CN108129400B - Deuterated oxalagori derivative and application thereof - Google Patents

Deuterated oxalagori derivative and application thereof Download PDF

Info

Publication number
CN108129400B
CN108129400B CN201711499447.8A CN201711499447A CN108129400B CN 108129400 B CN108129400 B CN 108129400B CN 201711499447 A CN201711499447 A CN 201711499447A CN 108129400 B CN108129400 B CN 108129400B
Authority
CN
China
Prior art keywords
fluoro
formula
dione
benzyl
trifluoromethyl
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.)
Active
Application number
CN201711499447.8A
Other languages
Chinese (zh)
Other versions
CN108129400A (en
Inventor
朱孝云
蒋维平
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.)
Jiaxing'an Dikang Biotechnology Co ltd
Original Assignee
Antikang Wuxi Biotechnology 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 Antikang Wuxi Biotechnology Co ltd filed Critical Antikang Wuxi Biotechnology Co ltd
Priority to CN201711499447.8A priority Critical patent/CN108129400B/en
Publication of CN108129400A publication Critical patent/CN108129400A/en
Priority to PCT/CN2019/076396 priority patent/WO2019129310A1/en
Application granted granted Critical
Publication of CN108129400B publication Critical patent/CN108129400B/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a method for preparing a group of deuterated oxarogalline derivatives, which are used as GnRH receptor antagonists and have the application of treating sex hormone related disease states of males and females. Also disclosed are compositions comprising a compound of the invention in combination with a pharmaceutically acceptable carrier, and methods of using the compositions to antagonize gonadotropin-releasing hormone in an individual.

Description

Deuterated oxalagori derivative and application thereof
Technical Field
The invention discloses a method for preparing a group of deuterated oxarogalline derivatives, which are used as GnRH receptor antagonists and have the purpose of treating sex hormone related diseases of males and females. Also disclosed are compositions comprising a compound of the invention in combination with a pharmaceutically acceptable carrier, and methods of using the compositions to antagonize gonadotropin-releasing hormone in an individual.
Technical Field
Gonadotropin releasing hormone (GnRH), also known as luteinizing hormone release; hormone (LHRH) is decapeptide (pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH)2) It plays an important role in human reproduction. GnRH is released from the hypothalamus and acts on the pituitary to stimulate the biosynthesis and release of Luteinizing Hormone (LH) and Follicle Stimulating Hormone (FSH). LH released from the pituitary is responsible for the regulation of sex steroid production in men and women, while FSH regulates spermatogenesis in men and follicular development in women.
GnRH antagonists are capable of inhibiting the effects of gonadotropins and are therefore of great interest. Currently, the major obstacles to the clinical use of such antagonists are their relatively low bioavailability and the side effects of histamine release.
CN200480019502.3 discloses pyrimidine-2, 4-dione derivatives as ghrelin receptor antagonists, and significant research in this field has been conducted. There remains a need in the art for effective small molecule GnRH receptor antagonists. There is also a need for pharmaceutical compositions containing such GnRH receptor antagonists and methods of using them to treat, for example, sex hormone related conditions. The present invention fulfills these needs and provides other related advantages.
Detailed Description
Briefly, the present invention is directed to compounds useful as gonadotropin releasing hormone (GnRH) receptor antagonists. The compounds of the present invention have the following structural formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or are stereoisomers, prodrugs or pharmaceutically acceptable salts thereof:
I. compound (I)
Figure BSA0000156715660000021
Figure BSA0000156715660000031
The compounds of the invention are generally used in the form of the free acid or free base. Alternatively, the compounds of the present invention may be used in the form of acid or base salts. Acid addition salts of the free amino compounds of the present invention may be prepared by methods well known in the art and may be prepared from organic and inorganic acids. Suitable organic acids include maleic acid, fumaric acid, benzoic acid, ascorbic acid, succinic acid, methanesulfonic acid, acetic acid, trifluoroacetic acid, oxalic acid, propionic acid, tartaric acid, salicylic acid, citric acid, gluconic acid, lactic acid, mandelic acid, phenylacetic acid, aspartic acid, stearic acid, palmitic acid, glycolic acid, glutamic acid, and benzenesulfonic acid. Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric and nitric acids. Base salts include salts formed with carboxylate anions and include salts formed with organic and inorganic cations such as those selected from alkali metal ions, alkaline earth metal ions (e.g., lithium, sodium, potassium, magnesium, barium, calcium), and ammonium ions, and substituted derivatives thereof (e.g., dibenzylammonium, benzylammonium, 2-hydroxyethylammonium, and the like). Thus, the terms "pharmaceutically acceptable salt" of structural formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) shall include and all acceptable salt forms.
In addition, prodrugs are also included within the scope of the present invention. A prodrug is any covalently bound carrier that releases a compound of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) in vivo when the prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner that enables the modification to be cleaved, either by conventional exchange or in vivo, to yield the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, is cleaved to yield the hydroxy, amino, or sulfhydryl group.
Thus, representative examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups of compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI). Further, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may be included.
For stereoisomers, the compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) may have chiral centers and may exist as racemates, racemic mixtures, as well as individual enantiomers or diastereomers. All isomeric forms are included in the present invention, including mixtures O thereof. Furthermore, certain crystalline forms of the compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) may exist in the form of polymorphs, which are also included in the present invention. In addition, some of the compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) may also form solvates with water or other organic solvents. Such solvates are likewise included within the scope of the present invention.
II. Application method
The GnRH receptor antagonists of the present invention are useful in a wide range of therapeutic applications and can be used to treat a variety of sex hormone-related disease states in both men and women, as well as mammals in general (also referred to herein as "subjects"). For example, such disease states include endometriosis, uterine leiomyomas, polycystic ovarian disease, hirsutism, precocious puberty, gonadal steroid-dependent neoplasias such as prostate, breast and ovary, pituitary gonadotroph adenomas, sleep disorders, irritable bowel syndrome, premenstrual syndrome, benign prostatic hypertrophy, infertility, and infertility (e.g., assisted reproductive therapy for in vitro fertilization). The compounds of the invention are also useful in the adjuvant treatment of growth hormone deficiency and short stature, and in the treatment of systemic lupus erythematosus. The compounds may also be used in combination with androgens, estrogens, progestins, and antiestrogens and antiprogestins for the treatment of endometriosis, fibroids, and for contraception, as well as in combination with angiotensin converting enzyme inhibitors, angiotensin II-receptor antagonists, or angiotensin peptidase (renin) inhibitors for the treatment of uterine leiomyoma. In addition, the compounds may also be used in combination with bisphosphonates and other agents for the treatment and/or prevention of disturbances of calcium, phosphorus and bone metabolism, and in combination with estrogens, progestins and/or androgens for the prevention or treatment of bone loss (bone loss) or symptoms of sexual hypofunction such as hot flashes which occur in the treatment with GnRH antagonists.
Use of a compound of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI), or a pharmaceutically acceptable salt, prodrug, stereoisomer or tautomer thereof, for the manufacture of a medicament for the treatment or prevention of a sex hormone related disease, wherein the disease state is cancer, benign prostatic hypertrophy, or uterine fibroids. Wherein the cancer is prostate cancer, uterine cancer, breast cancer or pituitary gonadotroph adenoma, and is further preferably prostate cancer.
Use of a compound of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or a pharmaceutically acceptable salt, prodrug, stereoisomer or tautomer thereof for the manufacture of a medicament for the treatment or prophylaxis of a sex hormone related disease, wherein the disease state is endometriosis, polycystic ovarian disease, uterine leiomyoma or precocious puberty, further preferably endometriosis and uterine leiomyoma.
A method of treating infertility in a subject, comprising administering to the subject an effective amount of a composition of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI), wherein the subject is in need of such treatment.
A method of treating lupus erythematosus, irritable bowel syndrome, premenstrual syndrome, hirsutism, short stature, or sleep disorders in a subject, comprising administering to the subject an effective amount of a composition of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI), wherein the subject is in need of such treatment.
III, preparation
Pure forms or suitable pharmaceutical compositions of the compounds of the present invention or pharmaceutically acceptable salts thereof may be administered by any acceptable mode of administration of agents that serve similar utilities. The pharmaceutical compositions of the present invention may be prepared by combining a compound of the present invention with a suitable pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into solid, semi-solid, liquid or gaseous form preparations such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres and aerosols. Typical routes of administration for the pharmaceutical compositions include, but are not limited to, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal administration. As used herein, the term parenteral includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. The pharmaceutical compositions of the present invention are formulated to allow the cleansing component contained therein to be bioavailable upon administration of the composition to a patient. The compound of the invention may be administered to a subject or patient in the form of one or more dosage units in a composition, wherein, for example, a tablet may be a single dosage unit and a container containing the compound in aerosol form may contain a plurality of dosage units. The actual methods of preparing the dosage forms are known, or will be known, to those skilled in the art. The compositions to be administered will contain, in any event, a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in order to treat the disease or condition of interest in accordance with the teachings of the present invention.
Pharmaceutical compositions
The pharmaceutical compositions of the present invention may be in solid or liquid form. In one aspect, the carrier is a microparticle, such that the composition is in the form of, for example, a tablet or powder. The carrier can be a liquid and the composition can be, for example, an oral syrup, an injectable liquid, or an aerosol suitable for administration, for example, by inhalation. When intended for oral administration, the pharmaceutical composition is preferably selected from solid or liquid forms, wherein semi-solid, semi-liquid, suspension and gel forms are included in the forms considered herein to be solid or liquid. For oral solid compositions, the pharmaceutical compositions can be formulated in the form of powders, granules, compressed tablets, pills, capsules, chewable tablets, powder tablets, and the like. Such solid compositions typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may also be present: binders, such as carboxymethyl cellulose, ethyl cellulose, microcrystalline cellulose, xanthan gum or gelatin; excipients, such as starch, lactose or dextrin; disintegrating agents, such as alginic acid, sodium alginate, Primogel, corn starch, and the like; lubricants, such as magnesium stearate or hydrogenated vegetable oil (Sterotex); glidants, such as colloidal silicon dioxide; sweetening agents, such as sucrose or saccharin; a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring; and a colorant.
When the pharmaceutical composition is in the form of a capsule, for example a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier, for example polyethylene glycol or an oil. The pharmaceutical composition may be in liquid form, such as a tincture, syrup, solution, emulsion, or suspension. Such liquids may be administered orally, or delivered by injection, as two examples. When intended for oral administration, the compositions preferably contain one or more of sweeteners, preservatives, dyes/colorants and flavor enhancers in addition to the compounds of the present invention. In compositions intended for administration by injection, one or more of surfactants, preservatives, wetting agents, dispersing agents, suspending agents, buffering agents, stabilizing agents and isotonicity agents may be included.
The liquid pharmaceutical compositions of the present invention, whether in solution, suspension or other similar form, may include one or more of the following adjuvants: sterile diluents, such as water for injection, physiological saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils (e.g., synthetic mono-or diglycerides which may be used as a solvent or suspending medium), polyethylene glycols, glycerol, propylene glycol and the like; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for adjusting tonicity such as sodium oxide or dextrose. Parenteral formulations can be packaged in ampules, disposable syringes or multi-dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. The injectable pharmaceutical composition is preferably sterile.
Liquid pharmaceutical compositions of the invention intended for parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage is obtained. The pharmaceutical compositions of the invention may be intended for topical administration, in which case the carrier preferably comprises a solution, emulsion, ointment or gel base. For example, the matrix may comprise one or more of: paraffin oil, lanolin, polyethylene glycol, beeswax, mineral oil, diluents (e.g., water and alcohol), and emulsifiers and stabilizers. Thickeners may be present in the pharmaceutical compositions for topical administration. If transdermal administration is intended, the composition may comprise a transdermal patch or an iontophoretic device.
The pharmaceutical compositions of the invention may be intended for rectal administration, in the form of suppositories for example, which will melt in the rectum and release the drug. Compositions for rectal administration may contain an oily base as a suitable non-irritating excipient. Such bases include, but are not limited to, lanolin, cocoa butter, and polyethylene glycols.
The pharmaceutical compositions of the present invention may include a variety of substances that alter the physical form of the solid or liquid dosage unit. For example, the composition may include a material that forms a coating shell around the active ingredient. The material forming the coating shell is generally inert and may be selected from, for example, sugars, shellac, and other enteric coating agents. Alternatively, the active ingredient may be encapsulated in a gelatin capsule.
The pharmaceutical compositions of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby aids in the delivery of the compound. Suitable agents with this capability include monoclonal or polyclonal antibodies, proteins or liposomes.
The pharmaceutical compositions of the present invention may be comprised of dosage units that can be administered in the form of an aerosol. The term aerosol is used to denote a variety of systems ranging from the jelly-like variety to systems consisting of pressurized packs. Delivery may be by liquefied or compressed gas, or by a suitable pump system that dispenses the active ingredient. Aerosols of the compounds of the invention may be delivered as single, two, or three phase systems to deliver the active ingredient. The delivery side of the aerosol comprises the necessary containers, actuators, valves, sub-containers, etc., which together may form a kit. Preferred aerosols are determined by one of skill in the art without undue experimentation.
The pharmaceutical compositions of the present invention can be prepared by methods well known in the pharmaceutical arts. For example, pharmaceutical compositions to be administered by injection may be prepared by combining a compound of the invention with sterile distilled water to form a solution. Surfactants may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compounds of the present invention, thereby facilitating dissolution or uniform suspension of the compounds in an aqueous delivery system.
The compounds of the present invention, or pharmaceutically acceptable salts thereof, are administered in a therapeutically effective amount, which will vary depending on a variety of factors, including the activity of the particular compound employed; metabolic stability and length of action of the compound; the age, weight, general health, sex, and diet of the patient; mode and time of administration; the rate of excretion; a pharmaceutical composition; the severity of the particular disorder or condition; and individuals undergoing therapy.
The compounds of the present invention or pharmaceutically acceptable derivatives thereof can also be administered simultaneously with, before, or after the administration of one or more other therapeutic agents. Such combination therapy includes the administration of a single pharmaceutical dosage formulation containing a compound of the present invention and one or more other active agents, as well as the administration of separate pharmaceutical dosage formulations of a compound of the present invention with each active agent itself. For example, a compound of the invention and another active agent can be administered to a patient in a single orally administered composition (e.g., a tablet or capsule), or each agent can be administered as a separate orally administered formulation. In the case of separate administration formulations, the compound of the invention and the one or more additional active agents may be administered substantially at the same time (i.e., simultaneously) or at separately staggered times (i.e., sequentially); combination therapy is understood to include all such regimens.
It is to be understood that in the present invention, combinations of substituents and/or variables of the formulas are permissible only if they result in stable compounds.
V. examples
Certain preferred embodiment aspects of the invention are shown by way of illustration in the following non-limiting examples.
Example 1.3- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione
Figure BSA0000156715660000081
Step 15- (2-fluoro-3-methoxy-phenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methylpyrimidine-2, 4- (1H, 3H) -dione
To a reaction flask was added 20g of 1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-5-iodo-6-methylpyrimidine-2, 4- (1H, 3H) -dione, 2-fluoro-3-methoxyphenylboronic acid 8.7g, 60ml acetone. The reaction mixture was cooled to 15 ℃ and potassium hydroxide/water solution (10.8g/64mL) was added and stirred. Degassing for 30min, then adding 0.142g of tri-tert-butylphosphine tetrafluoroborate, stirring and mixing at 45 ℃ for 20 min. 0.052g of palladium acetate is added into the reactor and stirred until the reaction is completed. Adding acetic acid 5.6g, stirring for 1hr, and stirring at 55 deg.C for 30 min. The reaction mixture was stirred at 25 ℃ for 2 hr. Filtering to obtain solid, washing with water, washing with methanol, and dryingDrying to obtain white solid with yield of 85%. ESI [ M + H ]]+427.4.
Step 2 (S) -Boc-phenylglycinol mesylate
Adding 17.5g of N-Boc-D-phenylglycinol, 11.4g of DIPEA and 80ml of tetrahydrofuran into a reaction bottle, and cooling to 0 ℃. Methanesulfonyl chloride was added dropwise to the reactor at 10 ℃. 26ml of THF were added. After the reaction was completed, 0.5N hydrochloric acid (3.6g concentrated hydrochloric acid/67 ml water) was added to the reactor, stirred at 10 ℃ and separated. Cyclohexane was added to the organic layer. White solid was obtained and dried to obtain (S) -Boc-phenylglycinol mesylate in 91% yield.
Step 33- ((2R) -amino-2-phenylethyl) -5- (2-fluoro-3-methoxy-phenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methylpyrimidine-2, 4- (1H, 3H) -dione
19.7g of (S) -Boc-phenylglycinol mesylate, 16.5g of 5- (2-fluoro-3-methoxy-phenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methylpyrimidine-2, 4- (1H, 3H) -dione, potassium carbonate 13.4g, 100ml DMF was added to the reaction flask. The temperature is raised to 55 ℃, after the reaction is finished, the reaction product is cooled to 20 ℃, 150ml of ethyl acetate is added for liquid separation, an ester layer is remained, and the reaction product is washed by water. Adding organic layer into reaction flask, adding 11.2g methanesulfonic acid, heating reaction solution to 60 deg.C, and stirring for 2 hr. Cooling the reaction liquid to room temperature, adding a potassium carbonate aqueous solution, adjusting the pH value, standing and layering. Rotating the ester layer to near dry, adding cyclohexane within 1hr, refluxing and pulping. Filtering to obtain white solid, yield 88%. ESI [ M + H ]]+546.2.
Step 43- [2(R) - { ethoxycarbonylpropyl-amino } -2-phenylethyl ] -5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methyl-pyrimidine-2, 4- (1H, 3H) -dione
30.7g 3- ((2R) -amino-2-phenylethyl) -5- (2-fluoro-3-methoxy-phenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methylpyrimidine-2, 4- (1H, 3H) -dione, 30ml DMF were added to the reaction vessel. Ethyl 4-bromobutyrate (11.8 g) and DIPEA (8.9 g) were added thereto, and the mixture was heated to 52 ℃ and stirred until the reaction was completed. To the reaction flask, 140ml of ethyl acetate and 140ml of water were added. The aqueous layer was separated and the ester layer was washed with water. An aqueous phosphoric acid solution (15g phosphoric acid/30 g water) was added and washed with water. 15ml of ethyl acetate were added, and 35g of potassium carbonate/43 g of aqueous solution were slowly added and stirred. The organic layer was separated, concentrated, flash column chromatographed, washed with ethanol/dichloromethane (1: 90), and the collected liquid was concentrated and used directly in the next step.
Step 5 (Oxagolide) 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl ] -5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methyl-pyrimidine-2, 4- (1H, 3H) -dione
60g of ethanol was added to the above concentrated solution, and the mixture was evaporated to dryness. 60g of ethanol was added, 6g of NaOH4.5g/water was added, and the mixture was heated to 35 ℃ until the reaction was completed. Heating 90g of water, concentrating under reduced pressure, adding 150g of water, and cooling to below 15 ℃. Adding hydrochloric acid solution dropwise until pH is 6.1, maintaining the temperature at room temperature, and stirring for 15 hr. Filtering, washing filter cake with water, drying to obtain white solid with yield of 86%. ESI [ M + H ]]+632.2.
Step 63- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione sodium salt
5g of 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, 15g sodium carbonate, 50mL of heavy water (D)2O), refluxing for 15hr, cooling, adding 150ml acetone, filtering, drying to obtain 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione sodium salt.
1H-NMR(CD3OD):δ1.71-1.79(m,2H),2.11-2.20(t,2H),2.50-2.54(t,2H),3.90(s,3H),4.16-4.35(m,3H),5.38-5.55(m,2H),6.61-7.66(m,11H)。
Step 73- [2(R) - { Hydroxycarbonylpropyl-amino) -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione
5g of 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, 15g sodium carbonate, 50mL weightWater (D)2O), refluxing for 15hr, cooling, adding hydrochloric acid to adjust ph to 6.1, stirring for 3hr, and filtering to obtain 3- [2(R) - { hydroxycarbonylpropyl-amino) -2-phenylethyl]-5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione. ESI [ M + H ]]+635.2.
Example 2.3- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione
Figure BSA0000156715660000101
Step 12-fluoro-3- (D)3-methoxy) phenylboronic acid
Cooling 11.2g of o-fluorophenol and 100ml of tetrahydrofuran to 0 ℃, adding 4.8g of NaH, stirring, dropwise adding 21.3g of deuterated iodomethane, and reacting for 1 hr. Adding sodium sulfite solution to quench, extracting and spin-drying to obtain 2-fluoro-benzene- (D3-methyl) ether with yield of 90%.
7.9g 2-fluoro-benzene- (D)3-methyl) ether and 10.4ml of PMDAT were dissolved in 60ml of THF solution, cooled to-78 ℃ and 34ml of 1.6M n-butyllithium were slowly added to the reaction solution, and reacted for 2 hr. 16.8ml of trimethyl borate was slowly added to the reaction mixture at-78 ℃ and then slowly warmed to room temperature and stirred for 8 hours. Adding 50ml 1N hydrochloric acid dropwise, acidifying for 1hr, extracting with ethyl acetate, and spin drying to obtain 2-fluoro-3- (D)3-methoxy) phenylboronic acid, yield 71%.
Step 25- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methylpyrimidine-2, 4- (1H, 3H) -dione
To a reaction flask was added 20g of 1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-5-iodo-6-methylpyrimidine-2, 4- (1H, 3H) -dione, 2-fluoro-3- (D)3-methoxy) phenylboronic acid 8.7g, 60ml acetone. The reaction mixture was cooled to 15 ℃ and potassium hydroxide/water solution (10.8g/64mL) was added and stirred. Degassing for 30min, then adding 0.142g of tri-tert-butylphosphine tetrafluoroborate, stirring and mixing at 45 ℃ for 20 min. 0.052g of palladium acetate is added into the reactor and stirred until the reaction is finishedAnd (4) completing. Adding acetic acid 5.6g, stirring for 1hr, and stirring at 55 deg.C for 30 min. The reaction mixture was stirred at 25 ℃ for 2 hr. Filtering to obtain solid, washing with water, washing with methanol, and drying to obtain white solid with yield of 85%]+430.4.
Step 33- ((2R) -amino-2-phenylethyl) -5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methylpyrimidine-2, 4- (1H, 3H) -dione
19.7g of (S) -Boc-phenylglycinol mesylate, 16.6g of 5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methylpyrimidine-2, 4- (1H, 3H) -dione, potassium carbonate 13.4g, 100ml DMF was added to the reaction flask. The temperature is raised to 55 ℃, after the reaction is finished, the reaction product is cooled to 20 ℃, 150ml of ethyl acetate is added for liquid separation, an ester layer is remained, and the reaction product is washed by water. Adding organic layer into reaction flask, adding 11.2g methanesulfonic acid, heating reaction solution to 60 deg.C, and stirring for 2 hr. Cooling the reaction liquid to room temperature, adding a potassium carbonate aqueous solution, adjusting the pH value, standing and layering. Rotating the ester layer to near dry, adding cyclohexane within 1hr, refluxing and pulping. Filtering to obtain white solid, yield 88%. ESI [ M + H ]]+549.2.
Step 43- [2(R) - { ethoxycarbonylpropyl-amino) -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione
30.9g 3- ((2R) -amino-2-phenylethyl) -5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methylpyrimidine-2, 4- (1H, 3H) -dione, 30ml DMF was added to the reaction kettle. Ethyl 4-bromobutyrate (11.8 g) and DIPEA (8.9 g) were added thereto, and the mixture was heated to 52 ℃ and stirred until the reaction was completed. To the reaction flask, 140ml of ethyl acetate and 140ml of water were added. The aqueous layer was separated and the ester layer was washed with water. An aqueous phosphoric acid solution (15g phosphoric acid/30 g water) was added and washed with water. 15ml of ethyl acetate were added, and 35g of potassium carbonate/43 g of aqueous solution were slowly added and stirred. The organic layer was separated, concentrated, flash column chromatographed, washed with ethanol/dichloromethane (1: 90), and the collected liquid was concentrated and used directly in the next step.
Step 53- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl)Yl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione
60g of ethanol was added to the above concentrated solution, and the mixture was evaporated to dryness. 60g of ethanol was added, 6g of NaOH4.5g/water was added, and the mixture was heated to 35 ℃ until the reaction was completed. Heating 90g of water, concentrating under reduced pressure, adding 150g of water, and cooling to below 15 ℃. Adding hydrochloric acid solution dropwise until pH is 6.1, maintaining the temperature at room temperature, and stirring for 15 hr. Filtering, washing filter cake with water, drying to obtain white solid with yield of 86%. ESI [ M + H ]]+635.2.
Step 63- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione sodium salt
5g of 3- [2(R) - { hydroxycarbonylpropyl-amino) -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, 15g sodium carbonate, 50mL water, refluxing for 5hr, cooling, adding 150mL acetone, and drying to give 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione sodium salt.
1H-NMR(CD3OD):δ1.71-1.79(m,2H),2.09(s,3H),2.12-2.21(t,2H),2.49-2.55(t,2H),4.15-4.33(m,3H),5.36-5.54(m,2H),6.58-7.65(m,11H)。
Example 3.3- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione
Figure BSA0000156715660000121
Step 13- [2(R) - { Hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione sodium salt
5g 3-[2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl group]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, 15g sodium carbonate, 50mL of heavy water (D)2O), refluxing for 15hr, cooling, adding 150ml acetone, and drying to obtain 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl group]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione sodium salt.
1H-NMR(CD3OD):δ1.72-1.80(m,2H),2.10-2.19(t,2H),2.51-2.56(t,2H),4.17-4.37(m,3H),5.39-5.54(m,2H),6.65-7.69(m,11H)。
Step 23- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione
5g of 3- [2(R) - { hydroxycarbonylpropyl-amino) -2-phenylethyl]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, 15g sodium carbonate, 50mL of heavy water (D)2O), refluxing for 15hr, cooling, adding hydrochloric acid to adjust ph to 6.1, stirring for 3hr, and filtering to obtain 3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl group]-5- [ 2-fluoro-3- (D)3-methoxy) phenyl]-1- [ 2-fluoro-6- (trifluoromethyl) benzyl]-6-(D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione. ESI [ M + H ]]+638.2.
Table compounds:
using a similar protocol to that above, the following table compounds and their individual enantiomers were prepared
Table 1: table compound structure.
Figure BSA0000156715660000131
Example 4 GnRH receptor antagonist Activity
By measuring the compound and [2 ]125I-Tyr5,DLeu6,NMeLeu7,Pro9-NEt]GnRH and hGnRHR competitive affinity for determining GnRH receptor antagonist activity, reference is made to the literature (struts, R.S.; Xie, Q.; Sullivan, S.K.; Reinhart, G.J.; Kohout, T.A. pharmacological. characterization of a novel peptide antagonist assay of the human gonadotropin-refining hormone receptor, NBI-42902.Endocrinology 2007, 148, 857-. For other membrane binding assays, stably transfected HEK293 cells were harvested by blowing tissue culture flasks on a hard surface and harvested by centrifugation at 1000 × g for 5 minutes. The cell pellet was resuspended in 5% sucrose and triturated with a polytron triturator for two 15 second trituration steps. Cell membrane fractions were collected by centrifugation at 3000Xg for 5 minutes to remove nuclei from the cell homogenate followed by centrifugation of the supernatant 44000Xg for 30 minutes. The cell membrane pellet was resuspended in GnRH binding buffer (10mM HEPES, pH 7.5, 150mM NaCl and 0.1% BSA) and aliquots were immediately frozen in an atmosphere and stored at-80 ℃. The protein content of the cell membrane suspension was determined using a Bio-Rad protein assay kit (Bio-Rad, Hercules, Calif.).
Competitive radioactive 20-shot ligand binding assays were performed with cell membrane preparations in Millpore 96-well filtration plates with filters pre-coated with 200. mu.l of 0.1% polyazaprop (Sigma, St. Louis. MO). Before use, the plates were washed 3 times with phosphate buffered saline. The membrane fraction in GnRH binding buffer was added to the wells together with 20 μ L of competitor ligand at various concentrations. The binding reaction was initiated by the addition of radioligand (0.1 nM in 50. mu.L GnRH binding buffer). The reaction was allowed to proceed at room temperature on a platform shaker for 90 minutes and was stopped by placing the assay plate on a Millipore vacuum manifold, aspirating the solvent, and washing 2 times with 200 μ Ι _ cold Phosphate Buffer (PBS). The filters in the wells were removed and counted in a gamma counter. Ki values were calculated from each competition binding curve using non-linear least squares regression and radioligand concentrations were corrected using the Cheng-Prusoff equation (Prism, GraphPad Software, San Diego, Calif.). The average Ki values were calculated from the inverse logarithm of the average pKi values for each receptor ligand pair. The measurement results (hGnRH-R Ki) are shown in Table 2 below.
EXAMPLE 5 Activity of antagonists to inhibit GnRH-stimulated phosphoinositide production
Reference is made to the literature for the protocol of this example (Benjamin, E.R.; Haftl, S.L.; Xanthos, D.N.; Crumley, G.; Hachicha, M.A minor worked column chromatography method for measuring receiver-treated accelerator culture. J.Biomol. screening 2004, 9, 343-. RBL-1 cell lines stably expressing human GnRH receptors were selected and inoculated into 96-well plates at a density of 1500 cells/well in the presence of inositol-free DMEM medium containing 10% dialyzed fetal bovine serum, 10mM HEPES buffer, 2mM L-glutamine, 1mM sodium pyruvate, 0.1mM non-essential amino acids, and 50. mu.g/mL penicillin-streptomycin solution. The RBL-1 cell line was cultured with 0.2. mu. Ci myo-2-, [ solution of3H]Inositol was labeled in the medium for 48 hours. The cell lines were washed once with PBS and pretreated for 50min at 37 ℃ with assay buffer 4mM KCl, 10mM LiCl, 140mM NaCl, 20mM HEPES, 8.3mM glucose, 1mM MgCl2,1mM CaCl2Cells were stimulated with GnRH (6nM), test compound was added and incubated at 37 ℃ for 60min and a blank control was set. Cells were extracted with 10mM formic acid at 4 ℃ for 30min, and the extracts were applied to Millipore (MAGVN0B50) plates containing 20. mu.g of AG1-X8 resin, which were washed once with water and once with 60mM ammonium formate/5 mM sodium triborate. Inositol phosphates were eluted with 1M ammonium formate/0.1M formic acid. The eluate was transferred to a LUMA plate and counted using a microplate scintillation counter. Measurement results (hGnRH-R IP IC)50) As in table 2 below.
Example 6 human liver microsomal enzyme CYP3A4 inhibitory Activity
Reference is made to the literature (ZLokarnik, G.; Grootenhuis, P.D.; Watson, J.B.high through serum P450 inhibition strategies in early drug DiscoVery. drug DiscoVery Today 2005, 10, 1443. the preparation of human liver microsomes is carried out by ultracentrifugation, the protein content being determined by Lowy et al using calf serum albumin as a standard sample (literature: OMURA T, SATO R.the calf serum-binding fragment of liver micro.II.Solubilization purification, and properties.J Biol m, 1964, 239: 2379. 2385). Determination of CYPs content by the OMURA method of the literatureAmount of the compound (A). The probe substrate used for CYP3a4 was testosterone. The positive inhibitor is ketoconazole with the concentration of 1 mu mol/L. The half Inhibitory Concentrations (IC) were determined from the different concentrations of the example compounds50) The results are shown in Table 2.
TABLE 2 GnRH antagonist in vitro test results
Figure BSA0000156715660000151
The results of the in vitro test of GnRH antagonists show that: the compounds of the examples all showed stronger affinity compared to the human GnRH affinity versus the control drug group, where example 2 was 2 times the affinity activity of oxalagril. The compounds of the examples also showed various degrees of improvement in the activity of inhibiting the production of inositol phosphate compared to the control group, wherein example 2 was about 2 times as much as oxalagril. The compound of the example has extremely weak inhibitory activity on human liver microsomal CYP3A4 enzyme, and taking the compound of the example 3 as an example, the enzymatic activity of the compound of the example 3 is 8-9 times that of the compound of the example 3 in oxalagogrel on human CYP3A 4. In summary, the example compounds 1-3 showed superior GnRH antagonist antagonistic activity in vitro and were more stable against liver microsomal enzymes in vitro.
Example 7 pharmacokinetic parameter testing
Injection and oral doses: 10 mg/kg; animals: a rat; water as a solvent, complete injection for 15min, and taking blood from veins after administration, time points: 0.25, 0.33, 0.5, 1, 1.5, 4, 8, 24hr. water as vehicle, gavage, post-administration venous blood draw, time points: centrifuging for 30min for blood sample at 0.25, 0.33, 0.5, 1, 1.5, 4, 8, 24hr, collecting plasma, and storing at-70 deg.C. Proteins were precipitated, injected, analyzed by LC-MS/MS, analyzed by the instrument Waters 2790 HPLC, containing a Micromass Quattro-LC module. Pharmacokinetic parameters were calculated using WinNonlin program, version 3.2 software processing. The results are shown in Table 3.
TABLE 3 GnRH antagonist in vivo pharmacokinetic parameters in rats
Figure BSA0000156715660000161
As can be seen from the data in table 3, the in vivo half-life of the compound of example is significantly prolonged compared to the control group, wherein example 3 is 2.5 times that of the control group. The maximum blood concentration of the compound of example 3 is 1.7 times that of oxa-rogue, and the absolute bioavailability is improved by nearly 2 times. Therefore, the compound of the embodiment has good metabolic stability in vivo and has better clinical development prospect.
EXAMPLE 8 evaluation of the therapeutic Effect of GnRH antagonists on hysteromyoma
The model is built by a female and progestogen loading method to build a rat experimental hysteromyoma model, SD rats are selected and randomly divided into two groups, namely a hysteromyoma model building group and a blank control group. The molding method refers to the literature report [ experimental study on treating hysteromyoma with oral liquid of Yiming Juan and Gongning, pharmacology and clinic of traditional Chinese medicine, 1996, 12 (4): 41-43]. The model building group is given with 0.06 mL/body (containing 0.12mg) of estradiol benzoate for intramuscular injection, 3 times/week (Monday, three and five), 16 weeks, and 0.05 mL/body (containing 1mg) of progesterone for 10 weeks, 2 times/week (Tuesday and four), and 7 weeks. The blank control group is given with 0.05 mL/piece of high-temperature sterilized peanut oil for intramuscular injection for 3 times/week for 16 weeks. Compared with the blank control group, the rats in the model group have obvious uterine smooth muscle proliferation, and the two rats have obvious difference (P is less than 0.001), which indicates that the scheme is successfully modeled.
Dosing was randomized into groups of 12 rats each. Gavage administration, example compound 2 and example compound 3, oxalagril, dose: 1.7mg/kg (low dose group) and 2.5mg/kg (high dose group). Water as vehicle, 1 time/day (10: 00 am) for 4 weeks. After the last administration, rats in each group were fasted for 14h at 20 o' clock late, and in the following morning, rats in each group were bled from the carotid artery after intravenous anesthesia with 10% chloral hydrate, and after bleeding, the rats were sacrificed by cervical dislocation and uterus was taken, and fixed with 10% phosphate buffered formalin.
Histological morphological examination of uterus was fixed in 10% formalin, paraffin-embedded sections, HE stained, and histological morphological changes of uterus were observed under microscope. Smooth muscle cell proliferation degree scoring standard: (-): the myometrium layer is not thickened, inflammatory infiltration is not seen, and the structure is normal. (+): the myometrium layer was not significantly thickened and was mildly inflammatory infiltrated. (++): the myometrium is slightly thickened, and inflammatory cell infiltration is obvious. (+++): the myometrium is obviously thickened, and inflammatory cells are heavily infiltrated. The significant efficiency rate is (n (-) + n (+))/13, and the test results are shown in table 4.
TABLE 4 Effect of GnRH antagonists on the pathological conditions in the hysteromyoma model rats
Figure BSA0000156715660000171
As can be seen from the results in table 4, the high dose groups in examples 2 and 3 have higher significant effective rates, the low dose groups in examples 2 and 3 have equivalent therapeutic effects to the high dose group of loragoid, and the low dose of loragoid has weak pathological improvement on the rats with the hysteromyoma model.
It will be appreciated by those skilled in the art that the present disclosure is not limited to the foregoing illustrative embodiments, but may be embodied in other specific forms without departing from the essential attributes thereof. It is therefore intended that the embodiments described herein are to be considered in all respects as illustrative and not restrictive, the embodiments referenced by the appended claims being other than the foregoing embodiments, the references being made to the appended claims rather than to the foregoing examples, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.
All patents, patent applications, and literature references cited in this specification are hereby incorporated by reference in their entirety. In the event of inconsistencies, the present disclosure, including definitions, will be convincing.

Claims (5)

1. A compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl ] -5- (2-fluoro-3-methoxyphenyl) -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6- (D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione,
3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl ] -5- [ 2-fluoro-3- (D3-methoxy) phenyl ] -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6-methyl-pyrimidine-2, 4- (1H, 3H) -dione, or
3- [2(R) - { hydroxycarbonylpropyl-amino } -2-phenylethyl ] -5- [ 2-fluoro-3- (D3-methoxy) phenyl ] -1- [ 2-fluoro-6- (trifluoromethyl) benzyl ] -6- (D3-methyl) -pyrimidine-2, 4- (1H, 3H) -dione.
2. A pharmaceutical composition comprising an effective amount of a compound of any one of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
3. Use of a composition according to claim 2 for the manufacture of a medicament for the treatment or prevention of a sex hormone related disorder, wherein the disorder is cancer, benign prostatic hypertrophy or uterine fibroids.
4. Use of a composition according to claim 3 for the manufacture of a medicament for the treatment or prevention of a gonadotropin related disease wherein said cancer is prostate cancer, uterine cancer, breast cancer or pituitary gonadotroph adenomas.
5. Use of a composition according to claim 2 for the manufacture of a medicament for the treatment or prevention of a gonadal-hormone related condition, wherein the condition is endometriosis, polycystic ovarian disease, uterine leiomyoma, precocious puberty, infertility, lupus erythematosus, irritable bowel syndrome, premenstrual syndrome, hirsutism, short stature or sleep disorders.
CN201711499447.8A 2017-12-29 2017-12-29 Deuterated oxalagori derivative and application thereof Active CN108129400B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201711499447.8A CN108129400B (en) 2017-12-29 2017-12-29 Deuterated oxalagori derivative and application thereof
PCT/CN2019/076396 WO2019129310A1 (en) 2017-12-29 2019-02-28 Deuterated elagolix derivative, pharmaceutical composition, and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201711499447.8A CN108129400B (en) 2017-12-29 2017-12-29 Deuterated oxalagori derivative and application thereof

Publications (2)

Publication Number Publication Date
CN108129400A CN108129400A (en) 2018-06-08
CN108129400B true CN108129400B (en) 2021-10-15

Family

ID=62400463

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201711499447.8A Active CN108129400B (en) 2017-12-29 2017-12-29 Deuterated oxalagori derivative and application thereof

Country Status (2)

Country Link
CN (1) CN108129400B (en)
WO (1) WO2019129310A1 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019036713A1 (en) 2017-08-18 2019-02-21 Abbvie Inc. Solid pharmaceutical formulations for treating endometriosis, uterine fibroids, polycystic ovary syndrome and adenomyosis
US20190054088A1 (en) 2017-08-18 2019-02-21 Abbvie Inc. Pharmaceutical Formulations for Treating Endometriosis, Uterine Fibroids, Polycystic Ovary Syndrome or Adenomyosis
CN108129400B (en) * 2017-12-29 2021-10-15 安帝康(无锡)生物科技有限公司 Deuterated oxalagori derivative and application thereof
CN108586359B (en) * 2018-06-26 2020-04-28 杭州科巢生物科技有限公司 Synthesis method of oxaagolide
EP3829584A4 (en) * 2018-08-01 2022-06-08 Abbvie Inc. Dosing regimens for elagolix
CA3126925A1 (en) * 2019-01-15 2020-07-23 Lupin Inc. Deuterated elagolix-like compositions and methods
CN111116492B (en) * 2019-01-25 2021-07-09 青岛吉澳医药科技有限公司 Deuterated benzylaminopyrimidinedione derivative and application thereof
CN111689907A (en) * 2019-03-16 2020-09-22 苏州鹏旭医药科技有限公司 Crystal form A of intermediate of elogolide and sodium salt thereof, preparation method and application
CN110372608A (en) * 2019-07-09 2019-10-25 丽珠集团新北江制药股份有限公司 A kind of preparation method of antagonists of gonadotropin-releasing hormone
CN110437159B (en) * 2019-08-13 2021-02-09 丽珠集团新北江制药股份有限公司 Gonadotropin releasing hormone antagonist intermediate and preparation method of antagonist sodium
CN112694445B (en) * 2019-10-22 2023-07-28 成都倍特药业股份有限公司 Purification method of oxaagole sodium intermediate
CN110759870B (en) * 2019-12-06 2021-03-23 上海博璞诺科技发展有限公司 Synthesis method of oxalagogri intermediate
CN113666943A (en) * 2020-05-15 2021-11-19 苏州泽璟生物制药股份有限公司 Deuterated tetrahydrothieno [3,4-d ] pyrimidinedione compound and pharmaceutical composition containing same
CN113912552A (en) * 2020-07-09 2022-01-11 成都苑东生物制药股份有限公司 Preparation method of amorphous form of oxalagogri sodium
CN112679442B (en) * 2021-01-08 2022-09-06 浙江乐普药业股份有限公司 Preparation method of oxaagolide sodium
CN114685379A (en) * 2022-04-29 2022-07-01 河北智恒医药科技股份有限公司 Preparation method and application of intermediate 1 and intermediate 2 of oxaagolide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1819829A (en) * 2003-07-07 2006-08-16 纽罗克里生物科学有限公司 Pyrimidine-2, 4-dione derivatives as gonadotropin-releasing hormone receptor antagonists

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108129400B (en) * 2017-12-29 2021-10-15 安帝康(无锡)生物科技有限公司 Deuterated oxalagori derivative and application thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1819829A (en) * 2003-07-07 2006-08-16 纽罗克里生物科学有限公司 Pyrimidine-2, 4-dione derivatives as gonadotropin-releasing hormone receptor antagonists

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GnRHa临床应用进展;陈春艳,等;《浙江中西医结合杂志》;20170820;第27卷(第8期);第724-727页 *

Also Published As

Publication number Publication date
CN108129400A (en) 2018-06-08
WO2019129310A1 (en) 2019-07-04

Similar Documents

Publication Publication Date Title
CN108129400B (en) Deuterated oxalagori derivative and application thereof
US10941152B2 (en) Pyrazole compounds as modulators of FSHR and uses thereof
US11365199B2 (en) Pyrazole compounds as modulators of FSHR and uses thereof
JPH11502538A (en) Gonadotropin-releasing hormone antagonist
CN100376246C (en) Pyrimidine-2,4-dione derivatives as gonadotropin-releasing hormone receptor antagonists
EP2094702B1 (en) Viral inhibitors
JPH11506471A (en) Gonadotropin-releasing hormone antagonist
JP2002510630A (en) Gonadotropin-releasing hormone antagonist
JP2002535244A (en) Non-peptide GnRH agent, preparation method thereof and intermediate
JPWO2006080533A1 (en) 3-amino-1,2,4-triazole derivatives
JP2002510629A (en) Gonadotropin-releasing hormone antagonist
JP2002505318A (en) Non-peptide GnRH factor
JP2002510685A (en) Gonadotropin-releasing hormone antagonist
WO2007011072A1 (en) Premature ovulation preventive agent
NO341019B1 (en) Propane-1,3-dione derivative and salt thereof, pharmaceutical composition, use in the manufacture of a medicament, and use in a method of treatment
JP2002510628A (en) Gonadotropin-releasing hormone antagonist
JP2002538208A (en) 6-azaindole compounds as gonadotropin-releasing hormone antagonists
JP2002539124A (en) 6-azaindole compounds as gonadotropin-releasing hormone antagonists
JP2002510631A (en) Gonadotropin-releasing hormone antagonist
JP2002510684A (en) Gonadotropin-releasing hormone antagonist
US5985892A (en) Antagonists of gonadotropin releasing hormone
JP2003526618A (en) 6-azaindole compounds as gonadotropin-releasing hormone antagonists
JP2002538205A (en) 6-azaindole compounds as gonadotropin-releasing hormone antagonists
US10138233B2 (en) Imidazole compounds as modulators of FSHR and uses thereof
TW201825489A (en) Pentacyclic compound as selective estrogen receptor down-regulator and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20180903

Address after: 201399 Pudong New Area, Shanghai Hui Nan Town, 2 New Culture Village 3 3

Applicant after: Antikang (Wuxi) Biotechnology Co.,Ltd.

Address before: 201399 Pudong New Area, Shanghai Hui Nan Town, 2 New Culture Village 3 3

Applicant before: Zhu Xiaoyun

TA01 Transfer of patent application right
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address

Address after: 314006 Floor 9, No. 152, Huixin Road, Daqiao Town, Nanhu District, Jiaxing City, Zhejiang Province

Patentee after: Jiaxing'an Dikang Biotechnology Co.,Ltd.

Address before: 201399 Pudong New Area, Shanghai Hui Nan Town, 2 New Culture Village 3 3

Patentee before: Antikang (Wuxi) Biotechnology Co.,Ltd.

CP03 Change of name, title or address