WO2021190425A1 - 转化生长因子-β受体抑制剂 - Google Patents

转化生长因子-β受体抑制剂 Download PDF

Info

Publication number
WO2021190425A1
WO2021190425A1 PCT/CN2021/081965 CN2021081965W WO2021190425A1 WO 2021190425 A1 WO2021190425 A1 WO 2021190425A1 CN 2021081965 W CN2021081965 W CN 2021081965W WO 2021190425 A1 WO2021190425 A1 WO 2021190425A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
alkyl
cycloalkyl
group
Prior art date
Application number
PCT/CN2021/081965
Other languages
English (en)
French (fr)
Inventor
俞智勇
李刚
王坤瑶
邱庆崇
季彬
孙钊
Original Assignee
杭州阿诺生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 杭州阿诺生物医药科技有限公司 filed Critical 杭州阿诺生物医药科技有限公司
Publication of WO2021190425A1 publication Critical patent/WO2021190425A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention provides a new type of heterocyclic compound, its preparation method and its application as TGF- ⁇ receptor (especially TGF ⁇ RI) antagonist.
  • Transforming growth factor- ⁇ (transforming growth factor- ⁇ , TGF- ⁇ ) is a multifunctional growth factor and cytokine. It is combined with activins, inhibitors, and Mullerian inhibitors. Substance, MIS) and bone morpho-genetic proteins (BMPs) and other related proteins form the transforming growth factor- ⁇ superfamily. It can participate in the regulation of a variety of biological processes, including cell proliferation, differentiation, development, and modification of extracellular matrix (including tumor matrix and immunosuppression, angiogenesis and fibrous tissue generation).
  • MIS transforming growth factor- ⁇
  • BMPs bone morpho-genetic proteins
  • TGF- ⁇ RI type I
  • TGF ⁇ RII type II
  • TGF ⁇ RIII type III
  • the signal transduction in the TGF- ⁇ signaling pathway is mainly through the combination of TGF- ⁇ ligand with the serine/threonine kinase receptor TGF ⁇ RI (ALK5) and TGF ⁇ RII on the cell surface to form a heterotetrameric complex, and then TGF ⁇ RII interacts with TGF ⁇ RI
  • ALK5 serine/threonine kinase receptor
  • TGF ⁇ RII TGF ⁇ RI
  • the glycine/serine region phosphorylates and activates the phosphorylation of TGF ⁇ RI to the intracellular signaling protein Smad protein molecule-Smad2/Smad3 that is connected to it.
  • TGF ⁇ RIII transforming growth factor- ⁇ III receptor
  • TGF- ⁇ plays a key role in the occurrence and progression of fibrosis in different organ systems such as heart, liver, lung, and kidney.
  • Abnormal TGF- ⁇ signaling is related to many human diseases, such as cancer, cardiovascular disease, inflammation, organ fibrotic disease, pancreatic disease, etc.
  • the dysregulation of TGF- ⁇ signaling is very common, which can promote tumor cell growth and differentiation, and regulate extracellular matrix and epithelial-mesenchymal transition.
  • TGF- ⁇ has been proven to play an important role in regulating anti-tumor immunity.
  • TGF- ⁇ has a strong inhibitory effect on the differentiation of T lymphocytes, and has a significant negative effect on dendritic cells, CD8 + T cells and NK cells. At the same time, it can enhance immunosuppressive T reg and bone marrow-derived suppressor cells (MDSC). ), thereby providing a favorable tumor microenvironment for tumor growth and metastasis. Therefore, inhibiting the signal transduction of a certain link in the TGF- ⁇ signaling pathway may become one of the effective methods for the treatment of tumors.
  • MDSC bone marrow-derived suppressor cells
  • TGF ⁇ RI As an important node in the TGF- ⁇ signaling pathway, TGF ⁇ RI (ALK5) is considered to be an important target for tumor treatment. By preventing the binding of TGF ⁇ RI to ligands, it inhibits the phosphoric acid of ALK5 on its downstream signaling protein (Smad2 or Smad3). In order to prevent and treat various related diseases mediated by ALK5, it can affect or block TGF- ⁇ signal transmission.
  • Cy represents a C 6 -C 10 aryl group or a 5-10 membered heteroaryl group substituted by 0, 1, 2 or 3 substituents, wherein the substituents are selected from: halogen, C 1 -C 6 alkyl , C 3 -C 6 cycloalkyl, halo C 1 -C 6 alkyl, -OR a , cyano, nitro, NR a R b , -SO 2 R a , -C(O)R a ,- C(O)OR a , -C(O)NR a R b , -P(O)R a R b ;
  • R 1 represents hydrogen, halogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, hydroxy (C 1 -C 6 alkyl), halogenated C 1 -C 6 alkyl;
  • R 2 represents hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, hydroxy (C 1 -C 6 alkyl), halogenated C 1 -C 6 alkyl, -OR a , cyano, Nitro, NR a R b , -SO 2 R a , -C(O)R a , -C(O)OR a , -C(O)NR a R b , -NR a C(O)R b , -SO 2 NR a R b , -NR a SO 2 R b ;
  • R 3 represents hydrogen, halogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, hydroxy (C 1 -C 6 alkyl), halogenated C 1 -C 6 alkyl, -OR a , cyanide Group, nitro group, NR a R b , -SO 2 R a , -C(O)R a , -C(O)OR a , -C(O)NR a R b , -NR a C(O)R b , -SO 2 NR a R b , -NR a SO 2 R b ;
  • R 4 represents hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, halogenated C 1 -C 6 alkyl, -OR a , -C(O)R a , -C(O)OR a , -C(O)NR a R b , hydroxyl (C 1 -C 6 alkyl);
  • R 5 represents hydrogen, C 1 -C 6 alkyl, C 2- C 6 alkene Group, C 3 -C 6 cycloalkyl, halo C 1 -C 6 alkyl; or R 4 , R 5 together with the atoms they are connected together form a 5-8 membered saturated and unsaturated ring, and the ring can also Randomly contain 0, 1 or 2 heteroatoms selected from N, O, S;
  • R a and R b each independently represent hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, or halogenated C 1 -C 6 alkyl; or R a, R b form together with the atoms to which they are attached a 5-8 membered ring which may also optionally contain 1 or 2 heteroatoms selected from N, O, S, heteroatoms;
  • n 0, 1, 2 or 3.
  • the compound of formula (I) has the following structure of formula (II):
  • R 1, R 3, R 4 , R 5, R a, R b and n have the meanings according to claim 1;
  • R 2 represents hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, hydroxy (C 1 -C 6 alkyl), halogenated C 1 -C 6 alkyl;
  • W represents CR L or N
  • R 6 represents hydrogen, halogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, halo C 1 -C 6 alkyl, -OR a , cyano, nitro, NR a R b ,- SO 2 R a , -C(O)R a , -C(O)OR a , -C(O)NR a R b , -P(O)R a R b ;
  • R L represents hydrogen, halogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, halo C 1 -C 6 alkyl, -OR a , cyano, nitro, NR a R b ,- SO 2 R a , -C(O)R a , -C(O)OR a , -C(O)NR a R b , -P(O)R a R b ;
  • o 0, 1, 2 or 3.
  • the compound of the present invention is selected from the following structures:
  • the compounds of the present invention can also be prepared in the form of pharmaceutically acceptable salts, which can be formed using, for example, the following inorganic or organic acids: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, acetic acid, glycolic acid , Lactic acid, pyruvic acid, malonic acid, succinic acid, glutaric acid, fumaric acid, malic acid, mandelic acid, tartaric acid, citric acid, ascorbic acid, palmitic acid, maleic acid, hydroxymaleic acid, benzoic acid, hydroxyl Benzoic acid, phenylacetic acid, cinnamic acid, salicylic acid, methanesulfonic acid, benzenesulfonic acid or toluenesulfonic acid.
  • inorganic or organic acids hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, acetic acid, glycolic acid , Lactic acid,
  • the pharmaceutically acceptable salt of the present invention can be prepared by conventional methods, for example, by dissolving the compound of the present invention in an organic solvent miscible with water (such as acetone, methanol, ethanol, and acetonitrile), and adding an excessive amount of organic acid or An aqueous inorganic acid solution is used to precipitate the salt from the resulting mixture, the solvent and remaining free acid are removed therefrom, and then the precipitated salt can be separated.
  • an organic solvent miscible with water such as acetone, methanol, ethanol, and acetonitrile
  • the compound of the present invention may include a solvate form, preferably, the solvate is a hydrate.
  • the present invention also provides the use of the compound of the present invention in the preparation of a medicine for preventing or treating diseases that can be regulated by inhibiting the activity of ALK5.
  • the disease is selected from cancer, tumor, inflammatory disease, autoimmune disease and immune-mediated disease.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, which comprises the formula of the present invention (I) Compound as active ingredient.
  • the present invention provides a method for preventing or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, which includes Of mammals administer the compounds of the invention.
  • cancer or tumors may include, but are not limited to, skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neurocytoma, rectal cancer , Colon cancer, familial adenomatous polyposis cancer, hereditary non-polyposis colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, hypopharyngeal cancer, tongue cancer, salivary gland cancer, gastric cancer, adenocarcinoma, medullary thyroid cancer, Papillary thyroid cancer, kidney cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, uterine body cancer, endometrial cancer, choriocarcinoma, testicular cancer, urinary cancer, melanoma, brain tumors such as glioblastoma, Astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodg
  • the compound of the present invention or a pharmaceutically acceptable salt thereof when administered in combination with another anticancer agent or checkpoint inhibitor for the treatment of cancer or tumor, the compound of the present invention or a pharmaceutically acceptable salt thereof can provide an enhanced anticancer effect.
  • anticancer agents for the treatment of cancer or tumors may include, but are not limited to, cell signal transduction inhibitors, chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carbohydrate Mustine, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, Mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectidine, dactinomycin, doxorubicin , Epirubicin, daunomycin, mitoxantrone, bleomycin, mitomycin C, ixabepilone, tamoxi
  • inflammatory diseases may include, but are not limited to, arthritis, rheumatoid arthritis, spondyloarthritis, gouty arthritis, osteoarthritis, juvenile arthritis , Other arthritic conditions, lupus, systemic lupus erythematosus (SLE), skin-related diseases, psoriasis, eczema, dermatitis, allergic dermatitis, pain, lung disease, lung inflammation, adult respiratory distress syndrome (ARDS) , Pulmonary sarcoidosis, chronic pulmonary inflammatory disease, chronic obstructive pulmonary disease (COPD), cardiovascular disease, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia reperfusion injury, inflammatory bowel disease, Crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, Sjogren's syndrome, autoimmune thyroid disease, urticaria (rubella), multiple sclerosis,
  • the compound of the present invention or a pharmaceutically acceptable salt thereof when administered in combination with another therapeutic agent for the treatment of inflammatory diseases, autoimmune diseases and immune-mediated diseases, the compound of the present invention or a pharmaceutically acceptable salt thereof can provide enhanced Therapeutic effect.
  • therapeutic agents for the treatment of inflammatory diseases, autoimmune diseases, and immune-mediated diseases may include, but are not limited to, steroid drugs (e.g., prednisone, hydroprednisolone, methylhydroprednisolone) Pine, cortisone, hydroxycortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-TNF ⁇ agents (e.g., Etanercept, Infliximab, Adali Monoclonal antibodies, etc.), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, etc.) and antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebas , Ketotifen, cetirizine, levocetirizine, fexofenadine, etc.), and at least one therapeutic agent selected from them may be included in the pharmaceutical composition of the present invention.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be administered orally or parenterally as an active ingredient, and its effective amount ranges from 0.1 to 2,000 mg/kg body weight/day in the case of mammals including humans (body weight of about 70 kg). It is preferably 1 to 1,000 mg/kg body weight/day, and administered in a single or 4 divided doses per day, or following/not following a predetermined time.
  • the dosage of the active ingredient can be adjusted according to a number of relevant factors (such as the condition of the subject to be treated, the type and severity of the disease, the rate of administration, and the doctor's opinion). In some cases, an amount less than the above dosage may be appropriate. If it does not cause harmful side effects, an amount larger than the above dose can be used and the amount can be administered in divided doses per day.
  • the pharmaceutical composition of the present invention can be formulated into dosage forms for oral administration or parenteral administration (including intramuscular, intravenous and subcutaneous routes) according to any of the conventional methods, such as tablets, granules, powders, capsules, syrups , Emulsion, microemulsion, solution or suspension.
  • the pharmaceutical composition of the present invention for oral administration can be prepared by mixing the active ingredient with a carrier such as the following: cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, hard Magnesium stearate, calcium stearate, gelatin, talc, surfactants, suspending agents, emulsifiers and diluents.
  • a carrier such as the following: cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, hard Magnesium stearate, calcium stearate, gelatin, talc, surfactants, suspending agents, emulsifiers and diluents.
  • carriers used in the injection composition of the present invention are water, salt solution, glucose solution, glucose-like solution, alcohol, glycol, ether (e.g., polyethylene glycol 400), oil, fatty acid, fatty acid ester,
  • the compounds of the present invention can be prepared in a variety of ways known to those skilled in the field of organic synthesis, and the compounds of the present invention can be synthesized using synthetic methods known in the field of organic synthetic chemistry or through variations known to those skilled in the art. .
  • the desired reaction can be carried out in a solvent or solvent mixture suitable for the kit materials used and suitable for the transformation achieved.
  • the present invention describes the cis- and trans- (or E- and Z-) geometric isomers of the compounds of the present invention, and they can be separated into a mixture of isomers or separate isomer forms.
  • the compounds of the present invention can be isolated in optically active or racemic form.
  • the compounds of the present invention may exist in a variety of tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecule and the chemical bonds between the atoms of the molecule are thus rearranged. It should be understood that all tautomeric forms that may exist are included in the present invention.
  • substituent is selected from, for example, the following substituents such as alkyl, cycloalkyl, aryl, heterocyclyl, halogen, hydroxy, Alkoxy, oxo, alkanoyl, aryloxy, alkanoyloxy, amino, alkylamino, arylamino, arylalkylamino, disubstituted amine groups (two of the amino substituents are selected (From alkyl, aryl or arylalkyl), alkanoylamino, aroylamino, aralkanoylamino, substituted alkanoylamino, substituted arylamino, substituted aralkanoylamino, thio, alkyl Thio, arylthio, arylalkylthio, arylthiocarbonyl, arylalkylthiocarbonyl, alkylsul
  • substituents such as alkyl, cycloalkyl,
  • alkyl or "alkylene” as used herein is intended to include branched or straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C 1 -C 6 alkyl means an alkyl group having 1 to 6 carbon atoms.
  • alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, tert-butyl) and Pentyl (e.g., n-pentyl, isopentyl, neopentyl).
  • Preferred alkyl groups are C 1 -C 6 alkyl groups, and more preferred alkyl groups are C 1 -C 4 alkyl groups.
  • alkenyl refers to a straight or branched hydrocarbon group containing one or more double bonds and usually having a length of 2 to 20 carbon atoms.
  • C 2 -C 8 alkenyl contains two to eight carbon atoms.
  • Alkenyl includes, but is not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, heptenyl, octenyl, and the like.
  • a preferred alkenyl group is a C 2 -C 8 alkenyl group, and a more preferred alkenyl group is a C 2 -C 6 alkenyl group.
  • alkynyl means a straight or branched hydrocarbon group containing one or more triple bonds and usually having a length of 2 to 20 carbon atoms.
  • C 2 -C 8 alkynyl contains two to eight carbon atoms.
  • a preferred alkynyl group is a C 2 -C 8 alkynyl group, and a more preferred alkenyl group is a C 2 -C 6 alkynyl group.
  • Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl, and the like.
  • alkoxy refers to -O-alkyl.
  • C 1 -C 6 alkoxy (or alkyloxy) is intended to include C 1 , C 2 , C 3 , C 4 , C 5 and C 6 alkoxy.
  • a preferred alkoxy group is a C 1 -C 6 alkoxy group, and a more preferred alkoxy group is a C 1 -C 4 alkoxy group.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (such as n-propoxy and isopropoxy), and tert-butoxy.
  • alkylthio or “thioalkoxy” means an alkyl group, as defined above, with the specified number of carbon atoms connected via a sulfur bridge; for example, methyl-S- and ethyl-S-.
  • aryl alone or as part of a larger part such as “aralkyl”, “aralkoxy” or “aryloxyalkyl”, refers to a monocyclic ring having a total of 5 to 12 ring members , A bicyclic or tricyclic ring system, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • a monocyclic aromatic group refers to a phenyl group
  • a bicyclic or more aromatic group refers to naphthyl, anthracenyl, etc.
  • the aryl bicyclic ring can also be a benzene ring fused with a cycloalkyl group or a ring Alkenyl, or fused with a cycloalkynyl group.
  • the preferred aryl group is a C 6 -C 12 aryl group.
  • "aryl" refers to an aromatic ring system, which includes, but is not limited to, phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl, and tetrahydronaphthalene base.
  • aralkyl or "arylalkyl” refers to an alkyl residue attached to an aryl ring. Non-limiting examples include benzyl, phenethyl, and the like.
  • the fused aryl group may be connected to another group at a suitable position on the cycloalkyl ring or aromatic ring.
  • Example The arrow line drawn from the ring system indicates that the bond can be connected to any suitable ring atom.
  • cycloalkyl refers to a monocyclic or bicyclic cyclic alkyl group.
  • the monocyclic cyclic alkyl group refers to a C 3 -C 8 cyclic alkyl group, including but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbornanyl.
  • Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included in the definition of "cycloalkyl”.
  • the bicyclic cyclic alkyl group includes a bridged ring, a spiro ring, or a fused ring cycloalkyl group.
  • the preferred cycloalkyl group is a C 3 -C 6 cycloalkyl group.
  • cycloalkenyl refers to a monocyclic or bicyclic cyclic alkenyl.
  • the monocyclic cyclic alkenyl group refers to a C 3 -C 8 cyclic alkenyl group, including but not limited to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, and norbornenyl.
  • Branched cycloalkenyl groups such as 1-methylcyclopropenyl and 2-methylcyclopropenyl are included in the definition of "cycloalkenyl”.
  • the bicyclic cyclic alkenyl group includes a bridged ring, a spiro ring, or a fused ring cyclic alkenyl group.
  • hydroxyalkyl refers to a branched and straight chain saturated aliphatic hydrocarbon group with the specified number of carbon atoms and at least one -OH is attached to the carbon atom of the hydrocarbon group.
  • hydroxyalkyl groups include, but are not limited to, hydroxymethyl, hydroxyethyl, 1-hydroxypropyl, 2-hydroxypropyl.
  • Halo or halogen includes fluorine, chlorine, bromine and iodine.
  • Haloalkyl is intended to include branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and substituted with one or more halogens.
  • haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoromethyl Propyl and heptachloropropyl.
  • haloalkyl groups also include "fluoroalkyl groups” intended to include branched and straight chain saturated aliphatic hydrocarbon groups having a designated number of carbon atoms and substituted with one or more fluorine atoms.
  • Haloalkoxy or "haloalkyloxy” means a haloalkyl group as defined above with the specified number of carbon atoms connected via an oxygen bridge.
  • C 1 -C 6 haloalkoxy is meant to include C 1 , C 2 , C 3 , C 4 , C 5 and C 6 haloalkoxy.
  • Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluoroethoxy.
  • haloalkylthio or “thiohaloalkoxy” means a haloalkyl group as defined above that has the specified number of carbon atoms connected by a sulfur bridge; for example, trifluoromethyl-S- and pentafluoroethyl -S-.
  • heteroaryl means a stable 3-membered, 4-membered, 5-membered, 6-membered, or 7-membered aromatic monocyclic or aromatic bicyclic ring or 7-membered, 8-membered, 9-membered, 10-membered, 11-membered, 12-membered , 13-membered or 14-membered aromatic polycyclic heterocyclic ring, which is fully unsaturated, partially unsaturated, and contains carbon atoms and 1, 2, 3 or 4 independently selected from N, O and S And includes any of the following polycyclic groups in which any heterocyclic ring as defined above is fused with a benzene ring. Nitrogen and sulfur heteroatoms can optionally be oxidized.
  • the nitrogen atom is substituted or unsubstituted (ie N or NR, where R is H or another substituent if defined).
  • the heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. If the resulting compound is stable, the heterocyclic group described herein may be substituted on a carbon or nitrogen atom.
  • the nitrogen in the heterocyclic ring can optionally be quaternized.
  • the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to each other.
  • the total number of S and O atoms in the heterocyclic ring is not more than one.
  • Preferred heteroaryl groups are 5-12 membered heteroaryl groups.
  • heteroaryl groups include, but are not limited to, acridinyl, azetidinyl, azetidinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothienyl, benzoxazole Group, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH -Carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3 -b] Tetrahydrofuryl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H
  • heterocycloalkyl refers to a monocyclic heterocycloalkyl system, or a bicyclic heterocycloalkyl system, and also includes spiro heterocycles or bridged heterocycloalkyls.
  • the monocyclic heterocycloalkyl refers to a 3-8 membered, and at least one saturated or unsaturated but not aromatic cyclic alkyl system selected from O, N, S, and P.
  • the bicyclic heterocycloalkyl system refers to a heterocycloalkyl group fused to a phenyl group, or a cycloalkyl group, or a cycloalkenyl group, or a heterocycloalkyl group, or a heteroaryl group.
  • a preferred heterocycloalkyl group is a 3-12 membered heterocycloalkyl group.
  • substitution means that at least one hydrogen atom is replaced by a non-hydrogen group, provided that the normal valence is maintained and the substitution results in a stable compound.
  • nitrogen atoms such as amines
  • these nitrogen atoms can be converted into N-oxides by treatment with an oxidizing agent (such as m-CPBA and/or hydrogen peroxide) to obtain the present invention
  • an oxidizing agent such as m-CPBA and/or hydrogen peroxide
  • any variable occurs more than once in any composition or formula of a compound, its definition at each occurrence is independent of its definition at every other occurrence.
  • the group may be optionally substituted with up to three R groups, and R is independently selected from R at each occurrence. definition.
  • substituents and/or variables are only allowed if the above-mentioned combinations produce stable compounds.
  • solvate means the physical association of a compound of the invention with one or more solvent molecules (whether organic or inorganic). This physical association includes hydrogen bonding. In some cases, such as when one or more solvent molecules are incorporated into the crystal lattice of the crystalline solid, the solvate will be able to be separated.
  • the solvent molecules in the solvate can be arranged in regular and/or disordered arrangements.
  • Solvates may contain stoichiometric or non-stoichiometric solvent molecules.
  • “Solvate” encompasses both solution-phase and isolatable solvates. Exemplary solvates include, but are not limited to, hydrate, ethanolate, methanolate, and isopropanolate. Solvation methods are well known in the art.
  • patient refers to an organism that is treated by the method of the present invention.
  • organisms preferably include, but are not limited to, mammals (e.g., murine, ape/monkey, horse, cow, pig, dog, cat, etc.) and most preferably refer to humans.
  • an effective amount means the amount of a drug or agent (ie, a compound of the present invention) that will elicit a biological or medical response of a tissue, system, animal, or human sought by, for example, a researcher or clinician.
  • therapeutically effective amount means an amount that results in an improved treatment, cure, prevention, or alleviation of a disease, disorder, or side effect, or reduction in disease, compared to a corresponding subject who has not received the above-mentioned amount. Or the rate of progression of the disease.
  • the effective amount can be given in one or more administrations, administrations or doses and is not intended to be limited by a particular formulation or route of administration. The term also includes within its scope an effective amount for enhancing normal physiological functions.
  • treatment includes any effect that leads to amelioration of a condition, disease, disorder, etc., such as alleviation, reduction, regulation, amelioration or elimination, or amelioration of its symptoms.
  • a certain compound or pharmaceutical composition can improve a certain disease, symptom or condition after administration, especially its severity is improved, the onset is delayed, the progression of the disease is slowed, or the duration of the disease is reduced. Regardless of fixed administration or temporary administration, continuous administration or intermittent administration, it can be attributed to or related to the administration.
  • prevention refers to preventing, blocking, eliminating the occurrence of a disease or interfering with or slowing the development of a disease before the occurrence of a disease or symptom.
  • composition refers to a combination of an active agent and an inert or active carrier, so that the composition is particularly suitable for in vivo or ex vivo diagnosis or treatment.
  • bases include, but are not limited to, alkali metal (e.g., sodium) hydroxide, alkaline earth metal (e.g., magnesium) hydroxide, ammonia, and the like.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium hydroxide
  • ammonia e.g., sodium
  • salt of the compound of the present invention is intended for therapeutic use, and the salt of the compound of the present invention is expected to be pharmaceutically acceptable.
  • salts of non-pharmaceutical acids and bases can also be used, for example, in the preparation or purification of pharmaceutical compounds.
  • immediate is used herein to refer to the following compounds, substances, compositions and/or dosage forms: within the scope of reasonable medical judgment, they are suitable for use in contact with human and animal tissues without excessive toxicity or irritation Sex, allergic reactions, and/or other problems or complications, and are commensurate with a reasonable benefit/risk ratio.
  • phrases "pharmaceutically acceptable carrier” and “pharmaceutically acceptable carrier” mean pharmaceutical substances, compositions or vehicles, such as liquid or solid fillers, diluents, excipients, manufacturing aids (e.g. Lubricants, talc, magnesium stearate, calcium stearate or zinc stearate or stearic acid) or solvent encapsulated substances, which involve carrying or transporting the active compound from one organ or part of the body to another organ or Parts of the body.
  • manufacturing aids e.g. Lubricants, talc, magnesium stearate, calcium stearate or zinc stearate or stearic acid
  • solvent encapsulated substances which involve carrying or transporting the active compound from one organ or part of the body to another organ or Parts of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not harmful to the patient.
  • acceptable refers to a prescription component or active ingredient that does not have undue harmful effects on the health of the general treatment target.
  • cancer refers to an abnormal growth of cells that cannot be controlled and which can metastasize (spread) under certain conditions.
  • This type of cancer includes, but is not limited to, solid tumors (such as bladder, intestine, brain, chest, uterus, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas or other endocrine organs (such as thyroid), prostate , Skin (melanoma) or hematoma (such as non-leukemic leukemia).
  • co-administration refers to administering several selected therapeutic drugs to a patient in the same or different modes of administration at the same or different times.
  • an “enhance” or “enhance”, as used herein, means that the expected result can be increased or prolonged in terms of potency or duration. Therefore, in terms of enhancing the therapeutic effect of a drug, the term “enhanced” refers to the ability of the drug to increase or extend the potency or duration of the drug in the system. "Synergy value” as used herein refers to the ability of another therapeutic drug to be maximized in an ideal system.
  • immune disease refers to a disease or symptom that produces an adverse or deleterious reaction to an endogenous or exogenous antigen. The result is usually cell dysfunction, or destruction and dysfunction, or destruction of organs or tissues that may produce immune symptoms.
  • subject or “subject” includes mammals and non-mammals.
  • Mammals include, but are not limited to, mammals: humans, non-human primates such as orangutans, apes and monkeys; agricultural animals such as cows, horses, goats, sheep, pigs; domestic animals such as rabbits and dogs; laboratory animals include rodents, Such as rats, mice and guinea pigs.
  • Non-mammalian animals include, but are not limited to, birds, fish, etc.
  • the selected mammal is a human.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ocular, pulmonary, transdermal, vaginal, and otic administration , Nasal administration and local administration.
  • parenteral administration includes intramuscular injection, subcutaneous injection, intravenous injection, intramedullary injection, ventricular injection, intraperitoneal injection, intralymphatic injection, and intranasal injection.
  • the mode of administration of the compounds described herein is a local rather than a systemic mode of administration.
  • the long-acting formulation is administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is administered through a targeted drug delivery system.
  • liposomes encapsulated by organ-specific antibodies In this specific embodiment, the liposomes are selectively targeted to specific organs and absorbed.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of one or more compounds of the present invention formulated together with one or more pharmaceutical carriers (additives) and/or diluents, and optionally one One or more of the above-mentioned other therapeutic agents.
  • the compound of the present invention can be administered by any suitable means for any of the above-mentioned purposes, for example, oral administration, such as tablets, pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried Dispersions), syrups and emulsions; sublingually; buccal; parenterally, such as by subcutaneous, intravenous, intramuscular or intrasternal injection or infusion techniques (for example, in a sterile injectable aqueous or non-aqueous solution or suspension Liquid form); nasal, including administration to the nasal membranes, such as by inhalation spray; topical, such as in the form of creams or ointments; or transrectally, such as in the form of suppositories. They can be administered alone, but are usually administered using a pharmaceutical carrier selected based on the chosen route of administration and standard pharmaceutical practice.
  • the pharmaceutical carrier is formulated according to many factors within the scope of those skilled in the art. These factors include, but are not limited to: the type and nature of the active agent formulated; the subject to whom the composition containing the active agent is to be administered; the expected route of administration of the composition; and the targeted therapeutic indication.
  • Pharmaceutical carriers include aqueous and non-aqueous liquid media and various solid and semi-solid dosage forms.
  • the above-mentioned carrier may include many different ingredients and additives in addition to the active agent, and the above-mentioned other ingredients are included in the formulation for various reasons known to those skilled in the art, such as stabilizing active agents, binders, and the like.
  • suitable pharmaceutical carriers and the factors involved in the selection of carriers please refer to many easily available sources, such as Allen LVJr.et al. Remington: The Science and Practice of Pharmacy(2Volumes), 22nd Edition (2012) ), Pharmaceutical Press.
  • the dosage regimen of the compound of the present invention varies depending on known factors, such as the pharmacodynamic properties of the specific agent and its administration mode and route; the species, age, sex, health status, medical condition and weight of the recipient. ; The nature and degree of symptoms; the types of simultaneous treatment; the frequency of treatment; the route of administration, the patient's renal and liver function, and the desired effect.
  • the daily oral dose of each active ingredient should be about 0.001 mg/day to about 10-5000 mg/day, preferably about 0.01 mg/day to about 1000 mg/day, and most preferably The ground is about 0.1 mg/day to about 250 mg/day.
  • the most preferred intravenous dose should be about 0.01 mg/kg/min to about 10 mg/kg/min.
  • the compounds of the present invention may be administered in a single daily dose, or the total daily dose may be administered in divided doses of two, three or four times daily.
  • the compound is usually selected as a suitable pharmaceutical diluent, excipient or carrier (herein Administered in the form of a mixture of drugs collectively referred to as drug carriers).
  • a dosage form (pharmaceutical composition) suitable for administration may contain about 1 mg to about 2000 mg of active ingredient per dosage unit.
  • the active ingredient will usually be present in an amount of about 0.1-95% by weight based on the total weight of the composition.
  • a typical capsule for oral administration contains at least one compound of the present invention (250 mg), lactose (75 mg) and magnesium stearate (15 mg). The mixture was passed through a 60-mesh screen and packed into No. 1 gelatin capsules.
  • a typical injectable preparation can be prepared by aseptically placing at least one compound of the present invention (250 mg) in a bottle, aseptically lyophilizing and sealing. For use, the contents of the bottle are mixed with 2 mL of physiological saline to produce an injectable preparation.
  • the scope of the present invention includes (alone or in combination with a pharmaceutical carrier) pharmaceutical compositions containing a therapeutically effective amount of at least one compound of the present invention as an active ingredient.
  • the compound of the present invention may be used alone, in combination with other compounds of the present invention, or in combination with one or more other therapeutic agents (e.g., anticancer agents or other pharmaceutically active substances).
  • the compound of the present invention (which can be used in a suitable hydrated form) and/or the pharmaceutical composition of the present invention is formulated into a pharmaceutical dosage form by conventional methods known to those skilled in the art.
  • the actual dosage level of the active ingredient in the pharmaceutical composition of the present invention can be changed, so as to obtain the amount of the active ingredient that is effective for realizing the desired therapeutic response, composition, and administration mode of a specific patient without being toxic to the patient.
  • the selected dosage level will depend on many factors, including the activity of the particular compound of the invention or its ester, salt or amide used; route of administration; time of administration; excretion rate of the particular compound used; rate and extent of absorption The duration of treatment; other drugs, compounds and/or substances used in combination with the specific compound used; the age, sex, weight, condition, general health and previous medical history of the patient being treated and other factors well known in the medical field.
  • a doctor or veterinarian with ordinary skill in the art can easily determine and prescribe an effective amount of the required pharmaceutical composition.
  • the physician or veterinarian can start a competition of the compound of the present invention used in the pharmaceutical composition at a level lower than the required level, and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of the compound of the invention will be the amount of the compound that is the lowest dose effective to produce a therapeutic effect.
  • Such effective doses usually depend on the above-mentioned factors.
  • oral, intravenous, intracerebroventricular, and subcutaneous doses of the compound of the present invention for patients range from about 0.01 to about 50 mg/kg body weight/day.
  • the effective daily dose of the active compound can be administered separately in two, three, four, five, six or more sub-doses at appropriate intervals throughout the day, optionally in unit dosage form.
  • the medication is administered once a day.
  • the compound of the present invention can be administered alone, it is preferable to administer the compound in the form of a pharmaceutical preparation (composition).
  • kits/product packaging is also described here.
  • kits can be composed of a conveyor, a medicine pack or a container box.
  • the container box can be divided into multiple compartments to accommodate one or more containers, such as vials, test tubes, and the like.
  • Each container contains all A single component in the method described. Suitable containers include bottles, vials, syringes and test tubes.
  • the container is made of acceptable materials such as glass or plastic.
  • the container may contain one or more of the compounds described herein.
  • the compounds may exist in the form of pharmaceutical components, or they may exist as a mixture with other ingredients described herein.
  • the container may have a sterile outlet (for example, the container may be an intravenous infusion bag or a bottle, and the stopper may be pierced by a hypodermic syringe needle).
  • kits may contain a compound, and instructions, labels, or operating instructions for the method of use described herein.
  • a typical kit may include one or more containers.
  • each container contains one or more materials (such as reagents, or concentrated mother liquor, and/ Or equipment).
  • materials include, but are not limited to, buffers, diluents, filters, needles, syringes, conveyors, bags, containers, bottles and/or test tubes, with a list of contents and/or instructions for use, and instructions for the built-in packaging. The entire set of instructions must be included.
  • the label can be displayed on the container or closely related to the container.
  • the appearance of a label on a container means that the label letters, numbers or other features are pasted, molded, or engraved on the container; the label can also appear in a container box or shipping box containing a variety of containers, such as in a product insert.
  • a label can be used to indicate a specific therapeutic use of the contents.
  • the label may also indicate instructions for use of the content, such as described in the above method.
  • the unit of weight-volume percentage in the present invention is well known to those skilled in the art, for example, refers to the weight of the solute in a 100 ml solution. Unless otherwise defined, all professional and scientific terms used in the text have the same meaning as those familiar to those skilled in the art. In addition, any methods and materials similar or equivalent to the content described can be applied to the method of the present invention. The preferred implementation methods and materials described in this article are for demonstration purposes only.
  • the relevant intermediates are commercially available (for example from Sigma Aldrich, Alfa).
  • the LC-MS experiment is measured under the following conditions:
  • Preparative HPLC usually uses an acidic method (gradient of acetonitrile and water, each containing 0.1% formic acid) with Thermo U3000 AFC-3000; column: Globalsil C-18 12nm, 250x 20mm, 10 ⁇ m, or equivalent; flow rate: 20mL/min, Separate.
  • N-bromosuccinimide (549 mg, 3.1 mmol) was added to N, N-dimethylformamide (5 mL) in which compound INT-1a (500 mg, 3.1 mmol) was dissolved, The resulting reaction liquid was stirred at the same temperature for 1.5 hours.
  • the reaction solution was poured into water (50 mL), and extracted with ethyl acetate (50 mL ⁇ 3); the combined organic phase was washed with water and saturated brine (100 mL each), dried over anhydrous sodium sulfate, and concentrated.
  • the reaction solution was quenched with a little water, poured into water (50mL), and extracted with ethyl acetate (50mL ⁇ 3); the combined organic phase was washed with water and saturated brine (100mL each), dried over anhydrous sodium sulfate, and concentrated .
  • Cyclopropyl methyl ketone (160mg, 1.9mmol) was added to a solution of sodium hydrogen (37mg, 1.6mmol, 60% w/w in kerosene) in tetrahydrofuran (10mL), and the resulting reaction solution was stirred at room temperature for half an hour After heating, it was raised to reflux; then, a solution of 4-fluoroacetophenone (250 mg, 1.6 mmol) in tetrahydrofuran (3 mL) was added dropwise. The resulting reaction solution was further stirred for half an hour under reflux.
  • 2,4-Dinitrophenylhydroxylamine (3.1g, 15.4mmol) was added to a solution of 4-(tert-butoxycarbonylamino)pyridine (3.0g, 15.4mmol) in 2-methyltetrahydrofuran (15mL); The resulting reaction solution was stirred at 40°C for 4 hours, then cooled to room temperature and stirred overnight. The reaction solution was filtered to obtain a yellow solid INT-4a (6.1 g, yield: 99.6%).
  • Diethyl oxalate (554mg, 3.8mmol) was added to a tetrahydrofuran (20mL) solution mixed with sodium hydrogen (74mg, 3.2mmol, 60% w/w in kerosene), and the resulting reaction solution was stirred at room temperature for half an hour. Heat to reflux; then add dropwise a solution of 4-fluoroacetophenone (500 mg, 3.6 mmol) in tetrahydrofuran (6 mL) previously dissolved. The resulting reaction solution was further stirred for half an hour under reflux.
  • Aqueous sodium hydroxide solution (3M, 1 mL) was added to a solution of compound 1a (147 mg, 0.31 mmol) in methanol (5 mL); the resulting reaction solution was stirred at 60° C. for 2 hours. After the reaction solution was cooled to room temperature, the pH was adjusted to 3-4 with 1M aqueous hydrochloric acid, and extracted with ethyl acetate (20 mL ⁇ 2). The combined organic phase was washed with saturated sodium bicarbonate and saturated brine (30 mL each), dried over anhydrous sodium sulfate, and concentrated to obtain a yellow oil 3a (122 mg, yield: 88.1%). MS(ESI): m/z 453.2(M+H) + .
  • TGF ⁇ RI kinase was purchased from Carna (Cat#09-141). Add 2 ⁇ l of enzyme solution prepared with kinase buffer (40mM Tris PH 7.5, 20mM MgCl 2 , 1mM DTT, 1mg/ml BSA) into a 384-well plate (Greiner, Cat#784075) (TGF ⁇ RI kinase is 25nM in the final reaction system) ). For the negative control, 2 ⁇ l kinase buffer was added. The compound was prepared as a 10 mM stock solution with DMSO, and the compound was diluted twice with DMSO according to the requirement of the initial concentration before the compound was detected.
  • p38 ⁇ kinase was purchased from SignalChem (Cat#M39-10BG). Add 2 ⁇ l of enzyme solution (p38 ⁇ kinase in the final reaction system) prepared with kinase buffer (40mM Tris PH 7.5, 20mM MgCl 2 , 0.05mM DTT, 0.1mg/ml BSA) into a 384-well plate (Greiner, Cat#784075) 0.8ng/ul). For the negative control, 2 ⁇ l kinase buffer was added. The compound was prepared as a 10 mM stock solution with DMSO, and the compound was diluted twice with DMSO according to the requirement of the initial concentration before the compound was detected.
  • HEK-Blue TGF ⁇ cells (Invivogen, Cat#hkb-tgfb), adjust the cell density to 1.25 ⁇ 10 6 cells/ml with 10% FBS DMEM, and add 40 ⁇ l per well to a 96-well plate.
  • the compound was prepared as a 10 mM stock solution with DMSO, and the compound was diluted twice with DMSO according to the requirement of the initial concentration before the compound was detected. Add 2.5 ⁇ l of the compound to 1 ml of complete medium, mix by pipetting, and dilute the compound 3-fold with DMEM medium containing 0.25% DMSO (the DMSO concentration in a 100 ⁇ l system is 0.1%).
  • SEAP detection uses Great EscApe SEAP chemiluminescence KIT (Clontech, Cat#631738) for detection, and cell viability detection uses CellTiter-Glo Luminescent Cell Viability Assay (Promega, Cat#G7573) for detection.
  • CellTiter-Glo Luminescent Cell Viability Assay Promega, Cat#G7573
  • Use Molecular Devices, SpectraMax i3x. to read the final chemiluminescence signal.
  • XLFIT data obtained using the 4-parameter curve fitting is performed aggregated IC 50.

Abstract

式(I)化合物及其药物组合物。式(I)化合物可用作TGF-β受体抑制剂,尤其是TGFβRI抑制剂,可以用于预防或治疗例如各种TGFβRI(ALK5)介导的相关疾病。

Description

转化生长因子-β受体抑制剂
本申请要求于2020年03月24日提交中国专利局、申请号为202010212752.X、发明名称为“转化生长因子-β受体抑制剂”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。
技术领域
本发明提供了一类新型杂环化合物,其制备方法及其作为TGF-β受体(特别是TGFβRI)拮抗剂的应用。
背景技术
转化生长因子-β(transforming growth factor-β,TGF-β)是一种多功能的生长因子和细胞因子,与活化素(activins)、抑制素(inhibins)、缪勒氏管抑制质(Mullerian inhibitor substance,MIS)和骨形成蛋白(bone morpho-genetic proteins,BMPs)等多种相关蛋白组成转化生长因子-β超家族。它能够参与调节多种生物过程,包括细胞的增殖、分化、发育以及细胞外基质的修饰(包括肿瘤基质和免疫抑制、血管生成和纤维组织生成)。
TGF-β主要有三种细胞受体,分别是I型(TGFβRI)、II型(TGFβRII)和III型(TGFβRIII)。TGF-β信号通路中的信号传导主要是通过TGF-β配体与细胞表面的丝氨酸/苏氨酸激酶受体TGFβRI(ALK5)和TGFβRII结合形成一个异源四聚体复合物,然后TGFβRII对TGFβRI的甘氨酸/丝氨酸区域(GS region)进行磷酸化并激活TGFβRI磷酸化与其连接的细胞内信号传导蛋白Smad蛋白分子——Smad2/Smad3。磷酸化的Smad2和Smad3蛋白与Smad4结合形成的复合物转移到细胞核,结合不同的转录因子和转录共激活剂或共抑制剂,来调节靶基因的转录,引起各种各样的转录反应,从而导致基因表达的改变。而转化生长因子-βIII型受体(TGFβRIII)则本身不能传递信号,但它能通过结合TGF-β并将其传递给TGFβRII来增强TGF-β配体与TGF-βII型受体的结合,从而间接的影响信号的传导。在生物体内,细胞因子、生长因子、微环境条件、激素、磷酸化和去磷酸化激酶等都能影响和控制TGF-β信号通路中的信号传导。
研究表明,TGF-β在心、肝、肺、肾等不同器官***纤维化的发生和进展中起着关键作用。异常的TGF-β信号和许多人类疾病有关,比如癌症、心血管疾病、炎症、器官纤维化疾病、胰腺疾病等。在人的肿瘤中,TGF-β信号的失调很常见,能促进肿瘤细胞生长和分化、调节细胞外基质和上皮间质转换。目前,随着肿瘤免疫疗法研究的不断深入,TGF-β已经被证实对调节抗肿瘤免疫 有重要作用。TGF-β对T淋巴细胞的分化有很强的抑制作用,对树状细胞、CD8 +T细胞和NK细胞有明显的负影响,同时它能增强免疫抑制T reg和骨髓源性抑制细胞(MDSC)的活性,从而为肿瘤生长和转移提供有利的肿瘤微环境。因此,抑制TGF-β信号通路中某个环节的信号传导有可能成为***的有效方法之一。
TGFβRI(ALK5)作为TGF-β信号通路中的一个重要节点被认为是***的一个重要靶点,通过阻止TGFβRI与配体的结合,来抑制ALK5对其下游信号蛋白(Smad2或Smad3)的磷酸化,从而影响或者阻断TGF-β信号的传导,以实现预防和治疗各种由ALK5介导的相关疾病。
目前,国际专利申请公开号WO2000/061576、WO2002/066462、WO2004/111036、WO2004/048382、WO2009/087224、WO2009/013335、WO2012/002680、WO2016/057278、WO2017/015425、WO2017/215506、WO2018/017633等和中国专利申请公开号CN107663206A等公开了用作TGF-β受体拮抗剂的化合物。然而,仍需要对TGF-β受体,尤其是对TGFβRI(ALK5)具有更好抑制效果的抑制剂;特别地,能够选择性抑制ALK5的拮抗剂具有重要的临床价值和治疗意义,但是目前对其报道很少。现有技术中迫切需要具有更好抑制效果的ALK5抑制剂,特别是能够选择性抑制ALK5的新型拮抗剂化合物。
发明内容
经过长期而深入的研究,我们意外地发现了一类具有良好的ALK5抑制活性的杂环化合物。
基于上述发现,在本发明的第一个方面,提供了式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物,
Figure PCTCN2021081965-appb-000001
其中:
Cy表示被0、1、2或3个取代基所取代的C 6-C 10芳基或者5-10元杂芳基,其中所述的取代基选自:卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
R 1表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基;
R 2表示氢、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-SO 2NR aR b、-NR aSO 2R b
R 3表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-SO 2NR aR b、-NR aSO 2R b
R 4表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、羟基(C 1-C 6烷基);R 5表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基;或者R 4、R 5连同其共同相连的原子一起形成5-8元饱和和不饱和环,该环中还可以任意地含有0、1或2个选自N、O、S的杂原子;
其中,R a、R b分别各自独立地表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基;或者R a、R b与其所连接的原子一起形成5-8元环,该环中还可以任意地含有0、1或2个选自N、O、S的杂原子;
其中n表示0、1、2或3。
优选地,所述式(I)化合物具有以下式(II)结构:
Figure PCTCN2021081965-appb-000002
其中:
R 1、R 3、R 4、R 5、R a、R b和n具有如权利要求1所述的定义;
R 2表示氢、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基;
W表示CR L或者N;
R 6表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
R L表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
o表示0、1、2或3。
优选地,本发明的化合物选自以下结构:
Figure PCTCN2021081965-appb-000003
本发明的化合物也可制备成可药用盐的形式,所述可药用盐可以使用例如以下的无机酸或有机酸而形成:盐酸、氢溴酸、硫酸、磷酸、硝酸、乙酸、乙 醇酸、乳酸、丙酮酸、丙二酸、琥珀酸、戊二酸、富马酸、苹果酸、扁桃酸、酒石酸、柠檬酸、抗坏血酸、棕榈酸、马来酸、羟基马来酸、苯甲酸、羟基苯甲酸、苯乙酸、肉桂酸、水杨酸、甲磺酸、苯磺酸或甲苯磺酸。
本发明的可药用盐可通过常规方法制备,例如通过将本发明的化合物溶解于与水可混溶的有机溶剂(例如丙酮、甲醇、乙醇和乙腈)中,向其中添加过量的有机酸或无机酸水溶液,以使得盐从所得混合物中沉淀,从中除去溶剂和剩余的游离酸,然后分离所沉淀的盐即可。
本发明化合物(或其可药用盐)可包括溶剂化物形式,优选地,所述溶剂化物为水合物。
本发明还提供了本发明的化合物在制备用于预防或治疗可通过抑制ALK5活性来调节之疾病的药物中的用途。优选地,所述疾病选自癌症、肿瘤、炎症性疾病、自身免疫性疾病和免疫介导性疾病。
此外,本发明提供了用于预防或治疗癌症、肿瘤、炎症性疾病、自身免疫性疾病、神经退行性疾病、注意力相关疾病或免疫介导性疾病的药物组合物,其包含本发明的式(I)化合物作为活性成分。
此外,本发明提供了一种用于预防或治疗癌症、肿瘤、炎症性疾病、自身免疫性疾病、神经退行性疾病、注意力相关疾病或免疫介导性疾病的方法,其包括向有此需要的哺乳动物施用本发明的化合物。
癌症或肿瘤的代表性实例可包括但不限于,皮肤癌、膀胱癌、卵巢癌、乳腺癌、胃癌、胰腺癌、***癌、结肠癌、肺癌、骨癌、脑癌、神经细胞瘤、直肠癌、结肠癌、家族性腺瘤性息肉性癌、遗传性非息肉性结直肠癌、食管癌、唇癌、喉癌、下咽癌、舌癌、唾液腺癌、胃癌、腺癌、甲状腺髓样癌、***状甲状腺癌、肾癌、肾实质癌、卵巢癌、***、子宫体癌、子宫内膜癌、绒毛膜癌、睾丸癌、泌尿癌、黑素瘤、脑肿瘤诸如成胶质细胞瘤、星形细胞瘤、脑膜瘤、成神经管细胞瘤和外周神经外胚层肿瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、伯基特淋巴瘤、急性淋巴性白血病(ALL)、慢性淋巴性白血病(CLL)、急性骨髓性白血病(AML)、慢性粒细胞白血病(CML)、成人T细胞白血病淋巴瘤、弥漫性大B细胞淋巴瘤(DLBCL)、肝细胞癌、胆囊癌、支气管癌、小细胞肺癌、非小细胞肺癌、多发性骨髓瘤、基底细胞瘤、畸胎瘤、成视网膜细胞瘤、脉络膜黑素瘤、***瘤、横纹肌肉瘤、颅咽管瘤、骨肉瘤、软骨肉瘤、肌肉瘤、脂肪肉瘤、纤维肉瘤、尤因肉瘤或浆细胞瘤。
当将本发明化合物或其可药用盐与另外的用于治疗癌症或肿瘤的抗癌剂或检查点抑制剂组合施用时,本发明化合物或其可药用盐可提供增强的抗癌作用。
用于治疗癌症或肿瘤的抗癌剂的代表性实例可包括但不限于细胞信号转导抑制剂、苯丁酸氮芥、美法仑、环磷酰胺、异环磷酰胺、白消安、卡莫司汀、洛莫司汀、链脲佐菌素、顺铂、卡铂、奥沙利铂、达卡巴嗪、替莫唑胺、丙卡巴肼、甲氨蝶呤、氟尿嘧啶、阿糖胞苷、吉西他滨、巯基嘌呤、氟达拉滨、长春碱、长春新碱、长春瑞滨、紫杉醇、多西紫杉醇、拓扑替康、伊立替康、依托泊苷、曲贝替定、更生霉素、多柔比星、表柔比星、道诺霉素、米托蒽醌、博来霉素、丝裂霉素C、伊沙匹隆、他莫昔芬、氟他胺、戈那瑞林类似物、甲地孕酮、强的松、***、甲泼尼龙、沙利度胺、干扰素α、亚叶酸钙、西罗莫司、西罗莫司脂化物、依维莫司、阿法替尼、alisertib、amuvatinib、阿帕替尼、阿西替尼、硼替佐米、波舒替尼、布立尼布、卡博替尼、西地尼布、crenolanib、克卓替尼、达拉菲尼、达可替尼、达努塞替、达沙替尼、多维替尼、厄洛替尼、foretinib、ganetespib、吉非替尼、依鲁替尼、埃克替尼、伊马替尼、iniparib、拉帕替尼、lenvatinib、linifanib、linsitinib、马赛替尼、momelotinib、莫替沙尼、来那替尼、尼罗替尼、niraparib、oprozomib、olaparib、帕唑帕尼、pictilisib、普纳替尼、quizartinib、瑞格菲尼、rigosertib、rucaparib、鲁索利替尼、塞卡替尼、saridegib、索拉非尼、舒尼替尼、替拉替尼、tivantinib、替沃扎尼、托法替尼、曲美替尼、凡德他尼、维利帕尼、威罗菲尼、维莫德吉、volasertib、阿仑单抗、贝伐单抗、贝伦妥单抗维多汀、卡妥索单抗、西妥昔单抗、地诺单抗、吉妥珠单抗、伊匹单抗、尼妥珠单抗、奥法木单抗、帕尼单抗、利妥昔单抗、托西莫单抗、曲妥珠单抗;检查点抑制剂,包括但不局限于抗PD-1抗体、抗PD-L1抗体、LAG3抗体、TIM-3抗体以及抗CTLA-4抗体或它们的任意组合。
炎症性疾病、自身免疫性疾病和免疫介导性疾病的代表性实例可包括但不限于,关节炎、类风湿性关节炎、脊柱关节炎、痛风性关节炎、骨关节炎、幼年型关节炎、其他关节炎性病症、狼疮、***性红斑狼疮(SLE)、皮肤相关疾病、银屑病、湿疹、皮炎、过敏性皮肤炎、疼痛、肺病、肺部炎症、成人呼吸窘迫综合征(ARDS)、肺结节病、慢性肺部炎症性疾病、慢性阻塞性肺病(COPD)、心血管疾病、动脉粥样硬化、心肌梗塞、充血性心力衰竭、心肌缺血再灌注损伤、炎性肠病、克罗恩病、溃疡性结肠炎、肠易激综合征、哮喘、干燥综合征、自身免疫甲状腺疾病、荨麻疹(风疹)、多发性硬化、硬皮症、器官移植排斥、异种移植、特发性血小板减少性紫癜(ITP)、帕金森病、阿尔兹海默病、糖尿病相关疾病、炎症、***性疾病、过敏性鼻炎、过敏性支气管炎、过敏性鼻窦炎、白血病、淋巴瘤、B细胞淋巴瘤、T细胞淋巴瘤、骨髓瘤、急性淋巴性白血病(ALL)、慢性淋巴性白血病(CLL)、急性髓性白血病(AML)、慢性髓性白血病(CML)、毛细胞白血病、何杰金氏病、非何杰金淋巴瘤、多发性骨髓瘤、 骨髓增生异常综合征(MDS)、骨髓增生性肿瘤(MPN)、弥漫性大B细胞淋巴瘤和滤泡性淋巴瘤。
当将本发明化合物或其可药用盐与另外的用于治疗炎症性疾病、自身免疫性疾病和免疫介导性疾病的治疗剂组合施用时,本发明化合物或其可药用盐可提供增强的治疗作用。
用于治疗炎症性疾病、自身免疫性疾病和免疫介导性疾病的治疗剂的代表性实例可包括但不限于,甾体药物(例如,强的松、氢化波尼松、甲基氢化波尼松、可的松、羟基可的松、倍他米松、***等)、甲氨蝶呤、来氟米特、抗TNFα剂(例如,依那西普、英夫利昔单抗、阿达利单抗等)、钙调神经磷酸酶抑制剂(例如,他克莫司、吡美莫司等)和抗组胺药(例如,苯海拉明、羟嗪、氯雷他定、依巴斯汀、酮替芬、西替利嗪、左西替利嗪、非索非那定等),并且选自其中的至少一种治疗剂可包含于本发明药物组合物中。
本发明的化合物或其可药用盐可作为活性成分通过口服或肠胃外施用,其有效量的范围为在哺乳动物包括人(体重约70kg)的情况下0.1至2,000mg/kg体重/天、优选1至1,000mg/kg体重/天,并且每天以单次或4次分次剂量,或者遵循/不遵循预定时间施用。活性成分的剂量可根据多个相关因素(例如待治疗对象的情况、疾病类型和严重性、施用速率和医生意见)进行调整。在某些情况下,小于以上剂量的量可能是合适的。如果不引起有害的副作用则可使用大于以上剂量的量并且该量可以每天以分次剂量施用。
可根据常规方法中的任何一种将本发明药物组合物配制成用于口服施用或肠胃外施用(包括肌内、静脉内和皮下途径)的剂型,例如片剂、颗粒、粉末、胶囊、糖浆、乳剂、微乳剂、溶液或混悬液。
用于口服施用的本发明药物组合物可通过将活性成分与例如以下的载体混合来制备:纤维素、硅酸钙、玉米淀粉、乳糖、蔗糖、右旋糖、磷酸钙、硬脂酸、硬脂酸镁、硬脂酸钙、明胶、滑石、表面活性剂、助悬剂、乳化剂和稀释剂。在本发明的注射组合物中采用的载体的实例是水、盐溶液、葡萄糖溶液、葡萄糖样溶液、醇、二醇、醚(例如,聚乙二醇400)、油、脂肪酸、脂肪酸酯、甘油酯、表面活性剂、助悬剂和乳化剂。
本发明描述示例性实施方案的过程中,本发明的其它特征将变得显而易见,给出所述实施方案用于说明本发明而不意欲成为其限制,以下实施例使用本发明所公开的方法制备、分离和表征。
可以用有机合成领域的技术人员已知的多种方式来制备本发明的化合物,可使用有机合成化学领域中已知的合成方法或通过本领域技术人员所了解的其 变化形式来合成本发明化合物。可以在适用于所使用试剂盒材料和适用于所实现转变的溶剂或溶剂混合物中实施所需反应。
具体实施方式
本发明人经过长期而深入的研究,发现了一类具有TGF-β受体抑制活性、尤其是ALK5抑制活性的如式(I)所示的杂环化合物。基于上述发现,发明人完成了本发明。
术语
如果无另外说明,用于本发明申请,包括说明书和权利要求书中的术语,定义如下。必须注意,在说明书和所附的权利要求书中,如果文中无另外清楚指示,无数词修饰的名词通常包括其复数意义。如果无另外说明,使用质谱、核磁、HPLC、蛋白化学、生物化学、重组DNA技术和药理的常规方法。
在说明书和权利要求书中,给定化学式或名称应涵盖所有立体和光学异构体及其中存在上述异构体的外消旋物。除非另外指明,否则所有手性(对映异构体和非对映异构体)和外消旋形式均在本发明范围内。所述化合物中还可存在C=C双键、C=N双键、环***等的许多几何异构体,且所有上述稳定异构体均涵盖于本发明内。本发明描述了本发明化合物的顺式-和反式-(或E-和Z-)几何异构体,且其可分离成异构体的混合物或分开的异构体形式。本发明化合物可以光学活性或外消旋形式加以分离。用于制备本发明化合物和其中制备的中间体的所有方法均视为本发明的部分。在制备对映异构体或非对映异构体产物时,其可通过常规方法(例如通过色谱或分段结晶)进行分离。取决于方法条件,以游离(中性)或盐形式获得本发明的终产物。这些终产物的游离形式和盐均在本发明的范围内。如果需要的话,则可将化合物的一种形式转化成另一种形式。可将游离碱或酸转化成盐;可将盐转化成游离化合物或另一种盐;可将本发明异构体化合物的混合物分离成单独的异构体。本发明化合物、其游离形式和盐可以多种互变异构体形式存在,其中氢原子转置到分子的其它部分上且由此分子的原子之间的化学键发生重排。应当理解的是,可存在的所有互变异构体形式均包括在本发明内。
除非另有定义,否则当取代基被标注为“任选取代的”时,所述取代基选自例如以下取代基,诸如烷基、环烷基、芳基、杂环基、卤素、羟基、烷氧基、氧 代、烷酰基、芳基氧基、烷酰基氧基、氨基、烷基氨基、芳基氨基、芳基烷基氨基、二取代的胺基团(其中2个氨基取代基选自烷基、芳基或芳基烷基)、烷酰基氨基、芳酰基氨基、芳烷酰基氨基、取代的烷酰基氨基、取代的芳基氨基、取代的芳烷酰基氨基、硫基、烷基硫基、芳基硫基、芳基烷基硫基、芳基硫羰基、芳基烷基硫羰基、烷基磺酰基、芳基磺酰基、芳基烷基磺酰基、磺酰氨基例如-SO 2NH 2、取代的磺酰氨基、硝基、氰基、羧基、氨基甲酰基例如-CONH 2、取代的氨基甲酰基例如-CONH烷基、-CONH芳基、-CONH芳基烷基或在氮上具有两个选自烷基、芳基或芳基烷基的取代基的情况、烷氧基羰基、芳基、取代的芳基、胍基、杂环基,例如吲哚基、咪唑基、呋喃基、噻吩基、噻唑基、吡咯烷基、吡啶基、嘧啶基、吡咯烷基、哌啶基、吗啉基、哌嗪基、高哌嗪基等和取代的杂环基。
本文使用的术语“烷基”或“亚烷基”意欲包括具有指定碳原子数的支链或直链饱和脂族烃基团。例如,“C 1-C 6烷基”表示具有1个至6个碳原子的烷基。烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(例如正丙基和异丙基)、丁基(例如正丁基、异丁基、叔丁基)和戊基(例如正戊基、异戊基、新戊基)。优选的烷基是C 1-C 6烷基,更优选的烷基是C 1-C 4烷基。
术语“烯基”表示含一个或多个双键且通常长度为2至20个碳原子的直链或支链的烃基。例如,“C 2-C 8烯基”含有两个至八个碳原子。烯基包括但不限于例如乙烯基、丙烯基、丁烯基、1-甲基-2-丁烯-1-基、庚烯基、辛烯基等。优选的烯基是C 2-C 8烯基,更优选的烯基是C 2-C 6烯基。
术语“炔基”表示含一个或多个叁键且通常长度为2至20个碳原子的直链或支链的烃基。例如,“C 2-C 8炔基”含有两个至八个碳原子。优选的炔基是C 2-C 8炔基,更优选的烯基是C 2-C 6炔基。代表性炔基包括但不限于例如乙炔基、1-丙炔基、1-丁炔基、庚炔基、辛炔基等。
术语“烷氧基”或“烷基氧基”是指-O-烷基。“C 1-C 6烷氧基”(或烷基氧基)意欲包括C 1、C 2、C 3、C 4、C 5和C 6烷氧基。优选的烷氧基是C 1-C 6烷氧基,更优选的烷氧基是C 1-C 4烷氧基。烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(例如正丙氧基和异丙氧基)和叔丁氧基。类似地,“烷基硫基”或“硫代烷氧基”表示具有指定数量碳原子的经硫桥连接的如上文所定义的烷基;例如甲基-S-和乙基-S-。
术语“羰基”是指由碳和氧两种原子通过双键连接而成的有机官能团(C=O)。
术语“芳基”,单独或作为较大部分诸如“芳烷基”、“芳烷氧基”或“芳基氧基 烷基”的部分,是指具有总计5至12个环成员的单环、二环或三环的环***,其中所述***中的至少一个环为芳族的且其中所述***中的每个环含有3至7个环成员。单环的芳香基指的是苯基,二环及二环以上的芳香基指萘基、蒽基等,同时此芳基二环也可为苯环融合了一个环烷基、或融合一个环烯基、或融合一个环炔基。优选的芳基是C 6-C 12芳基。在本发明的某些实施方案中,“芳基”是指芳族环***,其包括但不限于苯基、联苯基、茚满基、1-萘基、2-萘基和四氢萘基。术语“芳烷基”或“芳基烷基”是指连接至芳基环的烷基残基。非限制性实例包括苄基、苯乙基等。稠合的芳基可在环烷基环或芳族环的合适位置上连接至另一基团。例从环***中画出的箭头线表明键可连接至任意合适的环原子。
术语“环烷基”是指单环或二环的环状烷基。单环的环状烷基指C 3-C 8的环状烷基,包括但不限于环丙基、环丁基、环戊基、环己基和降莰烷基。支化环烷基诸如1-甲基环丙基和2-甲基环丙基包括在“环烷基”的定义中。二环的环状烷基包括桥环、螺环或融合环的环烷基。优选的环烷基是C 3-C 6环烷基。
术语“环烯基”是指单环或二环的环状烯基。单环的环状烯基指C 3-C 8的环状烯基,包括但不限于环丙烯基、环丁烯基、环戊烯基、环己烯基和降莰烯基。支化环烯基诸如1-甲基环丙烯基和2-甲基环丙烯基包括在“环烯基”的定义中。二环的环状烯基包括桥环、螺环或融合环的环状烯基。
术语“羟基烷基”是指具有指定碳原子数的支链和直链饱和脂族烃基团且烃基的碳原子上至少连接一个-OH。羟基烷基的实例包括但不限于羟甲基、羟乙基、1-羟基丙基、2-羟基丙基。
“卤代”或“卤素”包括氟、氯、溴和碘。“卤代烷基”意欲包括具有指定碳原子数且取代有1个或多个卤素的支链和直链饱和脂族烃基团。卤代烷基的实例包括但不限于氟甲基、二氟甲基、三氟甲基、三氯甲基、五氟乙基、五氯乙基、2,2,2-三氟乙基、七氟丙基和七氯丙基。卤代烷基的实例还包括意欲包括具有指定碳原子数且取代有1个或多个氟原子的支链和直链饱和脂族烃基团的“氟烷基”。
“卤代烷氧基”或“卤代烷基氧基”表示具有指定数量碳原子的经氧桥连接的如上文所定义的卤代烷基。例如,“C 1-C 6卤代烷氧基”意欲包括C 1、C 2、C 3、C 4、C 5和C 6卤代烷氧基。卤代烷氧基的实例包括但不限于三氟甲氧基、2,2,2-三氟乙氧基和五氟乙氧基。类似地,“卤代烷基硫基”或“硫代卤代烷氧基”表示具有指定数量碳原子的经硫桥连接的如上文所定义的卤代烷基;例如三氟甲基-S-和五氟乙基-S-。
术语“杂芳基”意指稳定的3元、4元、5元、6元、或7元芳香单环或芳香二环或7元、8元、9元、10元、11元、12元、13元或14元芳香多环杂环,其为完全不饱和的、部分不饱和的,且其含有碳原子和1个、2个、3个或4个独立地选自N、O和S的杂原子;且包括任何以下多环基团,其中上文所定义的任意杂环与苯环稠合。氮和硫杂原子可任选地被氧化。氮原子为取代的或未取代的(即N或NR,其中R为H或如果被定义,则为另一取代基)。杂环可在得到稳定结构的任何杂原子或碳原子处连接至其侧基。如果所得化合物是稳定的,则本文所述的杂环基可在碳或氮原子上被取代。杂环中的氮可任选地被季铵化。优选地,当杂环中S和O原子的总数超过1时,则这些杂原子彼此不相邻。优选地,杂环中S和O原子的总数不大于1。优选的杂芳基是5-12元杂芳基。杂芳基的实例包括但不限于吖啶基、氮杂环丁基、吖辛因基、苯并咪唑基、苯并呋喃基、苯并硫代呋喃基、苯并噻吩基、苯并噁唑基、苯并噁唑啉基、苯并噻唑基、苯并***基、苯并四唑基、苯并异噁唑基、苯并异噻唑基、苯并咪唑啉基、咔唑基、4aH-咔唑基、咔啉基、色满基、色烯基、噌啉基、十氢喹啉基、2H,6H-1,5,2-二噻嗪基、二氢呋喃并[2,3-b]四氢呋喃基、呋喃基、呋咱基、咪唑烷基、咪唑啉基、咪唑基、1H-吲唑基、咪唑并吡啶基、假吲哚基(indolenyl)、二氢吲哚基、吲嗪基、吲哚基、3H-吲哚基、靛红酰基(isatinoyl)、异苯并呋喃基、异色满基、异吲唑基、异二氢吲哚基、异吲哚基、异喹啉基、异噻唑基、异噻唑并吡啶基、异噁唑基、异噁唑并吡啶基、亚甲基二氧基苯基、吗啉基、二氮杂萘基、八氢异喹啉基、噁二唑基、1,2,3-噁二唑基、1,2,4-噁二唑基、1,2,5-噁二唑基、1,3,4-噁二唑基、噁唑烷基、噁唑基、噁唑并吡啶基、噁唑烷基、萘嵌间二氮杂苯基、羟吲哚基、嘧啶基、菲啶基、菲咯啉基、吩嗪基、吩噻嗪基、吩噁噻基、吩噁嗪基、酞嗪基、哌嗪基、哌啶基、哌啶酮基、4-哌啶酮基、胡椒基、喋啶基、嘌呤基、吡喃基、吡嗪基、吡唑烷基、吡唑啉基、吡唑并吡啶基、吡唑基、哒嗪基、吡啶并噁唑基、吡啶并咪唑基、吡啶并噻唑基、吡啶基、嘧啶基、吡咯烷基、吡咯啉基、2-吡咯烷酮基、2H-吡咯基、吡咯基、喹唑啉基、喹啉基、4H-喹嗪基、喹喔啉基、奎宁环基、四唑基、四氢呋喃基、四氢异喹啉基、四氢喹啉基、6H-1,2,5-噻二嗪基、1,2,3-噻二唑基、1,2,4-噻二唑基、1,2,5-噻二唑基、1,3,4-噻二唑基、噻蒽基、噻唑基、噻吩基、噻唑并吡啶基、噻吩并噻唑基、噻吩并噁唑基、噻吩并咪唑基、噻吩基、三嗪基、1,2,3-***基、1,2,4-***基、1,2,5-***基、1,3,4-***基和呫吨基、喹啉基、异喹啉基、酞嗪基、喹唑啉基、吲哚基、异吲哚基、二氢吲哚基、1H-吲唑基、苯并咪唑基、1,2,3,4-四氢喹啉基、1,2,3,4-四氢异喹啉基、5,6,7,8-四氢-喹啉基、2,3-二氢-苯并呋喃基、色满基、1,2,3,4-四氢-喹喔啉基和1,2,3,4-四氢-喹唑啉基。本发明还包括含有例 如上述杂环的稠环和螺环化合物。
本文使用的术语“杂环烷基”指的是一个单环杂环烷基体系,或为一个二环杂环烷基体系,同时还包括螺杂环或桥杂环烷基。单环的杂环烷基指的是3-8元、且至少含一个选自O、N、S、P的饱和或不饱和但不为芳香性的环状烷基体系。二环杂环烷基体系指的是一个杂环烷基融合到一个苯基、或一个环烷基、或一个环烯基、或一个杂环烷基、或一个杂芳基。优选的杂环烷基是3-12元杂环烷基。
本文中所用的术语“取代”意指至少一个氢原子被非氢基团替代,条件是维持正常化合价且所述取代得到稳定的化合物。本文所用的环双键为在两个相邻环原子之间形成的双键(例如C=C、C=N或N=N)。
在本发明化合物上存在氮原子(例如胺)的情形下,可通过使用氧化剂(例如m-CPBA和/或过氧化氢)进行处理来将这些氮原子转化成N-氧化物以获得本发明的其它化合物。因此,所显示和要求保护的氮原子视为均涵盖所显示氮及其N-氧化物以获得本发明衍生物。
当任何变量在化合物的任何组成或式中出现一次以上时,其每次出现时的定义均独立于其在其它每种情况下出现时的定义。因此,例如如果显示基团取代有0、1、2或3个R,则所述基团可任选地取代有至多三个R基团,且在每次出现时R独立地选自R的定义。此外,取代基和/或变量的组合仅在上述组合可产生稳定的化合物时才容许存在。
术语“溶剂化物”意指本发明化合物与一个或多个溶剂分子(无论有机的还是无机的)的物理缔合。该物理缔合包括氢键。在某些情形中,例如当一个或多个溶剂分子纳入结晶固体的晶格中时,溶剂化物将能够被分离。溶剂化物中的溶剂分子可按规则排列和/或无序排列存在。溶剂化物可包含化学计量或非化学计量的溶剂分子。“溶剂合物”涵盖溶液相和可分离的溶剂化物。示例性溶剂化物包括但不限于水合物、乙醇合物、甲醇合物和异丙醇合物。溶剂化方法是本领域公知的。
本文使用的术语“患者”是指通过本发明的方法进行治疗的有机体。这类有机体优选包括但不限于哺乳动物(例如鼠类、猿/猴、马、牛、猪、犬、猫等)且最优选是指人类。
本文使用的术语“有效量”意指将会引起例如研究人员或临床医师所寻求的组织、***、动物或人的生物学或医学响应的药物或药剂(即本发明化合物)的量。此外,术语“治疗有效量”意指这样的量:与未接受上述量的相应受试者相比,所 述量导致改善的治疗、治愈、预防或减轻疾病、病症或副作用,或降低在疾病或病症的进展速度。有效量可以一个或多个给药、施用或剂量给予且不意欲被特定的制剂或给药途径限制。该术语还包括在其范围内的增强正常生理机能的有效量。
本文使用的术语“治疗”包括导致改善病症、疾病、障碍等的任何效果,例如减轻、减少、调节、改善或消除,或改善其症状。如本文所用,某一化合物或药物组合物,在给药后可以使某一疾病、症状或情况得到改善,尤指其严重度得到改善,延迟发病,减缓病情进展,或减少病情持续时间。无论固定给药或临时给药、持续给药或断续给药,可以归因于或与给药有关的情况。本文使用的术语“预防”是指在疾病或症状发生之前防止、阻断、消除疾病发生或者干扰或减缓病程发展。
本文使用的术语“药物组合物”是指活性剂与惰性或活性的载体的组合,使得所述组合物尤其适用于体内或离体诊断或治疗。碱的实例包括但不限于碱金属(例如钠)氢氧化物、碱土金属(例如镁)氢氧化物、氨等。对于治疗用途,本发明化合物的盐对于治疗用途,本发明化合物的盐预期为是药用的。然而,非药用的酸和碱的盐也可用于例如药用化合物的制备或纯化中。
术语“药用”在本文中用于指如下那些化合物、物质、组合物和/或剂型:在合理医学判断的范围内,其适于与人类和动物的组织接触使用而无过高毒性、刺激性、过敏反应和/或其它问题或并发症,并与合理的益处/风险比相称。
本文使用的短语“药学上可接受的载体”、“可药用载体”意指药用物质、组合物或媒介物,诸如液体或固体填充剂、稀释剂、赋形剂、制造助剂(例如润滑剂、滑石、硬脂酸镁、硬脂酸钙或硬脂酸锌或硬脂酸)或溶剂包囊物质,其涉及将活性化合物从一个器官或身体的部分携带或运送至另一个器官或身体的部分。每种载体在与制剂的其它成分相容和对患者无害的意义上必须是“可接受的”。
特定药学及医学术语
术语“可接受的”,如本文所用,指一个处方组分或活性成分对一般治疗目标的健康没有过分的有害影响。
术语“癌症”,如本文所用,指一种不能控制的细胞的异常生长,并且在某种条件下能够转移(传播)。这种类型的癌症包括但不限于,实体肿瘤(如膀胱、肠、脑、胸、子宫、心脏、肾、肺、淋巴组织(淋巴瘤)、卵巢、胰腺或其它内分泌器官(如甲状腺)、***、皮肤(黑色素瘤)或血液瘤(如非白血性白血病)。
术语“联合给药”或其类似术语,如本文所用,指将几种所选的治疗药物给一个病人用药,以相同或不同的给药方式在相同或不同的时间给药。
术语“增强”或“能增强”,如本文所用,指预期的结果能够在效价或是持续时间方面都有增加或延长。因此,在增强药物的治疗效果方面,术语“能增强”指药物在***中有提高或延长效价或持续时间的能力。本文所用的“增效值”,指在理想的***中,能够最大限度地的增强另外一个治疗药物的能力。
术语“免疫性疾病”指对内源性或外源性抗原产生的不良或有害反应的疾病或症状。结果通常会造成细胞的功能障碍、或因此而破坏并造成机能障碍、或破坏可能产生免疫症状的器官或组织。
术语“试剂盒”与“产品包装”是同义词。
术语“受试者”或“对象”包括哺乳动物和非哺乳动物。哺乳动物包括但不限于,哺乳类:人、非人灵长类如猩猩、猿及猴类;农业动物如牛、马、山羊、绵羊、猪;家畜如兔、狗;实验动物包括啮齿类,如大鼠、小鼠及豚鼠等。非哺乳类动物包括但不限于,鸟、鱼等。在一优选例中,所选哺乳动物是人。
给药途径
适合的给药途径包括但不限于,口服、静脉注射、直肠、气雾剂、非肠道给药、眼部给药、肺部给药、经皮给药、***给药、耳道给药、鼻腔给药及局部给药。此外,仅作举例说明,肠道外给药,包括肌肉注射、皮下注射、静脉注射、髓内注射、心室注射、腹膜内注射、***内注射、及鼻内注射。
在一方面,此处描述的化合物给药方式是局部的而不是全身性的给药方式。在特定的具体实施例中,长效制剂通过植入给药(例如皮下或肌肉)或通过肌肉注射。此外,在另一具体化实施例中,药物通过靶向药物给药***来给药。例如,由器官特异性抗体包裹的脂质体。在这种具体实施例中,所述脂质体被选择性的导向特定器官并吸收。
药物组合物和剂量
本发明还提供药用组合物,其包含治疗有效量的与一种或多种药用载体(添加剂)和/或稀释剂一起配制的一种或多种本发明的化合物,和任选的一种或多种上述其它治疗剂。可通过任意合适方式给予本发明化合物以用于任意上述用途,例如口服,诸如片剂、丸剂、粉剂、颗粒剂、酏剂、酊剂、悬浮液(包括纳米悬 浮液、微悬浮液、喷雾干燥的分散液)、糖浆和乳液;经舌下;含服;经肠胃外,诸如通过皮下、静脉内、肌内或胸骨内注射或输注技术(例如以无菌可注射水性或非水性溶液或悬浮液形式);经鼻,包括向鼻膜给药,诸如通过吸入喷雾;局部,诸如以乳膏剂或软膏剂形式;或经直肠,诸如以栓剂形式。它们可单独给药,但通常使用基于所选给药途径和标准药学实践选择的药物载体给药。
根据本领域技术人员认识范围内的诸多因素来调配药用载体。这些因素包括,但不限于:所调配活性剂的类型和性质;含有活性剂的组合物所要给药的受试者;组合物的预期给药途径;及所靶向的治疗适应症。药用载体包括水性和非水性液体介质及各种固体和半固体剂型。
上述载体可包括除活性剂外的诸多不同成分和添加剂,上述其它成分出于本领域技术人员公知的各种原因包括于制剂中,例如稳定活性剂、粘合剂等。关于合适的药用载体和载体选择中所涉及的因素的描述可参见多个容易获得的来源,例如Allen L.V.Jr.et al.Remington:The Science and Practice of Pharmacy(2 Volumes),22nd Edition(2012),Pharmaceutical Press。
当然,本发明化合物的剂量方案取决于已知因素而有所变化,诸如具体药剂的药效学特性及其给药模式和途径;接受者的物种、年龄、性别、健康状况、医学病状和重量;症状的性质和程度;同时治疗的种类;治疗频率;给药途径、患者的肾和肝功能及期望效应。根据一般指导,当用于指定效应时,各活性成分的日口服剂量应为约0.001mg/天至约10-5000mg/天,优选地为约0.01mg/天至约1000mg/天,且最优选地为约0.1mg/天至约250mg/天。在恒速输注期间,静脉内最优选剂量应为约0.01mg/kg/分钟至约10mg/kg/分钟。本发明化合物可以单一日剂量给药,或可以每日两次、三次或四次的分开剂量给药总日剂量。
所述化合物通常以与根据预期给药形式(例如口服片剂、胶囊剂、酏剂和糖浆剂)适当地选择且与常规药学实践相符合的合适药物稀释剂、赋形剂或载体(在本文中统称为药物载体)的混合物形式进行给药。
适于给药的剂型(药物组合物)可含有约1毫克至约2000毫克活性成分/剂量单位。在这些医药组合物中,以组合物的总重量计,活性成分通常将以约0.1-95重量%的量存在。
用于口服给药的典型胶囊剂含有至少一种本发明化合物(250mg)、乳糖(75mg)和硬脂酸镁(15mg)。使该混合物穿过60目网筛,并包装成1号明胶胶囊。
典型的可注射制剂可如下制备:以无菌方式将至少一种本发明化合物(250mg)置于瓶中、以无菌方式冻干并密封。为进行使用,将瓶内容物与2mL生 理盐水混合,以产生可注射制剂。
本发明范围包括(单独或与药物载体组合)包含治疗有效量的至少一种本发明化合物作为活性成分的药物组合物。任选地,本发明化合物可单独使用、与本发明其它化合物组合使用或与一种或多种其它治疗剂(例如抗癌剂或其它药学活性物质)组合使用。
不考虑所选择的给药路径,通过本领域技术人员已知的常规方法来将本发的化合物(其可以合适的水合形式使用)和/或本发明的药物组合物配制成药用剂量形式。
可改变活性成分在本发明的药物组合物中的实际剂量水平,从而获得对于实现特定患者的期望的治疗响应、组成和给药模式有效的而对患者无毒的活性成分量。
选定的剂量水平会取决于多种因素,包括所用的本发明的特定化合物或其酯、盐或酰胺的活性;给药路径;给药时间;所用的特定化合物的***速率;吸收速率和程度;治疗的持续时间;与所用的特定化合物组合使用的其它药物、化合物和/或物质;所治疗的患者的年龄、性别、重量、状况、一般健康和先前的医学史等医学领域公知的因素。
具有本领域普通技术的医生或兽医可容易地确定并开出有效量的所需药物组合物。例如,为了达到所期望的治疗效果,医师或兽医可在低于所需的水平开始药物组合物中所用的本发明化合物的较量,并逐步增加剂量直至实现所期望的效果。通常,合适日剂量的本发明化合物将是有效产生治疗效果的最低剂量的化合物的量。此种有效剂量通常取决于上述因素。通常,口服、静脉内、脑室内和皮下剂量的用于患者的本发明化合物的范围为约0.01至约50mg/kg体重/天。如果需要的话,有效日剂量的活性化合物可以两个、三个、四个、五个、六个或更多个亚剂量在一天当中的适当的间隔分别给药,任选地呈单位剂型形式。在本发明的某些方面中,服药为每天一次给药。
虽然本发明化合物可单独给药,但优选以药物制剂(组合物)形式给予化合物。
试剂盒/产品包装
为了用于上述适应症的治疗,试剂盒/产品包装也在此进行描述。这些试剂盒可以由输送器、药包或容器盒组成,容器盒可被划分成多格,以容纳一种或多种容器,如管形瓶、试管及类似物等,每个容器中包含所述方法中的单独一 种成分。合适的容器包括瓶子,管形瓶,注射器和试管等。容器由可接受的玻璃或塑料等材料制作而成。
举例来讲,容器可装有一种或多种在此所述的化合物,化合物可能以药物组分形式存在,也可能与在本文中所述的其它成分组成混合物体存在。容器可有一个无菌输出口(例如容器可为静脉输液包或瓶,瓶塞可被皮下注射器针头刺破)。这样的试剂盒可带有一种化合物,及本文中所述的使用方法的说明、标签或操作说明。
一个典型的试剂盒可包括一种或多种容器,为适应商业推广和使用者对化合物使用的需求,每个容器装有一种或多种材料(如试剂,也可以是浓缩的母液,和/或器械)。这些材料包括但不局限于缓冲液,稀释液,滤器,针头,注射器,输送器,包,容器,瓶和/或试管,附有内容清单和/或使用说明书,内置包装也附有说明书。整套的说明都要包括在内。
标签可显示在容器上或与容器紧密相关。标签出现在容器上即指标签字母、数字或其它特征被粘贴、铸模、刻在容器上;标签也可出现在装有多种容器的容器盒或运输盒内,如在产品插页中。一个标签可用来提示内容物的某种特定治疗用途。标签也可标示内容物使用说明,诸如在上述方法中描述的。
在本说明书中被描述的所有特征(包括任何所述的权利要求、摘要和图),和/或任何方法或过程中涉及的所有步骤,均有可能以任意一种组合存在,除非某些特征或步骤在同一组合中是相互排斥的。
本发明提到的上述特征,或实施例提到的特征可以任意组合。本案说明书所揭示的所有特征可与任何组合物形式并用,说明书中所揭示的各个特征,可以任何可提供相同、均等或相似目的的替代性特征取代。因此除有特别说明,所揭示的特征仅为均等或相似特征的一般性例子。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件或按照制造厂商所建议的条件。除非另外说明,否则所有的百分数、比率、比例、或份数按重量计。
本发明中的重量体积百分比中的单位是本领域技术人员所熟知的,例如是指在100毫升的溶液中溶质的重量。除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。
在本发明的优选例中,提供但不局限于以下化合物:
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
具体实施例
当未包括制备途径时,相关中间体是市售的(例如来自Sigma Aldrich,Alfa)。
通用过程
使用市售试剂而不需进一步纯化。 1H-NMR谱在Bruker仪器上于500MHz记录。化学位移值以百万分率表示,即δ值。以下简写用于NMR信号的多重性:s=单峰,brs=宽峰,d=二重峰,t=三重峰,m=多重峰。耦合常数以J值列出,以Hz测量。NMR和质谱结果根据背景峰校正。色谱是指使用100筛目硅胶进行并在氮气压力(快速色谱)条件下完成的柱色谱。用于监测反应的TLC指使用特定流动相和来自Merck的硅胶F254作为固定相进行的TLC。
LC-MS实验在以下条件下测量:
仪器:Thermo U3000,ALLtech ELSD,MSQ,UV检测器结合ELSD和MSD(流出比为4:1)。柱:Waters X-Bridge C-18,3.5μm,4.6x50mm;柱温:30℃。梯度【时间(min)/溶剂B在A中(%)】:0.00/5.0,0.70/95,1.40/95,1.41/5,1.50/5。(溶剂A=0.01%三氟乙酸在水中;溶剂B=0.01%三氟乙酸在乙腈中)。UV检测:214/254/280/300nm;DAD检测:200-400nm;流速:4mL/min;MS:ESI,100-1500m/z
制备型HPLC通常使用酸性方法(乙腈和水的梯度,各含有0.1%甲酸)用Thermo U3000 AFC-3000;柱:Globalsil C-18 12nm,250x 20mm,10μm,或相当;流速:20mL/min,进行分离。
中间体的合成
化合物INT-1的制备:
Figure PCTCN2021081965-appb-000004
在回流条件下,将混有4-氟苯乙酮(2.5g,18mmol)和N,N-二甲基甲酰胺二甲基缩醛(2.4g,20mmol)搅拌4小时;反应液浓缩后,粗品用乙醇溶解(10mL),随后加入水合肼(1.1g,18mmol,80%w/w)。所得反应液在80℃条件下搅拌2小时,反应液浓缩后,所得粗品用硅胶柱层析(石油醚/乙酸乙酯=4/1~1/1)得到黄色固体INT-1a(1.84g,收率:62.7%)。MS(ESI):m/z 163.2(M+H) +.
在室温条件下,将N-溴代丁二酰亚胺(549mg,3.1mmol)加入至溶有化合物INT-1a(500mg,3.1mmol)的N,N-二甲基甲酰胺(5mL)中,所得反应液在相同温度下搅拌1.5小时。反应液倒入水(50mL)中,并用乙酸乙酯萃取(50mL×3);合并有机相用水和饱和食盐水(各100mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=4/1)得到白色固体INT-1b(553mg,收率:74.4%)。 1H NMR(500MHz,Chloroform-d)δ7.81–7.74(m,2H),7.63(s,1H),7.18–7.11(m,2H);MS(ESI):m/z 241.2(M+H) +.
在0℃条件下,将钠氢(38mg,0.95mmol,60%w/w在煤油)加入至溶有化合物INT-1c(200mg,0.83mmol)的无水四氢呋喃(8mL)溶液中,所得反应液在相同条件下搅拌20分钟;随后加入2-(三甲基硅烷基)乙氧甲基氯(207mg,1.24mmol),所得反应液在室温条件下进一步搅拌2小时。反应液加少许水淬灭后,倒入至水(50mL)中,并用乙酸乙酯萃取(50mL×3);合并有机相用水和饱和食盐水(各100mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=10/1)得到无色油状物INT-1(298mg,收率:96.7%)。
化合物INT-2的制备:
Figure PCTCN2021081965-appb-000005
将环丙甲基酮(160mg,1.9mmol)加入至混有钠氢(37mg,1.6mmol,60%w/w在煤油)的四氢呋喃(10mL)溶液中,所得反应液在室温条件下搅拌半小时后加热升至回流;随后滴加预先溶有4-氟苯乙酮(250mg,1.6mmol)的四氢呋喃(3mL)溶液。所得反应液在回流状态下进一步搅拌半小时。冷却至室温后, 将反应液倒入至1M盐酸水溶液,调节pH至7-8;并用乙酸乙酯萃取(30mL×2)。合并有机相用水和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=20/1~10/1)得到淡黄色油状物INT-2a(250mg,收率:74.8%)。MS(ESI):m/z 207.4(M+H) +
在回流条件下,将溶有化合物INT-2a(250mg,1.2mmol),水合肼(103mg,2.7mmol,80%w/w)的乙醇(5mL)溶液搅拌4小时。反应液冷却至室温后浓缩,所得粗品用二氯甲烷(30mL)溶解,并用水和饱和食盐水(各30mL)洗涤,无水硫酸钠干燥,浓缩得到白色固体INT-2b(200mg,收率:81.6%)。MS(ESI):m/z 203.5(M+H) +
在冰浴条件下,将N-碘代丁二酰亚胺(270mg,1.2mmol)加入至溶有化合物INT-2b(200mg,1.0mmol)的N,N-二甲基甲酰胺(5mL)中;所得反应液加热至90℃条件下搅拌16小时。反应液中加入二氯甲烷和饱和碳酸氢钠溶液(各30mL);有机相用水和饱和食盐水(各30mL)洗涤,无水硫酸钠干燥,浓缩得到淡黄色油状物INT-2c(300mg,收率:92.5%)。MS(ESI):m/z 329.2(M+H) +
从化合物INT-2c起,参考化合物INT-1合成中的最后一步,得到化合物INT-2。MS(ESI):m/z 459.1(M+H) +
化合物INT-3的制备:
Figure PCTCN2021081965-appb-000006
从乙酸乙酯和4-氟苯乙酮起,参考化合物INT-2a的合成得到化合物INT-3a。
从化合物INT-3a起,参考化合物INT-2的合成得到化合物INT-3。MS(ESI):m/z 433.0(M+H) +
化合物INT-4的制备:
Figure PCTCN2021081965-appb-000007
将2,4-二硝基苯基羟胺(3.1g,15.4mmol)加入至溶有4-(叔丁氧羰基氨基)吡啶(3.0g,15.4mmol)的2-甲基四氢呋喃(15mL)溶液;所得反应液在40℃条件下搅拌4小时,随后冷却至室温后搅拌过夜。反应液过滤得到黄色固体INT-4a(6.1g,收率:99.6%)。
将混有化合物INT-4a(1.0g,2.5mmol),丙炔酸乙酯(249mg,2.5mmol)和碳酸钾(703mg,5.0mmol)的N,N-二甲基甲酰胺(8mL)在室温条件下搅拌过夜。所得反应液倒入水(80mL)中,并用乙酸乙酯萃取(60mL×3)。合并有机相用水和饱和食盐水(各150mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=4/1)得到黄色固体INT-4b(378mg,收率:48.7%)。MS(ESI):m/z 306.1(M+H) +.
将溶有化合物INT-4b(378mg,1.24mmol)和三氟乙酸(1.41g,12.4mmol)的二氯甲烷(12mL)溶液在室温条件下搅拌过夜。所得反应液倒入水(30mL)中,用二氯甲烷萃取(20mL×2)。合并有机相用饱和碳酸氢钠,水和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩得到黄色固体INT-4c(254mg,收率:100%)。MS(ESI):m/z 206.5(M+H) +.
在冰浴条件下,将亚硝酸钠(128mg,1.86mmol)加入至溶有化合物INT-4c(254mg,1.24mmol)的溴化氢水溶液(5mL,48%w/w),并维持5分钟;随后在相同条件下加入溴化亚铜(355mg,2.48mmol),并进一步维持10分钟。所得反应液慢慢加热搅拌至50℃,反应20分钟。反应液倒入水(20mL)中,用乙酸乙酯萃取(30mL×2)。合并有机相用饱和碳酸氢钠,水和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=4/1)得到黄色固体INT-4(272mg,收率:81.7%)。 1H NMR(500MHz,DMSO-d 6)δ8.86(d,J=7.5Hz,1H),8.48(s,1H),8.24-8.20(m,1H),7.34(d,J=7.5Hz,1H),4.32(q,J=7.0Hz,2H),1.34(t,J=7.0Hz,3H);MS(ESI):m/z 269.3(M+H) +.
化合物INT-5的制备:
Figure PCTCN2021081965-appb-000008
将溶有化合物INT-4(100mg,0.37mmol)的硫酸水溶液(40%w/w,5mL)在100℃条件下搅拌5小时。反应液冷却至室温后倒入水(20mL)中,并用乙酸乙酯(30mL×2)萃取。合并有机相用饱和碳酸氢钠,水和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩得到黄色油状物INT-5(77mg,收率:94.6%)。MS(ESI):m/z 197.3(M+H) +.
化合物INT-6的制备:
Figure PCTCN2021081965-appb-000009
在冰浴条件下,将钠氢(72mg,1.8mmol,60%w/w在煤油)加入至溶有化合物INT-1b(300mg,1.24mmol)的四氢呋喃(8mL)溶液中,并维持20分钟;随后在相同条件下加入碘甲烷(353mg,2.5mmol)。所得反应液在搅拌中缓慢升至室温,并在室温条件下搅拌过夜。反应液用少许水淬灭,倒入水(40mL)中,并用乙酸乙酯萃取(40mL×2)。合并有机相浓缩后用制备HPLC分离得到黄色固体INT-6(149mg,收率:46.9%)。 1H NMR(500MHz,Chloroform-d)δ7.88–7.81(m,2H),7.45(s,1H),7.15–7.07(m,2H),3.92(s,3H).
化合物INT-7的制备:
Figure PCTCN2021081965-appb-000010
在0℃条件下,将溶有1H-吡唑-3-硼酸频哪酯(500mg,2.6mmol)和N,N-二异丙基乙胺(333mg,2.6mmol)的二氯甲烷(10mL)溶液搅拌半小时,随后加入2-(三甲基硅烷基)乙氧甲基氯(430mg,2.6mmol)。反应液在搅拌下缓慢升至室温,并在室温条件下继续搅拌过夜。反应液用二氯甲烷(40mL)稀释,并用饱和碳酸氢钠和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(二氯甲烷/甲醇=20/1)分离得到淡黄色油状物INT-7a(500 mg,收率:59.8%)。
在氮气气氛下,将混有化合物INT-7a(150mg,0.46mmol),2-溴-6-甲基吡啶(88mg,0.51mmol),Pd(PPh 3) 4(53mg,0.046mmol)和碳酸钾(192mg,1.38mmol)的1,4-二氧六环和水的混合溶液(3.3mL,v/v=10/1)在90℃条件下搅拌16小时。反应液冷却后用乙酸乙酯(30mL)稀释,硅藻土过滤。滤液浓缩后用硅胶柱层析(石油醚/乙酸乙酯=5/1)分离得到黄色油状物INT-7b(120mg,收率:89.6%)。MS(ESI):m/z 290.5(M+H) +
从化合物INT-7b起,参照化合物INT-1b的合成得到化合物INT-7。MS(ESI):m/z 368.3(M+H) +
化合物INT-8的制备:
Figure PCTCN2021081965-appb-000011
从化合物INT-7a和2-溴-6-三氟甲基吡啶起,参照化合物INT-7的合成得到化合物INT-8。MS(ESI):m/z 422.1(M+H) +
化合物INT-9的制备:
Figure PCTCN2021081965-appb-000012
从化合物INT-7a和2-溴-6-二氟甲基吡啶起,参照化合物INT-7的合成得到化合物INT-9。MS(ESI):m/z 404.1(M+H) +
化合物INT-10的制备:
Figure PCTCN2021081965-appb-000013
从6-三氟甲基吡啶-2-甲酸甲酯起,参照化合物INT-2的合成得到化合物 INT-10。MS(ESI):m/z 510.2(M+H) +
化合物INT-11的制备:
Figure PCTCN2021081965-appb-000014
从6-甲基吡啶-2-甲酸甲酯起,参照化合物INT-2的合成得到化合物INT-11。MS(ESI):m/z 456.1(M+H) +
化合物INT-12的制备:
Figure PCTCN2021081965-appb-000015
从4-氟苯乙酮和环戊甲基酮起,参照化合物INT-2的合成得到化合物INT-12。MS(ESI):m/z 487.0(M+H) +
化合物INT-13的制备:
Figure PCTCN2021081965-appb-000016
从化合物INT-4a和3-炔基-2-丁酮起,参照化合物INT-4的合成得到化合物INT-13。MS(ESI):m/z 239.0(M+H) +
化合物INT-14的制备:
Figure PCTCN2021081965-appb-000017
将草酸二乙酯(554mg,3.8mmol)加入至混有钠氢(74mg,3.2mmol,60%w/w在煤油)的四氢呋喃(20mL)溶液中,所得反应液在室温条件下搅拌半小时后加热升至回流;随后滴加预先溶有4-氟苯乙酮(500mg,3.6mmol)的四氢呋喃(6mL)溶液。所得反应液在回流状态下进一步搅拌半小时。冷却至室温后,将反应液倒入至1M盐酸水溶液,调节pH至7-8;并用乙酸乙酯萃取(30mL×2)。合并有机相用水和饱和食盐水(各50mL)洗涤,无水硫酸钠干燥,浓缩。所得粗品用硅胶柱层析(石油醚/乙酸乙酯=20/1~10/1)得到淡黄色油状物INT-14a(386mg,收率:45.1%)。MS(ESI):m/z 239.3(M+H) +
从化合物INT-14a起,参照化合物INT-2的合成得到化合物INT-14。MS(ESI):m/z 491.2(M+H) +
化合物INT-15的制备
Figure PCTCN2021081965-appb-000018
从4-氟苯乙酮和1,1-二氟乙酸乙酯起,参照化合物INT-14的合成得到化合物INT-15。MS(ESI):m/z 469.4(M+H) +
实施例化合物的合成
实施例1:
Figure PCTCN2021081965-appb-000019
在氮气氛围下,将混有化合物INT-4(10mg,37μmol),联硼酸频那醇酯(9.9mg,39μmol),Pd(PPh 3) 2Cl 2(2.6mg,3.7μmol)和醋酸钾(10.9mg,111μmol)的1,4-二氧六环溶液(5mL)在90℃条件下搅拌2小时。随后在反应液中加入化合物INT-1(15mg,40μmol),Pd(PPh 3) 4(4.7mg,4.0μmol)磷酸钾(25.7mg,121μmol)和水(0.5mL);所得混合液用氮气置换三次,并在100℃条件和氮气气氛下搅拌过夜。反应液冷却至室温,硅藻土过滤,浓缩。所得粗品用制备型薄层层析板(石油醚/乙酸乙酯=3/1)得到黄色油状物1a(16.7mg,收率:86.0%)。 MS(ESI):m/z 481.0(M+H) +.
将溶有化合物1a(16.7mg,35μmol)的三氟乙酸和二氯甲烷的混合溶液(2mL,v/v=1/1)溶液在室温下搅拌2小时。反应液中加入乙酸乙酯和水(各15mL),然后分液;有机相进一步用饱和碳酸氢钠和饱和食盐水洗涤(各10mL),浓缩。所得粗品用制备HPLC分离得到白色固体1(2.0mg,收率:16.4%)。 1H NMR(500MHz,DMSO-d6)δ13.33(s,1H),8.78(d,J=7.3Hz,1H),8.37(s,1H),7.84(s,1H),7.57–7.45(m,3H),7.38–7.16(m,3H),7.09–7.02(m,1H),4.15(q,J=7.1Hz,2H),1.15(t,J=7.1Hz,3H);MS(ESI):m/z 351.2(M+H) +.
实施例2:
Figure PCTCN2021081965-appb-000020
从化合物INT-5起,参照化合物1的合成得到化合物2。 1H NMR(500MHz,DMSO-d6)δ13.25(s,1H),8.57(d,J=7.2Hz,1H),8.03(brs,1H),7.93(d,J=2.0Hz,1H),7.58–7.43(m,3H),7.30–7.17(m,2H),6.66–6.62(m,1H),6.51–6.48(m,1H);MS(ESI):m/z 279.3(M+H) +.
实施例3:
Figure PCTCN2021081965-appb-000021
将氢氧化钠水溶液(3M,1mL)加入至溶有化合物1a(147mg,0.31mmol)的甲醇(5mL)溶液;所得反应液在60℃条件下搅拌2小时。反应液冷却至室温后,用1M盐酸水溶液调节pH至3-4,并用乙酸乙酯萃取(20mL×2)。合并有机相用饱和碳酸氢钠和饱和食盐水洗涤(各30mL),无水硫酸钠干燥,浓缩后得到黄色油状物3a(122mg,收率:88.1%)。MS(ESI):m/z 453.2(M+H) +.
在溶有化合物3a(122mg,0.27mmol)的1,4-二氧六环(4mL)溶液中加入 N,N-二异丙基乙胺(70mg,0.54mmol),HATU(123mg,0.32mmol)和氨(0.4M在1,4-二氧六环中,5mL);所得反应液在50℃条件下搅拌4小时。反应液浓缩,所得粗品用硅胶柱层析(石油醚/乙酸乙酯=1/1)得到淡黄色油状物3b(100mg,收率:82.2%)。
从化合物3b起,参照化合物1的最后一步反应得到化合物3。 1H NMR(500MHz,Chloroform-d)δ8.32–8.27(m,1H),8.27–8.23(m,1H),8.20(d,J=2.6Hz,1H),7.84(brs,1H),7.45–7.37(m,2H),7.28–7.24(m,2H),7.09–7.00(m,2H),6.71–6.65(m,1H);MS(ESI):m/z 322.3(M+H) +.
实施例4:
Figure PCTCN2021081965-appb-000022
从化合物INT-5和化合物INT-7起,参照化合物2的合成得到化合物4。 1H NMR(500MHz,DMSO-d6)δ8.61-8.56(m,1H),8.30(s,1H),7.95(s,1H),7.81(s,1H),7.73(t,J=7.5Hz,1H),7.27–7.22(m,1H),6.90–6.83(m,1H),6.53(s,1H),2.44(s,3H);MS(ESI):m/z 276.3(M+H) +.
实施例5:
Figure PCTCN2021081965-appb-000023
从化合物INT-5和化合物INT-8起,参照化合物2的合成得到化合物5。 1H NMR(500MHz,DMSO-d6)δ8.57(d,J=7.5Hz,1H),8.22(s,1H),8.16(t,J=8.0Hz,1H),8.11(d,J=8.0Hz,1H),7.98–7.94(m,1H),7.89–7.84(m,2H),6.89(d,J=7.5Hz,1H),6.50(s,1H);MS(ESI):m/z 330.3(M+H) +.
实施例6:
Figure PCTCN2021081965-appb-000024
从化合物INT-4和化合物6a起,参照化合物1a的合成得到化合物6a。MS(ESI):m/z 365.3(M+H) +.
从化合物6a起,参照化合物3b的合成得到化合物6。 1H NMR(500MHz,DMSO-d6)δ8.67(d,J=7.1Hz,1H),8.49(s,1H),8.20(s,1H),8.11(s,1H),7.64(brs,1H),7.51–7.41(m,2H),7.26–7.16(m,2H),7.03(brs,1H),6.78–6.72(m,1H),3.93(s,3H);MS(ESI):m/z 336.3(M+H) +.
实施例7:
Figure PCTCN2021081965-appb-000025
从化合物INT-4和化合INT-2起,参照化合物1a的合成得到化合物7a。MS(ESI):m/z 521.5(M+H) +.
从化合物7a起,参照化合物3的合成得到化合物7。 1H NMR(500MHz,DMSO-d6)δ12.88(brs,1H),8.70(d,J=7.0Hz,1H),8.52(s,1H),8.20(s,1H),7.69(s,1H),7.48–7.38(m,2H),7.19(t,J=9.0Hz,2H),7.02(s,1H),6.81–6.73(m,1H),1.95–1.83(m,1H),0.98–0.83(m,4H);MS(ESI):m/z 362.0(M+H) +.
实施例8:
Figure PCTCN2021081965-appb-000026
从化合物INT-4和化合INT-3起,参照化合物7的合成得到化合物8。 1H NMR(500MHz,Methanol-d4)δ8.51(d,J=7.2Hz,1H),8.44(s,1H),8.15(s,1H),7.45–7.38(m,2H),7.07(s,2H),6.73(s,1H),4.62(s,2H),2.40(s,3H);MS(ESI):m/z336.2(M+H) +.
实施例9:
Figure PCTCN2021081965-appb-000027
从化合物INT-4和化合INT-7起,参照化合物7的合成得到化合物9。 1H NMR(500MHz,DMSO-d6)δ13.35(brs,1H),8.66(s,1H),8.48(s,1H),8.40-8.24(m,1H),7.73(t,J=7.5Hz,1H),7.63(s,1H),7.22(s,1H),7.15-6.92(m,2H),2.39(s,3H);MS(ESI):m/z 319.1(M+H) +.
实施例10:
Figure PCTCN2021081965-appb-000028
从化合物INT-4和化合INT-8起,参照化合物7的合成得到化合物10。 1H NMR(500MHz,methanol-d4)δ8.51(d,J=7.0Hz,1H),8.42(s,1H),8.21(s,1H),8.10–7.98(m,3H),7.75–7.70(m,1H),7.14(d,J=7.0Hz,1H);MS(ESI):m/z 373.2(M+H) +.
实施例11:
Figure PCTCN2021081965-appb-000029
从化合物INT-4和化合INT-9起,参照化合物7的合成得到化合物11。 1H NMR(500MHz,Methanol-d4)δ8.52(d,J=7.0Hz,1H),8.42(s,1H),8.38(s,1H),8.05(s,1H),8.00(t,J=8.0Hz,1H),7.90–7.83(m,1H),7.65(d,J=7.5Hz,1H),7.12–7.08(m,1H),6.65(t,J=45Hz,1H);MS(ESI):m/z 355.0(M+H) +.
实施例12:
Figure PCTCN2021081965-appb-000030
从化合物INT-4和化合INT-10起,参照化合物7的合成得到化合物12。 1H  NMR(500MHz,Methanol-d4)δ8.54(d,J=7.5Hz,1H),8.45(s,1H),8.27(s,1H),7.98(t,J=7.5Hz,1H),7.65(d,J=7.5Hz,1H),7.04–6.99(m,1H),2.02–1.93(m,1H),1.04–0.95(m,2H),0.91–0.85(m,2H);MS(ESI):m/z 413.3(M+H) +.
实施例13:
Figure PCTCN2021081965-appb-000031
从化合物INT-4和化合INT-11起,参照化合物7的合成得到化合物13。 1H NMR(500MHz,Methanol-d4)δ8.70–8.42(m,2H),8.33(s,1H),7.72–7.55(m,1H),7.49–7.07(m,2H),7.01–6.85(m,1H),2.59–2.30(m,3H),2.09–1.81(m,1H),1.08–0.85(m,4H);MS(ESI):m/z 359.3(M+H) +.
实施例14:
Figure PCTCN2021081965-appb-000032
从化合物INT-4和化合INT-10起,参照化合物12的合成,将氨换成二甲胺得到化合物14。 1H NMR(500MHz,Methanol-d4)δ8.53(s,1H),8.26(s,1H),8.12-7.93(m,3H),7.64(s,1H),7.03-7.00(m,1H),3.20(s,6H),2.05-1.97(m,1H),1.05-0.95(m,2H),0.92-0.85(m,2H);MS(ESI):m/z 441.2(M+H) +.
实施例15:
Figure PCTCN2021081965-appb-000033
从化合物INT-4和化合INT-10起,参照化合物12的合成,将氨换成甲胺得到化合物15。 1H NMR(500MHz,Methanol-d4)δ8.52(s,1H),8.36(s,1H),8.27(s,1H),8.06-7.94(m,2H),7.64(d,J=7.5Hz,1H),7.00(dd,J=7.0,2.0Hz,1H),2.90(s,3H),1.99(s,1H),1.00(s,2H),0.91-0.84(m,2H);MS(ESI):m/z 427.2(M+H) +.
实施例16:
Figure PCTCN2021081965-appb-000034
从化合物INT-4和化合INT-2起,参照化合物7的合成,将氨换成乙醇胺得到化合物16。 1H NMR(500MHz,Methanol-d4)δ8.50(d,J=7.0Hz,1H),8.42(s,1H),8.31(s,1H),7.44-7.39(m,2H),7.08(t,J=8.5Hz,2H),6.78(d,J=7.0Hz,1H),3.71(t,J=6.0Hz,2H),3.49(t,J=6.0Hz,2H),2.04-1.96(m,1H),1.05-0.96(m,2H),0.92-0.85(m,2H);m/z 405.7(M+H) +.
实施例17:
Figure PCTCN2021081965-appb-000035
从化合物INT-4和化合INT-2起,参照化合物7的合成得到化合物17。 1H NMR(500MHz,Methanol-d4)δ8.51(d,J=7.0Hz,1H),8.44(s,1H),8.17(s,1H),7.42–7.38(m,2H),7.05(t,J=8.5Hz,2H),6.73(d,J=8.0Hz,1H),2.13–2.01(m,2H),1.89–1.71(m,5H),1.71–1.63(m,2H);m/z 389.7(M+H) +.
实施例18:
Figure PCTCN2021081965-appb-000036
从化合物INT-13和化合物INT-2起,参照化合物1的合成得到化合物18。 1H NMR(500MHz,Methanol-d4)δ8.60(d,J=7.2Hz,1H),8.55(s,1H),8.38(s,1H),7.42(m,2H),7.15-7.05(m,2H),6.96(s,1H),2.55(s,3H),2.12-1.96(m,1H),0.99(m,4H);m/z 361.2(M+H) +.
实施例19:
Figure PCTCN2021081965-appb-000037
从化合物7a起,参照化合物1合成中的最后一步得到化合物19a。m/z 391.2(M+H) +.
在冰浴条件下,将二异丁基氢化铝(83μL,2M在甲苯中)滴加至溶有化合物19a(40mg,0.10mmol)的二氯甲烷(1mL)溶液中;所得混合液液在相同条件下搅拌半小时。反应液随后加入到酒石酸钾钠溶液中(5mL,1M水溶液),经乙酸乙酯萃取;有机相减压浓缩后,所得粗品用制备HPLC分离得到白色固体19(5.2mg,收率:14.6%)。 1H NMR(500MHz,Methanol-d4)δ8.44(d,J=7.2Hz,1H),7.95(s,1H),7.70(s,1H),7.44(dd,J=8.5,5.4Hz,2H),7.08(m,2H),6.71(m,1H),4.76(s,2H),2.01(m,1H),0.94(m,4H);m/z 349.3(M+H) +.
实施例20:
Figure PCTCN2021081965-appb-000038
从化合物INT-14和化合物INT-4起,参照化合物1和化合物3的合成得到化合物20a。m/z 394.3(M+H) +.
从化合物20a起,参照化合物19的最后一步合成得到化合物20。 1H NMR(500MHz,Methanol-d4)δ13.31(s,1H),8.69(d,J=7.0Hz,1H),8.53–8.48(m,1H),8.09(s,1H),7.49–7.38(m,2H),7.29–7.13(m,2H),7.01–6.75(m,1H),4.49(s,2H);m/z 352.2(M+H) +.
实施例21:
Figure PCTCN2021081965-appb-000039
从化合物INT-15和化合物INT-4起,参照化合物1和化合物3的合成得到化合物21。 1HNMR(500MHz,Methanol-d4)δ8.56(d,J=7.0Hz,1H),8.44(s,1H),8.23(s,1H),7.42(dd,J=8.5,5.0Hz,2H),7.13(t,J=8.5Hz,2H),6.90–6.83(m,1H),1.36–1.19(m,1H);m/z 372.3(M+H) +.
测试实施例
化合物对TGFβRI激酶(ALK5)活性抑制作用的测定:
TGFβRI激酶购买自Carna(Cat#09-141)。在384孔板(Greiner,Cat#784075)中加入2μl用激酶缓冲液(40mM Tris PH 7.5,20mM MgCl 2,1mM DTT,1mg/ml BSA)配制的酶溶液(TGFβRI激酶在最终反应体系中为25nM)。对于阴性对照则加入2μl激酶缓冲液。将化合物用DMSO配制成10mM的储存液,在检测化合物之前根据起始浓度的需要,用DMSO对化合物进行二次稀释。取2.5μl化合物加入47.5μl ddH 2O中,按照3倍稀释方法进行稀释,每个化合物设置10个浓度点。化合物稀释完成后,取1μl加入酶溶液中。充分混匀后室温孵育10min(最终反应体系中DMSO浓度为1%)。对于阴性对照和阳性对照,则加入1μl含5%DMSO的水溶液。最后加入2μl含有ATP以及TGFβRI激酶多肽底物(Signalchem,Cat#T36-58)的混合液(ATP最终浓度为3.5μM,底物最终浓度为0.1μg/ul),充分混匀后置于28度反应120min。最后使用ADP-Glo Kinase Assay Kit(Promega,Cat#V9102)进行检测。使用Molecular Devices,SpectraMax i3x.读取最终化学发光信号。采用XLFIT对所得到的数据进行4参数曲线拟合并计算IC 50
化合物对p38α激酶活性抑制作用的测定:
p38α激酶购买自SignalChem(Cat#M39-10BG)。在384孔板(Greiner,Cat#784075)中加入2μl用激酶缓冲液(40mM Tris PH 7.5,20mM MgCl 2,0.05mM DTT,0.1mg/ml BSA)配制的酶溶液(p38α激酶在最终反应体系中为0.8ng/ul)。对于阴性对照则加入2μl激酶缓冲液。将化合物用DMSO配制成10mM的储存液,在检测化合物之前根据起始浓度的需要,用DMSO对化合物进行二次稀释。 取2.5μl化合物加入47.5μl ddH 2O中,按照3倍稀释方法进行稀释,每个化合物设置10个浓度点。化合物稀释完成后,取1μl加入酶溶液中。充分混匀后室温孵育10min(最终反应体系中DMSO浓度为1%)。对于阴性对照和阳性对照,则加入1μl含5%DMSO的水溶液。最后加入2μl含有ATP以及p38激酶多肽底物(Signalchem,Cat#P03-58)的混合液(ATP最终浓度为50μM,底物最终浓度为0.125μg/μl),充分混匀后置于25度反应120min。最后使用ADP-Glo Kinase Assay Kit(Promega,Cat#V9102)进行检测。使用Molecular Devices,SpectraMax i3x.读取最终化学发光信号。采用XLFIT对所得到的数据进行4参数曲线拟合并计算IC 50
实施例中所列化合物对于ALK5和p38α的激酶活性抑制作用的结果:
Figure PCTCN2021081965-appb-000040
*参比化合物是LY2157299(即galunisertib)
化合物对TGFβRI受体Smad信号通路的抑制活性的测定:
收集HEK-Blue TGFβcells(Invivogen,Cat#hkb-tgfb),并用10%FBS的DMEM调整细胞密度为1.25×10 6cells/ml,并以每孔40μl加入96孔板中。将化合物用DMSO配制成10mM的储存液,在检测化合物之前根据起始浓度的需要,用DMSO对化合物进行二次稀释。取2.5μl化合物加入1ml完全培养基中,吹 打混匀,用含0.25%DMSO的DMEM培养基对化合物进行3倍稀释(100μl体系中DMSO浓度为0.1%)。取40μl配制好的化合物至96孔细胞板中,阳性对照以及阴性对照中加入40ul含0.25%DMSO的培养基,于37℃5%CO 2培养箱中孵育2小时。每孔加入20μl人重组TGFβ1(Invivogen,cat#Rcyc-htgfb1),其最终浓度为0.1ng/ml。置于37℃5%CO 2中培养24小时。24小时后取60μl上清用于分泌型碱性磷酸酶(SEAP)检测,细胞板中补加60ul/well的培养基用于细胞活性检测。SEAP检测采用Great EscApe SEAP chemiluminescence KIT(Clontech,Cat#631738)进行检测,细胞活性检测采用CellTiter-Glo Luminescent Cell Viability Assay(Promega,Cat#G7573)进行检测。使用Molecular Devices,SpectraMax i3x.读取最终化学发光信号。采用XLFIT对所得到的数据进行4参数曲线拟合并计算IC 50
实施例中所列化合物对于TGFβRI受体Smad信号通路的抑制活性的结果:
Figure PCTCN2021081965-appb-000041
*参比化合物是LY2157299(即galunisertib)

Claims (9)

  1. 式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物:
    Figure PCTCN2021081965-appb-100001
    其中,Cy表示被0、1、2、3个取代基所取代的C 6-C 10芳基或者5-10元杂芳基,其中所述的取代基选自:卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
    R 1表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基;
    R 2表示氢、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-SO 2NR aR b、-NR aSO 2R b
    R 3表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-SO 2NR aR b、-NR aSO 2R b
    R 4表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、羟基(C 1-C 6烷基);
    R 5表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基;或者R 4、R 5与其所连接的原子一起形成5-8元饱和或不饱和环,该环中还可以任意地含有0、1或2个选自N、O、S的杂原子;
    其中,R a、R b分别各自独立地表示氢、C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、卤代C 1-C 6烷基;或者R a、R b与其所连接的原子一起形成5-8元环,该环中还可以任意地含有0、1或2个选自N、O、S的杂原 子;
    n表示0、1、2或3。
  2. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物,其中所述式(I)化合物具有以下式(II)结构:
    Figure PCTCN2021081965-appb-100002
    其中,R 1、R 3、R 4、R 5、R a、R b和n具有如权利要求1所述的定义;
    R 2表示氢、C 1-C 6烷基、C 3-C 6环烷基、羟基(C 1-C 6烷基)、卤代C 1-C 6烷基;
    W表示CR L或者N;
    R 6表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
    其中,R L表示氢、卤素、C 1-C 6烷基、C 3-C 6环烷基、卤代C 1-C 6烷基、-OR a、氰基、硝基、NR aR b、-SO 2R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-P(O)R aR b
    其中,o表示0、1、2或3。
  3. 如权利要求1-2任一项所述的化合物,其选自:
    Figure PCTCN2021081965-appb-100003
    Figure PCTCN2021081965-appb-100004
  4. 一种药物组合物,其包含权利要求1至3任一项所述的式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物,以及任选的药学上可接受的载体。
  5. 如权利要求4所述的药物组合物,其进一步包含另外的治疗剂和/或检查点抑制剂,所述另外的治疗剂优选地选自苯丁酸氮芥、美法仑、环磷酰胺、异环磷酰胺、白消安、卡莫司汀、洛莫司汀、链脲佐菌素、 顺铂、卡铂、奥沙利铂、达卡巴嗪、替莫唑胺、丙卡巴肼、甲氨蝶呤、氟尿嘧啶、阿糖胞苷、吉西他滨、巯基嘌呤、氟达拉滨、长春碱、长春新碱、长春瑞滨、紫杉醇、多西紫杉醇、拓扑替康、伊立替康、依托泊苷、曲贝替定、更生霉素、多柔比星、表柔比星、道诺霉素、米托蒽醌、博来霉素、丝裂霉素C、伊沙匹隆、他莫昔芬、氟他胺、戈那瑞林类似物、甲地孕酮、强的松、***、甲泼尼龙、沙利度胺、干扰素α、亚叶酸钙、西罗莫司、西罗莫司脂化物、依维莫司、阿法替尼、alisertib、amuvatinib、阿帕替尼、阿西替尼、硼替佐米、波舒替尼、布立尼布、卡博替尼、西地尼布、crenolanib、克卓替尼、达拉菲尼、达可替尼、达努塞替、达沙替尼、多维替尼、厄洛替尼、foretinib、ganetespib、吉非替尼、依鲁替尼、埃克替尼、伊马替尼、iniparib、拉帕替尼、lenvatinib、linifanib、linsitinib、马赛替尼、momelotinib、莫替沙尼、来那替尼、尼罗替尼、niraparib、oprozomib、olaparib、帕唑帕尼、pictilisib、普纳替尼、quizartinib、瑞格菲尼、rigosertib、rucaparib、鲁索利替尼、塞卡替尼、saridegib、索拉非尼、舒尼替尼、替拉替尼、tivantinib、替沃扎尼、托法替尼、曲美替尼、凡德他尼、维利帕尼、威罗菲尼、维莫德吉、volasertib、阿仑单抗、贝伐单抗、贝伦妥单抗维多汀、卡妥索单抗、西妥昔单抗、地诺单抗、吉妥珠单抗、伊匹单抗、尼妥珠单抗、奥法木单抗、帕尼单抗、利妥昔单抗、托西莫单抗、曲妥珠单抗;所述的检查点抑制剂优选地选自抗PD-1抗体、抗PD-L1抗体、LAG3抗体、TIM-3抗体及抗CTLA-4抗体。
  6. 如权利要求1-3任一项所述的式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物或者如权利要求4-5任一项所述的药物组合物在制备用于通过抑制TGF-β受体来预防或治疗的肿瘤、癌症、病毒感染、器官移植排斥、神经退行性疾病、注意力缺陷相关疾病或自身免疫性疾病的药物中的用途,其中所述TGF-β受体优选选自TGFβRI(ALK5)受体。
  7. 如权利要求6所述的用途,其中,所述肿瘤或癌症选自皮肤癌、膀胱癌、卵巢癌、乳腺癌、胃癌、胰腺癌、***癌、结肠癌、肺癌、骨癌、脑癌、神经细胞瘤、直肠癌、结肠癌、家族性腺瘤性息肉性癌、遗传性非息肉性结直肠癌、食管癌、唇癌、喉癌、下咽癌、舌癌、唾 液腺癌、胃癌、腺癌、甲状腺髓样癌、***状甲状腺癌、肾癌、肾实质癌、卵巢癌、***、子宫体癌、子宫内膜癌、绒毛膜癌、睾丸癌、泌尿癌、黑素瘤、脑肿瘤诸如成胶质细胞瘤、星形细胞瘤、脑膜瘤、成神经管细胞瘤和外周神经外胚层肿瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、伯基特淋巴瘤、急性淋巴性白血病(ALL)、慢性淋巴性白血病(CLL)、急性骨髓性白血病(AML)、慢性粒细胞白血病(CML)、成人T细胞白血病淋巴瘤、弥漫性大B细胞淋巴瘤(DLBCL)、肝细胞癌、胆囊癌、支气管癌、小细胞肺癌、非小细胞肺癌、多发性骨髓瘤、基底细胞瘤、畸胎瘤、成视网膜细胞瘤、脉络膜黑素瘤、***瘤、横纹肌肉瘤、颅咽管瘤、骨肉瘤、软骨肉瘤、肌肉瘤、脂肪肉瘤、纤维肉瘤、尤因肉瘤和浆细胞瘤。
  8. 一种预防或***、癌症、病毒感染、器官移植排斥、神经退行性疾病、注意力缺陷相关疾病或自身免疫性疾病的方法,其包括向有此需要的哺乳动物施用权利要求1-3任一项所述的化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物或者权利要求4或5所述的药物组合物。
  9. 一种抑制TGF-β受体活性的方法,其包括使TGF-β受体暴露于如权利要求1-3任一项所述的式(I)化合物或其药学上可接受的盐、前药、同位素衍生物、异构体、溶剂化物、或其代谢产物,其中所述TGF-β受体优选选自TGFβRI(ALK5)受体。
PCT/CN2021/081965 2020-03-24 2021-03-22 转化生长因子-β受体抑制剂 WO2021190425A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010212752.X 2020-03-24
CN202010212752 2020-03-24

Publications (1)

Publication Number Publication Date
WO2021190425A1 true WO2021190425A1 (zh) 2021-09-30

Family

ID=77891775

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/081965 WO2021190425A1 (zh) 2020-03-24 2021-03-22 转化生长因子-β受体抑制剂

Country Status (2)

Country Link
TW (1) TWI768781B (zh)
WO (1) WO2021190425A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114853754B (zh) * 2022-05-23 2023-04-18 云白药征武科技(上海)有限公司 一种硫代酰胺衍生物及其制备方法和应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017215506A1 (zh) * 2016-06-13 2017-12-21 南京明德新药研发股份有限公司 作为TGF-βR1抑制剂的苯并三氮唑衍生的α,β不饱和酰胺类化合物
WO2018017633A1 (en) * 2016-07-21 2018-01-25 Bristol-Myers Squibb Company TGF Beta RECEPTOR ANTAGONISTS
CN107663206A (zh) * 2016-07-29 2018-02-06 上海璎黎药业有限公司 含氮芳香杂环化合物、其制备方法、药物组合物和应用
CN110066277A (zh) * 2018-01-24 2019-07-30 上海璎黎药业有限公司 芳香杂环取代烯烃化合物、其制备方法、药物组合物和应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017215506A1 (zh) * 2016-06-13 2017-12-21 南京明德新药研发股份有限公司 作为TGF-βR1抑制剂的苯并三氮唑衍生的α,β不饱和酰胺类化合物
WO2018017633A1 (en) * 2016-07-21 2018-01-25 Bristol-Myers Squibb Company TGF Beta RECEPTOR ANTAGONISTS
CN107663206A (zh) * 2016-07-29 2018-02-06 上海璎黎药业有限公司 含氮芳香杂环化合物、其制备方法、药物组合物和应用
CN110066277A (zh) * 2018-01-24 2019-07-30 上海璎黎药业有限公司 芳香杂环取代烯烃化合物、其制备方法、药物组合物和应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LI XINGZHOU, LIU FENG, LI SONG: "Research Progress on Transforming Growth Factor β Signal and its Signal Pathway Blockers", FOREIGN MEDICAL SCIENCES SECTION OF PHARMACY, MILITARY MEDICAL SCIENCES, INSTITUT FÜR PHARMAKOLOGIE UND TOXIKOLOGIE, CN, vol. 33, no. 5, 31 October 2006 (2006-10-31), CN, pages 331 - 335, XP055853037, ISSN: 1001-0971, DOI: 10.13220/j.cnki.jipr.2006.05.005 *
SUN LI-QIAN, MENG LI-QIANG, YAN CHAO-QUN, CUI DONG-XIAO, MIAO JUN-QIU, CHEN JING-RUN, LIANG TAI-GANG, LI QING-SHAN: "3D-QSAR Studies on 4-([1,2,4]Triazolo[1,5-α] pyridin-6-yl)-5(3)-(6-methylpyridin-2-yl)imidazole Analogues as Potent Inhibitors of Transforming Growth Factor-β Type Ⅰ Receptor Kinase", JIEGOU HUAXUE - CHINESE JOURNAL OF STRUCTURAL CHEMISTRY, ZHONGGUO KEXUEYUAN FUJIAN WUZHI JIEGOU YANJIUSUO, CN, vol. 37, no. 4, 15 April 2018 (2018-04-15), CN, pages 517 - 530, XP055853038, ISSN: 0254-5861, DOI: 10.14102/j.cnki.0254-5861.2011-1766 *

Also Published As

Publication number Publication date
TW202144347A (zh) 2021-12-01
TWI768781B (zh) 2022-06-21

Similar Documents

Publication Publication Date Title
WO2019158070A1 (zh) A2a和/或a2b受体拮抗剂
CN111548343B (zh) 一种高活性csf1r抑制剂化合物的制备方法
CN110914277B (zh) 咪唑并[1,2-b]嘧啶并[4,5-d]哒嗪-5(6H)-酮类化合物及其应用
KR20150090044A (ko) 아자퀴나졸린 카복사미드 유도체
WO2022089398A1 (zh) 一种高活性的hpk1激酶抑制剂
CN112028891B (zh) 腺苷受体拮抗剂
CN112313220B (zh) Pd-l1拮抗剂化合物
WO2021190425A1 (zh) 转化生长因子-β受体抑制剂
WO2021129584A1 (zh) Pd-l1拮抗剂化合物
CN112592354B (zh) 一种异噁唑并嘧啶类杂环化合物的制备方法
CN111440148B (zh) 一种腺苷受体拮抗剂的制备方法
JP2024513538A (ja) Parp阻害剤としての置換融合二環式化合物及びその使用
JP2018536692A (ja) 新規なジヒドロピラノピリミジノン誘導体およびその用途{Novel dihydropyranopyrimidinone derivatives、and use thereof}
LU505117B1 (en) A pan-KRAS inhibitor compound
WO2023025225A1 (zh) 细胞周期蛋白k降解剂
WO2024022365A1 (zh) 一种Wnt通路抑制剂化合物
WO2022199561A1 (zh) Hpk1激酶抑制剂化合物
WO2022214008A1 (zh) 高活性hpk1激酶抑制剂
WO2023143546A1 (zh) Wnt通路抑制剂化合物
BR112018009880B1 (pt) Compostos e composições farmacêuticas moduladores dos receptores de quimiocina e seus usos

Legal Events

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

Ref document number: 21775613

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21775613

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 06/07/2023)