WO2022166980A1 - 杂芳基并哌啶类衍生物及其药物组合物和应用 - Google Patents

杂芳基并哌啶类衍生物及其药物组合物和应用 Download PDF

Info

Publication number
WO2022166980A1
WO2022166980A1 PCT/CN2022/075486 CN2022075486W WO2022166980A1 WO 2022166980 A1 WO2022166980 A1 WO 2022166980A1 CN 2022075486 W CN2022075486 W CN 2022075486W WO 2022166980 A1 WO2022166980 A1 WO 2022166980A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
methyl
tetrahydro
fluoropropyl
isoquinolin
Prior art date
Application number
PCT/CN2022/075486
Other languages
English (en)
French (fr)
Inventor
祖厚贤
王义乾
杜国龙
陈凯
徐晓峰
刘湘永
丁列明
王家炳
Original Assignee
贝达药业股份有限公司
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 贝达药业股份有限公司 filed Critical 贝达药业股份有限公司
Priority to CN202280009011.9A priority Critical patent/CN116669729A/zh
Publication of WO2022166980A1 publication Critical patent/WO2022166980A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of medicine, and in particular relates to a heteroaryl piperidine derivative and a pharmaceutical composition and application thereof.
  • Estrogen receptor is a transcriptional regulatory protein that mediates ligand-activated activation of multiple biological effects through its interaction with endogenous estrogens.
  • ER contains two isoforms, ER ⁇ and ER ⁇ .
  • ER ⁇ is mainly distributed in the uterus, ovary, testis, pituitary, kidney, epididymis and adrenal gland, while ER ⁇ is mainly distributed in the prostate, ovary, lung, bladder, brain and blood vessels. Because full agonists or full antagonists have serious side effects, the research of selective estrogen receptor modulator SERM came into being.
  • SERM acts as an agonist in certain tissues such as bone, liver, and ER ⁇ -concentrated areas of the cardiovascular system, and as an antagonist in other tissues such as breast. It can be either an agonist or an antagonist in the uterus (where ERa is more pronounced).
  • SERMs include Tamoxifen, Raloxifene, Bazedoxifene, Toremifene, etc.
  • SESDs Selective estrogen receptor downregulators
  • AF1 and AF2 Drugs (complete antagonists).
  • Fulvestrant is the only SERD drug currently approved for clinical use in the treatment of ER+ breast cancer, but it has poor druggable properties, is rapidly metabolized and must be administered by monthly intramuscular injections, in contrast to what has been seen in in vitro studies This limits the efficient degradation of ER compared to complete ER degradation ( ⁇ 50% ER degradation in clinical samples).
  • AstraZeneca's WO2018077630A patent discloses a series of SERD compounds, including early clinical drug AZD9833, wherein the structure of AZD9833 is:
  • Selective estrogen receptor down-regulators have shown some therapeutic advantages, but more orally available selective estrogen receptors still need to be developed, so that drug candidates have more excellent properties, such as better efficacy, lower side effects, oral Better absorption, better pharmacokinetic properties, etc., so that it can be better used for the prevention or treatment of estrogen receptor-related diseases.
  • the present invention provides an oral selective estrogen receptor compound with better pharmacokinetic properties and a pharmaceutical composition and application thereof.
  • the present invention provides a compound represented by formula (I), its stereoisomer, tautomer or pharmaceutically acceptable salt:
  • Ring A is selected from
  • D is CR 9 or N
  • E is CR 10 or N
  • R 1 is selected from H, F, Cl, Br, I, -CN, -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 CH(CH 3 ) 2 , -CH 2 OH, -CH2OCH3 , CH2CH2OH, -C ( CH3 )2OH, -CH(OH) CH ( CH3 ) 2 , -C ( CH3 ) 2CH2OH , -CH2CH2SO 2CH3 , -CH2OP ( O) ( OH ) 2 , -CH2F , -CHF2 , -CH2NH2 , -CH2NHSO2CH3 , -CH2NHCH3 , -CH2N ( CH3 ) 2 , -CF3 , -CH2CF3 , -CH2CHF2 , -CH( CH3 ) CN, -C ( CH3 ) 2CN, -CH2CN ,
  • R 2 is selected from H, C 1 -C 9 alkyl, C 3 -C 9 cycloalkyl, C 3 -C 9 heterocyclyl, C 6 -C 10 aryl, C 5 -C 10 heteroaryl, - (C 1 -C 6 alkylene)-(C 3 -C 9 cycloalkyl), -(C 1 -C 6 alkylene)-(C 3 -C 9 heterocyclyl), C(O)R b , C(O)N(R a ) 2 , SO 2 R a and SO 2 N(R a ) 2 , the C 1 -C 9 alkyl, C 3 -C 9 cycloalkyl, C 3 -C 9 heterocyclyl, C 6 -C 10 aryl, C 5 -C 10 heteroaryl, -(C 1 -C 6 alkylene)-(C 3 -C 9 cycloalkyl), -(C 1 - C 6 alkylene)-(
  • Each R 5 is the same or different and each is independently selected from H, F, Cl, Br, I, alkyl, haloalkyl, alkoxy, haloalkoxy, -CN, -NH2 , -NO2 , -COOH , -CHO, -OH, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; wherein the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally each independently Ground is selected from alkyl, F, Cl, Br, I, -CN, -NH2 , -NO2 , -OH, hydroxyalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl substituted with one or more substituents in the radical and heteroaryl;
  • R7 is H, F, Cl, Br, I, OH or NH2 ;
  • R8 is F or CN
  • R 9 is H, Cl, F, halogenated C 1 -C 6 alkoxy
  • R 10 is H, Cl, F, halogenated C 1 -C 6 alkoxy
  • R a is selected from H, C 1 -C 6 alkyl, C 2 -C 8 alkenyl, propargyl, C 3 -C 6 cycloalkyl and C 3 -C 6 heterocyclyl, said C 1 -C 6 alkyl, C 2 -C 8 alkenyl, propargyl, C 3 -C 6 cycloalkyl and C 3 -C 6 heterocyclyl optionally substituted with one or more groups independently selected from: F , Cl, Br, I, CN, OH, OCH 3 and SO 2 CH 3 ;
  • R b is selected from H, -O(C 1 -C 3 alkyl), C 1 -C 6 alkyl, C 2 -C 8 alkenyl, propargyl, -(C 1 -C 6 alkylene)- (C 3 -C 6 cycloalkyl), C 3 -C 6 cycloalkyl and C 3 -C 6 heterocyclyl, the -O(C 1 -C 3 alkyl), C 1 -C 6 alkyl , C 2 -C 8 alkenyl, propargyl, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), C 3 -C 6 cycloalkyl and C 3 -C 6 Heterocyclyl is optionally substituted with one or more groups independently selected from: F, Cl , Br, I, CN, -CH2F , -CHF2 , -CF3 , -CH2CF3 , -CH 2CH
  • n 0, 1, 2, 3 or 4;
  • p 1, 2 or 3.
  • the compound represented by formula (I) is further a compound represented by formula (Ib):
  • D is CR 9 or N
  • E is CR 10 or N
  • R7 is H, F, Cl, Br, I, OH or NH2 ;
  • R8 is F or CN
  • R 9 is H, Cl, F, or halogenated C 1 -C 6 alkoxy
  • R 10 is H, Cl, F, or halogenated C 1 -C 6 alkoxy
  • R 8 is F
  • R 9 and R 10 are not H or F at the same time
  • R 11 is Me, F or CH 2 F
  • R 12 is Me, F, CH 2 F, CHF 2 , CF 3 , CH 2 OMe or CH 2 OH;
  • R 13 is H or F
  • R12 and R13 together with the carbon atom to which they are attached form a cyclopropyl, cyclobutyl or oxetane ring;
  • p 1, 2 or 3.
  • the compound represented by formula (I) is further a compound represented by formula (Ic) or formula (Id):
  • D is CR9 and E is CR10 .
  • D is CR9 and E is N; or D is N and E is CR10 .
  • R 9 is Cl; or R 10 is Cl.
  • R 9 is H; or R 10 is H.
  • R 9 is F; or R 10 is F.
  • R 9 is -OCHF 2 or -OCHF 3 .
  • R 10 is -OCHF 2 or -OCHF 3 .
  • R 9 is Cl and R 10 is Cl.
  • R 11 is F.
  • R 12 is F or CH 2 OH.
  • R 13 is F.
  • R8 is F.
  • R7 is H or OH.
  • R7 is H.
  • p is 1.
  • p is 2.
  • the compound represented by the formula (I) is selected from:
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of at least one of the compounds of formula (I), formula (Ib), formula (Ic) or formula (Id) and at least one compound represented by formula (Ib), formula (Ib) or formula (Id).
  • a pharmaceutically acceptable excipient comprising a pharmaceutically acceptable excipient.
  • the present invention also provides the use of the compound represented by formula (I), formula (Ib), formula (Ic) or formula (Id) in preparing a medicament for treating estrogen receptor-related diseases.
  • the estrogen receptor-related disease is a cancer selected from the group consisting of breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and uterine cancer.
  • the present invention also provides a method of treating an estrogen receptor-related disease in a patient, comprising administering to said patient a therapeutically effective amount of Formula (I), Formula (Ib), Formula (Ic), or Formula (Id) above ) compounds shown.
  • the estrogen receptor-related disease is a cancer selected from the group consisting of breast cancer, lung cancer, ovarian cancer, endometrial cancer, prostate cancer, and uterine cancer.
  • halo and halogen as used herein refer to fluorine, chlorine, bromine or iodine.
  • Preferred halogen groups include fluorine, chlorine and bromine.
  • alkyl includes linear or branched monovalent saturated hydrocarbon groups. Alkyl groups as used herein may be optionally substituted with one to more substituents. Non-limiting examples of alkyl groups include, for example, alkyl groups including methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3-( 2-methyl)butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl, 2-methylpentyl and the like.
  • C 1-8 in "C 1-8 alkyl” refers to a linear or branched arrangement containing 1 , 2, 3, 4, 5, 6, 7 or 8 carbon atoms group.
  • Alkenyl and alkynyl groups include straight or branched chain alkenyl and alkynyl groups.
  • C 2-8 alkenyl and “C 2-8 alkynyl” refer to alkenyl groups containing 2, 3, 4, 5, 6, 7 or 8 carbon atoms arranged in a straight or branched chain or alkynyl.
  • Alkoxy refers to the oxyether form of the aforementioned straight or branched chain alkyl groups, ie, -O-alkyl.
  • Haloalkoxy means the aforementioned "alkoxy” is substituted with one or more halogens; non-limiting examples of haloalkoxy groups include, but are not limited to, for example, -OCH2F, - OCHF 2 , -OCF 3 , -OCH 2 CH 2 F, -OCH 2 CHF 2 , -OCH 2 CF 3 , -OCHFCH 3 , -OCF 2 CH 3 , -OCHFCH 2 F and the like.
  • compositions comprising "a” pharmaceutically acceptable excipient can be interpreted to mean that the composition includes “one or more” pharmaceutically acceptable excipients.
  • aromatic ring refers to an unsubstituted or substituted monocyclic, polycyclic or fused-ring aromatic group including carbon atoms, or unsubstituted or substituted including heteroatoms, such as A monocyclic, paracyclic or condensed aromatic group of N, O or S, when it is a polycyclic or condensed ring, at least one ring is aromatic.
  • the aromatic ring is a 5- to 10-membered monocyclic or bicyclic aromatic ring group. Examples of such aromatic rings include, but are not limited to, phenyl, pyridyl, pyrazolyl, pyrimidinyl, chromofuran, indolyl.
  • cycloalkyl refers to monocyclic and polycyclic ring systems containing only carbon atoms in the ring, and which may be optionally substituted with one or more substituents.
  • Cycloalkyl as used herein refers to and includes saturated or unsaturated non-aromatic ring systems.
  • the term cycloalkyl further includes bridged, fused and spiro ring systems.
  • Non-limiting examples of cycloalkyl groups include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, spiro[3.4]octyl, bicyclo[2.2.1]heptane, and the like.
  • heterocyclyl in the present invention, unless otherwise specified, refers to unsubstituted or substituted monocyclic and polycyclic ring systems consisting of carbon atoms and 1-3 heteroatoms selected from N, O or S. , and includes saturated or unsaturated ring systems as well as polycyclic systems having unsaturated and/or aromatic moieties. Wherein nitrogen or sulfur heteroatoms can be selectively oxidized, and nitrogen heteroatoms can be selectively quaternized.
  • the heterocyclyl group can be attached to any heteroatom or carbon atom to form a stable structure. It should be understood that polycyclic heterocycloalkyl groups additionally include fused, bridged, and spiro ring systems.
  • Heterocycloalkyl groups used herein may be optionally substituted with one to more substituents.
  • heterocyclyl groups include, but are not limited to, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidyl, tetrahydrofuranyl, dioxolanyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone and tetrahydro oxadiazolyl.
  • aryl refers to an unsubstituted or substituted monocyclic or polycyclic ring system containing carbon ring atoms, at least one of which is aromatic in nature.
  • Preferred aryl groups are monocyclic or bicyclic 6-10 membered aromatic ring systems. Phenyl and naphthyl are preferred aryl groups. The most preferred aryl group is phenyl.
  • heteroaryl in the present invention, unless otherwise specified, refers to an unsubstituted or substituted stable 5- or 6-membered monocyclic aromatic ring system or an unsubstituted or substituted 9- or 10-membered benzo
  • Heteroaryl groups can be attached to any heteroatom or carbon atom to form a stable structure.
  • heteroaryl groups include, but are not limited to, thienyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridyl Azinyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzothiazolyl, benzothiazolyl, benzene thiadiazolyl, benzotriazolyl adenine, quinolinyl or isoquinolinyl.
  • substituted refers to the replacement of one or more hydrogen atoms in a group with the same or a different substituent, respectively.
  • the substituents are independently selected from the group consisting of -F, -Cl, -Br, -I, -OH, trifluoromethoxy, ethoxy, propoxy, isopropoxy, n-butoxy group, isobutoxy, tert-butoxy, -SCH 3 , -SC 2 H 5 , carboxaldehyde, -C(OCH 3 ), cyano, nitro, -CF 3 , -OCF 3 , amino, dimethyl amino, methylthio, sulfonyl and acetyl groups.
  • substituted alkyl groups include, but are not limited to, 2-aminoethyl, 2-hydroxyethyl, pentachloroethyl, trifluoromethyl, methoxymethyl, pentafluoroethyl, and piperazinylmethyl.
  • substituted alkoxy groups include, but are not limited to, aminomethoxy, trifluoromethoxy, 2-diethylaminoethoxy, 2-ethoxycarbonylethoxy, 3-hydroxypropoxy.
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids.
  • compounds provided herein are acids, their corresponding salts can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic and organic bases.
  • Salts derived from inorganic bases include aluminum, ammonium, calcium, copper (high and low), ferric, ferrous, lithium, magnesium, manganese (high and low), potassium, sodium, zinc, and the like. The ammonium, calcium, magnesium, potassium and sodium salts are particularly preferred.
  • Nontoxic organic bases that can be derivatized into salts include primary, secondary, and tertiary amines, as well as cyclic amines and substituted amines, such as naturally occurring and synthetic substituted amines.
  • Other pharmaceutically acceptable nontoxic organic bases capable of forming salts including ion exchange resins and arginine, betaine, caffeine, choline, N',N'-dibenzylethylenediamine, diethylamine, 2 -Diethylaminoethanol, 2-Dimethylaminoethanol, Ethanolamine, Ethylenediamine, N-Ethylmorpholine, N-Ethylpiperidine, Reduced Glucosamine, Glucosamine, Histidine, Haramine, Isopropylamine , lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resin, procaine, purine, theobromine, triethylamine, trimethylamine, trip
  • acids include, for example, acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, isethionic acid, formic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, Hydroiodic acid, perchloric acid, hydrochloric acid, isethionic acid, propionic acid, glycolic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, oxalic acid, pamoic acid, pantothenic acid, phosphoric acid , succinic acid, sulfuric acid, 2-naphthalenesulfonic acid, cyclohexylaminesulfonic acid, salicylic acid, saccharinic acid,
  • the compound represented by formula (I) will be used as a medicine, it is preferable to use a certain purity, for example, at least 60% pure, more suitably at least 75% pure, particularly suitably at least 98% pure (% by weight) Compare).
  • Prodrugs of the compounds of the present invention are included within the scope of the present invention.
  • the prodrugs refer to functional derivatives that are readily converted into the desired compound in vivo.
  • any pharmaceutically acceptable salt, ester, salt of ester or other derivative of a compound of the present application which, upon administration to a recipient, is capable of providing, directly or indirectly, a compound of the present application or a pharmaceutically active metabolite thereof or Residues.
  • Particularly preferred derivatives or prodrugs are those that increase the bioavailability of the compounds of the present application when administered to a patient (eg, make orally administered compounds more readily absorbed into the bloodstream), or promote the delivery of the parent compound to biological organs or Those compounds that are delivered to the site of action (eg, the brain or lymphatic system). Therefore, the term "administration" in the treatment methods provided by the present invention refers to the administration of the compounds disclosed in the present invention that can treat different diseases, or, although not explicitly disclosed, can be transformed into the disclosed compounds in vivo after administration to a subject compounds of compounds. Conventional methods for the selection and preparation of suitable prodrug derivatives are described in, for example, Design of Prodrugs (Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985).
  • the compounds of the present invention may contain one or more asymmetric centers and may thereby give rise to diastereomers and optical isomers.
  • the present invention includes all possible diastereomers and racemic mixtures thereof, substantially pure resolved enantiomers thereof, all possible geometric isomers and pharmaceutically acceptable salts thereof.
  • the above formula (I) does not exactly define the stereoscopic structure of the compound at a certain position.
  • the present invention includes all stereoisomers of the compounds represented by formula (I) and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers and specific isolated stereoisomers are also included in the present invention. During synthetic procedures to prepare such compounds, or using racemization or epimerization procedures well known to those skilled in the art, the resulting product may be a mixture of stereoisomers.
  • the present invention includes any possible tautomer and pharmaceutically acceptable salts thereof, and mixtures thereof.
  • the present invention includes any possible solvates and polymorphs.
  • the type of solvent that forms the solvate is not particularly limited as long as the solvent is pharmaceutically acceptable.
  • water, ethanol, propanol, acetone and similar solvents can be used.
  • composition refers to a product comprising a specified amount of each of the specified ingredients, as well as any product produced directly or indirectly from a combination of the specified amounts of each of the specified ingredients. Accordingly, pharmaceutical compositions containing the compounds of the present invention as active ingredients and methods of preparing the compounds of the present invention are also part of the present invention. In addition, some of the crystalline forms of the compounds may exist as polymorphs, and such polymorphs are included in the present invention. In addition, some of the compounds may form solvates with water (ie, hydrates) or common organic solvents, and such solvates are also within the scope of this invention.
  • the pharmaceutical composition provided by the present invention comprises a compound represented by formula (I) (or a pharmaceutically acceptable salt thereof) as an active component, a pharmaceutically acceptable excipient and other optional therapeutic components or Accessories.
  • the pharmaceutical compositions of the present invention include oral, rectal, topical and Pharmaceutical compositions for parenteral (including subcutaneous, intramuscular, intravenous) administration.
  • the pharmaceutical compositions of the present invention may conveniently be presented in unit dosage form and prepared by any of the methods of preparation well known in the art of pharmacy.
  • the compound represented by formula (I) of the present invention can be used as an active component, mixed with a pharmaceutical carrier to form a pharmaceutical combination thing.
  • the pharmaceutical carrier can take a wide variety of forms depending on the desired mode of administration, eg, oral or injection (including intravenous). Accordingly, the pharmaceutical compositions of the present invention may be presented in discrete units suitable for oral administration, such as capsules, cachets or tablets containing a predetermined dose of the active ingredient.
  • the pharmaceutical compositions of the present invention may take the form of powders, granules, solutions, aqueous suspensions, non-aqueous liquids, oil-in-water emulsions, or water-in-oil emulsions.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof can also be administered by a controlled release manner and/or a delivery device.
  • the pharmaceutical composition of the present invention can be prepared by any pharmaceutical method. In general, such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more essential ingredients.
  • the pharmaceutical compositions are prepared by uniform intimate admixture of the active ingredient with liquid carriers or finely divided solid carriers, or a mixture of both.
  • the product can be easily prepared to the desired appearance.
  • the pharmaceutical composition of the present invention comprises a pharmaceutically acceptable carrier and a compound represented by formula (I) or its stereoisomer, tautomer, polymorph, solvate, pharmaceutically acceptable Salts and their prodrugs.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, combined with one or more other compounds with therapeutic activity are also included in the pharmaceutical composition of the present invention.
  • the pharmaceutical carrier employed in the present invention can be, for example, a solid carrier, a liquid carrier or a gaseous carrier.
  • Solid carriers include, but are not limited to, lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid.
  • Liquid carriers include, but are not limited to, syrup, peanut oil, olive oil and water.
  • Gaseous carriers including but not limited to carbon dioxide and nitrogen.
  • any pharmaceutically convenient medium can be employed. For example, water, glycols, oils, alcohols, flavor enhancers, preservatives, colorants, etc.
  • oral liquid preparations such as suspensions, elixirs and solutions
  • carriers such as starches, sugars, Microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, etc.
  • oral solid preparations such as powders, capsules and tablets.
  • tablets and capsules are preferred for oral formulations, where solid pharmaceutical carriers are employed.
  • the tablet coating can use standard aqueous or non-aqueous formulation techniques.
  • Tablets containing the compounds or pharmaceutical compositions of the present invention may be formed by compression or molding, optionally together with one or more accessory ingredients or adjuvants to form tablets.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by wetting the powdered compound or pharmaceutical composition with an inert liquid diluent and molding in a suitable machine.
  • each tablet contains about 0.05 mg to 5 g of active ingredient and each cachet or capsule contains about 0.05 mg to 5 g of active ingredient.
  • a formulation intended for oral administration to humans contains from about 0.5 mg to about 5 g of the active ingredient in admixture with suitable and conveniently metered adjunct materials comprising from about 5% to 95% of the total pharmaceutical composition.
  • a unit dosage form generally contains from about 1 mg to about 2 g of the active ingredient, typically 2 mg, 5 mg, 10 mg, 20 mg, 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 1000 mg.
  • the pharmaceutical composition suitable for parenteral administration provided by the present invention can be prepared as an aqueous solution or suspension by adding the active ingredient to water.
  • Suitable surfactants such as hydroxypropylcellulose may be included.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, preservatives may also be included in the pharmaceutical compositions of the present invention to prevent the growth of harmful microorganisms.
  • the present invention provides pharmaceutical compositions suitable for injection, including sterile aqueous solutions or dispersions.
  • the above-mentioned pharmaceutical compositions can be prepared in sterile powder form for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the final injectable form must be sterile and, for ease of injection, must be readily flowable.
  • the pharmaceutical compositions must be stable during manufacture and storage. Therefore, preferably, the pharmaceutical composition is to be preserved under conditions that are resistant to contamination by microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium, for example, water, ethanol, polyol (eg, glycerol, propylene glycol, liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.
  • compositions provided by the present invention may be in a form suitable for topical administration, eg, an aerosol, cream, ointment, lotion, dusting powder, or other similar dosage forms. Further, the pharmaceutical composition provided by the present invention can be in a form suitable for use in a transdermal drug delivery device.
  • These formulations can be prepared by conventional processing methods using the compound represented by formula (I) of the present invention, or a pharmaceutically acceptable salt thereof.
  • a cream or ointment is prepared with the desired consistency by adding about 5 to 10 wt % of a hydrophilic material and water.
  • the pharmaceutical composition provided by the present invention can be in the form of rectal administration by using a solid as a carrier.
  • Unit-dose suppositories are the most typical dosage form. Suitable excipients include cocoa butter and other materials commonly used in the art. Suppositories can be conveniently prepared by first mixing the pharmaceutical composition with softened or melted excipients, then cooling and moulding.
  • the above formulation formulation may also include, as appropriate, one or more additional adjuvant components, such as diluents, buffers, flavoring agents, binders, surfactants, Thickeners, lubricants and preservatives (including antioxidants), etc.
  • additional adjuvant components such as diluents, buffers, flavoring agents, binders, surfactants, Thickeners, lubricants and preservatives (including antioxidants), etc.
  • other adjuvants may also include osmotic enhancers that adjust the isotonic pressure of the drug and blood.
  • the pharmaceutical composition comprising the compound represented by formula (I), or a pharmaceutically acceptable salt thereof, can be prepared in the form of a powder or a concentrated solution.
  • the dosage level of the drug is about 0.01 mg/kg body weight to 150 mg/kg body weight per day, or 0.5 mg to 7 g per patient per day.
  • the effective treatment of the drug is at a dosage level of 0.01 mg/kg body weight to 50 mg/kg body weight per day, or 0.5mg to 3.5g per patient per day.
  • the specific dosage level and treatment regimen for any particular patient will depend on a variety of factors, including the activity of the specific compound used, age, body weight, general health, sex, diet, time of administration, route of administration, excretion rate, drug combination condition and the severity of the specific disease being treated.
  • PTSA p-toluenesulfonic acid
  • n-BuLi 2.5M in hexane
  • n-butyllithium 2.5mol/L n-hexane solution
  • K 2 CO 3 potassium carbonate
  • TFA trifluoroacetic acid
  • Toluene toluene
  • BINAP 1,1'-binaphthyl-2,2'-bisdiphenylphosphine
  • Pd 2 (dba) 3 tris(dibenzylideneacetone)dipalladium
  • t-BuONa sodium tert-butoxide
  • DIEA/DIPEA N,N-diisopropylethylamine
  • I 2 elemental iodine
  • i-PrMgCl isopropyl magnesium chloride (2mol/L tetrahydrofuran solution);
  • TBDPSCl tert-butyldiphenylchlorosilane
  • Tf 2 O trifluoromethanesulfonic anhydride
  • Acetone Acetone
  • TosCl 4-toluenesulfonyl chloride
  • Pd(OH) 2 /C palladium hydroxide supported on carbon
  • TBAF tetrabutylammonium fluoride
  • HOAc glacial acetic acid
  • LC-MS or LCMS Liquid Chromatography-Mass Spectrometry.
  • compound 1-7 (0.200g, 1eq), compound 4-3 (0.390g, 1.2eq), TFA (0.266g, 3eq) were dissolved in toluene (10ml), and heated in an oil bath. The temperature was raised to 120°C and stirred overnight. The reaction solution was concentrated, the residue was dissolved in dichloromethane, washed twice with saturated aqueous sodium bicarbonate solution, the aqueous phase was extracted twice with dichloromethane, the organic phases were combined, dried, concentrated, and the residue was purified by column chromatography to obtain 0.230 g Compound 4-4.
  • the raw material 4-3 is replaced with 5-1.
  • compound 1-7 (0.600g, 1eq), compound 1-8 (0.886g, 2eq), TFA (0.5mL) were dissolved in toluene (10ml), heated in an oil bath and heated to 120 Stir overnight at °C.
  • the reaction solution was concentrated, the residue was dissolved in dichloromethane, washed twice with saturated aqueous sodium bicarbonate solution, the aqueous phase was extracted twice with dichloromethane, the organic phases were combined, dried, concentrated, and the residue was purified by column chromatography to obtain 0.580 g Compounds 1-9.
  • the raw material 1-11 can be replaced with 2-11.
  • Example Example Compounds 14-16 can be synthesized:
  • Celltiter-glo luminescence viability assay kit (Promega, Cat#G7573) was used to detect the cell proliferation inhibitory activity of compounds on breast cancer cell line MCF7.
  • Cells in logarithmic growth phase were taken, trypsinized, seeded in 96-well cell culture plates at a cell density of 2000/well, and incubated overnight at 37°C in a humidified incubator with 5% CO2 .
  • compounds were formulated and serially diluted in DMSO and added to cells in 96-well plates at the following final concentrations: 1000, 333, 111, 37, 12, 4.1, 1.4, 0.45, 0.15 nM.
  • the same volume of DMSO was added to control wells as a solvent control.
  • the degradation activity of the compounds on ER proteins in MCF7 and T47D cells was detected by In-Cell Western method.
  • Cells in logarithmic growth phase were taken, trypsinized and collected, inoculated in 384-well plates and incubated overnight at 37°C, 5% CO 2 .
  • the compound was serially diluted with DMSO, and 120nL/well compound solution was added to the well plate (the final concentration of DMSO was 0.5%), so that the final compound concentration was 100, 33, 11, 3.7, 1.2, 0.41, 0.14, 0.046, 0.015, 0.001 nM and incubated overnight.
  • Cells were fixed, washed with PBS, permeabilized and blocked.
  • Detection method 6 male SD rats (150-300g). Randomly divided into 2 groups with 3 rats in each group. Of the 6 rats in each group, 3 were given a single intravenous injection of 1 mg/kg of the example compound, and the other 3 were given a single intragastric administration of 10 mg/kg of the example compound, respectively, within 5 min (for intravenous administration only) , 15min, 30min, 1h, 2h, 4h, 7h, 24h, blood was collected through the orbital venous plexus, centrifuged to separate the plasma, and stored in a -80°C refrigerator for later use.
  • Mouse PK studies (PO 5 mg/kg) were performed using female BALB/c mice (20-30 g) purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd.
  • the compound concentration was 0.5 mg/mL, as-is.
  • Oral administration was by oral gavage at 10 mL/kg.
  • Blood was drawn through the mouse orbital venous plexus, and 100 ⁇ L of whole blood was transferred to K2 - EDTA tubes per time point. Blood collection time was 30min, 1h, 2h, 4h, 7h and 24h. The collected whole blood samples were centrifuged and plasma separated, and stored in a -80°C refrigerator for later use.

Landscapes

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

Abstract

本发明属于医药领域,具体涉及一种杂芳基并哌啶类衍生物及其药物组合物和应用。本发明提供的如式(I)所示的杂芳基并哌啶类衍生物,不仅可以制备成口服制剂,且具有很好的药代动力学性质。

Description

杂芳基并哌啶类衍生物及其药物组合物和应用 技术领域
本发明属于医药领域,具体涉及一种杂芳基并哌啶类衍生物及其药物组合物和应用。
背景技术
雌激素受体(ER)是一种通过其与内源性雌激素的相互作用介导多种生物效应诱导的配体活化的转录调节蛋白,ER包含ERα和ERβ两种亚型。ERα主要分布在子宫、卵巢、睾丸、垂体、肾、附睾和肾上腺中,而ERβ主要分布在前列腺、卵巢、肺、膀胱、脑和血管中。由于全激动剂或全拮抗剂都有较严重的副作用,选择性雌激素受体调节剂SERM的研究应运而生。其“调节剂”是指SERM在某些组织如骨、肝、心血管系统ERβ集中区中表现为激动剂,而在另外一些组织如乳腺中表现为拮抗剂。其在子宫(ERα较显著区)中可以是激动剂,也可以是拮抗剂。目前已上市的SERM包括他莫昔芬(Tamoxifen)、雷洛昔芬(Raloxifene)、苯卓昔芬(Bazedoxifene)、托瑞米芬(Toremifene)等,但研究发现目前已上市的SERM仍有严重的副作用,如他莫昔芬和托瑞米芬长期服用会引起子宫内膜增生、息肉和子宫内膜癌等,而雷洛昔芬常见的副作用包括潮热、腿痛、乳房触痛和静脉栓塞等。因此研究并开发新型的化合物仍是亟需解决的问题。
选择性雌激素受体下调剂(SERD)是一类能够通过抑制雌激素受体两个转录激活域AF1和AF2的功能而阻断雌激素的活性的药物,是一类经典的抗雌激素类药物(完全拮抗剂)。氟维司群是目前被批准用于临床使用的唯一SERD类药物,用于治疗ER+乳腺癌,但其成药特性差,快速新陈代谢并且必须通过每月肌肉注射来施用,与体外研究中见到的完全ER降解相比,其限制了ER的有效降解(在临床样品中~50%ER降解)。
临床上对能够抑制雌激素受体活性、下调雌激素受体表达水平或诱导雌激素受体降解的药物需求强烈,以改善针对早期、转移性、或耐药性的乳腺癌或其它 与雌激素受体过度活性相关疾病的治疗效果。
AstraZeneca公司的WO2018077630A专利公开了一系列SERD类化合物,包括临床早期药物AZD9833,其中AZD9833的结构为:
Figure PCTCN2022075486-appb-000001
选择性雌激素受体下调剂已显示一定治疗优势,但仍然需要开发更多的可口服的选择性雌激素受体,使得候选药物具有更加优异的特性,如疗效更好、副作用更低、口服吸收更好、更优的药代动力学特性等,从而更好的用于预防或治疗雌激素受体相关疾病。
发明内容
为了解决上述问题,本发明提供一种药代动力学性质更好的口服选择性雌激素受体类化合物及其药物组合物和应用。
本发明提供了一种如式(I)所示化合物、其立体异构体、互变异构体或药学上可接受的盐:
Figure PCTCN2022075486-appb-000002
其中,
环A选自
Figure PCTCN2022075486-appb-000003
D是CR 9或N;
E是CR 10或N;
R 1选自H、F、Cl、Br、I、-CN、-CH 3、-CH 2CH 3、-CH(CH 3) 2、-CH 2CH(CH 3) 2、-CH 2OH、-CH 2OCH 3、CH 2CH 2OH、-C(CH 3) 2OH、-CH(OH)CH(CH 3) 2、-C(CH 3) 2CH 2OH、-CH 2CH 2SO 2CH 3、-CH 2OP(O)(OH) 2、-CH 2F、-CHF 2、-CH 2NH 2、-CH 2NHSO 2CH 3、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CF 3、-CH 2CF 3、-CH 2CHF 2、-CH(CH 3)CN、-C(CH 3) 2CN、-CH 2CN、-CO 2H、-COCH 3、-CO 2CH 3、-CO 2C(CH 3) 3、-COCH(OH)CH 3、-CONH 2、-CONHCH 3、-CONHCH 2CH 3、-CONHCH(CH 3) 2、-CON(CH 3) 2、-C(CH 3) 2CONH 2、-NH 2、-NHCH 3、-N(CH 3) 2、-NHCOCH 3、-N(CH 3)COCH 3、-NHS(O) 2CH 3、-N(CH 3)C(CH 3) 2CONH 2、-N(CH 3)CH 2CH 2S(O) 2CH 3、-NO 2、=O、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2OCH 3、-OCH 2CH 2OH、-OCH 2CH 2N(CH 3) 2、-OP(O)(OH) 2、-S(O) 2N(CH 3) 2、-SCH 3、-S(O) 2CH 3、-S(O) 2OH、环丙基、环丙基酰胺基、环丁基、氧杂环丁烷基、氮杂环丁烷基、(1-甲基氮杂环丁烷-3-基)氧基、N-甲基-N-氧杂环丁烷-3-基氨基、氮杂环丁烧-1-基甲基、苄基氧基苯基、吡咯烷-1-基、吡咯烷-1-基-甲酮基、哌嗪-1-基、吗啉代甲基、吗啉代-甲酮基和吗啉代;
R 2选自H、C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、-(C 1-C 6亚烷基)-(C 3-C 9环烷基)、-(C 1-C 6亚烷基)-(C 3-C 9杂环基)、C(O)R b、C(O)N(R a) 2、SO 2R a和SO 2N(R a) 2,所述C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、-(C 1-C 6亚烷基)-(C 3-C 9环烷基)、-(C 1-C 6亚烷基)-(C 3-C 9杂环基)任选取代有一个或多个F、Cl、Br、I、CN、OR a、N(R a) 2、C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、C(O)R b、C(O)N(R a) 2、SO 2R a和SO 2N(R a) 2
R 5各自相同或不同,其各自独立地选自H、F、Cl、Br、I、烷基、卤代烷基、烷氧基、卤代烷氧基、-CN、-NH 2、-NO 2、-COOH、-CHO、-OH、羟烷基、环烷基、杂环基、芳基和杂芳基;其中所述烷基、环烷基、杂环基、芳基和杂芳基任选各自独立地被选自烷基、F、Cl、Br、I、-CN、-NH 2、-NO 2、-OH、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R 7是H、F、Cl、Br、I、OH或NH 2
R 8是F或CN;
R 9是H、Cl、F、卤代的C 1-C 6烷氧基;
R 10是H、Cl、F、卤代的C 1-C 6烷氧基;
R a选自H、C 1-C 6烷基、C 2-C 8烯基、炔丙基、C 3-C 6环烷基和C 3-C 6杂环基,所述C 1-C 6烷基、C 2-C 8烯基、炔丙基、C 3-C 6环烷基和C 3-C 6杂环基任选取代有一个或多个独立选自以下的基团:F、Cl、Br、I、CN、OH、OCH 3和SO 2CH 3
R b选自H、-O(C 1-C 3烷基)、C 1-C 6烷基、C 2-C 8烯基、炔丙基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)、C 3-C 6环烷基和C 3-C 6杂环基,所述-O(C 1-C 3烷基)、C 1-C 6烷基、C 2-C 8烯基、炔丙基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)、C 3-C 6环烷基和C 3-C 6杂环基任选取代有一个或多个独立选自以下的基团:F、Cl、Br、I、CN、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CHF 2、-CH 2CH 2F、-OH、-OCH 3和-SO 2CH 3
m为0、1、2、3或4;
p为1、2或3。
一些实施方式中,式(Ⅰ)所示化合物进一步地为式(Ⅰb)所示化合物:
Figure PCTCN2022075486-appb-000004
其中,
D是CR 9或N;
E是CR 10或N;
R 7是H、F、Cl、Br、I、OH或NH 2
R 8是F或CN;
R 9是H、Cl、F、或卤代的C 1-C 6烷氧基;
R 10是H、Cl、F、或卤代的C 1-C 6烷氧基;
当R 8是F时,R 9、R 10不同时为H或F;
当D是N、E是CR 10且R 8是F时,R 10不为H或F;或者当E是N、D是CR 9且R 8是F时,R 9不为H或F;
R 11是Me、F或CH 2F;
R 12是Me、F、CH 2F、CHF 2、CF 3、CH 2OMe或CH 2OH;
R 13是H或F;或者
R 12和R 13连同它们附接至其上的碳原子一起形成环丙基环、环丁基环或氧杂环丁烷环;
p为1、2或3。
一些实施方式中,式(Ⅰ)所示化合物进一步地为式(Ⅰc)或式(Ⅰd)所示化合物:
Figure PCTCN2022075486-appb-000005
一些实施方式中,D是CR 9,E是CR 10
一些实施方式中,D是CR 9,E是N;或者D是N,E是CR 10
一些实施方式中,R 9是Cl;或R 10是Cl。
一些实施方式中,R 9是H;或R 10是H。
一些实施方式中,R 9是F;或R 10是F。
一些实施方式中,R 9是-OCHF 2或-OCHF 3
一些实施方式中,R 10是-OCHF 2或-OCHF 3
一些实施方式中,R 9是Cl,且R 10是Cl。
一些实施方式中,R 11是F。
一些实施方式中,R 12是F或CH 2OH。
一些实施方式中,R 13是F。
一些实施方式中,R 8是F。
一些实施方式中,R 7是H或OH。
一些实施方式中,R 7是H。
一些实施方式中,p为1。
一些实施方式中,p为2。
一些实施方式中,所述式(I)所示化合物选自:
5-(二氟甲氧基)-N-(1-(3-氟丙基)氮杂环丁烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
N-(3-(二氟甲氧基)-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
1-(3-氟丙基)-N-(4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)-3-(三氟甲氧基)苯基)氮杂环丁烷-3-胺;
5-(二氟甲氧基)-N-((S)-1-(3-氟丙基)吡咯烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
5-(二氟甲氧基)-N-((R)-1-(3-氟丙基)吡咯烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
(3R,4R)-4-((5-(二氟甲氧基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-基)氨基)-1-(3-氟丙基)吡咯烷-3-醇;
N-(3-(二氟甲氧基)-5-氟-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
3-((6S,8R)-6-(2-(二氟甲氧基)-6-氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
3-((6S,8R)-6-(3-(二氟甲氧基)-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
3-((6S,8R)-6-(2-(二氟甲氧基)-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
(S)-N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)吡咯烷-3-胺;
(3R,4R)-4-((3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)氨基)-1-(3-氟丙基)吡咯烷-3-醇;
(R)-N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)吡咯烷-3-胺;或
3-((6S,8R)-6-(2,6-二氯-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇。
本发明还提供了一种药物组合物,所述药物组合物包含治疗有效量的至少一种所述式(I)、式(Ib)、式(Ic)或式(Id)所示化合物和至少一种药学上可接受的辅料。
本发明还提供了式(I)、式(Ib)、式(Ic)或式(Id)所示化合物在制备用于治疗雌激素受体相关疾病的药物中的用途。
一些实施方式中,所述雌激素受体相关疾病为选自以下的癌症:乳腺癌、肺癌、卵巢癌、子宫内膜癌、前列腺癌和子宫癌。
本发明还提供了一种在患者中治疗雌激素受体相关疾病的方法,其包括向所述患者给予治疗有效量的上述式(I)、式(Ib)、式(Ic)或式(Id)所示化合物。
一些实施方式中,所述雌激素受体相关疾病为选自以下的癌症:乳腺癌、肺癌、卵巢癌、子宫内膜癌、前列腺癌和子宫癌。
定义
上述结构通式中使用的一般化学术语具有通常的含义。
例如,除非另有说明,本发明所用的术语“卤代”和“卤素”是指氟、氯、溴或碘。优选的卤素基团包括氟、氯和溴。
在本发明中,除非另有说明,“烷基”包括直链或支链的一价饱和烃基。本 文中使用的烷基基团可以任选地被一至多个取代基取代。烷基基团的非限制性实例包括例如烷基包括甲基、乙基、丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、3-(2-甲基)丁基、2-戊基、2-甲基丁基、新戊基、正己基、2-己基、2-甲基戊基等。类似的,“C 1-8烷基”中的“C 1-8”是指包含有1、2、3、4、5、6、7或8个碳原子的直链或支链形式排列的基团。
烯基和炔基包括直链或支链的烯基和炔基。同样地,“C 2-8烯基”和“C 2-8炔基”是指含有2、3、4、5、6、7或者8个碳原子以直链或支链形式排列的烯基或炔基。
“烷氧基”是指前述的直链或支链烷基的氧醚形式,即-O-烷基。
“卤代的烷氧基”是指前述的“烷氧基”被1个或多个卤素取代;卤代的烷氧基基团的非限制性实例包括但不限于例如-OCH 2F、-OCHF 2、-OCF 3、-OCH 2CH 2F、-OCH 2CHF 2、-OCH 2CF 3、-OCHFCH 3、-OCF 2CH 3、-OCHFCH 2F等。
在本发明中,“一”、“一个”、“该”、“至少一个”和“一个或多个”可互换使用。因此,例如,包含“一种”药学上可接受的赋形剂的组合物可以被解释为表示该组合物包括“一种或多种”药学上可接受的赋形剂。
术语“芳香环”,在本发明中,除非另有说明,是指未取代或取代的包括碳原子的单环、并环或稠环芳香基团,或者未取代或取代的包括杂原子,例如N、O或S的单环、并环或稠环芳香基团,当为并环或稠环时,至少有一个环具有芳香性。优选芳香环为5到10元的单环或双环的芳香环基团。这些芳香环的实例包括但不限于苯基、吡啶基、吡唑基、嘧啶基、苯并二氢呋喃、吲哚基。
术语“环烷基”,是指在环中仅含碳原子的单环和多环系统,并且可以任选地被一至多个取代基取代。本文中使用的环烷基是指并且包括饱和的或不饱和的非芳香的环状系统。术语环烷基另外包括桥环、稠环和螺环的环状系统。环烷基的非限制性实例包括例如环丙基、环丁基、环戊基、环己基、环己烯基、螺[3.4]辛基、双环[2.2.1]庚烷等。
术语“杂环基”,在本发明中,除非另有说明,是指由碳原子和1-3个选自N、O或S的杂原子组成的未取代或取代的单环和多环系统,并且包括饱和的或不饱和的环状系统以及具有不饱和部分和/或芳香部分的多环系统。其中氮或硫杂原子可以选择性地被氧化,并且氮杂原子可以选择性地被季铵化。该杂环基可 以被连接到任何的杂原子或碳原子上以形成稳定的结构。应该理解,多环杂环烷基基团另外包括稠环、桥环和螺环的环状系统。本文中使用的杂环烷基基团可以任选地被一至多个取代基取代。这些杂环基的实例包括但不限于氮杂环丁烷基、吡咯烷基、哌啶基、哌嗪基、氧代哌嗪基、氧代哌啶基、四氢呋喃基、二氧戊环基、四氢咪唑基、四氢噻唑基、四氢恶唑基、四氢吡喃基、吗啉基、硫代吗啉基、硫代吗啉基亚砜、硫代吗啉基砜基和四氢恶二唑基。
除非另有说明,本文所用的术语“芳基”是指含有碳环原子的未取代或取代的单环或多环环系,至少一个环具有芳香性。优选的芳基是单环或双环6-10元芳环系统。苯基和萘基是优选的芳基。最优选的芳基是苯基。
术语“杂芳基”,在本发明中,除非另有说明,是指未取代或取代的稳定的5元或6元单环芳族环系统或未取代或取代的9元或10元苯并稠合杂芳族环系统或双环杂芳族环系统,其由碳原子和1-4个选自N、O或S的杂原子组成,并且其中所述氮或硫杂原子可以选择性地被氧化,所述氮杂原子可以选择性地被季铵化。杂芳基可以连接在任何杂原子或碳原子上以形成稳定的结构。杂芳基的实例包括但不限于噻吩基、呋喃基、咪唑基、异恶唑基、恶唑基、吡唑基、吡咯基、噻唑基、噻二唑基、三唑基、吡啶基、哒嗪基、吲哚基、氮杂吲哚基、吲唑基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并异恶唑基、苯并噻唑基、苯并噻唑基、苯并噻二唑基、苯并三唑基腺嘌呤、喹啉基或异喹啉基。
术语“取代的”是指基团中的一个或多个氢原子分别被相同的或者不同的取代基所取代。典型的取代基包括但不限于卤素(F、Cl、Br或I)、C 1-8烷基、C 3-12环烷基、-OR 1、-SR 1、=O、=S、-C(O)R 1、-C(S)R 1、=NR 1、-C(O)OR 1、-C(S)OR 1、-NR 1R 2、-C(O)NR 1R 2、氰基、硝基、-S(O) 2R 1、-O-S(O 2)OR 1、-O-S(O) 2R 1、-OP(O)(OR 1)(OR 2);其中R 1和R 2独立地选自-H、C 1-6烷基、C 1-6卤代烷基。在一些实施例中,取代基独立地选自包含-F、-Cl、-Br、-I、-OH、三氟甲氧基、乙氧基、丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基、-SCH 3、-SC 2H 5、甲醛基、-C(OCH 3)、氰基、硝基、-CF 3、-OCF 3、氨基、二甲基氨基、甲硫基、磺酰基和乙酰基的基团。
取代烷基的实例包括但不限于2-氨基乙基、2-羟乙基、五氯乙基、三氟甲基、甲氧基甲基、五氟乙基和哌嗪基甲基。
取代烷氧基的实例包括但不限于氨基甲氧基、三氟甲氧基、2-二乙基氨基乙氧基、2-乙氧基羰基乙氧基、3-羟基丙氧基。
术语“药学上可接受的盐”是指从药学上可接受的无毒的碱或酸制备的盐。当本发明提供的化合物是酸时,可以从药学上可接受的无毒的碱,包括无机碱和有机碱,方便地制得其相应的盐。从无机碱衍生的盐包括铝、铵、钙、铜(高价和低价)、三价铁、亚铁、锂、镁、锰(高价和低价)、钾、钠、锌之类的盐。特别优选铵、钙、镁、钾和钠的盐。药学上可接受的能够衍生成盐的无毒有机碱包括伯胺、仲胺和叔胺,也包括环胺及含有取代基的胺,如天然存在的和合成的含取代基的胺。能够成盐的其他药学上可接受的无毒有机碱,包括离子交换树脂以及精氨酸、甜菜碱、咖啡因、胆碱、N',N'-二苄乙二胺、二乙胺、2-二乙氨基乙醇、2-二甲胺基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、还原葡萄糖胺、氨基葡萄糖、组氨酸、哈胺、异丙胺、赖氨酸,甲基葡萄糖胺、吗啉、哌嗪、哌啶、多胺树脂、普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨丁三醇等。
当本发明提供的化合物是碱时,可以从药学上可接受的无毒的酸,包括无机酸和有机酸,方便制得其相应的盐。这样的酸包括,如,醋酸、苯磺酸、苯甲酸、樟脑磺酸、柠檬酸、乙磺酸、羟乙基磺酸、甲酸、富马酸、葡萄糖酸、谷氨酸、氢溴酸、氢碘酸、高氯酸、盐酸、羟乙磺酸、丙酸、乙醇酸、乳酸、马来酸、苹果酸、扁桃酸、甲磺酸、黏酸、硝酸、草酸、扑酸、泛酸、磷酸、琥珀酸、硫酸、2-萘磺酸、环己胺磺酸、水杨酸、糖精酸、三氟乙酸、酒石酸和对甲苯磺酸等。较优地,柠檬酸、氢溴酸、甲酸、盐酸、马来酸、磷酸、硫酸和酒石酸。更优地,甲酸和盐酸。
由于式(I)所示化合物将作为药物应用,较优地,使用一定纯度,例如,至少为60%纯度,比较合适的纯度为至少75%,特别合适地纯度为至少98%(%是重量比)。
本发明化合物的药物前体包含在本发明的保护范围内。通常,所述药物前体是指很容易在体内转化成所需化合物的功能性衍生物。例如,本申请化合物的任何药学上可接受的盐、酯、酯的盐或其它衍生物,其在向受体施用后能够直接或间接地提供本申请的化合物或其具有药学活性的代谢物或残基。特别优选的衍生 物或前药是在施用于患者时可以提高本申请化合物生物利用度的那些化合物(例如,可以使口服的化合物更易于被吸收到血液中),或者促进母体化合物向生物器官或作用位点(例如脑部或淋巴系统)递送的那些化合物。因此,本发明提供的治疗方法中的术语“给药”是指施用能治疗不同疾病的本发明公开的化合物,或虽未明确公开但对受试者给药后能够在体内转化为本发明公开的化合物的化合物。有关选择和制备合适药物前体衍生物的常规方法,已记载在例如《药物前体设计》(Design of Prodrugs,ed.H.Bundgaard,Elsevier,1985)这类书中。
显然的,一个分子中任何取代基或特定位置的变量的定义是独立于分子中其他位置的。很容易理解,本领域技术人员可以通过现有技术手段及本发明中所述的方法来选择本发明中的化合物的取代基或取代形式,以获得化学上稳定且易于合成的化合物。
本发明所述化合物可能含有一个或多个不对称中心,并可能由此产生非对映异构体和光学异构体。本发明包括所有可能的非对映异构体及其外消旋混合物、其基本上纯的拆分对映异构体、所有可能的几何异构体及其药学上可接受的盐。
上述式(I)没有确切定义该化合物某一位置的立体结构。本发明包括式(I)所示化合物的所有立体异构体及其药学上可接受的盐。进一步地,立体异构体的混合物及分离出的特定的立体异构体也包括在本发明中。制备此类化合物的合成过程中,或使用本领域技术人员公知的外消旋化或差向异构化的过程中,制得的产品可以是立体异构体的混合物。
当式(I)所示化合物存在互变异构体时,除非特别声明,本发明包括任何可能的互变异构体和其药学上可接受的盐,及它们的混合物。
当式(I)所示化合物及其药学上可接受的盐存在溶剂化物或多晶型时,本发明包括任何可能的溶剂化物和多晶型。形成溶剂化物的溶剂类型没有特别的限定,只要该溶剂是药学上可以接受的。例如,水、乙醇、丙醇、丙酮等类似的溶剂都可以采用。
术语“组合物”,在本发明中,是指包括包含指定量的各指定成分的产品,以及直接或间接地由指定量的各指定成分的组合生产的任何产品。因此,含有本发明的化合物作为活性成分的药物组合物以及制备本发明化合物的方法也是本发明的一部分。此外,化合物的一些结晶形式可以多晶型存在,并且此多晶型包 括在本发明中。另外,一些化合物可以与水(即水合物)或常见的有机溶剂形成溶剂化物,并且此类溶剂化物也落入本发明的范围内。
本发明提供的药物组合物包括作为活性组分的式(I)所示化合物(或其药学上可接受的盐)、一种药学上可接受的赋形剂及其他可选的治疗组分或辅料。尽管任何给定的情况下,最适合的活性组分给药方式取决于接受给药的特定的主体、主体性质和病情严重程度,但是本发明的药物组合物包括适于口腔、直肠、局部和不经肠道(包括皮下给药、肌肉注射、静脉给药)给药的药物组合物。本发明的药物组合物可以方便地以本领域公知的单位剂型存在和药学领域公知的任何制备方法制备。
实际上,根据常规的药物混合技术,本发明式(I)所示化合物,或药物前体,或代谢物,或药学上可接受的盐,可以作为活性组分,与药物载体混合成药物组合物。所述药物载体可以采取各种各样的形式,这取决于期望采用的给药方式,例如,口服或注射(包括静脉注射)。因此,本发明的药物组合物可以采用适于口服给药的独立单元,如包含预定剂量的活性组分的胶囊剂、扁囊剂或片剂。进一步地,本发明的药物组合物可采用粉末、颗粒、溶液、水性悬浮液、非水液体、水包油型乳液,或油包水型乳液形式。另外,除了上述提到的常见的剂型,式(I)所示化合物或其药学上可接受的盐,也可以通过控释的方式和/或输送装置给药。本发明的药物组合物可以采用任何制药学上的方法制备。一般情况下,这种方法包括使活性组分和组成一个或多个必要成分的载体缔合的步骤。一般情况下,所述药物组合物经由活性组分与液体载体或精细分割的固体载体或两者的混合物经过统一的密切的混合制得。另外,该产品可以方便地制备成所需要的外观。
因此,本发明的药物组合物包括药学上可接受的载体和式(I)所示化合物或其立体异构体、互变异构体,多晶型物、溶剂化物、其药学上可接受的盐、其药物前体。式(I)所示化合物或其药学上可接受的盐,与其他一种或多种具有治疗活性的化合物的联合用药也包括在本发明的药物组合物中。
本发明采用的药物载体可以是,例如,固体载体、液体载体或气体载体。固体载体,包括但不限于乳糖、石膏粉、蔗糖、滑石粉、明胶、琼脂、果胶、阿拉伯胶、硬脂酸镁、硬脂酸。液体载体,包括但不限于糖浆、花生油、橄榄油和水。 气体载体,包括但不限于二氧化碳和氮气。制备药物口服制剂时,可以使用任何制药学上方便的介质。例如,水、乙二醇、油类、醇类、增味剂、防腐剂、着色剂等可用于口服的液体制剂如悬浮剂、酏剂和溶液剂;而载体,如淀粉类、糖类、微晶纤维素、稀释剂、造粒剂、润滑剂、粘合剂、崩解剂等可用于口服的固体制剂如散剂、胶囊剂和片剂。考虑到易于施用,口服制剂首选片剂和胶囊,在此应用固体药学载体。可选地,片剂包衣可使用标准的水制剂或非水制剂技术。
含有本发明化合物或药物组合物的片剂可通过压缩或模塑成型,可选地,可以与一种或多种辅助组分或辅药一起制成片剂。活性组分以自由流动的形式如粉末或颗粒,与粘合剂、润滑剂、惰性稀释剂、表面活性剂或分散剂混合,在适当的机器中,通过压缩可以制得压缩片。用一种惰性液体稀释剂浸湿粉末状的化合物或药物组合物,然后在适当的机器中,通过模塑可以制得模塑片。较优地,每个片剂含有大约0.05mg到5g的活性组分,每个扁囊剂或胶囊剂含有大约0.05mg到5g的活性组分。例如,拟用于人类口服给药的配方包含约0.5mg到约5g的活性组分,与合适且方便计量的辅助材料复合,该辅助材料约占药物组合物总量的5%至95%。单位剂型一般包含约1mg到约2g的活性组分,典型的是2mg、5mg、10mg、20mg、25mg、50mg、100mg、200mg、300mg、400mg、500mg、600mg、800mg或1000mg。
本发明提供的适用于胃肠外给药的药物组合物可将活性组分加入水中制备成水溶液或悬浮液。可以包含适当的表面活性剂如羟丙基纤维素。在甘油、液态聚乙二醇,及其在油中的混合物,也可以制得分散体系。进一步地,防腐剂也可以包含在本发明的药物组合物中用于防止有害的微生物生长。
本发明提供适用于注射的药物组合物,包括无菌水溶液或分散体系。进一步地,上述药物组合物可以制备成无菌粉末形式以用于即时配制无菌注射液或分散液。无论如何,最终的注射形式必须是无菌的,且为了易于注射,必须是易于流动的。此外,所述药物组合物在制备和储存过程中必须稳定。因此,优选地,所述药物组合物要在抗微生物如细菌和真菌污染的条件下保存。载体可以是溶剂或分散介质,例如,水、乙醇、多元醇(如甘油、丙二醇、液态聚乙二醇)、植物油及其适当的混合物。
本发明提供的药物组合物可以是适于局部用药的形式,例如,气溶胶、乳剂、 软膏、洗液、撒粉或其他类似的剂型。进一步地,本发明提供的药物组合物可以采用适于经皮给药设备使用的形式。利用本发明式(I)所示化合物,或其药学上可接受的盐,通过常规的加工方法,可以制备这些制剂。作为一个例子,乳剂或软膏通过加入约5wt%到10wt%的亲水性材料和水,制得具有预期一致性的乳剂或软膏。
本发明提供的药物组合物,可以以固体为载体,适用于直肠给药的形式。单位剂量的栓剂是最典型的剂型。适当的辅料包括本领域常用的可可脂和其他材料。栓剂可以方便地制备,首先药物组合物与软化或熔化的辅料混合,然后冷却和模具成型而制得。
除了上述提到的辅料组分外,上述制剂配方还可以包括,适当的,一种或多种附加的辅料组分,如稀释剂、缓冲剂、调味剂、粘合剂、表面活性剂、增稠剂、润滑剂和防腐剂(包括抗氧化剂)等。进一步地,其他的辅药还可以包括调节药物与血液等渗压的促渗剂。包含式(I)所示化合物,或其药学上可接受的盐的药物组合物,可以制备成粉剂或浓缩液的形式。
一般情况下,治疗上述所示的状况或不适,药物的剂量水平约为每天0.01mg/kg体重到150mg/kg体重,或者每个病人每天0.5mg到7g。例如,炎症、癌症、牛皮癣、过敏/哮喘、免疫系统的疾病和不适、中枢神经系统(CNS)的疾病和不适,有效治疗的药物剂量水平为每天0.01mg/kg体重到50mg/kg体重,或者每个病人每天0.5mg到3.5g。
但是,可以理解,可能需要比上述那些更低或更高的剂量。任何特定病人的具体剂量水平和治疗方案将取决于多种因素,包括所用具体化合物的活性、年龄、体重、综合健康状况、性别、饮食、给药时间、给药途径、排泄率、药物联用的情况和接受治疗的特定疾病的严重程度。
具体实施方式
为使上述内容更清楚、明确,本发明将用以下实施例来进一步阐述本发明的技术方案。以下实施例仅用于说明本发明的具体实施方式,以使本领域的技术人员能够理解本发明,但不用于限制本发明的保护范围。本发明的具体实施方式中,未作特别说明的技术手段或方法等为本领域的常规技术手段或方法等。
除非另有说明,本发明所有的一部分和百分比均按重量计算,所有温度均指摄氏度。
实施例中使用了下列缩略语:
DHP:3,4-二氢-2H-吡喃;
PTSA:对甲苯磺酸;
DCM:二氯甲烷;
n-BuLi(2.5M in hexane):正丁基锂(2.5mol/L的正己烷溶液);
HCl(4M in dioxane):盐酸(4mol/L的1,4-二氧六环溶液);
EA:乙酸乙酯;
K 2CO 3:碳酸钾;
CH 3CN/ACN:乙腈;
TFA:三氟乙酸;
Toluene:甲苯;
BINAP:1,1'-联萘-2,2'-双二苯膦;
Pd 2(dba) 3:三(二亚苄基丙酮)二钯;
t-BuONa:叔丁醇钠;
DIEA/DIPEA:N,N-二异丙基乙胺;
DMF:N,N-二甲基甲酰胺;
THF:四氢呋喃;
I 2:单质碘;
Na 2CO 3:碳酸钠;
i-PrMgCl(2M in THF):异丙基氯化镁(2mol/L的四氢呋喃溶液);
BrettPhos Pd G3:甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II);
NaH:氢化钠;
TBDPSCl:叔丁基二苯基氯硅烷;
Tf 2O:三氟甲磺酸酐;
2,6-Lutidine:2,6-二甲基吡啶;
KI:碘化钾;
Cs 2CO 3:碳酸铯;
CbzCl:氯甲酸苄酯;
Acetone:丙酮;
Et 3N/TEA:三乙胺;
TosCl:4-甲苯磺酰氯;
Pd(OH) 2/C:碳负载氢氧化钯;
TBAF:四丁基氟化铵;
HOAc:冰乙酸;
LC-MS或LCMS:液相色谱-质谱。
中间体1-7的制备
Figure PCTCN2022075486-appb-000006
步骤1:化合物1-2的合成
向500ml单口瓶中,加入化合物1-1(24.00g,1eq),DCM(200mL),DHP(15.21g,1.5eq),PTSA(2.08g,0.1eq)。室温下搅拌反应过夜。反应完毕后,用饱和碳酸氢钠溶液洗涤两次,有机相干燥,浓缩,柱层析纯化(PE:EA=100:0-50:50),得33.12g化合物1-2。
LC-MS[M+H] +:283。
步骤2:化合物1-4的合成
向1000mL三口瓶中,加入化合物1-2(23.5g,1eq),THF(200mL),氮气保护,降温至-70℃。向其中滴加n-BuLi(2.5M in hexane)(50mL,1.5eq),-70℃下搅拌0.5小时。向其中滴加1-3(29.54g,1.5eq)溶于THF(200mL)的溶液,滴加过程中控制内温在-50℃以下,滴加完毕后,缓慢升温至室温并搅拌2小时。
反应完毕后,用饱和氯化铵水溶液淬灭反应,乙酸乙酯萃取,浓缩,柱层析纯化(PE:EA=100:0-60:40),得26.80g化合物1-4。
LC-MS[M+H] +:362。
步骤3:化合物1-5的合成
向500mL单口瓶中,加入化合物1-4(10.00g,1eq),EA(80mL),HCl(4M in dioxane,35mL,5eq),室温下搅拌反应。反应完毕后,过滤反应液,滤饼烘干,得5.83g化合物1-5。
LC-MS[M+H] +:176。
步骤4:化合物1-7的合成
于50ml单口瓶中,将化合物1-5(0.500g,1eq),化合物1-6(0.786g,1.2eq),无水碳酸钾(1.170g,3eq)溶于乙腈(10ml)中,油浴锅加热升温至70℃搅拌过夜。将反应液过滤,滤饼用乙腈淋洗,收集滤液浓缩,残余物溶于二氯甲烷中,水洗两次,无水硫酸钠干燥,过滤,滤液柱层析纯化,浓缩得到0.600g化合物1-7。
LC-MS[M+H] +:258。
实施例3:化合物3的制备
Figure PCTCN2022075486-appb-000007
Figure PCTCN2022075486-appb-000008
步骤1:化合物3-2的合成
向500ml单口瓶中,加入化合物3-1(16.00g,1eq),H 2O(160mL),Na 2CO 3(29.24g,3eq),I 2(12.84g,1.1eq)。室温下搅拌反应过夜。反应完毕后,用2mol/L的HCl溶液调节反应液PH=1-2,滴加过程中析出固体,搅拌0.5小时,过滤,滤饼用水洗2-3次,收集滤饼,干燥,得15.70g化合物3-2。
LC-MS[M+H] +:300/302。
步骤2:化合物3-4的合成
向250ml单口瓶中,加入化合物3-2(10.00g,1eq),乙腈(50mL),K 2CO 3(13.83g,3eq),化合物3-3(7.63g,1.5eq)。85℃搅拌反应6小时,反应完毕后,过滤,滤液浓缩,柱层析纯化(PE:EA=100:0-70:30),得7.50g化合物3-4。
LC-MS[M+H] +:350/352。
步骤3:化合物3-5的合成
向50ml三口瓶中,加入化合物3-4(1.37g,1eq),THF(14mL),氮气保护,降温至0℃。向其中滴加i-PrMgCl(2.0M in THF)(3.9mL,2eq),滴加完毕后,0℃下搅拌反应1小时。向其中加入DMF(0.859g,3eq),加完后,0℃搅拌反应1小时。反应完毕后,用饱和氯化铵水溶液淬灭反应,EA萃取,有机相浓缩,柱层析纯化(PE:EA=100:0-60:40),得0.25g化合物3-5。
LC-MS[M+H] +:252/254。
步骤4:化合物3-6的合成
向50ml单口瓶中,加入化合物3-5(0.16g,1eq),Toluene(5mL),化合物1-7(0.16g,1eq),TFA(0.22g,3eq),120℃搅拌反应2小时。反应完毕后,EA稀释,有机相用饱和碳酸氢钠溶液洗涤,浓缩,得0.20g化合物3-6。
LC-MS[M+H] +:493/495。
步骤5:化合物3-8的合成
向100ml单口瓶中,加入化合物3-7(2.00g,1eq),CH 3CN(20mL),化合物1-14(2.40g,1.1eq),K 2CO 3(4.82g,3eq),H 2O(5mL)。70℃搅拌反应过夜,反应完毕后,浓缩反应液,残余物加DCM/H 2O萃取,有机相水洗、浓缩,得2.30g化合物3-8。
LC-MS[M+H] +:233。
步骤6:化合物3-9的合成
向250ml单口瓶中,加入化合物3-8(2.30g,1eq),DCM(30mL),HCl(4M in dioxane)(30mL)。室温搅拌反应过夜,反应完毕后,过滤,干燥滤饼,得1.60g化合物3-9。
LC-MS[M+H] +:133。
步骤7:化合物3的合成
向10ml单口瓶中,加入化合物3-6(0.15g,1eq),化合物3-9(125mg,2eq),BrettPhos Pd G3(69mg,0.25eq),t-BuONa(293mg,10eq),1,4-dioxane(5mL)。升温至70℃搅拌反应4小时。反应完毕后,浓缩,高压制备液相纯化,得9.4mg化合物3。
LC-MS[M+H] +:545。
实施例4化合物4的制备
Figure PCTCN2022075486-appb-000009
步骤1:化合物4-3的合成
向50ml单口瓶中,加入化合物4-1(2.00g,1eq),DMF(25mL),Cs 2CO 3(4.86g,1.5eq),KI(0.20g,0.12eq),化合物4-2(2.94g,1.5eq),升温至75℃搅拌反应过夜。反应完毕后,乙酸乙酯稀释,水洗,饱和食盐水洗,有机相干燥,浓缩,柱层析纯化(PE:EA=100:0-80:20),得0.50g化合物4-3。
1H NMR(500MHz,DMSO-d6)δ10.22(s,1H),7.78(d,J=8.2Hz,1H),7.71-7.64(m,2H),7.45(t,J=72.9Hz,1H).
步骤2:化合物4-4的合成
于50ml单口瓶中,将化合物1-7(0.200g,1eq),化合物4-3(0.390g,1.2eq),TFA(0.266g,3eq)溶于甲苯(10ml)中,油浴锅中加热升温至120℃搅拌过夜。将反应液浓缩,残余物溶于二氯甲烷中,饱和碳酸氢钠水溶液洗二次,水相用二氯甲烷萃取两次,合并有机相,干燥,浓缩,残余物柱层析纯化,得到0.230g化合物4-4。
LC-MS[M+H] +:490。
步骤3:化合物4-5的合成
于50ml单口瓶中,将化合物4-4(0.230g,1eq),DHP(0.059g,1.5eq),PTSA(0.004g,0.05eq)溶于DCM(5ml)中,室温搅拌过夜。将反应液用饱和碳酸 氢钠水溶液洗两次,无水硫酸钠干燥,过滤浓缩,残余物柱层析纯化,得到0.190g化合物4-5。
步骤4:化合物4-6的合成
向50ml单口瓶中,加入化合物4-5(190mg,1eq),化合物3-9(139mg,2.5eq),BrettPhos Pd G3(60mg,0.2eq),t-BuONa(191mg,6eq),1,4-二氧六环(5mL),氮气保护,升温至70℃搅拌反应2小时。反应完毕后,加入乙酸乙酯和水,搅拌分层,水相用乙酸乙酯萃取一次,合并有机相,水洗,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析纯化(DCM:MeOH=100:0-90:10),得100mg化合物4-6。
LC-MS[M+H] +:626。
步骤5:化合物4的合成
向50mL单口瓶中加入化合物4-6(100mg,1eq),DCM(5mL),TFA(1mL),室温下搅拌反应2小时。反应完毕后,浓缩反应液,残余物溶于DCM中,饱和碳酸氢钠洗两次,干燥,浓缩,残余物拌硅胶柱层析纯化(DCM:MeOH=100:0-90:10),得48mg化合物4。
LC-MS[M+H] +:542。
实施例5化合物5的制备
Figure PCTCN2022075486-appb-000010
按照类似实施例4的合成方法,将原料4-3替换成5-1即可。
实施例7化合物7的制备
Figure PCTCN2022075486-appb-000011
步骤1:化合物7-2的合成
向250ml单口瓶中,加入化合物7-1(2.00g,1eq),CH 3CN(50mL),K 2CO 3(4.45g,3eq),化合物1-14(2.42g,1.2eq)。70℃搅拌反应过夜,反应完毕后,浓缩反应液,残余物加EA/H 2O分层,有机相水洗两次,无水硫酸钠干燥,过滤,浓缩,得2.48g化合物7-2。
LC-MS[M+H] +:247。
步骤2:化合物7-3的合成
向250ml单口瓶中,加入化合物7-2(2.48g,1eq),EA(30mL),4M HCl/dioxane(30mL)。室温搅拌反应过夜,反应完毕后,过滤,干燥滤饼,得1.91g化合物7-3(盐酸盐)。
LC-MS[M+H] +:147。
步骤3:化合物7的合成
向50ml单口瓶中,加入化合物3-6(100mg,1eq),化合物7-3(92mg,2.5eq),BrettPhos Pd G3(37mg,0.2eq),t-BuONa(117mg,6eq),1,4-二氧六环(10mL),氮气保护,升温至70℃搅拌反应2小时。反应完毕后,加入乙酸乙酯和水,搅拌分层,水相用乙酸乙酯萃取一次,合并有机相,水洗,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析纯化(DCM:MeOH=100:0-90:10),得29mg化合物7。
LC-MS[M+H] +:557。
按照上述类似的方法,使用相应的原料,可以合成实施例1-2、实施例6、
实施例8-实施例11:
Figure PCTCN2022075486-appb-000012
Figure PCTCN2022075486-appb-000013
实施例12:化合物12的制备
Figure PCTCN2022075486-appb-000014
Figure PCTCN2022075486-appb-000015
步骤1:化合物1-9的合成
于50ml单口瓶中,将化合物1-7(0.600g,1eq),化合物1-8(0.886g,2eq),TFA(0.5mL)溶于甲苯(10ml)中,油浴锅中加热升温至120℃搅拌过夜。将反应液浓缩,残余物溶于二氯甲烷中,饱和碳酸氢钠水溶液洗二次,水相用二氯甲烷萃取两次,合并有机相,干燥,浓缩,残余物柱层析纯化,得到0.580g化合物1-9。
LC-MS[M+H] +:492/494。
步骤2:化合物1-10的合成
于50ml单口瓶中,将化合物1-9(0.200g,1eq),DHP(0.051g,1.5eq),PTSA(0.0035g,0.05eq)溶于DCM(5ml)中,室温搅拌过夜。将反应液用饱和碳酸氢钠水溶液洗两次,无水硫酸钠干燥,过滤浓缩,残余物柱层析纯化,得到0.170g化合物1-10。
LC-MS[M+H] +:576/578。
步骤3:化合物1-12的合成
于50ml单口瓶中,将化合物1-10(0.125g,1eq),化合物1-11(0.0559g,1.5eq),Pd 2(dba) 3(0.0397g,0.2eq),BINAP(0.027g,0.2eq),t-BuONa(0.062g,3eq)溶于甲苯(5ml)中,氮气保护下加热至100℃搅拌过夜。将反应液浓缩,残余物柱层析纯化(PE:EA=100:0-50:50),得到0.140g化合物1-12。
LC-MS[M+H] +:668。
步骤4:化合物1-13的合成
向50mL单口瓶中加入化合物1-12(0.140g,1eq),DCM(5mL),TFA(5mL),室温下搅拌反应5小时。反应完毕后,浓缩反应液,得到0.151g化合物1-13。
LC-MS[M+H] +:484。
步骤5:化合物12的合成
于50ml单口瓶中,将化合物1-13(0.100g,1eq),化合物1-14(0.033g,1.2eq)和DIPEA(0.057g,3eq)溶于DMF(5ml)中,室温搅拌过夜。反应完全,将反应液直接注入C18反相柱中(水:乙腈=95:5-0:100)得到20.6mg化合物12。
LC-MS[M+H] +:544。
实施例13:化合物13的制备
Figure PCTCN2022075486-appb-000016
Figure PCTCN2022075486-appb-000017
按照类似实施例12的合成方法,将原料1-11替换成2-11即可。
按照实施例12和实施例13类似的方法,使用相应的原料,可以合成实施例实施例化合物14-16:
Figure PCTCN2022075486-appb-000018
Figure PCTCN2022075486-appb-000019
药理实验
对照例1
Figure PCTCN2022075486-appb-000020
上述化合物按照WO2018077630A1实施例8制备得到。
实施例A:细胞增殖实验
在乳腺癌细胞系MCF7上使用celltiter-glo发光法活力检测试剂盒(Promega,Cat#G7573)检测化合物细胞增殖抑制活性。取对数生长期细胞,胰蛋白酶消化后,以2000/孔的细胞密度接种在96孔细胞培养板中,在37℃、5%CO 2的潮湿培养箱中孵育过夜。次日,在DMSO中配置化合物并连续稀释,以如下最终浓度加入到96孔板的细胞中:1000、333、111、37、12、4.1、1.4、0.45、0.15nM。将相同体积的DMSO作为溶剂对照加入对照孔中。细胞与化合物共孵育5天,在室温下将50μl的celltiter-glo工作液加入各孔中,避光振荡10分钟。使用PerkinElmer EnVision读取化学发光信号。使用Graph Pad Prism 8.0 软件的非线性回归方程Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))拟合细胞生长抑制的半数抑制浓度IC 50,部分实施例具体结果详见表1。
表1
Figure PCTCN2022075486-appb-000021
实施例B:靶点降解实验
用In-Cell Western方法检测化合物对MCF7和T47D细胞内ER蛋白的降解活性。取对数生长期细胞,胰蛋白酶消化并收集细胞,将其接种于384孔板,37℃、5%CO 2条件下孵育过夜。用DMSO连续稀释化合物,并向孔板中加入120nL/孔化合物溶液(DMSO终浓度为0.5%),使化合物最终浓度依次为100、33、11、3.7、1.2、0.41、0.14、0.046、0.015、0.001nM,并孵育过夜。固定细胞,PBS洗涤后进行透化、封闭。使用ER兔一抗和GAPDH鼠一抗在室温孵育,PBST洗涤3次后,用羊抗兔(800CW)和羊抗鼠(680RD)的荧光二抗于室温避光孵育,并用PBST洗涤3次,1000rpm,1min离心弃上清后,用Odyssey Clx读数,使用非线性回归方程Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))进行拟合求得IC 50,部分实施例具体结果详见表2。
表2
Figure PCTCN2022075486-appb-000022
实施例C:大鼠药代动力学试验
检测方法:雄性SD大鼠6只(150-300g)。随机分为2组,每组3只大鼠。每组的6只大鼠中,3只单次静脉注射1mg/kg的实施例化合物,另外3只单次灌胃给药10mg/kg的实施例化合物,分别在5min(仅对静脉给药)、15min、30min、 1h、2h、4h、7h、24h,通过眼眶静脉丛采血,离心并分离血浆,放入-80℃冰箱保存备用。
将上述血浆样品,通过乙腈沉淀蛋白后,取上清液,并与水1:1混合,取10μL至LC-MS/MS检测,化合物在SD大鼠的血浆中标准曲线的定量限为1.00-1000ng/mL。试验数据结果如表3所示:
表3
Figure PCTCN2022075486-appb-000023
实施例D:小鼠药代动力学试验
使用从北京维通利华实验动物技术有限公司购买得到的雌性BALB/c小鼠(20-30g)进行小鼠PK研究(PO 5mg/kg)。对于5mg/kg的口服(PO)剂量,化合物浓度为0.5mg/mL,现用现配。口服给药以10mL/kg通过口服灌胃进行。通过小鼠眼眶静脉丛采血,每个时间点将100μL的全血至K 2-EDTA管中。采血时间为30min、1h、2h、4h、7h和24h。将采集的全血样品,离心并分离血浆,放入-80℃冰箱保存备用。
将上述血浆样品,通过乙腈沉淀蛋白后,取上清液,并与水1:1混合,取10μL至LC-MS/MS检测。化合物在BALB/c小鼠的血浆中标准曲线的定量限为1.00-1000ng/mL。试验数据结果如表4所示:
表4
Figure PCTCN2022075486-appb-000024
虽然本发明已通过其实施方式进行了全面的描述,但是值得注意的是,各种变化和修改对于本领域技术人员都是显而易见的。这样的变化和修改都应该包括在本发明所附权利要求的范围内。

Claims (24)

  1. 一种如式(I)所示化合物、其立体异构体、互变异构体或药学上可接受的盐:
    Figure PCTCN2022075486-appb-100001
    其中,
    环A选自
    Figure PCTCN2022075486-appb-100002
    D是CR 9或N;
    E是CR 10或N;
    R 1选自H、F、Cl、Br、I、-CN、-CH 3、-CH 2CH 3、-CH(CH 3) 2、-CH 2CH(CH 3) 2、-CH 2OH、-CH 2OCH 3、CH 2CH 2OH、-C(CH 3) 2OH、-CH(OH)CH(CH 3) 2、-C(CH 3) 2CH 2OH、-CH 2CH 2SO 2CH 3、-CH 2OP(O)(OH) 2、-CH 2F、-CHF 2、-CH 2NH 2、-CH 2NHSO 2CH 3、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CF 3、-CH 2CF 3、-CH 2CHF 2、-CH(CH 3)CN、-C(CH 3) 2CN、-CH 2CN、-CO 2H、-COCH 3、-CO 2CH 3、-CO 2C(CH 3) 3、-COCH(OH)CH 3、-CONH 2、-CONHCH 3、-CONHCH 2CH 3、-CONHCH(CH 3) 2、-CON(CH 3) 2、-C(CH 3) 2CONH 2、-NH 2、-NHCH 3、-N(CH 3) 2、-NHCOCH 3、-N(CH 3)COCH 3、-NHS(O) 2CH 3、-N(CH 3)C(CH 3) 2CONH 2、-N(CH 3)CH 2CH 2S(O) 2CH 3、-NO 2、=O、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2OCH 3、-OCH 2CH 2OH、-OCH 2CH 2N(CH 3) 2、-OP(O)(OH) 2、-S(O) 2N(CH 3) 2、-SCH 3、-S(O) 2CH 3、-S(O) 2OH、环丙基、环丙基酰胺基、环丁基、氧杂环丁烷基、氮杂环丁烷基、(1-甲基氮杂环丁烷-3-基)氧基、N-甲基-N-氧杂环丁烷-3-基氨基、氮杂环丁烧-1-基甲基、苄基氧基苯基、吡咯烷-1-基、吡咯烷-1-基-甲酮基、哌嗪-1- 基、吗啉代甲基、吗啉代-甲酮基和吗啉代;
    R 2选自H、C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、-(C 1-C 6亚烷基)-(C 3-C 9环烷基)、-(C 1-C 6亚烷基)-(C 3-C 9杂环基)、C(O)R b、C(O)N(R a) 2、SO 2R a和SO 2N(R a) 2,所述C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、-(C 1-C 6亚烷基)-(C 3-C 9环烷基)、-(C 1-C 6亚烷基)-(C 3-C 9杂环基)任选取代有一个或多个F、Cl、Br、I、CN、OR a、N(R a) 2、C 1-C 9烷基、C 3-C 9环烷基、C 3-C 9杂环基、C 6-C 10芳基、C 5-C 10杂芳基、C(O)R b、C(O)N(R a) 2、SO 2R a和SO 2N(R a) 2
    R 5各自相同或不同,其各自独立地选自H、F、Cl、Br、I、烷基、卤代烷基、烷氧基、卤代烷氧基、-CN、-NH 2、-NO 2、-COOH、-CHO、-OH、羟烷基、环烷基、杂环基、芳基和杂芳基;其中所述烷基、环烷基、杂环基、芳基和杂芳基任选各自独立地被选自烷基、F、Cl、Br、I、-CN、-NH 2、-NO 2、-OH、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 7是H、F、Cl、Br、I、OH或NH 2
    R 8是F或CN;
    R 9是H、Cl、F、卤代的C 1-C 6烷氧基;
    R 10是H、Cl、F、卤代的C 1-C 6烷氧基;
    R a选自H、C 1-C 6烷基、C 2-C 8烯基、炔丙基、C 3-C 6环烷基和C 3-C 6杂环基,所述C 1-C 6烷基、C 2-C 8烯基、炔丙基、C 3-C 6环烷基和C 3-C 6杂环基任选取代有一个或多个独立选自以下的基团:F、Cl、Br、I、CN、OH、OCH 3和SO 2CH 3
    R b选自H、-O(C 1-C 3烷基)、C 1-C 6烷基、C 2-C 8烯基、炔丙基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)、C 3-C 6环烷基和C 3-C 6杂环基,所述-O(C 1-C 3烷基)、C 1-C 6烷基、C 2-C 8烯基、炔丙基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)、C 3-C 6环烷基和C 3-C 6杂环基任选取代有一个或多个独立选自以下的基团:F、Cl、Br、I、CN、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CHF 2、-CH 2CH 2F、-OH、-OCH 3和-SO 2CH 3
    m为0、1、2、3或4;
    p为1、2或3。
  2. 根据权利要求1所述的化合物,其特征在于,式(Ⅰ)所示化合物进一步地为式(Ⅰb)所示化合物:
    Figure PCTCN2022075486-appb-100003
    其中,
    D是CR 9或N;
    E是CR 10或N;
    R 7是H、F、Cl、Br、I、OH或NH 2
    R 8是F或CN;
    R 9是H、Cl、F、或卤代的C 1-C 6烷氧基;
    R 10是H、Cl、F、或卤代的C 1-C 6烷氧基;
    当R 8是F时,R 9、R 10不同时为H或F;
    当D是N、E是CR 10且R 8是F时,R 10不为H或F;或者当E是N、D是CR 9且R 8是F时,R 9不为H或F;
    R 11是Me、F或CH 2F;
    R 12是Me、F、CH 2F、CHF 2、CF 3、CH 2OMe或CH 2OH;
    R 13是H或F;或者
    R 12和R 13连同它们附接至其上的碳原子一起形成环丙基环、环丁基环或氧杂环丁烷环;
    p为1、2或3。
  3. 根据权利要求2所述的化合物,其特征在于,式(Ⅰ)所示化合物进一步地为式(Ⅰc)或式(Ⅰd)所示化合物:
    Figure PCTCN2022075486-appb-100004
  4. 根据权利要求1-3任一项所述的化合物,其特征在于,D是CR 9,E是CR 10
  5. 根据权利要求1-4任一项所述的化合物,其特征在于,D是CR 9,E是N;或者D是N,E是CR 10
  6. 根据权利要求1-5任一项所述的化合物,其特征在于,R 9是Cl;或R 10是Cl。
  7. 根据权利要求1-5任一项所述的化合物,其特征在于,R 9是H;或R 10是H。
  8. 根据权利要求1-5任一项所述的化合物,其特征在于,R 9是F;或R 10是F。
  9. 根据权利要求1-5任一项所述的化合物,其特征在于,R 9是-OCHF 2或-OCHF 3;或R 10是-OCHF 2或-OCHF 3
  10. 根据权利要求1-5任一项所述的化合物,其特征在于,R 9是Cl,且R 10是Cl。
  11. 根据权利要求2-5任一项所述的化合物,其特征在于,R 11是F。
  12. 根据权利要求2-5任一项所述的化合物,其特征在于,R 12是F或CH 2OH。
  13. 根据权利要求2-5任一项所述的化合物,其特征在于,R 13是F。
  14. 根据权利要求1-5任一项所述的化合物,其特征在于,R 8是F。
  15. 根据权利要求1-5任一项所述的化合物,其特征在于,R 7是H或OH。
  16. 根据权利要求1-5任一项所述的化合物,其特征在于,R 7是H。
  17. 根据权利要求1-5任一项所述的化合物,其特征在于,p为1。
  18. 根据权利要求1-5任一项所述的化合物,其特征在于,p为2。
  19. 根据权利要求1-5任一项所述的化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物选自:
    5-(二氟甲氧基)-N-(1-(3-氟丙基)氮杂环丁烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
    N-(3-(二氟甲氧基)-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
    1-(3-氟丙基)-N-(4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)-3-(三氟甲氧基)苯基)氮杂环丁烷-3-胺;
    5-(二氟甲氧基)-N-((S)-1-(3-氟丙基)吡咯烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
    5-(二氟甲氧基)-N-((R)-1-(3-氟丙基)吡咯烷-3-基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-胺;
    (3R,4R)-4-((5-(二氟甲氧基)-6-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)吡啶-3-基)氨基)-1-(3-氟丙基)吡咯烷-3-醇;
    N-(3-(二氟甲氧基)-5-氟-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
    3-((6S,8R)-6-(2-(二氟甲氧基)-6-氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
    3-((6S,8R)-6-(3-(二氟甲氧基)-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
    3-((6S,8R)-6-(2-(二氟甲氧基)-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇;
    N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺;
    (S)-N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)吡咯烷-3-胺;
    (3R,4R)-4-((3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)氨基)-1-(3-氟丙基)吡咯烷-3-醇;
    (R)-N-(3,5-二氯-4-((6S,8R)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉-6-基)苯基)-1-(3-氟丙基)吡咯烷-3-胺;或
    3-((6S,8R)-6-(2,6-二氯-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-8-甲基-3,6,8,9-四氢-7H-吡唑并[4,3-f]异喹啉-7-基)-2,2-二氟丙-1-醇。
  20. 一种药物组合物,其特征在于,所述药物组合物包含治疗有效量的至少一种权利要求1-19任一项所述的化合物或其药学上可接受的盐和至少一种药学上可接受的辅料。
  21. 权利要求1-19任一项所述的化合物或其药物上可接受的盐或权利要求20所述的药物组合物在制备用于治疗雌激素受体相关疾病的药物中的用途。
  22. 根据权利要去21所述的用途,其特征在于,所述雌激素受体相关疾病为选自以下的癌症:乳腺癌、肺癌、卵巢癌、子宫内膜癌、前列腺癌和子宫癌。
  23. 一种在患者中治疗雌激素受体相关疾病的方法,其特征在于,包括向所述患者给予治疗有效量的上述权利要求1-19任一项所述的化合物或其药物上可接受的盐或权利要求20所述的药物组合物。
  24. 根据权利要求23所述的方法,其特征在于,所述雌激素受体相关疾病为选自以下的癌症:乳腺癌、肺癌、卵巢癌、子宫内膜癌、前列腺癌和子宫癌。
PCT/CN2022/075486 2021-02-08 2022-02-08 杂芳基并哌啶类衍生物及其药物组合物和应用 WO2022166980A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280009011.9A CN116669729A (zh) 2021-02-08 2022-02-08 杂芳基并哌啶类衍生物及其药物组合物和应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021076009 2021-02-08
CNPCT/CN2021/076009 2021-02-08

Publications (1)

Publication Number Publication Date
WO2022166980A1 true WO2022166980A1 (zh) 2022-08-11

Family

ID=82742002

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/075486 WO2022166980A1 (zh) 2021-02-08 2022-02-08 杂芳基并哌啶类衍生物及其药物组合物和应用

Country Status (2)

Country Link
CN (1) CN116669729A (zh)
WO (1) WO2022166980A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170305909A1 (en) * 2016-04-20 2017-10-26 Astrazeneca Ab Chemical compounds
US20170320871A1 (en) * 2016-04-08 2017-11-09 Genentech, Inc. Tetrahydroisoquinoline estrogen receptor modulators and uses thereof
WO2019002441A1 (en) * 2017-06-29 2019-01-03 Astrazeneca Ab CHEMICAL COMPOUNDS
WO2019002442A1 (en) * 2017-06-29 2019-01-03 Astrazeneca Ab CHEMICAL COMPOUNDS
CN109843888A (zh) * 2016-10-24 2019-06-04 阿斯利康(瑞典)有限公司 可用于治疗癌症的6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉衍生物
WO2019228443A1 (en) * 2018-05-30 2019-12-05 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Selective estrogen receptor downregulators and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170320871A1 (en) * 2016-04-08 2017-11-09 Genentech, Inc. Tetrahydroisoquinoline estrogen receptor modulators and uses thereof
US20170305909A1 (en) * 2016-04-20 2017-10-26 Astrazeneca Ab Chemical compounds
CN109843888A (zh) * 2016-10-24 2019-06-04 阿斯利康(瑞典)有限公司 可用于治疗癌症的6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉衍生物
WO2019002441A1 (en) * 2017-06-29 2019-01-03 Astrazeneca Ab CHEMICAL COMPOUNDS
WO2019002442A1 (en) * 2017-06-29 2019-01-03 Astrazeneca Ab CHEMICAL COMPOUNDS
WO2019228443A1 (en) * 2018-05-30 2019-12-05 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Selective estrogen receptor downregulators and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SCOTT JAMES S., MOSS THOMAS A., BALAZS AMBER, BARLAAM BERNARD, BREED JASON, CARBAJO RODRIGO J., CHIARPARIN ELISABETTA, DAVEY PAUL : "Discovery of AZD9833, a Potent and Orally Bioavailable Selective Estrogen Receptor Degrader and Antagonist", JOURNAL OF MEDICINAL CHEMISTRY, vol. 63, no. 23, 10 December 2020 (2020-12-10), US , pages 14530 - 14559, XP055823030, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.0c01163 *
SCOTT JAMES S., MOSS THOMAS A., BARLAAM BERNARD, DAVEY PAUL R. J., FAIRLEY GARY, GANGL ERIC T., GREENWOOD RYAN D. R., HATOUM-MOKDA: "Addition of Fluorine and a Late-Stage Functionalization (LSF) of the Oral SERD AZD9833", ACS MEDICINAL CHEMISTRY LETTERS, vol. 11, no. 12, 10 December 2020 (2020-12-10), US , pages 2519 - 2525, XP055957319, ISSN: 1948-5875, DOI: 10.1021/acsmedchemlett.0c00505 *

Also Published As

Publication number Publication date
CN116669729A (zh) 2023-08-29

Similar Documents

Publication Publication Date Title
US20230062486A1 (en) Kras g12c inhibitor and pharmaceutical use thereof
CN111499634B (zh) 一种喹唑啉化合物及其在医药上的应用
KR102057877B1 (ko) 질소함유 헤테로고리 유도체 및 그의 약물에서의 용도
CN112390796B (zh) Kras g12c抑制剂及其在医药上的应用
JP2021512059A (ja) ブルトン型チロシンキナーゼ(btk)阻害剤のe3リガーゼリガンドとのコンジュゲーションによるbtkの分解および使用方法
WO2020216190A1 (zh) 一种喹唑啉化合物及其在医药上的应用
TWI762769B (zh) 肽醯精胺酸脫亞胺酶抑制劑及其用途
US20230203055A1 (en) Fused ring compound and application thereof in medicine
AU2017237929A1 (en) Pyrimidines and variants thereof, and uses therefor
CN116354936A (zh) 用作选择性aurora a抑制剂的新型杂环化合物
WO2022166983A1 (zh) 杂芳基并哌啶类衍生物及其药物组合物和应用
CN110746399A (zh) 具有降解雄激素受体活性的化合物
JP2023553291A (ja) Trpm3媒介性障害を治療するためのアリール誘導体
CN107074816A (zh) 一种杂环衍生物及其制备方法和在医药上的用途
WO2022166980A1 (zh) 杂芳基并哌啶类衍生物及其药物组合物和应用
WO2020029908A1 (zh) 螺桥环化合物、其药物组合物及其用途
WO2022161166A1 (zh) 靶向嵌合化合物、含其的药物组合物及其制备方法和用途
CN112759538B (zh) 3-(二甲氨基甲基)环己-4-醇衍生物及其制备方法和药物用途
WO2022002100A1 (zh) 新型苯并咪唑化合物
CN108864080B (zh) 作为选择性雌激素受体下调剂的四环类化合物及其应用
CA3136725A1 (en) Inhibitors of cd40-cd154 binding
WO2021073498A1 (zh) Egfr抑制剂、组合物及其制备方法
WO2020156162A1 (zh) Mek抑制剂及其在医药上的应用
WO2022012534A1 (zh) 含氮杂环化合物、药物组合物和应用
WO2023208165A1 (zh) 一种含氮杂环衍生物及其组合物和药学上的应用

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: 22749259

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280009011.9

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22749259

Country of ref document: EP

Kind code of ref document: A1