CN111362967A - Benzoxadiazatetetradecene derivatives and use thereof - Google Patents

Benzoxadiazatetetradecene derivatives and use thereof Download PDF

Info

Publication number
CN111362967A
CN111362967A CN202010347120.4A CN202010347120A CN111362967A CN 111362967 A CN111362967 A CN 111362967A CN 202010347120 A CN202010347120 A CN 202010347120A CN 111362967 A CN111362967 A CN 111362967A
Authority
CN
China
Prior art keywords
pharmaceutically acceptable
methyl
acceptable salt
derivative
cycloalkyl
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.)
Granted
Application number
CN202010347120.4A
Other languages
Chinese (zh)
Other versions
CN111362967B (en
Inventor
范文华
唐春雷
范为正
吴露婷
张晴
张永杰
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.)
Nanjing Leizheng Pharmaceutical Technology Co ltd
Jiangnan University
Original Assignee
Nanjing Leizheng Pharmaceutical Technology Co ltd
Jiangnan University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Leizheng Pharmaceutical Technology Co ltd, Jiangnan University filed Critical Nanjing Leizheng Pharmaceutical Technology Co ltd
Priority to CN202010347120.4A priority Critical patent/CN111362967B/en
Publication of CN111362967A publication Critical patent/CN111362967A/en
Application granted granted Critical
Publication of CN111362967B publication Critical patent/CN111362967B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs

Landscapes

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

Abstract

The invention discloses a benzoxadiazacyclotetradecene derivative and application thereof, belonging to the field of medicines. The benzoxadiazacyclotetradecene derivative with the structure shown in the general formula (I) has excellent anaplastic lymphoma enzyme (ALK) inhibition activity and excellent pharmacodynamic performance, and can remarkably prolong the large metabolic half-life of a medicament; can be safely and effectively used for treating anaplastic lymphoma kinase positive (ALK +) metastatic (advanced) non-small cell lung cancer (NSCLC) and the like, therebyProvides a new means for treating cancer, metabolic and immune diseases, cardiovascular diseases, neurological diseases and the like.

Description

Benzoxadiazatetetradecene derivatives and use thereof
Technical Field
The invention relates to a benzoxadiazacyclotetradecene derivative and application thereof, belonging to the field of medicines.
Background
It is well known that lung cancer is one of the most threatening malignancies to humans, with morbidity and mortality ranking first in men and second in women. Among lung cancers, the most common one belongs to non-small cell lung cancer (NSCLC), accounting for about 85% of lung cancers; wherein the positive incidence rate of Anaplastic Lymphoma Kinase (ALK) accounts for 3 to 5 percent of that of non-small cell lung cancer (NSCLC).
ALK is a member of the receptor tyrosine kinase superfamily and the amino acid sequence level is most closely related to members such as Ros-1, leukocyte tyrosine kinase, insulin receptor and cMet (liver growth factor receptor), as are all members of this gene family, which have extracellular ligand binding regions, transmembrane sequences and intracellular kinase catalytic/signaling regions. The signaling ligand properties of anaplastic lymphoma enzyme (ALK) have not been elucidated, and different mechanisms have been proposed in the literature (Stoica G. E. et a., ]. Bio l. chem.,2001,276, 16772-. Anaplastic lymphoma enzyme (ALK) is abundantly expressed primarily in the developing nervous system, and although its expression is retained in certain regions of the brain, spinal cord and eye, its relative content in adult animals tends to decrease. Anaplastic Lymphoma Kinase (ALK) plays an important role in oncology, and point mutations of the full-length ALK enzyme that cause activation of this enzyme and increase expression of the full-length enzyme have all been shown to result in neuroblastoma. Furthermore, fusion of ALK and other proteins due to gene translocation has also been shown to result in activated kinase regions associated with cancer. Many such ALK translocations leading to gene fusions are seen in lymphomas, the most common being the nuclear phosphoprotein NPM-ALK fusions seen in anaplastic large cell lymphomas. ALK and EML4 fusion produces a chimeric protein (EML4-ALK) that is thought to be associated with 3 to 5% of non-small cell lung adenocarcinoma (NSCLC).
In 2011, the ALK/ROS1/c-MET inhibitor crizotinib was approved for the treatment of locally advanced or metastatic NSCLC patients that were ALK-positive as detected by FDA-approved tests. Crizotinib also showed efficacy in treating NSCLC with ROS1 translocation, as clinically observed for other tyrosine kinase inhibitors, mutations in ALK and ROS1 that have conferred resistance to ALK inhibitors.
Therefore, ALK and ROS1 are attractive molecular targets for cancer therapeutic intervention. There remains a need to identify compounds with novel activity profiles for both wild-type and mutant forms of ALK and ROS 1.
Currently, the target drugs for ALK mutation are first-generation ALK targeting drug Crizotinib (Crizotinib), and second-generation ALK targeting drugs include Ceritinib (Ceritinib), aletinib (Alectinib), bugatinib (brigatinib) and the newly-marketed third-generation ALK targeting drug loratinib (loretinanib).
Loratinib (lorelatinb, PF-06463922) is a novel anti-drug resistant ALK inhibitor developed by fevered for the investigative treatment of anaplastic lymphoma kinase positive (ALK +) metastatic (advanced) non-small cell lung cancer (NSCLC). Moreover, the medicine has high blood brain barrier permeability, and is approved to exert better efficacy on the non-small cell lung cancer with central nervous system metastasis.
Figure BDA0002470516720000021
However, the drug has poor metabolic stability, short half-life period, large dosage, and easy generation of drug resistance and toxic and side effects.
Disclosure of Invention
In order to solve the problems, the inventor develops a novel benzoxadiazacyclotetradecene derivative used as an ALK inhibitor, and the compound has the advantages of lower toxic and side effects, effectively increased blood brain barrier passing rate, longer half life and better safety. The inhibitor is expected to have good curative effect, is expected to overcome the problems of drug resistance and toxic and side effects, and has good development prospect. The novel benzoxadiazacyclotetradecene derivatives and salts of the present invention are effective in inhibiting anaplastic lymphoma enzyme (ALK) and are useful for treating or ameliorating abnormal cell proliferative disorders.
The invention provides a novel benzoxadiazacyclotetradecene derivative serving as an anaplastic lymphoma enzyme (ALK) inhibitor, and the structural compound has better pharmacodynamic performance and higher metabolic stability.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof:
Figure BDA0002470516720000022
wherein when R is1When it is hydrogen, R2、R3Each independently selected from C3-C6Cycloalkyl, deuterated methyl; wherein cycloalkyl may be optionally mono-to pentasubstituted with the same or different substituents selected from halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group;
when R is1Is selected from C1-C4Alkyl radical, C3-C6Cycloalkyl, -CH2OR4、-COOR5、-COR6or-CH2OP(O)OR7OR8When R is2、R3Each independently selected from C1-C4Alkyl, deuterated methyl or C3-C6A cycloalkyl group; wherein saidThe alkyl and cycloalkyl groups may be optionally mono-to pentasubstituted with the same or different substituents selected from halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group; wherein R is4,R5,R6Are respectively and independently selected from hydrogen and C1-C4Alkyl or C3-C6A cycloalkyl group; r7,R8Each independently selected from hydrogen and C1-C4Alkyl or C3-C6A cycloalkyl group; wherein said alkyl, cycloalkyl may be optionally mono-to pentasubstituted with the following same or different substituents selected from: halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, wherein when R is1When it is hydrogen, R2、R3At least one of each is deuterated methyl and the other is selected from C1-C4Alkyl radical, C3-C6Cycloalkyl, deuterated methyl. Wherein, C3-C6Cycloalkyl is preferably cyclopropyl; c1-C4The alkyl group is preferably a methyl group.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, R4Preferably hydrogen, C1-C4An alkyl group. Further preferred are methyl and hydrogen.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, R5Preferably selected from hydrogen, methyl, ethyl or isopropyl;
the invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, R6Hydrogen is preferred.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, R7,R8Independently of one another, preferably hydrogen and methylEthyl, isopropyl, tert-butyl; further preferred are ethyl and tert-butyl.
The invention provides a benzoxadiazepitetradecene derivative with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof, which is selected from the following groups:
Figure BDA0002470516720000041
the invention relates to a benzoxadiazacyclotetradecene derivative with a structure shown in a general formula (I) or a pharmaceutically acceptable salt thereof, wherein the pharmaceutically acceptable salt is an inorganic salt or an organic salt, and the inorganic salt comprises hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate and acid phosphate; the organic salt is selected from acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate, maleate, lactate, malate, citrate, tartrate, methanesulfonate, benzenesulfonate, salicylate.
The invention also aims to provide a pharmaceutical composition, which contains the compound with the general formula (I) or the pharmaceutically acceptable salt thereof, and pharmaceutically acceptable carriers and pharmaceutical excipients.
The drug carrier comprises: microcapsules, microspheres, nanoparticles, liposomes.
The pharmaceutic adjuvant comprises: comprising solvents, propellants, solubilizers, cosolvents, emulsifiers, colorants, binders, disintegrants, fillers, lubricants, wetting agents, osmotic pressure regulators, stabilizers, glidants, flavoring agents, preservatives, suspending agents, coating materials, fragrances, anti-adhesives, integration agents, permeation enhancers, pH regulators, buffers, plasticizers, surfactants, foaming agents, antifoaming agents, thickeners, encapsulation agents, humectants, absorbents, diluents, flocculants and deflocculants, filter aids, and release retardants.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated as solid formulations for oral administration, including, but not limited to, capsules, tablets, pills, powders, granules, and the like.
The single dose of the compound of the present invention or a pharmaceutically acceptable salt thereof may be 0.01 to 1000mg, for example, 0.05 to 800mg, 0.1 to 500mg, 0.01 to 300mg, 0.01 to 200mg, 0.05 to 150mg, 0.05 to 50mg, etc.
In another aspect, the present invention provides the use of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use as an ALK inhibitor.
In another aspect, the present invention provides the use of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
The cancer mentioned in the invention can be selected from non-small cell lung cancer (NSCLC), squamous cell carcinoma, hormone refractory prostate cancer, papillary renal cell carcinoma, colorectal adenocarcinoma, neuroblastoma, Anaplastic Large Cell Lymphoma (ALCL), gastric cancer, metastatic brain cancer and the like.
The invention has the beneficial effects that:
the benzoxadiazacyclotetradecene derivative with the structure shown in the general formula (I) has a convenient synthesis method, good anaplastic lymphoma enzyme (ALK) inhibition activity and excellent pharmacodynamic performance, and can remarkably prolong the large metabolic half-life of a medicament.
The novel benzoxadiazacyclotetradecene derivative has the biological function of inhibiting ALK, is used for treating anaplastic lymphoma kinase positive (ALK +) metastatic (advanced) non-small cell lung cancer (NSCLC), and provides a new means for searching and treating cancers, metabolic and immune diseases, cardiovascular diseases, neurological diseases and the like.
Detailed Description
The technical solution of the present invention will be described in detail with reference to the following examples.
In the present invention, "C1-C4Alkyl "refers to a saturated straight or branched chain monovalent hydrocarbon group of 1 to 4 carbon atoms, respectively. Examples include, but are not limited to, methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2-methyl-1-propyl, 2-butyl, and 2-methyl-2-propyl.
In the present invention, "C3-C6Cycloalkyl "refers to cycloalkyl groups of 3 to 6 carbon atoms each.
In the present invention "administering" or "administering" an individual compound means providing a compound of the invention to an individual in need of treatment.
< Compound or pharmaceutically acceptable salt thereof >
The invention provides a novel benzoxadiazacyclotetradecene derivative serving as an anaplastic lymphoma enzyme (ALK) inhibitor or a pharmaceutically acceptable salt thereof, wherein the structural formula of the derivative is shown as a general formula (I):
Figure BDA0002470516720000061
the present invention also provides a process for the preparation of a compound of formula (I) by the reaction of:
reaction scheme 1:
Figure BDA0002470516720000062
wherein R is4Selected from hydrogen, C1-C4Alkyl or C3-C6Cycloalkyl when R4When hydrogen is obtained by reacting a compound of formula (II) with formaldehyde under tetrabutylammonium fluoride conditions, when R is4Is C1-C4Alkyl radical, C3-C6Cycloalkyl radicals can be formed by reaction with R4OCH2Br is obtained under alkaline conditions;
alternatively, reaction scheme 2:
Figure BDA0002470516720000063
wherein R is5Selected from hydrogen, C1-C4Alkyl or C3-C6Cycloalkyl radicals obtainable by reacting a compound of the formula (II) with R5OCOCl is obtained under alkaline conditions;
alternatively, reaction scheme 3:
Figure BDA0002470516720000064
wherein R is6Selected from hydrogen, C1-C4Alkyl or C3-C6Cycloalkyl when R6When hydrogen, by reaction of a compound of formula (II) with formic acid, when R is6Is C1-C4Alkyl or C3-C6Cycloalkyl radicals obtainable by reacting a compound of the formula (II) with R6COCl is obtained under alkaline condition;
alternatively, reaction scheme 4:
Figure BDA0002470516720000071
wherein R is7,R8Independently selected from hydrogen, C1-C4Alkyl or C3-C6A cycloalkyl group;
suitable bases include tertiary amine bases such as Diisopropylethylamine (DIEA) and triethylamine, inorganic bases, and the like.
< pharmaceutical composition >
The invention also provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, and a pharmaceutically acceptable carrier, excipient or diluent.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated as solid formulations for oral administration, including, but not limited to, capsules, tablets, pills, powders, granules, and the like. In these solid dosage forms, the compounds of general formula (I) according to the invention as active ingredient are mixed with at least one customary inert excipient (or carrier), for example with sodium citrate or dicalcium phosphate. Or mixing with the following components: (1) fillers or solubilizers, for example, starch, lactose, sucrose, glucose, mannitol, silicic acid, and the like; (2) binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose, gum arabic and the like; (3) humectants, such as glycerol and the like; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium carbonate, and the like; (5) a slow solvent such as paraffin and the like; (6) absorption accelerators such as quaternary ammonium compounds and the like; (7) wetting agents such as cetyl alcohol and glyceryl monostearate and the like; (8) adsorbents, for example, kaolin, and the like; (9) lubricants, for example, talc, calcium stearate, solid polyethylene glycols, sodium lauryl sulfate, and the like, or mixtures thereof. Capsules, tablets, pills, etc. may also contain buffering agents.
The solid dosage forms, e.g., tablets, dragees, capsules, pills, and granules, can be coated or microencapsulated with coating and shell materials such as enteric coatings and other crystalline forms of materials well known in the art. They may contain opacifying agents and the release of the active ingredient in such compositions may be delayed in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active ingredient may also be in microencapsulated form with one or more of the above excipients.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated in liquid dosage forms for oral administration, including, but not limited to, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, tinctures, and the like. In addition to the compounds of formula (I) or pharmaceutically acceptable salts thereof as active ingredients, the liquid dosage forms may contain inert diluents conventionally employed in the art, such as water and other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, and oils, particularly cottonseed oil, peanut oil, corn oil, olive oil, castor oil, sesame oil and the like or mixtures of such materials and the like. In addition to these inert diluents, the liquid dosage forms of the present invention may also include conventional adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, perfuming agents and the like.
Such suspending agents include, for example, ethoxylated stearyl alcohol, polyoxyethylene sorbitol, and sorbitan, microcrystalline cellulose, agar, and the like, or mixtures of these materials.
The compounds of the present invention and pharmaceutically acceptable salts thereof may be formulated for parenteral injection in dosage forms including, but not limited to, physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions and dispersions. Suitable carriers, diluents, solvents, excipients include water, ethanol, polyols and suitable mixtures thereof.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be formulated into dosage forms for topical administration, including, for example, ointments, powders, suppositories, drops, sprays, inhalants and the like. The compounds of the general formula (I) according to the invention or their pharmaceutically acceptable salts as active ingredients are mixed under sterile conditions with physiologically acceptable carriers and optionally preservatives, buffers and, if desired, propellants.
The pharmaceutical composition comprises the compound of the general formula (I) or pharmaceutically acceptable salt thereof as an active ingredient, and pharmaceutically acceptable carriers, excipients and diluents. In preparing the pharmaceutical compositions, the compounds of formula (I) or pharmaceutically acceptable salts thereof of the present invention are typically mixed with a pharmaceutically acceptable carrier, excipient or diluent. Wherein the content of the compound of the general formula (I) or the pharmaceutically acceptable salt thereof may be 0.01-1000mg, for example, 0.05-800mg, 0.1-500mg, 0.01-300mg, 0.01-200mg, 0.05-150mg, 0.05-50mg, etc.
< use >
The invention also provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer in a mammal.
A compound of general formula (I), or a pharmaceutically acceptable salt thereof, for use in a method of treating a metastatic (advanced) non-small cell lung cancer (NSCLC) disease in a mammal, including a human, by inhibiting anaplastic lymphoma enzyme (ALK) activity, which method comprises administering to said mammal, including a human, a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, as defined above.
A "therapeutically effective amount" is an amount of a compound of the invention effective to produce a biological or medical response (e.g., decrease or inhibit enzymatic protein activity, or ameliorate symptoms, alleviate a condition, slow or delay disease progression, or prevent disease) in an individual.
Cancers to which the present invention refers include lung cancer, bone cancer, membrane adenocarcinoma, skin cancer, cancer of the head or neck, skin melanoma or endobulbar melanoma, uterine cancer, ovarian cancer, rectal cancer, carcinoma of the biliary tree, stomach cancer, colon cancer, breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, hodgkin's disease, carcinoma of the esophagus, carcinoma of the small intestine, carcinoma of the endocrine system, carcinoma of the thyroid gland, carcinoma of the parathyroid gland, carcinoma of the adrenal gland, carcinoma of soft tissue, carcinoma of the urethra, carcinoma of the penis, carcinoma of the prostate, chronic or acute leukemia, lymphocytic lymphomas, cancer of the shoulder, carcinoma of the kidney or ureter, renal cell carcinoma, menamandoma, carcinoma of the Central Nervous System (CNS), primary central nervous system lymphoma, spinal axis carcinoma, brain stem glioma, pituitary adenoma, and the like.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be administered to mammals including humans, either orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously), topically (powders, ointments, drops) or intratumorally.
The compound or the pharmaceutically acceptable salt thereof can be independently administered or combined with other pharmaceutically acceptable therapeutic agents to be combined with other antitumor drugs. Such combination therapy may be achieved by the simultaneous, sequential or separate use of the individual components of the therapy. Such therapeutic agents include, but are not limited to: antineoplastic acting on DNA chemical structure, such as cisplatin, antineoplastic acting on nucleotide synthesis, such as methotrexate, 5-fluorouracil and the like, antineoplastic acting on nucleic acid transcription, such as adriamycin, epirubicin, aclacinomycin and the like, antineoplastic acting on tubulin synthesis, such as taxol, vinorelbine and the like, aromatase inhibitors, such as aminoglutethimide, letrozole, rennin and the like, cell signaling pathway inhibitors, such as ALK inhibitors crizotinib, ceritinib, exetinib, loratinib and the like. Anti-tumor monoclonal antibodies, immunosuppressants PD-1, PD-L1 and the like, and the components to be combined can be administered simultaneously or sequentially, in a single preparation form or in different preparation forms. Such combinations include not only combinations of one or other active agents of the compounds of the present invention, but also combinations of two or more other active agents of the compounds of the present invention.
The invention will be further illustrated with reference to the following specific examples. It is to be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental methods in the following examples, which are not specified under specific conditions, are generally performed under conventional conditions.
The following intermediate substances are involved in the specific embodiment and can be synthesized by the following process route:
intermediate 9: preparation of tert-butyl ((4-bromo-5-cyano-1-methyl-1H-pyrazol-3-yl) methyl) (methyl) carbamate
The route is as follows:
Figure BDA0002470516720000091
the specific process comprises the following steps:
step a: preparation of methyl 2, 4-dicarbonyl valerate (intermediate 1):
dimethyl oxalate (23.6g,0.2mol) and acetone (29.2g,0.2mol) in an ice salt bath are slowly dropped into 25% sodium methoxide methanol (200mL,0.2mol), the mixture is stirred for 8 hours at 0 ℃, the reaction solution is poured into 200mL of water, ethyl acetate is used for extraction, the pH of an aqueous phase is adjusted to be 2-3 by concentrated hydrochloric acid, the aqueous phase is extracted by ethyl acetate, an organic phase is washed by water, saturated salt is washed by water, and the product is concentrated after drying to obtain 26g of a product, wherein the yield is as follows: 90 percent.
Step b: preparation of methyl 3-methyl-1H-pyrazole-5-carboxylate (intermediate 2):
dissolving methyl 2, 4-dicarbonyl valerate (intermediate 1) (12g,0.076mol) in 100mL dichloromethane, slowly adding hydrazine hydrate (85%, 5.3g,0.091mol) dropwise under ice salt bath, stirring at 0 ℃ for 3 hours, pouring the reaction solution into 200mL water, extracting with dichloromethane, washing the organic phase with water, drying, and concentrating to obtain 8.8g of a product, wherein the yield is as follows: 83 percent.
Step c: preparation of methyl 1, 3-dimethyl-1H-pyrazole-5-carboxylate (intermediate 3):
dissolving 3-methyl-1H-pyrazole-5-carboxylic acid methyl ester (intermediate 2) (5g,0.036mol) in 50mL tetrahydrofuran, adding methyl iodide (6.1g,0.043mol), reacting at 50 ℃ for 3 hours, pouring the reaction solution into 100mL water, extracting with ethyl acetate, washing the organic phase with water, drying, and concentrating to obtain 4.5g of a product, wherein the yield is as follows: 82 percent.
Step d: preparation of methyl 4-bromo-1, 3-dimethyl-1H-pyrazole-5-carboxylate (intermediate 4):
methyl 1, 3-dimethyl-1H-pyrazole-5-carboxylate (intermediate 3) (1g,6.49mmol) was dissolved in 5mL DMF and NBS (1.73g,9.73mmol) was added portionwise and stirred at room temperature for 4 hours after the addition. And (3) dropwise adding the reaction solution into 50mL of ice water, separating out a white solid, filtering, washing a filter cake with water, and drying to obtain 1.1g of a product, wherein the yield is as follows: 73 percent.
Step e: preparation of 4-bromo-1, 3-dimethyl-1H-pyrazole-5-carboxamide (intermediate 5)
Adding 4-bromo-1, 3-dimethyl-1H-pyrazole-5-carboxylic acid methyl ester (intermediate 4) (1g,0.0043mol) into 10mL of ammonia water, stirring overnight at room temperature, precipitating a solid, performing suction filtration, and drying to obtain 0.65g of a product, wherein the yield is as follows: 69.5 percent.
Step f: preparation of 4-bromo-1, 3-dimethyl-1H-pyrazole-5-carbonitrile (intermediate 6)
Suspending 4-bromo-1, 3-dimethyl-1H-pyrazole-5-carboxamide (intermediate 5) (400mg,1.83mmol) in 5mL acetonitrile, adding phosphorus oxychloride (844mg,3.0eq), heating to 80 ℃ for reaction for 3 hours, cooling the reaction solution to room temperature, slowly pouring into ice water, precipitating a solid, filtering, washing with water, and drying to obtain a product 200mg, wherein the yield is 54.5%.
Step g: preparation of 4-bromo-3- (bromomethyl) -1-methyl-1H-pyrazole-5-carbonitrile (intermediate 7)
4-bromo-1, 3-dimethyl-1H-pyrazole-5-carbonitrile (intermediate 6) (500mg,2.5mmol), NBS (489mg,2.75mmol), AIBN (82mg,0.5mmol) was dissolved in 10mL of carbon tetrachloride, protected with nitrogen and refluxed overnight. After the reaction solution was cooled to room temperature, it was filtered, and the filtrate was spin-dried to obtain 600mg of the product with a yield of 86%.
Step h: preparation of 4-bromo-1-methyl-3- ((methylamino) methyl) -1H-pyrazole-5-carbonitrile (intermediate 8)
Dissolving 4-bromo-3- (bromomethyl) -1-methyl-1H-pyrazole-5-carbonitrile (intermediate 7) (1g,3.6mmol) in 10mL of methylamine alcohol solution, stirring overnight at room temperature, rotary evaporating to dryness, adjusting pH to 2-3 with dilute hydrochloric acid, washing with ethyl acetate to remove impurities, adjusting pH of the aqueous phase to 9-10 with 1M aqueous NaOH, extracting with ethyl acetate, combining the organic phases, washing with water, drying, and concentrating to give 600mg of product, yield: 73 percent.
Step i: preparation of tert-butyl ((4-bromo-5-cyano-1-methyl-1H-pyrazol-3-yl) methyl) (methyl) carbamate (intermediate 9)
4-bromo-1-methyl-3- ((methylamino) methyl) -1H-pyrazole-5-carbonitrile (intermediate 8) (500mg,2.18mmol) was dissolved in 10mL tetrahydrofuran, triethylamine (330mg,3.27mmol) and di-tert-butyl dicarbonate (713mg,3.27mmol) were added, stirring was carried out at room temperature for 3 hours, the reaction was diluted with ethyl acetate, washed with aqueous citric acid, washed with water, dried, and concentrated to give 680mg of product, yield: 95 percent.1H NMR(400MHz,CDCl3)δ4.46(br s,2H),4.01(s,3H),2.83(brs,3H),1.47(s,9H).
Intermediate 14: preparation of methyl (R) -2- (1- ((2-amino-5-bromopyridin-3-yl) oxy) ethyl) -4-fluorobenzoate
The route is as follows:
Figure BDA0002470516720000111
step j: preparation of (S) -1- (5-fluoro-2-iodophenyl) ethan-1-ol (intermediate 10)
A solution of (-) -DIPCI (5.7g,17.8mmol) in tetrahydrofuran (50mL) was cooled to-20 to-30 deg.C, then a solution of 1- (5-fluoro-2-iodophenyl) ethanone (3.13g,11.9mmol) in tetrahydrofuran (50mL) was added dropwise and the reaction was heated to room temperature. After 2h, cool to-30 deg.C, add another portion of (-) -DIPCl (3.8g,11.9mmol), incubate for 30 min, then warm to room temperature, concentrate to dryness after 1h, and dissolve the residue with methyl tert-butyl ether (100 mL). A solution of diethanolamine (3.1g,29.6mmol) in ethanol/tetrahydrofuran (7.5mL/15mL) was slowly added dropwise to the reaction solution under ice-cooling, heated under reflux for 2 hours, then cooled to room temperature, filtered, and the filtrate was concentrated. The residue was taken up in 7:3 heptane/ethyl acetate (100mL) to precipitate a solid which was filtered. This procedure was repeated until no more solids were observed after the liquid was concentrated. Purifying by column chromatography to obtain oily substance. The colorless oil was further purified by recrystallization from heptane to give 1.8g of white crystals, yield: 57 percent.
Step k: preparation of (S) -1- (5-fluoro-2-iodophenyl) methanesulfonic acid ethyl ester (intermediate 11)
A solution of intermediate 10(2.2g,8.3mmol) in methyl tert-butyl ether (35mL) was cooled to 0 deg.C, triethylamine (2.3mL,16.6mmol) and methanesulfonyl chloride (0.96mL,12.4mmol) were added dropwise in that order, and the reaction was heated to room temperature and stirred for 3 hours. Filter and wash the solid with ethyl acetate. The filtrate was concentrated to give 3.5g of a pale yellow oil, yield: 80 percent of
Step l: preparation of (R) -3- (1- (5- (5-fluoro-2-iodophenyl) ethoxy) pyridin-2-amine (intermediate 12)
Cesium carbonate (6.5g,20.1mmol) was added to a mixed solvent of 2-amino-3-hydroxypyridine (1.33g,12.1mmol) in methyltetrahydrofuran (60mL) and acetone (30mL), stirred at room temperature for 30 minutes, then heated to 40 ℃, and a solution of intermediate 11(3.44g,80mmol) in methyltetrahydrofuran (30mL) was added dropwise. The reaction was carried out at 80 ℃ for 24 hours. The reaction solution was filtered, the filtrate was concentrated, and column chromatography purification was performed to obtain 1.4g of a white solid, yield: 38 percent of
Step m: preparation of methyl (R) -2- (1- (((2-aminopyridin-3-yl) oxy) ethyl) -4-fluorobenzoate (intermediate 13)
Intermediate 12(2g,5.7mmol) was dissolved in 20mL of methanol and triethylamine (1.54mL,11.3mmol) was added to the solution, followed by Pd (dppf) C12(0.41g,0.57 mmol). The reaction was carried out at 100 ℃ for 16 hours under an atmosphere of carbon monoxide at a pressure of 100 psi. The reaction mixture was filtered, the filtrate was concentrated, and column chromatography purification gave 1.3g of a brown oil, yield: 79 percent.
Step n: preparation of methyl (R) -2- (1- ((2-amino-5-bromopyridin-3-yl) oxy) ethyl) -4-fluorobenzoate (intermediate 14)
Intermediate 13(1.3g,4.5mmol) was dissolved in 20mL acetonitrile and NBS (0.79g,4.5mmol) in acetonitrile (20mL solution) was added slowly at 0 ℃ and the mixture was stirred for 2h after addition was complete. Concentrate to dryness, dissolve the residue with ethyl acetate and wash with 2M aqueous sodium hydroxide and 10% aqueous sodium thiosulfate. Drying, concentration, column chromatography gave 1.2g, yield: 73 percent.
lHNMR(400MHz,DMSO-d6)δ:7.97-7.93(m,1H),7.61-7.51(m,1H),7.54(s,1H),7.29-7.24(m,1H),6.89(s,1H),6.25-6.08(m,3H),3.91(s,3H),1.59(d,J=8.0Hz,3H).
Intermediate 18: preparation of (10R) -7-amino-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecyl-3-carbonitrile
The route is as follows:
Figure BDA0002470516720000121
step o: preparation of methyl (R) -2- (1- ((5-amino-2- (3- ((((tert-butoxycarbonyl) (methyl) amino) methyl ] - -5-cyano-1-methyl-1H-pyrazol-4-) pyridin-3-yl) oxy) ethyl-4-fluorobenzoate (intermediate 15)
Intermediate 14(0.9g,2.4 mmols), intermediate 9(1.0g,3.0 mmols), pinacol diboron ester (0.9g,3.6 mmols), cesium fluoride (1.9g,12.6 mmols) were dissolved in a 9:1 methanol/water (12mL) solution, heated to 60 deg.C, and Pd (dppf) Cl was added under nitrogen protection2A solution of (175mg,0.24mmol) in toluene (1.5mL) was stirred at reflux for 3 h. Adding another part of Pd (dppf) Cl2A solution of (87mg,0.12mmol) in toluene (1.5mL) was stirred at 60 ℃ overnight. After cooling to room temperature, ethyl acetate was added, filtered, and the filtrate was washed with water and brine in sequence, dried, and concentrated. Purification by column chromatography gave 570mg of a yellow oil, yield: and 43 percent.
Step p preparation of (R) -2- (1- ((5-amino-2- (3- ((((tert-butoxycarbonyl) (methyl) amino) methyl ] -5-cyano-1-methyl-1H-pyrazol-4-yl) pyridin-3-yl) oxy) ethyl) -4-fluorobenzoic acid (intermediate 16)
Intermediate 15(2.7g,5mmol) was dissolved in methanol (20mL), a solution of sodium hydroxide (1.0g,25mmol) in water (2mL) was added, and the mixture was stirred at 40 ℃ for 3 hours. The reaction was diluted with water, concentrated to remove methanol, and the aqueous phase was washed with ethyl acetate. The aqueous phase was neutralized with dilute hydrochloric acid, extracted with ethyl acetate, dried, and concentrated to give 2g of a pale yellow solid, yield: 76 percent.
Step q: preparation of (R) -2- (1- ((5-amino-2- (5-cyano-1-methyl-3- ((methylamino) methyl) -1H-pyrazol-4-yl) pyridin-3-yl) oxy) ethyl) -4-fluorobenzoic acid (intermediate 17)
Intermediate 16(960mg,1.82mmol) was dissolved in 4M hydrogen chloride-dioxane solution (15mL) and the reaction was stirred at room temperature for 2.5 h. Concentration gave 700mg of an off-white solid, yield: 83.1 percent.
Step r: preparation of (10R) -7-amino-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (intermediate 18)
Intermediate 17(100mg,0.217mmol) was dissolved in DMF (10mL), DIPEA (42mg,0.325mmol) was added, the temperature was reduced to 0 ℃ and HATU (123.5mg,0.325mmol) was slowly added and reacted at 0 ℃ for 2 hours. Adding the reaction solution into water, extracting with ethyl acetate, washing the organic phase with water, drying, concentrating, and performing column chromatography to obtain 40mg, wherein the yield is as follows: 45 percent.
lHNMR(400MHz,DMSO-d6)δ:7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.19(s,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Intermediate 25: preparation of tert-butyl ((4-bromo-5-cyano-1-deuterated methyl-1H-pyrazol-3-yl) methyl) (cyclopropyl) carbamate
The route is as follows:
Figure BDA0002470516720000141
the synthesis method refers to the synthesis of intermediate 9, wherein iodomethane is replaced by deuterated iodomethane, methylamine is replaced by cyclopropylamine, ESI-MS M/z:358.2/360.2[ M + H ]]+
Intermediate 27: preparation of tert-butyl ((4-bromo-5-cyano-1-deuterated methyl-1H-pyrazol-3-yl) methyl) (deuterated methyl) carbamate
The route is as follows:
Figure BDA0002470516720000142
synthetic methods reference intermediate 25In which cyclopropylamine is replaced with deuterated methylamine, ESI-MS M/z:335.2/337.2[ M + H]+
Intermediate 34: preparation of tert-butyl ((4-bromo-5-cyano-1-cyclopropyl-1H-pyrazol-3-yl) methyl) (deuterated methyl) carbamate
The route is as follows:
Figure BDA0002470516720000151
the synthesis method refers to the synthesis of an intermediate 9, wherein methyl iodide is replaced by bromocyclopropane, methylamine is replaced by deuterated methylamine, ESI-MS M/z is 358.2/360.2[ M + H ]]+
Intermediate 36: preparation of tert-butyl ((4-bromo-5-cyano-1-cyclopropyl-1H-pyrazol-3-yl) methyl) (methyl) carbamate
Figure BDA0002470516720000152
Synthetic methods reference the synthesis of intermediate 25, where cyclopropylamine was replaced with methylamine, ESI-MS M/z:332.2/334.2[ M + H]+
Intermediate 38: preparation of tert-butyl ((4-bromo-5-cyano-1-methyl-1H-pyrazol-3-yl) methyl) (deuterated methyl) carbamate:
the route is as follows:
Figure BDA0002470516720000153
the synthesis method refers to the synthesis of an intermediate 9, wherein methylamine is replaced by deuterated methylamine, ESI-MS M/z is 332.2/334.2[ M + H ]]+
Intermediate 40: preparation of tert-butyl ((4-bromo-5-cyano-1-methyl-1H-pyrazol-3-yl) methyl) (cyclopropyl) carbamate
The route is as follows:
Figure BDA0002470516720000161
synthetic methods reference the synthesis of intermediate 9, where methylamine was replaced with cyclopropylamine, ESI-MS M/z 355.2/357.2[ M + H]+
Example 1: (10R) -7-hydroxymethyl-amino-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 1)
Figure BDA0002470516720000162
Intermediate 18(100mg,0.245mmol) was suspended in water (1mL), dichloromethane (1mL), methanol (1mL), and aqueous formaldehyde (37%, 0.4mL) and tetrabutylammonium fluoride (1M,0.08mL) were added. The reaction mixture was extracted with dichloromethane overnight at room temperature, the organic phase was washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and column purified to give 80mg of a solid in 74.7% yield.
ESI-MS m/z:437.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Example 2: (10R) -7-carbamic acid ethyl ester-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 2)
Figure BDA0002470516720000163
Intermediate 18(100mg,0.245mmol) was dissolved in THF, DIPEA (38mg,0.294mmol) was added, ethyl chloroformate (26.5mg,0.245mmol) was slowly added at 0 ℃, then stirred at room temperature overnight, the reaction was washed with water, extracted with ethyl acetate, the organic phase was dried and spun dry, and the column was purified to 35mg, yield: 29.7 percent.
ESI-MS m/z:479.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.51(s,1H),7.98(s,1H),7.83-7.80(m,1H),7.49-7.47(m,1H),7.21-7.16(m,2H),5.66(q,J=8.0Hz,1H),4.48(d,J=16.0Hz,1H),4.25(d,J=12.0Hz,1H),4.18(q,J=8.0Hz,2H),4.08(s,3H),3.00(s,3H),1.68(d,J=8.0Hz,3H),1.28(t,J=8.0Hz,3H).
Example 3: (10R) -7-Aminocarboxaldehyde-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 3)
Figure BDA0002470516720000171
Intermediate 18(100mg,0.245mmol) was slowly added to formic acid (113mg,2.45mmol) under ice-bath conditions, acetic anhydride (37mg,0.367mmol) was slowly added, and after the addition was completed, the mixture was stirred at room temperature overnight. Diluting the reaction solution with ethyl acetate, washing with water, drying, concentrating, and performing column chromatography to obtain a solid 42mg, wherein the yield is as follows: 39.2 percent.
ESI-MS m/z:435.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:10.42(d,J=12.0Hz,1H),9.23(s,1H),7.89(s,1H),7.81(d,J=8.0Hz,1H),7.50-7.46(m,1H),7.21-7.17(m,2H),5.73(q,J=8.0Hz,1H),4.48(d,J=16.0Hz,1H),4.23(d,J=12.0Hz,1H),4.08(s,3H),3.01(s,3H),1.72(d,J=8.0Hz,3H).
Example 4: (10R) -7-amino-12-fluoro-2-deuterated methyl-10-methyl-16-cyclopropyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 4)
Figure BDA0002470516720000172
Synthetic methods reference the synthesis of intermediate 18, with intermediate 9 replaced intermediate 25.
ESI-MS m/z:436.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.19(s,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),2.30-2.22(m,1H),1.68(d,J=8.0Hz,3H),0.83-0.56(m,4H).
Example 5: (10R) -7-amino-12-fluoro-2, 16-dideuteromethyl-10-methyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 5)
Figure BDA0002470516720000181
Synthetic methods reference the synthesis of intermediate 18, wherein intermediate 9 is replaced with intermediate 27.
ESI-MS m/z:413.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.19(s,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),1.68(d,J=8.0Hz,3H).
Example 6: (10R) -7-amino-12-fluoro-2-cyclopropyl-10-methyl-16-deuterated methyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 6)
Figure BDA0002470516720000182
Synthetic methods reference the synthesis of intermediate 18, wherein intermediate 9 is replaced with intermediate 34.
ESI-MS m/z:436.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.19(s,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),2.32-2.25(m,1H),1.68(d,J=8.0Hz,3H),0.81-0.58(m,4H).
Example 7: (10R) -7-hydroxymethyl-amino-12-fluoro-2, 10-dimethyl-16-deuterated methyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 7)
Figure BDA0002470516720000183
Synthetic methods reference the synthesis of example 1, wherein intermediate 9 is replaced with intermediate 38.
ESI-MS m/z:440.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),1.68(d,J=8.0Hz,3H).
Example 8: (10R) -7-hydroxymethyl-amino-12-fluoro-2, 16-dideuteromethyl-10-methyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 8)
Figure BDA0002470516720000191
Synthetic methods reference the synthesis of example 1, wherein intermediate 9 is replaced with intermediate 27.
ESI-MS m/z:443.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),1.68(d,J=8.0Hz,3H).
Example 9: (10R) -7-hydroxymethyl-amino-12-fluoro-2-deuterated methyl-10, 16-dimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 9)
Figure BDA0002470516720000192
Synthetic methods reference the synthesis of example 1, wherein intermediate 9 is replaced with intermediate 36.
ESI-MS m/z:440.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.65(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Example 10: (10R) -7-Aminomethylphosphonic acid di-tert-butyl-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 10)
Figure BDA0002470516720000193
Dissolving the intermediate 18(100mg,0.246mmol) in 1mL DMF, adding NaH (10mg,0.246mmol) under ice bath, reacting at 0 ℃ for 0.5h after the addition is finished, then adding DMF (0.5mL) of di-tert-butyl chloromethyl phosphate (67mg,0.246mmol) into the reaction solution, reacting at room temperature for 2h, pouring the reaction solution into water, extracting DCM, washing the organic phase with water, washing with saturated sodium chloride, drying with anhydrous sodium sulfate, and purifying the column to obtain 20mg of a solid, wherein the yield is: 13 percent.
ESI-MS m/z:629.4[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.63(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),2.99(s,3H),1.68(d,J=8.0Hz,3H),1.32(s,18H)
Example 11: (10R) -7-Aminophosphoric acid dihydromethyl ester-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathidiazacyclo-3-carbonitrile (Compound 11)
Figure BDA0002470516720000201
Compound 13(100mg,0.159mmol) was dissolved in 1mL of dilute hydrochloric acid (1M) and 1mL of tetrahydrofuran, stirred at room temperature for 1 hour, extracted with dichloromethane, dried and then spin-dried to give a solid 25mg, yield: 30.5 percent.
ESI-MS m/z:517.1[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.34(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Example 12: (10R) -7-Aminophosphoric acid diethyl ester-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathidiazacyclo-3-carbonitrile (Compound 12)
Figure BDA0002470516720000202
The synthesis was as in example 10, replacing di-tert-butyl chloromethyl phosphate with diethyl chloromethyl phosphate.
ESI-MS m/z:573.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),4.03(q,J=8.0Hz,4H),2.99(s,3H),1.68(d,J=8.0Hz,3H),1.41(t,J=8.0Hz,6H).
Example 13: (10R) -7-amino acid dimethyl ester-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 13)
Figure BDA0002470516720000211
The synthesis was as in example 10, but di-tert-butyl chloromethyl phosphate was replaced by dimethyl chloromethyl phosphate.
ESI-MS m/z:545.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.63(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),3.78(s,6H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Example 14: (10R) -7-Methoxymethylmethylamino-12-fluoro-2, 10, 16-trimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecane-3-carbonitrile (Compound 14)
Figure BDA0002470516720000212
Dissolving the intermediate 18(100mg,0.246mmol) in 1mL DMF, adding NaH (10mg,0.246mmol) under ice bath, reacting at 0 ℃ for 0.5h after the addition is finished, then adding bromomethyl methyl ether (30.7mg,0.246mmol), reacting at room temperature for 2h, pouring the reaction liquid into water, extracting with DCM, washing the organic phase with water, washing with saturated sodium chloride, drying with anhydrous sodium sulfate, and purifying with a column to obtain a solid 32mg, yield: 28.9 percent.
ESI-MS m/z:451.2[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),3.32(s,3H),2.99(s,3H),1.68(d,J=8.0Hz,3H).
Example 15: (10R) -7-Aminomethylphosphonic acid di-tert-butyl-12-fluoro-2, 10-dimethyl-16-deuterated methyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathidiazacyclo-3-carbonitrile (Compound 15)
Figure BDA0002470516720000213
The synthesis was as in example 10, with intermediate 9 replacing intermediate 38.
ESI-MS m/z:632.3[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),1.68(d,J=8.0Hz,3H),1.32(s,18H).
Example 16: (10R) -7-Aminomethylphosphonic acid di-tert-butyl-12-fluoro-2-deuterated methyl-10, 16-dimethyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizatetradecyl-3-carbonitrile (Compound 16)
Figure BDA0002470516720000221
The synthesis was as in example 10, with intermediate 9 replacing intermediate 36.
ESI-MS m/z:632.3[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.62(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),2.99(s,3H),1.68(d,J=8.0Hz,3H),1.32(s,18H).
Example 17: (10R) -7-Aminomethylphosphonic acid di-tert-butyl-12-fluoro-2, 10-dimethyl-16-cyclopropyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 17)
Figure BDA0002470516720000222
The synthesis was as in example 10, with intermediate 9 replacing intermediate 40.
ESI-MS m/z:655.3[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.28(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.36(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.04(s,3H),2.30-2.22(m,1H),1.68(d,J=8.0Hz,3H),1.32(s,18H),0.83-0.56(m,4H).
Example 18: (10R) -7-hydroxymethyl-amino-12-fluoro-2, 10-dimethyl-16-cyclopropyl-5-oxo-10, 15,16, 17-tetrahydro-2H-8, 4- (methine bridge) -pyrazolo [4,3-H ] [2,5,11] benzoxathizazepine-3-carbonitrile (Compound 18)
Figure BDA0002470516720000231
Synthetic methods reference the synthesis of example 1, wherein intermediate 9 is replaced with intermediate 40.
ESI-MS m/z:463.3[M+H]+
lHNMR(400MHz,DMSO-d6)δ:9.29(s,1H),7.61-7.58(m,2H),7.48-7.45(m,1H),7.20-7.16(m,1H),6.81(s,1H),6.39(d,J=4.0Hz,2H),5.61(q,J=8.0Hz,1H),4.44(d,J=16.0Hz,1H),4.20(d,J=12.0Hz,1H),4.03(s,3H),2.30-2.22(m,1H),1.67(d,J=8.0Hz,3H),0.83-0.56(m,4H).
Example 19 investigation of ALK inhibitory Effect of novel benzoxadiazacyclotetradecene derivatives
Enzyme analysis of wild type ALK and L1196M mutant ALK
Inhibition of the ALK enzyme by wild-type ALK and L1196M mutant ALK was measured by microfluidic mobility shift analysis. Reactions were performed in 96-well plates in 50 μ L volumes containing preactivated human recombinant wild-type (1.3nM) or L1196M (0.5nM) ALK kinase domain (amino acids 1093 to 1411), 1.5 μ M phosphorus receptor, 5' FAM-KKSRGDYMTMQIG-CONH2(CPCScientific, Sunnyva1e, Calif.), test compound (11-dose three-fold serial dilution, final 2% maple dimethyl) or pure dimethylsulfoxide, 1mM DTT, 0.002% Tween-20 with 5mM magnesium chloride/25 mM Hepes (pH 7.1), preincubated for 20 minutes, initiated by addition of ATP (60 μ M final concentration, km grade). The reaction was incubated at room temperature for 1 hour, stopped by the addition of 0.1M EDTA (pH8), and the extent of reaction (5% to 20% conversion without inhibitor) was determined by separating the fluorescently labeled peptide substrate from the phosphorylated product by LabChip EZ Reader II (Ca1iperLife Sciences, Hopkinton, Mass.) electrophoresis. Kinetic and crystallographic studies confirmed that the inhibitor is ATP competitive. Ki values were calculated by fitting the conversion to an equation for competitive inhibition using non-linear regression (GraphPad Prism, GraphPad software, San Diego, Calif.), and wild-type ATP K was experimentally determinedmATP K of 58. mu.M and L1196M kinasemThe value was 55 μ M. Self-preparing ATThe pase (baculovirus expressed), preactivated for 1 hour at room temperature in the presence of 2mM ATP, 10mM magnesium chloride and 4mM DTT/20mM Hepes (pH 7.5) using autophosphorylation of 16 μ M unactivated enzyme, confirmed complete phosphorylation of the ALK kinase domain (4 phosphates per protein molecule) by Q-TOF mass spectrometry.
Cellular Phospho-ALK (Tyr1604) ELISA for EML 4-ALK:
cell lines:
NIH-3T3EML4-ALK wt v1 and NIH-3T3EML4-ALK v1 Ll196M cells are human stable cell lines. The cells were stored in DMEM (Invitrogen, Car1sbad, CA) medium supplemented with 1% L-glutamine, 1% penicillin and streptomycin, 1. mu.g/mL puromycin, and 10% newborn bovine serum (NCS) in a T-75 flask in a 5% carbon dioxide incubator at 37 ℃.
And (3) analysis:
the cells were washed with PBS, suspended in DMEM medium supplemented with 0.5% NCS and 1% penicillin and streptomycin, seeded into 96-well plates at a density of 20,000 cells/well/100 μ L, and incubated at 37 ℃ in a 5% carbon dioxide incubator. After 20 hours of incubation, 100 μ L of test medium DMEM containing the indicated PF-compound concentration or control group (dimethyl sulfoxide) was added to the plate and incubated for 1 hour with an incubator. The medium was removed, and the lysate containing phosphatase inhibitor and phenylmethanesulfonyl fluoride (PMSF) was added to the wells and shaken at 4 ℃ for 30 minutes to obtain a protein lysate. Phosphorylation of ALK was then assessed using a pathscanphos-ALK (Tyr1604) chemiluminescent sandwich ELISA kit (Cell Signal Technology inc., cat #7020), see below:
Pho-ALK (Tyr1604) rabbit antibody was coated on 96-well micro-plates. 50 μ L of cell lysate was added to the antibody-coated plate and incubated at room temperature for 2 hours. Unbound material was removed by bulk washing with 0.1% Tween20/PBS and ALK mouse mAb was added to detect captured phospho-ALK (Tyr1604) and phospho-ALK fusion proteins. The binding check antibody was then recognized with anti-mouse IgG (HRP enzyme-bound antibody). Finally, a chemiluminescent agent was added and incubated for 10 minutes to allow signal development. The assay plate was read in luminescence mode using an Envision plate reader. Using four parameter analysisMethod for calculating IC by fitting concentration response curve50The value is obtained.
Ki and IC obtained by ALK enzyme analysis and cell Phospho-ALK (Tyr1604) ELISA analysis for WT EML4-ALK and L1196M EML4-ALK50The data are shown in Table 1.
TABLE 1 results of cytostatic Properties of different compounds
Figure BDA0002470516720000241
Figure BDA0002470516720000251
Through the analysis of enzyme activity data of the wild ALK and the L1196M mutant ALK, compounds 4, 5 and 6 show better effects than loratinib on the level of kinase and the level of cells, and compounds 1,2, 3, 7, 8, 9 and 18 show the same performance as loratinib on the level of kinase and the level of cells; although compounds 10, 11, 12, 13, 15,16,17 were slightly weaker than loratinib at the kinase level as phosphate prodrugs, among them, compounds 10, 11, 12, 13, 14 were all precursors of compound 1, compound 15 was a precursor of compound 7, compound 16 was a precursor of compound 9, and compound 17 was a precursor of compound 18, and their effects at the cellular level were comparable to loratinib.
Example 20 exploration of the metabolic stability of novel benzoxadiazacyclotetradecene derivatives
Evaluation of compound stability was performed using human liver microsomes. The liver microsomal enzyme stability of the example compounds was compared to Lorlatinib (PF-06463922).
Measurement System: the metabolic stability of the compounds of the present invention was tested using 1mM NADPH for liver microparticles mixed by men and women. The samples were analyzed using a mass spectrometer. HRMS was used to determine peak area response ratios (peak area corresponding to test compound or control divided by peak area of the analytical internal standard) without running a standard curve. In order to detect all possible metabolites, HRMS scans were performed at the appropriate m/z range.
The measurement conditions were as follows: the assay was performed with one incubation (N ═ 1) v. Test compounds were incubated at 37 ℃ in buffer containing 0.5 mg/ml liver microsomal protein. Reactions were initiated by addition of cofactors and samples taken at 0, 2,4, 8, 16, 24, 36, 48 hours, positive controls (5 μ M testosterone) were incubated in parallel and samples taken at 0, 2,4, 8, 16, 24, 36, 48 hours.
And (3) measuring quality control: the control compound testosterone was performed in parallel to confirm the enzymatic activity of the (liver) microsomes. After the final time point, the addition of NADPH to the reaction mixture was confirmed using fluorimetry. The T1/2 of the control met acceptable internal standards.
The analysis method comprises the following steps:
liquid chromatography column: thermo BDS Hypersil C1830X2.0 mM, 3 μm, with guard column M.P., buffer 25mM formic acid buffer, pH 3.5;
aqueous phase (a): 90% water, 10% buffer;
organic phase (B): 90% acetonitrile, 10% buffer;
flow rate: 300 microliter/min
Automatic sample injector: injection volume 10 microliter
See table 2 for gradient program.
TABLE 2 gradient program
Figure BDA0002470516720000252
Figure BDA0002470516720000261
By using human liver microsomes, examples 4, 5, 6 showed a metabolic half-life of greater than 36 hours, 10, 12, 15,16,17 showed a metabolic half-life of 24-36 hours, significantly greater than 20 hours for Lorlatinib (PF-06463922), and examples 1,2, 3, 7, 8, 9, 11, 13, 14, 18 showed similar metabolic half-lives for Lorlatinib (PF-06463922), as described in the present invention. The results show that the relatively long metabolic half-life makes them potentially useful for lowering therapeutic doses and extending the time interval between administrations.
Enzymatic Activity, IC from Compounds of examples 1-1750Values and metabolic half-life data show that for compounds of the general formula (I), the linker and substituent groups have important effects on the pharmacodynamic properties and metabolic stability of the compounds. While the invention has been illustrated by the foregoing specific embodiments, it is not to be construed as being limited thereby; but that the present invention encompass the generic aspects previously disclosed. Various modifications and embodiments can be made without departing from the spirit and scope of the invention.

Claims (10)

1. A benzoxadiazepitetradecene derivative or its pharmaceutically acceptable salt has a structure shown in general formula (I),
Figure FDA0002470516710000011
wherein when R is1When it is hydrogen, R2、R3Each independently selected from C3-C6Cycloalkyl, deuterated methyl; wherein cycloalkyl may be optionally mono-to pentasubstituted with the same or different substituents selected from halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group;
when R is1Is selected from C1-C4Alkyl radical, C3-C6Cycloalkyl, -CH2OR4、-COOR5、-COR6or-CH2OP(O)OR7OR8When R is2、R3Each independently selected from C1-C4Alkyl, deuterated methyl or C3-C6A cycloalkyl group; wherein said alkyl, cycloalkyl may optionally be mono-to pentasubstituted with the same or different substituents selected from halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group; wherein R is4,R5,R6Are respectively and independently selected from hydrogen and C1-C4Alkyl or C3-C6A cycloalkyl group; r7,R8Each independently selected from hydrogen and C1-C4Alkyl or C3-C6A cycloalkyl group; wherein said alkyl, cycloalkyl may be optionally mono-to pentasubstituted with the following same or different substituents selected from: halogen, trifluoromethyl, cyano, nitro, hydroxy or C1-C4An alkyl group.
2. A benzoxadiazepitetradecene derivative or its pharmaceutically acceptable salt has a structure shown in general formula (I),
Figure FDA0002470516710000012
R1is hydrogen; r2、R3At least one of each is deuterated methyl and the other is selected from C1-C4Alkyl radical, C3-C6Cycloalkyl, deuterated methyl.
3. The benzoxadiazacyclotetradecene derivative or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is4Selected from methyl and hydrogen.
4. The benzoxadiazacyclotetradecene derivative or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is5Selected from hydrogen, methyl, ethyl, isopropyl.
5. The benzoxadiazepidodecene derivative or a pharmaceutically acceptable salt thereof, according to claim 1, R7,R8Each independently selected from hydrogen, methyl, ethyl, isopropyl, tert-butyl.
6. The benzoxadiazepitetradecene derivative according to claim 1, or a pharmaceutically acceptable salt thereof, which is selected from the following compounds:
Figure FDA0002470516710000021
7. the benzoxadiazepitetradecene derivative or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 6, wherein said pharmaceutically acceptable salt is selected from an inorganic salt or an organic salt; wherein the inorganic salt is selected from hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate, and acid phosphate; the organic salt is selected from acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate, maleate, lactate, malate, citrate, tartrate, methanesulfonate, benzenesulfonate, salicylate.
8. A pharmaceutical composition comprising a benzoxadiazacyclotetradecene derivative according to any one of claims 1 to 7 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier and a pharmaceutically acceptable adjuvant.
9. The pharmaceutical composition of claim 8, wherein the pharmaceutically acceptable carrier comprises: microcapsules, microspheres, nanoparticles, liposomes.
10. Use of a benzoxadiazacyclotetradecene derivative according to any one of claims 1 to 7 or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of cancer.
CN202010347120.4A 2020-04-28 2020-04-28 Benzoxadiazatetetradecene derivatives and use thereof Active CN111362967B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010347120.4A CN111362967B (en) 2020-04-28 2020-04-28 Benzoxadiazatetetradecene derivatives and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010347120.4A CN111362967B (en) 2020-04-28 2020-04-28 Benzoxadiazatetetradecene derivatives and use thereof

Publications (2)

Publication Number Publication Date
CN111362967A true CN111362967A (en) 2020-07-03
CN111362967B CN111362967B (en) 2021-09-07

Family

ID=71203781

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010347120.4A Active CN111362967B (en) 2020-04-28 2020-04-28 Benzoxadiazatetetradecene derivatives and use thereof

Country Status (1)

Country Link
CN (1) CN111362967B (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112209879A (en) * 2020-10-19 2021-01-12 河北省药品医疗器械检验研究院 Sildenafil impurity and preparation method and application thereof
CN112724077A (en) * 2020-12-29 2021-04-30 武汉利昌医药科技有限公司 Synthetic method of Laolatinib intermediate
CN115246843A (en) * 2021-04-26 2022-10-28 苏州东南药业股份有限公司 Ten-quaternary fused ring derivatives and application thereof
US11542278B1 (en) 2020-05-05 2023-01-03 Nuvalent, Inc. Heteroaromatic macrocyclic ether chemotherapeutic agents
US11667649B2 (en) 2020-05-05 2023-06-06 Nuvalent, Inc. Heteroaromatic macrocyclic ether chemotherapeutic agents
WO2023245847A1 (en) * 2022-06-22 2023-12-28 中山大学 Use of boric acid small molecule compound in preparation of drugs for enhancing curative effect of immune checkpoint inhibitor and treating leukemia
WO2024036097A1 (en) 2022-08-12 2024-02-15 Nuvalent, Inc. Heteroaromatic macrocyclic ether compounds and isotopologues thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107207528A (en) * 2014-08-20 2017-09-26 苏州韬略生物科技有限公司 Big ring and its application method are substituted as kinase inhibitor
WO2018137679A1 (en) * 2017-01-25 2018-08-02 Teligene Ltd Process for the Preparation of (10R) -7- (2-aminoacetyl) amino-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15, 16, 17-tetrahydro-2H-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotetradecine-3-carbonitrile
WO2019073347A1 (en) * 2017-10-10 2019-04-18 Pfizer Inc. Crystalline form of lorlatinib free base hydrate
CN108699081B (en) * 2016-03-03 2019-10-18 深圳市塔吉瑞生物医药有限公司 A kind of macrocyclic compound and the composition comprising the compound

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107207528A (en) * 2014-08-20 2017-09-26 苏州韬略生物科技有限公司 Big ring and its application method are substituted as kinase inhibitor
CN108699081B (en) * 2016-03-03 2019-10-18 深圳市塔吉瑞生物医药有限公司 A kind of macrocyclic compound and the composition comprising the compound
WO2018137679A1 (en) * 2017-01-25 2018-08-02 Teligene Ltd Process for the Preparation of (10R) -7- (2-aminoacetyl) amino-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15, 16, 17-tetrahydro-2H-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotetradecine-3-carbonitrile
WO2019073347A1 (en) * 2017-10-10 2019-04-18 Pfizer Inc. Crystalline form of lorlatinib free base hydrate

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SHENGQUAN DUAN ET AL.: "Developing an Asymmetric Transfer Hydrogenation Process for (S)-5-Fluoro-3-methylisobenzofuran-1(3H)-one, a Key Intermediate to Lorlatinib", 《ORG. PROCESS RES. DEV》 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11542278B1 (en) 2020-05-05 2023-01-03 Nuvalent, Inc. Heteroaromatic macrocyclic ether chemotherapeutic agents
US11667649B2 (en) 2020-05-05 2023-06-06 Nuvalent, Inc. Heteroaromatic macrocyclic ether chemotherapeutic agents
CN112209879A (en) * 2020-10-19 2021-01-12 河北省药品医疗器械检验研究院 Sildenafil impurity and preparation method and application thereof
CN112724077A (en) * 2020-12-29 2021-04-30 武汉利昌医药科技有限公司 Synthetic method of Laolatinib intermediate
CN112724077B (en) * 2020-12-29 2023-07-11 武汉利昌医药科技有限公司 Synthesis method of loratidine intermediate
CN115246843A (en) * 2021-04-26 2022-10-28 苏州东南药业股份有限公司 Ten-quaternary fused ring derivatives and application thereof
WO2023245847A1 (en) * 2022-06-22 2023-12-28 中山大学 Use of boric acid small molecule compound in preparation of drugs for enhancing curative effect of immune checkpoint inhibitor and treating leukemia
WO2024036097A1 (en) 2022-08-12 2024-02-15 Nuvalent, Inc. Heteroaromatic macrocyclic ether compounds and isotopologues thereof

Also Published As

Publication number Publication date
CN111362967B (en) 2021-09-07

Similar Documents

Publication Publication Date Title
CN111362967B (en) Benzoxadiazatetetradecene derivatives and use thereof
TW202200562A (en) Quinoxaline dione derivative as irreversible inhibitor of KRAS G12C mutant protein
CN112608318B (en) Compound as protein kinase inhibitor and application thereof
CN110407856B (en) Macrocyclic compound and composition containing same
JP2022538917A (en) Heterocyclic compounds as BET inhibitors
JP7077323B2 (en) Quinazoline compound and its preparation method, use and pharmaceutical composition
CN115043842A (en) Amino-substituted bicyclic inhibitor and preparation method and application thereof
US20220289716A1 (en) Crystalline forms of a cd73 inhibitor
CN110520416B (en) Polysubstituted pyridone derivative, preparation method and medical application thereof
WO2020038433A1 (en) Egfr kinase inhibitor, preparation method therefor, and application thereof
WO2022083657A1 (en) Substituted benzo or pyridopyrimidine amine inhibitor, preparation method therefor, and application thereof
CN114885607B (en) Quinolinylphosphine oxide compounds, compositions and uses thereof
CN110938104A (en) Cyclic dinucleotide analogs, pharmaceutical compositions and uses thereof
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN114437116A (en) Heterocyclic compound and preparation method, pharmaceutical composition and application thereof
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
KR20230002706A (en) Crystalline RET inhibitors
CN110066272B (en) Substituted benzo [ d ] imidazoles and pharmaceutical compositions thereof
CN112805287B (en) BTK inhibitors containing furo [3,4-b ] pyrroles
CN114437077A (en) Compounds useful as kinase inhibitors and uses thereof
CN112142747A (en) Pyrazolone pyrimidine compound, preparation method and application thereof
JP2023036957A (en) Diarylpyrazole compound, composition comprising the compound, and use thereof
JP2024516194A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
CN115368378A (en) Substituted macrocyclic compounds, compositions containing the same and uses thereof
WO2021057867A1 (en) Class of cdk inhibitor based on organic arsine, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant