- (piperidin-3-yloxy) naphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-2)
The procedure of example 1 was followed except that compound B2 was reacted with N-Boc-3-hydroxypiperidine to give compound I-2 (36 mg, yield 61.2%) as a white solid. 1 H NMR(400MHz,Chloroform-d)δ8.75(d,J=4.8Hz,1H),8.29(d,J=9.0Hz,2H),7.64(s,2H),7.60(d,J=15.4Hz,2H),7.41(d,J=4.8Hz,1H),4.84(s,2H),3.63–3.54(m,1H),2.71(s,1H),2.62(s,1H),2.49(dt,J=14.2,6.8Hz,2H),2.36(s,2H),1.54(d,J=6.5Hz,2H),1.45–1.34(m,2H),1.22(s,3H),1.11(s,3H)。
Example 3
3- ((7- (4-chloro-1- (piperidin-4-ylmethoxy) naphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-3)
The preparation of example 1 was followed, except that compound B2 was reacted with N-Boc-4-piperidinemethanol to give compound I-3 (white solid 85mg, yield 51.6%). 1 H NMR(300MHz,Chloroform-d)δ8.74(d,J=4.8Hz,1H),8.28(ddd,J=17.2,8.2,1.5Hz,2H),7.68(ddd,J=9.8,8.0,1.4Hz,2H),7.59(d,J=8.3Hz,2H),7.37(d,J=4.8Hz,1H),4.83(s,2H),3.36(d,J=6.1Hz,2H),2.95(d,J=11.9Hz,2H),2.51–2.40(m,2H),2.36(s,2H),1.67–1.48(m,2H),1.46–1.36(m,3H),1.22(s,3H),1.11(s,3H)。
Example 4
3- ((7- (4-chloro-1- (piperidin-3-ylmethoxy) naphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-4)
The preparation of example 1 was followed, except that compound B2 was reacted with N-Boc-3-piperidinemethanol to give compound I-4 (white solid 85mg, yield 39.2%). 1 H NMR(300MHz,DMSO-d 6 )δ8.78(d,J=4.8Hz,1H),8.75(s,1H),8.26(t,J=9.3Hz,2H),7.92–7.75(m,3H),7.59–7.50(m,2H),4.81(s,2H),3.51–3.38(m,2H),3.11(d,J=12.0Hz,1H),2.97(d,J=12.1Hz,1H),2.77–2.60(m,1H),2.59(s,2H),2.37(t,J=12.0Hz,1H),2.05–1.90(m,1H),1.65–1.45(m,2H),1.41–1.22(m,2H),1.16(s,3H),1.01(s,3H)。
Example 5
3- ((7- (1- (azepan-4-yloxy) -4-chloronaphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-5)
With reference to the preparation of example 1, the only difference was that compound B2 was reacted with t-butyl 4-hydroxyazepan-1-carboxylate to give compound I-5 (67 mg, 45.5% yield as a white solid). 1 H NMR(300MHz,DMSO-d 6 )δ8.79(d,J=4.8Hz,1H),8.72–8.67(m,1H),8.28(dd,J=8.0,6.0Hz,2H),7.89–7.76(m,3H),7.59–7.53(m,2H),4.81(s,2H),3.96–3.88(m,1H),2.94–2.77(m,3H),2.63–2.54(m,3H),1.82–1.71(m,2H),1.62–1.50(m,2H),1.49–1.37(m,1H),1.18–1.12(m,4H),1.00(s,3H)。
Example 6
3- ((7- (1- (3-aminopropoxy) -4-chloronaphthalen-2-yl) thieno [3,2-b ] pyridin-2-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (I-6)
With reference to the preparation of example 1, the only difference was that compound B2 was reacted with tert-butyl N- (3-hydroxypropyl) carbamate to give compound I-6 (44 mg of white solid, 58.9% yield). 1 H NMR(400MHz,DMSO-d 6 )δ8.79(d,J=4.8Hz,1H),8.29(dd,J=15.8,8.3Hz,2H),7.90–7.81(m,4H),7.78(d,J=1.4Hz,1H),7.58–7.52(m,2H),4.81(s,2H),3.61(t,J=6.1Hz,2H),2.58(s,2H),2.55–2.51(m,2H),1.79–1.67(m,2H),1.16(s,3H),1.01(s,3H)。
Example 7
Compounds I-7 to I-20 were synthesized by the method described in example 1 and are shown in Table 2.
TABLE 2 Structure of Compounds I-7 to I-20 and analytical data
Example 8
Test of inhibitory Activity of Compounds against in vitro USP7 enzyme
1. Preparation work
Enzyme activity test buffer: buffers containing 50mM HEPES, 0.5mM EDTA, 100mM NaCl, 1mM TCEP, 0.1mg/mL BSA were prepared with deionized water and adjusted to pH 7.5 with sodium hydroxide solution.
Preparing a compound solution to be tested: the test compound (I-1 to I-20 prepared in examples 1 to 7) was weighed in an appropriate amount, dissolved in DMSO, and diluted 3 times in sequence to obtain 8 concentration test compound solutions. The appropriate amount of positive compound was weighed, dissolved in DMSO, and diluted 3-fold sequentially to obtain 8 positive compound solutions at a concentration (up to 250 μm). The compound solution and DMSO (blank) are diluted 62.5 times by enzyme activity test buffer to obtain compound solution to be tested.
Preparing a USP7 protein solution: commercially available USP7 solution (0.7. Mu.g/. Mu.l, recombinant USP7 Protein, active Motif, catalog No. 31525,Lot No:08215001) was diluted to 20pM with enzyme activity test buffer gradient to give a USP7 Protein solution for testing.
Preparing Ub-Rho solution: a commercially available Ub-Rho solution (2.490 mg/ml, boston Biochem, U-555-050,Lot No:DBFH2322121) was diluted 500-fold with an enzyme activity test buffer to give a Ub-Rho solution for testing.
2. Sample addition and incubation
To the experimental well of 384-well plate (Thermo Scientific, lot No: 1207363), USP7 protein solution (10. Mu.l) and a solution of a test compound at different concentrations (5. Mu.L) were added each of which was 4-fold wells, and the resulting mixture was incubated in a dark place at room temperature for 30 minutes. Ub-Rho solution (5. Mu.L) was added to 2 wells of each concentration, and enzyme activity test buffer (5. Mu.L) was added to the other 2 wells. After addition, 384-well plates were incubated at 37℃for 2h in the dark. The positive control was a USP7 inhibitor Compound 4 (CP 4) reported in literature (Nat Chem bio1,2018,14 (2), 118).
3. Reading and calculation
Fluorescence data were read with a multifunctional plate reader at excitation wavelength 485nm and emission wavelength 535 nm.
Inhibition% Blank (Ub-Rho) -F Blank (buffer) )-(F Compound (Ub-Rho) -F Compound (buffer) )]/(F Blank (Ub-Rho) -F Blank (buffer) )×100%
IC 50 The values were calculated using GraphPad Prism software.
4. Experimental results
The inhibitory activity data for some compounds against USP7 are shown in table 3 below.
TABLE 3 inhibition of USP7 by partial Compounds
Numbering of compounds IC 50 (nM)
CP4 26.43±0.43
I-1 130.9
I-2 147.9
I-3 414.3
I-4 951.5
I-5 50.4
As shown in Table 3, naphthalene compounds of the present invention are effective in inhibiting USP7 enzyme activity, IC 50 The values all reached nanomolar levels where the inhibitory activity of compound I-5 was close to that of positive control CP 4.
Example 9
Preparation of tablets
The compound I-5 (5 g) prepared in example 5 of the present invention, hydroxypropylmethyl cellulose E (150 g), starch (200 g), povidone K30 in an appropriate amount and magnesium stearate (1 g) were mixed, granulated and tableted.
In addition, the compounds I-1 to I-20 can be endowed with different pharmaceutical excipients according to the conventional preparation method of pharmacopoeia 2015 and prepared into capsules, powder, granules, pills, injections, syrups, oral liquids, inhalants, ointments, suppositories or patches and the like.

Claims (10)

1. Naphthalene compounds represented by formula (I) or pharmaceutically acceptable salts, esters, deuterates, stereoisomers or solvates thereof:
wherein L is 1 Selected from chemical bond, -CH 2 -, -O-, -S-or-NH-or- (CO) -;
ring a is selected from a 5-6 membered aryl or heteroaryl, a 3-6 membered cycloalkyl or heterocycloalkyl, which may optionally be substituted with (=o), halogen, -CN, -NO 2 、-CF 3 、-OR 3 、-SR 3 、-NR 3 R 4 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl;
R 1 selected from C 1-6 Alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, 5-6 membered aryl or heteroaryl, which alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl may optionally be substituted with (=o), halogen, -CN, -NO 2 、-CF 3 、-OR 3 、-SR 3 、-NR 3 R 4 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl;
R 2 selected from H, halogen, -CN, -CF 3 Or C 1-6 An alkyl group;
R 3 and R is 4 Each independently selected from H, C 1-6 Alkyl or C 3-8 Cycloalkyl groups.
2. The naphthalene compound according to claim 1, wherein said naphthalene compound is selected from the group consisting of pharmaceutically acceptable salts, esters, deuterides, stereoisomers and solvates thereofAny one of the following structural fragments:
3. the naphthalene compound according to claim 1, wherein R is selected from the group consisting of 1 Any one of the following structural fragments:
4. naphthalene compound according to claim 1, or a pharmaceutically acceptable salt, ester, deuterated, stereoisomer, or solvate thereof, wherein the compound is selected from any one of the following compounds:
5. the naphthalene compound of claim 1 or a pharmaceutically acceptable salt, ester, deuterate, stereoisomer, or solvate thereof, wherein the salt is an acid salt of at least one of the following acids: galactose diacid, D-glucuronic acid, glycerophosphate, hippuric acid, isethionic acid, lactobionic acid, maleic acid, 1, 5-naphthalene disulfonic acid, naphthalene-2-sulfonic acid, pivalic acid, terephthalic acid, thiocyanic acid, cholic acid, n-dodecyl sulfuric acid, benzenesulfonic acid, citric acid, D-glucose, glycolic acid, lactic acid, malic acid, malonic acid, mandelic acid, phosphoric acid, propionic acid, hydrochloric acid, sulfuric acid, tartaric acid, succinic acid, formic acid, hydroiodic acid, hydrobromic acid, methanesulfonic acid, nicotinic acid, nitric acid, orotic acid, oxalic acid, picric acid, L-pyroglutamic acid, saccharin acid, salicylic acid, gentisic acid, p-toluenesulfonic acid, valeric acid, palmitic acid, stearic acid, lauric acid, acetic acid, adipic acid, carbonic acid, 4-benzenesulfonic acid, ethanedisulfonic acid, ethylsuccinic acid, fumaric acid, 3-hydroxynaphthalene-2-carboxylic acid, 1-hydroxynaphthalene-carboxylic acid, oleic acid, undecylenic acid, ascorbic acid, camphoric acid, ethanesulfonic acid; or the salt is a salt of a compound with a metal ion or a pharmaceutically acceptable amine or ammonium ion.
6. The use of a compound according to any one of claims 1-4, or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate, or solvate thereof, for the preparation of a USP7 inhibitor.
7. The use of a compound according to any one of claims 1-4, or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate or solvate thereof, for the manufacture of a medicament for the prevention or treatment of a USP7 mediated disease.
8. The use according to claim 7, wherein the USP7 mediated disease is any one of a tumor, an infectious disease inflammatory disease or an autoimmune disease.
9. A pharmaceutical composition for the preparation of a medicament for the prevention or treatment of USP7 mediated diseases, comprising a compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt, ester, stereoisomer, deuterate or solvate thereof or a pharmaceutically acceptable carrier.
10. The pharmaceutical composition of claim 9, wherein the pharmaceutical composition is in the form of a capsule, powder, tablet, granule, pill, injection, syrup, oral liquid, inhalant, ointment, suppository, or patch.
CN202410068136.XA 2024-01-17 2024-01-17 Naphthalene compound and preparation method and medical application thereof Pending CN117865984A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410068136.XA CN117865984A (en) 2024-01-17 2024-01-17 Naphthalene compound and preparation method and medical application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410068136.XA CN117865984A (en) 2024-01-17 2024-01-17 Naphthalene compound and preparation method and medical application thereof

Publications (1)

Publication Number Publication Date
CN117865984A true CN117865984A (en) 2024-04-12

Family

ID=90584400

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410068136.XA Pending CN117865984A (en) 2024-01-17 2024-01-17 Naphthalene compound and preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN117865984A (en)

Similar Documents

Publication Publication Date Title
CA2927392C (en) Dna-pk inhibitors
JP6966423B2 (en) Condensed ring pyrimidine amino derivative, its production method, intermediate, pharmaceutical composition and application
AU2005332339B2 (en) Furanopyridine derivatives as ACK1 and Lck modulators
CA2934700A1 (en) Glutaminase inhibitors
JP6704422B2 (en) Quinazoline derivative salt and method for producing the same
JP2023535746A (en) Compounds used as CDK7 kinase inhibitors and their applications
CN113717156B (en) EGFR inhibitor, preparation method and application thereof
JP2020529968A (en) Substituted 5- and 6-membered heterocyclic compounds, their preparation methods, drug combinations and their use
EP3381925B1 (en) Deuterium-modified brigatinib derivatives, pharmaceutical compositions comprising same, and use thereof
CN110563703B (en) Compound for inducing PARP-1 degradation based on CRBN ligand, preparation method and application
CN112851663B (en) Parallel heterocyclic compound and application thereof
JP2022517723A (en) Macrocycle compound as a CDK inhibitor, its production method and its application in pharmaceutical products
EP1650194A1 (en) Protein kinase inhibitor
CN114524810B (en) Pyrimidine heterocyclic compounds, preparation method and application
CN103833756A (en) Pyridazinone compound as well as preparation method and application thereof
CN113166148B (en) Heterocyclic compounds as CDK-HDAC dual pathway inhibitors
Wu et al. Development and structure-activity relationship of tacrine derivatives as highly potent CDK2/9 inhibitors for the treatment of cancer
CN117865984A (en) Naphthalene compound and preparation method and medical application thereof
TW202045506A (en) Pyrazole compound
CN113999210B (en) Group of 2-phenylamino-4-triazolyl pyrimidine derivatives and application thereof
WO2023057371A1 (en) 1,2,4-oxadiazol-5-one derivatives for the treatment of cancer
CN117384161A (en) Compound for targeted degradation of CDK protein and application thereof
CN112174958B (en) Pyrido [2,3-d ] pyrimidine compound and preparation method and application thereof
CN113880814B (en) Pyrimidine amine compound and application thereof
CN117865983A (en) Indane compound, preparation method and medical 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