CN111333587A - Substituted pyrimidine-2, 4(1H,3H) -dione derivatives and uses thereof - Google Patents

Substituted pyrimidine-2, 4(1H,3H) -dione derivatives and uses thereof Download PDF

Info

Publication number
CN111333587A
CN111333587A CN202010281944.6A CN202010281944A CN111333587A CN 111333587 A CN111333587 A CN 111333587A CN 202010281944 A CN202010281944 A CN 202010281944A CN 111333587 A CN111333587 A CN 111333587A
Authority
CN
China
Prior art keywords
alkyl
radical
chf
compound
alkylene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202010281944.6A
Other languages
Chinese (zh)
Other versions
CN111333587B (en
Inventor
金传飞
钟文和
许腾飞
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dongguan Dongyangguang New Drug R & D Co ltd
Guangdong HEC Pharmaceutical
Original Assignee
Dongguan Dongyangguang New Drug R & D Co ltd
Sunshine Lake Pharma Co Ltd
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 Dongguan Dongyangguang New Drug R & D Co ltd, Sunshine Lake Pharma Co Ltd filed Critical Dongguan Dongyangguang New Drug R & D Co ltd
Priority to CN202010281944.6A priority Critical patent/CN111333587B/en
Publication of CN111333587A publication Critical patent/CN111333587A/en
Application granted granted Critical
Publication of CN111333587B publication Critical patent/CN111333587B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/52Two oxygen atoms
    • C07D239/54Two oxygen atoms as doubly bound oxygen atoms or as unsubstituted hydroxy radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

The invention belongs to the technical field of medicaments, and relates to substituted pyrimidine-2, 4(1H,3H) -diketone derivatives, application thereof and a pharmaceutical composition containing the compounds, which can be used as gonadotropin releasing hormone receptor antagonists. The invention also relates to methods for preparing such compounds and pharmaceutical compositions, and their use in the prevention or treatment of sex hormone dependent diseases, including but not limited to prostate cancer, endometriosis, uterine fibroids, precocious puberty, and the like.

Description

Substituted pyrimidine-2, 4(1H,3H) -dione derivatives and uses thereof
Technical Field
The invention belongs to the technical field of medicines, and relates to a compound and a pharmaceutical composition for preventing or treating sex hormone-dependent diseases, and a using method and application thereof. In particular, the present invention relates to substituted pyrimidine-2, 4(1H,3H) -dione derivatives which are useful as gonadotropin-releasing hormone receptor antagonists, and uses thereof.
Background
The secretion of anterior pituitary hormone is feedback-controlled by peripheral hormones secreted from the target organs of the respective hormones and by secretion-regulating hormones from the hypothalamus, which is the upper central organ of the anterior pituitary (hereinafter, these hormones are collectively referred to as "hypothalamic hormones" in the specification). Currently, it has been confirmed that as hypothalamic hormones, there are nine hormones including, for example, thyrotropin-releasing hormone (TRH) and gonadotropin-releasing hormone (GnRH, sometimes referred to as LH-RH (luteinizing hormone-releasing hormone)). These hypothalamic hormones are thought to exhibit their effects via receptors that are thought to be present in the anterior pituitary; and efforts have been made to find receptor-gene expression specific for these hormones, including in humans. Thus, antagonists or agonists that specifically and selectively act on these receptors should control the action of hypothalamic hormones and the secretion of anterior pituitary hormones. Therefore, such antagonists or agonists are expected to be useful for the prevention or treatment of anterior pituitary hormone-dependent diseases.
The compounds having GnRH antagonistic activity known so far are mostly peptide compounds, for example, GnRH-derived linear peptides (linear peptides) (US 5,140,009 and US 5,171,835), bicyclic peptide derivatives (Journal of medicinal Chemistry, Vol.36, pp.3265-3273(1993)), decapeptide compounds modified at position 5 or 6 (WO9846634A1) and decapeptide compounds modified at position 8 (EP 0277829B1), and the like.
Peptide compounds present a number of problems to be solved, which are associated with oral absorbability, dosage form, dose volume, drug stability, sustained action, metabolic stability, and the like. Therefore, there is a strong need for an oral GnRH antagonist, particularly an antagonist based on a non-peptide compound, which has an excellent therapeutic effect on sex hormone-dependent diseases such as prostate cancer, endometriosis, precocious puberty and the like, which exhibits not only an instant pituitary-gonadotropic action (acute action) but also excellent oral absorbability.
Disclosure of Invention
The following is a summary of some aspects of the invention only and is not intended to be limiting. These aspects and others are described more fully below. All references in this specification are incorporated herein by reference in their entirety. When the disclosure of the present specification differs from the cited documents, the disclosure of the present specification controls.
The present invention relates to a novel class of substituted pyrimidine-2, 4(1H,3H) -dione derivatives having unexpectedly superior GnRH antagonistic activity, which can be used as gonadotropin-releasing hormone receptor antagonists, and thus can be used for the prevention or treatment of sex hormone-dependent diseases, including, but not limited to, prostate cancer, endometriosis, uterine fibroids, precocious puberty, and the like.
The compound has stable property and good safety, and has pharmacodynamic and pharmacokinetic advantages, such as good brain/plasma ratio (brainnplama ratio), good bioavailability or good metabolic stability, and the like, so the compound has good clinical application prospect.
The invention also provides processes for the preparation of such compounds and pharmaceutical compositions containing them.
In one aspect, the invention relates to a compound that is a compound of formula (I) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt of a compound of formula (I), or prodrug thereof,
Figure BDA0002446925260000021
wherein R is1a、R1b、R1c、R1d、R1e、R2a、R2b、R3a、R3b、R3c、R3d、R3e、R4、R5a、R5b、R5c、R5dAnd R has the meaning as described in the present invention.
In some embodiments, each R is1a、R1b、R1c、R1dAnd R1eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group.
In some embodiments, each R is2aAnd R2bIndependently H, D, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl or C1-C6A haloalkyl group.
In some embodiments, each R is3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy radicalBase), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms.
In some embodiments, R4Is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group.
In some embodiments, each R is5a、R5b、R5cAnd R5dIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-C6CycloalkanesRadical, heterocyclic radical consisting of 3-8 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms.
In some embodiments, R is-C1-C6alkylene-C (═ O) NH2、-C1-C6alkylene-C (═ O) NHCH3、-C1-C6alkylene-C (═ O) N (CH)3)2、-C1-C6alkylene-C (═ O) - (C)1-C6Alkyl), -C1-C6alkylene-C (═ O) - (C)1-C6Alkoxy), -C1-C6alkylene-S (═ O)2NH2、-C1-C6alkylene-S (═ O)2NHCH3、-C1-C6alkylene-S (═ O)2N(CH3)2、-C1-C6alkylene-S (═ O)2-(C1-C6Alkyl) or
Figure BDA0002446925260000022
Wherein X, y and z have the meaning as indicated in the present invention.
In some embodiments, X is-O-, -S-, -C (═ O) -, -S (═ O)2-、-CH2-or-NRn-; wherein R isnHave the meaning as described in the present invention.
In some embodiments, RnIs H, D, C1-6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-10Aryl, 5-10-membered heteroaryl, -C (═ O) - (C)1-C6Alkyl), -C (═ O) - (C)3-C6Cycloalkyl), -C (═ O) - (3-8 atom heterocyclyl), -S (═ O)2-(C1-C6Alkyl), -S (═ O)2-(C3-C6Cycloalkyl) or-S (═ O)2- (heterocyclic groups of 3 to 8 atoms).
In some embodiments, y is 0, 1,2, or 3.
In some embodiments, z is 1,2, or 3.
In some embodiments, R is-C1-C4alkylene-C (═ O) NH2、-C1-C4alkylene-C (═ O) NHCH3、-C1-C4alkylene-C (═ O) N (CH)3)2、-C1-C4alkylene-C (═ O) - (C)1-C4Alkyl), -C1-C4alkylene-C (═ O) - (C)1-C4Alkoxy), -C1-C4alkylene-S (═ O)2NH2、-C1-C4alkylene-S (═ O)2NHCH3、-C1-C4alkylene-S (═ O)2N(CH3)2、-C1-C4alkylene-S (═ O)2-(C1-C4Alkyl) or
Figure BDA0002446925260000031
Wherein X, y and z have the meaning as indicated in the present invention.
In some embodiments, X is-O-, -S-, or-NRn-; wherein Rn has the meaning as described in the present invention.
In some embodiments, RnIs H, D, C1-4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, -C (═ O) - (C)1-C4Alkyl), -C (═ O) - (C)3-C6Cycloalkyl), -C (═ O) - (3-6 atom heterocyclyl), -S (═ O)2-(C1-C4Alkyl), -S (═ O)2-(C3-C6Cycloalkyl) or-S (═ O)2- (heterocyclic groups of 3 to 6 atoms).
In other embodiments, R is
Figure BDA0002446925260000032
Figure BDA0002446925260000033
In some embodiments, each R is2aAnd R2bIndependently H, D, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl or C1-C4A haloalkyl group.
In other embodiments, each R is2aAnd R2bIndependently H, D, methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3or-CH2CF2CHF2
In some embodiments, each R is3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C4Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms.
In other embodiments, each R is3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, indolyl or quinolinyl.
In some embodiments, R4Is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
In other embodiments, R4Is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, or 2-hydroxyethyl.
In some embodiments, each R is1a、R1b、R1c、R1dAnd R1eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
In other embodiments, each R is1a、R1b、R1c、R1dAnd R1eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, or 2-hydroxyethyl.
In some embodiments, each R is5a、R5b、R5cAnd R5dIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino, hydroxy-substituted C1-C4Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms.
In other embodiments, each R is5a、R5b、R5cAnd R5dIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propylAryloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, indolyl or quinolinyl.
In some embodiments, the compound of the present invention, which is a compound of formula (II) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (II), or a prodrug thereof,
Figure BDA0002446925260000051
wherein R is2a、R2b、R4、R5dAnd R has the meaning as described in the present invention.
In some embodiments, the compound of the present invention is a compound having one of the following structures or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt of the compound having one of the following structures, or a prodrug thereof:
Figure BDA0002446925260000052
Figure BDA0002446925260000061
Figure BDA0002446925260000071
in another aspect, the present invention relates to a pharmaceutical composition comprising a compound disclosed herein.
In some embodiments, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, or any combination thereof.
In a further aspect, the present invention relates to the use of a compound or pharmaceutical composition disclosed herein for the preparation of a medicament for the prevention or treatment of a sex hormone dependent disease.
In some embodiments, the sex hormone dependent disorder of the invention is a sex hormone dependent cancer, a bone metastasis of a sex hormone dependent cancer, prostatic hypertrophy, uterine fibroids, endometriosis, uterine fibroids, precocious puberty, amenorrhea, premenstrual syndrome, dysmenorrhea, multiple atrial ovarian syndrome, polycystic ovarian syndrome, acne, alopecia, alzheimer's disease, infertility, irritable bowel syndrome, a benign or malignant tumor that is hormone independent and sensitive to LH-RH (luteinizing hormone releasing hormone), or flushing.
In one aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the manufacture of a medicament for use as a reproduction regulator (reproduction regulator), a contraceptive, an ovulation inducer (ovulatory), or for the prevention of postoperative recurrence of sex hormone dependent cancer.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the manufacture of a medicament for antagonizing gonadotropin releasing hormone (GnRH).
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds of formula (I) or formula (II).
Biological test results show that the compound has stronger antagonistic activity on gonadotropin-releasing hormone (GnRH), so that the compound provided by the invention can be used as a better GnRH receptor antagonist.
Any embodiment of any aspect of the invention may be combined with other embodiments, as long as they do not contradict. Furthermore, in any embodiment of any aspect of the invention, any feature may be applicable to that feature in other embodiments, so long as they do not contradict.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. One skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
The following definitions, as used herein, should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be found in the descriptions of "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and JerryMarch, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to one or to more than one (i.e., to at least one) of the objects. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "stereoisomers" refers to compounds having the same chemical structure, but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers)), geometric (cis/trans) isomers, atropisomers, and the like.
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (lowenergy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization.
"pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and which are effective for their intended use.
The term "optionally substituted with … …", is used interchangeably with the term "unsubstituted or substituted with ….," i.e., the structure is unsubstituted or substituted with one or more substituents described herein, including but not limited to D, F, Cl, Br, I, N3,-CN,-NO2,-NH2,-OH,-SH,-COOH,-C(=O)NH2,-C(=O)NHCH3,-C(=O)N(CH3)2-C (═ O) - (alkyl), -C (═ O) - (alkoxy), alkyl, alkoxy, alkylthio, alkylamino, alkenyl, alkynyl, haloalkyl, haloalkoxy, hydroxy-substituted alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -alkylene-C (═ O) NH2-alkylene-C (═ O) NHCH3-alkylene-C (═ O) N (CH)3)2-alkylene-C (═ O) - (alkyl), -alkylene-C (═ O) - (alkoxy), -alkylene-S (═ O)2NH2-alkylene-S (═ O)2NHCH3-alkylene-S (═ O)2N(CH3)2-alkylene-S (═ O)2- (alkyl), -C (═ O) - (cycloalkyl), -C (═ O) - (heterocyclyl), -S (═ O)2- (alkyl), -S (═ O)2- (cycloalkyl), -S (═ O)2- (heterocyclyl) and the like.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-C6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The terms "halogen" and "halo" are used interchangeably herein to refer to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I).
The term "alkyl" or "alkyl group" as used herein, denotes a saturated straight chain containing from 1 to 20 carbon atomsOr a branched monovalent hydrocarbon group, wherein the alkyl group may be optionally substituted with one or more substituents described herein. In some embodiments, the alkyl group contains 1 to 6 carbon atoms; in other embodiments, the alkyl group contains 1 to 4 carbon atoms; in still other embodiments, the alkyl group contains 1 to 3 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl group (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) Tert-butyl (t-Bu, -C (CH)3)3) And so on.
The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon. Unless otherwise specified, the alkylene group contains 1 to 10 carbon atoms. In some embodiments, the alkylene group contains 1 to 6 carbon atoms; in other embodiments, the alkylene group contains 1 to 4 carbon atoms; in still other embodiments, the alkylene group contains 1 to 2 carbon atoms. Examples include, but are not limited to, methylene (-CH)2-) ethylene (-CH2CH2-) isopropylidene (-CH (CH)3)CH2-) and the like. The alkylene group is optionally substituted with one or more substituents described herein.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "trans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodimentIn one embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) 1-propenyl (i.e., propenyl, -CH ═ CH-CH)3) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. a carbon-carbon sp triple bond, wherein said alkynyl radical may optionally be substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH)2C.ident.CH), 1-propynyl (i.e., propynyl, -C.ident.C-CH)3) And so on.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In some embodiments, alkoxy groups contain 1 to 6 carbon atoms; in other embodiments, the alkoxy group contains 1 to 4 carbon atoms; in still other embodiments, alkoxy groups contain 1-3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein. Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-propyloxy, n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (isopropyloxy, i-PrO, i-propoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH)2CH(CH3)2) 2-butoxy (s-BuO, s-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) And so on.
The term "alkylthio" means an alkyl group attached to the rest of the molecule through a sulfur atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkylthio group contains 1 to 12 carbon atoms. In some embodiments, the alkylthio group contains 1 to 6 carbon atoms; in other embodiments, the alkylthio group contains 1 to 4 carbon atoms; in still other embodiments, the alkylthio group contains 1 to 3 carbon atoms. The alkylthio group may be optionally substituted with one or more substituents described herein. Examples of alkylthio groups include, but are not limited to, methylthio, ethylthio, and the like.
The term "alkylamino" or "alkylamino" denotes an amino group independently substituted with one or two alkyl groups, respectively, including "N-alkylamino" and "N, N-dialkylamino" wherein the alkyl groups have the meaning as described herein. Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino (methylamino), N-ethylamino (ethylamino), N-dimethylamino (dimethylamino), N-diethylamino (diethylamino), and the like. The alkylamino group is optionally substituted with one or more substituents described herein.
The term "hydroxy-substituted alkyl" denotes an alkyl group substituted with one or more hydroxy groups, wherein the alkyl group has the meaning as described herein; examples include, but are not limited to, hydroxymethyl, hydroxyethyl (e.g., 2-hydroxyethyl), 2-hydroxy-1-propyl, 3-hydroxy-1-propyl, 2, 3-dihydroxypropyl, and the like.
The term "haloalkyl" or "haloalkoxy" means an alkyl or alkoxy group substituted with one or more halogen atoms, wherein the alkyl and alkoxy groups have the meaning as described herein, examples of which include, but are not limited to, trifluoromethyl, difluoromethyl, trifluoromethoxy, and the like. In some embodiments, C1-C6The haloalkyl group containing a fluorine-substituted C1-C6Alkyl radical(ii) a In other embodiments, C1-C4The haloalkyl group containing a fluorine-substituted C1-C4An alkyl group; in still other embodiments, C1-C2The haloalkyl group containing a fluorine-substituted C1-C2An alkyl group.
The term "cycloalkyl" denotes a saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms. In some embodiments, cycloalkyl groups contain 3 to 10 ring carbon atoms, e.g., C3-C10A cycloalkyl group; in other embodiments, cycloalkyl groups contain 3 to 8 ring carbon atoms, e.g., C3-C8A cycloalkyl group; in still other embodiments, cycloalkyl groups contain 3-6 ring carbon atoms, e.g., C3-C6A cycloalkyl group. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like. Wherein, as described in the present invention, C3-C8Cycloalkyl radicals including C3-C6A cycloalkyl group; said C3-C6Cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The cycloalkyl group may be optionally substituted with one or more substituents described herein.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms; wherein the heterocyclic group is non-aromatic and does not contain any aromatic ring. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atoms of the ring may optionally be oxidized to the N-oxide. The term "heterocyclyl" may be used interchangeably with the term "heterocycle". As described herein, the heterocyclyl group may consist of 3 to 8 atoms or 3 to 6 atoms, optionally selected from C, N, O or S and at least one atom being N, O or S; wherein the heterocyclic group consisting of 3 to 8 atoms includes a heterocyclic group consisting of 3 to 6 atoms; the heterocyclic group consisting of 3 to 6 atoms includes a heterocyclic group consisting of 3 to 5 atoms. Utensil for cleaning buttockIn particular, the heterocyclic group consisting of 3 to 6 atoms includes, but is not limited to, ethylene oxide, aziridinyl, azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, thiazolidinyl, pyrazolidinyl, pyrazolinyl, oxazolidinyl, imidazolidinyl, tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, and the like. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
The term "aryl" denotes a monocyclic, bicyclic and tricyclic carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the terms "aromatic ring" or "aromatic ring". Examples of the aryl group may include phenyl, 2, 3-dihydro-1H-indenyl, naphthyl and anthryl. The aryl group may be optionally substituted with one or more substituents described herein. Unless otherwise stated, the group "C6-C10Aryl "represents an aryl group containing from 6 to 10 ring carbon atoms.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one ring contains 1,2,3 or 4 ring heteroatoms selected from nitrogen, oxygen, sulfur, and wherein the heteroaryl has one or more attachment points to the rest of the molecule. when-CH is present in the heteroaryl group2When it is a group, -CH2-the group may optionally be replaced by-C (═ O) -. Unless otherwise indicated, the heteroaryl group may be attached to the rest of the molecule (e.g., the main structure in the formula) via any reasonable site, which may be C or N. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". The heteroaryl group may be optionally substituted with one or more substituents described herein. In some embodiments, heteroaryl is 5-10 atom heteroaryl, meaning that heteroaryl contains 1-9 ring carbon atoms and 1,2,3, or 4 ring heteroatoms selected from O, S and NAn atom; in other embodiments, heteroaryl is 5-6 atom heteroaryl, meaning that heteroaryl contains 1-5 ring carbon atoms and 1,2,3, or 4 ring heteroatoms selected from O, S and N; examples of heteroaryl groups consisting of 5 to 6 atoms include, but are not limited to, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyrazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, thienyl, thiazolyl, triazolyl, tetrazolyl, and the like.
The term "j-k atoms" means that the cyclic group consists of j-k ring atoms including carbon atoms and/or heteroatoms such as O, N, S, P; j and k are each independently any non-zero natural number, and k > j; the term "j-k" includes j, k and any natural number therebetween. For example, "3-8 atoms", "3-6 atoms", "5-10 atoms", or "5-6 atoms" means that the cyclic group consists of 3-8, 3-6, 5-10, or 5-6 ring atoms, including carbon atoms and/or heteroatoms such as O, N, S, P.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I) or formula (II). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-C24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, descriptive acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-C8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, ethanolamine, or mixtures thereof. The term "hydrate" refers to an association of solvent molecules that is water.
When the solvent is water, the term "hydrate" may be used. In one embodiment, a molecule of a compound of the present invention may be associated with a molecule of water, such as a monohydrate; in another embodiment, one molecule of the compound of the present invention may be associated with more than one molecule of water, such as a dihydrate; in yet another embodiment, one molecule of the compound of the present invention may be associated with less than one molecule of water, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the biological effectiveness of the compound in its non-hydrated form.
The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
The term "prevent" or "prevention" refers to a reduction in the risk of acquiring a disease or disorder (i.e., arresting the development of at least one clinical symptom of a disease in a subject that may be facing or predisposed to facing such a disease, but who has not yet experienced or exhibited symptoms of the disease).
The term "therapeutically effective amount" means an amount of a compound that, when administered to a subject to treat a disease, is sufficient to effect treatment of the disease. The "therapeutically effective amount" may vary with the compound, the disease and the severity, as well as the condition, age, weight, sex, etc., of the subject to be treated.
Unless otherwise indicated, all suitable isotopic variations, stereoisomers, tautomers, solvates, metabolites, pharmaceutically acceptable salts and prodrugs thereof, of the compounds of the present invention are encompassed within the scope of the present invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
Nitroxides of the compounds of the present invention are also included within the scope of the present invention. The nitroxides of the compounds of the present invention may be prepared by oxidation of the corresponding nitrogen-containing basic species using a common oxidizing agent (e.g. hydrogen peroxide) in the presence of an acid such as acetic acid at elevated temperature, or by reaction with a peracid in a suitable solvent, for example peracetic acid in dichloromethane, ethyl acetate or methyl acetate, or 3-chloroperoxybenzoic acid in chloroform or dichloromethane.
The compounds of formula (I) or formula (II) may be present in the form of a salt. In some embodiments, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. In other embodiments, the salt need not be a pharmaceutically acceptable salt, and may be an intermediate useful in the preparation and/or purification of a compound of formula (I) or formula (II) and/or in the isolation of an enantiomer of a compound of formula (I) or formula (II).
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the invention relates to intermediates for the preparation of compounds of formula (I) or formula (II).
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The invention provides a pharmaceutical composition, which comprises a compound shown as a formula (I) or a formula (II) or an individual stereoisomer, a racemic or non-racemic mixture of isomers or a pharmaceutically acceptable salt or solvate thereof. In some embodiments of the invention, the pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier, adjuvant or vehicle, and optionally, other therapeutic and/or prophylactic ingredients.
Suitable carriers, adjuvants and excipients are well known to those skilled in the art and are described in detail, for example, in Ansel h.c.et al, Ansel's Pharmaceutical Dosage Forms and Drug delivery systems (2004) Lippincott, Williams & Wilkins, philidelphia; gennaro a.r.et al, Remington: the Science and Practice of Pharmacy (2000) Lippincott, Williams & Wilkins, Philadelphia; and Rowe R.C., Handbook of Pharmaceutical Excipients (2005) Pharmaceutical Press, Chicago.
It will also be appreciated that certain compounds of the invention may be present in free form or, if appropriate, in the form of a pharmaceutically acceptable derivative thereof, when used in therapy. Some non-limiting embodiments of pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, pharmaceutically acceptable excipients may be selected for their specific function in the composition. Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and provide alternative functions, depending on how many such excipients are present in the formulation and which other excipients are present in the formulation.
The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some commonly used methods in the art are described in Remington's Pharmaceutical Sciences (Mack publishing company).
Thus, in another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising the disclosed compounds may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein are generally formulated in a dosage form suitable for administration to a patient by a desired route. For example, dosage forms include those suitable for the following routes of administration: (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patches; (4) rectal administration, e.g., suppositories; (5) inhalation, such as aerosols, solutions, and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
In some embodiments, the compounds disclosed herein can be formulated in oral dosage forms. In other embodiments, the compounds disclosed herein may be formulated in an inhalation dosage form. In other embodiments, the compounds disclosed herein can be formulated for nasal administration. In still other embodiments, the compounds disclosed herein can be formulated for transdermal administration. In still other embodiments, the compounds disclosed herein may be formulated for topical administration.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, enteric-coated tablets, sugar-coated or film-coated tablets.
The pharmaceutical composition provided by the present invention may be provided in soft or hard capsules, which may be prepared from gelatin, methylcellulose, starch or calcium alginate.
The pharmaceutical compositions provided by the present invention may be administered parenterally by injection, infusion or implantation for local or systemic administration. Parenteral administration as used herein includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein can be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems and solid forms suitable for solution or suspension in a liquid prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical Science (see Remington: The Science and Practice of Pharmacy, supra).
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for administration to a patient by inhalation, such as a dry powder, aerosol, suspension, or solution composition.
Pharmaceutical compositions suitable for transdermal administration may be prepared as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch agent by iontophoresis, as generally described in Pharmaceutical Research,3(6),318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
Use of the Compounds and pharmaceutical compositions of the invention
The compounds and pharmaceutical compositions provided by the present invention have excellent GnRH antagonistic activity and low toxicity (e.g., acute toxicity, chronic toxicity, genetic toxicity, reproductive toxicity, cardiotoxin, drug interactions and carcinogenicity). Also, the compound or the pharmaceutical composition is excellent in oral absorbability, action sustainability, stability and pharmacokinetics. Furthermore, the compounds or pharmaceutical compositions are less affected by plasma components. Therefore, the compound or the pharmaceutical composition of the present invention can be safely used in mammals (e.g., human, monkey, cow, horse, dog, cat, rabbit, rat, mouse, etc.) to prevent and/or treat diseases depending on male or female hormones and diseases caused by excess of these hormones, etc. by controlling plasma sex hormone concentration using GnRH receptor antagonism to inhibit gonadotropin secretion.
Specifically, the compound or the pharmaceutical composition of the present invention is suitable for the prevention and/or treatment of sex hormone-dependent cancer (e.g., prostate cancer, uterine cancer, breast cancer, pituitary tumor, etc.), bone metastasis of sex hormone-dependent cancer, prostatic hypertrophy, uterine myoma, endometriosis, uterine fibroid, precocious puberty, amenorrhea, premenstrual syndrome, dysmenorrhea, multiple atrial ovarian syndrome (multilocular ovarian syndrome), polycystic ovarian syndrome, acne, alopecia, alzheimer's disease (alzheimer's disease, senile dementia of the alzheimer's type and a mixed type thereof), and the like. The compounds of the invention are also useful for regulating male and female reproduction (e.g., pregnancy regulators, menstrual cycle regulators, and the like). The compounds or pharmaceutical compositions of the invention may also be used as male or female contraceptives or as female ovulation inducers. Based on the rebound effect after drug withdrawal, the compound or the pharmaceutical composition of the invention can be used for treating infertility. Furthermore, the compounds or pharmaceutical compositions of the present invention can be used as agents for the prevention and/or treatment of hormone-independent and LH-RH sensitive benign or malignant tumors. Furthermore, the compound or the pharmaceutical composition of the present invention can be used as an agent for preventing and/or treating irritable bowel syndrome and preventing postoperative recurrence of sex hormone-dependent cancer (an agent for preventing postoperative recurrence of prostate cancer; an agent for preventing postoperative recurrence of breast cancer or ovarian cancer before or after menopause; particularly an agent for preventing postoperative recurrence of breast cancer or ovarian cancer before menopause).
Furthermore, the compounds or pharmaceutical compositions of the invention are suitable for regulating the estrus of animals, improving the quality of meat and promoting the growth of animals in animal husbandry. The compound of the present invention is also suitable for a fish spawning promoter (fisherspawningpromoter).
The compounds or pharmaceutical compositions of the invention may also be used to inhibit transient increases in plasma testosterone concentrations (flarepromeon) observed upon administration of GnRH superagonists such as leuprolide acetate. The compound of the present invention can be used in combination with a super-agonist such as leuprolide acetate, gonadorelin (gonadorelin), buserelin (buserelin), triptorelin (triptorelin), goserelin (goserelin), nalfrelin (nafarelin), histrelin (histrelin), deslorelin (deslorelin), mirerelin (meterelin) and goserelin (lecirelin). Especially preferred is leuprolide acetate.
It may also be particularly advantageous to use a compound or pharmaceutical composition of the invention in combination with at least one substance selected from the group consisting of steroidal or non-steroidal antiandrogenic agents or antiestrogenic agents, chemotherapeutic agents, GnRH antagonist peptides (antiangiogenic peptides), α -reductase inhibitors, α -receptor inhibitors, aromatase inhibitors, 17 β -hydroxysteroid dehydrogenase inhibitors, adrenoandrogen production inhibitors, kinase inhibitors, hormone therapy agents and agents inhibiting cell growth factors or their receptors.
The above-mentioned "chemotherapeutic agent" includes ifosfamide (ifosfamide), doxorubicin (adriamycin), pellomycin (polyplomycin), cisplatin (cissplatin), cyclophosphamide (cyclophosphamide), 5-FU, UFT, methotrexate (methotrexate), mitomycin C (mitomycin C), mitoxantrone (mitoxantrone), and the like. The above-mentioned "GnRH antagonistic peptide" includes non-oral GnRH antagonistic peptides such as cetrorelix (cetrorelix), ganirelix (ganirelix), abarelix (abarelix), and the like.
In addition to being beneficial for human therapy, the compounds and pharmaceutical compositions of the present invention may also find application in veterinary therapy for pets, animals of the introduced species and mammals in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compound of the present invention includes pharmaceutically acceptable derivatives thereof.
In some embodiments, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time according to a dosing regimen. The compounds disclosed herein may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the invention may be administered separately from the other therapeutic agents, by the same or different routes of administration, or in the same pharmaceutical composition.
General synthetic procedure
To illustrate the invention, the following examples are set forth. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention.
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I) or formula (II), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin Haojian Yunyu chemical Co., Ltd, Tianjin Shucheng chemical reagent factory, Wuhan Xin Huayuan scientific and technological development Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaolingyi factory.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N, N-dimethylacetamide and N, N-dimethylformamide were used as they were previously dried over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants.
1H NMR spectra were recorded using a Bruker 400MHz or 600MHz NMR spectrometer.1H NMR Spectrum in CDC13、DMSO-d6、CD3OD or acetone-d6TMS (0ppm) or chloroform (7.26ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (singlets, singlet), d (doublets ), t (triplets, triplets), q (quatets, quartets), m (multiplets ), br (broadpededwideams), brs (broadpedsinglets, wideadlets), dd (doublets, ddles of doublets), ddd (doublets ), ddt (doublets of doublets, doublets), dt (doublets of triplets, doublets), dq (doublets of quatts, doublets), td (triplets of doublets, triplets), tt (triplets of triplets, triplets), triplets (quadruplands ). Coupling constant J, expressed in Hertz (Hz).
Low resolution Mass Spectrometry (MS) data were determined using Agilent 6120 quadrupole HPLC-M (column model: Zorbax SB-C18,2.1 × 30mm,3.5 microns, 6min, flow rate of 0.6 mL/min. mobile phase: 5% -95% (CH with 0.1% formic acid)3CN) in (H containing 0.1% formic acid)2O) by electrospray ionization (ESI) at 210nm/254nm, with UV detection.
Pure compounds were detected by UV at 210nm/254nm using Agilent 1260pre-HPLC or Calesep pump 250pre-HPLC (column model: NOVASEP 50/80mm DAC).
The following acronyms are used throughout the invention:
Figure BDA0002446925260000161
the following synthetic schemes describe the steps for preparing the disclosed compounds, wherein R has the definitions described herein, unless otherwise indicated.
Intermediate Synthesis scheme 1
Formula (A), (B) and11) The intermediate compound shown can be prepared by the following processes: formula (A), (B) and1) Reacting the compound with urea to obtain a compound of the formula (A)2) A compound shown in the specification; then formula (A), (B), (C), (2) A compound of the formula3) Reacting the compound shown in the formula (A) to obtain a compound shown in the formula (B)4) The compounds shown. Formula (A), (B) and4) Brominating a compound shown in the formula (I) to obtain a compound shown in the formula (I)5) The compounds shown. Formula (A), (B) and5) A compound of the formula6) The compound shown in the formula (I) is reacted to obtain7) A compound shown in the specification; formula (A), (B) and7) A compound of the formula8) Reacting the compound shown in the formula (A) to obtain a compound shown in the formula (B)9) The compounds shown. Formula (A), (B) and9) Reacting the compound with boron tribromide to obtain the compound shown in the formula (A)10) The compounds shown. Formula (A), (B) and10) The protecting group on the amino group of the compound is shown in the formula (A)11) Intermediate compounds are shown.
Figure BDA0002446925260000171
Synthesis scheme 1
Formula (A), (B) and14) The compound shown can be prepared by the following steps: formula (A), (B) and11) A compound of the formula12) Reacting the compound shown in the formula (A) to obtain a compound shown in the formula (B)13) A compound shown in the specification; formula (A), (B) and13) Removing the protecting group of the compound to obtain a compound of the formula (I)14) The target product shown.
Figure BDA0002446925260000172
Wherein L is a leaving group such as: F. cl, Br, I,
Figure BDA0002446925260000181
Synthesis scheme 2
Formula (A), (B) and15) The compound shown can be prepared by the following steps: formula (A), (B) and14) Aminoalkylation of a compound of formula (I) to give a compound of formula (I)15) The target product shown.
Figure BDA0002446925260000182
The compounds, pharmaceutical compositions and uses thereof provided by the present invention are further illustrated below in connection with the examples.
Examples
EXAMPLE 1 Synthesis of (R) -4- (3- (3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanamide
Figure BDA0002446925260000183
Step 1) -Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) Urea
(2-fluoro-6- (trifluoromethyl) phenyl) methylamine (1.5g,25.89mmol) and urea (6.22g,104.00mmol) were added to a 100mL single neck round bottom flask, concentrated hydrochloric acid (12M,3mL) and water (11mL) were added and reacted at 100 ℃ for 12 hours; stopping the reaction, cooling to room temperature, adding ice water (20mL), and filtering to obtain a white solid; the resulting white solid was added to ethyl acetate (40mL), stirred for 10 minutes, filtered, and the filter cake was dried at 50 ℃ for 16 hours to give the title compound as a white solid (4.6g, 75%).
MS(ESI,pos.ion)m/z:237.2[M+H]+
1H NMR(600MHz,DMSO-d6)δ(ppm)7.63–7.57(m,3H),6.17(t,J=4.5Hz,1H),5.49(s,2H),4.37(d,J=4.7Hz,2H).
Step 2) Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
1- (2-fluoro-6- (trifluoromethyl) benzyl) urea (4.0g,17.0mmol) and sodium iodide (3.82g,25.5mmol) were added to a 100mL single neck round bottom flask and acetonitrile (35mL) was added; under the protection of nitrogen, cooling the mixture in a low-temperature bath at 0 ℃, dropwise adding diketene (2mL), then adding trimethylchlorosilane (3.3mL), slowly heating to 25 ℃, and continuing to react for 16 hours; water (30mL) was added and the reaction was continued for 5 hours; the reaction was stopped, filtered and the filter cake was dried at 50 ℃ overnight to give the title compound as a white solid (4.5g, 88%).
MS(ESI,pos.ion)m/z:303.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)8.96(s,1H),7.54(d,J=7.8Hz,1H),7.41(dd,J=13.2,7.9Hz,1H),7.28–7.22(m,1H),5.35(s,2H),2.15(s,3H).
Step 3) Synthesis of 5-bromo-1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione (4.5g,15.0mmol) and acetic acid (23mL) were added to a 100mL single-neck round-bottom flask at 25 ℃ followed by dropwise addition of liquid bromine (1.5mL) and reaction continued for 14H; the reaction was stopped, water (30mL) was added, stirred for 10 min, filtered and the filter cake dried at 50 ℃ overnight to give the title compound as a white solid (5.44g, 96%).
MS(ESI,pos.ion)m/z:381.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)8.75(s,1H),7.58(d,J=7.8Hz,1H),7.47(dd,J=13.2,7.9Hz,1H),7.32(s,1H),5.50(s,2H),2.44(s,3H).
Step 4) (R) -tert-butyl (2- (5-bromo-3- (2-fluoro-6- (trifluoromethyl) benzyl)-4-methyl-2, 6-dioxo- Synthesis of 2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate
5-bromo-1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione (2.2g,5.8mmol) and tetrahydrofuran (35mL) were added to a 100mL single neck round bottom flask at 25 deg.C, and (R) -tert-butyl (2-hydroxy-1-phenylethyl) carbamate (1.7g,7.2mmol) and triphenylphosphine (2.3g,8.7mmol) were added followed by diethyl azodicarboxylate (2.0g,8.5mmol) and the reaction was stirred for 14 hours; the reaction was stopped, spin-dried under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (3.42g, 99%).
MS(ESI,pos.ion)m/z:500.2[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)8.59(s,3H),8.15(s,1H),7.59(dd,J=13.2,7.8Hz,1H),7.54–7.47(m,1H),7.26(s,3H),5.35(d,J=4.9Hz,2H),4.92(dd,J=14.9,8.6Hz,1H),4.09(dd,J=19.4,10.4Hz,1H),4.01(dd,J=6.4,2.8Hz,1H),2.54(s,3H),1.32(s,9H).
Step 5) (R) -tert-butyl (2- (5- (2-fluoro-3-methoxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) - Synthesis of 4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate
(R) -tert-butyl (2- (5-bromo-3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (5.1g,16.7mmol), (2-fluoro-3-methoxy-phenyl) boronic acid (4.25g,25.0mmol), potassium carbonate (6.98g,50.0mmol), 2-dicyclohexylphosphonium-2 ',4',6' -triisopropylbiphenyl (810mg,1.67mmol), palladium acetate (190mg,0.83mmol) and toluene (160mL) were added to a 250mL single neck flask and reacted at 110 ℃ for 16 hours under nitrogen; the reaction was stopped, cooled to room temperature, filtered, the filtrate was spin-dried under reduced pressure and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a white solid (7.25g, 67%). MS (ESI, pos.ion) M/z 546.2[ M + H-100 ]]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.67(d,J=7.7Hz,1H),7.57(dt,J=19.7,8.2Hz,2H),7.30–7.21(m,6H),7.17(t,J=6.0Hz,2H),5.33(s,2H),4.94(d,J=7.0Hz,1H),4.04(dd,J=13.7,7.5Hz,2H),3.86(s,3H),2.10(s,3H),1.34(s,9H).
Step 6) (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3-hydroxyphenyl) -1- (2-fluoro-6- (trifluoromethyl) Synthesis of (yl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Adding (R) -tert-butyl (2- (5- (2-fluoro-3-acetoxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (7.25g,11.2mmol) and dichloromethane (60mL) to a 250mL single-neck round-bottom flask, dropwise adding boron tribromide (23mL) after cooling at-78 ℃ for half an hour under nitrogen protection, and continuing to stir for reaction for 3 hours; the reaction mixture was then transferred to 25 ℃ and reacted for 12 hours, the reaction was stopped, water (30mL) was added, the aqueous phase was extracted with dichloromethane (40mL) after separation, the organic phases were combined, dried by spinning under reduced pressure, and purified by column chromatography (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound as a white solid (5.82g, 98%).
MS(ESI,pos.ion)m/z:532.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)9.93(s,1H),7.94(s,2H),7.66(d,J=6.3Hz,1H),7.62–7.52(m,2H),7.35(d,J=8.1Hz,5H),7.05–6.93(m,2H),6.63–6.41(m,1H),5.33(s,2H),4.47(t,J=6.8Hz,1H),4.31–4.11(m,2H),2.13(s,3H).
Step 7) (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4- Synthesis of methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate
(R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3-hydroxyphenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione (1.68g,3.16mmol) and dichloromethane (18mL) were added to a 100mL single neck round bottom flask at 25 deg.C, followed by triethylamine (0.67mL,4.8mmol) and di-tert-butyl dicarbonate (697mg,3.16mmol), and the reaction was stirred for an additional 5 hours; the reaction was stopped, spin-dried under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a white solid (1.21g, 60%).
MS(ESI,pos.ion)m/z:532.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)9.86(s,1H),7.65(d,J=7.5Hz,1H),7.55(dt,J=19.8,8.1Hz,2H),7.25–7.20(m,6H),7.03–6.92(m,2H),5.31(s,2H),4.93(d,J=6.9Hz,1H),4.02(dd,J=14.1,7.0Hz,2H),2.09(s,3H),1.33(s,9H).
Step 8) (R) -methyl 4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (tris) Synthesis of fluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanoate
(R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.5g,0.79mmol) and N, N-dimethylformamide (8.0mL) were added to a 100mL single-neck round-bottom flask, potassium carbonate (221mg,1.58mmol) and methyl 4-bromobutyrate (286mg,1.58mmol) were added, the reaction was allowed to proceed at 70 ℃ for 12 hours, the reaction was stopped, water (50mL) was added after cooling to room temperature, ethyl acetate was added for extraction (50mL × 2), the organic phases were combined, column chromatography was performed under reduced pressure, and isolation and purification (petroleum ether/ethyl acetate (v/v) ═ 3/1) gave the title compound as a viscous liquid (456mg, 79%).
MS(ESI,pos.ion)m/z:632.7[M+H-100]+
1H NMR(400MHz,CDCl3)δ(ppm)7.57(d,J=7.8Hz,1H),7.40(d,J=9.7Hz,3H),7.34(t,J=7.5Hz,2H),7.27(d,J=9.6Hz,2H),7.12(t,J=8.0Hz,1H),6.99(t,J=7.9Hz,1H),6.84(t,J=6.7Hz,1H),4.46–4.31(m,1H),4.16–4.08(m,4H),3.70(s,3H),2.57(t,J=7.2Hz,2H),2.16(dd,J=6.6,5.0Hz,2H),2.10(s,3H),1.41(s,9H).
Step 9) (R) -4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) Synthesis of yl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butyric acid
(R) -methyl 4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanoate (456mg,0.62mmol) and methanol (10mL) were added to a 100mL single-neck round-bottom flask at 25 ℃, lithium hydroxide (45mg,1.84mmol, dissolved in 2mL water) was added, the reaction was continued for 14 hours with stirring, the reaction was stopped, citric acid was added to adjust pH to 5, water (50mL) was added, then ethyl acetate was added for extraction (40mL × 2), the organic phases were combined, dried under reduced pressure, and purified by column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (433mg, 97%).
MS(ESI,pos.ion)m/z:618.6[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.62–7.50(m,3H),7.28–7.19(m,5H),7.15(t,J=6.8Hz,2H),5.32(s,2H),4.97–4.88(m,1H),4.08(t,J=6.3Hz,2H),4.02(d,J=7.8Hz,2H),2.41(t,J=7.3Hz,2H),2.10(s,3H),2.00–1.95(m,2H),1.33(s,9H).
Step 10) (R) -4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoro-phenyl) Synthesis of methyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanamide
(R) -4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butyric acid (258mg,0.36mmol), ammonium chloride (48mg,0.89mmol), HATU (150mg,0.39mmol) and N, N-dimethylformamide (4mL) were added to a 100mL single-necked round-bottomed flask at 25 ℃ followed by diisopropylethylamine (139mg,1.08mmol), stirring for 12 hours, stopped, added water (30mL), then dichloromethane extracted (50mL × 2), the organic phases combined, rotary dried under reduced pressure, and isolated and purified (petroleum ether/ethyl acetate (v/v) ═ 1/2) to give the title compound as a white solid (231mg, 89.5%).
MS(ESI,pos.ion)m/z:617.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.6Hz,1H),7.60–7.49(m,2H),7.33(s,2H),7.26–7.22(m,6H),7.14(t,J=7.1Hz,2H),6.77(s,1H),5.32(s,2H),4.99–4.88(m,1H),4.08–3.99(m,4H),2.25(t,J=7.3Hz,2H),2.10(s,3H),1.98–1.92(m,2H),1.33(s,9H).
Step 11) (R) -4- (3- (3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl Synthesis of 1,2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanamide-2, 4-dioxo-2
(R) -4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) butanamide (256mg,0.36mmol) and dichloromethane (10mL) were added to a 100mL single-necked round bottom flask at 25 deg.C, followed by methanesulfonic acid (0.105g,1.07mmol), stirring for an additional 5 hours, reaction was stopped, saturated sodium bicarbonate solution (30mL) was added, followed by extraction with dichloromethane (30mL × 3), organic phases were combined, spin-dried under reduced pressure, and column chromatography purification (dichloromethane/methanol (v/v) ═ 20/1) gave the title compound as a white solid (189mg, 85.1%).
MS(ESI,pos.ion)m/z:617.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.65(d,J=6.8Hz,1H),7.60–7.52(m,2H),7.34(s,1H),7.26(t,J=4.9Hz,4H),7.20–7.08(m,3H),6.78(s,1H),6.76–6.59(m,1H),5.34(s,2H),4.16–4.10(m,1H),4.05(t,J=6.3Hz,2H),3.98–3.85(m,2H),2.25(t,J=7.4Hz,2H),2.09(s,3H),1.99–1.92(m,2H).
EXAMPLE 2 Synthesis of (R) -2- (3- (3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetamide
Figure BDA0002446925260000211
Step 1) (R) -methyl 2- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (tris) Synthesis of fluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetate
The title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.5g,0.79mmol), methyl 2-bromoacetate (242mg,1.58mmol) and potassium carbonate (221mg,1.58mmol) in N, N-dimethylformamide (6mL), and the crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 2/1) to give the title compound as a colorless viscous mass (542mg, 97%).
MS(ESI,pos.ion)m/z:604.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.67(d,J=7.7Hz,1H),7.62–7.51(m,2H),7.28–7.20(m,6H),7.17–7.08(m,2H),5.33(s,2H),4.96–4.84(m,3H),4.10–3.98(m,2H),3.72(s,3H),2.11(s,3H),1.34(s,9H).
Step 2) (R) -2- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) Synthesis of yl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetic acid
This step title compound was prepared as described in example 1, step 9 by reacting (R) -methyl 2- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetate (0.542g,0.77mmol) and lithium hydroxide (57mg,2.28mmol) in methanol (5mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (410mg, 77%).
MS(ESI,pos.ion)m/z:590.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.67(d,J=7.6Hz,1H),7.62–7.51(m,2H),7.25–7.22(m,6H),7.16–7.05(m,2H),5.33(s,2H),4.93(dd,J=11.0,3.3Hz,1H),4.79(s,2H),4.09–3.98(m,2H),2.11(s,3H),1.34(s,9H).
Step 3) (R) -4- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoro-phenyl) Synthesis of methyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetamide
This title compound was prepared as described in example 1, step 10 by reacting (R) -2- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetic acid (0.41g,0.59mmol), ammonium chloride (79mg,1.48mmol), HATU (251mg,0.65mmol) and diisopropylethylamine (0.3mL,2.0mmol) in N, N-dimethylformamide (6mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give the title compound as a white solid (328mg, 80.8%).
MS(ESI,pos.ion)m/z:589.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.67(d,J=7.6Hz,1H),7.62–7.47(m,3H),7.31–7.21(m,5H),7.17–7.11(m,1H),7.07(t,J=7.6Hz,1H),5.33(s,2H),5.00–4.89(m,1H),4.55(s,2H),4.12–3.96(m,2H),2.11(s,3H),1.34(s,9H).
Step 4) (R) -2- (3- (3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl Synthesis of 1,2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetamide-2, 4-dioxo-2
This step was prepared by the method described in example 1, step 11, reacting (R) -2- (3- (3- (2- ((tert-butoxycarbonyl) amino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -2-fluorophenoxy) acetamide (0.328g,0.48mmol) and methanesulfonic acid (0.14g,1.43mmol) in dichloromethane (6mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (233mg, 82.6%).
MS(ESI,pos.ion)m/z:589.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=6.7Hz,1H),7.59–7.53(m,2H),7.49(s,1H),7.43(s,1H),7.30–7.23(m,4H),7.22–7.16(m,1H),7.13(t,J=7.8Hz,1H),7.07(t,J=8.0Hz,1H),6.82–6.63(m,1H),5.36(s,2H),4.55(s,2H),4.14(dd,J=12.7,6.3Hz,1H),4.01–3.85(m,2H),2.11(s,3H).
EXAMPLE 3 Synthesis of (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- (oxetan-3-yloxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000221
Step 1) (R) -tert-butyl (2- (5- (2-fluoro-3- (oxetan-3-yloxy) phenyl) -3- (2-fluoro-6- (trifluoro-2-oxo) Synthesis of methyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate Become into
The title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.45g,0.71mmol), oxetan-3-yl 4-methylbenzenesulfonate (243mg,1.06mmol) and cesium carbonate (232mg,0.71mmol) in N, N-dimethylformamide (8mL) and the crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 2/1) to give the title compound as a white solid (349mg, 71%).
MS(ESI,pos.ion)m/z:588.0[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.62–7.51(m,2H),7.29–7.18(m,6H),7.12(dd,J=15.2,7.5Hz,1H),6.84(t,J=8.0Hz,1H),5.38–5.29(m,3H),4.95(dd,J=12.8,6.3Hz,3H),4.64–4.56(m,2H),4.09–3.96(m,2H),2.11(s,3H),1.34(s,9H).
Step 2) (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- (oxetan-3-yloxy) phenyl) -1- Synthesis of (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3- (oxetan-3-yloxy) phenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.33g,0.48mmol) and phosphoric acid (1.5mL) in dichloromethane (5mL) and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (148mg, 52%).
MS(ESI,pos.ion)m/z:588.0[M+H]+
1H NMR(600MHz,DMSO-d6)δ(ppm)7.65(dd,J=7.0,2.8Hz,1H),7.59–7.53(m,2H),7.29–7.22(m,6H),7.20–7.17(m,1H),7.15–7.11(m,1H),5.35(s,2H),5.18–5.10(m,1H),4.58–4.53(m,1H),4.13(dd,J=14.5,7.7Hz,1H),3.99–3.82(m,5H),2.10(s,3H).
Example Synthesis of 43- ((R) -2-amino-2-phenylethyl) -5- (2-fluoro-3- (((R) -tetrahydrofuran-3-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000231
Step 1) tert-butyl ((R) -2- (5- (2-fluoro-3- (((R) -tetrahydrofuran-3-yl) oxy) phenyl) -3- (2-fluoro-6-) (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamic acid Synthesis of esters
This title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.5g,0.79mmol), ((3S) -tetrahydrofuran-3-yl) 4-methylbenzenesulfonate (287mg,1.18mmol) and cesium carbonate (258mg,0.79mmol) in N, N-dimethylformamide (6mL) and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid Body (495mg, 89%).
MS(ESI,pos.ion)m/z:602.2[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.6Hz,1H),7.60–7.49(m,2H),7.28–7.20(m,6H),7.15(t,J=5.4Hz,2H),5.32(s,2H),5.08(s,1H),4.97–4.87(m,1H),4.10–3.98(m,2H),3.97–3.80(m,4H),3.80–3.74(m,1H),2.24(dd,J=13.8,6.0Hz,1H),2.10(s,3H),2.04–1.96(m,1H),1.33(s,9H).
Step 2)3- ((R) -2-amino-2-phenylethyl) -5- (2-fluoro-3- (((R) -tetrakis)Hydro furan-3-yl) oxy) benzene Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
The title compound was prepared as described in example 1, step 11 by reacting tert-butyl ((R) -2- (5- (2-fluoro-3- (((R) -tetrahydrofuran-3-yl) oxy) phenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.45g,0.64mmol) and methanesulfonic acid (189mg,1.93mmol) in dichloromethane (8mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (384mg, 99%).
MS(ESI,pos.ion)m/z:602.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.65(d,J=6.7Hz,1H),7.61–7.52(m,2H),7.29–7.23(m,4H),7.19(dd,J=7.8,3.8Hz,1H),7.14(t,J=5.3Hz,2H),6.79–6.61(m,1H),5.35(s,2H),5.08(s,1H),4.16–4.10(m,1H),3.95–3.82(m,5H),3.77(td,J=8.1,5.0Hz,1H),2.29–2.19(m,1H),2.09(s,3H),2.04–1.97(m,1H).
Example Synthesis of 53- ((R) -2-amino-2-phenylethyl) -5- (2-fluoro-3- (((S) -tetrahydrofuran-3-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000241
Step 1) tert-butyl ((R) -2- (5- (2-fluoro-3- (((S) -tetrahydrofuran-3-yl) oxy) phenyl) -3- (2-fluoro-6-) (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamic acid Synthesis of esters
This title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.5g,0.79mmol), ((3R) -tetrahydrofuran-3-yl) 4-methylbenzenesulfonate (287mg,1.18mmol) and cesium carbonate (258mg,0.79mmol) in N, N-dimethylformamide (6mL) and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid Body (484mg, 87%).
MS(ESI,pos.ion)m/z:602.2[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.62–7.50(m,2H),7.28–7.20(m,6H),7.15(t,J=5.9Hz,2H),5.33(s,2H),5.09(t,J=4.8Hz,1H),4.99–4.89(m,1H),4.11–3.73(m,7H),2.30–2.20(m,1H),2.11(s,3H),2.06–1.98(m,1H),1.34(s,9H).
Step 2)3- ((R) -2-amino-2-phenylethyl) -5- (2-fluoro-3- (((S) -tetrahydrofuran-3-yl) oxy) benzene Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
The title compound was prepared as described in example 1, step 11 by reacting tert-butyl ((R) -2- (5- (2-fluoro-3- (((S) -tetrahydrofuran-3-yl) oxy) phenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.47g,0.67mmol) and methanesulfonic acid (197mg,2.01mmol) in dichloromethane (8mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (371mg, 92%).
MS(ESI,pos.ion)m/z:602.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.65(d,J=6.8Hz,1H),7.60–7.52(m,2H),7.29–7.23(m,4H),7.21–7.17(m,1H),7.17–7.10(m,2H),6.80–6.61(m,1H),5.35(s,2H),5.08(t,J=4.7Hz,1H),4.16–4.09(m,1H),3.96–3.81(m,5H),3.81–3.74(m,1H),2.30–2.19(m,1H),2.10(s,3H),2.05–1.98(m,1H).
EXAMPLE 6 Synthesis of (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000251
Step 1) (R) -tert-butyl (2- (5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) phenyl) -3- (2-fluoro-6-) (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamic acid Synthesis of esters
The title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.5g,0.79mmol), tetrahydropyran-4-yl 4-methylbenzenesulfonate (304mg,1.18mmol) and cesium carbonate (258mg,0.79mmol) in N, N-dimethylformamide (8mL) and the crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (466mg, 82%).
MS(ESI,pos.ion)m/z:616.2[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.61–7.50(m,2H),7.25–7.20(m,7H),7.13(dd,J=15.3,7.6Hz,1H),5.32(s,2H),4.99–4.88(m,1H),4.59(s,1H),4.02(d,J=8.1Hz,1H),3.86(d,J=11.5Hz,2H),3.49(t,J=10.1Hz,2H),3.43–3.38(m,2H),2.10(s,3H),1.99(dd,J=8.9,3.9Hz,2H),1.67–1.57(m,2H),1.34(s,9H).
Step 2) (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) benzene Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) phenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (406mg,0.57mmol) and methanesulfonic acid (167mg,1.7mmol) in dichloromethane (6mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (310mg, 89%).
MS(ESI,pos.ion)m/z:616.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(s,1H),7.61–7.51(m,2H),7.30–7.21(m,5H),7.19(d,J=4.5Hz,1H),7.12(dd,J=15.8,8.0Hz,1H),6.71–6.69(m,1H),5.34(s,2H),4.58(d,J=1.1Hz,1H),4.12(t,J=6.1Hz,1H),3.96–3.93(m,2H),3.88–3.83(m,2H),3.48(t,J=9.9Hz,2H),2.09(s,3H),2.02–1.95(m,2H),1.62(d,J=8.9Hz,2H).
EXAMPLE 7 Synthesis of (R) -3- (2- (dimethylamino) -2-phenylethyl) -5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000261
(R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- ((tetrahydro-2H-pyran-4-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione (67mg,0.11mmol) and aqueous formaldehyde (0.09mL,1.0mmol, 37%) were added to a 100mL one-necked round-bottomed flask at 25 deg.C, then acetic acid (0.012mL,0.21mmol) was added and stirred for 2 hours, sodium cyanoborohydride (35mg,0.53mmol) was added and the reaction was continued for 12 hours with stirring, the reaction was stopped, a saturated sodium bicarbonate solution (20mL) was added, then dichloromethane was extracted (30mL column chromatography × 2), the organic phases were combined, spin-dried under reduced pressure, and isolated and purified (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound as a white solid (41mg, 59%).
MS(ESI,pos.ion)m/z:644.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.64(d,J=7.5Hz,1H),7.60–7.50(m,2H),7.27–7.20(m,4H),7.15–7.07(m,3H),6.66(m,1H),5.32(s,2H),4.58(td,J=8.3,4.3Hz,1H),4.51–4.35(m,1H),3.97(ddd,J=19.1,12.9,6.7Hz,1H),3.86(dd,J=9.6,5.6Hz,2H),3.79(t,J=7.6Hz,1H),3.52–3.44(m,2H),2.07(d,J=6.9Hz,2H),2.01(s,3H),1.98(s,3H),1.68–1.56(m,2H).
EXAMPLE 8 Synthesis of (R) -5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000262
Step 1) (R) -tert-butyl (2- (5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6-) (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamic acid Synthesis of esters
The title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.43g,0.68mmol), (1-acetyl-4-piperidinyl) 4-methylbenzenesulfonate (303mg,1.02mmol) and cesium carbonate (221mg,0.68mmol) in N, N-dimethylformamide (8mL) and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (R) ((R) (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-phenylethyl) in (8mL) 435mg, 84%).
MS(ESI,pos.ion)m/z:657.2[M+H-100]+
1H NMR(400MHz,CDCl3)δ(ppm)7.58(d,J=7.8Hz,1H),7.49–7.30(m,7H),7.13(t,J=7.9Hz,1H),7.04(t,J=7.8Hz,1H),6.90(dd,J=9.9,5.5Hz,1H),5.55(s,2H),4.45–4.30(m,1H),4.15–4.08(m,1H),3.81–3.60(m,4H),3.52–3.32(m,2H),2.13(s,3H),2.10(s,3H),1.97–1.86(m,4H),1.41(s,9H).
Step 2) (R) -5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenylethane Synthesis of 1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.35g,0.46mmol) and methanesulfonic acid (196mg,2.0mmol) in dichloromethane (6mL), separating the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (281mg, 93%).
MS(ESI,pos.ion)m/z:657.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=6.9Hz,1H),7.59–7.48(m,2H),7.30–7.23(m,5H),7.19(dd,J=10.3,5.1Hz,1H),7.15–7.09(m,1H),6.71(dt,J=12.8,6.7Hz,1H),5.35(s,2H),4.74–4.59(m,1H),4.16–4.08(m,1H),4.00–3.81(m,3H),3.72–3.62(m,1H),3.29–3.20(m,2H),2.10(s,3H),2.02(s,3H),1.94–1.85(m,2H),1.69–1.63(m,1H),1.58–1.51(m,1H).
Example 9 Synthesis of (R) -5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2- (dimethylamino) -2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000271
(R) -5- (3- ((1-acetylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione (204mg,0.31mmol) and aqueous formaldehyde (0.19mL,2.6mmol, 37%) were added to a 100mL one-necked round bottom flask at 25 deg.C, followed by addition of acetic acid (0.038mL,0.63mmol), stirring for 1 hour, addition of sodium cyanoborohydride (82mg,1.25mmol), stirring for 12 hours, stopping the reaction, addition of a saturated solution of sodium bicarbonate (20mL), followed by extraction with dichloromethane (30mL × 2), combining the organic phases, rotary drying under reduced pressure, and column chromatography purification (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound as a white solid (189mg, 88%).
MS(ESI,pos.ion)m/z:685.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.65(d,J=7.5Hz,1H),7.60–7.50(m,2H),7.26(brs,5H),7.16–7.10(m,3H),5.32(s,2H),4.62(d,J=3.3Hz,1H),4.49–4.35(m,1H),4.06–3.81(m,3H),3.72–3.64(m,1H),3.31–3.18(m,2H),2.09–2.00(m,12H),1.96–1.87(m,2H),1.69–1.62(m,1H),1.57–1.50(m,1H).
EXAMPLE 10 Synthesis of (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- ((1- (methylsulfonyl) piperidin-4-yl) oxy) phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000281
Step 1) (R) -tert-butyl (2- (5- (2-fluoro-3- ((1- (methylsulfonyl) piperidin-4-yl) oxy) phenyl) -3- (2- Fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) amino Synthesis of formic acid esters
This step was performed by the method described in example 1, step 8, using (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.3g,0.47mmol), (1-methanesulfonyl-4-piperidinyl) 4-methylbenzenesulfonate (237mg,0.71mmol) and cesium carbonate (154mg,0.47mmol) in N, N-dimethylformamide (6mL), and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (308mg, 81.8%).
MS(ESI,pos.ion)m/z:693.2[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.67(s,1H),7.62–7.51(m,2H),7.28–7.20(m,7H),7.18–7.12(m,1H),5.33(s,2H),4.97–4.89(m,1H),4.60–4.54(m,1H),4.11–3.96(m,2H),3.36(dd,J=6.7,4.5Hz,2H),3.18–3.12(m,2H),2.90(s,3H),2.11(s,3H),2.06–2.00(m,2H),1.79(ddd,J=9.1,7.9,4.0Hz,2H),1.33(s,9H).
Step 2) (R) -3- (2-amino-2-phenylethyl) -5- (2-fluoro-3- ((1- (methylsulfonyl) piperidin-4-yl) oxy) Synthesis of phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
The title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3- ((1- (methylsulfonyl) piperidin-4-yl) oxy) phenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.308g,0.39mmol) and methanesulfonic acid (114mg,1.16mmol) in dichloromethane (6mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (242mg, 89.9%).
MS(ESI,pos.ion)m/z:693.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.0Hz,1H),7.61–7.53(m,2H),7.30–7.22(m,6H),7.21–7.17(m,1H),7.16–7.11(m,1H),5.35(s,2H),4.57(s,1H),4.13(dd,J=7.5,4.7Hz,1H),3.91(ddd,J=16.0,14.1,7.7Hz,2H),3.36(s,2H),3.14(t,J=8.2Hz,2H),2.90(s,3H),2.10(s,3H),2.02(d,J=4.1Hz,2H),1.78(d,J=8.2Hz,2H).
EXAMPLE 11 Synthesis of (R) -3- (2-amino-2-phenylethyl) -5- (3- ((1- (cyclopropanoyl) piperidin-4-yl) oxy) -2-fluorophenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000291
Step 1) (R) -tert-butyl (2- (5- (3- ((1-cyclopropanylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-Dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamic acid ester Synthesis of acid esters
This step was performed by the method described in example 1, step 8, using (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.3g,0.475mmol), (1-cyclopropanoyl-4-piperidinyl) 4-methylbenzenesulfonate (230mg,0.71mmol) and cesium carbonate (154mg,0.47mmol) in N, N-dimethylformamide (6mL), and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (312mg, 83.9%).
MS(ESI,pos.ion)m/z:783.3[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.62–7.51(m,2H),7.30–7.20(m,7H),7.15(dd,J=15.2,7.6Hz,1H),5.32(s,2H),4.93(dt,J=14.9,7.3Hz,1H),4.65(td,J=7.1,3.7Hz,1H),4.11–3.80(m,5H),3.55(s,1H),2.11(s,3H),2.05–1.89(m,4H),1.33(s,9H),0.75–0.69(m,4H).
Step 2) (R) -3- (2-amino-2-phenylethyl) -5- (3- ((1- (cyclopropanoyl) piperidin-4-yl) oxy) -2-fluoro Synthesis of phenyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (3- ((1-cyclopropanylpiperidin-4-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.312g,0.4mmol) and methanesulfonic acid (117mg,1.19mmol) in dichloromethane (6mL), and the crude product was isolated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (237mg, 87.1%).
MS(ESI,pos.ion)m/z:683.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=6.9Hz,1H),7.59–7.53(m,2H),7.30–7.22(m,6H),7.19(dd,J=9.5,4.0Hz,1H),7.16–7.10(m,1H),5.35(s,2H),4.64(s,1H),4.13(dd,J=12.3,6.2Hz,1H),4.06–3.83(m,5H),3.59–3.49(m,1H),2.10(s,3H),2.00(dd,J=6.7,5.0Hz,2H),1.61(ddd,J=11.2,7.8,4.1Hz,2H),1.26–1.21(m,1H),0.71(dd,J=9.5,6.5Hz,4H).
EXAMPLE 12 Synthesis of (R) -5- (3- ((1-Acetylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000301
Step 1) (R) -tert-butyl (2- (5- (3- ((1-acetylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) amino Synthesis of formic acid esters
This step title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.4g,0.63mmol), (1-acetylazetidin-3-yl) 4-methylbenzenesulfonate (255mg,0.95mmol) and cesium carbonate (207mg,0.63mmol) in N, N-dimethylformamide (8mL) and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid Body (411mg, 89.1%).
MS(ESI,pos.ion)m/z:729.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.7Hz,1H),7.61–7.50(m,2H),7.29–7.20(m,6H),7.15(dd,J=15.4,7.8Hz,1H),6.95(t,J=7.9Hz,1H),5.32(s,2H),5.09(ddd,J=10.1,6.6,3.9Hz,1H),4.94(dt,J=15.8,7.8Hz,1H),4.57(dd,J=15.7,6.5Hz,1H),4.34–4.26(m,1H),4.18–4.12(m,1H),4.05–4.00(m,2H),3.86–3.78(m,1H),2.10(s,3H),1.80(s,3H),1.33(s,9H).
Step 2) (R) -5- (3- ((1-Acetylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenyl Synthesis of ethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step the title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (3- ((1-acetylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.4g,0.55mmol) and phosphoric acid (2mL) in dichloromethane (6mL), and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (316mg, 91.6%).
MS(ESI,pos.ion)m/z:629.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=6.6Hz,1H),7.61–7.53(m,2H),7.28–7.25(m,5H),7.19(dd,J=7.9,4.0Hz,1H),7.17–7.12(m,1H),6.95(t,J=8.1Hz,1H),5.35(s,2H),5.12–5.07(m,1H),4.57(dd,J=15.7,6.5Hz,1H),4.34–4.27(m,1H),4.15(dt,J=14.0,4.8Hz,2H),3.97–3.89(m,2H),3.82(dd,J=10.5,2.9Hz,1H),2.11(s,3H),1.81(s,3H).
EXAMPLE 13 Synthesis of (R) -5- (3- ((1-methanesulfonylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-phenylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
Figure BDA0002446925260000311
Step 1) (R) -tert-butyl (2- (5- (3- ((1-methanesulfonylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) Synthesis of carbamates
This step title compound was prepared as described in example 1, step 8 by reacting (R) -tert-butyl (2- (5- (2-fluoro-3-hydroxyphenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.35g,0.55mmol), (1-methanesulfonylazetidin-3-yl) 4-methylbenzenesulfonate (253mg,0.83mmol) and cesium carbonate (181mg,0.56mmol) in N, N-dimethylformamide (6mL) and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white color Solid (256mg, 60.4%).
MS(ESI,pos.ion)m/z:665.1[M+H-100]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=7.6Hz,1H),7.61–7.51(m,2H),7.29–7.20(m,6H),7.18–7.12(m,1H),7.00(t,J=8.1Hz,1H),5.33(s,2H),5.14–5.07(m,1H),4.99–4.89(m,1H),4.34(dd,J=15.4,7.8Hz,2H),4.06–3.96(m,4H),3.09(s,3H),2.11(s,3H),1.34(s,9H).
Step 2) (R) -5- (3- ((1-methanesulfonyl-azetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-amino-2-benzene Synthesis of ylethyl) -1- (2-fluoro-6- (trifluoromethyl) benzyl) -6-methylpyrimidine-2, 4(1H,3H) -dione
This step the title compound was prepared as described in example 1, step 11 by reacting (R) -tert-butyl (2- (5- (3- ((1-methanesulfonylazetidin-3-yl) oxy) -2-fluorophenyl) -3- (2-fluoro-6- (trifluoromethyl) benzyl) -4-methyl-2, 6-dioxo-2, 3-dihydropyrimidin-1 (6H) -yl) -1-phenylethyl) carbamate (0.24g,0.32mmol) and phosphoric acid (2mL) in dichloromethane (5mL), and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give the title compound as a white solid (159mg, 76.2%).
MS(ESI,pos.ion)m/z:665.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)7.66(d,J=5.5Hz,1H),7.62–7.52(m,2H),7.30–7.22(m,4H),7.21–7.10(m,2H),7.00(d,J=6.9Hz,1H),6.87–6.65(m,1H),5.35(s,2H),5.10(d,J=4.7Hz,1H),4.33(dd,J=14.2,6.9Hz,2H),4.16–4.09(m,1H),4.02–3.85(m,4H),3.08(s,3H),2.10(s,3H).
Biological assay
Example A: antagonism of humanized GnRH receptors by Compounds of the invention
Purpose of experiment
The antagonism of the compounds of the present invention against the transfected humanized GnRH receptor in RBL-1 cells (rat basophilic leukemia cells) was evaluated by fluorescence detection of cytosolic calcium flux.
Experimental procedure
The cells were suspended in HBSS buffer (Invitrogen ), 20mM HEPES buffer (Invitrogen ), and then 1.186 × 104The density of cells/well is averaged into the microwell reaction plate. The fluorescent probe (Fluo8 Direct, AAT biosquest) was mixed with HBSS buffer (20 mM HEPES buffer, pH 7.4 was added), and after mixing, each well was added, and the cells were incubated at 30 ℃ for 60 min. The reaction plate was then placed in a microplate reader (FlipR Tetra, Molecular Device) for testing, the test compound or HBSS buffer (blank control) was added, 8nM LH-RH (agonist ligand) was added after 5min, and the change in fluorescence intensity, which is proportional to the cytosolic calcium ion concentration, was measured. Fluorescence intensity of blank control group without compoundThe inhibition rate of the compound was calculated as 100% (inhibition rate of 0%). Measuring the inhibition rate of the compound at different concentrations, and calculating the IC50The corresponding Ki value is calculated by the following equation:
Ki=IC50/[1+(A/EC50A)]. Wherein, A represents the concentration of agonist ligand; EC (EC)50AEC representing agonist ligand50The value is obtained.
The results of the experiment are shown in table a below.
TABLE A antagonistic Effect of the Compounds of the invention on humanized GnRH receptors
Example No. 2 IC50(nM) Ki(nM) Example No. 2 IC50(nM) Ki(nM)
Example 1 1.187 0.326 Example 5 5.101 1.402
Example 2 2.329 0.640 Practice ofExample 8 1.907 0.524
Example 3 1.36 0.374 Example 12 4.414 1.213
Example 4 4.589 1.262 Example 13 3.644 1.002
The experimental result shows that the compound has better antagonistic action on the humanized GnRH receptor.
Example B: pharmacokinetic evaluation of the Compounds of the invention after intravenous injection or gavage in rats and dogs
The present invention evaluates the pharmacokinetic studies of the compounds of the invention in rats and/or dogs for details in the animal information given in table B.
Table B information sheet of the subject animals of the present invention
Figure BDA0002446925260000321
Test method
The compounds of the invention were administered to the animals in the form of 10% DMSO + 10% Kolliphor HS15+ 78% Saline + 2% (2% HCl) solution or 78% Saline + 2% (2% HCl) + 20% PEG400 solution, and the animals were fasted for 12h before administration and allowed free access to water. For the group administered by intravenous injection, the dose was 1mg/kg (rat) or 0.5mg/kg (dog)Post-dose, blood was taken intravenously (approximately 0.2mL blood volume) at the following time points: 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24h (dog) or 0.083, 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24h (rat), EDTA-K is added into the blood sampling tube in advance2Can be used as anticoagulant. For the gavage group, 5mg/kg (rat) or 2.5mg/kg (dog) was administered, and intravenous blood sampling (blood volume taken about 0.2mL) was performed at the following time points after administration: 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24h (dog) or 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24h (rat), EDTA-K is added into the blood sampling tube in advance2Can be used as anticoagulant. Blood samples were centrifuged at 12,000rpm for 2 minutes, plasma was collected and stored at-20 ℃ or-70 ℃.
The plasma samples collected above were processed (frozen plasma was thawed at room temperature, vortexed for 15s, 10-20. mu.L of plasma was taken, 120-150. mu.L of acetonitrile solution containing the internal standard was added, vortexed for 5min, centrifuged at 4,000rpm for 5min, 100. mu.L of supernatant was taken, and 120-150. mu.L of methanol/water (v/v. 1/1) was added and mixed) and then the concentration of the compounds in the plasma was analyzed by LC/MS/MS.
The analysis result shows that the compound of the invention has better pharmacokinetic property in rats and/or dogs. The compound of the invention has good drug property and good clinical application prospect.
In the description herein, references to the description of the term "one embodiment," "an embodiment," "some embodiments," "an example," "a specific example" or "some examples" or the like are intended to mean that a particular feature, structure, material, or characteristic described in connection with the embodiment, or example is included in at least one embodiment, or example of the invention. In this specification, a schematic representation of the above terms does not necessarily refer to the same embodiment, implementation, or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments, implementations, or examples. Furthermore, the various examples, embodiments, or examples described in this specification, as well as features of various examples, embodiments, or examples, may be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (10)

1. A compound which is a compound represented by formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug thereof,
Figure FDA0002446925250000011
wherein:
each R1a、R1b、R1c、R1dAnd R1eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group;
each R2aAnd R2bIndependently H, D, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl or C1-C6A haloalkyl group;
each R3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl、Br、I、-CN、-NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms;
R4is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group;
each R5a、R5b、R5cAnd R5dIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy group), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms;
r is-C1-C6alkylene-C (═ O) NH2、-C1-C6alkylene-C (═ O) NHCH3、-C1-C6alkylene-C (═ O) N (CH)3)2、-C1-C6alkylene-C (═ O) - (C)1-C6Alkyl), -C1-C6alkylene-C (═ O) - (C)1-C6Alkoxy), -C1-C6alkylene-S (═ O)2NH2、-C1-C6alkylene-S (═ O)2NHCH3、-C1-C6alkylene-S (═ O)2N(CH3)2、-C1-C6alkylene-S (═ O)2-(C1-C6Alkyl) or
Figure FDA0002446925250000012
X is-O-, -S-, -C (O) -, -S (O)2-、-CH2-or-NRn-;
RnIs H, D, C1-6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-C10Aryl, 5-10-membered heteroaryl, -C (═ O) - (C)1-C6Alkyl), -C (═ O) - (C)3-C6Cycloalkyl), -C (═ O) - (3-8 atom heterocyclyl), -S (═ O)2-(C1-C6Alkyl), -S (═ O)2-(C3-C6Cycloalkyl) or-S (═ O)2- (3-8 atom-constituting heterocyclic group);
y is 0, 1,2 or 3; and
z is 1,2 or 3.
2. The compound of claim 1, wherein R is-C1-C4alkylene-C (═ O) NH2、-C1-C4alkylene-C (═ O) NHCH3、-C1-C4alkylene-C (═ O) N (CH)3)2、-C1-C4alkylene-C (═ O) - (C)1-C4Alkyl), -C1-C4alkylene-C (═ O) - (C)1-C4Alkoxy), -C1-C4alkylene-S (═ O)2NH2、-C1-C4alkylene-S (═ O)2NHCH3、-C1-C4alkylene-S (═ O)2N(CH3)2、-C1-C4alkylene-S (═ O)2-(C1-C4Alkyl) or
Figure FDA0002446925250000021
X is-O-, -S-or-NRn-;
RnIs H, D, C1-4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, -C (═ O) - (C)1-C4Alkyl), -C (═ O) - (C)3-C6Cycloalkyl), -C (═ O) - (3-6 atom heterocyclyl), -S (═ O)2-(C1-C4Alkyl), -S (═ O)2-(C3-C6Cycloalkyl) or-S (═ O)2- (heterocyclic groups of 3 to 6 atoms).
3. A compound according to claim 1 or 2, wherein R is
Figure FDA0002446925250000022
Figure FDA0002446925250000023
4. The compound of claim 1, wherein each R2aAnd R2bIndependently H, D, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl or C1-C4A haloalkyl group;
each R3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C6Alkylamino, hydroxy-substituted C1-C4Alkyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-C10Aryl or heteroaryl of 5 to 10 atoms;
R4is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy group), C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
5. A compound according to claim 1 or 4, wherein each R is2aAnd R2bIndependently H, D, methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3or-CH2CF2CHF2
Each R3a、R3b、R3c、R3dAnd R3eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, indolyl or quinolinyl;
R4is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, or 2-hydroxyethyl.
6. The compound of claim 1, wherein each R1a、R1b、R1c、R1dAnd R1eIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, or 2-hydroxyethyl;
each R5a、R5b、R5cAnd R5dIndependently H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3Methyl, ethyl, n-propyl, isopropyl, allyl, propenyl, propargyl, propynyl, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2Methoxy, ethoxy, n-propyloxy, isopropyloxy, -OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2Methylthio, ethylthio, methylamino, dimethylamino, ethylamino, hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, indolyl or quinolinyl.
7. The compound of claim 1, which is a compound of formula (II) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt of a compound of formula (II), or prodrug thereof,
Figure FDA0002446925250000041
8. the compound of claim 1, which is a compound having one of the following structures or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound having one of the following structures, or a prodrug of the compound:
Figure FDA0002446925250000042
Figure FDA0002446925250000051
Figure FDA0002446925250000061
9. a pharmaceutical composition comprising a compound of any one of claims 1-8; and
the pharmaceutical composition optionally further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, or any combination thereof.
10. Use of a compound according to any one of claims 1 to 8 or a pharmaceutical composition according to claim 9 for the manufacture of a medicament for the prevention or treatment of a sex hormone dependent disease;
wherein the sex hormone dependent disease is a sex hormone dependent cancer, bone metastasis of a sex hormone dependent cancer, prostatic hypertrophy, hysteromyoma, endometriosis, uterine fibroids, precocious puberty, amenorrhea, premenstrual syndrome, dysmenorrhea, multi-atrial ovarian syndrome, polycystic ovarian syndrome, acne, alopecia, infertility or irritable bowel syndrome.
CN202010281944.6A 2020-04-11 2020-04-11 Substituted pyrimidine-2, 4 (1H, 3H) -dione derivatives and uses thereof Active CN111333587B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010281944.6A CN111333587B (en) 2020-04-11 2020-04-11 Substituted pyrimidine-2, 4 (1H, 3H) -dione derivatives and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010281944.6A CN111333587B (en) 2020-04-11 2020-04-11 Substituted pyrimidine-2, 4 (1H, 3H) -dione derivatives and uses thereof

Publications (2)

Publication Number Publication Date
CN111333587A true CN111333587A (en) 2020-06-26
CN111333587B CN111333587B (en) 2023-04-07

Family

ID=71180904

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010281944.6A Active CN111333587B (en) 2020-04-11 2020-04-11 Substituted pyrimidine-2, 4 (1H, 3H) -dione derivatives and uses thereof

Country Status (1)

Country Link
CN (1) CN111333587B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111995584A (en) * 2020-09-01 2020-11-27 常州工学院 Preparation method of oxalagrill intermediate
CN114057592A (en) * 2020-08-05 2022-02-18 沈阳化工研究院有限公司 Method for preparing 4-aminomethylbenzoic acid
CN115232077A (en) * 2021-04-22 2022-10-25 成都倍特药业股份有限公司 Oxagolide sodium related substance and preparation method thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055119A2 (en) * 2000-01-25 2001-08-02 Neurocrine Biosciences, Inc. Gonadotropin-releasing hormone receptor antagonists and methods relating thereto
CN1826119A (en) * 2003-07-07 2006-08-30 纽罗克里生物科学有限公司 Pyrimidine-2,4-dione derivatives as gonadotropin-releasing hormone receptor antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055119A2 (en) * 2000-01-25 2001-08-02 Neurocrine Biosciences, Inc. Gonadotropin-releasing hormone receptor antagonists and methods relating thereto
CN1826119A (en) * 2003-07-07 2006-08-30 纽罗克里生物科学有限公司 Pyrimidine-2,4-dione derivatives as gonadotropin-releasing hormone receptor antagonists

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114057592A (en) * 2020-08-05 2022-02-18 沈阳化工研究院有限公司 Method for preparing 4-aminomethylbenzoic acid
CN114057592B (en) * 2020-08-05 2023-08-22 沈阳化工研究院有限公司 Method for preparing 4-aminomethylbenzoic acid
CN111995584A (en) * 2020-09-01 2020-11-27 常州工学院 Preparation method of oxalagrill intermediate
CN111995584B (en) * 2020-09-01 2022-03-29 常州工学院 Preparation method of oxalagrill intermediate
CN115232077A (en) * 2021-04-22 2022-10-25 成都倍特药业股份有限公司 Oxagolide sodium related substance and preparation method thereof

Also Published As

Publication number Publication date
CN111333587B (en) 2023-04-07

Similar Documents

Publication Publication Date Title
CN111925379B (en) Nitrogen-containing heteroaryl substituted pyrimidinediones and uses thereof
JP6152098B2 (en) Therapeutically active compositions and methods of their use
JP6494624B2 (en) Substituted 4,5,6,7-tetrahydropyrazolo [1,5-A] pyrazine derivatives as casein kinase 1D / E inhibitors
CN111333587B (en) Substituted pyrimidine-2, 4 (1H, 3H) -dione derivatives and uses thereof
JP6196615B2 (en) Compounds, pharmaceutical compositions thereof, and their use as inhibitors for the treatment of cancer
WO2020236940A1 (en) Kras g12c inhibitors and uses thereof
CN111574534B (en) Phenyl-substituted thieno [2,3-d ] pyrimidine-2, 4(1H,3H) -diones and their use
TW202115065A (en) Kras mutant protein inhibitor
CN107635404B (en) MCT4 inhibitors for the treatment of disease
KR20030031483A (en) Substituted pyrrolopyridinone derivatives useful as phosphodiesterase inhibitors
JP2017057221A (en) Therapeutically active compositions and their methods of use
EA012926B1 (en) Pyrrolopyrimidine aselective antagonist compounds, their synthesis and use
EP3915991A1 (en) Pyrrolopyrimidine derivative, and pharmaceutical composition for preventing or treating protein kinase-related disease comprising same as active ingredient
CN113754679B (en) Substituted pyrimidinediones and their use
WO2022115439A1 (en) Kras g12c inhibitors and uses thereof
CN112142757B (en) Five-membered nitrogen-containing heteroaryl substituted pyrimidinedione compound and application thereof
CN111875614B (en) Heterocyclyl-substituted thieno [2,3-d ] pyrimidine-2, 4(1H,3H) -diones and their use
CN111943960B (en) Substituted pyrimidinediones and their use
CN111793078B (en) Bicyclic nitrogen-containing heteroaryl substituted pyrimidinediones and application thereof
JP2010540422A (en) Thienopyrimidine compounds
EP4055013B1 (en) Wdr5 inhibitors and modulators
CN111909168B (en) Nitrogenous heterocyclic group substituted pyrimidinediones and uses thereof
CN112047957B (en) Substituted pyrimidinediones and their use
AU2020393364A1 (en) Condensed thiophene derivatives as hypoxia inducible factor (HIF) inhibitors
CN116438181A (en) Sulfonamide substituted heteroaryl pyrimidinediones and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP01 Change in the name or title of a patent holder
CP01 Change in the name or title of a patent holder

Address after: Room 501, building 4, No.368, middle Zhen'an Road, Chang'an Town, Dongguan City, Guangdong Province, 523871

Patentee after: Dongguan dongyangguang new drug R & D Co.,Ltd.

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: Room 501, building 4, No.368, middle Zhen'an Road, Chang'an Town, Dongguan City, Guangdong Province, 523871

Patentee before: Dongguan dongyangguang new drug R & D Co.,Ltd.

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.