WO2023217064A1 - 一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用 - Google Patents

一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用 Download PDF

Info

Publication number
WO2023217064A1
WO2023217064A1 PCT/CN2023/092670 CN2023092670W WO2023217064A1 WO 2023217064 A1 WO2023217064 A1 WO 2023217064A1 CN 2023092670 W CN2023092670 W CN 2023092670W WO 2023217064 A1 WO2023217064 A1 WO 2023217064A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
hydrogen
alkyl
hydroxyl
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2023/092670
Other languages
English (en)
French (fr)
Inventor
蒋闪军
钱刚
陈锋
王贵涛
张楠
黄保华
Original Assignee
同宜医药(苏州)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 同宜医药(苏州)有限公司 filed Critical 同宜医药(苏州)有限公司
Priority to CN202380022412.2A priority Critical patent/CN118696048A/zh
Publication of WO2023217064A1 publication Critical patent/WO2023217064A1/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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings

Definitions

  • the invention belongs to the field of medicinal chemistry, and specifically relates to a camptothecin derivative, antibody-drug conjugates and pharmaceutical compositions obtained based on the derivative, and applications in the medical field.
  • Chemotherapy is one of the important treatments for cancer, but many chemotherapy drugs have no specific recognition function between tumor cells and normal cells, so they can cause severe systemic toxicity.
  • Antibody-drug conjugates can specifically recognize ligands of tumor targets and accurately deliver chemotherapy drugs to disease sites, reducing toxic damage to healthy tissues.
  • Antibody-drug conjugate is a representative of conjugated drugs at this stage. It generally consists of three parts: antibody or antibody ligand (antibody, Ab), small molecule cytotoxic drug (payload) and linkers that couple antibodies and drugs. Among them, as anti-tumor small molecule drugs, camptothecins (CPTs) can exhibit anti-tumor effects by inhibiting DNA topoisomerase I (TOP1). As a representative compound, exatecan developed by Daiichi Sankyo was initially used as a chemotherapy drug alone and has been advanced to clinical phase III. Its main indications are bone cancer, prostate cancer, breast cancer, pancreatic cancer, etc. However, due to severe side effects and narrow therapeutic window, directly administered ixotecan failed to be successfully launched.
  • Daiichi Sankyo developed ixotecan into an ADC and has successfully launched it on the market.
  • the first marketed ADC using ixotecan as a toxin is trastuzumab deruxtecan (DS-8201), which uses a single antibody to connect 8 toxins and mainly targets the HER2 target.
  • DS-8201 trastuzumab deruxtecan
  • ADCs using ixotecan as toxin designed for other targets such as Trop2 had to reduce the drug/antibody ratio (DAR) to 4 due to safety issues. The reduction in the number of drugs will undoubtedly reduce the therapeutic index of ADC.
  • camptothecin drugs and ADCs based on them such as large side effects, narrow therapeutic window, and low safety for individual targets
  • the present invention discovered better camptothecin derivatives and improved Its safety and effectiveness in ADC applications were confirmed, and an anti-tumor conjugate drug with excellent efficacy was obtained.
  • the invention provides a compound represented by formula I or a pharmaceutically acceptable salt, N-oxide, solvate, stereoisomer, tautomer, isotope label, metabolite or prodrug thereof ,in
  • X is -O-, -N(R 4 )-, C 3 -C 10 cycloalkylene or 3 to 10 membered heterocycloalkylene;
  • Y is hydroxyl, mercapto, amino, C 3 -C 10 cycloalkyl or 3 to 10 membered heterocycloalkyl;
  • R 1 is hydrogen, C 1 -C 10 alkyl or C 1 -C 10 haloalkyl
  • each R 2 is independently hydrogen, C 1 -C 10 alkyl or C 1 -C 10 haloalkyl;
  • Each R 3 is independently hydrogen, hydroxyl, amino, C 1 -C 10 alkyl or C 1 -C 10 haloalkyl; or two R 3 connected to the same carbon atom and the carbon atom to which it is connected jointly form Carbonyl group, C 3 -C 10 cycloalkylene group or 3 to 10 membered heterocycloalkylene group;
  • R 4 is hydrogen, C 1 -C 10 alkyl or C 1 -C 10 haloalkyl
  • k, l and m are each independently any integer from 0 to 5;
  • n is any integer from 1 to 5;
  • X is -O-, -NH-, cyclopropylene, cyclobutylene, cyclopentylene or cyclohexylene;
  • Y is hydroxyl, mercapto, amino, tetrahydropyrrolyl, tetrahydrofuryl, tetrahydrothienyl, piperidinyl, pyranyl or thiopyranyl;
  • R 1 is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
  • each R 2 is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
  • Each R 3 is independently hydrogen, hydroxyl, amino, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl; or, two connecting the same carbon atom R 3 and the carbon atom to which it is connected together form carbonyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydropyrrolyl, tetrahydrofuryl, tetrahydrothienyl, piperidyl, pyranyl or thiopyranyl base;
  • R 4 is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
  • k 0 or 1
  • l 0, 1, 2;
  • n 0 or 1
  • n 1, 2 or 3;
  • Y is not an amino group.
  • the compound of formula I is represented by formula IA, wherein
  • X is -O-, -N(R 4 )- or C 3 -C 10 cycloalkylene, preferably -O-, -N(R 4 )- or C 3 -C 6 cycloalkylene, more preferably - O-, -N(R 4 )- or C 4 -C 6 cycloalkylene;
  • Y is hydroxyl, mercapto or amino, preferably hydroxyl
  • R 1 is hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • each R 2 is independently hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • Each R 3 is independently hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • R 4 is hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • l is 0, 1, 2 or 3, preferably 0, 1 or 2;
  • n is 1, 2, 3 or 4, preferably 1, 2 or 3.
  • X is C 3 -C 10 cycloalkylene, preferably C 3 -C 6 cycloalkylene, more preferably C 4 -C 6 cycloalkylene;
  • Y is hydroxyl ;
  • R 1 is hydrogen; each R 3 is independently hydrogen; l is 0; n is 1, 2 or 3.
  • Y is hydroxyl, mercapto, amino or 3 to 10-membered heterocycloalkyl, preferably hydroxyl, mercapto, amino or 3 to 6-membered heterocycloalkyl, more preferably hydroxyl, mercapto, amino or 3 to 6-membered heterocycloalkyl;
  • R 1 is hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • Each R 3 is independently hydrogen, hydroxyl, amino or C 1 -C 10 alkyl, preferably hydrogen, hydroxyl, amino or C 1 -C 4 Alkyl group; or, two R 3 connected to the same carbon atom and the carbon atom to which they are connected together form a carbonyl group;
  • n is 1, 2, 3 or 4, preferably 1, 2 or 3;
  • n 1 and R 1 and R 3 are both hydrogen, Y is not amino;
  • Y is hydroxyl, mercapto or amino;
  • n is 1, 2 or 3.
  • the compound of formula I is represented by formula IC, wherein
  • X is -O- or -N(R 4 )-;
  • Y is hydroxyl, mercapto or amino, preferably hydroxyl
  • Each R 3 is independently hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • R 4 is hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen;
  • n 0 or 1
  • n 1, 2, 3 or 4, preferably 2 or 3.
  • X is -O- or -NH-; Y is hydroxyl; each R 3 is independently hydrogen or C 1 -C 10 alkyl, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen; m is 0 or 1; n is 2 or 3.
  • the present invention provides the following compounds or pharmaceutically acceptable salts, N-oxides, solvates, stereoisomers, tautomers, isotopic labels, metabolites or prodrugs thereof.
  • the present invention provides the compounds described in the ⁇ First Aspect> or the ⁇ Second Aspect> or their pharmaceutically acceptable salts, N-oxides, solvates, stereoisomers, tautomers, and isotopic labels.
  • the present invention provides an antibody-drug conjugate, which contains the compound described in the ⁇ first aspect> or the ⁇ second aspect> or a pharmaceutically acceptable salt, N-oxide, solvate, stereoisomer thereof Conforms, tautomers, isotopic labels, metabolites or prodrugs and antibodies or antibody-based ligands coupled thereto.
  • the antibody or antibody ligand is selected from the group consisting of anti-EGFRvIII antibody, anti-DLL-3 antibody, anti-PSMA antibody, anti-CD70 antibody, anti-MUC16 antibody, anti-ENPP3 antibody, anti-TDGF1 antibody, anti-ETBR antibody, and anti-MSLN antibody.
  • the present invention provides a pharmaceutical composition, which contains the compound described in the ⁇ first aspect> or the ⁇ second aspect> or a pharmaceutically acceptable salt, N-oxide, solvate, stereoisomer, Tautomers, isotopic labels, metabolites or prodrugs or antibody-drug conjugates described in ⁇ Fourth Aspect>.
  • the pharmaceutical composition further contains at least one pharmaceutically acceptable excipient.
  • the present invention provides the compounds described in the ⁇ First Aspect> or the ⁇ Second Aspect> or their pharmaceutically acceptable salts, N-oxides, solvates, stereoisomers, tautomers, and isotopic labels.
  • the antibody, metabolite or prodrug or the antibody-drug conjugate described in the ⁇ fourth aspect> or the pharmaceutical composition described in the ⁇ fifth aspect> is prepared for prevention and/or treatment at least partially by DNA topology Use in medicines for diseases or conditions mediated by isomerase I.
  • the present invention provides the compounds described in the ⁇ First Aspect> or the ⁇ Second Aspect> or their pharmaceutically acceptable salts, N-oxides, solvates, stereoisomers, tautomers, and isotopic labels.
  • the present invention provides a method for preventing and/or treating a disease or condition mediated at least in part by DNA topoisomerase I, comprising:
  • a prophylactically and/or therapeutically effective amount of the compound described in the ⁇ first aspect> or the ⁇ second aspect> or a pharmaceutically acceptable salt, N-oxide, solvate, stereoisomer, tautomer thereof is administered to an individual in need thereof.
  • the disease or condition mediated at least in part by DNA topoisomerase I is cancer, autoimmune disease, Cardiovascular, metabolic or neurological diseases.
  • the cancer is selected from lung cancer, kidney cancer, colon cancer, rectal cancer, ovarian cancer, breast cancer, liver cancer, Bladder cancer, prostate cancer, gastric cancer, cervical cancer, uterine cancer, colorectal cancer, esophageal cancer, endometrial cancer, salivary gland cancer, thyroid cancer, pancreatic cancer and bone cancer;
  • the autoimmune disease is selected from the group consisting of connective tissue diseases, Systemic sclerosis, rheumatoid arthritis and systemic lupus erythematosus;
  • the cardiovascular disease is selected from angina pectoris, myocardial infarction, stroke, heart attack, hypertensive heart disease, rheumatic heart disease heart disease, cardiomyopathy, cardiac arrhythmia and congenital heart disease;
  • the metabolic disease is selected from diabetes, gout, obesity, hypoglycemia, hyperglycemia and dyslipidemia;
  • the neurological disease is selected from diabetes, gout, obesity, hypoglycemia, hyperglycemia and
  • camptothecin derivatives On the basis of a comprehensive understanding of the structure-activity relationship (SAR) of camptothecin derivatives, the present invention designs and synthesizes a series of camptothecin derivatives with significant anti-tumor activity. Through cell experiments, it was found that this type of small molecule compounds can exhibit excellent anti-tumor activity.
  • SAR structure-activity relationship
  • alkyl refers to a saturated monovalent aliphatic hydrocarbon group, including straight-chain and branched groups of 1 to 10 carbon atoms, for example, 1 to 8 carbon atoms, 1 to 6 carbon atoms, etc. Carbon atoms or straight and branched chain groups of 1 to 4 carbon atoms.
  • C 1 -C 10 alkyl examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, and n-pentyl, n-hexyl base, n-heptyl, n-octyl, n-nonyl, n-decyl and various branched isomers, etc.
  • cycloalkyl refers to a saturated or partially unsaturated, monocyclic or polycyclic monovalent aliphatic hydrocarbon group, including 3 to 10 ring atoms, such as 3 to 6 ring atoms ( That is, 3 to 6 membered rings).
  • C 3 -C 10 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, and moieties thereof. Saturated derivatized groups, etc.
  • Cycloalkylene refers to a divalent aliphatic hydrocarbon group formed by removing one hydrogen atom on a ring atom from “cycloalkyl”.
  • heterocycloalkyl refers to a saturated or partially unsaturated, monocyclic or polycyclic monovalent aliphatic group, including 3 to 10 ring atoms, such as 3 to 6 ring atoms. , one or more ring atoms are heteroatoms selected from N, O or S(O) m (where m is 0, 1 or 2), and the remaining ring atoms are C.
  • Preferred heterocycloalkyl groups include 3 to 6 ring atoms, of which 1 to 3 ring atoms are heteroatoms; more preferred heterocycloalkyl groups include 4 to 6 ring atoms, of which 1 to 3 ring atoms are heteroatoms.
  • heterocycloalkyl groups include 5 or 6 ring atoms, 1 or 2 of which are heteroatoms.
  • Examples of "3- to 10-membered heterocycloalkyl” include, but are not limited to, oxetanyl, thiiryl, aziridinyl, oxetanyl, thietanyl, azetidinyl, tetrahydrofuranyl , tetrahydrothiophenyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, etc.
  • Heterocycloalkylene refers to a divalent aliphatic group formed by removing one hydrogen atom on the ring atom from “heterocycloalkyl”.
  • halogen means fluorine, chlorine, bromine and iodine, preferably fluorine, chlorine and bromine.
  • haloalkyl refers to a monovalent aliphatic hydrocarbon group formed after the "alkyl” is substituted by at least one halogen atom that is the same as or different from each other, including straight-chain and branched-chain groups of 1 to 10 carbon atoms.
  • Groups for example, may be straight-chain and branched groups of 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms.
  • C 1 -C 10 haloalkyl examples include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 1-fluoroethyl, 2-fluoroethyl, 1,1-difluoroethyl, 1,2-difluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, etc.
  • hydroxyl refers to the "-OH” group.
  • mercapto refers to the "-SH” group.
  • amino refers to the “-NH 2 " group.
  • pharmaceutically acceptable salts refer to salts prepared from compounds of the present invention and relatively non-toxic acids or bases.
  • base addition salts can be obtained by contacting the free form with a sufficient amount of base in pure solution or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include, but are not limited to, sodium salts, potassium salts, ammonium salts, calcium salts, magnesium salts, Organic amine salts or similar salts.
  • acid addition salts can be obtained by contacting the free form with a sufficient amount of acid in pure solution or in a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include, but are not limited to, inorganic acid salts (e.g., hydrochlorides, hydrobromides, hydroiodates, nitrates, carbonates, bicarbonates, phosphates , monohydrogen phosphate, dihydrogen phosphate, phosphite, sulfate, hydrogen sulfate, etc.), organic acid salts (such as acetate, propionate, isobutyrate, malonate, succinate , suberate, maleate, fumarate, citrate, tartrate, lactate, mandelate, benzoate, phthalate, methanesulfonate, benzene sulfonate acid salt, p-toluenesul
  • inorganic acid salts e.g., hydrochlorides, hydrobromides, hydroio
  • antibody-drug conjugate refers to a compound formed by covalently linking a biologically active small molecule drug to a macromolecular antibody (or antibody ligand) through a specific chemical linking fragment, wherein the small molecule drug is Molecular drugs exert pharmacological activity, while macromolecular antibodies mainly play a targeted delivery role, and some macromolecular antibodies also exert synergistic drug effects.
  • pharmaceutical composition refers to a pharmaceutically acceptable composition, which contains a small molecule drug as an active pharmaceutical ingredient (API), a macromolecular antibody (or antibody ligand), or a coupling of the two. substances, and other components (such as pharmaceutically acceptable excipients).
  • API active pharmaceutical ingredient
  • pharmaceutical compositions may be prepared using any method known to those skilled in the art. For example, conventional mixing, dissolving, granulating, emulsifying, grinding, encapsulating, embedding and/or lyophilizing processes.
  • auxiliary materials refer to auxiliary materials widely used in the field of pharmaceutical production.
  • the main purpose of using excipients is to provide a pharmaceutical composition that is safe to use, stable in nature and/or has specific functionality, and also to provide a method so that after the drug is administered to the subject, the active ingredient can be used in the desired manner. rate dissolution, or promote effective absorption of the active ingredient in the subject to whom it is administered.
  • Pharmaceutically acceptable excipients may be inert fillers or functional ingredients that provide a certain function for the pharmaceutical composition (such as stabilizing the overall pH value of the composition or preventing the degradation of the active ingredients in the composition).
  • “pharmaceutically acceptable excipients” include, but are not limited to, binders, suspending agents, emulsifiers, diluents (or fillers), granulating agents, adhesives, disintegrants, lubricants, anti-adhesive agents, Glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, enhancers, adsorbents, buffers, chelating agents, preservatives, colorants, flavoring agents, sweeteners, etc.
  • the compounds of the present invention or their pharmaceutically acceptable salts, N-oxides, solvates, stereoisomers, tautomers, isotopic labels, metabolites or prodrugs and antibody-drug conjugations based thereon
  • Both the drug and the pharmaceutical composition have excellent tumor cell killing effect, can be used as anti-tumor drugs to prevent and/or treat cancer, and have good clinical applications and medical uses.
  • the starting materials used in the present invention can be synthesized by methods known in the art, or purchased through conventional commercial means.
  • the isolation and purification of the compounds of the present invention can be achieved by methods well known to those skilled in the art, including but not limited to column chromatography (CC), high-performance liquid chromatography (HPLC), ultra-high performance liquid chromatography (UPLC), etc. .
  • the structural identification of the compounds of the present invention can be achieved by methods well known to those skilled in the art, including but not limited to nuclear magnetic resonance (NMR), mass spectrometry (MS), etc.
  • Example 1 N-((1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15 -Hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl)-4-(hydroxymethyl cyclohexane-1-carboxamide (compound 6A)
  • the target compound (81.70 mg, purity: 95.37%) was obtained.
  • the target compound (323.50 mg, purity: 95.98%) was obtained as a brown-green solid.
  • the target compound 50.4 mg, purity: 96.72%) was obtained as a brown solid.
  • Example 8 1-((1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15 -Hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl)-3-(2- Hydroxyethyl)urea (Compound 11A)
  • Compound 16A can be prepared using a similar method.
  • Example 12 N-((1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15 -Hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl)-4-(2- Hydroxyethyl)cyclohexane-1-carboxamide (Compound 5A)
  • Example 14 N-((1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15 -Hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl)-3-(3- Hydroxypropyl)cyclobutane-1-carboxamide (Compound 17A)
  • Dissolve compound 1 (5.00g, 35.6mmol, 1.00eq) in anhydrous THF (50.0mL), control the temperature to -10°C, add BH 3 .THF (1M, 35.6mL, 1.00eq) dropwise, and raise to 20°C and stir for 2 hours.
  • the reaction solution was cooled to -20°C, methanol (50.0 mL) was added and stirred for 15 minutes, then NaOH aqueous solution (10%, 7.13g, 17.8mmol, 0.500eq) and H 2 O 2 (7.70g, 67.9mmol, 6.53 mL, 1.90eq), raised to 20°C and stirred for 2 hours.
  • PCC pyridinium chlorochromate, 3.41g, 15.8mmol, 1.25eq
  • diatomaceous earth 3.41g
  • PCC pyridinium chlorochromate, 3.41g, 15.8mmol, 1.25eq
  • compound 2 (2.00g, 12.6mmol, 1.00eq) was added at 0°C.
  • DCM DCM (20.0 mL)
  • Compound 19 can be prepared using a similar method.
  • SK-OV-3 commercially available
  • 293T-FOLR1 constructed by the method described below
  • 293T commercially available
  • SK-BR-3 commercially available
  • Dx ixotecan, CAS: 171335-80-1, commercially available
  • Construction of cell model 293T-FOLR1 Construct a recombinant viral vector and recombinant virus particles containing the FR ⁇ encoding gene by expressing the FR ⁇ gene sequence, and then stably transfer it into the 293T cell line. By reducing the concentration in the culture medium during the transfection process serum concentration, add polybrene, and adjust the 5MOI value to improve virus transfection efficiency, and obtain a cell model 293T-FOLR1 that highly expresses FR ⁇ .
  • the next day use complete culture medium to prepare a working solution in a 1.5 mL EP tube.
  • the starting concentration is 10 ⁇ M and diluted 5 times for a total of 9 concentrations.
  • After the preparation is completed add it to a 96-well plate, 50 ⁇ l/well, in triplicate wells. Place the cell plate into a 37°C, 5% CO2 incubator and continue culturing for 72 hours.
  • CCK8 cell viability detection reagent (Abbkine, KTC011001), 15 ⁇ l/well, shake and mix well, let it stand in a dark place for 70 minutes, use the luminescence program of the microplate reader for detection, and use GraphPad Prims
  • the software calculates the IC 50 value, and the experimental results are shown in the table below. Note: “-” indicates that no relevant measurement has been performed.

Landscapes

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

Abstract

本发明属于药物化学领域,涉及一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用。该衍生物的结构如式(I)所示,具有良好的DNA拓扑异构酶(I)抑制活性以及肿瘤细胞杀伤作用,因此可用于预防和/或治疗至少部分与TOP1有关的疾病(如癌症),极具开发前景。

Description

一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用
相关申请的交叉引用
本发明要求2022年05月09日在中国提交的,名称为“一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用”、申请号为202210503019.2的发明专利申请和2023年04月24日在中国提交的,名称为“一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用”、申请号为202310447872.1的发明专利申请的优先权,通过引用的方式将该专利申请的全部内容并入本文。
技术领域
本发明属于药物化学领域,具体涉及一种喜树碱衍生物,以该衍生物为基础而得到的抗体-药物偶联物和药物组合物,以及在医药领域中的应用。
背景技术
化疗是癌症的重要治疗手段之一,但是很多化疗药物对于肿瘤细胞和正常细胞没有特异性识别功能,因此会导致严重的全身系统性毒性。而抗体-药物偶联物则能够特异性识别肿瘤靶点的配体,并将化疗药物精准递送至疾病部位,减少对于健康组织的毒性损伤。
抗体-药物偶联物(antibody-drug conjugate,ADC)是现阶段偶联药物中的代表,一般由三部分组成:抗体或抗体类配体(antibody,Ab)、小分子细胞毒药物(payload)以及将抗体和药物偶联起来的连接子(linker)。其中,作为抗肿瘤小分子药物,喜树碱类药物(camptothecins,CPTs)可通过抑制DNA拓扑异构酶I(TOP1)而呈现抗肿瘤作用。作为代表性化合物,由第一三共公司开发的伊喜替康(exatecan)前期作为化疗药物单独使用已推进至临床三期,主要适应症为骨癌、前列腺癌、乳腺癌、胰腺癌等,但因为副作用大、治疗窗窄,直接给药的伊喜替康最终未能成功上市。
为克服伊喜替康直接给药的副作用,第一三共公司将伊喜替康开发成为ADC来使用,并已成功上市。第一个上市的以伊喜替康为毒素的ADC为trastuzumab deruxtecan(DS-8201),其采用单个抗体连接8个毒素的方式,主要针对HER2靶点。但是,在后续临床研究中,针对其他靶点(如Trop2)设计的以伊喜替康为毒素的ADC却因为安全性问题而不得已将药物/抗体比率(DAR)降低至4。而药物数目的减少无疑会降低ADC的治疗指数。
发明内容
发明要解决的问题
针对目前直接给药的喜树碱类药物以及基于其的ADC存在副作用大、治疗窗窄、对于个别靶点的安全性低等问题,本发明发现了更优的喜树碱类衍生物,提高了其在ADC应用中的安全性、有效性,得到具有优异疗效的抗肿瘤偶联药物。
用于解决问题的方案
<第一方面>
本发明提供了一种如式I所示的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其中
若存在,X为-O-、-N(R4)-、C3-C10亚环烷基或3至10元亚杂环烷基;
Y为羟基、巯基、氨基、C3-C10环烷基或3至10元杂环烷基;
若存在,R1为氢、C1-C10烷基或C1-C10卤代烷基;
若存在,每一个R2各自独立地为氢、C1-C10烷基或C1-C10卤代烷基;
每一个R3各自独立地为氢、羟基、氨基、C1-C10烷基或C1-C10卤代烷基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基、C3-C10亚环烷基或3至10元亚杂环烷基;
若存在,R4为氢、C1-C10烷基或C1-C10卤代烷基;
k、l和m各自独立地为0至5中的任一整数;
n为1至5中的任一整数;
并且,当k为1,l和m均为0,n为1,R1和R3均为氢时,Y不为氨基;
所述杂环烷基和亚杂环烷基各自独立地包含1至4个杂原子或杂原子团,所述杂原子或杂原子团各自独立地为C(=O)、O、S、S(=O)、S(=O)2、N或NH。
优选地,在所述式I化合物中,当k和l均为0,m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
更优选地,在所述式I化合物中,
若存在,X为-O-、-NH-、亚环丙基、亚环丁基、亚环戊基或亚环己基;
Y为羟基、巯基、氨基、四氢吡咯基、四氢呋喃基、四氢噻吩基、哌啶基、吡喃基或噻喃基;
若存在,R1为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
若存在,每一个R2各自独立地为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
每一个R3各自独立地为氢、羟基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基、环丙基、环丁基、环戊基、环己基、四氢吡咯基、四氢呋喃基、四氢噻吩基、哌啶基、吡喃基或噻喃基;
若存在,R4为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
k为0或1;
l为0、1、2;
m为0或1;
n为1、2或3;
并且,当k为1,l和m均为0,n为1,R1和R3均为氢时,Y不为氨基。
进一步优选地,在所述式I化合物中,当k和l均为0,m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
或者,优选地,所述式I化合物如式IA所示,其中
X为-O-、-N(R4)-或C3-C10亚环烷基,优选-O-、-N(R4)-或C3-C6亚环烷基,更优选-O-、-N(R4)-或C4-C6亚环烷基;
Y为羟基、巯基或氨基,优选羟基;
R1为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
若存在,每一个R2各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
若存在,R4为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
l为0、1、2或3,优选0、1或2;
n为1、2、3或4,优选1、2或3。
更优选地,在所述式IA化合物中,X为C3-C10亚环烷基,优选C3-C6亚环烷基,更优选C4-C6亚环烷基;Y为羟基;R1为氢;每一个R3各自独立地为氢;l为0;n为1、2或3。
或者,更优选地,在所述式IA化合物中,X为-O-;Y为羟基;R1为氢;每一个R2各自独立地为氢;每一个R3各自独立地为氢;l为1或2;n为1或2。
或者,更优选地,在所述式IA化合物中,X为-NH-;Y为羟基;R1为氢;每一个R3各自独立地为氢;l为0;n为1、2或3。
或者,优选地,所述式I化合物如式IB所示,其中
Y为羟基、巯基、氨基或3至10元杂环烷基,优选羟基、巯基、氨基或3至6元杂环烷基,更优选羟基、巯基、氨基或3至6元杂环烷基;
R1为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
每一个R3各自独立地为氢、羟基、氨基或C1-C10烷基,优选氢、羟基、氨基或C1-C4 烷基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基;
n为1、2、3或4,优选1、2或3;
并且,当n为1,R1和R3均为氢时,Y不为氨基;
所述杂环烷基包含1至2个杂原子或杂原子团,所述杂原子或杂原子团各自独立地为O、S、S(=O)、S(=O)2、N或NH,优选O、S、N或NH。
更优选地,在所述式IB化合物中,Y为羟基、巯基或氨基;R1为氢;每一个R3各自独立地为氢、羟基或C1-C10烷基,优选氢、羟基或C1-C4烷基;优选地,每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基;n为1、2或3。
或者,优选地,所述式I化合物如式IC所示,其中
若存在,X为-O-或-N(R4)-;
Y为羟基、巯基或氨基,优选羟基;
每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
若存在,R4为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
m为0或1;
n为1、2、3或4,优选2或3。
更优选地,在所述式IC化合物中,若存在,X为-O-或-NH-;Y为羟基;每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;m为0或1;n为2或3。
进一步优选地,在所述式IC化合物中,当m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
<第二方面>
本发明提供了下列化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药。




<第三方面>
本发明提供了<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药在包含其的药物偶联物中作为细胞毒药物的用途。
<第四方面>
本发明提供了一种抗体-药物偶联物,其包含<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药以及与之偶联的抗体或抗体类配体。
优选地,所述抗体或抗体类配体选自抗EGFRvIII抗体、抗DLL-3抗体、抗PSMA抗体、抗CD70抗体、抗MUC16抗体、抗ENPP3抗体、抗TDGF1抗体、抗ETBR抗体、抗MSLN抗体、抗TIM-1抗体、抗LRRC15抗体、抗LIV-1抗体、抗CanAg/AFP抗体、抗cladin 18.2抗体、抗Mesothelin抗体、抗HER2(ErbB2)抗体、抗EGFR抗体、抗c-MET抗体、抗SLITRK6抗体、抗KIT/CD117抗体、抗STEAP1抗体、抗SLAMF7/CS1抗体、抗NaPi2B/SLC34A2抗体、抗GPNMB抗体、抗HER3(ErbB3)抗体、抗MUC1/CD227抗体、抗AXL抗体、抗CD166抗体、抗B7-H3(CD276)抗体、抗PTK7/CCK4抗体、抗PRLR抗体、抗EFNA4抗体、抗5T4抗体、抗NOTCH3抗体、抗Nectin 4抗体、抗TROP-2抗体、抗CD142抗体、抗CA6抗体、抗GPR20抗体、抗CD174抗体、抗CD71抗体、抗EphA2抗体、抗LYPD3抗体、抗FGFR2抗体、抗FGFR3抗体、抗FRα抗体、抗CEACAMs抗体、抗GCC抗体、抗Integrin Av抗体、抗CAIX抗体、抗P-cadherin抗体、抗GD3抗体、抗Cadherin 6抗体、抗LAMP1抗体、抗FLT3抗体、抗BCMA抗体、抗CD79b抗体、抗CD19抗体、抗CD33抗体、抗CD56抗体、抗CD74抗体、抗CD22抗体、抗CD30抗体、抗CD37抗体、抗CD138抗体、抗CD352抗体、抗CD25抗体、抗CD123抗体或抗CD47抗体。
<第五方面>
本发明提供了一种药物组合物,其包含<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者<第四方面>中所述的抗体-药物偶联物。
优选地,所述药物组合物还包含至少一种药学上可接受的辅料。
<第六方面>
本发明提供了<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者<第四方面>中所述的抗体-药物偶联物或者<第五方面>中所述的药物组合物在制备用于预防和/或治疗至少部分由DNA拓扑异构酶I介导的疾病或病症的药物中的应用。
<第七方面>
本发明提供了<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者<第四方面>中所述的抗体-药物偶联物或者<第五方面>中所述的药物组合物,其用于预防和/或治疗至少部分由DNA拓扑异构酶I介导的疾病或病症。
<第八方面>
本发明提供了一种用于预防和/或治疗至少部分由DNA拓扑异构酶I介导的疾病或病症的方法,其包括:
将预防和/或治疗有效量的<第一方面>或<第二方面>中所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者<第四方面>中所述的抗体-药物偶联物或者<第五方面>中所述的药物组合物施用于对其有需要的个体。
优选地,在<第六方面>、<第七方面>和/或<第八方面>中,所述至少部分由DNA拓扑异构酶I介导的疾病或病症为癌症、自身免疫性疾病、心血管疾病、代谢性疾病或神经性疾病。
更优选地,在<第六方面>、<第七方面>和/或<第八方面>中,所述癌症选自肺癌、肾癌、结肠癌、直肠癌、卵巢癌、乳腺癌、肝癌、膀胱癌、前列腺癌、胃癌、宫颈癌、子宫癌、结直肠癌、食道癌、子宫内膜癌、唾液腺癌、甲状腺癌、胰腺癌和骨癌;所述自身免疫性疾病选自结缔组织病、系统性硬化症、类风湿性关节炎和系统性红斑狼疮;所述心血管疾病选自心绞痛、心肌梗死、中风、心脏病发作、高血压性心脏病、风湿性心 脏病、心肌病、心脏心律失常和先天性心脏病;所述代谢性疾病选自糖尿病、痛风、肥胖症、低血糖症、高血糖症和血脂异常;所述神经性疾病选自阿尔茨海默病、帕金森病、亨廷顿病、头部损伤、多发性硬化症、眩晕、昏迷和癫痫。
发明的效果
在综合理解喜树碱衍生物的构效关系(SAR)的基础上,本发明设计合成出一系列具有显著抗肿瘤活性的喜树碱类衍生物。通过细胞实验发现,这类小分子化合物能够表现出优异的抗肿瘤活性。
具体实施方式
一般术语和定义
除非有相反陈述,否则在本发明中所使用的术语具有下述含义。
在本发明中,“烷基”是指饱和的一价脂族烃基团,包括1至10个碳原子的直链和支链基团,例如可以是1至8个碳原子、1至6个碳原子或1至4个碳原子的直链和支链基团。“C1-C10烷基”的实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基以及正戊基、正己基、正庚基、正辛基、正壬基、正癸基及其各种支链异构体等。
在本发明中,“环烷基”是指饱和或部分不饱和的、单环或多环的一价脂族烃基团,包括3至10个环原子,例如可以是3至6个环原子(即3至6元环)。“C3-C10环烷基”的实例包括但不限于环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基、环癸基及其部分不饱和的衍生基团等。“亚环烷基”是指从“环烷基”中取走环原子上的一个氢原子后形成的二价脂族烃基团。
在本发明中,“杂环烷基”是指饱和或部分不饱和的、单环或多环的一价脂族基团,包括3至10个环原子,例如可以是3至6个环原子,其中一个或多个环原子为选自N、O或S(O)m(其中m为0、1或2)的杂原子,其余环原子为C。优选的杂环烷基包括3至6个环原子,其中1至3个环原子是杂原子;更优选的杂环烷基包括4至6个环原子,其中1至3个环原子是杂原子;最优选的杂环烷基包括5或6个环原子,其中1或2个是杂原子。“3至10元杂环烷基”的实例包括但不限于氧杂环丙基、硫杂环丙基、吖丙啶基、氧杂环丁基、硫杂环丁基、吖丁啶基、四氢呋喃基、四氢噻吩基、吡咯烷基、四氢吡喃基、四氢噻喃基、哌啶基、哌嗪基、吗啉基、硫代吗啉基等。“亚杂环烷基”是指从“杂环烷基”中取走环原子上的一个氢原子后形成的二价脂族基团。
在本发明中,“卤素”是指氟、氯、溴和碘,优选氟、氯和溴。
在本发明中,“卤代烷基”是指“烷基”被至少一个彼此相同或不同的卤素原子取代后形成的一价脂族烃基团,包括1至10个碳原子的直链和支链基团,例如可以是1至8个碳原子、1至6个碳原子或1至4个碳原子的直链和支链基团。“C1-C10卤代烷基”的实例包括但不限于氟甲基、二氟甲基、三氟甲基、1-氟乙基、2-氟乙基、1,1-二氟乙基、1,2-二氟乙基、2,2-二氟乙基、2,2,2-三氟乙基、五氟乙基等。
在本发明中,“羟基”是指“-OH”基团。
在本发明中,“巯基”是指“-SH”基团。
在本发明中,“氨基”是指“-NH2”基团。
在本发明中,“羰基”是指“-C(=O)-”基团。
在本发明中,“药学上可接受的盐”是指由本发明中的化合物与相对无毒的酸或碱制备得到的盐。当本发明中的化合物含有相对偏酸性的官能团(例如羧基)时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与其游离形式接触的方式获得碱加成盐。药学上可接受的碱加成盐的非限制性实例包括但不限于钠盐、钾盐、铵盐、钙盐、镁盐、 有机胺盐或类似的盐。当本发明中的化合物含有相对偏碱性的官能团(例如叔胺基)时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与其游离形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的非限制性实例包括但不限于无机酸盐(例如盐酸盐、氢溴酸盐、氢碘酸盐、硝酸盐、碳酸盐、碳酸氢盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、亚磷酸盐、硫酸盐、硫酸氢盐等)、有机酸盐(例如乙酸盐、丙酸盐、异丁酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、马来酸盐、富马酸盐、柠檬酸盐、酒石酸盐、乳酸盐、扁桃酸盐、苯甲酸盐、邻苯二甲酸盐、甲磺酸盐、苯磺酸盐、对甲苯磺酸盐、葡糖醛酸等)以及氨基酸盐(例如精氨酸盐等)。药学上可接受的盐的具体形式还可参见Berge et al.,“Pharmaceutical Salts”,Journal of Pharmaceutical Science,1977,66:1-19)。
在本发明中,“抗体-药物偶联物”是指通过特定的化学连接片段将具有生物活性的小分子药物与大分子抗体(或抗体类配体)共价连接后形成的化合物,其中小分子药物发挥药理活性,而大分子抗体则主要发挥靶向投递作用,部分大分子抗体同时还发挥协同药效。
在本发明中,“药物组合物”是指可供药用的组合物,其包含作为活性药物成分(API)的小分子药物、大分子抗体(或抗体类配体)或二者的偶联物,以及其他组分(例如药学上可接受的辅料)。药物组合物可以使用本领域技术人员已知的任何方法来制备。例如,常规混合、溶解、造粒、乳化、磨细、包封、包埋和/或冻干工艺。
在本发明中,“药学上可接受的辅料”是指在药物生产领域中广泛采用的辅助物料。使用辅料的主要目的在于提供一种使用安全、性质稳定和/或具有特定功能性的药物组合物,还在于提供一种方法,以便在为受试者施用药物之后,活性成分能够以所期望的速率溶出,或者促进活性成分在接受给药的受试者体内得到有效吸收。药学上可接受的辅料可以是具有惰性的填充剂,也可以是为药物组合物提供某种功能(例如稳定组合物的整体pH值或防止组合物中活性成分的降解)的功效成分。“药学上可接受的辅料”的实例包括但不限于粘合剂、助悬剂、乳化剂、稀释剂(或填充剂)、成粒剂、胶粘剂、崩解剂、润滑剂、抗粘着剂、助流剂、润湿剂、胶凝剂、吸收延迟剂、溶解抑制剂、增强剂、吸附剂、缓冲剂、螯合剂、防腐剂、着色剂、矫味剂、甜味剂等。
本发明的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药以及基于其的抗体-药物偶联物和药物组合物均具有优良的肿瘤细胞杀伤作用,能够作为抗肿瘤药物,用于预防和/或治疗癌症,具有良好的临床应用和医药用途。
以下将结合具体实施例来阐述本发明的技术方案,下列实施例的提供旨在进一步说明本发明,而非用于限制本发明的范围。对本领域技术人员而言,在不脱离本发明的精神和范围的情况下,针对本发明的具体实施方式进行各种变化和改进将是显而易见的。
本发明所使用的起始原料可以通过本领域已知的方法来合成,或者通过常规的商业手段来购买。本发明的化合物的分离纯化可以通过本领域技术人员所熟知的方法来实现,包括但不限于柱色谱法(CC)、高效液相色谱法(HPLC)、超高效液相色谱法(UPLC)等。本发明的化合物的结构鉴定可以通过本领域技术人员所熟知的方法来实现,包括但不限于核磁共振(NMR)、质谱(MS)等。
缩略语表
目标化合物的制备和功能验证
实施例1:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-4-(羟甲基)环己烷-1-甲酰胺(化合物6A)
将4-(羟甲基)环己烷-1-甲酸(179.09mg,1.13mmol),HATU(322.88mg,0.85mmol),DIPEA(222.18mg,1.7mmol)和DMF(6mL)加入反应瓶中,氮气保护下,将体系降温至0~5℃,加入化合物1(301.23mg,0.56mmol),升温至15-20℃,搅拌反应0.5h后,取样,HPLC显示化合物1完全反应,加水(10mL)淬灭,反应液浑浊,过滤,得到褐色固体,经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(12mg,纯度:97.39%)。
1H-NMR(400MHz,DMSO-d6):δ8.38(d,J=8.7Hz,1H),7.82(d,J=11.0Hz,1H),7.33(s,1H),6.58(s,1H),5.58(dd,J=13.7,5.4Hz,1H),5.45(s,2H),5.18(dd,J=39.2,18.9Hz,2H),4.45(t,J=5.2Hz,1H),3.19(d,J=4.7Hz,2H),2.43(s,3H),2.32(dd,J=7.8,4.2Hz,1H),2.14(d,J=5.9Hz,2H),1.98-1.83(m,2H),1.73(dd,J=19.4,9.8Hz,2H),1.66-1.38(m,9H),0.90(t,J=7.3Hz,3H)。
MS(ESI+):m/z 576.3[M+H]+
实施例2:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-(2-羟乙氧基)丙酰胺(化合物2A)
按照实施例1中的方法,以3-(2-羟基乙氧基)丙酸和化合物1为原料,得到目标化合物(81.70mg,纯度:95.37%)。
1H-NMR(400MHz,DMSO-d6):δ8.53(d,J=8.7Hz,1H),7.82(d,J=10.9Hz,1H),7.33(s,1H),6.57(s,1H),5.67-5.55(m,1H),5.45(s,2H),5.22(q,J=19.0Hz,2H),4.52(s,1H),3.70(t,J=6.2Hz,2H),3.40(d,J=2.8Hz,4H),3.20(s,2H),2.44(d,J=10.6Hz,5H),2.17(m,2H),2.02-1.77(m,2H),0.90(t,J=7.3Hz,3H)。
MS(ESI+):m/z 552.2[M+H]+
实施例3:(S)-2-氨基-N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-甲基丁酰胺(化合物12A)
按照实施例1中的方法,以N-Boc-L-缬氨酸和化合物1为原料,得到作为淡黄色固体的化合物2(433.6mg)。产物未经进一步纯化,直接用于下一步反应。
将粗品化合物2(433.6mg,0.68mmol)溶解于等体积的TFA/DCM(12mL/12mL)中,反应1h后,取样,HPLC显示化合物2完全反应,减压除去溶剂,向残液中加入MTBE(24mL)后打浆,抽滤,烘干,得到作为褐色粉末状固体的目标化合物(293.15mg,纯度:98.15%)。
1H-NMR(400MHz,DMSO-d6):δ9.23(d,J=7.9Hz,1H),8.38(s,2H),7.84(d,J=10.9Hz,1H),7.34(s,1H),6.58(s,1H),5.63-5.54(m,1H),5.45(s,2H),5.41(d,J=18.9Hz,1H),5.12(d,J=18.8Hz,1H),3.68(s,1H),3.24(dd,J=32.5,14.6Hz,2H),2.43(s,3H),2.33(d,J=9.5Hz,1H),2.14(dd,J=11.6,7.0Hz,2H),1.90(td,J=13.1,6.4Hz,2H),0.91(s,9H)。
MS(ESI+):m/z 535.1[M+H]+
实施例4:(S)-2-氨基-N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)丙酰胺(化合物13A)
按照实施例3中的方法,以N-Boc-L-丙氨酸和化合物1为原料,得到作为棕绿色固体的目标化合物(323.50mg,纯度:95.98%)。
1H-NMR(400MHz,DMSO-d6):δ9.59(d,J=8.6Hz,1H),8.53(d,J=4.4Hz,3H),7.84(d,J=10.8Hz,1H),7.34(s,1H),5.66-5.56(m,1H),5.45(s,2H),5.25(d,J=8.3Hz,2H),4.04-3.96(m,1H),3.39-3.30(m,2H),2.43(s,3H),2.20(dd,J=11.0,5.4Hz,2H),1.95-1.83(m,2H),1.45(d,J=6.9Hz,4H),0.90(t,J=7.3Hz,3H)。
MS(ESI+):m/z 507.2[M+H]+
实施例5:(S)-2-氨基-N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-羟基丙酰胺(化合物14A)
按照实施例3中的方法,以N-Boc-O-叔丁基-L-丝氨酸和化合物1为原料,得到作为棕色固体的目标化合物(50.4mg,纯度:96.72%)。
1H-NMR(400MHz,DMSO-d6):δ8.99(d,J=8.6Hz,1H),8.23(d,J=3.8Hz,3H),7.87(d,J=11.0Hz,1H),7.35(s,1H),5.83-5.63(m,1H),5.45(s,2H),5.33(d,J=18.9Hz,1H),5.18(d,J=18.9Hz,1H),3.84(d,J=4.0Hz,1H),3.70(s,3H),3.19(dd,J=28.1,16.2Hz,2H),2.45(s,3H),2.22(dd,J=28.2,10.8Hz,2H),1.98-1.79(m,2H),0.91(t,J=7.3Hz,3H)。
MS(ESI+):m/z 523.3[M+H]+
实施例6:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-2,3-二羟基丙酰胺(化合物4A)
将2,2-二甲基-1,3-二氧杂环戊烷-4-甲酸(150mg,1.03mmol)溶于等体积的DCM/DMF混合溶剂(3mL)中,加入N-羟基琥珀酰亚胺(130.4mg,1.13mmol)和EDCI(394.0mg,2.06mmol),加毕搅拌2h,反应结束后,向反应液中加入DIPEA(110.30mg,0.20mmol),冰盐浴降温,加入化合物1(150.0mg,0.03mmol),加毕移去冰盐浴,搅拌2.5h后,取样,HPLC显示化合物1完全反应,加水(15mL)淬灭,再加入乙酸乙酯萃取,合并有机相,减压除去溶剂,得到化合物3粗品。
将粗品化合物3溶于1,4-二氧六环(2.5mL)中,冰盐浴冷却,再加入HCl的1,4-二氧六环溶液(4M,2.5mL),加毕升至室温,搅拌2h后,取样,HPLC显示化合物3完全反应,减压除去溶剂,再经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(32.4mg,纯度:96.57%)。
1H-NMR(400MHz,DMSO-d6):δ8.45-8.36(m,1H),7.89-7.74(m,1H),7.33(s,1H), 6.65-6.51(m,1H),5.76-5.51(m,1H),5.45(s,2H),5.35-4.89(m,2H),4.24-3.97(m,1H),3.81-3.63(m,3H),3.20-3.16(m,2H),2.42(s,3H),2.29-2.22(m,1H),2.20-2.11(m,1H),1.95-1.75(m,2H),0.89(t,J=7.3Hz,3H)。
MS(ESI+):m/z 524.2[M+H]+
实施例7:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-2-巯基乙酰胺(化合物10A)
将2-(三苯甲基硫基)乙酸(250mg,0.75mmol),T3P(957mg,3.0mmol),DIPEA(194mg,1.5mmol)溶于DMF(4mL)中,冰盐浴冷却,搅拌10min后,加入化合物1(200mg,0.37mmol),加毕移去冰盐浴,反应1h后,取样,HPLC显示化合物1完全反应,加水(10mL)淬灭,用乙酸乙酯萃取,合并有机相,经无水硫酸钠干燥,减压除去溶剂,得到化合物4粗品(200mg)。
将粗品化合物4(200mg,0.27mmol)溶于DCM(2mL)中,冰盐浴降温,先加入三乙基硅烷,搅拌10min,再缓慢滴加TFA(2mL),搅拌1h后,取样,HPLC显示化合物4完全反应,将反应液减压浓缩,向残液中加入MTBE打浆,抽滤,得到固体,再经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(72.1mg,纯度:91.23%)。
1H-NMR(400MHz,DMSO-d6):δ8.64(d,J=8.6Hz,1H),7.84(d,J=11.0Hz,1H),7.33(s,1H),5.57(dt,J=8.6,4.4Hz,1H),5.45(s,2H),5.27(dd,J=42.0,19.0Hz,2H),3.25-3.16(m,4H),2.89(t,J=8.1Hz,1H),2.43(s,3H),2.28-2.09(m,2H),1.96-1.79(m,2H),0.89(t,J=7.3Hz,3H)。
MS(ESI+):m/z 510.1[M+H]+
实施例8:1-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-(2-羟乙基)脲(化合物11A)
将2-((叔丁基二甲基硅基)氧基)乙胺(150.0mg,0.86mmol)加入到DCM(1.5mL)和饱和NaHCO3溶液(1.5mL)中,在冰水浴条件下加入三光气(84.0mg,0.29mmol),搅拌1.5h后反应结束,向反应液中加入DCM萃取,用饱和NaCl溶液洗涤,经无水Na2SO4干燥,减压除去溶剂,得到淡黄色油状液体。将油状液体溶于DCM(3mL)中,依次加 入三乙胺(57.0mg,0.56mmol)和化合物1(150.0mg,0.28mmol),搅拌2h后,取样,HPLC显示反应完成,加水(10mL)淬灭,再加入DCM萃取,合并有机相,经无水硫酸钠干燥,减压除去溶剂,得到化合物5(145.0mg)。
将化合物5(145.0mg,0.16mmol)溶于THF(0.5mL)中,冰水浴冷却,加入TBAF的THF溶液(1M,0.02mL),搅拌0.5h后,取样,HPLC显示反应完成,反应液经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(16.2mg,纯度:96.91%)。
1H-NMR(400MHz,DMSO-d6):δ7.78(d,J=11.0Hz,1H),7.32(s,1H),6.75(d,J=9.0Hz,1H),6.56(s,1H),6.03(t,J=5.6Hz,1H),5.45(s,2H),5.35(d,J=19.2Hz,2H),5.22(d,J=19.2Hz,1H),4.75(s,1H),3.46(d,J=5.4Hz,2H),3.20-3.10(m,4H),2.40(s,3H),2.25-2.09(m,2H),1.89(tt,J=14.1,7.2Hz,2H),0.90(t,J=7.3Hz,3H)。
MS(ESI+):m/z 523.2[M+H]+
实施例9:(1S,9S)-9-乙基-5-氟-9-羟基-1-((2-羟乙基)氨基)-4-甲基-1,2,3,9,12,15-六氢-10H,13H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-10,13-二酮(化合物8A)
将化合物1(100.05mg,0.19mmol)溶于DMF(3mL)和DCE(6mL)中,加入DIPEA(47.60mg,0.37mmol)和2-((叔丁基二甲基硅基)氧基)乙醛(47.6mg,0.27mmol),反应搅拌1h后,加入三乙酰氧基硼氢化钠(31mg,0.30mmol),反应5h后,经柱色谱纯化,冻干,得到化合物6(59.0mg)。粗品未经纯化,直接用于下一步反应。
向50mL单口瓶中加入粗品化合物6(59.0mg,0.10mmol),体积比为13:7:3的AcOH/H2O/THF混合溶液(0.5mL),反应搅拌10h后,取样,HPLC显示反应完成,经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(18.8mg,纯度:96.54%)。
1H-NMR(400MHz,DMSO-d6):δ8.88(d,J=96.6Hz,1H),7.92(d,J=10.8Hz,1H),7.38(s,1H),6.60(s,1H),5.55(d,J=19.0Hz,1H),5.49(s,2H),5.44(d,J=19.0Hz,1H),5.32(s,1H),5.12(s,1H),3.71(s,2H),3.31-3.12(m,4H),2.81(d,J=15.6Hz,1H),2.44(s,3H),2.20(t,J=14.0Hz,1H),1.91(qd,J=14.0,7.1Hz,2H),0.90(t,J=7.3Hz,3H)。
MS(ESI+):m/z 480.3[M+H]+
可采用类似方法制备得到化合物16A。
1H-NMR(400MHz,DMSO-d6):δ7.84(d,J=10.6Hz,1H),7.37(s,1H),5.26-5.56(m,4H),5.00(br s,1H),3.42-3.52(m,2H),3.19-3.32(m,2H),3.01-3.13(m,2H),2.63-2.74(m,1H),2.38(s,3H),2.13-2.25(m,1H),1.70-1.92(m,4H),0.85(t,J=7.3Hz,3H)。
MS(ESI+):m/z 494.3[M+H]+
实施例10:(S)-3-氨基-4-(((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)氨基)-4-氧代丁酸(化合物15A)
按照实施例3中的方法,以(S)-4-(叔丁氧基)-2-((叔丁氧羰基)氨基)-4-氧代丁酸和化合物1为原料,得到作为棕色固体的化合物24(93.2mg,纯度:98.05%)。
1H-NMR(400MHz,DMSO-d6):δ12.90(s,1H),9.01(d,J=8.7Hz,1H),8.30(s,2H),7.86(d,J=11.0Hz,1H),7.35(s,1H),6.58(s,1H),5.73-5.61(m,1H),5.46(s,2H),5.38(d,J=18.8Hz,1H),5.11(d,J=18.8Hz,1H),4.12-4.00(m,1H),3.21(d,J=26.5Hz,2H),2.86(dd,J=17.5,4.5Hz,1H),2.73(dd,J=17.5,8.1Hz,1H),2.45(s,4H),2.28(d,J=10.1Hz,1H),2.12(d,J=12.0Hz,1H),1.89(dt,J=10.6,6.8Hz,2H),0.91(t,J=7.3Hz,3H)。
MS(ESI+):m/z 551.2[M+H]+
实施例11:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-(羟甲基)环丁烷-1-甲酰胺(化合物1A)
向反应瓶中加入3-(羟甲基)环丁烷-1-甲酸(49mg,0.37mmol),DCM(15mL),化合物1(100mg,0.19mmol)和DMAP(80mg,0.48mmol),冰浴搅拌10min,再加入二环己基碳二亚胺(97mg,0.48mmol),N,N-二甲基甲酰胺(3mL),室温反应过夜,取样,TLC显示反应完成,将反应液减压浓缩,经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(22.8mg,纯度:95.7%)。
1H-NMR(400MHz,DMSO-d6):δ8.33(dd,J=8.7,3.5Hz,1H),7.79(d,J=10.9Hz,1H),7.31(d,J=1.1Hz,1H),6.52(s,1H),5.55(dd,J=12.6,7.1Hz,1H),5.43(s,2H),5.26-5.03(m,2H),4.54(t,J=5.3Hz,1H),4.45(t,J=5.3Hz,1H),3.42(dd,J=6.4,5.3Hz,1H),3.27-2.84(m,3H),2.45-2.35(m,3H),2.35-2.23(m,2H),2.23-2.04(m,3H),2.02-1.76(m,4H),1.24(s,2H),0.96-0.73(m,3H).
MS(ESI+):m/z 548.3[M+H]+
实施例12:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-4-(2-羟乙基)环己烷-1-甲酰胺(化合物5A)
向反应瓶中加入4-(2-羟乙基)环己烷-1-甲酸(97mg,0.56mmol),DCM(12mL),化合物1(150mg,0.28mmol)和DMAP(121mg,0.98mmol),冰浴冷却,在加入DCC(146mg,0.70mmol)和DMF(12mL),加毕移去冰浴,室温反应过夜,取样,TLC显示反应完成,将反应液浓缩,经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(23mg,纯度:96.5%)。
1H-NMR(400MHz,DMSO-d6):δ8.36(dd,J=16.4,8.7Hz,1H),7.80(d,J=10.9Hz,1H),7.31(s,1H),7.21(s,1H),6.66(s,1H),6.53(s,1H),5.56(p,J=5.0Hz,1H),5.43(s,1H),5.33(dd,J=5.4,4.2Hz,1H),5.25-5.07(m,2H),2.41(d,J=1.9Hz,2H),2.28(d,J=8.6Hz,2H),2.12(d,J=6.4Hz,2H),2.00(p,J=7.0Hz,1H),1.94-1.67(m,4H),1.66-1.37(m,5H),1.24(d,J=3.9Hz,6H),0.87(q,J=7.2Hz,3H)。
MS(ESI+):m/z 590.3[M+H]+
实施例13:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-(哌啶-4-基)丙酰胺(化合物9A)
将3-(1-(叔丁氧羰基)哌啶-4-基)丙酸(100mg,0.39mmol)加入到反应试管中,加入DMAP(34.5mg,0.58mmol),化合物1(48.5mg,0.39mmol)和DCM(2mL),冰浴冷却,再滴加DCC(42.7mg,0.43mmol)的DCM(3mL)溶液,常温反应过夜,取样,TLC显示反应完成,加入水(10mL)洗涤两遍,水相用DCM萃取,合并有机相,经无水硫酸钠干燥,过滤,收集滤液,旋干,得到化合物7(230mg)。粗品未经进一步纯化,直接用于下一步反应。
将化合物7(230mg,0.35mmol)溶解在DCM(10mL)中,加入TFA(15mL),室温搅拌2h后,TLC显示反应完成,将反应液浓缩,经制备型色谱纯化(乙腈/水:20%~90%,V/V),冻干,得到目标化合物(19mg,纯度97.2%)。
1H-NMR(400MHz,DMSO-d6):δ8.50(d,J=8.7Hz,1H),8.26(s,1H),7.82(d,J=11.1Hz,1H),7.68(s,1H),6.18(s,1H),5.54(dd,J=8.8,4.6Hz,1H),5.11(d,J=4.0Hz,1H),4.64(d,J=11.6Hz,1H),3.26-3.04(m,5H),2.79(t,J=12.6Hz,2H),2.43-2.35(m,3H),2.12(tdt,J=33.5,13.7,7.1Hz,5H),1.78(d,J=12.8Hz,3H),1.56(dt,J=37.3,7.8Hz,3H),1.22(d,J=14.6Hz,3H),0.86(t,J=7.2Hz,3H)。
MS(ESI+):m/z 575.2[M+H]+
实施例14:N-((1S,9S)-9-乙基-5-氟-9-羟基-4-甲基-10,13-二氧代-2,3,9,10,13,15-六氢-1H,12H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-1-基)-3-(3-羟丙基)环丁烷-1-甲酰胺 (化合物17A)
将化合物1(5.00g,35.6mmol,1.00eq)溶于无水THF(50.0mL)中,控温-10℃,滴加BH3.THF(1M,35.6mL,1.00eq),升至20℃并搅拌2小时。将反应液冷却至-20℃,加入甲醇(50.0mL)并搅拌15分钟,接着加入NaOH水溶液(10%,7.13g,17.8mmol,0.500eq)和H2O2(7.70g,67.9mmol,6.53mL,1.90eq),升至20℃并搅拌2小时。升至20℃并加入饱和Na2SO3(150mL),用DCM(150mL*3)萃取,合并有机相并用饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得无色油状化合物2(3.00g,18.9mmol)。
将PCC(氯铬酸吡啶盐,3.41g,15.8mmol,1.25eq)和硅藻土(3.41g)加入至DCM(20.0mL)中,0℃下加入化合物2(2.00g,12.6mmol,1.00eq)的DCM(20.0mL)溶液,20℃反应5小时。过滤并收集滤液浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得无色油状化合物3(800mg,5.12mmol)。
将化合物10(1.94g,7.68mmol,1.20eq)溶于THF(10.0mL)中,降温至-78℃,滴加n-BuLi(2.5M,3.07mL,1.20eq),30分钟左右滴完,然后滴加入化合物3(1.00g,6.40mmol,1.00eq)的THF(2.00mL)溶液,30分钟左右滴加完毕,升至20℃并搅拌2小时。反应完毕后加入饱和氯化铵(15.0mL)淬灭并用EA萃取(10.0mL*3),收集有机相并用饱和食盐水(15.0mL)洗,干燥并浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得无色油状化合物4(650mg,2.56mmol)。
将化合物4(800mg,3.15mmol,1.00eq)溶于EA(10.0mL)中,氮气保护下加入10%的Pd/C(800mg),氢气置换3次,氢气条件(30psi)下,25℃反应16小时。过滤并收集滤液,浓缩至干,得无色油状化合物5(500mg,1.95mmol)。
将化合物5(1.00g,3.90mmol,1.00eq)溶于DCM(10.0mL)中,加入TFA(15.4g,135mmol,10.0mL,34.6eq),25℃下反应12小时。反应液浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得无色油状化合物6(500mg,2.50mmol)。
将化合物6(500mg,2.50mmol,1.00eq)溶于THF(10.0mL)中,0℃下加入BH3.THF(1M,2.50mL,1.00eq),升至25℃反应2小时。0℃下加入甲醇(20.0mL)淬灭并搅拌15分钟,浓缩至干,加入水(20.0mL),并用EA(20.0mL*3)萃取,收集有机相并用饱和食盐水洗,无水硫酸钠干燥,过滤,滤液浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得无色油状化合物7(390mg,2.09mmol)。
将化合物7(390mg,2.09mmol,1.00eq)溶于THF(10.0mL),20℃下加入LiOH.H2O(2M,4.19mL,4.00eq),25℃下反应12小时。将反应液倒入冰水(20.0mL)中,并搅拌10分钟,用EA(20.0mL)萃取,水相用1N HCl调节pH至5-6,用EA萃取(20.0mL*3),收集有机相并用饱和食盐水洗,无水硫酸钠干燥,过滤,滤液浓缩至干,得黄色油状化合物8(140mg,粗品)。
将化合物8(80.0mg,505μmol,1.00eq)溶于DCM(5.00mL),0℃下加入TEA(153mg,1.52mmol,211μL,3.00eq)、HATU(211mg,556μmol,1.10eq)和化合物9(231mg,530μmol,1.05eq),升至25℃反应12小时。用水(20.0mL)淬灭,并用DCM(20.0mL*3)萃取,收集有机相并用饱和食盐水(20.0mL)洗,无水硫酸钠干燥,过滤,滤液浓缩至干,薄层色谱纯化,得黄色固体目标化合物(22.0mg,38.2μmol)。
1H-NMR(400MHz,DMSO-d6):δ8.36(br d,J=8.9Hz,1H),7.77(d,J=10.9Hz,1H),7.22-7.39(m,1H),6.52(s,1H),5.35-5.61(m,3H),5.00-5.19(m,2H),4.25-4.45(m,1H),2.99-3.29(m,4H),2.80-2.96(m,1H),2.28-2.42(m,4H),2.01-2.26(m,4H),1.69-1.95(m,4H),1.28-1.45(m,4H),0.87(br t,J=7.3Hz,3H)。
MS(ESI+):m/z 576.4[M+H]+
实施例15:(9S)-9-乙基-5-氟-9-羟基-1-(2-羟乙基)-4-甲基-1,2,3,9,12,15-六氢-10H,13H-苯并[de]吡喃并[3',4':6,7]吲嗪并[1,2-b]喹啉-10,13-二酮(化合物18)
将化合物1(5.00g,21.2mmol)溶于THF(50.0mL)中,氮气保护,-50℃下缓慢滴加KHMDS(1M,42.5mL),滴加完毕后-50℃搅拌反应3小时,加入化合物1a(6.08g,21.2mmol),升温至20℃,并反应12小时。反应完毕后降温至10℃,将反应液倒入至饱和氯化铵水溶液(100mL)中,并用EA(100mL*3)萃取,收集有机相并用饱和食盐水(100mL*2)洗,无水硫酸钠干燥,过滤,滤液浓缩至干,柱层析纯化(PE/EA=100/1~10/1),得黄色油状化合物2(1.00g,2.39mmol)。
将化合物2(1.00g,2.54mmol)溶于甲醇(20mL),氮气保护,25℃下缓慢滴加HCl甲醇(2M,25.4mL),升至60℃反应2小时。过滤,收集滤液并浓缩至干,得黄色固体化合物3(粗品,1.00g)。
将化合物3(1.00g,3.65mmol)和化合物4(961mg,3.65mmol)溶于甲苯(5.00mL),25℃下加入TsOH(31.4mg,182μmol),升至130℃反应12小时。反应液浓缩至干,薄 层色谱纯化,得黄色固体目标化合物(30.0mg)。
1H-NMR(400MHz,DMSO-d6):δ7.71(d,J=11.0Hz,1H),7.29(d,J=1.5Hz,1H),5.42(s,2H),5.27(s,2H),3.47-3.56(m,4H),2.99-3.13(m,2H),2.23-2.38(m,4H),1.80-1.97(m,3H),1.67-1.77(m,2H),0.87(t,J=7.3Hz,3H)。
MS(ESI+):m/z 465.3[M+H]+
可采用类似方法制备得到化合物19。
1H-NMR(400MHz,DMSO-d6):δ7.72(d,J=11.1Hz,1H),7.30(d,J=1.8Hz,1H),5.43(s,2H),5.38-5.30(m,1H),5.26-5.17(m,1H),5.10-4.76(m,4H),3.45(br d,J=5.1Hz,2H),3.34(br s,1H),3.16-2.95(m,2H),2.38-2.23(m,4H),1.98-1.80(m,3H),1.76-1.50(m,4H),0.87(t,J=7.2Hz,3H)。
MS(ESI+):m/z 479.2[M+H]+
实验例1:喜树碱衍生物的肿瘤细胞杀伤作用研究
为了检测本发明的化合物对于肿瘤细胞的杀伤作用,采用SK-OV-3(市售)、293T-FOLR1(通过下文描述的方法构建构建)、293T(市售)、SK-BR-3(市售)进行评估,并采用Dx(依喜替康,CAS:171335-80-1,市售)作为阳性对照。
细胞模型293T-FOLR1的构建:通过表达FRα的基因序列构建含有FRα编码基因的重组病毒载体及重组病毒颗粒,然后将其稳转到293T细胞株中,通过在转染过程中降低培养基中的血清浓度,添加polybrene,并调整5MOI值提高病毒转染效率,获得高表达FRα的细胞模型293T-FOLR1。
分别收集293T、293T-FOLR1、SK-OV-3、SK-BR-3细胞,离心计数后,采用完全培养基调整细胞密度为1x104个/mL,接种于细胞板的中间60个孔,100μl/孔,细胞数为1000细胞/孔,边缘孔加入100μl/孔的PBS。将96孔板放入37℃,5%CO2的培养箱中培养过夜。
次日,用完全培养基在1.5mL的EP管中配制工作溶液,起始浓度为10μM,采用5倍稀释,共9个浓度。配制完成后,加入到96孔板中,50μl/孔,三复孔。将细胞板放入37℃,5%CO2的培养箱中继续培养72小时。
第五天,取出细胞板,加入CCK8细胞活力检测试剂(Abbkine,KTC011001),15μl/孔,振荡混匀后,放于暗处静置70min,使用酶标仪的发光程序进行检测,使用GraphPad Prims软件计算IC50值,实验结果如下表所示。

注:“-”表示未进行相关测定。
实验结果表明,针对SK-OV-3细胞,化合物1A、8A、12A、18和19均优于阳性对 照;针对293T-FOLR1细胞,化合物16A和19表现出与阳性对照基本相当的杀伤效果,而化合物17A则显著优于阳性对照;针对293T细胞,大部分化合物的活性与阳性对照处于同一数量级水平,而化合物17A则显著优于阳性对照;针对SK-BR-3细胞,大部分化合物的活性与阳性对照处于同一数量级水平,而化合物18则显著优于阳性对照。因此,本发明的化合物对293T、293T-FOLR1、SK-OV-3、SK-BR-3细胞具有较好的杀伤效果,适合于开发成抗肿瘤药物。
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制。在不脱离本发明的原理和宗旨的情况下,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型,这些变化、修改、替换和变型均涵盖在本发明的范围之中。

Claims (18)

  1. 如式I所示的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其中
    若存在,X为-O-、-N(R4)-、C3-C10亚环烷基或3至10元亚杂环烷基;
    Y为羟基、巯基、氨基、C3-C10环烷基或3至10元杂环烷基;
    若存在,R1为氢、C1-C10烷基或C1-C10卤代烷基;
    若存在,每一个R2各自独立地为氢、C1-C10烷基或C1-C10卤代烷基;
    每一个R3各自独立地为氢、羟基、氨基、C1-C10烷基或C1-C10卤代烷基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基、C3-C10亚环烷基或3至10元亚杂环烷基;
    若存在,R4为氢、C1-C10烷基或C1-C10卤代烷基;
    k、l和m各自独立地为0至5中的任一整数;
    n为1至5中的任一整数;
    并且,当k为1,l和m均为0,n为1,R1和R3均为氢时,Y不为氨基;
    所述杂环烷基和亚杂环烷基各自独立地包含1至4个杂原子或杂原子团,所述杂原子或杂原子团各自独立地为C(=O)、O、S、S(=O)、S(=O)2、N或NH。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    当k和l均为0,m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
  3. 根据权利要求1所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,所述化合物如式IA所示,其中
    X为-O-、-N(R4)-或C3-C10亚环烷基,优选-O-、-N(R4)-或C3-C6亚环烷基,更优选-O-、 -N(R4)-或C4-C6亚环烷基;
    Y为羟基、巯基或氨基,优选羟基;
    R1为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    若存在,每一个R2各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    若存在,R4为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    l为0、1、2或3,优选0、1或2;
    n为1、2、3或4,优选1、2或3。
  4. 根据权利要求3所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    X为C3-C10亚环烷基,优选C3-C6亚环烷基,更优选C4-C6亚环烷基;
    Y为羟基;
    R1为氢;
    每一个R3各自独立地为氢;
    l为0;
    n为1、2或3。
  5. 根据权利要求3所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    X为-O-;
    Y为羟基;
    R1为氢;
    每一个R2各自独立地为氢;
    每一个R3各自独立地为氢;
    l为1或2;
    n为1或2。
  6. 根据权利要求3所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    X为-NH-;
    Y为羟基;
    R1为氢;
    每一个R3各自独立地为氢;
    l为0;
    n为1、2或3。
  7. 根据权利要求1所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,所述化合物如式IB所示,其中
    Y为羟基、巯基、氨基或3至10元杂环烷基,优选羟基、巯基、氨基或3至6元杂环烷基,更优选羟基、巯基、氨基或3至6元杂环烷基;
    R1为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    每一个R3各自独立地为氢、羟基、氨基或C1-C10烷基,优选氢、羟基、氨基或C1-C4烷基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基;
    n为1、2、3或4,优选1、2或3;
    并且,当n为1,R1和R3均为氢时,Y不为氨基;
    所述杂环烷基包含1至2个杂原子或杂原子团,所述杂原子或杂原子团各自独立地为O、S、S(=O)、S(=O)2、N或NH,优选O、S、N或NH。
  8. 根据权利要求7所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    Y为羟基、巯基或氨基;
    R1为氢;
    每一个R3各自独立地为氢、羟基或C1-C10烷基,优选氢、羟基或C1-C4烷基;优选地,每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基;
    n为1、2或3。
  9. 根据权利要求1所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,所述化合物如式IC所示,其中
    若存在,X为-O-或-N(R4)-;
    Y为羟基、巯基或氨基,优选羟基;
    每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    若存在,R4为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    m为0或1;
    n为1、2、3或4,优选2或3。
  10. 根据权利要求9所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    若存在,X为-O-或-NH-;
    Y为羟基;
    每一个R3各自独立地为氢或C1-C10烷基,优选氢或C1-C4烷基,更优选氢;
    m为0或1;
    n为2或3。
  11. 根据权利要求9或10所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    当m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
  12. 根据权利要求1所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    若存在,X为-O-、-NH-、亚环丙基、亚环丁基、亚环戊基或亚环己基;
    Y为羟基、巯基、氨基、四氢吡咯基、四氢呋喃基、四氢噻吩基、哌啶基、吡喃基或噻喃基;
    若存在,R1为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
    若存在,每一个R2各自独立地为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
    每一个R3各自独立地为氢、羟基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;或者,连接同一个碳原子的两个R3与其所连接的碳原子共同形成羰基、环丙基、环丁基、环戊基、环己基、四氢吡咯基、四氢呋喃基、四氢噻吩基、哌啶基、吡喃基或噻喃基;
    若存在,R4为氢、甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基;
    k为0或1;
    l为0、1、2;
    m为0或1;
    n为1、2或3;
    并且,当k为1,l和m均为0,n为1,R1和R3均为氢时,Y不为氨基。
  13. 根据权利要求12所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药,其特征在于,
    当k和l均为0,m为1,n为2,X为-NH-,R3均为氢时,Y不为羟基。
  14. 下列化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药:



  15. 根据权利要求1至14中任一项所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药在包含其的药物偶联物中作为细胞毒药物的用途。
  16. 一种抗体-药物偶联物,其包含根据权利要求1至14中任一项所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药以及与之偶联的抗体或抗体类配体;
    优选地,所述抗体或抗体类配体选自抗EGFRvIII抗体、抗DLL-3抗体、抗PSMA抗体、抗CD70抗体、抗MUC16抗体、抗ENPP3抗体、抗TDGF1抗体、抗ETBR抗体、抗MSLN抗体、抗TIM-1抗体、抗LRRC15抗体、抗LIV-1抗体、抗CanAg/AFP抗体、抗cladin 18.2抗体、抗Mesothelin抗体、抗HER2(ErbB2)抗体、抗EGFR抗体、抗c-MET抗体、抗SLITRK6抗体、抗KIT/CD117抗体、抗STEAP1抗体、抗SLAMF7/CS1抗体、抗NaPi2B/SLC34A2抗体、抗GPNMB抗体、抗HER3(ErbB3)抗体、抗MUC1/CD227抗体、抗AXL抗体、抗CD166抗体、抗B7-H3(CD276)抗体、抗PTK7/CCK4抗体、抗PRLR抗体、抗EFNA4抗体、抗5T4抗体、抗NOTCH3抗体、抗Nectin 4抗体、抗TROP-2抗体、抗CD142抗体、抗CA6抗体、抗GPR20抗体、抗CD174抗体、抗CD71抗体、抗EphA2抗体、抗LYPD3抗体、抗FGFR2抗体、抗FGFR3抗体、抗FRα抗体、抗CEACAMs抗体、抗GCC抗体、抗Integrin Av抗体、抗CAIX抗体、抗P-cadherin抗体、抗GD3抗体、抗Cadherin 6抗体、抗LAMP1抗体、抗FLT3抗体、抗BCMA抗体、抗CD79b抗体、抗CD19抗体、抗CD33抗体、抗CD56抗体、抗CD74抗体、抗CD22抗体、抗CD30抗体、抗CD37抗体、抗CD138抗体、抗CD352抗体、抗CD25抗体、抗CD123抗体或抗CD47抗体。
  17. 一种药物组合物,其包含根据权利要求1至14中任一项所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者根据权利要求16所述的抗体-药物偶联物;
    优选地,所述药物组合物还包含至少一种药学上可接受的辅料。
  18. 根据权利要求1至14中任一项所述的化合物或其药学上可接受的盐、N-氧化物、溶剂化物、立体异构体、互变异构体、同位素标记物、代谢产物或前药或者根据权利要求16所述的抗体-药物偶联物或者根据权利要求17所述的药物组合物在制备用于预防和/或治疗至少部分由DNA拓扑异构酶I介导的疾病或病症的药物中的应用;
    优选地,所述至少部分由DNA拓扑异构酶I介导的疾病或病症为癌症、自身免疫性疾病、心血管疾病、代谢性疾病或神经性疾病;
    更优选地,所述癌症选自肺癌、肾癌、结肠癌、直肠癌、卵巢癌、乳腺癌、肝癌、膀胱癌、前列腺癌、胃癌、宫颈癌、子宫癌、结直肠癌、食道癌、子宫内膜癌、唾液腺癌、甲状腺癌、胰腺癌和骨癌;
    更优选地,所述自身免疫性疾病选自结缔组织病、系统性硬化症、类风湿性关节炎 和系统性红斑狼疮;
    更优选地,所述心血管疾病选自心绞痛、心肌梗死、中风、心脏病发作、高血压性心脏病、风湿性心脏病、心肌病、心脏心律失常和先天性心脏病;
    更优选地,所述代谢性疾病选自糖尿病、痛风、肥胖症、低血糖症、高血糖症和血脂异常;
    更优选地,所述神经性疾病选自阿尔茨海默病、帕金森病、亨廷顿病、头部损伤、多发性硬化症、眩晕、昏迷和癫痫。
PCT/CN2023/092670 2022-05-09 2023-05-08 一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用 WO2023217064A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380022412.2A CN118696048A (zh) 2022-05-09 2023-05-08 一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210503019.2 2022-05-09
CN202210503019 2022-05-09
CN202310447872.1 2023-04-24
CN202310447872 2023-04-24

Publications (1)

Publication Number Publication Date
WO2023217064A1 true WO2023217064A1 (zh) 2023-11-16

Family

ID=88729746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/092670 WO2023217064A1 (zh) 2022-05-09 2023-05-08 一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用

Country Status (3)

Country Link
CN (1) CN118696048A (zh)
TW (1) TW202344252A (zh)
WO (1) WO2023217064A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024067811A1 (en) * 2022-09-30 2024-04-04 Beigene, Ltd. Ligand-drug conjugate of exatecan analogue, and medical use thereof
WO2024125627A1 (zh) * 2022-12-16 2024-06-20 上海齐鲁制药研究中心有限公司 喜树碱类化合物及其制备方法和用途
WO2024158996A3 (en) * 2023-01-25 2024-09-19 Zeno Management, Inc. Exatecan immunoconjugates
WO2024199462A1 (zh) * 2023-03-30 2024-10-03 上海齐鲁制药研究中心有限公司 依喜替康衍生物及其抗体药物偶联物

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111689980A (zh) * 2019-05-26 2020-09-22 四川百利药业有限责任公司 一种喜树碱药物及其抗体偶联物
CN112125915A (zh) * 2019-09-18 2020-12-25 四川百利药业有限责任公司 一种喜树碱衍生物及其偶联物
CN113827736A (zh) * 2020-06-08 2021-12-24 四川百利药业有限责任公司 一种带有高稳定性亲水连接单元的喜树碱类药物及其偶联物
WO2022015656A1 (en) * 2020-07-13 2022-01-20 Regeneron Pharmaceuticals, Inc. Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
WO2022056696A1 (zh) * 2020-09-15 2022-03-24 四川百利药业有限责任公司 一种喜树碱类药物及其抗体偶联物
WO2022068878A1 (zh) * 2020-09-30 2022-04-07 映恩生物制药(苏州)有限公司 一种抗肿瘤化合物及其制备方法和应用
WO2022078260A1 (zh) * 2020-10-12 2022-04-21 四川百利药业有限责任公司 一种喜树碱类衍生物及其配体-药物偶联物
WO2022166762A1 (zh) * 2021-02-05 2022-08-11 四川科伦博泰生物医药股份有限公司 喜树碱类化合物及其制备方法和应用
WO2022236136A1 (en) * 2021-05-07 2022-11-10 ALX Oncology Inc. Exatecan derivatives and antibody-drug conjugates thereof
WO2023004266A1 (en) * 2021-07-19 2023-01-26 Recurium Ip Holdings, Llc Immunoconjugates and methods
CN115850291A (zh) * 2021-09-24 2023-03-28 石药集团巨石生物制药有限公司 喜树碱衍生物及其用途

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111689980A (zh) * 2019-05-26 2020-09-22 四川百利药业有限责任公司 一种喜树碱药物及其抗体偶联物
CN112125915A (zh) * 2019-09-18 2020-12-25 四川百利药业有限责任公司 一种喜树碱衍生物及其偶联物
CN113827736A (zh) * 2020-06-08 2021-12-24 四川百利药业有限责任公司 一种带有高稳定性亲水连接单元的喜树碱类药物及其偶联物
WO2022015656A1 (en) * 2020-07-13 2022-01-20 Regeneron Pharmaceuticals, Inc. Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
WO2022056696A1 (zh) * 2020-09-15 2022-03-24 四川百利药业有限责任公司 一种喜树碱类药物及其抗体偶联物
WO2022068878A1 (zh) * 2020-09-30 2022-04-07 映恩生物制药(苏州)有限公司 一种抗肿瘤化合物及其制备方法和应用
WO2022078260A1 (zh) * 2020-10-12 2022-04-21 四川百利药业有限责任公司 一种喜树碱类衍生物及其配体-药物偶联物
WO2022166762A1 (zh) * 2021-02-05 2022-08-11 四川科伦博泰生物医药股份有限公司 喜树碱类化合物及其制备方法和应用
WO2022236136A1 (en) * 2021-05-07 2022-11-10 ALX Oncology Inc. Exatecan derivatives and antibody-drug conjugates thereof
WO2023004266A1 (en) * 2021-07-19 2023-01-26 Recurium Ip Holdings, Llc Immunoconjugates and methods
CN115850291A (zh) * 2021-09-24 2023-03-28 石药集团巨石生物制药有限公司 喜树碱衍生物及其用途

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024067811A1 (en) * 2022-09-30 2024-04-04 Beigene, Ltd. Ligand-drug conjugate of exatecan analogue, and medical use thereof
WO2024125627A1 (zh) * 2022-12-16 2024-06-20 上海齐鲁制药研究中心有限公司 喜树碱类化合物及其制备方法和用途
WO2024158996A3 (en) * 2023-01-25 2024-09-19 Zeno Management, Inc. Exatecan immunoconjugates
WO2024199462A1 (zh) * 2023-03-30 2024-10-03 上海齐鲁制药研究中心有限公司 依喜替康衍生物及其抗体药物偶联物

Also Published As

Publication number Publication date
CN118696048A (zh) 2024-09-24
TW202344252A (zh) 2023-11-16

Similar Documents

Publication Publication Date Title
WO2023217064A1 (zh) 一种喜树碱衍生物,基于其的抗体-药物偶联物和药物组合物,及其应用
CN111484477B (zh) 一种苯并吡啶酮杂环化合物及其用途
ES2929415T3 (es) Compuestos heterocíclicos tricíclicos como activadores de STING
EP4162954A1 (en) Camptothecin drug having high-stability hydrophilic connecting unit and conjugate thereof
CN108863963B (zh) 作为pd-l1抑制剂的杂环类化合物
WO2022078259A1 (zh) 一种氘代的喜树碱衍生物及其抗体药物偶联物
WO2016131414A1 (zh) 氘代鹅去氧胆酸衍生物以及包含该化合物的药物组合物
WO2020011246A1 (zh) 含苯环的化合物、其制备方法及应用
WO2021227938A1 (zh) 含非甾体抗炎药结构的氨基酸衍生物及其制备方法与应用
CN112300153A (zh) 一种杂环化合物、药物组合物和用途
WO2023280136A1 (zh) 氘甲基取代吡嗪并吡嗪并喹啉酮类衍生物、其制备方法及其在医药上的应用
JP2005525367A (ja) 塩酸イリノテカンの結晶多形
JP2021520409A (ja) 細胞障害性薬物の複合体及び前記複合体のプロドラッグの形態
TWI823420B (zh) 用作cdk激酶抑制劑的化合物及其應用
KR20240120744A (ko) 캄프토테신 화합물 및 이의 접합체
CN113816990A (zh) 修饰的氨基酸及其在adc中的应用
WO2016197946A1 (zh) 抗体药物偶联物、中间体、制备方法、药物组合物及应用
WO2022126593A1 (zh) 一种靶向trop2的抗体药物偶联物、其制备方法及应用
WO2023232145A1 (zh) 一种高喜树碱类小分子及其应用
CN114853735B (zh) 靶向泛素化降解trk的化合物及其制备方法、组合物和用途
WO2023025324A1 (zh) 作为p53调节剂的化合物
CN113024557B (zh) 一种Peganumine A生物碱结构简化物及其应用
CN115724784A (zh) 作为p53调节剂和/或hdac抑制剂的化合物
BR112021013099A2 (pt) Composto de benzotiofeno substituído contendo flúor, e composição farmacêutica e aplicação dos mesmos
WO2022126569A1 (zh) 一种靶向b7-h3的抗体药物偶联物、其制备方法及应用

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: 23802836

Country of ref document: EP

Kind code of ref document: A1