CN117603175A - Chalcone derivative with benzopyran structure, and preparation method and application thereof - Google Patents

Chalcone derivative with benzopyran structure, and preparation method and application thereof Download PDF

Info

Publication number
CN117603175A
CN117603175A CN202311595446.9A CN202311595446A CN117603175A CN 117603175 A CN117603175 A CN 117603175A CN 202311595446 A CN202311595446 A CN 202311595446A CN 117603175 A CN117603175 A CN 117603175A
Authority
CN
China
Prior art keywords
cancer
compound
membered
alkyl
alkyloxy
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
CN202311595446.9A
Other languages
Chinese (zh)
Inventor
于凡
赵炜
王瑞
金洪真
马冉
王丹阳
王琨
柯牧妍
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.)
Nankai University
Original Assignee
Nankai 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 Nankai University filed Critical Nankai University
Priority to CN202311595446.9A priority Critical patent/CN117603175A/en
Publication of CN117603175A publication Critical patent/CN117603175A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

The invention provides a chalcone derivative with a benzopyran structure, which has anti-inflammatory and anti-tumor activities, and can inhibit proliferation of tumor cells, promote apoptosis of the tumor cells, inhibit colony formation of the tumor cells and inhibit migration of the tumor cells.

Description

Chalcone derivative with benzopyran structure, and preparation method and application thereof
Technical Field
The invention belongs to the technical field of pharmaceutical compounds, and particularly relates to chalcone derivatives with benzopyran structures, a preparation method and pharmaceutical application thereof.
Background
Chalcones are simple chemical scaffolds for many natural compounds, widely distributed in vegetables, fruits, tea and other plants. Chalcone compounds have the common chemical scaffold 1,3 diaryl-2-propen-1-oneThe therapeutic use of chalcones dates back to thousands of years and treats different doctors through the use of plants and herbsDiseases such as cancer, inflammation and diabetes. Several chalcone-based compounds have been approved for clinical use, for example, metochalcone was once marketed as a cholagogue, while sofalcone was previously used as an antiulcer and mucosal protective drug.
Chalcones exhibit a broad range of biological activities, probably due to their small structure and michael acceptor character, which makes them resistant to and allow them to bind easily or reactively to different biomolecules. The biological activity of chalcone includes anticancer activity, cancer preventing effect, antiinflammatory activity, antibacterial activity, antituberculosis activity, antidiabetic activity, antioxidant activity, antibacterial activity, antiviral activity, antimalarial activity, neuroprotection, etc.
Many chalcone derivatives have also been prepared due to their ease of synthesis. These natural and synthetic compounds have shown many interesting biological activities with clinical potential against various diseases. Numerous studies have shown that they have a strong positive effect in reducing inflammation, modulating immune responses, and supporting and restoring normal function to cells.
Disclosure of Invention
The invention provides a compound with a structure shown in a formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof,
wherein R is 1 Is C 1-20 Alkyl, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl, 5-10 membered heterocyclyl, 5-10 membered heteroaryl, C 1-20 Alkyloxy, C 6-10 Arylcyclooxy, C 3-10 Cycloalkyloxy, 5-to 10-membered heterocyclyloxy or 5-to 10-membered heteroaryloxy, wherein the alkyl, aryl, cycloalkyl, heterocyclyl, heteroaryl, alkyloxy, aryl-aryloxy, cycloalkyloxy, heterocyclyloxy, heteroaryloxy is optionally substituted with one or more groups selected from halogen, C 1-10 Alkyloxy, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl group,5-10 membered heterocyclyl, 5-10 membered heteroaryl, C 6-10 Arylcyclooxy, C 3-10 A cycloalkyloxy, 5-10 membered heterocyclyloxy or 5-10 membered heteroaryloxy group;
R 2 is hydrogen, C 1-20 Alkyl, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl, 5-10 membered heterocyclyl, or 5-10 membered heteroaryl;
R 3 is hydrogen, halogen or C 1-6 Alkyl, or C 1-6 An alkyl oxy group;
each R 4 Identical or different, independently selected from hydrogen or C 1-12 An alkyl group.
In some embodiments of the invention, C 6-10 The aromatic ring group is preferably phenyl.
In some embodiments of the invention, C 3-10 Cycloalkyl is preferably C 3-7 Cycloalkyl, for example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl.
In some embodiments of the invention, the 5-10 membered heterocyclyl is preferably a 5-7 membered heterocyclyl, such as tetrahydrofuran, thiolane, pyrrolidine, dioxolane, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, imidazolidine, pyrazolidine, tetrahydropyran, piperidine, 1, 4-dioxane, piperazine, azepane, cyclohexane oxide, thietane, 1, 4-oxazaidine, 1, 4-thiazaidine.
In some embodiments of the invention, the 5-10 membered heteroaryl is preferably a 5-6 membered heteroaryl, such as furan, thiophene, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, imidazole, 1,2, 3-triazole, 1,2, 4-triazole, oxadiazole, thiadiazole, pyridine, pyridone, pyrimidine, pyridazine, pyrazine, triazine.
In some embodiments of the invention, the halogen is preferably F, cl or Br.
In some embodiments of the invention, in formula I, R 1 Is C 1-12 Alkyl, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, C 1-12 Alkyloxy, phenyloxy, C 3-7 Cycloalkyloxy, 5-to 7-membered heterocyclyloxy,5-6 membered heteroaryloxy, wherein the alkyl, phenyl, cycloalkyl, heterocyclyl, heteroaryl, alkyloxy, phenyloxy, cycloalkyloxy, heterocyclyloxy, heteroaryloxy is optionally substituted with one or more groups selected from F, cl, br, or the alkyloxy is optionally substituted with one or more groups selected from C 1-6 Alkyloxy, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyloxy, C 3-7 Cycloalkyl oxy, 5-7 membered heterocyclyloxy or 5-6 membered heteroaryloxy. Preferably, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, cyclopentyl, tetrahydrofuranyl, thiolanyl, pyrrolidinyl, tetrahydropyranyl, thietanyl, piperidinyl, C 1-8 An alkyloxy, phenyloxy, cyclohexyloxy, cyclopentyloxy, tetrahydrofuranyloxy, thiolanyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thiolanyloxy, piperidinyloxy, wherein the alkyloxy is optionally substituted with one or more groups selected from F, cl, br, C 1-6 Alkyloxy, phenyl, tetrahydrofuranyl, cyclohexyl, cyclopentyl, thiolanyl, pyrrolidinyl, tetrahydropyranyl, thiolanyl, piperidinyl, phenyloxy, tetrahydrofuranyloxy, cyclohexyloxy, cyclopentyloxy, thiolanyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thiolanyloxy, piperidinyloxy. More preferably, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, tetrahydrofuranyl, C 1-8 An alkyloxy, phenyloxy, cyclohexyloxy, tetrahydrofuranyloxy group, wherein the alkyloxy group is optionally substituted with one or more groups selected from F, cl, br, C 1-6 Alkyloxy, phenyl, tetrahydrofuranyl, cyclohexyl, phenyloxy, tetrahydrofuranyloxy, cyclohexyloxy.
In some embodiments of the invention, in formula I, R 2 Is hydrogen or C 1-12 An alkyl group; preferably R 2 Is hydrogen or C 1-4 An alkyl group; more preferably, R 2 Is methyl.
In some embodiments of the invention, in formula I, R 3 Is hydrogen or C 1-4 An alkyl group; preferably R 3 Is hydrogen.
In some embodiments of the invention, in formula I, R 4 Is hydrogen or C 1-4 An alkyl group; more preferably, R 4 Is methyl.
In some embodiments of the invention, in formula I, R 1 Is C 1-12 Alkyl, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, C 1-12 Alkyloxy, phenyloxy, C 3-7 Cycloalkyloxy, 5-7 membered heterocyclyloxy, 5-6 membered heteroaryloxy, wherein the alkyl, phenyl, cycloalkyl, heterocyclyl, heteroaryl, alkyloxy, phenyloxy, cycloalkyloxy, heterocyclyloxy, heteroaryloxy is optionally substituted with one or more groups selected from F, cl, br, or the alkyloxy is optionally substituted with one or more groups selected from C 1-6 Alkyloxy, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyloxy, C 3-7 A cycloalkyloxy, 5-7 membered heterocyclyloxy or 5-6 membered heteroaryloxy group; r is R 2 Is hydrogen or C 1-12 An alkyl group; r is R 3 Is hydrogen or C 1-4 An alkyl group; r is R 4 Is hydrogen or C 1-4 An alkyl group; preferably, R 2 Is hydrogen or C 1-4 An alkyl group; more preferably, R 2 Is methyl, R 3 Is hydrogen, R 4 Is methyl.
In some embodiments of the invention, in formula I, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, cyclopentyl, tetrahydrofuranyl, thiolanyl, pyrrolidinyl, tetrahydropyranyl, thietanyl, piperidinyl, C 1-8 An alkyloxy, phenyloxy, cyclohexyloxy, cyclopentyloxy, tetrahydrofuranyloxy, thiolanyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thiolanyloxy, piperidinyloxy, wherein the alkyloxy is optionally substituted with one or more groups selected from F, cl, br, C 1-6 Alkyloxy, phenyl, tetrahydrofuranyl, cyclohexyl, cyclopentyl, thiolanyl, pyrrolidinyl, tetrahydropyranyl, thiolanyl, piperidinyl, phenyloxy, tetrakisA radical substitution of hydrofuranyloxy, cyclohexyloxy, cyclopentyloxy, thiolanyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thietanyloxy, piperidinyloxy; r is R 2 Is hydrogen or C 1-12 An alkyl group; r is R 3 Is hydrogen or C 1-4 An alkyl group; r is R 4 Is hydrogen or C 1-4 An alkyl group; preferably, R 2 Is hydrogen or C 1-4 An alkyl group; more preferably, R 2 Is methyl, R 3 Is hydrogen, R 4 Is methyl.
In some embodiments of the invention, in formula I, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, tetrahydrofuranyl, C 1-8 An alkyloxy, phenyloxy, cyclohexyloxy, tetrahydrofuranyloxy group, wherein the alkyloxy group is optionally substituted with one or more groups selected from F, cl, br, C 1-6 Alkyl oxy, phenyl, tetrahydrofuranyl, cyclohexyl, phenyl oxy, tetrahydrofuranyloxy, cyclohexyloxy; r is R 2 Is methyl; r is R 3 Is hydrogen; r is R 4 Is methyl.
In some embodiments of the invention, the compound of formula I is selected from the following compounds:
the invention also discloses a method for preparing the compound shown in the formula I, which comprises the following steps: subjecting a compound of formula A and a compound of formula B to olefin metathesis reaction to obtain a compound of formula I, wherein R in formulas A and B 1 、R 2 、R 3 、R 4 Rx and Ry are independently selected from hydrogen and C as defined above 1-4 An alkyl group.
The compound of the formula I, the enantiomer, the pharmaceutically acceptable salt or the solvate thereof have anti-inflammatory and anti-tumor activities and have VEGF inhibiting activity.
The invention also provides the use of a compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, according to the invention, in the manufacture of a medicament for inhibiting inflammation.
The invention also provides the use of the compounds of formula I, their enantiomers, pharmaceutically acceptable salts or solvates thereof in the manufacture of an antitumor medicament.
The invention also provides the use of a compound of formula I of the invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, in the preparation of a VEGF inhibitor.
The invention also provides a pharmaceutical composition comprising a compound of formula I of the invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, optionally together with one or more pharmaceutically acceptable carriers. The pharmaceutically acceptable carrier is a variety of excipients commonly used or known in the pharmaceutical arts, including but not limited to: diluents, binders, antioxidants, pH adjusters, preservatives, lubricants, disintegrants, and the like.
The pharmaceutical composition contains the compound of formula I in an amount of 0.1 to 1000mg, preferably 1 to 500mg, more preferably 5 to 100mg, based on the compound of formula I.
The compound of the formula I (calculated as the compound of the formula I) in the pharmaceutical composition accounts for 0.01-95% of the mass of the pharmaceutical composition, and can be, for example, 0.1-10%, 0.3-5%, or 10-90% and the like according to different dosage forms.
The dosage form of the pharmaceutical composition may be in the form of an oral dosage form, such as a tablet, capsule, pill, powder, granule, suspension, syrup, etc.; it may also be in the form of an injectable administration, such as an injectable solution, powder injection, etc., by intravenous, intraperitoneal, subcutaneous or intramuscular route. All dosage forms used are well known to those of ordinary skill in the pharmaceutical arts.
Routes of administration of the pharmaceutical composition include, but are not limited to: oral administration; is taken orally; sublingual, sublingual; transdermal; lung; rectal; parenteral, e.g., by injection, including subcutaneous, intradermal, intramuscular, intravenous; by implantation into a reservoir or reservoir.
The dosage of the compound of formula I administered (based on the compound of formula I) will depend on the age, health and weight of the recipient, the type of combination drug, the frequency of treatment, the route of administration, and the like. The drug may be administered in a single daily dose, once daily, once every two days, once every three days, once every four days, or the total daily dose may be administered in divided doses of two, three or four times per day. The compounds of formula I are administered in an amount of 0.01-100 mg/kg/day (based on the compounds of formula I), for example 0.5 mg/kg/day, 1 mg/kg/day, 2 mg/kg/day, 5 mg/kg/day, etc.
The present invention also provides a method of inhibiting inflammation by administering to a patient in need thereof a compound of formula I of the present invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a compound of formula I of the present invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof.
The inflammation includes, but is not limited to, an autoimmune disease, disorder or condition, an inflammatory disease, disorder or condition; for example: idiopathic pulmonary fibrosis, inflammatory bowel disease (selected from crohn's disease and ulcerative colitis), rheumatoid arthritis, osteoarthritis, still's disease, sjogren's syndrome, systemic lupus erythematosus, multiple sclerosis, psoriasis, systemic sclerosis, acute respiratory distress syndrome, allergic rhinitis, asthma, ocular inflammatory diseases (e.g., allergic conjunctivitis, dry eye (dry eye) and uveitis), atopic dermatitis, interstitial cystitis, chronic prostatitis/chronic pelvic pain syndrome (chronic pelvic pain syndrome) (CP/CPPS), epidermocontact hypersensitivity (dermal contact hypersensitivy), eosinophilic gastrointestinal disorders (eosiniphilic gastrointestinal disorder), fibromyalgia, liver fibrosis, irritable bowel syndrome, ischemia reperfusion disease, renal fibrosis, pancreatitis, post-operative inflammation, seronegative spinal arthropathy (e.g., ankylosing spondylitis, psoriatic arthritis and reiter's syndrome), and inflammatory diseases (e.g., wegener's granulomatosis, sarcoidosis, leukosis, vascular-stoneley vasculitis, 35, vascular (35) and vascular inflammation (35) and the like).
The present invention also provides a method of treating a tumor by administering to a patient in need thereof a compound of formula I of the present invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a compound of formula I of the present invention, an enantiomer, a pharmaceutically acceptable salt or solvate thereof.
Such tumors include, but are not limited to, leukemias (e.g., acute leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, polycythemia vera), lymphomas (hodgkin's disease, non-hodgkin's disease), primary macroglobulinemia, heavy chain diseases, solid tumors such as sarcomas and cancers (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovial vioma, mesothelioma), ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer (e.g., triple negative breast cancer), ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary adenocarcinoma, bronchogenic carcinoma, medullary carcinoma, renal cell carcinoma, liver cancer, nile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, wilms' cell tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small cell lung cancer, bladder cancer, epithelial cancer, glioma, astrocytoma, medulloblastoma, craniopharyngeoma, ependymoma, pineal tumor, angioblastoma, auditory neuroma, oligodendroglioma, neuroblastoma, meningioma, melanoma, neuroblastoma, retinoblastoma), esophageal carcinoma, gall bladder cancer, renal carcinoma, multiple myeloma; preferably, the tumors include, but are not limited to: pancreatic cancer, liver cancer, lung cancer, stomach cancer, esophageal cancer, head and neck squamous cell carcinoma, prostate cancer, colon cancer, breast cancer (e.g., triple negative breast cancer), lymphoma, gall bladder cancer, kidney cancer, leukemia, multiple myeloma, ovarian cancer, cervical cancer, and glioma, and any combination thereof.
In the present disclosure, "and/or" will be seen as a specific disclosure of each of two specified features or components with or without the other. Thus, the term "and/or" as used in phrases such as "a and/or B" is intended to include "a and B", "a or B", "a" (alone) and "B" (alone). Likewise, the term "and/or" as used in phrases such as "A, B and/or C" is intended to encompass each of the following aspects: A. b and C; A. b or C; a or C; a or B; b or C; a and C; a and B; b and C; a (alone); b (alone); and C (alone).
"comprising" and "including" have the same meaning and are intended to be open and allow for the inclusion of additional elements or steps but not required. When the terms "comprising" or "including" are used herein, the terms "consisting of" and/or "consisting essentially of … …" are therefore also included and disclosed.
Alkyl: linear or branched saturated aliphatic groups. In the present invention, an alkyl group having 1 to 20 carbon atoms, preferably 1 to 12 carbon atoms, more preferably 1 to 8 carbon atoms, most preferably 1 to 4 carbon atoms, such as methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, 6-methylhept-2-yl, 5-ethyl-6-methylhept-2-yl and the like is preferable.
An alkyl oxy group: -O-alkyl, wherein alkyl is as defined above.
Cycloalkyl: saturated or partially unsaturated, monocyclic or polycyclic, cyclic alkyl groups. In the present invention, cycloalkyl groups having 3 to 10 carbon atoms, preferably 3 to 7 carbon atoms, are preferred. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbornyl (norbornyl), decalyl, and the like.
Cycloalkyloxy: -O-cycloalkyl, wherein cycloalkyl is as defined above.
A heterocyclic group: a stable 3 to 18 membered non-aromatic ring radical consisting of 2 to 12 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur, which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include spiro, fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl may optionally be oxidized; the nitrogen atom may optionally be quaternized; and the heterocyclyl groups may be partially or fully saturated. In the present invention, a 5-to 10-membered heterocyclic group is preferable, and a 5-to 7-membered heterocyclic group is more preferable. Examples of such heterocyclic groups include, but are not limited to, dioxolanyl, dioxanyl, thienyl [1,3] dithianyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, 1,2, 4-thiadiazol-5 (4H) -ylidene, tetrahydrofuranyl, trioxane, trithianyl, triazinidinyl (triazinyl), tetrahydropyranyl, thiomorpholinyl (thiomorpholinyl), 1-oxo-thiomorpholinyl, 1-dioxo-thiomorpholinyl, and 1, 6-dioxaspiro [4.5] decyl.
Heterocyclyloxy: -O-heterocyclyl, wherein heterocyclyl is as defined above.
Aromatic ring group: the hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. In the present invention, an aromatic ring group having 6 to 10 carbon atoms is preferable. Aromatic ring groups include, but are not limited to, aromatic ring groups derived from acetate, acenaphthene, acetenatorene, anthracene, azulene, benzene, fluoranthene, fluorene, asymmetric indacene (as-indacene), symmetric indacene (s-indacene), indane, indene, naphthalene, phenalene, phenanthrene, obsidiene (pleiadene), pyrene, and benzo [9,10] phenanthrene.
An aryloxygroup: -O-aryl, wherein aryl is as defined above.
Heteroaryl group: a 5 to 14 membered ring system radical comprising a hydrogen atom, 1 to 13 carbon atoms, 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur, and at least one aromatic ring, which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl group may optionally be oxidized; the nitrogen atom may optionally be quaternized. In the present invention, a heteroaryl group of 5 to 10 membered, more preferably 5 to 6 membered is preferable. Examples of heteroaryl groups include, but are not limited to, aza, acridine, benzimidazolyl, benzo [ d ] imidazolyl, benzimidazolopyrimidinyl, benzo [4,5] imidazo [1,2-a ] pyrimidinyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzo [ d ] isoxazolyl, benzothiadiazolyl, benzo [ b ] [1,4] dioxacycloheptadienyl, 1, 4-benzodioxanyl, benzonaphtofuranyl, benzoxazolyl, benzodioxolyl, benzodioxanyl, benzopyranyl, benzopyronyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothienyl), benzotriazolyl, benzo [4,6] imidazo [1,2-a ] pyridinyl benzoxazolinonyl, benzimidazolylsulfinyl (benzimidazolethionyl), carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothienyl, furanyl, furanonyl, isothiazolyl, imidazo [1,2-a ] pyridinyl, imidazo [1,2-a ] pyrimidinyl, imidazo [1,2-a ] pyrazinyl, imidazo [1,5-a ] pyrazinyl, imidazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 1, 5-naphthyridinyl, oxadiazolyl, 2-oxoazenyl, oxazolyl, oxiranyl, 1-oxopyridinyl, 1-oxopyrimidinyl, 1-oxopyrazinyl, 1-oxopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, 2, 3-naphthyridinyl, pteridinyl, pteridinonyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyridonyl, pyrazinyl, pyrimidinyl, pyrimidinonyl, pyridazinyl, pyrido [2,3-d ] pyrimidinonyl, pyrazolo [1,5-a ] pyrimidinyl, quinazolinyl, quinazolinonyl, quinoxalinyl, quinoxalinonyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, thieno [3,2-d ] pyrimidin-4-onyl, thieno [2,3-d ] pyrimidin-4-onyl, triazolyl, tetrazolyl, triazinyl and thienyl (i.e., thienyl).
Heteroaryloxy: -O-heteroaryl, wherein heteroaryl is as defined above.
The compounds of the invention may contain asymmetric or chiral centers and thus exist in different enantiomeric forms. All enantiomeric forms of these compounds, as well as mixtures thereof (including racemic mixtures), are contemplated as forming part of the present invention. Individual enantiomers of a compound may be prepared synthetically from commercially available starting materials that contain asymmetric or stereocenters or by preparing racemic mixtures followed by resolution procedures well known to those of ordinary skill in the art. These splitting methods are exemplified by: (1) Connecting the enantiomer mixture with a chiral auxiliary, separating the obtained diastereomer mixture by recrystallization or chromatography, and separating the optically pure product from the auxiliary; (2) forming a salt using an optically active resolving agent; or (3) directly separating the mixture of optical enantiomers on a chiral chromatographic column.
Drawings
FIG. 1 shows a statistical plot of the detection of IL-6 content in culture medium of RAW264.7 cells stimulated with LPS after treatment of RAW264.7 cells with the compound WR031-WR046, where P < 0.01 compared to LPS treated group, P < 0.001 compared to LPS treated group, P < 0.0001 compared to LPS treated group
FIG. 2 shows a statistical plot of the detection of TNF- α content in culture medium of RAW264.7 cells stimulated with LPS after treatment of RAW264.7 cells with compound WR031-WR046, where P < 0.01 compared to LPS treated group, P < 0.001 compared to LPS treated group, P < 0.0001 compared to LPS treated group
FIG. 3 IC50 (48 h) of compound WR034 for four TNBCs, data are plotted as a percentage relative to the DMSO control group
FIG. 4 results of effect of compounds WR034 and 5-Fu on apoptosis levels of Cal51 and MDA-MB-231 cells
FIG. 5 representative image and quantitative data analysis of colony formation of compound WR034 in Cal51 and MDA-MB-231 cells
FIG. 6 results of effect of compounds WR034 and 5-Fu on BT549 and MDA-MB-231 cell migration levels
FIG. 7 results of detection of VEGF-induced inhibition of HUVEC cell proliferation by Compounds WR033, WR034 and WR045 at gradient concentrations (2. Mu.M, 4. Mu.M, 6. Mu.M, 8. Mu.M and 10. Mu.M)
FIG. 8 results of detection of inhibition of VEGF-induced HUVEC cell proliferation by Compounds WR033, WR034 and WR045 at gradient concentrations (2. Mu.M, 4. Mu.M, 6. Mu.M, 8. Mu.M and 10. Mu.M)
FIG. 9 results of the effect of compounds WR033, WR034 and WR045 on VEGF-induced HUVEC cell migration levels
FIG. 10 results of experiments on the effect of Compounds WR033, WR034 and WR045 on in vitro angiogenesis of HUVEC cells
FIG. 11 results of experiments in which compounds WR034, WR037 and WR043 inhibited proliferation of colorectal cancer cells HCT116 and HCT8
Detailed Description
The invention is further described below with reference to examples. It should be noted that the examples should not be construed as limiting the scope of the present invention, and those skilled in the art will understand that any modifications and variations based on the present invention are within the scope of the present invention.
Conventional reagents used in the following examples are all commercially available. The biological experiments are all routine in the art, and can be performed according to the instruction of the corresponding experimental manual or the instruction of the kit.
CH 3 I: methyl iodide; k (K) 2 CO 3 : potassium carbonate; THF: tetrahydrofuran; BBr (BBr) 3 : boron tribromide; cuI: cuprous iodide; DMF: n, N-dimethylformamide; DEAD: diethyl azodicarboxylate; PPh (PPh) 3 : triphenylphosphine; eu (fod) 3 : europium tris (6,6,7,7,8,8,8-heptafluoro-2, 2-dimethyl-3, 5-octanedioic acid); ac (Ac) 2 O: acetic anhydride.
EXAMPLE 1 Synthesis of WR031 Compound
The specific experimental steps are as follows:
compound 1 (2 g,13.144 mmol), potassium carbonate (3.63 g,26.288 mmol), 3-chloro-3-methyl-1-butyne (2.29 g,22.33 mmol), cuprous iodide (0.13 g,0.66 mmol) and potassium iodide (3.27 g,19.72 mmol) were placed in sequence in a 250mL round bottom flask, dissolved in anhydrous DMF, and the reaction was stirred at ambient temperature. The reaction was monitored by TLC, after completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction solution was washed and extracted with saturated sodium chloride solution (100 mL. Times.3), the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated, and the crude product was purified by flash silica gel chromatography to give Compound 2 (2.324 g, 81%) as a pale yellow solid.
1 H NMR(400MHz,Chloroform-d)δ12.60(s,1H),7.63(d,J=8.9Hz,1H),6.88(d,J=2.5Hz,1H),6.66(dd,J=8.9,2.5Hz,1H),2.66(s,1H),2.56(s,3H),1.72(s,6H).
13 C NMR(100MHz,Chloroform-d)δ202.81,164.36,162.63,131.82,114.50,111.05,106.48,84.71,75.10,72.36,29.54,26.30.
Compound 2 (500 mg,2.28 mmol) was dissolved in Diethyllaniline (15 mL), placed in a 50mL round bottom flask, the reaction system was placed at 250℃and checked by TLC, after completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction solution was washed and extracted with sodium hydrogencarbonate solution (100 mL. Times.3) and saturated sodium chloride solution (100 mL. Times.3) in this order, the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound 3 (473 mg, 95%) as a white solid.
1 H NMR(400MHz,Chloroform-d)δ12.97(s,1H),7.50(d,J=8.8Hz,1H),6.70(dd,J=10.1,0.8Hz,1H),6.32(dd,J=8.8,0.8Hz,1H),5.57(d,J=10.1Hz,1H),2.53(s,3H),1.44(s,6H).
13 C NMR(100MHz,Chloroform-d)δ202.77,159.62,131.66,128.22,115.79,113.85,109.22,108.31,77.72,28.31,26.18.
Compound 3 (0.88 g,4.03 mmol) and sodium hydride (0.55 g,22.91 mmol) were dissolved in anhydrous DMF under argon, and then placed in a 50mL round bottom flask and stirred, allyl bromide (1.19 mL,10.31 mmol) was added under ice-bath, and after stirring for 30 minutes, the reaction was continued at ambient temperature. After completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction mixture was washed with a saturated sodium chloride solution (100 mL. Times.3) and extracted, and the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound 5 (784 mg, 88%) as a white solid.
1 H NMR(400MHz,Chloroform-d)δ7.53–7.41(m,1H),6.52(ddd,J=13.2,9.7,3.0Hz,2H),5.98(dddd,J=15.9,10.8,5.4,2.8Hz,1H),5.72–5.50(m,1H),5.35(ddd,J=17.1,3.0,1.5Hz,1H),5.20(ddd,J=10.5,3.0,1.5Hz,1H),4.29(dq,J=4.4,1.5Hz,2H),2.67–2.31(m,3H),1.59–0.87(m,6H).
13 C NMR(100MHz,Chloroform-d)δ198.32,157.86,155.24,132.98,131.03,130.56,125.59,118.12,116.75,115.08,112.75,76.87,76.61,30.35,28.00.
Compound 5 (0.88 g,3.11 mmol) and Eu (fod) 3 (0.16 mg,0.16 mmol) was dissolved in chloroform and placed in a 50mL round bottom flask with stirring at 60℃under reflux. Detection of the reaction by TLCAfter the reaction was completed, 200mL of ethyl acetate was added to the system, the reaction solution was washed and extracted with a saturated sodium chloride solution (100 mL. Times.3), and the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound 6 (0.695 g, 78%) as a white solid.
1 H NMR(400MHz,Chloroform-d)δ12.87(d,J=3.2Hz,1H),7.33(d,J=3.1Hz,1H),6.71(d,J=9.6Hz,1H),5.94(ddd,J=16.9,6.9,3.6Hz,1H),5.58(d,J=9.7Hz,1H),5.27–4.82(m,2H),3.27(d,J=6.3Hz,2H),2.54(d,J=3.2Hz,3H),1.44(d,J=3.0Hz,6H).
13 C NMR(100MHz,Chloroform-d)δ202.76,158.34,157.55,136.67,131.25,127.99,119.13,116.09,115.67,113.37,108.99,77.67,33.46,28.31,26.22.
Compound 6 (0.6 g,2.09 mmol) was dissolved in anhydrous N, N-dimethylformamide, and then placed in a 50mL round bottom flask and stirred, methyl iodide (0.26 mL,4.19 mmol) and sodium hydride (0.25 g,10.41 mmol) were slowly added dropwise under ice-bath, and after stirring for 30 minutes, the reaction system was allowed to continue at ambient temperature. After completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction mixture was washed with a saturated sodium chloride solution (100 mL. Times.3) and extracted, and the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound 7 (559 mg, 89%) as a white solid.
Compound 7 (330 mg,1.21 mmol) and benzaldehyde (535.8 mg,5.049 mmol) were dissolved in absolute ethanol and then placed in a 50mL round bottom flask and stirred, 4M sodium hydroxide solution (0.841 mL,3.367 mmol) was slowly added dropwise under ice-bath, after stirring for 30 min, the reaction was continued at 50 ℃. After completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction mixture was washed with a saturated sodium chloride solution (100 mL. Times.3) and extracted, and the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound 8 (698 mg, 86%) as a pale yellow solid.
1 H NMR(400MHz,Chloroform-d)δ7.73(d,J=15.8Hz,1H),7.67–7.50(m,3H),7.46–7.36(m,4H),6.65(d,J=10.0Hz,1H),6.03–5.87(m,1H),5.70(d,J=10.0Hz,1H),5.15–4.92(m,2H),3.74(s,3H),3.33(dd,J=6.6,1.6Hz,2H),1.46(s,6H).
13 C NMR(100MHz,Chloroform-d)δ190.89,155.18,155.03,142.91,136.36,135.29,131.50,130.42,130.18,128.91,128.43,126.33,124.96,124.12,116.77,115.82,114.49,77.26,76.89,63.53,33.67,28.14.
Intermediate 8 (50 mg,0.138 mmol) was dissolved in anhydrous dichloromethane and placed in a 50mL round bottom flask and stirred, 2-phenoxyethyl acrylate (2-phenoxyethyl acrylate) (27. Mu.L, 0.278 mmol) and Grubbs' second generation catalyst (23 mg,0.027 mmol) were slowly dropped under ice bath, and after stirring for 30 min, the reaction was allowed to continue at ambient temperature. After completion of the reaction, 200mL of ethyl acetate was added to the system, the reaction mixture was washed with a saturated sodium chloride solution (100 mL. Times.3) and extracted, and the organic phase was collected, treated with anhydrous sodium sulfate as a drying agent, filtered and concentrated. The crude product was purified by flash chromatography on silica gel to give compound WR031 (40.81 mg, 71%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ7.79–7.46(m,4H),7.43–7.34(m,4H),7.30–7.00(m,3H),6.90(dd,J=23.6,7.8Hz,3H),6.61(d,J=10.0Hz,1H),5.81(dt,J=15.5,1.6Hz,1H),5.66(d,J=10.0Hz,1H),4.44(t,J=4.8Hz,2H),4.15(t,J=4.8Hz,2H),3.71(s,3H),3.55–3.31(m,2H),1.40(s,6H).
13 C NMR(100MHz,CDCl 3 )δ262.40,190.65,166.47,158.51,155.48,155.24,147.60,143.15,135.21,131.94,130.57,130.26,129.52,128.93,128.47,126.14,125.24,121.56,121.14,116.59,114.74,114.63,94.32,77.25,65.90,63.57,62.68,32.36,28.17.。
EXAMPLE 2 preparation of other Compounds
Using a method similar to example 1, the following compounds were prepared:
1 H NMR(400MHz,CDCl 3 )δ7.74(dd,J=15.5,2.9Hz,1H),7.66–7.53(m,3H),7.40(dt,J=8.4,2.8Hz,4H),7.05(dtd,J=16.2,6.8,3.0Hz,1H),6.65(dd,J=10.0,2.9Hz,1H),5.79(dt,J=15.5,1.8Hz,1H),5.70(dd,J=10.0,3.0Hz,1H),4.78(s,1H),3.74(d,J=3.0Hz,3H),3.51–3.39(m,2H),1.95–1.63(m,3H),1.61–1.14(m,13H).
13 C NMR(100MHz,CDCl 3 )δ190.72,166.12,155.44,146.22,143.16,135.19,131.95,130.57,130.28,128.94,128.48,126.13,125.19,122.66,121.83,116.60,114.73,77.22,72.44,63.59,32.36,31.69,28.17,25.43,23.81。
1 H NMR(400MHz,CDCl 3 )δ7.74(dd,J=15.9,2.5Hz,1H),7.67–7.51(m,3H),7.40(dt,J=5.3,2.6Hz,4H),7.17–6.97(m,1H),6.64(dd,J=10.2,2.5Hz,1H),5.90–5.65(m,2H),3.73(dd,J=12.3,2.5Hz,6H),3.46(d,J=6.6Hz,2H),1.45(d,J=2.5Hz,6H).
13 C NMR(100MHz,CDCl 3 )δ190.70,167.09,155.46,155.25,146.96,143.18,135.19,131.90,130.58,130.29,128.94,128.49,126.11,125.21,121.70,116.60,114.74,77.24,63.60,51.50,32.29,28.19。
1 H NMR(400MHz,CDCl 3 )δ7.79–7.51(m,4H),7.40(q,J=5.3,4.3Hz,4H),6.91(dd,J=15.9,6.5Hz,1H),6.63(dd,J=10.1,6.0Hz,1H),6.08(d,J=16.2Hz,1H),5.74–5.65(m,1H),3.73(d,J=5.9Hz,3H),3.46(t,J=6.1Hz,2H),2.55(q,J=7.1Hz,2H),1.44(d,J=5.9Hz,6H),1.06(q,J=7.0Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ201.23,190.68,155.49,155.27,144.39,143.20,135.16,131.88,130.80,130.55,130.31,128.95,128.48,126.07,125.22,121.75,116.60,114.74,77.24,63.60,33.14,32.61,28.22,8.12。
1 H NMR(400MHz,CDCl 3 )δ7.81–7.49(m,4H),7.48–7.33(m,4H),7.05–6.88(m,1H),6.69–6.61(m,1H),5.79–5.66(m,2H),3.82–3.66(m,3H),3.48–3.37(m,2H),1.46(d,J=2.9Hz,15H).
13 C NMR(100MHz,CDCl 3 )δ190.74,166.03,155.40,155.25,145.30,143.15,135.20,131.93,130.57,130.28,128.94,128.48,126.15,125.17,123.86,122.00,116.61,114.71,80.13,77.20,63.58,32.25,28.16.。
1 H NMR(400MHz,CDCl 3 )δ7.74(dd,J=15.9,1.5Hz,1H),7.66–7.61(m,2H),7.56(dd,J=15.8,1.6Hz,1H),7.45–7.28(m,9H),7.13(dtd,J=15.2,6.6,1.6Hz,1H),6.64(dd,J=10.0,1.6Hz,1H),5.85(dd,J=15.6,1.7Hz,1H),5.70(dd,J=10.0,1.6Hz,1H),5.16(d,J=1.6Hz,2H),3.74(d,J=1.6Hz,3H),3.47(dt,J=6.6,1.7Hz,2H),1.44(d,J=1.6Hz,6H).
13 C NMR(100MHz,CDCl 3 )δ190.67,166.41,155.48,155.25,147.38,143.16,136.10,135.19,131.95,130.56,130.28,128.93,128.56,128.48,128.22,126.12,125.22,121.74,121.56,116.58,114.74,77.24,66.10,63.58,32.40,28.18.。
1 H NMR(400MHz,CDCl 3 )δ7.73(d,J=15.8Hz,1H),7.67–7.51(m,3H),7.47–7.33(m,4H),7.11(d,J=15.6Hz,1H),6.64(d,J=10.1Hz,1H),5.83(dt,J=15.7,1.6Hz,1H),5.69(d,J=10.0Hz,1H),4.26(t,J=4.6Hz,2H),3.74(s,3H),3.59(t,J=4.7Hz,2H),3.51–3.41(m,2H),3.37(s,3H),1.44(s,6H).
13 C NMR(100MHz,CDCl 3 )δ190.66,166.57,155.46,155.23,147.35,143.14,135.20,131.96,130.56,130.26,128.92,128.47,126.14,125.22,121.66,121.57,116.59,114.73,77.23,70.54,63.56,63.32,59.02,32.27,28.18.。
1 H NMR(400MHz,CDCl 3 )δ7.74(d,J=15.6Hz,1H),7.66–7.51(m,3H),7.40(dd,J=8.0,3.7Hz,4H),7.13–7.02(m,1H),6.64(d,J=10.0Hz,1H),5.80(d,J=15.6Hz,1H),5.70(d,J=10.0Hz,1H),4.16(q,J=7.1Hz,2H),3.74(s,3H),3.45(d,J=6.7Hz,2H),1.45(s,6H),1.26(t,J=7.1Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ190.69,166.64,155.45,155.24,146.61,143.16,135.19,131.91,130.57,130.27,128.93,128.47,126.13,125.20,122.12,121.72,116.60,114.73,77.22,63.57,60.22,32.28,28.17,14.28.。
1 H NMR(400MHz,CDCl 3 )δ7.74(dd,J=16.0,2.9Hz,1H),7.67–7.50(m,3H),7.48–7.32(m,4H),7.11(dddt,J=16.0,7.8,6.2,2.4Hz,1H),6.73–6.57(m,1H),5.89–5.78(m,1H),5.74–5.65(m,1H),4.20(dt,J=11.2,3.4Hz,1H),4.11(td,J=6.9,3.3Hz,1H),4.08–3.99(m,1H),3.87(ddd,J=7.1,4.4,2.6Hz,1H),3.82–3.70(m,4H),3.53–3.38(m,2H),2.08–1.79(m,3H),1.60(ddd,J=10.7,7.2,2.5Hz,1H),1.53–1.38(m,6H).
13 C NMR(100MHz,CDCl 3 )δ190.66,166.57,155.46,155.25,147.34,143.14,135.19,131.98,130.57,130.27,128.93,128.48,126.11,125.20,121.66,121.56,116.59,114.73,77.23,76.57,68.47,66.34,63.59,32.27,28.19,28.01,25.67.。
1 H NMR(400MHz,CDCl 3 )δ7.76(d,J=15.8Hz,1H),7.69–7.53(m,3H),7.48(s,1H),7.44–7.31(m,5H),7.30–7.18(m,2H),7.14–7.04(m,2H),6.67(d,J=9.9Hz,1H),6.01(dd,J=15.6,1.8Hz,1H),5.73(dd,J=10.0,1.8Hz,1H),3.76(d,J=1.8Hz,3H),3.62–3.49(m,2H),1.49(d,J=1.8Hz,6H).
13 C NMR(100MHz,CDCl 3 )δ190.68,164.97,155.57,155.28,150.75,149.02,143.25,135.19,131.99,130.61,130.31,129.41,128.95,128.49,126.13,125.72,125.32,121.66,121.36,116.61,114.82,77.34,63.60,32.56,28.23.。
1 H NMR(400MHz,CDCl 3 )δ7.74(d,J=15.8Hz,1H),7.66–7.52(m,3H),7.40(dd,J=8.2,3.9Hz,4H),7.06(dt,J=15.3,6.7Hz,1H),6.64(d,J=10.0Hz,1H),5.80(d,J=15.6Hz,1H),5.70(d,J=10.0Hz,1H),4.11(t,J=6.6Hz,2H),3.74(s,3H),3.45(d,J=6.7Hz,2H),1.62(dq,J=14.1,6.5Hz,2H),1.45(s,6H),1.37(q,J=7.5Hz,2H),0.92(t,J=7.4Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ190.70,166.75,155.45,155.25,146.54,143.16,135.19,131.92,130.56,130.27,128.93,128.47,126.14,125.20,122.13,121.73,116.60,114.72,77.22,64.15,63.57,32.35,30.72,28.17,19.17,13.74.。
1 H NMR(400MHz,CDCl 3 )δ7.73(d,J=15.7Hz,1H),7.67–7.51(m,3H),7.48–7.31(m,4H),7.05(dt,J=15.8,6.6Hz,1H),6.75–6.57(m,1H),5.81(d,J=15.5Hz,1H),5.70(d,J=10.0Hz,1H),4.03(dd,J=5.9,3.5Hz,2H),3.74(d,J=2.1Hz,3H),3.45(d,J=6.7Hz,2H),1.58(dt,J=12.1,6.4Hz,1H),1.45(s,6H),1.36(qd,J=7.1,2.3Hz,2H),1.28(d,J=6.4Hz,6H),0.88(dt,J=8.0,4.2Hz,6H).
13 C NMR(100MHz,CDCl 3 )δ190.69,166.84,155.47,155.26,146.39,143.15,135.22,131.92,130.52,130.25,128.92,128.46,126.17,125.21,122.21,121.74,116.61,114.71,77.21,66.68,63.54,38.81,32.44,30.43,28.94,28.16,23.79,22.97,14.05,11.00.。
1 H NMR(400MHz,CDCl 3 )δ7.75(d,J=15.8Hz,1H),7.67–7.53(m,4H),7.46–7.37(m,4H),6.65(dd,J=10.2,2.2Hz,1H),5.91(dd,J=15.5,1.9Hz,1H),5.88–5.76(m,1H),5.71(dd,J=10.2,2.0Hz,1H),3.75(d,J=2.2Hz,3H),3.60–3.49(m,2H),1.44(d,J=2.2Hz,6H).
13 C NMR(100MHz,CDCl 3 )δ190.58,162.81,155.75,155.28,152.41,143.31,135.15,132.02,130.56,130.33,128.94,128.47,126.03,125.39,120.51,118.60,116.51,114.83,77.24,66.66,66.32,65.97,63.58,33.08,28.12.。
1 H NMR(400MHz,CDCl 3 )δ7.73(d,J=15.8Hz,1H),7.67–7.50(m,3H),7.48–7.35(m,4H),7.06(dt,J=15.6,6.7Hz,1H),6.64(d,J=10.0Hz,1H),5.80(d,J=15.6Hz,1H),5.70(d,J=10.0Hz,1H),4.10(t,J=6.7Hz,2H),3.74(s,3H),3.53–3.36(m,2H),1.62(q,J=6.9Hz,2H),1.45(s,6H),1.37–1.20(m,18H),0.87(t,J=6.7Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ190.67,166.72,155.45,155.23,146.48,143.14,135.22,131.90,130.52,130.24,128.91,128.45,126.17,125.21,122.17,121.73,116.61,114.71,77.20,64.45,63.53,32.34,31.92,29.64,29.59,29.53,29.35,29.28,28.69,28.17,25.96,22.69,14.12.。
1 H NMR(400MHz,CDCl 3 )δ7.93–7.86(m,2H),7.74(d,J=15.8Hz,1H),7.65–7.49(m,4H),7.49–7.35(m,6H),7.14(dd,J=14.5,7.6Hz,1H),6.87(dt,J=15.3,1.6Hz,1H),6.66(d,J=10.0Hz,1H),5.71(d,J=10.0Hz,1H),3.75(s,3H),3.58(d,J=6.8Hz,2H),1.46(s,6H).
13 C NMR(100MHz,CDCl 3 )δ191.06,190.67,155.51,155.27,147.16,143.19,137.94,135.20,132.62,131.93,130.55,130.26,128.92,128.56,128.53,128.45,126.17,125.25,121.80,116.63,114.78,77.26,63.55,33.06,28.21.。
1 H NMR(400MHz,CDCl 3 )δ7.74(d,J=15.8Hz,1H),7.65–7.50(m,3H),7.47–7.33(m,4H),6.90(dd,J=14.6,7.9Hz,1H),6.65(d,J=10.0Hz,1H),6.13–6.01(m,1H),5.70(d,J=10.0Hz,1H),3.74(s,3H),3.52–3.41(m,2H),2.50(t,J=7.4Hz,2H),1.62(q,J=7.3Hz,2H),1.45(s,6H),0.91(t,J=7.4Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ200.72,190.66,155.48,155.24,144.45,143.18,135.20,131.85,131.11,130.50,130.26,128.92,128.45,126.14,125.24,121.73,116.61,114.72,77.22,63.53,41.93,32.59,28.20,17.67,13.82.。
example 3 anti-inflammatory Activity
1. Anti-inflammatory Activity assay
1) Experimental materials
And (3) cells: macrophage (RAW 264.7)
Cytokine kit: IL-6 detection kit and TNF-alpha detection kit
Stimulus: lipopolysaccharide (LPS)
Positive control: dexamethasone (Dex)
Solvent: dimethyl sulfoxide (DMSO)
Comparative compound: WR011WR022/>
2) Experimental procedure
Cell culture: RAW264.7 cells were resuscitated in T75 cell culture flasks and incubated at 37℃with 5% CO 2 Is cultured in a constant temperature incubator, and is subjected to passage treatment when the cell density in a cell culture flask reaches more than 99%, and is subjected to passage for at least 2 times. Cells were then plated in 24-well cell culture plates at a cell concentration of 0.5X10 per well 6 Overnight culture allowed the cells to adhere well.
And (3) drug treatment: cells of experimental group and positive control group were added with 20. Mu.M of compound and Dex, respectively, and negative control was added with DMSO as a solvent for dissolving the compound alone, at 37℃with 5% CO 2 After 1h incubation in a constant temperature incubator, the experimental group and the positive control group were incubated with 5. Mu.g/mL of LPS for 12h, and cell supernatants were collected for detection experiments of the cytokine TNF-alpha and IL-6 contents.
Cytokine detection: cytokine levels in serum were detected using ELISA kits for TNF- α, IL-6. mu.L of a capture antibody solution of TNF-. Alpha.and IL-6 was added to the 96-well plate, and incubated overnight at 4 ℃. Wash Buffer was washed 3 times, blocked with 200. Mu.L/well Assay reagent A, and washed 3 times at 37℃for 1 h. 100. Mu.L/well of TNF-alpha, IL-6 standard and sample were added, washed 3 times at 37℃for 2 hours, 100. Mu.L of diluted TNF-alpha, IL-6 antibody solution was added, washed 1 hour at 37℃for 3 times, 100. Mu.L of Avidin-HRP solution was added, washed 30 minutes at room temperature, 3 times at 100. Mu.L of TMB was added, and light was protected from the environment at room temperature for 30 minutes. 100. Mu.L of stop solution was added to terminate the reaction. The absorbance at 450nm was read with a microplate reader within 15min and cytokine levels were calculated from the standard curve.
2. Experimental results
As shown in the experimental results in FIG. 1 and FIG. 2, the tested compounds can down-regulate the content of IL-6 and TNF-alpha after LPS stimulates RAW264.7 cells, have obvious differences compared with LPS groups, and are superior to positive control dexamethasone and comparative compounds WR011 and WR022.
3. The inventors further carried out concentration gradient experiments using WR034, WR037, and WR043 as examples: by adopting the same experimental method, the experimental compound and positive control dexamethasone are respectively treated with 20 mu M, 10 mu M and 5 mu M, and the content of cytokines TNF-alpha and IL-6 is detected, so that the results show that the contents of WR034, WR037 and WR043 for inhibiting TNF-alpha and IL-6 generated under LPS stimulation have dose-response relation, the higher the concentration, the higher the inhibition intensity, and the positive control dexamethasone with the concentration is superior to that of the positive control dexamethasone with the corresponding concentration.
The experimental results show that the compound has the effect of inhibiting the generation of macrophage TNF-alpha and IL-6 under the stimulation of LPS.
Example 4 anti-tumor Activity
1. Anti-triple negative breast cancer cell (TNBC) proliferation experiment-CCK-8 detection
1) Experimental materials
And (3) cells: human breast cancer cells MDA-MB-231, MDA-MB-468, BT549 and Cal51
Cell proliferation assay kit: cell Counting Kit-8 (CCK 8) kit (Tao Shu TargetMol)
Solvent: dimethyl sulfoxide DMSO (Solaro Soy Bao)
2) Experimental procedure
Cell culture: resuscitates MDA-MB-231, MDA-MB-468, BT549, cal51 cells in T75 cell culture flasks, and 5% CO at 37deg.C 2 Is cultured in a constant temperature incubator, and is subjected to passage treatment when the cell density in a cell culture flask reaches more than 90%, and is subjected to passage for at least 2 times. The cells were then plated in 96-well cell culture plates, respectively, at a cell concentration of 1X 10 per well 4 Overnight culture allowed the cells to adhere well.
And (3) drug treatment: the cells of the experimental group were added with the compound WR034 to give a final concentration gradient of 1. Mu.M, 5. Mu.M, 10. Mu.M, 15. Mu.M, 20. Mu.M. Cells of the control group were added with DMSO, a solvent for dissolving the compound, and the blank group was added with cell-free medium at 37℃with 5% CO 2 Is incubated for 48h in a constant temperature incubator.
And (3) detection: after 48h, 10. Mu.L of CCK-8 solution was added to each well of the experimental, control and blank groups for incubation for 1-2h, and absorbance at 450nm was measured with a microplate reader for calculation of cell viability.
Cell viability = [ (As-Ab)/(Ac-Ab) ] ×100%
As: an experimental hole; ac: control wells; ab: blank hole
3) Experimental results
The experimental results are shown in fig. 3, and the experiment shows that the compound WR034 can obviously inhibit proliferation of four TNBC, and the IC50 is respectively: MDA-MB-468: 2.906. Mu.M; MDA-MB-231: 7.388. Mu.M; BT549: 7.771. Mu.M; cal51: 2.452. Mu.M.
2. Promote triple negative breast cancer cell apoptosis experiment
1) Experimental materials:
and (3) cells: human breast cancer cells Cal51, MDA-MB-231
Positive control drug: 5-fluorouracil (5-Fu, MCE)
Solvent: dimethyl sulfoxide DMSO (Solaro Soy Bao)
Reagent: annexin V-FITC/PI fluorescent double-dye apoptosis detection kit (Procell of Punuocele)
2) Experimental method
Cells were seeded in 12-well plates with MDA-MB-231 of 1.5X10 5 A/hole; cal51 is 2X 10 5 Cells were allowed to adhere well by overnight culture per well. DMSO was added to the blank, equivalent amount of compound WR034 was added to the test to give a final concentration of 5 μm and 10 μm, and 5-Fu was added to the positive control to give a final concentration of 10 μm. Placing into an incubator for culturing for 48 hours.
After 48h, cells from each well were collected into a centrifuge tube, centrifuged at 300 Xg for 5min, the supernatant was discarded, the cells were collected, washed once with PBS, and the cells were gently resuspended and countedA number. Taking 1×10 5 The resuspended cells were centrifuged at 300 Xg for 5min and the supernatant discarded. Cells were washed once with PBS, centrifuged, and the supernatant was discarded, and 100. Mu.L of diluted 1X Annexin V Binding Buffer resuspended cells were added. To the cell suspension, 2.5. Mu.L of Annexin V-FITC staining solution and 2.5. Mu.L of PI staining solution (50. Mu.g/mL) were added. After being mixed evenly by gentle vortex, the mixture is incubated for 15 to 20 minutes at room temperature in a dark place. 400 μl of diluted 1× Annexin V Binding Buffer was added and the samples were mixed. And (5) immediately starting the machine for detection. During detection by a flow cytometer, annexin V-FITC selects a FITC channel, and PI selects a PerCP/Cy5.5 channel.
3) Experimental results
The experimental results are shown in FIG. 4.
(1) In Cal51 cells, compound WR034 promotes apoptosis, and the percentage of apoptosis increases with increasing concentration of drug administered. At the same concentration, the apoptosis percentage of the cells in the group of the compound WR034 is higher than that of the cells in the group of the positive control drug 5-Fu.
In MDA-MB-231 cells, the compound WR034 can promote apoptosis, and the apoptosis percentage increases with the increase of the administration concentration, and the apoptosis percentage of cells of a low-concentration compound WR034 administration group is higher than that of cells of a positive control 5-Fu group.
3. Anti-triple negative breast cancer cell proliferation experiment-colony formation experiment
1) Experimental materials:
and (3) cells: human breast cancer cells Cal51, MDA-MB-231
Solvent: dimethyl sulfoxide DMSO (Solaro Soy Bao)
Reagent: 4% paraformaldehyde, 0.1% crystal violet staining solution (Solaro Soy Bao)
2) Experimental method
Cells were seeded in 12-well plates with MDA-MB-231 at 500/well; cal51 is 600 per well. Cells were treated with different concentrations of compound WR034 (final concentrations of 1.25 μm, 2.5 μm) and DMSO (control). The culture medium was changed every 3 days until visible colonies were formed. After the end of the experiment, cells were fixed with 4% paraformaldehyde for 30min, stained with 0.1% crystal violet for 30min, photographed and colony counted per well.
3) Experimental results
The experimental results are shown in fig. 5, the colony formation number in the administration group is reduced as compared with the control group, and the effect is enhanced as the concentration increases. Therefore, the compound WR034 inhibits the proliferation of Cal51 and MDA-MB-231 cells, and the inhibition effect is concentration-dependent.
4. Migration test for inhibiting triple negative breast cancer cells
1) Experimental materials:
and (3) cells: human breast cancer cell BT549, MDA-MB-231
Positive control drug: 5-fluorouracil (5-Fu, MCE)
Solvent: dimethyl sulfoxide DMSO (Solaro Soy Bao)
2) Experimental method
Cells were seeded in 6-well plates with MDA-MB-231 of 5.5X10 5 A/hole; BT549 is 3.5X10 5 Cells were allowed to adhere well by overnight culture per well. The wells were scored from one end to the other with 200 μl tips perpendicular to the lateral line on the surface of the well plate, washed with PBS to remove debris, DMSO was added to the blank, compound WR034 was added to the test to a final concentration of 10 μm, and 5-Fu was added to the positive control to a final concentration of 10 μm. The trace width was observed under a microscope at 0h and 12h, respectively, and photographed, and then imported into image J for result analysis.
3) Experimental results
The experimental results are shown in FIG. 6. The compound WR034 dosed group had reduced mobility compared to the control group, and the mobility was less than the 5-Fu dosed group. Thus, it can be seen that compound WR034 inhibits migration of both BT549 and MDA-MB-231 cell lines and this effect is superior to that of the positive control 5-Fu.
Example 5 anti-tumor angiogenesis Activity of Compounds
1. Compound concentration gradient test results
1) Experimental materials
And (3) cells: human Umbilical Vein Endothelial Cells (HUVEC)
Reagent: vascular Endothelial Growth Factor (VEGF) and cell viability detection kit (CCK-8)
Solvent: dimethyl sulfoxide (DMSO)
2) Experimental procedure
Cell culture: resuscitates HUVEC cells into T75 culture flask, and cultures in cell culture box at 37deg.C, carbon dioxide concentration of 5% and humidity of 90% until cell density reaches above 80%, and subcultures. Cells were plated in 96-well plates after passage 3, 5000 cells per well, and cultured overnight to allow adequate cell attachment.
And (3) drug treatment: after the cells had been sufficiently adherent, they were starved for 24h with cell culture medium containing 0.5% serum, and then treated for 48h with cell culture medium containing 0.5% serum with 50ng/mL VEGF and gradient concentrations (2. Mu.M, 4. Mu.M, 6. Mu.M, 8. Mu.M and 10. Mu.M) of the compound. Wherein the experimental group is a compound dissolved in DMSO, and the negative control group is two groups, namely the DMSO of the dissolved compound, one group contains 50ng/mL of VEGF, and the other group does not contain VEGF.
CCK-8 detection: after cells were treated with the drug for 48 hours, 10. Mu.L of CCK-8 was added to each well, incubated in a cell incubator for 2 hours, and then absorbance at 450nm was read with a microplate reader, and cell activity was calculated according to the formula.
3) Experimental results
After the VEGF induces the HUVEC cells, the VEGF-VEGFR2 signaling pathway of the HUVEC cells can be effectively activated, so that the proliferation of the HUVEC cells is promoted, and the tumor angiogenesis capacity is enhanced. As shown in FIG. 7, after HUVEC cells were induced by VEGF, the inhibition of proliferation by compounds WR033, WR034 and WR045 at gradient concentrations (2. Mu.M, 4. Mu.M, 6. Mu.M, 8. Mu.M and 10. Mu.M) was examined, and it was found that the inhibition was more pronounced with increasing concentrations of the compounds at each concentration. IC of Compound WR033 50 Around 6. Mu.M, each compound was treated with 6. Mu.M in the subsequent experiments. When the effect of the compound on the proliferation of HUVEC cells not induced by VEGF was examined, the results are shown in FIG. 8, and it was found that the inhibitory effect of the compound at various concentrations was not obvious, indicating the effect of the compound on the proliferation of HUVEC cellsIs achieved by inhibiting the VEGF-VEGFR2 signaling pathway.
2. Effect of Compounds on VEGF-induced HUVEC cell migration Capacity experiments
Experimental materials
And (3) cells: human Umbilical Vein Endothelial Cells (HUVEC)
Reagent: vascular Endothelial Growth Factor (VEGF)
Positive control: sorafenib (Sorafenib), sunitinib (Sunitinib)
Solvent: dimethyl sulfoxide (DMSO)
1) Experimental procedure
Cell culture: resuscitates HUVEC cells into T75 culture flask, and cultures in cell culture box at 37deg.C, carbon dioxide concentration of 5% and humidity of 90% until cell density reaches above 80%, and subcultures. Cells were plated in 6-well plates after passage 3, with a cell number of 1X 10 per well 6 And (3) culturing overnight to enable the cells to be fully attached.
And (3) drug treatment: after the cells had been sufficiently adherent, they were starved for 24h with cell culture medium containing 0.5% serum, then scored with a 200. Mu.L yellow gun, and subsequently treated for 12h with cell culture medium containing 0.5% serum and 6. Mu.M compound with 50ng/mL VEGF. Wherein the experimental group is a compound dissolved in DMSO, the positive control group is Sorafenib and Sunitinib dissolved in DMSO, the negative control group is cell culture medium containing 0.5% serum with 50ng/mL VEGF and DMSO for dissolving the compound.
Finally, observing and photographing under a microscope.
2) Experimental results
As shown in fig. 9, the compounds WR033, WR034 and WR045 all have remarkable inhibiting effect on HUVEC cell migration induced by VEGF, and the inhibiting effect of the compound WR034 is the best and is superior to that of the positive control drugs Sorafenib and Sunitinib.
3. Experiment of Effect of Compounds on HUVEC cell angiogenesis in vitro
Cell culture: resuscitate HUVEC cells in T75 flask and place at 37deg.C, and perform the oxidationCulturing in a cell culture box under the conditions of 5% of carbon concentration and 90% of humidity until the cell density reaches more than 80% for passage. Cells were plated in 12 well plates after passage 3 with a cell count of 1X 10 per well 5 And (3) culturing overnight to enable the cells to be fully attached.
And (3) drug treatment: after the cells were sufficiently adherent, the cells were starved with a cell culture medium containing 0.5% serum for 24 hours, and then treated with a cell culture medium containing 0.5% serum and 6 μm compound for 30 minutes. The cells were then digested for later use. Wherein the experimental group is a compound dissolved in DMSO, the positive control group is Sorafenib and Sunitinib dissolved in DMSO, and the negative control group is DMSO for dissolving the compound. No additional additions were required for this experiment because of the self-contained VEGF in matrigel.
In vitro angiogenesis: the matrigel and serum-free medium are mixed on ice, 50 mu L of diluted matrigel is spread on each 96-well plate hole, then the diluted matrigel is placed in a cell culture box for incubation for 45-60min to solidify, 30000 cells digested in advance are dispersed in 50 mu L of medium and spread on the solidified matrigel, then the fixed matrigel is placed in the cell culture box for incubation, and after 4 hours, observation and photographing are carried out under a microscope.
Experimental results:
as shown in fig. 10, compounds WR033, WR034 and WR045 all had significant inhibitory effect on HUVEC cell angiogenesis in vitro. And the inhibition effect of the compound WR034 is the best and is superior to that of the positive control medicines Sorafenib and Sunitinib.
Example 6 experiments to inhibit colorectal cancer cell proliferation
Using the same method as the experiment for the 1 st CCK-8 test in example 4, experiments were conducted by replacing colorectal cancer cell lines HCT116 and HCT8, and as shown in FIG. 11, WR034, WR037 and WR043 all inhibit proliferation of colorectal cancer cells at the tested concentrations, and the effects of WR034 and WR037 are superior to those of 5-FU.
The embodiments of the present invention have been described above. However, the present invention is not limited to the above embodiment. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (10)

1. A compound having the structure of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof,
wherein R is 1 Is C 1-20 Alkyl, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl, 5-10 membered heterocyclyl, 5-10 membered heteroaryl, C 1-20 Alkyloxy, C 6-10 Arylcyclooxy, C 3-10 Cycloalkyloxy, 5-to 10-membered heterocyclyloxy or 5-to 10-membered heteroaryloxy, wherein the alkyl, aryl, cycloalkyl, heterocyclyl, heteroaryl, alkyloxy, aryl-aryloxy, cycloalkyloxy, heterocyclyloxy, heteroaryloxy is optionally substituted with one or more groups selected from halogen, C 1-10 Alkyloxy, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl, 5-10 membered heterocyclyl, 5-10 membered heteroaryl, C 6-10 Arylcyclooxy, C 3-10 A cycloalkyloxy, 5-10 membered heterocyclyloxy or 5-10 membered heteroaryloxy group;
R 2 is hydrogen, C 1-20 Alkyl, C 6-10 Aromatic ring radical, C 3-10 Cycloalkyl, 5-10 membered heterocyclyl, or 5-10 membered heteroaryl;
R 3 is hydrogen, halogen or C 1-6 Alkyl, C 1-6 An alkyl oxy group;
each R 4 Identical or different, independently selected from hydrogen or C 1-12 An alkyl group.
2. A compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, according to claim 1 wherein C 6-10 The aromatic ring radical being phenyl, C 3-10 Cycloalkyl radicals are C 3-7 Cycloalkyl, 5-10 membered heterocyclyl is 5-7 membered heterocyclyl, 5-10 membered heteroaryl is 5-6 membered heteroaryl, halogenF, cl or Br.
3. A compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, according to claim 1 wherein R 1 Is C 1-12 Alkyl, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, C 1-12 Alkyloxy, phenyloxy, C 3-7 Cycloalkyloxy, 5-7 membered heterocyclyloxy, 5-6 membered heteroaryloxy, wherein the alkyl, phenyl, cycloalkyl, heterocyclyl, heteroaryl, alkyloxy, phenyloxy, cycloalkyloxy, heterocyclyloxy, heteroaryloxy is optionally substituted with one or more groups selected from F, cl, br, or the alkyloxy is optionally substituted with one or more groups selected from C 1-6 Alkyloxy, phenyl, C 3-7 Cycloalkyl, 5-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyloxy, C 3-7 A cycloalkyloxy, 5-7 membered heterocyclyloxy or 5-6 membered heteroaryloxy group;
preferably, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, cyclopentyl, tetrahydrofuranyl, thiolanyl, pyrrolidinyl, tetrahydropyranyl, thietanyl, piperidinyl, C 1-8 An alkyloxy, phenyloxy, cyclohexyloxy, cyclopentyloxy, tetrahydrofuranyloxy, thiolanyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thiolanyloxy, piperidinyloxy, wherein the alkyloxy is optionally substituted with one or more groups selected from F, cl, br, C 1-6 Alkyl oxygen, phenyl, tetrahydrofuran, cyclohexyl, cyclopentyl, thiaclopentalkyl, pyrrolidinyl, tetrahydropyranyl, thiacyclohexanyl, piperidinyl, phenyloxy, tetrahydrofuranyloxy, cyclohexyloxy, cyclopentyloxy, thiaclopentalkyloxy, pyrrolidinyloxy, tetrahydropyranyloxy, thiacyclohexanyloxy, piperidinyloxy;
more preferably, R 1 Is C 1-8 Alkyl, phenyl, cyclohexyl, tetrahydrofuranyl, C 1-8 Alkyloxy, phenyloxy, cyclohexyloxy, tetrahydrofuranyloxy, wherein alkyl isThe oxygen radical being optionally substituted by one or more radicals selected from F, cl, br, C 1-6 Alkyloxy, phenyl, tetrahydrofuranyl, cyclohexyl, phenyloxy, tetrahydrofuranyloxy, cyclohexyloxy.
4. A compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, as claimed in any one of claims 1 to 3 wherein R 2 Is hydrogen or C 1-12 An alkyl group;
preferably, R 2 Is hydrogen or C 1-4 An alkyl group;
more preferably, R 2 Is methyl.
5. A compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, as claimed in any one of claims 1 to 4 wherein R 3 Is hydrogen or C 1-4 An alkyl group;
preferably R 3 Is hydrogen.
6. A compound of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, as claimed in any one of claims 1 to 5 wherein R 4 Is hydrogen or C 1-4 An alkyl group;
preferably, R 4 Is methyl.
7. A compound of formula I according to claim 1, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, selected from the following compounds:
8. a pharmaceutical composition comprising a compound of the structure of formula I, an enantiomer, a pharmaceutically acceptable salt or solvate thereof, as claimed in any one of claims 1 to 7, and preferably further comprising one or more pharmaceutically acceptable carriers.
9. Use of a compound of the structure of formula I according to any one of claims 1 to 7, an enantiomer, a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition according to claim 8 for the preparation of a medicament for inhibiting inflammation or an anti-tumour medicament or a VEGF inhibitor.
10. The use according to claim 9, wherein the inflammation is selected from an autoimmune disease, disorder or condition, an inflammatory disease, disorder or condition; preferably, it is: idiopathic pulmonary fibrosis, inflammatory bowel disease (selected from crohn's disease and ulcerative colitis), rheumatoid arthritis, osteoarthritis, stell's disease, sjogren's syndrome, systemic lupus erythematosus, multiple sclerosis, psoriasis, systemic sclerosis, acute respiratory distress syndrome, allergic rhinitis, asthma, ocular inflammatory diseases (e.g., allergic conjunctivitis, dry eye (dry eye) and uveitis), atopic dermatitis, interstitial cystitis, chronic prostatitis/chronic pelvic pain syndrome (chronic pelvic pain syndrome) (CP/CPPS), epidermocontact hypersensitivity (dermal contact hypersensitivy), eosinophilic gastrointestinal disorder (eosiniphilic gastrointestinal disorder), fibromyalgia, liver fibrosis, irritable bowel syndrome, ischemia reperfusion disease, renal fibrosis, pancreatitis, post-operative inflammation, seronegative spinal arthropathy (e.g., ankylosing spondylitis, psoriatic arthritis and reiter's syndrome), and inflammation (e.g., wegener's granulomatosis, sarcoidosis, leukosis, vascular-stoneley arteritis (35, 35-35) and vascular chebulimitis (35);
The tumor is selected from leukemia (such as acute leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, polycythemia vera), lymphoma (Hodgkin's disease, non-Hodgkin's disease), primary macroglobulinemia, heavy chain disease, solid tumors such as sarcomas and cancers (such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovial vioma, mesothelioma), ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer (e.g., triple negative breast cancer), ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary adenocarcinoma, bronchogenic carcinoma, medullary carcinoma, renal cell carcinoma, liver cancer, nile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, wilms' cell tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small cell lung cancer, bladder cancer, epithelial cancer, glioma, astrocytoma, medulloblastoma, craniopharyngeoma, ependymoma, pineal tumor, angioblastoma, auditory neuroma, oligodendroglioma, neuroblastoma, meningioma, melanoma, neuroblastoma, retinoblastoma), esophageal carcinoma, gall bladder cancer, renal carcinoma, multiple myeloma; preferably, the tumors include, but are not limited to: pancreatic cancer, liver cancer, lung cancer, stomach cancer, esophageal cancer, head and neck squamous cell carcinoma, prostate cancer, colon cancer, breast cancer (e.g., triple negative breast cancer), lymphoma, gall bladder cancer, kidney cancer, leukemia, multiple myeloma, ovarian cancer, cervical cancer, and glioma.
CN202311595446.9A 2023-11-27 2023-11-27 Chalcone derivative with benzopyran structure, and preparation method and application thereof Pending CN117603175A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311595446.9A CN117603175A (en) 2023-11-27 2023-11-27 Chalcone derivative with benzopyran structure, and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311595446.9A CN117603175A (en) 2023-11-27 2023-11-27 Chalcone derivative with benzopyran structure, and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN117603175A true CN117603175A (en) 2024-02-27

Family

ID=89945754

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311595446.9A Pending CN117603175A (en) 2023-11-27 2023-11-27 Chalcone derivative with benzopyran structure, and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN117603175A (en)

Similar Documents

Publication Publication Date Title
CN107001347B (en) Oxa- loop coil analog derivative, preparation method and its application in medicine
KR102534185B1 (en) Biaryl derivatives, synthesis method thereof and pharmaceutical use thereof
CN104507921B (en) Benzocyclobutene derivative and preparation method and pharmaceutical application thereof
CN111171049B (en) Tyrosine kinase inhibitors and uses thereof
US20130109749A1 (en) Micheliolide derivatives, medicinal composition, producing method and usage thereof
JP7190036B2 (en) MACROCYCLIC TYROSINE KINASE INHIBITOR AND USE THEREOF
CN112979528B (en) Tegaserod water-soluble organic acid salt and preparation method and application thereof
US9029403B2 (en) Treatment of ovarian cancer with benzylidenebenzohydrazides
AU2021105895A4 (en) Lycoline B-aryl acrylate derivatives, preparation method and application thereof
CN112047880B (en) Azaflavone derivatives and application thereof as antitumor drugs
CN117603175A (en) Chalcone derivative with benzopyran structure, and preparation method and application thereof
CN117603035A (en) Chalcone derivative and preparation method and application thereof
CN115677662A (en) Indazole compound, preparation method and medical application thereof
CN115433207A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN102584679B (en) Benzocarbazole acylamide compound and preparation method and application thereof
WO2021143927A1 (en) Compound acting as bcr-abl inhibitor
CA2855245A1 (en) Treatment of ovarian cancer with 2-amino-4h-naphtho[1,2-b]pyran-3-carbonitriles
CN111825608A (en) Tetrahydroquinoline and tetrahydroisoquinoline compounds and application thereof
RU2798231C2 (en) Macrocyclic tyrosine kinase inhibitor and its use
CN115701429B (en) 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof
CN116239594B (en) 6- (imidazo [1,2-a ] pyridin-6-yl) quinazoline derivatives and uses thereof
CN112375112B (en) Benzimidazole derivative BI361 and preparation method and application thereof
CN114105977B (en) Estrogen receptor modulator compounds and uses thereof
KR20240004539A (en) Condensed ring compounds as WEE-1 inhibitors, preparation methods therefor, and uses thereof
WO2023116812A1 (en) Sulfonylurea compound

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